Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.257
Filter
1.
Cell Death Dis ; 15(6): 389, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38830896

ABSTRACT

Apolipoprotein O (APOO) plays a critical intracellular role in regulating lipid metabolism. Here, we investigated the roles of APOO in metabolism and atherogenesis in mice. Hepatic APOO expression was increased in response to hyperlipidemia but was inhibited after simvastatin treatment. Using a novel APOO global knockout (Apoo-/-) model, it was found that APOO depletion aggravated diet-induced obesity and elevated plasma cholesterol levels. Upon crossing with low-density lipoprotein receptor (LDLR) and apolipoprotein E (APOE) knockout hyperlipidemic mouse models, Apoo-/- Apoe-/- and Apoo-/- Ldlr-/- mice exhibited elevated plasma cholesterol levels, with more severe atherosclerotic lesions than littermate controls. This indicated the effects of APOO on cholesterol metabolism independent of LDLR and APOE. Moreover, APOO deficiency reduced cholesterol excretion through bile and feces while decreasing phospholipid unsaturation by inhibiting NRF2 and CYB5R3. Restoration of CYB5R3 expression in vivo by adeno-associated virus (AAV) injection reversed the reduced degree of phospholipid unsaturation while decreasing blood cholesterol levels. This represents the first in vivo experimental validation of the role of APOO in plasma cholesterol metabolism independent of LDLR and elucidates a previously unrecognized cholesterol metabolism pathway involving NRF2/CYB5R3. APOO may be a metabolic regulator of total-body cholesterol homeostasis and a target for atherosclerosis management. Apolipoprotein O (APOO) regulates plasma cholesterol levels and atherosclerosis through a pathway involving CYB5R3 that regulates biliary and fecal cholesterol excretion, independently of the LDL receptor. In addition, down-regulation of APOO may lead to impaired mitochondrial function, which in turn aggravates diet-induced obesity and fat accumulation.


Subject(s)
Cholesterol , NF-E2-Related Factor 2 , Receptors, LDL , Animals , Receptors, LDL/metabolism , Cholesterol/metabolism , NF-E2-Related Factor 2/metabolism , Mice , Mice, Knockout , Mice, Inbred C57BL , Lipid Metabolism , Male , Atherosclerosis/metabolism , Apolipoproteins/metabolism , Apolipoproteins/genetics , Humans , Liver/metabolism , Apolipoproteins E/metabolism , Hyperlipidemias/metabolism
2.
Fa Yi Xue Za Zhi ; 40(2): 172-178, 2024 Apr 25.
Article in English, Chinese | MEDLINE | ID: mdl-38847033

ABSTRACT

OBJECTIVES: To explore the biomarkers and potential mechanisms of chronic restraint stress-induced myocardial injury in hyperlipidemia ApoE-/- mice. METHODS: The hyperlipidemia combined with the chronic stress model was established by restraining the ApoE-/- mice. Proteomics and bioinformatics techniques were used to describe the characteristic molecular changes and related regulatory mechanisms of chronic stress-induced myocardial injury in hyperlipidemia mice and to explore potential diagnostic biomarkers. RESULTS: Proteomic analysis showed that there were 43 significantly up-regulated and 58 significantly down-regulated differentially expressed proteins in hyperlipidemia combined with the restraint stress group compared with the hyperlipidemia group. Among them, GBP2, TAOK3, TFR1 and UCP1 were biomarkers with great diagnostic potential. KEGG pathway enrichment analysis indicated that ferroptosis was a significant pathway that accelerated the myocardial injury in hyperlipidemia combined with restraint stress-induced model. The mmu_circ_0001567/miR-7a/Tfr-1 and mmu_circ_0001042/miR-7a/Tfr-1 might be important circRNA-miRNA-mRNA regulatory networks related to ferroptosis in this model. CONCLUSIONS: Chronic restraint stress may aggravate myocardial injury in hyperlipidemia mice via ferroptosis. Four potential biomarkers are selected for myocardial injury diagnosis, providing a new direction for sudden cardiac death (SCD) caused by hyperlipidemia combined with the restraint stress.


Subject(s)
Apolipoproteins E , Biomarkers , Disease Models, Animal , Hyperlipidemias , Restraint, Physical , Animals , Hyperlipidemias/metabolism , Hyperlipidemias/complications , Mice , Biomarkers/metabolism , Apolipoproteins E/genetics , Proteomics/methods , Stress, Psychological/complications , MicroRNAs/metabolism , MicroRNAs/genetics , Ferroptosis , Male , Myocardium/metabolism , Myocardium/pathology , Mice, Knockout , Uncoupling Protein 1/metabolism , Computational Biology
3.
Front Immunol ; 15: 1342350, 2024.
Article in English | MEDLINE | ID: mdl-38720901

ABSTRACT

Dyslipidemia is the most prevalent independent risk factor for patients with chronic kidney disease (CKD). Lipid-induced NLRP3 inflammasome activation in kidney-resident cells exacerbates renal injury by causing sterile inflammation. Nuclear factor erythroid 2-related factor 2 (Nrf2) is a transcription factor that modulates the cellular redox balance; however, the exact role of Nrf2 signaling and its regulation of the NLRP3 inflammasome in hyperlipidemia-induced kidney injury are poorly understood. In this study, we demonstrated that activation of the mtROS-NLRP3 inflammasome pathway is a critical contributor to renal tubular epithelial cell (RTEC) apoptosis under hyperlipidemia. In addition, the Nrf2/ARE signaling pathway is activated in renal tubular epithelial cells under hyperlipidemia conditions both in vivo and in vitro, and Nrf2 silencing accelerated palmitic acid (PA)-induced mtROS production, mitochondrial injury, and NLRP3 inflammasome activation. However, the activation of Nrf2 with tBHQ ameliorated mtROS production, mitochondrial injury, NLRP3 inflammasome activation, and cell apoptosis in PA-induced HK-2 cells and in the kidneys of HFD-induced obese rats. Furthermore, mechanistic studies showed that the potential mechanism of Nrf2-induced NLRP3 inflammasome inhibition involved reducing mtROS generation. Taken together, our results demonstrate that the Nrf2/ARE signaling pathway attenuates hyperlipidemia-induced renal injury through its antioxidative and anti-inflammatory effects through the downregulation of mtROS-mediated NLRP3 inflammasome activation.


Subject(s)
Epithelial Cells , Hyperlipidemias , Inflammasomes , Kidney Tubules , NF-E2-Related Factor 2 , NLR Family, Pyrin Domain-Containing 3 Protein , Signal Transduction , NF-E2-Related Factor 2/metabolism , Animals , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Inflammasomes/metabolism , Hyperlipidemias/metabolism , Hyperlipidemias/complications , Hyperlipidemias/immunology , Epithelial Cells/metabolism , Rats , Humans , Kidney Tubules/pathology , Kidney Tubules/metabolism , Male , Cell Line , Apoptosis , Antioxidant Response Elements , Mitochondria/metabolism , Disease Models, Animal , Rats, Sprague-Dawley
4.
J Med Chem ; 67(10): 7870-7890, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38739840

ABSTRACT

Activation of AMP-activated protein kinase (AMPK) is proposed to alleviate hyperlipidemia. With cordycepin and N6-(2-hydroxyethyl) adenosine (HEA) as lead compounds, a series of adenosine-based derivatives were designed, synthesized, and evaluated on activation of AMPK. Finally, compound V1 was identified as a potent AMPK activator with the lipid-lowering effect. Molecular docking and circular dichroism indicated that V1 exerted its activity by binding to the γ subunit of AMPK. V1 markedly decreased the serum low-density lipoprotein cholesterol levels in C57BL/6 mice, golden hamsters, and rhesus monkeys. V1 was selected as the clinical compound and concluded Phase 1 clinical trials. A single dose of V1 (2000 mg) increased AMPK activation in human erythrocytes after 5 and 12 h of treatment. RNA sequencing data suggested that V1 downregulated expression of genes involved in regulation of apoptotic process, lipid metabolism, endoplasmic reticulum stress, and inflammatory response in liver by activating AMPK.


Subject(s)
AMP-Activated Protein Kinases , Hyperlipidemias , Mice, Inbred C57BL , Animals , AMP-Activated Protein Kinases/metabolism , Hyperlipidemias/drug therapy , Hyperlipidemias/metabolism , Humans , Mice , Male , Macaca mulatta , Molecular Docking Simulation , Administration, Oral , Mesocricetus , Hypolipidemic Agents/pharmacology , Hypolipidemic Agents/chemistry , Hypolipidemic Agents/chemical synthesis , Hypolipidemic Agents/therapeutic use , Drug Discovery , Structure-Activity Relationship , Cricetinae
5.
Int J Biol Macromol ; 270(Pt 2): 132253, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38744359

ABSTRACT

Lipid metabolism plays an important role in energy homeostasis maintenance in response to stress. Nowadays, hyperlipidemia-related chronic diseases such as obesity, diabetes, atherosclerosis, and fatty liver pose significant health challenges. Dietary polysaccharides (DPs) have gained attention for their effective lipid-lowering properties. This review examines the multifaceted mechanisms that DPs employ to lower lipid levels in subjects with hyperlipidemia. DPs could directly inhibit lipid intake and absorption, promote lipid excretion, and regulate key enzymes involved in lipid metabolism pathways, including triglyceride and cholesterol anabolism and catabolism, fatty acid oxidation, and bile acid synthesis. Additionally, DPs indirectly improve lipid homeostasis by modulating gut microbiota composition and alleviating oxidative stress. Moreover, the lipid-lowering mechanisms of particular structural DPs (including ß-glucan, pectin, glucomannan, inulin, arabinoxylan, and fucoidan) are summarized. The relationship between the structure and lipid-lowering activity of DPs is also discussed based on current researches. Finally, potential breakthroughs and future directions in the development of DPs in lipid-lowering activity are discussed. The paper could provide a reference for further exploring the mechanism of DPs for lipid regulations and utilizing DPs as lipid-lowering dietary ingredients.


Subject(s)
Lipid Metabolism , Polysaccharides , Humans , Lipid Metabolism/drug effects , Polysaccharides/chemistry , Polysaccharides/pharmacology , Animals , Gastrointestinal Microbiome/drug effects , Hyperlipidemias/metabolism , Hyperlipidemias/drug therapy , Pectins/chemistry , Pectins/pharmacology
6.
Food Funct ; 15(11): 5955-5971, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38738998

ABSTRACT

The structural characteristics of fucoidans exhibit species and regional diversity. Previous studies have demonstrated that Laminaria japonica- and Ascophyllum nodosum-derived fucoidans have type I and type II fucosyl chains, respectively. These chemical differences may contribute to distinct hypolipidemic effects and mechanisms of action. Chemical analysis demonstrated that the percentage contents of sulfate, glucuronic acid, and galactose were higher in L. japonica-derived fucoidans than those of A. nodosum-derived fucoidans. In hyperlipidemic apolipoprotein E-deficient mice, both A. nodosum- and L. japonica-derived fucoidans significantly decreased the plasma and hepatic levels of total cholesterol and triglyceride, leading to the reduction of atherosclerotic plaques. Western blotting experiments demonstrated that these fucoidans significantly enhanced the expression and levels of scavenger receptor B type 1, cholesterol 7 alpha-hydroxylase A1, and peroxisome proliferator-activated receptor (PPAR)-α, contributing to circulating lipoprotein clearance and fatty acid degradation, respectively. Differentially, L. japonica-derived fucoidan significantly increased the LXR/ATP-binding cassette G8 signaling pathway in the small intestine, as revealed by real-time quantitative PCR, which may lead to further cholesterol and other lipid excretion. Collectively, these data are useful for understanding the hypolipidemic mechanisms of action of seaweed-derived fucoidans, and their potential application for the prevention and/or treatment of atherosclerotic cardiovascular diseases.


Subject(s)
Apolipoproteins E , Ascophyllum , Hypolipidemic Agents , Laminaria , Polysaccharides , Animals , Laminaria/chemistry , Ascophyllum/chemistry , Mice , Polysaccharides/pharmacology , Polysaccharides/chemistry , Hypolipidemic Agents/pharmacology , Apolipoproteins E/genetics , Male , Mice, Inbred C57BL , Triglycerides/blood , Triglycerides/metabolism , Cholesterol/blood , Cholesterol/metabolism , Mice, Knockout , PPAR alpha/metabolism , PPAR alpha/genetics , Hyperlipidemias/drug therapy , Hyperlipidemias/metabolism , Plant Extracts/pharmacology , Plant Extracts/chemistry , Liver/metabolism , Liver/drug effects , Humans , Edible Seaweeds
7.
Comput Biol Chem ; 110: 108088, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38685167

ABSTRACT

Cichorium glandulosum, a common traditional Chinese medicine used by Uyghur and Mongolian ethnic groups, is recognized for its potential to ameliorate metabolic disorders. However, the specific efficacy and mechanisms of Cichorium glandulosum in treating the comorbidity of hyperuricaemia and hyperlipidaemia remain unexplored. This study aims to explore the pharmacological effects and mechanisms of Cichorium glandulosum on this comorbidity through a combination of animal experiments, network pharmacology, and molecular docking techniques. A rat model of hyperuricaemia combined with hyperlipidaemia was established through a high-fat and high-purine diet, and the effective parts of the aqueous extract of Cichorium glandulosum to reduce uric acid and lipid levels were screened and the components of the parts were analysed by LC-MS/MS. The active components, core targets, and key pathways were analysed using network pharmacology and validated by molecular docking. Animal experimental results indicated that the n-butanol extract of Cichorium glandulosum showed a significant therapeutic effect on this comorbidity. Analysis of the n-butanol extract yielded 35 active ingredients and 138 intersecting targets related to diseases. Key targets identified through compound-target-pathway (C-T-P) and Protein-Protein Interaction (PPI) analyses included RELA, CASP3, PTGS2, TNF, and ESR1. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses revealed 2515 functional items and 164 pathways, respectively. Molecular docking demonstrated that isochlorogenic acid A, baicalin, chicoric acid, and lactucopicrin showed the highest binding affinity to RELA and PTGS2. The n-butanol fraction from the aqueous extract of Cichorium glandulosum was found to reduce uric acid and lipid levels effectively. In summary, Cichorium glandulosum has a therapeutic effect on hyperuricaemia combined with hyperlipidaemia through its multi-component, multi-target, and multi-pathway characteristics.


Subject(s)
Hyperlipidemias , Hyperuricemia , Molecular Docking Simulation , Network Pharmacology , Plant Extracts , Hyperuricemia/drug therapy , Hyperuricemia/metabolism , Animals , Hyperlipidemias/drug therapy , Hyperlipidemias/metabolism , Rats , Plant Extracts/chemistry , Plant Extracts/pharmacology , Male , Rats, Sprague-Dawley , Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use , Medicine, Chinese Traditional , Disease Models, Animal
8.
Am J Physiol Cell Physiol ; 326(6): C1563-C1572, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38586879

ABSTRACT

Atherosclerosis is an inflammatory disease of blood vessels involving the immune system. Natural killer T (NKT) cells, as crucial components of the innate and acquired immune systems, play critical roles in the development of atherosclerosis. However, the mechanism and clinical relevance of NKT cells in early atherosclerosis are largely unclear. The study investigated the mechanism influencing NKT cell function in apoE deficiency-induced early atherosclerosis. Our findings demonstrated that there were higher populations of NKT cells and interferon-gamma (IFN-γ)-producing NKT cells in the peripheral blood of patients with hyperlipidemia and in the aorta, blood, spleen, and bone marrow of early atherosclerotic mice compared with the control groups. Moreover, we discovered that the infiltration of CD80+ macrophages and CD1d expression on CD80+ macrophages in atherosclerotic mice climbed remarkably. CD1d expression increased in CD80+ macrophages stimulated by oxidized low-density lipoprotein (ox-LDL) ex vivo and in vitro. Ex vivo coculture of macrophages with NKT cells revealed that ox-LDL-induced CD80+ macrophages presented lipid antigen α-Galcer (alpha-galactosylceramide) to NKT cells via CD1d, enabling NKT cells to express more IFN-γ. Furthermore, a greater proportion of CD1d+ monocytes and CD1d+CD80+ monocytes were found in peripheral blood of hyperlipidemic patients compared with that of healthy donors. Positive correlations were found between CD1d+CD80+ monocytes and NKT cells or IFN-γ+ NKT cells in hyperlipidemic patients. Our findings illustrated that CD80+ macrophages stimulated NKT cells to secrete IFN-γ via CD1d-presenting α-Galcer, which may accelerate the progression of early atherosclerosis. Inhibiting lipid antigen presentation by CD80+ macrophages to NKT cells may be a promising immune target for the treatment of early atherosclerosis.NEW & NOTEWORTHY This work proposed the ox-LDL-CD80+ monocyte/macrophage-CD1d-NKT cell-IFN-γ axis in the progression of atherosclerosis. The proinflammatory IFN-γ+ NKT cells are closely related to CD1d+CD80+ monocytes in hyperlipidemic patients. Inhibiting CD80+ macrophages to present lipid antigens to NKT cells through CD1d blocking may be a new therapeutic target for atherosclerosis.


Subject(s)
Antigens, CD1d , Atherosclerosis , B7-1 Antigen , Hyperlipidemias , Lipoproteins, LDL , Macrophages , Natural Killer T-Cells , Animals , Humans , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , Antigens, CD1d/metabolism , Antigens, CD1d/immunology , Antigens, CD1d/genetics , Atherosclerosis/immunology , Atherosclerosis/metabolism , Atherosclerosis/pathology , Hyperlipidemias/immunology , Hyperlipidemias/metabolism , Lipoproteins, LDL/immunology , Lipoproteins, LDL/metabolism , Macrophages/immunology , Macrophages/metabolism , Male , Mice , B7-1 Antigen/metabolism , B7-1 Antigen/immunology , Interferon-gamma/metabolism , Interferon-gamma/immunology , Mice, Inbred C57BL , Female , Middle Aged
9.
Biomed Pharmacother ; 173: 116400, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38484560

ABSTRACT

Hyperlipidemia caused by abnormal lipid metabolism has reached epidemic proportions. This phenomenon is also common in companion animals. Previous studies showed that AEE significantly improves abnormal blood lipids in hyperlipidemia rats and mice, but its mechanism is still not clear enough. In this study, the mechanism and potential key pathways of AEE on improving hyperlipidemia in mice were investigated through the transcriptome and proteome study of ApoE-/- mice liver and the verification study on high-fat HepG2 cells. The results showed that AEE significantly decreased the serum TC and LDL-C levels of hyperlipidemia ApoE-/- mice, and significantly increased the enzyme activity of CYP7A1. After AEE intervention, the results of mice liver transcriptome and proteome showed that differential genes and proteins were enriched in lipid metabolism-related pathways. The results of RT-qPCR showed that AEE significantly regulated the expression of genes related to lipid metabolism in mice liver tissue. AEE significantly upregulated the protein expression of CYP7A1 in hyperlipidemia ApoE-/- mice liver tissue. The results in vitro showed that AEE significantly decreased the levels of TC and TG, and improved lipid deposition in high-fat HepG2 cells. AEE significantly increased the expression of CYP7A1 protein in high-fat HepG2 cells. AEE regulates the expression of genes related to lipid metabolism in high-fat HepG2 cells, mainly by FXR-SHP-CYP7A1 and FGF19-TFEB-CYP7A1 pathways. To sum up, AEE can significantly improve the hyperlipidemia status of ApoE-/- mice and the lipid deposition of high-fat HepG2 cells, and its main pathway is probably the bile acid metabolism-related pathway centered on CYP7A1.


Subject(s)
Hyperlipidemias , Mice , Rats , Animals , Hyperlipidemias/drug therapy , Hyperlipidemias/genetics , Hyperlipidemias/metabolism , Proteomics , Proteome/metabolism , Diet, High-Fat/adverse effects , Lipids , Lipid Metabolism/genetics , Gene Expression Profiling , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Liver/metabolism
10.
Aging (Albany NY) ; 16(5): 4095-4115, 2024 Mar 03.
Article in English | MEDLINE | ID: mdl-38441531

ABSTRACT

FoxO6, an identified factor, induces hyperlipidemia and hepatic steatosis during aging by activating hepatic lipoprotein secretion and lipogenesis leading to increased ApoC3 concentrations in the bloodstream. However, the intricate mechanisms underlying hepatic steatosis induced by elevated FoxO6 under hyperglycemic conditions remain intricate and require further elucidation. In order to delineate the regulatory pathway involving ApoC3 controlled by FoxO6 and its resultant functional impacts, we employed a spectrum of models including liver cell cultures, aged rats subjected to HFD, transgenic mice overexpressing FoxO6 (FoxO6-Tg), and FoxO6 knockout mice (FoxO6-KO). Our findings indicate that FoxO6 triggered ApoC3-driven lipid accumulation in the livers of aged rats on an HFD and in FoxO6-Tg, consequently leading to hepatic steatosis and hyperglycemia. Conversely, the absence of FoxO6 attenuated the expression of genes involved in lipogenesis, resulting in diminished hepatic lipid accumulation and mitigated hyperlipidemia in murine models. Additionally, the upregulation of FoxO6 due to elevated glucose levels led to increased ApoC3 expression, consequently instigating cellular triglyceride mediated lipid accumulation. The transcriptional activation of FoxO6 induced by both the HFD and high glucose levels resulted in hepatic steatosis by upregulating ApoC3 and genes associated with gluconeogenesis in aged rats and liver cell cultures. Our conclusions indicate that the upregulation of ApoC3 by FoxO6 promotes the development of hyperlipidemia, hyperglycemia, and hepatic steatosis in vivo, and in vitro. Taken together, our findings underscore the significance of FoxO6 in driving hyperlipidemia and hepatic steatosis specifically under hyperglycemic states by enhancing the expression of ApoC3 in aged rats.


Subject(s)
Fatty Liver , Hypercholesterolemia , Hyperglycemia , Hyperlipidemias , Animals , Mice , Rats , Diet, High-Fat/adverse effects , Fatty Liver/metabolism , Glucose/metabolism , Hyperglycemia/metabolism , Hyperlipidemias/metabolism , Liver/metabolism , Mice, Knockout , Mice, Transgenic , Transcription Factors/metabolism , Triglycerides/metabolism , Up-Regulation , Forkhead Transcription Factors/metabolism , Apolipoprotein C-III/metabolism
11.
Theranostics ; 14(5): 2036-2057, 2024.
Article in English | MEDLINE | ID: mdl-38505614

ABSTRACT

Background: ApoA5 mainly synthesized and secreted by liver is a key modulator of lipoprotein lipase (LPL) activity and triglyceride-rich lipoproteins (TRLs). Although the role of ApoA5 in extrahepatic triglyceride (TG) metabolism in circulation has been well documented, the relationship between ApoA5 and nonalcoholic fatty liver disease (NAFLD) remains incompletely understood and the underlying molecular mechanism still needs to be elucidated. Methods: We used CRISPR/Cas9 gene editing to delete Apoa5 gene from Syrian golden hamster, a small rodent model replicating human metabolic features. Then, the ApoA5-deficient (ApoA5-/-) hamsters were used to investigate NAFLD with or without challenging a high fat diet (HFD). Results: ApoA5-/- hamsters exhibited hypertriglyceridemia (HTG) with markedly elevated TG levels at 2300 mg/dL and hepatic steatosis on a regular chow diet, accompanied with an increase in the expression levels of genes regulating lipolysis and small adipocytes in the adipose tissue. An HFD challenge predisposed ApoA5-/- hamsters to severe HTG (sHTG) and nonalcoholic steatohepatitis (NASH). Mechanistic studies in vitro and in vivo revealed that targeting ApoA5 disrupted NR1D1 mRNA stability in the HepG2 cells and the liver to reduce both mRNA and protein levels of NR1D1, respectively. Overexpression of human NR1D1 by adeno-associated virus 8 (AAV8) in the livers of ApoA5-/- hamsters significantly ameliorated fatty liver without affecting plasma lipid levels. Moreover, restoration of hepatic ApoA5 or activation of UCP1 in brown adipose tissue (BAT) by cold exposure or CL316243 administration could significantly correct sHTG and hepatic steatosis in ApoA5-/- hamsters. Conclusions: Our data demonstrate that HTG caused by ApoA5 deficiency in hamsters is sufficient to elicit hepatic steatosis and HFD aggravates NAFLD by reducing hepatic NR1D1 mRNA and protein levels, which provides a mechanistic link between ApoA5 and NAFLD and suggests the new insights into the potential therapeutic approaches for the treatment of HTG and the related disorders due to ApoA5 deficiency in the clinical trials in future.


Subject(s)
Hyperlipidemias , Non-alcoholic Fatty Liver Disease , Animals , Cricetinae , Humans , Mice , Non-alcoholic Fatty Liver Disease/metabolism , Liver/metabolism , Triglycerides/metabolism , Hyperlipidemias/metabolism , Diet, High-Fat/adverse effects , Mesocricetus , RNA, Messenger/metabolism , Mice, Inbred C57BL , Nuclear Receptor Subfamily 1, Group D, Member 1/metabolism
12.
Int Immunopharmacol ; 132: 111856, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38537537

ABSTRACT

BACKGROUND AND AIMS: Inflammation and atherosclerosis (AS) are closely associated to Secreted Protein Acidic and Rich in Cysteine (SPARC) and its related factors. This study attempted to define the role and the potential mechanism of SPARC and its related factors in ameliorating hyperlipidemia and AS by aerobic exercise intervention. METHODS: The AS rat model was established with a high-fat diet plus vitamin D3 intraperitoneal injection. Treadmill exercises training (5 days/week at 14 m/min for 60 min/day) for 6 weeks was carried out for AS rat intervention method. Western blotting and qRT-PCR were used to analyze the mRNA and protein expression of SPARC and its related factors, respectively. H&E staining was applied to evaluate the morphological changes and inflammation damage. Von Kossa staining was used to measure the degree of vascular calcification. Fluorescence immunohistochemistry staining was used to detect the expression and distribution of SPARC signal molecules. RESULTS: SPARC was highly expressed and co-localization with the smooth muscle marker α-SMC in the AS rat. And its downstream factors, NF-κB, Caspase-1, IL-1ß and IL-18 were upregulated (P < 0.05 or P < 0.01), FNDC5 expression was downregulated in AS rat model. However, slight declined body weight, delayed AS progression, decreased hyperlipidemia and favorable morphology of skeletal muscle and blood vessels have been detected in AS rat with aerobic exercise intervention. Moreover, the expression of SPARC and its downstream factors were decreased (P < 0.05 or P < 0.01), while elevated the expression of FNDC5 (P < 0.01) was observed after aerobic exercise intervention. CONCLUSIONS: This study suggested that aerobic exercise ameliorated hyperlipidemia and AS by effectively inhibiting SPARC signal, and vascular smooth muscle cells may contribute greatly to the protection of AS.


Subject(s)
Atherosclerosis , Diet, High-Fat , Osteonectin , Physical Conditioning, Animal , Rats, Sprague-Dawley , Animals , Osteonectin/metabolism , Osteonectin/genetics , Atherosclerosis/therapy , Atherosclerosis/metabolism , Male , Rats , Signal Transduction , Disease Models, Animal , Hyperlipidemias/therapy , Hyperlipidemias/metabolism , Cholecalciferol/metabolism
13.
Cell Biol Int ; 48(6): 848-860, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38444077

ABSTRACT

Oxidized low-density lipoprotein (oxLDL), a key component in atherosclerosis and hyperlipidemia, is a risk factor for atherothrombosis in dyslipidemia, yet its mechanism is poorly understood. In this study, we used oxLDL-induced human aortic endothelial cells (HAECs) and high-fat diet (HFD)-fed mice as a hyperlipidemia model. Phosphatidylserine (PS) exposure, cytosolic Ca2+, reactive oxygen species (ROS), and lipid peroxidation were measured by flow cytometer. TMEM16F expression was detected by immunofluorescence, western blot, and reverse transcription polymerase chain reaction. Procoagulant activity (PCA) was measured by coagulation time, intrinsic/extrinsic factor Xase, and thrombin generation. We found that oxLDL-induced PS exposure and the corresponding PCA of HAECs were increased significantly compared with control, which could be inhibited over 90% by lactadherin. Importantly, TMEM16F expression in oxLDL-induced HAECs was upregulated by enhanced intracellular Ca2+ concentration, ROS, and lipid peroxidation, which led to PS exposure. Meanwhile, the knockdown of TMEM16F by short hairpin RNA significantly inhibited PS exposure in oxLDL-induced HAECs. Moreover, we observed that HFD-fed mice dramatically increased the progress of thrombus formation and accompanied upregulated TMEM16F expression by thromboelastography analysis, FeCl3-induced carotid artery thrombosis model, and western blot. Collectively, these results demonstrate that TMEM16F-mediated PS exposure may contribute to prothrombotic status under hyperlipidemic conditions, which may serve as a novel therapeutic target for the prevention of thrombosis in hyperlipidemia.


Subject(s)
Anoctamins , Endothelial Cells , Lipoproteins, LDL , Phosphatidylserines , Reactive Oxygen Species , Lipoproteins, LDL/pharmacology , Lipoproteins, LDL/metabolism , Animals , Humans , Phosphatidylserines/metabolism , Endothelial Cells/metabolism , Endothelial Cells/drug effects , Mice , Anoctamins/metabolism , Reactive Oxygen Species/metabolism , Mice, Inbred C57BL , Male , Hyperlipidemias/metabolism , Calcium/metabolism , Diet, High-Fat , Thrombosis/metabolism , Lipid Peroxidation/drug effects , Cells, Cultured , Blood Coagulation/drug effects
14.
J Ethnopharmacol ; 328: 118066, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38499259

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Gynostemma pentaphyllum (Thunb.) Makino has traditional applications in Chinese medicine to treat lipid abnormalities. Gypenosides (GPs), the main bioactive components of Gynostemma pentaphyllum, have been reported to exert hypolipidemic effects through multiple mechanisms. The lipid-lowering effects of GPs may be attributed to the aglycone portion resulting from hydrolysis of GPs by the gut microbiota. However, to date, there have been no reports on whether gypenoside aglycones (Agl), the primary bioactive constituents, can ameliorate hyperlipidemia by modulating the gut microbiota. AIM OF THE STUDY: This study explored the potential therapeutic effects of gypenoside aglycone (Agl) in a rat model of high-fat diet (HFD)-induced hyperlipidemia. METHODS: A hyperlipidemic rat model was established by feeding rats with a high-fat diet. Agl was administered orally, and serum lipid levels were analyzed. Molecular techniques, including RT-polymerase chain reaction (PCR) and fecal microbiota sequencing, were used to investigate the effects of Agl on lipid metabolism and gut microbiota composition. RESULTS: Agl administration significantly reduced serum levels of total cholesterol (TC), triglycerides (TG), and low-density lipoprotein cholesterol (LDL-C) and mitigated hepatic damage induced by HFD. Molecular investigations have revealed the modulation of key lipid metabolism genes and proteins by Agl. Notably, Agl treatment enriched the gut microbiota with beneficial genera, including Lactobacillus, Akkermansia, and Blautia and promoted specific shifts in Lactobacillus murinus, Firmicutes bacterium CAG:424, and Allobaculum stercoricanis. CONCLUSION: This comprehensive study established Agl as a promising candidate for the treatment of hyperlipidemia. It also exhibits remarkable hypolipidemic and hepatoprotective properties. The modulation of lipid metabolism-related genes, along with the restoration of gut microbiota balance, provides mechanistic insights. Thus, Agl has great potential for clinical applications in hyperlipidemia management.


Subject(s)
Gastrointestinal Microbiome , Hyperlipidemias , Rats , Animals , Diet, High-Fat/adverse effects , Gynostemma , Hyperlipidemias/drug therapy , Hyperlipidemias/metabolism , Triglycerides/metabolism , Lipid Metabolism , Cholesterol, LDL/metabolism , Plant Extracts
15.
Int J Mol Sci ; 25(5)2024 Mar 03.
Article in English | MEDLINE | ID: mdl-38474201

ABSTRACT

In recent years, the potent influence of tocotrienol (T3) on diminishing blood glucose and lipid concentrations in both Mus musculus (rats) and Homo sapiens (humans) has been established. However, the comprehensive exploration of tocotrienol's hypolipidemic impact and the corresponding mechanisms in aquatic species remains inadequate. In this study, we established a zebrafish model of a type 2 diabetes mellitus (T2DM) model through high-fat diet administration to zebrafish. In the T2DM zebrafish, the thickness of ocular vascular walls significantly increased compared to the control group, which was mitigated after treatment with T3. Additionally, our findings demonstrate the regulatory effect of T3 on lipid metabolism, leading to the reduced synthesis and storage of adipose tissue in zebrafish. We validated the expression patterns of genes relevant to these processes using RT-qPCR. In the T2DM model, there was an almost two-fold upregulation in pparγ and cyp7a1 mRNA levels, coupled with a significant downregulation in cpt1a mRNA (p < 0.01) compared to the control group. The ELISA revealed that the protein expression levels of Pparγ and Rxrα exhibited a two-fold elevation in the T2DM group relative to the control. In the T3-treated group, Pparγ and Rxrα protein expression levels consistently exhibited a two-fold decrease compared to the model group. Lipid metabolomics showed that T3 could affect the metabolic pathways of zebrafish lipid regulation, including lipid synthesis and decomposition. We provided experimental evidence that T3 could mitigate lipid accumulation in our zebrafish T2DM model. Elucidating the lipid-lowering effects of T3 could help to minimize the detrimental impacts of overfeeding in aquaculture.


Subject(s)
Diabetes Mellitus, Type 2 , Hyperlipidemias , Tocotrienols , Humans , Mice , Rats , Animals , Tocotrienols/metabolism , Zebrafish/metabolism , Diet, High-Fat , Hyperlipidemias/metabolism , Rice Bran Oil , Diabetes Mellitus, Type 2/metabolism , PPAR gamma/metabolism , RNA, Messenger/metabolism , Lipid Metabolism , Liver/metabolism
16.
Clin Sci (Lond) ; 138(4): 137-151, 2024 02 21.
Article in English | MEDLINE | ID: mdl-38299431

ABSTRACT

Hypercholesterolemia in pregnancy is a physiological process required for normal fetal development. In contrast, excessive pregnancy-specific hypercholesterolemia increases the risk of complications, such as preeclampsia. However, the underlying mechanisms are unclear. Toll-like receptor 4 (TLR4) is a membrane receptor modulated by high cholesterol levels, leading to endothelial dysfunction; but whether excessive hypercholesterolemia in pregnancy activates TLR4 is not known. We hypothesized that a high cholesterol diet (HCD) during pregnancy increases TLR4 activity in uterine arteries, leading to uterine artery dysfunction. Sprague Dawley rats were fed a control diet (n=12) or HCD (n=12) during pregnancy (gestational day 6-20). Vascular function was assessed in main uterine arteries using wire myography (vasodilation to methacholine and vasoconstriction to phenylephrine; with and without inhibitors for mechanistic pathways) and pressure myography (biomechanical properties). Exposure to a HCD during pregnancy increased maternal blood pressure, induced proteinuria, and reduced the fetal-to-placental weight ratio for both sexes. Excessive hypercholesterolemia in pregnancy also impaired vasodilation to methacholine in uterine arteries, whereby at higher doses, methacholine caused vasoconstriction instead of vasodilation in only the HCD group, which was prevented by inhibition of TLR4 or prostaglandin H synthase 1. Endothelial nitric oxide synthase expression and nitric oxide levels were reduced in HCD compared with control dams. Vasoconstriction to phenylephrine and biomechanical properties were similar between groups. In summary, excessive hypercholesterolemia in pregnancy impairs uterine artery function, with TLR4 activation as a key mechanism. Thus, TLR4 may be a target for therapy development to prevent adverse perinatal outcomes in complicated pregnancies.


Subject(s)
Hypercholesterolemia , Hyperlipidemias , Animals , Female , Male , Pregnancy , Rats , Hypercholesterolemia/metabolism , Hyperlipidemias/metabolism , Methacholine Chloride/metabolism , Phenylephrine/pharmacology , Phenylephrine/metabolism , Placenta , Rats, Sprague-Dawley , Toll-Like Receptor 4/metabolism , Uterine Artery/metabolism , Vasodilation/physiology
17.
Cardiovasc Ther ; 2024: 8649365, 2024.
Article in English | MEDLINE | ID: mdl-38375358

ABSTRACT

Cardiovascular disease (CVD) is the leading cause of morbidity and mortality worldwide, and hyperlipidemia is one major inducing factor of CVD. It is worthy to note that fucoidans are reported to have hypolipidemic activity with species specificity; however, the underlying mechanisms of action are far from clarification. This study is aimed at investigating the plasma lipid-lowering mechanisms of the fucoidan from L. japonica Aresch by detecting the levels of hepatic genes that are involved in lipid metabolism. Our results demonstrated that the fucoidan F3 significantly lowered total cholesterol and triglyceride in C57BL/6J mice fed a high-fat diet. In the mouse liver, fucoidan F3 intervention significantly increased the gene expression of peroxisome proliferator-activated receptor (PPAR) α, liver X receptor (LXR) α and ß, and ATP-binding cassette transporter (ABC) G1 and G8 and decreased the expression of proprotein convertase subtilisin/kexin type 9 (PCSK9), low-density lipoprotein receptor, cholesterol 7 alpha-hydroxylase A1, and sterol regulatory element-binding protein (SREBP) 1c and SREBP-2. These results demonstrated that the antihyperlipidemic effects of fucoidan F3 are related to its activation of PPARα and LXR/ABC signaling pathways and inactivation of SREBPs. In conclusion, fucoidan F3 may be explored as a potential compound for prevention or treatment of lipid disorders.


Subject(s)
Cardiovascular Diseases , Edible Seaweeds , Hyperlipidemias , Laminaria , Polysaccharides , Mice , Animals , Proprotein Convertase 9/metabolism , Hyperlipidemias/drug therapy , Hyperlipidemias/genetics , Hyperlipidemias/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , Sterol Regulatory Element Binding Protein 1/pharmacology , Mice, Inbred C57BL , Liver , Cholesterol/metabolism , Cholesterol/pharmacology , Cardiovascular Diseases/metabolism , Lipids
18.
J Ethnopharmacol ; 321: 117532, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38048892

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Poria cocos (Schw.) Wolf (Polyporaceae, P.cocos), which is born on the pine root, has a history of more than two thousand years of medicine in China. P.cocos was first recorded in the Shennong's Herbal Classic, studies have proved its lipid-lowering effect. AIM OF STUDY: The aim of study was to investigate the underlying mechanism of P.cocos extract on hyperlipidemia. MATERIALS AND METHODS: Male Sprague-Dawley (SD) rats aged 9-12 weeks were intraperitoneally (IP) injected with Triton-WR 1339 to establish an acute hyperlipidemia model. At 0 h and 20 h after the model was established, low and high doses of P.cocos extract or simvastatin were given twice. After 48 h, the rats were sacrificed, and liver and serum samples were collected for analysis. The cell model was constructed by treating L02 cells with 1% fat emulsion-10% FBS-RPMI 1640 medium for 48 h. At the same time, low and high doses of P.cocos extract and simvastatin were administered. Oil red O staining was used to evaluate the lipid accumulation in the cells, and H&E staining was used to evaluate the liver lesions of rats. Real-time quantitative PCR and western blotting were used to detect the expressions of lipid metabolism-related genes. RESULTS: P.cocos extract relieved lipid accumulation in vitro and alleviated hyperlipidemia in vivo. Both gene and protein expressions of peroxisome proliferator-activated receptor α (PPARα) were shown to be up-regulated by P.cocos extract. Additionally, P.cocos extract down-regulated the expressions of fatty acid synthesis-related genes sterol regulatory element-binding protein-1 (SREBP-1), Acetyl-CoA Carboxylase 1 (ACC1) and fatty acid synthase (FAS), while up-regulated the expressions of cholesterol metabolism-related genes liver X receptor-α (LXRα), ATP-binding cassette transporter A1 (ABCA1), cholesterol 7alpha-hydroxylase (CYP7A1) and low density lipoprotein receptor (LDLR), which were reversed by the treatment with the PPARα inhibitor GW6471. CONCLUSION: P.cocos extract ameliorates hyperlipidemia and lipid accumulation by regulating cholesterol homeostasis in hepatocytes through PPARα pathway. This study provides evidence that supplementation with P.cocos extract could be a potential strategy for the treatment of hyperlipidemia.


Subject(s)
Hyperlipidemias , Wolfiporia , Wolves , Rats , Male , Animals , PPAR alpha/genetics , PPAR alpha/metabolism , Wolves/metabolism , Rats, Sprague-Dawley , Liver , Lipid Metabolism , Hyperlipidemias/metabolism , Hepatocytes/metabolism , Lipids , Cholesterol/metabolism , Homeostasis , Simvastatin/pharmacology , Simvastatin/therapeutic use
19.
Biomed Pharmacother ; 170: 115960, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38039754

ABSTRACT

With the gradual improvement of individuals' living standards, there has been a concurrent escalation in the consumption of fats and sugars in the daily dietary habits. Consequently, an increasing number of individuals are afflicted by hyperlipidemia, a condition that, could elevate blood viscosity, thereby engendering serious complications in a long run. Traditional lipid-lowering medications, such as statins, manifest substantial side effects, thereby imposing a significant metabolic burden on the liver and kidneys. Conversely, antisense oligonucleotides (ASOs) exhibit attributes such as rapid absorption, prolonged efficacy, and minimal side effects. In light of these considerations, a novel ASO was meticulously designed, sebsequently, its efficacy and toxicity assessments were conducted both in vitro and in vivo. The results unequivocally demonstrate the effectiveness and safety of this ASO.


Subject(s)
Hyperlipidemias , Proprotein Convertase 9 , Humans , Proprotein Convertase 9/genetics , Proprotein Convertase 9/metabolism , Hyperlipidemias/drug therapy , Hyperlipidemias/metabolism , Oligonucleotides, Antisense/pharmacology , Oligonucleotides, Antisense/therapeutic use , Liver/metabolism
20.
J Sci Food Agric ; 104(4): 2417-2428, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-37989713

ABSTRACT

BACKGROUND: Hyperlipidemia is characterized by abnormally elevated blood lipids. Quinoa saponins (QS) have multiple pharmacological activities, including antitumor, bactericidal and immune-enhancing effects. However, the lipid-lowering effect and mechanisms of QS in vivo have been scarcely reported. METHODS: The effect of QS against hyperlipidemia induced by high-fat diet in rats was explored based on gut microbiota and serum non-targeted metabolomics. RESULTS: The study demonstrated that the supplementation of QS could reduce serum lipids, body weight, liver injury and inflammation. 16S rRNA sequencing demonstrated that QS mildly increased alpha-diversity, altered the overall structure of intestinal flora, decreased the relative richness of Firmicutes, the ratio of Firmicutes/Bacteroidetes (P < 0.05) and increased the relative richness of Actinobacteria, Bacteroidetes, Bifidobacterium, Roseburia and Coprococcus (P < 0.05). Simultaneously, metabolomics analysis showed that QS altered serum functional metabolites with respect to bile acid biosynthesis, arachidonic acid metabolism and taurine and hypotaurine metabolism, which were closely related to bile acid metabolism and fatty acid ß-oxidation. Furthermore, QS increased protein levels of farnesoid X receptor, peroxisome proliferator-activated receptor α and carnitine palmitoyltransferase 1, which were related to the screened metabolic pathways. Spearman correlation analysis showed that there was a correlation between gut microbiota and differential metabolites. CONCLUSION: QS could prevent lipid metabolism disorders in hyperlipidemic rats, which may be closely associated with the regulation of the gut microbiota and multiple metabolic pathways. This study may provide new evidence for QS as natural active substances for the prevention of hyperlipidemia. © 2023 Society of Chemical Industry.


Subject(s)
Chenopodium quinoa , Gastrointestinal Microbiome , Hyperlipidemias , Rats , Animals , Diet, High-Fat/adverse effects , Chenopodium quinoa/metabolism , Hyperlipidemias/drug therapy , Hyperlipidemias/etiology , Hyperlipidemias/metabolism , RNA, Ribosomal, 16S , Lipids/pharmacology , Metabolic Networks and Pathways , Bile Acids and Salts
SELECTION OF CITATIONS
SEARCH DETAIL
...