Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Crit Rev Biochem Mol Biol ; 56(5): 455-481, 2021 10.
Article in English | MEDLINE | ID: mdl-34182855

ABSTRACT

Over accumulation of lipids in adipose tissue disrupts metabolic homeostasis by affecting cellular processes. Endoplasmic reticulum (ER) stress is one such process affected by obesity. Biochemical and physiological alterations in adipose tissue due to obesity interfere with adipose ER functions causing ER stress. This is in line with increased irregularities in other cellular processes such as inflammation and autophagy, affecting overall metabolic integrity within adipocytes. Additionally, microRNAs (miRNAs), which can post-transcriptionally regulate genes, are differentially modulated in obesity. A better understanding and identification of such miRNAs could be used as novel therapeutic targets to fight against diseases. In this review, we discuss ways in which ER stress participates as a common molecular process in the pathogenesis of obesity-associated metabolic disorders. Moreover, our review discusses detailed underlying mechanisms through which ER stress and miRNAs contribute to metabolic alteration in adipose tissue in obesity. Hence, identifying mechanistic involvement of miRNAs-ER stress cross-talk in regulating adipose function during obesity could be used as a potential therapeutic approach to combat chronic diseases, including obesity.


Subject(s)
MicroRNAs , Adipose Tissue , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum Stress/genetics , Humans , MicroRNAs/genetics , Obesity/genetics
2.
Int J Mol Sci ; 24(10)2023 May 13.
Article in English | MEDLINE | ID: mdl-37240054

ABSTRACT

Uncoupling protein 1 (UCP1) plays a central role in thermogenic tissues by uncoupling cellular respiration to dissipate energy. Beige adipocytes, an inducible form of thermogenic cells in subcutaneous adipose tissue (SAT), have become a major focus in obesity research. We have previously shown that eicosapentaenoic acid (EPA) ameliorated high-fat diet (HFD)-induced obesity by activating brown fat in C57BL/6J (B6) mice at thermoneutrality (30 °C), independently of UCP1. Here, we investigated whether ambient temperature (22 °C) impacts EPA effects on SAT browning in wild-type (WT) and UCP1 knockout (KO) male mice and dissected underlying mechanisms using a cell model. We observed resistance to diet-induced obesity in UCP1 KO mice fed HFD at ambient temperature, with significantly higher expression of UCP1-independent thermogenic markers, compared to WT mice. These markers included the fibroblast growth factor 21 (FGF21) and sarco/endoplasmic reticulum Ca2+-ATPase 2b (SERCA2b), suggesting the indispensable role of temperature in beige fat reprogramming. Surprisingly, although EPA induced thermogenic effects in SAT-derived adipocytes harvested from both KO and WT mice, EPA only increased thermogenic gene and protein expression in the SAT of UCP1 KO mice housed at ambient temperature. Collectively, our findings indicate that the thermogenic effects of EPA, which are independent of UCP1, occur in a temperature-dependent manner.


Subject(s)
Adipose Tissue, Brown , Eicosapentaenoic Acid , Male , Animals , Mice , Temperature , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism , Eicosapentaenoic Acid/pharmacology , Eicosapentaenoic Acid/metabolism , Mice, Knockout , Mice, Inbred C57BL , Adipose Tissue, Brown/metabolism , Obesity/metabolism , Subcutaneous Fat/metabolism , Thermogenesis/genetics , Adipose Tissue, White/metabolism
3.
J Nutr ; 150(7): 1693-1704, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32271912

ABSTRACT

Inflammation is largely mediated by immune cells responding to invading pathogens, whereas metabolism is oriented toward producing usable energy for vital cell functions. Immunometabolic alterations are considered key determinants of chronic inflammation, which leads to the development of chronic diseases. Studies have demonstrated that macrophages and the NOD-, LRR-, and pyrin domain-containing protein 3 (NLRP3) inflammasome are activated in key metabolic tissues to contribute to increased risk for type 2 diabetes mellitus, Alzheimer disease, and liver diseases. Thus, understanding the tissue-/cell-type-specific regulation of the NLRP3 inflammasome is crucial for developing intervention strategies. Currently, most of the nutrients and bioactive compounds tested to determine their inflammation-reducing effects are limited to animal models. Future studies need to address how dietary compounds regulate immune and metabolic cell reprograming in humans.


Subject(s)
Gene Expression Regulation/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Brain/immunology , Humans , Inflammasomes , Liver/immunology , Macrophages , NLR Family, Pyrin Domain-Containing 3 Protein/genetics
4.
Proc Natl Acad Sci U S A ; 114(1): E67-E74, 2017 01 03.
Article in English | MEDLINE | ID: mdl-27994150

ABSTRACT

Double-positive (DP) thymocytes respond to intrathymic T-cell receptor (TCR) signals by undergoing positive selection and lineage differentiation into single-positive (SP) mature cells. Concomitant with these well-characterized events is the acquisition of a mature T-cell gene expression program characterized by the induction of the effector molecules IL-7Rα, S1P1, and CCR7, but the underlying mechanism remains elusive. We report here that transcription repressor Growth factor independent 1 (Gfi1) orchestrates the fidelity of the DP gene expression program and developmental maturation into SP cells. Loss of Gfi1 resulted in premature induction of effector genes and the transcription factors forkhead box protein O1 (Foxo1) and Klf2 in DP thymocytes and the accumulation of postselection intermediate populations and accelerated transition into SP cells. Strikingly, partial loss of Foxo1 function, but not restored survival fitness, rectified the dysregulated gene expression and thymocyte maturation in Gfi1-deficient mice. Our results establish the Gfi1-Foxo1 axis and the transcriptional circuitry that actively maintain DP identity and shape the proper generation of mature T cells.


Subject(s)
CD4-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/cytology , Cell Differentiation/genetics , DNA-Binding Proteins/genetics , Forkhead Box Protein O1/genetics , Gene Expression Regulation/immunology , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Transcription Factors/genetics , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , DNA-Binding Proteins/metabolism , Forkhead Box Protein O1/metabolism , Gene Expression Regulation/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Antigen, T-Cell/immunology , Thymus Gland/cytology , Transcription Factors/metabolism , Transcription, Genetic/genetics , Transcriptional Activation/genetics
5.
Ceylon Med J ; 64(3): 82-90, 2019 Sep 30.
Article in English | MEDLINE | ID: mdl-32115403

ABSTRACT

Introduction: Adolescents have high vulnerability for body image distortions which may result in many psychological and physical problems. Objectives: This study aimed to determine body image perception and dissatisfaction among rural schooling adolescents in Sri Lanka. Methods: Adolescents (aged 13-16 years) were selected using probability-based sampling framework. Self-administered figure rating scale was used to assess perceived current (PCBS) and ideal (IBS) body size and body discrepancy score (BDS = PCBS ­ IBS). Pubertal stage was assessed with pubertal development scale. Anthropometry was done according to WHO guidelines. Results: Of 3128 students studied, 47.8% were boys. Median PCBS was 4 (inter quartile range 1) for boys and 4 (2) for girls. Boys aspire a larger body size compared to girls and the respective median IBS were 5 (0) and 4 (1). More boys (70.4%, n = 1053) than girls (66.4%, n = 1084) were dissatisfied about their current body size; χ2 = 5.5, df = 1, p < 0.05. Overweight students recorded higher PCBS and were more dissatisfied with their body size when compared to others. Nearly half of boys and 29.6% girls wanted to have a larger body size than their current. The main influencing factor of body dissatisfaction was body mass index. Conclusions: Body dissatisfaction is evident even among rural adolescents, affecting boys more than girls. Many students show a desire to have a larger body size than their current. This study also provides valid and reliable tools to assess body dissatisfaction and pubertal development in Sinhala speaking adolescents.


Subject(s)
Body Dissatisfaction/psychology , Body Image/psychology , Rural Population/statistics & numerical data , Students/psychology , Adolescent , Female , Humans , Male , Psychiatric Status Rating Scales , Sri Lanka
6.
Ceylon Med J ; 64(3): 111-117, 2019 Sep 30.
Article in English | MEDLINE | ID: mdl-32120461

ABSTRACT

Introduction: Visceral adiposity index (VAI) is a mathematical index derived from the body mass index (BMI), waist circumference (WC), serum fasting triglyceride (TG) and high-density lipoprotein cholesterol (HDL-C). It reflects visceral adipocyte dysfunction (VAD) and is associated with cardiometabolic risk. Women with polycystic ovarian syndrome (PCOS) have adipocyte dysfunction, which is associated with metabolic disorders. Hirsutism in PCOS is considered to be due to high insulin levels which enhances androgen activity at the pilosebaceous unit. Objectives: To determine the association between VAI, hirsutism and cardiometabolic risk factors in patients with PCOS. Methods: A total of 99 patients aged 18-40 years with PCOS diagnosed by the Rotterdam consensus criteria-2003 and a hirsutism score of 8 or more according to the Ferriman-Gallway Score (FGS) were studied. BMI, WC, fasting lipid profile, serum leptin, insulin, sex hormone binding globulin (SHBG), free-androgen index (FAI), fasting blood glucose (FBG) and oral glucose tolerance test (OGTT) were determined. Homeostasis model assessment (HOMA)-beta, HOMA-insulin resistance (IR) and VAI were calculated. Diameter and rate of hair growth at sideburns and chin; density of hair at sideburns were measured. Data were analyzed by SPSS-22.0. Results: There was no significant association between parameters of hirsutism and VAI. There was a significant association between VAI and OGTT, FAI, systolic and diastolic blood pressure: but not between VAI and other metabolic parameters. Conclusion: Visceral adipocyte dysfunction is closely linked to glucose intolerance and blood pressure in women with PCOS. However, hirsutism is unlikely to be due to adipocyte dysfunction.


Subject(s)
Glucose Intolerance/etiology , Hirsutism/etiology , Intra-Abdominal Fat/physiopathology , Obesity, Abdominal/etiology , Polycystic Ovary Syndrome/physiopathology , Adolescent , Adult , Blood Pressure , Body Mass Index , Cross-Sectional Studies , Fasting/blood , Female , Humans , Insulin/blood , Polycystic Ovary Syndrome/blood , Polycystic Ovary Syndrome/complications , Risk Factors , Triglycerides/blood , Waist Circumference , Young Adult
7.
Proc Natl Acad Sci U S A ; 110(34): E3198-205, 2013 Aug 20.
Article in English | MEDLINE | ID: mdl-23918371

ABSTRACT

Foxp3(+) regulatory T (T(reg)) cells are essential for the maintenance of self-tolerance and immune homeostasis. The majority of T(reg) cells is generated in the thymus as a specific subset of CD4(+) T cells, known as thymus-derived or natural T(reg) (nT(reg)) cells, in response to signals from T-cell receptors, costimulatory molecules, and cytokines. Recent studies have identified intracellular signaling and transcriptional pathways that link these signals to Foxp3 induction, but how the production of these extrinsic factors is controlled remains poorly understood. Here, we report that the transcription repressor growth factor independent 1 (Gfi1) has a key inhibitory role in the generation of nT(reg) cells by a noncell-autonomous mechanism. T cell-specific deletion of Gfi1 results in aberrant expansion of thymic nT(reg) cells and increased production of cytokines. In particular, IL-2 overproduction plays an important role in driving the expansion of nT(reg) cells. In contrast, although Gfi1 deficiency elevated thymocyte apoptosis, Gfi1 repressed nT(reg) generation independently of its prosurvival effect. Consistent with an inhibitory role of Gfi1 in this process, loss of Gfi1 dampens antitumor immunity. These data point to a previously unrecognized extrinsic control mechanism that negatively shapes thymic generation of nT(reg) cells.


Subject(s)
DNA-Binding Proteins/immunology , Homeostasis/immunology , T-Lymphocytes, Regulatory/immunology , Thymus Gland/immunology , Transcription Factors/immunology , Analysis of Variance , Animals , Cells, Cultured , DNA-Binding Proteins/genetics , Flow Cytometry , Interleukin-2/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Knockout , Real-Time Polymerase Chain Reaction , Transcription Factors/genetics
8.
J Nutr ; 145(3): 411-7, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25733455

ABSTRACT

BACKGROUND: Obesity is associated with an overexpansion of adipose tissue, along with increases in blood pressure, glycemia, inflammation, and thrombosis. Research to develop nutritional interventions to prevent or treat obesity and its associated diseases is greatly needed. Previously, we demonstrated the ability of eicosapentaenoic acid (EPA) to prevent high-fat (HF) diet-induced obesity, insulin resistance, and inflammation in mice. OBJECTIVE: The objective of the current study was to determine the mechanisms mediating the anti-inflammatory and antilipogenic actions of EPA. METHODS: In a previous study, male C57BL/6J mice were fed a low-fat diet (10% of energy from fat), an HF diet (45% of energy from fat), or an HF diet supplemented with EPA (45% of energy from fat; 36 g/kg EPA; HF+EPA) for 11 wk or an HF diet for 6 wk and then switched to the HF+EPA diet for 5 wk. In this study, we used histology/immunohistochemistry, gene expression, and metabolomic analyses of white adipose tissue from these mice. In addition, cultured mouse 3T3-L1 adipocytes were treated with 100 µM EPA for 48 h and then used for extracellular flux assays with untreated 3T3-L1 adipocytes used as a control. RESULTS: Compared with the HF diet, the HF+EPA diet significantly reduced body weight, adiposity, adipocyte size, and macrophage infiltration into adipose tissue. No significant differences in overall body weight or fat pad weights were observed between HF-fed mice vs. those fed the HF+EPA diet for a short time after first inducing obesity with the HF diet. Interestingly, both histology and immunohistochemistry results showed a significantly lower mean adipocyte size and macrophage infiltration in mice fed the HF diet and then switched to the HF+EPA diet vs. those fed HF diets only. This indicated that EPA was able to prevent as well as reverse HF-diet-induced adipocyte inflammation and hypertrophy and that some of the metabolic effects of EPA were independent of body weight or adiposity. In addition, adipose tissue metabolomic data and cultured adipocyte extracellular flux bioenergetic assays indicated that EPA also regulated mitochondrial function by increasing fatty acid oxidation and oxygen consumption, respectively. CONCLUSION: With the use of mice and cultured adipocytes, we showed that EPA ameliorates HF-diet effects at least in part by increasing oxygen consumption and fatty acid oxidation and reducing adipocyte size, adipogenesis, and adipose tissue inflammation, independent of obesity.


Subject(s)
Adipocytes/drug effects , Diet, High-Fat/adverse effects , Eicosapentaenoic Acid/pharmacology , Inflammation/drug therapy , 3T3 Cells , Adiponectin/blood , Adipose Tissue/drug effects , Adipose Tissue/pathology , Adipose Tissue, White , Adiposity/drug effects , Animals , Body Weight/drug effects , Diet, Fat-Restricted , Dietary Fats/administration & dosage , Hypertrophy/pathology , Lipid Metabolism/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Mitochondria/drug effects , Mitochondria/metabolism , Obesity/drug therapy , Reactive Oxygen Species/metabolism
9.
Crit Rev Biochem Mol Biol ; 47(4): 379-90, 2012.
Article in English | MEDLINE | ID: mdl-22720713

ABSTRACT

The renin-angiotensin system (RAS) is classically known for its role in regulation of blood pressure, fluid and electrolyte balance. In this system, angiotensinogen (Agt), the obligate precursor of all bioactive angiotensin peptides, undergoes two enzymatic cleavages by renin and angiotensin converting enzyme (ACE) to produce angiotensin I (Ang I) and angiotensin II (Ang II), respectively. The contemporary view of RAS has become more complex with the discovery of additional angiotensin degradation pathways such as ACE2. All components of the RAS are expressed in and have independent regulation of adipose tissue. This local adipose RAS exerts important auto/paracrine functions in modulating lipogenesis, lipolysis, adipogenesis as well as systemic and adipose tissue inflammation. Mice with adipose-specific Agt overproduction have a 30% increase in plasma Agt levels and develop hypertension and insulin resistance, while mice with adipose-specific Agt knockout have a 25% reduction in Agt plasma levels, demonstrating endocrine actions of adipose RAS. Emerging evidence also points towards a role of RAS in regulation of energy balance. Because adipose RAS is overactivated in many obesity conditions, it is considered a potential candidate linking obesity to hypertension, insulin resistance and other metabolic derangements.


Subject(s)
Adipose Tissue/metabolism , Metabolic Diseases/metabolism , Renin-Angiotensin System/physiology , Adipose Tissue/immunology , Angiotensins/genetics , Angiotensins/metabolism , Animals , Humans , Insulin Resistance/genetics , Insulin Resistance/physiology , Metabolic Diseases/genetics , Renin-Angiotensin System/genetics
10.
Nutrients ; 16(1)2023 Dec 25.
Article in English | MEDLINE | ID: mdl-38201900

ABSTRACT

The plant-derived polyphenol curcumin alleviates the inflammatory and metabolic effects of obesity, in part, by reducing adipose tissue inflammation. We hypothesized that the benefits of curcumin supplementation on diet-induced obesity and systemic inflammation in mice occur through downregulation of white adipose tissue (WAT) inflammation. The hypothesis was tested in adipose tissue from high-fat diet-induced obese mice supplemented with or without curcumin and in 3T3-L1 adipocytes treated with or without curcumin. Male B6 mice were fed a high-fat diet (HFD, 45% kcal fat) with or without 0.4% (w/w) curcumin supplementation (HFC). Metabolic changes in these mice have been previously reported. Here, we determined the serum levels of the curcumin metabolites tetrahydrocurcumin (THC) and curcumin-O-glucuronide (COG) using mass spectrometry. Moreover, we determined interleukin 6 (IL-6) levels and proteomic changes in LPS-stimulated 3T3-L1 adipocytes treated with or without curcumin by using immunoassays and mass spectrometry, respectively, to gain further insight into any altered processes. We detected both curcumin metabolites, THC and COG, in serum samples from the curcumin-fed mice. Both curcumin and its metabolites reduced LPS-induced adipocyte IL-6 secretion and mRNA levels. Proteomic analyses indicated that curcumin upregulated EIF2 and mTOR signaling pathways. Overall, curcumin exerted anti-inflammatory effects in adipocytes, in part by reducing IL-6, and these effects may be linked to the upregulation of the mTOR signaling pathway, warranting additional mechanistic studies on the effects of curcumin and its metabolites on metabolic health.


Subject(s)
Curcumin , Glucuronides , Animals , Mice , Curcumin/pharmacology , Interleukin-6/genetics , Lipopolysaccharides , Proteomics , Adipocytes , Adipose Tissue, White , Anti-Inflammatory Agents/pharmacology , Inflammation/drug therapy , TOR Serine-Threonine Kinases , Obesity/drug therapy
11.
J Nutr Biochem ; 115: 109242, 2023 05.
Article in English | MEDLINE | ID: mdl-36442715

ABSTRACT

Obesity is associated with an imbalance of micro-and macro-nutrients, gut dysbiosis, and a "leaky" gut phenomenon. Polyphenols, such as curcumin, resveratrol, and anthocyanins may alleviate the systemic effects of obesity, potentially by improving gut microbiota, intestinal barrier integrity (IBI), and zinc homeostasis. The essential micronutrient zinc plays a crucial role in the regulation of enzymatic processes, including inflammation, maintenance of the microbial ecology, and intestinal barrier integrity. In this review, we focus on IBI- which prevents intestinal lipopolysaccharide (LPS) leakage - as a critical player in polyphenol-mediated protective effects against obesity-associated white adipose tissue (WAT) inflammation. This occurs through mechanisms that block the movement of the bacterial endotoxin LPS across the gut barrier. Available research suggests that polyphenols reduce WAT and systemic inflammation via crosstalk with inflammatory NF-κB, the mammalian target of rapamycin (mTOR) signaling and zinc homeostasis.


Subject(s)
Gastrointestinal Microbiome , Humans , Polyphenols/pharmacology , Lipopolysaccharides/pharmacology , Anthocyanins/pharmacology , Obesity/microbiology , Inflammation/drug therapy , Anti-Inflammatory Agents/pharmacology , Homeostasis , Zinc/pharmacology , Dysbiosis/microbiology
12.
Biochim Biophys Acta Mol Basis Dis ; 1868(10): 166477, 2022 10 01.
Article in English | MEDLINE | ID: mdl-35780942

ABSTRACT

Impaired metabolic functions underlie the pathophysiology of diabetes and obesity. The renin-angiotensin system (RAS) is one pathway related to the pathophysiology of both diseases. RAS activation in metabolically active tissues exerts pro-inflammatory effects via angiotensin II (Ang II), linked to dysfunction in cellular processes such as autophagy, which is associated with obesity and diabetes. Here, we determined whether RAS is involved in metabolic dysregulations in a Type 1 Diabetes (T1D) mouse model, treated with captopril, and in an obesity mouse model (Agt-Tg) that overexpresses angiotensinogen (Agt) in adipose tissue. T1D mice had lower plasma leptin, resistin and higher non-esterified fatty acids (NEFA) compared to wild type (Wt) mice, even under captopril treatment. Further, mRNA levels for Agt, At1, Insr, and Beclin1 were upregulated in muscle and liver of T1D mice with captopril compared to Wt. Moreover, autophagy markers LC3 and p62 proteins were decreased, regardless of captopril treatment in the liver from T1D mice. In obese Wt mice, captopril increased muscle Irs1 gene levels. Further, captopril reduced mRNA levels of At1, Insr, Ampk, Beclin1, Atg12, and Lc3 in the liver from both Wt and Agt-Tg mice, while Agt, At1, Insr, and Atg12 expression was reduced in Agt-Tg mice without captopril treatment. Irs1 expression was decreased in the liver from obese Wt mice treated with captopril. Our results suggest that captopril treatment upregulates components of RAS, insulin signaling, and autophagy in both muscle and liver, indicating potential utility of captopril in targeting both insulin sensitivity and autophagy in diabetes and obesity.


Subject(s)
Captopril , Diabetes Mellitus, Type 1 , Animals , Autophagy , Beclin-1/metabolism , Captopril/pharmacology , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/metabolism , Diet , Glucose/metabolism , Liver/metabolism , Mice , Mice, Obese , Muscles/metabolism , Obesity/drug therapy , Obesity/metabolism , RNA, Messenger/metabolism
13.
J Nutr ; 141(3): 512-4, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21270365

ABSTRACT

Systems genetics is a novel approach for identifying the complex genetic architecture of quantitative traits and gene-environment interactions via detection of connections from genetic variation through intermediate phenotypes to overlying systems level phenotypes. This symposium, conducted at the Experimental Biology 2010 conference, aimed at educating nutrition researchers about the use of systems genetics as a tool for linking genetic variation to nutrient metabolism and energy balance and their overlying effects on health and disease. Basic concepts of systems genetics and the analytical framework used in these studies were presented. Further, the utility of genetic reference populations for gene-environment interaction studies along with specific studies addressing genetic variation in responsiveness to nutrients were discussed.


Subject(s)
Biomedical Research/methods , Genetic Phenomena , Nutritional Sciences/trends , Obesity/prevention & control , Systems Biology/methods , Animals , Energy Metabolism , Genetic Variation , Humans , Nutrigenomics/methods
14.
Biomedicines ; 9(11)2021 Oct 27.
Article in English | MEDLINE | ID: mdl-34829779

ABSTRACT

Visceral obesity may be a driving factor in nonalcoholic fatty liver disease (NAFLD) development. Previous studies have shown that the omega-3 polyunsaturated fatty acid, eicosapentaenoic acid (EPA), ameliorates obesity in high-fat (HF) fed male, C57Bl/6 mice at thermoneutral conditions, independent of uncoupling protein 1 (UCP1). Our goals herein were to investigate sex-dependent mechanisms of EPA in the livers of wild type (WT) and UCP1 knockout (KO) male and female mice fed a HF diet (45% kcal fat; WT-HF, KO-HF) with or without supplementation of 36 g/kg EPA (WT-EPA, KO-EPA). KO significantly increased body weight in males, with no significant reductions with EPA in the WT or KO groups. In females, there were no significant differences in body weight among KO groups and no effects of EPA. In males, liver TGs were significantly higher in the KO-HF group and reduced with EPA, which was not observed in females. Accordingly, gene and protein markers of mitochondrial oxidation, peroxisomal biogenesis and oxidation, as well as metabolic futile cycles were sex-dependently impacted by KO and EPA supplementation. These findings suggest a genotypic difference in response to dietary EPA supplementation on the livers of male and female mice with diet-induced obesity and housed at thermoneutrality.

15.
J Nutr ; 140(11): 1915-22, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20861209

ABSTRACT

We investigated the effects of eicosapentaenoic acid (EPA) on prevention (P) and reversal (R) of high saturated-fat (HF) diet-induced obesity and glucose-insulin homeostasis. Male C57BL/6J mice were fed low-fat (LF; 10% energy from fat), HF (45% energy from fat), or a HF-EPA-P (45% energy from fat; 36 g/kg EPA) diet for 11 wk. A 4th group was initially fed HF for 6 wk followed by the HF-EPA-R diet for 5 wk. As expected, mice fed the HF diet developed obesity and glucose intolerance. In contrast, mice fed the HF-EPA-P diet maintained normal glucose tolerance despite weight gain compared with the LF group. Whereas the HF group developed hyperglycemia and hyperinsulinemia, both HF-EPA groups (P and R) exhibited normal glycemia and insulinemia. Further, plasma adiponectin concentration was lower in the HF group but was comparable in the LF and HF-EPA groups, suggesting a role of EPA in preventing and improving insulin resistance induced by HF feeding. Further analysis of adipose tissue adipokine levels and proteomic studies in cultured adipocytes indicated that dietary EPA supplementation of HF diets was associated with reduced adipose inflammation and lipogenesis and elevated markers of fatty acid oxidation. In C57BL/6J mice, EPA minimized saturated fat-induced insulin resistance and this is in part mediated by its effects on fatty acid oxidation and inflammation.


Subject(s)
Adipose Tissue/metabolism , Dietary Fats/adverse effects , Eicosapentaenoic Acid/administration & dosage , Glucose Intolerance/prevention & control , Inflammation Mediators/metabolism , Insulin Resistance , Obesity/prevention & control , 3T3-L1 Cells , Adipokines/metabolism , Adiponectin/blood , Animals , Biomarkers/blood , Biomarkers/metabolism , Body Weight , Fatty Liver/pathology , Fatty Liver/prevention & control , Gene Expression Regulation , Glucose Intolerance/blood , Hyperglycemia/blood , Hyperglycemia/prevention & control , Hyperinsulinism/blood , Hyperinsulinism/prevention & control , Inflammation Mediators/blood , Lipid Metabolism , Lipogenesis , Male , Mice , Mice, Inbred C57BL , Obesity/blood , Obesity/metabolism , Obesity/pathology
16.
Adipocyte ; 9(1): 360-373, 2020 12.
Article in English | MEDLINE | ID: mdl-32654628

ABSTRACT

Obesity is a complex disease of global epidemic proportions. Adipose tissue expansion and chronic low-grade inflammation, locally and systemically, are hallmark features of obesity. Obesity is associated with several other chronic diseases, which are also characterized by inflammation. Determination of adipocyte size and macrophage content in adipose tissue is a critical step in assessing changes in this tissue with obesity. Here, we introduce a complete standalone software package, AdipoGauge, to analyse microscopic images derived from haematoxylin and eosin (H&E)-stained and immunofluorescently stained histology sections of adipose tissue. The software package is a user-friendly application that does not require a vast knowledge of computer science or costly commercial tools. AdipoGauge includes analysing tools that are capable of cell counting and colour separation. Furthermore, it can quantify the cell data in images both with and without clear boundaries around the cells. It can also remove objects from the image that are not intended for analysis, such as blood vessels or partial cells at edges of slide sections. The simple and state-of-the-art graphical user interface requires minimal time and learning.


Subject(s)
Fluorescent Antibody Technique/methods , Image Processing, Computer-Assisted/methods , Immunohistochemistry/methods , Microscopy , Software , Adipocytes/pathology , Adipose Tissue/pathology , Animals , Humans , Macrophages/pathology , Mice , Microscopy/methods
17.
J Clin Aesthet Dermatol ; 13(7): 18-23, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32983332

ABSTRACT

BACKGROUND: The effectiveness of different combined oral contraceptive pills and metformin in reducing hirsutism in patients with polycystic ovary syndrome (PCOS) remains unclear. OBJECTIVE: We sought to determine the effects of ethinylestradiol (35µg)/cyproterone acetate (2mg) (EE/CPA) and ethinylestradiol (20µg)/desogestrel (0.15mg) (EE/DES), alone or with metformin, on hirsutism in PCOS. METHODS: A randomized, double-blind, triple-dummy study was conducted on women with PCOS and hirsutism (N=107) who received one of four drug combinations (Arm A: EE/CPA; Arm B: EE/DES; Arm C: EE/CPA plus metformin; or Arm D: EE/DES plus metformin). Hirsutism was assessed at baseline, six months, and 12 months by using five outcomes variables. RESULTS: No outcomes variable showed a significant difference between the four arms at 12 months. There was a significant reduction in both hair density and modified Ferriman-Gallwey score (mFGS) in Arm A, mFGS in Arm B, hair density in Arm C, and diameter of sideburn hair in Arm D, respectively. Separately, there was a significant increase noted in the hair growth rate of chin and an improvement in patients' perceptions of hirsutism in all four study arms. CONCLUSION: EE/CPA and EE/DES were equally effective in improving hirsutism in PCOS, with no added benefit from low-dose metformin. Sri Lanka Clinical Trials Registry (http://www.slctr.lk) registration no. SLCTR/2015/007.

18.
Cells ; 9(6)2020 05 26.
Article in English | MEDLINE | ID: mdl-32466437

ABSTRACT

Overactivation of the renin-angiotensin system (RAS) during obesity disrupts adipocyte metabolic homeostasis and induces endoplasmic reticulum (ER) stress and inflammation; however, underlying mechanisms are not well known. We propose that overexpression of angiotensinogen (Agt), the precursor protein of RAS in adipose tissue or treatment of adipocytes with Angiotensin II (Ang II), RAS bioactive hormone, alters specific microRNAs (miRNA), that target ER stress and inflammation leading to adipocyte dysfunction. Epididymal white adipose tissue (WAT) from B6 wild type (Wt) and transgenic male mice overexpressing Agt (Agt-Tg) in adipose tissue and adipocytes treated with Ang II were used. Small RNA sequencing and microarray in WAT identified differentially expressed miRNAs and genes, out of which miR-690 and mitogen-activated protein kinase kinase 3 (MAP2K3) were validated as significantly up- and down-regulated, respectively, in Agt-Tg, and in Ang II-treated adipocytes compared to respective controls. Additionally, the direct regulatory role of miR-690 on MAP2K3 was confirmed using mimic, inhibitors and dual-luciferase reporter assay. Downstream protein targets of MAP2K3 which include p38, NF-κB, IL-6 and CHOP were all reduced. These results indicate a critical post-transcriptional role for miR-690 in inflammation and ER stress. In conclusion, miR-690 plays a protective function and could be a useful target to reduce obesity.


Subject(s)
Angiotensin II/pharmacology , Endoplasmic Reticulum Stress , Inflammation/genetics , MicroRNAs/metabolism , 3T3-L1 Cells , Adipocytes/drug effects , Adipocytes/metabolism , Adipose Tissue, White/metabolism , Animals , Base Sequence , Biomarkers/metabolism , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/genetics , Gene Expression Regulation/drug effects , Inflammation/pathology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , Male , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Renin-Angiotensin System/drug effects , Renin-Angiotensin System/genetics , Reproducibility of Results , Signal Transduction/drug effects
19.
Cell Death Dis ; 11(2): 87, 2020 02 03.
Article in English | MEDLINE | ID: mdl-32015340

ABSTRACT

Metabolic syndrome (MetS) is a complex, emerging epidemic which disrupts the metabolic homeostasis of several organs, including liver, heart, pancreas, and adipose tissue. While studies have been conducted in these research areas, the pathogenesis and mechanisms of MetS remain debatable. Lines of evidence show that physiological systems, such as the renin-angiotensin system (RAS) and autophagy play vital regulatory roles in MetS. RAS is a pivotal system known for controlling blood pressure and fluid balance, whereas autophagy is involved in the degradation and recycling of cellular components, including proteins. Although RAS is activated in MetS, the interrelationship between RAS and autophagy varies in glucose homeostatic organs and their cross talk is poorly understood. Interestingly, autophagy is attenuated in the liver during MetS, whereas autophagic activity is induced in adipose tissue during MetS, indicating tissue-specific discordant roles. We discuss in vivo and in vitro studies conducted in metabolic tissues and dissect their tissue-specific effects. Moreover, our review will focus on the molecular mechanisms by which autophagy orchestrates MetS and the ways future treatments could target RAS in order to achieve metabolic homeostasis.


Subject(s)
Autophagy/physiology , Metabolic Syndrome/pathology , Renin-Angiotensin System/physiology , Adipose Tissue/metabolism , Adipose Tissue/pathology , Energy Metabolism , Heart Diseases/metabolism , Heart Diseases/pathology , Humans , Inflammation , Insulin Resistance , Liver/metabolism , Liver/pathology , Metabolic Syndrome/metabolism , Obesity/metabolism , Obesity/pathology
20.
Contemp Clin Trials Commun ; 16: 100453, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31650073

ABSTRACT

Mobile phone-based health interventions (mHealth) are viewed as an attractive approach to foster behaviour change, and found to be effective in promoting physical activity and healthy diets. The present study aims to investigate whether mHealth with advice for dietary and lifestyle modifications would reduce 10-year cardio vascular disease (CVD) risk among overweight or obese adults aged 35-64 years in Sri Lanka. A two-group parallel-arm randomized controlled trial (RCT) was conducted in Colombo district, recruiting 1200 individuals aged 35-64 years with a body mass index (BMI) of ≥25 kgm-2. Participants were randomly assigned either to mHealth package (intervention arm, n = 600) or usual care (control arm, n = 600). The intervention package contains a series of dietary and lifestyle improvement messages, a mobile application to register participants, and a web application to deliver these messages. Participants in the intervention arm receive 2 voice and 2 text messages per week to their mobile phones for a period of 12 months. The primary outcome (10-year CVD risk) will be assessed according to sex, age, smoking status, blood pressure, serum cholesterol and glycaemic status. Data are collected at enrollment and after 12 months of intervention on: dietary practices, physical activity, smoking, anthropometry, body composition, blood pressure, fasting plasma glucose, HbA1c and lipid profile. Analysis of effect will be performed by intention-to-treat principle, comparing the outcomes between intervention and control arms. The study resulted in a comprehensive mHealth nutrition and lifestyle package (mHENAL) and successfully completed recruitment and baseline assessment of participants. The message delivery is in progress.

SELECTION OF CITATIONS
SEARCH DETAIL