1
|
Kempegowda SN, Sugur K, Thimmulappa RK. Dysfunctional HDL Diagnostic Metrics for Cardiovascular Disease Risk Stratification: Are we Ready to Implement in Clinics? J Cardiovasc Transl Res 2024:10.1007/s12265-024-10559-x. [PMID: 39298091 DOI: 10.1007/s12265-024-10559-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 09/09/2024] [Indexed: 09/21/2024]
Abstract
Epidemiological studies have revealed that patients with higher levels of high-density lipoprotein cholesterol (HDL-C) were more resistant to cardiovascular diseases (CVD), and yet targeting HDL for CVD prevention, risk assessment, and pharmacological management has not proven to be very effective. The mechanistic investigations have demonstrated that HDL exerts anti-atherogenic functions via mediating reverse cholesterol transport, antioxidant action, anti-inflammatory activity, and anti-thrombotic activity. Contrary to expectations, however, adverse cardiovascular events were reported in clinical trials of drugs that raised HDL levels. This has sparked a debate between HDL quantity and quality. Patients with atherosclerotic CVD are associated with dysfunctional HDL, and the degree of HDL dysfunction is correlated with the severity of the disease, independent of HDL-C levels. This growing body of evidence has underscored the need for integrating HDL functional assays in clinical practice for CVD risk management. Because HDL exerts diverse athero-protective functions, there is no single method for capturing HDL functionality. This review critically evaluates the various techniques currently being used for monitoring HDL functionality and discusses key structural changes in HDL indicative of dysfunctional HDL and the technical challenges that need to be addressed to enable the integration of HDL function-based metrics in clinical practice for CVD risk estimation and the development of newer therapies targeting HDL function.
Collapse
Affiliation(s)
- Swetha N Kempegowda
- Department of Biochemistry, Centre of Excellence in Molecular Biology & Regenerative Medicine, JSS Medical College, JSS Academy of Higher Education & Research, Mysore, Karnataka, 570015, India
| | - Kavya Sugur
- Department of Biochemistry, Centre of Excellence in Molecular Biology & Regenerative Medicine, JSS Medical College, JSS Academy of Higher Education & Research, Mysore, Karnataka, 570015, India
| | - Rajesh K Thimmulappa
- Department of Biochemistry, Centre of Excellence in Molecular Biology & Regenerative Medicine, JSS Medical College, JSS Academy of Higher Education & Research, Mysore, Karnataka, 570015, India.
| |
Collapse
|
2
|
Berna-Rico E, Abbad-Jaime de Aragon C, Ballester-Martinez A, Perez-Bootello J, Solis J, Fernandez-Friera L, Llamas-Velasco M, Castellanos-Gonzalez M, Barderas MG, Azcarraga-Llobet C, Garcia-Mouronte E, de Nicolas-Ruanes B, Naharro-Rodriguez J, Jaen-Olasolo P, Gelfand JM, Mehta NN, Gonzalez-Cantero A. Monocyte-to-High-Density Lipoprotein Ratio Is Associated with Systemic Inflammation, Insulin Resistance, and Coronary Subclinical Atherosclerosis in Psoriasis: Results from 2 Observational Cohorts. J Invest Dermatol 2024; 144:2002-2012.e2. [PMID: 38460808 DOI: 10.1016/j.jid.2024.02.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 02/01/2024] [Accepted: 02/12/2024] [Indexed: 03/11/2024]
Abstract
Systemic inflammation or insulin resistance drive atherosclerosis. However, they are difficult to capture for assessing cardiovascular risk in clinical settings. The monocyte-to-high-density lipoprotein ratio (MHR) is an accessible biomarker that integrates inflammatory and metabolic information and has been associated with poorer cardiovascular outcomes. Our aim was to evaluate the association of MHR with the presence of subclinical atherosclerosis in patients with psoriasis. The study involved a European and an American cohort including 405 patients with the disease. Subclinical atherosclerosis was assessed by coronary computed tomography angiography. First, MHR correlated with insulin resistance through homeostatic model assessment for insulin resistance, with high-sensitivity CRP and with 18F-fluorodeoxyglucose uptake in spleen, liver, and bone marrow by positron emission tomography/computed tomography. MHR was associated with both the presence of coronary plaques >50% of the artery lumen and noncalcified coronary burden, beyond traditional cardiovascular risk factors (P < .05). In a noncalcified coronary burden prediction model accounting for cardiovascular risk factors, statins, and biologic treatment, MHR added value (area under the curve base model = 0.72 vs area under the curve base model plus MHR = 0.76, P = .04) within the American cohort. These results suggests that MHR may detect patients with psoriasis who have subclinical burden of cardiovascular disease and warrant more aggressive measures to reduce lifetime adverse cardiovascular outcomes.
Collapse
Affiliation(s)
- Emilio Berna-Rico
- Department of Dermatology, Hospital Universitario Ramon y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain.
| | - Carlota Abbad-Jaime de Aragon
- Department of Dermatology, Hospital Universitario Ramon y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Asuncion Ballester-Martinez
- Department of Dermatology, Hospital Universitario Ramon y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Javier Perez-Bootello
- Department of Dermatology, Hospital Universitario Ramon y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Jorge Solis
- Department of Cardiology, Hospital Universitario 12 de Octubre, Madrid, Spain; Department of Cardiology, Atria Clinic, Madrid, Spain; Centro Integral de Enfermedades Cardiovasculares (CIEC), Hospital Universitario HM Montepríncipe, HM Hospitales, Madrid, Spain; Facultad HM Hospitales de Ciencias de la Salud, Universidad Camilo José Cela, Madrid, Spain; CIBER de Enfermedades CardioVasculares (CIBERCV), Madrid, Spain
| | - Leticia Fernandez-Friera
- Department of Cardiology, Atria Clinic, Madrid, Spain; Centro Integral de Enfermedades Cardiovasculares (CIEC), Hospital Universitario HM Montepríncipe, HM Hospitales, Madrid, Spain; Facultad HM Hospitales de Ciencias de la Salud, Universidad Camilo José Cela, Madrid, Spain; CIBER de Enfermedades CardioVasculares (CIBERCV), Madrid, Spain
| | - Mar Llamas-Velasco
- Department of Dermatology, Hospital Universitario La Princesa, Madrid, Spain
| | | | - Maria G Barderas
- Department of Vascular Physiopathology, Hospital Nacional de Parapléjicos, Servicio de Salud de Castilla-La Mancha (SESCAM), Toledo, Spain
| | - Carlos Azcarraga-Llobet
- Department of Dermatology, Hospital Universitario Ramon y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Emilio Garcia-Mouronte
- Department of Dermatology, Hospital Universitario Ramon y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Belen de Nicolas-Ruanes
- Department of Dermatology, Hospital Universitario Ramon y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Jorge Naharro-Rodriguez
- Department of Dermatology, Hospital Universitario Ramon y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Pedro Jaen-Olasolo
- Department of Dermatology, Hospital Universitario Ramon y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Joel M Gelfand
- Department of Dermatology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA; Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Nehal N Mehta
- Department of Cardiology, George Washington Medical Center, Washington, District of Columbia, USA
| | - Alvaro Gonzalez-Cantero
- Department of Dermatology, Hospital Universitario Ramon y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain; Faculty of Medicine, Universidad Francisco de Vitoria, Madrid, Spain.
| |
Collapse
|
3
|
Namous H, Strillacci MG, Braz CU, Shanmuganayagam D, Krueger C, Peppas A, Soffregen WC, Reed J, Granada JF, Khatib H. ITGB2 is a central hub-gene associated with inflammation and early fibro-atheroma development in a swine model of atherosclerosis. ATHEROSCLEROSIS PLUS 2023; 54:30-41. [PMID: 38116576 PMCID: PMC10728570 DOI: 10.1016/j.athplu.2023.11.001] [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: 05/12/2023] [Revised: 09/14/2023] [Accepted: 11/09/2023] [Indexed: 12/21/2023]
Abstract
Background and aim The complex dynamic interplay between different biological pathways involved in atherosclerosis development has rendered the identification of specific therapeutic targets a challenging quest. We aimed to identify specific genes and mechanistic pathways associated with the early development of fibro-atheromas in a swine model of atherosclerosis. Methods The Wisconsin Miniature Swine™ model of Familial Hypercholesterolemia (WMS-FH, n = 11) and genetically related WMS controls (WMS-N, n = 11) were used. The infrarenal aorta was harvested from both groups for histopathologic and transcriptomic profiling at 12 months. Bioinformatic analysis was performed to identify hub genes and pathways central to disease pathophysiology. The expression of ITGB2, the top ranked hub gene, was manipulated in cell culture and the expression of interconnected genes was tested. Results Fibro-atheromatous lesions were documented in all WMS-FH aortic tissues and displayed internal elastic lamina (IEL) disruption, significant reduction of myofibroblast presence and disorganized collagen deposition. No fibro-atheromas were observed in the control group. A total of 266 differentially expressed genes (DEGs) were upregulated in WMS-FH aortic tissues, while 29 genes were downregulated. Top identified hub genes included ITGB2, C1QA, LCP2, SPI1, CSF1R, C5AR1, CTSS, MPEG1, C1QC, and CSF2RB. Overexpression of ITGB2 resulted in elevated expression of other interconnected genes expressed in porcine endothelial cells. Conclusion In a swine translational model of atherosclerosis, transcriptomic analysis identified ITGB2 as a central hub gene associated inflammation and early fibroatheroma development making it a potential therapeutic target at this stage of disease.
Collapse
Affiliation(s)
- Hadjer Namous
- Department of Animal and Dairy Sciences – University of Wisconsin Madison, WI, USA
| | | | - Camila Urbano Braz
- Department of Animal and Dairy Sciences – University of Wisconsin Madison, WI, USA
| | | | - Christian Krueger
- Department of Animal and Dairy Sciences – University of Wisconsin Madison, WI, USA
| | - Athanasios Peppas
- Skirball Center for Innovation, Cardiovascular Research Foundation, New York, NY, USA
| | - William C. Soffregen
- Northstar Preclinical and Pathology Services, LLC and Skirball Center for Innovation, Cardiovascular Research Foundation, New York, NY, USA
| | - Jess Reed
- Department of Animal and Dairy Sciences – University of Wisconsin Madison, WI, USA
| | - Juan F. Granada
- Skirball Center for Innovation, Cardiovascular Research Foundation, New York, NY, USA
| | - Hasan Khatib
- Department of Animal and Dairy Sciences – University of Wisconsin Madison, WI, USA
| |
Collapse
|
4
|
Zou D, Yang P, Liu J, Dai F, Xiao Y, Zhao A, Huang N. Constructing Mal-Efferocytic Macrophage Model and Its Atherosclerotic Spheroids and Rat Model for Therapeutic Evaluation. Adv Biol (Weinh) 2023; 7:e2200277. [PMID: 36721069 DOI: 10.1002/adbi.202200277] [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: 10/11/2022] [Revised: 11/27/2022] [Indexed: 02/02/2023]
Abstract
Efferocytosis, responsible for apoptotic cell clearance, is an essential factor against atherosclerosis. It is reported that efferocytosis is severely impaired in fibroatheroma, especially in vulnerable thin cap fibroatheroma. However, there is a shortage of studies on efferocytosis defects in cell and animal models. Here, the impacts of oxidized low density lipoprotein (ox-LDL) and glut 1 inhibitor (STF31) on efferocytosis of macrophages are studied, and an evaluation system is constructed. Through regulating the cell ratios and stimulus, three types of atherosclerotic spheroids are fabricated, and a necrotic core emerges with surrounding apoptotic cells. Rat models present a similar phenomenon in that substantial apoptotic cells are uncleared in time in vulnerable plaque, and the model period is shortened to 7 weeks. Mechanism studies reveal that ox-LDL, through mRNA and miRNA modulation, downregulates efferocytosis receptor (PPARγ/LXRα/MerTK), internalization molecule (SLC29a1), and upregulates the competitive receptor CD300a that inhibits efferocytosis receptor-ligand binding process. The foam cell differentiation has also confirmed that CD36 and Lp-PLA2 levels are significantly elevated, and macrophages present an interesting transition into prothrombic phenotype. Collectively, the atherosclerotic models featured by efferocytosis defect provide a comprehensive platform to evaluate the efficacy of medicine and biomaterials for atherosclerosis treatment.
Collapse
Affiliation(s)
- Dan Zou
- Key Laboratory for Advanced Technologies of Materials, Ministry of Education, Chengdu, 610031, P. R. China
- School of Material Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| | - Ping Yang
- Key Laboratory for Advanced Technologies of Materials, Ministry of Education, Chengdu, 610031, P. R. China
- School of Material Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| | - Jianan Liu
- Key Laboratory for Advanced Technologies of Materials, Ministry of Education, Chengdu, 610031, P. R. China
- School of Material Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| | - Fanfan Dai
- Key Laboratory for Advanced Technologies of Materials, Ministry of Education, Chengdu, 610031, P. R. China
- School of Material Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| | - Yangyang Xiao
- Key Laboratory for Advanced Technologies of Materials, Ministry of Education, Chengdu, 610031, P. R. China
- School of Material Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| | - Ansha Zhao
- Key Laboratory for Advanced Technologies of Materials, Ministry of Education, Chengdu, 610031, P. R. China
- School of Material Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| | - Nan Huang
- Key Laboratory for Advanced Technologies of Materials, Ministry of Education, Chengdu, 610031, P. R. China
- School of Material Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| |
Collapse
|
5
|
Guo X, Ma L. Inflammation in coronary artery disease-clinical implications of novel HDL-cholesterol-related inflammatory parameters as predictors. Coron Artery Dis 2023; 34:66-77. [PMID: 36317383 PMCID: PMC9742007 DOI: 10.1097/mca.0000000000001198] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 09/25/2022] [Indexed: 12/13/2022]
Abstract
Coronary artery disease (CAD) is the leading cause of death worldwide. Inflammation and atherosclerotic plaques are the primary pathological mechanisms of CAD. Upon stimulation by deposited lipids and damaged endothelium, innate and adaptive immune cells are activated and recruited to initiate plaque development. Therefore, inflammatory cells and mediators are used to identify inflammatory risk in CAD patients. HDL-cholesterol (HDL-C) is demonstrated to have anti-inflammatory roles in atherosclerosis by interfering with plasma membrane lipid rafts of immune cells. Based on this, novel inflammatory parameters such as monocyte to HDL-C ratio are explored to improve the risk estimation of CAD prognosis. Moreover, with the advance in treatment strategies targeting the inflammatory process in atherosclerosis, identifying CAD patients with increased inflammatory risk by novel inflammatory parameters is of great importance in guiding CAD management. Therefore, this review aims to summarize the current information regarding inflammatory activation and HDL-C in atherosclerosis with a particular focus on the clinical implication of the novel HDL-C-related inflammatory parameters in CAD.
Collapse
Affiliation(s)
- Xuantong Guo
- National Clinical Research Center for Cardiovascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lihong Ma
- National Clinical Research Center for Cardiovascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
6
|
Yang Y, Ding X, Chen F, Wu X, Chen Y, Zhang Q, Cao J, Wang J, Dai Y. Inhibition Effects of Nippostrongylus brasiliensis and Its Derivatives against Atherosclerosis in ApoE-/- Mice through Anti-Inflammatory Response. Pathogens 2022; 11:pathogens11101208. [PMID: 36297265 PMCID: PMC9610917 DOI: 10.3390/pathogens11101208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/18/2022] [Accepted: 10/18/2022] [Indexed: 11/16/2022] Open
Abstract
Atherosclerosis (AS) is a dominant and growing cause of death and disability worldwide that involves inflammation from its inception to the emergence of complications. Studies have demonstrated that intervention with helminth infections or derived products could modulate the host immune response and effectively prevent or mitigate the onset and progression of inflammation-related diseases. Therefore, to understand the molecular mechanisms underlying the development of atherosclerosis, we intervened in ApoE-/- mice maintained on a high-fat diet with Nippostrongylus brasiliensis (N. brasiliensis) infection and immunized with its derived products. We found that N. brasiliensis infection and its derived proteins had suitable protective effects both in the initial and progressive stages of atherosclerosis, effectively reducing aortic arch plaque areas and liver lipid contents and downregulating serum LDL levels, which may be associated with the significant upregulation of serum anti-inflammatory cytokines (IL-10 and IL-4) and the down-regulation of proinflammatory cytokines (TNF-α and IFN-γ) in the serum. In conclusion, these data highlighted the effective regulatory role of N. brasiliensis and its derived proteins in the development and progression of atherosclerosis. This could provide a promising new avenue for the prevention and treatment of atherosclerosis.
Collapse
Affiliation(s)
- Yougui Yang
- School of Public Health, Nanjing Medical University, Nanjing 211166, China
- Key Laboratory of National Health Commission on Parasitic Disease Control and Prevention, Key Laboratory of Jiangsu Province on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi 214064, China
| | - Xin Ding
- Key Laboratory of National Health Commission on Parasitic Disease Control and Prevention, Key Laboratory of Jiangsu Province on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi 214064, China
| | - Fuzhong Chen
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Xiaomin Wu
- Key Laboratory of National Health Commission on Parasitic Disease Control and Prevention, Key Laboratory of Jiangsu Province on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi 214064, China
- Microbiological Laboratory, Anhui Provincial Center for Disease Control and Prevention, Hefei 230601, China
| | - Yuying Chen
- School of Public Health, Nanjing Medical University, Nanjing 211166, China
- Key Laboratory of National Health Commission on Parasitic Disease Control and Prevention, Key Laboratory of Jiangsu Province on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi 214064, China
| | - Qiang Zhang
- Key Laboratory of National Health Commission on Parasitic Disease Control and Prevention, Key Laboratory of Jiangsu Province on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi 214064, China
| | - Jun Cao
- School of Public Health, Nanjing Medical University, Nanjing 211166, China
- Key Laboratory of National Health Commission on Parasitic Disease Control and Prevention, Key Laboratory of Jiangsu Province on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi 214064, China
| | - Junhong Wang
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
- Correspondence: (J.W.); (Y.D.)
| | - Yang Dai
- School of Public Health, Nanjing Medical University, Nanjing 211166, China
- Key Laboratory of National Health Commission on Parasitic Disease Control and Prevention, Key Laboratory of Jiangsu Province on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi 214064, China
- Correspondence: (J.W.); (Y.D.)
| |
Collapse
|
7
|
Li RY, Qin Q, Yang HC, Wang YY, Mi YX, Yin YS, Wang M, Yu CJ, Tang Y. TREM2 in the pathogenesis of AD: a lipid metabolism regulator and potential metabolic therapeutic target. Mol Neurodegener 2022; 17:40. [PMID: 35658903 PMCID: PMC9166437 DOI: 10.1186/s13024-022-00542-y] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 05/09/2022] [Indexed: 12/13/2022] Open
Abstract
Triggering receptor expressed on myeloid cells 2 (TREM2) is a single-pass transmembrane immune receptor that is mainly expressed on microglia in the brain and macrophages in the periphery. Recent studies have identified TREM2 as a risk factor for Alzheimer’s disease (AD). Increasing evidence has shown that TREM2 can affect lipid metabolism both in the central nervous system (CNS) and in the periphery. In the CNS, TREM2 affects the metabolism of cholesterol, myelin, and phospholipids and promotes the transition of microglia into a disease-associated phenotype. In the periphery, TREM2 influences lipid metabolism by regulating the onset and progression of obesity and its complications, such as hypercholesterolemia, atherosclerosis, and nonalcoholic fatty liver disease. All these altered lipid metabolism processes could influence the pathogenesis of AD through several means, including affecting inflammation, insulin resistance, and AD pathologies. Herein, we will discuss a potential pathway that TREM2 mediates lipid metabolism to influence the pathogenesis of AD in both the CNS and periphery. Moreover, we discuss the possibility that TREM2 may be a key factor that links central and peripheral lipid metabolism under disease conditions, including AD. This link may be due to impacts on the integrity of the blood–brain barrier, and we introduce potential pathways by which TREM2 affects the blood–brain barrier. Moreover, we discuss the role of lipids in TREM2-associated treatments for AD. We propose some potential therapies targeting TREM2 and discuss the prospect and limitations of these therapies.
Collapse
Affiliation(s)
- Rui-Yang Li
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, National Center for Neurological Disorders, Beijing, China
| | - Qi Qin
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, National Center for Neurological Disorders, Beijing, China
| | - Han-Chen Yang
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, National Center for Neurological Disorders, Beijing, China
| | - Ying-Ying Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Ying-Xin Mi
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, National Center for Neurological Disorders, Beijing, China
| | - Yun-Si Yin
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, National Center for Neurological Disorders, Beijing, China
| | - Meng Wang
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, National Center for Neurological Disorders, Beijing, China
| | - Chao-Ji Yu
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, National Center for Neurological Disorders, Beijing, China
| | - Yi Tang
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, National Center for Neurological Disorders, Beijing, China.
| |
Collapse
|
8
|
Tao Y, Geng Y, Dang W, Xu X, Zhao H, Zou L, Li Y. Mechanism of Endoplasmic Reticulum Stress Pathway in the Osteogenic Phenotypic Transformation of Aortic Valve Interstitial Cells. Front Endocrinol (Lausanne) 2022; 13:856331. [PMID: 35355558 PMCID: PMC8959129 DOI: 10.3389/fendo.2022.856331] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 02/07/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND AND PURPOSE Calcific Aortic Valve Disease (CAVD) is a crucial component of degenerative valvular disease in old age and with the increasing prevalence of the aging population. we hope that by modeling valvular osteogenesis and intervening with endoplasmic reticulum stress inhibitor TUDCA to observe the effect of endoplasmic reticulum stress on valve osteogenesis. METHODS In this study, rabbit heart valvular interstitial cells (VICs) were isolated and cultured. They treated with ox-LDL (Oxidized Low Density Lipoprotein) stimulation to establish a model of valvular osteogenic transformation. BMP2 (Bone Morphogenetic Protein 2), PERK (Protein kinase R-like endoplasmic reticulum kinase), CHOP (CCAAT/enhancer-binding protein homologous protein) and transcriptional regulatory factor ATF4 (Activating Transcription Factor 4 )were recorded after intervention with ER stress inhibitor TUDCA. The effects of er stress on valvular osteogenic transformation were analyzed. RESULT After stimulation of VICs with ox-LDL, the expression levels of BMP2, PERK, CHOP, and ATF4 increased. However, TUDCA treatment can alleviate the increased expression levels of BMP2, PERK ATF4, and CHOP under ox-LDL stimulation to a certain extent. CONCLUSION The endoplasmic reticulum stress signaling pathway is involved in ox-LDL-induced calcification of rabbit valve interstitial cells. Inhibition of endoplasmic reticulum stress using TUDCA can improve the progression of rabbit aortic valve calcification.
Collapse
Affiliation(s)
- Yiming Tao
- Department of Intensive Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- The Emergency Department, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yimin Geng
- Department of Intensive Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- The Emergency Department, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenpei Dang
- Department of Intensive Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- The Emergency Department, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xinxin Xu
- Department of Intensive Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- The Emergency Department, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hui Zhao
- Department of Intensive Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- The Emergency Department, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lijuan Zou
- Department of Intensive Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- The Emergency Department, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yongsheng Li
- Department of Intensive Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- The Emergency Department, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Yongsheng Li,
| |
Collapse
|
9
|
C/EBPβ is a key transcription factor of ox-LDL inducing THP-1 cells to release multiple pro-inflammatory cytokines. Inflamm Res 2021; 70:1191-1199. [PMID: 34605942 DOI: 10.1007/s00011-021-01509-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 09/21/2021] [Accepted: 09/23/2021] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVE CCAAT/enhancer binding protein β (C/EBPβ) plays an important role during atherogenesis. However, how C/EBPβ functions remains unclear. In this study, we explore the relationship between C/EBPβ and oxidized LDL-induced multiple pro-inflammatory cytokines released in monocytes. MATERIALS AND METHODS THP-1 cells (human monocyte cell line) were stimulated by ox-LDL, ChIP was used to detect the binding function of C/EBPβ to target genes, small interfering RNA was used to knock down the expression of C/EBPβ, Western Blot was used to detect protein expression, and ChIP-seq was used to detect different groups of C/EBPβ bound gene fragments. The integrative genomics viewer (IGV), model-based analysis of ChIP-seq (MACS) were used to visualize the results of ChIP-seq. GO (gene ontology), KEGG (Kyoto Encyclopedia of Genes and Genomes) and Reactome data bases enrichment analysis were performed by the ClusterProfiler software. Ingenuity pathway analysis (IPA) was used to analyze the results of ChIP-seq and to summarize the data within the database. RESULTS We identified C/EBPβ as a key protein that regulated IL-1β, IL-6 through database. Then our results confirmed that C/EBPβ could bind directly to the gene of IL-18 and C/EBPβ plays a role in the increased expression and secretion of IL-18 protein after ox-LDL stimulation of THP-1. Using ChIP-seq, we found that the enhanced transcriptional function of C/EBPβ after ox-LDL treatment triggered changes in C/EBPβ-regulated downstream pathways. In the ChIP-seq results, we extracted inflammatory cytokines with significant expression differences, and by comparing them with the database of inflammatory cytokines that C/EBPβ directly regulated, we screened five inflammatory cytokines, CXCL8, IL17B, TNFSF11, CSF3, and CCL2, and the results showed that knockdown of C/EBPβ expression inhibited ox-LDL-induced secretion of CXCL8, TNFSF11, CSF3, and CCL2 by THP-1. CONCLUSION Our results suggest that ox-LDL stimulation enhances C/EBPβ-regulated transcription in THP-1 and C/EBPβ upregulate the release of multiple pro-inflammatory cytokines including IL-18, IL-1β, and IL-6 through direct binding to genes.
Collapse
|
10
|
Checkouri E, Blanchard V, Meilhac O. Macrophages in Atherosclerosis, First or Second Row Players? Biomedicines 2021; 9:biomedicines9091214. [PMID: 34572399 PMCID: PMC8465019 DOI: 10.3390/biomedicines9091214] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/08/2021] [Accepted: 09/11/2021] [Indexed: 12/24/2022] Open
Abstract
Macrophages represent a cell type that has been widely described in the context of atherosclerosis since the earliest studies in the 17th century. Their role has long been considered to be preponderant in the onset and aggravation of atherosclerosis, in particular by participating in the establishment of a chronic inflammatory state by the release of pro-inflammatory cytokines and by uncontrolled engorgement of lipids resulting in the formation of foam cells and later of the necrotic core. However, recent evidence from mouse models using an elegant technique of tracing vascular smooth muscle cells (VSMCs) during plaque development revealed that resident VSMCs display impressive plastic properties in response to an arterial injury, allowing them to switch into different cell types within the plaque, including mesenchymal-like cells, macrophage-like cells and osteochondrogenic-like cells. In this review, we oppose the arguments in favor or against the influence of macrophages versus VSMCs in all stages of atherosclerosis including pre-atherosclerosis, formation of lipid-rich foam cells, development of the necrotic core and the fibrous cap as well as calcification and rupture of the plaque. We also analyze the relevance of animal models for the investigation of the pathophysiological mechanisms of atherosclerosis in humans, and discuss potential therapeutic strategies targeting either VSMCs or macrophage to prevent the development of cardiovascular events. Overall, although major findings have been made from animal models, efforts are still needed to better understand and therefore prevent the development of atherosclerotic plaques in humans.
Collapse
Affiliation(s)
- Eloïse Checkouri
- INSERM, UMR 1188 Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de La Réunion, 97400 Sainte-Clotilde, France; (E.C.); (V.B.)
- Habemus Papam, Food Industry, 97470 Saint-Benoit, France
| | - Valentin Blanchard
- INSERM, UMR 1188 Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de La Réunion, 97400 Sainte-Clotilde, France; (E.C.); (V.B.)
- Departments of Medicine, Centre for Heart Lung Innovation, Providence Healthcare Research Institute, St Paul’s Hospital, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Olivier Meilhac
- INSERM, UMR 1188 Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de La Réunion, 97400 Sainte-Clotilde, France; (E.C.); (V.B.)
- CHU de La Réunion, INSERM, CIC1410, 97500 Saint-Pierre, France
- Correspondence: ; Tel.: +33-262-93-8811
| |
Collapse
|
11
|
Quiros-Roldan E, Carriero C, Paghera S, Degli Antoni M, Fiorini C, Quaresima V, Castelli F, Imberti L. Symptoms and quality of life in HIV-infected patients with benign prostatic hyperplasia are improved by the consumption of a newly developed whole tomato-based food supplement. A phase II prospective, randomized double-blinded, placebo-controlled study. J Funct Foods 2021. [DOI: 10.1016/j.jff.2021.104495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|
12
|
Ackers I, Szymanski C, Silver MJ, Malgor R. Oxidized Low-Density Lipoprotein Induces WNT5A Signaling Activation in THP-1 Derived Macrophages and a Human Aortic Vascular Smooth Muscle Cell Line. Front Cardiovasc Med 2020; 7:567837. [PMID: 33330641 PMCID: PMC7710548 DOI: 10.3389/fcvm.2020.567837] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 10/22/2020] [Indexed: 12/13/2022] Open
Abstract
The pathogenesis of atherosclerosis is complex, evolves, and involves many cell types. Macrophages and vascular smooth muscle cells (VSMCs) are critically involved in atherosclerosis development and progression. Several studies have shown that WNT5A protein is abundantly expressed in human atherosclerotic lesions; however, the mechanism and role of WNT signaling pathway activation is not clearly known. Using THP-1 derived macrophages, and human aortic VSMC cells, we evaluated in vitro how oxidized low-density lipoprotein (oxLDL) and WNT5A signaling interact in these two cell lines. We used western blot, scratch assay, metabolic proliferation assay, as well as immunostaining to analyze the effect of Wnt signaling activation. The results demonstrated that oxLDL, as well as WNT5A (control), induced Disheveled-2 (DVL2) activation and Kif26b degradation, indicating activation of non-canonical Wnt signaling. We found that oxLDL and WNT5A induced FZD5-ROR2 co-localization at the cellular membrane in vitro in THP-1 derived macrophages. Box5 (FZD5 receptor antagonist) inhibited oxLDL-induced DVL2/JNK activation secondary to newly secreted WNT protein from THP-1 derived macrophages. We found that WNT3A (canonical Wnt) and WNT5A showed different roles in this VSMC cell line. These findings indicate that WNT5A is upregulated by oxLDL, promotes foam cell formation, and affects VSMC phenotype and migration in these two cell lines. Also, in these cell lines FZD5 signaling seems to be necessary for lipid accumulation and, through this mechanism, WNT5A could modulate foam cell formation. Thus, our results suggest that WNT5A may contribute to the pathogenesis of vascular disease through modulating macrophage and VSMC behavior.
Collapse
Affiliation(s)
- Ian Ackers
- Osteopathic Heritage Foundation, Translational Biomedical Sciences Program, Ohio University, Athens, OH, United States
| | - Candice Szymanski
- Department of Biomedical Sciences, Ohio University, Athens, OH, United States
| | | | - Ramiro Malgor
- Department of Biomedical Sciences, Ohio University, Athens, OH, United States
- The Diabetes Institute, Ohio University, Athens, OH, United States
- Molecular and Cellular Biology Graduate Program, Ohio University, Athens, OH, United States
| |
Collapse
|
13
|
Direct electrochemiluminescent immunosensing for an early indication of coronary heart disease using dual biomarkers. Anal Chim Acta 2020; 1110:82-89. [DOI: 10.1016/j.aca.2020.03.022] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 03/07/2020] [Accepted: 03/11/2020] [Indexed: 11/18/2022]
|
14
|
Lin F, Yin HB, Li XY, Zhu GM, He WY, Gou X. Bladder cancer cell‑secreted exosomal miR‑21 activates the PI3K/AKT pathway in macrophages to promote cancer progression. Int J Oncol 2019; 56:151-164. [PMID: 31814034 PMCID: PMC6910194 DOI: 10.3892/ijo.2019.4933] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Accepted: 11/08/2019] [Indexed: 12/11/2022] Open
Abstract
Tumour-associated macrophages (TAMs) compose a major component of the tumour microenvironment and form in this microenvironment prior to cancer metastasis. However, the detailed mechanisms of TAM remodelling in the context of bladder cancer have not been clearly defined. The present study collected exosomes from the conditioned medium of human bladder T24 cancer cells. The effects of macrophages treated with exosomes derived from T24 cells on bladder cancer cell migration and invasion were analysed by Transwell assays. The expression levels of endogenous and exosomal microRNA-21 (miR-21) were examined by reverse transcription-quantitative PCR, while the expression level of the target protein was analysed by western blot analysis. Luciferase reporter plasmids and mutants were used to confirm direct targeting. The effects of miR-21 on bladder cancer cell migration and invasion were analysed by Transwell and Matrigel assays following miR-21 transfection. It was identified that exosomes derived from bladder cancer cells polarized THP-1 cell-derived macrophages into the M2 phenotype, and TAM-mediated pro-migratory and pro-invasive activity was determined. Moreover, it was found that miR-21 was highly expressed in exosomes derived from bladder cancer cells as well as in macrophages treated with exosomes. In addition, macrophages transfected with miR-21 exhibited M2 polarization and promoted T24 cell migratory and invasive ability. Mechanistically, exosomal miR-21 derived from bladder cancer cells inhibited phosphatase and tensin homolog activation of the PI3K/AKT signalling pathway in macrophages and enhanced STAT3 expression to promote M2 phenotypic polarization. The present results suggest that exosomal miR-21 can promote cancer progression by polarizing TAMs.
Collapse
Affiliation(s)
- Fan Lin
- Department of Urology, The First Affiliated Hospital, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Hu-Bin Yin
- Department of Urology, The First Affiliated Hospital, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Xin-Yuan Li
- Department of Urology, The First Affiliated Hospital, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Gong-Min Zhu
- Department of Urology, The First Affiliated Hospital, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Wei-Yang He
- Department of Urology, The First Affiliated Hospital, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Xin Gou
- Department of Urology, The First Affiliated Hospital, Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|
15
|
Feng C, Chen Q, Fan M, Guo J, Liu Y, Ji T, Zhu J, Zhao X. Platelet-derived microparticles promote phagocytosis of oxidized low-density lipoprotein by macrophages, potentially enhancing foam cell formation. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:477. [PMID: 31700913 DOI: 10.21037/atm.2019.08.06] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Background The interaction between platelets and macrophages plays an important role in the development and progression of atherosclerosis (AS). This study aimed to investigate the role of platelet microparticles (PMPs) in the development of foam cells. Methods PMPs are generated by activating platelets with thrombin and separated by ultracentrifugation. The macrophages were treated with PMPs, the phagocytosis of oxidized low-density lipoprotein (Ox-LDL) and formation of foam cells were evaluated by flow cytometry and confocal microscopy, respectively, and the inflammatory factors cytokines in the supernatant were detected by ELISA. Results PMPs significantly increase the phagocytosis of Ox-LDL and elevated foam cell formation of macrophages. IL-1β content in the supernatant of macrophages peaked around 2-4 h and declined to normal level after 6-8 h; IL-6 content peaked at 4 h and then decreased to normal level. TNF-α content peaked at 2-4 h. Conclusions The microparticles from activated platelets can increase the phagocytosis of Ox-LDL and the production of inflammatory cytokines by macrophages, which is related to the development of AS.
Collapse
Affiliation(s)
- Can Feng
- Department of Cardiology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China.,Department of Cardiology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Qi Chen
- Department of Cardiology, Biomedical Research Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China
| | - Min Fan
- Department of Cardiology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Jun Guo
- Department of Cardiology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Yu Liu
- Department of Cardiology, Nanjing University Medical School Affiliated Nanjing Drum Tower Hospital, Nanjing 210008, China
| | - Tao Ji
- Department of Neurosurgery, Tenth Affiliated Hospital, Tongji University, Shanghai 200072, China
| | - Jiaqi Zhu
- Department of Cardiology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Xianxian Zhao
- Department of Cardiology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| |
Collapse
|
16
|
Chinchu JU, Mohan MC, Devi SJR, Kumar BP. Evaluation of anti-inflammatory effect of Varanadi Kashayam (decoction) in THP-1-derived macrophages. Ayu 2019; 39:243-249. [PMID: 31367148 PMCID: PMC6639814 DOI: 10.4103/ayu.ayu_53_18] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Background Varanadi Kashayam is an Ayurvedic polyherbal decoction containing 16 ingredients, for which the mechanisms of action involved in controlling chronic inflammatory conditions have not been evaluated. The inhibition of release of proinflammatory cytokines by lipopolysaccharide (LPS)-stimulated monocytes/macrophages is an ideal in vitro model for identifying anti-inflammatory molecules. Aim The aim of the study is to determine the anti-inflammatory effect of Varanadi Kashayam in THP-1-derived macrophages. Materials and Methods The efficacy of Varanadi Kashayam on monocyte cell differentiation was determined by quantitative polymerase chain reaction to assess the expression of differentiation markers MMP-9, CD36, CD11b and CD14. Further Varanadi Kashayam treated THP-1 macrophages were induced with LPS and the production of proinflammatory cytokines tumor necrosis factor-alpha (TNF-α) and interleukin-1beta (IL-1β) were measured and corresponding genes expressions were quantified. Results The results indicate that Varanadi Kashayam reduced the differentiation of THP-1 monocytes to macrophages and downregulated the expression of cell surface markers. Furthermore, it could decrease the release of proinflammatory cytokines from LPS-induced THP-1 macrophages and downregulated the expression of TNF-α and IL-1β genes. Conclusion The results obtained from this study suggest a possible mechanism of action of the herbal decoction in inflammatory processes and opens up the possibilities of identifying bioactive lead molecules with anti-inflammatory potentials.
Collapse
Affiliation(s)
- J U Chinchu
- Department of School of Biosciences, Inflammation Research Lab, Mahatma Gandhi University, Kottayam, Kerala, India
| | - Mohind C Mohan
- Department of School of Biosciences, Inflammation Research Lab, Mahatma Gandhi University, Kottayam, Kerala, India
| | - S J Rahitha Devi
- Department of School of Biosciences, Inflammation Research Lab, Mahatma Gandhi University, Kottayam, Kerala, India
| | - B Prakash Kumar
- Department of School of Biosciences, Inflammation Research Lab, Mahatma Gandhi University, Kottayam, Kerala, India
| |
Collapse
|
17
|
Zhou J, Ma W, Wang X, Liu H, Miao Y, Wang J, Du P, Chen Y, Zhang Y, Liu Z. Matrine Suppresses Reactive Oxygen Species (ROS)-Mediated MKKs/p38-Induced Inflammation in Oxidized Low-Density Lipoprotein (ox-LDL)-Stimulated Macrophages. Med Sci Monit 2019; 25:4130-4136. [PMID: 31156213 PMCID: PMC6561390 DOI: 10.12659/msm.917151] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Background The objective of this study was to study the anti-inflammatory effect and possibly involved molecular mechanisms of matrine on oxidized low-density lipoprotein (ox-LDL)-exposed macrophages. Material/Methods Cultured human macrophages (THP-1 cell line) were exposed to ox-LDL at final concentrations of 0, 25, 50, and 100 μg/mL. Several cells were then treated with matrine at serial diluted concentrations. 2,7-Dichlorodi-hydrofluorescein diacetate (DCFH-DA) staining was used to evaluate reactive oxygen species (ROS) production; a colorimetric method was used to determine the cellular antioxidant capacity; production of pro-inflammatory cytokines interleukin (IL)18 and tumor necrosis factor (TNF)α were determined by enzyme-linked immunosorbent assay (ELISA); and immunoblot assay was used to assess the relative protein phosphorylation and expression. Results ox-LDL exposure significantly elevated intracellular ROS level and supernatant IL18 and TNFα concentrations, but impaired total antioxidant capacity (TAC) of macrophages. The relative phosphorylations of MAPK kinase kinases (MKK)6, MKK3, and p38 mitogen-activated protein kinases (MAPK) were increased by ox-LDL exposure. The expression levels of IL18 and TNFα were also increased in ox-LDL-treated macrophages. The matrine treatment reduced intracellular ROS level and supernatant IL18 and TNFα concentrations and increased TAC in a concentration- dependent manner. The relative phosphorylations of MKK6, MKK3, and p38 MAPK were reduced after matrine administration. Moreover, the expression levels of IL18 and TNFα were also decreased by matrine treatment, in a concentration-dependent manner. Conclusions ox-LDL increases inflammatory response in macrophages by activating the ROS-mediated MKKs/p38 MAPK-induced inflammatory signaling pathway. Matrine suppresses ox-LDL-induced inflammatory by inhibiting the MKKs/p38 MAPK signaling pathway.
Collapse
Affiliation(s)
- Junli Zhou
- Department of Cardiology, Zhouzhi County People's Hospital, Xi'an, Shaanxi, China (mainland)
| | - Wangxia Ma
- Department of Cardiology, Zhouzhi County People's Hospital, Xi'an, Shaanxi, China (mainland)
| | - Xincheng Wang
- Department of Cardiology, Zhouzhi County People's Hospital, Xi'an, Shaanxi, China (mainland)
| | - Hongbo Liu
- Department of Cardiology, Zhouzhi County People's Hospital, Xi'an, Shaanxi, China (mainland)
| | - Youliang Miao
- Department of Cardiology, Zhouzhi County People's Hospital, Xi'an, Shaanxi, China (mainland)
| | - Juanli Wang
- Department of Cardiology, Zhouzhi County People's Hospital, Xi'an, Shaanxi, China (mainland)
| | - Peng Du
- Department of Cardiology, Zhouzhi County People's Hospital, Xi'an, Shaanxi, China (mainland)
| | - Yani Chen
- Department of Cardiology, Zhouzhi County People's Hospital, Xi'an, Shaanxi, China (mainland)
| | - Yong Zhang
- Department of Cardiology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, China (mainland)
| | - Zhongwei Liu
- Department of Cardiology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, China (mainland)
| |
Collapse
|
18
|
Dawodu D, Patecki M, Dumler I, Haller H, Kiyan Y. oxLDL inhibits differentiation of mesenchymal stem cells into osteoblasts via the CD36 mediated suppression of Wnt signaling pathway. Mol Biol Rep 2019; 46:3487-3496. [PMID: 30847850 DOI: 10.1007/s11033-019-04735-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 02/28/2019] [Indexed: 10/27/2022]
Abstract
Bone abnormalities as a consequence of osteoblast deregulation are associated with several diseases such as diabetes and chronic kidney disease. Important role for oxidized low density lipoproteins (oxLDL) in the pathophysiology of bone disorders has been reported. However, little is known about the effects and mechanisms of oxLDL on the process of osteoblastogenesis in human mesenchymal stem cells (MSCs). We show that oxLDL concentrations of ~ 10-25 µg protein (0.43-1.0 µM MDA/mg protein) inhibited the differentiation of MSCs to osteoblasts. We demonstrate that the underlying mechanism entails the suppression of the Wnt signaling through the down-regulation of β-catenin. Further, we show the association of scavenger receptor CD36 with the receptors LRP5/6 and Frizzled in mediating the oxLDL effects on the differentiation of MSCs to pre-osteoblasts. Inhibiting CD36 restored osteoblasts differentiation in the presence of oxLDL. Our findings suggest that oxLDL interferes with the canonical Wnt signaling pathway in a CD36 dependent manner leading to an inhibition of osteoblastogenesis.
Collapse
Affiliation(s)
- Damilola Dawodu
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Margret Patecki
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Inna Dumler
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Hermann Haller
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Yulia Kiyan
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
19
|
Leonhardt J, Große S, Marx C, Siwczak F, Stengel S, Bruns T, Bauer R, Kiehntopf M, Williams DL, Wang ZQ, Mosig AS, Weis S, Bauer M, Heller R. Candida albicans β-Glucan Differentiates Human Monocytes Into a Specific Subset of Macrophages. Front Immunol 2018; 9:2818. [PMID: 30555483 PMCID: PMC6284042 DOI: 10.3389/fimmu.2018.02818] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 11/14/2018] [Indexed: 12/12/2022] Open
Abstract
β-Glucan derived from cell walls of Candida albicans is a potent immune modulator. It has been shown to induce trained immunity in monocytes via epigenetic and metabolic reprogramming and to protect from lethal sepsis if applied prior to infection. Since β-glucan-trained monocytes have not been classified within the system of mononuclear phagocytes we analyzed these cells metabolically, phenotypically and functionally with a focus on monocyte-to-macrophage differentiation and compared them with naïve monocytes and other types of monocyte-derived cells such as classically (M1) or alternatively (M2) activated macrophages and monocyte-derived dendritic cells (moDCs). We show that β-glucan inhibits spontaneous apoptosis of monocytes independent from autocrine or paracrine M-CSF release and stimulates monocyte differentiation into macrophages. β-Glucan-differentiated macrophages exhibit increased cell size and granularity and enhanced metabolic activity when compared to naïve monocytes. Although β-glucan-primed cells expressed markers of alternative activation and secreted higher levels of IL-10 after lipopolysaccharide (LPS), their capability to release pro-inflammatory cytokines and to kill bacteria was unaffected. Our data demonstrate that β-glucan priming induces a population of immune competent long-lived monocyte-derived macrophages that may be involved in immunoregulatory processes.
Collapse
Affiliation(s)
- Julia Leonhardt
- Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany.,Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany
| | - Silke Große
- Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany.,Institute of Molecular Cell Biology, Center for Molecular Biomedicine, Jena University Hospital, Jena, Germany
| | - Christian Marx
- Leibniz Institute on Aging, Fritz Lipmann Institute, Jena, Germany
| | - Fatina Siwczak
- Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany.,Institute of Biochemistry II, Jena University Hospital, Jena, Germany
| | - Sven Stengel
- Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany.,Department of Internal Medicine IV, Jena University Hospital, Jena, Germany
| | - Tony Bruns
- Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany.,Department of Internal Medicine IV, Jena University Hospital, Jena, Germany
| | - Reinhard Bauer
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine, Jena University Hospital, Jena, Germany
| | - Michael Kiehntopf
- Institute of Clinical Chemistry and Laboratory Diagnostics Jena University Hospital, Jena, Germany
| | - David L Williams
- Department of Surgery and Center of Excellence in Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Zhao-Qi Wang
- Leibniz Institute on Aging, Fritz Lipmann Institute, Jena, Germany
| | - Alexander S Mosig
- Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany.,Institute of Biochemistry II, Jena University Hospital, Jena, Germany
| | - Sebastian Weis
- Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany.,Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany.,Center for Infectious Diseases and Infection Control, Jena University Hospital, Jena, Germany
| | - Michael Bauer
- Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany.,Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany
| | - Regine Heller
- Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany.,Institute of Molecular Cell Biology, Center for Molecular Biomedicine, Jena University Hospital, Jena, Germany
| |
Collapse
|
20
|
Varghese JF, Patel R, Yadav UCS. Sterol regulatory element binding protein (SREBP) -1 mediates oxidized low-density lipoprotein (oxLDL) induced macrophage foam cell formation through NLRP3 inflammasome activation. Cell Signal 2018; 53:316-326. [PMID: 30389501 DOI: 10.1016/j.cellsig.2018.10.020] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 10/26/2018] [Accepted: 10/29/2018] [Indexed: 11/27/2022]
Abstract
Macrophage foam cell formation (FCF) has long been known to play a critical role during atherosclerotic plaque development. In the presence of atherogenic molecules such as oxidized low-density lipoprotein (oxLDL) macrophages accumulate massive amounts of lipid through uptake. However, in the presence of oxLDL mechanism of dysregulated lipid homeostasis in the macrophages remains largely unknown. Herein we have investigated the role of Sterol regulatory element binding protein (SREBP)-1 in oxLDL-induced inflammation and altered lipid homeostasis in macrophages. The U937 monocytes and monocyte-derived macrophages (MDMs) were stimulated with different doses of oxLDL. MTT assay to study the effect of oxLDL on cell viability, Oil-Red-O (ORO) staining to observe cytosolic lipid accumulation, semi-quantitative PCR and Western blotting to analyze mRNA and protein expressions, respectively, and spectrophotometric assay to measure the lipid synthesizing enzyme's activity were performed. Our results indicate that oxLDL increased proliferation in monocytes and decreased the viability in MDMs in a time- and dose-dependent manner. The oxLDL (100 μg/ml) enhanced lipid accumulation via increased expressions of SREBP-1 and its downstream proteins such as fatty acid synthase (FAS) and 3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR) at both RNA and protein levels in monocytes as well as in MDMs. Inhibiting SREBP-1 by a synthetic inhibitor prevented excessive lipid accumulation by downregulating the expression of its downstream proteins. Further, oxLDL increased reactive oxygen species (ROS) levels, NLRP3 inflammasome activation and active interleukin 1β (IL-1β) release in both the cell types. The oxLDL-induced NLRP3 could be responsible for SREBP-1 and downstream proteins overexpression as siRNA silencing of NLRP3 decreased SERBP-1 levels. In summary, we have demonstrated that SREBP-1 could be a key player in oxLDL-induced excessive lipid accumulation leading to macrophage FCF via ROS-mediated NLRP3/IL-1β/SREBP-1 pathway.
Collapse
Affiliation(s)
- Johnna F Varghese
- Metabolic Disorders and Inflammatory Pathologies Laboratory, School of Life Sciences, Central University of Gujarat, Gandhinagar, Gujarat 382030, India
| | - Rohit Patel
- Metabolic Disorders and Inflammatory Pathologies Laboratory, School of Life Sciences, Central University of Gujarat, Gandhinagar, Gujarat 382030, India
| | - Umesh C S Yadav
- Metabolic Disorders and Inflammatory Pathologies Laboratory, School of Life Sciences, Central University of Gujarat, Gandhinagar, Gujarat 382030, India.
| |
Collapse
|
21
|
Dawodu D, Patecki M, Hegermann J, Dumler I, Haller H, Kiyan Y. oxLDL inhibits differentiation and functional activity of osteoclasts via scavenger receptor-A mediated autophagy and cathepsin K secretion. Sci Rep 2018; 8:11604. [PMID: 30072716 PMCID: PMC6072764 DOI: 10.1038/s41598-018-29963-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 07/19/2018] [Indexed: 12/24/2022] Open
Abstract
Resorptive activity of osteoclasts is important for maintaining bone homeostasis. Endogenous compounds such as oxidized low density lipoprotein (oxLDL) have been shown to disturb this activity. While some studies have investigated the effects of oxLDL on the process of osteoclastogenesis, the underlying mechanism are not fully understood. We show here that oxLDL concentrations of ~10-25 µg protein (0.43-1.0 µM MDA/mg protein) completely blocked the formation of functional osteoclasts. The underlying mechanism implies an inhibition of autophagy that in turn leads to a decreased fusion of cathepsin K (CatK)-loaded lysosomal vesicles with the ruffled border membrane. As result, a lower secretion of CatK and impaired protonation of the resorption lacunae by vacuolar-ATPase (v-ATPase) is observed in the presence of oxLDL. We demonstrate that scavenger receptor A (SR-A) mediates oxLDL effects on osteoclastogenesis and repressing this receptor partially rescued oxLDL effects. Collectively, our data provides an insight into the possible mechanism of oxLDL on osteoclastogenesis suggesting that it does not perturb the packaging of CatK and v-ATPase (V-a3) in the secretory lysosome, but inhibits the fusion of these lysosomes to the ruffled border. The relevance of our findings suggests a distinct link between oxLDL, autophagy and osteoclastogenesis.
Collapse
Affiliation(s)
- Damilola Dawodu
- Department of Nephrology and Hypertensiology, Hannover Medical School, Hannover, Germany
| | - Margret Patecki
- Department of Nephrology and Hypertensiology, Hannover Medical School, Hannover, Germany
| | - Jan Hegermann
- Research Core Unit Electron Microscopy, Hannover Medical School, Hannover, Germany
| | - Inna Dumler
- Department of Nephrology and Hypertensiology, Hannover Medical School, Hannover, Germany
| | - Hermann Haller
- Department of Nephrology and Hypertensiology, Hannover Medical School, Hannover, Germany
| | - Yulia Kiyan
- Department of Nephrology and Hypertensiology, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
22
|
Ackers I, Szymanski C, Duckett KJ, Consitt LA, Silver MJ, Malgor R. Blocking Wnt5a signaling decreases CD36 expression and foam cell formation in atherosclerosis. Cardiovasc Pathol 2018; 34:1-8. [PMID: 29474941 DOI: 10.1016/j.carpath.2018.01.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 01/12/2018] [Accepted: 01/19/2018] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND AND AIMS Wnt5a is a highly studied member of the Wnt family and recently has been implicated in the pathogenesis of atherosclerosis, but its precise role is unknown. Foam cell development is a critical process to atherosclerotic plaque formation. In the present study, we investigated the role of noncanonical Wnt5a signaling in the development of foam cells. METHODS Human carotid atherosclerotic tissue and THP-1-derived macrophages were used to investigate the contribution of Wnt5a signaling in the formation of foam cells. Immunohistochemistry was used to evaluate protein expression of scavenger receptors and noncanonical Wnt5a receptors [frizzled 5 (Fz5) and receptor tyrosine kinase-like orphan receptor 2 (Ror2)] in human atherosclerotic macrophages/foam cells. Changes in protein expression in response to Wnt5a stimulation/inhibition were determined by Western blot, and lipid accumulation was evaluated by fluorescent lipid droplet staining. RESULTS Wnt5a (P<.05), Fz5 (P<.01), and Ror2 (P<.01) were significantly expressed in advanced atherosclerotic lesions compared to less advanced lesions (N=10). Wnt5a, Fz5, and Ror2 were expressed in macrophages/foam cells within the plaque. In vitro studies revealed that Wnt5a significantly increased the expression of the lipid uptake receptor CD36 (P<.05) but not the lipid efflux receptor ATP-binding cassette transporter (P>.05). rWnt5a also significantly increased lipid accumulation in THP-1 macrophages (P<.05). Furthermore, inhibition of Wnt5a signaling with Box5 prevented lipid accumulation (P<.01) and prevented CD36 up-regulation (P<.01). CONCLUSIONS These results suggest a direct role for Wnt5a signaling in the pathogenesis of atherosclerosis, specifically the accumulation of lipid in macrophages and the formation of foam cells.
Collapse
Affiliation(s)
- Ian Ackers
- Heritage Fellow, Translational Biomedical Sciences Program, Ohio University, Athens, Ohio, USA
| | - Candice Szymanski
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio, USA
| | - K Jordan Duckett
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio, USA
| | - Leslie A Consitt
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio, USA; The Diabetes Institute, Ohio University, Athens, Ohio, USA
| | | | - Ramiro Malgor
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio, USA; The Diabetes Institute, Ohio University, Athens, Ohio, USA; Molecular and Cellular Biology Graduate Program, Ohio University, Athens, Ohio, USA.
| |
Collapse
|
23
|
Gargiulo S, Testa G, Gamba P, Staurenghi E, Poli G, Leonarduzzi G. Oxysterols and 4-hydroxy-2-nonenal contribute to atherosclerotic plaque destabilization. Free Radic Biol Med 2017; 111:140-150. [PMID: 28057601 DOI: 10.1016/j.freeradbiomed.2016.12.037] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 12/22/2016] [Accepted: 12/24/2016] [Indexed: 12/31/2022]
Abstract
A growing bulk of evidence suggests that cholesterol oxidation products, known as oxysterols, and 4-hydroxy-2-nonenal (HNE), the major proatherogenic components of oxidized low density lipoproteins (oxLDLs), significantly contribute to atherosclerotic plaque progression and destabilization, with eventual plaque rupture. These oxidized lipids are involved in various key steps of this complex process, mainly thanks to their ability to induce inflammation, oxidative stress, and apoptosis. This review summarizes the current knowledge of the effects induced by these compounds on vascular cells, after their accumulation in the arterial wall and in the atherosclerotic plaque.
Collapse
Affiliation(s)
- Simona Gargiulo
- Department of Clinical and Biological Sciences, University of Torino, San Luigi Hospital, 10043 Orbassano, Torino, Italy
| | - Gabriella Testa
- Department of Clinical and Biological Sciences, University of Torino, San Luigi Hospital, 10043 Orbassano, Torino, Italy
| | - Paola Gamba
- Department of Clinical and Biological Sciences, University of Torino, San Luigi Hospital, 10043 Orbassano, Torino, Italy
| | - Erica Staurenghi
- Department of Clinical and Biological Sciences, University of Torino, San Luigi Hospital, 10043 Orbassano, Torino, Italy
| | - Giuseppe Poli
- Department of Clinical and Biological Sciences, University of Torino, San Luigi Hospital, 10043 Orbassano, Torino, Italy
| | - Gabriella Leonarduzzi
- Department of Clinical and Biological Sciences, University of Torino, San Luigi Hospital, 10043 Orbassano, Torino, Italy.
| |
Collapse
|
24
|
Zhang M, Zhu H, Ding Y, Liu Z, Cai Z, Zou MH. AMP-activated protein kinase α1 promotes atherogenesis by increasing monocyte-to-macrophage differentiation. J Biol Chem 2017; 292:7888-7903. [PMID: 28330873 PMCID: PMC5427268 DOI: 10.1074/jbc.m117.779447] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 03/20/2017] [Indexed: 02/01/2023] Open
Abstract
Monocyte-to-macrophage differentiation, which can be initiated by physiological or atherogenic factors, is a pivotal process in atherogenesis, a disorder in which monocytes adhere to endothelial cells and subsequently migrate into the subendothelial spaces, where they differentiate into macrophages and macrophage-derived foam cells and cause atherosclerotic lesions. However, the monocyte-differentiation signaling pathways that are activated by atherogenic factors are poorly defined. Here we report that the AMP-activated protein kinase α1 (AMPKα1) in monocytes promotes atherosclerosis by increasing monocyte differentiation and survival. Exposure of monocytes to oxidized low-density lipoprotein, 7-ketocholesterol, phorbol 12-myristate 13-acetate, or macrophage colony-stimulated factor (M-CSF) significantly activated AMPK and promoted monocyte-to-macrophage differentiation. M-CSF-activated AMPK is via M-CSF receptor-dependent reactive oxygen species production. Consistently, genetic deletion of AMPKα1 or pharmacological inhibition of AMPK blunted monocyte-to-macrophage differentiation and promoted monocyte/macrophage apoptosis. Compared with apolipoprotein E knock-out (ApoE-/-) mice, which show impaired clearing of plasma lipoproteins and spontaneously develop atherosclerosis, ApoE-/-/AMPKα1-/- mice showed reduced sizes of atherosclerotic lesions and lesser numbers of macrophages in the lesions. Furthermore, aortic lesions were decreased in ApoE-/- mice transplanted with ApoE-/-/AMPKα1-/- bone marrow and in myeloid-specific AMPKα1-deficient ApoE-/- mice. Finally, rapamycin treatment, which abolished delayed monocyte differentiation in ApoE-/-/AMPKα1-/- mice, lost its atherosclerosis-lowering effects in these mice. Mechanistically, we found that AMPKα1 regulates FoxO3-dependent expression of both LC3 and ULK1, which are two important autophagy-related markers. Rapamycin treatment increased FoxO3 activity as well as LC3 and ULK1 expressions in macrophages from AMPKα1-/- mice. Our results reveal that AMPKα1 deficiency impairs autophagy-mediated monocyte differentiation and decreases monocyte/macrophage survival, which attenuates atherosclerosis in ApoE-/- mice in vivo.
Collapse
Affiliation(s)
- Miao Zhang
- From the Section of Molecular Medicine, Department of Medicine, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma 73104 and
| | - Huaiping Zhu
- the Center for Molecular and Translational Medicine, Georgia State University, Atlanta, Georgia 30302-5035
| | - Ye Ding
- the Center for Molecular and Translational Medicine, Georgia State University, Atlanta, Georgia 30302-5035
| | - Zhaoyu Liu
- the Center for Molecular and Translational Medicine, Georgia State University, Atlanta, Georgia 30302-5035
| | - Zhejun Cai
- the Center for Molecular and Translational Medicine, Georgia State University, Atlanta, Georgia 30302-5035
| | - Ming-Hui Zou
- the Center for Molecular and Translational Medicine, Georgia State University, Atlanta, Georgia 30302-5035
| |
Collapse
|
25
|
Verescakova H, Ambrozova G, Kubala L, Perecko T, Koudelka A, Vasicek O, Rudolph TK, Klinke A, Woodcock SR, Freeman BA, Pekarova M. Nitro-oleic acid regulates growth factor-induced differentiation of bone marrow-derived macrophages. Free Radic Biol Med 2017; 104:10-19. [PMID: 28063941 PMCID: PMC5329068 DOI: 10.1016/j.freeradbiomed.2017.01.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 12/20/2016] [Accepted: 01/03/2017] [Indexed: 01/05/2023]
Abstract
Many diseases accompanied by chronic inflammation are connected with dysregulated activation of macrophage subpopulations. Recently, we reported that nitro-fatty acids (NO2-FAs), products of metabolic and inflammatory reactions of nitric oxide and nitrite, modulate macrophage and other immune cell functions. Bone marrow cell suspensions were isolated from mice and supplemented with macrophage colony-stimulating factor (M-CSF) or granulocyte-macrophage colony-stimulating factor (GM-CSF) in combination with NO2-OA for different times. RAW 264.7 macrophages were used for short-term (1-5min) experiments. We discovered that NO2-OA reduces cell numbers, cell colony formation, and proliferation of macrophages differentiated with colony-stimulating factors (CSFs), all in the absence of toxicity. In a case of GM-CSF-induced bone marrow-derived macrophages (BMMs), NO2-OA acts via downregulation of signal transducer and activator of transcription 5 and extracellular signal-regulated kinase (ERK) activation. In the case of M-CSF-induced BMMs, NO2-OA decreases activation of M-CSFR and activation of related PI3K and ERK. Additionally, NO2-OA also attenuates activation of BMMs. In aggregate, we demonstrate that NO2-OA regulates the process of macrophage differentiation and that NO2-FAs represent a promising therapeutic tool in the treatment of inflammatory pathologies linked with increased accumulation of macrophages in inflamed tissues.
Collapse
Affiliation(s)
- Hana Verescakova
- Institute of Biophysics, Academy of Sciences of the Czech Republic, Brno, Czechia
| | - Gabriela Ambrozova
- Institute of Biophysics, Academy of Sciences of the Czech Republic, Brno, Czechia; International Clinical Research Center - Center of Biomolecular and Cellular Engineering, St. Anne's University Hospital, Brno, Czechia
| | - Lukas Kubala
- Institute of Biophysics, Academy of Sciences of the Czech Republic, Brno, Czechia; International Clinical Research Center - Center of Biomolecular and Cellular Engineering, St. Anne's University Hospital, Brno, Czechia
| | - Tomas Perecko
- Institute of Biophysics, Academy of Sciences of the Czech Republic, Brno, Czechia; International Clinical Research Center - Center of Biomolecular and Cellular Engineering, St. Anne's University Hospital, Brno, Czechia
| | - Adolf Koudelka
- Institute of Biophysics, Academy of Sciences of the Czech Republic, Brno, Czechia; International Clinical Research Center - Center of Biomolecular and Cellular Engineering, St. Anne's University Hospital, Brno, Czechia; Department of Animal Physiology and Immunology, Masaryk University, Brno, Czechia
| | - Ondrej Vasicek
- Institute of Biophysics, Academy of Sciences of the Czech Republic, Brno, Czechia; International Clinical Research Center - Center of Biomolecular and Cellular Engineering, St. Anne's University Hospital, Brno, Czechia
| | - Tanja K Rudolph
- Heart Centre, University Hospital of Cologne, Cologne, Germany
| | - Anna Klinke
- International Clinical Research Center - Center of Biomolecular and Cellular Engineering, St. Anne's University Hospital, Brno, Czechia; Heart Centre, University Hospital of Cologne, Cologne, Germany
| | - Steven R Woodcock
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Bruce A Freeman
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Michaela Pekarova
- Institute of Biophysics, Academy of Sciences of the Czech Republic, Brno, Czechia; International Clinical Research Center - Center of Biomolecular and Cellular Engineering, St. Anne's University Hospital, Brno, Czechia.
| |
Collapse
|
26
|
Abstract
In this chapter, we discuss the manner through which the immune system regulates the cardiovascular system in health and disease. We define the cardiovascular system and elements of atherosclerotic disease, the main focus in this chapter. Herein we elaborate on the disease process that can result in myocardial infarction (heart attack), ischaemic stroke and peripheral arterial disease. We have discussed broadly the homeostatic mechanisms in place that help autoregulate the cardiovascular system including the vital role of cholesterol and lipid clearance as well as the role lipid homeostasis plays in cardiovascular disease in the context of atherosclerosis. We then elaborate on the role played by the immune system in this setting, namely, major players from the innate and adaptive immune system, as well as discussing in greater detail specifically the role played by monocytes and macrophages.This chapter should represent an overview of the role played by the immune system in cardiovascular homeostasis; however further reading of the references cited can expand the reader's knowledge of the detail, and we point readers to many excellent reviews which summarise individual immune systems and their role in cardiovascular disease.
Collapse
Affiliation(s)
- Mohammed Shamim Rahman
- Division of Immunology and Inflammation, Department of Medicine, Imperial College London, London, UK
| | - Kevin Woollard
- Division of Immunology and Inflammation, Department of Medicine, Imperial College London, London, UK.
| |
Collapse
|
27
|
Thrombomodulin regulates monocye differentiation via PKCδ and ERK1/2 pathway in vitro and in atherosclerotic artery. Sci Rep 2016; 6:38421. [PMID: 27910925 PMCID: PMC5133669 DOI: 10.1038/srep38421] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 11/08/2016] [Indexed: 01/31/2023] Open
Abstract
Thrombomodulin (TM) modulates the activation of protein C and coagulation. Additionally, TM regulates monocyte migration and inflammation. However, its role on monocyte differentiation is still unknown. We investigated the effects of TM on monocyte differentiation. First, we found that TM was increased when THP-1 cells were treated with phorbol-12-myristate-13-acetate (PMA). Overexpression of TM enhanced the macrophage markers, CD14 and CD68 expression in PMA-induced THP-1. TM siRNA depressed the PMA-induced increase of p21Cip1/WAF1 via ERK1/2-NF-kB p65 signaling. TM regulated cytoskeletal reorganization via its interaction with paxillin, cofilin, LIMK1, and PYK2. In addition, PMA-induced p21Cip1/WAF1 expression, CD14-positive cell labeling intensity and ERK1/2 phosphorylation were markedly inhibited when protein kinase C-δ (PKCδ) was knocked down. We identified that TM directly interacts with PKCδ. PKCδ was highly expressed in human atherosclerotic arteries and colocalized with TM in CD68-positive infiltrated macrophages of plaques, indicating that the coordination between TM and PKCδ in macrophages participated in atherogenesis. TM may act as a scaffold for PKCδ docking, which keeps PKCδ in the region close to the monocyte membrane to promote the activation of ERK1/2. Taken together, our findings suggest that TM-PKCδ interaction may contribute to cardiovascular disorders by affecting monocye differentiation, which may develop future therapeutic applications.
Collapse
|
28
|
Therapeutic Strategies for Oxidative Stress-Related Cardiovascular Diseases: Removal of Excess Reactive Oxygen Species in Adult Stem Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:2483163. [PMID: 27668035 PMCID: PMC5030421 DOI: 10.1155/2016/2483163] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 08/17/2016] [Indexed: 02/07/2023]
Abstract
Accumulating evidence indicates that acute and chronic uncontrolled overproduction of oxidative stress-related factors including reactive oxygen species (ROS) causes cardiovascular diseases (CVDs), atherosclerosis, and diabetes. Moreover ROS mediate various signaling pathways underlying vascular inflammation in ischemic tissues. With respect to stem cell-based therapy, several studies clearly indicate that modulating antioxidant production at cellular levels enhances stem/progenitor cell functionalities, including proliferation, long-term survival in ischemic tissues, and complete differentiation of transplanted cells into mature vascular cells. Recently emerging therapeutic strategies involving adult stem cells, including endothelial progenitor cells (EPCs), for treating ischemic CVDs have highlighted the need to control intracellular ROS production, because it critically affects the replicative senescence of ex vivo expanded therapeutic cells. Better understanding of the complexity of cellular ROS in stem cell biology might improve cell survival in ischemic tissues and enhance the regenerative potentials of transplanted stem/progenitor cells. In this review, we will discuss the nature and sources of ROS, drug-based therapeutic strategies for scavenging ROS, and EPC based therapeutic strategies for treating oxidative stress-related CVDs. Furthermore, we will discuss whether primed EPCs pretreated with natural ROS-scavenging compounds are crucial and promising therapeutic strategies for vascular repair.
Collapse
|
29
|
Akerele OA, Cheema SK. Fatty acyl composition of lysophosphatidylcholine is important in atherosclerosis. Med Hypotheses 2015; 85:754-60. [PMID: 26604024 DOI: 10.1016/j.mehy.2015.10.013] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 09/24/2015] [Accepted: 10/14/2015] [Indexed: 12/11/2022]
Abstract
Atherosclerosis is a major cause of death for mankind. Although the pathophysiology of atherosclerosis is a complex and multifactorial process, growing body of evidence has identified phospholipids-mediated signaling as an important factor in the induction and progression of atherosclerosis. Lysophosphatidylcholine (LPC) is a major phospholipid in oxidized low-density lipoprotein, and is generally considered to be atherogenic. However, some studies have shown anti-atherogenic properties of LPC. The controversial findings surrounding the pro- or anti-atherogenic properties of LPC appear to be due to the chain length and the degree of saturation of the fatty acyl moiety of LPC. Studies have suggested that the presence of omega (n)-polyunsaturated fatty acids (PUFA) at the sn-1 position of LPC modulates the inflammatory response thereby making LPC anti-atherogenic. We have recently shown that feeding a diet high in n-3 PUFA resulted in the enrichment of LPC in both plasma and liver of C57BL/6 mice with n-3 PUFA. Others have also shown that supplementation with fish oil leads to preferential incorporation of n-3 PUFA into LPC. We also found that plasma obtained from mice fed a diet high in n-3 PUFA showed higher cholesterol efflux capacity compared to animals fed a low n-3 PUFA diet, despite no changes in high-density lipoprotein concentrations. We are therefore hypothesizing that n-3 PUFA enriched LPC has anti-atherogenic properties by promoting cholesterol efflux from macrophages and by reducing inflammation. Our anticipated long term objective is to establish that the fatty acyl moiety of LPC can be used as a potential biomarker for the risk of developing atherosclerosis. Validating this hypothesis would have a substantial impact on the public health with respect to early diagnosis of cardiovascular risks, and designing dietary based therapeutic strategies for the prevention and management of atherosclerosis and other heart related diseases.
Collapse
|
30
|
Abstract
Atherosclerosis, the build-up of occlusive, lipid-rich plaques in arterial walls, is a focal trigger of chronic coronary, intracranial, and peripheral arterial diseases, which together account for the leading causes of death worldwide. Although the directed treatment of atherosclerotic plaques remains elusive, macrophages are a natural target for new interventions because they are recruited to lipid-rich lesions, actively internalize modified lipids, and convert to foam cells with diseased phenotypes. In this work, we present a nanomedicine platform to counteract plaque development based on two building blocks: first, at the single macrophage level, sugar-based amphiphilic macromolecules (AMs) were designed to competitively block oxidized lipid uptake via scavenger receptors on macrophages; second, for sustained lesion-level intervention, AMs were fabricated into serum-stable core/shell nanoparticles (NPs) to rapidly associate with plaques and inhibit disease progression in vivo. An AM library was designed and fabricated into NP compositions that showed high binding and down-regulation of both MSR1 and CD36 scavenger receptors, yielding minimal accumulation of oxidized lipids. When intravenously administered to a mouse model of cardiovascular disease, these AM NPs showed a pronounced increase in lesion association compared with the control nanoparticles, causing a significant reduction in neointimal hyperplasia, lipid burden, cholesterol clefts, and overall plaque occlusion. Thus, synthetic macromolecules configured as NPs are not only effectively mobilized to lipid-rich lesions but can also be deployed to counteract atheroinflammatory vascular diseases, highlighting the promise of nanomedicines for hyperlipidemic and metabolic syndromes.
Collapse
|
31
|
Kang H, Park SH, Yun JM, Nam TG, Kim YE, Kim DO, Kim YJ. Effect of cinnamon water extract on monocyte-to-macrophage differentiation and scavenger receptor activity. Altern Ther Health Med 2014; 14:90. [PMID: 24602512 PMCID: PMC3973967 DOI: 10.1186/1472-6882-14-90] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2013] [Accepted: 02/28/2014] [Indexed: 12/14/2022]
Abstract
Background Water soluble cinnamon extract has been shown to increase insulin sensitivity and modulate macrophage activation, a desirable trait for the management of obesity or atherosclerosis. Our present study investigated whether cinnamon water extract (CWE) may influence the differentiation of monocytes into macrophages and the activity of macrophage scavenger receptors, commonly observed in atherosclerotic lesions. Methods We investigated the effect of CWE on the expression of various surface markers and the uptake of acetylated low density lipoprotein (LDL) in phorbol-12-myristate-13-acetate (PMA)-stimulated THP-1 cells. The protein levels of PMA or macrophage-colony stimulating factor (M-CSF)-stimulated type 1 macrophage scavenger receptor (SRA) were analyzed. Finally, the role of extracellar signal-related kinase (ERK) 1/2 in SRA synthesis and the effect of CWE on PMA-stimulated ERK1/2 were determined. Results CWE inhibited the differentiation of monocyte by decreasing the expression of CD11b, CD36 and SRA and the uptake of acetyl LDL. CWE suppressed the upregulation of SRA by M-CSF and modulated ERK1/2 activity, which was required for PMA-induced SRA synthesis. Conclusions Our results demonstrate that CWE was able to interfere with monocyte differentiation and macrophage scavenger activity, indicating its potential in preventing the development of atherosclerotic lesions.
Collapse
|
32
|
Kiyan Y, Tkachuk S, Hilfiker-Kleiner D, Haller H, Fuhrman B, Dumler I. oxLDL induces inflammatory responses in vascular smooth muscle cells via urokinase receptor association with CD36 and TLR4. J Mol Cell Cardiol 2013; 66:72-82. [PMID: 24239845 DOI: 10.1016/j.yjmcc.2013.11.005] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Revised: 10/31/2013] [Accepted: 11/05/2013] [Indexed: 10/26/2022]
Abstract
The pathogenesis of atherosclerosis involves an imbalanced lipid metabolism and a deregulated immune response culminating in chronic inflammation of the arterial wall. Recent studies show that endogenous ligands, such as modified plasma lipoproteins, can trigger pattern recognition receptors (PRR) of innate immunity for cellular and humoral reactions. The underlying molecular pathways remain less explored. In this study, we investigated the mechanisms of inflammatory effects of oxidized low-density lipoproteins (oxLDL) on human primary coronary artery smooth muscle cells (VSMC). We show that already low concentration of oxLDL initiated atherogenic signals triggering VSMC transition to proinflammatory phenotype. oxLDL impaired the expression of contractile proteins and myocardin in VSMC and initiated changes in cell functional responses, including expression of proinflammatory molecules. The effects of oxLDL were abolished by downregulation of the multifunctional urokinase receptor (uPAR). In response to oxLDL uPAR associated with CD36 and TLR4, the two main PRR for both pathogen and endogenous ligands. We demonstrate that uPAR association with CD36 and TLR4 mediated oxLDL-induced and NF-κB-dependent G-CSF and GM-CSF expression and changes in VSMC contractile proteins. uPAR-mediated release of G-CSF and GM-CSF by VSMC affected macrophage behavior and production of MCP-1. We provide evidence for functional relevance of our in vitro findings to in vivo human atherosclerotic tissues. Our data imply uPAR as a part of a PRR cluster interfering structurally and functionally with CD36 and TLR4 and responding to endogenous atherogenic ligands. They further point to specific function of each component of this cluster in mediating the ultimate signaling pattern.
Collapse
Affiliation(s)
- Yulia Kiyan
- Nephrology Department, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
| | - Sergey Tkachuk
- Nephrology Department, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
| | - Denise Hilfiker-Kleiner
- Cardiology Department, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
| | - Hermann Haller
- Nephrology Department, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
| | - Bianca Fuhrman
- The Lipid Research Laboratory, Technion Faculty of Medicine, and Rambam Medical Center, Haifa, Israel.
| | - Inna Dumler
- Nephrology Department, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
| |
Collapse
|
33
|
Urokinase-type plasminogen activator (uPA) modulates monocyte-to-macrophage differentiation and prevents Ox-LDL-induced macrophage apoptosis. Atherosclerosis 2013; 231:29-38. [DOI: 10.1016/j.atherosclerosis.2013.08.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Revised: 08/01/2013] [Accepted: 08/19/2013] [Indexed: 01/09/2023]
|
34
|
Electronegative LDL: a circulating modified LDL with a role in inflammation. Mediators Inflamm 2013; 2013:181324. [PMID: 24062611 PMCID: PMC3766570 DOI: 10.1155/2013/181324] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Accepted: 07/19/2013] [Indexed: 12/13/2022] Open
Abstract
Electronegative low density lipoprotein (LDL(−)) is a minor modified fraction of LDL found in blood. It comprises a heterogeneous population of LDL particles modified by various mechanisms sharing as a common feature increased electronegativity. Modification by oxidation is one of these mechanisms. LDL(−) has inflammatory properties similar to those of oxidized LDL (oxLDL), such as inflammatory cytokine release in leukocytes and endothelial cells. However, in contrast with oxLDL, LDL(−) also has some anti-inflammatory effects on cultured cells. The inflammatory and anti-inflammatory properties ascribed to LDL(−) suggest that it could have a dual biological effect.
Collapse
|
35
|
Oxysterols induce transition of monocytic cells to phenotypically mature dendritic cell-like cells. Biochem Biophys Res Commun 2013; 438:161-8. [DOI: 10.1016/j.bbrc.2013.07.046] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Accepted: 07/12/2013] [Indexed: 12/27/2022]
|
36
|
Lewis DR, Kholodovych V, Tomasini MD, Abdelhamid D, Petersen LK, Welsh WJ, Uhrich KE, Moghe PV. In silico design of anti-atherogenic biomaterials. Biomaterials 2013; 34:7950-9. [PMID: 23891521 DOI: 10.1016/j.biomaterials.2013.07.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Accepted: 07/01/2013] [Indexed: 01/10/2023]
Abstract
Atherogenesis, the uncontrolled deposition of modified lipoproteins in inflamed arteries, serves as a focal trigger of cardiovascular disease (CVD). Polymeric biomaterials have been envisioned to counteract atherogenesis based on their ability to repress scavenger mediated uptake of oxidized lipoprotein (oxLDL) in macrophages. Following the conceptualization in our laboratories of a new library of amphiphilic macromolecules (AMs), assembled from sugar backbones, aliphatic chains and poly(ethylene glycol) tails, a more rational approach is necessary to parse the diverse features such as charge, hydrophobicity, sugar composition and stereochemistry. In this study, we advance a computational biomaterials design approach to screen and elucidate anti-atherogenic biomaterials with high efficacy. AMs were quantified in terms of not only 1D (molecular formula) and 2D (molecular connectivity) descriptors, but also new 3D (molecular geometry) descriptors of AMs modeled by coarse-grained molecular dynamics (MD) followed by all-atom MD simulations. Quantitative structure-activity relationship (QSAR) models for anti-atherogenic activity were then constructed by screening a total of 1164 descriptors against the corresponding, experimentally measured potency of AM inhibition of oxLDL uptake in human monocyte-derived macrophages. Five key descriptors were identified to provide a strong linear correlation between the predicted and observed anti-atherogenic activity values, and were then used to correctly forecast the efficacy of three newly designed AMs. Thus, a new ligand-based drug design framework was successfully adapted to computationally screen and design biomaterials with cardiovascular therapeutic properties.
Collapse
Affiliation(s)
- Daniel R Lewis
- Department of Chemical and Biochemical Engineering, Rutgers University, NJ 08854, USA
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Bzowska M, Nogieć A, Skrzeczyńska-Moncznik J, Mickowska B, Guzik K, Pryjma J. Oxidized LDLs inhibit TLR-induced IL-10 production by monocytes: a new aspect of pathogen-accelerated atherosclerosis. Inflammation 2013; 35:1567-84. [PMID: 22556042 PMCID: PMC3397235 DOI: 10.1007/s10753-012-9472-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
It is widely accepted that oxidized low-density lipoproteins and local infections or endotoxins in circulation contribute to chronic inflammatory process at all stages of atherosclerosis. The hallmark cells of atherosclerotic lesions-monocytes and macrophages-are able to detect and integrate complex signals derived from lipoproteins and pathogens, and respond with a spectrum of immunoregulatory cytokines. In this study, we show strong inhibitory effect of oxLDLs on anti-inflammatory interleukin-10 production by monocytes responding to TLR2 and TLR4 ligands. In contrast, pro-inflammatory tumor necrosis factor secretion was even slightly increased, when stimulated with lipopolysaccharide from Porphyromonas gingivalis-an oral pathogen associated with atherosclerosis. The oxLDLs modulatory activity may be explained by altered recognition of pathogen-associated molecular patterns, which involves serum proteins, particularly vitronectin. We also suggest an interaction between vitronectin receptor, CD11b, and TLR2. The presented data support a novel pathway for pathogen-accelerated atherosclerosis, which relies on oxidized low-density lipoprotein-mediated modulation of anti-inflammatory response to TLR ligands.
Collapse
Affiliation(s)
- Małgorzata Bzowska
- Department of Immunology, Faculty of Biochemistry, Biophysics, and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland
| | - Anna Nogieć
- Department of Immunology, Faculty of Biochemistry, Biophysics, and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland
| | - Joanna Skrzeczyńska-Moncznik
- Department of Immunology, Faculty of Biochemistry, Biophysics, and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland
| | - Barbara Mickowska
- Malopolska Centre of Food Monitoring and Certification, Faculty of Food Technology, Agricultural University, Balicka 122, 30-149 Kraków, Poland
| | - Krzysztof Guzik
- Department of Immunology, Faculty of Biochemistry, Biophysics, and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland
| | - Juliusz Pryjma
- Department of Immunology, Faculty of Biochemistry, Biophysics, and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland
| |
Collapse
|
38
|
Oxidized LDLs inhibit TLR-induced IL-10 production by monocytes: a new aspect of pathogen-accelerated atherosclerosis. Inflammation 2013. [PMID: 22556042 DOI: 10.1007/s110753-012-9472-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
It is widely accepted that oxidized low-density lipoproteins and local infections or endotoxins in circulation contribute to chronic inflammatory process at all stages of atherosclerosis. The hallmark cells of atherosclerotic lesions-monocytes and macrophages-are able to detect and integrate complex signals derived from lipoproteins and pathogens, and respond with a spectrum of immunoregulatory cytokines. In this study, we show strong inhibitory effect of oxLDLs on anti-inflammatory interleukin-10 production by monocytes responding to TLR2 and TLR4 ligands. In contrast, pro-inflammatory tumor necrosis factor secretion was even slightly increased, when stimulated with lipopolysaccharide from Porphyromonas gingivalis-an oral pathogen associated with atherosclerosis. The oxLDLs modulatory activity may be explained by altered recognition of pathogen-associated molecular patterns, which involves serum proteins, particularly vitronectin. We also suggest an interaction between vitronectin receptor, CD11b, and TLR2. The presented data support a novel pathway for pathogen-accelerated atherosclerosis, which relies on oxidized low-density lipoprotein-mediated modulation of anti-inflammatory response to TLR ligands.
Collapse
|
39
|
Gao D, Pararasa C, Dunston CR, Bailey CJ, Griffiths HR. Palmitate promotes monocyte atherogenicity via de novo ceramide synthesis. Free Radic Biol Med 2012; 53:796-806. [PMID: 22640955 DOI: 10.1016/j.freeradbiomed.2012.05.026] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2011] [Revised: 04/30/2012] [Accepted: 05/15/2012] [Indexed: 11/21/2022]
Abstract
Elevated plasma free fatty acids (FAs) are associated with increased risk of cardiovascular disease. This study investigates the effects of the saturated FA palmitate and unsaturated FA oleate on monocyte phenotype and function. Incubation of human U937 and THP-1 monocytes with palmitate for 24h increased cell surface expression of integrin CD11b and scavenger receptor CD36 in a concentration-dependent manner with some decrease in mitochondrial reducing capacity at high concentration (300 μM). Monocytes incubated with palmitate, but not oleate, showed increased uptake of oxidized LDL and increased adhesion to rat aortic endothelium, particularly at bifurcations. The palmitate-induced increase in CD11b and CD36 expression was associated with increased cellular C16 ceramide and sphingomyelin, loss of reduced glutathione, and increased reactive oxygen species (ROS). Increased monocyte surface CD11b and CD36 was inhibited by fumonisin B1, an inhibitor of de novo ceramide synthesis, but not by the superoxide dismutase mimetic MnTBap. In contrast, MnTBap prevented the mitochondrial ROS increase and metabolic inhibition due to 300μM palmitate. This study demonstrates that in viable monocytes, palmitate but not oleate increases expression of surface CD11b and CD36. Palmitate increases monocyte adhesion to the aortic wall and promotes uptake of oxidized LDL and this involves de novo ceramide synthesis.
Collapse
Affiliation(s)
- Dan Gao
- School of Life and Health Sciences, Aston University, Birmingham B4 7ET, UK
| | | | | | | | | |
Collapse
|
40
|
Bhatt PM, Lewis CJ, House DL, Keller CM, Kohn LD, Silver MJ, McCall KD, Goetz DJ, Malgor R. Increased Wnt5a mRNA Expression in Advanced Atherosclerotic Lesions, and Oxidized LDL Treated Human Monocyte-Derived Macrophages. ACTA ACUST UNITED AC 2012; 5:1-7. [PMID: 25530821 PMCID: PMC4270053 DOI: 10.2174/1877382601205010001] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Objective Wnt5a is a secreted glycoprotein highly present in atherosclerotic lesions. Uptake of oxidized-low density lipoprotein (ox-LDL) by monocytes/macrophages plays a critical role in atherosclerosis. The objective of this study was to determine if Wnt5a mRNA expression correlates with the severity of atherosclerotic lesions, and if, ox-LDL can induce Wnt5a mRNA in macrophages. Methods Wnt5a mRNA in tissue sections from carotid arteries of patients undergoing endarterectomy was quantified via RT-PCR and correlated with plaque severity. Human monocyte-derived macrophages and differentiated THP-1 cells, a human monocytic cell line, were treated with ox-LDL or native-LDL. Subsequently, Wnt5a transcripts were quantified by RT-PCR. Results Regions of the arteries with more severe plaques had detectable and significant levels of Wnt5a mRNA, while regions of the arteries containing less vulnerable plaques had low or non-detectable Wnt5a. Ox-LDL, but not native-LDL, induced Wnt5a mRNA in both human monocyte-derived macrophages and differentiated THP-1 cells. Conclusion Our results demonstrate that the expression of Wnt5a correlates with the severity of atherosclerotic lesions, and that ox-LDL induces Wnt5a mRNA expression in human macrophages. These findings are consistent with the hypothesis that Wnt5a plays a critical role in atherosclerosis progression and that a source of Wnt5a is ox-LDL stimulated macrophages.
Collapse
Affiliation(s)
- Pooja M Bhatt
- Department of Biological Sciences, Molecular and Cellular Biology Graduate Program, Ohio University, Athens, Ohio
| | - Christopher J Lewis
- Department of Biological Sciences, Molecular and Cellular Biology Graduate Program, Ohio University, Athens, Ohio
| | - Denise L House
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio
| | - Chad M Keller
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio
| | - Leonard D Kohn
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio
| | | | - Kelly D McCall
- Department of Specialty Medicine, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio
| | - Douglas J Goetz
- Department of Chemical and Biomolecular Engineering, Ohio University, Athens, Ohio, USA
| | - Ramiro Malgor
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio
| |
Collapse
|
41
|
Leonarduzzi G, Gamba P, Gargiulo S, Biasi F, Poli G. Inflammation-related gene expression by lipid oxidation-derived products in the progression of atherosclerosis. Free Radic Biol Med 2012; 52:19-34. [PMID: 22037514 DOI: 10.1016/j.freeradbiomed.2011.09.031] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2011] [Revised: 09/16/2011] [Accepted: 09/24/2011] [Indexed: 12/31/2022]
Abstract
Vascular areas of atherosclerotic development persist in a state of inflammation, and any further inflammatory stimulus in the subintimal area elicits a proatherogenic response; this alters the behavior of the artery wall cells and recruits further inflammatory cells. In association with the inflammatory response, oxidative events are also involved in the development of atherosclerotic plaques. It is now unanimously recognized that lipid oxidation-derived products are key players in the initiation and progression of atherosclerotic lesions. Oxidized lipids, derived from oxidatively modified low-density lipoproteins (LDLs), which accumulate in the intima, strongly modulate inflammation-related gene expression, through involvement of various signaling pathways. In addition, considerable evidence supports a proatherogenic role of a large group of potent bioactive lipids called eicosanoids, which derive from oxidation of arachidonic acid, a component of membrane phospholipids. Of note, LDL lipid oxidation products might regulate eicosanoid production, modulating the enzymatic degradation of arachidonic acid by cyclooxygenases and lipoxygenases; these enzymes might also directly contribute to LDL oxidation. This review provides a comprehensive overview of current knowledge on signal transduction pathways and inflammatory gene expression, modulated by lipid oxidation-derived products, in the progression of atherosclerosis.
Collapse
|
42
|
Mat MC, Mohamed AS, Hamid SS. Primary human monocyte differentiation regulated by Nigella sativa pressed oil. Lipids Health Dis 2011; 10:216. [PMID: 22104447 PMCID: PMC3280944 DOI: 10.1186/1476-511x-10-216] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2011] [Accepted: 11/21/2011] [Indexed: 02/02/2023] Open
Abstract
Background Oxidized low density lipoprotein plays an important role in development of foam cells in atherosclerosis. The study was focused on regulation of primary human monocyte growth and CD11b expression in presence of Nigella sativa oil. Methods Primary human monocytes were isolated from whole blood and grown at 37°C and 5% CO2 saturation for five days prior to treatment with Nigella sativa oil. The cells were plated and washed before treatment with ox-LDL (10 μg/ml) as positive control and combined treatment of ox-LDL (10 μg/ml) and (140 ng/ml) Nigella sativa oil. The growth progression was monitored every 24 hours for 3 days. Results Macrophages showed reduced growth in comparison to monocytes 24 hours after treatment with Nigella sativa oil. The mean cell diameter was significantly different between untreated and treated condition in monocytes and macrophages (p < 0.001). Similarly, intracellular lipid accumulation was hindered in combined treatment with Nigella sativa oil. This was further supported by cell surface expression analysis, where CD11b was markedly reduced in cells treated with combination oxLDL and Nigella sativa oil compared to oxLDL alone. More cells differentiated into macrophage-like cells when monocytes were supplemented with oxidized LDL alone. Conclusions The finding provides preliminary evidence on regulation of cell growth and differentiation in monocyte and monocyte-derived macrophages by Nigella sativa oil. Further investigations need to be conducted to explain its mechanism in human monocyte.
Collapse
Affiliation(s)
- Mahaya C Mat
- Advanced Medical and Dental Institute, Universiti Sains Malaysia, Lot 1-8, PersiaranSeksyen 4/1, Bandar Putra Bertam, Kepala Batas 13200, Penang, Malaysia
| | | | | |
Collapse
|
43
|
Fuhrman B. The urokinase system in the pathogenesis of atherosclerosis. Atherosclerosis 2011; 222:8-14. [PMID: 22137664 DOI: 10.1016/j.atherosclerosis.2011.10.044] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2011] [Revised: 10/14/2011] [Accepted: 10/31/2011] [Indexed: 01/08/2023]
Abstract
Atherogenesis refers to the development of atheromatous plaques in the inner lining of the arteries. These atherosclerotic lesions are characterized by accumulation of monocyte-derived macrophage-foam cells loaded with cholesterol, which eventually undergo apoptotic death, leading finally to formation of the necrotic core of the plaque. Atheroma formation also involves the recruitment of smooth muscle cells (SMC) from the media into the intima, where they proliferate and form the neointima in a process called "remodeling". Cells in the advanced atherosclerotic plaques express high levels of the serine protease urokinase-type plasminogen activator (uPA) and its receptor (uPAR). uPA is a multi-functional multi-domain protein that is not only a regulator of fibrinolysis, but it is also associated with several acute and chronic pathologic conditions. uPA mediate the extracellular matrix (ECM) degradation, and plays a pivotal role in cell adhesion, migration and proliferation, during tissue remodeling. On cell surface uPA binds to the high affinity urokinase receptor, providing a strictly localized proteolysis of ECM proteins. The uPA/uPAR complex also activates intracellular signaling, thus regulating cellular function. An imbalance in the uPA/uPAR system leads to dis-orders in tissue structure and function. This review summarizes recent progress in understanding the role and mechanisms of the uPA/uPAR system in atherogenesis.
Collapse
Affiliation(s)
- Bianca Fuhrman
- The Lipid Research Laboratory, Technion Faculty of Medicine, Rambam Medical Center, Haifa 31096, Israel.
| |
Collapse
|
44
|
Corrêa CR, Dias-Melicio LA, Calvi SA, Lastória S, Soares AM. Activation of monocytes and cytokine production in patients with peripheral atherosclerosis obliterans. JOURNAL OF INFLAMMATION-LONDON 2011; 8:23. [PMID: 21875436 PMCID: PMC3182876 DOI: 10.1186/1476-9255-8-23] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2011] [Accepted: 08/29/2011] [Indexed: 11/21/2022]
Abstract
Background Arterial peripheral disease is a condition caused by the blocked blood flow resulting from arterial cholesterol deposits within the arms, legs and aorta. Studies have shown that macrophages in atherosclerotic plaque are highly activated, which makes these cells important antigen-presenting cells that develop a specific immune response, in which LDLox is the inducing antigen. As functional changes of cells which participate in the atherogenesis process may occur in the peripheral blood, the objectives of the present study were to evaluate plasma levels of anti-inflammatory and inflammatory cytokines including TNF-α, IFN-γ, interleukin-6 (IL-6), IL-10 and TGF-β in patients with peripheral arteriosclerosis obliterans, to assess the monocyte activation level in peripheral blood through the ability of these cells to release hydrogen peroxide (H2O2) and to develop fungicidal activity against Candida albicans (C. albicans) in vitro. Methods TNF-α, IFN-γ, IL-6, IL-10 and TGF-β from plasma of patients were detected by ELISA. Monocyte cultures activated in vitro with TNF-alpha and IFN-gamma were evaluated by fungicidal activity against C. albicans by culture plating and Colony Forming Unit (CFU) recovery, and by H2O2 production. Results Plasma levels of all cytokines were significantly higher in patients compared to those detected in control subjects. Control group monocytes did not release substantial levels of H2O2 in vitro, but these levels were significantly increased after activation with IFN-γ and TNF-α. Monocytes of patients, before and after activation, responded less than those of control subjects. Similar results were found when fungicidal activity was evaluated. The results seen in patients were always significantly smaller than among control subjects. Conclusions: The results revealed an unresponsiveness of patient monocytes in vitro probably due to the high activation process occurring in vivo as corroborated by high plasma cytokine levels.
Collapse
Affiliation(s)
- Camila R Corrêa
- Departamento de Microbiologia e Imunologia, UNESP - Univ Estadual Paulista, Instituto de Biociências - Campus Botucatu, CEP 18618-970, SP, Brasil.
| | | | | | | | | |
Collapse
|
45
|
Rosenblat M, Volkova N, Ward J, Aviram M. Paraoxonase 1 (PON1) inhibits monocyte-to-macrophage differentiation. Atherosclerosis 2011; 219:49-56. [PMID: 21798540 DOI: 10.1016/j.atherosclerosis.2011.06.054] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2011] [Revised: 06/09/2011] [Accepted: 06/29/2011] [Indexed: 11/16/2022]
Abstract
OBJECTIVE To analyze paraoxonase 1 (PON1) effect on monocyte-to-macrophage differentiation. METHODS AND RESULTS THP-1 monocytic cell-line and mouse peritoneal macrophages (MPM) were studied. Markers for monocytes differentiation included: morphological changes, CD11b and CD36 expression, and cellular oxidative stress. PON1KO MPM were more differentiated than control C57BL/6 MPM. Intraperitoneal injection of recombinant PON1 (rePON1) to C57BL/6 or to PON1KO mice significantly increased serum, MPM, and tissues PON1 activities. These effects were associated with a significant decrease in CD11b in C57BL/6 and PON1KO MPM (by 21% and 35%, respectively), in CD36 (by 35% and 38%, respectively), and in cellular total peroxides content (by 18% and 20%, respectively). rePON1 also significantly inhibited CD11b and CD36 expression, and cellular total peroxides during PMA-induced THP-1 monocytes differentiation, by 68%, 56% and 53%, respectively. Similar effects were observed upon using reconstituted HDL (rHDL) +rePON1, or human HDL +rePON1, in comparison to rHDL or to human HDL, as well as, HDL from C57BL/6 vs. PON1KO mice. Inhibition of monocyte-to-macrophage differentiation was demonstrated also by several dietary antioxidants such as vitamin E, gallic acid, or punicalagin (the major polyphenol in pomegranate). Whereas NADPH oxidase was not involved in PON1 anti-differentiation effect, mitochondrial complex I could be involved, as rotenone (complex I inhibitor) significantly decreased (by 77%) the expression of CD11b during THP-1 differentiation. Finally, blocking PON1 sulfhydryl group with N-ethylmalemide significantly attenuated PON1 inhibitory effect on THP-I monocyte-to-macrophage differentiation. CONCLUSION HDL-associated PON1 inhibits monocyte-to-macrophage differentiation, and this effect could be related to PON1 peroxidase-like activity which involves its free sulfhydryl group.
Collapse
Affiliation(s)
- Mira Rosenblat
- The Lipid Research Laboratory, Technion Faculty of Medicine, The Rappaport Family Institute for Research in the Medical Sciences, Rambam Medical Center, Haifa, Israel
| | | | | | | |
Collapse
|
46
|
Gupta H, Pawar D, Riva A, Bombardelli E, Morazzoni P. A randomized, double-blind, placebo-controlled trial to evaluate efficacy and tolerability of an optimized botanical combination in the management of patients with primary hypercholesterolemia and mixed dyslipidemia. Phytother Res 2011; 26:265-72. [PMID: 21674629 DOI: 10.1002/ptr.3542] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2010] [Revised: 03/30/2011] [Accepted: 04/17/2011] [Indexed: 11/12/2022]
Abstract
This study compared the efficacy and tolerability of an optimized botanical combination containing policosanol, tomato extract, orally bioavailable grape procyanidins and Oenothera biennis oil against placebo in the management of patients with primary hypercholesterolemia and mixed dyslipidemia. Such a combination is endowed with biological properties targeted to cholesterol control and vasoprotection. This randomized, double-blind, parallel-group trial consisted of a 6 week treatment period following 4 week baseline period, and a 2 week post-treatment follow-up. At baseline, both the groups were comparable to each other. Both the active treatment and the placebo group included 30 patients (active treatment: mean age 46.80 ± 7.43 years, nine males; placebo: mean age 45.50 ± 6.76 years, eight males). Significant reductions in the LDL-cholesterol (LDL-C; -17.33% from baseline, p < 0.001) and total cholesterol (TC; -13.38% from baseline, p < 0.0001) values over the treatment period were observed with the tested product. The treatment also resulted in reductions in C-reactive protein (CRP), malondialdehyde (MDA) and superoxide dismutase (SOD) values, which are indices of oxidative stress. This rational combination of different compounds is effective and safe in lowering the elevated LDL-C and TC values. It is also effective in the modulation of the oxidation indices values; however, a further long term study in a larger population would be needed in order to confirm these preliminary findings.
Collapse
Affiliation(s)
- H Gupta
- Department of Medicine, Grant Medical College Mumbai, Mumbai, India
| | | | | | | | | |
Collapse
|
47
|
Soehnlein O, Drechsler M, Hristov M, Weber C. Functional alterations of myeloid cell subsets in hyperlipidaemia: relevance for atherosclerosis. J Cell Mol Med 2009; 13:4293-303. [PMID: 19900213 PMCID: PMC4515047 DOI: 10.1111/j.1582-4934.2009.00965.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Atherosclerosis is a chronic inflammatory disease wherein the infiltration of myeloid cells of the vessel wall is a hallmark event. Lymphocytes, platelets and endothelial cells stand out as prominent suspects being involved in atherosclerosis. However, recent advances suggest a crucial role for myeloid leucocytes, specifically monocyte subsets, neutrophils, dendritic cells and endothelial progenitor cells. These cell types are not just rapidly recruited or already reside in the vascular wall, but also initiate and perpetuate core mechanisms in plaque formation and destabilization. Hyperlipidaemia is an independent risk factor for atherosclerosis. Herein, hyperlipidaemia skews myeloid cell haemostasis, phenotype and transcriptional regulation of pro-inflammatory factors ultimately promoting myeloid cell extravasation and atherosclerosis. We here review the role of myeloid cells in atherosclerosis as well as the effects of hyperlipidaemia on these cells.
Collapse
Affiliation(s)
- Oliver Soehnlein
- Institute for Molecular Cardiovascular Research, RWTH Aachen, Germany.
| | | | | | | |
Collapse
|
48
|
Retinoid X receptor agonists inhibit phorbol-12-myristate-13-acetate (PMA)-induced differentiation of monocytic THP-1 cells into macrophages. Mol Cell Biochem 2009; 335:283-9. [DOI: 10.1007/s11010-009-0278-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2009] [Accepted: 09/16/2009] [Indexed: 10/20/2022]
|
49
|
Spite M, Summers L, Porter TF, Srivastava S, Bhatnagar A, Serhan CN. Resolvin D1 controls inflammation initiated by glutathione-lipid conjugates formed during oxidative stress. Br J Pharmacol 2009; 158:1062-73. [PMID: 19422383 DOI: 10.1111/j.1476-5381.2009.00234.x] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND AND PURPOSE Inflammation is associated with oxidative stress and local generation of lipid peroxidation-derived aldehydes, such as 4-hydroxy-trans-2-nonenal (HNE). In most tissues, HNE is readily conjugated with glutathione and presently it is unknown whether glutathionyl-HNE (GS-HNE) plays a functional role in inflammation. Here, we sought to determine whether GS-HNE is a mediator of oxidative stress-initiated inflammation and if its actions can be regulated by the anti-inflammatory and pro-resolving lipid mediator, resolvin D1 (RvD1). EXPERIMENTAL APPROACH GS-HNE was administered intraperitoneally to mice and peritoneal lavages were assessed for leukocyte infiltration and lipid mediators were targeted by mediator-lipidomics. RvD1 was administered to mice treated with GS-HNE and leukocyte infiltration was assessed in the peritoneum. Superoxide production and CD11b modulation were measured in isolated human polymorphonuclear leukocytes incubated with GS-HNE. KEY RESULTS GS-HNE (1-10 microg) evoked infiltration of Gr-1(+) leukocytes into the peritoneum to form an inflammatory exudate. With isolated human polymorphonuclear leukocytes, GS-HNE stimulated both superoxide generation and CD11b expression. Among the lipid mediators, both cyclooxygenase- and lipoxygenase-derived pro-inflammatory eicosanoids, including prostaglandin E(2), leukotriene B(4) and cysteinyl leukotrienes, were generated in exudates of mice injected intraperitoneally with GS-HNE. RvD1, given i.v. in doses as low as 0.01-10.0 ng, sharply reduced GS-HNE-stimulated leukocyte infiltration ( approximately 30-70%). CONCLUSIONS AND IMPLICATIONS Glutathione conjugates of HNE, derived during oxidative stress, are pro-inflammatory in vivo. RvD1 protects against this oxidative stress-initiated inflammation.
Collapse
Affiliation(s)
- M Spite
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | |
Collapse
|
50
|
Saha P, Modarai B, Humphries J, Mattock K, Waltham M, Burnand KG, Smith A. The monocyte/macrophage as a therapeutic target in atherosclerosis. Curr Opin Pharmacol 2009; 9:109-18. [PMID: 19230773 DOI: 10.1016/j.coph.2008.12.017] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2008] [Revised: 12/22/2008] [Accepted: 12/31/2008] [Indexed: 12/31/2022]
Abstract
It is now clear that the monocyte/macrophage has a crucial role in the development of atherosclerosis. This cell appears to be involved in all stages of atherosclerotic plaque development and is increasingly seen as a candidate for therapeutic intervention and as a potential biomarker of disease progression and response to therapy. The main mechanisms related to the activity of the monocyte/macrophage that have been targeted for therapy are those that facilitate recruitment, cholesterol metabolism, inflammatory activity and oxidative stress. There is also increasing evidence that there is heterogeneity within the monocyte/macrophage population, which may have important implications for plaque development and regression. A better insight into how specific phenotypes may influence plaque progression should facilitate the development of novel methods of imaging and more refined treatments.
Collapse
Affiliation(s)
- Prakash Saha
- Academic Department of Surgery, Cardiovascular Division, King's College London, St Thomas' Hospital, London, United Kingdom
| | | | | | | | | | | | | |
Collapse
|