Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 2.468
1.
PLoS One ; 19(5): e0303060, 2024.
Article En | MEDLINE | ID: mdl-38723008

In the current study we investigated the impact of combination of rutin and vitamin A on glycated products, the glyoxalase system, oxidative markers, and inflammation in animals fed a high-fat high-fructose (HFFD) diet. Thirty rats were randomly divided into six groups (n = 5). The treatments, metformin (120 mg/kg), rutin (100 mg/kg), vitamin A (43 IU/kg), and a combination of rutin (100 mg/kg) and vitamin A (43 IU/kg) were given to relevant groups of rats along with high-fructose high-fat diet for 42 days. HbA1c, D-lactate, Glyoxylase-1, Hexokinase 2, malondialdehyde (MDA), glutathione peroxidase (GPx), catalase (CAT), nuclear transcription factor-B (NF-κB), interleukin-6 (IL-6), interleukin-8 (IL-8) and histological examinations were performed after 42 days. The docking simulations were conducted using Auto Dock package. The combined effects of rutin and vitamin A in treated rats significantly (p < 0.001) reduced HbA1c, hexokinase 2, and D-lactate levels while preventing cellular damage. The combination dramatically (p < 0.001) decreased MDA, CAT, and GPx in treated rats and decreased the expression of inflammatory cytokines such as IL-6 andIL-8, as well as the transcription factor NF-κB. The molecular docking investigations revealed that rutin had a strong affinity for several important biomolecules, including as NF-κB, Catalase, MDA, IL-6, hexokinase 2, and GPx. The results propose beneficial impact of rutin and vitamin A as a convincing treatment strategy to treat AGE-related disorders, such as diabetes, autism, alzheimer's, atherosclerosis.


Diet, High-Fat , Fructose , Hyperglycemia , Inflammation , Oxidative Stress , Rutin , Vitamin A , Animals , Rutin/pharmacology , Oxidative Stress/drug effects , Fructose/adverse effects , Rats , Diet, High-Fat/adverse effects , Vitamin A/pharmacology , Vitamin A/metabolism , Inflammation/metabolism , Inflammation/drug therapy , Inflammation/pathology , Male , Hyperglycemia/drug therapy , Hyperglycemia/metabolism , Hyperglycemia/chemically induced , Molecular Docking Simulation , Rats, Wistar , Disease Models, Animal , Glycosylation/drug effects , Metformin/pharmacology , Glycated Hemoglobin/metabolism , NF-kappa B/metabolism , Hexokinase/metabolism , Catalase/metabolism
2.
Front Immunol ; 15: 1375461, 2024.
Article En | MEDLINE | ID: mdl-38711514

Excess dietary fructose consumption has been long proposed as a culprit for the world-wide increase of incidence in metabolic disorders and cancer within the past decades. Understanding that cancer cells can gradually accumulate metabolic mutations in the tumor microenvironment, where glucose is often depleted, this raises the possibility that fructose can be utilized by cancer cells as an alternative source of carbon. Indeed, recent research has increasingly identified various mechanisms that show how cancer cells can metabolize fructose to support their proliferating and migrating needs. In light of this growing interest, this review will summarize the recent advances in understanding how fructose can metabolically reprogram different types of cancer cells, as well as how these metabolic adaptations can positively support cancer cells development and malignancy.


Fructose , Neoplasms , Tumor Microenvironment , Humans , Fructose/metabolism , Fructose/adverse effects , Neoplasms/metabolism , Neoplasms/etiology , Animals , Cellular Reprogramming/drug effects , Energy Metabolism/drug effects , Metabolic Reprogramming
3.
Biosci Rep ; 44(5)2024 May 29.
Article En | MEDLINE | ID: mdl-38660995

Several models of mice-fed high-fat diets have been used to trigger non-alcoholic steatohepatitis and some chemical substances, such as carbon tetrachloride. The present study aimed to evaluate the joint action of a high-fat diet and CCl4 in developing a short-term non-alcoholic steatohepatitis model. C57BL6/J mice were divided into two groups: standard diet-fed (SD), the high-fat diet-fed (HFD) and HFD + fructose-fed and carbon tetrachloride (HFD+CCl4). The animals fed with HFD+CCl4 presented increased lipid deposition compared with both SD and HFD mice. Plasma cholesterol was increased in animals from the HFD+CCl4 group compared with the SD and HFD groups, without significant differences between the SD and HFD groups. Plasma triglycerides showed no significant difference between the groups. The HFD+CCl4 animals had increased collagen deposition in the liver compared with both SD and HFD groups. Hydroxyproline was also increased in the HFD+CCl4 group. Liver enzymes, alanine aminotransferase and aspartate aminotransferase, were increased in the HFD+CCl4 group, compared with SD and HFD groups. Also, CCl4 was able to trigger an inflammatory process in the liver of HFD-fed animals by promoting an increase of ∼2 times in macrophage activity, ∼6 times in F4/80 gene expression, and pro-inflammatory cytokines (IL-1b and TNFa), in addition to an increase in inflammatory pathway protein phosphorylation (IKKbp). HFD e HFD+CCl4 animals increased glucose intolerance compared with SD mice, associated with reduced insulin-stimulated AKT activity in the liver. Therefore, our study has shown that short-term HFD feeding associated with fructose and CCl4 can trigger non-alcoholic steatohepatitis and cause damage to glucose metabolism.


Carbon Tetrachloride , Diet, High-Fat , Disease Models, Animal , Liver , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease , Animals , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Diet, High-Fat/adverse effects , Liver/metabolism , Liver/pathology , Male , Mice , Triglycerides/blood , Triglycerides/metabolism , Fructose/adverse effects
4.
Nutrients ; 16(7)2024 Mar 22.
Article En | MEDLINE | ID: mdl-38612947

We aimed to investigate how dietary fructose and sodium impact blood pressure and risk of hypertensive target organ damage 10 years later. Data from n = 3116 individuals were obtained from the Coronary Artery Risk Development in Young Adults (CARDIA) study. Four groups were identified based on the four possible combinations of the lower and upper 50th percentile for sodium (in mg) and fructose (expressed as percent of total daily calories). Differences among groups were ascertained and logistic regression analyses were used to assess the risk of hypertensive target organ damage (diastolic dysfunction, coronary calcification and albuminuria). Individuals in the low-fructose + low-sodium group were found to have lower SBP compared to those in the low-fructose + high-sodium and high-fructose + high-sodium groups (p < 0.05). The highest risk for hypertensive target organ damage was found for albuminuria only in the high-fructose + high-sodium group (OR = 3.328, p = 0.006) while female sex was protective across all groups against coronary calcification. Our findings highlight that sodium alone may not be the culprit for hypertension and hypertensive target organ damage, but rather when combined with an increased intake of dietary fructose, especially in middle-aged individuals.


Calcinosis , Hypertension , Middle Aged , Young Adult , Female , Humans , Coronary Vessels , Sodium , Albuminuria , Hypertension/epidemiology , Hypertension/etiology , Diet, Sodium-Restricted , Fructose/adverse effects
5.
Nutrients ; 16(7)2024 Mar 25.
Article En | MEDLINE | ID: mdl-38612973

Worldwide, childhood obesity cases continue to rise, and its prevalence is known to increase the risk of non-communicable diseases typically found in adults, such as cardiovascular disease and type 2 diabetes mellitus. Thus, comprehending its multiple causes to build healthier approaches and revert this scenario is urgent. Obesity development is strongly associated with high fructose intake since the excessive consumption of this highly lipogenic sugar leads to white fat accumulation and causes white adipose tissue (WAT) inflammation, oxidative stress, and dysregulated adipokine release. Unfortunately, the global consumption of fructose has increased dramatically in recent years, which is associated with the fact that fructose is not always evident to consumers, as it is commonly added as a sweetener in food and sugar-sweetened beverages (SSB). Therefore, here, we discuss the impact of excessive fructose intake on adipose tissue biology, its contribution to childhood obesity, and current strategies for reducing high fructose and/or free sugar intake. To achieve such reductions, we conclude that it is important that the population has access to reliable information about food ingredients via food labels. Consumers also need scientific education to understand potential health risks to themselves and their children.


Diabetes Mellitus, Type 2 , Pediatric Obesity , Child , Adult , Humans , Pediatric Obesity/epidemiology , Pediatric Obesity/etiology , Pediatric Obesity/prevention & control , Adipose Tissue , Adipose Tissue, White , Fructose/adverse effects
6.
PLoS One ; 19(4): e0301012, 2024.
Article En | MEDLINE | ID: mdl-38573884

Different functional foods with bioactive nutrients are being explored for the management of NAFLD. Whey proteins are rich in bioactive peptides and are suggested to show antioxidant and anti-inflammatory effects. We aim to test the hypothesis that the whey protein supplementation following a high fat-high fructose (HFHF) diet would protect against liver damage, inflammation, endotoxemia and steatosis in male Wistar rats. 36 rats were randomized into four groups for 8 weeks as the HFHF diet group, HFHF diet and whey protein isolate (WPI-200mg/kg/day) group (HFHF+WPI), control (C) group, and C+WPI (200mg/kg/day) group. Rats fed with a HFHF diet had higher final body weight compared to C and C+WPI groups (p = 0.002). Thus, WPI showed no significant effects for the body weight of rats with a HFHF diet. On the other hand, the HFHF+WPI group had significantly lower abdominal circumference when compared with the HFHF group (p<0,001). Higher serum CRP levels were observed in the groups with a HFHF diet (p<0,001) and WPI supplementation showed no effects on CRP levels. Whey protein supplementation resulted with lower total liver damage score in HFHF+WPI group compared with the HFHF diet group (p<0,001). Conversely, higher liver damage scores were observed with the C+WPI group compared to C group (p<0,001). HFHF diet resulted with higher expression of TLR-4 in the liver meanwhile WPI supplementation showed no effects on liver TLR-4 expression. We observed higher colon Occludin expression in HFHF+WPI and C+WPI groups compared with HFHF and C groups (p<0,001). Our results showed that, whey protein supplementation might help improve liver damage associated with a high fat-high fructose diet and increase the expression of Occludin in the small intestine and colon.


Fructose , Toll-Like Receptor 4 , Rats , Male , Animals , Whey Proteins/pharmacology , Rats, Wistar , Fructose/adverse effects , Occludin , Diet, High-Fat/adverse effects , Liver , Body Weight , Dietary Supplements
7.
J Nutr Biochem ; 128: 109626, 2024 Jun.
Article En | MEDLINE | ID: mdl-38527560

Along with the increasing prevalence of obesity worldwide, the deleterious effects of high-calorie diet are gradually recognized through more and more epidemiological studies. However, the concealed and chronic causality whitewashes its unhealthy character. Given an ingenious mechanism orchestrates the metabolic adaptation to high-fat high-fructose (HFF) diet and connive its lipotoxicity, in this study, an experimental rat/mouse model of obesity was induced and a comparative transcriptomic analysis was performed to probe the mystery. Our results demonstrated that HFF diet consumption altered the transcriptomic pattern as well as different high-calorie diet fed rat/mouse manifested distinct hepatic transcriptome. Validation with RT-qPCR and Western blotting confirmed that SREBP1-FASN involved in de novo lipogenesis partly mediated metabolic self-adaption. Moreover, hepatic ACSL1-CPT1A-CPT2 pathway involved in fatty acids ß-oxidation, played a key role in the metabolic adaption to HFF. Collectively, our findings enrich the knowledge of the chronic adaptation mechanisms and also shed light on future investigations. Meanwhile, our results also suggest that efforts to restore the fatty acids metabolic fate could be a promising avenue to fight against obesity and associated steatosis and insulin resistance challenged by HFF diet.


Diet, High-Fat , Fatty Acid Synthase, Type I , Fructose , Liver , Obesity , Sterol Regulatory Element Binding Protein 1 , Transcriptome , Animals , Fructose/adverse effects , Diet, High-Fat/adverse effects , Male , Liver/metabolism , Obesity/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , Sterol Regulatory Element Binding Protein 1/genetics , Lipogenesis , Mice, Inbred C57BL , Rats , Mice , Rats, Sprague-Dawley , Fatty Acids/metabolism
9.
Int J Mol Sci ; 25(6)2024 Mar 14.
Article En | MEDLINE | ID: mdl-38542281

Epilepsy ranks as the second-most prevalent neurological disease, and is characterized by seizures resulting in neurobiological and behavioral impairment. Naturally occurring in coffee beans or tea leaves, the alkaloid caffeine (CAF) is the most prevalent global stimulant. Caffeine has been observed to influence epileptic seizures and the efficacy of antiepileptic medications, with a notable impact on topiramate (TPM). This study aimed to explore the influence of CAF on TPM's anticonvulsant effects in zebrafish larvae within a PTZ-induced seizure model, concurrently determining TPM concentrations through a sophisticated analytical approach based on ultrahigh-performance liquid chromatography and subsequent mass spectrometric detection. Zebrafish larvae four days post-fertilization were incubated for 18 h with varying doses of TPM or combinations of CAF + TPM, and locomotor activity was then assessed. Seizures were induced by introducing a PTZ solution to achieve a final concentration of 20 mM. Utilizing liquid chromatography-mass spectrometry (LC-MS/MS), TPM levels in the larvae were quantified. CAF co-administration (especially in higher doses) with TPM caused a decrease in the average locomotor activity in the larvae compared to TPM alone. Moreover, CAF decreased TPM levels in the larvae at all investigated doses. In conclusion, these findings offer a novel perspective on the interplay between CAF and TPM, shedding light on previously unexplored facets. The potential impact of CAF consumption in assisting with epileptic seizure control, unless proven otherwise, suggests a noteworthy consideration for future research and clinical practices.


Epilepsy , Zebrafish , Animals , Topiramate/therapeutic use , Pentylenetetrazole/toxicity , Caffeine/pharmacology , Caffeine/therapeutic use , Chromatography, Liquid , Fructose/adverse effects , Tandem Mass Spectrometry , Seizures/chemically induced , Seizures/drug therapy , Anticonvulsants/adverse effects , Epilepsy/drug therapy
10.
EMBO Rep ; 25(4): 2097-2117, 2024 Apr.
Article En | MEDLINE | ID: mdl-38532128

High fructose intake during pregnancy increases insulin resistance (IR) and gestational diabetes mellitus (GDM) risk. IR during pregnancy primarily results from elevated hormone levels. We aim to determine the role of liver carbohydrate response element binding protein (ChREBP) in insulin sensitivity and lipid metabolism in pregnant mice and their offspring. Pregnant C57BL/6J wild-type mice and hepatocyte-specific ChREBP-deficient mice were fed with a high-fructose diet (HFrD) or normal chow diet (NC) pre-delivery. We found that the combination of HFrD with pregnancy excessively activates hepatic ChREBP, stimulating progesterone synthesis by increasing MTTP expression, which exacerbates IR. Increased progesterone levels upregulated hepatic ChREBP via the progesterone-PPARγ axis. Placental progesterone activated the progesterone-ChREBP loop in female offspring, contributing to IR and lipid accumulation. In normal dietary conditions, hepatic ChREBP modestly affected progesterone production and influenced IR during pregnancy. Our findings reveal the role of hepatic ChREBP in regulating insulin sensitivity and lipid homeostasis in both pregnant mice consuming an HFrD and female offspring, and suggest it as a potential target for managing gestational metabolic disorders, including GDM.


Insulin Resistance , Pregnancy , Female , Mice , Animals , Insulin Resistance/genetics , Fructose/adverse effects , Fructose/metabolism , Progesterone/metabolism , Mice, Inbred C57BL , Placenta/metabolism , Liver/metabolism , Lipids , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism
11.
Biochim Biophys Acta Mol Basis Dis ; 1870(3): 167029, 2024 03.
Article En | MEDLINE | ID: mdl-38325224

High fructose diets are associated with an increased risk of liver cancer. Previous studies in mice suggest increased lipogenesis is a key mechanism linking high fructose diets to liver tumour growth. However, these studies administered fructose to mice at supraphysiological levels. The aim of this study was to determine whether liver tumour growth and lipogenesis were altered in mice fed fructose at physiological levels. To test this, we injected male C57BL/6 mice with the liver carcinogen diethylnitrosamine and then fed them diets without fructose or fructose ranging from 10 to 20 % total calories. Results showed mice fed diets with ≥15 % fructose had significantly increased liver tumour numbers (2-4-fold) and total tumour burden (∼7-fold) vs mice fed no-fructose diets. However, fructose-associated tumour burden was not associated with lipogenesis. Conversely, unbiased metabolomic analyses revealed bile acids were elevated in the sera of mice fed a 15 % fructose diet vs mice fed a no-fructose diet. Using a syngeneic ectopic liver tumour model, we show that ursodeoxycholic acid, which decreases systemic bile acids, significantly reduced liver tumour growth in mice fed the 15 % fructose diet but not mice fed a no-fructose diet. These results point to a novel role for systemic bile acids in mediating liver tumour growth associated with a high fructose diet. Overall, our study shows fructose intake at or above normal human consumption (≥15 %) is associated with increased liver tumour numbers and growth and that modulating systemic bile acids inhibits fructose-associated liver tumour growth in mice.


Bile Acids and Salts , Liver Neoplasms , Humans , Mice , Male , Animals , Fructose/adverse effects , Mice, Inbred C57BL , Liver Neoplasms/chemically induced
12.
Eur J Pharmacol ; 967: 176356, 2024 Mar 15.
Article En | MEDLINE | ID: mdl-38325797

Accumulating evidence suggests that excess fructose uptake induces metabolic syndrome and kidney injury. Here, we primarily investigated the influence of catalpol on fructose-induced renal inflammation in mice and explored its potential mechanism. Treatment with catalpol improved insulin sensitivity and hyperuricemia in fructose-fed mice. Hyperuricemia induced by high-fructose diet was associated with increases in the expressions of urate reabsorptive transporter URAT1 and GLUT9. Treatment with catalpol decreased the expressions of URAT1 and GLUT9. Futhermore, treatment with catalpol ameliorated renal inflammatory cell infiltration and podocyte injury, and these beneficial effects were associated with inhibiting the production of inflammatory cytokines including IL-1ß, IL-18, IL-6 and TNF-α. Moreover, fructose-induced uric acid triggers an inflammatory response by activiting NLRP3 inflammasome, which then processes pro-inflammatory cytokines. Treatment with catalpol could inhibit the activation of NLRP3 inflammasome as well. Additionally, TLR4/MyD88 signaling was activated in fructose-fed mice, while treatment with catalpol inhibited this activation along with promoting NF-κB nuclear translocation in fructose-fed mice. Thus, our study demonstrated that catalpol could ameliorate renal inflammation in fructose-fed mice, attributing its beneficial effects to promoting uric acid excretion and inhibit the activation of TLR4/MyD88 signaling.


Hyperuricemia , Iridoid Glucosides , Nephritis , Mice , Animals , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Uric Acid/metabolism , Inflammasomes/metabolism , Hyperuricemia/chemically induced , Hyperuricemia/drug therapy , Hyperuricemia/metabolism , Myeloid Differentiation Factor 88/metabolism , Toll-Like Receptor 4/metabolism , Fructose/adverse effects , Adaptor Proteins, Signal Transducing/metabolism , NF-kappa B/metabolism , Cytokines/metabolism , Inflammation/drug therapy , Inflammation/metabolism
13.
Biochim Biophys Acta Mol Basis Dis ; 1870(4): 167074, 2024 Apr.
Article En | MEDLINE | ID: mdl-38354758

For the advancement of DKD treatment, identifying unrecognized residual risk factors is essential. We explored the impact of obesity diversity derived from different carbohydrate qualities, with an emphasis on the increasing trend of excessive fructose consumption and its effect on DKD progression. In this study, we utilized db/db mice to establish a novel diabetic model characterized by fructose overconsumption, aiming to uncover the underlying mechanisms of renal damage. Compared to the control diet group, the fructose-fed db/db mice exhibited more pronounced obesity yet demonstrated milder glucose intolerance. Plasma cystatin C levels were elevated in the fructose model compared to the control, and this elevation was accompanied by enhanced glomerular sclerosis, even though albuminuria levels and tubular lesions were comparable. Single-cell RNA sequencing of the whole kidney highlighted an increase in Lrg1 in glomerular endothelial cells (GECs) in the fructose model, which appeared to drive mesangial fibrosis through enhanced TGF-ß1 signaling. Our findings suggest that excessive fructose intake exacerbates diabetic kidney disease progression, mediated by aberrant Lrg1-driven crosstalk between GECs and mesangial cells.


Diabetic Nephropathies , Mesangial Cells , Mice , Animals , Endothelial Cells/pathology , Fructose/adverse effects , Diabetic Nephropathies/pathology , Mice, Inbred Strains , Obesity/complications , Cell Communication
14.
Nutr J ; 23(1): 16, 2024 Feb 02.
Article En | MEDLINE | ID: mdl-38302919

Labels do not disclose the excess-free-fructose/unpaired-fructose content in foods/beverages. Objective was to estimate excess-free-fructose intake using USDA loss-adjusted-food-availability (LAFA) data (1970-2019) for high fructose corn syrup (HFCS) and apple juice, major sources of excess-free-fructose, for comparison with malabsorption dosages (~ 5 g-children/ ~ 10 g-adults). Unlike sucrose and equimolar fructose/glucose, unpaired-fructose triggers fructose malabsorption and its health consequences. Daily intakes were calculated for HFCS that is generally-recognized-as-safe/ (55% fructose/45% glucose), and variants (65/35, 60/40) with higher fructose-to-glucose ratios (1.9:1, 1.5:1), as measured by independent laboratories. Estimations include consumer-level-loss (CLL) allowances used before (20%), and after, subjective, retroactively-applied increases (34%), as recommended by corn-refiners (~ 2012). No contributions from crystalline-fructose or agave syrup were included due to lack of LAFA data. High-excess-free-fructose-fruits (apples/pears/watermelons/mangoes) were not included. Eaten in moderation they are less likely to trigger malabsorption. Another objective was to identify potential parallel trends between excess-free-fructose intake and the "unexplained" US asthma epidemic. The fructose/gut-dysbiosis/lung axis is well documented, case-study evidence and epidemiological research link HFCS/apple juice intake with asthma, and unlike gut-dysbiosis/gut-fructosylation, childhood asthma prevalence data spans > 40 years. Results Excess-free-fructose daily intake for individuals consuming HFCS with an average 1.5:1 fructose-to-glucose ratio, ranged from 0.10 g/d in 1970, to 11.3 g/d in 1999, to 6.5 g/d in 2019, and for those consuming HFCS with an average 1.9:1 ratio, intakes ranged from 0.13 g/d to 16.9 g/d (1999), to 9.7 g/d in 2019, based upon estimates with a 20% CLL allowance. Intake exceeded dosages that trigger malabsorption (~ 5 g) around ~ 1980. By the early 1980's, tripled apple juice intake had added ~ 0.5 g to average-per-capita excess-free-fructose intake. Contributions were higher (~ 3.8 g /4-oz.) for individuals consuming apple juice consistent with a healthy eating pattern (4-oz. children, 8-oz. adults). The "unexplained" childhood asthma epidemic (1980-present) parallels increasing average-per-capita HFCS/apple juice intake trends and reflects epidemiological research findings. Conclusion Displacement of sucrose with HFCS, its ubiquitous presence in the US food-supply, the industry practice of adding more fructose to HFCS than generally-recognized-as-safe, and increased use of apple juice/crystalline fructose/agave syrup in foods/beverages has contributed to unprecedented excess-free-fructose intake levels, fructose malabsorption, gut-dysbiosis and gut-fructosylation (immunogen burden)-gateways to chronic disease.


Asthma , High Fructose Corn Syrup , Leukemia, Lymphocytic, Chronic, B-Cell , Malus , Adult , Humans , Child , High Fructose Corn Syrup/adverse effects , Fructose/adverse effects , Dysbiosis , Glucose , Chronic Disease , Asthma/epidemiology , Sucrose
15.
Nutrients ; 16(3)2024 Jan 26.
Article En | MEDLINE | ID: mdl-38337654

Consumption of foods with fiber and compounds can promote gastrointestinal health and reduce obesity complications. Therefore, treatment with common bean leaves (BL) against obesity was evaluated in mice with a high-fat and high-fructose diet (HFFD) for 14 weeks. The bromatological and phytochemical characterization of BL were determined. Afterwards, the animals were supplemented with BL (10%) or a standard diet (SD) as a strategy to encourage a healthy diet for 12 additional weeks. Changes in body composition, lipid profile, and intestinal integrity were analyzed. The characterization of BL stood out for its content of 27.2% dietary fiber, total phenolics (475.04 mg/100 g), and saponins (2.2 mg/100 g). The visceral adipose tissue (VAT) decreased in the BL group by 52% compared to the HFFD group. Additionally, triglyceride levels were 23% lower in the BL consumption group compared to the HFFD group. The improvement in lipid profile was attributed to the 1.77-fold higher fecal lipid excretion in the BL consumption group compared to the HFFD group and the inhibition of pancreatic lipase by 29%. Furthermore, BL supplementation reduced the serum levels of IL-6 (4.4-fold) and FITC-dextran by 50% compared with those in the HFFD group. Metabolic endotoxemia was inhibited after BL supplementation (-33%) compared to the HFFD group. BL consumption as a treatment in obese mice reduces adipose tissue accumulation and improves the lipid profile. Furthermore, we report for the first time that BL consumption improves intestinal integrity.


Diet, High-Fat , Fructose , Mice , Animals , Fructose/adverse effects , Fructose/metabolism , Diet, High-Fat/adverse effects , Mice, Inbred C57BL , Obesity/metabolism , Adipose Tissue/metabolism , Lipids , Eating
16.
J Pharm Pharmacol ; 76(5): 567-578, 2024 May 03.
Article En | MEDLINE | ID: mdl-38271051

OBJECTIVES: Accumulating evidence demonstrates that copper deficiency (CuD) is a risk factor for cardiovascular diseases, besides, fructose has been strongly linked to the development of cardiovascular diseases. However, how CuD or fructose causes cardiovascular diseases is not clearly delineated. The present study aims to investigate the mechanism of CuD or fructose on cardiac remodeling. METHODS: We established a model of CuD- or fructose-induced cardiac hypertrophy in 3-week-old male Sprague-Dawley (SD) rats by CuD diet supplemented with or without 30% fructose for 4 weeks. In vitro study was performed by treating cardiomyocytes with tetrathiomolydbate (TM) and fructose. Echocardiography, histology analysis, immunofluorescence, western blotting, and qPCR were performed. KEY FINDINGS: Our findings revealed that CuD caused noticeable cardiac hypertrophy either in the presence or absence of fructose supplement. Fructose exacerbated CuD-induced cardiac remodeling and intramyocardial lipid accumulation. Furthermore, we presented that the inhibition of autophagic flux caused by Ca2+ disturbance is the key mechanism by which CuD- or fructose-induced cardiac remodeling. The reduced expression of sarcoplasmic/endoplasmic reticulum Ca2+ ATPase 2a (SERCA2a) in cardiomyocytes accounts for the elevated cytoplasmic Ca2+ concentration. CONCLUSIONS: Collectively, our study suggested that fructose aggravated CuD-induced cardiac remodeling through the blockade of autophagic flux via SERCA2a decreasing-induced Ca2+ imbalance.


Cardiomegaly , Copper , Fructose , Myocytes, Cardiac , Rats, Sprague-Dawley , Sarcoplasmic Reticulum Calcium-Transporting ATPases , Ventricular Remodeling , Animals , Fructose/adverse effects , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Male , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/drug effects , Ventricular Remodeling/drug effects , Rats , Copper/metabolism , Copper/deficiency , Cardiomegaly/metabolism , Cardiomegaly/etiology , Calcium/metabolism , Disease Models, Animal , Autophagy/drug effects
17.
Nutr Metab Cardiovasc Dis ; 34(2): 281-293, 2024 Feb.
Article En | MEDLINE | ID: mdl-38176960

AIMS: In recent decades, there has been a rise in the consumption of sugars containing fructose, raising concerns about their association with metabolic disorders and obesity. We conducted a systematic review and meta-analysis of randomized controlled trials to assess the effects of a low-fructose diet on anthropometric and metabolic variables. DATA SYNTHESIS: We conducted a systematic review and meta-analysis of randomized controlled trials to assess the effects of low-fructose diets on anthropometric and metabolic factors. Relevant studies were identified by searching electronic databases such as PubMed, Scopus, and Web of Science up to January 2023. The quality of the included studies was assessed using the Cochrane risk-of-bias tool. Ten trials with varying intervention durations (ranging from 4 to 24 weeks) and a total of 750 participants were included. The analysis revealed that a low-fructose diet had no significant effect on weight but did have a significant impact on body mass index (SMD = -0.2; 95 % CI: -0.37, -0.04, P = 0.017) and waist circumference (SMD = -0.48; 95 % CI: -0.67, -0.29, P < 0.0001). Furthermore, a low-fructose diet significantly affected systolic blood pressure (SMD = -0.24; 95 % CI: -0.39, -0.09, P = 0.002), fasting blood glucose (SMD = -0.23; 95 % CI: -0.40, -0.07, P = 0.005), hemoglobin A1c (SMD = -0.62; 95 % CI: -0.93, -0.31, P < 0.0001), and triglyceride levels (SMD = -0.17; 95 % CI: -0.33, -0.02, P = 0.028). However, it had no significant effect on diastolic blood pressure, insulin levels, or homeostatic model assessment of insulin resistance. Subgroup analysis indicated that a low-fructose diet had a greater effect on healthy participants aged over 50 years. CONCLUSIONS: Meta-analysis results suggest that low-fructose diets significantly reduce body mass index, waist circumference, systolic blood pressure, fasting blood glucose, hemoglobin A1c, and triglyceride levels. Additionally, the results of the current study suggest that a low-fructose diet may be more effective in healthy individuals who are older than 50 years old compared to those younger than 50 years old.


Blood Glucose , Fructose , Humans , Middle Aged , Glycated Hemoglobin , Fructose/adverse effects , Triglycerides , Diet/adverse effects
18.
Poult Sci ; 103(3): 103466, 2024 Mar.
Article En | MEDLINE | ID: mdl-38277893

This study analyzed the formation of goose fatty liver due to endoplasmic reticulum stress (ERS) caused by 3 types of sugar. Transcriptome analysis was performed for liver tissues from geese fed a traditional diet (maize flour), geese overfed with traditional diet, and geese overfed with diet supplemented with glucose, fructose, or sucrose. Correlation analysis of the liver tissue transcriptomes showed that differentially expressed genes (DEGs) involved in ERS were significantly negatively correlated with DEGs involved in inflammation response in the sucrose overfeeding group, and significantly positively correlated with the DEGs involved in lipid metabolism in fructose overfeeding group. Goose primary hepatocytes were isolated in vitro and then treated with glucose or fructose. Some were also treated with ERS inhibitor 4-phenylbutyric acid (4-PBA). In the hepatocytes, mRNA expression of X-Box Binding Protein 1 (XBP1), activating transcription factor 6 (AFT6) and glucose-regulated protein 78 (GRP78) genes increased in the two sugar groups (glucose and fructose), but were suppressed by adding 4-PBA. The mRNA expression data, protein kinase contents, and triglyceride (TG) and very low-density lipoprotein (VLDL) concentrations all suggest that ERS regulates lipid deposition induced by glucose and fructose via elevating lipid synthesis, inhibiting fatty acid oxidation, and decreasing lipid transportation. In conclusion, glucose, or fructose cause ERS and then ERS causes lipid deposition in goose primary hepatocytes. Three types of sugar cause lipid accumulation and then lipid accumulation prevents ERS during goose fatty liver formation, which suggests a potential mechanism protects goose livers from ERS. The different sugars may induce lipid deposition in different ways.


Butylamines , Fatty Liver , Geese , Animals , Geese/metabolism , Sugars , Chickens/genetics , Fatty Liver/etiology , Fatty Liver/veterinary , Glucose/metabolism , Triglycerides/metabolism , Fructose/adverse effects , Fructose/metabolism , RNA, Messenger/metabolism , Endoplasmic Reticulum Stress , Sucrose
19.
Nutrients ; 16(1)2024 Jan 02.
Article En | MEDLINE | ID: mdl-38201980

(1) Background: Clinical results on the effects of excess sugar consumption on insulin sensitivity are conflicting, possibly due to differences in sugar type and the insulin sensitivity index (ISI) assessed. Therefore, we compared the effects of consuming four different sugars on insulin sensitivity indices derived from oral glucose tolerance tests (OGTT). (2) Methods: Young adults consumed fructose-, glucose-, high-fructose corn syrup (HFCS)-, sucrose-, or aspartame-sweetened beverages (SB) for 2 weeks. Participants underwent OGTT before and at the end of the intervention. Fasting glucose and insulin, Homeostatic Model Assessment-Insulin Resistance (HOMA-IR), glucose and insulin area under the curve, Surrogate Hepatic Insulin Resistance Index, Matsuda ISI, Predicted M ISI, and Stumvoll Index were assessed. Outcomes were analyzed to determine: (1) effects of the five SB; (2) effects of the proportions of fructose and glucose in all SB. (3) Results: Fructose-SB and the fructose component in mixed sugars negatively affected outcomes that assess hepatic insulin sensitivity, while glucose did not. The effects of glucose-SB and the glucose component in mixed sugar on muscle insulin sensitivity were more negative than those of fructose. (4) Conclusion: the effects of consuming sugar-SB on insulin sensitivity varied depending on type of sugar and ISI index because outcomes assessing hepatic insulin sensitivity were negatively affected by fructose, and outcomes assessing muscle insulin sensitivity were more negatively affected by glucose.


High Fructose Corn Syrup , Insulin Resistance , Young Adult , Humans , Glucose , Glucose Tolerance Test , Aspartame/pharmacology , Zea mays , Sucrose/pharmacology , Fructose/adverse effects , High Fructose Corn Syrup/adverse effects , Beverages , Insulin
20.
Aliment Pharmacol Ther ; 59(4): 475-491, 2024 Feb.
Article En | MEDLINE | ID: mdl-38169126

BACKGROUND: Obesity has reached epidemic proportions, with >40% of the US population affected. Although traditionally managed by lifestyle modification, and less frequently by bariatric therapies, there are significant pharmacological advancements. AIMS: To conduct a narrative review of the neurohormonal and physiological understanding of weight gain and obesity, and the development, clinical testing, indications, expected clinical outcomes, and associated risks of current FDA-approved and upcoming anti-obesity medications (AOMs). METHODS: We conducted a comprehensive review in PubMed for articles on pathophysiology and complications of obesity, including terms 'neurohormonal', 'obesity', 'incretin', and 'weight loss'. Next, we searched for clinical trial data of all FDA-approved AOMs, including both the generic and trade names of orlistat, phentermine/topiramate, bupropion/naltrexone, liraglutide, and semaglutide. Additional searches were conducted for tirzepatide and retatrutide - medications expecting regulatory approval. Searches included combinations of terms related to mechanism of action, indications, side effects, risks, and future directions. RESULTS: We reviewed the pathophysiology of obesity, including specific role of incretins and glucagon. Clinical data supporting the use of various FDA-approved medications for weight loss are presented, including placebo-controlled or, when available, head-to-head trials. Beneficial metabolic effects, including impact on liver disease, adverse effects and risks of medications are discussed, including altered gastrointestinal motility and risk for periprocedural aspiration. CONCLUSION: AOMs have established efficacy and effectiveness for weight loss even beyond 52 weeks. Further pharmacological options, such as dual and triple incretins, are probable forthcoming additions to clinical practice for combating obesity and its metabolic consequences such as metabolic dysfunction-associated steatotic liver disease.


Anti-Obesity Agents , Liver Diseases , Humans , Incretins/therapeutic use , Topiramate/therapeutic use , Fructose/adverse effects , Obesity/drug therapy , Anti-Obesity Agents/adverse effects , Weight Loss , Liver Diseases/drug therapy
...