51
|
Chin DD, Poon C, Wang J, Joo J, Ong V, Jiang Z, Cheng K, Plotkin A, Magee GA, Chung EJ. miR-145 micelles mitigate atherosclerosis by modulating vascular smooth muscle cell phenotype. Biomaterials 2021; 273:120810. [PMID: 33892346 PMCID: PMC8152375 DOI: 10.1016/j.biomaterials.2021.120810] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 03/31/2021] [Accepted: 04/04/2021] [Indexed: 12/14/2022]
Abstract
In atherosclerosis, resident vascular smooth muscle cells (VSMCs) in the blood vessels become highly plastic and undergo phenotypic switching from the quiescent, contractile phenotype to the migratory and proliferative, synthetic phenotype. Additionally, recent VSMC lineage-tracing mouse models of atherosclerosis have found that VSMCs transdifferentiate into macrophage-like and osteochondrogenic cells and make up to 70% of cells found in atherosclerotic plaques. Given VSMC phenotypic switching is regulated by microRNA-145 (miR-145), we hypothesized that nanoparticle-mediated delivery of miR-145 to VSMCs has the potential to mitigate atherosclerosis development by inhibiting plaque-propagating cell types derived from VSMCs. To test our hypothesis, we synthesized miR-145 micelles targeting the C-C chemokine receptor-2 (CCR2), which is highly expressed on synthetic VSMCs. When miR-145 micelles were incubated with human aortic VSMCs in vitro, >90% miR-145 micelles escaped the lysosomal pathway in 4 hours and released the miR cargo under cytosolic levels of glutathione, an endogenous reducing agent. As such, miR-145 micelles rescued atheroprotective contractile markers, myocardin, α-SMA, and calponin, in synthetic VSMCs in vitro. In early-stage atherosclerotic ApoE-/- mice, one dose of miR-145 micelles prevented lesion growth by 49% and sustained an increased level of miR-145 expression after 2 weeks post-treatment. Additionally, miR-145 micelles inhibited 35% and 43% plaque growth compared to free miR-145 and PBS, respectively, in mid-stage atherosclerotic ApoE-/- mice. Collectively, we present a novel therapeutic strategy and cell target for atherosclerosis, and present miR-145 micelles as a viable nanotherapeutic that can intervene atherosclerosis progression at both early and later stages of disease.
Collapse
Affiliation(s)
- Deborah D Chin
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, 90089, United States
| | - Christopher Poon
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, 90089, United States
| | - Jonathan Wang
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, 90089, United States
| | - Johan Joo
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, 90089, United States
| | - Victor Ong
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, 90089, United States
| | - Zhangjingyi Jiang
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, 90089, United States
| | - Kayley Cheng
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, 90089, United States
| | - Anastasia Plotkin
- Division of Vascular Surgery and Endovascular Therapy, Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, United States
| | - Gregory A Magee
- Division of Vascular Surgery and Endovascular Therapy, Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, United States
| | - Eun Ji Chung
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, 90089, United States; Division of Vascular Surgery and Endovascular Therapy, Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, United States; Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA, 90089, United States; Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, United States; Division of Nephrology and Hypertension, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, United States; Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90089, United States.
| |
Collapse
|
52
|
Zhao L, Zhang S, Su Q, Li S. Effects of withdrawing an atherogenic diet on the atherosclerotic plaque in rabbits. Exp Ther Med 2021; 22:751. [PMID: 34035848 PMCID: PMC8135140 DOI: 10.3892/etm.2021.10183] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 12/11/2020] [Indexed: 02/05/2023] Open
Abstract
Lifestyle interventions and pharmacotherapy are the most common of non-invasive treatments for atherosclerosis, but the individual effect of diet on plaques remains unclear. The current study aimed to investigate the effect of withdrawing the atherogenic diet on plaque in the aortas of rabbits. Experimental atheroma was induced in 33 rabbits using a 1% high cholesterol diet for 30 days (H-30 d) or 90 days (H-90 d, baseline group). After 90 days of the atherogenic diet, the remaining animals were divided into four groups: A total of 10 rabbits continued to consume the atherogenic diet for 50 days (H-90 d & H-50 d; n=5) or 140 days (H-90 d & H-140 d; n=5). Another 13 rabbits were switched to a chow diet for 50 days (H-90 d & C-50 d; n=7) or 140 days (H-90 d & C-140 d; n=6). A total of 10 age-matched rabbits in the control groups were fed a chow diet for 90 and 230 days, respectively. The en face or cross-sectional plaque areas were determined using oil red O staining and elastic van Gieson staining. Immunohistochemistry analyses were used to assess the macrophages or smooth muscle cell contents. When fed an atherogenic diet for 90 days, the rabbits' abdominal aortas exhibited severe atherosclerotic lesions (the median en face plaque area was 63.6%). After withdrawing the atherogenic diet, the plaque area did not shrink with feeding the chow diet compared with the baseline, but increased to 71.8 or 80.5% after 50 or 140 days, respectively. After removing cholesterol from the diet, the lipids content in the plaques increased during the first 50 days, and then decreased compared with the baseline group. Furthermore, withdrawing the atherogenic diet increased the total collagen content and the percentage of the smooth muscle cells, alleviated macrophage infiltration, decreased the vulnerable index and promoted the cross-linking of collagen. Feeding the rabbits an atherogenic diet followed by removal of cholesterol from the diet did not lead to the regression of established lesions but instead delayed the progression of the lesions and promoted the stabilization of the plaque.
Collapse
Affiliation(s)
- Lijun Zhao
- Department of General Practice, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Shifang Zhang
- Department of Pulmonary Disease, Institute of Respiratory Disease, Chengdu Second People's Hospital, Chengdu, Sichuan 610000, P.R. China
| | - Qiaoli Su
- Department of General Practice, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Shuangqing Li
- Department of General Practice, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
53
|
Lee CL, Lee JW, Daniel AR, Holbrook M, Hasapis S, Wright AO, Brownstein J, Da Silva Campos L, Ma Y, Mao L, Abraham D, Badea CT, Kirsch DG. Characterization of cardiovascular injury in mice following partial-heart irradiation with clinically relevant dose and fractionation. Radiother Oncol 2021; 157:155-162. [DOI: 10.1016/j.radonc.2021.01.023] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 01/14/2021] [Accepted: 01/15/2021] [Indexed: 12/16/2022]
|
54
|
Sun Y, Wu D, Zeng W, Chen Y, Guo M, Lu B, Li H, Sun C, Yang L, Jiang X, Gao Q. The Role of Intestinal Dysbacteriosis Induced Arachidonic Acid Metabolism Disorder in Inflammaging in Atherosclerosis. Front Cell Infect Microbiol 2021; 11:618265. [PMID: 33816331 PMCID: PMC8012722 DOI: 10.3389/fcimb.2021.618265] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 01/28/2021] [Indexed: 12/12/2022] Open
Abstract
Background Aging induced chronic systemic inflammatory response is an important risk factor for atherosclerosis (AS) development; however, the detailed mechanism is yet to be elucidated. Objective To explore the underlying mechanism of how aging aggravates AS advancement. Methods A young (five-week-old, YM) and aged group (32-week-old, OM) male apoE-/- mice with a high fat diet were used as models, and age-matched male wild-type C57BL/6J (WT) mice were used as controls. AS lesion size, serum lipid profile, cytokines, and gut microbiota-derived LPS were analyzed after 32 weeks of diet intervention. A correlation analysis between the 16S rRNA sequencing of the feces and serum metabolomics profiles was applied to examine the effect of their interactions on AS. Results ApoE-/- mice developed severe atherosclerosis and inflammation in the aorta compared to the WT groups, and aged apoE-/- mice suffered from a more severe AS lesion than their younger counterparts and had low-grade systemic inflammation. Furthermore, increased levels of serum LPS, decreased levels of SCFAs production, as well as dysfunction of the ileal mucosal barrier were detected in aged mice compared with their younger counterparts. There were significant differences in the intestinal flora composition among the four groups, and harmful bacteria such as Lachnospiraceae_FCS020, Ruminococcaceae_UCG-009, Acetatifactor, Lachnoclostridium and Lactobacillus_gasseri were significantly increased in the aged apoE-/- mice compared with the other groups. Concurrently, metabolomics profiling revealed that components involved in the arachidonic acid (AA) metabolic pathway such as 20-HETE, PGF2α, arachidonic acid, and LTB4 were significantly higher in the aged AS group than in the other groups. This suggested that metabolic abnormalities and disorders of intestinal flora occurred in AS mice. Conclusions Aging not only altered the gut microbiome community but also substantially disturbed metabolic conditions. Our results confirm that AA metabolism is associated with the imbalance of the intestinal flora in the AS lesions of aged mice. These findings may offer new insights regarding the role of gut flora disorders and its consequent metabolite changed in inflammaging during AS development.
Collapse
Affiliation(s)
- Yingxin Sun
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Danbin Wu
- Department of Rheumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wenyun Zeng
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yefei Chen
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, China
| | - Maojuan Guo
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Bin Lu
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Huhu Li
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Chun Sun
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lin Yang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xijuan Jiang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Qing Gao
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
55
|
Xiao H, Chen J, Duan L, Li S. Role of emerging vitamin K‑dependent proteins: Growth arrest‑specific protein 6, Gla‑rich protein and periostin (Review). Int J Mol Med 2021; 47:2. [PMID: 33448308 PMCID: PMC7834955 DOI: 10.3892/ijmm.2020.4835] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 10/21/2020] [Indexed: 01/27/2023] Open
Abstract
Vitamin K‑dependent proteins (VKDPs) are a group of proteins that need vitamin K to conduct carboxylation. Thus far, scholars have identified a total of 17 VKDPs in the human body. In this review, we summarize three important emerging VKDPs: Growth arrest‑specific protein 6 (Gas 6), Gla‑rich protein (GRP) and periostin in terms of their functions in physiological and pathological conditions. As examples, carboxylated Gas 6 and GRP effectively protect blood vessels from calcification, Gas 6 protects from acute kidney injury and is involved in chronic kidney disease, GRP contributes to bone homeostasis and delays the progression of osteoarthritis, and periostin is involved in all phases of fracture healing and assists myocardial regeneration in the early stages of myocardial infarction. However, periostin participates in the progression of cardiac fibrosis, idiopathic pulmonary fibrosis and airway remodeling of asthma. In addition, we discuss the relationship between vitamin K, VKDPs and cancer, and particularly the carboxylation state of VKDPs in cancer.
Collapse
Affiliation(s)
- Huiyu Xiao
- Department of Physiology, Dalian Medical University, Dalian, Liaoning 116044
| | - Jiepeng Chen
- Sungen Bioscience Co., Ltd., Shantou, Guangdong 515071, P.R. China
| | - Lili Duan
- Sungen Bioscience Co., Ltd., Shantou, Guangdong 515071, P.R. China
| | - Shuzhuang Li
- Department of Physiology, Dalian Medical University, Dalian, Liaoning 116044
| |
Collapse
|
56
|
van den Brule S, Rappe M, Ambroise J, Bouzin C, Dessy C, Paquot A, Muccioli GG, Lison D. Diesel exhaust particles alter the profile and function of the gut microbiota upon subchronic oral administration in mice. Part Fibre Toxicol 2021; 18:7. [PMID: 33563307 PMCID: PMC7871568 DOI: 10.1186/s12989-021-00400-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 01/29/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Ambient air pollution by particulate matters, including diesel exhaust particles (DEP), is a major cause of cardiovascular and metabolic mortality worldwide. The mechanisms by which DEP cause these adverse outcomes are not completely understood. Because the gut microbiota controls cardiovascular and metabolic health, we hypothesized that the fraction of inhaled DEP which reach the gut after mucociliary clearance and swallowing might induce gut dysbiosis and, in turn, contribute to aggravate or induce cardiovascular and metabolic diseases. RESULTS Female ApoE-/- mice fed a Western diet, and wild-type (C57Bl/6) mice fed standard diet were gavaged with DEP (SRM2975) doses corresponding to mucociliary clearance from inhalation exposure (200 or 1000 ng/day, 3 times a week for 3 months; and 40, 200 or 1000 ng/day, 3 times a week for 6 months, respectively). No mortality, overt systemic or digestive toxicity was observed. A dose-dependent alteration of the gut microbiota was recorded in both strains. In ApoE-/-, β-diversity was modified by DEP, but no significant modification of the relative abundance of the phyla, families or genera was identified. In C57BL/6 mice, DEP reduced α-diversity (Shannon and Simpson indices), and modified β-diversity, including a reduction of the Proteobacteria and Patescibacteria phyla, and an increase of the Campylobacterota phylum. In both mouse models, perturbation of the gut microbiota composition was associated with a dose-dependent reduction of bacterial short chain fatty acids (butyrate and propionate) in cecal content. However, DEP ingestion did not aggravate (ApoE-/-), or induce (C57BL/6 mice) atherosclerotic plaques, and no metabolic alteration (glucose tolerance, resistance to insulin, or lipidemia) was recorded. CONCLUSIONS We show here that oral exposure to DEP, at doses relevant for human health, changes the composition and function of the gut microbiota. These modifications were, however, not translated into ultimate atherosclerotic or metabolic outcomes.
Collapse
Affiliation(s)
- Sybille van den Brule
- Louvain centre for Toxicology and Applied Pharmacology, Institut de Recherche Expérimentale et Clinique, UCLouvain, Brussels, Belgium
| | - Margaux Rappe
- Louvain centre for Toxicology and Applied Pharmacology, Institut de Recherche Expérimentale et Clinique, UCLouvain, Brussels, Belgium
| | - Jérôme Ambroise
- Centre de Technologies Moléculaires Appliquées, Institut de Recherche Expérimentale et Clinique, UCLouvain, Brussels, Belgium
| | - Caroline Bouzin
- IREC Imaging Platform (2IP), Institut de Recherche Expérimentale et Clinique, UCLouvain, Brussels, Belgium
| | - Chantal Dessy
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique, UCLouvain and Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Adrien Paquot
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, UCLouvain, Brussels, Belgium
| | - Giulio G Muccioli
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, UCLouvain, Brussels, Belgium
| | - Dominique Lison
- Louvain centre for Toxicology and Applied Pharmacology, Institut de Recherche Expérimentale et Clinique, UCLouvain, Brussels, Belgium.
| |
Collapse
|
57
|
Demina EP, Smutova V, Pan X, Fougerat A, Guo T, Zou C, Chakraberty R, Snarr BD, Shiao TC, Roy R, Orekhov AN, Miyagi T, Laffargue M, Sheppard DC, Cairo CW, Pshezhetsky AV. Neuraminidases 1 and 3 Trigger Atherosclerosis by Desialylating Low-Density Lipoproteins and Increasing Their Uptake by Macrophages. J Am Heart Assoc 2021; 10:e018756. [PMID: 33554615 PMCID: PMC7955353 DOI: 10.1161/jaha.120.018756] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Background Chronic vascular disease atherosclerosis starts with an uptake of atherogenic modified low-density lipoproteins (LDLs) by resident macrophages, resulting in formation of arterial fatty streaks and eventually atheromatous plaques. Increased plasma sialic acid levels, increased neuraminidase activity, and reduced sialic acid LDL content have been previously associated with atherosclerosis and coronary artery disease in human patients, but the mechanism underlying this association has not been explored. Methods and Results We tested the hypothesis that neuraminidases contribute to development of atherosclerosis by removing sialic acid residues from glycan chains of the LDL glycoprotein and glycolipids. Atherosclerosis progression was investigated in apolipoprotein E and LDL receptor knockout mice with genetic deficiency of neuraminidases 1, 3, and 4 or those treated with specific neuraminidase inhibitors. We show that desialylation of the LDL glycoprotein, apolipoprotein B 100, by human neuraminidases 1 and 3 increases the uptake of human LDL by human cultured macrophages and by macrophages in aortic root lesions in Apoe-/- mice via asialoglycoprotein receptor 1. Genetic inactivation or pharmacological inhibition of neuraminidases 1 and 3 significantly delays formation of fatty streaks in the aortic root without affecting the plasma cholesterol and LDL levels in Apoe-/- and Ldlr-/- mouse models of atherosclerosis. Conclusions Together, our results suggest that neuraminidases 1 and 3 trigger the initial phase of atherosclerosis and formation of aortic fatty streaks by desialylating LDL and increasing their uptake by resident macrophages.
Collapse
Affiliation(s)
- Ekaterina P Demina
- Departments of Pediatrics and Biochemistry Sainte-Justine University Hospital Research CenterUniversity of Montreal Quebec Canada
| | - Victoria Smutova
- Departments of Pediatrics and Biochemistry Sainte-Justine University Hospital Research CenterUniversity of Montreal Quebec Canada
| | - Xuefang Pan
- Departments of Pediatrics and Biochemistry Sainte-Justine University Hospital Research CenterUniversity of Montreal Quebec Canada
| | - Anne Fougerat
- Departments of Pediatrics and Biochemistry Sainte-Justine University Hospital Research CenterUniversity of Montreal Quebec Canada
| | - Tianlin Guo
- Department of Chemistry University of Alberta Edmonton Alberta Canada
| | - Chunxia Zou
- Department of Chemistry University of Alberta Edmonton Alberta Canada
| | | | - Brendan D Snarr
- Departments of Medicine, Microbiology and Immunology McGill University Montreal Quebec Canada
| | - Tze C Shiao
- Department of Chemistry Université du Québec à Montréal Montreal Quebec Canada
| | - Rene Roy
- Department of Chemistry Université du Québec à Montréal Montreal Quebec Canada
| | | | - Taeko Miyagi
- Miyagi Cancer Center Research Institute Natori Miyagi Japan
| | - Muriel Laffargue
- Institut National de la Santé et de la Recherche MédicaleUMR 1048Institute of Metabolic and Cardiovascular Diseases Toulouse France
| | - Donald C Sheppard
- Departments of Medicine, Microbiology and Immunology McGill University Montreal Quebec Canada
| | | | - Alexey V Pshezhetsky
- Departments of Pediatrics and Biochemistry Sainte-Justine University Hospital Research CenterUniversity of Montreal Quebec Canada
| |
Collapse
|
58
|
Shemesh D, Rozenberg K, Rosenzweig T, Abookasis D. Single probe diffuse reflectance spectroscopy to assess the effect of sarcopoterium spinosum treatment on the cerebral tissue properties of ApoE knockout mouse. JOURNAL OF BIOPHOTONICS 2021; 14:e202000307. [PMID: 33084182 DOI: 10.1002/jbio.202000307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 10/11/2020] [Accepted: 10/13/2020] [Indexed: 06/11/2023]
Abstract
In this work, diffuse near-infrared light reflectance spectroscopy based on a single optical probe, contains central single collection fiber surrounded by a circular array of illumination fibers, was used to quantify cerebral tissue properties in ApoE knockout mice following Sarcopoterium spinosum treatment. Sarcopoterium spinosum, also known as Thorny burnet, is a Mediterranean plant widely used as a traditional therapy for the treatment of a variety of pathologies, primarily type 2 diabetes mellitus (T2D). While it's efficacy in the treatment of T2D, and of other components of metabolic syndrome, have already been validated by us, the aim of this study was to investigate the effects of Sarcopoterium spinosum extract (SSE) on dyslipidemia and vascular functions. We utilized ApoE deficient mice (ApoE-/- , Atherosclerosis-prone apolipoprotein E-deficient), who have a severe impairment in plasma lipoprotein clearance and thus develop alterations in blood lipid profile and are highly susceptible to atherogenic plaque formation. A total of 34 male mice were divided into five groups representing various genetic, dietary, and treatment configurations. Optical measurements were used to assess changes in diffused reflectance spectra, optical properties (absorption and scattering), and cerebral tissue chromophore contents. Specifically, significant improvement in cerebral hemoglobin level was observed in ApoE KO mice, fed an artherogenic diet (ATD), upon SSE treatment. Biochemical and histological analyses of ApoE-/- ATD mice showed elevated body weight and a high level of blood triglycerides, free fatty acids and cholesterol. In contrast, in SSE treated mice improvement was observed, suggesting beneficial effects of SSE. In ApoE-/- ATD mice group a higher levels of deoxyhemoglobin was monitored indicating that the rate of oxygen release to the tissue is low. This was supported by decrease in oxygen saturation. It was also shown a reduction in water content in the brain of ApoE KO. Mice fed with the atherogenic diet demonstrated increased water content as compared to STD-fed ApoE KO mice, while SSE administration reversed the effect of the diet. To our knowledge, no such study has been reported before.
Collapse
Affiliation(s)
- David Shemesh
- Department of Electrical and Electronics Engineering, Ariel University, Ariel, Israel
| | - Konstantin Rozenberg
- Departments of Molecular Biology and Nutritional Studies, Ariel University, Ariel, Israel
| | - Tovit Rosenzweig
- Departments of Molecular Biology and Nutritional Studies, Ariel University, Ariel, Israel
| | - David Abookasis
- Department of Electrical and Electronics Engineering, Ariel University, Ariel, Israel
| |
Collapse
|
59
|
Singh B, Kosuru R, Lakshmikanthan S, Sorci-Thomas M, Zhang D, Sparapani R, Vasquez-Vivar J, Chrzanowska M. Endothelial Rap1 (Ras-Association Proximate 1) Restricts Inflammatory Signaling to Protect From the Progression of Atherosclerosis. Arterioscler Thromb Vasc Biol 2021; 41:638-650. [PMID: 33267664 PMCID: PMC8105264 DOI: 10.1161/atvbaha.120.315401] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Small GTPase Rap1 (Ras-association proximate 1) is a novel, positive regulator of NO release and endothelial function with a potentially key role in mechanosensing of atheroprotective, laminar flow. Our objective was to delineate the role of Rap1 in the progression of atherosclerosis and its specific functions in the presence and absence of laminar flow, to better define its role in endothelial mechanisms contributing to plaque formation and atherogenesis. Approach and Results: In a mouse atherosclerosis model, endothelial Rap1B deletion exacerbates atherosclerotic plaque formation. In the thoracic aorta, where laminar shear stress-induced NO is otherwise atheroprotective, plaque area is increased in Athero-Rap1BiΔEC (atherogenic endothelial cell-specific, tamoxifen-inducible Rap1A+Rap1B knockout) mice. Endothelial Rap1 deficiency also leads to increased plaque size, leukocyte accumulation, and increased CAM (cell adhesion molecule) expression in atheroprone areas, whereas vascular permeability is unchanged. In endothelial cells, in the absence of protective laminar flow, Rap1 deficiency leads to an increased proinflammatory TNF-α (tumor necrosis factor alpha) signaling and increased NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells) activation and elevated inflammatory receptor expression. Interestingly, this increased signaling to NF-κB activation is corrected by AKTVIII-an inhibitor of Akt (protein kinase B) translocation to the membrane. Together, these data implicate Rap1 in restricting Akt-dependent signaling, preventing excessive cytokine receptor signaling and proinflammatory NF-κB activation. CONCLUSIONS Via 2 distinct mechanisms, endothelial Rap1 protects from the atherosclerosis progression in the presence and absence of laminar flow; Rap1-stimulated NO release predominates in laminar flow, and restriction of proinflammatory signaling predominates in the absence of laminar flow. Our studies provide novel insights into the mechanisms underlying endothelial homeostasis and reveal the importance of Rap1 signaling in cardiovascular disease.
Collapse
Affiliation(s)
- Bandana Singh
- Blood Research Institute, Versiti, Milwaukee, Wisconsin
| | - Ramoji Kosuru
- Blood Research Institute, Versiti, Milwaukee, Wisconsin
| | | | - Mary Sorci-Thomas
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
- Division of Endocrinology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - David Zhang
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Rodney Sparapani
- Division of Biostatistics, Institute for Health and Equity, Medical College of Wisconsin, Milwaukee, Wisconsin
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Jeannette Vasquez-Vivar
- Department of Biophysics and Redox Biology Program, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Magdalena Chrzanowska
- Blood Research Institute, Versiti, Milwaukee, Wisconsin
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
60
|
He Z, Kwek E, Hao W, Zhu H, Liu J, Ma KY, Chen ZY. Hawthorn fruit extract reduced trimethylamine-N-oxide (TMAO)-exacerbated atherogenesis in mice via anti-inflammation and anti-oxidation. Nutr Metab (Lond) 2021; 18:6. [PMID: 33413490 PMCID: PMC7789617 DOI: 10.1186/s12986-020-00535-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 12/14/2020] [Indexed: 02/07/2023] Open
Abstract
Background Trimethylamine-N-oxide (TMAO) is an independent risk factor for atherosclerosis. Consumption of hawthorn fruit is believed to be cardio-protective, yet whether it is able to suppress the TMAO-induced atherosclerosis remains unexplored. The present study was to investigate the effects of hawthorn fruit extract (HFE) on TMAO-exacerbated atherogenesis.
Methods Five groups of male Apolipoprotein E knock-out (ApoE−/−) mice were fed a low-fat diet (LFD), a Western high-fat diet (WD), or one of the three WDs containing 0.2% TMAO (WD + TMAO), 0.2% TMAO plus 1% HFE (WD + TMAO + L-HFE), or 0.2% TMAO plus 2% HFE (WD + TMAO + H-HFE), respectively. After 12-weeks of intervention, plasma levels of TMAO, lipid profile, inflammatory biomarkers, and antioxidant enzyme activities were measured. Atherosclerotic lesions in the thoracic aorta and aortic sinus were evaluated. The sterols and fatty acids in the liver and feces were extracted and measured. Hepatic expressions of inflammatory biomarkers and antioxidant enzymes were analyzed. Results Dietary TMAO accelerated atherogenesis, exacerbated inflammation, and reduced antioxidant capacities in the plasma and the liver. TMAO promoted hepatic cholesterol accumulation by inhibiting fecal excretion of acidic sterols. HFE could dose-dependently reduce the TMAO-aggravated atherosclerosis and inflammation. HFE was also able to reverse the TMAO-induced reduction in antioxidant capacity by up-regulating the expression of antioxidant enzymes including superoxide dismutase 1 (SOD1), SOD2, glutathione peroxidase 3 (GSH-Px3), and catalase (CAT) in the liver. Moreover, the hepatic cholesterol content was lowered by HFE via enhanced fecal excretion of neutral and acidic sterols. Conclusions The present results indicated that HFE was able to reduce the TMAO-exacerbated atherogenesis by attenuating inflammation and improving antioxidant capacity at least in mice. Graphic abstract ![]()
Collapse
Affiliation(s)
- Zouyan He
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China.,School of Public Health, Guangxi Medical University, Nanning, 530021, China
| | - Erika Kwek
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Wangjun Hao
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Hanyue Zhu
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Jianhui Liu
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China.,College of Food Science and Engineering, Nanjing University of Finance and Economics, Nanjing, 210023, China
| | - Ka Ying Ma
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Zhen-Yu Chen
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China.
| |
Collapse
|
61
|
Lavrynenko O, Titz B, Dijon S, Santos DD, Nury C, Schneider T, Guedj E, Szostak J, Kondylis A, Phillips B, Ekroos K, Martin F, Peitsch MC, Hoeng J, Ivanov NV. Ceramide ratios are affected by cigarette smoke but not heat-not-burn or e-vapor aerosols across four independent mouse studies. Life Sci 2020; 263:118753. [PMID: 33189821 DOI: 10.1016/j.lfs.2020.118753] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 11/03/2020] [Accepted: 11/10/2020] [Indexed: 11/16/2022]
Abstract
AIMS Smoking is an important risk factor for the development of chronic obstructive pulmonary disease and cardiovascular diseases. This study aimed to further elucidate the role of ceramides, as a key lipid class dysregulated in disease states. MAIN METHODS In this article we developed and validated LC-MS/MS method for ceramides (Cer(d18:1/16:0), Cer(d18:1/18:0), Cer(d18:1/24:0) and Cer(d18:1/24:1(15Z)) for the absolute quantification. We deployed it together with proteomics and transcriptomic analysis to assess the effects of cigarette smoke (CS) from the reference cigarette as well as aerosols from heat-not-burn (HnB) tobacco and e-vapor products in apolipoprotein E-deficient (ApoE-/-) mice over several time points. KEY FINDINGS In the lungs, CS exposure substantially elevated the ratios of Cer(d18:1/24:0) and Cer(d18:1/24:1) to Cer(d18:1/18:0) in two independent ApoE-/- mouse inhalation studies. Data from previous studies, in both ApoE-/- and wild-type mice, further confirmed the reproducibility of this finding. Elevation of these ceramide ratios was also observed in plasma/serum, the liver, and-for the Cer(d18:1/24:1(15Z)) to Cer(d18:1/18:0) ratio-the abdominal aorta. Also, the levels of acid ceramidase (Asah1) and glucocerebrosidase (Gba)-lysosomal enzymes involved in the hydrolysis of glucosylceramides-were consistently elevated in the lungs after CS exposure. In contrast, exposure to HnB tobacco product and e-vapor aerosols did not induce significant changes in the ceramide profiles or associated enzymes. SIGNIFICANCE Our work in mice contributes to the accumulating evidence on the importance of ceramide ratios as biologically relevant markers for respiratory disorders, adding to their already demonstrated role in cardiovascular disease risk assessment in humans.
Collapse
Affiliation(s)
- Oksana Lavrynenko
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland.
| | - Bjoern Titz
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland
| | - Sophie Dijon
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland
| | - Daniel Dos Santos
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland
| | - Catherine Nury
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland
| | - Thomas Schneider
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland
| | - Emmanuel Guedj
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland
| | - Justyna Szostak
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland
| | - Athanasios Kondylis
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland
| | - Blaine Phillips
- Philip Morris International Research Laboratories Pte. Ltd., Science Park II, 117406, Singapore
| | - Kim Ekroos
- Lipidomics Consulting Ltd., Irisviksvägen 31D, 02230 Esbo, Finland
| | - Florian Martin
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland
| | - Manuel C Peitsch
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland
| | - Julia Hoeng
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland
| | - Nikolai V Ivanov
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland
| |
Collapse
|
62
|
Lu X, Wang S, Feng S, Li H. CSE/H 2S system alleviates uremic accelerated atherosclerosis by regulating TGF-β/Smad3 pathway in 5/6 nephrectomy ApoE -/- mice. BMC Nephrol 2020; 21:527. [PMID: 33276745 PMCID: PMC7716493 DOI: 10.1186/s12882-020-02183-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 11/24/2020] [Indexed: 01/17/2023] Open
Abstract
Background Hydrogen sulfide (H2S) has been shown to inhibit the atherosclerosis development and progression. It is produced by cystathionine γ-lyase (CSE) in the cardiovascular system. In our previous study, it has been shown that CSE/H2S system plays a significant role in the changes of uremic accelerated atherosclerosis (UAAS), but the mechanism is not known clearly. Methods In this study, we explored the antagonism of CSE/H2S system in UAAS and identified its possible signaling molecules in ApoE−/− mice with 5/6 nephrectomy and fed with atherogenic diet. Mice were divided into sham operation group (sham group), UAAS group, sodium hydrosulfide group (UAAS+NaHS group) and propargylglycine group (UAAS+PPG group). Serum creatinine, urea nitrogen, lipid levels and lesion size of atherosclerotic plaque in the aortic roots were analyzed. Meanwhile, the expression of CSE, TGF-β and phosphorylation of Smad3 were detected. Results Compared with sham group, the aortic root of ApoE−/− mice in the UAAS group developed early atherosclerosis, the levels of total cholesterol, triglyceride, low-density lipoprotein-cholesterol, serum creatinine and urea nitrogen were also higher than that in the sham group. NaHS administration can inhibit the development of atherosclerosis, but PPG administration can accelerate the atherosclerosis development. Meanwhile, the protein expression levels of CSE and TGF-β and phosphorylation of Smad3 significantly decreased in the UAAS mice. Treatment of UAAS mice with NaHS inhibited TGF-β protein expression and Smad3 phosphorylation decrease, but PPG treatment had the opposite effect. Conclusions The CSE/H2S system is of great importance for treating atherosclerosis in patients with chronic kidney disease, and it may protect the vascular from atherosclerosis through the TGF-β/Smad pathway.
Collapse
Affiliation(s)
- Xiangxue Lu
- Department of Blood Purification, Beijing Chao-Yang Hospital, Capital Medical University, No. 8 Gongti South Road, Chaoyang District, Beijing, 100020, China
| | - Shixiang Wang
- Department of Blood Purification, Beijing Chao-Yang Hospital, Capital Medical University, No. 8 Gongti South Road, Chaoyang District, Beijing, 100020, China
| | - Sujuan Feng
- Department of Blood Purification, Beijing Chao-Yang Hospital, Capital Medical University, No. 8 Gongti South Road, Chaoyang District, Beijing, 100020, China
| | - Han Li
- Department of Blood Purification, Beijing Chao-Yang Hospital, Capital Medical University, No. 8 Gongti South Road, Chaoyang District, Beijing, 100020, China.
| |
Collapse
|
63
|
Inoue KI, Toyoda S, Jojima T, Abe S, Sakuma M, Inoue T. Time-restricted feeding prevents high-fat and high-cholesterol diet-induced obesity but fails to ameliorate atherosclerosis in apolipoprotein E-knockout mice. Exp Anim 2020; 70:194-202. [PMID: 33268668 PMCID: PMC8150245 DOI: 10.1538/expanim.20-0112] [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: 12/29/2022] Open
Abstract
One of the leading risk factors for atherosclerosis is obesity, which is commonly caused by a nutrient-rich Western-style diet, sedentary behaviors, and shift
work. Time-restricted (TR) feeding and intermittent fasting are both known to prevent overweight and adiposity, improve glucose tolerance, and decrease plasma
cholesterol in high-fat diet-induced obese mice. Here we examined the overall effects of TR feeding of a Western diet (fat, 40.5 Kcal%; cholesterol, 0.21 g%)
using 8-week-old Apoe−/− mice. Mice were assigned into three groups: (1) an ad libitum (AL) group fed an AL Western
diet, (2) a TR group with restricted access to a Western diet (15 h/day, 12:00 to 3:00 Zeitgeber time [ZT]); and (3) an Ex/TR group fed a TR Western diet and
subjected to physical exercise at 12:00 ZT. Mice in the AL group gained body weight rapidly during the 14-week observation period. With TR feeding, excessive
weight gain, liver adiposity, visceral fat, and brown adipose tissue volume were effectively suppressed. Although TR feeding failed to decrease Oil Red
O-stained aortic plaques in Apoe−/− mice, physical exercise significantly decreased them. Neither TR feeding with exercise nor that
without exercise decreased the mean area under the curve of the plasma cholesterol level or the fasting plasma glucose. Collectively, TR feeding of a Western
diet prevented the development of obesity but failed to ameliorate atherosclerosis in Apoe−/− mice.
Collapse
Affiliation(s)
- Ken-Ichi Inoue
- Comprehensive Research Facilities for Advanced Medical Science, Research Center for Advanced Medical Science, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Tochigi 321-0293, Japan.,Center of Regenerative Medicine, Dokkyo Medical University Hospital, 880 Kitakobayashi, Mibu, Tochigi 321-0293, Japan
| | - Shigeru Toyoda
- Department of Cardiovascular Medicine, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Tochigi 321-0293, Japan
| | - Teruo Jojima
- Department of Endocrinology and Metabolism, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Tochigi 321-0293, Japan
| | - Shichiro Abe
- Department of Cardiovascular Medicine, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Tochigi 321-0293, Japan
| | - Masashi Sakuma
- Department of Cardiovascular Medicine, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Tochigi 321-0293, Japan
| | - Teruo Inoue
- Comprehensive Research Facilities for Advanced Medical Science, Research Center for Advanced Medical Science, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Tochigi 321-0293, Japan.,Center of Regenerative Medicine, Dokkyo Medical University Hospital, 880 Kitakobayashi, Mibu, Tochigi 321-0293, Japan.,Department of Cardiovascular Medicine, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Tochigi 321-0293, Japan
| |
Collapse
|
64
|
Li W, Yu J, Xiao X, Zang L, Yang Y, Yu J, Huang Q, Niu X, Li W. Imperatorin reduces the inflammatory response of atherosclerosis by regulating MAPKs signaling pathway in vivo and in vitro. Int Immunopharmacol 2020; 90:107170. [PMID: 33218940 DOI: 10.1016/j.intimp.2020.107170] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 10/22/2020] [Accepted: 10/30/2020] [Indexed: 10/23/2022]
Abstract
Inflammation plays an important role in the process of atherosclerosis (AS). Inhibition of inflammation is an effective way to prevent AS. Imperatorin (IMP) is a kind of furan coumarin with various activities. In this study, the anti-inflammatory effect of IMP was explored in oxidized low-density lipoprotein (ox-LDL)-induced VSMCs and high fat diet (HFD)-induced ApoE-/- mice. The results showed that IMP attenuated the elevation of TNF-α, IL-6, MCP-1 and NO induced by ox-LDL in supernatant of VSMCs. IMP has normalized the levels of serum lipids (TC, TG, LDL-C and HDL-C) and attenuated inflammatory cytokines in serum. IMP also improved pathological changes and lipid accumulation in aorta. Matrix metalloproteinase-2 (MMP-2) expression in aorta was down-regulated by IMP. IMP could inhibit the phosphorylation of MAPKs pathway in the aorta and VSMCs, resulting in a significant decrease in the contents of p-ERK 1/2, p-JNK and p-P38. Overall, IMP could exert anti-inflammatory effects in vivo and in vitro to interfere with AS.
Collapse
Affiliation(s)
- Wenqi Li
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China; Key Laboratory of Material Basis Analysis of Chinese Medicine, Shaanxi Administration of Traditional Chinese Medicine, Xi'an 710061, PR China
| | - Jinjin Yu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China; Key Laboratory of Material Basis Analysis of Chinese Medicine, Shaanxi Administration of Traditional Chinese Medicine, Xi'an 710061, PR China
| | - Xin Xiao
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China; Key Laboratory of Material Basis Analysis of Chinese Medicine, Shaanxi Administration of Traditional Chinese Medicine, Xi'an 710061, PR China
| | - Lulu Zang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China; Key Laboratory of Material Basis Analysis of Chinese Medicine, Shaanxi Administration of Traditional Chinese Medicine, Xi'an 710061, PR China
| | - Yajie Yang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China; Key Laboratory of Material Basis Analysis of Chinese Medicine, Shaanxi Administration of Traditional Chinese Medicine, Xi'an 710061, PR China
| | - Jiabao Yu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China; Key Laboratory of Material Basis Analysis of Chinese Medicine, Shaanxi Administration of Traditional Chinese Medicine, Xi'an 710061, PR China
| | - Qiuxia Huang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China; Key Laboratory of Material Basis Analysis of Chinese Medicine, Shaanxi Administration of Traditional Chinese Medicine, Xi'an 710061, PR China
| | - Xiaofeng Niu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China; Key Laboratory of Material Basis Analysis of Chinese Medicine, Shaanxi Administration of Traditional Chinese Medicine, Xi'an 710061, PR China
| | - Weifeng Li
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China; Key Laboratory of Material Basis Analysis of Chinese Medicine, Shaanxi Administration of Traditional Chinese Medicine, Xi'an 710061, PR China.
| |
Collapse
|
65
|
Zhao J, Wang Z, Yuan Z, Lv S, Su Q. Baicalin ameliorates atherosclerosis by inhibiting NLRP3 inflammasome in apolipoprotein E-deficient mice. Diab Vasc Dis Res 2020; 17:1479164120977441. [PMID: 33269624 PMCID: PMC7919226 DOI: 10.1177/1479164120977441] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND NLR family pyrin domain containing 3 (NLRP3) inflammasome has been implicated in the development of atherosclerosis and several studies have suggested that inhibiting NLRP3 inflammasome could be a potential therapeutic approach to treat atherosclerosis. Baicalin is a flavone glycoside with anti-inflammation, anti-oxidative activities. The inhibition of NLRP3 inflammasome activation by baicalin has also been described. Therefore, the effects of baicalin on NLRP3 inflammasome activation and atherosclerosis were evaluated in present study. METHODS We established the apolipoprotein E-deficient atherosclerosis mice model. After baicalin treatment, the IL-1, IL-18, and reactive oxygen species (ROS) production, and the plaque area was monitored. We also measured the NLRP3, ASC, caspase-1, ICAM-1, and VCAM-1 expression in atherosclerosis mice after baicalin treatment. We silenced NLRP3 by administration of lentivirus expressing NLRP3 shRNA to atherosclerosis mice and monitored the IL-1, IL-18, and ROS production, and NLRP3 inflammasome activation. RESULTS Baicalin remarkably inhibited the production of IL-1, IL-18, mitochondria ROS, total ROS, ICAM-1, and VCAM-1. Baicalin reduced the expression of NLRP3 inflammasome and suppressed its activation. Baicalin significantly reduced the plaque area. Silencing NLRP3 resulted in decreased production of IL-1, IL-18, mitochondria ROS, total ROS, ICAM-1, and VCAM-1, and inhibition of NLRP3 inflammasome activation. CONCLUSION Baicalin ameliorated atherosclerosis by inhibiting NLRP3 inflammasome.
Collapse
Affiliation(s)
- Jingfei Zhao
- Department of Surgery, Third People’s Hospital of Liaocheng City, Liaocheng, Shandong, China
| | - Zhengtang Wang
- Department of Surgery, Third People’s Hospital of Liaocheng City, Liaocheng, Shandong, China
| | - Zhilu Yuan
- Department of Surgery, Third People’s Hospital of Liaocheng City, Liaocheng, Shandong, China
| | - Shuzhen Lv
- Department of Orthopedic, Third People’s Hospital of Liaocheng City, Liaocheng, Shandong, China
| | - Qingbo Su
- Department of Vascular Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Qingbo Su, Department of Vascular Surgery, Qilu Hospital of Shandong University, No. 107 Wenhua Xilu, Jinan, Shandong 250012, China.
| |
Collapse
|
66
|
Berenji Ardestani S, Eftedal I, Pedersen M, Jeppesen PB, Nørregaard R, Matchkov VV. Endothelial dysfunction in small arteries and early signs of atherosclerosis in ApoE knockout rats. Sci Rep 2020; 10:15296. [PMID: 32943715 PMCID: PMC7499202 DOI: 10.1038/s41598-020-72338-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 08/17/2020] [Indexed: 12/18/2022] Open
Abstract
Endothelial dysfunction is recognized as a major contributor to atherosclerosis and has been suggested to be evident far before plaque formation. Endothelial dysfunction in small resistance arteries has been suggested to initiate long before changes in conduit arteries. In this study, we address early changes in endothelial function of atherosclerosis prone rats. Male ApoE knockout (KO) rats (11- to 13-weeks-old) were subjected to either a Western or standard diet. The diet intervention continued for a period of 20–24 weeks. Endothelial function of pulmonary and mesenteric arteries was examined in vitro using an isometric myograph. We found that Western diet decreased the contribution of cyclooxygenase (COX) to control the vascular tone of both pulmonary and mesenteric arteries. These changes were associated with early stage atherosclerosis and elevated level of plasma total cholesterol, LDL and triglyceride in ApoE KO rats. Chondroid-transformed smooth muscle cells, calcifications, macrophages accumulation and foam cells were also observed in the aortic arch from ApoE KO rats fed Western diet. The ApoE KO rats are a new model to study endothelial dysfunction during the earlier stages of atherosclerosis and could help us improve preclinical drug development.
Collapse
Affiliation(s)
- Simin Berenji Ardestani
- Department of Clinical Medicine, Aarhus University, Palle Juul Jensens Boulevard 99, 8200, Aarhus N, Denmark. .,Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, NTNU Norwegian University of Science and Technology, Trondheim, Norway.
| | - Ingrid Eftedal
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, NTNU Norwegian University of Science and Technology, Trondheim, Norway.,Faculty of Nursing and Health Sciences, Nord University, Bodø, Norway
| | - Michael Pedersen
- Department of Clinical Medicine, Aarhus University, Palle Juul Jensens Boulevard 99, 8200, Aarhus N, Denmark
| | - Per Bendix Jeppesen
- Department of Clinical Medicine, Aarhus University, Palle Juul Jensens Boulevard 99, 8200, Aarhus N, Denmark
| | - Rikke Nørregaard
- Department of Clinical Medicine, Aarhus University, Palle Juul Jensens Boulevard 99, 8200, Aarhus N, Denmark
| | - Vladimir V Matchkov
- Department of Biomedicine, MEMBRANES, Health, Aarhus University, Aarhus, Denmark
| |
Collapse
|
67
|
Dybas J, Bulat K, Blat A, Mohaissen T, Wajda A, Mardyla M, Kaczmarska M, Franczyk-Zarow M, Malek K, Chlopicki S, Marzec KM. Age-related and atherosclerosis-related erythropathy in ApoE/LDLR -/- mice. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165972. [PMID: 32949768 DOI: 10.1016/j.bbadis.2020.165972] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 08/30/2020] [Accepted: 09/14/2020] [Indexed: 10/24/2022]
Abstract
In this work we applied a multimodal approach to define the age- and atherosclerosis-related biochemical and functional alterations in red blood cells (RBCs) in ApoE/LDLR-/- mice. Our results revealed that age-related changes in RBCs, such as decreases in RBC deformability and mean height, were more pronounced in ApoE/LDLR-/- mice than in age-matched control mice (C57BL/6J). The decreases in phospholipid content and level of lipid unsaturation were accompanied by an increase in cholesterol esters and esterified lipids in RBC membranes in aged C57BL/6J mice. The age-related decrease in the phospholipid content was more pronounced in ApoE/LDLR-/- mice. In contrast, the increase in the total lipid content in RBC membranes occurred only in ApoE/LDLR-/- mice with advanced atherosclerosis. The age-related alterations also included a decrease in the ratio of turns to α-helices in the secondary structure of hemoglobin (Hb) inside intact RBCs. On the other hand, an increase in the ratio of unordered conformations to α-helices of Hb was observed only in ApoE/LDLR-/- mice and occurred already at the age of 5-weeks. This was related to hypercholesterolemia and resulted in an increased oxygen-carrying capacity. In conclusion, progressive mechanical and functional alterations of RBCs in aged ApoE/LDLR-/- mice were more pronounced than in age-matched C57BL/6J mice. Although, several biochemical changes in RBCs in aged ApoE/LDLR-/- mice recapitulated age-dependent changes observed in control mice, some biochemical features of RBC membranes attributed to hypercholesterolemia were distinct and could contribute to the accelerated deterioration of RBC function in ApoE/LDLR-/- mice.
Collapse
Affiliation(s)
- Jakub Dybas
- Jagiellonian Center for Experimental Therapeutics, Jagiellonian University, 14 Bobrzyńskiego St., 30-348 Krakow, Poland
| | - Katarzyna Bulat
- Jagiellonian Center for Experimental Therapeutics, Jagiellonian University, 14 Bobrzyńskiego St., 30-348 Krakow, Poland
| | - Aneta Blat
- Jagiellonian Center for Experimental Therapeutics, Jagiellonian University, 14 Bobrzyńskiego St., 30-348 Krakow, Poland; Faculty of Chemistry, Jagiellonian University, 2 Gronostajowa Str., 30-387 Krakow, Poland
| | - Tasnim Mohaissen
- Jagiellonian Center for Experimental Therapeutics, Jagiellonian University, 14 Bobrzyńskiego St., 30-348 Krakow, Poland; Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna Str., 30-688 Krakow, Poland
| | - Aleksandra Wajda
- Jagiellonian Center for Experimental Therapeutics, Jagiellonian University, 14 Bobrzyńskiego St., 30-348 Krakow, Poland; Faculty of Materials Science and Ceramics, AGH University of Science and Technology, Mickiewicza 30, 30-059 Krakow, Poland
| | - Mateusz Mardyla
- Jagiellonian Center for Experimental Therapeutics, Jagiellonian University, 14 Bobrzyńskiego St., 30-348 Krakow, Poland; Jagiellonian University, University School of Physical Education in Krakow, 78 Jana Pawła II St., 31-571 Krakow, Poland
| | - Magdalena Kaczmarska
- Jagiellonian Center for Experimental Therapeutics, Jagiellonian University, 14 Bobrzyńskiego St., 30-348 Krakow, Poland
| | - Magdalena Franczyk-Zarow
- Department of Human Nutrition and Dietetics, Faculty of Food Technology, University of Agriculture, 122 Balicka St., 30-149 Krakow, Poland
| | - Kamilla Malek
- Faculty of Chemistry, Jagiellonian University, 2 Gronostajowa Str., 30-387 Krakow, Poland
| | - Stefan Chlopicki
- Jagiellonian Center for Experimental Therapeutics, Jagiellonian University, 14 Bobrzyńskiego St., 30-348 Krakow, Poland; Department of Experimental Pharmacology, Jagiellonian University Medical College, 16 Grzegorzecka St., 31-531 Krakow, Poland
| | - Katarzyna M Marzec
- Jagiellonian Center for Experimental Therapeutics, Jagiellonian University, 14 Bobrzyńskiego St., 30-348 Krakow, Poland.
| |
Collapse
|
68
|
Bhattacharjee S, Lee Y, Zhu B, Wu H, Chen Y, Chen H. Epsins in vascular development, function and disease. Cell Mol Life Sci 2020; 78:833-842. [PMID: 32930806 DOI: 10.1007/s00018-020-03642-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 08/14/2020] [Accepted: 09/03/2020] [Indexed: 12/15/2022]
Abstract
Epsins are a family of adaptor proteins involved in clathrin-dependent endocytosis. In the vasculature, epsins 1 and 2 are functionally redundant members of this family that are expressed in the endothelial cells of blood vessels and the lymphatic system throughout development and adulthood. These proteins contain a number of peptide motifs that allow them to interact with lipid moieties and a variety of proteins. These interactions facilitate the regulation of a wide range of cell signaling pathways. In this review, we focus on the involvement of epsins 1 and 2 in controlling vascular endothelial growth factor receptor signaling in angiogenesis and lymphangiogenesis. We also discuss the therapeutic implications of understanding the molecular mechanisms of epsin-mediated regulation in diseases such as atherosclerosis and diabetes.
Collapse
Affiliation(s)
- Sudarshan Bhattacharjee
- Vascular Biology Program, Harvard Medical School, Boston Children's Hospital and Department of Surgery, Boston, MA, 02115, USA
| | - Yang Lee
- Vascular Biology Program, Harvard Medical School, Boston Children's Hospital and Department of Surgery, Boston, MA, 02115, USA
| | - Bo Zhu
- Vascular Biology Program, Harvard Medical School, Boston Children's Hospital and Department of Surgery, Boston, MA, 02115, USA
| | - Hao Wu
- Vascular Biology Program, Harvard Medical School, Boston Children's Hospital and Department of Surgery, Boston, MA, 02115, USA
| | - Yabing Chen
- Department of Pathology, Birmingham Veterans Affairs Medical Center, University of Alabama at Birmingham and Research Department, Birmingham, AL, 35294, USA
| | - Hong Chen
- Vascular Biology Program, Harvard Medical School, Boston Children's Hospital and Department of Surgery, Boston, MA, 02115, USA.
| |
Collapse
|
69
|
Riedl KA, Kampf T, Herold V, Behr VC, Bauer WR. Wall shear stress analysis using 17.6 Tesla MRI: A longitudinal study in ApoE-/- mice with histological analysis. PLoS One 2020; 15:e0238112. [PMID: 32857805 PMCID: PMC7454980 DOI: 10.1371/journal.pone.0238112] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 08/10/2020] [Indexed: 12/31/2022] Open
Abstract
This longitudinal study was performed to evaluate the feasibility of detecting the interaction between wall shear stress (WSS) and plaque development. 20 ApoE-/- mice were separated in 12 mice with Western Diet and 8 mice with Chow Diet. Magnetic resonance (MR) scans at 17.6 Tesla and histological analysis were performed after one week, eight and twelve weeks. All in vivo MR measurements were acquired using a flow sensitive phase contrast method for determining vectorial flow. Histological sections were stained with Hematoxylin and Eosin, Elastica van Gieson and CD68 staining. Data analysis was performed using Ensight and a Matlab-based “Flow Tool”. The body weight of ApoE-/- mice increased significantly over 12 weeks. WSS values increased in the Western Diet group over the time period; in contrast, in the Chow Diet group the values decreased from the first to the second measurement point. Western Diet mice showed small plaque formations with elastin fragmentations after 8 weeks and big plaque formations after 12 weeks; Chow Diet mice showed a few elastin fragmentations after 8 weeks and small plaque formations after 12 weeks. Favored by high-fat diet, plaque formation results in higher values of WSS. With wall shear stress being a known predictor for atherosclerotic plaque development, ultra highfield MRI can serve as a tool for studying the causes and beginnings of atherosclerosis.
Collapse
Affiliation(s)
- Katharina A. Riedl
- Department of Experimental Physics V, University of Würzburg, Würzburg, Germany
- Department of Cardiology, University Heart & Vascular Center Hamburg, Hamburg, Germany
- * E-mail:
| | - Thomas Kampf
- Department of Experimental Physics V, University of Würzburg, Würzburg, Germany
- Department of Diagnostic and Interventional Neuroradiology, University Hospital Würzburg, Würzburg, Germany
| | - Volker Herold
- Department of Experimental Physics V, University of Würzburg, Würzburg, Germany
| | - Volker C. Behr
- Department of Experimental Physics V, University of Würzburg, Würzburg, Germany
| | - Wolfgang R. Bauer
- Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
70
|
Novel Mechanism of Cholesterol Transport by ABCA5 in Macrophages and Its Role in Dyslipidemia. J Mol Biol 2020; 432:4922-4941. [DOI: 10.1016/j.jmb.2020.07.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 07/02/2020] [Accepted: 07/10/2020] [Indexed: 01/24/2023]
|
71
|
A Timing Effect of 17-β Estradiol on Atherosclerotic Lesion Development in Female ApoE -/- Mice. Int J Mol Sci 2020; 21:ijms21134710. [PMID: 32630298 PMCID: PMC7369926 DOI: 10.3390/ijms21134710] [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: 06/04/2020] [Revised: 06/25/2020] [Accepted: 06/29/2020] [Indexed: 11/18/2022] Open
Abstract
Differences in size or composition of existing plaques at the initiation of estrogen (E2) therapy may underpin evidence of increased risk of atherosclerosis-associated clinical sequelae. We investigated whether E2 had divergent effects on actively-growing versus established-advanced atherosclerotic lesions. Eight weeks of subcutaneous bi-weekly injections of 3 µg/g 17β-estradiol (n = 18) or vehicle control (n = 22) were administered to female Apolipoprotein null-mice aged 25- or 45 weeks old. Histological assessment of lesion size within the brachiocephalic artery was conducted. Lesion composition was also assessed with acellular, calcification and fibrosis areas measured and other cellular features (intimal thickening, foam cells, lipid pools and cholesterol) scored (0–3) for severity. The comparison showed increased lesion size and calcified area with advancing age but no effect of E2. However, subtle changes in composition were observed following E2. Within the younger group, E2 increased intima thickening and acceleration of calcification. In the older group, E2 increased the thickness of the lesion cap. Therefore, this study shows different effects of E2 depending on the underlying stage of lesion development at the time of initiation of treatment. These divergent changes help explain the controversy of the adverse effects of E2 treatment in cardiovascular disease.
Collapse
|
72
|
Lee J, Choi JH. Deciphering Macrophage Phenotypes upon Lipid Uptake and Atherosclerosis. Immune Netw 2020; 20:e22. [PMID: 32655970 PMCID: PMC7327152 DOI: 10.4110/in.2020.20.e22] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 06/13/2020] [Accepted: 06/15/2020] [Indexed: 02/08/2023] Open
Abstract
In the progression of atherosclerosis, macrophages are the key immune cells for foam cell formation. During hyperlipidemic condition, phagocytic cells such as monocytes and macrophages uptake oxidized low-density lipoproteins (oxLDLs) accumulated in subintimal space, and lipid droplets are accumulated in their cytosols. In this review, we discussed the characteristics and phenotypic changes of macrophages in atherosclerosis and the effect of cytosolic lipid accumulation on macrophage phenotype. Due to macrophage plasticity, the inflammatory phenotypes triggered by oxLDL can be re-programmed by cytosolic lipid accumulation, showing downregulation of NF-κB activation followed by activation of anti-inflammatory genes, leading to tissue repair and homeostasis. We also discuss about various in vivo and in vitro models for atherosclerosis research and next generation sequencing technologies for foam cell gene expression profiling. Analysis of the phenotypic changes of macrophages during the progression of atherosclerosis with adequate approach may lead to exact understandings of the cellular mechanisms and hint therapeutic targets for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Jihye Lee
- Department of Life Science, College of Natural Sciences, Research Institute of Natural Sciences, Hanyang University, Seoul 04763, Korea
| | - Jae-Hoon Choi
- Department of Life Science, College of Natural Sciences, Research Institute of Natural Sciences, Hanyang University, Seoul 04763, Korea
| |
Collapse
|
73
|
Corbacho-Alonso N, Baldán-Martín M, López JA, Rodríguez-Sánchez E, Martínez PJ, Mourino-Alvarez L, Martin-Rojas T, Sastre-Oliva T, Madruga F, Vázquez J, Padial LR, Alvarez-Llamas G, Vivanco F, Ruiz-Hurtado G, Ruilope LM, Barderas MG. Novel molecular plasma signatures on cardiovascular disease can stratify patients throughout life. J Proteomics 2020; 222:103816. [DOI: 10.1016/j.jprot.2020.103816] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 03/30/2020] [Accepted: 05/06/2020] [Indexed: 02/08/2023]
|
74
|
Kefir peptides alleviate high-fat diet-induced atherosclerosis by attenuating macrophage accumulation and oxidative stress in ApoE knockout mice. Sci Rep 2020; 10:8802. [PMID: 32472055 PMCID: PMC7260220 DOI: 10.1038/s41598-020-65782-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 05/11/2020] [Indexed: 02/01/2023] Open
Abstract
In the past decade, the high morbidity and mortality of atherosclerotic disease have been prevalent worldwide. High-fat food consumption has been suggested to be an overarching factor for atherosclerosis incidence. This study aims to investigate the effects of kefir peptides on high-fat diet (HFD)-induced atherosclerosis in apolipoprotein E knockout (ApoE−/−) mice. 7-week old male ApoE−/− and normal C57BL/6 mice were randomly divided into five groups (n = 8). Atherosclerotic lesion development in ApoE−/− mice was established after fed the HFD for 12 weeks compared to standard chow diet (SCD)-fed C57BL/6 and ApoE−/− control groups. Kefir peptides oral administration significantly improved atherosclerotic lesion development by protecting against endothelial dysfunction, decreasing oxidative stress, reducing aortic lipid deposition, attenuating macrophage accumulation, and suppressing the inflammatory immune response compared with the HFD/ApoE−/− mock group. Moreover, the high dose of kefir peptides substantially inhibited aortic fibrosis and restored the fibrosis in the aorta root close to that observed in the C57BL/6 normal control group. Our findings show, for the first time, anti-atherosclerotic progression via kefir peptides consumption in HFD-fed ApoE−/− mice. The profitable effects of kefir peptides provide new perspectives for its use as an anti-atherosclerotic agent in the preventive medicine.
Collapse
|
75
|
Hossaini Nasr S, Rashidijahanabad Z, Ramadan S, Kauffman N, Parameswaran N, Zinn KR, Qian C, Arora R, Agnew D, Huang X. Effective atherosclerotic plaque inflammation inhibition with targeted drug delivery by hyaluronan conjugated atorvastatin nanoparticles. NANOSCALE 2020; 12:9541-9556. [PMID: 32314997 PMCID: PMC7234819 DOI: 10.1039/d0nr00308e] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Atherosclerosis is associated with inflammation in the arteries, which is a major cause of heart attacks and strokes. Reducing the extent of local inflammation at atherosclerotic plaques can be an attractive strategy to combat atherosclerosis. While statins can exhibit direct anti-inflammatory activities, the high dose required for such a therapy renders it unrealistic due to their low systemic bioavailabilities and potential side effects. To overcome this, a new hyaluronan (HA)-atorvastatin (ATV) conjugate was designed with the hydrophobic statin ATV forming the core of the nanoparticle (HA-ATV-NP). The HA on the NPs can selectively bind with CD44, a cell surface receptor overexpressed on cells residing in atherosclerotic plaques and known to play important roles in plaque development. HA-ATV-NPs exhibited significantly higher anti-inflammatory effects on macrophages compared to ATV alone in vitro. Furthermore, when administered in an apolipoprotein E (ApoE)-knockout mouse model of atherosclerosis following a 1-week treatment regimen, HA-ATV-NPs markedly decreased inflammation in advanced atherosclerotic plaques, which were monitored through contrast agent aided magnetic resonance imaging. These results suggest CD44 targeting with HA-ATV-NPs is an attractive strategy to reduce harmful inflammation in atherosclerotic plaques.
Collapse
Affiliation(s)
- Seyedmehdi Hossaini Nasr
- Department of Chemistry Michigan State University, East Lansing, Michigan 48824, USA. and Institute for Quantitative Health Science and Engineering Michigan State University, East Lansing, Michigan 48824, USA
| | - Zahra Rashidijahanabad
- Department of Chemistry Michigan State University, East Lansing, Michigan 48824, USA. and Institute for Quantitative Health Science and Engineering Michigan State University, East Lansing, Michigan 48824, USA
| | - Sherif Ramadan
- Department of Chemistry Michigan State University, East Lansing, Michigan 48824, USA. and Institute for Quantitative Health Science and Engineering Michigan State University, East Lansing, Michigan 48824, USA and Chemistry Department, Faculty of Science, Benha University, Benha, Qaliobiya 13518, Egypt
| | - Nate Kauffman
- Institute for Quantitative Health Science and Engineering Michigan State University, East Lansing, Michigan 48824, USA and Department of Biomedical Engineering Michigan State University, East Lansing, Michigan 48824, USA
| | | | - Kurt R Zinn
- Institute for Quantitative Health Science and Engineering Michigan State University, East Lansing, Michigan 48824, USA and Department of Biomedical Engineering Michigan State University, East Lansing, Michigan 48824, USA and Department of Radiology Michigan State University, East Lansing, Michigan 48824, USA
| | - Chunqi Qian
- Department of Radiology Michigan State University, East Lansing, Michigan 48824, USA
| | - Ripla Arora
- Institute for Quantitative Health Science and Engineering Michigan State University, East Lansing, Michigan 48824, USA and Department of Obstetrics, Gynecology and Reproductive Biology Michigan State University, East Lansing, Michigan 48824, USA
| | - Dalen Agnew
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, Michigan 48824, USA
| | - Xuefei Huang
- Department of Chemistry Michigan State University, East Lansing, Michigan 48824, USA. and Institute for Quantitative Health Science and Engineering Michigan State University, East Lansing, Michigan 48824, USA and Department of Biomedical Engineering Michigan State University, East Lansing, Michigan 48824, USA
| |
Collapse
|
76
|
Ibrahim N‘I, Fairus S, Zulfarina MS, Naina Mohamed I. The Efficacy of Squalene in Cardiovascular Disease Risk-A Systematic Review. Nutrients 2020; 12:E414. [PMID: 32033387 PMCID: PMC7071298 DOI: 10.3390/nu12020414] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 01/31/2020] [Indexed: 12/19/2022] Open
Abstract
INTRODUCTION Cardiovascular disease (CVD) is the leading cause of death worldwide. Squalene (SQ), an intermediate for the cholesterol biosynthesis, has been proposed to act similarly to statins via inhibition of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase in the liver. PURPOSE This paper explores the effects of SQ in CVD. METHODS A systematic review of the literature was performed to identify relevant studies about SQ and CVD. A comprehensive search in Medline and Scopus for relevant studies published between the years 1946 and 2019 was performed. The main inclusion criteria were that the study was published in English; that the study reported association or effect of SQ and CVD; and that CVD should be related to lifestyle variables, aging, or experimentally induced conditions. RESULTS The literature searches identified 5562 potentially relevant articles, whereby 21 studies met the inclusion criteria. There were three human studies and 18 animal experimental studies included in this paper. Only one human study reported positive outcome of SQ in CVD. The remaining two studies reported inconsistent and/or no effect. For animal studies, 15 studies reported positive effect while the remaining reported negative and/or no effect of SQ on various related parameters. CONCLUSIONS This evidence-based review emphasizes the potential of SQ being used for cardiovascular-related diseases. The effect of SQ, especially of plant-based warrants further exploration. Controlled human observational studies should be performed to provide comprehensive evidence.
Collapse
Affiliation(s)
- Nurul ‘Izzah Ibrahim
- Pharmacoepidemiology and Drug Safety Unit, Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Bandar Tun Razak, Cheras 56000, Kuala Lumpur, Malaysia
| | - Syed Fairus
- Malaysian Palm Oil Board (MPOB), No. 6 Persiaran Institusi, Bandar Baru Bangi, Kajang 43000, Selangor, Malaysia
| | - Mohamed S. Zulfarina
- Pharmacoepidemiology and Drug Safety Unit, Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Bandar Tun Razak, Cheras 56000, Kuala Lumpur, Malaysia
| | - Isa Naina Mohamed
- Pharmacoepidemiology and Drug Safety Unit, Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Bandar Tun Razak, Cheras 56000, Kuala Lumpur, Malaysia
| |
Collapse
|
77
|
Ji R, Gu Y, Zhang J, Gao C, Gao W, Zang X, Zhao Y. TRIM7 promotes proliferation and migration of vascular smooth muscle cells in atherosclerosis through activating c-Jun/AP-1. IUBMB Life 2020; 72:247-258. [PMID: 31625258 DOI: 10.1002/iub.2181] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 08/23/2019] [Indexed: 01/04/2023]
Abstract
Atherosclerosis (AS), with associated risk of stroke or cerebrovascular disease, is one of the most common causes of death globally. It has been well established that tripartite motif-containing protein 7 Tripartite Motif-containing 7 (Trim7), as an E3 ubiquitin protein ligase, is involved in protein ubiquitination and thus regulating cellular proliferation. Moreover, TRIM7 is upregulated in advanced carotid AS. However, the detailed mechanism of TRIM7 on regulation of AS remains unclear. In the present study, we firstly discovered that TRIM7 expression was robustly induced in platelet-derived growth factor type BB-treated vascular smooth muscle cells (VSMCs) and human atherosclerotic plaques. Functional approaches established that knockdown of TRIM7 inhibited proliferation and migration of VSMCs, as well as arrested the cell cycle at G1-S, thus suppressing AS progression. Our results also identified that c-Jun/activator protein 1 (AP-1) signaling pathway was activated by TRIM7. Moreover, gain- and loss-of-function studies revealed that TRIM7 could promote proliferation and migration of VSMCs via activation of c-Jun/AP-1 signaling pathway. Finally, by using atherogenic apolipoprotein E-deficient (apoE-/-) C57BL/6 mice with high-fat diet AS model, we demonstrated that interference of TRIM7 could effectively mitigate in vivo AS via inactivation of c-Jun/AP-1 signaling pathway. In general, activation of c-Jun/AP-1 signaling pathway via TRIM7 could be an important mechanism in AS progression, thus shedding light on the development of novel therapeutics to the treatment of the disease.
Collapse
MESH Headings
- Animals
- Atherosclerosis/genetics
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Cell Movement
- Cell Proliferation
- Cells, Cultured
- Humans
- JNK Mitogen-Activated Protein Kinases/genetics
- JNK Mitogen-Activated Protein Kinases/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Knockout, ApoE
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Transcription Factor AP-1/genetics
- Transcription Factor AP-1/metabolism
- Tripartite Motif Proteins/genetics
- Tripartite Motif Proteins/metabolism
- Ubiquitin-Protein Ligases/genetics
- Ubiquitin-Protein Ligases/metabolism
- Ubiquitination
Collapse
Affiliation(s)
- Rongjing Ji
- Department of Cardiology, FuWai Central China Cardiovascular Hospital, Zhengzhou, China
- Department of Cardiology, Medical School of Jinzhou Medical University, Jinzhou, China
- Department of Cardiology, The People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Yuanyuan Gu
- Department of neurology, The Third People's Hospital of Zhengzhou, Zhengzhou, China
| | - Jing Zhang
- Department of Cardiology, FuWai Central China Cardiovascular Hospital, Zhengzhou, China
| | - Chuanyu Gao
- Department of Cardiology, FuWai Central China Cardiovascular Hospital, Zhengzhou, China
| | - Wanli Gao
- Department of Cardiology, FuWai Central China Cardiovascular Hospital, Zhengzhou, China
| | - Xiaobiao Zang
- Department of Cardiology, FuWai Central China Cardiovascular Hospital, Zhengzhou, China
| | - Yonghui Zhao
- Department of Cardiology, FuWai Central China Cardiovascular Hospital, Zhengzhou, China
- Department of Cardiology, The People's Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
78
|
Fan Q, Liu Y, Rao J, Zhang Z, Xiao W, Zhu T, Chai X, Ye K, Ning N, Yin Z, Chai Y, Xu Y, Lan R, Verkhratsky A, Nie H. Anti-Atherosclerosis Effect of Angong Niuhuang Pill via Regulating Th17/Treg Immune Balance and Inhibiting Chronic Inflammatory on ApoE -/- Mice Model of Early and Mid-Term Atherosclerosis. Front Pharmacol 2020; 10:1584. [PMID: 32082145 PMCID: PMC7005527 DOI: 10.3389/fphar.2019.01584] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 12/06/2019] [Indexed: 12/03/2022] Open
Abstract
Angong Niuhuang Pill (ANP) is a well-known patented Chinese medicine which is used for hundreds of years for treating the central nervous system diseases. Atherosclerosis is a poly-aetiological chronic inflammatory vascular disease. Preventing inflammation is fundamental for treating atherosclerosis in early stages. In this study, we investigated the protective effects and possible mechanisms of ANP action on a high-fat diet induced early and mid-term atherosclerosis ApoE-/- mice. The effects of ANP were compared with accepted drug simvastatin. Twelve male C57BL/6J mice were used as the control group, and 60 male ApoE-/- mice were randomly divided into five groups: Model group, Simvastatin group, Low-, Medium-, and High-dose ANP group these groups received, respectively, saline, simvastatin (3.0mg/kg), low-dose ANP (0.25 g/kg), medium-dose ANP (0.50 g/kg), and high-dose ANP (1.0 g/kg), once every other day for 10 weeks. After administration, serum biochemical indices were detected by the automatic biochemical analyzer, the concentrations of IL-6 and IL-10 in the serum were assayed by ELISA, expression levels of IL-1β, TNF-α, MMP-2, MMP-9, CCL2, and its receptor CCR2 in the full-length aorta, and expression levels of transcription factors Foxp3, RORγt in the spleen were assayed via western blotting and RT-qPCR. Flow cytometry was used to analyze Th17 cells and Treg cells. Pathological and histological analysis was completed on aortic root. ANP decreased LDL/HDL ratio, concentrations of IL-6 while increased IL-10 in serum. Moreover, ANP down-regulated the expression levels of IL-1β, TNF-α, MMP-2, MMP-9, CCL2, and CCR2 receptor in the full-length aorta. In addition, ANP decreased Th17 cells and expression levels of transcription factor RORγt, increased Treg cells and expression levels of transcription factor Foxp3. ANP decreased content of collagen fibers and infiltration of inflammatory cells in the aortic root. In conclusion, we demonstrated that ANP has anti-atherosclerosis effects on a high-fat diet induced ApoE-/- mice early and mid-term AS model via regulating Th17/Treg balance, inhibiting chronic inflammation, reducing plaque collagen fibers, and reducing inflammatory cells infiltration, to exert its multi-channel multi-target anti-early and mid-term AS effects.
Collapse
Affiliation(s)
- Qinghong Fan
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Yujuan Liu
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Jiaoyu Rao
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Zhe Zhang
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Wei Xiao
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Tao Zhu
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Xiaomeng Chai
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Kaihe Ye
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Na Ning
- Guangzhou Baiyunshan Zhongyi Pharmaceutical Co., Ltd, Guangzhou, China
| | - Zhen Yin
- Guangzhou Baiyunshan Zhongyi Pharmaceutical Co., Ltd, Guangzhou, China
| | - Yushuang Chai
- Guangzhou Baiyunshan Zhongyi Pharmaceutical Co., Ltd, Guangzhou, China
| | - Yimin Xu
- Guangzhou Baiyunshan Zhongyi Pharmaceutical Co., Ltd, Guangzhou, China
| | - Ruirui Lan
- International Department, The Affiliated High School of SCNU, Guangzhou, China
| | - A Verkhratsky
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Hong Nie
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China.,International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou, China
| |
Collapse
|
79
|
Monteiro EB, Soares EDR, Trindade PL, de Bem GF, Resende ADC, Passos MMCDF, Soulage CO, Daleprane JB. Uraemic toxin-induced inflammation and oxidative stress in human endothelial cells: protective effect of polyphenol-rich extract from açaí. Exp Physiol 2020; 105:542-551. [PMID: 31876965 DOI: 10.1113/ep088080] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 12/24/2019] [Indexed: 12/30/2022]
Abstract
NEW FINDINGS What is the central question of this study? Does a polyphenol-rich extract from açaí have a potential role in preventing uraemic toxin-induced endothelial cell dysfunction? What is the main finding and its importance? Polyphenols from açaí prevented cell death, restored migratory capacity, protected from inflammation and contributed to the restoration of the antioxidant response in endothelial cells exposed to uraemic toxins. The protective role of açaí against toxic effects exerted by uraemic toxins presents a potential new therapeutic target in endothelial cells. ABSTRACT In chronic kidney disease (CKD), progressive loss of kidney function results in the accumulation of protein-bound uraemic toxins such as p-cresyl sulfate (pCS) and indoxyl sulfate (IS). Among strategies to ameliorate the harmful actions of uraemic toxins, phenolic compounds have been extensively studied. The main goal of this work was to evaluate the antioxidant and anti-inflammatory actions of phenolic-rich açaí seed extract (ASE) in response to endothelial dysfunction induced by IS and pCS, in human umbilical vein endothelial cells (HUVECs). Cells were treated with ASE (10 µg ml-1 ) in the presence or absence of IS (61 µg ml-1 ) and pCS (40 µg ml-1 ). Cell viability, cell death, cell migratory capacity and inflammatory biomarker expression were evaluated. Cellular antioxidant response was measured through the activity and expression of antioxidant enzymes, and oxidative damage was evaluated. IS and pCS lowered cell viability, triggered cell death and lowered the migratory capacity in endothelial cells (P < 0.05). ASE prevented cell death and restored the migratory capacity in cells exposed to IS. Both toxins up-regulated pro-inflammatory cytokine expression, and ASE was able to beneficially counteract this effect. Tumour necrosis factor-α secretion was greater in uraemic toxin-treated cells and ASE reversed this phenomenon in cells treated with both toxins concomitantly (P < 0.05). With regard to the antioxidant response, superoxide dismutase expression was strikingly lower in cells treated with both toxins, and ASE inhibited this harmful effect (P < 0.05). From the results, we conclude that ASE exerted protective effects on inflammation and oxidative stress caused by uraemic toxins (particularly by IS) in human endothelial cells.
Collapse
Affiliation(s)
- Elisa Bernardes Monteiro
- Nutrition and Genomics Laboratory, Basic and Experimental Nutrition Department, Institute of Nutrition, Rio de Janeiro State University, Rio de Janeiro, Brazil.,Université de Lyon, INSERM U1060, CarMeN, INSA de Lyon, Univ Lyon-1, F-69621, Villeurbanne, France
| | - Elaine Dos Ramos Soares
- Nutrition and Genomics Laboratory, Basic and Experimental Nutrition Department, Institute of Nutrition, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Patrícia Letícia Trindade
- Nutrition and Genomics Laboratory, Basic and Experimental Nutrition Department, Institute of Nutrition, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Graziele Freitas de Bem
- Laboratory of Cardiovascular Pharmacology and Medicinal Plants, Department of Pharmacology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Angela de Castro Resende
- Laboratory of Cardiovascular Pharmacology and Medicinal Plants, Department of Pharmacology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Magna Maria Cottini da Fonseca Passos
- Nutrition and Genomics Laboratory, Basic and Experimental Nutrition Department, Institute of Nutrition, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | | | - Julio Beltrame Daleprane
- Nutrition and Genomics Laboratory, Basic and Experimental Nutrition Department, Institute of Nutrition, Rio de Janeiro State University, Rio de Janeiro, Brazil
| |
Collapse
|
80
|
Vigne J, Cabella C, Dézsi L, Rustique E, Couffin AC, Aid R, Anizan N, Chauvierre C, Letourneur D, Le Guludec D, Rouzet F, Hyafil F, Mészáros T, Fülöp T, Szebeni J, Cordaro A, Oliva P, Mourier V, Texier I. Nanostructured lipid carriers accumulate in atherosclerotic plaques of ApoE -/- mice. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2020; 25:102157. [PMID: 31982616 DOI: 10.1016/j.nano.2020.102157] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 12/04/2019] [Accepted: 01/18/2020] [Indexed: 12/20/2022]
Abstract
Nanostructured lipid carriers (NLC) might represent an interesting approach for the identification and targeting of rupture-prone atherosclerotic plaques. In this study, we evaluated the biodistribution, targeting ability and safety of 64Cu-fonctionalized NLC in atherosclerotic mice. 64Cu-chelating-NLC (51.8±3.1 nm diameter) with low dispersity index (0.066±0.016) were produced by high pressure homogenization at tens-of-grams scale. 24 h after injection of 64Cu-chelated particles in ApoE-/- mice, focal regions of the aorta showed accumulation of particles on autoradiography that colocalized with Oil Red O lipid mapping. Signal intensity was significantly greater in aortas isolated from ApoE-/- mice compared to wild type (WT) control (8.95 [7.58, 10.16]×108 vs 4.59 [3.11, 5.03]×108 QL/mm2, P < 0.05). Moreover, NLC seemed safe in relevant biocompatibility studies. NLC could constitute an interesting platform with high clinical translation potential for targeted delivery and imaging purposes in atherosclerosis.
Collapse
Affiliation(s)
- Jonathan Vigne
- Université de Paris, LVTS, INSERM U1148, Paris, France; Nuclear Medicine Department, X. Bichat Hospital, APHP and DHU FIRE, Paris, France; Université de Paris, UMS34 FRIM, Paris, France
| | - Claudia Cabella
- Centro Ricerche Bracco, Bracco Imaging SpA, Colleretto Giacosa, Italy
| | - László Dézsi
- Nanomedicine Research and Education Center, Institute of Pathophysiology, Semmelweis University, Budapest, Hungary
| | | | | | - Rachida Aid
- Université de Paris, UMS34 FRIM, Paris, France
| | | | | | | | - Dominique Le Guludec
- Université de Paris, LVTS, INSERM U1148, Paris, France; Nuclear Medicine Department, X. Bichat Hospital, APHP and DHU FIRE, Paris, France; Université de Paris, UMS34 FRIM, Paris, France
| | - François Rouzet
- Université de Paris, LVTS, INSERM U1148, Paris, France; Nuclear Medicine Department, X. Bichat Hospital, APHP and DHU FIRE, Paris, France; Université de Paris, UMS34 FRIM, Paris, France
| | - Fabien Hyafil
- Université de Paris, LVTS, INSERM U1148, Paris, France; Nuclear Medicine Department, X. Bichat Hospital, APHP and DHU FIRE, Paris, France; Université de Paris, UMS34 FRIM, Paris, France
| | - Tamás Mészáros
- Nanomedicine Research and Education Center, Institute of Pathophysiology, Semmelweis University, Budapest, Hungary
| | - Tamás Fülöp
- Nanomedicine Research and Education Center, Institute of Pathophysiology, Semmelweis University, Budapest, Hungary
| | - János Szebeni
- Nanomedicine Research and Education Center, Institute of Pathophysiology, Semmelweis University, Budapest, Hungary
| | - Alessia Cordaro
- Centro Ricerche Bracco, Bracco Imaging SpA, Colleretto Giacosa, Italy
| | - Paolo Oliva
- Centro Ricerche Bracco, Bracco Imaging SpA, Colleretto Giacosa, Italy
| | | | | |
Collapse
|
81
|
Andreadou I, Schulz R, Badimon L, Adameová A, Kleinbongard P, Lecour S, Nikolaou PE, Falcão-Pires I, Vilahur G, Woudberg N, Heusch G, Ferdinandy P. Hyperlipidaemia and cardioprotection: Animal models for translational studies. Br J Pharmacol 2020; 177:5287-5311. [PMID: 31769007 DOI: 10.1111/bph.14931] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 10/30/2019] [Accepted: 11/06/2019] [Indexed: 12/12/2022] Open
Abstract
Hyperlipidaemia is a well-established risk factor for cardiovascular diseases and therefore, many animal model have been developed to mimic the human abnormal elevation of blood lipid levels. In parallel, extensive research for the alleviation of ischaemia/reperfusion injury has revealed that hyperlipidaemia is a major co-morbidity that attenuates the cardioprotective effect of conditioning strategies (preconditioning, postconditioning and remote conditioning) and that of pharmacological interventions by interfering with cardioprotective signalling pathways. In the present review article, we summarize the existing data on animal models of hypercholesterolaemia (total, low density and HDL abnormalities) and hypertriglyceridaemia used in ischaemia/reperfusion injury and protection from it. We also provide recommendations on preclinical animal models to be used for translations of the cardioprotective strategies into clinical practice. LINKED ARTICLES: This article is part of a themed issue on Risk factors, comorbidities, and comedications in cardioprotection. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v177.23/issuetoc.
Collapse
Affiliation(s)
- Ioanna Andreadou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Rainer Schulz
- Institute for Physiology, Justus-Liebig University Giessen, Giessen, Germany
| | - Lina Badimon
- Cardiovascular Program ICCC, Research Institute-Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain.,CIBERCV, Instituto Salud Carlos III, Madrid, Spain.,Cardiovascular Research Chair Autonomous University of Barcelona (UAB), Barcelona, Spain
| | - Adriana Adameová
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University in Bratislava, Bratislava, Slovak Republic.,Center of Experimental Medicine, Slovak Academy of Sciences, Institute for Heart Research, Bratislava, Slovak Republic
| | - Petra Kleinbongard
- Institut für Pathophysiologie, Westdeutsches Herz- und Gefäßzentrum, Universitätsklinikum Essen, Essen, Germany
| | - Sandrine Lecour
- Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | | | - Ines Falcão-Pires
- Unidade de Investigação Cardiovascular, Departamento de Cirurgia e Fisiologia, Faculdade de Medicina, Universidade do Porto, Porto, Portugal
| | - Gemma Vilahur
- Cardiovascular Program ICCC, Research Institute-Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain.,CIBERCV, Instituto Salud Carlos III, Madrid, Spain
| | - Nicholas Woudberg
- Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Gerd Heusch
- Institut für Pathophysiologie, Westdeutsches Herz- und Gefäßzentrum, Universitätsklinikum Essen, Essen, Germany
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.,Pharmahungary Group, Szeged, Hungary
| |
Collapse
|
82
|
Scarcello E, Herpain A, Tomatis M, Turci F, Jacques P, Lison D. Hydroxyl radicals and oxidative stress: the dark side of Fe corrosion. Colloids Surf B Biointerfaces 2020; 185:110542. [DOI: 10.1016/j.colsurfb.2019.110542] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 09/07/2019] [Accepted: 10/01/2019] [Indexed: 11/29/2022]
|
83
|
Scarcello E, Lison D. Are Fe-Based Stenting Materials Biocompatible? A Critical Review of In Vitro and In Vivo Studies. J Funct Biomater 2019; 11:jfb11010002. [PMID: 31877701 PMCID: PMC7151573 DOI: 10.3390/jfb11010002] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 12/18/2019] [Accepted: 12/19/2019] [Indexed: 02/06/2023] Open
Abstract
Fe-based materials have increasingly been considered for the development of biodegradable cardiovascular stents. A wide range of in vitro and in vivo studies should be done to fully evaluate their biocompatibility. In this review, we summarized and analyzed the findings and the methodologies used to assess the biocompatibility of Fe materials. The majority of investigators drew conclusions about in vitro Fe toxicity based on indirect contact results. The setup applied in these tests seems to overlook the possible effects of Fe corrosion and does not allow for understanding of the complexity of released chemical forms and their possible impact on tissue. It is in particular important to ensure that test setups or interpretations of in vitro results do not hide some important mechanisms, leading to inappropriate subsequent in vivo experiments. On the other hand, the sample size of existing in vivo implantations is often limited, and effects such as local toxicity or endothelial function are not deeply scrutinized. The main advantages and limitations of in vitro design strategies applied in the development of Fe-based alloys and the correlation with in vivo studies are discussed. It is evident from this literature review that we are not yet ready to define an Fe-based material as safe or biocompatible.
Collapse
|
84
|
Cornelissen A, Simsekyilmaz S, Liehn E, Rusu M, Schaaps N, Afify M, Florescu R, Almalla M, Borinski M, Vogt F. Apolipoprotein E deficient rats generated via zinc-finger nucleases exhibit pronounced in-stent restenosis. Sci Rep 2019; 9:18153. [PMID: 31796798 PMCID: PMC6890749 DOI: 10.1038/s41598-019-54541-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 10/22/2019] [Indexed: 12/17/2022] Open
Abstract
The long-term success of coronary stent implantation is limited by in-stent restenosis (ISR). In spite of a broad variety of animal models available, an ideal high-throughput model of ISR has been lacking. Apolipoprotein E (apoE) deficient rats enable the evaluation of human-sized coronary stents while at the same time providing an atherogenic phenotype. Whereas apoE deficient rats have been proposed as animal model of atherosclerosis, to date it is unknown whether they also develop pronounced ISR. We sought to assess ISR after abdominal aorta stent implantation in apoE deficient rats. A total of 42 rats (16 wildtype, 13 homozygous apoE−/− and 13 heterozygous apoE+/− rats) underwent abdominal aorta stent implantation. After 28 days blood samples were analyzed to characterize lipid profiles. ISR was assessed by histomorphometric means. Homozygous apoE−/− rats exhibited significantly higher total cholesterol and low-density cholesterol levels than wildtype apoE+/+ and heterozygous apoE+/− rats. ISR was significantly pronounced in homozygous apoE−/− rats as compared to wildtype apoE+/+ (p = <0.0001) and heterozygous apoE+/− rats (p = 0.0102) on western diet. Abdominal aorta stenting of apoE−/− rats is a reliable model to investigate ISR after stent implantation and thus can be used for the evaluation of novel stent concepts. Apolipoprotein E (apoE) deficient rats have been proposed as animal model of atherosclerosis. We investigated the development of restenosis 28 days after stent implantation into the abdominal aorta of wildtype apoE+/+, homozygous apoE−/− and heterozygous apoE+/− rats, respectively. Homozygous apoE−/− rats exhibited significantly higher LDL and significantly lower HDL cholesterol levels compared to wildtype apoE+/+ and heterozygous apoE+/− rats. Restenosis after stent implantation was significantly pronounced in western-diet-fed homozygous apoE−/− rats, accompanied by a significantly increased neointimal thickness. Thus, apoE knockout rats exhibit elevated restenosis and might provide a novel tool for testing of innovative stent concepts.
Collapse
Affiliation(s)
- Anne Cornelissen
- University Hospital RWTH Aachen, Division of Cardiology, Pneumology, Angiology and Critical Care, Aachen, Germany.
| | - Sakine Simsekyilmaz
- University Hospital RWTH Aachen, Division of Cardiology, Pneumology, Angiology and Critical Care, Aachen, Germany
| | - Elisa Liehn
- University Hospital RWTH Aachen, Division of Cardiology, Pneumology, Angiology and Critical Care, Aachen, Germany
| | - Mihaela Rusu
- University Hospital RWTH Aachen, Division of Cardiology, Pneumology, Angiology and Critical Care, Aachen, Germany
| | - Nicole Schaaps
- University Hospital RWTH Aachen, Division of Cardiology, Pneumology, Angiology and Critical Care, Aachen, Germany
| | - Mamdouh Afify
- University Hospital RWTH Aachen, Division of Cardiology, Pneumology, Angiology and Critical Care, Aachen, Germany
| | - Roberta Florescu
- University Hospital RWTH Aachen, Division of Cardiology, Pneumology, Angiology and Critical Care, Aachen, Germany
| | - Mohammad Almalla
- University Hospital RWTH Aachen, Division of Cardiology, Pneumology, Angiology and Critical Care, Aachen, Germany
| | - Mauricio Borinski
- University Hospital RWTH Aachen, Division of Cardiology, Pneumology, Angiology and Critical Care, Aachen, Germany
| | - Felix Vogt
- University Hospital RWTH Aachen, Division of Cardiology, Pneumology, Angiology and Critical Care, Aachen, Germany
| |
Collapse
|
85
|
Berenji Ardestani S, Matchkov VV, Eftedal I, Pedersen M. A Single Simulated Heliox Dive Modifies Endothelial Function in the Vascular Wall of ApoE Knockout Male Rats More Than Females. Front Physiol 2019; 10:1342. [PMID: 31695628 PMCID: PMC6817487 DOI: 10.3389/fphys.2019.01342] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 10/09/2019] [Indexed: 12/19/2022] Open
Abstract
Introduction The number of divers is rising every year, including an increasing number of aging persons with impaired endothelial function and concomitant atherosclerosis. While diving is an independent modulator of endothelial function, little is known about how diving affects already impaired endothelium. In this study, we questioned whether diving exposure leads to further damage of an already impaired endothelium. Methods A total of 5 male and 5 female ApoE knockout (KO) rats were exposed to simulated diving to an absolute pressure of 600 kPa in heliox gas (80% helium, 20% oxygen) for 1 h in a dry pressure chamber. 10 ApoE KO rats (5 males, 5 females) and 8 male Sprague-Dawley rats served as controls. Endothelial function was examined in vitro by isometric myography of pulmonary and mesenteric arteries. Lipid peroxidation in blood plasma, heart and lung tissue was used as measures of oxidative stress. Expression and phosphorylation of endothelial NO synthase were quantified by Western blot. Results and Conclusion A single simulated dive was found to induce endothelial dysfunction in the pulmonary arteries of ApoE KO rats, and this was more profound in male than female rats. Endothelial dysfunction in males was associated with changing in production or bioavailability of NO; while in female pulmonary arteries an imbalance in prostanoid signaling was observed. No effect of diving was found on mesenteric arteries from rats of either sex. Our findings suggest that changes in endothelial dysfunction were specific for pulmonary circulation. In future, human translation of these findings may suggest caution for divers who are elderly or have prior reduced endothelial function.
Collapse
Affiliation(s)
- Simin Berenji Ardestani
- Department of Clinical Medicine, Comparative Medicine Lab, Aarhus University, Aarhus, Denmark.,Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, NTNU: Norwegian University of Science and Technology, Trondheim, Norway
| | | | - Ingrid Eftedal
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, NTNU: Norwegian University of Science and Technology, Trondheim, Norway.,Faculty of Nursing and Health Sciences, Nord University, Bodø, Norway
| | - Michael Pedersen
- Department of Clinical Medicine, Comparative Medicine Lab, Aarhus University, Aarhus, Denmark
| |
Collapse
|
86
|
Zhu L, Zhang N, Yan R, Yang W, Cong G, Yan N, Ma W, Hou J, Yang L, Jia S. Hyperhomocysteinemia induces vascular calcification by activating the transcription factor RUNX2 via Krüppel-like factor 4 up-regulation in mice. J Biol Chem 2019; 294:19465-19474. [PMID: 31628194 DOI: 10.1074/jbc.ra119.009758] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 10/16/2019] [Indexed: 01/09/2023] Open
Abstract
One of the main characteristics of atherosclerosis is vascular calcification, which is linked to adverse cardiovascular events. Increased homocysteine (Hcy), a feature of hyperhomocysteinemia, is correlated with advanced vascular calcification and phenotypic switching of vascular smooth muscle cells (VSMCs). Oxidative stress and high phosphate levels also induce VSMC calcification, suggesting that the Krüppel-like factor 4 (KLF4) signaling pathway may also contribute to vascular calcification. In this study, we investigated this possibility and the role and mechanisms of Hcy in vascular calcification. We found that in atherosclerotic apolipoprotein E-deficient (ApoE-/-) mice, Hcy significantly increases vascular calcification in vivo, as well as VSMC calcification in vitro Of note, the Hcy-induced VSMC calcification was correlated with elevated KLF4 levels. Hcy promoted KLF4 expression in calcified atherosclerotic lesions in vivo and in calcified VSMCs in vitro shRNA-mediated KLF4 knockdown blocked the Hcy-induced up-regulation of runt-related transcription factor 2 (RUNX2) and VSMC calcification. RUNX2 inhibition abolished Hcy-induced VSMC calcification. Using ChIP analysis, we demonstrate that KLF4 interacts with RUNX2, an interaction promoted by Hcy stimulation. Our experiments also revealed that the KLF4 knockdown attenuates Hcy-induced RUNX2 transactivity, indicating that KLF4 is important in modulating RUNX2 transactivity. These findings support a role for Hcy in regulating vascular calcification through a KLF4-RUNX2 interaction and indicate that Hcy-induced, enhanced RUNX2 transactivity increases VSMC calcification. These insights reveal possible opportunities for developing interventions that prevent or manage vascular calcification.
Collapse
Affiliation(s)
- Lili Zhu
- Department of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia 750001, China.,Institute of Cardiovascular Diseases, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750001, China
| | - Na Zhang
- Department of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia 750001, China
| | - Ru Yan
- Institute of Cardiovascular Diseases, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750001, China
| | - Wenjuan Yang
- Heart Centre, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750001, China
| | - Guangzhi Cong
- Heart Centre, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750001, China
| | - Ning Yan
- Heart Centre, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750001, China
| | - Wanrui Ma
- Department of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia 750001, China
| | - Jianjun Hou
- Department of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia 750001, China
| | - Libo Yang
- Department of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia 750001, China
| | - Shaobin Jia
- Institute of Cardiovascular Diseases, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750001, China .,Heart Centre, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750001, China
| |
Collapse
|
87
|
Talib J, Hains PG, Tumanov S, Hodson MP, Robinson PJ, Stocker R. Barocycler-Based Concurrent Multiomics Method To Assess Molecular Changes Associated with Atherosclerosis Using Small Amounts of Arterial Tissue from a Single Mouse. Anal Chem 2019; 91:12670-12679. [PMID: 31509387 DOI: 10.1021/acs.analchem.9b01842] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Atherosclerosis is a complex, multifactorial disease characterized by the buildup of plaque in the arterial wall. Apolipoprotein E gene deficient (Apoe-/-) mice serve as a commonly used tool to elucidate the pathophysiology of atherosclerosis because of their propensity to spontaneously develop arterial lesions. To date, however, an integrated omics assessment of atherosclerotic lesions in individual Apoe-/- mice has been challenging because of the small amount of diseased and nondiseased tissue available. To address this current limitation, we developed a multiomics method (Multi-ABLE) based on the proteomic method called accelerated Barocycler lysis and extraction (ABLE) to assess the depth of information that can be obtained from arterial tissue derived from a single mouse by splitting ABLE to allow for a combined proteomics-metabolomics-lipidomics analysis (Multi-ABLE). The new method includes tissue lysis via pressure cycling technology (PCT) in a Barocycler, followed by proteomic analysis of half the sample by nanoLC-MS and sequential extraction of lipids (organic extract) and metabolites (aqueous extract) combined with HILIC and reversed phase chromatography and time-of-flight mass spectrometry on the other half. Proteomic analysis identified 845 proteins, 93 of which were significantly altered in lesion-containing arteries. Lipidomic and metabolomic analyses detected 851 lipid and 362 metabolite features, which included 215 and 65 identified lipids and metabolites, respectively. The Multi-ABLE method is the first to apply a concurrent multiomics pipeline to cardiovascular disease using small (<5 mg) tissue samples, and it is applicable to other diseases where limited size samples are available at specific points during disease progression.
Collapse
Affiliation(s)
- Jihan Talib
- Vascular Biology Division , Victor Chang Cardiac Research Institute , Lowy Packer Building, 405 Liverpool Street , Darlinghurst , New South Wales 2010 , Australia.,St Vincent's Clinical School , University of New South Wales Medicine , Camperdown , New South Wales 2050 , Australia
| | - Peter G Hains
- Cell Signalling Unit, Children's Medical Research Institute , The University of Sydney , 214 Hawkesbury Rd , Westmead , New South Wales 2145 , Australia
| | - Sergey Tumanov
- Vascular Biology Division , Victor Chang Cardiac Research Institute , Lowy Packer Building, 405 Liverpool Street , Darlinghurst , New South Wales 2010 , Australia
| | - Mark P Hodson
- Freedman Foundation Metabolomics Facility, Victor Chang Innovation Centre , Victor Chang Cardiac Research Institute , Lowy Packer Building, 405 Liverpool Street , Darlinghurst , New South Wales 2010 , Australia.,School of Pharmacy , University of Queensland , 20 Cornwall Street , Woolloongabba , Queensland 4102 , Australia
| | - Phillip J Robinson
- Cell Signalling Unit, Children's Medical Research Institute , The University of Sydney , 214 Hawkesbury Rd , Westmead , New South Wales 2145 , Australia
| | - Roland Stocker
- Vascular Biology Division , Victor Chang Cardiac Research Institute , Lowy Packer Building, 405 Liverpool Street , Darlinghurst , New South Wales 2010 , Australia.,St Vincent's Clinical School , University of New South Wales Medicine , Camperdown , New South Wales 2050 , Australia
| |
Collapse
|
88
|
Wang D, Yang Y, Lei Y, Tzvetkov NT, Liu X, Yeung AWK, Xu S, Atanasov AG. Targeting Foam Cell Formation in Atherosclerosis: Therapeutic Potential of Natural Products. Pharmacol Rev 2019; 71:596-670. [PMID: 31554644 DOI: 10.1124/pr.118.017178] [Citation(s) in RCA: 146] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Foam cell formation and further accumulation in the subendothelial space of the vascular wall is a hallmark of atherosclerotic lesions. Targeting foam cell formation in the atherosclerotic lesions can be a promising approach to treat and prevent atherosclerosis. The formation of foam cells is determined by the balanced effects of three major interrelated biologic processes, including lipid uptake, cholesterol esterification, and cholesterol efflux. Natural products are a promising source for new lead structures. Multiple natural products and pharmaceutical agents can inhibit foam cell formation and thus exhibit antiatherosclerotic capacity by suppressing lipid uptake, cholesterol esterification, and/or promoting cholesterol ester hydrolysis and cholesterol efflux. This review summarizes recent findings on these three biologic processes and natural products with demonstrated potential to target such processes. Discussed also are potential future directions for studying the mechanisms of foam cell formation and the development of foam cell-targeted therapeutic strategies.
Collapse
Affiliation(s)
- Dongdong Wang
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Yang Yang
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Yingnan Lei
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Nikolay T Tzvetkov
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Xingde Liu
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Andy Wai Kan Yeung
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Suowen Xu
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Atanas G Atanasov
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| |
Collapse
|
89
|
The lncRNA DAPK-IT1 regulates cholesterol metabolism and inflammatory response in macrophages and promotes atherogenesis. Biochem Biophys Res Commun 2019; 516:1234-1241. [DOI: 10.1016/j.bbrc.2019.06.113] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 06/20/2019] [Indexed: 01/07/2023]
|
90
|
An J, Naruse TK, Hinohara K, Soejima Y, Sawabe M, Nakagawa Y, Kuwahara K, Kimura A. MRTF-A regulates proliferation and survival properties of pro-atherogenic macrophages. J Mol Cell Cardiol 2019; 133:26-35. [DOI: 10.1016/j.yjmcc.2019.05.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 04/01/2019] [Accepted: 05/16/2019] [Indexed: 12/12/2022]
|
91
|
Li Y, Pan Y, Wu X, Li Y, Wang H, Zhu H, Jiang L. Dual-modality imaging of atherosclerotic plaques using ultrasmall superparamagnetic iron oxide labeled with rhodamine. Nanomedicine (Lond) 2019; 14:1935-1944. [PMID: 31355711 DOI: 10.2217/nnm-2019-0062] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Aim: The diagnosis of vulnerable atherosclerotic plaques remains challenging. This study labeled ultrasmall superparamagnetic iron oxide with rhodamine (USPIO-R) and evaluated USPIO-R for imaging atherosclerotic plaques. Methods: Apolipoprotein E-deficient mice were fed a high-fat diet and underwent MRI before and after an intravenous injection of USPIO-R. Subsequently, an aortic specimen from the mice was removed and sliced for fluorescence imaging and Prussian blue and immunofluorescent staining. Results: T2 signal loss appeared and persisted in the aortic plaque postinjection, and spontaneous fluorescence from the plaque was observed. The accumulated mechanism of USPIO-R by plaque was the macrophage internalization by Prussian blue and immunofluorescence. Conclusion: USPIO-R is a promising dual-modality probe for diagnosing and monitoring vulnerable atherosclerotic plaques.
Collapse
Affiliation(s)
- Yi Li
- Department of Nuclear Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, PR China
| | - Yutao Pan
- Department of Emergency & Trauma Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, PR China
| | - Xiaodong Wu
- Department of Nuclear Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, PR China
| | - Yuan Li
- Department of Nuclear Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, PR China
| | - Huoqiang Wang
- Department of Nuclear Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, PR China
| | - Hong Zhu
- Laboratory of Oral Microbiology, Shanghai Research Institute of Stomatology, Shanghai Key Laboratory of Stomatology, Ninth People’s Hospital, School of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, PR China
| | - Lei Jiang
- Department of Nuclear Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, PR China
| |
Collapse
|
92
|
Fimasartan reduces neointimal formation and inflammation after carotid arterial injury in apolipoprotein E knockout mice. Mol Med 2019; 25:33. [PMID: 31307370 PMCID: PMC6632006 DOI: 10.1186/s10020-019-0095-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 05/26/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The beneficial effects of angiotensin II type 1 receptor blockers (ARBs) on atherosclerosis have been demonstrated in numerous studies. We investigated the effects of fimasartan on reducing neointimal formation and systemic inflammation after carotid artery (CA) injury in Apolipoprotein E knockout (ApoE KO) mice. METHODS ApoE KO mice were randomly allocated to Group I (without CA injury), Group II (without CA injury + Fimasartan), Group III (CA injury), and Group IV (CA injury + Fimasartan). Fimasartan was orally administered everyday starting 3 days before iatrogenic left CA injury. RESULTS At 28 days, neointimal hyperplasia and the inflammatory cytokines including TNFα, IL-6, ICAM, and MMP-9 in the peripheral blood were significantly reduced in Groups II and IV compared to Groups I and III, respectively. All fimasartan-administered groups revealed significant increases of CD4+CD25+Foxp3+ regulatory T (Treg) cells with increased plasma levels of IL-10 and TGFβ. In addition, increased CD8+ T cells by fimasartan were correlated with reduced smooth muscle cell (SMC) proliferation in the neointima in Groups II and IV. Furthermore, the populations of Treg and CD8+ T cells in total splenocytes were increased in Groups II and IV compared to Groups I and III, respectively. The enlargement of spleens due to CA injury in the Group III was attenuated by fimasartan, as shown in the Group IV. These data indicate that fimasartan significantly reduced SMC proliferation in neointima and increased Treg cells in ApoE KO CA injury mice. CONCLUSIONS This study suggests fimasartan could be an efficient strategy for reduction of atherosclerotic progression, with a decrease in immune response and systemic inflammation.
Collapse
|
93
|
Miceli M, Baldi D, Cavaliere C, Soricelli A, Salvatore M, Napoli C. Peripheral artery disease: the new frontiers of imaging techniques to evaluate the evolution of regenerative medicine. Expert Rev Cardiovasc Ther 2019; 17:511-532. [PMID: 31220944 DOI: 10.1080/14779072.2019.1635012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Introduction: Stem cells (ESC, iPSC, MSC) are known to have intrinsic regenerative properties. In the last decades numerous findings have favored the development of innovative therapeutic protocols based on the use of stem cells (Regenerative Medicine/Cell Therapy) for the treatment of numerous diseases including PAD, with promising results in preclinical studies. So far, several clinical studies have shown a general improvement of the patient's clinical outcome, however they possess many critical issues caused by the non-randomized design of the limited number of patients examined, the type cells to be used, their dosage, the short duration of treatment and also their delivery strategy. Areas covered: In this context, the use of the most advanced molecular imaging techniques will allow the visualization of very important physio-pathological processes otherwise invisible with conventional techniques, such as angiogenesis, also providing important structural and functional data. Expert opinion: The new frontier of cell therapy applied to PAD, potentially able to stop or even the process that causes the disease, with particular emphasis on the clinical aspects that different types of cells involve and on the use of more innovative molecular imaging techniques now available.
Collapse
Affiliation(s)
| | | | | | - Andrea Soricelli
- a IRCCS SDN , Naples , Italy.,b Department of Exercise and Wellness Sciences , University of Naples Parthenope , Naples , Italy
| | | | - Claudio Napoli
- a IRCCS SDN , Naples , Italy.,c University Department of Advanced Medical and Surgical Sciences, Clinical Department of Internal Medicine and Specialty Medicine , Università degli Studi della Campania 'Luigi Vanvitelli' , Napes , Italy
| |
Collapse
|
94
|
Hyperlipidemia induces meibomian gland dysfunction. Ocul Surf 2019; 17:777-786. [PMID: 31201956 DOI: 10.1016/j.jtos.2019.06.002] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 05/28/2019] [Accepted: 06/10/2019] [Indexed: 11/20/2022]
Abstract
PURPOSE To investigate the pathological changes of the meibomian gland (MG) and ocular surface in Apolipoprotein E knockout (ApoE-/-) mice and to investigate the association of meibomian gland dysfunction (MGD) with hyperlipidemia. METHODS Total plasma cholesterol was measured in different ages of ApoE-/- and wild type (WT) mice, whilst the ocular surfaces were observed by slit-lamp biomicroscopy. MG sections were subjected to H&E staining, Oil Red O staining, TUNEL assay and immunostaining. Quantitate RT-PCR and Western blot analyses were performed to detect the relative gene expression in MGs. The 5-month-old ApoE-/- mice were administered with rosiglitazone or GW9662 + rosiglitazone via oral gavage for 2 months to determine their effect on MG pathological change. RESULTS We found eyelid abnormality, MG dropout, abnormal MG acinar morphology, dilated MG duct and plugging of the MG orifice in ApoE-/- mice. MG acini in ApoE-/- mice showed exaggerated lipid accumulation. Abnormal keratinization increased in MG duct, accompanied with decreased proliferation and increased apoptosis in ApoE-/- mice. Inflammatory cells infiltrated into the surrounding microenvironment of MG acini, and the NF-κB signaling pathway was activated in MG acinar cells. Oxidative stress was evident in MG acinar cells of ApoE-/- mice. Further investigation showed downregulation of PPAR-γ in MG acinar cells of ApoE-/- mice. PPAR-γ agonist rosiglitazone treatment reduced the morbidity of eyelid, as well as corneal pathological changes and MG inflammation in ApoE-/- mice. CONCLUSION MGD and hyperlipidemia are closely associated in ApoE-/- mice, which represent a new model to study the pathophysiology of MGD related to dyslipidemia.
Collapse
|
95
|
Gwon SY, Lee HM, Rhee KJ, Sung HJ. Microarray and proteome array in an atherosclerosis mouse model for identification of biomarkers in whole blood. Int J Med Sci 2019; 16:882-892. [PMID: 31337962 PMCID: PMC6643112 DOI: 10.7150/ijms.30082] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 05/02/2019] [Indexed: 11/24/2022] Open
Abstract
Cardiovascular disease (CVD) is highly fatal, and 80 percent of the mortality is attributed to heart attack and stroke. Atherosclerosis is a disease that increases a patient's risk to CVD and is characterized by atheroma formed by immune cells, lipids, and smooth muscle cells. When an atherosclerotic lesion grows and blocks blood vessels or when an atheroma ruptures and blocks blood vessels by embolism, sudden angina, or stroke can occur. It is therefore important to diagnose atherosclerosis early and prevent its progression to more severe disease. Although myeloperoxidase, plasma fibrinogen, cardiac troponin-I, and C-reactive protein have been considered as diagnostic markers for multiple cardiac risks, specific biomarkers for atherosclerosis have not been clearly determined yet. Particularly, reliable biomarkers for the diagnosis of atherosclerosis using whole blood are not yet available. In this study, we screened potential biomarker genes and proteins from whole blood of apolipoprotein E knockout (ApoE-/- ) mice maintained on a Western diet, by comparing them to ApoE+/+ mice. We used whole blood for microarray and proteome array. Candidate genes and proteins identified from each method were confirmed with quantitative real-time PCR and ELISA. Based on our data, we speculate that Lilrb4a, n-R5s136, and IL-5 are potential targets that can be developed into novel biomarkers of atherosclerosis. Our study contributes to the diagnosis of atherosclerosis using whole blood in clinical settings.
Collapse
Affiliation(s)
- Sun-Yeong Gwon
- Department of Biomedical Laboratory Science, College of Health Science, Eulji University, Seongnam-si, Gyeonggi-do, 13135, Republic of Korea
- Department of Biomedical Laboratory Science, College of Health Sciences, Yonsei University at Wonju, Wonju, Gangwon-do 26493
| | - Hae Min Lee
- Department of Senior Healthcare, BK21 plus Program, Graduated School, Eulji University, Daejeon, 34824, Republic of Korea
| | - Ki-Jong Rhee
- Department of Biomedical Laboratory Science, College of Health Sciences, Yonsei University at Wonju, Wonju, Gangwon-do 26493
| | - Ho Joong Sung
- Department of Biomedical Laboratory Science, College of Health Science, Eulji University, Seongnam-si, Gyeonggi-do, 13135, Republic of Korea
- Department of Senior Healthcare, BK21 plus Program, Graduated School, Eulji University, Daejeon, 34824, Republic of Korea
| |
Collapse
|
96
|
The Role of ApoE in HCV Infection and Comorbidity. Int J Mol Sci 2019; 20:ijms20082037. [PMID: 31027190 PMCID: PMC6515466 DOI: 10.3390/ijms20082037] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 04/21/2019] [Accepted: 04/23/2019] [Indexed: 02/07/2023] Open
Abstract
Hepatitis C virus (HCV) is an RNA virus that can efficiently establish chronic infection in humans. The overlap between the HCV replication cycle and lipid metabolism is considered to be one of the primary means by which HCV efficiently develops chronic infections. In the blood, HCV is complex with lipoproteins to form heterogeneous lipo-viro-particles (LVPs). Furthermore, apolipoprotein E (ApoE), which binds to receptors during lipoprotein transport and regulates lipid metabolism, is localized on the surface of LVPs. ApoE not only participate in the attachment and entry of HCV on the cell surface but also the assembly and release of HCV viral particles from cells. Moreover, in the blood, ApoE can also alter the infectivity of HCV and be used by HCV to escape recognition by the host immune system. In addition, because ApoE can also affect the antioxidant and immunomodulatory/anti-inflammatory properties of the host organism, the long-term binding and utilization of host ApoE during chronic HCV infection not only leads to liver lipid metabolic disorders but may also lead to increased morbidity and mortality associated with systemic comorbidities.
Collapse
|
97
|
Latifi Y, Moccetti F, Wu M, Xie A, Packwood W, Qi Y, Ozawa K, Shentu W, Brown E, Shirai T, McCarty OJ, Ruggeri Z, Moslehi J, Chen J, Druker BJ, López JA, Lindner JR. Thrombotic microangiopathy as a cause of cardiovascular toxicity from the BCR-ABL1 tyrosine kinase inhibitor ponatinib. Blood 2019; 133:1597-1606. [PMID: 30692122 PMCID: PMC6450432 DOI: 10.1182/blood-2018-10-881557] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 01/16/2019] [Indexed: 01/13/2023] Open
Abstract
The third-generation tyrosine kinase inhibitor (TKI) ponatinib has been associated with high rates of acute ischemic events. The pathophysiology responsible for these events is unknown. We hypothesized that ponatinib produces an endothelial angiopathy involving excessive endothelial-associated von Willebrand factor (VWF) and secondary platelet adhesion. In wild-type mice and ApoE-/- mice on a Western diet, ultrasound molecular imaging of the thoracic aorta for VWF A1-domain and glycoprotein-Ibα was performed to quantify endothelial-associated VWF and platelet adhesion. After treatment of wild-type mice for 7 days, aortic molecular signal for endothelial-associated VWF and platelet adhesion were five- to sixfold higher in ponatinib vs sham therapy (P < .001), whereas dasatinib had no effect. In ApoE-/- mice, aortic VWF and platelet signals were two- to fourfold higher for ponatinib-treated compared with sham-treated mice (P < .05) and were significantly higher than in treated wild-type mice (P < .05). Platelet and VWF signals in ponatinib-treated mice were significantly reduced by N-acetylcysteine and completely eliminated by recombinant ADAMTS13. Ponatinib produced segmental left ventricular wall motion abnormalities in 33% of wild-type and 45% of ApoE-/- mice and corresponding patchy perfusion defects, yet coronary arteries were normal on angiography. Instead, a global microvascular angiopathy was detected by immunohistochemistry and by intravital microscopy observation of platelet aggregates and nets associated with endothelial cells and leukocytes. Our findings reveal a new form of vascular toxicity for the TKI ponatinib that involves VWF-mediated platelet adhesion and a secondary microvascular angiopathy that produces ischemic wall motion abnormalities. These processes can be mitigated by interventions known to reduce VWF multimer size.
Collapse
Affiliation(s)
| | | | - Melinda Wu
- Knight Cardiovascular Institute
- Doernbecher Children's Hospital, and
| | | | | | - Yue Qi
- Knight Cardiovascular Institute
| | | | | | | | - Toshiaki Shirai
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR
| | - Owen J McCarty
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR
| | - Zaverio Ruggeri
- Department of Molecular and Experimental Medicine, Scripps Research Institute, La Jolla, CA
| | - Javid Moslehi
- Cardiovascular Division, Vanderbilt University, Nashville, TN
| | | | - Brian J Druker
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR; and
| | | | - Jonathan R Lindner
- Knight Cardiovascular Institute
- Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR
| |
Collapse
|
98
|
Yang Z, Li F, Yelamanchili D, Zeng Z, Rosales C, Youker KA, Shen H, Ferrari M, Mahmarian J, Pownall HJ, Hamilton DJ, Li Z. Vulnerable Atherosclerotic Plaque Imaging by Small‐Molecule High‐Affinity Positron Emission Tomography Radiopharmaceutical. ADVANCED THERAPEUTICS 2019. [DOI: 10.1002/adtp.201900005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Zhen Yang
- Center for BioenergeticsHouston Methodist Research Institute 6670 Bertner Avenue Houston TX 77030 USA
| | - Feng Li
- Center for BioenergeticsHouston Methodist Research Institute 6670 Bertner Avenue Houston TX 77030 USA
| | - Dedipya Yelamanchili
- Center for BioenergeticsHouston Methodist Research Institute 6670 Bertner Avenue Houston TX 77030 USA
| | - Zihua Zeng
- Department of Pathology & Genomic MedicineHouston Methodist Research Institute
| | - Corina Rosales
- Center for BioenergeticsHouston Methodist Research Institute 6670 Bertner Avenue Houston TX 77030 USA
| | - Keith A. Youker
- Houston Methodist DeBakey Heart & Vascular CenterHouston Methodist Research Institute
| | - Haifa Shen
- Department of NanomedicineHouston Methodist Research Institute
- Department of MedicineWeill Cornell Medical College 1300 York Avenue New York NY 10065 USA
| | - Mauro Ferrari
- Department of NanomedicineHouston Methodist Research Institute
- Department of MedicineWeill Cornell Medical College 1300 York Avenue New York NY 10065 USA
| | - John Mahmarian
- Houston Methodist DeBakey Heart & Vascular CenterHouston Methodist Research Institute
- Department of MedicineWeill Cornell Medical College 1300 York Avenue New York NY 10065 USA
| | - Henry J. Pownall
- Center for BioenergeticsHouston Methodist Research Institute 6670 Bertner Avenue Houston TX 77030 USA
- Department of MedicineWeill Cornell Medical College 1300 York Avenue New York NY 10065 USA
| | - Dale J. Hamilton
- Center for BioenergeticsHouston Methodist Research Institute 6670 Bertner Avenue Houston TX 77030 USA
- Department of MedicineWeill Cornell Medical College 1300 York Avenue New York NY 10065 USA
| | - Zheng Li
- Center for BioenergeticsHouston Methodist Research Institute 6670 Bertner Avenue Houston TX 77030 USA
- Department of RadiologyWeill Cornell Medical College 1300 York Avenue New York NY 10065 USA
| |
Collapse
|
99
|
Stasinopoulou M, Kadoglou NPE, Christodoulou E, Paronis E, Kostomitsopoulos NG, Valsami G, Liapis CD, Kakisis J. Statins’ Withdrawal Induces Atherosclerotic Plaque Destabilization in Animal Model—A “Rebound” Stimulation of Inflammation. J Cardiovasc Pharmacol Ther 2019; 24:377-386. [DOI: 10.1177/1074248419838499] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Marianna Stasinopoulou
- Center of Clinical, Experimental Surgery, and Translational Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Nikolaos P. E. Kadoglou
- Centre for Statistics in Medicine—Botnar Research Centre, University of Oxford, Oxford, United Kingdom
| | - Eirini Christodoulou
- Department of Pharmacy, Laboratory of Biopharmaceutics-Pharmacokinetics, National and Kapodistrian University of Athens, Athens, Greece
| | - Efthymios Paronis
- Center of Clinical, Experimental Surgery, and Translational Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Nikolaos G. Kostomitsopoulos
- Center of Clinical, Experimental Surgery, and Translational Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Georgia Valsami
- Department of Pharmacy, Laboratory of Biopharmaceutics-Pharmacokinetics, National and Kapodistrian University of Athens, Athens, Greece
| | - Christos D. Liapis
- Department of Vascular Surgery, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - John Kakisis
- Department of Vascular Surgery, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
100
|
Czaplińska M, Ćwiklińska A, Sakowicz-Burkiewicz M, Wieczorek E, Kuchta A, Kowalski R, Kortas-Stempak B, Dębska-Ślizień A, Jankowski M, Król E. Apolipoprotein E gene polymorphism and renal function are associated with apolipoprotein E concentration in patients with chronic kidney disease. Lipids Health Dis 2019; 18:60. [PMID: 30851738 PMCID: PMC6408819 DOI: 10.1186/s12944-019-1003-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 03/01/2019] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND Chronic kidney disease (CKD) associates with complex lipoprotein disturbances resulting in high cardiovascular risk. Apolipoprotein E (APOE) is a polymorphic protein with three common isoforms (E2; E3; E4) that plays a crucial role in lipoprotein metabolism, including hepatic clearance of chylomicrons and very low-density lipoprotein (VLDL) remnants, and reverse cholesterol transport. It demonstrates anti-atherogenic properties but data concerning the link between polymorphism and level of APOE in CKD patients are inconclusive. The aim of our research was to assess the relationship between APOE gene polymorphism and APOE concentration and its redistribution among lipoproteins along with CKD progression. METHODS 90 non-dialysed CKD patients were included into the study. Real time PCR was used for APOE genotyping. APOE level was measured in serum and in isolated lipoprotein fractions (VLDL; IDL + HDL; HDL). Kidney function was assessed using eGFR CKD-EPI formula. RESULTS The population was divided into three APOE genotype subgroups: E2(ε2ε3), E3(ε3ε3) and E4(ε3ε4). The highest APOE level was observed for the E2 subgroup (p < 0.001). APOE concentration positively correlated with eGFR value in the E2 subgroup (r = 0.7, p < 0.001) but inversely in the E3 subgroup (r = - 0.29, p = 0.02).). A lower concentration of APOE in the E2 subgroup was associated with its diminished contents in HDL and IDL + LDL particles. In the E3 subgroup, the higher concentration of APOE was related to the increased number of non-HDL lipoproteins. CONCLUSION In patients with CKD, APOE genotype as well as renal function are associated with the concentration of APOE and its redistribution among lipoprotein classes.
Collapse
Affiliation(s)
- Monika Czaplińska
- Clinic & Chair of Nephrology, Transplantology and Internal Medicine, Medical University of Gdańsk, Dębinki 7, 80-211 Gdańsk, Poland
| | - Agnieszka Ćwiklińska
- Department of Clinical Chemistry, Medical University of Gdańsk, Dębinki 7, 80-211 Gdańsk, Poland
| | | | - Ewa Wieczorek
- Department of Clinical Chemistry, Medical University of Gdańsk, Dębinki 7, 80-211 Gdańsk, Poland
| | - Agnieszka Kuchta
- Department of Clinical Chemistry, Medical University of Gdańsk, Dębinki 7, 80-211 Gdańsk, Poland
| | - Robert Kowalski
- Department of Therapy Monitoring and Pharmacogenetics, Medical University of Gdańsk, Dębinki 7, 80-211 Gdańsk, Poland
| | - Barbara Kortas-Stempak
- Department of Clinical Chemistry, Medical University of Gdańsk, Dębinki 7, 80-211 Gdańsk, Poland
| | - Alicja Dębska-Ślizień
- Clinic & Chair of Nephrology, Transplantology and Internal Medicine, Medical University of Gdańsk, Dębinki 7, 80-211 Gdańsk, Poland
| | - Maciej Jankowski
- Department of Clinical Chemistry, Medical University of Gdańsk, Dębinki 7, 80-211 Gdańsk, Poland
| | - Ewa Król
- Clinic & Chair of Nephrology, Transplantology and Internal Medicine, Medical University of Gdańsk, Dębinki 7, 80-211 Gdańsk, Poland
| |
Collapse
|