Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters











Database
Language
Publication year range
1.
Nat Commun ; 14(1): 8151, 2023 Dec 09.
Article in English | MEDLINE | ID: mdl-38071367

ABSTRACT

Aging is a major risk factor for metabolic disorders. Polyunsaturated fatty acid-derived bioactive lipids play critical roles as signaling molecules in metabolic processes. Nonetheless, their effects on age-related liver steatosis remain unknown. Here we show that senescent liver cells induce liver steatosis in a paracrine manner. Linoleic acid-derived 9-hydroxy-octadecadienoic acid (9-HODE) and 13-HODE increase in middle-aged (12-month-old) and aged (20-month-old) male mouse livers and conditioned medium from senescent hepatocytes and macrophages. Arachidonate 15-lipoxygenase, an enzyme for 13-HODE and 9-HODE production, is upregulated in senescent cells. A 9-HODE and 13-HODE mixture induces liver steatosis and activates SREBP1. Furthermore, catalase (CAT) is a direct target of 13-HODE, and its activity is decreased by 13-HODE. CAT overexpression reduces 13-HODE-induced liver steatosis and protects male mice against age-related liver steatosis. Therefore, 13-HODE produced by senescent hepatocytes and macrophages activates SREBP1 by directly inhibiting CAT activity and promotes liver steatosis.


Subject(s)
Fatty Liver , Linoleic Acids , Male , Mice , Animals , Catalase , Linoleic Acids/metabolism , Linoleic Acid , Liver/metabolism
2.
Cardiovasc Drugs Ther ; 37(3): 571-584, 2023 06.
Article in English | MEDLINE | ID: mdl-35796905

ABSTRACT

Cardiovascular diseases (CVDs) are the leading cause of death globally. Atherosclerosis is the basis of major CVDs - myocardial ischemia, heart failure, and stroke. Among numerous functional molecules, the transcription factor nuclear factor κB (NF-κB) has been linked to downstream target genes involved in atherosclerosis. The activation of the NF-κB family and its downstream target genes in response to environmental and cellular stress, hypoxia, and ischemia initiate different pathological events such as innate and adaptive immunity, and cell survival, differentiation, and proliferation. Thus, NF-κB is a potential therapeutic target in the treatment of atherosclerosis and related CVDs. Several biologics and small molecules as well as peptide/proteins have been shown to regulate NF-κB dependent signaling pathways. In this review, we will focus on the function of NF-κB in CVDs and the role of NF-κB inhibitors in the treatment of CVDs.


Subject(s)
Atherosclerosis , Cardiovascular Diseases , Heart Failure , Humans , NF-kappa B/metabolism , Signal Transduction/physiology
3.
Biochim Biophys Acta Mol Basis Dis ; 1868(8): 166413, 2022 08 01.
Article in English | MEDLINE | ID: mdl-35413401

ABSTRACT

BACKGROUND: The mechanisms underlying the progression of liver disease from simple hepatic steatosis to advanced nonalcoholic steatohepatitis (NASH) and liver fibrosis warrant further investigation. Increased mRNA levels of Annexin A2 protein (Anxa2) have been observed in patients with NASH. However, the role of Anxa2 in NASH remains unclear. METHODS: The protein levels of Anxa2 were analyzed in the livers of mice and patients with NASH. Anxa2-knockout and -knockdown mice were generated, and NASH was induced through a high fructose, palmitate, and cholesterol (FPC) diet or methionine- and choline-deficient (MCD) diet. FINDINGS: We found elevated expression of Anxa2 in the livers of patients and mice with NASH. Anxa2 knockdown but not knockout ameliorated liver fibrosis in both FPC and MCD diet-fed mice. Liver-specific Anxa2 overexpression increased collagen deposition in mice fed a normal diet. Mechanistically, Anxa2 overexpression in hepatocytes promoted hepatic stellate cell activation in a paracrine manner by increasing osteopontin expression. Notch inhibition suppressed the exogenous overexpression of Anxa2-induced osteopontin and endogenous Anxa2 expression. Additionally, Anxa2 overexpression accelerated the progression of nonalcoholic fatty liver disease (NAFLD) in mice fed a high-fat diet. Moreover, Anxa2 levels were higher in NAFLD patients with advanced liver fibrosis than in those with mild liver fibrosis, as determined using the Gene Expression Omnibus database. INTERPRETATION: In conclusion, we found increased Anxa2 expression in hepatocytes promoted liver fibrosis in NASH mice by increasing osteopontin expression. The Anxa2-Notch positive regulatory loop contributes to this process and represents a novel target for the treatment of NASH-related liver fibrosis.


Subject(s)
Annexin A2 , Non-alcoholic Fatty Liver Disease , Osteopontin , Animals , Annexin A2/genetics , Annexin A2/metabolism , Hepatocytes/metabolism , Humans , Liver Cirrhosis/pathology , Methionine/metabolism , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/pathology , Osteopontin/genetics , Osteopontin/metabolism
4.
Sci China Life Sci ; 64(3): 404-418, 2021 Mar.
Article in English | MEDLINE | ID: mdl-32804340

ABSTRACT

Targeting the white-to-brown fat conversion is important for developing potential strategies to counteract metabolic diseases; yet the mechanisms are not fully understood. Yes-associated-protein (YAP), a transcription co-activator, was demonstrated to regulate adipose tissue functions; however, its effects on browning of subcutaneous white adipose tissue (sWAT) are unclear. We demonstrated that YAP was highly expressed in cold-induced beige fat. Mechanistically, YAP was found as a target gene of miR-429, which downregulated YAP expression in vivo and in vitro. In addition, miR-429 level was decreased in cold-induced beige fat. Additionally, pharmacological inhibition of the interaction between YAP and transcriptional enhanced associate domains by verteporfin dampened the browning of sWAT. Although adipose tissue-specific YAP overexpression increased energy expenditure with increased basal uncoupling protein 1 expression, it had no additional effects on the browning of sWAT in young mice. However, we found age-related impairment of sWAT browning along with decreased YAP expression. Under these circumstances, YAP overexpression significantly improved the impaired WAT browning in middle-aged mice. In conclusion, YAP as a regulator of sWAT browning, was upregulated by lowering miR-429 level in cold-induced beige fat. Targeting the miR-429-YAP pathway could be exploited for therapeutic strategies for age-related impairment of sWAT browning.


Subject(s)
Adipose Tissue, Beige/metabolism , Adipose Tissue, White/metabolism , Cold Temperature , MicroRNAs/metabolism , YAP-Signaling Proteins/metabolism , 3T3-L1 Cells , Adipose Tissue, White/drug effects , Aging/metabolism , Animals , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Protein Domains , Transcription, Genetic , Uncoupling Protein 1/metabolism , YAP-Signaling Proteins/antagonists & inhibitors
5.
Am J Physiol Gastrointest Liver Physiol ; 316(4): G527-G538, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30789748

ABSTRACT

Hepatic steatosis is the beginning phase of nonalcoholic fatty liver disease, and hyperhomocysteinemia (HHcy) is a significant risk factor. Soluble epoxide hydrolase (sEH) hydrolyzes epoxyeicosatrienoic acids (EETs) and other epoxy fatty acids, attenuating their cardiovascular protective effects. However, the involvement of sEH in HHcy-induced hepatic steatosis is unknown. The current study aimed to explore the role of sEH in HHcy-induced lipid disorder. We fed 6-wk-old male mice a chow diet or 2% (wt/wt) high-metnionine diet for 8 wk to establish the HHcy model. A high level of homocysteine induced lipid accumulation in vivo and in vitro, which was concomitant with the increased activity and expression of sEH. Treatment with a highly selective specific sEH inhibitor (0.8 mg·kg-1·day-1 for the animal model and 1 µM for cells) prevented HHcy-induced lipid accumulation in vivo and in vitro. Inhibition of sEH activated the peroxisome proliferator-activated receptor-α (PPAR-α), as evidenced by elevated ß-oxidation of fatty acids and the expression of PPAR-α target genes in HHcy-induced hepatic steatosis. In primary cultured hepatocytes, the effect of sEH inhibition on PPAR-α activation was further confirmed by a marked increase in PPAR-response element luciferase activity, which was reversed by knock down of PPAR-α. Of note, 11,12-EET ligand dependently activated PPAR-α. Thus increased sEH activity is a key determinant in the pathogenesis of HHcy-induced hepatic steatosis, and sEH inhibition could be an effective treatment for HHcy-induced hepatic steatosis. NEW & NOTEWORTHY In the current study, we demonstrated that upregulation of soluble epoxide hydrolase (sEH) is involved in the hyperhomocysteinemia (HHcy)-caused hepatic steatosis in an HHcy mouse model and in murine primary hepatocytes. Improving hepatic steatosis in HHcy mice by pharmacological inhibition of sEH to activate peroxisome proliferator-activated receptor-α was ligand dependent, and sEH could be a potential therapeutic target for the treatment of nonalcoholic fatty liver disease.


Subject(s)
Enzyme Inhibitors/pharmacokinetics , Epoxide Hydrolases , Fatty Acids/metabolism , Fatty Liver , Hyperhomocysteinemia , PPAR alpha/metabolism , Animals , Disease Models, Animal , Drug Discovery , Epoxide Hydrolases/antagonists & inhibitors , Epoxide Hydrolases/metabolism , Fatty Liver/drug therapy , Fatty Liver/enzymology , Fatty Liver/etiology , Fatty Liver/metabolism , Hyperhomocysteinemia/complications , Hyperhomocysteinemia/metabolism , Ligands , Mice , Mice, Inbred C57BL , Up-Regulation
6.
Cell Mol Gastroenterol Hepatol ; 7(1): 211-231, 2019.
Article in English | MEDLINE | ID: mdl-30539788

ABSTRACT

BACKGROUND & AIMS: Nonalcoholic steatohepatitis (NASH) is an increasingly prevalent nonalcoholic fatty liver disease, characterized by inflammatory cell infiltration and hepatocellular damage. Mammalian target of rapamycin complex 1 (mTORC1) has been investigated extensively in the context of cancer, including hepatocellular carcinoma. However, the role of mTORC1 in NASH remains largely unknown. METHODS: mTORC1 activity in macrophages in human mild and severe NASH liver was compared. Mice with macrophage-specific deletion of the regulatory-associated protein of mTOR (Raptor) subunit and littermate controls were fed a high-fructose, palmitate, and cholesterol diet for 24 weeks or a methionine- and choline-deficient diet for 4 weeks to develop NASH. RESULTS: We report that in human beings bearing NASH, macrophage mTORC1 activity was lower in livers experiencing severe vs mild NASH liver. Moreover, macrophage mTORC1 disruption exacerbated the inflammatory response in 2 diet-induced NASH mouse models. Mechanistically, in response to apoptotic hepatocytes (AHs), macrophage polarization toward a M2 anti-inflammatory phenotype was inhibited in Raptor-deficient macrophages. During the digestion of AHs, macrophage mTORC1 was activated and coupled with dynamin-related protein 1 to facilitate the latter's phosphorylation, leading to mitochondrial fission-mediated calcium release. Ionomycin or A23187, calcium ionophores, prevented Raptor deficiency-mediated failure of lysosome acidification and subsequent lipolysis. Blocking dynamin-related protein 1-dependent mitochondria fission impaired lysosome function, resulting in reduced production of anti-inflammatory factors such as interleukins 10 and 13. CONCLUSIONS: Persistent mTORC1 deficiency in macrophages contributes to the progression of NASH by causing lysosome dysfunction and subsequently attenuating anti-inflammatory M2-like response in macrophages during clearance of AHs.


Subject(s)
Disease Progression , Lysosomes/pathology , Macrophages, Peritoneal/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Regulatory-Associated Protein of mTOR/deficiency , Animals , Apoptosis , Calcium/metabolism , Cholesterol , Choline , Diet , Dynamins/metabolism , Feeding Behavior , Fructose , GTP Phosphohydrolases/metabolism , Gene Deletion , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Inflammation/pathology , Lipolysis , Liver/metabolism , Liver/pathology , Lysosomes/metabolism , Macrophages, Peritoneal/pathology , Male , Mechanistic Target of Rapamycin Complex 1/metabolism , Methionine/deficiency , Mice, Inbred C57BL , Mice, Knockout , Palmitates , Phagosomes/metabolism , Phosphorylation , Phosphoserine/metabolism , Regulatory-Associated Protein of mTOR/metabolism
7.
Br J Pharmacol ; 175(8): 1190-1204, 2018 04.
Article in English | MEDLINE | ID: mdl-28771708

ABSTRACT

BACKGROUND AND PURPOSE: Atherosclerosis results from a maladaptive inflammatory response initiated by the intramural retention of LDL in susceptible areas of the arterial vasculature. The ω-3 polyunsaturated fatty acids (ω-3) have protective effects in atherosclerosis; however, their molecular mechanism is still largely unknown. The present study used a metabolomic approach to reveal the atheroprotective metabolites of ω-3 and investigate the underlying mechanisms. EXPERIMENTAL APPROACH: We evaluated the development of atherosclerosis in LDL receptor-deficient mice (LDLR-/- ) fed a Western-type diet (WTD) plus ω-3 and also LDLR-/- and fat-1 transgenic (LDLR-/- -fat-1tg ) mice fed a WTD. The profiles of ω-3 in the plasma were screened by LC-MS/MS using unbiased systematic metabolomics analysis. We also studied the effect of metabolites of eicosapentaenoic acid (EPA) on endothelial activation in vitro. KEY RESULTS: The ω-3 diet and fat-1 transgene decreased monocyte infiltration, inhibited the expression of pro-inflammatory genes and significantly attenuated atherosclerotic plaque formation and enhanced plaque stability in LDLR-/- mice. The content of 18-hydroxy-eicosapentaenoic acid (18-HEPE) and 17,18-epoxy-eicosatetraenoic acid (17,18-EEQ), from the cytochrome P450 pathway of EPA, was significantly higher in plasma from both ω-3-treated LDLR-/- and LDLR-/- -fat-1tg mice as compared with WTD-fed LDLR-/- mice. In vitro in endothelial cells, 18-HEPE or 17,18-EEQ decreased inflammatory gene expression induced by TNFα via NF-κB signalling and thereby inhibited monocyte adhesion to endothelial cells. CONCLUSIONS AND IMPLICATIONS: EPA protected against the development of atherosclerosis in atheroprone mice via the metabolites 18-HEPE and/or 17,18-EEQ, which reduced endothelial activation. These compounds may have therapeutic implications in atherosclerosis. LINKED ARTICLES: This article is part of a themed section on Spotlight on Small Molecules in Cardiovascular Diseases. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.8/issuetoc.


Subject(s)
Atherosclerosis/drug therapy , Fatty Acids, Omega-3/therapeutic use , Animals , Aorta/drug effects , Aorta/metabolism , Atherosclerosis/metabolism , Cadherins/genetics , Cells, Cultured , Cholesterol/blood , Diet, Western , Fatty Acids, Omega-3/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/physiology , Humans , Male , Metabolomics , Mice, Transgenic , Monocytes/drug effects , Monocytes/physiology , Receptors, LDL/genetics , Triglycerides/blood , Tumor Necrosis Factor-alpha
8.
Br J Pharmacol ; 174(14): 2358-2372, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28471490

ABSTRACT

BACKGROUND AND PURPOSE: The ω-3 polyunsaturated fatty acids (PUFAs) mediate protective effects on several metabolic disorders. However, the functions of their metabolites in the early stage of nonalcoholic fatty liver disease (NAFLD) are largely unknown. EXPERIMENTAL APPROACH: Mice were fed a control diet, high-fat diet (HFD) or ω-3 PUFA-enriched HFD (ω3HFD) for 4 days and phenotypes were analysed. LC-MS/MS was used to determine the eicosanoid profiles. Primary hepatocytes and peritoneal macrophages were used for the mechanism study. KEY RESULTS: In short-term HFD-fed mice, the significantly increased lipid accumulation in the liver was reversed by ω-3 PUFA supplementation. Metabolomics showed that the plasma concentrations of hydroxyeicosapentaenoic acids (HEPEs) and epoxyeicosatetraenoic acids (EEQs) were reduced by a short-term HFD and markedly increased by the ω3HFD. However, HEPE/EEQ treatment had no direct protective effect on hepatocytes. ω3HFD also significantly attenuated HFD-induced adipose tissue inflammation. Furthermore, the expression of pro-inflammatory cytokines and activation of the JNK pathway induced by palmitate were suppressed by HEPEs and EEQs in macrophages. 17,18-EEQ, 5-HEPE and 9-HEPE were identified as the effective components among these metabolites, as indicated by their greater suppression of the palmitate-induced expression of inflammatory factors, chemotaxis and JNK activation compared to other metabolites in macrophages. A mixture of 17,18-EEQ, 5-HEPE and 9-HEPE significantly ameliorated the short-term HFD-induced accumulation of macrophages in adipose tissue and hepatic steatosis. CONCLUSION AND IMPLICATIONS: 17,18-EEQ, 5-HEPE and 9-HEPE may be potential approaches to prevent NAFLD in the early stage by inhibiting the inflammatory response in adipose tissue macrophages via JNK signalling.


Subject(s)
Arachidonic Acids/pharmacology , Eicosapentaenoic Acid/analogs & derivatives , Non-alcoholic Fatty Liver Disease/prevention & control , Animals , Arachidonic Acids/administration & dosage , Arachidonic Acids/metabolism , Eicosapentaenoic Acid/administration & dosage , Eicosapentaenoic Acid/metabolism , Eicosapentaenoic Acid/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology
SELECTION OF CITATIONS
SEARCH DETAIL