1
|
Zumaraga MP, Desmarchelier C, Gleize B, Nowicki M, Ould-Ali D, Borel P. Characterization of the interindividual variability of lutein and zeaxanthin concentrations in the adipose tissue of healthy male adults and identification of combinations of genetic variants associated with it. Food Funct 2024; 15:9995-10006. [PMID: 39279719 DOI: 10.1039/d4fo03087g] [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: 09/18/2024]
Abstract
Lutein (L) and zeaxanthin (Z) are involved in visual function and could prevent age-related macular degeneration and chronic diseases and improve cognitive performances. Adipose tissue is the main storage site for these xanthophylls (Xanth). The factors affecting their concentrations in this tissue remain poorly understood but in animal models, genetic variations in apolipoprotein E and β-carotene oxygenase 2 have been associated with adipose tissue L concentration. Therefore, the aims of this study were to better characterize the interindividual variability of adipose tissue Xanth concentration and to identify single nucleotide polymorphisms (SNPs) associated with it. Periumbilical subcutaneous adipose tissue samples were collected on 6 occasions in 42 healthy adult males and L and Z concentrations were measured by HPLC. Participants had their whole genome genotyped and the associations of 3589 SNPs in 49 candidate genes with the concentrations of L and Z were measured. Mean L and Z concentrations were 281 ± 27 and 150 ± 14 nmol g-1 proteins, respectively. There was no significant correlation between plasma and adipose tissue Xanth concentrations, although the correlation for L approached significance (Pearson's r = 0.276, p = 0.077). Following univariate filtering, 109 and 97 SNPs were then entered into a partial least squares regression analysis to identify the combination of SNPs that explained best adipose tissue concentration of L and Z, respectively. A combination of 7 SNPs in ELOVL5, PPARG, ISX and ABCA1, explained 58% of the variability in adipose tissue L concentration while 11 SNPs located in or near PPARG, ABCA1, ELOVL5, CXCL8, IRS1, ISX, MC4R explained 53% of the variance in adipose tissue Z concentration. This suggests that some genetic variations influence the concentrations of these Xanth in adipose tissue and could therefore indirectly influence the health effects of these compounds. Clinical Trial Registry: https://ClinicalTrials.gov registration number NCT02100774.
Collapse
Affiliation(s)
- Mark Pretzel Zumaraga
- C2VN, Aix-Marseille Univ, INRAE, INSERM, 27, boulevard Jean Moulin, 13385 Marseille Cedex 5, France.
- Department of Science and Technology - Food and Nutrition Research Institute, Bicutan, Taguig City, Philippines
| | - Charles Desmarchelier
- C2VN, Aix-Marseille Univ, INRAE, INSERM, 27, boulevard Jean Moulin, 13385 Marseille Cedex 5, France.
- Institut Universitaire de France (IUF), France
| | - Beatrice Gleize
- C2VN, Aix-Marseille Univ, INRAE, INSERM, 27, boulevard Jean Moulin, 13385 Marseille Cedex 5, France.
| | - Marion Nowicki
- C2VN, Aix-Marseille Univ, INRAE, INSERM, 27, boulevard Jean Moulin, 13385 Marseille Cedex 5, France.
| | - Djaffar Ould-Ali
- Plastic & Anesthetic Surgery Department, Clinique Internationale du Parc Monceau, Paris, France
| | - Patrick Borel
- C2VN, Aix-Marseille Univ, INRAE, INSERM, 27, boulevard Jean Moulin, 13385 Marseille Cedex 5, France.
| |
Collapse
|
2
|
Soto-Catalán M, Opazo-Ríos L, Quiceno H, Lázaro I, Moreno JA, Gómez-Guerrero C, Egido J, Mas-Fontao S. Semaglutide Improves Liver Steatosis and De Novo Lipogenesis Markers in Obese and Type-2-Diabetic Mice with Metabolic-Dysfunction-Associated Steatotic Liver Disease. Int J Mol Sci 2024; 25:2961. [PMID: 38474208 DOI: 10.3390/ijms25052961] [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: 02/03/2024] [Revised: 02/26/2024] [Accepted: 02/28/2024] [Indexed: 03/14/2024] Open
Abstract
Metabolic-dysfunction-associated steatotic liver disease (MASLD) is a prevalent clinical condition associated with elevated morbidity and mortality rates. Patients with MASLD treated with semaglutide, a glucagon-like peptide-1 receptor agonist, demonstrate improvement in terms of liver damage. However, the mechanisms underlaying this beneficial effect are not yet fully elucidated. We investigated the efficacy of semaglutide in halting MASLD progression using a genetic mouse model of diabesity. Leptin-receptor-deficient mice with obesity and diabetes (BKS db/db) were either untreated or administered with semaglutide for 11 weeks. Changes in food and water intake, body weight and glycemia were monitored throughout the study. Body fat composition was assessed by dual-energy X-ray absorptiometry. Upon sacrifice, serum biochemical parameters, liver morphology, lipidomic profile and liver-lipid-related pathways were evaluated. The semaglutide-treated mice exhibited lower levels of glycemia, body weight, serum markers of liver dysfunction and total and percentage of fat mass compared to untreated db/db mice without a significant reduction in food intake. Histologically, semaglutide reduced hepatic steatosis, hepatocellular ballooning and intrahepatic triglycerides. Furthermore, the treatment ameliorated the hepatic expression of de novo lipogenesis markers and modified lipid composition by increasing the amount of polyunsaturated fatty acids. The administration of semaglutide to leptin-receptor-deficient, hyperphagic and diabetic mice resulted in the amelioration of MASLD, likely independently of daily caloric intake, suggesting a direct effect of semaglutide on the liver through modulation of the lipid profile.
Collapse
Affiliation(s)
- Manuel Soto-Catalán
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundación Jiménez Díaz, Spanish Biomedical Universidad Autónoma de Madrid, 28049 Madrid, Spain
- Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), 28029 Madrid, Spain
| | - Lucas Opazo-Ríos
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundación Jiménez Díaz, Spanish Biomedical Universidad Autónoma de Madrid, 28049 Madrid, Spain
- Facultad de Ciencias de la Salud, Universidad de Las Américas, Concepción-Talcahuano 4301099, Chile
| | - Hernán Quiceno
- Department of Pathology, Fundación Jiménez Díaz, 28040 Madrid, Spain
| | - Iolanda Lázaro
- Cardiovascular Risk and Nutrition Research Group, Epidemiology and Public Health Program, Hospital del Mar Medical Research Institute (IMIM), 08003 Barcelona, Spain
| | - Juan Antonio Moreno
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, 140471 Cordoba, Spain
- Maimónides Biomedical Research Institute of Cordoba (IMIBIC), Hospital Universitario Reina Sofía, 14004 Córdoba, Spain
| | - Carmen Gómez-Guerrero
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundación Jiménez Díaz, Spanish Biomedical Universidad Autónoma de Madrid, 28049 Madrid, Spain
- Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), 28029 Madrid, Spain
| | - Jesús Egido
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundación Jiménez Díaz, Spanish Biomedical Universidad Autónoma de Madrid, 28049 Madrid, Spain
- Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), 28029 Madrid, Spain
| | - Sebastian Mas-Fontao
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundación Jiménez Díaz, Spanish Biomedical Universidad Autónoma de Madrid, 28049 Madrid, Spain
- Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), 28029 Madrid, Spain
- Faculty of Medicine and Biomedicine, Universidad Alfonso X el Sabio (UAX), 28691 Madrid, Spain
| |
Collapse
|
3
|
Reijnders E, van der Laarse A, Ruhaak LR, Cobbaert CM. Closing the gaps in patient management of dyslipidemia: stepping into cardiovascular precision diagnostics with apolipoprotein profiling. Clin Proteomics 2024; 21:19. [PMID: 38429638 PMCID: PMC10908091 DOI: 10.1186/s12014-024-09465-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 02/14/2024] [Indexed: 03/03/2024] Open
Abstract
In persons with dyslipidemia, a high residual risk of cardiovascular disease remains despite lipid lowering therapy. Current cardiovascular risk prediction mainly focuses on low-density lipoprotein cholesterol (LDL-c) levels, neglecting other contributing risk factors. Moreover, the efficacy of LDL-c lowering by statins resulting in reduced cardiovascular risk is only partially effective. Secondly, from a metrological viewpoint LDL-c falls short as a reliable measurand. Both direct and calculated LDL-c tests produce inaccurate test results at the low end under aggressive lipid lowering therapy. As LDL-c tests underperform both clinically and metrologically, there is an urging need for molecularly defined biomarkers. Over the years, apolipoproteins have emerged as promising biomarkers in the context of cardiovascular disease as they are the functional workhorses in lipid metabolism. Among these, apolipoprotein B (ApoB), present on all atherogenic lipoprotein particles, has demonstrated to clinically outperform LDL-c. Other apolipoproteins, such as Apo(a) - the characteristic apolipoprotein of the emerging risk factor lipoprotein(a) -, and ApoC-III - an inhibitor of triglyceride-rich lipoprotein clearance -, have attracted attention as well. To support personalized medicine, we need to move to molecularly defined risk markers, like the apolipoproteins. Molecularly defined diagnosis and molecularly targeted therapy require molecularly measured biomarkers. This review provides a summary of the scientific validity and (patho)physiological role of nine serum apolipoproteins, Apo(a), ApoB, ApoC-I, ApoC-II, ApoC-III, ApoE and its phenotypes, ApoA-I, ApoA-II, and ApoA-IV, in lipid metabolism, their association with cardiovascular disease, and their potential as cardiovascular risk markers when measured in a multiplex apolipoprotein panel.
Collapse
Affiliation(s)
- Esther Reijnders
- Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Center, Leiden, the Netherlands.
| | - Arnoud van der Laarse
- Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - L Renee Ruhaak
- Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Christa M Cobbaert
- Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
4
|
Oladosu O, Esobi IC, Powell RR, Bruce T, Stamatikos A. Dissecting the Impact of Vascular Smooth Muscle Cell ABCA1 versus ABCG1 Expression on Cholesterol Efflux and Macrophage-like Cell Transdifferentiation: The Role of SR-BI. J Cardiovasc Dev Dis 2023; 10:416. [PMID: 37887863 PMCID: PMC10607678 DOI: 10.3390/jcdd10100416] [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: 08/29/2023] [Revised: 09/20/2023] [Accepted: 09/27/2023] [Indexed: 10/28/2023] Open
Abstract
Cholesterol-laden macrophages are recognized as a major contributor to atherosclerosis. However, recent evidence indicates that vascular smooth muscle cells (VSMC) that accumulate cholesterol and transdifferentiate into a macrophage-like cell (MLC) phenotype also play a role in atherosclerosis. Therefore, removing cholesterol from MLC may be a potential atheroprotective strategy. The two transporters which remove cholesterol from cells are ABCA1 and ABCG1, as they efflux cholesterol to apoAI and HDL, respectively. In this study, the well-characterized immortalized VSMC line MOVAS cells were edited to generate ABCA1- and ABCG1-knockout (KO) MOVAS cell lines. We cholesterol-loaded ABCA1-KO MOVAS cells, ABCG1-KO MOVAS cells, and wild-type MOVAS cells to convert cells into a MLC phenotype. When we measured apoAI- and HDL-mediated cholesterol efflux in these cells, we observed a drastic decrease in apoAI-mediated cholesterol efflux within ABCA1-KO MOVAS MLC, but HDL-mediated cholesterol efflux was only partially reduced in ABCG1-KO MOVAS cells. Since SR-BI also participates in HDL-mediated cholesterol efflux, we assessed SR-BI protein expression in ABCG1-KO MOVAS MLC and observed SR-BI upregulation, which offered a possible mechanism explaining why HDL-mediated cholesterol efflux remains maintained in ABCG1-KO MOVAS MLC. When we used lentivirus for shRNA-mediated knockdown of SR-BI in ABCG1-KO MOVAS MLC, this decreased HDL-mediated cholesterol efflux when compared to ABCG1-KO MOVAS MLC with unmanipulated SR-BI expression. Taken together, these major findings suggest that SR-BI expression in MLC of a VSMC origin plays a compensatory role in HDL-mediated cholesterol efflux when ABCG1 expression becomes impaired and provides insight on SR-BI demonstrating anti-atherogenic properties within VSMC/MLC.
Collapse
Affiliation(s)
- Olanrewaju Oladosu
- Department of Food, Nutrition, and Packaging Sciences, Clemson University, Clemson, SC 29634, USA; (O.O.); (I.C.E.)
| | - Ikechukwu C. Esobi
- Department of Food, Nutrition, and Packaging Sciences, Clemson University, Clemson, SC 29634, USA; (O.O.); (I.C.E.)
| | - Rhonda R. Powell
- Clemson Light Imaging Facility, Clemson University, Clemson, SC 29634, USA; (R.R.P.); (T.B.)
| | - Terri Bruce
- Clemson Light Imaging Facility, Clemson University, Clemson, SC 29634, USA; (R.R.P.); (T.B.)
| | - Alexis Stamatikos
- Department of Food, Nutrition, and Packaging Sciences, Clemson University, Clemson, SC 29634, USA; (O.O.); (I.C.E.)
| |
Collapse
|
5
|
Segrest JP, Davidson WS, Heinecke JW. Phospholipid transport by ABCA1: the extracellular translocase or alternating access model? Curr Opin Lipidol 2023; 34:208-213. [PMID: 37548415 PMCID: PMC10528508 DOI: 10.1097/mol.0000000000000895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
PURPOSE OF REVIEW ATP-binding cassette transporter A1 (ABCA1) plays a key role in high-density lipoprotein (HDL) biogenesis and cholesterol export from artery wall cells. Recent evidence challenges the generally accepted model for lipid transport by ABCA1, termed the alternating access mechanism, which proposes that phospholipid moves from the inner leaflet to the outer leaflet of the plasma membrane. RECENT FINDINGS In contrast to the standard model, our computer simulations of ABCA1 indicate that ABCA1 extracts phospholipid from the plasma membrane's outer leaflet. The lipid then diffuses into the interior of ABCA1 to contact a structure termed the 'gateway'. A conformational change opens the gateway and forces the lipid through a ring-shaped domain, the 'annulus orifice', into the base of an elongated hydrophobic tunnel in the transporter's extracellular domain. Engineered mutations in the gateway and annulus strongly inhibited lipid export by ABCA1 without affecting cell-surface expression levels of the transporter, strongly supporting the proposed model. SUMMARY Our demonstration that ABCA1 extracts lipid from the outer face of the plasma membrane and forces it into an elongated hydrophobic tunnel contrasts with the alternating access model, which flops phospholipid from the membrane's inner leaflet to its outer leaflet. These results suggest that ABCA1 is a phospholipid translocase that transports lipids by a mechanism distinct from that of other ABC transporters.
Collapse
Affiliation(s)
- Jere P. Segrest
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
| | - W. Sean Davidson
- Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati OH 45237
| | - Jay W. Heinecke
- Department of Medicine, University of Washington, Seattle, WA 98109
| |
Collapse
|
6
|
Yang W, Wang S, Zhao Y, Jiang Q, Loor JJ, Tian Y, Fan W, Li M, Zhang B, Cao J, Xu C. Regulation of cholesterol metabolism during high fatty acid-induced lipid deposition in calf hepatocytes. J Dairy Sci 2023:S0022-0302(23)00370-3. [PMID: 37419743 DOI: 10.3168/jds.2022-23136] [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: 12/11/2022] [Accepted: 02/23/2023] [Indexed: 07/09/2023]
Abstract
Cholesterol in the circulation is partly driven by changes in feed intake, but aspects of cholesterol metabolism during development of fatty liver are not well known. The objective of this study was to investigate mechanisms of cholesterol metabolism in calf hepatocytes challenged with high concentrations of fatty acids (FA). To address mechanistic insights regarding cholesterol metabolism, liver samples were collected from healthy control dairy cows (n = 6; 7-13 d in milk) and cows with fatty liver (n = 6; 7-11 d in milk). In vitro, hepatocytes isolated from 3 healthy female calves (1 d old) were challenged with or without a mix of 1.2 mM FA to induce metabolic stress. In addition, hepatocytes were processed with 10 µmol/L of the cholesterol synthesis inhibitor simvastatin or 6 µmol/L of the cholesterol intracellular transport inhibitor U18666A with or without the 1.2 mM FA mix. To evaluate the role of cholesterol addition, hepatocytes were treated with 0.147 mg/mL methyl-β-cyclodextrin (MβCD + FA) or 0.147 mg/mL MβCD with or without 10 and 100 µmol/L cholesterol before incubation with FA (CHO10 + FA and CHO100 + FA). In vivo data from liver biopsies were analyzed by 2-tailed unpaired Student's t-test. Data from in vitro calf hepatocytes were analyzed by one-way ANOVA. Compared with healthy cows, blood plasma total cholesterol and plasma low-density lipoprotein cholesterol content in cows with fatty liver was markedly lower, whereas the hepatic total cholesterol content did not differ. In contrast, compared with healthy controls, the triacylglycerol content in the liver and the content of FA, β-hydroxybutyrate, and aspartate aminotransferase in the plasma of cows with fatty liver were greater. The results revealed that both fatty liver in vivo and challenge of calf hepatocytes with 1.2 mM FA in vitro led to greater mRNA and protein abundance of sterol regulatory element binding transcription factor 1 (SREBF1) and fatty acid synthase (FASN). In contrast, mRNA and protein abundance of sterol regulatory element binding transcription factor 2 (SREBF2), acyl coenzyme A-cholesterol acyltransferase, and ATP-binding cassette subfamily A member 1 (ABCA1) were lower. Compared with the FA group, the cholesterol synthesis inhibitor simvastatin led to greater protein abundance of microsomal triglyceride transfer protein and mRNA abundance of SREBF2, 3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR), ACAT2, and lower ABCA1 and FASN protein abundance. In contrast, compared with the FA group, the cholesterol intracellular transport inhibitor U18666A + FA led to greater total cholesterol concentration and greater protein and mRNA abundance of FASN. Compared with the MβCD + FA group, the addition of 10 µmol/L cholesterol led to greater concentration of cholesteryl ester and excretion of apolipoprotein B100, and greater protein and mRNA abundance of ABCA1 and microsomal triglyceride transfer protein, and lower concentration of malondialdehyde. Overall, a reduction in cholesterol synthesis promoted FA metabolism in hepatocytes likely to relieve the oxidative stress caused by the high FA load. The data suggest that maintenance of normal cholesterol synthesis promotes very low-density lipoprotein excretion and can reduce lipid accumulation and oxidative stress in dairy cows that experience fatty liver.
Collapse
Affiliation(s)
- Wei Yang
- College of Veterinary Medicine, China Agricultural University, Haidian District, Beijing 100193, China; Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Shuang Wang
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Yingying Zhao
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Qianming Jiang
- Mammalian NutriPhysioGenomics, Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana, IL 61801
| | - Juan J Loor
- Mammalian NutriPhysioGenomics, Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana, IL 61801
| | - Yan Tian
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Wenwen Fan
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Ming Li
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Bingbing Zhang
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Jie Cao
- College of Veterinary Medicine, China Agricultural University, Haidian District, Beijing 100193, China.
| | - Chuang Xu
- College of Veterinary Medicine, China Agricultural University, Haidian District, Beijing 100193, China; Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China.
| |
Collapse
|
7
|
Martinez AE, Weissberger G, Kuklenyik Z, He X, Meuret C, Parekh T, Rees JC, Parks BA, Gardner MS, King SM, Collier TS, Harrington MG, Sweeney MD, Wang X, Zlokovic BV, Joe E, Nation DA, Schneider LS, Chui HC, Barr JR, Han SD, Krauss RM, Yassine HN. The small HDL particle hypothesis of Alzheimer's disease. Alzheimers Dement 2023; 19:391-404. [PMID: 35416404 PMCID: PMC10563117 DOI: 10.1002/alz.12649] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 02/08/2022] [Accepted: 02/12/2022] [Indexed: 01/03/2023]
Abstract
We propose the hypothesis that small high-density lipoprotein (HDL) particles reduce the risk of Alzheimer's disease (AD) by virtue of their capacity to exchange lipids, affecting neuronal membrane composition and vascular and synaptic functions. Concentrations of small HDLs in cerebrospinal fluid (CSF) and plasma were measured in 180 individuals ≥60 years of age using ion mobility methodology. Small HDL concentrations in CSF were positively associated with performance in three domains of cognitive function independent of apolipoprotein E (APOE) ε4 status, age, sex, and years of education. Moreover, there was a significant correlation between levels of small HDLs in CSF and plasma. Further studies will be aimed at determining whether specific components of small HDL exchange across the blood, brain, and CSF barriers, and developing approaches to exploit small HDLs for therapeutic purposes.
Collapse
Affiliation(s)
- Ashley E. Martinez
- Department of Medicine, University of Southern California, Los Angeles, California, USA
| | - Gali Weissberger
- The Interdisciplinary Department of Social Sciences, Bar Ilan University, Israel
| | - Zsuzsanna Kuklenyik
- Clinical Chemistry Branch, Division of Laboratory Sciences, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Xulei He
- Department of Medicine, University of Southern California, Los Angeles, California, USA
| | - Cristiana Meuret
- Department of Medicine, University of Southern California, Los Angeles, California, USA
| | - Trusha Parekh
- Department of Medicine, University of Southern California, Los Angeles, California, USA
| | - Jon C. Rees
- Clinical Chemistry Branch, Division of Laboratory Sciences, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Bryan A. Parks
- Clinical Chemistry Branch, Division of Laboratory Sciences, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Michael S. Gardner
- Clinical Chemistry Branch, Division of Laboratory Sciences, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Sarah M. King
- Departments of Pediatrics and Medicine, University of California, San Francisco, California, USA
| | | | - Michael G. Harrington
- Department of Neurology, University of Southern California, Los Angeles, California, USA
| | - Melanie D. Sweeney
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
| | - Xinhui Wang
- Department of Neurology, University of Southern California, Los Angeles, California, USA
| | - Berislav V. Zlokovic
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
| | - Elizabeth Joe
- Department of Neurology, University of Southern California, Los Angeles, California, USA
| | - Daniel A. Nation
- Irvine, Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, California, USA
| | - Lon S. Schneider
- Department of Neurology, University of Southern California, Los Angeles, California, USA
- Department of Psychiatry and Behavioral Sciences, University of Southern California, Los Angeles, California, USA
| | - Helena C. Chui
- Department of Neurology, University of Southern California, Los Angeles, California, USA
| | - John R. Barr
- Clinical Chemistry Branch, Division of Laboratory Sciences, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - S. Duke Han
- Department of Family Medicine, University of Southern California, Los Angeles, California, USA
| | - Ronald M. Krauss
- Departments of Pediatrics and Medicine, University of California, San Francisco, California, USA
| | - Hussein N. Yassine
- Department of Medicine, University of Southern California, Los Angeles, California, USA
- Department of Neurology, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
8
|
Girotti AW, Korytowski W. Trafficking of oxidative stress-generated lipid hydroperoxides: pathophysiological implications. Free Radic Res 2023; 57:130-139. [PMID: 37171212 PMCID: PMC10405667 DOI: 10.1080/10715762.2023.2213817] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/26/2023] [Accepted: 05/05/2023] [Indexed: 05/13/2023]
Abstract
Lipid hydroperoxides (LOOHs) are reactive intermediates that arise during peroxidation of unsaturated phospholipids, glycolipids and cholesterol in biological membranes and lipoproteins. Non-physiological lipid peroxidation (LPO) typically occurs under oxidative stress conditions associated with pathologies such as atherogenesis, neurodegeneration, and carcinogenesis. As key intermediates in the LPO process, LOOHs are susceptible to one-electron versus two-electron reductive turnover, the former exacerbating membrane or lipoprotein damage/dysfunction and the latter diminishing it. A third possible LOOH fate is translocation to an acceptor membrane/lipoprotein, where one- or two-electron reduction may then ensue. In the case of cholesterol (Ch)-derived hydroperoxides (ChOOHs), translocation can be specifically stimulated by StAR family trafficking proteins, which are normally involved in Ch homeostasis and Ch-mediated steroidogenesis. In this review, we discuss how these processes can be impaired by StAR-mediated ChOOH and Ch co-trafficking to mitochondria of vascular macrophages and steroidogenic cells, respectively. The protective effects of endogenous selenoperoxidase, GPx4, are also discussed. This is the first known example of detrimental ChOOH transfer via a natural Ch trafficking pathway and inhibition thereof by GPx4.
Collapse
Affiliation(s)
- Albert W. Girotti
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, USA
| | | |
Collapse
|
9
|
León-Reyes G, Argoty-Pantoja AD, Rivera-Paredez B, Hidalgo-Bravo A, Flores YN, Salmerón J, Velázquez-Cruz R. Interaction between SIDT2 and ABCA1 Variants with Nutrients on HDL-c Levels in Mexican Adults. Nutrients 2023; 15:370. [PMID: 36678241 PMCID: PMC9861312 DOI: 10.3390/nu15020370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 11/09/2022] [Accepted: 11/15/2022] [Indexed: 01/13/2023] Open
Abstract
Previous studies have reported that the SIDT2 and ABCA1 genes are involved in lipid metabolism. We aimed to analyze the association-the gene x gene interaction between rs17120425 and rs1784042 on SIDT2 and rs9282541 on ABCA1 and their diet interaction on the HDL-c serum levels-in a cohort of 1982 Mexican adults from the Health Workers Cohort Study. Demographic and clinical data were collected through a structured questionnaire and standardized procedures. Genotyping was performed using a predesigned TaqMan assay. The associations and interactions of interest were estimated using linear and logistic regression. Carriers of the rs17120425-A and rs1784042-A alleles had slightly higher blood HDL-c levels compared to the non-carriers. In contrast, rs9282541-A was associated with low blood HDL-c levels (OR = 1.34, p = 0.013). The rs1784042 x rs9282541 interaction was associated with high blood HDL-c levels (p = 3.4 × 10-4). Premenopausal women who carried at least one rs17120425-A allele and consumed high dietary fat, protein, monounsaturated, or polyunsaturated fatty acids levels had higher HDL-c levels than the non-carriers. These results support the association between the genetic variants on SIDT2 and ABCA1 with HDL-c levels and suggest gene-gene and gene-diet interactions over HDL-c concentrations in Mexican adults. Our findings could be a platform for developing clinical and dietary strategies for improving the health of the Mexican population.
Collapse
Affiliation(s)
- Guadalupe León-Reyes
- Genomics of Bone Metabolism Laboratory, National Institute of Genomic Medicine (INMEGEN), Mexico City 14610, Morelos, Mexico
| | - Anna D. Argoty-Pantoja
- Research Center in Policies, Population and Health, School of Medicine, National Autonomous University of Mexico (UNAM), Mexico City 04510, Morelos, Mexico
| | - Berenice Rivera-Paredez
- Research Center in Policies, Population and Health, School of Medicine, National Autonomous University of Mexico (UNAM), Mexico City 04510, Morelos, Mexico
| | - Alberto Hidalgo-Bravo
- Department of Genetics, National Institute of Rehabilitation (INR), Mexico City 014389, Morelos, Mexico
| | - Yvonne N. Flores
- Epidemiological and Health Services Research Unit, Morelos, Mexican Institute of Social Security, Cuernavaca 62000, Morelos, Mexico
- Department of Health Policy and Management and Kaiser Permanente Center for Health Equity, Fielding School of Public Health, Los Angeles, University of California, Los Angeles, CA 90095, USA
- Cancer Prevention and Control Research Center, Fielding School of Public Health and Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA 90095, USA
| | - Jorge Salmerón
- Research Center in Policies, Population and Health, School of Medicine, National Autonomous University of Mexico (UNAM), Mexico City 04510, Morelos, Mexico
| | - Rafael Velázquez-Cruz
- Genomics of Bone Metabolism Laboratory, National Institute of Genomic Medicine (INMEGEN), Mexico City 14610, Morelos, Mexico
| |
Collapse
|
10
|
Segrest JP, Tang C, Song HD, Jones MK, Davidson WS, Aller SG, Heinecke JW. ABCA1 is an extracellular phospholipid translocase. Nat Commun 2022; 13:4812. [PMID: 35974019 PMCID: PMC9381790 DOI: 10.1038/s41467-022-32437-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 07/29/2022] [Indexed: 11/18/2022] Open
Abstract
Production of high density lipoprotein (HDL) requires ATP-binding cassette transporter A1 (ABCA1) to drive phospholipid (PL) from the plasma membrane into extracellular apolipoprotein A-I. Here, we use simulations to show that domains of ABCA1 within the plasma membrane remove PL from the membrane’s outer leaflet. In our simulations, after the lipid diffuses into the interior of ABCA1’s outward-open cavity, PL extracted by the gateway passes through a ring-shaped domain, the annulus orifice, which forms the base of an elongated hydrophobic tunnel in the transporter’s extracellular domain. Engineered mutations in the gateway and annulus strongly inhibit lipid export by ABCA1 without affecting cell-surface expression levels. Our finding that ABCA1 extracts lipid from the outer face of the plasma membrane and forces it through its gateway and annulus into an elongated hydrophobic tunnel contrasts with the alternating access model, which proposes that ABCA1 flops PL substrate from the inner leaflet to the outer leaflet of the membrane. Consistent with our model, ABCA1 lacks the charged amino acid residues in the transmembrane domain found in the floppase members of the ABC transporter family. ATP-binding cassette transporter A1 (ABCA1) drives phospholipid (PL) from the plasma membrane into extracellular apolipoprotein A-I, for the production of high density lipoprotein (HDL). Here, the authors use simulations to assess the mechanism of ABCA1 function and show that ABCA1 extracts lipid from the outer face of the plasma membrane.
Collapse
Affiliation(s)
- Jere P Segrest
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Chongren Tang
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Hyun D Song
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Martin K Jones
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - W Sean Davidson
- Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Stephen G Aller
- Department of Pharmacology & Toxicology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jay W Heinecke
- Department of Medicine, University of Washington, Seattle, WA, USA
| |
Collapse
|
11
|
Noveir SD, Kerman BE, Xian H, Meuret C, Smadi S, Martinez AE, Johansson J, Zetterberg H, Parks BA, Kuklenyik Z, Mack WJ, Johansson JO, Yassine HN. Effect of the ABCA1 agonist CS-6253 on amyloid-β and lipoprotein metabolism in cynomolgus monkeys. Alzheimers Res Ther 2022; 14:87. [PMID: 35751102 PMCID: PMC9229758 DOI: 10.1186/s13195-022-01028-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 06/07/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Inducing brain ATP-binding cassette 1 (ABCA1) activity in Alzheimer's disease (AD) mouse models is associated with improvement in AD pathology. The purpose of this study was to investigate the effects of the ABCA1 agonist peptide CS-6253 on amyloid-β peptides (Aβ) and lipoproteins in plasma and cerebrospinal fluid (CSF) of cynomolgus monkeys, a species with amyloid and lipoprotein metabolism similar to humans. METHODS CS-6253 peptide was injected intravenously into cynomolgus monkeys at various doses in three different studies. Plasma and CSF samples were collected at several time points before and after treatment. Levels of cholesterol, triglyceride (TG), lipoprotein particles, apolipoproteins, and Aβ were measured using ELISA, ion-mobility analysis, and asymmetric-flow field-flow fractionation (AF4). The relationship between the change in levels of these biomarkers was analyzed using multiple linear regression models and linear mixed-effects models. RESULTS Following CS-6253 intravenous injection, within minutes, small plasma high-density lipoprotein (HDL) particles were increased. In two independent experiments, plasma TG, apolipoprotein E (apoE), and Aβ42/40 ratio were transiently increased following CS-6253 intravenous injection. This change was associated with a non-significant decrease in CSF Aβ42. Both plasma total cholesterol and HDL-cholesterol levels were reduced following treatment. AF4 fractionation revealed that CS-6253 treatment displaced apoE from HDL to intermediate-density- and low density-lipoprotein (IDL/LDL)-sized particles in plasma. In contrast to plasma, CS-6253 had no effect on the assessed CSF apolipoproteins or lipids. CONCLUSIONS Treatment with the ABCA1 agonist CS-6253 appears to favor Aβ clearance from the brain.
Collapse
Affiliation(s)
- Sasan D Noveir
- Departments of Medicine and Neurology, University of Southern California, Los Angeles, CA, 90033, USA
| | - Bilal E Kerman
- Departments of Medicine and Neurology, University of Southern California, Los Angeles, CA, 90033, USA
| | - Haotian Xian
- Departments of Medicine and Neurology, University of Southern California, Los Angeles, CA, 90033, USA
| | - Cristiana Meuret
- Departments of Medicine and Neurology, University of Southern California, Los Angeles, CA, 90033, USA
| | - Sabrina Smadi
- Departments of Medicine and Neurology, University of Southern California, Los Angeles, CA, 90033, USA
| | - Ashley E Martinez
- Departments of Medicine and Neurology, University of Southern California, Los Angeles, CA, 90033, USA
| | | | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Bryan A Parks
- Centers for Disease Control and Prevention, Atlanta, GA, 30341, USA
| | | | - Wendy J Mack
- Department of Population and Public Health Sciences, University of Southern California, Los Angeles, CA, 90033, USA
| | | | - Hussein N Yassine
- Departments of Medicine and Neurology, University of Southern California, Los Angeles, CA, 90033, USA.
| |
Collapse
|
12
|
Vitamin D Counteracts Lipid Accumulation, Augments Free Fatty Acid-Induced ABCA1 and CPT-1A Expression While Reducing CD36 and C/EBPβ Protein Levels in Monocyte-Derived Macrophages. Biomedicines 2022; 10:biomedicines10040775. [PMID: 35453525 PMCID: PMC9028184 DOI: 10.3390/biomedicines10040775] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/16/2022] [Accepted: 03/25/2022] [Indexed: 12/15/2022] Open
Abstract
The biologically active form of vitamin D, calcitriol (VD3), has received great attention for its extraskeletal effects, such as a protective role on the cardiovascular system. The aim of the present work is to test the capacity of VD3 to affect lipid metabolism and fatty acid accumulation in an in vitro model of monocyte (THP-1)-derived macrophages. Cells were treated for 24 h with oleic/palmitic acid (500 μM, 2:1 ratio) and different VD3 concentrations (0.1, 1, 10, 50 and 100 nM). Lipid accumulation was quantified spectrophotometrically (excitation: 544 nm, emission: 590 nm). C/EBPβ, PPAR-γ1, CD36, CPT-1A, and ABCA1 protein levels were assessed by ELISA kits at different time-points (1, 2, 4, 8, and 24 h). VD3 at 50 and 100 nM significantly reduced fatty acids accumulation in macrophages by 27% and 32%, respectively. In addition, tested at 50 nM, VD3 decreased CD36, PPAR-γ1, and C/EBPβ, while it increased ABCA1 and CPT-1A protein levels in free fatty acid-exposed cells. In conclusion, VD3 reduced fatty acid accumulation in THP-1-derived macrophages exposed to lipid excess. The anti-atherogenic effect of VD3 could be ascribable to the regulation of proteins involved in lipid transport and clearance.
Collapse
|
13
|
Ahmadi A, Bagheri Ekta M, Sahebkar A. Mechanisms of antidiabetic drugs and cholesterol efflux: a clinical perspective. Drug Discov Today 2022; 27:1679-1688. [PMID: 35182734 DOI: 10.1016/j.drudis.2022.02.006] [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: 08/18/2021] [Revised: 01/09/2022] [Accepted: 02/14/2022] [Indexed: 11/26/2022]
Abstract
Reverse cholesterol transport (RCT) is a physiological process that reduces excess cholesterol in the body. Cholesterol efflux (CE), an important step in RCT, is mainly mediated by ATP-binding cassette transporters A1 and G1 and has a significant role in atheroprotection. Moreover, impairments in CE can lead to the development of diabetes and fatty liver disease. In this review, we summarize the possible effects of hypoglycemic agents on CE and how this might influence atherosclerosis and dyslipidemia-related pathologies. Newer antidiabetic agents could have significant potential for targeting CE and preventing or alleviating atherosclerosis, obesity, and liver steatosis, and simultaneously improving insulin secretion. However, more research is warranted to interpret the clinical relevance of these data.
Collapse
Affiliation(s)
- Ali Ahmadi
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad 9177948567, Iran
| | - Mariam Bagheri Ekta
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, A.P. Avtsyn Research Institute of Human Morphology, 3 Tsyurupy Street, 117418 Moscow, Russian Federation
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; School of Medicine, The University of Western Australia, Perth, WA, Australia; Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
14
|
She J, Gu T, Pang X, Liu Y, Tang L, Zhou X. Natural Products Targeting Liver X Receptors or Farnesoid X Receptor. Front Pharmacol 2022; 12:772435. [PMID: 35069197 PMCID: PMC8766425 DOI: 10.3389/fphar.2021.772435] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 11/22/2021] [Indexed: 12/18/2022] Open
Abstract
Nuclear receptors (NRs) are a superfamily of transcription factors induced by ligands and also function as integrators of hormonal and nutritional signals. Among NRs, the liver X receptors (LXRs) and farnesoid X receptor (FXR) have been of significance as targets for the treatment of metabolic syndrome-related diseases. In recent years, natural products targeting LXRs and FXR have received remarkable interests as a valuable source of novel ligands encompassing diverse chemical structures and bioactive properties. This review aims to survey natural products, originating from terrestrial plants and microorganisms, marine organisms, and marine-derived microorganisms, which could influence LXRs and FXR. In the recent two decades (2000-2020), 261 natural products were discovered from natural resources such as LXRs/FXR modulators, 109 agonists and 38 antagonists targeting LXRs, and 72 agonists and 55 antagonists targeting FXR. The docking evaluation of desired natural products targeted LXRs/FXR is finally discussed. This comprehensive overview will provide a reference for future study of novel LXRs and FXR agonists and antagonists to target human diseases, and attract an increasing number of professional scholars majoring in pharmacy and biology with more in-depth discussion.
Collapse
Affiliation(s)
- Jianglian She
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, China.,College of Earth and Planetary Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Tanwei Gu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Xiaoyan Pang
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, China
| | - Yonghong Liu
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, China.,College of Earth and Planetary Sciences, University of Chinese Academy of Sciences, Beijing, China.,Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, China
| | - Lan Tang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Xuefeng Zhou
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, China.,Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, China
| |
Collapse
|
15
|
Loughran R, Emerling BM. Mechanistic roles of mutant p53 governing lipid metabolism. Adv Biol Regul 2022; 83:100839. [PMID: 34840111 PMCID: PMC8858851 DOI: 10.1016/j.jbior.2021.100839] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 11/16/2021] [Indexed: 01/03/2023]
Abstract
Metabolic reprogramming of cancer cells by various acquired mutations provides support for rapid proliferation and growth in the tumor microenvironment. Mutations in the TP53 gene are the most common mutation found across all human cancers. Commonly referred to as "the guardian of the genome", p53 has a well-established role as a tumor suppressor by mediating checkpoint integrity and protecting cells from DNA damage. To date, the many functional roles of p53 extending beyond its classical function and exerting control over metabolic processes continues to confound the field. Recently, emerging roles for p53 in mediating lipid metabolism have come to light with intriguing metabolic roles in regulating cholesterol homeostasis and lipid droplet formation. Herein, we will seek to unify the mechanisms by which absence of functional p53, as well as stable mutant forms of p53, exert control over these lipid metabolism programs. Of equal importance, synthetic lethal phenotypes in the context of mutant p53 and aberrant lipid homeostasis offer new possible targets in the therapeutic landscape. This review aims to characterize the mechanisms by which p53 exerts control over these pathways and examine how precision medicine may benefit from tumor subtyping of p53 mutations.
Collapse
Affiliation(s)
- Ryan Loughran
- Cell and Molecular Biology of Cancer Program, Sanford Burnham Prebys, La Jolla, CA 92037, USA
| | - Brooke M. Emerling
- Cell and Molecular Biology of Cancer Program, Sanford Burnham Prebys, La Jolla, CA 92037, USA,Correspondence:
| |
Collapse
|
16
|
Zheng A, Dubuis G, Georgieva M, Ferreira CSM, Serulla M, Del Carmen Conde Rubio M, Trofimenko E, Mercier T, Decosterd L, Widmann C. HDLs extract lipophilic drugs from cells. J Cell Sci 2021; 135:273878. [PMID: 34981808 PMCID: PMC8919334 DOI: 10.1242/jcs.258644] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 12/14/2021] [Indexed: 11/20/2022] Open
Abstract
High-density lipoproteins (HDLs) prevent cell death induced by a variety of cytotoxic drugs. The underlying mechanisms are however still poorly understood. Here, we present evidence that HDLs efficiently protect cells against thapsigargin (TG), a sarco/endoplasmic reticulum (ER) Ca2+-ATPase (SERCA) inhibitor, by extracting the drug from cells. Drug efflux could also be triggered to some extent by low-density lipoproteins and serum. HDLs did not reverse the non-lethal mild ER stress response induced by low TG concentrations or by SERCA knockdown, but HDLs inhibited the toxic SERCA-independent effects mediated by high TG concentrations. HDLs could extract other lipophilic compounds, but not hydrophilic substances. This work shows that HDLs utilize their capacity of loading themselves with lipophilic compounds, akin to their ability to extract cellular cholesterol, to reduce the cell content of hydrophobic drugs. This can be beneficial if lipophilic xenobiotics are toxic but may be detrimental to the therapeutic benefit of lipophilic drugs such as glibenclamide. Summary: HDLs, akin to their capacity for extracting cholesterol, can remove lipophilic compounds from cells, thus protecting the cells when these compounds are toxic.
Collapse
Affiliation(s)
- Adi Zheng
- Department of Biomedical Sciences, University of Lausanne, Bugnon 7, 1005 Lausanne, Switzerland
| | - Gilles Dubuis
- Department of Biomedical Sciences, University of Lausanne, Bugnon 7, 1005 Lausanne, Switzerland
| | - Maria Georgieva
- Department of Biomedical Sciences, University of Lausanne, Bugnon 7, 1005 Lausanne, Switzerland
| | | | - Marc Serulla
- Department of Biomedical Sciences, University of Lausanne, Bugnon 7, 1005 Lausanne, Switzerland
| | | | - Evgeniya Trofimenko
- Department of Biomedical Sciences, University of Lausanne, Bugnon 7, 1005 Lausanne, Switzerland
| | - Thomas Mercier
- Laboratory of Clinical Pharmacology, Lausanne University Hospital (CHUV) and University of Lausanne, Switzerland
| | - Laurent Decosterd
- Laboratory of Clinical Pharmacology, Lausanne University Hospital (CHUV) and University of Lausanne, Switzerland
| | - Christian Widmann
- Department of Biomedical Sciences, University of Lausanne, Bugnon 7, 1005 Lausanne, Switzerland
| |
Collapse
|
17
|
Jeon S, Kim SH, Jeong J, Lee DK, Lee S, Kim S, Kim G, Maruthupandy M, Cho WS. ABCG1 and ABCG4 as key transporters in the development of pulmonary alveolar proteinosis by nanoparticles. JOURNAL OF HAZARDOUS MATERIALS 2021; 420:126595. [PMID: 34256330 DOI: 10.1016/j.jhazmat.2021.126595] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 06/30/2021] [Accepted: 07/04/2021] [Indexed: 06/13/2023]
Abstract
Pulmonary alveolar proteinosis (PAP) has been reported in rodents treated with nanoparticles (NPs). However, little is known about the type of NPs producing PAP and their toxicity mechanisms. Here, we assembled seven PAP-inducing NPs and TiO2 NPs as a negative control. At 1 and 6 months after a single intratracheal instillation in rats, pulmonary inflammation and the gene expression of ATP-binding cassette (ABC) transporters and related genes were evaluated in separated alveolar macrophages (AMs). One month after intratracheal instillation, seven NPs (Eu2O3, In2O3, Pr6O11, Sm2O3, Tb4O7, and NiO) caused PAP, but only In2O3 NPs caused persistent PAP at 6 months after treatment. The levels of phospholipids, indicators of PAP, showed good correlations with the gene expression profile of five transporters (ABCA1, ABCB4, ABCB8, ABCG1, and ABCG4), which effluxing phospholipids in AMs. Among them, ABCG1 and ABCG4 might be key transporters involved in PAP development because both showed a negative correlation with the magnitude of PAP, while others might be compensatory transporters for PAP recovery, as they showed a positive correlation. In conclusion, the identification of seven PAP-producing NPs implies that PAP may be an emerging occupational disease and that ABCG1 and ABCG4 may be therapeutic targets for PAP.
Collapse
Affiliation(s)
- Soyeon Jeon
- Lab of Toxicology, Department of Health Sciences, The Graduate School of Dong-A University, 37, Nakdong-daero 550 beon-gil Saha-gu, Busan 49315, Republic of Korea
| | - Sung-Hyun Kim
- Lab of Toxicology, Department of Health Sciences, The Graduate School of Dong-A University, 37, Nakdong-daero 550 beon-gil Saha-gu, Busan 49315, Republic of Korea
| | - Jiyoung Jeong
- Lab of Toxicology, Department of Health Sciences, The Graduate School of Dong-A University, 37, Nakdong-daero 550 beon-gil Saha-gu, Busan 49315, Republic of Korea
| | - Dong-Keun Lee
- Lab of Toxicology, Department of Health Sciences, The Graduate School of Dong-A University, 37, Nakdong-daero 550 beon-gil Saha-gu, Busan 49315, Republic of Korea
| | - Sinuk Lee
- Lab of Toxicology, Department of Health Sciences, The Graduate School of Dong-A University, 37, Nakdong-daero 550 beon-gil Saha-gu, Busan 49315, Republic of Korea
| | - Songyeon Kim
- Lab of Toxicology, Department of Health Sciences, The Graduate School of Dong-A University, 37, Nakdong-daero 550 beon-gil Saha-gu, Busan 49315, Republic of Korea
| | - Gyuri Kim
- Lab of Toxicology, Department of Health Sciences, The Graduate School of Dong-A University, 37, Nakdong-daero 550 beon-gil Saha-gu, Busan 49315, Republic of Korea
| | - Muchuchamy Maruthupandy
- Lab of Toxicology, Department of Health Sciences, The Graduate School of Dong-A University, 37, Nakdong-daero 550 beon-gil Saha-gu, Busan 49315, Republic of Korea
| | - Wan-Seob Cho
- Lab of Toxicology, Department of Health Sciences, The Graduate School of Dong-A University, 37, Nakdong-daero 550 beon-gil Saha-gu, Busan 49315, Republic of Korea.
| |
Collapse
|
18
|
Van Valkenburgh J, Meuret C, Martinez AE, Kodancha V, Solomon V, Chen K, Yassine HN. Understanding the Exchange of Systemic HDL Particles Into the Brain and Vascular Cells Has Diagnostic and Therapeutic Implications for Neurodegenerative Diseases. Front Physiol 2021; 12:700847. [PMID: 34552500 PMCID: PMC8450374 DOI: 10.3389/fphys.2021.700847] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 07/29/2021] [Indexed: 12/02/2022] Open
Abstract
High-density lipoproteins (HDLs) are complex, heterogenous lipoprotein particles, consisting of a large family of apolipoproteins, formed in subspecies of distinct shapes, sizes, and functions and are synthesized in both the brain and the periphery. HDL apolipoproteins are important determinants of Alzheimer’s disease (AD) pathology and vascular dementia, having both central and peripheral effects on brain amyloid-beta (Aβ) accumulation and vascular functions, however, the extent to which HDL particles (HLD-P) can exchange their protein and lipid components between the central nervous system (CNS) and the systemic circulation remains unclear. In this review, we delineate how HDL’s structure and composition enable exchange between the brain, cerebrospinal fluid (CSF) compartment, and vascular cells that ultimately affect brain amyloid metabolism and atherosclerosis. Accordingly, we then elucidate how modifications of HDL-P have diagnostic and therapeutic potential for brain vascular and neurodegenerative diseases.
Collapse
Affiliation(s)
- Juno Van Valkenburgh
- Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Cristiana Meuret
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Ashley E Martinez
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Vibha Kodancha
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Victoria Solomon
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Kai Chen
- Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Hussein N Yassine
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
19
|
Girotti AW, Korytowski W. Pathophysiological potential of lipid hydroperoxide intermembrane translocation: Cholesterol hydroperoxide translocation as a special case. Redox Biol 2021; 46:102096. [PMID: 34418596 PMCID: PMC8379493 DOI: 10.1016/j.redox.2021.102096] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 08/04/2021] [Accepted: 08/05/2021] [Indexed: 10/29/2022] Open
Abstract
Peroxidation of unsaturated phospholipids, glycolipids, and cholesterol in biological membranes under oxidative stress conditions can underlie a variety of pathological conditions, including atherogenesis, neurodegeneration, and carcinogenesis. Lipid hydroperoxides (LOOHs) are key intermediates in the peroxidative process. Nascent LOOHs may either undergo one-electron reduction to exacerbate membrane damage/dysfunction or two-electron reduction to attenuate this. Another possibility is LOOH translocation to an acceptor site, followed by either of these competing reductions. Cholesterol (Ch)-derived hydroperoxides (ChOOHs) have several special features that will be highlighted in this review. In addition to being susceptible to one-electron vs. two-electron reduction, ChOOHs can translocate from a membrane of origin to another membrane, where such turnover may ensue. Intracellular StAR family proteins have been shown to deliver not only Ch to mitochondria, but also ChOOHs. StAR-mediated transfer of free radical-generated 7-hydroperoxycholesterol (7-OOH) results in impairment of (a) Ch utilization in steroidogenic cells, and (b) anti-atherogenic reverse Ch transport in vascular macrophages. This is the first known example of how a peroxide derivative can be recognized by a natural lipid trafficking pathway with deleterious consequences. For each example above, we will discuss the underlying mechanism of oxidative damage/dysfunction, and how this might be mitigated by antioxidant intervention.
Collapse
Affiliation(s)
- Albert W Girotti
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, USA.
| | | |
Collapse
|
20
|
Nyandwi JB, Ko YS, Jin H, Yun SP, Park SW, Kim HJ. Rosmarinic Acid Increases Macrophage Cholesterol Efflux through Regulation of ABCA1 and ABCG1 in Different Mechanisms. Int J Mol Sci 2021; 22:8791. [PMID: 34445501 PMCID: PMC8395905 DOI: 10.3390/ijms22168791] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 08/06/2021] [Accepted: 08/13/2021] [Indexed: 01/12/2023] Open
Abstract
Lipid dysregulation in diabetes mellitus escalates endothelial dysfunction, the initial event in the development and progression of diabetic atherosclerosis. In addition, lipid-laden macrophage accumulation in the arterial wall plays a significant role in the pathology of diabetes-associated atherosclerosis. Therefore, inhibition of endothelial dysfunction and enhancement of macrophage cholesterol efflux is the important antiatherogenic mechanism. Rosmarinic acid (RA) possesses beneficial properties, including its anti-inflammatory, antioxidant, antidiabetic and cardioprotective effects. We previously reported that RA effectively inhibits diabetic endothelial dysfunction by inhibiting inflammasome activation in endothelial cells. However, its effect on cholesterol efflux remains unknown. Therefore, in this study, we aimed to assess the effect of RA on cholesterol efflux and its underlying mechanisms in macrophages. RA effectively reduced oxLDL-induced cholesterol contents under high glucose (HG) conditions in macrophages. RA enhanced ATP-binding cassette transporter A1 (ABCA1) and G1 (ABCG1) expression, promoting macrophage cholesterol efflux. Mechanistically, RA differentially regulated ABCA1 expression through JAK2/STAT3, JNK and PKC-p38 and ABCG1 expression through JAK2/STAT3, JNK and PKC-ERK1/2/p38 in macrophages. Moreover, RA primarily stabilized ABCA1 rather than ABCG1 protein levels by impairing protein degradation. These findings suggest RA as a candidate therapeutic to prevent atherosclerotic cardiovascular disease complications related to diabetes by regulating cholesterol efflux in macrophages.
Collapse
Affiliation(s)
- Jean-Baptiste Nyandwi
- Department of Pharmacology, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (J.-B.N.); (Y.S.K.); (H.J.); (S.P.Y.); (S.W.P.)
- Department of Convergence Medical Science (BK21 Plus), Gyeongsang National University, Jinju 52727, Korea
- Department of Pharmacy, School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, Kigali 4285, Rwanda
| | - Young Shin Ko
- Department of Pharmacology, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (J.-B.N.); (Y.S.K.); (H.J.); (S.P.Y.); (S.W.P.)
| | - Hana Jin
- Department of Pharmacology, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (J.-B.N.); (Y.S.K.); (H.J.); (S.P.Y.); (S.W.P.)
| | - Seung Pil Yun
- Department of Pharmacology, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (J.-B.N.); (Y.S.K.); (H.J.); (S.P.Y.); (S.W.P.)
| | - Sang Won Park
- Department of Pharmacology, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (J.-B.N.); (Y.S.K.); (H.J.); (S.P.Y.); (S.W.P.)
- Department of Convergence Medical Science (BK21 Plus), Gyeongsang National University, Jinju 52727, Korea
| | - Hye Jung Kim
- Department of Pharmacology, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (J.-B.N.); (Y.S.K.); (H.J.); (S.P.Y.); (S.W.P.)
- Department of Convergence Medical Science (BK21 Plus), Gyeongsang National University, Jinju 52727, Korea
| |
Collapse
|
21
|
Zhao Y, Qian Y, Sun Z, Shen X, Cai Y, Li L, Wang Z. Role of PI3K in the Progression and Regression of Atherosclerosis. Front Pharmacol 2021; 12:632378. [PMID: 33767629 PMCID: PMC7985550 DOI: 10.3389/fphar.2021.632378] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 01/29/2021] [Indexed: 12/11/2022] Open
Abstract
Phosphatidylinositol 3 kinase (PI3K) is a key molecule in the initiation of signal transduction pathways after the binding of extracellular signals to cell surface receptors. An intracellular kinase, PI3K activates multiple intracellular signaling pathways that affect cell growth, proliferation, migration, secretion, differentiation, transcription and translation. Dysregulation of PI3K activity, and as aberrant PI3K signaling, lead to a broad range of human diseases, such as cancer, immune disorders, diabetes, and cardiovascular diseases. A growing number of studies have shown that PI3K and its signaling pathways play key roles in the pathophysiological process of atherosclerosis. Furthermore, drugs targeting PI3K and its related signaling pathways are promising treatments for atherosclerosis. Therefore, we have reviewed how PI3K, an important regulatory factor, mediates the development of atherosclerosis and how targeting PI3K can be used to prevent and treat atherosclerosis.
Collapse
Affiliation(s)
- Yunyun Zhao
- Department of Pathology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Yongjiang Qian
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Zhen Sun
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Xinyi Shen
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Yaoyao Cai
- Department of Pathology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Lihua Li
- Department of Pathology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Zhongqun Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| |
Collapse
|
22
|
Jiang S, Cai Q, Zhang D, Fan J, Hu S, Venners SA. Effect of ABCG1 gene DNA methylations on the lipid-lowering efficacy of simvastatin. Pharmacogenomics 2020; 22:27-39. [PMID: 33356546 DOI: 10.2217/pgs-2020-0068] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: We investigated the effect of ABCG1 gene DNA methylation in the lipid-lowering efficacy of simvastatin. Materials & methods: An extreme sampling approach was used to select 211 individuals from the top and bottom 15% of adjusted lipid-lowering response residuals to simvastatin after eight consecutive weeks. DNA methylation was measured before treatment by the MethylTarget bisulfite sequencing method. Results: ABCG1_A DNA methylations were negatively associated with baseline high-density lipoprotein cholesterol (HDL-C) and the change in HDL-C after treatment. ABCG1_C methylations were also related to the change in triglyceride and HDL-C. Moreover, mean ABCG1_A and ABCG1_C methylations explain 7.2% of the ΔTC (total cholesterol) and 17.5% of the ΔHDL-C level variability, respectively. Conclusion: DNA methylations at the ABCG1 gene play significant inhibitory effects in the lipid-lowering therapy of simvastatin.
Collapse
Affiliation(s)
- Shanqun Jiang
- School of Life Sciences, Anhui University, Hefei, 230601, China.,Institute of Physical Science & Information Technology, Anhui University, Hefei, 230601, China.,Institute of Biomedicine, Anhui Medical University, Hefei, 230032, China
| | - Qianru Cai
- School of Life Sciences, Anhui University, Hefei, 230601, China
| | - Di Zhang
- School of Life Sciences, Anhui University, Hefei, 230601, China
| | - Juanlin Fan
- School of Life Sciences, Anhui University, Hefei, 230601, China
| | - Shengnan Hu
- School of Life Sciences, Anhui University, Hefei, 230601, China
| | - Scott A Venners
- Faculty of Health Sciences, Simon Fraser University, Burnaby, BC V5A1S6, Canada
| |
Collapse
|
23
|
Castaño D, Rattanasopa C, Monteiro-Cardoso VF, Corlianò M, Liu Y, Zhong S, Rusu M, Liehn EA, Singaraja RR. Lipid efflux mechanisms, relation to disease and potential therapeutic aspects. Adv Drug Deliv Rev 2020; 159:54-93. [PMID: 32423566 DOI: 10.1016/j.addr.2020.04.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 04/29/2020] [Accepted: 04/30/2020] [Indexed: 02/06/2023]
Abstract
Lipids are hydrophobic and amphiphilic molecules involved in diverse functions such as membrane structure, energy metabolism, immunity, and signaling. However, altered intra-cellular lipid levels or composition can lead to metabolic and inflammatory dysfunction, as well as lipotoxicity. Thus, intra-cellular lipid homeostasis is tightly regulated by multiple mechanisms. Since most peripheral cells do not catabolize cholesterol, efflux (extra-cellular transport) of cholesterol is vital for lipid homeostasis. Defective efflux contributes to atherosclerotic plaque development, impaired β-cell insulin secretion, and neuropathology. Of these, defective lipid efflux in macrophages in the arterial walls leading to foam cell and atherosclerotic plaque formation has been the most well studied, likely because a leading global cause of death is cardiovascular disease. Circulating high density lipoprotein particles play critical roles as acceptors of effluxed cellular lipids, suggesting their importance in disease etiology. We review here mechanisms and pathways that modulate lipid efflux, the role of lipid efflux in disease etiology, and therapeutic options aimed at modulating this critical process.
Collapse
|
24
|
Fernández‐Vega B, García M, Olivares L, Álvarez L, González‐Fernández A, Artime E, Fernández‐Vega Cueto A, Cobo T, Coca‐Prados M, Vega JA, González‐Iglesias H. The association study of lipid metabolism gene polymorphisms with AMD identifies a protective role for APOE-E2 allele in the wet form in a Northern Spanish population. Acta Ophthalmol 2020; 98:e282-e291. [PMID: 31654486 DOI: 10.1111/aos.14280] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 09/28/2019] [Indexed: 12/12/2022]
Abstract
PURPOSE To elucidate the potential role of eleven single nucleotide polymorphisms (SNPs) in the most relevant lipid metabolism genes in Northern Spanish patients with age-related macular degeneration (AMD). METHODS A case-control study of 228 unrelated native Northern Spanish patients diagnosed with AMD (73 dry and 155 wet) and 95 healthy controls was performed. DNA was isolated from peripheral blood and genotyped for the SNPs APOE rs429358 and rs7412; CTEP rs3764261; LIPC rs10468017 and rs493258; LPL rs12678919; ABCA1 rs1883025; ABCA4 rs76157638, rs3112831 and rs1800555; and SCARB1 rs5888, using TaqMan probes. An additional association study of ε2, ε3 and ε4 major isoforms of APOE gene with AMD has been carried out. RESULTS The allele and genotype frequencies for each of the eleven sequence variants in the lipid metabolism genes did not show significant differences when comparing AMD cases and controls. Statistical analysis revealed that APOE-ε2 carrier genotypes were less frequently observed in patients with wet AMD compared to controls (5.8% versus 13.7%, respectively: p = 3.28 × 10-2 ; OR = 0.42, 95% CI: 0.19-0.95). The frequency of the allele T of rs10468017 (LIPC gene) was lower in dry AMD cases compared to controls (15.8 versus 27.9%, respectively: p = 8.4 × 10-3 OR = 0.57, 95% CI: 0.33-0.98). CONCLUSIONS Our results suggest a protective role for APOE-ε2 allele to wet AMD in the Northern Spanish population.
Collapse
Affiliation(s)
- Beatriz Fernández‐Vega
- Instituto Oftalmológico Fernández‐Vega Oviedo Spain
- Instituto Universitario Fernández‐Vega (Fundación de Investigación Oftalmológica, Universidad de Oviedo) Oviedo Spain
- Departamento de Morfología y Biología Celular Grupo SINPOS Universidad de Oviedo Oviedo Spain
| | - Montserrat García
- Instituto Oftalmológico Fernández‐Vega Oviedo Spain
- Instituto Universitario Fernández‐Vega (Fundación de Investigación Oftalmológica, Universidad de Oviedo) Oviedo Spain
| | - Lorena Olivares
- Instituto Universitario Fernández‐Vega (Fundación de Investigación Oftalmológica, Universidad de Oviedo) Oviedo Spain
| | - Lydia Álvarez
- Instituto Universitario Fernández‐Vega (Fundación de Investigación Oftalmológica, Universidad de Oviedo) Oviedo Spain
| | - Adrián González‐Fernández
- Instituto Universitario Fernández‐Vega (Fundación de Investigación Oftalmológica, Universidad de Oviedo) Oviedo Spain
| | - Enol Artime
- Instituto Universitario Fernández‐Vega (Fundación de Investigación Oftalmológica, Universidad de Oviedo) Oviedo Spain
| | - Andrés Fernández‐Vega Cueto
- Instituto Oftalmológico Fernández‐Vega Oviedo Spain
- Instituto Universitario Fernández‐Vega (Fundación de Investigación Oftalmológica, Universidad de Oviedo) Oviedo Spain
| | - Teresa Cobo
- Departamento de Cirugía y Especialidades Médico‐Quirúrgicas Universidad de Oviedo Oviedo Spain
| | - Miguel Coca‐Prados
- Department of Ophthalmology and Visual Science Yale University School of Medicine New Haven CT USA
| | - José A. Vega
- Departamento de Morfología y Biología Celular Grupo SINPOS Universidad de Oviedo Oviedo Spain
- Facultad de Ciencias de la Salud Universidad Autónoma de Chile Santiago de Chile Chile
| | - Héctor González‐Iglesias
- Instituto Oftalmológico Fernández‐Vega Oviedo Spain
- Instituto Universitario Fernández‐Vega (Fundación de Investigación Oftalmológica, Universidad de Oviedo) Oviedo Spain
| |
Collapse
|
25
|
Han QA, Su D, Shi C, Liu P, Wang Y, Zhu B, Xia X. Urolithin A attenuated ox-LDL-induced cholesterol accumulation in macrophages partly through regulating miR-33a and ERK/AMPK/SREBP1 signaling pathways. Food Funct 2020; 11:3432-3440. [PMID: 32236173 DOI: 10.1039/c9fo02471a] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Promoting cholesterol efflux from foam cells represents one of the therapeutic strategies for ameliorating atherosclerosis. Urolithin A (UA) has been shown before to attenuate ox-LDL induced endothelial dysfunction in endothelial cells with its anti-inflammatory properties. The aim of this study was to investigate whether UA could promote cholesterol efflux via modulating related microRNA (miR) and signaling pathways. RAW264.7 cells were treated with 50 μg mL-1 ox-LDL to induce foam cell formation. After treatment with UA at different concentrations, intercellular and extracellular cholesterol levels were determined. Expression of Erk1/2, AMPKα and their phosphorylation forms, and SREBP1, was analyzed by western-blotting. The effect of UA on miR-33a expression and the involvement of miR-33a in cholesterol efflux regulation were also investigated. UA reduced ox-LDL induced cholesterol accumulation in macrophage cells and promoted cholesterol efflux from cells. Compared with ox-LDL treated cells, UA treatment reduced the level of phosphorylated ERK1/2, increased the expression of phosphorylated AMPKα and decreased the SREBP1 expression. Moreover, UA decreased the miR-33a expression at the transcriptional level but increased the transcriptional expression of ATP-binding cassette transporter A1 (ABCA1) and ABCG1, two genes contributing to reverse cholesterol transport. Furthermore, pre-miR-33a attenuated cholesterol efflux induced by UA. Collectively, UA promoted the reverse cholesterol transport in macrophage-derived foam cells and interfered with cholesterol metabolism possibly through regulating the miRNA-33 expression and interaction with the ERK/AMPKα/SREBP1 signaling pathway.
Collapse
Affiliation(s)
- Qi-An Han
- Department of Food Quality and Safety, School of Food Science and Technology, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, Liaoning, 116034 China.
| | | | | | | | | | | | | |
Collapse
|
26
|
Mechanisms and regulation of cholesterol homeostasis. Nat Rev Mol Cell Biol 2019; 21:225-245. [DOI: 10.1038/s41580-019-0190-7] [Citation(s) in RCA: 450] [Impact Index Per Article: 90.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/24/2019] [Indexed: 12/14/2022]
|
27
|
Liao S, Zhou Q, Zhang Y. Elastic aortic wrap reduced aortic stiffness by partially alleviating the impairment of cholesterol efflux capacity in pigs. J Diabetes Metab Disord 2019; 17:101-109. [PMID: 30918842 DOI: 10.1007/s40200-018-0345-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 07/17/2018] [Indexed: 12/11/2022]
Abstract
Purpose Metabolic syndrome patients exhibit impaired cholesterol efflux capacity. Previous studies have shown a positive association between aortic stiffness and metabolic syndrome. However, it is unknown whether cholesterol efflux capacity participates in the process of aortic stiffness. This study sought to determine the effect of metabolic syndrome on aortic stiffening, and to investigate the effectiveness of aortic wraps on aortic compliance and the underlying mechanisms. Methods In a swine model of metabolic syndrome, we compared the cholesterol efflux capacity and aortic compliance responding to diet modifications and aortic wrap applications. Results Metabolic syndrome induced by high cholesterol diet significantly decreased cholesterol efflux capacity and aortic compliance. Elastic aortic wrap application increased aortic compliance and partially restored cholesterol efflux capacity via ATP binding cassette transporter A1 (ABCA1) pathway. Conclusions Cholesterol efflux plays a role in aortic stiffening. Elastic aortic wrap application could be a potential treatment for aortic stiffness related to metabolic syndrome.
Collapse
Affiliation(s)
- Shutan Liao
- 1Rural Clinical School, University of New South Wales, Sydney, NSW Australia.,2The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Qing Zhou
- 3Department of Cardiac Surgery, The First Affiliated Hospital of Nanchang University, 53 Taohua Road, Nanchang, 330008 Jiangxi China
| | - Yang Zhang
- 3Department of Cardiac Surgery, The First Affiliated Hospital of Nanchang University, 53 Taohua Road, Nanchang, 330008 Jiangxi China
| |
Collapse
|
28
|
Storti F, Klee K, Todorova V, Steiner R, Othman A, van der Velde-Visser S, Samardzija M, Meneau I, Barben M, Karademir D, Pauzuolyte V, Boye SL, Blaser F, Ullmer C, Dunaief JL, Hornemann T, Rohrer L, den Hollander A, von Eckardstein A, Fingerle J, Maugeais C, Grimm C. Impaired ABCA1/ABCG1-mediated lipid efflux in the mouse retinal pigment epithelium (RPE) leads to retinal degeneration. eLife 2019; 8:45100. [PMID: 30864945 PMCID: PMC6435327 DOI: 10.7554/elife.45100] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 03/12/2019] [Indexed: 01/04/2023] Open
Abstract
Age-related macular degeneration (AMD) is a progressive disease of the retinal pigment epithelium (RPE) and the retina leading to loss of central vision. Polymorphisms in genes involved in lipid metabolism, including the ATP-binding cassette transporter A1 (ABCA1), have been associated with AMD risk. However, the significance of retinal lipid handling for AMD pathogenesis remains elusive. Here, we study the contribution of lipid efflux in the RPE by generating a mouse model lacking ABCA1 and its partner ABCG1 specifically in this layer. Mutant mice show lipid accumulation in the RPE, reduced RPE and retinal function, retinal inflammation and RPE/photoreceptor degeneration. Data from human cell lines indicate that the ABCA1 AMD risk-conferring allele decreases ABCA1 expression, identifying the potential molecular cause that underlies the genetic risk for AMD. Our results highlight the essential homeostatic role for lipid efflux in the RPE and suggest a pathogenic contribution of reduced ABCA1 function to AMD.
Collapse
Affiliation(s)
- Federica Storti
- Lab for Retinal Cell Biology, Department of Ophthalmology, University of Zurich, Schlieren, Switzerland
| | - Katrin Klee
- Lab for Retinal Cell Biology, Department of Ophthalmology, University of Zurich, Schlieren, Switzerland.,Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Vyara Todorova
- Lab for Retinal Cell Biology, Department of Ophthalmology, University of Zurich, Schlieren, Switzerland.,Neuroscience Center Zurich, University of Zurich, Zurich, Switzerland
| | - Regula Steiner
- Institute of Clinical Chemistry, University of Zurich, Schlieren, Switzerland
| | - Alaa Othman
- Institute of Clinical Chemistry, University of Zurich, Schlieren, Switzerland
| | | | - Marijana Samardzija
- Lab for Retinal Cell Biology, Department of Ophthalmology, University of Zurich, Schlieren, Switzerland
| | - Isabelle Meneau
- Department of Ophthalmology, University Hospital Zurich, Zurich, Switzerland
| | - Maya Barben
- Lab for Retinal Cell Biology, Department of Ophthalmology, University of Zurich, Schlieren, Switzerland
| | - Duygu Karademir
- Lab for Retinal Cell Biology, Department of Ophthalmology, University of Zurich, Schlieren, Switzerland.,Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Valda Pauzuolyte
- Lab for Retinal Cell Biology, Department of Ophthalmology, University of Zurich, Schlieren, Switzerland
| | - Sanford L Boye
- Department of Ophthalmology, University of Florida, Gainesville, United States
| | - Frank Blaser
- Department of Ophthalmology, University Hospital Zurich, Zurich, Switzerland
| | - Christoph Ullmer
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Joshua L Dunaief
- Department of Ophthalmology, Scheie Eye Institute, University of Pennsylvania, Philadelphia, United States
| | - Thorsten Hornemann
- Institute of Clinical Chemistry, University of Zurich, Schlieren, Switzerland
| | - Lucia Rohrer
- Institute of Clinical Chemistry, University of Zurich, Schlieren, Switzerland
| | - Anneke den Hollander
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, Netherlands.,Department of Ophthalmology, Radboud University Medical Center, Nijmegen, Netherlands
| | | | - Jürgen Fingerle
- Natural and Medical Sciences Institute, University of Tübingen, Tübingen, Germany
| | - Cyrille Maugeais
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Christian Grimm
- Lab for Retinal Cell Biology, Department of Ophthalmology, University of Zurich, Schlieren, Switzerland.,Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland.,Neuroscience Center Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
29
|
Label-free quantification of protein expression in the rainbow trout (Oncorhynchus mykiss) in response to short-term exposure to heat stress. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY D-GENOMICS & PROTEOMICS 2019; 30:158-168. [PMID: 30851505 DOI: 10.1016/j.cbd.2019.02.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 01/21/2019] [Accepted: 02/26/2019] [Indexed: 12/25/2022]
Abstract
Rainbow trout (Oncorhynchus mykiss) are a cold-water salmonid species that is highly susceptible to heat stress. Summer temperature stress is a common issue in trout aquaculture. To better understand the molecular mechanisms of the heat-stress response in the trout, we used label-free quantitative proteome techniques to identify differentially expressed proteins in the livers of rainbow trout exposed to heat stress. We identified 3362 proteins and 152 differentially expressed proteins (p < 0.05; fold-change >2). Of these, 37 were uniquely expressed in the heat-stress group and 35 were uniquely expressed in the control group. In addition, 42 proteins were significantly upregulated (fold-change >2) and 38 proteins were significantly downregulated (fold-change >2). GO (Gene Ontology) analysis indicated that these differentially expressed proteins were primarily expressed in the nucleus, extracellular matrix, and cytoplasm, and were associated with a variety of functions, including protein binding/bridging and enzyme facilitation. KEGG (Kyoto Encyclopedia of Genes and Genomes) analysis of the differentially expressed proteins showed that, during high temperature stress, many biological processes were extensively altered, particularly the estrogen signaling pathway, the complement and coagulation cascades, and the platelet activation pathway. Our study focused on the identification of a systematic approach for the characterization of regulatory networks. Our results provide a framework for further studies of the heat-stress response in fish.
Collapse
|
30
|
Vaidya M, Jentsch JA, Peters S, Keul P, Weske S, Gräler MH, Mladenov E, Iliakis G, Heusch G, Levkau B. Regulation of ABCA1-mediated cholesterol efflux by sphingosine-1-phosphate signaling in macrophages. J Lipid Res 2019; 60:506-515. [PMID: 30655318 DOI: 10.1194/jlr.m088443] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 01/16/2019] [Indexed: 12/21/2022] Open
Abstract
Sphingolipid and cholesterol metabolism are closely associated at the structural, biochemical, and functional levels. Although HDL-associated sphingosine-1-phosphate (S1P) contributes to several HDL functions, and S1P signaling regulates glucose and lipid metabolism, no study has addressed the involvement of S1P in cholesterol efflux. Here, we show that sphingosine kinase (Sphk) activity was induced by the LXR agonist 22(R)-hydroxycholesterol and required for the stimulation of ABCA1-mediated cholesterol efflux to apolipoprotein A-I. In support, pharmacological Sphk inhibition and Sphk2 but not Sphk1 deficiency abrogated efflux. The involved mechanism included stimulation of both transcriptional and functional ABCA1 regulatory pathways and depended for the latter on the S1P receptor 3 (S1P3). Accordingly, S1P3-deficient macrophages were resistant to 22(R)-hydroxycholesterol-stimulated cholesterol efflux. The inability of excess exogenous S1P to further increase efflux was consistent with tonic S1P3 signaling by a pool of constitutively generated Sphk-derived S1P dynamically regulating cholesterol efflux. In summary, we have established S1P as a previously unrecognized intermediate in LXR-stimulated ABCA1-mediated cholesterol efflux and identified S1P/S1P3 signaling as a positive-feedback regulator of cholesterol efflux. This constitutes a novel regulatory mechanism of cholesterol efflux by sphingolipids.
Collapse
Affiliation(s)
- Mithila Vaidya
- Institute for Pathophysiology, University of Duisburg-Essen, Duisburg, Germany.,West German Heart and Vascular Center University of Duisburg-Essen, Duisburg, Germany
| | - Julian A Jentsch
- Institute for Pathophysiology, University of Duisburg-Essen, Duisburg, Germany.,West German Heart and Vascular Center University of Duisburg-Essen, Duisburg, Germany
| | - Susann Peters
- Institute for Pathophysiology, University of Duisburg-Essen, Duisburg, Germany.,West German Heart and Vascular Center University of Duisburg-Essen, Duisburg, Germany
| | - Petra Keul
- Institute for Pathophysiology, University of Duisburg-Essen, Duisburg, Germany.,West German Heart and Vascular Center University of Duisburg-Essen, Duisburg, Germany
| | - Sarah Weske
- Institute for Pathophysiology, University of Duisburg-Essen, Duisburg, Germany.,West German Heart and Vascular Center University of Duisburg-Essen, Duisburg, Germany
| | - Markus H Gräler
- Department of Anesthesiology and Intensive Care Medicine University Hospital Jena, Jena, Germany.,Center for Sepsis Control and Care, University Hospital Jena, Jena, Germany.,Center for Molecular Biomedicine University Hospital Jena, Jena, Germany
| | - Emil Mladenov
- Institute of Medical Radiation Biology University Hospital Essen, University of Duisburg-Essen, Duisburg, Germany
| | - George Iliakis
- Institute of Medical Radiation Biology University Hospital Essen, University of Duisburg-Essen, Duisburg, Germany
| | - Gerd Heusch
- Institute for Pathophysiology, University of Duisburg-Essen, Duisburg, Germany.,West German Heart and Vascular Center University of Duisburg-Essen, Duisburg, Germany
| | - Bodo Levkau
- Institute for Pathophysiology, University of Duisburg-Essen, Duisburg, Germany .,West German Heart and Vascular Center University of Duisburg-Essen, Duisburg, Germany
| |
Collapse
|
31
|
Roizenblatt M, Naranjit N, Maia M, Gehlbach PL. The Question of a Role for Statins in Age-Related Macular Degeneration. Int J Mol Sci 2018; 19:ijms19113688. [PMID: 30469381 PMCID: PMC6274767 DOI: 10.3390/ijms19113688] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 11/18/2018] [Accepted: 11/19/2018] [Indexed: 01/22/2023] Open
Abstract
Age-related macular degeneration (AMD) is the leading cause of irreversible central vision loss in patients over the age of 65 years in industrialized countries. Epidemiologic studies suggest that high dietary fat intake is a risk factor for the development and progression of both vascular and retinal disease. These, and other associations, suggest a hypothesis linking elevated cholesterol and AMD progression. It follows, therefore, that cholesterol-lowering medications, such as statins, may influence the onset and progression of AMD. However, the findings have been inconclusive as to whether statins play a role in AMD. Due to the significant public health implications of a potential inhibitory effect of statins on the onset and progression of AMD, it is important to continually evaluate emerging findings germane to this question.
Collapse
Affiliation(s)
- Marina Roizenblatt
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
- Department of Ophthalmology, Federal University of São Paulo, São Paulo 04023-062, Brazil.
- Vision Institute, IPEPO, Department of Ophthalmology, Paulista Medical School, Federal University of São Paulo, São Paulo 04038-032, Brazil.
| | - Nara Naranjit
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| | - Mauricio Maia
- Department of Ophthalmology, Federal University of São Paulo, São Paulo 04023-062, Brazil.
- Vision Institute, IPEPO, Department of Ophthalmology, Paulista Medical School, Federal University of São Paulo, São Paulo 04038-032, Brazil.
| | - Peter L Gehlbach
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| |
Collapse
|
32
|
Heo W, Lee ES, Cho HT, Kim JH, Lee JH, Yoon SM, Kwon HT, Yang S, Kim YJ. Lactobacillus plantarum LRCC 5273 isolated from Kimchi ameliorates diet-induced hypercholesterolemia in C57BL/6 mice. Biosci Biotechnol Biochem 2018; 82:1964-1972. [DOI: 10.1080/09168451.2018.1497939] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
ABSTRACT
This study was designed to select potent cholesterol-lowering probiotic strains on HepG2 cell and investigate the effect of selected strain, Lactobacillus plantarum LRCC 5273 and LRCC 5279 in hypercholesterolemic mice. In the results, LP5273 group showed significantly reduced total and LDL cholesterol compared to HCD group. In addition to significantly up-regulated hepatic mRNA expression of LXR-α and CYP7A1, intestinal LXR-α and ABCG5 were significantly up-regulated in LP5273 group. With activation of hepatic and intestinal LXR-α and its target genes, fecal cholesterol and bile acid excretion were increased in LP5273 fed mice. These results suggest that LP5273 ameliorates hypercholesterolemia in mice through the activation of hepatic and intestinal LXR-α, resulting in enhancement of fecal cholesterol and bile acids excretion in the small intestine. The results of present study suggest mechanistic evidences for hypocholesterolemic effects of L. plantarum spp., and may contribute to future researches for prevention of hypercholesterolemia and cardiovascular disease.
Collapse
Affiliation(s)
- Wan Heo
- Department of Food and Biotechnology, Korea University, Sejong, Korea
| | - Eui Seop Lee
- Department of Food and Biotechnology, Korea University, Sejong, Korea
| | - Hyung Taek Cho
- Department of Food and Biotechnology, Korea University, Sejong, Korea
| | - Jun Ho Kim
- Department of Food Science and Biotechnology, Andong National University, Andong, Korea
| | - Jin Hyup Lee
- Department of Food and Biotechnology, Korea University, Sejong, Korea
| | | | | | | | - Young-Jun Kim
- Department of Food and Biotechnology, Korea University, Sejong, Korea
| |
Collapse
|
33
|
Impairment of trophoblast survival and differentiation by LXR ligands is prevented by cholesterol but not ABCA1 silencing. Placenta 2018; 69:50-56. [DOI: 10.1016/j.placenta.2018.07.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 07/02/2018] [Accepted: 07/06/2018] [Indexed: 01/31/2023]
|
34
|
Statin as a novel pharmacotherapy of pulmonary alveolar proteinosis. Nat Commun 2018; 9:3127. [PMID: 30087322 PMCID: PMC6081448 DOI: 10.1038/s41467-018-05491-z] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 06/22/2018] [Indexed: 12/20/2022] Open
Abstract
Pulmonary alveolar proteinosis (PAP) is a syndrome of reduced GM-CSF-dependent, macrophage-mediated surfactant clearance, dysfunctional foamy alveolar macrophages, alveolar surfactant accumulation, and hypoxemic respiratory failure for which the pathogenetic mechanism is unknown. Here, we examine the lipids accumulating in alveolar macrophages and surfactant to define the pathogenesis of PAP and evaluate a novel pharmacotherapeutic approach. In PAP patients, alveolar macrophages have a marked increase in cholesterol but only a minor increase in phospholipids, and pulmonary surfactant has an increase in the ratio of cholesterol to phospholipids. Oral statin therapy is associated with clinical, physiological, and radiological improvement in autoimmune PAP patients, and ex vivo statin treatment reduces cholesterol levels in explanted alveolar macrophages. In Csf2rb−/− mice, statin therapy reduces cholesterol accumulation in alveolar macrophages and ameliorates PAP, and ex vivo statin treatment increases cholesterol efflux from macrophages. These results support the feasibility of statin as a novel pathogenesis-based pharmacotherapy of PAP. Pulmonary alveolar proteinosis (PAP) is associated with defective macrophage clearance of surfactant. Here, the authors show that patients with PAP have altered cholesterol-to-phospholipid ratio in their surfactant, and that more importantly, statin therapy and reduction of cholesterol accumulation in macrophages can ameliorate PAP in both humans and mice.
Collapse
|
35
|
Zanoni P, Velagapudi S, Yalcinkaya M, Rohrer L, von Eckardstein A. Endocytosis of lipoproteins. Atherosclerosis 2018; 275:273-295. [PMID: 29980055 DOI: 10.1016/j.atherosclerosis.2018.06.881] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 06/04/2018] [Accepted: 06/22/2018] [Indexed: 02/06/2023]
Abstract
During their metabolism, all lipoproteins undergo endocytosis, either to be degraded intracellularly, for example in hepatocytes or macrophages, or to be re-secreted, for example in the course of transcytosis by endothelial cells. Moreover, there are several examples of internalized lipoproteins sequestered intracellularly, possibly to exert intracellular functions, for example the cytolysis of trypanosoma. Endocytosis and the subsequent intracellular itinerary of lipoproteins hence are key areas for understanding the regulation of plasma lipid levels as well as the biological functions of lipoproteins. Indeed, the identification of the low-density lipoprotein (LDL)-receptor and the unraveling of its transcriptional regulation led to the elucidation of familial hypercholesterolemia as well as to the development of statins, the most successful therapeutics for lowering of cholesterol levels and risk of atherosclerotic cardiovascular diseases. Novel limiting factors of intracellular trafficking of LDL and the LDL receptor continue to be discovered and to provide drug targets such as PCSK9. Surprisingly, the receptors mediating endocytosis of high-density lipoproteins or lipoprotein(a) are still a matter of controversy or even new discovery. Finally, the receptors and mechanisms, which mediate the uptake of lipoproteins into non-degrading intracellular itineraries for re-secretion (transcytosis, retroendocytosis), storage, or execution of intracellular functions, are largely unknown.
Collapse
Affiliation(s)
- Paolo Zanoni
- Institute for Clinical Chemistry, University and University Hospital Zurich, Zurich, Switzerland; Centre for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Srividya Velagapudi
- Institute for Clinical Chemistry, University and University Hospital Zurich, Zurich, Switzerland; Centre for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Mustafa Yalcinkaya
- Institute for Clinical Chemistry, University and University Hospital Zurich, Zurich, Switzerland; Centre for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Lucia Rohrer
- Institute for Clinical Chemistry, University and University Hospital Zurich, Zurich, Switzerland; Centre for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Arnold von Eckardstein
- Institute for Clinical Chemistry, University and University Hospital Zurich, Zurich, Switzerland; Centre for Integrative Human Physiology, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
36
|
Hu J, Luo T, Xi D, Guo K, Hu L, Zhao J, Chen S, Guo Z. Silencing ZAP70 prevents HSP65-induced reverse cholesterol transport and NF-κB activation in T cells. Biomed Pharmacother 2018; 102:271-277. [DOI: 10.1016/j.biopha.2018.03.082] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Revised: 03/14/2018] [Accepted: 03/14/2018] [Indexed: 11/30/2022] Open
|
37
|
Sun Y, Kopp S, Strutz J, Gali CC, Zandl-Lang M, Fanaee-Danesh E, Kirsch A, Cvitic S, Frank S, Saffery R, Björkhem I, Desoye G, Wadsack C, Panzenboeck U. Gestational diabetes mellitus modulates cholesterol homeostasis in human fetoplacental endothelium. Biochim Biophys Acta Mol Cell Biol Lipids 2018; 1863:968-979. [PMID: 29778664 DOI: 10.1016/j.bbalip.2018.05.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 05/07/2018] [Accepted: 05/15/2018] [Indexed: 02/06/2023]
Abstract
Gestational diabetes mellitus (GDM) is associated with excessive oxidative stress which may affect placental vascular function. Cholesterol homeostasis is crucial for maintaining fetoplacental endothelial function. We aimed to investigate whether and how GDM affects cholesterol metabolism in human fetoplacental endothelial cells (HPEC). HPEC were isolated from fetal term placental arterial vessels of GDM or control subjects. Cellular reactive oxygen species (ROS) were detected by H2DCFDA fluorescent dye. Oxysterols were quantified by gas chromatography-mass spectrometry analysis. Genes and proteins involved in cholesterol homeostasis were detected by real-time PCR and immunoblotting, respectively. Cholesterol efflux was determined from [3H]-cholesterol labeled HPEC and [14C]-acetate was used as cholesterol precursor to measure cholesterol biosynthesis and esterification. We detected enhanced formation of ROS and of specific, ROS-derived oxysterols in HPEC isolated from GDM versus control pregnancies. ROS-generated oxysterols were simultaneously elevated in cord blood of GDM neonates. Liver-X receptor activation in control HPEC by synthetic agonist TO901319, 7-ketocholesterol, or 7β-hydroxycholesterol upregulated ATP-binding cassette transporters (ABC)A1 and ABCG1 expression, accompanied by increased cellular cholesterol efflux. Upregulation of ABCA1 and ABCG1 and increased cholesterol release to apoA-I and HDL3 (78 ± 17%, 40 ± 9%, respectively) were also observed in GDM versus control HPEC. The LXR antagonist GGPP reversed ABCA1 and ABCG1 upregulation and reduced the increased cholesterol efflux in GDM HPEC. Similar total cellular cholesterol levels were detected in control and GDM HPEC, while GDM enhanced cholesterol biosynthesis along with upregulated 3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR) and sterol O-acyltransferase 1 (SOAT1) mRNA and protein levels. Our results suggest that in GDM cellular cholesterol homeostasis in the fetoplacental endothelium is modulated via LXR activation and helps to maintain its proper functionality.
Collapse
Affiliation(s)
- Yidan Sun
- Immunology and Pathophysiology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Austria
| | - Susanne Kopp
- Department of Obstetrics and Gynecology, Medical University of Graz, Austria
| | - Jasmin Strutz
- Department of Obstetrics and Gynecology, Medical University of Graz, Austria
| | - Chaitanya Chakravarthi Gali
- Immunology and Pathophysiology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Austria
| | - Martina Zandl-Lang
- Immunology and Pathophysiology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Austria
| | - Elham Fanaee-Danesh
- Immunology and Pathophysiology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Austria
| | - Andrijana Kirsch
- Institute of Molecular Biology and Biochemistry, Medical University of Graz, Austria
| | - Silvija Cvitic
- Department of Obstetrics and Gynecology, Medical University of Graz, Austria
| | - Saša Frank
- Institute of Molecular Biology and Biochemistry, Medical University of Graz, Austria
| | - Richard Saffery
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Australia; University of Melbourne, Department of Pediatrics, Melbourne, Australia
| | - Ingemar Björkhem
- Division of Clinical Chemistry, Karolinska Institute, Huddinge University Hospital, Sweden
| | - Gernot Desoye
- Department of Obstetrics and Gynecology, Medical University of Graz, Austria
| | - Christian Wadsack
- Department of Obstetrics and Gynecology, Medical University of Graz, Austria; BioTechMed-Graz, Graz, Austria.
| | - Ute Panzenboeck
- Immunology and Pathophysiology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Austria.
| |
Collapse
|
38
|
Marques LR, Diniz TA, Antunes BM, Rossi FE, Caperuto EC, Lira FS, Gonçalves DC. Reverse Cholesterol Transport: Molecular Mechanisms and the Non-medical Approach to Enhance HDL Cholesterol. Front Physiol 2018; 9:526. [PMID: 29867567 PMCID: PMC5962737 DOI: 10.3389/fphys.2018.00526] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 04/24/2018] [Indexed: 11/16/2022] Open
Abstract
Dyslipidemia (high concentrations of LDL-c and low concentrations of HDL-c) is a major cause of cardiovascular events, which are the leading cause of death in the world. On the other hand, nutrition and regular exercise can be an interesting strategy to modulate lipid profile, acting as prevention or treatment, inhibiting the risk of diseases due to its anti-inflammatory and anti-atherogenic characteristics. Additionally, the possibility of controlling different training variables, such as type, intensity and recovery interval, can be used to maximize the benefits of exercise in promoting cardiovascular health. However, the mechanisms by which exercise and nutrients act in the regulation of cholesterol and its fractions, such as reverse cholesterol transport, receptors and transcription factors involved, such as PPARs and their role related to exercise, deserve further discussion. Therefore, the objective of this review is to debate about non-medical approaches to increase HDL-c, such as nutritional and training strategies, and to discuss the central mechanisms involved in the modulation of lipid profile during exercise, as well as that can be controlled by physical trainers or sports specialists in attempt to maximize the benefits promoted by exercise. The search for papers was performed in the databases: Medline (Pubmed), Science Direct, Scopus, Sport Discus, Web of Science, Scielo and Lilacs until February 2016.
Collapse
Affiliation(s)
- Leandro R Marques
- Exercise and Immunometabolism Research Group, Department of Physical Education, Universidade Estadual Paulista, Presidente Prudente, Brazil
| | - Tiego A Diniz
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Barbara M Antunes
- Exercise and Immunometabolism Research Group, Department of Physical Education, Universidade Estadual Paulista, Presidente Prudente, Brazil
| | - Fabrício E Rossi
- Immunometabolism of Skeletal Muscle and Exercise Research Group, Department of Physical Education, Federal University of Piauí, Teresina, Brazil
| | - Erico C Caperuto
- Human Movement Laboratory, Universidade São Judas Tadeu, São Paulo, Brazil
| | - Fábio S Lira
- Exercise and Immunometabolism Research Group, Department of Physical Education, Universidade Estadual Paulista, Presidente Prudente, Brazil
| | - Daniela C Gonçalves
- Department of Biosciences, Universidade Federal de São Paulo, Santos, Brazil
| |
Collapse
|
39
|
Liu B, He Z, Wang J, Xin Z, Wang J, Li F, Fu Y. Taraxasterol Inhibits LPS-Induced Inflammatory Response in BV2 Microglia Cells by Activating LXRα. Front Pharmacol 2018; 9:278. [PMID: 29670526 PMCID: PMC5893773 DOI: 10.3389/fphar.2018.00278] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Accepted: 03/12/2018] [Indexed: 12/20/2022] Open
Abstract
Neuroinflammation plays a critical role in the development of neurodegenerative diseases. Taraxasterol, a pentacyclic-triterpene isolated from Taraxacum officinale, has been reported to have anti-inflammatory effect. The aim of this study was to investigate the anti-inflammatory effects and mechanism of taraxasterol in LPS-stimulated BV2 microglia cells. BV2 microglia cells were treated with taraxasterol 12 h before LPS stimulation. The effects of taraxasterol on LPS-induced TNF-α and IL-1β production were detected by ELISA. The effects of taraxasterol on LXRα, ABCA1, TLR4, and NF-κB expression were detected by western blot analysis. The results showed that taraxasterol dose-dependently inhibited LPS-induced TNF-α and IL-1β production and NF-κB activation. Taraxasterol also disrupted the formation of lipid rafts and inhibited translocation of TLR4 into lipid rafts. Furthermore, taraxasterol was found to activate LXRα-ABCA1 signaling pathway which induces cholesterol efflux from cells. In addition, our results showed that the anti-inflammatory effect of taraxasterol was attenuated by transfection with LXRα siRNA. In conclusion, these results suggested that taraxasterol inhibits LPS-induced inflammatory response in BV2 microglia cells by activating LXRα-ABCA1 signaling pathway.
Collapse
Affiliation(s)
- Bin Liu
- Cardiovascular Disease Center, First Hospital of Jilin University, Changchun, China
| | - Zhaoqi He
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Jingjing Wang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Zhuoyuan Xin
- Department of Pathogenobiology, The Key Laboratory of Zoonosis, Chinese Ministry of Education, College of Basic Medicine, Jilin University, Changchun, China
| | - Jiaxin Wang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Fan Li
- Department of Pathogenobiology, The Key Laboratory of Zoonosis, Chinese Ministry of Education, College of Basic Medicine, Jilin University, Changchun, China
| | - Yunhe Fu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, China.,Department of Pathogenobiology, The Key Laboratory of Zoonosis, Chinese Ministry of Education, College of Basic Medicine, Jilin University, Changchun, China
| |
Collapse
|
40
|
Abstract
Macrophages are critical to organ structure and function in health and disease. To determine mechanisms by which granulocyte/macrophage-colony stimulating factor (GM-CSF) signaling normally maintains surfactant homeostasis and how its disruption causes pulmonary alveolar proteinosis (PAP), we evaluated lipid composition in alveolar macrophages and lung surfactant, macrophage-mediated surfactant clearance kinetics/dynamics, and cholesterol-targeted pharmacotherapy of PAP in vitro and in vivo. Without GM-CSF signaling, surfactant-exposed macrophages massively accumulated cholesterol ester-rich lipid-droplets and surfactant had an increased proportion of cholesterol. GM-CSF regulated cholesterol clearance in macrophages in constitutive, dose-dependent, and reversible fashion but did not affect phospholipid clearance. PPARγ-agonist therapy increased cholesterol clearance in macrophages and reduced disease severity in PAP mice. Results demonstrate that GM-CSF is required for cholesterol clearance in macrophages, identify reduced cholesterol clearance as the primary macrophage defect driving PAP pathogenesis, and support the feasibility of translating pioglitazone as a novel pharmacotherapy of PAP.
Collapse
|
41
|
Monzel JV, Budde T, Meyer Zu Schwabedissen HE, Schwebe M, Bien-Möller S, Lütjohann D, Kroemer HK, Jedlitschky G, Grube M. Doxorubicin enhances oxysterol levels resulting in a LXR-mediated upregulation of cardiac cholesterol transporters. Biochem Pharmacol 2017; 144:108-119. [PMID: 28807695 DOI: 10.1016/j.bcp.2017.08.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 08/09/2017] [Indexed: 12/20/2022]
Abstract
The anthracycline-mediated cardiotoxicity is still not completely understood. To examine the impact of cholesterol metabolism and transport in this context, cholesterol and oxysterol levels as well as the expression of the cholesterol transporters ABCA1 and ABCG1 were analyzed in doxorubicin-treated HL-1 murine cardiomyocytes as well as in mouse model for acute doxorubicin-induced cardiotoxicity. Doxorubicin-treated HL-1 cells exhibited enhanced cholesterol (153±20% of control), oxysterol (24S-hydroxycholesterol: 206±29% of control) and cholesterol precursor levels (lathosterol: 122±12% of control; desmosterol: 188±10% of control) indicating enhanced cholesterol synthesis. Moreover, abca1 and abcg1 were upregulated on mRNA, protein and functional level caused by a doxorubicin-mediated activation of the nuclear receptor LXR. In addition, the oxysterols not only induced the abca1 and abcg1 in HL-1 cells but also enhanced the expression of endothelin-1 and transforming growth factor-β, which have already been identified as important factors in doxorubicin-induced cardiotoxicity. These in vitro findings were verified in a murine model for acute doxorubicin-induced cardiotoxicity, demonstrating elevated cardiac (2.1±0.2vs. 3.6±1.0ng/mg) and systemic cholesterol levels (105.0±8.4vs. 130.0±4.3mg/dl), respectively, as well as enhanced oxysterol levels such as cardiac 24S-hydroxycholesterol (2.1±0.2vs. 3.6±1.0ng/mg). In line with these findings cardiac mRNA expression of abca1 (303% of control) and abcg1 (161% of control) was induced. Taken together, our data demonstrate enhanced cholesterol and oxysterol levels by doxorubicin, resulting in a LXR-dependent upregulation of abca1 and abcg1. In this context, the cytotoxic effects of oxysterols and their impact on cardiac gene expression should be considered as an important factor in doxorubicin-induced cardiotoxicity.
Collapse
Affiliation(s)
- Judith V Monzel
- Dept. of Pharmacology at the Center of Drug Absorption and Transport (C_DAT), University Medicine, Greifswald, Germany
| | - Thomas Budde
- Dept. of Pharmacology at the Center of Drug Absorption and Transport (C_DAT), University Medicine, Greifswald, Germany
| | | | - Matthias Schwebe
- Dept. of Pharmacology at the Center of Drug Absorption and Transport (C_DAT), University Medicine, Greifswald, Germany
| | - Sandra Bien-Möller
- Dept. of Pharmacology at the Center of Drug Absorption and Transport (C_DAT), University Medicine, Greifswald, Germany
| | - Dieter Lütjohann
- Institute for Clinical Chemistry and Clinical Pharmacology, University of Bonn, Germany
| | - Heyo K Kroemer
- Dept. of Pharmacology at the Center of Drug Absorption and Transport (C_DAT), University Medicine, Greifswald, Germany
| | - Gabriele Jedlitschky
- Dept. of Pharmacology at the Center of Drug Absorption and Transport (C_DAT), University Medicine, Greifswald, Germany
| | - Markus Grube
- Dept. of Pharmacology at the Center of Drug Absorption and Transport (C_DAT), University Medicine, Greifswald, Germany.
| |
Collapse
|
42
|
Millar CL, Duclos Q, Blesso CN. Effects of Dietary Flavonoids on Reverse Cholesterol Transport, HDL Metabolism, and HDL Function. Adv Nutr 2017; 8:226-239. [PMID: 28298268 PMCID: PMC5347106 DOI: 10.3945/an.116.014050] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Strong experimental evidence confirms that HDL directly alleviates atherosclerosis. HDL particles display diverse atheroprotective functions in reverse cholesterol transport (RCT), antioxidant, anti-inflammatory, and antiapoptotic processes. In certain inflammatory disease states, however, HDL particles may become dysfunctional and proatherogenic. Flavonoids show the potential to improve HDL function through their well-documented effects on cellular antioxidant status and inflammation. The aim of this review is to summarize the basic science and clinical research examining the effects of dietary flavonoids on RCT and HDL function. Based on preclinical studies that used cell culture and rodent models, it appears that many flavonoids (e.g., anthocyanidins, flavonols, and flavone subclasses) influence RCT and HDL function beyond simple HDL cholesterol concentration by regulating cellular cholesterol efflux from macrophages and hepatic paraoxonase 1 expression and activity. In clinical studies, dietary anthocyanin intake is associated with beneficial changes in serum biomarkers related to HDL function in a variety of human populations (e.g., in those who are hyperlipidemic, hypertensive, or diabetic), including increased HDL cholesterol concentration, as well as HDL antioxidant and cholesterol efflux capacities. However, clinical research on HDL functionality is lacking for some flavonoid subclasses (e.g., flavanols, flavones, flavanones, and isoflavones). Although there has been a tremendous effort to develop HDL-targeted drug therapies, more research is warranted on how the intake of foods or specific nutrients affects HDL function.
Collapse
|
43
|
Li N, Wang X, Xu Y, Lin Y, Zhu N, Liu P, Lu D, Si S. Identification of a Novel Liver X Receptor Agonist that Regulates the Expression of Key Cholesterol Homeostasis Genes with Distinct Pharmacological Characteristics. Mol Pharmacol 2017; 91:264-276. [PMID: 28087808 DOI: 10.1124/mol.116.105213] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2016] [Accepted: 01/06/2017] [Indexed: 11/22/2022] Open
Abstract
Activation of liver X receptor (LXR) is associated with cholesterol metabolism and anti-inflammatory processes, which makes it beneficial to antiatherosclerosis therapy. Nevertheless, existing agonists that target LXR, for example TO901317, are related to unwanted side effects. In the present study, using a screening method we identified IMB-808, which displayed potent dual LXRα/β agonistic activity. In vitro, IMB-808 effectively increased the expressing quantity of genes related to reverse cholesterol transport process as well as those associated with cholesterol metabolism pathway in multiple cell lines. Additionally, IMB-808 remarkably promoted cholesterol efflux from RAW264.7 as well as THP-1 macrophage cells and reduced cellular lipid accumulation accordingly. Interestingly, compared with TO901317, IMB-808 almost did not increase the expressing quantity of genes related to lipogenesis in HepG2 cells, which indicated that IMB-808 could exhibit fewer internal lipogenic side effects with a characteristic of selective LXR agonist. Furthermore, in comparison with the full LXR agonist TO901317, IMB-808 recruits coregulators differently and possesses a distinct predictive binding pattern for the LXR ligand-binding domain. In summary, our study demonstrated that IMB-808 could act as an innovative partial LXR agonist that avoids common lipogenic side effects, providing insight for the design of novel LXR modulators. Our data indicate that this compound might be used as a promising therapeutic agent for the prospective treatment of atherosclerosis in the future.
Collapse
Affiliation(s)
- Ni Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences, Beijing, People's Republic of China (N.L., Y.L., D.L.); and Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China (N.L., X.W., Y.X., N.Z., P.L., S.S.)
| | - Xiao Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences, Beijing, People's Republic of China (N.L., Y.L., D.L.); and Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China (N.L., X.W., Y.X., N.Z., P.L., S.S.)
| | - Yanni Xu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences, Beijing, People's Republic of China (N.L., Y.L., D.L.); and Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China (N.L., X.W., Y.X., N.Z., P.L., S.S.)
| | - Yuan Lin
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences, Beijing, People's Republic of China (N.L., Y.L., D.L.); and Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China (N.L., X.W., Y.X., N.Z., P.L., S.S.)
| | - Ningyu Zhu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences, Beijing, People's Republic of China (N.L., Y.L., D.L.); and Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China (N.L., X.W., Y.X., N.Z., P.L., S.S.)
| | - Peng Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences, Beijing, People's Republic of China (N.L., Y.L., D.L.); and Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China (N.L., X.W., Y.X., N.Z., P.L., S.S.)
| | - Duo Lu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences, Beijing, People's Republic of China (N.L., Y.L., D.L.); and Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China (N.L., X.W., Y.X., N.Z., P.L., S.S.)
| | - Shuyi Si
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences, Beijing, People's Republic of China (N.L., Y.L., D.L.); and Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China (N.L., X.W., Y.X., N.Z., P.L., S.S.)
| |
Collapse
|
44
|
Hu X, Fu Y, Lu X, Zhang Z, Zhang W, Cao Y, Zhang N. Protective Effects of Platycodin D on Lipopolysaccharide-Induced Acute Lung Injury by Activating LXRα-ABCA1 Signaling Pathway. Front Immunol 2017; 7:644. [PMID: 28096801 PMCID: PMC5206804 DOI: 10.3389/fimmu.2016.00644] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 12/13/2016] [Indexed: 12/29/2022] Open
Abstract
The purpose of this study was to investigate the protective effects of platycodin D (PLD) on lipopolysaccharide (LPS)-induced acute lung injury (ALI) and clarify the possible mechanism. An LPS-induced ALI model was used to confirm the anti-inflammatory activity of PLD in vivo. The A549 lung epithelial cells were used to investigate the molecular mechanism and targets of PLD in vitro. In vivo, the results showed that PLD significantly attenuated lung histopathologic changes, myeloperoxidase activity, and pro-inflammatory cytokines levels, including TNF-α, IL-1β, and IL-6. In vitro, PLD inhibited LPS-induced IL-6 and IL-8 production in LPS-stimulated A549 lung epithelial cells. Western blot analysis showed that PLD suppressed LPS-induced NF-κB and IRF3 activation. Moreover, PLD did not act though affecting the expression of TLR4. We also showed that PLD disrupted the formation of lipid rafts by depleting cholesterol and prevented LPS-induced TLR4 trafficking to lipid rafts, thereby blocking LPS-induced inflammatory response. Finally, PLD activated LXRα–ABCA1-dependent cholesterol efflux. Knockdown of LXRα abrogated the anti-inflammatory effects of PLD. The anti-inflammatory effects of PLD was associated with upregulation of the LXRα–ABCA1 pathway, which resulted in disrupting lipid rafts by depleting cholesterol and reducing translocation of TLR4 to lipid rafts.
Collapse
Affiliation(s)
- Xiaoyu Hu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University , Changchun , China
| | - Yunhe Fu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University , Changchun , China
| | - Xiaojie Lu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University , Changchun , China
| | - Zecai Zhang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University , Changchun , China
| | - Wenlong Zhang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University , Changchun , China
| | - Yongguo Cao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University , Changchun , China
| | - Naisheng Zhang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University , Changchun , China
| |
Collapse
|
45
|
Li N, Wang X, Liu P, Lu D, Jiang W, Xu Y, Si S. E17110 promotes reverse cholesterol transport with liver X receptor β agonist activity in vitro. Acta Pharm Sin B 2016; 6:198-204. [PMID: 27175330 PMCID: PMC4856951 DOI: 10.1016/j.apsb.2016.03.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 03/04/2016] [Accepted: 03/17/2016] [Indexed: 12/11/2022] Open
Abstract
Liver X receptor (LXR) plays an important role in reverse cholesterol transport (RCT), and activation of LXR could reduce atherosclerosis. In the present study we used a cell-based screening method to identify new potential LXRβ agonists. A novel benzofuran-2-carboxylate derivative was identified with LXRβ agonist activity: E17110 showed a significant activation effect on LXRβ with an EC50 value of 0.72 μmol/L. E17110 also increased the expression of ATP-binding cassette transporter A1 (ABCA1) and G1 (ABCG1) in RAW264.7 macrophages. Moreover, E17110 significantly reduced cellular lipid accumulation and promoted cholesterol efflux in RAW264.7 macrophages. Interestingly, we found that the key amino acids in the LXRβ ligand-binding domain had distinct interactions with E17110 as compared to TO901317. These results suggest that E17110 was identified as a novel compound with LXRβ agonist activity in vitro via screening, and could be developed as a potential anti-atherosclerotic lead compound.
Collapse
Key Words
- ABCA1
- ABCA1, ATP-binding cassette transporter A1
- ABCG1
- ABCG1, ATP-binding cassette transporter G1
- ApoA-I, apolipoprotein A-I
- Atherosclerosis
- Cholesterol efflux
- GAPDH, glyceraldehyde-phosphate dehydrogenase
- HDL, high-density lipoprotein
- LBD, ligand-binding domain
- LXR, liver X receptor
- LXRE, LXR response element
- LXRβ
- NR, nuclear receptor
- RCT, reverse cholesterol transport
- RXR, retinoid X receptor
- Reverse cholesterol transport
- ox-LDL, oxidized low-density lipoprotein
Collapse
Affiliation(s)
- Ni Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences, Beijing 100050, China
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Xiao Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Peng Liu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Duo Lu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences, Beijing 100050, China
| | - Wei Jiang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yanni Xu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Corresponding authors. Fax: +86 10 63180604.
| | - Shuyi Si
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Corresponding authors. Fax: +86 10 63180604.
| |
Collapse
|
46
|
Yongsakulchai P, Settasatian C, Settasatian N, Komanasin N, Kukongwiriyapan U, Cote ML, Intharapetch P, Senthong V. Association of combined genetic variations in PPARγ , PGC-1α , and LXRα with coronary artery disease and severity in Thai population. Atherosclerosis 2016; 248:140-8. [DOI: 10.1016/j.atherosclerosis.2016.03.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Revised: 02/10/2016] [Accepted: 03/03/2016] [Indexed: 12/11/2022]
|
47
|
Ledda A, González M, Gulfo J, Díaz Ludovico I, Ramella N, Toledo J, Garda H, Grasa M, Esteve M. Decreased OxLDL uptake and cholesterol efflux in THP1 cells elicited by cortisol and by cortisone through 11β-hydroxysteroid dehydrogenase type 1. Atherosclerosis 2016; 250:84-94. [PMID: 27187933 DOI: 10.1016/j.atherosclerosis.2016.04.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 04/04/2016] [Accepted: 04/24/2016] [Indexed: 10/21/2022]
Abstract
BACKGROUND AND AIMS Data about glucocorticoids role in the development of atherosclerosis are controversial showing different effects in human than in experimental animal models. Atherosclerosis is the result of a chronic inflammatory response to an injured endothelium where an uncontrolled uptake of OxLDL by macrophages triggers the development of foam cells, the main component of fatty streaks in atherosclerotic plaque. There are few data about the direct effect of glucocorticoids in macrophages of atherosclerotic plaque. The aim of the study was to elucidate the role of glucocorticoids in the development of foam cells in atherosclerosis initiation. METHODS For this purpose we used THP1 cells differentiated to macrophages with phorbol esters and incubated with OxLDL alone or with cortisol or cortisone. THP1 cells were also incubated with cortisone plus an inhibitor of 11β-hydroxysteroid dehydrogenase 1 (11βHSD1) activity to determine the role of this enzyme on glucocorticoid action in this process. RESULTS Ours results showed that cortisol and cortisone decreased significantly the inflammation promoted by OxLDL, and also diminished the expression of genes involved in influx and efflux of cholesterol resulting in a reduced lipid accumulation. Likewise cortisol and cortisone decreased 11βHSD1 expression in THP1 cells. The presence of the inhibitor of 11βHSD1 abolished all the effects elicited by cortisone. CONCLUSION Our results indicate a direct effect of glucocorticoids on macrophages braking atherosclerosis initiation, reducing pro-inflammatory markers and OxLDL uptake and cholesterol re-esterification, but also inhibiting cholesterol output. These effects appear to be mediated, at least in part, by 11βHSD1 activity.
Collapse
Affiliation(s)
- Angelo Ledda
- Department of Nutrition and Food Sciences, Faculty of Biology, University of Barcelona, Barcelona, Spain; CIBER Obesity and Nutrition, Institute of Health Carlos III, Madrid, Spain
| | - Marina González
- INIBIOLP-CONICET, Facultad Cs. Médicas, Universidad Nacional de La Plata, La Plata, Argentina
| | - José Gulfo
- Department of Nutrition and Food Sciences, Faculty of Biology, University of Barcelona, Barcelona, Spain; CIBER Obesity and Nutrition, Institute of Health Carlos III, Madrid, Spain
| | - Ivo Díaz Ludovico
- INIBIOLP-CONICET, Facultad Cs. Médicas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Nahuel Ramella
- INIBIOLP-CONICET, Facultad Cs. Médicas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Juan Toledo
- INIBIOLP-CONICET, Facultad Cs. Médicas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Horacio Garda
- INIBIOLP-CONICET, Facultad Cs. Médicas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Mar Grasa
- Department of Nutrition and Food Sciences, Faculty of Biology, University of Barcelona, Barcelona, Spain; CIBER Obesity and Nutrition, Institute of Health Carlos III, Madrid, Spain
| | - Montserrat Esteve
- Department of Nutrition and Food Sciences, Faculty of Biology, University of Barcelona, Barcelona, Spain; CIBER Obesity and Nutrition, Institute of Health Carlos III, Madrid, Spain.
| |
Collapse
|
48
|
Zhang Y, Si Y, Zhai L, Guo S, Zhao J, Sang H, Pang X, Zhang X, Chen A, Qin S. Celastrus Orbiculatus Thunb. Reduces Lipid Accumulation by Promoting Reverse Cholesterol Transport in Hyperlipidemic Mice. Lipids 2016; 51:677-92. [DOI: 10.1007/s11745-016-4145-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 03/15/2016] [Indexed: 02/03/2023]
|
49
|
Baldán Á, de Aguiar Vallim TQ. miRNAs and High-Density Lipoprotein metabolism. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:2053-2061. [PMID: 26869447 DOI: 10.1016/j.bbalip.2016.01.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 01/28/2016] [Accepted: 01/29/2016] [Indexed: 12/16/2022]
Abstract
Altered lipoprotein metabolism plays a key role during atherogenesis. For over 50years, epidemiological data have fueled the proposal that HDL-cholesterol (HDL-c) in circulation is inversely correlated to cardiovascular risk. However, the atheroprotective role of HDL is currently the focus of much debate and remains an active field of research. The emerging picture from research in the past decade suggests that HDL function, rather than HDL-c content, is important in disease. Recent developments demonstrate that miRNAs play an important role in fine-tuning the expression of key genes involved in HDL biogenesis, lipidation, and clearance, as well as in determining the amounts of HDL-c in circulation. Thus, it has been proposed that miRNAs that affect HDL metabolism might be exploited therapeutically in patients. Whether HDL-based therapies, alone or in combination with LDL-based treatments (e.g. statins), provide superior outcomes in patients has been recently questioned by human genetics studies and clinical trials. The switch in focus from "HDL-cholesterol" to "HDL function" opens a new paradigm to understand the physiology and therapeutic potential of HDL, and to find novel modulators of cardiovascular risk. In this review we summarize the current knowledge on the regulation of HDL metabolism and function by miRNAs. This article is part of a Special Issue entitled: MicroRNAs and lipid/energy metabolism and related diseases edited by Carlos Fernández-Hernando and Yajaira Suárez.
Collapse
Affiliation(s)
- Ángel Baldán
- Edward A. Doisy Department of Biochemistry & Molecular Biology, Center for Cardiovascular Research, and Liver Center, 1100 S. Grand Blvd., Saint Louis University, Saint Louis, MO 63104, United States.
| | - Thomas Q de Aguiar Vallim
- Department of Medicine, Division of Cardiology, 650 Charles E. Young Drive S, A2-237 CHS, UCLA Los Angeles, Los Angeles, CA 90095, United States.
| |
Collapse
|
50
|
Pleiotropic effects of antitumour alkylphospholipids on cholesterol transport and metabolism. Exp Cell Res 2015; 340:81-90. [PMID: 26712518 DOI: 10.1016/j.yexcr.2015.12.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 12/15/2015] [Accepted: 12/18/2015] [Indexed: 11/20/2022]
Abstract
BACKGROUND Alkylphospholipid (APL) analogs are a new class of membrane-directed synthetic compounds with a variety of biological actions and clinical applications. In particular, these agents are promising candidates in cancer treatment. We have demonstrated that after prolonged treatment APLs alter intracellular cholesterol traffic and metabolism in human tumor-cell lines, leading to an accumulation of cholesterol inside the cell. After further investigation concerning the mode of action of APLs, we have explored the influence of several APLs on novel aspects of cholesterol and lipoprotein homeostasis using hepatoma HepG2 cells and THP1-derived macrophages. METHODS Quantitative real-time PCR analysis with a pathway-focused PCR array system was performed to measure relative changes in the mRNA expression of a number of genes related to cholesterol transport and metabolism. We compared the gene-expression profiles of HepG2 cells treated with miltefosine, edelfosine or perifosine for 6h and 24h with the profile of control cells. We also analysed particular genes of interest in both HepG2 and macrophage-like THP1 cells using specific PCR assays. Immunoblots were used to confirm protein-expression changes. Measurement of ABCA1-mediated cholesterol efflux was determined using apoA1 as cholesterol acceptor. RESULTS We found global changes in gene-expression patterns to maintain cholesterol homeostasis after exposure of cells to APLs. The pathways for cholesterol biosynthesis and LDL-cholesterol uptake were both transcriptionally upregulated by the three APLs assayed. Conversely, major pathways involved in the catabolism of cholesterol to bile acids and lipoprotein-associated cholesterol export were impaired after APL incubation, which may well contribute to the higher cell-cholesterol levels induced by these compounds. CONCLUSION Incubation of cells with different APLs stimulated cholesterol biosynthesis and uptake at the same time as it depressed common pathways for excess cholesterol removal in tumor cells, ultimately leading to altered cholesterol homeostasis.
Collapse
|