1
|
Chen X, Kang Y, Tang C, Zhang L, Guo L. TLR4 promotes smooth muscle cell-derived foam cells formation by inducing receptor-independent macropinocytosis. Biosci Biotechnol Biochem 2024; 89:22-32. [PMID: 39455413 DOI: 10.1093/bbb/zbae153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 10/22/2024] [Indexed: 10/28/2024]
Abstract
Foam cells are primarily formed through scavenger receptors that mediate the uptake of various modified low-density lipoproteins (LDL) into cells. In addition to the receptor-dependent pathway, macropinocytosis is an essential nonreceptor endocytic pathway for vascular smooth muscle cells (VSMCs) to take up lipids. However, the molecular mechanisms underlying this process remain unclear. Primary cultured VSMCs were stimulated with 200 ng/mL lipopolysaccharide (LPS) and 200 µg/mL native LDL (nLDL). We observed a significant increase in Toll-like receptor 4 (TLR4) protein expression and a significant activation of macropinocytosis, which correlated with the highest uptake of nLDL and intracellular lipid deposition in WT VSMCs. However, macropinocytosis was inhibited and lipid accumulation decreased after treatment with macropinocytosis inhibitors and Syk inhibitors in WT VSMCs. Consistently, TLR4 knockout significantly suppressed macropinocytosis and lipid droplets accumulation in VSMCs. Taken together, our findings suggest a critical role of TLR4/Syk signaling in promoting receptor-independent macropinocytosis leading to VSMC-derived foam cells formation.
Collapse
MESH Headings
- Toll-Like Receptor 4/metabolism
- Pinocytosis/drug effects
- Animals
- Foam Cells/metabolism
- Foam Cells/cytology
- Foam Cells/drug effects
- Syk Kinase/metabolism
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/metabolism
- Signal Transduction
- Lipopolysaccharides/pharmacology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/cytology
- Mice
- Lipoproteins, LDL/metabolism
- Lipoproteins, LDL/pharmacology
- Cells, Cultured
- Mice, Knockout
- Mice, Inbred C57BL
Collapse
Affiliation(s)
- Xue Chen
- Department of Rheumatology and Clinical Immunology, Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Yulai Kang
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Chunhua Tang
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Lili Zhang
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Lu Guo
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| |
Collapse
|
2
|
Saki N, Haybar H, Maniati M, Davari N, Javan M, Moghimian-Boroujeni B. Modification macrophage to foam cells in atherosclerosis disease: some factors stimulate or inhibit this process. J Diabetes Metab Disord 2024; 23:1687-1697. [PMID: 39610485 PMCID: PMC11599683 DOI: 10.1007/s40200-024-01482-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 07/16/2024] [Indexed: 11/30/2024]
Abstract
Background Atherosclerosis is an arterial blood vessel disease that begins and progresses by turning macrophages into foam cells. Uptake of oxidized low-density lipoprotein (ox-LDL), cholesterol esterification and cholesterol efflux are the most important factors in the formation of foam cells and play an important role in atherosclerosis. Methods The present study is based on the data obtained from the PubMed database (1961-2024) using the MeSH search terms "Atherosclerosis", "Macrophages" and "Foam cells". Reviews for writing the main text and non-English-language articles were excluded. Result The interaction between ox-LDL and macrophages plays an important role in plaque initiation and promotion processes. Macrophages abnormally digest ox-LDL, resulting in the accumulation of lipids and formation of foam cells. This is an important step in the development of atherosclerosis. Also, several other factors such as inflammatory factors, growth factors, hormones, etc. can play an important role in the development of atherosclerotic lesions or counteract it by affecting the formation of foam cells. Conclusion Several factors can affect the progression of atherosclerosis by affecting macrophage activity or its conversion to foam cells. Also, some of these factors play a protective role against the development and atherosclerosis progression. In this paper, we reviewed some of these factors and their effect on atherosclerosis.
Collapse
Affiliation(s)
- Najmaldin Saki
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Habib Haybar
- Cardiology Department, Medical College, Golestan Hospital, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mahmood Maniati
- School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Nader Davari
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammadreza Javan
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Iranian Blood Transfusion Organization (IBTO), Tehran, Iran
| | - Bahareh Moghimian-Boroujeni
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
3
|
Pourteymour S, Fan J, Majhi RK, Guo S, Sun X, Huang Z, Liu Y, Winter H, Bäcklund A, Skenteris NT, Chernogubova E, Werngren O, Li Z, Skogsberg J, Li Y, Matic L, Hedin U, Maegdefessel L, Ehrenborg E, Tian Y, Jin H. PIEZO1 targeting in macrophages boosts phagocytic activity and foam cell apoptosis in atherosclerosis. Cell Mol Life Sci 2024; 81:331. [PMID: 39107572 PMCID: PMC11335255 DOI: 10.1007/s00018-024-05372-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 06/20/2024] [Accepted: 07/17/2024] [Indexed: 08/22/2024]
Abstract
The rising incidences of atherosclerosis have necessitated efforts to identify novel targets for therapeutic interventions. In the present study, we observed increased expression of the mechanosensitive calcium channel Piezo1 transcript in mouse and human atherosclerotic plaques, correlating with infiltration of PIEZO1-expressing macrophages. In vitro administration of Yoda1, a specific agonist for PIEZO1, led to increased foam cell apoptosis and enhanced phagocytosis by macrophages. Mechanistically, PIEZO1 activation resulted in intracellular F-actin rearrangement, elevated mitochondrial ROS levels and induction of mitochondrial fragmentation upon PIEZO1 activation, as well as increased expression of anti-inflammatory genes. In vivo, ApoE-/- mice treated with Yoda1 exhibited regression of atherosclerosis, enhanced stability of advanced lesions, reduced plaque size and necrotic core, increased collagen content, and reduced expression levels of inflammatory markers. Our findings propose PIEZO1 as a novel and potential therapeutic target in atherosclerosis.
Collapse
Affiliation(s)
- Shirin Pourteymour
- Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden.
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Blindern, PO Box 1046, 0317, Oslo, Norway.
| | - Jingxue Fan
- Department of Cardiology, The First Affiliated Hospital, Cardiovascular Institute, Harbin Medical University, Harbin, People's Republic of China
| | - Rakesh Kumar Majhi
- Department of Microbiology, Tumor and Cell Biology, Division of Clinical Microbiology, Karolinska Institutet, Stockholm, Sweden
| | - Shuyuan Guo
- Department of Cardiology, The First Affiliated Hospital, Cardiovascular Institute, Harbin Medical University, Harbin, People's Republic of China
| | - Xin Sun
- Department of Cardiology, Shenzhen Cardiovascular Minimally Invasive Medical Engineering Technology Research and Development Center, Shenzhen People's Hospital, Shenzhen, People's Republic of China
| | - Zhen Huang
- Department of Cardiology, The First Affiliated Hospital, Cardiovascular Institute, Harbin Medical University, Harbin, People's Republic of China
| | - Ying Liu
- Department of Cardiology, The First Affiliated Hospital, Cardiovascular Institute, Harbin Medical University, Harbin, People's Republic of China
| | - Hanna Winter
- Department of Vascular and Endovascular Surgery, Technical University Munich, Munich, Germany
| | - Alexandra Bäcklund
- Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
| | - Nikolaos-Taxiarchis Skenteris
- Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | | | - Olivera Werngren
- Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
| | - Zhaolong Li
- Department of Vascular and Endovascular Surgery, Technical University Munich, Munich, Germany
| | - Josefin Skogsberg
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Yuhuang Li
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Ljubica Matic
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Ulf Hedin
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Lars Maegdefessel
- Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
- Department of Vascular and Endovascular Surgery, Technical University Munich, Munich, Germany
| | - Ewa Ehrenborg
- Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
| | - Ye Tian
- Department of Cardiology, The First Affiliated Hospital, Cardiovascular Institute, Harbin Medical University, Harbin, People's Republic of China.
| | - Hong Jin
- Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden.
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
4
|
Tobeh NS, Bruce KD. Emerging Alzheimer's disease therapeutics: promising insights from lipid metabolism and microglia-focused interventions. Front Aging Neurosci 2023; 15:1259012. [PMID: 38020773 PMCID: PMC10630922 DOI: 10.3389/fnagi.2023.1259012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 10/03/2023] [Indexed: 12/01/2023] Open
Abstract
More than 55 million people suffer from dementia, with this number projected to double every 20 years. In the United States, 1 in 3 aged individuals dies from Alzheimer's disease (AD) or another type of dementia and AD kills more individuals than breast cancer and prostate cancer combined. AD is a complex and multifactorial disease involving amyloid plaque and neurofibrillary tangle formation, glial cell dysfunction, and lipid droplet accumulation (among other pathologies), ultimately leading to neurodegeneration and neuronal death. Unfortunately, the current FDA-approved therapeutics do not reverse nor halt AD. While recently approved amyloid-targeting antibodies can slow AD progression to improve outcomes for some patients, they are associated with adverse side effects, may have a narrow therapeutic window, and are expensive. In this review, we evaluate current and emerging AD therapeutics in preclinical and clinical development and provide insight into emerging strategies that target brain lipid metabolism and microglial function - an approach that may synergistically target multiple mechanisms that drive AD neuropathogenesis. Overall, we evaluate whether these disease-modifying emerging therapeutics hold promise as interventions that may be able to reverse or halt AD progression.
Collapse
Affiliation(s)
- Nour S Tobeh
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Kimberley D Bruce
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
5
|
Lin HP, Singla B, Ahn W, Ghoshal P, Blahove M, Cherian-Shaw M, Chen A, Haller A, Hui DY, Dong K, Zhou J, White J, Stranahan AM, Jasztal A, Lucas R, Stansfield BK, Fulton D, Chlopicki S, Csányi G. Receptor-independent fluid-phase macropinocytosis promotes arterial foam cell formation and atherosclerosis. Sci Transl Med 2022; 14:eadd2376. [PMID: 36130017 PMCID: PMC9645012 DOI: 10.1126/scitranslmed.add2376] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Accumulation of lipid-laden foam cells in the arterial wall plays a central role in atherosclerotic lesion development, plaque progression, and late-stage complications of atherosclerosis. However, there are still fundamental gaps in our knowledge of the underlying mechanisms leading to foam cell formation in atherosclerotic arteries. Here, we investigated the role of receptor-independent macropinocytosis in arterial lipid accumulation and pathogenesis of atherosclerosis. Genetic inhibition of fluid-phase macropinocytosis in myeloid cells (LysMCre+ Nhe1fl/fl) and repurposing of a Food and Drug Administration (FDA)-approved drug that inhibits macrophage macropinocytosis substantially decreased atherosclerotic lesion development in low-density lipoprotein (LDL) receptor-deficient and Apoe-/- mice. Stimulation of macropinocytosis using genetic (H-RASG12V) and physiologically relevant approaches promoted internalization of unmodified native (nLDL) and modified [e.g., acetylated (ac) and oxidized (ox) LDL] lipoproteins in both wild-type and scavenger receptor (SR) knockout (Cd36-/-/Sra-/-) macrophages. Pharmacological inhibition of macropinocytosis in hypercholesterolemic wild-type and Cd36-/-/Sra-/- mice identified an important role of macropinocytosis in LDL uptake by lesional macrophages and development of atherosclerosis. Furthermore, serial section high-resolution imaging, LDL immunolabeling, and three-dimensional (3D) reconstruction of subendothelial foam cells provide visual evidence of lipid macropinocytosis in both human and murine atherosclerotic arteries. Our findings complement the SR paradigm of atherosclerosis and identify a therapeutic strategy to counter the development of atherosclerosis and cardiovascular disease.
Collapse
Affiliation(s)
- Hui-Ping Lin
- Vascular Biology Center, Medical College of Georgia, Augusta University, USA
| | - Bhupesh Singla
- Vascular Biology Center, Medical College of Georgia, Augusta University, USA
| | - WonMo Ahn
- Vascular Biology Center, Medical College of Georgia, Augusta University, USA
| | - Pushpankur Ghoshal
- Vascular Biology Center, Medical College of Georgia, Augusta University, USA
| | - Maria Blahove
- Vascular Biology Center, Medical College of Georgia, Augusta University, USA
| | - Mary Cherian-Shaw
- Vascular Biology Center, Medical College of Georgia, Augusta University, USA
| | - Alex Chen
- Vascular Biology Center, Medical College of Georgia, Augusta University, USA
| | - April Haller
- Department of Pathology, University of Cincinnati College of Medicine, USA
| | - David Y. Hui
- Department of Pathology, University of Cincinnati College of Medicine, USA
| | - Kunzhe Dong
- Department of Pharmacology & Toxicology, Medical College of Georgia, Augusta University, USA
| | - Jiliang Zhou
- Department of Pharmacology & Toxicology, Medical College of Georgia, Augusta University, USA
| | - Joseph White
- Department of Pathology, Medical College of Georgia, Augusta University, USA
| | - Alexis M. Stranahan
- Department of Neuroscience & Regenerative Medicine, Medical College of Georgia, Augusta University, USA
| | - Agnieszka Jasztal
- Jagiellonian Centre for Experimental Therapeutics, Jagiellonian University, Krakow, Poland
| | - Rudolf Lucas
- Vascular Biology Center, Medical College of Georgia, Augusta University, USA
- Department of Pharmacology & Toxicology, Medical College of Georgia, Augusta University, USA
| | - Brian K. Stansfield
- Vascular Biology Center, Medical College of Georgia, Augusta University, USA
- Department of Pediatrics, Medical College of Georgia, Augusta University, USA
| | - David Fulton
- Vascular Biology Center, Medical College of Georgia, Augusta University, USA
- Department of Pharmacology & Toxicology, Medical College of Georgia, Augusta University, USA
| | - Stefan Chlopicki
- Jagiellonian Centre for Experimental Therapeutics, Jagiellonian University, Krakow, Poland
| | - Gábor Csányi
- Vascular Biology Center, Medical College of Georgia, Augusta University, USA
- Department of Pharmacology & Toxicology, Medical College of Georgia, Augusta University, USA
| |
Collapse
|
6
|
Miyazaki T. Pinocytotic engulfment of lipoproteins by macrophages. Front Cardiovasc Med 2022; 9:957897. [PMID: 36105534 PMCID: PMC9464914 DOI: 10.3389/fcvm.2022.957897] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 08/11/2022] [Indexed: 11/22/2022] Open
Abstract
Atherosclerosis is a major cause of acute coronary syndrome and stroke. Foam cell formation in macrophages is involved in controlling plaque stability and the pathogenesis of atherosclerosis. Accordingly, many studies have examined the processes of lipid incorporation, such as scavenger receptor-mediated uptake of oxidized low-density lipoprotein, in cells. In addition to receptor-mediated machinery, growing evidence has suggested that pinocytosis, which is a receptor-independent endocytic pathway, is associated with foam cell formation when a sufficient number of lipoproteins is accumulated around cells. Pinocytotic engulfment of nanoparticles is initiated by plasma membrane ruffling in a phosphatidylinositol-3 kinase-dependent manner. Subsequent to pinosome closure, the majority of pinosomes are internalized through endocytic processes, and they can be recycled into the plasma membrane. These pinocytotic processes are modulated by small GTPases and their cytoskeletal rearrangement. Moreover, pinocytotic abilities may vary between immunological subsets in cells. Accordingly, macrophages may show diverse pinocytotic abilities depending on the surrounding microenvironment. This review summarizes the current understanding of pinocytotic engulfment of lipoprotein in macrophages, and discusses how this endocytic process is governed under hypercholesterolemic conditions.
Collapse
|
7
|
Mamillapalli R, Toffoloni N, Habata S, Qunhua H, Atwani R, Stachenfeld N, Taylor HS. Endometriosis promotes atherosclerosis in a murine model. Am J Obstet Gynecol 2022; 227:248.e1-248.e8. [PMID: 35351413 PMCID: PMC9308711 DOI: 10.1016/j.ajog.2022.03.040] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 03/14/2022] [Accepted: 03/23/2022] [Indexed: 11/24/2022]
Abstract
BACKGROUND Epidemiologic studies have demonstrated an association between endometriosis and the subsequent development of cardiovascular disease. The direct effect of endometriosis on the progression of atherosclerotic, if any, has not been previously characterized. Endometriosis leads to systemic inflammation that could have consequences for cardiovascular health. Here, we reported the effects of endometriosis on the development of atherosclerosis in a murine model. OBJECTIVE This study aimed to determine the contribution of endometriosis in promoting cardiovascular disease in a murine model of endometriosis. STUDY DESIGN Endometriosis was induced in 18 apolipoprotein E-null mice, the standard murine model used to study atherosclerosis. Mice of the same strain were used as controls (n=18) and underwent sham surgery without inducing endometriosis. The formation of endometriotic lesions was confirmed after 25 weeks of induction. Atherosclerotic lesions were subjected to hematoxylin and eosin staining followed by measurement of the aortic root luminal area and wall thickness. The whole aorta was isolated, and Oil Red O staining was performed to quantify the lipid deposits or plaque formation; moreover, biochemical assays were carried out in serum to determine the levels of lipids and inflammatory-related cytokines. RESULTS Apolipoprotein E mice with endometriosis exhibited increased aortic atherosclerosis compared with controls as measured using Oil Red O staining (7.9% vs 3.1%, respectively; P=.0004). Mice with endometriosis showed a significant 50% decrease in the aortic luminal area compared with sham mice (0.85 mm2 vs 1.46 mm2; P=.03) and a significant increase in aortic root wall thickness (0.22 mm vs 0.15 mm; P=.04). There was no difference in the lipoprotein profile (P<.05) between mice with endometriosis and sham mice. The serum levels of inflammatory cytokines interleukin 1 alpha, interleukin 6, interferon gamma, and vascular endothelial growth factor were significantly (P<.05)increased in the endometriosis mice. CONCLUSION Our study used a murine model to determine the effect of endometriosis on atherosclerosis. Inflammation-related cytokines interleukin 1 alpha, interleukin 6, interferon gamma, and vascular endothelial growth factor (angiogenic factor) released by endometriotic lesions may contribute to the increased cardiovascular risks in women with endometriosis. To reduce the risk of cardiovascular disease, early identification and treatment of endometriosis are essential. Future treatments targeting inflammatory cytokines may help reduce the long-term risk of cardiovascular disease in women with endometriosis.
Collapse
Affiliation(s)
- Ramanaiah Mamillapalli
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, CT.
| | - Nikoletta Toffoloni
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, CT
| | - Shutaro Habata
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, CT
| | - Huang Qunhua
- Department of Surgery (Cardiac Surgery), Yale School of Medicine, New Haven, CT
| | - Rula Atwani
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, CT
| | - Nina Stachenfeld
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, CT
| | - Hugh S Taylor
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, CT.
| |
Collapse
|
8
|
Xiang P, Blanchard V, Francis GA. Smooth Muscle Cell—Macrophage Interactions Leading to Foam Cell Formation in Atherosclerosis: Location, Location, Location. Front Physiol 2022; 13:921597. [PMID: 35795646 PMCID: PMC9251363 DOI: 10.3389/fphys.2022.921597] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Accepted: 05/30/2022] [Indexed: 11/13/2022] Open
Abstract
Cholesterol-overloaded cells or “foam cells” in the artery wall are the biochemical hallmark of atherosclerosis, and are responsible for much of the growth, inflammation and susceptibility to rupture of atherosclerotic lesions. While it has previously been thought that macrophages are the main contributor to the foam cell population, recent evidence indicates arterial smooth muscle cells (SMCs) are the source of the majority of foam cells in both human and murine atherosclerosis. This review outlines the timeline, site of appearance and proximity of SMCs and macrophages with lipids in human and mouse atherosclerosis, and likely interactions between SMCs and macrophages that promote foam cell formation and removal by both cell types. An understanding of these SMC-macrophage interactions in foam cell formation and regression is expected to provide new therapeutic targets to reduce the burden of atherosclerosis for the prevention of coronary heart disease, stroke and peripheral vascular disease.
Collapse
|
9
|
Evans BR, Yerly A, van der Vorst EPC, Baumgartner I, Bernhard SM, Schindewolf M, Döring Y. Inflammatory Mediators in Atherosclerotic Vascular Remodeling. Front Cardiovasc Med 2022; 9:868934. [PMID: 35600479 PMCID: PMC9114307 DOI: 10.3389/fcvm.2022.868934] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 04/11/2022] [Indexed: 12/23/2022] Open
Abstract
Atherosclerotic vascular disease remains the most common cause of ischemia, myocardial infarction, and stroke. Vascular function is determined by structural and functional properties of the arterial vessel wall, which consists of three layers, namely the adventitia, media, and intima. Key cells in shaping the vascular wall architecture and warranting proper vessel function are vascular smooth muscle cells in the arterial media and endothelial cells lining the intima. Pathological alterations of this vessel wall architecture called vascular remodeling can lead to insufficient vascular function and subsequent ischemia and organ damage. One major pathomechanism driving this detrimental vascular remodeling is atherosclerosis, which is initiated by endothelial dysfunction allowing the accumulation of intimal lipids and leukocytes. Inflammatory mediators such as cytokines, chemokines, and modified lipids further drive vascular remodeling ultimately leading to thrombus formation and/or vessel occlusion which can cause major cardiovascular events. Although it is clear that vascular wall remodeling is an elementary mechanism of atherosclerotic vascular disease, the diverse underlying pathomechanisms and its consequences are still insufficiently understood.
Collapse
Affiliation(s)
- Bryce R. Evans
- Division of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Anaïs Yerly
- Division of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Emiel P. C. van der Vorst
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich (LMU), Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
- Institute for Molecular Cardiovascular Research (IMCAR) and Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, Aachen, Germany
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht, Netherlands
| | - Iris Baumgartner
- Division of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Sarah Maike Bernhard
- Division of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Marc Schindewolf
- Division of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Yvonne Döring
- Division of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich (LMU), Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
- *Correspondence: Yvonne Döring
| |
Collapse
|
10
|
Alahmadi A, Ramji DP. Monitoring Modified Lipoprotein Uptake and Macropinocytosis Associated with Macrophage Foam Cell Formation. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2419:247-255. [PMID: 35237968 DOI: 10.1007/978-1-0716-1924-7_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Macrophage foam cell formation plays a crucial role in the initiation and progression of atherosclerosis. Macrophages uptake native and modified low density lipoprotein (LDL) through either receptor-dependent or receptor-independent mechanisms to transform into lipid laden foam cells. Foam cells are involved in the formation of fatty streak that is seen during the early stages of atherosclerosis development and therefore represents a promising therapeutic target. Normal or modified lipoproteins labeled with fluorescent dyes such as 1,1'-dioctadecyl-3-3-3',3'-tetramethylindocarbocyanine perchlorate (Dil) are often used to monitor their internalization during foam cell formation. In addition, the fluorescent dye Lucifer Yellow (LY) is widely used as a marker for macropinocytosis activity. In this chapter, we describe established methods for monitoring modified lipoprotein uptake and macropinocytosis during macrophage foam cell formation.
Collapse
Affiliation(s)
- Alaa Alahmadi
- Cardiff School of Biosciences, Cardiff University, Cardiff, UK.
| | - Dipak P Ramji
- Cardiff School of Biosciences, Cardiff University, Cardiff, UK
| |
Collapse
|
11
|
Siddiqui H, Yevstigneyev N, Madani G, McCormick S. Approaches to Visualising Endocytosis of LDL-Related Lipoproteins. Biomolecules 2022; 12:biom12020158. [PMID: 35204658 PMCID: PMC8961563 DOI: 10.3390/biom12020158] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 12/20/2021] [Accepted: 01/12/2022] [Indexed: 02/04/2023] Open
Abstract
Endocytosis is the process by which molecules are actively transported into cells. It can take on a variety of forms depending on the cellular machinery involved ranging from specific receptor-mediated endocytosis to the less selective and actin-driven macropinocytosis. The plasma lipoproteins, which deliver lipids and other cargo to cells, have been intensely studied with respect to their endocytic uptake. One of the first molecules to be visualised undergoing endocytosis via a receptor-mediated, clathrin-dependent pathway was low-density lipoprotein (LDL). The LDL molecule has subsequently been shown to be internalised through multiple endocytic pathways. Dissecting the pathways of lipoprotein endocytosis has been crucial to understanding the regulation of plasma lipid levels and how lipids enter cells in the arterial wall to promote atherosclerosis. It has also aided understanding of the dysregulation that occurs in plasma lipid levels when molecules involved in uptake are defective, as is the case in familial hypercholesterolemia (FH). The aim of this review is to outline the many endocytic pathways utilised for lipoprotein uptake. It explores the various experimental approaches that have been applied to visualise lipoprotein endocytosis with an emphasis on LDL and its more complex counterpart, lipoprotein(a) [Lp(a)]. Finally, we look at new developments in lipoprotein visualisation that hold promise for scrutinising endocytic pathways to finer detail in the future.
Collapse
Affiliation(s)
- Halima Siddiqui
- Department of Biochemistry, School of Biomedical Sciences, University of Otago, Dunedin 9016, New Zealand; (H.S.); (N.Y.); (G.M.)
- HeartOtago, School of Biomedical Sciences, University of Otago, Dunedin 9016, New Zealand
| | - Nikita Yevstigneyev
- Department of Biochemistry, School of Biomedical Sciences, University of Otago, Dunedin 9016, New Zealand; (H.S.); (N.Y.); (G.M.)
- HeartOtago, School of Biomedical Sciences, University of Otago, Dunedin 9016, New Zealand
| | - Golnoush Madani
- Department of Biochemistry, School of Biomedical Sciences, University of Otago, Dunedin 9016, New Zealand; (H.S.); (N.Y.); (G.M.)
- HeartOtago, School of Biomedical Sciences, University of Otago, Dunedin 9016, New Zealand
| | - Sally McCormick
- Department of Biochemistry, School of Biomedical Sciences, University of Otago, Dunedin 9016, New Zealand; (H.S.); (N.Y.); (G.M.)
- HeartOtago, School of Biomedical Sciences, University of Otago, Dunedin 9016, New Zealand
- Correspondence:
| |
Collapse
|
12
|
Jin P, Gao D, Cong G, Yan R, Jia S. Role of PCSK9 in Homocysteine-Accelerated Lipid Accumulation in Macrophages and Atherosclerosis in ApoE -/- Mice. Front Cardiovasc Med 2021; 8:746989. [PMID: 34660746 PMCID: PMC8517151 DOI: 10.3389/fcvm.2021.746989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 09/03/2021] [Indexed: 11/30/2022] Open
Abstract
Background: Homocysteine (Hcy) has been established as an independent risk factor for atherosclerosis, and the involvement of hyperhomocysteinemia (HHcy) in atherosclerotic lesions is complex. Proprotein convertase subtilisin kexin 9 (PCSK9) has vital importance in lipid metabolism, and its inhibitors have intense lipid-lowering and anti-atherosclerotic effects. However, the underlying effect of PCSK9 on HHcy-accelerated dyslipidemia of macrophages is still uncertain. The purpose of this study was to investigate the potential role of PCSK9 in Hcy-induced lipid accumulation and atherosclerotic lesions. Methods:In vitro, gene and protein expressions were assessed by real-time quantitative PCR and western blot in THP-1 macrophages with Hcy incubation. Lipid accumulation and cholesterol efflux were evaluated with Hcy treatment. SBC-115076 was used to examine the role of PCSK9 in ATP-binding cassette transporter A1 and G1 (ABCA1 and ABCG1)-dependent cholesterol efflux. In vivo, lesion area, lipid deposition and collagen contents were determined in aortas of ApoE−/− mice under a methionine diet. SBC-115076 was subcutaneously injected to explore the potential effects of PCSK9 inhibition on alleviating the severity of HHcy-related atherosclerotic lesions. Results: In THP-1 macrophages, Hcy dose- and time-dependently promoted PCSK9 gene and protein levels without regulating the translation of Low-density lipoprotein receptor (LDLR). SBC-115076 used to inhibit PCSK9 largely alleviated lipid accumulation and reversed the cholesterol efflux to apolipoprotein-I(apoA-I) and high-density lipoprotein (HDL) mediated by ABCA1 and ABCG1. In ApoE−/− mice, methionine diet induced HHcy caused larger lesion area and more lipid accumulation in aortic roots. SBC-115076 reduced atherosclerotic severity by reducing the lesion area and lipid accumulation and increasing expressions of ABCA1 and ABCG1 in macrophages from atherosclerotic plaque. In addition, SBC-115076 decreased plasma Hcy level and lipid profiles significantly. Conclusion: PCSK9 promoted lipid accumulation via inhibiting cholesterol efflux mediated by ABCA1 and ABCG1 from macrophages and accelerated atherosclerotic lesions under HHcy treatment. Inhibiting PCSK9 may have anti-atherogenic properties in HHcy-accelerated atherosclerosis.
Collapse
Affiliation(s)
- Ping Jin
- Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Dengfeng Gao
- Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Guangzhi Cong
- Heart Center and Cardiovascular Institute, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Ru Yan
- Heart Center and Cardiovascular Institute, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Shaobin Jia
- Heart Center and Cardiovascular Institute, General Hospital of Ningxia Medical University, Yinchuan, China
| |
Collapse
|
13
|
Lefevre JG, Koh YWH, Wall AA, Condon ND, Stow JL, Hamilton NA. LLAMA: a robust and scalable machine learning pipeline for analysis of large scale 4D microscopy data: analysis of cell ruffles and filopodia. BMC Bioinformatics 2021; 22:410. [PMID: 34412593 PMCID: PMC8375126 DOI: 10.1186/s12859-021-04324-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 08/10/2021] [Indexed: 11/24/2022] Open
Abstract
Background With recent advances in microscopy, recordings of cell behaviour can result in terabyte-size datasets. The lattice light sheet microscope (LLSM) images cells at high speed and high 3D resolution, accumulating data at 100 frames/second over hours, presenting a major challenge for interrogating these datasets. The surfaces of vertebrate cells can rapidly deform to create projections that interact with the microenvironment. Such surface projections include spike-like filopodia and wave-like ruffles on the surface of macrophages as they engage in immune surveillance. LLSM imaging has provided new insights into the complex surface behaviours of immune cells, including revealing new types of ruffles. However, full use of these data requires systematic and quantitative analysis of thousands of projections over hundreds of time steps, and an effective system for analysis of individual structures at this scale requires efficient and robust methods with minimal user intervention. Results We present LLAMA, a platform to enable systematic analysis of terabyte-scale 4D microscopy datasets. We use a machine learning method for semantic segmentation, followed by a robust and configurable object separation and tracking algorithm, generating detailed object level statistics. Our system is designed to run on high-performance computing to achieve high throughput, with outputs suitable for visualisation and statistical analysis. Advanced visualisation is a key element of LLAMA: we provide a specialised tool which supports interactive quality control, optimisation, and output visualisation processes to complement the processing pipeline. LLAMA is demonstrated in an analysis of macrophage surface projections, in which it is used to i) discriminate ruffles induced by lipopolysaccharide (LPS) and macrophage colony stimulating factor (CSF-1) and ii) determine the autonomy of ruffle morphologies. Conclusions LLAMA provides an effective open source tool for running a cell microscopy analysis pipeline based on semantic segmentation, object analysis and tracking. Detailed numerical and visual outputs enable effective statistical analysis, identifying distinct patterns of increased activity under the two interventions considered in our example analysis. Our system provides the capacity to screen large datasets for specific structural configurations. LLAMA identified distinct features of LPS and CSF-1 induced ruffles and it identified a continuity of behaviour between tent pole ruffling, wave-like ruffling and filopodia deployment. Supplementary Information The online version contains supplementary material available at 10.1186/s12859-021-04324-z.
Collapse
Affiliation(s)
- James G Lefevre
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Yvette W H Koh
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Adam A Wall
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Nicholas D Condon
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Jennifer L Stow
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Nicholas A Hamilton
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia. .,Research Computing Centre, The University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
14
|
Chaves LD, Abyad S, Honan AM, Bryniarski MA, McSkimming DI, Stahura CM, Wells SC, Ruszaj DM, Morris ME, Quigg RJ, Yacoub R. Unconjugated p-cresol activates macrophage macropinocytosis leading to increased LDL uptake. JCI Insight 2021; 6:144410. [PMID: 33914709 PMCID: PMC8262368 DOI: 10.1172/jci.insight.144410] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 04/28/2021] [Indexed: 01/03/2023] Open
Abstract
Patients with chronic kidney disease (CKD) and end-stage renal disease suffer from increased cardiovascular events and cardiac mortality. Prior studies have demonstrated that a portion of this enhanced risk can be attributed to the accumulation of microbiota-derived toxic metabolites, with most studies focusing on the sulfonated form of p-cresol (PCS). However, unconjugated p-cresol (uPC) itself was never assessed due to rapid and extensive first-pass metabolism that results in negligible serum concentrations of uPC. These reports thus failed to consider the host exposure to uPC prior to hepatic metabolism. In the current study, not only did we measure the effect of altering the intestinal microbiota on lipid accumulation in coronary arteries, but we also examined macrophage lipid uptake and handling pathways in response to uPC. We found that atherosclerosis-prone mice fed a high-fat diet exhibited significantly higher coronary artery lipid deposits upon receiving fecal material from CKD mice. Furthermore, treatment with uPC increased total cholesterol, triglycerides, and hepatic and aortic fatty deposits in non-CKD mice. Studies employing an in vitro macrophage model demonstrated that uPC exposure increased apoptosis whereas PCS did not. Additionally, uPC exhibited higher potency than PCS to stimulate LDL uptake and only uPC induced endocytosis- and pinocytosis-related genes. Pharmacological inhibition of varying cholesterol influx and efflux systems indicated that uPC increased macrophage LDL uptake by activating macropinocytosis. Overall, these findings indicate that uPC itself had a distinct effect on macrophage biology that might have contributed to increased cardiovascular risk in patients with CKD.
Collapse
Affiliation(s)
- Lee D Chaves
- Department of Medicine, Division of Nephrology, Jacobs School of Medicine and Biomedical Sciences, and.,Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo, New York, USA
| | - Sham Abyad
- Department of Medicine, Division of Nephrology, Jacobs School of Medicine and Biomedical Sciences, and
| | - Amanda M Honan
- Department of Medicine, Division of Nephrology, Jacobs School of Medicine and Biomedical Sciences, and
| | - Mark A Bryniarski
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo, New York, USA
| | - Daniel I McSkimming
- Department of Medicine, Bioinformatics and Computational Biology Core, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Corrine M Stahura
- Department of Medicine, Division of Nephrology, Jacobs School of Medicine and Biomedical Sciences, and
| | - Steven C Wells
- Department of Medicine, Division of Nephrology, Jacobs School of Medicine and Biomedical Sciences, and
| | - Donna M Ruszaj
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo, New York, USA
| | - Marilyn E Morris
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo, New York, USA
| | - Richard J Quigg
- Department of Medicine, Division of Nephrology, Jacobs School of Medicine and Biomedical Sciences, and
| | - Rabi Yacoub
- Department of Medicine, Division of Nephrology, Jacobs School of Medicine and Biomedical Sciences, and
| |
Collapse
|
15
|
Macrophages bind LDL using heparan sulfate and the perlecan protein core. J Biol Chem 2021; 296:100520. [PMID: 33684447 PMCID: PMC8027565 DOI: 10.1016/j.jbc.2021.100520] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 03/03/2021] [Accepted: 03/04/2021] [Indexed: 12/25/2022] Open
Abstract
The retention of low-density lipoprotein (LDL) is a key process in the pathogenesis of atherosclerosis and largely mediated via smooth-muscle cell-derived extracellular proteoglycans including the glycosaminoglycan chains. Macrophages can also internalize lipids via complexes with proteoglycans. However, the role of polarized macrophage-derived proteoglycans in binding LDL is unknown and important to advance our understanding of the pathogenesis of atherosclerosis. We therefore examined the identity of proteoglycans, including the pendent glycosaminoglycans, produced by polarized macrophages to gain insight into the molecular basis for LDL binding. Using the quartz crystal microbalance with dissipation monitoring technique, we established that classically activated macrophage (M1)- and alternatively activated macrophage (M2)-derived proteoglycans bind LDL via both the protein core and heparan sulfate (HS) in vitro. Among the proteoglycans secreted by macrophages, we found perlecan was the major protein core that bound LDL. In addition, we identified perlecan in the necrotic core as well as the fibrous cap of advanced human atherosclerotic lesions in the same regions as HS and colocalized with M2 macrophages, suggesting a functional role in lipid retention in vivo. These findings suggest that macrophages may contribute to LDL retention in the plaque by the production of proteoglycans; however, their contribution likely depends on both their phenotype within the plaque and the presence of enzymes, such as heparanase, that alter the secreted protein structure.
Collapse
|
16
|
Li J, Meng Q, Fu Y, Yu X, Ji T, Chao Y, Chen Q, Li Y, Bian H. Novel insights: Dynamic foam cells derived from the macrophage in atherosclerosis. J Cell Physiol 2021; 236:6154-6167. [PMID: 33507545 DOI: 10.1002/jcp.30300] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 12/22/2020] [Accepted: 01/15/2021] [Indexed: 12/12/2022]
Abstract
Atherosclerosis can be regarded as a chronic disease derived from the interaction between disordered lipoproteins and an unsuitable immune response. The evolution of foam cells is not only a significant pathological change in the early stage of atherosclerosis but also a key stage in the occurrence and development of atherosclerosis. The formation of foam cells is mainly caused by the imbalance among lipids uptake, lipids treatment, and reverse cholesterol transport. Although a large number of studies have summarized the source of foam cells and the mechanism of foam cells formation, we propose a new idea about foam cells in atherosclerosis. Rather than an isolated microenvironment, the macrophage multiple lipid uptake pathways, lipid internalization, lysosome, mitochondria, endoplasmic reticulum, neutral cholesterol ester hydrolase (NCEH), acyl-coenzyme A-cholesterol acyltransferase (ACAT), and reverse cholesterol transport are mutually influential, and form a dynamic process under multi-factor regulation. The macrophage takes on different uptake lipid statuses depending on multiple uptake pathways and intracellular lipids, lipid metabolites versus pro-inflammatory factors. Except for NCEH and ACAT, the lipid internalization of macrophages also depends on multicellular organelles including the lysosome, mitochondria, and endoplasmic reticulum, which are associated with each other. A dynamic balance between esterification and hydrolysis of cholesterol for macrophages is essential for physiology and pathology. Therefore, we propose that the foam cell in the process of atherosclerosis may be dynamic under multi-factor regulation, and collate this study to provide a holistic and dynamic idea of the foam cell.
Collapse
Affiliation(s)
- Jun Li
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Qinghai Meng
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yu Fu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xichao Yu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Tingting Ji
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ying Chao
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Qi Chen
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yu Li
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Huimin Bian
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Key Laboratory of Therapeutic Material of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
17
|
Golforoush P, Yellon DM, Davidson SM. Mouse models of atherosclerosis and their suitability for the study of myocardial infarction. Basic Res Cardiol 2020; 115:73. [PMID: 33258000 PMCID: PMC7704510 DOI: 10.1007/s00395-020-00829-5] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 10/28/2020] [Indexed: 12/17/2022]
Abstract
Atherosclerotic plaques impair vascular function and can lead to arterial obstruction and tissue ischaemia. Rupture of an atherosclerotic plaque within a coronary artery can result in an acute myocardial infarction, which is responsible for significant morbidity and mortality worldwide. Prompt reperfusion can salvage some of the ischaemic territory, but ischaemia and reperfusion (IR) still causes substantial injury and is, therefore, a therapeutic target for further infarct limitation. Numerous cardioprotective strategies have been identified that can limit IR injury in animal models, but none have yet been translated effectively to patients. This disconnect prompts an urgent re-examination of the experimental models used to study IR. Since coronary atherosclerosis is the most prevalent morbidity in this patient population, and impairs coronary vessel function, it is potentially a major confounder in cardioprotective studies. Surprisingly, most studies suggest that atherosclerosis does not have a major impact on cardioprotection in mouse models. However, a major limitation of atherosclerotic animal models is that the plaques usually manifest in the aorta and proximal great vessels, and rarely in the coronary vessels. In this review, we examine the commonly used mouse models of atherosclerosis and their effect on coronary artery function and infarct size. We conclude that none of the commonly used strains of mice are ideal for this purpose; however, more recently developed mouse models of atherosclerosis fulfil the requirement for coronary artery lesions, plaque rupture and lipoprotein patterns resembling the human profile, and may enable the identification of therapeutic interventions more applicable in the clinical setting.
Collapse
MESH Headings
- Animals
- Aortic Diseases/complications
- Aortic Diseases/genetics
- Aortic Diseases/metabolism
- Aortic Diseases/pathology
- Atherosclerosis/complications
- Atherosclerosis/genetics
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Coronary Artery Disease/complications
- Coronary Artery Disease/genetics
- Coronary Artery Disease/metabolism
- Coronary Artery Disease/pathology
- Diet, High-Fat
- Disease Models, Animal
- Genetic Predisposition to Disease
- Mice, Knockout, ApoE
- Myocardial Infarction/etiology
- Myocardial Infarction/genetics
- Myocardial Infarction/metabolism
- Myocardial Infarction/pathology
- Myocardium/pathology
- Phenotype
- Plaque, Atherosclerotic
- Receptors, LDL/deficiency
- Receptors, LDL/genetics
- Rupture, Spontaneous
- Scavenger Receptors, Class B/deficiency
- Scavenger Receptors, Class B/genetics
- Species Specificity
Collapse
Affiliation(s)
- Pelin Golforoush
- The Hatter Cardiovascular Institute, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Derek M Yellon
- The Hatter Cardiovascular Institute, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, 67 Chenies Mews, London, WC1E 6HX, UK.
| |
Collapse
|
18
|
Su X, Peng D. New insight into sortilin in controlling lipid metabolism and the risk of atherogenesis. Biol Rev Camb Philos Soc 2020; 95:232-243. [PMID: 31625271 DOI: 10.1111/brv.12561] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 09/16/2019] [Accepted: 09/18/2019] [Indexed: 01/24/2023]
Affiliation(s)
- Xin Su
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Daoquan Peng
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| |
Collapse
|
19
|
Doodnauth SA, Grinstein S, Maxson ME. Constitutive and stimulated macropinocytosis in macrophages: roles in immunity and in the pathogenesis of atherosclerosis. Philos Trans R Soc Lond B Biol Sci 2020; 374:20180147. [PMID: 30967001 DOI: 10.1098/rstb.2018.0147] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Macrophages respond to several stimuli by forming florid membrane ruffles that lead to fluid uptake by macropinocytosis. This type of induced macropinocytosis, executed by a variety of non-malignant and malignant cells, is initiated by transmembrane receptors and is involved in nutrient acquisition and mTOR signalling. However, macrophages also perform a unique type of constitutive ruffling and macropinocytosis that is dependent on the presence of extracellular calcium. Calcium-sensing receptors are responsible for this activity. This distinct form of macropinocytosis enables macrophages to continuously sample their microenvironment for antigenic molecules and for pathogen- and danger-associated molecular patterns, as part of their immune surveillance functions. Interestingly, even within the monocyte lineage, there are differences in macropinocytic ability that reflect the polarized functional roles of distinct macrophage subsets. This review discusses the shared and distinct features of both induced and constitutive macropinocytosis displayed by the macrophage lineage and their roles in physiology, immunity and pathophysiology. In particular, we analyse the role of macropinocytosis in the uptake of modified low-density lipoprotein (LDL) and its contribution to foam cell and atherosclerotic plaque formation. We propose a combined role of scavenger receptors and constitutive macropinocytosis in oxidized LDL uptake, a process we have termed 'receptor-assisted macropinocytosis'. This article is part of the Theo Murphy meeting issue 'Macropinocytosis'.
Collapse
Affiliation(s)
- Sasha A Doodnauth
- 1 Princess Margaret Cancer Center, University Health Network , Toronto, ON , Canada M5G 1L7.,2 Department of Medical Biophysics, University of Toronto , Toronto, ON , Canada M5G 1L7
| | - Sergio Grinstein
- 3 Program in Cell Biology, Hospital for Sick Children , 686 Bay Street, Toronto, ON , Canada M5G 0A4.,4 Department of Biochemistry, University of Toronto , 1 King's Circle, Toronto, ON , Canada M5S 1A8.,5 Keenan Research Centre of the Li Ka Shing Knowledge Institute , St. Michael's Hospital, 290 Victoria Street, Toronto, ON , Canada M5C 1N8
| | - Michelle E Maxson
- 3 Program in Cell Biology, Hospital for Sick Children , 686 Bay Street, Toronto, ON , Canada M5G 0A4
| |
Collapse
|
20
|
Brophy ML, Dong Y, Tao H, Yancey PG, Song K, Zhang K, Wen A, Wu H, Lee Y, Malovichko MV, Sithu SD, Wong S, Yu L, Kocher O, Bischoff J, Srivastava S, Linton MF, Ley K, Chen H. Myeloid-Specific Deletion of Epsins 1 and 2 Reduces Atherosclerosis by Preventing LRP-1 Downregulation. Circ Res 2019; 124:e6-e19. [PMID: 30595089 DOI: 10.1161/circresaha.118.313028] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
RATIONALE Atherosclerosis is, in part, caused by immune and inflammatory cell infiltration into the vascular wall, leading to enhanced inflammation and lipid accumulation in the aortic endothelium. Understanding the molecular mechanisms underlying this disease is critical for the development of new therapies. Our recent studies demonstrate that epsins, a family of ubiquitin-binding endocytic adaptors, are critical regulators of atherogenicity. Given the fundamental contribution lesion macrophages make to fuel atherosclerosis, whether and how myeloid-specific epsins promote atherogenesis is an open and significant question. OBJECTIVE We will determine the role of myeloid-specific epsins in regulating lesion macrophage function during atherosclerosis. METHODS AND RESULTS We engineered myeloid cell-specific epsins double knockout mice (LysM-DKO) on an ApoE-/- background. On Western diet, these mice exhibited marked decrease in atherosclerotic lesion formation, diminished immune and inflammatory cell content in aortas, and reduced necrotic core content but increased smooth muscle cell content in aortic root sections. Epsins deficiency hindered foam cell formation and suppressed proinflammatory macrophage phenotype but increased efferocytosis and anti-inflammatory macrophage phenotype in primary macrophages. Mechanistically, we show that epsin loss specifically increased total and surface levels of LRP-1 (LDLR [low-density lipoprotein receptor]-related protein 1), an efferocytosis receptor with antiatherosclerotic properties. We further show that epsin and LRP-1 interact via epsin's ubiquitin-interacting motif domain. ox-LDL (oxidized LDL) treatment increased LRP-1 ubiquitination, subsequent binding to epsin, and its internalization from the cell surface, suggesting that epsins promote the ubiquitin-dependent internalization and downregulation of LRP-1. Crossing ApoE-/-/LysM-DKO mice onto an LRP-1 heterozygous background restored, in part, atherosclerosis, suggesting that epsin-mediated LRP-1 downregulation in macrophages plays a pivotal role in propelling atherogenesis. CONCLUSIONS Myeloid epsins promote atherogenesis by facilitating proinflammatory macrophage recruitment and inhibiting efferocytosis in part by downregulating LRP-1, implicating that targeting epsins in macrophages may serve as a novel therapeutic strategy to treat atherosclerosis.
Collapse
Affiliation(s)
- Megan L Brophy
- From the Vascular Biology Program and Department of Surgery, Boston Children's Hospital (M.L.B., Y.D., K.S., K.Z., A.W., H.W., Y.L., S.W., L.Y., J.B., H.C.), Harvard Medical School, MA.,Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center (M.L.B.)
| | - Yunzhou Dong
- From the Vascular Biology Program and Department of Surgery, Boston Children's Hospital (M.L.B., Y.D., K.S., K.Z., A.W., H.W., Y.L., S.W., L.Y., J.B., H.C.), Harvard Medical School, MA
| | - Huan Tao
- Atherosclerosis Research Unit, Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (H.T., P.G.Y., M.F.L.)
| | - Patricia G Yancey
- Atherosclerosis Research Unit, Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (H.T., P.G.Y., M.F.L.)
| | - Kai Song
- From the Vascular Biology Program and Department of Surgery, Boston Children's Hospital (M.L.B., Y.D., K.S., K.Z., A.W., H.W., Y.L., S.W., L.Y., J.B., H.C.), Harvard Medical School, MA
| | - Kun Zhang
- From the Vascular Biology Program and Department of Surgery, Boston Children's Hospital (M.L.B., Y.D., K.S., K.Z., A.W., H.W., Y.L., S.W., L.Y., J.B., H.C.), Harvard Medical School, MA.,Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China (K.Z.)
| | - Aiyun Wen
- From the Vascular Biology Program and Department of Surgery, Boston Children's Hospital (M.L.B., Y.D., K.S., K.Z., A.W., H.W., Y.L., S.W., L.Y., J.B., H.C.), Harvard Medical School, MA
| | - Hao Wu
- From the Vascular Biology Program and Department of Surgery, Boston Children's Hospital (M.L.B., Y.D., K.S., K.Z., A.W., H.W., Y.L., S.W., L.Y., J.B., H.C.), Harvard Medical School, MA
| | - Yang Lee
- From the Vascular Biology Program and Department of Surgery, Boston Children's Hospital (M.L.B., Y.D., K.S., K.Z., A.W., H.W., Y.L., S.W., L.Y., J.B., H.C.), Harvard Medical School, MA
| | - Marina V Malovichko
- Division of Cardiovascular Medicine, Department of Medicine, University of Louisville, KY (M.V.M., S.D.S., S.S.)
| | - Srinivas D Sithu
- Division of Cardiovascular Medicine, Department of Medicine, University of Louisville, KY (M.V.M., S.D.S., S.S.)
| | - Scott Wong
- From the Vascular Biology Program and Department of Surgery, Boston Children's Hospital (M.L.B., Y.D., K.S., K.Z., A.W., H.W., Y.L., S.W., L.Y., J.B., H.C.), Harvard Medical School, MA
| | - Lili Yu
- From the Vascular Biology Program and Department of Surgery, Boston Children's Hospital (M.L.B., Y.D., K.S., K.Z., A.W., H.W., Y.L., S.W., L.Y., J.B., H.C.), Harvard Medical School, MA
| | - Olivier Kocher
- Department of Pathology and Center for Vascular Biology Research, Beth Israel Medical Deaconess Medical Center (O.K.), Harvard Medical School, MA
| | - Joyce Bischoff
- From the Vascular Biology Program and Department of Surgery, Boston Children's Hospital (M.L.B., Y.D., K.S., K.Z., A.W., H.W., Y.L., S.W., L.Y., J.B., H.C.), Harvard Medical School, MA
| | - Sanjay Srivastava
- Division of Cardiovascular Medicine, Department of Medicine, University of Louisville, KY (M.V.M., S.D.S., S.S.)
| | - MacRae F Linton
- Atherosclerosis Research Unit, Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (H.T., P.G.Y., M.F.L.)
| | - Klaus Ley
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, CA (K.L.)
| | - Hong Chen
- From the Vascular Biology Program and Department of Surgery, Boston Children's Hospital (M.L.B., Y.D., K.S., K.Z., A.W., H.W., Y.L., S.W., L.Y., J.B., H.C.), Harvard Medical School, MA
| |
Collapse
|
21
|
Interleukin 10 promotes macrophage uptake of HDL and LDL by stimulating fluid-phase endocytosis. Biochim Biophys Acta Mol Cell Biol Lipids 2019; 1865:158537. [PMID: 31676439 DOI: 10.1016/j.bbalip.2019.158537] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 09/06/2019] [Accepted: 09/19/2019] [Indexed: 01/01/2023]
Abstract
OBJECTIVE Highly elevated plasma levels of interleukin-10 (IL-10) are causally associated with "Disappearing HDL Syndrome" and low plasma LDL-cholesterol, but the underlying mechanism is poorly understood. Fluid-phase endocytosis, a process highly dependent on actin dynamics, enables cells to internalize relatively high amounts of extracellular fluids and solutes. We sought to investigate whether IL-10 induces lipoprotein uptake by fluid-phase endocytosis in macrophages. METHODS AND RESULTS Macrophages (RAW264.7, Kupffer and human) were incubated with vehicle (PBS) or IL-10 (20 ng/ml) for 7 days. Uptake of HDL, LDL, and/or fluid-phase endocytosis probes (albumin-Alexa680®, 70 kDa FITC-Dextran and Lucifer Yellow, LY) was evaluated by FACS. Intracellular cofilin and phosphorylated cofilin (p-cofilin) levels were determined by immunoblotting. Macrophage uptake of lipoproteins and probes was non-saturable and increased after IL-10 incubation (p < 0.0001). Furthermore, pre-incubation with fluid-phase endocytosis inhibitors (LY294002, Latrunculin A, and Amiloride) significantly reduced uptake (p < 0.05). IL-10 increased the cofilin/p-cofilin ratio (p = 0.021), signifying increased cofilin activation and hence filamentous actin. Consistently, phalloidin staining revealed increased filamentous actin in macrophages after IL-10 treatment (p = 0.0018). Finally, RNA-seq analysis demonstrated enrichment of gene sets related to actin filament dynamics, membrane ruffle formation and endocytosis in IL-10-treated macrophages (p < 0.05). IL-10 did not alter mRNA levels of Ldlr, Vldlr, Scarb1, Cd36 or Lrp1. In primary human monocyte-derived macrophages and murine Kupffer cells, IL-10 incubation also increased uptake of lipoproteins, albumin and LY (p < 0.01). CONCLUSIONS Interleukin-10 induces the uptake of HDL and LDL by fluid-phase endocytosis by increasing actin-filament rearrangement in macrophages, thus providing a plausible mechanism contributing to "Disappearing HDL Syndrome".
Collapse
|
22
|
Miyazaki T, Miyazaki A. Impact of Dysfunctional Protein Catabolism on Macrophage Cholesterol Handling. Curr Med Chem 2019; 26:1631-1643. [PMID: 29589525 DOI: 10.2174/0929867325666180326165234] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 11/24/2017] [Accepted: 12/26/2017] [Indexed: 12/18/2022]
Abstract
Protein catabolism in macrophages, which is accomplished mainly through autophagy- lysosomal degradation, ubiquitin-proteasome system, and calpains, is disturbed in atheroprone vessels. Moreover, growing evidence suggests that defects in protein catabolism interfere with cholesterol handling in macrophages. Indeed, decreases in autophagy facilitate the deposition of cholesterol in atheroprone macrophages and the subsequent development of vulnerable atherosclerotic plaques due to impaired catabolism of lipid droplets and limited efferocytic clearance of dead cells. The proteasome is responsible for the degradation of ATP-binding cassette transporters, which leads to impaired cholesterol efflux from macrophages. Overactivation of conventional calpains contributes to excessive processing of functional proteins, thereby accelerating receptor-mediated uptake of oxidized low-density lipoproteins (LDLs) and slowing cholesterol efflux. Furthermore, calpain-6, an unconventional nonproteolytic calpain in macrophages, potentiates pinocytotic uptake of native LDL and attenuates the efferocytic clearance of dead cells. Herein, we focus on recent progress in understanding how defective protein catabolism is associated with macrophage cholesterol handling and subsequent atherogenesis.
Collapse
Affiliation(s)
- Takuro Miyazaki
- Department of Biochemistry, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan
| | - Akira Miyazaki
- Department of Biochemistry, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan
| |
Collapse
|
23
|
Håversen L, Sundelin JP, Mardinoglu A, Rutberg M, Ståhlman M, Wilhelmsson U, Hultén LM, Pekny M, Fogelstrand P, Bentzon JF, Levin M, Borén J. Vimentin deficiency in macrophages induces increased oxidative stress and vascular inflammation but attenuates atherosclerosis in mice. Sci Rep 2018; 8:16973. [PMID: 30451917 PMCID: PMC6242955 DOI: 10.1038/s41598-018-34659-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 09/27/2018] [Indexed: 12/14/2022] Open
Abstract
The aim was to clarify the role of vimentin, an intermediate filament protein abundantly expressed in activated macrophages and foam cells, in macrophages during atherogenesis. Global gene expression, lipid uptake, ROS, and inflammation were analyzed in bone-marrow derived macrophages from vimentin-deficient (Vim-/-) and wild-type (Vim+/+) mice. Atherosclerosis was induced in Ldlr-/- mice transplanted with Vim-/- and Vim+/+ bone marrow, and in Vim-/- and Vim+/+ mice injected with a PCSK9 gain-of-function virus. The mice were fed an atherogenic diet for 12-15 weeks. We observed impaired uptake of native LDL but increased uptake of oxLDL in Vim-/- macrophages. FACS analysis revealed increased surface expression of the scavenger receptor CD36 on Vim-/- macrophages. Vim-/- macrophages also displayed increased markers of oxidative stress, activity of the transcription factor NF-κB, secretion of proinflammatory cytokines and GLUT1-mediated glucose uptake. Vim-/- mice displayed decreased atherogenesis despite increased vascular inflammation and increased CD36 expression on macrophages in two mouse models of atherosclerosis. We demonstrate that vimentin has a strong suppressive effect on oxidative stress and that Vim-/- mice display increased vascular inflammation with increased CD36 expression on macrophages despite decreased subendothelial lipid accumulation. Thus, vimentin has a key role in regulating inflammation in macrophages during atherogenesis.
Collapse
Affiliation(s)
- Liliana Håversen
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, University of Gothenburg, and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Jeanna Perman Sundelin
- Strategic planning and operations, Cardiovascular and metabolic diseases, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Adil Mardinoglu
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
- Centre for Host-Microbiome Interactions, Dental Institute, King's College London, London, SE1 9RT, United Kingdom
| | - Mikael Rutberg
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, University of Gothenburg, and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Marcus Ståhlman
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, University of Gothenburg, and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Ulrika Wilhelmsson
- Department of Clinical Neuroscience/Center for Brain Repair, University of Gothenburg, Gothenburg, Sweden
| | - Lillemor Mattsson Hultén
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, University of Gothenburg, and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Milos Pekny
- Department of Clinical Neuroscience/Center for Brain Repair, University of Gothenburg, Gothenburg, Sweden
| | - Per Fogelstrand
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, University of Gothenburg, and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Jacob Fog Bentzon
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark, and Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Malin Levin
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, University of Gothenburg, and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Jan Borén
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, University of Gothenburg, and Sahlgrenska University Hospital, Gothenburg, Sweden.
| |
Collapse
|
24
|
Abstract
PURPOSE OF REVIEW Lipoprotein-induced intracellular lipid accumulation (foam cell formation) is a trigger of atherogenesis at the subendothelial arterial cell level. The purpose of this review is to describe the recent data related to the possible mechanisms of LDL-induced formation of lipid-laden foam cells and their role in the onset and development of atherosclerotic lesion. RECENT FINDINGS The most interesting current studies are related to the factors affecting foam cell formation. SUMMARY The phenomenon of lipid accumulation in cultured cells became the basis for creating a cellular test system that has already been successfully applied for development of drugs possessing direct antiatherosclerotic activity, and then the efficacy of these drugs was demonstrated in clinical studies. Moreover, this test system could be used for diagnostic assessing lipoproteins atherogenicity.
Collapse
Affiliation(s)
- Alexander N Orekhov
- Laboratory of Angiopatology, Institute of General Pathology and Pathophysiology
- Institute for Atherosclerosis Research, Skolkovo Innovative Center, Moscow, Russia
| |
Collapse
|
25
|
Wang X, Gray Z, Willette-Brown J, Zhu F, Shi G, Jiang Q, Song NY, Dong L, Hu Y. Macrophage inducible nitric oxide synthase circulates inflammation and promotes lung carcinogenesis. Cell Death Discov 2018; 4:46. [PMID: 29844930 PMCID: PMC5967330 DOI: 10.1038/s41420-018-0046-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 02/13/2018] [Accepted: 02/28/2018] [Indexed: 12/26/2022] Open
Abstract
Human lung squamous cell carcinoma (SCC) is highly associated with increased pulmonary macrophage infiltration. Previously, we showed that marked pulmonary infiltrating macrophages were required for spontaneous lung SCC development in a mouse model (L-IkkαKA/KA, KA/KA) that resembles human lung SCC. Interestingly the lung SCC-associated macrophages specifically express elevated inducible nitric oxide synthase (NOS2). However, the role of macrophage NOS2 in lung carcinogenesis has not been explored. Here, we show that NOS2 ablation inhibits macrophage infiltration, fibrosis, and SCC development in the lungs of KA/KA mice. Macrophage NOS2 was found to circulate inflammation and enhance macrophage migration and survival. NOS2 promotes foamy macrophage formation characterized with impaired lipid metabolism. NOS2 null bone marrow transplantation reduces foamy macrophage numbers and carcinogenesis in KA/KA chimaeras. This finding sheds light on a new mechanism by which macrophage NOS2 increases pulmonary inflammatory responses and macrophage survival and impairs macrophage lipid metabolism, thereby promoting lung SCC formation.
Collapse
Affiliation(s)
- Xin Wang
- 1Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MA 21701 USA.,4The Respiratory Department, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong 250013 China
| | - Zane Gray
- 1Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MA 21701 USA
| | - Jami Willette-Brown
- 1Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MA 21701 USA
| | - Feng Zhu
- 1Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MA 21701 USA
| | - Gongping Shi
- 1Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MA 21701 USA
| | - Qun Jiang
- 2Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892 USA
| | - Na-Young Song
- 1Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MA 21701 USA
| | - Liang Dong
- 3Department of Respiratory Medicine, Qilu Hospital of Shandong University, 107#, Wenhua Xi Road, Jinan, 250012 Shandong China
| | - Yinling Hu
- 1Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MA 21701 USA
| |
Collapse
|
26
|
Chung TW, Choi HJ, Park MJ, Choi HJ, Lee SO, Kim KJ, Kim CH, Hong C, Kim KH, Joo M, Ha KT. The function of cancer-shed gangliosides in macrophage phenotype: involvement with angiogenesis. Oncotarget 2018; 8:4436-4448. [PMID: 28032600 PMCID: PMC5354844 DOI: 10.18632/oncotarget.13878] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 12/05/2016] [Indexed: 11/25/2022] Open
Abstract
Tumor-derived gangliosides in the tumor microenvironment are involved in the malignant progression of cancer. However, the molecular mechanisms underlying the effects of gangliosides shed from tumors on macrophage phenotype remain unknown. Here, we showed that ganglioside GM1 highly induced the activity and expression of arginase-1 (Arg-1), a major M2 macrophage marker, compared to various gangliosides in bone marrow-derived macrophages (BMDM), peritoneal macrophages and Raw264.7 macrophage cells. We found that GM1 bound to macrophage mannose receptor (MMR/CD206) and common gamma chain (γc). In addition, GM1 increased Arg-1 expression through CD206 and γc-mediated activation of Janus kinase 3 (JAK3) and signal transducer and activator of transcription- 6 (STAT-6). Interestingly, GM1-stimulated macrophages secreted monocyte chemoattractant protein-1 (MCP-1/CCL2) through a CD206/γc/STAT6-mediated signaling pathway and induced angiogenesis. Moreover, the angiogenic effect of GM1-treated macrophages was diminished by RS102895, an MCP-1 receptor (CCR2) antagonist. From these results we suggest that tumor-shed ganglioside is a secretory factor regulating the phenotype of macrophages and consequently enhancing angiogenesis.
Collapse
Affiliation(s)
- Tae-Wook Chung
- Korean Medical Research Center for Healthy Aging and Yangsan, Gyeongsangnam-do, Republic of Korea.,School of Korean Medicine, Pusan National University, Yangsan, Gyeongsangnam-do, Republic of Korea
| | - Hee-Jung Choi
- Korean Medical Research Center for Healthy Aging and Yangsan, Gyeongsangnam-do, Republic of Korea.,School of Korean Medicine, Pusan National University, Yangsan, Gyeongsangnam-do, Republic of Korea
| | - Mi-Ju Park
- Korean Medical Research Center for Healthy Aging and Yangsan, Gyeongsangnam-do, Republic of Korea
| | - Hee-Jin Choi
- Korean Medical Research Center for Healthy Aging and Yangsan, Gyeongsangnam-do, Republic of Korea.,School of Korean Medicine, Pusan National University, Yangsan, Gyeongsangnam-do, Republic of Korea
| | - Syng-Ook Lee
- Department of Food Science and Technology, Keimyung University, Daegu, Republic of Korea
| | - Keuk-Jun Kim
- Department of Clinical Pathology, TaeKyeung University, Gyeongsan, Gyeongsangbuk-do, Republic of Korea
| | - Cheorl-Ho Kim
- Department of Biological Science, Sungkyunkwan University, Suwon, Kyunggi-do, Republic of Korea
| | - Changwan Hong
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan, Gyeongsangnam-do, Republic of Korea
| | - Kyun-Ha Kim
- School of Korean Medicine, Pusan National University, Yangsan, Gyeongsangnam-do, Republic of Korea
| | - Myungsoo Joo
- Korean Medical Research Center for Healthy Aging and Yangsan, Gyeongsangnam-do, Republic of Korea.,School of Korean Medicine, Pusan National University, Yangsan, Gyeongsangnam-do, Republic of Korea
| | - Ki-Tae Ha
- Korean Medical Research Center for Healthy Aging and Yangsan, Gyeongsangnam-do, Republic of Korea.,School of Korean Medicine, Pusan National University, Yangsan, Gyeongsangnam-do, Republic of Korea
| |
Collapse
|
27
|
Head T, Daunert S, Goldschmidt-Clermont PJ. The Aging Risk and Atherosclerosis: A Fresh Look at Arterial Homeostasis. Front Genet 2017; 8:216. [PMID: 29312440 PMCID: PMC5735066 DOI: 10.3389/fgene.2017.00216] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 12/04/2017] [Indexed: 12/14/2022] Open
Abstract
A considerable volume of research over the last decade has focused on understanding the fundamental mechanisms for the progression of atherosclerosis-the underlying cause for the vast majority of all cardiovascular (CVD)-related complications. Aging is the dominant risk factor for clinically significant atherosclerotic lesion formation, yet the heightened impact of aging on the disease is not accounted for by changes in traditional risk factors, such as lack of physical activity, smoking, hypertension, hyperlipidemia, or diabetes mellitus. This review will examine the pathological and biochemical processes of atherosclerotic plaque formation and growth, with particular focus on the aging risk vis-a-vis arterial homeostasis. Particular focus will be placed on the impact of a number of important contributors to arterial homeostasis including bone marrow (BM)-derived vascular progenitor cells, differential monocyte subpopulations, and the role of cellular senescence. Finally, this review will explore many critical observations in the way the disease process has been reassessed both by clinicians and researchers, and will highlight recent advances in this field that have provided a greater understanding of this aging-driven disease.
Collapse
Affiliation(s)
- Trajen Head
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Sylvia Daunert
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, United States
| | | |
Collapse
|
28
|
Miyazaki T, Miyazaki A. Emerging roles of calpain proteolytic systems in macrophage cholesterol handling. Cell Mol Life Sci 2017; 74:3011-3021. [PMID: 28432377 PMCID: PMC11107777 DOI: 10.1007/s00018-017-2528-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 04/12/2017] [Accepted: 04/13/2017] [Indexed: 01/21/2023]
Abstract
Calpains are Ca2+-dependent intracellular proteases that play central roles in the post-translational processing of functional proteins. In mammals, calpain proteolytic systems comprise the endogenous inhibitor calpastatin as well as 15 homologues of the catalytic subunits and two homologues of the regulatory subunits. Recent pharmacological and gene targeting studies in experimental animal models have revealed the contribution of conventional calpains, which consist of the calpain-1 and -2 isozymes, to atherosclerotic diseases. During atherogenesis, conventional calpains facilitate the CD36-dependent uptake of oxidized low-density lipoprotein (LDL), and block cholesterol efflux through ATP-binding cassette transporters in lesional macrophages, allowing the expansion of lipid-enriched atherosclerotic plaques. In addition, calpain-6, an unconventional non-proteolytic calpain, in macrophages reportedly potentiates pinocytotic uptake of native LDL, and attenuates the efferocytic clearance of apoptotic and necrotic cell corpses from the lesions. Herein, we discuss the recent progress that has been made in our understanding of how calpain contributes to atherosclerosis, in particular focusing on macrophage cholesterol handling.
Collapse
Affiliation(s)
- Takuro Miyazaki
- Department of Biochemistry, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, 142-8555, Japan.
| | - Akira Miyazaki
- Department of Biochemistry, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, 142-8555, Japan
| |
Collapse
|
29
|
Anzinger JJ, Jin X, Palmer CS, Dagur P, Barthwal MK, Kruth HS. Measurement of Aortic Cell Fluid-Phase Pinocytosis in vivo by Flow Cytometry. J Vasc Res 2017; 54:195-199. [PMID: 28618422 DOI: 10.1159/000475934] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 04/10/2017] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVE Fluid-phase pinocytosis is a receptor-independent mechanism of endocytosis that occurs in all mammalian cells and may be a mechanism for the uptake of LDL by macrophages. As there are currently no methods for the measurement of fluid-phase pinocytosis by individual aortic cells in vivo, we sought to identify a suitable method. METHODS ApoE-/- mice were retro-orbitally injected with AngioSPARK fluorescent nanoparticles specifically designed to not interact with cells. After 24 h, mice were sacrificed, and the aortas were isolated and then digested to analyze aortic cell uptake of AngioSPARK by flow cytometry. RESULTS CD11b-expressing aortic macrophages from mice injected with AngioSPARK showed high levels of fluid-phase pinocytosis compared to aortic cells not expressing CD11b (4,393.7 vs. 408.3 mean fluorescence intensity [MFI], respectively). CONCLUSION This new technique allows for the measurement of fluid-phase pinocytosis by aortic cells in vivo, making it possible to examine the cell-signaling molecules and drugs that affect this process. Published by S. Karger AG, Basel.
Collapse
Affiliation(s)
- Joshua J Anzinger
- Section of Experimental Atherosclerosis, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, USA
| | | | | | | | | | | |
Collapse
|
30
|
Brophy ML, Dong Y, Wu H, Rahman HNA, Song K, Chen H. Eating the Dead to Keep Atherosclerosis at Bay. Front Cardiovasc Med 2017; 4:2. [PMID: 28194400 PMCID: PMC5277199 DOI: 10.3389/fcvm.2017.00002] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 01/12/2017] [Indexed: 12/22/2022] Open
Abstract
Atherosclerosis is the primary cause of coronary heart disease (CHD), ischemic stroke, and peripheral arterial disease. Despite effective lipid-lowering therapies and prevention programs, atherosclerosis is still the leading cause of mortality in the United States. Moreover, the prevalence of CHD in developing countries worldwide is rapidly increasing at a rate expected to overtake those of cancer and diabetes. Prominent risk factors include the hardening of arteries and high levels of cholesterol, which lead to the initiation and progression of atherosclerosis. However, cell death and efferocytosis are critical components of both atherosclerotic plaque progression and regression, yet, few currently available therapies focus on these processes. Thus, understanding the causes of cell death within the atherosclerotic plaque, the consequences of cell death, and the mechanisms of apoptotic cell clearance may enable the development of new therapies to treat cardiovascular disease. Here, we review how endoplasmic reticulum stress and cholesterol metabolism lead to cell death and inflammation, how dying cells affect plaque progression, and how autophagy and the clearance of dead cells ameliorates the inflammatory environment of the plaque. In addition, we review current research aimed at alleviating these processes and specifically targeting therapeutics to the site of the plaque.
Collapse
Affiliation(s)
- Megan L Brophy
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Karp Family Research Laboratories, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Yunzhou Dong
- Karp Family Research Laboratories, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital , Boston, MA , USA
| | - Hao Wu
- Karp Family Research Laboratories, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital , Boston, MA , USA
| | - H N Ashiqur Rahman
- Karp Family Research Laboratories, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital , Boston, MA , USA
| | - Kai Song
- Karp Family Research Laboratories, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital , Boston, MA , USA
| | - Hong Chen
- Karp Family Research Laboratories, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital , Boston, MA , USA
| |
Collapse
|
31
|
Ruparelia N, Chai JT, Fisher EA, Choudhury RP. Inflammatory processes in cardiovascular disease: a route to targeted therapies. Nat Rev Cardiol 2016; 14:133-144. [PMID: 27905474 DOI: 10.1038/nrcardio.2016.185] [Citation(s) in RCA: 345] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Inflammatory processes are firmly established as central to the development and complications of cardiovascular diseases. Elevated levels of inflammatory markers have been shown to be predictive of future cardiovascular events. The specific targeting of these processes in experimental models has been shown to attenuate myocardial and arterial injury, reduce disease progression, and promote healing. However, the translation of these observations and the demonstration of clear efficacy in clinical practice have been disappointing. A major limitation might be that tools currently used to measure 'inflammation' are insufficiently precise and do not provide information about disease site and activity, or discriminate between functionally important activation pathways. The challenge, therefore, is to make measures of inflammation that are more meaningful, and which can guide specific targeted therapies. In this Review, we consider the roles of inflammatory processes in the related pathologies of atherosclerosis and acute myocardial infarction, by providing an evaluation of the known and emerging inflammatory pathways. We highlight contemporary techniques to characterize and quantify inflammation, and consider how they might be used to guide specific treatments. Finally, we discuss emerging opportunities in the field, including their current limitations and challenges that are the focus of ongoing study.
Collapse
Affiliation(s)
- Neil Ruparelia
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Headley Way, Oxford, OX3 9DU, UK
| | - Joshua T Chai
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Headley Way, Oxford, OX3 9DU, UK.,Acute Vascular Imaging Centre, University of Oxford, John Radcliffe Hospital, Headley Way, Oxford, OX3 9DU, UK
| | - Edward A Fisher
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Headley Way, Oxford, OX3 9DU, UK.,The Center for the Prevention of Cardiovascular Disease and the Division of Cardiology, Department of Medicine, New York University School of Medicine, New York, New York 10016, USA
| | - Robin P Choudhury
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Headley Way, Oxford, OX3 9DU, UK.,Acute Vascular Imaging Centre, University of Oxford, John Radcliffe Hospital, Headley Way, Oxford, OX3 9DU, UK
| |
Collapse
|
32
|
Miyazaki T, Tonami K, Hata S, Aiuchi T, Ohnishi K, Lei XF, Kim-Kaneyama JR, Takeya M, Itabe H, Sorimachi H, Kurihara H, Miyazaki A. Calpain-6 confers atherogenicity to macrophages by dysregulating pre-mRNA splicing. J Clin Invest 2016; 126:3417-32. [PMID: 27525442 DOI: 10.1172/jci85880] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 07/07/2016] [Indexed: 01/10/2023] Open
Abstract
Macrophages contribute to the development of atherosclerosis through pinocytotic deposition of native LDL-derived cholesterol in macrophages in the vascular wall. Inhibiting macrophage-mediated lipid deposition may have protective effects in atheroprone vasculature, and identifying mechanisms that potentiate this process may inform potential therapeutic interventions for atherosclerosis. Here, we report that dysregulation of exon junction complex-driven (EJC-driven) mRNA splicing confers hyperpinocytosis to macrophages during atherogenesis. Mechanistically, we determined that inflammatory cytokines induce an unconventional nonproteolytic calpain, calpain-6 (CAPN6), which associates with the essential EJC-loading factor CWC22 in the cytoplasm. This association disturbs the nuclear localization of CWC22, thereby suppressing the splicing of target genes, including those related to Rac1 signaling. CAPN6 deficiency in LDL receptor-deficient mice restored CWC22/EJC/Rac1 signaling, reduced pinocytotic deposition of native LDL in macrophages, and attenuated macrophage recruitment into the lesions, generating an atheroprotective phenotype in the aorta. In macrophages, the induction of CAPN6 in the atheroma interior limited macrophage movements, resulting in a decline in cell clearance from the lesions. Consistent with this finding, we observed that myeloid CAPN6 contributed to atherogenesis in a murine model of bone marrow transplantation. Furthermore, macrophages from advanced human atheromas exhibited increased CAPN6 induction and impaired CWC22 nuclear localization. Together, these results indicate that CAPN6 promotes atherogenicity in inflamed macrophages by disturbing CWC22/EJC systems.
Collapse
|
33
|
Zhong LY, Cayabyab FS, Tang CK, Zheng XL, Peng TH, Lv YC. Sortilin: A novel regulator in lipid metabolism and atherogenesis. Clin Chim Acta 2016; 460:11-7. [PMID: 27312323 DOI: 10.1016/j.cca.2016.06.013] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2016] [Revised: 06/07/2016] [Accepted: 06/11/2016] [Indexed: 11/26/2022]
Abstract
Several lines of evidence have shown that SORT1 gene within 1p13.3 locus is an important modulator of the low-density lipoprotein-cholesterol (LDL-C) level and atherosclerosis risk. Here, we summarize the effects of SORT1, which codes for sortilin, on lipid metabolism and development of atherosclerosis and explore the mechanisms underlying sortilin effects on lipid metabolism especially in hepatocytes and macrophages. Recent epidemiological evidence demonstrated that sortilin has been implicated as the causative factor and regulates lipid metabolism in vivo. Hepatic sortilin overexpression leads to both increased and decreased LDL-C levels by several different mechanisms, suggesting the complex roles of sortilin in hepatic lipid metabolism. Macrophage sortilin causes internalization of LDL and probably a reduction in cholesterol efflux, resulting in the intracellular accumulation of excessive lipids. In addition, sortilin deficiency in an atherosclerotic mouse model results in decreased aortic atherosclerotic lesion. Sortilin involves in lipid metabolism, promotes the development of atherosclerosis, and possibly becomes a potential therapeutic target for atherosclerosis treatment.
Collapse
Affiliation(s)
- Li-Yuan Zhong
- Laboratory of Clinical Anatomy, University of South China, Hengyang 421001, China
| | - Francisco S Cayabyab
- Department of Surgery, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Chao-Ke Tang
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Life Science Research Center, University of South China, Hengyang 421001, China
| | - Xi-Long Zheng
- Department of Biochemistry and Molecular Biology, The Libin Cardiovascular Institute of Alberta, The University of Calgary, Health Sciences Center, 3330 Hospital Dr NW, Calgary, Alberta T2N 4N1, Canada
| | - Tian-Hong Peng
- Laboratory of Clinical Anatomy, University of South China, Hengyang 421001, China.
| | - Yun-Cheng Lv
- Laboratory of Clinical Anatomy, University of South China, Hengyang 421001, China.
| |
Collapse
|
34
|
Choi HJ, Choi HJ, Park MJ, Lee JY, Jeong SI, Lee S, Kim KH, Joo M, Jeong HS, Kim JE, Ha KT. The inhibitory effects of Geranium thunbergii on interferon-γ- and LPS-induced inflammatory responses are mediated by Nrf2 activation. Int J Mol Med 2015; 35:1237-45. [PMID: 25761198 PMCID: PMC4380198 DOI: 10.3892/ijmm.2015.2128] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 03/02/2015] [Indexed: 01/08/2023] Open
Abstract
Geranium thunbergii Sieb. et Zucc. (GT; which belongs to the Geraniaceae family) has been used as a traditional medicine in East Asia for the treatment of inflammatory diseases, including arthritis and diarrhea. However, the underlying mechanisms of the anti-inflammatory effects of GT remain poorly understood. In the present study, we examined the mechanisms responsible for the anti-inflammatory activity of GT in macrophages. The results revealed that GT significantly inhibited the lipopolysaccharide (LPS)- and interferon-γ (IFN-γ)-induced expression of pro-inflammatory genes, such as inducible nitric oxide synthase, tumor necrosis factor-α and interleukin-1β, as shown by RT-PCR. However, the inhibitory effects of GT on LPS- and IFN-γ-induced inflammation were associated with an enhanced nuclear factor erythroid 2-related factor 2 (Nrf2) activity, but not with the suppression of nuclear factor (NF)-κB activity, as shown by western blot analysis. In addition, in bone marrow-derived macrophages (BMDM) isolated from Nrf2 knockout mice, GT did not exert any inhibitory effect on the LPS- and IFN-γ-induced inflammation. Taken together, our findings indicate that the anti-inflammatory effects of GT may be associated with the activation of Nrf2, an anti-inflammatory transcription factor.
Collapse
Affiliation(s)
- Hee-Jin Choi
- Department of Pathology, College of Korean Medicine, Dongguk University, Siksa-dong, Ilsan, Gyeonggi-do, Republic of Korea
| | - Hee-Jung Choi
- Department of Korean Medical Science, School of Korean Medicine and Korean Medicine Research Center for Healthy Aging, Pusan National University, Mulgeum-eup, Yangsan, Gyeongsangnam-do, Republic of Korea
| | - Mi-Ju Park
- Department of Korean Medical Science, School of Korean Medicine and Korean Medicine Research Center for Healthy Aging, Pusan National University, Mulgeum-eup, Yangsan, Gyeongsangnam-do, Republic of Korea
| | - Ji-Yeon Lee
- Department of Korean Medical Science, School of Korean Medicine and Korean Medicine Research Center for Healthy Aging, Pusan National University, Mulgeum-eup, Yangsan, Gyeongsangnam-do, Republic of Korea
| | - Seung-Il Jeong
- Jeonju Biomaterials Institute, Jeonju, Republic of Korea
| | - Seongoo Lee
- Department of Korean Pathology, College of Korean Medicine, Sangji University, Wonju, Gangwon, Republic of Korea
| | - Kyun Ha Kim
- Department of Korean Medical Science, School of Korean Medicine and Korean Medicine Research Center for Healthy Aging, Pusan National University, Mulgeum-eup, Yangsan, Gyeongsangnam-do, Republic of Korea
| | - Myungsoo Joo
- Department of Korean Medical Science, School of Korean Medicine and Korean Medicine Research Center for Healthy Aging, Pusan National University, Mulgeum-eup, Yangsan, Gyeongsangnam-do, Republic of Korea
| | - Han-Sol Jeong
- Department of Korean Medical Science, School of Korean Medicine and Korean Medicine Research Center for Healthy Aging, Pusan National University, Mulgeum-eup, Yangsan, Gyeongsangnam-do, Republic of Korea
| | - Jai-Eun Kim
- Department of Pathology, College of Korean Medicine, Dongguk University, Siksa-dong, Ilsan, Gyeonggi-do, Republic of Korea
| | - Ki-Tae Ha
- Department of Korean Medical Science, School of Korean Medicine and Korean Medicine Research Center for Healthy Aging, Pusan National University, Mulgeum-eup, Yangsan, Gyeongsangnam-do, Republic of Korea
| |
Collapse
|
35
|
Michael DR, Davies TS, Laubertová L, Gallagher H, Ramji DP. The phosphoinositide 3-kinase signaling pathway is involved in the control of modified low-density lipoprotein uptake by human macrophages. Lipids 2015; 50:253-60. [PMID: 25663263 PMCID: PMC4339697 DOI: 10.1007/s11745-015-3993-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2014] [Accepted: 01/19/2015] [Indexed: 02/05/2023]
Abstract
The transformation of macrophages into lipid-loaded foam cells is a critical early event in the pathogenesis of atherosclerosis. Both receptor-mediated uptake of modified LDL, mediated primarily by scavenger receptors-A (SR-A) and CD36 along with other proteins such as lipoprotein lipase (LPL), and macropinocytosis contribute to macrophage foam cell formation. The signaling pathways that are involved in the control of foam cell formation are not fully understood. In this study, we have investigated the role of phosphoinositide 3-kinase (PI3K) in relation to foam cell formation in human macrophages. The pan PI3K inhibitor LY294002 attenuated the uptake of modified LDL and macropinocytosis, as measured by Lucifer Yellow uptake, by human macrophages. In addition, the expression of SR-A, CD36 and LPL was attenuated by LY294002. The use of isoform-selective PI3K inhibitors showed that PI3K-β, -γ and -δ were all required for the expression of SR-A and CD36 whereas only PI3K-γ was necessary in the case of LPL. These studies reveal a pivotal role of PI3K in the control of macrophage foam cell formation and provide further evidence for their potential as therapeutic target against atherosclerosis.
Collapse
Affiliation(s)
- Daryn R. Michael
- Cardiff School of Biosciences, Cardiff University, Sir Martin Evans Building, Museum Avenue, Cardiff, CF10 3AX UK
| | - Thomas S. Davies
- Cardiff School of Biosciences, Cardiff University, Sir Martin Evans Building, Museum Avenue, Cardiff, CF10 3AX UK
| | - Lucia Laubertová
- Cardiff School of Biosciences, Cardiff University, Sir Martin Evans Building, Museum Avenue, Cardiff, CF10 3AX UK
- Institute of Medical Biochemistry, Jessenius Faculty of Medicine, Comenius University, Malá Hora 4, 036 01 Martin, Slovakia
| | - Hayley Gallagher
- Cardiff School of Biosciences, Cardiff University, Sir Martin Evans Building, Museum Avenue, Cardiff, CF10 3AX UK
| | - Dipak P. Ramji
- Cardiff School of Biosciences, Cardiff University, Sir Martin Evans Building, Museum Avenue, Cardiff, CF10 3AX UK
| |
Collapse
|
36
|
Patel KM, Strong A, Tohyama J, Jin X, Morales CR, Billheimer J, Millar J, Kruth H, Rader DJ. Macrophage sortilin promotes LDL uptake, foam cell formation, and atherosclerosis. Circ Res 2015; 116:789-96. [PMID: 25593281 DOI: 10.1161/circresaha.116.305811] [Citation(s) in RCA: 138] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
RATIONALE Noncoding gene variants at the SORT1 locus are strongly associated with low-density lipoprotein cholesterol (LDL-C) levels, as well as with coronary artery disease. SORT1 encodes a protein called sortilin, and hepatic sortilin modulates LDL metabolism by targeting apolipoprotein B-containing lipoproteins to the lysosome. Sortilin is also expressed in macrophages, but its role in macrophage uptake of LDL and in atherosclerosis independent of plasma LDL-C levels is unknown. OBJECTIVE To determine the effect of macrophage sortilin expression on LDL uptake, foam cell formation, and atherosclerosis. METHODS AND RESULTS We crossed Sort1(-/-) mice onto a humanized Apobec1(-/-); hAPOB transgenic background and determined that Sort1 deficiency on this background had no effect on plasma LDL-C levels but dramatically reduced atherosclerosis in the aorta and aortic root. To test whether this effect was a result of macrophage sortilin deficiency, we transplanted Sort1(-/-);LDLR(-/-) or Sort1(+/+);LDLR(-/-) bone marrow into Ldlr(-/-) mice and observed a similar reduction in atherosclerosis in mice lacking hematopoetic sortilin without an effect on plasma LDL-C levels. In an effort to determine the mechanism by which hematopoetic sortilin deficiency reduced atherosclerosis, we found no effect of sortilin deficiency on macrophage recruitment or lipopolysaccharide-induced cytokine release in vivo. In contrast, sortilin-deficient macrophages had significantly reduced uptake of native LDL ex vivo and reduced foam cell formation in vivo, whereas sortilin overexpression in macrophages resulted in increased LDL uptake and foam cell formation. CONCLUSIONS Macrophage sortilin deficiency protects against atherosclerosis by reducing macrophage uptake of LDL. Sortilin-mediated uptake of native LDL into macrophages may be an important mechanism of foam cell formation and contributor to atherosclerosis development.
Collapse
Affiliation(s)
- Kevin M Patel
- From the Department of Medicine (K.M.P., A.S., J.T., J.B., J.M., D.J.R.) and Department of Genetics (D.J.R.), Perelman School of Medicine at the University of Pennsylvania, Philadelphia; Section of Experimental Atherosclerosis, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD (X.J., H.K.); and Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec, Canada (C.R.M.)
| | - Alanna Strong
- From the Department of Medicine (K.M.P., A.S., J.T., J.B., J.M., D.J.R.) and Department of Genetics (D.J.R.), Perelman School of Medicine at the University of Pennsylvania, Philadelphia; Section of Experimental Atherosclerosis, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD (X.J., H.K.); and Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec, Canada (C.R.M.)
| | - Junichiro Tohyama
- From the Department of Medicine (K.M.P., A.S., J.T., J.B., J.M., D.J.R.) and Department of Genetics (D.J.R.), Perelman School of Medicine at the University of Pennsylvania, Philadelphia; Section of Experimental Atherosclerosis, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD (X.J., H.K.); and Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec, Canada (C.R.M.)
| | - Xueting Jin
- From the Department of Medicine (K.M.P., A.S., J.T., J.B., J.M., D.J.R.) and Department of Genetics (D.J.R.), Perelman School of Medicine at the University of Pennsylvania, Philadelphia; Section of Experimental Atherosclerosis, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD (X.J., H.K.); and Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec, Canada (C.R.M.)
| | - Carlos R Morales
- From the Department of Medicine (K.M.P., A.S., J.T., J.B., J.M., D.J.R.) and Department of Genetics (D.J.R.), Perelman School of Medicine at the University of Pennsylvania, Philadelphia; Section of Experimental Atherosclerosis, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD (X.J., H.K.); and Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec, Canada (C.R.M.)
| | - Jeffery Billheimer
- From the Department of Medicine (K.M.P., A.S., J.T., J.B., J.M., D.J.R.) and Department of Genetics (D.J.R.), Perelman School of Medicine at the University of Pennsylvania, Philadelphia; Section of Experimental Atherosclerosis, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD (X.J., H.K.); and Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec, Canada (C.R.M.)
| | - John Millar
- From the Department of Medicine (K.M.P., A.S., J.T., J.B., J.M., D.J.R.) and Department of Genetics (D.J.R.), Perelman School of Medicine at the University of Pennsylvania, Philadelphia; Section of Experimental Atherosclerosis, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD (X.J., H.K.); and Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec, Canada (C.R.M.)
| | - Howard Kruth
- From the Department of Medicine (K.M.P., A.S., J.T., J.B., J.M., D.J.R.) and Department of Genetics (D.J.R.), Perelman School of Medicine at the University of Pennsylvania, Philadelphia; Section of Experimental Atherosclerosis, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD (X.J., H.K.); and Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec, Canada (C.R.M.)
| | - Daniel J Rader
- From the Department of Medicine (K.M.P., A.S., J.T., J.B., J.M., D.J.R.) and Department of Genetics (D.J.R.), Perelman School of Medicine at the University of Pennsylvania, Philadelphia; Section of Experimental Atherosclerosis, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD (X.J., H.K.); and Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec, Canada (C.R.M.).
| |
Collapse
|