Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25.414
Filter
1.
Cell Mol Life Sci ; 81(1): 329, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39090270

ABSTRACT

Decidualisation of the endometrium is a key event in early pregnancy, which enables embryo implantation. Importantly, the molecular processes impairing decidualisation in obese mothers are yet to be characterised. We hypothesise that impaired decidualisation in obese mice is mediated by the upregulation of leptin modulators, the suppressor of cytokine signalling 3 (SOCS3) and the protein tyrosine phosphatase non-receptor type 2 (PTPN2), together with the disruption of progesterone (P4)-signal transducer and activator of transcription (STAT3) signalling. After feeding mice with chow diet (CD) or high-fat diet (HFD) for 16 weeks, we confirmed the downregulation of P4 and oestradiol (E2) steroid receptors in decidua from embryonic day (E) 6.5 and decreased proliferation of stromal cells from HFD. In vitro decidualised mouse endometrial stromal cells (MESCs) and E6.5 deciduas from the HFD showed decreased expression of decidualisation markers, followed by the upregulation of SOCS3 and PTPN2 and decreased phosphorylation of STAT3. In vivo and in vitro leptin treatment of mice and MESCs mimicked the results observed in the obese model. The downregulation of Socs3 and Ptpn2 after siRNA transfection of MESCs from HFD mice restored the expression level of decidualisation markers. Finally, DIO mice placentas from E18.5 showed decreased labyrinth development and vascularisation and fetal growth restricted embryos. The present study revealed major defects in decidualisation in obese mice, characterised by altered uterine response to E2 and P4 steroid signalling. Importantly, altered hormonal response was associated with increased expression of leptin signalling modulators SOCS3 and PTPN2. Elevated levels of SOCS3 and PTPN2 were shown to molecularly affect decidualisation in obese mice, potentially disrupting the STAT3-PR regulatory molecular hub.


Subject(s)
Decidua , Fetal Growth Retardation , Leptin , Mice, Obese , Placenta , Protein Tyrosine Phosphatase, Non-Receptor Type 2 , STAT3 Transcription Factor , Signal Transduction , Suppressor of Cytokine Signaling 3 Protein , Animals , Female , Suppressor of Cytokine Signaling 3 Protein/metabolism , Suppressor of Cytokine Signaling 3 Protein/genetics , Pregnancy , Leptin/metabolism , Decidua/metabolism , Decidua/pathology , Protein Tyrosine Phosphatase, Non-Receptor Type 2/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 2/genetics , Mice , Placenta/metabolism , STAT3 Transcription Factor/metabolism , Fetal Growth Retardation/metabolism , Fetal Growth Retardation/pathology , Obesity/metabolism , Obesity/pathology , Progesterone/metabolism , Mice, Inbred C57BL , Diet, High-Fat/adverse effects , Stromal Cells/metabolism
2.
FASEB J ; 38(15): e23857, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39114953

ABSTRACT

Atherosclerotic plaque formation is largely attributed to the impaired efferocytosis, which is known to be associated with the pathologic upregulation of cluster of differentiation 47 (CD47), a key antiphagocytic molecule. By gene expression omnibus (GEO) datasets analysis, we identified that four miRNAs are aberrantly downregulated in atherosclerosis, coronary artery disease, and obesity. Of them, hsa-miR-299-3p (miR-299-3p) was predicted to target the 3'UTR of human CD47 mRNA by bioinformatics analysis. Further, we demonstrated that miR-299-3p negatively regulates CD47 expression by binding to the target sequence "CCCACAU" in the 3'UTR of CD47 mRNA through luciferase reporter assay and site-directed mutagenesis. Additionally, we found that miR-299-3p was downregulated by ~32% in foam cells in response to oxidized low-density lipoprotein (ox-LDL) stimulation, thus upregulating CD47 and contributing to the impaired efferocytosis. Whereas, restoration of miR-299-3p reversed the ox-LDL-induced upregulation of CD47, thereby facilitating efferocytosis. In high-fat diet (HFD) fed ApoE-/- mice, we discovered that miR-299-3p was downregulated thus leading to upregulation of CD47 in abdominal aorta. Conversely, miR-299-3p restoration potently suppressed HFD-induced upregulation of CD47 and promoted phagocytosis of foam cells by macrophages in atherosclerotic plaques, thereby reducing necrotic core, increasing plaque stability, and mitigating atherosclerosis. Conclusively, we identify miR-299-3p as a negative regulator of CD47, and reveal a molecular mechanism whereby the ox-LDL-induced downregulation of miR-299-3p leads to the upregulation of CD47 in foam cells thus contributing to the impaired efferocytosis in atherosclerosis, and propose miR-299-3p can potentially serve as an inhibitor of CD47 to promote efferocytosis and ameliorate atherosclerosis.


Subject(s)
Atherosclerosis , CD47 Antigen , Efferocytosis , MicroRNAs , Animals , Humans , Mice , 3' Untranslated Regions , Atherosclerosis/metabolism , Atherosclerosis/genetics , Atherosclerosis/pathology , CD47 Antigen/metabolism , CD47 Antigen/genetics , Diet, High-Fat/adverse effects , Foam Cells/metabolism , Foam Cells/pathology , Lipoproteins, LDL/metabolism , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/metabolism
3.
Nat Commun ; 15(1): 6622, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39103342

ABSTRACT

Sex steroids modulate the distribution of mammalian white adipose tissues. Moreover, WAT remodeling requires adipocyte progenitor cells. Nevertheless, the sex-dependent mechanisms regulating adipocyte progenitors remain undetermined. Here, we uncover Cxcr4 acting in a sexually dimorphic manner to affect a pool of proliferating cells leading to restriction of female fat mass. We find that deletion of Cxcr4 in Pparγ-expressing cells results in female, not male, lipodystrophy, which cannot be restored by high-fat diet consumption. Additionally, Cxcr4 deletion is associated with a loss of a pool of proliferating adipocyte progenitors. Cxcr4 loss is accompanied by the upregulation of estrogen receptor alpha in adipose-derived PPARγ-labelled cells related to estradiol hypersensitivity and stalled adipogenesis. Estrogen removal or administration of antiestrogens restores WAT accumulation and dynamics of adipose-derived cells in Cxcr4-deficient mice. These findings implicate Cxcr4 as a female adipogenic rheostat, which may inform strategies to target female adiposity.


Subject(s)
Adipocytes , Adipogenesis , Adiposity , PPAR gamma , Receptors, CXCR4 , Stem Cells , Animals , Receptors, CXCR4/metabolism , Receptors, CXCR4/genetics , Female , Male , Mice , Adipocytes/metabolism , Adipocytes/cytology , Stem Cells/metabolism , Stem Cells/cytology , PPAR gamma/metabolism , PPAR gamma/genetics , Mice, Knockout , Adipose Tissue, White/metabolism , Adipose Tissue, White/cytology , Diet, High-Fat/adverse effects , Estrogen Receptor alpha/metabolism , Estrogen Receptor alpha/genetics , Mice, Inbred C57BL , Estradiol/pharmacology , Estradiol/metabolism , Cell Proliferation , Sex Factors , Sex Characteristics
4.
Clin Exp Med ; 24(1): 176, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39105860

ABSTRACT

Biotin and arginine play crucial roles in lipid metabolism and may offer promising interventions against obesity. This study examined the combined effect of magnesium biotinate (MgB) and inositol-stabilized arginine silicate complex (ASI) on obesity-related oxidative imbalance, inflammation, lipid metabolism and neuromodulation in rats on a high-fat diet (HFD). Forty rats were divided into five groups: (a) control: rats were fed a standard diet containing 12% of energy from fat; (b) HFD: rats were fed the HFD with 42% of energy from fat; (c) HFD + MgB: rats were fed the HFD and given 0.31 mg/kg body weight (BW) MgB, (d) HFD + ASI: rats were fed the HFD and were given 12.91 mg/kg BW ASI), and (e) HFD + MgB + ASI: rats were fed the HFD and given 0.31 mg/kg BW MgB and 12.91 mg/kg BW ASI). The combined administration of MgB and ASI reduced the levels of serum cholesterol, free fatty acid (FFA), and malondialdehyde (MDA), as well as liver inflammatory cytokines, sterol regulatory element-binding protein 1-c (SREBP-1c), and 3-hydroxy-3-methylglutaryl-CoA reductase (HMGR) proteins (P < 0.001) compared to HFD rats without supplementation. Moreover, this combination increased the activities of antioxidant enzymes (P < 0.05) and boosted the brain-derived neurotrophic factor (BDNF), serotonin, dopamine (P < 0.001), as well as liver insulin receptor substrate 1 (IRS-1) and peroxisome proliferator-activated receptor gamma (PPAR-γ) (P < 0.001). These findings suggest that combining MgB and ASI could deter liver fat accumulation and enhance lipid metabolism in HFD-fed rats by modulating various metabolic pathways and neuromodulators related to energy metabolism. This combination demonstrates potential in addressing obesity and its related metabolic dysfunctions.


Subject(s)
Antioxidants , Arginine , Diet, High-Fat , Animals , Diet, High-Fat/adverse effects , Rats , Arginine/pharmacology , Arginine/metabolism , Male , Antioxidants/pharmacology , Antioxidants/metabolism , Silicates/pharmacology , Obesity/metabolism , Inflammation/metabolism , Lipid Metabolism/drug effects , Neurotransmitter Agents/metabolism , Liver/metabolism , Liver/drug effects , Oxidative Stress/drug effects , Disease Models, Animal
5.
Nat Commun ; 15(1): 6697, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39107299

ABSTRACT

The skeleton has been suggested to function as an endocrine organ controlling whole organism energy balance, however the mediators of this effect and their molecular links remain unclear. Here, utilizing Schnurri-3-/- (Shn3-/-) mice with augmented osteoblast activity, we show Shn3-/-mice display resistance against diet-induced obesity and enhanced white adipose tissue (WAT) browning. Conditional deletion of Shn3 in osteoblasts but not adipocytes recapitulates lean phenotype of Shn3-/-mice, indicating this phenotype is driven by skeleton. We further demonstrate osteoblasts lacking Shn3 can secrete cytokines to promote WAT browning. Among them, we identify a C-terminal fragment of SLIT2 (SLIT2-C), primarily secreted by osteoblasts, as a Shn3-regulated osteokine that mediates WAT browning. Lastly, AAV-mediated Shn3 silencing phenocopies the lean phenotype and augmented glucose metabolism. Altogether, our findings establish a novel bone-fat signaling axis via SHN3 regulated SLIT2-C production in osteoblasts, offering a potential therapeutic target to address both osteoporosis and metabolic syndrome.


Subject(s)
Adipose Tissue, White , Bone and Bones , Diet, High-Fat , Intercellular Signaling Peptides and Proteins , Mice, Knockout , Obesity , Osteoblasts , Animals , Obesity/metabolism , Obesity/genetics , Obesity/etiology , Adipose Tissue, White/metabolism , Osteoblasts/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Mice , Diet, High-Fat/adverse effects , Bone and Bones/metabolism , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Male , Adipose Tissue, Brown/metabolism , Mice, Inbred C57BL , Adipocytes/metabolism , Signal Transduction
6.
PLoS One ; 19(8): e0308445, 2024.
Article in English | MEDLINE | ID: mdl-39110747

ABSTRACT

The LBX1 gene is located near a single nucleotide polymorphism that is highly associated with susceptibility to adolescent idiopathic scoliosis and is considered one of the strongest candidate genes involved in the pathogenesis of this condition. We have previously found that loss of LBX1 from skeletal muscle results not only in spinal deformity but also in lean body mass, suggesting a potential role for LBX1 in energy metabolism. The purpose of the present study was to test this hypothesis by analyzing the phenotype of mice lacking LBX1 in skeletal muscle with a focus on energy metabolism. We found that loss of LBX1 rendered mice more resistant to high-fat diet-induced obesity, despite comparable food intake between mutant and control mice. Notably, the mutant mice exhibited improved glucose tolerance, increased maximal aerobic capacity, and higher core body temperature compared to control mice. In addition, we found that overexpression of LBX1 decreased glucose uptake in cultured cells. Taken together, our data show that LBX1 functions as a negative regulator of energy metabolism and that loss of LBX1 from skeletal muscle increases systemic energy expenditure resulting in lean body mass. The present study thus suggests a potential association between LBX1 dysfunction and lean body mass in patients with adolescent idiopathic scoliosis.


Subject(s)
Energy Metabolism , Muscle, Skeletal , Animals , Mice , Muscle, Skeletal/metabolism , Obesity/metabolism , Obesity/genetics , Diet, High-Fat/adverse effects , Glucose/metabolism , Male , Humans , Mice, Knockout , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Scoliosis/genetics , Scoliosis/metabolism , Mice, Inbred C57BL
7.
Metabolomics ; 20(5): 96, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-39110263

ABSTRACT

INTRODUCTION: Ginseng berry (GB) has previously been demonstrated to improve systemic insulin resistance and regulate hepatic glucose metabolism and steatosis in mice with diet-induced obesity (DIO). OBJECTIVES: In this study, the role of GB in metabolism was assessed using metabolomics analysis on the total liver metabolites of DIO mice. METHODS: Metabolomic profiling was performed using capillary electrophoresis time-of-flight mass spectrometry (CE-TOF/MS) of liver tissue from mice on a 12-wk normal chow diet (NC), high-fat diet (HFD), and HFD supplemented with 0.1% GB (HFD + GB). The detected metabolites, its pathways, and functions were analyzed through partial least square discriminant analysis (PLS-DA), the small molecular pathway database (SMPDB), and MetaboAnalyst 5.0. RESULTS: The liver metabolite profiles of NC, HFD, and GB-fed mice (HFD + GB) were highly compartmentalized. Metabolites involved in major liver functions, such as mitochondrial function, gluconeogenesis/glycolysis, fatty acid metabolism, and primary bile acid biosynthesis, showed differences after GB intake. The metabolites that showed significant correlations with fasting blood glucose (FBG), insulin, and homeostatic model assessment for insulin resistance (HOMA-IR) were highly associated with mitochondrial membrane function, energy homeostasis, and glucose metabolism. Ginseng berry intake increased the levels of metabolites involved in mitochondrial membrane function, decreased the levels of metabolites related to glucose metabolism, and was highly correlated with metabolic phenotypes. CONCLUSION: This study demonstrated that long-term intake of GB changed the metabolite of hepatosteatotic livers in DIO mice, normalizing global liver metabolites involved in mitochondrial function and glucose metabolism and indicating the potential mechanism of GB in ameliorating hyperglycemia in DIO mice.


Subject(s)
Diet, High-Fat , Glucose , Liver , Metabolomics , Obesity , Panax , Animals , Panax/metabolism , Panax/chemistry , Mice , Metabolomics/methods , Liver/metabolism , Glucose/metabolism , Male , Obesity/metabolism , Mice, Inbred C57BL , Mitochondria/metabolism , Mitochondria/drug effects , Mice, Obese , Insulin Resistance , Fruit/metabolism , Fruit/chemistry , Metabolome/drug effects , Mitochondria, Liver/metabolism , Mitochondria, Liver/drug effects
8.
Gen Physiol Biophys ; 43(5): 411-421, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39140684

ABSTRACT

S-adenosylmethionine (SAM) is the main methyl group donor and has antioxidant potential. In this study, preventive and regressive potential of SAM were investigated in high fat/high cholesterol (HFHC) diet-induced non-alcoholic fatty liver disease (NAFLD) in guinea pigs. They were injected with SAM (50 mg/kg, i.p.) for 6 weeks along with HFHC diet or 4 weeks after HFHC diet. Serum transaminase activities, total cholesterol (TC), triglyceride (TG), cytochrome p450-2E1 (CYP2E1) and hydroxyproline (Hyp) levels, prooxidative and antioxidative parameters, protein expressions of α-smooth muscle actin (α-SMA) and transforming growth factor-ß1 (TGF-ß1) together with histopathological changes were examined in the liver. SAM treatment diminished HFHC diet-induced increases in serum transaminase activities and hepatic TC, TG, CYP2E1, Hyp, α-SMA and TGF-ß1 expressions and ameliorated prooxidant-antioxidant balance. Histopathological scores for hepatic steatosis, inflammation, and fibrosis were decreased by SAM treatment. Increases in TC, diene conjugate levels, and lipid vacuoles within the tunica media of the aorta were reduced in HFHC-fed animals treated with SAM. These protective effects were also detected in the regression period of HFHC-guinea pigs due to SAM. In conclusion, SAM treatment was found to be effective in prevention and regression of HFHC-induced hepatic and aortic lesions together with decreases in oxidative stress in guinea pigs with NAFLD.


Subject(s)
Diet, High-Fat , Liver , Oxidative Stress , S-Adenosylmethionine , Animals , Guinea Pigs , Oxidative Stress/drug effects , Diet, High-Fat/adverse effects , Male , S-Adenosylmethionine/metabolism , S-Adenosylmethionine/pharmacology , Liver/drug effects , Liver/metabolism , Liver/pathology , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Aortic Diseases/prevention & control , Aortic Diseases/pathology , Aortic Diseases/metabolism , Aortic Diseases/etiology , Aorta/drug effects , Aorta/pathology , Aorta/metabolism
9.
Front Endocrinol (Lausanne) ; 15: 1432928, 2024.
Article in English | MEDLINE | ID: mdl-39104812

ABSTRACT

We recently reported that a novel chimeric peptide (GEP44) targeting both the glucagon-like peptide-1 receptor (GLP-1R) and neuropeptide Y1- and Y2 receptor (Y1R and Y2R) reduced energy intake and body weight (BW) in diet-induced obese (DIO) rats. We hypothesized that GEP44 reduces energy intake and BW primarily through a GLP-1R dependent mechanism. To test this hypothesis, GLP-1R+/+ mice and GLP-1R null (GLP-1R-/-) mice were fed a high fat diet for 4 months to elicit diet-induced obesity prior to undergoing a sequential 3-day vehicle period, 3-day drug treatment (5, 10, 20 or 50 nmol/kg; GEP44 vs the selective GLP-1R agonist, exendin-4) and a 3-day washout. Energy intake, BW, core temperature and activity were measured daily. GEP44 (10, 20 and 50 nmol/kg) reduced BW after 3-day treatment in DIO male GLP-1R+/+ mice by -1.5 ± 0.6, -1.3 ± 0.4 and -1.9 ± 0.4 grams, respectively (P<0.05), with similar effects being observed in female GLP-1R+/+ mice. These effects were absent in male and female DIO GLP-1R-/- mice suggesting that GLP-1R signaling contributes to GEP44-elicited reduction of BW. Further, GEP44 decreased energy intake in both male and female DIO GLP-1R+/+ mice, but GEP44 appeared to produce more consistent effects across multiple doses in males. In GLP-1R-/- mice, the effects of GEP44 on energy intake were only observed in males and not females, suggesting that GEP44 may reduce energy intake, in part, through a GLP-1R independent mechanism in males. In addition, GEP44 reduced core temperature and activity in both male and female GLP-1R+/+ mice suggesting that it may also reduce energy expenditure. Lastly, we show that GEP44 reduced fasting blood glucose in DIO male and female mice through GLP-1R. Together, these findings support the hypothesis that the chimeric peptide, GEP44, reduces energy intake, BW, core temperature, and glucose levels in male and female DIO mice primarily through a GLP-1R dependent mechanism.


Subject(s)
Body Weight , Diet, High-Fat , Energy Intake , Glucagon-Like Peptide-1 Receptor , Mice, Obese , Obesity , Animals , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/metabolism , Female , Male , Mice , Obesity/drug therapy , Obesity/metabolism , Diet, High-Fat/adverse effects , Energy Intake/drug effects , Body Weight/drug effects , Mice, Knockout , Mice, Inbred C57BL
10.
Lipids Health Dis ; 23(1): 242, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39123208

ABSTRACT

It had been observed that homozygous albumin knockout mice (Alb-/-) exhibit low plasma free fatty acid (FFA) concentration and improved blood glucose regulation. However, it was not yet known to what extent heterozygous albumin knockout (Alb+/-) mice would display a similar phenotype. Alb-/-, Alb+/-, and wild-type (WT) female mice were studied on a low-fat diet (LFD) or high-fat diet (HFD). On both diets, decreased plasma FFA concentration, and improved glucose tolerance test were observed in Alb-/-, but not in Alb+/-, compared to WT. Plasma adiponectin concentration showed greater elevation in Alb-/- than Alb+/-. Consistent with that, adiponectin gene expression was significantly higher in Alb-/- mice than in Alb+/- and WT mice. A dose-dependent response was observed for hepatic Acadl gene expression showing higher Acadl gene expression in Alb-/- mice than in Alb+/- and WT mice. In conclusion, although female Alb+/- mice exhibited some slight differences from WT mice (e.g., increased plasma adiponectin and hepatic Acadl gene expression), Alb+/- mice did not exhibit improved glucoregulation in comparison to WT mice, indicating that a minor suppression of albumin expression is not sufficient to improve glucoregulation. Furthermore, it is now clear that although the response of female mice to HFD might be unique from how males generally respond, still the complete albumin deficiency in Alb-/- mice and the associated FFA reduction is capable of improving glucoregulation in females on this diet. The present results have implications for the role of albumin and FFA in the regulation of metabolism.


Subject(s)
Adiponectin , Albumins , Blood Glucose , Diet, High-Fat , Fatty Acids, Nonesterified , Mice, Knockout , Animals , Female , Adiponectin/genetics , Adiponectin/metabolism , Adiponectin/blood , Mice , Fatty Acids, Nonesterified/blood , Fatty Acids, Nonesterified/metabolism , Diet, High-Fat/adverse effects , Albumins/metabolism , Albumins/genetics , Blood Glucose/metabolism , Liver/metabolism , Diet, Fat-Restricted , Glucose Tolerance Test , Serum Albumin/metabolism , Serum Albumin/genetics , Gene Expression Regulation , Mice, Inbred C57BL
11.
Cell Mol Life Sci ; 81(1): 343, 2024 Aug 12.
Article in English | MEDLINE | ID: mdl-39129011

ABSTRACT

The coordination of food intake, energy storage, and expenditure involves complex interactions between hypothalamic neurons and peripheral tissues including pancreatic islets, adipocytes, muscle, and liver. Previous research shows that deficiency of the transcription factor Alx3 alters pancreatic islet-dependent glucose homeostasis. In this study we carried out a comprehensive assessment of metabolic alterations in Alx3 deficiency. We report that Alx3-deficient mice exhibit decreased food intake without changes in body weight, along with reduced energy expenditure and altered respiratory exchange ratio. Magnetic resonance imaging reveals increased adiposity and decreased muscle mass, which was associated with markers of motor and sympathetic denervation. By contrast, Alx3-deficient mice on a high-fat diet show attenuated weight gain and improved insulin sensitivity, compared to control mice. Gene expression analysis demonstrates altered lipogenic and lipolytic gene profiles. In wild type mice Alx3 is expressed in hypothalamic arcuate nucleus neurons, but not in major peripheral metabolic organs. Functional diffusion-weighted magnetic resonance imaging reveals selective hypothalamic responses to fasting in the arcuate nucleus of Alx3-deficient mice. Additionally, altered expression of proopiomelanocortin and melanocortin-3 receptor mRNA in the hypothalamus suggests impaired regulation of feeding behavior. This study highlights the crucial role for Alx3 in governing food intake, energy homeostasis, and metabolic nutrient partitioning, thereby influencing body mass composition.


Subject(s)
Body Composition , Eating , Energy Metabolism , Homeodomain Proteins , Homeostasis , Hypothalamus , Mice, Knockout , Animals , Energy Metabolism/genetics , Hypothalamus/metabolism , Mice , Eating/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Diet, High-Fat , Transcription Factors/metabolism , Transcription Factors/genetics , Male , Mice, Inbred C57BL , Neurons/metabolism , Pro-Opiomelanocortin/metabolism , Pro-Opiomelanocortin/genetics , Insulin Resistance/genetics , Arcuate Nucleus of Hypothalamus/metabolism
12.
Sci Rep ; 14(1): 18377, 2024 08 08.
Article in English | MEDLINE | ID: mdl-39112593

ABSTRACT

The effects of calcitonin gene-related peptide (CGRP) on atherosclerosis remain unclear. We used apolipoprotein E-deficient (ApoE-/-) mice to generate double-knockout ApoE-/-:CGRP-/- mice lacking alpha CGRP. ApoE-/-:CGRP-/- mice exhibited larger atherosclerotic plaque areas, peritoneal macrophages with enhanced migration functions, and elevated levels of the inflammatory cytokine tumor necrosis factor (TNF)-⍺. Thus, we also explored whether inhibiting TNF-⍺ could improve atherosclerosis in ApoE-/-:CGRP-/- mice by administering etanercept intraperitoneally once a week (5 mg/kg) alongside a high-fat diet for 2 weeks. This treatment led to significant reductions in aortic root lesion size, atherosclerotic plaque area and macrophage migration in ApoE-/-:CGRP-/- mice compared with mice treated with human IgG (5 mg/kg). We further examined whether results observed in ApoE-/-:CGRP-/- mice could similarly be obtained by administering a humanized monoclonal CGRP antibody, galcanezumab, to ApoE-/- mice. ApoE-/- mice were subcutaneously administered galcanezumab at an initial dose of 50 mg/kg, followed by a dose of 30 mg/kg in the second week. Galcanezumab administration did not affect systolic blood pressure, serum lipid levels, or macrophage migration but led to a significant increase in lipid deposition at the aortic root. These findings suggest that alpha CGRP plays a critical role in inhibiting the progression of atherosclerosis.


Subject(s)
Apolipoproteins E , Atherosclerosis , Calcitonin Gene-Related Peptide , Mice, Knockout , Plaque, Atherosclerotic , Animals , Atherosclerosis/metabolism , Atherosclerosis/genetics , Atherosclerosis/pathology , Calcitonin Gene-Related Peptide/metabolism , Mice , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Plaque, Atherosclerotic/pathology , Plaque, Atherosclerotic/metabolism , Plaque, Atherosclerotic/genetics , Diet, High-Fat/adverse effects , Tumor Necrosis Factor-alpha/metabolism , Male , Mice, Knockout, ApoE , Disease Models, Animal , Humans , Antibodies, Monoclonal, Humanized/pharmacology , Etanercept/pharmacology , Mice, Inbred C57BL , Cell Movement/drug effects , Aorta/metabolism , Aorta/pathology , Aorta/drug effects
13.
Biol Pharm Bull ; 47(8): 1429-1436, 2024.
Article in English | MEDLINE | ID: mdl-39135238

ABSTRACT

Farnesoid X receptor (FXR) is a nuclear receptor that regulates the synthesis and enterohepatic circulation of bile acids (BAs). It also regulates lipid and carbohydrate metabolism, making FXR ligands potential therapeutic agents for systemic and/or hepatic metabolic disorders. We previously synthesized a series of FXR antagonists and showed that oral administration of FLG249 reduced the expression of several FXR target genes in the mouse ileum. Here, we investigated the effects of FLG249 on lipid metabolism in mice fed a high-fat diet (HFD). When FLG249 was administered for 4 weeks to HFD-induced obese mice, it altered the expression of genes related to BA metabolism, ceramide synthesis and fatty acid ß-oxidation, improving lipid metabolism in the liver and ileum without decreasing body weight. These findings suggest that FLG249 has the potential to be a low toxicity pharmaceutical compound and likely acts as a nonsteroidal FXR antagonist to improve lipid metabolism disorders.


Subject(s)
Cholesterol , Diet, High-Fat , Liver , Mice, Inbred C57BL , Obesity , Receptors, Cytoplasmic and Nuclear , Triglycerides , Animals , Diet, High-Fat/adverse effects , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/metabolism , Male , Liver/metabolism , Liver/drug effects , Obesity/drug therapy , Obesity/metabolism , Obesity/blood , Cholesterol/blood , Triglycerides/blood , Lipid Metabolism/drug effects , Bile Acids and Salts/metabolism , Mice , Mice, Obese , Ileum/metabolism , Ileum/drug effects
14.
Sci Rep ; 14(1): 18724, 2024 08 12.
Article in English | MEDLINE | ID: mdl-39134597

ABSTRACT

ATP6AP2 knockout in the renal nephron impairs receptor-mediated endocytosis, increasing urinary albumin and glucose excretion and impairing weight gain. Nonesterified fatty acids (NEFA) in urine are bound to albumin and reabsorbed in the proximal tubule through receptor-mediated endocytosis by the megalin-cubilin complex. We hypothesized that ATP6AP2 knockout increases urinary NEFA excretion through a reduction in megalin. Ten-week-old male C57BL/6 mice with nephron specific inducible ATP6AP2 knockout and noninduced controls were fed either normal diet (ND 12% fat) or high fat diet (HFD 45% fat) for 6 months. ATP6AP2 knockout significantly increased urine albumin:creatinine ratio in both ND and HFD fed mice while normalized urine NEFA concentration increased 489% and 259% in ND and HFD knockout mice compared to respective controls. Knockout decreased renal cortical megalin mRNA by 47% on ND and 49% on HFD while megalin protein expression decreased by 36% and 44% respectively. At the same time, markers of mTOR activity were increased while autophagy was impaired. Our results indicate that nephron specific ATP6AP2 knockout increases urinary NEFA excretion in the setting of impaired receptor-mediated endocytosis. Further investigation should determine whether ATP6AP2 contributes to obesity related ectopic lipid deposition in the proximal tubule.


Subject(s)
Low Density Lipoprotein Receptor-Related Protein-2 , Mice, Inbred C57BL , Mice, Knockout , Nephrons , Animals , Low Density Lipoprotein Receptor-Related Protein-2/genetics , Low Density Lipoprotein Receptor-Related Protein-2/metabolism , Male , Mice , Nephrons/metabolism , Diet, High-Fat , Fatty Acids, Nonesterified/metabolism , Kidney Cortex/metabolism , Vacuolar Proton-Translocating ATPases/genetics , Vacuolar Proton-Translocating ATPases/metabolism , Fatty Acids/metabolism , Prorenin Receptor
15.
Cells ; 13(15)2024 Jul 24.
Article in English | MEDLINE | ID: mdl-39120278

ABSTRACT

Sex differences may play a role in the etiopathogenesis and severity of metabolic dysfunction-associated steatotic liver disease (MASLD), a disorder characterized by excessive fat accumulation associated with increased inflammation and oxidative stress. We previously observed the development of steatosis specifically in female rats fed a high-fat diet enriched with liquid fructose (HFHFr) for 12 weeks. The aim of this study was to better characterize the observed sex differences by focusing on the antioxidant and cytoprotective pathways related to the KEAP1/NRF2 axis. The KEAP1/NRF2 signaling pathway, autophagy process (LC3B and LAMP2), and endoplasmic reticulum stress response (XBP1) were analyzed in liver homogenates in male and female rats that were fed a 12-week HFHFr diet. In females, the HFHFr diet resulted in the initial activation of the KEAP1/NRF2 pathway, which was not followed by the modulation of downstream molecular targets; this was possibly due to the increase in KEAP1 levels preventing the nuclear translocation of NRF2 despite its cytosolic increase. Interestingly, while in both sexes the HFHFr diet resulted in an increase in the levels of LC3BII/LC3BI, a marker of autophagosome formation, only males showed a significant upregulation of LAMP2 and XBP1s; this did not occur in females, suggesting impaired autophagic flux in this sex. Overall, our results suggest that males are characterized by a greater ability to cope with an HFHFr metabolic stimulus mainly through an autophagic-mediated proteostatic process while in females, this is impaired. This might depend at least in part upon the fine modulation of the cytoprotective and antioxidant KEAP1/NRF2 pathway resulting in sex differences in the occurrence and severity of MASLD. These results should be considered to design effective therapeutics for MASLD.


Subject(s)
Diet, High-Fat , Fructose , Kelch-Like ECH-Associated Protein 1 , NF-E2-Related Factor 2 , Sex Characteristics , Signal Transduction , Animals , NF-E2-Related Factor 2/metabolism , Female , Male , Diet, High-Fat/adverse effects , Signal Transduction/drug effects , Rats , Kelch-Like ECH-Associated Protein 1/metabolism , Autophagy/drug effects , X-Box Binding Protein 1/metabolism , X-Box Binding Protein 1/genetics , Disease Models, Animal , Fatty Liver/metabolism , Fatty Liver/pathology , Liver/metabolism , Liver/pathology , Liver/drug effects , Endoplasmic Reticulum Stress/drug effects , Rats, Wistar , Oxidative Stress/drug effects , Microtubule-Associated Proteins
16.
Nutrients ; 16(15)2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39125288

ABSTRACT

Young-onset colorectal cancer is an increasing concern worldwide due to the growing prevalence of Westernized lifestyles in childhood and adolescence. Environmental factors during early life, particularly early-life nutrition, significantly contribute to the increasing incidence. Recently, there have been reports of beneficial effects, including anti-inflammation and anti-cancer, of a unique fungus (Antrodia camphorate, AC) native to Taiwan. The objective of this study is to investigate the impact of AC supplementation in early life on the development of young-onset intestinal tumorigenesis. APC1638N mice were fed with a high-fat diet (HF) at 4-12 weeks of age, which is equivalent to human childhood/adolescence, before switching to a normal maintenance diet for an additional 12 weeks up to 24 weeks of age, which is equivalent to young to middle adulthood in humans. Our results showed that the body weight in the HF groups significantly increased after 8 weeks of feeding (p < 0.05). Following a switch to a normal maintenance diet, the change in body weight persisted. AC supplementation significantly suppressed tumor incidence and multiplicity in females (p < 0.05) and reduced IGF-1 and Wnt/ß-catenin signaling (p < 0.05). Moreover, it altered the gut microbiota, suppressed inflammatory responses, and created a microenvironment towards suppressing tumorigenesis later in life.


Subject(s)
Carcinogenesis , Diet, High-Fat , Dietary Supplements , Gastrointestinal Microbiome , Animals , Gastrointestinal Microbiome/drug effects , Female , Mice , Male , Diet, High-Fat/adverse effects , Carcinogenesis/drug effects , Polyporales , Mice, Inbred C57BL , Wnt Signaling Pathway/drug effects , Insulin-Like Growth Factor I/metabolism , Colorectal Neoplasms/prevention & control , Disease Models, Animal , Adenomatous Polyposis Coli Protein/genetics
17.
Nutrients ; 16(15)2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39125385

ABSTRACT

More effective treatments for hepatitis viral infections have led to a reduction in the incidence of liver cirrhosis. A high-fat diet can lead to chronic hepatitis and liver fibrosis, but the effects of lipid intake on liver disease status, including hepatitis C virus and alcohol, after elimination of the cause are unclear. To investigate the effects, we used a rat cirrhosis model and a high-fat diet in this study. Male Wistar rats were administered carbon tetrachloride for 5 weeks. At 12 weeks of age, one group was sacrificed. The remaining rats were divided into four groups according to whether or not they were administered carbon tetrachloride for 5 weeks, and whether they were fed a high-fat diet or control diet. At 12 weeks of age, liver fibrosis became apparent and then improved in the groups where carbon tetrachloride was discontinued, while it worsened in the groups where carbon tetrachloride was continued. Liver fibrosis was notable in both the carbon tetrachloride discontinuation and continuation groups due to the administration of a high-fat diet. In addition, liver precancerous lesions were observed in all groups, and tumor size and multiplicity were higher in the high-fat diet-fed groups. The expression of genes related to inflammation and lipogenesis were upregulated in rats fed a high-fat diet compared to their controls. The results suggest that a high-fat diet worsens liver fibrosis and promotes liver carcinogenesis, presumably through enhanced inflammation and lipogenesis, even after eliminating the underlying cause of liver cirrhosis.


Subject(s)
Carbon Tetrachloride , Diet, High-Fat , Disease Models, Animal , Liver Cirrhosis , Rats, Wistar , Animals , Diet, High-Fat/adverse effects , Male , Liver Cirrhosis/etiology , Rats , Liver/pathology , Liver/metabolism , Liver Neoplasms/pathology , Liver Neoplasms/etiology , Carcinogenesis , Lipogenesis
18.
Nutrients ; 16(15)2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39125449

ABSTRACT

We aimed to characterize the anti-obesity and anti-atherosclerosis effects of Lactobacillus curvatus HY7601 and Lactobacillus plantarum KY1032 using high-fat diet (HFD)-fed obese C57BL/6 mice. We divided the mice into control (CON), HFD, HFD with 108 CFU/kg/day probiotics (HFD + KL, HY7301:KY1032 = 1:1), and HFD with 109 CFU/kg/day probiotics (HFD + KH, HY7301:KY1032 = 1:1) groups and fed/treated them during 7 weeks. The body mass, brown adipose tissue (BAT), inguinal white adipose tissue (iWAT), and epididymal white adipose tissue (eWAT) masses and the total cholesterol and triglyceride concentrations were remarkably lower in probiotic-treated groups than in the HFD group in a dose-dependent manner. In addition, the expression of uncoupling protein 1 in the BAT, iWAT, and eWAT was significantly higher in probiotic-treated HFD mice than in the HFD mice, as demonstrated by immunofluorescence staining and Western blotting. We also measured the expression of cholesterol transport genes in the liver and jejunum and found that the expression of those encoding liver-X-receptor α, ATP-binding cassette transporters G5 and G8, and cholesterol 7α-hydroxylase were significantly higher in the HFD + KH mice than in the HFD mice. Thus, a Lactobacillus HY7601 and KY1032 mixture with 109 CFU/kg/day concentration can assist with body weight regulation through the management of lipid metabolism and thermogenesis.


Subject(s)
Cholesterol , Diet, High-Fat , Energy Metabolism , Lactobacillus , Mice, Inbred C57BL , Probiotics , Animals , Diet, High-Fat/adverse effects , Probiotics/pharmacology , Probiotics/administration & dosage , Cholesterol/metabolism , Cholesterol/blood , Energy Metabolism/drug effects , Male , Mice , Adipose Tissue, Brown/metabolism , Adipose Tissue, Brown/drug effects , Obesity/metabolism , Obesity/microbiology , Adipose Tissue, White/metabolism , Uncoupling Protein 1/metabolism , Uncoupling Protein 1/genetics , Adipose Tissue/metabolism , Liver/metabolism , Lactobacillus plantarum , Jejunum/metabolism , Jejunum/drug effects , Jejunum/microbiology
19.
Int J Mol Sci ; 25(15)2024 Jul 24.
Article in English | MEDLINE | ID: mdl-39125617

ABSTRACT

Progression of metabolic dysfunction-associated steatites liver disease (MASLD) to steatohepatitis (MASH) is driven by stress-inducing lipids that promote liver inflammation and fibrosis, and MASH can lead to cirrhosis and hepatocellular carcinoma. Previously, we showed coordinated defenses regulated by transcription factors, nuclear factor erythroid 2-related factor-1 (Nrf1) and -2 (Nrf2), protect against hepatic lipid stress. Here, we investigated protective effects of hepatocyte Nrf1 and Nrf2 against MASH-linked liver fibrosis and tumorigenesis. Male and female mice with flox alleles for genes encoding Nrf1 (Nfe2l1), Nrf2 (Nfe2l2), or both were fed a MASH-inducing diet enriched with high fat, fructose, and cholesterol (HFFC) or a control diet for 24-52 weeks. During this period, hepatocyte Nrf1, Nrf2, or combined deficiency for ~7 days, ~7 weeks, and ~35 weeks was induced by administering mice hepatocyte-targeting adeno-associated virus (AAV) expressing Cre recombinase. The effects on MASH, markers of liver fibrosis and proliferation, and liver tumorigenesis were compared to control mice receiving AAV-expressing green fluorescent protein. Also, to assess the impact of Nrf1 and Nrf2 induction on liver fibrosis, HFFC diet-fed C57bl/6J mice received weekly injections of carbon tetrachloride, and from week 16 to 24, mice were treated with the Nrf2-activating drug bardoxolone, hepatocyte overexpression of human NRF1 (hNRF1), or both, and these groups were compared to control. Compared to the control diet, 24-week feeding with the HFFC diet increased bodyweight as well as liver weight, steatosis, and inflammation. It also increased hepatocyte proliferation and a marker of liver damage, p62. Hepatocyte Nrf1 and combined deficiency increased liver steatosis in control diet-fed but not HFFC diet-fed mice, and increased liver inflammation under both diet conditions. Hepatocyte Nrf1 deficiency also increased hepatocyte proliferation, whereas combined deficiency did not, and this also occurred for p62 level in control diet-fed conditions. In 52-week HFFC diet-fed mice, 35 weeks of hepatocyte Nrf1 deficiency, but not combined deficiency, resulted in more liver tumors in male mice, but not in female mice. In contrast, hepatocyte Nrf2 deficiency had no effect on any of these parameters. However, in the 15-week CCL4-exposed and 24-week HFFC diet-fed mice, Nrf2 induction with bardoxolone reduced liver steatosis, inflammation, fibrosis, and proliferation. Induction of hepatic Nrf1 activity with hNRF1 enhanced the effect of bardoxolone on steatosis and may have stimulated liver progenitor cells. Physiologic Nrf1 delays MASLD progression, Nrf2 induction alleviates MASH, and combined enhancement synergistically protects against steatosis and may facilitate liver repair.


Subject(s)
Hepatocytes , NF-E2-Related Factor 2 , Animals , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , Mice , Hepatocytes/metabolism , Male , Female , Disease Progression , Mice, Inbred C57BL , Fatty Liver/metabolism , Fatty Liver/pathology , Fatty Liver/genetics , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Liver Cirrhosis/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/pathology , NF-E2-Related Factor 1/metabolism , NF-E2-Related Factor 1/genetics , Nuclear Respiratory Factor 1/metabolism , Nuclear Respiratory Factor 1/genetics , Diet, High-Fat/adverse effects , Liver/metabolism , Liver/pathology , Humans
20.
Int J Mol Sci ; 25(15)2024 Jul 28.
Article in English | MEDLINE | ID: mdl-39125804

ABSTRACT

Obesity is an emerging public health problem. Chronic low-grade inflammation is considered a major promotor of obesity-induced secondary diseases such as cardiovascular and fatty liver disease, type 2 diabetes mellitus, and several cancer entities. Most preliminary studies on obesity-induced immune responses have been conducted in male rodents. Sex-specific differences between men and women in obesity-induced immune dysregulation have not yet been fully outlined but are highly relevant to optimizing prevention strategies for overweight-associated complications. In this study, we fed C57BL/6 female vs. male mice with either standard chow or an obesity-inducing diet (OD). Blood and spleen immune cells were isolated and analyzed by flow cytometry. Lean control mice showed no sex bias in systemic and splenic immune cell composition, whereas the immune responses to obesity were significantly distinct between female and male mice. While immune cell alterations in male OD mice were characterized by a significant reduction in T cells and an increase in myeloid-derived suppressor cells (MDSC), female OD mice displayed preserved T cell numbers. The sex-dependent differences in obesity-induced T cell dysregulation were associated with varying susceptibility to body weight gain and fatty liver disease: Male mice showed significantly more hepatic inflammation and histopathological stigmata of fatty liver in comparison to female OD mice. Our findings indicate that sex impacts susceptibility to obesity-induced T cell dysregulation, which might explain sex-dependent different incidences in the development of obesity-associated secondary diseases. These results provide novel insights into the understanding of obesity-induced chronic inflammation from a sex-specific perspective. Given that most nutrition, exercise, and therapeutic recommendations for the prevention of obesity-associated comorbidities do not differentiate between men and women, the data of this study are clinically relevant and should be taken into consideration in future trials and treatment strategies.


Subject(s)
Mice, Inbred C57BL , Obesity , T-Lymphocytes , Animals , Obesity/immunology , Obesity/complications , Obesity/etiology , Female , Male , Mice , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Diet, High-Fat/adverse effects , Sex Factors , Spleen/immunology , Spleen/pathology , Sex Characteristics , Myeloid-Derived Suppressor Cells/immunology , Myeloid-Derived Suppressor Cells/metabolism , Fatty Liver/etiology , Fatty Liver/immunology , Fatty Liver/pathology , Inflammation/immunology , Inflammation/pathology , Inflammation/etiology
SELECTION OF CITATIONS
SEARCH DETAIL