Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 3.293
Filtrer
1.
J Agric Food Chem ; 72(37): 20261-20272, 2024 Sep 18.
Article de Anglais | MEDLINE | ID: mdl-39241169

RÉSUMÉ

Pectin, a natural polysaccharide predominantly sourced from the cell walls of terrestrial plants, is widely regarded for its gelling, thickening, and stabilizing properties, which have extensive applications in the food, pharmaceutical, and biotechnological industries. This review discusses the mechanistic pathways by which pectin mediates its lipid-lowering properties, such as pectin's antioxidant activity, the modulation of gut microbiota, its anti-inflammatory properties, its capacity to bind bile acids and cholesterol, and its impact on the expression of genes associated with lipid metabolism. To enhance its hypolipidemic properties, chemical, physical, and enzymatic modification techniques are explored. Additionally, the synergistic effects of pectin in combination with other bioactive compounds such as phytosterols and polyphenols, as well as its potential in nanocarrier-mediated delivery systems for lipid-lowering agents, are highlighted. The review also conducts a critical analysis of the safety and regulatory considerations associated with pectin use, emphasizing the necessity for comprehensive toxicological evaluations and adherence to regulatory standards. This paper underscores the growing potential of pectin not only as a dietary fiber but also as a multifaceted agent for ameliorating hyperlipidemia, catalyzing a shift toward more targeted and efficacious lipid-lowering strategies.


Sujet(s)
Vecteurs de médicaments , Pectine , Pectine/composition chimique , Humains , Animaux , Vecteurs de médicaments/composition chimique , Hypolipémiants/composition chimique , Hypolipémiants/pharmacologie , Métabolisme lipidique/effets des médicaments et des substances chimiques , Hyperlipidémies/traitement médicamenteux , Hyperlipidémies/métabolisme , Nanoparticules/composition chimique
2.
J Agric Food Chem ; 72(39): 21548-21559, 2024 Oct 02.
Article de Anglais | MEDLINE | ID: mdl-39226078

RÉSUMÉ

The simultaneous enhancement of lipophagy and mitochondrial biogenesis has emerged as a promising strategy for lipid lowering. The transcription factor EB (TFEB) exhibits a dual role, whereby it facilitates the degradation of lipid droplets (LDs) through the process of lipophagy while simultaneously stimulating mitochondrial biogenesis to support the utilization of lipophagy products. The purpose of this study was to explore the effect of astragaloside I (AS I) on hyperlipidemia and elucidate its underlying mechanism. AS I improved serum total cholesterol and triglyceride levels and reduced hepatic steatosis and lipid accumulation in db/db mice. AS I enhanced the fluorescence colocalization of LDs and autophagosomes and promoted the proteins and genes related to the autolysosome. Moreover, AS I increased the expression of mitochondrial biogenesis-related proteins and genes, indicating that AS I promoted lipophagy and mitochondrial biogenesis. Mechanistically, AS I inhibits the protein level of p-TFEB (ser211) expression and promotes TFEB nuclear translocation. The activation of TFEB by AS I was impeded upon the introduction of the mammalian target of rapamycin (mTOR) agonist MHY1485. The inhibition of p-mTOR by AS I and the activation of TFEB were no longer observed after administration of the Akt agonist SC-79, which indicated that AS I activated TFEB to promote lipophagy-dependent on the Akt/mTOR pathway and may be a potentially effective pharmaceutical and food additive for the treatment of hyperlipidemia.


Sujet(s)
Autophagie , Facteurs de transcription à motifs basiques hélice-boucle-hélice et à glissière à leucines , Hyperlipidémies , Souris de lignée C57BL , Biogenèse des organelles , Protéines proto-oncogènes c-akt , Saponines , Sérine-thréonine kinases TOR , Animaux , Sérine-thréonine kinases TOR/métabolisme , Sérine-thréonine kinases TOR/génétique , Facteurs de transcription à motifs basiques hélice-boucle-hélice et à glissière à leucines/métabolisme , Facteurs de transcription à motifs basiques hélice-boucle-hélice et à glissière à leucines/génétique , Souris , Saponines/pharmacologie , Protéines proto-oncogènes c-akt/métabolisme , Protéines proto-oncogènes c-akt/génétique , Hyperlipidémies/traitement médicamenteux , Hyperlipidémies/métabolisme , Hyperlipidémies/génétique , Mâle , Humains , Autophagie/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Mitochondries/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Triglycéride/métabolisme , Triterpènes/pharmacologie
3.
Nature ; 634(8033): 457-465, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39231480

RÉSUMÉ

Hyperlipidaemia is a major risk factor of atherosclerotic cardiovascular disease (ASCVD). Risk of cardiovascular events depends on cumulative lifetime exposure to low-density lipoprotein cholesterol (LDL-C) and, independently, on the time course of exposure to LDL-C, with early exposure being associated with a higher risk1. Furthermore, LDL-C fluctuations are associated with ASCVD outcomes2-4. However, the precise mechanisms behind this increased ASCVD risk are not understood. Here we find that early intermittent feeding of mice on a high-cholesterol Western-type diet (WD) accelerates atherosclerosis compared with late continuous exposure to the WD, despite similar cumulative circulating LDL-C levels. We find that early intermittent hyperlipidaemia alters the number and homeostatic phenotype of resident-like arterial macrophages. Macrophage genes with altered expression are enriched for genes linked to human ASCVD in genome-wide association studies. We show that LYVE1+ resident macrophages are atheroprotective, and identify biological pathways related to actin filament organization, of which alteration accelerates atherosclerosis. Using the Young Finns Study, we show that exposure to cholesterol early in life is significantly associated with the incidence and size of carotid atherosclerotic plaques in mid-adulthood. In summary, our results identify early intermittent exposure to cholesterol as a strong determinant of accelerated atherosclerosis, highlighting the importance of optimal control of hyperlipidaemia early in life, and providing insights into the underlying biological mechanisms. This knowledge will be essential to designing effective therapeutic strategies to combat ASCVD.


Sujet(s)
Athérosclérose , Hyperlipidémies , Macrophages , Animaux , Macrophages/métabolisme , Souris , Athérosclérose/anatomopathologie , Athérosclérose/métabolisme , Mâle , Hyperlipidémies/métabolisme , Hyperlipidémies/anatomopathologie , Humains , Femelle , Cholestérol LDL/sang , Cholestérol LDL/métabolisme , Régime occidental/effets indésirables , Plaque d'athérosclérose/anatomopathologie , Plaque d'athérosclérose/métabolisme , Finlande , Souris de lignée C57BL , Phénotype , Étude d'association pangénomique , Adulte d'âge moyen
4.
Atherosclerosis ; 397: 118582, 2024 10.
Article de Anglais | MEDLINE | ID: mdl-39260002

RÉSUMÉ

BACKGROUND AND AIMS: Lysyl oxidase (LOX) catalyzes the crosslinking of collagen and elastin to maintain tensile strength and structural integrity of the vasculature. Excessive LOX activity increases vascular stiffness and the severity of occlusive diseases. Herein, we investigated the mechanisms by which LOX controls atherogenesis and osteogenic differentiation of vascular smooth muscle cells (SMC) in hyperlipidemic mice. METHODS: Gene inactivation of Lox in SMC was achieved in conditional knockout mice after tamoxifen injections. Atherosclerosis burden and vascular calcification were assessed in hyperlipidemic conditional [Loxf/fMyh11-CreERT2ApoE-/-] and sibling control mice [Loxwt/wtMyh11-CreERT2ApoE-/-]. Mechanistic studies were performed with primary aortic SMC from Lox mutant and wild type mice. RESULTS: Inactivation of Lox in SMCs decreased > 70 % its RNA expression and protein level in the aortic wall and significantly reduced LOX activity without compromising vascular structure and function. Moreover, LOX deficiency protected mice against atherosclerotic burden (13 ± 2 versus 23 ± 1 %, p < 0.01) and plaque calcification (5 ± 0.4 versus 11.8 ± 3 %, p < 0.05) compared to sibling controls. Interestingly, gene inactivation of Lox in SMCs preserved the contractile phenotype of vascular SMC under hyperlipidemic conditions as demonstrated by single-cell RNA sequencing and immunofluorescence. Mechanistically, the absence of LOX in SMC prevented excessive collagen crosslinking and the subsequent activation of the pro-osteogenic FAK/ß-catenin signaling axis. CONCLUSIONS: Lox inactivation in SMC protects mice against atherosclerosis and plaque calcification by reducing SMC modulation and FAK/ß-catenin signaling.


Sujet(s)
Athérosclérose , Modèles animaux de maladie humaine , Hyperlipidémies , Souris knockout , Muscles lisses vasculaires , Myocytes du muscle lisse , Plaque d'athérosclérose , Lysyloxidase , Calcification vasculaire , Animaux , Lysyloxidase/métabolisme , Lysyloxidase/génétique , Myocytes du muscle lisse/enzymologie , Myocytes du muscle lisse/anatomopathologie , Myocytes du muscle lisse/métabolisme , Athérosclérose/génétique , Athérosclérose/enzymologie , Athérosclérose/anatomopathologie , Athérosclérose/métabolisme , Muscles lisses vasculaires/anatomopathologie , Muscles lisses vasculaires/enzymologie , Muscles lisses vasculaires/métabolisme , Calcification vasculaire/génétique , Calcification vasculaire/anatomopathologie , Calcification vasculaire/enzymologie , Calcification vasculaire/prévention et contrôle , Calcification vasculaire/métabolisme , Hyperlipidémies/génétique , Hyperlipidémies/enzymologie , Hyperlipidémies/complications , Hyperlipidémies/métabolisme , Souris , Ostéogenèse , Cellules cultivées , Maladies de l'aorte/anatomopathologie , Maladies de l'aorte/génétique , Maladies de l'aorte/enzymologie , Maladies de l'aorte/prévention et contrôle , Maladies de l'aorte/métabolisme , Aorte/anatomopathologie , Aorte/enzymologie , Aorte/métabolisme , Mâle , Souris de lignée C57BL , bêta-Caténine/métabolisme , Transduction du signal , Protéines de la matrice extracellulaire
5.
Nat Cardiovasc Res ; 3(5): 594-611, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-39195940

RÉSUMÉ

Dysregulation of the hematopoietic niche during hyperlipidemia facilitates pathologic leukocyte production, driving atherogenesis. Although definitive hematopoiesis occurs primarily in the bone marrow, during atherosclerosis this also occurs in the spleen. Cells of the bone marrow niche, particularly endothelial cells, have been studied in atherosclerosis, although little is known about how splenic endothelial cells respond to the atherogenic environment. Here we show unique dysregulated pathways in splenic compared to bone marrow endothelial cells during atherosclerosis, including perturbations of lipid metabolism and endocytic trafficking pathways. As part of this response, we identify the mixed lineage kinase domain-like (MLKL) protein as a repressor of splenic, but not bone marrow, myelopoiesis. Silencing MLKL in splenic endothelial cells results in inefficient endosomal trafficking and lipid accumulation, ultimately promoting the production of myeloid cells that participate in plaque development. These studies identify endocytic trafficking by MLKL as a key mechanism of splenic endothelial cell maintenance, splenic hematopoiesis and, subsequently, atherosclerosis.


Sujet(s)
Athérosclérose , Cellules endothéliales , Hyperlipidémies , Protein kinases , Rate , Rate/anatomopathologie , Rate/métabolisme , Protein kinases/métabolisme , Protein kinases/génétique , Cellules endothéliales/métabolisme , Cellules endothéliales/anatomopathologie , Animaux , Athérosclérose/anatomopathologie , Athérosclérose/métabolisme , Hyperlipidémies/métabolisme , Hyperlipidémies/anatomopathologie , Souris de lignée C57BL , Modèles animaux de maladie humaine , Mâle , Myélopoïèse , Humains , Cellules cultivées , Métabolisme lipidique , Souris , Plaque d'athérosclérose/anatomopathologie , Plaque d'athérosclérose/métabolisme , Souris invalidées pour les gènes ApoE , Endocytose/physiologie , Endosomes/métabolisme , Niche de cellules souches/physiologie
6.
Alcohol Alcohol ; 59(5)2024 Jul 21.
Article de Anglais | MEDLINE | ID: mdl-39127890

RÉSUMÉ

AIMS: Widespread brain metabolite abnormalities in those with alcohol use disorder (AUD) were reported in numerous studies, but the effects of the pro-atherogenic conditions of hypertension, type 2 diabetes mellitus, hepatitis C seropositivity, and hyperlipidemia on metabolite levels were not considered. These conditions were associated with brain metabolite abnormalities in those without AUD. We predicted treatment-seeking individuals with AUD and pro-atherogenic conditions (Atherogenic+) demonstrate lower regional metabolite markers of neuronal viability [N-acetylaspartate (NAA)] and cell membrane turnover/synthesis [choline-containing compounds (Cho)], compared with those with AUD without pro-atherogenic conditions (Atherogenic-) and healthy controls (CON). METHODS: Atherogenic+ (n = 59) and Atherogenic- (n = 51) and CON (n = 49) completed a 1.5 T proton magnetic resonance spectroscopic imaging study. Groups were compared on NAA, Cho, total creatine, and myoinositol in cortical gray matter (GM), white matter (WM), and select subcortical regions. RESULTS: Atherogenic+ had lower frontal GM and temporal WM NAA than CON. Atherogenic+ showed lower parietal GM, frontal, parietal and occipital WM and lenticular nuclei NAA level than Atherogenic- and CON. Atherogenic- showed lower frontal GM and WM NAA than CON. Atherogenic+ had lower Cho level than CON in the frontal GM, parietal WM, and thalamus. Atherogenic+ showed lower frontal WM and cerebellar vermis Cho than Atherogenic- and CON. CONCLUSIONS: Findings suggest proatherogenic conditions in those with AUD were associated with increased compromise of neuronal integrity and cell membrane turnover/synthesis. The greater metabolite abnormalities observed in Atherogenic+ may relate to increased oxidative stress-related compromise of neuronal and glial cell structure and/or impaired arterial vasoreactivity/lumen viability.


Sujet(s)
Alcoolisme , Athérosclérose , Encéphale , Humains , Mâle , Femelle , Adulte d'âge moyen , Alcoolisme/métabolisme , Alcoolisme/anatomopathologie , Encéphale/métabolisme , Encéphale/imagerie diagnostique , Adulte , Athérosclérose/métabolisme , Acide aspartique/analogues et dérivés , Acide aspartique/métabolisme , Diabète de type 2/métabolisme , Choline/métabolisme , Hypertension artérielle/métabolisme , Hyperlipidémies/métabolisme , Inositol/métabolisme , Spectroscopie par résonance magnétique , Créatine/métabolisme
7.
Discov Med ; 36(187): 1721-1731, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39190387

RÉSUMÉ

BACKGROUND: Hyperlipidemia is one of the main causes of aggravated hepatic ischemia-reperfusion injury (IRI). Simvastatin (SIM), a lipid-lowering drug, has been shown to effectively alleviate IRI caused by hyperlipidemia. However, the regulatory mechanism by which SIM alleviates hyperlipidemia-induced hepatic IRI is still not clear. This study aims to explore the potential mechanisms of SIM in inhibiting hyperlipidemia-induced hepatic IRI, providing new therapeutic strategies for the alleviation of hepatic IRI. METHODS: An animal model of hyperlipidemia was induced by feeding mice a high-fat diet for 8 weeks. Subsequently, a hepatic IRI animal model of hyperlipidemia was established by occluding the hepatic artery and portal vein for one hour, followed by reperfusion for 6 or 12 h. Enzyme linked immunosorbent assay, Western blotting, hematoxylin-eosin (H&E) staining, immunohistochemistry, immunofluorescence, and Terminal-deoxynucleoitidyl Transferase Mediated Nick End Labeling assay, were used to evaluate liver injury, neutrophil extracellular traps (NETs) formation, and related molecular mechanisms. RESULTS: Hepatic IRI was accelerated by hyperlipidemia, which enhanced the expression of oxidized low-density lipoprotein (oxLDL) and Macrophage-1antigen (Mac-1), leading to the promotion of NETs formation and apoptosis of liver cells. The administration of simvastatin reduced the levels of oxLDL and Mac-1, decreased the formation of NETs, and alleviated hepatic IRI induced by hyperlipidemia. CONCLUSIONS: Simvastatin reduced hyperlipidemia-induced hepatic IRI by inhibiting the formation of NETs through the regulation of the oxLDL/Mac-1 pathway.


Sujet(s)
Alimentation riche en graisse , Pièges extracellulaires , Hyperlipidémies , Foie , Lésion d'ischémie-reperfusion , Simvastatine , Animaux , Simvastatine/pharmacologie , Simvastatine/usage thérapeutique , Lésion d'ischémie-reperfusion/prévention et contrôle , Lésion d'ischémie-reperfusion/anatomopathologie , Lésion d'ischémie-reperfusion/traitement médicamenteux , Lésion d'ischémie-reperfusion/métabolisme , Souris , Pièges extracellulaires/effets des médicaments et des substances chimiques , Pièges extracellulaires/métabolisme , Mâle , Alimentation riche en graisse/effets indésirables , Foie/anatomopathologie , Foie/effets des médicaments et des substances chimiques , Foie/métabolisme , Hyperlipidémies/traitement médicamenteux , Hyperlipidémies/métabolisme , Hyperlipidémies/anatomopathologie , Hyperlipidémies/complications , Souris de lignée C57BL , Modèles animaux de maladie humaine , Lipoprotéines LDL/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques
8.
Food Funct ; 15(18): 9210-9223, 2024 Sep 16.
Article de Anglais | MEDLINE | ID: mdl-39158509

RÉSUMÉ

Monascus has the ability to produce secondary metabolites, such as monacolin K (MK), known for its physiological functions, including lipid-lowering effects. Widely utilized in industries such as health food and medicine, MK is a significant compound derived from Monascus. Quinoa, recognized by the Food and Agriculture Organization of the United Nations as "the only plant food that can meet human basic nutritional needs by itself", possesses dual advantages of high nutritional value and medicinal food homology. This study employed animal experiments to investigate the hypolipidemic activity of Monascus-fermented quinoa (MFQ) and explored the molecular mechanism underlying the lipid-lowering effect of MFQ on hyperlipidemic mice through transcriptomic and metabolomic analyses. The results demonstrated that high-dose MFQ intervention (1600 mg kg-1 d-1) effectively decreased weight gain in hyperlipidemic mice without significant changes in cardiac index, renal index, or spleen index. Moreover, hepatic steatosis in mice was significantly improved. Serum levels of total cholesterol, triglycerides, and low-density lipoprotein cholesterol were markedly reduced, demonstrating that the lipid-lowering effect of MFQ was comparable to the drug control lovastatin. Conversely, both low-dose MFQ (400 mg kg-1 d-1) and unfermented quinoa exhibited no significant lipid-lowering effect. Integrated analysis of the transcriptome and metabolome suggested that MFQ may regulate amino acid levels in hyperlipidemic mice by influencing metabolic pathways such as phenylalanine, tyrosine, and tryptophan metabolism. This regulation alleviates hyperlipidemia induced by a high-fat diet, resulting in a significant reduction in blood lipid levels in mice.


Sujet(s)
Acides aminés , Chenopodium quinoa , Hyperlipidémies , Monascus , Animaux , Monascus/métabolisme , Hyperlipidémies/traitement médicamenteux , Hyperlipidémies/métabolisme , Souris , Chenopodium quinoa/composition chimique , Mâle , Acides aminés/métabolisme , Fermentation , Souris de lignée C57BL , Hypolipémiants/pharmacologie , Triglycéride/métabolisme , Triglycéride/sang , Aliments fermentés/microbiologie
9.
Food Chem ; 461: 140715, 2024 Dec 15.
Article de Anglais | MEDLINE | ID: mdl-39178542

RÉSUMÉ

Hyperlipidemia, an elevated level of cholesterol and/or triglycerides, has become a major public health problem worldwide. Although drugs intervention is effective in treating hyperlipidemia, most of them have adverse side effects. Peptides from natural plants with high anti-hyperlipidemic activity and a strong safety profile have emerged as promising candidates to prevent and ameliorate hyperlipidemia. This review summarizes the recent advances in plant-derived anti-hyperlipidemic peptides in terms of their sources, production, purification, identification, and activity evaluation. The focus is extended to their potential anti-hyperlipidemic mechanisms and structure-function relationships. Bioactive peptides derived from various plant sources, especially peptides containing hydrophobic and/or acidic amino acids, have shown remarkable effects in hyperlipidemic treatment. Their anti-hyperlipidemic effects are mediated by various mechanisms, including regulation of cholesterol metabolism and triglyceride metabolism, inhibition of inflammation-related metabolic syndrome, and modulation of the gut microbiota. Further evaluation of the stability, bioavailability, and clinical efficacy of these peptides is recommended.


Sujet(s)
Hyperlipidémies , Hypolipémiants , Peptides , Humains , Hypolipémiants/composition chimique , Hypolipémiants/pharmacologie , Hyperlipidémies/traitement médicamenteux , Hyperlipidémies/métabolisme , Peptides/composition chimique , Peptides/pharmacologie , Animaux , Relation structure-activité , Extraits de plantes/composition chimique , Extraits de plantes/pharmacologie , Cholestérol/métabolisme , Protéines végétales/composition chimique , Protéines végétales/pharmacologie , Plantes/composition chimique
10.
Exp Mol Med ; 56(8): 1869-1886, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39198543

RÉSUMÉ

Hyperlipidemia and hypertension might play a role in cardiac fibrosis, in which a heterogeneous population of fibroblasts seems important. However, it is unknown whether CD34+ progenitor cells are involved in the pathogenesis of heart fibrosis. This study aimed to explore the mechanism of CD34+ cell differentiation in cardiac fibrosis during hyperlipidemia. Through the analysis of transcriptomes from 50,870 single cells extracted from mouse hearts and 76,851 single cells from human hearts, we have effectively demonstrated the evolving cellular landscape throughout cardiac fibrosis. Disturbances in lipid metabolism can accelerate the development of fibrosis. Through the integration of bone marrow transplantation models and lineage tracing, our study showed that hyperlipidemia can expedite the differentiation of non-bone marrow-derived CD34+ cells into fibroblasts, particularly FABP4+ fibroblasts, in response to angiotensin II. Interestingly, the partial depletion of CD34+ cells led to a notable reduction in triglycerides in the heart, mitigated fibrosis, and improved cardiac function. Furthermore, immunostaining of human heart tissue revealed colocalization of CD34+ cells and fibroblasts. Mechanistically, our investigation of single-cell RNA sequencing data through pseudotime analysis combined with in vitro cellular studies revealed the crucial role of the PPARγ/Akt/Gsk3ß pathway in orchestrating the differentiation of CD34+ cells into FABP4+ fibroblasts. Through our study, we generated valuable insights into the cellular landscape of CD34+ cell-derived cells in the hypertrophic heart with hyperlipidemia, indicating that the differentiation of non-bone marrow-derived CD34+ cells into FABP4+ fibroblasts during this process accelerates lipid accumulation and promotes heart failure via the PPARγ/Akt/Gsk3ß pathway.


Sujet(s)
Antigènes CD34 , Différenciation cellulaire , Protéines de liaison aux acides gras , Fibroblastes , Fibrose , Métabolisme lipidique , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Humains , Souris , Animaux , Antigènes CD34/métabolisme , Protéines de liaison aux acides gras/métabolisme , Protéines de liaison aux acides gras/génétique , Myocarde/métabolisme , Myocarde/anatomopathologie , Protéines proto-oncogènes c-akt/métabolisme , Hyperlipidémies/métabolisme , Hyperlipidémies/anatomopathologie , Mâle , Transduction du signal , Récepteur PPAR gamma/métabolisme , Glycogen synthase kinase 3 beta/métabolisme , Modèles animaux de maladie humaine
11.
J Biochem Mol Toxicol ; 38(8): e23802, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39132808

RÉSUMÉ

Hyperlipidemic pancreatitis (HP) is an inflammatory injury of the pancreas triggered by elevated serum triglyceride (TG) levels. The mechanistic target of rapamycin (mTOR) signaling pathway plays a crucial role in regulating lipid homeostasis and inflammation. This study aimed to investigate whether the activity of mTOR complex 2 (mTORC2) affects the progression of HP and its underlying mechanisms. In vivo, a high-fat diet and retrograde administration of sodium taurocholate were employed to establish the HP models in rats, with pancreatic tissue pathology evaluated. The expression of Rictor and peroxisome proliferator-activator receptor (PPAR) was examined. The serum levels of TG, fatty acid metabolites, inflammatory and lipid metabolism-related factors were determined. In vitro, pancreatic acinar cells (PACs) were exposed to palmitic acid and cholecystokinin-8. PAC apoptosis, pyroptosis, and ferroptosis were assessed. In the HP models, rats and PACs exhibited upregulated Rictor and downregulated PPARα, and Rictor knockdown promoted PPARα expression. In vivo, Rictor knockdown decreased the serum levels of TG, α-amylase, total cholesterol, low-density lipoprotein cholesterol, lactate dehydrogenase, and inflammatory factors, while increasing high-density lipoprotein cholesterol levels. Rictor knockdown increased ACOX1 and CPT1α and decreased SREBP-1, CD36, SCD1, ACLY, and ACACA. Rictor knockdown reduced damage to pancreatic tissue structure. In vitro, Rictor knockdown inhibited PAC apoptosis, pyroptosis, and ferroptosis. Treatment with the PPARα antagonist GW6471 abolished the beneficial effects of Rictor knockdown. Rictor/mTORC2 deficiency reduces serum TG levels, maintains lipid homeostasis, and suppresses inflammation by inhibiting PPARα expression. Weakening mTORC2 activity holds promise as a novel therapeutic strategy for HP.


Sujet(s)
Hyperlipidémies , Métabolisme lipidique , Complexe-2 cible mécanistique de la rapamycine , Récepteur PPAR alpha , Pancréatite , Rat Sprague-Dawley , Animaux , Récepteur PPAR alpha/métabolisme , Récepteur PPAR alpha/génétique , Rats , Pancréatite/métabolisme , Pancréatite/anatomopathologie , Pancréatite/induit chimiquement , Pancréatite/génétique , Hyperlipidémies/métabolisme , Hyperlipidémies/génétique , Métabolisme lipidique/effets des médicaments et des substances chimiques , Mâle , Complexe-2 cible mécanistique de la rapamycine/métabolisme , Techniques de knock-down de gènes
12.
Fa Yi Xue Za Zhi ; 40(3): 227-236, 2024 Jun 25.
Article de Anglais, Chinois | MEDLINE | ID: mdl-39166303

RÉSUMÉ

OBJECTIVES: To screen biomarkers for forensic identification of acute myocardial infarction (AMI) by non-targeted metabolomic studies on changes of urine metabolites in rats with AMI. METHODS: The rat models of the sham surgery group, AMI group and hyperlipidemia + acute myocardial infarction (HAMI) group were established. Ultra-high performance liquid chromatography-mass spectrometry (UPLC-MS) was used to analyze the changes of urine metabolic spectrometry in AMI rats. Principal component analysis, partial least squares-discriminant analysis, and orthogonal partial least squares-discriminant analysis were used to screen differential metabolites. The MetaboAnalyst database was used to analyze the metabolic pathway enrichment and access the predictive ability of differential metabolites. RESULTS: A total of 40 and 61 differential metabolites associated with AMI and HAMI were screened, respectively. Among them, 22 metabolites were common in both rat models. These small metabolites were mainly concentrated in the niacin and nicotinamide metabolic pathways. Within the 95% confidence interval, the area under the curve (AUC) values of receiver operator characteristic curve for N8-acetylspermidine, 3-methylhistamine, and thymine were greater than 0.95. CONCLUSIONS: N8-acetylspermidine, 3-methylhistamine, and thymine can be used as potential biomarkers for AMI diagnosis, and abnormal metabolism in niacin and nicotinamide may be the main causes of AMI. This study can provide reference for the mechanism and causes of AMI identification.


Sujet(s)
Marqueurs biologiques , Modèles animaux de maladie humaine , Métabolomique , Infarctus du myocarde , Animaux , Infarctus du myocarde/métabolisme , Infarctus du myocarde/urine , Rats , Métabolomique/méthodes , Mâle , Marqueurs biologiques/urine , Marqueurs biologiques/métabolisme , Chromatographie en phase liquide à haute performance , Rat Sprague-Dawley , Analyse en composantes principales , Analyse discriminante , Spectrométrie de masse/méthodes , Acide nicotinique/métabolisme , Acide nicotinique/urine , Hyperlipidémies/métabolisme , Nicotinamide/urine , Nicotinamide/métabolisme , Nicotinamide/analogues et dérivés , Voies et réseaux métaboliques , Courbe ROC , Méthode des moindres carrés , Médecine légale/méthodes , Métabolome
13.
Tissue Cell ; 90: 102498, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39079452

RÉSUMÉ

Atherosclerosis (AS) is a common disease seriously detrimental to human health. AS is a chronic progressive disease related to inflammatory reactions. The present study aimed to characterize and evaluate the effects of adipose tissue stem cells (ADSCs) in high-fat diet-induced atherosclerosis in a rat model. The present study comprises thirty-six rats and they were divided into three groups: the control group, the high-fat diet (HFD) group; which received a high-fat diet, and the high-fat diet + stem cells (HFD+SC) group; which was fed with a high-fat diet along with the administration of intravenous ADSCs. Food was given to the animals for 20 weeks to establish dyslipidemia models. After 20 weeks, animals were sacrificed by cervical dislocation; blood was collected to measure total cholesterol (TC), triglycerides (TG), low-density lipoprotein (LDL), and high-density lipoprotein (HDL); aortae were collected to detect morphologic changes. Rats of the HFD group showed a significant increase in body weight (B.Wt), altered lipid profile increased expression of inducible nitric oxide synthase (iNOS), and decreased expression of endothelial nitric oxide synthase (eNOS). However, in HFD+SC there was a significant decrease in body weight gain and an improvement in lipid profile. Histopathological and ultrastructural variations observed in the aorta of the HFD group when treated with ADSCs showed preserved normal histological architecture and reduced atherosclerosis compared with the HFD group. This was evidenced by laboratory, histological, immunohistochemical, and morphometric studies. Thus, ADSCs reduced TC, TG, and LDL, reduced the expression of iNOS, and increased the expression of eNOS. The high-fat diet was likely to cause damage to the wall of blood vessels. Systemically transplanted ADSCs could home to the aorta, and further protect the aorta from HFD-induced damage.


Sujet(s)
Tissu adipeux , Athérosclérose , Hyperlipidémies , Transplantation de cellules souches mésenchymateuses , Cellules souches mésenchymateuses , Animaux , Mâle , Tissu adipeux/cytologie , Athérosclérose/anatomopathologie , Athérosclérose/thérapie , Rats , Hyperlipidémies/thérapie , Hyperlipidémies/anatomopathologie , Hyperlipidémies/métabolisme , Cellules souches mésenchymateuses/métabolisme , Alimentation riche en graisse/effets indésirables , Nitric oxide synthase type III/métabolisme , Aorte/anatomopathologie , Nitric oxide synthase type II/métabolisme
14.
J Orthop Surg Res ; 19(1): 393, 2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-38970109

RÉSUMÉ

BACKGROUND: To aim of this study is to assess the mechanism through which Desertliving Cistanche modulates the PI3K/AKT signaling pathway in the treatment of hyperlipidemic osteoporosis in ovariectomized rats. METHODS: We randomly assigned specific-pathogen-free (SPF) rats into five groups (n = 10 per group). The normal control group received a standard diet, while the model group, atorvastatin group, diethylstilbestrol group, and treatment group were fed a high-fat diet. Four weeks later, bilateral ovariectomies were conducted, followed by drug interventions. After six weeks of treatment, relevant indicators were compared and analyzed. RESULTS: Compared to the normal control group, rats in the model group exhibited blurred trabecular morphology, disorganized osteocytes, significantly elevated levels of bone-specific alkaline phosphatase (BALP), bone Gla-protein (BGP), total cholesterol (TC), tumor necrosis factor-α (TNF-α), and receptor activator of NF-κB ligand (RANKL). Also, the model group revealed significantly reduced levels of ultimate load, fracture load, estradiol (E2), bone mineral density (BMD), osteoprotegerin (OPG), and phosphoinositide 3-kinase (PI3K) and protein kinase B (Akt) in femoral tissue. The atorvastatin group presented with higher TC and TNF-α levels compared to the normal control group. Conversely, the treatment group demonstrated enhanced trabecular morphology, denser structure, smaller bone marrow cavities, and reduced BALP, BGP, TC, TNF-α, and RANKL levels. Furthermore, the treatment group exhibited higher levels of E2, BMD, OPG, and PI3K and Akt in bone tissue compared to the model group. The treatment group also had lower TC and TNF-α levels than the atorvastatin group. Biomechanical analysis indicated that after administration of Desertliving Cistanche, the treatment group had reduced body mass, increased ultimate and fracture load of the femur, denser bone structure, smaller bone marrow cavities, and altered periosteal arrangement compared to the model group. CONCLUSION: Our study revealed that Desertliving Cistanche demonstrated significant efficacy in preventing and treating postmenopausal hyperlipidemic osteoporosis in rats.


Sujet(s)
Cistanche , Hyperlipidémies , Ostéoporose , Ovariectomie , Phosphatidylinositol 3-kinases , Protéines proto-oncogènes c-akt , Transduction du signal , Animaux , Ovariectomie/effets indésirables , Femelle , Protéines proto-oncogènes c-akt/métabolisme , Transduction du signal/physiologie , Phosphatidylinositol 3-kinases/métabolisme , Hyperlipidémies/complications , Hyperlipidémies/métabolisme , Ostéoporose/étiologie , Ostéoporose/métabolisme , Rats , Rat Sprague-Dawley , Densité osseuse/effets des médicaments et des substances chimiques , Répartition aléatoire
15.
Biochem Biophys Res Commun ; 730: 150387, 2024 Oct 20.
Article de Anglais | MEDLINE | ID: mdl-39002201

RÉSUMÉ

Uvaol (UV), a pentacyclic triterpene found in olives and virgin olive oil, is known for its anti-inflammatory and antioxidant effects in various disease models. While olive oil is reported to reduce obesity and insulin resistance, the specific impact of UV on liver lipid metabolism and its molecular mechanisms are not fully understood. In this study, hepatic lipid accumulation was measured using oil red O staining, and protein expression levels in liver cells were assessed via Western blot analysis. Apoptosis was evaluated through cell viability and caspase 3 activity assays. UV treatment reduced lipid accumulation, fatty acid uptake, apoptosis, and ER stress in palmitate-treated liver cells. Additionally, UV enhanced fatty acid oxidation. Mechanistically, increased SIRT6 expression and autophagy were observed in UV-treated cells. SIRT6-targeted siRNA or 3-methyladenine blocked the effects of UV in hyperlipidemic cells. In conclusion, UV improves SIRT6/autophagy signaling, reducing lipid deposition and apoptosis in liver cells under high lipid conditions. This in vitro study provides strong evidence for potential therapeutic strategies for hepatic steatosis.


Sujet(s)
Apoptose , Stress du réticulum endoplasmique , Hépatocytes , Hyperlipidémies , Métabolisme lipidique , Transduction du signal , Sirtuines , Hépatocytes/métabolisme , Hépatocytes/effets des médicaments et des substances chimiques , Hépatocytes/effets des radiations , Métabolisme lipidique/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Stress du réticulum endoplasmique/effets des médicaments et des substances chimiques , Hyperlipidémies/métabolisme , Hyperlipidémies/traitement médicamenteux , Sirtuines/métabolisme , Sirtuines/génétique , Apoptose/effets des médicaments et des substances chimiques , Apoptose/effets des radiations , Autophagie/effets des médicaments et des substances chimiques , Protein Tyrosine Phosphatase, Non-Receptor Type 1/métabolisme , Protein Tyrosine Phosphatase, Non-Receptor Type 1/antagonistes et inhibiteurs , Protein Tyrosine Phosphatase, Non-Receptor Type 1/génétique , Humains , Animaux , Triterpènes pentacycliques/pharmacologie
16.
Front Immunol ; 15: 1371477, 2024.
Article de Anglais | MEDLINE | ID: mdl-39007149

RÉSUMÉ

Background: A high-fat diet (HFD) contributes to various metabolic disorders and obesity, which are major contributors to cardiovascular disease. As an essential regulator for heart homeostasis, cardiac resident macrophages may go awry and contribute to cardiac pathophysiology upon HFD. Thus, to better understand how HFD induced cardiac dysfunction, this study intends to explore the transcriptional and functional changes in cardiac resident macrophages of HFD mice. Methods: C57BL/6J female mice that were 6 weeks old were fed with HFD or normal chow diet (NCD) for 16 weeks. After an evaluation of cardiac functions by echocardiography, mouse hearts were harvested and cardiac resident CCR2- macrophages were sorted, followed by Smart sequencing. Bioinformatics analysis including GO, KEGG, and GSEA analyses were employed to elucidate transcriptional and functional changes. Results: Hyperlipidemia and obesity were observed easily upon HFD. The mouse hearts also displayed more severe fibrosis and diastolic dysfunction in HFD mice. Smart sequencing and functional analysis revealed metabolic dysfunctions, especially lipid-related genes and pathways. Besides this, antigen-presentation-related gene such as Ctsf and inflammation, particularly for NF-κB signaling and complement cascades, underwent drastic changes in cardiac resident macrophages. GO cellular compartment analysis was also performed and showed specific organelle enrichment trends of the involved genes. Conclusion: Dysregulated metabolism intertwines with inflammation in cardiac resident macrophages upon HFD feeding in mice, and further research on crosstalk among organelles could shed more light on potential mechanisms.


Sujet(s)
Alimentation riche en graisse , Macrophages , Souris de lignée C57BL , Myocarde , Animaux , Alimentation riche en graisse/effets indésirables , Souris , Macrophages/immunologie , Macrophages/métabolisme , Femelle , Myocarde/métabolisme , Myocarde/immunologie , Obésité/immunologie , Obésité/métabolisme , Hyperlipidémies/immunologie , Hyperlipidémies/métabolisme
17.
Int J Biol Macromol ; 276(Pt 1): 133771, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38992531

RÉSUMÉ

Sargassum fusiforme is a brown seaweed that grows abundantly along the rocky coastlines of Asian countries. The polysaccharides derived from Sargassum fusiforme (SFPS) have received much interest due to their various bioactivities, such as hypolipidemic, hypoglycemic, and antioxidant activities. In this study, we extracted and purified SFPS, and obtained the ultrasonic degradation product (SFPSUD). The lipid regulatory effects of SFPS and SFPSUD were investigated in a zebrafish model fed a high-fat diet. The results showed that SFPS significantly decreased the levels of total cholesterol (TC) and triglycerides (TG), and increased the activities of lipoprotein lipase (LPL) and hepatic lipase (HL). SFPSUD was more effective than the SFPS in reducing the TC and TG levels in zebrafish, as well as increasing the LPL and HL activities. Histopathological observations of zebrafish livers showed that SFPSUD significantly improved lipid metabolism disorder in the hepatocytes. The possible lipid-lowering mechanism in zebrafish associated with SFPS and SFPSUD may involve acceleration of the lipid metabolism rate by increasing the activities of LPL and HL. Thus, SFPSUD could be tested as a highly effective hypolipidemic drug. Our results suggest that SFPS and SFPSUD have potential uses as functional foods for the prevention and treatment of hyperlipidemia. Ultrasound can be effectively applied to degrade SFPS to improve its physicochemical properties and bioactivities.


Sujet(s)
Alimentation riche en graisse , Hypolipémiants , Métabolisme lipidique , Polyosides , Sargassum , Danio zébré , Animaux , Sargassum/composition chimique , Polyosides/pharmacologie , Polyosides/composition chimique , Hypolipémiants/pharmacologie , Hypolipémiants/composition chimique , Métabolisme lipidique/effets des médicaments et des substances chimiques , Foie/effets des médicaments et des substances chimiques , Foie/métabolisme , Foie/anatomopathologie , Lipoprotein lipase/métabolisme , Triglycéride/sang , Triglycéride/métabolisme , Hyperlipidémies/traitement médicamenteux , Hyperlipidémies/métabolisme , Cholestérol/sang , Cholestérol/métabolisme , Triacylglycerol lipase/métabolisme , Edible Seaweeds
18.
Food Chem ; 459: 140264, 2024 Nov 30.
Article de Anglais | MEDLINE | ID: mdl-39068825

RÉSUMÉ

Hyperlipidemia, a condition characterized by elevated levels of lipids in the blood, poses a significant risk factor for various health disorders, notably cardiovascular diseases. Phytochemical compounds are promising alternatives to the current lipid-lowering drugs, which cause many undesirable effects. Based on in vivo and clinical studies, combining phytochemicals with other phytochemicals, prebiotics, and probiotics and their encapsulation in nanoparticles is more safe and effective for managing hyperlipidemia than monotherapy. To this end, the results obtained and the mechanisms of action of these combinations were examined in detail in this review.


Sujet(s)
Hyperlipidémies , Hypolipémiants , Composés phytochimiques , Humains , Hypolipémiants/pharmacologie , Hypolipémiants/composition chimique , Composés phytochimiques/composition chimique , Composés phytochimiques/pharmacologie , Animaux , Hyperlipidémies/traitement médicamenteux , Hyperlipidémies/métabolisme , Probiotiques/pharmacologie , Prébiotiques/analyse , Nanoparticules/composition chimique
19.
Life Sci ; 352: 122891, 2024 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-38977060

RÉSUMÉ

There is a growing body of evidence suggesting that the composition of intestinal flora plays a significant role in regulating lipid metabolism. 2', 3', 5'-tri-O-acetyl-N6-(3-hydroxyphenyl) adenosine (IMMH007) is a new candidate compound for regulating blood cholesterol and other lipids. In this study, we conducted metagenomic and metabolomic analyses on samples from high-fat diet-fed (HFD) hamsters treated with IMMH007. Our findings revealed that IMM-H007 reversed the imbalance of gut microbiota caused by a high-fat diet. Additionally, it activated adiponectin receptor and pantothenate and CoA biosynthesis pathway-related genes, which are known to regulate lipid and glucose metabolism. Furthermore, IMM-H007 promotes cholesterol metabolism by reducing the abundance of genes and species associated with 7α-dehydroxylation and bile salt hydrolase (BSH). Metabolomics and pharmacological studies have shown that IMM-H007 effectively improved glucose and lipid metabolism disorders caused by HFD, reduced the aggregation of secondary bile acids (SBAs), significantly increased the content of hyodeoxycholic acid (HDCA), and also activated the expression of VDR in the small intestine. As a result, there was a reduction in the leakage of diamine oxidase (DAO) into the bloodstream in hamsters, accompanied by an upregulation of ZO-1 expression in the small intestine. The results suggested that IMM-H007 regulated glucose and lipid metabolism, promoted cholesterol metabolism through activating the expression of VDR, inhibiting inflammatory and improving the permeability of the intestinal barrier. Thus, our study provides new understanding of how IMM-H007 interacts with intestinal function, microbiota, and relevant targets, shedding light on its mechanism of action.


Sujet(s)
Adénosine , Alimentation riche en graisse , Microbiome gastro-intestinal , Hyperlipidémies , Métabolisme lipidique , Animaux , Alimentation riche en graisse/effets indésirables , Mâle , Cricetinae , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Métabolisme lipidique/effets des médicaments et des substances chimiques , Hyperlipidémies/traitement médicamenteux , Hyperlipidémies/métabolisme , Adénosine/métabolisme , Voies et réseaux métaboliques/effets des médicaments et des substances chimiques , Mesocricetus , Intestins/effets des médicaments et des substances chimiques , Intestins/microbiologie , Transcriptome/effets des médicaments et des substances chimiques
20.
Int J Mol Sci ; 25(13)2024 Jul 06.
Article de Anglais | MEDLINE | ID: mdl-39000541

RÉSUMÉ

Type 2 diabetes (T2D) is a chronic metabolic disorder characterized by hyperglycemia and dyslipidemia. The termite fungus comb is an integral component of nests of termites, which are a global pest. Termite fungus comb polysaccharides (TFCPs) have been identified to possess antioxidant, anti-aging, and immune-enhancing properties. However, their physicochemical characteristics and their role in fighting diabetes have not been previously reported. In the current study, TFCPs were isolated and structurally characterized. The yield of TFCPs was determined to be 2.76%, and it was found to be composed of a diverse array of polysaccharides with varying molecular weights. The hypoglycemic and hypolipidemic effects of TFCPs, as well as their potential mechanisms of action, were investigated in a T2D mouse model. The results demonstrated that oral administration of TFCPs could alleviate fasting blood glucose levels, insulin resistance, hyperlipidemia, and the dysfunction of pancreatic islets in T2D mice. In terms of mechanisms, the TFCPs enhanced hepatic glycogenesis and glycolysis while inhibiting gluconeogenesis. Additionally, the TFCPs suppressed hepatic de novo lipogenesis and promoted fatty acid oxidation. Furthermore, the TFCPs altered the composition of the gut microbiota in the T2D mice, increasing the abundance of beneficial bacteria such as Allobaculum and Faecalibaculum, while reducing the levels of pathogens like Mailhella and Acetatifactor. Overall, these findings suggest that TFCPs may exert anti-diabetic effects by regulating hepatic glucose and lipid metabolism and the composition of the gut microbiota. These findings suggest that TFCPs can be used as a promising functional ingredient for the prevention and treatment of T2D.


Sujet(s)
Diabète de type 2 , Microbiome gastro-intestinal , Hyperglycémie , Hyperlipidémies , Métabolisme lipidique , Foie , Animaux , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Diabète de type 2/métabolisme , Diabète de type 2/traitement médicamenteux , Souris , Hyperlipidémies/traitement médicamenteux , Hyperlipidémies/métabolisme , Métabolisme lipidique/effets des médicaments et des substances chimiques , Hyperglycémie/traitement médicamenteux , Hyperglycémie/métabolisme , Foie/métabolisme , Foie/effets des médicaments et des substances chimiques , Polysaccharides fongiques/pharmacologie , Mâle , Diabète expérimental/traitement médicamenteux , Diabète expérimental/métabolisme , Glucose/métabolisme , Hypoglycémiants/pharmacologie , Hypoglycémiants/usage thérapeutique , Termitomyces/métabolisme , Glycémie/métabolisme , Polyosides/pharmacologie , Souris de lignée C57BL
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE