Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25.439
Filter
1.
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
2.
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
3.
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
4.
Signal Transduct Target Ther ; 9(1): 214, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39117631

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) is a serious threat to public health, but its underlying mechanism remains poorly understood. In screening important genes using Gene Importance Calculator (GIC) we developed previously, ribosomal modification protein rimK-like family member A (RIMKLA) was predicted as one essential gene but its functions remained largely unknown. The current study determined the roles of RIMKLA in regulating glucose and lipid metabolism. RIMKLA expression was reduced in livers of human and mouse with NAFLD. Hepatic RIMKLA overexpression ameliorated steatosis and hyperglycemia in obese mice. Hepatocyte-specific RIMKLA knockout aggravated high-fat diet (HFD)-induced dysregulated glucose/lipid metabolism in mice. Mechanistically, RIMKLA is a new protein kinase that phosphorylates betaine-homocysteine S-methyltransferase 1 (BHMT1) at threonine 45 (Thr45) site. Upon phosphorylation at Thr45 and activation, BHMT1 eliminated homocysteine (Hcy) to inhibit the activity of transcription factor activator protein 1 (AP1) and its induction on fatty acid synthase (FASn) and cluster of differentiation 36 (CD36) gene transcriptions, concurrently repressing lipid synthesis and uptake in hepatocytes. Thr45 to alanine (T45A) mutation inactivated BHMT1 to abolish RIMKLA's repression on Hcy level, AP1 activity, FASn/CD36 expressions, and lipid deposition. BHMT1 overexpression rescued the dysregulated lipid metabolism in RIMKLA-deficient hepatocytes. In summary, RIMKLA is a novel protein kinase that phosphorylates BHMT1 at Thr45 to repress lipid synthesis and uptake. Under obese condition, inhibition of RIMKLA impairs BHMT1 activity to promote hepatic lipid deposition.


Subject(s)
Betaine-Homocysteine S-Methyltransferase , Lipid Metabolism , Non-alcoholic Fatty Liver Disease , Animals , Mice , Humans , Betaine-Homocysteine S-Methyltransferase/genetics , Betaine-Homocysteine S-Methyltransferase/metabolism , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Lipid Metabolism/genetics , Diet, High-Fat/adverse effects , Hepatocytes/metabolism , Male , Mice, Knockout , Phosphorylation/genetics
5.
Nat Commun ; 15(1): 6768, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39117652

ABSTRACT

Light is fundamental for biological life, with most mammals possessing light-sensing photoreceptors in various organs. Opsin3 is highly expressed in adipose tissue which has extensive communication with other organs, particularly with the brain through the sympathetic nervous system (SNS). Our study reveals a new light-triggered crosstalk between adipose tissue and the hypothalamus. Direct blue-light exposure to subcutaneous white fat improves high-fat diet-induced metabolic abnormalities in an Opsin3-dependent manner. Metabolomic analysis shows that blue light increases circulating levels of histidine, which activates histaminergic neurons in the hypothalamus and stimulates brown adipose tissue (BAT) via SNS. Blocking central actions of histidine and denervating peripheral BAT blunts the effects of blue light. Human white adipocytes respond to direct blue light stimulation in a cell-autonomous manner, highlighting the translational relevance of this pathway. Together, these data demonstrate a light-responsive metabolic circuit involving adipose-hypothalamus communication, offering a potential strategy to alleviate obesity-induced metabolic abnormalities.


Subject(s)
Adipose Tissue, Brown , Hypothalamus , Light , Animals , Hypothalamus/metabolism , Hypothalamus/radiation effects , Humans , Adipose Tissue, Brown/metabolism , Male , Mice , Obesity/metabolism , Mice, Inbred C57BL , Diet, High-Fat/adverse effects , Rod Opsins/metabolism , Sympathetic Nervous System/metabolism , Adipose Tissue/metabolism , Neurons/metabolism , Neurons/radiation effects , Adipose Tissue, White/metabolism , Adipose Tissue, White/radiation effects , Adipocytes, White/metabolism , Adipocytes, White/radiation effects
6.
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
7.
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
8.
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
9.
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
10.
Mol Med ; 30(1): 113, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39095693

ABSTRACT

BACKGROUND: To explore whether nobiletin has a protective effect on high-fat diet (HFD)-induced enteric nerve injury and its underlying mechanism. METHODS: An obesity model was induced by a HFD. Nobiletin (100 mg/kg and 200 mg/kg) and vehicle were administered by gastric gavage for 4 weeks. Lee's index, body weight, OGTT and intestinal propulsion assays were performed before sacrifice. After sampling, lipids were detected using Bodipy 493/503; lipid peroxidation was detected using MDA and SOD kits and the expression of PGP 9.5, Trem2, GFAP, ß-tubulin 3, Bax, Bcl2, Nestin, P75 NTR, SOX10 and EDU was detected using immunofluorescence. The GDNF, p-AKT, AKT, p-FOXO3a, FOXO3a and P21 proteins were detected using western blotting. The relative mRNA expression levels of NOS2 were detected via qPCR. Primary enteric neural stem cells (ENSCs) were cultured. After ENSCs were treated with palmitic acid (PA) and nobiletin, CCK-8 and caspase-3/7 activity assays were performed to evaluate proliferation and apoptosis. RESULTS: HFD consumption caused colon lipid accumulation and peroxidation, induced enteric nerve damage and caused intestinal motor dysfunction. However, nobiletin reduced lipid accumulation and peroxidation in the colon; promoted Trem2, ß-tubulin 3, Nestin, P75NTR, SOX10 and Bcl2 expression; inhibited Bax and GFAP expression; reduced NOS2 mRNA transcription; and regulated the GDNF/AKT/FOXO3a/P21 pathway. Nobiletin also promoted PA-induced impairment of ENSCs. CONCLUSIONS: Nobiletin restored HFD-induced enteric nerve injury, which may be associated with inhibiting enteric nerve apoptosis, promoting enteric nerve survival and regulating the GDNF/AKT/FOXO3a/P21 pathway.


Subject(s)
Diet, High-Fat , Enteric Nervous System , Flavones , Forkhead Box Protein O3 , Glial Cell Line-Derived Neurotrophic Factor , Proto-Oncogene Proteins c-akt , Signal Transduction , Animals , Forkhead Box Protein O3/metabolism , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Diet, High-Fat/adverse effects , Signal Transduction/drug effects , Male , Flavones/pharmacology , Flavones/therapeutic use , Enteric Nervous System/metabolism , Enteric Nervous System/drug effects , Neuroglia/metabolism , Neuroglia/drug effects , Mice , Disease Models, Animal , Rats , Obesity/metabolism , Obesity/drug therapy , Apoptosis/drug effects
11.
Sci Prog ; 107(3): 368504241269431, 2024.
Article in English | MEDLINE | ID: mdl-39090965

ABSTRACT

Pork is one type of the most frequently consumed meat with about 30% globally. Thus, the questions regarding to the health effects of diet with high fat content from lard are raised. Here, we developed a model of mice fed with high fat (HF) from lard to investigate and have more insights on the effects of long-time feeding with HF on health. The results showed that 66 days on HF induced a significant gain in the body weight of mice, and this weight gain was associated to the deposits in the white fat, but not brown fat. The glucose tolerance, not insulin resistance, in mice was decreased by the HF diet, and this was accompanied with significantly higher blood levels of total cholesterol and triglycerides. Furthermore, the weight gains in mice fed with HF seemed to link to increased mRNA levels of adipose biomarkers in lipogenesis, including Acly and Acaca genes, in white fat tissues. Thus, our study shows that a diet with high fat from lard induced the increase in body weight, white fat depots' expansion, disruption of glucose tolerance, blood dyslipidemia, and seemed to start affecting the mRNA expression of some adipose biomarkers in a murine model.


Subject(s)
Biomarkers , Diet, High-Fat , Dietary Fats , RNA, Messenger , Animals , Mice , Diet, High-Fat/adverse effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Biomarkers/metabolism , Biomarkers/blood , Male , Dietary Fats/metabolism , Insulin Resistance , Adipose Tissue/metabolism , Body Weight , Mice, Inbred C57BL , Weight Gain , Adipose Tissue, White/metabolism , Triglycerides/blood , Triglycerides/metabolism
12.
J Neuroinflammation ; 21(1): 191, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39095788

ABSTRACT

OBJECTIVE: Obesity represents a significant global health challenge characterized by chronic low-grade inflammation and metabolic dysregulation. The hypothalamus, a key regulator of energy homeostasis, is particularly susceptible to obesity's deleterious effects. This study investigated the role of the immunoproteasome, a specialized proteasomal complex implicated in inflammation and cellular homeostasis, during metabolic diseases. METHODS: The levels of the immunoproteasome ß5i subunit were analyzed by immunostaining, western blotting, and proteasome activity assay in mice fed with either a high-fat diet (HFD) or a regular diet (CHOW). We also characterized the impact of autophagy inhibition on the levels of the immunoproteasome ß5i subunit and the activation of the AKT pathway. Finally, through confocal microscopy, we analyzed the contribution of ß5i subunit inhibition on mitochondrial function by flow cytometry and mitophagy assay. RESULTS: Using an HFD-fed obese mouse model, we found increased immunoproteasome levels in hypothalamic POMC neurons. Furthermore, we observed that palmitic acid (PA), a major component of saturated fats found in HFD, increased the levels of the ß5i subunit of the immunoproteasome in hypothalamic neuronal cells. Notably, the increase in immunoproteasome expression was associated with decreased autophagy, a critical cellular process in maintaining homeostasis and suppressing inflammation. Functionally, PA disrupted the insulin-glucose axis, leading to reduced AKT phosphorylation and increased intracellular glucose levels in response to insulin due to the upregulation of the immunoproteasome. Mechanistically, we identified that the protein PTEN, a key regulator of insulin signaling, was reduced in an immunoproteasome-dependent manner. To further investigate the potential therapeutic implications of these findings, we used ONX-0914, a specific immunoproteasome inhibitor. We demonstrated that this inhibitor prevents PA-induced insulin-glucose axis imbalance. Given the interplay between mitochondrial dysfunction and metabolic disturbances, we explored the impact of ONX-0914 on mitochondrial function. Notably, ONX-0914 preserved mitochondrial membrane potential and attenuated mitochondrial ROS production in the presence of PA. Moreover, we found that ONX-0914 reduced mitophagy in the presence of PA. CONCLUSIONS: Our findings strongly support the pathogenic involvement of the immunoproteasome in hypothalamic neurons in the context of HFD-induced obesity and metabolic disturbances. Targeting the immunoproteasome highlights a promising therapeutic strategy to mitigate the detrimental effects of obesity on the insulin-glucose axis and cellular homeostasis. This study provides valuable insights into the mechanisms driving obesity-related metabolic diseases and offers potential avenues for developing novel therapeutic interventions.


Subject(s)
Diet, High-Fat , Hypothalamus , Mice, Inbred C57BL , Neurons , Obesity , Proteasome Endopeptidase Complex , Animals , Diet, High-Fat/adverse effects , Mice , Hypothalamus/metabolism , Obesity/metabolism , Neurons/metabolism , Neurons/drug effects , Proteasome Endopeptidase Complex/metabolism , Male , Metabolic Diseases/metabolism , Metabolic Diseases/etiology , Oligopeptides
13.
Nat Commun ; 15(1): 6585, 2024 Aug 03.
Article in English | MEDLINE | ID: mdl-39097566

ABSTRACT

In type 1 diabetes, high-fat meals require more insulin to prevent hyperglycemia while meals followed by aerobic exercises require less insulin to prevent hypoglycemia, but the adjustments needed vary between individuals. We propose a decision support system with reinforcement learning to personalize insulin doses for high-fat meals and postprandial aerobic exercises. We test this system in a single-arm 16-week study in 15 adults on multiple daily injections therapy (NCT05041621). The primary objective of this study is to assess the feasibility of the novel learning algorithm. This study looks at glucose outcomes and patient reported outcomes. The postprandial incremental area under the glucose curve is improved from the baseline to the evaluation period for high-fat meals (378 ± 222 vs 38 ± 223 mmol/L/min, p = 0.03) and meals followed by exercises (-395 ± 192 vs 132 ± 181 mmol/L/min, p = 0.007). The postprandial time spent below 3.9 mmol/L is reduced after high-fat meals (5.3 ± 1.6 vs 1.8 ± 1.5%, p = 0.003) and meals followed by exercises (5.3 ± 1.2 vs 1.4 ± 1.1%, p = 0.003). Our study shows the feasibility of automatically personalizing insulin doses for high-fat meals and postprandial exercises. Randomized controlled trials are warranted.


Subject(s)
Blood Glucose , Diabetes Mellitus, Type 1 , Exercise , Insulin , Meals , Postprandial Period , Humans , Diabetes Mellitus, Type 1/therapy , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/blood , Insulin/administration & dosage , Male , Female , Adult , Exercise/physiology , Blood Glucose/metabolism , Proof of Concept Study , Middle Aged , Hypoglycemic Agents/administration & dosage , Diet, High-Fat/adverse effects , Reinforcement, Psychology , Precision Medicine/methods , Hypoglycemia/prevention & control , Algorithms , Young Adult
14.
Nat Commun ; 15(1): 6506, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39090079

ABSTRACT

The lack of an appropriate preclinical model of metabolic dysfunction-associated steatotic liver disease (MASLD) that recapitulates the whole disease spectrum impedes exploration of disease pathophysiology and the development of effective treatment strategies. Here, we develop a mouse model (Streptozotocin with high-fat diet, STZ + HFD) that gradually develops fatty liver, metabolic dysfunction-associated steatohepatitis (MASH), hepatic fibrosis, and hepatocellular carcinoma (HCC) in the context of metabolic dysfunction. The hepatic transcriptomic features of STZ + HFD mice closely reflect those of patients with obesity accompanying type 2 diabetes mellitus, MASH, and MASLD-related HCC. Dietary changes and tirzepatide administration alleviate MASH, hepatic fibrosis, and hepatic tumorigenesis in STZ + HFD mice. In conclusion, a murine model recapitulating the main histopathologic, transcriptomic, and metabolic alterations observed in MASLD patients is successfully established.


Subject(s)
Carcinoma, Hepatocellular , Diet, High-Fat , Disease Models, Animal , Liver Neoplasms , Animals , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/genetics , Male , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Liver Neoplasms/genetics , Mice , Diet, High-Fat/adverse effects , Mice, Inbred C57BL , Humans , Liver/metabolism , Liver/pathology , Fatty Liver/metabolism , Fatty Liver/pathology , Streptozocin , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Transcriptome , Obesity/metabolism , Obesity/complications , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Non-alcoholic Fatty Liver Disease/complications
15.
Front Endocrinol (Lausanne) ; 15: 1440070, 2024.
Article in English | MEDLINE | ID: mdl-39145314

ABSTRACT

Previous studies indicate that CNS administration of oxytocin (OT) reduces body weight in high fat diet-induced obese (DIO) rodents by reducing food intake and increasing energy expenditure (EE). We recently demonstrated that hindbrain (fourth ventricular [4V]) administration of OT elicits weight loss and elevates interscapular brown adipose tissue temperature (TIBAT, a surrogate measure of increased EE) in DIO mice. What remains unclear is whether OT-elicited weight loss requires increased sympathetic nervous system (SNS) outflow to IBAT. We hypothesized that OT-induced stimulation of SNS outflow to IBAT contributes to its ability to activate BAT and elicit weight loss in DIO mice. To test this hypothesis, we determined the effect of disrupting SNS activation of IBAT on the ability of 4V OT administration to increase TIBAT and elicit weight loss in DIO mice. We first determined whether bilateral surgical SNS denervation to IBAT was successful as noted by ≥ 60% reduction in IBAT norepinephrine (NE) content in DIO mice. NE content was selectively reduced in IBAT at 1-, 6- and 7-weeks post-denervation by 95.9 ± 2.0, 77.4 ± 12.7 and 93.6 ± 4.6% (P<0.05), respectively and was unchanged in inguinal white adipose tissue, pancreas or liver. We subsequently measured the effects of acute 4V OT (1, 5 µg ≈ 0.99, 4.96 nmol) on TIBAT in DIO mice following sham or bilateral surgical SNS denervation to IBAT. We found that the high dose of 4V OT (5 µg ≈ 4.96 nmol) elevated TIBAT similarly in sham mice as in denervated mice. We subsequently measured the effects of chronic 4V OT (16 nmol/day over 29 days) or vehicle infusions on body weight, adiposity and food intake in DIO mice following sham or bilateral surgical denervation of IBAT. Chronic 4V OT reduced body weight by 5.7 ± 2.23% and 6.6 ± 1.4% in sham and denervated mice (P<0.05), respectively, and this effect was similar between groups (P=NS). OT produced corresponding reductions in whole body fat mass (P<0.05). Together, these findings support the hypothesis that sympathetic innervation of IBAT is not necessary for OT-elicited increases in BAT thermogenesis and reductions of body weight and adiposity in male DIO mice.


Subject(s)
Adipose Tissue, Brown , Adiposity , Diet, High-Fat , Mice, Inbred C57BL , Obesity , Oxytocin , Sympathetic Nervous System , Animals , Oxytocin/pharmacology , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/metabolism , Adipose Tissue, Brown/innervation , Male , Mice , Obesity/metabolism , Sympathetic Nervous System/drug effects , Diet, High-Fat/adverse effects , Adiposity/drug effects , Body Weight/drug effects , Weight Loss/drug effects , Mice, Obese , Energy Metabolism/drug effects , Norepinephrine/metabolism
16.
Article in English | MEDLINE | ID: mdl-39147447

ABSTRACT

Obesity is a well-known risk factor for testicular function; however, dulaglutide's effect on the testis in obesity has received little attention. Currently, clinicians prescribe the antidiabetic drug dulaglutide only off-label for weight management in non-diabetics. Investigating the impact of this novel compound on obesity is critical for determining whether it has any disruptive effects on testicular cells. We used a well-known animal model of high-fat diet-induced obesity in this investigation, and testicular dysfunction was determined by sperm DNA damage, spermatocyte chromosomal abnormalities, and spermiogram analysis. Following a 12-week high-fat diet challenge, mice were randomly assigned to dulaglutide (0.6 mg/kg/day) or saline treatments for five weeks. Testes and sperm cells were collected 24 h after the last dulaglutide injection. Untreated obese mice had a lower testes/body weight ratio, more sperm DNA damage, diakinesis-metaphase I chromosomal abnormalities, a lower sperm count/motility, more cell morphological defects, and an altered testicular redox balance. In obese mice, dulaglutide injection efficiently restored all disturbed parameters to their control levels. Dulaglutide injection into healthy mice exhibited no significant harmful effects at the applied regimen. As a result, we infer that dulaglutide therapy might bring obese men additional benefits by recovering testicular dysfunction induced by obesity.


Subject(s)
Diet, High-Fat , Disease Models, Animal , Glucagon-Like Peptides , Immunoglobulin Fc Fragments , Obesity , Recombinant Fusion Proteins , Testis , Animals , Male , Immunoglobulin Fc Fragments/pharmacology , Obesity/drug therapy , Glucagon-Like Peptides/analogs & derivatives , Glucagon-Like Peptides/pharmacology , Glucagon-Like Peptides/therapeutic use , Diet, High-Fat/adverse effects , Mice , Recombinant Fusion Proteins/pharmacology , Testis/drug effects , Testis/pathology , Testis/metabolism , DNA Damage/drug effects , Spermatozoa/drug effects , Hypoglycemic Agents/pharmacology , Sperm Motility/drug effects , Mice, Inbred C57BL , Chromosome Aberrations/drug effects , Testicular Diseases/drug therapy
17.
J Biochem Mol Toxicol ; 38(9): e23809, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39148263

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) is an alarming ailment that leads to severe liver damage and increases the risk of serious health conditions. The prevalence of NAFLD due to oxidative stress could be mitigated by plant-derived antioxidants. This study aims to investigate the effects of syringic acid (SA) on NAFLD in a high-fat diet (HFD) rat model. Twenty-four rats were randomly divided into four groups (n = 6): normal control, HFD, SA-administered HFD, and positive control SA on a normal diet. Rats in the normal control and positive control groups received a normal diet, and the remaining groups received an HFD for 8 weeks. SA (20 mg/kg b.w.) was orally (gavage) administered for 8 weeks. Lipid profiles were controlled by SA against HFD-fed rats (p < 0.05). SA reduced the serum aspartate aminotransferase and alanine aminotransferase levels by 70%-190%. SA also suppressed pro-inflammatory cytokines and attenuated histopathological and immunohistochemical changes against HFD-fed rats. SA reversed oxidative stress by suppressing the malondialdehyde formation by 82% and replenished the nonenzymatic and enzymatic antioxidant activities (p < 0.05). Gene expressions of nuclear factor-erythroid 2-related factor/heme oxygenase 1 (Nrf2/HO-1) were elevated in SA-treated rats. Ameliorative effects of SA on NAFLD induced by an HFD in rats were prominent through the reversal of oxidative stress and inflammation, regulated by an intrinsic mechanism of defense against oxidative stress, the Nrf2/HO-1 pathway.


Subject(s)
Gallic Acid , Heme Oxygenase (Decyclizing) , NF-E2-Related Factor 2 , Non-alcoholic Fatty Liver Disease , Signal Transduction , Animals , NF-E2-Related Factor 2/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/pathology , Non-alcoholic Fatty Liver Disease/prevention & control , Rats , Male , Signal Transduction/drug effects , Gallic Acid/analogs & derivatives , Gallic Acid/pharmacology , Heme Oxygenase (Decyclizing)/metabolism , Oxidative Stress/drug effects , Heme Oxygenase-1/metabolism , Diet, High-Fat/adverse effects , Rats, Sprague-Dawley , Antioxidants/pharmacology , Liver/metabolism , Liver/drug effects , Liver/pathology
18.
PeerJ ; 12: e17891, 2024.
Article in English | MEDLINE | ID: mdl-39148685

ABSTRACT

Objective: This study aims to investigate the effects of semaglutide on gut microbiota, cognitive function, and inflammation in obese mice. Method: Twenty-four C57BL/6J male mice were randomly assigned to three groups: a normal-chow diet group (NCD, n = 8), high-fat diet group (HFD, n = 8), and HFD+semaglutide group (Sema, n = 8). The mice were fed a HFD to establish an animal model of obesity and then administered with semaglutide or saline for 12 weeks. Cognitive function was assessed using the Morris water maze test. Serum pro-inflammatory cytokines were measured. 16S rRNA gene sequencing technology was used to explore gut microbiota characteristics in obese mice. Result: Obese mice showed significant cognitive impairment and inflammation. Semaglutide improved cognitive function and attenuated inflammation induced by a HFD diet. The abundance of gut microbiota was significantly changed in the HFD group, including decreased Akkermansia, Muribaculaceae, Coriobacteriaceae_UCG_002, Clostridia_UCG_014 and increased Romboutsia, Dubosiella, Enterorhabdus. Whereas semaglutide could dramatically reverse the relative abundance of these gut microbiota. Correlation analysis suggested that cognitive function was positively correlated with Muribaculaceae and Clostridia_UCG_014, and negatively associated with Romboutsia and Dubosiella. Romboutsia was positively correlated with TNFα, IL-6 and IL-1ß. While Clostridia_UCG_014 was negatively related to TNFα, IL-6 and IL-1ß. Conclusions: For the first time semaglutide displayed different regulatory effects on HFD-induced gut microbiota dysbiosis. Semaglutide could regulate the structure and composition of gut microbiota associated with cognitive function and inflammation. Thus, affecting gut microbiota might be a potential mechanism of semaglutide in attenuating cognitive function and inflammation.


Subject(s)
Cognition , Diet, High-Fat , Gastrointestinal Microbiome , Glucagon-Like Peptides , Inflammation , Mice, Inbred C57BL , Obesity , Animals , Gastrointestinal Microbiome/drug effects , Glucagon-Like Peptides/pharmacology , Mice , Male , Obesity/microbiology , Obesity/drug therapy , Inflammation/drug therapy , Cognition/drug effects , Diet, High-Fat/adverse effects , Mice, Obese , Disease Models, Animal , Cytokines/metabolism , Cytokines/blood , Cognitive Dysfunction/drug therapy
19.
J Agric Food Chem ; 72(32): 18013-18026, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39088205

ABSTRACT

Glucose and lipid metabolism dysregulation in skeletal muscle contributes to the development of metabolic disorders. The efficacy of fucoxanthin in alleviating lipid metabolic disorders in skeletal muscle remains poorly understood. In this study, we systematically investigated the impact of fucoxanthin on mitigating lipid deposition and insulin resistance in skeletal muscle employing palmitic acid-induced lipid deposition in C2C12 cells and ob/ob mice. Fucoxanthin significantly alleviated PA-induced skeletal muscle lipid deposition and insulin resistance. In addition, fucoxanthin prominently upregulated the expression of lipid metabolism-related genes (Pparα and Cpt-1), promoting fatty acid ß-oxidation metabolism. Additionally, fucoxanthin significantly increased the expression of Pgc-1α and Tfam, elevated the mtDNA/nDNA ratio, and reduced ROS levels. Further, we identified pyruvate kinase muscle isozyme 1 (PKM1) as a high-affinity protein for fucoxanthin by drug affinity-responsive target stability and LC-MS and confirmed their robust interaction by CETSA, microscale thermophoresis, and circular dichroism. Supplementation with pyruvate, the product of PKM1, significantly attenuated the beneficial effects of fucoxanthin on lipid deposition and insulin resistance. Mechanistically, fucoxanthin reduced glucose glycolysis rate and enhanced mitochondrial biosynthesis and fatty acid ß-oxidation through inhibiting PKM1 activity, thereby alleviating lipid metabolic stress. These findings present a novel clinical strategy for treating metabolic diseases using fucoxanthin.


Subject(s)
Insulin Resistance , Lipid Metabolism , Muscle, Skeletal , Pyruvate Kinase , Xanthophylls , Animals , Mice , Muscle, Skeletal/metabolism , Muscle, Skeletal/drug effects , Xanthophylls/pharmacology , Lipid Metabolism/drug effects , Pyruvate Kinase/metabolism , Pyruvate Kinase/genetics , Male , Humans , Mice, Inbred C57BL , Diet, High-Fat/adverse effects
20.
Zhonghua Zhong Liu Za Zhi ; 46(8): 737-745, 2024 Aug 23.
Article in Chinese | MEDLINE | ID: mdl-39143796

ABSTRACT

Objective: To investigate the effect of high-fat and low-carbohydrate diet combined with radiotherapy on the tumor microenvironment of mice with lung xenografts. Methods: C57BL/6J mice were selected to establish the Lewis lung cancer model, and they were divided into the normal diet group, the high-fat and low-carbohydrate diet group, the normal diet + radiotherapy group, and the high-fat and low-carbohydrate diet + radiotherapy group, with 18 mice in each group. The mice in the normal diet group and the normal diet + radiotherapy group were fed with the normal diet with 12.11% fat for energy supply, and the mice in the high-fat and low-carbohydrate diet group and the high-fat and low-carbohydrate diet + radiotherapy group were fed with high-fat and low-carbohydratediet with 45.00% fat for energy. On the 12th to 14th days, the tumor sites of the mice in the normal diet + radiotherapy group and the high-fat and low-carbohydrate diet + radiotherapy group were treated with radiotherapy, and the irradiation dose was 24 Gy/3f. The body weight, tumor volume, blood glucose and blood ketone level, liver and kidney function, and survival status of the mice were observed and monitored. Immunohistochemical staining was used to detect the tumor-associated microangiogenesis molecule (CD34) and lymphatic endothelial hyaluronan receptor 1 (LYVE-1), Sirius staining was used to detect collagen fibers, and multiplex immunofluorescence was used to detect CD8 and programmed death-1 (PD-1). Expression of immune cell phenotypes (CD3, CD4, CD8, and Treg) was detected by flow cytometry. Results: On the 27th day after inoculation, the body weigh of the common diet group was(24.78±2.22)g, which was significantly higher than that of the common diet + radiotherapy group [(22.15±0.48)g, P=0.030] and high-fat low-carbohydrate diet + radiotherapy group [(22.02±0.77)g, P=0.031)]. On the 15th day after inoculation, the tumor volume of the high-fat and low-carbohydrate diet + radiotherapy group was (220.88±130.05) mm3, which was significantly smaller than that of the normal diet group [(504.37±328.48) mm3, P=0.042)] and the high-fat, low-carbohydrate diet group [(534.26±230.42) mm3, P=0.016], but there was no statistically significant difference compared with the normal diet + radiotherapy group [(274.64±160.97) mm3]. In the 4th week, the blood glucose values of the mice in the high-fat and low-carbohydrate diet group were lower than those in the normal diet group, with the value being (8.00±0.36) mmol/L and (9.57±0.40) mmol/L, respectively, and the difference was statistically significant (P<0.05). The blood ketone values of the mice in the high-fat and low-carbohydrate diet group were higher than those in the normal diet group, with the value being (1.00±0.20) mmol/L and (0.63±0.06) mmol/L, respectively, in the second week. In the third week, the blood ketone values of the two groups of mice were (0.90±0.17) mmol/L and (0.70±0.10) mmol/L, respectively, and the difference was statistically significant (P<0.05). On the 30th day after inoculation, there were no significant differences in aspartate aminotransferase, alanine aminotransferase, creatinine, and urea between the normal diet group and the high-fat, low-carbohydrate diet group (all P>0.05). The hearts, livers, spleens, lungs, and kidneys of the mice in each group had no obvious toxic changes and tumor metastasis. In the high-fat and low-carbohydrate diet + radiotherapy group, the expression of CD8 was up-regulated in the tumor tissues of mice, and the expressions of PD-1, CD34, LYVE-1, and collagen fibers were down-regulated. The proportion of CD8+ T cells in the paratumoral lymph nodes of the high-fat and low-carbohydrate diet + radiotherapy group was (25.13±0.97)%, higher than that of the normal diet group [(20.60±2.23)%, P<0.050] and the normal diet + radiotherapy group [(19.26±3.07)%, P<0.05], but there was no statistically significant difference with the high-fat and low-carbohydrate diet group [(22.03±1.75)%, P>0.05]. The proportion, of CD4+ T cells in the lymph nodes adjacent to the tumor in the normal diet + radiotherapy group (31.33±5.16)% and the high-fat and low-carbohydrate diet + radiotherapy group (30.63±1.70)% were higher than that in the normal diet group [(20.27±2.15)%, P<0.05] and the high-fat and low-carbohydrate diet group (23.70±2.62, P<0.05). Treg cells accounted for the highest (16.58±5.10)% of T cells in the para-tumor lymph nodes of the normal diet + radiotherapy group, but compared with the normal diet group, the high-fat and low-carbohydrate diet group, and the high-fat and low-carbohydrate diet + radiotherapy group, there was no statistically significant difference (all P>0.05). Conclusion: High-fat and low-carbohydrate diet plus radiotherapy can enhance the recruitment and function of immune effector cells in the tumor microenvironment, inhibit tumor microangiogenesis, and thus inhibit tumor growth.


Subject(s)
Carcinoma, Lewis Lung , Diet, Carbohydrate-Restricted , Diet, High-Fat , Mice, Inbred C57BL , Tumor Microenvironment , Animals , Carcinoma, Lewis Lung/radiotherapy , Carcinoma, Lewis Lung/metabolism , Mice , Diet, High-Fat/adverse effects , Diet, Carbohydrate-Restricted/methods , Tumor Burden , Lung Neoplasms/radiotherapy , Lung Neoplasms/metabolism , Lung Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL