1
|
Zhang S, Feng X, Li CH, Zheng YM, Wang MY, Li JJ, Dai YP, Jing N, Zhou JW, Wang G. Mediator MED23 controls oligodendrogenesis and myelination by modulating Sp1/P300-directed gene programs. Cell Discov 2024; 10:102. [PMID: 39402028 PMCID: PMC11473658 DOI: 10.1038/s41421-024-00730-8] [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: 03/28/2023] [Accepted: 08/26/2024] [Indexed: 10/17/2024] Open
Abstract
Gaining the molecular understanding for myelination development and regeneration has been a long-standing goal in neurological research. Mutations in the transcription cofactor Mediator Med23 subunit are often associated with intellectual disability and white matter defects, although the precise functions and mechanisms of Mediator in myelination remain unclear. In this study, we generated a mouse model carrying an Med23Q649R mutation that has been identified in a patient with hypomyelination features. The MED23Q649R mouse model develops white matter thinning and cognitive decline, mimicking common clinical phenotypes. Further, oligodendrocyte-lineage specific Med23 knockout mice verified the important function of MED23 in regulating central nervous system myelination and postinjury remyelination. Utilizing the in vitro cellular differentiation assay, we found that the oligodendrocyte progenitor cells, either carrying the Q649R mutation or lacking Med23, exhibit significant deficits in their capacity to differentiate into mature oligodendrocytes. Gene profiling combined with reporter assays demonstrated that Mediator Med23 controls Sp1-directed gene programs related to oligodendrocyte differentiation and cholesterol metabolism. Integrative analysis demonstrated that Med23 modulates the P300 binding to Sp1-targeted genes, thus orchestrating the H3K27 acetylation and enhancer activation for the oligodendrocyte lineage progression. Collectively, our findings identified the critical role for the Mediator Med23 in oligodendrocyte fate determination and provide mechanistic insights into the myelination pathogenesis associated with MED23 mutations.
Collapse
Affiliation(s)
- Shuai Zhang
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xue Feng
- Laboratory Animal Resource Center, Fudan University, Shanghai, China
| | - Chong-Hui Li
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Yuan-Ming Zheng
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Zhongshan Hospital, Fudan University, Shanghai, China
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Meng-Ya Wang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Jun-Jie Li
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yun-Peng Dai
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Zhongshan Hospital, Fudan University, Shanghai, China
| | - Naihe Jing
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
- Guangzhou Laboratory, Guangzhou, Guangdong, China
| | - Jia-Wei Zhou
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Gang Wang
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
2
|
Rimmen EV, Mjøs SA, Søfteland E, Gudbrandsen OA. Intake of herring oil, but not of anchovy oil, resulted in a lower serum cholesterol concentration in male Zucker Diabetic Sprague Dawley rats. Br J Nutr 2024; 132:547-557. [PMID: 39297194 DOI: 10.1017/s0007114524001454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2024]
Abstract
Patients with type 2 diabetes have increased risks for dyslipidaemia and subsequently for developing vascular complications. A recent meta-analysis found that cetoleic acid (C22:1n-11) rich fish oils resulted in lower cholesterol concentration in rodents. The aim was to investigate the effect of consuming fish oils with or without cetoleic acid on serum cholesterol concentration in diabetic rats and to elucidate any effects on cholesterol metabolism. Eighteen male Zucker Diabetic Sprague Dawley rats were fed diets containing herring oil (HERO) or anchovy oil (ANCO) or a control diet with soyabean oil for 5 weeks. The HERO diet contained 0·70 % cetoleic acid, with no cetoleic acid in the ANCO diet. The HERO and ANCO diets contained 0·35 and 0·37 wt% EPA + DHA, respectively. Data were analysed using one-way ANOVA. The serum total cholesterol concentration was 14 % lower in the HERO group compared with ANCO and Control groups (P = 0·023). The HERO group had a higher faecal excretion of bile acids (P = 0·0036), but the cholesterol production in the liver, the hepatic secretion of VLDL and the liver's capacity to take up cholesterol were similar to controls. The ANCO diet did not affect the serum cholesterol concentration, but the hepatic cholesterol biosynthesis, the clearance of lipoprotein cholesterol and the excretion of bile acids in faeces were higher than in the Control group. To conclude, consumption of herring oil, but not of anchovy oil, led to a lower cholesterol concentration in a type 2 diabetes rat model.
Collapse
Affiliation(s)
- Eira V Rimmen
- Dietary Protein Research Group, Centre for Nutrition, Department of Clinical Medicine, University of Bergen, Bergen 5021, Norway
| | - Svein Are Mjøs
- Department of Chemistry, University of Bergen, 5020 Bergen, Norway
| | - Eirik Søfteland
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Oddrun A Gudbrandsen
- Dietary Protein Research Group, Centre for Nutrition, Department of Clinical Medicine, University of Bergen, Bergen 5021, Norway
| |
Collapse
|
3
|
Bouchard J, Raj P, Yu L, Sobhi B, Malalgoda M, Malunga L, Netticadan T, Joseph Thandapilly S. Oat protein modulates cholesterol metabolism and improves cardiac systolic function in high fat, high sucrose fed rats. Appl Physiol Nutr Metab 2024; 49:738-750. [PMID: 38477294 DOI: 10.1139/apnm-2023-0440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2024]
Abstract
Oats are recognized to provide many health benefits that are mainly associated with its dietary fibre, β-glucan. However, the protein derived from oats is largely understudied with respect to its ability to maintain health and attenuate risk factors of chronic diseases. The goal of the current study was to investigate the metabolic effects of oat protein consumption in lieu of casein as the protein source in high fat, high sucrose (HF/HS) fed Wistar rats. Four-week-old rats were divided into three groups and were fed three different experimental diets: a control diet with casein as the protein source, an HF/HS diet with casein, or an HF/HS diet with oat protein for 16 weeks. Heart structure and function were determined by echocardiography. Blood pressure measurements, an oral glucose tolerance test, and markers of cholesterol metabolism, oxidative stress, inflammation, and liver and kidney damage were also performed. Our study results show that incorporation of oat protein in the diet was effective in preserving systolic heart function in HF/HS fed rats. Oat protein significantly reduced serum total and low-density lipoprotein cholesterol levels. Furthermore, oat protein normalized liver HMG-CoAR activity, which, to our knowledge, is the first time this has been reported in the literature. Therefore, our research suggests that oat protein can provide hypocholesterolemic and cardioprotective benefits in a diet-induced model of metabolic syndrome.
Collapse
Affiliation(s)
- Jenny Bouchard
- Morden Research and Development Centre, Agriculture and Agri-Food Canada, Morden, MB R6M 1Y5, Canada
- Canadian Centre for Agri-Food Research in Health and Medicine, Winnipeg, MB R2H 2A6, Canada
- Richardson Center for Food Technology and Research, Winnipeg, MB R3T 2N2, Canada
- Department of Food and Human Nutritional Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Pema Raj
- Canadian Centre for Agri-Food Research in Health and Medicine, Winnipeg, MB R2H 2A6, Canada
| | - Liping Yu
- Morden Research and Development Centre, Agriculture and Agri-Food Canada, Morden, MB R6M 1Y5, Canada
- Canadian Centre for Agri-Food Research in Health and Medicine, Winnipeg, MB R2H 2A6, Canada
| | - Babak Sobhi
- Morden Research and Development Centre, Agriculture and Agri-Food Canada, Morden, MB R6M 1Y5, Canada
- Richardson Center for Food Technology and Research, Winnipeg, MB R3T 2N2, Canada
| | - Maneka Malalgoda
- Department of Food and Human Nutritional Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Lovemore Malunga
- Morden Research and Development Centre, Agriculture and Agri-Food Canada, Morden, MB R6M 1Y5, Canada
- Canadian Centre for Agri-Food Research in Health and Medicine, Winnipeg, MB R2H 2A6, Canada
- Richardson Center for Food Technology and Research, Winnipeg, MB R3T 2N2, Canada
- Department of Food and Human Nutritional Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Thomas Netticadan
- Morden Research and Development Centre, Agriculture and Agri-Food Canada, Morden, MB R6M 1Y5, Canada
- Canadian Centre for Agri-Food Research in Health and Medicine, Winnipeg, MB R2H 2A6, Canada
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Sijo Joseph Thandapilly
- Morden Research and Development Centre, Agriculture and Agri-Food Canada, Morden, MB R6M 1Y5, Canada
- Canadian Centre for Agri-Food Research in Health and Medicine, Winnipeg, MB R2H 2A6, Canada
- Richardson Center for Food Technology and Research, Winnipeg, MB R3T 2N2, Canada
- Department of Food and Human Nutritional Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| |
Collapse
|
4
|
Swan J, Szabó Z, Peters J, Kummu O, Kemppi A, Rahtu-Korpela L, Konzack A, Hakkola J, Pasternack A, Ritvos O, Kerkelä R, Magga J. Inhibition of activin receptor 2 signalling ameliorates metabolic dysfunction-associated steatotic liver disease in western diet/L-NAME induced cardiometabolic disease. Biomed Pharmacother 2024; 175:116683. [PMID: 38705130 DOI: 10.1016/j.biopha.2024.116683] [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: 01/09/2024] [Revised: 04/19/2024] [Accepted: 04/29/2024] [Indexed: 05/07/2024] Open
Abstract
OBJECTIVE Blockade of activin 2 receptor (ACVR2) signaling has been shown to improve insulin sensitivity and aid in weight loss. Inhibition of ACVR2 signaling restores cardiac function in multiple heart failure models. However, its potential in the treatment of obesity-related cardiometabolic disease remains unknown. Here, we investigated targeting ACVR2 signaling in cardiometabolic disease manifested with metabolic dysfunction-associated steatotic liver disease (MASLD). METHODS Mice were fed a high-fat, high-sugar diet combined with the administration of nitric oxide synthase inhibitor L-NAME in drinking water, which causes hypertensive stress. For the last eight weeks, the mice were treated with the soluble ACVR2B decoy receptor (sACVR2B-Fc). RESULTS sACVR2B-Fc protected against the development of comorbidities associated with cardiometabolic disease. This was most pronounced in the liver where ACVR2 blockade attenuated the development of MASLD including cessation of pro-fibrotic activation. It also significantly reduced total plasma cholesterol levels, impeded brown adipose tissue whitening, and improved cardiac diastolic function. In vitro, ACVR2 ligands activin A, activin B and GDF11 induced profibrotic signaling and the proliferation of human cardiac fibroblasts. CONCLUSIONS Blockade of ACVR2B exerts broad beneficial effects for therapy of cardiometabolic disease. By reducing obesity, ameliorating cardiovascular deterioration and restraining MASLD, blockade of ACVR2B signaling proves a potential target in MASLD and its comorbidities.
Collapse
Affiliation(s)
- Julia Swan
- Research Unit of Biomedicine and Internal Medicine, University of Oulu, Aapistie 5, Oulu 90220, Finland; Biocenter Oulu, University of Oulu, Aapistie 5, Oulu 90220, Finland.
| | - Zoltán Szabó
- Research Unit of Biomedicine and Internal Medicine, University of Oulu, Aapistie 5, Oulu 90220, Finland
| | - Juliana Peters
- Research Unit of Biomedicine and Internal Medicine, University of Oulu, Aapistie 5, Oulu 90220, Finland
| | - Outi Kummu
- Research Unit of Biomedicine and Internal Medicine, University of Oulu, Aapistie 5, Oulu 90220, Finland; Biocenter Oulu, University of Oulu, Aapistie 5, Oulu 90220, Finland; Medical Research Centre Oulu, Oulu University Hospital and University of Oulu, Aapistie 5, Oulu 90220, Finland
| | - Anna Kemppi
- Research Unit of Biomedicine and Internal Medicine, University of Oulu, Aapistie 5, Oulu 90220, Finland
| | - Lea Rahtu-Korpela
- Research Unit of Biomedicine and Internal Medicine, University of Oulu, Aapistie 5, Oulu 90220, Finland
| | - Anja Konzack
- Research Unit of Biomedicine and Internal Medicine, University of Oulu, Aapistie 5, Oulu 90220, Finland; Biocenter Oulu, University of Oulu, Aapistie 5, Oulu 90220, Finland; Medical Research Centre Oulu, Oulu University Hospital and University of Oulu, Aapistie 5, Oulu 90220, Finland
| | - Jukka Hakkola
- Research Unit of Biomedicine and Internal Medicine, University of Oulu, Aapistie 5, Oulu 90220, Finland; Biocenter Oulu, University of Oulu, Aapistie 5, Oulu 90220, Finland; Medical Research Centre Oulu, Oulu University Hospital and University of Oulu, Aapistie 5, Oulu 90220, Finland
| | - Arja Pasternack
- Department of Physiology, Faculty of Medicine, University of Helsinki, Haartmaninkatu 8, Helsinki 00014, Finland
| | - Olli Ritvos
- Department of Physiology, Faculty of Medicine, University of Helsinki, Haartmaninkatu 8, Helsinki 00014, Finland
| | - Risto Kerkelä
- Research Unit of Biomedicine and Internal Medicine, University of Oulu, Aapistie 5, Oulu 90220, Finland; Biocenter Oulu, University of Oulu, Aapistie 5, Oulu 90220, Finland; Medical Research Centre Oulu, Oulu University Hospital and University of Oulu, Aapistie 5, Oulu 90220, Finland
| | - Johanna Magga
- Research Unit of Biomedicine and Internal Medicine, University of Oulu, Aapistie 5, Oulu 90220, Finland; Biocenter Oulu, University of Oulu, Aapistie 5, Oulu 90220, Finland.
| |
Collapse
|
5
|
Abdelhamid MS, Sherif MH, Abaza HR, El‐Maghraby LMM, Watad SH, Awad AE. Zingiber officinale extract maximizes the efficacy of simvastatin as a hypolipidemic drug in obese male rats. Food Sci Nutr 2024; 12:1940-1954. [PMID: 38455204 PMCID: PMC10916669 DOI: 10.1002/fsn3.3889] [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: 07/31/2023] [Revised: 11/14/2023] [Accepted: 11/21/2023] [Indexed: 03/09/2024] Open
Abstract
Obesity became a serious public health problem with enormous socioeconomic implications among the Egyptian population. The present investigation aimed to explore the efficacy of Zingiber officinale extract as a hypolipidemic agent combined with the commercially well-known anti-obesity drug simvastatin in obese rats. Thirty-five male Wister rats were randomly divided into five groups as follows: group I received a standard balanced diet for ten weeks; high-fat diet was orally administered to rats in groups II-V for ten weeks. From the fifth week to the tenth week, group III orally received simvastatin (40 mg/kg B.W.), group IV orally received Z. officinale root extract (400 mg/kg B.W.), and group V orally received simvastatin (20 mg/kg B.W.) plus Z. officinale extract (200 mg/kg B.W.) separately. Liver and kidney function tests, lipid profiles, serum glucose, insulin, and leptin were determined. Quantitative RT-PCR analysis of PPAR-γ, iNOS, HMG-CoA reductase, and GLUT-4 genes was carried out. Caspase 3 was estimated in liver and kidney tissues immunohistochemically. Liver and kidney tissues were examined histologically. The administration of Z. officinale extract plus simvastatin to high-fat diet-fed rats caused a significant reduction in the expression of HMG-coA reductase and iNOS by 41.81% and 88.05%, respectively, compared to highfat diet (HFD)-fed rats that received simvastatin only. Otherwise, a significant increase was noticed in the expression of PPAR-γ and GLUT-4 by 33.3% and 138.81%, respectively, compared to those that received simvastatin only. Immunohistochemistry emphasized that a combination of Z. officinale extract plus simvastatin significantly suppressed caspase 3 in the hepatic tissue of high-fat diet-fed rats. Moreover, the best results of lipid profile indices and hormonal indicators were obtained when rats received Z. officinale extract plus simvastatin. Z. officinale extract enhanced the efficiency of simvastatin as a hypolipidemic drug in obese rats due to the high contents of flavonoid and phenolic ingredients.
Collapse
Affiliation(s)
| | | | - Hazem R. Abaza
- Biochemistry Department, Faculty of ScienceZagazig UniversityZagazigEgypt
| | | | - Shimaa H. Watad
- Biochemistry Department, Faculty of ScienceZagazig UniversityZagazigEgypt
| | - Ahmed E. Awad
- Agricultural Biochemistry Department, Faculty of AgricultureZagazig UniversityZagazigEgypt
| |
Collapse
|
6
|
Kumar R, Chhillar N, Gupta DS, Kaur G, Singhal S, Chauhan T. Cholesterol Homeostasis, Mechanisms of Molecular Pathways, and Cardiac Health: A Current Outlook. Curr Probl Cardiol 2024; 49:102081. [PMID: 37716543 DOI: 10.1016/j.cpcardiol.2023.102081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 09/11/2023] [Indexed: 09/18/2023]
Abstract
The metabolism of lipoproteins, which regulate the transit of the lipid to and from tissues, is crucial to maintaining cholesterol homeostasis. Cardiac remodeling is referred to as a set of molecular, cellular, and interstitial changes that, following injury, affect the size, shape, function, mass, and geometry of the heart. Acetyl coenzyme A (acetyl CoA), which can be made from glucose, amino acids, or fatty acids, is the precursor for the synthesis of cholesterol. In this article, the authors explain concepts behind cardiac remodeling, its clinical ramifications, and the pathophysiological roles played by numerous various components, such as cell death, neurohormonal activation, oxidative stress, contractile proteins, energy metabolism, collagen, calcium transport, inflammation, and geometry. The levels of cholesterol are traditionally regulated by 2 biological mechanisms at the transcriptional stage. First, the SREBP transcription factor family regulates the transcription of crucial rate-limiting cholesterogenic and lipogenic proteins, which in turn limits cholesterol production. Immune cells become activated, differentiated, and divided, during an immune response with the objective of eradicating the danger signal. In addition to creating ATP, which is used as energy, this process relies on metabolic reprogramming of both catabolic and anabolic pathways to create metabolites that play a crucial role in regulating the response. Because of changes in signal transduction, malfunction of the sarcoplasmic reticulum and sarcolemma, impairment of calcium handling, increases in cardiac fibrosis, and progressive loss of cardiomyocytes, oxidative stress appears to be the primary mechanism that causes the transition from cardiac hypertrophy to heart failure. De novo cholesterol production, intestinal cholesterol absorption, and biliary cholesterol output are consequently crucial processes in cholesterol homeostasis. In the article's final section, the pharmacological management of cardiac remodeling is explored. The route of treatment is explained in different steps: including, promising, and potential strategies. This chapter offers a brief overview of the history of the study of cholesterol absorption as well as the different potential therapeutic targets.
Collapse
Affiliation(s)
| | - Neelam Chhillar
- Deparetment of Biochemistry, School of Medicine, DY Patil University, Navi Mumbai, India
| | - Dhruv Sanjay Gupta
- Department of Pharmacology, SPP School of Pharmacy & Technology Management, SVKM's NMIMS, Mumbai, India
| | - Ginpreet Kaur
- Department of Pharmacology, SPP School of Pharmacy & Technology Management, SVKM's NMIMS, Mumbai, India
| | - Shailey Singhal
- Cluster of Applied Sciences, University of Petroleum and Energy Studies, Dehradun, India
| | - Tanya Chauhan
- Division of Forensic Biology, National Forensic Sciences University, Delhi Campus (LNJN NICFS) Delhi, India
| |
Collapse
|
7
|
Itkonen A, Hakkola J, Rysä J. Adverse outcome pathway for pregnane X receptor-induced hypercholesterolemia. Arch Toxicol 2023; 97:2861-2877. [PMID: 37642746 PMCID: PMC10504106 DOI: 10.1007/s00204-023-03575-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 08/02/2023] [Indexed: 08/31/2023]
Abstract
Pharmaceuticals and environmental contaminants contribute to hypercholesterolemia. Several chemicals known to cause hypercholesterolemia, activate pregnane X receptor (PXR). PXR is a nuclear receptor, classically identified as a sensor of chemical environment and regulator of detoxification processes. Later, PXR activation has been shown to disrupt metabolic functions such as lipid metabolism and recent findings have shown PXR activation to promote hypercholesterolemia through multiple mechanisms. Hypercholesterolemia is a major causative risk factor for atherosclerosis and greatly promotes global health burden. Metabolic disruption by PXR activating chemicals leading to hypercholesterolemia represents a novel toxicity pathway of concern and requires further attention. Therefore, we constructed an adverse outcome pathway (AOP) by collecting the available knowledge considering the molecular mechanisms for PXR-mediated hypercholesterolemia. AOPs are tools of modern toxicology for systematizing mechanistic knowledge to assist health risk assessment of chemicals. AOPs are formalized and structured linear concepts describing a link between molecular initiating event (MIE) and adverse outcome (AO). MIE and AO are connected via key events (KE) through key event relationships (KER). We present a plausible route of how PXR activation (MIE) leads to hypercholesterolemia (AO) through direct regulation of cholesterol synthesis and via activation of sterol regulatory element binding protein 2-pathway.
Collapse
Affiliation(s)
- Anna Itkonen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, 70211, Kuopio, Finland
| | - Jukka Hakkola
- Research Unit of Biomedicine and Internal Medicine, Biocenter Oulu, Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Jaana Rysä
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, 70211, Kuopio, Finland.
| |
Collapse
|
8
|
Kim N, Lee S, Jung EJ, Jung EY, Chang UJ, Jin CM, Suh HJ, Choi HS. Yeast-Hydrolysate-Derived 1-Methyl-1,2,3,4-tetrahydro-β-carboline-3-carboxylic Acid Inhibits Fat Accumulation during Adipocyte Differentiation. Foods 2023; 12:3466. [PMID: 37761175 PMCID: PMC10528377 DOI: 10.3390/foods12183466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 09/14/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
This study aimed to investigate the impact of yeast hydrolysate (YH) on lipogenesis, elucidate its mechanistic action, and identify the active compounds responsible for its anti-adipogenic effects. YH (2 mg/mL) significantly reduced Oil Red O-stained lipids. YH (2 mg/mL) also downregulated C/EBPβ and upregulated KLF2, both of which are early adipogenic factors. Moreover, YH (2 mg/mL) decreased C/EBPα, PPARγ, FABP4, FAS, ACC, and HMGCR mRNA expression. Additionally, YH significantly downregulated SEBP1c and SREBP2 and their target genes, which govern fatty acid and cholesterol metabolism; however, 2 mg/mL YH had a greater suppressive effect on SREBP1c than on SREBP2. YH (2 mg/mL) also significantly reduced the mRNA level of G6PD and malic enzyme, which are enzymes that synthesize NADPH for lipid synthesis, compared with the control. Furthermore, 1-methyl-1,2,3,4-tetrahydro-β-carboline-3-carboxylic acid (MTCA) was identified as the active compound with anti-adipogenic effects using solvent fractionation and chromatographic analysis of YH, and 1.1 μg/mL MTCA significantly downregulated SREBP1c/SREBP2 mRNAs by 47.8% and 69.2%, respectively, along with the target genes FAS, ACC, and HMGCR by 79.0%, 77.0%, and 40.9%, respectively. Collectively, YH effectively suppressed adipogenic lipid storage by downregulating SREBP- and NADPH-synthesizing genes. These findings suggest that YH containing MTCA has the potential to act as an anti-obesity agent.
Collapse
Affiliation(s)
- Nari Kim
- Department of Integrated Biomedical and Life Science, Graduate School, Korea University, Seoul 02841, Republic of Korea; (N.K.); (S.L.)
| | - Sekyung Lee
- Department of Integrated Biomedical and Life Science, Graduate School, Korea University, Seoul 02841, Republic of Korea; (N.K.); (S.L.)
- Transdisciplinary Major in Learning Health Systems, Department of Healthcare Sciences, Graduate School, Korea University, Seoul 02841, Republic of Korea
| | - Eun-Jin Jung
- Department of Food and Biotechnology, Korea University, Sejong 30019, Republic of Korea;
| | - Eun Young Jung
- Department of Home Economic Education, Jeonju University, Jeonju 55069, Republic of Korea;
| | - Un-Jae Chang
- Department of Food and Nutrition, Dongduk Women’s University, Seoul 02748, Republic of Korea;
| | - Cheng-Min Jin
- Analysis and Research Department, NeuroVIS, Inc., Hwaseong-si 18469, Republic of Korea;
| | - Hyung Joo Suh
- Department of Integrated Biomedical and Life Science, Graduate School, Korea University, Seoul 02841, Republic of Korea; (N.K.); (S.L.)
- Transdisciplinary Major in Learning Health Systems, Department of Healthcare Sciences, Graduate School, Korea University, Seoul 02841, Republic of Korea
| | - Hyeon-Son Choi
- Department of Food Nutrition, Sangmyung University, Hongjimun 2-Gil 20, Jongno-Gu, Seoul 03016, Republic of Korea
| |
Collapse
|
9
|
Iyer DR, Venkatraman J, Tanguy E, Vitale N, Mahapatra NR. Chromogranin A and its derived peptides: potential regulators of cholesterol homeostasis. Cell Mol Life Sci 2023; 80:271. [PMID: 37642733 PMCID: PMC11072126 DOI: 10.1007/s00018-023-04908-3] [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: 03/03/2023] [Revised: 08/02/2023] [Accepted: 08/03/2023] [Indexed: 08/31/2023]
Abstract
Chromogranin A (CHGA), a member of the granin family of proteins, has been an attractive therapeutic target and candidate biomarker for several cardiovascular, neurological, and inflammatory disorders. The prominence of CHGA stems from the pleiotropic roles of several bioactive peptides (e.g., catestatin, pancreastatin, vasostatins) generated by its proteolytic cleavage and by their wide anatomical distribution. These peptides are emerging as novel modulators of cardiometabolic diseases that are often linked to high blood cholesterol levels. However, their impact on cholesterol homeostasis is poorly understood. The dynamic nature of cholesterol and its multitudinous roles in almost every aspect of normal body function makes it an integral component of metabolic physiology. A tightly regulated coordination of cholesterol homeostasis is imperative for proper functioning of cellular and metabolic processes. The deregulation of cholesterol levels can result in several pathophysiological states. Although studies till date suggest regulatory roles for CHGA and its derived peptides on cholesterol levels, the mechanisms by which this is achieved still remain unclear. This review aims to aggregate and consolidate the available evidence linking CHGA with cholesterol homeostasis in health and disease. In addition, we also look at common molecular regulatory factors (viz., transcription factors and microRNAs) which could govern the expression of CHGA and genes involved in cholesterol homeostasis under basal and pathological conditions. In order to gain further insights into the pathways mediating cholesterol regulation by CHGA/its derived peptides, a few prospective signaling pathways are explored, which could act as primers for future studies.
Collapse
Affiliation(s)
- Dhanya R Iyer
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, 600036, India
| | - Janani Venkatraman
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, 600036, India
| | - Emeline Tanguy
- Institut des Neurosciences Cellulaires et Intégratives, CNRS UPR 3212 and Université de Strasbourg, 5 Rue Blaise Pascal, 67000, Strasbourg, France
| | - Nicolas Vitale
- Institut des Neurosciences Cellulaires et Intégratives, CNRS UPR 3212 and Université de Strasbourg, 5 Rue Blaise Pascal, 67000, Strasbourg, France.
| | - Nitish R Mahapatra
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, 600036, India.
| |
Collapse
|
10
|
Kim MH, Lee EJ, Kim SJ, Jung YJ, Park WJ, Park I. Macrophage inhibitory cytokine-1 aggravates diet-induced gallstone formation via increased ABCG5/ABCG8 expression. PLoS One 2023; 18:e0287146. [PMID: 37310967 DOI: 10.1371/journal.pone.0287146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 05/30/2023] [Indexed: 06/15/2023] Open
Abstract
Macrophage inhibitory cytokine 1 (MIC-1), which is overproduced in various human cancers and associated with cachexia, acts on the hypothalamus to suppress appetite and reduce body weight. We investigated the mechanisms through which MIC-1 affects bile acid metabolism and gallstone formation, which are poorly understood. Over 6 weeks, male C57BL/6 mice fed either standard chow or a lithogenic diet were intraperitoneally injected with phosphate-buffered saline (PBS) or MIC-1 (200 μg/kg/week). Among lithogenic diet-fed mice, MIC-1 treatment resulted in increased gallstone formation compared with PBS treatment. Compared with PBS treatment, MIC-1 treatment decreased hepatic cholesterol and bile acid levels and reduced expression of HMG-CoA reductase (HMGCR), the master cholesterol metabolism regulator sterol regulatory element-binding protein 2, cholesterol 7α-hydroxylase (CYP7A1), mitochondrial sterol 27-hydroxylase, and oxysterol 7α-hydroxylase. Compared with PBS treatment, MIC-1 treatment had no effect on small heterodimer partner, farnesoid X receptor, or pregnane X receptor expression, and extracellular signal-related kinase and c-Jun N-terminal kinase phosphorylation decreased, suggesting that these factors do not contribute to the MIC-1-induced reduction in CYP7A1 expression. Compared with PBS treatment, MIC-1 treatment increased AMP-activated protein kinase (AMPK) phosphorylation. Treatment with the AMPK activator 5-aminoimidazole-4-carboxamide ribonucleoside (AICAR) reduced CYP7A1 and HMGCR expression, whereas the AMPK inhibitor Compound C reversed MIC-1-induced reductions in CYP7A1 and HMGCR expression. Furthermore, in MIC-1-treated mice, total biliary cholesterol levels increased together with increased ATP-binding cassette subfamily G (ABCG)5 and ABCG8 expression. Compared with PBS treatment, MIC-1 treatment did not affect expression of liver X receptors α and β, liver receptor homolog 1, hepatocyte nuclear factor 4α, or NR1I3 (also known as constitutive androstane receptor), which are upstream of ABCG5/8; however, MIC-1 treatment increased ABCG5/8 expression and promoter activities. Our study indicates that MIC-1 influences gallstone formation by increasing AMPK phosphorylation, reducing CYP7A1 and HMGCR expression, and increasing ABCG5 and ABCG8 expression.
Collapse
Affiliation(s)
- Min Hee Kim
- Department of Biochemistry, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Eun-Ji Lee
- Department of Biochemistry, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Su-Jeong Kim
- Department of Biochemistry, Chung-Ang University College of Medicine, Seoul, Republic of Korea
| | - Yun-Jae Jung
- Department of Microbiology, Lee Gil Ya Cancer and Diabetes Institute, College of Medicine, Gachon University, Incheon, Republic of Korea
- Department of Health Science and Technology, Gachon Advanced Institute for Health Science & Technology, Gachon University, Incheon, Republic of Korea
| | - Woo-Jae Park
- Department of Biochemistry, Chung-Ang University College of Medicine, Seoul, Republic of Korea
| | - Inkeun Park
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
11
|
Zhang ZF, Song TT, Chen JF, Lv GY. Recovery of a hypolipidemic polysaccharide from artificially cultivated Sanghuangporus vaninii with an effective method. Front Nutr 2023; 9:1095556. [PMID: 36712537 PMCID: PMC9880258 DOI: 10.3389/fnut.2022.1095556] [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: 11/11/2022] [Accepted: 12/28/2022] [Indexed: 01/15/2023] Open
Abstract
In this study, an effective method was developed to extract the polysaccharide from Sanghuangporus vaninii (PFSV) by destroying the cell wall. Box-Behnken design was employed to determine the optimal processing conditions as follows: processing temperature (80°C), processing time (0.81 h) and amount of HCl (1.5 ml). Under these conditions, the yield of PFSV reached 5.94 ± 0.16%. The purified polysaccharide (PFSV-2) was found to be a hetero-polysaccharide with an average molecular weight of 20.377 kDa. The backbone of PFSV-2 was composed of an →6)-α-Galp-(1→ and →2,6)-β-Manp-(1→ and →2)-α-Fucp-(1→ and was branched of t-α-Manp-(1→ at position 2 of residue B. PFSV-2 showed hypolipidemic activity by decreasing lipid accumulation and the levels of total cholesterol and triglycerides in zebrafish larvae. Furthermore, PFSV-2 downregulated the pparg, fasn, and HMGCRb genes and upregulated the pparab and acaca genes. These findings suggested PFSV-2 may be a promising candidate in lipid regulation therapy.
Collapse
|
12
|
Nanoparticulate System for the Transdermal Delivery of Catechin as an Antihypercholesterol: In Vitro and In Vivo Evaluations. Pharmaceuticals (Basel) 2022; 15:ph15091142. [PMID: 36145363 PMCID: PMC9505170 DOI: 10.3390/ph15091142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/22/2022] [Accepted: 09/01/2022] [Indexed: 11/16/2022] Open
Abstract
Gambir (Uncaria Gambir, Roxb.) contains catechins that is often empirically used to treat various diseases. Catechins can reduce cholesterol levels by inhibiting coenzyme HMG-CoA reductase that plays a role in cholesterol metabolism. Research has been carried out covering the optimization of transethosomal catechins, the formulation of Transethosomal Catechin Gel (TCG) and Non-Transethosomal Catechin Gel (NTCG), which were then tested for catechin permeation from these gel preparations in vitro using Franz’s diffusion cell with PTFE membranes. The anti-hypercholesterol activity test was carried out with Simvastatin orally as a positive control using 25 male Wistar rats (Rattus norvegicus). The catechin transetosomes have a size of 176.1 ± 5.8 nm, Zeta potential −11.6 ± 5.28, and Entrapment Efficacy of 96.77% ± 0.05. The result of cumulative catechins that permeated from TCG and NTCG were and 172.454 ± 5.287 and 112.741 ± 2.241 μg respectively. Permeation test graphs showed similar permeation and flux profiles. TCG can reduce total cholesterol and LDL (Low Density Lipoprotein) values in rats by 39.77% and 51.52% respectively during 14 days of use.
Collapse
|
13
|
Smith CJ, Sinnott-Armstrong N, Cichońska A, Julkunen H, Fauman EB, Würtz P, Pritchard JK. Integrative analysis of metabolite GWAS illuminates the molecular basis of pleiotropy and genetic correlation. eLife 2022; 11:e79348. [PMID: 36073519 PMCID: PMC9536840 DOI: 10.7554/elife.79348] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 09/06/2022] [Indexed: 11/15/2022] Open
Abstract
Pleiotropy and genetic correlation are widespread features in genome-wide association studies (GWAS), but they are often difficult to interpret at the molecular level. Here, we perform GWAS of 16 metabolites clustered at the intersection of amino acid catabolism, glycolysis, and ketone body metabolism in a subset of UK Biobank. We utilize the well-documented biochemistry jointly impacting these metabolites to analyze pleiotropic effects in the context of their pathways. Among the 213 lead GWAS hits, we find a strong enrichment for genes encoding pathway-relevant enzymes and transporters. We demonstrate that the effect directions of variants acting on biology between metabolite pairs often contrast with those of upstream or downstream variants as well as the polygenic background. Thus, we find that these outlier variants often reflect biology local to the traits. Finally, we explore the implications for interpreting disease GWAS, underscoring the potential of unifying biochemistry with dense metabolomics data to understand the molecular basis of pleiotropy in complex traits and diseases.
Collapse
Affiliation(s)
- Courtney J Smith
- Department of Genetics, Stanford University School of MedicineStanfordUnited States
| | - Nasa Sinnott-Armstrong
- Department of Genetics, Stanford University School of MedicineStanfordUnited States
- Herbold Computational Biology Program, Fred Hutchinson Cancer Research CenterSeattleUnited States
| | | | | | - Eric B Fauman
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development and MedicalCambridgeUnited States
| | | | - Jonathan K Pritchard
- Department of Genetics, Stanford University School of MedicineStanfordUnited States
- Department of Biology, Stanford UniversityStanfordUnited States
| |
Collapse
|
14
|
Huang Y, Li S, Han Z, Du J, Liu X, Zhu Z, Zheng L, Han S, Shi H, Wang X, Liu X, Jiang Z, Li Y, Li X, Gu X, Han D, Li D. Tyrosol Ameliorates Liver Inflammatory Response in a Mouse Model of Nonalcoholic Fatty Liver Disease (NFALD) by Regulating JAK1/STAT3. Nat Prod Commun 2022. [DOI: 10.1177/1934578x221111033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is becoming one of the major health issues globally. NAFLD is usually associated with obesity and other metabolic syndromes, and there is no specific cure to address this issue so far. Featured by lipid accumulation in hepatocytes, and later progressing to fibrosis, inflammatory responses are involved in the various levels of the pathological changes. In the present study, we added a natural compound, tyrosol, which in our previous study had demonstrated anti-inflammatory properties, to a high-fat diet-induced NAFLD mouse model and investigated whether tyrosol could mitigate the liver damage by attenuating the inflammation response. The treatment with tyrosol significantly improved the liver function and decreased the fasting glucose level in NAFLD mice. Morphologically, our results showed that tyrosol could reduce the fat deposition and lipid droplets accumulation in liver tissue. The key regulating factors, JAK1 and STAT3, were increased in NAFLD mice, but tyrosol treatment could effectively prevent the upregulation. The expression levels of pro-inflammatory cytokine genes in liver tissue of the NAFLD mice were upregulated, which could be effectively prevented by the treatment with tyrosol. Overall, in the high-fat diet-induced NAFLD mouse model, tyrosol could improve the liver function and, more importantly, ameliorate the inflammatory response triggered by the high-fat diet. Although our data here are consistent with the previous report that tyrosol could exert beneficial effects on the NAFLD animal model, we also provide solid evidence that tyrosol is able to conquer the inflammatory response in liver, which is related to the high-fat-diet feeding. Tyrosol could be a promising candidate for the treatment of NAFLD in the future.
Collapse
Affiliation(s)
- Yinuo Huang
- Department of Gastroenterology, Beijing FengTai You'anmen Hospital, Beijing, China
| | - Shuai Li
- Department of Gastroenterology, Beijing FengTai You'anmen Hospital, Beijing, China
| | - Zifei Han
- School of Statistics, University of International Business and Economics, Beijing, China
| | - Juan Du
- Department of Gastroenterology, Beijing FengTai You'anmen Hospital, Beijing, China
| | - Xin Liu
- Department of Gastroenterology, Beijing FengTai You'anmen Hospital, Beijing, China
| | - Zhiyuan Zhu
- Department of Gastroenterology, Beijing FengTai You'anmen Hospital, Beijing, China
| | - Lixia Zheng
- Department of Gastroenterology, Beijing FengTai You'anmen Hospital, Beijing, China
| | - Suyan Han
- Department of Gastroenterology, Beijing FengTai You'anmen Hospital, Beijing, China
| | - Hongbo Shi
- Department of Gastroenterology, Beijing FengTai You'anmen Hospital, Beijing, China
| | - Xiaojuan Wang
- Department of Gastroenterology, Beijing FengTai You'anmen Hospital, Beijing, China
| | - Xinmei Liu
- Department of Gastroenterology, Beijing FengTai You'anmen Hospital, Beijing, China
| | - Zhihong Jiang
- Department of Gastroenterology, Beijing FengTai You'anmen Hospital, Beijing, China
| | - Yuxiang Li
- Department of Gastroenterology, Beijing FengTai You'anmen Hospital, Beijing, China
| | - Xiaoli Li
- Department of Gastroenterology, Beijing FengTai You'anmen Hospital, Beijing, China
| | - Xin Gu
- Department of Gastroenterology, Beijing FengTai You'anmen Hospital, Beijing, China
| | - Dazheng Han
- The First Affiliated Hospital, Henan University, Kaifeng, China
| | - Donghai Li
- Department of Gastroenterology, Beijing FengTai You'anmen Hospital, Beijing, China
| |
Collapse
|
15
|
Inflammation Induced by Lipopolysaccharide and Palmitic Acid Increases Cholesterol Accumulation via Enhancing Myeloid Differentiation Factor 88 Expression in HepG2 Cells. Pharmaceuticals (Basel) 2022; 15:ph15070813. [PMID: 35890112 PMCID: PMC9322353 DOI: 10.3390/ph15070813] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 06/21/2022] [Accepted: 06/27/2022] [Indexed: 02/01/2023] Open
Abstract
Recently, multiple studies have shown that chronic inflammation disturbs cholesterol homeostasis and promotes its accumulation in the liver. The underlying molecular mechanism remains to be revealed. The relationship between the toll-like receptor 4 (TLR4) inflammatory signaling pathway and cholesterol accumulation was investigated in HepG2 cells treated with lipopolysaccharide (LPS) or palmitic acid (PA) for different lengths of time. In addition, the effects of pretreatment with 20μmol/L ST2825 (MyD88 inhibitor) were also studied in LPS- or PA-treated HepG2 cells and myeloid differentiation factor 88 (MyD88)-overexpressing HEK293T cells. The intracellular total and free cholesterol levels were measured using a commercial kit and filipin staining, respectively. The expression levels of sterol regulatory element-binding protein-2 (SREBP-2) and components in the TLR4 signaling pathway were determined using Western blotting. The treatments with LPS for 12 h and with PA for 24 h significantly increased the contents of intracellular total and free cholesterol, as well as the expression levels of SREBP-2 and components in the TLR4 signaling pathway. The inhibition of MyD88 by ST2825 significantly decreased the cholesterol content and the expression levels of SREBP-2 and components of the TLR4/MyD88/NF-κB pathway in HepG2 cells, as well as MyD88-overexpressing HEK293T cells. These results indicated that LPS and PA treatments increase SREBP-2-mediated cholesterol accumulation via the activation of the TLR4/MyD88/NF-κB signaling pathway in HepG2 cells.
Collapse
|
16
|
Santos-Sánchez G, Cruz-Chamorro I, Bollati C, Bartolomei M, Pedroche J, Millán F, Millán-Linares MDC, Capriotti AL, Cerrato A, Laganà A, Arnoldi A, Carrillo-Vico A, Lammi C. A Lupinus angustifolius protein hydrolysate exerts hypocholesterolemic effects in Western diet-fed ApoE -/- mice through the modulation of LDLR and PCSK9 pathways. Food Funct 2022; 13:4158-4170. [PMID: 35316320 DOI: 10.1039/d1fo03847h] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Lupin protein hydrolysates (LPHs) are gaining attention in the food and nutraceutical industries due to their several beneficial health effects. Recently, we have shown that LPH treatment reduces liver cholesterol and triglyceride levels in hypercholesterolemic mice. The aim of this study was to elucidate the effects of LPH treatment on the molecular mechanism underlying liver cholesterol metabolism in ApoE-/- mice fed the Western diet. After identifying the composition of the peptide within the LPH mixture and determining its ability to reduce HMGCoAR activity in vitro, its effect on the LDLR and PCSK9 pathways was measured in liver tissue from the same mice. Thus, the LPH reduced the protein levels of HMGCoAR and increased the phosphorylated inactive form of HMGCoAR and the pHMGCoAR/HMGCoAR ratio, which led to the deactivation of de novo cholesterol synthesis. Furthermore, the LPH decreased the protein levels of SREBP2, a key upstream transcription factor involved in the expression of HMGCoAR and LDLR. Consequently, LDLR protein levels decreased in the liver of LPH-treated animals. Interestingly, the LPH also increased the protein levels of pAMPK responsible for HMGCoAR phosphorylation. Furthermore, the LPH controlled the PSCK9 signal pathway by decreasing its transcription factor, the HNF1-α protein. Consequently, lower PSCK9 protein levels were found in the liver of LPH-treated mice. This is the first study elucidating the molecular mechanism at the basis of the hypocholesterolemic effects exerted by the LPH in an in vivo model. All these findings point out LPHs as a future lipid-lowering ingredient to develop new functional foods.
Collapse
Affiliation(s)
- Guillermo Santos-Sánchez
- Department of Pharmaceutical Sciences, University of Milan, 20133 Milan, Italy. .,Departamento de Bioquímica Médica y Biología Molecular e Inmunología, Universidad de Sevilla, 41009 Seville, Spain. .,Instituto de Biomedicina de Sevilla, IBiS (Universidad de Sevilla, HUVR, Junta de Andalucía, CSIC), 41013 Seville, Spain
| | - Ivan Cruz-Chamorro
- Department of Pharmaceutical Sciences, University of Milan, 20133 Milan, Italy. .,Departamento de Bioquímica Médica y Biología Molecular e Inmunología, Universidad de Sevilla, 41009 Seville, Spain. .,Instituto de Biomedicina de Sevilla, IBiS (Universidad de Sevilla, HUVR, Junta de Andalucía, CSIC), 41013 Seville, Spain
| | - Carlotta Bollati
- Department of Pharmaceutical Sciences, University of Milan, 20133 Milan, Italy.
| | - Martina Bartolomei
- Department of Pharmaceutical Sciences, University of Milan, 20133 Milan, Italy.
| | - Justo Pedroche
- Department of Food & Health, Instituto de la Grasa, CSIC, Ctra, Utrera Km 1, 41013 Seville, Spain
| | - Francisco Millán
- Department of Food & Health, Instituto de la Grasa, CSIC, Ctra, Utrera Km 1, 41013 Seville, Spain
| | - María Del Carmen Millán-Linares
- Departamento de Bioquímica Médica y Biología Molecular e Inmunología, Universidad de Sevilla, 41009 Seville, Spain. .,Department of Food & Health, Instituto de la Grasa, CSIC, Ctra, Utrera Km 1, 41013 Seville, Spain
| | - Anna Laura Capriotti
- Dipartimento di Chimica, Sapienza Università di Roma, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Andrea Cerrato
- Dipartimento di Chimica, Sapienza Università di Roma, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Aldo Laganà
- Dipartimento di Chimica, Sapienza Università di Roma, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Anna Arnoldi
- Department of Pharmaceutical Sciences, University of Milan, 20133 Milan, Italy.
| | - Antonio Carrillo-Vico
- Departamento de Bioquímica Médica y Biología Molecular e Inmunología, Universidad de Sevilla, 41009 Seville, Spain. .,Instituto de Biomedicina de Sevilla, IBiS (Universidad de Sevilla, HUVR, Junta de Andalucía, CSIC), 41013 Seville, Spain
| | - Carmen Lammi
- Department of Pharmaceutical Sciences, University of Milan, 20133 Milan, Italy.
| |
Collapse
|
17
|
Charan J, Riyad P, Ram H, Purohit A, Ambwani S, Kashyap P, Singh G, Hashem A, Abd_Allah EF, Gupta VK, Kumar A, Panwar A. Ameliorations in dyslipidemia and atherosclerotic plaque by the inhibition of HMG-CoA reductase and antioxidant potential of phytoconstituents of an aqueous seed extract of Acacia senegal (L.) Willd in rabbits. PLoS One 2022; 17:e0264646. [PMID: 35239727 PMCID: PMC8893677 DOI: 10.1371/journal.pone.0264646] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 02/14/2022] [Indexed: 11/19/2022] Open
Abstract
The assigned work was aimed to examine the capability of phytoconstituents of an aqueous seed extract of Acacia senegal (L.) Willd to inhibit HMG-CoA reductase and regression of the atherosclerotic plaque. The chemical fingerprinting of the test extract was assessed by LC-MS/MS. Consequently, the analyses of in-vitro, in-vivo, and in-silico were executed by using the standard protocols. The in-vitro assessment of the test extract revealed 74.1% inhibition of HMG-CoA reductase. In-vivo assessments of the test extract indicated that treated hypercholesterolemic rabbits exhibited a significant (P≤0.001) amelioration in the biomarker indices of the dyslipidaemia i.e., atherogenic index, Castelli risk index(I&II), atherogenic coefficient along with lipid profile. Subsequently, significant reductions were observed in the atherosclerotic plaque and antioxidant levels. The in-silico study of molecular docking shown interactions capabilities of the leading phytoconstituents of the test extract i.e., eicosanoic acid, linoleic acid, and flavan-3-ol with target protein of HMG-CoA reductase. The values of RSMF and potential energy of top docked complexes were show significant interactions. Accordingly, the free energy of solvation, interaction angle, radius of gyration and SASA were shown significant stabilities of top docked complex. The cumulative data of results indicate phytoconstituents of an aqueous seed extract of Acacia senegal have capabilities to inhibit the HMG-CoA reductase and improve the levels of antioxidants.
Collapse
Affiliation(s)
- Jaykaran Charan
- Department of Pharmacology, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Priyanka Riyad
- Department of Zoology, Jai Narain Vyas University, Jodhpur, Rajasthan, India
| | - Heera Ram
- Department of Zoology, Jai Narain Vyas University, Jodhpur, Rajasthan, India
| | - Ashok Purohit
- Department of Zoology, Jai Narain Vyas University, Jodhpur, Rajasthan, India
| | - Sneha Ambwani
- Department of Pharmacology, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Priya Kashyap
- University School of Biotechnology, GGS Indraprastha University, New Delhi, India
| | - Garima Singh
- Department of Botany, Pachhunga University College, Aizawl, Mizoram, India
| | - Abeer Hashem
- Botany and Microbiology Department, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Elsayed Fathi Abd_Allah
- Plant Production Department, College of Food and Agricultural Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Vijai Kumar Gupta
- Center for Safe and Improved Food & Biorefining and Advanced Biomaterials Research Center, SRUC, Kings Buildings, Scotland, United Kingdom
| | - Ashok Kumar
- Centre for Systems biology and bioinformatics, Panjab University Chandigarh, Punjab, India
| | - Anil Panwar
- Centre for Systems biology and bioinformatics, Panjab University Chandigarh, Punjab, India
| |
Collapse
|
18
|
Abstract
Cholesterol is an essential component of eukaryotic cellular membranes. It is also an important precursor for making other molecules needed by the body. Cholesterol homeostasis plays an essential role in human health. Having high cholesterol can increase the chances of getting heart disease. As a result of the risks associated with high cholesterol, it is imperative that studies are conducted to determine the best course of action to reduce whole body cholesterol levels. Mathematical models can provide direction on this. By examining existing models, the suitable reactions or processes for drug targeting to lower whole-body cholesterol can be determined. This paper examines existing models in the literature that, in total, cover most of the processes involving cholesterol metabolism and transport, including: the absorption of cholesterol in the intestine; the cholesterol biosynthesis in the liver; the storage and transport of cholesterol between the intestine, the liver, blood vessels, and peripheral cells. The findings presented in these models will be discussed for potential combination to form a comprehensive model of cholesterol within the entire body, which is then taken as an in-silico patient for identifying drug targets, screening drugs, and designing intervention strategies to regulate cholesterol levels in the human body.
Collapse
|
19
|
Pan X. Cholesterol Metabolism in Chronic Kidney Disease: Physiology, Pathologic Mechanisms, and Treatment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1372:119-143. [PMID: 35503178 PMCID: PMC11106795 DOI: 10.1007/978-981-19-0394-6_9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
High plasma levels of lipids and/or lipoproteins are risk factors for atherosclerosis, nonalcoholic fatty liver disease (NAFLD), obesity, and diabetes. These four conditions have also been identified as risk factors leading to the development of chronic kidney disease (CKD). Although many pathways that generate high plasma levels of these factors have been identified, most clinical and physiologic dysfunction results from aberrant assembly and secretion of lipoproteins. The results of several published studies suggest that elevated levels of low-density lipoprotein (LDL)-cholesterol are a risk factor for atherosclerosis, myocardial infarction, coronary artery calcification associated with type 2 diabetes, and NAFLD. Cholesterol metabolism has also been identified as an important pathway contributing to the development of CKD; clinical treatments designed to alter various steps of the cholesterol synthesis and metabolism pathway are currently under study. Cholesterol synthesis and catabolism contribute to a multistep process with pathways that are regulated at the cellular level in renal tissue. Cholesterol metabolism may also be regulated by the balance between the influx and efflux of cholesterol molecules that are capable of crossing the membrane of renal proximal tubular epithelial cells and podocytes. Cellular accumulation of cholesterol can result in lipotoxicity and ultimately kidney dysfunction and failure. Thus, further research focused on cholesterol metabolism pathways will be necessary to improve our understanding of the impact of cholesterol restriction, which is currently a primary intervention recommended for patients with dyslipidemia.
Collapse
Affiliation(s)
- Xiaoyue Pan
- Department of Foundations of Medicine, New York University Long Island School of Medicine, Mineola, NY, USA.
| |
Collapse
|
20
|
Silva JYGD, Rodrigues PAS, Holanda MO, Silva BBD, Lima CLS, Lira SM, Moura LFWG, Paim RTT, Florean EOPT, Marques MMM, Oriá RB, Mendes FNP, Vieira IGP, Guedes MIF. Hypolipidemic and reduced nitrergic effects of p-hydroxycinnamic diesters extracted from Copernicia prunifera in mice challenged by a high-fat diet. Biomed Pharmacother 2021; 142:111930. [PMID: 34416631 DOI: 10.1016/j.biopha.2021.111930] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 07/05/2021] [Accepted: 07/12/2021] [Indexed: 11/27/2022] Open
Abstract
Dyslipidemia is a chronic non-transmissible condition that has increased due to an unhealthy lifestyle. Statins have been used as the standard treatment to control hyperlipidemia. However, side effects and high costs may be associated with its prolonged treatment, so plants derivatives have been an attractive therapy to overcome these problems. Among the compounds extracted from plants, the p-hydroxycinnamic diesters (HCE), present in carnauba wax (CW), have been found with good pharmacological properties. Therefore, this study aimed to evaluate the potential anti-hypercholesterolemic and possible toxicological effects of HCE in C57BL/6J mice under a high-fat (HF) diet. Male C57BL/6J mice were fed during 60 days under the HF diet and therefore were either treated with HCE (200 and 400 mg/kg) or simvastatin (20 mg/kg) or received saline (controls) by gavage for 30 days under the same diet. HCE treatment was able to reduce serum total cholesterol and LDL levels. Besides, this compound increased liver X receptor (LXR) and but not significantly affected IL-1β and TNF-α liver mRNA transcription activity. In conclusion, HCE treatment was found safe and may attenuate the deleterious effects of dyslipidemia due to chronic feeding with western diets.
Collapse
Affiliation(s)
- José Ytalo Gomes da Silva
- Laboratory of Biotechnology & Molecular Biology, State University of Ceara, Fortaleza 60.714.903, Brazil.
| | | | - Marcelo Oliveira Holanda
- Laboratory of Biotechnology & Molecular Biology, State University of Ceara, Fortaleza 60.714.903, Brazil
| | - Bruno Bezerra da Silva
- Laboratory of Biotechnology & Molecular Biology, State University of Ceara, Fortaleza 60.714.903, Brazil
| | - Carla Laine Silva Lima
- Laboratory of Biotechnology & Molecular Biology, State University of Ceara, Fortaleza 60.714.903, Brazil
| | - Sandra Machado Lira
- Laboratory of Biotechnology & Molecular Biology, State University of Ceara, Fortaleza 60.714.903, Brazil
| | | | | | | | | | - Reinaldo Barreto Oriá
- Laboratory of Tissue healing, Ontogeny and Nutrition, Department of Morphology and Institute of Biomedicine, Federal University of Ceara, Fortaleza 60.430-270, Brazil
| | | | - Icaro Gusmão Pinto Vieira
- Laboratory of Biotechnology & Molecular Biology, State University of Ceara, Fortaleza 60.714.903, Brazil
| | | |
Collapse
|
21
|
Wu J, Zhang F, Ruan H, Chang X, Wang J, Li Z, Jin W, Shi Y. Integrating Network Pharmacology and RT-qPCR Analysis to Investigate the Mechanisms Underlying ZeXie Decoction-Mediated Treatment of Non-alcoholic Fatty Liver Disease. Front Pharmacol 2021; 12:722016. [PMID: 34566646 PMCID: PMC8458890 DOI: 10.3389/fphar.2021.722016] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 08/26/2021] [Indexed: 01/14/2023] Open
Abstract
ZeXie Decoction (ZXD) is a traditional Chinese medicine composed of Alisma orientalis (Sam.) Juzep. and Atractylodes macrocephala Koidz. ZXD has been widely used to treat non-alcoholic fatty liver disease (NAFLD). The mechanistic basis for the pharmacological activity of ZXD, however, remains poorly understood. In this study, we used a network pharmacology approach and investigated the association between ZXD and NAFLD. We identified the active ingredients of ZXD and screened the potential targets of these ingredients, after which a database of relevant NAFLD-related targets were constructed and several enrichment analyses were performed. Furthermore, the ethanol and aqueous extracts of ZXD were prepared and experimental pharmacology validation was conducted using RT-qPCR of the non-alcoholic fatty liver disease (NAFLD) model in Sprague-Dawley (SD) rats. As a result, a herb-compound-target-pathway network model was developed, and HMGCR, SREBP-2, MAPK1, and NF-κBp65 targets were validated. The gene expression results of these four targets were consistent with those of the network pharmacology prediction. Using an integration strategy, we revealed that ZXD could treat NAFLD by targeting HMGCR, SREBP-2, MAPK1, and NF-κBp65.
Collapse
Affiliation(s)
- Jiashuo Wu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fangqing Zhang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Haonan Ruan
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaoyan Chang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jingxun Wang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhuangzhuang Li
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Weiyi Jin
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,College of Public Health, Hebei Medical University, Shijiazhuang, China
| | - Yue Shi
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
22
|
Santos-Sánchez G, Cruz-Chamorro I, Álvarez-Ríos AI, Fernández-Santos JM, Vázquez-Román MV, Rodríguez-Ortiz B, Álvarez-Sánchez N, Álvarez-López AI, Millán-Linares MDC, Millán F, Pedroche J, Fernández-Pachón MS, Lardone PJ, Guerrero JM, Bejarano I, Carrillo-Vico A. Lupinus angustifolius Protein Hydrolysates Reduce Abdominal Adiposity and Ameliorate Metabolic Associated Fatty Liver Disease (MAFLD) in Western Diet Fed-ApoE -/- Mice. Antioxidants (Basel) 2021; 10:antiox10081222. [PMID: 34439470 PMCID: PMC8388992 DOI: 10.3390/antiox10081222] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 07/21/2021] [Accepted: 07/26/2021] [Indexed: 12/11/2022] Open
Abstract
Metabolic-associated fatty liver disease (MAFLD) is the most important cause of liver disease worldwide. It is characterized by the accumulation of fat in the liver and is closely associated with abdominal obesity. In addition, oxidative stress and inflammation are significant features involved in MAFLD. Recently, our group demonstrated that lupin protein hydrolysates (LPHs) had lipid lowering, antioxidant, and anti-inflammatory effects. Sixty male mice fed with a Western diet were intragastrically treated with LPHs (or vehicle) for 12 weeks. Liver and adipose tissue lipid accumulation and hepatic inflammatory and oxidant status were evaluated. A significant decrease in steatosis was observed in LPHs-treated mice, which presented a decreased gene expression of CD36 and LDL-R, crucial markers in MAFLD. In addition, LPHs increased the hepatic total antioxidant capacity and reduced the hepatic inflammatory status. Moreover, LPHs-treated mice showed a significant reduction in abdominal adiposity. This is the first study to show that the supplementation with LPHs markedly ameliorates the generation of the steatotic liver caused by the intake of a Western diet and reduces abdominal obesity in ApoE−/− mice. Future clinical trials should shed light on the effects of LPHs on MAFLD.
Collapse
Affiliation(s)
- Guillermo Santos-Sánchez
- Instituto de Biomedicina de Sevilla, IBiS (Universidad de Sevilla, HUVR, Junta de Andalucía, CSIC), 41013 Seville, Spain; (G.S.-S.); (B.R.-O.); (N.Á.-S.); (A.I.Á.-L.); (P.J.L.); (J.M.G.); (I.B.)
- Departamento de Bioquímica Médica y Biología Molecular e Inmunología, Universidad de Sevilla, 41009 Seville, Spain
| | - Ivan Cruz-Chamorro
- Instituto de Biomedicina de Sevilla, IBiS (Universidad de Sevilla, HUVR, Junta de Andalucía, CSIC), 41013 Seville, Spain; (G.S.-S.); (B.R.-O.); (N.Á.-S.); (A.I.Á.-L.); (P.J.L.); (J.M.G.); (I.B.)
- Departamento de Bioquímica Médica y Biología Molecular e Inmunología, Universidad de Sevilla, 41009 Seville, Spain
- Correspondence: (I.C.-C.); (A.C.-V.); Tel.: +34-955-923-106 (I.C.-C. & A.C.-V.)
| | - Ana Isabel Álvarez-Ríos
- Departamento de Bioquímica Clínica, Unidad de Gestión de Laboratorios, Hospital Universitario Virgen del Rocío, 41013 Seville, Spain;
| | - José María Fernández-Santos
- Departamento Citología e Histología Normal y Patológica, Universidad de Sevilla, 41009 Seville, Spain; (J.M.F.-S.); (M.V.V.-R.)
| | - María Victoria Vázquez-Román
- Departamento Citología e Histología Normal y Patológica, Universidad de Sevilla, 41009 Seville, Spain; (J.M.F.-S.); (M.V.V.-R.)
| | - Beatriz Rodríguez-Ortiz
- Instituto de Biomedicina de Sevilla, IBiS (Universidad de Sevilla, HUVR, Junta de Andalucía, CSIC), 41013 Seville, Spain; (G.S.-S.); (B.R.-O.); (N.Á.-S.); (A.I.Á.-L.); (P.J.L.); (J.M.G.); (I.B.)
| | - Nuria Álvarez-Sánchez
- Instituto de Biomedicina de Sevilla, IBiS (Universidad de Sevilla, HUVR, Junta de Andalucía, CSIC), 41013 Seville, Spain; (G.S.-S.); (B.R.-O.); (N.Á.-S.); (A.I.Á.-L.); (P.J.L.); (J.M.G.); (I.B.)
| | - Ana Isabel Álvarez-López
- Instituto de Biomedicina de Sevilla, IBiS (Universidad de Sevilla, HUVR, Junta de Andalucía, CSIC), 41013 Seville, Spain; (G.S.-S.); (B.R.-O.); (N.Á.-S.); (A.I.Á.-L.); (P.J.L.); (J.M.G.); (I.B.)
- Departamento de Bioquímica Médica y Biología Molecular e Inmunología, Universidad de Sevilla, 41009 Seville, Spain
| | | | - Francisco Millán
- Department of Food & Health, Instituto de la Grasa, CSIC, Ctra, Utrera Km 1, 41013 Seville, Spain; (F.M.); (J.P.)
| | - Justo Pedroche
- Department of Food & Health, Instituto de la Grasa, CSIC, Ctra, Utrera Km 1, 41013 Seville, Spain; (F.M.); (J.P.)
| | - María Soledad Fernández-Pachón
- Área de Nutrición y Bromatología, Departamento de Biología Molecular e Ingeniería Bioquímica, Universidad Pablo de Olavide, Ctra Utrera Km 1, 41013 Seville, Spain;
| | - Patricia Judith Lardone
- Instituto de Biomedicina de Sevilla, IBiS (Universidad de Sevilla, HUVR, Junta de Andalucía, CSIC), 41013 Seville, Spain; (G.S.-S.); (B.R.-O.); (N.Á.-S.); (A.I.Á.-L.); (P.J.L.); (J.M.G.); (I.B.)
- Departamento de Bioquímica Médica y Biología Molecular e Inmunología, Universidad de Sevilla, 41009 Seville, Spain
| | - Juan Miguel Guerrero
- Instituto de Biomedicina de Sevilla, IBiS (Universidad de Sevilla, HUVR, Junta de Andalucía, CSIC), 41013 Seville, Spain; (G.S.-S.); (B.R.-O.); (N.Á.-S.); (A.I.Á.-L.); (P.J.L.); (J.M.G.); (I.B.)
- Departamento de Bioquímica Médica y Biología Molecular e Inmunología, Universidad de Sevilla, 41009 Seville, Spain
- Departamento de Bioquímica Clínica, Unidad de Gestión de Laboratorios, Hospital Universitario Virgen del Rocío, 41013 Seville, Spain;
| | - Ignacio Bejarano
- Instituto de Biomedicina de Sevilla, IBiS (Universidad de Sevilla, HUVR, Junta de Andalucía, CSIC), 41013 Seville, Spain; (G.S.-S.); (B.R.-O.); (N.Á.-S.); (A.I.Á.-L.); (P.J.L.); (J.M.G.); (I.B.)
- Departamento de Bioquímica Médica y Biología Molecular e Inmunología, Universidad de Sevilla, 41009 Seville, Spain
| | - Antonio Carrillo-Vico
- Instituto de Biomedicina de Sevilla, IBiS (Universidad de Sevilla, HUVR, Junta de Andalucía, CSIC), 41013 Seville, Spain; (G.S.-S.); (B.R.-O.); (N.Á.-S.); (A.I.Á.-L.); (P.J.L.); (J.M.G.); (I.B.)
- Departamento de Bioquímica Médica y Biología Molecular e Inmunología, Universidad de Sevilla, 41009 Seville, Spain
- Correspondence: (I.C.-C.); (A.C.-V.); Tel.: +34-955-923-106 (I.C.-C. & A.C.-V.)
| |
Collapse
|
23
|
Du H, Li C, Wang Z, He Y, Wang Y, Zhou H, Wan H, Yang J. Effects of Danhong injection on dyslipidemia and cholesterol metabolism in high-fat diets fed rats. JOURNAL OF ETHNOPHARMACOLOGY 2021; 274:114058. [PMID: 33766756 DOI: 10.1016/j.jep.2021.114058] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 03/14/2021] [Accepted: 03/19/2021] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Danhong injection (DHI) is a Chinese medical injection applied to the clinical treatment of cardiovascular diseases that has anti-inflammatory, antiplatelet aggregation and antithrombotic effects. This study aimed to explore the effects of DHI on dyslipidemia and cholesterol metabolism in high-fat diet-fed rats. METHODS Sprague Dawley (SD) rats were randomly divided into six groups: normal group (Normal); hyperlipidemia model group (Model); DHI-treated groups at doses of 1.0 mL/kg, 2.0 mL/kg, 4.0 mL/kg; and simvastatin positive control group (2.0 mg/kg). The hypolipidemic effects of DHI were evaluated by measuring serum lipid levels, hepatic function and oxidative stress, respectively. And pathological changes in liver tissues were determined using hematoxylin-eosin (H&E) and oil red O staining. Moreover, the mRNA and protein expression levels of cholesterol metabolism related genes were detected by real-time PCR (RT-PCR) and Western blot. RESULTS Compared with the Model group, DHI treatment markedly decreased the liver index and improved the pathological morphology of liver tissues. DHI treatment dose-dependently decreased the levels of total cholesterol (TC), triglycerides (TG), low-density lipoprotein cholesterol (LDL-C), malondialdehyde (MDA), and free fatty acids (FFA) in serum or liver tissues (P < 0.01 or P < 0.05), and increased the high-density lipoprotein cholesterol (HDL-C) and tripeptide glutathione (GSH) (P < 0.01 or P < 0.05). The activities of superoxide dismutase (SOD) and glutathione peroxidase (GSH-PX) were increased in the DHI-treated groups (P < 0.01 or P < 0.05), while the alanine transaminase (ALT) and aspartate transaminase (AST) were decreased (P < 0.01 or P < 0.05). Furthermore, the expression levels of LDL receptor (LDLR), cholesterol 7-α-hydroxylase (CYP7A1), liver X receptor α (LXRα), and peroxisome proliferator-activated receptor α (PPARα) were dose-dependently upregulated in the DHI-treated groups, whereas the expression of sterol regulatory element-binding protein-2 (SREBP-2) was downregulated. CONCLUSIONS Our study demonstrated that DHI markedly ameliorated hyperlipidemia rats by regulating serum lipid levels, inhibiting hepatic lipid accumulation and steatosis, improving hepatic dysfunction, and reducing oxidative stress. The potential mechanism was also tentatively investigated and may be related to the promotion of bile acid synthesis via activation of the PPARα-LXRα-CYP7A1 pathway. Therefore, DHI could be regarded as a potential hypolipidemic drug for the treatment of hyperlipidemia.
Collapse
Affiliation(s)
- Haixia Du
- Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Chang Li
- Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Zhixiong Wang
- Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Yu He
- Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Yu Wang
- Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Huifen Zhou
- Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Haitong Wan
- Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Jiehong Yang
- Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| |
Collapse
|
24
|
Jin BR, Lee M, An HJ. Nodakenin represses obesity and its complications via the inhibition of the VLDLR signalling pathway in vivo and in vitro. Cell Prolif 2021; 54:e13083. [PMID: 34165214 PMCID: PMC8349651 DOI: 10.1111/cpr.13083] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/22/2021] [Accepted: 05/29/2021] [Indexed: 12/11/2022] Open
Abstract
Objectives Nodakenin (NK) is a coumarin glucoside that is found in the roots of Angelicae gigas. A limited number of studies have been conducted on the pharmacological activities of NK. Although NK is an important natural resource having anti‐inflammatory and antioxidant effects, no investigation has been conducted to examine the effects of NK on obesity and obesity‐induced inflammation. Materials and Methods The present study investigated the therapeutic effects of NK treatment on obesity and its complications, and its mechanism of action using differentiated 3T3‐L1 adipocytes and high‐fat diet (HFD)‐induced obese mice. Oil red O staining, western blot assay, qRT‐PCR assay, siRNA transfection, enzyme‐linked immunosorbent assay, H&E staining, immunohistochemistry, molecular docking and immunofluorescence staining were utilized. Results Treatment with NK demonstrated anti‐adipogenesis effects via the regulation of adipogenic transcription factors and genes associated with triglyceride synthesis in differentiated 3T3‐L1 adipocytes. Compared with the control group, the group administered NK showed a suppression in weight gain, dyslipidaemia and the development of fatty liver in HFD‐induced obese mice. In addition, NK administration inhibited adipogenic differentiation and obesity‐induced inflammation and oxidative stress via the suppression of the VLDLR and MEK/ERK1/2 pathways. This is the first study that has documented the interaction between NK and VLDLR structure. Conclusion These results demonstrate the potential of NK as a natural product‐based therapeutic candidate for the treatment of obesity and its complications by targeting adipogenesis and adipose tissue inflammation‐associated markers.
Collapse
Affiliation(s)
- Bo-Ram Jin
- Department of Pharmacology, College of Korean Medicine, Sangji University, Wonju, Korea
| | - Minho Lee
- Department of Life Science, Dongguk University-Seoul, Goyang-si, Korea
| | - Hyo-Jin An
- Department of Pharmacology, College of Korean Medicine, Sangji University, Wonju, Korea
| |
Collapse
|
25
|
High-Efficacy α,β-Dehydromonacolin S Improves Hepatic Steatosis and Suppresses Gluconeogenesis Pathway in High-Fat Diet-Induced Obese Rats. Pharmaceuticals (Basel) 2021; 14:ph14040375. [PMID: 33920678 PMCID: PMC8073358 DOI: 10.3390/ph14040375] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 04/14/2021] [Accepted: 04/15/2021] [Indexed: 12/24/2022] Open
Abstract
Isolated α,β-dehydromonacolin S (C5) from soil-derived fungus Aspergillus sclerotiorum PSU-RSPG178 was recently shown to exhibit an inhibitory effect against 3-hydroxy-3-methylglutaryl coenzyme A reductase (HMGR) activity in vitro. In this study, we investigated the effects of C5 on lipid-lowering, hepatic steatosis, and hepatic gluconeogenesis in vivo. The control rats received a daily dose of either vehicle or C5 at 10 mg/kg, while the high-fat diet-induced obese (HFD) rats were administered vehicle; 1, 3, or 10 mg/kg C5; or 10 mg/kg lovastatin (LO) for 6 weeks. C5 significantly improved dyslipidemia and diminished liver enzymes, HMGR activity, insulin resistance, and hepatic steatosis, comparable to LO without any hepatotoxicity and nephrotoxicity in HFD rats. A higher efficacy of C5 in lipid-lowering activity and anti-hepatic steatosis was associated with a significant decrease in genes involved in lipid metabolism including sterol regulatory element binding protein (SREBP) 1c, SREBP2, liver X receptor alpha (LXRα), and peroxisome proliferator-activated receptor (PPAR) gamma (PPARγ) together with an increase in the PPAR alpha (PPARα). Correspondingly, C5 was able to down-regulate the lipid transporters cluster of differentiation 36 (CD36) and Niemann-Pick C1 Like 1 (NPC1L1), increase the antioxidant superoxide dismutase gene expression, and decrease the proinflammatory cytokines, tumor necrosis factor alpha (TNFα) and interleukin 1 beta (IL-1β). Impairment of hepatic gluconeogenesis and insulin resistance in HFD rats was restored by C5 through down-regulation of the gluconeogenic genes phosphoenolpyruvate carboxykinase (PEPCK) and glucose-6-phosphatase (G6Pase), and the activation of AMP-dependent kinase serine (AMPK) and serine/threonine protein kinase B (Akt). Collectively, this novel C5 may be a therapeutic option for treating dyslipidemia, hepatic steatosis, and reducing potential risk for diabetes mellitus.
Collapse
|
26
|
Tsai WT, Nakamura Y, Akasaka T, Katakura Y, Tanaka Y, Shirouchi B, Jiang Z, Yuan X, Sato M. Soyasaponin ameliorates obesity and reduces hepatic triacylglycerol accumulation by suppressing lipogenesis in high-fat diet-fed mice. J Food Sci 2021; 86:2103-2117. [PMID: 33864648 DOI: 10.1111/1750-3841.15696] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 02/18/2021] [Accepted: 02/28/2021] [Indexed: 01/26/2023]
Abstract
Soyasaponins are triterpenoid glycosides found in soybean. We investigated whether soyasaponin ameliorates lipid metabolism and its possible mechanisms. In C57BL/6J mice fed a high-fat diet (HFD), soyasaponin (SAP) was orally administered for 9 weeks. Additionally, we evaluated the effect of soyasapogenols on 3T3-L1 adipocytes. In HFD-fed mice, the SAP significantly reduced body weight by 7% and relative adipose tissue weight by 35%. X-ray computed tomography demonstrated that the SAP reduced visceral and subcutaneous adipose tissue weights during week 3 of feeding. The SAP reduced sterol regulatory element-binding protein-1c (SREBP-1c) mRNA levels by 32% in the epididymal adipose tissue, significantly decreasing the triacylglycerol (TAG) content by 37% and SREBP-1c and fatty acid synthase mRNA levels by 52% and 61%, respectively, in the liver. In 3T3-L1 adipocytes, soyasapogenol B significantly decreased lipid droplets. The SAP containing soyasaponin A and B as conjugates demonstrate anti-obesity effects by suppressing adipocyte differentiation and lipogenesis, with a preventive effect on hepatic TAG accumulation by suppressing lipogenesis. PRACTICAL APPLICATION: Soyasaponin is one of the oleanane triterpenoids in soybeans. We have demonstrated that soyasaponin potently reduces body weight and white adipose tissue weight, and hepatic triacylglycerol accumulation in high-fat diet-fed mice. Thus, soyasaponin is a beneficial compound to prevent obesity and fatty liver.
Collapse
Affiliation(s)
- Wei-Ting Tsai
- Laboratory of Nutrition Chemistry, Department of Bioscience and Biotechnology, Faculty of Agriculture, Graduate School, Kyushu University, Fukuoka, Japan
| | - Yuki Nakamura
- Laboratory of Nutrition Chemistry, Department of Bioscience and Biotechnology, Faculty of Agriculture, Graduate School, Kyushu University, Fukuoka, Japan
| | - Taiki Akasaka
- Center for Advanced Instrumental and Educational Supports, Faculty of Agriculture, Kyushu University, Fukuoka, Japan
| | - Yoshinori Katakura
- Laboratory of Cellular Regulation Technology, Department of Bioscience and Biotechnology, Faculty of Agriculture, Graduate School, Kyushu University, Fukuoka, Japan
| | - Yasutake Tanaka
- Laboratory of Nutrition Chemistry, Department of Bioscience and Biotechnology, Faculty of Agriculture, Graduate School, Kyushu University, Fukuoka, Japan
| | - Bungo Shirouchi
- Laboratory of Nutrition Chemistry, Department of Bioscience and Biotechnology, Faculty of Agriculture, Graduate School, Kyushu University, Fukuoka, Japan
| | - Zhe Jiang
- Laboratory of Nutrition Chemistry, Department of Bioscience and Biotechnology, Faculty of Agriculture, Graduate School, Kyushu University, Fukuoka, Japan
| | - Xingyu Yuan
- Laboratory of Nutrition Chemistry, Department of Bioscience and Biotechnology, Faculty of Agriculture, Graduate School, Kyushu University, Fukuoka, Japan
| | - Masao Sato
- Laboratory of Nutrition Chemistry, Department of Bioscience and Biotechnology, Faculty of Agriculture, Graduate School, Kyushu University, Fukuoka, Japan
| |
Collapse
|
27
|
Funes AK, Simón L, Colombo R, Avena MV, Monclús M, Crescitelli J, Cabrillana ME, Conte MI, Cayado N, Boarelli P, Fornés MW, Saez Lancellotti TE. Impact of high fat diet on the sterol regulatory element-binding protein 2 cholesterol pathway in the testicle. Mol Hum Reprod 2021; 27:6206393. [PMID: 33787903 DOI: 10.1093/molehr/gaab023] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 03/18/2021] [Indexed: 12/30/2022] Open
Abstract
Male fertility has been shown to be dependent on cholesterol homeostasis. This lipid is essential for testosterone synthesis and spermatogenesis, but its levels must be maintained in an optimal range for proper testicular function. In particular, sperm cells' development is very sensitive to high cholesterol levels, noticeably during acrosomal formation. The aim of this work was to study whether the molecular pathway that regulates intracellular cholesterol, the sterol regulatory element-binding protein (SREBP) pathway, is affected in the testicles of animals under a fat diet. To investigate this, we took advantage of the non-obese hypercholesterolemia (HC) model in New Zealand rabbits that displays poor sperm and seminal quality. The testicular expression of SREBP isoform 2 (SREBP2) and its target molecules 3-hydroxy-3-methyl-glutaryl-coenzyme A reductase (HMGCR) and low-density lipoprotein receptor (LDLR) were studied under acute (6 months) and chronic (more than 12 months) fat intake by RT-PCR, western blot and immunofluorescence. Our findings showed that fat consumption promoted down-regulation of the SREBP2 pathway in the testicle at 6 months, but upregulation after a chronic period. This was consistent with load of testicular cholesterol, assessed by filipin staining. In conclusion, the intracellular pathway that regulates cholesterol levels in the testicle is sensitive to dietary fats, and behaves differently depending on the duration of consumption: it has a short-term protective effect, but became deregulated in the long term, ultimately leading to a detrimental situation. These results will contribute to the understanding of the basic mechanisms of the effect of fat consumption in humans with idiopathic infertility.
Collapse
Affiliation(s)
- Abi K Funes
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM) †. Instituto de Histología y Embriología (IHEM), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Centro Científico y Tecnológico (CCT), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza, Argentina.,Laboratorio de Biología Molecular del Metabolismo & Nutrición (bMeNu)†, Instituto de Histología y Embriología (IHEM), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Centro Científico y Tecnológico (CCT), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza, Argentina
| | - Layla Simón
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM) †. Instituto de Histología y Embriología (IHEM), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Centro Científico y Tecnológico (CCT), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza, Argentina
| | - Regina Colombo
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM) †. Instituto de Histología y Embriología (IHEM), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Centro Científico y Tecnológico (CCT), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza, Argentina.,Laboratorio de Biología Molecular del Metabolismo & Nutrición (bMeNu)†, Instituto de Histología y Embriología (IHEM), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Centro Científico y Tecnológico (CCT), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza, Argentina
| | - María Virginia Avena
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM) †. Instituto de Histología y Embriología (IHEM), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Centro Científico y Tecnológico (CCT), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza, Argentina.,Laboratorio de Biología Molecular del Metabolismo & Nutrición (bMeNu)†, Instituto de Histología y Embriología (IHEM), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Centro Científico y Tecnológico (CCT), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza, Argentina
| | - María Monclús
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM) †. Instituto de Histología y Embriología (IHEM), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Centro Científico y Tecnológico (CCT), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza, Argentina.,Instituto de Investigaciones. Facultad de Ciencias Médicas. Universidad del Aconcagua. Mendoza, Argentina
| | - Julieta Crescitelli
- Laboratorio de Biología Molecular del Metabolismo & Nutrición (bMeNu)†, Instituto de Histología y Embriología (IHEM), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Centro Científico y Tecnológico (CCT), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza, Argentina.,Instituto de Investigaciones. Facultad de Ciencias Médicas. Universidad del Aconcagua. Mendoza, Argentina
| | - María E Cabrillana
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM) †. Instituto de Histología y Embriología (IHEM), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Centro Científico y Tecnológico (CCT), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza, Argentina
| | - María Inés Conte
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM) †. Instituto de Histología y Embriología (IHEM), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Centro Científico y Tecnológico (CCT), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza, Argentina
| | - Niubys Cayado
- Instituto de Investigaciones. Facultad de Ciencias Médicas. Universidad del Aconcagua. Mendoza, Argentina.,Laboratorio de Oncología, Instituto de Medicina y Biología Experimental de Cuyo (IMBECU), Centro Científico y Tecnológico (CCT), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza, Argentina
| | - Paola Boarelli
- Laboratorio de Enfermedades Metabólicas (LEM), Universidad Juan Agustín Maza, Mendoza, Argentina
| | - Miguel W Fornés
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM) †. Instituto de Histología y Embriología (IHEM), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Centro Científico y Tecnológico (CCT), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza, Argentina
| | - Tania E Saez Lancellotti
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM) †. Instituto de Histología y Embriología (IHEM), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Centro Científico y Tecnológico (CCT), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza, Argentina.,Laboratorio de Biología Molecular del Metabolismo & Nutrición (bMeNu)†, Instituto de Histología y Embriología (IHEM), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Centro Científico y Tecnológico (CCT), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza, Argentina.,Instituto de Investigaciones. Facultad de Ciencias Médicas. Universidad del Aconcagua. Mendoza, Argentina
| |
Collapse
|
28
|
Bagherieh M, Kheirollahi A, Zamani-Garmsiri F, Emamgholipour S, Meshkani R. Folic acid ameliorates palmitate-induced inflammation through decreasing homocysteine and inhibiting NF-κB pathway in HepG2 cells. Arch Physiol Biochem 2021:1-8. [PMID: 33596128 DOI: 10.1080/13813455.2021.1878539] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Prevention of inflammation is one of the possible remedy procedure for steatohepatitis during NAFLD. In this study, we researched the folic acid (FA) potency to attenuate the inflammation of palmitate-treated HepG2 cells and the related signalling pathways. METHODS The molecular mechanisms related to FA anti-inflammatory effect in palmitate and Hcy-treated HepG2 cell line were assessed. RESULTS The results indicated that while palmitate enhances the expression and secretion of TNF-α, IL-6, and IL-1β, and also intracellular ROS level, FA at concentrations of 25, 50, and 75 µg/mL significantly reversed these effects in HepG2 cells. In addition, FA could ameliorate inflammation and decrease ROS production induced by Hcy. Furthermore, FA pre-treatment suppress palmitate -induced (NF-κB) p65 level in palmitate or Hcy stimulated cells. CONCLUSIONS Overall, these results suggest that FA reduces inflammation in HepG2 cells through decreasing ROS and Hcy concentration level resulting in inhibiting the NF-κB pathway.
Collapse
Affiliation(s)
- Molood Bagherieh
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Asma Kheirollahi
- Department of Comparative Biosciences, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Fahimeh Zamani-Garmsiri
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Solaleh Emamgholipour
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Meshkani
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
29
|
Liu YS, Yuan MH, Zhang CY, Liu HM, Liu JR, Wei AL, Ye Q, Zeng B, Li MF, Guo YP, Guo L. Puerariae Lobatae radix flavonoids and puerarin alleviate alcoholic liver injury in zebrafish by regulating alcohol and lipid metabolism. Biomed Pharmacother 2020; 134:111121. [PMID: 33341668 DOI: 10.1016/j.biopha.2020.111121] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 11/27/2020] [Accepted: 12/07/2020] [Indexed: 01/01/2023] Open
Abstract
Exessive drinking is commonly associated with a wide spectrum of liver injuries. The term alcoholic liver disease (ALD) is generally used to refer to this spectrum of hepatic abnormalities, and the term hepatic steatosis denotes early lesions. Puerariae Lobatae Radix (PLR) is a common traditional Chinese medicine and has been widely used as an efficient treatment for alcohol-induced damage. Flavonoids are the principal components of PLR that could potentially be responsible for the activation of alcohol metabolism and lipid-lowering effects. However, little is known about the mechanisms underlying their activity against alcoholic injury. In this study, PLR flavonoids (PLF) were obtained by microwave extraction. A 2% ethanol solution was used to establish a model of alcoholic fatty liver disease by exposure of zebrafish larvae for 32 h, and then the zebrafish were administered PLF and puerarin. The results showed that PLF and puerarin significantly decreased lipid accumulation and the levels of total cholesterol and triglycerides in zebrafish larvae. Moreover, PLF and puerarin downregulated the expression of genes related to alcohol and lipid metabolism (CYP2y3, CYP3a65, ADH8a, ADH8b, HMGCRB, and FASN), endoplasmic reticulum stress, and DNA damage (CHOP, EDEM1, GADD45αa, and ATF6) and reduced levels of inflammatory factors (IL-1β, TNF-α) in zebrafish larvae. PLF and puerarin increased the phosphorylation of AMP-activated protein kinase-α (AMPKα) and decreased the total protein level of ACC1. The findings suggested that PLF and puerarin alleviated alcohol-induced hepatic steatosis in zebrafish larvae by regulating alcohol and lipid metabolism, which was closely related to the regulation of the AMPKα-ACC signaling pathway. In conclusion, the study provided a possible therapeutic drug for ALD treatment.
Collapse
Affiliation(s)
- Yu-Shi Liu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Ming-Hao Yuan
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Cun-Yan Zhang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Hong-Mei Liu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Juan-Ru Liu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Ai-Ling Wei
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Qiang Ye
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Bin Zeng
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Mei-Feng Li
- School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yi-Ping Guo
- Quantitative and Systems Biology Graduate Program, School of Natural Sciences, University of California, Merced, CA, 95343, USA.
| | - Li Guo
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
30
|
Ramchoun M, Khouya T, Alibrahim EA, Hmidani A, Sellam K, Amrani S, Harnafi H, Benlyas M, Kasbi Chadli F, Ouguerram K, Alem C. Thymus atlanticus polyphenol-rich extract regulates cholesterol metabolism by inhibiting its biosynthesis without affecting its excretion in hamsters fed a high-fat diet. Arch Physiol Biochem 2020; 129:618-625. [PMID: 33320714 DOI: 10.1080/13813455.2020.1854308] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
CONTEXT Thymus atlanticus has been reported to have significant hypolipidaemic effect in animal models. However, the mechanism of this hypolipidaemic action still unknown. OBJECTIVE To determinate the possible mechanism(s) of hypolipidaemic action of a Thymus atlanticus polyphenol-rich extract (PRE). MATERIALS AND METHODS Plasma, faecal, and liver cholesterol, bile acid content in the faeces, and gene expression level of HMG-CoA reductase, CYP7A1, ABCG5 and ABCG8 were analysed after 9 weeks in hamsters feeding normal diet, high-fat diet (HFD) or HFD supplemented with 400 mg/kg body weight/day of PRE. RESULTS PRE significantly decreased total cholesterol content (p < .05) and HMG-CoA reductase expression (p < .05), but did not affect the faecal cholesterol, bile acid contents and CYP7A1 and ABCG5/G8 expression (p > .05). CONCLUSION We can conclude that the T. atlanticus extract is efficient in the alleviation of chronic hyperlipidaemia by acting as cholesterol biosynthesis inhibitor.
Collapse
Affiliation(s)
- Mhamed Ramchoun
- Laboratory of Biotechnology and Sustainable Development of Natural Resources, Polydisciplinary Faculty, University Sultan Moulay Slimane, Beni Mellal, Morocco
- Biochemistry and Natural Substances Team, Department of Biology, Faculty of Sciences & Techniques, University Moulay Ismail, Errachidia, Morocco
- Laboratory of Biochemistry and Biotechnologies, Department of Biology, Faculty of Sciences, University Mohamed I, Oujda, Morocco
| | - Tarik Khouya
- Biochemistry and Natural Substances Team, Department of Biology, Faculty of Sciences & Techniques, University Moulay Ismail, Errachidia, Morocco
| | - Eid Alabed Alibrahim
- INSERM U1063, Stress Oxydant et Pathologies Métaboliques, Université d'Angers, Université Bretagne-Loire, Angers, France
| | - Abdelbassat Hmidani
- Biochemistry and Natural Substances Team, Department of Biology, Faculty of Sciences & Techniques, University Moulay Ismail, Errachidia, Morocco
| | - Khalid Sellam
- Biochemistry and Natural Substances Team, Department of Biology, Faculty of Sciences & Techniques, University Moulay Ismail, Errachidia, Morocco
| | - Souliman Amrani
- Laboratory of Biochemistry and Biotechnologies, Department of Biology, Faculty of Sciences, University Mohamed I, Oujda, Morocco
| | - Hicham Harnafi
- Laboratory of Biochemistry and Biotechnologies, Department of Biology, Faculty of Sciences, University Mohamed I, Oujda, Morocco
| | - Mohamed Benlyas
- Biochemistry and Natural Substances Team, Department of Biology, Faculty of Sciences & Techniques, University Moulay Ismail, Errachidia, Morocco
| | - Fatima Kasbi Chadli
- INRA, UMR 1280, Physiologie des Adaptations Nutritionnelles, Nantes, France
- CRNH, Western Human Nutrition Research Centre, CHU Hôtel-Dieu, Nantes, France
| | - Khadija Ouguerram
- INRA, UMR 1280, Physiologie des Adaptations Nutritionnelles, Nantes, France
- CRNH, Western Human Nutrition Research Centre, CHU Hôtel-Dieu, Nantes, France
| | - Chakib Alem
- Biochemistry and Natural Substances Team, Department of Biology, Faculty of Sciences & Techniques, University Moulay Ismail, Errachidia, Morocco
| |
Collapse
|
31
|
Protective Role of Picralima nitida Seed Extract in High-Fat High-Fructose-Fed Rats. Adv Pharmacol Pharm Sci 2020; 2020:5206204. [PMID: 33163962 PMCID: PMC7604582 DOI: 10.1155/2020/5206204] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 09/22/2020] [Accepted: 10/03/2020] [Indexed: 12/23/2022] Open
Abstract
Picralima nitida is a therapeutic herb used in ethnomedicine for the management of several disease conditions including diabetes. This study examined the potential palliative effect of aqueous seed extract of Picralima nitida (APN) on dyslipidemia, hyperglycemia, oxidative stress, insulin resistance, and the expression of some metabolic genes in high-fat high-fructose-fed rats. Experimental rats (2 months old) were fed a control diet or a high-fat diet with 25% fructose (HFHF diet) in their drinking water for nine weeks. APN was administered orally during the last four weeks. Anthropometric and antioxidant parameters, lipid profile, plasma glucose, and insulin levels and the relative expression of some metabolic genes were assessed. APN caused a significant decrease (P < 0.05) in weight gained, body mass index, insulin resistance, plasma glucose, and insulin levels. High-density lipoprotein cholesterol level was significantly increased (P < 0.05), while triacylglycerol, cholesterol, low-density lipoprotein, cardiac index, atherogenic index, coronary artery index, and malondialdehyde levels in plasma and liver samples were also significantly decreased (P < 0.05) by APN at all experimental doses when compared to the group fed with an HFHF diet only. APN also significantly (P < 0.05) upregulated the relative expression of glucokinase, carnitine palmitoyltransferase-1 (CPT-1), and leptin at 400 mg/kg body weight when compared to the group fed with an HFHF diet only. This study showed that APN alleviated dyslipidemia, hyperglycemia, and oxidant effect associated with the intake of a high-fat high-fructose diet.
Collapse
|
32
|
Miyamoto JÉ, Reginato A, Portovedo M, Dos Santos RM, Stahl MA, Le Stunff H, Latorraca MQ, de Barros Reis MA, Arantes VC, Doneda DL, Ignacio-Souza LM, Torsoni AS, Grimaldi R, Ribeiro APB, Torsoni MA, Milanski M. Interesterified palm oil impairs glucose homeostasis and induces deleterious effects in liver of Swiss mice. Metabolism 2020; 112:154350. [PMID: 32910938 DOI: 10.1016/j.metabol.2020.154350] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 06/30/2020] [Accepted: 08/30/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Interesterified fats have largely replaced the partially hydrogenated oils which are the main dietary source of trans fat in industrialized food. This process promotes a random rearrangement of the native fatty acids and the results are different triacylglycerol (TAG) molecules without generating trans isomers. The role of interesterified fats in metabolism remains unclear. We evaluated metabolic parameters, glucose homeostasis and inflammatory markers in mice fed with normocaloric and normolipidic diets or hypercaloric and high-fat diet enriched with interesterified palm oil. METHODS Male Swiss mice were randomly divided into four experimental groups and submitted to either normolipidic palm oil diet (PO), normolipidic interesterified palm oil diet (IPO), palm oil high-fat diet (POHF) or interesterified palm oil high-fat diet (IPOHF) during an 8 weeks period. RESULTS When compared to the PO group, IPO group presented higher body mass, hyperglycemia, impaired glucose tolerance, evidence of insulin resistance and greater production of glucose in basal state during pyruvate in situ assay. We also observed higher protein content of hepatic PEPCK and increased cytokine mRNA expression in the IPO group when compared to PO. Interestingly, IPO group showed similar parameters to POHF and IPOHF groups. CONCLUSION The results indicate that substitution of palm oil for interesterified palm oil even on normocaloric and normolipidic diet could negatively modulate metabolic parameters and glucose homeostasis as well as cytokine gene expression in the liver and white adipose tissue. This data support concerns about the effects of interesterified fats on health and could promote further discussions about the safety of the utilization of this unnatural fat by food industry.
Collapse
Affiliation(s)
- Josiane Érica Miyamoto
- School of Applied Sciences, University of Campinas, UNICAMP, Limeira, Brazil; Obesity and Comorbidities Research Center, University of Campinas, UNICAMP, Campinas, Brazil
| | - Andressa Reginato
- School of Applied Sciences, University of Campinas, UNICAMP, Limeira, Brazil; Obesity and Comorbidities Research Center, University of Campinas, UNICAMP, Campinas, Brazil
| | - Mariana Portovedo
- School of Applied Sciences, University of Campinas, UNICAMP, Limeira, Brazil; Obesity and Comorbidities Research Center, University of Campinas, UNICAMP, Campinas, Brazil
| | - Raísa Magno Dos Santos
- School of Applied Sciences, University of Campinas, UNICAMP, Limeira, Brazil; Obesity and Comorbidities Research Center, University of Campinas, UNICAMP, Campinas, Brazil
| | | | - Hervé Le Stunff
- Paris-Saclay Institute of Neuroscience, CNRS UMR 9197, Université Paris-Sud, University Paris Saclay, Orsay, France
| | | | | | | | - Diego Luiz Doneda
- Physiology Laboratory, Department of Basic Health Sciences, Federal University of Mato Grosso, Cuiabá, Brazil
| | - Leticia Martins Ignacio-Souza
- School of Applied Sciences, University of Campinas, UNICAMP, Limeira, Brazil; Obesity and Comorbidities Research Center, University of Campinas, UNICAMP, Campinas, Brazil
| | - Adriana Souza Torsoni
- School of Applied Sciences, University of Campinas, UNICAMP, Limeira, Brazil; Obesity and Comorbidities Research Center, University of Campinas, UNICAMP, Campinas, Brazil
| | - Renato Grimaldi
- School of Food Engineering, University of Campinas, UNICAMP, Campinas, Brazil
| | | | - Marcio Alberto Torsoni
- School of Applied Sciences, University of Campinas, UNICAMP, Limeira, Brazil; Obesity and Comorbidities Research Center, University of Campinas, UNICAMP, Campinas, Brazil
| | - Marciane Milanski
- School of Applied Sciences, University of Campinas, UNICAMP, Limeira, Brazil; Obesity and Comorbidities Research Center, University of Campinas, UNICAMP, Campinas, Brazil.
| |
Collapse
|
33
|
Chen F, Zhou Y, Yang K, Shen M, Wang Y. NPY stimulates cholesterol synthesis acutely by activating the SREBP2-HMGCR pathway through the Y1 and Y5 receptors in murine hepatocytes. Life Sci 2020; 262:118478. [PMID: 32976883 DOI: 10.1016/j.lfs.2020.118478] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 09/07/2020] [Accepted: 09/17/2020] [Indexed: 12/12/2022]
Abstract
AIMS The development of non-alcoholic fatty liver disease (NAFLD) is partially attributed to disturbance in cholesterol metabolism and sympathetic overactivity. Excessive levels of the sympathetic neurotransmitter neuropeptide Y (NPY) positively correlated with both NAFLD and cholesterol accumulation. We wanted to determine, for the first time, whether NPY promotes cholesterol accumulation directly in hepatocytes and elucidate the underlying mechanism. MAIN METHODS In vivo, NPY was injected through the hepatic portal vein into SD rats. One hour later, serum and liver tissues were collected. In vitro, BRL-3A hepatocytes were treated with NPY, and with Y1, Y2, Y5, receptor antagonists as well as with extracellular signal-regulated protein kinase 1 and 2 (ERK1/2) antagonist, respectively. Cholesterol content was measured by coupled enzyme method. Precursor sterol-regulatory element binding protein 2 (pSREBP2), mature SREBP2 (mSREBP2), HMG-CoA reductase (HMGCR), ERK1/2, pERK1/2, cAMP-dependent protein kinase (PKA), and pPKA protein expression levels were examined by western blotting. KEY FINDINGS In rats, intraportal vein injection of NPY activates pSREBP2, mSREBP2, and HMGCR protein expression, and induces hepatic cholesterol accumulation. In BRL-3A cells, we observed that NPY increases cholesterogenic protein expression and cholesterol synthesis through Y1 and Y5 receptors. This effect is mediated by the activation of the ERK1/2 signaling pathway. SIGNIFICANCE We demonstrated, for the first time, that NPY can activate the cholesterogenic pathway and elucidated the underlying mechanism. Thus, NPY and NPY receptors might be new targets for the treatment of NAFLD and dyslipidemia.
Collapse
Affiliation(s)
- Fu Chen
- Department of General Surgery, Fourth Affiliated Hospital of China Medical University, No. 4, Chongshan East Road, Huanggu District, Shenyang City, Liaoning Province 110032, China
| | - Yong Zhou
- Department of General Surgery, Fourth Affiliated Hospital of China Medical University, No. 4, Chongshan East Road, Huanggu District, Shenyang City, Liaoning Province 110032, China
| | - Keyu Yang
- Department of General Surgery, Fourth Affiliated Hospital of China Medical University, No. 4, Chongshan East Road, Huanggu District, Shenyang City, Liaoning Province 110032, China
| | - Mingyang Shen
- Department of General Surgery, Fourth Affiliated Hospital of China Medical University, No. 4, Chongshan East Road, Huanggu District, Shenyang City, Liaoning Province 110032, China
| | - Yong Wang
- Department of General Surgery, Fourth Affiliated Hospital of China Medical University, No. 4, Chongshan East Road, Huanggu District, Shenyang City, Liaoning Province 110032, China.
| |
Collapse
|
34
|
Neuroprotective role of alendronate against APP processing and neuroinflammation in mice fed a high fat diet. Brain Res Bull 2020; 161:197-212. [PMID: 32389800 DOI: 10.1016/j.brainresbull.2020.04.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 04/04/2020] [Accepted: 04/16/2020] [Indexed: 12/17/2022]
Abstract
Obesity and consumption of diet rich in fat are known to contribute to the development of Alzheimer's disease (AD) which is a complex and multifactorial neurodegenerative disease and a leading cause of mortality with unmet medical needs. Hypercholesterolemia was discovered to increase neuropathological changes along with cognitive decline in AD mouse models but still the underlying mechanism is elusive. Furthermore, isoprenoids, the crucial products of Mevalonate-pathway produced by the action of farnesyl pyrophosphate synthase (FPPS) enzyme, are also demonstrated to play a key role in AD. Nevertheless, bisphosphonates target this enzyme in order to treat osteoporosis and also found to alleviate dementia in such patients. As per the cited inhibitory action of alendronate, against acetylcholinesterase and cholesterol level, we hypothesized to explore the potential of alendronate against high fat diet (HFD) induced neuropathologies and cognitive disabilities in AD mouse model. Here we noticed that in mice provided with HFD for 14 weeks, spatial memory was compromised as interpreted in different behavioral paradigms. Together with cognitive depletion, there was observed a provoking effect on amyloid precursor protein (APP)-processing via amyloidogenic pathway due to enhanced β-site APP cleaving enzyme-1 (BACE-1) level which in turn leads to augmented release of amyloid beta (Aβ) in hippocampus of HFD mice. Relevant to these, significant elevation in hippocampal level of neuroinflammatory cytokines, oxidative stress markers and isoprenoids and serum cholesterol were also found after HFD exposure. Marked reversal of cognitive impairment, enhanced APP-processing, neuroinflammation along with other neuropathological alterations in hippocampus was demonstrated following oral administration of alendronate (1.76 mg/kg) for 15 days despite of HFD treatment. These changes were noted to be due to modulation of isoprenoids and cholesterol level by alendronate. Supporting these, histopathological analysis done by congo red revealed the reduced Aβ deposition in hippocampus of drug treated HFD mice The current outcomes provide important implications for the contribution of Mevalonate-pathway and HFD for the onset of AD and also support alendronate as a prominent intervention for amelioration of AD-like pathologies.
Collapse
|
35
|
Abstract
Currently, the prevention and treatment of CVD have been a global focus since CVD is the number one cause of mortality and morbidity. In the pathogenesis of CVD, it was generally thought that impaired cholesterol homeostasis might be a risk factor. Cholesterol homeostasis is affected by exogenous factors (i.e. diet) and endogenous factors (i.e. certain receptors, enzymes and transcription factors). In this context, the number of studies investigating the potential mechanisms of dietary fatty acids on cholesterol homeostasis have increased in recent years. As well, the cluster of differentiation 36 (CD36) receptor is a multifunctional membrane receptor involved in fatty acid uptake, lipid metabolism, atherothrombosis and inflammation. CD36 is proposed to be a crucial molecule for cholesterol homeostasis in various mechanisms including absorption/reabsorption, synthesis, and transport of cholesterol and bile acids. Moreover, it has been reported that the amount of fatty acids and fatty acid pattern of the diet influence the CD36 level and CD36-mediated cholesterol metabolism principally in the liver, intestine and macrophages. In these processes, CD36-mediated cholesterol and lipoprotein homeostasis might be impaired by dietary SFA and trans-fatty acids, whereas ameliorated by MUFA in the diet. The effects of PUFA on CD36-mediated cholesterol homeostasis are controversial depending on the amount of n-3 PUFA and n-6 PUFA, and the n-3:n-6 PUFA ratio. Thus, since the CD36 receptor is suggested to be a novel nutrient-sensitive biomarker, the role of CD36 and dietary fatty acids in cholesterol metabolism might be considered in medical nutrition therapy in the near future. Therefore, the novel nutritional target of CD36 and interventions that focus on dietary fatty acids and potential mechanisms underlying cholesterol homeostasis are discussed in this review.
Collapse
|
36
|
Madduma Hewage S, Prashar S, Debnath SC, O K, Siow YL. Inhibition of Inflammatory Cytokine Expression Prevents High-Fat Diet-Induced Kidney Injury: Role of Lingonberry Supplementation. Front Med (Lausanne) 2020; 7:80. [PMID: 32292787 PMCID: PMC7119336 DOI: 10.3389/fmed.2020.00080] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 02/25/2020] [Indexed: 12/21/2022] Open
Abstract
Chronic low-grade inflammation is a major stimulus for progression of chronic kidney disease (CKD) in individuals consuming high-fat diet. Currently, there are limited treatment options for CKD other than controlling the progression rate and its associated complications. Lingonberry (Vaccinium vitis-idaea L.) is rich in anthocyanins with demonstrated anti-inflammatory effect. In the current study, we investigated the potential renal protective effect of lingonberry and its anthocyanin (cyanidin-3-glucoside) in high-fat diet fed obese mice and in human proximal tubular cells. Prolonged consumption of high-fat diets is strongly associated with obesity, abnormal lipid and glucose metabolism. Mice (C57BL/6J) fed a high-fat diet (62% kcal fat) for 12 weeks developed renal injury as indicated by an elevation of blood urea nitrogen (BUN) level as well as an increase in renal kidney injury molecule-1 (KIM-1), neutrophil gelatinase-associated lipocalin (NGAL) and renin expression. Those mice displayed an activation of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and increased expression of inflammatory cytokines-monocyte chemoattractant-1 (MCP-1), tumor necrosis factor alpha (TNF-α), interleukin-6 (IL-6) in the kidneys. Mice fed a high-fat diet also had a significant elevation of inflammatory cytokine levels in the plasma. Dietary supplementation of lingonberry for 12 weeks not only attenuated high-fat diet-induced renal inflammatory response but also reduced kidney injury. Such a treatment improved plasma lipid and glucose profiles, reduced plasma inflammatory cytokine levels but did not affect body weight gain induced by high-fat diet feeding. Lingonberry extract or its active component cyanidin-3-glucoside effectively inhibited palmitic acid-induced NF-κB activation and inflammatory cytokine expression in proximal tubular cells. These results suggest that lingonberry supplementation can reduce inflammatory response and prevent chronic kidney injury. Such a renal protective effect by lingonberry and its active component may be mediated, in part, through NF-κB signaling pathway.
Collapse
Affiliation(s)
- Susara Madduma Hewage
- Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB, Canada.,Department of Physiology & Pathophysiology, University of Manitoba, Winnipeg, MB, Canada
| | - Suvira Prashar
- Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB, Canada.,Agriculture and Agri-Food Canada, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB, Canada
| | - Samir C Debnath
- Agriculture and Agri-Food Canada, St. John's Research and Development Centre, St. John's, NL, Canada
| | - Karmin O
- Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB, Canada.,Department of Physiology & Pathophysiology, University of Manitoba, Winnipeg, MB, Canada.,Department of Animal Science, University of Manitoba, Winnipeg, MB, Canada
| | - Yaw L Siow
- Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB, Canada.,Department of Physiology & Pathophysiology, University of Manitoba, Winnipeg, MB, Canada.,Agriculture and Agri-Food Canada, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB, Canada
| |
Collapse
|
37
|
Zhao Y, Cao X, Fu L, Gao J. n-3 PUFA reduction caused by fabp2 deletion interferes with triacylglycerol metabolism and cholesterolhomeostasis in fish. Appl Microbiol Biotechnol 2020; 104:2149-2161. [PMID: 31950220 DOI: 10.1007/s00253-020-10366-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 12/29/2019] [Accepted: 01/09/2020] [Indexed: 01/20/2023]
Abstract
Fatty acid-binding protein 2 (Fabp2), which is involved in the transport of long-chain fatty acids, is widely studied in mammals. Nevertheless, the role of this protein in teleost fish is mostly unknown. Here, we produced a fabp2-/- zebrafish (KO) animal model. Compared with wild-type zebrafish (WT), KO had a markedly decreased content of intestinal n-3 poly-unsaturated fatty acids (n-3 PUFAs) and increased levels of intestinal, hepatic, and serum triacylglycerols (TAG). The intestinal transcriptome analysis of KO and WT revealed an obviously disrupted TAG metabolism and up-regulated bile secretion in KO. Expression levels of the genes related to fatty acid transport and cholesterol (CL) absorption in the intestine of KO were significantly lower than those of WT, while the expression levels of genes related to intestinal TAG synthesis and hepatic CL synthesis were in the opposite direction. To confirm these findings, we further established fabp2 transgenic zebrafish (TG). Compared with WT, TG had a markedly increased content of intestinal n-3 PUFAs, a significantly decreased level of hepatic TAG, and significantly higher expression of genes related to fatty acid transport and CL absorption in the intestine. In conclusion, this study suggests that teleost fish fabp2 could promote intestinal n-3 PUFA absorption to mediate TAG synthesis and CL homeostasis, by regulating the genes involved in lipid metabolism.
Collapse
Affiliation(s)
- Yan Zhao
- College of Fisheries, Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education/Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, 430070, China
| | - Xiaojuan Cao
- College of Fisheries, Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education/Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, 430070, China
- Hubei Provincial Engineering Laboratory for Pond Aquaculture, No. 1 Shizishan Stress, Hongshan District, Wuhan, 430070, Hubei Province, China
| | - Lele Fu
- College of Fisheries, Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education/Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, 430070, China
| | - Jian Gao
- College of Fisheries, Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education/Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, 430070, China.
- Hubei Provincial Engineering Laboratory for Pond Aquaculture, No. 1 Shizishan Stress, Hongshan District, Wuhan, 430070, Hubei Province, China.
| |
Collapse
|
38
|
Chen G, Wu K, Zhao T, Ling S, Liu W, Luo Z. miR-144 Mediates High Fat-Induced Changes of Cholesterol Metabolism via Direct Regulation of C/EBPα in the Liver and Isolated Hepatocytes of Yellow Catfish. J Nutr 2020; 150:464-474. [PMID: 31724712 DOI: 10.1093/jn/nxz282] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 09/11/2019] [Accepted: 10/24/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND microRNAs (miRNAs) post-transcriptionally regulate gene expression and act as important modulators of cholesterol homeostasis. OBJECTIVE The study explores the mechanism by which miRNAs mediate high fat-induced changes of cholesterol metabolism in yellow catfish. METHODS Yellow catfish (weight: 3.79 ± 0.16 g, 3 mo old, mixed sex) were fed 2 diets containing lipids at 11.3% [control (CON)] or 15.4% [high-fat diet (HFD)] (by weight) for 8 wk. Cholesterol content was measured; hematoxylin-eosin (H&E) staining, qPCR assays, and small RNA sequencing were conducted in the liver. Hepatocytes were isolated from separate, untreated fish and incubated for 24 h in control solution or palmitic acid (PA; 0.25 mM)/oleic acid (OA; 0.5 mM) after 4 h pretreatment with or without miR-144 inhibitor/mimic (40 nM). Cholesterol content was measured; qPCR assays and Western blotting were conducted in the hepatocytes. HEK293T cells were co-transfected with plasmids to validate miR-144 target genes. The promoter activities of miR-144 were analyzed in HEK293T cells with PA (0.25 mM) or OA (0.25 or 0.5 mM) treatment for 24 h. Luciferase activity assays, electrophoretic mobility shift assay, and Western blotting were conducted in HEK293T cells. RESULTS Compared with CON, HFD induced hepatic cholesterol accumulation (31.5%), and upregulated miR-144 expression (8.40-fold, P < 0.05). miR-144 directly targeted hydroxymethylglutaryl-CoA reductase (hmgcr), cholesterol 7α-monooxygenase A1 (cyp7a1), and adenosine triphosphate binding cassette transporter A1 (abca1) in HEK293T cells. In the hepatocytes of yellow catfish, miR-144 inversely regulated the expression of hmgcr, cyp7a1, and abca1 (30.3-78.5%, P < 0.05); loss of miR-144 function alleviated PA- or OA-induced cholesterol accumulation (19.5-61.1%, P < 0.05). We also characterized the C/EBPα binding site in the miR-144 promoter, and found that C/EBPα positively regulated miR-144 expression through binding to the miR-144 promoter. CONCLUSIONS miR-144 mediated HFD-induced changes in the liver and hepatocytes of yellow catfish, suggesting a possible mechanism for HFD-induced dysfunction in cholesterol metabolism.
Collapse
Affiliation(s)
- Guanghui Chen
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Fishery College, Huazhong Agricultural University, Wuhan, China
| | - Kun Wu
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Fishery College, Huazhong Agricultural University, Wuhan, China
| | - Tao Zhao
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Fishery College, Huazhong Agricultural University, Wuhan, China
| | - Shicheng Ling
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Fishery College, Huazhong Agricultural University, Wuhan, China
| | - Wei Liu
- Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan, China
| | - Zhi Luo
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Fishery College, Huazhong Agricultural University, Wuhan, China.,Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| |
Collapse
|
39
|
Xu Y, Ai C, Jiang P, Sun X, Liu Y, Jiang G, Song S. Oligosaccharides from Gracilaria lemaneiformis better attenuated high fat diet-induced metabolic syndrome by promoting the Bacteroidales proliferation. Food Funct 2020; 11:1049-1062. [DOI: 10.1039/c9fo01996k] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Reduction in the degree of polymerization of polysaccharides can improve its bioactivity.
Collapse
Affiliation(s)
- Yuxin Xu
- School of Food Science and Technology; National Engineering Research Center of Seafood
- Dalian Polytechnic University
- Dalian 116034
- P. R. China
| | - Chunqing Ai
- School of Food Science and Technology; National Engineering Research Center of Seafood
- Dalian Polytechnic University
- Dalian 116034
- P. R. China
- National & Local Joint Engineering Laboratory for Marine Bioactive Polysaccharide Development and Application
| | - Pingrui Jiang
- School of Food Science and Technology; National Engineering Research Center of Seafood
- Dalian Polytechnic University
- Dalian 116034
- P. R. China
| | - Xiaona Sun
- School of Food Science and Technology; National Engineering Research Center of Seafood
- Dalian Polytechnic University
- Dalian 116034
- P. R. China
| | - Yili Liu
- School of Food Science and Technology; National Engineering Research Center of Seafood
- Dalian Polytechnic University
- Dalian 116034
- P. R. China
| | - Guoping Jiang
- School of Food Science and Technology; National Engineering Research Center of Seafood
- Dalian Polytechnic University
- Dalian 116034
- P. R. China
| | - Shuang Song
- School of Food Science and Technology; National Engineering Research Center of Seafood
- Dalian Polytechnic University
- Dalian 116034
- P. R. China
- National & Local Joint Engineering Laboratory for Marine Bioactive Polysaccharide Development and Application
| |
Collapse
|
40
|
Transfer of the Septin Ring to Cytokinetic Remnants in ER Stress Directs Age-Sensitive Cell-Cycle Re-entry. Dev Cell 2019; 51:173-191.e5. [PMID: 31564614 DOI: 10.1016/j.devcel.2019.08.017] [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: 11/17/2018] [Revised: 05/19/2019] [Accepted: 08/23/2019] [Indexed: 02/06/2023]
Abstract
During cell division, the inheritance of a functional endoplasmic reticulum (ER) is ensured by the endoplasmic reticulum stress surveillance (ERSU) pathway. Activation of ERSU causes the septin ring to mislocalize, which blocks ER inheritance and cytokinesis. Here, we uncover that the septin ring in fact translocates to previously utilized cell division sites called cytokinetic remnants (CRMs). This unconventional translocation requires Nba1, a negative polarity regulator that normally prevents repolarization and re-budding at CRMs. Furthermore, septin ring translocation relies on the recruitment and activation of a key ERSU component Slt2 by Bem1, without activating Cdc42. Failure to transfer all septin subunits to CRMs delays the cell's ability to re-enter the cell cycle when ER homeostasis is restored and hinders cell growth after ER stress recovery. Thus, these deliberate but unprecedented rearrangements of cell polarity factors during ER stress safeguard cell survival and the timely cell-cycle re-entry upon ER stress recovery.
Collapse
|
41
|
Wang T, Xiao X, Regenstein JM, Wu W, Zhou Y, Wang S, Cheng Y, Wu X, Bao B. Effect on lipid metabolism of mice continuously fed a crab-containing diet. FOOD BIOSCI 2019. [DOI: 10.1016/j.fbio.2019.100422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
42
|
Wang K, Yu XH, Tang YJ, Tang YL, Liang XH. Obesity: An emerging driver of head and neck cancer. Life Sci 2019; 233:116687. [PMID: 31348948 DOI: 10.1016/j.lfs.2019.116687] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Revised: 07/22/2019] [Accepted: 07/22/2019] [Indexed: 02/05/2023]
Abstract
Obesity has become pandemic and emerged as one of the most critical global health care problems worldwide since last century. Recent studies have demonstrated that there may be a causal link between obesity and higher risks and mortality of cancers, including prostate, breast, colon, and thyroid cancers, head and neck cancer (HNC). This review focuses on the relationship between obesity and HNC, and the molecular mechanism of abnormal lipid metabolism in HNC. Elucidating the mechanism may open up new possibilities for strategies to reduce risk and mortality of HNC in an increasingly obese population.
Collapse
Affiliation(s)
- Ke Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Oral And Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Xiang-Hua Yu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Oral And Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Ya-Jie Tang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China; Hubei Key Laboratory of Industrial Microbiology, Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China.
| | - Ya-Ling Tang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Oral Pathology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| | - Xin-Hua Liang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Oral And Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
43
|
Jones RS, Tu C, Zhang M, Qu J, Morris ME. Characterization and Proteomic-Transcriptomic Investigation of Monocarboxylate Transporter 6 Knockout Mice: Evidence of a Potential Role in Glucose and Lipid Metabolism. Mol Pharmacol 2019; 96:364-376. [PMID: 31436537 DOI: 10.1124/mol.119.116731] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 06/27/2019] [Indexed: 12/14/2022] Open
Abstract
Monocarboxylate transporter 6 [(MCT6), SLC16A5] is an orphan transporter with no known endogenous substrates or physiological role. Previous in vitro and in vivo experiments investigated MCT6 substrate/inhibitor specificity in Xenopus laevis oocytes; however, these data remain limited. Transcriptomic changes in the livers of mice undergoing different dieting schemes have suggested that Mct6 plays a role in glucose and lipid metabolism. The objectives of this study were 1) to develop a novel knockout (KO) mouse model (Mct6-/-) using CRISPR/Cas9 technology, 2) to characterize the KO animal model by examining physiological and biochemical parameters, and 3) to understand the physiological role of MCT6 in vivo through global proteomic and liver transcriptomic profiling. mRNA tissue analysis demonstrated knockout of Mct6, which showed greater than 90% knockdown of Mct6 (Slc16a5) gene expression in all major tissues analyzed when normalized to Mct6+/+ mice. Proteomic analyses identified greater than 4000 unique proteins in kidney, liver, and colon tissues, among which 51, 38, and 241 proteins were significantly altered, respectively (for each tissue), between Mct6+/+ and Mct6-/- mice. Additionally, Mct6-/- mice demonstrated significant changes in 199 genes in the liver compared with Mct6+/+ mice. In silico biological pathway analyses revealed significant changes in proteins and genes involved in glucose and lipid metabolism-associated pathways. This study is the first to provide evidence for an association of Mct6 in the regulation of glucose and lipid metabolism. SIGNIFICANCE STATEMENT: This paper focuses on elucidating the innate biological role of an orphan transporter in vivo, which has not been investigated thus far. Using efficient and high-throughput technologies, such as CRISPR/Cas9 gene editing, liquid chromatography-tandem mass spectrometry-based proteomic and RNA-sequencing transcriptomic analyses, our laboratory provides the first existence and characterization of a Mct6 knockout mouse model. The evidence gathered in this paper, as well as other laboratories, support the importance of MCT6 in regulating a variety of glucose and lipid metabolic pathways, which may indicate its significance in metabolic diseases.
Collapse
Affiliation(s)
- Robert S Jones
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, New York (R.S.J., C.T., J.Q., M.E.M.); and New York State Center of Excellence in Bioinformatics and Life Sciences, Buffalo, New York (C.T., M.Z., J.Q.)
| | - Chengjian Tu
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, New York (R.S.J., C.T., J.Q., M.E.M.); and New York State Center of Excellence in Bioinformatics and Life Sciences, Buffalo, New York (C.T., M.Z., J.Q.)
| | - Ming Zhang
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, New York (R.S.J., C.T., J.Q., M.E.M.); and New York State Center of Excellence in Bioinformatics and Life Sciences, Buffalo, New York (C.T., M.Z., J.Q.)
| | - Jun Qu
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, New York (R.S.J., C.T., J.Q., M.E.M.); and New York State Center of Excellence in Bioinformatics and Life Sciences, Buffalo, New York (C.T., M.Z., J.Q.)
| | - Marilyn E Morris
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, New York (R.S.J., C.T., J.Q., M.E.M.); and New York State Center of Excellence in Bioinformatics and Life Sciences, Buffalo, New York (C.T., M.Z., J.Q.)
| |
Collapse
|
44
|
Zhu Z, Huang Y, Luo X, Wu Q, He J, Li S, Barba FJ. Modulation of lipid metabolism and colonic microbial diversity of high-fat-diet C57BL/6 mice by inulin with different chain lengths. Food Res Int 2019; 123:355-363. [PMID: 31284986 DOI: 10.1016/j.foodres.2019.05.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 04/30/2019] [Accepted: 05/02/2019] [Indexed: 12/28/2022]
Abstract
The physicochemical properties, biological functions and microbial degradation of inulins differ according to their degree of polymerization. However, the relationship between inulin activities and its effect on gut microbiota remains unknown. In this study, high fat diet with inulin (1 or 5 g/kg·bw), either with short or long chains groups were administered to different groups of mice (n = 10) for 10 weeks in order to investigate the effect of inulin on the microbial diversity of the animals. Litchi pericarp procyanidins (LPPC) were used for comparison purposes. Furthermore, the lipid metabolism and key regulator genes in mice were determined. The results indicated that natural inulin (1 g/kg·bw) ingestion reduced the body weight of fat mice between week 6-9. Glutathione peroxidase (GSH-Px) activity in liver was remarkably higher after adding long chain inulin (5 g/kg·bw) compared to high-fat-diet mice. Moreover, high dose of natural inulin regulated malondialdehyde and advanced glycation end-products levels in mice liver. Likewise, the high dose of short-chain inulin increased sterol response element binding protein 1 (SREBP-1), β-Hydroxy β-methylglutaryl-CoA (HMG-CoA) and ATP-binding cassette transporter A1 (ABCA1) genetic expression. A significant change on the abundance of six genera in gut microbial profile suggested that inulin has the ability to modulate the lipid metabolism regardless of chain length, mainly due to its impact on colon microbiota variety.
Collapse
Affiliation(s)
- Zhenzhou Zhu
- College of Food Science and Engineering, Wuhan Polytechnic University, Wuhan 430023, China; Hubei Key Laboratory for Processing and Transformation of Agricultural Products, Wuhan Polytechnic University, Wuhan 430023, China
| | - Yuqi Huang
- College of Food Science and Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Xiao Luo
- College of Food Science and Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Qian Wu
- College of Food Science and Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Jingren He
- College of Food Science and Engineering, Wuhan Polytechnic University, Wuhan 430023, China; Hubei Key Laboratory for Processing and Transformation of Agricultural Products, Wuhan Polytechnic University, Wuhan 430023, China
| | - Shuyi Li
- College of Food Science and Engineering, Wuhan Polytechnic University, Wuhan 430023, China; Hubei Key Laboratory for Processing and Transformation of Agricultural Products, Wuhan Polytechnic University, Wuhan 430023, China.
| | - Francisco J Barba
- Nutrition and Food Science Area, Preventive Medicine and Public Health, Food Sciences, Toxicology and Forensic Medicine Department, Faculty of Pharmacy, Universitat de València, Avda. Vicent Andrés Estellés, s/n, 46100 Burjassot, València, Spain.
| |
Collapse
|
45
|
Acharya P, Talahalli RR. n-3 Fatty Acids Abrogate Dyslipidemia-Induced Changes in Bile Acid Uptake, Synthesis, and Transport in Young and Aged Dyslipidemic Rats. Lipids 2019; 54:39-51. [DOI: 10.1002/lipd.12125] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Revised: 11/01/2018] [Accepted: 12/20/2018] [Indexed: 01/05/2023]
Affiliation(s)
- Pooja Acharya
- Department of Biochemistry; CSIR-Central Food Technological Research Institute; Mysore, 570020 Karnataka India
- Academy of Scientific and Innovative Research (AcSIR); Ghaziabad, 201002 Uttar Pradesh India
| | - Ramaprasad R. Talahalli
- Department of Biochemistry; CSIR-Central Food Technological Research Institute; Mysore, 570020 Karnataka India
- Academy of Scientific and Innovative Research (AcSIR); Ghaziabad, 201002 Uttar Pradesh India
| |
Collapse
|
46
|
Li Q, Zhang H, Zou J, Feng X, Feng D. Bisphenol A induces cholesterol biosynthesis in HepG2 cells via SREBP-2/HMGCR signaling pathway. J Toxicol Sci 2019; 44:481-491. [DOI: 10.2131/jts.44.481] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Affiliation(s)
- Qingrong Li
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-sen University, China
| | - Hongmin Zhang
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-sen University, China
| | - Jun Zou
- Department of Cardiology, Affiliated NanHai Hospital of Southern Medical University, China
| | - Xiang Feng
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-sen University, China
| | - Dan Feng
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-sen University, China
- Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, China
| |
Collapse
|
47
|
Tang R, Yang Q, Lin S, Feng Y, Yang J, Lv Q, Wu G, Hu J. Preventive or Curative Administration of Taurine Regulates Lipid Metabolism in the Liver of Rats with Alcoholic Liver Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1155:119-131. [PMID: 31468391 DOI: 10.1007/978-981-13-8023-5_11] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Excessive consumption causes alcoholic liver disease (ALD), which injures hepatocytes and induces imbalance of lipid metabolism. Taurine is known to protect the liver from various liver injuries, and relieve lipid profile. Our previous studies also found that taurine can prevent or cure ALD, reduce fat deposition, but the mechanism remains unclear. In the present study, ALD rat model was established by administration of alcohol, pyrazole and high fat diet. Two percent taurine was administered at the same time or after ALD model establishment. Serum activities of alanine aminotransferase (ALT) and aspartate aminotransferase (AST), serum and hepatic TC, TG, HDL-C and LDL-C were analyzed. Real-Time RT-PCR was conducted to detect the mRNA expressions of fatty acid synthetase (FAS), acetyl-CoA catboxylase (ACC), carnitine palmitoyl transferase 1 (CPT-1), 3-Hydroxy-3-methyl glutaric acid acyl Coenzyme A reductase (HMGCR), peroxisome proliferators activated receptor α (PPARα) and sterol regulatory element-binding protein 1c (SREBP-1c). The results showed that serum ALT, AST, serum and hepatic TC, TG and LDL-C were higher, while HDL-C in ALD model rats was lower than normal rats, the changes of which can be significantly relieved by taurine administration. mRNA expressions of ACC, FAS, CPT-1, HMGCR, PPARα and SREBP-1c which were significantly changed by ethanol can also be regulated by taurine. The results indicated that taurine can prevent and repair hepatic injury of ALD rats and balance lipid metabolism indexes in the liver, the mechanisms may involves in the regulation of related enzymes and transcriptional regulators participated in lipid metabolism.
Collapse
Affiliation(s)
- Riyi Tang
- Liaoning Provincial Key Laboratory of Zoonosis, College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang, Liaoning, People's Republic of China
| | - Qunhui Yang
- Liaoning Provincial Key Laboratory of Zoonosis, College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang, Liaoning, People's Republic of China
| | - Shumei Lin
- Liaoning Provincial Key Laboratory of Zoonosis, College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang, Liaoning, People's Republic of China
| | - Ying Feng
- Liaoning Provincial Key Laboratory of Zoonosis, College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang, Liaoning, People's Republic of China
| | - Jiancheng Yang
- Liaoning Provincial Key Laboratory of Zoonosis, College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang, Liaoning, People's Republic of China
| | - Qiufeng Lv
- Liaoning Provincial Key Laboratory of Zoonosis, College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang, Liaoning, People's Republic of China
| | - Gaofeng Wu
- Liaoning Provincial Key Laboratory of Zoonosis, College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang, Liaoning, People's Republic of China
| | - Jianmin Hu
- Liaoning Provincial Key Laboratory of Zoonosis, College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang, Liaoning, People's Republic of China
| |
Collapse
|
48
|
Sid V, Shang Y, Siow YL, Hewage SM, House JD, O K. Folic Acid Supplementation Attenuates Chronic Hepatic Inflammation in High-Fat Diet Fed Mice. Lipids 2018; 53:709-716. [PMID: 30252139 DOI: 10.1002/lipd.12084] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 08/09/2018] [Accepted: 08/20/2018] [Indexed: 02/06/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common form of chronic liver disease worldwide. Hepatic inflammation is an important pathogenic mediator of NAFLD. There is currently no pharmacological agent approved for the treatment of NAFLD. Folic acid is a water-soluble B vitamin that has been shown to have lipid-lowering and antioxidant effects. The objective of this study was to investigate the effect of folic acid supplementation on hepatic inflammation and to identify the underlying mechanisms. Male C57BL/6 J mice were fed a control diet (10% kcal fat), a high-fat diet (HFD) (60% kcal fat), or a HFD supplemented with folic acid (26 mg/kg diet) for 8 weeks. HFD feeding led to increased body mass gain, lipid accumulation, activation of transcription factor nuclear factor-κB (NF-κB), and elevation of inflammatory cytokine gene expression in the liver. Folic acid supplementation attenuated hepatic lipid accumulation and aggregation of inflammatory foci induced by HFD feeding. This was associated with a significant reduction of NF-κB activation and inflammatory cytokine expression. These results suggest that the hepatoprotective effect of folic acid in NAFLD may be attributed, in part, to its anti-inflammatory action.
Collapse
Affiliation(s)
- Victoria Sid
- Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Research Centre, 351 Tache Avenue, Winnipeg, MB, Canada.,Department of Physiology and Pathophysiology, University of Manitoba, 727 McDermot Avenue, Winnipeg, MB, Canada
| | - Yue Shang
- Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Research Centre, 351 Tache Avenue, Winnipeg, MB, Canada.,Department of Animal Science, University of Manitoba, 12 Dafoe Road, Winnipeg, MB, Canada
| | - Yaw L Siow
- Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Research Centre, 351 Tache Avenue, Winnipeg, MB, Canada.,Department of Physiology and Pathophysiology, University of Manitoba, 727 McDermot Avenue, Winnipeg, MB, Canada.,Agriculture and Agri-Food Canada, 351 Tache Avenue, Winnipeg, MB, Canada
| | - Susara Madduma Hewage
- Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Research Centre, 351 Tache Avenue, Winnipeg, MB, Canada.,Department of Physiology and Pathophysiology, University of Manitoba, 727 McDermot Avenue, Winnipeg, MB, Canada
| | - James D House
- Department of Animal Science, University of Manitoba, 12 Dafoe Road, Winnipeg, MB, Canada
| | - Karmin O
- Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Research Centre, 351 Tache Avenue, Winnipeg, MB, Canada.,Department of Physiology and Pathophysiology, University of Manitoba, 727 McDermot Avenue, Winnipeg, MB, Canada.,Department of Animal Science, University of Manitoba, 12 Dafoe Road, Winnipeg, MB, Canada
| |
Collapse
|
49
|
Sid V, Siow YL, Shang Y, Woo CW, O K. High-fat diet consumption reduces hepatic folate transporter expression via nuclear respiratory factor-1. J Mol Med (Berl) 2018; 96:1203-1213. [PMID: 30178194 DOI: 10.1007/s00109-018-1688-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 08/03/2018] [Accepted: 08/17/2018] [Indexed: 12/13/2022]
Abstract
Folate is an essential micronutrient for biological function. The liver, a primary organ for folate metabolism and storage, plays an important role in folate homeostasis. Proton-coupled folate transporter (PCFT) and reduced folate carrier (RFC) are the major folate transporters responsible for folate uptake at basolateral membrane of hepatocytes. Low serum folate levels are frequently associated with obesity. We investigated the mechanism that regulated folate status in a mouse model with diet-induced obesity. Mice (C57BL/6J) were fed a high-fat diet (60% kcal fat) for 8 weeks. Mice displayed increased hepatic lipid accumulation and decreased folate levels in the liver and serum compared to mice fed a normal chow diet (10% kcal fat). High-fat diet-fed mice had low expression of PCFT and RFC and decreased nuclear respiratory factor-1 (NRF-1)/DNA-binding activity. Treatment with NRF-1 siRNA or palmitic acid reduced folate transporter expression in hepatocytes. Inhibition of NRF-1 mediated folate transporter expression significantly reduced intracellular folate levels. These results suggest that chronic consumption of high-fat diets impairs folate transporter expression via NRF-1-dependent mechanism, leading to reduced hepatic folate storage. Understanding the regulation of folate homeostasis in obesity may have an important implication in current guideline of folate intake. KEY MESSAGES: Serum and liver folate levels are decreased in diet-induced obese mice. Chronic high-fat diet consumption impairs expression of hepatic PCFT and RFC. NRF-1 regulates hepatic folate transporters expression and folate levels.
Collapse
Affiliation(s)
- Victoria Sid
- St. Boniface Hospital Research Centre, Winnipeg, Canada
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Canada
| | - Yaw L Siow
- St. Boniface Hospital Research Centre, Winnipeg, Canada
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Canada
- Agriculture and Agri-Food Canada, Winnipeg, Canada
| | - Yue Shang
- St. Boniface Hospital Research Centre, Winnipeg, Canada
- Department of Animal Science, University of Manitoba, Winnipeg, Canada
| | - Connie W Woo
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, SAR, China.
| | - Karmin O
- St. Boniface Hospital Research Centre, Winnipeg, Canada.
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Canada.
- Department of Animal Science, University of Manitoba, Winnipeg, Canada.
- Laboratory of Integrative Biology, CCARM, St. Boniface Hospital Research Centre, 351 Tache Avenue, Winnipeg, Manitoba, R2H 2A6, Canada.
| |
Collapse
|
50
|
Lai Y, Zhou C, Huang P, Dong Z, Mo C, Xie L, Lin H, Zhou Z, Deng G, Liu Y, Chen Y, Huang S, Wu Z, Sun X, Gao L, Lv Z. Polydatin alleviated alcoholic liver injury in zebrafish larvae through ameliorating lipid metabolism and oxidative stress. J Pharmacol Sci 2018; 138:46-53. [DOI: 10.1016/j.jphs.2018.08.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2018] [Revised: 08/18/2018] [Accepted: 08/22/2018] [Indexed: 02/07/2023] Open
|