1
|
Aguilar V, Le Master E, Paul A, Ahn SJ, Lazarko D, Febbraio M, Mehta D, Lee JC, Levitan I. Endothelial Stiffening Induced by CD36-Mediated Lipid Uptake Leads to Endothelial Barrier Disruption and Contributes to Atherosclerotic Lesions. Arterioscler Thromb Vasc Biol 2025; 45:e201-e216. [PMID: 40207364 DOI: 10.1161/atvbaha.124.322244] [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: 11/27/2024] [Accepted: 03/21/2025] [Indexed: 04/11/2025]
Abstract
BACKGROUND Endothelial stiffening induced by Western diet was proposed to be an important factor in vascular dysfunction. In this study, we determine the role of endothelial CD36 (cluster of differentiation 36) in stiffening, disruption of aortic endothelial barrier, and atherosclerosis in mouse models of obesity and hypercholesterolemia. METHODS To address this goal, we generated an endothelial-specific inducible knockdown mouse model of CD36, Cdh5.CreERT2CD36fl/fl, on C57/BL6J wild-type and LDLR-/- genetic backgrounds. Endothelial stiffness is assessed by atomic force microscopy; endothelial barrier integrity is assessed by imaging VE (vascular endothelium)-cadherin junctions and by penetration of Evans blue dye into the aortic wall. Atherosclerotic plaques are quantified using oil red O staining. RESULTS Endothelial-specific downregulation of CD36 abrogates stiffening of aortic endothelium induced by Western diet in Cdh5.CreERT2CD36fl/fl and in Cdh5.CreERT2CD36fl/flLDLR-/- mice. Prevention of Western diet-induced endothelial stiffening by downregulation of CD36 is associated with a protective effect against endothelial barrier disruption in both mouse models and with a significant decrease in the areas of atherosclerotic lesions in Cdh5.CreERT2CD36fl/flLDLR-/- mice. Mechanistically, stiffening of human aortic endothelial cells in vitro is induced by saturated fatty acids, particularly palmitic acid (PA), which results in activation of RhoA. Both PA-induced endothelial stiffening and RhoA activation are abrogated by CD36 siRNA. Furthermore, PA-induced endothelial stiffening of excised aortas ex vivo is lost in aortas isolated from mice, where endothelial CD36 is downregulated. We also demonstrate that PA-induced activation of RhoA and endothelial stiffening require expressing an RhoA-inhibitory protein, Rho-GDI1 (Rho guanosine dissociation inhibitor 1). Finally, we discover that PA disrupts the colocalization of RhoA with Rho-GDI1. CONCLUSIONS We conclude that stiffening of the aortic endothelium by CD36-mediated uptake of fatty acids contributes significantly to Western diet-induced vascular dysfunction and atherosclerosis. We further propose that fatty acids may activate RhoA by inducing its dissociation from Rho-GDI1.
Collapse
MESH Headings
- Animals
- CD36 Antigens/genetics
- CD36 Antigens/metabolism
- CD36 Antigens/deficiency
- Mice, Inbred C57BL
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Atherosclerosis/genetics
- Atherosclerosis/physiopathology
- Atherosclerosis/etiology
- Disease Models, Animal
- Mice, Knockout
- Cadherins/metabolism
- Cadherins/genetics
- Plaque, Atherosclerotic
- rhoA GTP-Binding Protein/metabolism
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/physiopathology
- Endothelium, Vascular/pathology
- Vascular Stiffness
- Endothelial Cells/metabolism
- Endothelial Cells/pathology
- Humans
- Diet, Western
- Male
- Obesity/metabolism
- Obesity/genetics
- Obesity/pathology
- Cells, Cultured
- Antigens, CD/metabolism
- Antigens, CD/genetics
- Signal Transduction
- Hypercholesterolemia/metabolism
- Hypercholesterolemia/genetics
- Hypercholesterolemia/pathology
- Receptors, LDL/genetics
- Receptors, LDL/deficiency
- Aorta/metabolism
- Aorta/pathology
- Aorta/physiopathology
- Aortic Diseases/metabolism
- Aortic Diseases/pathology
- Aortic Diseases/genetics
- Aortic Diseases/physiopathology
- Aortic Diseases/prevention & control
- Mice
- Capillary Permeability
Collapse
Affiliation(s)
- Victor Aguilar
- Departments of Medicine (V.A., E.L.M., A.P., S.J.A., D.L., I.L.), University of Illinois at Chicago
- Biomedical Engineering (V.A., J.C.L., I.L.), University of Illinois at Chicago
| | - Elizabeth Le Master
- Departments of Medicine (V.A., E.L.M., A.P., S.J.A., D.L., I.L.), University of Illinois at Chicago
| | - Amit Paul
- Departments of Medicine (V.A., E.L.M., A.P., S.J.A., D.L., I.L.), University of Illinois at Chicago
| | - Sang Joon Ahn
- Departments of Medicine (V.A., E.L.M., A.P., S.J.A., D.L., I.L.), University of Illinois at Chicago
| | - Dana Lazarko
- Departments of Medicine (V.A., E.L.M., A.P., S.J.A., D.L., I.L.), University of Illinois at Chicago
| | - Maria Febbraio
- Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada (M.F.)
| | - Dolly Mehta
- Pharmacology (D.M.), University of Illinois at Chicago
| | - James C Lee
- Biomedical Engineering (V.A., J.C.L., I.L.), University of Illinois at Chicago
| | - Irena Levitan
- Departments of Medicine (V.A., E.L.M., A.P., S.J.A., D.L., I.L.), University of Illinois at Chicago
- Biomedical Engineering (V.A., J.C.L., I.L.), University of Illinois at Chicago
| |
Collapse
|
2
|
Li J, Wang K, Starodubtseva MN, Nadyrov E, Kapron CM, Hoh J, Liu J. Complement factor H in molecular regulation of angiogenesis. MEDICAL REVIEW (2021) 2024; 4:452-466. [PMID: 39444793 PMCID: PMC11495524 DOI: 10.1515/mr-2023-0048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 06/07/2024] [Indexed: 10/25/2024]
Abstract
Angiogenesis, the process of formation of new capillaries from existing blood vessels, is required for multiple physiological and pathological processes. Complement factor H (CFH) is a plasma protein that inhibits the alternative pathway of the complement system. Loss of CFH enhances the alternative pathway and increases complement activation fragments with pro-angiogenic capacity, including complement 3a, complement 5a, and membrane attack complex. CFH protein contains binding sites for C-reactive protein, malondialdehyde, and endothelial heparan sulfates. Dysfunction of CFH prevents its interaction with these molecules and initiates pro-angiogenic events. Mutations in the CFH gene have been found in patients with age-related macular degeneration characterized by choroidal neovascularization. The Cfh-deficient mice show an increase in angiogenesis, which is decreased by administration of recombinant CFH protein. In this review, we summarize the molecular mechanisms of the anti-angiogenic effects of CFH and the regulatory mechanisms of CFH expression. The therapeutic potential of recombinant CFH protein in angiogenesis-related diseases has also been discussed.
Collapse
Affiliation(s)
- Jiang Li
- Laboratory of Translational Medicine in Microvascular Regulation, Institute of Microvascular Medicine, Medical Research Center, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong Province, China
- Medical Research Center, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong Province, China
- Shandong Provincial Key Medical and Health Laboratory of Translational Medicine in Microvascular Aging, Jinan, Shandong Province, China
| | - Kaili Wang
- Laboratory of Translational Medicine in Microvascular Regulation, Institute of Microvascular Medicine, Medical Research Center, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong Province, China
- Shandong Provincial Key Medical and Health Laboratory of Translational Medicine in Microvascular Aging, Jinan, Shandong Province, China
| | - Maria N. Starodubtseva
- Gomel State Medical University, Gomel, Belarus
- Institute of Radiobiology of NAS of Belarus, Gomel, Belarus
| | | | | | - Josephine Hoh
- Department of Ophthalmology, Yale School of Medicine, New Haven, CT, USA
| | - Ju Liu
- Laboratory of Translational Medicine in Microvascular Regulation, Institute of Microvascular Medicine, Medical Research Center, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong Province, China
- Gomel State Medical University, Gomel, Belarus
- Shandong Provincial Key Medical and Health Laboratory of Translational Medicine in Microvascular Aging, Jinan, Shandong Province, China
| |
Collapse
|
3
|
Tong KL, Mahmood Zuhdi AS, Wong PF. The role of miR-134-5p in 7-ketocholesterol-induced human aortic endothelial dysfunction. EXCLI JOURNAL 2024; 23:1073-1090. [PMID: 39391056 PMCID: PMC11464864 DOI: 10.17179/excli2024-7342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 08/22/2024] [Indexed: 10/12/2024]
Abstract
Atherosclerotic cardiovascular diseases are the leading causes of morbidity and mortality worldwide. In our previous study, a panel of miRNA including miR-134-5p was deregulated in young acute coronary syndrome (ACS) patients. However, the roles of these ACS-associated miRNAs in endothelial dysfunction, an early event preceding atherosclerosis, remain to be investigated. In the present study, human aortic endothelial cells (HAECs) were treated with 7-ketocholesterol (7-KC) to induce endothelial dysfunction. Following treatment with 20 μg/ml 7-KC, miR-134-5p was significantly up-regulated and endothelial nitric oxide synthase (eNOS) expression was suppressed. Endothelial barrier disruption was evidenced by the deregulation of adhesion molecules including the activation of focal adhesion kinase (FAK), down-regulation of VE-cadherin, up-regulation of adhesion molecules (E-selectin and ICAM-1), increased expression of inflammatory genes (IL1B, IL6 and COX2) and AKT activation. Knockdown of miR-134-5p in 7-KC-treated HAECs attenuated the suppression of eNOS, the activation of AKT, the down-regulation of VE-cadherin and the up-regulation of E-selectin. In addition, the interaction between miR-134-5p and FOXM1 mRNA was confirmed by the enrichment of FOXM1 transcripts in the pull-down miRNA-mRNA complex. Knockdown of miR-134-5p increased FOXM1 expression whereas transfection with mimic miR-134-5p decreased FOXM1 protein expression. In summary, the involvement of an ACS-associated miRNA, miR-134-5p in endothelial dysfunction was demonstrated. Findings from this study could pave future investigations into utilizing miRNAs as a supplementary tool in ACS diagnosis or as targets for the development of therapeutics.
Collapse
Affiliation(s)
- Kind-Leng Tong
- Department of Pharmacology, Faculty of Medicine, Universiti Malaya, 50603 Kuala Lumpur, Malaysia
| | | | - Pooi-Fong Wong
- Department of Pharmacology, Faculty of Medicine, Universiti Malaya, 50603 Kuala Lumpur, Malaysia
| |
Collapse
|
4
|
Calvo MJ, Parra H, Santeliz R, Bautista J, Luzardo E, Villasmil N, Martínez MS, Chacín M, Cano C, Checa-Ros A, D'Marco L, Bermúdez V, De Sanctis JB. The Placental Role in Gestational Diabetes Mellitus: A Molecular Perspective. TOUCHREVIEWS IN ENDOCRINOLOGY 2024; 20:10-18. [PMID: 38812661 PMCID: PMC11132656 DOI: 10.17925/ee.2024.20.1.5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 08/01/2023] [Indexed: 05/31/2024]
Abstract
During pregnancy, women undergo several metabolic changes to guarantee an adequate supply of glucose to the foetus. These metabolic modifications develop what is known as physiological insulin resistance. When this process is altered, however, gestational diabetes mellitus (GDM) occurs. GDM is a multifactorial disease, and genetic and environmental factors play a crucial role in its aetiopathogenesis. GDM has been linked to both macroscopic and molecular alterations in placental tissues that affect placental physiology. This review summarizes the role of the placenta in the development of GDM from a molecular perspective, including hormonal and pro-inflammatory changes. Inflammation and hormonal imbalance, the characteristics dominating the GDM microenvironment, are responsible for placental changes in size and vascularity, leading to dysregulation in maternal and foetal circulations and to complications in the newborn. In conclusion, since the hormonal mechanisms operating in GDM have not been fully elucidated, more research should be done to improve the quality of life of patients with GDM and their future children.
Collapse
Affiliation(s)
- María José Calvo
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Heliana Parra
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Raquel Santeliz
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Jordan Bautista
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Eliana Luzardo
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Nelson Villasmil
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - María Sofía Martínez
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Maricamen Chacín
- Facultad de Ciencias de la Salud, Barranquilla, Universidad Simón Bolívar, Barranquilla, Colombia
| | - Clímaco Cano
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Ana Checa-Ros
- Research Group on Cardiorenal and Metabolic Diseases, Departamento de Medicina y Cirugía, Facultad de Ciencias de la Salud, Universidad Cardenal Herrera-CEU, CEU Universities, Valencia, Spain
- School of Life and Health Sciences, Aston University, Birmingham, United Kingdom
| | - Luis D'Marco
- Research Group on Cardiorenal and Metabolic Diseases, Departamento de Medicina y Cirugía, Facultad de Ciencias de la Salud, Universidad Cardenal Herrera-CEU, CEU Universities, Valencia, Spain
| | - Valmore Bermúdez
- Facultad de Ciencias de la Salud, Barranquilla, Universidad Simón Bolívar, Barranquilla, Colombia
| | - Juan Bautista De Sanctis
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| |
Collapse
|
5
|
Kiseleva D, Kolmogorov V, Cherednichenko V, Khovantseva U, Bogatyreva A, Markina Y, Gorelkin P, Erofeev A, Markin A. Effect of LDL Extracted from Human Plasma on Membrane Stiffness in Living Endothelial Cells and Macrophages via Scanning Ion Conductance Microscopy. Cells 2024; 13:358. [PMID: 38391971 PMCID: PMC10887070 DOI: 10.3390/cells13040358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 02/02/2024] [Accepted: 02/16/2024] [Indexed: 02/24/2024] Open
Abstract
Mechanical properties of living cells play a crucial role in a wide range of biological functions and pathologies, including atherosclerosis. We used low-stress Scanning Ion-Conductance Microscopy (SICM) correlated with confocal imaging and demonstrated the topographical changes and mechanical properties alterations in EA.hy926 and THP-1 exposed to LDL extracted from CVD patients' blood samples. We show that the cells stiffened in the presence of LDL, which also triggered caveolae formation. Endothelial cells accumulated less cholesterol in the form of lipid droplets in comparison to THP-1 cells based on fluorescence intensity data and biochemical analysis; however, the effect on Young's modulus is higher. The cell stiffness is closely connected to the distribution of lipid droplets along the z-axis. In conclusion, we show that the sensitivity of endothelial cells to LDL is higher compared to that of THP-1, triggering changes in the cytoskeleton and membrane stiffness which may result in the increased permeability of the intima layer due to loss of intercellular connections and adhesion.
Collapse
Affiliation(s)
- Diana Kiseleva
- Department of Biophysics, Faculty of Biology, Lomonosov Moscow State University, 119991 Moscow, Russia
- Petrovsky National Research Center of Surgery, 119991 Moscow, Russia; (V.C.); (A.B.); (Y.M.)
| | - Vasilii Kolmogorov
- Laboratory of Biophysics, National University of Science and Technology MISIS, Leninskiy Prospect, 4, 119049 Moscow, Russia
| | - Vadim Cherednichenko
- Petrovsky National Research Center of Surgery, 119991 Moscow, Russia; (V.C.); (A.B.); (Y.M.)
| | - Ulyana Khovantseva
- Petrovsky National Research Center of Surgery, 119991 Moscow, Russia; (V.C.); (A.B.); (Y.M.)
| | - Anastasia Bogatyreva
- Petrovsky National Research Center of Surgery, 119991 Moscow, Russia; (V.C.); (A.B.); (Y.M.)
| | - Yuliya Markina
- Petrovsky National Research Center of Surgery, 119991 Moscow, Russia; (V.C.); (A.B.); (Y.M.)
| | - Petr Gorelkin
- Laboratory of Biophysics, National University of Science and Technology MISIS, Leninskiy Prospect, 4, 119049 Moscow, Russia
| | - Alexander Erofeev
- Laboratory of Biophysics, National University of Science and Technology MISIS, Leninskiy Prospect, 4, 119049 Moscow, Russia
| | - Alexander Markin
- Petrovsky National Research Center of Surgery, 119991 Moscow, Russia; (V.C.); (A.B.); (Y.M.)
- Medical Institute, Peoples’ Friendship University of Russia named after Patrice Lumumba (RUDN University), 117198 Moscow, Russia
| |
Collapse
|
6
|
Dias IHK, Shokr H. Oxysterols as Biomarkers of Aging and Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1440:307-336. [PMID: 38036887 DOI: 10.1007/978-3-031-43883-7_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/02/2023]
Abstract
Oxysterols derive from either enzymatic or non-enzymatic oxidation of cholesterol. Even though they are produced as intermediates of bile acid synthesis pathway, they are recognised as bioactive compounds in cellular processes. Therefore, their absence or accumulation have been shown to be associated with disease phenotypes. This chapter discusses the contribution of oxysterol to ageing, age-related diseases such as neurodegeneration and various disorders such as cancer, cardiovascular disease, diabetes, metabolic and ocular disorders. It is clear that oxysterols play a significant role in development and progression of these diseases. As a result, oxysterols are being investigated as suitable markers for disease diagnosis purposes and some drug targets are in development targeting oxysterol pathways. However, further research will be needed to confirm the suitability of these potentials.
Collapse
Affiliation(s)
- Irundika H K Dias
- Aston Medical School, College of Health and Life Sciences, Aston University, Birmingham, UK.
| | - Hala Shokr
- Manchester Pharmacy School, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| |
Collapse
|
7
|
Mahmood T, Miles JR, Minnier J, Tavori H, DeBarber AE, Fazio S, Shapiro MD. Effect of PCSK9 inhibition on plasma levels of small dense low density lipoprotein-cholesterol and 7-ketocholesterol. J Clin Lipidol 2024; 18:e50-e58. [PMID: 37923663 PMCID: PMC10957330 DOI: 10.1016/j.jacl.2023.10.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 09/19/2023] [Accepted: 10/20/2023] [Indexed: 11/07/2023]
Abstract
BACKGROUND Oxidized forms of cholesterol (oxysterols) are implicated in atherogenesis and can accumulate in the body via direct absorption from food or through oxidative reactions of endogenous cholesterol, inducing the formation of LDL particles loaded with oxidized cholesterol. It remains unknown whether drastic reductions in LDL-cholesterol (LDL-C) are associated with changes in circulating oxysterols and whether small dense LDL (sdLDL) are more likely to carry these oxysterols and susceptible to the effects of PCSK9 inhibition (PCSK9i). OBJECTIVE We investigate the effect of LDL-C reduction accomplished via PCSK9i on changes in plasma levels of sdLDL-cholesterol (sdLDL-C) and a common, stable oxysterol, 7-ketocholesterol (7-KC), among 134 patients referred to our Preventive Cardiology clinic. METHODS Plasma lipid panel, sdLDL-C, and 7-KC measurements were obtained from patients before and after initiation of PCSK9i. RESULTS The intervention caused a significant lowering of LDL-C (-55.4 %). The changes in sdLDL-C levels (mean reduction 51.4 %) were highly correlated with the reductions in LDL-C levels (R = 0.829, p < 0.001). Interestingly, whereas changes in plasma free 7-KC levels with PCSK9i treatment were much smaller than (-6.6 %) and did not parallel those of LDL-C and sdLDL-C levels, they did significantly correlate with changes in triglycerides and very low-density lipoprotein-cholesterol (VLDL-C) levels (R = 0.219, p = 0.025). CONCLUSION Our findings suggest a non-preferential clearance of LDL subparticles as a consequence of LDL receptor upregulation caused by PCSK9 inhibition. Moreover, the lack of significant reduction in 7-KC with PCSK9i suggests that 7-KC may be in part carried by VLDL and lost during lipoprotein processing leading to LDL formation.
Collapse
Affiliation(s)
- Tahir Mahmood
- Oregon Health & Science University, Knight Cardiovascular Institute, Center for Preventive Cardiology, Portland, OR, USA (Dr Mahmood, Miles, Minnier, Tavori and Fazio)
| | - Joshua R Miles
- Oregon Health & Science University, Knight Cardiovascular Institute, Center for Preventive Cardiology, Portland, OR, USA (Dr Mahmood, Miles, Minnier, Tavori and Fazio)
| | - Jessica Minnier
- Oregon Health & Science University, Knight Cardiovascular Institute, Center for Preventive Cardiology, Portland, OR, USA (Dr Mahmood, Miles, Minnier, Tavori and Fazio); Oregon Health & Science University, OHSU-PSU School of Public Health, Portland, OR, USA (Dr Minnier)
| | - Hagai Tavori
- Oregon Health & Science University, Knight Cardiovascular Institute, Center for Preventive Cardiology, Portland, OR, USA (Dr Mahmood, Miles, Minnier, Tavori and Fazio)
| | - Andrea E DeBarber
- Oregon Health & Science University, University Shared Resources, Portland, OR, USA (Dr DeBarber)
| | - Sergio Fazio
- Oregon Health & Science University, Knight Cardiovascular Institute, Center for Preventive Cardiology, Portland, OR, USA (Dr Mahmood, Miles, Minnier, Tavori and Fazio)
| | - Michael D Shapiro
- Wake Forest University School of Medicine, Section on Cardiovascular Medicine, Center for Prevention of Cardiovascular Disease, Winston-Salem, NC, USA (Dr Shapiro).
| |
Collapse
|
8
|
Domingues N, Gaifem J, Matthiesen R, Saraiva DP, Bento L, Marques ARA, Soares MIL, Sampaio J, Klose C, Surma MA, Almeida MS, Rodrigues G, Gonçalves PA, Ferreira J, E Melo RG, Pedro LM, Simons K, Pinho E Melo TMVD, Cabral MG, Jacinto A, Silvestre R, Vaz W, Vieira OV. Cholesteryl hemiazelate identified in CVD patients causes in vitro and in vivo inflammation. J Lipid Res 2023; 64:100419. [PMID: 37482218 PMCID: PMC10450993 DOI: 10.1016/j.jlr.2023.100419] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 07/13/2023] [Accepted: 07/14/2023] [Indexed: 07/25/2023] Open
Abstract
Oxidation of PUFAs in LDLs trapped in the arterial intima plays a critical role in atherosclerosis. Though there have been many studies on the atherogenicity of oxidized derivatives of PUFA-esters of cholesterol, the effects of cholesteryl hemiesters (ChEs), the oxidation end products of these esters, have not been studied. Through lipidomics analyses, we identified and quantified two ChE types in the plasma of CVD patients and identified four ChE types in human endarterectomy specimens. Cholesteryl hemiazelate (ChA), the ChE of azelaic acid (n-nonane-1,9-dioic acid), was the most prevalent ChE identified in both cases. Importantly, human monocytes, monocyte-derived macrophages, and neutrophils exhibit inflammatory features when exposed to subtoxic concentrations of ChA in vitro. ChA increases the secretion of proinflammatory cytokines such as interleukin-1β and interleukin-6 and modulates the surface-marker profile of monocytes and monocyte-derived macrophage. In vivo, when zebrafish larvae were fed with a ChA-enriched diet, they exhibited neutrophil and macrophage accumulation in the vasculature in a caspase 1- and cathepsin B-dependent manner. ChA also triggered lipid accumulation at the bifurcation sites of the vasculature of the zebrafish larvae and negatively impacted their life expectancy. We conclude that ChA behaves as an endogenous damage-associated molecular pattern with inflammatory and proatherogenic properties.
Collapse
Affiliation(s)
- Neuza Domingues
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, (NMS, FCM), Universidade Nova de Lisboa, Lisboa, Portugal
| | - Joana Gaifem
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Portugal and ICVS/3B's, PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Rune Matthiesen
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, (NMS, FCM), Universidade Nova de Lisboa, Lisboa, Portugal
| | - Diana P Saraiva
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, (NMS, FCM), Universidade Nova de Lisboa, Lisboa, Portugal
| | - Luís Bento
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, (NMS, FCM), Universidade Nova de Lisboa, Lisboa, Portugal
| | - André R A Marques
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, (NMS, FCM), Universidade Nova de Lisboa, Lisboa, Portugal
| | - Maria I L Soares
- Department of Chemistry, Coimbra Chemistry Centre, Institute of Molecular Sciences, University of Coimbra, Coimbra, Portugal
| | | | | | | | - Manuel S Almeida
- Hospital Santa Cruz, Centro Hospitalar de Lisboa Ocidental, Carnaxide, Portugal
| | - Gustavo Rodrigues
- Hospital Santa Cruz, Centro Hospitalar de Lisboa Ocidental, Carnaxide, Portugal
| | | | - Jorge Ferreira
- Hospital Santa Cruz, Centro Hospitalar de Lisboa Ocidental, Carnaxide, Portugal
| | - Ryan Gouveia E Melo
- Department of Vascular Surgery, Hospital de Santa Maria, Centro Hospitalar Universitario Lisboa Norte (CHULN), Lisboa, Portugal
| | - Luís Mendes Pedro
- Department of Vascular Surgery, Hospital de Santa Maria, Centro Hospitalar Universitario Lisboa Norte (CHULN), Lisboa, Portugal
| | | | - Teresa M V D Pinho E Melo
- Department of Chemistry, Coimbra Chemistry Centre, Institute of Molecular Sciences, University of Coimbra, Coimbra, Portugal
| | - M Guadalupe Cabral
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, (NMS, FCM), Universidade Nova de Lisboa, Lisboa, Portugal
| | - Antonio Jacinto
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, (NMS, FCM), Universidade Nova de Lisboa, Lisboa, Portugal
| | - Ricardo Silvestre
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Portugal and ICVS/3B's, PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Winchil Vaz
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, (NMS, FCM), Universidade Nova de Lisboa, Lisboa, Portugal
| | - Otília V Vieira
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, (NMS, FCM), Universidade Nova de Lisboa, Lisboa, Portugal.
| |
Collapse
|
9
|
Pan J, Wang B, Pu X, Qiu C, Li D, Wu Z, Zhang H, He Y. lncRNA GAPLINC regulates vascular endothelial cell apoptosis in atherosclerosis. Arch Med Sci 2023; 20:216-232. [PMID: 38414459 PMCID: PMC10895973 DOI: 10.5114/aoms/169383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 07/08/2023] [Indexed: 02/29/2024] Open
Abstract
Introduction In this study, we investigated the role of the long non-coding RNA GAPLINC in atherosclerosis under oxidized low-density lipoprotein (ox-LDL) treatment. Material and methods We utilized ox-LDL exposed human aortic endothelial cells as an in-vitro model. The expression level of GAPLINC was quantified by qPCR in different times and concentrations of ox-LDL treatment conditions. Cell apoptosis rate and cell cycle profiles were assessed by flow cytometry and TUNEL assay. The target association was confirmed using a luciferase reporter assay and Western blot. Results We found that GAPLINC expression was induced by ox-LDL treatment, but cell proliferation ability was significantly inhibited. We further confirmed that overexpressing of lncRNA GAPLINC in ox-LDL-exposed HAECs decreased cell proliferation by increasing cell apoptosis and arresting cell cycle in G2/M and S phase. Importantly, the detailed mechanistic analysis elucidated that LncRNA GAPLINC acts as a decoy to sequester miR-183-5p to prevent it from binding to target PDCD4. MiR-183-5p targets GAPLINC, and PDCD4 is a downstream target of miR-183-5p, and the cellular effects of this direct interaction were confirmed by a rescue assay experiment. Conclusions The present study demonstrates that upregulation of lncRNA GAPLINC represses the binding of miR-183-5p to the PDCD4 promoter region and then promotes PDCD4 expression, thereby inducing cell apoptosis and suppressing endothelial cell proliferation.
Collapse
Affiliation(s)
- Jun Pan
- Department of Vascular Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Bing Wang
- Department of Vascular Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xibin Pu
- Department of Vascular Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Department of General Surgery, Haiyan People's Hospital. Haiyan, China
| | - Chenyang Qiu
- Department of Vascular Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Donglin Li
- Department of Vascular Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Ziheng Wu
- Department of Vascular Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Honkun Zhang
- Department of Vascular Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yangyan He
- Department of Vascular Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
10
|
Higashi Y. Endothelial Function in Dyslipidemia: Roles of LDL-Cholesterol, HDL-Cholesterol and Triglycerides. Cells 2023; 12:1293. [PMID: 37174693 PMCID: PMC10177132 DOI: 10.3390/cells12091293] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 04/24/2023] [Accepted: 04/28/2023] [Indexed: 05/15/2023] Open
Abstract
Dyslipidemia is associated with endothelial dysfunction. Endothelial dysfunction is the initial step for atherosclerosis, resulting in cardiovascular complications. It is clinically important to break the process of endothelial dysfunction to cardiovascular complications in patients with dyslipidemia. Lipid-lowering therapy enables the improvement of endothelial function in patients with dyslipidemia. It is likely that the relationships of components of a lipid profile such as low-density lipoprotein cholesterol, high-density lipoprotein cholesterol and triglycerides with endothelial function are not simple. In this review, we focus on the roles of components of a lipid profile in endothelial function.
Collapse
Affiliation(s)
- Yukihito Higashi
- Department of Regenerative Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima 743-8551, Japan; ; Tel.: +81-82-257-5831
- Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima 734-8553, Japan
| |
Collapse
|
11
|
Lin J, de Rezende VL, de Aguiar da Costa M, de Oliveira J, Gonçalves CL. Cholesterol metabolism pathway in autism spectrum disorder: From animal models to clinical observations. Pharmacol Biochem Behav 2023; 223:173522. [PMID: 36717034 DOI: 10.1016/j.pbb.2023.173522] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 12/18/2022] [Accepted: 01/24/2023] [Indexed: 01/29/2023]
Abstract
Autism Spectrum Disorder (ASD) is a neurodevelopmental disorder characterized by a persistent impairment of social skills, including aspects of perception, interpretation, and response, combined with restricted and repetitive behavior. ASD is a complex and multifactorial condition, and its etiology could be attributed to genetic and environmental factors. Despite numerous clinical and experimental studies, no etiological factor, biomarker, and specific model of transmission have been consistently associated with ASD. However, an imbalance in cholesterol levels has been observed in many patients, more specifically, a condition of hypocholesterolemia, which seems to be shared between ASD and ASD-related genetic syndromes such as fragile X syndrome (FXS), Rett syndrome (RS), and Smith- Lemli-Opitz (SLO). Furthermore, it is known that alterations in cholesterol levels lead to neuroinflammation, oxidative stress, impaired myelination and synaptogenesis. Thus, the aim of this review is to discuss the cholesterol metabolic pathways in the ASD context, as well as in genetic syndromes related to ASD, through clinical observations and animal models. In fact, SLO, FXS, and RS patients display early behavioral markers of ASD followed by cholesterol disturbances. Several studies have demonstrated the role of cholesterol in psychiatric conditions and how its levels modulate brain neurodevelopment. This review suggests an important relationship between ASD pathology and cholesterol metabolism impairment; thus, some strategies could be raised - at clinical and pre-clinical levels - to explore whether cholesterol metabolism disturbance has a generally adverse effect in exacerbating the symptoms of ASD patients.
Collapse
Affiliation(s)
- Jaime Lin
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Victória Linden de Rezende
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Maiara de Aguiar da Costa
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Jade de Oliveira
- Laboratory for Research in Metabolic Disorders and Neurodegenerative Diseases, Graduate Program in Health Sciences, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Cinara Ludvig Gonçalves
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil.
| |
Collapse
|
12
|
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
|
13
|
Li H, Xie X, Zhang L, He Y, Liu H, Qiang D, Bai G, Li L, Tang Y. Ultra-high-performance liquid chromatography-tandem mass spectrometry analysis of serum metabolomic characteristics in people with different vitamin D levels. Open Med (Wars) 2023; 18:20230658. [PMID: 36874363 PMCID: PMC9979004 DOI: 10.1515/med-2023-0658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 01/14/2023] [Accepted: 01/14/2023] [Indexed: 03/05/2023] Open
Abstract
Vitamin D is a fat-soluble vitamin with multiple functions. However, the metabolism of people with different vitamin D concentrations is still unclear. Herein, we collected clinical data and analysed the serum metabolome of people with 25-hydroxyvitamin D (25[OH]D) ≥40 ng/mL (A), 30 ng/mL ≤25(OH)D <40 ng/mL (B) and 25(OH)D <30 ng/mL (C) by the ultra-high-performance liquid chromatography-tandem mass spectrometry method. We found that haemoglobin A1c, fasting blood glucose, fasting insulin, homeostasis model assessment of insulin resistance and thioredoxin interaction protein were enhanced, while HOMA-β was reduced with the decrease of 25(OH)D concentration. In addition, people in the C group were diagnosed with prediabetes or diabetes. Metabolomics analysis showed that seven, thirty-four and nine differential metabolites were identified in the groups B vs A, C vs A and C vs B, respectively. Metabolites associated with cholesterol metabolism and bile acid biosynthesis, such as 7-ketolithocholic acid, 12-ketolithocholic acid, apocholic acid, N-arachidene glycine and d-mannose 6-phosphate, were significantly upregulated in the C group compared with the A or B groups. In conclusion, the disorder of vitamin D metabolism may be related to cholesterol metabolism and bile acid biosynthesis. This study provided a basis for exploring the possible mechanism leading to abnormal vitamin D metabolism.
Collapse
Affiliation(s)
- Huan Li
- Department of Endocrinology, The First People's Hospital of Yinchuan, Yinchuan City, 750001, Ningxia Hui Autonomous Region, China
| | - Xiaomin Xie
- Department of Endocrinology, The First People's Hospital of Yinchuan, Yinchuan City, Liqun West Street 2, 750001, Ningxia Hui Autonomous Region, China
| | - Li Zhang
- Department of Endocrinology, The First People's Hospital of Yinchuan, Yinchuan City, 750001, Ningxia Hui Autonomous Region, China
| | - Yanting He
- Department of Endocrinology, The First People's Hospital of Yinchuan, Yinchuan City, 750001, Ningxia Hui Autonomous Region, China
| | - Huili Liu
- Department of Endocrinology, The First People's Hospital of Yinchuan, Yinchuan City, 750001, Ningxia Hui Autonomous Region, China
| | - Dan Qiang
- Department of Endocrinology, The First People's Hospital of Yinchuan, Yinchuan City, 750001, Ningxia Hui Autonomous Region, China
| | - Guirong Bai
- Department of Endocrinology, The First People's Hospital of Yinchuan, Yinchuan City, 750001, Ningxia Hui Autonomous Region, China
| | - Ling Li
- Department of Endocrinology, The First People's Hospital of Yinchuan, Yinchuan City, 750001, Ningxia Hui Autonomous Region, China
| | - Yanpan Tang
- Department of Endocrinology, The First People's Hospital of Yinchuan, Yinchuan City, 750001, Ningxia Hui Autonomous Region, China
| |
Collapse
|
14
|
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: 12] [Impact Index Per Article: 4.0] [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
|
15
|
Uchikawa T, Matoba T, Kawahara T, Baba I, Katsuki S, Koga JI, Hashimoto Y, Yamasaki R, Ichi I, Akita H, Tsutsui H. Dietary 7-ketocholesterol exacerbates myocardial ischemia-reperfusion injury in mice through monocyte/macrophage-mediated inflammation. Sci Rep 2022; 12:14902. [PMID: 36050346 PMCID: PMC9436973 DOI: 10.1038/s41598-022-19065-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 08/24/2022] [Indexed: 11/22/2022] Open
Abstract
Emerging evidence suggests that 7-ketocholesterol (7-KC), one of the most abundant dietary oxysterols, causes inflammation and cardiovascular diseases. Here we show the deteriorating effects of dietary 7-KC on myocardial ischemia-reperfusion (IR) injury and detailed the molecular mechanisms. A high-fat high-cholesterol diet containing 7-KC (7KWD) for 3 weeks increased the plasma 7-KC level compared with high-fat high-cholesterol diet in mice. In wild-type mice but not in CCR2-/- mice, dietary 7-KC increased the myocardial infarct size after IR. Flow cytometry revealed that the ratio of Ly-6Chigh inflammatory monocytes to total monocytes was increased in the 7KWD group. Unbiased RNA sequencing using murine primary macrophages revealed that 7-KC regulated the expression of transcripts related to inflammation and cholesterol biosynthesis. We further validated that in vitro, 7-KC induced endoplasmic reticulum stress, mitochondrial reactive oxygen species production, and nuclear factor-kappa B activation, which are associated with increased mRNA levels of proinflammatory cytokines. Administration of N-acetyl-L-cysteine or siRNA-mediated knockdown of PKR-like endoplasmic reticulum kinase or endoplasmic reticulum oxidase 1α suppressed the levels of 7-KC-induced inflammation. Dietary 7-KC exacerbates myocardial IR injury through monocyte/macrophage-mediated inflammation. Endoplasmic reticulum stress and oxidative stress are involved in the 7-KC-induced proinflammatory response in macrophages.
Collapse
Affiliation(s)
- Tomoki Uchikawa
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-Ku, Fukuoka, 812-8582, Japan
- Division of Cardiovascular Medicine, Faculty of Medical Sciences, Research Institute of Angiocardiology, Kyushu University, Fukuoka, Japan
| | - Tetsuya Matoba
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-Ku, Fukuoka, 812-8582, Japan.
| | - Takuro Kawahara
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-Ku, Fukuoka, 812-8582, Japan
- Division of Cardiovascular Medicine, Faculty of Medical Sciences, Research Institute of Angiocardiology, Kyushu University, Fukuoka, Japan
| | - Isashi Baba
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-Ku, Fukuoka, 812-8582, Japan
- Division of Cardiovascular Medicine, Faculty of Medical Sciences, Research Institute of Angiocardiology, Kyushu University, Fukuoka, Japan
| | - Shunsuke Katsuki
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Jun-Ichiro Koga
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Yu Hashimoto
- Department of Neurology, Graduate School of Medical Sciences, Neurological Institute, Kyushu University, Fukuoka, Japan
| | - Ryo Yamasaki
- Department of Neurology, Graduate School of Medical Sciences, Neurological Institute, Kyushu University, Fukuoka, Japan
| | - Ikuyo Ichi
- Graduate School of Humanities and Science, Ochanomizu University, Tokyo, Japan
| | - Hidetaka Akita
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Hiroyuki Tsutsui
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-Ku, Fukuoka, 812-8582, Japan
- Division of Cardiovascular Medicine, Faculty of Medical Sciences, Research Institute of Angiocardiology, Kyushu University, Fukuoka, Japan
| |
Collapse
|
16
|
Sarkar P, Kumar GA, Shrivastava S, Chattopadhyay A. Chronic cholesterol depletion increases F-actin levels and induces cytoskeletal reorganization via a dual mechanism. J Lipid Res 2022; 63:100206. [PMID: 35390404 PMCID: PMC9096963 DOI: 10.1016/j.jlr.2022.100206] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 03/22/2022] [Accepted: 03/28/2022] [Indexed: 12/24/2022] Open
Abstract
Previous work from us and others has suggested that cholesterol is an important lipid in the context of the organization of the actin cytoskeleton. However, reorganization of the actin cytoskeleton upon modulation of membrane cholesterol is rarely addressed in the literature. In this work, we explored the signaling crosstalk between cholesterol and the actin cytoskeleton by using a high-resolution confocal microscopic approach to quantitatively measure changes in F-actin content upon cholesterol depletion. Our results show that F-actin content significantly increases upon chronic cholesterol depletion, but not during acute cholesterol depletion. In addition, utilizing inhibitors targeting the cholesterol biosynthetic pathway at different steps, we show that reorganization of the actin cytoskeleton could occur due to the synergistic effect of multiple pathways, including prenylated Rho GTPases and availability of membrane phosphatidylinositol 4,5-bisphosphate. These results constitute one of the first comprehensive dissections of the mechanistic basis underlying the interplay between cellular actin levels and cholesterol biosynthesis. We envision these results will be relevant for future understating of the remodeling of the actin cytoskeleton in pathological conditions with altered cholesterol.
Collapse
Affiliation(s)
- Parijat Sarkar
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India
| | - G Aditya Kumar
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India
| | | | | |
Collapse
|
17
|
Levitan I. Evaluating membrane structure by Laurdan imaging: Disruption of lipid packing by oxidized lipids. CURRENT TOPICS IN MEMBRANES 2021; 88:235-256. [PMID: 34862028 DOI: 10.1016/bs.ctm.2021.10.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Impact of different lipids on membrane structure/lipid order is critical for multiple biological processes. Laurdan microscopy provides a unique tool to assess this property in heterogeneous biological membranes. This review describes the general principles of the approach and its application in model membranes and cells. It also provides an in-depth discussion of the insights obtained using Laurdan microscopy to evaluate the differential effects of cholesterol, oxysterols and oxidized phospholipids on lipid packing of ordered and disordered domains in vascular endothelial cells.
Collapse
Affiliation(s)
- Irena Levitan
- Division of Pulmonary and Critical Care, Department of Medicine, University of Illinois at Chicago, Chicago, IL, United States.
| |
Collapse
|
18
|
Ayee MA, Levitan I. Lipoprotein-Induced Increases in Cholesterol and 7-Ketocholesterol Result in Opposite Molecular-Scale Biophysical Effects on Membrane Structure. Front Cardiovasc Med 2021; 8:715932. [PMID: 34336964 PMCID: PMC8322651 DOI: 10.3389/fcvm.2021.715932] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 06/21/2021] [Indexed: 11/13/2022] Open
Abstract
Under hypercholesterolemic conditions, exposure of cells to lipoproteins results in a subtle membrane increase in the levels of cholesterol and 7-ketocholesterol, as compared to normal conditions. The effect of these physiologically relevant concentration increases on multicomponent bilayer membranes was investigated using coarse-grained molecular dynamics simulations. Significant changes in the structural and dynamic properties of the bilayer membranes resulted from these subtle increases in sterol levels, with both sterol species inducing decreases in the lateral area and inhibiting lateral diffusion to varying extents. Cholesterol and 7-ketocholesterol, however, exhibited opposite effects on lipid packing and orientation. The results from this study indicate that the subtle increases in membrane sterol levels induced by exposure to lipoproteins result in molecular-scale biophysical perturbation of membrane structure.
Collapse
Affiliation(s)
- Manuela A.A. Ayee
- Department of Engineering, Dordt University, Sioux Center, IA, United States
| | - Irena Levitan
- Division of Pulmonary and Critical Care, Department of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
19
|
ROCK Inhibition as Potential Target for Treatment of Pulmonary Hypertension. Cells 2021; 10:cells10071648. [PMID: 34209333 PMCID: PMC8303917 DOI: 10.3390/cells10071648] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/17/2021] [Accepted: 06/23/2021] [Indexed: 02/07/2023] Open
Abstract
Pulmonary hypertension (PH) is a cardiovascular disease caused by extensive vascular remodeling in the lungs, which ultimately leads to death in consequence of right ventricle (RV) failure. While current drugs for PH therapy address the sustained vasoconstriction, no agent effectively targets vascular cell proliferation and tissue inflammation. Rho-associated protein kinases (ROCKs) emerged in the last few decades as promising targets for PH therapy, since ROCK inhibitors demonstrated significant anti-remodeling and anti-inflammatory effects. In this review, current aspects of ROCK inhibition therapy are discussed in relation to the treatment of PH and RV dysfunction, from cell biology to preclinical and clinical studies.
Collapse
|
20
|
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: 24] [Impact Index Per Article: 6.0] [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
|
21
|
Yemanyi F, Baidouri H, Burns AR, Raghunathan V. Dexamethasone and Glucocorticoid-Induced Matrix Temporally Modulate Key Integrins, Caveolins, Contractility, and Stiffness in Human Trabecular Meshwork Cells. Invest Ophthalmol Vis Sci 2021; 61:16. [PMID: 33170205 PMCID: PMC7686803 DOI: 10.1167/iovs.61.13.16] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Purpose To determine the temporal effects of dexamethasone (DEX) and glucocorticoid-induced matrix (GIM) on integrins/integrin adhesomes, caveolins, cytoskeletal-related proteins, and stiffness in human trabecular meshwork (hTM) cells. Methods Primary hTM cells were plated on plastic dishes (TCP), treated with vehicle (Veh) or 100 nM DEX in 1% serum media for 1, 3, 5, and 7 day(s). Concurrently, hTM cells were also plated on vehicle control matrices (VehMs) and GIMs for similar time points; VehMs and GIMs had been generated from chronic cultures of Veh-/DEX-stimulated hTM cells and characterized biochemically. Subsets of cells prior to plating on TCP or VehMs / GIMs served as baseline. Protein expression of mechanoreceptors, cytoskeletal-related proteins, and elastic moduli of hTM cells were determined. Results Compared with Veh, DEX temporally overexpressed αV, β3, and β5 integrins from day 3 to day 7, and integrin linked kinase at day 7, in hTM cells. However, DEX decreased β1 integrin at day 1 and day 7, while increasing Cavin1 at day 7, in a time-independent manner. Further, DEX temporally upregulated α-smooth muscle actin(α-SMA) and RhoA at day 7 and day 5, respectively; while temporally downregulating Cdc42 at day 3 and day 7 in hTM cells. Conversely, GIM showed increased immunostaining of fibronectin extra-domain A and B isoforms. Compared with VehM, GIM temporally increased αV integrin, Cavin1, and RhoA from day 3 to day 7, at day 3 and day 7, and at day 5, respectively, in hTM cells. Further, GIM overexpressed α-SMA at day 3 and day 7, and stiffened hTM cells from day 1 to day 7, in a time-independent fashion. Conclusions Our data highlight crucial mechanoreceptors, integrin adhesomes, and actin-related proteins that may temporally sustain fibrotic phenotypes precipitated by DEX and/or GIM in hTM cells.
Collapse
Affiliation(s)
- Felix Yemanyi
- Department of Basic Sciences, University of Houston College of Optometry, Houston, Texas, United States
| | - Hasna Baidouri
- Department of Basic Sciences, University of Houston College of Optometry, Houston, Texas, United States
| | - Alan R Burns
- Department of Basic Sciences, University of Houston College of Optometry, Houston, Texas, United States
| | - VijayKrishna Raghunathan
- Department of Basic Sciences, University of Houston College of Optometry, Houston, Texas, United States.,Department of Biomedical Engineering, Cullen College of Engineering, University of Houston, Houston, Texas, United States
| |
Collapse
|
22
|
Zhang X, Alhasani RH, Zhou X, Reilly J, Zeng Z, Strang N, Shu X. Oxysterols and retinal degeneration. Br J Pharmacol 2021; 178:3205-3219. [PMID: 33501641 DOI: 10.1111/bph.15391] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 01/13/2021] [Accepted: 01/19/2021] [Indexed: 12/11/2022] Open
Abstract
Retinal degeneration, characterised by the progressive death of retinal neurons, is the most common cause of visual impairment. Oxysterols are the cholesterol derivatives produced via enzymatic and/or free radical oxidation that regulate cholesterol homeostasis in the retina. Preclinical and clinical studies have suggested a connection between oxysterols and retinal degeneration. Here, we summarise early and recent work related to retina oxysterol-producing enzymes and the distribution of oxysterols in the retina. We examine the impact of loss of oxysterol-producing enzymes on retinal pathology and explore the molecular mechanisms associated with the toxic or protective roles of individual oxysterols in different types of retinal degeneration. We conclude that increased efforts to better understand the oxysterol-associated pathophysiology will help in the development of effective retinal degeneration therapies. LINKED ARTICLES: This article is part of a themed issue on Oxysterols, Lifelong Health and Therapeutics. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.16/issuetoc.
Collapse
Affiliation(s)
- Xun Zhang
- Department of Biological and Biomedical Sciences, Glasgow Caledonian University, Glasgow, UK
| | - Reem Hasaballah Alhasani
- Department of Biological and Biomedical Sciences, Glasgow Caledonian University, Glasgow, UK.,Department of Biology, Faculty of Applied Science, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Xinzhi Zhou
- Department of Biological and Biomedical Sciences, Glasgow Caledonian University, Glasgow, UK
| | - James Reilly
- Department of Biological and Biomedical Sciences, Glasgow Caledonian University, Glasgow, UK
| | - Zhihong Zeng
- College of Biological and Environmental Engineering, Changsha University, Changsha, Hunan, China
| | - Niall Strang
- Department of Vision Science, Glasgow Caledonian University, Glasgow, UK
| | - Xinhua Shu
- Department of Biological and Biomedical Sciences, Glasgow Caledonian University, Glasgow, UK.,Department of Vision Science, Glasgow Caledonian University, Glasgow, UK.,School of Basic Medical Sciences, Shaoyang University, Shaoyang, Hunan, China
| |
Collapse
|
23
|
Gianazza E, Brioschi M, Martinez Fernandez A, Casalnuovo F, Altomare A, Aldini G, Banfi C. Lipid Peroxidation in Atherosclerotic Cardiovascular Diseases. Antioxid Redox Signal 2021; 34:49-98. [PMID: 32640910 DOI: 10.1089/ars.2019.7955] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Significance: Atherosclerotic cardiovascular diseases (ACVDs) continue to be a primary cause of mortality worldwide in adults aged 35-70 years, occurring more often in countries with lower economic development, and they constitute an ever-growing global burden that has a considerable socioeconomic impact on society. The ACVDs encompass diverse pathologies such as coronary artery disease and heart failure (HF), among others. Recent Advances: It is known that oxidative stress plays a relevant role in ACVDs and some of its effects are mediated by lipid oxidation. In particular, lipid peroxidation (LPO) is a process under which oxidants such as reactive oxygen species attack unsaturated lipids, generating a wide array of oxidation products. These molecules can interact with circulating lipoproteins, to diffuse inside the cell and even to cross biological membranes, modifying target nucleophilic sites within biomolecules such as DNA, lipids, and proteins, and resulting in a plethora of biological effects. Critical Issues: This review summarizes the evidence of the effect of LPO in the development and progression of atherosclerosis-based diseases, HF, and other cardiovascular diseases, highlighting the role of protein adduct formation. Moreover, potential therapeutic strategies targeted at lipoxidation in ACVDs are also discussed. Future Directions: The identification of valid biomarkers for the detection of lipoxidation products and adducts may provide insights into the improvement of the cardiovascular risk stratification of patients and the development of therapeutic strategies against the oxidative effects that can then be applied within a clinical setting.
Collapse
Affiliation(s)
- Erica Gianazza
- Proteomics Unit, Monzino Cardiology Center IRCCS, Milan, Italy
| | - Maura Brioschi
- Proteomics Unit, Monzino Cardiology Center IRCCS, Milan, Italy
| | | | | | | | - Giancarlo Aldini
- Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
| | - Cristina Banfi
- Proteomics Unit, Monzino Cardiology Center IRCCS, Milan, Italy
| |
Collapse
|
24
|
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
|
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
|
Zhang L, She ZG, Li H, Zhang XJ. Non-alcoholic fatty liver disease: a metabolic burden promoting atherosclerosis. Clin Sci (Lond) 2020; 134:1775-1799. [PMID: 32677680 DOI: 10.1042/cs20200446] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 06/06/2020] [Accepted: 06/29/2020] [Indexed: 02/07/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) has become the fastest growing chronic liver disease, with a prevalence of up to 25% worldwide. Individuals with NAFLD have a high risk of disease progression to cirrhosis, hepatocellular carcinoma (HCC), and liver failure. With the exception of intrahepatic burden, cardiovascular disease (CVD) and especially atherosclerosis (AS) are common complications of NAFLD. Furthermore, CVD is a major cause of death in NAFLD patients. Additionally, AS is a metabolic disorder highly associated with NAFLD, and individual NAFLD pathologies can greatly increase the risk of AS. It is increasingly clear that AS-associated endothelial cell damage, inflammatory cell activation, and smooth muscle cell proliferation are extensively impacted by NAFLD-induced systematic dyslipidemia, inflammation, oxidative stress, the production of hepatokines, and coagulations. In clinical trials, drug candidates for NAFLD management have displayed promising effects for the treatment of AS. In this review, we summarize the key molecular events and cellular factors contributing to the metabolic burden induced by NAFLD on AS, and discuss therapeutic strategies for the improvement of AS in individuals with NAFLD.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Model Animal of Wuhan University, Luojia Mount Wuchang, Wuhan 430072, China
| | - Zhi-Gang She
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Model Animal of Wuhan University, Luojia Mount Wuchang, Wuhan 430072, China
| | - Hongliang Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Model Animal of Wuhan University, Luojia Mount Wuchang, Wuhan 430072, China
- Basic Medical School, Wuhan University, Wuhan 430071, China
- Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Xiao-Jing Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Model Animal of Wuhan University, Luojia Mount Wuchang, Wuhan 430072, China
| |
Collapse
|
27
|
Li HY, Yuan Y, Fu YH, Wang Y, Gao XY. Hypoxia-inducible factor-1α: A promising therapeutic target for vasculopathy in diabetic retinopathy. Pharmacol Res 2020; 159:104924. [PMID: 32464323 DOI: 10.1016/j.phrs.2020.104924] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 05/04/2020] [Accepted: 05/10/2020] [Indexed: 02/07/2023]
Abstract
Diabetic retinopathy (DR) is a serious condition that can cause blindness in diabetic patients. It is a neurovascular disease, but the pathogenesis leading to the onset of this disease is still not completely understood. However, hypoxia with subsequent neovascularization is a characteristic phenomenon observed with DR. Cellular response to hypoxia is mediated by the transcriptional regulator hypoxia-inducible factor (HIF). Long-term research has shown that one isotype of HIF, HIF-1α, may play a pivotal role under hypoxic conditions, and an increasing number of studies have shown that HIF-1α and its target genes contribute to retinal neovascularization. Therefore, targeting HIF-1α may lead to more effective DR treatments. This review describes the possible mechanisms of HIF-1α in neovascularization of DR. Furthermore, various inhibitors of HIF-1α that may have viable potential in the treatment of DR are also discussed.
Collapse
Affiliation(s)
- Hui-Yao Li
- Department of Endocrinology, First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Yue Yuan
- Department of Endocrinology, First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Yu-Hong Fu
- Department of Endocrinology, First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Ying Wang
- Department of Endocrinology, First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Xin-Yuan Gao
- Department of Endocrinology, First Affiliated Hospital of Harbin Medical University, Harbin 150001, China.
| |
Collapse
|
28
|
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
|
29
|
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
|
30
|
Anderson A, Campo A, Fulton E, Corwin A, Jerome WG, O'Connor MS. 7-Ketocholesterol in disease and aging. Redox Biol 2020; 29:101380. [PMID: 31926618 PMCID: PMC6926354 DOI: 10.1016/j.redox.2019.101380] [Citation(s) in RCA: 120] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 11/06/2019] [Accepted: 11/10/2019] [Indexed: 02/08/2023] Open
Abstract
7-Ketocholesterol (7KC) is a toxic oxysterol that is associated with many diseases and disabilities of aging, as well as several orphan diseases. 7KC is the most common product of a reaction between cholesterol and oxygen radicals and is the most concentrated oxysterol found in the blood and arterial plaques of coronary artery disease patients as well as various other disease tissues and cell types. Unlike cholesterol, 7KC consistently shows cytotoxicity to cells and its physiological function in humans or other complex organisms is unknown. Oxysterols, particularly 7KC, have also been shown to diffuse through membranes where they affect receptor and enzymatic function. Here, we will explore the known and proposed mechanisms of pathologies that are associated with 7KC, as well speculate about the future of 7KC as a diagnostic and therapeutic target in medicine.
Collapse
|
31
|
Couto NF, Rezende L, Fernandes-Braga W, Alves AP, Agero U, Alvarez-Leite J, Damasceno NRT, Castro-Gomes T, Andrade LO. OxLDL alterations in endothelial cell membrane dynamics leads to changes in vesicle trafficking and increases cell susceptibility to injury. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2019; 1862:183139. [PMID: 31812625 DOI: 10.1016/j.bbamem.2019.183139] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 11/14/2019] [Accepted: 11/27/2019] [Indexed: 02/07/2023]
Abstract
Plasma membrane repair (PMR) is an important process for cell homeostasis, especially for cells under constant physical stress. Repair involves a sequence of Ca2+-dependent events, including lysosomal exocytosis and subsequent compensatory endocytosis. Cholesterol sequestration from plasma membrane causes actin cytoskeleton reorganization and polymerization, increasing cell stiffness, which leads to exocytosis and reduction of a peripheral pool of lysosomes involved in PMR. These changes in mechanical properties are similar to those observed in cells exposed to oxidized Low Density Lipoprotein (oxLDL), a key molecule during atherosclerosis development. Using a human umbilical vein endothelial cell line (EAhY926) we evaluated the influence of mechanical modulation induced by oxLDL in PMR and its effect in endothelial fragility. Similar to MβCD (a drug capable of sequestering cholesterol) treatment, oxLDL exposure led to actin reorganization and de novo polymerization, as well as an increase in cell rigidity and lysosomal exocytosis. Additionally, for both MβCD and oxLDL treated cells, there was an initial increase in endocytic events, likely triggered by the peak of exocytosis induced by both treatments. However, no further endocytic events were observed, suggesting that constitutive endocytosis is blocked upon treatment and that the reorganized cytoskeleton function as a mechanical barrier to membrane traffic. Finally, the increase in cell rigidity renders cells more prone to mechanical injury. Together, these data show that mechanical modulation induced by oxLDL exposure not only alters membrane traffic in cells, but also makes them more susceptible to mechanical injury, which may likely contribute to the initial steps of atherosclerosis development.
Collapse
Affiliation(s)
- Natália Fernanda Couto
- Department of Morphology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Luisa Rezende
- Department of Morphology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Weslley Fernandes-Braga
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Ana Paula Alves
- Department of Physics, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Ubirajara Agero
- Department of Physics, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Jacqueline Alvarez-Leite
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Thiago Castro-Gomes
- Department of Parasitology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Luciana O Andrade
- Department of Morphology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil.
| |
Collapse
|
32
|
Li L, Zhong S, Shen X, Li Q, Xu W, Tao Y, Yin H. Recent development on liquid chromatography-mass spectrometry analysis of oxidized lipids. Free Radic Biol Med 2019; 144:16-34. [PMID: 31202785 DOI: 10.1016/j.freeradbiomed.2019.06.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 05/21/2019] [Accepted: 06/05/2019] [Indexed: 12/13/2022]
Abstract
Polyunsaturated fatty acids (PUFAs) in the cellular membrane can be oxidized by various enzymes or reactive oxygen species (ROS) to form many oxidized lipids. These metabolites are highly bioactive, participating in a variety of physiological and pathophysiological processes. Mass spectrometry (MS), coupled with Liquid Chromatography, has been increasingly recognized as an indispensable tool for the analysis of oxidized lipids due to its excellent sensitivity and selectivity. We will give an update on the understanding of the molecular mechanisms related to generation of various oxidized lipids and recent progress on the development of LC-MS in the detection of these bioactive lipids derived from fatty acids, cholesterol esters, and phospholipids. The purpose of this review is to provide an overview of the formation mechanisms and technological advances in LC-MS for the study of oxidized lipids in human diseases, and to shed new light on the potential of using oxidized lipids as biomarkers and mechanistic clues of pathogenesis related to lipid metabolism. The key technical problems associated with analysis of oxidized lipids and challenges in the field will also discussed.
Collapse
Affiliation(s)
- Luxiao Li
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai, 200031, China; University of Chinese Academy of Sciences, CAS, Beijing, 100049, China
| | - Shanshan Zhong
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai, 200031, China; University of Chinese Academy of Sciences, CAS, Beijing, 100049, China
| | - Xia Shen
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai, 200031, China; University of Chinese Academy of Sciences, CAS, Beijing, 100049, China; School of Life Science and Technology, ShanghaiTech University, Shanghai, 200031, China
| | - Qiujing Li
- Department of Pharmacy, Zhangzhou Health Vocational College, Zhangzhou, 363000, China
| | - Wenxin Xu
- Department of Medical Technology, Zhangzhou Health Vocational College, Zhangzhou, 363000, China
| | - Yongzhen Tao
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai, 200031, China
| | - Huiyong Yin
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai, 200031, China; University of Chinese Academy of Sciences, CAS, Beijing, 100049, China; School of Life Science and Technology, ShanghaiTech University, Shanghai, 200031, China; Key Laboratory of Food Safety Risk Assessment, Ministry of Health, Beijing, 100000, China.
| |
Collapse
|
33
|
Chaqour B. Caught between a "Rho" and a hard place: are CCN1/CYR61 and CCN2/CTGF the arbiters of microvascular stiffness? J Cell Commun Signal 2019; 14:21-29. [PMID: 31376071 DOI: 10.1007/s12079-019-00529-3] [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: 07/11/2019] [Accepted: 07/26/2019] [Indexed: 12/18/2022] Open
Abstract
The extracellular matrix (ECM) is a deformable dynamic structure that dictates the behavior, function and integrity of blood vessels. The composition, density, chemistry and architecture of major globular and fibrillar proteins of the matrisome regulate the mechanical properties of the vasculature (i.e., stiffness/compliance). ECM proteins are linked via integrins to a protein adhesome directly connected to the actin cytoskeleton and various downstream signaling pathways that enable the cells to respond to external stimuli in a coordinated manner and maintain optimal tissue stiffness. However, cardiovascular risk factors such as diabetes, dyslipidemia, hypertension, ischemia and aging compromise the mechanical balance of the vascular wall. Stiffening of large blood vessels is associated with well-known qualitative and quantitative changes of fibrillar and fibrous macromolecules of the vascular matrisome. However, the mechanical properties of the thin-walled microvasculature are essentially defined by components of the subendothelial matrix. Cellular communication network (CCN) 1 and 2 proteins (aka Cyr61 and CTGF, respectively) of the CCN protein family localize in and act on the pericellular matrix of microvessels and constitute primary candidate markers and regulators of microvascular compliance. CCN1 and CCN2 bind various integrin and non-integrin receptors and initiate signaling pathways that regulate connective tissue remodeling and response to injury, the associated mechanoresponse of vascular cells, and the subsequent inflammatory response. The CCN1 and CCN2 genes are themselves responsive to mechanical stimuli in vascular cells, wherein mechanotransduction signaling converges into the common Rho GTPase pathway, which promotes actomyosin-based contractility and cellular stiffening. However, CCN1 and CCN2 each exhibit unique functional attributes in these processes. A better understanding of their synergistic or antagonistic effects on the maintenance (or loss) of microvascular compliance in physiological and pathological situations will assist more broadly based studies of their functional properties and translational value.
Collapse
Affiliation(s)
- Brahim Chaqour
- Department of Cell Biology and Department of Ophthalmology, State University of New York - SUNY Downstate Medical Center, 450 Clarkson Avenue, MSC 5, Brooklyn, NY, 11203, USA.
| |
Collapse
|
34
|
Liu J, Wada Y, Katsura M, Tozawa H, Erwin N, Kapron CM, Bao G, Liu J. Rho-Associated Coiled-Coil Kinase (ROCK) in Molecular Regulation of Angiogenesis. Am J Cancer Res 2018; 8:6053-6069. [PMID: 30613282 PMCID: PMC6299434 DOI: 10.7150/thno.30305] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Accepted: 10/16/2018] [Indexed: 02/06/2023] Open
Abstract
Identified as a major downstream effector of the small GTPase RhoA, Rho-associated coiled-coil kinase (ROCK) is a versatile regulator of multiple cellular processes. Angiogenesis, the process of generating new capillaries from the pre-existing ones, is required for the development of various diseases such as cancer, diabetes and rheumatoid arthritis. Recently, ROCK has attracted attention for its crucial role in angiogenesis, making it a promising target for new therapeutic approaches. In this review, we summarize recent advances in understanding the role of ROCK signaling in regulating the permeability, migration, proliferation and tubulogenesis of endothelial cells (ECs), as well as its functions in non-ECs which constitute the pro-angiogenic microenvironment. The therapeutic potential of ROCK inhibitors in angiogenesis-related diseases is also discussed.
Collapse
|
35
|
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
|
36
|
Röhrl C, Stangl H. Cholesterol metabolism-physiological regulation and pathophysiological deregulation by the endoplasmic reticulum. Wien Med Wochenschr 2018; 168:280-285. [PMID: 29488036 PMCID: PMC6132555 DOI: 10.1007/s10354-018-0626-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 01/30/2018] [Indexed: 12/22/2022]
Abstract
Cholesterol is an essential lipid for mammalian cells and its homeostasis is tightly regulated. Disturbance of cellular cholesterol homeostasis is linked to atherosclerosis and cardiovascular diseases. A central role in the sensing and regulation of cholesterol homeostasis is attributed to the endoplasmic reticulum (ER). This organelle harbours inactive transcription factors, which sense ER cholesterol levels and initiate transcriptional responses after activation and translocation into the nucleus. Thereupon, these responses enable adaption to high or low cellular cholesterol levels. Besides the abovementioned canonical functions, ER stress-induced by metabolic burden-and the resulting unfolded protein response influence cholesterol metabolism relevant to metabolic disorders. This review summarizes basic as well as recent knowledge on the role of the ER in terms of regulation of cholesterol metabolism.
Collapse
Affiliation(s)
- Clemens Röhrl
- Department of Medical Chemistry, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Währingerstraße 10, 1090, Vienna, Austria
| | - Herbert Stangl
- Department of Medical Chemistry, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Währingerstraße 10, 1090, Vienna, Austria.
| |
Collapse
|
37
|
Le Master E, Huang RT, Zhang C, Bogachkov Y, Coles C, Shentu TP, Sheng Y, Fancher IS, Ng C, Christoforidis T, Subbaiah PV, Berdyshev E, Qain Z, Eddington DT, Lee J, Cho M, Fang Y, Minshall RD, Levitan I. Proatherogenic Flow Increases Endothelial Stiffness via Enhanced CD36-Mediated Uptake of Oxidized Low-Density Lipoproteins. Arterioscler Thromb Vasc Biol 2018; 38:64-75. [PMID: 29025707 PMCID: PMC5746473 DOI: 10.1161/atvbaha.117.309907] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 09/26/2017] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Disturbed flow (DF) is well-known to induce endothelial dysfunction and synergistically with plasma dyslipidemia facilitate plaque formation. Little is known, however, about the synergistic impact of DF and dyslipidemia on endothelial biomechanics. Our goal was to determine the impact of DF on endothelial stiffness and evaluate the role of dyslipidemia/oxLDL (oxidized low-density lipoprotein) in this process. APPROACH AND RESULTS Endothelial elastic modulus of intact mouse aortas ex vivo and of human aortic endothelial cells exposed to laminar flow or DF was measured using atomic force microscopy. Endothelial monolayer of the aortic arch is found to be significantly stiffer than the descending aorta (4.2+1.1 versus 2.5+0.2 kPa for aortic arch versus descending aorta) in mice maintained on low-fat diet. This effect is significantly exacerbated by short-term high-fat diet (8.7+2.5 versus 4.5+1.2 kPa for aortic arch versus descending aorta). Exposure of human aortic endothelial cells to DF in vitro resulted in 50% increase in oxLDL uptake and significant endothelial stiffening in the presence but not in the absence of oxLDL. DF also increased the expression of oxLDL receptor CD36 (cluster of differentiation 36), whereas downregulation of CD36 abrogated DF-induced endothelial oxLDL uptake and stiffening. Furthermore, genetic deficiency of CD36 abrogated endothelial stiffening in the aortic arch in vivo in mice fed either low-fat diet or high-fat diet. We also show that the loss of endothelial stiffening in CD36 knockout aortas is not mediated by the loss of CD36 in circulating cells. CONCLUSIONS DF facilitates endothelial CD36-dependent uptake of oxidized lipids resulting in local increase of endothelial stiffness in proatherogenic areas of the aorta.
Collapse
Affiliation(s)
- Elizabeth Le Master
- From the Division of Pulmonary and Critical Care (E.L.M., C.Z., T.-P.S., I.S.F., I.L.), Division of Endocrinology (P.V.S.), Division of Hematology and Oncology, Department of Medicine (Y.S., Z.Q.), and Departments of Bioengineering (E.L.M., T.-P.S., C.N., T.C., D.T.E., J.L., M.C., I.L.), Pharmacology (Y.B., C.C., R.D.M., I.L.), and Anesthesiology (R.D.M.), University of Illinois at Chicago; Department of Medicine, University of Chicago, IL (R.-T.H., Y.F.); and Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, CO (E.B.)
| | - Ru-Ting Huang
- From the Division of Pulmonary and Critical Care (E.L.M., C.Z., T.-P.S., I.S.F., I.L.), Division of Endocrinology (P.V.S.), Division of Hematology and Oncology, Department of Medicine (Y.S., Z.Q.), and Departments of Bioengineering (E.L.M., T.-P.S., C.N., T.C., D.T.E., J.L., M.C., I.L.), Pharmacology (Y.B., C.C., R.D.M., I.L.), and Anesthesiology (R.D.M.), University of Illinois at Chicago; Department of Medicine, University of Chicago, IL (R.-T.H., Y.F.); and Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, CO (E.B.)
| | - Chongxu Zhang
- From the Division of Pulmonary and Critical Care (E.L.M., C.Z., T.-P.S., I.S.F., I.L.), Division of Endocrinology (P.V.S.), Division of Hematology and Oncology, Department of Medicine (Y.S., Z.Q.), and Departments of Bioengineering (E.L.M., T.-P.S., C.N., T.C., D.T.E., J.L., M.C., I.L.), Pharmacology (Y.B., C.C., R.D.M., I.L.), and Anesthesiology (R.D.M.), University of Illinois at Chicago; Department of Medicine, University of Chicago, IL (R.-T.H., Y.F.); and Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, CO (E.B.)
| | - Yedida Bogachkov
- From the Division of Pulmonary and Critical Care (E.L.M., C.Z., T.-P.S., I.S.F., I.L.), Division of Endocrinology (P.V.S.), Division of Hematology and Oncology, Department of Medicine (Y.S., Z.Q.), and Departments of Bioengineering (E.L.M., T.-P.S., C.N., T.C., D.T.E., J.L., M.C., I.L.), Pharmacology (Y.B., C.C., R.D.M., I.L.), and Anesthesiology (R.D.M.), University of Illinois at Chicago; Department of Medicine, University of Chicago, IL (R.-T.H., Y.F.); and Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, CO (E.B.)
| | - Cassandre Coles
- From the Division of Pulmonary and Critical Care (E.L.M., C.Z., T.-P.S., I.S.F., I.L.), Division of Endocrinology (P.V.S.), Division of Hematology and Oncology, Department of Medicine (Y.S., Z.Q.), and Departments of Bioengineering (E.L.M., T.-P.S., C.N., T.C., D.T.E., J.L., M.C., I.L.), Pharmacology (Y.B., C.C., R.D.M., I.L.), and Anesthesiology (R.D.M.), University of Illinois at Chicago; Department of Medicine, University of Chicago, IL (R.-T.H., Y.F.); and Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, CO (E.B.)
| | - Tzu-Pin Shentu
- From the Division of Pulmonary and Critical Care (E.L.M., C.Z., T.-P.S., I.S.F., I.L.), Division of Endocrinology (P.V.S.), Division of Hematology and Oncology, Department of Medicine (Y.S., Z.Q.), and Departments of Bioengineering (E.L.M., T.-P.S., C.N., T.C., D.T.E., J.L., M.C., I.L.), Pharmacology (Y.B., C.C., R.D.M., I.L.), and Anesthesiology (R.D.M.), University of Illinois at Chicago; Department of Medicine, University of Chicago, IL (R.-T.H., Y.F.); and Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, CO (E.B.)
| | - Yue Sheng
- From the Division of Pulmonary and Critical Care (E.L.M., C.Z., T.-P.S., I.S.F., I.L.), Division of Endocrinology (P.V.S.), Division of Hematology and Oncology, Department of Medicine (Y.S., Z.Q.), and Departments of Bioengineering (E.L.M., T.-P.S., C.N., T.C., D.T.E., J.L., M.C., I.L.), Pharmacology (Y.B., C.C., R.D.M., I.L.), and Anesthesiology (R.D.M.), University of Illinois at Chicago; Department of Medicine, University of Chicago, IL (R.-T.H., Y.F.); and Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, CO (E.B.)
| | - Ibra S Fancher
- From the Division of Pulmonary and Critical Care (E.L.M., C.Z., T.-P.S., I.S.F., I.L.), Division of Endocrinology (P.V.S.), Division of Hematology and Oncology, Department of Medicine (Y.S., Z.Q.), and Departments of Bioengineering (E.L.M., T.-P.S., C.N., T.C., D.T.E., J.L., M.C., I.L.), Pharmacology (Y.B., C.C., R.D.M., I.L.), and Anesthesiology (R.D.M.), University of Illinois at Chicago; Department of Medicine, University of Chicago, IL (R.-T.H., Y.F.); and Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, CO (E.B.)
| | - Carlos Ng
- From the Division of Pulmonary and Critical Care (E.L.M., C.Z., T.-P.S., I.S.F., I.L.), Division of Endocrinology (P.V.S.), Division of Hematology and Oncology, Department of Medicine (Y.S., Z.Q.), and Departments of Bioengineering (E.L.M., T.-P.S., C.N., T.C., D.T.E., J.L., M.C., I.L.), Pharmacology (Y.B., C.C., R.D.M., I.L.), and Anesthesiology (R.D.M.), University of Illinois at Chicago; Department of Medicine, University of Chicago, IL (R.-T.H., Y.F.); and Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, CO (E.B.)
| | - Theodore Christoforidis
- From the Division of Pulmonary and Critical Care (E.L.M., C.Z., T.-P.S., I.S.F., I.L.), Division of Endocrinology (P.V.S.), Division of Hematology and Oncology, Department of Medicine (Y.S., Z.Q.), and Departments of Bioengineering (E.L.M., T.-P.S., C.N., T.C., D.T.E., J.L., M.C., I.L.), Pharmacology (Y.B., C.C., R.D.M., I.L.), and Anesthesiology (R.D.M.), University of Illinois at Chicago; Department of Medicine, University of Chicago, IL (R.-T.H., Y.F.); and Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, CO (E.B.)
| | - Pappasani V Subbaiah
- From the Division of Pulmonary and Critical Care (E.L.M., C.Z., T.-P.S., I.S.F., I.L.), Division of Endocrinology (P.V.S.), Division of Hematology and Oncology, Department of Medicine (Y.S., Z.Q.), and Departments of Bioengineering (E.L.M., T.-P.S., C.N., T.C., D.T.E., J.L., M.C., I.L.), Pharmacology (Y.B., C.C., R.D.M., I.L.), and Anesthesiology (R.D.M.), University of Illinois at Chicago; Department of Medicine, University of Chicago, IL (R.-T.H., Y.F.); and Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, CO (E.B.)
| | - Evgeny Berdyshev
- From the Division of Pulmonary and Critical Care (E.L.M., C.Z., T.-P.S., I.S.F., I.L.), Division of Endocrinology (P.V.S.), Division of Hematology and Oncology, Department of Medicine (Y.S., Z.Q.), and Departments of Bioengineering (E.L.M., T.-P.S., C.N., T.C., D.T.E., J.L., M.C., I.L.), Pharmacology (Y.B., C.C., R.D.M., I.L.), and Anesthesiology (R.D.M.), University of Illinois at Chicago; Department of Medicine, University of Chicago, IL (R.-T.H., Y.F.); and Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, CO (E.B.)
| | - Zhijian Qain
- From the Division of Pulmonary and Critical Care (E.L.M., C.Z., T.-P.S., I.S.F., I.L.), Division of Endocrinology (P.V.S.), Division of Hematology and Oncology, Department of Medicine (Y.S., Z.Q.), and Departments of Bioengineering (E.L.M., T.-P.S., C.N., T.C., D.T.E., J.L., M.C., I.L.), Pharmacology (Y.B., C.C., R.D.M., I.L.), and Anesthesiology (R.D.M.), University of Illinois at Chicago; Department of Medicine, University of Chicago, IL (R.-T.H., Y.F.); and Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, CO (E.B.)
| | - David T Eddington
- From the Division of Pulmonary and Critical Care (E.L.M., C.Z., T.-P.S., I.S.F., I.L.), Division of Endocrinology (P.V.S.), Division of Hematology and Oncology, Department of Medicine (Y.S., Z.Q.), and Departments of Bioengineering (E.L.M., T.-P.S., C.N., T.C., D.T.E., J.L., M.C., I.L.), Pharmacology (Y.B., C.C., R.D.M., I.L.), and Anesthesiology (R.D.M.), University of Illinois at Chicago; Department of Medicine, University of Chicago, IL (R.-T.H., Y.F.); and Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, CO (E.B.)
| | - James Lee
- From the Division of Pulmonary and Critical Care (E.L.M., C.Z., T.-P.S., I.S.F., I.L.), Division of Endocrinology (P.V.S.), Division of Hematology and Oncology, Department of Medicine (Y.S., Z.Q.), and Departments of Bioengineering (E.L.M., T.-P.S., C.N., T.C., D.T.E., J.L., M.C., I.L.), Pharmacology (Y.B., C.C., R.D.M., I.L.), and Anesthesiology (R.D.M.), University of Illinois at Chicago; Department of Medicine, University of Chicago, IL (R.-T.H., Y.F.); and Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, CO (E.B.)
| | - Michael Cho
- From the Division of Pulmonary and Critical Care (E.L.M., C.Z., T.-P.S., I.S.F., I.L.), Division of Endocrinology (P.V.S.), Division of Hematology and Oncology, Department of Medicine (Y.S., Z.Q.), and Departments of Bioengineering (E.L.M., T.-P.S., C.N., T.C., D.T.E., J.L., M.C., I.L.), Pharmacology (Y.B., C.C., R.D.M., I.L.), and Anesthesiology (R.D.M.), University of Illinois at Chicago; Department of Medicine, University of Chicago, IL (R.-T.H., Y.F.); and Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, CO (E.B.)
| | - Yun Fang
- From the Division of Pulmonary and Critical Care (E.L.M., C.Z., T.-P.S., I.S.F., I.L.), Division of Endocrinology (P.V.S.), Division of Hematology and Oncology, Department of Medicine (Y.S., Z.Q.), and Departments of Bioengineering (E.L.M., T.-P.S., C.N., T.C., D.T.E., J.L., M.C., I.L.), Pharmacology (Y.B., C.C., R.D.M., I.L.), and Anesthesiology (R.D.M.), University of Illinois at Chicago; Department of Medicine, University of Chicago, IL (R.-T.H., Y.F.); and Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, CO (E.B.)
| | - Richard D Minshall
- From the Division of Pulmonary and Critical Care (E.L.M., C.Z., T.-P.S., I.S.F., I.L.), Division of Endocrinology (P.V.S.), Division of Hematology and Oncology, Department of Medicine (Y.S., Z.Q.), and Departments of Bioengineering (E.L.M., T.-P.S., C.N., T.C., D.T.E., J.L., M.C., I.L.), Pharmacology (Y.B., C.C., R.D.M., I.L.), and Anesthesiology (R.D.M.), University of Illinois at Chicago; Department of Medicine, University of Chicago, IL (R.-T.H., Y.F.); and Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, CO (E.B.)
| | - Irena Levitan
- From the Division of Pulmonary and Critical Care (E.L.M., C.Z., T.-P.S., I.S.F., I.L.), Division of Endocrinology (P.V.S.), Division of Hematology and Oncology, Department of Medicine (Y.S., Z.Q.), and Departments of Bioengineering (E.L.M., T.-P.S., C.N., T.C., D.T.E., J.L., M.C., I.L.), Pharmacology (Y.B., C.C., R.D.M., I.L.), and Anesthesiology (R.D.M.), University of Illinois at Chicago; Department of Medicine, University of Chicago, IL (R.-T.H., Y.F.); and Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, CO (E.B.).
| |
Collapse
|
38
|
Zhang C, Adamos C, Oh MJ, Baruah J, Ayee MAA, Mehta D, Wary KK, Levitan I. oxLDL induces endothelial cell proliferation via Rho/ROCK/Akt/p27 kip1 signaling: opposite effects of oxLDL and cholesterol loading. Am J Physiol Cell Physiol 2017; 313:C340-C351. [PMID: 28701359 DOI: 10.1152/ajpcell.00249.2016] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 07/05/2017] [Accepted: 07/05/2017] [Indexed: 12/19/2022]
Abstract
Oxidized modifications of LDL (oxLDL) play a key role in the development of endothelial dysfunction and atherosclerosis. However, the underlying mechanisms of oxLDL-mediated cellular behavior are not completely understood. Here, we compared the effects of two major types of oxLDL, copper-oxidized LDL (Cu2+-oxLDL) and lipoxygenase-oxidized LDL (LPO-oxLDL), on proliferation of human aortic endothelial cells (HAECs). Cu2+-oxLDL enhanced HAECs' proliferation in a dose- and degree of oxidation-dependent manner. Similarly, LPO-oxLDL also enhanced HAEC proliferation. Mechanistically, both Cu2+-oxLDL and LPO-oxLDL enhance HAEC proliferation via activation of Rho, Akt phosphorylation, and a decrease in the expression of cyclin-dependent kinase inhibitor 1B (p27kip1). Both Cu2+-oxLDL or LPO-oxLDL significantly increased Akt phosphorylation, whereas an Akt inhibitor, MK2206, blocked oxLDL-induced increase in HAEC proliferation. Blocking Rho with C3 or its downstream target ROCK with Y27632 significantly inhibited oxLDL-induced Akt phosphorylation and proliferation mediated by both Cu2+- and LPO-oxLDL. Activation of RhoA was blocked by Rho-GDI-1, which also abrogated oxLDL-induced Akt phosphorylation and HAEC proliferation. In contrast, blocking Rac1 in these cells had no effect on oxLDL-induced Akt phosphorylation or cell proliferation. Moreover, oxLDL-induced Rho/Akt signaling downregulated cell cycle inhibitor p27kip1 Preloading these cells with cholesterol, however, prevented oxLDL-induced Akt phosphorylation and HAEC proliferation. These findings provide a new understanding of the effects of oxLDL on endothelial proliferation, which is essential for developing new treatments against neovascularization and progression of atherosclerosis.
Collapse
Affiliation(s)
- Chongxu Zhang
- Division of Pulmonary and Critical Care, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; and
| | - Crystal Adamos
- Division of Pulmonary and Critical Care, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; and
| | - Myung-Jin Oh
- Division of Pulmonary and Critical Care, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; and
| | - Jugajyoti Baruah
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois
| | - Manuela A A Ayee
- Division of Pulmonary and Critical Care, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; and
| | - Dolly Mehta
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois
| | - Kishore K Wary
- Department of Pharmacology, 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; and
| |
Collapse
|
39
|
Ayee MAA, LeMaster E, Shentu TP, Singh DK, Barbera N, Soni D, Tiruppathi C, Subbaiah PV, Berdyshev E, Bronova I, Cho M, Akpa BS, Levitan I. Molecular-Scale Biophysical Modulation of an Endothelial Membrane by Oxidized Phospholipids. Biophys J 2017; 112:325-338. [PMID: 28122218 DOI: 10.1016/j.bpj.2016.12.002] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 11/29/2016] [Accepted: 12/01/2016] [Indexed: 12/31/2022] Open
Abstract
The influence of two bioactive oxidized phospholipids on model bilayer properties, membrane packing, and endothelial cell biomechanics was investigated computationally and experimentally. The truncated tail phospholipids, 1-palmitoyl-2-(5-oxovaleroyl)-sn-glycero-3-phosphocholine (POVPC) and 1-palmitoyl-2-glutaroyl-sn-glycero-3-phosphocholine (PGPC), are two major oxidation products of the unsaturated phospholipid 1-palmitoyl-2-arachidonoyl-sn-glycero-phosphocholine. A combination of coarse-grained molecular dynamics simulations, Laurdan multiphoton imaging, and atomic force microscopy microindentation experiments was used to determine the impact of POVPC and PGPC on the structure of a multicomponent phospholipid bilayer and to assess the consequences of their incorporation on membrane packing and endothelial cell stiffness. Molecular simulations predicted differential bilayer perturbation effects of the two oxidized phospholipids based on the chemical identities of their truncated tails, including decreased bilayer packing, decreased bilayer bending modulus, and increased water penetration. Disruption of lipid order was consistent with Laurdan imaging results indicating that POVPC and PGPC decrease the lipid packing of both ordered and disordered membrane domains. Computational predictions of a larger membrane perturbation effect by PGPC correspond to greater stiffness of PGPC-treated endothelial cells observed by measuring cellular elastic moduli using atomic force microscopy. Our results suggest that disruptions in membrane structure by oxidized phospholipids play a role in the regulation of overall endothelial cell stiffness.
Collapse
Affiliation(s)
- Manuela A A Ayee
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Elizabeth LeMaster
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Tzu Pin Shentu
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Dev K Singh
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Nicolas Barbera
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Dheeraj Soni
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois
| | | | - Papasani V Subbaiah
- Division of Endocrinology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | | | | | - Michael Cho
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois
| | - Belinda S Akpa
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, North Carolina
| | - Irena Levitan
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois.
| |
Collapse
|
40
|
Vascular endothelial growth factor modified macrophages transdifferentiate into endothelial-like cells and decrease foam cell formation. Biosci Rep 2017; 37:BSR20170002. [PMID: 28536311 PMCID: PMC5479018 DOI: 10.1042/bsr20170002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 05/20/2017] [Accepted: 05/22/2017] [Indexed: 12/30/2022] Open
Abstract
Macrophages are largely involved in the whole process of atherosclerosis from an initiation lesion to an advanced lesion. Endothelial disruption is the initial step and macrophage-derived foam cells are the hallmark of atherosclerosis. Promotion of vascular integrity and inhibition of foam cell formation are two important strategies for preventing atherosclerosis. How can we inhibit even the reverse negative role of macrophages in atherosclerosis? The present study was performed to investigate if overexpressing endogenous human vascular endothelial growth factor (VEGF) could facilitate transdifferentiation of macrophages into endothelial-like cells (ELCs) and inhibit foam cell formation. We demonstrated that VEGF-modified macrophages which stably overexpressed human VEGF (hVEGF165) displayed a high capability to alter their phenotype and function into ELCs in vitro. Exogenous VEGF could not replace endogenous VEGF to induce the transdifferentiation of macrophages into ELCs in vitro. We further showed that VEGF-modified macrophages significantly decreased cytoplasmic lipid accumulation after treatment with oxidized LDL (ox-LDL). Moreover, down-regulation of CD36 expression in these cells was probably one of the mechanisms of reduction in foam cell formation. Our results provided the in vitro proof of VEGF-modified macrophages as atheroprotective therapeutic cells by both promotion of vascular repair and inhibition of foam cell formation.
Collapse
|
41
|
Miller YI, Shyy JYJ. Context-Dependent Role of Oxidized Lipids and Lipoproteins in Inflammation. Trends Endocrinol Metab 2017; 28:143-152. [PMID: 27931771 PMCID: PMC5253098 DOI: 10.1016/j.tem.2016.11.002] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 10/26/2016] [Accepted: 11/02/2016] [Indexed: 01/13/2023]
Abstract
Oxidized low-density lipoprotein (OxLDL), which contains hundreds of different oxidized lipid molecules, is a hallmark of hyperlipidemia and atherosclerosis. The same oxidized lipids found in OxLDL are also formed in apoptotic cells, and are present in tissues as well as in the circulation under pathological conditions. In many disease contexts, oxidized lipids constitute damage signals, or patterns, that activate pattern-recognition receptors (PRRs) and significantly contribute to inflammation. Here, we review recent discoveries and emerging trends in the field of oxidized lipids and the regulation of inflammation, focusing on oxidation products of polyunsaturated fatty acids esterified into cholesteryl esters (CEs) and phospholipids (PLs). We also highlight context-dependent activation and biased agonism of Toll-like receptor-4 (TLR4) and the NLRP3 inflammasome, among other signaling pathways activated by oxidized lipids.
Collapse
Affiliation(s)
- Yury I Miller
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA.
| | - John Y-J Shyy
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
42
|
Li GY, Wang YJ, Zhao TJ, Peng Y. Signaling pathways related to role of hepatic sinusoidal endothelial cells in liver fibrosis. Shijie Huaren Xiaohua Zazhi 2016; 24:3933-3939. [DOI: 10.11569/wcjd.v24.i28.3933] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Liver fibrosis is the pathological preprocess of liver cirrhosis or liver cancer progressing from chronic liver disease. Hepatic sinusoidal endothelial cells (HSECs) are involved in the formation and development of liver fibrosis through multiple signaling pathways. In this paper, we summarize and elaborate these signaling pathways including Rho-GTPase, CXCR7-Id1/FGFR1-CXCR4, VEGFR-2/p38 mitogen-activated protein kinases (MAPK), MAPK, and TLRs. Based on these signaling pathways, we put forward new ideas for the prevention and treatment of liver fibrosis.
Collapse
|
43
|
Choi SH, Sviridov D, Miller YI. Oxidized cholesteryl esters and inflammation. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1862:393-397. [PMID: 27368140 DOI: 10.1016/j.bbalip.2016.06.020] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 06/08/2016] [Accepted: 06/24/2016] [Indexed: 11/30/2022]
Abstract
The oxidation hypothesis of atherosclerosis proposes that oxidized LDL is a major causative factor in the development of atherosclerosis. Although this hypothesis has received strong mechanistic support and many animal studies demonstrated profound atheroprotective effects of antioxidants, which reduce LDL oxidation, the results of human clinical trials with antioxidants were mainly negative, except in selected groups of patients with clearly increased systemic oxidative stress. We propose that even if reducing lipoprotein oxidation in humans might be difficult to achieve, deeper understanding of mechanisms by which oxidized LDL promotes atherosclerosis and targeting these specific mechanisms will offer novel approaches to treatment of cardiovascular disease. In this review article, we focus on oxidized cholesteryl esters (OxCE), which are a major component of minimally and extensively oxidized LDL and of human atherosclerotic lesions. OxCE and OxCE-protein covalent adducts induce profound biological effects. Among these effects, OxCE activate macrophages via toll-like receptor-4 (TLR4) and spleen tyrosine kinase and induce macropinocytosis resulting in lipid accumulation, generation of reactive oxygen species and secretion of inflammatory cytokines. Specific inhibition of OxCE-induced TLR4 activation, as well as blocking other inflammatory effects of OxCE, may offer novel treatments of atherosclerosis and cardiovascular disease. This article is part of a Special Issue entitled: Lipid modification and lipid peroxidation products in innate immunity and inflammation edited by Christoph J. Binder.
Collapse
Affiliation(s)
- Soo-Ho Choi
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Dmitri Sviridov
- Baker IDI Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
| | - Yury I Miller
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
44
|
Sobrevia L, Salsoso R, Fuenzalida B, Barros E, Toledo L, Silva L, Pizarro C, Subiabre M, Villalobos R, Araos J, Toledo F, González M, Gutiérrez J, Farías M, Chiarello DI, Pardo F, Leiva A. Insulin Is a Key Modulator of Fetoplacental Endothelium Metabolic Disturbances in Gestational Diabetes Mellitus. Front Physiol 2016; 7:119. [PMID: 27065887 PMCID: PMC4815008 DOI: 10.3389/fphys.2016.00119] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 03/15/2016] [Indexed: 12/11/2022] Open
Abstract
Gestational diabetes mellitus (GDM) is a disease of the mother that associates with altered fetoplacental vascular function. GDM-associated maternal hyperglycaemia result in fetal hyperglycaemia, a condition that leads to fetal hyperinsulinemia and altered L-arginine transport and synthesis of nitric oxide, i.e., endothelial dysfunction. These alterations in the fetoplacental endothelial function are present in women with GDM that were under diet or insulin therapy. Since these women and their newborn show normal glycaemia at term, other factors or conditions could be altered and/or not resolved by restoring normal level of circulating D-glucose. GDM associates with metabolic disturbances, such as abnormal handling of the locally released vasodilator adenosine, and biosynthesis and metabolism of cholesterol lipoproteins, or metabolic diseases resulting in endoplasmic reticulum stress and altered angiogenesis. Insulin acts as a potent modulator of all these phenomena under normal conditions as reported in primary cultures of cells obtained from the human placenta; however, GDM and the role of insulin regarding these alterations in this disease are poorly understood. This review focuses on the potential link between insulin and endoplasmic reticulum stress, hypercholesterolemia, and angiogenesis in GDM in the human fetoplacental vasculature. Based in reports in primary culture placental endothelium we propose that insulin is a factor restoring endothelial function in GDM by reversing ERS, hypercholesterolaemia and angiogenesis to a physiological state involving insulin activation of insulin receptor isoforms and adenosine receptors and metabolism in the human placenta from GDM pregnancies.
Collapse
Affiliation(s)
- Luis Sobrevia
- Cellular and Molecular Physiology Laboratory, Division of Obstetrics and Gynecology, Faculty of Medicine, School of Medicine, Pontificia Universidad Católica de ChileSantiago, Chile; Faculty of Medicine and Biomedical Sciences, University of Queensland Centre for Clinical Research, University of QueenslandHerston, QLD, Australia; Department of Physiology, Faculty of Pharmacy, Universidad de SevillaSeville, Spain
| | - Rocío Salsoso
- Cellular and Molecular Physiology Laboratory, Division of Obstetrics and Gynecology, Faculty of Medicine, School of Medicine, Pontificia Universidad Católica de ChileSantiago, Chile; Department of Physiology, Faculty of Pharmacy, Universidad de SevillaSeville, Spain
| | - Bárbara Fuenzalida
- Cellular and Molecular Physiology Laboratory, Division of Obstetrics and Gynecology, Faculty of Medicine, School of Medicine, Pontificia Universidad Católica de Chile Santiago, Chile
| | - Eric Barros
- Cellular and Molecular Physiology Laboratory, Division of Obstetrics and Gynecology, Faculty of Medicine, School of Medicine, Pontificia Universidad Católica de Chile Santiago, Chile
| | - Lilian Toledo
- Cellular and Molecular Physiology Laboratory, Division of Obstetrics and Gynecology, Faculty of Medicine, School of Medicine, Pontificia Universidad Católica de Chile Santiago, Chile
| | - Luis Silva
- Cellular and Molecular Physiology Laboratory, Division of Obstetrics and Gynecology, Faculty of Medicine, School of Medicine, Pontificia Universidad Católica de Chile Santiago, Chile
| | - Carolina Pizarro
- Cellular and Molecular Physiology Laboratory, Division of Obstetrics and Gynecology, Faculty of Medicine, School of Medicine, Pontificia Universidad Católica de Chile Santiago, Chile
| | - Mario Subiabre
- Cellular and Molecular Physiology Laboratory, Division of Obstetrics and Gynecology, Faculty of Medicine, School of Medicine, Pontificia Universidad Católica de Chile Santiago, Chile
| | - Roberto Villalobos
- Cellular and Molecular Physiology Laboratory, Division of Obstetrics and Gynecology, Faculty of Medicine, School of Medicine, Pontificia Universidad Católica de Chile Santiago, Chile
| | - Joaquín Araos
- Cellular and Molecular Physiology Laboratory, Division of Obstetrics and Gynecology, Faculty of Medicine, School of Medicine, Pontificia Universidad Católica de Chile Santiago, Chile
| | - Fernando Toledo
- Department of Basic Sciences, Faculty of Sciences, Universidad del Bío-Bío Chillán, Chile
| | - Marcelo González
- Vascular Physiology Laboratory, Department of Physiology, Faculty of Biological Sciences, Universidad de ConcepciónConcepción, Chile; Group of Research and Innovation in Vascular Health (GRIVAS-Health)Chillán, Chile
| | - Jaime Gutiérrez
- Cellular and Molecular Physiology Laboratory, Division of Obstetrics and Gynecology, Faculty of Medicine, School of Medicine, Pontificia Universidad Católica de ChileSantiago, Chile; Cellular Signaling and Differentiation Laboratory, Health Sciences Faculty, Universidad San SebastiánSantiago, Chile
| | - Marcelo Farías
- Cellular and Molecular Physiology Laboratory, Division of Obstetrics and Gynecology, Faculty of Medicine, School of Medicine, Pontificia Universidad Católica de Chile Santiago, Chile
| | - Delia I Chiarello
- Cellular and Molecular Physiology Laboratory, Division of Obstetrics and Gynecology, Faculty of Medicine, School of Medicine, Pontificia Universidad Católica de Chile Santiago, Chile
| | - Fabián Pardo
- Cellular and Molecular Physiology Laboratory, Division of Obstetrics and Gynecology, Faculty of Medicine, School of Medicine, Pontificia Universidad Católica de Chile Santiago, Chile
| | - Andrea Leiva
- Cellular and Molecular Physiology Laboratory, Division of Obstetrics and Gynecology, Faculty of Medicine, School of Medicine, Pontificia Universidad Católica de Chile Santiago, Chile
| |
Collapse
|