1
|
Klersy T, Achner L, Fels B, Rezende F, Lopez M, Alenina N, Spiecker F, Stölting I, Häuser W, Reinberger T, Aherrahrou Z, Kuenne C, Vahldieck C, Matschl U, Hille S, Bader M, Brandes RP, Müller OJ, Kusche-Vihrog K, Raasch W. The anti-atherosclerotic effect of chronic AT1 receptor blocker treatment also depends on the ACE2/Ang(1-7)/Mas axis. Biomed Pharmacother 2025; 186:117990. [PMID: 40106968 DOI: 10.1016/j.biopha.2025.117990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 03/06/2025] [Accepted: 03/13/2025] [Indexed: 03/22/2025] Open
Abstract
Blockade of AT1-receptors by telmisartan (TEL) has anti-atherosclerotic efficacy. We investigated to what extent the ACE2/Ang1-7/Mas axis-dependent mechanism contributes to the TEL-induced protection of endothelial function. Atherosclerosis was induced in C57BL/6 N, Mas-knock out (ko), and Ace2-ko mice by AAV-PCSK9DY (2 ×1011 VG) injections plus Western diet (WD) feeding (12w). Mice were treated (12w) with TEL or vehicle. Controls received no PCSK9DY, chow-feeding, and vehicle-treatment. In the aortae of mice, the plaque burden was determined, RNAseq analyses were performed and functional properties were assessed by quantifying the mechanical properties of the endothelial surface by Atomic Force Microscopy. Regardless of strain, plaque burden and total cholesterol were increased upon AAV-PCSK9DY+WD but decreased by TEL. Cortical stiffness was also enhanced in all strains by AAV-PCSK9DY+WD but reduced under TEL only in the C57BL/6 N, while remaining still high in both knockout strains. Plasma NO negatively correlated with cortical stiffness in C57BL/6 N, but not in transgenic mice. TNFα plasma levels and aortic MMP12 expression was increased in PCSK9DY/WD vehicle-treated controls and was normalized by TEL in C57BL/6 N but not in Mas-ko and Ace2-ko mice. We conclude that TEL-induced reduction of endothelial stiffness occurred only in the C57BL/6 N but not in the Mas-ko and Ace2-ko mice. We suggest that the protective TEL effect is partly due to an Ang(1-7)/ACE2/Mas axis mediated mechanism. Since Mmp12 has well-known proatherogenic properties but was not altered in the two transgenic mouse lines, follow-up studies are required to further elucidate the correlation between Mmp12 and the Ang(1-7)/ACE2/Mas axis with respect to atherosclerosis.
Collapse
Affiliation(s)
- Tobias Klersy
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Germany
| | - Leonie Achner
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Germany
| | - Benedikt Fels
- Institute for Physiology, University Lübeck, Germany
| | - Flavia Rezende
- Institute for Cardiovascular Physiology, Faculty of Medicine, Goethe-University Frankfurt, Germany; DZHK (German Center for Cardiovascular Research) Partner site Rhine-Main, Germany
| | - Melina Lopez
- Institute for Cardiovascular Physiology, Faculty of Medicine, Goethe-University Frankfurt, Germany; DZHK (German Center for Cardiovascular Research) Partner site Rhine-Main, Germany
| | - Natalia Alenina
- Max-Delbrück-Center for Molecular Medicine (MDC), Berlin, Germany
| | - Frauke Spiecker
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Germany
| | - Ines Stölting
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Germany
| | - Walter Häuser
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Germany
| | - Tobias Reinberger
- Institute for Cardiogenetics, University Lübeck; University of Lübeck, Germany; DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Germany
| | - Zouhair Aherrahrou
- Institute for Cardiogenetics, University Lübeck; University of Lübeck, Germany; DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Germany
| | - Carsten Kuenne
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | | | - Urte Matschl
- Department Virus Immunology, Leibniz Institute for Virology, Hamburg, Germany
| | - Susanne Hille
- DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Germany; Department of Internal Medicine V, University Hospital Schleswig-Holstein, Campus Kiel, Germany
| | - Michael Bader
- Max-Delbrück-Center for Molecular Medicine (MDC), Berlin, Germany; DZHK (German Centre for Cardiovascular Research), partner site, Berlin, Germany; Center for Structural and Cell Biology in Medicine, Institute for Biology, University of Lübeck, Lübeck, Germany; Charité - University Medicine Berlin, Berlin, Germany
| | - Ralf P Brandes
- Institute for Cardiovascular Physiology, Faculty of Medicine, Goethe-University Frankfurt, Germany; DZHK (German Center for Cardiovascular Research) Partner site Rhine-Main, Germany
| | - Oliver J Müller
- DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Germany; Department of Internal Medicine V, University Hospital Schleswig-Holstein, Campus Kiel, Germany
| | - Kristina Kusche-Vihrog
- Institute for Physiology, University Lübeck, Germany; DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Germany
| | - Walter Raasch
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Germany; DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Germany; CBBM (Centre for Brain, Behavior and Metabolism), University of Lübeck, Germany.
| |
Collapse
|
2
|
Zhu L, Liu Z, Liu J, Li Z, Bao Y, Sun X, Zhao W, Zhou A, Wu H. NCOA4 linked to endothelial cell ferritinophagy and ferroptosis:a key regulator aggravate aortic endothelial inflammation and atherosclerosis. Redox Biol 2025; 79:103465. [PMID: 39700692 PMCID: PMC11729014 DOI: 10.1016/j.redox.2024.103465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 11/26/2024] [Accepted: 12/09/2024] [Indexed: 12/21/2024] Open
Abstract
Atherosclerosis (AS) is associated with a high incidence of cardiovascular events, yet the mechanisms underlying this association remain unclear. Our previous study found that Atherosclerotic endothelial injury is closely associated with ferroptosis in ApoE-/- mice. Ferroptosis is a novel mode of cell death induced by decreased antioxidant capacity of the organism and accumulation of reactive oxygen species. Nuclear receptor coactivator 4 (NCOA4)-mediated ferritinophagy is an important regulator of sudden ferroptosis in cells. However, the role of NCOA4 in AS and the exact mechanism by which it regulates the ferritinophagy response remain unclear. Herein, we report that NCOA4 expression is elevated in ApoE-/- mice and endothelial cells and is significantly correlated with AS. NCOA4 expression promoted ferroptosis, and was positively correlated with ferritinophagy response. Mechanistically, our findings indicate that LOX-1 is a key upstream target that influences the function of NCOA4. The specific pathway is related to the activation of cGAS-STING signaling to upregulate NCOA4 expression. Moreover, our findings demonstrate the "Gualou-Xiebai" herb pair can regulate LOX-1 to inhibit ferroptosis. Collectively, our results provide evidence of a connection between NCOA4-mediated promotion of AS and suggest that targeting upstream molecules regulating NCOA4 could be a potential therapy for AS.
Collapse
Affiliation(s)
- Li Zhu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China; Anhui Province Key Laboratory of Bioactive Natural Products
| | - Zijian Liu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China; Anhui Province Key Laboratory of Bioactive Natural Products
| | - Jiahui Liu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China; Anhui Province Key Laboratory of Bioactive Natural Products
| | - Zhenglong Li
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China; Anhui Province Key Laboratory of Bioactive Natural Products
| | - Youli Bao
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China; Anhui Province Key Laboratory of Bioactive Natural Products
| | - Xin Sun
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China; Anhui Province Key Laboratory of Bioactive Natural Products
| | - Wenchen Zhao
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, 15219, USA
| | - An Zhou
- Anhui Province Key Laboratory of Bioactive Natural Products.
| | - Hongfei Wu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China; Anhui Province Key Laboratory of Bioactive Natural Products.
| |
Collapse
|
3
|
Noels H, Jankowski V, Schunk SJ, Vanholder R, Kalim S, Jankowski J. Post-translational modifications in kidney diseases and associated cardiovascular risk. Nat Rev Nephrol 2024; 20:495-512. [PMID: 38664592 DOI: 10.1038/s41581-024-00837-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2024] [Indexed: 07/21/2024]
Abstract
Patients with chronic kidney disease (CKD) are at an increased cardiovascular risk compared with the general population, which is driven, at least in part, by mechanisms that are uniquely associated with kidney disease. In CKD, increased levels of oxidative stress and uraemic retention solutes, including urea and advanced glycation end products, enhance non-enzymatic post-translational modification events, such as protein oxidation, glycation, carbamylation and guanidinylation. Alterations in enzymatic post-translational modifications such as glycosylation, ubiquitination, acetylation and methylation are also detected in CKD. Post-translational modifications can alter the structure and function of proteins and lipoprotein particles, thereby affecting cellular processes. In CKD, evidence suggests that post-translationally modified proteins can contribute to inflammation, oxidative stress and fibrosis, and induce vascular damage or prothrombotic effects, which might contribute to CKD progression and/or increase cardiovascular risk in patients with CKD. Consequently, post-translational protein modifications prevalent in CKD might be useful as diagnostic biomarkers and indicators of disease activity that could be used to guide and evaluate therapeutic interventions, in addition to providing potential novel therapeutic targets.
Collapse
Affiliation(s)
- Heidi Noels
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, Aachen, Germany.
- Aachen-Maastricht Institute for Cardiorenal Disease (AMICARE), University Hospital RWTH Aachen, Aachen, Germany.
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, Netherlands.
| | - Vera Jankowski
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, Aachen, Germany
- Aachen-Maastricht Institute for Cardiorenal Disease (AMICARE), University Hospital RWTH Aachen, Aachen, Germany
| | - Stefan J Schunk
- Department of Internal Medicine IV, Nephrology and Hypertension, Saarland University, Homburg/Saar, Germany
| | - Raymond Vanholder
- Nephrology Section, Department of Internal Medicine and Paediatrics, University Hospital, Ghent, Belgium
- European Kidney Health Alliance (EKHA), Brussels, Belgium
| | - Sahir Kalim
- Department of Medicine, Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Joachim Jankowski
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, Aachen, Germany.
- Aachen-Maastricht Institute for Cardiorenal Disease (AMICARE), University Hospital RWTH Aachen, Aachen, Germany.
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, Netherlands.
| |
Collapse
|
4
|
Wen SY, Zhi X, Liu HX, Wang X, Chen YY, Wang L. Is the suppression of CD36 a promising way for atherosclerosis therapy? Biochem Pharmacol 2024; 219:115965. [PMID: 38043719 DOI: 10.1016/j.bcp.2023.115965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 11/29/2023] [Accepted: 11/30/2023] [Indexed: 12/05/2023]
Abstract
Atherosclerosis is the main underlying pathology of many cardiovascular diseases and is marked by plaque formation in the artery wall. It has posed a serious threat to the health of people all over the world. CD36 acts as a significant regulator of lipid homeostasis, which is closely associated with the onset and progression of atherosclerosis and may be a new therapeutic target. The abnormal overexpression of CD36 facilitates lipid accumulation, foam cell formation, inflammation, endothelial apoptosis, and thrombosis. Numerous natural products and lipid-lowering agents are found to target the suppression of CD36 or inhibit the upregulation of CD36 to prevent and treat atherosclerosis. Here, the structure, expression regulation and function of CD36 in atherosclerosis and its related pharmacological therapies are reviewed. This review highlights the importance of drugs targeting CD36 suppression in the treatment and prevention of atherosclerosis, in order to develop new therapeutic strategies and potential anti-atherosclerotic drugs both preclinically and clinically.
Collapse
Affiliation(s)
- Shi-Yuan Wen
- College of Basic Medical Sciences, Shanxi Medical University, Taiyuan, China
| | - Xiaoyan Zhi
- College of Basic Medical Sciences, Shanxi Medical University, Taiyuan, China
| | - Hai-Xin Liu
- School of Traditional Chinese Materia Medica, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Xiaohui Wang
- College of Basic Medical Sciences, Shanxi Medical University, Taiyuan, China
| | - Yan-Yan Chen
- School of Medicine, Jiangsu University, Zhenjiang, China.
| | - Li Wang
- College of Basic Medical Sciences, Shanxi Medical University, Taiyuan, China.
| |
Collapse
|
5
|
Schoch L, Alcover S, Padró T, Ben-Aicha S, Mendieta G, Badimon L, Vilahur G. Update of HDL in atherosclerotic cardiovascular disease. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE ARTERIOSCLEROSIS 2023; 35:297-314. [PMID: 37940388 DOI: 10.1016/j.arteri.2023.10.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 10/13/2023] [Indexed: 11/10/2023]
Abstract
Epidemiologic evidence supported an inverse association between HDL (high-density lipoprotein) cholesterol (HDL-C) levels and atherosclerotic cardiovascular disease (ASCVD), identifying HDL-C as a major cardiovascular risk factor and postulating diverse HDL vascular- and cardioprotective functions beyond their ability to drive reverse cholesterol transport. However, the failure of several clinical trials aimed at increasing HDL-C in patients with overt cardiovascular disease brought into question whether increasing the cholesterol cargo of HDL was an effective strategy to enhance their protective properties. In parallel, substantial evidence supports that HDLs are complex and heterogeneous particles whose composition is essential for maintaining their protective functions, subsequently strengthening the "HDL quality over quantity" hypothesis. The following state-of-the-art review covers the latest understanding as per the roles of HDL in ASCVD, delves into recent advances in understanding the complexity of HDL particle composition, including proteins, lipids and other HDL-transported components and discusses on the clinical outcomes after the administration of HDL-C raising drugs with particular attention to CETP (cholesteryl ester transfer protein) inhibitors.
Collapse
Affiliation(s)
- Leonie Schoch
- Cardiovascular Program, Institut de Recerca, Hospital de la Santa Creu I Sant Pau, IIB Sant Pau, 08025 Barcelona, Spain; Faculty of Medicine, University of Barcelona (UB), 08036 Barcelona, Spain
| | - Sebastián Alcover
- Cardiovascular Program, Institut de Recerca, Hospital de la Santa Creu I Sant Pau, IIB Sant Pau, 08025 Barcelona, Spain
| | - Teresa Padró
- Cardiovascular Program, Institut de Recerca, Hospital de la Santa Creu I Sant Pau, IIB Sant Pau, 08025 Barcelona, Spain
| | | | - Guiomar Mendieta
- Cardiology Unit, Cardiovascular Clinical Institute, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Lina Badimon
- Cardiovascular Program, Institut de Recerca, Hospital de la Santa Creu I Sant Pau, IIB Sant Pau, 08025 Barcelona, Spain; Cardiovascular Research Chair, UAB, 08025 Barcelona, Spain; CiberCV, Institute of Health Carlos III, Madrid, Spain
| | - Gemma Vilahur
- Cardiovascular Program, Institut de Recerca, Hospital de la Santa Creu I Sant Pau, IIB Sant Pau, 08025 Barcelona, Spain; CiberCV, Institute of Health Carlos III, Madrid, Spain.
| |
Collapse
|
6
|
Koh S, Kim T, Kang D, Kim D. Effectiveness of Regular Aerobic Exercise in Improving Vascular Stiffness in Elderly Korean Women. J Clin Med 2023; 12:6119. [PMID: 37834763 PMCID: PMC10573532 DOI: 10.3390/jcm12196119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 09/16/2023] [Accepted: 09/20/2023] [Indexed: 10/15/2023] Open
Abstract
The purpose of this study was to determine the effects of aerobic exercise on carotid to femoral pulse wave velocity (cf-PWV), cell adhesion molecules (intracellular adhesion molecules (ICAM-1), vascular cell adhesion molecules (VCAM-1), endothelial selectin (E-selectin), and oxidized LDL in elderly women aged 70-85 years, and to identify the effect of and correlation with vascular stiffness. Forty participants were recruited and divided into three groups; vascular stiffness (VSG, n = 14), obesity (OG, n = 14), and normal (NG, n = 12). All groups were given a 16-week intervention of aerobic exercise, and the data collected before and after exercise were analyzed using SPSS Ver. 23.0. Two-way repeated-measures ANOVA was used to evaluate between-group and time-dependent interaction effects. One-way ANOVA was used to evaluate between-group variations. In addition, the significance was tested using a post hoc test (Scheffe). The within-group variations by time before and after exercise were examined using a paired t-test, and correlation analysis was performed using Pearson correlation coefficients. Simple regression analysis was performed for variables showing significant differences. The results indicate interaction effects for cf-PWV (p < 0.001), VCAM-1 (p < 0.01), E-selectin (p < 0.05), and oxidized LDL (p < 0.001). The rate of change of cf-PWV was positively correlated with that of VCAM-1 (r = 0.352, p < 0.05) and that of oxidized LDL (r = 0.325, p < 0.05) with statistical significance. To determine the effect of the rate of change of cf-PWV on the rate of change of VCAM-1, the variables were tested, and the coefficient of determination in the regression analysis was 0.124, indicating that 12.4% of the tested variables fit the standard regression line. The variables for the effect of the rate of change of cf-PWV on the rate of change of oxidized LDL were also tested, and the coefficient of determination in the regression analysis was 0.106, indicating that 10.6% of the tested variables fit the standard regression line. Thus, the 16-week regular and consistent aerobic exercise program had significant effects on the cf-PWV, ICAM-1, VCAM-1, E-selectin, and oxidized LDL in elderly Korean women with vascular stiffness, suggesting improvements in vascular stiffness, based on which the intervention is predicted to contribute to the prevention of vascular dysfunction by lowering the risk of cardiovascular disease due to atherosclerosis, as well as having a positive effect in the prevention of impairment of vascular endothelial cells.
Collapse
Affiliation(s)
| | | | | | - Doyeon Kim
- Department of Physical Education, Pusan National University, Busan 46241, Republic of Korea
| |
Collapse
|
7
|
Khan MA, Mohammad I, Banerjee S, Tomar A, Varughese KI, Mehta JL, Chandele A, Arockiasamy A. Oxidized LDL receptors: a recent update. Curr Opin Lipidol 2023:00041433-990000000-00037. [PMID: 37171285 DOI: 10.1097/mol.0000000000000884] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
PURPOSE OF REVIEW LDL in its oxidized form, or 'oxLDL', is now generally acknowledged to be highly proatherogenic and to play a significant role in atherosclerotic plaque formation. Therefore, there has been increasing interest in understanding the significance of oxLDL and its receptors in different phases of atherosclerosis, leading to the accumulation of additional data at the cellular, structural, and physiological levels. This review focuses on the most recent discoveries about these receptors and how they influence lipid absorption, metabolism, and inflammation in various cell types. RECENT FINDINGS Two crystal structures of lectin-like oxLDL receptor-1 (LOX-1), one with a small molecule inhibitor and the other with a monoclonal antibody have been published. We recently demonstrated that the 'surface site' of LOX1, adjacent to the positively charged 'basic spine region' that facilitates oxLDL binding, is a targetable site for drug development. Further, recent human studies showed that soluble LOX-1 holds potential as a biomarker for cardiovascular disease diagnosis, prognosis, and assessing the efficacy of therapy. SUMMARY Receptor-mediated oxLDL uptake results in cellular dysfunction of various cell types involved in atherogenesis and plaque development. The current advancements clearly demonstrate that targeting oxLDL-LOX-1 axis may lead to development of future therapeutics for the treatment of atherosclerotic cardiovascular and cerebrovascular diseases.
Collapse
Affiliation(s)
- Mohd Azeem Khan
- Membrane Protein Biology Group, International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi, India
| | - Irshad Mohammad
- Membrane Protein Biology Group, International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi, India
| | - Sohom Banerjee
- Membrane Protein Biology Group, International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi, India
| | - Akanksha Tomar
- Membrane Protein Biology Group, International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi, India
| | - Kottayil I Varughese
- Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences
| | - Jawahar L Mehta
- Division of Cardiology, University of Arkansas for Medical Sciences and the VA Medical Center, Little Rock, Arkansas, USA
| | - Anmol Chandele
- ICGEB-Emory Vaccine Centre, International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi, India
| | - Arulandu Arockiasamy
- Membrane Protein Biology Group, International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi, India
| |
Collapse
|
8
|
Aguilar VM, Paul A, Lazarko D, Levitan I. Paradigms of endothelial stiffening in cardiovascular disease and vascular aging. Front Physiol 2023; 13:1081119. [PMID: 36714307 PMCID: PMC9874005 DOI: 10.3389/fphys.2022.1081119] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 12/22/2022] [Indexed: 01/13/2023] Open
Abstract
Endothelial cells, the inner lining of the blood vessels, are well-known to play a critical role in vascular function, while endothelial dysfunction due to different cardiovascular risk factors or accumulation of disruptive mechanisms that arise with aging lead to cardiovascular disease. In this review, we focus on endothelial stiffness, a fundamental biomechanical property that reflects cell resistance to deformation. In the first part of the review, we describe the mechanisms that determine endothelial stiffness, including RhoA-dependent contractile response, actin architecture and crosslinking, as well as the contributions of the intermediate filaments, vimentin and lamin. Then, we review the factors that induce endothelial stiffening, with the emphasis on mechanical signals, such as fluid shear stress, stretch and stiffness of the extracellular matrix, which are well-known to control endothelial biomechanics. We also describe in detail the contribution of lipid factors, particularly oxidized lipids, that were also shown to be crucial in regulation of endothelial stiffness. Furthermore, we discuss the relative contributions of these two mechanisms of endothelial stiffening in vasculature in cardiovascular disease and aging. Finally, we present the current state of knowledge about the role of endothelial stiffening in the disruption of endothelial cell-cell junctions that are responsible for the maintenance of the endothelial barrier.
Collapse
Affiliation(s)
- Victor M. Aguilar
- Department of Medicine, Division of Pulmonary and Critical Care, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
- Richard and Loan Hill Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL, United States
| | - Amit Paul
- Department of Medicine, Division of Pulmonary and Critical Care, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Dana Lazarko
- Department of Medicine, Division of Pulmonary and Critical Care, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Irena Levitan
- Department of Medicine, Division of Pulmonary and Critical Care, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
- Richard and Loan Hill Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
9
|
Caveolin-1 is a primary determinant of endothelial stiffening associated with dyslipidemia, disturbed flow, and ageing. Sci Rep 2022; 12:17822. [PMID: 36280774 PMCID: PMC9592578 DOI: 10.1038/s41598-022-20713-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 09/16/2022] [Indexed: 01/20/2023] Open
Abstract
Endothelial stiffness is emerging as a major determinant in endothelial function. Here, we analyzed the role of caveolin-1 (Cav-1) in determining the stiffness of endothelial cells (EC) exposed to oxidized low density lipoprotein (oxLDL) under static and hemodynamic conditions in vitro and of aortic endothelium in vivo in mouse models of dyslipidemia and ageing. Elastic moduli of cultured ECs and of the endothelial monolayer of freshly isolated mouse aortas were measured using atomic force microscopy (AFM). We found that a loss of Cav-1 abrogates the uptake of oxLDL and oxLDL-induced endothelial stiffening, as well as endothelial stiffening induced by disturbed flow (DF), which was also oxLDL dependent. Mechanistically, Cav-1 is required for the expression of CD36 (cluster of differentiation 36) scavenger receptor. Genetic deletion of Cav-1 abrogated endothelial stiffening observed in the DF region of the aortic arch, and induced by a high fat diet (4-6 weeks) and significantly blunted endothelial stiffening that develops with advanced age. This effect was independent of stiffening of the sub-endothelium layer. Additionally, Cav-1 expression significantly increased with age. No differences in elastic modulus were observed between the sexes in advanced aged wild type and Cav-1 knockout mice. Taken together, this study demonstrates that Cav-1 plays a critical role in endothelial stiffening induced by oxLDL in vitro and by dyslipidemia, disturbed flow and ageing in vivo.
Collapse
|
10
|
Rezende L, Couto NFD, Fernandes-Braga W, Epshtein Y, Alvarez-Leite JI, Levitan I, Andrade LDO. OxLDL induces membrane structure rearrangement leading to biomechanics alteration and migration deficiency in macrophage. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2022; 1864:183951. [PMID: 35504320 DOI: 10.1016/j.bbamem.2022.183951] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 04/20/2022] [Accepted: 04/25/2022] [Indexed: 11/30/2022]
Abstract
Cholesterol sequestration from plasma membrane has been shown to induce lipid packing disruption, causing actin cytoskeleton reorganization and polymerization, increasing cell stiffness and inducing lysosomal exocytosis in non-professional phagocytes. Similarly, oxidized form of low-density lipoprotein (oxLDL) has also been shown to disrupt lipid organization and packing in endothelial cells, leading to biomechanics alterations that interfere with membrane injury and repair. For macrophages, much is known about oxLDL effects in cell activation, cytokine production and foam cell formation. However, little is known about its impact in the organization of macrophage membrane structured domains and cellular mechanics, the focus of the present study. Treatment of bone marrow-derived macrophages (BMDM) with oxLDL not only altered membrane structure, and potentially the distribution of raft domains, but also induced actin rearrangement, diffuse integrin distribution and cell shrinkage, similarly to observed upon treatment of these cells with MβCD. Those alterations led to decreased migration efficiency. For both treatments, higher co-localization of actin cytoskeleton and GM1 was observed, indicating a similar mechanism of action involving raft-like domain dynamics. Lastly, like MβCD treatment, oxLDL also induced lysosomal spreading in BMDM. We propose that OxLDL induced re-organization of membrane/cytoskeleton complex in macrophages can be attributed to the insertion of oxysterols into the membrane, which lead to changes in lipid organization and disruption of membrane structure, similar to the effect of cholesterol depletion by MβCD treatment. These results indicate that oxLDL can induce physical alterations in the complex membrane/cytoskeleton of macrophages, leading to significant biomechanical changes that compromise cell behavior.
Collapse
Affiliation(s)
- Luisa Rezende
- Department of Morphology/Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Natalia Fernanda Do Couto
- Department of Morphology/Federal University of Minas Gerais, Belo Horizonte, MG, Brazil; Department of Medicine, University of Illinois at Chicago, Chicago, USA
| | - Weslley Fernandes-Braga
- Department of Biochemistry and Immunology/Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Yulia Epshtein
- Department of Medicine, University of Illinois at Chicago, Chicago, USA
| | | | - Irena Levitan
- Department of Medicine, University of Illinois at Chicago, Chicago, USA
| | | |
Collapse
|
11
|
Achner L, Klersy T, Fels B, Reinberger T, Schmidt CX, Groß N, Hille S, Müller OJ, Aherrahrou Z, Kusche-Vihrog K, Raasch W. AFM-based nanoindentation indicates an impaired cortical stiffness in the AAV-PCSK9 DY atherosclerosis mouse model. Pflugers Arch 2022; 474:993-1002. [PMID: 35648220 PMCID: PMC9393126 DOI: 10.1007/s00424-022-02710-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 05/22/2022] [Indexed: 12/23/2022]
Abstract
Investigating atherosclerosis and endothelial dysfunction has mainly become established in genetically modified ApoE−/− or LDL-R−/− mice transgenic models. A new AAV-PCSK9DYDY mouse model with no genetic modification has now been reported as an alternative atherosclerosis model. Here, we aimed to employ this AAV-PCSK9DY mouse model to quantify the mechanical stiffness of the endothelial surface, an accepted hallmark for endothelial dysfunction and forerunner for atherosclerosis. Ten-week-old male C57BL/6 N mice were injected with AAV-PCSK9DY (0.5, 1 or 5 × 1011 VG) or saline as controls and fed with Western diet (1.25% cholesterol) for 3 months. Total cholesterol (TC) and triglycerides (TG) were measured after 6 and 12 weeks. Aortic sections were used for atomic force microscopy (AFM) measurements or histological analysis using Oil-Red-O staining. Mechanical properties of in situ endothelial cells derived from ex vivo aorta preparations were quantified using AFM-based nanoindentation. Compared to controls, an increase in plasma TC and TG and extent of atherosclerosis was demonstrated in all groups of mice in a viral load-dependent manner. Cortical stiffness of controls was 1.305 pN/nm and increased (10%) in response to viral load (≥ 0.5 × 1011 VG) and positively correlated with the aortic plaque content and plasma TC and TG. For the first time, we show changes in the mechanical properties of the endothelial surface and thus the development of endothelial dysfunction in the AAV-PCSK9DY mouse model. Our results demonstrate that this model is highly suitable and represents a good alternative to the commonly used transgenic mouse models for studying atherosclerosis and other vascular pathologies.
Collapse
Affiliation(s)
- Leonie Achner
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Tobias Klersy
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Benedikt Fels
- Institute for Physiology, University Lübeck, Lübeck, Germany
| | - Tobias Reinberger
- DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/, Lübeck, Germany.,Institute for Cardiogenetics, University Lübeck, Lübeck, Germany
| | - Cosima X Schmidt
- Institute of Neurobiology, University of Lübeck, Lübeck, Germany
| | - Natalie Groß
- Institute for Experimental Dermatology (LIED), University of Lübeck, Lübeck, Germany
| | - Susanne Hille
- DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/, Lübeck, Germany.,Department of Internal Medicine III, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Oliver J Müller
- DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/, Lübeck, Germany.,Department of Internal Medicine III, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Zouhair Aherrahrou
- DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/, Lübeck, Germany.,Institute for Cardiogenetics, University Lübeck, Lübeck, Germany
| | - Kristina Kusche-Vihrog
- Institute for Physiology, University Lübeck, Lübeck, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/, Lübeck, Germany
| | - Walter Raasch
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany. .,DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/, Lübeck, Germany. .,CBBM (Centre for Brain, Behavior and Metabolism), University of Lübeck, Lübeck, Germany.
| |
Collapse
|
12
|
Ahn SJ, Le Master E, Lee JC, Phillips SA, Levitan I, Fancher IS. Differential effects of obesity on visceral versus subcutaneous adipose arteries: role of shear-activated Kir2.1 and alterations to the glycocalyx. Am J Physiol Heart Circ Physiol 2022; 322:H156-H166. [PMID: 34890278 PMCID: PMC8742723 DOI: 10.1152/ajpheart.00399.2021] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 12/07/2021] [Accepted: 12/07/2021] [Indexed: 02/03/2023]
Abstract
Obesity imposes well-established deficits to endothelial function. We recently showed that obesity-induced endothelial dysfunction was mediated by disruption of the glycocalyx and a loss of Kir channel flow sensitivity. However, obesity-induced endothelial dysfunction is not observed in all vascular beds: visceral adipose arteries (VAAs), but not subcutaneous adipose arteries (SAAs), exhibit endothelial dysfunction. To determine whether differences in SAA versus VAA endothelial function observed in obesity are attributed to differential impairment of Kir channels and alterations to the glycocalyx, mice were fed a normal rodent diet, or a high-fat Western diet to induce obesity. Flow-induced vasodilation (FIV) was measured ex vivo. Functional downregulation of endothelial Kir2.1 was accomplished by transducing adipose arteries from mice and obese humans with adenovirus containing a dominant-negative Kir2.1 construct. Kir function was tested in freshly isolated endothelial cells seeded in a flow chamber for electrophysiological recordings under fluid shear. Atomic force microscopy was used to assess biophysical properties of the glycocalyx. Endothelial dysfunction was observed in VAAs of obese mice and humans. Downregulating Kir2.1 blunted FIV in SAAs, but had no effect on VAAs, from obese mice and humans. Obesity abolished Kir shear sensitivity in VAA endothelial cells and significantly altered the VAA glycocalyx. In contrast, Kir shear sensitivity was observed in SAA endothelial cells from obese mice and effects on SAA glycocalyx were less pronounced. We reveal distinct differences in Kir function and alterations to the glycocalyx that we propose contribute to the dichotomy in SAA versus VAA endothelial function with obesity.NEW & NOTEWORTHY We identified a role for endothelial Kir2.1 in the differences observed in VAA versus SAA endothelial function with obesity. The endothelial glycocalyx, a regulator of Kir activation by shear, is unequally perturbed in VAAs as compared with SAAs, which we propose results in a near complete loss of VAA endothelial Kir shear sensitivity and endothelial dysfunction. We propose that these differences underly the preserved endothelial function of SAA in obese mice and humans.
Collapse
Affiliation(s)
- Sang Joon Ahn
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Elizabeth Le Master
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - James C Lee
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, Illinois
| | - Shane A Phillips
- Department of Physical Therapy, College of Applied Health Sciences, University of Illinois at Chicago, Chicago, Illinois
| | - Irena Levitan
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Ibra S Fancher
- Department of Kinesiology and Applied Physiology, College of Health Sciences, University of Delaware, Newark, Delaware
| |
Collapse
|
13
|
Zeng Y, Du X, Yao X, Qiu Y, Jiang W, Shen J, Li L, Liu X. Mechanism of cell death of endothelial cells regulated by mechanical forces. J Biomech 2021; 131:110917. [PMID: 34952348 DOI: 10.1016/j.jbiomech.2021.110917] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 12/13/2021] [Accepted: 12/14/2021] [Indexed: 12/26/2022]
Abstract
Cell death of endothelial cells (ECs) is a common devastating consequence of various vascular-related diseases. Atherosclerosis, hypertension, sepsis, diabetes, cerebral ischemia and cardiac ischemia/reperfusion injury, and chronic kidney disease remain major causes of morbidity and mortality worldwide, in which ECs are constantly subjected to a great amount of dynamic changed mechanical forces including shear stress, extracellular matrix stiffness, mechanical stretch and microgravity. A thorough understanding of the regulatory mechanisms by which the mechanical forces controlled the cell deaths including apoptosis, autophagy, and pyroptosis is crucial for the development of new therapeutic strategies. In the present review, experimental and clinical data highlight that nutrient depletion, oxidative stress, tumor necrosis factor-α, high glucose, lipopolysaccharide, and homocysteine possess cytotoxic effects in many tissues and induce apoptosis of ECs, and that sphingosine-1-phosphate protects ECs. Nevertheless, EC apoptosis in the context of those artificial microenvironments could be enhanced, reduced or even reversed along with the alteration of patterns of shear stress. An appropriate level of autophagy diminishes EC apoptosis to some extent, in addition to supporting cell survival upon microenvironment challenges. The intervention of pyroptosis showed a profound effect on atherosclerosis. Further cell and animal studies are required to ascertain whether the alterations in the levels of cell deaths and their associated regulatory mechanisms happen at local lesion sites with considerable mechanical force changes, for preventing senescence and cell deaths in the vascular-related diseases.
Collapse
Affiliation(s)
- Ye Zeng
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China.
| | - Xiaoqiang Du
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xinghong Yao
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yan Qiu
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China
| | - Wenli Jiang
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China
| | - Junyi Shen
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China
| | - Liang Li
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xiaoheng Liu
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
14
|
Moreno-Cañadas R, Luque-Martín L, Arroyo AG. Intravascular Crawling of Patrolling Monocytes: A Lèvy-Like Motility for Unique Search Functions? Front Immunol 2021; 12:730835. [PMID: 34603307 PMCID: PMC8485030 DOI: 10.3389/fimmu.2021.730835] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 08/30/2021] [Indexed: 12/12/2022] Open
Abstract
Patrolling monocytes (PMo) are the organism’s preeminent intravascular guardians by their continuous search of damaged endothelial cells and harmful microparticles for their removal and to restore homeostasis. This surveillance is accomplished by PMo crawling on the apical side of the endothelium through regulated interactions of integrins and chemokine receptors with their endothelial ligands. We propose that the search mode governs the intravascular motility of PMo in vivo in a similar way to T cells looking for antigen in tissues. Signs of damage to the luminal side of the endothelium (local death, oxidized LDL, amyloid deposits, tumor cells, pathogens, abnormal red cells, etc.) will change the diffusive random towards a Lèvy-like crawling enhancing their recognition and clearance by PMo damage receptors as the integrin αMβ2 and CD36. This new perspective can help identify new actors to promote unique PMo intravascular actions aimed at maintaining endothelial fitness and combating harmful microparticles involved in diseases as lung metastasis, Alzheimer’s angiopathy, vaso-occlusive disorders, and sepsis.
Collapse
Affiliation(s)
- Rocío Moreno-Cañadas
- Molecular Biomedicine Department, Centro de Investigaciones Biológicas Margarita Salas (CIB-CSIC), Madrid, Spain
| | - Laura Luque-Martín
- Molecular Biomedicine Department, Centro de Investigaciones Biológicas Margarita Salas (CIB-CSIC), Madrid, Spain
| | - Alicia G Arroyo
- Molecular Biomedicine Department, Centro de Investigaciones Biológicas Margarita Salas (CIB-CSIC), Madrid, Spain
| |
Collapse
|
15
|
Li J, Fang Y, Wu D. Mechanical forces and metabolic changes cooperate to drive cellular memory and endothelial phenotypes. CURRENT TOPICS IN MEMBRANES 2021; 87:199-253. [PMID: 34696886 PMCID: PMC8639155 DOI: 10.1016/bs.ctm.2021.07.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Endothelial cells line the innermost layer of arterial, venous, and lymphatic vascular tree and accordingly are subject to hemodynamic, stretch, and stiffness mechanical forces. Normally quiescent, endothelial cells have a hemodynamic set point and become "activated" in response to disturbed hemodynamics, which may signal impending nutrient or gas depletion. Endothelial cells in the majority of tissue beds are normally inactivated and maintain vessel barrier functions, are anti-inflammatory, anti-coagulant, and anti-thrombotic. However, under aberrant mechanical forces, endothelial signaling transforms in response, resulting cellular changes that herald pathological diseases. Endothelial cell metabolism is now recognized as the primary intermediate pathway that undergirds cellular transformation. In this review, we discuss the various mechanical forces endothelial cells sense in the large vessels, microvasculature, and lymphatics, and how changes in environmental mechanical forces result in changes in metabolism, which ultimately influence cell physiology, cellular memory, and ultimately disease initiation and progression.
Collapse
Affiliation(s)
- Jin Li
- Committee on Molecular Metabolism and Nutrition, Biological Sciences Division, University of Chicago, Chicago, IL, United States; Department of Medicine, Biological Sciences Division, University of Chicago, Chicago, IL, United States
| | - Yun Fang
- Committee on Molecular Metabolism and Nutrition, Biological Sciences Division, University of Chicago, Chicago, IL, United States; Department of Medicine, Biological Sciences Division, University of Chicago, Chicago, IL, United States
| | - David Wu
- Committee on Molecular Metabolism and Nutrition, Biological Sciences Division, University of Chicago, Chicago, IL, United States; Department of Medicine, Biological Sciences Division, University of Chicago, Chicago, IL, United States.
| |
Collapse
|
16
|
Noels H, Lehrke M, Vanholder R, Jankowski J. Lipoproteins and fatty acids in chronic kidney disease: molecular and metabolic alterations. Nat Rev Nephrol 2021; 17:528-542. [PMID: 33972752 DOI: 10.1038/s41581-021-00423-5] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/22/2021] [Indexed: 02/06/2023]
Abstract
Chronic kidney disease (CKD) induces modifications in lipid and lipoprotein metabolism and homeostasis. These modifications can promote, modulate and/or accelerate CKD and secondary cardiovascular disease (CVD). Lipid and lipoprotein abnormalities - involving triglyceride-rich lipoproteins, LDL and/or HDL - not only involve changes in concentration but also changes in molecular structure, including protein composition, incorporation of small molecules and post-translational modifications. These alterations modify the function of lipoproteins and can trigger pro-inflammatory and pro-atherogenic processes, as well as oxidative stress. Serum fatty acid levels are also often altered in patients with CKD and lead to changes in fatty acid metabolism - a key process in intracellular energy production - that induce mitochondrial dysfunction and cellular damage. These fatty acid changes might not only have a negative impact on the heart, but also contribute to the progression of kidney damage. The presence of these lipoprotein alterations within a biological environment characterized by increased inflammation and oxidative stress, as well as the competing risk of non-atherosclerotic cardiovascular death as kidney function declines, has important therapeutic implications. Additional research is needed to clarify the pathophysiological link between lipid and lipoprotein modifications, and kidney dysfunction, as well as the genesis and/or progression of CVD in patients with kidney disease.
Collapse
Affiliation(s)
- Heidi Noels
- Institute for Molecular Cardiovascular Research, RWTH Aachen University, University Hospital, Aachen, Germany
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, Netherlands
| | - Michael Lehrke
- Department of Internal Medicine I, RWTH Aachen University, University Hospital, Aachen, Germany
| | - Raymond Vanholder
- Nephrology Section, Department of Internal Medicine and Pediatrics, University Hospital, Ghent, Belgium
| | - Joachim Jankowski
- Institute for Molecular Cardiovascular Research, RWTH Aachen University, University Hospital, Aachen, Germany.
- Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre, Maastricht University, Maastricht, Netherlands.
| |
Collapse
|
17
|
Fatty acids and evolving roles of their proteins in neurological, cardiovascular disorders and cancers. Prog Lipid Res 2021; 83:101116. [PMID: 34293403 DOI: 10.1016/j.plipres.2021.101116] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 07/04/2021] [Accepted: 07/14/2021] [Indexed: 01/03/2023]
Abstract
The dysregulation of fat metabolism is involved in various disorders, including neurodegenerative, cardiovascular, and cancers. The uptake of long-chain fatty acids (LCFAs) with 14 or more carbons plays a pivotal role in cellular metabolic homeostasis. Therefore, the uptake and metabolism of LCFAs must constantly be in tune with the cellular, metabolic, and structural requirements of cells. Many metabolic diseases are thought to be driven by the abnormal flow of fatty acids either from the dietary origin and/or released from adipose stores. Cellular uptake and intracellular trafficking of fatty acids are facilitated ubiquitously with unique combinations of fatty acid transport proteins and cytoplasmic fatty acid-binding proteins in every tissue. Extensive data are emerging on the defective transporters and metabolism of LCFAs and their clinical implications. Uptake and metabolism of LCFAs are crucial for the brain's functional development and cardiovascular health and maintenance. In addition, data suggest fatty acid metabolic transporter can normalize activated inflammatory response by reprogramming lipid metabolism in cancers. Here we review the current understanding of how LCFAs and their proteins contribute to the pathophysiology of three crucial diseases and the mechanisms involved in the processes.
Collapse
|
18
|
Li F, Yan K, Wu L, Zheng Z, Du Y, Liu Z, Zhao L, Li W, Sheng Y, Ren L, Tang C, Zhu L. Single-cell RNA-seq reveals cellular heterogeneity of mouse carotid artery under disturbed flow. Cell Death Discov 2021; 7:180. [PMID: 34282126 PMCID: PMC8290019 DOI: 10.1038/s41420-021-00567-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/26/2021] [Accepted: 05/13/2021] [Indexed: 12/28/2022] Open
Abstract
Disturbed blood flow (d-flow) has been known to induce changes of the cells in the arterial wall, increasing the risk of atherosclerosis. However, the heterogeneity of the vascular cell populations under d-flow remains less understood. To generate d-flow in vivo, partial carotid artery ligation (PCL) was performed. Seven days after ligation, single-cell RNA sequencing of nine left carotid arteries (LCA) from the PCL group (10,262 cells) or control group (14,580 cells) was applied and a single-cell atlas of gene expression was constructed. The integrated analysis identified 15 distinct carotid cell clusters, including 10 d-flow-relevant subpopulations. Among endothelial cells, at least four subpopulations were identified, including Klk8hi ECs, Lrp1hi ECs, Dkk2hi ECs, and Cd36hi ECs. Analysis of GSVA and single-cell trajectories indicated that the previously undescribed Dkk2hi ECs subpopulation was mechanosensitive and potentially transformed from Klk8hi ECs under d-flow. D-flow-induced Spp1hi VSMCs subpopulation that appeared to be endowed with osteoblast differentiation, suggesting a role in arterial stiffness. Among the infiltrating cell subpopulations, Trem2hi Mφ, Birc5hi Mφ, DCs, CD4+ T cells, CXCR6+ T cells, NK cells, and granulocytes were identified under d-flow. Of note, the novel Birc5hi Mφ was identified as a potential contributor to the accumulation of macrophages in atherosclerosis. Finally, Dkk2hi ECs, and Cd36hi ECs were also found in the proatherosclerotic area of the aorta where the d-flow occurs. In conclusion, we presented a comprehensive single-cell atlas of all cells in the carotid artery under d-flow, identified previously unrecognized cell subpopulations and their gene expression signatures, and suggested their specialized functions.
Collapse
Affiliation(s)
- Fengchan Li
- Cyrus Tang Hematology Center, Suzhou, Jiangsu, China
| | - Kunmin Yan
- Cyrus Tang Hematology Center, Suzhou, Jiangsu, China
| | - Lili Wu
- Cyrus Tang Hematology Center, Suzhou, Jiangsu, China
| | - Zhong Zheng
- Cyrus Tang Hematology Center, Suzhou, Jiangsu, China
| | - Yun Du
- Cyrus Tang Hematology Center, Suzhou, Jiangsu, China
| | - Ziting Liu
- Cyrus Tang Hematology Center, Suzhou, Jiangsu, China
| | - Luyao Zhao
- Cyrus Tang Hematology Center, Suzhou, Jiangsu, China
| | - Wei Li
- Cyrus Tang Hematology Center, Suzhou, Jiangsu, China
| | - Yulan Sheng
- Cyrus Tang Hematology Center, Suzhou, Jiangsu, China
| | - Lijie Ren
- Cyrus Tang Hematology Center, Suzhou, Jiangsu, China
| | - Chaojun Tang
- Cyrus Tang Hematology Center, Suzhou, Jiangsu, China.
- Collaborative Innovation Center of Hematology, Suzhou, Jiangsu, China.
- Suzhou Key Laboratory of Thrombosis and Vascular Diseases, Suzhou, Jiangsu, China.
- National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.
| | - Li Zhu
- Cyrus Tang Hematology Center, Suzhou, Jiangsu, China.
- Collaborative Innovation Center of Hematology, Suzhou, Jiangsu, China.
- Suzhou Key Laboratory of Thrombosis and Vascular Diseases, Suzhou, Jiangsu, China.
- National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.
- State Key Laboratory of Radiation Medicine and Protection Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
19
|
Zhang X, Fan J, Li H, Chen C, Wang Y. CD36 Signaling in Diabetic Cardiomyopathy. Aging Dis 2021; 12:826-840. [PMID: 34094645 PMCID: PMC8139204 DOI: 10.14336/ad.2020.1217] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 12/17/2020] [Indexed: 12/14/2022] Open
Abstract
Cluster of differentiation 36 (CD36), also referred to as scavenger receptor B2, has been shown to serve multiple functions in lipid metabolism, inflammatory signaling, oxidative stress, and energy reprogramming. As a scavenger receptor, CD36 interacts with various ligands, such as oxidized low-density lipoprotein (oxLDL), thrombospondin 1 (TSP-1), and fatty acid (FA), thereby activating specific downstream signaling pathways. Cardiac CD36 is mostly expressed on the surface of cardiomyocytes and endothelial cells. The pathophysiological process of diabetic cardiomyopathy (DCM) encompasses diverse metabolic abnormalities, such as enhanced transfer of cardiac myocyte sarcolemmal FA, increased levels of advanced glycation end-products, elevation in oxidative stress, impaired insulin signaling cascade, disturbance in calcium handling, and microvascular rarefaction which are closely related to CD36 signaling. This review presents a summary of the CD36 signaling pathway that acts mainly as a long-chain FA transporter in cardiac myocytes and functions as a receptor to bind to numerous ligands in endothelial cells. Finally, we summarize the recent basic research and clinical findings regarding CD36 signaling in DCM, suggesting a promising strategy to treat this condition.
Collapse
Affiliation(s)
- Xudong Zhang
- Division of Cardiology, Tongji Hospital, Tongji Medical College and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| | - Jiahui Fan
- Division of Cardiology, Tongji Hospital, Tongji Medical College and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| | - Huaping Li
- Division of Cardiology, Tongji Hospital, Tongji Medical College and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| | - Chen Chen
- Division of Cardiology, Tongji Hospital, Tongji Medical College and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Wang
- Division of Cardiology, Tongji Hospital, Tongji Medical College and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
20
|
Mahmoud M, Mayer M, Cancel LM, Bartosch AM, Mathews R, Tarbell JM. The glycocalyx core protein Glypican 1 protects vessel wall endothelial cells from stiffness-mediated dysfunction and disease. Cardiovasc Res 2021; 117:1592-1605. [PMID: 32647868 PMCID: PMC8152694 DOI: 10.1093/cvr/cvaa201] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 05/22/2020] [Accepted: 07/02/2020] [Indexed: 12/25/2022] Open
Abstract
AIMS Arterial stiffness is an underlying risk factor and a hallmark of cardiovascular diseases. The endothelial cell (EC) glycocalyx is a glycan rich surface layer that plays a key role in protecting against EC dysfunction and vascular disease. However, the mechanisms by which arterial stiffness promotes EC dysfunction and vascular disease are not fully understood, and whether the mechanism involves the protective endothelial glycocalyx is yet to be determined. We hypothesized that endothelial glycocalyx protects the endothelial cells lining the vascular wall from dysfunction and disease in response to arterial stiffness. METHODS AND RESULTS Cells cultured on polyacrylamide (PA) gels of substrate stiffness 10 kPa (mimicking the subendothelial stiffness of aged, unhealthy arteries) showed a significant inhibition of glycocalyx expression compared to cells cultured on softer PA gels (2.5 kPa, mimicking the subendothelial stiffness of young, healthy arteries). Specifically, gene and protein analyses revealed that a glycocalyx core protein Glypican 1 was inhibited in cells cultured on stiff PA gels. These cells had enhanced endothelial cell dysfunction as determined by enhanced cell inflammation (enhanced inflammatory gene expression, monocyte adhesion, and inhibited nitric oxide expression), proliferation, and EndMT. Removal of Glypican 1 using gene-specific silencing with siRNA or gene overexpression using a plasmid revealed that Glypican 1 is required to protect against stiffness-mediated endothelial cell dysfunction. Consistent with this, using a model of age-mediated stiffness, older mice exhibited a reduced expression of Glypican 1 and enhanced endothelial cell dysfunction compared to young mice. Glypican 1 gene deletion in knockout mice (GPC1-/-) exacerbated endothelial dysfunction in young mice, which normally had high endothelial expression, but not in old mice that normally expressed low levels. Endothelial cell dysfunction was exacerbated in young, but not aged, Glypican 1 knockout mice (GPC1-/-). CONCLUSION Arterial stiffness promotes EC dysfunction and vascular disease at least partly through the suppression of the glycocalyx protein Glypican 1. Glypican 1 contributes to the protection against endothelial cell dysfunction and vascular disease in endothelial cells.
Collapse
Affiliation(s)
- Marwa Mahmoud
- Department of Biomedical Engineering, The City College of New York, New York, NY, USA
| | - Mariya Mayer
- Department of Biomedical Engineering, The City College of New York, New York, NY, USA
| | - Limary M Cancel
- Department of Biomedical Engineering, The City College of New York, New York, NY, USA
| | - Anne Marie Bartosch
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Rick Mathews
- Oregon Health & Science University, School of Medicine, Portland, OR, USA
| | - John M Tarbell
- Department of Biomedical Engineering, The City College of New York, New York, NY, USA
| |
Collapse
|
21
|
Abstract
Apocynin is a naturally occurring acetophenone, found in the roots of Apocynum cannabinum and Picrorhiza kurroa. Various chemical and pharmaceutical modifications have been carried out to enhance the absorption and duration of action of apocynin, like, formulation of chitosan-based apocynin-loaded solid lipid nanoparticles, chitosan-oligosaccharide based nanoparticles, and biodegradable polyanhydride nanoparticles. Apocynin has been subjected to a wide range of experimental screening and has proved to be useful for amelioration of a variety of disorders, like diabetic complications, neurodegeneration, cardiovascular disorders, lung cancer, hepatocellular cancer, pancreatic cancer, and pheochromocytoma. Apocynin has been primarily reported as an NADPH oxidase (NOX) inhibitor and prevents translocation of its p47phox subunit to the plasma membrane, observed in neurodegeneration and hypertension. However, recent studies highlight its off-target effects that it is able to function as a scavenger of non-radical oxidant species, which is relevant for its activity against NOX 4 mediated production of hydrogen peroxide. Additionally, apocynin has shown inhibition of eNOS-dependent superoxide production in diabetic cardiomyopathy, reduction of NLRP3 activation and TGFβ/Smad signaling in diabetic nephropathy, diminished VEGF expression and decreased retinal NF-κB activation in diabetic retinopathy, inhibition of P38/MAPK/Caspase3 pathway in pheochromocytoma, inhibition of AKT-GSK3β and ERK1/2 pathways in pancreatic cancer, and decreased FAK/PI3K/Akt signaling in hepatocellular cancer. This review aims to discuss the pharmacokinetics and mechanisms of the pharmacological actions of apocynin.
Collapse
Affiliation(s)
- Shreya R Savla
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS, Mumbai, India
| | - Ankit P Laddha
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS, Mumbai, India
| | - Yogesh A Kulkarni
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS, Mumbai, India
| |
Collapse
|
22
|
Çakıcı M, Yeşil Dirisu G, Karaaziz M, Buran A, Çakıcı E. Psychoactive substance abuse: prevalence, risk factors and relation to acculturation in adults living in North Cyprus, 2003–2018. CURRENT PSYCHOLOGY 2021. [DOI: 10.1007/s12144-020-01225-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
23
|
Le Master E, Ahn SJ, Levitan I. Mechanisms of endothelial stiffening in dyslipidemia and aging: Oxidized lipids and shear stress. CURRENT TOPICS IN MEMBRANES 2020; 86:185-215. [PMID: 33837693 PMCID: PMC8168803 DOI: 10.1016/bs.ctm.2020.08.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Vascular stiffening of the arterial walls is well-known as a key factor in aging and the development of cardiovascular disease; however, the role of endothelial stiffness in vascular dysfunction is still an emerging topic. In this review, the authors discuss the impact of dyslipidemia, oxidized lipids, substrate stiffness, age and pro-atherogenic disturbed flow have on endothelial stiffness. Furthermore, we investigate several mechanistic pathways that are key contributors in endothelial stiffness and discuss their physiological effects in the onset of atherogenesis in the disturbed flow regions of the aortic vasculature. The findings in this chapter describe a novel paradigm of synergistic interaction of plasma dyslipidemia/oxidized lipids and pro-atherogenic disturbed shear stress, as well as aging has on endothelial stiffness and vascular dysfunction.
Collapse
Affiliation(s)
- Elizabeth Le Master
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Sang Joon Ahn
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Irena Levitan
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of Illinois at Chicago, Chicago, IL, United States.
| |
Collapse
|
24
|
Abstract
PURPOSE OF REVIEW Atherosclerosis is a chronic disease characterized by lipid retention and inflammation in the artery wall. The retention and oxidation of low-density lipoprotein (LDL) in sub-endothelial space play a critical role in atherosclerotic plaque formation and destabilization. Oxidized LDL (ox-LDL) and other modified LDL particles are avidly taken up by endothelial cells, smooth muscle cells, and macrophages mainly through several scavenger receptors, including CD36 which is a class B scavenger receptor and membrane glycoprotein. RECENT FINDINGS Animal studies performed on CD36-deficient mice suggest that deficiency of CD36 prevents the development of atherosclerosis, though with some debate. CD36 serves as a signaling hub protein at the crossroad of inflammation, lipid metabolism, and fatty acid metabolism. In addition, the level of soluble CD36 (unattached to cells) in the circulating blood was elevated in patients with atherosclerosis and other metabolic disorders. We performed a state-of-the-art review on the structure, ligands, functions, and regulation of CD36 in the context of atherosclerosis by focusing on the pathological role of CD36 in the dysfunction of endothelial cells, smooth muscle cells, monocytes/macrophages, and platelets. Finally, we highlight therapeutic possibilities to target CD36 expression/activity in atherosclerosis.
Collapse
|
25
|
Fancher IS, Le Master E, Ahn SJ, Adamos C, Lee JC, Berdyshev E, Dull RO, Phillips SA, Levitan I. Impairment of Flow-Sensitive Inwardly Rectifying K + Channels via Disruption of Glycocalyx Mediates Obesity-Induced Endothelial Dysfunction. Arterioscler Thromb Vasc Biol 2020; 40:e240-e255. [PMID: 32698687 DOI: 10.1161/atvbaha.120.314935] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVE To determine if endothelial dysfunction in a mouse model of diet-induced obesity and in obese humans is mediated by the suppression of endothelial Kir (inwardly rectifying K+) channels. Approach and Results: Endothelial dysfunction, observed as reduced dilations to flow, occurred after feeding mice a high-fat, Western diet for 8 weeks. The functional downregulation of endothelial Kir2.1 using dominant-negative Kir2.1 construct resulted in substantial reductions in the response to flow in mesenteric arteries of lean mice, whereas no effect was observed in arteries of obese mice. Overexpressing wild-type-Kir2.1 in endothelium of arteries from obese mice resulted in full recovery of the flow response. Exposing freshly isolated endothelial cells to fluid shear during patch-clamp electrophysiology revealed that the flow-sensitivity of Kir was virtually abolished in cells from obese mice. Atomic force microscopy revealed that the endothelial glycocalyx was stiffer and the thickness of the glycocalyx layer reduced in arteries from obese mice. We also identified that the length of the glycocalyx is critical to the flow-activation of Kir. Overexpressing Kir2.1 in endothelium of arteries from obese mice restored flow- and heparanase-sensitivity, indicating an important role for heparan sulfates in the flow-activation of Kir. Furthermore, the Kir2.1-dependent component of flow-induced vasodilation was lost in the endothelium of resistance arteries of obese humans obtained from biopsies collected during bariatric surgery. CONCLUSIONS We conclude that obesity-induced impairment of flow-induced vasodilation is attributed to the loss of flow-sensitivity of endothelial Kir channels and propose that the latter is mediated by the biophysical alterations of the glycocalyx.
Collapse
Affiliation(s)
- Ibra S Fancher
- From the Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine (I.S.F., E.L.M., S.J.A., C.A., I.L.), University of Illinois at Chicago
| | - Elizabeth Le Master
- From the Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine (I.S.F., E.L.M., S.J.A., C.A., I.L.), University of Illinois at Chicago
| | - Sang Joon Ahn
- From the Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine (I.S.F., E.L.M., S.J.A., C.A., I.L.), University of Illinois at Chicago
| | - Crystal Adamos
- From the Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine (I.S.F., E.L.M., S.J.A., C.A., I.L.), University of Illinois at Chicago
| | - James C Lee
- Departement of Bioengineering (J.C.L.), University of Illinois at Chicago
| | - Evgeny Berdyshev
- Division of Pulmonary, Critical Care and Sleep Medicine, Departement of Medicine, National Jewish Health, Denver, CO (E.B.)
| | - Randal O Dull
- Department of Anesthesiology, University of Arizona College of Medicine, Banner-University Medical Center, Tucson (R.O.D.)
| | - Shane A Phillips
- Department of Physical Therapy (S.A.P.), University of Illinois at Chicago
| | - Irena Levitan
- From the Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine (I.S.F., E.L.M., S.J.A., C.A., I.L.), University of Illinois at Chicago
| |
Collapse
|
26
|
Shao Y, Saredy J, Yang WY, Sun Y, Lu Y, Saaoud F, Drummer C, Johnson C, Xu K, Jiang X, Wang H, Yang X. Vascular Endothelial Cells and Innate Immunity. Arterioscler Thromb Vasc Biol 2020; 40:e138-e152. [PMID: 32459541 PMCID: PMC7263359 DOI: 10.1161/atvbaha.120.314330] [Citation(s) in RCA: 179] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In addition to the roles of endothelial cells (ECs) in physiological processes, ECs actively participate in both innate and adaptive immune responses. We previously reported that, in comparison to macrophages, a prototypic innate immune cell type, ECs have many innate immune functions that macrophages carry out, including cytokine secretion, phagocytic function, antigen presentation, pathogen-associated molecular patterns-, and danger-associated molecular patterns-sensing, proinflammatory, immune-enhancing, anti-inflammatory, immunosuppression, migration, heterogeneity, and plasticity. In this highlight, we introduce recent advances published in both ATVB and many other journals: (1) several significant characters classify ECs as novel immune cells not only in infections and allograft transplantation but also in metabolic diseases; (2) several new receptor systems including conditional danger-associated molecular pattern receptors, nonpattern receptors, and homeostasis associated molecular patterns receptors contribute to innate immune functions of ECs; (3) immunometabolism and innate immune memory determine the innate immune functions of ECs; (4) a great induction of the immune checkpoint receptors in ECs during inflammations suggests the immune tolerogenic functions of ECs; and (5) association of immune checkpoint inhibitors with cardiovascular adverse events and cardio-oncology indicates the potential contributions of ECs as innate immune cells.
Collapse
Affiliation(s)
- Ying Shao
- Centers of Inflammation, Translational & Clinical Lung Research, Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140
| | - Jason Saredy
- Metabolic Disease Research, Cardiovascular Research, Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140
| | - William Y. Yang
- Metabolic Disease Research, Cardiovascular Research, Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140
| | - Yu Sun
- Centers of Inflammation, Translational & Clinical Lung Research, Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140
| | - Yifan Lu
- Centers of Inflammation, Translational & Clinical Lung Research, Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140
| | - Fatma Saaoud
- Centers of Inflammation, Translational & Clinical Lung Research, Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140
| | - Charles Drummer
- Centers of Inflammation, Translational & Clinical Lung Research, Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140
| | - Candice Johnson
- Centers of Inflammation, Translational & Clinical Lung Research, Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140
| | - Keman Xu
- Centers of Inflammation, Translational & Clinical Lung Research, Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140
| | - Xiaohua Jiang
- Centers of Inflammation, Translational & Clinical Lung Research, Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140
- Metabolic Disease Research, Cardiovascular Research, Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140
| | - Hong Wang
- Metabolic Disease Research, Cardiovascular Research, Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140
| | - Xiaofeng Yang
- Centers of Inflammation, Translational & Clinical Lung Research, Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140
- Metabolic Disease Research, Cardiovascular Research, Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140
| |
Collapse
|
27
|
Le Master E, Levitan I. Endothelial stiffening in dyslipidemia. Aging (Albany NY) 2020; 11:299-300. [PMID: 30674709 PMCID: PMC6366977 DOI: 10.18632/aging.101778] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 01/15/2019] [Indexed: 12/12/2022]
Affiliation(s)
- Elizabeth Le Master
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Irena Levitan
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA
| |
Collapse
|
28
|
Bogachkov YY, Chen L, Le Master E, Fancher IS, Zhao Y, Aguilar V, Oh MJ, Wary KK, DiPietro LA, Levitan I. LDL induces cholesterol loading and inhibits endothelial proliferation and angiogenesis in Matrigels: correlation with impaired angiogenesis during wound healing. Am J Physiol Cell Physiol 2020; 318:C762-C776. [PMID: 31995410 DOI: 10.1152/ajpcell.00495.2018] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Hypercholesterolemia is a major risk factor for adverse cardiovascular outcomes, but its effect on angiogenesis and wound healing is not well understood. In this study, using a combination of mass spectrometry and laurdan two-photon imaging, we show that elevated levels of low-density lipoprotein (LDL), like those seen in hypercholesterolemic patients, lead to an increase in both free cholesterol and cholesterol esters, as well as increase in lipid order of endothelial cell membranes. Notably, these effects are distinct and opposite to the lack of cholesterol loading and the disruption of lipid order observed in our earlier studies in response to oxidized LDL (oxLDL). The same pathological level of LDL leads to a significant inhibition of endothelial proliferation and cell cycle arrest in G2/M phase, whereas oxLDL enhances endothelial proliferation in S phase of the cycle. LDL but not oxLDL suppresses the expression of vascular endothelial growth factor receptor-2 while enhancing the expression of vascular endothelial growth factor (VEGF). Furthermore, we show that aged (8-10 mo) hypercholesterolemic apolipoprotein E-deficient (ApoE-/-) mice display delayed wound closure compared with age-matched C57/BL6 wild-type controls following a skin punch biopsy. The delay in wound healing is associated with a decreased expression of cluster of differentiation 31 platelet endothelial cell adhesion molecule endothelial marker and decreased angiogenesis within the wound bed. Furthermore, decreased endothelial responsiveness to the growth factors VEGF and basic fibroblast growth factor is observed in ApoE-/- mice in Matrigel plugs and in Matrigels with high levels of LDL in wild-type mice. We propose that plasma hypercholesterolemia is antiangiogenic due to elevated levels of LDL.
Collapse
Affiliation(s)
- Yedida Y Bogachkov
- Division of Pulmonary and Critical Care, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois.,Department of Cellular and Molecular Pharmacology, University of Illinois at Chicago, Chicago, Illinois
| | - Lin Chen
- Center for Wound Healing and Tissue Regeneration, College of Dentistry, University of Illinois at Chicago, Chicago, Illinois
| | - Elizabeth Le Master
- Division of Pulmonary and Critical Care, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Ibra S Fancher
- Division of Pulmonary and Critical Care, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Yan Zhao
- Center for Wound Healing and Tissue Regeneration, College of Dentistry, University of Illinois at Chicago, Chicago, Illinois
| | - Victor Aguilar
- Division of Pulmonary and Critical Care, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Myung-Jin Oh
- Division of Pulmonary and Critical Care, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Kishore K Wary
- Department of Cellular and Molecular Pharmacology, University of Illinois at Chicago, Chicago, Illinois
| | - Luisa A DiPietro
- Center for Wound Healing and Tissue Regeneration, College of Dentistry, University of Illinois at Chicago, Chicago, Illinois
| | - Irena Levitan
- Division of Pulmonary and Critical Care, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois.,Department of Cellular and Molecular Pharmacology, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
29
|
Lu HS, Schmidt AM, Hegele RA, Mackman N, Rader DJ, Weber C, Daugherty A. Annual Report on Sex in Preclinical Studies: Arteriosclerosis, Thrombosis, and Vascular Biology Publications in 2018. Arterioscler Thromb Vasc Biol 2019; 40:e1-e9. [PMID: 31869272 DOI: 10.1161/atvbaha.119.313556] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Hong S Lu
- From the Saha Cardiovascular Research Center and Department of Physiology, University of Kentucky, Lexington (H.S.L., A.D.)
| | - Ann Marie Schmidt
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University Langone Medical Center, New York, NY (A.M.S.)
| | - Robert A Hegele
- Department of Medicine and Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada (R.A.H.)
| | - Nigel Mackman
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC (N.M.)
| | - Daniel J Rader
- Departments of Medicine and Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia (D.J.R.)
| | - Christian Weber
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität (LMU) and German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany (C.W.)
| | - Alan Daugherty
- From the Saha Cardiovascular Research Center and Department of Physiology, University of Kentucky, Lexington (H.S.L., A.D.)
| |
Collapse
|
30
|
Li X, Zhang T, Geng J, Wu Z, Xu L, Liu J, Tian J, Zhou Z, Nie J, Bai X. Advanced Oxidation Protein Products Promote Lipotoxicity and Tubulointerstitial Fibrosis via CD36/β-Catenin Pathway in Diabetic Nephropathy. Antioxid Redox Signal 2019; 31:521-538. [PMID: 31084358 DOI: 10.1089/ars.2018.7634] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Aims: Diabetic nephropathy (DN) is the principal cause of mortality and morbidity in diabetic patients, the progression of which correlates best with tubulointerstitial fibrosis (TIF). Advanced oxidation protein products (AOPPs) have been detected in patients with chronic renal failure, causing injuries to proximal tubular epithelial cells. CD36, a known receptor for AOPP, is an important modulator of lipid homeostasis, predisposing to renal tubular damage. However, whether AOPPs induce lipotoxicity via the CD36 receptor pathway remains unknown. Herein, we tested the hypothesis that AOPPs accumulation in diabetes incurs lipotoxicity, causing renal TIF via the CD36 signaling pathway. Results: In DN patients and diabetic mice in vivo, AOPPs overload induces lipogenesis (upregulation of CD36 and sterol regulatory element-binding protein 1), fibrosis (upregulation of Fibronectin), and renal function decline (increased serum creatinine and N-acetyl-β-d-glucosaminidase, decreased estimated glomerular filtration rate). In HK-2 cells in vitro, high glucose stimulated AOPPs-induced lipotoxicity, apoptosis, and fibrosis via the CD36 receptor pathway. In addition, apocynin abrogated AOPPs-induced lipid accumulation and CD36 inhibition significantly mitigated AOPPs-induced mitochondrial injuries, lipotoxicity, and renal fibrosis. Further, we provide mechanistic evidence that AOPPs overload induces the enrichment of β-catenin binding the CD36 promoter region. Innovation and Conclusion: Our data reveal a major role of AOPPs in triggering lipotoxicity and fibrosis via CD36-dependent Wnt/β-catenin activation, providing new evidence for understanding the role of lipid accumulation in DN.
Collapse
Affiliation(s)
- Xiao Li
- 1Department of Emergency, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Ting Zhang
- 2Division of Nephrology, State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Guangdong Provincial Institute of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Jian Geng
- 3Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Zhuguo Wu
- 4Department of Internal Medicine, the Second Clinical Medical College, Guangdong Medical University, Dongguan, Guangdong, People's Republic of China
| | - Liting Xu
- 2Division of Nephrology, State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Guangdong Provincial Institute of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Jixing Liu
- 2Division of Nephrology, State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Guangdong Provincial Institute of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Jianwei Tian
- 2Division of Nephrology, State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Guangdong Provincial Institute of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Zhanmei Zhou
- 2Division of Nephrology, State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Guangdong Provincial Institute of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Jing Nie
- 2Division of Nephrology, State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Guangdong Provincial Institute of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Xiaoyan Bai
- 2Division of Nephrology, State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Guangdong Provincial Institute of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| |
Collapse
|
31
|
Schnitzler JG, Dallinga-Thie GM, Kroon J. The Role of (Modified) Lipoproteins in Vascular Function: A Duet Between Monocytes and the Endothelium. Curr Med Chem 2019; 26:1594-1609. [PMID: 29546830 DOI: 10.2174/0929867325666180316121015] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 12/05/2017] [Accepted: 12/06/2017] [Indexed: 12/24/2022]
Abstract
Over the last century, many studies have demonstrated that low-density lipoprotein (LDL) is a key risk factor of cardiovascular diseases (CVD) related to atherosclerosis. Thus, for these CVD patients, LDL lowering agents are commonly used in the clinic to reduce the risk for CVD. LDL, upon modification, will develop distinct inflammatory and proatherogenic potential, leading to impaired endothelial integrity, influx of immune cells and subsequent increased foam cell formation. LDL can also directly affect peripheral monocyte composition, rendering them in a more favorable position to migrate and accumulate in the subendothelial space. It has become apparent that other lipoprotein particles, such as triglyceride- rich lipoproteins or remnants (TRL) and lipoprotein(a) [Lp(a)] may also impact on atherogenic pathways. Evidence is accumulating that Lp(a) can promote peripheral monocyte activation, eventually leading to increased transmigration through the endothelium. Similarly, remnant cholesterol has been identified to play a key role in endothelial dysfunction and monocyte behavior. In this review, we will discuss recent developments in understanding the role of different lipoproteins in the context of inflammation at both the level of the monocyte and the endothelium.
Collapse
Affiliation(s)
- Johan G Schnitzler
- Department of Vascular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Geesje M Dallinga-Thie
- Department of Vascular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands.,Department of Experimental Vascular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Jeffrey Kroon
- Department of Vascular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands.,Department of Experimental Vascular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
32
|
Silverstein RL. Oxidized Lipid Uptake by Scavenger Receptor CD36 (Cluster of Differentiation 36) Modulates Endothelial Surface Properties and May Contribute to Atherogenesis. Arterioscler Thromb Vasc Biol 2018; 38:4-5. [PMID: 29282244 DOI: 10.1161/atvbaha.117.310351] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Roy L Silverstein
- From the Department of Medicine, Medical College of Wisconsin, and Blood Research Institute, Blood Center of Wisconsin, Milwaukee
| |
Collapse
|
33
|
Hassanisaber H, Jafari L, Campeau MA, Drevelle O, Lauzon MA, Langelier E, Faucheux N, Rouleau L. The effect of substrate bulk stiffness on focal and fibrillar adhesion formation in human abdominal aortic endothelial cells. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2018; 98:572-583. [PMID: 30813060 DOI: 10.1016/j.msec.2018.12.130] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 12/07/2018] [Accepted: 12/28/2018] [Indexed: 01/01/2023]
Abstract
Endothelial cell (EC) dysfunction contributes to atherosclerosis, which is associated with arterial stiffening and fibronectin (FN) deposition, by ECs and smooth muscle cells (SMCs). The effect of stiffness on the EC/FN interaction and fibrillar adhesion formation has been poorly studied. An in vitro model was prepared that included FN-coated polydimethylsiloxane (PDMS) films with similar hydrophobicity and roughness but distinct Young's modulus values, mimicking healthy (1.0 MPa) and atherosclerotic (2.8 MPa) arteries. Human aortic abdominal endothelial cells (HAAECs) seeded on 1.0 MPa PDMS films spread over time and reached their maximum surface area faster than on 2.8 MPa PDMS films. In addition, HAAECs appeared to organize focal adhesion more rapidly on 1.0 MPa PDMS films, despite the similar cell binding domain accessibility to adsorbed FN. Interestingly, we also observed up to a ~5-fold increase in the percentage of HAAECs that had a well-developed fibrillar adhesion on 1.0 MPa compared to 2.8 MPa PDMS films as verified by integrin α5 subunits, tensin, and FN staining. This variation did not affect EC migration. These results suggest that there are favourable conditions for FN matrix assembly by ECs in early atherosclerosis rather than at advanced stages. Our in vitro model will therefore be helpful to understand the influence of bulk stiffness on cells involved in atherosclerosis.
Collapse
Affiliation(s)
- H Hassanisaber
- Cell-biomaterial Biohybrid Systems, Department of Chemical and Biotechnological Engineering, Université de Sherbrooke, 2500 boul. de l'Université, Sherbrooke, Québec, J1K 2R1, Canada.
| | - L Jafari
- Department of Mechanical Engineering, Université de Sherbrooke, 2500 boul. Université, Sherbrooke, Québec, J1K 2R1, Canada.
| | - M A Campeau
- Department of Chemical Engineering, McGill University, 3610 Rue University, Montréal, Québec, H3A 0C5, Canada.
| | - O Drevelle
- Cell-biomaterial Biohybrid Systems, Department of Chemical and Biotechnological Engineering, Université de Sherbrooke, 2500 boul. de l'Université, Sherbrooke, Québec, J1K 2R1, Canada; Thermal Plasma and Nanomaterial Synthesis Laboratory, Department of Chemical and Biotechnological Engineering, Université de Sherbrooke, 2500 boul. Université, Sherbrooke, Québec J1K 2R1, Canada.
| | - M-A Lauzon
- 3D Cell Culture Systems Laboratory, Department of Chemical and Biotechnological Engineering, Université de Sherbrooke, 2500 boul. Université, Sherbrooke, Québec J1K 2R1, Canada.
| | - E Langelier
- Department of Mechanical Engineering, Université de Sherbrooke, 2500 boul. Université, Sherbrooke, Québec, J1K 2R1, Canada; Clinical Research Centre of Centre Hospitalier Universitaire de Sherbrooke, 12e Avenue N, Sherbrooke, Québec J1H 5N4, Canada.
| | - N Faucheux
- Cell-biomaterial Biohybrid Systems, Department of Chemical and Biotechnological Engineering, Université de Sherbrooke, 2500 boul. de l'Université, Sherbrooke, Québec, J1K 2R1, Canada; Clinical Research Centre of Centre Hospitalier Universitaire de Sherbrooke, 12e Avenue N, Sherbrooke, Québec J1H 5N4, Canada.
| | - L Rouleau
- Department of Mechanical Engineering, Université de Sherbrooke, 2500 boul. Université, Sherbrooke, Québec, J1K 2R1, Canada.
| |
Collapse
|
34
|
Affiliation(s)
- Chantal M. Boulanger
- From the INSERM UMR-970, Paris Cardiovascular Research Center, Paris Descartes University, France
| |
Collapse
|
35
|
Lee FY, Sun CK, Sung PH, Chen KH, Chua S, Sheu JJ, Chung SY, Chai HT, Chen YL, Huang TH, Huang CR, Li YC, Luo CW, Yip HK. Daily melatonin protects the endothelial lineage and functional integrity against the aging process, oxidative stress, and toxic environment and restores blood flow in critical limb ischemia area in mice. J Pineal Res 2018; 65:e12489. [PMID: 29570854 DOI: 10.1111/jpi.12489] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 02/27/2018] [Indexed: 12/13/2022]
Abstract
We tested the hypothesis that daily melatonin treatment protects endothelial lineage and functional integrity against the aging process, oxidative stress/endothelial denudation (ED), and toxic environment and restored blood flow in murine critical limb ischemia (CLI). In vitro study using HUVECs, in vivo models (ie, CLI through left femoral artery ligation and ED through carotid artery wire injury), and model of lipopolysaccharide-induced aortic injury in young (3 months old) and aged (8 months old) mice were used to elucidate effects of melatonin treatment on vascular endothelial integrity. In vitro study showed that menadione-induced oxidative stress (NOX-1/NOX-2), inflammation (TNF-α/NF-kB), apoptosis (cleaved caspase-3/PARP), and mitochondrial damage (cytosolic cytochrome c) in HUVECs were suppressed by melatonin but reversed by SIRT3-siRNA (all P < .001). In vivo, reduced numbers of circulating endothelial progenitor cells (EPCs) (C-kit/CD31+/Sca-1/KDR+/CXCR4/CD34+), and angiogenesis (Matrigel assay of bone marrow-derived EPC and ex vivo aortic ring cultures) in older (compared with younger) mice were significantly reversed through daily melatonin administration (20 mg/kg/d, ip) (all P < .001). Aortic vasorelaxation and nitric oxide release were impaired in older mice and reversed in age-match mice receiving melatonin (all P < .01). ED-induced intimal/medial hyperplasia, reduced blood flow to ischemic limb, and angiogenesis (reduced CD31+/vWF+ cells/small vessel number) were improved after daily melatonin treatment (all P < .0001). Lipopolysaccharide-induced aortic endothelial cell detachment, which was more severe in aged mice, was also alleviated after daily melatonin treatment (P < .0001). Daily melatonin treatment protected both structural and functional integrity of vascular endothelium against aging-, oxidative stress-, lipopolysaccharide-, and ischemia-induced damage probably through upregulating the SIRT signaling pathway.
Collapse
Affiliation(s)
- Fan-Yen Lee
- Division of thoracic and Cardiovascular Surgery, Department of Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
- Center for Shockwave Medicine and Tissue Engineering, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
- Division of Cardiovascular Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Cheuk-Kwan Sun
- Department of Emergency Medicine, E-Da Hospital, I-Shou University School of Medicine for International Students, Kaohsiung, Taiwan
| | - Pei-Hsun Sung
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Kuan-Hung Chen
- Department of Anesthesiology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Sarah Chua
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Jiunn-Jye Sheu
- Division of thoracic and Cardiovascular Surgery, Department of Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Sheng-Ying Chung
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Han-Tan Chai
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Yi-Ling Chen
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Tien-Hung Huang
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chi-Ruei Huang
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Yi-Chen Li
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chi-Wen Luo
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Hon-Kan Yip
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
- Center for Shockwave Medicine and Tissue Engineering, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
- Department of Nursing, Asia University, Taichung, Taiwan
| |
Collapse
|
36
|
Le Master E, Fancher IS, Lee J, Levitan I. Comparative analysis of endothelial cell and sub-endothelial cell elastic moduli in young and aged mice: Role of CD36. J Biomech 2018; 76:263-268. [PMID: 29954596 DOI: 10.1016/j.jbiomech.2018.06.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 05/22/2018] [Accepted: 06/09/2018] [Indexed: 10/14/2022]
Abstract
OBJECTIVE To perform comparative analysis of the role of scavenger receptor CD36 on endothelial vs. sub-endothelial elastic modulus (stiffness) in the aortas of young and aged mice. APPROACHES AND RESULTS Elastic moduli of endothelial and sub-endothelial layers of freshly isolated mouse aortas were quantified using atomic force microscopy. In young mice (4-6 months old), we found that while endothelial stiffness is markedly reduced in aortas of CD36-/-mice, as compared to WT controls, no difference between CD36-/- and WT aortas is observed in the stiffness of the sub-endothelial layer in denuded arteries. Additionally, inhibition of myosin phosphorylation also decreases the elastic modulus in the EC, but not the sub-EC layer in WT mice. Moreover, inhibiting CD36 mediated uptake of oxLDL in intact WT aortas abrogated oxLDL-induced endothelial stiffening. Further analysis of aged mice (22-25 months) revealed that aging resulted not only in significant stiffening of the denuded arteries, as was previously known, but also a comparable increase in the elastic modulus of the endothelial layer. Most significantly, this stiffening in the EC layer is dependent on CD36, whereas the denuded layer is not affected. CONCLUSIONS Our results show that the role CD36 in stiffening of cellular components of intact aortas is endothelial-specific and that genetic deficiency of CD36 protects against endothelial stiffening in aged mice. Moreover, these data suggest that endothelial stiffness in intact mouse aortas depends more on the expression of CD36 than on the stiffness of the sub-endothelial layer.
Collapse
Affiliation(s)
- Elizabeth Le Master
- Division of Pulmonary and Critical Care, Department of Medicine, University of Illinois at Chicago, United States; Bioengineering, University of Illinois at Chicago, United States
| | - Ibra S Fancher
- Division of Pulmonary and Critical Care, Department of Medicine, University of Illinois at Chicago, United States
| | - James Lee
- Bioengineering, University of Illinois at Chicago, United States
| | - Irena Levitan
- Division of Pulmonary and Critical Care, Department of Medicine, University of Illinois at Chicago, United States; Bioengineering, University of Illinois at Chicago, United States.
| |
Collapse
|