Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 12.807
Filter
1.
FASEB J ; 38(10): e23664, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38775797

ABSTRACT

Adipogenesis, a pivotal cellular process involving the differentiation of mesenchymal stem cells (MSCs) to mature adipocytes, plays a significant role in various physiological functions. Dysregulation of adipogenesis is implicated in conditions such as obesity. However, the complete molecular understanding of adipogenesis remains elusive. This study aimed to uncover the novel role of lamina-associated polypeptide 2 alpha (LAP2α) in human adipose-derived stem cells (hASCs) adipogenesis and its impact on high-fat diet (HFD)-induced obesity and associated metabolic disturbances. LAP2α expression was assessed during the adipogenic differentiation of hASCs using RT-qPCR and western blotting. The functional role of LAP2α in adipogenesis was explored both in vitro and in vivo through loss- and gain-of-function studies. Moreover, mice with HFD-induced obesity received lentivirus injection to assess the effect of LAP2α knockdown on fat accumulation. Molecular mechanisms underlying LAP2α in adipogenic differentiation were investigated using RT-qPCR, Western blotting, immunofluorescence staining, and Oil Red O staining. LAP2α expression was upregulated during hASCs adipogenic differentiation. LAP2α knockdown hindered adipogenesis, while LAP2α overexpression promoted adipogenic differentiation. Notably, LAP2α deficiency resisted HFD-induced obesity, improved glucose intolerance, mitigated insulin resistance, and prevented fatty liver development. Mechanistically, LAP2α knockdown attenuated signal transducer and activator of transcription 3 (STAT3) activation by reducing the protein level of phosphorylated STAT3. A STAT3 activator (Colivelin) counteracted the negative impact of LAP2α deficiency on hASCs adipogenic differentiation. Taken together, our current study established LAP2α as a crucial regulator of hASCs adipogenic differentiation, unveiling a new therapeutic target for obesity prevention.


Subject(s)
Adipogenesis , Diet, High-Fat , Mesenchymal Stem Cells , Obesity , Humans , Diet, High-Fat/adverse effects , Obesity/metabolism , Obesity/genetics , Obesity/etiology , Animals , Mice , Mesenchymal Stem Cells/metabolism , Male , Cell Differentiation , Mice, Inbred C57BL , Adipose Tissue/metabolism , Adipose Tissue/cytology , Adipocytes/metabolism , Cells, Cultured , Gene Knockdown Techniques , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , DNA-Binding Proteins , Membrane Proteins
2.
JCI Insight ; 9(9)2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38716728

ABSTRACT

The importance of the proper localization of most receptors at the cell surface is often underestimated, although this feature is essential for optimal receptor response. Endospanin 1 (Endo1) (also known as OBRGRP or LEPROT) is a protein generated from the same gene as the human leptin receptor and regulates the trafficking of proteins to the surface, including the leptin receptor. The systemic role of Endo1 on whole-body metabolism has not been studied so far. Here, we report that general Endo1-KO mice fed a high-fat diet develop metabolically healthy obesity with lipid repartitioning in organs and preferential accumulation of fat in adipose tissue, limited systematic inflammation, and better controlled glucose homeostasis. Mechanistically, Endo1 interacts with the lipid translocase CD36, thus regulating its surface abundance and lipid uptake in adipocytes. In humans, the level of Endo1 transcripts is increased in the adipose tissue of patients with obesity, but low levels rather correlate with a profile of metabolically healthy obesity. We suggest here that Endo1, most likely by controlling CD36 cell surface abundance and lipid uptake in adipocytes, dissociates obesity from diabetes and that its absence participates in metabolically healthy obesity.


Subject(s)
Adipose Tissue , CD36 Antigens , Diet, High-Fat , Mice, Knockout , Obesity , Animals , Female , Humans , Male , Mice , Adipocytes/metabolism , Adipose Tissue/metabolism , CD36 Antigens/metabolism , CD36 Antigens/genetics , Diet, High-Fat/adverse effects , Glucose/metabolism , Lipid Metabolism/genetics , Mice, Inbred C57BL , Obesity/metabolism , Obesity/genetics
3.
PLoS One ; 19(5): e0300292, 2024.
Article in English | MEDLINE | ID: mdl-38718051

ABSTRACT

The aim of the study was to investigate the effect of returning to a balanced diet combined with chromium picolinate (CrPic) or chromium nanoparticles (CrNPs) supplementation at a pharmacologically relevant dose of 0.3 mg/kg body weight on the expression level of selected genes and bone turnover markers in the blood and bones of rats fed an obese diet. The results of the study showed that chronic intake of a high-fat obesogenic diet negatively affects bone turnover by impairing processes of both synthesis and degradation of bones. The switch to a healthy diet proved insufficient to regulate bone metabolism disorders induced by an obesogenic diet, even when it was supplemented with chromium, irrespective of its form. Supplementation with CrPic with no change in diet stimulated bone metabolism only at the molecular level, towards increased osteoclastogenesis (bone resorption). In contrast, CrNPs added to the high-fat diet effectively regulated bone turnover by increasing both osteoblastogenesis and osteoclastogenesis, with these changes directed more towards bone formation. The results of the study suggest that unfavourable changes in bone metabolism induced by chronic intake of a high-fat diet can be mitigated by supplementation with CrNPs, whereas a change in eating habits fails to achieve a similar effect.


Subject(s)
Bone Remodeling , Chromium , Diet, High-Fat , Animals , Diet, High-Fat/adverse effects , Rats , Chromium/administration & dosage , Chromium/pharmacology , Male , Bone Remodeling/drug effects , Nanoparticles/chemistry , Dietary Fiber/pharmacology , Picolinic Acids/pharmacology , Picolinic Acids/administration & dosage , Dietary Supplements , Bone and Bones/metabolism , Bone and Bones/drug effects , Rats, Wistar , Metal Nanoparticles/chemistry , Metal Nanoparticles/administration & dosage , Osteogenesis/drug effects
4.
Clin Exp Pharmacol Physiol ; 51(6): e13869, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38725222

ABSTRACT

Treatment with erythropoietin (EPO) can correct anaemia in chronic kidney disease (CKD) patients; however, up to 10% exhibit resistance or hyporesponsiveness to EPO. Non-alcoholic fatty liver disease (NAFLD), prevalent liver disease in CKD patients, may limit EPO response because of thrombopoietin deficiency, iron homeostasis disorder and inflammation. Therefore, we hypothesized NAFLD is a risk factor for EPO responsiveness. To test our hypothesis, we evaluated the effect of EPO in healthy rats and rats with NAFLD induced by a high-fat, high-carbohydrate (HFHC) diet. After 12 weeks on the HFHC diet, NAFLD rats showed lower erythroid response to EPO treatment than healthy rats. We, then, determined that the primary cause of EPO hyporesponsiveness could be iron deficiency associated with inflammation, which reduces erythroid cell production. Specifically, the concentrations of hepcidin, ferritin, transferrin and white blood cells in NAFLD rats were 12.8-, 16.4-, 2.51- and 1.40-fold higher than those in healthy rats, respectively. However, erythroid cell types in the bone marrow of NAFLD rats were significantly reduced. In conclusion, our data suggest that NAFLD could be a risk factor for EPO responsiveness, which is attributed to functional iron deficiency associated with inflammation.


Subject(s)
Erythropoietin , Non-alcoholic Fatty Liver Disease , Animals , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/pathology , Rats , Male , Rats, Sprague-Dawley , Diet, High-Fat/adverse effects , Hepcidins/metabolism
5.
World J Microbiol Biotechnol ; 40(7): 206, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38755297

ABSTRACT

The significance of microorganisms occurring in foods is predominantly targeted due to their application for identifying a novel range of the bacterial spectrum. Diverse microbial species are capable of exhibiting potential pharmacological activities like antimicrobial and anticancer. Microbial strains capable of reducing obesity-related syndromes have also been reported. In the present study, the hypocholesterolemic efficacy of Bacillus amyloliquefaciens isolated from dairy products was scrutinised by in vitro (3T3-L1 adipose cells) and in vivo (high-fat diet-induced obese Wistar albino rats) methods. Potential cholesterol-lowering isolates were screened using a plate assay method and optimised by physical parameters. Molecular identification of the topmost five cholesterol-lowering isolates was acquired by amplification of the 16 S rRNA gene region. Bacillus amyloliquefaciens strain KAVK1, followed by strains KAVK2, KAVK3, KAVK4, and KAVK5 were molecularly determined. Further, cholesterol-lowering strains degraded the spectral patterns determined by the side chain of a cholesterol molecule. The anti-lipase activity was demonstrated using the porcine pancreatic lipase inhibitory method and compared with the reference compound Atorvastatin. Lyophilised strain KAVK1 revealed maximum pancreatic lipase inhibition. Strain KAVK1 attenuated lipid accumulation in 3T3-L1 adipose cell line predicted by Oil Red O staining method. Significant reduction of body weight and change in lipid profile was recognised after the supplement of KAVK1 to obese rats. Histopathological changes in organs were predominantly marked. The result of this study implies that the cholesterol-lowering B. amyloliquefaciens KAVK1 strain was used to treat hypercholesterolemia.


Subject(s)
3T3-L1 Cells , Anticholesteremic Agents , Bacillus amyloliquefaciens , Diet, High-Fat , Lipid Metabolism , Obesity , RNA, Ribosomal, 16S , Rats, Wistar , Animals , Bacillus amyloliquefaciens/metabolism , Diet, High-Fat/adverse effects , Mice , Obesity/microbiology , Rats , Anticholesteremic Agents/pharmacology , Lipid Metabolism/drug effects , RNA, Ribosomal, 16S/genetics , Male , Disease Models, Animal , Cholesterol/metabolism , Lipase/metabolism , Adipocytes/metabolism , Adipocytes/drug effects
6.
FASEB J ; 38(10): e23656, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38752523

ABSTRACT

Coronavirus disease 2019 (COVID-19) is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Obesity is a major risk factor for the development of COVID-19. Angiotensin-converting enzyme 2 (ACE2) is an essential receptor for cell entry of SARS-CoV-2. The receptor-binding domain of the S1 subunit (S1-RBD protein) in the SARS-CoV-2 spike glycoprotein binds to ACE2 on host cells, through which the virus enters several organs, including the lungs. Considering these findings, recombinant ACE2 might be utilized as a decoy protein to attenuate SARS-CoV-2 infection. Here, we examined whether obesity increases ACE2 expression in the lungs and whether recombinant ACE2 administration diminishes the entry of S1-RBD protein into lung cells. We observed that high-fat diet-induced obesity promoted ACE2 expression in the lungs by increasing serum levels of LPS derived from the intestine. S1-RBD protein entered the lungs specifically through ACE2 expressed in host lungs and that the administration of recombinant ACE2 attenuated this entry. We conclude that obesity makes hosts susceptible to recombinant SARS-CoV-2 spike proteins due to elevated ACE2 expression in lungs, and this model of administering S1-RBD protein can be applied to new COVID-19 treatments.


Subject(s)
Angiotensin-Converting Enzyme 2 , COVID-19 , Diet, High-Fat , Lung , Obesity , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Animals , Spike Glycoprotein, Coronavirus/metabolism , Spike Glycoprotein, Coronavirus/genetics , Diet, High-Fat/adverse effects , Mice , Angiotensin-Converting Enzyme 2/metabolism , Angiotensin-Converting Enzyme 2/genetics , Lung/metabolism , Lung/virology , SARS-CoV-2/metabolism , Obesity/metabolism , COVID-19/metabolism , COVID-19/virology , Mice, Inbred C57BL , Virus Internalization , Male , Humans , Mice, Obese , Recombinant Proteins/metabolism
7.
Front Endocrinol (Lausanne) ; 15: 1387964, 2024.
Article in English | MEDLINE | ID: mdl-38742193

ABSTRACT

The high prevalence of obesity has become a pressing global public health problem and there exists a strong association between increased BMI and mortality at a BMI of 25 kg/m2 or higher. The prevalence of obesity is higher among middle-aged adults than among younger groups and the combination of aging and obesity exacerbate systemic inflammation. Increased inflammatory cytokines such as interleukin 6 and tumor necrosis factor alpha (TNFα) are hallmarks of obesity, and promote the secretion of hepatic C-reactive protein (CRP) which further induces systematic inflammation. The neuropeptide oxytocin has been shown to have anti-obesity and anti-inflammation effects, and also suppress sweet-tasting carbohydrate consumption in mammals. Previously, we have shown that the Japanese herbal medicine Kamikihito (KKT), which is used to treat neuropsychological stress disorders in Japan, functions as an oxytocin receptors agonist. In the present study, we further investigated the effect of KKT on body weight (BW), food intake, inflammation, and sweet preferences in middle-aged obese mice. KKT oral administration for 12 days decreased the expression of pro-inflammatory cytokines in the liver, and the plasma CRP and TNFα levels in obese mice. The effect of KKT administration was found to be different between male and female mice. In the absence of sucrose, KKT administration decreased food intake only in male mice. However, while having access to a 30% sucrose solution, both BW and food intake was decreased by KKT administration in male and female mice; but sucrose intake was decreased in female mice alone. In addition, KKT administration decreased sucrose intake in oxytocin deficient lean mice, but not in the WT lean mice. The present study demonstrates that KKT ameliorates chronic inflammation, which is strongly associated with aging and obesity, and decreases food intake in male mice as well as sucrose intake in female mice; in an oxytocin receptor dependent manner.


Subject(s)
Diet, High-Fat , Drugs, Chinese Herbal , Inflammation , Mice, Inbred C57BL , Obesity , Animals , Obesity/metabolism , Obesity/drug therapy , Male , Mice , Diet, High-Fat/adverse effects , Inflammation/metabolism , Female , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use , Sucrose/administration & dosage , Food Preferences/drug effects , Body Weight/drug effects , Oxytocin/pharmacology , Medicine, Kampo , East Asian People
8.
FASEB J ; 38(10): e23669, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38747734

ABSTRACT

Amomum xanthioides (AX) has been used as an edible herbal medicine to treat digestive system disorders in Asia. Additionally, Lactobacillus casei is a well-known probiotic commonly used in fermentation processes as a starter. The current study aimed to investigate the potential of Lactobacillus casei-fermented Amomum xanthioides (LAX) in alleviating metabolic disorders induced by high-fat diet (HFD) in a mouse model. LAX significantly reduced the body and fat weight, outperforming AX, yet without suppressing appetite. LAX also markedly ameliorated excessive lipid accumulation and reduced inflammatory cytokine (IL-6) levels in serum superior to AX in association with UCP1 activation and adiponectin elevation. Furthermore, LAX noticeably improved the levels of fasting blood glucose, serum insulin, and HOMA-IR through positive regulation of glucose transporters (GLUT2, GLUT4), and insulin receptor gene expression. In conclusion, the fermentation of AX demonstrates a pronounced mitigation of overnutrition-induced metabolic dysfunction, including hyperlipidemia, hyperglycemia, hyperinsulinemia, and obesity, compared to non-fermented AX. Consequently, we proposed that the fermentation of AX holds promise as a potential candidate for effectively ameliorating metabolic disorders.


Subject(s)
Amomum , Diet, High-Fat , Fermentation , Lacticaseibacillus casei , Obesity , Animals , Diet, High-Fat/adverse effects , Mice , Obesity/metabolism , Male , Lacticaseibacillus casei/metabolism , Amomum/chemistry , Mice, Inbred C57BL , Probiotics/pharmacology , Uncoupling Protein 1/metabolism , Insulin Resistance , Mice, Obese , Adiponectin/metabolism , Insulin/metabolism , Insulin/blood , Blood Glucose/metabolism
9.
Biochem Biophys Res Commun ; 716: 150026, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38701557

ABSTRACT

BACKGROUND: Previous in vivo and in vitro studies have demonstrated that estrogen receptor agonist G-1 regulates glucose and lipid metabolism. This study focused on the effects of G-1 on cardiometabolic syndrome and anti-obesity under a high fat diet (HFD). METHODS: Bilateral ovariectomized female mice were fed an HFD for 6 weeks, and treated them with G-1. A cardiomyocyte insulin resistance model was used to simulate the in vivo environment. The main outcome measures were blood glucose, body weight, and serum insulin levels to assess insulin resistance, while cardiac function and degree of fibrosis were assessed by cardiac ultrasound and pathological observations. We also examined the expression of p-AMPK, p-AKT, and GLUT4 in mice hearts and in vitro models to explore the mechanism by which G-1 regulates insulin signaling. RESULTS: G-1 reduced body weight in mice on an HFD, but simultaneously increased blood glucose and promoted insulin resistance, resulting in myocardial damage. This damage included disordered cardiomyocytes, massive accumulation of glycogen, extensive fibrosis of the heart, and thickening of the front and rear walls of the left ventricle. At the molecular level, G-1 enhances gluconeogenesis and promotes glucose production by increasing the activity of pyruvate carboxylase (PC) while inhibiting GLUT4 translocation via the AMPK/TBC1D1 pathway, thereby limiting glucose uptake. CONCLUSION: Despite G-1's the potential efficacy in weight reduction, the concomitant induction of insulin resistance and cardiac impairment in conjunction with an HFD raises significant concerns. Therefore, comprehensive studies of its safety profile and effects under specific conditions are essential prior to clinical use.


Subject(s)
Diet, High-Fat , Insulin Resistance , Mice, Inbred C57BL , Ovariectomy , Receptors, G-Protein-Coupled , Animals , Female , Diet, High-Fat/adverse effects , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/metabolism , Mice , Glucose Transporter Type 4/metabolism , Receptors, Estrogen/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Insulin/metabolism , Insulin/blood
10.
Int J Oral Sci ; 16(1): 39, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38740741

ABSTRACT

The aim of this study was to explore the impact of chronic apical periodontitis (CAP) on atherosclerosis in apoE-/- mice fed high-fat diet (HFD). This investigation focused on the gut microbiota, metabolites, and intestinal barrier function to uncover potential links between oral health and cardiovascular disease (CVD). In this study, CAP was shown to exacerbate atherosclerosis in HFD-fed apoE-/- mice, as evidenced by the increase in plaque size and volume in the aortic walls observed via Oil Red O staining. 16S rRNA sequencing revealed significant alterations in the gut microbiota, with harmful bacterial species thriving while beneficial species declining. Metabolomic profiling indicated disruptions in lipid metabolism and primary bile acid synthesis, leading to elevated levels of taurochenodeoxycholic acid (TCDCA), taurocholic acid (TCA), and tauroursodeoxycholic acid (TDCA). These metabolic shifts may contribute to atherosclerosis development. Furthermore, impaired intestinal barrier function, characterized by reduced mucin expression and disrupted tight junction proteins, was observed. The increased intestinal permeability observed was positively correlated with the severity of atherosclerotic lesions, highlighting the importance of the intestinal barrier in cardiovascular health. In conclusion, this research underscores the intricate interplay among oral health, gut microbiota composition, metabolite profiles, and CVD incidence. These findings emphasize the importance of maintaining good oral hygiene as a potential preventive measure against cardiovascular issues, as well as the need for further investigations into the intricate mechanisms linking oral health, gut microbiota, and metabolic pathways in CVD development.


Subject(s)
Atherosclerosis , Diet, High-Fat , Dysbiosis , Gastrointestinal Microbiome , Animals , Diet, High-Fat/adverse effects , Atherosclerosis/metabolism , Mice , Male , Periapical Periodontitis/metabolism , Periapical Periodontitis/microbiology , Apolipoproteins E/metabolism , Mice, Inbred C57BL , RNA, Ribosomal, 16S
11.
Sci Rep ; 14(1): 10855, 2024 05 13.
Article in English | MEDLINE | ID: mdl-38740782

ABSTRACT

Type 2 diabetes mellitus (T2DM) is a chronic inflammatory disease that can compromise the functioning of various organs, including the salivary glands (SG). The purinergic system is one of the most important inflammatory pathways in T2DM condition, and P2X7R and P2X4R are the primary purinergic receptors in SG that regulate inflammatory homeostasis. This study aimed to evaluate P2X7R and P2X4R expression, and morphological changes in the submandibular gland (SMG) in T2DM. Twenty-four 5-week-old mice were randomly assigned to control (CON) and diabetes mellitus (DM) groups (n = 12 each). Body weight, diet, and blood glucose levels were monitored weekly. The histomorphology of the SMG and the expression of the P2X7R, and P2X7R was evaluated by immunohistochemistry (IHC) staining and reverse transcription-quantitative polymerase chain reaction (RT-qPCR) at 11 and 13 weeks of age. Our findings indicate a significant increase in food consumption, body weight, and blood glucose levels in the DM group. Although a significant increase in P2X7R and P2X4R expression was observed in the DM groups, the receptor location remained unchanged. We also observed a significant increase in the acinar area in the DM13w group, and a significant decrease in the ductal area in the DM11w and DM13w groups. Targeting purinergic receptors may offer novel therapeutic methods for diabetic complications.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2 , Diet, High-Fat , Receptors, Purinergic P2X4 , Receptors, Purinergic P2X7 , Submandibular Gland , Animals , Submandibular Gland/metabolism , Submandibular Gland/pathology , Receptors, Purinergic P2X4/metabolism , Receptors, Purinergic P2X4/genetics , Receptors, Purinergic P2X7/metabolism , Receptors, Purinergic P2X7/genetics , Diet, High-Fat/adverse effects , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Mice , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Male , Blood Glucose/metabolism , Body Weight , Streptozocin , Mice, Inbred C57BL
12.
Front Endocrinol (Lausanne) ; 15: 1388361, 2024.
Article in English | MEDLINE | ID: mdl-38745946

ABSTRACT

Introduction: The pathogenesis of Post-Transplant Diabetes Mellitus (PTDM) is complex and multifactorial and it resembles that of Type-2 Diabetes Mellitus (T2DM). One risk factor specific to PTDM differentiates both entities: the use of immunosuppressive therapy. Specifically, Tacrolimus interacts with obesity and insulin resistance (IR) in accelerating the onset of PTDM. In a genotypic model of IR, the obese Zucker rats, Tacrolimus is highly diabetogenic by promoting the same changes in beta-cell already modified by IR. Nevertheless, genotypic animal models have their limitations and may not resemble the real pathophysiology of diabetes. In this study, we have evaluated the interaction between beta-cell damage and Tacrolimus in a non-genotypic animal model of obesity and metabolic syndrome. Methods: Sprague Dawley rats were fed a high-fat enriched diet during 45 days to induce obesity and metabolic dysregulation. On top of this established obesity, the administration of Tacrolimus (1mg/kg/day) during 15 days induced severe hyperglycaemia and changes in morphological and structural characteristics of the pancreas. Results: Obese animals administered with Tacrolimus showed increased size of islets of Langerhans and reduced beta-cell proliferation without changes in apoptosis. There were also changes in beta-cell nuclear factors such as a decrease in nuclear expression of MafA and a nuclear overexpression of FoxO1A, PDX-1 and NeuroD1. These animals also showed increased levels of pancreatic insulin and glucagon. Discussion: This model could be evidence of the relationship between the T2DM and PTDM physiopathology and, eventually, the model may be instrumental to study the pathogenesis of T2DM.


Subject(s)
Disease Models, Animal , Metabolic Syndrome , Obesity , Rats, Sprague-Dawley , Tacrolimus , Animals , Tacrolimus/pharmacology , Metabolic Syndrome/metabolism , Metabolic Syndrome/pathology , Metabolic Syndrome/chemically induced , Obesity/metabolism , Obesity/pathology , Rats , Male , Immunosuppressive Agents/adverse effects , Immunosuppressive Agents/pharmacology , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Insulin-Secreting Cells/drug effects , Phenotype , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/metabolism , Insulin Resistance , Diet, High-Fat/adverse effects
13.
Front Cell Infect Microbiol ; 14: 1347716, 2024.
Article in English | MEDLINE | ID: mdl-38716198

ABSTRACT

High-fat diets (HFDs), a prevailing daily dietary style worldwide, induce chronic low-grade inflammation in the central nervous system and peripheral tissues, promoting a variety of diseases including pathologies associated with neuroinflammation. However, the mechanisms linking HFDs to inflammation are not entirely clear. Here, using a Drosophila HFD model, we explored the mechanism of HFD-induced inflammation in remote tissues. We found that HFDs activated the IMD/NFκB immune pathway in the head through remodeling of the commensal gut bacteria. Removal of gut microbiota abolished such HFD-induced remote inflammatory response. Further experiments revealed that HFDs significantly increased the abundance of Acetobacter malorum in the gut, and the re-association of this bacterium was sufficient to elicit inflammatory response in remote tissues. Mechanistically, Acetobacter malorum produced a greater amount of peptidoglycan (PGN), a well-defined microbial molecular pattern that enters the circulation and remotely activates an inflammatory response. Our results thus show that HFDs trigger inflammation mediated by a bacterial molecular pattern that elicits host immune response.


Subject(s)
Diet, High-Fat , Drosophila Proteins , Gastrointestinal Microbiome , Inflammation , NF-kappa B , Signal Transduction , Animals , Diet, High-Fat/adverse effects , NF-kappa B/metabolism , Inflammation/metabolism , Drosophila Proteins/metabolism , Acetobacter/metabolism , Drosophila/microbiology , Peptidoglycan/metabolism , Disease Models, Animal , Drosophila melanogaster/microbiology
14.
Redox Rep ; 29(1): 2347139, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38718286

ABSTRACT

OBJECTIVES: The objective of this study was to investigate whether skeletal muscle cystathionine γ-lyase (CTH) contributes to high-fat diet (HFD)-induced metabolic disorders using skeletal muscle Cth knockout (CthΔskm) mice. METHODS: The CthΔskm mice and littermate Cth-floxed (Cthf/f) mice were fed with either HFD or chow diet for 13 weeks. Metabolomics and transcriptome analysis were used to assess the impact of CTH deficiency in skeletal muscle. RESULTS: Metabolomics coupled with transcriptome showed that CthΔskm mice displayed impaired energy metabolism and some signaling pathways linked to insulin resistance (IR) in skeletal muscle although the mice had normal insulin sensitivity. HFD led to reduced CTH expression and impaired energy metabolism in skeletal muscle in Cthf/f mice. CTH deficiency and HFD had some common pathways enriched in the aspects of amino acid metabolism, carbon metabolism, and fatty acid metabolism. CthΔskm+HFD mice exhibited increased body weight gain, fasting blood glucose, plasma insulin, and IR, and reduced glucose transporter 4 and CD36 expression in skeletal muscle compared to Cthf/f+HFD mice. Impaired mitochondria and irregular arrangement in myofilament occurred in CthΔskm+HFD mice. Omics analysis showed differential pathways enriched between CthΔskm mice and Cthf/f mice upon HFD. More severity in impaired energy metabolism, reduced AMPK signaling, and increased oxidative stress and ferroptosis occurred in CthΔskm+HFD mice compared to Cthf/f+HFD mice. DISCUSSION: Our results indicate that skeletal muscle CTH expression dysregulation contributes to metabolism disorders upon HFD.


Subject(s)
Cystathionine gamma-Lyase , Diet, High-Fat , Hyperglycemia , Insulin Resistance , Muscle, Skeletal , Obesity , Animals , Insulin Resistance/physiology , Muscle, Skeletal/metabolism , Mice , Obesity/metabolism , Cystathionine gamma-Lyase/metabolism , Cystathionine gamma-Lyase/genetics , Cystathionine gamma-Lyase/deficiency , Diet, High-Fat/adverse effects , Hyperglycemia/metabolism , Mice, Knockout , Male , Energy Metabolism
15.
Hepatol Commun ; 8(5)2024 May 01.
Article in English | MEDLINE | ID: mdl-38704651

ABSTRACT

BACKGROUND: Alcohol-associated liver disease is a complex disease regulated by genetic and environmental factors such as diet and sex. The combination of high-fat diet and alcohol consumption has synergistic effects on liver disease progression. Female sex hormones are known to protect females from liver disease induced by high-fat diet. In contrast, they promote alcohol-mediated liver injury. We aimed to define the role of female sex hormones on liver disease induced by a combination of high-fat diet and alcohol. METHODS: Wild-type and protein arginine methyltransferase (Prmt)6 knockout female mice were subjected to gonadectomy (ovariectomy, OVX) or sham surgeries and then fed western diet and alcohol in the drinking water. RESULTS: We found that female sex hormones protected mice from western diet/alcohol-induced weight gain, liver steatosis, injury, and fibrosis. Our data suggest that these changes are, in part, mediated by estrogen-mediated induction of arginine methyltransferase PRMT6. Liver proteome changes induced by OVX strongly correlated with changes induced by Prmt6 knockout. Using Prmt6 knockout mice, we confirmed that OVX-mediated weight gain, steatosis, and injury are PRMT6 dependent, while OVX-induced liver fibrosis is PRMT6 independent. Proteomic and gene expression analyses revealed that estrogen signaling suppressed the expression of several components of the integrin pathway, thus reducing integrin-mediated proinflammatory (Tnf, Il6) and profibrotic (Tgfb1, Col1a1) gene expression independent of PRMT6 levels. Integrin signaling inhibition using Arg-Gly-Asp peptides reduced proinflammatory and profibrotic gene expression in mice, suggesting that integrin suppression by estrogen is protective against fibrosis development. CONCLUSIONS: Taken together, estrogen signaling protects mice from liver disease induced by a combination of alcohol and high-fat diet through upregulation of Prmt6 and suppression of integrin signaling.


Subject(s)
Estradiol , Integrins , Mice, Knockout , Protein-Arginine N-Methyltransferases , Signal Transduction , Animals , Mice , Female , Signal Transduction/drug effects , Protein-Arginine N-Methyltransferases/metabolism , Protein-Arginine N-Methyltransferases/genetics , Integrins/metabolism , Diet, High-Fat/adverse effects , Ovariectomy , Ethanol/adverse effects , Liver Cirrhosis, Alcoholic/metabolism , Liver Cirrhosis, Alcoholic/prevention & control , Liver Cirrhosis, Alcoholic/pathology , Liver/metabolism , Liver/pathology , Liver/drug effects , Mice, Inbred C57BL , Disease Models, Animal
16.
Biol Res ; 57(1): 23, 2024 May 06.
Article in English | MEDLINE | ID: mdl-38705984

ABSTRACT

Obesity, associated with the intake of a high-fat diet (HFD), and anxiety are common among those living in modern urban societies. Recent studies suggest a role of microbiome-gut-brain axis signaling, including a role for brain serotonergic systems in the relationship between HFD and anxiety. Evidence suggests the gut microbiome and the serotonergic brain system together may play an important role in this response. Here we conducted a nine-week HFD protocol in male rats, followed by an analysis of the gut microbiome diversity and community composition, brainstem serotonergic gene expression (tph2, htr1a, and slc6a4), and anxiety-related defensive behavioral responses. We show that HFD intake decreased alpha diversity and altered the community composition of the gut microbiome in association with obesity, increased brainstem tph2, htr1a and slc6a4 mRNA expression, including in the caudal part of the dorsomedial dorsal raphe nucleus (cDRD), a subregion previously associated with stress- and anxiety-related behavioral responses, and, finally, increased anxiety-related defensive behavioral responses. The HFD increased the Firmicutes/Bacteroidetes ratio relative to control diet, as well as higher relative abundances of Blautia, and decreases in Prevotella. We found that tph2, htr1a and slc6a4 mRNA expression were increased in subregions of the dorsal raphe nucleus in the HFD, relative to control diet. Specific bacterial taxa were associated with increased serotonergic gene expression in the cDRD. Thus, we propose that HFD-induced obesity is associated with altered microbiome-gut-serotonergic brain axis signaling, leading to increased anxiety-related defensive behavioral responses in rats.


Subject(s)
Anxiety , Brain-Gut Axis , Diet, High-Fat , Gastrointestinal Microbiome , Animals , Male , Diet, High-Fat/adverse effects , Gastrointestinal Microbiome/physiology , Anxiety/microbiology , Brain-Gut Axis/physiology , Rats , Rats, Sprague-Dawley , Obesity/microbiology , Obesity/psychology , Obesity/metabolism , Signal Transduction/physiology , Behavior, Animal/physiology
17.
Cells ; 13(9)2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38727268

ABSTRACT

Treatment strategies for steatohepatitis are of special interest given the high prevalence of obesity and fatty liver disease worldwide. This study aimed to investigate the potential therapeutic mechanism of L-carnitine (LC) and Ginkgo biloba leaf extract (GB) supplementation in ameliorating the adverse effects of hyperlipidemia and hepatosteatosis induced by a high-cholesterol diet (HCD) in an animal model. The study involved 50 rats divided into five groups, including a control group, a group receiving only an HCD, and three groups receiving an HCD along with either LC (300 mg LC/kg bw), GB (100 mg GB/kg bw), or both. After eight weeks, various parameters related to lipid and glucose metabolism, antioxidant capacity, histopathology, immune reactivity, and liver ultrastructure were measured. LC + GB supplementation reduced serum total cholesterol, triglyceride, low-density lipoprotein cholesterol, glucose, insulin, HOMA-IR, alanine transaminase, and aspartate transaminase levels and increased high-density lipoprotein cholesterol levels compared with those in the HCD group. Additionally, treatment with both supplements improved antioxidant ability and reduced lipid peroxidation. The histological examination confirmed that the combination therapy reduced liver steatosis and fibrosis while also improving the appearance of cell organelles in the ultrastructural hepatocytes. Finally, the immunohistochemical analysis indicated that cotreatment with LC + GB upregulated the immune expression of GLP-1 and ß-Cat in liver sections that were similar to those of the control animals. Mono-treatment with LC or GB alone substantially but not completely protected the liver tissue, while the combined use of LC and GB may be more effective in treating liver damage caused by high cholesterol than either supplement alone by regulating hepatic oxidative stress and the protein expression of GLP-1 and ß-Cat.


Subject(s)
Carnitine , Dietary Supplements , Dyslipidemias , Ginkgo biloba , Liver , Plant Extracts , Animals , Liver/drug effects , Liver/pathology , Liver/metabolism , Carnitine/pharmacology , Male , Rats , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Dyslipidemias/drug therapy , Dyslipidemias/metabolism , Fatty Liver/drug therapy , Fatty Liver/pathology , Fatty Liver/metabolism , Rats, Sprague-Dawley , Lipid Metabolism/drug effects , Antioxidants/pharmacology , Diet, High-Fat/adverse effects , Ginkgo Extract
18.
FASEB J ; 38(9): e23643, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38703030

ABSTRACT

Secreted phospholipase A2s are involved in the development of obesity, type 2 diabetes mellitus (T2DM) and cardiovascular disease, which have become serious and growing health concerns worldwide. Integration of genome-wide association study and gene co-expression networks analysis showed that the secreted phospholipase A2 group XIIA (PLA2G12A) may participate in hepatic lipids metabolism. Nevertheless, the role of PLA2G12A in lipid metabolism and its potential mechanism remain elusive. Here, we used AAV9 vector carrying human PLA2G12A gene to exogenously express hPLA2G12A in the liver of mice. We demonstrated that the overexpression of hPLA2G12A resulted in a significant decrease in serum lipid levels in wild-type mice fed with chow diet or high-fat diet (HFD). Moreover, hPLA2G12A treatment protected against diet-induced obesity and insulin resistance in mice fed a HFD. Notably, we found that hPLA2G12A treatment confers protection against obesity and hyperlipidemia independent of its enzymatic activity, but rather by increasing physical activity and energy expenditure. Furthermore, we demonstrated that hPLA2G12A treatment induced upregulation of ApoC2 and Cd36 and downregulation of Angptl8, which contributed to the increase in clearance of circulating triglycerides and hepatic uptake of fatty acids without affecting hepatic de novo lipogenesis, very low-density lipoprotein secretion, or intestinal lipid absorption. Our study highlights the potential of PLA2G12A gene therapy as a promising approach for treating obesity, insulin resistance and T2DM.


Subject(s)
Diet, High-Fat , Energy Metabolism , Insulin Resistance , Mice, Inbred C57BL , Obesity , Triglycerides , Animals , Obesity/metabolism , Obesity/etiology , Mice , Triglycerides/metabolism , Triglycerides/blood , Male , Diet, High-Fat/adverse effects , Humans , Liver/metabolism , Lipid Metabolism
19.
Adipocyte ; 13(1): 2339418, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38706095

ABSTRACT

A Disintegrin And Metalloproteinase domain-containing protein 10 (ADAM10), is involved in several metabolic and inflammatory pathways. We speculated that ADAM10 plays a modulatory role in adipose tissue inflammation and metabolism. To this end, we studied adipose tissue-specific ADAM10 knock-out mice (aKO). While young, regular chow diet-fed aKO mice showed increased insulin sensitivity, following prolonged (33 weeks) high-fat diet (HFD) exposure, aKO mice developed obesity and insulin resistance. Compared to controls, aKO mice showed less inflammatory adipokine profile despite the significant increase in adiposity. In brown adipose tissue, aKO mice on HFD had changes in CD8+ T cell populations indicating a lesser inflammatory pattern. Following HFD, both aKO and control littermates demonstrated decreased adipose tissue pro-inflammatory macrophages, and increased anti-inflammatory accumulation, without differences between the genotypes. Collectively, our observations indicate that selective deletion of ADAM10 in adipocytes results in a mitigated inflammatory response, leading to increased insulin sensitivity in young mice fed with regular diet. This state of insulin sensitivity, following prolonged HFD, facilitates energy storage resulting in increased fat accumulation which ultimately leads to the development of a phenotype of obesity and insulin resistance. In conclusion, the data indicate that ADAM10 has a modulatory effect of inflammation and whole-body energy metabolism.


Subject(s)
ADAM10 Protein , Adipose Tissue , Diet, High-Fat , Mice, Knockout , Animals , Male , Mice , ADAM10 Protein/metabolism , ADAM10 Protein/genetics , Adipocytes/metabolism , Adipose Tissue/metabolism , Amyloid Precursor Protein Secretases/metabolism , Diet, High-Fat/adverse effects , Inflammation/metabolism , Insulin Resistance , Membrane Proteins/metabolism , Membrane Proteins/genetics , Obesity/metabolism , Obesity/etiology , Phenotype
20.
Physiol Res ; 73(2): 273-284, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38710057

ABSTRACT

Lifestyle intervention encompassing nutrition and physical activity are effective strategies to prevent progressive lipid deposition in the liver. This study aimed to explore the effect of dietary change, and/or high-intensity interval training (HIIT) on hepatic lipid accumulation in high fat diet (HFD)-induced obese rats. We divided lean rats into lean control (LC) or HIIT groups (LH), and obese rats into obese normal chow diet (ND) control (ONC) or HIIT groups (ONH) and obese HFD control (OHC) or HIIT groups (OHH). We found that dietary or HIIT intervention significantly decreased body weight and the risk of dyslipidemia, prevented hepatic lipid accumulation. HIIT significantly improved mitochondrial fatty acid oxidation through upregulating mitochondrial enzyme activities, mitochondrial function and AMPK/PPARalpha/CPT1alpha pathway, as well as inhibiting hepatic de novo lipogenesis in obese HFD rats. These findings indicate that dietary alone or HIIT intervention powerfully improve intrahepatic storage of fat in diet induced obese rats. Keywords: Obesity, Exercise, Diet, Mitochondrial function, Lipid deposition.


Subject(s)
Diet, High-Fat , High-Intensity Interval Training , Lipid Metabolism , Liver , Obesity , Rats, Sprague-Dawley , Animals , Obesity/metabolism , Obesity/therapy , Male , Diet, High-Fat/adverse effects , Rats , Liver/metabolism , Physical Conditioning, Animal/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...