1
|
Bao Y, Zhang H, Wang D, Yan P, Shao S, Zhang Z, Liu B, Li N. The Pathological Factors Involved in Current In Vitro Atherosclerotic Models. TISSUE ENGINEERING. PART B, REVIEWS 2024; 30:530-544. [PMID: 38258801 DOI: 10.1089/ten.teb.2023.0272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Cardiovascular disease stemmed from atherosclerosis (AS) is well recognized to be the predominant cause of global death. To comprehensively clarify the pathogenesis of AS, exploit effective drugs, as well as develop therapeutic solutions, various atherosclerotic models were constructed in vitro and widely utilized by the scientific community. Compared with animal models, the in vitro atherosclerotic models play a prominent role not only in the targeted research of single pathological factor related to AS in the human derived system, but also in the combined study on multipathological factors leading to AS, thereby contributing tremendously to the in-depth elucidation of atherosclerotic pathological process. In the current review, a variety of pathological factors incorporated into the existing atherosclerotic models in vitro are broadly elaborated, including the pathological mechanism, in vitro simulation approaches, and the desired improvement perspectives for reproducing each pathological factor. In addition, this review also summarizes the advantages and disadvantages of current atherosclerotic models as well as their potential functionality. Finally, the promising aspects for future atherosclerotic models in vitro with potential advances are also discussed.
Collapse
Affiliation(s)
- Yuxin Bao
- School of Biomedical Engineering, Faculty of Medicine, Dalian University of Technology, Dalian, China
| | - Hangyu Zhang
- School of Biomedical Engineering, Faculty of Medicine, Dalian University of Technology, Dalian, China
| | - Danbo Wang
- Cancer Hospital of Dalian University of Technology, Shenyang, China
| | - Peishi Yan
- Department of Cardiology, Central Hospital of Dalian University of Technology, Dalian, China
| | - Shuai Shao
- School of Biomedical Engineering, Faculty of Medicine, Dalian University of Technology, Dalian, China
| | - Zhengyao Zhang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, China
| | - Bo Liu
- Cancer Hospital of Dalian University of Technology, Shenyang, China
- School of Basic Medical Sciences, Faculty of Medicine, Dalian University of Technology, Dalian, China
- Liaoning Key Lab of Integrated Circuit and Biomedical Electronic System, Dalian University of Technology, Dalian, China
| | - Na Li
- Cancer Hospital of Dalian University of Technology, Shenyang, China
- School of Basic Medical Sciences, Faculty of Medicine, Dalian University of Technology, Dalian, China
- Liaoning Key Lab of Integrated Circuit and Biomedical Electronic System, Dalian University of Technology, Dalian, China
| |
Collapse
|
2
|
Ahmed IU, Myerscough MR. HDL and plaque regression in a multiphase model of early atherosclerosis. Math Biosci 2024; 373:109208. [PMID: 38759951 DOI: 10.1016/j.mbs.2024.109208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 05/09/2024] [Accepted: 05/11/2024] [Indexed: 05/19/2024]
Abstract
Atherosclerosis is a chronic disease of the arteries characterised by the accumulation of lipids and lipid-engorged cells in the artery wall. Early plaque growth is aggravated by the deposition of low density lipoproteins (LDL) in the wall and the subsequent immune response. High density lipoproteins (HDL) counterbalance the effects of LDL by accepting cholesterol from macrophages and removing it from the plaque. In this paper, we develop a free boundary multiphase model to investigate the effects of LDL and HDL on early plaque development. We examine how the rates of LDL and HDL deposition affect cholesterol accumulation in macrophages, and how this impacts cell death rates and emigration. We identify a region of LDL-HDL parameter space where plaque growth stabilises for low LDL and high HDL influxes, due to macrophage emigration and HDL clearance that counterbalances the influx of new cells and cholesterol. We explore how the efferocytic uptake of dead cells and the recruitment of new macrophages affect plaque development for a range of LDL and HDL influxes. Finally, we consider how changes in the LDL-HDL profile can change the course of plaque development. We show that changes towards lower LDL and higher HDL can slow plaque growth and even induce regression. We find that these changes have less effect on larger, more established plaques, and that temporary changes will only slow plaque growth in the short term.
Collapse
Affiliation(s)
- Ishraq U Ahmed
- School of Mathematics and Statistics, University of Sydney, Australia.
| | - Mary R Myerscough
- School of Mathematics and Statistics, University of Sydney, Australia
| |
Collapse
|
3
|
Christensen JJ, Arnesen EK, Rundblad A, Telle-Hansen VH, Narverud I, Blomhoff R, Bogsrud MP, Retterstøl K, Ulven SM, Holven KB. Dietary fat quality, plasma atherogenic lipoproteins, and atherosclerotic cardiovascular disease: An overview of the rationale for dietary recommendations for fat intake. Atherosclerosis 2024; 389:117433. [PMID: 38219649 DOI: 10.1016/j.atherosclerosis.2023.117433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 11/29/2023] [Accepted: 12/20/2023] [Indexed: 01/16/2024]
Abstract
The scientific evidence supporting the current dietary recommendations for fat quality keeps accumulating; however, a paradoxical distrust has taken root among many researchers, clinicians, and in parts of the general public. One explanation for this distrust may relate to an incomplete overview of the totality of the evidence for the link between fat quality as a dietary exposure, and health outcomes such as atherosclerotic cardiovascular disease (ASCVD). Therefore, the main aim of the present narrative review was to provide a comprehensive overview of the rationale for dietary recommendations for fat intake, limiting our discussion to ASCVD as outcome. Herein, we provide a core framework - a causal model - that can help us understand the evidence that has accumulated to date, and that can help us understand new evidence that may become available in the future. The causal model for fat quality and ASCVD is comprised of three key research questions (RQs), each of which determine which scientific methods are most appropriate to use, and thereby which lines of evidence that should feed into the causal model. First, we discuss the link between low-density lipoprotein (LDL) particles and ASCVD (RQ1); we draw especially on evidence from genetic studies, randomized controlled trials (RCTs), epidemiology, and mechanistic studies. Second, we explain the link between dietary fat quality and LDL particles (RQ2); we draw especially on metabolic ward studies, controlled trials (randomized and non-randomized), and mechanistic studies. Third, we explain the link between dietary fat quality, LDL particles, and ASCVD (RQ3); we draw especially on RCTs in animals and humans, epidemiology, population-based changes, and experiments of nature. Additionally, the distrust over dietary recommendations for fat quality may partly relate to an unclear understanding of the scientific method, especially as applied in nutrition research, including the process of developing dietary guidelines. We therefore also aimed to clarify this process. We discuss how we assess causality in nutrition research, and how we progress from scientific evidence to providing dietary recommendations.
Collapse
Affiliation(s)
- Jacob J Christensen
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.
| | - Erik Kristoffer Arnesen
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Amanda Rundblad
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | | | - Ingunn Narverud
- Norwegian National Advisory Unit on Familial Hypercholesterolemia, Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital, Oslo, Norway
| | - Rune Blomhoff
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Martin P Bogsrud
- Unit for Cardiac and Cardiovascular Genetics, Oslo University Hospital, Oslo, Norway
| | - Kjetil Retterstøl
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway; The Lipid Clinic, Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital, Oslo, Norway
| | - Stine M Ulven
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Kirsten B Holven
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway; Norwegian National Advisory Unit on Familial Hypercholesterolemia, Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
4
|
Arya P, Sharma V, Thapliyal S, Sagar R, Singh P. Preclinical models of atherosclerosis: An overview. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2024; 27:535-542. [PMID: 38629090 PMCID: PMC11017846 DOI: 10.22038/ijbms.2024.74352.16148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 11/11/2023] [Indexed: 04/19/2024]
Abstract
Atherosclerosis is a primary cause of illness and death globally and its mechanism is still unclear. Different animal models have been created to evaluate the progression of atherosclerosis, allowing researchers to carefully control the circumstances of the experiment as well as the nutrition and environmental risk factors. To investigate the negative effects of various interventions, pathophysiological alterations might be generated utilizing genetic or pharmacological methods. These models' molecular and pathophysiological mechanisms have been clarified through experiments, and they have served as platforms for the creation of new drugs. Different models can be employed to address various research problems, each with its own benefits and drawbacks. In the current review study, various species of atherosclerosis models are discussed, along with the viability of using them in experiments.
Collapse
Affiliation(s)
- Priyanka Arya
- Galgotias College of Pharmacy, Greater Noida, U.P., India
| | - Vikram Sharma
- Galgotias College of Pharmacy, Greater Noida, U.P., India
| | - Surabhi Thapliyal
- Department of Pharmacology, All India Institute of Medical Sciences, Rishikesh 249203, India
| | | | - Priyanka Singh
- Galgotias College of Pharmacy, Greater Noida, U.P., India
| |
Collapse
|
5
|
Fu Y, Deng Y, Zhang J, Chua SL, Khoo BL. Biofilms exacerbate atherogenesis through macrophage-induced inflammatory responses in a fibrous plaque microsystem model. Acta Biomater 2023; 168:333-345. [PMID: 37385520 DOI: 10.1016/j.actbio.2023.06.028] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 06/19/2023] [Accepted: 06/20/2023] [Indexed: 07/01/2023]
Abstract
BACKGROUND Microbes have been implicated in atherosclerosis development and progression, but the impact of bacterial-based biofilms on fibrous plaque rupture remains poorly understood. RESULTS Here, we developed a comprehensive atherosclerotic model to reflect the progression of fibrous plaque under biofilm-induced inflammation (FP-I). High expressions of biofilm-specific biomarkers algD, pelA and pslB validated the presence of biofilms. Biofilm promotes the polarization of macrophages towards a pro-inflammatory (M1) phenotype, as demonstrated by an increase in M1 macrophage-specific marker CD80 expression in CD68+ macrophages. The increase in the number of intracellular lipid droplets (LDs) and foam cell percentage highlighted the potential role of biofilms on lipid synthesis or metabolic pathways in macrophage-derived foam cells. In addition, collagen I production by myofibroblasts associated with the fibrous cap was significantly reduced along with the promotion of apoptosis of myofibroblasts, indicating that biofilms affect the structural integrity of the fibrous cap and potentially undermine its strength. CONCLUSION We validated the unique role of biofilm-based inflammation in exacerbating fibrous plaque damage in the FP-I model, increasing fibrous plaque instability and risk of thrombosis. Our results lay the foundation for mechanistic studies of the role of biofilms in fibrous plaques, allowing the evaluation of preclinical combination strategies for drug therapy. STATEMENT OF SIGNIFICANCE A microsystem-based model was developed to reveal interactions in fibrous plaque during biofilm-induced inflammation (FP-I). Real-time assessment of biofilm formation and its role in fibrous plaque progression was achieved. The presence of biofilms enhanced the expression of pro-inflammatory (M1) specific marker CD80, lipid droplets, and foam cells and reduced anti-inflammatory (M2) specific marker CD206 expression. Fibrous plaque exposure to biofilm-based inflammation reduced collagen I expression and increased apoptosis marker Caspase-3 expression significantly. Overall, we demonstrate the unique role of biofilm-based inflammation in exacerbating fibrous plaque damage in the FP-I model, promoting fibrous plaque instability and enhanced thrombosis risk. Our findings lay the groundwork for mechanistic studies, facilitating the evaluation of preclinical drug combination strategies.
Collapse
Affiliation(s)
- Yatian Fu
- Department of Biomedical Engineering, City University of Hong Kong; Hong Kong Center for Cerebro-Cardiovascular Health Engineering (COCHE)
| | - Yanlin Deng
- Department of Biomedical Engineering, City University of Hong Kong
| | - Jing Zhang
- Department of Biomedical Engineering, City University of Hong Kong
| | - Song Lin Chua
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR China; State Key Laboratory of Chemical Biology and Drug Discovery, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR China; Shenzhen Key Laboratory of Food Biological Safety Control; Research Centre for Deep Space Explorations (RCDSE), The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR China
| | - Bee Luan Khoo
- Department of Biomedical Engineering, City University of Hong Kong; Hong Kong Center for Cerebro-Cardiovascular Health Engineering (COCHE); City University of Hong Kong - Futian Shenzhen Research Institute.
| |
Collapse
|
6
|
Srivastava RAK. A Review of Progress on Targeting LDL Receptor-Dependent and -Independent Pathways for the Treatment of Hypercholesterolemia, a Major Risk Factor of ASCVD. Cells 2023; 12:1648. [PMID: 37371118 DOI: 10.3390/cells12121648] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/10/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023] Open
Abstract
Since the discovery of the LDL receptor in 1973 by Brown and Goldstein as a causative protein in hypercholesterolemia, tremendous amounts of effort have gone into finding ways to manage high LDL cholesterol in familial hypercholesterolemic (HoFH and HeFH) individuals with loss-of-function mutations in the LDL receptor (LDLR) gene. Statins proved to be the first blockbuster drug, helping both HoFH and HeFH individuals by inhibiting the cholesterol synthesis pathway rate-limiting enzyme HMG-CoA reductase and inducing the LDL receptor. However, statins could not achieve the therapeutic goal of LDL. Other therapies targeting LDLR include PCSK9, which lowers LDLR by promoting LDLR degradation. Inducible degrader of LDLR (IDOL) also controls the LDLR protein, but an IDOL-based therapy is yet to be developed. Among the LDLR-independent pathways, such as angiopoietin-like 3 (ANGPTL3), apolipoprotein (apo) B, apoC-III and CETP, only ANGPTL3 offers the advantage of treating both HoFH and HeFH patients and showing relatively better preclinical and clinical efficacy in animal models and hypercholesterolemic individuals, respectively. While loss-of-LDLR-function mutations have been known for decades, gain-of-LDLR-function mutations have recently been identified in some individuals. The new information on gain of LDLR function, together with CRISPR-Cas9 genome/base editing technology to target LDLR and ANGPTL3, offers promise to HoFH and HeFH individuals who are at a higher risk of developing atherosclerotic cardiovascular disease (ASCVD).
Collapse
Affiliation(s)
- Rai Ajit K Srivastava
- Integrated Pharma Solutions LLC, Boston, MA 02101-02117, USA
- College of Professional Studies, Northeastern University, Boston, MA 02101-02117, USA
| |
Collapse
|
7
|
Poznyak AV, Sukhorukov VN, Eremin II, Nadelyaeva II, Orekhov AN. Diagnostics of atherosclerosis: Overview of the existing methods. Front Cardiovasc Med 2023; 10:1134097. [PMID: 37229223 PMCID: PMC10203409 DOI: 10.3389/fcvm.2023.1134097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 04/11/2023] [Indexed: 05/27/2023] Open
Abstract
Atherosclerosis was and remains an extremely common and serious health problem. Since the elderly are most at risk of cardiovascular risk, and the average life expectancy is increasing, the spread of atherosclerosis and its consequences increases as well. One of the features of atherosclerosis is its asymptomaticity. This factor makes it difficult to make a timely diagnosis. This entails the lack of timely treatment and even prevention. To date, in the arsenal of physicians, there is only a limited set of methods to suspect and fully diagnose atherosclerosis. In this review, we have tried to briefly describe the most common and effective methods for diagnosing atherosclerosis.
Collapse
|
8
|
Huang D, Gao W, Zhong X, Wu H, Zhou Y, Ma Y, Qian J, Ge J. Epigenetically altered macrophages promote development of diabetes-associated atherosclerosis. Front Immunol 2023; 14:1196704. [PMID: 37215106 PMCID: PMC10196132 DOI: 10.3389/fimmu.2023.1196704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 04/21/2023] [Indexed: 05/24/2023] Open
Abstract
Background Atherosclerosis (AS) risk is elevated in diabetic patients, but the underlying mechanism such as involvement of epigenetic control of foam macrophages remains unclear. We have previously shown the importance of immune regulation on endothelial cells to AS development in diabetes. In this study, we examined the hypothesis that diabetes may promote AS through modification of the epigenetic status of macrophages. Methods We employed the Laser Capture Microdissection (LCM) method to evaluate the expression levels of key epigenetic regulators in both endothelial cells and macrophages at the AS lesions of patients. We then assessed the correlation between the significantly altered epigenetic regulator and serum levels of low-density Lipoprotein (LDL), triglycerides (TRIG) and high-density Lipoprotein (HDL) in patients. In vitro, the effects of high glucose on glucose utilization, lactate production, succinate levels, oxygen consumption and polarization in either undifferentiated or differentiated bone marrow-derived macrophages (BMDMs) were analyzed. The effects of depleting this significantly altered epigenetic regulator in macrophages on AS development were assessed in AS-prone diabetic mice. Results Histone deacetylase 3 (HDAC3) was identified as the most significantly altered epigenetic regulator in macrophages from the AS lesions in human diabetic patients. The levels of HDAC3 positively correlated with high serum LDL and TRIG, as well as low serum HDL. High glucose significantly increased glucose utilization, lactate production, succinate levels and oxygen consumption in cultured macrophages, and induced proinflammatory M1-like polarization. Macrophage depletion of HDAC3 significantly attenuated AS severity in AS-prone diabetic mice. Conclusion Epigenetically altered macrophages promote development of diabetes-associated AS, which could be prevented through HDAC3 depletion.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Junbo Ge
- *Correspondence: Juying Qian, ; Junbo Ge,
| |
Collapse
|
9
|
Wang K, Han L, Tan Y, Hong H, Fan H, Luo Y. Novel Hypocholesterolemic Peptides Derived from Silver Carp Muscle: The Modulatory Effects on Enterohepatic Cholesterol Metabolism In Vitro and In Vivo. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:5565-5575. [PMID: 36997503 DOI: 10.1021/acs.jafc.2c09092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
This research aimed to investigate the effect of silver carp hydrolysates (SCHs) on hypercholesterolemia and enterohepatic cholesterol metabolism. Results showed that in vitro gastrointestinal digestion products of Alcalase-SCH (GID-Alcalase) exhibited the highest inhibitory activity of cholesterol absorption mainly through downregulating the expression of essential genes related to cholesterol transport in a Caco-2 monolayer. After being absorbed by the Caco-2 monolayer, GID-Alcalase increased the low-density lipoprotein (LDL) uptake in HepG2 cells by enhancing the protein level of the LDL receptor (LDLR). The in vivo experiment showed that long-term intervention of Alcalase-SCH ameliorated hypercholesterolemia in ApoE-/- mice fed with a Western diet (WD). After transepithelial transport, four novel peptides (TKY, LIL, FPK, and IAIM) were identified, and these peptides possessed dual hypocholesterolemic functions including inhibition of cholesterol absorption and promotion of peripheric LDL uptake. Our results indicated for the first time the potential of SCHs as functional food ingredients for the management of hypercholesterolemia.
Collapse
Affiliation(s)
- Kai Wang
- Beijing Laboratory for Food Quality and Safety, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Lihua Han
- Beijing Laboratory for Food Quality and Safety, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Yuqing Tan
- Beijing Laboratory for Food Quality and Safety, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Hui Hong
- Beijing Laboratory for Food Quality and Safety, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Hongbing Fan
- Department of Animal and Food Sciences, University of Kentucky, Lexington, Kentucky 40546, United States
| | - Yongkang Luo
- Beijing Laboratory for Food Quality and Safety, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
- National Research and Development Center for Freshwater Fish Processing, Jiangxi Normal University, Nanchang, Jiangxi 330022, China
| |
Collapse
|
10
|
Basheer S, Malik IR, Awan FR, Sughra K, Roshan S, Khalil A, Iqbal MJ, Parveen Z. Histological and Microscopic Analysis of Fats in Heart, Liver Tissue, and Blood Parameters in Experimental Mice. Genes (Basel) 2023; 14:515. [PMID: 36833442 PMCID: PMC9957320 DOI: 10.3390/genes14020515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 02/08/2023] [Accepted: 02/14/2023] [Indexed: 02/22/2023] Open
Abstract
The intake of various types and amounts of dietary fats influences metabolic and cardiovascular health. Hence, this study evaluated the impact of routinely consumed Pakistani dietary fats on their cardiometabolic impact. For this, we made four groups of mice, each comprising 5 animals: (1) C-ND: Control mice on a normal diet, (2) HFD-DG: High-fat diet mice on a normal diet plus 10% (w/w) desi ghee, (3) HFD-O: Mice on normal diet plus 10% (w/w) plant oil (4) HFD-BG: Mice on normal diet plus 10% (w/w) banaspati ghee. Mice were fed for 16 weeks, and blood, liver, and heart samples were collected for biochemical, histological, and electron microscopic analysis. The physical factors indicated that mice fed on HFD gained more body weight than the C-ND group. Blood parameters do not show significant differences, but overall, the glucose and cholesterol concentrations were raised in the mice fed with a fat-rich diet, with the highest concentrations in the HFD-BG group. The mice fed with HFD-BG and HFD-O had more lipid droplets in the liver, compared to HFD-DG and C-ND.
Collapse
Affiliation(s)
- Sehrish Basheer
- Department of Biotechnology, University of Sargodha, Sargodha 40100, Pakistan
- Diabetes and Cardio-Metabolic Disorders Lab, Health Biotechnology Division, National Institute for Biotechnology and Genetic Engineering (NIBGE), Jhang Road, Faisalabad 38000, Pakistan
| | - Imran Riaz Malik
- Department of Biotechnology, University of Sargodha, Sargodha 40100, Pakistan
| | - Fazli Rabbi Awan
- Diabetes and Cardio-Metabolic Disorders Lab, Health Biotechnology Division, National Institute for Biotechnology and Genetic Engineering (NIBGE), Jhang Road, Faisalabad 38000, Pakistan
| | - Kalsoom Sughra
- Department of Biochemistry and Biotechnology, University of Gujrat, Gujrat 50700, Pakistan
| | - Sadia Roshan
- Department of Zoology, University of Gujrat, Gujrat 50700, Pakistan
| | - Adila Khalil
- Diabetes and Cardio-Metabolic Disorders Lab, Health Biotechnology Division, National Institute for Biotechnology and Genetic Engineering (NIBGE), Jhang Road, Faisalabad 38000, Pakistan
| | - Muhammad Javed Iqbal
- Diabetes and Cardio-Metabolic Disorders Lab, Health Biotechnology Division, National Institute for Biotechnology and Genetic Engineering (NIBGE), Jhang Road, Faisalabad 38000, Pakistan
| | - Zahida Parveen
- Department of Biochemistry, Abdul Wali Khan University, Mardan 23200, Pakistan
| |
Collapse
|
11
|
Wahl L, Raschke M, Wittmann J, Regler A, Heelemann S, Brandsch C, Stangl GI, Vervuert I. Effects of atherogenic diet supplemented with fermentable carbohydrates on metabolic responses and plaque formation in coronary arteries using a Saddleback pig model. PLoS One 2022; 17:e0275214. [PMID: 36206259 PMCID: PMC9543622 DOI: 10.1371/journal.pone.0275214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 09/12/2022] [Indexed: 11/17/2022] Open
Abstract
Fermentable carbohydrates are gaining interest in the field of human nutrition because of their benefits in obesity-related comorbidities. The aim of this study was to investigate the influence of fermentable carbohydrates, such as pectin and inulin, in an atherogenic diet on metabolic responses and plaque formation in coronary arteries using a Saddleback pig model. Forty-eight healthy pigs aged five months were divided into four feeding groups (n = 10) and one baseline group (n = 8). Three feeding groups received an atherogenic diet (38% crisps, 10% palm fat, and 2% sugar with or without supplementation of 5% pectin or inulin), and one group received a conventional diet over 15 weeks. Feed intake, weight gain, body condition score, and back fat thickness were monitored regularly. Blood and fecal samples were collected monthly to assess the metabolites associated with high cardiovascular risk and fat content, respectively. At the end of 15 weeks, the coronary arteries of the pigs were analyzed for atherosclerotic plaque formation. Independent of supplementation, significant changes were observed in lipid metabolism, such as an increase in triglycerides, bile acids, and cholesterol in serum, in all groups fed atherogenic diets in comparison to the conventional group. Serum metabolome analysis showed differentiation of the feeding groups by diet (atherogenic versus conventional diet) but not by supplementation with pectin or inulin. Cardiovascular lesions were found in all feeding groups and in the baseline group. Supplementation of pectin or inulin in the atherogenic diet had no significant impact on cardiovascular lesion size. Saddleback pigs can develop naturally occurring plaques in coronary arteries. Therefore, this pig model offers potential for further research on the effects of dietary intervention on obesity-related comorbidities, such as cardiovascular lesions, in humans.
Collapse
Affiliation(s)
- Lisa Wahl
- Institute of Animal Nutrition, Nutrition Diseases and Dietetics, Leipzig University, Leipzig, Germany,Competence Cluster of Nutrition and Cardiovascular Health (nutriCARD), Halle-Jena-Leipzig, Germany
| | - Melina Raschke
- Competence Cluster of Nutrition and Cardiovascular Health (nutriCARD), Halle-Jena-Leipzig, Germany,Institute of Agricultural and Nutritional Science, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | | | | | | | - Corinna Brandsch
- Competence Cluster of Nutrition and Cardiovascular Health (nutriCARD), Halle-Jena-Leipzig, Germany,Institute of Agricultural and Nutritional Science, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Gabriele I. Stangl
- Competence Cluster of Nutrition and Cardiovascular Health (nutriCARD), Halle-Jena-Leipzig, Germany,Institute of Agricultural and Nutritional Science, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Ingrid Vervuert
- Institute of Animal Nutrition, Nutrition Diseases and Dietetics, Leipzig University, Leipzig, Germany,Competence Cluster of Nutrition and Cardiovascular Health (nutriCARD), Halle-Jena-Leipzig, Germany,* E-mail:
| |
Collapse
|
12
|
Zhu X, Zhao L, Wang Y, Hu X, Zhu Y, Yang X. Dietary titanium dioxide particles (E171) promote diet-induced atherosclerosis through reprogramming gut microbiota-mediated choline metabolism in APOE -/- mice. JOURNAL OF HAZARDOUS MATERIALS 2022; 436:129179. [PMID: 35739712 DOI: 10.1016/j.jhazmat.2022.129179] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 05/09/2022] [Accepted: 05/15/2022] [Indexed: 06/15/2023]
Abstract
Food-grade titanium dioxide (E171) has been reported to induce changes in some intestinal metabolites related to development of atherosclerosis (AS). However, little is known about the effects of chronic dietary intake of E171 on AS development, particularly in AS-prone populations with high-choline western diet (HCD). Herein, we disclosed that E171 obviously exacerbated HCD-induced AS through increasing production of trimethylamine (TMA) and pro-atherogenic trimethylamine-N-oxide (TMAO) via remodeling gut microbiota structure in APOE-/- mice. Oral administration of 40 mg/kg E171 daily for 4 months significantly increased the atherosclerotic lesion area, especially in the HCD group. Mechanistic studies revealed that E171 induced much more TMAO production by increasing the gut microbial expression of choline TMA lyases (CutC/D), which converted dietary choline to TMA by a glycyl radical reaction. The 16S rDNA sequencing analysis demonstrated that bacterial strains expressing CutC/D were enriched by E171 in HCD-fed mice. In contrast, gut microbiota depletion eliminated the impact of E171 on choline/TMA/TMAO pathway and AS progression, indicating gut flora shifts were responsible for the exacerbation effects of E171 ingestion on HCD-induced AS. All the results emphasized the alarming role of E171 on AS progression and stated the importance of reevaluating the impact of food additives on the development of chronic diseases.
Collapse
Affiliation(s)
- Xiaoqiang Zhu
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Lijun Zhao
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Yiqian Wang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xiuwen Hu
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Yanhong Zhu
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China.
| | - Xiangliang Yang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China.
| |
Collapse
|
13
|
da Silva WB, Teixeira ALDS, Oliveira KG, Gonzaga CN, de Souza AM, Almosny NRP. Lipid profile, apolipoproteins A-1 and B in owl monkeys (Aotus infulatus) in captivity. J Med Primatol 2022; 51:367-373. [PMID: 35916434 DOI: 10.1111/jmp.12607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 07/15/2022] [Accepted: 07/20/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Owl monkeys (Aotus infulatus) are frequently affected by heart diseases and, as in humans, dyslipidemia is one of the predisposing factors for adverse cardiovascular events. In view of this, the study of the lipid profile and plasma apolipoproteins can contribute to the clinical management of this neotropical primate species. METHODS Lipid profile as well as A-1 and B apolipoprotein values were analyzed in 60 owl monkeys, studying their relationship with body biometry and the presence of cardiac alterations. RESULTS Animals suspected of having heart disease did not show significant differences (p < .05) in terms of biometry or in relation to lipid profile and apolipoproteins A-1 and B values; however, higher values of LDL and ApoB and ApoB/ApoA-1 were observed in this group. CONCLUSIONS This study is the first to describe the lipid profile and apolipoprotein values in owl monkeys, and further work will be needed to better elucidate the worthiness of LDL, ApoB, and the ApoB/ApoA-1 ratio in this primate species.
Collapse
Affiliation(s)
- Wellington Bandeira da Silva
- Faculty of Veterinary Medicine, Fluminense Federal University, Niterói, Brazil.,National Primate Center/Evandro Chagas Institute, Ananindeua, Brazil
| | | | | | | | | | | |
Collapse
|
14
|
Wang K, Fu Z, Li X, Hong H, Zhan X, Guo X, Luo Y, Tan Y. Whey protein hydrolysate alleviated atherosclerosis and hepatic steatosis by regulating lipid metabolism in apoE -/- mice fed a Western diet. Food Res Int 2022; 157:111419. [PMID: 35761665 DOI: 10.1016/j.foodres.2022.111419] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 05/05/2022] [Accepted: 05/24/2022] [Indexed: 11/16/2022]
Abstract
Whey protein hydrolysate (WPH) has been proved to possess various biological activities associated with the amelioration of cardiovascular disease (CVD). The objective of this study was to investigate the anti-atherosclerotic and hepatoprotective effects of WPH on apolipoprotein E knockout (apoE-/-) mice fed with a Western diet for 15 weeks. Results revealed that WPH markedly inhibited the development of atherosclerotic lesions in the aorta and steatosis injury in the liver. The serum lipid and inflammation levels were both reduced after WPH supplemented in apoE-/- mice. In addition, WPH inhibited the lipid accumulation in the liver, thereby decreasing the hepatic inflammation level and oxidative stress injury. Mechanism investigation revealed that WPH down-regulated the expression of cholesterol biosynthesis genes while up-regulated the expression of cholesterol uptake and excretion genes in the liver. Meanwhile, the de novo lipogenesis was inhibited while the fatty acids β-oxidation was activated in the liver by WPH supplementation. Notably, the n-3 polyunsaturated fatty acid (PUFA)/n-6 PUFA ratio in serum and liver of the WPH-H group were 2.69-fold (p < 0.01) and 3.64-fold (p < 0.01) higher than that of the Model group. Collectively, our results proved WPH possesses potent anti-atherosclerotic and hepatoprotective activities and has the potential to be used as a novel functional ingredient for the management of CVD.
Collapse
Affiliation(s)
- Kai Wang
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China.
| | - Zixin Fu
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China.
| | - Xiaoyi Li
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China.
| | - Hui Hong
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China.
| | - Xin Zhan
- Tianjin Milkyway Import and Export Corp, Tianjin 300457, China.
| | - Xiaohong Guo
- Hebei Dongkang Dairy Co., Ltd, Shijiazhuang 052160, China.
| | - Yongkang Luo
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China.
| | - Yuqing Tan
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China.
| |
Collapse
|
15
|
Liu J, Zhang W, Li Y, Li X, Li Y, Guo F. Flavonoids extract from the seeds of Psoralea corylifolia L. (PFE) alleviates atherosclerosis in high-fat diet-induced LDLR -/- mice. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 98:153983. [PMID: 35152088 DOI: 10.1016/j.phymed.2022.153983] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 11/12/2021] [Accepted: 02/06/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND The seeds of Psoralea corylifolia L., a traditional medicine popular used in China and India, have been recommended in the treatment of leucoderma, psoriasis, osteoporosis, and gynecological bleeding. Our previous studies have found that flavonoid extract from the seeds of Psoralea corylifolia L. could activate fat browning and correct the disorder of glucose and lipid metabolism in obese mice. PURPOSE The present study aimed to investigate the anti-atherosclerosis of flavonoids extract from the seeds of Psoralea corylifolia L. METHODS Leukocyte adhesion assay, RT-PCR, Western blot analysis, and immunofluorescent assay were carried out in ox-LDL induced endothelium injury and foam cells formation in vitro. Flavonoids from the seeds of P. corylifolia L. (PFE) was administrated 150 and 300 mg/kg/day in HFD-induced LDLR-/- mice for 12 weeks. RESULTS Flavonoids from the seeds of P. corylifolia L. (PFE) could prevent leukocyte adhesion to the endothelium by inhibiting mRNA and protein expression of these adhesion molecules (VCAM-1, ICAM-1, and E-selectin). PFE could also prevent ox-LDL stimulated inflammation in HUVECs by inhibiting the NF-κB pathway. In addition, PFE significantly ameliorated ox-LDL induced macrophages-oriented foam cells formation through inducing cholesterol efflux via PPARγ-ABCA1/ABCG1. In HFD-induced LDLR-/- mice, PFE reversed the serum profile and circulated inflammation level. Meanwhile, PFE could remarkably alleviate atherosclerotic lesion sizes and intraplaque macrophage infiltration in aortic roots. CONCLUSION Flavonoids from the seeds of P. corylifolia L. could alleviate atherosclerosis by preventing endothelium injury, attenuating vascular inflammation, and alleviating the formation of foam cells.
Collapse
Affiliation(s)
- Jingwen Liu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, PR China
| | - Wen Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, PR China
| | - Yahui Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, PR China
| | - Xiaoye Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, PR China
| | - Yiming Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, PR China.
| | - Fujiang Guo
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, PR China.
| |
Collapse
|
16
|
Zaina S, Lund G. Clonal hematopoiesis of indeterminate potential and the evolutionary lottery in chromosome 2: does that make human atherosclerosis special? Curr Opin Lipidol 2021; 32:389-391. [PMID: 34751167 DOI: 10.1097/mol.0000000000000785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- Silvio Zaina
- Department of Medical Sciences, Division of Health Sciences, Leon Campus, University of Guanajuato, Leon
| | - Gertrud Lund
- Department of Genetic Engineering, CINVESTAV Irapuato Unit, Irapuato, Mexico
| |
Collapse
|
17
|
Munekata PES, Pateiro M, Conte-Junior CA, Domínguez R, Nawaz A, Walayat N, Movilla Fierro E, Lorenzo JM. Marine Alkaloids: Compounds with In Vivo Activity and Chemical Synthesis. Mar Drugs 2021; 19:374. [PMID: 34203532 PMCID: PMC8306672 DOI: 10.3390/md19070374] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/24/2021] [Accepted: 06/24/2021] [Indexed: 12/15/2022] Open
Abstract
Marine alkaloids comprise a class of compounds with several nitrogenated structures that can be explored as potential natural bioactive compounds. The scientific interest in these compounds has been increasing in the last decades, and many studies have been published elucidating their chemical structure and biological effects in vitro. Following this trend, the number of in vivo studies reporting the health-related properties of marine alkaloids has been increasing and providing more information about the effects in complex organisms. Experiments with animals, especially mice and zebrafish, are revealing the potential health benefits against cancer development, cardiovascular diseases, seizures, Alzheimer's disease, mental health disorders, inflammatory diseases, osteoporosis, cystic fibrosis, oxidative stress, human parasites, and microbial infections in vivo. Although major efforts are still necessary to increase the knowledge, especially about the translation value of the information obtained from in vivo experiments to clinical trials, marine alkaloids are promising candidates for further experiments in drug development.
Collapse
Affiliation(s)
- Paulo E. S. Munekata
- Centro Tecnológico de la Carne de Galicia, Parque Tecnológico de Galicia, rúa Galicia No. 4, San Cibrao das Viñas, 32900 Ourense, Spain; (P.E.S.M.); (M.P.); (R.D.)
| | - Mirian Pateiro
- Centro Tecnológico de la Carne de Galicia, Parque Tecnológico de Galicia, rúa Galicia No. 4, San Cibrao das Viñas, 32900 Ourense, Spain; (P.E.S.M.); (M.P.); (R.D.)
| | - Carlos A. Conte-Junior
- Centro de Tecnologia, Programa de Pós-Graduação em Ciência de Alimentos, Instituto de Química, Universidade Federal do Rio de Janeiro, Avenida Athos da Silveira Ramos 149, Cidade Universitária, Rio de Janeiro 21941-909, RJ, Brazil;
| | - Rubén Domínguez
- Centro Tecnológico de la Carne de Galicia, Parque Tecnológico de Galicia, rúa Galicia No. 4, San Cibrao das Viñas, 32900 Ourense, Spain; (P.E.S.M.); (M.P.); (R.D.)
| | - Asad Nawaz
- Jiangsu Key Laboratory of Crop Genetics and Physiology, College of Agriculture, Yangzhou University, Yangzhou 225009, China;
| | - Noman Walayat
- Department of Food Science and Engineering, College of Ocean, Zhejiang University of Technology, Hangzhou 310014, China;
| | | | - José M. Lorenzo
- Centro Tecnológico de la Carne de Galicia, Parque Tecnológico de Galicia, rúa Galicia No. 4, San Cibrao das Viñas, 32900 Ourense, Spain; (P.E.S.M.); (M.P.); (R.D.)
- Área de Tecnología de los Alimentos, Facultad de Ciencias de Ourense, Universidad de Vigo, 32004 Ourense, Spain
| |
Collapse
|
18
|
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
|
19
|
Porwal M, Gautam SK, Khan NA, Maheshwari KK. Evaluation of Toxicity and Antihyperlipidemic Activity of Spondias Mombin l. Leaves Methanolic Extract in Laboratory Rats. Cardiovasc Hematol Disord Drug Targets 2020; 20:289-296. [PMID: 33115396 DOI: 10.2174/1871529x20999201027232556] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 07/13/2020] [Accepted: 09/25/2020] [Indexed: 11/22/2022]
Abstract
AIMS To study the toxicological profile and anti-hyperlipidemic effects of Spondias mombin leaves methanolic extract in experimental rats. BACKGROUND Preventing high levels of lipids or its recurrence is currently one of the key aims of clinical and experimental studies. OBJECTIVE This study was carried out to investigate the toxicological profile and anti-hyperlipidemic effects of methanolic extract of leaves of Spondias mombin. METHODS The acute toxicity study was carried out where the limited dose of 2000 mg/kg body weight was administered to five rats at 48 h intervals. The interpretation was prepared and recorded for 24 h. In the sub-acute toxicity study, rats were treated with 250, 500, and 1000 mg/kg doses of the extract every 24 h for 28 days. The hematological, biochemical, and histopathological tests of treated animals were carried out at the end of the test. The anti-hyperlipidemic activity of plant extract (100, 200 mg/kg) was studied on Triton-X-100 induced hyperlipidemia in rats. Histopathological changes in the liver of rats were examined. RESULTS For acute and subacute treatment, the extract did not reveal any signs of toxicity or mortality, or any significant effects on hematological, biochemical parameters, and histopathology of organs. The extract demonstrated an important anti-hyperlipidemic result by decreasing the serum levels of cholesterol, TGs, LDL, VLDL, and enhancing HDL. CONCLUSION Taking up the evidence of the experimental study, we can conclude that the methanolic extract of Spondias mombin leaves helps in declining hyperlipidemia in rats and it can be safely used for a period of 28 days to treat hyperlipidemia.
Collapse
Affiliation(s)
- Mayur Porwal
- Department of Pharmacology, Iftm University, Moradabad, India
| | | | - Najam A Khan
- Department of Pharmacology, Iftm University, Moradabad, India
| | | |
Collapse
|
20
|
Huang D, Gao W, Zhong X, Ge J. NLRP3 activation in endothelia promotes development of diabetes-associated atherosclerosis. Aging (Albany NY) 2020; 12:18181-18191. [PMID: 32966239 PMCID: PMC7585081 DOI: 10.18632/aging.103666] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 06/22/2020] [Indexed: 01/24/2023]
Abstract
Inflammatory damage to endothelial cells plays a pivotal role in the diabetes-provoked atherosclerosis (AS). PYD domains-containing protein 3 (NLRP3) induces formation of inflammasome activates caspase-1, which subsequently cleaves the precursor form of IL-1β (pro-IL-1β) into the processed, secreted form IL-1β to promote the immune responses in AS. However, it is not known whether NLRP3 activation specifically in endothelial cells causes AS. Here, in an in vitro model for AS, we showed that NLRP3-depleted human aortic endothelial cells (HAECs) became resistant to apoptotic cell death, maintained proliferative potential and reduced reactive oxygen species (ROS) production upon treatment with oxidized low-density lipoprotein (ox-LDL). Next, the role of NLRP3 in endothelial cells in the development of diabetes-associated AS was assessed in endothelial cell-specific NLRP3 mutant, ApoE (-/-) mice (APOEKO/Tie2p-Cre/NLRP3MKO), compared to control ApoE (-/-) mice (APOEKO), supplied with either high-fat diet (HFD), or normal diet (ND). We found that endothelia-specific NLRP3-depletion significantly attenuated AS severity in mice treated with HFD, likely through reduced apoptotic death of endothelial cells and production of ROS. Together, our data suggest that NLRP3 activation in endothelial cells promotes development of diabetes-associated AS.
Collapse
Affiliation(s)
- Dong Huang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
| | - Wei Gao
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
| | - Xin Zhong
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
| |
Collapse
|
21
|
Poznyak AV, Silaeva YY, Orekhov AN, Deykin AV. Animal models of human atherosclerosis: current progress. ACTA ACUST UNITED AC 2020; 53:e9557. [PMID: 32428130 PMCID: PMC7266502 DOI: 10.1590/1414-431x20209557] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 02/20/2020] [Indexed: 12/24/2022]
Abstract
Atherosclerosis retains the leading position among the causes of global morbidity and mortality worldwide, especially in the industrialized countries. Despite the continuing efforts to investigate disease pathogenesis and find the potential points of effective therapeutic intervention, our understanding of atherosclerosis mechanisms remains limited. This is partly due to the multifactorial nature of the disease pathogenesis, when several factors so different as altered lipid metabolism, increased oxidative stress, and chronic inflammation act together leading to the formation and progression of atherosclerotic plaques. Adequate animal models are currently indispensable for studying these processes and searching for novel therapies. Animal models based on rodents, such as mice and rats, and rabbits represent important tools for studying atherosclerosis. Currently, genetically modified animals allow for previously unknown possibilities in modelling the disease and its most relevant aspects. In this review, we describe the recent progress made in creating such models and discuss the most important findings obtained with them to date.
Collapse
Affiliation(s)
- A V Poznyak
- Institute for Atherosclerosis Research, Skolkovo Innovative Center, Moscow, Russia
| | - Y Y Silaeva
- Core Facility Centre, Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia
| | - A N Orekhov
- Institute for Atherosclerosis Research, Skolkovo Innovative Center, Moscow, Russia
| | - A V Deykin
- Core Facility Centre, Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
22
|
Silva G, Ferraresi C, de Almeida RT, Motta ML, Paixão T, Ottone VO, Fonseca IA, Oliveira MX, Rocha-Vieira E, Dias-Peixoto MF, Esteves EA, Coimbra CC, Amorim FT, Magalhães FDC. Insulin resistance is improved in high-fat fed mice by photobiomodulation therapy at 630 nm. JOURNAL OF BIOPHOTONICS 2020; 13:e201960140. [PMID: 31707768 DOI: 10.1002/jbio.201960140] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Revised: 10/24/2019] [Accepted: 11/06/2019] [Indexed: 06/10/2023]
Abstract
Photobiomodulation therapy (PBMT) in the infrared spectrum exerts positive effects on glucose metabolism, but the use of PBMT at the red spectrum has not been assessed. Male Swiss albino mice were divided into low-fat control and high-fat diet (HFD) for 12 weeks and were treated with red (630 nm) PBMT or no treatment (Sham) during weeks 9 to 12. PBMT was delivered at 31.19 J/cm2 , 60 J total dose per day for 20 days. In HFD-fed mice, PBMT improved glucose tolerance, insulin resistance and fasting hyperinsulinemia. PBMT also reduced adiposity and inflammatory infiltrate in adipose tissue. Phosphorylation of Akt in epididymal adipose tissue and rectus femoralis muscle was improved by PBMT. In epididymal fat PBMT reversed the reduced phosphorylation of AS160 and the reduced Glut4 content. In addition, PBMT reversed the alterations caused by HFD in rectus femoralis muscle on proteins involved in mitochondrial dynamics and β-oxidation. In conclusion, PBMT at red spectrum improved insulin resistance and glucose metabolism in HFD-fed mice.
Collapse
Affiliation(s)
- Gabriela Silva
- Programa Multicêntrico de Pós-Graduação em Ciências Fisiológicas, Faculdade de Ciências Básicas e da Saúde, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, Brazil
| | - Cleber Ferraresi
- Post-Graduation Program in Biomedical Engineering, Universidade Brasil, São Paulo, Brazil
| | - Rodrigo T de Almeida
- Programa Multicêntrico de Pós-Graduação em Ciências Fisiológicas, Faculdade de Ciências Básicas e da Saúde, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, Brazil
| | - Mariana L Motta
- Programa Multicêntrico de Pós-Graduação em Ciências Fisiológicas, Faculdade de Ciências Básicas e da Saúde, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, Brazil
| | - Thiago Paixão
- Programa Multicêntrico de Pós-Graduação em Ciências Fisiológicas, Faculdade de Ciências Básicas e da Saúde, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, Brazil
| | - Vinicius O Ottone
- Programa Multicêntrico de Pós-Graduação em Ciências Fisiológicas, Faculdade de Ciências Básicas e da Saúde, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, Brazil
| | - Ivana A Fonseca
- Programa Multicêntrico de Pós-Graduação em Ciências Fisiológicas, Faculdade de Ciências Básicas e da Saúde, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, Brazil
| | - Murilo X Oliveira
- Programa de Pós-Graduação em Reabilitação e Desempenho Funcional, Physiotherapy Department, Faculdade de Ciências Básicas e da Saúde, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, Brazil
| | - Etel Rocha-Vieira
- Programa Multicêntrico de Pós-Graduação em Ciências Fisiológicas, Faculdade de Ciências Básicas e da Saúde, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, Brazil
| | - Marco F Dias-Peixoto
- Programa Multicêntrico de Pós-Graduação em Ciências Fisiológicas, Faculdade de Ciências Básicas e da Saúde, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, Brazil
| | - Elizabethe A Esteves
- Programa Multicêntrico de Pós-Graduação em Ciências Fisiológicas, Faculdade de Ciências Básicas e da Saúde, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, Brazil
| | - Cândido C Coimbra
- Programa Multicêntrico de Pós-Graduação em Ciências Fisiológicas, Faculdade de Ciências Básicas e da Saúde, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, Brazil
- Endocrinology Laboratory, Biological Sciences Institute, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Fabiano T Amorim
- Programa Multicêntrico de Pós-Graduação em Ciências Fisiológicas, Faculdade de Ciências Básicas e da Saúde, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, Brazil
- Department of Heath, Exercise and Sports Science, University of New Mexico, Albuquerque, New Mexico
| | - Flávio de Castro Magalhães
- Programa Multicêntrico de Pós-Graduação em Ciências Fisiológicas, Faculdade de Ciências Básicas e da Saúde, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, Brazil
- Department of Heath, Exercise and Sports Science, University of New Mexico, Albuquerque, New Mexico
| |
Collapse
|
23
|
Sari G, Meester EJ, van der Zee LC, Wouters K, van Lennep JR, Peppelenbosch M, Boonstra A, Van der Heiden K, Mulder MMT, Vanwolleghem T. A mouse model of humanized liver shows a human-like lipid profile, but does not form atherosclerotic plaque after western type diet. Biochem Biophys Res Commun 2020; 524:510-515. [PMID: 32014257 DOI: 10.1016/j.bbrc.2020.01.067] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Accepted: 01/11/2020] [Indexed: 01/12/2023]
Abstract
Mouse models are a crucial and often used tool to provide insight into the underlying mechanisms of human atherosclerosis. However, mice profoundly differ from humans in lipoprotein synthesis and metabolism, key factors in atherosclerotic plaque formation. Mouse models often require genetic and dietary modifications to mimic human pathophysiology, shifting from a high-density lipoprotein to an low-density lipoprotein dominant lipoprotein profile. We examined the suitability of mice with a humanized liver as a model for lipoprotein studies and studies on plaque formation, given the central role of hepatocytes in lipoprotein synthesis and metabolism. Our results show a progressive humanization of the mouse liver and a humanized lipoprotein profile. However, no atherosclerotic plaque formation was observed in the studied time frame, despite presence of functional macrophages and application of a high cholesterol western-type diet. The humanized-liver mouse model therefore might require further modifications to induce atherosclerosis, yet seems a valuable model for in vivo studies on lipoprotein metabolism.
Collapse
Affiliation(s)
- Gulce Sari
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, Rotterdam, the Netherlands.
| | - Eric J Meester
- Department of Biomedical Engineering, Thoraxcenter, Erasmus University Medical Center, Rotterdam, the Netherlands; Department of Radiology & Nuclear Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Leonie C van der Zee
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Kristiaan Wouters
- Department of Internal Medicine, CARIM School for Cardiovascular Diseases, MUMC+, Maastricht, the Netherlands
| | - Jeanine R van Lennep
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Maikel Peppelenbosch
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Andre Boonstra
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Kim Van der Heiden
- Department of Biomedical Engineering, Thoraxcenter, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Monique M T Mulder
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Thomas Vanwolleghem
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, Rotterdam, the Netherlands; Laboratory of Experimental Medicine and Pediatrics, Faculty of Medicine and Health Sciences, University of Antwerp and Department of Gastroenterology and Hepatology, Antwerp University Hospital, Antwerp, Belgium.
| |
Collapse
|
24
|
Zhao H, Li Y, He L, Pu W, Yu W, Li Y, Wu YT, Xu C, Wei Y, Ding Q, Song BL, Huang H, Zhou B. In Vivo AAV-CRISPR/Cas9-Mediated Gene Editing Ameliorates Atherosclerosis in Familial Hypercholesterolemia. Circulation 2019; 141:67-79. [PMID: 31779484 DOI: 10.1161/circulationaha.119.042476] [Citation(s) in RCA: 127] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Mutations in low-density lipoprotein (LDL) receptor (LDLR) are one of the main causes of familial hypercholesterolemia, which induces atherosclerosis and has a high lifetime risk of cardiovascular disease. The clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 system is an effective tool for gene editing to correct gene mutations and thus to ameliorate disease. METHODS The goal of this work was to determine whether in vivo somatic cell gene editing through the CRISPR/Cas9 system delivered by adeno-associated virus (AAV) could treat familial hypercholesterolemia caused by the Ldlr mutant in a mouse model. We generated a nonsense point mutation mouse line, LdlrE208X, based on a relevant familial hypercholesterolemia-related gene mutation. The AAV-CRISPR/Cas9 was designed to correct the point mutation in the Ldlr gene in hepatocytes and was delivered subcutaneously into LdlrE208X mice. RESULTS We found that homogeneous LdlrE208X mice (n=6) exhibited severe atherosclerotic phenotypes after a high-fat diet regimen and that the Ldlr mutation was corrected in a subset of hepatocytes after AAV-CRISPR/Cas9 treatment, with LDLR protein expression partially restored (n=6). Compared with the control groups (n=6 each group), the AAV-CRISPR/Cas9 with targeted single guide RNA group (n=6) had significant reductions in total cholesterol, total triglycerides, and LDL cholesterol in the serum, whereas the aorta had smaller atherosclerotic plaques and a lower degree of macrophage infiltration. CONCLUSIONS Our work shows that in vivo AAV-CRISPR/Cas9-mediated Ldlr gene correction can partially rescue LDLR expression and effectively ameliorate atherosclerosis phenotypes in Ldlr mutants, providing a potential therapeutic approach for the treatment of patients with familial hypercholesterolemia.
Collapse
Affiliation(s)
- Huan Zhao
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences (H.Z., Y.L., L.H., W.P., W.Y., Y.L., B.Z.)
| | - Yan Li
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences (H.Z., Y.L., L.H., W.P., W.Y., Y.L., B.Z.)
| | - Lingjuan He
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences (H.Z., Y.L., L.H., W.P., W.Y., Y.L., B.Z.)
| | - Wenjuan Pu
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences (H.Z., Y.L., L.H., W.P., W.Y., Y.L., B.Z.)
| | - Wei Yu
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences (H.Z., Y.L., L.H., W.P., W.Y., Y.L., B.Z.)
| | - Yi Li
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences (H.Z., Y.L., L.H., W.P., W.Y., Y.L., B.Z.)
| | - Yan-Ting Wu
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, China (Y.-T.W., C.X., H.H.).,Shanghai Key Laboratory of Embryo Original Diseases, China (Y.-T.W., C.X., H.H.)
| | - Chenming Xu
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, China (Y.-T.W., C.X., H.H.).,Shanghai Key Laboratory of Embryo Original Diseases, China (Y.-T.W., C.X., H.H.)
| | - Yuda Wei
- CAS Key Laboratory of Nutrition, Metabolism, and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences (Y.W., Q.D., B.Z.)
| | - Qiurong Ding
- CAS Key Laboratory of Nutrition, Metabolism, and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences (Y.W., Q.D., B.Z.)
| | - Bao-Liang Song
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Institute for Advanced Studies, Wuhan University, China (B.-L.S.)
| | - Hefeng Huang
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, China (Y.-T.W., C.X., H.H.).,Shanghai Key Laboratory of Embryo Original Diseases, China (Y.-T.W., C.X., H.H.)
| | - Bin Zhou
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences (H.Z., Y.L., L.H., W.P., W.Y., Y.L., B.Z.).,CAS Key Laboratory of Nutrition, Metabolism, and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences (Y.W., Q.D., B.Z.).,School of Life Science and Technology, ShanghaiTech University, China (B.Z.).,Key Laboratory of Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, China (B.Z.).,Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, China (B.Z.).,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing (B.Z.)
| |
Collapse
|
25
|
Barreiro E, Wang X, Tang J. COPD: preclinical models and emerging therapeutic targets. Expert Opin Ther Targets 2019; 23:829-838. [DOI: 10.1080/14728222.2019.1667976] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Esther Barreiro
- Respiratory Medicine Department, Muscle Wasting and Cachexia in Chronic Respiratory Diseases and Lung Cancer Research Group, Institute of Medical Research of Hospital del Mar (IMIM)-Hospital del Mar, Parc de Salut Mar, Barcelona Biomedical Research Park (PRBB), Barcelona, Spain
- Department of Health Sciences (CEXS), Universitat Pompeu Fabra, Barcelona, Spain
- Centro de Investigación en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III (ISCIII), Barcelona, Spain
| | - Xuejie Wang
- Respiratory Medicine Department, Muscle Wasting and Cachexia in Chronic Respiratory Diseases and Lung Cancer Research Group, Institute of Medical Research of Hospital del Mar (IMIM)-Hospital del Mar, Parc de Salut Mar, Barcelona Biomedical Research Park (PRBB), Barcelona, Spain
- Department of Medicine, Autonomous University of Barcelona, Barcelona, Spain
| | - Jun Tang
- Respiratory Medicine Department, Muscle Wasting and Cachexia in Chronic Respiratory Diseases and Lung Cancer Research Group, Institute of Medical Research of Hospital del Mar (IMIM)-Hospital del Mar, Parc de Salut Mar, Barcelona Biomedical Research Park (PRBB), Barcelona, Spain
- Centro de Investigación en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III (ISCIII), Barcelona, Spain
- Department of Medicine, Autonomous University of Barcelona, Barcelona, Spain
| |
Collapse
|
26
|
Silvério de Barros R, Dias GS, Paula do Rosario A, Paladino FV, Lopes GH, Campos AH. Gremlin-1 potentiates the dedifferentiation of VSMC in early stages of atherosclerosis. Differentiation 2019; 109:28-33. [PMID: 31494396 DOI: 10.1016/j.diff.2019.08.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 08/12/2019] [Accepted: 08/27/2019] [Indexed: 02/08/2023]
Abstract
Vascular smooth muscle cells (VSMC) are highly specialized, and exhibit a contractile phenotype when mature and fully differentiated, being responsible for vessel homeostasis and blood pressure control. In response to pro-atherogenic stimuli VSMC alter their state of differentiation, increase proliferation and migration, resulting in SMC phenotypes ranging from contractile to synthetic. This variability is observed in cell morphology and expression level of marker genes for differentiation status. There is growing evidence that bone morphogenetic protein (BMP) signaling is involved in vascular diseases, including atherosclerosis. Here, we evaluated in vitro the role of specific agonists/antagonists belonging to the BMP pathway on dedifferentiation of VSMC harvested during early stages of atherosclerosis. RESULTS: Comparing primary VSMC isolated from aortas of susceptible ApoE-/- animals fed 8 weeks of western diet with their littermate controls fed usual diet, we observed that recombinant BMP4 was able to reduce SM22-alpha and alpha actin gene expression indicating dedifferentiation was under way. Unexpectedly, treatment with recombinant Gremlin-1, a known BMP antagonist, also reduced 4-6.5 folds gene expression of SM22-alpha, alpha-actin and, calponin, exclusively in VSMC from ApoE-/- animals, independently on the diet consumed. CONCLUSION: Our data show that BMP4 is capable of modulating of SM22-alpha and alpha actin gene expression, indicative of cell dedifferentiation in VSMC. Additionally, we report for first time that Gremlin-1 acts independently of the BMP pathway and selectively on VSMC from susceptible animals, reducing the expression of all genes evaluated.
Collapse
Affiliation(s)
- Renata Silvério de Barros
- Hospital Israelita Albert Einstein, São Paulo, Brazil; Centro de Pesquisa Experimental, Av Albert Einstein, 627. Morumbi, 2S/Bloco A, São Paulo, SP, CEP 05651-901, Brazil
| | - Grazielle Suhett Dias
- Hospital Israelita Albert Einstein, São Paulo, Brazil; Centro de Pesquisa Experimental, Av Albert Einstein, 627. Morumbi, 2S/Bloco A, São Paulo, SP, CEP 05651-901, Brazil
| | - Ana Paula do Rosario
- Hospital Israelita Albert Einstein, São Paulo, Brazil; Centro de Pesquisa Experimental, Av Albert Einstein, 627. Morumbi, 2S/Bloco A, São Paulo, SP, CEP 05651-901, Brazil
| | - Fernanda Vieira Paladino
- Hospital Israelita Albert Einstein, São Paulo, Brazil; Centro de Pesquisa Experimental, Av Albert Einstein, 627. Morumbi, 2S/Bloco A, São Paulo, SP, CEP 05651-901, Brazil
| | | | - Alexandre Holthausen Campos
- Hospital Israelita Albert Einstein, São Paulo, Brazil; Centro de Pesquisa Experimental, Av Albert Einstein, 627. Morumbi, 2S/Bloco A, São Paulo, SP, CEP 05651-901, Brazil.
| |
Collapse
|
27
|
Zhou PL, Li M, Han XW, Bi YH, Zhang WG, Wu ZY, Wu G. Perilipin 5 deficiency promotes atherosclerosis progression through accelerating inflammation, apoptosis, and oxidative stress. J Cell Biochem 2019; 120:19107-19123. [PMID: 31297870 DOI: 10.1002/jcb.29238] [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: 07/30/2017] [Accepted: 11/09/2017] [Indexed: 01/11/2023]
Abstract
Excessive plasma triglyceride (TG) and cholesterol levels promote the progression of several prevalent cardiovascular risk factors, including atherosclerosis, which is a leading death cause. Perilipin 5 (Plin5), an important perilipin protein, is abundant in tissues with very active lipid catabolism and is involved in the regulation of oxidative stress. Although inflammation and oxidative stress play a critical role in atherosclerosis development, the underlying mechanisms are complex and not completely understood. In the present study, we demonstrated the role of Plin5 in high-fat-diet-induced atherosclerosis in apolipoprotein E null (ApoE-/- ) mice. Our results suggested that Plin5 expressions increased in the artery tissues of ApoE-/- mice. ApoE/Plin5 double knockout (ApoE-/- Plin5-/- ) exacerbated severer atherogenesis, accompanied with significantly disturbed plasma metabolic profiles, such as elevated TG, total cholesterol, and low-density lipoprotein cholesterol levels and reduced high-density lipoprotein cholesterol contents. ApoE-/- Plin5-/- exhibited a higher number of inflammatory monocytes and neutrophils, as well as overexpression of cytokines and chemokines linked with an inflammatory response. Consistently, the IκBα/nuclear factor kappa B pathway was strongly activated in ApoE-/- Plin5-/- . Notably, apoptosis was dramatically induced by ApoE-/- Plin5-/- , as evidenced by increased cleavage of Caspase-3 and Poly (ADP-ribose) polymerase-2. In addition, ApoE-/- Plin5-/- contributed to oxidative stress generation in the aortic tissues, which was linked with the activation of phosphatidylinositol 3-kinase/protein kinase B and mitogen-activated protein kinases pathways. In vitro, oxidized low-density lipoprotein (ox-LDL) increased Plin5 expression in RAW264.7 cells. Its knockdown enhanced inflammation, apoptosis, oxidative stress, and lipid accumulation, while promotion of Plin5 markedly reduced all the effects induced by ox-LDL in cells. These studies strongly supported that Plin5 could be a new regulator against atherosclerosis, providing new insights on therapeutic solutions.
Collapse
Affiliation(s)
- Peng-Li Zhou
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Min Li
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xin-Wei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yong-Hua Bi
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Wen-Guang Zhang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zheng-Yang Wu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Gang Wu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
28
|
Schick D, Babendreyer A, Wozniak J, Awan T, Noels H, Liehn E, Bartsch JW, Vlacil AK, Grote K, Zayat R, Goetzenich A, Ludwig A, Dreymueller D. Elevated expression of the metalloproteinase ADAM8 associates with vascular diseases in mice and humans. Atherosclerosis 2019; 286:163-171. [PMID: 30910225 DOI: 10.1016/j.atherosclerosis.2019.03.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 02/17/2019] [Accepted: 03/12/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND AIMS Members of the family of a disintegrin and metalloproteinases (ADAMs) and their substrates have been previously shown to modulate the inflammatory response in cardiac diseases, but studies investigating the relevance of ADAM8 are still rare. Our aim is to provide evidence for the inflammatory dysregulation of ADAM8 in vascular diseases and its association with disease severity. METHODS Western-type diet fed Apoe-/- and Ldlr-/- mice and artery ligation served as murine model for atherosclerosis and myocardial infarction, respectively. Human bypass grafts were used to study the association with coronary artery disease (CAD), with the simplified acute physiology score II (SAPS II) as a measure of postoperative organ dysfunction. Human primary vascular and blood cells were analyzed under basal and inflammatory conditions. mRNA levels were determined by RT-qPCR, ADAM8 protein levels by ELISA, immunohistochemistry or flow cytometry. RESULTS ADAM8/ADAM8 expression is associated with atherosclerosis and CAD such as myocardial infarction in both mice and humans, especially in endothelial cells and leukocytes. We observed a strong in vivo and in vitro correlation of ADAM8 with the vascular disease markers VCAM-1, ICAM-1, TNF, IL-6, and CCL-2. Serum analysis revealed a significant elevation of soluble ADAM8 serum levels correlating with soluble CXCL16 levels and SAPS II. CONCLUSIONS We demonstrate a general association of ADAM8 with cardiovascular diseases in mice and humans predominantly acting in endothelial cells and leukocytes. The correlation with postoperative organ dysfunctions in CAD patients highlights the value of further studies investigating the specific function of ADAM8 in cardiovascular diseases.
Collapse
Affiliation(s)
- Daniel Schick
- Institute of Pharmacology and Toxicology, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Aaron Babendreyer
- Institute of Pharmacology and Toxicology, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Justyna Wozniak
- Institute of Pharmacology and Toxicology, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Tanzeela Awan
- Institute of Pharmacology and Toxicology, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Heidi Noels
- Institute of Molecular Cardiovascular Research, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074, Aachen, Germany
| | - Elisa Liehn
- Institute of Molecular Cardiovascular Research, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074, Aachen, Germany; National Heart Center Singapore, Singapore, Human Genetic Laboratory, University of Medicine Craiova, Romania
| | - Jörg-W Bartsch
- Department of Neurosurgery, Philipps University Marburg, University Hospital Marburg, Baldingerstrasse, 35033, Marburg, Germany
| | - Ann-Kathrin Vlacil
- Clinic for Internal Medicine, Cardiology, Philipps University Marburg, University Hospital Marburg, Marburg, Germany
| | - Karsten Grote
- Clinic for Internal Medicine, Cardiology, Philipps University Marburg, University Hospital Marburg, Marburg, Germany
| | - Rashad Zayat
- Department of Thoracic and Cardiovascular Surgery, RWTH Aachen University Hospital, Pauwelsstr. 30, 52066, Aachen, Germany
| | - Andreas Goetzenich
- Department of Thoracic and Cardiovascular Surgery, RWTH Aachen University Hospital, Pauwelsstr. 30, 52066, Aachen, Germany
| | - Andreas Ludwig
- Institute of Pharmacology and Toxicology, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Daniela Dreymueller
- Institute of Experimental and Clinical Pharmacology and Toxicology, PZMS, ZHMB, Saarland University, UKS Bldg. 46, 66421, Homburg, Germany.
| |
Collapse
|
29
|
Yang L, Gao C. MiR-590 Inhibits Endothelial Cell Apoptosis by Inactivating the TLR4/NF-κB Pathway in Atherosclerosis. Yonsei Med J 2019; 60:298-307. [PMID: 30799593 PMCID: PMC6391523 DOI: 10.3349/ymj.2019.60.3.298] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 10/30/2018] [Accepted: 11/28/2018] [Indexed: 01/01/2023] Open
Abstract
PURPOSE Previous study has well documented the anti-apoptotic effects of miR-590 on oxidized low-density lipoprotein (ox-LDL)-treated endothelial cells (ECs). However, the mechanism underlying the anti-apoptotic effects of miR-590 in ox-LDL-treated ECs remains to be further addressed. MATERIALS AND METHODS ApoE-/- mice fed with a high-fat diet (HFD) and human aortic endothelial cells (HAECs) treated with ox-LDL were used as in vivo and in vitro models of atherosclerosis. The expressions of miR-590 and toll-like receptor 4 (TLR4) were detected by quantitative real-time PCR and Western blot, respectively. Atherosclerotic lesion analysis was performed using Evans blue and hematoxylin-eosin staining. Cell proliferation was assessed by MTT assay. Apoptosis was examined using flow cytometry analysis and Western blot analysis of Cleaved poly (ADP-ribose) polymerase (PARP) and Cleaved Caspase-3 levels. The effect of miR-590 on TLR4/nuclear factor kappa B (NF-κB) pathway was evaluated by Western blot. Binding between miR-590 and TLR4 was confirmed by luciferase reporter assay and Western blot. RESULTS miR-590 was downregulated in the aorta tissues from HFD-fed apoE-/- mice and ox-LDL-treated HAECs. miR-590 overexpression inhibited atherosclerotic lesion in HFD-induced apoE-/- mice and promoted proliferation and inhibited apoptosis of ox-LDL-treated HAECs. Additionally, TLR4 was identified as a direct target of miR-590 in ox-LDL-treated HAECs. Moreover, anti-miR-590 reversed TLR4 knockdown-mediated promotion of cell proliferation and suppression of apoptosis in ox-LDL-treated HAECs. miR-590 overexpression suppressed the TLR4/NF-κB pathway, and inhibition of the TLR4/NF-κB pathway promoted cell proliferation and impeded apoptosis in ox-LDL-treated HAECs. CONCLUSION miR-590 promoted proliferation and blocked ox-LDL-induced apoptosis in HAECs through inhibition of the TLR4/NF-κB pathway.
Collapse
Affiliation(s)
- Lei Yang
- Department of Emergency, Zhengzhou University People's Hospital, Zhengzhou, China
| | - Chuanyu Gao
- Department of Coronary Heart Disease, Zhengzhou University People's Hospital, Fuwai Central China Cardiovascular Hospital, Zhengzhou, China.
| |
Collapse
|
30
|
Hall S, Agrawal DK. Delivery of viral vectors for gene therapy in intimal hyperplasia and restenosis in atherosclerotic swine. Drug Deliv Transl Res 2018; 8:918-927. [PMID: 28707263 DOI: 10.1007/s13346-017-0409-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cardiovascular diseases including atherosclerosis are a major financial and health burden globally. Inflammation associated with atherosclerosis results in the development of plaques that can rupture causing thrombosis, stroke, or death. The most widely used treatment for the removal of atherosclerotic plaques is percutaneous transluminal coronary angioplasty (PTCA) with or without stenting. Although this is a safer and minimally invasive method, restenosis and intimal hyperplasia after interventional procedure remains a major hurdle and more refined approaches are needed. Studies in large animal models such as pigs have facilitated a greater understanding of the underlying mechanisms of the disease and provided novel targets for therapeutic intervention. In pre-clinical studies, viral vector gene therapy has emerged as a promising option for the reduction and/or prevention of restenosis and intimal hyperplasia. Although studies in animal models have generated promising results, clinical trials have yet to prove the clinical efficacy of gene therapy in coronary artery diseases. In this review, we examined and critically reviewed the most recent advances in viral vector gene therapy obtained from studies using porcine model of atherosclerosis.
Collapse
Affiliation(s)
- Sannette Hall
- Department of Clinical and Translational Science, School of Medicine, Creighton University, Omaha, NE, 68178, USA
| | - Devendra K Agrawal
- Department of Clinical and Translational Science, School of Medicine, Creighton University, Omaha, NE, 68178, USA. .,Department of Clinical and Translational Science, The Peekie Nash Carpenter Endowed Chair in Medicine, School of Medicine, Creighton University, CRISS II Room 510, 2500 California Plaza, Omaha, NE, 68178, USA.
| |
Collapse
|
31
|
Yang S, Liu L, Meng L, Hu X. Capsaicin is beneficial to hyperlipidemia, oxidative stress, endothelial dysfunction, and atherosclerosis in Guinea pigs fed on a high-fat diet. Chem Biol Interact 2018; 297:1-7. [PMID: 30342015 DOI: 10.1016/j.cbi.2018.10.006] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 09/27/2018] [Accepted: 10/16/2018] [Indexed: 12/31/2022]
Abstract
Capsaicin has anti-inflammatory and antioxidant effects, as well as some benefits on the cardiovascular system. The exact effects of capsaicin on atherosclerosis are poorly understood. To investigate the effects of capsaicin on hyperlipidemia and atherosclerosis in guinea pigs fed on a high-fat diet, as well as its potential mechanisms. Guinea pigs (n = 48) were randomly divided into six groups (n = 8/group): normal diet (control); high fat diet (model); model + low-dose capsaicin (2.5 mg/kg); model + moderate-dose capsaicin (5 mg/kg); model + high-dose capsaicin (10 mg/kg), and model + simvastatin (1.5 mg/kg) (positive control). After 14 weeks, serum lipids, apolipoprotein B100, malondialdehyde (MDA), superoxide dismutase (SOD), nitric oxide (NO), and endothelin-1 were measured. Aortic atherosclerotic lesions were histologically examined. eNOS and iNOS were assessed by immunohistochemistry. The model group developed severe dyslipidemia and associated histologic changes and endothelial dysfunction. All doses of capsaicin decreased total cholesterol, triglycerides, low-density lipoprotein cholesterol, and apolipoprotein B-100, and increased high-density lipoprotein cholesterol (all P < 0.05). Capsaicin alleviated the plaque area (-17.9-70.5%), plaque area to intima ratio (-18.0-73.6%), and intima thickness (-20.5-83.6%) (all P < 0.05). Capsaicin decreased MDA (-45.5-76.1%), ET-1 (-19.6-51.6%), and average gray value (AGV) of eNOS (-10.9-48.8%), and increased SOD activity (+31.7-76.1%), NO (+11.2-36.8%), and AGV of iNOS (+6.8-+93.0%) (all P < 0.05). Similar changes were observed with simvastatin. Capsaicin is beneficial to hyperlipidemia and atherosclerosis in guinea pigs fed on a high-fat diet. Reduced oxidative stress and endothelial dysfunction were involved in these benefits. This could represent a novel approach to prevent cardiovascular diseases.
Collapse
Affiliation(s)
- Siyuan Yang
- Division of Cardiac Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China.
| | - Lin Liu
- Department of Respiratory & Critical Care Medicine, Guizhou Provincial People's Hospital, Guiyang, Guizhou, 550002, China
| | - Like Meng
- School of Principle Medicine, Guizhou Medical University, Guiyang, Guizhou, 550025, China
| | - Xuanyi Hu
- Division of Cardiac Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China
| |
Collapse
|
32
|
Tarasco E, Pellegrini G, Whiting L, Lutz TA. Phenotypical heterogeneity in responder and nonresponder male ApoE*3Leiden.CETP mice. Am J Physiol Gastrointest Liver Physiol 2018; 315:G602-G617. [PMID: 29975550 DOI: 10.1152/ajpgi.00081.2018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The metabolic syndrome (MetS) is a major health issue worldwide and is associated with obesity, insulin resistance, and hypercholesterolemia. Several animal models were used to describe the MetS; however, many of them do not mimic well the MetS pathophysiology in humans. The ApoE*3Leiden.CETP mouse model overcomes part of this limitation, since they have a humanised lipoprotein metabolism and a heterogeneous response to MetS, similar to humans. The reported heterogeneity among them and their common classification refer to responder (R) and nonresponder (NR) mice; R mice show increased body weight, cholesterol, and triglycerides levels, whereas NR mice do not show this expected phenotype when fed a Western type diet. To define better the differences between R and NR mice, we focused on feeding behavior, body weight gain, glucose tolerance, and lipid parameters, and on an extensive pathological examination along with liver histology analysis. Our data confirmed that R mice resemble the pathological features of the human MetS: obesity, dysplipidemia, and glucose intolerance. NR mice do not develop the full dysmetabolic phenotype because of a severe inflammatory hepatic condition, which may heavily affect liver function. We conclude that R and NR mice are metabolically different and that NR mice have indications of severely impaired liver function. Hence, it is critical to identify and separate the respective mice to decrease data heterogeneity. Clinical chemistry and histological analysis should be used to confirm retrospectively the animals' classification. Moreover, we point out that NR mice may not be an appropriate control for studies involving ApoE*3Leiden.CETP R mice. NEW & NOTEWORTHY When compared with some other animal models, ApoE*3Leiden.CETP mice are better models to describe the metabolic syndrome. However, there is phenotypic heterogeneity between "responder" and "nonresponder" mice, the latter showing some evidence of hepatic pathology. A full phenotypic characterization and eventually postmortem analysis of the liver are warranted.
Collapse
Affiliation(s)
- Erika Tarasco
- Institute of Veterinary Physiology, Vetsuisse Faculty University of Zurich , Zurich , Switzerland.,Zurich Center for Integrative Human Physiology, University of Zurich , Zurich , Switzerland
| | - Giovanni Pellegrini
- Laboratory for Animal Model Pathology, Institute of Veterinary Pathology, Vetsuisse Faculty University of Zurich , Zurich , Switzerland
| | - Lynda Whiting
- Institute of Drug and Discovery Biology, University of Monash , Victoria , Australia
| | - Thomas A Lutz
- Institute of Veterinary Physiology, Vetsuisse Faculty University of Zurich , Zurich , Switzerland.,Zurich Center for Integrative Human Physiology, University of Zurich , Zurich , Switzerland
| |
Collapse
|
33
|
Wu L, Yao Q, Lin P, Li Y, Li H. Comparative transcriptomics reveals specific responding genes associated with atherosclerosis in rabbit and mouse models. PLoS One 2018; 13:e0201618. [PMID: 30067832 PMCID: PMC6070260 DOI: 10.1371/journal.pone.0201618] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 07/18/2018] [Indexed: 11/18/2022] Open
Abstract
Mouse and rabbit are frequently employed species for atherosclerosis research. With respect to modeling human atherosclerosis, it has been observed that variations in phenotype under commonly used atherogenic conditions are partial or no congruence between two species. However, genome-wide molecular variations are still lacking. To understand the differences between rabbit and mouse in developing atherosclerosis, here from aspect of orthologs, we compared the genome-wide expression profiles of two species under the same atherosclerosis driven factors: high-fat diet or LDLR deficiency. Our results illuminated that: 1) LDLR-deficiency induced different gene expression changes in rabbit and mouse. WHHL rabbit had more significantly differential expressed genes and the most of genes were down-regulated. 2) Some genes and functions were commonly dysregulated in high-fat fed rabbit and mouse models, such as lipid metabolism and inflammation process. However, high-fat intake in rabbit produced more differentially expressed genes and more serious functional effects. 3) Specific differential expression genes were revealed for rabbit and mouse related with high-fat intake. In the aspect of lipoprotein metabolism, APOA4 and APOB was the major responding gene in rabbit and mice, respectively. The expression change of APOA4 and APOB in human atherosclerosis was more similar to rabbit, and therefore rabbit might be a better animal model for investigating human lipoprotein metabolism related diseases. In conclusion, our comparative transcriptome analysis revealed species-specific expression regulation that could partially explain the different phenotypes between rabbit and mouse, which was helpful for model selection to study atherosclerosis.
Collapse
Affiliation(s)
- Leilei Wu
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Qianlan Yao
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Ping Lin
- CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yixue Li
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
- CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
- Collaborative Innovation Center of Genetics and Development, Fudan University, Shanghai, China
| | - Hong Li
- CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
34
|
The potential atheroprotective role of plant MIR156a as a repressor of monocyte recruitment on inflamed human endothelial cells. J Nutr Biochem 2018; 57:197-205. [DOI: 10.1016/j.jnutbio.2018.03.026] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 03/21/2018] [Accepted: 03/27/2018] [Indexed: 02/04/2023]
|
35
|
Ziegler T, Bähr A, Howe A, Klett K, Husada W, Weber C, Laugwitz KL, Kupatt C, Hinkel R. Tβ4 Increases Neovascularization and Cardiac Function in Chronic Myocardial Ischemia of Normo- and Hypercholesterolemic Pigs. Mol Ther 2018; 26:1706-1714. [PMID: 29929787 DOI: 10.1016/j.ymthe.2018.06.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 06/05/2018] [Accepted: 06/05/2018] [Indexed: 12/11/2022] Open
Abstract
Translations of new therapeutic options for cardiovascular disease from animal studies into a clinical setting have been hampered, in part by an improper reflection of a relevant patient population in animal models. In this study, we investigated the impact of thymosin β4 (Tβ4), which promotes collateralization and capillarization, during hypercholesterolemia, a known risk factor of coronary artery disease. Initial in vitro results highlighted an improved endothelial cell function upon Tβ4 treatment under control conditions and during hypercholesterolemic stress (scratch area [pixels]: oxidized low-density lipoprotein [oxLDL], 191,924 ± 7,717; and oxLDL + Tβ4, 105,621 ± 11,245). To mimic the common risk factor of hypercholesterolemia in vivo, pigs on regular (NC) or high-fat (HC) diet underwent chronic myocardial ischemia followed by recombinant adeno-associated virus (rAAV)-mediated transduction of Tβ4 or LacZ as a control. We show that Tβ4 overexpression improves capillarization and collateralization (collaterals: NC + rAAV.LacZ, 2.1 ± 0.5; NC + rAAV.Tβ4, 6.7 ± 0.5; HC + rAAV.LacZ, 3.0 ± 0.3; and HC + rAAV.Tβ4, 6.0 ± 0.4), ultimately leading to an improved myocardial function in both diet groups (ejection fraction [EF] at day 56 [%]: NC + rAAV.LacZ, 26 ± 1.1; NC + rAAV.Tβ4, 45 ± 1.5; HC + rAAV.LacZ, 26 ± 2.5; and HC + rAAV.Tβ4, 41 ± 2.6). These results demonstrate the potency of Tβ4 in a patient-relevant large animal model of chronic myocardial ischemia.
Collapse
Affiliation(s)
- Tilman Ziegler
- Klinik und Poliklinik Innere Medizin I, Klinikum rechts der Isar - Technical University of Munich, 81675 Munich, Germany; DZHK (German Centre for Cardiovascular Research) - partner site Munich Heart Alliance, 81675 Munich, Germany
| | - Andrea Bähr
- Klinik und Poliklinik Innere Medizin I, Klinikum rechts der Isar - Technical University of Munich, 81675 Munich, Germany; DZHK (German Centre for Cardiovascular Research) - partner site Munich Heart Alliance, 81675 Munich, Germany
| | - Andrea Howe
- Klinik und Poliklinik Innere Medizin I, Klinikum rechts der Isar - Technical University of Munich, 81675 Munich, Germany
| | - Katharina Klett
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität (LMU), 80336 Munich, Germany; DZHK (German Centre for Cardiovascular Research) - partner site Munich Heart Alliance, 81675 Munich, Germany
| | - Wira Husada
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität (LMU), 80336 Munich, Germany
| | - Christian Weber
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität (LMU), 80336 Munich, Germany; DZHK (German Centre for Cardiovascular Research) - partner site Munich Heart Alliance, 81675 Munich, Germany
| | - Karl-Ludwig Laugwitz
- Klinik und Poliklinik Innere Medizin I, Klinikum rechts der Isar - Technical University of Munich, 81675 Munich, Germany; DZHK (German Centre for Cardiovascular Research) - partner site Munich Heart Alliance, 81675 Munich, Germany
| | - Christian Kupatt
- Klinik und Poliklinik Innere Medizin I, Klinikum rechts der Isar - Technical University of Munich, 81675 Munich, Germany; DZHK (German Centre for Cardiovascular Research) - partner site Munich Heart Alliance, 81675 Munich, Germany
| | - Rabea Hinkel
- Klinik und Poliklinik Innere Medizin I, Klinikum rechts der Isar - Technical University of Munich, 81675 Munich, Germany; Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität (LMU), 80336 Munich, Germany; DZHK (German Centre for Cardiovascular Research) - partner site Munich Heart Alliance, 81675 Munich, Germany.
| |
Collapse
|
36
|
Huwiler A, Zangemeister-Wittke U. The sphingosine 1-phosphate receptor modulator fingolimod as a therapeutic agent: Recent findings and new perspectives. Pharmacol Ther 2018; 185:34-49. [DOI: 10.1016/j.pharmthera.2017.11.001] [Citation(s) in RCA: 165] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
37
|
Boshtam M, Khanahmad Shahreza H, Feizollahzadeh S, Rahimmanesh I, Asgary S. Expression and purification of biologically active recombinant rabbit monocyte chemoattractant protein1 in Escherichia coli. FEMS Microbiol Lett 2018; 365:4955552. [PMID: 29596634 DOI: 10.1093/femsle/fny070] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 03/26/2018] [Indexed: 12/22/2022] Open
Abstract
Monocyte chemoattractant protein 1 (MCP1) with recruiting monocytes is an important factor at the beginning of inflammatory disorders such as atherosclerosis which seems its blocking preclude this process and help improvement of related diseases. To perform clinical research in this field, MCP1 protein is required but firstly, animal studies should be done. As the rabbit is a suitable model for many inflammatory disorders, and Escherichia coli BL21(DE3) (BL21) cell is a high-efficiency host for protein expression, we decided to produce recombinant rabbit MCP1 (rRMCP1) in BL21/pET28a system. After codon usage, a construct containing RMCP1 sequence was synthesized, cloned into the pET28a plasmid, and overexpressed in BL21 cells. Followed that, with changing expression condition such as cell concentration before the induction, time period, temperature, shaking rate and inducer concentration (IPTG), rRMCP1 expression was optimized, and purified by Ni-NTA. The biological activity of the expressed protein was verified using monocyte migration assay. Using this expression system, nearly 28 mg/mL rRMCP1 was produced at 26°C/180 rpm for 24 h in LB broth medium with 1 mM IPTG. Therefore, we were succeeded to express the intermediate level of rRMCP1 with this method. This amount of protein is sufficient for biological researches in the laboratory.
Collapse
Affiliation(s)
- Maryam Boshtam
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan 8174643446, Iran
| | - Hossein Khanahmad Shahreza
- Genetic and Molecular Biology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan 8174643446, Iran
| | - Sadegh Feizollahzadeh
- Faculty of Paramedical, Urmia University of Medical Sciences, Urmia 5756115198, Iran
| | - Ilnaz Rahimmanesh
- Genetic and Molecular Biology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan 8174643446, Iran
| | - Sedigheh Asgary
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan 8174643446, Iran
| |
Collapse
|
38
|
Li ZM, Xu SW, Liu PQ. Salvia miltiorrhizaBurge (Danshen): a golden herbal medicine in cardiovascular therapeutics. Acta Pharmacol Sin 2018; 39:802-824. [PMID: 29698387 PMCID: PMC5943903 DOI: 10.1038/aps.2017.193] [Citation(s) in RCA: 340] [Impact Index Per Article: 48.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Accepted: 12/31/2017] [Indexed: 02/07/2023]
Abstract
Salvia miltiorrhiza Burge (Danshen) is an eminent medicinal herb that possesses broad cardiovascular and cerebrovascular protective actions and has been used in Asian countries for many centuries. Accumulating evidence suggests that Danshen and its components prevent vascular diseases, in particular, atherosclerosis and cardiac diseases, including myocardial infarction, myocardial ischemia/reperfusion injury, arrhythmia, cardiac hypertrophy and cardiac fibrosis. The published literature indicates that lipophilic constituents (tanshinone I, tanshinone IIa, tanshinone IIb, cryptotanshinone, dihydrotanshinone, etc) as well as hydrophilic constituents (danshensu, salvianolic acid A and B, protocatechuic aldehyde, etc) contribute to the cardiovascular protective actions of Danshen, suggesting a potential synergism among these constituents. Herein, we provide a systematic up-to-date review on the cardiovascular actions and therapeutic potential of major pharmacologically active constituents of Danshen. These bioactive compounds will serve as excellent drug candidates in small-molecule cardiovascular drug discovery. This article also provides a scientific rationale for understanding the traditional use of Danshen in cardiovascular therapeutics.
Collapse
Affiliation(s)
- Zhuo-ming Li
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences; National and Local United Engineering Lab of Druggability and New Drugs Evaluation, Sun Yat-Sen University, Guangzhou 510006, China
| | - Suo-wen Xu
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York, 14642, USA
| | - Pei-qing Liu
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences; National and Local United Engineering Lab of Druggability and New Drugs Evaluation, Sun Yat-Sen University, Guangzhou 510006, China
| |
Collapse
|
39
|
Colleter R, Dedouit F, Duchesne S, Gérard P, Dercle L, Poilpré P, Gendrot V, Rousseau H, Crubézy É, Telmon N, Mokrane FZ. Study of a seventeenth-century French artificial mummy: autopsical, native, and contrast-injected CT investigations. Int J Legal Med 2018; 132:1405-1413. [PMID: 29594348 DOI: 10.1007/s00414-018-1830-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 03/21/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND A lead coffin was fortuitously discovered in a church called "Eglise des Toussaints" in Rennes (French Brittany). A collaborative taskforce investigated this extraordinary discovery. A multi-disciplinary team of experts from the National Institute for Preventive Archeological Research (INRAP) and Rangueil University Hospital of Toulouse was created, including anthropologists, archeologists, forensic pathologists, radiologists, and pathologists. The inscription on the lead coffin specified that the body belonged to "Messer Louys de Bruslon, Lord of Plessis," a nobleman who died on November 1, 1661. Multiple holes were visible in the lead coffin, and deterioration threatened the mummy. We opened the lead coffin and discovered an excellently preserved mummy, except for mostly skeletonized upper and lower limbs. The mummy was conserved in several layers of shrouds. Vegetal embalming material covered the head and filled the face, the thorax, and the abdomen. The embalmers had removed all thoracic and abdominal organs and conserved some pelvic organs (e.g., the bladder). METHODS Multi-slice computed tomography (MSCT) scanner evaluated the mummy, at each step of our analysis. The excellent preservation of abdominal vascular axes led us to perform a CT angiography using Angiofil®, an oily contrast agent developed for postmortem imaging, before an autopsy. RESULTS Sub-diaphragmatic arteries, including the abdominal aorta and iliac arteries, were excellently preserved. The vascular contrast agent filled all arteries. The native CT, CT angiography, and autopsy did not detect any vascular lesion. CONCLUSION Our study, based on rare archeological material, allowed a complete examination of an excellently preserved seventeenth-century mummy, using MSCT, angiography, and an autopsy. We did not detect any arterial lesion and proposed a comprehensive description of the embalmment process.
Collapse
Affiliation(s)
- Rozenn Colleter
- INRAP National Institute of Preventive Archaeological Research (INRAP, France), 37 rue du Bignon, CS 67737, 35577, Cesson-Sévigné, France
- AMIS Laboratory: University of Toulouse, French National Center for Scientific Research, UMR 5288, 37 allées Jules Guesde, 31073, Toulouse, France
| | - Fabrice Dedouit
- AMIS Laboratory: University of Toulouse, French National Center for Scientific Research, UMR 5288, 37 allées Jules Guesde, 31073, Toulouse, France
- Unit of Forensic and Anthropological Imaging, Centre Universitaire Romand de Médecine Légale (CURML), Chemin de la Vulliette 4, CH-1000, Lausanne 25, Switzerland
| | - Sylvie Duchesne
- INRAP National Institute of Preventive Archaeological Research (INRAP, France), 37 rue du Bignon, CS 67737, 35577, Cesson-Sévigné, France
- AMIS Laboratory: University of Toulouse, French National Center for Scientific Research, UMR 5288, 37 allées Jules Guesde, 31073, Toulouse, France
| | - Patrice Gérard
- AMIS Laboratory: University of Toulouse, French National Center for Scientific Research, UMR 5288, 37 allées Jules Guesde, 31073, Toulouse, France
| | - Laurent Dercle
- Gustave Roussy Institute, Université Paris-Saclay, F-94805, Villejuif, France
- New York Presbyterian Hospital, Columbia University, New York City, NY, USA
| | - Pierre Poilpré
- INRAP National Institute of Preventive Archaeological Research (INRAP, France), 37 rue du Bignon, CS 67737, 35577, Cesson-Sévigné, France
| | - Véronique Gendrot
- French Regional Archaeological Service, Bretagne Avenue Charles Foulon, 35700, Rennes, France
- French National Center for Scientific Research, UMR 6566, Rennes, France
| | - Hervé Rousseau
- Radiology Department, Rangueil University Hospital, 1 Avenue du Professeur Jean Poulhes, 31059 Cedex, Toulouse, France
| | - Éric Crubézy
- AMIS Laboratory: University of Toulouse, French National Center for Scientific Research, UMR 5288, 37 allées Jules Guesde, 31073, Toulouse, France
| | - Norbert Telmon
- AMIS Laboratory: University of Toulouse, French National Center for Scientific Research, UMR 5288, 37 allées Jules Guesde, 31073, Toulouse, France
- Forensic Department, Rangueil University Hospital, 1 avenue du Professeur Jean Poulhes, 31059 Cedex, Toulouse, France
| | - Fatima-Zohra Mokrane
- AMIS Laboratory: University of Toulouse, French National Center for Scientific Research, UMR 5288, 37 allées Jules Guesde, 31073, Toulouse, France.
- New York Presbyterian Hospital, Columbia University, New York City, NY, USA.
- Radiology Department, Rangueil University Hospital, 1 Avenue du Professeur Jean Poulhes, 31059 Cedex, Toulouse, France.
| |
Collapse
|
40
|
Chlamydia pneumoniae Infection Exacerbates Atherosclerosis in ApoB100only/LDLR -/- Mouse Strain. BIOMED RESEARCH INTERNATIONAL 2018; 2018:8325915. [PMID: 29770337 PMCID: PMC5889898 DOI: 10.1155/2018/8325915] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 01/31/2018] [Accepted: 02/18/2018] [Indexed: 11/25/2022]
Abstract
Aims Hyperlipidaemia model animals have been used to elucidate the role of Chlamydia pneumoniae (Cpn) infection in atherosclerosis. The aims of this study were to investigate the proatherogenic effect of multiple Cpn infections in ApoB100only/LDLR−/− mice which based on lipid profile can be regarded as the most suitable mouse model of human hypercholesterolemia and to compare the lesion development to that in a major atherosclerosis model ApoE−/− mice. Methods and Results Aorta samples of ApoB100only/LDLR−/− mice infected three times with Cpn were subjected to morphometric analyses. Morphometric evaluation disclosed that Cpn infections exacerbated atherosclerosis development in the aortic root and descending aorta of the mice fed with normal diet. Viable Cpn was detected in the ascending aorta by RT-PCR. Chlamydial 16SrRNA expression showed the presence of viable Cpn in the aorta of infected animals. A similar rate of acceleration of atherosclerosis was observed when the infection protocol was applied in ApoB100only/LDLR−/− and in ApoE−/− mice. Conclusion Similar to ApoE−/− mice, ApoB100only/LDLR−/− mice with more human-relevant serum lipoprotein composition develop increased atherosclerosis after Cpn infections; thus this mouse strain can be used as a model of infection-related atherosclerosis enhancement and can provide further evidence for the proatherogenic influence of Cpn in mice.
Collapse
|
41
|
Du X, Jiang S, Zeng X, Zhang J, Pan K, Zhou J, Xie Y, Kan H, Song W, Sun Q, Zhao J. Air pollution is associated with the development of atherosclerosis via the cooperation of CD36 and NLRP3 inflammasome in ApoE -/- mice. Toxicol Lett 2018; 290:123-132. [PMID: 29571893 DOI: 10.1016/j.toxlet.2018.03.022] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 03/13/2018] [Accepted: 03/19/2018] [Indexed: 12/22/2022]
Abstract
Previous studies have indicated that the main air pollutant fine particulate matter (≤2.5 μm; PM2.5) exposure is associated with the development of atherosclerosis. Although the mechanism is not fully illustrated, the inflammatory responses play an important role. The present study aimed to explore whether PM2.5-exacerbated atherosclerosis was mediated by the cooperation of cluster of differentiation 36 (CD36) and nucleotide-binding oligomerization domain-like receptor protein (NLRP3) inflammasome in apolipoprotein E-/- (ApoE-/-) mice. Thirty-two ApoE-/- mice were randomly divided into two groups. One group was fed with high fat chow (HFC) for 10 weeks to establish atherosclerotic model, and the other was fed with normal chow (NC). From week 11, the mice were exposed to concentrated PM2.5 (PM) or filtered air (FA) using Shanghai Meteorological and Environmental Animal Exposure System for 16 weeks. In both NC and HFC groups, PM2.5 exposure induced the formation of atherosclerosis plaque. Similarly, PM mice appeared higher lipid content in the aortic root than that in the FA mice. Compared with the FA mice, PM mice appeared a decrease in high density lipoprotein-cholesterol (HDL-C) and apolipoprotein A1 along with an increase in apolipoprotein B, low density lipoprotein-cholesterol (LDL-C) and oxidized low-density lipoprotein (ox-LDL). Moreover, PM2.5 exposure induced increase of CD36 in serum and aorta. In both NC and HFC groups, NLRP3 inflammasome activation-related indicators were activated or increased in the aorta of the PM mice when compared with the FA mice. The cooperation of CD36 and NLRP3 inflammasome activation may be the potential mechanisms linkixposed to concentrated PM2.5 (PM) or filtered air (FA) using Shanghai Meteorological and Environmental Animal Exposure System for 16 weeks. In both NC and HFC groups, PM2.5 exposure induced the formation of atherosclerosis plaque. Similarly, PM mice appeared higher lipid content in the aortic root than that in the FA mice. Compared with the FA mice, PM mice appeared a decrease in high density lipoprotein-cholesterol (HDL-C) and apolipoprotein A1 along with an increase in apolipoprotein B, low density lipoprotein-cholesterol (LDL-C) and oxidized low-density lipoprotein (ox-LDL). Moreover, PM2.5 exposure induced increase of CD36 in serum and aorta. In both NC and HFC groups, NLRP3 inflammasome activation-related indicators were activated or increased in the aorta of the PM mice when compared with the FA mice. The cooperation of CD36 and NLRP3 inflammasome activation may be the potential mechanisms linking air pollution and HFC-induced atherosclerosis even in the mice with NC intake.
Collapse
Affiliation(s)
- Xihao Du
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai, China
| | - Shuo Jiang
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai, China
| | - Xuejiao Zeng
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai, China
| | - Jia Zhang
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai, China
| | - Kun Pan
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai, China
| | - Ji Zhou
- Shanghai Key Laboratory of Meteorology and Health, Shanghai, China
| | - Yuquan Xie
- Department of Cardiology, Xinhua Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200092, China
| | - Haidong Kan
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai, China
| | - Weimin Song
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai, China
| | - Qinghua Sun
- Division of Environmental Health Sciences, College of Public Health, The Ohio State University, Columbus, OH, USA
| | - Jinzhuo Zhao
- Department of Environmental Health, School of Public Health and the Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai, China; Shanghai Key Laboratory of Meteorology and Health, Shanghai, China.
| |
Collapse
|
42
|
Hadj Ahmed S, Kharroubi W, Kaoubaa N, Zarrouk A, Batbout F, Gamra H, Najjar MF, Lizard G, Hininger-Favier I, Hammami M. Correlation of trans fatty acids with the severity of coronary artery disease lesions. Lipids Health Dis 2018; 17:52. [PMID: 29544473 PMCID: PMC5856295 DOI: 10.1186/s12944-018-0699-3] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 03/07/2018] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Nutritional choices, which include the source of dietary fatty acids (FA), have an important significant impact on coronary artery disease (CAD). We aimed to determine on patients with CAD the relationships between Trans fatty acids (Trans FA) and different CAD associated parameters such as inflammatory and oxidative stress parameters in addition to Gensini score as a vascular severity index. METHODS Fatty acid profiles were established by gas chromatography from 111 CAD patients compared to 120 age-matched control group. Lipid peroxidation biomarkers, oxidative stress, inflammatory parameters and Gensini score were studied. RESULTS Our study showed a significant decrease of the antioxidant parameters levels such as erythrocyte glutathione peroxydase (GPx) and superoxide dismutase (SOD) activities, plasma antioxidant status (FRAP) and thiol (SH) groups in CAD patients. On the other hand, catalase activity, conjugated dienes and malondialdehyde were increased. Plasmatic and erythrocyte Trans FA were also increased in CAD patients compared to controls. Furthermore, divergent associations of these Trans FA accumulations were observed with low-density lipoprotein-cholesterol/ high-density lipoprotein-cholesterol (LDL-C/HDL-C) ratio, Apolipoprotein B (ApoB), lipid peroxidation parameters, high-sensitivity C Reactive Protein (hs-CRP), Interleukin 6 (IL-6), tumor necrosis factor alpha (TNF-α) and Gensini score. Especially, elaidic acid (C18:1 trans 9), trans C18:2 isomers and trans 11 eicosanoic acid are correlated with these parameters. Trans FA are also associated with oxidative stress, confirmed by a positive correlation between C20:1 trans 11 and GPx in erythrocytes. CONCLUSIONS High level of Trans FA was highly associated with the induction of inflammation, oxidative stress and lipoperoxidation which appear to be based on the vascular severity and might be of interest to assess the stage and progression of atherosclerosis. The measurement of these Trans FA would be of great value for the screening of lipid metabolism disorders in CAD patients.
Collapse
Affiliation(s)
- Samia Hadj Ahmed
- Research Laboratory LR12ES05 LR-NAFS 'Nutrition - Functional Food & Vascular Health' Faculty of Medicine, University of Monastir, Avicene st, 5019, Monastir, Tunisia.
| | - Wafa Kharroubi
- Research Laboratory LR12ES05 LR-NAFS 'Nutrition - Functional Food & Vascular Health' Faculty of Medicine, University of Monastir, Avicene st, 5019, Monastir, Tunisia
| | - Nadia Kaoubaa
- Research Laboratory LR12ES05 LR-NAFS 'Nutrition - Functional Food & Vascular Health' Faculty of Medicine, University of Monastir, Avicene st, 5019, Monastir, Tunisia
| | - Amira Zarrouk
- Research Laboratory LR12ES05 LR-NAFS 'Nutrition - Functional Food & Vascular Health' Faculty of Medicine, University of Monastir, Avicene st, 5019, Monastir, Tunisia
| | - Fathi Batbout
- Department of Cardiology, CHU Fattouma Bourguiba, Monastir, Tunisia
| | - Habib Gamra
- Department of Cardiology, CHU Fattouma Bourguiba, Monastir, Tunisia
| | | | - Gérard Lizard
- Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA 7270 / INSERM, University of Bourgogne Franche-Comté, Dijon, France
| | - Isabelle Hininger-Favier
- Laboratory of Fundamental and Applied Bioenergetic, INSERM, Grenoble Alpes University, F-38041, Grenoble, France
| | - Mohamed Hammami
- Research Laboratory LR12ES05 LR-NAFS 'Nutrition - Functional Food & Vascular Health' Faculty of Medicine, University of Monastir, Avicene st, 5019, Monastir, Tunisia
| |
Collapse
|
43
|
Thrombosis-on-a-chip: Prospective impact of microphysiological models of vascular thrombosis. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2018; 5:29-34. [DOI: 10.1016/j.cobme.2017.12.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
44
|
Dorighello GG, Inada NM, Paim BA, Pardo-Andreu GL, Vercesi AE, Oliveira HCF. Mangifera indica L. extract (Vimang®) reduces plasma and liver cholesterol and leucocyte oxidative stress in hypercholesterolemic LDL receptor deficient mice. Cell Biol Int 2018; 42:747-753. [PMID: 29427465 DOI: 10.1002/cbin.10950] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 02/03/2018] [Indexed: 01/16/2023]
Abstract
Cardiovascular diseases are major causes of death worldwide. Beyond the classical cholesterol risk factor, other conditions such as oxidative stress are well documented to promote atherosclerosis. The Mangifera indica L. extract (Vimang®) was reported to present antioxidant and hypocholesterolemic properties. Thus, here we evaluate the effects of Vimang treatment on risk factors of the atherosclerosis prone model of familial hypercholesterolemia, the LDL receptor knockout mice. Mice were treated with Vimang during 2 weeks and were fed a cholesterol-enriched diet during the second week. The Vimang treated mice presented significantly reduced levels of plasma (15%) and liver (20%) cholesterol, increased plasma total antioxidant capacity (10%) and decreased reactive oxygen species (ROS) production by spleen mononuclear cells (50%), P < 0.05 for all. In spite of these benefits, the average size of aortic atherosclerotic lesions stablished in this short experimental period did not change significantly in Vimang treated mice. Therefore, in this study we demonstrated that Vimang has protective effects on systemic and tissue-specific risk factors, but it is not sufficient to promote a reduction in the initial steps of atherosclerosis development. In addition, we disclosed a new antioxidant target of Vimang, the spleen mononuclear cells that might be relevant for more advanced stages of atherosclerosis.
Collapse
Affiliation(s)
- Gabriel G Dorighello
- Department of Structural and Functional Biology, Biology Institute, State University of Campinas, Campinas, São Paulo, Brazil
| | - Natália M Inada
- Faculty of Medical Sciences, Department of Clinical Pathology, State University of Campinas, Campinas, São Paulo, Brazil
| | - Bruno A Paim
- Faculty of Medical Sciences, Department of Clinical Pathology, State University of Campinas, Campinas, São Paulo, Brazil
| | - Gilberto L Pardo-Andreu
- Centro de Estudio para las Investigaciones y Evaluaciones Biológicas, Instituto de Farmacia y Alimentos de La Universidad de La Habana, La Habana, Cuba
| | - Anibal E Vercesi
- Faculty of Medical Sciences, Department of Clinical Pathology, State University of Campinas, Campinas, São Paulo, Brazil
| | - Helena C F Oliveira
- Department of Structural and Functional Biology, Biology Institute, State University of Campinas, Campinas, São Paulo, Brazil
| |
Collapse
|
45
|
Li J, Yao Q, Feng F, He S, Lin P, Yang L, Yang C, Li H, Li Y. Systematic identification of rabbit LncRNAs reveals functional roles in atherosclerosis. Biochim Biophys Acta Mol Basis Dis 2018; 1864:2266-2273. [PMID: 29317334 DOI: 10.1016/j.bbadis.2017.12.040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 12/04/2017] [Accepted: 12/28/2017] [Indexed: 12/24/2022]
Abstract
Long noncoding RNAs (lncRNAs) have been gradually emerging as important regulators in various biological processes and diseases, while the contributions of lncRNAs to atherosclerosis remain largely unknown. Our previous work has discovered atherosclerosis associated protein-coding genes by transcriptome sequencing of rabbit models. Here we investigated the roles of lncRNAs in atherosclerosis. We defined a stringent set of 3736 multi-exonic lncRNA transcripts in rabbits. All lncRNAs are firstly reported and 609 (16.3%) of them are conserved in 13 species. Rabbit lncRNAs have similar characteristics to lncRNAs in other mammals, such as relatively short length, low expression, and highly tissue-specificity. The integrative analysis of lncRNAs and co-expressed genes characterize diverse functions of lncRNAs. Comparing two kinds of atherosclerosis models (LDLR-deficient WHHL rabbits and cholesterol-fed NZW rabbits) with their corresponding controls, we found the expression changes of two rabbit models were similar in aorta in but different in liver. The shared change in aorta revealed a subset of lncRNAs involved in immune response, while the cholesterol-fed NZW rabbits showed broader lncRNA expression changes in skeletal muscle system compared to WHHL rabbits. These atherosclerosis-associated lncRNAs and genes provide hits for the experimental validation of lncRNA functions. In summary, our study systematically identified rabbit lncRNAs for the first time and provides new insights for understanding the functions of lncRNAs in atherosclerosis. This article is part of a Special Issue entitled: Accelerating Precision Medicine through Genetic and Genomic Big Data Analysis edited by Yudong Cai & Tao Huang.
Collapse
Affiliation(s)
- Jia Li
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; CAS Key Laboratory for Computational Biology, CAS-MPG Partner Institute for Computing Biology, Shanghai Institute for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Qianlan Yao
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200031, China
| | - Fangyoumin Feng
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; CAS Key Laboratory for Computational Biology, CAS-MPG Partner Institute for Computing Biology, Shanghai Institute for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Sheng He
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; CAS Key Laboratory for Computational Biology, CAS-MPG Partner Institute for Computing Biology, Shanghai Institute for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Ping Lin
- CAS Key Laboratory for Computational Biology, CAS-MPG Partner Institute for Computing Biology, Shanghai Institute for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Liguang Yang
- CAS Key Laboratory for Computational Biology, CAS-MPG Partner Institute for Computing Biology, Shanghai Institute for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Chuhua Yang
- CAS Key Laboratory for Computational Biology, CAS-MPG Partner Institute for Computing Biology, Shanghai Institute for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Hong Li
- CAS Key Laboratory for Computational Biology, CAS-MPG Partner Institute for Computing Biology, Shanghai Institute for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China.
| | - Yixue Li
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; CAS Key Laboratory for Computational Biology, CAS-MPG Partner Institute for Computing Biology, Shanghai Institute for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200031, China.
| |
Collapse
|
46
|
Boshtam M, Asgary S, Rahimmanesh I, Kouhpayeh S, Naderi J, Hejazi Z, Mohammad-Dezashibi H, Pieper IL, Khanahmad H. Display of human and rabbit monocyte chemoattractant protein-1 on human embryonic kidney 293T cell surface. Res Pharm Sci 2018; 13:430-439. [PMID: 30271445 PMCID: PMC6082034 DOI: 10.4103/1735-5362.236836] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Monocyte chemoattractant protein-1 (MCP-1/CCL2) is a protein that is secreted immediately upon endothelial injury, and thereby it plays a key role in inflammation via recruitment of leucocytes to the site of inflammation at the beginning and throughout the inflammatory processes. Aim of this study was to develop two separate cell lines displaying either human MCP-1 (HMCP-1) or rabbit MCP-1 (RMCP-1) on their surface. A DNA fragment containing HMCP-1- or RMCP-1-encoding sequence was inserted into a pcDNA plasmid. Escherichia coli cells strain TOP 10F' was separately transformed with the pcDNA/RMCP-1 or /HMCP-1 ligation mixture. Following the cloning and construct verification, human embryonic kidney cell line (HEK 293T) was transfected with either of the linearized plasmids. Plasmid integration into the genomic DNA of HEK 293T cells was verified by polymerase chain reaction (PCR). HMCP-1 and RMCP-1 expression was evaluated at RNA and protein levels by real-time PCR and flow cytometry, respectively. PCR products of the expected sizes were amplified from the chromosomal DNA of transfected HEK 293T cells, i.e. 644 bp for H-MCP1 and 737 bp for RMCP-1 constructs. Real-time PCR revealed that the copy numbers of RMCP1 and HMCP1 mRNA per cell were 294 and 500, respectively. Flow cytometry analysis indicated 85% for RMCP-1 and 87% for HMCP-1 expression levels on the surface of transfected cells, when compared with an isotype control. The experiments thus confirmed that the MCP-1 genes were integrated into the HEK 293T genomic DNA and the encoded proteins were stably expressed on the cell surface.
Collapse
Affiliation(s)
- Maryam Boshtam
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| | - Seddigheh Asgary
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| | - Ilnaz Rahimmanesh
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| | - Shirin Kouhpayeh
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| | - Jamal Naderi
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| | - Zahra Hejazi
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| | - Hoda Mohammad-Dezashibi
- Department of Genetics and Molecular Biology, School of Medicine, Semnan University of Medical Sciences, Semnan, I.R. Iran
| | - Ina Laura Pieper
- Institute of Life Science, Swansea University Medical School, Swansea, United Kingdom
| | - Hossein Khanahmad
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| |
Collapse
|
47
|
Animal models of atherosclerosis. Eur J Pharmacol 2017; 816:3-13. [DOI: 10.1016/j.ejphar.2017.05.010] [Citation(s) in RCA: 387] [Impact Index Per Article: 48.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 04/07/2017] [Accepted: 05/04/2017] [Indexed: 12/31/2022]
|
48
|
Shafik NM, Baalash A, Ebeid AM. Synergistic Cardioprotective Effects of Combined Chromium Picolinate and Atorvastatin Treatment in Triton X-100-Induced Hyperlipidemia in Rats: Impact on Some Biochemical Markers. Biol Trace Elem Res 2017; 180:255-264. [PMID: 28409410 DOI: 10.1007/s12011-017-1010-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 03/31/2017] [Indexed: 11/26/2022]
Abstract
Hyperlipidemia is one of the major risk factors for atherosclerosis and ischemic heart disease. Chromium (Cr) mineral is playing a crucial role in glucose and lipid homeostasis. The aim of this study was to evaluate the protective effects of combined chromium picolinate (CrPic) and atorvastatin treatment against hyperlipidemia-induced cardiac injury. Seventy-five male albino rats were divided into five groups (15 rats each). Hyperlipidemia was induced by intraperitoneal injection of a single dose of Triton X-100 (300 mg/kg body weight (b.w) (group ІІ). Treatment of hyperlipidemic rats was induced by daily administration of CrPic at a dose of 200 μg/kg b.w/day (group ІІІ), atorvastatin at a dose of 10 mg/kg/day (group IV), and combined treatment with both (group V) by gavage for 7 days. At the end of experiment, serum and heart tissues were obtained. Hyperlipidemia was confirmed by histopathology of heart tissues, marked serum dyslipidemia, increased atherogenic indices, and values of ischemia-modified albumin. In addition to increased values of proprotein convertase subtilisin/kexin type 9, activity of 3-hydroxy-3-methylglutaryl coenzyme A reductase enzyme and high relative expression levels of pentraxin-3 were observed. However, paraoxonase-1 activity was markedly decreased in the hyperlipidemic group. Significant improvement in all assessed parameters was observed in the rat group treated with both CrPic and atorvastatin. It can be concluded that combined CrPic and atorvastatin treatments had synergistic cardioprotective effects against hyperlipidemia which may be through modulating atherosclerosis as well as cardiac and aortic damage and/or activation of anti-inflammatory and anti-oxidant pathways, thus reversing endothelial dysfunction.
Collapse
Affiliation(s)
- Noha M Shafik
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Tanta University, Tanta, 31111, Egypt.
| | - Amal Baalash
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Tanta University, Tanta, 31111, Egypt
| | - Abla M Ebeid
- Department of Clinical Pharmacy, Faculty of Pharmacy, Al-Delta University, Gamasa, Egypt
| |
Collapse
|
49
|
Korber M, Klein I, Daum G. Steryl ester synthesis, storage and hydrolysis: A contribution to sterol homeostasis. Biochim Biophys Acta Mol Cell Biol Lipids 2017; 1862:1534-1545. [DOI: 10.1016/j.bbalip.2017.09.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 08/25/2017] [Accepted: 09/05/2017] [Indexed: 02/01/2023]
|
50
|
CD80 Is Upregulated in a Mouse Model with Shear Stress-Induced Atherosclerosis and Allows for Evaluating CD80-Targeting PET Tracers. Mol Imaging Biol 2017; 19:90-99. [PMID: 27430577 DOI: 10.1007/s11307-016-0987-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
PURPOSE A shear stress-induced atherosclerosis mouse model was characterized for its expression of inflammation markers with focus on CD80. With this model, we evaluated two positron emission tomography (PET) radiotracers targeting CD80 as well as 2-deoxy-2-[18F]fluoro-D-mannose ([18F]FDM) in comparison with 2-deoxy-2-[18F]fluoro-D-glucose ([18F]FDG). PROCEDURE A flow constrictive cuff implanted around the common carotid artery in apolipoprotein E knockout mice resulted in plaque formation. CD80 expression levels and plaque histopathology were evaluated. Serial PET/X-ray computed tomography scans were performed to follow inflammation. RESULTS Plaque formation with increased levels of CD80 was observed. Histologically, plaques presented macrophage-rich and large necrotic areas covered by a thin fibrous cap. Of the CD80-specific tracers, one displayed an increased uptake in plaques by PET. Both [18F]FDG and [18F]FDM accumulated in atherosclerotic plaques. CONCLUSION This mouse model presented, similar to humans, an increased expression of CD80 which renders it suitable for non-invasively targeting CD80-positive immune cells and evaluating CD80-specific radiotracers.
Collapse
|