Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.439
Filter
1.
Clin Transl Sci ; 17(6): e13760, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38847320

ABSTRACT

Metabolic dysfunction-associated steatohepatitis (MASH) is the severe form of non-alcoholic fatty liver disease which has a high potential to progress to cirrhosis and hepatocellular carcinoma, yet adequate effective therapies are lacking. Hypoadiponectinemia is causally involved in the pathogenesis of MASH. This study investigated the pharmacological effects of adiponectin replacement therapy with the adiponectin-derived peptide ALY688 (ALY688-SR) in a mouse model of MASH. Human induced pluripotent stem (iPS) cell-derived hepatocytes were used to test cytotoxicity and signaling of unmodified ALY688 in vitro. High-fat diet with low methionine and no added choline (CDAHF) was used to induce MASH and test the effects of ALY688-SR in vivo. Histological MASH activity score (NAS) and fibrosis score were determined to assess the effect of ALY688-SR. Transcriptional characterization of mice through RNA sequencing was performed to indicate potential molecular mechanisms involved. In cultured hepatocytes, ALY688 efficiently induced adiponectin-like signaling, including the AMP-activated protein kinase and p38 mitogen-activated protein kinase pathways, and did not elicit cytotoxicity. Administration of ALY688-SR in mice did not influence body weight but significantly ameliorated CDAHF-induced hepatic steatosis, inflammation, and fibrosis, therefore effectively preventing the development and progression of MASH. Mechanistically, ALY688-SR treatment markedly induced hepatic expression of genes involved in fatty acid oxidation, whereas it significantly suppressed the expression of pro-inflammatory and pro-fibrotic genes as demonstrated by transcriptomic analysis. ALY688-SR may represent an effective approach in MASH treatment. Its mode of action involves inhibition of hepatic steatosis, inflammation, and fibrosis, possibly via canonical adiponectin-mediated signaling.


Subject(s)
Adiponectin , Disease Models, Animal , Hepatocytes , Non-alcoholic Fatty Liver Disease , Animals , Adiponectin/metabolism , Adiponectin/pharmacology , Adiponectin/deficiency , Mice , Humans , Hepatocytes/metabolism , Hepatocytes/drug effects , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/prevention & control , Non-alcoholic Fatty Liver Disease/pathology , Non-alcoholic Fatty Liver Disease/etiology , Male , Mice, Inbred C57BL , Signal Transduction/drug effects , Diet, High-Fat/adverse effects , Metabolism, Inborn Errors/metabolism , Metabolism, Inborn Errors/drug therapy , Metabolism, Inborn Errors/pathology , Metabolic Diseases/drug therapy , Metabolic Diseases/metabolism , Metabolic Diseases/prevention & control , Metabolic Diseases/etiology , Liver/metabolism , Liver/drug effects , Liver/pathology , Fatty Liver/prevention & control , Fatty Liver/metabolism , Fatty Liver/drug therapy , Fatty Liver/pathology
2.
Redox Biol ; 74: 103230, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38875959

ABSTRACT

α-Ketoglutarate (AKG), a crucial intermediate in the tricarboxylic acid cycle, has been demonstrated to mitigate hyperlipidemia-induced dyslipidemia and endothelial damage. While hyperlipidemia stands as a major trigger for non-alcoholic fatty liver disease, the protection of AKG on hyperlipidemia-induced hepatic metabolic disorders remains underexplored. This study aims to investigate the potential protective effects and mechanisms of AKG against hepatic lipid metabolic disorders caused by acute hyperlipidemia. Our observations indicate that AKG effectively alleviates hepatic lipid accumulation, mitochondrial dysfunction, and loss of redox homeostasis in P407-induced hyperlipidemia mice, as well as in palmitate-injured HepG2 cells and primary hepatocytes. Mechanistic insights reveal that the preventive effects are mediated by activating the AMPK-PGC-1α/Nrf2 pathway. In conclusion, our findings shed light on the role and mechanism of AKG in ameliorating abnormal lipid metabolic disorders in hyperlipidemia-induced fatty liver, suggesting that AKG, an endogenous mitochondrial nutrient, holds promising potential for addressing hyperlipidemia-induced fatty liver conditions.


Subject(s)
AMP-Activated Protein Kinases , Hyperlipidemias , Ketoglutaric Acids , NF-E2-Related Factor 2 , Oxidative Stress , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Signal Transduction , Animals , Hyperlipidemias/metabolism , Hyperlipidemias/drug therapy , Hyperlipidemias/complications , Mice , Oxidative Stress/drug effects , Humans , NF-E2-Related Factor 2/metabolism , AMP-Activated Protein Kinases/metabolism , Ketoglutaric Acids/metabolism , Ketoglutaric Acids/pharmacology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Signal Transduction/drug effects , Hep G2 Cells , Mitochondria/metabolism , Mitochondria/drug effects , Male , Lipid Metabolism/drug effects , Hepatocytes/metabolism , Hepatocytes/drug effects , Fatty Liver/metabolism , Fatty Liver/etiology , Fatty Liver/drug therapy , Fatty Liver/prevention & control , Fatty Liver/pathology , Disease Models, Animal , Liver/metabolism , Liver/drug effects , Liver/pathology
3.
Int J Mol Sci ; 25(10)2024 May 07.
Article in English | MEDLINE | ID: mdl-38791103

ABSTRACT

Menopause is characterized by a reduction in sex hormones in women and is associated with metabolic changes, including fatty liver and insulin resistance. Lifestyle changes, including a balanced diet and physical exercise, are necessary to prevent these undesirable changes. Strength training (ST) has been widely used because of the muscle and metabolic benefits it provides. Our study aims to evaluate the effects of ST on hepatic steatosis and insulin resistance in ovariectomized mice fed a high-fat diet (HFD) divided into four groups as follows: simulated sedentary surgery (SHAM-SED), trained simulated surgery (SHAM-EXE), sedentary ovariectomy (OVX-SED), and trained ovariectomy (OVX-EXE). They were fed an HFD for 9 weeks. ST was performed thrice a week. ST efficiently reduced body weight and fat percentage and increased lean mass in OVX mice. Furthermore, ST reduced the accumulation of ectopic hepatic lipids, increased AMPK phosphorylation, and inhibited the de novo lipogenesis pathway. OVX-EXE mice also showed a better glycemic profile, associated with greater insulin sensitivity identified by the euglycemic-hyperinsulinemic clamp, and reduced markers of hepatic oxidative stress compared with sedentary animals. Our data support the idea that ST can be indicated as a non-pharmacological treatment approach to mitigate metabolic changes resulting from menopause.


Subject(s)
Diet, High-Fat , Fatty Liver , Insulin Resistance , Ovariectomy , Resistance Training , Animals , Female , Ovariectomy/adverse effects , Diet, High-Fat/adverse effects , Mice , Fatty Liver/metabolism , Fatty Liver/prevention & control , Physical Conditioning, Animal , Oxidative Stress , Liver/metabolism , Mice, Inbred C57BL , Body Weight , Lipogenesis
4.
Radiat Res ; 202(1): 1-10, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38772553

ABSTRACT

As the number of cancer survivors increases and the risk of accidental radiation exposure rises, there is a pressing need to characterize the delayed effects of radiation exposure and develop medical countermeasures. Radiation has been shown to damage adipose progenitor cells and increase liver fibrosis, such that it predisposes patients to developing metabolic-associated fatty liver disease (MAFLD) and insulin resistance. The risk of developing these conditions is compounded by the global rise of diets rich in carbohydrates and fats. Radiation persistently increases the signaling cascade of transforming growth factor ß (TGFß), leading to heightened fibrosis as characteristic of the delayed effects of radiation exposure. We investigate here a potential radiation medical countermeasure, IPW-5371, a small molecule inhibitor of TGFßRI kinase (ALK5). We found that mice exposed to sub-lethal whole-body irradiation and chronic Western diet consumption but treated with IPW-5371 had a similar body weight, food consumption, and fat mass compared to control mice exposed to radiation. The IPW-5371 treated mice maintained lower fibrosis and fat accumulation in the liver, were more responsive to insulin and had lower circulating triglycerides and better muscle endurance. Future studies are needed to verify the improvement by IPW-5371 on the structure and function of other metabolically active tissues such as adipose and skeletal muscle, but these data demonstrate that IPW-5371 protects liver and whole-body health in rodents exposed to radiation and a Western diet, and there may be promise in using IPW-5371 to prevent the development of MAFLD.


Subject(s)
Fatty Liver , Insulin Resistance , Animals , Mice , Fatty Liver/prevention & control , Fatty Liver/pathology , Male , Mice, Inbred C57BL , Receptors, Transforming Growth Factor beta/metabolism , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Diet , Whole-Body Irradiation/adverse effects
5.
Front Endocrinol (Lausanne) ; 15: 1282231, 2024.
Article in English | MEDLINE | ID: mdl-38756999

ABSTRACT

Introduction: Cigarettes containing nicotine (Nic) are a risk factor for the development of cardiovascular and metabolic diseases. We reported that Nic delivered via injections or e-cigarette vapor led to hepatic steatosis in mice fed with a high-fat diet. High-fructose corn syrup (HFCS) is the main sweetener in sugar-sweetened beverages (SSBs) in the US. Increased consumption of SSBs with HFCS is associated with increased risks of non-alcoholic fatty liver disease (NAFLD). Nicotinamide riboside (NR) increases mitochondrial nicotinamide adenine dinucleotide (NAD+) and protects mice against hepatic steatosis. This study evaluated if Nic plus Coca-Cola™ (Coke) with HFCS can cause hepatic steatosis and that can be protected by NR. Methods: C57BL/6J mice received twice daily intraperitoneal (IP) injections of Nic or saline and were given Coke (HFCS), or Coke with sugar, and NR supplementation for 10 weeks. Results: Our results show that Nic+Coke caused increased caloric intake and induced hepatic steatosis, and the addition of NR prevented these changes. Western blot analysis showed lipogenesis markers were activated (increased cleavage of the sterol regulatory element-binding protein 1 [SREBP1c] and reduction of phospho-Acetyl-CoA Carboxylase [p-ACC]) in the Nic+Coke compared to the Sal+Water group. The hepatic detrimental effects of Nic+Coke were mediated by decreased NAD+ signaling, increased oxidative stress, and mitochondrial damage. NR reduced oxidative stress and prevented mitochondrial damage by restoring protein levels of Sirtuin1 (Sirt1) and peroxisome proliferator-activated receptor coactivator 1-alpha (PGC1) signaling. Conclusion: We conclude that Nic+Coke has an additive effect on producing hepatic steatosis, and NR is protective. This study suggests concern for the development of NAFLD in subjects who consume nicotine and drink SSBs with HFCS.


Subject(s)
Mice, Inbred C57BL , Niacinamide , Nicotine , Pyridinium Compounds , Animals , Pyridinium Compounds/pharmacology , Mice , Niacinamide/analogs & derivatives , Niacinamide/pharmacology , Male , Non-alcoholic Fatty Liver Disease/prevention & control , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Non-alcoholic Fatty Liver Disease/etiology , Fatty Liver/prevention & control , Fatty Liver/metabolism , Fatty Liver/chemically induced , High Fructose Corn Syrup/adverse effects , Liver/metabolism , Liver/drug effects , Liver/pathology , Oxidative Stress/drug effects
6.
Nutr Res ; 126: 180-192, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38759501

ABSTRACT

Metabolic dysfunction-associated steatotic liver disease (MASLD) has attracted increasing attention from the scientific community because of its severe but silent progression and the lack of specific treatment. Glucolipotoxicity triggers endoplasmic reticulum (ER) stress with decreased beta-oxidation and enhanced lipogenesis, promoting the onset of MASLD, whereas regular physical exercise can prevent MASLD by preserving ER and mitochondrial function. Thus, the hypothesis of this study was that high-intensity interval training (HIIT) could prevent the development of MASLD in high-fat (HF)-fed C57BL/6J mice by maintaining insulin sensitivity, preventing ER stress, and promoting beta-oxidation. Forty male C57BL/6J mice (3 months old) comprised 4 experimental groups: the control (C) diet group, the C diet + HIIT (C-HIIT) group, the HF diet group, and the HF diet + HIIT (HF-HIIT) group. HIIT sessions lasted 12 minutes and were performed 3 times weekly by trained mice. The diet and exercise protocols lasted for 10 weeks. The HIIT protocol prevented weight gain and maintained insulin sensitivity in the HF-HIIT group. A chronic HF diet increased ER stress-related gene and protein expression, but HIIT helped to maintain ER homeostasis, preserve mitochondrial ultrastructure, and maximize beta-oxidation. The increased sirtuin-1/peroxisome proliferator-activated receptor-gamma coactivator 1-alpha expression implies that HIIT enhanced mitochondrial biogenesis and yielded adequate mitochondrial dynamics. High hepatic fibronectin type III domain containing 5/irisin agreed with the antilipogenic and anti-inflammatory effects observed in the HF-HIIT group, reinforcing the antisteatotic effects of HIIT. Thus, we confirmed that practicing HIIT 3 times per week maintained insulin sensitivity, prevented ER stress, and enhanced hepatic beta-oxidation, impeding MASLD development in this mouse model even when consuming high energy intake from saturated fatty acids.


Subject(s)
Diet, High-Fat , Endoplasmic Reticulum Stress , High-Intensity Interval Training , Insulin Resistance , Liver , Mice, Inbred C57BL , Mitochondria, Liver , Physical Conditioning, Animal , Animals , Diet, High-Fat/adverse effects , Male , Liver/metabolism , Mitochondria, Liver/metabolism , Mice , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/therapy , Fatty Liver/prevention & control , Oxidation-Reduction
7.
Nutrients ; 16(9)2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38732511

ABSTRACT

Prenatal alcohol exposure (AE) affects cognitive development. However, it is unclear whether prenatal AE influences the metabolic health of offspring and whether postnatal AE exacerbates metabolic deterioration resulting from prenatal AE. Choline is a semi-essential nutrient that has been demonstrated to mitigate the cognitive impairment of prenatal AE. This study investigated how maternal choline supplementation (CS) may modify the metabolic health of offspring with prenatal and postnatal AE (AE/AE). C57BL/6J female mice were fed either a Lieber-DeCarli diet with 1.4% ethanol between embryonic day (E) 9.5 and E17.5 or a control diet. Choline was supplemented with 4 × concentrations versus the control throughout pregnancy. At postnatal week 7, offspring mice were exposed to 1.4% ethanol for females and 3.9% ethanol for males for 4 weeks. AE/AE increased hepatic triglyceride accumulation in male offspring only, which was normalized by prenatal CS. Prenatal CS also improved glucose tolerance compared to AE/AE animals. AE/AE suppressed hepatic gene expression of peroxisome proliferator activated receptor alpha (Ppara) and low-density lipoprotein receptor (Ldlr), which regulate fatty acid catabolism and cholesterol reuptake, respectively, in male offspring. However, these changes were not rectified by prenatal CS. In conclusion, AE/AE led to an increased risk of steatosis and was partially prevented by prenatal CS in male mice.


Subject(s)
Choline , Dietary Supplements , Ethanol , Liver , Mice, Inbred C57BL , Prenatal Exposure Delayed Effects , Animals , Female , Pregnancy , Choline/administration & dosage , Male , Liver/metabolism , Liver/drug effects , Mice , Fatty Liver/prevention & control , Fatty Liver/etiology , Triglycerides/metabolism , PPAR alpha/metabolism , Receptors, LDL/genetics , Receptors, LDL/metabolism , Glucose Intolerance/prevention & control , Lipid Metabolism/drug effects
8.
Biomed Pharmacother ; 174: 116520, 2024 May.
Article in English | MEDLINE | ID: mdl-38581924

ABSTRACT

A combination of liver and heart dysfunction worsens the prognosis of human survival. The aim of this study was to investigate whether empagliflozin (a sodium-glucose transporter-2 inhibitor) has beneficial effects not only on cardiac and renal function but also on hepatic function. Adult (6-month-old) male spontaneously hypertensive rats (SHR) were fed a high-fat diet (60% fat) for four months to induce hepatic steatosis and mild heart failure. For the last two months, the rats were treated with empagliflozin (empa, 10 mg.kg-1.day-1 in the drinking water). Renal function and oral glucose tolerance test were analyzed in control (n=8), high-fat diet (SHR+HF, n=10), and empagliflozin-treated (SHR+HF+empa, n=9) SHR throughout the study. Metabolic parameters and echocardiography were evaluated at the end of the experiment. High-fat diet feeding increased body weight and visceral adiposity, liver triglyceride and cholesterol concentrations, and worsened glucose tolerance. Although the high-fat diet did not affect renal function, it significantly worsened cardiac function in a subset of SHR rats. Empagliflozin reduced body weight gain but not visceral fat deposition. It also improved glucose sensitivity and several metabolic parameters (plasma insulin, uric acid, and HDL cholesterol). In the liver, empagliflozin reduced ectopic lipid accumulation, lipoperoxidation, inflammation and pro-inflammatory HETEs, while increasing anti-inflammatory EETs. In addition, empagliflozin improved cardiac function (systolic, diastolic and pumping) independent of blood pressure. The results of our study suggest that hepatoprotection plays a decisive role in the beneficial effects of empagliflozin in preventing the progression of cardiac dysfunction induced by high-fat diet feeding.


Subject(s)
Benzhydryl Compounds , Diet, High-Fat , Glucosides , Liver , Rats, Inbred SHR , Sodium-Glucose Transporter 2 Inhibitors , Animals , Glucosides/pharmacology , Benzhydryl Compounds/pharmacology , Male , Diet, High-Fat/adverse effects , Liver/drug effects , Liver/metabolism , Liver/pathology , Rats , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Cardiotonic Agents/pharmacology , Blood Pressure/drug effects , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Fatty Liver/prevention & control , Fatty Liver/drug therapy , Blood Glucose/metabolism , Blood Glucose/drug effects , Protective Agents/pharmacology , Hypertension/drug therapy
9.
Br J Pharmacol ; 181(15): 2545-2565, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38599607

ABSTRACT

BACKGROUND AND PURPOSE: Bardoxolone methyl (2-cyano-3,12-dioxooleana-1,9(11)-dien-28-oic acid methyl ester, CDDO-Me) is a potent activator of nuclear factor erythroid 2-related factor 2 (Nrf2), which induces the expression of antioxidative-associated genes. CDDO-Me exerts protective effects against chronic inflammatory diseases in the kidneys and lungs. However, its pharmacological effects on metabolic dysfunction-associated steatohepatitis (MASH) caused by fat accumulation remain unknown. In this study, we examined the hepatoprotective effects of CDDO-Me in a diet-induced MASH mouse model and elucidated its pharmacological mechanisms using RNA-seq analysis. EXPERIMENTAL APPROACH: CDDO-Me was orally administered to mice fed a choline-deficient, L-amino acid-defined, high-fat diet (CDAHFD), and histological, biochemical, and transcriptomic analyses were performed on livers of mice that developed MASH. KEY RESULTS: CDDO-Me administration induced the expression of antioxidant genes and cholesterol transporters downstream of Nrf2 and significantly prevented the symptoms of MASH. Whole-transcriptome analysis revealed that CDDO-Me inhibited the inflammatory pathway that led to phagocyte recruitment, in addition to activating the Nrf2-dependent pathway. Among inflammatory pathways, CC chemokine ligands (CCL)3 and CCL4, which are downstream of NF-κB and are associated with the recruitment of macrophages expressing CC chemokine receptors (CCR)1 and CCR5, were released into the blood in MASH mice. However, CDDO-Me directly inhibited the expression of CCL3-CCR1 and CCL4-CCR5 in macrophages. CONCLUSIONS AND IMPLICATIONS: Overall, we revealed the potent hepatoprotective effect of CDDO-Me in a MASH mouse model and demonstrated that its pharmacological effects were closely associated with a reduction of macrophage infiltration, through CCL3-CCR1 and CCL4-CCR5 inhibition, in addition to Nrf2-mediated hepatoprotective effects.


Subject(s)
Diet, High-Fat , Macrophages , Mice, Inbred C57BL , Oleanolic Acid , Animals , Oleanolic Acid/analogs & derivatives , Oleanolic Acid/pharmacology , Oleanolic Acid/therapeutic use , Mice , Male , Macrophages/drug effects , Macrophages/metabolism , Diet, High-Fat/adverse effects , Fatty Liver/drug therapy , Fatty Liver/prevention & control , Fatty Liver/metabolism , NF-E2-Related Factor 2/metabolism , Receptors, CCR1/antagonists & inhibitors , Receptors, CCR1/metabolism , Liver/drug effects , Liver/metabolism , Liver/pathology , Receptors, CCR5
10.
Hepatol Commun ; 8(5)2024 May 01.
Article in English | MEDLINE | ID: mdl-38619434

ABSTRACT

BACKGROUND: Recent reports have unveiled the potential utility of l-carnitine to alleviate metabolic dysfunction-associated steatohepatitis (MASH) by enhancing mitochondrial metabolic function. However, its efficacy at preventing the development of HCC has not been assessed fully. METHODS: l-carnitine (2 g/d) was administered to 11 patients with MASH for 10 weeks, and blood liver function tests were performed. Five patients received a serial liver biopsy, and liver histology and hepatic gene expression were evaluated using this tissue. An atherogenic plus high-fat diet MASH mouse model received long-term l-carnitine administration, and liver histology and liver tumor development were evaluated. RESULTS: Ten-week l-carnitine administration significantly improved serum alanine transaminase and aspartate transaminase levels along with a histological improvement in the NAFLD activity score, while steatosis and fibrosis were not improved. Gene expression profiling revealed a significant improvement in the inflammation and profibrotic gene signature as well as the recovery of lipid metabolism. Long-term l-carnitine administration to atherogenic plus high-fat diet MASH mice substantially improved liver histology (inflammation, steatosis, and fibrosis) and significantly reduced the incidence of liver tumors. l-carnitine directly reduced the expression of the MASH-associated and stress-induced transcriptional factor early growth response 1. Early growth response 1 activated the promoter activity of neural precursor cell expressed, developmentally downregulated protein 9 (NEDD9), an oncogenic protein. Thus, l-carnitine reduced the activation of the NEDD9, focal adhesion kinase 1, and AKT oncogenic signaling pathway. CONCLUSIONS: Short-term l-carnitine administration ameliorated MASH through its anti-inflammatory effects. Long-term l-carnitine administration potentially improved the steatosis and fibrosis of MASH and may eventually reduce the risk of HCC.


Subject(s)
Carcinoma, Hepatocellular , Fatty Liver , Liver Neoplasms , Humans , Animals , Mice , Liver Neoplasms/prevention & control , Carcinoma, Hepatocellular/prevention & control , Fatty Liver/drug therapy , Fatty Liver/prevention & control , Carnitine/pharmacology , Carnitine/therapeutic use , Fibrosis , Inflammation , Adaptor Proteins, Signal Transducing
11.
Biomed Pharmacother ; 174: 116582, 2024 May.
Article in English | MEDLINE | ID: mdl-38642504

ABSTRACT

The aim of this study was to investigate whether the therapeutic effect of theabrownin extracted from Qingzhuan tea (QTB) on metabolic dysfunction-associated steatosis liver disease (MASLD) is related to the regulation of intestinal microbiota and its metabolite short-chain fatty acids (SCFAs). Mice were divided into four groups and received normal diet (ND), high-fat diet (HFD) and HFD+QTB (180, 360 mg/kg) for 8 weeks. The results showed that QTB significantly reduced the body weight of HFD mice, ameliorated liver lipid and dyslipidemia, and increased the level of intestinal SCFAs in HFD mice. The results of 16 S rRNA showed that the relative abundance of Bacteroides, Blautia and Lachnoclostridium and their main metabolites acetate and propionate were significantly increased after QTB intervention. The relative abundance of Colidextribacter, Faecalibaculum and Lactobacillus was significantly reduced. QTB can also significantly up-regulate the expression of ATGL, PPARα, FFAR2 and FFAR3, and inhibit the expression of LXRα, SREBP-1c, FAS and HMGCR genes. This makes it possible to act as a prebiotic to prevent MASLD.


Subject(s)
Catechin/analogs & derivatives , Diet, High-Fat , Gastrointestinal Microbiome , Mice, Inbred C57BL , Tea , Animals , Gastrointestinal Microbiome/drug effects , Diet, High-Fat/adverse effects , Male , Tea/chemistry , Mice , Fatty Acids, Volatile/metabolism , Liver/drug effects , Liver/metabolism , Lipid Metabolism/drug effects , Dyslipidemias/drug therapy , Dyslipidemias/prevention & control , Fatty Liver/prevention & control , Fatty Liver/drug therapy
12.
Diabetes Obes Metab ; 26(6): 2339-2348, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38504118

ABSTRACT

AIM: Dipeptidyl peptidase-4 (DPP-4) inhibitors suppress the inactivation of incretin hormones and lower blood glucose levels by inhibiting DPP-4 function. Sodium-glucose cotransporter 2 (SGLT2) inhibitors lower blood glucose levels in an insulin-independent manner by inhibiting renal reabsorption of glucose. DPP-4 and SGLT2 inhibitors each have the potential to improve hepatic steatosis; however, their combined effects remain unclear. In this study, we examined the effects of the combination of these drugs on hepatic steatosis using high-fat diet-fed mice. METHOD: C57BL/6J male mice were fed a 60% high-fat diet for 2 months to induce hepatic steatosis. Mice were divided into four groups (control; DPP-4 inhibitor anagliptin; SGLT2 inhibitor luseogliflozin; anagliptin and luseogliflozin combination), and the effects of each drug and their combination on hepatic steatosis after a 4-week intervention were evaluated. RESULTS: There were no differences in blood glucose levels among the four groups. Anagliptin suppresses inflammation- and chemokine-related gene expression. It also improved macrophage fractionation in the liver. Luseogliflozin reduced body weight, hepatic gluconeogenesis and blood glucose levels in the oral glucose tolerance test. The combination treatment improved hepatic steatosis without interfering with the effects of anagliptin and luseogliflozin, respectively, and fat content and inflammatory gene expression in the liver were significantly improved in the combination group compared with the other groups. CONCLUSION: The combination therapy with the DPP-4 inhibitor anagliptin and the SGLT2 inhibitor luseogliflozin inhibits fat deposition in the liver via anti-inflammatory effects during the early phase of diet-induced liver steatosis.


Subject(s)
Diet, High-Fat , Dipeptidyl-Peptidase IV Inhibitors , Fatty Liver , Sodium-Glucose Transporter 2 Inhibitors , Animals , Male , Mice , Blood Glucose/drug effects , Blood Glucose/metabolism , Diet, High-Fat/adverse effects , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Drug Synergism , Drug Therapy, Combination , Fatty Liver/prevention & control , Fatty Liver/drug therapy , Glucosides/pharmacology , Glucosides/therapeutic use , Liver/drug effects , Liver/metabolism , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/etiology , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Sodium-Glucose Transporter 2 Inhibitors/therapeutic use , Sorbitol/analogs & derivatives , Sorbitol/pharmacology , Sorbitol/therapeutic use
13.
Nutrients ; 16(6)2024 Mar 21.
Article in English | MEDLINE | ID: mdl-38542814

ABSTRACT

(1) Background: Modulators of the Neuropeptide Y (NPY) system are involved in energy metabolism, but the effect of NPY receptor antagonists on metabolic-dysfunction-associated steatotic liver disease (MASLD), a common obesity-related comorbidity, are largely unknown. In this study, we report on the effects of antagonists of the NPY-2 receptor (Y2R) in comparison with empagliflozin and semaglutide, substances that are known to be beneficial in MASLD. (2) Methods: Diet-induced obese (DIO) male Wistar rats were randomized into the following treatment groups: empagliflozin, semaglutide ± PYY3-36, the Y2R antagonists JNJ 31020028 and a food-restricted group, as well as a control group. After a treatment period of 8 weeks, livers were weighed and histologically evaluated. QrtPCR was performed to investigate liver inflammation and de novo lipogenesis (in liver and adipose tissue). Serum samples were analysed for metabolic parameters. (3) Results: Semaglutide + PYY3-36 led to significant weight loss, reduced liver steatosis (p = 0.05), and decreased inflammation, insulin resistance, and leptin levels. JNJ-31020028 prevented steatosis (p = 0.03) without significant weight loss. Hepatic downregulation of de novo lipogenesis-regulating genes (SREBP1 and MLXIPL) was observed in JNJ-31020028-treated rats (p ≤ 0.0001). Food restriction also resulted in significantly reduced weight, steatosis, and hepatic de novo lipogenesis. (4) Conclusions: Body weight reduction (e.g., by food restriction or drugs like semaglutide ± PYY3-36) is effective in improving liver steatosis in DIO rats. Remarkably, the body-weight-neutral Y2R antagonists may be effective in preventing liver steatosis through a reduction in de novo lipogenesis, making this drug class a candidate for the treatment of (early) MASLD.


Subject(s)
Benzamides , Benzhydryl Compounds , Fatty Liver , Glucagon-Like Peptides , Glucosides , Piperazines , Receptors, Neuropeptide Y , Rats , Male , Animals , Receptors, Neuropeptide Y/metabolism , Rats, Wistar , Obesity/complications , Obesity/drug therapy , Diet , Fatty Liver/drug therapy , Fatty Liver/etiology , Fatty Liver/prevention & control , Weight Loss , Inflammation
14.
Nutr Res ; 123: 111-119, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38310647

ABSTRACT

Chaenomeles sinensis (Thouin) Koehne fruit is a rich source of medicinally and nutritionally important natural phytochemicals that benefit human health. Based on the information provided, we hypothesized that Chaenomeles sinensis (Thouin) Koehne fruit polyphenols (CSFP) possessed in vivo protective effect of on high-fat diet (HFD)-induced obesity and hepatic steatosis. Specific pathogen-free male C57BL/6J mice were randomly divided into 3 groups and fed with a low-fat diet, HFD, or HFD supplemented with CSFP by intragastric administration for 14 weeks. Obesity-related biochemical indexes and hepatic gene expression profile were determined. The findings of this study demonstrated notable reductions in body weight gain, serum triglycerides, total cholesterol, low-density lipoprotein cholesterol, and steatosis grade in the group supplemented with CSFP compared with the HFD group. Gene expression analysis provided insights into the molecular mechanisms, demonstrating that CSFP downregulated the expression of key genes involved in lipogenesis (e.g., Fas, Fads2, Scd1) and upregulated the genes associated with fatty acid oxidation (e.g., Pparα, Cpt1a, Acox1), while also suppressing genes implicated in cholesterol homeostasis (e.g., HMGCoR, Insig1, AdipoR2). These molecular changes suggest that CSFP exerts protective effects by modulating hepatic lipid metabolism pathways, thereby mitigating the metabolic derangements associated with HFD-induced obesity and hepatic steatosis.


Subject(s)
Fatty Liver , Rosaceae , Humans , Male , Mice , Animals , Lipid Metabolism , Diet, High-Fat/adverse effects , Fruit/chemistry , Polyphenols/pharmacology , Polyphenols/therapeutic use , Polyphenols/analysis , Mice, Inbred C57BL , Fatty Liver/etiology , Fatty Liver/prevention & control , Fatty Liver/metabolism , Obesity/drug therapy , Obesity/etiology , Obesity/metabolism , Liver/metabolism , Cholesterol
15.
J Sci Food Agric ; 104(9): 5462-5473, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38348948

ABSTRACT

BACKGROUND: Obesity has been demonstrated as a risk factor that seriously affects health. Insoluble dietary fiber (IDF), as a major component of dietary fiber, has positive effects on obesity, inflammation and diabetes. RESULTS: In this study, complex IDF was prepared using 50% enoki mushroom IDF, 40% carrot IDF, and 10% oat IDF. The effects and potential mechanism of complex IDF on obesity were investigated in C57BL/6 mice fed a high-fat diet. The results showed that feeding diets containing 5% complex IDF for 8 weeks significantly reduced mouse body weight, epididymal lipid index, and ectopic fat deposition, and improved mouse liver lipotoxicity (reduced serum levels of alanine aminotransferase, aspartate aminotransferase, and alkaline phosphatase), fatty liver, and short-chain fatty acid composition. High-throughput sequencing of 16S rRNA and analysis of fecal metabolomics showed that the intervention with complex IDF reversed the high-fat-diet-induced dysbiosis of gut microbiota, which is associated with obesity and intestinal inflammation, and affected metabolic pathways, such as primary bile acid biosynthesis, related to fat digestion and absorption. CONCLUSION: Composite IDF intervention can effectively inhibit high-fat-diet-induced obesity and related symptoms and affect the gut microbiota and related metabolic pathways in obesity. Complex IDF has potential value in the prevention of obesity and metabolic syndrome. © 2024 Society of Chemical Industry.


Subject(s)
Diet, High-Fat , Dietary Fiber , Gastrointestinal Microbiome , Liver , Mice, Inbred C57BL , Obesity , Animals , Dietary Fiber/metabolism , Diet, High-Fat/adverse effects , Obesity/metabolism , Obesity/diet therapy , Obesity/microbiology , Mice , Male , Liver/metabolism , Humans , Bacteria/classification , Bacteria/isolation & purification , Bacteria/metabolism , Bacteria/genetics , Fatty Liver/prevention & control , Fatty Liver/metabolism , Fatty Liver/etiology , Avena/chemistry , Daucus carota/chemistry
16.
Nutr Res ; 124: 43-54, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38367426

ABSTRACT

Kimchi is a traditional fermented food that contains abundant nutrients and functional ingredients with various health benefits. We previously reported that kimchi active components suppress hepatic steatosis caused by endoplasmic reticulum (ER) stress in vitro and in vivo. Therefore, we assessed the effect of kimchi on the inhibition of hepatic steatosis caused by ER stress in HepG2 cells and C57BL/6N mice to verify the hypothesis that kimchi may potentially inhibit nonalcoholic fatty liver disease. We investigated the effect of kimchi on cell viability and triglyceride concentrations in cells and on lipid profile, lipid accumulation, and expression of related genes in cells and mice with hepatic steatosis. A mechanistic study was also performed using the liver X receptor α agonist T0901317 and the AMP-activated protein kinase agonist AICAR. Kimchi was noncytotoxic and effectively reduced triglyceride concentrations and suppressed hepatic steatosis-related gene expression in cells and mice. Additionally, kimchi recovered weight loss, lowered the serum and liver tissue lipid profiles, suppressed lipid accumulation, and reduced the effects of T0901317 and AICAR on lipogenic gene expression in tunicamycin-treated mice. Our results highlight that kimchi could prevent hepatic steatosis caused by ER stress in cells and mice.


Subject(s)
Aminoimidazole Carboxamide/analogs & derivatives , Benzenesulfonamides , Endoplasmic Reticulum Stress , Fermented Foods , Fluorocarbons , Liver , Mice, Inbred C57BL , Triglycerides , Animals , Endoplasmic Reticulum Stress/drug effects , Humans , Hep G2 Cells , Triglycerides/blood , Triglycerides/metabolism , Male , Liver/metabolism , Liver/drug effects , Mice , Aminoimidazole Carboxamide/pharmacology , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/prevention & control , Non-alcoholic Fatty Liver Disease/etiology , Sulfonamides/pharmacology , Ribonucleotides/pharmacology , AMP-Activated Protein Kinases/metabolism , Lipid Metabolism/drug effects , Cell Survival/drug effects , Liver X Receptors/metabolism , Tunicamycin/pharmacology , Lipogenesis/drug effects , Fatty Liver/drug therapy , Fatty Liver/prevention & control
17.
Clin Sci (Lond) ; 138(5): 327-349, 2024 03 06.
Article in English | MEDLINE | ID: mdl-38381799

ABSTRACT

Metabolic dysfunction-associated steatohepatitis (MASH) is always accompanied with hepatic fibrosis that could potentially progress to liver cirrhosis and hepatocellular carcinoma. Employing a rat model, we evaluated the role of human placental extract (HPE) to arrest the progression of hepatic fibrosis to cirrhosis in patients with MASH. SHRSP5/Dmcr rats were fed with a high-fat and high-cholesterol diet for 4 weeks and evaluated for the development of steatosis. The animals were divided into control and treated groups and received either saline or HPE (3.6 ml/kg body weight) subcutaneously thrice a week. A set of animals were killed at the end of 6th, 8th, and 12th weeks from the beginning of the experiment. Serum aspartate aminotransferase (AST), alanine aminotransferase (ALT), hepatic malondialdehyde (MDA), and glutathione content were measured. Immunohistochemical staining was performed for α-smooth muscle actin (α-SMA), 4-hydroxy-2-nonenal (4-HNE), collagen type I, and type III. Control rats depicted progression of liver fibrosis at 6 weeks, advanced fibrosis and bridging at 8 weeks, and cirrhosis at 12 weeks, which were significantly decreased in HPE-treated animals. Treatment with HPE maintained normal levels of MDA and glutathione in the liver. There was marked decrease in the staining intensity of α-SMA, 4-HNE, and collagen type I and type III in HPE treated rats compared with control animals. The results of the present study indicated that HPE treatment mediates immunotropic, anti-inflammatory, and antioxidant responses and attenuates hepatic fibrosis and early cirrhosis. HPE depicts therapeutic potential to arrest the progression of MASH towards cirrhosis.


Subject(s)
Fatty Liver , Non-alcoholic Fatty Liver Disease , Placental Extracts , Humans , Pregnancy , Rats , Female , Animals , Placental Extracts/metabolism , Placental Extracts/therapeutic use , Collagen Type I/metabolism , Placenta/metabolism , Liver Cirrhosis/drug therapy , Liver Cirrhosis/prevention & control , Fatty Liver/drug therapy , Fatty Liver/prevention & control , Liver/metabolism , Fibrosis , Glutathione/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Diet, High-Fat
18.
Poult Sci ; 103(2): 103286, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38100949

ABSTRACT

In this study, we evaluated the enrichment efficiency of lutein in eggs and its function in preventing fatty liver hemorrhagic syndrome (FLHS) in aged laying hens. Five groups of laying hens (65 wk old) were fed basal diets supplemented with 0, 30, 60, 90, or 120 mg/kg of lutein. The supplementation period lasted 12 wk followed by 2 wk of lutein depletion in feed. The results revealed that lutein efficiently enriched the egg yolks and improved their color with a significant increase in relative redness (P < 0.001). Lutein accumulation increased in the egg yolk until day 10, then depletion reached a minimum level after 14 d. Overall, zeaxanthin content in all the groups was similar throughout the experimental period. However, triglycerides and total cholesterol were significantly decreased in the liver (P < 0.05) but not significantly different in the serum (P > 0.05). In the serum, the lipid metabolism enzyme acetyl-CoA synthetase was significantly reduced (P < 0.05), whereas dipeptidyl-peptidase 4 was not significantly different (P > 0.05), and there was no statistical difference of either enzyme in the liver (P > 0.05). Regarding oxidation and inflammation-related indexes, malondialdehyde, tumor necrosis factors alpha, interleukin-6, and interleukin-1 beta were decreased, whereas superoxide dismutase and total antioxidant capacity increased in the liver (P < 0.001). The function of lutein for the same indexes in serum was limited. It was concluded that lutein efficiently enriched the egg yolk of old laying hens to improve their color and reached the highest level on day 10 without being subject to a significant conversion into zeaxanthin. At the same time, lutein prevented liver steatosis in aged laying hens by exerting strong antioxidant and anti-inflammatory functions, but also through the modulation of lipid metabolism, which may contribute to reducing the incidence of FLHS in poultry.


Subject(s)
Abnormalities, Multiple , Craniofacial Abnormalities , Fatty Liver , Growth Disorders , Heart Septal Defects, Ventricular , Lutein , Female , Animals , Lutein/metabolism , Antioxidants/metabolism , Chickens/metabolism , Zeaxanthins/metabolism , Dietary Supplements/analysis , Diet/veterinary , Egg Yolk/metabolism , Fatty Liver/prevention & control , Fatty Liver/veterinary , Animal Feed/analysis
19.
Int J Mol Sci ; 24(24)2023 Dec 09.
Article in English | MEDLINE | ID: mdl-38139133

ABSTRACT

Berberine (BBR) is a natural alkaloid with multiple biotical effects that has potential as a treatment for fatty liver hemorrhagic syndrome (FLHS). However, the mechanism underlying the protective effect of BBR against FLHS remains unclear. The present study aimed to investigate the effect of BBR on FLHS induced by a high-energy, low-protein (HELP) diet and explore the involvement of the gut microbiota and bile acid metabolism in the protective effects. A total of 90 healthy 140-day-old Hy-line laying hens were randomly divided into three groups, including a control group (fed a basic diet), a HELP group (fed a HELP diet), and a HELP+BBR group (high-energy, high-protein diet supplemented with BBR instead of maize). Our results show that BBR supplementation alleviated liver injury and hepatic steatosis in laying hens. Moreover, BBR supplementation could significantly regulate the gut's microbial composition, increasing the abundance of Actinobacteria and Romboutsia. In addition, the BBR supplement altered the profile of bile acid. Furthermore, the gut microbiota participates in bile acid metabolism, especially taurochenodeoxycholic acid and α-muricholic acid. BBR supplementation could regulate the expression of genes and proteins related to glucose metabolism, lipid synthesis (FAS, SREBP-1c), and bile acid synthesis (FXR, CYP27a1). Collectively, our findings demonstrate that BBR might be a potential feed additive for preventing FLHS by regulating the gut microbiota and bile acid metabolism.


Subject(s)
Berberine , Fatty Liver , Gastrointestinal Microbiome , Animals , Female , Berberine/pharmacology , Berberine/therapeutic use , Berberine/metabolism , Diet, Protein-Restricted , Chickens , Fatty Liver/drug therapy , Fatty Liver/etiology , Fatty Liver/prevention & control , Liver/metabolism , Bile Acids and Salts/metabolism
20.
J Med Food ; 26(12): 911-918, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37971778

ABSTRACT

The health benefits of soy foods are attributed to the high-quality protein and the bioactive compounds such as isoflavones. We previously reported that feeding obese (fa/fa) Zucker rats soy protein concentrates (SPCs) with low isoflavone (LIF) and high isoflavone (HIF) for 9 weeks significantly reduced liver steatosis compared to a casein control (C) diet. The current study extended the dietary treatments to 18 weeks to investigate the long-term effect of LIF and HIF SPC diets. 6-week-old male lean (L, n = 21) and obese (O, n = 21) Zucker rats were fed a casein C diet, LIF and HIF SPC diets for 18 weeks and body weight (BW) was recorded twice weekly. Rats were killed after 18 weeks to measure liver steatosis and serum aspartate aminotransferase and alanine aminotransferase. Obese rats had significantly greater final BW, liver weight, liver weight as the percentage of BW, and steatosis score compared to lean rats in all three dietary groups. The obese high-isoflavones (OHIF) group had significantly higher BW compared to obese control (OC) group (P < .0001) and obese low-isoflavones (OLIF) group (P = .01). OC group had significantly greater liver weight, liver weight as the percentage of BW, and liver steatosis score compared to OLIF (P = .0077, P < .0001 and P < .0001, respectively) and OHIF (P = .0094, P < .0001, and P < .0001, respectively) groups. Taken together, long-term feeding of SPC diets protected against liver steatosis regardless of isoflavone levels.


Subject(s)
Fatty Liver , Isoflavones , Male , Rats , Animals , Soybean Proteins , Caseins/pharmacology , Isoflavones/pharmacology , Rats, Zucker , Fatty Liver/prevention & control , Liver/metabolism , Obesity/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...