1
|
Butyrate Lowers Cellular Cholesterol through HDAC Inhibition and Impaired SREBP-2 Signalling. Int J Mol Sci 2022; 23:ijms232415506. [PMID: 36555149 PMCID: PMC9779842 DOI: 10.3390/ijms232415506] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 11/25/2022] [Accepted: 11/30/2022] [Indexed: 12/13/2022] Open
Abstract
In animal studies, HDAC inhibitors such as butyrate have been reported to reduce plasma cholesterol, while conferring protection from diabetes, but studies on the underlying mechanisms are lacking. This study compares the influence of butyrate and other HDAC inhibitors to that of statins on cholesterol metabolism in multiple cell lines, but primarily in HepG2 hepatic cells due to the importance of the liver in cholesterol metabolism. Sodium butyrate reduced HepG2 cholesterol content, as did sodium valproate and the potent HDAC inhibitor trichostatin A, suggesting HDAC inhibition as the exacting mechanism. In contrast to statins, which increase SREBP-2 regulated processes, HDAC inhibition downregulated SREBP-2 targets such as HMGCR and the LDL receptor. Moreover, in contrast to statin treatment, butyrate did not increase cholesterol uptake by HepG2 cells, consistent with its failure to increase LDL receptor expression. Sodium butyrate also reduced ABCA1 and SRB1 protein expression in HepG2 cells, but these effects were not consistent across all cell types. Overall, the underlying mechanism of cell cholesterol lowering by sodium butyrate and HDAC inhibition is consistent with impaired SREBP-2 signalling, and calls into question the possible use of butyrate for lowering of serum LDL cholesterol in humans.
Collapse
|
2
|
Inflammatory gene silencing in activated monocytes by a cholesterol tagged-miRNA/siRNA: a novel approach to ameliorate diabetes induced inflammation. Cell Tissue Res 2022; 389:219-240. [PMID: 35604451 DOI: 10.1007/s00441-022-03637-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 05/09/2022] [Indexed: 11/02/2022]
Abstract
There is a major unmet need for the development of effective therapies for diabetes induced inflammation. Increased adenosine-uridine rich elements (AREs) containing mRNAs of inflammatory molecules are reported in inflamed monocytes. Destabilizing these inflammatory mRNAs by the miR-16 could reduce inflammation. DNA microarrays and in vitro cell studies showed that exogenous miR16 and its mimic treatment, in LPS/PMA induced monocytes, significantly downregulated several ARE containing inflammatory cytokine mRNAs similar to those seen in the normal monocytes. Ingenuity pathway analyses showed exogenous miR-16 or its synthetic mimic treatment alleviates inflammatory responses. To selectively target uptake, especially to inflamed cells, one of the CD36 substrate cholesterol was tagged to miR16/siRNA. Cholesterol tagged miR-16/ARE-siRNA showed enhanced uptake in CD36 expressing inflamed cells. In LPS or PMA, treated monocytes, candidate genes expressions levels such as IL-6, IL-8, IL-12β, IP-10, and TNF-α mRNA were increased, as measured by RT-qPCR as seen in primary monocytes of diabetes patients. Exogenous miR16 or ARE-siRNA transfection reduced mRNAs of pro-inflammatory cytokines levels in monocyte, and its adhesion. Increased uptake of cholesterol tagged miR-16 through the CD36 receptor was observed. This destabilizes numerous inflammatory ARE containing mRNAs and alleviates inflammatory responses. Cholesterol-tagged miR-16 and its mimic are novel anti-inflammatory molecules that can be specifically targeted to, via through CD36 expressing, "inflamed" cells and thus serve as therapeutic candidates to alleviate inflammatory diseases.
Collapse
|
3
|
Dotan I, Yang J, Ikeda J, Roth Z, Pollock-Tahiri E, Desai H, Sivasubramaniyam T, Rehal S, Rapps J, Li YZ, Le H, Farber G, Alchami E, Xiao C, Karim S, Gronda M, Saikali MF, Tirosh A, Wagner KU, Genest J, Schimmer AD, Gupta V, Minden MD, Cummins CL, Lewis GF, Robbins C, Jongstra-Bilen J, Cybulsky M, Woo M. Macrophage Jak2 deficiency accelerates atherosclerosis through defects in cholesterol efflux. Commun Biol 2022; 5:132. [PMID: 35169231 PMCID: PMC8847578 DOI: 10.1038/s42003-022-03078-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Accepted: 01/26/2022] [Indexed: 12/11/2022] Open
Abstract
Atherosclerosis is a chronic inflammatory condition in which macrophages play a major role. Janus kinase 2 (JAK2) is a pivotal molecule in inflammatory and metabolic signaling, and Jak2V617F activating mutation has recently been implicated with enhancing clonal hematopoiesis and atherosclerosis. To determine the essential in vivo role of macrophage (M)-Jak2 in atherosclerosis, we generate atherosclerosis-prone ApoE-null mice deficient in M-Jak2. Contrary to our expectation, these mice exhibit increased plaque burden with no differences in macrophage proliferation, recruitment or bone marrow clonal expansion. Notably, M-Jak2-deficient bone marrow derived macrophages show a significant defect in cholesterol efflux. Pharmacologic JAK2 inhibition with ruxolitinib also leads to defects in cholesterol efflux and accelerates atherosclerosis. Liver X receptor agonist abolishes the efflux defect and attenuates the accelerated atherosclerosis that occurs with M-Jak2 deficiency. Macrophages of individuals with the Jak2V617F mutation show increased efflux which is normalized when treated with a JAK2 inhibitor. Together, M-Jak2-deficiency leads to accelerated atherosclerosis primarily through defects in cholesterol efflux from macrophages.
Collapse
Affiliation(s)
- Idit Dotan
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada.,Institute of Endocrinology, Beilinson Campus, Rabin Medical Center, Petach Tikva, Israel
| | - Jiaqi Yang
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
| | - Jiro Ikeda
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
| | - Ziv Roth
- Program in Cell Biology, Peter Gilgan Centre for Research and Learning, Hospital for Sick Children, Toronto, Canada
| | - Evan Pollock-Tahiri
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
| | - Harsh Desai
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
| | | | - Sonia Rehal
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
| | - Josh Rapps
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
| | - Yu Zhe Li
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
| | - Helen Le
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
| | - Gedaliah Farber
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
| | - Edouard Alchami
- Department of Immunology, University of Toronto, Toronto, Canada
| | - Changting Xiao
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
| | - Saraf Karim
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
| | - Marcela Gronda
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Michael F Saikali
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, Canada
| | - Amit Tirosh
- Endocrine Cancer Genomics Center, Sheba Medical Center, Tel Hashomer, Israel
| | - Kay-Uwe Wagner
- Department of Oncology, Wayne State University School of Medicine and Tumor Biology Program, Barbara Ann Karmanos Cancer Institute, Detroit, MI, USA
| | - Jacques Genest
- Research Institute of the McGill University Health Centre, Royal Victoria Hospital, Montreal, QC, Canada
| | - Aaron D Schimmer
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Vikas Gupta
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Mark D Minden
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Carolyn L Cummins
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, Canada
| | - Gary F Lewis
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
| | - Clinton Robbins
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada.,Department of Immunology, University of Toronto, Toronto, Canada
| | - Jenny Jongstra-Bilen
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada.,Department of Immunology, University of Toronto, Toronto, Canada
| | - Myron Cybulsky
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada.,Department of Immunology, University of Toronto, Toronto, Canada
| | - Minna Woo
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada. .,Department of Immunology, University of Toronto, Toronto, Canada. .,Division of Endocrinology and Metabolism, Department of Medicine, University Health Network and Sinai Health System, University of Toronto, Toronto, Canada.
| |
Collapse
|
4
|
Oleacein may intensify the efflux of oxLDL from human macrophages by increasing the expression of the SRB1 receptor, as well as ABCA1 and ABCG1 transporters. J Funct Foods 2021. [DOI: 10.1016/j.jff.2021.104373] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
5
|
Khiabani Rad M, Vazifeh Shiran N, Mohammadi MH, Hamidpour M. Evaluating the effect of cinnamon and rosuvastatin, on the formation of foam cells in macrophages co-cultured with platelets. ADVANCES IN TRADITIONAL MEDICINE 2021. [DOI: 10.1007/s13596-020-00537-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
6
|
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
|
7
|
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
|
8
|
Wang Y, Tang W, Yang P, Shin H, Li Q. Hepatic NPC1L1 promotes hyperlipidemia in LDL receptor deficient mice. Biochem Biophys Res Commun 2018; 499:626-633. [DOI: 10.1016/j.bbrc.2018.03.200] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Accepted: 03/27/2018] [Indexed: 10/17/2022]
|
9
|
Yang F, Du Y, Zhang J, Jiang Z, Wang L, Hong B. Low-density lipoprotein upregulate SR-BI through Sp1 Ser702 phosphorylation in hepatic cells. BIOCHIMICA ET BIOPHYSICA ACTA 2016; 1861:1066-1075. [PMID: 27320013 DOI: 10.1016/j.bbalip.2016.06.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 05/04/2016] [Accepted: 06/10/2016] [Indexed: 01/25/2023]
Abstract
Scavenger receptor class B type I (SR-BI) is one of the key proteins in the process of reverse cholesterol transport (RCT), and its major function is to uptake high density lipoprotein (HDL) cholesterol from plasma into liver cells. The regulation of SR-BI expression is important for controlling serum lipid content and reducing the risks of cardiovascular diseases. Here we found that SR-BI expression was significantly increased by LDL in vivo and in vitro, and the transcription factor specific protein 1 (Sp1) plays a critical role in this process. Results from co-immunoprecipitation experiments indicate that the activation of SR-BI was associated with Sp1-recruited protein complexes in the promoter region of SR-BI, where histone acetyltransferase p300 was recruited and histone deacetylase HDAC1 was dismissed. As a result, histone acetylation increased, leading to activation of SR-BI transcription. With further investigation, we found that LDL phosphorylated Sp1 through ERK1/2 pathway, which affected Sp1 protein complexes formation in SR-BI promoter. Using mass spectrometry and site directed mutagenesis, a new Sp1 phosphorylation site Ser702 was defined to be associated with Sp1-HDAC1 interaction and may be important in SR-BI activation, shedding light on the knowledge of delicate mechanism of hepatic HDL receptor SR-BI gene modulation by LDL.
Collapse
Affiliation(s)
- Fan Yang
- Key Laboratory of Biotechnology of Antibiotics of Ministry of Health, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Tiantan Xili, Beijing 100050, China
| | - Yu Du
- Key Laboratory of Biotechnology of Antibiotics of Ministry of Health, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Tiantan Xili, Beijing 100050, China
| | - Jin Zhang
- Key Laboratory of Biotechnology of Antibiotics of Ministry of Health, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Tiantan Xili, Beijing 100050, China
| | - Zhibo Jiang
- Key Laboratory of Biotechnology of Antibiotics of Ministry of Health, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Tiantan Xili, Beijing 100050, China
| | - Li Wang
- Key Laboratory of Biotechnology of Antibiotics of Ministry of Health, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Tiantan Xili, Beijing 100050, China.
| | - Bin Hong
- Key Laboratory of Biotechnology of Antibiotics of Ministry of Health, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Tiantan Xili, Beijing 100050, China.
| |
Collapse
|
10
|
IgE-tailpiece associates with α-1-antitrypsin (A1AT) to protect IgE from proteolysis without compromising its ability to interact with FcεRI. Sci Rep 2016; 6:20509. [PMID: 26842628 PMCID: PMC4740804 DOI: 10.1038/srep20509] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 01/05/2016] [Indexed: 11/08/2022] Open
Abstract
Several splice variants of IgE exist in human plasma, including a variant called IgE-tailpiece (IgE-tp) that differs from classical IgE by the replacement of two carboxy-terminal amino acids with eight novel residues that include an ultimate cysteine. To date, the role of the secreted IgE-tp isoform in human immunity is unknown. We show that levels of IgE-tp are raised in helminth-infected donors, and that both the classical form of IgE (IgE-c) and IgE-tp interact with polymers of the serine protease inhibitor alpha-1-antitrypsin (A1AT). The association of IgE-tp with A1AT polymers in plasma protects the antibody from serine protease-mediated degradation, without affecting the functional interaction of IgE-tp with important receptors, including FcεR1. That polymers of A1AT protect IgE from degradation by helminth proteases may explain why these common and normally non-disease causing polymorphic variants of A1AT have been retained by natural selection. The observation that IgE can be complexed with polymeric forms of A1AT may therefore have important consequences for our understanding of the pathophysiology of pulmonary diseases that arise either as a consequence of A1AT-deficiency or through IgE-mediated type 1 hypersensitivity responses.
Collapse
|
11
|
Shen WJ, Hu J, Hu Z, Kraemer FB, Azhar S. Scavenger receptor class B type I (SR-BI): a versatile receptor with multiple functions and actions. Metabolism 2014; 63:875-86. [PMID: 24854385 PMCID: PMC8078058 DOI: 10.1016/j.metabol.2014.03.011] [Citation(s) in RCA: 101] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Revised: 03/12/2014] [Accepted: 03/18/2014] [Indexed: 11/16/2022]
Abstract
Scavenger receptor class B type I (SR-BI), is a physiologically relevant HDL receptor that mediates selective uptake of lipoprotein (HDL)-derived cholesteryl ester (CE) in vitro and in vivo. Mammalian SR-BI is a 509-amino acid, ~82 kDa glycoprotein that contains N- and C-terminal cytoplasmic domains, two-transmembrane domains, as well as a large extracellular domain containing 5-6 cysteine residues and multiple sites for N-linked glycosylation. The size and structural characteristics of SR-BI, however, vary considerably among lower vertebrates and insects. Recently, significant progress has been made in understanding the molecular mechanisms involved in the posttranscriptional/posttranslational regulation of SR-BI in a tissue specific manner. The purpose of this review is to summarize the current body of knowledge about the events and molecules connected with the posttranscriptional/posttranslational regulation of SR-BI and to update the molecular and functional characteristics of the insect SR-BI orthologs.
Collapse
MESH Headings
- Animals
- Biological Transport
- Gene Expression Regulation
- Glycosylation
- Humans
- Insect Proteins/chemistry
- Insect Proteins/genetics
- Insect Proteins/metabolism
- Lipoproteins, HDL/chemistry
- Lipoproteins, HDL/genetics
- Lipoproteins, HDL/metabolism
- Liver/metabolism
- Organ Specificity
- Protein Conformation
- Protein Processing, Post-Translational
- Receptors, Lipoprotein/chemistry
- Receptors, Lipoprotein/genetics
- Receptors, Lipoprotein/metabolism
- Scavenger Receptors, Class B/chemistry
- Scavenger Receptors, Class B/genetics
- Scavenger Receptors, Class B/metabolism
- Species Specificity
Collapse
Affiliation(s)
- Wen-Jun Shen
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, Palo Alto, California 94304; Division of Endocrinology, Stanford University, Stanford, California 94305
| | - Jie Hu
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, Palo Alto, California 94304
| | - Zhigang Hu
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, Palo Alto, California 94304; Division of Endocrinology, Stanford University, Stanford, California 94305
| | - Fredric B Kraemer
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, Palo Alto, California 94304; Division of Endocrinology, Stanford University, Stanford, California 94305
| | - Salman Azhar
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, Palo Alto, California 94304; Division of Gastroenterology and Hepatology, Stanford University, Stanford, California 94305.
| |
Collapse
|
12
|
Nagy L, Szanto A, Szatmari I, Széles L. Nuclear hormone receptors enable macrophages and dendritic cells to sense their lipid environment and shape their immune response. Physiol Rev 2012; 92:739-89. [PMID: 22535896 DOI: 10.1152/physrev.00004.2011] [Citation(s) in RCA: 169] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
A key issue in the immune system is to generate specific cell types, often with opposing activities. The mechanisms of differentiation and subtype specification of immune cells such as macrophages and dendritic cells are critical to understand the regulatory principles and logic of the immune system. In addition to cytokines and pathogens, it is increasingly appreciated that lipid signaling also has a key role in differentiation and subtype specification. In this review we explore how intracellular lipid signaling via a set of transcription factors regulates cellular differentiation, subtype specification, and immune as well as metabolic homeostasis. We introduce macrophages and dendritic cells and then we focus on a group of transcription factors, nuclear receptors, which regulate gene expression upon receiving lipid signals. The receptors we cover are the ones with a recognized physiological function in these cell types and ones which heterodimerize with the retinoid X receptor. These are as follows: the receptor for a metabolite of vitamin A, retinoic acid: retinoic acid receptor (RAR), the vitamin D receptor (VDR), the fatty acid receptor: peroxisome proliferator-activated receptor γ (PPARγ), the oxysterol receptor liver X receptor (LXR), and their obligate heterodimeric partner, the retinoid X receptor (RXR). We discuss how they can get activated and how ligand is generated and eliminated in these cell types. We also explore how activation of a particular target gene contributes to biological functions and how the regulation of individual target genes adds up to the coordination of gene networks. It appears that RXR heterodimeric nuclear receptors provide these cells with a coordinated and interrelated network of transcriptional regulators for interpreting the lipid milieu and the metabolic changes to bring about gene expression changes leading to subtype and functional specification. We also show that these networks are implicated in various immune diseases and are amenable to therapeutic exploitation.
Collapse
Affiliation(s)
- Laszlo Nagy
- Department of Biochemistry and Molecular Biology, University of Debrecen, Medical and Health Science Center, Egyetem tér 1, Debrecen, Hungary.
| | | | | | | |
Collapse
|
13
|
Research Advances of Cholesterol Efflux in Atherosclerosis*. PROG BIOCHEM BIOPHYS 2012. [DOI: 10.3724/sp.j.1206.2011.00301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
14
|
Fournier N, Attia N, Rousseau-Ralliard D, Vedie B, Destaillats F, Grynberg A, Paul JL. Deleterious impact of elaidic fatty acid on ABCA1-mediated cholesterol efflux from mouse and human macrophages. Biochim Biophys Acta Mol Cell Biol Lipids 2011; 1821:303-12. [PMID: 22074701 DOI: 10.1016/j.bbalip.2011.10.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2011] [Revised: 09/07/2011] [Accepted: 10/05/2011] [Indexed: 10/16/2022]
Abstract
Consumption of trans fatty acids (TFA) increase cardiovascular risk more than do saturated FA, but the mechanisms explaining their atherogenicity are still unclear. We investigated the impact of membrane incorporation of TFA on cholesterol efflux by exposing J774 mouse macrophages or human monocyte-derived macrophages (HMDM) to media enriched or not (standard medium) with industrially produced elaidic (trans-9 18:1) acid, naturally produced vaccenic (trans-11 18:1) acid (34 h, 70 μM) or palmitic acid. In J774 macrophages, elaidic and palmitic acid, but not vaccenic acid, reduced ABCA1-mediated efflux by ~23% without affecting aqueous diffusion, SR-BI or ABCG1-mediated pathways, and this effect was maintained in cholesterol-loaded cells. The impact of elaidic acid on the ABCA1 pathway was weaker in cholesterol-normal HMDM, but elaidic acid induced a strong reduction of ABCA1-mediated efflux in cholesterol-loaded cells (-36%). In J774 cells, the FA supplies had no impact on cellular free cholesterol or cholesteryl ester masses, the abundance of ABCA1 mRNA or the total and plasma membrane ABCA1 protein content. Conversely, TFA or palmitic acid incorporation induced strong modifications of the membrane FA composition with a decrease in the ratio of (cis-monounsaturated FA+polyunsaturated FA):(saturated FA+TFA), with elaidic and vaccenic acids representing each 20% and 13% of the total FA composition, respectively. Moreover, we demonstrated that cellular ATP was required for the effect of elaidic acid, suggesting that it contributes to atherogenesis by impairing ABCA1-mediated cholesterol efflux in macrophages, likely by decreasing the membrane fluidity, which could thereby reduce ATPase activity and the function of the transporter.
Collapse
Affiliation(s)
- Natalie Fournier
- Univ Paris-Sud, EA 4529, UFR de Pharmacie, 92296 Châtenay-Malabry, France.
| | | | | | | | | | | | | |
Collapse
|
15
|
Zhao Y, Pennings M, Vrins CL, Calpe-Berdiel L, Hoekstra M, Kruijt JK, Ottenhoff R, Hildebrand RB, van der Sluis R, Jessup W, Le Goff W, Chapman MJ, Huby T, Groen AK, Van Berkel TJ, Van Eck M. Hypocholesterolemia, foam cell accumulation, but no atherosclerosis in mice lacking ABC-transporter A1 and scavenger receptor BI. Atherosclerosis 2011; 218:314-22. [DOI: 10.1016/j.atherosclerosis.2011.07.096] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2011] [Revised: 06/23/2011] [Accepted: 07/14/2011] [Indexed: 10/17/2022]
|
16
|
Ji A, Meyer JM, Cai L, Akinmusire A, de Beer MC, Webb NR, van der Westhuyzen DR. Scavenger receptor SR-BI in macrophage lipid metabolism. Atherosclerosis 2011; 217:106-12. [PMID: 21481393 DOI: 10.1016/j.atherosclerosis.2011.03.017] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2010] [Revised: 03/07/2011] [Accepted: 03/11/2011] [Indexed: 12/15/2022]
Abstract
OBJECTIVE To investigate the mechanisms by which macrophage scavenger receptor BI (SR-BI) regulates macrophage cholesterol homeostasis and protects against atherosclerosis. METHODS AND RESULTS The expression and function of SR-BI was investigated in cultured mouse bone marrow-derived macrophages (BMM). SR-BI, the other scavenger receptors SRA and CD36 and the ATP-binding cassette transporters ABCA1 and ABCG1 were each distinctly regulated during BMM differentiation. SR-BI levels increased transiently to significant levels during culture. SR-BI expression in BMM was reversibly down-regulated by lipid loading with modified LDL; SR-BI was shown to be present both on the cell surface as well as intracellularly. BMM exhibited selective HDL CE uptake, however, this was not dependent on SR-BI or another potential candidate glycosylphosphatidylinositol anchored high density lipoprotein binding protein 1 (GPIHBP1). SR-BI played a significant role in facilitating bidirectional cholesterol flux in non lipid-loaded cells. SR-BI expression enhanced both cell cholesterol efflux and cholesterol influx from HDL, but did not lead to altered cellular cholesterol mass. SR-BI-dependent efflux occurred to larger HDL particles but not to smaller HDL(3). Following cholesterol loading, ABCA1 and ABCG1 were up-regulated and served as the major contributors to cholesterol efflux, while SR-BI expression was down-regulated. CONCLUSION Our results suggest that SR-BI plays a significant role in macrophage cholesterol flux that may partly account for its effects on atherogenesis.
Collapse
Affiliation(s)
- Ailing Ji
- Department of Internal Medicine, University of Kentucky, Lexington, KY 40536, USA.
| | | | | | | | | | | | | |
Collapse
|
17
|
Zhao Y, Van Berkel TJ, Van Eck M. Relative roles of various efflux pathways in net cholesterol efflux from macrophage foam cells in atherosclerotic lesions. Curr Opin Lipidol 2010; 21:441-53. [PMID: 20683325 DOI: 10.1097/mol.0b013e32833dedaa] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW Cholesterol efflux mechanisms are essential for macrophage cholesterol homeostasis. HDL, an important cholesterol efflux acceptor, comprises a class of heterogeneous particles that induce cholesterol efflux via distinct pathways. This review focuses on the understanding of the different cholesterol efflux pathways and physiological acceptors involved, and their regulation in atherosclerotic lesions. RECENT FINDINGS The synergistic interactions of ATP-binding cassette transporters A1 and G1 as well as ATP-binding cassette transporter A1 and scavenger receptor class B type I are essential for cellular cholesterol efflux and the prevention of macrophage foam cell formation. However, the importance of aqueous diffusion should also not be underestimated. Significant progress has been made in understanding the mechanisms underlying ATP-binding cassette A1-mediated cholesterol efflux and regulation of its expression and trafficking. Conditions locally in the atherosclerotic lesion, for example, lipids, cytokines, oxidative stress, and hypoxia, as well as systemic factors, including inflammation and diabetes, critically influence the expression of cholesterol transporters on macrophage foam cells. Furthermore, HDL modification and remodeling in atherosclerosis, inflammation, and diabetes impairs its function as an acceptor for cellular cholesterol. SUMMARY Recent advances in the understanding of the regulation of cholesterol transporters and their acceptors in atherosclerotic lesions indicate that HDL-based therapies should aim to enhance the activity of cholesterol transporters and improve both the quantity and quality of HDL.
Collapse
Affiliation(s)
- Ying Zhao
- Division of Biopharmaceutics, Leiden/Amsterdam Center for Drug Research, University of Leiden, Leiden, The Netherlands
| | | | | |
Collapse
|
18
|
Abstract
Dysregulation of cholesterol balance contributes significantly to atherosclerotic cardiovascular disease (ASCVD), the leading cause of death in the United States. The intestine has the unique capability to act as a gatekeeper for entry of cholesterol into the body, and inhibition of intestinal cholesterol absorption is now widely regarded as an attractive non-statin therapeutic strategy for ASCVD prevention. In this chapter we discuss the current state of knowledge regarding sterol transport across the intestinal brush border membrane. The purpose of this work is to summarize substantial progress made in the last decade in regards to protein-mediated sterol trafficking, and to discuss this in the context of human disease.
Collapse
Affiliation(s)
| | - Liqing Yu
- Address correspondence to: Liqing Yu, M.D., Ph.D., Department of Pathology Section on Lipid Sciences, Wake Forest University School of Medicine, Medical Center Blvd, Winston-Salem, NC 27157-1040, Tel: 336-716-0920, Fax: 336-716-6279,
| |
Collapse
|
19
|
Gantman A, Fuhrman B, Aviram M, Hayek T. High glucose stimulates macrophage SR-BI expression and induces a switch in its activity from cholesterol efflux to cholesterol influx. Biochem Biophys Res Commun 2010; 391:523-8. [DOI: 10.1016/j.bbrc.2009.11.091] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2009] [Accepted: 11/14/2009] [Indexed: 12/29/2022]
|
20
|
Larrede S, Quinn CM, Jessup W, Frisdal E, Olivier M, Hsieh V, Kim MJ, Van Eck M, Couvert P, Carrie A, Giral P, Chapman MJ, Guerin M, Le Goff W. Stimulation of Cholesterol Efflux by LXR Agonists in Cholesterol-Loaded Human Macrophages Is ABCA1-Dependent but ABCG1-Independent. Arterioscler Thromb Vasc Biol 2009; 29:1930-6. [DOI: 10.1161/atvbaha.109.194548] [Citation(s) in RCA: 155] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Sandra Larrede
- From INSERM, UMR S939 (S.L., E.F., M.O., P.C., A.C., P.G., M.J.C., M.G., W.L.G.), Paris, France; Université Pierre et Marie Curie–Paris6 (S.L., E.F., M.O., P.C., A.C., P.G., M.J.C., M.G., W.L.G.), UMR S939 Paris, France; AP-HP, Groupe hospitalier Pitié-Salpétrière, Service de Biochimie Endocrinienne et Oncologique (P.C., A.C.) and Service d’Endocrinologie-Métabolisme (P.G.), Paris, France; the Centre for Vascular Research (C.M.Q., W.J., V.H., M.-J.K.), School of Medical Sciences, University of
| | - Carmel M. Quinn
- From INSERM, UMR S939 (S.L., E.F., M.O., P.C., A.C., P.G., M.J.C., M.G., W.L.G.), Paris, France; Université Pierre et Marie Curie–Paris6 (S.L., E.F., M.O., P.C., A.C., P.G., M.J.C., M.G., W.L.G.), UMR S939 Paris, France; AP-HP, Groupe hospitalier Pitié-Salpétrière, Service de Biochimie Endocrinienne et Oncologique (P.C., A.C.) and Service d’Endocrinologie-Métabolisme (P.G.), Paris, France; the Centre for Vascular Research (C.M.Q., W.J., V.H., M.-J.K.), School of Medical Sciences, University of
| | - Wendy Jessup
- From INSERM, UMR S939 (S.L., E.F., M.O., P.C., A.C., P.G., M.J.C., M.G., W.L.G.), Paris, France; Université Pierre et Marie Curie–Paris6 (S.L., E.F., M.O., P.C., A.C., P.G., M.J.C., M.G., W.L.G.), UMR S939 Paris, France; AP-HP, Groupe hospitalier Pitié-Salpétrière, Service de Biochimie Endocrinienne et Oncologique (P.C., A.C.) and Service d’Endocrinologie-Métabolisme (P.G.), Paris, France; the Centre for Vascular Research (C.M.Q., W.J., V.H., M.-J.K.), School of Medical Sciences, University of
| | - Eric Frisdal
- From INSERM, UMR S939 (S.L., E.F., M.O., P.C., A.C., P.G., M.J.C., M.G., W.L.G.), Paris, France; Université Pierre et Marie Curie–Paris6 (S.L., E.F., M.O., P.C., A.C., P.G., M.J.C., M.G., W.L.G.), UMR S939 Paris, France; AP-HP, Groupe hospitalier Pitié-Salpétrière, Service de Biochimie Endocrinienne et Oncologique (P.C., A.C.) and Service d’Endocrinologie-Métabolisme (P.G.), Paris, France; the Centre for Vascular Research (C.M.Q., W.J., V.H., M.-J.K.), School of Medical Sciences, University of
| | - Maryline Olivier
- From INSERM, UMR S939 (S.L., E.F., M.O., P.C., A.C., P.G., M.J.C., M.G., W.L.G.), Paris, France; Université Pierre et Marie Curie–Paris6 (S.L., E.F., M.O., P.C., A.C., P.G., M.J.C., M.G., W.L.G.), UMR S939 Paris, France; AP-HP, Groupe hospitalier Pitié-Salpétrière, Service de Biochimie Endocrinienne et Oncologique (P.C., A.C.) and Service d’Endocrinologie-Métabolisme (P.G.), Paris, France; the Centre for Vascular Research (C.M.Q., W.J., V.H., M.-J.K.), School of Medical Sciences, University of
| | - Victar Hsieh
- From INSERM, UMR S939 (S.L., E.F., M.O., P.C., A.C., P.G., M.J.C., M.G., W.L.G.), Paris, France; Université Pierre et Marie Curie–Paris6 (S.L., E.F., M.O., P.C., A.C., P.G., M.J.C., M.G., W.L.G.), UMR S939 Paris, France; AP-HP, Groupe hospitalier Pitié-Salpétrière, Service de Biochimie Endocrinienne et Oncologique (P.C., A.C.) and Service d’Endocrinologie-Métabolisme (P.G.), Paris, France; the Centre for Vascular Research (C.M.Q., W.J., V.H., M.-J.K.), School of Medical Sciences, University of
| | - Mi-Jurng Kim
- From INSERM, UMR S939 (S.L., E.F., M.O., P.C., A.C., P.G., M.J.C., M.G., W.L.G.), Paris, France; Université Pierre et Marie Curie–Paris6 (S.L., E.F., M.O., P.C., A.C., P.G., M.J.C., M.G., W.L.G.), UMR S939 Paris, France; AP-HP, Groupe hospitalier Pitié-Salpétrière, Service de Biochimie Endocrinienne et Oncologique (P.C., A.C.) and Service d’Endocrinologie-Métabolisme (P.G.), Paris, France; the Centre for Vascular Research (C.M.Q., W.J., V.H., M.-J.K.), School of Medical Sciences, University of
| | - Miranda Van Eck
- From INSERM, UMR S939 (S.L., E.F., M.O., P.C., A.C., P.G., M.J.C., M.G., W.L.G.), Paris, France; Université Pierre et Marie Curie–Paris6 (S.L., E.F., M.O., P.C., A.C., P.G., M.J.C., M.G., W.L.G.), UMR S939 Paris, France; AP-HP, Groupe hospitalier Pitié-Salpétrière, Service de Biochimie Endocrinienne et Oncologique (P.C., A.C.) and Service d’Endocrinologie-Métabolisme (P.G.), Paris, France; the Centre for Vascular Research (C.M.Q., W.J., V.H., M.-J.K.), School of Medical Sciences, University of
| | - Philippe Couvert
- From INSERM, UMR S939 (S.L., E.F., M.O., P.C., A.C., P.G., M.J.C., M.G., W.L.G.), Paris, France; Université Pierre et Marie Curie–Paris6 (S.L., E.F., M.O., P.C., A.C., P.G., M.J.C., M.G., W.L.G.), UMR S939 Paris, France; AP-HP, Groupe hospitalier Pitié-Salpétrière, Service de Biochimie Endocrinienne et Oncologique (P.C., A.C.) and Service d’Endocrinologie-Métabolisme (P.G.), Paris, France; the Centre for Vascular Research (C.M.Q., W.J., V.H., M.-J.K.), School of Medical Sciences, University of
| | - Alain Carrie
- From INSERM, UMR S939 (S.L., E.F., M.O., P.C., A.C., P.G., M.J.C., M.G., W.L.G.), Paris, France; Université Pierre et Marie Curie–Paris6 (S.L., E.F., M.O., P.C., A.C., P.G., M.J.C., M.G., W.L.G.), UMR S939 Paris, France; AP-HP, Groupe hospitalier Pitié-Salpétrière, Service de Biochimie Endocrinienne et Oncologique (P.C., A.C.) and Service d’Endocrinologie-Métabolisme (P.G.), Paris, France; the Centre for Vascular Research (C.M.Q., W.J., V.H., M.-J.K.), School of Medical Sciences, University of
| | - Philippe Giral
- From INSERM, UMR S939 (S.L., E.F., M.O., P.C., A.C., P.G., M.J.C., M.G., W.L.G.), Paris, France; Université Pierre et Marie Curie–Paris6 (S.L., E.F., M.O., P.C., A.C., P.G., M.J.C., M.G., W.L.G.), UMR S939 Paris, France; AP-HP, Groupe hospitalier Pitié-Salpétrière, Service de Biochimie Endocrinienne et Oncologique (P.C., A.C.) and Service d’Endocrinologie-Métabolisme (P.G.), Paris, France; the Centre for Vascular Research (C.M.Q., W.J., V.H., M.-J.K.), School of Medical Sciences, University of
| | - M. John Chapman
- From INSERM, UMR S939 (S.L., E.F., M.O., P.C., A.C., P.G., M.J.C., M.G., W.L.G.), Paris, France; Université Pierre et Marie Curie–Paris6 (S.L., E.F., M.O., P.C., A.C., P.G., M.J.C., M.G., W.L.G.), UMR S939 Paris, France; AP-HP, Groupe hospitalier Pitié-Salpétrière, Service de Biochimie Endocrinienne et Oncologique (P.C., A.C.) and Service d’Endocrinologie-Métabolisme (P.G.), Paris, France; the Centre for Vascular Research (C.M.Q., W.J., V.H., M.-J.K.), School of Medical Sciences, University of
| | - Maryse Guerin
- From INSERM, UMR S939 (S.L., E.F., M.O., P.C., A.C., P.G., M.J.C., M.G., W.L.G.), Paris, France; Université Pierre et Marie Curie–Paris6 (S.L., E.F., M.O., P.C., A.C., P.G., M.J.C., M.G., W.L.G.), UMR S939 Paris, France; AP-HP, Groupe hospitalier Pitié-Salpétrière, Service de Biochimie Endocrinienne et Oncologique (P.C., A.C.) and Service d’Endocrinologie-Métabolisme (P.G.), Paris, France; the Centre for Vascular Research (C.M.Q., W.J., V.H., M.-J.K.), School of Medical Sciences, University of
| | - Wilfried Le Goff
- From INSERM, UMR S939 (S.L., E.F., M.O., P.C., A.C., P.G., M.J.C., M.G., W.L.G.), Paris, France; Université Pierre et Marie Curie–Paris6 (S.L., E.F., M.O., P.C., A.C., P.G., M.J.C., M.G., W.L.G.), UMR S939 Paris, France; AP-HP, Groupe hospitalier Pitié-Salpétrière, Service de Biochimie Endocrinienne et Oncologique (P.C., A.C.) and Service d’Endocrinologie-Métabolisme (P.G.), Paris, France; the Centre for Vascular Research (C.M.Q., W.J., V.H., M.-J.K.), School of Medical Sciences, University of
| |
Collapse
|
21
|
Rocha-Perugini V, Lavie M, Delgrange D, Canton J, Pillez A, Potel J, Lecoeur C, Rubinstein E, Dubuisson J, Wychowski C, Cocquerel L. The association of CD81 with tetraspanin-enriched microdomains is not essential for Hepatitis C virus entry. BMC Microbiol 2009; 9:111. [PMID: 19476617 PMCID: PMC2694809 DOI: 10.1186/1471-2180-9-111] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2008] [Accepted: 05/28/2009] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Three percent of the world's population is chronically infected with hepatitis C virus (HCV) and thus at risk of developing liver cancer. Although precise mechanisms regulating HCV entry into hepatic cells are still unknown, several cell surface proteins have been identified as entry factors for this virus. Among these molecules, the tetraspanin CD81 is essential for HCV entry. Interestingly, CD81 is also required for Plasmodium infection. A major characteristic of tetraspanins is their ability to interact with each other and other transmembrane proteins to build tetraspanin-enriched microdomains (TEM). RESULTS In our study, we describe a human hepatoma Huh-7 cell clone (Huh-7w7) which has lost CD81 expression and can be infected by HCV when human CD81 (hCD81) or mouse CD81 (mCD81) is ectopically expressed. We took advantage of these permissive cells expressing mCD81 and the previously described MT81/MT81w mAbs to analyze the role of TEM-associated CD81 in HCV infection. Importantly, MT81w antibody, which only recognizes TEM-associated mCD81, did not strongly affect HCV infection. Furthermore, cholesterol depletion, which inhibits HCV infection and reduces total cell surface expression of CD81, did not affect TEM-associated CD81 levels. In addition, sphingomyelinase treatment, which also reduces HCV infection and cell surface expression of total CD81, raised TEM-associated CD81 levels. CONCLUSION In contrast to Plasmodium infection, our data show that association of CD81 with TEM is not essential for the early steps of HCV life cycle, indicating that these two pathogens, while using the same molecules, invade their host by different mechanisms.
Collapse
Affiliation(s)
- Vera Rocha-Perugini
- Institut de Biologie de Lille, CNRS-UMR8161, Université Lille Nord de France, Institut Pasteur de Lille, Lille, France.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Niemeier A, Kovacs WJ, Strobl W, Stangl H. Atherogenic diet leads to posttranslational down-regulation of murine hepatocyte SR-BI expression. Atherosclerosis 2009; 202:169-75. [DOI: 10.1016/j.atherosclerosis.2008.04.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2007] [Revised: 04/03/2008] [Accepted: 04/17/2008] [Indexed: 02/06/2023]
|
23
|
Abstract
PURPOSE OF REVIEW The lipid efflux pathway is important for both HDL formation and the reverse cholesterol transport pathway. This review is focused on recent findings on the mechanism of lipid efflux and its regulation, particularly in macrophages. RECENT FINDINGS Significant progress has been made on understanding the sequence of events that accompany the interaction of apolipoproteins A-I with cell surface ATP-binding cassette transporter A1 and its subsequent lipidation. Continued research on the regulation of ATP-binding cassette transporter A1 and ATP-binding cassette transporter G1 expression and traffic has also generated new paradigms for the control of lipid efflux from macrophages and its contribution to reverse cholesterol transport. In addition, the mobilization of cholesteryl esters from lipid droplets represents a new step in the control of cholesterol efflux. SUMMARY The synergy between lipid transporters is a work in progress, but its importance in reverse cholesterol transport is clear. The regulation of efflux implies both the regulation of relevant transporters and the cellular trafficking of cholesterol.
Collapse
Affiliation(s)
- Yves L Marcel
- Lipoprotein and Atherosclerosis Research Group, University of Ottawa Heart Institute, Ottawa, Ontario, Canada.
| | | | | |
Collapse
|
24
|
Functional LCAT is not required for macrophage cholesterol efflux to human serum. Atherosclerosis 2008; 204:141-6. [PMID: 18922527 DOI: 10.1016/j.atherosclerosis.2008.08.038] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2008] [Revised: 07/31/2008] [Accepted: 08/14/2008] [Indexed: 11/20/2022]
Abstract
OBJECTIVES To evaluate the capacity of serum from carriers of LCAT gene mutations to promote cell cholesterol efflux through the ABCA1, ABCG1, and SR-BI pathways. METHODS Serum was obtained from 41 carriers of mutant LCAT alleles (14 carriers of two mutant LCAT alleles and 27 heterozygotes) and 10 non-carrier relatives (controls). The capacity of serum to promote cholesterol efflux was tested in pathway-specific cell models. RESULTS LCAT deficient sera were significantly more efficient than control sera in promoting cell cholesterol efflux via ABCA1 (3.1+/-0.3% for carriers of two mutant LCAT alleles and 2.6+/-0.2% for heterozygotes vs. 1.5+/-0.4% for controls), and less efficient in promoting ABCG1- and SR-BI-mediated cholesterol efflux. The enhanced capacity of LCAT deficient serum for ABCA1 efflux is explained by the increased content of prebeta-HDL, as indicated by the significant positive correlation between ABCA1 efflux and serum prebeta-HDL content (R=0.468, P<0.001). Moreover, chymase treatment of LCAT deficient serum selectively degraded prebeta-HDL and completely abolished ABCA1 efflux. Despite the remarkable reductions in serum HDL levels, LCAT deficient sera were as effective as control sera in removing mass cholesterol from cholesterol-loaded macrophages. CONCLUSIONS Serum from carriers of LCAT gene mutations has the same capacity of control serum to decrease the cholesterol content of cholesterol-loaded macrophages due to a greater cholesterol efflux capacity via ABCA1.
Collapse
|
25
|
Enhanced removal of cholesterol from macrophage foam cells to serum from type IV hypertriglyceridemic subjects. Atherosclerosis 2008; 198:49-56. [DOI: 10.1016/j.atherosclerosis.2007.09.023] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2007] [Revised: 09/07/2007] [Accepted: 09/18/2007] [Indexed: 11/22/2022]
|
26
|
Yvan-Charvet L, Pagler TA, Wang N, Senokuchi T, Brundert M, Li H, Rinninger F, Tall AR. SR-BI inhibits ABCG1-stimulated net cholesterol efflux from cells to plasma HDL. J Lipid Res 2008; 49:107-14. [DOI: 10.1194/jlr.m700200-jlr200] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
27
|
Wang X, Collins HL, Ranalletta M, Fuki IV, Billheimer JT, Rothblat GH, Tall AR, Rader DJ. Macrophage ABCA1 and ABCG1, but not SR-BI, promote macrophage reverse cholesterol transport in vivo. J Clin Invest 2007; 117:2216-24. [PMID: 17657311 PMCID: PMC1924499 DOI: 10.1172/jci32057] [Citation(s) in RCA: 451] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2007] [Accepted: 05/10/2007] [Indexed: 01/21/2023] Open
Abstract
Macrophage ATP-binding cassette transporter A1 (ABCA1), scavenger receptor class B type I (SR-BI), and ABCG1 have been shown to promote cholesterol efflux to extracellular acceptors in vitro and influence atherosclerosis in mice, but their roles in mediating reverse cholesterol transport (RCT) from macrophages in vivo are unknown. Using an assay of macrophage RCT in mice, we found that primary macrophages lacking ABCA1 had a significant reduction in macrophage RCT in vivo, demonstrating the importance of ABCA1 in promoting macrophage RCT, however substantial residual RCT exists in the absence of macrophage ABCA1. Using primary macrophages deficient in SR-BI expression, we found that macrophage SR-BI, which was shown to promote cholesterol efflux in vitro, does not contribute to macrophage RCT in vivo. To investigate whether macrophage ABCG1 is involved in macrophage RCT in vivo, we used ABCG1-overexpressing, -knockdown, and -knockout macrophages. We show that increased macrophage ABCG1 expression significantly promoted while knockdown or knockout of macrophage ABCG1 expression significantly reduced macrophage RCT in vivo. Finally, we show that there was a greater decrease in macrophage RCT from cells where both ABCA1 and ABCG1 expression were knocked down than from ABCG1-knockdown cells. These results demonstrate that ABCA1 and ABCG1, but not SR-BI, promote macrophage RCT in vivo and are additive in their effects.
Collapse
Affiliation(s)
- Xun Wang
- Institute for Translational Medicine and Therapeutics and Cardiovascular Institute, School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.
Division of Molecular Medicine, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, New York, USA
| | - Heidi L. Collins
- Institute for Translational Medicine and Therapeutics and Cardiovascular Institute, School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.
Division of Molecular Medicine, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, New York, USA
| | - Mollie Ranalletta
- Institute for Translational Medicine and Therapeutics and Cardiovascular Institute, School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.
Division of Molecular Medicine, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, New York, USA
| | - Ilia V. Fuki
- Institute for Translational Medicine and Therapeutics and Cardiovascular Institute, School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.
Division of Molecular Medicine, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, New York, USA
| | - Jeffrey T. Billheimer
- Institute for Translational Medicine and Therapeutics and Cardiovascular Institute, School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.
Division of Molecular Medicine, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, New York, USA
| | - George H. Rothblat
- Institute for Translational Medicine and Therapeutics and Cardiovascular Institute, School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.
Division of Molecular Medicine, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, New York, USA
| | - Alan R. Tall
- Institute for Translational Medicine and Therapeutics and Cardiovascular Institute, School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.
Division of Molecular Medicine, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, New York, USA
| | - Daniel J. Rader
- Institute for Translational Medicine and Therapeutics and Cardiovascular Institute, School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.
Division of Molecular Medicine, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, New York, USA
| |
Collapse
|
28
|
Temel RE, Tang W, Ma Y, Rudel LL, Willingham MC, Ioannou YA, Davies JP, Nilsson LM, Yu L. Hepatic Niemann-Pick C1-like 1 regulates biliary cholesterol concentration and is a target of ezetimibe. J Clin Invest 2007; 117:1968-78. [PMID: 17571164 PMCID: PMC1888567 DOI: 10.1172/jci30060] [Citation(s) in RCA: 284] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2006] [Accepted: 04/10/2007] [Indexed: 12/13/2022] Open
Abstract
Niemann-Pick C1-like 1 (NPC1L1) is required for cholesterol absorption. Intestinal NPC1L1 appears to be a target of ezetimibe, a cholesterol absorption inhibitor that effectively lowers plasma LDL-cholesterol in humans. However, human liver also expresses NPC1L1. Hepatic function of NPC1L1 was previously unknown, but we recently discovered that NPC1L1 localizes to the canalicular membrane of primate hepatocytes and that NPC1L1 facilitates cholesterol uptake in hepatoma cells. Based upon these findings, we hypothesized that hepatic NPC1L1 allows the retention of biliary cholesterol by hepatocytes and that ezetimibe disrupts hepatic function of NPC1L1. To test this hypothesis, transgenic mice expressing human NPC1L1 in hepatocytes (L1-Tg mice) were created. Hepatic overexpression of NPC1L1 resulted in a 10- to 20-fold decrease in biliary cholesterol concentration, but not phospholipid and bile acid concentrations. This decrease was associated with a 30%-60% increase in plasma cholesterol, mainly because of the accumulation of apoE-rich HDL. Biliary and plasma cholesterol concentrations in these animals were virtually returned to normal with ezetimibe treatment. These findings suggest that in humans, ezetimibe may reduce plasma cholesterol by inhibiting NPC1L1 function in both intestine and liver, and hepatic NPC1L1 may have evolved to protect the body from excessive biliary loss of cholesterol.
Collapse
Affiliation(s)
- Ryan E. Temel
- Department of Pathology, Section on Lipid Sciences, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA.
Department of Human Genetics, Mount Sinai School of Medicine, New York, New York, USA.
Division of Gastroenterology and Hepatology, Department of Medicine, Karolinska Institute at Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Weiqing Tang
- Department of Pathology, Section on Lipid Sciences, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA.
Department of Human Genetics, Mount Sinai School of Medicine, New York, New York, USA.
Division of Gastroenterology and Hepatology, Department of Medicine, Karolinska Institute at Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Yinyan Ma
- Department of Pathology, Section on Lipid Sciences, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA.
Department of Human Genetics, Mount Sinai School of Medicine, New York, New York, USA.
Division of Gastroenterology and Hepatology, Department of Medicine, Karolinska Institute at Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Lawrence L. Rudel
- Department of Pathology, Section on Lipid Sciences, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA.
Department of Human Genetics, Mount Sinai School of Medicine, New York, New York, USA.
Division of Gastroenterology and Hepatology, Department of Medicine, Karolinska Institute at Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Mark C. Willingham
- Department of Pathology, Section on Lipid Sciences, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA.
Department of Human Genetics, Mount Sinai School of Medicine, New York, New York, USA.
Division of Gastroenterology and Hepatology, Department of Medicine, Karolinska Institute at Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Yiannis A. Ioannou
- Department of Pathology, Section on Lipid Sciences, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA.
Department of Human Genetics, Mount Sinai School of Medicine, New York, New York, USA.
Division of Gastroenterology and Hepatology, Department of Medicine, Karolinska Institute at Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Joanna P. Davies
- Department of Pathology, Section on Lipid Sciences, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA.
Department of Human Genetics, Mount Sinai School of Medicine, New York, New York, USA.
Division of Gastroenterology and Hepatology, Department of Medicine, Karolinska Institute at Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Lisa-Mari Nilsson
- Department of Pathology, Section on Lipid Sciences, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA.
Department of Human Genetics, Mount Sinai School of Medicine, New York, New York, USA.
Division of Gastroenterology and Hepatology, Department of Medicine, Karolinska Institute at Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Liqing Yu
- Department of Pathology, Section on Lipid Sciences, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA.
Department of Human Genetics, Mount Sinai School of Medicine, New York, New York, USA.
Division of Gastroenterology and Hepatology, Department of Medicine, Karolinska Institute at Karolinska University Hospital Huddinge, Stockholm, Sweden
| |
Collapse
|
29
|
Umetani M, Domoto H, Gormley AK, Yuhanna IS, Cummins CL, Javitt NB, Korach KS, Shaul PW, Mangelsdorf DJ. 27-Hydroxycholesterol is an endogenous SERM that inhibits the cardiovascular effects of estrogen. Nat Med 2007; 13:1185-92. [PMID: 17873880 DOI: 10.1038/nm1641] [Citation(s) in RCA: 305] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2007] [Accepted: 08/08/2007] [Indexed: 01/18/2023]
Abstract
The cardioprotective effects of estrogen are mediated by receptors expressed in vascular cells. Here we show that 27-hydroxycholesterol (27HC), an abundant cholesterol metabolite that is elevated with hypercholesterolemia and found in atherosclerotic lesions, is a competitive antagonist of estrogen receptor action in the vasculature. 27HC inhibited both the transcription-mediated and the non-transcription-mediated estrogen-dependent production of nitric oxide by vascular cells, resulting in reduced estrogen-induced vasorelaxation of rat aorta. Furthermore, increasing 27HC levels in mice by diet-induced hypercholesterolemia, pharmacologic administration or genetic manipulation (by knocking out the gene encoding the catabolic enzyme CYP7B1) decreased estrogen-dependent expression of vascular nitric oxide synthase and repressed carotid artery reendothelialization. As well as antiestrogenic effects, there were proestrogenic actions of 27HC that were cell-type specific, indicating that 27HC functions as an endogenous selective estrogen receptor modulator (SERM). Taken together, these studies point to 27HC as a contributing factor in the loss of estrogen protection from vascular disease.
Collapse
MESH Headings
- Animals
- Aorta, Thoracic/drug effects
- Binding, Competitive/drug effects
- Cardiotonic Agents/antagonists & inhibitors
- Cardiotonic Agents/metabolism
- Cardiotonic Agents/pharmacology
- Cell Culture Techniques
- Cell Line
- Cells, Cultured
- Cholesterol, Dietary/administration & dosage
- DNA, Complementary
- Dose-Response Relationship, Drug
- Drug Administration Schedule
- Estrogens/metabolism
- Estrogens/pharmacology
- Female
- Glutathione Transferase/metabolism
- Humans
- Hydroxycholesterols/administration & dosage
- Hydroxycholesterols/blood
- Hydroxycholesterols/pharmacology
- Inhibitory Concentration 50
- Injections, Subcutaneous
- Kidney/cytology
- Kinetics
- Male
- Mice
- Mice, Knockout
- Nitric Oxide/antagonists & inhibitors
- Nitric Oxide Synthase Type II/antagonists & inhibitors
- Nitric Oxide Synthase Type III
- RNA, Messenger/metabolism
- Receptors, Estrogen/antagonists & inhibitors
- Receptors, Estrogen/genetics
- Recombinant Fusion Proteins/antagonists & inhibitors
- Selective Estrogen Receptor Modulators/administration & dosage
- Selective Estrogen Receptor Modulators/blood
- Selective Estrogen Receptor Modulators/pharmacology
- Vasodilation/drug effects
Collapse
Affiliation(s)
- Michihisa Umetani
- Department of Pharmacology and Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, 6001 Forest Park Road, Dallas, Texas 75390-9050, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Weibel GL, Alexander ET, Joshi MR, Rader DJ, Lund-Katz S, Phillips MC, Rothblat GH. Wild-type ApoA-I and the Milano variant have similar abilities to stimulate cellular lipid mobilization and efflux. Arterioscler Thromb Vasc Biol 2007; 27:2022-9. [PMID: 17615385 DOI: 10.1161/atvbaha.107.148403] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE The present study is a comparative investigation of cellular lipid mobilization and efflux to lipid-free human apoA-I and apoA-I(Milano), reconstituted high-density lipoprotein (rHDL) particles containing these proteins and serum isolated from mice expressing human apoA-I or apoA-I(Milano). METHODS AND RESULTS Cholesterol and phospholipid efflux to these acceptors was measured in cell systems designed to assess the contributions of ATP-binding cassette A1 (ABCA1), scavenger receptor type BI (SRBI), and cellular lipid content to cholesterol and phospholipid efflux. Acceptors containing the Milano variant of apoA-I showed no functional increase in lipid efflux in all assays when compared with wild-type apoA-I. In fact, in some systems, acceptors containing the Milano variant of apoA-I promoted significantly less efflux than the acceptors containing wild-type apoA-I (apoA-I(wt)). Additionally, intracellular cholesteryl ester hydrolysis in macrophage foam cells was not different in the presence of either apoA-I(Milano) or apoA-I(wt). CONCLUSION Collectively these studies suggest that if the Milano variant of apoA-I offers greater atheroprotection than wild-type apoA-I, it is not attributable to greater cellular lipid mobilization.
Collapse
Affiliation(s)
- Ginny L Weibel
- Division of Gasteroenterology and Nutrition, The Children's Hospital of Philadelphia, Philadelphia, PA 19104-4399, USA.
| | | | | | | | | | | | | |
Collapse
|
31
|
Rideout TC, Yuan Z, Bakovic M, Liu Q, Li RK, Mine Y, Fan MZ. Guar gum consumption increases hepatic nuclear SREBP2 and LDL receptor expression in pigs fed an atherogenic diet. J Nutr 2007; 137:568-72. [PMID: 17311941 DOI: 10.1093/jn/137.3.568] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
To gain insight into the regulation of hepatic sterol-responsive genes that are thought to mediate the hypocholesterolemic effects of guar gum (GG) consumption, the mRNA and protein expression of sterol regulatory element binding protein 2 (SREBP2), LDL receptor (LDLr), and scavenger receptor class B, type 1 (SR-B1) were examined in pigs consuming an atherogenic control diet or the control diet supplemented with 10% GG. Compared with the control group, GG consumption reduced (P < 0.05) plasma total cholesterol and LDL cholesterol concentrations by 27 and 37%, respectively. Furthermore, hepatic free cholesterol concentration was lower (P < 0.05) in the GG-fed pigs in comparison with the control group. GG consumption increased hepatic LDLr mRNA (1.5-fold of the control, P = 0.09) and protein (2-fold of the control, P < 0.05) expression in comparison with the control group. However, GG consumption reduced hepatic SR-B1 mRNA to 36% of the control (P < 0.05) expression but did not affect (P = 0.19) SR-B1 protein abundance in comparison with the control group. Although SREBP2 mRNA expression was similar (P = 0.89) in the 2 groups, GG consumption increased (P < 0.05) the expression of the cytoplasmic precursor (3-fold of the control) and nuclear active forms (1.5-fold of the control) of SREBP2. We conclude that the hypocholesterolemic effects of GG consumption are related to a reduction in hepatic free cholesterol concentration and associated increases in nuclear active SREBP2 expression and hepatic LDLr abundance.
Collapse
Affiliation(s)
- Todd C Rideout
- Centre for Nutrition Modelling, Department of Animal and Poultry Science, University of Guelph, Guelph, Ontario, Canada N1G 2W1.
| | | | | | | | | | | | | |
Collapse
|
32
|
Yvan-Charvet L, Matsuura F, Wang N, Bamberger MJ, Nguyen T, Rinninger F, Jiang XC, Shear CL, Tall AR. Inhibition of cholesteryl ester transfer protein by torcetrapib modestly increases macrophage cholesterol efflux to HDL. Arterioscler Thromb Vasc Biol 2007; 27:1132-8. [PMID: 17322101 DOI: 10.1161/atvbaha.106.138347] [Citation(s) in RCA: 174] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE This study examines the effects of pharmacological inhibition of cholesteryl ester transfer protein (CETP) on the ability of high-density lipoprotein particles (HDL) to promote net cholesterol efflux from human THP-1 macrophage foam cells. METHODS AND RESULTS Two groups of 8 healthy, moderately hyperlipidemic subjects received the CETP inhibitor torcetrapib at 60 or 120 mg daily for 8 weeks. Torcetrapib increased HDL cholesterol levels in both groups by 50% and 60%, respectively. Compared with baseline, torcetrapib 60 mg daily increased HDL-mediated net cholesterol efflux from foam cells primarily by increasing HDL concentrations, whereas 120 mg daily torcetrapib increased cholesterol efflux both by increasing HDL concentration and by causing increased efflux at matched HDL concentrations. There was an increased content of lecithin:cholesterol acyltransferase (LCAT) and apolipoprotein E (apoE) in HDL-2 only at the 120 mg dose. ABCG1 activity was responsible for 40% to 50% of net cholesterol efflux to both control and T-HDL. CONCLUSIONS These data indicate that inhibition of CETP by torcetrapib causes a modest increase in the ability of HDL to promote net cholesterol efflux at the 60 mg dose, and a more dramatic increase at the 120 mg dose in association with enhanced particle functionality.
Collapse
Affiliation(s)
- Laurent Yvan-Charvet
- Division of Molecular Medicine, Department of Medicine, Columbia University, 630 West 168 St, New York, NY 10032, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Favari E, Gomaraschi M, Zanotti I, Bernini F, Lee-Rueckert M, Kovanen PT, Sirtori CR, Franceschini G, Calabresi L. A Unique Protease-sensitive High Density Lipoprotein Particle Containing the Apolipoprotein A-IMilano Dimer Effectively Promotes ATP-binding Cassette A1-mediated Cell Cholesterol Efflux. J Biol Chem 2007; 282:5125-32. [PMID: 17164237 DOI: 10.1074/jbc.m609336200] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Carriers of the apolipoprotein A-I(Milano) (A-I(M)) variant present with severe reductions of plasma HDL levels, not associated with premature coronary heart disease (CHD). Sera from 14 A-I(M) carriers and matched controls were compared for their ability to promote ABCA1-driven cholesterol efflux from J774 macrophages and human fibroblasts. When both cell types are stimulated to express ABCA1, the efflux of cholesterol through this pathway is greater with A-I(M) than control sera (3.4 +/- 1.0% versus 2.3 +/- 1.0% in macrophages; 5.2 +/- 2.4% versus 1.9 +/- 0.1% in fibroblasts). A-I(M) and control sera are instead equally effective in removing cholesterol from unstimulated cells and from fibroblasts not expressing ABCA1. The A-I(M) sera contain normal amounts of apoA-I-containing prebeta-HDL and varying concentrations of a unique small HDL particle containing a single molecule of the A-I(M) dimer; chymase treatment of serum degrades both particles and abolishes ABCA1-mediated cholesterol efflux. The serum content of chymase-sensitive HDL correlates strongly and significantly with ABCA1-mediated cholesterol efflux (r = 0.542, p = 0.004). The enhanced capacity of A-I(M) serum for ABCA1 cholesterol efflux is thus explained by the combined occurrence in serum of normal amounts of apoA-I-containing prebeta-HDL, together with a unique protease-sensitive, small HDL particle containing the A-I(M) dimer, both effective in removing cell cholesterol via ABCA1.
Collapse
Affiliation(s)
- Elda Favari
- Department of Pharmacological and Biological Sciences, and Applied Chemistries, University of Parma, Viale delle Scienze 27A, 43100 Parma, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Brundert M, Heeren J, Bahar-Bayansar M, Ewert A, Moore KJ, Rinninger F. Selective uptake of HDL cholesteryl esters and cholesterol efflux from mouse peritoneal macrophages independent of SR-BI. J Lipid Res 2006; 47:2408-21. [PMID: 16926440 DOI: 10.1194/jlr.m600136-jlr200] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Scavenger receptor class B type I (SR-BI) mediates the selective uptake of HDL cholesteryl esters (CEs) and facilitates the efflux of unesterified cholesterol. SR-BI expression in macrophages presumably plays a role in atherosclerosis. The role of SR-BI for selective CE uptake and cholesterol efflux in macrophages was explored. Macrophages and HDL originated from wild-type (WT) or SR-BI knockout (KO; homozygous) mice. For uptake, macrophages were incubated in medium containing 125I-/3H-labeled HDL. For lipid removal, [3H]cholesterol efflux was analyzed using HDL as acceptor. Selective uptake of HDL CE ([3H]cholesteryl oleyl ether - 125I-tyramine cellobiose) was similar in WT and SR-BI KO macrophages. Radiolabeled SR-BI KO-HDL yielded a lower rate of selective uptake compared with WT-HDL in WT and SR-BI KO macrophages. Cholesterol efflux was similar in WT and SR-BI KO cells using HDL as acceptor. SR-BI KO-HDL more efficiently promoted cholesterol removal compared with WT-HDL from both types of macrophages. Macrophages selectively take up HDL CE independently of SR-BI. Additionally, in macrophages, there is substantial cholesterol efflux that is not mediated by SR-BI. Therefore, SR-BI-independent mechanisms mediate selective CE uptake and cholesterol removal. SR-BI KO-HDL is an inferior donor for selective CE uptake compared with WT-HDL, whereas SR-BI KO-HDL more efficiently promotes cholesterol efflux.
Collapse
Affiliation(s)
- May Brundert
- University Hospital Hamburg Eppendorf, Hamburg, Germany
| | | | | | | | | | | |
Collapse
|
35
|
Kapadia SB, Barth H, Baumert T, McKeating JA, Chisari FV. Initiation of hepatitis C virus infection is dependent on cholesterol and cooperativity between CD81 and scavenger receptor B type I. J Virol 2006; 81:374-83. [PMID: 17050612 PMCID: PMC1797271 DOI: 10.1128/jvi.01134-06] [Citation(s) in RCA: 202] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
In the past several years, a number of cellular proteins have been identified as candidate entry receptors for hepatitis C virus (HCV) by using surrogate models of HCV infection. Among these, the tetraspanin CD81 and scavenger receptor B type I (SR-BI), both of which localize to specialized plasma membrane domains enriched in cholesterol, have been suggested to be key players in HCV entry. In the current study, we used a recently developed in vitro HCV infection system to demonstrate that both CD81 and SR-BI are required for authentic HCV infection in vitro, that they function cooperatively to initiate HCV infection, and that CD81-mediated HCV entry is, in part, dependent on membrane cholesterol.
Collapse
Affiliation(s)
- Sharookh B Kapadia
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | | | | | | | |
Collapse
|
36
|
Jessup W, Gelissen IC, Gaus K, Kritharides L. Roles of ATP binding cassette transporters A1 and G1, scavenger receptor BI and membrane lipid domains in cholesterol export from macrophages. Curr Opin Lipidol 2006; 17:247-57. [PMID: 16680029 DOI: 10.1097/01.mol.0000226116.35555.eb] [Citation(s) in RCA: 200] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW The initial steps of reverse cholesterol transport involve export of cholesterol from peripheral cells to plasma lipoproteins for subsequent delivery to the liver. The review discusses recent developments in our understanding of how these steps occur, with particular emphasis on the macrophage, the major site of cellular cholesterol accumulation in atherosclerosis. RECENT FINDINGS ATP binding cassette transporter (ABC) A1 exports cholesterol and phospholipid to lipid-free apolipoproteins, while ATP binding cassette transporter G1 and scavenger receptor BI export cholesterol to phospholipid-containing acceptors. ABCA1-dependent cholesterol export involves an initial interaction of apolipoprotein AI with lipid raft membrane domains, although ABCA1 and most exported cholesterol are not raft associated. ABCG1 exports cholesterol to HDL and other phospholipid-containing acceptors. These include particles generated during lipidation of apoAI by ABCA1, suggesting that the two transporters cooperate in cholesterol export. Scavenger receptor BI is atheroprotective, mediating clearance of HDL cholesterol by the liver. The relative contributions of scavenger receptor BI and ABCG to cholesterol export to HDL from macrophages is unclear and may depend on cellular cholesterol status and the cholesterol gradient between cell and acceptor. SUMMARY The presence of distinct pathways for cholesterol efflux to lipid-free apolipoprotein AI and phospholipid-containing HDL species clarifies our understanding of reverse cholesterol transport, and provides new opportunities for its therapeutic manipulation.
Collapse
Affiliation(s)
- Wendy Jessup
- Centre for Vascular Research, at the School of Medical Sciences, University of New South Wales, Sydney, Australia.
| | | | | | | |
Collapse
|
37
|
Trogan E, Feig JE, Dogan S, Rothblat GH, Angeli V, Tacke F, Randolph GJ, Fisher EA. Gene expression changes in foam cells and the role of chemokine receptor CCR7 during atherosclerosis regression in ApoE-deficient mice. Proc Natl Acad Sci U S A 2006; 103:3781-6. [PMID: 16537455 PMCID: PMC1450154 DOI: 10.1073/pnas.0511043103] [Citation(s) in RCA: 286] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2005] [Indexed: 12/31/2022] Open
Abstract
Atherosclerosis regression is an important clinical goal. In previous studies of regression in mice, the rapid loss of plaque foam cells was explained by emigration to lymph nodes, a process reminiscent of dendritic cells. In the present study, plaque-containing arterial segments from apoE-/- mice were transplanted into WT recipient normolipidemic mice or apoE-/- mice. Three days after transplant, in the WT regression environment, plaque size decreased by approximately 40%, and foam cell content by approximately 75%. In contrast, both parameters increased in apoE-/- recipients. Foam cells were isolated by laser capture microdissection. In WT recipients, there were 3- to 6-fold increases in foam cells of mRNA for liver X receptor alpha and cholesterol efflux factors ABCA1 and SR-BI. Although liver X receptor alpha was induced, there was no detectable expression of its putative activator, peroxisome proliferator-activated receptor gamma. Expression levels of VCAM or MCP-1 were reduced to 25% of levels in pretransplant or apoE-/- recipient samples, but there was induction at the mRNA and protein levels of chemokine receptor CCR7, an essential factor for dendritic cell migration. Remarkably, when CCR7 function was abrogated in vivo by treatment of WT recipients with antibodies to CCR7 ligands CCL19 and CCL21, lesion size and foam cell content were substantially preserved. In summary, in foam cells during atherosclerosis regression, there is induction of CCR7 and a requirement for its function. Taken with the other gene expression data, these results in vivo point to complex relationships among the immune system, nuclear hormone receptors, and inflammation during regression.
Collapse
MESH Headings
- ATP Binding Cassette Transporter 1
- ATP-Binding Cassette Transporters/genetics
- Animals
- Aorta, Thoracic/metabolism
- Aorta, Thoracic/pathology
- Aorta, Thoracic/transplantation
- Apolipoproteins E/deficiency
- Apolipoproteins E/genetics
- Atherosclerosis/genetics
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Atherosclerosis/therapy
- Biological Transport, Active
- Chemokine CCL19
- Chemokine CCL21
- Chemokines, CC/antagonists & inhibitors
- Cholesterol/metabolism
- DNA-Binding Proteins/genetics
- Dyslipidemias/genetics
- Dyslipidemias/metabolism
- Dyslipidemias/pathology
- Dyslipidemias/therapy
- Foam Cells/drug effects
- Foam Cells/metabolism
- Foam Cells/pathology
- Gene Expression/drug effects
- Humans
- In Vitro Techniques
- Inflammation/pathology
- Ligands
- Liver X Receptors
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Orphan Nuclear Receptors
- PPAR gamma/agonists
- PPAR gamma/genetics
- PPAR gamma/metabolism
- Pioglitazone
- Receptors, CCR7
- Receptors, Chemokine/metabolism
- Receptors, Cytoplasmic and Nuclear/genetics
- Thiazolidinediones/pharmacology
Collapse
Affiliation(s)
- Eugene Trogan
- *Marc and Ruti Bell Vascular Biology Program, Leon H. Charney Division of Cardiology/Department of Medicine, New York University School of Medicine, New York, NY 10016
- Graduate School of Biological Sciences and
| | - Jonathan E. Feig
- *Marc and Ruti Bell Vascular Biology Program, Leon H. Charney Division of Cardiology/Department of Medicine, New York University School of Medicine, New York, NY 10016
| | - Snjezana Dogan
- *Marc and Ruti Bell Vascular Biology Program, Leon H. Charney Division of Cardiology/Department of Medicine, New York University School of Medicine, New York, NY 10016
| | - George H. Rothblat
- Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
| | - Véronique Angeli
- Department of Gene and Cell Medicine, Mount Sinai School of Medicine, New York, NY 10029; and
| | - Frank Tacke
- Department of Gene and Cell Medicine, Mount Sinai School of Medicine, New York, NY 10029; and
| | - Gwendalyn J. Randolph
- Department of Gene and Cell Medicine, Mount Sinai School of Medicine, New York, NY 10029; and
| | - Edward A. Fisher
- *Marc and Ruti Bell Vascular Biology Program, Leon H. Charney Division of Cardiology/Department of Medicine, New York University School of Medicine, New York, NY 10016
| |
Collapse
|
38
|
Duong M, Collins HL, Jin W, Zanotti I, Favari E, Rothblat GH. Relative Contributions of ABCA1 and SR-BI to Cholesterol Efflux to Serum From Fibroblasts and Macrophages. Arterioscler Thromb Vasc Biol 2006; 26:541-7. [PMID: 16410457 DOI: 10.1161/01.atv.0000203515.25574.19] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objectives—
Cholesterol efflux is achieved by several mechanisms. This study examines contributions of these pathways to efflux to human serum.
Methods and Results—
Human fibroblasts were stably transfected with SR-BI while ABCA1 was upregulated. Quantitation of cholesterol efflux to human serum demonstrated that there was efflux from cells without either protein. Expression of ABCA1 produced a small increase in efflux, whereas SR-BI expression had a dramatic impact. To quantitate ABCA1 and SR-BI contribution, fibroblasts were pretreated with Probucol and BLT-1 to, respectively, inhibit these efflux proteins. Exposing SR-BI–expressing fibroblasts to BLT-1 inhibited efflux by 67%. Probucol pretreatment of ABCA1-expressing fibroblasts reduced efflux to serum by 26%. A large fraction of total efflux was uninhibited. For both J774 and mouse peritoneal macrophages, contributions of either ABCA1 or SR-BI to efflux to serum were low, with background/uninhibited efflux contributing from 70% to 90% of total efflux.
Conclusions—
We have shown that ABCA1-mediated efflux to serum responds to the pool of lipid-free/poor apolipoproteins, whereas phospholipid-containing particles mediate SR-BI efflux. Although SR-BI and ABCA1 contribute to efflux from fibroblasts and cholesterol-enriched macrophages, a large proportion of the total efflux to human serum is mediated by a mechanism that is neither SR-BI nor ABCA1.
Collapse
Affiliation(s)
- MyNgan Duong
- GI and Nutrition, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | | | | | | | | | | |
Collapse
|
39
|
Zimetti F, Weibel GK, Duong M, Rothblat GH. Measurement of cholesterol bidirectional flux between cells and lipoproteins. J Lipid Res 2006; 47:605-13. [PMID: 16327021 DOI: 10.1194/jlr.m500466-jlr200] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We developed an assay that quantitates bidirectional cholesterol flux between cells and lipoproteins. Incubating Fu5AH cells with increasing concentrations of human serum resulted in increased influx and efflux; however, influx was 2- to 3-fold greater at all serum concentrations. With apolipoprotein B (apoB)-depleted serum, the ratio of influx to efflux (I/E) was close to 1, indicating cholesterol exchange. The apoB fraction of serum induced influx and little efflux, with I/E > 1. Using block lipid transport-1 to block scavenger receptor class B type I (SR-BI)-mediated flux with different acceptors, we determined that 50% to 70% of efflux was via SR-BI. With HDL, 90% of influx was via SR-BI, whereas with LDL or serum, 20% of influx was SR-BI-mediated. Cholesterol-enriched hepatoma cells produced increased efflux without a change in influx, resulting in reduced I/E. The assay was applied to cholesterol-normal and -enriched mouse peritoneal macrophages exposed to serum or LDL. The enrichment enhanced efflux without shifts in influx. With cholesterol-enriched macrophages, HDL efflux was enhanced and influx was greatly reduced. With all lipoproteins, cholesterol enrichment of murine peritoneal macrophages led to a reduced I/E. We conclude that this assay can simultaneously and accurately quantitate cholesterol bidirectional flux and can be applied to a variety of cells exposed to isolated lipoproteins or serum.
Collapse
Affiliation(s)
- Francesca Zimetti
- Gastrointestinal and Nutrition Division, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | | | | | | |
Collapse
|
40
|
Gelissen IC, Harris M, Rye KA, Quinn C, Brown AJ, Kockx M, Cartland S, Packianathan M, Kritharides L, Jessup W. ABCA1 and ABCG1 Synergize to Mediate Cholesterol Export to ApoA-I. Arterioscler Thromb Vasc Biol 2006; 26:534-40. [PMID: 16357317 DOI: 10.1161/01.atv.0000200082.58536.e1] [Citation(s) in RCA: 327] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE To study the acceptor specificity for human ABCG1 (hABCG1)-mediated cholesterol efflux. METHODS AND RESULTS Cells overexpressing hABCG1 were created in Chinese Hamster Ovary (CHO-K1) cells and characterized in terms of lipid composition. hABCG1 expressed in these cells formed homodimers and was mostly present intracellularly. Cholesterol efflux from hABCG1 cells to HDL2 and HDL3 was increased but not to lipid-free apolipoproteins. A range of phospholipid containing acceptors apart from high-density lipoprotein (HDL) subclasses were also efficient in mediating ABCG1-dependent export of cholesterol. Importantly, a buoyant phospholipid-containing fraction generated from incubation of lipid-free apoA-I with macrophages was nearly as efficient as HDL2. The capacity of acceptors to induce ABCG1-mediated efflux was strongly correlated with their total phospholipid content, suggesting that acceptor phospholipids drive ABCG1-mediated efflux. Most importantly, acceptors for ABCG1-mediated cholesterol export could be generated from incubation of cells with lipid-free apoA-I through the action of ABCA1 alone. CONCLUSIONS These results indicate a synergistic relationship between ABCA1 and ABCG1 in peripheral tissues, where ABCA1 lipidates any lipid-poor/free apoA-I to generate nascent or pre-beta-HDL. These particles in turn may serve as substrates for ABCG1-mediated cholesterol export.
Collapse
Affiliation(s)
- Ingrid C Gelissen
- Centre for Vascular Research, School of Medical Sciences, University of New South Wales, Kensington, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Panzenboeck U, Kratzer I, Sovic A, Wintersperger A, Bernhart E, Hammer A, Malle E, Sattler W. Regulatory effects of synthetic liver X receptor- and peroxisome-proliferator activated receptor agonists on sterol transport pathways in polarized cerebrovascular endothelial cells. Int J Biochem Cell Biol 2006; 38:1314-29. [PMID: 16530456 DOI: 10.1016/j.biocel.2006.01.013] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2005] [Revised: 12/20/2005] [Accepted: 01/17/2006] [Indexed: 01/03/2023]
Abstract
The blood-brain barrier contributes to maintain brain cholesterol metabolism and protects this uniquely balanced system from exchange with plasma lipoprotein cholesterol. Brain capillary endothelial cells, representing a physiological barrier to the central nervous system, express apolipoprotein A-I (apoA-I, the major high-density lipoprotein (HDL)-associated apolipoprotein), ATP-binding cassette transporter A1 (ABCA1), and scavenger receptor, class B, type I (SR-BI), proteins that promote cellular cholesterol mobilization. Liver X receptors (LXRs) and peroxisome-proliferator activated receptors (PPARs) are regulators of cholesterol transport, and activation of LXRs and PPARs has potential therapeutic implications for lipid-related neurodegenerative diseases. To clarify the functional impact of LXR/PPAR activation, sterol transport along the: (i) ABCA1/apoA-I and (ii) SR-BI/HDL pathway was investigated in primary, polarized brain capillary endothelial cells, an in vitro model of the blood-brain barrier. Activation of LXR (24(S)OH-cholesterol, TO901317), PPARalpha (bezafibrate, fenofibrate), and PPARgamma (troglitazone, pioglitazone) modulated expression of apoA-I, ABCA1, and SR-BI on mRNA and/or protein levels without compromising transendothelial electrical resistance or tight junction protein expression. LXR-agonists and troglitazone enhanced basolateral-to-apical cholesterol mobilization in the absence of exogenous sterol acceptors. Along with the induction of cell surface-located ABCA1, several agonists enhanced cholesterol mobilization in the presence of exogenous apoA-I, while efflux of 24(S)OH-cholesterol (the major brain cholesterol metabolite) in the presence of exogenous HDL remained unaffected. Summarizing, in cerebrovascular endothelial cells apoA-I, ABCA1, and SR-BI represent drug targets for LXR and PPAR-agonists to interfere with cholesterol homeostasis at the periphery of the central nervous system.
Collapse
MESH Headings
- ATP Binding Cassette Transporter 1
- ATP-Binding Cassette Transporters/genetics
- ATP-Binding Cassette Transporters/metabolism
- Animals
- Apolipoprotein A-I/genetics
- Apolipoprotein A-I/metabolism
- Biological Transport/drug effects
- Biological Transport/physiology
- Cell Polarity/physiology
- Cells, Cultured
- Clofibric Acid/chemical synthesis
- Clofibric Acid/pharmacology
- DNA-Binding Proteins/agonists
- DNA-Binding Proteins/metabolism
- Endothelium, Vascular/cytology
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/metabolism
- Immunoblotting
- Lipoproteins, HDL/metabolism
- Lipoproteins, HDL3
- Liver X Receptors
- Microscopy, Fluorescence
- Models, Biological
- Orphan Nuclear Receptors
- Peroxisome Proliferator-Activated Receptors/agonists
- Peroxisome Proliferator-Activated Receptors/metabolism
- Pioglitazone
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, Cytoplasmic and Nuclear/agonists
- Receptors, Cytoplasmic and Nuclear/metabolism
- Scavenger Receptors, Class B/genetics
- Scavenger Receptors, Class B/metabolism
- Signal Transduction/drug effects
- Sterols/chemistry
- Sterols/metabolism
- Swine
- Thiazolidinediones/chemical synthesis
- Thiazolidinediones/pharmacology
- Transcription, Genetic/drug effects
- Transcription, Genetic/genetics
Collapse
Affiliation(s)
- Ute Panzenboeck
- Medical University Graz, Center of Molecular Medicine, Institute of Molecular Biology and Biochemistry, Harrachgasse 21, A-8010 Graz, Austria.
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Towns R, Menon KMJ. The role of cyclic AMP response element binding protein in transactivation of scavenger receptor class B type I promoter in transfected cells and in primary cultures of rat theca-interstitial cells. Mol Cell Endocrinol 2005; 245:23-30. [PMID: 16298471 DOI: 10.1016/j.mce.2005.09.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2005] [Accepted: 09/23/2005] [Indexed: 11/30/2022]
Abstract
In the ovary, lutropin (LH) stimulates the selective uptake and transport of cholesterol for steroid biosynthesis from HDL particles via the scavenger receptor class B type I (SR-BI). Furthermore the expression of SR-BI mRNA in the ovary is stimulated by LH and cyclic AMP (cAMP). Since the promoter of the rat SR-BI gene is devoid of consensus cyclic AMP response element (CRE) sequences, this study examined if cAMP response element binding protein (CREB) plays a role in the transactivation of SR-BI promoter (SR-BIpr). The transactivation of SR-BIpr was examined in transfected 293T cells and human granulosa SVOG-4o cells, and in primary cultures of rat theca-interstitial cells infected with adenoviral constructs containing the SR-BIpr and a luciferase reporter gene. Dose-related increases in SR-BRpr activity ranging from 2- to 4-fold was induces by 293T cells co-transfected with the catalytic subunit of protein kinase A (cPKA). Co-transfections with CREB and cPKA produced a concentration-dependent increase ranging from 6- to 32-fold. The cAMP-mediated transactivation was significantly attenuated by co-transfection with CREB M1, a non-phosphorylatable, dominant-negative form of CREB. An increase in transactivation of SR-BIpr activity was also seen in SVOG-4o cells co-transfected with CREB. In primary cultures of rat theca-interstitial (T-I) cells infected with an adenoviral construct of SR-BIpr, forskolin produced a marked increase in promoter activity. These data indicate that stimulation of the cAMP-PKA-CREB pathway enhances rat SR-BIpr activity and substantiate the role of CREB as an intermediary in this process. The absence of canonical CRE sequences in the rat SR-BIpr suggests that the activation of SR-BI by CREB may occur either through non-canonical CRE sequences or through additional transcription factors that cooperate with CREB in the activation of SR-BI promoter activity.
Collapse
MESH Headings
- Animals
- Cell Line
- Cells, Cultured
- Cholesterol, HDL/physiology
- Colforsin/pharmacology
- Cyclic AMP/physiology
- Cyclic AMP Response Element-Binding Protein/genetics
- Cyclic AMP Response Element-Binding Protein/physiology
- Cyclic AMP-Dependent Protein Kinases/genetics
- Cyclic AMP-Dependent Protein Kinases/physiology
- Female
- Gene Expression Regulation/drug effects
- Humans
- Luteinizing Hormone/physiology
- Promoter Regions, Genetic/drug effects
- Promoter Regions, Genetic/genetics
- Promoter Regions, Genetic/physiology
- RNA, Messenger/genetics
- RNA, Messenger/physiology
- Rats
- Rats, Sprague-Dawley
- Scavenger Receptors, Class B/genetics
- Scavenger Receptors, Class B/physiology
- Theca Cells/physiology
- Transcriptional Activation
- Transfection
Collapse
Affiliation(s)
- Roberto Towns
- Department of Obstetrics and Gynecology, 6428 Medical Science I, 1150 West Medical Center Drive, University of Michigan Medical School, Ann Arbor, 48109-0617, USA
| | | |
Collapse
|
43
|
Westendorf T, Graessler J, Kopprasch S. Hypochlorite-oxidized low-density lipoprotein upregulates CD36 and PPARgamma mRNA expression and modulates SR-BI gene expression in murine macrophages. Mol Cell Biochem 2005; 277:143-52. [PMID: 16132726 DOI: 10.1007/s11010-005-5873-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2005] [Accepted: 04/20/2005] [Indexed: 12/16/2022]
Abstract
The uptake of oxidized low-density lipoprotein (oxLDL) by scavenger receptors of macrophages with resulting foam cell formation is considered a critical event in atherogenesis. Since hypochlorite-oxidized LDL (HOCl-LDL) has been shown to be recognized by macrophages and evidence was provided that HOCl-LDL is internalized via class B scavenger receptors CD36 and SR-BI, the regulatory relationships between CD36, SR-BI, and the nuclear transcription factor PPARgamma in murine macrophages (RAW 264.7) on exposure to HOCl-LDL were examined. Using the highly sensitive real-time RT-PCR we could demonstrate that HOCl-LDL upregulated CD36 and PPARgamma levels dose- and time dependently while modulating SR-BI message levels differently in dependence on HOCl-LDL concentration and incubation time. On exposure of macrophages to HOCl-LDL but not native LDL in varying concentrations, a significant positive correlation between CD36 and PPARgamma (rho = 0.603, p = 0.001) was observed indicating the presence of a positive feedback mechanism by which HOCl-LDL could promote its own uptake. The transcriptional expression of SR-BI in macrophages was not significantly related to PPARgamma mRNA levels after treatment with HOCl-LDL suggesting a differential regulation of the two members of the scavenger receptor class B family in response to HOCl-LDL.
Collapse
Affiliation(s)
- Thomas Westendorf
- Department of Internal Medicine III, Pathological Biochemistry, Carl Gustav Carus Medical School, University of Technology, Dresden, Germany
| | | | | |
Collapse
|
44
|
Llaverias G, Lacasa D, Vázquez-Carrera M, Sánchez RM, Laguna JC, Alegret M. Cholesterol regulation of genes involved in sterol trafficking in human THP-1 macrophages. Mol Cell Biochem 2005; 273:185-91. [PMID: 16013454 DOI: 10.1007/s11010-005-0624-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Modulation of the expression of genes involved in the control of cholesterol homeostasis by sterols in macrophages is crucial to foam cell formation. To characterize this regulation in THP-1 macrophages, we examined the effect of sterol loading and unloading on the expression of a number of genes that participate in lipoprotein uptake and cholesterol efflux. Sterol loading by exposure to acetylated LDL for 24 h resulted in an increase in free and esterified cholesterol of 1.4 and 1.8-fold, respectively. Under these conditions, the mRNA levels for SR-A were reduced a 59%, while those of CYP27 were increased by 4.6-fold. However, the expression of other genes involved in cholesterol efflux (ABCA1, ABCG1 and CLA-1) was not modified, despite a high intracellular cholesterol accumulation specially in the form of esterified cholesterol. On the other hand, HDL exposure reduced intracellular cholesterol content to 70%, and caused an increase in the expression of CD36 (78%), SR-A (51%) and CLA-1 (136%). Conversely, the expression of ABCA1, ABCG1 and CYP27 was decreased by 49, 67 and 57%, respectively. These findings indicate that in THP-1 macrophages, the expression of genes for receptors involved in lipoprotein binding and uptake tends to decrease upon cholesterol loading and to increase by cholesterol depletion, while the opposite pattern is found regarding the mRNA levels for proteins involved in cholesterol efflux.
Collapse
Affiliation(s)
- Gemma Llaverias
- Unitat de Farmacologia, Departament de Farmacologia i Química Terapéutica, Facultat de Farmàcia, Universitat de Barcelona, Spain
| | | | | | | | | | | |
Collapse
|