1
|
Willemsen S, Yengej FAY, Puschhof J, Rookmaaker MB, Verhaar MC, van Es J, Beumer J, Clevers H. A comprehensive transcriptome characterization of individual nuclear receptor pathways in the human small intestine. Proc Natl Acad Sci U S A 2024; 121:e2411189121. [PMID: 39475639 PMCID: PMC11551338 DOI: 10.1073/pnas.2411189121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 09/24/2024] [Indexed: 11/13/2024] Open
Abstract
Nuclear receptors (NRs) are widely expressed transcription factors that bind small, lipophilic compounds and regulate diverse biological processes. In the small intestine, NRs are known to act as sensors that control transcriptional responses to endogenous and exogenous signals, yet their downstream effects have not been characterized extensively. Here, we investigate the activation of six different NRs individually in human intestinal organoids using small molecules agonists. We observe changes in key enterocyte functions such as lipid, glucose, and amino acid absorption, the regulation of electrolyte balance, and drug metabolism. Our findings reinforce PXR, LXR, FXR, and PPARα as regulators of lipid absorption. Furthermore, known hepatic effects of AHR and VDR activation were recapitulated in the human small intestine. Finally, we identify unique target genes for intestinal PXR activation (ERG28, TMEM97, and TM7SF2), LXR activation (RAB6B), and VDR activation (CA12). This study provides an unbiased and comprehensive transcriptomic description of individual NR pathways in the human small intestine. By gaining a deeper understanding of the effects of individual NRs, we might better harness their pharmacological and therapeutic potential.
Collapse
Affiliation(s)
- Sam Willemsen
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences, Utrecht3584 CT, The Netherlands
- University Medical Centre Utrecht, Utrecht3584 CX, The Netherlands
- Oncode Institute, Utrecht3584 CT, The Netherlands
| | - Fjodor A. Yousef Yengej
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences, Utrecht3584 CT, The Netherlands
- University Medical Centre Utrecht, Utrecht3584 CX, The Netherlands
| | - Jens Puschhof
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences, Utrecht3584 CT, The Netherlands
- University Medical Centre Utrecht, Utrecht3584 CX, The Netherlands
- Oncode Institute, Utrecht3584 CT, The Netherlands
- Junior Research Group Epithelium Microbiome Interactions, German Cancer Research Center, Heidelberg69120, Germany
| | | | | | - Johan van Es
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences, Utrecht3584 CT, The Netherlands
- University Medical Centre Utrecht, Utrecht3584 CX, The Netherlands
- Oncode Institute, Utrecht3584 CT, The Netherlands
| | - Joep Beumer
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences, Utrecht3584 CT, The Netherlands
- University Medical Centre Utrecht, Utrecht3584 CX, The Netherlands
- Oncode Institute, Utrecht3584 CT, The Netherlands
- Institute of Human Biology, Roche Innovation Center Basel, Basel4058, Switzerland
| | - Hans Clevers
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences, Utrecht3584 CT, The Netherlands
- University Medical Centre Utrecht, Utrecht3584 CX, The Netherlands
- Oncode Institute, Utrecht3584 CT, The Netherlands
- The Princess Máxima Center for Pediatric Oncology, Utrecht3584 CS, The Netherlands
- Pharma, Research and Early Development of F. Hoffmann-La Roche Ltd, BaselCH-4070, Switzerland
| |
Collapse
|
2
|
Ding J, Nguyen AT, Lohman K, Hensley MT, Parker D, Hou L, Taylor J, Voora D, Sawyer JK, Boudyguina E, Bancks MP, Bertoni A, Pankow JS, Rotter JI, Goodarzi MO, Tracy RP, Murdoch DM, Duprez D, Rich SS, Psaty BM, Siscovick D, Newgard CB, Herrington D, Hoeschele I, Shea S, Stein JH, Patel M, Post W, Jacobs D, Parks JS, Liu Y. LXR signaling pathways link cholesterol metabolism with risk for prediabetes and diabetes. J Clin Invest 2024; 134:e173278. [PMID: 38747290 PMCID: PMC11093600 DOI: 10.1172/jci173278] [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: 06/21/2023] [Accepted: 03/20/2024] [Indexed: 05/19/2024] Open
Abstract
BACKGROUNDPreclinical studies suggest that cholesterol accumulation leads to insulin resistance. We previously reported that alterations in a monocyte cholesterol metabolism transcriptional network (CMTN) - suggestive of cellular cholesterol accumulation - were cross-sectionally associated with obesity and type 2 diabetes (T2D). Here, we sought to determine whether the CMTN alterations independently predict incident prediabetes/T2D risk, and correlate with cellular cholesterol accumulation.METHODSMonocyte mRNA expression of 11 CMTN genes was quantified among 934 Multi-Ethnic Study of Atherosclerosis (MESA) participants free of prediabetes/T2D; cellular cholesterol was measured in a subset of 24 monocyte samples.RESULTSDuring a median 6-year follow-up, lower expression of 3 highly correlated LXR target genes - ABCG1 and ABCA1 (cholesterol efflux) and MYLIP (cholesterol uptake suppression) - and not other CMTN genes, was significantly associated with higher risk of incident prediabetes/T2D. Lower expression of the LXR target genes correlated with higher cellular cholesterol levels (e.g., 47% of variance in cellular total cholesterol explained by ABCG1 expression). Further, adding the LXR target genes to overweight/obesity and other known predictors significantly improved prediction of incident prediabetes/T2D.CONCLUSIONThese data suggest that the aberrant LXR/ABCG1-ABCA1-MYLIP pathway (LAAMP) is a major T2D risk factor and support a potential role for aberrant LAAMP and cellular cholesterol accumulation in diabetogenesis.FUNDINGThe MESA Epigenomics and Transcriptomics Studies were funded by NIH grants 1R01HL101250, 1RF1AG054474, R01HL126477, R01DK101921, and R01HL135009. This work was supported by funding from NIDDK R01DK103531 and NHLBI R01HL119962.
Collapse
Affiliation(s)
- Jingzhong Ding
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | | | - Kurt Lohman
- Department of Medicine, Division of Cardiology, and
| | | | - Daniel Parker
- Department of Medicine, Division of Geriatrics, Duke University, Durham, North Carolina, USA
| | - Li Hou
- Department of Medicine, Division of Cardiology, and
| | - Jackson Taylor
- Department of Biological, Geological, and Environmental Sciences, Cleveland State University, Cleveland, Ohio, USA
| | - Deepak Voora
- Department of Medicine, Division of Cardiology, and
| | - Janet K. Sawyer
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Elena Boudyguina
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Michael P. Bancks
- Department of Epidemiology and Prevention, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Alain Bertoni
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - James S. Pankow
- Division of Epidemiology and Community Health, University of Minnesota, Minneapolis, Minnesota, USA
| | - Jerome I. Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, USA
| | - Mark O. Goodarzi
- Division of Endocrinology, Diabetes and Metabolism, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Russell P. Tracy
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, Vermont, USA
| | - David M. Murdoch
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Duke University, Durham, North Carolina, USA
| | - Daniel Duprez
- Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Stephen S. Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia, USA
| | - Bruce M. Psaty
- Cardiovascular Health Research Unit, Departments of Medicine, Epidemiology, and Health Systems and Population Health, University of Washington, Seattle, Washington, USA
| | | | - Christopher B. Newgard
- Department of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina, USA
| | - David Herrington
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Ina Hoeschele
- Fralin Life Sciences Institute, Virginia Tech, Blacksburg, Virginia, USA
| | - Steven Shea
- Department of Medicine, Columbia University, New York, New York, USA
| | - James H. Stein
- School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, USA
| | - Manesh Patel
- Department of Medicine, Division of Cardiology, and
| | - Wendy Post
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - David Jacobs
- Division of Epidemiology and Community Health, University of Minnesota, Minneapolis, Minnesota, USA
| | - John S. Parks
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Yongmei Liu
- Department of Medicine, Division of Cardiology, and
| |
Collapse
|
3
|
Greco S, Campigotto M, D’Amuri A, Fabbri N, Passaro A. Dyslipidemia, Cholangitis and Fatty Liver Disease: The Close Underexplored Relationship: A Narrative Review. J Clin Med 2024; 13:2714. [PMID: 38731243 PMCID: PMC11084647 DOI: 10.3390/jcm13092714] [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: 04/10/2024] [Revised: 04/25/2024] [Accepted: 04/30/2024] [Indexed: 05/13/2024] Open
Abstract
In assessing individual cardiovascular risk, dyslipidemia is known for emerging as a pivotal factor significantly contributing to major cardiovascular events. However, dyslipidemic patients frequently present with concurrent medical conditions, each with varying frequencies of occurrence; cholangitis, whether acute or chronic, and hepatic steatosis, along with associated conditions, are strongly associated with specific forms of dyslipidemia, and these associations are reasonably well elucidated. Conversely, evidence linking biliary disease to hepatic steatosis is comparatively scant. This narrative review aims to bridge this gap in knowledge concerning the interplay between dyslipidemia, cholangitis, and hepatic steatosis. By addressing this gap, clinicians can better identify patients at heightened risk of future major cardiovascular events, facilitating more targeted interventions and management strategies. The review delves into the intricate relationships between dyslipidemia and these hepatic and biliary clinical conditions, shedding light on potential mechanisms underlying their associations. Understanding these complex interactions is crucial for optimizing cardiovascular risk assessment as well and devising tailored treatment approaches for patients with dyslipidemia and associated hepatic disorders. Moreover, elucidating these connections empowers clinicians with the knowledge needed to navigate the multifaceted landscape of cardiovascular risk assessment and management effectively. By exploring the intricate relationships between dyslipidemia, cholangitis, and hepatic steatosis (without forgetting the possible clinical consequences of hepatic steatosis itself), this review not only contributes to the existing body of knowledge but also offers insights into potential avenues for further research and clinical practice. Thus, it serves as a valuable resource for healthcare professionals striving to enhance patient care and outcomes in the context of cardiovascular disease and associated hepatic conditions.
Collapse
Affiliation(s)
- Salvatore Greco
- Department of Translational Medicine, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, FE, Italy;
- Department of Internal Medicine, Ospedale del Delta, Via Valle Oppio 2, 44023 Lagosanto, FE, Italy
| | - Michele Campigotto
- Gastroenterology and Digestive Endoscopy Unit, ASUGI, Cattinara University Hospital, Strada di Fiume 447, 34149 Trieste, TS, Italy;
| | - Andrea D’Amuri
- General Medicine Unit, Medical Department, ASST Mantova, Ospedale Carlo Poma, Strada Lago Paiolo 10, 46100 Mantova, MN, Italy;
| | - Nicolò Fabbri
- Department of General Surgery, Ospedale del Delta, Via Valle Oppio 2, 44023 Lagosanto, FE, Italy;
| | - Angelina Passaro
- Department of Translational Medicine, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, FE, Italy;
| |
Collapse
|
4
|
Wu D, Khan FA, Zhang K, Pandupuspitasari NS, Negara W, Guan K, Sun F, Huang C. Retinoic acid signaling in development and differentiation commitment and its regulatory topology. Chem Biol Interact 2024; 387:110773. [PMID: 37977248 DOI: 10.1016/j.cbi.2023.110773] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 10/11/2023] [Accepted: 10/20/2023] [Indexed: 11/19/2023]
Abstract
Retinoic acid (RA), the derivative of vitamin A/retinol, is a signaling molecule with important implications in health and disease. It is a well-known developmental morphogen that functions mainly through the transcriptional activity of nuclear RA receptors (RARs) and, uncommonly, through other nuclear receptors, including peroxisome proliferator-activated receptors. Intracellular RA is under spatiotemporally fine-tuned regulation by synthesis and degradation processes catalyzed by retinaldehyde dehydrogenases and P450 family enzymes, respectively. In addition to dictating the transcription architecture, RA also impinges on cell functioning through non-genomic mechanisms independent of RAR transcriptional activity. Although RA-based differentiation therapy has achieved impressive success in the treatment of hematologic malignancies, RA also has pro-tumor activity. Here, we highlight the relevance of RA signaling in cell-fate determination, neurogenesis, visual function, inflammatory responses and gametogenesis commitment. Genetic and post-translational modifications of RAR are also discussed. A better understanding of RA signaling will foster the development of precision medicine to improve the defects caused by deregulated RA signaling.
Collapse
Affiliation(s)
- Di Wu
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong, 226001, China
| | - Faheem Ahmed Khan
- Research Center for Animal Husbandry, National Research and Innovation Agency, Jakarta Pusat, 10340, Indonesia
| | - Kejia Zhang
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong, 226001, China
| | | | - Windu Negara
- Research Center for Animal Husbandry, National Research and Innovation Agency, Jakarta Pusat, 10340, Indonesia
| | - Kaifeng Guan
- School of Advanced Agricultural Sciences, Peking University, Beijing, 100871, China.
| | - Fei Sun
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong, 226001, China.
| | - Chunjie Huang
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong, 226001, China.
| |
Collapse
|
5
|
Li Z, Zheng D, Zhang T, Ruan S, Li N, Yu Y, Peng Y, Wang D. The roles of nuclear receptors in cholesterol metabolism and reverse cholesterol transport in nonalcoholic fatty liver disease. Hepatol Commun 2024; 8:e0343. [PMID: 38099854 PMCID: PMC10727660 DOI: 10.1097/hc9.0000000000000343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Accepted: 10/28/2023] [Indexed: 12/18/2023] Open
Abstract
As the most prevalent chronic liver disease globally, NAFLD encompasses a pathological process that ranges from simple steatosis to NASH, fibrosis, cirrhosis, and HCC, closely associated with numerous extrahepatic diseases. While the initial etiology was believed to be hepatocyte injury caused by lipid toxicity from accumulated triglycerides, recent studies suggest that an imbalance of cholesterol homeostasis is of greater significance. The role of nuclear receptors in regulating liver cholesterol homeostasis has been demonstrated to be crucial. This review summarizes the roles and regulatory mechanisms of nuclear receptors in the 3 main aspects of cholesterol production, excretion, and storage in the liver, as well as their cross talk in reverse cholesterol transport. It is hoped that this review will offer new insights and theoretical foundations for the study of the pathogenesis and progression of NAFLD and provide new research directions for extrahepatic diseases associated with NAFLD.
Collapse
|
6
|
Zhang Z, Zhang B, Jiang X, Yu Y, Cui Y, Luo H, Wang B. Hyocholic acid retards renal fibrosis by regulating lipid metabolism and inflammatory response in a sheep model. Int Immunopharmacol 2023; 122:110670. [PMID: 37481851 DOI: 10.1016/j.intimp.2023.110670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 07/15/2023] [Accepted: 07/16/2023] [Indexed: 07/25/2023]
Abstract
The kidneys are vital organs that regulate metabolic homeostasis in the body, filter waste products from the blood, and remove extrahepatic bile acids. We previously found that the dietary supplementation of hyocholic acid alleviated the sheep body lipid deposition and decreased kidney weight. This study evaluated hyocholic acid's (HCA) roles and mechanisms on lipid metabolism and anti-inflammatory function in the kidney under a high-energy diet. Histomicrograph showing the apparent improvement by HCA by attenuating structural damage. The HCA treatment reduced the renal accumulation of cholesterol. Bile acid receptors such as LXR and FXR were activated at the protein level. HCA significantly altered several genes related to immune response (NF-κB, IL-6, and MCP1) and fibrosis (TGF-β, Col1α1, and α-SMA). These significant changes correlated with renal lipid accumulation. The KEGG pathways including non-alcoholic fatty liver disease, insulin resistance, TNF signaling pathway, and Th17 cell differentiation were enriched and NF-κB, IL-6, and TGF-β were identified as the core interconnected genes. This study revealed that HCA plays an efficient role in alleviating kidney lipids accumulation and inflammatory response through crucial genes such as FXR, LXR, HMGCR, NF-κB, IL-6, MCP1, and TGF-β, and expand our understanding of HCA's role in kidney function. In conclusion, HCA mitigated kidney fibrosis, lipid metabolism disorders and immune responses induced by a high-energy diet by regulating a potential LXR/SREBP2/TGF-β-NF-κB signaling pathway.
Collapse
Affiliation(s)
- Zeping Zhang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, PR China
| | - Boyan Zhang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, PR China
| | - Xianzhe Jiang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, PR China
| | - Yue Yu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, PR China
| | - Yimeng Cui
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, PR China
| | - Hailing Luo
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, PR China
| | - Bing Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, PR China.
| |
Collapse
|
7
|
Kim H, Park C, Kim TH. Targeting Liver X Receptors for the Treatment of Non-Alcoholic Fatty Liver Disease. Cells 2023; 12:cells12091292. [PMID: 37174692 PMCID: PMC10177243 DOI: 10.3390/cells12091292] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 04/29/2023] [Accepted: 04/29/2023] [Indexed: 05/15/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) refers to a range of conditions in which excess lipids accumulate in the liver, possibly leading to serious hepatic manifestations such as steatohepatitis, fibrosis/cirrhosis and cancer. Despite its increasing prevalence and significant impact on liver disease-associated mortality worldwide, no medication has been approved for the treatment of NAFLD yet. Liver X receptors α/β (LXRα and LXRβ) are lipid-activated nuclear receptors that serve as master regulators of lipid homeostasis and play pivotal roles in controlling various metabolic processes, including lipid metabolism, inflammation and immune response. Of note, NAFLD progression is characterized by increased accumulation of triglycerides and cholesterol, hepatic de novo lipogenesis, mitochondrial dysfunction and augmented inflammation, all of which are highly attributed to dysregulated LXR signaling. Thus, targeting LXRs may provide promising strategies for the treatment of NAFLD. However, emerging evidence has revealed that modulating the activity of LXRs has various metabolic consequences, as the main functions of LXRs can distinctively vary in a cell type-dependent manner. Therefore, understanding how LXRs in the liver integrate various signaling pathways and regulate metabolic homeostasis from a cellular perspective using recent advances in research may provide new insights into therapeutic strategies for NAFLD and associated metabolic diseases.
Collapse
Affiliation(s)
- Hyejin Kim
- College of Pharmacy, Sookmyung Women's University, Seoul 04310, Republic of Korea
| | - Chaewon Park
- College of Pharmacy, Sookmyung Women's University, Seoul 04310, Republic of Korea
| | - Tae Hyun Kim
- College of Pharmacy, Sookmyung Women's University, Seoul 04310, Republic of Korea
- Drug Information Research Institute, Sookmyung Women's University, Seoul 04310, Republic of Korea
- Muscle Physiome Research Center, Sookmyung Women's University, Seoul 04310, Republic of Korea
| |
Collapse
|
8
|
Cassim Bawa FN, Gopoju R, Xu Y, Hu S, Zhu Y, Chen S, Jadhav K, Zhang Y. Retinoic Acid Receptor Alpha (RARα) in Macrophages Protects from Diet-Induced Atherosclerosis in Mice. Cells 2022; 11:3186. [PMID: 36291054 PMCID: PMC9600071 DOI: 10.3390/cells11203186] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/07/2022] [Accepted: 10/09/2022] [Indexed: 08/13/2023] Open
Abstract
Retinoic acid signaling plays an important role in regulating lipid metabolism and inflammation. However, the role of retinoic acid receptor alpha (RARα) in atherosclerosis remains to be determined. In the current study, we investigated the role of macrophage RARα in the development of atherosclerosis. Macrophages isolated from myeloid-specific Rarα-/- (RarαMac-/-) mice showed increased lipid accumulation and inflammation and reduced cholesterol efflux compared to Rarαfl/fl (control) mice. All-trans retinoic acid (AtRA) induced ATP-binding cassette subfamily A member 1 (Abca1) and Abcg1 expression and cholesterol efflux in both RarαMac-/- mice and Rarαfl/fl mice. In Ldlr-/- mice, myeloid ablation of RARα significantly reduced macrophage Abca1 and Abcg1 expression and cholesterol efflux, induced inflammatory genes, and aggravated Western diet-induced atherosclerosis. Our data demonstrate that macrophage RARα protects against atherosclerosis, likely via inducing cholesterol efflux and inhibiting inflammation.
Collapse
Affiliation(s)
- Fathima N. Cassim Bawa
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272, USA
- School of Biomedical Sciences, Kent State University, Kent, OH 44240, USA
| | - Raja Gopoju
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272, USA
| | - Yanyong Xu
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272, USA
| | - Shuwei Hu
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272, USA
| | - Yingdong Zhu
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272, USA
- School of Biomedical Sciences, Kent State University, Kent, OH 44240, USA
| | - Shaoru Chen
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272, USA
| | - Kavita Jadhav
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272, USA
| | - Yanqiao Zhang
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272, USA
| |
Collapse
|
9
|
Nuclear Receptors in Pregnancy and Outcomes: Clinical Perspective. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1390:3-19. [DOI: 10.1007/978-3-031-11836-4_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
10
|
Rozhkova AV, Dmitrieva VG, Nosova EV, Dergunov AD, Limborska SA, Dergunova LV. Genomic Variants and Multilevel Regulation of ABCA1, ABCG1, and SCARB1 Expression in Atherogenesis. J Cardiovasc Dev Dis 2021; 8:jcdd8120170. [PMID: 34940525 PMCID: PMC8707585 DOI: 10.3390/jcdd8120170] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 11/28/2021] [Accepted: 11/29/2021] [Indexed: 12/12/2022] Open
Abstract
Atheroprotective properties of human plasma high-density lipoproteins (HDLs) are determined by their involvement in reverse cholesterol transport (RCT) from the macrophage to the liver. ABCA1, ABCG1, and SR-BI cholesterol transporters are involved in cholesterol efflux from macrophages to lipid-free ApoA-I and HDL as a first RCT step. Molecular determinants of RCT efficiency that may possess diagnostic and therapeutic meaning remain largely unknown. This review summarizes the progress in studying the genomic variants of ABCA1, ABCG1, and SCARB1, and the regulation of their function at transcriptional and post-transcriptional levels in atherosclerosis. Defects in the structure and function of ABCA1, ABCG1, and SR-BI are caused by changes in the gene sequence, such as single nucleotide polymorphism or various mutations. In the transcription initiation of transporter genes, in addition to transcription factors, long noncoding RNA (lncRNA), transcription activators, and repressors are also involved. Furthermore, transcription is substantially influenced by the methylation of gene promoter regions. Post-transcriptional regulation involves microRNAs and lncRNAs, including circular RNAs. The potential biomarkers and targets for atheroprotection, based on molecular mechanisms of expression regulation for three transporter genes, are also discussed in this review.
Collapse
Affiliation(s)
- Alexandra V. Rozhkova
- Department of Molecular Bases of Human Genetics, Institute of Molecular Genetics of National Research Center “Kurchatov Institute”, 123182 Moscow, Russia; (A.V.R.); (V.G.D.); (E.V.N.); (S.A.L.); (L.V.D.)
| | - Veronika G. Dmitrieva
- Department of Molecular Bases of Human Genetics, Institute of Molecular Genetics of National Research Center “Kurchatov Institute”, 123182 Moscow, Russia; (A.V.R.); (V.G.D.); (E.V.N.); (S.A.L.); (L.V.D.)
| | - Elena V. Nosova
- Department of Molecular Bases of Human Genetics, Institute of Molecular Genetics of National Research Center “Kurchatov Institute”, 123182 Moscow, Russia; (A.V.R.); (V.G.D.); (E.V.N.); (S.A.L.); (L.V.D.)
| | - Alexander D. Dergunov
- Laboratory of Structural Fundamentals of Lipoprotein Metabolism, National Medical Research Center for Therapy and Preventive Medicine, 101990 Moscow, Russia
- Correspondence:
| | - Svetlana A. Limborska
- Department of Molecular Bases of Human Genetics, Institute of Molecular Genetics of National Research Center “Kurchatov Institute”, 123182 Moscow, Russia; (A.V.R.); (V.G.D.); (E.V.N.); (S.A.L.); (L.V.D.)
| | - Liudmila V. Dergunova
- Department of Molecular Bases of Human Genetics, Institute of Molecular Genetics of National Research Center “Kurchatov Institute”, 123182 Moscow, Russia; (A.V.R.); (V.G.D.); (E.V.N.); (S.A.L.); (L.V.D.)
| |
Collapse
|
11
|
Desmosterol suppresses macrophage inflammasome activation and protects against vascular inflammation and atherosclerosis. Proc Natl Acad Sci U S A 2021; 118:2107682118. [PMID: 34782454 DOI: 10.1073/pnas.2107682118] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/11/2021] [Indexed: 12/14/2022] Open
Abstract
Cholesterol biosynthetic intermediates, such as lanosterol and desmosterol, are emergent immune regulators of macrophages in response to inflammatory stimuli or lipid overloading, respectively. However, the participation of these sterols in regulating macrophage functions in the physiological context of atherosclerosis, an inflammatory disease driven by the accumulation of cholesterol-laden macrophages in the artery wall, has remained elusive. Here, we report that desmosterol, the most abundant cholesterol biosynthetic intermediate in human coronary artery lesions, plays an essential role during atherogenesis, serving as a key molecule integrating cholesterol homeostasis and immune responses in macrophages. Depletion of desmosterol in myeloid cells by overexpression of 3β-hydroxysterol Δ24-reductase (DHCR24), the enzyme that catalyzes conversion of desmosterol to cholesterol, promotes the progression of atherosclerosis. Single-cell transcriptomics in isolated CD45+CD11b+ cells from atherosclerotic plaques demonstrate that depletion of desmosterol increases interferon responses and attenuates the expression of antiinflammatory macrophage markers. Lipidomic and transcriptomic analysis of in vivo macrophage foam cells demonstrate that desmosterol is a major endogenous liver X receptor (LXR) ligand involved in LXR/retinoid X receptor (RXR) activation and thus macrophage foam cell formation. Decreased desmosterol accumulation in mitochondria promotes macrophage mitochondrial reactive oxygen species production and NLR family pyrin domain containing 3 (NLRP3)-dependent inflammasome activation. Deficiency of NLRP3 or apoptosis-associated speck-like protein containing a CARD (ASC) rescues the increased inflammasome activity and atherogenesis observed in desmosterol-depleted macrophages. Altogether, these findings underscore the critical function of desmosterol in the atherosclerotic plaque to dampen inflammation by integrating with macrophage cholesterol metabolism and inflammatory activation and protecting from disease progression.
Collapse
|
12
|
Espinoza C, Fuenzalida B, Leiva A. Increased Fetal Cardiovascular Disease Risk: Potential Synergy Between Gestational Diabetes Mellitus and Maternal Hypercholesterolemia. Curr Vasc Pharmacol 2021; 19:601-623. [PMID: 33902412 DOI: 10.2174/1570161119666210423085407] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 01/27/2021] [Accepted: 03/16/2021] [Indexed: 01/25/2023]
Abstract
Cardiovascular diseases (CVD) remain a major cause of death worldwide. Evidence suggests that the risk for CVD can increase at the fetal stages due to maternal metabolic diseases, such as gestational diabetes mellitus (GDM) and maternal supraphysiological hypercholesterolemia (MSPH). GDM is a hyperglycemic, inflammatory, and insulin-resistant state that increases plasma levels of free fatty acids and triglycerides, impairs endothelial vascular tone regulation, and due to the increased nutrient transport, exposes the fetus to the altered metabolic conditions of the mother. MSPH involves increased levels of cholesterol (mainly as low-density lipoprotein cholesterol) which also causes endothelial dysfunction and alters nutrient transport to the fetus. Despite that an association has already been established between MSPH and increased CVD risk, however, little is known about the cellular processes underlying this relationship. Our knowledge is further obscured when the simultaneous presentation of MSPH and GDM takes place. In this context, GDM and MSPH may substantially increase fetal CVD risk due to synergistic impairment of placental nutrient transport and endothelial dysfunction. More studies on the separate and/or cumulative role of both processes are warranted to suggest specific treatment options.
Collapse
Affiliation(s)
- Cristian Espinoza
- Faculty of Biological Sciences, Pontificia Universidad Catolica de Chile, Santiago 8330024, Chile
| | - Barbara Fuenzalida
- Institute of Biochemistry and Molecular Medicine, University of Bern, CH-3012 Bern, Switzerland
| | - Andrea Leiva
- School of Medical Technology, Health Sciences Faculty, Universidad San Sebastian, Providencia 7510157, Chile
| |
Collapse
|
13
|
Horn CL, Morales AL, Savard C, Farrell GC, Ioannou GN. Role of Cholesterol-Associated Steatohepatitis in the Development of NASH. Hepatol Commun 2021; 6:12-35. [PMID: 34558856 PMCID: PMC8710790 DOI: 10.1002/hep4.1801] [Citation(s) in RCA: 116] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 07/08/2021] [Accepted: 07/14/2021] [Indexed: 12/11/2022] Open
Abstract
The rising prevalence of nonalcoholic fatty liver disease (NAFLD) and NAFLD-related cirrhosis in the United States and globally highlights the need to better understand the mechanisms causing progression of hepatic steatosis to fibrosing steatohepatitis and cirrhosis in a small proportion of patients with NAFLD. Accumulating evidence suggests that lipotoxicity mediated by hepatic free cholesterol (FC) overload is a mechanistic driver for necroinflammation and fibrosis, characteristic of nonalcoholic steatohepatitis (NASH), in many animal models and also in some patients with NASH. Diet, lifestyle, obesity, key genetic polymorphisms, and hyperinsulinemia secondary to insulin resistance are pivotal drivers leading to aberrant cholesterol signaling, which leads to accumulation of FC within hepatocytes. FC overload in hepatocytes can lead to ER stress, mitochondrial dysfunction, development of toxic oxysterols, and cholesterol crystallization in lipid droplets, which in turn lead to hepatocyte apoptosis, necrosis, or pyroptosis. Activation of Kupffer cells and hepatic stellate cells by hepatocyte signaling and cholesterol loading contributes to this inflammation and leads to hepatic fibrosis. Cholesterol accumulation in hepatocytes can be readily prevented or reversed by statins. Observational studies suggest that use of statins in NASH not only decreases the substantially increased cardiovascular risk, but may ameliorate liver pathology. Conclusion: Hepatic FC loading may result in cholesterol-associated steatohepatitis and play an important role in the development and progression of NASH. Statins appear to provide significant benefit in preventing progression to NASH and NASH-cirrhosis. Randomized controlled trials are needed to demonstrate whether statins or statin/ezetimibe combination can effectively reverse steatohepatitis and liver fibrosis in patients with NASH.
Collapse
Affiliation(s)
- Christian L Horn
- Division of Gastroenterology and Hepatology, Department of Medicine, San Antonio Military Medical Center, Fort Sam Houston, TX, USA
| | - Amilcar L Morales
- Division of Gastroenterology and Hepatology, Department of Medicine, San Antonio Military Medical Center, Fort Sam Houston, TX, USA
| | - Christopher Savard
- Division of Gastroenterology, Department of Medicine, Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA.,Division of Gastroenterology, Department of Medicine, University of Washington, Seattle, WA, USA.,Research and Development, Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA
| | - Geoffrey C Farrell
- Liver Research Group, ANU Medical School, Australian National University at the Canberra Hospital, Garran, ACT, Australia
| | - George N Ioannou
- Division of Gastroenterology, Department of Medicine, Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA.,Division of Gastroenterology, Department of Medicine, University of Washington, Seattle, WA, USA.,Research and Development, Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA
| |
Collapse
|
14
|
Manna LB, Williamson C. Nuclear receptors, gestational metabolism and maternal metabolic disorders. Mol Aspects Med 2021; 78:100941. [PMID: 33455843 DOI: 10.1016/j.mam.2021.100941] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Accepted: 01/04/2021] [Indexed: 12/20/2022]
Abstract
Normal pregnancy is characterised by a gradual alteration in metabolism that results in elevated serum bile acids, dyslipidaemia and impaired glucose tolerance in the third trimester. Nuclear receptors play important roles in regulating metabolic pathways that influence alterations in these parameters. There is evidence for altered function of FXR and LXR in gestation; these nuclear receptors play an integral role in bile acid and lipid homeostasis. There is some evidence for influence of clock genes in late pregnancy metabolic changes, and this may be linked to alterations in placental gene expression and function, thereby influencing fetal growth. This article will review the current data from human studies and investigation of animal models to illustrate the role of nuclear receptors (namely LXR, FXR, PPARs and clock genes) in gestational alterations in metabolism and the ways this may influence susceptibility to metabolic disorders of pregnancy such as gestational diabetes mellitus and intrahepatic cholestasis of pregnancy.
Collapse
Affiliation(s)
- Luiza Borges Manna
- Division of Women and Children's Health, King's College London, London, United Kingdom
| | - Catherine Williamson
- Division of Women and Children's Health, King's College London, London, United Kingdom.
| |
Collapse
|
15
|
Zhang X, Wang K, Zhu L, Wang Q. Reverse Cholesterol Transport Pathway and Cholesterol Efflux in Diabetic Retinopathy. J Diabetes Res 2021; 2021:8746114. [PMID: 34746320 PMCID: PMC8564209 DOI: 10.1155/2021/8746114] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 09/16/2021] [Accepted: 10/01/2021] [Indexed: 11/21/2022] Open
Abstract
Cholesterol esters, synthesized from cholesterol with long-chain fatty acids, are essential components of plasma lipoproteins and cell membranes that participate in various metabolic processes in the body. Cholesterol can be excreted through the cholesterol reverse transport (RCT) pathway when excessive cholesterol is produced in the extrahepatic cells, which is regulated by the liver X receptor (LXR) and its downstream regulators ATP-binding cassette subfamily A member 1 (ABCA1) and ATP-binding cassette subfamily G member 1 (ABCG1) genes. Abnormal cholesterol metabolism is closely associated with the development of diabetic retinopathy (DR). However, the precise underlying mechanism of the RCT pathway in the pathogenesis of DR is still not fully understood. This review focused on cholesterol metabolism, with a particular emphasis on the RCT pathway and its correlation with the development of DR. Particular attention has been paid to the key regulators of the RCT pathway: LXR, ABCA1, and ABCG1 genes and their potential therapeutic targets in the management of DR.
Collapse
Affiliation(s)
- Xinyuan Zhang
- Beijing Institute of Ophthalmology, Department of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, China
- Beijing Retinal and Choroidal Vascular Study Group, China
| | - Kaiyue Wang
- Beijing Institute of Ophthalmology, Department of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, China
| | - Ling Zhu
- Save Sight Institute, University of Sydney, Australia
| | - Qiyun Wang
- Beijing Institute of Ophthalmology, Department of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, China
- Beijing Retinal and Choroidal Vascular Study Group, China
| |
Collapse
|
16
|
Park HS, Lee K, Kim SH, Hong MJ, Jeong NJ, Kim MS. Luteolin improves hypercholesterolemia and glucose intolerance through LXRα-dependent pathway in diet-induced obese mice. J Food Biochem 2020; 44:e13358. [PMID: 32598492 DOI: 10.1111/jfbc.13358] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 06/08/2020] [Accepted: 06/08/2020] [Indexed: 01/06/2023]
Abstract
Luteolin, a naturally derived flavonoid, exerts beneficial effects such as antitumor, antioxidant, and anti-inflammatory effects. However, the molecular mechanism underlying the effect of luteolin in hypercholesterolemia remains unclear. In this study, we demonstrated that luteolin upregulated the expression of liver X receptor (LXR) α, ATP-binding cassette transporter G1 (ABCG1), and scavenger receptor class B member 1 (SRB1), which play a major role in cholesterol efflux, in HepG2 hepatocytes. Luteolin-stimulated expression of ABCG1 and SRB1 was reversed by inhibitory compound of LXRα. Luteolin administration also upregulated the expression of ABCG1, and SRB1 as well as cholesterol 7 α-hydroxylase (Cyp7α1) in the liver of diet-induced obese mice. Luteolin decreased the level of blood cholesterol and non-high-density lipoprotein cholesterol in obese mice. In addition, luteolin ameliorated glucose intolerance and reduced expression of gluconeogenesis-associated enzymes in an LXRα-dependent manner. PRACTICAL APPLICATIONS: Luteolin is known to possess various pharmacological activities. This research revealed that luteolin ameliorates hypercholesterolemia and glucose intolerance in diet-induced obesity. The results indicate that the potential properties of luteolin in cholesterol metabolism could be explained, at least in part, as being due to upregulated expression of ABCG1, and SRB1 through activation of liver X receptor, LXRα signaling pathway in HepG2 cells.
Collapse
Affiliation(s)
- Hee-Sook Park
- Research group of Healthcare, Korea Food Research Institute, Wanju-gun, Republic of Korea
| | - Kyunhee Lee
- Research group of Healthcare, Korea Food Research Institute, Wanju-gun, Republic of Korea.,Department of Food Biotechnology, Korea University of Science & Technology, Wanju-gun, Republic of Korea
| | - Soon-Hee Kim
- Research group of Healthcare, Korea Food Research Institute, Wanju-gun, Republic of Korea
| | - Moon Ju Hong
- Research group of Healthcare, Korea Food Research Institute, Wanju-gun, Republic of Korea.,Department of Food Biotechnology, Korea University of Science & Technology, Wanju-gun, Republic of Korea
| | - Nam-Joo Jeong
- Research group of Healthcare, Korea Food Research Institute, Wanju-gun, Republic of Korea
| | - Myung-Sunny Kim
- Research group of Healthcare, Korea Food Research Institute, Wanju-gun, Republic of Korea.,Department of Food Biotechnology, Korea University of Science & Technology, Wanju-gun, Republic of Korea
| |
Collapse
|
17
|
Kitano VJF, Ohyama Y, Hayashida C, Ito J, Okayasu M, Sato T, Ogasawara T, Tsujita M, Kakino A, Shimada J, Sawamura T, Hakeda Y. LDL uptake-dependent phosphatidylethanolamine translocation to the cell surface promotes fusion of osteoclast-like cells. J Cell Sci 2020; 133:jcs243840. [PMID: 32295848 DOI: 10.1242/jcs.243840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 03/25/2020] [Indexed: 11/20/2022] Open
Abstract
Osteoporosis is associated with vessel diseases attributed to hyperlipidemia, and bone resorption by multinucleated osteoclasts is related to lipid metabolism. In this study, we generated low-density lipoprotein receptor (LDLR)/lectin-like oxidized LDL receptor-1 (LOX-1, also known as Olr1) double knockout (dKO) mice. We found that, like LDLR single KO (sKO), LDLR/LOX-1 dKO impaired cell-cell fusion of osteoclast-like cells (OCLs). LDLR/LOX-1 dKO and LDLR sKO preosteoclasts exhibited decreased uptake of LDL. The cell surface cholesterol levels of both LDLR/LOX-1 dKO and LDLR sKO osteoclasts were lower than the levels of wild-type OCLs. Additionally, the amount of phosphatidylethanolamine (PE) on the cell surface was attenuated in LDLR/LOX-1 dKO and LDLR sKO preosteoclasts, whereas the PE distribution in wild-type OCLs was concentrated on the filopodia in contact with neighboring cells. Abrogation of the ATP binding cassette G1 (ABCG1) transporter, which transfers PE to the cell surface, caused decreased PE translocation to the cell surface and subsequent cell-cell fusion. The findings of this study indicate the involvement of a novel cascade (LDLR∼ABCG1∼PE translocation to cell surface∼cell-cell fusion) in multinucleation of OCLs.
Collapse
Affiliation(s)
- Victor J F Kitano
- Division of Oral Anatomy, Meikai University School of Dentistry, Sakado, Saitama 350-0283, Japan
- Division of Oral and Maxillofacial Surgery, Meikai University School of Dentistry, Sakado, Saitama 350-0283, Japan
| | - Yoko Ohyama
- Division of Oral Anatomy, Meikai University School of Dentistry, Sakado, Saitama 350-0283, Japan
- Division of Oral and Maxillofacial Surgery, Meikai University School of Dentistry, Sakado, Saitama 350-0283, Japan
| | - Chiyomi Hayashida
- Division of Oral Anatomy, Meikai University School of Dentistry, Sakado, Saitama 350-0283, Japan
| | - Junta Ito
- Josai University, Faculty of Pharmacy and Pharmaceutical Sciences, Department of Clinical Dietetics and Human Nutrition, Sakado, Saitama 350-0295, Japan
| | - Mari Okayasu
- Division of Oral-maxillofacial Surgery, Dentistry and Orthodontics, The University of Tokyo Hospital, Hongo, Tokyo 113-8655, Japan
| | - Takuya Sato
- Division of Oral Anatomy, Meikai University School of Dentistry, Sakado, Saitama 350-0283, Japan
| | - Toru Ogasawara
- Division of Oral-maxillofacial Surgery, Dentistry and Orthodontics, The University of Tokyo Hospital, Hongo, Tokyo 113-8655, Japan
| | - Maki Tsujita
- Department of Biochemistry, Graduate School of Medical Sciences, Nagoya City University, Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 467-8601, Japan
| | - Akemi Kakino
- Department of Physiology, Shinshu University School of Medicine, Matsumoto, Nagano 390-8621, Japan
| | - Jun Shimada
- Division of Oral and Maxillofacial Surgery, Meikai University School of Dentistry, Sakado, Saitama 350-0283, Japan
| | - Tatsuya Sawamura
- Department of Physiology, Shinshu University School of Medicine, Matsumoto, Nagano 390-8621, Japan
| | - Yoshiyuki Hakeda
- Division of Oral Anatomy, Meikai University School of Dentistry, Sakado, Saitama 350-0283, Japan
| |
Collapse
|
18
|
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: 3.6] [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
|
19
|
Hansmann E, Mennillo E, Yoda E, Verreault M, Barbier O, Chen S, Tukey RH. Differential Role of Liver X Receptor (LXR) α and LXR β in the Regulation of UDP-Glucuronosyltransferase 1A1 in Humanized UGT1 Mice. Drug Metab Dispos 2020; 48:255-263. [PMID: 31980500 PMCID: PMC7065491 DOI: 10.1124/dmd.119.090068] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 01/14/2020] [Indexed: 12/17/2022] Open
Abstract
Liver X receptors (LXRs), LXRα and LXRβ, are nuclear receptors that regulate the metabolism of cholesterol and bile acids and are activated by oxysterols. Humanized UGT1 (hUGT1) mice express the 9-human UGT1A genes associated with the UGT1 locus in a Ugt1-null background. The expression of UGT1A1 is developmentally delayed in the liver and intestines, resulting in the accumulation of serum bilirubin during the neonatal period. Induction of UGT1A1 in newborn hUGT1 mice leads to rapid reduction in total serum bilirubin (TSB) levels, a phenotype measurement that allows for an accurate prediction on UGT1A1 expression. When neonatal hUGT1 mice were treated by oral gavage with the LXR agonist T0901317, TSB levels were dramatically reduced. To determine the LXR contribution to the induction of UGT1A1 and the lowering of TSB levels, experiments were conducted in neonatal hUGT1/Lxrα -/- , hUGT1/Lxrβ -/- , and hUGT1/Lxrαβ -/- mice treated with T0901317. Induction of liver UGT1A1 was dependent upon LXRα, with the induction pattern paralleling induction of LXRα-specific stearoyl CoA desaturase 1. However, the actions of T0901317 were also shown to display a lack of specificity for LXR, with the induction of liver UGT1A1 in hUGT1/Lxrαβ -/- mice, a result associated with activation of both pregnane X receptor and constitutive androstane receptor. However, the LXR agonist GW3965 was highly selective toward LXRα, showing no impact on lowering TSB values or inducing UGT1A1 in hUGT1/Lxrα -/- mice. An LXR-specific enhancer site on the UGT1A1 gene was identified, along with convincing evidence that LXRα is crucial in maintaining constitutive expression of UGT1A1 in adult hUGT1 mice. SIGNIFICANCE STATEMENT: It has been established that activation of LXRα, and not LXRβ, is responsible for the induction of liver UGT1A1 and metabolism of serum bilirubin in neonatal hUGT1 mice. Although induction of the human UGT1A1 gene is initiated at a newly characterized LXR enhancer site, allelic deletion of the Lxrα gene drastically reduces the constitutive expression of liver UGT1A1 in adult hUGT1 mice. Combined, these findings indicate that LXRα is critical for the developmental expression of UGT1A1.
Collapse
Affiliation(s)
- Eva Hansmann
- Laboratory of Environmental Toxicology, Department of Pharmacology, University of California, San Diego, La Jolla, California (E.H., E.M., E.Y., S.C., R.H.T.); Division of Health Chemistry, Department of Healthcare and Regulatory Sciences, School of Pharmacy, Showa University, Shinagawa-ku, Tokyo, Japan (E.Y.); and Laboratory of Moléculaire Pharmacology, Centre de Recherche du CHU de Québec, Faculté of Pharmacie, Université Laval Québec, Québec, Canada (M.V., O.B.)
| | - Elvira Mennillo
- Laboratory of Environmental Toxicology, Department of Pharmacology, University of California, San Diego, La Jolla, California (E.H., E.M., E.Y., S.C., R.H.T.); Division of Health Chemistry, Department of Healthcare and Regulatory Sciences, School of Pharmacy, Showa University, Shinagawa-ku, Tokyo, Japan (E.Y.); and Laboratory of Moléculaire Pharmacology, Centre de Recherche du CHU de Québec, Faculté of Pharmacie, Université Laval Québec, Québec, Canada (M.V., O.B.)
| | - Emiko Yoda
- Laboratory of Environmental Toxicology, Department of Pharmacology, University of California, San Diego, La Jolla, California (E.H., E.M., E.Y., S.C., R.H.T.); Division of Health Chemistry, Department of Healthcare and Regulatory Sciences, School of Pharmacy, Showa University, Shinagawa-ku, Tokyo, Japan (E.Y.); and Laboratory of Moléculaire Pharmacology, Centre de Recherche du CHU de Québec, Faculté of Pharmacie, Université Laval Québec, Québec, Canada (M.V., O.B.)
| | - Mélanie Verreault
- Laboratory of Environmental Toxicology, Department of Pharmacology, University of California, San Diego, La Jolla, California (E.H., E.M., E.Y., S.C., R.H.T.); Division of Health Chemistry, Department of Healthcare and Regulatory Sciences, School of Pharmacy, Showa University, Shinagawa-ku, Tokyo, Japan (E.Y.); and Laboratory of Moléculaire Pharmacology, Centre de Recherche du CHU de Québec, Faculté of Pharmacie, Université Laval Québec, Québec, Canada (M.V., O.B.)
| | - Olivier Barbier
- Laboratory of Environmental Toxicology, Department of Pharmacology, University of California, San Diego, La Jolla, California (E.H., E.M., E.Y., S.C., R.H.T.); Division of Health Chemistry, Department of Healthcare and Regulatory Sciences, School of Pharmacy, Showa University, Shinagawa-ku, Tokyo, Japan (E.Y.); and Laboratory of Moléculaire Pharmacology, Centre de Recherche du CHU de Québec, Faculté of Pharmacie, Université Laval Québec, Québec, Canada (M.V., O.B.)
| | - Shujuan Chen
- Laboratory of Environmental Toxicology, Department of Pharmacology, University of California, San Diego, La Jolla, California (E.H., E.M., E.Y., S.C., R.H.T.); Division of Health Chemistry, Department of Healthcare and Regulatory Sciences, School of Pharmacy, Showa University, Shinagawa-ku, Tokyo, Japan (E.Y.); and Laboratory of Moléculaire Pharmacology, Centre de Recherche du CHU de Québec, Faculté of Pharmacie, Université Laval Québec, Québec, Canada (M.V., O.B.)
| | - Robert H Tukey
- Laboratory of Environmental Toxicology, Department of Pharmacology, University of California, San Diego, La Jolla, California (E.H., E.M., E.Y., S.C., R.H.T.); Division of Health Chemistry, Department of Healthcare and Regulatory Sciences, School of Pharmacy, Showa University, Shinagawa-ku, Tokyo, Japan (E.Y.); and Laboratory of Moléculaire Pharmacology, Centre de Recherche du CHU de Québec, Faculté of Pharmacie, Université Laval Québec, Québec, Canada (M.V., O.B.)
| |
Collapse
|
20
|
Sun MMG, Beier F. Liver X Receptor activation regulates genes involved in lipid homeostasis in developing chondrocytes. OSTEOARTHRITIS AND CARTILAGE OPEN 2020; 2:100030. [DOI: 10.1016/j.ocarto.2020.100030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 01/16/2020] [Indexed: 02/07/2023] Open
|
21
|
LongShengZhi Capsule Reduces Established Atherosclerotic Lesions in apoE-Deficient Mice by Ameliorating Hepatic Lipid Metabolism and Inhibiting Inflammation. J Cardiovasc Pharmacol 2020; 73:105-117. [PMID: 30540683 DOI: 10.1097/fjc.0000000000000642] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Disorders of lipid metabolism and inflammation play an important role in atherosclerosis. LongShengZhi (LSZ) capsule, a Chinese herbal medicine, has been used for treatment of patients with vascular diseases for many years. In this article, we determined the effect of LSZ on the progression of established atherosclerotic lesions in apoE-deficient (apoE) mice. ApoE mice were prefed high-fat diet (HFD) for 8 weeks to induce atherosclerosis, then started with LSZ treatment contained in HFD for 10 weeks. Although LSZ had little effect on HFD-induced hypercholesterolemia, it substantially reduced en face and sinus aortic lesions. The reduction of lesions was associated with reduced macrophage/foam cell accumulation by activating ABCA1/ABCG1 expression. LSZ maintained the integrity of arterial wall by increasing collagen or smooth muscle cell content and inhibiting cell apoptosis. LSZ also attenuated HFD-induced fatty liver by down-regulating expression of lipogenic and cholesterol synthetic genes while activating expression of triglyceride catabolism genes. Moreover, LSZ demonstrated potent anti-inflammatory effects. In vivo, LSZ reduced serum TNF-α levels, infiltration of neutrophils, Kupffer cells, and expression of inflammatory cytokines in the liver. In vitro, it inhibited lipopolysaccharide or palmitate-induced expression of inflammatory cytokines in macrophages. Therefore, LSZ reduces atherosclerosis by ameliorating hepatic lipid metabolism and inhibiting inflammation.
Collapse
|
22
|
Lake NJ, Taylor RL, Trahair H, Harikrishnan KN, Curran JE, Almeida M, Kulkarni H, Mukhamedova N, Hoang A, Low H, Murphy AJ, Johnson MP, Dyer TD, Mahaney MC, Göring HHH, Moses EK, Sviridov D, Blangero J, Jowett JBM, Bozaoglu K. TRAK2, a novel regulator of ABCA1 expression, cholesterol efflux and HDL biogenesis. Eur Heart J 2019; 38:3579-3587. [PMID: 28655204 DOI: 10.1093/eurheartj/ehx315] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 05/25/2017] [Indexed: 12/28/2022] Open
Abstract
Aims The recent failures of HDL-raising therapies have underscored our incomplete understanding of HDL biology. Therefore there is an urgent need to comprehensively investigate HDL metabolism to enable the development of effective HDL-centric therapies. To identify novel regulators of HDL metabolism, we performed a joint analysis of human genetic, transcriptomic, and plasma HDL-cholesterol (HDL-C) concentration data and identified a novel association between trafficking protein, kinesin binding 2 (TRAK2) and HDL-C concentration. Here we characterize the molecular basis of the novel association between TRAK2 and HDL-cholesterol concentration. Methods and results Analysis of lymphocyte transcriptomic data together with plasma HDL from the San Antonio Family Heart Study (n = 1240) revealed a significant negative correlation between TRAK2 mRNA levels and HDL-C concentration, HDL particle diameter and HDL subspecies heterogeneity. TRAK2 siRNA-mediated knockdown significantly increased cholesterol efflux to apolipoprotein A-I and isolated HDL from human macrophage (THP-1) and liver (HepG2) cells by increasing the mRNA and protein expression of the cholesterol transporter ATP-binding cassette, sub-family A member 1 (ABCA1). The effect of TRAK2 knockdown on cholesterol efflux was abolished in the absence of ABCA1, indicating that TRAK2 functions in an ABCA1-dependent efflux pathway. TRAK2 knockdown significantly increased liver X receptor (LXR) binding at the ABCA1 promoter, establishing TRAK2 as a regulator of LXR-mediated transcription of ABCA1. Conclusion We show, for the first time, that TRAK2 is a novel regulator of LXR-mediated ABCA1 expression, cholesterol efflux, and HDL biogenesis. TRAK2 may therefore be an important target in the development of anti-atherosclerotic therapies.
Collapse
Affiliation(s)
- Nicole J Lake
- Baker IDI Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia.,Murdoch Children's Research Institute, 50 Flemington Road, Parkville, VIC 3052, Australia
| | - Rachael L Taylor
- Baker IDI Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
| | - Hugh Trahair
- Baker IDI Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
| | - K N Harikrishnan
- Baker IDI Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia.,Department of Pathology, University of Melbourne, Grattan Street, Parkville, VIC 3010, Australia
| | - Joanne E Curran
- South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, One West University Blvd. Brownsville, Texas 78520, USA
| | - Marcio Almeida
- South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, One West University Blvd. Brownsville, Texas 78520, USA
| | - Hemant Kulkarni
- South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, One West University Blvd. Brownsville, Texas 78520, USA
| | - Nigora Mukhamedova
- Baker IDI Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
| | - Anh Hoang
- Baker IDI Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
| | - Hann Low
- Baker IDI Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
| | - Andrew J Murphy
- Baker IDI Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
| | - Matthew P Johnson
- South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, One West University Blvd. Brownsville, Texas 78520, USA
| | - Thomas D Dyer
- South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, One West University Blvd. Brownsville, Texas 78520, USA
| | - Michael C Mahaney
- South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, One West University Blvd. Brownsville, Texas 78520, USA
| | - Harald H H Göring
- South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, One West University Blvd. Brownsville, Texas 78520, USA
| | - Eric K Moses
- University of Western Australia, 35 Stirling Hwy, Crawley, WA 6009, Australia.,Curtin University, Kent St, Bentley, WA 6102, Australia
| | - Dmitri Sviridov
- Baker IDI Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
| | - John Blangero
- South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, One West University Blvd. Brownsville, Texas 78520, USA
| | - Jeremy B M Jowett
- Baker IDI Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
| | - Kiymet Bozaoglu
- Baker IDI Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia.,Murdoch Children's Research Institute, 50 Flemington Road, Parkville, VIC 3052, Australia.,Department of Paediatrics, University of Melbourne, 50 Flemington Road, Parkville,VIC 3052, Australia
| |
Collapse
|
23
|
Wang D, Yang Y, Lei Y, Tzvetkov NT, Liu X, Yeung AWK, Xu S, Atanasov AG. Targeting Foam Cell Formation in Atherosclerosis: Therapeutic Potential of Natural Products. Pharmacol Rev 2019; 71:596-670. [PMID: 31554644 DOI: 10.1124/pr.118.017178] [Citation(s) in RCA: 146] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Foam cell formation and further accumulation in the subendothelial space of the vascular wall is a hallmark of atherosclerotic lesions. Targeting foam cell formation in the atherosclerotic lesions can be a promising approach to treat and prevent atherosclerosis. The formation of foam cells is determined by the balanced effects of three major interrelated biologic processes, including lipid uptake, cholesterol esterification, and cholesterol efflux. Natural products are a promising source for new lead structures. Multiple natural products and pharmaceutical agents can inhibit foam cell formation and thus exhibit antiatherosclerotic capacity by suppressing lipid uptake, cholesterol esterification, and/or promoting cholesterol ester hydrolysis and cholesterol efflux. This review summarizes recent findings on these three biologic processes and natural products with demonstrated potential to target such processes. Discussed also are potential future directions for studying the mechanisms of foam cell formation and the development of foam cell-targeted therapeutic strategies.
Collapse
Affiliation(s)
- Dongdong Wang
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Yang Yang
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Yingnan Lei
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Nikolay T Tzvetkov
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Xingde Liu
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Andy Wai Kan Yeung
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Suowen Xu
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Atanas G Atanasov
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| |
Collapse
|
24
|
Grinman DY, Careaga VP, Wellberg EA, Dansey MV, Kordon EC, Anderson SM, Maier MS, Burton G, MacLean PS, Rudolph MC, Pecci A. Liver X receptor-α activation enhances cholesterol secretion in lactating mammary epithelium. Am J Physiol Endocrinol Metab 2019; 316:E1136-E1145. [PMID: 30964702 PMCID: PMC6620573 DOI: 10.1152/ajpendo.00548.2018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 04/01/2019] [Accepted: 04/04/2019] [Indexed: 11/22/2022]
Abstract
Liver X receptors (LXRs) are ligand-dependent transcription factors activated by cholesterol metabolites. These receptors induce a suite of target genes required for de novo synthesis of triglycerides and cholesterol transport in many tissues. Two different isoforms, LXRα and LXRβ, have been well characterized in liver, adipocytes, macrophages, and intestinal epithelium among others, but their contribution to cholesterol and fatty acid efflux in the lactating mammary epithelium is poorly understood. We hypothesize that LXR regulates lipogenesis during milk fat production in lactation. Global mRNA analysis of mouse mammary epithelial cells (MECs) revealed multiple LXR/RXR targets upregulated sharply early in lactation compared with midpregnancy. LXRα is the primary isoform, and its protein levels increase throughout lactation in MECs. The LXR agonist GW3965 markedly induced several genes involved in cholesterol transport and lipogenesis and enhanced cytoplasmic lipid droplet accumulation in the HC11 MEC cell line. Importantly, in vivo pharmacological activation of LXR increased the milk cholesterol percentage and induced sterol regulatory element-binding protein 1c (Srebp1c) and ATP-binding cassette transporter a7 (Abca7) expression in MECs. Cumulatively, our findings identify LXRα as an important regulator of cholesterol incorporation into the milk through key nodes of de novo lipogenesis, suggesting a potential therapeutic target in women with difficulty initiating lactation.
Collapse
Affiliation(s)
- Diego Y Grinman
- Instituto de Fisiología, Biología Molecular y Neurociencias, CONICET, Universidad de Buenos Aires , Buenos Aires , Argentina
| | - Valeria P Careaga
- Unidad De Microanálisis Y Métodos Físicos Aplicados a la Química Orgánica, CONICET, Universidad de Buenos Aires , Buenos Aires , Argentina
- Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Química Orgánica, Facultad de Ciencias Exactas y Naturales , Universidad de Buenos Aires, Buenos Aires , Argentina
| | - Elizabeth A Wellberg
- Department of Pathology, University of Colorado, Anschutz Medical Campus, Denver, Colorado
| | - María V Dansey
- Unidad De Microanálisis Y Métodos Físicos Aplicados a la Química Orgánica, CONICET, Universidad de Buenos Aires , Buenos Aires , Argentina
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires , Buenos Aires , Argentina
| | - Edith C Kordon
- Instituto de Fisiología, Biología Molecular y Neurociencias, CONICET, Universidad de Buenos Aires , Buenos Aires , Argentina
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires , Buenos Aires , Argentina
| | - Steven M Anderson
- Department of Pathology, University of Colorado, Anschutz Medical Campus, Denver, Colorado
| | - Marta S Maier
- Unidad De Microanálisis Y Métodos Físicos Aplicados a la Química Orgánica, CONICET, Universidad de Buenos Aires , Buenos Aires , Argentina
- Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Química Orgánica, Facultad de Ciencias Exactas y Naturales , Universidad de Buenos Aires, Buenos Aires , Argentina
| | - Gerardo Burton
- Unidad De Microanálisis Y Métodos Físicos Aplicados a la Química Orgánica, CONICET, Universidad de Buenos Aires , Buenos Aires , Argentina
- Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Química Orgánica, Facultad de Ciencias Exactas y Naturales , Universidad de Buenos Aires, Buenos Aires , Argentina
| | - Paul S MacLean
- Division of Endocrinology, Metabolism, and Diabetes, University of Colorado, Anschutz Medical Campus, Denver, Colorado
| | - Michael C Rudolph
- Division of Endocrinology, Metabolism, and Diabetes, University of Colorado, Anschutz Medical Campus, Denver, Colorado
| | - Adali Pecci
- Instituto de Fisiología, Biología Molecular y Neurociencias, CONICET, Universidad de Buenos Aires , Buenos Aires , Argentina
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires , Buenos Aires , Argentina
| |
Collapse
|
25
|
Saida–Tamiya K, Tamiya M, Sekiya G, Isobe K, Kitazawa T, Isaka N, Matsukawa A, Kawahara K, Komuro A, Ishiguro M. Structural requirements of cholenamide derivatives as the LXR ligands. Bioorg Med Chem Lett 2019; 29:1330-1335. [DOI: 10.1016/j.bmcl.2019.03.051] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 03/15/2019] [Accepted: 03/30/2019] [Indexed: 11/27/2022]
|
26
|
Leonurine: From Gynecologic Medicine to Pleiotropic Agent. Chin J Integr Med 2019; 26:152-160. [DOI: 10.1007/s11655-019-3453-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/05/2018] [Indexed: 12/11/2022]
|
27
|
Bang BR, Li M, Tsai KN, Aoyagi H, Lee SA, Machida K, Aizaki H, Jung JU, Ou JHJ, Saito T. Regulation of Hepatitis C Virus Infection by Cellular Retinoic Acid Binding Proteins through the Modulation of Lipid Droplet Abundance. J Virol 2019; 93:e02302-18. [PMID: 30728260 PMCID: PMC6450116 DOI: 10.1128/jvi.02302-18] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 01/31/2019] [Indexed: 02/08/2023] Open
Abstract
Retinoid (vitamin A) is an essential diet constituent that governs a broad range of biological processes. Its biologically active metabolite, all-trans retinoic acid (ATRA), exhibits a potent antiviral property by enhancing both innate and adaptive antiviral immunity against a variety of viral pathogens, such as, but not limited to, HIV, respiratory syncytial virus (RSV), herpes simplex virus (HSV), and measles. Even though the hepatocyte is highly enriched with retinoid and its metabolite ATRA, it supports the establishment of efficient hepatitis C virus (HCV) replication. Here, we demonstrate the hepatocyte-specific cell-intrinsic mechanism by which ATRA exerts either a proviral or antiviral effect, depending on how it engages cellular retinoic acid binding proteins (CRABPs). We found that the engagement of CRABP1 by ATRA potently supported viral infection by promoting the accumulation of lipid droplets (LDs), which robustly enhanced the formation of a replication complex on the LD-associated endoplasmic reticulum (ER) membrane. In contrast, ATRA binding to CRABP2 potently inhibited HCV via suppression of LD accumulation. However, this antiviral effect of CRABP2 was abrogated due to the functional and quantitative predominance of CRABP1 in the hepatocytes. In summary, our study demonstrates that CRABPs serve as an on-off switch that modulates the efficiency of the HCV life cycle and elucidates how HCV evades the antiviral properties of ATRA via the exploitation of CRABP1 functionality.IMPORTANCE ATRA, a biologically active metabolite of vitamin A, exerts pleiotropic biological effects, including the activation of both innate and adaptive immunity, thereby serving as a potent antimicrobial compound against numerous viral pathogens. Despite the enrichment of hepatocytes with vitamin A, HCV still establishes an efficient viral life cycle. Here, we discovered that the hepatocellular response to ATRA creates either a proviral or an antiviral environment depending on its engagement with CRABP1 or -2, respectively. CRABP1 supports the robust replication of HCV, while CRABP2 potently inhibits the efficiency of viral replication. Our biochemical, genetic, and microscopic analyses reveal that the pro- and antiviral effects of CRABPs are mediated by modulation of LD abundance, where HCV establishes the platform for viral replication and assembly on the LD-associated ER membrane. This study uncovered a cell-intrinsic mechanism by which HCV exploits the proviral function of CRABP1 to establish an efficient viral life cycle.
Collapse
Affiliation(s)
- Bo-Ram Bang
- Department of Medicine, Division of Gastrointestinal and Liver Diseases, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Meng Li
- Bioinformatics Service, Norris Medical Library, University of Southern California, Los Angeles, California, USA
| | - Kuen-Nan Tsai
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Haruyo Aoyagi
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, Japan
| | - Shin-Ae Lee
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Keigo Machida
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
- Southern California Research Center for ALPD and Cirrhosis, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Hideki Aizaki
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, Japan
| | - Jae U Jung
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Jing-Hsiung James Ou
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Takeshi Saito
- Department of Medicine, Division of Gastrointestinal and Liver Diseases, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
- USC Research Center for Liver Diseases, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
- Southern California Research Center for ALPD and Cirrhosis, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
28
|
Liu J, Guo K, Hu L, Luo T, Ma Y, Zhang Y, Lai W, Guo Z. ZAP70 deficiency promotes reverse cholesterol transport through MAPK/ERK pathway in Jurkat cell. Mol Immunol 2019; 107:21-28. [PMID: 30639475 DOI: 10.1016/j.molimm.2019.01.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 12/12/2018] [Accepted: 01/02/2019] [Indexed: 01/02/2023]
Abstract
BACKGROUND Lots of studies have demonstrated that immune cells could regulate reverse cholesterol transport (RCT). However, neither T cell receptor (TCR) signalling nor Zeta-chain associated protein 70 (ZAP70) have been demonstrated to be associated with RCT. To investigate this association, we used a ZAP70-deficient Jurkat-derived mutant, P116 cell line, to detect the effect of ZAP70 on RCT and inflammatory response. ZAP70 deficiency improved cholesterol efflux capacity by 14%. Meanwhile, mRNA and proteins expression of RCT regulatory proteins such as ABCA1, ABCG1 and SR-BI were increased in P116 cells. ZAP70-deficiency had no influence on LXR-α and PPAR-γ. Regarding the inflammatory response, the mRNA expression and secretion of pro-atherosclerotic cytokines, TNF-α, IFN-γ, IL-2 and IL-6, were significantly decreased in the ZAP70-deficient cell line. Activation of MAP kinases cascades, as determined by of ERK, JNK and p38 MAPK phosphorylation, were found to be inhibited in the absence of ZAP70. Specific inhibition of ERK, JNK and p38 MAPK activity was also found to decreased TNF-α, IFN-γ, and IL-6 secretion. However, only the ERK inhibition was observed to reduce IL-2 secretion, improve cholesterol efflux capacity and increase expression of ABCA1, ABCG1 and SR-BI without increasing LXR-α and PPAR-γ. Using ChIP assay to detect the binding of LXR-α to LXRE, which promotes the expression of ABCG1, we found that inhibiting ERK improved binding without increasing LXR-α levels. Thus, we speculate that ZAP70-deficiency may improve RCT and decrease the inflammatory response of T cells. Furthermore, these effects are probably achieved via ERK signalling pathway.
Collapse
Affiliation(s)
- Jichen Liu
- Department of Cardiology, Huiqiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, People's Republic of China
| | - Kai Guo
- Department of Cardiology, Huiqiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, People's Republic of China
| | - Lu Hu
- Department of Cardiology, Huiqiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, People's Republic of China
| | - Tiantian Luo
- Department of Cardiology, Huiqiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, People's Republic of China
| | - Yusheng Ma
- Department of Cardiology, Huiqiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, People's Republic of China
| | - Yanan Zhang
- Department of Cardiology, Huiqiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, People's Republic of China
| | - Wenyan Lai
- Department of Cardiology, Huiqiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, People's Republic of China
| | - Zhigang Guo
- Department of Cardiology, Huiqiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, People's Republic of China.
| |
Collapse
|
29
|
Yu XH, Zhang DW, Zheng XL, Tang CK. Cholesterol transport system: An integrated cholesterol transport model involved in atherosclerosis. Prog Lipid Res 2018; 73:65-91. [PMID: 30528667 DOI: 10.1016/j.plipres.2018.12.002] [Citation(s) in RCA: 150] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 10/30/2018] [Accepted: 12/01/2018] [Indexed: 02/07/2023]
Abstract
Atherosclerosis, the pathological basis of most cardiovascular disease (CVD), is closely associated with cholesterol accumulation in the arterial intima. Excessive cholesterol is removed by the reverse cholesterol transport (RCT) pathway, representing a major antiatherogenic mechanism. In addition to the RCT, other pathways are required for maintaining the whole-body cholesterol homeostasis. Thus, we propose a working model of integrated cholesterol transport, termed the cholesterol transport system (CTS), to describe body cholesterol metabolism. The novel model not only involves the classical view of RCT but also contains other steps, such as cholesterol absorption in the small intestine, low-density lipoprotein uptake by the liver, and transintestinal cholesterol excretion. Extensive studies have shown that dysfunctional CTS is one of the major causes for hypercholesterolemia and atherosclerosis. Currently, several drugs are available to improve the CTS efficiently. There are also several therapeutic approaches that have entered into clinical trials and shown considerable promise for decreasing the risk of CVD. In recent years, a variety of novel findings reveal the molecular mechanisms for the CTS and its role in the development of atherosclerosis, thereby providing novel insights into the understanding of whole-body cholesterol transport and metabolism. In this review, we summarize the latest advances in this area with an emphasis on the therapeutic potential of targeting the CTS in CVD patients.
Collapse
Affiliation(s)
- Xiao-Hua Yu
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Medical Research Experiment Center, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, China
| | - Da-Wei Zhang
- Department of Pediatrics and Group on the Molecular and Cell Biology of Lipids, University of Alberta, Alberta, Canada
| | - Xi-Long Zheng
- Department of Biochemistry and Molecular Biology, Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, University of Calgary, Health Sciences Center, 3330 Hospital Dr NW, Calgary, Alberta T2N 4N1, Canada
| | - Chao-Ke Tang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Medical Research Experiment Center, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, China.
| |
Collapse
|
30
|
Anastasius M, Luquain-Costaz C, Kockx M, Jessup W, Kritharides L. A critical appraisal of the measurement of serum 'cholesterol efflux capacity' and its use as surrogate marker of risk of cardiovascular disease. Biochim Biophys Acta Mol Cell Biol Lipids 2018; 1863:1257-1273. [PMID: 30305243 DOI: 10.1016/j.bbalip.2018.08.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 08/02/2018] [Accepted: 08/03/2018] [Indexed: 12/15/2022]
Abstract
The 'cholesterol efflux capacity (CEC)' assay is a simple in vitro measure of the capacities of individual sera to promote the first step of the reverse cholesterol transport pathway, the delivery of cellular cholesterol to plasma HDL. This review describes the cell biology of this model and critically assesses its application as a marker of cardiovascular risk. We describe the pathways for cell cholesterol export, current cell models used in the CEC assay with their limitations and consider the contribution that measurement of serum CEC provides to our understanding of HDL function in vivo.
Collapse
Affiliation(s)
- Malcolm Anastasius
- ANZAC Research Institute, Concord Repatriation General Hospital, University of Sydney, Sydney, NSW, Australia
| | | | - Maaike Kockx
- ANZAC Research Institute, Concord Repatriation General Hospital, University of Sydney, Sydney, NSW, Australia
| | - Wendy Jessup
- ANZAC Research Institute, Concord Repatriation General Hospital, University of Sydney, Sydney, NSW, Australia
| | - Leonard Kritharides
- ANZAC Research Institute, Concord Repatriation General Hospital, University of Sydney, Sydney, NSW, Australia; Cardiology Department, Concord Repatriation General Hospital, University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
31
|
Critical Role of the Human ATP-Binding Cassette G1 Transporter in Cardiometabolic Diseases. Int J Mol Sci 2017; 18:ijms18091892. [PMID: 28869506 PMCID: PMC5618541 DOI: 10.3390/ijms18091892] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 08/30/2017] [Accepted: 08/30/2017] [Indexed: 12/15/2022] Open
Abstract
ATP-binding cassette G1 (ABCG1) is a member of the large family of ABC transporters which are involved in the active transport of many amphiphilic and lipophilic molecules including lipids, drugs or endogenous metabolites. It is now well established that ABCG1 promotes the export of lipids, including cholesterol, phospholipids, sphingomyelin and oxysterols, and plays a key role in the maintenance of tissue lipid homeostasis. Although ABCG1 was initially proposed to mediate cholesterol efflux from macrophages and then to protect against atherosclerosis and cardiovascular diseases (CVD), it becomes now clear that ABCG1 exerts a larger spectrum of actions which are of major importance in cardiometabolic diseases (CMD). Beyond a role in cellular lipid homeostasis, ABCG1 equally participates to glucose and lipid metabolism by controlling the secretion and activity of insulin and lipoprotein lipase. Moreover, there is now a growing body of evidence suggesting that modulation of ABCG1 expression might contribute to the development of diabetes and obesity, which are major risk factors of CVD. In order to provide the current understanding of the action of ABCG1 in CMD, we here reviewed major findings obtained from studies in mice together with data from the genetic and epigenetic analysis of ABCG1 in the context of CMD.
Collapse
|
32
|
Cocci P, Mosconi G, Palermo FA. Pregnane X receptor (PXR) signaling in seabream primary hepatocytes exposed to extracts of seawater samples collected from polycyclic aromatic hydrocarbons (PAHs)-contaminated coastal areas. MARINE ENVIRONMENTAL RESEARCH 2017; 130:181-186. [PMID: 28760623 DOI: 10.1016/j.marenvres.2017.07.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 07/04/2017] [Accepted: 07/18/2017] [Indexed: 06/07/2023]
Abstract
Polycyclic aromatic hydrocarbons (PAHs) are persistent organic pollutants damaging to the marine environment and the wildlife. Herein, we investigated the effects of extracts from coastal seawaters (central Adriatic sea, Italy), showing high concentrations of PAHs, on pregnane X receptor (PXR)-transcriptional regulation of the cytochrome P450 3A (CYP3A) gene using seabream primary hepatocytes. The results show that concentrated extracts of seawater with original ΣPAH concentrations above the putative threshold of 30 ng L-1 increased expression of PXR and its main target gene, CYP3A. Similar results were observed for LXR and its target gene SREBP-1c suggesting pathway cross-talk. These data are further supported by the finding of multiple PXR and LXR response elements in the putative promoters of their target genes. Overall, our data indicate the capacity of seawater extracts, containing environmentally relevant levels of PAHs, to affect multiple pathways, including lipid and cholesterol metabolism.
Collapse
Affiliation(s)
- Paolo Cocci
- School of Biosciences and Veterinary Medicine, University of Camerino, Via Gentile III Da Varano, I-62032 Camerino, MC, Italy
| | - Gilberto Mosconi
- School of Biosciences and Veterinary Medicine, University of Camerino, Via Gentile III Da Varano, I-62032 Camerino, MC, Italy
| | - Francesco Alessandro Palermo
- School of Biosciences and Veterinary Medicine, University of Camerino, Via Gentile III Da Varano, I-62032 Camerino, MC, Italy.
| |
Collapse
|
33
|
Kober AC, Manavalan APC, Tam-Amersdorfer C, Holmér A, Saeed A, Fanaee-Danesh E, Zandl M, Albrecher NM, Björkhem I, Kostner GM, Dahlbäck B, Panzenboeck U. Implications of cerebrovascular ATP-binding cassette transporter G1 (ABCG1) and apolipoprotein M in cholesterol transport at the blood-brain barrier. Biochim Biophys Acta Mol Cell Biol Lipids 2017; 1862:573-588. [PMID: 28315462 DOI: 10.1016/j.bbalip.2017.03.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 03/07/2017] [Accepted: 03/12/2017] [Indexed: 02/03/2023]
Abstract
Impaired cholesterol/lipoprotein metabolism is linked to neurodegenerative diseases such as Alzheimer's disease (AD). Cerebral cholesterol homeostasis is maintained by the highly efficient blood-brain barrier (BBB) and flux of the oxysterols 24(S)-hydroxycholesterol and 27-hydroxycholesterol, potent liver-X-receptor (LXR) activators. HDL and their apolipoproteins are crucial for cerebral lipid transfer, and loss of ATP binding cassette transporters (ABC)G1 and G4 results in toxic accumulation of oxysterols in the brain. The HDL-associated apolipoprotein (apo)M is positively correlated with pre-β HDL formation in plasma; its presence and function in the brain was thus far unknown. Using an in vitro model of the BBB, we examined expression, regulation, and functions of ABCG1, ABCG4, and apoM in primary porcine brain capillary endothelial cells (pBCEC). RT Q-PCR analyses and immunoblotting revealed that in addition to ABCA1 and scavenger receptor, class B, type I (SR-BI), pBCEC express high levels of ABCG1, which was up-regulated by LXR activation. Immunofluorescent staining, site-specific biotinylation and immunoprecipitation revealed that ABCG1 is localized both to early and late endosomes and on apical and basolateral plasma membranes. Using siRNA interference to silence ABCG1 (by 50%) reduced HDL-mediated [3H]-cholesterol efflux (by 50%) but did not reduce [3H]-24(S)-hydroxycholesterol efflux. In addition to apoA-I, pBCEC express and secrete apoM mainly to the basolateral (brain) compartment. HDL enhanced expression and secretion of apoM by pBCEC, apoM-enriched HDL promoted cellular cholesterol efflux more efficiently than apoM-free HDL, while apoM-silencing diminished cellular cholesterol release. We suggest that ABCG1 and apoM are centrally involved in regulation of cholesterol metabolism/turnover at the BBB.
Collapse
Affiliation(s)
| | | | | | - Andreas Holmér
- Department of Translational Medicine, University Hospital SUS, Malmö, Lund University, Sweden
| | - Ahmed Saeed
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska University Hospital, Karolinska Institute Huddinge, Huddinge, Sweden
| | - Elham Fanaee-Danesh
- Institute of Pathophysiology and Immunology, Medical University of Graz, Graz, Austria
| | - Martina Zandl
- Institute of Pathophysiology and Immunology, Medical University of Graz, Graz, Austria
| | | | - Ingemar Björkhem
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska University Hospital, Karolinska Institute Huddinge, Huddinge, Sweden
| | - Gerhard M Kostner
- Institute of Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Björn Dahlbäck
- Department of Translational Medicine, University Hospital SUS, Malmö, Lund University, Sweden
| | - Ute Panzenboeck
- Institute of Pathophysiology and Immunology, Medical University of Graz, Graz, Austria.
| |
Collapse
|
34
|
de Aguiar Vallim TQ, Lee E, Merriott DJ, Goulbourne CN, Cheng J, Cheng A, Gonen A, Allen RM, Palladino END, Ford DA, Wang T, Baldán Á, Tarling EJ. ABCG1 regulates pulmonary surfactant metabolism in mice and men. J Lipid Res 2017; 58:941-954. [PMID: 28264879 DOI: 10.1194/jlr.m075101] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 03/03/2017] [Indexed: 12/27/2022] Open
Abstract
Idiopathic pulmonary alveolar proteinosis (PAP) is a rare lung disease characterized by accumulation of surfactant. Surfactant synthesis and secretion are restricted to epithelial type 2 (T2) pneumocytes (also called T2 cells). Clearance of surfactant is dependent upon T2 cells and macrophages. ABCG1 is highly expressed in both T2 cells and macrophages. ABCG1-deficient mice accumulate surfactant, lamellar body-loaded T2 cells, lipid-loaded macrophages, B-1 lymphocytes, and immunoglobulins, clearly demonstrating that ABCG1 has a critical role in pulmonary homeostasis. We identify a variant in the ABCG1 promoter in patients with PAP that results in impaired activation of ABCG1 by the liver X receptor α, suggesting that ABCG1 basal expression and/or induction in response to sterol/lipid loading is essential for normal lung function. We generated mice lacking ABCG1 specifically in either T2 cells or macrophages to determine the relative contribution of these cell types on surfactant lipid homeostasis. These results establish a critical role for T2 cell ABCG1 in controlling surfactant and overall lipid homeostasis in the lung and in the pathogenesis of human lung disease.
Collapse
Affiliation(s)
- Thomas Q de Aguiar Vallim
- Department of Medicine, University of California Los Angeles, Los Angeles, CA 90095.,Department of Biological Chemistry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095.,Molecular Biology Institute, University of California Los Angeles, Los Angeles, CA 90095.,Johnson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA 90095
| | - Elinor Lee
- Department of Medicine, University of California Los Angeles, Los Angeles, CA 90095.,Division of Pulmonary and Critical Care Medicine, University of California Los Angeles, Los Angeles, CA 90095
| | - David J Merriott
- Department of Biological Chemistry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095
| | | | - Joan Cheng
- Department of Biological Chemistry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095
| | - Angela Cheng
- Department of Biological Chemistry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095
| | - Ayelet Gonen
- Department of Medicine, University of California San Diego, La Jolla, CA 92093
| | - Ryan M Allen
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University, St. Louis, MO 63104
| | - Elisa N D Palladino
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University, St. Louis, MO 63104.,Center for Cardiovascular Research, School of Medicine, Saint Louis University, St. Louis, MO 63104
| | - David A Ford
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University, St. Louis, MO 63104.,Center for Cardiovascular Research, School of Medicine, Saint Louis University, St. Louis, MO 63104
| | - Tisha Wang
- Department of Medicine, University of California Los Angeles, Los Angeles, CA 90095.,Division of Pulmonary and Critical Care Medicine, University of California Los Angeles, Los Angeles, CA 90095
| | - Ángel Baldán
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University, St. Louis, MO 63104
| | - Elizabeth J Tarling
- Department of Medicine, University of California Los Angeles, Los Angeles, CA 90095 .,Molecular Biology Institute, University of California Los Angeles, Los Angeles, CA 90095.,Johnson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA 90095
| |
Collapse
|
35
|
Cao X, Zhang L, Chen C, Wang Q, Guo L, Ma Q, Deng P, Zhu G, Li B, Pi Y, Long C, Zhang L, Yu Z, Zhou Z, Li J. The critical role of ABCG1 and PPARγ/LXRα signaling in TLR4 mediates inflammatory responses and lipid accumulation in vascular smooth muscle cells. Cell Tissue Res 2016; 368:145-157. [DOI: 10.1007/s00441-016-2518-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 09/27/2016] [Indexed: 12/15/2022]
|
36
|
Zhang L, Chen Y, Yang X, Yang J, Cao X, Li X, Li L, Miao QR, Hajjar DP, Duan Y, Han J. MEK1/2 inhibitors activate macrophage ABCG1 expression and reverse cholesterol transport-An anti-atherogenic function of ERK1/2 inhibition. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:1180-1191. [PMID: 27365310 DOI: 10.1016/j.bbalip.2016.06.017] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 06/03/2016] [Accepted: 06/24/2016] [Indexed: 02/07/2023]
Abstract
Expression of ATP-binding cassette transporter G1 (ABCG1), a molecule facilitating cholesterol efflux to HDL, is activated by liver X receptor (LXR). In this study, we investigated if inhibition of ERK1/2 can activate macrophage ABCG1 expression and functions. MEK1/2 inhibitors, PD98059 and U0126, increased ABCG1 mRNA and protein expression, and activated the natural ABCG1 promoter but not the promoter with the LXR responsive element (LXRE) deletion. Inhibition of ABCG1 expression by ABCG1 siRNA did enhance the formation of macrophage/foam cells and it attenuated the inhibitory effect of MEK1/2 inhibitors on foam cell formation. MEK1/2 inhibitors activated macrophage cholesterol efflux to HDL in vitro, and they enhanced reverse cholesterol transport (RCT) in vivo. ApoE deficient (apoE(-/-)) mice receiving U0126 treatment had reduced sinus lesions in the aortic root which was associated with activated macrophage ABCG1 expression in the lesion areas. MEK1/2 inhibitors coordinated the RXR agonist, but not the LXR agonist, to induce ABCG1 expression. Furthermore, induction of ABCG1 expression by MEK1/2 inhibitors was associated with activation of SIRT1, a positive regulator of LXR activity, and inactivation of SULT2B1 and RIP140, two negative regulators of LXR activity. Taken together, our study suggests that MEK1/2 inhibitors activate macrophage ABCG1 expression/RCT, and inhibit foam cell formation and lesion development by multiple mechanisms, supporting the concept that ERK1/2 inhibition is anti-atherogenic.
Collapse
Affiliation(s)
- Ling Zhang
- Department of Cardiology, Xijing Hospital, the 4th Military Medical University, Xi'an, China
| | - Yuanli Chen
- College of Biomedical Engineering, Hefei University of Technology, Hefei, China; School of Medicine, Nankai University, Tianjin, China
| | - Xiaoxiao Yang
- College of Life Sciences, Nankai University, Tianjin, China
| | - Jie Yang
- College of Life Sciences, Nankai University, Tianjin, China
| | - Xingyue Cao
- College of Life Sciences, Nankai University, Tianjin, China
| | - Xiaoju Li
- College of Life Sciences, Nankai University, Tianjin, China
| | - Luyuan Li
- College of Pharmacy, Nankai University, Tianjin, China
| | | | | | - Yajun Duan
- College of Biomedical Engineering, Hefei University of Technology, Hefei, China; College of Life Sciences, Nankai University, Tianjin, China; State Key Laboratory of Medicinal Chemical Biology, Collaborative Innovation Center of Biotherapy, Nankai University, Tianjin, China.
| | - Jihong Han
- College of Biomedical Engineering, Hefei University of Technology, Hefei, China; College of Life Sciences, Nankai University, Tianjin, China; State Key Laboratory of Medicinal Chemical Biology, Collaborative Innovation Center of Biotherapy, Nankai University, Tianjin, China.
| |
Collapse
|
37
|
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: 1.1] [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
|
38
|
Lipid droplet-associated proteins in atherosclerosis (Review). Mol Med Rep 2016; 13:4527-34. [PMID: 27082419 PMCID: PMC4878557 DOI: 10.3892/mmr.2016.5099] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 01/29/2016] [Indexed: 01/01/2023] Open
Abstract
Accumulation of atherosclerotic plaques in arterial walls leads to major cardiovascular diseases and stroke. Macrophages/foam cells are central components of atherosclerotic plaques, which populate the arterial wall in order to remove harmful modified low‑density lipoprotein (LDL) particles, resulting in the accumulation of lipids, mostly LDL‑derived cholesterol ester, in cytosolic lipid droplets (LDs). At present, LDs are recognized as dynamic organelles that govern cellular metabolic processes. LDs consist of an inner core of neutral lipids surrounded by a monolayer of phospholipids and free cholesterol, and contain LD‑associated proteins (LDAPs) that regulate LD functions. Foam cells are characterized by an aberrant accumulation of cytosolic LDs, and are considered a hallmark of atherosclerotic lesions through all stages of development. Previous studies have investigated the mechanisms underlying foam cell formation, aiming to discover therapeutic strategies that target foam cells and intervene against atherosclerosis. It is well established that LDAPs have a major role in the pathogenesis of metabolic diseases caused by dysfunction of lipid metabolism, and several studies have linked LDAPs to the development of atherosclerosis. In this review, several foam cell‑targeting pathways have been described, with an emphasis on the role of LDAPs in cholesterol mobilization from macrophages. In addition, the potential of LDAPs as therapeutic targets to prevent the progression and/or facilitate the regression of the disease has been discussed.
Collapse
|
39
|
Retinoic acid induces macrophage cholesterol efflux and inhibits atherosclerotic plaque formation in apoE-deficient mice. Br J Nutr 2015. [PMID: 26201974 DOI: 10.1017/s0007114515002159] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
It has been suggested that retinoic acid (RA) has a potential role in the prevention of atherosclerotic CVD. In the present study, we used J774A.1 cell lines and primary peritoneal macrophages to investigate the protective effects of RA on foam cell formation and atherogenesis in apoE-deficient (apoE- / -) mice. A total of twenty male apoE- / - mice (n 10 animals per group), aged 8 weeks, were fed on a high-fat diet (HFD) and treated with vehicle or 9-cis-RA for 8 weeks. The atherosclerotic plaque area in the aortic sinus of mice in the 9-cis-RA group was 40·7 % less than that of mice in the control group (P< 0·01). Mouse peritoneal macrophages from the 9-cis-RA group had higher protein expression levels of ATP-binding cassette transporter A1 (ABCA1) and G1 (ABCG1) than those from the control group. Serum total and LDL-cholesterol concentrations were lower in the 9-cis-RA group than in the control group (P< 0·05). In vitro studies showed that incubation of cholesterol-loaded J774A.1 macrophages with 9-cis-RA (0·1, 1 and 10 μmol/l) induced cholesterol efflux in a dose-dependent manner. The 9-cis-RA treatment markedly attenuated lipid accumulation in macrophages exposed to oxidised LDL. Moreover, treatment with 9-cis-RA significantly increased the protein expression levels of ABCA1 and ABCG1 in J774A.1 macrophages in a dose-dependent manner. Furthermore, 9-cis-RA dose-dependently enhanced the protein expression level of liver X receptor-α (LXRα), the upstream regulator of ABCA1 and ABCG1. Taken together, the present results show that 9-cis-RA suppresses foam cell formation and prevents HFD-induced atherogenesis via the LXRα-dependent up-regulation of ABCA1 and ABCG1.
Collapse
|
40
|
Lanuti M, Talamonti E, Maccarrone M, Chiurchiù V. Activation of GPR55 Receptors Exacerbates oxLDL-Induced Lipid Accumulation and Inflammatory Responses, while Reducing Cholesterol Efflux from Human Macrophages. PLoS One 2015; 10:e0126839. [PMID: 25970609 PMCID: PMC4430319 DOI: 10.1371/journal.pone.0126839] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 04/08/2015] [Indexed: 11/18/2022] Open
Abstract
The G protein-coupled receptor GPR55 has been proposed as a new cannabinoid receptor associated with bone remodelling, nervous system excitability, vascular homeostasis as well as in several pathophysiological conditions including obesity and cancer. However, its physiological role and underlying mechanism remain unclear. In the present work, we demonstrate for the first time its presence in human macrophages and its increased expression in ox-LDL-induced foam cells. In addition, pharmacological activation of GPR55 by its selective agonist O-1602 increased CD36- and SRB-I-mediated lipid accumulation and blocked cholesterol efflux by downregulating ATP-binding cassette (ABC) transporters ABCA1 and ABCG1, as well as enhanced cytokine- and pro-metalloprotease-9 (pro-MMP-9)-induced proinflammatory responses in foam cells. Treatment with cannabidiol, a selective antagonist of GPR55, counteracted these pro-atherogenic and proinflammatory O-1602-mediated effects. Our data suggest that GPR55 could play deleterious role in ox-LDL-induced foam cells and could be a novel pharmacological target to manage atherosclerosis and other related cardiovascular diseases.
Collapse
Affiliation(s)
- Mirko Lanuti
- European Center for Brain Research (CERC), IRCCS, Santa Lucia Foundation, Rome, Italy
| | - Emanuela Talamonti
- European Center for Brain Research (CERC), IRCCS, Santa Lucia Foundation, Rome, Italy
- Center of Integrated Research, Campus Bio-Medico University of Rome, Rome, Italy
| | - Mauro Maccarrone
- European Center for Brain Research (CERC), IRCCS, Santa Lucia Foundation, Rome, Italy
- Center of Integrated Research, Campus Bio-Medico University of Rome, Rome, Italy
- * E-mail: (VC); (MM)
| | - Valerio Chiurchiù
- European Center for Brain Research (CERC), IRCCS, Santa Lucia Foundation, Rome, Italy
- Center of Integrated Research, Campus Bio-Medico University of Rome, Rome, Italy
- * E-mail: (VC); (MM)
| |
Collapse
|
41
|
Abstract
Dyslipidaemia is a major risk factor for cardiovascular diseases. Pharmacological lowering of LDL-C levels using statins reduces cardiovascular risk. However, a substantial residual risk persists especially in patients with type 2 diabetes mellitus. Because of the inverse association observed in epidemiological studies of HDL-C with the risk for cardiovascular diseases, novel therapeutic strategies to raise HDL-C levels or improve HDL functionality are developed as complementary therapy for cardiovascular diseases. However, until now most therapies targeting HDL-C levels failed in clinical trials because of side effects or absence of clinical benefits. This chapter will highlight the emerging small molecules currently developed and tested in clinical trials to pharmacologically modulate HDL-C and functionality including new CETP inhibitors (anacetrapib, evacetrapib), novel PPAR agonists (K-877, CER-002, DSP-8658, INT131 and GFT505), LXR agonists (ATI-111, LXR-623, XL-652) and RVX-208.
Collapse
|
42
|
Uto-Kondo H, Ayaori M, Nakaya K, Takiguchi S, Yakushiji E, Ogura M, Terao Y, Ozasa H, Sasaki M, Komatsu T, Sotherden GM, Hosoai T, Sakurada M, Ikewaki K. Citrulline increases cholesterol efflux from macrophages in vitro and ex vivo via ATP-binding cassette transporters. J Clin Biochem Nutr 2014; 55:32-9. [PMID: 25120277 PMCID: PMC4078065 DOI: 10.3164/jcbn.13-76] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Accepted: 04/13/2014] [Indexed: 01/27/2023] Open
Abstract
Reverse cholesterol transport (RCT) is a mechanism critical to the anti-atherogenic property of HDL. Although citrulline contributes to the amelioration of atherosclerosis via endothelial nitric oxide production, it remains unclear whether it affects RCT. This study was undertaken to clarify the effects of citrulline on expressions of specific transporters such as ATP binding cassette transporters (ABC)A1 and ABCG1, and the cholesterol efflux from macrophages to apolipoprotein (apo) A-I or HDL in vitro and ex vivo. Citrulline increased ABCA1 and ABCG1 mRNA and protein levels in THP-1 macrophages, translating into enhanced apoA-I- and HDL-mediated cholesterol efflux. In the human crossover study, 8 healthy male volunteers (age 30-49 years) consumed either 3.2 g/day citrulline or placebo for 1 week. Citrulline consumption brought about significant increases in plasma levels of citrulline and arginine. Supporting the in vitro data, monocyte-derived macrophages (MDM) differentiated under autologous post-citrulline sera demonstrated enhancement of both apoA-I- and HDL-mediated cholesterol efflux through increased ABCA1 and ABCG1 expressions, compared to MDM differentiated under pre-citrulline sera. However, the placebo did not modulate these parameters. Therefore, in addition to improving endothelium function, citrulline might have an anti-atherogenic property by increasing RCT of HDL.
Collapse
Affiliation(s)
- Harumi Uto-Kondo
- Division of Anti-aging and Vascular Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan
| | - Makoto Ayaori
- Division of Anti-aging and Vascular Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan
| | - Kazuhiro Nakaya
- Division of Anti-aging and Vascular Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan
| | - Shunichi Takiguchi
- Division of Anti-aging and Vascular Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan
| | - Emi Yakushiji
- Division of Anti-aging and Vascular Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan
| | - Masatsune Ogura
- Division of Anti-aging and Vascular Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan
| | - Yoshio Terao
- Division of Cardiology, Jikei University School of Medicine, 3-25-8 Nishi-Shinbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Hideki Ozasa
- Division of Cardiology, Jikei University School of Medicine, 3-25-8 Nishi-Shinbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Makoto Sasaki
- Division of Anti-aging and Vascular Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan
| | - Tomohiro Komatsu
- Division of Anti-aging and Vascular Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan
| | - Grace Megumi Sotherden
- Division of Anti-aging and Vascular Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan
| | - Tamaki Hosoai
- Division of Anti-aging and Vascular Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan
| | - Masami Sakurada
- Tokorozawa Heart Center, 2-6-61 Kamiarai, Tokorozawa, Saitama 359-1142, Japan
| | - Katsunori Ikewaki
- Division of Anti-aging and Vascular Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan
| |
Collapse
|
43
|
Gu HM, Wang FQ, Zhang DW. Caveolin-1 interacts with ATP binding cassette transporter G1 (ABCG1) and regulates ABCG1-mediated cholesterol efflux. Biochim Biophys Acta Mol Cell Biol Lipids 2014; 1841:847-58. [DOI: 10.1016/j.bbalip.2014.02.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Revised: 02/06/2014] [Accepted: 02/12/2014] [Indexed: 01/19/2023]
|
44
|
Li N, Xu Y, Feng T, Liu C, Li Y, Wang X, Si S. Identification of a Selective Agonist for Liver X Receptor α (LXRα) via Screening of a Synthetic Compound Library. ACTA ACUST UNITED AC 2013; 19:566-74. [DOI: 10.1177/1087057113516004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Liver X receptor α (LXRα) plays an important role in reverse cholesterol transport (RCT), and activation of LXRα could reduce atherosclerosis. In the present study, we developed a screening method to identify new potential LXRα agonists using an LXRα-GAL4 chimera reporter assay. A novel analogue of N,N-disubstituted 2,8-diazaspiro[4.5]decane, IMB-151, was identified as an LXRα agonist by using this method. IMB-151 showed a significant activation effect on LXRα, with an EC50 value of 1.47 µM. IMB-151 also increased the expression of ATP-binding cassette transporter A1 (ABCA1) and G1 (ABCG1) in RAW264.7 macrophages. The upregulating effects of IMB-151 on ABCA1 and ABCG1 markedly decreased when coincubated with geranylgeranyl pyrophosphate (GGPP) ammonium salt or LXRα small interfering RNA (siRNA). Our data indicated that the upregulation of ABCA1 and ABCG1 by IMB-151 depended on activation of LXRα. Moreover, IMB-151 significantly reduced cellular lipid accumulation and increased cholesterol efflux in RAW264.7 macrophages. Interestingly, IMB-151 slightly increased sterol response element binding protein 1c (SREBP-1c) protein expression levels in HepG2 cells compared with TO901317, and this indicated that IMB-151 might have less lipogenesis side effect in vivo. These results suggested that IMB-151 was identified as a selective agonist for LXRα by using a screening method and could be used as a potential antiatherosclerotic lead compound in the future.
Collapse
Affiliation(s)
- Ni Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yanni Xu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tingting Feng
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chang Liu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yongzhen Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiao Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shuyi Si
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
45
|
Higham A, Lea S, Plumb J, Maschera B, Simpson K, Ray D, Singh D. The role of the liver X receptor in chronic obstructive pulmonary disease. Respir Res 2013; 14:106. [PMID: 24118845 PMCID: PMC3852990 DOI: 10.1186/1465-9921-14-106] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Accepted: 09/25/2013] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND There is a need for novel anti-inflammatory therapies to treat COPD. The liver X receptor (LXR) is a nuclear hormone receptor with anti-inflammatory properties. METHODS We investigated LXR gene and protein expression levels in alveolar macrophages and whole lung tissue from COPD patients and controls, the effect of LXR activation on the suppression of inflammatory mediators from LPS stimulated COPD alveolar macrophages, and the effect of LXR activation on the induction of genes associated with alternative macrophage polarisation. RESULTS The levels of LXR mRNA were significantly increased in whole lung tissue extracts in COPD patients and smokers compared to non-smokers. The expression of LXR protein was significantly increased in small airway epithelium and alveolar epithelium in COPD patients compared to controls. No differences in LXR mRNA and protein levels were observed in alveolar macrophages between patient groups. The LXR agonist GW3965 significantly induced the expression of the LXR dependent genes ABCA1 and ABCG1 in alveolar macrophage cultures. In LPS stimulated alveolar macrophages, GW3965 suppressed the production of CXCL10 and CCL5, whilst stimulating IL-10 production. CONCLUSIONS GW3965 did not significantly suppress the production of TNFα, IL-1β, or CXCL8. Our major finding is that LXR activation has anti-inflammatory effects on CXC10, CCL5 and IL-10 production from alveolar macrophages.
Collapse
Affiliation(s)
- Andrew Higham
- The University of Manchester, NIHR Translational Research Facility, University Hospital of South Manchester Foundation Trust, Southmoor Road, Manchester M23 9LT, UK
| | - Simon Lea
- The University of Manchester, NIHR Translational Research Facility, University Hospital of South Manchester Foundation Trust, Southmoor Road, Manchester M23 9LT, UK
| | - Jonathan Plumb
- The University of Manchester, NIHR Translational Research Facility, University Hospital of South Manchester Foundation Trust, Southmoor Road, Manchester M23 9LT, UK
| | - Barbara Maschera
- GlaxoSmithKline, Respiratory CEDD, Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, UK
| | - Karen Simpson
- GlaxoSmithKline, Respiratory CEDD, Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, UK
| | - David Ray
- The University of Manchester, NIHR Translational Research Facility, University Hospital of South Manchester Foundation Trust, Southmoor Road, Manchester M23 9LT, UK
| | - Dave Singh
- The University of Manchester, NIHR Translational Research Facility, University Hospital of South Manchester Foundation Trust, Southmoor Road, Manchester M23 9LT, UK
| |
Collapse
|
46
|
D'Amore S, Vacca M, Graziano G, D'Orazio A, Cariello M, Martelli N, Di Tullio G, Salvia R, Grandaliano G, Belfiore A, Pellegrini F, Palasciano G, Moschetta A. Nuclear receptors expression chart in peripheral blood mononuclear cells identifies patients with Metabolic Syndrome. Biochim Biophys Acta Mol Basis Dis 2013; 1832:2289-301. [PMID: 24060638 DOI: 10.1016/j.bbadis.2013.09.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Revised: 09/11/2013] [Accepted: 09/15/2013] [Indexed: 01/27/2023]
Abstract
BACKGROUND Nuclear receptors are a class of 48 ligand-activated transcription factors identified as key players of metabolic and developmental processes. Most of these receptors are potential targets for pharmacological strategies in the Metabolic Syndrome. In the present study, we analyzed changes in the mRNA expression of nuclear receptors in the peripheral blood mononuclear cells of patients with Metabolic Syndrome, in order to identify novel biomarkers of disease and candidate targets for putative therapeutical approaches. METHODS AND RESULTS We enrolled thirty healthy controls (14 M:16 F) and thirty naïve patients (16 M: 14 F; >3 criteria for Metabolic Syndrome upon Adult Treatment Panel III) without organ damage. Using quantitative real-time PCR, we assessed the expression patterns of nuclear receptors in peripheral blood mononuclear cells. 33/48 nuclear receptors were expressed in peripheral blood mononuclear cells. In patients with Metabolic Syndrome, we found a significant down-regulation of the entire PPAR, NR4A and RAR families, together with a repression of RXRα, VDR, and Rev-Erbα. Furthermore, we performed a novel statistical analysis with classification trees, which allowed us to depict a predictive core of nuclear receptor expression patterns characterizing subjects with Metabolic Syndrome. Random Forest Analysis identified NOR1 and PPARδ, which were both reduced in peripheral blood mononuclear cells and specifically in CD14(+) cells (mostly monocytes), as classifiers of Metabolic Syndrome, with high specificity and sensitivity. CONCLUSIONS Our results point to the use of PPAR and NR4A mRNA levels in the overall peripheral blood mononuclear cells as biomarkers of Metabolic Syndrome and bona fide putative targets of pharmacological therapy.
Collapse
Affiliation(s)
- Simona D'Amore
- Clinica Medica "A. Murri", "Aldo Moro" University of Bari, Italy; National Cancer Research Center, IRCCS Oncologico Giovanni Paolo II, Bari, Italy; Laboratory of Lipid Metabolism and Cancer, Consorzio Mario Negri Sud, Santa Maria Imbaro (Chieti), Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Adiponectin increases macrophages cholesterol efflux and suppresses foam cell formation in patients with type 2 diabetes mellitus. Atherosclerosis 2013; 229:62-70. [DOI: 10.1016/j.atherosclerosis.2013.01.017] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Revised: 01/04/2013] [Accepted: 01/15/2013] [Indexed: 01/05/2023]
|
48
|
Pannu PS, Allahverdian S, Francis GA. Oxysterol generation and liver X receptor-dependent reverse cholesterol transport: not all roads lead to Rome. Mol Cell Endocrinol 2013; 368:99-107. [PMID: 22884520 DOI: 10.1016/j.mce.2012.07.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2012] [Revised: 06/30/2012] [Accepted: 07/27/2012] [Indexed: 12/31/2022]
Abstract
Cell cholesterol metabolism is a tightly regulated process, dependent in part on activation of nuclear liver X receptors (LXRs) to increase expression of genes mediating removal of excess cholesterol from cells in the reverse cholesterol transport pathway. LXRs are thought to be activated predominantly by oxysterols generated enzymatically from cholesterol in different cell organelles. Defects resulting in slowed release of cholesterol from late endosomes and lysosomes or reduction in sterol-27-hydroxylase activity lead to specific blocks in oxysterol production and impaired LXR-dependent gene activation. This block does not appear to be compensated by oxysterol production in other cell compartments. The purpose of this review is to summarize current knowledge about oxysterol-dependent activation by LXR of genes involved in reverse cholesterol transport, and what these defects of cell cholesterol homeostasis can teach us about the critical pathways of oxysterol generation for expression of LXR-dependent genes.
Collapse
Affiliation(s)
- Parveer S Pannu
- Department of Medicine, UBC James Hogg Research Centre, Institute of Heart and Lung Health at St. Paul's Hospital, Vancouver, BC, Canada V6Z 1Y6.
| | | | | |
Collapse
|
49
|
Abstract
PURPOSE OF REVIEW To offer a comprehensive review on the role of ABCG1 in cellular sterol homeostasis. RECENT FINDINGS Early studies with Abcg1 mice indicated that ABCG1 was crucial for tissue lipid homeostasis, especially in the lung. More recent studies have demonstrated that loss of ABCG1 has wide-ranging consequences and impacts lymphocyte and stem cell proliferation, endothelial cell function, macrophage foam cell formation, as well as insulin secretion from pancreatic β cells. Recent studies have also demonstrated that ABCG1 functions as an intracellular lipid transporter, localizes to intracellular vesicles/endosomes, and that the transmembrane domains are sufficient for localization and transport function. SUMMARY ABCG1 plays a crucial role in maintaining intracellular sterol and lipid homeostasis. Loss of this transporter has significant, cell-type-specific consequences ranging from effects on cellular proliferation, to surfactant production and/or insulin secretion. Elucidation of the mechanisms by which ABCG1 affects intracellular sterol flux/movement should provide important information that may link ABCG1 to diseases of dysregulated tissue lipid homeostasis.
Collapse
Affiliation(s)
- Elizabeth J Tarling
- Departments of Biological Chemistry and Medicine, David Geffen School of Medicine at UCLA, University of California, Los Angeles, CA 90095-1737, USA.
| |
Collapse
|
50
|
Yoon HS, Ju JH, Lee JE, Park HJ, Lee JM, Shin HK, Holzapfel W, Park KY, Do MS. The probiotic Lactobacillus rhamnosus BFE5264 and Lactobacillus plantarum NR74 promote cholesterol efflux and suppress inflammation in THP-1 cells. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2013; 93:781-787. [PMID: 22806829 DOI: 10.1002/jsfa.5797] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Revised: 05/15/2012] [Accepted: 06/06/2012] [Indexed: 06/01/2023]
Abstract
BACKGROUND The balance between the rate of cholesterol uptake/accumulation and the rate of cholesterol efflux is reflected in the amount of lipid accumulation in macrophages. Based upon the fact that liver X receptors (LXRs) play a role in cholesterol efflux, we studied the effects of probiotics on cholesterol efflux and anti-inflammatory action in macrophages. We confirmed changes in LXR expression by treatment of LXR-transfected CHO-K1 cells with lactic acid bacteria (LAB), and co-cultured THP-1 cells with LAB to investigate changes in cholesterol efflux and inflammation. RESULTS The experiment with CHO-K1 cells showed upregulation of LXR-β by LAB. Treatment of THP-1 cells with LAB promoted LXR expression in THP-1, which eventually led to significant upregulation of ABCA1 and ABCG1 expression. The treatment with live LAB also significantly promoted cholesterol efflux. LAB suppressed expression of interleukin (IL)-1β and tumor necrosis factor (TNF)-α, which resulted from activation of LXR. CONCLUSION Our study shows that Lactobacillus rhamnosus BFE5264 and Lactobacillus plantarum NR74 activated LXR and induced cholesterol efflux by promoting expression of ABCA1 and ABCG1. Both strains also suppressed proinflammatory cytokines including IL-1β and TNF-α. This study could account for the observation that LAB may block foam cell formation by cholesterol efflux and immune modulation.
Collapse
Affiliation(s)
- Hong-sup Yoon
- School of Life Science, Handong Global University, Pohang, Gyeongbuk 791-708, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|