Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
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
2.
Mol Nutr Food Res ; 68(6): e2300634, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38402434

ABSTRACT

The discovery of metabolically active brown adipose tissue (BAT) in human adults and the worldwide increase in obesity and obesity-related chronic noncommunicable diseases (NCDs) has made BAT a therapeutic target in the last two decades. The potential of BAT to oxidize fatty acids rapidly and increase energy expenditure inversely correlates with adiposity, insulin and glucose resistance, and cardiovascular and metabolic diseases. Currently, BAT is recognized by a new molecular signature; several BAT-derived molecules that act positively on target tissues have been identified and collectively called batokines. Bioactive compounds present in foods are endowed with thermogenic properties that increase BAT activation signaling. Understanding the mechanisms that lead to BAT activation and the batokines secreted by it within the thermogenic state is fundamental for its recruitment and management of obesity and NCDs. This review contributes to recent updates on the morphophysiology of BAT, its endocrine role in obesity, and the main bioactive compounds present in foods involved in classical and nonclassical thermogenic pathways activation.


Subject(s)
Adipose Tissue, Brown , Obesity , Humans , Adipose Tissue, Brown/metabolism , Obesity/metabolism , Energy Metabolism , Glucose/metabolism , Signal Transduction , Thermogenesis , Adipocytes, Brown/metabolism
3.
J Am Nutr Assoc ; 43(2): 201-212, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37611162

ABSTRACT

Introduction: Chronic kidney disease (CKD) promotes gut dysbiosis, and enteric glial reactivity, a feature of intestinal inflammation. Brazil nut modulated enteric glial profile in healthy animals and could modulate these cells in 5/6 nephrectomized rats.Methods: A 5/6 nephrectomy-induced CKD and Sham-operated rats were divided as follows: CKD and Sham received a standard diet and CKD-BN and Sham-BN received a 5% Brazil nut enriched-diet. The protein content of glial fibrillary acid protein (GFAP), enteric glial marker, and GPx protein content and activity were assessed in the colon. The major phyla of gut microbiota were assessed.Results: CKD-BN group presented a decrease in GFAP content (p = 0.0001). The CKD-BN group modulated the abundance of Firmicutes, increasing its proportion compared to the CKD group. The CKD-BN group showed increased GPx activity in the colon (p = 0.0192), despite no significant difference in protein content.Conclusion: Brazil nut-enriched diet consumption decreased enteric glial reactivity and modulated gut microbiota in the CKD experimental model.


Subject(s)
Bertholletia , Gastrointestinal Microbiome , Renal Insufficiency, Chronic , Rats , Animals , Diet , Neuroglia/metabolism , Renal Insufficiency, Chronic/metabolism
4.
Nutrition ; 117: 112253, 2024 Jan.
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
5.
J Nutr Biochem ; 117: 109336, 2023 07.
Article in English | MEDLINE | ID: mdl-36990367

ABSTRACT

This study aimed to evaluate the preventive and therapeutic effects of coffee consumption on molecular changes and adipose tissue remodeling in a murine model of high-fat diet-induced obesity. Three-month-old C57BL/6 mice were initially divided into three groups, namely, control (C), high-fat (HF), and coffee prevention (HF-CP) groups, and the HF group was subdivided at the end of the 10th week into two subgroups, an HF group and a coffee treatment (HF-CT) group; thus, a total of four groups were investigated at the 14th week of the experiment. The HF-CP group had lower body mass than the HF group (-7%, P < .05) and a better distribution of adipose tissue. Both groups that received coffee (HF-CP and HF-CT) showed improved glucose metabolism compared with the HF group. Coffee consumption also attenuated adipose tissue inflammation and showed decreased macrophage infiltration and lower IL-6 levels compared with the HF group (HF-CP: -337% %, P < .05; HF-CT: -275%, P < .05). Hepatic steatosis and inflammation were attenuated in the HF-CP and HF-CT groups. The HF-CP group showed more pronounced expression of genes involved in adaptive thermogenesis and mitochondrial biogenesis (PPARγ, Prdm16, Pcg1α, ß3-adrenergic receptor, Ucp-1, and Opa-1) than the other experimental groups. Preventive coffee consumption associated with a high-fat diet ameliorates the metabolic profile related to the development of obesity and its comorbidities.


Subject(s)
Adipose Tissue, Brown , Diet, High-Fat , Mice , Animals , Adipose Tissue, Brown/metabolism , Diet, High-Fat/adverse effects , Coffee , Mice, Inbred C57BL , Obesity/metabolism , Adipose Tissue/metabolism , Inflammation/metabolism
6.
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
7.
Foods ; 13(1)2023 Dec 23.
Article in English | MEDLINE | ID: mdl-38201092

ABSTRACT

Despite advances in diagnosis and therapy, breast cancer remains the leading cause of death in many countries. Green tea (GT) has been proposed to play a crucial role in cancer chemoprevention. Although extensive research has been conducted on GT phytochemicals, most experimental studies concentrate mainly on commercial formulations or isolated catechins. This study presents a comparative investigation into the anticancer properties of green tea extract (GTE) and epigallocatechin-3-gallate (EGCG) in a three-dimensional (3D) MCF-7 breast cancer cell culture. MCF-7 spheroids were exposed to GTE or EGCG, and effects on 3D culture formation, growth, cell viability, and migration were examined. GTE inhibits cell migration and the formation of breast cancer spheroids more effectively than EGCG, while inducing more pronounced morphological changes in the spheroids' structure. These findings suggest that the food matrix improves GTE effects on breast cancer spheroids, supporting the hypothesis that a mixture of phytochemicals might enhance its anticancer potential.

8.
Sci Rep ; 12(1): 20855, 2022 12 02.
Article in English | MEDLINE | ID: mdl-36460743

ABSTRACT

The main goal of this study was to evaluate the reno-protective effects of a phenolic-rich Açaí seed extract (ASE) in mice with kidney failure. Kidney failure was induced chemically with an adenine-rich diet (0.25% w/w for 4 weeks) in male CD1 Swiss mice. Mice were then provided daily with ASE (at a dose of ~ 350 mg/kg/day) in drinking water for 4 weeks. Adenine mice exhibited renal dysfunction evidenced by increased proteinuria, increased uremia, extensive tubular atrophy and kidney fibrosis associated with overexpression of pro-fibrotic genes (collagen 1a1, transforming growth factor ß1, TGF-ß1) and markers of tubular injury (such as Kidney injury molecule-1, KIM-1). ASE was able to beneficially counteract all these effects. ASE improved oxidative damage and fibrosis by decreasing carbonylated protein and MDA concentrations, as well as collagen deposition in renal tissue. ASE decreased the expression of TGF-ß1 gene and the abundance of protein TGF-ß1 in kidneys. It further decreased both expression and urinary excretion of tubular injury biomarkers, e.g., KIM-1 and Neutrophil gelatinase-associated lipocalin. CKD ASE-treated mice exhibited higher polyphenol content and total antioxidant capacity compared to control mice. ASE further prevented the expression of profibrotic genes in HK2 human tubular cells exposed to uremic toxins. Taken together, these findings suggest that ASE exerted potent reno-protective and anti-fibrotic effects through its antioxidant activity and the modulation of the TGF-ß1 pathway.


Subject(s)
Polyphenols , Renal Insufficiency , Humans , Male , Mice , Animals , Polyphenols/pharmacology , Transforming Growth Factor beta1/genetics , Kidney , Antioxidants/pharmacology , Adenine , Fibrosis , Plant Extracts/pharmacology
9.
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
10.
Br J Nutr ; 127(3): 344-352, 2022 02 14.
Article in English | MEDLINE | ID: mdl-33787474

ABSTRACT

Accumulating evidence indicates that dietary phenolic compounds can prevent obesity-related disorders. We investigated whether the consumption of polyphenol-rich jabuticaba peel and seed powder (JPSP) could ameliorate the progression of diet-induced obesity in mice. Male mice were fed a control diet or a high-fat (HF) diet for 9 weeks. After this period, mice were fed control, HF or HF diets supplemented with 5 % (HF-J5), 10 % (HF-J10) or 15 % (HF-J15) of JPSP, for 4 additional weeks. Supplementation with JPSP not only attenuated HF-induced weight gain and fat accumulation but also ameliorated the pro-inflammatory response associated with obesity, as evidenced by the absence of mast cells in the visceral depot accompanied by lower IL-6 and TNF-α at the tissue and circulating levels. JPSP-supplemented mice also exhibited smaller-sized adipocytes, reduced levels of leptin and higher levels of adiponectin, concomitant with improved glucose metabolism and insulin sensitivity. The magnitude of the observed effects was dependent on JPSP concentration with HF-J10- and HF-J15-fed mice showing metabolic profiles similar to control. This study reveals that the consumption of JPSP protects against the dysfunction of the adipose tissue and metabolic disturbances in obese mice. Thus, these findings indicate the therapeutic potential of the phenolic-rich JPSP in preventing obesity-related disorders.


Subject(s)
Diet, High-Fat , Obesity , Adipose Tissue/metabolism , Animals , Diet, High-Fat/adverse effects , Male , Mice , Mice, Obese , Obesity/metabolism , Phenols/pharmacology , Phenols/therapeutic use , Powders/metabolism , Powders/therapeutic use
11.
Chem Biol Interact ; 351: 109721, 2022 Jan 05.
Article in English | MEDLINE | ID: mdl-34715092

ABSTRACT

Obesity is recognized as an independent risk factor for cardiovascular diseases and is an important contributor to cardiac mortality. Açaí seed extract (ASE), rich in proanthocyanidins, has been shown to have potential anti-obesity effects. This study aimed to investigate the therapeutic effect of ASE in cardiovascular remodeling associated with obesity and compare it with that of rosuvastatin. Male C57BL/6 mice were fed a high-fat diet or a standard diet for 12 weeks. The ASE (300 mg/kg/day) and rosuvastatin (20 mg/kg/day) treatments started in the 8th week until the 12th week, totaling 4 weeks of treatment. Our data showed that treatment with ASE and rosuvastatin reduced body weight, ameliorated lipid profile, and improved cardiovascular remodeling. Treatment with ASE but not rosuvastatin reduced hyperglycemia and oxidative stress by reducing immunostaining of 8-isoprostane and increasing SOD-1 and GPx expression in HFD mice. ASE and rosuvastatin reduced NOX4 expression, increased SIRT-1 and Nrf2 expression and catalase and GPx activities, and improved vascular and cardiac remodeling in HFD mice. The therapeutic effect of ASE was similar to that of rosuvastatin in reducing dyslipidemia and cardiovascular remodeling but was superior in reducing oxidative damage and hyperglycemia, suggesting that ASE was a promising natural product for the treatment of cardiovascular alterations associated with obesity.


Subject(s)
Antioxidants/therapeutic use , Cardiomegaly/drug therapy , Obesity/metabolism , Oxidative Stress/drug effects , Plant Extracts/therapeutic use , Ventricular Remodeling/drug effects , Animals , Cardiomegaly/etiology , Diet, High-Fat , Euterpe/chemistry , Male , Mice, Inbred C57BL , Mice, Obese , Obesity/complications , Proanthocyanidins/therapeutic use , Seeds/chemistry
12.
Appl Physiol Nutr Metab ; 46(12): 1469-1475, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34192478

ABSTRACT

This study evaluated the effect of green banana flour (GBF) consumption on obesity-related conditions in mice fed high-fat diets. GBF was prepared using stage 1 green banana pulp, which was dehydrated and milled. Mice were fed a control diet (n = 20; 10% of energy from lipids) or a high-fat diet (n = 20; 50% of energy from lipids). After 10 weeks, mice were divided into 4 groups based on feed: standard chow (SC; n = 10), standard with 15% GBF (SB; n = 10), high-fat diet (HF; n = 10) and high-fat diet with 15% GBF (HFB; n = 10) for 4 weeks. HFB exhibited lower gains in body weight (-21%; p < 0.01) and in all fat pads (p < 0.01) compared with the HF group. SC, SB, and HFB showed smaller retroperitoneal white adipose tissue diameters (p < 0.001). SB and HFB-treated mice showed lower levels of leptin, IL-6, and TNF-α compared with the SC and HF groups (p < 0.01). In the GBF-fed groups, there was a reduction in the abundance of Firmicutes (SB: -22%; HFB: -23%) and an increase in Bacteroidetes (SB: +25%; HFB: +29%) compared with their counterparts. We demonstrated that GBF consumption attenuated inflammation and improved metabolic status, adipose tissue remodeling, and the gut microbiota profile of obese mice. Novelty: Green banana flour (GBF) consumption, rich in resistant starch, regulates body weight in mice fed high-fat diets. GBF consumption improves fat pad distribution in mice fed high-fat diets. GBF improves obesity-associated systemic inflammation and regulates gut microbiota profile in mice fed high-fat diets.


Subject(s)
Food, Fortified , Gastrointestinal Microbiome , Inflammation/physiopathology , Musa , Obesity/microbiology , Obesity/physiopathology , Adiposity , Animals , Diet, High-Fat , Disease Models, Animal , Interleukin-6/blood , Leptin/blood , Male , Mice, Inbred C57BL , Mice, Obese , Obesity/prevention & control , Tumor Necrosis Factor-alpha/blood , Weight Gain
13.
Nutr Res ; 91: 26-35, 2021 07.
Article in English | MEDLINE | ID: mdl-34130208

ABSTRACT

Current evidence suggests that high fructose intake results in gut dysbiosis, leading to endotoxemia and NAFLD onset. Thus, the hypothesis of the study was that an enhanced Proteobacteria proportion in the cecal microbiota could be the most prominent trigger of NAFLD through enhanced endotoxin (LPS) in adult high-fructose-fed C57BL/6 mice. Male C57BL/6 mice received a control diet (n = 10, C: 76% of energy as carbohydrates, 0% as fructose) or high-fructose diet (n = 10, HFRU: 76% of energy as carbohydrate, 50% as fructose) for 12 weeks. Outcomes included biochemical analyses, 16S rDNA PCR amplification, hepatic stereology, and RT-qPCR. The groups showed similar body masses during the whole experiment. However, the HFRU group showed greater water intake and blood pressure than the C group. The HFRU group showed a significantly lower amount of Bacteroidetes and a predominant rise in Proteobacteria, implying increased LPS. The HFRU group also showed enhanced de novo lipogenesis (Chrebp expression), while beta-oxidation was decreased (Ppar-alpha expression). These results agree with the deposition of fat droplets within hepatocytes and the enhanced hepatic triacylglycerol concentrations, as observed in the photomicrographs, where the HFRU group had a higher volume density of steatosis than the C group. Thus, we confirmed that a rise in the Proteobacteria phylum proportion was the most prominent alteration in gut-liver axis-induced hepatic steatosis in HFRU-fed C57BL/6 mice. Gut dysbiosis and fatty liver were observed even in the absence of overweight in this dietary adult mouse model.


Subject(s)
Diet/adverse effects , Dysbiosis/microbiology , Fructose/adverse effects , Gastrointestinal Microbiome , Liver , Non-alcoholic Fatty Liver Disease/microbiology , Proteobacteria/growth & development , Animals , Body Weight , Cecum/microbiology , Dietary Sugars/adverse effects , Disease Models, Animal , Dysbiosis/etiology , Endotoxemia/etiology , Endotoxemia/microbiology , Feeding Behavior , Lipid Metabolism , Lipopolysaccharides , Liver/metabolism , Male , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/etiology , Triglycerides/metabolism
14.
Chem Biol Interact ; 336: 109369, 2021 Feb 25.
Article in English | MEDLINE | ID: mdl-33422521

ABSTRACT

Consumption of foods rich in phenolic compounds can be beneficial for health. This study aimed to examine whether the consumption of a phenolic-rich smoothie, based on juçara, strawberry and banana, ameliorates metabolic status and liver damage of diet-induced obese mice. Forty male C57BL/6J mice were assigned into four groups (n = 10) and fed control diet with free access to water (C) or phenolic-rich smoothie (C-S), or fed high-fat diet with free access to water (HF) or phenolic-rich smoothie (HF-S) for five weeks. HF and HF-S groups had higher body weight gains than the C group, however the HF had a greater adipose index, higher plasma levels of glucose, insulin and leptin, as well as higher plasma and hepatic steatosis than C, C-S and HF-S groups. The liver oxidative stress markers were reduced in C-S and HF-S groups and the activity of catalase and glutathione peroxidase were higher compared with their counterparts. The present study suggests that regular consumption of a phenolic-rich smoothie improves the liver antioxidant status, prevents metabolic disorders and ameliorates non-alcoholic fatty liver disease caused by high-fat diet consumption.


Subject(s)
Antioxidants/therapeutic use , Non-alcoholic Fatty Liver Disease/drug therapy , Obesity/drug therapy , Phenols/therapeutic use , Animals , Antioxidants/chemistry , Diet, High-Fat/adverse effects , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/chemically induced , Phenols/chemistry
15.
J Pharm Pharmacol ; 72(12): 1921-1932, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32856322

ABSTRACT

OBJECTIVES: Obesity is considered a risk factor for the development of non-alcoholic fatty liver disease (NAFLD). The hydroalcoholic extract obtained from the açai seed (ASE), rich in proanthocyanidins, has been shown a potential body weight regulator with antioxidant properties. This study aimed to investigate the therapeutic effect of ASE in obesity-associated NAFLD and compare it with Rosuvastatin. METHODS: Male C57BL/6 mice received a high-fat diet or standard diet for 12 weeks. The treatments with ASE (300 mg/kg per day) or rosuvastatin (20 mg/kg per day) began in the eighth week until the 12th week. KEY FINDINGS: Our data show that the treatments with ASE and rosuvastatin reduced body weight and hyperglycaemia, improved lipid profile and attenuated hepatic steatosis in HFD mice. ASE and Rosuvastatin reduced HMGCoA-Reductase and SREBP-1C and increased ABGC8 and pAMPK expressions in the liver. Additionally, ASE, but not Rosuvastatin, reduced NPC1L1 and increased ABCG5 and PPAR-α expressions. ASE and rosuvastatin increased SIRT-1 expression and antioxidant defence, although only ASE was able to decrease the oxidative damage in hepatic tissue. CONCLUSIONS: The therapeutic effect of ASE was similar to that of rosuvastatin in reducing dyslipidemia and hepatic steatosis but was better in reducing oxidative damage and hyperglycaemia.


Subject(s)
Euterpe , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Hypolipidemic Agents/pharmacology , Liver/drug effects , Non-alcoholic Fatty Liver Disease/prevention & control , Obesity/drug therapy , Plant Extracts/pharmacology , Rosuvastatin Calcium/pharmacology , Animals , Diet, High-Fat , Disease Models, Animal , Dyslipidemias/metabolism , Dyslipidemias/prevention & control , Euterpe/chemistry , Hyperglycemia/metabolism , Hyperglycemia/prevention & control , Hypolipidemic Agents/isolation & purification , Lipid Metabolism/drug effects , Liver/metabolism , Liver/pathology , Male , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Obesity/etiology , Obesity/metabolism , Obesity/pathology , Oxidative Stress/drug effects , Plant Extracts/isolation & purification , Seeds
16.
J Endocrinol ; 247(1): 11-24, 2020 10.
Article in English | MEDLINE | ID: mdl-32698143

ABSTRACT

Fructose dietary intake affects the composition of the intestinal microbiota and influences the development of hepatic steatosis. Endotoxins produced by gram-negative bacteria alter intestinal permeability and cause bacterial translocation. This study evaluated the effects of gut microbiota modulation by a purified PPAR-alpha agonist (WY14643), a DPP-4 inhibitor (linagliptin), or their association on intestinal barrier integrity, endotoxemia, and hepatic energy metabolism in high-fructose-fed C57BL/6 mice. Fifty mice were divided to receive the control diet (C group) or the high-fructose diet (HFRU) for 12 weeks. Subsequently, the HFRU group was divided to initiate the treatment with PPAR-alpha agonist (3.5 mg/kg/BM) and DPP-4 inhibitor (15 mg/kg/BM). The HFRU group had glucose intolerance, endotoxemia, and dysbiosis (with increased Proteobacteria) without changes in body mass in comparison with the C group. HFRU group showed damaged intestinal ultrastructure, which led to liver inflammation and marked hepatic steatosis in the HFRU group when compared to the C group. PPAR-alpha activation and DPP-4 inhibition countered glucose intolerance, endotoxemia, and dysbiosis, ameliorating the ultrastructure of the intestinal barrier and reducing Tlr4 expression in the liver of treated animals. These beneficial effects suppressed lipogenesis and mitigated hepatic steatosis. In conclusion, the results herein propose a role for PPAR-alpha activation, DPP-4 inhibition, and their association in attenuating hepatic steatosis by gut-liver axis modulation in high-fructose mice model. These observations suggest these treatments as potential targets to treat hepatic steatosis and avoid its progression.


Subject(s)
Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Fructose/administration & dosage , Gastrointestinal Microbiome/drug effects , Linagliptin/pharmacology , Liver/drug effects , PPAR alpha/physiology , Animals , Blood Glucose/analysis , Diet , Endotoxemia/prevention & control , Fatty Liver/prevention & control , Gastrointestinal Microbiome/physiology , Intestines/drug effects , Intestines/ultrastructure , Lipogenesis/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , PPAR alpha/drug effects , Peroxisome Proliferators , Pyrimidines/pharmacology
17.
Int J Biol Macromol ; 145: 1066-1072, 2020 Feb 15.
Article in English | MEDLINE | ID: mdl-31730978

ABSTRACT

This study aimed to investigate the effect of resistant starch from green banana (GB) on steatosis and short-chain fatty acid (SCFAs) production in high fat diet-induced obesity in mice. High-fat green banana group (HFB) exhibited lower gains in BM (body mass; -6%; P < 0.01) compared with High-fat diet group (HF). Additionally, HFB mice showed reduction in liver steatosis (-28%, P < 0.01) with reduction of 93% in hepatic triacylglycerol (P < 0.01) compared to HF-diet-fed mice. In addition, the protein abundance of AMPKp/AMPK, HMGCoA-r and FAS were downregulated in livers of HFB mice (P < 0.01), relatively to the HF-diet-fed mice. ABCG8 and ABCG5 were up-regulated in HFB group compared to HF group (P < 0.01). Furthermore, the HFB fed-mice produced the highest amount of SCFAs (p < 0.05) compared to its counterpart HFD. In conclusion, we demonstrated that resistant starch from GB improved metabolic parameters by modulating the expression of key proteins involved in liver lipid metabolism.


Subject(s)
Diet, High-Fat/adverse effects , Dietary Carbohydrates/therapeutic use , Fatty Acids, Volatile/metabolism , Musa/chemistry , Non-alcoholic Fatty Liver Disease/metabolism , Starch/administration & dosage , Animals , Dietary Supplements , Disease Models, Animal , Eating , Fasting , Glucose/metabolism , Glucose Tolerance Test , Lipid Metabolism/drug effects , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Obesity/metabolism , Plant Extracts/pharmacology , Triglycerides/metabolism
18.
Exp Physiol ; 105(3): 542-551, 2020 03.
Article in English | MEDLINE | ID: mdl-31876965

ABSTRACT

NEW FINDINGS: What is the central question of this study? Does a polyphenol-rich extract from açaí have a potential role in preventing uraemic toxin-induced endothelial cell dysfunction? What is the main finding and its importance? Polyphenols from açaí prevented cell death, restored migratory capacity, protected from inflammation and contributed to the restoration of the antioxidant response in endothelial cells exposed to uraemic toxins. The protective role of açaí against toxic effects exerted by uraemic toxins presents a potential new therapeutic target in endothelial cells. ABSTRACT: In chronic kidney disease (CKD), progressive loss of kidney function results in the accumulation of protein-bound uraemic toxins such as p-cresyl sulfate (pCS) and indoxyl sulfate (IS). Among strategies to ameliorate the harmful actions of uraemic toxins, phenolic compounds have been extensively studied. The main goal of this work was to evaluate the antioxidant and anti-inflammatory actions of phenolic-rich açaí seed extract (ASE) in response to endothelial dysfunction induced by IS and pCS, in human umbilical vein endothelial cells (HUVECs). Cells were treated with ASE (10 µg ml-1 ) in the presence or absence of IS (61 µg ml-1 ) and pCS (40 µg ml-1 ). Cell viability, cell death, cell migratory capacity and inflammatory biomarker expression were evaluated. Cellular antioxidant response was measured through the activity and expression of antioxidant enzymes, and oxidative damage was evaluated. IS and pCS lowered cell viability, triggered cell death and lowered the migratory capacity in endothelial cells (P < 0.05). ASE prevented cell death and restored the migratory capacity in cells exposed to IS. Both toxins up-regulated pro-inflammatory cytokine expression, and ASE was able to beneficially counteract this effect. Tumour necrosis factor-α secretion was greater in uraemic toxin-treated cells and ASE reversed this phenomenon in cells treated with both toxins concomitantly (P < 0.05). With regard to the antioxidant response, superoxide dismutase expression was strikingly lower in cells treated with both toxins, and ASE inhibited this harmful effect (P < 0.05). From the results, we conclude that ASE exerted protective effects on inflammation and oxidative stress caused by uraemic toxins (particularly by IS) in human endothelial cells.


Subject(s)
Euterpe/chemistry , Human Umbilical Vein Endothelial Cells/drug effects , Inflammation/drug therapy , Oxidative Stress/drug effects , Plant Extracts/pharmacology , Polyphenols/pharmacology , Protective Agents/pharmacology , Antioxidants , Biomarkers/metabolism , Cell Death/drug effects , Cells, Cultured , Cytokines/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Inflammation/metabolism , Renal Insufficiency, Chronic/drug therapy , Renal Insufficiency, Chronic/metabolism , Superoxide Dismutase/metabolism
19.
Life Sci ; 228: 316-322, 2019 Jul 01.
Article in English | MEDLINE | ID: mdl-31022407

ABSTRACT

Body adiposity is an important risk factor for the development of chronic non-transmissible diseases. Studies on the process of adipogenesis have been extensively performed in vivo and in vitro models to describe the molecular and cellular bases of adipose tissue development and the effect of natural products in this process. The açai seed extract (ASE) has been evidenced as a potential regulator of body mass. In our work high-fat diet-fed mice treated with ASE (300 mg/Kg/d) (HFD-ASE) showed a lower adipose index (-32.63%, p < 0.001) than the high-fat diet-fed mice group (HFD) and the adipocytes from the HFD group were considerably enlarged (p < 0.001) compared to those in the control group (CG) and HFD-ASE group (+175% and +123%, respectively). We also evaluated the effects of ASE on the modulation of adipogenesis in 3T3-L1 cells. ASE exposure (25 and 100 µg/mL) led to a decrease of 26.6 (p < 0.05) in proliferation and also inhibited pre-adipocyte differentiation through the decreasing expression (p < 0.05) of transcription factors and adipogenic proteins such as PPARÉ£, SREBP-1, and FAS. These results show that the ASE reduce adipogenesis and suppress lipid accumulation in the in vivo model and in 3T3-L1 adipocytes and reinforce ASE as a potential strategy to modulate adipogenesis.


Subject(s)
Adipocytes/drug effects , Adipogenesis/drug effects , Anti-Obesity Agents/pharmacology , Diet, High-Fat/adverse effects , Euterpe/chemistry , Plant Extracts/pharmacology , 3T3-L1 Cells , Adipocytes/metabolism , Adiposity/drug effects , Animals , Anti-Obesity Agents/chemistry , Lipid Metabolism/drug effects , Male , Mice , Mice, Inbred C57BL , Obesity/metabolism , Obesity/prevention & control , Plant Extracts/chemistry , Seeds/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...