1
|
Tang Y, Tong W, Peng Y, Sun S. Targeting cholesterol-driven pyroptosis: a promising strategy for the prevention and treatment of atherosclerosis. Mol Biol Rep 2025; 52:459. [PMID: 40372511 DOI: 10.1007/s11033-025-10554-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2025] [Accepted: 04/28/2025] [Indexed: 05/16/2025]
Abstract
Funding Pyroptosis is a type of programmed cell death (PCD) pathway distinguished by inflammation. It is activated by specific inflammasomes. Once activated, it causes the physical breakdown of the cell, along with the discharge of pro-inflammatory cytokines, such as interleukin-1β (IL-1β) and interleukin-18 (IL-18). Abundant evidence has demonstrated the existence of pyroptotic cell death within atherosclerotic plaques, which has significance for the development of atherosclerosis (AS). As a result, pyroptosis has become a new and important topic in cardiovascular disease (CVD) research. Cholesterol, it is recognized to have a connection with inflammation, exerts a crucial function in the development process of AS, and has been linked to the initiation of pyroptosis. This review aims to briefly summarize the fundamental aspects of pyroptosis and the influence of cholesterol-related inflammation in AS. Additionally, this review will explore potential therapeutic approaches based on pyroptosis that could be utilized for the prevention and treatment of AS.
Collapse
Affiliation(s)
- Yuehong Tang
- Institute of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Wenjuan Tong
- Department of Gynecology and Obstetrics, First Affiliated Hospital, University of South China, Hengyang, Hunan, 421001, China
| | - Yujiao Peng
- Institute of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Shaowei Sun
- Institute of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, China.
| |
Collapse
|
2
|
Akyol O, Akyol S, Chou MC, Chen S, Liu CK, Selek S, Soares JC, Chen CH. Lipids and lipoproteins may play a role in the neuropathology of Alzheimer's disease. Front Neurosci 2023; 17:1275932. [PMID: 38033552 PMCID: PMC10687420 DOI: 10.3389/fnins.2023.1275932] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 10/30/2023] [Indexed: 12/02/2023] Open
Abstract
Alzheimer's disease (AD) and other classes of dementia are important public health problems with overwhelming social, physical, and financial effects for patients, society, and their families and caregivers. The pathophysiology of AD is poorly understood despite the extensive number of clinical and experimental studies. The brain's lipid-rich composition is linked to disturbances in lipid homeostasis, often associated with glucose and lipid abnormalities in various neurodegenerative diseases, including AD. Moreover, elevated low-density lipoprotein (LDL) cholesterol levels may be related to a higher probability of AD. Here, we hypothesize that lipids, and electronegative LDL (L5) in particular, may be involved in the pathophysiology of AD. Although changes in cholesterol, triglyceride, LDL, and glucose levels are seen in AD, the cause remains unknown. We believe that L5-the most electronegative subfraction of LDL-may be a crucial factor in understanding the involvement of lipids in AD pathology. LDL and L5 are internalized by cells through different receptors and mechanisms that trigger separate intracellular pathways. One of the receptors involved in L5 internalization, LOX-1, triggers apoptotic pathways. Aging is associated with dysregulation of lipid homeostasis, and it is believed that alterations in lipid metabolism contribute to the pathogenesis of AD. Proposed mechanisms of lipid dysregulation in AD include mitochondrial dysfunction, blood-brain barrier disease, neuronal signaling, inflammation, and oxidative stress, all of which lead ultimately to memory loss through deficiency of synaptic integration. Several lipid species and their receptors have essential functions in AD pathogenesis and may be potential biomarkers.
Collapse
Affiliation(s)
- Omer Akyol
- Molecular Cardiology, Vascular and Medicinal Research, The Texas Heart Institute, Houston, TX, United States
| | | | - Mei-Chuan Chou
- Department of Neurology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shioulan Chen
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ching-Kuan Liu
- Institute of Precision Medicine, College of Medicine, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Salih Selek
- Department of Psychiatry and Behavioral Sciences, UTHealth Houston McGovern Medical School, Houston, TX, United States
| | - Jair C. Soares
- Department of Psychiatry and Behavioral Sciences, UTHealth Houston McGovern Medical School, Houston, TX, United States
| | - Chu-Huang Chen
- Molecular Cardiology, Vascular and Medicinal Research, The Texas Heart Institute, Houston, TX, United States
| |
Collapse
|
3
|
Lorey MB, Youssef A, Äikäs L, Borrelli M, Hermansson M, Assini JM, Kemppainen A, Ruhanen H, Ruuth M, Matikainen S, Kovanen PT, Käkelä R, Boffa MB, Koschinsky ML, Öörni K. Lipoprotein(a) induces caspase-1 activation and IL-1 signaling in human macrophages. Front Cardiovasc Med 2023; 10:1130162. [PMID: 37293282 PMCID: PMC10244518 DOI: 10.3389/fcvm.2023.1130162] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 05/02/2023] [Indexed: 06/10/2023] Open
Abstract
Introduction Lipoprotein(a) (Lp(a)) is an LDL-like particle with an additional apolipoprotein (apo)(a) covalently attached. Elevated levels of circulating Lp(a) are a risk factor for atherosclerosis. A proinflammatory role for Lp(a) has been proposed, but its molecular details are incompletely defined. Methods and results To explore the effect of Lp(a) on human macrophages we performed RNA sequencing on THP-1 macrophages treated with Lp(a) or recombinant apo(a), which showed that especially Lp(a) induces potent inflammatory responses. Thus, we stimulated THP-1 macrophages with serum containing various Lp(a) levels to investigate their correlations with cytokines highlighted by the RNAseq, showing significant correlations with caspase-1 activity and secretion of IL-1β and IL-18. We further isolated both Lp(a) and LDL particles from three donors and then compared their atheroinflammatory potentials together with recombinant apo(a) in primary and THP-1 derived macrophages. Compared with LDL, Lp(a) induced a robust and dose-dependent caspase-1 activation and release of IL-1β and IL-18 in both macrophage types. Recombinant apo(a) strongly induced caspase-1 activation and IL-1β release in THP-1 macrophages but yielded weak responses in primary macrophages. Structural analysis of these particles revealed that the Lp(a) proteome was enriched in proteins associated with complement activation and coagulation, and its lipidome was relatively deficient in polyunsaturated fatty acids and had a high n-6/n-3 ratio promoting inflammation. Discussion Our data show that Lp(a) particles induce the expression of inflammatory genes, and Lp(a) and to a lesser extent apo(a) induce caspase-1 activation and IL-1 signaling. Major differences in the molecular profiles between Lp(a) and LDL contribute to Lp(a) being more atheroinflammatory.
Collapse
Affiliation(s)
- Martina B. Lorey
- Atherosclerosis Research Laboratory, Wihuri Research Institute, Helsinki, Finland
- Molecular and Integrative Biosciences, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Amer Youssef
- Robarts Research Institute, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON, Canada
| | - Lauri Äikäs
- Atherosclerosis Research Laboratory, Wihuri Research Institute, Helsinki, Finland
| | - Matthew Borrelli
- Department of Physiology & Pharmacology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON, Canada
| | - Martin Hermansson
- Atherosclerosis Research Laboratory, Wihuri Research Institute, Helsinki, Finland
| | - Julia M. Assini
- Robarts Research Institute, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON, Canada
- Department of Biochemistry, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON, Canada
| | - Aapeli Kemppainen
- Atherosclerosis Research Laboratory, Wihuri Research Institute, Helsinki, Finland
| | - Hanna Ruhanen
- Molecular and Integrative Biosciences, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
- Helsinki University Lipidomics Unit (HiLIPID), Helsinki Institute of Life Science (HiLIFE) and Biocenter Finland, Helsinki, Finland
| | - Maija Ruuth
- Atherosclerosis Research Laboratory, Wihuri Research Institute, Helsinki, Finland
| | - Sampsa Matikainen
- Helsinki Rheumatic Disease and Inflammation Research Group, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Petri T. Kovanen
- Atherosclerosis Research Laboratory, Wihuri Research Institute, Helsinki, Finland
| | - Reijo Käkelä
- Molecular and Integrative Biosciences, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
- Helsinki University Lipidomics Unit (HiLIPID), Helsinki Institute of Life Science (HiLIFE) and Biocenter Finland, Helsinki, Finland
| | - Michael B. Boffa
- Robarts Research Institute, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON, Canada
- Department of Biochemistry, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON, Canada
| | - Marlys L. Koschinsky
- Robarts Research Institute, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON, Canada
- Department of Physiology & Pharmacology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON, Canada
| | - Katariina Öörni
- Atherosclerosis Research Laboratory, Wihuri Research Institute, Helsinki, Finland
- Molecular and Integrative Biosciences, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| |
Collapse
|
4
|
Benitez S, Puig N, Rives J, Solé A, Sánchez-Quesada JL. Can Electronegative LDL Act as a Multienzymatic Complex? Int J Mol Sci 2023; 24:ijms24087074. [PMID: 37108253 PMCID: PMC10138509 DOI: 10.3390/ijms24087074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/06/2023] [Accepted: 04/10/2023] [Indexed: 04/29/2023] Open
Abstract
Electronegative LDL (LDL(-)) is a minor form of LDL present in blood for which proportions are increased in pathologies with increased cardiovascular risk. In vitro studies have shown that LDL(-) presents pro-atherogenic properties, including a high susceptibility to aggregation, the ability to induce inflammation and apoptosis, and increased binding to arterial proteoglycans; however, it also shows some anti-atherogenic properties, which suggest a role in controlling the atherosclerotic process. One of the distinctive features of LDL(-) is that it has enzymatic activities with the ability to degrade different lipids. For example, LDL(-) transports platelet-activating factor acetylhydrolase (PAF-AH), which degrades oxidized phospholipids. In addition, two other enzymatic activities are exhibited by LDL(-). The first is type C phospholipase activity, which degrades both lysophosphatidylcholine (LysoPLC-like activity) and sphingomyelin (SMase-like activity). The second is ceramidase activity (CDase-like). Based on the complementarity of the products and substrates of these different activities, this review speculates on the possibility that LDL(-) may act as a sort of multienzymatic complex in which these enzymatic activities exert a concerted action. We hypothesize that LysoPLC/SMase and CDase activities could be generated by conformational changes in apoB-100 and that both activities occur in proximity to PAF-AH, making it feasible to discern a coordinated action among them.
Collapse
Affiliation(s)
- Sonia Benitez
- Cardiovascular Biochemistry Group, Research Institute of the Hospital de la Santa Creu i Sant Pau (IIB Sant Pau), 08041 Barcelona, Spain
- CIBER of Diabetes and Related Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Núria Puig
- Cardiovascular Biochemistry Group, Research Institute of the Hospital de la Santa Creu i Sant Pau (IIB Sant Pau), 08041 Barcelona, Spain
- Biochemistry and Molecular Biology Department, Universitat Autònoma de Barcelona, 08193 Cerdanyola, Spain
| | - José Rives
- Cardiovascular Biochemistry Group, Research Institute of the Hospital de la Santa Creu i Sant Pau (IIB Sant Pau), 08041 Barcelona, Spain
- Biochemistry and Molecular Biology Department, Universitat Autònoma de Barcelona, 08193 Cerdanyola, Spain
| | - Arnau Solé
- Cardiovascular Biochemistry Group, Research Institute of the Hospital de la Santa Creu i Sant Pau (IIB Sant Pau), 08041 Barcelona, Spain
- Biochemistry and Molecular Biology Department, Universitat Autònoma de Barcelona, 08193 Cerdanyola, Spain
| | - José Luis Sánchez-Quesada
- Cardiovascular Biochemistry Group, Research Institute of the Hospital de la Santa Creu i Sant Pau (IIB Sant Pau), 08041 Barcelona, Spain
- CIBER of Diabetes and Related Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
5
|
Hong P, Wang Q, Chen G. Cholesterol induces inflammation and reduces glucose utilization. Open Med (Wars) 2023; 18:20230701. [PMID: 37197354 PMCID: PMC10183724 DOI: 10.1515/med-2023-0701] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 03/29/2023] [Accepted: 04/03/2023] [Indexed: 05/19/2023] Open
Abstract
Cholesterol stimulates inflammation and affects the normal function of islet tissues. However, the precise mechanism underlying the effects of cholesterol on islet cells requires clarification. In this study, we explored the role of cholesterol in glucose utilization in pancreatic cells. Beta-TC-6 cells and mice were treated with cholesterol. We used glucose detection kits to identify the glucose content in the cell culture supernatant and mouse serum and an enzyme-linked immunosorbent assay was used to detect insulin levels in the serum. Glucose-6-phosphatase catalytic subunit 2 (G6PC2), 78 kDa glucose-regulated protein (GRP78), 94 kDa glucose-regulated protein (GRP94), nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3), caspase-1 (casp1), and interleukin-1β (IL-1β) expression levels were detected using immunofluorescence, immunohistochemistry, western blotting, and reverse transcription-quantitative polymerase chain reaction. Hematoxylin-eosin staining was used to detect the histological alterations in pancreatic tissues. Cholesterol decreased beta-TC-6 cell glucose utilization; enhanced pancreatic tissue pathological alterations; increased glucose and insulin levels in mouse serum; increased G6PC2, GRP78, GRP94, and NLRP3 expression levels; and elevated casp1 and pro-IL-1β cleavage. Cholesterol can attenuate glucose utilization efficiency in beta-TC-6 cells and mice, which may be related to endoplasmic reticulum stress and inflammation.
Collapse
Affiliation(s)
- Pingping Hong
- Department of Endocrinology, Shaoxing Central Hospital, Shaoxing312000, Zhejiang, P.R. China
| | - Qing Wang
- Department of Clinical Laboratory Centre, Shaoxing People’s Hospital, Shaoxing312000, Zhejiang, P.R. China
| | - Guoping Chen
- Department of Endocrinology, Deqing People’s Hospital, No. 120 Yingxi South Road, Wukang Town, Deqing County, Huzhou City313200, Zhejiang, P.R. China
| |
Collapse
|
6
|
Wang Y, Jin Z, Sun J, Chen X, Xie P, Zhou Y, Wang S. The role of activated monocyte IFN/SIGLEC1 signalling in Graves' disease. J Endocrinol 2022; 255:1-9. [PMID: 35695299 DOI: 10.1530/joe-21-0453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 06/08/2022] [Indexed: 11/08/2022]
Abstract
Graves' disease (GD) is characterized by dysregulation of the immune system with aberrant immune cell function. However, there have been few previous studies on the role of monocytes in the pathology of GD. The object of this study was to investigate whether and how monocytes participate in GD pathology. CD14+ monocytes were isolated from untreated initial GD patients and healthy controls. Then, RNA-seq was performed to investigate changes in global mRNA expression in monocytes and found that type I interferon (IFN) signalling was among the top upregulated signalling pathways in GD monocytes. Type I IFN-induced sialic acid-binding immunoglobulin-like lectin1 (SIGLEC1) expression was significantly upregulated in untreated GD patients and correlated with thyroid parameters. Patient serum SIGLEC1 concentrations were reduced after anti-thyroid drug treatment. Inhibiting SIGLEC1 expression could inhibit proinflammatory cytokine (IL-1β, IL-6, IL-8, IL-10 and M-CSF) expression in monocytes. In conclusion, our study suggested that type I IFN-mediated monocyte activation could have a deleterious effect on the pathogenesis of GD. These observations indicated that the inhibition of type I IFN-activated monocytes/macrophages could have a therapeutic effect on GD remission.
Collapse
Affiliation(s)
- Yanqiu Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhou Jin
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiajun Sun
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinxin Chen
- Department of Endocrine and Metabolic Diseases, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Shanghai, China
| | - Pu Xie
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yulin Zhou
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shu Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
7
|
Lorey MB, Öörni K, Kovanen PT. Modified Lipoproteins Induce Arterial Wall Inflammation During Atherogenesis. Front Cardiovasc Med 2022; 9:841545. [PMID: 35310965 PMCID: PMC8927694 DOI: 10.3389/fcvm.2022.841545] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 01/26/2022] [Indexed: 12/15/2022] Open
Abstract
Circulating apolipoprotein B-containing lipoproteins, notably the low-density lipoproteins, enter the inner layer of the arterial wall, the intima, where a fraction of them is retained and modified by proteases, lipases, and oxidizing agents and enzymes. The modified lipoproteins and various modification products, such as fatty acids, ceramides, lysophospholipids, and oxidized lipids induce inflammatory reactions in the macrophages and the covering endothelial cells, initiating an increased leukocyte diapedesis. Lipolysis of the lipoproteins also induces the formation of cholesterol crystals with strong proinflammatory properties. Modified and aggregated lipoproteins, cholesterol crystals, and lipoproteins isolated from human atherosclerotic lesions, all can activate macrophages and thereby induce the secretion of proinflammatory cytokines, chemokines, and enzymes. The extent of lipoprotein retention, modification, and aggregation have been shown to depend largely on differences in the composition of the circulating lipoprotein particles. These properties can be modified by pharmacological means, and thereby provide opportunities for clinical interventions regarding the prevention and treatment of atherosclerotic vascular diseases.
Collapse
Affiliation(s)
- Martina B. Lorey
- Atherosclerosis Research Laboratory, Wihuri Research Institute, Helsinki, Finland
- Molecular and Integrative Biosciences, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Katariina Öörni
- Atherosclerosis Research Laboratory, Wihuri Research Institute, Helsinki, Finland
- Molecular and Integrative Biosciences, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
- *Correspondence: Katariina Öörni
| | - Petri T. Kovanen
- Atherosclerosis Research Laboratory, Wihuri Research Institute, Helsinki, Finland
| |
Collapse
|
8
|
Chang PY, Chang SF, Chang TY, Su HM, Lu SC. Synergistic effects of electronegative-LDL- and palmitic-acid-triggered IL-1β production in macrophages via LOX-1- and voltage-gated-potassium-channel-dependent pathways. J Nutr Biochem 2021; 97:108767. [PMID: 34052311 DOI: 10.1016/j.jnutbio.2021.108767] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 03/16/2021] [Accepted: 04/16/2021] [Indexed: 12/22/2022]
Abstract
Electronegative LDL (LDL(-)) and free fatty acids (FFAs) are circulating risk factors for cardiovascular diseases (CVDs) and have been associated with inflammation. Interleukin-1 beta (IL-1β) represents a key cytokine in the development of CVD; however, the initial trigger of IL-1β in CVD remains to be explored. In this study, we investigated the combined effects of LDL(-) from the plasma of ST-segment elevation myocardial infarction (STEMI) patients or diet-induced hypercholesterolemic rabbits and bovine serum albumin bound palmitic acid (PA-BSA) on IL-1β production in macrophages. Macrophages derived from THP-1 cells or human peripheral blood mononuclear cells were independently treated with LDL(-), PA-BSA or cotreated with LDL(-) and PA-BSA. The results showed that nLDL and/or PA-BSA had no effect on IL-1β, and LDL(-) slightly increased IL-1β; however, cotreatment with LDL(-) and PA-BSA resulted in abundant secretion of IL-1β in macrophages. Rabbit LDL(-) induced the elevation of cellular pro-IL-1β and p-Iκ-Bα, but PA-BSA had no effect on pro-IL-1β or p-Iκ-Bα. In potassium-free buffer, LDL(-)-induced IL-1β reached a level similar to that induced by cotreatment with LDL(-) and PA-BSA. Moreover, LDL(-) and PA-BSA-induced IL-1β was inhibited in lectin-type oxidized LDL receptor-1 (LOX-1) knockdown cells and by blockers of voltage-gated potassium (Kv) channels. LDL(-) from diet-induced hypercholesterolemic rabbit had a similar effect as STEMI LDL(-) on IL-1β in macrophages. These results show that PA-BSA cooperates with LDL(-) to trigger IL-1β production in macrophages via a mechanism involving the LOX-1 and Kv channel pathways, which may play crucial roles in the regulation of inflammation in CVD.
Collapse
Affiliation(s)
- Po-Yuan Chang
- Cardiovascular Center and Division of Cardiology, Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Shwu-Fen Chang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Ting-Yu Chang
- Department of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hui-Min Su
- Department of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Shao-Chun Lu
- Department of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
9
|
The selective NLRP3 inhibitor MCC950 hinders atherosclerosis development by attenuating inflammation and pyroptosis in macrophages. Sci Rep 2021; 11:19305. [PMID: 34588488 PMCID: PMC8481539 DOI: 10.1038/s41598-021-98437-3] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Accepted: 08/30/2021] [Indexed: 11/16/2022] Open
Abstract
NLRP3 inflammasome is a vital player in macrophages pyroptosis, which is a type of proinflammatory cell-death and takes part in the pathogenesis of atherosclerosis. In this study, we used apoE−/− mice and ox-LDL induced THP-1 derived macrophages to explore the mechanisms of MCC950, a selective NLRP3 inhibitor in treating atherosclerosis. For the in vivo study, MCC950 was intraperitoneal injected to 8-week-old apoE−/− mice fed with high-fat diet for 12 weeks. For the in vitro study, THP-1 derived macrophages were treated with ox-LDL and MCC950 for 48 h. MCC950 administration reduced plaque areas and macrophages contents, but did not improve the serum lipid profiles in aortic root of apoE−/− mice. MCC950 inhibited the activation of NLRP3/ASC/Caspase-1/GSDMD-N axis, and alleviated macrophages pyroptosis and the production of IL-1β and IL-18 both in aorta and in cell lysates. However, MCC950 did not affect the expression of TLR4 or the mRNA levels of NLRP3 inflammasome and its downstream proteins, suggesting that MCC950 had no effects on the priming of NLRP3 inflammasome activation in macrophages. The anti-atherosclerotic mechanisms of MCC950 on attenuating macrophages inflammation and pyroptosis involved in inhibiting the assembly and activation of NLRP3 inflammasome, rather than interrupting its priming.
Collapse
|
10
|
Sun C, Zheng W, Liang L, Liu Z, Sun W, Tang R. Ezetimibe Improves Rosuvastatin Effects on Inflammation and Vascular Endothelial Function in Acute Coronary Syndrome Patients Undergoing PCI. J Interv Cardiol 2021; 2021:2995602. [PMID: 34566523 PMCID: PMC8443370 DOI: 10.1155/2021/2995602] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 07/26/2021] [Accepted: 08/18/2021] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Little is known of the acute effects of ezetimibe in patients with acute coronary syndrome (ACS) undergoing PCI. We investigated whether ezetimibe improves inflammation and vascular endothelial function in patients with ACS undergoing PCI. METHODS We randomized 171 patients with ACS undergoing PCI to receive ezetimibe 10 mg/day plus rosuvastatin 20 mg/day (combination group, n = 81) versus rosuvastatin 20 mg/day (rosuvastatin group, n = 90). Lipid profile, type II secretory phospholipase A2 (sPLA2-IIa), interleukin-1β (IL-1β), vascular cell adhesion molecule-1 (VCAM-1), and intercellular cell adhesion molecule-1 (ICAM-1) were measured at baseline and after 7 days. Three months after PCI, clinical outcomes were examined. RESULT The levels of sPLA2-IIa and IL-1β reduced significantly in both groups, but more when ezetimibe and rosuvastatin were coadministered (sPLA2-IIa: 6.16 ± 2.67 vs. 7.42 ± 3.53 ng/ml, p=0.01; IL-1β: 37.39 ± 26.25 vs. 48.98 ± 32.26 pg/ml, p=0.01). A significant rise of VCAM-1 and ICAM-1 was observed on day 7 after PCI in the both groups, but was less in the combination group (VCAM-1: 918.28 ± 235.31 vs. 988.54 ± 194.41 ng/ml, p=0.03; ICAM-1: 213.01 ± 100.15 vs. 246.88 ± 105.71 ng/ml, p=0.03). Patients in the combination versus rosuvastatin group appeared to suffer from less major adverse events. Periprocedural therapy of ezetimibe improves rosuvastatin effects on proinflammatory responses and endothelial function associated with ACS patients undergoing PCI. This trial is registered with https://clinicaltrials.gov/ct2/show/ChiCTR-IPR-17012219 (Chinese Clinical Trial Registry, http://www.chictr.org.cn on 02/08/2017).
Collapse
Affiliation(s)
- Changqing Sun
- Department of Cardiology, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China
| | - Wuyang Zheng
- Department of Cardiology, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China
| | - Ling Liang
- Department of Cardiology, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China
| | - Zuheng Liu
- Department of Cardiology, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China
| | - Wenchao Sun
- Department of Cardiology, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China
| | - Rong Tang
- Department of Cardiology, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China
| |
Collapse
|
11
|
Structure and Dynamics of Oxidized Lipoproteins In Vivo: Roles of High-Density Lipoprotein. Biomedicines 2021; 9:biomedicines9060655. [PMID: 34201176 PMCID: PMC8229488 DOI: 10.3390/biomedicines9060655] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 06/03/2021] [Accepted: 06/04/2021] [Indexed: 01/30/2023] Open
Abstract
Oxidative modification of lipoproteins is implicated in the occurrence and development of atherosclerotic lesions. Earlier studies have elucidated on the mechanisms of foam cell formation and lipid accumulation in these lesions, which is mediated by scavenger receptor-mediated endocytosis of oxidized low-density lipoprotein (oxLDL). Mounting clinical evidence has supported the involvement of oxLDL in cardiovascular diseases. High-density lipoprotein (HDL) is known as anti-atherogenic; however, recent studies have shown circulating oxidized HDL (oxHDL) is related to cardiovascular diseases. A modified structure of oxLDL, which was increased in the plasma of patients with acute myocardial infarction, was characterized. It had two unique features: (1) a fraction of oxLDL accompanied oxHDL, and (2) apoA1 was heavily modified, while modification of apoB, and the accumulation of oxidized phosphatidylcholine (oxPC) and lysophosphatidylcholine (lysoPC) was less pronounced. When LDL and HDL were present at the same time, oxidized lipoproteins actively interacted with each other, and oxPC and lysoPC were transferred to another lipoprotein particle and enzymatically metabolized rapidly. This brief review provides a novel view on the dynamics of oxLDL and oxHDL in circulation.
Collapse
|
12
|
Cavalcante MF, Adorne MD, Turato WM, Kemmerer M, Uchiyama MK, Asbahr ACC, Alves ADCS, Farsky SHP, Drewes C, Spatti MC, Kazuma SM, Boss M, Guterres SS, Araki K, Brüne B, Namgaladze D, Pohlmann AR, Abdalla DSP. scFv-Anti-LDL(-)-Metal-Complex Multi-Wall Functionalized-Nanocapsules as a Promising Tool for the Prevention of Atherosclerosis Progression. Front Med (Lausanne) 2021; 8:652137. [PMID: 33959626 PMCID: PMC8095373 DOI: 10.3389/fmed.2021.652137] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 03/17/2021] [Indexed: 11/13/2022] Open
Abstract
Atherosclerosis can be originated from the accumulation of modified cholesterol-rich lipoproteins in the arterial wall. The electronegative LDL, LDL(-), plays an important role in the pathogenesis of atherosclerosis once this cholesterol-rich lipoprotein can be internalized by macrophages, contributing to the formation of foam cells, and provoking an immune-inflammatory response. Herein, we engineered a nanoformulation containing highly pure surface-functionalized nanocapsules using a single-chain fragment variable (scFv) reactive to LDL(-) as a ligand and assessed whether it can affect the LDL(-) uptake by primary macrophages and the progression of atherosclerotic lesions in Ldlr -/- mice. The engineered and optimized scFv-anti-LDL(-)-MCMN-Zn nanoformulation is internalized by human and murine macrophages in vitro by different endocytosis mechanisms. Moreover, macrophages exhibited lower LDL(-) uptake and reduced mRNA and protein levels of IL1B and MCP1 induced by LDL(-) when treated with this new nanoformulation. In a mouse model of atherosclerosis employing Ldlr -/- mice, intravenous administration of scFv-anti-LDL(-)-MCMN-Zn nanoformulation inhibited atherosclerosis progression without affecting vascular permeability or inducing leukocytes-endothelium interactions. Together, these findings suggest that a scFv-anti-LDL(-)-MCMN-Zn nanoformulation holds promise to be used in future preventive and therapeutic strategies for atherosclerosis.
Collapse
Affiliation(s)
- Marcela Frota Cavalcante
- Department of Clinical and Toxicological Analysis, Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Márcia Duarte Adorne
- Department of Organic Chemistry, Chemistry Institute, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Walter Miguel Turato
- Department of Clinical and Toxicological Analysis, Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Marina Kemmerer
- Faculty of Medicine, Institute of Biochemistry I, Goethe-University Frankfurt, Frankfurt, Germany
| | - Mayara Klimuk Uchiyama
- Department of Fundamental Chemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Ana Carolina Cavazzin Asbahr
- Department of Organic Chemistry, Chemistry Institute, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Aline de Cristo Soares Alves
- Department of Production and Control of Medicines, Faculty of Pharmacy, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Sandra Helena Poliselli Farsky
- Department of Clinical and Toxicological Analysis, Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Carine Drewes
- Department of Clinical and Toxicological Analysis, Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Marina Cecília Spatti
- Department of Clinical and Toxicological Analysis, Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Soraya Megumi Kazuma
- Department of Clinical and Toxicological Analysis, Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Marcel Boss
- Faculty of Medicine, Institute of Biochemistry I, Goethe-University Frankfurt, Frankfurt, Germany
| | - Silvia Stanisçuaski Guterres
- Department of Production and Control of Medicines, Faculty of Pharmacy, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Koiti Araki
- Department of Fundamental Chemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Bernhard Brüne
- Faculty of Medicine, Institute of Biochemistry I, Goethe-University Frankfurt, Frankfurt, Germany
| | - Dmitry Namgaladze
- Faculty of Medicine, Institute of Biochemistry I, Goethe-University Frankfurt, Frankfurt, Germany
| | - Adriana Raffin Pohlmann
- Department of Organic Chemistry, Chemistry Institute, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Dulcineia Saes Parra Abdalla
- Department of Clinical and Toxicological Analysis, Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
13
|
Ke LY, Law SH, Mishra VK, Parveen F, Chan HC, Lu YH, Chu CS. Molecular and Cellular Mechanisms of Electronegative Lipoproteins in Cardiovascular Diseases. Biomedicines 2020; 8:biomedicines8120550. [PMID: 33260304 PMCID: PMC7760527 DOI: 10.3390/biomedicines8120550] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/23/2020] [Accepted: 11/26/2020] [Indexed: 02/06/2023] Open
Abstract
Dysregulation of glucose and lipid metabolism increases plasma levels of lipoproteins and triglycerides, resulting in vascular endothelial damage. Remarkably, the oxidation of lipid and lipoprotein particles generates electronegative lipoproteins that mediate cellular deterioration of atherosclerosis. In this review, we examined the core of atherosclerotic plaque, which is enriched by byproducts of lipid metabolism and lipoproteins, such as oxidized low-density lipoproteins (oxLDL) and electronegative subfraction of LDL (LDL(−)). We also summarized the chemical properties, receptors, and molecular mechanisms of LDL(−). In combination with other well-known markers of inflammation, namely metabolic diseases, we concluded that LDL(−) can be used as a novel prognostic tool for these lipid disorders. In addition, through understanding the underlying pathophysiological molecular routes for endothelial dysfunction and inflammation, we may reassess current therapeutics and might gain a new direction to treat atherosclerotic cardiovascular diseases, mainly targeting LDL(−) clearance.
Collapse
Affiliation(s)
- Liang-Yin Ke
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807378, Taiwan; (L.-Y.K.); (S.H.L.); (V.K.M.); (F.P.)
- Graduate Institute of Medicine, College of Medicine and Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
- Center for Lipid Biosciences, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807377, Taiwan; (H.-C.C.); (Y.-H.L.)
| | - Shi Hui Law
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807378, Taiwan; (L.-Y.K.); (S.H.L.); (V.K.M.); (F.P.)
| | - Vineet Kumar Mishra
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807378, Taiwan; (L.-Y.K.); (S.H.L.); (V.K.M.); (F.P.)
| | - Farzana Parveen
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807378, Taiwan; (L.-Y.K.); (S.H.L.); (V.K.M.); (F.P.)
| | - Hua-Chen Chan
- Center for Lipid Biosciences, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807377, Taiwan; (H.-C.C.); (Y.-H.L.)
| | - Ye-Hsu Lu
- Center for Lipid Biosciences, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807377, Taiwan; (H.-C.C.); (Y.-H.L.)
- Division of Cardiology, Department of International Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807377, Taiwan
| | - Chih-Sheng Chu
- Center for Lipid Biosciences, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807377, Taiwan; (H.-C.C.); (Y.-H.L.)
- Division of Cardiology, Department of International Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807377, Taiwan
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 80145, Taiwan
- Correspondence: ; Tel.: +886-73121101 (ext. 2297); Fax: +886-73111996
| |
Collapse
|
14
|
Chu CS, Law SH, Lenzen D, Tan YH, Weng SF, Ito E, Wu JC, Chen CH, Chan HC, Ke LY. Clinical Significance of Electronegative Low-Density Lipoprotein Cholesterol in Atherothrombosis. Biomedicines 2020; 8:biomedicines8080254. [PMID: 32751498 PMCID: PMC7460408 DOI: 10.3390/biomedicines8080254] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/25/2020] [Accepted: 07/28/2020] [Indexed: 02/06/2023] Open
Abstract
Despite the numerous risk factors for atherosclerotic cardiovascular diseases (ASCVD), cumulative evidence shows that electronegative low-density lipoprotein (L5 LDL) cholesterol is a promising biomarker. Its toxicity may contribute to atherothrombotic events. Notably, plasma L5 LDL levels positively correlate with the increasing severity of cardiovascular diseases. In contrast, traditional markers such as LDL-cholesterol and triglyceride are the therapeutic goals in secondary prevention for ASCVD, but that is controversial in primary prevention for patients with low risk. In this review, we point out the clinical significance and pathophysiological mechanisms of L5 LDL, and the clinical applications of L5 LDL levels in ASCVD can be confidently addressed. Based on the previously defined cut-off value by receiver operating characteristic curve, the acceptable physiological range of L5 concentration is proposed to be below 1.7 mg/dL. When L5 LDL level surpass this threshold, clinically relevant ASCVD might be present, and further exams such as carotid intima-media thickness, pulse wave velocity, exercise stress test, or multidetector computed tomography are required. Notably, the ultimate goal of L5 LDL concentration is lower than 1.7 mg/dL. Instead, with L5 LDL greater than 1.7 mg/dL, lipid-lowering treatment may be required, including statin, ezetimibe or PCSK9 inhibitor, regardless of the low-density lipoprotein cholesterol (LDL-C) level. Since L5 LDL could be a promising biomarker, we propose that a high throughput, clinically feasible methodology is urgently required not only for conducting a prospective, large population study but for developing therapeutics strategies to decrease L5 LDL in the blood.
Collapse
Affiliation(s)
- Chih-Sheng Chu
- Center for Lipid Biosciences, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807377, Taiwan;
- Division of Cardiology, Department of International Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807377, Taiwan
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 80145, Taiwan
| | - Shi Hui Law
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807378, Taiwan; (S.H.L.); (D.L.); (Y.-H.T.); (E.I.)
| | - David Lenzen
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807378, Taiwan; (S.H.L.); (D.L.); (Y.-H.T.); (E.I.)
| | - Yong-Hong Tan
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807378, Taiwan; (S.H.L.); (D.L.); (Y.-H.T.); (E.I.)
| | - Shih-Feng Weng
- Department of Healthcare Administration and Medical Informatics, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807378, Taiwan;
| | - Etsuro Ito
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807378, Taiwan; (S.H.L.); (D.L.); (Y.-H.T.); (E.I.)
- Department of Biology, Waseda University, Tokyo 162-8480, Japan
- Waseda Research Institute for Science and Engineering, Waseda University, Tokyo 162-8480, Japan
| | - Jung-Chou Wu
- Division of Cardiology, Department of Internal Medicine, Pingtung Christian Hospital, Pingtung 90059, Taiwan;
| | - Chu-Huang Chen
- Vascular and Medicinal Research, Texas Heart Institute, Houston, TX 77030, USA;
| | - Hua-Chen Chan
- Center for Lipid Biosciences, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807377, Taiwan;
- Correspondence: (H.-C.C.); (L.-Y.K.); Tel.: +886-73121101 (ext. 2296); Fax: +886-73111996 (L.-Y.K.)
| | - Liang-Yin Ke
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807378, Taiwan; (S.H.L.); (D.L.); (Y.-H.T.); (E.I.)
- Graduate Institute of Medicine, College of Medicine, & Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
- Correspondence: (H.-C.C.); (L.-Y.K.); Tel.: +886-73121101 (ext. 2296); Fax: +886-73111996 (L.-Y.K.)
| |
Collapse
|
15
|
Akyol O, Chowdhury I, Akyol HR, Tessier K, Vural H, Akyol S. Why are cardiovascular diseases more common among patients with severe mental illness? The potential involvement of electronegative low-density lipoprotein (LDL) L5. Med Hypotheses 2020; 142:109821. [PMID: 32417641 DOI: 10.1016/j.mehy.2020.109821] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 04/22/2020] [Accepted: 05/05/2020] [Indexed: 02/07/2023]
Abstract
Despite tremendous efforts of experimental and clinical studies and knowledge, the pathophysiology of severe mental illness (SMI), including bipolar disorder (BD), unipolar depression (mood disorders, MD), and schizophrenia (SCZ), remains poorly understood. Besides their chronic course and high prevalence in society, mental and somatic comorbidities are really serious problems; patients with these disorders have increased risk of cardiovascular (CV) diseases (CVD) including coronary artery diseases (CAD, i.e. myocardial infarction and angina), stroke, sudden cardiac death, hypertension, cardiomyopathy, arrhythmia, and thromboembolic disease. Although it is determined that triglycerides, cholesterol, glucose, and low-density lipoprotein (LDL) levels are increased in MD and SCZ, the underlying reason remains unknown. Considering this, we propose that electronegative LDL (L5) is probably the main crucial element to understanding CVD induced by SMI and to discovering novel remedial approaches for these diseases. When it is hypothesized that L5 is greatly presupposed in CV system abnormalities, it follows that the anti-L5 therapies and even antioxidant treatment options may open new therapeutic opportunities to prevent CVD diseases secondary to SMI. In this review article, we tried to bring a very original subject to the attention of readers who are interested in lipoprotein metabolism in terms of experimental, clinical, and cell culture studies that corroborate the involvement of L5 in physiopathology of CVD secondary to SMI and also the new therapeutic approaches for these disorders.
Collapse
Affiliation(s)
- Omer Akyol
- Michigan Math & Science Academy, Department of Science, Warren, MI, USA.
| | - Imtihan Chowdhury
- Michigan Math & Science Academy, High School, 11th grade, Warren, MI, USA
| | - Hafsa Rana Akyol
- Illinois Institute of Technology, Biology, Sophomore, Chicago, IL, USA
| | - Kylie Tessier
- Michigan Math & Science Academy, High School, 11th grade, Warren, MI, USA
| | - Huseyin Vural
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, RWTH University Hospital Aachen, Aachen, Germany
| | - Sumeyya Akyol
- Beaumont Health, Beaumont Research Institute, Royal Oak, MI, USA
| |
Collapse
|
16
|
Chen C, Ke L, Chan H, Chu C, Lee A, Lin K, Lee M, Hsiao P, Chen C, Shin S. Electronegative low-density lipoprotein of patients with metabolic syndrome induces pathogenesis of aorta through disruption of the stimulated by retinoic acid 6 cascade. J Diabetes Investig 2020; 11:535-544. [PMID: 31597015 PMCID: PMC7232312 DOI: 10.1111/jdi.13158] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 10/01/2019] [Accepted: 10/06/2019] [Indexed: 12/26/2022] Open
Abstract
AIMS/INTRODUCTION Electronegative low-density lipoprotein (L5) is the most atherogenic fraction of low-density lipoprotein and is elevated in people with metabolic syndrome (MetS), whereas the retinol-binding protein 4 receptor (stimulated by retinoic acid 6 [STRA6]) cascade is disrupted in various organs of patients with obesity-related diseases. Our objective was to investigate whether L5 from MetS patients capably induces pathogenesis of aorta through disrupting the STRA6 cascade. MATERIAL AND METHODS We examined the in vivo and in vitro effects of L5 on the STRA6 cascade and aortic atherogenic markers. To investigate the role of this cascade on atherosclerotic formation, crbp1 transfection was carried out in vitro. RESULTS This study shows that L5 activates atherogenic markers (p38 mitogen-activated protein kinases, pSmad2 and matrix metallopeptidase 9) and simultaneously suppresses STRA6 signals (STRA6, cellular retinol-binding protein 1, lecithin-retinol acyltransferase, retinoic acid receptor-α and retinoid X receptor-α) in aortas of L5-injected mice and L5-treated human aortic endothelial cell lines and human aortic smooth muscle cell lines. These L5-induced changes of the STRA6 cascade and atherogenic markers were reversed in aortas of LOX1-/- mice and in LOX1 ribonucleic acid-silenced human aortic endothelial cell lines and human aortic smooth muscle cell lines. Furthermore, crbp1 gene transfection reversed the disruption of the STRA6 cascade, the phosphorylation of p38 mitogen-activated protein kinases and Smad2, and the elevation of matrix metallopeptidase 9 in L5-treated human aortic endothelial cell lines. CONCLUSIONS This study shows that L5 from MetS patients induces atherogenic markers by disrupting STRA6 signaling. Suppression of STRA6 might be one novel pathogenesis of aorta in patients with MetS.
Collapse
Affiliation(s)
- Chao‐Hung Chen
- School of MedicineCollege of MedicineKaohsiung Medical UniversityKaohsiungTaiwan
- Divison of Endocrinology and MetabolismDepartment of Internal MedicineKaohsiung Medical University HospitalKaohsiungTaiwan
| | - Liang‐Yin Ke
- Lipid Science and Aging Research CenterKaohsiung Medical UniversityKaohsiungTaiwan
- Department of Medical Laboratory Science and BiotechnologyCollege of Health SciencesKaohsiung Medical UniversityKaohsiungTaiwan
| | - Hua‐Chen Chan
- Department of Medical Laboratory Science and BiotechnologyCollege of Health SciencesKaohsiung Medical UniversityKaohsiungTaiwan
| | - Chih‐Sheng Chu
- Division of CardiologyDepartment of Internal MedicineKaohsiung Medical University HospitalKaohsiungTaiwan
| | - An‐Sheng Lee
- Department of MedicineMackay Medical CollegeNew TaipeiTaiwan
| | - Kun‐Der Lin
- School of MedicineCollege of MedicineKaohsiung Medical UniversityKaohsiungTaiwan
- Divison of Endocrinology and MetabolismDepartment of Internal MedicineKaohsiung Medical University HospitalKaohsiungTaiwan
- Vascular and Medical ResearchTexas Heart InstituteHoustonTexasUSA
| | - Mei‐Yueh Lee
- School of MedicineCollege of MedicineKaohsiung Medical UniversityKaohsiungTaiwan
- Divison of Endocrinology and MetabolismDepartment of Internal MedicineKaohsiung Medical University HospitalKaohsiungTaiwan
| | - Pi‐Jung Hsiao
- School of MedicineCollege of MedicineKaohsiung Medical UniversityKaohsiungTaiwan
- Divison of Endocrinology and MetabolismDepartment of Internal MedicineKaohsiung Medical University HospitalKaohsiungTaiwan
| | - Chu‐Huang Chen
- Lipid Science and Aging Research CenterKaohsiung Medical UniversityKaohsiungTaiwan
- Vascular and Medical ResearchTexas Heart InstituteHoustonTexasUSA
- Department of Internal MedicineKaohsiung Ta‐Tung Municipal HospitalKaohsiung Medical University HospitalKaohsiungTaiwan
| | - Shyi‐Jang Shin
- School of MedicineCollege of MedicineKaohsiung Medical UniversityKaohsiungTaiwan
- Divison of Endocrinology and MetabolismDepartment of Internal MedicineKaohsiung Medical University HospitalKaohsiungTaiwan
- Department of Medical Laboratory Science and BiotechnologyCollege of Health SciencesKaohsiung Medical UniversityKaohsiungTaiwan
| |
Collapse
|
17
|
Bäck M, Yurdagul A, Tabas I, Öörni K, Kovanen PT. Inflammation and its resolution in atherosclerosis: mediators and therapeutic opportunities. Nat Rev Cardiol 2020; 16:389-406. [PMID: 30846875 DOI: 10.1038/s41569-019-0169-2] [Citation(s) in RCA: 635] [Impact Index Per Article: 127.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Atherosclerosis is a lipid-driven inflammatory disease of the arterial intima in which the balance of pro-inflammatory and inflammation-resolving mechanisms dictates the final clinical outcome. Intimal infiltration and modification of plasma-derived lipoproteins and their uptake mainly by macrophages, with ensuing formation of lipid-filled foam cells, initiate atherosclerotic lesion formation, and deficient efferocytotic removal of apoptotic cells and foam cells sustains lesion progression. Defective efferocytosis, as a sign of inadequate inflammation resolution, leads to accumulation of secondarily necrotic macrophages and foam cells and the formation of an advanced lesion with a necrotic lipid core, indicative of plaque vulnerability. Resolution of inflammation is mediated by specialized pro-resolving lipid mediators derived from omega-3 fatty acids or arachidonic acid and by relevant proteins and signalling gaseous molecules. One of the major effects of inflammation resolution mediators is phenotypic conversion of pro-inflammatory macrophages into macrophages that suppress inflammation and promote healing. In advanced atherosclerotic lesions, the ratio between specialized pro-resolving mediators and pro-inflammatory lipids (in particular leukotrienes) is strikingly low, providing a molecular explanation for the defective inflammation resolution features of these lesions. In this Review, we discuss the mechanisms of the formation of clinically dangerous atherosclerotic lesions and the potential of pro-resolving mediator therapy to inhibit this process.
Collapse
Affiliation(s)
- Magnus Bäck
- Department of Cardiology, Karolinska University Hospital, Stockholm, Sweden
| | - Arif Yurdagul
- Columbia University Irving Medical Center, New York, NY, USA
| | - Ira Tabas
- Columbia University Irving Medical Center, New York, NY, USA
| | - Katariina Öörni
- Atherosclerosis Research Laboratory, Wihuri Research Institute, Helsinki, Finland.,Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Petri T Kovanen
- Atherosclerosis Research Laboratory, Wihuri Research Institute, Helsinki, Finland.
| |
Collapse
|
18
|
Corrêa R, Silva LFF, Ribeiro DJS, Almeida RDN, Santos IDO, Corrêa LH, de Sant'Ana LP, Assunção LS, Bozza PT, Magalhães KG. Lysophosphatidylcholine Induces NLRP3 Inflammasome-Mediated Foam Cell Formation and Pyroptosis in Human Monocytes and Endothelial Cells. Front Immunol 2020; 10:2927. [PMID: 31998284 PMCID: PMC6962110 DOI: 10.3389/fimmu.2019.02927] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 11/28/2019] [Indexed: 12/04/2022] Open
Abstract
Foam cells are specialized lipid-loaded macrophages derived from monocytes and are a key pathological feature of atherosclerotic lesions. Lysophosphatidylcholine (LPC) is a major lipid component of the plasma membrane with a broad spectrum of proinflammatory activities and plays a key role in atherosclerosis. However, the role of LPC in lipid droplet (LD) biogenesis and the modulation of inflammasome activation is still poorly understood. In the present study, we investigated whether LPC can induce foam cell formation through an analysis of LD biogenesis and determined whether the cell signaling involved in this process is mediated by the inflammasome activation pathway in human endothelial cells and monocytes. Our results showed that LPC induced foam cell formation in both types of cells by increasing LD biogenesis via a NLRP3 inflammasome-dependent pathway. Furthermore, LPC induced pyroptosis in both cells and the activation of the inflammasome with IL-1β secretion, which was dependent on potassium efflux and lysosomal damage in human monocytes. The present study described the IL-1β secretion and foam cell formation triggered by LPC via an inflammasome-mediated pathway in human monocytes and endothelial cells. Our results will help improve our understanding of the relationships among LPC, LD biogenesis, and NLRP3 inflammasome activation in the pathogenesis of atherosclerosis.
Collapse
Affiliation(s)
- Rafael Corrêa
- Laboratory of Immunology and Inflammation, Department of Cell Biology, University of Brasília, Brasilia, Brazil
| | - Luís Felipe Fonseca Silva
- Laboratory of Immunology and Inflammation, Department of Cell Biology, University of Brasília, Brasilia, Brazil
| | | | - Raquel das Neves Almeida
- Laboratory of Immunology and Inflammation, Department of Cell Biology, University of Brasília, Brasilia, Brazil
| | - Igor de Oliveira Santos
- Laboratory of Immunology and Inflammation, Department of Cell Biology, University of Brasília, Brasilia, Brazil
| | - Luís Henrique Corrêa
- Laboratory of Immunology and Inflammation, Department of Cell Biology, University of Brasília, Brasilia, Brazil
| | - Lívia Pimentel de Sant'Ana
- Laboratory of Immunology and Inflammation, Department of Cell Biology, University of Brasília, Brasilia, Brazil
| | | | - Patrícia T Bozza
- Laboratory of Immunopharmacology, Institute of Oswaldo Cruz-Fiocruz, Rio de Janeiro, Brazil
| | - Kelly Grace Magalhães
- Laboratory of Immunology and Inflammation, Department of Cell Biology, University of Brasília, Brasilia, Brazil
| |
Collapse
|
19
|
Tripodi GL, Prieto MB, Abdalla DSP. Inflammasome Activation in Human Macrophages Induced by a LDL (-) Mimetic Peptide. Inflammation 2019; 43:722-730. [PMID: 31858317 DOI: 10.1007/s10753-019-01159-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The inflammasome is responsible for maturation of interleukin-1β (IL-1β) and interleukin-18 (IL-18) contributing to the inflammatory process in atherosclerosis. It is shown here that an electronegative low-density lipoprotein [LDL (-)] apoB-100 mimetic peptide can activate the transcriptional and posttranslational signs needed for complete inflammasome activation. This peptide, named p2C7, can activate the Toll-like receptor 4 (TLR4) that induces NF-κB activation and the transcription of inflammasome components. After blocking TLR4 with a neutralizing antibody, inflammasome component (NLRP3, CASP1, and ASC) and IL1b and IL18 gene downregulation occurred in human-derived macrophages stimulated with p2C7 or LDL (-). Moreover, the posttranslational signal was activated by the interaction between p2C7 and the lectin-type oxidized LDL receptor 1 (LOX-1), as demonstrated by the induction of caspase-1 cleavage in macrophages. The blockage of either TLR4 or LOX-1 decreased IL-1β and IL-18 secretion by human-derived macrophages as both pathways are necessary for complete inflammasome activation. These findings suggest a mechanism by which macrophages transduce the pro-inflammatory signal provided by LDL (-) ApoB-100 and its mimetic peptides to activate the inflammasome protein complex what may be relevant for the inflammatory process in atherosclerosis.
Collapse
Affiliation(s)
- Gustavo Luis Tripodi
- Department of Clinical and Toxicological Analyses, Faculty of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, 05508-000, Brazil
| | - Marcela Bach Prieto
- Department of Clinical and Toxicological Analyses, Faculty of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, 05508-000, Brazil
| | - Dulcineia Saes Parra Abdalla
- Department of Clinical and Toxicological Analyses, Faculty of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, 05508-000, Brazil.
| |
Collapse
|
20
|
Electronegative LDL from Rabbits Fed with Atherogenic Diet Is Highly Proinflammatory. Mediators Inflamm 2019; 2019:6163130. [PMID: 31534437 PMCID: PMC6724430 DOI: 10.1155/2019/6163130] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 06/23/2019] [Accepted: 07/03/2019] [Indexed: 12/11/2022] Open
Abstract
Electronegative low-density lipoprotein (LDL(-)) has been found in the plasma of familial hypercholesterolemia and acute myocardial infarction and has been implicated in atherosclerosis and cardiovascular disease. However, less is known about the involvement of LDL(-) in atherosclerosis-related inflammation. This study aims at investigating the inducibility of LDL(-) by atherogenic diet in rabbits and at exploring the proinflammatory potential of the diet-induced LDL(-) in macrophages. Rabbits were fed with an atherogenic diet; LDL was isolated from plasma by NaBr density gradient ultracentrifugation and was then resolved into nLDL and LDL(-) by anion-exchange chromatography. Isolated nLDL and LDL(-) were directly used or incubated with 10 μM CuSO4 for 24 h to produce copper- (Cu-) ox-nLDL and Cu-ox-LDL(-). The effects of these LDLs on inflammation were evaluated in THP-1-derived macrophages. Macrophages were treated with nLDL, LDL(-), and extensively oxidized LDL (ox-LDL), then the levels of interleukin- (IL-) 1β, IL-6, and tumor necrosis factor- (TNF-) α in a culture medium were determined by ELISA, and the levels of total and phosphorylated IκB, p65, p38, JNK, and ERK in cell lysates were determined by Western blotting. The LDL(-) induced significantly higher levels of IL-1β, IL-6, and TNF-α in the medium. The levels of phosphorylated/total IκB, p65, p38, JNK, and ERK were also upregulated by LDL(-). In contrast, nLDL, Cu-ox-nLDL, and Cu-ox-LDL(-) exhibited much less effect. Knockdown of lectin-type oxidized LDL receptor- (LOX-) 1 resulted in significant reduction in LDL(-)-induced IL-1β, IL-6, and TNF-α. In addition, these LDL(-) effects were also markedly attenuated by inhibition of NF-κB and ERK1/2. The data suggested that LDL(-) induced inflammation through LOX-1-, NF-κB-, and ERK1/2-dependent pathways. Taken together, our results show that rabbits fed with atherogenic diet produce a highly proinflammatory LDL(-) that is more potent in inducing inflammation than nLDL and extensively oxidize LDL in macrophages. The results thus provide a novel link between diet-induced hypercholesterolemia and inflammation.
Collapse
|
21
|
Puig N, Estruch M, Jin L, Sanchez-Quesada JL, Benitez S. The Role of Distinctive Sphingolipids in the Inflammatory and Apoptotic Effects of Electronegative LDL on Monocytes. Biomolecules 2019; 9:biom9080300. [PMID: 31344975 PMCID: PMC6722802 DOI: 10.3390/biom9080300] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 07/17/2019] [Accepted: 07/20/2019] [Indexed: 01/18/2023] Open
Abstract
Electronegative low-density lipoprotein (LDL(-)) is a minor LDL subfraction that is present in blood with inflammatory and apoptotic effects. We aimed to evaluate the role of sphingolipids ceramide (Cer), sphingosine (Sph), and sphingosine-1-phosphate (S1P) in the LDL(-)-induced effect on monocytes. Total LDL was subfractioned into native LDL and LDL(-) by anion-exchange chromatography and their sphingolipid content evaluated by mass spectrometry. LDL subfractions were incubated with monocytes in the presence or absence of enzyme inhibitors: chlorpromazine (CPZ), d-erythro-2-(N-myristoyl amino)-1-phenyl-1-propanol (MAPP), and N,N-dimethylsphingosine (DMS), which inhibit Cer, Sph, and S1P generation, respectively. After incubation, we evaluated cytokine release by enzyme-linked immunosorbent assay (ELISA) and apoptosis by flow cytometry. LDL(-) had an increased content in Cer and Sph compared to LDL(+). LDL(-)-induced cytokine release from cultured monocytes was inhibited by CPZ and MAPP, whereas DMS had no effect. LDL(-) promoted monocyte apoptosis, which was inhibited by CPZ, but increased with the addition of DMS. LDL enriched with Sph increased cytokine release in monocytes, and when enriched with Cer, reproduced both the apoptotic and inflammatory effects of LDL(-). These observations indicate that Cer content contributes to the inflammatory and apoptotic effects of LDL(-) on monocytes, whereas Sph plays a more important role in LDL(-)-induced inflammation, and S1P counteracts apoptosis.
Collapse
Affiliation(s)
- Núria Puig
- Cardiovascular Biochemistry. Biomedical Research Institute Sant Pau (IIB-Sant Pau), Barcelona, Spain. C/Sant Quinti 77-79, 08041 Barcelona, Spain
- Molecular Biology and Biochemistry Department, Universitat Autònoma de Barcelona (UAB) Faculty of Medicine. Building M. Cerdanyola del Vallès, 08193 Barcelona, Spain
| | - Montserrat Estruch
- Cardiovascular Biochemistry. Biomedical Research Institute Sant Pau (IIB-Sant Pau), Barcelona, Spain. C/Sant Quinti 77-79, 08041 Barcelona, Spain
| | - Lei Jin
- Cardiovascular Biochemistry. Biomedical Research Institute Sant Pau (IIB-Sant Pau), Barcelona, Spain. C/Sant Quinti 77-79, 08041 Barcelona, Spain
| | - Jose Luis Sanchez-Quesada
- Cardiovascular Biochemistry. Biomedical Research Institute Sant Pau (IIB-Sant Pau), Barcelona, Spain. C/Sant Quinti 77-79, 08041 Barcelona, Spain
- CIBER of Diabetes and Metabolic Diseases (CIBERDEM), 28029 Madrid, Spain
| | - Sonia Benitez
- Cardiovascular Biochemistry. Biomedical Research Institute Sant Pau (IIB-Sant Pau), Barcelona, Spain. C/Sant Quinti 77-79, 08041 Barcelona, Spain.
| |
Collapse
|
22
|
Rivas-Urbina A, Rull A, Ordóñez-Llanos J, Sánchez-Quesada JL. Electronegative LDL: An Active Player in Atherogenesis or a By- Product of Atherosclerosis? Curr Med Chem 2019; 26:1665-1679. [PMID: 29600751 DOI: 10.2174/0929867325666180330093953] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Revised: 11/12/2017] [Accepted: 12/11/2017] [Indexed: 12/16/2022]
Abstract
Low-density lipoproteins (LDLs) are the major plasma carriers of cholesterol. However, LDL particles must undergo various molecular modifications to promote the development of atherosclerotic lesions. Modified LDL can be generated by different mechanisms, but as a common trait, show an increased electronegative charge of the LDL particle. A subfraction of LDL with increased electronegative charge (LDL(-)), which can be isolated from blood, exhibits several pro-atherogenic characteristics. LDL(-) is heterogeneous, due to its multiple origins but is strongly related to the development of atherosclerosis. Nevertheless, the implication of LDL(-) in a broad array of pathologic conditions is complex and in some cases anti-atherogenic LDL(-) properties have been reported. In fact, several molecular modifications generating LDL(-) have been widely studied, but it remains unknown as to whether these different mechanisms are specific or common to different pathological disorders. In this review, we attempt to address these issues examining the most recent findings on the biology of LDL(-) and discussing the relationship between this LDL subfraction and the development of different diseases with increased cardiovascular risk. Finally, the review highlights the importance of minor apolipoproteins associated with LDL(-) which would play a crucial role in the different properties displayed by these modified LDL particles.
Collapse
Affiliation(s)
- Andrea Rivas-Urbina
- Cardiovascular Biochemistry Group, Research Institute of the Hospital de Sant Pau (IIB Sant Pau), Barcelona, Spain.,Biochemistry and Molecular Biology Department, Universitat Autònoma de Barcelona, Cerdanyola, Spain
| | - Anna Rull
- Cardiovascular Biochemistry Group, Research Institute of the Hospital de Sant Pau (IIB Sant Pau), Barcelona, Spain.,Hospital Universitari Joan XXIII, IISPV, Universitat Rovira i Virgili, Tarragona, Spain
| | - Jordi Ordóñez-Llanos
- Cardiovascular Biochemistry Group, Research Institute of the Hospital de Sant Pau (IIB Sant Pau), Barcelona, Spain.,Biochemistry and Molecular Biology Department, Universitat Autònoma de Barcelona, Cerdanyola, Spain
| | - José Luis Sánchez-Quesada
- Cardiovascular Biochemistry Group, Research Institute of the Hospital de Sant Pau (IIB Sant Pau), Barcelona, Spain.,CIBERDEM. Institute of Health Carlos III, Madrid 28029, Spain
| |
Collapse
|
23
|
Marin-Palma D, Sirois CM, Urcuqui-Inchima S, Hernandez JC. Inflammatory status and severity of disease in dengue patients are associated with lipoprotein alterations. PLoS One 2019; 14:e0214245. [PMID: 30901375 PMCID: PMC6430398 DOI: 10.1371/journal.pone.0214245] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 03/09/2019] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION The triggering of severe dengue has been associated with an exacerbated inflammatory process characterized by the production of pro-inflammatory cytokines such as IL-1β/IL-18, which are the product of inflammasome activation. Furthermore, alteration in the levels of high-density (HDL) and low-density lipoproteins (LDL) has been observed; and HDL are known to have immunomodulatory properties, including the regulation of inflammasomes. While HDL would be expected to counteract hyperactivation of the inflammasome, the relationship between HDL and dengue severity, has not previously been explored. METHODOLOGY We conducted a cross-sectional study of 30 patients with dengue and 39 healthy controls matched by sex and age. Lipid profile and levels of C-reactive protein were quantified. Serum levels of IL-1β, IL-6, IL-10, IL-18, and TNF-α, were assessed by ELISA. Expression of inflammasome-related genes in PBMC was quantified by qPCR. RESULTS Dengue patients presented an alteration in the parameters of the lipid profile, with a significant decrease in HDL levels, which was more pronounced in dengue patients with warning signs. Moreover, a decrease in the expression of the inflammasome-related genes NLRP1, NLRC4, caspase-1, IL-1β and IL-18 was observed, as well as an increase in serum levels of C-reactive protein and IL-10 in dengue patients versus healthy donors. Significant positive correlations between LDL levels and the relative expression of NLRP3, NLRC4, IL-1β and IL-18, were found. CONCLUSION The results suggest that there is a relationship between the alteration of LDL and HDL with the imbalance in the inflammatory response, which could be associated with the severity of dengue.
Collapse
Affiliation(s)
- Damariz Marin-Palma
- Infettare, Facultad de Medicina, Universidad Cooperativa de Colombia, Medellin, Colombia
- Grupo Inmunovirologia, Facultad de Medicina, Universidad de Antioquia, UdeA, Medellín, Colombia
| | - Cherilyn M. Sirois
- Department of Biology & Chemistry, Springfield College, Springfield, MA, United States of America
| | - Silvio Urcuqui-Inchima
- Grupo Inmunovirologia, Facultad de Medicina, Universidad de Antioquia, UdeA, Medellín, Colombia
| | - Juan C. Hernandez
- Infettare, Facultad de Medicina, Universidad Cooperativa de Colombia, Medellin, Colombia
- * E-mail:
| |
Collapse
|
24
|
Witkowski A, Carta S, Lu R, Yokoyama S, Rubartelli A, Cavigiolio G. Oxidation of methionine residues in human apolipoprotein A-I generates a potent pro-inflammatory molecule. J Biol Chem 2019; 294:3634-3646. [PMID: 30635405 DOI: 10.1074/jbc.ra118.005663] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 12/27/2018] [Indexed: 02/06/2023] Open
Abstract
Amyloid deposits of apolipoprotein A-I (apoA-I) and inflammation are common in atherosclerotic arteries. In this study, we investigated the interplay between oxidation of apoA-I methionine residues (Met(O)-ApoA-I), a known amyloidogenic modification of apoA-I, and the inflammatory response of immune cells. Soluble pre-fibrillar Met(O)-ApoA-I, but not apoA-I, induced intracellular accumulation of pro-interleukin (IL)-1β and secretion of the pro-inflammatory cytokines tumor necrosis factor α (TNFα) and IL-6 in mouse bone marrow-derived macrophages (BMDMs) and human primary monocytes. Additionally, secretion of mature IL-1β was also activated in human monocytes. The pro-inflammatory activity of Met(O)-ApoA-I was Toll-like receptor 4 (TLR4)-dependent and CD36-independent and was solely determined by oxidation of apoA-I methionine residues, in particular Met-86 and Met-148. In contrast, amyloid fibrils or reconstituted high-density lipoproteins (HDLs) generated from Met(O)-ApoA-I did not induce cytokine production in BMDMs. Although lipid-free Met(O)-ApoA-I remained functional in extracting lipids from cells and generating HDL, it gained strong pro-inflammatory properties that may aggravate local inflammation in the arteries and atherosclerosis. Our study indicates that oxidation of apoA-I methionine residues produces a potent danger-associated molecular pattern capable of stimulating pro-inflammatory cytokine secretion at levels similar to those induced by known pathogen-associated molecular patterns, such as lipopolysaccharide.
Collapse
Affiliation(s)
- Andrzej Witkowski
- From the UCSF Benioff Children's Hospital Oakland Research Institute, Oakland, California 94609
| | - Sonia Carta
- the Cell Biology Unit, Ospedale Policlinico San Martino, 16132 Genova, Italy, and
| | - Rui Lu
- Food and Nutritional Sciences, Chubu University, Kasugai 487-8501, Japan
| | - Shinji Yokoyama
- Food and Nutritional Sciences, Chubu University, Kasugai 487-8501, Japan
| | - Anna Rubartelli
- the Cell Biology Unit, Ospedale Policlinico San Martino, 16132 Genova, Italy, and
| | - Giorgio Cavigiolio
- From the UCSF Benioff Children's Hospital Oakland Research Institute, Oakland, California 94609,
| |
Collapse
|
25
|
Ligi D, Benitez S, Croce L, Rivas-Urbina A, Puig N, Ordóñez-Llanos J, Mannello F, Sanchez-Quesada JL. Electronegative LDL induces MMP-9 and TIMP-1 release in monocytes through CD14 activation: Inhibitory effect of glycosaminoglycan sulodexide. Biochim Biophys Acta Mol Basis Dis 2018; 1864:3559-3567. [PMID: 30254012 DOI: 10.1016/j.bbadis.2018.09.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 09/03/2018] [Accepted: 09/17/2018] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Electronegative LDL (LDL(-)) is involved in atherosclerosis through the activation of the TLR4/CD14 inflammatory pathway in monocytes. Matrix metalloproteinases (MMP) and their inhibitors (tissue inhibitors of metalloproteinase [TIMP]) are also crucially involved in atherosclerosis, but their modulation by LDL(-) has never been investigated. The aim of this study was to examine the ability of LDL(-) to release MMPs and TIMPs in human monocytes and to determine whether sulodexide (SDX), a glycosaminoglycan-based drug, was able to affect their secretion. APPROACH AND RESULTS Native LDL (LDL(+)) and LDL(-) separated by anion-exchange chromatography were added to THP1-CD14 monocytes in the presence or absence of SDX for 24 h. A panel of 9 MMPs and 4 TIMPs was analyzed in cell supernatants with multiplex immunoassays. The gelatinolytic activity of MMP-9 was assessed by gelatin zymography. LDL(-) stimulated the release of MMP-9 (13-fold) and TIMP-1 (4-fold) in THP1-CD14 monocytes, as well as the gelatinolytic activity of MMP-9. Co-incubation of monocytes with LDL(-) and SDX for 24 h significantly reduced both the release of MMP-9 and TIMP-1 and gelatinase activity. In THP1 cells not expressing CD14, no effect of LDL(-) on MMP-9 or TIMP-1 release was observed. The uptake of DiI-labeled LDL(-) was higher than that of DiI-LDL(+) in THP1-CD14 but not in THP1 cells. This increase was inhibited by SDX. Experiments in microtiter wells coated with SDX demonstrated a specific interaction of LDL(-) with SDX. CONCLUSIONS LDL(-) induced the release of MMP-9 and TIMP-1 in monocytes through CD14. SDX affects the ability of LDL(-) to promote TIMP-1 and MMP-9 release by its interaction with LDL(-).
Collapse
Affiliation(s)
- Daniela Ligi
- Department of Biomolecular Sciences, Section of Clinical Biochemistry and Molecular Genetics, University Carlo Bo Urbino, Italy
| | - Sonia Benitez
- Cardiovascular Biochemistry, Biomedical Research Institute Sant Pau (IIB-Sant Pau), Barcelona, Spain, C/Sant Antoni M. Claret 167, 08025 Barcelona, Spain; Molecular Biology and Biochemistry Department, Universitat Autònoma de Barcelona (UAB). Cerdanyola del Vallès, Spain
| | - Lidia Croce
- Department of Biomolecular Sciences, Section of Clinical Biochemistry and Molecular Genetics, University Carlo Bo Urbino, Italy
| | - Andrea Rivas-Urbina
- Cardiovascular Biochemistry, Biomedical Research Institute Sant Pau (IIB-Sant Pau), Barcelona, Spain, C/Sant Antoni M. Claret 167, 08025 Barcelona, Spain; Molecular Biology and Biochemistry Department, Universitat Autònoma de Barcelona (UAB). Cerdanyola del Vallès, Spain
| | - Núria Puig
- Cardiovascular Biochemistry, Biomedical Research Institute Sant Pau (IIB-Sant Pau), Barcelona, Spain, C/Sant Antoni M. Claret 167, 08025 Barcelona, Spain; Molecular Biology and Biochemistry Department, Universitat Autònoma de Barcelona (UAB). Cerdanyola del Vallès, Spain
| | - Jordi Ordóñez-Llanos
- Cardiovascular Biochemistry, Biomedical Research Institute Sant Pau (IIB-Sant Pau), Barcelona, Spain, C/Sant Antoni M. Claret 167, 08025 Barcelona, Spain; Molecular Biology and Biochemistry Department, Universitat Autònoma de Barcelona (UAB). Cerdanyola del Vallès, Spain
| | - Ferdinando Mannello
- Department of Biomolecular Sciences, Section of Clinical Biochemistry and Molecular Genetics, University Carlo Bo Urbino, Italy.
| | - Jose Luis Sanchez-Quesada
- Cardiovascular Biochemistry, Biomedical Research Institute Sant Pau (IIB-Sant Pau), Barcelona, Spain, C/Sant Antoni M. Claret 167, 08025 Barcelona, Spain; CIBER of Diabetes and Metabolic Diseases (CIBERDEM).
| |
Collapse
|
26
|
Chu CS, Chan HC, Tsai MH, Stancel N, Lee HC, Cheng KH, Tung YC, Chan HC, Wang CY, Shin SJ, Lai WT, Yang CY, Dixon RA, Chen CH, Ke LY. Range of L5 LDL levels in healthy adults and L5's predictive power in patients with hyperlipidemia or coronary artery disease. Sci Rep 2018; 8:11866. [PMID: 30089847 PMCID: PMC6082876 DOI: 10.1038/s41598-018-30243-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 07/24/2018] [Indexed: 12/24/2022] Open
Abstract
Electronegative L5 low-density lipoprotein (LDL) level may be a useful biomarker for predicting cardiovascular disease. We determined the range of plasma L5 levels in healthy adults (n = 35) and examined the power of L5 levels to differentiate patients with coronary artery disease (CAD; n = 40) or patients with hyperlipidemia (HLP) without evidence of CAD (n = 35) from healthy adults. The percent L5 in total LDL (L5%) was quantified by using fast-protein liquid chromatography with an anion-exchange column. Receiver operating characteristic curve analysis was performed to determine cut-off values for L5 levels. The mean L5% and plasma concentration of L5 (ie, [L5]) were significantly higher in patients with HLP or CAD than in healthy adults (P < 0.001). The ranges of L5% and [L5] in healthy adults were determined to be <1.6% and <1.7 mg/dL, respectively. In individuals with L5% >1.6%, the odds ratio was 9.636 for HLP or CAD. In individuals with [L5] >1.7 mg/dL, the odds ratio was 17.684 for HLP or CAD. The power of L5% or [L5] to differentiate patients with HLP or CAD from healthy adults was superior to that of the LDL/high-density lipoprotein ratio. The ranges of L5% and [L5] in healthy adults determined here may be clinically useful in preventing and treating cardiovascular disease.
Collapse
Affiliation(s)
- Chih-Sheng Chu
- Lipid Science and Aging Research Center, Kaohsiung Medical University (KMU), Kaohsiung, Taiwan.,Center for Lipid Biosciences, KMU Hospital, KMU, Kaohsiung, Taiwan.,Department of Internal Medicine, KMU Hospital, KMU, Kaohsiung, Taiwan
| | - Hua-Chen Chan
- Center for Lipid Biosciences, KMU Hospital, KMU, Kaohsiung, Taiwan.,Vascular and Medicinal Research, Texas Heart Institute, Houston, TX, USA
| | - Ming-Hsien Tsai
- Center for Lipid Biosciences, KMU Hospital, KMU, Kaohsiung, Taiwan
| | - Nicole Stancel
- Vascular and Medicinal Research, Texas Heart Institute, Houston, TX, USA
| | - Hsiang-Chun Lee
- Lipid Science and Aging Research Center, Kaohsiung Medical University (KMU), Kaohsiung, Taiwan.,Department of Internal Medicine, KMU Hospital, KMU, Kaohsiung, Taiwan
| | - Kai-Hung Cheng
- Center for Lipid Biosciences, KMU Hospital, KMU, Kaohsiung, Taiwan.,Department of Internal Medicine, KMU Hospital, KMU, Kaohsiung, Taiwan
| | - Yi-Ching Tung
- Department of Public Health and Environmental Medicine, KMU, Kaohsiung, Taiwan
| | - Hsiu-Chuan Chan
- Lipid Science and Aging Research Center, Kaohsiung Medical University (KMU), Kaohsiung, Taiwan
| | - Chung-Ya Wang
- Center for Lipid Biosciences, KMU Hospital, KMU, Kaohsiung, Taiwan
| | - Shyi-Jang Shin
- Lipid Science and Aging Research Center, Kaohsiung Medical University (KMU), Kaohsiung, Taiwan.,Center for Lipid Biosciences, KMU Hospital, KMU, Kaohsiung, Taiwan.,Department of Internal Medicine, KMU Hospital, KMU, Kaohsiung, Taiwan
| | - Wen-Ter Lai
- Lipid Science and Aging Research Center, Kaohsiung Medical University (KMU), Kaohsiung, Taiwan.,Department of Internal Medicine, KMU Hospital, KMU, Kaohsiung, Taiwan
| | - Chao-Yuh Yang
- Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Richard A Dixon
- Department of Molecular Cardiology, Texas Heart Institute, Houston, TX, USA
| | - Chu-Huang Chen
- Lipid Science and Aging Research Center, Kaohsiung Medical University (KMU), Kaohsiung, Taiwan. .,Center for Lipid Biosciences, KMU Hospital, KMU, Kaohsiung, Taiwan. .,Department of Internal Medicine, KMU Hospital, KMU, Kaohsiung, Taiwan. .,New York Heart Research Foundation, Mineola, NY, USA.
| | - Liang-Yin Ke
- Lipid Science and Aging Research Center, Kaohsiung Medical University (KMU), Kaohsiung, Taiwan. .,Center for Lipid Biosciences, KMU Hospital, KMU, Kaohsiung, Taiwan. .,Department of Internal Medicine, KMU Hospital, KMU, Kaohsiung, Taiwan. .,Department of Medical Laboratory Science and Biotechnology, KMU, Kaohsiung, Taiwan.
| |
Collapse
|
27
|
Ke LY, Chan HC, Chan HC, Kalu FCU, Lee HC, Lin IL, Jhuo SJ, Lai WT, Tsao CR, Sawamura T, Dixon RA, Chen CH, Chu CS, Shin SJ. Electronegative Low-Density Lipoprotein L5 Induces Adipose Tissue Inflammation Associated With Metabolic Syndrome. J Clin Endocrinol Metab 2017; 102:4615-4625. [PMID: 29029093 DOI: 10.1210/jc.2017-01657] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 09/20/2017] [Indexed: 02/06/2023]
Abstract
CONTEXT Electronegative low-density lipoprotein (LDL) L5 is a naturally occurring, atherogenic entity found at elevated levels in the plasma of patients with metabolic syndrome (MetS) in the absence of elevated plasma LDL levels. OBJECTIVE To investigate the role of L5 in the mechanism of adipose tissue inflammation associated with MetS. PATIENTS/SETTING Plasma LDL isolated from patients with MetS (n = 29) and controls (n = 29) with similar plasma LDL levels was separated into five subfractions, L1 to L5, with increasing electronegativity. DESIGN We examined the invivo effects of L5 on adipose tissue in mice and the in vitro effects of L5 on adipocytokine signaling and monocytes. RESULTS Tail-vein injection of human L5 but not L1 into C57BL/6 mice induced the accumulation of F4/80+ and CD11c+ M1 macrophages. The effects of L5 were attenuated in mice deficient for L5's receptor, lectin-like oxidized LDL receptor 1 (LOX-1). L5 but not L1 induced human adipocytes to release inflammatory adipocytokines. Incubating human THP-1 monocytes with LDL-free culture media from L5-treated adipocytes enhanced the migration of monocytes by 300-fold (P < 0.001 vs L1-treated adipocyte media)-effects that were attenuated by LOX-1 neutralizing antibody. Migrated cells were positive for mature macrophage marker PM-2K, indicating the transformation of monocytes into macrophages. The infiltration of M1 macrophages in adipose tissue was also observed in a previously established hamster model of endogenously elevated L5. CONCLUSIONS L5 induces adipose inflammation through LOX-1 by promoting macrophage maturation and infiltration into adipose tissue. Elevated plasma L5 levels may be a novel etiology of adipose tissue inflammation in patients with MetS.
Collapse
Affiliation(s)
- Liang-Yin Ke
- Vascular and Medicinal Research, Texas Heart Institute
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Taiwan
- Lipid Science and Aging Research Center, Kaohsiung Medical University, Taiwan
- Center for Lipid Biosciences, Kaohsiung Medical University Hospital, Taiwan
| | - Hua-Chen Chan
- Vascular and Medicinal Research, Texas Heart Institute
- Lipid Science and Aging Research Center, Kaohsiung Medical University, Taiwan
- Center for Lipid Biosciences, Kaohsiung Medical University Hospital, Taiwan
| | - Hsiu-Chuan Chan
- Lipid Science and Aging Research Center, Kaohsiung Medical University, Taiwan
| | - Franklin Chikodi Udo Kalu
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Taiwan
| | - Hsiang-Chun Lee
- Lipid Science and Aging Research Center, Kaohsiung Medical University, Taiwan
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Taiwan
| | - I-Ling Lin
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Taiwan
| | - Shih-Jie Jhuo
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Taiwan
| | - Wen-Ter Lai
- Lipid Science and Aging Research Center, Kaohsiung Medical University, Taiwan
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Taiwan
| | - Chen-Rong Tsao
- Division of Cardiology, Department of Internal Medicine, Feng Yuan Hospital, Ministry of Health, Taiwan
| | - Tatsuya Sawamura
- Department of Physiology, School of Medicine, Shinshu University, Japan
| | | | - Chu-Huang Chen
- Vascular and Medicinal Research, Texas Heart Institute
- Lipid Science and Aging Research Center, Kaohsiung Medical University, Taiwan
- Center for Lipid Biosciences, Kaohsiung Medical University Hospital, Taiwan
- New York Heart Research Foundation
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Taiwan
| | - Chih-Sheng Chu
- Lipid Science and Aging Research Center, Kaohsiung Medical University, Taiwan
- Center for Lipid Biosciences, Kaohsiung Medical University Hospital, Taiwan
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Taiwan
| | - Shyi-Jang Shin
- Lipid Science and Aging Research Center, Kaohsiung Medical University, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Taiwan
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kaohsiung Medical University Hospital, Taiwan
| |
Collapse
|
28
|
Lehti S, Nguyen SD, Belevich I, Vihinen H, Heikkilä HM, Soliymani R, Käkelä R, Saksi J, Jauhiainen M, Grabowski GA, Kummu O, Hörkkö S, Baumann M, Lindsberg PJ, Jokitalo E, Kovanen PT, Öörni K. Extracellular Lipids Accumulate in Human Carotid Arteries as Distinct Three-Dimensional Structures and Have Proinflammatory Properties. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 188:525-538. [PMID: 29154769 DOI: 10.1016/j.ajpath.2017.09.019] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 09/11/2017] [Accepted: 09/26/2017] [Indexed: 12/12/2022]
Abstract
Lipid accumulation is a key characteristic of advancing atherosclerotic lesions. Herein, we analyzed the ultrastructure of the accumulated lipids in endarterectomized human carotid atherosclerotic plaques using three-dimensional (3D) electron microscopy, a method never used in this context before. 3D electron microscopy revealed intracellular lipid droplets and extracellular lipoprotein particles. Most of the particles were aggregated, and some connected to needle-shaped or sheet-like cholesterol crystals. Proteomic analysis of isolated extracellular lipoprotein particles revealed that apolipoprotein B is their main protein component, indicating their origin from low-density lipoprotein, intermediate-density lipoprotein, very-low-density lipoprotein, lipoprotein (a), or chylomicron remnants. The particles also contained small exchangeable apolipoproteins, complement components, and immunoglobulins. Lipidomic analysis revealed differences between plasma lipoproteins and the particles, thereby indicating involvement of lipolytic enzymes in their generation. Incubation of human monocyte-derived macrophages with the isolated extracellular lipoprotein particles or with plasma lipoproteins that had been lipolytically modified in vitro induced intracellular lipid accumulation and triggered inflammasome activation in them. Taken together, extracellular lipids accumulate in human carotid plaques as distinct 3D structures that include aggregated and fused lipoprotein particles and cholesterol crystals. The particles originate from plasma lipoproteins, show signs of lipolytic modifications, and associate with cholesterol crystals. By inducing intracellular cholesterol accumulation (ie, foam cell formation) and inflammasome activation, the extracellular lipoprotein particles may actively enhance atherogenesis.
Collapse
Affiliation(s)
- Satu Lehti
- Atherosclerosis Research Laboratory, Wihuri Research Institute, Helsinki, Finland
| | - Su D Nguyen
- Atherosclerosis Research Laboratory, Wihuri Research Institute, Helsinki, Finland
| | - Ilya Belevich
- Electron Microscopy Unit, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Helena Vihinen
- Electron Microscopy Unit, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Hanna M Heikkilä
- Molecular Neurology, Research Programs Unit, University of Helsinki, Helsinki, Finland
| | - Rabah Soliymani
- Clinical Proteomics Core Facility, Medicum-Biochemistry and Developmental Biology, School of Medicine, University of Helsinki, Helsinki, Finland
| | - Reijo Käkelä
- Helsinki University Lipidomics Unit, Department of Biosciences, University of Helsinki, Helsinki, Finland
| | - Jani Saksi
- Molecular Neurology, Research Programs Unit, University of Helsinki, Helsinki, Finland
| | - Matti Jauhiainen
- National Institute for Health and Welfare, Helsinki, Finland; Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Gregory A Grabowski
- Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Kiniksa Pharmaceuticals, Ltd., Wellesley, Massachusetts
| | - Outi Kummu
- Medical Microbiology and Immunology, Research Unit of Biomedicine, University of Oulu, Oulu, Finland
| | - Sohvi Hörkkö
- Medical Microbiology and Immunology, Research Unit of Biomedicine, University of Oulu, Oulu, Finland; Medical Research Center and Nordlab Oulu, University Hospital and University of Oulu, Oulu, Finland
| | - Marc Baumann
- Clinical Proteomics Core Facility, Medicum-Biochemistry and Developmental Biology, School of Medicine, University of Helsinki, Helsinki, Finland
| | - Perttu J Lindsberg
- Molecular Neurology, Research Programs Unit, University of Helsinki, Helsinki, Finland; Clinical Neurosciences, Neurology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Eija Jokitalo
- Electron Microscopy Unit, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Petri T Kovanen
- Atherosclerosis Research Laboratory, Wihuri Research Institute, Helsinki, Finland
| | - Katariina Öörni
- Atherosclerosis Research Laboratory, Wihuri Research Institute, Helsinki, Finland; Helsinki University Lipidomics Unit, Department of Biosciences, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
29
|
Estruch M, Miñambres I, Sanchez-Quesada JL, Soler M, Pérez A, Ordoñez-Llanos J, Benitez S. Increased inflammatory effect of electronegative LDL and decreased protection by HDL in type 2 diabetic patients. Atherosclerosis 2017; 265:292-298. [PMID: 28734591 DOI: 10.1016/j.atherosclerosis.2017.07.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 07/04/2017] [Accepted: 07/12/2017] [Indexed: 12/19/2022]
Abstract
BACKGROUND AND AIMS Type 2 diabetic patients have an increased proportion of electronegative low-density lipoprotein (LDL(-)), an inflammatory LDL subfraction present in blood, and dysfunctional high-density lipoprotein (HDL). We aimed at examining the inflammatory effect of LDL(-) on monocytes and the counteracting effect of HDL in the context of type 2 diabetes. METHODS This was a cross-sectional study in which the population comprised 3 groups (n = 12 in each group): type 2 diabetic patients with good glycaemic control (GC-T2DM patients), type 2 diabetic patients with poor glycaemic control (PC-T2DM), and a control group. Total LDL, HDL, and monocytes were isolated from plasma of these subjects. LDL(-) was isolated from total LDL by anion-exchange chromatography. LDL(-) from the three groups of subjects was added to monocytes in the presence or absence of HDL, and cytokines released by monocytes were quantified by ELISA. RESULTS LDL(-) proportion and plasma inflammatory markers were increased in PC-T2DM patients. LDL(-) from PC-T2DM patients induced the highest IL1β, IL6, and IL10 release in monocytes compared to LDL(-) from GC-T2DM and healthy subjects, and presented the highest content of non-esterified fatty acids (NEFA). In turn, HDL from PC-T2DM patients showed the lowest ability to inhibit LDL(-)-induced cytokine release in parallel to an impaired ability to decrease NEFA content in LDL(-). CONCLUSIONS Our findings show an imbalance in the pro- and anti-inflammatory effects of lipoproteins from T2DM patients, particularly in PC-T2DM.
Collapse
Affiliation(s)
- Montserrat Estruch
- Cardiovascular Biochemistry, Biomedical Research Institute Sant Pau (IIB-Sant Pau), C/Sant Antoni M. Claret 167, 08025 Barcelona, Spain
| | - Inka Miñambres
- Endocrinology and Nutrition Department, Hospital de la Santa Creu i Sant Pau Barcelona, C/Sant Quintí 89, 08026 Barcelona, Spain
| | - Jose Luis Sanchez-Quesada
- Cardiovascular Biochemistry, Biomedical Research Institute Sant Pau (IIB-Sant Pau), C/Sant Antoni M. Claret 167, 08025 Barcelona, Spain; Molecular Biology and Biochemistry Department, Universitat Autònoma de Barcelona (UAB) Faculty of Medicine, Building M. Cerdanyola del Vallès, Spain
| | - Marta Soler
- Flow Cytometry Platform, Biomedical Research Institute Sant Pau (IIB-Sant Pau), C/Sant Antoni M. Claret 167, 08025 Barcelona, Spain
| | - Antonio Pérez
- Endocrinology and Nutrition Department, Hospital de la Santa Creu i Sant Pau Barcelona, C/Sant Quintí 89, 08026 Barcelona, Spain
| | - Jordi Ordoñez-Llanos
- Molecular Biology and Biochemistry Department, Universitat Autònoma de Barcelona (UAB) Faculty of Medicine, Building M. Cerdanyola del Vallès, Spain; Biochemistry Department, Hospital de la Santa Creu i Sant Pau Barcelona, C/Sant Quintí 89, 08026 Barcelona, Spain
| | - Sonia Benitez
- Cardiovascular Biochemistry, Biomedical Research Institute Sant Pau (IIB-Sant Pau), C/Sant Antoni M. Claret 167, 08025 Barcelona, Spain; Molecular Biology and Biochemistry Department, Universitat Autònoma de Barcelona (UAB) Faculty of Medicine, Building M. Cerdanyola del Vallès, Spain.
| |
Collapse
|
30
|
Yang TC, Chang PY, Lu SC. L5-LDL from ST-elevation myocardial infarction patients induces IL-1β production via LOX-1 and NLRP3 inflammasome activation in macrophages. Am J Physiol Heart Circ Physiol 2016; 312:H265-H274. [PMID: 27864235 DOI: 10.1152/ajpheart.00509.2016] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 10/28/2016] [Accepted: 11/11/2016] [Indexed: 12/24/2022]
Abstract
L5-LDL, the most electronegative LDL associated with major cardiovascular risks, significantly rises in patients with ST-segment elevation myocardial infarction (STEMI). The inflammatory nature of atherosclerotic vascular diseases has prompted us to investigate whether L5-LDL induces the production of inflammatory cytokines, especially vascular ischemia-related interleukin (IL)-1β, in the pathogenesis of STEMI. Clinical data showed that plasma levels of L5-LDL and IL-1β were higher in the STEMI patients than in the controls (P < 0.05). In THP-1-derived human macrophages, L5-LDL significantly increased the levels of both IL-1β and cleaved caspase-1, indicating the activation of NOD-like receptor pyrin domain containing 3 (NLRP3) inflammasomes by L5-LDL. Knockdown of NLRP3 and its adaptor protein apoptosis-associated speck-like protein containing a CARD (ASC) resulted in decreased L5-LDL-induced IL-1β. Furthermore, knock down of the lectin-type oxidized LDL receptor (LOX-1) in THP-1 cells attenuated L5-LDL-induced activation of NF-κB and caspase-1, leading to subsequent inhibition of IL-1β in macrophages. Furthermore, blockade LOX-1 with neutralizing antibody also inhibited L5-LDL-induced IL-1β in human peripheral blood mononuclear cell-derived macrophages. In conclusion, L5-LDL induces IL-1β production in macrophages by activation of NF-κB and caspase-1 through the LOX-1-dependent pathway. This study represents the evidence linking L5-LDL and the inflammatory cytokine IL-1β in STEMI, and identifies L5-LDL as a novel therapeutic target in acute myocardial infarction. NEW & NOTEWORTHY This study represents the evidence linking L5-LDL and the inflammatory cytokine IL-1β in ST-segment elevation myocardial infarction (STEMI). We elucidate the molecular mechanism underlying L5-LDL-induced production of IL-1β in macrophages. The results showed that L5-LDL induced activation of caspase-1 and NF-κB through the lectin-type oxidized LDL receptor (LOX-1)-dependent pathway, leading to the production of IL-1β.
Collapse
Affiliation(s)
- Tzu-Ching Yang
- Department of Biochemistry and Molecular Biology, National Taiwan University College of Medicine, Taipei, Taiwan; and
| | - Po-Yuan Chang
- Cardiovasccular Center and Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - Shao-Chun Lu
- Department of Biochemistry and Molecular Biology, National Taiwan University College of Medicine, Taipei, Taiwan; and
| |
Collapse
|
31
|
Chen CH, Ke LY, Chan HC, Lee AS, Lin KD, Chu CS, Lee MY, Hsiao PJ, Hsu C, Chen CH, Shin SJ. Electronegative low density lipoprotein induces renal apoptosis and fibrosis: STRA6 signaling involved. J Lipid Res 2016; 57:1435-46. [PMID: 27256691 PMCID: PMC4959859 DOI: 10.1194/jlr.m067215] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Indexed: 02/07/2023] Open
Abstract
Dyslipidemia has been proven to capably develop and aggravate chronic kidney disease. We also report that electronegative LDL (L5) is the most atherogenic LDL. On the other hand, retinoic acid (RA) and RA receptor (RAR) agonist are reported to be beneficial in some kidney diseases. “Stimulated by retinoic acid 6” (STRA6), one retinol-binding protein 4 receptor, was recently identified to regulate retinoid homeostasis. Here, we observed that L5 suppressed STRA6 cascades [STRA6, cellular retinol-binding protein 1 (CRBP1), RARs, retinoid X receptor α, and retinol, RA], but L5 simultaneously induced apoptosis and fibrosis (TGFβ1, Smad2, collagen 1, hydroxyproline, and trichrome) in kidneys of L5-injected mice and L5-treated renal tubular cells. These L5-induced changes of STRA6 cascades, renal apoptosis, and fibrosis were reversed in kidneys of LOX1−/− mice. LOX1 RNA silencing and inhibitor of c-Jun N-terminal kinase and p38MAPK rescued the suppression of STRA6 cascades and apoptosis and fibrosis in L5-treated renal tubular cells. Furthermore, crbp1 gene transfection reversed downregulation of STRA6 cascades, apoptosis, and fibrosis in L5-treated renal tubular cells. For mimicking STRA6 deficiency, efficient silencing of STRA6 RNA was performed and was found to repress STRA6 cascades and caused apoptosis and fibrosis in L1-treated renal tubular cells. In summary, this study reveals that electronegative L5 can cause kidney apoptosis and fibrosis via the suppression of STRA6 cascades, and implicates that STRA6 signaling may be involved in dyslipidemia-mediated kidney disease.
Collapse
Affiliation(s)
- Chao-Hung Chen
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Liang-Yin Ke
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan Lipid Science and Aging Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hua-Chen Chan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan Lipid Science and Aging Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - An-Sheng Lee
- Department of Medicine, Mackay Medical College, New Taipei, Taiwan
| | - Kun-Der Lin
- Divisions of Endocrinology and Metabolism Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Chih-Sheng Chu
- Cardiology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Mei-Yueh Lee
- Divisions of Endocrinology and Metabolism Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Pi-Jung Hsiao
- Divisions of Endocrinology and Metabolism Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan Departments of Internal Medicine College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chin Hsu
- Physiology, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chu-Huang Chen
- Lipid Science and Aging Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan Department of Vascular and Medical Research, Texas Heart Institute, Houston, TX
| | - Shyi-Jang Shin
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan Lipid Science and Aging Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan Divisions of Endocrinology and Metabolism Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan Departments of Internal Medicine College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
32
|
Estruch M, Sanchez-Quesada JL, Ordoñez-Llanos J, Benitez S. Inflammatory intracellular pathways activated by electronegative LDL in monocytes. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:963-969. [PMID: 27235719 DOI: 10.1016/j.bbalip.2016.05.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 05/10/2016] [Accepted: 05/23/2016] [Indexed: 12/12/2022]
Abstract
AIMS Electronegative LDL (LDL(-)) is a plasma LDL subfraction that induces cytokine release in monocytes through toll-like receptor 4 (TLR4) activation. However, the intracellular pathways induced by LDL(-) downstream TLR4 activation are unknown. We aimed to identify the pathways activated by LDL(-) leading to cytokine release in monocytes. METHODS AND RESULTS We determined LDL(-)-induced activation of several intracellular kinases in protein extracts from monocytes using a multikinase ELISA array. LDL(-) induced higher p38 mitogen-activated protein kinase (MAPK) phosphorylation than native LDL. This was corroborated by a specific cell-based assay and it was dependent on TLR4 and phosphoinositide 3-kinase (PI3k)/Akt pathway. P38 MAPK activation was involved in cytokine release promoted by LDL(-). A specific ELISA showed that LDL(-) activated cAMP response-element binding (CREB) in a p38 MAPK dependent manner. P38 MAPK was also involved in the nuclear factor kappa-B (NF-kB) and activating protein-1 (AP-1) activation by LDL(-). We found that NF-kB, AP-1 and CREB inhibitors decreased LDL(-)-induced cytokine release, mainly on MCP1, IL6 and IL10 release, respectively. CONCLUSIONS LDL(-) promotes p38 MAPK phosphorylation through TLR4 and PI3k/Akt pathways. Phosphorylation of p38 MAPK is involved in NF-kB, AP-1 and CREB activation, leading to LDL(-)-induced cytokine release in monocytes.
Collapse
Affiliation(s)
- Montserrat Estruch
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), Barcelona, Spain, C/Sant Antoni M. Claret 167, 08025 Barcelona, Spain.
| | - Jose Luis Sanchez-Quesada
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), Barcelona, Spain, C/Sant Antoni M. Claret 167, 08025 Barcelona, Spain; Molecular Biology and Biochemistry Department, Universitat Autònoma de Barcelona (UAB) Faculty of Medicine, Building M. Cerdanyola del Vallès, Spain.
| | - Jordi Ordoñez-Llanos
- Molecular Biology and Biochemistry Department, Universitat Autònoma de Barcelona (UAB) Faculty of Medicine, Building M. Cerdanyola del Vallès, Spain; Biochemistry Department, Hospital de la Santa Creu i Sant Pau Barcelona, C/Sant Quintí 89, 08026 Barcelona, Spain.
| | - Sonia Benitez
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), Barcelona, Spain, C/Sant Antoni M. Claret 167, 08025 Barcelona, Spain; Molecular Biology and Biochemistry Department, Universitat Autònoma de Barcelona (UAB) Faculty of Medicine, Building M. Cerdanyola del Vallès, Spain.
| |
Collapse
|
33
|
Hörl G, Froehlich H, Ferstl U, Ledinski G, Binder J, Cvirn G, Stojakovic T, Trauner M, Koidl C, Tafeit E, Amrein K, Scharnagl H, Jürgens G, Hallström S. Simvastatin Efficiently Lowers Small LDL-IgG Immune Complex Levels: A Therapeutic Quality beyond the Lipid-Lowering Effect. PLoS One 2016; 11:e0148210. [PMID: 26840480 PMCID: PMC4739583 DOI: 10.1371/journal.pone.0148210] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2015] [Accepted: 01/14/2016] [Indexed: 01/28/2023] Open
Abstract
We investigated a polyethylene glycol non-precipitable low-density lipoprotein (LDL) subfraction targeted by IgG and the influence of statin therapy on plasma levels of these small LDL-IgG-immune complexes (LDL-IgG-IC). LDL-subfractions were isolated from 6 atherosclerotic subjects and 3 healthy individuals utilizing iodixanol density gradient ultracentrifugation. Cholesterol, apoB and malondialdehyde (MDA) levels were determined in each fraction by enzymatic testing, dissociation-enhanced lanthanide fluorescence immunoassay and high-performance liquid chromatography, respectively. The levels of LDL-IgG-IC were quantified densitometrically following lipid electrophoresis, particle size distribution was assessed with dynamic light scattering and size exclusion chromatography. The influence of simvastatin (40 mg/day for three months) on small LDL-IgG-IC levels and their distribution among LDL-subfractions (salt gradient separation) were investigated in 11 patients with confirmed coronary artery disease (CAD). We demonstrate that the investigated LDL-IgG-IC are small particles present in atherosclerotic patients and healthy subjects. In vitro assembly of LDL-IgG-IC resulted in particle density shifts indicating a composition of one single molecule of IgG per LDL particle. Normalization on cholesterol levels revealed MDA values twice as high for LDL-subfractions rich in small LDL-IgG-IC if compared to dominant LDL-subfractions. Reactivity of affinity purified small LDL-IgG-IC to monoclonal antibody OB/04 indicates a high degree of modified apoB and oxidative modification. Simvastatin therapy studied in the CAD patients significantly lowered LDL levels and to an even higher extent, small LDL-IgG-IC levels without affecting their distribution. In conclusion simvastatin lowers levels of small LDL-IgG-IC more effectively than LDL-cholesterol and LDL-apoB levels in atherosclerotic patients. This antiatherogenic effect may additionally contribute to the known beneficial effects of this drug in the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Gerd Hörl
- Institute of Physiological Chemistry, Center of Physiological Medicine, Medical University of Graz, Graz, Austria
- * E-mail:
| | - Harald Froehlich
- Division of Angiology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Ulrika Ferstl
- Institute of Physiological Chemistry, Center of Physiological Medicine, Medical University of Graz, Graz, Austria
| | - Gerhard Ledinski
- Institute of Physiological Chemistry, Center of Physiological Medicine, Medical University of Graz, Graz, Austria
| | - Josepha Binder
- Division of Cardiology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Gerhard Cvirn
- Institute of Physiological Chemistry, Center of Physiological Medicine, Medical University of Graz, Graz, Austria
| | - Tatjana Stojakovic
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Michael Trauner
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Christoph Koidl
- Institute of Hygiene, Medical University of Graz, Graz, Austria
| | - Erwin Tafeit
- Institute of Physiological Chemistry, Center of Physiological Medicine, Medical University of Graz, Graz, Austria
| | - Karin Amrein
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Hubert Scharnagl
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Günther Jürgens
- Institute of Physiological Chemistry, Center of Physiological Medicine, Medical University of Graz, Graz, Austria
| | - Seth Hallström
- Institute of Physiological Chemistry, Center of Physiological Medicine, Medical University of Graz, Graz, Austria
| |
Collapse
|