Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 382
Filter
1.
Inflammopharmacology ; 31(3): 1361-1372, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36877411

ABSTRACT

Cyclophosphamide (CP) is a chemotherapeutic agent that causes pulmonary damage by generating free radicals and pro-inflammatory cytokines. Pulmonary damage has a high mortality rate due to the severe inflammation and edema occurred in lung. PPARγ/Sirt 1 signaling has been shown to be cytoprotective effect against cellular inflammatory stress and oxidative injury. Protocatechuic acid (PCA) is a potent Sirt1 activator and exhibits antioxidant as well as anti-inflammatory properties. The current study aims to investigate the therapeutic impacts of PCA against CP-induced pulmonary damage in rats. Rats were assigned randomly into 4 experimental groups. The control group was injected with a single i.p injection of saline. CP group was injected with a single i.p injection of CP (200 mg/kg). PCA groups were administered orally with PCA (50 and 100 mg/kg; p.o.) once daily for 10 consecutive days after CP injection. PCA treatment resulted in a significant decrease in the protein levels of MDA, a marker of lipid peroxidation, NO and MPO along with a significant increase in GSH and catalase protein levels. Moreover, PCA downregulated anti-inflammatory markers as IL-17, NF-κB, IKBKB, COX-2, TNF-α, and PKC and upregulated cytoprotective defenses as PPARγ, and SIRT1. In addition, PCA administration ameliorated FoxO-1 elevation, increased Nrf2 gene expression, and reduced air alveoli emphysema, bronchiolar epithelium hyperplasia and inflammatory cell infiltration induced by CP. PCA might represent a promising adjuvant to prevent pulmonary damage in patients receiving CP due to its antioxidant and anti-inflammatory effects with cytoprotective defenses.


Subject(s)
Antioxidants , Peroxisome Proliferators , Rats , Animals , Antioxidants/pharmacology , Antioxidants/metabolism , Peroxisome Proliferators/pharmacology , Sirtuin 1/metabolism , PPAR gamma/metabolism , Cyclophosphamide/adverse effects , Lung , Oxidative Stress , Anti-Inflammatory Agents/pharmacology
2.
Poult Sci ; 102(3): 102437, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36621096

ABSTRACT

The objective of the following study was to investigate the effects of naturally oxidized corn oil on the antioxidant capacity and lipid metabolism of broilers. A total of 450, 1-day-old Arbor Acres male broilers were randomly divided into 5 treatments with 6 replicate cages and 15 birds/cage. The dietary treatment array consisted of ratios of naturally oxidized corn oil to non-oxidized corn oil from 0:100, 25:75, 50:50, 75:25, and 100:0, respectively. Serum, liver, and abdominal fat samples were taken at 42 d. The results showed that the liver organ index, liver catalase (CAT) activity, malondialdehyde (MDA) content, and the serum aspartate aminotransferase (AST) content had significant quadratic relationships with the ratio of naturally oxidized corn oil (P < 0.05). Inflammatory infiltrating cells appeared in the liver of the 50% and 75% oxidized corn oil group. The percentage of abdominal fat, and serum free fatty acids (FFA) content increased linearly with the increased proportion of oxidized corn oil (P < 0.05). The mRNA expression of NADH quinone oxidoreductase 1 (NQO-1), nuclear factor kappa B (NF-κB), toll-like receptor-4 (TLR-4), peroxisome proliferators activate receptor-α (PPARα), carnitine acyltransferase (CPT1), and acyl-coenzyme oxidase (ACO) of the liver increased linearly while oxidized corn oil increased in the diet (P < 0.05). Diets containing 100% oxidized corn oil significantly changed the mRNA expression of liver Caveolin compared with other treatment groups (P < 0.05). Taken together, this study demonstrated that naturally oxidized corn oil could change liver lipid metabolism and accelerate lipid deposition of broilers by upregulating PPARα.


Subject(s)
Corn Oil , Peroxisome Proliferators , Male , Animals , Corn Oil/metabolism , Peroxisome Proliferators/metabolism , Peroxisome Proliferators/pharmacology , Lipid Metabolism , Chickens/physiology , PPAR alpha/genetics , PPAR alpha/metabolism , PPAR alpha/pharmacology , Diet/veterinary , Liver/metabolism , Antioxidants/metabolism , RNA, Messenger/metabolism , Dietary Supplements/analysis , Animal Feed/analysis , Clinical Trials, Veterinary as Topic
3.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 54(6): 1112-1120, 2023 Nov 20.
Article in Chinese | MEDLINE | ID: mdl-38162058

ABSTRACT

Objective: To investigate the renoprotective effects of a Sichuan dark tea-based medicated dietary formula (alternatively referred to as Qing, or clarity in Chinese) on mice with diet-induced obesity (DIO) and to explore the specific mechanisms involved. Methods: Male C57BL/6 mice were randomly assigned to three groups, a control group, a DIO group, and a Qing treatment group, or the Qing group, with 8 mice in each group. The mice in the control group were given normal maintenance feed and purified water, and the other two groups were fed a high-fat diet for 12 weeks to establish the DIO model. After that, high-fat diet continued in the DIO group, while the Qing group was given Qing at the same time for 12 weeks, during which period the weight of the mice was monitored and recorded every week. The mice were sacrificed after 12 weeks. Serum samples were collected and the levels of triglyceride (TG), total cholesterol (TC), alanine aminotransferase (ALT), aspartate aminotransferase (AST), and albumin were measured to evaluate liver function. In addition, renal lipids were extracted to determine the levels of TG and TC in the kidney and periodic acid-Schiff (PAS) and oil red O stainings were performed to evaluate kidney pathological injury. Western blot was performed to determine the phosphorylated AMPK (pAMPK)/AMPK ratio in the kidney tissue. RT-qPCR and Western blot were used to determine the expression of proteins related to fatty acid oxidation, including acetyl-CoA carboxylase 1 (ACC1), carnitine acyltransferase 1 (CTP1), peroxisome proliferators-activated receptor γ (PPARγ), peroxisome proliferators-activated receptor-1 α (PPAR1α), sterol-regulatory element binding proteins (SREBP-1), and key proteins related to lipid synthesis, including fatty acid synthase (FASN) and stearoyl-coenzyme A desaturase 1 (stearoyl-CoA desaturase) in the kidney tissue. 16SrRNA and metabolomics were applied to analyze the gut microbiota in the intestinal contents and its metabolites. Results: Compared with those of the control group, the levels of liver mass (P=0.0003), serum ALT (P<0.0001) and AST (P=0.0001), and kidney TC (P=0.0191) and TG (P=0.0101) of the DIO group were significantly increased and there was lipid deposition in the kidney. Compared with those of the DIO group, mice in the Qing group showed effective reduction in liver mass (P=0.0316) and improvements in the abnormal serum levels of AST (P=0.0012) and ALT (P=0.0027) and kidney TC (P=0.0200) and TG (P=0.0499). In addition, mice in the Qing group showed significant improvement in lipid deposition in the kidney. Qing group showed increased pAMPK/AMPK ratio in comparison with that of the DIO group. In comparison with those of the control group, mice in the DIO group had upregulated expression of lipid synthesis-related genes and proteins (SREBP-1, FASN, and SCD1). As for the fatty acid oxidation-related genes and proteins, DIO mice showed upregulated expression of ACC1 and downregulated expression of CPT1A, PPARγ, and PGC1α in comparison with those of the control group. In the Qing goup, improvements in regard to all these changes were observed. The Qing group demonstrated improvement in the disrupted homeostasis of the gut microbiota. Short-chain fatty acids in the cecal contents, especially isovaleric acid and propionic acid, were also restored. Conclusion: Sichuan dark tea-based medicated dietary formula may improve renal lipid metabolism by regulating gut microbiota and the levels of intestinal short-chain fatty acids, thereby protecting obesity-related kidney injury. Isovaleric acid and propionic acid may be the metabolites key to its regulation of gut microbiota.


Subject(s)
Gastrointestinal Microbiome , Lipid Metabolism Disorders , Male , Animals , Mice , Lipid Metabolism/genetics , Liver , Propionates/metabolism , Propionates/pharmacology , AMP-Activated Protein Kinases/metabolism , AMP-Activated Protein Kinases/pharmacology , Sterol Regulatory Element Binding Protein 1/metabolism , PPAR gamma/metabolism , PPAR gamma/pharmacology , Peroxisome Proliferators/metabolism , Peroxisome Proliferators/pharmacology , Mice, Inbred C57BL , Obesity/drug therapy , Diet, High-Fat/adverse effects , Lipid Metabolism Disorders/metabolism , Triglycerides , Tea/metabolism
4.
Biol Pharm Bull ; 45(10): 1458-1465, 2022.
Article in English | MEDLINE | ID: mdl-36184503

ABSTRACT

Obesity is currently the most common cause of metabolic diseases including type 2 diabetes and hyperlipidemia. Obesity results from excess lipid accumulation in adipose tissue. Several studies have investigated the inhibitory effects of natural plant-derived products on adipocyte differentiation and lipid accumulation. In this study, we examined the effect of hydrolysable tannins composed of gallic acid and glucose on adipocyte differentiation in 3T3-L1 cells. 1,2,3,4,6-Penta-O-galloyl-ß-D-glucose (PGG) (1), a representative gallotannin, inhibited lipid accumulation in 3T3-L1 cells, whereas ellagitannins (tellimagrandin I, eugeniin and casuarictin) did not. The expression of adipocyte differentiation-related genes, including peroxisome proliferator activator γ2 (Pparγ2), CCAAT/enhancer binding protein α (C/EBPα) and adipocyte fatty acid binding protein (aP2), was significantly suppressed in PGG (1)-treated 3T3-L1 cells beginning at day 2 after induction of differentiation. While PGG (1) did not directly reduce Pparγ2 expression, it reduced the expression of its target genes in mature adipocytes. In addition, PGG (1) treatment inhibited mitotic clonal expansion, one of earliest events of adipocyte differentiation. These findings indicate that PGG (1) has an inhibitory effect on adipocyte differentiation through the suppression of mitotic clonal expansion.


Subject(s)
Diabetes Mellitus, Type 2 , Hydrolyzable Tannins , 3T3-L1 Cells , Adipocytes , Adipogenesis , Animals , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Cell Differentiation , Diabetes Mellitus, Type 2/metabolism , Fatty Acid-Binding Proteins/genetics , Fatty Acid-Binding Proteins/metabolism , Fatty Acid-Binding Proteins/pharmacology , Gallic Acid/pharmacology , Glucose/metabolism , Hydrolyzable Tannins/metabolism , Hydrolyzable Tannins/pharmacology , Lipids , Mice , Obesity/metabolism , PPAR gamma/genetics , PPAR gamma/metabolism , Peroxisome Proliferators/metabolism , Peroxisome Proliferators/pharmacology
5.
Hum Exp Toxicol ; 41: 9603271221129852, 2022.
Article in English | MEDLINE | ID: mdl-36137816

ABSTRACT

Lipid metabolism is an important biochemical process in the body. Recent studies have found that environmental endocrine disruptors play an important role in the regulation of lipid metabolism. Bisphenol A (BPA), a common environmental endocrine disruptor, has adverse effects on lipid metabolism, but the mechanism is still unclear. This study aimed to investigate the effects of gestational BPA exposure on hepatic lipid metabolism and its possible mechanism in male offspring. The pregnant Sprague-Dawley rats were exposed to BPA (0, 0.05, 0.5, 5 mg/kg/day) from day 5 to day 19 of gestation to investigate the levels of triglyceride (TG) and total cholesterol (TC), and the expression of liver lipid metabolism-related genes in male offspring rats. The results showed that compared with the control group, the TG and TC levels in serum and liver in BPA-exposed groups was increased. And the expressions of liver fatty acid oxidation related genes, such as peroxisome proliferators-activated receptor α (PPARα) and carnitine palmitoyl transferase 1α (CPT1α), were down-regulated. However, the expressions of fatty acid synthesis related genes, such as sterol regulatory element binding proteins 1 (SREBP-1), acetyl-CoA carboxylase 1 (ACC1), fatty acid synthase (FAS) and stearoyl-CoA desaturase 1 (SCD-1), were up-regulated. The increased protein levels of mTOR and p-CRTC2 suggested that CREB-regulated transcription coactivator 2 (CRTC2) might be an important mediator in the mTOR/SREBP-1 pathway. In conclusion, these results demonstrated that mTOR/CRTC2/SREBP-1 could be affected by gestational BPA exposure, which may involve in the lipid metabolic disorders in later life.


Subject(s)
Endocrine Disruptors , Lipid Metabolism , Acetyl-CoA Carboxylase/metabolism , Acetyl-CoA Carboxylase/pharmacology , Animals , Benzhydryl Compounds , Carnitine/pharmacology , Cholesterol , Endocrine Disruptors/toxicity , Fatty Acid Synthases/metabolism , Fatty Acid Synthases/pharmacology , Fatty Acids/pharmacology , Female , Liver , Male , PPAR alpha/metabolism , Peroxisome Proliferators/metabolism , Peroxisome Proliferators/pharmacology , Phenols , Pregnancy , Rats , Rats, Sprague-Dawley , Stearoyl-CoA Desaturase/metabolism , Stearoyl-CoA Desaturase/pharmacology , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 1/metabolism , Sterol Regulatory Element Binding Protein 1/pharmacology , TOR Serine-Threonine Kinases/metabolism , Transferases/metabolism , Transferases/pharmacology , Triglycerides
6.
Food Funct ; 13(20): 10610-10622, 2022 Oct 17.
Article in English | MEDLINE | ID: mdl-36168843

ABSTRACT

Brassica rapa L., an edible, feeding and medicinal plant cultivated on the Tibetan plateau with altitudes above 3800 m, has several pharmacological effects. However, its therapeutic effects against memory impairment and central fatigue have yet to be conclusively established. In this study, the Y-maze and Morris water maze tasks revealed that Brassica rapa L. aqueous extract (BE) significantly ameliorated cognitive deficits of sleep deprivation (SD)-treated mice. Moreover, BE treatment partially alleviated SD-induced reductions in the levels of peripheral energy metabolism, and significantly decreased inflammatory factor levels in serum and hippocampus. In addition, BE treatment significantly relieved central fatigue and stabilized the excitability as well as activities of neurons by regulating the levels of hypothalamus tryptophan metabolites and striatum neurotransmitters. The neuroprotective effects of BE were also confirmed using glutamate-treated HT22 cells, whereby BE pretreatment significantly attenuated intracellular ROS production and mitochondrial depolarization via adenosine 5'-monophosphate activated protein kinase/peroxisome proliferators-activated receptors (AMPK/PPAR-γ) signaling pathways. Thus, BE might probably prevent SD-induced learning and memory deficits by inhibiting neuroinflammation and restoring mitochondrial energy metabolism in the hippocampus. These findings imply that BE is a potential complementary therapy for those suffering from deficient sleep or neurometabolic disorders, although this needs verification by prospective clinical studies.


Subject(s)
Brassica napus , Brassica rapa , Neuroprotective Agents , AMP-Activated Protein Kinases/metabolism , Adenosine/therapeutic use , Animals , Cognition , Fatigue/metabolism , Glutamates/metabolism , Hippocampus/metabolism , Maze Learning , Memory Disorders/drug therapy , Memory Disorders/metabolism , Memory Disorders/prevention & control , Mice , Neuroinflammatory Diseases , Neuroprotective Agents/pharmacology , Peroxisome Proliferator-Activated Receptors/metabolism , Peroxisome Proliferators/metabolism , Peroxisome Proliferators/pharmacology , Peroxisome Proliferators/therapeutic use , Prospective Studies , Reactive Oxygen Species/metabolism , Sleep Deprivation/complications , Sleep Deprivation/drug therapy , Sleep Deprivation/metabolism , Tibet , Tryptophan/metabolism
7.
J Biomed Sci ; 29(1): 75, 2022 Sep 29.
Article in English | MEDLINE | ID: mdl-36175886

ABSTRACT

BACKGROUND: The extra-intestinal effects of probiotics for preventing allergic diseases are well known. However, the probiotic components that interact with host target molecules and have a beneficial effect on allergic asthma remain unknown. Lactobacillus gasseri attenuates allergic airway inflammation through the activation of peroxisome proliferator- activated receptor γ (PPARγ) in dendritic cells. Therefore, we aimed to isolate and investigate the immunomodulatory effect of the PPARγ activation component from L. gasseri. METHODS: Culture supernatants of L. gasseri were fractionated and screened for the active component for allergic asthma. The isolated component was subjected to in vitro functional assays and then cloned. The crystal structure of this component protein was determined using X-ray crystallography. Intrarectal inoculation of the active component-overexpressing Clear coli (lipopolysaccharide-free Escherichia coli) and intraperitoneal injection of recombinant component protein were used in a house dust mite (HDM)-induced allergic asthma mouse model to investigate the protective effect. Recombinant mutant component proteins were assayed, and their structures were superimposed to identify the detailed mechanism of alleviating allergic inflammation. RESULTS: A moonlighting protein, glycolytic glyceraldehyde 3-phosphate dehydrogenase (GAPDH), LGp40, that has multifunctional effects was purified from cultured L. gasseri, and the crystal structure was determined. Both intrarectal inoculation of LGp40-overexpressing Clear coli and intraperitoneal administration of recombinant LGp40 protein attenuated allergic inflammation in a mouse model of allergic asthma. However, CDp40, GAPDH isolated from Clostridium difficile did not possess this anti-asthma effect. LGp40 redirected allergic M2 macrophages toward the M1 phenotype and impeded M2-prompted Th2 cell activation through glycolytic activity that induced immunometabolic changes. Recombinant mutant LGp40, without enzyme activity, showed no protective effect against HDM-induced airway inflammation. CONCLUSIONS: We found a novel mechanism of moonlighting LGp40 in the reversal of M2-prompted Th2 cell activation through glycolytic activity, which has an important immunoregulatory role in preventing allergic asthma. Our results provide a new strategy for probiotics application in alleviating allergic asthma.


Subject(s)
Asthma , Lactobacillus gasseri , Animals , Asthma/therapy , Disease Models, Animal , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Glyceraldehyde-3-Phosphate Dehydrogenases/pharmacology , Inflammation , Lung , Macrophages/metabolism , Mice , PPAR gamma/metabolism , Peroxisome Proliferators/metabolism , Peroxisome Proliferators/pharmacology , Pyroglyphidae
8.
Mol Cell Endocrinol ; 557: 111722, 2022 11 01.
Article in English | MEDLINE | ID: mdl-35917881

ABSTRACT

Peroxisome proliferator-activated receptor α (PPARα) is a key mediator of lipid metabolism and metabolic stress in the liver. A recent study revealed that PPARα-dependent long non-coding RNAs (lncRNAs) play an important role in modulating metabolic stress and inflammation in the livers of fasted mice. Here hepatic lncRNA 3930402G23Rik (G23Rik) was found to have active peroxisome proliferator response elements (PPREs) within its promoter and is directly regulated by PPARα. Although G23Rik RNA was expressed to varying degrees in several tissues, the PPARα-dependent regulation of this lncRNA was only observed in the liver. Pharmacological activation of PPARα induced PPARα recruitment at the G23Rik promoter and a pronounced increase in hepatic G23Rik lncRNA expression. A G23Rik-null mouse line was developed to further characterize the function of this lncRNA in the liver. G23Rik-null mice were more susceptible to hepatic lipid accumulation in response to acute fasting. Histological analysis further revealed a pronounced buildup of lipid droplets and a significant increase in neutral triglycerides and lipids as indicated by enhanced oil red O staining of liver sections. Hepatic cholesterol, non-esterified fatty acid, and triglyceride levels were significantly elevated in G23Rik-null mice and associated with induction of the lipid-metabolism related gene Cd36. These findings provide evidence for a lncRNA dependent mechanism by which PPARα attenuates hepatic lipid accumulation in response to metabolic stress through lncRNA G23Rik induction.


Subject(s)
Fasting , Lipid Metabolism , Liver , RNA, Long Noncoding , Animals , Fatty Acids, Nonesterified/metabolism , Fatty Acids, Nonesterified/pharmacology , Lipid Metabolism/genetics , Liver/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , PPAR alpha/genetics , PPAR alpha/metabolism , Peroxisome Proliferators/metabolism , Peroxisome Proliferators/pharmacology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Triglycerides/metabolism
9.
Neurotherapeutics ; 19(5): 1546-1565, 2022 09.
Article in English | MEDLINE | ID: mdl-35917087

ABSTRACT

Abnormal productions of amyloid beta (Aß) plaque and chronic neuroinflammation are commonly observed in the brain of patients with Alzheimer's disease, and both of which induce neuronal cell death, loss of memory, and cognitive dysfunction. However, many of the drugs targeting the production of Aß peptides have been unsuccessful in treating Alzheimer's disease. In this study, we identified synthetic novel peroxisome proliferator-activating receptor (PPAR) agonist, DTMB, which can ameliorate the chronic inflammation and Aß pathological progression of Alzheimer's disease. We discovered that DTMB attenuated the proinflammatory cytokine production of microglia by reducing the protein level of NF-κB. DTMB also improved the learning and memory defects and reduced the amount of Aß plaque in the brain of 5xFAD mice. This reduction in Aß pathology was attributed to the changes in gliosis and chronic inflammation level. Additionally, bulk RNA-sequencing showed that genes related to inflammation and cognitive function were changed in the hippocampus and cortex of DTMB-treated mice. Our findings demonstrate that DTMB has the potential to be a novel therapeutic agent for Alzheimer's disease.


Subject(s)
Alzheimer Disease , Receptors, Artificial , Mice , Animals , Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Microglia/metabolism , Amyloid beta-Peptides/metabolism , Peroxisome Proliferator-Activated Receptors/metabolism , Peroxisome Proliferator-Activated Receptors/pharmacology , Peroxisome Proliferator-Activated Receptors/therapeutic use , Mice, Transgenic , NF-kappa B/metabolism , Peroxisome Proliferators/metabolism , Peroxisome Proliferators/pharmacology , Peroxisome Proliferators/therapeutic use , Receptors, Artificial/metabolism , Receptors, Artificial/therapeutic use , Disease Models, Animal , Plaque, Amyloid/metabolism , Inflammation/drug therapy , Inflammation/metabolism , Cytokines/metabolism , RNA/metabolism , RNA/pharmacology , RNA/therapeutic use
10.
Ecotoxicol Environ Saf ; 231: 113173, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35007830

ABSTRACT

A growing body of evidence associated particulate matter (PM) exposure with lipid metabolism disorders, yet, the underlying mechanism remains to be elucidated. Among the major lipid metabolism modulators, peroxisome proliferator-activated receptor (PPAR) alpha plays an important role. In the current study, an individually ventilated cage (IVC) system was used to expose C57/B6 mice to real-ambient PM for six weeks, with or without co-treatment of PPAR alpha agonist WY14,643. The general parameters, liver and adipose tissue pathology, serum lipids, metal deposition and lipid profile of liver were assessed. The results indicated that six weeks of real-ambient PM exposure induced dyslipidemia, including increased serum triglycerides (TG) and decreased high density lipoprotein cholesterol (HDL-C) level, along with steatosis in liver, increased size of adipocytes in white adipose tissue (WAT) and whitening of brown adipose tissue (BAT). ICP-MS results indicated increased Cr and As deposition in liver. Lipidomics analysis revealed that glycerophospholipids and cytochrome P450 pathway were most significantly affected by PM exposure. Several lipid metabolism-related genes, including CYP4A14 in liver and UCP1 in BAT were downregulated following PM exposure. WY14,643 treatment alleviated PM-induced dyslipidemia, liver steatosis and whitening of BAT, while enhancing CD36, SLC27A1, CYP4A14 and UCP1 expression. In conclusion, PPAR alpha pathway participates in PM-induced lipid metabolism disorder, PPAR alpha agonist WY14,643 treatment exerted protective effects on PM-induced dyslipidemia, liver steatosis and whitening of BAT, but not on increased adipocyte size of WAT.


Subject(s)
Lipid Metabolism Disorders , PPAR alpha , Adipose Tissue, Brown/metabolism , Animals , Lipid Metabolism , Lipid Metabolism Disorders/metabolism , Mice , PPAR alpha/genetics , PPAR alpha/metabolism , Particulate Matter/metabolism , Peroxisome Proliferators/metabolism , Peroxisome Proliferators/pharmacology
11.
Theranostics ; 11(10): 4710-4727, 2021.
Article in English | MEDLINE | ID: mdl-33754023

ABSTRACT

Background: Telomere shortening and dysfunction may cause metabolic disorders, tissue damage and age-dependent pathologies. However, little is known about the association of telomere-associated protein Rap1 with mitochondrial energy metabolism and cardiac aging. Methods: Echocardiography was performed to detect cardiac structure and function in Rap1+/+ and Rap1-/- mice at different ages (3 months, 12 months and 20 months). Telomere length, DNA damage, cardiac senescence and cardiomyocyte size were analyzed using the real-time PCR, Western blotting, senescence associated ß-galactosidase assay and wheat germ agglutinin staining, respectively. Western blotting was also used to determine the level of cardiac fatty acid metabolism related key enzymes in mouse and human myocardium. Chromatin immunoprecipitation assay was used to verify the direct link between p53 and PPARα. The p53 inhibitor, Pifithrin-α and PPARα activator WY14643 were utilized to identify the effects of Rap1/p53/PPARα signaling pathway. Results: Telomere was shortened concomitant with extensive DNA damage in aged Rap1-/- mouse hearts, evidenced by reduced T/S ratios and increased nuclear γH2AX. Meanwhile, the aging-associated phenotypes were pronounced as reflected by altered mitochondrial ultrastructure, enhanced senescence, cardiac hypertrophy and dysfunction. Mechanistically, acetylated p53 and nuclear p53 was enhanced in the Rap1-/- mouse hearts, concomitant with reduced PPARα. Importantly, p53 directly binds to the promoter of PPARα in mouse hearts and suppresses the transcription of PPARα. In addition, aged Rap1-/- mice exhibited reduced cardiac fatty acid metabolism. Pifithrin-α alleviated cardiac aging and enhanced fatty acid metabolism in the aged Rap1-/- mice. Activating PPARα with WY14643 in primarily cultured Rap1-/- cardiomyocytes restored maximal oxygen consumption rates. Reduced Rap1 expression and impaired p53/PPARα signaling also presented in aged human myocardium. Conclusion: In summary, Rap1 may link telomere biology to fatty acid metabolism and aging-related cardiac pathologies via modulating the p53/PPARα signaling pathway, which could represent a therapeutic target in preventing/attenuating cardiac aging.


Subject(s)
Aging/genetics , Cardiomegaly/genetics , Cellular Senescence/genetics , Myocytes, Cardiac/metabolism , PPAR alpha/genetics , Telomere-Binding Proteins/genetics , Tumor Suppressor Protein p53/genetics , Animals , Benzothiazoles/pharmacology , Cardiomegaly/diagnostic imaging , Cardiomegaly/physiopathology , DNA Damage , Echocardiography , Fatty Acids/metabolism , Heart Diseases/diagnostic imaging , Heart Diseases/genetics , Heart Diseases/physiopathology , Histones/metabolism , Mice , Mice, Knockout , Mitochondria, Heart/metabolism , Mitochondria, Heart/ultrastructure , Open Field Test , PPAR alpha/metabolism , Peroxisome Proliferators/pharmacology , Pyrimidines/pharmacology , Shelterin Complex , Signal Transduction , Telomere/metabolism , Telomere Homeostasis , Telomere-Binding Proteins/metabolism , Toluene/analogs & derivatives , Toluene/pharmacology , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/metabolism
12.
Nat Commun ; 11(1): 5847, 2020 11 17.
Article in English | MEDLINE | ID: mdl-33203882

ABSTRACT

Exploring the molecular mechanisms that prevent inflammation during caloric restriction may yield promising therapeutic targets. During fasting, activation of the nuclear receptor peroxisome proliferator-activated receptor α (PPARα) promotes the utilization of lipids as an energy source. Herein, we show that ligand activation of PPARα directly upregulates the long non-coding RNA gene Gm15441 through PPARα binding sites within its promoter. Gm15441 expression suppresses its antisense transcript, encoding thioredoxin interacting protein (TXNIP). This, in turn, decreases TXNIP-stimulated NLR family pyrin domain containing 3 (NLRP3) inflammasome activation, caspase-1 (CASP1) cleavage, and proinflammatory interleukin 1ß (IL1B) maturation. Gm15441-null mice were developed and shown to be more susceptible to NLRP3 inflammasome activation and to exhibit elevated CASP1 and IL1B cleavage in response to PPARα agonism and fasting. These findings provide evidence for a mechanism by which PPARα attenuates hepatic inflammasome activation in response to metabolic stress through induction of lncRNA Gm15441.


Subject(s)
Inflammasomes/genetics , Liver/pathology , PPAR alpha/agonists , RNA, Long Noncoding/metabolism , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cells, Cultured , Fasting , Gene Expression Regulation , HEK293 Cells , Humans , Inflammasomes/metabolism , Liver/drug effects , Liver/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , PPAR alpha/genetics , PPAR alpha/metabolism , Peroxisome Proliferators/pharmacology , Promoter Regions, Genetic , Pyrimidines/pharmacology , RNA, Long Noncoding/genetics , Thioredoxins/genetics , Thioredoxins/metabolism
13.
Blood Adv ; 4(18): 4522-4537, 2020 09 22.
Article in English | MEDLINE | ID: mdl-32946570

ABSTRACT

Arterial thrombosis is the underlying cause for a number of cardiovascular-related events. Although dietary supplementation that includes polyunsaturated fatty acids (PUFAs) has been proposed to elicit cardiovascular protection, a mechanism for antithrombotic protection has not been well established. The current study sought to investigate whether an omega-6 essential fatty acid, docosapentaenoic acid (DPAn-6), and its oxidized lipid metabolites (oxylipins) provide direct cardiovascular protection through inhibition of platelet reactivity. Human and mouse blood and isolated platelets were treated with DPAn-6 and its 12-lipoxygenase (12-LOX)-derived oxylipins, 11-hydroxy-docosapentaenoic acid and 14-hydroxy-docosapentaenoic acid, to assess their ability to inhibit platelet activation. Pharmacological and genetic approaches were used to elucidate a role for DPA and its oxylipins in preventing platelet activation. DPAn-6 was found to be significantly increased in platelets following fatty acid supplementation, and it potently inhibited platelet activation through its 12-LOX-derived oxylipins. The inhibitory effects were selectively reversed through inhibition of the nuclear receptor peroxisome proliferator activator receptor-α (PPARα). PPARα binding was confirmed using a PPARα transcription reporter assay, as well as PPARα-/- mice. These approaches confirmed that selectivity of platelet inhibition was due to effects of DPA oxylipins acting through PPARα. Mice administered DPAn-6 or its oxylipins exhibited reduced thrombus formation following vessel injury, which was prevented in PPARα-/- mice. Hence, the current study demonstrates that DPAn-6 and its oxylipins potently and effectively inhibit platelet activation and thrombosis following a vascular injury. Platelet function is regulated, in part, through an oxylipin-induced PPARα-dependent manner, suggesting that targeting PPARα may represent an alternative strategy to treat thrombotic-related diseases.


Subject(s)
Arachidonate 12-Lipoxygenase , Blood Platelets , Animals , Arachidonate 12-Lipoxygenase/genetics , Arachidonate 12-Lipoxygenase/pharmacology , Lipids , Mice , PPAR alpha/genetics , PPAR alpha/pharmacology , Peroxisome Proliferators/pharmacology
14.
Int J Mol Sci ; 20(23)2019 Nov 25.
Article in English | MEDLINE | ID: mdl-31775380

ABSTRACT

Human cytochrome P450 1B1 (CYP1B1)-mediated biotransformation of endobiotics and xenobiotics plays an important role in the progression of human breast cancer. In this study, we investigated the effects of WY-14643, a peroxisome proliferator-activated receptor α (PPARα) agonist, on CYP1B1 expression and the related mechanism in MCF7 breast cancer cells. We performed quantitative reverse transcription-polymerase chain reaction, transient transfection, and chromatin immunoprecipitation to evaluate the effects of PPARα on peroxisome proliferator response element (PPRE)-mediated transcription. WY-14643 increased the protein and mRNA levels of CYP1B1, as well as promoter activity, in MCF-7 cells. Moreover, WY-14643 plus GW6471, a PPARα antagonist, significantly inhibited the WY-14643-mediated increase in CYP1B1 expression. PPARα knockdown by a small interfering RNA markedly suppressed the induction of CYP1B1 expression by WY-14643, suggesting that WY-14643 induces CYP1B1 expression via a PPARα-dependent mechanism. Bioinformatics analysis identified putative PPREs (-833/-813) within the promoter region of the CYP1B1 gene. Inactivation of these putative PPREs by deletion mutagenesis suppressed the WY-14643-mediated induction of CYP1B1 promoter activation. Furthermore, WY-14643 induced PPARα to assume a form capable of binding specifically to the PPRE-binding site in the CYP1B1 promoter. Our findings suggest that WY-14643 induces the expression of CYP1B1 through activation of PPARα.


Subject(s)
Breast Neoplasms/metabolism , Cytochrome P-450 CYP1B1/genetics , Gene Expression Regulation, Neoplastic/drug effects , PPAR alpha/metabolism , Peroxisome Proliferators/pharmacology , Pyrimidines/pharmacology , Signal Transduction/drug effects , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Proliferation , Cytochrome P-450 CYP1B1/metabolism , Female , Humans , PPAR alpha/genetics , Promoter Regions, Genetic , Response Elements , Tumor Cells, Cultured
15.
Am J Physiol Gastrointest Liver Physiol ; 312(3): G283-G299, 2017 Mar 01.
Article in English | MEDLINE | ID: mdl-28082284

ABSTRACT

Peroxisome proliferator-activated receptor-α (PPARA) is a nuclear transcription factor and key mediator of systemic lipid metabolism. Prolonged activation in rodents causes hepatocyte proliferation and hepatocellular carcinoma. Little is known about the contribution of nonparenchymal cells (NPCs) to PPARA-mediated cell proliferation. NPC contribution to PPARA agonist-induced hepatomegaly was assessed in hepatocyte (Ppara△Hep)- and macrophage (Ppara△Mac)-specific Ppara null mice. Mice were treated with the agonist Wy-14643 for 14 days, and response of conditional null mice was compared with conventional knockout mice (Ppara-/- ). Wy-14643 treatment caused weight loss and severe hepatomegaly in wild-type and Ppara△Mac mice, and histological analysis revealed characteristic hepatocyte swelling; Ppara△Hep and Ppara-/- mice were protected from these effects. Ppara△Mac serum chemistries, as well as aspartate aminotransferase and alanine aminotransferase levels, matched wild-type mice. Agonist-treated Ppara△Hep mice had elevated serum cholesterol, phospholipids, and triglycerides when compared with Ppara-/- mice, indicating a possible role for extrahepatic PPARA in regulating circulating lipid levels. BrdU labeling confirmed increased cell proliferation only in wild-type and Ppara△Mac mice. Macrophage PPARA disruption did not impact agonist-induced upregulation of lipid metabolism, cell proliferation, or DNA damage and repair-related gene expression, whereas gene expression was repressed in Ppara△Hep mice. Interestingly, downregulation of inflammatory cytokines IL-15 and IL-18 was dependent on macrophage PPARA. Cell type-specific regulation of target genes was confirmed in primary hepatocytes and Kupffer cells. These studies conclusively show that cell proliferation is mediated exclusively by PPARA activation in hepatocytes and that Kupffer cell PPARA has an important role in mediating the anti-inflammatory effects of PPARA agonists.


Subject(s)
Cell Proliferation/drug effects , Hepatocytes/metabolism , Kupffer Cells/metabolism , PPAR alpha/metabolism , Animals , Cholesterol/blood , Hepatocytes/drug effects , Kupffer Cells/drug effects , Lipid Metabolism/drug effects , Lipid Metabolism/physiology , Mice , Mice, Knockout , PPAR alpha/agonists , PPAR alpha/genetics , Peroxisome Proliferators/pharmacology , Pyrimidines/pharmacology , Weight Loss/drug effects , Weight Loss/physiology
16.
Brain Res Bull ; 127: 156-163, 2016 10.
Article in English | MEDLINE | ID: mdl-27646482

ABSTRACT

The peroxisome proliferator-activated receptor alpha (PPARα) is a member of the nuclear receptor superfamily that has been suggested as a modulator of several physiological functions. The PPARα recognizes as an endogenous ligand the anorexic lipid mediator oleoylethanolamide (OEA) which displays wake-inducing properties. Despite that recent evidence indicates that activation of PPARα by synthetic agonists such as Wy14643 enhances waking as well as the extracellular contents of wake-related neurotransmitters, the role of PPARα in sleep recovery after prolonged waking has not been fully described. Thus, the aim of this study was to characterize if PPARα regulates sleep rebound after total sleep deprivation (TSD). We report that after 6h of TSD activation of PPARα by pharmacological systemic administration of OEA (10, 20 or 30mg/Kg, i.p.) promoted alertness by blocking the sleep rebound after TSD. Besides, wake-linked compounds such as dopamine, norepinephrine, serotonin, or adenosine collected from nucleus accumbens were enhanced after TSD in OEA-treated animals. These sleep and neurochemical results were mimicked after injection of PPARα agonist Wy14643 (10, 20, 30mg/Kg, i.p.). However, similar findings from the sham of vehicle groups were observed if PPARα antagonist MK-886 was administered to rats (10, 20, 30mg/Kg, i.p.). Our results strengthened the hypothesis that PPARα might modulate sleep and neurochemical homeostasis after sleep deprivation.


Subject(s)
Homeostasis/drug effects , PPAR alpha/metabolism , Peroxisome Proliferators/pharmacology , Sleep/drug effects , Wakefulness-Promoting Agents/pharmacology , Adenosine/metabolism , Animals , Biogenic Monoamines/metabolism , Brain/drug effects , Brain/metabolism , Dose-Response Relationship, Drug , Endocannabinoids/pharmacology , Homeostasis/physiology , Indoles/pharmacology , Male , Oleic Acids/pharmacology , PPAR alpha/agonists , PPAR alpha/antagonists & inhibitors , Pyrimidines/pharmacology , Rats, Wistar , Sleep/physiology , Sleep Deprivation/metabolism , Wakefulness/drug effects , Wakefulness/physiology
17.
BMC Evol Biol ; 16(1): 147, 2016 Jul 16.
Article in English | MEDLINE | ID: mdl-27421266

ABSTRACT

BACKGROUND: In the duplication-degeneration-complementation (DDC) model, a duplicated gene has three possible fates: it may lose functionality through the accumulation of mutations (nonfunctionalization), acquire a new function (neofunctionalization), or each duplicate gene may retain a subset of functions of the ancestral gene (subfunctionalization). The role that promoter evolution plays in retention of duplicated genes in eukaryotic genomes is not well understood. Fatty acid-binding proteins (Fabp) belong to a multigene family that are highly conserved in sequence and function, but differ in their gene regulation, suggesting selective pressure is exerted via regulatory elements in the promoter. RESULTS: In this study, we describe the PPAR regulation of zebrafish fabp1a, fabp1b.1, and fabp1b.2 promoters and compare them to the PPAR regulation of the spotted gar fabp1 promoter, representative of the ancestral fabp1 gene. Evolution of the fabp1 promoter was inferred by sequence analysis, and differential PPAR-agonist activation of fabp1 promoter activity in zebrafish liver and intestine explant cells, and in HEK293A cells transiently transfected with wild-type and mutated fabp1promoter-reporter gene constructs. The promoter activity of spotted gar fabp1, representative of the ancestral fabp1, was induced by both PPARα- and PPARγ-specific agonists, but displayed a biphasic response to PPARα activation. Zebrafish fabp1a was PPARα-selective, fabp1b.1 was PPARγ-selective, and fabp1b.2 was not regulated by PPAR. CONCLUSIONS: The zebrafish fabp1 promoters underwent two successive rounds of subfunctionalization with respect to PPAR regulation leading to retention of three zebrafish fabp1 genes with stimuli-specific regulation. Using a pharmacological approach, we demonstrated here the divergent regulation of the zebrafish fabp1a, fabp1b.1, and fabp1b.2 with regard to subfunctionalization of PPAR regulation following two rounds of gene duplication.


Subject(s)
Fatty Acid-Binding Proteins/genetics , Gene Duplication , Genes, Duplicate , Peroxisome Proliferators , Zebrafish Proteins/genetics , Animals , Gene Expression Regulation , Humans , Mutation , PPAR alpha/genetics , Peroxisome Proliferators/pharmacology , Promoter Regions, Genetic , Response Elements , Zebrafish
18.
Wei Sheng Wu Xue Bao ; 56(10): 1606-15, 2016 Oct 04.
Article in Chinese | MEDLINE | ID: mdl-29741349

ABSTRACT

Objective: To study the effect of peroxisome proliferations (PPs) on the development and pathogenicity of rice blast fungus Magnaporthe oryzae. Methods: The peroxisomal proliferation and the expression of peroxisomal biogenesis related genes were detected in M. oryzae strain Guy11 under the induction of 6 PPs. Vegetative growth, conidial germination, appressoria formation and pathogenicity of the strain treated with PPs were compared with those of the control. Results: Induced by 6 PPs, the quantity of peroxisome and the expression of PEX14, PEX8 and PEX11 were significantly increased. Vegetative growth, conidial germination, appressorial formation and pathogenicity were inhibited by the majority of the PPs. Of them, 2, 4-D and aspirin (ASA) exhibited higher inhibition rates than others. Further, the inhibition of 2, 4-D and Aspirin to the vegetative growth of Δpex5 and Δpex7 mutants of M. oryzae was found significantly increased than that of the wild type strain. Conclusion: PPs could induce peroxisome proliferation in M. oryzae, inhibit the growth and development and reduce the pathogenicity of the fungus. This is the first investigation on the effects of PPs to filamentous fungi.


Subject(s)
Magnaporthe/drug effects , Magnaporthe/growth & development , Peroxisome Proliferators/pharmacology , Plant Diseases/microbiology , Fungal Proteins/genetics , Fungal Proteins/metabolism , Magnaporthe/genetics , Magnaporthe/pathogenicity , Oryza/microbiology , Peroxisomes/drug effects , Peroxisomes/genetics , Peroxisomes/metabolism , Spores, Fungal/drug effects , Spores, Fungal/genetics , Spores, Fungal/growth & development , Virulence/drug effects
19.
Mol Nutr Food Res ; 59(8): 1573-83, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25913736

ABSTRACT

SCOPE: Omega-3 polyunsaturated fatty acids (n-3 PUFA) found in fish oil activate PPAR-α, stimulate peroxisomal fatty acid (FA) ß-oxidation and prevent impairments on glucose homeostasis. METHODS AND RESULTS: Glucose metabolism and FA oxidation were studied in C57/Bl6 mice fed with diets containing either 3.6 and 31.5% fish oil or lard. To assess the effects of peroxisomal proliferation on FA oxidation independent of n-3 PUFA intake, mice were treated with the PPAR-α agonist WY-14643. n-3 PUFA-fed mice were protected from glucose intolerance and dyslipidemia compared to animals fed a lard-based high-fat diet. Most importantly, mice fed on the hyperlipidic diet based on fish oil as well as the WY-14643 treated mice showed twofold increase of odd, medium-chain, dicarboxylic acylcarnitines in the liver suggesting that not only ß-oxidation, but also α- and ω-oxidation of FA were increased. Finally, an oxidation assay using liver homogenates and palmitic acid as substrate revealed an over tenfold increased production of similar acylcarnitines, indicating that FA are their precursors. CONCLUSION: This study shows at the metabolite level that peroxisome proliferation induced either by fish oil or WY-14643 is associated with increased α- and ω-oxidation of FA producing specific acylcarnitines that can be utilized as biomarkers of peroxisomal FA oxidation.


Subject(s)
Carnitine/analogs & derivatives , Diet, High-Fat/adverse effects , Dietary Fats, Unsaturated/metabolism , Fatty Acids, Omega-3/metabolism , Liver/metabolism , Overweight/metabolism , Peroxisomes/metabolism , Animals , Biomarkers/chemistry , Biomarkers/metabolism , Carnitine/chemistry , Carnitine/metabolism , Dietary Fats/adverse effects , Dietary Fats, Unsaturated/adverse effects , Dietary Fats, Unsaturated/therapeutic use , Fatty Acids, Omega-3/therapeutic use , Fish Oils/therapeutic use , Gene Expression Regulation/drug effects , Glucose Intolerance/etiology , Glucose Intolerance/prevention & control , Hyperlipidemias/etiology , Hyperlipidemias/prevention & control , Liver/drug effects , Liver/enzymology , Male , Mice, Inbred C57BL , Molecular Weight , Overweight/etiology , Overweight/physiopathology , Overweight/prevention & control , Oxidation-Reduction , Peroxisome Proliferators/pharmacology , Peroxisomes/drug effects , Peroxisomes/enzymology , Pyrimidines/pharmacology
20.
Biochem J ; 467(3): 453-60, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25695641

ABSTRACT

Insulin-like growth factor (IGF)-binding protein-2 (IGFBP-2), one of the most abundant circulating IGFBPs, is known to attenuate the biological action of IGF-1. Although the effect of IGFBP-2 in preventing metabolic disorders is well known, its regulatory mechanism remains unclear. In the present study, we demonstrated the transcriptional regulation of the Igfbp-2 gene by peroxisome-proliferator-activated receptor (PPAR) α in the liver. During fasting, both Igfbp-2 and PPARα expression levels were increased. Wy14643, a selective PPARα agonist, significantly induced Igfbp-2 gene expression in primary cultured hepatocytes. However, Igfbp-2 gene expression in Pparα null mice was not affected by fasting or Wy14643. In addition, through transient transfection and chromatin immunoprecipitation assay in fasted livers, we determined that PPARα bound to the putative PPAR-responsive element between -511 bp and -499 bp on the Igfbp-2 gene promoter, indicating that the Igfbp-2 gene transcription is activated directly by PPARα. To explore the role of PPARα in IGF-1 signalling, we treated primary cultured hepatocytes with Wy14643 and observed a decrease in the number of IGF-1 receptors (IGF-1Rs) and in Akt phosphorylation. No inhibition was observed in the hepatocytes isolated from Pparα null mice. These results suggest that PPARα controls IGF-1 signalling through the up-regulation of hepatic Igfbp-2 transcription during fasting and Wy14643 treatment.


Subject(s)
Fasting/metabolism , Insulin-Like Growth Factor Binding Protein 2/genetics , Animals , Cells, Cultured , Gene Expression Regulation/drug effects , Hepatocytes/drug effects , Hepatocytes/metabolism , Insulin-Like Growth Factor I/metabolism , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , PPAR alpha/deficiency , PPAR alpha/genetics , PPAR gamma/agonists , Peroxisome Proliferators/pharmacology , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Pyrimidines/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rosiglitazone , Signal Transduction , Thiazolidinediones/pharmacology , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...