1
|
Liu A, Liu X, Wei Y, Xiang X, Chen Y, Zheng Z, Xu C, Yang S, Zhao K. Novel Insights into Causal Effects of Serum Lipids and Apolipoproteins on Cardiovascular Morpho-Functional Phenotypes. Cardiovasc Toxicol 2024:10.1007/s12012-024-09930-w. [PMID: 39394502 DOI: 10.1007/s12012-024-09930-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Accepted: 10/02/2024] [Indexed: 10/13/2024]
Abstract
Previous observational studies have explored the association between serum lipids, apolipoproteins, and adverse ventricular/aortic structure and function. However, whether a causal link exists is uncertain. This study employed a two-sample Mendelian randomization (MR), colocalization, reverse, and multivariable MR (MVMR) approach to examine the causal associations among five serum lipids, two apolipoproteins, and 32 cardiac magnetic resonance (CMR) traits. Utilizing single-nucleotide polymorphisms (SNPs) linked to serum lipids and apolipoproteins as instrumental variables. CMR traits from seven independent genome-wide association studies served as preclinical endophenotypes, offering insights into aortic and cardiac structure/function. The primary analysis utilized a random-effects inverse variance method (IVW), followed by sensitivity and validation analyses. In the primary IVW MR analyses, genetically predicted low-density lipoprotein cholesterol (LDL-C) levels were positively correlated with increased descending aorta strain (DAo strain) (β = 0.098; P = 2.69E-07) and ascending aorta strain (AAo strain) (β = 0.079; P = 5.19E-05). Genetically predicted high-density lipoprotein cholesterol (HDL-C) levels were positively correlated with left ventricular radial peak diastolic strain rate (LV-PDSRll) (β = 0.176; P = 2.89E-05) and the left ventricular longitudinal peak diastolic strain rate (LV-PDSRrr) (β = 0.059; P = 2.44E-06), and negatively correlated with left ventricular regional wall thickness (LVRWT). While apolipoprotein B (ApoB) levels were positively correlated with AAo strain (β = 0.076; P = 1.16E-05), DAo strain (β = 0.065; P = 2.77E-05). A shared causal variant was identified to demonstrate the associations of ApoB with AAo strain and DAo strain using colocalization analysis. Sensitivity analyses confirmed the robustness of these associations. Targeting lipid and apolipoprotein levels through interventions may provide novel strategies for the primary prevention of CVDs.
Collapse
Affiliation(s)
- Ankang Liu
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital South Campus, Shanghai, China
| | - Xiaohong Liu
- Department of Radiology, Shanghai Eighth People's Hospital, No. 8. Caobao Road, Xuhui District, Shanghai, 200235, China
| | - Yuanhao Wei
- School of Public Health, Harbin Medical University, Harbin, China
| | - Xiqiao Xiang
- Department of PET-CT Imaging Center, Shanghai Jiao Tong University Affiliated Sixth People's Hospital South Campus, Shanghai, China
| | - Yi Chen
- Department of PET-CT Imaging Center, Shanghai Jiao Tong University Affiliated Sixth People's Hospital South Campus, Shanghai, China
| | - Ziwei Zheng
- Department of Ultrasound Medicine, Shanghai Eighth People's Hospital, No. 8. Caobao Road, Xuhui District, Shanghai, 200235, China
| | - Changde Xu
- Department of PET-CT Imaging Center, Shanghai Jiao Tong University Affiliated Sixth People's Hospital South Campus, Shanghai, China
| | - Shaoling Yang
- Department of Ultrasound Medicine, Shanghai Eighth People's Hospital, No. 8. Caobao Road, Xuhui District, Shanghai, 200235, China.
| | - Kun Zhao
- Department of PET-CT Imaging Center, Shanghai Jiao Tong University Affiliated Sixth People's Hospital South Campus, Shanghai, China.
| |
Collapse
|
2
|
Wang J, Wu Q, Wang X, Liu H, Chen M, Xu L, Zhang Z, Li K, Li W, Zhong J. Targeting Macrophage Phenotypes and Metabolism as Novel Therapeutic Approaches in Atherosclerosis and Related Cardiovascular Diseases. Curr Atheroscler Rep 2024; 26:573-588. [PMID: 39133247 PMCID: PMC11392985 DOI: 10.1007/s11883-024-01229-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/22/2024] [Indexed: 08/13/2024]
Abstract
PURPOSE OF THE REVIEW Macrophage accumulation and activation function as hallmarks of atherosclerosis and have complex and intricate dynamics throughout all components and stages of atherosclerotic plaques. In this review, we focus on the regulatory roles and underlying mechanisms of macrophage phenotypes and metabolism in atherosclerosis. We highlight the diverse range of macrophage phenotypes present in atherosclerosis and their potential roles in progression and regression of atherosclerotic plaque. Furthermore, we discuss the challenges and opportunities in developing therapeutic strategies for preventing and treating atherosclerotic cardiovascular disease. RECENT FINDINGS Dysregulation of macrophage polarization between the proinflammatory M1 and anti-inflammatory M2 phenotypealters the immuno-inflammatory response during atherosclerosis progression, leading to plaque initiation, growth, and ultimately rupture. Altered metabolism of macrophage is a key feature for their function and the subsequent progression of atherosclerotic cardiovascular disease. The immunometabolism of macrophage has been implicated to macrophage activation and metabolic rewiring of macrophages within atherosclerotic lesions, thereby shifting altered macrophage immune-effector and tissue-reparative function. Targeting macrophage phenotypes and metabolism are potential therapeutic strategies in the prevention and treatment of atherosclerosis and atherosclerotic cardiovascular diseases. Understanding the precise function and metabolism of specific macrophage subsets and their contributions to the composition and growth of atherosclerotic plaques could reveal novel strategies to delay or halt development of atherosclerotic cardiovascular diseases and their associated pathophysiological consequences. Identifying biological stimuli capable of modulating macrophage phenotypes and metabolism may lead to the development of innovative therapeutic approaches for treating patients with atherosclerosis and coronary artery diseases.
Collapse
Affiliation(s)
- Juan Wang
- Beijing Key Laboratory of Hypertension, Heart Center of Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China.
| | - Qiang Wu
- Senior Department of Cardiology, the Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
- Journal of Geriatric Cardiology Editorial Office, Chinese PLA General Hospital, Beijing, China
| | - Xinyu Wang
- Beijing Key Laboratory of Hypertension, Heart Center of Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Hongbin Liu
- Department of Cardiology, the Second Medical Center, National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| | - Mulei Chen
- Beijing Key Laboratory of Hypertension, Heart Center of Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Li Xu
- Beijing Key Laboratory of Hypertension, Heart Center of Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Ze Zhang
- National Institute of Biological Sciences, Beijing, China
| | - Kuibao Li
- Beijing Key Laboratory of Hypertension, Heart Center of Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Weiming Li
- Beijing Key Laboratory of Hypertension, Heart Center of Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China.
| | - Jiuchang Zhong
- Beijing Key Laboratory of Hypertension, Heart Center of Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
3
|
Kumar M, Ali W, Yadav K, Kaumri S, Mishra S, Nardi P, Iellamo F, Bernardini S, Pradhan A, Perrone MA. High-Density Lipoprotein-Associated Paraoxonase-1 (PON-1) and Scavenger Receptor Class B Type 1 (SRB-1) in Coronary Artery Disease: Correlation with Disease Severity. J Clin Med 2024; 13:5480. [PMID: 39336967 PMCID: PMC11432482 DOI: 10.3390/jcm13185480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 09/04/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024] Open
Abstract
Background: Coronary artery disease (CAD) is the leading cause of death worldwide. High-Density lipoprotein (HDL) is a well-established marker associated with CAD. The current research goes beyond the conventional HDL-C measurement in previous studies and dives into the functional intricacies of HDL. By understanding how HDL works, rather than just how much of it exists, we can better tailor diagnostic and therapeutic strategies for CAD and related conditions. Hence, the current study quantifies the serum levels of two novel HDL-associated markers, Paraoxonase-1 (PON-1) and Scavenger Receptor Class B Type 1 (SRB-1), in CAD cases vs. controls. Methods: A total of 92 subjects, including 69 CAD and 23 healthy controls, were included, based on the prevalence of the disease. Further, based on the severity of the disease, CAD cases were subcategorized as CAD-I, -II, and -III. Serum PON-1 and SRB-1 levels were measured and compared between patient and control groups. Results: The levels of PON-1 and SRB-1 (32.6 ng/mL and 12.49 ng/mL) were significantly lower in CAD patients vs. the healthy control, at 60.36 ng/mL and 15.85 ng/mL, respectively (p < 0.000). A further intergroup comparison showed a statistically significant difference between the CAT-I and -III for PON-1 (p < 0.025), the CAT-I and -III, and CAT-II and -III for SRB-1 (p < 0.000). The receiver operating characteristics (ROC) curve showed cutoff values of 48.20 ng/mL and 14.90 ng/mL for PON-1 and SRB-1. Conclusions: The current study found that serum levels of HDL-associated PON-1 and SRB-1 are significantly lower in CAD cases, and were also inversely related to the increasing severity of coronary artery disease. This inference implies that serum PON-1 and SRB-1 could be used as non-invasive tools for the identification of coronary atherosclerosis and risk assessment in CAD cases.
Collapse
Affiliation(s)
- Manish Kumar
- Department of Pathology, King George Medical University, Lucknow 226003, Uttar Pradesh, India; (M.K.); (S.K.)
| | - Wahid Ali
- Department of Pathology, King George Medical University, Lucknow 226003, Uttar Pradesh, India; (M.K.); (S.K.)
| | - Kusum Yadav
- Department of Pathology, Dr. Ram Manohar Lohia Institute of Medical Sciences, Lucknow 226003, Uttar Pradesh, India; (K.Y.); (S.M.)
| | - Swati Kaumri
- Department of Pathology, King George Medical University, Lucknow 226003, Uttar Pradesh, India; (M.K.); (S.K.)
| | - Sridhar Mishra
- Department of Pathology, Dr. Ram Manohar Lohia Institute of Medical Sciences, Lucknow 226003, Uttar Pradesh, India; (K.Y.); (S.M.)
| | - Paolo Nardi
- Department of Surgical Sciences, University of Rome Tor Vergata, 00133 Rome, Italy;
| | - Ferdinando Iellamo
- Division of Cardiology and CardioLab, Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (F.I.); (M.A.P.)
| | - Sergio Bernardini
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy;
| | - Akshyaya Pradhan
- Lari Cardiology Center, King George Medical University, Lucknow 226003, Uttar Pradesh, India;
| | - Marco Alfonso Perrone
- Division of Cardiology and CardioLab, Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (F.I.); (M.A.P.)
| |
Collapse
|
4
|
Qiu X, Feng Y. Echinacoside activates Nrf2/PPARγ signaling pathway to modulate mitochondrial fusion-fission balance to ameliorate ox-LDL-induced dysfunction of coronary artery endothelial cells. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024:10.1007/s00210-024-03233-1. [PMID: 38916831 DOI: 10.1007/s00210-024-03233-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 06/08/2024] [Indexed: 06/26/2024]
Abstract
As a cardiovascular disease, coronary heart disease (CHD) is characterized by poor prognosis and increasing morbidity and mortality rates. Echinacoside (ECH) can protect against multiple cardiovascular diseases due to its antioxidant and anti-inflammatory properties. However, the role of ECH in CHD remains unclear. In ECH-treated human coronary artery endothelial cells (HCAECs), cell viability, NO production, endothelial nitric oxide synthase (eNOS) expression, and angiogenesis ability were detected using cell counting kit-8 (CCK-8) assay, diaminofluorescein-FM diacetate (DAF-FM DA) staining, western blot, and tube formation assay, respectively. The activities of oxidative stress markers were detected using dichloro-dihydro-fluorescein diacetate (DCFH-DA) assay and corresponding assay kits. Cell apoptosis was detected utilizing flow cytometry and caspase3 assay. Western blot was used to detect the expressions of Nrf2/PPARγ signaling pathway- and mitochondrial dynamics-related proteins. Mitochondrial membrane potential and mitochondrial fusion and fission were detected using JC-1 staining and immunofluorescence (IF) assay. In this study, ECH was found to revive the viability, ameliorate the endothelial dysfunction, suppress oxidative stress, and inhibit the apoptosis in ox-LDL-induced HCAECs via activating Nrf2/PPARγ signaling pathway, which were all abolished following the treatment of Nrf2 inhibitor ML385. It was also identified that ECH regulated mitochondrial fusion-fission balance in ox-LDL-induced HCAECs through the activation of Nrf2/PPARγ signaling pathway. In summary, ECH activated Nrf2/PPARγ signaling pathway to regulate mitochondrial fusion-fission balance, thereby improving ox-LDL-induced dysfunction of HCAECs.
Collapse
Affiliation(s)
- Xiandi Qiu
- Department of Cardiovascular Medicine, The Ninth People's Hospital of Chongqing, Chongqing, China
| | - Yuxing Feng
- Department of Neurology, The Ninth People's Hospital of Chongqing, No. 69 Jialing Village, Beibei District, Chongqing, 400700, China.
| |
Collapse
|
5
|
Frey K, Rohrer L, Frommelt F, Ringwald M, Potapenko A, Goetze S, von Eckardstein A, Wollscheid B. Mapping the dynamic high-density lipoprotein synapse. Atherosclerosis 2023; 380:117200. [PMID: 37619408 DOI: 10.1016/j.atherosclerosis.2023.117200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 07/27/2023] [Accepted: 07/28/2023] [Indexed: 08/26/2023]
Abstract
BACKGROUND AND AIMS Heterogeneous high-density lipoprotein (HDL) particles, which can contain hundreds of proteins, affect human health and disease through dynamic molecular interactions with cell surface proteins. How HDL mediates its long-range signaling functions and interactions with various cell types is largely unknown. Due to the complexity of HDL, we hypothesize that multiple receptors engage with HDL particles resulting in condition-dependent receptor-HDL interaction clusters at the cell surface. METHODS Here we used the mass spectrometry-based and light-controlled proximity labeling strategy LUX-MS in a discovery-driven manner to decode HDL-receptor interactions. RESULTS Surfaceome nanoscale organization analysis of hepatocytes and endothelial cells using LUX-MS revealed that the previously known HDL-binding protein scavenger receptor B1 (SCRB1) is embedded in a cell surface protein community, which we term HDL synapse. Modulating the endothelial HDL synapse, composed of 60 proteins, by silencing individual members, showed that the HDL synapse can be assembled in the absence of SCRB1 and that the members are interlinked. The aminopeptidase N (AMPN) (also known as CD13) was identified as an HDL synapse member that directly influences HDL uptake into the primary human aortic endothelial cells (HAECs). CONCLUSIONS Our data indicate that preformed cell surface residing protein complexes modulate HDL function and suggest new theragnostic opportunities.
Collapse
Affiliation(s)
- Kathrin Frey
- Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland; Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland; Institute for Clinical Chemistry University Hospital Zurich, Zurich, Switzerland.
| | - Lucia Rohrer
- Institute for Clinical Chemistry University Hospital Zurich, Zurich, Switzerland
| | - Fabian Frommelt
- Institute of Molecular Systems Biology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Meret Ringwald
- Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Anton Potapenko
- Institute for Clinical Chemistry University Hospital Zurich, Zurich, Switzerland
| | - Sandra Goetze
- Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland; Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland; ETH PHRT Swiss Multi-Omics Center (SMOC), Switzerland
| | | | - Bernd Wollscheid
- Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland; Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland; ETH PHRT Swiss Multi-Omics Center (SMOC), Switzerland.
| |
Collapse
|
6
|
Abstract
Epidemiologic studies detected an inverse relationship between HDL (high-density lipoprotein) cholesterol (HDL-C) levels and atherosclerotic cardiovascular disease (ASCVD), identifying HDL-C as a major risk factor for ASCVD and suggesting atheroprotective functions of HDL. However, the role of HDL-C as a mediator of risk for ASCVD has been called into question by the failure of HDL-C-raising drugs to reduce cardiovascular events in clinical trials. Progress in understanding the heterogeneous nature of HDL particles in terms of their protein, lipid, and small RNA composition has contributed to the realization that HDL-C levels do not necessarily reflect HDL function. The most examined atheroprotective function of HDL is reverse cholesterol transport, whereby HDL removes cholesterol from plaque macrophage foam cells and delivers it to the liver for processing and excretion into bile. Indeed, in several studies, HDL has shown inverse associations between HDL cholesterol efflux capacity and ASCVD in humans. Inflammation plays a key role in the pathogenesis of atherosclerosis and vulnerable plaque formation, and a fundamental function of HDL is suppression of inflammatory signaling in macrophages and other cells. Oxidation is also a critical process to ASCVD in promoting atherogenic oxidative modifications of LDL (low-density lipoprotein) and cellular inflammation. HDL and its proteins including apoAI (apolipoprotein AI) and PON1 (paraoxonase 1) prevent cellular oxidative stress and LDL modifications. Importantly, HDL in humans with ASCVD is oxidatively modified rendering HDL dysfunctional and proinflammatory. Modification of HDL with reactive carbonyl species, such as malondialdehyde and isolevuglandins, dramatically impairs the antiatherogenic functions of HDL. Importantly, treatment of murine models of atherosclerosis with scavengers of reactive dicarbonyls improves HDL function and reduces systemic inflammation, atherosclerosis development, and features of plaque instability. Here, we discuss the HDL antiatherogenic functions in relation to oxidative modifications and the potential of reactive dicarbonyl scavengers as a therapeutic approach for ASCVD.
Collapse
Affiliation(s)
- MacRae F. Linton
- 1. Department of Medicine, Division of Cardiovascular Medicine, Atherosclerosis Research Unit, Vanderbilt University School of Medicine, Nashville, TN 37232
- 2. Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Patricia G. Yancey
- 1. Department of Medicine, Division of Cardiovascular Medicine, Atherosclerosis Research Unit, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Huan Tao
- 1. Department of Medicine, Division of Cardiovascular Medicine, Atherosclerosis Research Unit, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Sean S. Davies
- 2. Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232
| |
Collapse
|
7
|
Peña-de-la-Sancha P, Muñoz-García A, Espínola-Zavaleta N, Bautista-Pérez R, Mejía AM, Luna-Luna M, López-Olmos V, Rodríguez-Pérez JM, Fragoso JM, Carreón-Torres E, Pérez-Méndez Ó. Eicosapentaenoic and Docosahexaenoic Acid Supplementation Increases HDL Content in n-3 Fatty Acids and Improves Endothelial Function in Hypertriglyceridemic Patients. Int J Mol Sci 2023; 24:5390. [PMID: 36982461 PMCID: PMC10049536 DOI: 10.3390/ijms24065390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 03/08/2023] [Accepted: 03/09/2023] [Indexed: 03/18/2023] Open
Abstract
High-density lipoproteins (HDLs) are known to enhance vascular function through different mechanisms, including the delivery of functional lipids to endothelial cells. Therefore, we hypothesized that omega-3 (n-3) eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) content of HDLs would improve the beneficial vascular effects of these lipoproteins. To explore this hypothesis, we performed a placebo-controlled crossover clinical trial in 18 hypertriglyceridemic patients without clinical symptoms of coronary heart disease who received highly purified EPA 460 mg and DHA 380 mg, twice a day for 5 weeks or placebo. After 5 weeks of treatment, patients followed a 4-week washout period before crossover. HDLs were isolated using sequential ultracentrifugation for characterization and determination of fatty acid content. Our results showed that n-3 supplementation induced a significant decrease in body mass index, waist circumference as well as triglycerides and HDL-triglyceride plasma concentrations, whilst HDL-cholesterol and HDL-phospholipids significantly increased. On the other hand, HDL, EPA, and DHA content increased by 131% and 62%, respectively, whereas 3 omega-6 fatty acids significantly decreased in HDL structures. In addition, the EPA-to-arachidonic acid (AA) ratio increased more than twice within HDLs suggesting an improvement in their anti-inflammatory properties. All HDL-fatty acid modifications did not affect the size distribution or the stability of these lipoproteins and were concomitant with a significant increase in endothelial function assessed using a flow-mediated dilatation test (FMD) after n-3 supplementation. However, endothelial function was not improved in vitro using a model of rat aortic rings co-incubated with HDLs before or after treatment with n-3. These results suggest a beneficial effect of n-3 on endothelial function through a mechanism independent of HDL composition. In conclusion, we demonstrated that EPA and DHA supplementation for 5 weeks improved vascular function in hypertriglyceridemic patients, and induced enrichment of HDLs with EPA and DHA to the detriment of some n-6 fatty acids. The significant increase in the EPA-to-AA ratio in HDLs is indicative of a more anti-inflammatory profile of these lipoproteins.
Collapse
Affiliation(s)
- Paola Peña-de-la-Sancha
- Department of Molecular Biology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico
| | - Adolfo Muñoz-García
- Department of Molecular Biology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico
| | - Nilda Espínola-Zavaleta
- Department of Nuclear Medicine, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico
- Department of Echocardiography, ABC Medical Center, I.A.P, Mexico City 01120, Mexico
| | - Rocío Bautista-Pérez
- Department of Molecular Biology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico
| | - Ana María Mejía
- Blood Bank, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico
| | - María Luna-Luna
- Department of Molecular Biology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico
| | - Victoria López-Olmos
- Department of Molecular Biology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico
| | - José-Manuel Rodríguez-Pérez
- Department of Molecular Biology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico
| | - José-Manuel Fragoso
- Department of Molecular Biology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico
| | - Elizabeth Carreón-Torres
- Department of Molecular Biology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico
| | - Óscar Pérez-Méndez
- Department of Molecular Biology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico
- Tecnologico de Monterrey, School of Engineering and Sciences, Mexico City 14380, Mexico
| |
Collapse
|
8
|
Song B, Dang H, Dong R. Differential Expression of LOXL1-AS1 in Coronary Heart Disease and its Regulatory Mechanism in ox-LDL-Induced Human Coronary Artery Endothelial Cell Pyroptosis. Cardiovasc Drugs Ther 2023; 37:75-87. [PMID: 34633594 DOI: 10.1007/s10557-021-07274-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/29/2021] [Indexed: 01/14/2023]
Abstract
OBJECTIVE Coronary heart disease (CHD) is a notable contributor to the burden of human health. Dysregulated long non-coding RNAs (lncRNAs) are implicated in the pathogenesis of CHD. This study investigated the expression pattern of lncRNA LOXL1-AS1 in CHD and its regulatory mechanism in oxidized low-density lipoprotein (ox-LDL)-induced human coronary artery endothelial cell (HCAEC) pyroptosis. METHODS Serum was collected from 62 CHD patients and 62 healthy volunteers for the detection of LOXL1-AS1 expression. The value of LOXL1-AS1 in CHD diagnosis and major cardiovascular adverse event (MACE) prediction was analyzed using the ROC curve. LOXL1-AS1, miR-16-5p, and SNX16 expressions in ox-LDL-treated HCAECs were determined using RT-qPCR. NLPR3, cleaved-caspase-1, and GSDMD-N protein levels were measured using Western blot. IL-1β and IL-18 concentrations were measured using ELISA. The binding relationships between LOXL1-AS1 and miR-16-5p and miR-16-5p and SNX16 were verified. Functional rescue experiment was performed to verify the role of miR-16-5p in HCAEC pyroptosis. RESULTS LOXL1-AS1 was highly expressed in CHD patients. LOXL1-AS1 had diagnostic value for CHD and predictive value for MACE occurrence. ox-LDL-treated HCAECs showed reduced viability, increased IL-1β and IL-18 concentrations, and elevated NLPR3, cleaved-caspase-1, and GSDMD-N levels. LOXL1-AS1 silencing promoted cell viability and reduced pyroptosis. LOXL1-AS1 bound to miR-16-5p and miR-16-5p targeted SNX16. Inhibition of miR-16-5p reversed the inhibitory effect of LOXL1-AS1 silencing on HCAEC pyroptosis. CONCLUSION LOXL1-AS1 was elevated in CHD patients with a good diagnostic value for CHD and predictive value for MACE. LOXL1-AS1 downregulated miR-16-5p expression by binding to miR-16-5p to enhance ox-LDL-induced HCAEC pyroptosis, which may be associated with upregulation of SNX16 transcription.
Collapse
Affiliation(s)
- Bangrong Song
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Anzhen Road, Chaoyang, Beijing, 100029, People's Republic of China
| | - Haiming Dang
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Anzhen Road, Chaoyang, Beijing, 100029, People's Republic of China
| | - Ran Dong
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Anzhen Road, Chaoyang, Beijing, 100029, People's Republic of China.
| |
Collapse
|
9
|
Klobučar I, Stadler JT, Klobučar L, Lechleitner M, Trbušić M, Pregartner G, Berghold A, Habisch H, Madl T, Marsche G, Frank S, Degoricija V. Associations between Endothelial Lipase, High-Density Lipoprotein, and Endothelial Function Differ in Healthy Volunteers and Metabolic Syndrome Patients. Int J Mol Sci 2023; 24:2073. [PMID: 36768410 PMCID: PMC9916974 DOI: 10.3390/ijms24032073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/13/2023] [Accepted: 01/17/2023] [Indexed: 01/21/2023] Open
Abstract
Metabolic syndrome (MS) is characterized by endothelial- and high-density lipoprotein (HDL) dysfunction and increased endothelial lipase (EL) serum levels. We examined the associations between EL serum levels, HDL (serum levels, lipid content, and function), and endothelial function in healthy volunteers (HV) and MS patients. Flow-mediated dilation (FMD), nitroglycerin-mediated dilation (NMD), serum levels of HDL subclasses (measured by nuclear magnetic resonance (NMR) spectroscopy), and EL serum levels differed significantly between HV and MS patients. The serum levels of triglycerides in large HDL particles were significantly positively correlated with FMD and NMD in HV, but not in MS patients. Cholesterol (C) and phospholipid (PL) contents of large HDL particles, calculated as HDL1-C/HDL1-apoA-I and HDL1-PL/HDL1-apoA-I, respectively, were significantly negatively correlated with FMD in HV, but not in MS patients. Cholesterol efflux capacity and arylesterase activity of HDL, as well as EL, were correlated with neither FMD nor NMD. EL was significantly negatively correlated with HDL-PL/HDL-apoA-I in HV, but not in MS patients, and with serum levels of small dense HDL containing apolipoprotein A-II in MS patients, but not in HV. We conclude that MS modulates the association between HDL and endothelial function, as well as between EL and HDL. HDL cholesterol efflux capacity and arylesterase activity, as well as EL serum levels, are not associated with endothelial function in HV or MS patients.
Collapse
Affiliation(s)
- Iva Klobučar
- Department of Cardiology, Sisters of Charity University Hospital Centre, 10000 Zagreb, Croatia
| | - Julia T. Stadler
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, 8010 Graz, Austria
| | - Lucija Klobučar
- Department of Medicine, University Hospital Centre Osijek, 31000 Osijek, Croatia
| | - Margarete Lechleitner
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria
| | - Matias Trbušić
- Department of Cardiology, Sisters of Charity University Hospital Centre, 10000 Zagreb, Croatia
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Gudrun Pregartner
- Institute for Medical Informatics, Statistics und Documentation, Medical University of Graz, 8036 Graz, Austria
| | - Andrea Berghold
- Institute for Medical Informatics, Statistics und Documentation, Medical University of Graz, 8036 Graz, Austria
| | - Hansjörg Habisch
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria
| | - Tobias Madl
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria
- BioTechMed-Graz, 8010 Graz, Austria
| | - Gunther Marsche
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, 8010 Graz, Austria
- BioTechMed-Graz, 8010 Graz, Austria
| | - Saša Frank
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria
- BioTechMed-Graz, 8010 Graz, Austria
| | - Vesna Degoricija
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
- Department of Medicine, Sisters of Charity University Hospital Centre, 10000 Zagreb, Croatia
| |
Collapse
|
10
|
Shishikura D, Octavia Y, Hayat U, Thondapu V, Barlis P. Atherogenesis and Inflammation. Interv Cardiol 2022. [DOI: 10.1002/9781119697367.ch1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
|
11
|
Li Y, Shan J. Study on the correlation between high density lipoprotein and lower extremities deep venous thrombosis in patients undergoing hip arthroplasty. Phlebology 2022; 37:516-521. [PMID: 35575216 DOI: 10.1177/02683555221090309] [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] [Indexed: 11/15/2022]
Abstract
OBJECTIVE To explore the relationship between high density lipoprotein (HDL) and lower extremities deep venous thrombosis (DVT) in patients undergoing hip arthroplasty. METHODS A total of 348 patients undergoing hip arthroplasty in our hospital were enrolled, and divided into observation (n = 154, 44.25%) and control (n = 194, 55.75%) groups according to the occurrence of lower extremities DVT. The presence of DVT was assessed 1 day before surgery and routinely every 2 days after surgery. The factors of DVT were analyzed by single factor analysis, multivariate logistic regression analysis, and Pearson correlation. RESULTS The age and body mass index in the observation group were significantly higher (p = .045, p = .041, respectively), while HDL-C was significantly lower (p = .032) than the control group. Increase age, high BMI, low apolipoprotein-A1 level and low HDL-C level were risk factors for lower extremities DVT. The mean HDL-C in the observation and control groups was 0.91 ± 0.27 and 1.19 ± 0.37, respectively, the adjusted odds ratio was 1.050; 95% CI 1.010-1.092, p = .014. CONCLUSION Elderly patients with high BMI and low HDL-C level undergoing hip arthroplasty are at risk of lower extremities DVT, and should be paid attention to clinically.
Collapse
Affiliation(s)
- Yong Li
- Department of Orthopaedic, 117858Dongyang People's Hospital, Dongyang, China
| | - Junbiao Shan
- Department of Orthopaedic, 117858Dongyang People's Hospital, Dongyang, China
| |
Collapse
|
12
|
Wang H, Liu H, Zhao X, Chen X. Heterogeneous nuclear ribonucleoprotein U-actin complex derived from extracellular vesicles facilitates proliferation and migration of human coronary artery endothelial cells by promoting RNA polymerase II transcription. Bioengineered 2022; 13:11469-11486. [PMID: 35535400 PMCID: PMC9276035 DOI: 10.1080/21655979.2022.2066754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Coronary artery disease (CAD) represents a fatal public threat. The involvement of extracellular vesicles (EVs) in CAD has been documented. This study explored the regulation of embryonic stem cells (ESCs)-derived EVs-hnRNPU-actin complex in human coronary artery endothelial cell (HCAEC) growth. Firstly, in vitro HCAEC hypoxia models were established. EVs were extracted from ESCs by ultracentrifugation. HCAECs were treated with EVs and si-VEGF for 24 h under hypoxia, followed by assessment of cell proliferation, apoptosis, migration, and tube formation. Uptake of EVs by HCAECs was testified. Additionally, hnRNPU, VEGF, and RNA Pol II levels were determined using Western blotting and CHIP assays. Interaction between hnRNPU and actin was evaluated by Co-immunoprecipitation assay. HCAEC viability and proliferation were lowered, apoptosis was enhanced, wound fusion was decreased, and the number of tubular capillary structures was reduced under hypoxia, whereas ESC-EVs treatment counteracted these effects. Moreover, EVs transferred hnRNPU into HCAECs. EVs-hnRNPU-actin complex increased RNA Pol II level on the VEGF gene promoter and promoted VEGF expression in HCAECs. Inhibition of hnRNPU or VEGF both annulled the promotion of EVs on HCAEC growth. Collectively, ESC-EVs-hnRNPU-actin increased RNA Pol II phosphorylation and VEGF expression, thus promoting HCAEC growth.
Collapse
Affiliation(s)
- Han Wang
- Department of Cardiovascular, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Hengdao Liu
- Department of Cardiovascular, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Xi Zhao
- Department of Cardiovascular, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Xiaowei Chen
- Department of Cardiovascular, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| |
Collapse
|
13
|
Laka K, Makgoo L, Mbita Z. Cholesterol-Lowering Phytochemicals: Targeting the Mevalonate Pathway for Anticancer Interventions. Front Genet 2022; 13:841639. [PMID: 35391801 PMCID: PMC8981032 DOI: 10.3389/fgene.2022.841639] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 03/07/2022] [Indexed: 11/15/2022] Open
Abstract
There are a plethora of cancer causes and the road to fully understanding the carcinogenesis process remains a dream that keeps changing. However, a list of role players that are implicated in the carcinogens process is getting lengthier. Cholesterol is known as bad sterol that is heavily linked with cardiovascular diseases; however, it is also comprehensively associated with carcinogenesis. There is an extensive list of strategies that have been used to lower cholesterol; nevertheless, the need to find better and effective strategies remains vastly important. The role played by cholesterol in the induction of the carcinogenesis process has attracted huge interest in recent years. Phytochemicals can be dubbed as magic tramp cards that humans could exploit for lowering cancer-causing cholesterol. Additionally, the mechanisms that are regulated by phytochemicals can be targeted for anticancer drug development. One of the key role players in cancer development and suppression, Tumour Protein 53 (TP53), is crucial in regulating the biogenesis of cholesterol and is targeted by several phytochemicals. This minireview covers the role of p53 in the mevalonate pathway and how bioactive phytochemicals target the mevalonate pathway and promote p53-dependent anticancer activities.
Collapse
Affiliation(s)
| | | | - Zukile Mbita
- Department of Biochemistry, Microbiology and Biotechnology, University of Limpopo, Sovenga, South Africa
| |
Collapse
|
14
|
Hatton SL, Pandey MK. Fat and Protein Combat Triggers Immunological Weapons of Innate and Adaptive Immune Systems to Launch Neuroinflammation in Parkinson's Disease. Int J Mol Sci 2022; 23:1089. [PMID: 35163013 PMCID: PMC8835271 DOI: 10.3390/ijms23031089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/12/2022] [Accepted: 01/14/2022] [Indexed: 01/27/2023] Open
Abstract
Parkinson's disease (PD) is the second-most common neurodegenerative disease in the world, affecting up to 10 million people. This disease mainly happens due to the loss of dopaminergic neurons accountable for memory and motor function. Partial glucocerebrosidase enzyme deficiency and the resultant excess accumulation of glycosphingolipids and alpha-synuclein (α-syn) aggregation have been linked to predominant risk factors that lead to neurodegeneration and memory and motor defects in PD, with known and unknown causes. An increasing body of evidence uncovers the role of several other lipids and their association with α-syn aggregation, which activates the innate and adaptive immune system and sparks brain inflammation in PD. Here, we review the emerging role of a number of lipids, i.e., triglyceride (TG), diglycerides (DG), glycerophosphoethanolamines (GPE), polyunsaturated fatty acids (PUFA), sphingolipids, gangliosides, glycerophospholipids (GPL), and cholesterols, and their connection with α-syn aggregation as well as the induction of innate and adaptive immune reactions that trigger neuroinflammation in PD.
Collapse
Affiliation(s)
- Shelby Loraine Hatton
- Cincinnati Children’s Hospital Medical Center, Division of Human Genetics, 3333 Burnet Avenue, Cincinnati, OH 45229, USA;
| | - Manoj Kumar Pandey
- Cincinnati Children’s Hospital Medical Center, Division of Human Genetics, 3333 Burnet Avenue, Cincinnati, OH 45229, USA;
- Department of Pediatrics, Division of Human Genetics, College of Medicine, University of Cincinnati, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| |
Collapse
|
15
|
HDL and Endothelial Function. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1377:27-47. [DOI: 10.1007/978-981-19-1592-5_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
16
|
Tracz J, Luczak M. Applying Proteomics and Integrative "Omics" Strategies to Decipher the Chronic Kidney Disease-Related Atherosclerosis. Int J Mol Sci 2021; 22:7492. [PMID: 34299112 PMCID: PMC8305100 DOI: 10.3390/ijms22147492] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/06/2021] [Accepted: 07/08/2021] [Indexed: 02/07/2023] Open
Abstract
Patients with chronic kidney disease (CKD) are at increased risk of atherosclerosis and premature mortality, mainly due to cardiovascular events. However, well-known risk factors, which promote "classical" atherosclerosis are alone insufficient to explain the high prevalence of atherosclerosis-related to CKD (CKD-A). The complexity of the molecular mechanisms underlying the acceleration of CKD-A is still to be defied. To obtain a holistic picture of these changes, comprehensive proteomic approaches have been developed including global protein profiling followed by functional bioinformatics analyses of dysregulated pathways. Furthermore, proteomics surveys in combination with other "omics" techniques, i.e., transcriptomics and metabolomics as well as physiological assays provide a solid ground for interpretation of observed phenomena in the context of disease pathology. This review discusses the comprehensive application of various "omics" approaches, with emphasis on proteomics, to tackle the molecular mechanisms underlying CKD-A progression. We summarize here the recent findings derived from global proteomic approaches and underline the potential of utilizing integrative systems biology, to gain a deeper insight into the pathogenesis of CKD-A and other disorders.
Collapse
Affiliation(s)
| | - Magdalena Luczak
- European Centre for Bioinformatics and Genomics, Institute of Bioorganic Chemistry, Polish Academy of Sciences, 61-704 Poznan, Poland;
| |
Collapse
|
17
|
Martínez-Beamonte R, Sánchez-Marco J, Lázaro G, Barco M, Herrero-Continente T, Serrano-Megías M, Botaya D, Arnal C, Barranquero C, Surra JC, Osada J, Navarro MA. Dietary Avian Proteins Are Comparable to Soybean Proteins on the Atherosclerosis Development and Fatty Liver Disease in Apoe-Deficient Mice. Nutrients 2021; 13:nu13061838. [PMID: 34072167 PMCID: PMC8227708 DOI: 10.3390/nu13061838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 05/22/2021] [Accepted: 05/24/2021] [Indexed: 12/05/2022] Open
Abstract
Background and aim: The type and amount of dietary protein has become a topic of renewed interest in light of their involvement in metabolic diseases, atherosclerosis and thrombosis. However, little attention has been devoted to the effect of avian proteins despite their wide human consumption. The aim was to investigate the influence of chicken and turkey as sources of protein compared with that of soybean on atherosclerosis and fatty liver disease. Methods and results: To this purpose, male and female Apoe-deficient were fed purified Western diets differing in their protein sources for 12 weeks. After this period, blood, liver, aortic tree and heart base samples were taken for analyses of plasma lipids and atherosclerosis. Plasma triglycerides, non-esterified fatty acids, esterified cholesterol levels and radical oxygen species in lipoproteins changed depending on the diet and sex. Females consuming the turkey protein-containing diet showed decreased atherosclerotic foci, as evidenced by the en face atherosclerosis analyses. The presence of macrophages and smooth muscle cells in plaques were not modified, and no changes were observed in hepatic lipid droplets in the studied groups either. Paraoxonase activity was higher in the group consuming turkey protein without sex differences, but only in females, it was significantly associated with aortic lesion areas. Conclusions: Compared to soybean protein, the consumption of avian proteins depending on sex resulted in similar or lower atherosclerosis development and comparable hepatic steatosis.
Collapse
Affiliation(s)
- Roberto Martínez-Beamonte
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, E-50013 Zaragoza, Spain; (R.M.-B.); (J.S.-M.); (G.L.); (M.B.); (T.H.-C.); (C.B.); (M.A.N.)
- Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, E-50013 Zaragoza, Spain; (C.A.); (J.C.S.)
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, E-28029 Madrid, Spain
| | - Javier Sánchez-Marco
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, E-50013 Zaragoza, Spain; (R.M.-B.); (J.S.-M.); (G.L.); (M.B.); (T.H.-C.); (C.B.); (M.A.N.)
- Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, E-50013 Zaragoza, Spain; (C.A.); (J.C.S.)
| | - Gonzalo Lázaro
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, E-50013 Zaragoza, Spain; (R.M.-B.); (J.S.-M.); (G.L.); (M.B.); (T.H.-C.); (C.B.); (M.A.N.)
| | - María Barco
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, E-50013 Zaragoza, Spain; (R.M.-B.); (J.S.-M.); (G.L.); (M.B.); (T.H.-C.); (C.B.); (M.A.N.)
| | - Tania Herrero-Continente
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, E-50013 Zaragoza, Spain; (R.M.-B.); (J.S.-M.); (G.L.); (M.B.); (T.H.-C.); (C.B.); (M.A.N.)
| | | | - David Botaya
- Aves Nobles y Derivados-Aldelis, E-50197 Zaragoza, Spain; (M.S.-M.); (D.B.)
| | - Carmen Arnal
- Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, E-50013 Zaragoza, Spain; (C.A.); (J.C.S.)
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, E-28029 Madrid, Spain
- Departamento de Patología Animal, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, E-50013 Zaragoza, Spain
| | - Cristina Barranquero
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, E-50013 Zaragoza, Spain; (R.M.-B.); (J.S.-M.); (G.L.); (M.B.); (T.H.-C.); (C.B.); (M.A.N.)
- Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, E-50013 Zaragoza, Spain; (C.A.); (J.C.S.)
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, E-28029 Madrid, Spain
| | - Joaquín C. Surra
- Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, E-50013 Zaragoza, Spain; (C.A.); (J.C.S.)
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, E-28029 Madrid, Spain
- Departamento de Producción Animal y Ciencia de los Alimentos, Escuela Politécnica Superior de Huesca, Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, E-22071 Huesca, Spain
| | - Jesús Osada
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, E-50013 Zaragoza, Spain; (R.M.-B.); (J.S.-M.); (G.L.); (M.B.); (T.H.-C.); (C.B.); (M.A.N.)
- Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, E-50013 Zaragoza, Spain; (C.A.); (J.C.S.)
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, E-28029 Madrid, Spain
- Department of Biochemistry and Molecular Biology, Veterinary School, University of Zaragoza, Miguel Servet, 177, E-50013 Zaragoza, Spain
- Correspondence: ; Tel.: +34-976-761644
| | - María A. Navarro
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, E-50013 Zaragoza, Spain; (R.M.-B.); (J.S.-M.); (G.L.); (M.B.); (T.H.-C.); (C.B.); (M.A.N.)
- Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, E-50013 Zaragoza, Spain; (C.A.); (J.C.S.)
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, E-28029 Madrid, Spain
| |
Collapse
|
18
|
Lin FY, Lin YW, Shih CM, Lin SJ, Tung YT, Li CY, Chen YH, Lin CY, Tsai YT, Huang CY. A Novel Relative High-Density Lipoprotein Index to Predict the Structural Changes in High-Density Lipoprotein and Its Ability to Inhibit Endothelial-Mesenchymal Transition. Int J Mol Sci 2021; 22:ijms22105210. [PMID: 34069162 PMCID: PMC8157136 DOI: 10.3390/ijms22105210] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 05/12/2021] [Accepted: 05/13/2021] [Indexed: 12/12/2022] Open
Abstract
Therapeutic elevation of high-density lipoprotein (HDL) is thought to minimize atherogenesis in subjects with dyslipidemia. However, this is not the case in clinical practice. The function of HDL is not determined by its concentration in the plasma but by its specific structural components. We previously identified an index for the prediction of HDL functionality, relative HDL (rHDL) index, and preliminarily explored that dysfunctional HDL (rHDL index value > 2) failed to rescue the damage to endothelial progenitor cells (EPCs). To confirm the effectiveness of the rHDL index for predicting HDL functions, here we evaluated the effects of HDL from patients with different rHDL index values on the endothelial–mesenchymal transition (EndoMT) of EPCs. We also analyzed the lipid species in HDL with different rHDL index values and investigated the structural differences that affect HDL functions. The results indicate that HDL from healthy adults and subjects with an rHDL index value < 2 protected transforming growth factor (TGF)-β1-stimulated EndoMT by modulating Smad2/3 and Snail activation. HDL from subjects with an rHDL index value > 2 failed to restore the functionality of TGF-β1-treated EPCs. Lipidomic analysis demonstrated that HDL with different rHDL index values may differ in the composition of triglycerides, phosphatidylcholine, and phosphatidylinositol. In conclusion, we confirmed the applicability of the rHDL index value to predict HDL function and found structural differences that may affect the function of HDL, which warrants further in-depth studies.
Collapse
Affiliation(s)
- Feng-Yen Lin
- Taipei Heart Institute, Taipei Medical University, Taipei 110, Taiwan; (F.-Y.L.); (C.-M.S.); (S.-J.L.)
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- Division of Cardiology, Department of Internal Medicine and Cardiovascular Research Center, Taipei Medical University Hospital, Taipei 110, Taiwan
| | - Yi-Wen Lin
- Institute of Oral Biology, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan;
| | - Chun-Ming Shih
- Taipei Heart Institute, Taipei Medical University, Taipei 110, Taiwan; (F.-Y.L.); (C.-M.S.); (S.-J.L.)
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- Division of Cardiology, Department of Internal Medicine and Cardiovascular Research Center, Taipei Medical University Hospital, Taipei 110, Taiwan
| | - Shing-Jong Lin
- Taipei Heart Institute, Taipei Medical University, Taipei 110, Taiwan; (F.-Y.L.); (C.-M.S.); (S.-J.L.)
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- Division of Cardiology, Department of Internal Medicine and Cardiovascular Research Center, Taipei Medical University Hospital, Taipei 110, Taiwan
| | - Yu-Tang Tung
- Graduate Institute of Biotechnology, National Chung Hsing University, Taichung 238, Taiwan;
| | - Chi-Yuan Li
- Department of Anesthesiology and Graduate Institute of Clinical Medical Science, China Medical University and Hospital, Taichung 406, Taiwan;
| | - Yung-Hsiang Chen
- Graduate Institute of Integrated Medicine, China Medical University, Taichung 406, Taiwan;
- Department of Psychology, College of Medical and Health Science, Asia University, Taichung 413, Taiwan
| | - Cheng-Yen Lin
- Healthcare Information and Management Department, Ming Chuan University, Taoyuan 333, Taiwan;
| | - Yi-Ting Tsai
- Taipei Heart Institute, Taipei Medical University, Taipei 110, Taiwan; (F.-Y.L.); (C.-M.S.); (S.-J.L.)
- Division of Cardiovascular Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei 115, Taiwan
- Correspondence: (Y.-T.T.); (C.-Y.H.)
| | - Chun-Yao Huang
- Taipei Heart Institute, Taipei Medical University, Taipei 110, Taiwan; (F.-Y.L.); (C.-M.S.); (S.-J.L.)
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- Division of Cardiology, Department of Internal Medicine and Cardiovascular Research Center, Taipei Medical University Hospital, Taipei 110, Taiwan
- Correspondence: (Y.-T.T.); (C.-Y.H.)
| |
Collapse
|
19
|
Vickers KC, Michell DL. HDL-small RNA Export, Transport, and Functional Delivery in Atherosclerosis. Curr Atheroscler Rep 2021; 23:38. [PMID: 33983531 DOI: 10.1007/s11883-021-00930-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/14/2021] [Indexed: 12/21/2022]
Abstract
PURPOSE OF REVIEW This review highlights recent advances on the mechanisms and impact of HDL-small non-coding RNAs (sRNA) on intercellular communication in atherosclerosis. RECENT FINDINGS Studies demonstrate that HDL-microRNAs (miRNA) are significantly altered in atherosclerotic cardiovascular disease (ASCVD), and are responsive to diet, obesity, and diabetes. Immune cells, pancreatic beta cells, and neurons are shown to export miRNAs to HDL. In turn, HDL can deliver functional miRNAs to recipient hepatocytes and endothelial cells regulating adhesion molecule expression, cytokines, and angiogenesis. With high-throughput sRNA sequencing, we now appreciate the full sRNA signature on circulating HDL, including the transport of rRNA and tRNA-derived fragments. Strikingly, HDL were highly enriched with exogenous microbial sRNAs. HDL transport a diverse set of host and non-host sRNAs that are altered in cardiometabolic diseases. Given the bioactivity of these sRNAs, they likely contribute to cellular communication within atherosclerotic lesions, and are potential disease biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Kasey C Vickers
- Department of Medicine, Vanderbilt University Medical Center, 2220 Pierce Ave. 312 Preston Research Building, Nashville, TN, 37232, USA.
| | - Danielle L Michell
- Department of Medicine, Vanderbilt University Medical Center, 2220 Pierce Ave. 312 Preston Research Building, Nashville, TN, 37232, USA
| |
Collapse
|
20
|
Yamamoto H, Yokota A, Suzuki N, Tachibana M, Tsutsumi Y. Gastric perforation caused by secondary systemic amyloidosis. Clin Case Rep 2021; 9:e04254. [PMID: 34084518 PMCID: PMC8142406 DOI: 10.1002/ccr3.4254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 04/17/2021] [Accepted: 04/19/2021] [Indexed: 11/06/2022] Open
Abstract
Amyloid A amyloidosis secondary to chronic inflammation involves systemic organs and tissues, including the gastrointestinal tract. In the present case, massive amyloid deposit caused gastric perforation. IgM co-deposition in the glomeruli was another finding of note.
Collapse
Affiliation(s)
- Hiroto Yamamoto
- Department of General MedicineShimada Municipal HospitalShimadaJapan
- Department of Diagnostic PathologyShimada Municipal HospitalShimadaJapan
| | - Akihiko Yokota
- Department of GastroenterologyShimada Municipal HospitalShimadaJapan
| | - Noriyuki Suzuki
- Department of NephrologyShimada Municipal HospitalShimadaJapan
| | | | - Yutaka Tsutsumi
- Department of Diagnostic PathologyShimada Municipal HospitalShimadaJapan
- Diagnostic Pathology ClinicPathos TsutsumiInazawaJapan
| |
Collapse
|
21
|
Mahrooz A, Shokri Y, Variji A, Zargari M, Alizadeh A, Mehtarian E. Improved risk assessment of coronary artery disease by substituting paraoxonase 1 activity for HDL-C: Novel cardiometabolic biomarkers based on HDL functionality. Nutr Metab Cardiovasc Dis 2021; 31:1166-1176. [PMID: 33579580 DOI: 10.1016/j.numecd.2020.12.026] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 10/28/2020] [Accepted: 12/22/2020] [Indexed: 11/17/2022]
Abstract
BACKGROUND AND AIMS Developing laboratory assays to evaluate HDL functions and improve cardiovascular disease (CVD) risk assessment has recently emerged as a challenge. The present study was conducted to help predict the risk of coronary artery disease (CAD) by investigating new cardiometabolic risk factors based on substituting paraoxonase 1 (PON1) as a critical enzyme in the functionality of HDL for that of HDL-C. METHODS AND RESULTS The present study recruited 274 subjects undergoing diagnostic coronary angiography, 92 without significant CAD (non-CAD), and 182 with a severe CAD. The diagnostic accuracy of the new biomarkers in non-CAD versus multi-vessel disease was obtained in descending order of AUC as 0.72 (P < 0.001) for log (TG/PON1), 0.70 (P < 0.001) for nonHDL-C/PON1, and 0.67 (P < 0.001) for LDL-C/PON1. After performing a multivariate adjustment for age, gender, BMI, statin therapy, and diabetes mellitus, the increased odds of CAD remained significant for the new cardiometabolic ratios as independent variables [adjusted OR = 1.47 (1.15-1.88), p = 0.002 for LDL-C/PON1; adjusted OR = 2.15 (1.41-3.5), p = 0.009 for nonHDL-C/PON1; adjusted OR = 5.03 (2.14-13.02), p = 0.004 for log (TG/PON1)]. CAD was diagnosed with an optimal discriminating cutoff of 1.84 for LDL-C/PON1, 2.8 for nonHDL-C/PON1, and 0.48 for log (TG/PON1). CONCLUSIONS To improve CAD's risk assessment, the PON1 activity was proposed as an alternative to HDL-C in the commonly used atherogenic lipid ratios. Substituting the PON1 activity for the HDL-C concentration can provide an index of the HDL activity. The present study sought to exploit the lipoprotein-related risk factors of CAD from a more effective perspective.
Collapse
Affiliation(s)
- Abdolkarim Mahrooz
- Molecular and Cell Biology Research Center, Mazandaran University of Medical Sciences, Sari, Iran; Department of Clinical Biochemistry and Genetics, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Yasaman Shokri
- Department of Clinical Biochemistry and Genetics, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Atena Variji
- Department of Clinical Biochemistry and Genetics, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mehryar Zargari
- Molecular and Cell Biology Research Center, Mazandaran University of Medical Sciences, Sari, Iran; Department of Clinical Biochemistry and Genetics, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Ahad Alizadeh
- Metabolic Diseases Research Center, Research Institute for Prevention of Non-Communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran.
| | - Ehsan Mehtarian
- Department of Clinical Biochemistry and Genetics, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
22
|
Morris G, Puri BK, Bortolasci CC, Carvalho A, Berk M, Walder K, Moreira EG, Maes M. The role of high-density lipoprotein cholesterol, apolipoprotein A and paraoxonase-1 in the pathophysiology of neuroprogressive disorders. Neurosci Biobehav Rev 2021; 125:244-263. [PMID: 33657433 DOI: 10.1016/j.neubiorev.2021.02.037] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 01/29/2021] [Accepted: 02/23/2021] [Indexed: 12/11/2022]
Abstract
Lowered high-density lipoprotein (HDL) cholesterol has been reported in major depressive disorder, bipolar disorder, first episode of psychosis, and schizophrenia. HDL, its major apolipoprotein component, ApoA1, and the antioxidant enzyme paraoxonase (PON)1 (which is normally bound to ApoA1) all have anti-atherogenic, antioxidant, anti-inflammatory, and immunomodulatory roles, which are discussed in this paper. The paper details the pathways mediating the anti-inflammatory effects of HDL, ApoA1 and PON1 and describes the mechanisms leading to compromised HDL and PON1 levels and function in an environment of chronic inflammation. The molecular mechanisms by which changes in HDL, ApoA1 and PON1 might contribute to the pathophysiology of the neuroprogressive disorders are explained. Moreover, the anti-inflammatory actions of ApoM-mediated sphingosine 1-phosphate (S1P) signalling are reviewed as well as the deleterious effects of chronic inflammation and oxidative stress on ApoM/S1P signalling. Finally, therapeutic interventions specifically aimed at improving the levels and function of HDL and PON1 while reducing levels of inflammation and oxidative stress are considered. These include the so-called Mediterranean diet, extra virgin olive oil, polyphenols, flavonoids, isoflavones, pomegranate juice, melatonin and the Mediterranean diet combined with the ketogenic diet.
Collapse
Affiliation(s)
- Gerwyn Morris
- Deakin University, IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia
| | | | - Chiara C Bortolasci
- Deakin University, IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Deakin University, CMMR Strategic Research Centre, School of Medicine, Geelong, Victoria, Australia.
| | - Andre Carvalho
- Deakin University, IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada
| | - Michael Berk
- Deakin University, IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Orygen, The National Centre of Excellence in Youth Mental Health, The Department of Psychiatry and The Florey Institute for Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Ken Walder
- Deakin University, IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Deakin University, CMMR Strategic Research Centre, School of Medicine, Geelong, Victoria, Australia
| | - Estefania G Moreira
- Post-Graduation Program in Health Sciences, State University of Londrina, Londrina, PR, Brazil
| | - Michael Maes
- Deakin University, IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Department of Psychiatry, King Chulalongkorn University Hospital, Bangkok, Thailand; Department of Psychiatry, Medical University of Plovdiv, Plovdiv, Bulgaria
| |
Collapse
|
23
|
Ben-Aicha S, Escate R, Casaní L, Padró T, Peña E, Arderiu G, Mendieta G, Badimón L, Vilahur G. High-density lipoprotein remodelled in hypercholesterolaemic blood induce epigenetically driven down-regulation of endothelial HIF-1α expression in a preclinical animal model. Cardiovasc Res 2021; 116:1288-1299. [PMID: 31504272 PMCID: PMC7243281 DOI: 10.1093/cvr/cvz239] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 07/24/2019] [Accepted: 08/29/2019] [Indexed: 12/12/2022] Open
Abstract
AIMS High-density lipoproteins (HDLs) are circulating micelles that transport proteins, lipids, and miRNAs. HDL-transported miRNAs (HDL-miRNAs) have lately received attention but their effects on vascular cells are not fully understood. Additionally, whether cardiovascular risk factors affect HDL-miRNAs levels and miRNA transfer to recipient cells remains equally poorly known. Here, we have investigated the changes induced by hypercholesterolaemia on HDL-miRNA levels and its effect on recipient endothelial cells (ECs). METHODS AND RESULTS Pigs were kept on a high-fat diet (HC; n = 10) or a normocholesterolaemic chow (NC; n = 10) for 10 days reaching cholesterol levels of 321.0 (229.7-378.5) mg/dL and 74.0 (62.5-80.2) mg/dL, respectively. HDL particles were isolated, purified, and quantified. HDL-miRNA profiling (n = 149 miRNAs) of HC- and NC-HDLs was performed by multipanel qPCR. Cell cultures of porcine aortic ECs were used to determine whether HDL-miRNAs were delivered to ECs. Potential target genes modulated by miRNAs were identified by bioinformatics and candidate miRNAs were validated by molecular analysis. In vivo effects in the coronary arteries of normocholesterolaemic swine administered HC- or NC-HDLs were analysed. Among the HDL-miRNAs, four were found in different amounts in HC- and NC-HDL (P < 0.05). miR-126-5p and -3p and miR-30b-5p (2.7×, 1.7×, and 1.3×, respectively) were found in higher levels and miR-103a-3p and miR-let-7g-5p (-1.6×, -1.4×, respectively) in lower levels in HC-HDL. miR-126-5p and -3p were transferred from HC-HDL to EC (2.5×; P < 0.05), but not from NC-HDL, by a SRB1-mediated mechanism. Bioinformatics revealed that HIF1α was the miR-126 target gene with the highest predictive value, which was accordingly found to be markedly reduced in HC-HDL-treated ECs and in miR126 mimic transfected ECs. In vivo validation confirmed that HIF1α was diminished in the coronary endothelial layer of NC pigs administered HC-HDL vs. those administered NC-HDL (P < 0.05). CONCLUSION Hypercholesterolaemia induces changes in the miRNA content of HDL enhancing miR126 and its delivery to ECs with the consequent down-regulation of its target gene HIF1α.
Collapse
Affiliation(s)
- Soumaya Ben-Aicha
- Cardiovascular Program-ICCC, Research Institute-Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain.,School of Medicine, University of Barcelona (UB), Barcelona, Spain
| | - Rafael Escate
- Cardiovascular Program-ICCC, Research Institute-Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain.,Centro de Investigación Biomédica en Red Cardiovascular (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Laura Casaní
- Cardiovascular Program-ICCC, Research Institute-Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain
| | - Teresa Padró
- Cardiovascular Program-ICCC, Research Institute-Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain.,Centro de Investigación Biomédica en Red Cardiovascular (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Esther Peña
- Cardiovascular Program-ICCC, Research Institute-Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain.,Centro de Investigación Biomédica en Red Cardiovascular (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Gemma Arderiu
- Cardiovascular Program-ICCC, Research Institute-Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain
| | - Guiomar Mendieta
- Cardiovascular Program-ICCC, Research Institute-Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain.,School of Medicine, University of Barcelona (UB), Barcelona, Spain.,Cardiology Department, Hospital Clinic Barcelona, Barcelona, Spain
| | - Lina Badimón
- Cardiovascular Program-ICCC, Research Institute-Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain.,Centro de Investigación Biomédica en Red Cardiovascular (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain.,Cardiovascular Research Chair, Universidad Autónoma Barcelona (UAB), Barcelona, Spain
| | - Gemma Vilahur
- Cardiovascular Program-ICCC, Research Institute-Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain.,Centro de Investigación Biomédica en Red Cardiovascular (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
24
|
Cimen S, Dursun M, Sulukaya M, Besiroglu H. Could the monocyte/HDL cholesterol ratio be an early marker of erectile dysfunction? Aging Male 2020; 23:694-699. [PMID: 30777468 DOI: 10.1080/13685538.2019.1574735] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
OBJECTIVE Research evaluating the relationship between the Monocyte/HDL-C ratio (MHR) and ED is limited in number. We aimed for determining whether MHR could be an early marker of ED in our study. MATERIAL AND METHODS Eighty-nine patients diagnosed with ED during the period of April 2018-November 2018 and 100 patients that did not have any systemic diseases and erectile dysfunction were included in our study. The presence of ED was evaluated with International Erectile Function Index (IIEF-5). Peripheral blood samples were collected from the patients for complete blood count and biochemical analyses. The relationship between ED and MHR was evaluated for both groups. RESULTS Body mass indices (BMI) of the ED group and the control group were significantly different in statistical terms. While a significant difference was observed for fasting blood glucose, total testosterone, luteinizing hormone values between the ED group and the control group, and follicle stimulant hormone values were found to be similar with each other. MHR values of two groups were found to be significantly different. DISCUSSION MHR can be used as a marker for the determination of ED's severity and for its monitoring. We also believe that a large series of clinical and research studies are required for it to be applicable in clinical practice. Being the first prospective study in literature, we believe that our study would provide important contributions to the literature.
Collapse
Affiliation(s)
- Serhan Cimen
- Department of Urology, Malatya Education and Research Hospital, Malatya, Turkey
| | - Murat Dursun
- Department of Urology, Malatya Education and Research Hospital, Malatya, Turkey
| | - Muhammed Sulukaya
- Department of Urology, Malatya Education and Research Hospital, Malatya, Turkey
| | - Huseyin Besiroglu
- Department of Urology, Catalca Ilyas Cokay State Hospital, Istanbul, Turkey
| |
Collapse
|
25
|
A Role for Caveolin-3 in the Pathogenesis of Muscular Dystrophies. Int J Mol Sci 2020; 21:ijms21228736. [PMID: 33228026 PMCID: PMC7699313 DOI: 10.3390/ijms21228736] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 11/16/2020] [Accepted: 11/17/2020] [Indexed: 12/14/2022] Open
Abstract
Caveolae are the cholesterol-rich small invaginations of the plasma membrane present in many cell types including adipocytes, endothelial cells, epithelial cells, fibroblasts, smooth muscles, skeletal muscles and cardiac muscles. They serve as specialized platforms for many signaling molecules and regulate important cellular processes like energy metabolism, lipid metabolism, mitochondria homeostasis, and mechano-transduction. Caveolae can be internalized together with associated cargo. The caveolae-dependent endocytic pathway plays a role in the withdrawal of many plasma membrane components that can be sent for degradation or recycled back to the cell surface. Caveolae are formed by oligomerization of caveolin proteins. Caveolin-3 is a muscle-specific isoform, whose malfunction is associated with several diseases including diabetes, cancer, atherosclerosis, and cardiovascular diseases. Mutations in Caveolin-3 are known to cause muscular dystrophies that are collectively called caveolinopathies. Altered expression of Caveolin-3 is also observed in Duchenne’s muscular dystrophy, which is likely a part of the pathological process leading to muscle weakness. This review summarizes the major functions of Caveolin-3 in skeletal muscles and discusses its involvement in the pathology of muscular dystrophies.
Collapse
|
26
|
Bonacina F, Da Dalt L, Catapano AL, Norata GD. Metabolic adaptations of cells at the vascular-immune interface during atherosclerosis. Mol Aspects Med 2020; 77:100918. [PMID: 33032828 PMCID: PMC7534736 DOI: 10.1016/j.mam.2020.100918] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 09/28/2020] [Accepted: 09/28/2020] [Indexed: 12/20/2022]
Abstract
Metabolic reprogramming is a physiological cellular adaptation to intracellular and extracellular stimuli that couples to cell polarization and function in multiple cellular subsets. Pathological conditions associated to nutrients overload, such as dyslipidaemia, may disturb cellular metabolic homeostasis and, in turn, affect cellular response and activation, thus contributing to disease progression. At the vascular/immune interface, the site of atherosclerotic plaque development, many of these changes occur. Here, an intimate interaction between endothelial cells (ECs), vascular smooth muscle cells (VSMCs) and immune cells, mainly monocytes/macrophages and lymphocytes, dictates physiological versus pathological response. Furthermore, atherogenic stimuli trigger metabolic adaptations both at systemic and cellular level that affect the EC layer barrier integrity, VSMC proliferation and migration, monocyte infiltration, macrophage polarization, lymphocyte T and B activation. Rewiring cellular metabolism by repurposing “metabolic drugs” might represent a pharmacological approach to modulate cell activation at the vascular immune interface thus contributing to control the immunometabolic response in the context of cardiovascular diseases.
Collapse
Affiliation(s)
- F Bonacina
- Department of Excellence of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy.
| | - L Da Dalt
- Department of Excellence of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy.
| | - A L Catapano
- Department of Excellence of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy; IRCSS Multimedica, Milan, Italy.
| | - G D Norata
- Department of Excellence of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy; IRCCS, Ospedale Bassini, Cinisello Balsamo, Italy.
| |
Collapse
|
27
|
Belyaeva VS, Stepenko YV, Lyubimov II, Kulikov AL, Tietze AA, Kochkarova IS, Martynova OV, Pokopeyko ON, Krupen’kina LA, Nagikh AS, Pokrovskiy VM, Patrakhanov EA, Belashova AV, Lebedev PR, Gureeva AV. Non-hematopoietic erythropoietin-derived peptides for atheroprotection and treatment of cardiovascular diseases. RESEARCH RESULTS IN PHARMACOLOGY 2020. [DOI: 10.3897/rrpharmacology.6.58891] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Relevance: Cardiovascular diseases continue to be the leading cause of premature adult death.Lipid profile and atherogenesis: Dislipidaemia leads to subsequent lipid accumulation and migration of immunocompetent cells into the vessel intima. Macrophages accumulate cholesterol forming foam cells – the morphological substrate of atherosclerosis in its initial stage.Inflammation and atherogenesis: Pro-inflammatory factors provoke oxidative stress, vascular wall damage and foam cells formation.Endothelial and mitochondrial dysfunction in the development of atherosclerosis: Endothelial mitochondria are some of the organelles most sensitive to oxidative stress. Damaged mitochondria produce excess superoxide and H2O2, which are the main factors of intracellular damage, further increasing endothelial dysfunction.Short non-hematopoietic erythropoietin-based peptides as innovative atheroprotectors: Research in recent decades has shown that erythropoietin has a high cytoprotective activity, which is mainly associated with exposure to the mitochondrial link and has been confirmed in various experimental models. There is also a short-chain derivative, the 11-amino acid pyroglutamate helix B surface peptide (PHBSP), which selectively binds to the erythropoietin heterodymic receptor and reproduces its cytoprotective properties. This indicates the promising use of short-chain derivatives of erythropoietin for the treatment and prevention of atherosclerotic vascular injury. In the future, it is planned to study the PHBSP derivatives, the modification of which consists in adding RGD and PGP tripeptides with antiaggregant properties to the original 11-member peptide.
Collapse
|
28
|
Holmannova D, Borsky P, Borska L, Andrys C, Hamakova K, Rehacek V, Svadlakova T, Malkova A, Beranek M, Palicka V, Krejsek J, Fiala Z. Metabolic Syndrome, Clusterin and Elafin in Patients with Psoriasis Vulgaris. Int J Mol Sci 2020; 21:ijms21165617. [PMID: 32764517 PMCID: PMC7460615 DOI: 10.3390/ijms21165617] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 07/21/2020] [Accepted: 08/04/2020] [Indexed: 12/15/2022] Open
Abstract
Background: Psoriasis is a pathological condition characterized by immune system dysfunction and inflammation. Patients with psoriasis are more likely to develop a wide range of disorders associated with inflammation. Serum levels of various substances and their combinations have been associated with the presence of the disease (psoriasis) and have shown the potential to reflect its activity. The aim of the present study is to contribute to the elucidation of pathophysiological links between psoriasis, its pro-inflammatory comorbidity metabolic syndrome (MetS), and the expression of clusterin and elafin, which are reflected in the pathophysiological “portfolio” of both diseases. Material and methods: Clinical examinations (PASI score), ELISA (clusterin, elafin), and biochemical analyses (parameters of MetS) were performed. Results: We found that patients with psoriasis were more often afflicted by MetS, compared to the healthy controls. Clusterin and elafin levels were higher in the patients than in the controls but did not correlate to the severity of psoriasis. Conclusion: Our data suggest that patients with psoriasis are more susceptible to developing other systemic inflammatory diseases, such as MetS. The levels of clusterin and elafin, which are tightly linked to inflammation, were significantly increased in the patients, compared to the controls, but the presence of MetS in patients did not further increase these levels.
Collapse
Affiliation(s)
- Drahomira Holmannova
- Institute of Hygiene and Preventive Medicine, Faculty of Medicine in Hradec Kralove, Charles University, 50038 Hradec Kralove, Czech Republic; (D.H.); (T.S.); (A.M.); (M.B.); (Z.F.)
| | - Pavel Borsky
- Institute of Hygiene and Preventive Medicine, Faculty of Medicine in Hradec Kralove, Charles University, 50038 Hradec Kralove, Czech Republic; (D.H.); (T.S.); (A.M.); (M.B.); (Z.F.)
- Institute of Pathological Physiology, Faculty of Medicine in Hradec Kralove, Charles University, 50003 Hradec Kralove, Czech Republic;
- Correspondence:
| | - Lenka Borska
- Institute of Pathological Physiology, Faculty of Medicine in Hradec Kralove, Charles University, 50003 Hradec Kralove, Czech Republic;
| | - Ctirad Andrys
- Institute of Clinical Immunology and Allergology, University Hospital and Faculty of Medicine in Hradec Kralove, Charles University, 50005 Hradec Kralove, Czech Republic; (C.A.); (J.K.)
| | - Kvetoslava Hamakova
- Clinic of Dermatology and Venereology, University Hospital Hradec Kralove, 50005 Hradec Králové, Czech Republic;
| | - Vit Rehacek
- Transfusion Center, University Hospital, 50005 Hradec Kralove, Czech Republic;
| | - Tereza Svadlakova
- Institute of Hygiene and Preventive Medicine, Faculty of Medicine in Hradec Kralove, Charles University, 50038 Hradec Kralove, Czech Republic; (D.H.); (T.S.); (A.M.); (M.B.); (Z.F.)
- Institute of Clinical Immunology and Allergology, University Hospital and Faculty of Medicine in Hradec Kralove, Charles University, 50005 Hradec Kralove, Czech Republic; (C.A.); (J.K.)
| | - Andrea Malkova
- Institute of Hygiene and Preventive Medicine, Faculty of Medicine in Hradec Kralove, Charles University, 50038 Hradec Kralove, Czech Republic; (D.H.); (T.S.); (A.M.); (M.B.); (Z.F.)
| | - Martin Beranek
- Institute of Hygiene and Preventive Medicine, Faculty of Medicine in Hradec Kralove, Charles University, 50038 Hradec Kralove, Czech Republic; (D.H.); (T.S.); (A.M.); (M.B.); (Z.F.)
- Institute of Clinical Biochemistry and Diagnostics, University Hospital Hradec Kralove and Faculty of Medicine in Hradec Kralove, Charles University, 50005 Hradec Kralove, Czech Republic;
| | - Vladimir Palicka
- Institute of Clinical Biochemistry and Diagnostics, University Hospital Hradec Kralove and Faculty of Medicine in Hradec Kralove, Charles University, 50005 Hradec Kralove, Czech Republic;
| | - Jan Krejsek
- Institute of Clinical Immunology and Allergology, University Hospital and Faculty of Medicine in Hradec Kralove, Charles University, 50005 Hradec Kralove, Czech Republic; (C.A.); (J.K.)
| | - Zdenek Fiala
- Institute of Hygiene and Preventive Medicine, Faculty of Medicine in Hradec Kralove, Charles University, 50038 Hradec Kralove, Czech Republic; (D.H.); (T.S.); (A.M.); (M.B.); (Z.F.)
| |
Collapse
|
29
|
Takaeko Y, Matsui S, Kajikawa M, Maruhashi T, Yamaji T, Harada T, Han Y, Hashimoto H, Kihara Y, Hida E, Chayama K, Goto C, Aibara Y, Yusoff FM, Kishimoto S, Nakashima A, Higashi Y. Relationship between high-density lipoprotein cholesterol levels and endothelial function in women: a cross-sectional study. BMJ Open 2020; 10:e038121. [PMID: 32641366 PMCID: PMC7342861 DOI: 10.1136/bmjopen-2020-038121] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVES The purpose of this study was to evaluate the relationship between high-density lipoprotein cholesterol (HDL-C) levels and endothelial function in women. DESIGN Cross-sectional study. SETTING 22 university hospitals and affiliated clinics in Japan. PARTICIPANTS 1719 Japanese women aged 17-90 years who were not receiving lipid-lowering therapy. MEASURES We evaluated flow-mediated vasodilation (FMD) and serum levels of HDL-C. All participants were divided into four groups by HDL-C level: low HDL-C (<40 mg/dL), moderate HDL-C (40-59 mg/dL), high HDL-C (60-79 md/dL) and extremely high HDL-C (≥80 mg/dL). RESULTS Univariate regression analysis revealed a significant relationship between FMD and HDL-C (r=0.12, p<0.001). FMD values were significantly smaller in the low HDL-C group (5.2%±3.8%) and moderate HDL-C group (5.2%±3.8%) than in the extremely high HDL-C group (6.7%±3.4%) (p=0.024 and p=0.003, respectively), while there was no significant difference in FMD between the high HDL-C group and the extremely high HDL-C group. Multiple logistic regression analysis did not show a significant association between HDL-C levels and FMD. CONCLUSIONS Endothelial function increased in relation to HDL-C levels. However, there was no association of HDL-C levels with endothelial function after adjustment of traditional cardiovascular risk factors in women. TRIAL REGISTRATION NUMBER UMIN000012950; Results.
Collapse
Affiliation(s)
- Yuji Takaeko
- Department of Cardiovascular Medicine, Hiroshima University Faculty of Medicine Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Shogo Matsui
- Department of Cardiovascular Medicine, Hiroshima University Faculty of Medicine Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Masato Kajikawa
- Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan
| | - Tatsuya Maruhashi
- Department of Cardiovascular Medicine, Hiroshima University Faculty of Medicine Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Takayuki Yamaji
- Department of Cardiovascular Medicine, Hiroshima University Faculty of Medicine Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Takahiro Harada
- Department of Cardiovascular Medicine, Hiroshima University Faculty of Medicine Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Yiming Han
- Department of Cardiovascular Medicine, Hiroshima University Faculty of Medicine Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Haruki Hashimoto
- Department of Cardiovascular Medicine, Hiroshima University Faculty of Medicine Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Yasuki Kihara
- Department of Cardiovascular Medicine, Hiroshima University Faculty of Medicine Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Eisuke Hida
- Department of Biostatistics and Data Science, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kazuaki Chayama
- Department of Gastroenterology and Metabolism, Hiroshima University Faculty of Medicine Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Chikara Goto
- Department of Physical Therapy, Hiroshima International University, HigashiHiroshima, Hiroshima, Japan
| | - Yoshiki Aibara
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima, Japan
| | - Farina Mohamad Yusoff
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima, Japan
| | - Shinji Kishimoto
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima, Japan
| | - Ayumu Nakashima
- Department of Stem Cell Biology and Medicine, Hiroshima University Faculty of Medicine Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Yukihito Higashi
- Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima, Japan
| |
Collapse
|
30
|
Zaric B, Obradovic M, Trpkovic A, Banach M, Mikhailidis DP, Isenovic ER. Endothelial Dysfunction in Dyslipidaemia: Molecular Mechanisms and Clinical Implications. Curr Med Chem 2020; 27:1021-1040. [DOI: 10.2174/0929867326666190903112146] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 08/23/2019] [Accepted: 08/23/2019] [Indexed: 12/13/2022]
Abstract
The endothelium consists of a monolayer of Endothelial Cells (ECs) which form
the inner cellular lining of veins, arteries, capillaries and lymphatic vessels. ECs interact with
the blood and lymph. The endothelium fulfils functions such as vasodilatation, regulation of
adhesion, infiltration of leukocytes, inhibition of platelet adhesion, vessel remodeling and
lipoprotein metabolism. ECs synthesize and release compounds such as Nitric Oxide (NO),
metabolites of arachidonic acid, Reactive Oxygen Species (ROS) and enzymes that degrade
the extracellular matrix. Endothelial dysfunction represents a phenotype prone to atherogenesis
and may be used as a marker of atherosclerotic risk. Such dysfunction includes impaired
synthesis and availability of NO and an imbalance in the relative contribution of endothelialderived
relaxing factors and contracting factors such as endothelin-1 and angiotensin. This
dysfunction appears before the earliest anatomic evidence of atherosclerosis and could be an
important initial step in further development of atherosclerosis. Endothelial dysfunction was
historically treated with vitamin C supplementation and L-arginine supplementation. Short
term improvement of the expression of adhesion molecule and endothelial function during
antioxidant therapy has been observed. Statins are used in the treatment of hyperlipidaemia, a
risk factor for cardiovascular disease. Future studies should focus on identifying the mechanisms
involved in the beneficial effects of statins on the endothelium. This may help develop
drugs specifically aimed at endothelial dysfunction.
Collapse
Affiliation(s)
- Bozidarka Zaric
- Laboratory of Radiobiology and Molecular Genetics, Vinca Institute of Nuclear Sciences, University of Belgrade, Mike Petrovica Alasa 12-14, 11000 Belgrade, Serbia
| | - Milan Obradovic
- Laboratory of Radiobiology and Molecular Genetics, Vinca Institute of Nuclear Sciences, University of Belgrade, Mike Petrovica Alasa 12-14, 11000 Belgrade, Serbia
| | - Andreja Trpkovic
- Laboratory of Radiobiology and Molecular Genetics, Vinca Institute of Nuclear Sciences, University of Belgrade, Mike Petrovica Alasa 12-14, 11000 Belgrade, Serbia
| | - Maciej Banach
- Department of Hypertension, WAM University Hospital in Lodz, Medical University of Lodz, Lodz, Poland
| | - Dimitri P. Mikhailidis
- Department of Clinical Biochemistry, Royal Free Campus, University College London Medical School, University College London (UCL), London, United Kingdom
| | - Esma R. Isenovic
- Laboratory of Radiobiology and Molecular Genetics, Vinca Institute of Nuclear Sciences, University of Belgrade, Mike Petrovica Alasa 12-14, 11000 Belgrade, Serbia
| |
Collapse
|
31
|
Kwaifa IK, Bahari H, Yong YK, Noor SM. Endothelial Dysfunction in Obesity-Induced Inflammation: Molecular Mechanisms and Clinical Implications. Biomolecules 2020; 10:biom10020291. [PMID: 32069832 PMCID: PMC7072669 DOI: 10.3390/biom10020291] [Citation(s) in RCA: 155] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 10/04/2019] [Accepted: 10/05/2019] [Indexed: 12/21/2022] Open
Abstract
Obesity is characterized by the excessive deposition of fat that may interfere with the normal metabolic process of the body. It is a chronic condition associated with various metabolic syndromes, whose prevalence is grossly increasing, and affects both children and adults. Accumulation of excessive macronutrients on the adipose tissues promotes the secretion and release of inflammatory mediators, including interleukin-6 (IL-6), interleukin 1β, tumor necrotic factor-α (TNF-α), leptin, and stimulation of monocyte chemoattractant protein-1 (MCP-1), which subsequently reduce the production of adiponectin thereby initiating a proinflammatory state. During obesity, adipose tissue synthesizes and releases a large number of hormones and cytokines that alter the metabolic processes, with a profound influence on endothelial dysfunction, a situation associated with the formation of atherosclerotic plaque. Endothelial cells respond to inflammation and stimulation of MCP-1, which is described as the activation of adhesion molecules leading to proliferation and transmigration of leukocytes, which facilitates their increase in atherogenic and thromboembolic potentials. Endothelial dysfunction forms the cornerstone of this discussion, as it has been considered as the initiator in the progression of cardiovascular diseases in obesity. Overexpression of proinflammatory cytokines with subsequent reduction of anti-inflammatory markers in obesity, is considered to be the link between obesity-induced inflammation and endothelial dysfunction. Inhibition of inflammatory mechanisms and management and control of obesity can assist in reducing the risks associated with cardiovascular complications.
Collapse
Affiliation(s)
- Ibrahim Kalle Kwaifa
- Department of Pathology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia (UPM), Selangor 43400, Malaysia;
- Department of Haematology, School of Medical Laboratory Sciences, College of Health Sciences, Usmanu Danfodiyo University (UDU), Sokoto, North-Western 2346, Nigeria
| | - Hasnah Bahari
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia (UPM), Selangor 43400, Malaysia; (H.B.); (Y.K.Y.)
| | - Yoke Keong Yong
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia (UPM), Selangor 43400, Malaysia; (H.B.); (Y.K.Y.)
| | - Sabariah Md Noor
- Department of Pathology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia (UPM), Selangor 43400, Malaysia;
- Correspondence: ; Tel.: +60-193220798
| |
Collapse
|
32
|
Wu J, Dong T, Chen T, Sun J, Luo J, He J, Wei L, Zeng B, Zhang H, Li W, Liu J, Chen X, Su M, Ni Y, Jiang Q, Zhang Y, Xi Q. Hepatic exosome-derived miR-130a-3p attenuates glucose intolerance via suppressing PHLPP2 gene in adipocyte. Metabolism 2020; 103:154006. [PMID: 31715176 DOI: 10.1016/j.metabol.2019.154006] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 09/17/2019] [Accepted: 09/24/2019] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Glucose and lipid metabolism disorders are a major risk factor for type II diabetes and cardiovascular diseases. Evidence has indicated that the interplay between the liver and adipose tissue is crucial in maintaining energy homeostasis. Recently, the interaction between two distant endocrine organs mainly focuses on the regulation of hormones and receptors. However, as a novel carrier in the inter-tissue communication, exosomes plays a role in liver-fat crosstalk, but its effects on glucose and lipid metabolisms are still unclear. In this study, we sought to investigate the effects of hepatic exosome-derived miR-130a-3p in the regulation of glucose/lipid metabolism in adipose tissues. MEASURE In vivo, we constructed generalized miR-130a-3p knockout (130KO) and overexpressed (130OE) mice. Wild type (WT), 130KO and 130OE mice (n = 10) were assigned to a randomized controlled trial and were fed diets with either 10% (standard diet, SD) or 60% (high-fat diet, HFD) of total calories from fat (lard). Next, hepatic exosomes were extracted from WT-SD, 130KO-SD and 130OE-SD mice (WT-EXO, KO-EXO, OE-EXO), and 130KO mice were injected with 100 mg hepatic exosomes of different sources via tail-vein (once every 48 h) for 28 days, fed with HFD. In vitro, 3T3-L1 cells were treated with miR-130a-3p mimics, inhibitor and hepatic exosomes. Growth performance and glucose and lipid metabolic profiles were examined. RESULTS After feeding with HFD, the weights of 130KO mice were markedly higher than WT mice. Over-expression of miR-130a-3p in 130OE mice and intravenous injection of 130OE-EXO in 130KO mice contributed to a positive correlation with the recovery of insulin resistance. In addition, miR-130a-3p mimics and 130OE-EXO treatment of 3T3-L1 cells exhibited decreasing generations of lipid droplets and increasing glucose uptake. Conversely, inhibition of miR-130a-3p in vitro and in vivo resulted in opposite phenotype changes. Furthermore, PHLPP2 was identified as a direct target of miR-130a-3p, and the hepatic exosome-derived miR-130a-3p could improve glucose intolerance via suppressing PHLPP2 to activate AKT-AS160-GLUT4 signaling pathway in adipocytes. CONCLUSIONS We demonstrated that hepatic exosome-derived miR-130a regulated energy metabolism in adipose tissues, and elucidated a new molecular mechanism that hepatic exosome-derived miR-130a-3p is a crucial participant in organismic energy homeostasis through mediating crosstalk between the liver and adipose tissues.
Collapse
Affiliation(s)
- Jiahan Wu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center For Breeding Swine Industry, Guangdong Province Research Center of Woody Forage Engineering and Technology, College of Animal Science, South China Agricultural University, 483 Wushan Road, Guangzhou 510642, China
| | - Tao Dong
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center For Breeding Swine Industry, Guangdong Province Research Center of Woody Forage Engineering and Technology, College of Animal Science, South China Agricultural University, 483 Wushan Road, Guangzhou 510642, China
| | - Ting Chen
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center For Breeding Swine Industry, Guangdong Province Research Center of Woody Forage Engineering and Technology, College of Animal Science, South China Agricultural University, 483 Wushan Road, Guangzhou 510642, China
| | - Jiajie Sun
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center For Breeding Swine Industry, Guangdong Province Research Center of Woody Forage Engineering and Technology, College of Animal Science, South China Agricultural University, 483 Wushan Road, Guangzhou 510642, China
| | - Junyi Luo
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center For Breeding Swine Industry, Guangdong Province Research Center of Woody Forage Engineering and Technology, College of Animal Science, South China Agricultural University, 483 Wushan Road, Guangzhou 510642, China
| | - Jiajian He
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center For Breeding Swine Industry, Guangdong Province Research Center of Woody Forage Engineering and Technology, College of Animal Science, South China Agricultural University, 483 Wushan Road, Guangzhou 510642, China
| | - Limin Wei
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center For Breeding Swine Industry, Guangdong Province Research Center of Woody Forage Engineering and Technology, College of Animal Science, South China Agricultural University, 483 Wushan Road, Guangzhou 510642, China
| | - Bin Zeng
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center For Breeding Swine Industry, Guangdong Province Research Center of Woody Forage Engineering and Technology, College of Animal Science, South China Agricultural University, 483 Wushan Road, Guangzhou 510642, China
| | - Haojie Zhang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center For Breeding Swine Industry, Guangdong Province Research Center of Woody Forage Engineering and Technology, College of Animal Science, South China Agricultural University, 483 Wushan Road, Guangzhou 510642, China
| | - Weite Li
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center For Breeding Swine Industry, Guangdong Province Research Center of Woody Forage Engineering and Technology, College of Animal Science, South China Agricultural University, 483 Wushan Road, Guangzhou 510642, China
| | - Jie Liu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center For Breeding Swine Industry, Guangdong Province Research Center of Woody Forage Engineering and Technology, College of Animal Science, South China Agricultural University, 483 Wushan Road, Guangzhou 510642, China
| | - Xingping Chen
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center For Breeding Swine Industry, Guangdong Province Research Center of Woody Forage Engineering and Technology, College of Animal Science, South China Agricultural University, 483 Wushan Road, Guangzhou 510642, China
| | - Mei Su
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center For Breeding Swine Industry, Guangdong Province Research Center of Woody Forage Engineering and Technology, College of Animal Science, South China Agricultural University, 483 Wushan Road, Guangzhou 510642, China
| | - Yuechun Ni
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center For Breeding Swine Industry, Guangdong Province Research Center of Woody Forage Engineering and Technology, College of Animal Science, South China Agricultural University, 483 Wushan Road, Guangzhou 510642, China
| | - Qingyan Jiang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center For Breeding Swine Industry, Guangdong Province Research Center of Woody Forage Engineering and Technology, College of Animal Science, South China Agricultural University, 483 Wushan Road, Guangzhou 510642, China
| | - Yongliang Zhang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center For Breeding Swine Industry, Guangdong Province Research Center of Woody Forage Engineering and Technology, College of Animal Science, South China Agricultural University, 483 Wushan Road, Guangzhou 510642, China.
| | - Qianyun Xi
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center For Breeding Swine Industry, Guangdong Province Research Center of Woody Forage Engineering and Technology, College of Animal Science, South China Agricultural University, 483 Wushan Road, Guangzhou 510642, China.
| |
Collapse
|
33
|
The Role and Function of HDL in Patients with Chronic Kidney Disease and the Risk of Cardiovascular Disease. Int J Mol Sci 2020; 21:ijms21020601. [PMID: 31963445 PMCID: PMC7014265 DOI: 10.3390/ijms21020601] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 01/09/2020] [Accepted: 01/10/2020] [Indexed: 02/06/2023] Open
Abstract
Chronic kidney disease (CKD) is a worldwide health problem with steadily increasing occurrence. Significantly elevated cardiovascular morbidity and mortality have been observed in CKD. Cardiovascular diseases are the most important and frequent cause of death of CKD patients globally. The presence of CKD is related to disturbances in lipoprotein metabolism whose consequences are dyslipidemia and the accumulation of atherogenic particles. CKD not only fuels the reduction of high-density lipoprotein (HDL) cholesterol concentration, but also it modifies the composition of this lipoprotein. The key role of HDL is the participation in reverse cholesterol transport from peripheral tissues to the liver. Moreover, HDL prevents the oxidation of low-density lipoprotein (LDL) cholesterol by reactive oxygen species (ROS) and protects against the adverse effects of oxidized LDL (ox-LDL) on the endothelium. Numerous studies have demonstrated the ability of HDL to promote the production of nitric oxide (NO) by endothelial cells (ECs) and to exert antiapoptotic and anti-inflammatory effects. Increasing evidence suggests that in patients with chronic inflammatory disorders, HDLs may lose important antiatherosclerotic properties and become dysfunctional. So far, no therapeutic strategy to raise HDL, or alter the ratio of HDL subfractions, has been successful in slowing the progression of CKD or reducing cardiovascular disease in patients either with or without CKD.
Collapse
|
34
|
Li J, Zhou C, Xu H, Brook RD, Liu S, Yi T, Wang Y, Feng B, Zhao M, Wang X, Zhao Q, Chen J, Song X, Wang T, Liu S, Zhang Y, Wu R, Gao J, Pan B, Pennathur S, Rajagopalan S, Huo Y, Zheng L, Huang W. Ambient Air Pollution Is Associated With HDL (High-Density Lipoprotein) Dysfunction in Healthy Adults. Arterioscler Thromb Vasc Biol 2020; 39:513-522. [PMID: 30700134 DOI: 10.1161/atvbaha.118.311749] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Objective- We aimed to assess whether exposure to higher levels of ambient air pollution impairs HDL (high-density lipoprotein) function and to elucidate the underlying biological mechanisms potentially involved. Approach and Results- In the Beijing AIRCHD study (Air Pollution and Cardiovascular Dysfunction in Healthy Adults), 73 healthy adults (23.3±5.4 years) were followed-up with 4 repeated study visits in 2014 to 2016. During each visit, ambient air pollution concentrations, HDL function metrics, and parameters of inflammation and oxidative stress were measured. Average daily concentrations of ambient particulate matter in diameter <2.5 μm were 62.9 µg/m3 (8.1-331.0 µg/m3). We observed significant decreases in HDL cholesterol efflux capacity of 2.3% (95% CI, -4.3 to -0.3) to 5.0% (95% CI, -7.6 to -2.4) associated with interquartile range increases in moving average concentrations of particulate matter in diameter <2.5 μm and traffic-related air pollutants (black carbon, nitrogen dioxide, and carbon monoxide) during the 1 to 7 days before each participant's clinic visit. Higher ambient air pollutant levels were also associated with significant reductions in circulating HDL cholesterol and apoA-I (apolipoprotein A-I), as well as elevations in HDL oxidation index, oxidized LDL (low-density lipoprotein), malondialdehyde, and high-sensitivity C-reactive protein. Conclusions- Higher ambient air pollution concentrations were associated with impairments in HDL functionality, potentially because of systemic inflammation and oxidative stress. These novel findings further our understanding of the mechanisms whereby air pollutants promote cardiometabolic disorders.
Collapse
Affiliation(s)
- Jianping Li
- From the Division of Cardiology, Peking University First Hospital, Beijing (J.L., S.L., T.Y., Y.H.).,Key Laboratory of Molecular Cardiovascular Sciences of Ministry of Education, Health Science Center (J.L., C.Z., H.X., S.L., T.Y., B.F., M.Z., X.W., Q.Z., S.L., Y.Z., R.W., X.S., T.W., J.G., B.P., Y.H., L.Z., W.H.), Peking University, Beijing
| | - Changping Zhou
- Institute of Cardiovascular Sciences (C.Z., M.Z., X.W., J.G., B.P., L.Z.), Peking University School of Basic Medical Sciences, Beijing.,Institute of Systems Biomedicine (C.Z., M.Z., X.W., J.G., B.P., L.Z.), Peking University School of Basic Medical Sciences, Beijing.,Key Laboratory of Molecular Cardiovascular Sciences of Ministry of Education, Health Science Center (J.L., C.Z., H.X., S.L., T.Y., B.F., M.Z., X.W., Q.Z., S.L., Y.Z., R.W., X.S., T.W., J.G., B.P., Y.H., L.Z., W.H.), Peking University, Beijing
| | - Hongbing Xu
- Department of Occupational and Environmental Health, Peking University School of Public Health, Peking University Institute of Environmental Medicine (H.X., B.F., Q.Z., S.L., Y.Z., R.W., X.S., T.W., W.H., J.C.).,Key Laboratory of Molecular Cardiovascular Sciences of Ministry of Education, Health Science Center (J.L., C.Z., H.X., S.L., T.Y., B.F., M.Z., X.W., Q.Z., S.L., Y.Z., R.W., X.S., T.W., J.G., B.P., Y.H., L.Z., W.H.), Peking University, Beijing
| | - Robert D Brook
- Division of Cardiovascular Medicine (R.D.B.), University of Michigan, Ann Arbor
| | - Shengcong Liu
- From the Division of Cardiology, Peking University First Hospital, Beijing (J.L., S.L., T.Y., Y.H.).,Key Laboratory of Molecular Cardiovascular Sciences of Ministry of Education, Health Science Center (J.L., C.Z., H.X., S.L., T.Y., B.F., M.Z., X.W., Q.Z., S.L., Y.Z., R.W., X.S., T.W., J.G., B.P., Y.H., L.Z., W.H.), Peking University, Beijing
| | - Tieci Yi
- From the Division of Cardiology, Peking University First Hospital, Beijing (J.L., S.L., T.Y., Y.H.)
| | - Yang Wang
- Department of Prevention and Health Care, Hospital of Health Science Center (Y.W.), Peking University, Beijing
| | - Baihuan Feng
- Department of Occupational and Environmental Health, Peking University School of Public Health, Peking University Institute of Environmental Medicine (H.X., B.F., Q.Z., S.L., Y.Z., R.W., X.S., T.W., W.H., J.C.)
| | - Mingming Zhao
- Institute of Cardiovascular Sciences (C.Z., M.Z., X.W., J.G., B.P., L.Z.), Peking University School of Basic Medical Sciences, Beijing.,Institute of Systems Biomedicine (C.Z., M.Z., X.W., J.G., B.P., L.Z.), Peking University School of Basic Medical Sciences, Beijing
| | - Xu Wang
- Institute of Cardiovascular Sciences (C.Z., M.Z., X.W., J.G., B.P., L.Z.), Peking University School of Basic Medical Sciences, Beijing.,Institute of Systems Biomedicine (C.Z., M.Z., X.W., J.G., B.P., L.Z.), Peking University School of Basic Medical Sciences, Beijing
| | - Qian Zhao
- Department of Occupational and Environmental Health, Peking University School of Public Health, Peking University Institute of Environmental Medicine (H.X., B.F., Q.Z., S.L., Y.Z., R.W., X.S., T.W., W.H., J.C.)
| | - Jie Chen
- Department of Occupational and Environmental Health, Peking University School of Public Health, Peking University Institute of Environmental Medicine (H.X., B.F., Q.Z., S.L., Y.Z., R.W., X.S., T.W., W.H., J.C.).,Institute for Risk Assessment Sciences (J.C.), University Medical Centre Utrecht, University of Utrecht, the Netherlands.,Julius Centre for Health Sciences and Primary Care (J.C.), University Medical Centre Utrecht, University of Utrecht, the Netherlands
| | - Xiaoming Song
- Department of Occupational and Environmental Health, Peking University School of Public Health, Peking University Institute of Environmental Medicine (H.X., B.F., Q.Z., S.L., Y.Z., R.W., X.S., T.W., W.H., J.C.).,Key Laboratory of Molecular Cardiovascular Sciences of Ministry of Education, Health Science Center (J.L., C.Z., H.X., S.L., T.Y., B.F., M.Z., X.W., Q.Z., S.L., Y.Z., R.W., X.S., T.W., J.G., B.P., Y.H., L.Z., W.H.), Peking University, Beijing
| | - Tong Wang
- Department of Occupational and Environmental Health, Peking University School of Public Health, Peking University Institute of Environmental Medicine (H.X., B.F., Q.Z., S.L., Y.Z., R.W., X.S., T.W., W.H., J.C.).,Key Laboratory of Molecular Cardiovascular Sciences of Ministry of Education, Health Science Center (J.L., C.Z., H.X., S.L., T.Y., B.F., M.Z., X.W., Q.Z., S.L., Y.Z., R.W., X.S., T.W., J.G., B.P., Y.H., L.Z., W.H.), Peking University, Beijing
| | - Shuo Liu
- Department of Occupational and Environmental Health, Peking University School of Public Health, Peking University Institute of Environmental Medicine (H.X., B.F., Q.Z., S.L., Y.Z., R.W., X.S., T.W., W.H., J.C.)
| | - Yi Zhang
- Department of Occupational and Environmental Health, Peking University School of Public Health, Peking University Institute of Environmental Medicine (H.X., B.F., Q.Z., S.L., Y.Z., R.W., X.S., T.W., W.H., J.C.).,Key Laboratory of Molecular Cardiovascular Sciences of Ministry of Education, Health Science Center (J.L., C.Z., H.X., S.L., T.Y., B.F., M.Z., X.W., Q.Z., S.L., Y.Z., R.W., X.S., T.W., J.G., B.P., Y.H., L.Z., W.H.), Peking University, Beijing
| | - Rongshan Wu
- Department of Occupational and Environmental Health, Peking University School of Public Health, Peking University Institute of Environmental Medicine (H.X., B.F., Q.Z., S.L., Y.Z., R.W., X.S., T.W., W.H., J.C.).,Key Laboratory of Molecular Cardiovascular Sciences of Ministry of Education, Health Science Center (J.L., C.Z., H.X., S.L., T.Y., B.F., M.Z., X.W., Q.Z., S.L., Y.Z., R.W., X.S., T.W., J.G., B.P., Y.H., L.Z., W.H.), Peking University, Beijing
| | - Jianing Gao
- Institute of Cardiovascular Sciences (C.Z., M.Z., X.W., J.G., B.P., L.Z.), Peking University School of Basic Medical Sciences, Beijing.,Institute of Systems Biomedicine (C.Z., M.Z., X.W., J.G., B.P., L.Z.), Peking University School of Basic Medical Sciences, Beijing.,Key Laboratory of Molecular Cardiovascular Sciences of Ministry of Education, Health Science Center (J.L., C.Z., H.X., S.L., T.Y., B.F., M.Z., X.W., Q.Z., S.L., Y.Z., R.W., X.S., T.W., J.G., B.P., Y.H., L.Z., W.H.), Peking University, Beijing
| | - Bing Pan
- Institute of Cardiovascular Sciences (C.Z., M.Z., X.W., J.G., B.P., L.Z.), Peking University School of Basic Medical Sciences, Beijing.,Institute of Systems Biomedicine (C.Z., M.Z., X.W., J.G., B.P., L.Z.), Peking University School of Basic Medical Sciences, Beijing.,Key Laboratory of Molecular Cardiovascular Sciences of Ministry of Education, Health Science Center (J.L., C.Z., H.X., S.L., T.Y., B.F., M.Z., X.W., Q.Z., S.L., Y.Z., R.W., X.S., T.W., J.G., B.P., Y.H., L.Z., W.H.), Peking University, Beijing
| | | | - Sanjay Rajagopalan
- Division of Cardiovascular Medicine, Case Western Reserve Medical School, Cleveland OH (S.R.), Peking University, Beijing
| | - Yong Huo
- From the Division of Cardiology, Peking University First Hospital, Beijing (J.L., S.L., T.Y., Y.H.).,Key Laboratory of Molecular Cardiovascular Sciences of Ministry of Education, Health Science Center (J.L., C.Z., H.X., S.L., T.Y., B.F., M.Z., X.W., Q.Z., S.L., Y.Z., R.W., X.S., T.W., J.G., B.P., Y.H., L.Z., W.H.), Peking University, Beijing
| | - Lemin Zheng
- Institute of Cardiovascular Sciences (C.Z., M.Z., X.W., J.G., B.P., L.Z.), Peking University School of Basic Medical Sciences, Beijing.,Institute of Systems Biomedicine (C.Z., M.Z., X.W., J.G., B.P., L.Z.), Peking University School of Basic Medical Sciences, Beijing.,Key Laboratory of Molecular Cardiovascular Sciences of Ministry of Education, Health Science Center (J.L., C.Z., H.X., S.L., T.Y., B.F., M.Z., X.W., Q.Z., S.L., Y.Z., R.W., X.S., T.W., J.G., B.P., Y.H., L.Z., W.H.), Peking University, Beijing
| | - Wei Huang
- Department of Occupational and Environmental Health, Peking University School of Public Health, Peking University Institute of Environmental Medicine (H.X., B.F., Q.Z., S.L., Y.Z., R.W., X.S., T.W., W.H., J.C.).,Key Laboratory of Molecular Cardiovascular Sciences of Ministry of Education, Health Science Center (J.L., C.Z., H.X., S.L., T.Y., B.F., M.Z., X.W., Q.Z., S.L., Y.Z., R.W., X.S., T.W., J.G., B.P., Y.H., L.Z., W.H.), Peking University, Beijing
| |
Collapse
|
35
|
Radulović S, Gottschalk B, Hörl G, Zardoya-Laguardia P, Schilcher I, Hallström S, Vujić N, Schmidt K, Trieb M, Graier WF, Malli R, Kratky D, Marsche G, Frank S. Endothelial lipase increases eNOS activating capacity of high-density lipoprotein. Biochim Biophys Acta Mol Cell Biol Lipids 2020; 1865:158612. [PMID: 31923467 PMCID: PMC7116681 DOI: 10.1016/j.bbalip.2020.158612] [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: 06/28/2019] [Revised: 12/30/2019] [Accepted: 12/31/2019] [Indexed: 12/26/2022]
Abstract
Endothelial lipase (EL) changes structural and functional properties of high-density lipoprotein (HDL). HDL is a relevant modulator of endothelial nitric oxide synthase (eNOS) activity, but the effect of EL on HDL induced eNOS-activation has not yet been investigated. Here, we examined the impact of EL-modified HDL (EL-HDL) on eNOS activity, subcellular trafficking, and eNOS- dependent vasorelaxation. EL-HDL and empty virus (EV)-HDL as control were isolated from human serum incubated with EL-overexpressing or EV infected HepG2 cells. EL-HDL exhibited higher capacity to induce eNOS phosphorylation at Ser1177 and eNOS activity in EA.hy 926 cells, as well as eNOS-dependent vasorelaxation of mouse aortic rings compared to control HDL. As revealed by confocal and structured illumination-microscopy EL-HDL-driven induction of eNOS was accompanied by an increased eNOS-GFP targeting to the plasma membrane and a lower eNOS-GFP colocalization with Golgi and mitochondria. Widefield microscopy of filipin stained cells revealed that EL-HDL lowered cellular free cholesterol (FC) and as found by thin-layer chromatography increased cellular cholesterol ester (CE) content. Additionally, cholesterol efflux capacity, acyl-coenzyme A: cholesterol acyltransferase activity, and HDL particle uptake were comparable between EL-HDL and control HDL. In conclusion, EL increases eNOS activating capacity of HDL, a phenomenon accompanied by an enrichment of the plasma membrane eNOS pool, a decreased cell membrane FC and increased cellular CE content.
Collapse
Affiliation(s)
- Snježana Radulović
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria
| | - Benjamin Gottschalk
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria
| | - Gerd Hörl
- Otto Loewi Research Center, Division of Physiological Chemistry, Medical University of Graz, Neue Stiftingtalstraße 6/3, 8010 Graz, Austria
| | - Pablo Zardoya-Laguardia
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria
| | - Irene Schilcher
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria
| | - Seth Hallström
- Otto Loewi Research Center, Division of Physiological Chemistry, Medical University of Graz, Neue Stiftingtalstraße 6/3, 8010 Graz, Austria
| | - Nemanja Vujić
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria
| | - Kurt Schmidt
- Department of Pharmacology and Toxicology, University of Graz, Graz, Austria
| | - Markus Trieb
- Otto Loewi Research Center, Division of Experimental and Clinical Pharmacology, Medical University of Graz, Universitätsplatz 4, 8010 Graz, Austria
| | - Wolfgang F Graier
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria; BioTechMed-Graz, Mozartgasse 12/II, 8010 Graz, Austria
| | - Roland Malli
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria; BioTechMed-Graz, Mozartgasse 12/II, 8010 Graz, Austria
| | - Dagmar Kratky
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria; BioTechMed-Graz, Mozartgasse 12/II, 8010 Graz, Austria
| | - Gunther Marsche
- Otto Loewi Research Center, Division of Experimental and Clinical Pharmacology, Medical University of Graz, Universitätsplatz 4, 8010 Graz, Austria; BioTechMed-Graz, Mozartgasse 12/II, 8010 Graz, Austria
| | - Saša Frank
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria; BioTechMed-Graz, Mozartgasse 12/II, 8010 Graz, Austria.
| |
Collapse
|
36
|
Mutual Influences between Nitric Oxide and Paraoxonase 1. Antioxidants (Basel) 2019; 8:antiox8120619. [PMID: 31817387 PMCID: PMC6943684 DOI: 10.3390/antiox8120619] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 12/02/2019] [Accepted: 12/03/2019] [Indexed: 12/13/2022] Open
Abstract
One of the best consolidated paradigms in vascular pharmacology is that an uncontrolled excess of oxidizing chemical species causes tissue damage and loss of function in the endothelial and subendothelial layers. The fact that high-density lipoproteins play an important role in preventing such an imbalance is integrated into that concept, for which the expression and activity of paraoxonases is certainly crucial. The term paraoxonase (aryldialkyl phosphatase, EC 3.1.8.1) encompasses at least three distinct isoforms, with a wide variation in substrate affinity, cell and fluid localization, and biased expression of polymorphism. The purpose of this review is to determine the interactions that paraoxonase 1 has with nitric oxide synthase, its reaction product, nitric oxide (nitrogen monoxide, NO), and its derived reactive species generated in an oxidative medium, with a special focus on its pathological implications.
Collapse
|
37
|
Nezu T, Hosomi N, Kitagawa K, Nagai Y, Nakagawa Y, Aoki S, Kagimura T, Maruyama H, Origasa H, Minematsu K, Uchiyama S, Matsumoto M. Effect of Statin on Stroke Recurrence Prevention at Different Infarction Locations: A Post Hoc Analysis of The J-STARS Study. J Atheroscler Thromb 2019; 27:524-533. [PMID: 31554765 PMCID: PMC7355099 DOI: 10.5551/jat.51391] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Aim: Posterior circulation stroke (PCS) has different clinical features and prognosis compared with anterior circulation stroke (ACS), and whether the effect of statin therapy on stroke prevention differs according to infarction location remains unclear. This post hoc analysis of the J-STARS study aimed to compare the usefulness of statin at different infarction locations (i.e., ACS and PCS). Methods: In the J-STARS study, 1578 patients were randomly assigned to the pravastatin or control group. The subjects were divided into two subgroups (ACS and PCS groups) based on the arteries responsible for the infarction. Cox proportional hazards models were used to investigate whether the all stroke recurrence rate was different between the ACS and PCS groups. Results: The PCS group (n = 499) had a significantly higher prevalence of diabetes than the ACS group (n = 1022) (30.7% vs. 19.8%, P < 0.001). During the follow-up (4.9 ± 1.4 years), the incidence of all stroke was significantly lower in the pravastatin group than in the control group among patients with PCS (adjusted hazard ratio [HR] 0.46, 95% confidence interval [CI] 0.25–0.83, P = 0.009); however, the stroke recurrence rates were not significantly different between both groups among patients with ACS (adjusted HR 1.32, 95% CI 0.93–1.88, P = 0.123). A significant interaction between the ACS and PCS groups in terms of pravastatin effects was noted (P = 0.003 for interaction). Conclusions: Pravastatin significantly reduced the recurrence rate of all stroke among patients with PCS. Thus, the effect of statin on the recurrence of stroke may differ according to infarction location.
Collapse
Affiliation(s)
- Tomohisa Nezu
- Department of Clinical Neuroscience and Therapeutics, Hiroshima University Graduate School of Biomedical and Health Sciences
| | - Naohisa Hosomi
- Department of Clinical Neuroscience and Therapeutics, Hiroshima University Graduate School of Biomedical and Health Sciences
| | - Kazuo Kitagawa
- Department of Neurology, Tokyo Women's Medical University School of Medicine
| | - Yoji Nagai
- Center for Clinical Research, Kobe University Hospital
| | - Yoko Nakagawa
- Division of Medical Statistics, Translational Research Informatics Center, Foundation for Biomedical Research and Innovation
| | - Shiro Aoki
- Department of Clinical Neuroscience and Therapeutics, Hiroshima University Graduate School of Biomedical and Health Sciences
| | - Tatsuo Kagimura
- Division of Medical Statistics, Translational Research Informatics Center, Foundation for Biomedical Research and Innovation
| | - Hirofumi Maruyama
- Department of Clinical Neuroscience and Therapeutics, Hiroshima University Graduate School of Biomedical and Health Sciences
| | - Hideki Origasa
- Division of Biostatistics and Clinical Epidemiology, University of Toyama Graduate School of Medicine and Pharmaceutical Sciences
| | | | - Shinichiro Uchiyama
- Clinical Research Center for Medicine, International University of Health and Welfare, Center for Brain and Cerebral Vessels, Sanno Hospital and Sanno Medical Center
| | - Masayasu Matsumoto
- Department of Clinical Neuroscience and Therapeutics, Hiroshima University Graduate School of Biomedical and Health Sciences.,Sakai City Medical Center, Sakai City Hospital Organization
| | | |
Collapse
|
38
|
Xu W, Qian M, Huang C, Cui P, Li W, Du Q, Yi S, Shi X, Guo Y, Zheng J, Liu D, Lin D. Comparison of Mechanisms of Endothelial Cell Protections Between High-Density Lipoprotein and Apolipoprotein A-I Mimetic Peptide. Front Pharmacol 2019; 10:817. [PMID: 31379582 PMCID: PMC6659106 DOI: 10.3389/fphar.2019.00817] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 06/24/2019] [Indexed: 01/23/2023] Open
Abstract
Apolipoprotein A-I (apoA-I) mimetic peptide, D-4F, exhibits anti-atherogenic effects similar to high-density lipoprotein (HDL). However, it remains elusive whether D-4F and HDL share similar molecular mechanisms underlying anti-atherogenic effects and endothelial cell protections. We here compared the metabolic changes in endothelial cells induced by D-4F and HDL against oxidized low-density lipoprotein (ox-LDL), which may be of benefit to understanding the protective mechanisms of HDL and D-4F. Functional assays, including wound healing, transwell migration, and tube formation, were used to evaluate the pro-angiogenic effects of HDL and D-4F. NMR-based metabolomic analysis was employed to explore the protective mechanisms underlying HDL and D-4F. Partial least-squares discriminant analysis (PLS-DA) was performed to assess metabolic profiles, and orthogonal PLS-DA (OPLS-DA) was carried out to identify characteristic metabolites. Moreover, significantly altered metabolic pathways were also analyzed. We found that ox-LDL impaired the migration and tube formation of endothelial cells. Metabolomic analysis showed that ox-LDL triggered oxidative stress, impaired glycolysis, and enhanced glycerophospholipid metabolism. Both HDL and D-4F improved the migration and angiogenesis of endothelial cells, alleviated oxidative stress, and ameliorated disordered glycolysis impaired by ox-LDL. Strikingly, HDL partially attenuated the disturbed glycerophospholipid metabolism, whereas D-4F did not show this effect. In summary, although D-4F shared the similar protective effects with HDL on the migration and angiogenesis of endothelial cells, it could not deduce the molecular mechanisms of HDL completely. Nevertheless, D-4F possesses the potentiality to be exploited as clinically applicable agent for endothelial cell protection and cardiovascular disease treatment.
Collapse
Affiliation(s)
- Wenqi Xu
- Key Laboratory for Chemical Biology of Fujian Province, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, China
| | - Mingming Qian
- Department of Cardiology, The Affiliated Cardiovascular Hospital of Xiamen University, Medical College of Xiamen University, Xiamen, China
| | - Caihua Huang
- Exercise and Health Laboratory, Xiamen University of Technology, Xiamen, China
| | - Pengfei Cui
- Key Laboratory for Chemical Biology of Fujian Province, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, China
| | - Wei Li
- Department of Cardiology, The Affiliated Cardiovascular Hospital of Xiamen University, Medical College of Xiamen University, Xiamen, China
| | - Qian Du
- Department of Cardiology, The Affiliated Cardiovascular Hospital of Xiamen University, Medical College of Xiamen University, Xiamen, China
| | - Shenghui Yi
- Key Laboratory for Chemical Biology of Fujian Province, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, China
| | - Xiaohe Shi
- Key Laboratory for Chemical Biology of Fujian Province, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, China
| | - Yansong Guo
- Department of Cardiology, Fujian Provincial Hospital, Provincial Clinical Medicine College, Fujian Cardiovascular Institute, Fujian Provincial Key Laboratory of Cardiovascular Disease, Fujian Provincial Center for Geriatrics, Fujian Medical University, Fuzhou, China
| | - Jianlan Zheng
- Department of Ob/Gyn and Neonatal and Reproductive Medicine, The People's Liberation Army 174th Hospital and The Affiliated Hospital of Xiamen University, Xiamen, China
| | - Donghui Liu
- Department of Cardiology, The Affiliated Cardiovascular Hospital of Xiamen University, Medical College of Xiamen University, Xiamen, China.,Department of Cardiology, Fujian Provincial Hospital, Provincial Clinical Medicine College, Fujian Cardiovascular Institute, Fujian Provincial Key Laboratory of Cardiovascular Disease, Fujian Provincial Center for Geriatrics, Fujian Medical University, Fuzhou, China
| | - Donghai Lin
- Key Laboratory for Chemical Biology of Fujian Province, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, China
| |
Collapse
|
39
|
Trakaki A, Sturm GJ, Pregartner G, Scharnagl H, Eichmann TO, Trieb M, Knuplez E, Holzer M, Stadler JT, Heinemann A, Sturm EM, Marsche G. Allergic rhinitis is associated with complex alterations in high-density lipoprotein composition and function. Biochim Biophys Acta Mol Cell Biol Lipids 2019; 1864:1280-1292. [PMID: 31185305 DOI: 10.1016/j.bbalip.2019.06.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 05/17/2019] [Accepted: 06/05/2019] [Indexed: 12/20/2022]
Abstract
Despite strong evidence that high-density lipoproteins (HDLs) modulate the immune response, the role of HDL in allergies is still poorly understood. Many patients with allergic rhinitis (AR) develop a late-phase response, characterized by infiltration of monocytes and eosinophils into the nasal submucosa. Functional impairment of HDL in AR-patients may insufficiently suppress inflammation and cell infiltration, but the effect of AR on the composition and function of HDL is not understood. We used apolipoprotein (apo) B-depleted serum as well as isolated HDL from AR-patients (n = 43) and non-allergic healthy controls (n = 20) for detailed compositional and functional characterization of HDL. Both AR-HDL and apoB-depleted serum of AR-patients showed decreased anti-oxidative capacity and impaired ability to suppress monocyte nuclear factor-κB expression and pro-inflammatory cytokine secretion, such as interleukin (IL)-4, IL-6, IL-8, tumor necrosis factor alpha and IL-1 beta. Sera of AR-patients showed decreased paraoxonase and cholesteryl-ester transfer protein activities, increased lipoprotein-associated phospholipase A2 activity, while lecithin-cholesterol acyltransferase activity and cholesterol efflux capacity were not altered. Surprisingly, apoB-depleted serum and HDL from AR-patients showed an increased ability to suppress eosinophil effector responses upon eotaxin-2/CCL24 stimulation. Mass spectrometry and biochemical analyses showed reduced levels of apoA-I and phosphatidylcholine, but increased levels of apoA-II, triglycerides and lyso-phosphatidylcholine in AR-HDL. The changes in AR-HDL composition were associated with altered functional properties. In conclusion, AR alters HDL composition linked to decreased anti-oxidative and anti-inflammatory properties but improves the ability of HDL to suppress eosinophil effector responses.
Collapse
Affiliation(s)
- Athina Trakaki
- Division of Pharmacology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Universitätsplatz 4, 8010 Graz, Austria
| | - Gunter J Sturm
- Department of Dermatology and Venerology, Medical University of Graz, Auenbruggerplatz 8, 8036 Graz, Austria; Allergy Outpatient Clinic Reumannplatz, Vienna, Austria
| | - Gudrun Pregartner
- Institute of Medical Informatics, Statistics and Documentation, Medical University of Graz, Auenbruggerplatz 2/9/V, 8036 Graz, Austria
| | - Hubert Scharnagl
- Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
| | - Thomas O Eichmann
- Institute of Molecular Biosciences, University of Graz, Graz, Austria; Center for Explorative Lipidomics, BioTechMed-Graz, Graz, Austria
| | - Markus Trieb
- Division of Pharmacology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Universitätsplatz 4, 8010 Graz, Austria
| | - Eva Knuplez
- Division of Pharmacology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Universitätsplatz 4, 8010 Graz, Austria
| | - Michael Holzer
- Division of Pharmacology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Universitätsplatz 4, 8010 Graz, Austria
| | - Julia T Stadler
- Division of Pharmacology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Universitätsplatz 4, 8010 Graz, Austria
| | - Akos Heinemann
- Division of Pharmacology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Universitätsplatz 4, 8010 Graz, Austria.; BioTechMed Graz, Mozartgasse 12/II, 8010 Graz, Austria
| | - Eva M Sturm
- Division of Pharmacology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Universitätsplatz 4, 8010 Graz, Austria..
| | - Gunther Marsche
- Division of Pharmacology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Universitätsplatz 4, 8010 Graz, Austria.; BioTechMed Graz, Mozartgasse 12/II, 8010 Graz, Austria.
| |
Collapse
|
40
|
Abstract
PURPOSE OF REVIEW Plasma levels of HDL cholesterol are a biomarker of cardiovascular health but not a therapeutic target, as demonstrated by the failure of pharmacological modulation of HDL cholesterol to prevent or treat atherosclerotic cardiovascular disease. In health, HDL particles exert pleiotropic effects against atherosclerosis, including cholesterol removal from foam cells, vasodilatory effects through vascular endothelial cell nitric oxide production, decreased vascular inflammation and oxidative damage, endothelial cell proliferation and antiapoptotic effects. RECENT FINDINGS These functional effects of HDL are independent of the cholesterol mass and are related to the proteome and lipidome. In disease states and with the ageing process, HDL components are extensively modified and may no longer play a beneficial role but are retained in the atheroma and contribute to atherosclerosis. We have recently shown that desmocollin 1 (DSC1) acts as an apolipoprotein (apo) A-I binding protein that is highly expressed in atherosclerotic plaques and inhibits atheroprotective HDL functions by retaining apoA-I. The apoA-I retention hypothesis proposes that macrophages express DSC1 in a maladaptive process that renders apoA-I inactive and contributes to atherosclerosis. SUMMARY HDL loses their beneficial properties in ageing and disease states. Novel pathways may present new therapeutic avenues to restore their biological functions.
Collapse
|
41
|
Summerhill V, Karagodin V, Grechko A, Myasoedova V, Orekhov A. Vasculoprotective Role of Olive Oil Compounds via Modulation of Oxidative Stress in Atherosclerosis. Front Cardiovasc Med 2018; 5:188. [PMID: 30622950 PMCID: PMC6308304 DOI: 10.3389/fcvm.2018.00188] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 12/10/2018] [Indexed: 12/14/2022] Open
Abstract
Existing evidence supports the significant role of oxidative stress in the endothelial injury, and there is a direct link between increased oxidative stress, and the development of endothelial dysfunction. Endothelial dysfunction precedes the development of atherosclerosis and subsequent cardiovascular disease (CVD). The overproduction of reactive oxygen species facilitates the processes, such as oxidative modification of low-density lipoproteins and phospholipids, reduction in the NOS-derived nitric oxide, and the functional disruption of high-density lipids that are profoundly involved in atherogenesis, inflammation, and thrombus formation in vascular cells. Thus, under oxidative stress conditions, endothelial dysfunction was found to be associated with the following endothelial alterations: reduced nitric oxide bioavailability, increased anticoagulant properties, increased platelet aggregation, increased expression of adhesion molecules, chemokines, and cytokines. In this review, we summarized the evidence indicating that endothelial damage triggered by oxidation can be diminished or reversed by the compounds of olive oil, a readily available antioxidant food source. Olive oil bioactive compounds exhibited a potent capability to attenuate oxidative stress and improve endothelial function through their anti-inflammatory, anti-oxidant, and anti-thrombotic properties, therefore reducing the risk and progression of atherosclerosis. Also, their molecular mechanisms of action were explored to establish the potential preventive and/or therapeutic alternatives to the pharmacological remedies available.
Collapse
Affiliation(s)
- Volha Summerhill
- Skolkovo Innovative Center, Institute for Atherosclerosis Research Moscow, Moscow, Russia
| | - Vasilyi Karagodin
- Department of Commodity Research and Expertise, Plekhanov Russian University of Economics, Moscow, Russia
| | - Andrey Grechko
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, Moscow, Russia
| | - Veronika Myasoedova
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - Alexander Orekhov
- Skolkovo Innovative Center, Institute for Atherosclerosis Research Moscow, Moscow, Russia
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Moscow, Russia
| |
Collapse
|
42
|
Morin EE, Li XA, Schwendeman A. HDL in Endocrine Carcinomas: Biomarker, Drug Carrier, and Potential Therapeutic. Front Endocrinol (Lausanne) 2018; 9:715. [PMID: 30555417 PMCID: PMC6283888 DOI: 10.3389/fendo.2018.00715] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Accepted: 11/12/2018] [Indexed: 12/14/2022] Open
Abstract
High-density lipoprotein (HDL) have long been studied for their protective role against cardiovascular diseases, however recently relationship between HDL and cancer came into focus. Several epidemiological studies have shown an inverse correlation between HDL-cholesterol (HDL-C) and cancer risk, and some have even implied that HDL-C can be used as a predictive measure for survival prognosis in for specific sub-population of certain types of cancer. HDL itself is an endogenous nanoparticle capable of removing excess cholesterol from the periphery and returning it to the liver for excretion. One of the main receptors for HDL, scavenger receptor type B-I (SR-BI), is highly upregulated in endocrine cancers, notably due to the high demand for cholesterol by cancer cells. Thus, the potential to exploit administration of cholesterol-free reconstituted or synthetic HDL (sHDL) to deplete cholesterol in endocrine cancer cell and stunt their growth of use chemotherapeutic drug loaded sHDL to target payload delivery to cancer cell has become increasingly attractive. This review focuses on the role of HDL and HDL-C in cancer and application of sHDLs as endocrine cancer therapeutics.
Collapse
Affiliation(s)
- Emily E. Morin
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, United States
- BioInterfaces Institute, University of Michigan, Ann Arbor, MI, United States
| | - Xiang-An Li
- Department of Physiology, Saha Cardiovascular Research Center, College of Medicine, University of Kentucky, Lexington, KY, United States
| | - Anna Schwendeman
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, United States
- BioInterfaces Institute, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
43
|
Maternal supraphysiological hypercholesterolemia associates with endothelial dysfunction of the placental microvasculature. Sci Rep 2018; 8:7690. [PMID: 29769708 PMCID: PMC5955926 DOI: 10.1038/s41598-018-25985-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 04/27/2018] [Indexed: 01/24/2023] Open
Abstract
Maternal physiological or supraphysiological hypercholesterolemia (MPH, MSPH) occurs during pregnancy. MSPH is associated with foetal endothelial dysfunction and atherosclerosis. However, the potential effects of MSPH on placental microvasculature are unknown. The aim of this study was to determine whether MSPH alters endothelial function in the placental microvasculature both ex vivo in venules and arterioles from the placental villi and in vitro in primary cultures of placental microvascular endothelial cells (hPMEC). Total cholesterol < 280 mg/dL indicated MPH, and total cholesterol ≥280 mg/dL indicated MSPH. The maximal relaxation to histamine, calcitonin gene-related peptide and adenosine was reduced in MSPH venule and arteriole rings. In hPMEC from MSPH placentas, nitric oxide synthase (NOS) activity and L-arginine transport were reduced without changes in arginase activity or the protein levels of endothelial NOS (eNOS), human cationic amino acid 1 (hCAT-1), hCAT-2A/B or arginase II compared with hPMEC from MPH placentas. In addition, it was shown that adenosine acts as a vasodilator of the placental microvasculature and that NOS is active in hPMEC. We conclude that MSPH alters placental microvascular endothelial function via a NOS/L-arginine imbalance. This work also reinforces the concept that placental endothelial cells from the macro- and microvasculature respond differentially to the same pathological condition.
Collapse
|
44
|
Abstract
The systematic study of nanoparticle-biological interactions requires particles to be reproducibly dispersed in relevant fluids along with further development in the identification of biologically relevant structural details at the materials-biology interface. Here, we develop a biocompatible long-term colloidally stable water dispersion of few-layered graphene nanoflakes in the biological exposure medium in which it will be studied. We also report the study of the orientation and functionality of key proteins of interest in the biolayer (corona) that are believed to mediate most of the early biological interactions. The evidence accumulated shows that graphene nanoflakes are rich in effective apolipoprotein A-I presentation, and we are able to map specific functional epitopes located in the C-terminal portion that are known to mediate the binding of high-density lipoprotein to binding sites in receptors that are abundant in the liver. This could suggest a way of connecting the materials' properties to the biological outcomes.
Collapse
|
45
|
Detecting the functional complexities between high-density lipoprotein mimetics. Biomaterials 2018; 170:58-69. [PMID: 29653287 DOI: 10.1016/j.biomaterials.2018.04.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 04/03/2018] [Accepted: 04/05/2018] [Indexed: 01/05/2023]
Abstract
High-density lipoprotein (HDL) is a key regulator of lipid homeostasis through its native roles like reverse cholesterol transport. The reconstitution of this natural nanoparticle (NP) has become a nexus between nanomedicine and multi-disease therapies, for which a major portion of HDL functionality is attributed to its primary scaffolding protein, apolipoprotein A1 (apoA1). ApoA1-mimetic peptides were formulated as cost-effective alternatives to apoA1-based therapies; reverse-4F (r4F) is one such peptide used as part of a nanoparticle platform. While similarities between r4F- and apoA1-based HDL-mimetic nanoparticles have been identified, key functional differences native to HDL have remained undetected. In the present study, we executed a multidisciplinary approach to uncover these differences by exploring the form, function, and medical applicability of engineered HDL-mimetic NPs (eHNPs) made from r4F (eHNP-r4F) and from apoA1 (eHNP-A1). Comparative analyses of the eHNPs through computational molecular dynamics (MD), advanced microfluidic NP synthesis and screening technologies, and in vivo animal model studies extracted distinguishable eHNP characteristics: the eHNPs share identical structural and compositional characteristics with distinct differences in NP stability and organization; eHNP-A1 could more significantly stimulate anti-inflammatory responses characteristic of the scavenger receptor class B type 1 (SR-B1) mediated pathways; and eHNP-A1 could outperform eHNP-r4F in the delivery of a model hydrophobic drug to an in vivo tumor. The biomimetic microfluidic technologies and MD simulations uniquely enabled our comparative analysis through which we determined that while eHNP-r4F is a capable NP with properties mimicking natural eHNP-A1, challenges remain in reconstituting the full functionality of NPs naturally derived from humans.
Collapse
|
46
|
Vaisar T, Couzens E, Hwang A, Russell M, Barlow CE, DeFina LF, Hoofnagle AN, Kim F. Type 2 diabetes is associated with loss of HDL endothelium protective functions. PLoS One 2018; 13:e0192616. [PMID: 29543843 PMCID: PMC5854245 DOI: 10.1371/journal.pone.0192616] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 01/27/2018] [Indexed: 11/18/2022] Open
Abstract
Aims/Hypothesis One of the hallmarks of diabetes is impaired endothelial function. Previous studies showed that HDL can exert protective effects on endothelium stimulating NO production and protecting from inflammation and suggested that HDL in obese people with diabetes and dyslipidemia may have lower endothelial protective function. We aimed to investigate whether type 2 diabetes impairs HDL endothelium protective functions in people with otherwise normal lipid profile. Methods In a case-control study (n = 41 per group) nested in the Cooper Center Longitudinal Study we tested the ability of HDL to protect endothelium by stimulating endothelial nitric oxide synthase activity and suppressing NFκB-mediated inflammatory response in endothelial cells. In parallel we measured HDL protein composition, sphinogosine-1-phosphate and P-selectin. Results Despite similar levels of plasma HDL-C the HDL in individuals with type 2 diabetes lost almost 40% of its ability to stimulate eNOS activity (P<0.001) and 20% of its ability to suppress TNFα-dependent NFκB-mediated inflammatory response in endothelial cells (P<0.001) compared to non-T2D controls despite similar BMI and lipid profile (HDL-C, LDL-C, TC, TG). Significantly, the ability of HDL to stimulate eNOS activity was negatively associated with plasma levels of P-selectin, an established marker of endothelial dysfunction (r = −0.32, P<0.001). Furthermore, sphingosine-1-phosphate (S1P) levels were decreased in diabetic plasma (P = 0.017) and correlated with HDL-mediated eNOS activation. Conclusions/Interpretations Collectively, our data suggest that HDL in individuals with type 2 diabetes loses its ability to maintain proper endothelial function independent of HDL-C, perhaps due to loss of S1P, and may contribute to development of diabetic complications.
Collapse
Affiliation(s)
- Tomáš Vaisar
- UW Medicine Diabetes Institute, Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Erica Couzens
- UW Medicine Diabetes Institute, Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Arnold Hwang
- UW Medicine Diabetes Institute, Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Michael Russell
- UW Medicine Diabetes Institute, Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | | | - Laura F DeFina
- The Cooper Institute, Dallas, Texas, United States of America
| | - Andrew N Hoofnagle
- Department of Laboratory Medicine, University of Washington, Seattle, Washington, United States of America
| | - Francis Kim
- UW Medicine Diabetes Institute, Department of Medicine, University of Washington, Seattle, Washington, United States of America
| |
Collapse
|
47
|
High-density lipoprotein (HDL) promotes angiogenesis via S1P3-dependent VEGFR2 activation. Angiogenesis 2018; 21:381-394. [PMID: 29450744 DOI: 10.1007/s10456-018-9603-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 02/06/2018] [Indexed: 02/05/2023]
Abstract
High-density lipoprotein (HDL) has previously been shown to promote angiogenesis. However, the mechanisms by which HDL enhances the formation of blood vessels remain to be defined. To address this, the effects of HDL on the proliferation, transwell migration and tube formation of human umbilical vein endothelial cells were investigated. By examining the abundance and phosphorylation (i.e., activation) of the vascular endothelial growth factor receptor VEGFR2 and modulating the activity of the sphingosine-1 phosphate receptors S1P1-3 and VEGFR2, we characterized mechanisms controlling angiogenic responses in response to HDL exposure. Here, we report that HDL dose-dependently increased endothelial proliferation, migration and tube formation. These events were in association with increased VEGFR2 abundance and rapid VEGFR2 phosphorylation at Tyr1054/Tyr1059 and Tyr1175 residues in response to HDL. Blockade of VEGFR2 activation by the VEGFR2 inhibitor SU1498 markedly abrogated the pro-angiogenic capacity of HDL. Moreover, the S1P3 inhibitor suramin prevented VEGFR2 expression and abolished endothelial migration and tube formation, while the S1P1 agonist CYM-5442 and the S1P2 inhibitor JTE-013 had no effect. Last, the role of S1P3 was further confirmed in regulation of S1P-induced endothelial proliferation, migration and tube formation via up-regulation and activation of VEGFR2. Together, these findings argue that HDL promotes angiogenesis via S1P3-dependent up-regulation and activation of VEGFR2 and also suggest that the S1P-S1P3-VEGFR2 signaling cascades as a novel target for HDL-modulating therapy implicated in vascular remodeling and functional recovery in atherosclerotic diseases such as myocardial infarction and ischemic stroke.
Collapse
|
48
|
Ganjali S, Momtazi AA, Banach M, Kovanen PT, Stein EA, Sahebkar A. HDL abnormalities in familial hypercholesterolemia: Focus on biological functions. Prog Lipid Res 2017; 67:16-26. [DOI: 10.1016/j.plipres.2017.05.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 04/25/2017] [Accepted: 05/10/2017] [Indexed: 02/07/2023]
|
49
|
Shinnakasu A, Yamamoto K, Kurano M, Arimura H, Arimura A, Kikuti A, Hashiguchi H, Deguchi T, Nishio Y. The Combination Therapy of Fenofibrate and Ezetimibe Improved Lipid Profile and Vascular Function Compared with Statins in Patients with Type 2 Diabetes. J Atheroscler Thromb 2017; 24:735-748. [PMID: 28450679 PMCID: PMC5517547 DOI: 10.5551/jat.39446] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
AIM Elevated level of serum triglyceride (TG) is a characteristic of type 2 diabetes. We evaluated the clinical significance of intervention for the serum TG levels in the fasting and postprandial states in patients with type 2 diabetes. METHODS Fifty patients with type 2 diabetes, treated with statins, were selected and divided into two groups. One group was treated with a combination of fenofibrate and ezetimibe (F/E group) and the other group with statins (statin group) for 12 weeks. The lipoprotein profile of both groups was compared using high-performance liquid chromatography, and the vascular function was assessed using flow-mediated dilation (FMD) at the forearm. RESULTS The levels of very low-density lipoprotein (VLDL) cholesterol, malondialdehyde low-density lipoprotein (MDA-LDL), total TG, chylomicron-TG, VLDL-TG, and HDL-TG decreased in the F/E group, whereas those of HDL cholesterol increased. Furthermore, the peak particle size of LDL increased, but that of HDL decreased in the F/E group. The combination treatment significantly improved the FMD. The change in the cholesterol level in a very small fraction of HDL was a significant independent predictor for determining the improvement of FMD (p<0.01). CONCLUSIONS Compared with the treatment with statins, the treatment with the combination of fenofibrate and ezetimibe effectively controlled the LDL cholesterol and TG levels, increased the HDL cholesterol level, especially in its small fraction, and improved vascular function of patients with type 2 diabetes.
Collapse
Affiliation(s)
- Atsushi Shinnakasu
- Department of Diabetes and Endocrine Medicine, Kagoshima University Graduate School of Medical and Dental Sciences
| | - Kiyoaki Yamamoto
- Department of Diabetes and Endocrine Medicine, Kagoshima University Graduate School of Medical and Dental Sciences
| | - Mihoko Kurano
- Department of Diabetes and Endocrine Medicine, Kagoshima University Graduate School of Medical and Dental Sciences
| | - Hiroshi Arimura
- Department of Diabetes and Endocrine Medicine, Kagoshima University Graduate School of Medical and Dental Sciences
| | - Aiko Arimura
- Department of Diabetes and Endocrine Medicine, Kagoshima University Graduate School of Medical and Dental Sciences
| | - Akira Kikuti
- Department of Diabetes and Endocrine Medicine, Kagoshima University Graduate School of Medical and Dental Sciences
| | - Hiroshi Hashiguchi
- Department of Diabetes and Endocrine Medicine, Kagoshima University Graduate School of Medical and Dental Sciences
| | - Takahisa Deguchi
- Department of Diabetes and Endocrine Medicine, Kagoshima University Graduate School of Medical and Dental Sciences
| | - Yoshihiko Nishio
- Department of Diabetes and Endocrine Medicine, Kagoshima University Graduate School of Medical and Dental Sciences
| |
Collapse
|
50
|
Li J, Tan M, Xiang Q, Zhou Z, Yan H. Thrombin-activated platelet-derived exosomes regulate endothelial cell expression of ICAM-1 via microRNA-223 during the thrombosis-inflammation response. Thromb Res 2017; 154:96-105. [PMID: 28460288 DOI: 10.1016/j.thromres.2017.04.016] [Citation(s) in RCA: 131] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 04/09/2017] [Accepted: 04/16/2017] [Indexed: 12/19/2022]
Abstract
Platelet activation and endothelial damage play essential roles in atherosclerosis. The pathophysiology of this process is mediated by chemokines and exosomes, two critical players in cell communication. Thrombin-activated platelet-derived exosomes have protective effects on atherosclerosis and endothelial inflammation. To confirm these findings, we isolated exosomes using differential ultracentrifugation and transmission electron microscopy. The exosomes were identified using NanoSight-tracking analysis. Immunofluorescence staining and western blotting were performed to assess exosome uptake and intercellular adhesion molecule-1 (ICAM-1) expression in human umbilical vein endothelial cells (HUVECs). We found that the levels of miR-223, miR-339 and miR-21 were elevated in thrombin-activated platelet exosomes. This finding was verified in our atherosclerosis mouse model. We also found that miR-223 transfection in HUVECs inhibited ICAM-1 expression under TNF-α stimulation. Furthermore, the miR-223 inhibitor blocked the downregulating effects of exosomes on ICAM-1 expression. We examined the key proteins of two classical signaling pathways, MAPK and NF-κB, and found that miR-223 inhibited the phosphorylation of p38, JNK and ERK and blocked the nuclear translocation of NF-κB p65. Our results suggest that thrombin-activated platelet-derived exosomes inhibit ICAM-1 expression during inflammation. MiR-223 may mediate this process via regulation of the NF-κB and MAPK pathways.
Collapse
Affiliation(s)
- Jiannan Li
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ming Tan
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qinqin Xiang
- State Key Laboratory of Cardiovascular Disease, Beijing Key Laboratory for Molecular Diagnostics of Cardiovascular Diseases, Diagnostic Laboratory Service, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhou Zhou
- State Key Laboratory of Cardiovascular Disease, Beijing Key Laboratory for Molecular Diagnostics of Cardiovascular Diseases, Diagnostic Laboratory Service, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Hongbing Yan
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|