Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 181
Filter
1.
Lifestyle Genom ; 17(1): 82-92, 2024.
Article in English | MEDLINE | ID: mdl-38952113

ABSTRACT

INTRODUCTION: This study aims to investigate if a mixture of functional lipids (FLs), containing conjugated linoleic acid (CLA), tocopherols (TPs), and phytosterols (PSs), prevents some lipid alterations induced by high-fat (HF) diets, without adverse effects. METHODS: Male CF1 mice (n = 6/group) were fed (4 weeks) with control (C), HF, or HF + FL diets. RESULTS: FL prevented the overweight induced by the HF diet and reduced the adipose tissue (AT) weight, associated with lower energy efficiency. After the intervention period, the serum triacylglycerol (TAG) levels in both HF diets underwent a decrease associated with an enhanced LPL activity (mainly in muscle). The beneficial effect of the FL mixture on body weight gain and AT weight might be attributed to the decreased lipogenesis, denoted by the lower mRNA levels of SREBP1-c and ACC in AT, as well as by an exacerbated lipid catabolism, reflected by increased mRNA levels of PPARα, ATGL, HSL, and UCP2 in AT. Liver TAG levels were reduced in the HF + FL group due to an elevated lipid oxidation associated with a higher CPT-1 activity and mRNA levels of PPARα and CPT-1a. Moreover, genes linked to fatty acid biosynthesis (SREBP1-c and ACC) showed decreased mRNA levels in both HF diets, this finding being more pronounced in the HF + FL group. CONCLUSION: The administration of an FL mixture (CLA + TP + PS) prevented some lipid alterations induced by a HF diet, avoiding frequent deleterious effects of CLA in mice through the modulation of gene expression related to the regulation of lipid metabolism.


Subject(s)
Diet, High-Fat , Linoleic Acids, Conjugated , Lipid Metabolism , Liver , PPAR alpha , Sterol Regulatory Element Binding Protein 1 , Triglycerides , Animals , Diet, High-Fat/adverse effects , Mice , Male , Triglycerides/metabolism , Liver/metabolism , Liver/drug effects , Lipid Metabolism/drug effects , PPAR alpha/metabolism , PPAR alpha/genetics , Sterol Regulatory Element Binding Protein 1/metabolism , Sterol Regulatory Element Binding Protein 1/genetics , Linoleic Acids, Conjugated/pharmacology , Lipogenesis/drug effects , Carnitine O-Palmitoyltransferase/metabolism , Carnitine O-Palmitoyltransferase/genetics , Uncoupling Protein 2/metabolism , Uncoupling Protein 2/genetics , Phytosterols/pharmacology , Adipose Tissue/metabolism , Adipose Tissue/drug effects , Weight Gain/drug effects , Lipoprotein Lipase/metabolism , Lipoprotein Lipase/genetics
2.
Mol Cell Endocrinol ; 585: 112177, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38373652

ABSTRACT

AIM: To evaluate the effects of PPARα and PPARγ activation (alone or in combination) on the gut-liver axis, emphasizing the integrity of the intestinal barrier and hepatic steatosis in mice fed a high saturated fat diet. METHODS: Male C57BL/6J were fed a control diet (C) or a high-fat diet (HF) for ten weeks. Then, a four-week treatment started: HF-α (WY14643), HF-γ (low-dose pioglitazone), and HF-αγ (combination). RESULTS: The HF caused overweight, insulin resistance, impaired gut-liver axis, and marked hepatic steatosis. Treatments reduced body mass, improved glucose homeostasis, and restored the gut microbiota diversity and intestinal barrier gene expression. Treatments also lowered the plasma lipopolysaccharide concentrations and favored beta-oxidation genes, reducing macrophage infiltration and steatosis in the liver. CONCLUSION: Treatment with PPAR agonists modulated the gut microbiota and rescued the integrity of the intestinal barrier, alleviating hepatic steatosis. These results show that these agonists can contribute to metabolic-associated fatty liver disease treatment.


Subject(s)
Diet, High-Fat , Non-alcoholic Fatty Liver Disease , Male , Animals , Mice , Diet, High-Fat/adverse effects , PPAR alpha/genetics , PPAR alpha/metabolism , Obesity/metabolism , Mice, Inbred C57BL , Liver/metabolism , Non-alcoholic Fatty Liver Disease/metabolism
3.
Nutrition ; 117: 112253, 2024 01.
Article in English | MEDLINE | ID: mdl-37944411

ABSTRACT

OBJECTIVE: The aim of this study was to investigate the role of peroxisome proliferator-activated receptor (PPAR) activation (single PPARα or PPARγ, and dual PPARα/γ) on UCP1-dependent and -independent thermogenic pathways and mitochondrial metabolism in the subcutaneous white adipose tissue of mice fed a high-fat diet. METHODS: Male C57BL/6 mice received either a control diet (10% lipids) or a high-fat diet (HF; 50% lipids) for 12 wk. The HF group was divided to receive the treatments for 4 wk: HFγ (pioglitazone, 10 mg/kg), HFα (WY-14643, 3.5 mg/kg), and HFα/γ (tesaglitazar, 4 mg/kg). RESULTS: The HF group was overweight, insulin resistant, and had subcutaneous white adipocyte dysfunction. Treatment with PPARα and PPARα/γ reduced body mass, mitigated insulin resistance, and induced browning with increased UCP1-dependent and -independent thermogenesis activation and improved mitochondrial metabolism to support the beige adipocyte phenotype. CONCLUSION: PPARα and dual PPARα/γ activation recruited UCP1+ beige adipocytes and favored UCP1-independent thermogenesis, yielding body mass and insulin sensitivity normalization. Preserved mitochondrial metabolism emerges as a potential target for obesity treatment using PPAR agonists, with possible clinical applications.


Subject(s)
Adipocytes, Beige , Insulin Resistance , Animals , Male , Mice , Adipocytes, Beige/metabolism , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Diet, High-Fat/adverse effects , Lipids , Mice, Inbred C57BL , Mitochondrial Dynamics , PPAR alpha/metabolism , Thermogenesis , Uncoupling Protein 1/metabolism
4.
Acta Cir Bras ; 38: e387023, 2023.
Article in English | MEDLINE | ID: mdl-38055403

ABSTRACT

PURPOSE: Cerebral ischemia-reperfusion (I/R) is a neurovascular disorder that leads to brain injury. In mice, Fasudil improves nerve injury induced by I/R. However, it is unclear if this is mediated by increased peroxisome proliferator-activated receptor-α (PPARα) expression and reduced oxidative damage. This study aimed to investigate the neuroprotective mechanism of action of Fasudil. METHODS: MCAO (Middle cerebral artery occlusion) was performed in male C57BL/6J wild-type and PPARα KO mice between September 2021 to April 2023. Mice were treated with Fasudil and saline; 2,3,5-Triphenyltetrazolium chloride (TTC) staining was performed to analyze cerebral infarction. PPARα and Rho-associated protein kinase (ROCK) expression were detected using Western blot, and the expression of NADPH subunit Nox2 mRNA was detected using real-time polymerase chain reaction. The NADPH oxidase activity level and reactive oxygen species (ROS) content were also investigated. RESULTS: After cerebral ischemia, the volume of cerebral necrosis was reduced in wild-type mice treated with Fasudil. The expression of PPARα was increased, while ROCK was decreased. Nox2 mRNA expression, NADPH oxidase activity, and ROS content decreased. There were no significant changes in cerebral necrosis volumes, NADPH oxidase activity, and ROS content in the PPARα KO mice treated with Fasudil. CONCLUSIONS: In mice, the neuroprotective effect of Fasudil depends on the expression of PPARα induced by ROCK-PPARα-NOX axis-mediated reduction in ROS and associated oxidative damage.


Subject(s)
Brain Ischemia , Reperfusion Injury , Mice , Male , Animals , PPAR alpha/physiology , Reactive Oxygen Species/metabolism , Neuroprotection , Reperfusion Injury/drug therapy , Reperfusion Injury/prevention & control , Reperfusion Injury/genetics , Mice, Inbred C57BL , Ischemia , Brain Ischemia/drug therapy , Brain Ischemia/prevention & control , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Reperfusion , Necrosis , RNA, Messenger
5.
Food Res Int ; 173(Pt 1): 113252, 2023 11.
Article in English | MEDLINE | ID: mdl-37803565

ABSTRACT

Sorghum BRS 305 (Sorghum bicolor L. Moench) is a cereal with high tannins and anthocyanins content and keep better the resistant starch when submitted to dry heat treatment. Our objective was to investigate the effects of BRS 305 dry heat treatment whole sorghum flour on satiety and antioxidant response in brain and adipose tissue of Wistar rats fed with a high fat high fructose diet (HFHF). Male Wistar rats were divided in two groups: control (n = 8) and HFHF (n = 16) for eight weeks. After, animals of HFHF group were divided: HFHF (n = 8) and HFHF + BRS 305 sorghum whole flour (n = 8), for 10 weeks. Sorghum consumption reduced gene expression of leptin, resistin, and endocannabinoid receptor 1 type (CB1) in adipose and brain tissues compared to HFHF group. In brain, sorghum consumption also promotes reduction in neuropeptide Y (NPY) gene expression. BRS305 sorghum consumption improved gene expression of sirtuin-1 (SIRT1) in adipose tissue, and in the brain increased heat shock protein 72 (HSP72), erythroid-derived nuclear factor 2 (NRF2), peroxisome proliferator-activated receptor alpha (PPARα), superoxide dismutase (SOD) and catalase activity compared to HFHF. In silicoanalysis showed interaction with PPARα, CB1, and leptin receptors. Advanced glycation end products (AGEs) concentrations in group HFHF + sorghum did not differ from HFHF group. Advanced glycation end products receptors (RAGEs) concentrations did not differ among experimental groups. Then, BRS 305 sorghum submitted to dry treatment was able to modulate gene expression of markers related to satiety and improve antioxidant capacity of rats fed with HFHF diet.


Subject(s)
Antioxidants , Sorghum , Rats , Male , Animals , Rats, Wistar , Antioxidants/analysis , Sorghum/chemistry , Flour/analysis , Edible Grain/chemistry , Fructose/analysis , PPAR alpha , Anthocyanins/analysis , Diet, High-Fat/adverse effects , Brain , Glycation End Products, Advanced/analysis
6.
Ann Hepatol ; 28(3): 101082, 2023.
Article in English | MEDLINE | ID: mdl-36893888

ABSTRACT

INTRODUCTION AND OBJECTIVES: As a fatal clinical syndrome, acute liver failure (ALF) is characterized by overwhelming liver inflammation and hepatic cell death. Finding new therapeutic methods has been a challenge in ALF research. VX-765 is a known pyroptosis inhibitor and has been reported to prevent damage in a variety of diseases by reducing inflammation. However, the role of VX-765 in ALF is still unclear. MATERIALS AND METHODS: ALF model mice were treated with D-galactosamine (D-GalN) and lipopolysaccharide (LPS). LO2 cells were stimulated with LPS. Thirty subjects were enrolled in clinical experiments. The levels of inflammatory cytokines, pyroptosis-associated proteins and peroxisome proliferator-activated receptor α (PPARα) were detected using quantitative reverse transcription-polymerase chain reaction (qRT‒PCR), western blotting and immunohistochemistry. An automatic biochemical analyzer was used to determine the serum aminotransferase enzyme levels. Hematoxylin and eosin (HE) staining was used to observe the pathological features of the liver. RESULTS: With the progression of ALF, the expression levels of interleukin (IL) -1ß, IL-18, caspase-1, and serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) were increased. VX-765 could reduce the mortality rate of ALF mice, relieve liver pathological damage, and reduce inflammatory responses to protect against ALF. Further experiments showed that VX-765 could protect against ALF through PPARα, and this protective effect against ALF was reduced in the context of PPARα inhibition. CONCLUSIONS: As ALF progresses, inflammatory responses and pyroptosis deteriorate gradually. VX-765 can inhibit pyroptosis and reduce inflammatory responses to protect against ALF by upregulating PPARα expression, thus providing a possible therapeutic strategy for ALF.


Subject(s)
Liver Failure, Acute , PPAR alpha , Mice , Animals , PPAR alpha/genetics , PPAR alpha/metabolism , Pyroptosis , Lipopolysaccharides/metabolism , Lipopolysaccharides/pharmacology , Liver Failure, Acute/chemically induced , Liver Failure, Acute/prevention & control , Liver/pathology , Inflammation/prevention & control , Inflammation/metabolism , Tumor Necrosis Factor-alpha/metabolism , Mice, Inbred C57BL
7.
Int J Mol Sci ; 24(6)2023 Mar 10.
Article in English | MEDLINE | ID: mdl-36982395

ABSTRACT

Metabolic syndrome (MetS) is a cluster of factors that increase the risk of developing diabetes, stroke, and heart failure. The pathophysiology of injury by ischemia/reperfusion (I/R) is highly complex and the inflammatory condition plays an important role by increasing matrix remodeling and cardiac apoptosis. Natriuretic peptides (NPs) are cardiac hormones with numerous beneficial effects mainly mediated by a cell surface receptor named atrial natriuretic peptide receptor (ANPr). Although NPs are powerful clinical markers of cardiac failure, their role in I/R is still controversial. Peroxisome proliferator-activated receptor α agonists exert cardiovascular therapeutic actions; however, their effect on the NPs' signaling pathway has not been extensively studied. Our study provides important insight into the regulation of both ANP and ANPr in the hearts of MetS rats and their association with the inflammatory conditions caused by damage from I/R. Moreover, we show that pre-treatment with clofibrate was able to decrease the inflammatory response that, in turn, decreases myocardial fibrosis, the expression of metalloprotease 2 and apoptosis. Treatment with clofibrate is also associated with a decrease in ANP and ANPr expression.


Subject(s)
Metabolic Syndrome , Reperfusion Injury , Rats , Animals , Atrial Natriuretic Factor/metabolism , PPAR alpha/agonists , Clofibrate/pharmacology , Metabolic Syndrome/complications , Metabolic Syndrome/drug therapy , Reperfusion Injury/drug therapy , Reperfusion Injury/metabolism , Receptors, Atrial Natriuretic Factor/metabolism , Natriuretic Peptides , Ischemia , Arrhythmias, Cardiac , Inflammation/drug therapy
8.
Clin Transl Oncol ; 25(3): 601-610, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36348225

ABSTRACT

Obesity may create a mitogenic microenvironment that influences tumor initiation and progression. The obesity-associated adipokine, leptin regulates energy metabolism and has been implicated in cancer development. It has been shown that some cell types other than adipocytes can express leptin and leptin receptors in tumor microenvironments. It has been shown that peroxisome proliferator-activated receptors (PPAR) agonists can affect leptin levels and vice versa leptin can affect PPARs. Activation of PPARs affects the expression of several genes involved in aspects of lipid metabolism. In addition, PPARs regulate cancer cell progression through their action on the tumor cell proliferation, metabolism, and cellular environment. Some studies have shown an association between obesity and several types of cancer, including breast cancer. There is some evidence that suggests that there is crosstalk between PPARs and leptin during the development of breast cancer. Through a systematic review of previous studies, we have reviewed the published relevant articles regarding leptin signaling in breast cancer and its crosstalk with peroxisome proliferator-activated receptors α and γ.


Subject(s)
Breast Neoplasms , Peroxisome Proliferator-Activated Receptors , Humans , Female , Peroxisome Proliferator-Activated Receptors/agonists , Peroxisome Proliferator-Activated Receptors/metabolism , Leptin , PPAR alpha , Obesity , Signal Transduction , Tumor Microenvironment
9.
Mol Cell Endocrinol ; 562: 111839, 2023 02 15.
Article in English | MEDLINE | ID: mdl-36581062

ABSTRACT

AIM: To evaluate the effects of single PPARα or PPARγ activation, and their synergism (combined PPARα/γ activation) upon the gut-adipose tissue axis, focusing on the endotoxemia and upstream interscapular brown adipose tissue (iBAT) function in high-saturated fat-fed mice. METHODS: Male C57BL/6 mice received a control diet (C, 10% lipids) or a high-fat diet (HF, 50% lipids) for 12 weeks. Then, the HF group was divided to receive the treatments for four weeks: HFγ (pioglitazone, 10 mg/kg), HFα (WY-14643, 3.5 mg/kg), and HFα/γ (tesaglitazar, 4 mg/kg). RESULTS: The HF group exhibited overweight, oral glucose intolerance, gut dysbiosis, altered gut permeability, and endotoxemia, culminating in iBAT whitening. The downregulation of LPS-Tlr4 signaling underpinned reduced inflammation and improved lipid metabolism in iBAT in the HFα/γ group, the unique to show normalized body mass and increased energy expenditure. CONCLUSION: PPARα/γ synergism treated obesity by ameliorating the gut-adipose tissue axis, where restored gut microbiota and permeability controlled endotoxemia and rescued iBAT whitening through favored thermogenesis.


Subject(s)
Endotoxemia , PPAR alpha , Animals , Male , Mice , Adipose Tissue, Brown/metabolism , Diet, High-Fat , Lipids , Mice, Inbred C57BL , Obesity/metabolism , PPAR alpha/metabolism , PPAR gamma/metabolism
10.
Exp Physiol ; 107(12): 1454-1466, 2022 12.
Article in English | MEDLINE | ID: mdl-36114682

ABSTRACT

NEW FINDINGS: What is the central question of this study? What is the effect of an obesogenic diet on the control of hydromineral balance in rats? What is the main finding and its importance? The results showed that, when dehydrated, rats fed a high-fat diet drink less water than their control-diet-fed counterparts. Changes in aquaporin-7 and peroxisome proliferator-activated receptor α expression in the white adipose tissue might be involved. ABSTRACT: High-fat diet (HFD) increases fat accumulation, glycaemia and blood triglycerides and is used as a model to study obesity. Besides the metabolic changes, obesity likely affects water intake. We assessed the effects of HFD on behavioural and hormonal responses to water deprivation. Additionally, we measured if the adipose tissue is differentially affected by water deprivation in control and HFD-fed rats. HFD rats showed a decreased basal water intake when compared to control-fed rats. When subjected to 48 h of water deprivation, as expected, both control and HFD rats drank more water than the hydrated rats. However, the increase in water intake was lessened in HFD dehydrated rats. Similarly, the increase in haematocrit in dehydrated rats was less pronounced in HFD dehydrated rats. These results suggest that HFD diminishes drinking behaviour. White adipose tissue weight, glycaemia and plasma glycerol concentration were increased in HFD rats; however, after 48 h of water deprivation, these parameters were significantly decreased in dehydrated HFD rats, when compared to controls. The increase in adipose tissue caused by HFD may mitigate the effects of dehydration, possibly through the increased production of metabolic water caused by lipolysis in the adipocytes. Oxytocin possibly mediates the lipolytic response, since both its secretion and receptor expression are affected by dehydration in both control and HFD rats, which suggests that oxytocin signalling is maintained in these conditions. Changes in mediators of lipolysis, such as aquaporin-7 and peroxisome proliferator-activated receptor α, might contribute to the different effects observed in control and HFD rats.


Subject(s)
Dehydration , Diet, High-Fat , Rats , Male , Animals , Rats, Wistar , Water Deprivation , PPAR alpha , Oxytocin , Obesity/metabolism , Water
11.
J Appl Oral Sci ; 30: e20220076, 2022.
Article in English | MEDLINE | ID: mdl-35830121

ABSTRACT

OBJECTIVE: The purpose of this study is to investigate the pathogenic role of PPARα in periodontal antigen treated gingival cells in vitro and in experimental periodontitis in vivo . METHODOLOGY: Gingival fibroblasts, gingival epithelial cells and splenocytes were isolated from C57BL/6J wild type (WT) mice and treated with fixed P. gingivalis at for 48 hours. The mRNA levels of PPARs, TNFα, IL-1ß and IL-10 were detected by Real-time quantitative PCR. Silk ligatures after being soaked in the P.gingivalis suspension were tied around both maxillary second molars of WT mice or PPARα knock-out (KO) mice for two weeks. PPARα agonist fenofibrate and vehicle control were injected into the different side of the palatal gingiva on days 3, 6, and 9. At day 14, bone resorption and gingival mRNA expression levels of PPARs, TNFα, IL-1ß and IL-10 were measured by micro-computed tomography and RT-qPCR respectively. RESULTS: P. gingivalis treatment downregulated the expression of PPARα, but not PPARß or PPARγ, and increased the expression of TNF-α and IL-1ß in Gingival fibroblasts, gingival epithelial cells and splenocytes from WT mice. Gingival mRNA levels of PPARα were significantly decreased in experimental periodontitis in WT mice. The bone loss of PPARα KO mice in experimental periodontitis was significantly higher than WT mice and was not reduced by fenofibrate treatment. Gingival TNFα protein expressions were significantly increased by P. gingivalis associated ligation and decreased by fenofibrate treatment in WT mice but not in PPARα KO mice. CONCLUSION: This study suggests that PPARα plays an essential role in periodontitis.


Subject(s)
Alveolar Bone Loss , Fenofibrate , PPAR alpha , Periodontitis , Alveolar Bone Loss/pathology , Animals , Fenofibrate/pharmacology , Gingiva/pathology , Interleukin-10/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , PPAR alpha/metabolism , Periodontitis/metabolism , Periodontitis/pathology , Porphyromonas gingivalis/metabolism , RNA, Messenger/metabolism , Tumor Necrosis Factor-alpha/metabolism , X-Ray Microtomography
12.
An Acad Bras Cienc ; 94(2): e20210236, 2022.
Article in English | MEDLINE | ID: mdl-35703694

ABSTRACT

The objective of this study is to assess, in zebrafish, the effects of combining linseed oil (LO) and clove leaf essential oil (CLEO) on the incorporation of fatty acids in the muscle, oxidative markers, lipid peroxidation and expression of the PPAR-α (Peroxisome Proliferator-Activated Receptor-α) and the SREBP-2 (Sterol Regulatory Element Binding Protein-2) genes. Six diets were prepared, containing combinations of LO (3, 6 and 9%) and CLEO (0.5 and 1%): 3% LO + 0.5% CLEO; 3% LO + 1% CLEO; 6% LO + 0.5% CLEO; 6% LO + 1% CLEO; 9% LO + 0.5% CLEO; 9% LO + 1% CLEO. Results showed increase in the incorporation of n-3 fatty acids in the muscle concomitantly with the addition of LO and CLEO. The activities of superoxide dismutase and catalase were reduced and the glutathione content had increased. Lipid peroxidation was lower in the treatment with 1% CLEO, regardless of LO content. The expression of the PPAR-α and the SREBP-2 genes was higher in animals fed 9% LO + 0.5% CLEO. Therefore, for a greater incorporation and protection against the oxidative damages of n-3 fatty acids, a combined use of 9% LO with 0.5% CLEO is recommended for zebrafish.


Subject(s)
Fatty Acids, Omega-3 , Oils, Volatile , Syzygium , Animals , Fatty Acids/analysis , Fatty Acids, Omega-3/analysis , Fatty Acids, Omega-3/metabolism , Linseed Oil/chemistry , Linseed Oil/metabolism , Linseed Oil/pharmacology , Lipid Peroxidation , Liver/metabolism , Muscles/metabolism , Oils, Volatile/metabolism , Oxidative Stress , PPAR alpha/analysis , PPAR alpha/metabolism , Plant Leaves/metabolism , Sterol Regulatory Element Binding Protein 1/analysis , Sterol Regulatory Element Binding Protein 1/metabolism , Zebrafish/metabolism
13.
J Endocrinol ; 254(1): 37-49, 2022 06 08.
Article in English | MEDLINE | ID: mdl-35674008

ABSTRACT

The aim of this study was to evaluate the paternal programming of sex-dependent alterations in fetoplacental growth and placental lipid metabolism regulated by peroxisome proliferator-activated receptor (PPAR) target genes in F1 diabetic males born from F0 pregestational diabetic rats. F1 control and diabetic male rats were mated with control female rats. On day 21 of gestation, F2 male and female fetoplacental growth, placental lipid levels, and protein and mRNA levels of genes involved in lipid metabolism and transport were evaluated. Fetal but not placental weight was increased in the diabetic group. Triglyceride, cholesterol and free fatty acid levels were increased in placentas of male fetuses from the diabetic group. The mRNA levels of Pparα and Pparγ coactivator 1α (Pgc-1α) were increased only in placentas of male fetuses from the diabetic group. Protein levels of PPARα and PGC-1α were decreased only in placentas of male fetuses from the diabetic group. No differences were found in Pparγ mRNA and protein levels in placentas from the diabetic group. The mRNA levels of genes involved in lipid synthesis showed no differences between groups, whereas the mRNA levels of genes involved in lipid oxidation and transport were increased only in placentas of male fetuses from the diabetic group. In conclusion, paternal diabetes programs fetal overgrowth and sex-dependent effects on the regulation of lipid metabolism in the placenta, where only placentas of male fetuses show an increase in lipid accumulation and mRNA expression of enzymes involved in lipid oxidation and transport pathways.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes, Gestational , Animals , Diabetes Mellitus, Experimental/metabolism , Diabetes, Gestational/genetics , Diabetes, Gestational/metabolism , Female , Fetal Macrosomia/metabolism , Humans , Male , PPAR alpha/genetics , PPAR alpha/metabolism , PPAR gamma/genetics , PPAR gamma/metabolism , Placenta/metabolism , Pregnancy , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Triglycerides/metabolism
14.
Food Funct ; 13(11): 5996-6007, 2022 Jun 06.
Article in English | MEDLINE | ID: mdl-35575219

ABSTRACT

Aim: To investigate the effects of egg white hydrolysate (EWH) on the lipid and glycemic metabolism disruption in the white adipose tissue (WAT) dysfunction induced by mercury (Hg). Experimental: Wistar rats were treated for 60 days: control (saline, intramuscular - i.m.); hydrolysate (EWH, gavage, 1 g kg-1 day-1); mercury (HgCl2, i.m., 1st dose 4.6 µg kg-1, subsequent doses 0.07 µg kg-1 day-1) and hydrolysate-mercury (EWH-HgCl2). Hg level and histological analyses were performed in epididymal WAT (eWAT), pancreas and liver. GRP78, CHOP, PPARα, PPARγ, leptin, adiponectin, and CD11 mRNA expressions were analyzed in eWAT. The plasma lipid profile, glucose, and insulin levels were measured. Antioxidant status was also evaluated in the plasma and liver. Results: EWH intake prevented the reduced eWAT weight, adipocyte size, insulin levels, and antioxidant defenses and the increased glucose and triglyceride levels induced by Hg exposure; hepatic glutathione levels were higher in rats co-treated with EWH. The increased mRNA expression of CHOP, PPARα, and leptin induced by Hg was reduced in co-treated rats. EWH did not modify the elevated mRNA expression of GRP78, PPARγ and adiponectin in Hg-treated rats. Increased levels of Hg were found in the liver; the co-treatment did not alter this parameter. EWH prevented the morphological and metabolic disorder induced by Hg, by improving antioxidant defenses, inactivating pro-apoptotic pathways and normalizing the mRNA expression of PPARs and adipokines. Its effects enabled an increase in insulin levels and a normal balance between the fat storage and expenditure mechanisms in WAT. Conclusions: EWH may have potential benefits in the prevention and management of Hg-related metabolic disorders.


Subject(s)
Insulins , Mercury , Adiponectin/genetics , Adiponectin/metabolism , Adipose Tissue , Adipose Tissue, White/metabolism , Animals , Antioxidants/pharmacology , Egg White , Glucose/metabolism , Insulins/metabolism , Insulins/pharmacology , Leptin/metabolism , Lipids/pharmacology , Mercury/metabolism , Mercury/pharmacology , PPAR alpha/metabolism , PPAR gamma/genetics , PPAR gamma/metabolism , RNA, Messenger/metabolism , Rats , Rats, Wistar
15.
Nutrients ; 14(9)2022 May 09.
Article in English | MEDLINE | ID: mdl-35565950

ABSTRACT

In patients with age-related macular degeneration (AMD), the crucial retinal pigment epithelial (RPE) cells are characterized by mitochondria that are structurally and functionally defective. Moreover, deficient expression of the mRNA-editing enzyme Dicer is noted specifically in these cells. This Dicer deficit up-regulates expression of Alu RNA, which in turn damages mitochondria-inducing the loss of membrane potential, boosting oxidant generation, and causing mitochondrial DNA to translocate to the cytoplasmic region. The cytoplasmic mtDNA, in conjunction with induced oxidative stress, triggers a non-canonical pathway of NLRP3 inflammasome activation, leading to the production of interleukin-18 that acts in an autocrine manner to induce apoptotic death of RPE cells, thereby driving progression of dry AMD. It is proposed that measures which jointly up-regulate mitophagy and mitochondrial biogenesis (MB), by replacing damaged mitochondria with "healthy" new ones, may lessen the adverse impact of Alu RNA on RPE cells, enabling the prevention or control of dry AMD. An analysis of the molecular biology underlying mitophagy/MB and inflammasome activation suggests that nutraceuticals or drugs that can activate Sirt1, AMPK, Nrf2, and PPARα may be useful in this regard. These include ferulic acid, melatonin urolithin A and glucosamine (Sirt1), metformin and berberine (AMPK), lipoic acid and broccoli sprout extract (Nrf2), and fibrate drugs and astaxanthin (PPARα). Hence, nutraceutical regimens providing physiologically meaningful doses of several or all of the: ferulic acid, melatonin, glucosamine, berberine, lipoic acid, and astaxanthin, may have potential for control of dry AMD.


Subject(s)
Berberine , Macular Degeneration , Melatonin , Thioctic Acid , AMP-Activated Protein Kinases/metabolism , Berberine/pharmacology , DNA, Mitochondrial/metabolism , Dietary Supplements , Glucosamine , Humans , Inflammasomes/metabolism , Macular Degeneration/drug therapy , Melatonin/metabolism , Mitochondria/metabolism , Mitophagy , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Organelle Biogenesis , Oxidative Stress , PPAR alpha/metabolism , RNA/metabolism , Retinal Pigment Epithelium/metabolism , Sirtuin 1/metabolism
16.
Bioorg Med Chem Lett ; 70: 128804, 2022 08 15.
Article in English | MEDLINE | ID: mdl-35598791

ABSTRACT

In current work, we prepared a series of nine 4-benzyloxy-5-benzylidene-1,3-thiazolidine-2,4-diones using a two-step pathway. Compounds 1-9 were tested in vitro using a set of three proteins recognized as important targets in diabetes and related diseases: PPARα, PPARγ, and GLUT-4. Compounds 1-3, 5, and 7 showed significant increases in the mRNA expression of PPARγ and GLUT-4, whereas compounds 1-3 did it over PPARα. Compounds 1-3 were identified as a dual PPAR α/γ modulators and were selected for evaluating the in vivo antidiabetic action at 100 mg/kg dose, being orally actives and decreasing blood glucose concentration in a hyperglycemic mice model, as well as reducing the triacylglycerides levels in normolipidemic rats. Docking and molecular dynamics studies were conducted to clarify the dual effect and binding mode of compounds 1-3 on both PPARs. Compounds 2 and 3 exhibited robust in vitro and in vivo efficacy and could be considered dual PPAR modulators with antidiabetic and antidyslipidemic effects.


Subject(s)
Hypoglycemic Agents , PPAR gamma , Animals , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Lipids , Mice , PPAR alpha/metabolism , PPAR gamma/metabolism , Rats , Thiazolidines/pharmacology
17.
World J Gastroenterol ; 28(17): 1814-1829, 2022 May 07.
Article in English | MEDLINE | ID: mdl-35633911

ABSTRACT

BACKGROUND: Obesity and comorbidities onset encompass gut dysbiosis, altered intestinal permeability, and endotoxemia. Treatments that target gut dysbiosis can cope with obesity and nonalcoholic fatty liver disease (NAFLD) management. Peroxisome proliferator-activated receptor (PPAR)-alpha activation and dipeptidyl-peptidase-4 (DPP-4) inhibition alleviate NAFLD, but the mechanism may involve gut microbiota modulation and merits further investigation. AIM: To address the effects of PPAR-alpha activation and DPP-4 inhibition (isolated or combined) upon the gut-liver axis, emphasizing inflammatory pathways in NAFLD management in high-fat-fed C57BL/6J mice. METHODS: Male C57BL/6J mice were fed a control diet (C, 10% of energy as lipids) or a high-fat diet (HFD, 50% of energy as lipids) for 12 wk, when treatments started, forming the groups: C, HF, HFA (HFD + PPAR-alpha agonist WY14643, 2.5 mg/kg body mass), HFL (HFD + DPP-4 inhibitor linagliptin, 15 mg/kg body mass), and HFC (HFD + the combination of WY14643 and linagliptin). RESULTS: The HFD was obesogenic compared to the C diet. All treatments elicited significant body mass loss, and the HFC group showed similar body mass to the C group. All treatments tackled oral glucose intolerance and raised plasma glucagon-like peptide-1 concentrations. These metabolic benefits restored Bacteroidetes/Firmicutes ratio, resulting in increased goblet cells per area of the large intestine and reduced lipopolysaccharides concentrations in treated groups. At the gene level, treated groups showed higher intestinal Mucin 2, Occludin, and Zo-1 expression than the HFD group. The reduced endotoxemia suppressed inflammasome and macrophage gene expression in the liver of treated animals. These observations complied with the mitigation of liver steatosis and reduced hepatic triacylglycerol, reassuring the role of the proposed treatments on NAFLD mitigation. CONCLUSION: PPAR alpha activation and DPP-4 inhibition (isolated or combined) tackled NAFLD in diet-induced obese mice by restoration of gut-liver axis. The reestablishment of the intestinal barrier and the rescued phylogenetic gut bacteria distribution mitigated liver steatosis through anti-inflammatory signals. These results can cope with NAFLD management by providing pre-clinical evidence that drugs used to treat obesity comorbidities can help to alleviate this silent and harmful liver disease.


Subject(s)
Dipeptidyl-Peptidase IV Inhibitors , Endotoxemia , Non-alcoholic Fatty Liver Disease , Obesity , PPAR alpha , Animals , Dipeptidyl Peptidase 4/metabolism , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Dysbiosis/drug therapy , Dysbiosis/metabolism , Endotoxemia/complications , Endotoxemia/drug therapy , Linagliptin/pharmacology , Linagliptin/therapeutic use , Lipid Metabolism/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Non-alcoholic Fatty Liver Disease/metabolism , Obesity/complications , Obesity/drug therapy , Obesity/metabolism , PPAR alpha/agonists , PPAR alpha/metabolism , Phylogeny
18.
Eur Rev Med Pharmacol Sci ; 26(6): 2158-2164, 2022 03.
Article in English | MEDLINE | ID: mdl-35363365

ABSTRACT

OBJECTIVE: Peroxisome proliferator-activated receptor alpha (PPARα) is a nuclear transcription factor with a role in gene expression changes associated to lipid metabolism. PPARα polymorphic variants have been previously correlated to serum lipid profile but in Mexico, there is no previous report about that association. For this reason, the aim of this study was to investigate the relationship between PPARα polymorphic variants and lipids level in serum in a Mexican population. PATIENTS AND METHODS: Two-hundred and forty women from the Northeast region of Mexico were included in the study. Anthropometric characteristics and serum lipid profile (such as triglycerides, total cholesterol, LDL cholesterol and HDL cholesterol) were evaluated. Genomic DNA extraction and purification were made from blood samples. Real-time PCR and TaqMan probes were used for genotyping of rs1800206 and rs4253778 single nucleotide polymorphisms (SNPs). RESULTS: Linear regression analysis (adjusted by age and body mass index (BMI)) showed a significant statistical association of rs4253778 with total cholesterol (p=0.034) and LDL cholesterol (p=0.037). Any significant association was found between rs1800206 and lipid levels. CONCLUSIONS: These results suggested that rs4253778 (C allele) is associated with high levels of total cholesterol and LDL in a Mexican women population.


Subject(s)
PPAR alpha/genetics , Polymorphism, Single Nucleotide , Cholesterol, HDL , Cholesterol, LDL , Female , Genotype , Humans , Mexico/epidemiology , Triglycerides
19.
J Appl Oral Sci ; 30: e20210648, 2022.
Article in English | MEDLINE | ID: mdl-35293424

ABSTRACT

OBJECTIVE: Inflammation-related immune responses and bone metabolism lead to extensive tooth loss in periodontitis.. This study aims to investigate the effect of peroxisome proliferator-activated receptor (PPAR) alpha agonist anti-inflammatory treatment in vitro and in ligature-induced experimental periodontitis in vivo . METHODOLOGY: Splenocytes were isolated from C57BL/6J mice and cultured for 48 hours under the following conditions: control, P. gingivalis lipopolysaccharide (LPS) (1 µg/ml); experimental, LPS (1 µg/ml) + PPARα agonist (fenofibrate) at 1, 10, 50, 100 µM. MRNA and secreted protein levels of TNF-α expression were detected by RT-qPCR and ELISA, respectively. Silk ligatures (7-0) were tied around maxillary second molars of C57BL/6J mice for two weeks. Optimized doses of fenofibrate (50 µM) and vehicle control were injected into the contralateral side of the palatal gingiva on days three, six, and nine. At day 14, bone resorption, osteoclastogenesis, and gingival mRNA expression levels of TNF-α, IL-1ß, IL-6, and RANKL/OPG were measured by micro-computed tomography, Tartrate-resistant acid phosphatase (TRAP) staining, and Real-time quantitative PCR, respectively. RESULTS: TNF-α expression in cultured spleen cells were significantly increased in the presence of LPS, when compared with the control group, and significantly reduced by fenofibrate treatment in a dose-dependent manner from 1-100 µM (p<0.05). Gingival mRNA levels of TNF-α, IL-1ß, IL-6, and the ratio of RANKL/OPG, were significantly decreased after injection of fenofibrate, when compared to the control side (p<0.05). Periodontal bone loss and TRAP positive cell formation were significantly decreased on the side with an injection of fenofibrate, as compared to the control side (p<0.05). CONCLUSIONS: An anti-inflammatory treatment, PPARα agonist, inhibited inflammation and periodontal bone loss in ligature-induced experimental periodontitis.


Subject(s)
Alveolar Bone Loss , Periodontitis , Alveolar Bone Loss/drug therapy , Alveolar Bone Loss/prevention & control , Animals , Mice , Mice, Inbred C57BL , PPAR alpha/therapeutic use , Periodontitis/drug therapy , Periodontitis/metabolism , X-Ray Microtomography
20.
Autophagy ; 18(10): 2397-2408, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35220898

ABSTRACT

Mutations in the mitochondrial genome (mtDNA) are ubiquitous in humans and can lead to a broad spectrum of disorders. However, due to the presence of multiple mtDNA molecules in the cell, co-existence of mutant and wild-type mtDNAs (termed heteroplasmy) can mask disease phenotype unless a threshold of mutant molecules is reached. Importantly, the mutant mtDNA level can change across lifespan as mtDNA segregates in an allele- and cell-specific fashion, potentially leading to disease. Segregation of mtDNA is mainly evident in hepatic cells, resulting in an age-dependent increase of mtDNA variants, including non-synonymous potentially deleterious mutations. Here we modeled mtDNA segregation using a well-established heteroplasmic mouse line with mtDNA of NZB/BINJ and C57BL/6N origin on a C57BL/6N nuclear background. This mouse line showed a pronounced age-dependent NZB mtDNA accumulation in the liver, thus leading to enhanced respiration capacity per mtDNA molecule. Remarkably, liver-specific atg7 (autophagy related 7) knockout abolished NZB mtDNA accumulat ion, resulting in close-to-neutral mtDNA segregation through development into adulthood. prkn (parkin RBR E3 ubiquitin protein ligase) knockout also partially prevented NZB mtDNA accumulation in the liver, but to a lesser extent. Hence, we propose that age-related liver mtDNA segregation is a consequence of macroautophagic clearance of the less-fit mtDNA. Considering that NZB/BINJ and C57BL/6N mtDNAs have a level of divergence comparable to that between human Eurasian and African mtDNAs, these findings have potential implications for humans, including the safe use of mitochondrial replacement therapy.Abbreviations: Apob: apolipoprotein B; Atg1: autophagy-related 1; Atg7: autophagy related 7; Atp5a1: ATP synthase, H+ transporting, mitochondrial F1 complex, alpha subunit 1; BL6: C57BL/6N mouse strain; BNIP3: BCL2/adenovirus E1B interacting protein 3; FCCP: carbonyl cyanide 4-(trifluoromethoxy)phenylhydrazone; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; MAP1LC3A: microtubule-associated protein 1 light chain 3 alpha; MAP1LC3B: microtubule-associated protein 1 light chain 3 beta; mt-Atp8: mitochondrially encoded ATP synthase 8; MT-CO1: mitochondrially encoded cytochrome c oxidase I; MT-CO2: mitochondrially encoded cytochrome c oxidase II; mt-Co3: mitochondrially encoded cytochrome c oxidase III; mt-Cytb: mitochondrially encoded cytochrome b; mtDNA: mitochondrial DNA; MUL1: mitochondrial ubiquitin ligase activator of NFKB 1; nDNA: nuclear DNA; Ndufa9: NADH:ubiquinone oxireductase subunit A9; NDUFB8: NADH:ubiquinone oxireductase subunit B8; Nnt: nicotinamide nucleotide transhydrogenase; NZB: NZB/BINJ mouse strain; OXPHOS: oxidative phosphorylation; PINK1: PTEN induced putative kinase 1; Polg2: polymerase (DNA directed), gamma 2, accessory subunit; Ppara: peroxisome proliferator activated receptor alpha; Ppia: peptidylprolyl isomerase A; Prkn: parkin RBR E3 ubiquitin protein ligase; P10: post-natal day 10; P21: post-natal day 21; P100: post-natal day 100; qPCR: quantitative polymerase chain reaction; Rpl19: ribosomal protein L19; Rps18: ribosomal protein S18; SD: standard deviation; SEM: standard error of the mean; SDHB: succinate dehydrogenase complex, subunit B, iron sulfur (Ip); SQSTM1: sequestosome 1; Ssbp1: single-stranded DNA binding protein 1; TFAM: transcription factor A, mitochondrial; Tfb1m: transcription factor B1, mitochondrial; Tfb2m: transcription factor B2, mitochondrial; TOMM20: translocase of outer mitochondrial membrane 20; UQCRC2: ubiquinol cytochrome c reductase core protein 2; WT: wild-type.


Subject(s)
Mitophagy , NADP Transhydrogenases , Adenosine Triphosphate , Adult , Animals , Apolipoproteins/metabolism , Apolipoproteins B/metabolism , Autophagy/genetics , Carbon Dioxide/metabolism , Carbonyl Cyanide p-Trifluoromethoxyphenylhydrazone , Cytochromes b/metabolism , DNA, Mitochondrial/genetics , DNA-Binding Proteins/metabolism , Electron Transport Complex III , Electron Transport Complex IV/metabolism , Humans , Iron/metabolism , Liver/metabolism , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Mitochondrial Proteins , NAD/metabolism , NADP Transhydrogenases/metabolism , PPAR alpha/metabolism , Peptidylprolyl Isomerase/metabolism , Protein Kinases/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Ribosomal Proteins/metabolism , Sequestosome-1 Protein/metabolism , Succinate Dehydrogenase/metabolism , Sulfur/metabolism , Transcription Factors/metabolism , Ubiquinone , Ubiquitin-Protein Ligases/metabolism , Ubiquitins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL