1
|
Benitez S, Puig N, Camps-Renom P, Sánchez-Quesada JL. Atherogenic circulating lipoproteins in ischemic stroke. Front Cardiovasc Med 2024; 11:1470364. [PMID: 39713216 PMCID: PMC11659270 DOI: 10.3389/fcvm.2024.1470364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 11/22/2024] [Indexed: 12/24/2024] Open
Abstract
The fundamental role of qualitative alterations of lipoproteins in the early development of atherosclerosis has been widely demonstrated. Modified low-density lipoproteins (LDL), such as oxidized LDL (oxLDL), small dense LDL (sdLDL), and electronegative LDL [LDL(-)], are capable of triggering the atherogenic process, favoring the subendothelial accumulation of cholesterol and promoting inflammatory, proliferative, and apoptotic processes characteristic of atherosclerotic lesions. In contrast, high-density lipoprotein (HDL) prevents and/or reverses these atherogenic effects. However, LDL's atherogenic and HDL's anti-atherogenic actions may result altered in certain pathological conditions. The molecular mechanisms underlying the impaired effects of altered lipoproteins have been studied in numerous in vitro and in vivo studies, and have been extensively analyzed in coronary atherosclerosis, especially in the context of pathologies such as dyslipidemia, diabetes, obesity, and metabolic syndrome. However, the corresponding studies are scarcer in the field of ischemic stroke, despite carotid arteriosclerosis progression underlies at least 20% of ischemic strokes. The present review relates qualitative alterations of LDL and HDL with the development of carotid arteriosclerosis and the occurrence of ischemic stroke.
Collapse
Affiliation(s)
- Sonia Benitez
- Cardiovascular Biochemistry Group, Institut de Recerca Hospital de Sant Pau (IR Sant Pau), Barcelona, Spain
- CIBER-Diabetes and Metabolic Diseases (CIBERDEM), Madrid, Spain
| | - Núria Puig
- Cardiovascular Biochemistry Group, Institut de Recerca Hospital de Sant Pau (IR Sant Pau), Barcelona, Spain
| | - Pol Camps-Renom
- Stroke Unit, Department of Neurology, Hospital de La Santa Creu I Sant Pau, IR Sant Pau, Barcelona, Spain
| | - José Luis Sánchez-Quesada
- Cardiovascular Biochemistry Group, Institut de Recerca Hospital de Sant Pau (IR Sant Pau), Barcelona, Spain
- CIBER-Diabetes and Metabolic Diseases (CIBERDEM), Madrid, Spain
| |
Collapse
|
2
|
Akyol O, Yang CY, Woodside DG, Chiang HH, Chen CH, Gotto AM. Comparative Analysis of Atherogenic Lipoproteins L5 and Lp(a) in Atherosclerotic Cardiovascular Disease. Curr Atheroscler Rep 2024; 26:317-329. [PMID: 38753254 PMCID: PMC11192678 DOI: 10.1007/s11883-024-01209-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/05/2024] [Indexed: 06/22/2024]
Abstract
PURPOSE OF REVIEW Low-density lipoprotein (LDL) poses a risk for atherosclerotic cardiovascular disease (ASCVD). As LDL comprises various subtypes differing in charge, density, and size, understanding their specific impact on ASCVD is crucial. Two highly atherogenic LDL subtypes-electronegative LDL (L5) and Lp(a)-induce vascular cell apoptosis and atherosclerotic changes independent of plasma cholesterol levels, and their mechanisms warrant further investigation. Here, we have compared the roles of L5 and Lp(a) in the development of ASCVD. RECENT FINDINGS Lp(a) tends to accumulate in artery walls, promoting plaque formation and potentially triggering atherosclerosis progression through prothrombotic or antifibrinolytic effects. High Lp(a) levels correlate with calcific aortic stenosis and atherothrombosis risk. L5 can induce endothelial cell apoptosis and increase vascular permeability, inflammation, and atherogenesis, playing a key role in initiating atherosclerosis. Elevated L5 levels in certain high-risk populations may serve as a distinctive predictor of ASCVD. L5 and Lp(a) are both atherogenic lipoproteins contributing to ASCVD through distinct mechanisms. Lp(a) has garnered attention, but equal consideration should be given to L5.
Collapse
Affiliation(s)
- Omer Akyol
- Molecular Cardiology Research Laboratories, Vascular and Medicinal Research, The Texas Heart Institute, Houston, Texas, 77030, USA
| | - Chao-Yuh Yang
- Department of Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, Texas, 77030, USA
| | - Darren G Woodside
- Molecular Cardiology Research Laboratories, The Texas Heart Institute, Houston, TX, 77030, USA
| | - Huan-Hsing Chiang
- Molecular Cardiology Research Laboratories, Vascular and Medicinal Research, The Texas Heart Institute, Houston, Texas, 77030, USA
| | - Chu-Huang Chen
- Molecular Cardiology Research Laboratories, Vascular and Medicinal Research, The Texas Heart Institute, Houston, Texas, 77030, USA.
| | | |
Collapse
|
3
|
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
|
4
|
Presence of Ceramidase Activity in Electronegative LDL. Int J Mol Sci 2022; 24:ijms24010165. [PMID: 36613609 PMCID: PMC9820682 DOI: 10.3390/ijms24010165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 11/25/2022] [Accepted: 12/17/2022] [Indexed: 12/24/2022] Open
Abstract
Electronegative low-density lipoprotein (LDL(-)) is a minor modified fraction of human plasma LDL with several atherogenic properties. Among them is increased bioactive lipid mediator content, such as lysophosphatidylcholine (LPC), non-esterified fatty acids (NEFA), ceramide (Cer), and sphingosine (Sph), which are related to the presence of some phospholipolytic activities, including platelet-activating factor acetylhydrolase (PAF-AH), phospholipase C (PLC), and sphingomyelinase (SMase), in LDL(-). However, these enzymes' activities do not explain the increased Sph content, which typically derives from Cer degradation. In the present study, we analyzed the putative presence of ceramidase (CDase) activity, which could explain the increased Sph content. Thin layer chromatography (TLC) and lipidomic analysis showed that Cer, Sph, and NEFA spontaneously increased in LDL(-) incubated alone at 37 °C, in contrast with native LDL(+). An inhibitor of neutral CDase prevented the formation of Sph and, in turn, increased Cer content in LDL(-). In addition, LDL(-) efficiently degraded fluorescently labeled Cer (NBD-Cer) to form Sph and NEFA. These observations defend the existence of the CDase-like activity's association with LDL(-). However, neither the proteomic analysis nor the Western blot detected the presence of an enzyme with known CDase activity. Further studies are thus warranted to define the origin of the CDase-like activity detected in LDL(-).
Collapse
|
5
|
Gaggini M, Gorini F, Vassalle C. Lipids in Atherosclerosis: Pathophysiology and the Role of Calculated Lipid Indices in Assessing Cardiovascular Risk in Patients with Hyperlipidemia. Int J Mol Sci 2022; 24:ijms24010075. [PMID: 36613514 PMCID: PMC9820080 DOI: 10.3390/ijms24010075] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/15/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
The role of lipids is essential in any phase of the atherosclerotic process, which is considered a chronic lipid-related and inflammatory condition. The traditional lipid profile (including the evaluation of total cholesterol, triglycerides, high-density lipoprotein, and low-density lipoprotein) is a well-established tool to assess the risk of atherosclerosis and as such has been widely used as a pillar of cardiovascular disease prevention and as a target of pharmacological treatments in clinical practice over the last decades. However, other non-traditional lipids have emerged as possible alternative predictors of cardiometabolic risk in addition to traditional single or panel lipids, as they better reflect the overall interaction between lipid/lipoprotein fractions. Therefore, this review deals with the lipid involvement characterizing the pathophysiology of atherosclerosis, discussing some recently proposed non-traditional lipid indices and, in the light of available knowledge, their actual potential as new additive tools to better stratify cardiovascular risk in patients with hyperlipidemia as well as possible therapeutic targets in the clinical practice.
Collapse
Affiliation(s)
- Melania Gaggini
- Institute of Clinical Physiology, National Research Council, Via Moruzzi 1, 56124 Pisa, Italy
| | - Francesca Gorini
- Institute of Clinical Physiology, National Research Council, Via Moruzzi 1, 56124 Pisa, Italy
| | - Cristina Vassalle
- Fondazione CNR—Regione Toscana G Monasterio, Via Moruzzi 1, 56124 Pisa, Italy
- Correspondence:
| |
Collapse
|
6
|
Chen DY, Sawamura T, Dixon RAF, Sánchez-Quesada JL, Chen CH. Autoimmune Rheumatic Diseases: An Update on the Role of Atherogenic Electronegative LDL and Potential Therapeutic Strategies. J Clin Med 2021; 10:1992. [PMID: 34066436 PMCID: PMC8124242 DOI: 10.3390/jcm10091992] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 04/20/2021] [Accepted: 05/02/2021] [Indexed: 12/12/2022] Open
Abstract
Atherosclerosis has been linked with an increased risk of atherosclerotic cardiovascular disease (ASCVD). Autoimmune rheumatic diseases (AIRDs) are associated with accelerated atherosclerosis and ASCVD. However, the mechanisms underlying the high ASCVD burden in patients with AIRDs cannot be explained only by conventional risk factors despite disease-specific factors and chronic inflammation. Nevertheless, the normal levels of plasma low-density lipoprotein (LDL) cholesterol observed in most patients with AIRDs do not exclude the possibility of increased LDL atherogenicity. By using anion-exchange chromatography, human LDL can be divided into five increasingly electronegative subfractions, L1 to L5, or into electropositive and electronegative counterparts, LDL (+) and LDL (-). Electronegative L5 and LDL (-) have similar chemical compositions and can induce adverse inflammatory reactions in vascular cells. Notably, the percentage of L5 or LDL (-) in total LDL is increased in normolipidemic patients with AIRDs. Electronegative L5 and LDL (-) are not recognized by the normal LDL receptor but instead signal through the lectin-like oxidized LDL receptor 1 (LOX-1) to activate inflammasomes involving interleukin 1β (IL-1β). Here, we describe the detailed mechanisms of AIRD-related ASCVD mediated by L5 or LDL (-) and discuss the potential targeting of LOX-1 or IL-1β signaling as new therapeutic modalities for these diseases.
Collapse
Affiliation(s)
- Der-Yuan Chen
- Translational Medicine Center, China Medical University Hospital, Taichung 404, Taiwan;
- Rheumatology and Immunology Center, China Medical University Hospital, Taichung 404, Taiwan
- College of Medicine, China Medical University, Taichung 404, Taiwan
| | - Tatsuya Sawamura
- Department of Molecular Pathophysiology, Shinshu University School of Medicine, Matsumoto 390-8621, Japan;
- Department of Life Innovation, Institute for Biomedical Sciences, Shinshu University, Matsumoto 390-8621, Japan
| | - Richard A. F. Dixon
- Molecular Cardiology Research Laboratories, Texas Heart Institute, Houston, TX 77030, USA;
| | - José Luis Sánchez-Quesada
- Cardiovascular Biochemistry Group, Biomedical Research Institute IIB Sant Pau, 08041 Barcelona, Spain;
- CIBER of Diabetes and Metabolic Diseases (CIBERDEM), 08041 Barcelona, Spain
| | - Chu-Huang Chen
- Department of Life Innovation, Institute for Biomedical Sciences, Shinshu University, Matsumoto 390-8621, Japan
- Vascular and Medicinal Research, Texas Heart Institute, Houston, TX 77030, USA
- New York Heart Research Foundation, Mineola, NY 11501, USA
| |
Collapse
|
7
|
Changes in the Composition and Function of Lipoproteins after Bariatric Surgery in Patients with Severe Obesity. J Clin Med 2021; 10:jcm10081716. [PMID: 33923393 PMCID: PMC8071565 DOI: 10.3390/jcm10081716] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 04/14/2021] [Indexed: 11/30/2022] Open
Abstract
The effect of bariatric surgery on lipid profile and the qualitative characteristics of lipoproteins was analyzed in morbidly obese subjects. Thirteen obese patients underwent bariatric surgery. Plasma samples were obtained before surgery and at 6 and 12 months after the intervention. Thirteen healthy subjects comprised the control group. Lipid profile, hsCRP, and the composition and functional characteristics of VLDL, LDL, and HDL were assessed. At baseline, plasma from subjects with obesity had more triglycerides, VLDLc, and hsCRP, and less HDLc than the control group. These levels progressively normalized after surgery, although triglyceride and hsCRP levels remained higher than those in the controls. The main differences in lipoprotein composition between the obese subjects and the controls were increased apoE in VLDL, and decreased cholesterol and apoJ and increased apoC-III content in HDL. The pro-/anti-atherogenic properties of LDL and HDL were altered in the subjects with obesity at baseline compared with the controls, presenting smaller LDL particles that are more susceptible to modification and smaller HDL particles with decreased antioxidant capacity. Bariatric surgery normalized the composition of lipoproteins and improved the qualitative characteristics of LDL and HDL. In summary, patients with obesity present multiple alterations in the qualitative properties of lipoproteins compared with healthy subjects. Bariatric surgery reverted most of these alterations.
Collapse
|
8
|
Going "Green" in the Prevention and Management of Atherothrombotic Diseases: The Role of Dietary Polyphenols. J Clin Med 2021; 10:jcm10071490. [PMID: 33916712 PMCID: PMC8038361 DOI: 10.3390/jcm10071490] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/24/2021] [Accepted: 03/27/2021] [Indexed: 02/06/2023] Open
Abstract
During the 20th century processed and ready-to-eat foods became routinely consumed resulting in a sharp rise of fat, salt, and sugar intake in people's diets. Currently, the global incidence of obesity, raised blood lipids, hypertension, and diabetes in an increasingly aged population contributes to the rise of atherothrombotic events and cardiovascular diseases (CVD) mortality. Drug-based therapies are valuable strategies to tackle and help manage the socio-economic impact of atherothrombotic disorders though not without adverse side effects. The inclusion of fresh fruits and vegetables rich in flavonoids to human diets, as recommended by WHO offers a valuable nutritional strategy, alternative to drug-based therapies, to be explored in the prevention and management of atherothrombotic diseases at early stages. Though polyphenols are mostly associated to color and taste in foods, food flavonoids are emerging as modulators of cholesterol biosynthesis, appetite and food intake, blood pressure, platelet function, clot formation, and anti-inflammatory signaling, supporting the health-promoting effects of polyphenol-rich diets in mitigating the impact of risk factors in atherothrombotic disorders and CVD events. Here we overview the current knowledge on the effect of polyphenols particularly of flavonoid intake on the atherothrombotic risk factors and discuss the caveats and challenges involved with current experimental cell-based designs.
Collapse
|
9
|
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
|
10
|
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
|
11
|
Signaling Pathways and Key Genes Involved in Regulation of foam Cell Formation in Atherosclerosis. Cells 2020; 9:cells9030584. [PMID: 32121535 PMCID: PMC7140394 DOI: 10.3390/cells9030584] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 02/22/2020] [Accepted: 02/25/2020] [Indexed: 02/07/2023] Open
Abstract
Atherosclerosis is associated with acute cardiovascular conditions, such as ischemic heart disease, myocardial infarction, and stroke, and is the leading cause of morbidity and mortality worldwide. Our understanding of atherosclerosis and the processes triggering its initiation is constantly improving, and, during the last few decades, many pathological processes related to this disease have been investigated in detail. For example, atherosclerosis has been considered to be a chronic inflammation triggered by the injury of the arterial wall. However, recent works showed that atherogenesis is a more complex process involving not only the immune system, but also resident cells of the vessel wall, genetic factors, altered hemodynamics, and changes in lipid metabolism. In this review, we focus on foam cells that are crucial for atherosclerosis lesion formation. It has been demonstrated that the formation of foam cells is induced by modified low-density lipoprotein (LDL). The beneficial effects of the majority of therapeutic strategies with generalized action, such as the use of anti-inflammatory drugs or antioxidants, were not confirmed by clinical studies. However, the experimental therapies targeting certain stages of atherosclerosis, among which are lipid accumulation, were shown to be more effective. This emphasizes the relevance of future detailed investigation of atherogenesis and the importance of new therapies development.
Collapse
|
12
|
Scuruchi M, Potì F, Rodríguez-Carrio J, Campo GM, Mandraffino G. Biglycan and atherosclerosis: Lessons from high cardiovascular risk conditions. Biochim Biophys Acta Mol Cell Biol Lipids 2020; 1865:158545. [PMID: 31672572 DOI: 10.1016/j.bbalip.2019.158545] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 09/26/2019] [Accepted: 10/17/2019] [Indexed: 02/07/2023]
Abstract
Atherosclerosis (ATH) is a chronic, dynamic, evolutive process involving morphological and structural subversion of artery walls, leading to the formation of atherosclerotic plaques. ATH generally initiates during the childhood, occurring as a result of a number of changes in the intima tunica and in the media of arteries. A key event occurring during the pathobiology of ATH is the accumulation of lipoproteins in the sub-intimal spaces mediated by extracellular matrix (ECM) molecules, especially by the chondroitin sulfate/dermatan sulfate (CS/DS) -containing proteoglycans (CS/DSPGs). Among them, the proteoglycan biglycan (BGN) is critically involved in the onset and progression of ATH and evidences show that BGN represents the missing link between the pro-atherogenic status induced by both traditional and non-traditional cardiovascular risk factors and the development and progression of vascular damage. In the light of these findings, the role of BGN in dyslipidemia, hypertension, cigarette smoking, diabetes, chronic kidney disease and inflammatory status is briefly analyzed and discussed in order to shed new light on the underlying mechanisms governing the association between BGN and ATH.
Collapse
Affiliation(s)
- Michele Scuruchi
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Francesco Potì
- Department of Medicine and Surgery-Unit of Neurosciences, University of Parma, Parma, Italy
| | - Javier Rodríguez-Carrio
- Area of Immunology, Department of Functional Biology, Faculty of Medicine, University of Oviedo, Oviedo, Spain; Instituto de Investigación Sanitaria Del Principado de Asturias (ISPA), Oviedo, Spain; Bone and Mineral Research Unit, Instituto Reina Sofía de Investigación Nefrológica, REDinREN Del ISCIII, Hospital Universitario Central de Asturias, Oviedo, Spain
| | | | - Giuseppe Mandraffino
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy.
| |
Collapse
|
13
|
Orekhov AN, Nikiforov NG, Sukhorukov VN, Kubekina MV, Sobenin IA, Wu WK, Foxx KK, Pintus S, Stegmaier P, Stelmashenko D, Kel A, Gratchev AN, Melnichenko AA, Wetzker R, Summerhill VI, Manabe I, Oishi Y. Role of Phagocytosis in the Pro-Inflammatory Response in LDL-Induced Foam Cell Formation; a Transcriptome Analysis. Int J Mol Sci 2020; 21:ijms21030817. [PMID: 32012706 PMCID: PMC7037225 DOI: 10.3390/ijms21030817] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 01/14/2020] [Accepted: 01/23/2020] [Indexed: 12/25/2022] Open
Abstract
Excessive accumulation of lipid inclusions in the arterial wall cells (foam cell formation) caused by modified low-density lipoprotein (LDL) is the earliest and most noticeable manifestation of atherosclerosis. The mechanisms of foam cell formation are not fully understood and can involve altered lipid uptake, impaired lipid metabolism, or both. Recently, we have identified the top 10 master regulators that were involved in the accumulation of cholesterol in cultured macrophages induced by the incubation with modified LDL. It was found that most of the identified master regulators were related to the regulation of the inflammatory immune response, but not to lipid metabolism. A possible explanation for this unexpected result is a stimulation of the phagocytic activity of macrophages by modified LDL particle associates that have a relatively large size. In the current study, we investigated gene regulation in macrophages using transcriptome analysis to test the hypothesis that the primary event occurring upon the interaction of modified LDL and macrophages is the stimulation of phagocytosis, which subsequently triggers the pro-inflammatory immune response. We identified genes that were up- or downregulated following the exposure of cultured cells to modified LDL or latex beads (inert phagocytosis stimulators). Most of the identified master regulators were involved in the innate immune response, and some of them were encoding major pro-inflammatory proteins. The obtained results indicated that pro-inflammatory response to phagocytosis stimulation precedes the accumulation of intracellular lipids and possibly contributes to the formation of foam cells. In this way, the currently recognized hypothesis that the accumulation of lipids triggers the pro-inflammatory response was not confirmed. Comparative analysis of master regulators revealed similarities in the genetic regulation of the interaction of macrophages with naturally occurring LDL and desialylated LDL. Oxidized and desialylated LDL affected a different spectrum of genes than naturally occurring LDL. These observations suggest that desialylation is the most important modification of LDL occurring in vivo. Thus, modified LDL caused the gene regulation characteristic of the stimulation of phagocytosis. Additionally, the knock-down effect of five master regulators, such as IL15, EIF2AK3, F2RL1, TSPYL2, and ANXA1, on intracellular lipid accumulation was tested. We knocked down these genes in primary macrophages derived from human monocytes. The addition of atherogenic naturally occurring LDL caused a significant accumulation of cholesterol in the control cells. The knock-down of the EIF2AK3 and IL15 genes completely prevented cholesterol accumulation in cultured macrophages. The knock-down of the ANXA1 gene caused a further decrease in cholesterol content in cultured macrophages. At the same time, knock-down of F2RL1 and TSPYL2 did not cause an effect. The results obtained allowed us to explain in which way the inflammatory response and the accumulation of cholesterol are related confirming our hypothesis of atherogenesis development based on the following viewpoints: LDL particles undergo atherogenic modifications that, in turn, accompanied by the formation of self-associates; large LDL associates stimulate phagocytosis; as a result of phagocytosis stimulation, pro-inflammatory molecules are secreted; these molecules cause or at least contribute to the accumulation of intracellular cholesterol. Therefore, it became obvious that the primary event in this sequence is not the accumulation of cholesterol but an inflammatory response.
Collapse
Affiliation(s)
- Alexander N. Orekhov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 8 Baltiiskaya Street, 125315 Moscow, Russia
- Laboratory of Infection Pathology and Molecular Microecology, Institute of Human Morphology, 3 Tsyurupa Street, 117418 Moscow, Russia
- Correspondence: (A.N.O.); (V.I.S.)
| | - Nikita G. Nikiforov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 8 Baltiiskaya Street, 125315 Moscow, Russia
- Laboratory of Medical Genetics, Institute of Experimental Cardiology, National Medical Research Center of Cardiology, 15A 3-rd Cherepkovskaya Street, 121552 Moscow, Russia
- Centre of Collective Usage, Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilova Street, 119334 Moscow, Russia
| | - Vasily N. Sukhorukov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 8 Baltiiskaya Street, 125315 Moscow, Russia
- Laboratory of Infection Pathology and Molecular Microecology, Institute of Human Morphology, 3 Tsyurupa Street, 117418 Moscow, Russia
| | - Marina V. Kubekina
- Centre of Collective Usage, Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilova Street, 119334 Moscow, Russia
| | - Igor A. Sobenin
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 8 Baltiiskaya Street, 125315 Moscow, Russia
- Laboratory of Medical Genetics, Institute of Experimental Cardiology, National Medical Research Center of Cardiology, 15A 3-rd Cherepkovskaya Street, 121552 Moscow, Russia
| | - Wei-Kai Wu
- Department of Internal Medicine, National Taiwan University Hospital, Bei-Hu Branch, Taipei 10002, Taiwan
| | - Kathy K. Foxx
- Kalen Biomedical, LLC, Montgomery Village, MD 20886, USA
| | - Sergey Pintus
- BIOSOFT.RU, LLC, 630090 Novosibirsk, Russia
- Institute of Computational Technologies, 630090 Novosibirsk, Russia
| | | | - Daria Stelmashenko
- BIOSOFT.RU, LLC, 630090 Novosibirsk, Russia
- geneXplain GmbH, 38302 Wolfenbüttel, Germany
| | - Alexander Kel
- BIOSOFT.RU, LLC, 630090 Novosibirsk, Russia
- geneXplain GmbH, 38302 Wolfenbüttel, Germany
- Institute of Chemical Biology and Fundamental Medicine, 630090 Novosibirsk, Russia
| | - Alexei N. Gratchev
- N. N. Blokhin National Medical Research Center of Oncology, 24 Kashirskoye sh., 115478 Moscow, Russia
| | - Alexandra A. Melnichenko
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 8 Baltiiskaya Street, 125315 Moscow, Russia
- Laboratory of Medical Genetics, Institute of Experimental Cardiology, National Medical Research Center of Cardiology, 15A 3-rd Cherepkovskaya Street, 121552 Moscow, Russia
| | - Reinhard Wetzker
- Department of Anaesthesiology and Intensive Care Medicine, University Hospital Jena, Am Klinikum 1, D-07747 Jena, Germany
| | - Volha I. Summerhill
- Department of Basic Research, Institute for Atherosclerosis Research, 121609 Moscow, Russia
- Correspondence: (A.N.O.); (V.I.S.)
| | - Ichiro Manabe
- Department of Aging Research, Graduate School of Medicine, Chiba University, Chiba 263-8522, Japan
| | - Yumiko Oishi
- Department of Biochemistry & Molecular Biology, Nippon Medical School, Tokyo 113-8602, Japan
| |
Collapse
|
14
|
Familial Combined Hyperlipidemia (FCH) Patients with High Triglyceride Levels Present with Worse Lipoprotein Function Than FCH Patients with Isolated Hypercholesterolemia. Biomedicines 2020; 8:biomedicines8010006. [PMID: 31935793 PMCID: PMC7168323 DOI: 10.3390/biomedicines8010006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 01/02/2020] [Accepted: 01/03/2020] [Indexed: 02/06/2023] Open
Abstract
Lipoprotein characteristics were analyzed in familial combined hyperlipidemia (FCH) patients before and after statin treatment. Twenty-six FCH patients were classified according to the presence (HTG group, n = 13) or absence (normotriglyceridemic (NTG) group, n = 13) of hypertriglyceridemia. Fifteen healthy subjects comprised the control group. Lipid profile, inflammation markers, and qualitative characteristics of lipoproteins were assessed. Both groups of FCH subjects showed high levels of plasma C-reactive protein (CRP), lipoprotein-associated phospholipase A2 (Lp-PLA2) activity and apolipoprotein J. Statins reverted the increased levels of Lp-PLA2 and CRP. Lipoprotein composition alterations detected in FCH subjects were much more frequent in the HTG group, leading to dysfunctional low-density lipoproteins (LDL) and high-density lipoproteins (HDL). In the HTG group, LDL was smaller, more susceptible to oxidation, and contained more electronegative LDL (LDL(-)) compared to the NTG and control groups. Regarding HDL, the HTG group had less Lp-PLA2 activity than the NTG and control groups. HDL from both FCH groups was less anti-inflammatory than HDL from the control group. Statins increased LDL size, decreased LDL(-), and lowered Lp-PLA2 in HDL from HTG. In summary, pro-atherogenic alterations were more frequent and severe in the HTG group. Statins improved some alterations, but many remained unchanged in HTG.
Collapse
|
15
|
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
|
16
|
Itabe H, Kato R, Sawada N, Obama T, Yamamoto M. The Significance of Oxidized Low-Density Lipoprotein in Body Fluids as a Marker Related to Diseased Conditions. Curr Med Chem 2019. [PMID: 29521196 DOI: 10.2174/0929867325666180307114855] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Oxidatively modified low-density lipoprotein (oxLDL) is known to be involved in various diseases, including cardiovascular diseases. The presence of oxLDL in the human circulatory system and in atherosclerotic lesions has been demonstrated using monoclonal antibodies. Studies have shown the significance of circulating oxLDL in various systemic diseases, including acute myocardial infarction and diabetic mellitus. Several different enzyme-linked immunosorbent assay (ELISA) procedures to measure oxLDL were utilized. Evidence has been accumulating that reveals changes in oxLDL levels under certain pathological conditions. Since oxLDL concentration tends to correlate with low-density lipoprotein (LDL)-cholesterol, the ratio of ox-LDL and LDL rather than oxLDL concentration alone has also been focused. In addition to circulating plasma, LDL and oxLDL are found in gingival crevicular fluid (GCF), where the ratio of oxLDL to LDL in GCF is much higher than in plasma. LDL and oxLDL levels in GCF show an increase in diabetic patients and periodontal patients, suggesting that GCF might be useful in examining systemic conditions. GCF oxLDL increased when the teeth were affected by periodontitis. It is likely that oxLDL levels in plasma and GCF could reflect oxidative stress and transfer efficacy in the circulatory system.
Collapse
Affiliation(s)
- Hiroyuki Itabe
- Division of Biological Chemistry, Department of Molecular Biology, Showa University School of Pharmacy, Tokyo, Japan
| | - Rina Kato
- Division of Biological Chemistry, Department of Molecular Biology, Showa University School of Pharmacy, Tokyo, Japan
| | - Naoko Sawada
- Division of Biological Chemistry, Department of Molecular Biology, Showa University School of Pharmacy, Tokyo, Japan
| | - Takashi Obama
- Division of Biological Chemistry, Department of Molecular Biology, Showa University School of Pharmacy, Tokyo, Japan
| | - Matsuo Yamamoto
- Department of Periodontology, Showa University School of Dentistry, Tokyo, Japan
| |
Collapse
|
17
|
Pan M, Huo Y, Wang C, Zhang Y, Dai Z, Li B. Positively charged peptides from casein hydrolysate show strong inhibitory effects on LDL oxidation and cellular lipid accumulation in Raw264.7 cells. Int Dairy J 2019. [DOI: 10.1016/j.idairyj.2018.09.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
18
|
Dias IHK, Ferreira R, Gruber F, Vitorino R, Rivas-Urbina A, Sanchez-Quesada JL, Vieira Silva J, Fardilha M, de Freitas V, Reis A. Sulfate-based lipids: Analysis of healthy human fluids and cell extracts. Chem Phys Lipids 2019; 221:53-64. [PMID: 30910732 DOI: 10.1016/j.chemphyslip.2019.03.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 03/15/2019] [Accepted: 03/15/2019] [Indexed: 12/12/2022]
Abstract
Sulfate-based lipids (SL) have been proposed as players in inflammation, immunity and infection. In spite of the many biochemical processes linked to SL, analysis on this class of lipids has only focused on specific SL sub-classes in individual fluids or cells leaving a range of additional SL in other biological samples unaccounted for. This study describes the mass spectrometry screening of SL in lipid extracts of human fluids (saliva, plasma, urine, seminal fluid) and primary human cells (RBC, neutrophils, fibroblasts and skin epidermal) using targeted precursor ion scanning (PIS) approach. The PIS 97 mass spectra reveal a wide diversity of SL including steroid sulfates, sulfoglycolipids and other unidentified SL, as well as metabolites such as taurines, sulfated polyphenols and hypurate conjugates. Semi-quantification of SL revealed that plasma exhibited the highest content of SL whereas seminal fluid and epithelial cells contained the highest sulphur to phosphorous (S/P) ratio. The complexity of biofluids and cells sulfateome presented in this study highlight the importance of expanding the panel of synthetic sulfate-based lipid standards. Also, the heterogenous distribution of SL provides evidence for the interplay of sulfotransferases/sulfatases, opening new avenues for biomarker discovery in oral health, cardiovascular, fertility and dermatology research areas.
Collapse
Affiliation(s)
| | - Rita Ferreira
- Departamento de Quimica, Research Unit of Química Orgânica, Produtos Naturais e Agro-alimentares (QOPNA), Universidade de Aveiro, 3810-193, Aveiro, Portugal
| | - Florian Gruber
- Medical University of Vienna, Department of Dermatology, Vienna, Austria; Christian Doppler Laboratory for Biotechnology of Skin Aging, Vienna, Austria
| | - Rui Vitorino
- Unidade de Investigação Cardiovascular, Departamento de Cirurgia e Fisiologia, Faculdade de Medicina, Universidade do Porto, 4200-319, Porto, Portugal; Department of Medical Sciences, Institute of Biomedicine - iBiMED, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Andrea Rivas-Urbina
- Cardiovascular Biochemistry, Biomedical Research Institute IIB Sant Pau, Sant Antoni Ma Claret, 167, Barcelona, Spain
| | - José Luis Sanchez-Quesada
- Cardiovascular Biochemistry, Biomedical Research Institute IIB Sant Pau, Sant Antoni Ma Claret, 167, Barcelona, Spain
| | - Joana Vieira Silva
- Department of Medical Sciences, Institute of Biomedicine - iBiMED, University of Aveiro, 3810-193, Aveiro, Portugal; Reproductive Genetics & Embryo-fetal Development Group, Institute for Innovation and Health Research (I3S), University of Porto, Porto, Portugal; Unit for Multidisciplinary Research in Biomedicine (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Margarida Fardilha
- Department of Medical Sciences, Institute of Biomedicine - iBiMED, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Victor de Freitas
- LAQV/REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Rua do Campo Alegre 687, 4169-007, Porto, Portugal
| | - Ana Reis
- LAQV/REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Rua do Campo Alegre 687, 4169-007, Porto, Portugal.
| |
Collapse
|
19
|
Sukhorukov V, Gudelj I, Pučić-Baković M, Zakiev E, Orekhov A, Kontush A, Lauc G. Glycosylation of human plasma lipoproteins reveals a high level of diversity, which directly impacts their functional properties. Biochim Biophys Acta Mol Cell Biol Lipids 2019; 1864:643-653. [PMID: 30641224 DOI: 10.1016/j.bbalip.2019.01.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 12/18/2018] [Accepted: 01/06/2019] [Indexed: 01/25/2023]
Abstract
AIMS Human plasma lipoproteins are known to contain various glycan structures whose composition and functional importance are starting to be recognized. We assessed N-glycosylation of human plasma HDL and LDL and the role of their glycomes in cellular cholesterol metabolism. METHODS N-glycomic profiles of native and neuraminidase-treated HDL and LDL were obtained using HILIC-UHPLC-FLD. Relative abundance of the individual chromatographic peaks was quantitatively expressed as a percentage of total integrated area and N-glycan structures present in each peak were elucidated by MALDI-TOF MS. The capacity of HDL to mediate cellular efflux of cholesterol and the capacity of LDL to induce cellular accumulation of cholesteryl esters were evaluated in THP-1 cells. RESULTS HILIC-UHPLC-FLD analysis of HDL and LDL N-glycans released by PNGase F resulted in 22 and 18 distinct chromatographic peaks, respectively. The majority of N-glycans present in HDL (~70%) and LDL (~60%) were sialylated with one or two sialic acid residues. The most abundant N-glycan structure in both HDL and LDL was a complex type biantennary N-glycan with one sialic acid (A2G2S1). Relative abundances of several N-glycan structures were dramatically altered by the neuraminidase treatment, which selectively removed sialic acid residues. Native HDL displayed significantly greater efficacy in removing cellular cholesterol from THP-1 cells as compared to desialylated HDL (p < 0.05). Cellular accumulation of cholesteryl esters in THP-1 cells was significantly higher after incubations with desialylated LDL particles as compared to native LDL (p < 0.05). CONCLUSIONS N-glycome of human plasma lipoproteins reveals a high level of diversity, which directly impacts functional properties of the lipoproteins.
Collapse
Affiliation(s)
- Vasily Sukhorukov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia; National Institute for Health and Medical Research (INSERM), UMR 1166 ICAN, Paris F-75013, France; Sorbonne University, Paris F-75013, France; AP-HP, Groupe hospitalier Pitié-Salpétrière, Paris F-75013, France; Federal State Budget Institution of Sciences Institute of Gene Biology, Russian Academy of Sciences, Vavilova Str., 34/5, Moscow 119334, Russia
| | - Ivan Gudelj
- Genos Glycoscience Research Laboratory, Borongajska cesta 83H, HR-10 000 Zagreb, Croatia
| | - Maja Pučić-Baković
- Genos Glycoscience Research Laboratory, Borongajska cesta 83H, HR-10 000 Zagreb, Croatia
| | - Emile Zakiev
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia; National Institute for Health and Medical Research (INSERM), UMR 1166 ICAN, Paris F-75013, France; Sorbonne University, Paris F-75013, France; AP-HP, Groupe hospitalier Pitié-Salpétrière, Paris F-75013, France; Federal State Budget Institution of Sciences Institute of Gene Biology, Russian Academy of Sciences, Vavilova Str., 34/5, Moscow 119334, Russia
| | - Alexander Orekhov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia; Institute for Atherosclerosis Research, Skolkovo Innovative Center, 121609 Moscow, Russia
| | - Anatol Kontush
- National Institute for Health and Medical Research (INSERM), UMR 1166 ICAN, Paris F-75013, France; Sorbonne University, Paris F-75013, France; AP-HP, Groupe hospitalier Pitié-Salpétrière, Paris F-75013, France.
| | - Gordan Lauc
- Genos Glycoscience Research Laboratory, Borongajska cesta 83H, HR-10 000 Zagreb, Croatia
| |
Collapse
|
20
|
Rodríguez M, Guardiola M, Oliva I, Carles Vallvé J, Ferré R, Masana L, Parra S, Ribalta J, Castro A. Low-density lipoprotein net charge is a risk factor for atherosclerosis in lupus patients independent of lipid concentrations. Int J Rheum Dis 2018; 22:480-487. [PMID: 30450745 DOI: 10.1111/1756-185x.13445] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 09/17/2018] [Accepted: 10/21/2018] [Indexed: 01/20/2023]
Abstract
AIMS Patients with systemic lupus erythematosus (SLE) suffer from accelerated atherosclerosis. Their most common cause of death is a cardiovascular disease (CVD), in spite of the presence of moderate lipid alterations and normal cardiovascular risk scores. However, cholesterol still accumulates in the arteries of SLE patients, so we aim to identify additional factors that may help explain the residual risk that exists in these patients. We focus on investigating whether the net charge contributes significantly to both the development and the progression of atherosclerosis in patients with SLE. METHODS The lipoproteins from 78 patients with SLE and 32 controls were isolated via sequential ultracentrifugation. Lipoprotein subclasses distributions were analyzed via nuclear magnetic resonance spectroscopy and the net charges of very low-density lipoprotein (VLDL), intermediate-density lipoprotein (IDL), low-density lipoprotein (LDL) and high-density lipoprotein (HDL) were measured using a Zetasizer Nano-ZS. The degree of atherosclerosis (carotid intima-media thickness [cIMT]) was determined in all the participants. RESULTS Each lipoprotein class exhibited a negative net charge. IDL and LDL net charge correlated negatively with cIMT (r = -0.274, P = 0.034; r = -0.288; P = 0.033, respectively) in patients with SLE. This effect was independent of age, body mass index (BMI), gender, tobacco consumption, high-sensitivity C-reactive protein (hsCRP), lipid concentration and lipoprotein particle number. LDL net charge explained 4% of the cIMT variability among these patients; this contribution was also independent of age, BMI, gender, tobacco consumption, lipids levels, apolipoproteins and hsCRP. CONCLUSIONS Low-density lipoprotein net charge may be considered a new independent contributor to subclinical atherosclerosis in SLE patients. The observed relationship was independent of lipid concentrations and extends the prominent role that IDL and LDL play in cardiovascular risk.
Collapse
Affiliation(s)
- Marina Rodríguez
- Departament de Medicina i Cirurgia, Unitat de Recerca en Lípids i Arteriosclerosi, Universitat Rovira i Virgili, Reus, Spain.,Institut d'Investigació Sanitària Pere Virgili, Reus, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Spain
| | - Montse Guardiola
- Departament de Medicina i Cirurgia, Unitat de Recerca en Lípids i Arteriosclerosi, Universitat Rovira i Virgili, Reus, Spain.,Institut d'Investigació Sanitària Pere Virgili, Reus, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Spain
| | - Iris Oliva
- Departament de Medicina i Cirurgia, Unitat de Recerca en Lípids i Arteriosclerosi, Universitat Rovira i Virgili, Reus, Spain.,Institut d'Investigació Sanitària Pere Virgili, Reus, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Spain
| | - Joan Carles Vallvé
- Departament de Medicina i Cirurgia, Unitat de Recerca en Lípids i Arteriosclerosi, Universitat Rovira i Virgili, Reus, Spain.,Institut d'Investigació Sanitària Pere Virgili, Reus, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Spain
| | - Raimon Ferré
- Departament de Medicina i Cirurgia, Unitat de Recerca en Lípids i Arteriosclerosi, Universitat Rovira i Virgili, Reus, Spain.,Institut d'Investigació Sanitària Pere Virgili, Reus, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Spain.,Unitat de Medicina Vascular i Metabolisme (UVASMET), Hospital Universitari Sant Joan de Reus, Reus, Spain
| | - Lluís Masana
- Departament de Medicina i Cirurgia, Unitat de Recerca en Lípids i Arteriosclerosi, Universitat Rovira i Virgili, Reus, Spain.,Institut d'Investigació Sanitària Pere Virgili, Reus, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Spain.,Unitat de Medicina Vascular i Metabolisme (UVASMET), Hospital Universitari Sant Joan de Reus, Reus, Spain
| | - Sandra Parra
- Institut d'Investigació Sanitària Pere Virgili, Reus, Spain.,Unitat de Malalties Autoinmunes, Medicina Interna, Hospital Universitari Sant Joan de Reus, Reus, Spain
| | - Josep Ribalta
- Departament de Medicina i Cirurgia, Unitat de Recerca en Lípids i Arteriosclerosi, Universitat Rovira i Virgili, Reus, Spain.,Institut d'Investigació Sanitària Pere Virgili, Reus, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Spain
| | - Antoni Castro
- Institut d'Investigació Sanitària Pere Virgili, Reus, Spain.,Unitat de Malalties Autoinmunes, Medicina Interna, Hospital Universitari Sant Joan de Reus, Reus, Spain
| |
Collapse
|
21
|
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
|
22
|
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
|
23
|
Abstract
PURPOSE OF REVIEW Lipoprotein-induced intracellular lipid accumulation (foam cell formation) is a trigger of atherogenesis at the subendothelial arterial cell level. The purpose of this review is to describe the recent data related to the possible mechanisms of LDL-induced formation of lipid-laden foam cells and their role in the onset and development of atherosclerotic lesion. RECENT FINDINGS The most interesting current studies are related to the factors affecting foam cell formation. SUMMARY The phenomenon of lipid accumulation in cultured cells became the basis for creating a cellular test system that has already been successfully applied for development of drugs possessing direct antiatherosclerotic activity, and then the efficacy of these drugs was demonstrated in clinical studies. Moreover, this test system could be used for diagnostic assessing lipoproteins atherogenicity.
Collapse
Affiliation(s)
- Alexander N Orekhov
- Laboratory of Angiopatology, Institute of General Pathology and Pathophysiology
- Institute for Atherosclerosis Research, Skolkovo Innovative Center, Moscow, Russia
| |
Collapse
|
24
|
Orekhov AN, Ivanova EA, Melnichenko AA, Sobenin IA. Circulating desialylated low density lipoprotein. COR ET VASA 2017. [DOI: 10.1016/j.crvasa.2016.10.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
25
|
Akyol S, Lu J, Akyol O, Akcay F, Armutcu F, Ke LY, Chen CH. The role of electronegative low-density lipoprotein in cardiovascular diseases and its therapeutic implications. Trends Cardiovasc Med 2016; 27:239-246. [PMID: 28040327 DOI: 10.1016/j.tcm.2016.11.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 11/01/2016] [Accepted: 11/15/2016] [Indexed: 11/30/2022]
Abstract
Cardiovascular disease (CVD) is a health problem of great concern to both the public and medical authorities. Low-density lipoprotein (LDL) has been reported to play an important role in both the development and progression of CVD, but studies are underway to determine how LDL exerts its effects. In recent years, it has been found that LDL has several subfractions, each of which affects endothelial function differently; L5, the most electronegative fraction, has been shown to be unique in that it induces an atherogenic response. This review examines the current knowledge concerning the relationships between L5 and CVD and highlights the role of L5 in the pathophysiology of CVD, especially with regards to atherosclerosis.
Collapse
Affiliation(s)
- Sumeyya Akyol
- Vascular & Medicinal Research, Texas Heart Institute, 6770 Bertner Avenue, MC 2-255, Houston, TX 77030, USA; Department of Medical Biology, Faculty of Medicine, Turgut Ozal University, Ankara, Turkey.
| | - Jonathan Lu
- Vascular & Medicinal Research, Texas Heart Institute, 6770 Bertner Avenue, MC 2-255, Houston, TX 77030, USA
| | - Omer Akyol
- Vascular & Medicinal Research, Texas Heart Institute, 6770 Bertner Avenue, MC 2-255, Houston, TX 77030, USA; Department of Medical Biochemistry, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Fatih Akcay
- Department of Medical Biochemistry, Faculty of Medicine, Ataturk University, Erzurum, Turkey
| | - Ferah Armutcu
- Department of Medical Biochemistry, Cerrahpasa Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Liang-Yin Ke
- Lipid Science and Aging Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan; Center for Lipid Biosciences, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan; Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung, Taiwan.
| | - Chu-Huang Chen
- Vascular & Medicinal Research, Texas Heart Institute, 6770 Bertner Avenue, MC 2-255, Houston, TX 77030, USA; Lipid Science and Aging Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan; Center for Lipid Biosciences, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| |
Collapse
|
26
|
Cavalcante MF, Kazuma SM, Bender EA, Adorne MD, Ullian M, Veras MM, Saldiva PHN, Maranhão AQ, Guterres SS, Pohlmann AR, Abdalla DSP. A nanoformulation containing a scFv reactive to electronegative LDL inhibits atherosclerosis in LDL receptor knockout mice. Eur J Pharm Biopharm 2016; 107:120-9. [PMID: 27378286 DOI: 10.1016/j.ejpb.2016.07.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Revised: 06/24/2016] [Accepted: 07/01/2016] [Indexed: 01/21/2023]
Abstract
Atherosclerosis is a chronic inflammatory disease responsible for the majority of cases of myocardial infarction and ischemic stroke. The electronegative low-density lipoprotein, a modified subfraction of native LDL, is pro-inflammatory and plays an important role in atherogenesis. To investigate the effects of a nanoformulation (scFv anti-LDL(-)-MCMN-Zn) containing a scFv reactive to LDL(-) on the inhibition of atherosclerosis, its toxicity was evaluated in vitro and in vivo and further it was also administered weekly to LDL receptor knockout mice. The scFv anti-LDL(-)-MCMN-Zn nanoformulation did not induce cell death in RAW 264.7 macrophages and HUVECs. The 5mg/kg dose of scFv anti-LDL(-)-MCMN-Zn did not cause any typical signs of toxicity and it was chosen for the evaluation of its atheroprotective effect in Ldlr(-/-) mice. This nanoformulation significantly decreased the atherosclerotic lesion area at the aortic sinus, compared with that in untreated mice. In addition, the Il1b mRNA expression and CD14 protein expression were downregulated in the atherosclerotic lesions at the aortic arch of Ldlr(-/-) mice treated with scFv anti-LDL(-)-MCMN-Zn. Thus, the scFv anti-LDL(-)-MCMN-Zn nanoformulation inhibited the progression of atherosclerotic lesions, indicating its potential use in a future therapeutic strategy for atherosclerosis.
Collapse
Affiliation(s)
- Marcela Frota Cavalcante
- Department of Clinical and Toxicological Analyses, Faculty of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Soraya Megumi Kazuma
- Department of Clinical and Toxicological Analyses, Faculty of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Eduardo André Bender
- Department of Organic Chemistry, Chemistry Institute, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Márcia Duarte Adorne
- Department of Organic Chemistry, Chemistry Institute, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Mayara Ullian
- Department of Clinical and Toxicological Analyses, Faculty of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Mariana Matera Veras
- LIM5, Department of Pathology, Medicine School, University of Sao Paulo, Sao Paulo, SP, Brazil
| | | | - Andrea Queiroz Maranhão
- Molecular Immunology Laboratory, Department of Cell Biology, University of Brasilia, Brasilia, Distrito Federal, Brazil
| | - Silvia Stanisçuaski Guterres
- Department of Production and Control of Medicines, Faculty of Pharmacy, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Adriana Raffin Pohlmann
- Department of Organic Chemistry, Chemistry Institute, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Dulcineia Saes Parra Abdalla
- Department of Clinical and Toxicological Analyses, Faculty of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil.
| |
Collapse
|
27
|
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
|
28
|
Thermal stability of human plasma electronegative low-density lipoprotein: A paradoxical behavior of low-density lipoprotein aggregation. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:1015-1024. [PMID: 27233433 DOI: 10.1016/j.bbalip.2016.05.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 04/08/2016] [Accepted: 05/21/2016] [Indexed: 12/20/2022]
Abstract
Low-density lipoprotein (LDL) aggregation is central in triggering atherogenesis. A minor fraction of electronegative plasma LDL, termed LDL(-), plays a special role in atherogenesis. To better understand this role, we analyzed the kinetics of aggregation, fusion and disintegration of human LDL and its fractions, LDL(+) and LDL(-). Thermal denaturation of LDL was monitored by spectroscopy and electron microscopy. Initially, LDL(-) aggregated and fused faster than LDL(+), but later the order reversed. Most LDL(+) disintegrated and precipitated upon prolonged heating. In contrast, LDL(-) partially retained lipoprotein morphology and formed soluble aggregates. Biochemical analysis of all fractions showed no significant degradation of major lipids, mild phospholipid oxidation, and an increase in non-esterified fatty acid (NEFA) upon thermal denaturation. The main baseline difference between LDL subfractions was higher content of NEFA in LDL(-). Since NEFA promote lipoprotein fusion, increased NEFA content can explain rapid initial aggregation and fusion of LDL(-) but not its resistance to extensive disintegration. Partial hydrolysis of apoB upon heating was similar in LDL subfractions, suggesting that minor proteins importantly modulate LDL disintegration. Unlike LDL(+), LDL(-) contains small amounts of apoA-I and apoJ. Addition of exogenous apoA-I to LDL(+) hampered lipoprotein aggregation, fusion and precipitation, while depletion of endogenous apoJ had an opposite effect. Therefore, the initial rapid aggregation of LDL(-) is apparently counterbalanced by the stabilizing effects of minor proteins such as apoA-I and apoJ. These results help identify key determinants for LDL aggregation, fusion and coalescence into lipid droplets in vivo.
Collapse
|
29
|
Ke LY, Chan HC, Chen CC, Lu J, Marathe GK, Chu CS, Chan HC, Wang CY, Tung YC, McIntyre TM, Yen JH, Chen CH. Enhanced Sphingomyelinase Activity Contributes to the Apoptotic Capacity of Electronegative Low-Density Lipoprotein. J Med Chem 2016; 59:1032-40. [DOI: 10.1021/acs.jmedchem.5b01534] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Liang-Yin Ke
- Vascular
and Medicinal Research, Texas Heart Institute, Houston, Texas 77030, United States
| | - Hua-Chen Chan
- Vascular
and Medicinal Research, Texas Heart Institute, Houston, Texas 77030, United States
| | - Chih-Chieh Chen
- Institute
of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung, Taiwan 80424
| | - Jonathan Lu
- Vascular
and Medicinal Research, Texas Heart Institute, Houston, Texas 77030, United States
| | - Gopal K. Marathe
- Departments of Cellular & Molecular Medicine, Lerner Research Institute, Cleveland Clinic Lerner College of Medicine, Cleveland, Ohio 44195, United States
- Department
of Studies in Biochemistry, Manasagangothri, University of Mysore, Mysore-570006, India
| | | | | | | | | | - Thomas M. McIntyre
- Departments of Cellular & Molecular Medicine, Lerner Research Institute, Cleveland Clinic Lerner College of Medicine, Cleveland, Ohio 44195, United States
| | | | - Chu-Huang Chen
- Vascular
and Medicinal Research, Texas Heart Institute, Houston, Texas 77030, United States
- New York Heart Research
Foundation, Mineola, New York 11501, United States
| |
Collapse
|
30
|
Estruch M, Rajamäki K, Sanchez-Quesada JL, Kovanen PT, Öörni K, Benitez S, Ordoñez-Llanos J. Electronegative LDL induces priming and inflammasome activation leading to IL-1β release in human monocytes and macrophages. Biochim Biophys Acta Mol Cell Biol Lipids 2015; 1851:1442-9. [PMID: 26327597 DOI: 10.1016/j.bbalip.2015.08.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Revised: 08/05/2015] [Accepted: 08/26/2015] [Indexed: 10/23/2022]
Abstract
BACKGROUND Electronegative LDL (LDL(−)), a modified LDL fraction found in blood, induces the release of inflammatory mediators in endothelial cells and leukocytes. However, the inflammatory pathways activated by LDL(−) have not been fully defined. We aim to study whether LDL(−) induced release of the first-wave proinflammatory IL-1β in monocytes and monocyte-derived macrophages (MDM) and the mechanisms involved. METHODS LDL(−) was isolated from total LDL by anion exchange chromatography. Monocytes and MDM were isolated from healthy donors and stimulated with LDL(+) and LDL(−) (100 mg apoB/L). RESULTS In monocytes, LDL(−) promoted IL-1β release in a time-dependent manner, obtaining at 20 h-incubation the double of IL-1β release induced by LDL(−) than by native LDL. LDL(−)-induced IL-1β release involved activation of the CD14-TLR4 receptor complex. LDL(−) induced priming, the first step of IL-1β release, since it increased the transcription of pro-IL-1β (8-fold) and NLRP3 (3-fold) compared to native LDL. Several findings show that LDL(−) induced inflammasome activation, the second step necessary for IL-1β release. Preincubation of monocytes with K+ channel inhibitors decreased LDL(−)-induced IL-1β release. LDL(−) induced formation of the NLRP3-ASC complex. LDL(−) triggered 2-fold caspase-1 activation compared to native LDL and IL-1β release was strongly diminished in the presence of the caspase-1 inhibitor Z-YVAD. In MDM, LDL(−) promoted IL-1β release, which was also associated with caspase-1 activation. CONCLUSIONS LDL(−) promotes release of biologically active IL-1β in monocytes and MDM by induction of the two steps involved: priming and NLRP3 inflammasome activation. SIGNIFICANCE By IL-1β release, LDL(−) could regulate inflammation in atherosclerosis.
Collapse
Affiliation(s)
- M Estruch
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), Barcelona (Spain). C/Sant Antoni M. Claret, 167 08025 Barcelona, Spain.
| | - K Rajamäki
- Wihuri Research Institute (WRI). Haartmaninkatu, 8 FI-00290 Helsinki, Finland.
| | - J L 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.
| | - P T Kovanen
- Wihuri Research Institute (WRI). Haartmaninkatu, 8 FI-00290 Helsinki, Finland.
| | - K Öörni
- Wihuri Research Institute (WRI). Haartmaninkatu, 8 FI-00290 Helsinki, Finland.
| | - S 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.
| | - J 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.
| |
Collapse
|
31
|
Ivanova EA, Bobryshev YV, Orekhov AN. LDL electronegativity index: a potential novel index for predicting cardiovascular disease. Vasc Health Risk Manag 2015; 11:525-32. [PMID: 26357481 PMCID: PMC4559248 DOI: 10.2147/vhrm.s74697] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
High cardiovascular risk conditions are frequently associated with altered plasma lipoprotein profile, such as elevated low-density lipoprotein (LDL) and LDL cholesterol and decreased high-density lipoprotein. There is, however, accumulating evidence that specific subclasses of LDL may play an important role in cardiovascular disease development, and their relative concentration can be regarded as a more relevant risk factor. LDL particles undergo multiple modifications in plasma that can lead to the increase of their negative charge. The resulting electronegative LDL [LDL(–)] subfraction has been demonstrated to be especially atherogenic, and became a subject of numerous recent studies. In this review, we discuss the physicochemical properties of LDL(–), methods of its detection, atherogenic activity, and relevance of the LDL electronegativity index as a potential independent predictor of cardiovascular risk.
Collapse
Affiliation(s)
- Ekaterina A Ivanova
- Department of Pediatric Nephrology and Growth and Regeneration, Katholieke Universiteit Leuven and University Hospitals Leuven, Leuven, Belgium
| | - Yuri V Bobryshev
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Sciences, Moscow, Russia ; Faculty of Medicine, School of Medical Sciences, University of New South Wales, Kensington, Sydney, NSW, Australia
| | - Alexander N Orekhov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Sciences, Moscow, Russia ; Institute for Atherosclerosis Research, Skolkovo Innovative Center, Moscow, Russia ; Department of Biophysics, Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
32
|
Revuelta-López E, Cal R, Julve J, Rull A, Martínez-Bujidos M, Perez-Cuellar M, Ordoñez-Llanos J, Badimon L, Sanchez-Quesada JL, Llorente-Cortés V. Hypoxia worsens the impact of intracellular triglyceride accumulation promoted by electronegative low-density lipoprotein in cardiomyocytes by impairing perilipin 5 upregulation. Int J Biochem Cell Biol 2015; 65:257-67. [DOI: 10.1016/j.biocel.2015.06.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Revised: 06/03/2015] [Accepted: 06/12/2015] [Indexed: 10/23/2022]
|
33
|
Lu M, Gursky O. Aggregation and fusion of low-density lipoproteins in vivo and in vitro. Biomol Concepts 2015; 4:501-18. [PMID: 25197325 DOI: 10.1515/bmc-2013-0016] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Low-density lipoproteins (LDLs, also known as 'bad cholesterol') are the major carriers of circulating cholesterol and the main causative risk factor of atherosclerosis. Plasma LDLs are 20- to 25-nm nanoparticles containing a core of cholesterol esters surrounded by a phospholipid monolayer and a single copy of apolipoprotein B (550 kDa). An early sign of atherosclerosis is the accumulation of LDL-derived lipid droplets in the arterial wall. According to the widely accepted 'response-to-retention hypothesis', LDL binding to the extracellular matrix proteoglycans in the arterial intima induces hydrolytic and oxidative modifications that promote LDL aggregation and fusion. This enhances LDL uptake by the arterial macrophages and triggers a cascade of pathogenic responses that culminate in the development of atherosclerotic lesions. Hence, LDL aggregation, fusion, and lipid droplet formation are important early steps in atherogenesis. In vitro, a variety of enzymatic and nonenzymatic modifications of LDL can induce these reactions and thereby provide useful models for their detailed analysis. Here, we summarize current knowledge of the in vivo and in vitro modifications of LDLs leading to their aggregation, fusion, and lipid droplet formation; outline the techniques used to study these reactions; and propose a molecular mechanism that underlies these pro-atherogenic processes. Such knowledge is essential in identifying endogenous and exogenous factors that can promote or prevent LDL aggregation and fusion in vivo and to help establish new potential therapeutic targets to decelerate or even block these pathogenic reactions.
Collapse
Affiliation(s)
- Mengxiao Lu
- Department of Physiology and Biophysics, Boston University School of Medicine, W321, 700 Albany Street, Boston, MA 02118, USA.
| | | |
Collapse
|
34
|
|
35
|
Abstract
PURPOSE OF REVIEW Decreased size and increased density of LDL have been associated with increased coronary heart disease (CHD) risk. Elevated plasma concentrations of small dense LDL (sdLDL) correlate with high plasma triglycerides and low HDL cholesterol levels. This review highlights recent findings about the metabolism and composition of LDL subfractions. RECENT FINDINGS The development of an automated assay has recently made possible the assessment of the CHD risk associated with sdLDL in large clinical trials and has demonstrated convincingly that sdLDL cholesterol levels are a more significant independent determinant of CHD risk than total LDL cholesterol. Metabolic studies have revealed that sdLDL particles originate through the delipidation of larger atherogenic VLDL and large LDL and from direct de novo production by the liver. Proteins associated with LDL, in addition to apolipoprotein (apo) B, include the C apolipoproteins, apoA-I, apoA-IV, apoD, apoE, apoF, apoH, apoJ, apoL-1, apoM, α-1 antitrypsin, migration inhibitory factor-related protein 8, lysosome C, prenylcysteine oxidase 1, paraoxonase 1, transthyretin, serum amyloid A4, and fibrinogen α chain. The role of the increasing number of LDL-associated proteins remains unclear; however, the data do indicate that LDL particles not only transport lipids but also carry proteins involved in inflammation and thrombosis. The sdLDL proteome in diabetic individuals differs significantly from that of larger LDL, being enriched in apoC-III. SUMMARY Progress in our understanding of the composition and metabolism of LDL subfractions strengthens the association between sdLDL and CHD risk.
Collapse
Affiliation(s)
- Margaret R Diffenderfer
- Cardiovascular Nutrition Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, Massachusetts, USA
| | | |
Collapse
|
36
|
Electronegative LDL: a circulating modified LDL with a role in inflammation. Mediators Inflamm 2013; 2013:181324. [PMID: 24062611 PMCID: PMC3766570 DOI: 10.1155/2013/181324] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Accepted: 07/19/2013] [Indexed: 12/13/2022] Open
Abstract
Electronegative low density lipoprotein (LDL(−)) is a minor modified fraction of LDL found in blood. It comprises a heterogeneous population of LDL particles modified by various mechanisms sharing as a common feature increased electronegativity. Modification by oxidation is one of these mechanisms. LDL(−) has inflammatory properties similar to those of oxidized LDL (oxLDL), such as inflammatory cytokine release in leukocytes and endothelial cells. However, in contrast with oxLDL, LDL(−) also has some anti-inflammatory effects on cultured cells. The inflammatory and anti-inflammatory properties ascribed to LDL(−) suggest that it could have a dual biological effect.
Collapse
|
37
|
The Induction of Cytokine Release in Monocytes by Electronegative Low-Density Lipoprotein (LDL) Is Related to Its Higher Ceramide Content than Native LDL. Int J Mol Sci 2013; 14:2601-16. [PMID: 23358250 PMCID: PMC3588005 DOI: 10.3390/ijms14022601] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Revised: 01/05/2013] [Accepted: 01/16/2013] [Indexed: 12/21/2022] Open
Abstract
Electronegative low-density lipoprotein (LDL(−)) is a minor modified LDL subfraction that is present in blood. LDL(−) promotes inflammation and is associated with the development of atherosclerosis. We previously reported that the increase of cytokine release promoted by this lipoprotein subfraction in monocytes is counteracted by high-density lipoprotein (HDL). HDL also inhibits a phospholipase C-like activity (PLC-like) intrinsic to LDL(−). The aim of this work was to assess whether the inhibition of the PLC-like activity by HDL could decrease the content of ceramide (CER) and diacylglycerol (DAG) generated in LDL(−). This knowledge would allow us to establish a relationship between these compounds and the inflammatory activity of LDL(−). LDL(−) incubated at 37 °C for 20 h increased its PLC-like activity and, subsequently, the amount of CER and DAG. We found that incubating LDL(−) with HDL decreased both products in LDL(−). Native LDL was modified by lipolysis with PLC or by incubation with CER-enriched or DAG-enriched liposomes. The increase of CER in native LDL significantly increased cytokine release, whereas the enrichment in DAG did not show these inflammatory properties. These data point to CER, a resultant product of the PLC-like activity, as a major determinant of the inflammatory activity induced by LDL(−) in monocytes.
Collapse
|