101
|
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
|
102
|
Foam Cell Induction Activates AMPK But Uncouples Its Regulation of Autophagy and Lysosomal Homeostasis. Int J Mol Sci 2020; 21:ijms21239033. [PMID: 33261140 PMCID: PMC7730064 DOI: 10.3390/ijms21239033] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 11/23/2020] [Accepted: 11/26/2020] [Indexed: 12/30/2022] Open
Abstract
The dysregulation of macrophage lipid metabolism drives atherosclerosis. AMP-activated protein kinase (AMPK) is a master regulator of cellular energetics and plays essential roles regulating macrophage lipid dynamics. Here, we investigated the consequences of atherogenic lipoprotein-induced foam cell formation on downstream immunometabolic signaling in primary mouse macrophages. A variety of atherogenic low-density lipoproteins (acetylated, oxidized, and aggregated forms) activated AMPK signaling in a manner that was in part due to CD36 and calcium-related signaling. In quiescent macrophages, basal AMPK signaling was crucial for maintaining markers of lysosomal homeostasis as well as levels of key components in the lysosomal expression and regulation network. Moreover, AMPK activation resulted in targeted upregulation of members of this network via transcription factor EB. However, in lipid-induced macrophage foam cells, neither basal AMPK signaling nor its activation affected lysosomal-associated programs. These results suggest that while the sum of AMPK signaling in cultured macrophages may be anti-atherogenic, atherosclerotic input dampens the regulatory capacity of AMPK signaling.
Collapse
|
103
|
Ma X, Liu H, Chen F. Functioning of Long Noncoding RNAs Expressed in Macrophage in the Development of Atherosclerosis. Front Pharmacol 2020; 11:567582. [PMID: 33381026 PMCID: PMC7768882 DOI: 10.3389/fphar.2020.567582] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 09/22/2020] [Indexed: 12/26/2022] Open
Abstract
Chronic inflammation is part of the pathological process during atherosclerosis (AS). Due to the abundance of monocytes/macrophages within the arterial plaque, monocytes/macrophages have become a critical cellular target in AS studies. In recent decades, a number of long noncoding RNAs (lncRNAs) have been found to exert regulatory roles on the macrophage metabolism and macrophage plasticity, consequently promoting or suppressing atherosclerotic inflammation. In this review, we provide a comprehensive overview of lncRNAs in macrophage biology, highlighting the potential role of lncRNAs in AS based on recent findings, with the aim to identify disease biomarkers and future therapeutic interventions for AS.
Collapse
Affiliation(s)
- Xirui Ma
- Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huifang Liu
- Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fengling Chen
- Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
104
|
Navas-Madroñal M, Castelblanco E, Camacho M, Consegal M, Ramirez-Morros A, Sarrias MR, Perez P, Alonso N, Galán M, Mauricio D. Role of the Scavenger Receptor CD36 in Accelerated Diabetic Atherosclerosis. Int J Mol Sci 2020; 21:ijms21197360. [PMID: 33028031 PMCID: PMC7583063 DOI: 10.3390/ijms21197360] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 09/28/2020] [Accepted: 10/01/2020] [Indexed: 01/22/2023] Open
Abstract
Diabetes mellitus entails increased atherosclerotic burden and medial arterial calcification, but the precise mechanisms are not fully elucidated. We aimed to investigate the implication of CD36 in inflammation and calcification processes orchestrated by vascular smooth muscle cells (VSMCs) under hyperglycemic and atherogenic conditions. We examined the expression of CD36, pro-inflammatory cytokines, endoplasmic reticulum (ER) stress markers, and mineralization-regulating enzymes by RT-PCR in human VSMCs, cultured in a medium containing normal (5 mM) or high glucose (22 mM) for 72 h with or without oxidized low-density lipoprotein (oxLDL) (24 h). The uptake of 1,1′-dioctadecyl-3,3,3′,3-tetramethylindocarbocyanine perchlorate-fluorescently (DiI) labeled oxLDL was quantified by flow cytometry and fluorimetry and calcification assays were performed in VSMC cultured in osteogenic medium and stained by alizarin red. We observed induction in the expression of CD36, cytokines, calcification markers, and ER stress markers under high glucose that was exacerbated by oxLDL. These results were confirmed in carotid plaques from subjects with diabetes versus non-diabetic subjects. Accordingly, the uptake of DiI-labeled oxLDL was increased after exposure to high glucose. The silencing of CD36 reduced the induction of CD36 and the expression of calcification enzymes and mineralization of VSMC. Our results indicate that CD36 signaling is partially involved in hyperglycemia and oxLDL-induced vascular calcification in diabetes.
Collapse
MESH Headings
- Aged
- Atherosclerosis/genetics
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- CD36 Antigens/genetics
- CD36 Antigens/metabolism
- Calcinosis/genetics
- Calcinosis/metabolism
- Calcinosis/pathology
- Diabetes Complications/genetics
- Diabetes Complications/metabolism
- Diabetes Complications/pathology
- Diabetes Mellitus/genetics
- Diabetes Mellitus/metabolism
- Diabetes Mellitus/pathology
- Female
- Flow Cytometry
- Glucose/adverse effects
- Humans
- Hyperglycemia/genetics
- Hyperglycemia/metabolism
- Hyperglycemia/pathology
- Inflammation/genetics
- Inflammation/metabolism
- Inflammation/pathology
- Lipoproteins, LDL/genetics
- Lipoproteins, LDL/metabolism
- Male
- Middle Aged
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Receptors, Scavenger/genetics
- Receptors, Scavenger/metabolism
Collapse
Affiliation(s)
- Miquel Navas-Madroñal
- Sant Pau Biomedical Research Institute (IIB Sant Pau), Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (M.N.-M.); (M.C.); (M.C.)
| | - Esmeralda Castelblanco
- Department of Endocrinology & Nutrition, Hospital de la Santa Creu i Sant Pau & Sant Pau Biomedical Research Institute (IIB Sant Pau), 08041 Barcelona, Spain;
- Center for Biomedical Research on Diabetes and Associated Metabolic Diseases (CIBERDEM), 08025 Barcelona, Spain;
| | - Mercedes Camacho
- Sant Pau Biomedical Research Institute (IIB Sant Pau), Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (M.N.-M.); (M.C.); (M.C.)
- Center for Biomedical Research on Cardiovascular Disease (CIBERCV), 28029 Madrid, Spain
| | - Marta Consegal
- Sant Pau Biomedical Research Institute (IIB Sant Pau), Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (M.N.-M.); (M.C.); (M.C.)
| | - Anna Ramirez-Morros
- Department of Endocrinology & Nutrition, University Hospital and Health Sciences Research Institute Germans Trias i Pujol, 08916 Badalona, Spain;
| | - Maria Rosa Sarrias
- Innate Immunity Group, Health Sciences Research Institute Germans Trias i Pujol, Center for Biomedical Research on Liver and Digestive Diseases (CIBEREHD), 28029 Madrid, Spain;
| | - Paulina Perez
- Department of Angiology & Vascular Surgery, University Hospital and Health Sciences Germans Trias i Pujol, Autonomous University of Barcelona, 08916 Badalona, Spain;
| | - Nuria Alonso
- Center for Biomedical Research on Diabetes and Associated Metabolic Diseases (CIBERDEM), 08025 Barcelona, Spain;
- Department of Endocrinology & Nutrition, University Hospital and Health Sciences Research Institute Germans Trias i Pujol, 08916 Badalona, Spain;
| | - María Galán
- Sant Pau Biomedical Research Institute (IIB Sant Pau), Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (M.N.-M.); (M.C.); (M.C.)
- Department of Endocrinology & Nutrition, Hospital de la Santa Creu i Sant Pau & Sant Pau Biomedical Research Institute (IIB Sant Pau), 08041 Barcelona, Spain;
- Correspondence: (M.G.); (D.M.); Tel.: +34-93-556-56-22 (M.G.); +34-93-556-56-61 (D.M.); Fax: +34-93-556-55-59 (M.G.); +34-93-556-56-02 (D.M.)
| | - Dídac Mauricio
- Center for Biomedical Research on Diabetes and Associated Metabolic Diseases (CIBERDEM), 08025 Barcelona, Spain;
- Center for Biomedical Research on Cardiovascular Disease (CIBERCV), 28029 Madrid, Spain
- Correspondence: (M.G.); (D.M.); Tel.: +34-93-556-56-22 (M.G.); +34-93-556-56-61 (D.M.); Fax: +34-93-556-55-59 (M.G.); +34-93-556-56-02 (D.M.)
| |
Collapse
|
105
|
Yoshii D, Nakagawa T, Komohara Y, Kawaguchi H, Yamada S, Tanimoto A. Phenotypic Changes in Macrophage Activation in a Model of Nonalcoholic Fatty Liver Disease using Microminipigs. J Atheroscler Thromb 2020; 28:844-851. [PMID: 33012740 PMCID: PMC8326174 DOI: 10.5551/jat.57703] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Aim:
Non-alcoholic fatty liver disease (NAFLD) is one of the most common chronic liver disorders associated with metabolic syndrome, and its prevalence has been on the rise. The pathogenesis of NAFLD has not yet been sufficiently elucidated due to the multifactorial nature of the disease, although the activation of macrophages/Kupffer cells is considered to be involved. We previously reported an animal model of NAFLD using Microminipigs
TM
(µMPs) fed high-fat diets containing cholesterol with or without cholic acid. The aim of this study was to investigate the phenotypic changes of macrophages that occur during the development of NAFLD.
Methods:
Immunohistochemistry of macrophages, lymphocytes, and stellate cells was performed using liver samples, and the density of positive cells was analyzed.
Results:
The number of Iba-1-positive macrophages increased with increasing cholesterol content in the diet. The numbers of CD163-positive macrophages and CD204-positive macrophages also increased with increasing cholesterol content in the diet; however, the proportion of CD204-positive macrophages among Iba-1-positive macrophages was significantly reduced by cholic acid supplementation.
Conclusion:
The results suggest that lipid accumulation induced macrophage recruitment in swine livers, and that the number of M2-like macrophages increased at the early stage of NAFLD, while the number of M1-like macrophages increased at the late stage of NAFLD, resulting in a liver condition like non-alcoholic steatohepatitis. We provide evidence of the phenotypic changes that occur in macrophages during the development of NAFLD that has never been reported before using µMPs.
Collapse
Affiliation(s)
- Daiki Yoshii
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University
| | - Takenobu Nakagawa
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University
| | - Yoshihiro Komohara
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University.,Center for Metabolic Regulation of Healthy Aging, Kumamoto University
| | - Hiroaki Kawaguchi
- Department of Hygiene and Health Promotion Medicine, Kagoshima University Graduate School of Medical and Dental Sciences
| | - Sohsuke Yamada
- Department of Pathology and Laboratory Medicine, Kanazawa Medical University
| | - Akihide Tanimoto
- Department of Pathology, Kagoshima University Graduate School of Medical and Dental Sciences
| |
Collapse
|
106
|
Belyaeva VS, Stepenko YV, Lyubimov II, Kulikov AL, Tietze AA, Kochkarova IS, Martynova OV, Pokopeyko ON, Krupen’kina LA, Nagikh AS, Pokrovskiy VM, Patrakhanov EA, Belashova AV, Lebedev PR, Gureeva AV. Non-hematopoietic erythropoietin-derived peptides for atheroprotection and treatment of cardiovascular diseases. RESEARCH RESULTS IN PHARMACOLOGY 2020. [DOI: 10.3897/rrpharmacology.6.58891] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Relevance: Cardiovascular diseases continue to be the leading cause of premature adult death.Lipid profile and atherogenesis: Dislipidaemia leads to subsequent lipid accumulation and migration of immunocompetent cells into the vessel intima. Macrophages accumulate cholesterol forming foam cells – the morphological substrate of atherosclerosis in its initial stage.Inflammation and atherogenesis: Pro-inflammatory factors provoke oxidative stress, vascular wall damage and foam cells formation.Endothelial and mitochondrial dysfunction in the development of atherosclerosis: Endothelial mitochondria are some of the organelles most sensitive to oxidative stress. Damaged mitochondria produce excess superoxide and H2O2, which are the main factors of intracellular damage, further increasing endothelial dysfunction.Short non-hematopoietic erythropoietin-based peptides as innovative atheroprotectors: Research in recent decades has shown that erythropoietin has a high cytoprotective activity, which is mainly associated with exposure to the mitochondrial link and has been confirmed in various experimental models. There is also a short-chain derivative, the 11-amino acid pyroglutamate helix B surface peptide (PHBSP), which selectively binds to the erythropoietin heterodymic receptor and reproduces its cytoprotective properties. This indicates the promising use of short-chain derivatives of erythropoietin for the treatment and prevention of atherosclerotic vascular injury. In the future, it is planned to study the PHBSP derivatives, the modification of which consists in adding RGD and PGP tripeptides with antiaggregant properties to the original 11-member peptide.
Collapse
|
107
|
Cholesterol Efflux Efficiency of Reconstituted HDL Is Affected by Nanoparticle Lipid Composition. Biomedicines 2020; 8:biomedicines8100373. [PMID: 32977626 PMCID: PMC7598155 DOI: 10.3390/biomedicines8100373] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 09/17/2020] [Accepted: 09/21/2020] [Indexed: 12/31/2022] Open
Abstract
Cardiovascular disease (CVD), the leading cause of mortality worldwide is primarily caused by atherosclerosis, which is promoted by the accumulation of low-density lipoproteins into the intima of large arteries. Multiple nanoparticles mimicking natural HDL (rHDL) have been designed to remove cholesterol excess in CVD therapy. The goal of this investigation was to assess the cholesterol efflux efficiency of rHDLs with different lipid compositions, mimicking different maturation stages of high-density lipoproteins (HDLs) occurring in vivo. Methods: the cholesterol efflux activity of soybean PC (Soy-PC), 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC), DPPC:Chol:1-palmitoyl-2-hydroxy-sn-glycero-3-phosphocholine (LysoPC) and DPPC:18:2 cholesteryl ester (CE):LysoPC rHDLs was determined in several cell models to investigate the contribution of lipid composition to the effectiveness of cholesterol removal. Results: DPPC rHDLs are the most efficient particles, inducing cholesterol efflux in all cellular models and in all conditions the effect was potentiated when the ABCA1 transporter was upregulated. Conclusions: DPPC rHDLs, which resemble nascent HDL, are the most effective particles in inducing cholesterol efflux due to the higher physical binding affinity of cholesterol to the saturated long-chain-length phospholipids and the favored cholesterol transfer from a highly positively curved bilayer, to an accepting planar bilayer such as DPPC rHDLs. The physicochemical characteristics of rHDLs should be taken into consideration to design more efficient nanoparticles to promote cholesterol efflux.
Collapse
|
108
|
Biomimetic 3D Models for Investigating the Role of Monocytes and Macrophages in Atherosclerosis. Bioengineering (Basel) 2020; 7:bioengineering7030113. [PMID: 32947976 PMCID: PMC7552756 DOI: 10.3390/bioengineering7030113] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/12/2020] [Accepted: 09/14/2020] [Indexed: 01/08/2023] Open
Abstract
Atherosclerosis, the inflammation of artery walls due to the accumulation of lipids, is the most common underlying cause for cardiovascular diseases. Monocytes and macrophages are major cells that contribute to the initiation and progression of atherosclerotic plaques. During this process, an accumulation of LDL-laden macrophages (foam cells) and an alteration in the extracellular matrix (ECM) organization leads to a local vessel stiffening. Current in vitro models are carried out onto two-dimensional tissue culture plastic and cannot replicate the relevant microenvironments. To bridge the gap between in vitro and in vivo conditions, we utilized three-dimensional (3D) collagen matrices that allowed us to mimic the ECM stiffening during atherosclerosis by increasing collagen density. First, human monocytic THP-1 cells were embedded into 3D collagen matrices reconstituted at low and high density. Cells were subsequently differentiated into uncommitted macrophages (M0) and further activated into pro- (M1) and anti-inflammatory (M2) phenotypes. In order to mimic atherosclerotic conditions, cells were cultured in the presence of oxidized LDL (oxLDL) and analyzed in terms of oxLDL uptake capability and relevant receptors along with their cytokine secretomes. Although oxLDL uptake and larger lipid size could be observed in macrophages in a matrix dependent manner, monocytes showed higher numbers of oxLDL uptake cells. By analyzing major oxLDL uptake receptors, both monocytes and macrophages expressed lectin-like oxidized low-density lipoprotein receptor-1 (LOX1), while enhanced expression of scavenger receptor CD36 could be observed only in M2. Notably, by analyzing the secretome of macrophages exposed to oxLDL, we demonstrated that the cells could, in fact, secrete adipokines and growth factors in distinct patterns. Besides, oxLDL appeared to up-regulate MHCII expression in all cells, while an up-regulation of CD68, a pan-macrophage marker, was found only in monocytes, suggesting a possible differentiation of monocytes into a pro-inflammatory macrophage. Overall, our work demonstrated that collagen density in the plaque could be one of the major factors driving atherosclerotic progression via modulation of monocyte and macrophages behaviors.
Collapse
|
109
|
Soppert J, Lehrke M, Marx N, Jankowski J, Noels H. Lipoproteins and lipids in cardiovascular disease: from mechanistic insights to therapeutic targeting. Adv Drug Deliv Rev 2020; 159:4-33. [PMID: 32730849 DOI: 10.1016/j.addr.2020.07.019] [Citation(s) in RCA: 188] [Impact Index Per Article: 37.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 07/20/2020] [Accepted: 07/22/2020] [Indexed: 12/12/2022]
Abstract
With cardiovascular disease being the leading cause of morbidity and mortality worldwide, effective and cost-efficient therapies to reduce cardiovascular risk are highly needed. Lipids and lipoprotein particles crucially contribute to atherosclerosis as underlying pathology of cardiovascular disease and influence inflammatory processes as well as function of leukocytes, vascular and cardiac cells, thereby impacting on vessels and heart. Statins form the first-line therapy with the aim to block cholesterol synthesis, but additional lipid-lowering drugs are sometimes needed to achieve low-density lipoprotein (LDL) cholesterol target values. Furthermore, beyond LDL cholesterol, also other lipid mediators contribute to cardiovascular risk. This review comprehensively discusses low- and high-density lipoprotein cholesterol, lipoprotein (a), triglycerides as well as fatty acids and derivatives in the context of cardiovascular disease, providing mechanistic insights into their role in pathological processes impacting on cardiovascular disease. Also, an overview of applied as well as emerging therapeutic strategies to reduce lipid-induced cardiovascular burden is provided.
Collapse
Affiliation(s)
- Josefin Soppert
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital Aachen, Aachen, Germany
| | - Michael Lehrke
- Medical Clinic I, University Hospital Aachen, Aachen, Germany
| | - Nikolaus Marx
- Medical Clinic I, University Hospital Aachen, Aachen, Germany
| | - Joachim Jankowski
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital Aachen, Aachen, Germany; Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre, Maastricht University, the Netherlands
| | - Heidi Noels
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital Aachen, Aachen, Germany; Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, the Netherlands.
| |
Collapse
|
110
|
Dietary Apigenin Reduces Induction of LOX-1 and NLRP3 Expression, Leukocyte Adhesion, and Acetylated Low-Density Lipoprotein Uptake in Human Endothelial Cells Exposed to Trimethylamine-N-Oxide. J Cardiovasc Pharmacol 2020; 74:558-565. [PMID: 31815868 DOI: 10.1097/fjc.0000000000000747] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
By inducing vascular inflammation, trimethylamine-N-oxide (TMAO) is associated with endothelial dysfunction, atherosclerosis, and enhanced risk of cardiovascular diseases in humans. However, the underlying mechanisms are unknown. Expression of several genes related to arteriosclerosis, inflammasomes, and endothelial dysfunction was quantified by polymerase chain reaction after exposure to TMAO. LOX-1, ICAM-1, and NLRP3 were also quantified by Western blot, whereas leukocytic adhesion was examined using fluorescently labeled U937 cells. Scavenger receptors, adhesion molecules, and other genes associated with atherosclerosis were induced in endothelial cells exposed to TMAO. On the other hand, apigenin, a flavonoid that is abundant in parsley and celery, prevents initial arteriosclerosis events in endothelial cells. Apigenin reversed the effects of TMAO on mRNA expression of LOX-1, SREC, SR-PSOX, NLRP3, ASC, TXNIP, VCAM-1, ICAM-1, and MCP-1, as well as protein expression of LOX-1, the adhesion molecule ICAM-1, and the inflammasome protein NLRP3. Apigenin also suppressed leukocyte adhesion and uptake of acetylated low-density lipoprotein. The data indicate that expression of scavenger receptors and adhesion molecules in response to TMAO, along with formation of NLRP3 inflammasomes, may drive endothelial dysfunction through uptake of acetylated low-density lipoprotein and lymphocyte adhesion. Apigenin reverses these effects, implying that it may also prevent arteriosclerosis.
Collapse
|
111
|
Increased serum levels of advanced glycation end products due to induced molting in hen layers trigger a proinflammatory response by peripheral blood leukocytes. Poult Sci 2020; 99:3452-3462. [PMID: 32616239 PMCID: PMC7597842 DOI: 10.1016/j.psj.2020.04.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 04/01/2020] [Accepted: 04/03/2020] [Indexed: 12/17/2022] Open
Abstract
Induced molting (IM), a severe detriment to animal welfare, is still used in the poultry industry in some countries to increase or rejuvenate egg production and is responsible for several physiological perturbations, possibly including reactive oxidative stress, a form of metabolic stress. Because metabolic stress has been shown to induce a proinflammatory response involved in attempts to restore homeostasis, we hypothesized that similar responses followed IM. To confirm this hypothesis, we initially confirmed the establishment of oxidative stress during IM in 75-wk-old layers by demonstrating increased production of advanced glycation end products (AGE). Concomitant with increased oxidative metabolites, cellular stress was demonstrated in peripheral blood leukocytes (PBL) by increased levels of stress gene products (the glucocorticoid receptor, sirtuin-1, and heat shock protein 70 mRNA). Increased expression of stress proteins in PBL was followed by a proinflammatory response as demonstrated by increased levels of proinflammatory gene products (IL-6 and IL-1β mRNA); increased expression of these gene products was also demonstrated in direct response to AGE in vitro, thus establishing a direct link between oxidative and cellular stress. To establish a possible pathway for inducing a proinflammatory response by PBL, we showed that AGE increased a time dependent expression of galactin-3, Toll-like receptor-4, and nuclear factor - κB, all involved in the proinflammatory activation pathway. In vivo, AGE formed complexes with increased levels of circulating acute phase proteins (lysozyme and transferrin), products of a proinflammatory immune response, thereby demonstrating an effector response to cope with the consequences of oxidative stress. Thus, the harmful consequences of IM for animal welfare are extended here by demonstrating the activation of a resource-demanding proinflammatory response.
Collapse
|
112
|
Meyer CE, Liu J, Craciun I, Wu D, Wang H, Xie M, Fussenegger M, Palivan CG. Segregated Nanocompartments Containing Therapeutic Enzymes and Imaging Compounds within DNA-Zipped Polymersome Clusters for Advanced Nanotheranostic Platform. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e1906492. [PMID: 32130785 DOI: 10.1002/smll.201906492] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 12/22/2019] [Indexed: 06/10/2023]
Abstract
Nanotheranostics is an emerging field that brings together nanoscale-engineered materials with biological systems providing a combination of therapeutic and diagnostic strategies. However, current theranostic nanoplatforms have serious limitations, mainly due to a mismatch between the physical properties of the selected nanomaterials and their functionalization ease, loading ability, or overall compatibility with bioactive molecules. Herein, a nanotheranostic system is proposed based on nanocompartment clusters composed of two different polymersomes linked together by DNA. Careful design and procedure optimization result in clusters segregating the therapeutic enzyme human Dopa decarboxylase (DDC) and fluorescent probes for the detection unit in distinct but colocalized nanocompartments. The diagnostic compartment provides a twofold function: trackability via dye loading as the imaging component and the ability to attach the cluster construct to the surface of cells. The therapeutic compartment, loaded with active DDC, triggers the cellular expression of a secreted reporter enzyme via production of dopamine and activation of dopaminergic receptors implicated in atherosclerosis. This two-compartment nanotheranostic platform is expected to provide the basis of a new treatment strategy for atherosclerosis, to expand versatility and diversify the types of utilizable active molecules, and thus by extension expand the breadth of attainable applications.
Collapse
Affiliation(s)
- Claire E Meyer
- Department of Chemistry, University of Basel, Mattenstrasse 24a, Basel, 4002, Switzerland
| | - Juan Liu
- Department of Chemistry, University of Basel, Mattenstrasse 24a, Basel, 4002, Switzerland
| | - Ioana Craciun
- Department of Chemistry, University of Basel, Mattenstrasse 24a, Basel, 4002, Switzerland
| | - Dalin Wu
- Department of Chemistry, University of Basel, Mattenstrasse 24a, Basel, 4002, Switzerland
| | - Hui Wang
- Department of Biosystems Science Engineering, ETHZ, Mattenstrasse 26, Basel, 4058, Switzerland
| | - Mingqi Xie
- Department of Biosystems Science Engineering, ETHZ, Mattenstrasse 26, Basel, 4058, Switzerland
| | - Martin Fussenegger
- Department of Biosystems Science Engineering, ETHZ, Mattenstrasse 26, Basel, 4058, Switzerland
| | - Cornelia G Palivan
- Department of Chemistry, University of Basel, Mattenstrasse 24a, Basel, 4002, Switzerland
| |
Collapse
|
113
|
Padgett LE, Araujo DJ, Hedrick CC, Olingy CE. Functional crosstalk between T cells and monocytes in cancer and atherosclerosis. J Leukoc Biol 2020; 108:297-308. [PMID: 32531833 DOI: 10.1002/jlb.1mir0420-076r] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 04/03/2020] [Accepted: 05/04/2020] [Indexed: 12/12/2022] Open
Abstract
Monocytes and monocyte-derived cells, including Mϕs and dendritic cells, exhibit a diverse array of phenotypic states that are dictated by their surrounding microenvironment. These cells direct T cell activation and function via cues that range from being immunosuppressive to immunostimulatory. Solid tumors and atherosclerotic plaques represent two pathological niches with distinct immune microenvironments. While monocytes and their progeny possess a phenotypic spectrum found within both disease contexts, most within tumors are pro-tumoral and support evasion of host immune responses by tumor cells. In contrast, monocyte-derived cells within atherosclerotic plaques are usually pro-atherogenic, pro-inflammatory, and predominantly directed against self-antigens. Consequently, cancer immunotherapies strive to enhance the immune response against tumor antigens, whereas atherosclerosis treatments seek to dampen the immune response against lipid antigens. Insights into monocyte-T cell interactions within these niches could thus inform therapeutic strategies for two immunologically distinct diseases. Here, we review monocyte diversity, interactions between monocytes and T cells within tumor and plaque microenvironments, how certain therapies have leveraged these interactions, and novel strategies to assay such associations.
Collapse
Affiliation(s)
- Lindsey E Padgett
- Division of Inflammation Biology, La Jolla Institute for Immunology, La Jolla, California, USA
| | - Daniel J Araujo
- Division of Inflammation Biology, La Jolla Institute for Immunology, La Jolla, California, USA
| | - Catherine C Hedrick
- Division of Inflammation Biology, La Jolla Institute for Immunology, La Jolla, California, USA
| | - Claire E Olingy
- Division of Inflammation Biology, La Jolla Institute for Immunology, La Jolla, California, USA
| |
Collapse
|
114
|
Roy A, Saqib U, Wary K, Baig MS. Macrophage neuronal nitric oxide synthase (NOS1) controls the inflammatory response and foam cell formation in atherosclerosis. Int Immunopharmacol 2020; 83:106382. [DOI: 10.1016/j.intimp.2020.106382] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 02/20/2020] [Accepted: 03/04/2020] [Indexed: 01/07/2023]
|
115
|
Resveratrol Treatment Is Associated with Lipid Regulation and Inhibition of Lipoprotein-Associated Phospholipase A2 (Lp-PLA2) in Rabbits Fed a High-Fat Diet. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:9641582. [PMID: 32595754 PMCID: PMC7256704 DOI: 10.1155/2020/9641582] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 03/10/2020] [Accepted: 04/18/2020] [Indexed: 12/16/2022]
Abstract
The effects of resveratrol on various conditions have been widely studied previously. This paper aimed to investigate the influence of resveratrol on atherosclerosis (AS). Twenty-four New Zealand male rabbits were randomly and equally assigned to the normal diet group (NDG), fat diet group (FDG), and fat diet with resveratrol group (80 mg/kg/d, RFG). Biochemical indicators from blood samples were analyzed at baseline and 3 months to investigate the effects of resveratrol on blood lipid, lipoprotein-associated phospholipase A2 (Lp-PLA2), liver, and renal function. The indicators including alanine aminotransferase (ALT), aspartate aminotransferase (AST), creatinine (CREA), triglycerides (TG), total cholesterol (TC), high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein cholesterol (LDL-C), and Lp-PLA2. At 3 months, arteries were stained with hematoxylin and eosin to study the influence of resveratrol on the aortic intima, smooth muscle layer, and the intima/media ratio. Comparisons of weight, ALT, AST, CREA, TG, TC, HDL-C, LDL-C, and Lp-PLA2 among the three groups showed no significant difference at baseline. However, at the end of 3 months, significant differences were observed in AST, CREA, TC, HDL-C, LDL-C, and Lp-PLA2 between the three groups (P < 0.05). In pairwise comparison, CREA, TC, LDL-C, and Lp-PLA2 had significant differences between any two groups (P < 0.05). In addition, there were significant differences in the AST and HDL-C levels between RFG and NDG groups (P < 0.05). Meanwhile, the HDL-C levels were also significantly different between the FDG and NDG groups (P < 0.01). The histologic analysis also showed that the thickness of the aortic intima and the ratio of the intima and aortic tunica media (P < 0.05) significantly decreased in RFG compared to FDG. Resveratrol may have an antiatherosclerosis effect on a rabbit model of AS.
Collapse
|
116
|
Basu A, Dvorina N, Baldwin WM, Mazumder B. High-fat diet-induced GAIT element-mediated translational silencing of mRNAs encoding inflammatory proteins in macrophage protects against atherosclerosis. FASEB J 2020; 34:6888-6906. [PMID: 32232901 DOI: 10.1096/fj.201903119r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 02/28/2020] [Accepted: 03/16/2020] [Indexed: 11/11/2022]
Abstract
Previously, we identified a mechanism of inflammation control directed by ribosomal protein L13a and "GAIT" (Gamma Activated Inhibitor of Translation) elements in target mRNAs and showed that its elimination in myeloid cell-specific L13a knockout mice (L13a KO) increased atherosclerosis susceptibility and severity. Here, we investigated the mechanistic basis of this endogenous defense against atherosclerosis. We compared molecular and cellular aspects of atherosclerosis in high-fat diet (HFD)-fed L13a KO and intact (control) mice. HFD treatment of control mice induced release of L13a from 60S ribosome, formation of RNA-binding complex, and subsequent GAIT element-mediated translational silencing. Atherosclerotic plaques from HFD-treated KO mice showed increased infiltration of M1 type inflammatory macrophages. Macrophages from KO mice showed increased phagocytic activity and elevated expression of LDL receptor and pro-inflammatory mediators. NanoString analysis of the plaques from KO mice showed upregulation of a number of mRNAs encoding inflammatory proteins. Bioinformatics analysis suggests the presence of the potential GAIT elements in the 3'UTRs of several of these mRNAs. Macrophage induces L13a/GAIT-dependent translational silencing of inflammatory genes in response to HFD as an endogenous defense against atherosclerosis in ApoE-/- model.
Collapse
Affiliation(s)
- Abhijit Basu
- Department of Biology, Geology and Environmental Sciences, Center for Gene Regulation in Health and Disease, Cleveland State University, Cleveland, OH, USA
| | - Nina Dvorina
- Department of Inflammation and Immunity, Lerner College of Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - William M Baldwin
- Department of Inflammation and Immunity, Lerner College of Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Barsanjit Mazumder
- Department of Biology, Geology and Environmental Sciences, Center for Gene Regulation in Health and Disease, Cleveland State University, Cleveland, OH, USA
| |
Collapse
|
117
|
Signaling Pathways and Key Genes Involved in Regulation of foam Cell Formation in Atherosclerosis. Cells 2020; 9:cells9030584. [PMID: 32121535 PMCID: PMC7140394 DOI: 10.3390/cells9030584] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 02/22/2020] [Accepted: 02/25/2020] [Indexed: 02/07/2023] Open
Abstract
Atherosclerosis is associated with acute cardiovascular conditions, such as ischemic heart disease, myocardial infarction, and stroke, and is the leading cause of morbidity and mortality worldwide. Our understanding of atherosclerosis and the processes triggering its initiation is constantly improving, and, during the last few decades, many pathological processes related to this disease have been investigated in detail. For example, atherosclerosis has been considered to be a chronic inflammation triggered by the injury of the arterial wall. However, recent works showed that atherogenesis is a more complex process involving not only the immune system, but also resident cells of the vessel wall, genetic factors, altered hemodynamics, and changes in lipid metabolism. In this review, we focus on foam cells that are crucial for atherosclerosis lesion formation. It has been demonstrated that the formation of foam cells is induced by modified low-density lipoprotein (LDL). The beneficial effects of the majority of therapeutic strategies with generalized action, such as the use of anti-inflammatory drugs or antioxidants, were not confirmed by clinical studies. However, the experimental therapies targeting certain stages of atherosclerosis, among which are lipid accumulation, were shown to be more effective. This emphasizes the relevance of future detailed investigation of atherogenesis and the importance of new therapies development.
Collapse
|
118
|
Feriani A, Hachani R, Tir M, Ghazouani L, Mufti A, Borgi MA, Allagui MS. Bifenthrin exerts proatherogenic effects via arterial accumulation of native and oxidized LDL in rats: the beneficial role of vitamin E and selenium. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2020; 27:5651-5660. [PMID: 30465240 DOI: 10.1007/s11356-018-3771-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 11/15/2018] [Indexed: 06/09/2023]
Abstract
The purpose of this study was to investigate, for the first time, the effects of Bifenthrin (Bif) chronic exposure on plasmatic and aortic lipid parameters disturbance and their pro-atherogenic possibility in Wistar rats. The ameliorative role of vitamin E (Vit E) and selenium (Se) were also targeted. Thus, rats were treated by gastric gavage with combination of Vit E (100 mg/kg/bw) and Se (0.25 mg/kg/bw) in alone and co-treated groups for 90 days. Apart from control and Vit E-Se groups, all the groups were subjected to Bif (3 mg/kg, via gavage) toxicity. Results showed that Bif increased markedly plasmatic and aortic total cholesterol, LDL-cholesterol, native LDL-apoB-100, and oxidized-LDL, compared to the control. Moreover, Bif treatment significantly increased the plasmatic levels of the pro-inflammatory cytokines TNF-α, IL-2, and IL-6. In addition, the densitometric quantification of protein bands showed that the amount of hepatic native LDL-receptor protein decreased significantly in the intoxicated rats compared to the control group. The expression of arterial LDL receptors (LDLRs) and scavenger receptors (CD36) was amplified owing to Bif toxicity. This harmful effect was confirmed by histological study using Oil-Red-O staining. Owing to their antioxidant capacities, Vit E and Se have maintained all the changes in plasma and aorta lipids and prevented the pro-atherogenic effect observed in Bif-treated animals.
Collapse
Affiliation(s)
- Anouar Feriani
- Unité de Biochimie Macromoléculaire et Génétique, Faculté des Sciences de Gafsa, cité Zarroug, Université de Gafsa, 2112, Gafsa, Tunisia.
| | - Rafik Hachani
- Unité de Physiologie Intégrée, Laboratoire de Pathologies Vasculaires, Faculté des Sciences de Bizerte, Université de Carthage, 7021, Jarzouna, Tunisia
- Laboratoire d'Etude de la Microcirculation (EA 3509), Faculté de Médecine Lariboisière-St. Louis, Université Paris VII, Paris, France
| | - Meriam Tir
- Unité de Physiologie et Environnement Aquatique, Faculté des Sciences de Tunis, Université Tunis EL Manar, 2092, Tunis, Tunisia
| | - Lakhdar Ghazouani
- Unité de Biochimie Macromoléculaire et Génétique, Faculté des Sciences de Gafsa, cité Zarroug, Université de Gafsa, 2112, Gafsa, Tunisia
| | - Afoua Mufti
- Unité de Biochimie Macromoléculaire et Génétique, Faculté des Sciences de Gafsa, cité Zarroug, Université de Gafsa, 2112, Gafsa, Tunisia
| | - Mohamed Ali Borgi
- Unité de Biochimie Macromoléculaire et Génétique, Faculté des Sciences de Gafsa, cité Zarroug, Université de Gafsa, 2112, Gafsa, Tunisia
| | - Mohamed Salah Allagui
- Laboratoire d'Ecophysiologie Animale, Faculté des Sciences de Sfax, 3018, Sfax, Tunisia
| |
Collapse
|
119
|
Lamberti G, De Smet CH, Angelova M, Kremser L, Taub N, Herrmann C, Hess MW, Rainer J, Tancevski I, Schweigreiter R, Kofler R, Schmiedinger T, Vietor I, Trajanoski Z, Ejsing CS, Lindner HH, Huber LA, Stasyk T. LAMTOR/Ragulator regulates lipid metabolism in macrophages and foam cell differentiation. FEBS Lett 2020; 594:31-42. [PMID: 31423582 PMCID: PMC7003824 DOI: 10.1002/1873-3468.13579] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 07/30/2019] [Accepted: 07/31/2019] [Indexed: 11/08/2022]
Abstract
Late endosomal/lysosomal adaptor and MAPK and mTOR activator (LAMTOR/Ragulator) is a scaffold protein complex that anchors and regulates multiprotein signaling units on late endosomes/lysosomes. To identify LAMTOR-modulated endolysosomal proteins, primary macrophages were derived from bone marrow of conditional knockout mice carrying a specific deletion of LAMTOR2 in the monocyte/macrophage cell lineage. Affymetrix-based transcriptomic analysis and quantitative iTRAQ-based organelle proteomic analysis of endosomes derived from macrophages were performed. Further analyses showed that LAMTOR could be a novel regulator of foam cell differentiation. The lipid droplet formation phenotype observed in macrophages was additionally confirmed in MEFs, where lipidomic analysis identified cholesterol esters as specifically downregulated in LAMTOR2 knockout cells. The data obtained indicate a function of LAMTOR2 in lipid metabolism.
Collapse
Affiliation(s)
- Giorgia Lamberti
- Division of Cell BiologyBiocenterMedical University of InnsbruckAustria
| | - Cedric H. De Smet
- Division of Cell BiologyBiocenterMedical University of InnsbruckAustria
| | - Mihaela Angelova
- Division of BioinformaticsBiocenterMedical University of InnsbruckAustria
| | - Leopold Kremser
- Division of Clinical BiochemistryBiocenterMedical University of InnsbruckAustria
| | - Nicole Taub
- Division of Cell BiologyBiocenterMedical University of InnsbruckAustria
| | - Caroline Herrmann
- Division of Cell BiologyBiocenterMedical University of InnsbruckAustria
| | - Michael W. Hess
- Division of Histology and EmbryologyMedical University of InnsbruckAustria
| | - Johannes Rainer
- Division of Molecular PathophysiologyBiocenterMedical University of InnsbruckAustria
| | - Ivan Tancevski
- Department of Internal MedicineMedical University of InnsbruckAustria
| | | | - Reinhard Kofler
- Division of Molecular PathophysiologyBiocenterMedical University of InnsbruckAustria
| | - Thomas Schmiedinger
- Department of Therapeutic Radiology and OncologyMedical University of InnsbruckAustria
| | - Ilja Vietor
- Division of Cell BiologyBiocenterMedical University of InnsbruckAustria
| | - Zlatko Trajanoski
- Division of BioinformaticsBiocenterMedical University of InnsbruckAustria
| | - Christer S. Ejsing
- Department of Biochemistry and Molecular BiologyVillum Center for Bioanalytical SciencesUniversity of Southern DenmarkOdenseDenmark
| | - Herbert H. Lindner
- Division of Clinical BiochemistryBiocenterMedical University of InnsbruckAustria
| | - Lukas A. Huber
- Division of Cell BiologyBiocenterMedical University of InnsbruckAustria
- Austrian Drug Screening Institute, ADSIInnsbruckAustria
| | - Taras Stasyk
- Division of Cell BiologyBiocenterMedical University of InnsbruckAustria
| |
Collapse
|
120
|
Kobos L, Shannahan J. Biocorona‐induced modifications in engineered nanomaterial–cellular interactions impacting biomedical applications. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2019; 12:e1608. [PMID: 31788989 DOI: 10.1002/wnan.1608] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 09/18/2019] [Accepted: 09/29/2019] [Indexed: 02/06/2023]
Affiliation(s)
- Lisa Kobos
- School of Health Sciences College of Human and Health Sciences, Purdue University West Lafayette Indiana
| | - Jonathan Shannahan
- School of Health Sciences College of Human and Health Sciences, Purdue University West Lafayette Indiana
| |
Collapse
|
121
|
Ding X, Zheng L, Yang B, Wang X, Ying Y. Luteolin Attenuates Atherosclerosis Via Modulating Signal Transducer And Activator Of Transcription 3-Mediated Inflammatory Response. DRUG DESIGN DEVELOPMENT AND THERAPY 2019; 13:3899-3911. [PMID: 31819365 PMCID: PMC6874161 DOI: 10.2147/dddt.s207185] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 09/26/2019] [Indexed: 12/14/2022]
Abstract
Background Inflammatory factors play a crucial role throughout the development and progression of atherosclerosis, which has been considered as a chronic vascular inflammatory disease. Luteolin, a natural flavonoid which exists in many natural medicinal materials, has anti-inflammatory, anti-fibrotic and other pharmacological effects. Recently, the protective effects of luteolin on the cardiovascular disease have been reported. However, there is a paucity of studies on anti-atherosclerosis. Therefore, the anti-atherosclerosis potential of luteolin remains to be elucidated. Method ApoE-/- mice were fed with a high-fat diet to induce atherosclerosis in an animal model, where they were treated with oral administration of luteolin for 12 weeks. Primary mouse peritoneal macrophages challenged with oxidized low-density lipoprotein (oxLDL) were used for in vitro mechanistic study. The effectiveness of luteolin in the ApoE-/- mouse model of atherosclerosis was estimated in the aortic sinus and enface, and the underlying mechanisms were explored by molecular modeling study and siRNA-induced gene silencing. Results Our results showed that luteolin remarkably attenuated atherosclerosis in high-fat diet-induced ApoE-/- mouse via alleviating inflammation. We further found that luteolin decreased oxLDL-induced inflammation by inhibiting signal transducer and activator of transcription 3 (STAT3) in vitro, respectively. Further molecular modeling analysis indicated that luteolin interacted with STAT3 primarily through hydrogen bond interaction. Conclusion Luteolin could be a promising candidate molecule for atherosclerosis, and STAT3 may be a potential therapeutic target that could prevent the development of atherosclerosis.
Collapse
Affiliation(s)
- Xiaoji Ding
- Department of Pharmacy, Tongde Hospital of Zhejiang Province, Hangzhou 310012, Zhejiang, People's Republic of China
| | - Lulu Zheng
- Department of Pharmacy, Tongde Hospital of Zhejiang Province, Hangzhou 310012, Zhejiang, People's Republic of China
| | - Bo Yang
- Department of Pharmacy, Tongde Hospital of Zhejiang Province, Hangzhou 310012, Zhejiang, People's Republic of China
| | - Xiaodong Wang
- Department of Vascular Surgery, Tongde Hospital of Zhejiang Province, Hangzhou 310012, Zhejiang, People's Republic of China
| | - Yin Ying
- Department of Pharmacy, Tongde Hospital of Zhejiang Province, Hangzhou 310012, Zhejiang, People's Republic of China
| |
Collapse
|
122
|
Enos N, Takenaka H, Scott S, Salfity HVN, Kirk M, Egar MW, Sarria DA, Slayback-Barry D, Belecky-Adams T, Chernoff EAG. Meningeal Foam Cells and Ependymal Cells in Axolotl Spinal Cord Regeneration. Front Immunol 2019; 10:2558. [PMID: 31736973 PMCID: PMC6838144 DOI: 10.3389/fimmu.2019.02558] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 10/15/2019] [Indexed: 12/01/2022] Open
Abstract
A previously unreported population of foam cells (foamy macrophages) accumulates in the invasive fibrotic meninges during gap regeneration of transected adult Axolotl spinal cord (salamander Ambystoma mexicanum) and may act beneficially. Multinucleated giant cells (MNGCs) also occurred in the fibrotic meninges. Actin-label localization and transmission electron microscopy showed characteristic foam cell and MNGC podosome and ruffled border-containing sealing ring structures involved in substratum attachment, with characteristic intermediate filament accumulations surrounding nuclei. These cells co-localized with regenerating cord ependymal cell (ependymoglial) outgrowth. Phase contrast-bright droplets labeled with Oil Red O, DiI, and DyRect polar lipid live cell label showed accumulated foamy macrophages to be heavily lipid-laden, while reactive ependymoglia contained smaller lipid droplets. Both cell types contained both neutral and polar lipids in lipid droplets. Foamy macrophages and ependymoglia expressed the lipid scavenger receptor CD36 (fatty acid translocase) and the co-transporter toll-like receptor-4 (TLR4). Competitive inhibitor treatment using the modified fatty acid Sulfo-N-succinimidyl Oleate verified the role of the lipid scavenger receptor CD36 in lipid uptake studies in vitro. Fluoromyelin staining showed both cell types took up myelin fragments in situ during the regeneration process. Foam cells took up DiI-Ox-LDL and DiI-myelin fragments in vitro while ependymoglia took up only DiI-myelin in vitro. Both cell types expressed the cysteine proteinase cathepsin K, with foam cells sequestering cathepsin K within the sealing ring adjacent to the culture substratum. The two cell types act as sinks for Ox-LDL and myelin fragments within the lesion site, with foamy macrophages showing more Ox-LDL uptake activity. Cathepsin K activity and cellular localization suggested that foamy macrophages digest ECM within reactive meninges, while ependymal cells act from within the spinal cord tissue during outgrowth into the lesion site, acting in complementary fashion. Small MNGCs also expressed lipid transporters and showed cathepsin K activity. Comparison of 3H-glucosamine uptake in ependymal cells and foam cells showed that only ependymal cells produce glycosaminoglycan and proteoglycan-containing ECM, while the cathepsin studies showed both cell types remove ECM. Interaction of foam cells and ependymoglia in vitro supported the dispersion of ependymal outgrowth associated with tissue reconstruction in Axolotl spinal cord regeneration.
Collapse
Affiliation(s)
- Nathaniel Enos
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, United States
| | - Hidehito Takenaka
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, United States
| | - Sarah Scott
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, United States
| | - Hai V N Salfity
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, United States
| | - Maia Kirk
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, United States
| | - Margaret W Egar
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, United States
| | - Deborah A Sarria
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, United States
| | - Denise Slayback-Barry
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, United States
| | - Teri Belecky-Adams
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, United States
| | - Ellen A G Chernoff
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, United States
| |
Collapse
|
123
|
Herrero-Fernandez B, Gomez-Bris R, Somovilla-Crespo B, Gonzalez-Granado JM. Immunobiology of Atherosclerosis: A Complex Net of Interactions. Int J Mol Sci 2019; 20:E5293. [PMID: 31653058 PMCID: PMC6862594 DOI: 10.3390/ijms20215293] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 10/21/2019] [Accepted: 10/22/2019] [Indexed: 02/07/2023] Open
Abstract
Cardiovascular disease is the leading cause of mortality worldwide, and atherosclerosis the principal factor underlying cardiovascular events. Atherosclerosis is a chronic inflammatory disease characterized by endothelial dysfunction, intimal lipid deposition, smooth muscle cell proliferation, cell apoptosis and necrosis, and local and systemic inflammation, involving key contributions to from innate and adaptive immunity. The balance between proatherogenic inflammatory and atheroprotective anti-inflammatory responses is modulated by a complex network of interactions among vascular components and immune cells, including monocytes, macrophages, dendritic cells, and T, B, and foam cells; these interactions modulate the further progression and stability of the atherosclerotic lesion. In this review, we take a global perspective on existing knowledge about the pathogenesis of immune responses in the atherosclerotic microenvironment and the interplay between the major innate and adaptive immune factors in atherosclerosis. Studies such as this are the basis for the development of new therapies against atherosclerosis.
Collapse
Affiliation(s)
- Beatriz Herrero-Fernandez
- LamImSys Lab. Instituto de Investigación Hospital 12 de Octubre (imas12), 28041 Madrid, Spain.
- Departamento de Fisiología. Facultad de Medicina. Universidad Autónoma de Madrid (UAM), 28029 Madrid, Spain.
| | - Raquel Gomez-Bris
- LamImSys Lab. Instituto de Investigación Hospital 12 de Octubre (imas12), 28041 Madrid, Spain.
| | | | - Jose Maria Gonzalez-Granado
- LamImSys Lab. Instituto de Investigación Hospital 12 de Octubre (imas12), 28041 Madrid, Spain.
- Departamento de Fisiología. Facultad de Medicina. Universidad Autónoma de Madrid (UAM), 28029 Madrid, Spain.
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), 28029 Madrid, Spain.
- CIBER de Enfermedades Cardiovasculares, 28029 Madrid, Spain.
| |
Collapse
|
124
|
Xiao Y, Chen B, Yang K, Wang Q, Liu P, Gu Y, Zhong Q, Liu Z, He Y, Liu Q. Down-regulation of MARCO associates with tumor progression in hepatocellular carcinoma. Exp Cell Res 2019; 383:111542. [PMID: 31381879 DOI: 10.1016/j.yexcr.2019.111542] [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: 03/22/2019] [Revised: 07/26/2019] [Accepted: 07/28/2019] [Indexed: 12/25/2022]
Abstract
Hepatocellular carcinoma(HCC) is a malignant tumor with high mortality due to lack of early diagnostic methods and effective treatments, and the molecular mechanisms are intricate and remain unclear. In the present study, the role of macrophage receptor with collagenous structure (MARCO) in tumor advancement of HCC was investigated. We examined expression level of MARCO in HCC samples, corresponding adjacent nontumor tissues and six hepatoma cell lines by polymerase chain reaction and immunohistochemistry (IHC). Clinical information of HCC patients was also analyzed. The role of MARCO involved in HCC progression via multiple functional experiments in vitro and in vivo was investigated. Bioinformatics analysis was conducted to further explore biological functions of MARCO. We found MARCO was suggestively down-regulated in HCC and associated with favorable prognosis, and MARCO upregulation oppressed tumor cell migration and invasion. Besides, overexpression of MARCO not only promoted apoptosis of hepatoma cells but also suppressed proliferation in vivo and in vitro. Furthermore, gene set enrichment analysis (GSEA) analysis suggested that MARCO may be related to the P53 signaling pathway, and this prediction was confirmed in this study as well. In sum, our study indicated that MARCO was involved in HCC progression and it can be defined as a novel probable biomarker as well as treatment target for HCC.
Collapse
Affiliation(s)
- Yusha Xiao
- Department of General Surgery, Research Center of Digestive Diseases, Zhongnan Hospital of Wuhan University, Hubei, China
| | - Baiyang Chen
- Department of General Surgery, Research Center of Digestive Diseases, Zhongnan Hospital of Wuhan University, Hubei, China
| | - Kang Yang
- Department of Urology, Renmin Hospital of Wuhan University, Hubei, China
| | - Quanxiong Wang
- Department of General Surgery, Research Center of Digestive Diseases, Zhongnan Hospital of Wuhan University, Hubei, China
| | - Pengpeng Liu
- Department of General Surgery, Research Center of Digestive Diseases, Zhongnan Hospital of Wuhan University, Hubei, China
| | - Yang Gu
- Department of General Surgery, Research Center of Digestive Diseases, Zhongnan Hospital of Wuhan University, Hubei, China
| | - Qiu Zhong
- Department of General Surgery, Research Center of Digestive Diseases, Zhongnan Hospital of Wuhan University, Hubei, China
| | - Zhisu Liu
- Department of General Surgery, Research Center of Digestive Diseases, Zhongnan Hospital of Wuhan University, Hubei, China
| | - Yueming He
- Department of General Surgery, Research Center of Digestive Diseases, Zhongnan Hospital of Wuhan University, Hubei, China.
| | - Quanyan Liu
- Department of General Surgery, Research Center of Digestive Diseases, Zhongnan Hospital of Wuhan University, Hubei, China.
| |
Collapse
|
125
|
Nagarajan UM, Sikes JD, Burris RL, Jha R, Popovic B, Fraungruber P, Hennings L, Haggerty CL, Nagarajan S. Genital Chlamydia infection in hyperlipidemic mouse models exacerbates atherosclerosis. Atherosclerosis 2019; 290:103-110. [PMID: 31604170 DOI: 10.1016/j.atherosclerosis.2019.09.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 09/11/2019] [Accepted: 09/26/2019] [Indexed: 01/21/2023]
Abstract
BACKGROUND AND AIMS Atherosclerosis is a chronic inflammatory disease, and recent studies have shown that infection at remote sites can contribute to the progression of atherosclerosis in hyperlipidemic mouse models. In this report, we tested the hypothesis that genital Chlamydia infection could accelerate the onset and progression of atherosclerosis. METHODS Apolipoprotein E (Apoe-/-) and LDL receptor knockout (Ldlr-/-) mice on a high-fat diet were infected intra-vaginally with Chlamydia muridarum. Atherosclerotic lesions on the aortic sinuses and in the descending aorta were assessed at 8-weeks post-infection. Systemic, macrophage, and vascular site inflammatory responses were assessed and quantified. RESULTS Compared to the uninfected groups, infected Apoe-/- and Ldlr-/- mice developed significantly more atherosclerotic lesions in the aortic sinus and in the descending aorta. Increased lesions were associated with higher circulating levels of serum amyloid A-1, IL-1β, TNF-α, and increased VCAM-1 expression in the aortic sinus, suggesting an association with inflammatory responses observed during C. muridarum infection. Genital infection courses were similar in Apoe-/-, Ldlr-/-, and wild type mice. Further, Apoe-/- mice developed severe uterine pathology with increased dilatations. Apoe-deficiency also augmented cytokine/chemokine response in C. muridarum infected macrophages, suggesting that the difference in macrophage response could have contributed to the genital pathology in Apoe-/- mice. CONCLUSIONS Overall, these studies demonstrate that genital Chlamydia infection exacerbates atherosclerotic lesions in hyperlipidemic mouse and suggest a novel role for Apoe in full recovery of uterine anatomy after chlamydial infection.
Collapse
Affiliation(s)
- Uma M Nagarajan
- Department of Pediatrics and Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Department of Pediatrics, University of Arkansas for Medical Sciences, Arkansas Children's Hospital Research Institute, Little Rock, AR, USA
| | - James D Sikes
- Department of Pediatrics, University of Arkansas for Medical Sciences, Arkansas Children's Hospital Research Institute, Little Rock, AR, USA
| | - Ramona L Burris
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Arkansas Children's Hospital Research Institute, Little Rock, AR, USA
| | - Rajneesh Jha
- Department of Pediatrics, University of Arkansas for Medical Sciences, Arkansas Children's Hospital Research Institute, Little Rock, AR, USA
| | - Branimir Popovic
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Pamelia Fraungruber
- Department of Pathology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Leah Hennings
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Catherine L Haggerty
- Department of Epidemiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA, USA
| | - Shanmugam Nagarajan
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Arkansas Children's Hospital Research Institute, Little Rock, AR, USA; Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA; Department of Pathology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
126
|
Sun Y, Gao W, Liu Z, Yang H, Cao W, Tong L, Tang B. Luminescence-Resonance-Energy-Transfer-Based Luminescence Nanoprobe for In Situ Imaging of CD36 Activation and CD36–oxLDL Binding in Atherogenesis. Anal Chem 2019; 91:9770-9776. [DOI: 10.1021/acs.analchem.9b01398] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Yuhui Sun
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan 250014, PR China
| | - Wen Gao
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan 250014, PR China
| | - Zhenhua Liu
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan 250014, PR China
| | - Huazhen Yang
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan 250014, PR China
| | - Wenhua Cao
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan 250014, PR China
| | - Lili Tong
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan 250014, PR China
| | - Bo Tang
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan 250014, PR China
| |
Collapse
|
127
|
Calderon Moreno R, Navas-Acien A, Escolar E, Nathan DM, Newman J, Schmedtje JF, Diaz D, Lamas GA, Fonseca V. Potential Role of Metal Chelation to Prevent the Cardiovascular Complications of Diabetes. J Clin Endocrinol Metab 2019; 104:2931-2941. [PMID: 30869793 PMCID: PMC9136707 DOI: 10.1210/jc.2018-01484] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 03/07/2019] [Indexed: 02/12/2023]
Abstract
CONTEXT For decades, there has been epidemiologic evidence linking chronic toxic metal exposure with cardiovascular disease, suggesting a therapeutic role for metal chelation. Given the lack of compelling scientific evidence, however, the indications for metal chelation were never clearly defined. To determine the safety and efficacy of chelation therapy, the National Institutes of Health funded the Trial to Assess Chelation Therapy (TACT). TACT was the first double-blind, randomized, controlled trial to demonstrate an improvement in cardiovascular outcomes with edetate disodium therapy in patients with prior myocardial infarction. The therapeutic benefit was striking among the prespecified subgroup of patients with diabetes. DESIGN We review the published literature focusing on the atherogenic nature of diabetes, as well as available evidence from clinical trials, complete and in progress, of metal chelation with edetate disodium therapy in patients with diabetes. RESULTS The TACT results support the concept that ubiquitous toxic metals such as lead and cadmium may be modifiable risk factors for cardiovascular disease, particularly in patients with diabetes. CONCLUSIONS The purpose of this review is to discuss the potential mechanisms unifying the pathogenesis of atherogenic factors in diabetes with toxic metal exposure, and the potential role of metal chelation.
Collapse
Affiliation(s)
| | - Ana Navas-Acien
- Columbia University Mailman School of Public Health, New York, New York
| | - Esteban Escolar
- Department of Medicine, Columbia University Division of Cardiology at Mount Sinai Medical Center, Miami Beach, Florida
| | - David M Nathan
- Diabetes Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Jonathan Newman
- Department of Medicine, New York University School of Medicine, New York, New York
| | | | - Denisse Diaz
- Department of Medicine, Columbia University Division of Cardiology at Mount Sinai Medical Center, Miami Beach, Florida
- Correspondence and Reprint Requests: Denisse Diaz, MD, Mount Sinai Medical Center, 4300 Alton Road, Miami Beach, Florida 33140. E-mail:
| | - Gervasio A Lamas
- Department of Medicine, Columbia University Division of Cardiology at Mount Sinai Medical Center, Miami Beach, Florida
| | - Vivian Fonseca
- Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana
| |
Collapse
|
128
|
Zhang J, Nie S, Zu Y, Abbasi M, Cao J, Li C, Wu D, Labib S, Brackee G, Shen CL, Wang S. Anti-atherogenic effects of CD36-targeted epigallocatechin gallate-loaded nanoparticles. J Control Release 2019; 303:263-273. [PMID: 30999008 PMCID: PMC6579691 DOI: 10.1016/j.jconrel.2019.04.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Revised: 04/03/2019] [Accepted: 04/12/2019] [Indexed: 12/28/2022]
Abstract
Intimal macrophages play a critical role in atherosclerotic lesion initiation and progression by taking up oxidized low-density lipoprotein (oxLDL) and promoting inflammatory process. 1-(Palmitoyl)-2-(5-keto-6-octene-dioyl) phosphatidylcholine (KOdiA-PC), a major type of oxidized phosphatidylcholines (PC) found on oxLDL, has a high binding affinity to the macrophage scavenger receptor CD36 and participates in CD36-mediated recognition and uptake of oxLDL by intimal macrophages. We successfully synthesized epigallocatechin gallate (EGCG)-loaded nanoparticles (Enano), which were composed of EGCG, PC, (+) alpha-tocopherol acetate, and surfactant. We also incorporated KOdiA-PC on the surface of Enano to make ligand-coated Enano (L-Enano) to target intimal macrophages. The objectives of this study were to determine the anti-atherogenic effects of Enano and L-Enano in LDL receptor null (LDLr-/-) mice. Our in vitro data demonstrated that L-Enano had a higher binding affinity to mouse peritoneal macrophages than Enano. This high binding affinity was diminished by CD36 antibodies or knockdown of the CD36 receptor in mouse peritoneal macrophages, confirming the specific binding of L-Enano to the macrophage CD36 receptor. LDLr-/- mice were randomly divided to six groups and received weekly tail vein injection with PBS, EGCG, void nanoparticles (Vnano), Enano, ligand-coated Vnano (L-Vnano), or L-Enano once per week for 22 weeks. The dose of EGCG was 25 mg per kg body weight. L-Enano at 20 μg/mL significantly decreased production of monocyte chemoattractant protein-1, tumor necrosis factor alpha, and interleukin-6 from mouse macrophages, while having no effect on their plasma levels compared to the PBS control. There were no significant differences in blood lipid profiles among six treatment groups. Mice treated with L-Enano also had significantly smaller lesion surface areas on aortic arches compared to the PBS control. Liver EGCG content was decreased by treatments in the order of EGCG>Enano>L-Enano. Native EGCG had inhibitory effects on liver and body fat accumulation. This molecular target approach signals an important step towards inhibiting atherosclerosis development via targeted delivery of bioactive compounds to intimal macrophages.
Collapse
Affiliation(s)
- Jia Zhang
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX 79409, USA
| | - Shufang Nie
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX 79409, USA
| | - Yujiao Zu
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX 79409, USA
| | - Mehrnaz Abbasi
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX 79409, USA
| | - Jun Cao
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX 79409, USA; College of Food Science and Technology, Hainan University, Haikou 570228, China
| | - Chuan Li
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX 79409, USA; College of Food Science and Technology, Hainan University, Haikou 570228, China
| | - Dayong Wu
- Nutrition Immunology Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA 02111, USA
| | - Safaa Labib
- Department of Pathology, Texas Tech University Health Sciences Center, Lubbock, TX 70430, USA
| | - Gordon Brackee
- Laboratory Animal Resources Center, Texas Tech University Health Sciences Center, Lubbock, TX 79416, USA; Comparative Biology Resources Center, University of Rhode Island, Kingston, RI 02881, USA
| | - Chwan-Li Shen
- Department of Pathology, Texas Tech University Health Sciences Center, Lubbock, TX 70430, USA
| | - Shu Wang
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX 79409, USA.
| |
Collapse
|
129
|
Milic J, Tian Y, Bernhagen J. Role of the COP9 Signalosome (CSN) in Cardiovascular Diseases. Biomolecules 2019; 9:biom9060217. [PMID: 31195722 PMCID: PMC6628250 DOI: 10.3390/biom9060217] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 06/02/2019] [Accepted: 06/03/2019] [Indexed: 12/19/2022] Open
Abstract
The constitutive photomorphogenesis 9 (COP9) signalosome (CSN) is an evolutionarily conserved multi-protein complex, consisting of eight subunits termed CSN1-CSN8. The main biochemical function of the CSN is the control of protein degradation via the ubiquitin-proteasome-system through regulation of cullin-RING E3-ligase (CRL) activity by deNEDDylation of cullins, but the CSN also serves as a docking platform for signaling proteins. The catalytic deNEDDylase (isopeptidase) activity of the complex is executed by CSN5, but only efficiently occurs in the three-dimensional architectural context of the complex. Due to its positioning in a central cellular pathway connected to cell responses such as cell-cycle, proliferation, and signaling, the CSN has been implicated in several human diseases, with most evidence available for a role in cancer. However, emerging evidence also suggests that the CSN is involved in inflammation and cardiovascular diseases. This is both due to its role in controlling CRLs, regulating components of key inflammatory pathways such as nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), and complex-independent interactions of subunits such as CSN5 with inflammatory proteins. In this case, we summarize and discuss studies suggesting that the CSN may have a key role in cardiovascular diseases such as atherosclerosis and heart failure. We discuss the implicated molecular mechanisms ranging from inflammatory NF-κB signaling to proteotoxicity and necrosis, covering disease-relevant cell types such as myeloid and endothelial cells or cardiomyocytes. While the CSN is considered to be disease-exacerbating in most cancer entities, the cardiovascular studies suggest potent protective activities in the vasculature and heart. The underlying mechanisms and potential therapeutic avenues will be critically discussed.
Collapse
Affiliation(s)
- Jelena Milic
- Chair of Vascular Biology, Institute for Stroke and Dementia Research (ISD), Klinikum der Universität München (KUM), Ludwig-Maximilians-University (LMU), 81377 Munich, Germany.
| | - Yuan Tian
- Chair of Vascular Biology, Institute for Stroke and Dementia Research (ISD), Klinikum der Universität München (KUM), Ludwig-Maximilians-University (LMU), 81377 Munich, Germany.
| | - Jürgen Bernhagen
- Chair of Vascular Biology, Institute for Stroke and Dementia Research (ISD), Klinikum der Universität München (KUM), Ludwig-Maximilians-University (LMU), 81377 Munich, Germany.
- Munich Heart Alliance, 80802 Munich, Germany.
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany.
| |
Collapse
|
130
|
DOPG small unilamellar vesicles function as nano-carriers targeting the clustered lectin-like oxidized LDL receptor (LOX-1) on the cell surface. J Drug Deliv Sci Technol 2019. [DOI: 10.1016/j.jddst.2019.03.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
131
|
LI TT, WANG ZB, LI Y, CAO F, YANG BY, KUANG HX. The mechanisms of traditional Chinese medicine underlying the prevention and treatment of atherosclerosis. Chin J Nat Med 2019; 17:401-412. [DOI: 10.1016/s1875-5364(19)30048-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Indexed: 02/07/2023]
|
132
|
Ahmed M, Baumgartner R, Aldi S, Dusart P, Hedin U, Gustafsson B, Caidahl K. Human serum albumin-based probes for molecular targeting of macrophage scavenger receptors. Int J Nanomedicine 2019; 14:3723-3741. [PMID: 31190821 PMCID: PMC6535103 DOI: 10.2147/ijn.s197990] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Accepted: 03/01/2019] [Indexed: 02/05/2023] Open
Abstract
Background: Inflammation and accumulation of macrophages are key features of unstable atherosclerotic plaques. The ability of macrophages to take up molecular probes can be exploited in new clinical imaging methods for the detection of unstable atherosclerotic lesions. We investigated whether modifications of human serum albumin (HSA) could be used to target macrophages efficiently in vitro. Materials and methods: Maleylated and aconitylated HSA were compared with unmodified HSA. Fluorescent or radiolabeled (89Zr) modified HSA was used in in vitro experiments to study cellular uptake by differentiated THP-1 cells and primary human macrophages. The time course of uptake was evaluated by flow cytometry, confocal microscopy, real-time microscopy and radioactivity measurements. The involvement of scavenger receptors (SR-A1, SR-B2, LOX-1) was assessed by knockdown experiments using RNA interference, by blocking experiments and by assays of competition by modified low-density lipoprotein. Results: Modified HSA was readily taken up by different macrophages. Uptake was mediated nonexclusively via the scavenger receptor SR-A1 (encoded by the MSR1 gene). Knockdown of CD36 and ORL1 had no influence on the uptake. Modified HSA was preferentially taken up by human macrophages compared with other vascular cell types such as endothelial cells and smooth muscle cells. Conclusions: Modified 89Zr-labeled HSA probes were recognized by different subsets of polarized macrophages, and maleylated HSA may be a promising radiotracer for radionuclide imaging of macrophage-rich inflammatory vascular diseases.
Collapse
Affiliation(s)
- Mona Ahmed
- Department of Molecular Medicine and Surgery, Karolinska Institutet, SE 17176, Stockholm, Sweden
| | - Roland Baumgartner
- Department of Medicine Solna, Karolinska Institutet, SE 17176, Stockholm, Sweden
| | - Silvia Aldi
- Section for Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE 17177, Stockholm, Sweden
| | - Philip Dusart
- Department of Cellular and Clinical Proteomics, Science for Life Laboratory, Kungliga Tekniska Högskolan (KTH), SE 17165, Stockholm, Sweden
| | - Ulf Hedin
- Department of Molecular Medicine and Surgery, Karolinska Institutet, SE 17176, Stockholm, Sweden
| | - Björn Gustafsson
- Department of Molecular Medicine and Surgery, Karolinska Institutet, SE 17176, Stockholm, Sweden
| | - Kenneth Caidahl
- Department of Molecular Medicine and Surgery, Karolinska Institutet, SE 17176, Stockholm, Sweden
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, SE 41345, Gothenburg, Sweden
| |
Collapse
|
133
|
Ma Z, Ketelhuth DFJ, Wirström T, Ohki T, Forteza MJ, Wang H, Grill V, Wollheim CB, Björklund A. Increased uptake of oxLDL does not exert lipotoxic effects in insulin-secreting cells. J Mol Endocrinol 2019; 62:159-168. [PMID: 30917339 DOI: 10.1530/jme-18-0146] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 02/27/2019] [Indexed: 01/13/2023]
Abstract
Modified lipoproteins can negatively affect beta cell function and survival. However, the mechanisms behind interactions of modified lipoproteins with beta cells - and in particular, relationships to increased uptake - are only partly clarified. By over-expressing the scavenger receptor CD36 (Tet-on), we increased the uptake of fluorescent low-density modified lipoprotein (oxLDL) into insulin-secreting INS-1 cells. The magnitude of uptake followed the degree of CD36 over-expression. CD36 over-expression increased concomitant efflux of 3H-cholesterol in proportion to the cellular contents of 3H-cholesterol. Exposure to concentrations of oxLDL from 20 to 100 µg/mL dose-dependently increased toxicity (evaluated by MTT) as well as apoptosis. However, the increased uptake of oxLDL due to CD36 over-expression did not exert additive effects on oxLDL toxicity - neither on viability, nor on glucose-induced insulin release and cellular content. Reciprocally, blocking CD36 receptors by Sulfo-N-Succinimidyl Oleate decreased the uptake of oxLDL but did not diminish the toxicity. Pancreatic islets of CD36-/- mice displayed reduced uptake of 3H-cholesterol-labeled oxLDL vs wild type but similar toxicity to oxLDL. OxLDL was found to increase the expression of CD36 in islets and INS-1 cells. In summary, given the experimental conditions, our results indicate that (1) increased uptake of oxLDL is not responsible for toxicity of oxLDL, (2) increased efflux of the cholesterol moiety of oxLDL counterbalances, at least in part, increased uptake and (3) oxLDL participates in the regulation of CD36 in pancreatic islets and in INS-1 cells.
Collapse
Affiliation(s)
- Z Ma
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - D F J Ketelhuth
- Department of Medicine, Centre for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - T Wirström
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - T Ohki
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - M J Forteza
- Department of Medicine, Centre for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - H Wang
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - V Grill
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Institute of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - C B Wollheim
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - A Björklund
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
134
|
Vrieling F, Wilson L, Rensen PCN, Walzl G, Ottenhoff THM, Joosten SA. Oxidized low-density lipoprotein (oxLDL) supports Mycobacterium tuberculosis survival in macrophages by inducing lysosomal dysfunction. PLoS Pathog 2019; 15:e1007724. [PMID: 30998773 PMCID: PMC6490946 DOI: 10.1371/journal.ppat.1007724] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 04/30/2019] [Accepted: 03/21/2019] [Indexed: 02/07/2023] Open
Abstract
Type 2 diabetes mellitus (DM) is a major risk factor for developing tuberculosis (TB). TB-DM comorbidity is expected to pose a serious future health problem due to the alarming rise in global DM incidence. At present, the causal underlying mechanisms linking DM and TB remain unclear. DM is associated with elevated levels of oxidized low-density lipoprotein (oxLDL), a pathologically modified lipoprotein which plays a key role during atherosclerosis development through the formation of lipid-loaded foamy macrophages, an event which also occurs during progression of the TB granuloma. We therefore hypothesized that oxLDL could be a common factor connecting DM to TB. To study this, we measured oxLDL levels in plasma samples of healthy controls, TB, DM and TB-DM patients, and subsequently investigated the effect of oxLDL treatment on human macrophage infection with Mycobacterium tuberculosis (Mtb). Plasma oxLDL levels were significantly elevated in DM patients and associated with high triglyceride levels in TB-DM. Strikingly, incubation with oxLDL strongly increased macrophage Mtb load compared to native or acetylated LDL (acLDL). Mechanistically, oxLDL -but not acLDL- treatment induced macrophage lysosomal cholesterol accumulation and increased protein levels of lysosomal and autophagy markers, while reducing Mtb colocalization with lysosomes. Importantly, combined treatment of acLDL and intracellular cholesterol transport inhibitor (U18666A) mimicked the oxLDL-induced lysosomal phenotype and impaired macrophage Mtb control, illustrating that the localization of lipid accumulation is critical. Collectively, these results demonstrate that oxLDL could be an important DM-associated TB-risk factor by causing lysosomal dysfunction and impaired control of Mtb infection in human macrophages. Tuberculosis (TB) is an infectious disease of the lungs caused by a bacterium, Mycobacterium tuberculosis (Mtb), and is responsible for over a million deaths per year worldwide. Population studies have demonstrated that type 2 diabetes mellitus (DM) is a risk factor for TB as it triples the risk of developing the disease. DM is a metabolic disorder which is generally associated with obesity, and is characterized by resistance to the pancreatic hormone insulin and high blood glucose and lipid levels. As the global incidence of DM is rising at an alarming rate, especially in regions where TB is common, it is important to understand precisely how DM increases the risk of developing TB. Both TB and DM are associated with the development of foamy macrophages, lipid-loaded white blood cells, which can be the result of a specific lipoprotein particle called oxidized low-density lipoprotein (oxLDL). Here, we demonstrated that DM patients have high blood levels of oxLDL, and generating foamy macrophages with oxLDL supported Mtb survival after infection as a result of faulty intracellular cholesterol accumulation. Our results propose a proof of concept for oxLDL as a risk factor for TB development, encouraging future studies on lipid-lowering therapies for TB-DM.
Collapse
Affiliation(s)
- Frank Vrieling
- Department of Infectious Diseases, Leiden University Medical Center, Albinusdreef 2, ZA Leiden, The Netherlands
| | - Louis Wilson
- Department of Infectious Diseases, Leiden University Medical Center, Albinusdreef 2, ZA Leiden, The Netherlands
| | - Patrick C. N. Rensen
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Albinusdreef 2, ZA Leiden, The Netherlands
| | - Gerhard Walzl
- DST/NRF Center of Excellence for Biomedical Tuberculosis Research, SA MRC Center for TB Research, Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences Stellenbosch University, Francie van Zijl Drive, Tygerberg, Cape Town, South Africa
| | - Tom H. M. Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Albinusdreef 2, ZA Leiden, The Netherlands
| | - Simone A. Joosten
- Department of Infectious Diseases, Leiden University Medical Center, Albinusdreef 2, ZA Leiden, The Netherlands
- * E-mail:
| |
Collapse
|
135
|
Oppi S, Lüscher TF, Stein S. Mouse Models for Atherosclerosis Research-Which Is My Line? Front Cardiovasc Med 2019; 6:46. [PMID: 31032262 PMCID: PMC6473202 DOI: 10.3389/fcvm.2019.00046] [Citation(s) in RCA: 123] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 03/26/2019] [Indexed: 12/24/2022] Open
Abstract
Atherosclerosis is one of the primary causes of cardiovascular disease and mortality. This chronic immunometabolic disease evolves during decades in humans and encompasses different organs and immune cell types, as well as local and systemic processes that promote the progression of the disease. The most frequently used animal model to study these atherogenic processes and inter-organ crosstalk in a short time frame are genetically modified mouse models. Some models have been used throughout the last decades, and some others been developed recently. These models have important differences in cholesterol and lipoprotein metabolism, reverse cholesterol transport pathway, obesity and diabetes as well as inflammatory processes. Therefore, the disease develops and progresses differently in the various mouse models. Since atherosclerosis is a multifaceted disease and many processes contribute to its progression, the choice of the right mouse model is important to study specific aspects of the disease. We will describe the different mouse models and provide a roadmap to facilitate current and future atherosclerosis researchers to choose the right model depending on their scientific question.
Collapse
Affiliation(s)
- Sara Oppi
- Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland
| | - Thomas F. Lüscher
- Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland
- Heart Division, Royal Brompton & Harefield Hospitals and Imperial College, London, United Kingdom
| | - Sokrates Stein
- Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
136
|
Abstract
PURPOSE OF REVIEW Smooth muscle cells (SMCs) are the major cell type in human atherosclerosis-prone arteries and take up excess lipids, thereby contributing to luminal occlusion. Here we provide a focused review on pathways by which smooth muscle cells (SMCs) can become foam cells in atherosclerosis. RECENT FINDINGS A synthesis of recent and older investigations provides key mechanistic insights into SMC foam cell formation. LDL and other apoB-containing lipoproteins are modified by a diverse array of oxidative, enzymatic, and nonenzymatic processes present in the arterial intima. These modifications of LDL all promote the aggregation of LDL (agLDL), a key finding from analysis of arterial lesion particles. Scavenger receptor and phagocytic capacity of SMCs can vary greatly, perhaps related to differences in SMC phenotype or in-vitro cell culture environments, and can be increased with exposure to cytokines, growth factors, and cholesterol. Macrophages promote the formation of SMC foam cells in direct or indirect co-culture models. SUMMARY SMCs contribute significantly to the foam cell population in atherosclerosis. Further investigation and identification of key mechanisms of SMC foam cell formation will help drive new therapeutics to reduce cardiovascular disease.
Collapse
MESH Headings
- Animals
- Apolipoproteins B/genetics
- Apolipoproteins B/metabolism
- Atherosclerosis/genetics
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Cell Differentiation
- Coculture Techniques
- Cytokines/pharmacology
- Foam Cells/drug effects
- Foam Cells/metabolism
- Foam Cells/pathology
- Gene Expression
- Humans
- Intercellular Signaling Peptides and Proteins/pharmacology
- Lipoproteins, LDL/genetics
- Lipoproteins, LDL/metabolism
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Phagocytosis
- Protein Aggregates/drug effects
- Receptors, Scavenger/genetics
- Receptors, Scavenger/metabolism
- Tunica Intima/drug effects
- Tunica Intima/metabolism
- Tunica Intima/pathology
Collapse
Affiliation(s)
- Collin S Pryma
- Department of Medicine, Centre for Heart Lung Innovation, Providence Healthcare Research Institute at St. Paul's Hospital, University of British Columbia, Vancouver, BC, Canada
| | | | | | | |
Collapse
|
137
|
Park J, Gao H, Wang Y, Hu H, Simon DI, Steinmetz NF. S100A9-targeted tobacco mosaic virus nanoparticles exhibit high specificity toward atherosclerotic lesions in ApoE -/- mice. J Mater Chem B 2019; 7:1842-1846. [PMID: 32255046 PMCID: PMC7147689 DOI: 10.1039/c8tb02276c] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
We integrate a biocompatible plant virus-based nanotechnology (tobacco mosaic virus, TMV) with S100A9-targeting peptides for its application in imaging and diagnosis of atherosclerosis. S100A9-targeted TMV nanoparticles exhibit remarkable specificity to S100A9 and targeting of atherosclerosis lesions in ApoE-/- mice.
Collapse
Affiliation(s)
- Jooneon Park
- Department of Nanoengineering, University of California San Diego, La Jolla, CA 92093, USA.
| | | | | | | | | | | |
Collapse
|
138
|
Takemura Y, Okamoto M, Hasegawa M, Hatanaka K, Kubota S. Protamine may have anti-atherogenic potential by inhibiting the binding of oxidized-low density lipoprotein to LOX-1. Biosci Biotechnol Biochem 2019; 83:1094-1101. [PMID: 30871430 DOI: 10.1080/09168451.2019.1588096] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Oxidized low-density lipoprotein (ox-LDL) leads to atherosclerosis via lectin-like oxidized lipoprotein receptor-1 (LOX-1), one of the major receptor for ox-LDL. Inhibition of the binding of ox-LDL to LOX-1 decreases the proinflammatory and atherosclerotic events. The aim of the present study was to investigate whether protamine, a polybasic nuclear protein, interferes the binding of ox-LDL to LOX-1. Using sandwich ELISA with newly generated antibody, we measured the blocking effect of protamine on the binding of ox-LDL to LOX-1. Protamine dose-dependently inhibited the binding of ox-LDL to LOX-1. DiI-labeled ox-LDL uptake assay in two types of cultured human endothelial cells was performed with fluorescence microplate reader. Activation of extracellular-signal-regulated kinase (ERK)1/2 by ox-LDL was analyzed by immunoblotting. We found that protamine suppressed uptake of ox-LDL in endothelial cells and inhibited ERK1/2 activation by ox-LDL. These results suggest that protamine may possess anti-atherogenic potential by inhibiting ox-LDL binding to LOX-1 through electrostatic interactions.
Collapse
Affiliation(s)
- Yukitoshi Takemura
- a Institute of Industrial Science , The University of Tokyo , Tokyo , Japan.,b Department of Pharmaceutical Sciences , Teikyo Heisei University , Tokyo , Japan
| | - Masaki Okamoto
- b Department of Pharmaceutical Sciences , Teikyo Heisei University , Tokyo , Japan
| | - Makoto Hasegawa
- b Department of Pharmaceutical Sciences , Teikyo Heisei University , Tokyo , Japan
| | - Kenichi Hatanaka
- a Institute of Industrial Science , The University of Tokyo , Tokyo , Japan
| | - Shunichiro Kubota
- b Department of Pharmaceutical Sciences , Teikyo Heisei University , Tokyo , Japan.,c Department of Life Sciences , Graduate School of Arts and Sciences, The University of Tokyo , Tokyo , Japan
| |
Collapse
|
139
|
Fan J, Liu L, Liu Q, Cui Y, Yao B, Zhang M, Gao Y, Fu Y, Dai H, Pan J, Qiu Y, Liu CH, He F, Wang Y, Zhang L. CKIP-1 limits foam cell formation and inhibits atherosclerosis by promoting degradation of Oct-1 by REGγ. Nat Commun 2019; 10:425. [PMID: 30683852 PMCID: PMC6347643 DOI: 10.1038/s41467-018-07895-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 12/05/2018] [Indexed: 01/12/2023] Open
Abstract
Atherosclerosis-related cardiovascular diseases are the leading cause of mortality worldwide. Macrophages uptake modified lipoproteins and transform into foam cells, triggering an inflammatory response and thereby promoting plaque formation. Here we show that casein kinase 2-interacting protein-1 (CKIP-1) is a suppressor of foam cell formation and atherosclerosis. Ckip-1 deficiency in mice leads to increased lipoprotein uptake and foam cell formation, indicating a protective role of CKIP-1 in this process. Ablation of Ckip-1 specifically upregulates the transcription of scavenger receptor LOX-1, but not that of CD36 and SR-A. Mechanistically, CKIP-1 interacts with the proteasome activator REGγ and targets the transcriptional factor Oct-1 for degradation, thereby suppressing the transcription of LOX-1 by Oct-1. Moreover, Ckip-1-deficient mice undergo accelerated atherosclerosis, and bone marrow transplantation reveals that Ckip-1 deficiency in hematopoietic cells is sufficient to increase atherosclerotic plaque formation. Therefore, CKIP-1 plays an essential anti-atherosclerotic role through regulation of foam cell formation and cholesterol metabolism. In atherosclerotic plaques, transformation of macrophages into foam cells is a key step in initiating the inflammatory response. Here Fan et al. show that casein kinase 2-interacting protein-1 (CKIP-1) limits foam cell formation and atherosclerosis by preventing expression of the scavenger receptor LOX-1 through REGγ-mediated degradation of Oct-1.
Collapse
Affiliation(s)
- Jiao Fan
- State Key Laboratory of Proteomics, National Center of Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 100850, China.,Institute of Geriatrics, National Clinical Research Center of Geriatrics Disease, Chinese PLA General Hospital, Beijing, 100853, China
| | - Lifeng Liu
- Department of Cardiology, Chinese PLA General Hospital, Beijing, 100853, China
| | - Qingyan Liu
- Center of Therapeutic Research for Liver Cancer, 302 Military Hospital of China, Beijing, 100039, China
| | - Yu Cui
- State Key Laboratory of Proteomics, National Center of Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 100850, China
| | - Binwei Yao
- Department of Experimental Pathology, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Minghua Zhang
- Clinical Pharmacy Laboratory, Chinese PLA General Hospital, Beijing, 100853, China
| | - Yabing Gao
- Department of Experimental Pathology, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Yesheng Fu
- State Key Laboratory of Proteomics, National Center of Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 100850, China
| | - Hongmiao Dai
- State Key Laboratory of Proteomics, National Center of Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 100850, China
| | - Jingkun Pan
- Institute of Geriatrics, National Clinical Research Center of Geriatrics Disease, Chinese PLA General Hospital, Beijing, 100853, China
| | - Ya Qiu
- Institute of Geriatrics, National Clinical Research Center of Geriatrics Disease, Chinese PLA General Hospital, Beijing, 100853, China
| | - Cui Hua Liu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Fuchu He
- State Key Laboratory of Proteomics, National Center of Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 100850, China
| | - Yu Wang
- Department of Cardiology, Chinese PLA General Hospital, Beijing, 100853, China.
| | - Lingqiang Zhang
- State Key Laboratory of Proteomics, National Center of Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 100850, China.
| |
Collapse
|
140
|
Nahon JE, Hoekstra M, van Hulst S, Manta C, Goerdt S, Geerling JJ, Géraud C, Van Eck M. Hematopoietic Stabilin-1 deficiency does not influence atherosclerosis susceptibility in LDL receptor knockout mice. Atherosclerosis 2019; 281:47-55. [PMID: 30658191 DOI: 10.1016/j.atherosclerosis.2018.12.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 12/02/2018] [Accepted: 12/05/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND AIMS Stabilin-1 (STAB1) is a scavenger receptor expressed on alternatively activated macrophages and sinusoidal endothelial cells. Its ligands include oxidized low-density lipoprotein (LDL) and the extracellular matrix glycoprotein SPARC and it is present in both human and murine atherosclerotic lesions. We aimed to investigate the effect of specific deletion of STAB1 in bone marrow-derived cells, including macrophages on atherosclerotic lesion formation in mice. METHODS Lethally irradiated hypercholesterolemic LDL receptor knockout mice received either wildtype (WT) or STAB1 knockout (STAB1 KO) bone marrow. Bone marrow transplanted mice were fed a Western-type diet for 9 weeks to induce atherosclerotic lesion formation. RESULTS Interestingly, LDL receptor knockout mice reconstituted with STAB1 KO bone marrow showed increased body weight gain (two-way ANOVA: p < 0.001) and larger white adipocyte cell sizes (43% increase in cell area; p < 0.05) as compared to WT bone marrow transplanted mice, which correlated positively (r = 0.82; p < 0.001). This was paralleled by a significant increase in white adipose tissue relative mRNA expression levels of the adipokine leptin (+94% p < 0.05). Despite these changes, no differences in serum lipid levels, the extent of in vivo macrophage foam cell formation or circulating leukocyte concentrations were observed. Moreover, the size and composition of atherosclerotic lesions was not different between the two experimental groups. CONCLUSIONS Bone marrow-specific Stabilin-1 deletion does not affect the susceptibility for atherosclerosis in mice. However, the increased body weight gain and adipocyte cell size highlight a potential role for leukocyte STAB1 in the development of metabolic disorders.
Collapse
Affiliation(s)
- Joya E Nahon
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR), Gorlaeus Laboratories, Einsteinweg 55, 2333CC, Leiden, the Netherlands
| | - Menno Hoekstra
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR), Gorlaeus Laboratories, Einsteinweg 55, 2333CC, Leiden, the Netherlands.
| | - Silvia van Hulst
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR), Gorlaeus Laboratories, Einsteinweg 55, 2333CC, Leiden, the Netherlands
| | - Calin Manta
- Department of Dermatology, Venereology, and Allergology, University Medical Center and Medical Faculty Mannheim, Heidelberg University, And Center of Excellence in Dermatology, Mannheim, Germany
| | - Sergij Goerdt
- Department of Dermatology, Venereology, and Allergology, University Medical Center and Medical Faculty Mannheim, Heidelberg University, And Center of Excellence in Dermatology, Mannheim, Germany; European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Janine J Geerling
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR), Gorlaeus Laboratories, Einsteinweg 55, 2333CC, Leiden, the Netherlands
| | - Cyrill Géraud
- Department of Dermatology, Venereology, and Allergology, University Medical Center and Medical Faculty Mannheim, Heidelberg University, And Center of Excellence in Dermatology, Mannheim, Germany; European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Section of Clinical and Molecular Dermatology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Miranda Van Eck
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR), Gorlaeus Laboratories, Einsteinweg 55, 2333CC, Leiden, the Netherlands
| |
Collapse
|
141
|
Sukhorukov V, Gudelj I, Pučić-Baković M, Zakiev E, Orekhov A, Kontush A, Lauc G. Glycosylation of human plasma lipoproteins reveals a high level of diversity, which directly impacts their functional properties. Biochim Biophys Acta Mol Cell Biol Lipids 2019; 1864:643-653. [PMID: 30641224 DOI: 10.1016/j.bbalip.2019.01.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 12/18/2018] [Accepted: 01/06/2019] [Indexed: 01/25/2023]
Abstract
AIMS Human plasma lipoproteins are known to contain various glycan structures whose composition and functional importance are starting to be recognized. We assessed N-glycosylation of human plasma HDL and LDL and the role of their glycomes in cellular cholesterol metabolism. METHODS N-glycomic profiles of native and neuraminidase-treated HDL and LDL were obtained using HILIC-UHPLC-FLD. Relative abundance of the individual chromatographic peaks was quantitatively expressed as a percentage of total integrated area and N-glycan structures present in each peak were elucidated by MALDI-TOF MS. The capacity of HDL to mediate cellular efflux of cholesterol and the capacity of LDL to induce cellular accumulation of cholesteryl esters were evaluated in THP-1 cells. RESULTS HILIC-UHPLC-FLD analysis of HDL and LDL N-glycans released by PNGase F resulted in 22 and 18 distinct chromatographic peaks, respectively. The majority of N-glycans present in HDL (~70%) and LDL (~60%) were sialylated with one or two sialic acid residues. The most abundant N-glycan structure in both HDL and LDL was a complex type biantennary N-glycan with one sialic acid (A2G2S1). Relative abundances of several N-glycan structures were dramatically altered by the neuraminidase treatment, which selectively removed sialic acid residues. Native HDL displayed significantly greater efficacy in removing cellular cholesterol from THP-1 cells as compared to desialylated HDL (p < 0.05). Cellular accumulation of cholesteryl esters in THP-1 cells was significantly higher after incubations with desialylated LDL particles as compared to native LDL (p < 0.05). CONCLUSIONS N-glycome of human plasma lipoproteins reveals a high level of diversity, which directly impacts functional properties of the lipoproteins.
Collapse
Affiliation(s)
- Vasily Sukhorukov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia; National Institute for Health and Medical Research (INSERM), UMR 1166 ICAN, Paris F-75013, France; Sorbonne University, Paris F-75013, France; AP-HP, Groupe hospitalier Pitié-Salpétrière, Paris F-75013, France; Federal State Budget Institution of Sciences Institute of Gene Biology, Russian Academy of Sciences, Vavilova Str., 34/5, Moscow 119334, Russia
| | - Ivan Gudelj
- Genos Glycoscience Research Laboratory, Borongajska cesta 83H, HR-10 000 Zagreb, Croatia
| | - Maja Pučić-Baković
- Genos Glycoscience Research Laboratory, Borongajska cesta 83H, HR-10 000 Zagreb, Croatia
| | - Emile Zakiev
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia; National Institute for Health and Medical Research (INSERM), UMR 1166 ICAN, Paris F-75013, France; Sorbonne University, Paris F-75013, France; AP-HP, Groupe hospitalier Pitié-Salpétrière, Paris F-75013, France; Federal State Budget Institution of Sciences Institute of Gene Biology, Russian Academy of Sciences, Vavilova Str., 34/5, Moscow 119334, Russia
| | - Alexander Orekhov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia; Institute for Atherosclerosis Research, Skolkovo Innovative Center, 121609 Moscow, Russia
| | - Anatol Kontush
- National Institute for Health and Medical Research (INSERM), UMR 1166 ICAN, Paris F-75013, France; Sorbonne University, Paris F-75013, France; AP-HP, Groupe hospitalier Pitié-Salpétrière, Paris F-75013, France.
| | - Gordan Lauc
- Genos Glycoscience Research Laboratory, Borongajska cesta 83H, HR-10 000 Zagreb, Croatia
| |
Collapse
|
142
|
Kawanishi N, Mizokami T, Yada K, Suzuki K. Exercise training suppresses scavenger receptor CD36 expression in kupffer cells of nonalcoholic steatohepatitis model mice. Physiol Rep 2018; 6:e13902. [PMID: 30516003 PMCID: PMC6279959 DOI: 10.14814/phy2.13902] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 06/01/2018] [Accepted: 06/06/2018] [Indexed: 12/13/2022] Open
Abstract
Although nonalcoholic steatohepatitis (NASH) is an important component of the metabolic syndrome, scavenger receptor CD36 also modulates NASH development. This study aimed to clarify whether exercise training suppresses CD36 expression in a mouse model of NASH. Male C57BL/6 mice were divided into four groups: normal diet (ND) sedentary, ND exercise, high-fat diet and high-fructose water (HFF) sedentary, and HFF exercise groups. The exercise groups were trained on a motorized treadmill at running speeds of 15-20 m/min for 60 min/day, 5 times/week for 16 weeks. CD36 cell surface expression of hepatic resident macrophages, peroxisome proliferator-activated receptor (PPAR)-γ protein, and mRNA levels in the liver were increased in HFF sedentary mice but were attenuated in HFF exercise mice. Hepatic resident macrophages were significantly lower in HFF exercise mice than in HFF sedentary mice. Our findings indicated that exercise training reduced macrophage quantity in the liver, and downregulated CD36 and PPAR-γ expression in liver and macrophages.
Collapse
Affiliation(s)
- Noriaki Kawanishi
- Graduate School of Sport SciencesWaseda UniversityTokorozawaSaitamaJapan
- Faculty of Advanced EngineeringChiba Institute of TechnologyNarashinoChibaJapan
- Research Fellow of the Japan Society for the Promotion of SciencesChiyoda‐ku, TokyoJapan
| | - Tsubasa Mizokami
- Graduate School of Sport SciencesWaseda UniversityTokorozawaSaitamaJapan
| | - Koichi Yada
- Faculty of Sport SciencesWaseda UniversityTokorozawaSaitamaJapan
| | - Katsuhiko Suzuki
- Faculty of Sport SciencesWaseda UniversityTokorozawaSaitamaJapan
| |
Collapse
|
143
|
Grajchen E, Hendriks JJA, Bogie JFJ. The physiology of foamy phagocytes in multiple sclerosis. Acta Neuropathol Commun 2018; 6:124. [PMID: 30454040 PMCID: PMC6240956 DOI: 10.1186/s40478-018-0628-8] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 11/02/2018] [Indexed: 12/15/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic disease of the central nervous system characterized by massive infiltration of immune cells, demyelination, and axonal loss. Active MS lesions mainly consist of macrophages and microglia containing abundant intracellular myelin remnants. Initial studies showed that these foamy phagocytes primarily promote MS disease progression by internalizing myelin debris, presenting brain-derived autoantigens, and adopting an inflammatory phenotype. However, more recent studies indicate that phagocytes can also adopt a beneficial phenotype upon myelin internalization. In this review, we summarize and discuss the current knowledge on the spatiotemporal physiology of foamy phagocytes in MS lesions, and elaborate on extrinsic and intrinsic factors regulating their behavior. In addition, we discuss and link the physiology of myelin-containing phagocytes to that of foamy macrophages in other disorders such atherosclerosis.
Collapse
Affiliation(s)
- Elien Grajchen
- Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium/School of Life Sciences, Transnationale Universiteit Limburg, Diepenbeek, Belgium
| | - Jerome J A Hendriks
- Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium/School of Life Sciences, Transnationale Universiteit Limburg, Diepenbeek, Belgium
| | - Jeroen F J Bogie
- Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium/School of Life Sciences, Transnationale Universiteit Limburg, Diepenbeek, Belgium.
| |
Collapse
|
144
|
Benito-Vicente A, Uribe KB, Jebari S, Galicia-Garcia U, Ostolaza H, Martin C. Familial Hypercholesterolemia: The Most Frequent Cholesterol Metabolism Disorder Caused Disease. Int J Mol Sci 2018; 19:ijms19113426. [PMID: 30388787 PMCID: PMC6275065 DOI: 10.3390/ijms19113426] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 10/21/2018] [Accepted: 10/29/2018] [Indexed: 12/18/2022] Open
Abstract
Cholesterol is an essential component of cell barrier formation and signaling transduction involved in many essential physiologic processes. For this reason, cholesterol metabolism must be tightly controlled. Cell cholesterol is mainly acquired from two sources: Dietary cholesterol, which is absorbed in the intestine and, intracellularly synthesized cholesterol that is mainly synthesized in the liver. Once acquired, both are delivered to peripheral tissues in a lipoprotein dependent mechanism. Malfunctioning of cholesterol metabolism is caused by multiple hereditary diseases, including Familial Hypercholesterolemia, Sitosterolemia Type C and Niemann-Pick Type C1. Of these, familial hypercholesterolemia (FH) is a common inherited autosomal co-dominant disorder characterized by high plasma cholesterol levels. Its frequency is estimated to be 1:200 and, if untreated, increases the risk of premature cardiovascular disease. This review aims to summarize the current knowledge on cholesterol metabolism and the relation of FH to cholesterol homeostasis with special focus on the genetics, diagnosis and treatment.
Collapse
Affiliation(s)
- Asier Benito-Vicente
- Departamento de Bioquímica, Instituto Biofisika (UPV/EHU, CSIC), Universidad del País Vasco, Apdo.644, 48080 Bilbao, Spain.
| | - Kepa B Uribe
- Departamento de Bioquímica, Instituto Biofisika (UPV/EHU, CSIC), Universidad del País Vasco, Apdo.644, 48080 Bilbao, Spain.
| | - Shifa Jebari
- Departamento de Bioquímica, Instituto Biofisika (UPV/EHU, CSIC), Universidad del País Vasco, Apdo.644, 48080 Bilbao, Spain.
| | - Unai Galicia-Garcia
- Departamento de Bioquímica, Instituto Biofisika (UPV/EHU, CSIC), Universidad del País Vasco, Apdo.644, 48080 Bilbao, Spain.
| | - Helena Ostolaza
- Departamento de Bioquímica, Instituto Biofisika (UPV/EHU, CSIC), Universidad del País Vasco, Apdo.644, 48080 Bilbao, Spain.
| | - Cesar Martin
- Departamento de Bioquímica, Instituto Biofisika (UPV/EHU, CSIC), Universidad del País Vasco, Apdo.644, 48080 Bilbao, Spain.
| |
Collapse
|
145
|
Yang Q, Yin RX, Cao XL, Huang F, Zhou YJ, Chen WX. ANGPTL4 variants and their haplotypes are associated with serum lipid levels, the risk of coronary artery disease and ischemic stroke and atorvastatin cholesterol-lowering responses. Nutr Metab (Lond) 2018; 15:70. [PMID: 30323852 PMCID: PMC6173870 DOI: 10.1186/s12986-018-0308-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 09/26/2018] [Indexed: 12/20/2022] Open
Abstract
Background This study aimed to assess the association between the angiopoietin-like protein 4 gene (ANGPTL4) single nucleotide polymorphisms (SNPs) and serum lipid levels, the risk of coronary artery disease (CAD) and ischemic stroke (IS), and response to atorvastatin therapy in a Southern Chinese Han population. Methods Genotypes of the ANGPTL4 rs4076317, rs7255436, rs1044250 and rs2967605 SNPs in 1,654 unrelated subjects (CAD, 568; IS, 537; and controls, 549) were determined by the Snapshot technology. Another group of 724 hyperlipidemic patients was selected and treated with atorvastatin calcium tablet 20 mg/day for 8 weeks. Results The rs2967605 CT/TT genotypes were associated with a decreased risk of CAD (adjusted OR = 0.68, 95% CI = 0.47-0.99, P = 0.043 for CT/TT vs. CC) and IS (adjusted OR = 0.55, 95% CI = 0.38-0.80, P = 0.020 for CT/TT vs. CC). There was no significant association between the four SNPs and angiographic severity of CAD. The subjects with the rs4076317 CG/CC genotypes in controls had higher total cholesterol (TC) and low-density lipoprotein cholesterol (LDL-C) levels than the subjects with the GG genotype (P < 0.001; a P < 0.0018 was regarded statistically significant by the Bonferroni correction). The subjects with rs4076317CG/GG genotypes had lower TC and LDL-C levels than the subjects with CC genotype after atorvastatin treatment (P < 0.001). Conclusions The observed associations suggest that the ANGPTL4 variants have a potential role on serum lipid levels and atherosclerosis-related diseases in the Chinese Han population, especially the ANGPTL4 rs4076317 and rs2967605 SNPs.
Collapse
Affiliation(s)
- Qian Yang
- 1Department of Cardiology, Institute of Cardiovascular Diseases, the First Affiliated Hospital, Guangxi Medical University, Nanning, 530021 Guangxi People's Republic of China
| | - Rui-Xing Yin
- 1Department of Cardiology, Institute of Cardiovascular Diseases, the First Affiliated Hospital, Guangxi Medical University, Nanning, 530021 Guangxi People's Republic of China
| | - Xiao-Li Cao
- 2Department of Neurology, the First Affiliated Hospital, Guangxi Medical University, Nanning, 530021 Guangxi People's Republic of China
| | - Feng Huang
- 1Department of Cardiology, Institute of Cardiovascular Diseases, the First Affiliated Hospital, Guangxi Medical University, Nanning, 530021 Guangxi People's Republic of China
| | - Yi-Jiang Zhou
- 1Department of Cardiology, Institute of Cardiovascular Diseases, the First Affiliated Hospital, Guangxi Medical University, Nanning, 530021 Guangxi People's Republic of China
| | - Wu-Xian Chen
- 1Department of Cardiology, Institute of Cardiovascular Diseases, the First Affiliated Hospital, Guangxi Medical University, Nanning, 530021 Guangxi People's Republic of China
| |
Collapse
|
146
|
Zhao ZW, Zhang M, Chen LY, Gong D, Xia XD, Yu XH, Wang SQ, Ou X, Dai XY, Zheng XL, Zhang DW, Tang CK. Heat shock protein 70 accelerates atherosclerosis by downregulating the expression of ABCA1 and ABCG1 through the JNK/Elk-1 pathway. Biochim Biophys Acta Mol Cell Biol Lipids 2018; 1863:806-822. [DOI: 10.1016/j.bbalip.2018.04.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 03/30/2018] [Accepted: 04/15/2018] [Indexed: 12/14/2022]
|
147
|
Maréchal L, Laviolette M, Rodrigue-Way A, Sow B, Brochu M, Caron V, Tremblay A. The CD36-PPARγ Pathway in Metabolic Disorders. Int J Mol Sci 2018; 19:1529. [PMID: 29883404 PMCID: PMC5983591 DOI: 10.3390/ijms19051529] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Revised: 05/08/2018] [Accepted: 05/16/2018] [Indexed: 12/21/2022] Open
Abstract
Uncovering the biological role of nuclear receptor peroxisome proliferator-activated receptors (PPARs) has greatly advanced our knowledge of the transcriptional control of glucose and energy metabolism. As such, pharmacological activation of PPARγ has emerged as an efficient approach for treating metabolic disorders with the current use of thiazolidinediones to improve insulin resistance in diabetic patients. The recent identification of growth hormone releasing peptides (GHRP) as potent inducers of PPARγ through activation of the scavenger receptor CD36 has defined a novel alternative to regulate essential aspects of lipid and energy metabolism. Recent advances on the emerging role of CD36 and GHRP hexarelin in regulating PPARγ downstream actions with benefits on atherosclerosis, hepatic cholesterol biosynthesis and fat mitochondrial biogenesis are summarized here. The response of PPARγ coactivator PGC-1 is also discussed in these effects. The identification of the GHRP-CD36-PPARγ pathway in controlling various tissue metabolic functions provides an interesting option for metabolic disorders.
Collapse
Affiliation(s)
- Loïze Maréchal
- Research Center, CHU Sainte-Justine, Montréal, QC H3T 1C5, Canada.
- Department of Physiology, Faculty of Medicine, University of Montreal, Montréal, QC H3T 1J4, Canada.
| | - Maximilien Laviolette
- Research Center, CHU Sainte-Justine, Montréal, QC H3T 1C5, Canada.
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, University of Montreal, Montréal, QC H3T 1J4, Canada.
| | - Amélie Rodrigue-Way
- Research Center, CHU Sainte-Justine, Montréal, QC H3T 1C5, Canada.
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, University of Montreal, Montréal, QC H3T 1J4, Canada.
| | - Baly Sow
- Research Center, CHU Sainte-Justine, Montréal, QC H3T 1C5, Canada.
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, University of Montreal, Montréal, QC H3T 1J4, Canada.
| | - Michèle Brochu
- Department of Physiology, Faculty of Medicine, University of Montreal, Montréal, QC H3T 1J4, Canada.
| | - Véronique Caron
- Research Center, CHU Sainte-Justine, Montréal, QC H3T 1C5, Canada.
| | - André Tremblay
- Research Center, CHU Sainte-Justine, Montréal, QC H3T 1C5, Canada.
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, University of Montreal, Montréal, QC H3T 1J4, Canada.
- Centre de Recherche en Reproduction et Fertilité, University of Montreal, Saint Hyacinthe, QC J2S 7C6, Canada.
- Department of Obstetrics & Gynecology, Faculty of Medicine, University of Montreal, Montréal, QC H3T 1C5, Canada.
| |
Collapse
|
148
|
Lemos BS, Medina-Vera I, Malysheva OV, Caudill MA, Fernandez ML. Effects of Egg Consumption and Choline Supplementation on Plasma Choline and Trimethylamine-N-Oxide in a Young Population. J Am Coll Nutr 2018; 37:716-723. [PMID: 29764315 DOI: 10.1080/07315724.2018.1466213] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Background: Plasma trimethylamine-N-oxide (TMAO) concentrations have been associated with cardiovascular disease risk. Eggs are a rich source of choline, which is a precursor of TMAO.Objective: The effects of egg intake versus daily choline supplementation were evaluated on plasma choline and TMAO in a young, healthy population.Methods: Thirty participants (14 males, 16 females; 25.6 ± 2.3 years; body mass index = 24.3 ± 2.9 kg/m2) were enrolled in this 13-week crossover intervention. After a 2-week washout, participants were randomized to consume either 3 eggs/d or a choline bitartrate supplement (∼ 400 mg choline total in eggs or supplement) for 4 weeks. Following a 3-week washout, participants were switched to the alternate treatment. Dietary records were measured at the end of each period. Plasma TMAO and choline were measured at baseline and at the end of each dietary intervention. Gene expression of scavenger receptors associated with plasma TMAO were quantified at the end of each intervention.Results: Compared to the choline supplement, intake of total fat, cholesterol, selenium, and vitamin E were higher (p < 0.05), whereas carbohydrate intake was lower (p < 0.001) with consumption of 3 eggs/d. Fasting plasma choline increased 20% (p = 0.023) with egg intake, while no changes were observed with choline supplementation. Plasma TMAO levels were not different between dietary treatments or compared to baseline.Conclusions: Dietary choline appears to be more bioavailable via egg consumption when compared to a choline supplement. Plasma TMAO concentrations were not affected in healthy participants after 4 weeks of taking ∼400 mg/d choline either via eggs or choline supplementation.
Collapse
Affiliation(s)
- Bruno S Lemos
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT, US
| | - Isabel Medina-Vera
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT, US
| | - Olga V Malysheva
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, US
| | - Marie A Caudill
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, US
| | - Maria Luz Fernandez
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT, US
| |
Collapse
|
149
|
Poynter SJ, Monjo AL, DeWitte-Orr SJ. Identification of three class A scavenger receptors from rainbow trout (Oncorhynchus mykiss): SCARA3, SCARA4, and SCARA5. FISH & SHELLFISH IMMUNOLOGY 2018; 76:121-125. [PMID: 29471060 DOI: 10.1016/j.fsi.2018.02.029] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 02/06/2018] [Accepted: 02/14/2018] [Indexed: 06/08/2023]
Abstract
Class A scavenger receptors (SR-As) are a family of five surface receptors whose functions in mammals are associated with innate immunity; however, their role in fish immunity requires further elucidation. The present study identifies, performs sequence analysis, and constitutive transcript expression analysis for three SR-A family members, SCARA3, SCARA4 and SCARA5, from rainbow trout. This work will provide a basis for future studies on SR-A function and their role in innate immunity in this economically important fish.
Collapse
Affiliation(s)
- S J Poynter
- Department of Biology, University of Waterloo, Waterloo, Ontario, Canada
| | - A L Monjo
- Department of Biology, Wilfrid Laurier University, Waterloo, Ontario, Canada
| | - S J DeWitte-Orr
- Department of Biology, Wilfrid Laurier University, Waterloo, Ontario, Canada; Department of Health Sciences, Wilfrid Laurier University, Waterloo, Ontario, Canada.
| |
Collapse
|
150
|
Grajeda-Iglesias C, Rom O, Hamoud S, Volkova N, Hayek T, Abu-Saleh N, Aviram M. Leucine supplementation attenuates macrophage foam-cell formation: Studies in humans, mice, and cultured macrophages. Biofactors 2018; 44:245-262. [PMID: 29399895 DOI: 10.1002/biof.1415] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 01/06/2018] [Accepted: 01/09/2018] [Indexed: 01/07/2023]
Abstract
Whereas atherogenicity of dietary lipids has been largely studied, relatively little is known about the possible contribution of dietary amino acids to macrophage foam-cell formation, a hallmark of early atherogenesis. Recently, we showed that leucine has antiatherogenic properties in the macrophage model system. In this study, an in-depth investigation of the role of leucine in macrophage lipid metabolism was conducted by supplementing humans, mice, or cultured macrophages with leucine. Macrophage incubation with serum obtained from healthy adults supplemented with leucine (5 g/d, 3 weeks) significantly decreased cellular cholesterol mass by inhibiting the rate of cholesterol biosynthesis and increasing cholesterol efflux from macrophages. Similarly, leucine supplementation to C57BL/6 mice (8 weeks) resulted in decreased cholesterol content in their harvested peritoneal macrophages (MPM) in relation with reduced cholesterol biosynthesis rate. Studies in J774A.1 murine macrophages revealed that leucine dose-dependently decreased cellular cholesterol and triglyceride mass. Macrophages treated with leucine (0.2 mM) showed attenuated uptake of very low-density lipoproteins and triglyceride biosynthesis rate, with a concurrent down-regulation of diacylglycerol acyltransferase-1, a key enzyme catalyzing triglyceride biosynthesis in macrophages. Similar effects were observed when macrophages were treated with α-ketoisocaproate, a key leucine metabolite. Finally, both in vivo and in vitro leucine supplementation significantly improved macrophage mitochondrial respiration and ATP production. The above studies, conducted in human, mice, and cultured macrophages, highlight a protective role for leucine attenuating macrophage foam-cell formation by mechanisms related to the metabolism of cholesterol, triglycerides, and energy production. © 2018 BioFactors, 44(3):245-262, 2018.
Collapse
Affiliation(s)
- Claudia Grajeda-Iglesias
- The Lipid Research Laboratory, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Oren Rom
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Shadi Hamoud
- Department of Internal Medicine E, Rambam Health Care Campus and Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Nina Volkova
- The Lipid Research Laboratory, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Tony Hayek
- The Lipid Research Laboratory, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
- Department of Internal Medicine E, Rambam Health Care Campus and Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Niroz Abu-Saleh
- The Lipid Research Laboratory, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Michael Aviram
- The Lipid Research Laboratory, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| |
Collapse
|