1
|
Cao D, Khan Z, Li X, Saito S, Bernstein EA, Victor AR, Ahmed F, Hoshi AO, Veiras LC, Shibata T, Che M, Cai L, Yamashita M, Temel RE, Giani JF, Luthringer DJ, Divakaruni AS, Okwan-Duodu D, Bernstein KE. Macrophage angiotensin-converting enzyme reduces atherosclerosis by increasing peroxisome proliferator-activated receptor α and fundamentally changing lipid metabolism. Cardiovasc Res 2023; 119:1825-1841. [PMID: 37225143 PMCID: PMC10681664 DOI: 10.1093/cvr/cvad082] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 03/21/2023] [Accepted: 04/05/2023] [Indexed: 05/26/2023] Open
Abstract
AIMS The metabolic failure of macrophages to adequately process lipid is central to the aetiology of atherosclerosis. Here, we examine the role of macrophage angiotensin-converting enzyme (ACE) in a mouse model of PCSK9-induced atherosclerosis. METHODS AND RESULTS Atherosclerosis in mice was induced with AAV-PCSK9 and a high-fat diet. Animals with increased macrophage ACE (ACE 10/10 mice) have a marked reduction in atherosclerosis vs. WT mice. Macrophages from both the aorta and peritoneum of ACE 10/10 express increased PPARα and have a profoundly altered phenotype to process lipids characterized by higher levels of the surface scavenger receptor CD36, increased uptake of lipid, increased capacity to transport long chain fatty acids into mitochondria, higher oxidative metabolism and lipid β-oxidation as determined using 13C isotope tracing, increased cell ATP, increased capacity for efferocytosis, increased concentrations of the lipid transporters ABCA1 and ABCG1, and increased cholesterol efflux. These effects are mostly independent of angiotensin II. Human THP-1 cells, when modified to express more ACE, increase expression of PPARα, increase cell ATP and acetyl-CoA, and increase cell efferocytosis. CONCLUSION Increased macrophage ACE expression enhances macrophage lipid metabolism, cholesterol efflux, efferocytosis, and it reduces atherosclerosis. This has implications for the treatment of cardiovascular disease with angiotensin II receptor antagonists vs. ACE inhibitors.
Collapse
Affiliation(s)
- DuoYao Cao
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, USA
| | - Zakir Khan
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, USA
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, USA
| | - Xiaomo Li
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, USA
| | - Suguru Saito
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, USA
| | - Ellen A Bernstein
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, USA
| | - Aaron R Victor
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, USA
| | - Faizan Ahmed
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, USA
| | - Aoi O Hoshi
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, USA
| | - Luciana C Veiras
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, USA
| | - Tomohiro Shibata
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, USA
| | - Mingtian Che
- Biobank and Pathology Shared Resource, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Lei Cai
- Saha Cardiovascular Research Center and Department of Physiology, University of Kentucky, Lexington, KY 40536, USA
| | - Michifumi Yamashita
- Saha Cardiovascular Research Center and Department of Physiology, University of Kentucky, Lexington, KY 40536, USA
| | - Ryan E Temel
- Saha Cardiovascular Research Center and Department of Physiology, University of Kentucky, Lexington, KY 40536, USA
| | - Jorge F Giani
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, USA
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, USA
| | - Daniel J Luthringer
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, USA
| | - Ajit S Divakaruni
- Department of Molecular and Medical Pharmacology, UCLA David Geffen School of Medicine, Los Angeles, CA 90095, USA
| | - Derick Okwan-Duodu
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, USA
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, USA
| | - Kenneth E Bernstein
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, USA
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, USA
| |
Collapse
|
2
|
Zhao Y, Zhang L, Liu L, Zhou X, Ding F, Yang Y, Du S, Wang H, Van Eck M, Wang J. Specific Loss of ABCA1 (ATP-Binding Cassette Transporter A1) Suppresses TCR (T-Cell Receptor) Signaling and Provides Protection Against Atherosclerosis. Arterioscler Thromb Vasc Biol 2022; 42:e311-e326. [PMID: 36252122 DOI: 10.1161/atvbaha.122.318226] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND ABCA1 (ATP-binding cassette transporter A1) mediates cholesterol efflux to apo AI to maintain cellular cholesterol homeostasis. The current study aims to investigate whether T-cell-specific deletion of ABCA1 modulates the phenotype/function of T cells and the development of atherosclerosis. METHODS Mice with T-cell-specific deletion of ABCA1 on low-density lipoprotein receptor knockout (Ldlr-/-) background (Abca1CD4-/CD4-Ldlr-/-) were generated by multiple steps of (cross)-breedings among Abca1flox/flox, CD4-Cre, and Ldlr-/- mice. RESULTS Deletions of ABCA1 greatly suppressed cholesterol efflux to apo AI but slightly reduced membrane lipid rafts on T cells probably due to the upregulation of ABCG1. Moreover, ABCA1 deficiency impaired TCR (T-cell receptor) signaling and inhibited the survival and proliferation of T cells as well as the formation of effector memory T cells. Despite the comparable levels of plasma total cholesterol after Western-type diet feeding, Abca1CD4-/CD4-Ldlr-/- mice showed significantly attenuated arterial accumulations of T cells and smaller atherosclerotic lesions than Abca1+/+Ldlr-/-controls, which were associated with reduced surface CCR5 (CC motif chemokine receptor 5) and CXCR3 (CXC motif chemokine receptor 3), decreased antiapoptotic Bcl-2 (B-cell lymphoma 2) and Bcl-xL (B-cell lymphoma extra-large), and hampered abilities to produce IL (interleukin)-2 and IFN (interferon)-γ by ABCA1-deficient T cells. CONCLUSIONS ABCA1 is essential for T-cell cholesterol homeostasis. Deletion of ABCA1 in T cells impairs TCR signaling, suppresses the survival, proliferation, differentiation, and function of T cells, thereby providing atheroprotection in vivo.
Collapse
Affiliation(s)
- Ying Zhao
- Department of Pathophysiology (Y.Z., L.Z., L.L., F.D., Y.Y., S.D.), Soochow Medical College of Soochow University, Suzhou, China
| | - Lili Zhang
- Department of Pathophysiology (Y.Z., L.Z., L.L., F.D., Y.Y., S.D.), Soochow Medical College of Soochow University, Suzhou, China
| | - Limin Liu
- Department of Pathophysiology (Y.Z., L.Z., L.L., F.D., Y.Y., S.D.), Soochow Medical College of Soochow University, Suzhou, China
| | - Xuan Zhou
- Department of Immunology (X.Z.), Soochow Medical College of Soochow University, Suzhou, China
| | - Fangfang Ding
- Department of Pathophysiology (Y.Z., L.Z., L.L., F.D., Y.Y., S.D.), Soochow Medical College of Soochow University, Suzhou, China
| | - Yan Yang
- Department of Pathophysiology (Y.Z., L.Z., L.L., F.D., Y.Y., S.D.), Soochow Medical College of Soochow University, Suzhou, China
| | - Shiyu Du
- Department of Pathophysiology (Y.Z., L.Z., L.L., F.D., Y.Y., S.D.), Soochow Medical College of Soochow University, Suzhou, China
| | - Hongmin Wang
- School of Biology & Basic Medical Sciences, and Institutes of Biology & Medical Sciences (H.W., J.W.), Soochow Medical College of Soochow University, Suzhou, China
| | - Miranda Van Eck
- Division of BioTherapeutics (M.V.E.), Leiden Academic Centre for Drug Research, Leiden University, the Netherlands.,Division of Systems Pharmacology and Pharmacy (M.V.E.), Leiden Academic Centre for Drug Research, Leiden University, the Netherlands.,Pharmacy Leiden, the Netherlands (M.V.E.)
| | - Jun Wang
- School of Biology & Basic Medical Sciences, and Institutes of Biology & Medical Sciences (H.W., J.W.), Soochow Medical College of Soochow University, Suzhou, China
| |
Collapse
|
3
|
FTY720 Reduces Lipid Accumulation by Upregulating ABCA1 through Liver X Receptor and Sphingosine Kinase 2 Signaling in Macrophages. Int J Mol Sci 2022; 23:ijms232314617. [PMID: 36498944 PMCID: PMC9740778 DOI: 10.3390/ijms232314617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 11/15/2022] [Accepted: 11/16/2022] [Indexed: 11/24/2022] Open
Abstract
Formation of foam cells as a result of excess lipid accumulation by macrophages is a pathological hallmark of atherosclerosis. Fingolimod (FTY720) is an immunosuppressive agent used in clinical settings for the treatment of multiple sclerosis and has been reported to inhibit atherosclerotic plaque development. However, little is known about the effect of FTY720 on lipid accumulation leading to foam cell formation. In this study, we investigated the effects of FTY720 on lipid accumulation in murine macrophages. FTY720 treatment reduced lipid droplet formation and increased the expression of ATP-binding cassette transporter A1 (ABCA1) in J774 mouse macrophages. FTY720 also enhanced the expression of liver X receptor (LXR) target genes such as FASN, APOE, and ABCG1. In addition, FTY720-induced upregulation of ABCA1 was abolished by knockdown of sphingosine kinase 2 (SphK2) expression. Furthermore, we found that FTY720 treatment induced histone H3 lysine 9 (H3K9) acetylation, which was lost in SphK2-knockdown cells. Taken together, FTY720 induces ABCA1 expression through SphK2-mediated acetylation of H3K9 and suppresses lipid accumulation in macrophages, which provides novel insights into the mechanisms of action of FTY720 on atherosclerosis.
Collapse
|
4
|
Kotlyarov S, Kotlyarova A. Clinical Significance of Lipid Transport Function of ABC Transporters in the Innate Immune System. MEMBRANES 2022; 12:1083. [PMID: 36363640 PMCID: PMC9698216 DOI: 10.3390/membranes12111083] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 10/25/2022] [Accepted: 10/28/2022] [Indexed: 06/16/2023]
Abstract
ABC transporters are a large family of proteins that transport a variety of substrates across cell plasma membranes. Because of this, they are involved in many physiological processes. It is of interest to note that many ABC transporters are involved in the transport of various lipids. In addition, this function may be related to the innate immune system. The evidence that ABC transporters are involved in the regulation of the innate immune system through the transport of various substances greatly enhances the understanding of their clinical significance. ABC transporters are involved in the cellular homeostasis of cholesterol as well as in the regulation of its content in lipid rafts. Through these mechanisms, they can regulate the function of membrane proteins, including receptors of the innate immune system. By regulating lipid transport, some members of ABC transporters are involved in phagocytosis. In addition, ABC transporters are involved in the transport of lipopolysaccharide, lipid mediators of inflammation, and perform other functions in the innate immune system.
Collapse
Affiliation(s)
- Stanislav Kotlyarov
- Department of Nursing, Ryazan State Medical University, 390026 Ryazan, Russia
| | - Anna Kotlyarova
- Department of Pharmacy Management and Economics, Ryazan State Medical University, 390026 Ryazan, Russia
| |
Collapse
|
5
|
Keul P, Peters S, von Wnuck Lipinski K, Schröder NH, Nowak MK, Duse DA, Polzin A, Weske S, Gräler MH, Levkau B. Sphingosine-1-Phosphate (S1P) Lyase Inhibition Aggravates Atherosclerosis and Induces Plaque Rupture in ApoE−/− Mice. Int J Mol Sci 2022; 23:ijms23179606. [PMID: 36077004 PMCID: PMC9455951 DOI: 10.3390/ijms23179606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 08/09/2022] [Accepted: 08/20/2022] [Indexed: 11/17/2022] Open
Abstract
Altered plasma sphingosine-1-phosphate (S1P) concentrations are associated with clinical manifestations of atherosclerosis. However, whether long-term elevation of endogenous S1P is pro- or anti-atherogenic remains unclear. Here, we addressed the impact of permanently high S1P levels on atherosclerosis in cholesterol-fed apolipoprotein E-deficient (ApoE−/−) mice over 12 weeks. This was achieved by pharmacological inhibition of the S1P-degrading enzyme S1P lyase with 4-deoxypyridoxine (DOP). DOP treatment dramatically accelerated atherosclerosis development, propagated predominantly unstable plaque phenotypes, and resulted in frequent plaque rupture with atherothrombosis. Macrophages from S1P lyase-inhibited or genetically deficient mice had a defect in cholesterol efflux to apolipoprotein A-I that was accompanied by profoundly downregulated cholesterol transporters ATP-binding cassette transporters ABCA1 and ABCG1. This was dependent on S1P signaling through S1PR3 and resulted in dramatically enhanced atherosclerosis in ApoE−/−/S1PR3−/− mice, where DOP treatment had no additional effect. Thus, high endogenous S1P levels promote atherosclerosis, compromise cholesterol efflux, and cause genuine plaque rupture.
Collapse
Affiliation(s)
- Petra Keul
- Institute for Molecular Medicine III, University Hospital Düsseldorf, University of Düsseldorf, 40225 Düsseldorf, Germany
| | - Susann Peters
- Institute for Molecular Medicine III, University Hospital Düsseldorf, University of Düsseldorf, 40225 Düsseldorf, Germany
| | - Karin von Wnuck Lipinski
- Institute for Molecular Medicine III, University Hospital Düsseldorf, University of Düsseldorf, 40225 Düsseldorf, Germany
| | - Nathalie H. Schröder
- Institute for Molecular Medicine III, University Hospital Düsseldorf, University of Düsseldorf, 40225 Düsseldorf, Germany
| | - Melissa K. Nowak
- Institute for Molecular Medicine III, University Hospital Düsseldorf, University of Düsseldorf, 40225 Düsseldorf, Germany
| | - Dragos A. Duse
- Institute for Molecular Medicine III, University Hospital Düsseldorf, University of Düsseldorf, 40225 Düsseldorf, Germany
| | - Amin Polzin
- Division of Cardiology, Pulmonology, and Vascular Medicine, Heinrich Heine University Medical Center Düsseldorf, 40225 Düsseldorf, Germany
| | - Sarah Weske
- Institute for Molecular Medicine III, University Hospital Düsseldorf, University of Düsseldorf, 40225 Düsseldorf, Germany
| | - Markus H. Gräler
- Department of Anesthesiology and Intensive Care Medicine, Center for Sepsis Control and Care and Center for Molecular Biomedicine, University Hospital Jena, 07743 Jena, Germany
| | - Bodo Levkau
- Institute for Molecular Medicine III, University Hospital Düsseldorf, University of Düsseldorf, 40225 Düsseldorf, Germany
- Correspondence: ; Tel.: +49-211-88-12611
| |
Collapse
|
6
|
Potential Therapeutic Agents That Target ATP Binding Cassette A1 (ABCA1) Gene Expression. Drugs 2022; 82:1055-1075. [PMID: 35861923 DOI: 10.1007/s40265-022-01743-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/20/2022] [Indexed: 11/03/2022]
Abstract
The cholesterol efflux protein ATP binding cassette protein A1 (ABCA) and apolipoprotein A1 (apo A1) are key constituents in the process of reverse-cholesterol transport (RCT), whereby excess cholesterol in the periphery is transported to the liver where it can be converted primarily to bile acids for either use in digestion or excreted. Due to their essential roles in RCT, numerous studies have been conducted in cells, mice, and humans to more thoroughly understand the pathways that regulate their expression and activity with the goal of developing therapeutics that enhance RCT to reduce the risk of cardiovascular disease. Many of the drugs and natural compounds examined target several transcription factors critical for ABCA1 expression in both macrophages and the liver. Likewise, several miRNAs target not only ABCA1 but also the same transcription factors that are critical for its high expression. However, after years of research and many preclinical and clinical trials, only a few leads have proven beneficial in this regard. In this review we discuss the various transcription factors that serve as drug targets for ABCA1 and provide an update on some important leads.
Collapse
|
7
|
Truslow JG, Goto S, Homilius M, Mow C, Higgins JM, MacRae CA, Deo RC. Cardiovascular Risk Assessment Using Artificial Intelligence-Enabled Event Adjudication and Hematologic Predictors. Circ Cardiovasc Qual Outcomes 2022; 15:e008007. [PMID: 35477255 PMCID: PMC9208816 DOI: 10.1161/circoutcomes.121.008007] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Researchers routinely evaluate novel biomarkers for incorporation into clinical risk models, weighing tradeoffs between cost, availability, and ease of deployment. For risk assessment in population health initiatives, ideal inputs would be those already available for most patients. We hypothesized that common hematologic markers (eg, hematocrit), available in an outpatient complete blood count without differential, would be useful to develop risk models for cardiovascular events. METHODS We developed Cox proportional hazards models for predicting heart attack, ischemic stroke, heart failure hospitalization, revascularization, and all-cause mortality. For predictors, we used 10 hematologic indices (eg, hematocrit) from routine laboratory measurements, collected March 2016 to May 2017 along with demographic data and diagnostic codes. As outcomes, we used neural network-based automated event adjudication of 1 028 294 discharge summaries. We trained models on 23 238 patients from one hospital in Boston and evaluated them on 29 671 patients from a second one. We assessed calibration using Brier score and discrimination using Harrell's concordance index. In addition, to determine the utility of high-dimensional interactions, we compared our proportional hazards models to random survival forest models. RESULTS Event rates in our cohort ranged from 0.0067 to 0.075 per person-year. Models using only hematology indices had concordance index ranging from 0.60 to 0.80 on an external validation set and showed the best discrimination when predicting heart failure (0.80 [95% CI, 0.79-0.82]) and all-cause mortality (0.78 [0.77-0.80]). Compared with models trained only on demographic data and diagnostic codes, models that also used hematology indices had better discrimination and calibration. The concordance index of the resulting models ranged from 0.75 to 0.85 and the improvement in concordance index ranged up to 0.072. Random survival forests had minimal improvement over proportional hazards models. CONCLUSIONS We conclude that low-cost, ubiquitous inputs, if biologically informative, can provide population-level readouts of risk.
Collapse
Affiliation(s)
- James G Truslow
- One Brave Idea and Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA (J.G.T., S.G., M.H., C.A.M., R.C.D.)
| | - Shinichi Goto
- One Brave Idea and Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA (J.G.T., S.G., M.H., C.A.M., R.C.D.).,Department of Medicine (S.G., M.H., C.A.M., R.C.D.), Harvard Medical School, Boston, MA
| | - Max Homilius
- Department of Medicine (S.G., M.H., C.A.M., R.C.D.), Harvard Medical School, Boston, MA
| | - Christopher Mow
- Center for Systems Biology, Massachusetts General Hospital (C.M., J.M.H.), Harvard Medical School, Boston, MA.,Partners Healthcare Enterprise Research Information Systems, Boston, MA (C.M.)
| | - John M Higgins
- Center for Systems Biology, Massachusetts General Hospital (C.M., J.M.H.), Harvard Medical School, Boston, MA.,Department of Pathology, Massachusetts General Hospital (J.M.H.), Harvard Medical School, Boston, MA.,Department of Systems Biology (J.M.H.), Harvard Medical School, Boston, MA
| | - Calum A MacRae
- One Brave Idea and Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA (J.G.T., S.G., M.H., C.A.M., R.C.D.).,Department of Medicine (S.G., M.H., C.A.M., R.C.D.), Harvard Medical School, Boston, MA
| | - Rahul C Deo
- One Brave Idea and Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA (J.G.T., S.G., M.H., C.A.M., R.C.D.).,Department of Medicine (S.G., M.H., C.A.M., R.C.D.), Harvard Medical School, Boston, MA
| |
Collapse
|
8
|
Chen L, Zhao ZW, Zeng PH, Zhou YJ, Yin WJ. Molecular mechanisms for ABCA1-mediated cholesterol efflux. Cell Cycle 2022; 21:1121-1139. [PMID: 35192423 PMCID: PMC9103275 DOI: 10.1080/15384101.2022.2042777] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The maintenance of cellular cholesterol homeostasis is essential for normal cell function and viability. Excessive cholesterol accumulation is detrimental to cells and serves as the molecular basis of many diseases, such as atherosclerosis, Alzheimer's disease, and diabetes mellitus. The peripheral cells do not have the ability to degrade cholesterol. Cholesterol efflux is therefore the only pathway to eliminate excessive cholesterol from these cells. This process is predominantly mediated by ATP-binding cassette transporter A1 (ABCA1), an integral membrane protein. ABCA1 is known to transfer intracellular free cholesterol and phospholipids to apolipoprotein A-I (apoA-I) for generating nascent high-density lipoprotein (nHDL) particles. nHDL can accept more free cholesterol from peripheral cells. Free cholesterol is then converted to cholesteryl ester by lecithin:cholesterol acyltransferase to form mature HDL. HDL-bound cholesterol enters the liver for biliary secretion and fecal excretion. Although how cholesterol is transported by ABCA1 to apoA-I remains incompletely understood, nine models have been proposed to explain this effect. In this review, we focus on the current view of the mechanisms underlying ABCA1-mediated cholesterol efflux to provide an important framework for future investigation and lipid-lowering therapy.
Collapse
Affiliation(s)
- Lei Chen
- Department of Cardiology, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Zhen-Wang Zhao
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Peng-Hui Zeng
- Department of Clinical Laboratory, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Ying-Jie Zhou
- Department of Clinical Laboratory, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Wen-Jun Yin
- Department of Clinical Laboratory, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China,CONTACT Wen-Jun Yin Department of Clinical Laboratory, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan421001, China
| |
Collapse
|
9
|
Nguyen MA, Hoang HD, Rasheed A, Duchez AC, Wyatt H, Lynn Cottee M, Graber TE, Susser L, Robichaud S, Berber İ, Geoffrion M, Ouimet M, Kazan H, Maegdefessel L, Mulvihill EE, Alain T, Rayner KJ. miR-223 Exerts Translational Control of Proatherogenic Genes in Macrophages. Circ Res 2022; 131:42-58. [PMID: 35611698 PMCID: PMC9213086 DOI: 10.1161/circresaha.121.319120] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
A significant burden of atherosclerotic disease is driven by inflammation. Recently, microRNAs (miRNAs) have emerged as important factors driving and protecting from atherosclerosis. miR-223 regulates cholesterol metabolism and inflammation via targeting both cholesterol biosynthesis pathway and NFkB signaling pathways; however, its role in atherosclerosis has not been investigated. We hypothesize that miR-223 globally regulates core inflammatory pathways in macrophages in response to inflammatory and atherogenic stimuli thus limiting the progression of atherosclerosis.
Collapse
Affiliation(s)
- My-Anh Nguyen
- University of Ottawa Heart Institute, Canada (M.-A.N., A.R., A.-C.D., H.W., M.L.C., L.S., S.R., M.G., M.O., E.E.M., K.J.R.).,Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Canada (M.-A.N., H.-D.H., A.R., M.L.C., L.S., S.R., M.O., E.E.M., T.A., K.J.R.)
| | - Huy-Dung Hoang
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, Canada (H.-D.H., T.E.G., T.A.).,Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Canada (M.-A.N., H.-D.H., A.R., M.L.C., L.S., S.R., M.O., E.E.M., T.A., K.J.R.)
| | - Adil Rasheed
- University of Ottawa Heart Institute, Canada (M.-A.N., A.R., A.-C.D., H.W., M.L.C., L.S., S.R., M.G., M.O., E.E.M., K.J.R.).,Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Canada (M.-A.N., H.-D.H., A.R., M.L.C., L.S., S.R., M.O., E.E.M., T.A., K.J.R.)
| | - Anne-Claire Duchez
- University of Ottawa Heart Institute, Canada (M.-A.N., A.R., A.-C.D., H.W., M.L.C., L.S., S.R., M.G., M.O., E.E.M., K.J.R.)
| | - Hailey Wyatt
- University of Ottawa Heart Institute, Canada (M.-A.N., A.R., A.-C.D., H.W., M.L.C., L.S., S.R., M.G., M.O., E.E.M., K.J.R.).,Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Canada (M.-A.N., H.-D.H., A.R., M.L.C., L.S., S.R., M.O., E.E.M., T.A., K.J.R.)
| | - Mary Lynn Cottee
- University of Ottawa Heart Institute, Canada (M.-A.N., A.R., A.-C.D., H.W., M.L.C., L.S., S.R., M.G., M.O., E.E.M., K.J.R.)
| | - Tyson E Graber
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, Canada (H.-D.H., T.E.G., T.A.)
| | - Leah Susser
- University of Ottawa Heart Institute, Canada (M.-A.N., A.R., A.-C.D., H.W., M.L.C., L.S., S.R., M.G., M.O., E.E.M., K.J.R.).,Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Canada (M.-A.N., H.-D.H., A.R., M.L.C., L.S., S.R., M.O., E.E.M., T.A., K.J.R.)
| | - Sabrina Robichaud
- University of Ottawa Heart Institute, Canada (M.-A.N., A.R., A.-C.D., H.W., M.L.C., L.S., S.R., M.G., M.O., E.E.M., K.J.R.).,Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Canada (M.-A.N., H.-D.H., A.R., M.L.C., L.S., S.R., M.O., E.E.M., T.A., K.J.R.)
| | - İbrahim Berber
- Electrical and Computer Engineering Graduate Program, Antalya Bilim University, Turkey (I.B.)
| | - Michele Geoffrion
- University of Ottawa Heart Institute, Canada (M.-A.N., A.R., A.-C.D., H.W., M.L.C., L.S., S.R., M.G., M.O., E.E.M., K.J.R.)
| | - Mireille Ouimet
- University of Ottawa Heart Institute, Canada (M.-A.N., A.R., A.-C.D., H.W., M.L.C., L.S., S.R., M.G., M.O., E.E.M., K.J.R.).,Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Canada (M.-A.N., H.-D.H., A.R., M.L.C., L.S., S.R., M.O., E.E.M., T.A., K.J.R.)
| | - Hilal Kazan
- Department of Computer Engineering, Antalya Bilim University, Turkey (H.K.)
| | - Lars Maegdefessel
- Department of Vascular and Endovascular Surgery, Technical University Munich, Germany (L.M.).,Department of Medicine, Karolinska Institute, Stockholm, Sweden (L.M.)
| | - Erin E Mulvihill
- University of Ottawa Heart Institute, Canada (M.-A.N., A.R., A.-C.D., H.W., M.L.C., L.S., S.R., M.G., M.O., E.E.M., K.J.R.).,Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Canada (M.-A.N., H.-D.H., A.R., M.L.C., L.S., S.R., M.O., E.E.M., T.A., K.J.R.)
| | - Tommy Alain
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, Canada (H.-D.H., T.E.G., T.A.).,Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Canada (M.-A.N., H.-D.H., A.R., M.L.C., L.S., S.R., M.O., E.E.M., T.A., K.J.R.)
| | - Katey J Rayner
- University of Ottawa Heart Institute, Canada (M.-A.N., A.R., A.-C.D., H.W., M.L.C., L.S., S.R., M.G., M.O., E.E.M., K.J.R.).,Centre for Infection, Immunity & Inflammation, Faculty of Medicine, University of Ottawa, Canada (K.J.R.).,Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Canada (M.-A.N., H.-D.H., A.R., M.L.C., L.S., S.R., M.O., E.E.M., T.A., K.J.R.)
| |
Collapse
|
10
|
Esobi I, Olanrewaju O, Echesabal-Chen J, Stamatikos A. Utilizing the LoxP-Stop-LoxP System to Control Transgenic ABC-Transporter Expression In Vitro. Biomolecules 2022; 12:679. [PMID: 35625607 PMCID: PMC9138957 DOI: 10.3390/biom12050679] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 04/20/2022] [Accepted: 05/06/2022] [Indexed: 12/02/2022] Open
Abstract
ABCA1 and ABCG1 are two ABC-transporters well-recognized to promote the efflux of cholesterol to apoAI and HDL, respectively. As these two ABC-transporters are critical to cholesterol metabolism, several studies have assessed the impact of ABCA1 and ABCG1 expression on cellular cholesterol homeostasis through ABC-transporter ablation or overexpressing ABCA1/ABCG1. However, for the latter, there are currently no well-established in vitro models to effectively induce long-term ABC-transporter expression in a variety of cultured cells. Therefore, we performed proof-of-principle in vitro studies to determine whether a LoxP-Stop-LoxP (LSL) system would provide Cre-inducible ABC-transporter expression. In our studies, we transfected HEK293 cells and the HEK293-derived cell line 293-Cre cells with ABCA1-LSL and ABCG1-LSL-based plasmids. Our results showed that while the ABCA1/ABCG1 protein expression was absent in the transfected HEK293 cells, the ABCA1 and ABCG1 protein expression was detected in the 293-Cre cells transfected with ABCA1-LSL and ABCG1-LSL, respectively. When we measured cholesterol efflux in transfected 293-Cre cells, we observed an enhanced apoAI-mediated cholesterol efflux in 293-Cre cells overexpressing ABCA1, and an HDL2-mediated cholesterol efflux in 293-Cre cells constitutively expressing ABCG1. We also observed an appreciable increase in HDL3-mediated cholesterol efflux in ABCA1-overexpressing 293-Cre cells, which suggests that ABCA1 is capable of effluxing cholesterol to small HDL particles. Our proof-of-concept experiments demonstrate that the LSL-system can be used to effectively regulate ABC-transporter expression in vitro, which, in turn, allows ABCA1/ABCG1-overexpression to be extensively studied at the cellular level.
Collapse
Affiliation(s)
| | | | | | - Alexis Stamatikos
- Department of Food, Nutrition, and Packaging Sciences, Clemson University, Clemson, SC 29634, USA; (I.E.); (O.O.); (J.E.-C.)
| |
Collapse
|
11
|
Ouweneel AB, Reiche ME, Snip OSC, Wever R, van der Wel EJ, Schaftenaar FH, Kauerova S, Lutgens E, Van Eck M, Hoekstra M. Apolipoprotein A1 deficiency in mice primes bone marrow stem cells for T cell lymphopoiesis. J Cell Sci 2022; 135:272619. [PMID: 34698355 PMCID: PMC8645231 DOI: 10.1242/jcs.258901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 10/14/2021] [Indexed: 11/20/2022] Open
Abstract
The bone marrow has emerged as a potentially important target in cardiovascular disease as it generates all leukocytes involved in atherogenesis. In the current study, we evaluated whether a change in bone marrow functionality underlies the increased atherosclerosis susceptibility associated with high-density lipoprotein (HDL) deficiency. We found that HDL deficiency in mice due to the genetic lack of hepatocyte-derived apolipoprotein A1 (APOA1) was associated with an increase in the Lin−Sca-1+Kit+ (LSK) bone marrow stem cell population and lymphoid-primed multipotent progenitor numbers, which translated into a higher production and systemic flux of T cell subsets. In accordance with APOA1 deficiency-associated priming of stem cells to increase T lymphocyte production, atherogenic diet-fed low-density lipoprotein receptor knockout mice transplanted with bone marrow from APOA1-knockout mice displayed marked lymphocytosis as compared to wild-type bone marrow recipients. However, atherosclerotic lesion sizes and collagen contents were similar in the two groups of bone marrow recipients. In conclusion, systemic lack of APOA1 primes bone marrow stem cells for T cell lymphopoiesis. Our data provide novel evidence for a regulatory role of HDL in bone marrow functioning in normolipidemic mice. Summary: Changes in cholesterol metabolism, that is, in high-density lipoprotein levels, can significantly impact leukocyte numbers via modulating bone marrow functionality.
Collapse
Affiliation(s)
- Amber B Ouweneel
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, 2333CC Leiden, The Netherlands
| | - Myrthe E Reiche
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, 1105AZ Amsterdam, The Netherlands
| | - Olga S C Snip
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, 2333CC Leiden, The Netherlands
| | - Robbert Wever
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, 2333CC Leiden, The Netherlands
| | - Ezra J van der Wel
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, 2333CC Leiden, The Netherlands
| | - Frank H Schaftenaar
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, 2333CC Leiden, The Netherlands
| | - Soňa Kauerova
- Laboratory for Atherosclerosis Research, Institute for Clinical and Experimental Medicine, 12111 Prague, Czech Republic
| | - Esther Lutgens
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, 1105AZ Amsterdam, The Netherlands
| | - Miranda Van Eck
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, 2333CC Leiden, The Netherlands
| | - Menno Hoekstra
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, 2333CC Leiden, The Netherlands
| |
Collapse
|
12
|
Gündoğdu Y, Anaforoğlu İ. Effects of Smoking on Diabetic Nephropathy. FRONTIERS IN CLINICAL DIABETES AND HEALTHCARE 2022; 3:826383. [PMID: 36992741 PMCID: PMC10012135 DOI: 10.3389/fcdhc.2022.826383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/28/2022] [Indexed: 11/13/2022]
Abstract
Diabetes is a systemic metabolic disease with serious complications that cause significant stress on the healthcare system. Diabetic kidney disease is the primary cause of end stage renal disease globally and its progression is accelerated by various factors. Another major healthcare hazard is tobacco consumption and smoking has deleterious effects on renal physiology. Prominent factors are defined as sympathetic activity, atherosclerosis, oxidative stress and dyslipidemia. This review aims to enlighten the mechanism underlying the cumulative negative effect of simultaneous exposure to hyperglycemia and nicotine.
Collapse
Affiliation(s)
- Yasemin Gündoğdu
- School of Medicine, Department of Internal Medicine, Acıbadem Mehmet Ali Aydınlar University, Istanbul, Turkey
| | - İnan Anaforoğlu
- School of Medicine, Department of Endocrinology and Metabolism, Acıbadem Mehmet Ali Aydınlar University, Istanbul, Turkey
| |
Collapse
|
13
|
Araki MVR, Silva YCO, Rodrigues TAR, Bajano FF, de Souza BB, Costa FF, Costa VP, de Melo MB, de Vasconcellos JPC. Association of ABCA1 (rs2472493) and GAS7 (rs9913911) gene variants with primary open-angle glaucoma in a Brazilian population. Mol Vis 2022; 28:1-10. [PMID: 35400990 PMCID: PMC8942454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 02/20/2022] [Indexed: 11/11/2022] Open
Abstract
Purpose Glaucoma is the world's leading cause of irreversible blindness, with primary open-angle glaucoma (POAG) being the most prevalent subtype. In recent years, there have been advances in knowledge about the genetics involved in POAG, but genetic studies in admixed populations, such as Brazilians, are still rare. This study aimed to evaluate the association of single nucleotide variants (SNV) of the ABCA1 (rs2472493) and GAS7 (rs9913911) genes with POAG in a sample of the Brazilian population. Furthermore, the study aimed to evaluate the relationship between these SNVs and the need for surgical intervention in glaucoma control. Methods A cross-sectional association study with 1,009 subjects (505 patients with POAG and 504 controls) was performed. Participants underwent a comprehensive ocular examination, including the need for surgical procedures for intraocular pressure control. Genotyping of SNVs was performed using the TaqMan genotyping assay. Results SNV rs9913911 of GAS7 was found to be associated with POAG in the presence of the risk allele A (p = 0.0004) and the AA genotype (p = 0.002). There was no association between SNV rs2472493 of ABCA1 for either the allele risk or genotypes. However, the combination of these variants showed an additive effect on the risk for POAG: ABCA1(GG) + GAS7(AA; p = 0.02), ABCA1(GG) + GAS7(AG; p = 0.003), and ABCA1(AG) + GAS7(AG; p = 0.004). Also, POAG patients carrying the AA genotype of the GAS7 gene required antiglaucomatous surgery more frequently than those without the AA genotype (p = 0.01). Conclusions In a Brazilian population sample, there was an association identified between SNV rs9913911 (GAS7) and the risk of POAG, and an additive effect was found when GAS7 was combined with SNV rs2472493 (ABCA1). There was an association between SNV rs9913911 (GAS7) and the risk for antiglaucomatous surgery.
Collapse
Affiliation(s)
- Manoel Vinicius Rocha Araki
- Department of Ophthalmology, Faculty of Medical Sciences, University of Campinas – UNICAMP, Campinas – SP, Brazil
| | - Yuri Carvalho Oiamore Silva
- Laboratory of Human Genetics, Center for Molecular Biology and Genetic Engineering, University of Campinas – UNICAMP, Campinas – SP, Brazil
| | - Thiago Adalton Rosa Rodrigues
- Laboratory of Human Genetics, Center for Molecular Biology and Genetic Engineering, University of Campinas – UNICAMP, Campinas – SP, Brazil
| | - Flavia Fialho Bajano
- Laboratory of Human Genetics, Center for Molecular Biology and Genetic Engineering, University of Campinas – UNICAMP, Campinas – SP, Brazil
| | - Bruno Batista de Souza
- Laboratory of Human Genetics, Center for Molecular Biology and Genetic Engineering, University of Campinas – UNICAMP, Campinas – SP, Brazil
| | | | - Vital Paulino Costa
- Department of Ophthalmology, Faculty of Medical Sciences, University of Campinas – UNICAMP, Campinas – SP, Brazil
| | - Mônica Barbosa de Melo
- Laboratory of Human Genetics, Center for Molecular Biology and Genetic Engineering, University of Campinas – UNICAMP, Campinas – SP, Brazil
| | | |
Collapse
|
14
|
Javadifar A, Ghezeldasht SA, Rahimi H, Valizadeh N, Borojerdi ZR, Vahidi Z, Rezaee SR. Possible deterioration of Apolipoproteins expression by HTLV-1 infection in favor of infected leukemic cells in adult T-cell leukemia/lymphoma (ATLL). GENE REPORTS 2022. [DOI: 10.1016/j.genrep.2022.101531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
15
|
HDL Is Not Dead Yet. Biomedicines 2022; 10:biomedicines10010128. [PMID: 35052806 PMCID: PMC8773442 DOI: 10.3390/biomedicines10010128] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/04/2022] [Accepted: 01/05/2022] [Indexed: 12/11/2022] Open
Abstract
High-density lipoprotein cholesterol (HDL-C) levels are inversely correlated with coronary heart disease (CHD) in multiple epidemiological studies, but whether HDL is causal or merely associated with CHD is unclear. Recent trials for HDL-raising drugs were either not effective in reducing CHD events or, if beneficial in reducing CHD events, were not conclusive as the findings could be attributed to the drugs’ LDL-reducing activity. Furthermore, the first large Mendelian randomization study did not causally relate HDL-C levels to decreased CHD. Thus, the hypothesis that HDL is protective against CHD has been rightfully challenged. However, subsequent Mendelian randomization studies found HDL characteristics that are causally related to decreased CHD. Many aspects of HDL structure and function, especially in reverse cholesterol transport, may be better indicators of HDL’s protective activity than simply measuring HDL-C. Cholesterol efflux capacity is associated with lower levels of prevalent and incident CHD, even after adjustment for HDL-C and apolipoprotein A-1 levels. Also, subjects with very high levels of HDL-C, including those with rare mutations that disrupt hepatic HDL uptake and reverse cholesterol transport, may be at higher risk for CHD than those with moderate levels. We describe here several cell-based and cell-free in vitro assays of HDL structure and function that may be used in clinical studies to determine which of HDL’s functions are best associated with protection against CHD. We conclude that the HDL hypothesis may need revision based on studies of HDL structure and function, but that the HDL hypothesis is not dead yet.
Collapse
|
16
|
Xian X, Wang Y, Liu G. Genetically Engineered Hamster Models of Dyslipidemia and Atherosclerosis. Methods Mol Biol 2022; 2419:433-459. [PMID: 35237980 DOI: 10.1007/978-1-0716-1924-7_26] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Animal models of human diseases play an extremely important role in biomedical research. Among them, mice are widely used animal models for translational research, especially because of ease of generation of genetically engineered mice. However, because of the great differences in biology between mice and humans, translation of findings to humans remains a major issue. Therefore, the exploration of models with biological and metabolic characteristics closer to those of humans has never stopped.Although pig and nonhuman primates are biologically similar to humans, their genetic engineering is technically difficult, the cost of breeding is high, and the experimental time is long. As a result, the application of these species as model animals, especially genetically engineered model animals, in biomedical research is greatly limited.In terms of lipid metabolism and cardiovascular diseases, hamsters have several characteristics different from rats and mice, but similar to those in humans. The hamster is therefore an ideal animal model for studying lipid metabolism and cardiovascular disease because of its small size and short reproduction period. However, the phenomenon of zygote division, which was unexpectedly blocked during the manipulation of hamster embryos for some unknown reasons, had plagued researchers for decades and no genetically engineered hamsters have therefore been generated as animal models of human diseases for a long time. After solving the problem of in vitro development of hamster zygotes, we successfully prepared enhanced green fluorescent protein (eGFP) transgenic hamsters by microinjection of lentiviral vectors into the zona pellucida space of zygotes. On this basis, we started the development of cardiovascular disease models using the hamster embryo culture system combined with the novel genome editing technique of clustered regularly interspaced short palindromic repeats (CRISPR )/CRISPR associated protein 9 (Cas9). In this chapter, we will introduce some of the genetically engineered hamster models with dyslipidemia and the corresponding characteristics of these models. We hope that the genetically engineered hamster models can be further recognized and complement other genetically engineered animal models such as mice, rats, and rabbits. This will lead to new avenues and pathways for the study of lipid metabolism and its related diseases.
Collapse
Affiliation(s)
- Xunde Xian
- Institute of Cardiovascular Sciences, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Yuhui Wang
- Institute of Cardiovascular Sciences, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, School of Basic Medical Sciences, Peking University, Beijing, China
| | - George Liu
- Institute of Cardiovascular Sciences, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, School of Basic Medical Sciences, Peking University, Beijing, China.
| |
Collapse
|
17
|
Rozhkova AV, Dmitrieva VG, Nosova EV, Dergunov AD, Limborska SA, Dergunova LV. Genomic Variants and Multilevel Regulation of ABCA1, ABCG1, and SCARB1 Expression in Atherogenesis. J Cardiovasc Dev Dis 2021; 8:jcdd8120170. [PMID: 34940525 PMCID: PMC8707585 DOI: 10.3390/jcdd8120170] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 11/28/2021] [Accepted: 11/29/2021] [Indexed: 12/12/2022] Open
Abstract
Atheroprotective properties of human plasma high-density lipoproteins (HDLs) are determined by their involvement in reverse cholesterol transport (RCT) from the macrophage to the liver. ABCA1, ABCG1, and SR-BI cholesterol transporters are involved in cholesterol efflux from macrophages to lipid-free ApoA-I and HDL as a first RCT step. Molecular determinants of RCT efficiency that may possess diagnostic and therapeutic meaning remain largely unknown. This review summarizes the progress in studying the genomic variants of ABCA1, ABCG1, and SCARB1, and the regulation of their function at transcriptional and post-transcriptional levels in atherosclerosis. Defects in the structure and function of ABCA1, ABCG1, and SR-BI are caused by changes in the gene sequence, such as single nucleotide polymorphism or various mutations. In the transcription initiation of transporter genes, in addition to transcription factors, long noncoding RNA (lncRNA), transcription activators, and repressors are also involved. Furthermore, transcription is substantially influenced by the methylation of gene promoter regions. Post-transcriptional regulation involves microRNAs and lncRNAs, including circular RNAs. The potential biomarkers and targets for atheroprotection, based on molecular mechanisms of expression regulation for three transporter genes, are also discussed in this review.
Collapse
Affiliation(s)
- Alexandra V. Rozhkova
- Department of Molecular Bases of Human Genetics, Institute of Molecular Genetics of National Research Center “Kurchatov Institute”, 123182 Moscow, Russia; (A.V.R.); (V.G.D.); (E.V.N.); (S.A.L.); (L.V.D.)
| | - Veronika G. Dmitrieva
- Department of Molecular Bases of Human Genetics, Institute of Molecular Genetics of National Research Center “Kurchatov Institute”, 123182 Moscow, Russia; (A.V.R.); (V.G.D.); (E.V.N.); (S.A.L.); (L.V.D.)
| | - Elena V. Nosova
- Department of Molecular Bases of Human Genetics, Institute of Molecular Genetics of National Research Center “Kurchatov Institute”, 123182 Moscow, Russia; (A.V.R.); (V.G.D.); (E.V.N.); (S.A.L.); (L.V.D.)
| | - Alexander D. Dergunov
- Laboratory of Structural Fundamentals of Lipoprotein Metabolism, National Medical Research Center for Therapy and Preventive Medicine, 101990 Moscow, Russia
- Correspondence:
| | - Svetlana A. Limborska
- Department of Molecular Bases of Human Genetics, Institute of Molecular Genetics of National Research Center “Kurchatov Institute”, 123182 Moscow, Russia; (A.V.R.); (V.G.D.); (E.V.N.); (S.A.L.); (L.V.D.)
| | - Liudmila V. Dergunova
- Department of Molecular Bases of Human Genetics, Institute of Molecular Genetics of National Research Center “Kurchatov Institute”, 123182 Moscow, Russia; (A.V.R.); (V.G.D.); (E.V.N.); (S.A.L.); (L.V.D.)
| |
Collapse
|
18
|
Liu G, Lai P, Guo J, Wang Y, Xian X. Genetically-engineered hamster models: applications and perspective in dyslipidemia and atherosclerosis-related cardiovascular disease. MEDICAL REVIEW (BERLIN, GERMANY) 2021; 1:92-110. [PMID: 37724074 PMCID: PMC10388752 DOI: 10.1515/mr-2021-0004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 08/03/2021] [Indexed: 09/20/2023]
Abstract
Cardiovascular disease is the leading cause of morbidity and mortality in both developed and developing countries, in which atherosclerosis triggered by dyslipidemia is the major pathological basis. Over the past 40 years, small rodent animals, such as mice, have been widely used for understanding of human atherosclerosis-related cardiovascular disease (ASCVD) with the advantages of low cost and ease of maintenance and manipulation. However, based on the concept of precision medicine and high demand of translational research, the applications of mouse models for human ASCVD study would be limited due to the natural differences in metabolic features between mice and humans even though they are still the most powerful tools in this research field, indicating that other species with biological similarity to humans need to be considered for studying ASCVD in future. With the development and breakthrough of novel gene editing technology, Syrian golden hamster, a small rodent animal replicating the metabolic characteristics of humans, has been genetically modified, suggesting that gene-targeted hamster models will provide new insights into the precision medicine and translational research of ASCVD. The purpose of this review was to summarize the genetically-modified hamster models with dyslipidemia to date, and their potential applications and perspective for ASCVD.
Collapse
Affiliation(s)
- George Liu
- Institute of Cardiovascular Sciences and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, School of Basic Medical Sciences, Peking University 38 Xueyuan Road, Beijing 100191, China
| | - Pingping Lai
- Institute of Cardiovascular Sciences and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, School of Basic Medical Sciences, Peking University 38 Xueyuan Road, Beijing 100191, China
| | - Jiabao Guo
- Institute of Cardiovascular Sciences and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, School of Basic Medical Sciences, Peking University 38 Xueyuan Road, Beijing 100191, China
| | - Yuhui Wang
- Institute of Cardiovascular Sciences and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, School of Basic Medical Sciences, Peking University 38 Xueyuan Road, Beijing 100191, China
| | - Xunde Xian
- Institute of Cardiovascular Sciences and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, School of Basic Medical Sciences, Peking University 38 Xueyuan Road, Beijing 100191, China
| |
Collapse
|
19
|
Guevara-Cruz M, Medina-Vera I, Cu-Cañetas TE, Cordero-Chan Y, Torres N, Tovar AR, Márquez-Mota C, Talamantes-Gómez JM, Pérez-Monter C, Lugo R, Gutiérrez-Solis AL, Avila-Nava A. Chaya Leaf Decreased Triglycerides and Improved Oxidative Stress in Subjects With Dyslipidemia. Front Nutr 2021; 8:666243. [PMID: 34368206 PMCID: PMC8343181 DOI: 10.3389/fnut.2021.666243] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 06/28/2021] [Indexed: 12/14/2022] Open
Abstract
Chaya is an edible leaf popular in Mexico and Central America because of its high nutritional value. Studies in animal models have demonstrated the beneficial effects of Chaya, which include reduction of circulating lipids and increase in antioxidant activity. However, its hypolipidemic and antioxidant effects have not been demonstrated in humans. Thus, the aim of the present study was to evaluate the effect of Chaya on the lipid profile, lipid peroxidation, inflammation, and peripheral blood mononuclear cell gene expression in a population with dyslipidemia. We performed a single-arm trial in 30 participants with dyslipidemia who consumed 500 mL of Chaya beverage per day over a 6-week period. Interestingly, we observed a significant decrease in serum triglyceride concentration (P < 0.05) and an increase in plasma antioxidant activity and polyphenol concentration (P < 0.005) after 6 weeks of Chaya consumption. This was accompanied by a reduction in the oxidative stress marker MDA (P < 0.0001) and by an increase in the antioxidant enzyme CAT expression in peripheral blood mononuclear cells (P < 0.001). Altogether, our results demonstrate that consumption of Chaya has hypotriglyceridemic and antioxidant effects in subjects with dyslipidemia.
Collapse
Affiliation(s)
- Martha Guevara-Cruz
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Nutrición y Ciencias Médicas Salvador Zubirán, Ciudad de México, Mexico.,Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, Mexico
| | - Isabel Medina-Vera
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, Mexico.,Departamento de Metodología, Instituto Nacional de Pediatría, Ciudad de México, Mexico
| | | | | | - Nimbe Torres
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Nutrición y Ciencias Médicas Salvador Zubirán, Ciudad de México, Mexico
| | - Armando R Tovar
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Nutrición y Ciencias Médicas Salvador Zubirán, Ciudad de México, Mexico
| | - Claudia Márquez-Mota
- Departamento de Nutrición Animal y Bioquímica, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México (FMVZ-UNAM), Ciudad de México, Mexico
| | - José Moisés Talamantes-Gómez
- Departamento de Nutrición Animal y Bioquímica, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México (FMVZ-UNAM), Ciudad de México, Mexico
| | - Carlos Pérez-Monter
- Departamento de Gastroenterología, Instituto Nacional de Nutrición y Ciencias Médicas Salvador Zubirán, Ciudad de México, Mexico
| | - Roberto Lugo
- Hospital Regional de Alta Especialidad de la Península de Yucatán, Mérida, Mexico
| | | | - Azalia Avila-Nava
- Hospital Regional de Alta Especialidad de la Península de Yucatán, Mérida, Mexico
| |
Collapse
|
20
|
Kotlyarov S, Kotlyarova A. The Role of ABC Transporters in Lipid Metabolism and the Comorbid Course of Chronic Obstructive Pulmonary Disease and Atherosclerosis. Int J Mol Sci 2021; 22:6711. [PMID: 34201488 PMCID: PMC8269124 DOI: 10.3390/ijms22136711] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 06/12/2021] [Accepted: 06/18/2021] [Indexed: 12/11/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) ranks among the leading causes of morbidity and mortality worldwide. COPD rarely occurs in isolation and is often combined with various diseases. It is considered that systemic inflammation underlies the comorbid course of COPD. The data obtained in recent years have shown the importance of violations of the cross-links of lipid metabolism and the immune response, which are links in the pathogenesis of both COPD and atherosclerosis. The role of lipid metabolism disorders in the pathogenesis of the comorbid course of COPD and atherosclerosis and the participation of ATP-binding cassette (ABC) transporters in these processes is discussed in this article. It is known that about 20 representatives of a large family of ABC transporters provide lipid homeostasis of cells by moving lipids inside the cell and in its plasma membrane, as well as removing lipids from the cell. It was shown that some representatives of the ABC-transporter family are involved in various links of the pathogenesis of COPD and atherosclerosis, which can determine their comorbid course.
Collapse
Affiliation(s)
- Stanislav Kotlyarov
- Department of Nursing, Ryazan State Medical University, 390026 Ryazan, Russia
| | - Anna Kotlyarova
- Department of Pharmacology and Pharmacy, Ryazan State Medical University, 390026 Ryazan, Russia;
| |
Collapse
|
21
|
Thomas C, Gautier T, Masson D. Non-lipogenic ABCA1 inducers: The holy grail in cardio-metabolic diseases? EBioMedicine 2021; 66:103324. [PMID: 33857903 PMCID: PMC8050846 DOI: 10.1016/j.ebiom.2021.103324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 03/19/2021] [Indexed: 11/20/2022] Open
Affiliation(s)
- Charles Thomas
- LNC UMR1231, University Bourgogne Franche-Comté, Dijon F-21000, France; LNC UMR1231, INSERM, Dijon F-21000, France; LipSTIC LabEx, FCS Bourgogne-Franche Comté, Dijon F-21000, France
| | - Thomas Gautier
- LNC UMR1231, University Bourgogne Franche-Comté, Dijon F-21000, France; LNC UMR1231, INSERM, Dijon F-21000, France; LipSTIC LabEx, FCS Bourgogne-Franche Comté, Dijon F-21000, France
| | - David Masson
- LNC UMR1231, University Bourgogne Franche-Comté, Dijon F-21000, France; LNC UMR1231, INSERM, Dijon F-21000, France; LipSTIC LabEx, FCS Bourgogne-Franche Comté, Dijon F-21000, France; Laboratory of Clinical Chemistry, CHU Dijon, Dijon F-21000, France.
| |
Collapse
|
22
|
Lewandowski CT, Khan MW, BenAissa M, Dubrovskyi O, Ackerman-Berrier M, LaDu MJ, Layden BT, Thatcher GRJ. Metabolomic analysis of a selective ABCA1 inducer in obesogenic challenge provides a rationale for therapeutic development. EBioMedicine 2021; 66:103287. [PMID: 33752129 PMCID: PMC8010624 DOI: 10.1016/j.ebiom.2021.103287] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 03/02/2021] [Accepted: 03/02/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Therapeutic agents with novel mechanisms of action are needed to combat the growing epidemic of type 2 diabetes (T2D) and related metabolic syndromes. Liver X receptor (LXR) agonists possess preclinical efficacy yet produce side effects due to excessive lipogenesis. Anticipating that many beneficial and detrimental effects of LXR agonists are mediated by ABCA1 and SREPB1c expression, respectively, we hypothesized that a phenotypic optimization strategy prioritizing selective ABCA1 induction would identify an efficacious lead compound with an improved side effect profile over existing LXRβ agonists. METHODS We synthesized and characterized a novel small molecule for selective induction of ABCA1 vs. SREBP1c in vitro. This compound was evaluated in both wild-type mice and a high-fat diet (HFD) mouse model of obesity-driven diabetes through functional, biochemical, and metabolomic analysis. FINDINGS Six weeks of oral administration of our lead compound attenuated weight gain, glucose intolerance, insulin signaling deficits, and adiposity. Global metabolomics revealed suppression of gluconeogenesis, free fatty acids, and pro-inflammatory metabolites. Target identification linked these beneficial effects to selective LXRβ agonism and PPAR/RXR antagonism. INTERPRETATION Our observations in the HFD model, combined with the absence of lipogenesis and neutropenia in WT mice, support this novel approach to therapeutic development for T2D and related conditions.
Collapse
Affiliation(s)
- Cutler T Lewandowski
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Md Wasim Khan
- Department of Medicine, University of Illinois at Chicago, 835 S. Wolcott St., Chicago, IL 60612, USA
| | - Manel BenAissa
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Oleksii Dubrovskyi
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Martha Ackerman-Berrier
- Department of Pharmacology and Toxicology, University of Arizona, 1295N. Martin, Tucson, AZ 85721, USA
| | - Mary Jo LaDu
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, USA
| | - Brian T Layden
- Department of Medicine, University of Illinois at Chicago, 835 S. Wolcott St., Chicago, IL 60612, USA.
| | - Gregory R J Thatcher
- Department of Pharmacology and Toxicology, University of Arizona, 1295N. Martin, Tucson, AZ 85721, USA.
| |
Collapse
|
23
|
Ben Aissa M, Lewandowski CT, Ratia KM, Lee SH, Layden BT, LaDu MJ, Thatcher GRJ. Discovery of Nonlipogenic ABCA1 Inducing Compounds with Potential in Alzheimer's Disease and Type 2 Diabetes. ACS Pharmacol Transl Sci 2021; 4:143-154. [PMID: 33615168 PMCID: PMC7887740 DOI: 10.1021/acsptsci.0c00149] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Indexed: 02/07/2023]
Abstract
Selective liver X receptor (LXR) agonists have been extensively pursued as therapeutics for Alzheimer's disease and related dementia (ADRD) and, for comorbidities such as type 2 diabetes (T2D) and cerebrovascular disease (CVD), disorders with underlying impaired insulin signaling, glucose metabolism, and cholesterol mobilization. The failure of the LXR-focused approach led us to pursue a novel strategy to discover nonlipogenic ATP-binding cassette transporter A1 (ABCA1) inducers (NLAIs): screening for ABCA1-luciferase activation in astrocytoma cells and counterscreening against lipogenic gene upregulation in hepatocarcinoma cells. Beneficial effects of LXRβ agonists mediated by ABCA1 include the following: control of cholesterol and phospholipid efflux to lipid-poor apolipoproteins forming beneficial peripheral HDL and HDL-like particles in the brain and attenuation of inflammation. While rare, ABCA1 variants reduce plasma HDL and correlate with an increased risk of ADRD and CVD. In secondary assays, NLAI hits enhanced cholesterol mobilization and positively impacted in vitro biomarkers associated with insulin signaling, inflammatory response, and biogenic properties. In vivo target engagement was demonstrated after oral administration of NLAIs in (i) mice fed a high-fat diet, a model for obesity-linked T2D, (ii) mice administered LPS, and (iii) mice with accelerated oxidative stress. The lack of adverse effects on lipogenesis and positive effects on multiple biomarkers associated with T2D and ADRD supports this novel phenotypic approach to NLAIs as a platform for T2D and ADRD drug discovery.
Collapse
Affiliation(s)
- Manel Ben Aissa
- Department
of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago (UIC), Chicago, Illinois 60612, United States
- UICentre
(Drug Discovery @ UIC), University of Illinois
at Chicago (UIC), Chicago, Illinois 60612, United States
| | - Cutler T. Lewandowski
- Department
of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago (UIC), Chicago, Illinois 60612, United States
| | - Kiira M. Ratia
- HTS
Screening Facility, Research Resources Center, University of Illinois at Chicago (UIC), Chicago, Illinois 60612, United States
| | - Sue H. Lee
- Department
of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago (UIC), Chicago, Illinois 60612, United States
| | - Brian T. Layden
- Department
of Medicine, University of Illinois at Chicago
(UIC), Chicago, Illinois 60612, United States
| | - Mary Jo LaDu
- Department
of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago (UIC), Chicago, Illinois 60612, United States
| | - Gregory R. J. Thatcher
- Department
of Pharmacology & Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona 85721, United States
| |
Collapse
|
24
|
Xu (许艳妮) Y, Liu (刘畅) C, Han (韩小婉) X, Jia (贾晓健) X, Li (李永臻) Y, Liu (刘超) C, Li (李霓) N, Liu (刘伦铭) L, Liu (刘鹏) P, Jiang (姜新海) X, Wang (王伟志) W, Wang (王潇) X, Li (李依宁) Y, Chen (陈明珠) M, Luo (罗金雀) J, Zuo (左璇) X, Han (韩江雪) J, Wang (王丽) L, Du (杜郁) Y, Xu (徐扬) Y, Jiang (蒋建东) JD, Hong (洪斌) B, Si (司书毅) S. E17241 as a Novel ABCA1 (ATP-Binding Cassette Transporter A1) Upregulator Ameliorates Atherosclerosis in Mice. Arterioscler Thromb Vasc Biol 2021; 41:e284-e298. [PMID: 33441025 DOI: 10.1161/atvbaha.120.314156] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Yanni Xu (许艳妮)
- NHC Key Laboratory of Biotechnology of Antibiotics, National Center for New Microbial Drug Screening, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS&PUMC), Beijing, China (Y.X., C.L., X.H., X. Jia, Y.L., C.L., N.L., L.L., P.L., X. Jiang, W.W., X.W., Y.L., M.C., J.L., X.Z., J.H., L.W., Y.D., Y.X., J.-D.J., B.H., S.S.)
| | - Chang Liu (刘畅)
- NHC Key Laboratory of Biotechnology of Antibiotics, National Center for New Microbial Drug Screening, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS&PUMC), Beijing, China (Y.X., C.L., X.H., X. Jia, Y.L., C.L., N.L., L.L., P.L., X. Jiang, W.W., X.W., Y.L., M.C., J.L., X.Z., J.H., L.W., Y.D., Y.X., J.-D.J., B.H., S.S.)
| | - Xiaowan Han (韩小婉)
- NHC Key Laboratory of Biotechnology of Antibiotics, National Center for New Microbial Drug Screening, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS&PUMC), Beijing, China (Y.X., C.L., X.H., X. Jia, Y.L., C.L., N.L., L.L., P.L., X. Jiang, W.W., X.W., Y.L., M.C., J.L., X.Z., J.H., L.W., Y.D., Y.X., J.-D.J., B.H., S.S.).,State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, CAMS&PUMC, Beijing, China (X.H., N.L., J.-D.J.)
| | - Xiaojian Jia (贾晓健)
- NHC Key Laboratory of Biotechnology of Antibiotics, National Center for New Microbial Drug Screening, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS&PUMC), Beijing, China (Y.X., C.L., X.H., X. Jia, Y.L., C.L., N.L., L.L., P.L., X. Jiang, W.W., X.W., Y.L., M.C., J.L., X.Z., J.H., L.W., Y.D., Y.X., J.-D.J., B.H., S.S.)
| | - Yongzhen Li (李永臻)
- NHC Key Laboratory of Biotechnology of Antibiotics, National Center for New Microbial Drug Screening, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS&PUMC), Beijing, China (Y.X., C.L., X.H., X. Jia, Y.L., C.L., N.L., L.L., P.L., X. Jiang, W.W., X.W., Y.L., M.C., J.L., X.Z., J.H., L.W., Y.D., Y.X., J.-D.J., B.H., S.S.)
| | - Chao Liu (刘超)
- NHC Key Laboratory of Biotechnology of Antibiotics, National Center for New Microbial Drug Screening, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS&PUMC), Beijing, China (Y.X., C.L., X.H., X. Jia, Y.L., C.L., N.L., L.L., P.L., X. Jiang, W.W., X.W., Y.L., M.C., J.L., X.Z., J.H., L.W., Y.D., Y.X., J.-D.J., B.H., S.S.)
| | - Ni Li (李霓)
- NHC Key Laboratory of Biotechnology of Antibiotics, National Center for New Microbial Drug Screening, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS&PUMC), Beijing, China (Y.X., C.L., X.H., X. Jia, Y.L., C.L., N.L., L.L., P.L., X. Jiang, W.W., X.W., Y.L., M.C., J.L., X.Z., J.H., L.W., Y.D., Y.X., J.-D.J., B.H., S.S.).,State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, CAMS&PUMC, Beijing, China (X.H., N.L., J.-D.J.)
| | - Lunming Liu (刘伦铭)
- NHC Key Laboratory of Biotechnology of Antibiotics, National Center for New Microbial Drug Screening, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS&PUMC), Beijing, China (Y.X., C.L., X.H., X. Jia, Y.L., C.L., N.L., L.L., P.L., X. Jiang, W.W., X.W., Y.L., M.C., J.L., X.Z., J.H., L.W., Y.D., Y.X., J.-D.J., B.H., S.S.)
| | - Peng Liu (刘鹏)
- NHC Key Laboratory of Biotechnology of Antibiotics, National Center for New Microbial Drug Screening, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS&PUMC), Beijing, China (Y.X., C.L., X.H., X. Jia, Y.L., C.L., N.L., L.L., P.L., X. Jiang, W.W., X.W., Y.L., M.C., J.L., X.Z., J.H., L.W., Y.D., Y.X., J.-D.J., B.H., S.S.)
| | - Xinhai Jiang (姜新海)
- NHC Key Laboratory of Biotechnology of Antibiotics, National Center for New Microbial Drug Screening, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS&PUMC), Beijing, China (Y.X., C.L., X.H., X. Jia, Y.L., C.L., N.L., L.L., P.L., X. Jiang, W.W., X.W., Y.L., M.C., J.L., X.Z., J.H., L.W., Y.D., Y.X., J.-D.J., B.H., S.S.)
| | - Weizhi Wang (王伟志)
- NHC Key Laboratory of Biotechnology of Antibiotics, National Center for New Microbial Drug Screening, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS&PUMC), Beijing, China (Y.X., C.L., X.H., X. Jia, Y.L., C.L., N.L., L.L., P.L., X. Jiang, W.W., X.W., Y.L., M.C., J.L., X.Z., J.H., L.W., Y.D., Y.X., J.-D.J., B.H., S.S.)
| | - Xiao Wang (王潇)
- NHC Key Laboratory of Biotechnology of Antibiotics, National Center for New Microbial Drug Screening, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS&PUMC), Beijing, China (Y.X., C.L., X.H., X. Jia, Y.L., C.L., N.L., L.L., P.L., X. Jiang, W.W., X.W., Y.L., M.C., J.L., X.Z., J.H., L.W., Y.D., Y.X., J.-D.J., B.H., S.S.)
| | - Yining Li (李依宁)
- NHC Key Laboratory of Biotechnology of Antibiotics, National Center for New Microbial Drug Screening, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS&PUMC), Beijing, China (Y.X., C.L., X.H., X. Jia, Y.L., C.L., N.L., L.L., P.L., X. Jiang, W.W., X.W., Y.L., M.C., J.L., X.Z., J.H., L.W., Y.D., Y.X., J.-D.J., B.H., S.S.)
| | - Mingzhu Chen (陈明珠)
- NHC Key Laboratory of Biotechnology of Antibiotics, National Center for New Microbial Drug Screening, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS&PUMC), Beijing, China (Y.X., C.L., X.H., X. Jia, Y.L., C.L., N.L., L.L., P.L., X. Jiang, W.W., X.W., Y.L., M.C., J.L., X.Z., J.H., L.W., Y.D., Y.X., J.-D.J., B.H., S.S.)
| | - Jinque Luo (罗金雀)
- NHC Key Laboratory of Biotechnology of Antibiotics, National Center for New Microbial Drug Screening, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS&PUMC), Beijing, China (Y.X., C.L., X.H., X. Jia, Y.L., C.L., N.L., L.L., P.L., X. Jiang, W.W., X.W., Y.L., M.C., J.L., X.Z., J.H., L.W., Y.D., Y.X., J.-D.J., B.H., S.S.)
| | - Xuan Zuo (左璇)
- NHC Key Laboratory of Biotechnology of Antibiotics, National Center for New Microbial Drug Screening, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS&PUMC), Beijing, China (Y.X., C.L., X.H., X. Jia, Y.L., C.L., N.L., L.L., P.L., X. Jiang, W.W., X.W., Y.L., M.C., J.L., X.Z., J.H., L.W., Y.D., Y.X., J.-D.J., B.H., S.S.)
| | - Jiangxue Han (韩江雪)
- NHC Key Laboratory of Biotechnology of Antibiotics, National Center for New Microbial Drug Screening, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS&PUMC), Beijing, China (Y.X., C.L., X.H., X. Jia, Y.L., C.L., N.L., L.L., P.L., X. Jiang, W.W., X.W., Y.L., M.C., J.L., X.Z., J.H., L.W., Y.D., Y.X., J.-D.J., B.H., S.S.)
| | - Li Wang (王丽)
- NHC Key Laboratory of Biotechnology of Antibiotics, National Center for New Microbial Drug Screening, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS&PUMC), Beijing, China (Y.X., C.L., X.H., X. Jia, Y.L., C.L., N.L., L.L., P.L., X. Jiang, W.W., X.W., Y.L., M.C., J.L., X.Z., J.H., L.W., Y.D., Y.X., J.-D.J., B.H., S.S.)
| | - Yu Du (杜郁)
- NHC Key Laboratory of Biotechnology of Antibiotics, National Center for New Microbial Drug Screening, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS&PUMC), Beijing, China (Y.X., C.L., X.H., X. Jia, Y.L., C.L., N.L., L.L., P.L., X. Jiang, W.W., X.W., Y.L., M.C., J.L., X.Z., J.H., L.W., Y.D., Y.X., J.-D.J., B.H., S.S.)
| | - Yang Xu (徐扬)
- NHC Key Laboratory of Biotechnology of Antibiotics, National Center for New Microbial Drug Screening, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS&PUMC), Beijing, China (Y.X., C.L., X.H., X. Jia, Y.L., C.L., N.L., L.L., P.L., X. Jiang, W.W., X.W., Y.L., M.C., J.L., X.Z., J.H., L.W., Y.D., Y.X., J.-D.J., B.H., S.S.)
| | - Jian-Dong Jiang (蒋建东)
- NHC Key Laboratory of Biotechnology of Antibiotics, National Center for New Microbial Drug Screening, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS&PUMC), Beijing, China (Y.X., C.L., X.H., X. Jia, Y.L., C.L., N.L., L.L., P.L., X. Jiang, W.W., X.W., Y.L., M.C., J.L., X.Z., J.H., L.W., Y.D., Y.X., J.-D.J., B.H., S.S.).,State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, CAMS&PUMC, Beijing, China (X.H., N.L., J.-D.J.)
| | - Bin Hong (洪斌)
- NHC Key Laboratory of Biotechnology of Antibiotics, National Center for New Microbial Drug Screening, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS&PUMC), Beijing, China (Y.X., C.L., X.H., X. Jia, Y.L., C.L., N.L., L.L., P.L., X. Jiang, W.W., X.W., Y.L., M.C., J.L., X.Z., J.H., L.W., Y.D., Y.X., J.-D.J., B.H., S.S.)
| | - Shuyi Si (司书毅)
- NHC Key Laboratory of Biotechnology of Antibiotics, National Center for New Microbial Drug Screening, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS&PUMC), Beijing, China (Y.X., C.L., X.H., X. Jia, Y.L., C.L., N.L., L.L., P.L., X. Jiang, W.W., X.W., Y.L., M.C., J.L., X.Z., J.H., L.W., Y.D., Y.X., J.-D.J., B.H., S.S.)
| |
Collapse
|
25
|
Ouweneel AB, Zhao Y, Calpe-Berdiel L, Lammers B, Hoekstra M, Van Berkel TJC, Van Eck M. Impact of bone marrow ATP-binding cassette transporter A1 deficiency on atherogenesis is independent of the presence of the low-density lipoprotein receptor. Atherosclerosis 2021; 319:79-85. [PMID: 33494008 DOI: 10.1016/j.atherosclerosis.2021.01.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 12/03/2020] [Accepted: 01/05/2021] [Indexed: 01/20/2023]
Abstract
BACKGROUND AND AIMS There is extensive evidence from bone marrow transplantation studies that hematopoietic ATP binding cassette A1 (Abca1) is atheroprotective in low-density lipoprotein receptor (Ldlr) deficient mice. In contrast, studies using lysosyme M promoter-driven deletion of Abca1 in Ldlr deficient mice failed to show similar effects. It was hypothesized that the discrepancy between these studies might be due to the presence of Ldlr in bone marrow-derived cells in the transplantation model. In this study, we aim to determine the contribution of Ldlr to the atheroprotective effect of hematopoietic Abca1 in the murine bone marrow transplantation model. METHODS Wild-type, Ldlr-/-, Abca1-/-, and Abca1-/-Ldlr-/- bone marrow was transplanted into hypercholesterolemic Ldlr-/- mice. RESULTS Bone marrow Lldr deficiency did not influence the effects of Abca1 on macrophage cholesterol efflux, foam cell formation, monocytosis or plasma cholesterol. Ldlr deficiency did reduce circulating and peritoneal lymphocyte counts, albeit only in animals lacking Abca1 in bone marrow-derived cells. Importantly, the effects of Abca1 deficiency on atherosclerosis susceptibility were unaltered by the presence or absence of Ldlr. Bone marrow Ldlr deficiency did lead to marginally but consistently decreased atherosclerosis, regardless of Abca1 deficiency. Thus, Ldlr expression on bone marrow-derived cells does, to a minimal extent, influence atherosclerotic lesion development, albeit independent of Abca1. CONCLUSIONS This study provides novel insight into the relative impact of Ldlr and Abca1 in bone marrow-derived cells on macrophage foam cell formation and atherosclerosis development in vivo. We have shown that Ldlr and Abca1 differentially and independently influence atherosclerosis development in a murine bone marrow transplantation model of atherosclerosis.
Collapse
Affiliation(s)
- Amber B Ouweneel
- Division of BioTherapeutics, Leiden Academic Center for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, the Netherlands.
| | - Ying Zhao
- Division of BioTherapeutics, Leiden Academic Center for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, the Netherlands
| | - Laura Calpe-Berdiel
- Division of BioTherapeutics, Leiden Academic Center for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, the Netherlands
| | - Bart Lammers
- Division of BioTherapeutics, Leiden Academic Center for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, the Netherlands
| | - Menno Hoekstra
- Division of BioTherapeutics, Leiden Academic Center for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, the Netherlands
| | - Theo J C Van Berkel
- Division of BioTherapeutics, Leiden Academic Center for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, the Netherlands
| | - Miranda Van Eck
- Division of BioTherapeutics, Leiden Academic Center for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, the Netherlands
| |
Collapse
|
26
|
Rafiei A, Ferns GA, Ahmadi R, Khaledifar A, Rahimzadeh-Fallah T, Mohmmad-Rezaei M, Emami S, Bagheri N. Expression levels of miR-27a, miR-329, ABCA1, and ABCG1 genes in peripheral blood mononuclear cells and their correlation with serum levels of oxidative stress and hs-CRP in the patients with coronary artery disease. IUBMB Life 2020; 73:223-237. [PMID: 33263223 DOI: 10.1002/iub.2421] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 10/27/2020] [Accepted: 11/19/2020] [Indexed: 12/17/2022]
Abstract
Atherosclerosis is a chronic inflammatory disease with high mortality worldwide. The reverse cholesterol transport pathway in macrophage plays an important role in the pathogenesis of coronary artery disease (CAD) and is strongly controlled by regulatory factors. The microRNAs can promote or prevent the formation of atherosclerotic lesions by post-transcriptional regulation of vital genes in this pathway. Therefore, this study was conducted to investigate the relationship between the expression levels of miR-27a, miR-329, ABCA1, and ABCG1 genes and serum levels of hs-CRP, ox-LDL, and indices of oxidative stress in the patients with established CAD and controls. A total of 84 subjects (42 patients with CAD and 42 controls) were included in this study. Expression levels of miR-27a-3p, miR-329-3p, ABCA1, and ABCG1 genes in the peripheral blood mononuclear cells (PBMCs) and serum concentration of hs-CRP and ox-LDL were measured by real time-PCR and ELISA, respectively. Also, oxidative stress parameters in the serum were evaluated by ferric-reducing antioxidant power (FRAP) and malondialdehyde (MDA) assays. ABCA1 and ABCG1 gene expression in PBMC and serum concentration of FRAP were significantly lower in the CAD group compared to the control group. Expression levels of miR-27a and miR-329 and serum levels of hs-CRP, ox-LDL, and MDA were significantly higher in the CAD group compared to the control group. Serum levels of hs-CRP, ox-LDL, and expression level of miR-27a have inversely related to ABCA1 and ABCG1 gene expression in all the subjects. An increase in the expression levels of miR-27a and miR-329 may lead to the progression of atherosclerosis plaque by downregulating the expression of ABCA1 and ABCG1 genes.
Collapse
Affiliation(s)
- Ali Rafiei
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Gordon A Ferns
- Division of Medical Education, Brighton & Sussex Medical School, Brighton, UK
| | - Reza Ahmadi
- Clinical Biochemistry Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Arsalan Khaledifar
- Department of Cardiology, School of Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Tina Rahimzadeh-Fallah
- Medical Biotechnology Research Center, School of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Mina Mohmmad-Rezaei
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Shohreh Emami
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Nader Bagheri
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
27
|
Galle-Treger L, Moreau M, Ballaire R, Poupel L, Huby T, Sasso E, Troise F, Poti F, Lesnik P, Le Goff W, Gautier EL, Huby T. Targeted invalidation of SR-B1 in macrophages reduces macrophage apoptosis and accelerates atherosclerosis. Cardiovasc Res 2020; 116:554-565. [PMID: 31119270 DOI: 10.1093/cvr/cvz138] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 01/30/2019] [Accepted: 05/16/2019] [Indexed: 12/15/2022] Open
Abstract
AIMS SR-B1 is a cholesterol transporter that exerts anti-atherogenic properties in liver and peripheral tissues in mice. Bone marrow (BM) transfer studies suggested an atheroprotective role in cells of haematopoietic origin. Here, we addressed the specific contribution of SR-B1 in the monocyte/macrophage. METHODS AND RESULTS We generated mice deficient for SR-B1 in monocytes/macrophages (Lysm-Cre × SR-B1f/f) and transplanted their BM into Ldlr-/- mice. Fed a cholesterol-rich diet, these mice displayed accelerated aortic atherosclerosis characterized by larger macrophage-rich areas and decreased macrophage apoptosis compared with SR-B1f/f transplanted controls. These findings were reproduced in BM transfer studies using another atherogenic mouse recipient (SR-B1 KOliver × Cholesteryl Ester Transfer Protein). Haematopoietic reconstitution with SR-B1-/- BM conducted in parallel generated similar results to those obtained with Lysm-Cre × SR-B1f/f BM; thus suggesting that among haematopoietic-derived cells, SR-B1 exerts its atheroprotective role primarily in monocytes/macrophages. Consistent with our in vivo data, free cholesterol (FC)-induced apoptosis of macrophages was diminished in the absence of SR-B1. This effect could not be attributed to differential cellular cholesterol loading. However, we observed that expression of apoptosis inhibitor of macrophage (AIM) was induced in SR-B1-deficient macrophages, and notably upon FC-loading. Furthermore, we demonstrated that macrophages were protected from FC-induced apoptosis by AIM. Finally, AIM protein was found more present within the macrophage-rich area of the atherosclerotic lesions of SR-B1-deficient macrophages than controls. CONCLUSION Our findings suggest that macrophage SR-B1 plays a role in plaque growth by controlling macrophage apoptosis in an AIM-dependent manner.
Collapse
Affiliation(s)
| | - Martine Moreau
- Sorbonne Université, INSERM, UMR_S 1166 ICAN, F-75013, Paris, France
| | | | - Lucie Poupel
- Sorbonne Université, INSERM, UMR_S 1166 ICAN, F-75013, Paris, France
| | - Thomas Huby
- Sorbonne Université, INSERM, UMR_S 1166 ICAN, F-75013, Paris, France
| | - Emanuele Sasso
- Ceinge Biotecnologie Avanzate S.C.R.L, Via Gaetano Salvatore 486, 80145, Napoli, Italy.,Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, 80131, Napoli, Italy
| | - Fulvia Troise
- Ceinge Biotecnologie Avanzate S.C.R.L, Via Gaetano Salvatore 486, 80145, Napoli, Italy
| | - Francesco Poti
- Department of Medicine and Surgery, Unit of Neurosciences, University of Parma, Parma, Italy
| | - Philippe Lesnik
- Sorbonne Université, INSERM, UMR_S 1166 ICAN, F-75013, Paris, France
| | - Wilfried Le Goff
- Sorbonne Université, INSERM, UMR_S 1166 ICAN, F-75013, Paris, France
| | | | - Thierry Huby
- Sorbonne Université, INSERM, UMR_S 1166 ICAN, F-75013, Paris, France
| |
Collapse
|
28
|
Castaño D, Rattanasopa C, Monteiro-Cardoso VF, Corlianò M, Liu Y, Zhong S, Rusu M, Liehn EA, Singaraja RR. Lipid efflux mechanisms, relation to disease and potential therapeutic aspects. Adv Drug Deliv Rev 2020; 159:54-93. [PMID: 32423566 DOI: 10.1016/j.addr.2020.04.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 04/29/2020] [Accepted: 04/30/2020] [Indexed: 02/06/2023]
Abstract
Lipids are hydrophobic and amphiphilic molecules involved in diverse functions such as membrane structure, energy metabolism, immunity, and signaling. However, altered intra-cellular lipid levels or composition can lead to metabolic and inflammatory dysfunction, as well as lipotoxicity. Thus, intra-cellular lipid homeostasis is tightly regulated by multiple mechanisms. Since most peripheral cells do not catabolize cholesterol, efflux (extra-cellular transport) of cholesterol is vital for lipid homeostasis. Defective efflux contributes to atherosclerotic plaque development, impaired β-cell insulin secretion, and neuropathology. Of these, defective lipid efflux in macrophages in the arterial walls leading to foam cell and atherosclerotic plaque formation has been the most well studied, likely because a leading global cause of death is cardiovascular disease. Circulating high density lipoprotein particles play critical roles as acceptors of effluxed cellular lipids, suggesting their importance in disease etiology. We review here mechanisms and pathways that modulate lipid efflux, the role of lipid efflux in disease etiology, and therapeutic options aimed at modulating this critical process.
Collapse
|
29
|
Morgan PK, Fang L, Lancaster GI, Murphy AJ. Hematopoiesis is regulated by cholesterol efflux pathways and lipid rafts: connections with cardiovascular diseases. J Lipid Res 2020; 61:667-675. [PMID: 31471447 PMCID: PMC7193969 DOI: 10.1194/jlr.tr119000267] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 08/08/2019] [Indexed: 12/11/2022] Open
Abstract
Lipid rafts are highly ordered regions of the plasma membrane that are enriched in cholesterol and sphingolipids and play important roles in many cells. In hematopoietic stem and progenitor cells (HSPCs), lipid rafts house receptors critical for normal hematopoiesis. Lipid rafts also can bind and sequester kinases that induce negative feedback pathways to limit proliferative cytokine receptor cycling back to the cell membrane. Modulation of lipid rafts occurs through an array of mechanisms, with optimal cholesterol efflux one of the major regulators. As such, cholesterol homeostasis also regulates hematopoiesis. Increased lipid raft content, which occurs in response to changes in cholesterol efflux in the membrane, can result in prolonged receptor occupancy in the cell membrane and enhanced signaling. In addition, certain diseases, like diabetes, may contribute to lipid raft formation and affect cholesterol retention in rafts. In this review, we explore the role of lipid raft-related mechanisms in hematopoiesis and CVD (specifically, atherosclerosis) and discuss how defective cholesterol efflux pathways in HSPCs contribute to expansion of lipid rafts, thereby promoting myelopoiesis and thrombopoiesis. We also discuss the utility of cholesterol acceptors in contributing to lipid raft regulation and disruption, and highlight the potential to manipulate these pathways for therapeutic gain in CVD as well as other disorders with aberrant hematopoiesis.jlr;61/5/667/F1F1f1.
Collapse
Affiliation(s)
- Pooranee K Morgan
- Division of Immunometabolism,Baker Heart and Diabetes Institute, Melbourne, Australia; School of Life Sciences,La Trobe University, Bundoora, Australia
| | - Longhou Fang
- Center for Cardiovascular Regeneration,Houston Methodist, Houston, TX
| | - Graeme I Lancaster
- Division of Immunometabolism,Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Andrew J Murphy
- Division of Immunometabolism,Baker Heart and Diabetes Institute, Melbourne, Australia; School of Life Sciences,La Trobe University, Bundoora, Australia
| |
Collapse
|
30
|
Stamatikos A, Knight E, Vojtech L, Bi L, Wacker BK, Tang C, Dichek DA. Exosome-Mediated Transfer of Anti-miR-33a-5p from Transduced Endothelial Cells Enhances Macrophage and Vascular Smooth Muscle Cell Cholesterol Efflux. Hum Gene Ther 2020; 31:219-232. [PMID: 31842627 DOI: 10.1089/hum.2019.245] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Atherosclerosis is a disease of large- and medium-sized arteries that is caused by cholesterol accumulation in arterial intimal cells, including macrophages and smooth muscle cells (SMC). Cholesterol accumulation in these cells can be prevented or reversed in preclinical models-and atherosclerosis reduced-by transgenesis that increases expression of molecules that control cholesterol efflux, including apolipoprotein AI (apoAI) and ATP-binding cassette subfamily A, member 1 (ABCA1). In a previous work, we showed that transduction of arterial endothelial cells (EC)-with a helper-dependent adenovirus (HDAd) expressing apoAI-enhanced EC cholesterol efflux in vitro and decreased atherosclerosis in vivo. Similarly, overexpression of ABCA1 in cultured EC increased cholesterol efflux and decreased inflammatory gene expression. These EC-targeted gene-therapy strategies might be improved by concurrent upregulation of cholesterol-efflux pathways in other intimal cell types. Here, we report modification of this strategy to enable delivery of therapeutic nucleic acids to cells of the sub-endothelium. We constructed an HDAd (HDAdXMoAntimiR33a5p) that expresses an antagomiR directed at miR-33a-5p (a microRNA that suppresses cholesterol efflux by silencing ABCA1). HDAdXMoAntimiR33a5p contains a sequence motif that enhances uptake of anti-miR-33a-5p into exosomes. Cultured EC release exosomes containing small RNA, including miR-33a-5p. After transduction with HDAdXMoAntimiR33a5p, EC-derived exosomes containing anti-miR-33a-5p accumulate in conditioned medium (CM). When this CM is added to macrophages or SMC, anti-miR-33a-5p is detected in these target cells. Exosome-mediated transfer of anti-miR-33a-5p reduces miR-33a-5p by ∼65-80%, increases ABCA1 protein by 1.6-2.2-fold, and increases apoAI-mediated cholesterol efflux by 1.4-1.6-fold (all p ≤ 0.01). These effects were absent in macrophages and SMC incubated in exosome-depleted CM. EC transduced with HDAdXMoAntimiR33a5p release exosomes that can transfer anti-miR-33a-5p to other intimal cell types, upregulating cholesterol efflux from these cells. This strategy provides a platform for genetic modification of intimal and medial cells, using a vector that transduces only EC.
Collapse
Affiliation(s)
- Alexis Stamatikos
- Department of Medicine, University of Washington, Seattle, Washington
| | - Ethan Knight
- Department of Medicine, University of Washington, Seattle, Washington
| | - Lucia Vojtech
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington
| | - Lianxiang Bi
- Department of Medicine, University of Washington, Seattle, Washington
| | - Bradley K Wacker
- Department of Medicine, University of Washington, Seattle, Washington
| | - Chongren Tang
- Department of Medicine, University of Washington, Seattle, Washington
| | - David A Dichek
- Department of Medicine, University of Washington, Seattle, Washington
| |
Collapse
|
31
|
ABC Transporters, Cholesterol Efflux, and Implications for Cardiovascular Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1276:67-83. [DOI: 10.1007/978-981-15-6082-8_6] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
32
|
Huesca-Gómez C, Torres-Paz YE, Martínez-Alvarado R, Fuentevilla-Álvarez G, Del Valle-Mondragón L, Torres-Tamayo M, Soto ME, Gamboa R. Association between the transporters ABCA1/G1 and the expression of miR-33a/144 and the carotid intima media thickness in patients with arterial hypertension. Mol Biol Rep 2019; 47:1321-1329. [PMID: 31853766 DOI: 10.1007/s11033-019-05229-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 12/07/2019] [Indexed: 12/22/2022]
Abstract
ATP-binding cassette membrane transporters (ABC), functions as an outflow facilitator of phospholipids and cellular cholesterol, playing an important role in the development of atherosclerosis and arterial hypertension. ABC's transporters could post-transcriptionally regulated by miRs. Evaluate the association in the transporters ABCA1 and ABCG1 with the expression of miR-33a and miR-144 and the carotid intima media thickness (cIMT) in patients with essential arterial hypertension. The miR-33a-5p, miR-144-3p and mRNA ABCA1 and ABCG1 expression in monocytes from Mexican hypertensive patients were examined by RT-PCR. The miR-33a and miR-144 expression in monocytes and mRNA ABCA1 and ABCG1 from Mexican hypertensive patients were examined by RT-PCR. This study involved 84 subjects (42 normotensive subjects and 42 patients with essential hypertension). Our study revealed that miR-33a expression (p = 0.001) and miR-144 (p = 0.985) were up-regulated, meanwhile, ABCA1 and ABCG1 transporters were down-regulated (p = 0.007 and p = 0.550 respectively) in hypertensive patients compared with the control group. The trend remains for miR33a/ABCA1 in presence of cIMT. Moreover, an inverse correlation was found with the expression levels of ABCA1 and ABCG1 as well as in HDL-C with miR-33a and miR-144. Our results showed an increase in the expression of miR-33a and miR-144 and an inverse correlation in their target ABCA1 and ABCG1; it may be associated with essential arterial hypertension in patients with cIMT and as consequence for atheromatous plaque.
Collapse
Affiliation(s)
- Claudia Huesca-Gómez
- Department of Physiology, Instituto Nacional de Cardiología "Ignacio Chávez", Juan Badiano No 1, Col. Sección XVI, C.P. 14080, Mexico City, Mexico
| | - Yazmín Estela Torres-Paz
- Department of Physiology, Instituto Nacional de Cardiología "Ignacio Chávez", Juan Badiano No 1, Col. Sección XVI, C.P. 14080, Mexico City, Mexico
| | - Rocío Martínez-Alvarado
- Department of Endocrinology, Instituto Nacional de Cardiología "Ignacio Chávez", Mexico City, Mexico
| | - Giovanny Fuentevilla-Álvarez
- Department of Physiology, Instituto Nacional de Cardiología "Ignacio Chávez", Juan Badiano No 1, Col. Sección XVI, C.P. 14080, Mexico City, Mexico
| | | | - Margarita Torres-Tamayo
- Department of Endocrinology, Instituto Nacional de Cardiología "Ignacio Chávez", Mexico City, Mexico
| | - Ma Elena Soto
- Department of Immunology, Instituto Nacional de Cardiología "Ignacio Chávez", Mexico City, Mexico
| | - Ricardo Gamboa
- Department of Physiology, Instituto Nacional de Cardiología "Ignacio Chávez", Juan Badiano No 1, Col. Sección XVI, C.P. 14080, Mexico City, Mexico.
| |
Collapse
|
33
|
Hu W, Jia Y, Kang Q, Peng H, Ma H, Zhang S, Hiromori Y, Kimura T, Nakanishi T, Zheng L, Qiu Y, Zhang Z, Wan Y, Hu J. Screening of House Dust from Chinese Homes for Chemicals with Liver X Receptors Binding Activities and Characterization of Atherosclerotic Activity Using an in Vitro Macrophage Cell Line and ApoE-/- Mice. ENVIRONMENTAL HEALTH PERSPECTIVES 2019; 127:117003. [PMID: 31724879 PMCID: PMC6927504 DOI: 10.1289/ehp5039] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 10/21/2019] [Accepted: 10/23/2019] [Indexed: 05/18/2023]
Abstract
BACKGROUND Atherosclerotic cardiovascular disease has become the leading cause of death worldwide, and environmental pollutants are increasingly recognized as risk factors for atherosclerosis. Liver X receptors (LXRs) play a central role in atherosclerosis; however, LXR activity of organic pollutants and associated potential risk of atherosclerosis have not yet been characterized. OBJECTIVES This study aimed to explore whether LXR-antagonistic chemicals are present in indoor house dust and, if so, to characterize this activity in relation to changes in macrophages in vitro and cardiovascular disease indicators in vivo in an atherosclerosis ApoE-/- mouse model. METHODS We used a His-LXRα-pull-down assay and a nontarget high-resolution mass spectrometry method to screen house dust collected from Chinese homes for LXRα- and LXRβ-antagonist activity. A chemical identified in this manner was assessed for its ability to induce cholesterol efflux and foam cell formation in RAW264.7 macrophages, to down-regulate the expression of two LXR-dependent genes, ABCA1 and ABCG1, and finally to induce atherosclerotic lesions in vivo using an ApoE-/- mouse model. RESULTS We identified the flame retardants triphenyl phosphate (TPHP) and 2-ethylhexyl diphenyl phosphate (EHDPP) in house dust samples and demonstrated their ability to antagonize LXRs. The potency of TPHP was similar to that of the LXR-antagonist SR9238. TPHP could also inhibit cholesterol efflux and promote foam cell formation in RAW264.7 macrophages and mouse peritoneal macrophages and significantly promoted atherosclerotic lesion formation in the ApoE-/- mouse model. CONCLUSIONS We found LXR-antagonist chemicals in environmental samples of indoor dust from Chinese homes. One of the chemicals, TPHP, was able to promote the development of atherosclerotic lesions in the ApoE-/- mouse model. These results highlight the need to assess the LXR-antagonist activities of pollutants in future environmental management programs. https://doi.org/10.1289/EHP5039.
Collapse
Affiliation(s)
- Wenxin Hu
- Laboratory for Earth Surface Processes, College of Urban and Environmental Sciences, Peking University, Beijing, China
| | - Yingting Jia
- Laboratory for Earth Surface Processes, College of Urban and Environmental Sciences, Peking University, Beijing, China
| | - Qiyue Kang
- Laboratory for Earth Surface Processes, College of Urban and Environmental Sciences, Peking University, Beijing, China
| | - Hui Peng
- Department of Chemistry, University of Toronto, Toronto, Ontario, Canada
| | - Haojia Ma
- Laboratory for Earth Surface Processes, College of Urban and Environmental Sciences, Peking University, Beijing, China
| | - Shiyi Zhang
- Laboratory for Earth Surface Processes, College of Urban and Environmental Sciences, Peking University, Beijing, China
| | - Youhei Hiromori
- Laboratory of Hygienic Chemistry and Molecular Toxicology, Gifu Pharmaceutical University, Gifu, Gifu, Japan
- Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka, Mie, Japan
| | - Tomoki Kimura
- Department of Life Science, Faculty of Science and Engineering, Setsunan University, Ikedanakamachi, Neyagawa, Japan
| | - Tsuyoshi Nakanishi
- Laboratory of Hygienic Chemistry and Molecular Toxicology, Gifu Pharmaceutical University, Gifu, Gifu, Japan
| | - Lemin Zheng
- School of Basic Medical Sciences, Peking University Health Science Center, Peking University, Beijing, China
| | - Yifu Qiu
- Institute of Molecular Medicine, Peking University, Beijing, China
| | - Zhaobin Zhang
- Laboratory for Earth Surface Processes, College of Urban and Environmental Sciences, Peking University, Beijing, China
| | - Yi Wan
- Laboratory for Earth Surface Processes, College of Urban and Environmental Sciences, Peking University, Beijing, China
| | - Jianying Hu
- Laboratory for Earth Surface Processes, College of Urban and Environmental Sciences, Peking University, Beijing, China
| |
Collapse
|
34
|
Nguyen MA, Wyatt H, Susser L, Geoffrion M, Rasheed A, Duchez AC, Cottee ML, Afolayan E, Farah E, Kahiel Z, Côté M, Gadde S, Rayner KJ. Delivery of MicroRNAs by Chitosan Nanoparticles to Functionally Alter Macrophage Cholesterol Efflux in Vitro and in Vivo. ACS NANO 2019; 13:6491-6505. [PMID: 31125197 DOI: 10.1021/acsnano.8b09679] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
The prevention and treatment of cardiovascular diseases (CVD) has largely focused on lowering circulating LDL cholesterol, yet a significant burden of atherosclerotic disease remains even when LDL is low. Recently, microRNAs (miRNAs) have emerged as exciting therapeutic targets for cardiovascular disease. miRNAs are small noncoding RNAs that post-transcriptionally regulate gene expression by degradation or translational inhibition of target mRNAs. A number of miRNAs have been found to modulate all stages of atherosclerosis, particularly those that promote the efflux of excess cholesterol from lipid-laden macrophages in the vessel wall to the liver. However, one of the major challenges of miRNA-based therapy is to achieve tissue-specific, efficient, and safe delivery of miRNAs in vivo. We sought to develop chitosan nanoparticles (chNPs) that can deliver functional miRNA mimics to macrophages and to determine if these nanoparticles can alter cholesterol efflux and reverse cholesterol transport in vivo. We developed chNPs with a size range of 150-200 nm via the ionic gelation method using tripolyphosphate (TPP) as a cross-linker. In this method, negatively charged miRNAs were encapsulated in the nanoparticles by ionic interactions with polymeric components. We then optimized the efficiency of intracellular delivery of different formulations of chitosan/TPP/miRNA to mouse macrophages. Using a well-defined miRNA with roles in macrophage cholesterol metabolism, we tested whether chNPs could deliver functional miRNAs to macrophages. We find chNPs can transfer exogenous miR-33 to naïve macrophages and reduce the expression of ABCA1, a potent miR-33 target gene, both in vitro and in vivo, confirming that miRNAs delivered via nanoparticles can escape the endosomal system and function in the RISC complex. Because miR-33 and ABCA1 play a key role in regulating the efflux of cholesterol from macrophages, we also confirmed that macrophages treated with miR-33-loaded chNPs exhibited reduced cholesterol efflux to apolipoprotein A1, further confirming functional delivery of the miRNA. In vivo, mice treated with miR33-chNPs showed decreased reverse cholesterol transport (RCT) to the plasma, liver, and feces. In contrast, when efflux-promoting miRNAs were delivered via chNPs, ABCA1 expression and cholesterol efflux into the RCT pathway were improved. Over all, miRNAs can be efficiently delivered to macrophages via nanoparticles, where they can function to regulate ABCA1 expression and cholesterol efflux, suggesting that these miRNA nanoparticles can be used in vivo to target atherosclerotic lesions.
Collapse
Affiliation(s)
- My-Anh Nguyen
- University of Ottawa Heart Institute , Ottawa , Ontario K1Y 4W7 , Canada
| | - Hailey Wyatt
- University of Ottawa Heart Institute , Ottawa , Ontario K1Y 4W7 , Canada
| | - Leah Susser
- University of Ottawa Heart Institute , Ottawa , Ontario K1Y 4W7 , Canada
| | - Michele Geoffrion
- University of Ottawa Heart Institute , Ottawa , Ontario K1Y 4W7 , Canada
| | - Adil Rasheed
- University of Ottawa Heart Institute , Ottawa , Ontario K1Y 4W7 , Canada
| | - Anne-Claire Duchez
- University of Ottawa Heart Institute , Ottawa , Ontario K1Y 4W7 , Canada
| | - Mary Lynn Cottee
- University of Ottawa Heart Institute , Ottawa , Ontario K1Y 4W7 , Canada
| | - Esther Afolayan
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine , University of Ottawa , Ottawa , Ontario K1H 8M5 , Canada
| | - Eliya Farah
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine , University of Ottawa , Ottawa , Ontario K1H 8M5 , Canada
| | - Zaina Kahiel
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine , University of Ottawa , Ottawa , Ontario K1H 8M5 , Canada
| | - Marceline Côté
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine , University of Ottawa , Ottawa , Ontario K1H 8M5 , Canada
| | - Suresh Gadde
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine , University of Ottawa , Ottawa , Ontario K1H 8M5 , Canada
| | - Katey J Rayner
- University of Ottawa Heart Institute , Ottawa , Ontario K1Y 4W7 , Canada
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine , University of Ottawa , Ottawa , Ontario K1H 8M5 , Canada
| |
Collapse
|
35
|
Yu XH, Zhang DW, Zheng XL, Tang CK. Cholesterol transport system: An integrated cholesterol transport model involved in atherosclerosis. Prog Lipid Res 2018; 73:65-91. [PMID: 30528667 DOI: 10.1016/j.plipres.2018.12.002] [Citation(s) in RCA: 136] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 10/30/2018] [Accepted: 12/01/2018] [Indexed: 02/07/2023]
Abstract
Atherosclerosis, the pathological basis of most cardiovascular disease (CVD), is closely associated with cholesterol accumulation in the arterial intima. Excessive cholesterol is removed by the reverse cholesterol transport (RCT) pathway, representing a major antiatherogenic mechanism. In addition to the RCT, other pathways are required for maintaining the whole-body cholesterol homeostasis. Thus, we propose a working model of integrated cholesterol transport, termed the cholesterol transport system (CTS), to describe body cholesterol metabolism. The novel model not only involves the classical view of RCT but also contains other steps, such as cholesterol absorption in the small intestine, low-density lipoprotein uptake by the liver, and transintestinal cholesterol excretion. Extensive studies have shown that dysfunctional CTS is one of the major causes for hypercholesterolemia and atherosclerosis. Currently, several drugs are available to improve the CTS efficiently. There are also several therapeutic approaches that have entered into clinical trials and shown considerable promise for decreasing the risk of CVD. In recent years, a variety of novel findings reveal the molecular mechanisms for the CTS and its role in the development of atherosclerosis, thereby providing novel insights into the understanding of whole-body cholesterol transport and metabolism. In this review, we summarize the latest advances in this area with an emphasis on the therapeutic potential of targeting the CTS in CVD patients.
Collapse
Affiliation(s)
- Xiao-Hua Yu
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Medical Research Experiment Center, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, China
| | - Da-Wei Zhang
- Department of Pediatrics and Group on the Molecular and Cell Biology of Lipids, University of Alberta, Alberta, Canada
| | - Xi-Long Zheng
- Department of Biochemistry and Molecular Biology, Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, University of Calgary, Health Sciences Center, 3330 Hospital Dr NW, Calgary, Alberta T2N 4N1, Canada
| | - Chao-Ke Tang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Medical Research Experiment Center, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, China.
| |
Collapse
|
36
|
Estrada-Luna D, Ortiz-Rodriguez MA, Medina-Briseño L, Carreón-Torres E, Izquierdo-Vega JA, Sharma A, Cancino-Díaz JC, Pérez-Méndez O, Belefant-Miller H, Betanzos-Cabrera G. Current Therapies Focused on High-Density Lipoproteins Associated with Cardiovascular Disease. Molecules 2018; 23:molecules23112730. [PMID: 30360466 PMCID: PMC6278283 DOI: 10.3390/molecules23112730] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 10/20/2018] [Accepted: 10/21/2018] [Indexed: 02/06/2023] Open
Abstract
High-density lipoproteins (HDL) comprise a heterogeneous family of lipoprotein particles divided into subclasses that are determined by density, size and surface charge as well as protein composition. Epidemiological studies have suggested an inverse correlation between High-density lipoprotein-cholesterol (HDL-C) levels and the risk of cardiovascular diseases and atherosclerosis. HDLs promote reverse cholesterol transport (RCT) and have several atheroprotective functions such as anti-inflammation, anti-thrombosis, and anti-oxidation. HDLs are considered to be atheroprotective because they are associated in serum with paraoxonases (PONs) which protect HDL from oxidation. Polyphenol consumption reduces the risk of chronic diseases in humans. Polyphenols increase the binding of HDL to PON1, increasing the catalytic activity of PON1. This review summarizes the evidence currently available regarding pharmacological and alternative treatments aimed at improving the functionality of HDL-C. Information on the effectiveness of the treatments has contributed to the understanding of the molecular mechanisms that regulate plasma levels of HDL-C, thereby promoting the development of more effective treatment of cardiovascular diseases. For that purpose, Scopus and Medline databases were searched to identify the publications investigating the impact of current therapies focused on high-density lipoproteins.
Collapse
Affiliation(s)
- Diego Estrada-Luna
- Instituto Nacional de Cardiología "Ignacio Chávez" Juan Badiano No. 1, Belisario Domínguez Sección 16, 14080 Tlalpan, Mexico City, Mexico.
| | - María Araceli Ortiz-Rodriguez
- Facultad de Nutrición, Universidad Autónoma del Estado de Morelos, UAEM, Calle Río Iztaccihuatl S/N, Vista Hermosa, 62350 Cuernavaca, Morelos, Mexico.
| | - Lizett Medina-Briseño
- Universidad de la Sierra Sur, UNSIS, Miahuatlán de Porfirio Díaz, 70800 Oaxaca, Mexico.
| | - Elizabeth Carreón-Torres
- Instituto Nacional de Cardiología "Ignacio Chávez" Juan Badiano No. 1, Belisario Domínguez Sección 16, 14080 Tlalpan, Mexico City, Mexico.
| | - Jeannett Alejandra Izquierdo-Vega
- Área Académica de Medicina, Instituto de Ciencias de la Salud, Universidad Autónoma del Estado de Hidalgo, Carretera Actopan-Tilcuautla, Ex-Hacienda La Concepción S/N, San Agustín Tlaxiaca, 42160 Hidalgo, Mexico.
| | - Ashutosh Sharma
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Queretaro, Epigmenio Gonzalez 500, 76130 Queretaro, Mexico.
| | - Juan Carlos Cancino-Díaz
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas del Instituto Politécnico Nacional, 11340 Ciudad de México, Mexico.
| | - Oscar Pérez-Méndez
- Instituto Nacional de Cardiología "Ignacio Chávez" Juan Badiano No. 1, Belisario Domínguez Sección 16, 14080 Tlalpan, Mexico City, Mexico.
| | | | - Gabriel Betanzos-Cabrera
- Área Académica de Medicina, Instituto de Ciencias de la Salud, Universidad Autónoma del Estado de Hidalgo, Carretera Actopan-Tilcuautla, Ex-Hacienda La Concepción S/N, San Agustín Tlaxiaca, 42160 Hidalgo, Mexico.
| |
Collapse
|
37
|
Stamatikos A, Dronadula N, Ng P, Palmer D, Knight E, Wacker BK, Tang C, Kim F, Dichek DA. ABCA1 Overexpression in Endothelial Cells In Vitro Enhances ApoAI-Mediated Cholesterol Efflux and Decreases Inflammation. Hum Gene Ther 2018; 30:236-248. [PMID: 30079772 DOI: 10.1089/hum.2018.120] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Atherosclerosis, a disease of blood vessels, is driven by cholesterol accumulation and inflammation. Gene therapy that removes cholesterol from blood vessels and decreases inflammation is a promising approach for prevention and treatment of atherosclerosis. In previous work, we reported that helper-dependent adenoviral (HDAd) overexpression of apolipoprotein A-I (apoAI) in endothelial cells (ECs) increases cholesterol efflux in vitro and reduces atherosclerosis in vivo. However, the effect of HDAdApoAI on atherosclerosis is partial. To improve this therapy, we considered concurrent overexpression of ATP-binding cassette subfamily A, member 1 (ABCA1), a protein that is required for apoAI-mediated cholesterol efflux. Before attempting combined apoAI/ABCA1 gene therapy, we tested whether an HDAd that expresses ABCA1 (HDAdABCA1) increases EC cholesterol efflux, whether increased cholesterol efflux alters normal EC physiology, and whether ABCA1 overexpression in ECs has anti-inflammatory effects. HDAdABCA1 increased EC ABCA1 protein (∼3-fold; p < 0.001) and apoAI-mediated cholesterol efflux (2.3-fold; p = 0.007). Under basal culture conditions, ABCA1 overexpression did not alter EC proliferation, metabolism, migration, apoptosis, nitric oxide production, or inflammatory gene expression. However, in serum-starved, apoAI-treated EC, ABCA1 overexpression had anti-inflammatory effects: decreased inflammatory gene expression (∼50%; p ≤ 0.02 for interleukin [IL]-6, tumor necrosis factor [TNF]-α, and vascular cell adhesion protein-1); reduced lipid-raft Toll-like receptor 4 (80%; p = 0.001); and a trend towards increased nitric oxide production (∼55%; p = 0.1). In ECs stimulated with lipopolysaccharide, ABCA1 overexpression markedly decreased inflammatory gene expression (∼90% for IL-6 and TNF-α; p < 0.001). Therefore, EC ABCA1 overexpression has no toxic effects and counteracts the two key drivers of atherosclerosis: cholesterol accumulation and inflammation. In vivo testing of HDAdABCA1 is warranted.
Collapse
Affiliation(s)
- Alexis Stamatikos
- 1 Division of Cardiology, Department of Medicine, University of Washington, Seattle, Washington
| | - Nagadhara Dronadula
- 1 Division of Cardiology, Department of Medicine, University of Washington, Seattle, Washington
| | - Philip Ng
- 2 Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Donna Palmer
- 2 Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Ethan Knight
- 1 Division of Cardiology, Department of Medicine, University of Washington, Seattle, Washington
| | - Bradley K Wacker
- 1 Division of Cardiology, Department of Medicine, University of Washington, Seattle, Washington
| | - Chongren Tang
- 1 Division of Cardiology, Department of Medicine, University of Washington, Seattle, Washington
| | - Francis Kim
- 1 Division of Cardiology, Department of Medicine, University of Washington, Seattle, Washington
| | - David A Dichek
- 1 Division of Cardiology, Department of Medicine, University of Washington, Seattle, Washington
| |
Collapse
|
38
|
Anastasius M, Luquain-Costaz C, Kockx M, Jessup W, Kritharides L. A critical appraisal of the measurement of serum 'cholesterol efflux capacity' and its use as surrogate marker of risk of cardiovascular disease. Biochim Biophys Acta Mol Cell Biol Lipids 2018; 1863:1257-1273. [PMID: 30305243 DOI: 10.1016/j.bbalip.2018.08.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 08/02/2018] [Accepted: 08/03/2018] [Indexed: 12/15/2022]
Abstract
The 'cholesterol efflux capacity (CEC)' assay is a simple in vitro measure of the capacities of individual sera to promote the first step of the reverse cholesterol transport pathway, the delivery of cellular cholesterol to plasma HDL. This review describes the cell biology of this model and critically assesses its application as a marker of cardiovascular risk. We describe the pathways for cell cholesterol export, current cell models used in the CEC assay with their limitations and consider the contribution that measurement of serum CEC provides to our understanding of HDL function in vivo.
Collapse
Affiliation(s)
- Malcolm Anastasius
- ANZAC Research Institute, Concord Repatriation General Hospital, University of Sydney, Sydney, NSW, Australia
| | | | - Maaike Kockx
- ANZAC Research Institute, Concord Repatriation General Hospital, University of Sydney, Sydney, NSW, Australia
| | - Wendy Jessup
- ANZAC Research Institute, Concord Repatriation General Hospital, University of Sydney, Sydney, NSW, Australia
| | - Leonard Kritharides
- ANZAC Research Institute, Concord Repatriation General Hospital, University of Sydney, Sydney, NSW, Australia; Cardiology Department, Concord Repatriation General Hospital, University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
39
|
Mahmoodi K, Kamali K, Ghaznavi H, Soltanpour MS. The C-565T Polymorphism (rs2422493) of the ATP-binding Cassette Transporter A1 Gene Contributes to the Development and Severity of Coronary Artery Disease in an Iranian Population. Oman Med J 2018; 33:309-315. [PMID: 30038730 DOI: 10.5001/omj.2018.57] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Objectives ATP-binding cassette transporter A1 (ABCA1) plays a pivotal role in reverse cholesterol transport from peripheral tissues back to the liver. Abnormalities in ABCA1 function may lead to dyslipidemia and coronary artery disease (CAD). We investigated the role of C-565T (rs2422493) promoter polymorphism of ABCA1 gene in the development and severity of CAD in an Iranian subpopulation. Methods Our study population consisted of 110 angiographically-confirmed CAD patients and 110 matched controls. The severity of CAD was expressed based on the number of stenotic vessels. Genotyping of C-565T promoter polymorphism was performed using the polymerase chain reaction followed by restriction fragments length polymorphism analysis methods. Lipid profile was determined by routine colorimetric methods. Results The distribution of ABCA1 C-565T genotypes (p = 0.035) and alleles (p = 0.017) was significantly different between the CAD and control groups. In univariate analysis (with genotype CC as reference), the TT genotype was significantly associated with an increased risk of CAD (odds ratio = 3.83; 95% confidence interval: 1.29-11.30, p = 0.014), but the CT genotype was not (p = 0.321). A multiple binary logistic regression analysis revealed that smoking, hypertension, triglyceride, cholesterol, high-density lipoprotein cholesterol, low-density lipoprotein cholesterol, and ABCA1 C-565T dominant genotype were significant and independent risk factors for CAD development (p < 0.050). The ABCA1 C-565T polymorphism affected the severity of CAD in TT homozygote state (p = 0.028). However, no significant correlation was seen between this common polymorphism and lipid profile in the study population (p > 0.050). Conclusions: Our study indicated that ABCA1 C-565T polymorphism is a significant risk factor for development and severity of CAD in our population.
Collapse
Affiliation(s)
- Khalil Mahmoodi
- Department of Cardiology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Koorosh Kamali
- Department of Public Health, School of Public Health, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Habib Ghaznavi
- Health Promotion Research Centre, Department of Pharmacology, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Mohammad Soleiman Soltanpour
- Department of Medical Laboratory Sciences, School of Paramedical Sciences, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
40
|
Akinyemiju T, Do AN, Patki A, Aslibekyan S, Zhi D, Hidalgo B, Tiwari HK, Absher D, Geng X, Arnett DK, Irvin MR. Epigenome-wide association study of metabolic syndrome in African-American adults. Clin Epigenetics 2018; 10:49. [PMID: 29643945 PMCID: PMC5891946 DOI: 10.1186/s13148-018-0483-2] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 03/27/2018] [Indexed: 01/10/2023] Open
Abstract
Background The high prevalence of obesity among US adults has resulted in significant increases in associated metabolic disorders such as diabetes, dyslipidemia, and high blood pressure. Together, these disorders constitute metabolic syndrome, a clinically defined condition highly prevalent among African-Americans. Identifying epigenetic alterations associated with metabolic syndrome may provide additional information regarding etiology beyond current evidence from genome-wide association studies. Methods Data on metabolic syndrome and DNA methylation was assessed on 614 African-Americans from the Hypertension Genetic Epidemiology Network (HyperGEN) study. Metabolic syndrome was defined using the joint harmonized criteria, and DNA methylation was assessed using the Illumina HumanMethylation450K Bead Chip assay on DNA extracted from buffy coat. Linear mixed effects regression models were used to examine the association between CpG methylation at > 450,000 CpG sites and metabolic syndrome adjusted for study covariates. Replication using DNA from a separate sample of 69 African-Americans, as well as meta-analysis combining both cohorts, was conducted. Results Two differentially methylated CpG sites in the IGF2BP1 gene on chromosome 17 (cg06638433; p value = 3.10 × 10− 7) and the ABCG1 gene on chromosome 21 (cg06500161; p value = 2.60 × 10− 8) were identified. Results for the ABCG1 gene remained statistically significant in the replication dataset and meta-analysis. Conclusion Metabolic syndrome was consistently associated with increased methylation in the ABCG1 gene in the discovery and replication datasets, a gene that encodes a protein in the ATP-binding cassette transporter family and is involved in intra- and extra-cellular signaling and lipid transport. Electronic supplementary material The online version of this article (10.1186/s13148-018-0483-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tomi Akinyemiju
- 1Department of Epidemiology, University of Kentucky, Lexington, KY USA
| | - Anh N Do
- 2Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL USA
| | - Amit Patki
- 3Department of Biostatistics, University of Alabama at Birmingham, Birmingham, AL USA
| | - Stella Aslibekyan
- 2Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL USA
| | - Degui Zhi
- 4School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX USA.,5School of Public Health, University of Texas Health Science Center at Houston, Houston, TX USA
| | - Bertha Hidalgo
- 2Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL USA
| | - Hemant K Tiwari
- 3Department of Biostatistics, University of Alabama at Birmingham, Birmingham, AL USA
| | - Devin Absher
- 6HudsonAlpha Institute for Biotechnology, Huntsville, AL USA
| | - Xin Geng
- 4School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX USA
| | - Donna K Arnett
- 7College of Public Health, University of Kentucky, Lexington, KY USA
| | - Marguerite R Irvin
- 2Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL USA
| |
Collapse
|
41
|
Browning KL, Lind TK, Maric S, Malekkhaiat-Häffner S, Fredrikson GN, Bengtsson E, Malmsten M, Cárdenas M. Human Lipoproteins at Model Cell Membranes: Effect of Lipoprotein Class on Lipid Exchange. Sci Rep 2017; 7:7478. [PMID: 28785025 PMCID: PMC5547137 DOI: 10.1038/s41598-017-07505-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 06/29/2017] [Indexed: 01/19/2023] Open
Abstract
High and low density lipoproteins (HDL and LDL) are thought to play vital roles in the onset and development of atherosclerosis; the biggest killer in the western world. Key issues of initial lipoprotein (LP) interactions at cellular membranes need to be addressed including LP deposition and lipid exchange. Here we present a protocol for monitoring the in situ kinetics of lipoprotein deposition and lipid exchange/removal at model cellular membranes using the non-invasive, surface sensitive methods of neutron reflection and quartz crystal microbalance with dissipation. For neutron reflection, lipid exchange and lipid removal can be distinguished thanks to the combined use of hydrogenated and tail-deuterated lipids. Both HDL and LDL remove lipids from the bilayer and deposit hydrogenated material into the lipid bilayer, however, the extent of removal and exchange depends on LP type. These results support the notion of HDL acting as the ‘good’ cholesterol, removing lipid material from lipid-loaded cells, whereas LDL acts as the ‘bad’ cholesterol, depositing lipid material into the vascular wall.
Collapse
Affiliation(s)
- K L Browning
- Department of Pharmacy, Uppsala University, Uppsala, Sweden
| | - T K Lind
- Department of Biomedical Sciences and Biofilms, Malmö University, Malmö, Sweden
| | - S Maric
- Department of Biomedical Sciences and Biofilms, Malmö University, Malmö, Sweden
| | | | - G N Fredrikson
- Department of Clinical Sciences, Malmö, Lund University, Malmö, Sweden
| | - E Bengtsson
- Department of Clinical Sciences, Malmö, Lund University, Malmö, Sweden
| | - M Malmsten
- Department of Pharmacy, Uppsala University, Uppsala, Sweden. .,Department of Pharmacy, University of Copenhagen, Copenhagen, Denmark.
| | - M Cárdenas
- Department of Biomedical Sciences and Biofilms, Malmö University, Malmö, Sweden.
| |
Collapse
|
42
|
Pamir N, Hutchins PM, Ronsein GE, Wei H, Tang C, Das R, Vaisar T, Plow E, Schuster V, Koschinsky ML, Reardon CA, Weinberg R, Dichek DA, Marcovina S, Getz GS, Heinecke JW. Plasminogen promotes cholesterol efflux by the ABCA1 pathway. JCI Insight 2017; 2:92176. [PMID: 28768900 DOI: 10.1172/jci.insight.92176] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 06/20/2017] [Indexed: 12/20/2022] Open
Abstract
Using genetic and biochemical approaches, we investigated proteins that regulate macrophage cholesterol efflux capacity (CEC) and ABCA1-specific CEC (ABCA1 CEC), 2 functional assays that predict cardiovascular disease (CVD). Macrophage CEC and the concentration of HDL particles were markedly reduced in mice deficient in apolipoprotein A-I (APOA1) or apolipoprotein E (APOE) but not apolipoprotein A-IV (APOA4). ABCA1 CEC was markedly reduced in APOA1-deficient mice but was barely affected in mice deficient in APOE or APOA4. High-resolution size-exclusion chromatography of plasma produced 2 major peaks of ABCA1 CEC activity. The early-eluting peak, which coeluted with HDL, was markedly reduced in APOA1- or APOE-deficient mice. The late-eluting peak was modestly reduced in APOA1-deficient mice but little affected in APOE- or APOA4-deficient mice. Ion-exchange chromatography and shotgun proteomics suggested that plasminogen (PLG) accounted for a substantial fraction of the ABCA1 CEC activity in the peak not associated with HDL. Human PLG promoted cholesterol efflux by the ABCA1 pathway, and PLG-dependent efflux was inhibited by lipoprotein(a) [Lp(a)]. Our observations identify APOA1, APOE, and PLG as key determinants of CEC. Because PLG and Lp(a) associate with human CVD risk, interplay among the proteins might affect atherosclerosis by regulating cholesterol efflux from macrophages.
Collapse
Affiliation(s)
- Nathalie Pamir
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Patrick M Hutchins
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | | | - Hao Wei
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Chongren Tang
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Riku Das
- Department of Molecular Cardiology, Cleveland Clinic, Cleveland, Ohio, USA
| | - Tomas Vaisar
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Edward Plow
- Department of Molecular Cardiology, Cleveland Clinic, Cleveland, Ohio, USA
| | - Volker Schuster
- Hospital for Children and Adolescents, Medical Faculty of Leipzig University, Leipzig, Germany
| | - Marlys L Koschinsky
- Robarts Research Institute, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| | | | - Richard Weinberg
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - David A Dichek
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Santica Marcovina
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Godfrey S Getz
- Department of Pathology, University of Chicago, Chicago, Illinois, USA
| | - Jay W Heinecke
- Department of Medicine, University of Washington, Seattle, Washington, USA
| |
Collapse
|
43
|
Jeong SJ, Lee MN, Oh GT. The Role of Macrophage Lipophagy in Reverse Cholesterol Transport. Endocrinol Metab (Seoul) 2017; 32:41-46. [PMID: 28345315 PMCID: PMC5368120 DOI: 10.3803/enm.2017.32.1.41] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 02/20/2017] [Accepted: 02/27/2017] [Indexed: 12/23/2022] Open
Abstract
Macrophage cholesterol efflux is a central step in reverse cholesterol transport, which helps to maintain cholesterol homeostasis and to reduce atherosclerosis. Lipophagy has recently been identified as a new step in cholesterol ester hydrolysis that regulates cholesterol efflux, since it mobilizes cholesterol from lipid droplets of macrophages via autophagy and lysosomes. In this review, we briefly discuss recent advances regarding the mechanisms of the cholesterol efflux pathway in macrophage foam cells, and present lipophagy as a therapeutic target in the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Se Jin Jeong
- Immune and Vascular Cell Network Research Center, National Creative Initiatives, Department of Life Sciences, Ewha Womans University, Seoul, Korea
| | - Mi Ni Lee
- Immune and Vascular Cell Network Research Center, National Creative Initiatives, Department of Life Sciences, Ewha Womans University, Seoul, Korea
| | - Goo Taeg Oh
- Immune and Vascular Cell Network Research Center, National Creative Initiatives, Department of Life Sciences, Ewha Womans University, Seoul, Korea.
| |
Collapse
|
44
|
Gabunia K, Herman AB, Ray M, Kelemen SE, England RN, DeLa Cadena R, Foster WJ, Elliott KJ, Eguchi S, Autieri MV. Induction of MiR133a expression by IL-19 targets LDLRAP1 and reduces oxLDL uptake in VSMC. J Mol Cell Cardiol 2017; 105:38-48. [PMID: 28257760 DOI: 10.1016/j.yjmcc.2017.02.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 02/21/2017] [Accepted: 02/27/2017] [Indexed: 10/20/2022]
Abstract
The transformation of vascular smooth muscle cells [VSMC] into foam cells leading to increased plaque size and decreased stability is a key, yet understudied step in atherogenesis. We reported that Interleukin-19 (IL-19), a novel, anti-inflammatory cytokine, attenuates atherosclerosis by anti-inflammatory effects on VSMC. In this work we report that IL-19 induces expression of miR133a, a muscle-specific miRNA, in VSMC. Although previously unreported, we report that miR133a can target and reduce mRNA abundance, mRNA stability, and protein expression of Low Density Lipoprotein Receptor Adaptor Protein 1, (LDLRAP1), an adaptor protein which functions to internalize the LDL receptor. Mutations in this gene lead to LDL receptor malfunction and cause the Autosomal Recessive Hypercholesterolemia (ARH) disorder in humans. Herein we show that IL-19 reduces lipid accumulation in VSMC, and LDLRAP1 expression and oxLDL uptake in a miR133a-dependent mechanism. We show that LDLRAP1 is expressed in plaque and neointimal VSMC of mouse and human injured arteries. Transfection of miR133a and LDLRAP1 siRNA into VSMC reduces their proliferation and uptake of oxLDL. miR133a is significantly increased in plasma from hyperlipidemic compared with normolipidemic patients. Expression of miR133a in IL-19 stimulated VSMC represents a previously unrecognized link between vascular lipid metabolism and inflammation, and may represent a therapeutic opportunity to combat vascular inflammatory diseases.
Collapse
Affiliation(s)
- Khatuna Gabunia
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States
| | - Allison B Herman
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States
| | - Mitali Ray
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States
| | - Sheri E Kelemen
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States
| | - Ross N England
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States
| | - Raul DeLa Cadena
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States
| | - William J Foster
- Departments of Ophthalmology & Bioengineering, Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States
| | - Katherine J Elliott
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States
| | - Satoru Eguchi
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States
| | - Michael V Autieri
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States.
| |
Collapse
|
45
|
Pulakazhi Venu VK, Adijiang A, Seibert T, Chen YX, Shi C, Batulan Z, O'Brien ER. Heat shock protein 27-derived atheroprotection involves reverse cholesterol transport that is dependent on GM-CSF to maintain ABCA1 and ABCG1 expression in ApoE -/- mice. FASEB J 2017; 31:2364-2379. [PMID: 28232480 DOI: 10.1096/fj.201601188r] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 01/30/2017] [Indexed: 11/11/2022]
Abstract
Recently, we demonstrated that heat shock protein (HSP)-27 is protective against the development of experimental atherosclerosis, reducing plaque cholesterol content by more than 30%. Moreover, elevated HSP-27 levels are predictive of relative freedom from clinical cardiovascular events. HSP-27 signaling occurs via the activation of NF-κB, which induces a marked up-regulation in expression of granulocyte-monocyte colony-stimulating factor (GM-CSF), a cytokine that is known to alter ABC transporters involved in reverse cholesterol transport (RCT). Therefore, we hypothesized that HSP-27-derived GM-CSF has a potent role in impeding plaque formation by promoting macrophage RCT and sought to better characterize this pathway. Treatment of THP-1 cells, RAW-Blue cells, and primary macrophages with recombinant HSP-27 resulted in NF-κB activation via TLR-4 and was inhibited by various pharmacologic blockers of this pathway. Moreover, HSP-27-induced upregulation of GM-CSF expression was dependent on TLR-4 signaling. Recombinant (r)HSP-27 treatment of ApoE-/- female (but not male) mice for 4 wk yielded reductions in plaque area and cholesterol clefts of 33 and 47%, respectively, with no effect on GM-CSF-/-ApoE-/- mice. With 12 wk of rHSP-27 treatment, both female and male mice showed reductions in plaque burden (55 and 42%, respectively) and a 60% reduction in necrotic core area but no treatment effect in GM-CSF-/-ApoE-/- mice. In vitro functional studies revealed that HSP-27 enhanced the expression of ABCA1 and ABCG1, as well as facilitated cholesterol efflux in vitro by ∼10%. These novel findings establish a paradigm for HSP-27-mediated RCT and set the stage for the development of HSP-27 atheroprotective therapeutics.-Pulakazhi Venu, V. K., Adijiang, A., Seibert, T., Chen, Y.-X., Shi, C., Batulan, Z., O'Brien, E. R. Heat shock protein 27-derived atheroprotection involves reverse cholesterol transport that is dependent on GM-CSF to maintain ABCA1 and ABCG1 expression in ApoE-/- mice.
Collapse
Affiliation(s)
- Vivek Krishna Pulakazhi Venu
- Department of Cardiac Sciences, Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Ayinuer Adijiang
- Department of Cardiac Sciences, Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Tara Seibert
- Department of Cardiac Sciences, Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Yong-Xiang Chen
- Department of Cardiac Sciences, Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Chunhua Shi
- Department of Cardiac Sciences, Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Zarah Batulan
- Department of Cardiac Sciences, Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Edward R O'Brien
- Department of Cardiac Sciences, Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
46
|
Hoekstra M, Van Berkel TJ. Functionality of High-Density Lipoprotein as Antiatherosclerotic Therapeutic Target. Arterioscler Thromb Vasc Biol 2016; 36:e87-e94. [DOI: 10.1161/atvbaha.116.308262] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- Menno Hoekstra
- From the Division of Biopharmaceutics, Cluster BioTherapeutics, Leiden Academic Centre for Drug Research, Gorlaeus Laboratories, The Netherlands
| | - Theo J.C. Van Berkel
- From the Division of Biopharmaceutics, Cluster BioTherapeutics, Leiden Academic Centre for Drug Research, Gorlaeus Laboratories, The Netherlands
| |
Collapse
|
47
|
Gui Y, Yao S, Yan H, Hu L, Yu C, Gao F, Xi C, Li H, Ye Y, Wang Y. A novel small molecule liver X receptor transcriptional regulator, nagilactone B, suppresses atherosclerosis in apoE-deficient mice. Cardiovasc Res 2016; 112:502-14. [DOI: 10.1093/cvr/cvw183] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 06/23/2016] [Indexed: 11/14/2022] Open
|
48
|
Acyl-CoA:Diacylglycerol Acyltransferase 1 Expression Level in the Hematopoietic Compartment Impacts Inflammation in the Vascular Plaques of Atherosclerotic Mice. PLoS One 2016; 11:e0156364. [PMID: 27223895 PMCID: PMC4880185 DOI: 10.1371/journal.pone.0156364] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 05/12/2016] [Indexed: 01/09/2023] Open
Abstract
The final step of triacylglycerol synthesis is catalyzed by acyl-CoA:diacylglycerol acyltransferases (DGATs). We have previously shown that ApoE-/-Dgat1-/- mice are protected from developing atherosclerosis in association with reduced foam cell formation. However, the role of DGAT1, specifically in myeloid and other hematopoietic cell types, in determining this protective phenotype is unknown. To address this question, we reconstituted the bone marrow of irradiated Ldlr–/–mice with that from wild-type (WT→ Ldlr–/–) and Dgat1–/–(Dgat1–/–→ Ldlr–/–) donor mice. We noted that DGAT1 in the hematopoietic compartment exerts a sex-specific effect on systemic cholesterol homeostasis. However, both male and female Dgat1–/–→ Ldlr–/–mice had higher circulating neutrophil and lower lymphocyte counts than control mice, suggestive of a classical inflammatory phenotype. Moreover, specifically examining the aortae of these mice revealed that Dgat1–/–→ Ldlr–/–mice have atherosclerotic plaques with increased macrophage content. This increase was coupled to a reduced plaque collagen content, leading to a reduced collagen-to-macrophage ratio. Together, these findings point to a difference in the inflammatory contribution to plaque composition between Dgat1–/–→ Ldlr–/–and control mice. By contrast, DGAT1 deficiency did not affect the transcriptional responses of cultured macrophages to lipoprotein treatment in vitro, suggesting that the alterations seen in the plaques of Dgat1–/–→ Ldlr–/–mice in vivo do not reflect a cell intrinsic effect of DGAT1 in macrophages. We conclude that although DGAT1 in the hematopoietic compartment does not impact the overall lipid content of atherosclerotic plaques, it exerts reciprocal effects on inflammation and fibrosis, two processes that control plaque vulnerability.
Collapse
|
49
|
Demina EP, Miroshnikova VV, Schwarzman AL. Role of the ABC transporters A1 and G1, key reverse cholesterol transport proteins, in atherosclerosis. Mol Biol 2016. [DOI: 10.1134/s0026893316020047] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
50
|
Affiliation(s)
- Hong Lu
- From the Saha Cardiovascular Research Center, University of Kentucky, Lexington.
| | - Alan Daugherty
- From the Saha Cardiovascular Research Center, University of Kentucky, Lexington
| |
Collapse
|