Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 95
Filter
Add more filters

Publication year range
1.
J Nanobiotechnology ; 22(1): 13, 2024 Jan 03.
Article in English | MEDLINE | ID: mdl-38167034

ABSTRACT

In recent years, the environmental health issue of microplastics has aroused an increasingly significant concern. Some studies suggested that exposure to polystyrene microplastics (PS-MPs) may lead to renal inflammation and oxidative stress in animals. However, little is known about the essential effects of PS-MPs with high-fat diet (HFD) on renal development and microenvironment. In this study, we provided the single-cell transcriptomic landscape of the kidney microenvironment induced by PS-MPs and HFD in mouse models by unbiased single-cell RNA sequencing (scRNA-seq). The kidney injury cell atlases in mice were evaluated after continued PS-MPs exposure, or HFD treated for 35 days. Results showed that PS-MPs plus HFD treatment aggravated the kidney injury and profibrotic microenvironment, reshaping mouse kidney cellular components. First, we found that PS-MPs plus HFD treatment acted on extracellular matrix organization of renal epithelial cells, specifically the proximal and distal convoluted tubule cells, to inhibit renal development and induce ROS-driven carcinogenesis. Second, PS-MPs plus HFD treatment induced activated PI3K-Akt, MAPK, and IL-17 signaling pathways in endothelial cells. Besides, PS-MPs plus HFD treatment markedly increased the proportions of CD8+ effector T cells and proliferating T cells. Notably, mononuclear phagocytes exhibited substantial remodeling and enriched in oxidative phosphorylation and chemical carcinogenesis pathways after PS-MPs plus HFD treatment, typified by alterations tissue-resident M2-like PF4+ macrophages. Multispectral immunofluorescence and immunohistochemistry identified PF4+ macrophages in clear cell renal cell carcinoma (ccRCC) and adjacent normal tissues, indicating that activate PF4+ macrophages might regulate the profibrotic and pro-tumorigenic microenvironment after renal injury. In conclusion, this study first systematically revealed molecular variation of renal cells and immune cells in mice kidney microenvironment induced by PS-MPs and HFD with the scRNA-seq approach, which provided a molecular basis for decoding the effects of PS-MPs on genitourinary injury and understanding their potential profibrotic and carcinogenesis in mammals.


Subject(s)
Microplastics , Polystyrenes , Mice , Animals , Microplastics/toxicity , Plastics , Single-Cell Gene Expression Analysis , Diet, High-Fat/adverse effects , Endothelial Cells , Phosphatidylinositol 3-Kinases , Kidney , Carcinogenesis , Mammals , Tumor Microenvironment
2.
J Fish Biol ; 104(6): 2068-2080, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38596840

ABSTRACT

The pervasive utilization of plastics and their integration into ecosystems has resulted in significant environmental issues, particularly the pollution of microplastics (MPs). In aquaculture, high-fat feed (HFD) is frequently employed to enhance the energy intake and economic fish production. This study utilized zebrafish as a model organism to investigate the impact of concurrent exposure to HFD and MPs on fish intestinal pathology damage and intestinal microbiome. The experimental design involved the division of zebrafish into two groups: one receiving a normal diet (ND) and the other receiving HFD. The zebrafish were exposed to a control group, as well as polystyrene (PS) MPs of varying sizes (5 and 50 µm). Histopathological examination revealed that the combination of 5 µm MPs and HFD resulted in the most significant damage to the zebrafish intestinal tract. Furthermore, gut microbiome assays indicated that exposure to MPs and HFD altered the composition of the gut microbiome. This study demonstrates that in aquaculture, the issue of HFD must be considered alongside concerns about MPs contamination, as both factors appear to have a combined effect on the intestinal pathology damage and intestinal microbiome. The findings of this research offer valuable insights for the improvement of fish farming practices.


Subject(s)
Gastrointestinal Microbiome , Intestines , Microplastics , Polystyrenes , Water Pollutants, Chemical , Zebrafish , Animals , Zebrafish/microbiology , Microplastics/toxicity , Polystyrenes/toxicity , Polystyrenes/adverse effects , Gastrointestinal Microbiome/drug effects , Intestines/pathology , Intestines/microbiology , Intestines/drug effects , Water Pollutants, Chemical/toxicity , Water Pollutants, Chemical/adverse effects , Aquaculture , Diet, High-Fat/adverse effects , Animal Feed/analysis
3.
Biochem Biophys Res Commun ; 686: 149163, 2023 12 17.
Article in English | MEDLINE | ID: mdl-37924667

ABSTRACT

The expression of brain-derived neurotrophic factor (BDNF) is observed not only in the brain, but also in peripheral tissues including white adipose tissues (WATs). Here, we showed that the mRNA expression of Bdnf in inguinal WAT (iWAT) and epididymal WAT (eWAT) increased within 2 weeks of feeding mice with a high-fat diet (HFD). In mice on a 2-week HFD, the induction of Bdnf expression in WATs was significantly correlated with increases in body weight, suggesting that Bdnf expression may increase at an early stage of obesity. The mRNA expression of hypoxia-inducible factor 1α and platelet-derived growth factor, which are involved in neovascularization and the subsequent expansion of adipose tissues, increased in the iWAT of mice on the 2-week HFD. We also found that the expression of macrophage marker F4/80 in iWAT increased under the HFD. Interestingly, HFD-induced Bdnf expression in iWAT was not observed when macrophages were removed by the administration of clodronate liposomes. Accordingly, mice receiving clodronate liposomes also exhibited a significant reduction in the HFD-induced increase in body weight. In conclusion, increased body weight in HFD-induced obese model mice was accompanied by the induction of Bdnf expression in iWAT and was probably mediated by macrophages. Our findings imply a novel function for BDNF in iWAT at an early stage of obesity.


Subject(s)
Brain-Derived Neurotrophic Factor , Diet, High-Fat , Mice , Animals , Diet, High-Fat/adverse effects , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Clodronic Acid , Liposomes/metabolism , Adipose Tissue, White/metabolism , Adipose Tissue/metabolism , Obesity/etiology , Obesity/metabolism , Body Weight , Macrophages/metabolism , RNA, Messenger/metabolism , Mice, Inbred C57BL
4.
Fish Shellfish Immunol ; 137: 108803, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37164123

ABSTRACT

Extensive use of microplastics (MPs) threatens the safety of aquatic environments and hydrobionts. Increasing the weight of economic fish through high-fat diet (HFD) to increase production is common in aquaculture. However, little is known about the combined effects of MPs and HFD in fish. The aim of this study was to investigate the relationship between adiposity and MP bioaccumulation in fish. Using zebrafish as a vertebrate model, the content of polystyrene (PS) MPs in zebrafish tissues exposed to 5 and 50 µm of 1000 µg/L PS MPs was detected via confocal Raman spectroscopy in normal diet (ND) and HFD. The content of PS MPs in HFD group was significantly higher than that in ND group. The levels of hepatic lipids were significantly elevated in zebrafish subjected to HFD treatment, and this effect was aggravated by exposure to 5 µm PS MPs, and even caused liver injury. Transcriptomic analysis revealed that exposure to PS MPs interferes with hepatic lipid metabolism and energy homeostasis in zebrafish. These results suggests that in addition to controlling the use and performing proper recycling of plastic products in our daily life, we should not blindly increase the weight of fish through HFD. This aids protect the quality of economic fish and prevent MPs from being consumed by humans through the food chain. This study explored the interaction between fish feed culture and environmental pollutants to provide important reference for fish culture.


Subject(s)
Polystyrenes , Water Pollutants, Chemical , Humans , Animals , Polystyrenes/toxicity , Microplastics/toxicity , Plastics , Zebrafish/metabolism , Bioaccumulation , Lipid Metabolism , Diet, High-Fat/adverse effects , Water Pollutants, Chemical/toxicity
5.
Int Endod J ; 56(2): 164-178, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36261317

ABSTRACT

AIM: To analyse the effects of melatonin (ME) treatment on oxidative stress and insulin resistance (IR) in rats with apical periodontitis (AP) fed a high-fat diet (HFD). METHODOLOGY: Eighty 60-day-old rats were divided into eight groups: control (CN), AP, HFD with AP (HFDAP), control with ME (CNME), AP with ME (APME), HFD with ME (HFDME) and HFD with AP+ME (HFDAPME). The animals from the HFD groups were fed a HFD throughout the experimental period. On day 7, the animals from the AP groups were subjected to experimental AP, and after 70 days, the ME groups were treated for 30 days. Glycaemia, insulinaemia, homeostatic model assessment for IR index, tumour necrosis factor-α (TNF-α), and interleukin-6 were analysed in plasma using biochemical tests and enzyme-linked immunosorbent assay. Thiobarbituric acid-reactive substances (TBARS), carbonyl protein (CP), superoxide dismutase (SOD), catalase, glutathione peroxidase, glutathione (GSH) and total antioxidant capacity (ferric reducing antioxidant power [FRAP]) were analysed in the gastrocnemius muscle. RESULTS: (1) Association of AP and HDF exacerbated IR, and ME treatment improved this alteration; (2) AP and HFD and their association showed increased TNF-α, and ME reversed it; (3) TBARS increased in the AP and HFDAP groups, and ME reversed only in the group with the association of disease and diet; (4) CP increased in all HFD groups and improved in the ME groups; (5) GSH activity decreased in all experimental groups, and ME increased this parameter only in the CN and AP groups; (6) FRAP did not change between the groups, but ME treatment increased its activity in the AP and HFD groups; (7) ME increased SOD in the CN and AP groups. CONCLUSION: Apical periodontitis and HFD promoted IR, and the association of AP with diet promoted IR exacerbation; this resistance might have been caused by an increase in TNF-α. AP promoted more intense changes in lipid oxidative damage than in protein oxidative damage. In non-enzymatic antioxidant defence, it was observed that both AP and HFD and their association promoted a decrease in GSH levels. Overall, ME treatment reversed changes such as oxidative stress and IR.


Subject(s)
Insulin Resistance , Melatonin , Periapical Periodontitis , Rats , Animals , Antioxidants/pharmacology , Melatonin/pharmacology , Melatonin/therapeutic use , Insulin Resistance/physiology , Tumor Necrosis Factor-alpha/metabolism , Diet, High-Fat/adverse effects , Thiobarbituric Acid Reactive Substances/metabolism , Thiobarbituric Acid Reactive Substances/pharmacology , Rats, Wistar , Oxidative Stress , Glutathione/metabolism , Superoxide Dismutase/metabolism
6.
Odontology ; 111(3): 687-696, 2023 Jul.
Article in English | MEDLINE | ID: mdl-36567367

ABSTRACT

To evaluate the effects of melatonin (MEL) on the expression of toll-like receptor-4 (TLR4); myeloid differentiation primary response protein-88 (MyD88); TIR-domain-containing adapter-inducing interferon-ß (TRIF); IFN regulatory-factor-3 (IRF-3); nuclear factor kappa-B (NF-κB); plasma concentrations of interleukin-1ß (IL-1ß) and lipopolysaccharide (LPS); and lipid profile of rats with apical periodontitis (AP) fed on a high-fat diet (HFD). Eighty 60-day-old rats were divided into eight groups: control, AP, HFD, HFDAP, CNMEL, APMEL, HFDMEL and HFDAPMEL. HFD groups were fed on a HFD for 107 days. On day 7, experimental AP was induced in the AP groups, and after 70 days, MEL (5 mg/kg) was administered to the MEL groups for 30 days. Plasma concentrations of LPS and IL-1ß were analyzed using enzyme-linked immunosorbent assay, and the lipid profile was analyzed using biochemical tests. The expression of proteins involved in the TLR4 pathway (TLR4, MyD88, TRIF, IRF-3 and NF-κB) in the gastrocnemius muscle (GM) was evaluated using western blotting and qRT-PCR. Treatment with MEL decreased IRF-3 protein expression in GM and IL-1ß plasma concentration in the APMEL and HFDMEL groups. Reduction in LPS plasma concentration was reported only in the HFDMEL group. Additionally, a decrease in LDL and an increase in HDL were observed in the HFDMEL and HFDAPMEL groups. Treatment with MEL exhibited anti-inflammatory and anti-hyperlipidemic effects attributed to HFD and AP by reducing the plasma concentrations of IL-1ß and LPS in addition to reducing IRF-3 protein expression in the GM, which is associated with the production of inflammatory cytokines.


Subject(s)
Melatonin , Periapical Periodontitis , Rats , Animals , NF-kappa B/metabolism , Toll-Like Receptor 4/metabolism , Lipopolysaccharides/pharmacology , Melatonin/pharmacology , Interleukin-1beta/metabolism , Myeloid Differentiation Factor 88/metabolism , Diet, High-Fat/adverse effects , Interferon Regulatory Factor-3/metabolism , Adaptor Proteins, Vesicular Transport/metabolism , Muscle, Skeletal/metabolism
7.
Int J Mol Sci ; 24(17)2023 Aug 30.
Article in English | MEDLINE | ID: mdl-37686267

ABSTRACT

Prenatal high-fat diet (HFD) or exposure to microplastics can affect the accumulation of liver fat in offspring. We sought to determine the effects of maternal HFD intake and microplastic exposure on fatty liver injury through oxidative stress in pups. Pregnant female Sprague-Dawley rats were randomly divided into maternal HFD (experimental group) or normal control diet (NCD; control group) groups with or without microplastic exposure. As a result, the following groups were established: HFD-L (HFD + microplastics, 5 µm, 100 µg/L), HFD-H (HFD + microplastics, 5 µm, 1000 µg/L), NCD-L (NCD + microplastics, 5 µm, 100 µg/L), and NCD-H (NCD + microplastics, 5 µm, 1000 µg/L). The pups were sacrificed on postnatal day 7 (PD7). Liver histology revealed increased hepatic lipid accumulation in pups in the HFD-L and HFD-H groups compared to those in the HFD, NCD-L, NCD-H, and NCD groups on PD7. Similarly, liver TUNEL staining and cellular apoptosis were found to increase in pups in the HFD-L and HFD-H groups compared to those in the HFD, NCD-L, NCD-H, and NCD groups. The expression levels of malondialdehyde, a lipid peroxidation marker, were high in the HFD, HFD-L, and HFD-H groups; however, the highest expression was observed in the HFD-H group (p < 0.05). The levels of glutathione peroxidase, an antioxidant enzyme, decreased in the HFD, HFD-L, and HFD-H groups (p < 0.05). Overall, oxidative stress with cellular apoptosis plays a vital role in liver injury in offspring after maternal intake of HFD and exposure to microplastic; such findings may shed light on future therapeutic strategies.


Subject(s)
Diet, High-Fat , Noncommunicable Diseases , Female , Male , Rats , Pregnancy , Animals , Rats, Sprague-Dawley , Diet, High-Fat/adverse effects , Microplastics , Plastics , Liver , Oxidative Stress , Vitamins
8.
Int J Mol Sci ; 24(9)2023 May 05.
Article in English | MEDLINE | ID: mdl-37176029

ABSTRACT

Clinical studies have shown that periodontitis is associated with non-alcoholic fatty liver disease (NAFLD). However, it remains unclear if periodontitis contributes to the progression of NAFLD. In this study, we generated a mouse model with high-fat diet (HFD)-induced metabolic syndrome (MetS) and NAFLD and oral P. gingivalis inoculation-induced periodontitis. Results showed that the presence of periodontitis increased insulin resistance and hepatic inflammation and exacerbated the progression of NAFLD. To determine the role of sphingolipid metabolism in the association between NAFLD and periodontitis, we also treated mice with imipramine, an inhibitor of acid sphingomyelinase (ASMase), and demonstrated that imipramine treatment significantly alleviated insulin resistance and hepatic inflammation, and improved NAFLD. Studies performed in vitro showed that lipopolysaccharide (LPS) and palmitic acid (PA), a major saturated fatty acid associated with MetS and NAFLD, synergistically increased the production of ceramide, a bioactive sphingolipid involved in NAFLD progression in macrophages but imipramine effectively reversed the ceramide production stimulated by LPS and PA. Taken together, this study showed for the first time that the presence of periodontitis contributed to the progression of NAFLD, likely due to alterations in sphingolipid metabolism that led to exacerbated insulin resistance and hepatic inflammation. This study also showed that targeting ASMase with imipramine improves NAFLD by reducing insulin resistance and hepatic inflammation.


Subject(s)
Insulin Resistance , Metabolic Syndrome , Non-alcoholic Fatty Liver Disease , Periodontitis , Mice , Animals , Non-alcoholic Fatty Liver Disease/complications , Non-alcoholic Fatty Liver Disease/metabolism , Metabolic Syndrome/complications , Metabolic Syndrome/metabolism , Liver/metabolism , Lipopolysaccharides/pharmacology , Imipramine/pharmacology , Periodontitis/complications , Periodontitis/metabolism , Palmitic Acid/pharmacology , Diet, High-Fat/adverse effects , Sphingolipids/metabolism , Ceramides/metabolism , Inflammation/metabolism , Mice, Inbred C57BL
9.
FASEB J ; 35(2): e21216, 2021 02.
Article in English | MEDLINE | ID: mdl-33230896

ABSTRACT

Obesity has emerged as a major risk factor for insulin resistance leading to the development of type 2 diabetes (T2D). The condition is characterized by high circulating levels of the adipose-derived hormone leptin and a state of chronic low-grade inflammation. Pro-inflammatory signaling in the hypothalamus is associated with a decrease of central leptin- and insulin action leading to impaired systemic glucose tolerance. Intriguingly, leptin not only regulates body weight and glucose homeostasis but also acts as a pro-inflammatory cytokine. Here we demonstrate that increasing leptin levels (62,5 µg/kg/d, PEGylated leptin) in mice fed a high-fat diet (HFD) exacerbated body weight gain and aggravated hypothalamic micro- as well as astrogliosis. In contrast, administration of a predetermined dose of a long-acting leptin antagonist (100 µg/kg/d, PESLAN) chosen to block excessive leptin signaling during diet-induced obesity (DIO) showed the opposite effect and significantly improved glucose tolerance as well as decreased the total number of microglia and astrocytes in the hypothalamus of mice fed HFD. These results suggest that high levels of leptin, such as in obesity, worsen HFD-induced micro-and astrogliosis, whereas the partial reduction of hyperleptinemia in DIO mice may have beneficial metabolic effects and improves hypothalamic gliosis.


Subject(s)
Glucose Intolerance/metabolism , Leptin/metabolism , Obesity/metabolism , Animals , Anti-Obesity Agents/pharmacology , Anti-Obesity Agents/therapeutic use , Diet, High-Fat/adverse effects , Gliosis/drug therapy , Gliosis/metabolism , Glucose Intolerance/drug therapy , Hypothalamus/metabolism , Hypothalamus/pathology , Leptin/analogs & derivatives , Leptin/antagonists & inhibitors , Male , Mice , Mice, Inbred C57BL , Obesity/drug therapy , Polyethylene Glycols/chemistry
10.
Environ Toxicol ; 37(2): 362-372, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34755918

ABSTRACT

Polystyrene nanoparticles (PS-NPs) as an issue of global environmental concern, have been shown to induce hepatic toxicity via triggering oxidative injury and inflammation. Non-alcoholic fatty liver disease (NAFLD) is initiated when excessive lipid is accumulated in the liver and will proceed to liver fibrosis with repeatedly chronic liver injury. In this study, we examined whether intravenous injection of PS-NPs could enhance the hepatic toxicity and potentiate the development of liver fibrosis in experimental high fat diet (HFD)-induced mice. The results demonstrated that PS-NPs could aggravate chronic hepatitis by interfere with liver lipid metabolism in HFD induced mice. Further, hepatic tissue in PS-NPs treated HFD mice displayed substantially lowered superoxide dismutase (SOD) activity, which confirming the oxidative stress induced by PS-NPs. PS-NPs exposure also resulted in the up-regulation of inflammation response in liver, as evidenced by the enhanced infiltration of Kupffer cells (KCs) and elevated expression of pro-inflammatory related indicators. Meanwhile, Masson trichrome staining revealed that PS-NPs could aggravate steatohepatitis with higher collagen fiber in HFD fed mice. Our data suggests that PS-NPs can induce oxidative stress and inflammation in HDF-induced experimental mice and further aggravate liver fibrosis, which highlight the potential health risks of PS-NPs.


Subject(s)
Diet, High-Fat , Non-alcoholic Fatty Liver Disease , Animals , Diet, High-Fat/adverse effects , Liver/metabolism , Liver Cirrhosis/metabolism , Mice , Mice, Inbred C57BL , Microplastics , Non-alcoholic Fatty Liver Disease/pathology , Oxidative Stress , Polystyrenes/toxicity
11.
Int J Mol Sci ; 23(24)2022 Dec 08.
Article in English | MEDLINE | ID: mdl-36555220

ABSTRACT

High-fat exposure leads to impaired intestinal barrier function by disrupting the function of intestinal stem cells (ISCs); however, the exact mechanism of this phenomenon is still not known. We hypothesize that high concentrations of deoxycholic acid (DCA) in response to a high-fat diet (HFD) affect aryl hydrocarbon receptor (AHR) signalling in ISCs and the intestinal barrier. For this purpose, C57BL/6J mice feeding on a low-fat diet (LFD), an HFD, an HFD with the bile acid binder cholestyramine, and a LFD with the DCA were studied. We found that high-fat feeding induced an increase in faecal DCA concentrations. An HFD or DCA diet disrupted the differentiation function of ISCs by downregulating AHR signalling, which resulted in decreased goblet cells (GCs) and MUC2, and these changes were reversed by cholestyramine. In vitro experiments showed that DCA downregulated the differentiation function of ISCs, which was reversed by the AHR agonist 6-formylindolo [3,2-b]carbazole (FICZ). Mechanistically, DCA caused a reduction in indoleamine 2,3-dioxygenase 1 (IDO1) in Paneth cells, resulting in paracrine deficiency of the AHR ligand kynurenine in crypts. We demonstrated for the first time that DCA disrupts intestinal mucosal barrier function by interfering with AHR signalling in ISCs. Supplementation with AHR ligands may be a new therapeutic target for HFD-related impaired intestinal barrier function.


Subject(s)
Cholestyramine Resin , Receptors, Aryl Hydrocarbon , Mice , Animals , Receptors, Aryl Hydrocarbon/metabolism , Mice, Inbred C57BL , Diet, High-Fat/adverse effects , Deoxycholic Acid/pharmacology , Stem Cells/metabolism
12.
Int J Mol Sci ; 23(11)2022 May 25.
Article in English | MEDLINE | ID: mdl-35682613

ABSTRACT

Bile acid metabolism, involved with the digestion and absorption of nutrients in the gut, is linked to the gut microbiota community, greatly impacting the host's metabolism. We examined the hypothesis that the modulation of bile acid metabolism by dietary fat contents, gallbladder removal (GBX; cholecystectomy), and bile acid sequestrant (BAS; cholestyramine) treatment could alter energy, glucose, and lipid metabolism through the changes in the gut microbiota. Mice were randomly assigned to the following six groups: (1) Sham GBX surgery (Sham) + low fat/high carbohydrate diet (LFD), (2) Sham + high fat diet (HFD), (3) Sham + HFD + BAS, (4) GBX + LFD, (5) GBX + HFD, and (6) GBX + HFD + BAS. BAS groups received 2% cholestyramine. After an 8-week intervention, energy, glucose, and lipid metabolism, and the gut microbiota community were measured. HFD groups exhibited higher body weight gain than LFD, and GBX increased the weight gain comped to Sham groups regardless of BAS in HFD (p < 0.05). Homeostatic model assessment for insulin resistance (HOMA-IR) was higher in HFD than LFD, and GBX increased it regardless of BAS. Serum lipid profiles were worsened in GBX + HFD compared to Sham + LFD, whereas BAS alleviated them, except for serum HDL cholesterol. Hepatic tumor-necrosis-factor-α (TNF-α) mRNA expression and lipid peroxide contents increased with GBX and BAS treatment compared to Sham and no BAS treatment (p < 0.05). Hepatic mRNA expression of sterol regulatory element-binding transcription factor 1c (SREBP1c) and peroxisome proliferator-activated receptor gamma (PPAR-γ) exhibited the same trend as that of tumor necrosis factor-α (TNF-α). The α-diversity of gut bacteria decreased in GBX + HFD and increased in GBX + HFD + BAS. Akkermentia, Dehalobacterium, SMB53, and Megamonas were high in the Sham + LFD, and Veillonella and Streptococcus were rich in the Sham + HFD, while Oscillospira and Olsenella were high in Sham + HFD + BAS (p < 0.05). GBX + LFD increased Lactobacillus and Sutterella while GBX + HFD + BAS elevated Clostridium, Alistipes, Blautia, Eubacterium, and Coprobacillus (p < 0.05). In conclusion, the modulation of bile acid metabolism influences energy, glucose, and lipid metabolisms, and it might be linked to changes in the gut microbiota by bile acid metabolism modulation.


Subject(s)
Dietary Fats , Gastrointestinal Microbiome , Animals , Bile Acids and Salts/metabolism , Cholecystectomy , Cholestyramine Resin/metabolism , Cholestyramine Resin/pharmacology , Diet, High-Fat/adverse effects , Dietary Fats/pharmacology , Glucose/metabolism , Hypolipidemic Agents/pharmacology , Lipid Metabolism , Liver/metabolism , Mice , Mice, Inbred C57BL , RNA, Messenger/metabolism , Tumor Necrosis Factor-alpha/metabolism , Weight Gain
13.
J Lipid Res ; 62: 100095, 2021.
Article in English | MEDLINE | ID: mdl-34214600

ABSTRACT

The pleiotropic actions of adiponectin in improving cell survival and metabolism have motivated the development of small-molecule therapeutic agents for treating diabetes and lipotoxicity. AdipoRon is a synthetic agonist of the adiponectin receptors, yet is limited by its poor solubility and bioavailability. In this work, we expand on the protective effects of AdipoRon in pancreatic ß-cells and examine how structural modifications could affect the activity, pharmacokinetics, and bioavailability of this small molecule. We describe a series of AdipoRon analogs containing amphiphilic ethylene glycol (PEG) chains. Among these, AdipoRonPEG5 induced pleiotropic effects in mice under insulinopenic and high-fat diet (HFD) conditions. While both AdipoRon and AdipoRonPEG5 substantially attenuate palmitate-induced lipotoxicity in INS-1 cells, only AdipoRonPEG5 treatment is accompanied by a significant reduction in cytotoxic ceramides. In vivo, AdipoRonPEG5 can substantially reduce pancreatic, hepatic, and serum ceramide species, with a concomitant increase in the corresponding sphingoid bases and improves insulin sensitivity of mice under HFD feeding conditions. Furthermore, hyperglycemia in streptozotocin (STZ)-induced insulinopenic adiponectin-null mice is also attenuated upon AdipoRonPEG5 treatment. Our results suggest that AdipoRonPEG5 is more effective in reducing ceramides and dihydroceramides in the liver of HFD-fed mice than AdipoRon, consistent with its potent activity in activating ceramidase in vitro in INS-1 cells. Additionally, these results indicate that the beneficial effects of AdipoRonPEG5 can be partially attributed to improved pharmacokinetics as compared with AdipoRon, thus suggesting that further derivatization may improve affinity and tissue-specific targeting.


Subject(s)
Diet, High-Fat/adverse effects , Glucose/metabolism , Lipid Metabolism/drug effects , Piperidines/pharmacology , Animals , Insulin Resistance , Liver/chemistry , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Piperidines/administration & dosage , Piperidines/chemistry , Polyethylene Glycols/chemistry
14.
J Biol Regul Homeost Agents ; 35(2): 641-655, 2021.
Article in English | MEDLINE | ID: mdl-33902274

ABSTRACT

Previous studies have shown that high-fat diet (HFD) may aggravate periodontitis, however the underlining mechanism remains to be further clarified. This study aims to explore whether HFD promotes periodontitis by inducing periodontal microbiota dysbiosis or stem cell dysfunction. A high-fat diet was given to four-week-old male Sprague-Dawley rats for 12 weeks. Periodontitis was induced during the latter 4 weeks. At the end of the 12th week, samples were collected after euthanasia. Maxillae were harvested for histological or microbial analysis. The microbial 16S rRNA gene sequencing was performed with the Illumina MiSeq platform. The data was analyzed through RDP Classifier against the SILVA database. The mandible molars were harvested for isolating periodontal ligament stem cells (PDLSCs). The protein level of p27, p21, and p16, which are negative regulators of the cell cycle, in PDLSCs were detected. Markers of osteogenic differentiation and pro-inflammatory mediators were detected by real-time polymerase chain reaction. Activation of pro-inflammatory signaling pathways was detected by Western blotting. We found that HFD significantly increased ligature-induced alveolar bone loss. HFD resulted in a less diverse periodontal microbiota, with increased proportions of Lactococcus, Bacillus, Alloprevotella, Carnobacterium, and Exiguobacterium and decreased proportion of Nitrospira. HFD increased the protein levels of p27, p16, and p21, and upregulated the expression of osteogenic biomarkers, IL-1ß and IL-10 with the ERK1/2 signaling pathway activated in PDLSCs.


Subject(s)
Osteogenesis , Periodontitis , Animals , Cell Differentiation , Diet, High-Fat/adverse effects , Dysbiosis , Male , Periodontal Ligament , Periodontitis/etiology , RNA, Ribosomal, 16S , Rats , Rats, Sprague-Dawley , Stem Cells
15.
J Clin Periodontol ; 48(8): 1137-1148, 2021 08.
Article in English | MEDLINE | ID: mdl-33998036

ABSTRACT

AIM: To characterize gingival metabolome in high-fat diet (HFD)-induced obesity in mice with/without periodontitis. METHODS: HFD-induced obesity mouse model was established by 16-week feeding, and a lean control group was fed with low-fat diet (n = 21/group). Both models were induced for periodontitis on the left sides by molar ligation for 10 days, whereas the right sides were used as controls. Gingival metabolome and arginine metabolism were analysed by non-targeted/targeted liquid chromatography-mass spectrometry. RESULTS: Of 2247 reference features, presence of periodontitis altered 165 in lean versus 885 in HFD mice; and HFD altered 525 in absence versus 1435 in presence of periodontitis. Compared with healthy condition, periodontitis and HFD had distinct effects on gingival metabolome. Metabolomic impacts of periodontitis were generally greater in HFD mice versus lean controls. K-medoids clustering showed that HFD amplified the impacts of periodontitis on gingival metabolome in both intensity and extensity. Ten metabolic pathways were enriched, including 2 specific to periodontitis, 5 specific to HFD and 3 shared ones. Targeted validation on arginine metabolism confirmed the additive effects between HFD and periodontitis. CONCLUSION: The obese population consuming excessive HFD display amplified metabolic response to periodontitis, presenting a metabolic susceptibility to exacerbated periodontal destruction.


Subject(s)
Diet, High-Fat , Periodontitis , Animals , Diet, High-Fat/adverse effects , Metabolome , Mice , Mice, Inbred C57BL , Obesity/complications , Periodontitis/etiology , Rodentia
16.
Oral Dis ; 27(5): 1243-1256, 2021 Jul.
Article in English | MEDLINE | ID: mdl-32989808

ABSTRACT

OBJECTIVES: The aim of the study was to investigate the effect of obesity on the tissue and molecular reactions of alveolar bone in response to orthodontic force and its underlying mechanisms. METHODS: Sixty-four rats were randomly divided into normal diet (ND) and high-fat diet (HFD) groups for eight weeks of dietary treatment. OTM was induced using nickel-titanium springs between the upper left first molar and incisor. After 1, 3, 7, and 14 days of OTM, the maxillary alveolar bone and gingival tissues were harvested and analyzed. RESULTS: Compared with the ND rats, the HFD rats had greater OTM distance, serum levels of tartrate-resistant acid phosphatase (TRAP), and tumor necrosis factor α (TNF-α), as well as significant alveolar bone loss and bone architecture deterioration on both the compression and tension sides (p < .05 for all). This response was linked to the increased osteoclast numbers and functional activity and decreased osteoblast activity in the periodontal ligament, gingival tissue, and alveolar bone. CONCLUSIONS: HFD-induced obesity promoted mechanically induced alveolar bone remodeling and detrimental changes in alveolar bone microstructure by increasing osteoclastogenesis and regulating inflammatory cytokine expression. The increased alveolar bone remodeling in the obese rats lead to an accelerated OTM.


Subject(s)
Diet, High-Fat , Tooth Movement Techniques , Animals , Bone Remodeling , Diet, High-Fat/adverse effects , Obesity/etiology , Osteoclasts , Rats
17.
Biochem Biophys Res Commun ; 533(1): 181-187, 2020 11 26.
Article in English | MEDLINE | ID: mdl-32958248

ABSTRACT

In recent years, the obese and overweight population has increased rapidly, which has become a worldwide public health problem. However, effective medication is lacking. Our previous study identified a novel peptide, PDBSN (GLSVADLAESIMKNL), that could significantly restrict adipocyte differentiation in vitro, but its in vivo function has not been determined. Thus, in this study, we encapsulated the peptide into liposomes attached with two ligands (visceral-adipose-tissue-targeting peptide and cell-penetrating peptide) to improve stability and specificity. We then tested the peptide's function in HFD (high-fat diet)-induced obese mice and found that PDBSN could reduce weight gain and improve insulin resistance as well as lipid homeostasis. These results suggest that PDBSN may be a potential candidate for anti-obesity drug discovery.


Subject(s)
Anti-Obesity Agents/therapeutic use , L-Lactate Dehydrogenase/therapeutic use , Lipid Metabolism/drug effects , Obesity/drug therapy , Peptide Fragments/therapeutic use , AMP-Activated Protein Kinases/metabolism , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animals , Anti-Obesity Agents/administration & dosage , Diet, High-Fat/adverse effects , Enzyme Activation/drug effects , Glucose/metabolism , Homeostasis/drug effects , L-Lactate Dehydrogenase/administration & dosage , Liposomes , Male , Mice, Inbred C57BL , Obesity/etiology , Obesity/metabolism , Peptide Fragments/administration & dosage
18.
Part Fibre Toxicol ; 17(1): 2, 2020 01 10.
Article in English | MEDLINE | ID: mdl-31924220

ABSTRACT

BACKGROUND: Previous studies have shown that inhalation of welding fumes may induce pulmonary and systemic inflammation and organ accumulation of metal, to which spermatogenesis and endocrine function may be sensitive. Also obesity may induce low-grade systemic inflammation. This study aimed to investigate the effects on sperm production of inhaled metal nanoparticles from stainless steel welding, and the potential exacerbation by intake of a high fat diet. Both the inbred Brown Norway and the outbred Sprague Dawley rat strains were included to study the influence of strain on the detection of toxicity. Rats were fed regular or high fat (HF) diet for 24 weeks and were exposed to 20 mg/m3 of gas metal arc-stainless steel (GMA-SS) welding fumes or filtered air for 3 h/day, 4 days/week for 5 weeks, during weeks 7-12. Outcomes were assessed upon termination of exposure (week 12) and after recovery (week 24). RESULTS: At week 12, the GMA-SS exposure induced pulmonary inflammation in both strains, without consistent changes in markers of systemic inflammation (CRP, MCP-1, IL-6 and TNFα). GMA-SS exposure lowered daily sperm production compared to air controls in Sprague Dawley rats, but only in GMA-SS Brown Norway rats also fed the HF diet. Overall, HF diet rats had lower serum testosterone levels compared to rats on regular diet. Metal content in the testes was assessed in a limited number of samples in Brown Norway rats, but no increase was obsedrved. At week 24, bronchoalveolar lavage cell counts had returned to background levels for GMA-SS exposed Sprague Dawley rats but remained elevated in Brown Norway rats. GMA-SS did not affect daily sperm production statistically significantly at this time point, but testicular weights were lowered in GMA-SS Sprague Dawley rats. Serum testosterone remained lowered in Sprague Dawley rats fed the HF diet. CONCLUSION: Exposure to GMA-SS welding fumes lowered sperm production in two strains of rats, whereas high fat diet lowered serum testosterone. The effect on sperm counts was likely not mediated by inflammation or lowered testosterone levels. The studied reproductive outcomes seemed more prone to disruption in the Sprague Dawley compared to the Brown Norway strain.


Subject(s)
Air Pollutants/toxicity , Diet, High-Fat/adverse effects , Inhalation Exposure/adverse effects , Spermatogenesis/drug effects , Testosterone/blood , Welding , Animals , Biomarkers/metabolism , Pneumonia/chemically induced , Pneumonia/metabolism , Rats, Sprague-Dawley , Species Specificity , Sperm Count , Stainless Steel
19.
Int J Mol Sci ; 21(20)2020 Oct 15.
Article in English | MEDLINE | ID: mdl-33076522

ABSTRACT

Current therapeutic options for obesity often require pharmacological intervention with dietary restrictions. Obesity is associated with underlying inflammation due to increased tissue macrophage infiltration, and recent evidence shows that inflammation can drive obesity, creating a feed forward mechanism. Therefore, targeting obesity-induced macrophage infiltration may be an effective way of treating obesity. Here, we developed cargo-less liposomes (UTS-001) using 1,2-dioleoyl-sn-glycero-3-phosphocholine, DOPC (synthetic phosphatidylcholine) as a single-agent to manage weight gain and related glucose disorders due to high fat diet (HFD) consumption in mice. UTS-001 displayed potent immunomodulatory properties, including reducing resident macrophage number in both fat and liver, downregulating liver markers involved in gluconeogenesis, and increasing marker involved in thermogenesis. As a result, UTS-001 significantly enhanced systemic glucose tolerance in vivo and insulin-stimulated cellular glucose uptake in vitro, as well as reducing fat accumulation upon ad libitum HFD consumption in mice. UTS-001 targets tissue residence macrophages to suppress tissue inflammation during HFD-induced obesity, resulting in improved weight control and glucose metabolism. Thus, UTS-001 represents a promising therapeutic strategy for body weight management and glycaemic control.


Subject(s)
Liposomes/therapeutic use , Obesity/drug therapy , Phosphatidylcholines/therapeutic use , 3T3-L1 Cells , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animals , Diet, High-Fat/adverse effects , Gluconeogenesis , Liposomes/chemistry , Liposomes/pharmacology , Liver/drug effects , Liver/metabolism , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Obesity/etiology , Phosphatidylcholines/chemistry , Phosphatidylcholines/pharmacology , Thermogenesis
20.
Bioconjug Chem ; 26(8): 1597-605, 2015 Aug 19.
Article in English | MEDLINE | ID: mdl-26086474

ABSTRACT

Regulation of cholesterol and bile acid homeostasis has been attracting attention as a pharmaceutical target for the treatment of diseases, such as hypercholesterolaemia and type 2 diabetes. In recent years, small bile acid analogues have been developed for the purpose of apical sodium-dependent bile acid transporter (ASBT) inhibition. Here, we designed a novel hydrophilic ASBT inhibitor using oligomeric bile acid with a high affinity with ASBT. Polyacrylic acid-tetraDOCA conjugates (PATD) have the ability to bind to ASBT in order to induce hypocholesterolemic effects. Both the viability and the functionality of PATD were evaluated in vitro, showing that PATDs were effective in inhibiting the increases of cholesterol in the blood and oil in the liver induced by high fat diet (HFD). The results indicated that the newly developed biomaterials with oligomeric bile acids and a hydrophilic polymer are potent therapeutic agents for hyperlipidemia.


Subject(s)
Acrylic Resins/chemistry , Anticholesteremic Agents/chemical synthesis , Anticholesteremic Agents/pharmacology , Drug Design , Hypercholesterolemia/drug therapy , Organic Anion Transporters, Sodium-Dependent/antagonists & inhibitors , Symporters/antagonists & inhibitors , Animals , Apoptosis/drug effects , Bile Acids and Salts/metabolism , Cell Proliferation/drug effects , Cholesterol/metabolism , Diet, High-Fat/adverse effects , Dogs , Glucose Tolerance Test , Hypercholesterolemia/etiology , Hypercholesterolemia/metabolism , Madin Darby Canine Kidney Cells , Male , Mice , Mice, Inbred C57BL , Molecular Dynamics Simulation , Organic Anion Transporters, Sodium-Dependent/metabolism , Symporters/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL