1
|
Pandey AK, Ezewudo E, Hoque N, Pandey AT, Menon S, Simon N, Rasouli B, Habibi E, Sarker SD, Nahar L, Hasan R. A review of food hydrocolloids on cardiovascular health: Alginate, astragalus polysaccharides, carrageenan, fucoidan, lunasin, and psyllium. Int J Biol Macromol 2025; 315:144505. [PMID: 40409656 DOI: 10.1016/j.ijbiomac.2025.144505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2025] [Revised: 05/06/2025] [Accepted: 05/20/2025] [Indexed: 05/25/2025]
Abstract
Food hydrocolloids, derived from natural sources such as plants, algae, and microbes, possess bioactive properties that significantly contribute to cardiovascular health. This review focuses on six key hydrocolloids: alginate, astragalus polysaccharides, carrageenan, fucoidan, lunasin, and psyllium, while also considering other important natural hydrocoloids such as short chain fatty acids (SCFAs), plant-derived food hydrocolloids, plant-derived gums, plant-derived mucilages, pectin, modified citrus pectin, inulin, naringenin, chia seeds, gelatine, whey protein, casein, microbial exopolysaccharides and gums, ulvan, and laminarin. Alginate, from brown seaweed, aids in cardiac tissue regeneration and repair. Astragalus polysaccharides, from the Astragalus plant, provide antioxidant, anti-inflammatory, and immunomodulatory benefits. Carrageenan, sourced from red seaweed, supports lipid profile balance and heart health. Fucoidan, another brown seaweed derivative, offers antihypertensive and lipid-lowering effects. Lunasin, a peptide found in soybeans, oats, and barley, is known for its cholesterol-lowering properties and anti-inflammatory effects. Psyllium, rich in soluble fiber, helps lower LDL cholesterol and improve overall cardiovascular function. These hydrocolloids, along with other mentioned compounds, are utilized in drug formulations, cosmetics, processed foods, and dietary supplements, enhancing food texture and stability while delivering health benefits. Upon consumption, they can be absorbed into the bloodstream or metabolized by gut microbiota into bioactive metabolites. This review examines their effects on cardiovascular function, highlighting their mechanisms in regulating vascular tone, blood pressure, vascular inflammation, and cardiac function. It consolidates current research, emphasizing the potential of these hydrocolloids and related compounds in the prevention and management of cardiovascular diseases (CVDs).
Collapse
Affiliation(s)
- Ajay K Pandey
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA, USA; Augusta University/University of Georgia Medical Partnership, University of Georgia Health Science Campus, Athens, GA, USA
| | - Emmanuella Ezewudo
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA, USA
| | - Nazia Hoque
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA, USA; Department of Pharmacy, East West University, Dhaka, Bangladesh
| | - Anjali T Pandey
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA, USA; Department of Psychology, St. Bonaventure University, St. Bonaventure, NY, USA
| | - Sreelakshmi Menon
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA, USA
| | - Nimi Simon
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA, USA
| | | | - Emran Habibi
- Medicinal Plants Research Centre, Institute of Herbal Medicines and Metabolic Disorders, Mazandaran University of Medical Sciences, Sari, Iran; Centre for Natural Products Discovery, School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool L3 3AF, UK
| | - Satyajit D Sarker
- Centre for Natural Products Discovery, School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool L3 3AF, UK
| | - Lutfun Nahar
- Laboratory of Growth Regulators, Palacký University and Institute of Experimental Botany, The Czech Academy of Sciences, Šlechtitelů 27, Olomouc, Czech Republic.
| | - Raquibul Hasan
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA, USA.
| |
Collapse
|
2
|
Wang X, Shi SR, Sun MM, Zhang XY, Zhang XH, Song SL, Yin F, Guo SD. Mechanisms of action of Fucus vesiculosus-derived fucoidan on improving dyslipidemia in New Zealand rabbits fed a high-fat diet. Int J Biol Macromol 2025; 314:144148. [PMID: 40368205 DOI: 10.1016/j.ijbiomac.2025.144148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 03/30/2025] [Accepted: 05/10/2025] [Indexed: 05/16/2025]
Abstract
Dyslipidemia is a major contributor to various diseases, including atherosclerotic cardiovascular disease and obesity. Treatment strategies for dyslipidemia continue to evolve as our understanding of this metabolic disorder and potential therapeutic candidates advance. Notably, fucoidan demonstrates promising effects in ameliorating dyslipidemia in rodents, although their lipid metabolism differs significantly from humans. This study, investigates the lipid-regulatory effects of Fucus vesiculosus-derived fucoidan (FvF) and elucidates the underlying mechanisms of action using New Zealand rabbits fed a high-fat diet, whose lipid profiles closely resemble those of patients with dyslipidemia. The results demonstrate that FvF intervention ameliorates dyslipidemia and lipid deposition in a dose-dependent manner. Mechanistically, FvF intervention modulates the expression levels of multiple molecules involved in lipid transport, fatty acid synthesis and beta-oxidation, and redox balance, as revealed by quantitative reverse transcription polymerase chain reaction, western blotting, and proteomic analysis. This study is the first to report that FvF, consisting of alternating [→4)-α-L-Fucp(1 → 3)-α-L-Fucp(1→] glycosyls ameliorates dyslipidemia by directly modulating lipid metabolism and indirectly attenuating oxidative stress. These findings suggest that FvF holds significant potential as a candidate for the treatment of lipid disorder-related diseases.
Collapse
Affiliation(s)
- Xue Wang
- Institute of Lipid Metabolism and Atherosclerosis, School of Pharmacy, Shandong Second Medical University, Weifang 261053, China
| | - Shan-Rui Shi
- Institute of Lipid Metabolism and Atherosclerosis, School of Pharmacy, Shandong Second Medical University, Weifang 261053, China
| | - Min-Min Sun
- School of Stomatology, Shandong Second Medical University, Weifang 261053, China
| | - Xue-Ying Zhang
- Institute of Lipid Metabolism and Atherosclerosis, School of Pharmacy, Shandong Second Medical University, Weifang 261053, China
| | - Xu-Hang Zhang
- Institute of Lipid Metabolism and Atherosclerosis, School of Pharmacy, Shandong Second Medical University, Weifang 261053, China
| | - Shi-Lin Song
- Institute of Lipid Metabolism and Atherosclerosis, School of Pharmacy, Shandong Second Medical University, Weifang 261053, China
| | - Fan Yin
- Institute of Lipid Metabolism and Atherosclerosis, School of Pharmacy, Shandong Second Medical University, Weifang 261053, China
| | - Shou-Dong Guo
- Institute of Lipid Metabolism and Atherosclerosis, School of Pharmacy, Shandong Second Medical University, Weifang 261053, China.
| |
Collapse
|
3
|
Feng Q, Ma J, Jiang X, Wei W, Xu D, Cao Y, Pei H. Therapeutic potential of fucoidan in atherosclerosis: a review. Food Funct 2025. [PMID: 40353291 DOI: 10.1039/d4fo05388e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2025]
Abstract
Atherosclerosis (AS) is a slowly progressive disease that significantly increases the risk of cardiovascular diseases. The development of AS is closely associated with various factors, including disturbances in lipid metabolism, endothelial damage, inflammation, and the formation of unstable plaques. AS is strongly linked to diseases with high incidence and mortality, such as ischemic heart disease and stroke, which pose significant economic burdens. Recent studies have focused on identifying effective treatments for preventing and reversing AS. New evidence indicates that fucoidan, a polysaccharide derived from rockweed, possesses lipid-lowering, antioxidant, anti-inflammatory, endothelial-protective and prebiotic properties that align with the pathophysiology of AS, making it a promising therapeutic candidate. This review systematically presents recent progress in understanding the anti-atherosclerotic effects of fucoidan, particularly its underlying mechanisms. These mechanisms involve the regulation of lipid levels, reduction of vascular inflammation, enhancement of antioxidant defenses, and protection of the vascular endothelium. These insights are essential for improving cardiovascular and cerebrovascular health.
Collapse
Affiliation(s)
- Qiujian Feng
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
- Beijing University of Chinese Medicine, Beijing, China
| | - Jinye Ma
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xuefan Jiang
- Beijing University of Chinese Medicine, Beijing, China
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Wei Wei
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Dongchen Xu
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yu Cao
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Hui Pei
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
4
|
Chen P, Hao L, Li Q, Wang H, Chen H, Zhang M, Jia A, Jiang H, Dai H, Zhang R. Identification of targets and comparative study of administration methods for the lipid-lowering effects of fucoidan from Saccharina japonica. Int J Biol Macromol 2025; 290:139102. [PMID: 39716710 DOI: 10.1016/j.ijbiomac.2024.139102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/21/2024] [Accepted: 12/20/2024] [Indexed: 12/25/2024]
Abstract
The lipid-lowering activity of fucoidan has been widely reported, but the exploration of its mechanisms is relatively limited, and studies on its direct targets are even scarcer. Additionally, it is unclear whether different administration methods affect the lipid-lowering activity of fucoidan. In current study, we used fucoidan derived from Saccharina japonica (SJF) to investigate its targets. The results showed that not only did SJF directly inhibit the Niemann-Pick C1-like 1 (NPC1L1)-mediated cholesterol transport, but it also reduced the solubility of cholesterol in mixed micelles, thereby interfering with the cholesterol uptake. Furthermore, SJF not only directly inhibited the activity of pancreatic triglyceride lipase (PTL), but also interfered with the overall catalytic process facilitated by colipase, thereby reducing the absorption of triglycerides. Moreover, comparative studies on the lipid-lowering activity of SJF administered via different methods demonstrated that dietary supplementation with SJF provided better lipid-lowering effects compared with gavage administration. Our research not only elucidates the targets and mechanisms of SJF but also provides theoretical basis for the selection of administration methods for fucoidan in lipid-lowering therapy.
Collapse
Affiliation(s)
- Pengwei Chen
- Hainan Key Laboratory for Research and Development of Natural Products from Li Folk Medicine, Institute of Tropical Bioscience and Biotechnology, Chinese Academy of Tropical Agricultural Sciences, Haikou 571101, China
| | - Li Hao
- Department of Gerontology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 528051, China
| | - Quancai Li
- Marine Biomedical Research Institute of Qingdao, Ocean University of China, Qingdao 266003, China
| | - Hao Wang
- Hainan Key Laboratory for Research and Development of Natural Products from Li Folk Medicine, Institute of Tropical Bioscience and Biotechnology, Chinese Academy of Tropical Agricultural Sciences, Haikou 571101, China
| | - Huiqin Chen
- Hainan Key Laboratory for Research and Development of Natural Products from Li Folk Medicine, Institute of Tropical Bioscience and Biotechnology, Chinese Academy of Tropical Agricultural Sciences, Haikou 571101, China
| | - Miao Zhang
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China
| | - Ang Jia
- The First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121000, China
| | - Hongfei Jiang
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China.
| | - Haofu Dai
- Hainan Key Laboratory for Research and Development of Natural Products from Li Folk Medicine, Institute of Tropical Bioscience and Biotechnology, Chinese Academy of Tropical Agricultural Sciences, Haikou 571101, China.
| | - Renshuai Zhang
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China.
| |
Collapse
|
5
|
Zhao W, Shen Y, Bao Y, Monroig Ó, Zhu T, Sun P, Tocher DR, Zhou Q, Jin M. Fucoidan alleviates hepatic lipid deposition by modulating the Perk-Eif2α-Atf4 axis via Sirt1 activation in Acanthopagrus schlegelii. Int J Biol Macromol 2024; 282:137266. [PMID: 39505163 DOI: 10.1016/j.ijbiomac.2024.137266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 11/03/2024] [Accepted: 11/04/2024] [Indexed: 11/08/2024]
Abstract
With the increasing use of high-fat diets (HFD), fatty liver disease has become common in fish, and fucoidan is of interest as a natural sulfated polysaccharide with lipid-lowering activity. To explore the molecular regulatory mechanisms of fucoidan's alleviation of HFD-induced lipid deposition in liver, black seabream (Acanthopagrus schlegelii) was used to construct in vivo and in vitro HFD models. In vivo HFD stimulated the protein kinase RNA-like endoplasmic reticulum kinase (Perk) pathway, and up-regulated proliferator-activated receptor gamma (Pparγ) nuclear translocation and expression of lipogenic genes, while it down-regulated Ppar alpha (Pparα) nuclear translocation and expression of lipolytic genes. However, fucoidan reversed these effects of HFD and significantly alleviated HFD-induced lipid accumulation in liver. Moreover, after sirtuin 1 (sirt1) knockdown, these effects of fucoidan disappeared. In the in vitro HFD model, GSK2606414 (GSK)-specific inhibition of the Perk pathway, decreased Pparγ nuclear translocation and increased Pparα nuclear translocation. Overall, fucoidan mitigated HFD-induced, Perk pathway-mediated lipid deposition in the liver of black seabream by activating Sirt1. The findings provided a new prospect for the application of green polysaccharides in aquatic animal feeds.
Collapse
Affiliation(s)
- Wenli Zhao
- Laboratory of Fish and Shellfish Nutrition, School of Marine Sciences, Ningbo University, Ningbo 315211, China; Key Laboratory of Aquacultural Biotechnology Ministry of Education, Ningbo University, Ningbo 315211, China; Key Laboratory of Green Mariculture (Co-construction by Ministry and Province), Ministry of Agriculture and Rural, Ningbo University, Ningbo 315211, China
| | - Yuedong Shen
- Laboratory of Fish and Shellfish Nutrition, School of Marine Sciences, Ningbo University, Ningbo 315211, China; Key Laboratory of Aquacultural Biotechnology Ministry of Education, Ningbo University, Ningbo 315211, China; Key Laboratory of Green Mariculture (Co-construction by Ministry and Province), Ministry of Agriculture and Rural, Ningbo University, Ningbo 315211, China
| | - Yangguang Bao
- Laboratory of Fish and Shellfish Nutrition, School of Marine Sciences, Ningbo University, Ningbo 315211, China; Key Laboratory of Aquacultural Biotechnology Ministry of Education, Ningbo University, Ningbo 315211, China; Key Laboratory of Green Mariculture (Co-construction by Ministry and Province), Ministry of Agriculture and Rural, Ningbo University, Ningbo 315211, China
| | - Óscar Monroig
- Instituto de Acuicultura Torre de la Sal (IATS), CSIC, 12595 Ribera de Cabanes, Castellon, Spain
| | - Tingting Zhu
- Laboratory of Fish and Shellfish Nutrition, School of Marine Sciences, Ningbo University, Ningbo 315211, China; Key Laboratory of Aquacultural Biotechnology Ministry of Education, Ningbo University, Ningbo 315211, China; Key Laboratory of Green Mariculture (Co-construction by Ministry and Province), Ministry of Agriculture and Rural, Ningbo University, Ningbo 315211, China
| | - Peng Sun
- Laboratory of Fish and Shellfish Nutrition, School of Marine Sciences, Ningbo University, Ningbo 315211, China; Key Laboratory of Aquacultural Biotechnology Ministry of Education, Ningbo University, Ningbo 315211, China; Key Laboratory of Green Mariculture (Co-construction by Ministry and Province), Ministry of Agriculture and Rural, Ningbo University, Ningbo 315211, China
| | - Douglas R Tocher
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Institute of Marine Sciences, Shantou University, Shantou 515063, China
| | - Qicun Zhou
- Laboratory of Fish and Shellfish Nutrition, School of Marine Sciences, Ningbo University, Ningbo 315211, China; Key Laboratory of Aquacultural Biotechnology Ministry of Education, Ningbo University, Ningbo 315211, China; Key Laboratory of Green Mariculture (Co-construction by Ministry and Province), Ministry of Agriculture and Rural, Ningbo University, Ningbo 315211, China
| | - Min Jin
- Laboratory of Fish and Shellfish Nutrition, School of Marine Sciences, Ningbo University, Ningbo 315211, China; Key Laboratory of Aquacultural Biotechnology Ministry of Education, Ningbo University, Ningbo 315211, China; Key Laboratory of Green Mariculture (Co-construction by Ministry and Province), Ministry of Agriculture and Rural, Ningbo University, Ningbo 315211, China.
| |
Collapse
|
6
|
Li X, Li M. Unlocking Cholesterol Metabolism in Metabolic-Associated Steatotic Liver Disease: Molecular Targets and Natural Product Interventions. Pharmaceuticals (Basel) 2024; 17:1073. [PMID: 39204178 PMCID: PMC11358954 DOI: 10.3390/ph17081073] [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: 07/07/2024] [Revised: 08/08/2024] [Accepted: 08/14/2024] [Indexed: 09/03/2024] Open
Abstract
Metabolic-associated steatotic liver disease (MASLD), the hepatic manifestation of metabolic syndrome, represents a growing global health concern. The intricate pathogenesis of MASLD, driven by genetic, metabolic, epigenetic, and environmental factors, leads to considerable clinical variability. Dysregulation of hepatic lipid metabolism, particularly cholesterol homeostasis, is a critical factor in the progression of MASLD and its more severe form, metabolic dysfunction-associated steatohepatitis (MASH). This review elucidates the multifaceted roles of cholesterol metabolism in MASLD, focusing on its absorption, transportation, biosynthesis, efflux, and conversion. We highlight recent advancements in understanding these processes and explore the therapeutic potential of natural products such as curcumin, berberine, and resveratrol in modulating cholesterol metabolism. By targeting key molecular pathways, these natural products offer promising strategies for MASLD management. Finally, this review also covers the clinical studies of natural products in MASLD, providing new insights for future research and clinical applications.
Collapse
Affiliation(s)
| | - Meng Li
- Institute of Digestive Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China;
| |
Collapse
|
7
|
Liu T, Wang X, Wang YM, Sui FR, Zhang XY, Liu HD, Ma DY, Liu XX, Guo SD. A comparative study of the hypolipidemic effects and mechanisms of action of Laminaria japonica- and Ascophyllum nodosum-derived fucoidans in apolipoprotein E-deficient mice. Food Funct 2024; 15:5955-5971. [PMID: 38738998 DOI: 10.1039/d3fo05521c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2024]
Abstract
The structural characteristics of fucoidans exhibit species and regional diversity. Previous studies have demonstrated that Laminaria japonica- and Ascophyllum nodosum-derived fucoidans have type I and type II fucosyl chains, respectively. These chemical differences may contribute to distinct hypolipidemic effects and mechanisms of action. Chemical analysis demonstrated that the percentage contents of sulfate, glucuronic acid, and galactose were higher in L. japonica-derived fucoidans than those of A. nodosum-derived fucoidans. In hyperlipidemic apolipoprotein E-deficient mice, both A. nodosum- and L. japonica-derived fucoidans significantly decreased the plasma and hepatic levels of total cholesterol and triglyceride, leading to the reduction of atherosclerotic plaques. Western blotting experiments demonstrated that these fucoidans significantly enhanced the expression and levels of scavenger receptor B type 1, cholesterol 7 alpha-hydroxylase A1, and peroxisome proliferator-activated receptor (PPAR)-α, contributing to circulating lipoprotein clearance and fatty acid degradation, respectively. Differentially, L. japonica-derived fucoidan significantly increased the LXR/ATP-binding cassette G8 signaling pathway in the small intestine, as revealed by real-time quantitative PCR, which may lead to further cholesterol and other lipid excretion. Collectively, these data are useful for understanding the hypolipidemic mechanisms of action of seaweed-derived fucoidans, and their potential application for the prevention and/or treatment of atherosclerotic cardiovascular diseases.
Collapse
Affiliation(s)
- Tian Liu
- Institute of Lipid Metabolism and Atherosclerosis, School of Pharmacy, Shandong Second Medical University, 7166# Baotongxi Street, Weifang 261053, Shandong Province, China.
| | - Xue Wang
- Institute of Lipid Metabolism and Atherosclerosis, School of Pharmacy, Shandong Second Medical University, 7166# Baotongxi Street, Weifang 261053, Shandong Province, China.
| | - Yan-Ming Wang
- Institute of Lipid Metabolism and Atherosclerosis, School of Pharmacy, Shandong Second Medical University, 7166# Baotongxi Street, Weifang 261053, Shandong Province, China.
| | - Feng-Rong Sui
- Institute of Lipid Metabolism and Atherosclerosis, School of Pharmacy, Shandong Second Medical University, 7166# Baotongxi Street, Weifang 261053, Shandong Province, China.
| | - Xue-Ying Zhang
- Institute of Lipid Metabolism and Atherosclerosis, School of Pharmacy, Shandong Second Medical University, 7166# Baotongxi Street, Weifang 261053, Shandong Province, China.
| | - Hai-Di Liu
- Institute of Lipid Metabolism and Atherosclerosis, School of Pharmacy, Shandong Second Medical University, 7166# Baotongxi Street, Weifang 261053, Shandong Province, China.
| | - Dong-Yue Ma
- Institute of Lipid Metabolism and Atherosclerosis, School of Pharmacy, Shandong Second Medical University, 7166# Baotongxi Street, Weifang 261053, Shandong Province, China.
| | - Xiao-Xiao Liu
- Institute of Lipid Metabolism and Atherosclerosis, School of Pharmacy, Shandong Second Medical University, 7166# Baotongxi Street, Weifang 261053, Shandong Province, China.
| | - Shou-Dong Guo
- Institute of Lipid Metabolism and Atherosclerosis, School of Pharmacy, Shandong Second Medical University, 7166# Baotongxi Street, Weifang 261053, Shandong Province, China.
| |
Collapse
|
8
|
Yin D, Zhong Y, Liu H, Hu J. Lipid metabolism regulation by dietary polysaccharides with different structural properties. Int J Biol Macromol 2024; 270:132253. [PMID: 38744359 DOI: 10.1016/j.ijbiomac.2024.132253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 04/28/2024] [Accepted: 05/07/2024] [Indexed: 05/16/2024]
Abstract
Lipid metabolism plays an important role in energy homeostasis maintenance in response to stress. Nowadays, hyperlipidemia-related chronic diseases such as obesity, diabetes, atherosclerosis, and fatty liver pose significant health challenges. Dietary polysaccharides (DPs) have gained attention for their effective lipid-lowering properties. This review examines the multifaceted mechanisms that DPs employ to lower lipid levels in subjects with hyperlipidemia. DPs could directly inhibit lipid intake and absorption, promote lipid excretion, and regulate key enzymes involved in lipid metabolism pathways, including triglyceride and cholesterol anabolism and catabolism, fatty acid oxidation, and bile acid synthesis. Additionally, DPs indirectly improve lipid homeostasis by modulating gut microbiota composition and alleviating oxidative stress. Moreover, the lipid-lowering mechanisms of particular structural DPs (including β-glucan, pectin, glucomannan, inulin, arabinoxylan, and fucoidan) are summarized. The relationship between the structure and lipid-lowering activity of DPs is also discussed based on current researches. Finally, potential breakthroughs and future directions in the development of DPs in lipid-lowering activity are discussed. The paper could provide a reference for further exploring the mechanism of DPs for lipid regulations and utilizing DPs as lipid-lowering dietary ingredients.
Collapse
Affiliation(s)
- Dafang Yin
- State Key Laboratory of Food Science and Resources, China-Canada Joint Laboratory of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, 235 Nanjing East Road, Nanchang, Jiangxi 330047, China
| | - Yadong Zhong
- State Key Laboratory of Food Science and Resources, China-Canada Joint Laboratory of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, 235 Nanjing East Road, Nanchang, Jiangxi 330047, China
| | - Huan Liu
- State Key Laboratory of Food Science and Resources, China-Canada Joint Laboratory of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, 235 Nanjing East Road, Nanchang, Jiangxi 330047, China
| | - Jielun Hu
- State Key Laboratory of Food Science and Resources, China-Canada Joint Laboratory of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, 235 Nanjing East Road, Nanchang, Jiangxi 330047, China.
| |
Collapse
|
9
|
Xue Q, Wang B, Feng J, Li C, Yu M, Zhao Y, Qi Z. Structural characterization and immune-enhancing effects of a novel polysaccharide extracted from Sargassum fusiforme. Int J Biol Macromol 2024; 270:132497. [PMID: 38763236 DOI: 10.1016/j.ijbiomac.2024.132497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 02/12/2024] [Accepted: 05/16/2024] [Indexed: 05/21/2024]
Abstract
To alleviate the adverse effects of chemotherapy and bolster immune function, a novel polysaccharide derived from Sargassum fusiforme named as SFP-αII. The structural composition of SFP-αII predominantly consisted of guluronic and mannuronic acids in a molar ratio of 33.8:66.2, with an average molecular weight of 16.5 kDa. Its structure was primarily characterized by →4)-α-GulA-(1 → and →4)-β-ManA-(1 → linkages confirmed by FT-IR, methylation, and NMR analyses. The absence of a triple-helix structure was in SFP-αII was confirmed using circular dichroism and Congo red dye assays. The dimensions varied with lengths ranging from 20 nm up to 3 μm revealed by atomic force microscopy (AFM). SFP-αII has been found to enhance immunomodulatory activity in cyclophosphamide (CTX)-induced immunosuppressed mice. This was evidenced by improvements in immune organ indices, cytokine levels, and the release of nitric oxide (NO). Specifically, SFP-αII mitigated immunosuppression by upregulating the secretion of IL-1β (167.3 %) and TNF-α (227.1 %) at a dose of 400 mg/kg, compared with the CTX group in macrophages. Ultimately, SFP-αII may serve as a mechanism for immune enhancement through modulation of TLR4-mediated NF-κB and MAPK signaling pathways. This integration of traditional Chinese and Western medicine, leveraging SFP-αII as a potential functional food could be pivotal in alleviating immunosuppressive side effects in CTX treatment.
Collapse
Affiliation(s)
- Qinbing Xue
- Engineering Research Center for Medicine, Ministry of Education, Harbin University of Commerce, Harbin, China
| | - Bing Wang
- School of Food Engineering, Harbin University of Commerce, Harbin, China
| | - Jie Feng
- Engineering Research Center for Medicine, Ministry of Education, Harbin University of Commerce, Harbin, China
| | - Chaoyu Li
- Engineering Research Center for Medicine, Ministry of Education, Harbin University of Commerce, Harbin, China
| | - Miao Yu
- Engineering Research Center for Medicine, Ministry of Education, Harbin University of Commerce, Harbin, China.
| | - Yan Zhao
- Medical Imaging Department, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Zheng Qi
- Engineering Research Center for Medicine, Ministry of Education, Harbin University of Commerce, Harbin, China.
| |
Collapse
|
10
|
Zhang Y, Zhang XY, Shi SR, Ma CN, Lin YP, Song WG, Guo SD. Natural products in atherosclerosis therapy by targeting PPARs: a review focusing on lipid metabolism and inflammation. Front Cardiovasc Med 2024; 11:1372055. [PMID: 38699583 PMCID: PMC11064802 DOI: 10.3389/fcvm.2024.1372055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 04/09/2024] [Indexed: 05/05/2024] Open
Abstract
Inflammation and dyslipidemia are critical inducing factors of atherosclerosis. Peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors and control the expression of multiple genes that are involved in lipid metabolism and inflammatory responses. However, synthesized PPAR agonists exhibit contrary therapeutic effects and various side effects in atherosclerosis therapy. Natural products are structural diversity and have a good safety. Recent studies find that natural herbs and compounds exhibit attractive therapeutic effects on atherosclerosis by alleviating hyperlipidemia and inflammation through modulation of PPARs. Importantly, the preparation of natural products generally causes significantly lower environmental pollution compared to that of synthesized chemical compounds. Therefore, it is interesting to discover novel PPAR modulator and develop alternative strategies for atherosclerosis therapy based on natural herbs and compounds. This article reviews recent findings, mainly from the year of 2020 to present, about the roles of natural herbs and compounds in regulation of PPARs and their therapeutic effects on atherosclerosis. This article provides alternative strategies and theoretical basis for atherosclerosis therapy using natural herbs and compounds by targeting PPARs, and offers valuable information for researchers that are interested in developing novel PPAR modulators.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Endocrinology and Metabolism, Guiqian International General Hospital, Guiyang, China
| | - Xue-Ying Zhang
- Institute of Lipid Metabolism and Atherosclerosis, School of Pharmacy, Shandong Second Medical University, Weifang, China
| | - Shan-Rui Shi
- Institute of Lipid Metabolism and Atherosclerosis, School of Pharmacy, Shandong Second Medical University, Weifang, China
| | - Chao-Nan Ma
- Institute of Lipid Metabolism and Atherosclerosis, School of Pharmacy, Shandong Second Medical University, Weifang, China
| | - Yun-Peng Lin
- Department of General Surgery, Qixia Traditional Chinese Medicine Hospital in Shandong Province, Yantai, China
| | - Wen-Gang Song
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Shou-Dong Guo
- Institute of Lipid Metabolism and Atherosclerosis, School of Pharmacy, Shandong Second Medical University, Weifang, China
| |
Collapse
|
11
|
Zhang Y, Liu T, Qu ZJ, Wang X, Song WG, Guo SD. Laminaria japonica Aresch-Derived Fucoidan Ameliorates Hyperlipidemia by Upregulating LXRs and Suppressing SREBPs. Cardiovasc Ther 2024; 2024:8649365. [PMID: 38375358 PMCID: PMC10876302 DOI: 10.1155/2024/8649365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 01/22/2024] [Accepted: 01/31/2024] [Indexed: 02/21/2024] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of morbidity and mortality worldwide, and hyperlipidemia is one major inducing factor of CVD. It is worthy to note that fucoidans are reported to have hypolipidemic activity with species specificity; however, the underlying mechanisms of action are far from clarification. This study is aimed at investigating the plasma lipid-lowering mechanisms of the fucoidan from L. japonica Aresch by detecting the levels of hepatic genes that are involved in lipid metabolism. Our results demonstrated that the fucoidan F3 significantly lowered total cholesterol and triglyceride in C57BL/6J mice fed a high-fat diet. In the mouse liver, fucoidan F3 intervention significantly increased the gene expression of peroxisome proliferator-activated receptor (PPAR) α, liver X receptor (LXR) α and β, and ATP-binding cassette transporter (ABC) G1 and G8 and decreased the expression of proprotein convertase subtilisin/kexin type 9 (PCSK9), low-density lipoprotein receptor, cholesterol 7 alpha-hydroxylase A1, and sterol regulatory element-binding protein (SREBP) 1c and SREBP-2. These results demonstrated that the antihyperlipidemic effects of fucoidan F3 are related to its activation of PPARα and LXR/ABC signaling pathways and inactivation of SREBPs. In conclusion, fucoidan F3 may be explored as a potential compound for prevention or treatment of lipid disorders.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Endocrinology and Metabolism, Guiqian International General Hospital, Guiyang 550018, China
| | - Tian Liu
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, Weifang 261053, China
| | - Ze-Jie Qu
- Cardiology Department, Qingzhou People's Hospital, Weifang 262500, China
| | - Xue Wang
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, Weifang 261053, China
| | - Wen-Gang Song
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, China
| | - Shou-Dong Guo
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, Weifang 261053, China
| |
Collapse
|
12
|
Li Y, Xiong Z, Jiang WL, Tian D, Zhou H, Hou Q, Xiao L, Zhang M, Huang L, Zhong L, Zhou L, Zeng GG. An innovative viewpoint on the existing and prospectiveness of SR-B1. Curr Probl Cardiol 2024; 49:102226. [PMID: 38040207 DOI: 10.1016/j.cpcardiol.2023.102226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 11/28/2023] [Indexed: 12/03/2023]
Abstract
Scavenger Receptor Class B Type 1 (SR-B1), a receptor protein expressed on the cell membrane, plays a crucial role in the metabolism and transport of cholesterol and other lipids, contributing significantly to the homeostasis of lipid levels within the body. Bibliometric analysis involves the application of mathematical and statistical methods to quantitatively analyze different types of documents. It involves the analysis of structural and temporal trends in scholarly articles, coupled with the identification of subject emphasis and variations. Through a bibliometric analysis, this study examines the historical background, current research trends, and future directions in the exploration of SR-B1. By offering insights into the research status and development of SR-B1, this paper aims to assist researchers in identifying novel pathways and areas of investigation in this field of study. Following the screening process, it can be concluded that research on SR-B1 has consistently remained a topic of significant interest over the past 17 years. Interestingly, SR-B1 has recently garnered attention in areas beyond its traditional research focus, including the field of cancer. The primary objective of this review is to provide a concise and accessible overview of the development process of SR-B1 that can help readers who are not well-versed in SR-B1 research quickly grasp its key aspects. Furthermore, this review aims to offer insights and suggestions to researchers regarding potential future research directions and areas of emphasis relating to SR-B1.
Collapse
Affiliation(s)
- Yonggui Li
- The Second Affiliated Hospital, Department of Digestive Internal Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Zhijie Xiong
- The Second Affiliated Hospital, Department of Digestive Internal Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Wan-Li Jiang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China; Departments of Clinical Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| | - Dandan Tian
- School of Nursing, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| | - Haiyou Zhou
- The Second Affiliated Hospital, Department of Digestive Internal Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, China; Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China; 2020 Grade Excellent Doctor Class of Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| | - Qin Hou
- Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| | - Liang Xiao
- The Second Affiliated Hospital, Department of Digestive Internal Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, China; Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China; 2020 Grade Excellent Doctor Class of Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| | - Mengjie Zhang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China; Departments of Clinical Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| | - Liubin Huang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China; Departments of Clinical Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| | - Lianping Zhong
- The Second Affiliated Hospital, Department of Digestive Internal Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, China; Department of Gastroenterology, the Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Li Zhou
- Department of Pathology, Chongqing Public Health Medical Center, Southwest University Public Health Hospital, Chongqing, China
| | - Guang-Gui Zeng
- The Second Affiliated Hospital, Department of Digestive Internal Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, China; Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China; 2020 Grade Excellent Doctor Class of Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China.
| |
Collapse
|
13
|
Deng C, Pan J, Zhu H, Chen ZY. Effect of Gut Microbiota on Blood Cholesterol: A Review on Mechanisms. Foods 2023; 12:4308. [PMID: 38231771 DOI: 10.3390/foods12234308] [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: 10/13/2023] [Revised: 11/24/2023] [Accepted: 11/27/2023] [Indexed: 01/19/2024] Open
Abstract
The gut microbiota serves as a pivotal mediator between diet and human health. Emerging evidence has shown that the gut microbiota may play an important role in cholesterol metabolism. In this review, we delve into five possible mechanisms by which the gut microbiota may influence cholesterol metabolism: (1) the gut microbiota changes the ratio of free bile acids to conjugated bile acids, with the former being eliminated into feces and the latter being reabsorbed back into the liver; (2) the gut microbiota can ferment dietary fiber to produce short-chain fatty acids (SCFAs) which are absorbed and reach the liver where SCFAs inhibit cholesterol synthesis; (3) the gut microbiota can regulate the expression of some genes related to cholesterol metabolism through their metabolites; (4) the gut microbiota can convert cholesterol to coprostanol, with the latter having a very low absorption rate; and (5) the gut microbiota could reduce blood cholesterol by inhibiting the production of lipopolysaccharides (LPS), which increases cholesterol synthesis and raises blood cholesterol. In addition, this review will explore the natural constituents in foods with potential roles in cholesterol regulation, mainly through their interactions with the gut microbiota. These include polysaccharides, polyphenolic entities, polyunsaturated fatty acids, phytosterols, and dicaffeoylquinic acid. These findings will provide a scientific foundation for targeting hypercholesterolemia and cardiovascular diseases through the modulation of the gut microbiota.
Collapse
Affiliation(s)
- Chuanling Deng
- School of Food Science and Engineering/National Technical Center (Foshan) for Quality Control of Famous and Special Agricultural Products (CAQS-GAP-KZZX043), Foshan University, Foshan 528011, China
| | - Jingjin Pan
- School of Food Science and Engineering/National Technical Center (Foshan) for Quality Control of Famous and Special Agricultural Products (CAQS-GAP-KZZX043), Foshan University, Foshan 528011, China
| | - Hanyue Zhu
- School of Food Science and Engineering/National Technical Center (Foshan) for Quality Control of Famous and Special Agricultural Products (CAQS-GAP-KZZX043), Foshan University, Foshan 528011, China
| | - Zhen-Yu Chen
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, NT, Hong Kong, China
| |
Collapse
|
14
|
He Z, Zhang Z, Xu P, Dirsch VM, Wang L, Wang K. Laminarin Reduces Cholesterol Uptake and NPC1L1 Protein Expression in High-Fat Diet (HFD)-Fed Mice. Mar Drugs 2023; 21:624. [PMID: 38132943 PMCID: PMC10744832 DOI: 10.3390/md21120624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 11/27/2023] [Accepted: 11/27/2023] [Indexed: 12/23/2023] Open
Abstract
Aberrantly high dietary cholesterol intake and intestinal cholesterol uptake lead to dyslipidemia, one of the risk factors for cardiovascular diseases (CVDs). Based on previous studies, laminarin, a polysaccharide found in brown algae, has hypolipidemic activity, but its underlying mechanism has not been elucidated. In this study, we investigated the effect of laminarin on intestinal cholesterol uptake in vitro, as well as the lipid and morphological parameters in an in vivo model of high-fat diet (HFD)-fed mice, and addressed the question of whether Niemann-Pick C1-like 1 protein (NPC1L1), a key transporter mediating dietary cholesterol uptake, is involved in the mechanistic action of laminarin. In in vitro studies, BODIPY-cholesterol-labeled Caco-2 cells were examined using confocal microscopy and a fluorescence reader. The results demonstrated that laminarin inhibited cholesterol uptake into Caco-2 cells in a concentration-dependent manner (EC50 = 20.69 μM). In HFD-fed C57BL/6J mice, laminarin significantly reduced the serum levels of total cholesterol (TC), total triglycerides (TG), and low-density lipoprotein cholesterol (LDL-C). It also decreased hepatic levels of TC, TG, and total bile acids (TBA) while promoting the excretion of fecal cholesterol. Furthermore, laminarin significantly reduced local villous damage in the jejunum of HFD mice. Mechanistic studies revealed that laminarin significantly downregulated NPC1L1 protein expression in the jejunum of HFD-fed mice. The siRNA-mediated knockdown of NPC1L1 attenuated the laminarin-mediated inhibition of cholesterol uptake in Caco-2 cells. This study suggests that laminarin significantly improves dyslipidemia in HFD-fed mice, likely by reducing cholesterol uptake through a mechanism that involves the downregulation of NPC1L1 expression.
Collapse
Affiliation(s)
- Zhuoqian He
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, Qingdao 266073, China; (Z.H.); (P.X.); (K.W.)
| | - Zhongyin Zhang
- Department of Medicinal Chemistry, School of Pharmacy, Qingdao University Medical College, Qingdao 266073, China;
| | - Pengfei Xu
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, Qingdao 266073, China; (Z.H.); (P.X.); (K.W.)
| | - Verena M. Dirsch
- Department of Pharmaceutical Sciences, Faculty of Life Sciences, University of Vienna, 1090 Vienna, Austria;
| | - Limei Wang
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, Qingdao 266073, China; (Z.H.); (P.X.); (K.W.)
- Institute of Innovative Drugs, Qingdao University, Qingdao 266071, China
| | - Kewei Wang
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, Qingdao 266073, China; (Z.H.); (P.X.); (K.W.)
- Institute of Innovative Drugs, Qingdao University, Qingdao 266071, China
| |
Collapse
|
15
|
Development and characterization of a fucoidan-based nanoemulsion using Nigella sativa oil for improvement of anti-obesity activity of fucoxanthin in an obese rat model. Int J Biol Macromol 2023; 235:123867. [PMID: 36870664 DOI: 10.1016/j.ijbiomac.2023.123867] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 02/23/2023] [Accepted: 02/24/2023] [Indexed: 03/06/2023]
Abstract
The anti-obesity activity of encapsulated fucoxanthin in fucoidan-based nanoemulsion was investigated. Then, high-fat diet (HFD) induced-obese rats were fed along with different treatments including administration of encapsulated fucoxanthin (10 mg/kg and 50 mg/kg/day), fucoidan (70 mg/kg), Nigella sativa oil (250 mg/kg), metformin (200 mg/kg), and free form of fucoxanthin (50 mg/kg) by oral gavage daily for 7 weeks. The study discovered that fucoidan-based nanoemulsions with a low and high dose of fucoxanthin had droplet size in the range of 181.70-184.87 nm and encapsulation efficacy of 89.94-91.68 %, respectively. Also exhibited 75.86 % and 83.76 % fucoxanthin in vitro release. The TEM images and FTIR spectera confirmed the particle size and encapsulation of fucoxanthin, respectively. Moreover, in vivo results revealed that encapsulated fucoxanthin reduced body and liver weight compared with a HFD group (p < 0.05). Biochemical parameters (FBS, TG, TC, HDL, LDL) and liver enzymes (ALP, AST, and ALT) were decreased after fucoxanthin and fucoidan administration. According to the histopathological analysis, fucoxanthin and fucoidan attenuated lipid accumulation in the liver.
Collapse
|
16
|
A Comprehensive Review of the Cardioprotective Effect of Marine Algae Polysaccharide on the Gut Microbiota. Foods 2022; 11:foods11223550. [PMID: 36429141 PMCID: PMC9689188 DOI: 10.3390/foods11223550] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 10/30/2022] [Accepted: 11/04/2022] [Indexed: 11/10/2022] Open
Abstract
Cardiovascular disease (CVD) is the number one cause of death worldwide. Recent evidence has demonstrated an association between the gut microbiota and CVD, including heart failure, cerebrovascular illness, hypertension, and stroke. Marine algal polysaccharides (MAPs) are valuable natural sources of diverse bioactive compounds. MAPs have many pharmaceutical activities, including antioxidant, anti-inflammatory, immunomodulatory, and antidiabetic effects. Most MAPs are not utilized in the upper gastrointestinal tract; however, they are fermented by intestinal flora. The relationship between MAPs and the intestinal microbiota has drawn attention in CVD research. Hence, this review highlights the main action by which MAPs are known to affect CVD by maintaining homeostasis in the gut microbiome and producing gut microbiota-generated functional metabolites and short chain fatty acids. In addition, the effects of trimethylamine N-oxide on the gut microbiota composition, bile acid signaling properties, and CVD prevention are also discussed. This review supports the idea that focusing on the interactions between the host and gut microbiota may be promising for the prevention or treatment of CVD. MAPs are a potential sustainable source for the production of functional foods or nutraceutical products for preventing or treating CVD.
Collapse
|
17
|
Zaitseva OO, Sergushkina MI, Khudyakov AN, Polezhaeva TV, Solomina ON. Seaweed sulfated polysaccharides and their medicinal properties. ALGAL RES 2022. [DOI: 10.1016/j.algal.2022.102885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
18
|
Savla SR, Prabhavalkar KS, Bhatt LK. Liver X Receptor: a potential target in the treatment of atherosclerosis. Expert Opin Ther Targets 2022; 26:645-658. [PMID: 36003057 DOI: 10.1080/14728222.2022.2117610] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Liver X receptors (LXRs) are master regulators of atherogenesis. Their anti-atherogenic potential has been attributed to their role in the inhibition of macrophage-mediated inflammation and promotion of reverse cholesterol transport. Owing to the significance of their anti-atherogenic potential, it is essential to develop and test new generation LXR agonists, both synthetic and natural, to identify potential LXR-targeted therapeutics for the future. AREAS COVERED This review describes the role of LXRs in atherosclerotic development, provides a summary of LXR agonists and future directions for atherosclerosis research. We searched PubMed, Scopus and Google Scholar for relevant reports, from last 10 years, using atherosclerosis, liver X receptor, and LXR agonist as keywords. EXPERT OPINION LXRα has gained widespread recognition as a regulator of cholesterol homeostasis and expression of inflammatory genes. Further research using models of cell type-specific knockout and specific agonist-targeted LXR isoforms is warranted. Enthusiasm for therapeutic value of LXR agonists has been tempered due to LXRα-mediated induction of hepatic lipogenesis. LXRα agonism and LXRβ targeting, gut-specific inverse LXR agonists, investigations combining LXR agonists with other lipogenesis mitigating agents, like IDOL antagonists and synthetic HDL, and targeting ABCA1, M2 macrophages and LXRα phosphorylation, remain as promising possibilities.
Collapse
Affiliation(s)
- Shreya R Savla
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai 400056, India
| | - Kedar S Prabhavalkar
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai 400056, India
| | - Lokesh K Bhatt
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai 400056, India
| |
Collapse
|
19
|
Méndez-Del Villar M, Pérez-Rubio KG, Hernández-Corona DM, Cortez-Navarrete M. Therapeutic Effect of Fucoidan on Metabolic Diseases: Experimental Data and Clinical Evidence. J Med Food 2022; 25:1011-1020. [PMID: 35984868 DOI: 10.1089/jmf.2022.0005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The rising prevalence of metabolic diseases represents a major challenge to public health worldwide. Therefore, there is a strong need to conduct research on the effectiveness of complementary and alternative therapies for metabolic disorders. Fucoidan is a fucose-enriched and sulfated polysaccharide extracted from ubiquitous brown seaweed. The antihypertensive, antidiabetic, antiobesity, and hypolipidemic effects of fucoidan have been reported in preclinical research and clinical trials. This study aims to review the mechanisms of action and the experimental and clinical use of different types of fucoidan for the treatment of metabolic diseases.
Collapse
Affiliation(s)
- Miriam Méndez-Del Villar
- Multidisciplinary Health Research Center, Biomedical Sciences Department, University Center of Tonala, University of Guadalajara, Tonala, Jalisco, Mexico
| | - Karina G Pérez-Rubio
- Institute of Experimental and Clinical Therapeutics, Department of Physiology, Health Science University Center, University of Guadalajara, Guadalajara, Jalisco, Mexico
| | - Diana M Hernández-Corona
- Multidisciplinary Health Research Center, Biomedical Sciences Department, University Center of Tonala, University of Guadalajara, Tonala, Jalisco, Mexico
| | - Marisol Cortez-Navarrete
- Institute of Experimental and Clinical Therapeutics, Department of Physiology, Health Science University Center, University of Guadalajara, Guadalajara, Jalisco, Mexico
| |
Collapse
|
20
|
Yang H, Feng L, Xu L, Jiang D, Zhai F, Tong G, Xing Y. Intervention of Shugan Xiaozhi Decoction on Nonalcoholic Fatty Liver Disease via Mediating Gut-Liver Axis. BIOMED RESEARCH INTERNATIONAL 2022; 2022:4801695. [PMID: 35837380 PMCID: PMC9276511 DOI: 10.1155/2022/4801695] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 06/08/2022] [Indexed: 11/18/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a chronic liver disease with an increasing incidence rate but few therapies. Shugan Xiaozhi decoction (SX) has demonstrated beneficial effects in treating NAFLD with an unclear mechanism. This study was aimed at investigating the therapeutic mechanism of SX on high-fat diet-induced NAFLD rats via the gut-liver axis. Hepatic steatosis and integrity of intestinal mucosa in NAFLD rats were assessed by histopathological staining. The level of lipid and inflammation were estimated by enzyme-linked immunosorbent assay. Western Blotting was used to detect apolipoprotein (apo) B48 expression. 16S rRNA analysis was used to measure the changes of gut microbial composition after SX treatment. The expressions of zona occludens 1 protein (ZO-1), occludin, and secretory immunoglobulin A (sIgA) in the colon were detected by immunostaining to investigate the intestinal barrier function. Our study found that SX reduced hepatic steatosis, the levels of alanine aminotransferase, aspartate aminotransferase, total cholesterol, and triglyceride and apoB48 expression but increased peroxisome proliferator activated receptor α (PPARα) level. Moreover, SX altered the diversity of gut microbiota, upregulating the relative abundance of f_Prevotellaceae, while downregulating f_Bacteroidales_ S24-7, f_Lachnospiraceae, f_Ruminococcaceae, f_Erysipelotrichaceae, and f_Desulfovibrionaceae. By increasing the expression of ZO-1 and occludin and decreasing the level of proinflammatory factors, including sIgA, lipopolysaccharide, tumor necrosis factor-α, interleukin-1β, monocyte chemotactic protein-1, and transforming growth factor-β1, SX improved intestinal mucosal integrity and barrier function. Our study illustrated that the gut-liver axis was a potential way for SX to ameliorate NAFLD, that is, by regulating the expression of PPARα, apoB48, and modulating gut microbiota to protect the intestinal barrier function, and thus alleviate lipid deposition and inflammatory response in the liver.
Collapse
Affiliation(s)
- Huili Yang
- Hepatology Department, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong 518033, China
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong 518033, China
| | - Lian Feng
- Hepatology Department, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong 518033, China
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong 518033, China
| | - Linyi Xu
- Hepatology Department, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong 518033, China
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong 518033, China
| | - Dansheng Jiang
- Hepatology Department, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong 518033, China
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong 518033, China
| | - Fenfen Zhai
- Shenzhen Futian Center for Chronic Disease Control, Shenzhen, Guangdong 518048, China
| | - Guangdong Tong
- Hepatology Department, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong 518033, China
| | - Yufeng Xing
- Hepatology Department, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong 518033, China
| |
Collapse
|
21
|
Lin P, Chen S, Zhong S. Nutritional and Chemical Composition of Sargassum zhangii and the Physical and Chemical Characterization, Binding Bile Acid, and Cholesterol-Lowering Activity in HepG2 Cells of Its Fucoidans. Foods 2022; 11:foods11121771. [PMID: 35741969 PMCID: PMC9223202 DOI: 10.3390/foods11121771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 06/11/2022] [Accepted: 06/13/2022] [Indexed: 11/16/2022] Open
Abstract
Fucoidan is a marine sulfated polysaccharide that is rich in Sargassum and has a wide range of biological activities. In this study, the chemical composition and bile acid binding ability of six crude fucoidans were compared, the nutrition and chemical composition of Sargassum zhangii were analyzed, and fucoidan from Sargassum zhangii was extracted and purified. The purified fractions (ZF1, ZF2, and ZF3) were analyzed by physicochemical characterization, and the ability of binding bile acid and cholesterol lowering in HepG2 cells were evaluated. The results showed that the contents of sulfate in crude fucoidan from Sargassum Zhangii (ZF) was as high as13.63%. Its ability of binding bile acid was better than other five crude fucoidans. Sargassum zhangii was a kind of brown seaweed with high carbohydrate, and low fat and rich in minerals. The sulfate content of ZF1, ZF2, and ZF3 was 3.29%, 19.39%, and 18.89% respectively, and the molecular weight (Mw) was 4.026 × 105, 2.893 × 105, and 3.368 × 105, respectively. Three fucoidans all contained the characteristic absorption bands of polysaccharides and sulfate groups and were rich in fucose. Three fucoidans can bind to bile acid, and ZF2 showed the best binding capability. In vitro experiments showed that ZF1, ZF2, and ZF3 could reduce intracellular total cholesterol (TC) content in HepG2 cells without affecting their viability. ZF2 showed the best ability to reduce TC.
Collapse
Affiliation(s)
- Peichun Lin
- School of Chemistry and Environment, Guangdong Ocean University, Zhanjiang 524088, China;
| | - Suhua Chen
- School of Chemistry and Environment, Guangdong Ocean University, Zhanjiang 524088, China;
- Correspondence: ; Tel.: +86-759-239-6026
| | - Siyan Zhong
- School of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China;
| |
Collapse
|
22
|
Wang K, Xu X, Wei Q, Yang Q, Zhao J, Wang Y, Li X, Ji K, Song S. Application of fucoidan as treatment for cardiovascular and cerebrovascular diseases. Ther Adv Chronic Dis 2022; 13:20406223221076891. [PMID: 35432845 PMCID: PMC9008857 DOI: 10.1177/20406223221076891] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 01/12/2022] [Indexed: 11/17/2022] Open
Abstract
Fucoidan is a marine polysaccharide. In recent years, fucoidan has attracted wide-scale attention from the pharmaceutical industries due to its diverse biological activities such as lipid-lowering, anti-atherosclerosis, and anticoagulation. This review clarifies the pharmacological effects of fucoidan in the treatment of human cardiovascular and cerebrovascular diseases. Fucoidan exerts a hypolipidemic effect by increasing the reverse transport of cholesterol, inhibiting lipid synthesis, reducing lipid accumulation, and increasing lipid metabolism. Inflammation, anti-oxidation, and so on have a regulatory effect in the process of atherosclerosis endothelial cells, macrophages, smooth muscle cells, and so on; fucoidan can not only prevent thrombosis through anticoagulation and regulate platelet activation, but also promote the dissolution of formed thrombi. Fucoidan has a neuroprotective effect, and also has a positive effect on the prognosis of the cardiovascular and cerebrovascular. The prospects of applying fucoidan in cardio-cerebrovascular diseases are reviewed to provide some theoretical bases and inspirations for its full-scale development and utilization.
Collapse
Affiliation(s)
- Ke Wang
- Marine College, Shandong University, Weihai,
ChinaHeping Hospital Affiliated to Changzhi Medical College, Changzhi,
China
| | - Xueli Xu
- Binzhou Inspection and Testing Center, Binzhou,
China
| | - Qiang Wei
- Marine College, Shandong University, Weihai,
China
| | - Qiong Yang
- Marine College, Shandong University, Weihai,
China
| | - Jiarui Zhao
- Marine College, Shandong University, Weihai,
China
| | - Yuan Wang
- Marine College, Shandong University, Weihai,
China
| | - Xia Li
- Marine College, Shandong University, Weihai,
China
| | - Kai Ji
- Department of Plastic Surgery, China-Japan
Friendship Hospital, Beijing 100029, China
| | - Shuliang Song
- Marine College, Shandong University, Weihai
264209, China
| |
Collapse
|
23
|
Seaweed Exhibits Therapeutic Properties against Chronic Diseases: An Overview. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12052638] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Seaweeds or marine macroalgae are known for producing potentially bioactive substances that exhibit a wide range of nutritional, therapeutic, and nutraceutical properties. These compounds can be applied to treat chronic diseases, such as cancer, cardiovascular disease, osteoporosis, neurodegenerative diseases, and diabetes mellitus. Several studies have shown that consumption of seaweeds in Asian countries, such as Japan and Korea, has been correlated with a lower incidence of chronic diseases. In this study, we conducted a review of published papers on seaweed consumption and chronic diseases. We used the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) method for this study. We identified and screened research articles published between 2000 and 2021. We used PubMed and ScienceDirect databases and identified 107 articles. This systematic review discusses the potential use of bioactive compounds of seaweed to treat chronic diseases and identifies gaps where further research in this field is needed. In this review, the therapeutic and nutraceutical properties of seaweed for the treatment of chronic diseases such as neurodegenerative diseases, obesity, diabetes, cancer, liver disease, cardiovascular disease, osteoporosis, and arthritis were discussed. We concluded that further study on the identification of bioactive compounds of seaweed, and further study at a clinical level, are needed.
Collapse
|
24
|
Li Y, Miao M, Yin F, Shen N, Yu WQ, Guo S. The polysaccharide-peptide complex from mushroom Cordyceps militaris ameliorates atherosclerosis by modulating the lncRNA-miRNA-mRNA axis. Food Funct 2022; 13:3185-3197. [DOI: 10.1039/d1fo03285b] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Polysaccharides from mushroom Cordyceps militaris are found to have pleiotropic bioactivities, suggesting a potential role in prevention of atherosclerosis. However, the underlying mechanisms of action are not clear. In this...
Collapse
|
25
|
Zhang S, Hong F, Ma C, Yang S. Hepatic Lipid Metabolism Disorder and Atherosclerosis. Endocr Metab Immune Disord Drug Targets 2021; 22:590-600. [PMID: 34931971 DOI: 10.2174/1871530322666211220110810] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 07/28/2021] [Accepted: 11/01/2021] [Indexed: 11/22/2022]
Abstract
Lipid metabolism disorder plays a fundamental role in the pathogenesis of atherosclerosis. As the largest metabolic organ of the human body, liver has a key role in lipid metabolism by influencing fat production, fat decomposition, and the intake and secretion of serum lipoproteins. Numerous clinical and experimental studies have indicated that the dysfunction of hepatic lipid metabolism is closely tied to the onset of atherosclerosis. However, the identity and functional role of hepatic lipid metabolism responsible for these associations remain unknown. This review presented that cholesterol synthesis, cholesterol transport, and the metabolism of triglyceride, lipoproteins, and fatty acids are all associated with hepatic lipid metabolism and atherosclerosis. Moreover, we also discussed the roles of gut microbiota, inflammatory response, and oxidative stress in the pathological association between hepatic lipid metabolism and atherosclerosis. These significant evidences support strongly that hepatic lipid metabolism disorders may increase the risk of atherosclerosis.
Collapse
Affiliation(s)
- Sen Zhang
- Department of Physiology, College of Medicine, Nanchang University, Nanchang, China
| | - Fenfang Hong
- Experimental Center of Pathogen Biology, Nanchang University, Nanchang, China
| | - Chen Ma
- Department of Physiology, College of Medicine, Nanchang University, Nanchang, China
| | - Shulong Yang
- Department of Physiology, College of Medicine, Nanchang University, Nanchang, China
| |
Collapse
|
26
|
Yu WQ, Yin F, Shen N, Lin P, Xia B, Li YJ, Guo SD. Polysaccharide CM1 from Cordyceps militaris hinders adipocyte differentiation and alleviates hyperlipidemia in LDLR (+/-) hamsters. Lipids Health Dis 2021; 20:178. [PMID: 34895241 PMCID: PMC8667404 DOI: 10.1186/s12944-021-01606-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 11/26/2021] [Indexed: 12/15/2022] Open
Abstract
Background Cordyceps militaris is cultured widely as an edible mushroom and accumulating evidence in mice have demonstrated that the polysaccharides of Cordyceps species have lipid-lowering effects. However, lipid metabolism in mice is significantly different from that in humans, making a full understanding of the mechanisms at play critical. Methods After 5 months, the hamsters were weighed and sampled under anesthesia after overnight fasting. The lipid-lowering effect and mechanisms of the polysaccharide CM1 was investigated by cellular and molecular technologies. Furthermore, the effect of the polysaccharide CM1 (100 μg/mL) on inhibiting adipocyte differentiation was investigated in vitro. Results CM1, a polysaccharide from C. militaris, significantly decreased plasma total cholesterol, triglyceride and epididymal fat index in LDLR(+/−) hamsters, which have a human-like lipid profile. After 5 months’ administration, CM1 decreased the plasma level of apolipoprotein B48, modulated the expression of key genes and proteins in liver, small intestine, and epididymal fat. CM1 also inhibited preadipocyte differentiation in 3T3-L1 cells by downregulating the key genes involved in lipid droplet formation. Conclusions The polysaccharide CM1 lowers lipid and adipocyte differentiation by several pathways, and it has potential applications for hyperlipidemia prevention. Supplementary Information The online version contains supplementary material available at 10.1186/s12944-021-01606-6.
Collapse
Affiliation(s)
- Wen-Qian Yu
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, Baotongxi street 7166#, Weifang, Shandong province, China
| | - Fan Yin
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, Baotongxi street 7166#, Weifang, Shandong province, China
| | - Nuo Shen
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, Baotongxi street 7166#, Weifang, Shandong province, China
| | - Ping Lin
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, Baotongxi street 7166#, Weifang, Shandong province, China
| | - Bin Xia
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, Baotongxi street 7166#, Weifang, Shandong province, China
| | - Yan-Jie Li
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, Baotongxi street 7166#, Weifang, Shandong province, China.
| | - Shou-Dong Guo
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, Baotongxi street 7166#, Weifang, Shandong province, China.
| |
Collapse
|
27
|
Liang B, Cai XY, Gu N. Marine Natural Products and Coronary Artery Disease. Front Cardiovasc Med 2021; 8:739932. [PMID: 34621803 PMCID: PMC8490644 DOI: 10.3389/fcvm.2021.739932] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 08/24/2021] [Indexed: 12/18/2022] Open
Abstract
Coronary artery disease is the major cause of mortality worldwide, especially in low- and middle-income earners. To not only reduce angina symptoms and exercise-induced ischemia but also prevent cardiovascular events, pharmacological intervention strategies, including antiplatelet drugs, anticoagulant drugs, statins, and other lipid-lowering drugs, and renin-angiotensin-aldosterone system blockers, are conducted. However, the existing drugs for coronary artery disease are incomprehensive and have some adverse reactions. Thus, it is necessary to look for new drug research and development. Marine natural products have been considered a valuable source for drug discovery because of their chemical diversity and biological activities. The experiments and investigations indicated that several marine natural products, such as organic small molecules, polysaccharides, proteins, and bioactive peptides, and lipids were effective for treating coronary artery disease. Here, we particularly discussed the functions and mechanisms of active substances in coronary artery disease, including antiplatelet, anticoagulant, lipid-lowering, anti-inflammatory, and antioxidant activities.
Collapse
Affiliation(s)
- Bo Liang
- Nanjing University of Chinese Medicine, Nanjing, China
| | - Xin-Yi Cai
- Nanjing University of Chinese Medicine, Nanjing, China
| | - Ning Gu
- Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
28
|
Human cholesteryl ester transport protein transgene promotes macrophage reverse cholesterol transport in C57BL/6 mice and phospholipid transfer protein gene knockout mice. J Physiol Biochem 2021; 77:683-694. [PMID: 34403126 DOI: 10.1007/s13105-021-00834-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 07/27/2021] [Indexed: 02/07/2023]
Abstract
Cholesteryl ester transfer protein (CETP) and phospholipid transfer protein (PLTP) belong to the same gene family. Liver-specific expression of CETP improves reverse cholesterol transport (RCT) and PLTP knockout (KO) decreases RCT in mice. In this study, we investigate the effect of CETP transgene (CETP-tg) on RCT and whether CETP-tg can partially restore RCT efficiency in PLTP KO mice. Several rounds of crossing were carried out to produce colonies of wild type (WT), CETP-tg, PLTP KO, and CETP-tg × PLTP KO mice were obtained after several generations of reproduction. The efficiency of RCT was detected using [3H]-cholesterol-laden macrophages, and the underlying mechanisms were investigated by multiple techniques. Our data demonstrated that CETP-tg significantly increased the transport rate of [3H]-cholesterol from macrophages to plasma and liver, and finally the excretion through feces compared to the WT littermates. The RCT improving effect of CETP-tg was similar in PLTPKO mice. Furthermore, CETP-tg did not affect the expression of RCT-related proteins, such as low-density lipoprotein receptor. The mechanisms of improving RCT may be attributed to the low level of oxidized lipids in CETP-tg mouse and CETP-mediated lipid transport. Collectively, CETP-tg improves RCT in mice, and CETP can not compensate for PLTP deficiency.
Collapse
|
29
|
Polysaccharide Structures and Their Hypocholesterolemic Potential. Molecules 2021; 26:molecules26154559. [PMID: 34361718 PMCID: PMC8348680 DOI: 10.3390/molecules26154559] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 07/23/2021] [Accepted: 07/24/2021] [Indexed: 12/12/2022] Open
Abstract
Several classes of polysaccharides have been described to have hypocholesterolemic potential, namely cholesterol bioaccessibility and bioavailability. This review will highlight the main mechanisms by which polysaccharides are known to affect cholesterol homeostasis at the intestine, namely the effect (i) of polysaccharide viscosity and its influence on cholesterol bioaccessibility; (ii) on bile salt sequestration and its dependence on the structural diversity of polysaccharides; (iii) of bio-transformations of polysaccharides and bile salts by the gut microbiota. Different quantitative structure–hypocholesterolemic activity relationships have been explored depending on the mechanism involved, and these were based on polysaccharide physicochemical properties, such as sugar composition and ramification degree, linkage type, size/molecular weight, and charge. The information gathered will support the rationalization of polysaccharides’ effect on cholesterol homeostasis and highlight predictive rules towards the development of customized hypocholesterolemic functional food.
Collapse
|
30
|
Abu Hafsa SH, Khalel MS, El-Gindy YM, Hassan AA. Nutritional potential of marine and freshwater algae as dietary supplements for growing rabbits. ITALIAN JOURNAL OF ANIMAL SCIENCE 2021. [DOI: 10.1080/1828051x.2021.1928557] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Salma H. Abu Hafsa
- Livestock Research Department, Arid Lands Cultivation Research Institute, City of Scientific Research and Technological Applications, New Borg El-Arab, Alexandria, Egypt
| | - Mohamed S. Khalel
- Animal Production Research Institute, Agricultural Research Center, Dokki, Egypt
| | - Yassmine M. El-Gindy
- Department of Animal and Fish Production, Faculty of Agriculture, Alexandria University, Alexandria, Egypt
| | - Ayman A. Hassan
- Animal Production Research Institute, Agricultural Research Center, Dokki, Egypt
| |
Collapse
|
31
|
Lin P, Ji HH, Li YJ, Guo SD. Macrophage Plasticity and Atherosclerosis Therapy. Front Mol Biosci 2021; 8:679797. [PMID: 34026849 PMCID: PMC8138136 DOI: 10.3389/fmolb.2021.679797] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 04/12/2021] [Indexed: 12/15/2022] Open
Abstract
Atherosclerosis is a chronic disease starting with the entry of monocytes into the subendothelium and the subsequent differentiation into macrophages. Macrophages are the major immune cells in atherosclerotic plaques and are involved in the dynamic progression of atherosclerotic plaques. The biological properties of atherosclerotic plaque macrophages determine lesion size, composition, and stability. The heterogenicity and plasticity of atherosclerotic macrophages have been a hotspot in recent years. Studies demonstrated that lipids, cytokines, chemokines, and other molecules in the atherosclerotic plaque microenvironment regulate macrophage phenotype, contributing to the switch of macrophages toward a pro- or anti-atherosclerosis state. Of note, M1/M2 classification is oversimplified and only represent two extreme states of macrophages. Moreover, M2 macrophages in atherosclerosis are not always protective. Understanding the phenotypic diversity and functions of macrophages can disclose their roles in atherosclerotic plaques. Given that lipid-lowering therapy cannot completely retard the progression of atherosclerosis, macrophages with high heterogeneity and plasticity raise the hope for atherosclerosis regression. This review will focus on the macrophage phenotypic diversity, its role in the progression of the dynamic atherosclerotic plaque, and finally discuss the possibility of treating atherosclerosis by targeting macrophage microenvironment.
Collapse
Affiliation(s)
- Ping Lin
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, Weifang, China
| | - Hong-Hai Ji
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, Weifang, China
| | - Yan-Jie Li
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, Weifang, China
| | - Shou-Dong Guo
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, Weifang, China
| |
Collapse
|
32
|
Xia J, Yu P, Zeng Z, Ma M, Zhang G, Wan D, Gong D, Deng S, Wang J. High Dietary Intervention of Lauric Triglyceride Might be Harmful to Its Improvement of Cholesterol Metabolism in Obese Rats. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:4453-4463. [PMID: 33844520 DOI: 10.1021/acs.jafc.1c00745] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Hypercholesterolemia is often considered to be a major risk factor for atherosclerosis, and medium-chain fatty acids have been found to reduce the total cholesterol (TC) level and maintain low-density lipoprotein cholesterol (LDL-c) stability. However, we unexpectedly found that the levels of TC and LDL-c were increased in obese rats treated with high-dose lauric triglycerides (LT). The study aimed to investigate the effect and mechanism of LT on cholesterol metabolism in obese rats. Our results showed that LT intervention could reduce cholesterol biosynthesis by downregulating the expression of HMG-CoA reductase in obese rats. LT increased the expression levels of PPARγ1, LXRα, ABCA1, and ABCG8 in the liver. These results indicated that LT could improve the lipid transfer and bile acid efflux. However, LT significantly increased the expression of PCSK 9, resulting in accelerated degradation of LDLR, thus reducing the transport of very LDL (VLDL) and LDL to the liver. Together with the increased expression of NPC1L1 protein, LT impaired the uptake of VLDL/LDL by the liver and increased the reabsorption of sterols, leading to an increase in the levels of TC and LDL-c in obese rats.
Collapse
Affiliation(s)
- Jiaheng Xia
- Jiangxi Province Key Laboratory of Edible and Medicinal Resources Exploitation, School of Resource and Environmental and Chemical Engineering, Nanchang University, Nanchang 330031, China
| | - Ping Yu
- Jiangxi Province Key Laboratory of Edible and Medicinal Resources Exploitation, School of Resource and Environmental and Chemical Engineering, Nanchang University, Nanchang 330031, China
| | - Zheling Zeng
- Jiangxi Province Key Laboratory of Edible and Medicinal Resources Exploitation, School of Resource and Environmental and Chemical Engineering, Nanchang University, Nanchang 330031, China
- State Key Laboratory of Food Science and Technology, Jiangxi Province Key Laboratory of Edible and Medicinal Resources Exploitation, School of Food Science and Technology, Nanchang University, Nanchang 330031, China
| | - Maomao Ma
- State Key Laboratory of Food Science and Technology, Jiangxi Province Key Laboratory of Edible and Medicinal Resources Exploitation, School of Food Science and Technology, Nanchang University, Nanchang 330031, China
| | - Guohua Zhang
- State Key Laboratory of Food Science and Technology, Jiangxi Province Key Laboratory of Edible and Medicinal Resources Exploitation, School of Food Science and Technology, Nanchang University, Nanchang 330031, China
| | - Dongman Wan
- School of Food Science and Technology, Nanchang University, Nanchang 330031, China
| | - Deming Gong
- State Key Laboratory of Food Science and Technology, Jiangxi Province Key Laboratory of Edible and Medicinal Resources Exploitation, New Zealand Institute of Natural Medicine Research, 8 Ha Crescent, Auckland 2104, New Zealand
| | - Shuguang Deng
- Jiangxi Province Key Laboratory of Edible and Medicinal Resources Exploitation, School for Engineering of Matter, Transport and Energy, Arizona State University, Tempe, Arizona 85284, United States
| | - Jun Wang
- Jiangxi Province Key Laboratory of Edible and Medicinal Resources Exploitation, School of Resource and Environmental and Chemical Engineering, Nanchang University, Nanchang 330031, China
| |
Collapse
|
33
|
Zhou MX, Tian X, Wu ZQ, Li K, Li ZJ. Fuzhuan brick tea supplemented with areca nuts: Effects on serum and gut microbiota in mice. J Food Biochem 2021; 45:e13737. [PMID: 33876445 DOI: 10.1111/jfbc.13737] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 03/16/2021] [Accepted: 04/02/2021] [Indexed: 11/28/2022]
Abstract
Areca nut and Fuzhuan brick tea, a type of natural plant products, have obvious effects of fat reduction and weight loss; however, there is no report on their synergistic effect. This study investigated the effects of Fuzhuan brick tea supplemented with different concentrations of areca nut (5% (LAF), 10% (MAF), and 20% (HAF)) on serum and gut microbiota in Kunming (KM) mice. The results showed that Fuzhuan brick tea supplemented with areca nuts (AFTs) could reduce weight, prevent the accumulation of fat, inhibit the increase in the levels of serum triglyceride, total cholesterol, low-density lipoprotein cholesterol, blood glucose, free fatty acid, insulin, and total bile acid, alleviate the decrease in high-density lipoprotein cholesterol level, and regulate the composition of gut microbiota by high-fat diet intervention. The HAF group with 20% areca nut content showed the best effect. These results could provide a novel approach to prevent obesity and hyperlipidemia. PRACTICAL APPLICATIONS: Consumption of areca nut and tea is widespread in Asia and other regions. As a controversial raw material, the damage due to areca nut to oral mucosa health has often aroused public concern and heated discussion; however, its medicinal value has been confirmed in terms of its pharmacological effects in various aspects. Fuzhuan brick tea, a type of traditional postfermented dark tea, has been confirmed to exert effects of antiobesity. Therefore, the areca nut and Fuzhuan brick tea, as a type of natural plant products, have obvious effects of fat reduction and weight loss; however, their synergistic effect has not been reported. To our knowledge, this study is the first to explore the effects of the Fuzhuan brick tea supplemented with areca nuts (AFTs) on serum and gut microbiota in mice. On the premise of exerting their beneficial effects (especially in terms of easing food stagnation and eliminating indigestion) and reducing their toxic and side effects, the effects of AFTs on health were further clarified, which could provide a novel direction for the development and utilization of areca nut. Moreover, our research would increase public understanding of areca nut and provide guidance to the Fuzhuan brick tea processing industry.
Collapse
Affiliation(s)
- Ming-Xi Zhou
- College of Food Science and Technology, Hunan Agricultural University, Changsha, China
| | - Xing Tian
- College of Food Science and Technology, Hunan Agricultural University, Changsha, China.,College of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Zhong-Qin Wu
- College of Food Science and Technology, Hunan Agricultural University, Changsha, China
| | - Ke Li
- College of Food Science and Technology, Hunan Agricultural University, Changsha, China
| | - Zong-Jun Li
- College of Food Science and Technology, Hunan Agricultural University, Changsha, China
| |
Collapse
|
34
|
Martín-Mateos R, Albillos A. The Role of the Gut-Liver Axis in Metabolic Dysfunction-Associated Fatty Liver Disease. Front Immunol 2021; 12:660179. [PMID: 33936094 PMCID: PMC8085382 DOI: 10.3389/fimmu.2021.660179] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 03/29/2021] [Indexed: 12/12/2022] Open
Abstract
The complex interplay between the gut microbiota, the intestinal barrier, the immune system and the liver is strongly influenced by environmental and genetic factors that can disrupt the homeostasis leading to disease. Among the modulable factors, diet has been identified as a key regulator of microbiota composition in patients with metabolic syndrome and related diseases, including the metabolic dysfunction-associated fatty liver disease (MAFLD). The altered microbiota disrupts the intestinal barrier at different levels inducing functional and structural changes at the mucus lining, the intercellular junctions on the epithelial layer, or at the recently characterized vascular barrier. Barrier disruption leads to an increased gut permeability to bacteria and derived products which challenge the immune system and promote inflammation. All these alterations contribute to the pathogenesis of MAFLD, and thus, therapeutic approaches targeting the gut-liver-axis are increasingly being explored. In addition, the specific changes induced in the intestinal flora may allow to characterize distinctive microbial signatures for non-invasive diagnosis, severity stratification and disease monitoring.
Collapse
Affiliation(s)
| | - Agustín Albillos
- Department of Gastroenterology and Hepatology, Hospital Universitario Ramón y Cajal, Universidad de Alcalá, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
35
|
Sharma PP, Baskaran V. Polysaccharide (laminaran and fucoidan), fucoxanthin and lipids as functional components from brown algae (Padina tetrastromatica) modulates adipogenesis and thermogenesis in diet-induced obesity in C57BL6 mice. ALGAL RES 2021. [DOI: 10.1016/j.algal.2021.102187] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
36
|
Ji C, Zhang Z, Zhang B, Chen J, Liu R, Song D, Li W, Lin N, Zou X, Wang J, Guo S. Purification, characterization, and in vitro antitumor activity of a novel glucan from the purple sweet potato Ipomoea Batatas (L.) Lam. Carbohydr Polym 2021; 257:117605. [DOI: 10.1016/j.carbpol.2020.117605] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 12/17/2020] [Accepted: 12/30/2020] [Indexed: 12/13/2022]
|
37
|
Purification, structural characterization, and PCSK9 secretion inhibitory effect of the novel alkali-extracted polysaccharide from Cordyceps militaris. Int J Biol Macromol 2021; 179:407-417. [PMID: 33662421 DOI: 10.1016/j.ijbiomac.2021.02.191] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 02/21/2021] [Accepted: 02/25/2021] [Indexed: 01/17/2023]
Abstract
One novel alkali-extracted polysaccharide, CM3-SII, was obtained from the fruiting body of C. militaris via column chromatography. Its structural characteristics were investigated via chemical and spectroscopic methods. The backbone of CM3-SII was composed of →4)-β-D-Manp(1→, →6)-β-D-Manp(1→, and →6)-α-D-Manp(1→ glycosyls, and branching at the O-4 positions of →6)-β-D-Manp(1→ glycosyls with β-D-Galp, (1→2) linked-β-D-Galf, and →2,6)-α-D-Manp(1→ residues. Furthermore, O-6 and O-2 positions of the →2,6)-α-D-Manp(1→ residues were substituted with methyl and β-D-Galp, respectively. This polysaccharide significantly enhanced the intracellular protein expression of low-density lipoprotein receptor and proprotein convertase subtilisin/kexin type 9 (PCSK9) via regulating sterol regulatory element-binding protein 2 in hepatoma Huh7 cells. Of note, CM3-SII significantly decreased PCSK9 secretion at the concentration of 200 μg/mL. Collectively, CM3-SII is different from the previously reported alkali-extracted polysaccharides isolated from the fruiting body of C. militaris, and it may have potential application in hypolipidemia or as a pharmaceutical additive.
Collapse
|
38
|
Fan Y, Yan LT, Yao Z, Xiong GY. Biochanin A Regulates Cholesterol Metabolism Further Delays the Progression of Nonalcoholic Fatty Liver Disease. Diabetes Metab Syndr Obes 2021; 14:3161-3172. [PMID: 34276221 PMCID: PMC8277457 DOI: 10.2147/dmso.s315471] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 06/23/2021] [Indexed: 12/11/2022] Open
Abstract
PURPOSE To discover the possible target of biochanin A (BCA) in the lipid metabolism pathway and further explore its mechanism to nonalcoholic fatty liver disease (NAFLD). METHODS We adopted a high-fat and high-glucose diet for 12 weeks to build the NAFLD rat model, which was then treated with different proportions of BCA for 4 weeks. General condition, body weight, Lee index, and liver index were then evaluated. Furthermore, blood lipid level and insulin resistance (IR) were detected. Moreover, hematoxylin and eosin and oil red O staining were used to observe the pathological changes in the liver. Finally, Western blotting was used to detect the protein expression levels of CYP7A1, HMGCR, LDLR, PPAR-α, PPAR-γ, and SREBP-1c in the liver. RESULTS The vital signs of rats in each group were stable. The treatment with BCA effectively reduced Lee index and liver index (F = 104.781, P < 0.05); however, the weight was not effected in each group. Additionally, BCA effectively reduced the related lipid metabolism indexes of NAFLD, such as total cholesterol (TC), triglyceride (TG), low-density lipoprotein (LDL), blood glucose, insulin, IR (F =12.463 (TC), 6.909 [TG], and 15.3 effected 75 [LDL], P < 0.05), and increased HDL (F = 11.580, P < 0.05). We observed that BCA could significantly improve steatosis and inflammatory cell infiltration in liver slices. Furthermore, BCA significantly increased the CYP7A1, LDLR, and PPAR-α protein expression in the liver and downregulated the HMGCR, SREBP-1c, and PPAR-γ protein expression. CONCLUSION BCA could delay the liver damage of NAFLD induced by a high-fat diet, regulate the blood lipid level, and improve the expression of lipid metabolism-related genes in rats.
Collapse
Affiliation(s)
- Yan Fan
- Department of Anatomy, Histology, and Embryology, School of Basic Medical Science, Kunming Medical University, Kunming, Yunnan, 650500, People’s Republic of China
| | - Long-Teng Yan
- Key Laboratory of Microcosmic Syndrome Differentiation, School of Basic Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan, 650500, People’s Republic of China
| | - Zheng Yao
- Key Laboratory of Microcosmic Syndrome Differentiation, School of Basic Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan, 650500, People’s Republic of China
- Correspondence: Zheng Yao; Guang-Yi Xiong Key Laboratory of Microcosmic Syndrome Differentiation, School of Basic Medicine, Yunnan University of Chinese Medicine, No. 1076, Yuhua Road, Chenggong District, Kunming, Yunnan, 650500, People’s Republic of ChinaTel/Fax +86 189 0871 9365 Email ;
| | - Guang-Yi Xiong
- Key Laboratory of Microcosmic Syndrome Differentiation, School of Basic Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan, 650500, People’s Republic of China
| |
Collapse
|
39
|
Rossi R, Vizzarri F, Ratti S, Palazzo M, Casamassima D, Corino C. Effects of Long-Term Supplementation with Brown Seaweeds and Polyphenols in Rabbit on Meat Quality Parameters. Animals (Basel) 2020; 10:ani10122443. [PMID: 33419317 PMCID: PMC7766534 DOI: 10.3390/ani10122443] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/16/2020] [Accepted: 12/17/2020] [Indexed: 11/16/2022] Open
Abstract
The objective of the present study was to evaluate the effects of dam and offspring dietary supplementation with a natural feed additive on the growth performance and meat quality parameters of growing rabbits. The growing rabbits are selected from lactating does receiving a control diet (C) or diets supplemented with 0.3% (SP1) and 0.6% (SP2) of feed additive containing brown seaweeds (Laminaria spp.) and plant extracts. In the postweaning phase, the growing rabbits remained in the treatment group defined by their does and the trial lasted 42 days. The average daily feed intake and feed conversion ratio were improved in the rabbit fed 0.6% of the natural feed additive. The cholesterol content tended to be lower in Longissimus lumborum (LL) muscle and decrease in Semimembranosus (SM) muscle (in SP2 -41.36% than controls). The α tocopherol and retinol content were enhanced in both muscles of rabbit fed the natural mixture (SP1 and SP2 groups). An improvement of sensory attributes of texture was observed in both muscles from rabbit fed natural mixture. In conclusion, long term supplementation of both lactating does and offspring with the high dosage of brown seaweed and plant polyphenols improves growth performance and enhances meat nutritional and sensory parameters.
Collapse
Affiliation(s)
- Raffaella Rossi
- Dipartimento di Medicina Veterinaria, Università Degli Studi di Milano, Via dell’Università 6, 26900 Lodi, Italy; (S.R.); (C.C.)
- Correspondence:
| | - Francesco Vizzarri
- Department of Agricultural and Environmental Science, Università di Bari Aldo Moro, Via G. Amendola 165/A, 70126 Bari, Italy;
| | - Sabrina Ratti
- Dipartimento di Medicina Veterinaria, Università Degli Studi di Milano, Via dell’Università 6, 26900 Lodi, Italy; (S.R.); (C.C.)
| | - Marisa Palazzo
- Department of Agricultural, Environmental and Food Sciences, Università Degli Studi del Molise, Via F. De Sanctis 1, 86100 Campobasso, Italy; (M.P.); (D.C.)
| | - Donato Casamassima
- Department of Agricultural, Environmental and Food Sciences, Università Degli Studi del Molise, Via F. De Sanctis 1, 86100 Campobasso, Italy; (M.P.); (D.C.)
| | - Carlo Corino
- Dipartimento di Medicina Veterinaria, Università Degli Studi di Milano, Via dell’Università 6, 26900 Lodi, Italy; (S.R.); (C.C.)
| |
Collapse
|
40
|
Yu XC, Fu Y, Bi YH, Zhang WW, Li J, Ji T, Chao Y, Meng QH, Chen Q, Ma MH, Zhang YH, Shan J, Bian HM. Alisol B 23-acetate activates ABCG5/G8 in the jejunum via the LXRα/ACAT2 pathway to relieve atherosclerosis in ovariectomized ApoE -/- mice. Aging (Albany NY) 2020; 12:25744-25766. [PMID: 33234731 PMCID: PMC7803561 DOI: 10.18632/aging.104185] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 10/06/2020] [Indexed: 12/20/2022]
Abstract
Phytosterols have been shown to improve blood lipid levels and treat atherosclerosis. This research investigated the effects of phytosterol Alisol B 23-acetate (AB23A) on jejunum lipid metabolism and atherosclerosis. The results show that intragastric administration of AB23A can significantly reduce atherosclerotic plaque area and lipid accumulation in the jejunum of ovariectomized ApoE-/- mice fed a high-fat diet and can also improve the lipid mass spectra of the plasma and jejunum. In vitro studies have shown that AB23A can increase cholesterol outflow in Caco-2 cells exposed to high fat concentrations and increase the expression of ATP-binding cassette transfer proteins G5/G8 (ABCG5/G8), the liver X receptor α (LXRα). Furthermore, inhibition of LXRα can significantly eliminate the active effect of AB23A on decreasing intracellular lipid accumulation. We also confirmed that AB23A has a negative effect on Acyl-CoA cholesterol acyltransferase 2 (ACAT2) in Caco-2 cells cultured in the high concentrations of fat, and we found that AB23A further reduces ACAT2 expression in cells treated with the ACAT2 inhibitor pyripyropene or transfected with ACAT2 siRNA. In conclusion, we confirmed that AB23A can reduce the absorption of dietary lipids in the jejunum by affecting the LXRα-ACAT2-ABCG5/G8 pathway and ultimately exert an anti-atherosclerotic effect.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily G, Member 5/drug effects
- ATP Binding Cassette Transporter, Subfamily G, Member 5/metabolism
- ATP Binding Cassette Transporter, Subfamily G, Member 8/drug effects
- ATP Binding Cassette Transporter, Subfamily G, Member 8/metabolism
- Animals
- Aorta/drug effects
- Aorta/metabolism
- Aorta/pathology
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Caco-2 Cells
- Cholestenones/pharmacology
- Cholesterol/metabolism
- Cholesterol Esters/metabolism
- Diet, High-Fat
- Female
- Glycerophospholipids/metabolism
- Humans
- Jejunum/drug effects
- Jejunum/metabolism
- Jejunum/pathology
- Lipid Droplets/drug effects
- Lipid Droplets/metabolism
- Lipid Droplets/pathology
- Lipid Metabolism/drug effects
- Lipoproteins/drug effects
- Lipoproteins/metabolism
- Liver X Receptors/drug effects
- Liver X Receptors/metabolism
- Mice
- Mice, Knockout, ApoE
- Ovariectomy
- Plaque, Atherosclerotic/metabolism
- Plaque, Atherosclerotic/pathology
- Sterol O-Acyltransferase/drug effects
- Sterol O-Acyltransferase/metabolism
- Triglycerides/metabolism
- Sterol O-Acyltransferase 2
Collapse
Affiliation(s)
- Xi-Chao Yu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yu Fu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yun-Hui Bi
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Wei-Wei Zhang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jun Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Tingting Ji
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Ying Chao
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Qing-Hai Meng
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Qi Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Meng-Hua Ma
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yu-Han Zhang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jinjun Shan
- Institute of Pediatrics, Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Hui-Min Bian
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- National Standard Laboratory of Pharmacology of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Key Laboratory of Therapeutic Material of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| |
Collapse
|
41
|
Li J, Guo C, Wu J. Fucoidan: Biological Activity in Liver Diseases. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2020; 48:1617-1632. [PMID: 33148007 DOI: 10.1142/s0192415x20500809] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Fucoidan is a type of polysaccharide rich in sulfuric acid groups and is mainly found in brown algae. Due to its extensive biological activities, such as anticoagulant, antitumor, antithrombotic, antiviral, anti-oxidant and enhancing immune function, fucoidan has gradually become a research hotspot. Under the scientific guidance of modern medical theory, fucoidan and its mechanism in oxidative stress, carbohydrate and lipid metabolism, inflammatory response, tumor proliferation, and metastasis have become a new research direction and an important basis as an effective liver protection drug. In this paper, we discuss the important role of fucoidan in viral hepatitis, liver fibrosis, liver cancer, nonalcoholic fatty liver and liver injury induced by drugs and ischemia and briefly discuss its underlying mechanism. We supplement the theoretical basis for its clinical application and provide effective targets for the development of follow-up dominant drugs.
Collapse
Affiliation(s)
- Jingjing Li
- Department of Gastroenterology, Putuo People's Hospital, Tongji University School of Medicine, Shanghai 200060, P. R. China
| | - Chuanyong Guo
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Shanghai 200072, P. R. China
| | - Jianye Wu
- Department of Gastroenterology, Putuo People's Hospital, Tongji University School of Medicine, Shanghai 200060, P. R. China
| |
Collapse
|
42
|
Cao Q, Zhao J, Xing M, Xiao H, Zhang Q, Liang H, Ji A, Song S. Current Research Landscape of Marine-Derived Anti-Atherosclerotic Substances. Mar Drugs 2020; 18:md18090440. [PMID: 32854344 PMCID: PMC7551282 DOI: 10.3390/md18090440] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/19/2020] [Accepted: 08/20/2020] [Indexed: 12/18/2022] Open
Abstract
Atherosclerosis is a chronic disease characterized by lipid accumulation and chronic inflammation of the arterial wall, which is the pathological basis for coronary heart disease, cerebrovascular disease and thromboembolic disease. Currently, there is a lack of low-cost therapeutic agents that effectively slow the progression of atherosclerosis. Therefore, the development of new drugs is urgently needed. The research and development of marine-derived drugs have gained increasing interest from researchers across the world. Many marine organisms provide a rich material basis for the development of atherosclerotic drugs. This review focuses on the latest technological advances in the structures and mechanisms of action of marine-derived anti-atherosclerotic substances and the challenges of the application of these substances including marine polysaccharides, proteins and peptides, polyunsaturated fatty acids and small molecule compounds. Here, we describe the theoretical basis of marine biological resources in the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Qi Cao
- Marine College, Shandong University, Weihai 264209, China; (Q.C.); (J.Z.); (M.X.); (H.X.); (Q.Z.); (H.L.)
| | - Jiarui Zhao
- Marine College, Shandong University, Weihai 264209, China; (Q.C.); (J.Z.); (M.X.); (H.X.); (Q.Z.); (H.L.)
| | - Maochen Xing
- Marine College, Shandong University, Weihai 264209, China; (Q.C.); (J.Z.); (M.X.); (H.X.); (Q.Z.); (H.L.)
| | - Han Xiao
- Marine College, Shandong University, Weihai 264209, China; (Q.C.); (J.Z.); (M.X.); (H.X.); (Q.Z.); (H.L.)
| | - Qian Zhang
- Marine College, Shandong University, Weihai 264209, China; (Q.C.); (J.Z.); (M.X.); (H.X.); (Q.Z.); (H.L.)
| | - Hao Liang
- Marine College, Shandong University, Weihai 264209, China; (Q.C.); (J.Z.); (M.X.); (H.X.); (Q.Z.); (H.L.)
| | - Aiguo Ji
- Marine College, Shandong University, Weihai 264209, China; (Q.C.); (J.Z.); (M.X.); (H.X.); (Q.Z.); (H.L.)
- School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
- Correspondence: (A.J.); (S.S.)
| | - Shuliang Song
- Marine College, Shandong University, Weihai 264209, China; (Q.C.); (J.Z.); (M.X.); (H.X.); (Q.Z.); (H.L.)
- Correspondence: (A.J.); (S.S.)
| |
Collapse
|
43
|
Han AR, Kim JH, Kim E, Cui J, Chai IS, Zhang G, Lee Y. Hypotriglyceridemic effects of brown seaweed consumption via regulation of bile acid excretion and hepatic lipogenesis in high fat diet-induced obese mice. Nutr Res Pract 2020; 14:580-592. [PMID: 33282121 PMCID: PMC7683202 DOI: 10.4162/nrp.2020.14.6.580] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 06/03/2020] [Accepted: 07/21/2020] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND/OBJECTIVES The present study aimed to further investigate the potential health beneficial effects of long-term seaweed supplementation on lipid metabolism and hepatic functions in DIO mice. MATERIALS/METHODS Four brown seaweeds (Undaria pinnatifida [UP], Laminaria japonica [LJ], Sargassum fulvellum [SF], or Hizikia fusiforme [HF]) were added to a high fat diet (HFD) at a 5% ratio and supplemented to C57BL/6N mice for 16 weeks. Triglycerides (TGs) and total cholesterol (TC) in the liver, feces, and plasma were measured. Fecal bile acid (BA) levels in feces were monitored. Hepatic insulin signaling- and lipogenesis-related proteins were evaluated by Western blot analysis. RESULTS Fasting blood glucose levels were significantly reduced in the LJ, SF, and HF groups compared to the HFD group by the end of 16-week feeding period. Plasma TG levels and hepatic lipid accumulation were significantly reduced in all 4 seaweed supplemented groups, whereas plasma TC levels were only suppressed in the UP and HF groups compared to the HFD group. Fecal BA levels were significantly elevated by UP, LJ, and SF supplementation compared to HFD feeding only. Lastly, regarding hepatic insulin signaling-related proteins, phosphorylation of 5′-AMP-activated protein kinase was significantly up-regulated by all 4 types of seaweed, whereas phosphorylation of protein kinase B was up-regulated only in the SF and HF groups. Lipogenesis-related proteins in the liver were effectively down-regulated by HF supplementation in DIO mice. CONCLUSIONS Brown seaweed consumption showed hypotriglyceridemic effects in the prolonged DIO mouse model. Specifically, combinatory regulation of BA excretion and lipogenesis-related proteins in the liver by seaweed supplementation contributed to the reduction of plasma and hepatic TG levels, which inhibited hyperglycemia in DIO mice. Thus, the discrepant and species-specific functions of brown seaweeds provide novel insights for the selection of future targets for therapeutic agents.
Collapse
Affiliation(s)
- A-Reum Han
- Department of Food Science and Nutrition, Jeju National University, Jeju 63243, Korea.,Yerae Elementary School, Jeju 63537, Korea
| | - Jae-Hoon Kim
- College of Veterinary Medicine, Jeju National University, Jeju 63243, Korea
| | - Eunyoung Kim
- Department of Food Science and Nutrition, Jeju National University, Jeju 63243, Korea
| | - Jiamei Cui
- Department of Food Science and Nutrition, Jeju National University, Jeju 63243, Korea
| | - In-Suk Chai
- Department of Food Science and Nutrition, Jeju National University, Jeju 63243, Korea
| | - Guiguo Zhang
- Department of Animal Nutrition, Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Taian 271018, China
| | - Yunkyoung Lee
- Department of Food Science and Nutrition, Jeju National University, Jeju 63243, Korea
| |
Collapse
|
44
|
Li T, Yin J, Ji Y, Lin P, Li Y, Yang Z, Hu S, Wang J, Zhang B, Koshti S, Wang J, Ji C, Guo S. Setosphapyrone C and D accelerate macrophages cholesterol efflux by promoting LXRα/ABCA1 pathway. Arch Pharm Res 2020; 43:788-797. [PMID: 32779151 DOI: 10.1007/s12272-020-01255-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 07/22/2020] [Indexed: 02/08/2023]
Abstract
LXRα agonists have attracted significant attention due to their potential biological activities on promoting cholesterol efflux. This study was designed to investigate whether setosphapyrone C and D have potential lipid-lowering capacity and the underlying mechanisms in vitro. Our data showed that setosphapyrone C and D had weak cytotoxicity compared to the liver X receptor α (LXRα) agonist T0901317. In RAW 264.7 macrophages, setosphapyrone C and D significantly enhanced [3H]-cholesterol efflux by ~ 21.3% and 32.4%, respectively; furthermore, setosphapyrone C and D enhanced the protein levels of ATP-binding cassette transporter (ABC) A1 and LXRα by 58% and 69%, and 60% and 70% (8 µM), respectively; however, they had no effect on the protein levels of ABCG1 and scavenger receptor B type 1; additionally, they had minor effect on the mRNA expression of lipogenic genes. Of note, setosphapyrone C and D significantly enhanced LXRα/ABCA1pathway in mice primary macrophages. In BRL cells, setosphapyrone C and D significantly improved the protein levels of ABCA1 and ABCG1; setosphapyrone D significantly enhanced the protein expression of low-density lipoprotein. Collectively, setosphapyrone C and D with weak cytotoxicity exhibited effective lipid-lowering effect via enhancing LXRα/ABC pathways. Setosphapyrones possess potential application for the treatment of hyperlipidemic diseases.
Collapse
Affiliation(s)
- Ting Li
- College of Pharmacy Engineering Research Center for Medicine, Harbin University of Commerce, 150076, Harbin, China.,Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, 261053, Weifang, China
| | - Jiayu Yin
- College of Pharmacy Engineering Research Center for Medicine, Harbin University of Commerce, 150076, Harbin, China.,Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, 261053, Weifang, China
| | - Yubin Ji
- College of Pharmacy Engineering Research Center for Medicine, Harbin University of Commerce, 150076, Harbin, China
| | - Ping Lin
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, 261053, Weifang, China
| | - Yanjie Li
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, 261053, Weifang, China
| | - Zixun Yang
- College of Pharmacy Engineering Research Center for Medicine, Harbin University of Commerce, 150076, Harbin, China.,Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, 261053, Weifang, China
| | - Shumei Hu
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, 261053, Weifang, China
| | - Jin Wang
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, 261053, Weifang, China
| | - Baihui Zhang
- College of Pharmacy Engineering Research Center for Medicine, Harbin University of Commerce, 150076, Harbin, China.,Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, 261053, Weifang, China
| | - Saloni Koshti
- Department of Physiology, University of Alberta, T6G2R3, Edmonton, Canada
| | - Junfeng Wang
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica/RNAM Center for Marine Microbiology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 510220, Guangzhou, China.
| | - Chenfeng Ji
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, 261053, Weifang, China.
| | - Shoudong Guo
- College of Pharmacy Engineering Research Center for Medicine, Harbin University of Commerce, 150076, Harbin, China. .,Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, 261053, Weifang, China.
| |
Collapse
|
45
|
Zhao J, Cao Q, Xing M, Xiao H, Cheng Z, Song S, Ji A. Advances in the Study of Marine Products with Lipid-Lowering Properties. Mar Drugs 2020; 18:E390. [PMID: 32726987 PMCID: PMC7459887 DOI: 10.3390/md18080390] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 07/22/2020] [Accepted: 07/23/2020] [Indexed: 12/18/2022] Open
Abstract
With twice the number of cancer's deaths, cardiovascular diseases have become the leading cause of death worldwide. Atherosclerosis, in particular, is a progressive, chronic inflammatory cardiovascular disease caused by persistent damage to blood vessels due to elevated cholesterol levels and hyperlipidemia. This condition is characterized by an increase in serum cholesterol, triglycerides, and low-density lipoprotein, and a decrease in high-density lipoprotein. Although existing therapies with hypolipidemic effects can improve the living standards of patients with cardiovascular diseases, the drugs currently used in clinical practice have certain side effects, which insists on the need for the development of new types of drugs with lipid-lowering effects. Some marine-derived substances have proven hypolipidemic activities with fewer side effects and stand as a good alternative for drug development. Recently, there have been thousands of studies on substances with lipid-lowering properties of marine origin, and some are already implemented in clinical practice. Here, we summarize the active components of marine-derived products having a hypolipidemic effect. These active constituents according to their source are divided into algal, animal, plant and microbial and contribute to the development and utilization of marine medicinal products with hypolipidemic effects.
Collapse
Affiliation(s)
- Jiarui Zhao
- Marine College, Shandong University, Weihai 264209, China; (J.Z.); (Q.C.); (M.X.); (H.X.); (Z.C.)
| | - Qi Cao
- Marine College, Shandong University, Weihai 264209, China; (J.Z.); (Q.C.); (M.X.); (H.X.); (Z.C.)
| | - Maochen Xing
- Marine College, Shandong University, Weihai 264209, China; (J.Z.); (Q.C.); (M.X.); (H.X.); (Z.C.)
| | - Han Xiao
- Marine College, Shandong University, Weihai 264209, China; (J.Z.); (Q.C.); (M.X.); (H.X.); (Z.C.)
| | - Zeyu Cheng
- Marine College, Shandong University, Weihai 264209, China; (J.Z.); (Q.C.); (M.X.); (H.X.); (Z.C.)
| | - Shuliang Song
- Marine College, Shandong University, Weihai 264209, China; (J.Z.); (Q.C.); (M.X.); (H.X.); (Z.C.)
| | - Aiguo Ji
- Marine College, Shandong University, Weihai 264209, China; (J.Z.); (Q.C.); (M.X.); (H.X.); (Z.C.)
- School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| |
Collapse
|
46
|
Korolenko TA, Bgatova NP, Ovsyukova MV, Shintyapina A, Vetvicka V. Hypolipidemic Effects of β-Glucans, Mannans, and Fucoidans: Mechanism of Action and Their Prospects for Clinical Application. Molecules 2020; 25:molecules25081819. [PMID: 32316136 PMCID: PMC7221696 DOI: 10.3390/molecules25081819] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 04/10/2020] [Accepted: 04/10/2020] [Indexed: 12/17/2022] Open
Abstract
The search for lipid-lowering drugs is important for clinical medicine. This review summarizes our research findings regarding the hypolipidemic activity of polysaccharides. There are several validated agents altering lipid levels which reduce the risk of atherosclerotic cardiovascular events. Nonetheless, for many people, the risk of such an event remains unacceptably high despite treatment with these agents. This situation has prompted the search for new therapies to reduce the residual cardiovascular risk. The lipid-lowering effect of β-glucans consumed with food was demonstrated in patients with atherosclerosis. The mechanism of the protective effect of β-glucans is poorly studied. The effects of β-glucans are mediated by Toll-like receptors, by dectin-1, and possibly by other receptors. Nevertheless, the mechanism of the protective action of β-glucan in lipemic mice has been studied insufficiently. This review will present up-to-date information regarding experimental hypolipidemic polysaccharide compounds that hold promise for medicine. Phagocyte-specific chitotriosidase in humans contributes to innate immune responses against chitin-containing fungi. This enzyme has been first described in patients with Gaucher disease and serves as an important diagnostic biomarker. It has been reported that, in mice, chitin particles of certain size are recognized by macrophages through Toll-like receptors, dectin-1, and to a lesser extent through mannose receptor.
Collapse
Affiliation(s)
- Tatiana A. Korolenko
- Department of Clinical Neuroscience, Behavior and Neurotechnologies, Institute of Physiology and Basic Medicine, Timakov St. 4, Novosibirsk 630117, Russia; (T.A.K.); (M.V.O.)
| | - Nataliya P. Bgatova
- Laboratory of Ultrastructural Research, Department of Experimental Pharmacology, Research Institute of Clinical and Experimental Lymphology—Branch of the Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Novosibirsk 630117, Russia;
| | - Marina V. Ovsyukova
- Department of Clinical Neuroscience, Behavior and Neurotechnologies, Institute of Physiology and Basic Medicine, Timakov St. 4, Novosibirsk 630117, Russia; (T.A.K.); (M.V.O.)
| | - Alexandra Shintyapina
- Institute of Molecular Biology and Biophysics, Federal Research Center, Timakov St. 2, Novosibirsk 630117, Russia;
| | - Vaclav Vetvicka
- Department of Pathology, University of Louisville, Louisville, KY 40292, USA
- Correspondence:
| |
Collapse
|
47
|
Li T, Hu S, Pang X, Wang J, Yin J, Li F, Wang J, Yang X, Xia B, Liu Y, Song W, Guo S. The marine-derived furanone reduces intracellular lipid accumulation in vitro by targeting LXRα and PPARα. J Cell Mol Med 2020; 24:3384-3398. [PMID: 31981312 PMCID: PMC7131916 DOI: 10.1111/jcmm.15012] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 12/30/2019] [Accepted: 01/10/2020] [Indexed: 12/22/2022] Open
Abstract
Recent studies have demonstrated that commercially available lipid-lowering drugs cause various side effects; therefore, searching for anti-hyperlipidaemic compounds with lower toxicity is a research hotspot. This study was designed to investigate whether the marine-derived compound, 5-hydroxy-3-methoxy-5-methyl-4-butylfuran-2(5H)-one, has an anti-hyperlipidaemic activity, and the potential underlying mechanism in vitro. Results showed that the furanone had weaker cytotoxicity compared to positive control drugs. In RAW 264.7 cells, the furanone significantly lowered ox-LDL-induced lipid accumulation (~50%), and its triglyceride (TG)-lowering effect was greater than that of liver X receptor (LXR) agonist T0901317. In addition, it significantly elevated the protein levels of peroxisome proliferator-activated receptors (PPARα) and ATP-binding cassette (ABC) transporters, which could be partially inhibited by LXR antagonists, GSK2033 and SR9243. In HepG2 cells, it significantly decreased oleic acid-induced lipid accumulation, enhanced the protein levels of low-density lipoprotein receptor (LDLR), ABCG5, ABCG8 and PPARα, and reduced the expression of sterol regulatory element-binding protein 2 (~32%). PPARα antagonists, GW6471 and MK886, could significantly inhibit the furanone-induced lipid-lowering effect. Furthermore, the furanone showed a significantly lower activity on the activation of the expression of lipogenic genes compared to T0901317. Taken together, the furanone exhibited a weak cytotoxicity but had powerful TC- and TG-lowering effects most likely through targeting LXRα and PPARα, respectively. These findings indicate that the furanone has a potential application for the treatment of dyslipidaemia.
Collapse
Affiliation(s)
- Ting Li
- Institute of Lipid Metabolism and AtherosclerosisSchool of PharmacyInnovative Drug Research CentreWeifang Medical UniversityWeifangChina
| | - Shu‐Mei Hu
- Institute of Lipid Metabolism and AtherosclerosisSchool of PharmacyInnovative Drug Research CentreWeifang Medical UniversityWeifangChina
| | - Xiao‐Yan Pang
- Institute of Lipid Metabolism and AtherosclerosisSchool of PharmacyInnovative Drug Research CentreWeifang Medical UniversityWeifangChina
| | - Jun‐feng Wang
- CAS Key Laboratory of Tropical Marine Bio‐resources and Ecology/Guangdong Key Laboratory of Marine Materia Medica/RNAM Center for Marine MicrobiologySouth China Sea Institute of OceanologyChinese Academy of SciencesGuangzhouChina
| | - Jia‐Yu Yin
- Institute of Lipid Metabolism and AtherosclerosisSchool of PharmacyInnovative Drug Research CentreWeifang Medical UniversityWeifangChina
| | - Fa‐Hui Li
- Institute of Lipid Metabolism and AtherosclerosisSchool of PharmacyInnovative Drug Research CentreWeifang Medical UniversityWeifangChina
| | - Jin Wang
- Institute of Lipid Metabolism and AtherosclerosisSchool of PharmacyInnovative Drug Research CentreWeifang Medical UniversityWeifangChina
| | - Xiao‐Qian Yang
- Institute of Lipid Metabolism and AtherosclerosisSchool of PharmacyInnovative Drug Research CentreWeifang Medical UniversityWeifangChina
| | - Bin Xia
- Institute of Lipid Metabolism and AtherosclerosisSchool of PharmacyInnovative Drug Research CentreWeifang Medical UniversityWeifangChina
| | - Yong‐Hong Liu
- CAS Key Laboratory of Tropical Marine Bio‐resources and Ecology/Guangdong Key Laboratory of Marine Materia Medica/RNAM Center for Marine MicrobiologySouth China Sea Institute of OceanologyChinese Academy of SciencesGuangzhouChina
| | - Wei‐Guo Song
- Institute of Lipid Metabolism and AtherosclerosisSchool of PharmacyInnovative Drug Research CentreWeifang Medical UniversityWeifangChina
| | - Shou‐Dong Guo
- Institute of Lipid Metabolism and AtherosclerosisSchool of PharmacyInnovative Drug Research CentreWeifang Medical UniversityWeifangChina
| |
Collapse
|
48
|
Fernando IPS, Ryu B, Ahn G, Yeo IK, Jeon YJ. Therapeutic potential of algal natural products against metabolic syndrome: A review of recent developments. Trends Food Sci Technol 2020. [DOI: 10.1016/j.tifs.2020.01.020] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
49
|
Xia B, Lin P, Ji Y, Yin J, Wang J, Yang X, Li T, Yang Z, Li F, Guo S. Ezetimibe promotes CYP7A1 and modulates PPARs as a compensatory mechanism in LDL receptor-deficient hamsters. Lipids Health Dis 2020; 19:24. [PMID: 32035489 PMCID: PMC7007651 DOI: 10.1186/s12944-020-1202-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 01/24/2020] [Indexed: 11/10/2022] Open
Abstract
Background The LDL-C lowering effect of ezetimibe has been attributed primarily to increased catabolism of LDL-C via up-regulation of LDL receptor (LDLR) and decreased cholesterol absorption. Recently, ezetimibe has been demonstrated to have reverse cholesterol transport (RCT) promoting effects in mice, hamsters and humans. However, the underlying mechanisms are still not clear. The aim of this study is to investigate whether ezetimibe improves RCT-related protein expression in LDLR−/− hamsters. Methods A high-fat diet was used to induce a human-like hyperlipidemia in LDLR−/− hamsters. Lipid profiles were assayed by commercially available kits, and the effects of ezetimibe on lipid metabolism-related protein expression were carried out via western blot. Results Our data demonstrated that ezetimibe administration significantly reduced plasma total cholesterol (~ 51.6% reduction, P < 0.01) and triglyceride (from ~ 884.1 mg/dL to ~ 277.3 mg/dL) levels in LDLR−/− hamsters fed a high-fat diet. Ezetimibe administration (25 mg/kg/d) significantly promoted the protein expression of cholesterol 7 alpha-hydroxylase A1 (CYP7A1), LXRβ and peroxisome proliferator-activated receptor (PPAR) γ; and down-regulated the protein expression of PPARα and PPARβ. However, it showed no significant effect on sterol regulatory element-binding protein (SREBP)-1c, SREBP-2, proprotein convertase subtilisin/kexin type 9 (PCSK9), Niemann-Pick C1-like 1 (NPC1L1), and ATP-biding cassette (ABC) G5/G8. Conclusion Ezetimibe may accelerate the transformation from cholesterol to bile acid via promoting CYP7A1 and thereby enhance RCT. As a compensatory mechanism of TG lowering, ezetimibe promoted the protein expression of PPARγ and decreased PPARα and β. These results are helpful in explaining the lipid-lowering effects of ezetimibe and the potential compensatory mechanisms.
Collapse
Affiliation(s)
- Bin Xia
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, 7166# Baotongxi Street, Weifang, 261053, Shandong Province, China
| | - Ping Lin
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, 7166# Baotongxi Street, Weifang, 261053, Shandong Province, China
| | - Yubin Ji
- College of Pharmacy Engineering Research Center for Medicine, Harbin University of Commerce, Harbin, 150076, China
| | - Jiayu Yin
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, 7166# Baotongxi Street, Weifang, 261053, Shandong Province, China.,College of Pharmacy Engineering Research Center for Medicine, Harbin University of Commerce, Harbin, 150076, China
| | - Jin Wang
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, 7166# Baotongxi Street, Weifang, 261053, Shandong Province, China
| | - Xiaoqian Yang
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, 7166# Baotongxi Street, Weifang, 261053, Shandong Province, China.,College of Pharmacy Engineering Research Center for Medicine, Harbin University of Commerce, Harbin, 150076, China
| | - Ting Li
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, 7166# Baotongxi Street, Weifang, 261053, Shandong Province, China.,College of Pharmacy Engineering Research Center for Medicine, Harbin University of Commerce, Harbin, 150076, China
| | - Zixun Yang
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, 7166# Baotongxi Street, Weifang, 261053, Shandong Province, China.,College of Pharmacy Engineering Research Center for Medicine, Harbin University of Commerce, Harbin, 150076, China
| | - Fahui Li
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, 7166# Baotongxi Street, Weifang, 261053, Shandong Province, China.
| | - Shoudong Guo
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, 7166# Baotongxi Street, Weifang, 261053, Shandong Province, China. .,College of Pharmacy Engineering Research Center for Medicine, Harbin University of Commerce, Harbin, 150076, China.
| |
Collapse
|
50
|
Zhao F, Liu Q, Cao J, Xu Y, Pei Z, Fan H, Yuan Y, Shen X, Li C. A sea cucumber (Holothuria leucospilota) polysaccharide improves the gut microbiome to alleviate the symptoms of type 2 diabetes mellitus in Goto-Kakizaki rats. Food Chem Toxicol 2019; 135:110886. [PMID: 31626838 DOI: 10.1016/j.fct.2019.110886] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 10/01/2019] [Accepted: 10/10/2019] [Indexed: 12/13/2022]
Abstract
Diabetes mellitus has become a worldwide concern in recent years. In this study, the effect of Holothuria leucospilota polysaccharide (HLP) on type 2 diabetes mellitus (T2DM) was investigated in Goto-Kakizaki (GK) rats. The results showed that HLP significantly improved glucose intolerance and regulated blood lipid and hormone levels (p < 0.05). Pathological analysis showed that HLP repaired the impairments of the pancreas and colon in diabetic rats. In addition, a high dose of HLP (200 mg/kg) significantly upregulated the gene expression of peroxisome proliferator-activated receptor-α (PPAR-α), peroxisome proliferator-activated receptor-γ (PPAR-γ), phosphoinositide 3-kinase (PI3K), protein kinase B (PKB/AKT), glucose transporter-4 (GLUT4) and anti-apoptotic (Bcl-2), and downregulated the mRNA levels of pro-apoptotic (Bax) and cluster of differentiation 36 (CD36) in diabetic rats (p < 0.05). Furthermore, HLP treatment increased the short-chain fatty acid-producing bacteria and decreased the opportunistic bacterial pathogen in the feces of diabetic rats. These results demonstrated that HLP has the potential to ameliorate T2DM in GK rats.
Collapse
Affiliation(s)
- Fuqiang Zhao
- Engineering Research Center of Utilization of Tropical Polysaccharide Resources of Ministry of Education, College of Food Science and Engineering, Hainan University, Haikou, 570228, China
| | - Qibing Liu
- School of Basic Medicine and Life Science, Hainan Medical University, Haikou, 571199, China
| | - Jun Cao
- Engineering Research Center of Utilization of Tropical Polysaccharide Resources of Ministry of Education, College of Food Science and Engineering, Hainan University, Haikou, 570228, China
| | - Yunsheng Xu
- Marine Food Engineering Technology Research Center of Hainan Province, Hainan Tropical Ocean University, Sanya, 572022, China
| | - Zhisheng Pei
- Marine Food Engineering Technology Research Center of Hainan Province, Hainan Tropical Ocean University, Sanya, 572022, China
| | - Haofei Fan
- School of Basic Medicine and Life Science, Hainan Medical University, Haikou, 571199, China
| | - Yiqiong Yuan
- Engineering Research Center of Utilization of Tropical Polysaccharide Resources of Ministry of Education, College of Food Science and Engineering, Hainan University, Haikou, 570228, China
| | - Xuanri Shen
- Engineering Research Center of Utilization of Tropical Polysaccharide Resources of Ministry of Education, College of Food Science and Engineering, Hainan University, Haikou, 570228, China; Collaborative Innovation Center of Seafood Deep Processing, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, 116034, China
| | - Chuan Li
- Engineering Research Center of Utilization of Tropical Polysaccharide Resources of Ministry of Education, College of Food Science and Engineering, Hainan University, Haikou, 570228, China; Collaborative Innovation Center of Seafood Deep Processing, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, 116034, China.
| |
Collapse
|