1
|
Abd Rahman IZ, Nor Hisam NS, Aminuddin A, Hamid AA, Kumar J, Ugusman A. Evaluating the Potential of Plukenetia volubilis Linneo (Sacha Inchi) in Alleviating Cardiovascular Disease Risk Factors: A Mini Review. Pharmaceuticals (Basel) 2023; 16:1588. [PMID: 38004453 PMCID: PMC10675584 DOI: 10.3390/ph16111588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 10/27/2023] [Accepted: 11/03/2023] [Indexed: 11/26/2023] Open
Abstract
Plukenetia volubilis Linneo or Sacha Inchi (SI), a traditional natural remedy indigenous to Peru and Brazil, has garnered global attention due to its exceptional nutritional composition. Its protective effects against various non-communicable diseases, notably cardiovascular disease (CVD), have become a subject of interest in recent research. This comprehensive review summarizes the existing evidence from 15 relevant articles concerning the impact of SI on common CVD risk factors, including dyslipidemia, obesity, diabetes, and hypertension. The relevant articles were derived from comprehensive searches on PubMed, Scopus, Google Scholar, and Web of Science using predefined criteria and keywords related to the topic. Overall, SI demonstrated positive effects in attenuating dyslipidemia, obesity, diabetes, and hypertension. The multifaceted mechanisms responsible for the protective effects of SI against these CVD risk factors are primarily attributed to its antioxidative and anti-inflammatory properties. While preclinical studies dominate the current scientific literature on SI, there are limited clinical trials to corroborate these findings. Therefore, future well-designed, large-scale randomized clinical trials are highly recommended to establish the efficacy of SI and determine its optimal dosage, potential drug and food interactions, and practical integration into preventive strategies and dietary interventions for the high-risk populations.
Collapse
Affiliation(s)
- Izzat Zulhilmi Abd Rahman
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Kuala Lumpur 56000, Malaysia; (I.Z.A.R.); (N.S.N.H.); (A.A.H.); (J.K.)
| | - Nur Syahidah Nor Hisam
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Kuala Lumpur 56000, Malaysia; (I.Z.A.R.); (N.S.N.H.); (A.A.H.); (J.K.)
- Programme of Biomedical Science, Centre for Toxicology & Health Risk Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia
| | - Amilia Aminuddin
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Kuala Lumpur 56000, Malaysia; (I.Z.A.R.); (N.S.N.H.); (A.A.H.); (J.K.)
| | - Adila A. Hamid
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Kuala Lumpur 56000, Malaysia; (I.Z.A.R.); (N.S.N.H.); (A.A.H.); (J.K.)
| | - Jaya Kumar
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Kuala Lumpur 56000, Malaysia; (I.Z.A.R.); (N.S.N.H.); (A.A.H.); (J.K.)
| | - Azizah Ugusman
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Kuala Lumpur 56000, Malaysia; (I.Z.A.R.); (N.S.N.H.); (A.A.H.); (J.K.)
| |
Collapse
|
2
|
Coppola A, Lombari P, Mazzella E, Capolongo G, Simeoni M, Perna AF, Ingrosso D, Borriello M. Zebrafish as a Model of Cardiac Pathology and Toxicity: Spotlight on Uremic Toxins. Int J Mol Sci 2023; 24:ijms24065656. [PMID: 36982730 PMCID: PMC10052014 DOI: 10.3390/ijms24065656] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/13/2023] [Accepted: 03/14/2023] [Indexed: 03/18/2023] Open
Abstract
Chronic kidney disease (CKD) is an increasing health care problem. About 10% of the general population is affected by CKD, representing the sixth cause of death in the world. Cardiovascular events are the main mortality cause in CKD, with a cardiovascular risk 10 times higher in these patients than the rate observed in healthy subjects. The gradual decline of the kidney leads to the accumulation of uremic solutes with a negative effect on every organ, especially on the cardiovascular system. Mammalian models, sharing structural and functional similarities with humans, have been widely used to study cardiovascular disease mechanisms and test new therapies, but many of them are rather expensive and difficult to manipulate. Over the last few decades, zebrafish has become a powerful non-mammalian model to study alterations associated with human disease. The high conservation of gene function, low cost, small size, rapid growth, and easiness of genetic manipulation are just some of the features of this experimental model. More specifically, embryonic cardiac development and physiological responses to exposure to numerous toxin substances are similar to those observed in mammals, making zebrafish an ideal model to study cardiac development, toxicity, and cardiovascular disease.
Collapse
Affiliation(s)
- Annapaola Coppola
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Patrizia Lombari
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Elvira Mazzella
- Department of Translational Medical Science, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Giovanna Capolongo
- Department of Translational Medical Science, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Mariadelina Simeoni
- Department of Translational Medical Science, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Alessandra F. Perna
- Department of Translational Medical Science, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Diego Ingrosso
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Margherita Borriello
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
- Correspondence:
| |
Collapse
|
3
|
Zhong R, Zhu Y, Zhang H, Huo Y, Huang Y, Cheng W, Liang P. Integrated lipidomic and transcriptomic analyses reveal the mechanism of large yellow croaker roe phospholipids on lipid metabolism in normal-diet mice. Food Funct 2022; 13:12852-12869. [PMID: 36444685 DOI: 10.1039/d2fo02736d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Large yellow croaker roe phospholipids (LYCRPLs) could regulate the accumulation of triglycerides and blood lipid levels. However, there exists little research on the mechanism of LYCRPLs on lipid metabolism in normal-diet mice. In this work, the mice on a normal diet were given low-dose, medium-dose, and high-dose LYCRPLs by intragastric administration for 6 weeks. At the same time, the physiological and biochemical indicators of the mice were determined, and the histomorphological observation of the liver and epididymal fat was carried out. In addition, we examined the gene expression and lipid metabolites in the liver of mice using transcriptomic and lipidomic and performed a correlation analysis. The results showed that LYCRPLs regulated the lipid metabolism of normal-diet mice by affecting the expression of the glycerolipid metabolism pathway, insulin resistance pathway, and cholesterol metabolism pathway. This study not only elucidated the main pathway by which LYCRPLs regulate lipid metabolism, but also laid a foundation for exploring LYCRPLs as functional food supplements.
Collapse
Affiliation(s)
- Rongbin Zhong
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, 350002 Fujian, China. .,Engineering Research Centre of Fujian-Taiwan Special Marine Food Processing and Nutrition, Ministry of Education, Fuzhou, 350002, Fujian, China
| | - Yujie Zhu
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, 350002 Fujian, China. .,Engineering Research Centre of Fujian-Taiwan Special Marine Food Processing and Nutrition, Ministry of Education, Fuzhou, 350002, Fujian, China
| | - Huadan Zhang
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, 350002 Fujian, China. .,Engineering Research Centre of Fujian-Taiwan Special Marine Food Processing and Nutrition, Ministry of Education, Fuzhou, 350002, Fujian, China
| | - Yuming Huo
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, 350002 Fujian, China. .,Engineering Research Centre of Fujian-Taiwan Special Marine Food Processing and Nutrition, Ministry of Education, Fuzhou, 350002, Fujian, China
| | - Ying Huang
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, 350002 Fujian, China. .,Engineering Research Centre of Fujian-Taiwan Special Marine Food Processing and Nutrition, Ministry of Education, Fuzhou, 350002, Fujian, China
| | - Wenjian Cheng
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, 350002 Fujian, China. .,Engineering Research Centre of Fujian-Taiwan Special Marine Food Processing and Nutrition, Ministry of Education, Fuzhou, 350002, Fujian, China
| | - Peng Liang
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, 350002 Fujian, China. .,Engineering Research Centre of Fujian-Taiwan Special Marine Food Processing and Nutrition, Ministry of Education, Fuzhou, 350002, Fujian, China
| |
Collapse
|
4
|
Balamurugan K, Medishetti R, Rao P, K RV, Chatti K, Parsa KV. Protocol to evaluate hyperlipidemia in zebrafish larvae. STAR Protoc 2022; 3:101819. [DOI: 10.1016/j.xpro.2022.101819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
|
5
|
Verwilligen RAF, Mulder L, Rodenburg FJ, Van Dijke A, Hoekstra M, Bussmann J, Van Eck M. Stabilin 1 and 2 are important regulators for cellular uptake of apolipoprotein B-containing lipoproteins in zebrafish. Atherosclerosis 2022; 346:18-25. [PMID: 35247629 DOI: 10.1016/j.atherosclerosis.2022.02.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 02/07/2022] [Accepted: 02/16/2022] [Indexed: 01/01/2023]
Abstract
BACKGROUND AND AIMS Scavenger receptors form a superfamily of membrane-bound receptors that bind and internalize different types of ligands, including pro-atherogenic oxidized low-density lipoproteins (oxLDLs). In vitro studies have indicated a role for the liver sinusoidal endothelial cell receptors stabilin 1 (stab1) and 2 (stab2) in oxLDL clearance. In this study, we evaluated the potential role of stab1 and stab2 in lipoprotein uptake in zebrafish, an upcoming model for studying cholesterol metabolism and atherosclerosis. METHODS Lipoproteins were injected in the duct of Cuvier of wild-type (ABTL) or stab1 and stab2 mutant (stab1-/-stab2-/-) zebrafish larvae at 3 days post-fertilization. To examine the effect of stabilin deficiency on lipoprotein and cholesterol metabolism, zebrafish larvae were challenged with a high cholesterol diet (HCD; 4% w/w) for 10 days. RESULTS Lipoprotein injections showed impaired uptake of both LDL and oxLDL into the vessel wall of caudal veins of stab1-/-stab2-/- zebrafish, which was paralleled by redistribution to tissue macrophages. Total body cholesterol levels did not differ between HCD-fed stab1-/-stab2-/- and ABTL zebrafish. However, stab1-/-stab2-/- larvae exhibited 1.4-fold higher mRNA expression levels of ldlra involved in (modified) LDL uptake, whereas the expression levels of scavenger receptors scarb1 and cd36 were significantly decreased. CONCLUSIONS We have shown that stabilins 1 and 2 have an important scavenging function for apolipoprotein B-containing lipoproteins in zebrafish and that combined deficiency of these two proteins strongly upregulates the clearance of lipoproteins by macrophages within the caudal vein. Our current study highlights the use of zebrafish as model to study lipoprotein metabolism and liver sinusoidal endothelial cell function.
Collapse
Affiliation(s)
- Robin A F Verwilligen
- Division BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, the Netherlands.
| | - Lindsay Mulder
- Division BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, the Netherlands
| | | | - Amy Van Dijke
- Division BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, the Netherlands
| | - Menno Hoekstra
- Division BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, the Netherlands
| | - Jeroen Bussmann
- Division BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, the Netherlands
| | - Miranda Van Eck
- Division BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, the Netherlands
| |
Collapse
|
6
|
Kulkarni A, Ibrahim S, Haider I, Basha A, Montgomery E, Ermis E, Mirmira RG, Anderson RM. A Novel 2-Hit Zebrafish Model to Study Early Pathogenesis of Non-Alcoholic Fatty Liver Disease. Biomedicines 2022; 10:479. [PMID: 35203687 PMCID: PMC8962409 DOI: 10.3390/biomedicines10020479] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 02/15/2022] [Accepted: 02/15/2022] [Indexed: 01/27/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is one of the most common liver diseases in adults. NAFLD progresses from benign liver fat accumulation to liver inflammation and cirrhosis, and ultimately leads to liver failure. Although several rodent models have been established for studying NAFLD, they have limitations that include cost, speed of disease development, key dissimilarities, and poor amenability to pharmacological screens. Here, we present a novel 2-hit zebrafish model to replicate aspects of NAFLD pathogenesis. We fed zebrafish larvae a high-fat diet (HFD) to drive liver fat accumulation (first hit). Next, we exacerbated liver-specific inflammation using a transgenic line (fabp10-CETI-PIC3) that induces the expression of proinflammatory cytokines following induction with doxycycline (second hit). These hits promoted fat accumulation and liver inflammation, as demonstrated by the high expression of inflammatory cytokines, macrophage infiltration, stress induction, and hepatic lipid droplet accumulation. Furthermore, zebrafish in this paradigm showed deranged glucose metabolism. To validate a small-molecule screening approach, we treated HFD-fed fish with pioglitazone, a drug shown to be beneficial for NAFLD in humans, and measured a sharp reduction in liver lipid accumulation. These results demonstrate new utility for zebrafish in modeling early NAFLD pathogenesis and demonstrate their feasibility for in vivo screening of new pharmacological interventions.
Collapse
Affiliation(s)
- Abhishek Kulkarni
- Department of Medicine, The University of Chicago, Chicago, IL 60637, USA; (A.K.); (A.B.); (E.M.); (E.E.)
| | - Sara Ibrahim
- The Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (S.I.); (I.H.)
| | - Isra Haider
- The Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (S.I.); (I.H.)
| | - Amina Basha
- Department of Medicine, The University of Chicago, Chicago, IL 60637, USA; (A.K.); (A.B.); (E.M.); (E.E.)
| | - Emma Montgomery
- Department of Medicine, The University of Chicago, Chicago, IL 60637, USA; (A.K.); (A.B.); (E.M.); (E.E.)
| | - Ebru Ermis
- Department of Medicine, The University of Chicago, Chicago, IL 60637, USA; (A.K.); (A.B.); (E.M.); (E.E.)
| | - Raghavendra G. Mirmira
- Department of Medicine, The University of Chicago, Chicago, IL 60637, USA; (A.K.); (A.B.); (E.M.); (E.E.)
| | - Ryan M. Anderson
- Department of Medicine, The University of Chicago, Chicago, IL 60637, USA; (A.K.); (A.B.); (E.M.); (E.E.)
| |
Collapse
|
7
|
Biomarker profiling of postmortem blood for diabetes mellitus and discussion of possible applications of metabolomics for forensic casework. Int J Legal Med 2022; 136:1075-1090. [DOI: 10.1007/s00414-021-02767-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 12/14/2021] [Indexed: 10/19/2022]
|
8
|
Liu W, Liu J, Xing S, Li X, Han L, Liu K, Wei T, Zhou M. Marine Phospholipids from Fishery By‐Products Attenuate Atherosclerosis. EUR J LIPID SCI TECH 2021. [DOI: 10.1002/ejlt.202000276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Wenjie Liu
- School of Chemistry and Chemical Engineering Qilu University of Technology (Shandong Academy of Sciences) # 3501 Daxue Road Jinan Shandong 250353 China
| | - Jianmin Liu
- School of Chemistry and Chemical Engineering Qilu University of Technology (Shandong Academy of Sciences) # 3501 Daxue Road Jinan Shandong 250353 China
| | - Shu Xing
- School of Chemistry and Chemical Engineering Qilu University of Technology (Shandong Academy of Sciences) # 3501 Daxue Road Jinan Shandong 250353 China
| | - Xiaobin Li
- Biology Institute Qilu University of Technology (Shandong Academy of Sciences) 28799 Jingshidong Road Jinan Shandong 250103 China
| | - Liwen Han
- Biology Institute Qilu University of Technology (Shandong Academy of Sciences) 28799 Jingshidong Road Jinan Shandong 250103 China
| | - Kechun Liu
- Biology Institute Qilu University of Technology (Shandong Academy of Sciences) 28799 Jingshidong Road Jinan Shandong 250103 China
| | - Tao Wei
- School of Chemistry and Chemical Engineering Qilu University of Technology (Shandong Academy of Sciences) # 3501 Daxue Road Jinan Shandong 250353 China
| | - Mingyang Zhou
- School of Chemistry and Chemical Engineering Qilu University of Technology (Shandong Academy of Sciences) # 3501 Daxue Road Jinan Shandong 250353 China
| |
Collapse
|
9
|
Hu HJ, Qiu J, Zhang C, Tang ZH, Qu SL, Jiang ZS. Hydrogen sulfide improves ox‑LDL‑induced expression levels of Lp‑PLA 2 in THP‑1 monocytes via the p38MAPK pathway. Mol Med Rep 2021; 23:358. [PMID: 33760156 PMCID: PMC7974332 DOI: 10.3892/mmr.2021.11997] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Accepted: 01/22/2021] [Indexed: 12/22/2022] Open
Abstract
Hydrogen sulfide (H2S) exerts an anti‑atherosclerotic effect and decreases foam cell formation. Lipoprotein‑associated phospholipase A2 (Lp‑PLA2) is a key factor involved in foam cell formation. However, the association between H2S and Lp‑PLA2 expression levels with respect to foam cell formation has not yet been elucidated. The present study investigated whether H2S can affect foam cell formation and potential signalling pathways via regulation of the expression and activity of Lp‑PLA2. Using human monocytic THP‑1 cells as a model system, it was observed that oxidized low‑density lipoprotein (ox‑LDL) not only upregulates the expression level and activity of Lp‑PLA2, it also downregulates the expression level and activity of Cystathionine γ lyase. Exogenous supplementation of H2S decreased the expression and activity of Lp‑PLA2 induced by ox‑LDL. Moreover, ox‑LDL induced the expression level and activity of Lp‑PLA2 via activation of the p38MAPK signalling pathway. H2S blocked the expression levels and activity of Lp‑PLA2 induced by ox‑LDL via inhibition of the p38MAPK signalling pathway. Furthermore, H2S inhibited Lp‑PLA2 activity by blocking the p38MAPK signaling pathway and significantly decreased lipid accumulation in ox‑LDL‑induced macrophages, as detected by Oil Red O staining. The results of the present study indicated that H2S inhibited ox‑LDL‑induced Lp‑PLA2 expression levels and activity by blocking the p38MAPK signalling pathway, thereby improving foam cell formation. These findings may provide novel insights into the role of H2S intervention in the progression of atherosclerosis.
Collapse
Affiliation(s)
- Heng-Jing Hu
- Department of Cardiology Laboratory, First Affiliated Hospital of University of South China, Hengyang, Hunan 421001, P.R. China
| | - Jie Qiu
- Department of Cardiology Laboratory, Huazhong University of Science and Technology Tongji Medical College First Clinical College, Wuhan, Hubei 430000, P.R. China
| | - Chi Zhang
- Institute of Cardiovascular Disease and Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Zhi-Han Tang
- Institute of Cardiovascular Disease and Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Shun-Lin Qu
- Institute of Cardiovascular Disease and Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Zhi-Sheng Jiang
- Department of Cardiology Laboratory, First Affiliated Hospital of University of South China, Hengyang, Hunan 421001, P.R. China
| |
Collapse
|
10
|
Tang D, Geng F, Yu C, Zhang R. Recent Application of Zebrafish Models in Atherosclerosis Research. Front Cell Dev Biol 2021; 9:643697. [PMID: 33718384 PMCID: PMC7947229 DOI: 10.3389/fcell.2021.643697] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 02/05/2021] [Indexed: 12/13/2022] Open
Abstract
Atherosclerotic cardiovascular disease is one of the leading causes of death worldwide. Establishing animal models of atherosclerosis is of great benefit for studying its complicated pathogenesis and screening and evaluating related drugs. Although researchers have generated a variety of models for atherosclerosis study in rabbits, mice and rats, the limitations of these models make it difficult to monitor the development of atherosclerosis, and these models are unsuitable for large scale screening of potential therapeutic targets. On the contrast, zebrafish can fulfill these purposes thanks to their fecundity, rapid development ex utero, embryonic transparency, and conserved lipid metabolism process. Thus, zebrafish have become a popular alternative animal model for atherosclerosis research. In this mini review, we summarize different zebrafish models used to study atherosclerosis, focusing on the latest applications of these models to the dynamic monitoring of atherosclerosis progression, mechanistic study of therapeutic intervention and drug screening, and assessment of the impacts of other risk factors.
Collapse
Affiliation(s)
- Dandan Tang
- School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Fang Geng
- School of Life Sciences, Fudan University, Shanghai, China
| | - Chunxiao Yu
- School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Ruilin Zhang
- School of Basic Medical Sciences, Wuhan University, Wuhan, China
| |
Collapse
|
11
|
Ka J, Jin SW. Zebrafish as an Emerging Model for Dyslipidemia and Associated Diseases. J Lipid Atheroscler 2020; 10:42-56. [PMID: 33537252 PMCID: PMC7838516 DOI: 10.12997/jla.2021.10.1.42] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 11/06/2020] [Accepted: 11/30/2020] [Indexed: 01/03/2023] Open
Abstract
Dyslipidemia related diseases such as hyperlipidemia and atherosclerosis are the leading cause of death in humans. While cellular and molecular basis on the pathophysiology of dyslipidemia has been extensively investigated over decades, we still lack comprehensive understanding on the etiology of dyslipidemia due to the complexity and the innate multimodality of the diseases. While mouse has been the model organism of choice to investigate the pathophysiology of human dyslipidemia, zebrafish, a small freshwater fish which has traditionally used to study vertebrate development, has recently emerged as an alternative model organism. In this review, we will provide comprehensive perspective on zebrafish as a model organism for human dyslipidemia; we will discuss the attributes of zebrafish as a model, and compare the lipid metabolism in zebrafish and humans. In addition, we will summarize current landscape of zebrafish-based dyslipidemia research.
Collapse
Affiliation(s)
- Jun Ka
- Cell Logistics Research Center and School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Suk-Won Jin
- Cell Logistics Research Center and School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Korea.,Yale Cardiovascular Research Center and Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
12
|
Li P, Huang J, Xiao N, Cai X, Yang Y, Deng J, Zhang LH, Du B. Sacha inchi oil alleviates gut microbiota dysbiosis and improves hepatic lipid dysmetabolism in high-fat diet-fed rats. Food Funct 2020; 11:5827-5841. [PMID: 32648886 DOI: 10.1039/d0fo01178a] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Dietary ω-3 polyunsaturated fatty acids (PUFAs) are beneficial for humans against the development of hyperlipidaemia, but the underlying mechanisms are still poorly understood. Here, we demonstrated that oral consumption of sacha inchi oil, which is rich in α-linolenic acid, alleviated dyslipidemia, hepatic steatosis and inflammatory infiltration in high-fat diet (HFD)-fed rats. Sacha inchi oil administration reversed gut microbiota dysbiosis and altered the gut microbiota metabolome and in particular prevented bile acid dysmetabolism caused by a HFD. Sacha inchi oil intake ameliorated hepatic lipid dysmetabolism in HFD-fed rats, via potentiating the biosynthesis and reuptake of bile acids, reducing the de novo lipogenesis, promoting fatty acid beta-oxidation, and alleviating the dysregulation of glycerolipid, glycerophospholipid, and sphingolipid metabolisms. The results showed that dietary sacha inchi oil can alleviate gut microbiota dysbiosis and reduce lipid dysmetabolism in HFD rats, and provide novel insights into the molecular mechanisms by which plant-derived ω-3 PUFAs prevent the development of hyperlipidaemia.
Collapse
Affiliation(s)
- Pan Li
- College of Food Science, South China Agricultural University, Guangzhou 510642, China.
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Vedder VL, Aherrahrou Z, Erdmann J. Dare to Compare. Development of Atherosclerotic Lesions in Human, Mouse, and Zebrafish. Front Cardiovasc Med 2020; 7:109. [PMID: 32714944 PMCID: PMC7344238 DOI: 10.3389/fcvm.2020.00109] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 05/26/2020] [Indexed: 12/19/2022] Open
Abstract
Cardiovascular diseases, such as atherosclerosis, are the leading cause of death worldwide. Although mice are currently the most commonly used model for atherosclerosis, zebrafish are emerging as an alternative, especially for inflammatory and lipid metabolism studies. Here, we review the history of in vivo atherosclerosis models and highlight the potential for future studies on inflammatory responses in lipid deposits in zebrafish, based on known immune reactions in humans and mice, in anticipation of new zebrafish models with more advanced atherosclerotic plaques.
Collapse
Affiliation(s)
- Viviana L Vedder
- Institute for Cardiogenetics, University of Lübeck, Lübeck, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Lübeck, Germany.,University Heart Centre Lübeck, Lübeck, Germany
| | - Zouhair Aherrahrou
- Institute for Cardiogenetics, University of Lübeck, Lübeck, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Lübeck, Germany.,University Heart Centre Lübeck, Lübeck, Germany
| | - Jeanette Erdmann
- Institute for Cardiogenetics, University of Lübeck, Lübeck, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Lübeck, Germany.,University Heart Centre Lübeck, Lübeck, Germany
| |
Collapse
|
14
|
Jinnouchi H, Guo L, Sakamoto A, Torii S, Sato Y, Cornelissen A, Kuntz S, Paek KH, Fernandez R, Fuller D, Gadhoke N, Surve D, Romero M, Kolodgie FD, Virmani R, Finn AV. Diversity of macrophage phenotypes and responses in atherosclerosis. Cell Mol Life Sci 2020; 77:1919-1932. [PMID: 31720740 PMCID: PMC11104939 DOI: 10.1007/s00018-019-03371-3] [Citation(s) in RCA: 127] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Revised: 10/29/2019] [Accepted: 11/05/2019] [Indexed: 12/12/2022]
Abstract
The presence of macrophages within the plaque is a defining hallmark of atherosclerosis. Macrophages are exposed to various microenvironments such as oxidized lipids and cytokines which effect their phenotypic differentiation and activation. Classically, macrophages have been divided into two groups: M1 and M2 macrophages induced by T-helper 1 and T-helper 2 cytokines, respectively. However, for a decade, greater phenotypic heterogeneity and plasticity of these cells have since been reported in various models. In addition to M1 and M2 macrophage phenotypes, the concept of additional macrophage phenotypes such as M (Hb), Mox, and M4 has emerged. Understanding the mechanisms and functions of distinct phenotype of macrophages can lead to determination of their potential role in atherosclerotic plaque pathogenesis. However, there are still many unresolved controversies regarding their phenotype and function with respect to atherosclerosis. Here, we summarize and focus on the differential subtypes of macrophages in atherosclerotic plaques and their differing functional roles based upon microenvironments such as lipid, intraplaque hemorrhage, and plaque regression.
Collapse
Affiliation(s)
| | - Liang Guo
- CVPath Institute, 19 Firstfield Road, Gaithersburg, MD, 20878, USA
| | - Atsushi Sakamoto
- CVPath Institute, 19 Firstfield Road, Gaithersburg, MD, 20878, USA
| | - Sho Torii
- CVPath Institute, 19 Firstfield Road, Gaithersburg, MD, 20878, USA
| | - Yu Sato
- CVPath Institute, 19 Firstfield Road, Gaithersburg, MD, 20878, USA
| | - Anne Cornelissen
- CVPath Institute, 19 Firstfield Road, Gaithersburg, MD, 20878, USA
| | - Salome Kuntz
- CVPath Institute, 19 Firstfield Road, Gaithersburg, MD, 20878, USA
| | - Ka Hyun Paek
- CVPath Institute, 19 Firstfield Road, Gaithersburg, MD, 20878, USA
| | - Raquel Fernandez
- CVPath Institute, 19 Firstfield Road, Gaithersburg, MD, 20878, USA
| | - Daniela Fuller
- CVPath Institute, 19 Firstfield Road, Gaithersburg, MD, 20878, USA
| | - Neel Gadhoke
- CVPath Institute, 19 Firstfield Road, Gaithersburg, MD, 20878, USA
| | - Dipti Surve
- CVPath Institute, 19 Firstfield Road, Gaithersburg, MD, 20878, USA
| | - Maria Romero
- CVPath Institute, 19 Firstfield Road, Gaithersburg, MD, 20878, USA
| | - Frank D Kolodgie
- CVPath Institute, 19 Firstfield Road, Gaithersburg, MD, 20878, USA
| | - Renu Virmani
- CVPath Institute, 19 Firstfield Road, Gaithersburg, MD, 20878, USA
| | - Aloke V Finn
- CVPath Institute, 19 Firstfield Road, Gaithersburg, MD, 20878, USA.
| |
Collapse
|
15
|
Williams MB, Watts SA. Current basis and future directions of zebrafish nutrigenomics. GENES AND NUTRITION 2019; 14:34. [PMID: 31890052 PMCID: PMC6935144 DOI: 10.1186/s12263-019-0658-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 12/16/2019] [Indexed: 12/19/2022]
Abstract
This review investigates the current state of nutrigenomics in the zebrafish animal models. The zebrafish animal model has been used extensively in the study of disease onset and progression and associated molecular changes. In this review, we provide a synopsis of nutrigenomics using the zebrafish animal model. Obesity and dyslipidemia studies describe the genomics of dietary-induced obesity in relation to high-fat/high-calorie diets. Inflammation and cardiovascular studies describe dietary effects on the expression of acute inflammatory markers and resulting chronic inflammatory issues including atherosclerosis. We also evaluated the genomic response to bioactive dietary compounds associated with metabolic disorders. Carbohydrate metabolism and β-cell function studies describe the impacts of high-carbohydrate dietary challenges on nutritional programming. We also report tumorigenesis in relation to dietary carcinogen exposure studies that can result in permanent genomic changes. Vitamin and mineral deficiency studies demonstrate transgenerational genomic impacts of micronutrients in the diet and temporal expression changes. Circadian rhythm studies describe the relation between metabolism and natural temporal cycles of gene expression that impacts health. Bone formation studies describe the role of dietary composition that influences bone reabsorption regulation. Finally, this review provides future directions in the use of the zebrafish model for nutrigenomic and nutrigenetic research.
Collapse
Affiliation(s)
- Michael B Williams
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL 35294 USA
| | - Stephen A Watts
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL 35294 USA
| |
Collapse
|
16
|
Luo H, Li QQ, Wu N, Shen YG, Liao WT, Yang Y, Dong E, Zhang GM, Liu BR, Yue XZ, Tang XQ, Yang HS. Chronological in vivo imaging reveals endothelial inflammation prior to neutrophils accumulation and lipid deposition in HCD-fed zebrafish. Atherosclerosis 2019; 290:125-135. [DOI: 10.1016/j.atherosclerosis.2019.09.017] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 09/20/2019] [Accepted: 09/25/2019] [Indexed: 12/20/2022]
|
17
|
Yu Z, Wang N, Ahn DU, Ma M. Long Term Egg Yolk Consumption Alters Lipid Metabolism and Attenuates Hyperlipidemia in Mice Fed a High‐Fat Diet Based on Lipidomics Analysis. EUR J LIPID SCI TECH 2019. [DOI: 10.1002/ejlt.201800496] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Zhihui Yu
- National R&D Center for Egg Processing, College of Food Science and Technology, Huazhong Agricultural UniversityWuhan 430070HubeiChina
| | - Ning Wang
- College of Food and Biological Engineering, Henan University of Animal Husbandry and EconomyZhengzhou 450046HenanChina
| | - Dong U. Ahn
- Department of Animal Science, Iowa State UniversityAmesIA50011USA
| | - Meihu Ma
- National R&D Center for Egg Processing, College of Food Science and Technology, Huazhong Agricultural UniversityWuhan 430070HubeiChina
| |
Collapse
|
18
|
Momtazi-Borojeni AA, Abdollahi E, Nikfar B, Chaichian S, Ekhlasi-Hundrieser M. Curcumin as a potential modulator of M1 and M2 macrophages: new insights in atherosclerosis therapy. Heart Fail Rev 2019; 24:399-409. [DOI: 10.1007/s10741-018-09764-z] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
19
|
Gonen A, Choi SH, Miu P, Agatisa-Boyle C, Acks D, Taylor AM, McNamara CA, Tsimikas S, Witztum JL, Miller YI. A monoclonal antibody to assess oxidized cholesteryl esters associated with apoAI and apoB-100 lipoproteins in human plasma. J Lipid Res 2018; 60:436-445. [PMID: 30563909 PMCID: PMC6358287 DOI: 10.1194/jlr.d090852] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 12/15/2018] [Indexed: 11/20/2022] Open
Abstract
Atherosclerosis is associated with increased lipid peroxidation, leading to generation of multiple oxidation-specific epitopes (OSEs), contributing to the pathogenesis of atherosclerosis and its clinical manifestation. Oxidized cholesteryl esters (OxCEs) are a major class of OSEs found in human plasma and atherosclerotic tissue. To evaluate OxCEs as a candidate biomarker, we generated a novel mouse monoclonal Ab (mAb) specific to an OxCE modification of proteins. The mAb AG23 (IgG1) was raised in C57BL6 mice immunized with OxCE-modified keyhole limpet hemocyanin, and hybridomas were screened against OxCE-modified BSA. This method ensures mAb specificity to the OxCE modification, independent of a carrier protein. AG23 specifically stained human carotid artery atherosclerotic lesions. An ELISA method, with AG23 as a capture and either anti-apoAI or anti-apoB-100 as the detection Abs, was developed to assay apoAI and apoB-100 lipoproteins that have one or more OxCE epitopes. OxCE-apoA or OxCE-apoB did not correlate with the well-established oxidized phospholipid-apoB biomarker. In a cohort of subjects treated with atorvastatin, OxCE-apoA was significantly lower than in the placebo group, independent of the apoAI levels. These results suggest the potential diagnostic utility of a new biomarker assay to measure OxCE-modified lipoproteins in patients with CVD.
Collapse
Affiliation(s)
- Ayelet Gonen
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Soo-Ho Choi
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Phuong Miu
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Colin Agatisa-Boyle
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Daniel Acks
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Angela M Taylor
- Cardiovascular Research Center, Department of Medicine, University of Virginia, Charlottesville, VA 22908
| | - Coleen A McNamara
- Cardiovascular Research Center, Department of Medicine, University of Virginia, Charlottesville, VA 22908
| | - Sotirios Tsimikas
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Joseph L Witztum
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Yury I Miller
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| |
Collapse
|
20
|
CHEN Z, ZANG L, WU Y, NAKAYAMA H, SHIMADA Y, SHRESTHA R, ZHAO Y, MIURA Y, CHIBA H, HUI SP, NISHIMURA N. Lipidomic Profiling on Oxidized Phospholipids in Type 2 Diabetes Mellitus Model Zebrafish. ANAL SCI 2018; 34:1201-1208. [DOI: 10.2116/analsci.18p281] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Affiliation(s)
- Zhen CHEN
- Faculty of Health Sciences, Hokkaido University
| | - Liqing ZANG
- Graduate School of Regional Innovation Studies, Mie University
| | - Yue WU
- Faculty of Health Sciences, Hokkaido University
| | - Hiroko NAKAYAMA
- Graduate School of Regional Innovation Studies, Mie University
| | - Yasuhito SHIMADA
- Department of Integrative Pharmacology, Graduate School of Medicine, Mie University
- Mie University Zebrafish Drug Screening Center
| | | | - Yaoyao ZHAO
- Faculty of Health Sciences, Hokkaido University
| | | | - Hitoshi CHIBA
- Department of Nutrition, Sapporo University of Health Sciences
| | | | | |
Collapse
|
21
|
Interleukin-1 genotypes modulate the long-term effect of lipoprotein(a) on cardiovascular events: The Ioannina Study. J Clin Lipidol 2018; 12:338-347. [DOI: 10.1016/j.jacl.2017.12.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 12/11/2017] [Accepted: 12/13/2017] [Indexed: 12/22/2022]
|
22
|
Anti-hypercholesterolemic Effect of Berbamine Isolated from Rhizoma Coptidis in Hypercholesterolemic Zebrafish Induced by High-Cholesterol Diet. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2018; 17:292-306. [PMID: 29755560 PMCID: PMC5937099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The anti-hypercholesterolemic effect of berbamine (BBM) isolated from Rhizoma Coptidis (RC) was investigated in hypercholesterolemic zebrafish model induced by high-cholesterol (HC) diet. Zebrafish embryo assay revealed no significant difference in morphology and cell death with the treatment of BBM less than 20 μg/mL. In zebrafish larvae, the fluorescently labeled cholesterol in caudal artery was reduced dose-dependently after BBM treatment. For adult zebrafish, administration of 0.2% BBM exhibited a significant decrease in plasma total cholesterol (TC), triglyceride (TG) and low-density lipoprotein cholesterol (LDL-c) levels by 37%, 38% and 28%, respectively, along with a fall in lipid content in liver. Further investigation suggested that the mRNA expression of 3-hydroxy-3-methylglutaryl-coenzyme A reductase (HMGCR) and microsomal triglyceride transfer protein (MTP) in liver were down-regulated and the transcription levels of liver gene low-density lipoprotein receptor (LDLR) and cytochrome P450 polypeptide 1a of subfamily A of family 7 (CYP7A1a) were significantly up-regulated with BBM treatment. Histological study showed that BBM can alleviate hepatic steatosis induced by HC diet. These data suggested that BBM has anti-hypercholesterolemic and hepatoprotective effects. The mechanism probably related to the up-regulation of cholesterol transport and bile acid synthesis as well as inhibition of cholesterol synthesis and lipoprotein assembly or secretion.
Collapse
|
23
|
Liu C, Kim YS, Kim J, Pattison J, Kamaid A, Miller YI. Modeling hypercholesterolemia and vascular lipid accumulation in LDL receptor mutant zebrafish. J Lipid Res 2017; 59:391-399. [PMID: 29187523 DOI: 10.1194/jlr.d081521] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 11/11/2017] [Indexed: 12/11/2022] Open
Abstract
Elevated plasma LDL cholesterol is the dominant risk factor for the development of atherosclerosis and cardiovascular disease. Deficiency in the LDL receptor (LDLR) is a major cause of familial hypercholesterolemia in humans, and the LDLR knockout mouse is a major animal model of atherosclerosis. Here we report the generation and characterization of an ldlr mutant zebrafish as a new animal model to study hypercholesterolemia and vascular lipid accumulation, an early event in the development of human atherosclerosis. The ldlr mutant zebrafish were characterized by activated SREBP-2 pathway and developed moderate hypercholesterolemia when fed a normal diet. However, a short-term, 5-day feeding of ldlr mutant larvae with a high-cholesterol diet (HCD) resulted in exacerbated hypercholesterolemia and accumulation of vascular lipid deposits. Lomitapide, an inhibitor of apoB lipoprotein secretion, but not the antioxidant probucol, significantly reduced accumulation of vascular lipid deposits in HCD-fed ldlr mutant larvae. Furthermore, ldlr mutants were defective in hepatic clearance of lipopolysaccharides, resulting in reduced survival. Taken together, our data suggest that the ldlr knockout zebra-fish is a versatile model for studying the function of the LDL receptor, hypercholesterolemia, and related vascular pathology in the context of early atherosclerosis.
Collapse
Affiliation(s)
- Chao Liu
- Department of Medicine, University of California, San Diego, La Jolla, CA
| | - Young Sook Kim
- Department of Medicine, University of California, San Diego, La Jolla, CA.,Korean Medicine Convergence Research Division, Korea Institute of Oriental Medicine Republic of Korea, Daejeon, South Korea
| | - Jungsu Kim
- Department of Medicine, University of California, San Diego, La Jolla, CA
| | - Jennifer Pattison
- Department of Medicine, University of California, San Diego, La Jolla, CA
| | - Andrés Kamaid
- Korean Medicine Convergence Research Division, Korea Institute of Oriental Medicine Republic of Korea, Daejeon, South Korea.,Korean Medicine Convergence Research Division, Korea Institute of Oriental Medicine Republic of Korea, Daejeon, South Korea
| | - Yury I Miller
- Department of Medicine, University of California, San Diego, La Jolla, CA
| |
Collapse
|
24
|
Bochkov V, Gesslbauer B, Mauerhofer C, Philippova M, Erne P, Oskolkova OV. Pleiotropic effects of oxidized phospholipids. Free Radic Biol Med 2017; 111:6-24. [PMID: 28027924 DOI: 10.1016/j.freeradbiomed.2016.12.034] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 12/21/2016] [Accepted: 12/22/2016] [Indexed: 12/25/2022]
Abstract
Oxidized phospholipids (OxPLs) are increasingly recognized to play a role in a variety of normal and pathological states. OxPLs were implicated in regulation of inflammation, thrombosis, angiogenesis, endothelial barrier function, immune tolerance and other important processes. Rapidly accumulating evidence suggests that OxPLs are biomarkers of atherosclerosis and other pathologies. In addition, successful application of experimental drugs based on structural scaffold of OxPLs in animal models of inflammation was recently reported. This review briefly summarizes current knowledge on generation, methods of quantification and biological activities of OxPLs. Furthermore, receptor and cellular mechanisms of these effects are discussed. The goal of the review is to give a broad overview of this class of lipid mediators inducing pleiotropic biological effects.
Collapse
Affiliation(s)
- Valery Bochkov
- Institute of Pharmaceutical Sciences, Department of Pharmaceutical Chemistry, University of Graz, Austria.
| | - Bernd Gesslbauer
- Institute of Pharmaceutical Sciences, Department of Pharmaceutical Chemistry, University of Graz, Austria
| | - Christina Mauerhofer
- Institute of Pharmaceutical Sciences, Department of Pharmaceutical Chemistry, University of Graz, Austria
| | - Maria Philippova
- Signaling Laboratory, Department of Biomedicine, Basel University Hospital, Basel, Switzerland
| | - Paul Erne
- Signaling Laboratory, Department of Biomedicine, Basel University Hospital, Basel, Switzerland
| | - Olga V Oskolkova
- Institute of Pharmaceutical Sciences, Department of Pharmaceutical Chemistry, University of Graz, Austria.
| |
Collapse
|
25
|
Gut P, Reischauer S, Stainier DYR, Arnaout R. LITTLE FISH, BIG DATA: ZEBRAFISH AS A MODEL FOR CARDIOVASCULAR AND METABOLIC DISEASE. Physiol Rev 2017; 97:889-938. [PMID: 28468832 PMCID: PMC5817164 DOI: 10.1152/physrev.00038.2016] [Citation(s) in RCA: 202] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 01/09/2017] [Accepted: 01/10/2017] [Indexed: 12/17/2022] Open
Abstract
The burden of cardiovascular and metabolic diseases worldwide is staggering. The emergence of systems approaches in biology promises new therapies, faster and cheaper diagnostics, and personalized medicine. However, a profound understanding of pathogenic mechanisms at the cellular and molecular levels remains a fundamental requirement for discovery and therapeutics. Animal models of human disease are cornerstones of drug discovery as they allow identification of novel pharmacological targets by linking gene function with pathogenesis. The zebrafish model has been used for decades to study development and pathophysiology. More than ever, the specific strengths of the zebrafish model make it a prime partner in an age of discovery transformed by big-data approaches to genomics and disease. Zebrafish share a largely conserved physiology and anatomy with mammals. They allow a wide range of genetic manipulations, including the latest genome engineering approaches. They can be bred and studied with remarkable speed, enabling a range of large-scale phenotypic screens. Finally, zebrafish demonstrate an impressive regenerative capacity scientists hope to unlock in humans. Here, we provide a comprehensive guide on applications of zebrafish to investigate cardiovascular and metabolic diseases. We delineate advantages and limitations of zebrafish models of human disease and summarize their most significant contributions to understanding disease progression to date.
Collapse
Affiliation(s)
- Philipp Gut
- Nestlé Institute of Health Sciences, EPFL Innovation Park, Lausanne, Switzerland; Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and Cardiovascular Research Institute and Division of Cardiology, Department of Medicine, University of California San Francisco, San Francisco, California
| | - Sven Reischauer
- Nestlé Institute of Health Sciences, EPFL Innovation Park, Lausanne, Switzerland; Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and Cardiovascular Research Institute and Division of Cardiology, Department of Medicine, University of California San Francisco, San Francisco, California
| | - Didier Y R Stainier
- Nestlé Institute of Health Sciences, EPFL Innovation Park, Lausanne, Switzerland; Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and Cardiovascular Research Institute and Division of Cardiology, Department of Medicine, University of California San Francisco, San Francisco, California
| | - Rima Arnaout
- Nestlé Institute of Health Sciences, EPFL Innovation Park, Lausanne, Switzerland; Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and Cardiovascular Research Institute and Division of Cardiology, Department of Medicine, University of California San Francisco, San Francisco, California
| |
Collapse
|
26
|
|
27
|
Byun YS, Yang X, Bao W, DeMicco D, Laskey R, Witztum JL, Tsimikas S. Oxidized Phospholipids on Apolipoprotein B-100 and Recurrent Ischemic Events Following Stroke or Transient Ischemic Attack. J Am Coll Cardiol 2017; 69:147-158. [DOI: 10.1016/j.jacc.2016.10.057] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 09/29/2016] [Accepted: 10/12/2016] [Indexed: 01/08/2023]
|
28
|
Lauriano ER, Pergolizzi S, Capillo G, Kuciel M, Alesci A, Faggio C. Immunohistochemical characterization of Toll-like receptor 2 in gut epithelial cells and macrophages of goldfish Carassius auratus fed with a high-cholesterol diet. FISH & SHELLFISH IMMUNOLOGY 2016; 59:250-255. [PMID: 27818343 DOI: 10.1016/j.fsi.2016.11.003] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 10/28/2016] [Accepted: 11/02/2016] [Indexed: 06/06/2023]
Abstract
Toll-like receptors (TLRs) are a group of pattern recognition molecules that play a crucial role in innate immunity. The structural conservation of the archaic TLR system suggests that the regulation of the immune response might be similar in fish and mammals. Several TLRs (TLR-1, -2, and -4) are expressed by activated macrophages, "foam cells" in human atherosclerotic lesions. To date, 20 different TLRs were identified in more than a dozen different fish species. In this study we found that feeding goldfish, Carrassius auratus, a high-cholesterol diet (HCD) resulted macrophage foam cell formation in the intestinal tissues. The expression of TLR2 has been found in foam cells and in the cytoplasm of enterocytes, however the staining was more intense at the apical surface of polarized intestinal epithelial cells and in the lamina propria. In the intestinal epithelial cells and in the lamina propria cells of the control fish the TLR2 was expressed at low levels. The intestinal epithelium is directly involved in the mucosal immune response through its expression of proinflammatory genes, release of inflammatory cytokines, and recruitment of inflammatory cells.
Collapse
Affiliation(s)
- E R Lauriano
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno d'Alcontres 31, 98166 Messina, Italy
| | - S Pergolizzi
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno d'Alcontres 31, 98166 Messina, Italy
| | - G Capillo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno d'Alcontres 31, 98166 Messina, Italy
| | - M Kuciel
- Department of Comparative Anatomy, Institute of Zoology, Jagiellonian University, Gronostajowa 9, 30-387 Krakow, Poland
| | - A Alesci
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno d'Alcontres 31, 98166 Messina, Italy
| | - C Faggio
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno d'Alcontres 31, 98166 Messina, Italy.
| |
Collapse
|
29
|
Zhu L, Fang L. AIBP: A Novel Molecule at the Interface of Cholesterol Transport, Angiogenesis, and Atherosclerosis. Methodist Debakey Cardiovasc J 2016; 11:160-5. [PMID: 26634023 DOI: 10.14797/mdcj-11-3-160] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Cardiovascular disease, which is often driven by hypercholesterolemia and subsequent coronary atherosclerosis, is the number-one cause of morbidity and mortality in the United States. Based on long-term epidemiological studies, high-density lipoprotein cholesterol (HDL-C) levels are inversely correlated with risk for coronary artery disease (CAD). HDL-mediated reverse cholesterol transport (RCT) is responsible for cholesterol removal from the peripheral tissues and return to the liver for final elimination.1 In atherosclerosis, intraplaque angiogenesis promotes plaque growth and increases plaque vulnerability. Conceivably, the acceleration of RCT and disruption of intraplaque angiogenesis would inhibit atherosclerosis and reduce CAD. We have identified a protein called apoA-I binding protein (AIBP) that augments HDL functionality by accelerating cholesterol efflux. Furthermore, AIBP inhibits vascular endothelial growth factor receptor 2 activation in endothelial cells and limits angiogenesis.2 The following discusses the prospect of using AIBP as a novel therapeutic approach for the treatment of CAD.
Collapse
Affiliation(s)
- Laurence Zhu
- Houston Methodist Research Institute, Houston Methodist Hospital, Houston, Texas
| | - Longhou Fang
- Houston Methodist Research Institute, Houston Methodist Hospital, Houston, Texas
| |
Collapse
|
30
|
Dennis EA. Liberating Chiral Lipid Mediators, Inflammatory Enzymes, and LIPID MAPS from Biological Grease. J Biol Chem 2016; 291:24431-24448. [PMID: 27555328 DOI: 10.1074/jbc.x116.723791] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
In 1970, it was well accepted that the central role of lipids was in energy storage and metabolism, and it was assumed that amphipathic lipids simply served a passive structural role as the backbone of biological membranes. As a result, the scientific community was focused on nucleic acids, proteins, and carbohydrates as information-containing molecules. It took considerable effort until scientists accepted that lipids also "encode" specific and unique biological information and play a central role in cell signaling. Along with this realization came the recognition that the enzymes that act on lipid substrates residing in or on membranes and micelles must also have important signaling roles, spurring curiosity into their potentially unique modes of action differing from those acting on water-soluble substrates. This led to the creation of the concept of "surface dilution kinetics" for describing the mechanism of enzymes acting on lipid substrates, as well as the demonstration that lipid enzymes such as phospholipase A2 (PLA2) contain allosteric activator sites for specific phospholipids as well as for membranes. As our understanding of phospholipases advanced, so did the understanding that many of the lipids released by these enzymes are chiral information-containing signaling molecules; for example, PLA2 regulates the generation of precursors for the biosynthesis of eicosanoids and other bioactive lipid mediators of inflammation and resolution underlying disease progression. The creation of the LIPID MAPS initiative in 2003 and the ensuing development of the lipidomics field have revealed that lipid metabolites are central to human metabolism. Today lipids are recognized as key mediators of health and disease as we enter a new era of biomarkers and personalized medicine. This article is my personal "reflection" on these scientific advances.
Collapse
Affiliation(s)
- Edward A Dennis
- From the Department of Chemistry and Biochemistry and Department of Pharmacology, School of Medicine, University of California at San Diego, La Jolla, California 92093-0601.
| |
Collapse
|
31
|
Freshwater clam extracts alleviate dyslipidaemia of tilapia fed a high-fat diet as an animal model. J Funct Foods 2016. [DOI: 10.1016/j.jff.2016.06.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
|
32
|
Chattopadhyay A, Navab M, Hough G, Grijalva V, Mukherjee P, Fogelman HR, Hwang LH, Faull KF, Lusis AJ, Reddy ST, Fogelman AM. Tg6F ameliorates the increase in oxidized phospholipids in the jejunum of mice fed unsaturated LysoPC or WD. J Lipid Res 2016; 57:832-47. [PMID: 26965826 DOI: 10.1194/jlr.m064352] [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] [Received: 10/07/2015] [Indexed: 12/13/2022] Open
Abstract
Mouse chow supplemented with lysophosphatidylcholine with oleic acid at sn-1 and a hydroxyl group at sn-2 (LysoPC 18:1) increased LysoPC 18:1 in tissue of the jejunum of LDL receptor (LDLR)-null mice by 8.9 ± 1.7-fold compared with chow alone. Western diet (WD) contained dramatically less phosphatidylcholine 18:1 or LysoPC 18:1 compared with chow, but feeding WD increased LysoPC 18:1 in the jejunum by 7.5 ± 1.4-fold compared with chow. Feeding LysoPC 18:1 or feeding WD increased oxidized phospholipids in the jejunum by 5.2 ± 3.0-fold or 8.6 ± 2.2-fold, respectively, in LDLR-null mice (P < 0.0004), and 2.6 ± 1.5-fold or 2.4 ± 0.92-fold, respectively, in WT C57BL/6J mice (P < 0.0001). Adding 0.06% by weight of a concentrate of transgenic tomatoes expressing the 6F peptide (Tg6F) decreased LysoPC 18:1 in the jejunum of LDLR-null mice on both diets (P < 0.0001), and prevented the increase in oxidized phospholipids in the jejunum in LDLR-null and WT mice on both diets (P < 0.008). Tg6F decreased inflammatory cells in the villi of the jejunum, decreased dyslipidemia, and decreased systemic inflammation in LDLR-null and WT mice on both diets. We conclude that Tg6F reduces diet-induced inflammation by reducing the content of unsaturated LysoPC and oxidized phospholipids in the jejunum of mice.
Collapse
Affiliation(s)
- Arnab Chattopadhyay
- Departments of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095-1736
| | - Mohamad Navab
- Departments of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095-1736
| | - Greg Hough
- Departments of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095-1736
| | - Victor Grijalva
- Departments of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095-1736
| | - Pallavi Mukherjee
- Departments of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095-1736
| | - Hannah R Fogelman
- Departments of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095-1736
| | - Lin H Hwang
- Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095-1736
| | - Kym F Faull
- Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095-1736
| | - Aldons J Lusis
- Departments of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095-1736 Human Genetics, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095-1736 Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095-1736
| | - Srinivasa T Reddy
- Departments of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095-1736 Molecular and Medical Pharmacology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095-1736 Obstetrics and Gynecology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095-1736
| | - Alan M Fogelman
- Departments of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095-1736
| |
Collapse
|
33
|
Choi SH, Kim J, Gonen A, Viriyakosol S, Miller YI. MD-2 binds cholesterol. Biochem Biophys Res Commun 2016; 470:877-80. [PMID: 26806306 DOI: 10.1016/j.bbrc.2016.01.126] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 01/21/2016] [Indexed: 12/22/2022]
Abstract
Cholesterol is a structural component of cellular membranes, which is transported from liver to peripheral cells in the form of cholesterol esters (CE), residing in the hydrophobic core of low-density lipoprotein. Oxidized CE (OxCE) is often found in plasma and in atherosclerotic lesions of subjects with cardiovascular disease. Our earlier studies have demonstrated that OxCE activates inflammatory responses in macrophages via toll-like receptor-4 (TLR4). Here we demonstrate that cholesterol binds to myeloid differentiation-2 (MD-2), a TLR4 ancillary molecule, which is a binding receptor for bacterial lipopolysaccharide (LPS) and is indispensable for LPS-induced TLR4 dimerization and signaling. Cholesterol binding to MD-2 was competed by LPS and by OxCE-modified BSA. Furthermore, soluble MD-2 in human plasma and MD-2 in mouse atherosclerotic lesions carried cholesterol, the finding supporting the biological significance of MD-2 cholesterol binding. These results help understand the molecular basis of TLR4 activation by OxCE and mechanisms of chronic inflammation in atherosclerosis.
Collapse
Affiliation(s)
- Soo-Ho Choi
- Department of Medicine, University of California, 9500 Gilman Drive, La Jolla, San Diego, CA 92093, USA
| | - Jungsu Kim
- Department of Medicine, University of California, 9500 Gilman Drive, La Jolla, San Diego, CA 92093, USA
| | - Ayelet Gonen
- Department of Medicine, University of California, 9500 Gilman Drive, La Jolla, San Diego, CA 92093, USA
| | - Suganya Viriyakosol
- Department of Medicine, University of California, 9500 Gilman Drive, La Jolla, San Diego, CA 92093, USA
| | - Yury I Miller
- Department of Medicine, University of California, 9500 Gilman Drive, La Jolla, San Diego, CA 92093, USA.
| |
Collapse
|
34
|
Schlegel A. Zebrafish Models for Dyslipidemia and Atherosclerosis Research. Front Endocrinol (Lausanne) 2016; 7:159. [PMID: 28018294 PMCID: PMC5159437 DOI: 10.3389/fendo.2016.00159] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 12/02/2016] [Indexed: 11/30/2022] Open
Abstract
Atherosclerotic cardiovascular disease is the leading cause of death. Elevated circulating concentrations of lipids are a central pathogenetic driver of atherosclerosis. While numerous effective therapies for this condition have been developed, there is substantial unmet need for this pandemic illness. Here, I will review nutritional, physiological, genetic, and pathological discoveries in the emerging zebrafish model for studying dyslipidemia and atherosclerosis. The technical and physiological advantages and the pharmacological potential of this organism for discovery and validation of dyslipidemia and atherosclerosis targets are stressed through summary of recent findings. An emerging literature shows that zebrafish, through retention of a cetp ortholog gene and high sensitivity to ingestion of excess cholesterol, rapidly develops hypercholesterolemia, with a pattern of distribution of lipid species in lipoprotein particles similar to humans. Furthermore, recent studies leveraging the optical transparency of zebrafish larvae to monitor the fate of these ingested lipids have provided exciting insights to the development of dyslipidemia and atherosclerosis. Future directions for investigation are considered, with particular attention to the potential for in vivo cell biological study of atherosclerotic plaques.
Collapse
Affiliation(s)
- Amnon Schlegel
- University of Utah Molecular Medicine Program, School of Medicine, University of Utah, Salt Lake City, UT, USA
- Department of Internal Medicine, Division of Endocrinology, Metabolism and Diabetes, University of Utah, Salt Lake City, UT, USA
- Department of Biochemistry, School of Medicine, University of Utah, Salt Lake City, UT, USA
- Department of Nutrition and Integrative Physiology, College of Health, University of Utah, Salt Lake City, UT, USA
- *Correspondence: Amnon Schlegel,
| |
Collapse
|
35
|
Seto SW, Kiat H, Lee SMY, Bensoussan A, Sun YT, Hoi MPM, Chang D. Zebrafish models of cardiovascular diseases and their applications in herbal medicine research. Eur J Pharmacol 2015; 768:77-86. [PMID: 26494630 DOI: 10.1016/j.ejphar.2015.10.031] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 10/02/2015] [Accepted: 10/16/2015] [Indexed: 01/12/2023]
Abstract
The zebrafish (Danio rerio) has recently become a powerful animal model for cardiovascular research and drug discovery due to its ease of maintenance, genetic manipulability and ability for high-throughput screening. Recent advances in imaging techniques and generation of transgenic zebrafish have greatly facilitated in vivo analysis of cellular events of cardiovascular development and pathogenesis. More importantly, recent studies have demonstrated the functional similarity of drug metabolism systems between zebrafish and humans, highlighting the clinical relevance of employing zebrafish in identifying lead compounds in Chinese herbal medicine with potential beneficial cardiovascular effects. This paper seeks to summarise the scope of zebrafish models employed in cardiovascular studies and the application of these research models in Chinese herbal medicine to date.
Collapse
Affiliation(s)
- Sai-Wang Seto
- National Institute of Complementary Medicine, Western Sydney University, Campbelltown, NSW, Australia
| | - Hosen Kiat
- Faculty of Medicine, University of New South Wales, NSW, Australia; School of Medicine, Western Sydney University, Locked Bag 1797, Penrith, NSW, Australia; Faculty of Medicine and Health Sciences, Macquarie University, NSW, Australia
| | - Simon M Y Lee
- State Key Laboratory Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Alan Bensoussan
- National Institute of Complementary Medicine, Western Sydney University, Campbelltown, NSW, Australia
| | - Yu-Ting Sun
- National Institute of Complementary Medicine, Western Sydney University, Campbelltown, NSW, Australia
| | - Maggie P M Hoi
- State Key Laboratory Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Dennis Chang
- National Institute of Complementary Medicine, Western Sydney University, Campbelltown, NSW, Australia.
| |
Collapse
|
36
|
Abstract
Initiation and progression of atherosclerosis depend on local inflammation and accumulation of lipids in the vascular wall. Although many cells are involved in the development and progression of atherosclerosis, macrophages are fundamental contributors. For nearly a decade, the phenotypic heterogeneity and plasticity of macrophages has been studied. In atherosclerotic lesions, macrophages are submitted to a large variety of micro-environmental signals, such as oxidized lipids and cytokines, which influence the phenotypic polarization and activation of macrophages resulting in a dynamic plasticity. The macrophage phenotype spectrum is characterized, at the extremes, by the classical M1 macrophages induced by T-helper 1 (Th-1) cytokines and by the alternative M2 macrophages induced by Th-2 cytokines. M2 macrophages can be further classified into M2a, M2b, M2c, and M2d subtypes. More recently, additional plaque-specific macrophage phenotypes have been identified, termed as Mox, Mhem, and M4. Understanding the mechanisms and functional consequences of the phenotypic heterogeneity of macrophages will contribute to determine their potential role in lesion development and plaque stability. Furthermore, research on macrophage plasticity could lead to novel therapeutic approaches to counteract cardiovascular diseases such as atherosclerosis. The present review summarizes our current knowledge on macrophage subsets in atherosclerotic plaques and mechanism behind the modulation of the macrophage phenotype.
Collapse
Affiliation(s)
- Sophie Colin
- Université Lille 2, Lille, France; Inserm, U1011, Lille, France; Institut Pasteur de Lille, Lille, France; European Genomic Institute for Diabetes (EGID), FR 3508, Lille, France
| | | | | |
Collapse
|
37
|
Astarita G, Kendall AC, Dennis EA, Nicolaou A. Targeted lipidomic strategies for oxygenated metabolites of polyunsaturated fatty acids. BIOCHIMICA ET BIOPHYSICA ACTA 2015; 1851:456-68. [PMID: 25486530 PMCID: PMC4323855 DOI: 10.1016/j.bbalip.2014.11.012] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Revised: 11/19/2014] [Accepted: 11/26/2014] [Indexed: 12/13/2022]
Abstract
Oxidation of polyunsaturated fatty acids (PUFA) through enzymatic or non-enzymatic free radical-mediated reactions can yield an array of lipid metabolites including eicosanoids, octadecanoids, docosanoids and related species. In mammals, these oxygenated PUFA mediators play prominent roles in the physiological and pathological regulation of many key biological processes in the cardiovascular, renal, reproductive and other systems including their pivotal contribution to inflammation. Mass spectrometry-based technology platforms have revolutionized our ability to analyze the complex mixture of lipid mediators found in biological samples, with increased numbers of metabolites that can be simultaneously quantified from a single sample in few analytical steps. The recent development of high-sensitivity and high-throughput analytical tools for lipid mediators affords a broader view of these oxygenated PUFA species, and facilitates research into their role in health and disease. In this review, we illustrate current analytical approaches for a high-throughput lipidomic analysis of eicosanoids and related mediators in biological samples. This article is part of a Special Issue entitled "Oxygenated metabolism of PUFA: analysis and biological relevance."
Collapse
Affiliation(s)
- Giuseppe Astarita
- Waters Corporation, Milford, MA, USA; Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC, USA.
| | - Alexandra C Kendall
- Manchester Pharmacy School, Faculty of Medical and Human Sciences, The University of Manchester, Stopford Building, Oxford Road, Manchester M13 9PT, UK
| | - Edward A Dennis
- Department of Chemistry/Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0601, USA; Department of Pharmacology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0601, USA
| | - Anna Nicolaou
- Manchester Pharmacy School, Faculty of Medical and Human Sciences, The University of Manchester, Stopford Building, Oxford Road, Manchester M13 9PT, UK.
| |
Collapse
|
38
|
Zeb A. Chemistry and liquid chromatography methods for the analyses of primary oxidation products of triacylglycerols. Free Radic Res 2015; 49:549-64. [PMID: 25824968 DOI: 10.3109/10715762.2015.1022540] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Triacylglycerols (TAGs) are one of the major components of the cells in higher biological systems, which can act as an energy reservoir in the living cells. The unsaturated fatty acid moiety is the key site of oxidation and formation of oxidation compounds. The TAG free radical generates several primary oxidation compounds. These include hydroperoxides, hydroxides, epidioxides, hydroperoxy epidioxides, hydroxyl epidioxides, and epoxides. The presence of these oxidized TAGs in the cell increases the chances of several detrimental processes. For this purpose, several liquid chromatography (LC) methods were reported in their analyses. This review is therefore focused on the chemistry, oxidation, extraction, and the LC methods reported in the analyses of oxidized TAGs. The studies on thin-layer chromatography were mostly focused on the total oxidized TAGs separation and employ hexane as major solvent. High-performance LC (HPLC) methods were discussed in details along with their merits and demerits. It was found that most of the HPLC methods employed isocratic elution with methanol and acetonitrile as major solvents with an ultraviolet detector. The coupling of HPLC with mass spectrometry (MS) highly increases the efficiency of analysis as well as enables reliable structural elucidation. The use of MS was found to be helpful in studying the oxidation chemistry of TAGs and needs to be extended to the complex biological systems.
Collapse
Affiliation(s)
- A Zeb
- Department of Biotechnology, University of Malakand , Chakdara , Pakistan
| |
Collapse
|
39
|
Study on the deformation of endothelial cells using a bio-inspired in vitro disease model. Microvasc Res 2015; 98:172-82. [DOI: 10.1016/j.mvr.2014.02.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Revised: 12/12/2013] [Accepted: 02/03/2014] [Indexed: 01/12/2023]
|
40
|
Paglia G, Angel P, Williams JP, Richardson K, Olivos HJ, Thompson JW, Menikarachchi L, Lai S, Walsh C, Moseley A, Plumb RS, Grant D, Palsson BO, Langridge J, Geromanos S, Astarita G. Ion mobility-derived collision cross section as an additional measure for lipid fingerprinting and identification. Anal Chem 2015; 87:1137-44. [PMID: 25495617 PMCID: PMC4302848 DOI: 10.1021/ac503715v] [Citation(s) in RCA: 225] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2014] [Accepted: 12/13/2014] [Indexed: 02/07/2023]
Abstract
Despite recent advances in analytical and computational chemistry, lipid identification remains a significant challenge in lipidomics. Ion-mobility spectrometry provides an accurate measure of the molecules' rotationally averaged collision cross-section (CCS) in the gas phase and is thus related to ionic shape. Here, we investigate the use of CCS as a highly specific molecular descriptor for identifying lipids in biological samples. Using traveling wave ion mobility mass spectrometry (MS), we measured the CCS values of over 200 lipids within multiple chemical classes. CCS values derived from ion mobility were not affected by instrument settings or chromatographic conditions, and they were highly reproducible on instruments located in independent laboratories (interlaboratory RSD < 3% for 98% of molecules). CCS values were used as additional molecular descriptors to identify brain lipids using a variety of traditional lipidomic approaches. The addition of CCS improved the reproducibility of analysis in a liquid chromatography-MS workflow and maximized the separation of isobaric species and the signal-to-noise ratio in direct-MS analyses (e.g., "shotgun" lipidomics and MS imaging). These results indicate that adding CCS to databases and lipidomics workflows increases the specificity and selectivity of analysis, thus improving the confidence in lipid identification compared to traditional analytical approaches. The CCS/accurate-mass database described here is made publicly available.
Collapse
Affiliation(s)
- Giuseppe Paglia
- Istituto
Zooprofilattico Sperimentale della Puglia e Della Basilicata, Foggia, Italy
- Center
for Systems Biology, University of Iceland, Reykjavik, Iceland
| | - Peggi Angel
- Protea
Biosciences Group, Inc., Morgantown, West Virginia 26505, United States
| | | | | | | | - J. Will Thompson
- Duke
Proteomics Core Facility, Durham, North Carolina 27708, United States
| | - Lochana Menikarachchi
- Department
of Pharmaceutical Sciences, University of
Connecticut, Storrs, Connecticut 06268, United States
| | - Steven Lai
- Waters
Corporation, Milford, Massachusetts 01757, United States
| | - Callee Walsh
- Protea
Biosciences Group, Inc., Morgantown, West Virginia 26505, United States
| | - Arthur Moseley
- Duke
Proteomics Core Facility, Durham, North Carolina 27708, United States
| | - Robert S. Plumb
- Waters
Corporation, Milford, Massachusetts 01757, United States
- Computational
and Systems Medicine, Department of Surgery and Cancer, Faculty of
Medicine, Imperial College London, London, United Kingdom
| | - David
F. Grant
- Department
of Pharmaceutical Sciences, University of
Connecticut, Storrs, Connecticut 06268, United States
| | - Bernhard O. Palsson
- Computational
and Systems Medicine, Department of Surgery and Cancer, Faculty of
Medicine, Imperial College London, London, United Kingdom
| | - James Langridge
- Waters
Corporation, Milford, Massachusetts 01757, United States
| | - Scott Geromanos
- Waters
Corporation, Milford, Massachusetts 01757, United States
| | - Giuseppe Astarita
- Waters
Corporation, Milford, Massachusetts 01757, United States
- Department
of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC 20057, United States
| |
Collapse
|
41
|
Schlegel A, Gut P. Metabolic insights from zebrafish genetics, physiology, and chemical biology. Cell Mol Life Sci 2015; 72:2249-60. [PMID: 25556679 PMCID: PMC4439526 DOI: 10.1007/s00018-014-1816-8] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 12/15/2014] [Accepted: 12/19/2014] [Indexed: 01/01/2023]
Abstract
Metabolic diseases—atherosclerotic cardiovascular disease, type 2 diabetes mellitus, obesity, and non-alcoholic fatty liver disease––have reached pandemic proportions. Across gene, cell, organ, organism, and social-environmental scales, fundamental discoveries of the derangements that occur in these diseases are required to develop effective new treatments. Here we will review genetic, physiological, pathological and chemical biological discoveries in the emerging zebrafish model for studying metabolism and metabolic diseases. We present a synthesis of recent studies using forward and reverse genetic tools to make new contributions to our understanding of lipid trafficking, diabetes pathogenesis and complications, and to β-cell biology. The technical and physiological advantages and the pharmacological potential of this organism for discovery and validation of metabolic disease targets are stressed by our summary of recent findings. We conclude by arguing that metabolic research using zebrafish will benefit from adoption of conventional blood and tissue metabolite measurements, employment of modern imaging techniques, and development of more rigorous metabolic flux methods.
Collapse
Affiliation(s)
- Amnon Schlegel
- University of Utah Molecular Medicine Program, School of Medicine, University of Utah, 15 North 2030 East, Room 3240B, Salt Lake City, UT, 84112, USA,
| | | |
Collapse
|
42
|
Abstract
Macrophage accumulation within the vascular wall is a hallmark of atherosclerosis. In atherosclerotic lesions, macrophages respond to various environmental stimuli, such as modified lipids, cytokines, and senescent erythrocytes, which can modify their functional phenotypes. The results of studies on human atherosclerotic plaques demonstrate that the relative proportions of macrophage subsets within a plaque might be a better indicator of plaque phenotype and stability than the total number of macrophages. Understanding the function of specific macrophage subsets and their contribution to the composition and growth of atherosclerotic plaques would aid the identification of novel strategies to delay or halt the development of the disease and its associated pathophysiological consequences. However, most studies aimed at characterizing the phenotypes of human macrophages are performed in vitro and, therefore, their functional relevance to human pathology remains uncertain. In this Review, the diverse range of macrophage phenotypes in atherosclerotic lesions and their potential roles in both plaque progression and stability are discussed, with an emphasis on human pathology.
Collapse
Affiliation(s)
- Giulia Chinetti-Gbaguidi
- INSERM U1011, Institut Pasteur de Lille, 1, Rue du Professeur Calmette, BP 245, Lille 59019, France
| | - Sophie Colin
- INSERM U1011, Institut Pasteur de Lille, 1, Rue du Professeur Calmette, BP 245, Lille 59019, France
| | - Bart Staels
- INSERM U1011, Institut Pasteur de Lille, 1, Rue du Professeur Calmette, BP 245, Lille 59019, France
| |
Collapse
|
43
|
O'Hare EA, Wang X, Montasser ME, Chang YPC, Mitchell BD, Zaghloul NA. Disruption of ldlr causes increased LDL-c and vascular lipid accumulation in a zebrafish model of hypercholesterolemia. J Lipid Res 2014; 55:2242-53. [PMID: 25201834 DOI: 10.1194/jlr.m046540] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Hyperlipidemia and arterial cholesterol accumulation are primary causes of cardiovascular events. Monogenic forms of hyperlipidemia and recent genome-wide association studies indicate that genetics plays an important role. Zebrafish are a useful model for studying the genetic susceptibility to hyperlipidemia owing to conservation of many components of lipoprotein metabolism, including those related to LDL, ease of genetic manipulation, and in vivo observation of lipid transport and vascular calcification. We sought to develop a genetic model for lipid metabolism in zebrafish, capitalizing on one well-understood player in LDL cholesterol (LDL-c) transport, the LDL receptor (ldlr), and an established in vivo model of hypercholesterolemia. We report that morpholinos targeted against the gene encoding ldlr effectively suppressed its expression in embryos during the first 8 days of development. The ldlr morphants exhibited increased LDL-c levels that were exacerbated by feeding a high cholesterol diet. Increased LDL-c was ameliorated in morphants upon treatment with atorvastatin. Furthermore, we observed significant vascular and liver lipid accumulation, vascular leakage, and plaque oxidation in ldlr-deficient embryos. Finally, upon transcript analysis of several cholesterol-regulating genes, we observed changes similar to those seen in mammalian systems, suggesting that cholesterol regulation may be conserved in zebrafish. Taken together, these observations indicate conservation of ldlr function in zebrafish and demonstrate the utility of transient gene knockdown in embryos as a genetic model for hyperlipidemia.
Collapse
Affiliation(s)
- Elizabeth A O'Hare
- Department of Medicine, Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, MD
| | - Xiaochun Wang
- Department of Medicine, Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, MD
| | - May E Montasser
- Department of Medicine, Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, MD
| | - Yen-Pei C Chang
- Department of Medicine, Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, MD
| | - Braxton D Mitchell
- Department of Medicine, Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, MD
| | - Norann A Zaghloul
- Department of Medicine, Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, MD
| |
Collapse
|
44
|
Cruz-Garcia L, Schlegel A. Lxr-driven enterocyte lipid droplet formation delays transport of ingested lipids. J Lipid Res 2014; 55:1944-58. [PMID: 25030662 DOI: 10.1194/jlr.m052845] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Liver X receptors (Lxrs) are master regulators of cholesterol catabolism, driving the elimination of cholesterol from the periphery to the lumen of the intestine. Development of pharmacological agents to activate Lxrs has been hindered by synthetic Lxr agonists' induction of hepatic lipogenesis and hypertriglyceridemia. Elucidating the function of Lxrs in regulating enterocyte lipid handling might identify novel aspects of lipid metabolism that are pharmacologically amenable. We took a genetic approach centered on the single Lxr gene nr1h3 in zebrafish to study the role of Lxr in enterocyte lipid metabolism. Loss of nr1h3 function causes anticipated gene regulatory changes and cholesterol intolerance, collectively reflecting high evolutionary conservation of zebrafish Lxra function. Intestinal nr1h3 activation delays transport of absorbed neutral lipids, with accumulation of neutral lipids in enterocyte cytoplasmic droplets. This delay in transport of ingested neutral lipids protects animals from hypercholesterolemia and hepatic steatosis induced by a high-fat diet. On a gene regulatory level, Lxra induces expression of acsl3a, which encodes acyl-CoA synthetase long-chain family member 3a, a lipid droplet-anchored protein that directs fatty acyl chains into lipids. Forced overexpression of acls3a in enterocytes delays, in part, the appearance of neutral lipids in the vasculature of zebrafish larvae. Activation of Lxr in the intestine cell-autonomously regulates the rate of delivery of absorbed lipids by inducting a temporary lipid intestinal droplet storage depot.
Collapse
Affiliation(s)
- Lourdes Cruz-Garcia
- University of Utah Molecular Medicine (U2M2) Program,University of Utah, Salt Lake City, UT 84112 Department of Internal Medicine, Division of Endocrinology, Metabolism, and Diabetes,University of Utah, Salt Lake City, UT 84112
| | - Amnon Schlegel
- University of Utah Molecular Medicine (U2M2) Program,University of Utah, Salt Lake City, UT 84112 Department of Internal Medicine, Division of Endocrinology, Metabolism, and Diabetes,University of Utah, Salt Lake City, UT 84112 Department of Biochemistry, School of Medicine, University of Utah, Salt Lake City, UT 84112
| |
Collapse
|
45
|
Davies SS, Guo L. Lipid peroxidation generates biologically active phospholipids including oxidatively N-modified phospholipids. Chem Phys Lipids 2014; 181:1-33. [PMID: 24704586 DOI: 10.1016/j.chemphyslip.2014.03.002] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Revised: 02/28/2014] [Accepted: 03/18/2014] [Indexed: 12/25/2022]
Abstract
Peroxidation of membranes and lipoproteins converts "inert" phospholipids into a plethora of oxidatively modified phospholipids (oxPL) that can act as signaling molecules. In this review, we will discuss four major classes of oxPL: mildly oxygenated phospholipids, phospholipids with oxidatively truncated acyl chains, phospholipids with cyclized acyl chains, and phospholipids that have been oxidatively N-modified on their headgroups by reactive lipid species. For each class of oxPL we will review the chemical mechanisms of their formation, the evidence for their formation in biological samples, the biological activities and signaling pathways associated with them, and the catabolic pathways for their elimination. We will end by briefly highlighting some of the critical questions that remain about the role of oxPL in physiology and disease.
Collapse
Affiliation(s)
- Sean S Davies
- Division of Clinical Pharmacology, Department of Pharmacology, Vanderbilt University, United States.
| | - Lilu Guo
- Division of Clinical Pharmacology, Department of Pharmacology, Vanderbilt University, United States
| |
Collapse
|
46
|
Ravandi A, Leibundgut G, Hung MY, Patel M, Hutchins PM, Murphy RC, Prasad A, Mahmud E, Miller YI, Dennis EA, Witztum JL, Tsimikas S. Release and capture of bioactive oxidized phospholipids and oxidized cholesteryl esters during percutaneous coronary and peripheral arterial interventions in humans. J Am Coll Cardiol 2014; 63:1961-71. [PMID: 24613321 DOI: 10.1016/j.jacc.2014.01.055] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Revised: 01/27/2014] [Accepted: 01/29/2014] [Indexed: 12/20/2022]
Abstract
OBJECTIVES This study sought to assess whether oxidized lipids are released downstream from obstructive plaques after percutaneous coronary and peripheral interventions using distal protection devices. BACKGROUND Oxidation of lipoproteins generates multiple bioactive oxidized lipids that affect atherothrombosis and endothelial function. Direct evidence of their role during therapeutic procedures, which may result in no-reflow phenomenon, myocardial infarction, and stroke, is lacking. METHODS The presence of specific oxidized lipids was assessed in embolized material captured by distal protection filter devices during uncomplicated saphenous vein graft, carotid, renal, and superficial femoral artery interventions. The presence of oxidized phospholipids (OxPL) and oxidized cholesteryl esters (OxCE) was evaluated in 24 filters using liquid chromatography, tandem mass spectrometry, enzyme-linked immunosorbent assays, and immunostaining. RESULTS Phosphatidylcholine-containing OxPL, including (1-palmitoyl-2-[9-oxo-nonanoyl] PC), representing a major phosphatidylcholine-OxPL molecule quantitated within plaque material, [1-palmitoyl-2-(5-oxo-valeroyl)-sn-glycero-3-phosphocholine], and 1-palmitoyl-2-glutaroyl-sn-glycero-3-phosphocholine, were identified in the extracted lipid portion from all vascular beds. Several species of OxCE, such as keto, hydroperoxide, hydroxy, and epoxy cholesteryl ester derivatives from cholesteryl linoleate and cholesteryl arachidonate, were also present. The presence of OxPL was confirmed using enzyme-linked immunoassays and immunohistochemistry of captured material. CONCLUSIONS This study documents the direct release and capture of OxPL and OxCE during percutaneous interventions from multiple arterial beds in humans. Entrance of bioactive oxidized lipids into the microcirculation may mediate adverse clinical outcomes during therapeutic procedures.
Collapse
Affiliation(s)
- Amir Ravandi
- St. Boniface Hospital Research Centre, University of Manitoba, Winnipeg, Manitoba, Canada; Department of Medicine, University of California-San Diego, La Jolla, California
| | - Gregor Leibundgut
- Department of Medicine, University of California-San Diego, La Jolla, California; University of Basel, Basel, Switzerland
| | - Ming-Yow Hung
- Department of Medicine, University of California-San Diego, La Jolla, California; Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Division of Cardiology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Mitul Patel
- Department of Medicine, University of California-San Diego, La Jolla, California
| | - Patrick M Hutchins
- Department of Pharmacology, University of Colorado Denver, Aurora, Colorado
| | - Robert C Murphy
- Department of Pharmacology, University of Colorado Denver, Aurora, Colorado
| | - Anand Prasad
- Department of Medicine, University of California-San Diego, La Jolla, California; Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Ehtisham Mahmud
- Department of Medicine, University of California-San Diego, La Jolla, California
| | - Yury I Miller
- Department of Medicine, University of California-San Diego, La Jolla, California
| | - Edward A Dennis
- Department of Pharmacology and Chemistry and Biochemistry, University of California, La Jolla, California
| | - Joseph L Witztum
- Department of Medicine, University of California-San Diego, La Jolla, California
| | - Sotirios Tsimikas
- Department of Medicine, University of California-San Diego, La Jolla, California.
| |
Collapse
|
47
|
Fang L, Liu C, Miller YI. Zebrafish models of dyslipidemia: relevance to atherosclerosis and angiogenesis. Transl Res 2014; 163:99-108. [PMID: 24095954 PMCID: PMC3946603 DOI: 10.1016/j.trsl.2013.09.004] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Revised: 09/07/2013] [Accepted: 09/10/2013] [Indexed: 01/07/2023]
Abstract
Lipid and lipoprotein metabolism in zebrafish and in humans are remarkably similar. Zebrafish express all major nuclear receptors, lipid transporters, apolipoproteins and enzymes involved in lipoprotein metabolism. Unlike mice, zebrafish express cetp and the Cetp activity is detected in zebrafish plasma. Feeding zebrafish a high cholesterol diet, without any genetic intervention, results in significant hypercholesterolemia and robust lipoprotein oxidation, making zebrafish an attractive animal model to study mechanisms relevant to early development of human atherosclerosis. These studies are facilitated by the optical transparency of zebrafish larvae and the availability of transgenic zebrafish expressing fluorescent proteins in endothelial cells and macrophages. Thus, vascular processes can be monitored in live animals. In this review article, we discuss recent advances in using dyslipidemic zebrafish in atherosclerosis-related studies. We also summarize recent work connecting lipid metabolism with regulation of angiogenesis, the work that considerably benefited from using the zebrafish model. These studies uncovered the role of aibp, abca1, abcg1, mtp, apoB, and apoC2 in regulation of angiogenesis in zebrafish and paved the way for future studies in mammals, which may suggest new therapeutic approaches to modulation of excessive or diminished angiogenesis contributing to the pathogenesis of human disease.
Collapse
Affiliation(s)
- Longhou Fang
- Department of Medicine, University of California, San Diego, La Jolla, Calif
| | - Chao Liu
- Department of Medicine, University of California, San Diego, La Jolla, Calif
| | - Yury I Miller
- Department of Medicine, University of California, San Diego, La Jolla, Calif.
| |
Collapse
|
48
|
Choi SH, Yin H, Ravandi A, Armando A, Dumlao D, Kim J, Almazan F, Taylor AM, McNamara CA, Tsimikas S, Dennis EA, Witztum JL, Miller YI. Polyoxygenated cholesterol ester hydroperoxide activates TLR4 and SYK dependent signaling in macrophages. PLoS One 2013; 8:e83145. [PMID: 24376657 PMCID: PMC3871536 DOI: 10.1371/journal.pone.0083145] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Accepted: 10/30/2013] [Indexed: 12/30/2022] Open
Abstract
Oxidation of low-density lipoprotein (LDL) is one of the major causative mechanisms in the development of atherosclerosis. In previous studies, we showed that minimally oxidized LDL (mmLDL) induced inflammatory responses in macrophages, macropinocytosis and intracellular lipid accumulation and that oxidized cholesterol esters (OxCEs) were biologically active components of mmLDL. Here we identified a specific OxCE molecule responsible for the biological activity of mmLDL and characterized signaling pathways in macrophages in response to this OxCE. Using liquid chromatography – tandem mass spectrometry and biological assays, we identified an oxidized cholesteryl arachidonate with bicyclic endoperoxide and hydroperoxide groups (BEP-CE) as a specific OxCE that activates macrophages in a TLR4/MD-2-dependent manner. BEP-CE induced TLR4/MD-2 binding and TLR4 dimerization, phosphorylation of SYK, ERK1/2, JNK and c-Jun, cell spreading and uptake of dextran and native LDL by macrophages. The enhanced macropinocytosis resulted in intracellular lipid accumulation and macrophage foam cell formation. Bone marrow-derived macrophages isolated from TLR4 and SYK knockout mice did not respond to BEP-CE. The presence of BEP-CE was demonstrated in human plasma and in the human plaque material captured in distal protection devices during percutaneous intervention. Our results suggest that BEP-CE is an endogenous ligand that activates the TLR4/SYK signaling pathway. Because BEP-CE is present in human plasma and human atherosclerotic lesions, BEP-CE-induced and TLR4/SYK-mediated macrophage responses may contribute to chronic inflammation in human atherosclerosis.
Collapse
Affiliation(s)
- Soo-Ho Choi
- Department of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Huiyong Yin
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
- Key Laboratory of Food Safety Risk Assessment, Ministry of Health, Beijing, China
- School of Life Science and Technology, Shanghai Tech University, Shanghai, China
| | - Amir Ravandi
- Institute of Cardiovascular Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Aaron Armando
- Department of Pharmacology, University of California San Diego, La Jolla, California, United States of America
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, California, United States of America
| | - Darren Dumlao
- Department of Pharmacology, University of California San Diego, La Jolla, California, United States of America
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, California, United States of America
| | - Jungsu Kim
- Department of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Felicidad Almazan
- Department of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Angela M. Taylor
- Cardiovascular Research Center, Department of Medicine, University of Virginia, Charlottesville, Virginia, United States of America
| | - Coleen A. McNamara
- Cardiovascular Research Center, Department of Medicine, University of Virginia, Charlottesville, Virginia, United States of America
| | - Sotirios Tsimikas
- Department of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Edward A. Dennis
- Department of Pharmacology, University of California San Diego, La Jolla, California, United States of America
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, California, United States of America
| | - Joseph L. Witztum
- Department of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Yury I. Miller
- Department of Medicine, University of California San Diego, La Jolla, California, United States of America
- * E-mail:
| |
Collapse
|
49
|
Wang Z, Mao Y, Cui T, Tang D, Wang XL. Impact of a combined high cholesterol diet and high glucose environment on vasculature. PLoS One 2013; 8:e81485. [PMID: 24349075 PMCID: PMC3857185 DOI: 10.1371/journal.pone.0081485] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Accepted: 10/21/2013] [Indexed: 11/26/2022] Open
Abstract
AIMS Vascular complications are the leading cause of mortality and morbidity in patients with diabetes. However, proper animal models of diabetic vasculopathy that recapitulate the accelerated progression of vascular lesions in human are unavailable. In the present study, we developed a zebrafish model of diabetic vascular complications and the methodology for quantifying vascular lesion formation real-time in the living diabetic zebrafish. METHODS AND RESULTS Wild type zebrafish (AB) and transgenic zebrafish lines of fli1:EGFP, lyz:EGFP, gata1:dsRed, double transgenic zebrafish of gata1:dsRed/fli1:EGFP were exposed to high cholesterol diet and 3% glucose (HCD-HG) for 10 days. The zebrafish model with HCD-HG treatment was characterized by significantly increased tissue levels of insulin, glucagon, glucose, total triglyceride and cholesterol. Confocal microscopic analysis further revealed that the diabetic larvae developed clearly thickened endothelial layers, distinct perivascular lipid depositions, substantial accumulations of inflammatory cells in the injured vasculature, and a decreased velocity of blood flow. Moreover, the vascular abnormalities were improved by the treatment of pioglitazone and metformin. CONCLUSION A combination of high cholesterol diet and high glucose exposure induces a rapid onset of vascular complications in zebrafish similar to the early atherosclerotic vascular injuries in mammalian diabetic models, suggesting that zebrafish may be used as a novel animal model for diabetic vasculopathy.
Collapse
Affiliation(s)
- Zemin Wang
- Shandong University Qilu Hospital Research Center for Cell Therapy, Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital of Shandong University, Jinan, P. R. China
| | - Yun Mao
- Shandong University Qilu Hospital Research Center for Cell Therapy, Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital of Shandong University, Jinan, P. R. China
| | - Taixing Cui
- Shandong University Qilu Hospital Research Center for Cell Therapy, Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital of Shandong University, Jinan, P. R. China
- Department of Cell Biology and Anatomy, South Carolina University, Columbus, South Carolina, United States of America
| | - Dongqi Tang
- Shandong University Qilu Hospital Research Center for Cell Therapy, Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital of Shandong University, Jinan, P. R. China
- Center for Stem Cell and Regenerative Medicine, The Second Hospital of Shandong University, Jinan, P.R. China
| | - Xing Li Wang
- Shandong University Qilu Hospital Research Center for Cell Therapy, Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital of Shandong University, Jinan, P. R. China
| |
Collapse
|
50
|
Meguro S, Hasumura T, Hase T. Coffee polyphenols exert hypocholesterolemic effects in zebrafish fed a high-cholesterol diet. Nutr Metab (Lond) 2013; 10:61. [PMID: 24220226 PMCID: PMC3871761 DOI: 10.1186/1743-7075-10-61] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2013] [Accepted: 09/26/2013] [Indexed: 11/29/2022] Open
Abstract
Background Hypercholesterolemia is an important risk factor for the development of coronary artery disease. Some dietary polyphenols, such as coffee polyphenols (CPPs), reduce cholesterol levels. The mechanism of this cholesterol-lowering effect is not fully understood, although 5-CQA, a major component of CPPs, reportedly inhibits cholesterol biosynthesis. Here, we investigated the mechanism of the cholesterol-lowering effect of CPPs on the basis of cholesterol metabolism–related gene expression in the liver. We also examined the effects of CPPs on vascular lipid accumulation in zebrafish with high cholesterol diet–induced hypercholesterolemia. Methods Over 14 weeks, adult zebrafish were fed a control diet, a high-cholesterol diet, or the latter diet supplemented with CPPs. To measure the extent of vascular lipid accumulation, for 10 days larval zebrafish (which are optically transparent) were fed these same diets with the addition of a fluorescent cholesteryl ester. Results In adult zebrafish, addition of CPPs to a high-cholesterol diet significantly suppressed the increase in plasma and liver cholesterol levels seen when fish ingested the same diet lacking CPPs. Transcription levels of the liver genes hmgcra (encoding 3-hydroxy-3-methylglutaryl-coenzyme A reductase A, a rate-limiting enzyme in cholesterol biosynthesis) and mtp (encoding microsomal triglyceride transfer protein, a lipid transfer protein required for assembly and secretion of lipoproteins) were significantly lower in fish fed the CPP-containing diet than in fish fed the unsupplemented high-cholesterol diet. In contrast, the expression level of the liver gene cyp7a1a (encoding the cytochrome P450 polypeptide 1a of subfamily A of family 7, a rate-limiting enzyme for bile acid biosynthesis) increased significantly upon consumption of the CPP-containing diet. In larval fish, accumulation of fluorescently labeled cholesterol in the caudal artery was greatly reduced on the CPP-containing diet. Conclusions CPP ingestion suppressed cholesterol accumulation in the plasma, liver, and vascular system of zebrafish. Downregulation of cholesterol and lipoprotein synthesis and upregulation of bile acid synthesis in the liver may be the fundamental underlying mechanisms by which CPPs exert their hypocholesterolemic effects. CPP intake may help prevent and manage hypercholesterolemia in humans, and further investigations along these lines using a variety of CPP dose rates are warranted.
Collapse
|