Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 2.653
1.
Toxicology ; 504: 153798, 2024 May.
Article En | MEDLINE | ID: mdl-38588857

Bisphenol S (BPS) is a commonly detected chemical raw material in water, which poses significant threats to both the ecological environment and human health. Despite being recognized as a typical endocrine disruptor and a substitute for Bisphenol A, the toxicological effects of BPS remain nonnegligible. In order to comprehensively understand the health impacts of BPS, a long-term (154 days) exposure experiment was conducted on mice, during which the physiological indicators of the liver, intestine, and blood were observed. The findings revealed that exposure to BPS resulted in dysbiosis of the gut microbiota, obesity, hepatic lipid accumulation, intestinal lesions, and dyslipidemia. Furthermore, there exists a significant correlation between gut microbiota and indicators of host health. Consequently, the identification of specific gut microbiota can be considered as potential biomarkers for the evaluation of risk associated with BPS. This study will effectively address the deficiency in toxicological data pertaining to BPS. The novel BPS data obtained from this research can serve as a valuable reference for professionals in the field.


Dysbiosis , Dyslipidemias , Gastrointestinal Microbiome , Lipid Metabolism , Liver , Obesity , Phenols , Sulfones , Animals , Phenols/toxicity , Gastrointestinal Microbiome/drug effects , Dyslipidemias/chemically induced , Dysbiosis/chemically induced , Liver/drug effects , Liver/metabolism , Liver/pathology , Mice , Obesity/chemically induced , Obesity/metabolism , Lipid Metabolism/drug effects , Male , Sulfones/toxicity , Endocrine Disruptors/toxicity , Intestines/drug effects , Intestines/microbiology
2.
In Vivo ; 38(3): 1152-1161, 2024.
Article En | MEDLINE | ID: mdl-38688615

BACKGROUND/AIM: The global prevalence of type 2 diabetes (T2D) continues to increase, necessitating the need for understanding the causes of its development. The widespread use of high-fructose corn syrup (HFCS) in drinks and diets is suspected to play a role in metabolic disorders. Although many studies have reported on the effects of excessive HFCS and excessive energy intakes in middle-aged individuals, few have focused on energy restriction. This study aimed to investigate the effects of excessive HFCS drink intake under energy restriction on developing T2D in early middle-aged mice. MATERIALS AND METHODS: Early middle-aged mice were divided in HFCS and control groups; they were provided either 10% HFCS water or deionized water ad libitum for 12 weeks, respectively. Total energy intake was controlled using a standard rodent diet. Oral glucose tolerance test (OGTT), insulin tolerance test (ITT), tissue weight measurements, serum parameter analyses, and mRNA expression assessments were performed. RESULTS: No increase in body and adipose tissue weight was observed with excessive HFCS intake under energy restriction. Moreover, serum lipid parameters did not differ from those of controls. However, in the OGTT and ITT, the HFCS group showed higher blood glucose levels than the control group. Moreover, the pancreatic weight and insulin II mRNA expression were reduced. CONCLUSION: The excessive HFCS drink intake under energy restriction did not induce obesity; however, it induced impaired glucose tolerance, indicating its negative effects on the pancreas in early middle-aged mice. When translated in human physiology, our results show that even if one does not become obese, excessive HFCS may affect the overall metabolic mechanism; these effects may vary depending on age.


Blood Glucose , Glucose Tolerance Test , High Fructose Corn Syrup , Animals , High Fructose Corn Syrup/adverse effects , High Fructose Corn Syrup/administration & dosage , Mice , Male , Diabetes Mellitus, Type 2/chemically induced , Diabetes Mellitus, Type 2/etiology , Diabetes Mellitus, Type 2/metabolism , Energy Intake , Disease Models, Animal , Insulin/blood , Body Weight/drug effects , Insulin Resistance , Obesity/etiology , Obesity/metabolism , Obesity/chemically induced
3.
Obes Rev ; 25(5): e13717, 2024 May.
Article En | MEDLINE | ID: mdl-38463003

Potent incretin-based therapy shows promise for the treatment of obesity along with reduced incidence of cardiovascular events in patients with preexisting cardiovascular disease and obesity. This study assessed the efficacy and safety of the incretin-based obesity treatments, once-weekly subcutaneous semaglutide 2.4 mg and tirzepatide 10 or 15 mg, in people with obesity without diabetes. Of the 744 records identified, seven randomized controlled trials (n = 5140) were included. Five studies (n = 3288) investigated semaglutide and two studies (n = 1852) investigated tirzepatide. The treatment effect, shown as placebo-subtracted difference, on body weight was -15.0% (95% CI, -17.8 to -12.2) with -12.9% (95% CI, -14.7 to -11.1) for semaglutide and -19.2% (95% CI, -22.2 to -16.2) for tirzepatide. The treatment effect on waist circumference was -11.4 cm (95% CI, -13.7 to -9.2) with -9.7 cm (95% CI, -10.8 to -8.5) for semaglutide and -14.6 cm (95% CI, -15.8 to -13.4) for tirzepatide. The adverse events related to semaglutide and tirzepatide were primarily of mild-to-moderate severity and mostly gastrointestinal, which was more frequent during the dose-titration period and leveled off during the treatment period. This emphasizes that once-weekly subcutaneous semaglutide 2.4 mg and tirzepatide 10 or 15 mg induce large reductions in body weight and waist circumference and are generally well-tolerated.


Diabetes Mellitus, Type 2 , Gastric Inhibitory Polypeptide , Glucagon-Like Peptide-2 Receptor , Glucagon-Like Peptides , Incretins , Humans , Incretins/therapeutic use , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/epidemiology , Waist Circumference , Body Weight , Obesity/drug therapy , Obesity/chemically induced , Glucagon-Like Peptide-1 Receptor
4.
Diabetes Obes Metab ; 26(5): 1850-1867, 2024 May.
Article En | MEDLINE | ID: mdl-38468148

There are conflicting data on the weight-reducing potential of metformin (MTF) in nondiabetic patients with obesity. The purpose of this systematic review and meta-analysis was to evaluate the effect of MTF on weight and cardiometabolic parameters in adults with overweight/obesity with or without nonalcoholic fatty liver disease (NAFLD) (CRD42018085512). We included randomized controlled trials (RCTs) in adults without diabetes mellitus, with mean body mass index (BMI) ≥ 25 kg/m2, with or without NAFLD, comparing MTF to placebo/control, lifestyle modification (LSM) or a US Food and Drug Administration-approved anti-obesity drug, reporting on weight or metabolic parameters, and extending over at least 3 months. We conducted a systematic search in MEDLINE, EMBASE, PubMed and the Cochrane Library without time limitation (until March 2022). We screened and selected eligible articles, abstracted relevant data, and assessed the risk of bias. All steps were in duplicate and independently. We conducted a random-effects model meta-analysis using Review Manager version 5.3, with prespecified subgroup analyses in case of heterogeneity. We identified 2650 citations and included 49 trials (55 publications). Compared to placebo, MTF was associated with a significant reduction in BMI (mean difference [MD] -0.56 [-0.74, -0.37] kg/m2; p < 0.0001), at doses ranging from 500 to 2550 mg/day, and with a significant percentage change in BMI of -2.53% (-2.90, -2.17) at the dose 1700 mg/day. There was no interaction by baseline BMI, MTF dose or duration, nor presence or absence of NAFLD. There was no significant difference between MTF and LSM. Orlistat was more effective than MTF (at doses of 1000-1700 mg/day) in terms of weight loss, with an MD in BMI of -3.17 (-5.88; -0.47) kg/m2, favouring the former. Compared to placebo/control, MTF improved insulin parameters, while no effect was detected when compared to LSM. A few small trials showed heterogenous effects on liver parameters in patients with NAFLD treated with MTF compared to placebo/control. There was a large variability in the expression of outcome measures and RCTs were of low quality. In conclusion, MTF was associated with a modest weight reduction in obese nondiabetic patients. Further high-quality and better powered studies are needed to examine the impact of MTF in patients with insulin resistance and NAFLD.


Metformin , Non-alcoholic Fatty Liver Disease , Adult , Humans , Metformin/therapeutic use , Non-alcoholic Fatty Liver Disease/complications , Non-alcoholic Fatty Liver Disease/drug therapy , Randomized Controlled Trials as Topic , Obesity/complications , Obesity/drug therapy , Obesity/chemically induced , Overweight/complications , Overweight/drug therapy , Weight Loss
5.
Biochem Pharmacol ; 222: 116074, 2024 Apr.
Article En | MEDLINE | ID: mdl-38395265

Olanzapine, a widely prescribed atypical antipsychotic, poses a great risk to the patient's health by fabricating a plethora of severe metabolic and cardiovascular adverse effects eventually reducing life expectancy and patient compliance. Its heterogenous receptor binding profile has made it difficult to point out a specific cause or treatment for the related side effects. Growing body of evidence suggest that transient receptor potential (TRP) channel subfamily Ankyrin 1 (TRPA1) has pivotal role in pathogenesis of type 2 diabetes and obesity. With this background, we aimed to investigate the role of pharmacological manipulations of TRPA1 channels in antipsychotic (olanzapine)-induced metabolic alterations in female mice using allyl isothiocyanate (AITC) and HC-030031 (TRPA1 agonist and antagonist, respectively). It was found that after 6 weeks of treatment, AITC prevented olanzapine-induced alterations in body weight and adiposity; serum, and liver inflammatory markers; glucose and lipid metabolism; and hypothalamic appetite regulation, nutrient sensing, inflammatory and TRPA1 channel signaling regulating genes. Furthermore, several of these effects were absent in the presence of HC-030031 (TRPA1 antagonist) indicating protective role of TRPA1 agonism in attenuating olanzapine-induced metabolic alterations. Supplementary in-depth studies are required to study TRPA1 channel effect on other aspects of olanzapine-induced metabolic alterations.


Acetanilides , Antipsychotic Agents , Diabetes Mellitus, Type 2 , Purines , Transient Receptor Potential Channels , Mice , Humans , Female , Animals , TRPA1 Cation Channel , Olanzapine , Antipsychotic Agents/toxicity , Isothiocyanates/pharmacology , Obesity/chemically induced , Obesity/drug therapy , Liver/metabolism
6.
Int J Biol Macromol ; 262(Pt 1): 130018, 2024 Mar.
Article En | MEDLINE | ID: mdl-38331057

The potential prebiotic feature of Bletilla striata polysaccharides (BSP) has been widely accepted, while the beneficial effect of BSP on high-fat-diet-induced obesity is unclear. Moreover, the "crosstalk" between microbiota and metabolomic profile in high-fat-diet-induced obese mice supplemented with BSP still need to be further explored. The present study attempted to illustrate the effect of BSP and/or composite polysaccharides on high-fat-diet-induced obese mice by combining multi-matrix (feces, urine, liver) metabolomics and gut microbiome. The results showed that BSP and/or composite polysaccharides were able to reduce the abnormal weight gain induced by high-fat diet. A total of 175 molecules were characterized by proton nuclear magnetic resonance (1H NMR) in feces, urine and liver, suggesting that multi-matrix metabolomics could provide a comprehensive view of metabolic regulatory mechanism of BSP in high-fat-diet-induced obese mice. Several pathways were altered in response to BSP supplementation, mainly pertaining to amino acid, purine, pyrimidine, ascorbate and aldarate metabolisms. In addition, BSP ameliorated high-fat-diet-induced imbalanced gut microbiome, by lowering the ratio of Firmicutes/Bacteroidetes. Significant correlations were illustrated between particular microbiota's features and specific metabolites. Overall, the anti-obesity effect of BSP could be attributed to the amelioration of the disorders of gut microbiota and to the regulation of the "gut-liver axis" metabolism.


Diet, High-Fat , Gastrointestinal Microbiome , Animals , Mice , Diet, High-Fat/adverse effects , Mice, Obese , Obesity/etiology , Obesity/chemically induced , Polysaccharides/chemistry , Dietary Supplements , Mice, Inbred C57BL
7.
Int J Biol Macromol ; 263(Pt 2): 130485, 2024 Apr.
Article En | MEDLINE | ID: mdl-38423434

The effects of seaweed cellulose (SC) on high fat-sugar diet (HFSD)-induced glucolipid metabolism disorders in mice and potential mechanisms were investigated. SC was isolated from dealginated residues of giant kelp (Macrocystis pyrifera), with a crystallinity index of 85.51 % and an average particle size of 678.2 nm. Administering SC to C57BL/6 mice at 250 or 500 mg/kg BW/day via intragastric gavage for six weeks apparently inhibited the development of HFSD-induced obesity, dyslipidemia, insulin resistance, oxidative stress and liver damage. Notably, SC intervention partially restored the structure and composition of the gut microbiota altered by the HFSD, substantially lowering the Firmicutes to Bacteroidetes ratio, and greatly increasing the relative abundance of Lactobacillus, Bifidobacterium, Oscillospira, Bacteroides and Akkermansia, which contributed to improved short-chain fatty acid (SCFA) production. Supplementing with a higher dose of SC led to more significant increases in total SCFA (67.57 %), acetate (64.56 %), propionate (73.52 %) and butyrate (66.23 %) concentrations in the rectal contents of HFSD-fed mice. The results indicated that highly crystalline SC microparticles could modulate gut microbiota dysbiosis and ameliorate HFSD-induced obesity and related metabolic syndrome in mice. Furthermore, particle size might have crucial impact on the prebiotic effects of cellulose as insoluble dietary fiber.


Gastrointestinal Microbiome , Hyperlipidemias , Metabolic Diseases , Animals , Mice , Sugars/pharmacology , Cellulose/pharmacology , Mice, Inbred C57BL , Obesity/etiology , Obesity/chemically induced , Fatty Acids, Volatile/metabolism , Diet , Diet, High-Fat/adverse effects
8.
Environ Sci Pollut Res Int ; 31(10): 15872-15884, 2024 Feb.
Article En | MEDLINE | ID: mdl-38302837

Glyphosate-based herbicides (GBH) are the most widely used pesticides globally. Studies have indicated that they may increase the risk of various organic dysfunctions. Herein, we verified whether exposure to GBH during puberty increases the susceptibility of male and female mice to obesity when they are fed a high-fat diet (HFD) in adulthood. From the 4th-7th weeks of age, male and female C57Bl/6 mice received water (CTL group) or 50 mg GBH /kg body weight (BW; GBH group). From the 8th-21st weeks of age, the mice were fed a standard diet or a HFD. It was found that pubertal GBH exposure exacerbated BW gains and hyperphagia induced by HFD, but only in female GBH-HFD mice. These female mice also exhibited high accumulation of perigonadal and subcutaneous fat, as well as reduced lean body mass. Both male and female GBH-HFD displayed hypertrophic white adipocytes. However, only in females, pubertal GBH exposure aggravated HFD-induced fat accumulation in brown adipocytes. Furthermore, GBH increased plasma cortisol levels by 80% in GBH-HFD males, and 180% in GBH-HFD females. In conclusion, pubertal GBH exposure aggravated HFD-induced obesity, particularly in adult female mice. This study provides novel evidence that GBH misprograms lipid metabolism, accelerating the development of obesity when individuals are challenged by a second metabolic stressor, such as an obesogenic diet.


Diet, High-Fat , Herbicides , Mice , Male , Female , Animals , Diet, High-Fat/adverse effects , Glyphosate , Herbicides/toxicity , Obesity/chemically induced , Lipid Metabolism
9.
Hum Exp Toxicol ; 43: 9603271241236346, 2024.
Article En | MEDLINE | ID: mdl-38394684

INTRODUCTION: In bipolar women who took lithium during pregnancy, several epidemiology studies have reported small increases in a rare fetal cardiac defect termed Ebstein's anomaly. METHODS: Behavioral, environmental, and lifestyle-associated risk factors associated with bipolar disorder and health insurance status were determined from an Internet search. The search was conducted from October 1, 2023, through October 14, 2023. The search terms employed included the following: bipolar, bipolar disorder, mood disorders, pregnancy, congenital heart defects, Ebstein's anomaly, diabetes, hypertension, Medicaid, Medicaid patients, alcohol use, cigarette smoking, marijuana, cocaine, methamphetamine, narcotics, nutrition, diet, obesity, body mass index, environment, environmental exposures, poverty, socioeconomic status, divorce, unemployment, and income. No quotes, special fields, truncations, etc., were used in the searches. No filters of any kind were used in the searches. RESULTS: Women who remain on lithium in the United States throughout their pregnancy are likely to be experiencing mania symptoms and/or suicidal ideation refractory to other drugs. Pregnant women administered the highest doses of lithium salts would be expected to have been insufficiently responsive to lower doses. Any small increases in the retrospectively determined risk of fetal cardiac anomalies in bipolar women taking lithium salts cannot be disentangled from potential developmental effects resulting from very high rates of cigarette smoking, poor diet, alcohol abuse, ingestion of illegal drugs like cocaine or opioids, marijuana smoking, obesity, and poverty. CONCLUSIONS: The small risks in fetal cardiac abnormalities reported in the epidemiology literature do not establish a causal association for lithium salts and Ebstein's anomaly.


Cocaine , Ebstein Anomaly , Teratogenesis , Humans , Pregnancy , Female , Lithium/toxicity , Ebstein Anomaly/chemically induced , Ebstein Anomaly/epidemiology , Teratogens , Salts , Retrospective Studies , Antimanic Agents , Obesity/epidemiology , Obesity/chemically induced
10.
Behav Brain Res ; 463: 114885, 2024 Apr 12.
Article En | MEDLINE | ID: mdl-38296202

The main cause of second-generation antipsychotic (SGA)-induced obesity is considered due to the antagonism of serotonin 2c receptors (5-HT2cR) and activation of ghrelin receptor type 1a (GHSR1a) signalling. It is reported that 5-HT2cR interacted with GHSR1a, however it is unknown whether one of the SGA olanzapine alters the 5-HT2cR/GHSR1a interaction, affecting orexigenic neuropeptide signalling in the hypothalamus. We found that olanzapine treatment increased average energy intake and body weight gain in mice; olanzapine treatment also increased orexigenic neuropeptide (NPY) and GHSR1a signaling molecules, pAMPK, UCP2, FOXO1 and pCREB levels in the hypothalamus. By using confocal fluorescence resonance energy transfer (FRET) technology, we found that 5-HT2cR interacted/dimerised with the GHSR1a in the hypothalamic neurons. As 5-HT2cR antagonist, both olanzapine and S242084 decreased the interaction between 5-HT2cR and GHSR1a and activated GHSR1a signaling. The 5-HT2cR agonist lorcaserin counteracted olanzapine-induced attenuation of interaction between 5-HT2cR and GHSR1a and inhibited activation of GHSR1a signalling and NPY production. These findings suggest that 5-HT2cR antagonistic effect of olanzapine in inhibition of the interaction of 5-HT2cR and GHSR1a, activation GHSR1a downstream signaling and increasing hypothalamic NPY, which may be the important neuronal molecular mechanism underlying olanzapine-induced obesity and target for prevention metabolic side effects of antipsychotic management in psychiatric disorders.


Antipsychotic Agents , Neuropeptides , Animals , Mice , Antipsychotic Agents/adverse effects , Hypothalamus/metabolism , Neurons/metabolism , Neuropeptides/metabolism , Obesity/chemically induced , Obesity/metabolism , Olanzapine/adverse effects
11.
Toxicol Sci ; 198(2): 210-220, 2024 Mar 26.
Article En | MEDLINE | ID: mdl-38291899

Microplastics (MP) derived from the weathering of polymers, or synthesized in this size range, have become widespread environmental contaminants and have found their way into water supplies and the food chain. Despite this awareness, little is known about the health consequences of MP ingestion. We have previously shown that the consumption of polystyrene (PS) beads was associated with intestinal dysbiosis and diabetes and obesity in mice. To further evaluate the systemic metabolic effects of PS on the gut-liver-adipose tissue axis, we supplied C57BL/6J mice with normal water or that containing 2 sizes of PS beads (0.5 and 5 µm) at a concentration of 1 µg/ml. After 13 weeks, we evaluated indices of metabolism and liver function. As observed previously, mice drinking the PS-containing water had a potentiated weight gain and adipose expansion. Here we found that this was associated with an increased abundance of adipose F4/80+ macrophages. These exposures did not cause nonalcoholic fatty liver disease but were associated with decreased liver:body weight ratios and an enrichment in hepatic farnesoid X receptor and liver X receptor signaling. PS also increased hepatic cholesterol and altered both hepatic and cecal bile acids. Mice consuming PS beads and treated with the berry anthocyanin, delphinidin, demonstrated an attenuated weight gain compared with those mice receiving a control intervention and also exhibited a downregulation of cyclic adenosine monophosphate (cAMP) and peroxisome proliferator-activated receptor (PPAR) signaling pathways. This study highlights the obesogenic role of PS in perturbing the gut-liver-adipose axis and altering nuclear receptor signaling and intermediary metabolism. Dietary interventions may limit the adverse metabolic effects of PS consumption.


Non-alcoholic Fatty Liver Disease , Plastics , Animals , Mice , Plastics/metabolism , Plastics/pharmacology , Polystyrenes/toxicity , Polystyrenes/metabolism , Microplastics/metabolism , Microplastics/pharmacology , Mice, Inbred C57BL , Liver , Non-alcoholic Fatty Liver Disease/metabolism , Obesity/chemically induced , Obesity/metabolism , Weight Gain
12.
Am J Psychiatry ; 181(1): 26-38, 2024 Jan 01.
Article En | MEDLINE | ID: mdl-38161305

Psychotropic drug-related weight gain (PDWG) is a common occurrence and is highly associated with non-initiation, discontinuation, and dissatisfaction with psychiatric drugs. Moreover, PDWG intersects with the elevated risk for obesity and associated morbidity that has been amply reported in the psychiatric population. Evidence indicates that differential liability for PDWG exists for antipsychotics, antidepressants, and anticonvulsants. During the past two decades, agents within these classes have become available with significantly lower or no liability for PDWG and as such should be prioritized. Although lithium is associated with weight gain, the overall extent of weight gain is significantly lower than previously estimated. The benefit of lifestyle and behavioral modification for obesity and/or PDWG in psychiatric populations is established, with effectiveness similar to that in the general population. Metformin is the most studied pharmacological treatment in the prevention and treatment of PDWG, and promising data are emerging for glucagon-like peptide-1 (GLP-1) receptor agonists (e.g., liraglutide, exenatide, semaglutide). Most pharmacologic antidotes for PDWG are supported with low-confidence data (e.g., topiramate, histamine-2 receptor antagonists). Future vistas for pharmacologic treatment for PDWG include large, adequately controlled studies with GLP-1 receptor agonists and possibly GLP-1/glucose-dependent insulinotropic polypeptide co-agonists (e.g., tirzepatide) as well as specific dietary modifications.


Diabetes Mellitus, Type 2 , Hypoglycemic Agents , Humans , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Diabetes Mellitus, Type 2/drug therapy , Liraglutide/therapeutic use , Weight Gain , Obesity/chemically induced , Obesity/drug therapy , Psychotropic Drugs/adverse effects , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/therapeutic use
13.
Int J Mol Sci ; 25(1)2024 Jan 04.
Article En | MEDLINE | ID: mdl-38203845

Excess body weight constitutes one of the major health challenges for societies and healthcare systems worldwide. Besides the type of diet, calorie intake and the lack of physical exercise, recent data have highlighted a possible association between endocrine-disrupting chemicals (EDCs), such as bisphenol A, phthalates and their analogs, and obesity. EDCs represent a heterogeneous group of chemicals that may influence the hormonal regulation of body mass and adipose tissue morphology. Based on the available data from mechanistic, animal and epidemiological studies including meta-analyses, the weight of evidence points towards the contribution of EDCs to the development of obesity, associated disorders and obesity-related adipose tissue dysfunction by (1) impacting adipogenesis; (2) modulating epigenetic pathways during development, enhancing susceptibility to obesity; (3) influencing neuroendocrine signals responsible for appetite and satiety; (4) promoting a proinflammatory milieu in adipose tissue and inducing a state of chronic subclinical inflammation; (5) dysregulating gut microbiome and immune homeostasis; and (6) inducing dysfunction in thermogenic adipose tissue. Critical periods of exposure to obesogenic EDCs are the prenatal, neonatal, pubertal and reproductive periods. Interestingly, EDCs even at low doses may promote epigenetic transgenerational inheritance of adult obesity in subsequent generations. The aim of this review is to summarize the available evidence on the role of obesogenic EDCs, specifically BPA and phthalate plasticizers, in the development of obesity, taking into account in vitro, animal and epidemiologic studies; discuss mechanisms linking EDCs to obesity; analyze the effects of EDCs on obesity in critical chronic periods of exposure; and present interesting perspectives, challenges and preventive measures in this research area.


Benzhydryl Compounds , Endocrine Disruptors , Phenols , Phthalic Acids , Animals , Female , Pregnancy , Endocrine Disruptors/toxicity , Obesity/chemically induced , Weight Gain , Humans
14.
Environ Sci Pollut Res Int ; 31(6): 8291-8311, 2024 Feb.
Article En | MEDLINE | ID: mdl-38165541

Arsenic-contaminated drinking water can induce various disorders by disrupting lipid and glucose metabolism in adipose tissue, leading to insulin resistance. It inhibits adipocyte development and exacerbates insulin resistance, though the precise impact on lipid synthesis and lipolysis remains unclear. This review aims to explore the processes and pathways involved in adipogenesis and lipolysis within adipose tissue concerning arsenic-induced diabetes. Although arsenic exposure is linked to type 2 diabetes, the specific role of adipose tissue in its pathogenesis remains uncertain. The review delves into arsenic's effects on adipose tissue and related signaling pathways, such as SIRT3-FOXO3a, Ras-MAP-AP-1, PI(3)-K-Akt, endoplasmic reticulum stress proteins, CHOP10, and GPCR pathways, emphasizing the role of adipokines. This analysis relies on existing literature, striving to offer a comprehensive understanding of different adipokine categories contributing to arsenic-induced diabetes. The findings reveal that arsenic detrimentally impacts white adipose tissue (WAT) by reducing adipogenesis and promoting lipolysis. Epidemiological studies have hinted at a potential link between arsenic exposure and obesity development, with limited research suggesting a connection to lipodystrophy. Further investigations are needed to elucidate the mechanistic association between arsenic exposure and impaired adipose tissue function, ultimately leading to insulin resistance.


Arsenic , Diabetes Mellitus, Type 2 , Insulin Resistance , Humans , Arsenic/toxicity , Diabetes Mellitus, Type 2/metabolism , Obesity/chemically induced , Adipose Tissue/metabolism , Lipids/toxicity
15.
Sci Total Environ ; 914: 169919, 2024 Mar 01.
Article En | MEDLINE | ID: mdl-38199361

Di-2-ethylhexyl phthalate (DEHP), a widely utilized plasticizer, has been described as a potential obesogen based on in vivo disruption of hepatic lipid homeostasis and in vitro promotion of lipid accumulation. However, limited literature exists regarding the specific ramifications of DEHP exposure on obese individuals, and the precise mechanisms underlying the adverse effects of DEHP exposure remain unclear. This study aimed to assess the impact of DEHP on hepatic lipid metabolism in obese mice by comparing them to normal mice. Following a 10-week DEHP exposure period, the obese mice exhibited higher blood lipid levels, more severe hepatic steatosis, and more infiltrations of inflammatory cells in liver tissue than normal mice. Interestingly, the body weight of the mice exhibited no significant alteration. In addition, transcriptomic analyses revealed that both lipogenesis and fatty acid oxidation contributed to hepatic lipid metabolism dysregulation following DEHP exposure. More specifically, alterations in the transcription of genes associated with hepatic lipid metabolism were linked to the different responses to DEHP exposure observed in normal and obese mice. Additionally, the outcomes of in vitro experiments validated the in vivo findings and demonstrated that DEHP exposure could modify hepatic lipid metabolism in normal mice by activating the LXR/SREBP-1c signaling pathway to promote lipogenesis. At the same time, DEHP exposure led to inhibition of the Camkkß/AMPK pathway to suppress ß-fatty acid oxidation. Conversely, in obese mice, DEHP exposure was found to be associated with the stimulation of both lipogenesis and fatty acid oxidation via activation of the LXR/SREBP-1c and PPAR-α signaling pathways, respectively. The findings presented in this study first elucidate the contrasting mechanisms underlying DEHP-induced liver damage in obese and normal mice, thereby offering valuable insights into the pathogenesis of DEHP-induced liver damage in individuals with obesity.


Diethylhexyl Phthalate , Lipid Metabolism , Phthalic Acids , Animals , Mice , Diethylhexyl Phthalate/metabolism , Fatty Acids/metabolism , Lipids , Liver/metabolism , Mice, Obese , Obesity/chemically induced , Obesity/metabolism , Peroxisome Proliferator-Activated Receptors/metabolism , Signal Transduction , Sterol Regulatory Element Binding Protein 1/metabolism
16.
Diabetes Obes Metab ; 26(4): 1346-1354, 2024 Apr.
Article En | MEDLINE | ID: mdl-38240127

AIM: To identify the sociodemographic, clinical and laboratory determinants relating to patient adherence to liraglutide treatment among individuals with overweight or obesity. METHODS: We retrospectively analysed patients with overweight or obesity who were treated with liraglutide between 2019 and 2022. Over a 6-month follow-up period, measurements of body mass index, sociodemographic characteristics, clinical and laboratory data, and prescription records for liraglutide were collected. Treatment adherence was assessed using the proportion of days covered (PDC) measure, with a PDC ≥80% indicating high adherence. RESULTS: The study population included 1890 participants (78.1% female, mean age 46 ± 12 years). At the end of the follow-up period, 84.9% of the participants exhibited low adherence to liraglutide treatment. Adherence to treatment improved with age (p = 0.04, odds ratio [OR] 1.013, confidence interval [CI] 1.001-1.025). Significant weight loss during treatment increased the likelihood of high adherence (p < 0.001, OR 1.251, CI 1.167-1.341). Individuals with a higher socioeconomic status displayed greater adherence (p = 0.023, OR 1.906, CI 1.091-3.328). Greater adherence was also seen in non-smokers (p = 0.047, OR 0.725, CI 0.528-0.996). CONCLUSIONS: Only 15.1% of study participants exhibited high adherence to treatment (PDC ≥80%) after 6 months of follow-up. Further research is needed to explore approaches to enhance adherence to liraglutide, including strategies to educate and support patients in their efforts to achieve and maintain weight loss with the use of this drug.


Diabetes Mellitus, Type 2 , Liraglutide , Humans , Female , Adult , Middle Aged , Male , Liraglutide/therapeutic use , Overweight/complications , Overweight/drug therapy , Overweight/chemically induced , Hypoglycemic Agents/therapeutic use , Retrospective Studies , Diabetes Mellitus, Type 2/drug therapy , Obesity/complications , Obesity/drug therapy , Obesity/chemically induced , Weight Loss
17.
Environ Sci Pollut Res Int ; 31(1): 549-563, 2024 Jan.
Article En | MEDLINE | ID: mdl-38015390

Exposure to particulate matter (PM) has been linked to metabolic diseases. However, the effects of PM with an aerodynamic diameter ≤ 1.0 µm (PM1) on metabolic diseases remain unclear. This study is aimed at assessing the associations of PM1 with metabolic disease risk and quantifying the concentration-response (C-R) relationship of PM1 with metabolic disease risk. A national cross-sectional study was conducted, including 12,495 middle-aged and older adults in 123 Chinese cities. The two-year average concentration of PM1 was evaluated using satellite-based spatiotemporal models. Metabolic diseases, including abdominal obesity, diabetes, hypertension, dyslipidemia, and metabolic syndrome, were identified based on physical examination, blood standard biochemistry examination, and self-reported disease histories. Generalized linear models and C-R curves were used to evaluate the associations of PM1 with metabolic diseases. A total of 12,495 participants were included in this study, with a prevalence of 45.73% for abdominal obesity, 20.22% for diabetes, 42.46% for hypertension, 41.01% for dyslipidemia, and 33.78% for metabolic syndrome. The mean ± standard deviation age of participants was 58.79 ± 13.14 years. In addition to dyslipidemia, exposure to PM1 was associated with increased risks of abdominal obesity, diabetes, hypertension, and metabolic syndrome. Each 10 µg/m3 increase in PM1 concentrations was associated with 39% (odds ratio (OR) = 1.39, 95% confidence interval (CI) 1.33, 1.46) increase in abdominal obesity, 18% (OR = 1.18, 95%CI 1.12, 1.25) increase in diabetes, 11% (OR = 1.11, 95%CI 1.06, 1.16) increase in hypertension, and 25% (OR = 1.25, 95%CI 1.19, 1.31) in metabolic syndrome, respectively. C-R curves showed that the OR values of abdominal obesity, diabetes, hypertension, and metabolic syndrome were increased gradually with the increase of PM1 concentrations. Subgroup analysis indicated that exposure to PM1 was associated with increased metabolic disease risks among participants with different lifestyles and found that solid fuel users were more susceptible to PM1 than clean fuel users. This national cross-sectional study indicated that exposure to higher PM1 might increase abdominal obesity, diabetes, hypertension, and metabolic syndrome risk, and solid fuel use might accelerate the adverse effects of PM1 on metabolic syndrome risk. Further longitudinal cohort studies are warranted to establish a causal inference between PM1 exposure and metabolic disease risk.


Air Pollutants , Air Pollution , Diabetes Mellitus , Dyslipidemias , Hypertension , Metabolic Diseases , Metabolic Syndrome , Middle Aged , Humans , Aged , Particulate Matter/analysis , Prevalence , Metabolic Syndrome/epidemiology , Cross-Sectional Studies , Obesity, Abdominal/epidemiology , Obesity, Abdominal/chemically induced , Cities , Hypertension/epidemiology , Hypertension/chemically induced , Metabolic Diseases/epidemiology , Obesity/chemically induced , Diabetes Mellitus/chemically induced , Dyslipidemias/epidemiology , Dyslipidemias/chemically induced , Environmental Exposure/analysis , Air Pollutants/analysis , China/epidemiology , Air Pollution/analysis
18.
Environ Res ; 242: 117637, 2024 Feb 01.
Article En | MEDLINE | ID: mdl-37993047

BACKGROUND: Exposure to air pollution is associated with adverse cardiometabolic health effects and increased mortality, even at low concentrations. Some of the biological mechanisms through which air pollution can affect cardiometabolic health overlap with health outcomes associated with diet quality and changes in diet. OBJECTIVE: The objective of this study is to investigate associations of air pollutants at average concentrations below the World Health Organization, 2021 air quality guidelines with cardiometabolic outcomes. Furthermore, potential interaction between air pollutants and diet quality will be assessed. METHODS: 82 individuals with obesity participated in a combined weight loss and weight loss maintenance study for a total of 33 weeks. A secondary analysis was conducted incorporating air pollution measurements. Data were analysed with linear mixed-effects models. RESULTS: A total of 17 significant associations were observed for single pollutants with 10 cardiometabolic outcomes, predominantly related to blood lipids, hormones, and glucose regulation. Diet quality, as measured by the Baltic Sea Diet score, did not appear to mediate the association of air pollution with cardiometabolic outcomes, however, diet quality was observed to significantly modify the association of PM2.5 with total cholesterol, and the associations of NO and O3 with ghrelin. DISCUSSION: These findings suggest that exposure to ambient air pollutants, especially particulate matter, at levels below World Health Organization, 2021 air quality guidelines, were associated with changes in cardiometabolic risk factors. Diet may be a personal-level approach for individuals to modify the impact of exposure to air pollution on cardiometabolic health.


Air Pollutants , Air Pollution , Cardiovascular Diseases , Humans , Air Pollution/adverse effects , Air Pollution/analysis , Air Pollutants/toxicity , Particulate Matter/analysis , Obesity/epidemiology , Obesity/chemically induced , Diet , Weight Loss , Environmental Exposure/analysis
19.
J Biochem Mol Toxicol ; 38(1): e23561, 2024 Jan.
Article En | MEDLINE | ID: mdl-37942807

The incidence of metabolic diseases is increasing alarmingly in recent times. Parallel to nutritional excess and sedentary lifestyle, the random usage of several endocrine disrupting chemicals including plasticizers is reported to be closely associated with metabolic diseases. Diethyl phthalate (DEP) is a widely used plasticizer in a host of consumer and daily care products. Adipose tissue plays a central role in energy storage and whole-body metabolism. The impairment of adipose function is critically implicated in the pathogenesis of insulin resistance, diabetes, and related metabolic diseases. Recently, exposure to certain phthalate esters has been linked to the development of obesity and diabetes, although there are contradictions and the mechanisms are not clearly understood. In an effort to ascertain the metabolic consequences of chronic phthalate exposure and the underlying mechanism, the present study was designed to examine the effects of long-term dietary consumption of DEP in adipocytes. DEP-treated mice were hyperglycemic but nonobese; their body weight initially increased which subsequently was reduced compared to control. DEP exposure at lower levels impaired adipogenesis by downregulating the key transcription factor, peroxisome proliferator-activated receptor γ and its downstream insulin-sensitizing adipokine, adiponectin, thereby severely compromising adipocyte function. The activation of master regulator nuclear factor κB led to rise in proinflammatory cytokines. We found that DEP triggered intrinsic apoptotic pathways through activated cytochrome c-Apaf1-caspase 9-caspase 3 axis in adipocytes. Taken together, our data revealed that chronic administration of dietary DEP could unleash adverse metabolic outcomes by initiating oxidative stress, inflammation, and apoptosis in the adipocytes, thus leading to adipose tissue dysfunction.


Diabetes Mellitus , Insulin Resistance , Phthalic Acids , Mice , Animals , Plasticizers/toxicity , Plasticizers/metabolism , Adipocytes , Obesity/chemically induced , Obesity/metabolism , Diabetes Mellitus/metabolism , Apoptosis , Inflammation/chemically induced , Inflammation/metabolism
20.
Toxicol Appl Pharmacol ; 482: 116788, 2024 01.
Article En | MEDLINE | ID: mdl-38086441

Environmental chemicals, such as plasticizers, have been linked to increased rates of obesity, according to epidemiological studies. Acetyl triethyl citrate (ATEC) is a plasticizer that is commonly utilized in pharmaceutical products and food packaging as a non-phthalate alternative. Due to its direct contact with the human body and high leakage rate from the polymers, assessment of the potential risk of ATEC exposure at environmentally relevant low doses to human health is needed. Male C57BL/6 J mice were fed diets containing ATEC at doses of either 0.1 or 10 µg/kg per day in a period of 12 weeks to mimic the real exposure environment. The findings suggest that in C57BL/6 J mice, ATEC exposure resulted in increased body weight gain, body fat percentage, and benign hepatocytes, as well as adipocyte size. Consistent with in vivo models, ATEC treatment obviously stimulated the increase of intracellular lipid load in both mouse and human hepatocytes. Mechanically, ATEC induced the transcriptional expression of genes involved in de novo lipogenesis and lipid uptake. Using both enzyme inhibitor and small interfering RNA (siRNA) transfection, we found that stearoyl-coenzyme A desaturase 1 (SCD1) played a significant role in ATEC-induced intracellular lipid accumulation. This study for the first time provided initial evidence suggesting the obesogenic and fatty liver-inducing effect of ATEC at low doses near human exposure levels, and ATEC might be a potential environmental obesogen and its effect on human health need to be further evaluated.


Citrates , Lipogenesis , Plasticizers , Male , Mice , Humans , Animals , Plasticizers/toxicity , Mice, Inbred C57BL , Obesity/chemically induced , Obesity/metabolism , Lipids , Liver , Stearoyl-CoA Desaturase/genetics , Stearoyl-CoA Desaturase/metabolism
...