1
|
Chini A, Guha P, Rishi A, Bhat N, Covarrubias A, Martinez V, Devejian L, Nguyen BN, Mandal SS. HDLR-SR-BI Expression and Cholesterol Uptake are Regulated via Indoleamine-2,3-dioxygenase 1 in Macrophages under Inflammation. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2025; 41:11253-11271. [PMID: 40309829 DOI: 10.1021/acs.langmuir.4c03005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2025]
Abstract
Macrophages play crucial roles in inflammation, and their dysfunction is a contributing factor to various human diseases. Maintaining the balance of cholesterol and lipid metabolism is central to macrophage function, and any disruption in this balance increases the risk of conditions such as cardiovascular disease, atherosclerosis, and others. HDLR-SR-BI (SR-BI) is pivotal for reverse cholesterol transport and cholesterol homeostasis. Our studies demonstrate that the expression of SR-BI is reduced along with a decrease in cholesterol uptake in macrophages, both of which are regulated by the activation of NF-κB. Furthermore, we have discovered that indoleamine-2,3-dioxygenase 1 (IDO1), which is a critical player in tryptophan (Trp) catabolism, is crucial to the regulation of SR-BI expression. Inflammation leads to elevated levels of IDO1 and the associated Trp catabolite kynurenine (KYN) in macrophages. Interestingly, knockdown or inhibition of IDO1 results in the downregulation of LPS-induced inflammation, decreased KYN levels, and the restoration of SR-BI expression as well as cholesterol uptake in macrophages. Beyond LPS, stimulation with pro-inflammatory cytokine IFNγ exhibits similar trends in inflammatory response, IDO1 regulation, and cholesterol uptake in macrophages. These observations suggest that IDO1 plays a critical role in SR-BI expression and cholesterol uptake in macrophages under inflammation.
Collapse
Affiliation(s)
- Avisankar Chini
- Gene Regulation and Epigenetics Research Laboratory, Department of Chemistry and Biochemistry, The University of Texas at Arlington, Arlington, Texas 76019, United States
| | - Prarthana Guha
- Gene Regulation and Epigenetics Research Laboratory, Department of Chemistry and Biochemistry, The University of Texas at Arlington, Arlington, Texas 76019, United States
| | - Ashcharya Rishi
- Gene Regulation and Epigenetics Research Laboratory, Department of Chemistry and Biochemistry, The University of Texas at Arlington, Arlington, Texas 76019, United States
| | - Nagashree Bhat
- Gene Regulation and Epigenetics Research Laboratory, Department of Chemistry and Biochemistry, The University of Texas at Arlington, Arlington, Texas 76019, United States
| | - Angel Covarrubias
- Gene Regulation and Epigenetics Research Laboratory, Department of Chemistry and Biochemistry, The University of Texas at Arlington, Arlington, Texas 76019, United States
| | - Valeria Martinez
- Gene Regulation and Epigenetics Research Laboratory, Department of Chemistry and Biochemistry, The University of Texas at Arlington, Arlington, Texas 76019, United States
| | - Lucine Devejian
- Gene Regulation and Epigenetics Research Laboratory, Department of Chemistry and Biochemistry, The University of Texas at Arlington, Arlington, Texas 76019, United States
| | - Bao Nhi Nguyen
- Gene Regulation and Epigenetics Research Laboratory, Department of Chemistry and Biochemistry, The University of Texas at Arlington, Arlington, Texas 76019, United States
| | - Subhrangsu S Mandal
- Gene Regulation and Epigenetics Research Laboratory, Department of Chemistry and Biochemistry, The University of Texas at Arlington, Arlington, Texas 76019, United States
| |
Collapse
|
2
|
Del Bianco V, Ferreira GDS, Bochi APG, Pinto PR, Rodrigues LG, Furukawa LNS, Okamoto MM, Almeida JA, da Silveira LKR, Santos AS, Bispo KCS, Capelozzi VL, Correa-Giannella ML, da Silva AA, Velosa APP, Nakandakare ER, Machado UF, Teodoro WPR, Passarelli M, Catanozi S. Aerobic Exercise Training Protects Against Insulin Resistance, Despite Low-Sodium Diet-Induced Increased Inflammation and Visceral Adiposity. Int J Mol Sci 2024; 25:10179. [PMID: 39337664 PMCID: PMC11432465 DOI: 10.3390/ijms251810179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 09/18/2024] [Accepted: 09/20/2024] [Indexed: 09/30/2024] Open
Abstract
Dietary sodium restriction increases plasma triglycerides (TG) and total cholesterol (TC) concentrations as well as causing insulin resistance and stimulation of the renin-angiotensin-aldosterone system (RAAS) and the sympathetic nervous system. Stimulation of the angiotensin II type-1 receptor (AT1) is associated with insulin resistance, inflammation, and the inhibition of adipogenesis. The current study investigated whether aerobic exercise training (AET) mitigates or inhibits the adverse effects of dietary sodium restriction on adiposity, inflammation, and insulin sensitivity in periepididymal adipose tissue. LDL receptor knockout mice were fed either a normal-sodium (NS; 1.27% NaCl) or a low-sodium (LS; 0.15% NaCl) diet and were either subjected to AET for 90 days or kept sedentary. Body mass, blood pressure (BP), hematocrit, plasma TC, TG, glucose and 24-hour urinary sodium (UNa) concentrations, insulin sensitivity, lipoprotein profile, histopathological analyses, and gene and protein expression were determined. The results were evaluated using two-way ANOVA. Differences were not observed in BP, hematocrit, diet consumption, and TC. The LS diet was found to enhance body mass, insulin resistance, plasma glucose, TG, LDL-C, and VLDL-TG and reduce UNa, HDL-C, and HDL-TG, showing a pro-atherogenic lipid profile. In periepididymal adipose tissue, the LS diet increased tissue mass, TG, TC, AT1 receptor, pro-inflammatory macro-phages contents, and the area of adipocytes; contrarily, the LS diet decreased anti-inflammatory macrophages, protein contents and the transcription of genes related to insulin sensitivity. The AET prevented insulin resistance, but did not protect against dyslipidemia, adipose tissue pro-inflammatory profile, increased tissue mass, AT1 receptor expression, TG, and TC induced by the LS diet.
Collapse
Affiliation(s)
- Vanessa Del Bianco
- Laboratorio de Lipides (LIM-10), Hospital das Clinicas (HCFMUSP) da Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo 01246 000, Brazil; (V.D.B.); (G.d.S.F.); (A.P.G.B.); (P.R.P.); (L.G.R.); (E.R.N.); (M.P.)
| | - Guilherme da Silva Ferreira
- Laboratorio de Lipides (LIM-10), Hospital das Clinicas (HCFMUSP) da Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo 01246 000, Brazil; (V.D.B.); (G.d.S.F.); (A.P.G.B.); (P.R.P.); (L.G.R.); (E.R.N.); (M.P.)
| | - Ana Paula Garcia Bochi
- Laboratorio de Lipides (LIM-10), Hospital das Clinicas (HCFMUSP) da Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo 01246 000, Brazil; (V.D.B.); (G.d.S.F.); (A.P.G.B.); (P.R.P.); (L.G.R.); (E.R.N.); (M.P.)
| | - Paula Ramos Pinto
- Laboratorio de Lipides (LIM-10), Hospital das Clinicas (HCFMUSP) da Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo 01246 000, Brazil; (V.D.B.); (G.d.S.F.); (A.P.G.B.); (P.R.P.); (L.G.R.); (E.R.N.); (M.P.)
| | - Letícia Gomes Rodrigues
- Laboratorio de Lipides (LIM-10), Hospital das Clinicas (HCFMUSP) da Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo 01246 000, Brazil; (V.D.B.); (G.d.S.F.); (A.P.G.B.); (P.R.P.); (L.G.R.); (E.R.N.); (M.P.)
| | - Luzia Naoko Shinohara Furukawa
- Laboratory of Renal Pathophysiology, Department of Internal Medicine, School of Medicine, University of São Paulo, São Paulo 01246 000, Brazil;
| | - Maristela Mitiko Okamoto
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508 000, Brazil; (M.M.O.); (U.F.M.)
| | - Jaíne Alves Almeida
- Rheumatology Division of the Hospital das Clinicas, University of São Paulo Medical School, São Paulo 01246 000, Brazil; (J.A.A.); (L.K.R.d.S.); (A.P.P.V.); (W.P.R.T.)
| | - Lizandre Keren Ramos da Silveira
- Rheumatology Division of the Hospital das Clinicas, University of São Paulo Medical School, São Paulo 01246 000, Brazil; (J.A.A.); (L.K.R.d.S.); (A.P.P.V.); (W.P.R.T.)
| | - Aritania Sousa Santos
- Laboratorio de Carboidratos e Radioimunoensaios (Laboratorio de Investigações Médicas, LIM-18), Faculdade de Medicina, Universidade de Sao Paulo (FMUSP), Sao Paulo 01246 000, Brazil; (A.S.S.); (M.L.C.-G.)
| | - Kely Cristina Soares Bispo
- Department of Pathology of the Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, FMUSP, Sao Paulo 01246 000, Brazil; (K.C.S.B.); (V.L.C.)
| | - Vera Luiza Capelozzi
- Department of Pathology of the Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, FMUSP, Sao Paulo 01246 000, Brazil; (K.C.S.B.); (V.L.C.)
| | - Maria Lucia Correa-Giannella
- Laboratorio de Carboidratos e Radioimunoensaios (Laboratorio de Investigações Médicas, LIM-18), Faculdade de Medicina, Universidade de Sao Paulo (FMUSP), Sao Paulo 01246 000, Brazil; (A.S.S.); (M.L.C.-G.)
| | - Alexandre Alves da Silva
- Department of Physiology and Biophysics, Mississippi Center for Obesity Research, Cardiorenal and Metabolic Diseases Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA;
| | - Ana Paula Pereira Velosa
- Rheumatology Division of the Hospital das Clinicas, University of São Paulo Medical School, São Paulo 01246 000, Brazil; (J.A.A.); (L.K.R.d.S.); (A.P.P.V.); (W.P.R.T.)
| | - Edna Regina Nakandakare
- Laboratorio de Lipides (LIM-10), Hospital das Clinicas (HCFMUSP) da Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo 01246 000, Brazil; (V.D.B.); (G.d.S.F.); (A.P.G.B.); (P.R.P.); (L.G.R.); (E.R.N.); (M.P.)
| | - Ubiratan Fabres Machado
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508 000, Brazil; (M.M.O.); (U.F.M.)
| | - Walcy Paganelli Rosolia Teodoro
- Rheumatology Division of the Hospital das Clinicas, University of São Paulo Medical School, São Paulo 01246 000, Brazil; (J.A.A.); (L.K.R.d.S.); (A.P.P.V.); (W.P.R.T.)
| | - Marisa Passarelli
- Laboratorio de Lipides (LIM-10), Hospital das Clinicas (HCFMUSP) da Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo 01246 000, Brazil; (V.D.B.); (G.d.S.F.); (A.P.G.B.); (P.R.P.); (L.G.R.); (E.R.N.); (M.P.)
- Programa de Pós Graduação em Medicina, Universidade Nove de Julho, Sao Paulo 01525 000, Brazil
| | - Sergio Catanozi
- Laboratorio de Lipides (LIM-10), Hospital das Clinicas (HCFMUSP) da Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo 01246 000, Brazil; (V.D.B.); (G.d.S.F.); (A.P.G.B.); (P.R.P.); (L.G.R.); (E.R.N.); (M.P.)
| |
Collapse
|
3
|
Knaack DA, Chang J, Thomas MJ, Sorci-Thomas MG, Chen Y, Sahoo D. Scavenger receptor class B type I is required for efficient glucose uptake and metabolic homeostasis in adipocytes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.21.554190. [PMID: 37662321 PMCID: PMC10473602 DOI: 10.1101/2023.08.21.554190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Obesity is a worldwide epidemic and places individuals at a higher risk for developing comorbidities that include cardiovascular disease and type 2 diabetes. Adipose tissue contains adipocytes that are responsible for lipid metabolism and reducing misdirected lipid storage. Adipocytes facilitate this process through insulin-mediated uptake of glucose and its subsequent metabolism into triglycerides for storage. During obesity, adipocytes become insulin resistant and have a reduced ability to mediate glucose import, thus resulting in whole-body metabolic dysfunction. Scavenger receptor class B type I (SR-BI) has been implicated in glucose uptake in skeletal muscle and adipocytes via its native ligands, apolipoprotein A-1 and high-density lipoproteins. Further, SR-BI translocation to the cell surface in adipocytes is sensitive to insulin stimulation. Using adipocytes differentiated from ear mesenchymal stem cells isolated from wild-type and SR-BI knockout (SR-BI -/- ) mice as our model system, we tested the hypothesis that SR-BI is required for insulin-mediated glucose uptake and regulation of energy balance in adipocytes. We demonstrated that loss of SR-BI in adipocytes resulted in inefficient glucose uptake regardless of cell surface expression levels of glucose transporter 4 compared to WT adipocytes. We also observed reduced glycolytic capacity, increased lipid biosynthesis, and dysregulated expression of lipid metabolism genes in SR-BI -/- adipocytes compared to WT adipocytes. These results partially support our hypothesis and suggest a novel role for SR-BI in glucose uptake and metabolic homeostasis in adipocytes.
Collapse
|
4
|
Cook JR, Ausiello J. Functional ACE2 deficiency leading to angiotensin imbalance in the pathophysiology of COVID-19. Rev Endocr Metab Disord 2022; 23:151-170. [PMID: 34195965 PMCID: PMC8245275 DOI: 10.1007/s11154-021-09663-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/04/2021] [Indexed: 02/07/2023]
Abstract
SARS-CoV-2, the virus responsible for COVID-19, uses angiotensin converting enzyme 2 (ACE2) as its primary cell-surface receptor. ACE2 is a key enzyme in the counter-regulatory pathway of the broader renin-angiotensin system (RAS) that has been implicated in a broad array of human pathology. The RAS is composed of two competing pathways that work in opposition to each other: the "conventional" arm involving angiotensin converting enzyme (ACE) generating angiotensin-2 and the more recently identified ACE2 pathway that generates angiotensin (1-7). Following the original SARS pandemic, additional studies suggested that coronaviral binding to ACE2 resulted in downregulation of the membrane-bound enzyme. Given the similarities between the two viruses, many have posited a similar process with SARS-CoV-2. Proponents of this ACE2 deficiency model argue that downregulation of ACE2 limits its enzymatic function, thereby skewing the delicate balance between the two competing arms of the RAS. In this review we critically examine this model. The available data remain incomplete but are consistent with the possibility that the broad multisystem dysfunction of COVID-19 is due in large part to functional ACE2 deficiency leading to angiotensin imbalance with consequent immune dysregulation and endothelial cell dysfunction.
Collapse
Affiliation(s)
- Joshua R Cook
- New York-Presbyterian Hospital and the Columbia University Irving Medical Center, New York, NY, USA
| | - John Ausiello
- New York-Presbyterian Hospital and the Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
5
|
Xu H, Thomas MJ, Kaul S, Kallinger R, Ouweneel AB, Maruko E, Oussaada SM, Jongejan A, Cense HA, Nieuwdorp M, Serlie MJ, Goldberg IJ, Civelek M, Parks BW, Lusis AJ, Knaack D, Schill RL, May SC, Reho JJ, Grobe JL, Gantner B, Sahoo D, Sorci-Thomas MG. Pcpe2, a Novel Extracellular Matrix Protein, Regulates Adipocyte SR-BI-Mediated High-Density Lipoprotein Uptake. Arterioscler Thromb Vasc Biol 2021; 41:2708-2725. [PMID: 34551590 PMCID: PMC8551036 DOI: 10.1161/atvbaha.121.316615] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 08/24/2021] [Indexed: 01/22/2023]
Abstract
Objective To investigate the role of adipocyte Pcpe2 (procollagen C-endopeptidase enhancer 2) in SR-BI (scavenger receptor class BI)-mediated HDL-C (high-density lipoprotein cholesterol) uptake and contributions to adipose lipid storage. Approach and Results Pcpe2, a glycoprotein devoid of intrinsic proteolytic activity, is believed to participate in extracellular protein-protein interactions, supporting SR-BI- mediated HDL-C uptake. In published studies, Pcpe2 deficiency increased the development of atherosclerosis by reducing SR-BI-mediated HDL-C catabolism, but the biological impact of this deficiency on adipocyte SR-BI-mediated HDL-C uptake is unknown. Differentiated cells from Ldlr-/-/Pcpe2-/- (Pcpe2-/-) mouse adipose tissue showed elevated SR-BI protein levels, but significantly reduced HDL-C uptake compared to Ldlr-/- (control) adipose tissue. SR-BI-mediated HDL-C uptake was restored by preincubation of cells with exogenous Pcpe2. In diet-fed mice lacking Pcpe2, significant reductions in visceral, subcutaneous, and brown adipose tissue mass were observed, despite elevations in plasma triglyceride and cholesterol concentrations. Significant positive correlations exist between adipose mass and Pcpe2 expression in both mice and humans. Conclusions Overall, these findings reveal a novel and unexpected function for Pcpe2 in modulating SR-BI expression and function as it relates to adipose tissue expansion and cholesterol balance in both mice and humans.
Collapse
Affiliation(s)
- Hao Xu
- Department of Medicine, Division of Endocrinology and Molecular Medicine
| | - Michael J. Thomas
- Pharmacology & Toxicology and
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Sushma Kaul
- Department of Medicine, Division of Endocrinology and Molecular Medicine
| | | | - Amber B. Ouweneel
- Department of Medicine, Division of Endocrinology and Molecular Medicine
| | - Elisa Maruko
- Department of Medicine, Division of Endocrinology and Molecular Medicine
| | - Sabrina M. Oussaada
- Department of Endocrinology and Metabolism, Amsterdam University Medical Centers, Academic Medical Center, Amsterdam, the Netherlands
| | - Aldo Jongejan
- Department of Bioinformatics, Amsterdam University Medical Centers, Academic Medical Center, Amsterdam, the Netherlands
| | - Huib A. Cense
- Department of Surgery, Rode Kruis Ziekenhuis, Beverwijk, the Netherlands
| | - Max Nieuwdorp
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, Academic Medical Center, Amsterdam, the Netherlands
| | - Mireille J. Serlie
- Department of Endocrinology and Metabolism, Amsterdam University Medical Centers, Academic Medical Center, Amsterdam, the Netherlands
| | - Ira J. Goldberg
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, New York University Langone School of Medicine, New York, NY
| | - Mete Civelek
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA
| | - Brian W. Parks
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI
| | - Aldons J. Lusis
- Department of Medicine, Human Genetics, Microbiology, Immunology and Molecular Genetics, University of California Los Angeles, California
| | - Darcy Knaack
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Rebecca L. Schill
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Sarah C. May
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - John J. Reho
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
- Comprehensive Rodent Metabolic Phenotyping Core
| | - Justin L. Grobe
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
- Comprehensive Rodent Metabolic Phenotyping Core
- Department of Biomedical Engineering
| | - Benjamin Gantner
- Department of Medicine, Division of Endocrinology and Molecular Medicine
| | - Daisy Sahoo
- Department of Medicine, Division of Endocrinology and Molecular Medicine
- Pharmacology & Toxicology and
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Mary G. Sorci-Thomas
- Department of Medicine, Division of Endocrinology and Molecular Medicine
- Pharmacology & Toxicology and
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
6
|
Sharma B, Hussain T, Khan MA, Jaiswal V. Exploring AT2R and its polymorphism in different diseases: An approach to develop AT2R as a drug target beyond hypertension. Curr Drug Targets 2021; 23:99-113. [PMID: 34365920 DOI: 10.2174/1389450122666210806125919] [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: 02/01/2021] [Revised: 05/30/2021] [Accepted: 06/02/2021] [Indexed: 11/22/2022]
Abstract
The Angiotensin II type 2 receptor (AT2R) is one of the critical components of the renin-angiotensin system (RAS), which performs diverse functions like inhibiting cell differentiation, cell proliferation, vasodilatation, reduces oxidative stress and inflammation. AT2R is relatively less studied in comparison to other components of RAS despite its uniqueness (sex-linked) and diverse functions. The AT2R is differentially expressed in different tissues, and its gene polymorphisms are associated with several diseases. The molecular mechanism behind the association of AT2R and its gene polymorphisms with the diseases remains to be fully understood, which hinders the development of AT2R as a drug target. Single nucleotide polymorphisms (SNPs) in AT2R are found at different locations (exons, introns, promoter, and UTR regions) and were studied for association with different diseases. There may be different mechanisms behind these associations as some AT2R SNP variants were associated with differential expression, the SNPs (A1675G/A1332G) affect the alternate splicing of AT2R mRNA, A1332G genotype results in shortening of the AT2R mRNA and subsequently defective protein. Few SNPs were found to be associated with the diseases in either females (C4599A) or males (T1334C). Several other SNPs were expected to be associated with other similar/related diseases, but studies have not been done yet. The present review emphasizes on the significance of AT2R and its polymorphisms associated with the diseases to explore the precise role of AT2R in different diseases and the possibility to develop AT2R as a potential drug target.
Collapse
Affiliation(s)
- Bhanu Sharma
- Faculty of Applied Sciences and Biotechnology Shoolini University of Biotechnology and Management Sciences, Post Box No. 9, Head post Office, Solan, Himachal Pradesh. India
| | - Tahir Hussain
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas. United States
| | - Mohammed Azhar Khan
- Faculty of Applied Sciences and Biotechnology Shoolini University of Biotechnology and Management Sciences, Post Box No. 9, Head post Office, Solan, Himachal Pradesh. India
| | - Varun Jaiswal
- Department of Food and Nutrition, College of BioNano Technology, Gachon University, Gyeonggi-do 13120. South Korea
| |
Collapse
|
7
|
Rivera K, Quiñones V, Amigo L, Santander N, Salas-Pérez F, Xavier A, Fernández-Galilea M, Carrasco G, Cabrera D, Arrese M, Busso D, Andia ME, Rigotti A. Lipoprotein receptor SR-B1 deficiency enhances adipose tissue inflammation and reduces susceptibility to hepatic steatosis during diet-induced obesity in mice. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1866:158909. [PMID: 33631309 DOI: 10.1016/j.bbalip.2021.158909] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 02/05/2021] [Accepted: 02/17/2021] [Indexed: 12/31/2022]
Abstract
Scavenger receptor class B type 1 (SR-B1) is a membrane lipoprotein receptor/lipid transporter involved in the pathogenesis of atherosclerosis, but its role in obesity and fatty liver development is unclear. Here, we determined the effects of SR-B1 deficiency on plasma metabolic and inflammatory parameters as well as fat deposition in adipose tissue and liver during obesity. To induce obesity, we performed high-fat diet (HFD) exposure for 12 weeks in male SR-B1 knock-out (SR-B1-/-, n = 14) and wild-type (WT, n = 12) mice. Compared to HFD-fed WT mice, plasma from HFD-fed SR-B1-/- animals exhibited increased total cholesterol, triglycerides (TG) and tumor necrosis factor-α (TNF-α) levels. In addition, hypertrophied adipocytes and macrophage-containing crown-like structures (CLS) were observed in adipose tissue from HFD-fed SR-B1 deficient mice. Remarkably, liver from obese SR-B1-/- mice showed attenuated TG content, dysregulation in hepatic peroxisome proliferator-activated receptors (PPARs) expression, increased hepatic TG secretion, and altered hepatic fatty acid (FA) composition. In conclusion, we show that SR-B1 deficiency alters the metabolic environment of obese mice through modulation of liver and adipose tissue lipid accumulation. Our findings provide the basis for further elucidation of SR-B1's role in obesity and fatty liver, two major public health issues that increase the risk of advanced chronic diseases and overall mortality.
Collapse
Affiliation(s)
- Katherine Rivera
- Department of Nutrition, Diabetes, and Metabolism, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile; Millennium Nucleus for Cardiovascular Magnetic Resonance, Santiago 7820436, Chile.
| | - Verónica Quiñones
- Department of Nutrition, Diabetes, and Metabolism, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile.
| | - Ludwig Amigo
- Department of Nutrition, Diabetes, and Metabolism, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile.
| | - Nicolás Santander
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA 94143, USA.
| | - Francisca Salas-Pérez
- Instituto de Ciencias de la Salud, Universidad de O'Higgins, Rancagua 2820000, Chile.
| | - Aline Xavier
- Millennium Nucleus for Cardiovascular Magnetic Resonance, Santiago 7820436, Chile; Biomedical Imaging Center, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile.
| | - Marta Fernández-Galilea
- Centre for Nutrition Research, School of Pharmacy and Nutrition, University of Navarra, Pamplona 31008, Spain; IDISNA, Navarra's Health Research Institute, Pamplona 31008, Spain.
| | - Gonzalo Carrasco
- Department of Pathology, Hospital Clínico Universidad de Chile, Santiago 8320000, Chile.
| | - Daniel Cabrera
- Department of Gastroenterology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile; Faculty of Medical Sciences, School of Medicine, Universidad Bernardo O Higgins, Santiago 8370854, Chile.
| | - Marco Arrese
- Department of Gastroenterology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile; Centro de Envejecimiento y Regeneración (CARE), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.
| | - Dolores Busso
- Biomedical Research and Innovation Center, Faculty of Medicine, Universidad de los Andes, Santiago 111711, Chile.
| | - Marcelo E Andia
- Millennium Nucleus for Cardiovascular Magnetic Resonance, Santiago 7820436, Chile; Biomedical Imaging Center, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile.
| | - Attilio Rigotti
- Department of Nutrition, Diabetes, and Metabolism, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile; Millennium Nucleus for Cardiovascular Magnetic Resonance, Santiago 7820436, Chile; Center of Molecular Nutrition and Chronic Diseases, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile.
| |
Collapse
|
8
|
Insulin Rescued MCP-1-Suppressed Cholesterol Efflux to Large HDL2 Particles via ABCA1, ABCG1, SR-BI and PI3K/Akt Activation in Adipocytes. Cardiovasc Drugs Ther 2021; 36:665-678. [PMID: 33740174 PMCID: PMC9270268 DOI: 10.1007/s10557-021-07166-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/24/2021] [Indexed: 12/02/2022]
Abstract
Purpose Intracellular cholesterol imbalance plays an important role in adipocyte dysfunction of obesity. However, it is unclear whether obesity induced monocyte chemoattractant protein-1 (MCP-1) causes the adipocyte cholesterol imbalance. In this study, we hypothesize that MCP-1 impairs cholesterol efflux of adipocytes to HDL2 and insulin rescues this process. Methods We recruited coronary artery disease (CAD) patients with obesity and overweight to analyze the association between MCP-1 and HDL2-C by Pearson correlation coefficients. We performed [3H]-cholesterol efflux assay to demonstrate the effect of MCP-1 and insulin on cholesterol efflux from 3T3-L1 adipocytes to large HDL2 particles. Western blot, RT-qPCR, cell-surface protein assay, and confocal microscopy were performed to determine the regulatory mechanism. Results Plasma MCP-1 concentrations were negatively correlated with HDL2-C in CAD patients with obesity and overweight (r = −0.60, p < 0.001). In differentiated 3T3-L1 adipocytes, MCP-1 reduced cholesterol efflux to large HDL2 particles by 55.4% via decreasing ATP-binding cassette A1 (ABCA1), ABCG1, and scavenger receptor class B type I (SR-BI) expression. Intriguingly, insulin rescued MCP-1 mediated-inhibition of cholesterol efflux to HDL2 in an Akt phosphorylation-dependent manner. The rescue efficacy of insulin was 138.2% for HDL2. Moreover, insulin increased mRNA and protein expression of ABCA1, ABCG1, and SR-BI at both transcriptional and translational levels via the PI3K/Akt activation. Conclusions These findings indicate that MCP-1 impairs cholesterol efflux to large HDL2 particles in adipocytes, which is reversed by insulin via the upregulation of ABCA1, ABCG1, and SR-BI. Therefore, insulin might improve cholesterol imbalance by an anti-inflammatory effect in adipocytes. Clinical trial registration number: ChiCTR2000033297; Date of registration: 2020/05/ 27; Retrospectively registered. Supplementary Information The online version contains supplementary material available at 10.1007/s10557-021-07166-2.
Collapse
|
9
|
SR-BI deficiency disassociates obesity from hepatic steatosis and glucose intolerance development in high fat diet-fed mice. J Nutr Biochem 2021; 89:108564. [DOI: 10.1016/j.jnutbio.2020.108564] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 11/24/2020] [Accepted: 11/24/2020] [Indexed: 01/05/2023]
|
10
|
Curley S, Gall J, Byrne R, Yvan‐Charvet L, McGillicuddy FC. Metabolic Inflammation in Obesity—At the Crossroads between Fatty Acid and Cholesterol Metabolism. Mol Nutr Food Res 2020; 65:e1900482. [DOI: 10.1002/mnfr.201900482] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 06/16/2020] [Indexed: 12/11/2022]
Affiliation(s)
- Sean Curley
- Cardiometabolic Research Group UCD Diabetes Complications Research Centre UCD Conway Institute UCD School of Medicine University College Dublin Dublin 4 Ireland
| | - Julie Gall
- University of Nice Unité Mixte de Recherce (UMR) Institut National de la Santé et de la Recherche Médicale U1065 062104 Nice Cedex 3 France
| | - Rachel Byrne
- Cardiometabolic Research Group UCD Diabetes Complications Research Centre UCD Conway Institute UCD School of Medicine University College Dublin Dublin 4 Ireland
| | - Laurent Yvan‐Charvet
- University of Nice Unité Mixte de Recherce (UMR) Institut National de la Santé et de la Recherche Médicale U1065 062104 Nice Cedex 3 France
| | - Fiona C. McGillicuddy
- Cardiometabolic Research Group UCD Diabetes Complications Research Centre UCD Conway Institute UCD School of Medicine University College Dublin Dublin 4 Ireland
| |
Collapse
|
11
|
Zhang T, Chen J, Tang X, Luo Q, Xu D, Yu B. Interaction between adipocytes and high-density lipoprotein:new insights into the mechanism of obesity-induced dyslipidemia and atherosclerosis. Lipids Health Dis 2019; 18:223. [PMID: 31842884 PMCID: PMC6913018 DOI: 10.1186/s12944-019-1170-9] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Accepted: 12/09/2019] [Indexed: 12/19/2022] Open
Abstract
Obesity is the most common nutritional disorder worldwide and is associated with dyslipidemia and atherosclerotic cardiovascular disease. The hallmark of dyslipidemia in obesity is low high density lipoprotein (HDL) cholesterol (HDL-C) levels. Moreover, the quality of HDL is also changed in the obese setting. However, there are still some disputes on the explanations for this phenomenon. There is increasing evidence that adipose tissue, as an energy storage tissue, participates in several metabolism activities, such as hormone secretion and cholesterol efflux. It can influence overall reverse cholesterol transport and plasma HDL-C level. In obesity individuals, the changes in morphology and function of adipose tissue affect plasma HDL-C levels and HDL function, thus, adipose tissue should be the main target for the treatment of HDL metabolism in obesity. In this review, we will summarize the cross-talk between adipocytes and HDL related to cardiovascular disease and focus on the new insights of the potential mechanism underlying obesity and HDL dysfunction.
Collapse
Affiliation(s)
- Tianhua Zhang
- Department of Cardiovascular Medicine, the Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, People's Republic of China
| | - Jin Chen
- Department of Cardiovascular Medicine, the Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, People's Republic of China
| | - Xiaoyu Tang
- Department of Cardiovascular Medicine, the Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, People's Republic of China
| | - Qin Luo
- Department of Cardiovascular Medicine, the Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, People's Republic of China
| | - Danyan Xu
- Department of Cardiovascular Medicine, the Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, People's Republic of China
| | - Bilian Yu
- Department of Cardiovascular Medicine, the Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, People's Republic of China.
| |
Collapse
|
12
|
Wang W, Yan Z, Hu J, Shen WJ, Azhar S, Kraemer FB. Scavenger receptor class B, type 1 facilitates cellular fatty acid uptake. Biochim Biophys Acta Mol Cell Biol Lipids 2019; 1865:158554. [PMID: 31678516 DOI: 10.1016/j.bbalip.2019.158554] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 09/21/2019] [Accepted: 09/25/2019] [Indexed: 02/07/2023]
Abstract
SR-B1 belongs to the class B scavenger receptor, or CD36 super family. SR-B1 and CD36 share an affinity for a wide array of ligands. Although they exhibit similar ligand binding specificity, SR-B1 and CD36 have some very specific lipid transport functions. Whereas SR-B1 primarily facilitates the selective delivery of cholesteryl esters (CEs) and cholesterol from HDL particles to the liver and non-placental steroidogenic tissues, as well as participating in cholesterol efflux from cells, CD36 primarily mediates the uptake of long-chain fatty acids in high fatty acid-requiring organs such as the heart, skeletal muscle and adipose tissue. However, CD36 also mediates cholesterol efflux and facilitates selective lipoprotein-CE delivery, although less efficiently than SR-B1. Interestingly, the ability or efficiency of SR-B1 to mediate fatty acid uptake has not been reported. In this paper, using overexpression and siRNA-mediated knockdown of SR-B1, we show that SR-B1 possesses the ability to facilitate fatty acid uptake. Moreover, this function is not blocked by BLT-1, a specific chemical inhibitor of HDL-CE uptake activity of SR-B1, nor by sulfo-N-succinimidyl oleate, which inhibits fatty acid uptake by CD36. Attenuated fatty acid uptake was also observed in primary adipocytes isolated from SR-B1 knockout mice. In conclusion, facilitation of fatty acid uptake is an additional function that is mediated by SR-B1.
Collapse
Affiliation(s)
- Wei Wang
- Division of Endocrinology, Gerontology and Metabolism, Stanford University, Stanford, CA 94305, United States of America; Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304, United States of America; Department of Endocrinology, Peking University First Hospital, Beijing, China
| | - Zhe Yan
- Division of Endocrinology, Gerontology and Metabolism, Stanford University, Stanford, CA 94305, United States of America; Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304, United States of America; Department of Endocrinology and Metabolism, West China Hospital of Sichuan University, Chengdu, China
| | - Jie Hu
- Division of Endocrinology, Gerontology and Metabolism, Stanford University, Stanford, CA 94305, United States of America; Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304, United States of America
| | - Wen-Jun Shen
- Division of Endocrinology, Gerontology and Metabolism, Stanford University, Stanford, CA 94305, United States of America; Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304, United States of America.
| | - Salman Azhar
- Division of Endocrinology, Gerontology and Metabolism, Stanford University, Stanford, CA 94305, United States of America; Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304, United States of America
| | - Fredric B Kraemer
- Division of Endocrinology, Gerontology and Metabolism, Stanford University, Stanford, CA 94305, United States of America; Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304, United States of America.
| |
Collapse
|
13
|
Liu X, Long X, Gao Y, Liu W, Hayashi T, Mizuno K, Hattori S, Fujisaki H, Ogura T, Onodera S, Wang DO, Ikejima T. Type I collagen inhibits adipogenic differentiation via YAP activation in vitro. J Cell Physiol 2019; 235:1821-1837. [PMID: 31432516 DOI: 10.1002/jcp.29100] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 06/27/2019] [Indexed: 12/18/2022]
Abstract
Extracellular matrix (ECM) has a marked influence on adipose tissue development. Adipose tissue formation is initiated with proliferation of preadipocytes and migration before undergoing further differentiation into mature adipocytes. Previous studies showed that collagen I (col I) provides a good substratum for 3T3-L1 preadipocytes to grow and migrate. However, it remains unclear whether and how col I regulates adipogenic differentiation of preadipocytes. This study reports that lipid accumulation, representing in vitro adipogenesis of the 3T3-L1 preadipocytes or the mouse primary adipocyte precursor cells derived from subcutaneous adipose tissue in the inguinal region is inhibited by the culture on col I, owing to downregulation of adipogenic factors. Previous study shows that col I enhances 3T3-L1 cell migration via stimulating the nuclear translocation of yes-associated protein (YAP). In this study, we report that downregulation of YAP is associated with in vitro adipogenesis of preadipocytes as well as with in vivo adipose tissue of high-fat diet fed mice. Increased expression of YAP in the cells cultured on col I-coated dishes is correlated with repression of adipogenic differentiation processes. The inactivation of YAP using YAP inhibitor, verteporfin, or YAP small-interfering RNA enhanced adipogenic differentiation and reversed the inhibitory effect of col I. Activation of YAP either by the transfection of YAP plasmid or the silence of large tumor suppressor 1 (LATS1), an inhibitory kinase of YAP, inhibited adipogenic differentiation. The results indicate that col I inhibits adipogenic differentiation via YAP activation in vitro.
Collapse
Affiliation(s)
- Xiaoling Liu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China
| | - Xinyu Long
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China
| | - Yanfang Gao
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China
| | - Weiwei Liu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China
| | - Toshihiko Hayashi
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China.,Department of Chemistry and Life Science, School of Advan ced Engineering, Kogakuin University, Nakanomachi, Hachioji, Tokyo, Japan
| | | | | | | | | | - Satoshi Onodera
- Medical Research Institute of Curing Mibyo, Narusedai, Machida, Tokyo, Japan
| | - Dan Ohtan Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China
| | - Takashi Ikejima
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China.,Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| |
Collapse
|
14
|
Guo X, Liao J, Huang X, Wang Y, Huang W, Liu G. Reversal of adipose tissue loss by probucol in mice with deficiency of both scavenger receptor class B type 1 and LDL receptor on high fat diet. Biochem Biophys Res Commun 2018; 497:930-936. [PMID: 28522295 DOI: 10.1016/j.bbrc.2017.05.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 05/03/2017] [Indexed: 01/01/2023]
Abstract
Scavenger receptor class B type 1(SR-B1) and low density lipoprotein receptor (LDLR) play vital roles in cholesterol homeostasis. Previous studies indicated a strong link between cholesterol and adipose tissue (AT). In this study, adult male SR-B1 and LDLR double knockout (DKO) mice were fed with high fat diet (HFD) for 3 months. Interestingly, we found severe loss of AT in DKO mice fed with HFD. To reverse the AT loss in DKO mice, 1% probucol was added in HFD. In DKO mice on HFD, plasma total cholesterol (TC) and free cholesterol (FC) levels were increased 6 and 15 folds respectively compared with wild type (WT) mice. We found severe loss of AT in whole body of DKO mice compared with WT or single KO mice. In AT of DKO mice, histology showed the very small size of adipocytes and infiltration of inflammatory cells; Genes expressions related to fatty acid uptake, lipogenesis and adipogenesis were decreased; TUNEL analysis and related genes expressions of endoplasmic reticulum (ER) stress and inflammation were significantly higher than those of WT or single KO mice. Probucol could reduce increased TC and FC levels, and reverse the loss of fat and apoptosis of AT in DKO mice. AT loss in DKO mice with HFD was probably due to high levels of FC which led to apoptosis induced by ER stress and inflammation of AT. This study provided a novel utility of probucol in rescue of fat loss in DKO mice.
Collapse
Affiliation(s)
- Xin Guo
- Institute of Cardiovascular Sciences and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Peking University Health Science Center, Beijing, 100191, China
| | - Jiawei Liao
- Institute of Cardiovascular Sciences and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Peking University Health Science Center, Beijing, 100191, China
| | - Xiaomin Huang
- Institute of Cardiovascular Sciences and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Peking University Health Science Center, Beijing, 100191, China
| | - Yuhui Wang
- Institute of Cardiovascular Sciences and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Peking University Health Science Center, Beijing, 100191, China
| | - Wei Huang
- Institute of Cardiovascular Sciences and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Peking University Health Science Center, Beijing, 100191, China.
| | - George Liu
- Institute of Cardiovascular Sciences and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Peking University Health Science Center, Beijing, 100191, China.
| |
Collapse
|
15
|
Morel E, Ghezzal S, Lucchi G, Truntzer C, Pais de Barros JP, Simon-Plas F, Demignot S, Mineo C, Shaul PW, Leturque A, Rousset M, Carrière V. Cholesterol trafficking and raft-like membrane domain composition mediate scavenger receptor class B type 1-dependent lipid sensing in intestinal epithelial cells. Biochim Biophys Acta Mol Cell Biol Lipids 2017; 1863:199-211. [PMID: 29196159 DOI: 10.1016/j.bbalip.2017.11.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 11/13/2017] [Accepted: 11/27/2017] [Indexed: 02/02/2023]
Abstract
Scavenger receptor Class B type 1 (SR-B1) is a lipid transporter and sensor. In intestinal epithelial cells, SR-B1-dependent lipid sensing is associated with SR-B1 recruitment in raft-like/ detergent-resistant membrane domains and interaction of its C-terminal transmembrane domain with plasma membrane cholesterol. To clarify the initiating events occurring during lipid sensing by SR-B1, we analyzed cholesterol trafficking and raft-like domain composition in intestinal epithelial cells expressing wild-type SR-B1 or the mutated form SR-B1-Q445A, defective in membrane cholesterol binding and signal initiation. These features of SR-B1 were found to influence both apical cholesterol efflux and intracellular cholesterol trafficking from plasma membrane to lipid droplets, and the lipid composition of raft-like domains. Lipidomic analysis revealed likely participation of d18:0/16:0 sphingomyelin and 16:0/0:0 lysophosphatidylethanolamine in lipid sensing by SR-B1. Proteomic analysis identified proteins, whose abundance changed in raft-like domains during lipid sensing, and these included molecules linked to lipid raft dynamics and signal transduction. These findings provide new insights into the role of SR-B1 in cellular cholesterol homeostasis and suggest molecular links between SR-B1-dependent lipid sensing and cell cholesterol and lipid droplet dynamics.
Collapse
Affiliation(s)
- Etienne Morel
- Centre de Recherche des Cordeliers, INSERM, UMPC Université Paris 6, Université Paris Descartes Paris 5, CNRS, F-75006 Paris, France
| | - Sara Ghezzal
- Centre de Recherche des Cordeliers, INSERM, UMPC Université Paris 6, Université Paris Descartes Paris 5, CNRS, F-75006 Paris, France
| | - Géraldine Lucchi
- Clinical Innovation Proteomic Platform CLIPP, Université Bourgogne Franche-Comté, F-21000 Dijon, France
| | - Caroline Truntzer
- Clinical Innovation Proteomic Platform CLIPP, Université Bourgogne Franche-Comté, F-21000 Dijon, France
| | - Jean-Paul Pais de Barros
- Plateforme de Lipidomique, INSERM UMR1231, Université de Bourgogne Franche Comté, F-21000 Dijon, France
| | - Françoise Simon-Plas
- Agroécologie, AgroSup Dijon, CNRS, INRA, Université Bourgogne Franche-Comté, F-21000 Dijon, France
| | - Sylvie Demignot
- Centre de Recherche des Cordeliers, INSERM, UMPC Université Paris 6, Université Paris Descartes Paris 5, CNRS, F-75006 Paris, France; EPHE, PSL Research University, F-75006 Paris, France
| | - Chieko Mineo
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas, Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Philip W Shaul
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas, Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Armelle Leturque
- Centre de Recherche des Cordeliers, INSERM, UMPC Université Paris 6, Université Paris Descartes Paris 5, CNRS, F-75006 Paris, France
| | - Monique Rousset
- Centre de Recherche des Cordeliers, INSERM, UMPC Université Paris 6, Université Paris Descartes Paris 5, CNRS, F-75006 Paris, France
| | - Véronique Carrière
- Centre de Recherche des Cordeliers, INSERM, UMPC Université Paris 6, Université Paris Descartes Paris 5, CNRS, F-75006 Paris, France.
| |
Collapse
|
16
|
Abstract
PURPOSE OF REVIEW Scavenger receptor BI (SR-BI) is classically known for its role in antiatherogenic reverse cholesterol transport as it selectively takes up cholesterol esters from HDL. Here, we have highlighted recent literature that describes novel functions for SR-BI in physiology and disease. RECENT FINDINGS A large population-based study has revealed that patients heterozygous for the P376L mutant form of SR-BI showed significantly increased levels of plasma HDL-cholesterol and had increased risk of cardiovascular disease, demonstrating that SR-BI in humans is a significant determinant of cardiovascular disease. Furthermore, SR-BI has been shown to modulate the susceptibility to LPS-induced tissue injury and the ability of sphingosine 1 phosphate to interact with its receptor, linking SR-BI to the regulation of inflammation. In addition, important domains within the molecule (Trp-415) as well as novel regulators (procollagen C-endopeptidase enhancer protein 2) of SR-BI's selective uptake function have recently been identified. Moreover, relatively high expression levels of the SR-BI protein have been observed in a variety of cancer tissues, which is associated with a reduced overall survival rate. SUMMARY The HDL receptor SR-BI is a potential therapeutic target not only in the cardiovascular disease setting, but also in inflammatory conditions as well as in cancer.
Collapse
Affiliation(s)
- Menno Hoekstra
- Division of Biopharmaceutics, Cluster BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden, The Netherlands, , Tel: +31-71-5276582
| | - Mary Sorci-Thomas
- Division of Endocrinology, Associate in Pharmacology and Toxicology, Medical College of Wisconsin, Senior Adjunct Investigator at the Blood Research Institute, Blood Center of Wisconsin, , Tel: 414-955-5728
| |
Collapse
|
17
|
Hoekstra M. SR-BI as target in atherosclerosis and cardiovascular disease - A comprehensive appraisal of the cellular functions of SR-BI in physiology and disease. Atherosclerosis 2017; 258:153-161. [DOI: 10.1016/j.atherosclerosis.2017.01.034] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 01/25/2017] [Accepted: 01/27/2017] [Indexed: 12/12/2022]
|
18
|
Lindahl M, Petrlova J, Dalla-Riva J, Wasserstrom S, Rippe C, Domingo-Espin J, Kotowska D, Krupinska E, Berggreen C, Jones HA, Swärd K, Lagerstedt JO, Göransson O, Stenkula KG. ApoA-I Milano stimulates lipolysis in adipose cells independently of cAMP/PKA activation. J Lipid Res 2015; 56:2248-59. [PMID: 26504176 DOI: 10.1194/jlr.m054767] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Indexed: 11/20/2022] Open
Abstract
ApoA-I, the main protein component of HDL, is suggested to be involved in metabolic homeostasis. We examined the effects of Milano, a naturally occurring ApoA-I variant, about which little mechanistic information is available. Remarkably, high-fat-fed mice treated with Milano displayed a rapid weight loss greater than ApoA-I WT treated mice, and a significantly reduced adipose tissue mass, without an inflammatory response. Further, lipolysis in adipose cells isolated from mice treated with either WT or Milano was increased. In primary rat adipose cells, Milano stimulated cholesterol efflux and increased glycerol release, independently of β-adrenergic stimulation and phosphorylation of hormone sensitive lipase (Ser563) and perilipin (Ser522). Stimulation with Milano had a significantly greater effect on glycerol release compared with WT but similar effect on cholesterol efflux. Pharmacological inhibition or siRNA silencing of ABCA1 did not diminish Milano-stimulated lipolysis, although binding to the cell surface was decreased, as analyzed by fluorescence microscopy. Interestingly, methyl-β-cyclodextrin, a well-described cholesterol acceptor, dose-dependently stimulated lipolysis. Together, these results suggest that decreased fat mass and increased lipolysis following Milano treatment in vivo is partly explained by a novel mechanism at the adipose cell level comprising stimulation of lipolysis independently of the canonical cAMP/protein kinase A signaling pathway.
Collapse
Affiliation(s)
- Maria Lindahl
- Medical Protein Science, Lund University, 221 84 Lund, Sweden Glucose Transport and Protein Trafficking, Lund University, 221 84 Lund, Sweden
| | - Jitka Petrlova
- Medical Protein Science, Lund University, 221 84 Lund, Sweden
| | | | | | - Catarina Rippe
- Cellular Biomechanics, Lund University, 221 84 Lund, Sweden
| | | | - Dorota Kotowska
- Glucose Transport and Protein Trafficking, Lund University, 221 84 Lund, Sweden
| | - Ewa Krupinska
- Medical Protein Science, Lund University, 221 84 Lund, Sweden
| | | | - Helena A Jones
- Molecular Endocrinology, Department of Experimental Medical Science, Biomedical Center, Lund University, 221 84 Lund, Sweden
| | - Karl Swärd
- Cellular Biomechanics, Lund University, 221 84 Lund, Sweden
| | | | - Olga Göransson
- Protein Phosphorylation, Lund University, 221 84 Lund, Sweden
| | - Karin G Stenkula
- Glucose Transport and Protein Trafficking, Lund University, 221 84 Lund, Sweden
| |
Collapse
|
19
|
Murphy AJ, Yvan-Charvet L. Adipose modulation of ABCG1 uncovers an intimate link between sphingomyelin and triglyceride storage. Diabetes 2015; 64:689-92. [PMID: 25713191 DOI: 10.2337/db14-1553] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Andrew J Murphy
- Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | - Laurent Yvan-Charvet
- Institut National de la Santé et de la Recherche Médicale U1065, Centre Mediterranéen de Médecine Moléculaire, Nice, France
| |
Collapse
|
20
|
Gonçalves P, Araújo JR, Martel F. Antipsychotics-induced metabolic alterations: focus on adipose tissue and molecular mechanisms. Eur Neuropsychopharmacol 2015; 25:1-16. [PMID: 25523882 DOI: 10.1016/j.euroneuro.2014.11.008] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Revised: 10/14/2014] [Accepted: 11/13/2014] [Indexed: 12/19/2022]
Abstract
The use of antipsychotic drugs for the treatment of mood disorders and psychosis has increased dramatically over the last decade. Despite its consumption being associated with beneficial neuropsychiatric effects in patients, atypical antipsychotics (which are the most frequently prescribed antipsychotics) use is accompanied by some secondary adverse metabolic effects such as weight gain, dyslipidemia and glucose intolerance. The molecular mechanisms underlying these adverse effects are not fully understood but have been suggested to involve a dysregulation of adipose tissue homeostasis. As such, the aim of this paper is to review and discuss the role of adipose tissue in the development of secondary adverse metabolic effects induced by atypical antipsychotics. Data analyzed in this article suggest that atypical antipsychotics may increase adipose tissue (particularly visceral adipose tissue) lipogenesis, differentiation/hyperplasia, pro-inflammatory mediator secretion and insulin resistance and decrease adipose tissue lipolysis. Consequently, patients receiving antipsychotic medication could be at risk of developing obesity, type 2 diabetes and cardiovascular disease. A better knowledge of the impact of these drugs on adipose tissue homeostasis may unveil strategies to develop novel antipsychotic drugs with less adverse metabolic effects and to develop adjuvant therapies (e.g. behavioral and nutritional therapies) to neuropsychiatric patients receiving antipsychotic medication.
Collapse
Affiliation(s)
- Pedro Gonçalves
- INSERM (French Institute of Health and Medical Research), Unit 1151, INEM (Research Center in Molecular Medicine), Faculty of Medicine of Paris Descartes University, Paris, France
| | - João Ricardo Araújo
- INSERM (French Institute of Health and Medical Research), Unit 786, Molecular Microbial Pathogenesis Unit, Institut Pasteur, Paris, France
| | - Fátima Martel
- Department of Biochemistry (U38-FCT), Faculty of Medicine, University of Porto, Porto, Portugal.
| |
Collapse
|
21
|
Ecto-F1-ATPase/P2Y pathways in metabolic and vascular functions of high density lipoproteins. Atherosclerosis 2015; 238:89-100. [DOI: 10.1016/j.atherosclerosis.2014.11.017] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Revised: 11/12/2014] [Accepted: 11/13/2014] [Indexed: 12/15/2022]
|
22
|
Martineau C, Kevorkova O, Brissette L, Moreau R. Scavenger receptor class B, type I (Scarb1) deficiency promotes osteoblastogenesis but stunts terminal osteocyte differentiation. Physiol Rep 2014; 2:2/10/e12117. [PMID: 25281615 PMCID: PMC4254088 DOI: 10.14814/phy2.12117] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Scavenger receptor class B type I (SR-BI), the Scarb1 gene product, is a high-density lipoprotein (HDL) receptor which was shown to influence bone metabolism. Its absence in mice is associated with alterations of the glucocorticoid/adrenocorticotropic hormone axis, and translated in high bone mass and enhanced bone formation. Since the cellular alterations underlying the enhanced bone formation remain unknown, we investigated Scarb1-deficient marrow stromal cells (MSC) behavior in vitro. No difference in HDL3, cholesteryl ester (CE) or estradiol (E) association/binding was measured between Scarb1-null and wild-type (WT) cells. Scarb1 genic expression was down-regulated twofold following osteogenic treatment. Neither WT nor null cell proliferation was influenced by HDL3 exposure whereas this condition decreased genic expression of osteoblastic marker osterix (Sp7), and osteocyte markers sclerostin (Sost) and dentin matrix protein 1 (Dmp1) independently of genotype. Sost and Dmp1 basal expression in null cells was 40% and 50% that of WT cells; accordingly, osteocyte density was 20% lower in vertebrae from Scarb1-null mice. Genic expression of co-receptors for Wnt signaling, namely LDL-related protein (Lrp) 5 and Lrp8, was increased, respectively, by two- and threefold, and of transcription target-genes axis inhibition protein 2 (Axin2) and lymphoid enhancer-binding factor 1 (Lef1) over threefold. Gene expression of Wnt signaling agonist Wnt5a and of the antagonist dickkopfs-related protein 1 (Dkk1) were found to be increased 10- to 20-fold in null MSC. These data suggest alterations of Wnt pathways in Scarb1-deficient MSC potentially explaining their enhanced function, hence contributing to the high bone mass observed in these mice.
Collapse
Affiliation(s)
- Corine Martineau
- Laboratoire du Métabolisme Osseux, BioMed, Département des Sciences Biologiques, Université du Québec à Montréal, Montréal, H3C 3P8, Quebec, Canada
| | - Olha Kevorkova
- Laboratoire du Métabolisme Osseux, BioMed, Département des Sciences Biologiques, Université du Québec à Montréal, Montréal, H3C 3P8, Quebec, Canada
| | - Louise Brissette
- Laboratoire du Métabolisme des Lipoprotéines, Département des Sciences Biologiques, BioMed, Université du Québec à Montréal, Montréal, H3C 3P8, Quebec, Canada
| | - Robert Moreau
- Laboratoire du Métabolisme Osseux, BioMed, Département des Sciences Biologiques, Université du Québec à Montréal, Montréal, H3C 3P8, Quebec, Canada
| |
Collapse
|
23
|
Aguilar D, Fernandez ML. Hypercholesterolemia induces adipose dysfunction in conditions of obesity and nonobesity. Adv Nutr 2014; 5:497-502. [PMID: 25469381 PMCID: PMC4188221 DOI: 10.3945/an.114.005934] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
It is well known that hypercholesterolemia can lead to atherosclerosis and coronary heart disease. Adipose tissue represents an active endocrine and metabolic site, which might be involved in the development of chronic disease. Because adipose tissue is a key site for cholesterol metabolism and the presence of hypercholesterolemia has been shown to induce adipocyte cholesterol overload, it is critical to investigate the role of hypercholesterolemia on normal adipose function. Studies in preadipocytes revealed that cholesterol accumulation can impair adipocyte differentiation and maturation by affecting multiple transcription factors. Hypercholesterolemia has been observed to cause adipocyte hypertrophy, adipose tissue inflammation, and disruption of endocrine function in animal studies. Moreover, these effects can also be observed in obesity-independent conditions as confirmed by clinical trials. In humans, hypercholesterolemia disrupts adipose hormone secretion of visfatin, leptin, and adiponectin, adipokines that play a central role in numerous metabolic pathways and regulate basic physiologic responses such as appetite and satiety. Remarkably, treatment with cholesterol-lowering drugs has been shown to restore adipose tissue endocrine function. In this review the role of hypercholesterolemia on adipose tissue differentiation and maturation, as well as on hormone secretion and physiologic outcomes, in obesity and non–obesity conditions is presented.
Collapse
|
24
|
Qiu Y, Liu S, Chen HT, Yu CH, Teng XD, Yao HT, Xu GQ. Upregulation of caveolin-1 and SR-B1 in mice with non-alcoholic fatty liver disease. Hepatobiliary Pancreat Dis Int 2013; 12:630-636. [PMID: 24322749 DOI: 10.1016/s1499-3872(13)60099-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) is one of the most frequent causes of liver diseases, with markedly increased prevalence. However, its mechanisms are not clear. The present study was undertaken to illustrate the role of caveolin-1 (cav1) and the scavenger receptor class B type 1 (SR-B1) in NAFLD. METHODS Adult male C57BL/6 mice were fed with a normal diet or high fat and cholesterol (HFC) diet for 14 weeks. The mice were sacrificed to collect plasma and harvest the liver; their plasma lipid concentration was measured. Hepatic cav1 and SR-B1 mRNA and protein expression were determined by real-time quantitative polymerase chain reaction (qPCR) and Western blotting, respectively. In order to study cav1 and SR-B1 distribution and change in hepatocytes, immunohistochemical analysis was performed. RESULTS HFC diet increased plasma lipids, induced NAFLD and increased the liver/body weight ratio. Compared to the control mice (n=6), the mRNA and protein levels of cav1 and SR-B1 in liver tissue of the NAFLD mice (n=12) increased significantly (cav1 mRNA: 1.536+/-0.226 vs 0.980+/-0.272, P<0.05; protein: 0.643+/-0.240 vs 0.100+/-0.130, P<0.01; SR-B1 mRNA: 1.377+/-0.125 vs 0.956+/-0.151, P<0.01; protein: 2.156+/-0.507 vs 0.211+/-0.211, P<0.01). Furthermore, both cav1 and SR-B1 immunoreactivity increased and their distribution was also changed, mainly in the plasma membrane of hepatocytes, cytoplasm and membrane of lipid droplets and around. CONCLUSION NAFLD is associated with increased concentration of plasma lipids and upregulation of hepatic cav1 and SR-B1 gene and protein expressions, which indicate that cav1 and SR-B1 might play crucial roles in the pathogenesis of NAFLD.
Collapse
Affiliation(s)
- Yan Qiu
- Department of Gastroenterology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China.
| | | | | | | | | | | | | |
Collapse
|
25
|
Marcus Y, Shefer G, Stern N. Adipose tissue renin-angiotensin-aldosterone system (RAAS) and progression of insulin resistance. Mol Cell Endocrinol 2013; 378:1-14. [PMID: 22750719 DOI: 10.1016/j.mce.2012.06.021] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2012] [Accepted: 06/24/2012] [Indexed: 12/31/2022]
Abstract
This review focuses on the expression of the key components of the renin-angiotensin-aldosterone axis in fat tissue. At the center of this report is the role of RAAS in normal and excessive fat mass enlargement, the leading etiology of insulin resistance. Understanding the expression and regulation of RAAS components in various fat depots allows insight not only into the processes by which these complex patterns are modified by the enlargement of adipose tissue, but also into their impact on local and systemic response to insulin.
Collapse
Affiliation(s)
- Yonit Marcus
- Institute of Endocrinology, Metabolism and Hypertension, Tel Aviv Sourasky Medical Center, Tel Aviv 64239, Israel
| | | | | |
Collapse
|
26
|
Röhrl C, Stangl H. HDL endocytosis and resecretion. Biochim Biophys Acta Mol Cell Biol Lipids 2013; 1831:1626-33. [PMID: 23939397 PMCID: PMC3795453 DOI: 10.1016/j.bbalip.2013.07.014] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Revised: 07/22/2013] [Accepted: 07/26/2013] [Indexed: 12/23/2022]
Abstract
HDL removes excess cholesterol from peripheral tissues and delivers it to the liver and steroidogenic tissues via selective lipid uptake without catabolism of the HDL particle itself. In addition, endocytosis of HDL holo-particles has been debated for nearly 40years. However, neither the connection between HDL endocytosis and selective lipid uptake, nor the physiological relevance of HDL uptake has been delineated clearly. This review will focus on HDL endocytosis and resecretion and its relation to cholesterol transfer. We will discuss the role of HDL endocytosis in maintaining cholesterol homeostasis in tissues and cell types involved in atherosclerosis, focusing on liver, macrophages and endothelium. We will critically summarize the current knowledge on the receptors mediating HDL endocytosis including SR-BI, F1-ATPase and CD36 and on intracellular HDL transport routes. Dependent on the tissue, HDL is either resecreted (retro-endocytosis) or degraded after endocytosis. Finally, findings on HDL transcytosis across the endothelial barrier will be summarized. We suggest that HDL endocytosis and resecretion is a rather redundant pathway under physiologic conditions. In case of disturbed lipid metabolism, however, HDL retro-endocytosis represents an alternative pathway that enables tissues to maintain cellular cholesterol homeostasis.
Collapse
Affiliation(s)
- Clemens Röhrl
- Department of Medical Chemistry, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Vienna, Austria
| | - Herbert Stangl
- Department of Medical Chemistry, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
27
|
Li H, Li H, Guo H, Liu F. Cholesterol suppresses adipocytic differentiation of mouse adipose-derived stromal cells via PPARγ2 signaling. Steroids 2013; 78:454-61. [PMID: 23454216 DOI: 10.1016/j.steroids.2013.02.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2012] [Revised: 01/24/2013] [Accepted: 02/04/2013] [Indexed: 12/14/2022]
Abstract
The effects of cholesterol on cell proliferation and adipocytic differentiation have been evaluated for the first time in mouse adipose-derived stromal cells (mASCs). Cholesterol loading by using cholesterol:methyl- β-cyclodextrin (Chol:MβCD) complexes promoted cellular levels of free cholesterol (FC) and cholesteryl ester (CE), induced high cell proliferation of mASCs dose-dependently. Compared with control cells, cholesterol-treated mASCs showed an impaired differentiation process in both dose- and time-dependent manners, based on reduced oil red O-stained lipid droplets, SREBP-1, PPARγ2 and aP2 expression levels. The involvement of SREBP-1-mediated PPARγ2 in the effects of cholesterol on mASC adipogenesis was elucidated. These results point to cholesterol as a modulator of adipogenesis, which separate cholesterol itself from other components of modified lipoproteins.
Collapse
Affiliation(s)
- Haifang Li
- College of Life Sciences, Shandong Agricultural University, Tai'an 271018, China.
| | | | | | | |
Collapse
|
28
|
Jeyakumar S, Yasmeen R, Reichert B, Ziouzenkova O. Metabolism of Vitamin A in White Adipose Tissue and Obesity. OXIDATIVE STRESS AND DISEASE 2013. [DOI: 10.1201/b14569-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
29
|
Kotani K, Sakane N, Taniguchi N. Association between angiotensin II Type 2 receptor gene A/C3123 polymorphism and high-density lipoprotein cholesterol with hypertension in asymptomatic women. Med Princ Pract 2013; 22:65-9. [PMID: 22869520 PMCID: PMC5586706 DOI: 10.1159/000339892] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2012] [Accepted: 06/04/2012] [Indexed: 12/05/2022] Open
Abstract
OBJECTIVE The present study investigated the association between the angiotensin II type 2 receptor (AT2R) gene adenine/cytosine (A/C)-3123 polymorphism and cardiometabolic variables in subjects with and without hypertension. METHODS Cardiometabolic variables, in addition to genotyping by an allele-specific DNA assay, were measured in 161 asymptomatic community-dwelling Japanese women (age range 30-83 years). They were divided into hypertensive (n = 82, age 50-81 years) and nonhypertensive (n = 79, age 30-83 years) subjects. RESULTS The A-allele carriers (n = 53) showed significantly lower high-density lipoprotein cholesterol (HDL-C) levels than the non-A-allele carriers (n = 26) among nonhypertensive subjects (1.45 ± 0.38 vs. 1.66 ± 0.33 mmol/l, p = 0.02). Even when multiple-adjusted analyses were performed, the HDL-C levels continued to differ significantly and independently of other variables, including the body mass index and insulin resistance index, between A-allele and non-A-allele carriers. However, this association was not observed among hypertensive subjects. CONCLUSION The present study demonstrated that A-allele carriers had significantly lower HDL-C levels than did non-A-allele carries among nonhypertensive women, while this association was not observed among hypertensive women. This indicates that the A/C3124 polymorphism may be a marker associated with HDL metabolism by hypertension. This was a small study, so further research is warranted to confirm the observed association.
Collapse
Affiliation(s)
- Kazuhiko Kotani
- Division of Preventive Medicine, Clinical Research Institute, National Hospital Organization Kyoto Medical Center, Kyoto, Japan.
| | | | | |
Collapse
|
30
|
Wang H, Peng DQ. New insights into the mechanism of low high-density lipoprotein cholesterol in obesity. Lipids Health Dis 2011; 10:176. [PMID: 21988829 PMCID: PMC3207906 DOI: 10.1186/1476-511x-10-176] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2011] [Accepted: 10/12/2011] [Indexed: 02/06/2023] Open
Abstract
Obesity, a significant risk factor for various chronic diseases, is universally related to dyslipidemia mainly represented by decreasing high-density lipoprotein cholesterol (HDL-C), which plays an indispensible role in development of cardiovascular disease (CVD). However, the mechanisms underlying obesity and low HDL-C have not been fully elucidated. Previous studies have focused on the alteration of HDL catabolism in circulation following elevated triglyceride (TG). But recent findings suggested that liver and fat tissue played pivotal role in obesity related low HDL-C. Some new molecular pathways like microRNA have also been proposed in the regulation of HDL metabolism in obesity. This article will review recent advances in understanding of the potential mechanism of low HDL-C in obesity.
Collapse
Affiliation(s)
- Hao Wang
- Departments of Cardiology, the Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, PR China
| | | |
Collapse
|
31
|
Curcumin promotes cholesterol efflux from adipocytes related to PPARgamma-LXRalpha-ABCA1 passway. Mol Cell Biochem 2011; 358:281-5. [PMID: 21748336 DOI: 10.1007/s11010-011-0978-z] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2011] [Accepted: 06/29/2011] [Indexed: 12/20/2022]
Abstract
Curcumin affects the functions of adipocytes. But it is not known whether curcumin has some effect on the cholesterol efflux process of adipocytes. Rabbit subcutaneous adipocytes were incubated with 5, 10 and 20 μg/ml curcumin for 24 h. The cholesterol efflux onto apoAI was assessed, and the peroxisome proliferators-activated receptor (PPAR) γ, liver X receptor (LXR) α and ATP-binding cassette transporter A1 (ABCA1) mRNA expression in adipocytes were quantified by reverse-transcription polymerase chain reaction (RT-PCR). Curcumin increased the cholesterol efflux from adipocytes in dose-dependent manner. The increased expression of PPARγ, LXRα and ABCA1 caused by curcumin were parallel. When the adipocytes were pre-treated by GW9662, the increased expression of PPARγ induced by curcumin was partially prevented, subsequent to the down-regulation of LXRα and ABCA1. Curcumin can affect the cholesterol efflux from adipocytes by regulating the PPARγ-LXR-ABCA1 passway.
Collapse
|
32
|
Cheng Q, Leung PS. An update on the islet renin-angiotensin system. Peptides 2011; 32:1087-95. [PMID: 21396973 DOI: 10.1016/j.peptides.2011.03.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2011] [Revised: 02/28/2011] [Accepted: 03/01/2011] [Indexed: 12/11/2022]
Abstract
The traditional renin-angiotensin system (RAS) components have been studied extensively since the rate-limiting component of RAS, renin, was first characterized. The ongoing identification of various novel RAS components and signaling pathways continues to elaborate the complexity of this system. Regulation of RAS according to the conventional and contemporary views of its functions in various tissues under pathophysiological conditions is a main treatment strategy for many metabolic diseases. The local pancreatic RAS, first proposed to exist in pancreatic islets two decades ago, could regulate islet function and glycemic control via influences on islet cell mass, inflammation, and ion channels. Insulin secretion, the major function of pancreatic islets, is controlled by numerous factors. Among these factors and of particular interest are glucagon-like peptide-1 (GLP-1) and vitamin D, which may regulate islet function by directly binding receptors on islet beta cells. These factors may work with local RAS signaling in islets to protect and maintain islet function under diabetic and hyperglycemic conditions. In this concise review, the local islet RAS will be discussed with particular attention being paid to recent notable findings.
Collapse
Affiliation(s)
- Qianni Cheng
- Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | | |
Collapse
|
33
|
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
|
34
|
Parent-offspring transmission of adipocytokine levels and their associations with metabolic traits. PLoS One 2011; 6:e18182. [PMID: 21483749 PMCID: PMC3070726 DOI: 10.1371/journal.pone.0018182] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2010] [Accepted: 02/22/2011] [Indexed: 12/17/2022] Open
Abstract
Adipose tissue secreted cytokines (adipocytokines) have significant effects on the physiology and pathology of human metabolism relevant to diabetes and cardiovascular disease. We determined the relationship of the pattern of these circulating hormones with obesity-related phenotypes and whether such pattern is transmitted from parent to offspring. A combined total of 403 individuals from 156 consenting Saudi families divided into initial (119 families with 123 adults and 131 children) and replication (37 families with 58 adults and 91 children) cohorts were randomly selected from the RIYADH Cohort study. Anthropometrics were evaluated and metabolic measures such as fasting serum glucose, lipid profiles, insulin, leptin, adiponectin, resistin, tumor necrosis factor alpha (TNFα), activated plasminogen activator inhibitor 1 (aPAI1), high sensitivity C-reactive protein (hsCRP) and angiotensin II were also assessed. Parent-offspring regressions revealed that with the exception of hsCRP, all hormones measured showed evidence for significant inheritance. Principal component (PC) analysis of standardized hormone levels demonstrated surprising heritability of the three most common axes of variation. PC1, which explained 21% of the variation, was most strongly loaded on levels of leptin, TNFα, insulin, and aPAI1, and inversely with adiponectin. It was significantly associated with body mass index (BMI) and phenotypically stronger in children, and showed a heritability of ∼50%, after adjustment for age, gender and generational effects. We conclude that adipocytokines are highly heritable and their pattern of co-variation significantly influences BMI as early as the pre-teen years. Investigation at the genomic scale is required to determine the variants affecting the regulation of the hormones studied.
Collapse
|
35
|
Seneff S, Wainwright G, Mascitelli L. Is the metabolic syndrome caused by a high fructose, and relatively low fat, low cholesterol diet? Arch Med Sci 2011; 7:8-20. [PMID: 22291727 PMCID: PMC3258689 DOI: 10.5114/aoms.2011.20598] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2010] [Revised: 08/15/2010] [Accepted: 08/23/2010] [Indexed: 01/24/2023] Open
Abstract
The metabolic syndrome (MetS) is manifested by a lipid triad which includes elevated serum triglycerides, small LDL particles, and low high-density lipoprotein (HDL) cholesterol, by central obesity (central adiposity), insulin resistance, glucose intolerance and elevated blood pressure, and it is associated with an increased risk of type 2 diabetes and coronary heart disease. We have developed a new hypothesis regarding MetS as a consequence of a high intake in carbohydrates and food with a high glycemic index, particularly fructose, and relatively low intake of cholesterol and saturated fat. We support our arguments through animal studies which have shown that exposure of the liver to increased quantities of fructose leads to rapid stimulation of lipogenesis and accumulation of triglycerides. The adipocytes store triglycerides in lipid droplets, leading to adipocyte hypertrophy. Adipocyte hypertrophy is associated with macrophage accumulation in adipose tissue. An important modulator of obesity-associated macrophage responses in white adipose tissue is the death of adipocytes. Excess exposure to fructose intake determines the liver to metabolize high doses of fructose, producing increased levels of fructose end products, like glyceraldehyde and dihydroxyacetone phosphate, that can converge with the glycolytic pathway. Fructose also leads to increased levels of advanced glycation end products. The macrophages exposed to advanced glycation end products become dysfunctional and, on entry into the artery wall, contribute to plaque formation and thrombosis.
Collapse
Affiliation(s)
- Stephanie Seneff
- Department of Electrical Engineering and Computer Science, MIT, Cambridge, MA, USA
| | | | - Luca Mascitelli
- Medical Service, Comando Brigata Alpina “Julia”, Udine, Italy
| |
Collapse
|
36
|
Toh SA, Millar JS, Billheimer J, Fuki I, Naik SU, Macphee C, Walker M, Rader DJ. PPARγ activation redirects macrophage cholesterol from fecal excretion to adipose tissue uptake in mice via SR-BI. Biochem Pharmacol 2011; 81:934-41. [PMID: 21291868 DOI: 10.1016/j.bcp.2011.01.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2010] [Revised: 01/18/2011] [Accepted: 01/20/2011] [Indexed: 01/26/2023]
Abstract
PPARγ agonists, used in the treatment of Type 2 diabetes, can raise HDL-cholesterol, therefore could potentially stimulate macrophage-to-feces reverse cholesterol transport (RCT). We aimed to test whether PPARγ activation promotes macrophage RCT in vivo. Macrophage RCT was assessed in mice using cholesterol loaded/(3)H-cholesterol labeled macrophages. PPARγ agonist GW7845 (20 mg/kg/day) did not change (3)H-tracer plasma appearance, but surprisingly decreased fecal (3)H-free sterol excretion by 43% (P<0.01) over 48h. Total free cholesterol efflux from macrophages to serum (collected from control and GW7845 groups) was not different, although ABCA1-mediated efflux was significantly higher with GW7845. To determine the effect of PPARγ activation on HDL cholesterol uptake by different tissues, the metabolic fate of HDL labeled with (3)H-cholesteryl ether (CE) was also measured. We observed two-fold increase in HDL derived (3)H-CE uptake by adipose tissue (P<0.005) with concomitant 22% decrease in HDL derived (3)H-CE uptake by the liver (P<0.05) in GW7845 treated wild type mice. This was associated with a significant increase in SR-BI protein expression in adipose tissue, but not liver. The same experiment in SR-BI knockout mice, showed no difference in HDL derived (3)H-CE uptake by adipose tissue or liver. In conclusion, PPARγ activation decreases the fecal excretion of macrophage derived cholesterol in mice. This is not due to inhibition of cholesterol efflux from macrophages, but rather involves redirection of effluxed cholesterol from liver towards adipose tissue uptake via SR-BI. This represents a novel mechanism for regulation of RCT and may extend the therapeutic implications of these ligands.
Collapse
Affiliation(s)
- Sue-Anne Toh
- Division of Endocrinology, Diabetes and Metabolism, University of Pennsylvania School of Medicine, 421 Curie Blvd., Philadelphia, PA 19104, USA.
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Yvan-Charvet L, Quignard-Boulangé A. Role of adipose tissue renin–angiotensin system in metabolic and inflammatory diseases associated with obesity. Kidney Int 2011; 79:162-8. [DOI: 10.1038/ki.2010.391] [Citation(s) in RCA: 150] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
38
|
Yu BL, Zhao SP, Hu JR. Cholesterol imbalance in adipocytes: a possible mechanism of adipocytes dysfunction in obesity. Obes Rev 2010; 11:560-7. [PMID: 20025694 DOI: 10.1111/j.1467-789x.2009.00699.x] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Studies of the past decade have increased our understanding of the role of adipose tissue dysfunction in obesity and obesity-related insulin resistance and type 2 diabetes. Although adipose tissue is the body's largest pool of free cholesterol, adipocytes have limited activity in cholesterol synthetic pathway. Thus, the majority of adipocyte cholesterol originates from circulating lipoproteins. To maintain cholesterol homeostasis, adipocytes have developed multiple pathways for cholesterol efflux. Several transcriptional factors, such as sterol regulatory element-binding proteins and liver X receptors may be responsible for the regulation of cholesterol homeostasis in adipocytes. Most notably, because altering cholesterol balance profoundly modifies adipocyte metabolism in a way resembling that seen in hypertrophied adipocytes, cholesterol imbalance is recognized as a characteristic for enlarged adipocytes per se in the obese state. In addition, plasma membrane cholesterol normalization by chromium picolinate can fully restore insulin-stimulated glucose transport, further supporting the role of the adipocyte cholesterol imbalance in obesity and insulin resistance.
Collapse
Affiliation(s)
- B-L Yu
- Department of Cardiology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | | | | |
Collapse
|
39
|
Zhang Y, McGillicuddy FC, Hinkle CC, O’Neill S, Glick JM, Rothblat GH, Reilly MP. Adipocyte modulation of high-density lipoprotein cholesterol. Circulation 2010; 121:1347-55. [PMID: 20212278 PMCID: PMC2925122 DOI: 10.1161/circulationaha.109.897330] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
BACKGROUND Adipose harbors a large depot of free cholesterol. However, a role for adipose in cholesterol lipidation of high-density lipoprotein (HDL) in vivo is not established. We present the first evidence that adipocytes support transfer of cholesterol to HDL in vivo as well as in vitro and implicate ATP-binding cassette subfamily A member 1 (ABCA1) and scavenger receptor class B type I (SR-BI), but not ATP-binding cassette subfamily G member 1 (ABCG1), cholesterol transporters in this process. METHODS AND RESULTS Cholesterol efflux from wild-type, ABCA1(-/-), SR-BI(-/-), and ABCG1(-/-) adipocytes to apolipoprotein A-I (apoA-I) and HDL3 were measured in vitro. 3T3L1 adipocytes, labeled with (3)H-cholesterol, were injected intraperitoneally into wild-type, apoA-I transgenic, and apoA-I(-/-) mice, and tracer movement onto plasma HDL was monitored. Identical studies were performed with labeled wild-type, ABCA1(-/-), or SR-BI(-/-) mouse embryonic fibroblast adipocytes. The effect of tumor necrosis factor-alpha on transporter expression and cholesterol efflux was monitored during adipocyte differentiation. Cholesterol efflux to apoA-I and HDL3 was impaired in ABCA1(-/-) and SR-BI(-/-) adipocytes, respectively, with no effect observed in ABCG1(-/-) adipocytes. Intraperitoneal injection of labeled 3T3L1 adipocytes resulted in increased HDL-associated (3)H-cholesterol in apoA-I transgenic mice but reduced levels in apoA-I(-/-) animals. Intraperitoneal injection of labeled ABCA1(-/-) or SR-BI(-/-) adipocytes reduced plasma counts relative to their respective controls. Tumor necrosis factor-alpha reduced both ABCA1 and SR-BI expression and impaired cholesterol efflux from partially differentiated adipocytes. CONCLUSIONS These data suggest a novel metabolic function of adipocytes in promoting cholesterol transfer to HDL in vivo and implicate adipocyte SR-BI and ABCA1, but not ABCG1, in this process. Furthermore, adipocyte modulation of HDL may be impaired in adipose inflammatory disease states such as type 2 diabetes mellitus.
Collapse
Affiliation(s)
- YuZhen Zhang
- Cardiovascular Institute, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
- Institute of Translational Medicine and Therapeutics, the Departments of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
| | - Fiona C. McGillicuddy
- Cardiovascular Institute, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
- Institute of Translational Medicine and Therapeutics, the Departments of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
- Nutrigenomics Research Group, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | - Christine C. Hinkle
- Cardiovascular Institute, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
- Institute of Translational Medicine and Therapeutics, the Departments of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
| | - Sean O’Neill
- Cardiovascular Institute, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
- Institute of Translational Medicine and Therapeutics, the Departments of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
| | - Jane M. Glick
- Cell and Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
| | - George H. Rothblat
- Division of Nutrition, Children's Hospital of Philadelphia, Philadelphia, PA 19104
| | - Muredach P. Reilly
- Cardiovascular Institute, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
- Institute of Translational Medicine and Therapeutics, the Departments of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
| |
Collapse
|
40
|
Béaslas O, Cueille C, Delers F, Chateau D, Chambaz J, Rousset M, Carrière V. Sensing of dietary lipids by enterocytes: a new role for SR-BI/CLA-1. PLoS One 2009; 4:e4278. [PMID: 19169357 PMCID: PMC2627924 DOI: 10.1371/journal.pone.0004278] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2008] [Accepted: 12/17/2008] [Indexed: 11/23/2022] Open
Abstract
Background The intestine is responsible for absorbing dietary lipids and delivering them to the organism as triglyceride-rich lipoproteins (TRL). It is important to determine how this process is regulated in enterocytes, the absorptive cells of the intestine, as prolonged postprandial hypertriglyceridemia is a known risk factor for atherosclerosis. During the postprandial period, dietary lipids, mostly triglycerides (TG) hydrolyzed by pancreatic enzymes, are combined with bile products and reach the apical membrane of enterocytes as postprandial micelles (PPM). Our aim was to determine whether these micelles induce, in enterocytes, specific early cell signaling events that could control the processes leading to TRL secretion. Methodology/Principal Findings The effects of supplying PPM to the apex of Caco-2/TC7 enterocytes were analyzed. Micelles devoid of TG hydrolysis products, like those present in the intestinal lumen in the interprandial period, were used as controls. The apical delivery of PPM specifically induced a number of cellular events that are not induced by interprandial micelles. These early events included the trafficking of apolipoprotein B, a structural component of TRL, from apical towards secretory domains, and the rapid, dose-dependent activation of ERK and p38MAPK. PPM supply induced the scavenger receptor SR-BI/CLA-1 to cluster at the apical brush border membrane and to move from non-raft to raft domains. Competition, inhibition or knockdown of SR-BI/CLA-1 impaired the PPM-dependent apoB trafficking and ERK activation. Conclusions/Significance These results are the first evidence that enterocytes specifically sense postprandial dietary lipid-containing micelles. SR-BI/CLA-1 is involved in this process and could be a target for further study with a view to modifying intestinal TRL secretion early in the control pathway.
Collapse
Affiliation(s)
- Olivier Béaslas
- Université Pierre et Marie Curie - Paris 6, UMR S 872, Les Cordeliers, Paris, France
- INSERM, U 872, Paris, France
- Université Paris Descartes, UMR S 872, Paris, France
| | - Carine Cueille
- Université Pierre et Marie Curie - Paris 6, UMR S 872, Les Cordeliers, Paris, France
- INSERM, U 872, Paris, France
- Université Paris Descartes, UMR S 872, Paris, France
| | - François Delers
- Université Pierre et Marie Curie - Paris 6, UMR S 872, Les Cordeliers, Paris, France
- INSERM, U 872, Paris, France
- Université Paris Descartes, UMR S 872, Paris, France
| | - Danielle Chateau
- Université Pierre et Marie Curie - Paris 6, UMR S 872, Les Cordeliers, Paris, France
- INSERM, U 872, Paris, France
- Université Paris Descartes, UMR S 872, Paris, France
| | - Jean Chambaz
- Université Pierre et Marie Curie - Paris 6, UMR S 872, Les Cordeliers, Paris, France
- INSERM, U 872, Paris, France
- Université Paris Descartes, UMR S 872, Paris, France
| | - Monique Rousset
- Université Pierre et Marie Curie - Paris 6, UMR S 872, Les Cordeliers, Paris, France
- INSERM, U 872, Paris, France
- Université Paris Descartes, UMR S 872, Paris, France
| | - Véronique Carrière
- Université Pierre et Marie Curie - Paris 6, UMR S 872, Les Cordeliers, Paris, France
- INSERM, U 872, Paris, France
- Université Paris Descartes, UMR S 872, Paris, France
- * E-mail:
| |
Collapse
|
41
|
Abstract
A local renin-angiotensin system (RAS) has been proposed in adipocytes. Adipocytes are a suggested source of components of the RAS, with regulation of their production related to obesity-hypertension. Both angiotensin type 1 and 2 receptors have been localized to adipocytes. Angiotensin II has been demonstrated to regulate adipocyte growth and differentiation, lipid metabolism, and expression and release of adipokines and RAS components, and to promote oxidative stress. Differences in regional expression of RAS components in visceral versus subcutaneous adipose tissue have been suggested as a link between abdominal obesity and cardiovascular disease. Finally, several studies support antihypertensive efficacy of RAS blockade in patients with type 2 diabetes and obesity. Future studies should address the role of adipocyte-specific deficiency of RAS components to definitively determine the relevance of the adipose RAS to normal physiology and to the development of hypertension.
Collapse
Affiliation(s)
- Lisa A Cassis
- Graduate Center for Nutritional Sciences, Wethington Building, Room 521b, 900 South Limestone Street, University of Kentucky, Lexington, KY 40536-0200, USA.
| | | | | | | |
Collapse
|
42
|
Scavenger receptor of class B expressed by osteoblastic cells are implicated in the uptake of cholesteryl ester and estradiol from LDL and HDL3. J Bone Miner Res 2008; 23:326-37. [PMID: 17967141 DOI: 10.1359/jbmr.071022] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
UNLABELLED Lipoproteins transport many vitamins and hormones that have been shown to be necessary for bone formation. However, the metabolism of LDL and HDL3 by bone-forming osteoblastic cells remains unknown. Here we report that osteoblastic cells express scavenger receptors of class B that are implicated in the uptake of cholesterol and estradiol from LDL and HDL3. INTRODUCTION The bone tissue is continuously remodeled, and its integrity requires a balance between osteoclastic bone resorption and osteoblastic bone formation. Recent studies have reported the importance of triglyceride-rich lipoproteins for the delivery of lipophilic vitamins necessary for normal bone metabolism. However, the ability of osteoblastic cells to process low- and high-density lipoproteins (LDL and HDL3) and the receptors involved remain unknown. MATERIALS AND METHODS Binding, competition, degradation, and selective uptake assays with LDL and HDL3 radiolabeled in their protein and lipid moieties or with [3H]estradiol were conducted on human osteoblasts (MG-63 cell line and primary cultures of human osteoblasts [hOB cells]) and on mouse osteoblasts (MC3T3-E1 cell line and primary cultures of murine osteoblasts [mOB cells]). The expression of scavenger receptors (SRs) by osteoblastic cells was determined by RT-PCR and Western immunoblotting, and cellular localization was assessed by sucrose gradient fractionation. RESULTS Osteoblastic cells were able to bind, internalize, and degrade HDL3 and LDL and are capable of selectively taking up cholesteryl esters (CEs) from these lipoproteins. Also, we provide evidence that osteoblastic cells express SR-BI, SR-BII, and CD36 (SR-Bs receptors) and that these receptors are localized in membrane lipid rafts or caveolin-rich membranes. The selective uptake of CE from LDL and HDL3 by osteoblastic cells was strongly inhibited by the known SR-B ligand oxidized LDL, indicating that SR-B receptors are responsible for the selective uptake. Finally, estradiol carried by LDL and HDL3 was selectively transferred to the osteoblastic cells also through SR-B receptors. CONCLUSIONS Overall, our results suggest a novel mechanism for the routing of cholesterol and estradiol to osteoblasts involving the metabolism of LDL and HDL3 by SR-B receptors.
Collapse
|
43
|
Plummer MR, Hasty AH. Atherosclerotic lesion formation and triglyceride storage in obese apolipoprotein AI-deficient mice. J Nutr Biochem 2008; 19:664-73. [PMID: 18280133 DOI: 10.1016/j.jnutbio.2007.08.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2007] [Revised: 07/19/2007] [Accepted: 08/30/2007] [Indexed: 11/18/2022]
Abstract
Obese leptin-deficient (ob/ob) mice have increased levels of high-density lipoprotein (HDL) and a unique lipoprotein referred to as low-density lipoprotein (LDL)/HDL1. When crossed onto an apolipoprotein AI (apoAI)-deficient (-/-) background, ob/ob;apoAI-/- mice accumulate LDL/HDL1 in the absence of traditional HDL. To determine the role of LDL/HDL1 in atherosclerosis, C57BL/6, apoAI-/-, ob/ob and ob/ob;apoAI-/- mice were placed on butterfat diet. After 20 weeks, all four groups had a significant increase in total cholesterol levels. The cholesterol in C57BL/6 mice was carried on very low-density lipoprotein (VLDL) and LDL and, in ob/ob and ob/ob;apoAI-/- mice, on HDL and LDL/HDL1. Atherosclerotic lesion area was similar among C57BL/6, ob/ob and ob/ob;apoAI-/- groups despite their dissimilar lipoprotein profiles. Hepatic triglyceride production and VLDL clearance rates were similar among the four groups. The ob/ob;apoAI-/- group had a significant decrease in liver weight and an increase in white adipose tissue (WAT) weight compared to the ob/ob group. Hepatic scavenger receptor class B type I (SR-BI) levels were decreased in both liver and WAT in ob/ob;apoAI-/- compared to ob/ob mice. Conclusions regarding the atherogenicity of LDL/HDL1 were confounded by the differences in lipoprotein profiles among the four groups. However, our studies provide support for the concept that apoAI and SR-BI assist in the partitioning of lipid from adipose tissue to the liver.
Collapse
Affiliation(s)
- Michelle R Plummer
- Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | | |
Collapse
|
44
|
Abstract
CONTEXT Obesity is increasing in prevalence and it is important to understand factors that regulate adipose tissue lipid metabolism. Recently, endogenous expression of apolipoprotein E (apoE) in adipose tissue has been shown to have important effects on adipocyte lipid flux and gene expression. Adipose tissue is also a physiological target of angiotensin II (AII). OBJECTIVE The aim of the current study was to evaluate a potential regulatory effect for AII on adipose tissue apoE expression. RESULTS Infusion of AII into mice for 3 d significantly reduced apoE expression in adipocytes from freshly isolated adipose tissue. ApoE expression was unchanged by the AII infusion in the stromovascular fraction. In isolated human adipocytes, treatment with AII significantly reduced cellular and secreted apoprotein E (by 20-60%). Suppression of apoE expression was observed in sc adipocytes obtained from nonobese (body mass index < 30 kg/m(2)) donors, and in sc and omental adipocytes obtained from obese (body mass index > 30 kg/m(2)) donors. Evaluation of the effect of AII in matched sets of sc and omental adipocytes from three separate donors showed lower overall apoE expression in omental adipocytes in two of the donors, and a concordant down-regulation of apoE expression in sc and omental adipocytes from all three subjects. The specific AT(1) receptor blocker, valsartan, eliminated the effect of AII on adipocyte apoE expression. CONCLUSION Both apoE and components of the renin-angiotensin system are expressed in adipose tissue, and each has important effects on adipocyte lipid metabolism and gene expression. The regulatory interaction we have identified between these two pathways has important implications for a complete understanding of adipose tissue lipid homeostasis.
Collapse
Affiliation(s)
- Poornima Rao
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612, USA
| | | | | |
Collapse
|