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










Publication year range
1.
Mol Biol Rep ; 50(2): 1033-1044, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36383337

ABSTRACT

BACKGROUND: Inorganic arsenic [As(III)] and hexavalent chromium [Cr(VI)] can potentially affect metabolic functions. These heavy metal(s)/metalloids can also affect the gut microbial architecture which affects metabolic health. Here, we assessed the effects of short-term exposure of As(III) and Cr(VI) on key transcription factors in adipose tissues and on selected gut microbial abundances to understand the possible modulatory role of these toxicants on host metabolic health. METHODS AND RESULTS: qRT-PCR based relative bacterial abundance studies in cecal samples, gene expression analysis for gut wall integrity in ileum and colon and adipogenesis, lipolysis, and thermogenic genes in gonadal white and brown adipose tissue (gWAT and BAT), along with tissue oxidative stress parameters have been performed. As(III) and Cr(VI) exposure reduced beneficial Lactobacilli, Bifidobacteria, Akkermansia, Lachenospiraceae, Fecalibacterium, Eubacterium, and clostridium coccoid group while increasing lipopolysaccharides producing Enterobacteriaceae abundances. It also impaired structural features and expression of key tight junction and mucin production genes in ileum and colon (Cld-2, Cld-4, ZO-1, ZO-2, MUC-2 and - 4). In gWAT it inhibited adipogenesis (PPARγ, FASN, SREBP1a), lipolysis (HSL, ACOX-1), and thermogenesis (UCP-1, PGC1a, PRDM-16, PPARa) related genes expression, whereas in BAT, it enhanced adipogenesis and reduced thermogenesis. These exposures also reduces the endogenous antioxidants levels in these tissues and promote pro-inflammatory cytokines genes expression (TLRs, IL-6, MCP-1). The combinatorial exposure appears to have more deleterious effects. CONCLUSION: These effects of As(III) and Cr(VI) may not directly be linked to their known toxicological effects, instead, more intriguing crosstalk with gut microbial ecosystem hold the key.


Subject(s)
Arsenic , Mice , Animals , Arsenic/metabolism , Ecosystem , Dysbiosis/metabolism , Chromium/toxicity , Chromium/metabolism , Adipose Tissue, White/metabolism , Thermogenesis
2.
Food Funct ; 12(22): 11526-11536, 2021 Nov 15.
Article in English | MEDLINE | ID: mdl-34705006

ABSTRACT

Scope. Given the global epidemic of diabesity (co-existence of both diabetes and obesity), novel approaches that target gut hormone secretion and their modulation may offer the dual benefits of increased efficacy and limited side effects. In the present study, we tested the hypothesis that agonism of Transient Receptor Potential Ankyrin 1 (TRPA1), using a dietary activator, has a modulatory role in high fat diet (HFD)-induced dysregulation of post-prandial gut hormone responses and prevention of metabolic alterations. Methods and results. The effect of HFD on TRPA1 expression in different parts of the gut using immunohistochemistry, western blotting and RT-PCR was studied. Dietary TRPA1 agonist, Allicin Rich Garlic Juice (ARGJ), was co-administered along with HFD in mice for three months and various metabolic health parameters, relative gut hormone levels and inflammation were observed. The HFD caused substantial reduction in gut TRPA1 expression along with dysregulation in post-prandial normalization of gut hormone levels, particularly GLP-1, precipitating hunger phenotype, altered glucose homeostasis, hepatic inflammation and fat accumulation. TRPA1 agonism through ARGJ co-supplementation prevented HFD-induced dysregulation in post-prandial normalization of gut hormone levels and averted metabolic and inflammatory complications in peripheral tissues. Conclusion. Our findings provide evidence that ARGJ (diet-based TRPA1 agonism) can be employed as a feasible strategy, as nutraceuticals or food, to prevent HFD-induced metabolic complications.


Subject(s)
Diet, High-Fat/adverse effects , Disulfides/pharmacology , Gastrointestinal Microbiome/drug effects , Inflammation , Sulfinic Acids/pharmacology , TRPA1 Cation Channel/agonists , Animals , Inflammation/metabolism , Inflammation/physiopathology , Male , Mice
3.
Child Care Health Dev ; 46(5): 571-575, 2020 09.
Article in English | MEDLINE | ID: mdl-32585729

ABSTRACT

BACKGROUND: An increasing prevalence of childhood obesity is reported worldwide. Few data are available regarding childhood obesity in North India. The present study aimed to study the prevalence of overweight/obesity among adolescents aged 10-14, in schools of Chandigarh, and to examine associated factors. METHODS: Nine co-educational schools were chosen to include both government and private schools in Chandigarh. We randomly sampled students from different subsections/batches of classes fifth to ninth (aged 10-14), and those present on the day were measured and completed questionnaires. Obesity was classified according to the methods recommended by the Indian Association of Pediatrics (IAP) growth charts committee. RESULTS: A total of 1,030 participants were included, 502 students from government and 528 students from private schools. The overall prevalence of overweight and obesity evaluated by using age-specific body mass index (BMI) cut-offs was found to be 9.9% and 14.0%, respectively. The prevalence of overweight (adult equivalent of 23) was 10.3% in boys and 9.4% in girls and that of obesity (adult equivalent of 27) was found to be 13.3% and 14.7%, respectively, in boys and girls. In univariate analyses, statistically significant associations were found between the risk of obesity and gender, socio-economic status (SES) and reported physical activity. CONCLUSION: We found significant levels of overweight and obesity among children aged 10-14 and found associations with SES, gender and reported physical activity as has been previously reported elsewhere.


Subject(s)
Pediatric Obesity/epidemiology , Adolescent , Body Mass Index , Child , Exercise , Female , Humans , India , Male , Pediatric Obesity/diagnosis , Pediatric Obesity/psychology , Prevalence , Risk Factors , Sedentary Behavior , Sex Factors , Social Class , Surveys and Questionnaires
4.
Front Pharmacol ; 9: 1244, 2018.
Article in English | MEDLINE | ID: mdl-30505271

ABSTRACT

Glucagon mediated mechanisms have been shown to play clinically significant role in energy expenditure. The present study was designed to understand whether pharmacological mimicking of cold using menthol (TRPM8 modulator) can induce glucagon-mediated energy expenditure to prevent weight gain and related complications. Acute oral and topical administration of TRPM8 agonists (menthol and icilin) increased serum glucagon concentration which was prevented by pre-treatment with AMTB, a TRPM8 blocker. Chronic administration of menthol (50 and 100 mg/kg/day for 12 weeks) to HFD fed animals prevented weight gain, insulin resistance, adipose tissue hypertrophy and triacylglycerol deposition in liver. These effects were not restricted to oral administration, but also observed upon the topical application of menthol (10% w/v). The metabolic alterations caused by menthol in liver and adipose tissue mirrored the known effects of glucagon, such as increased glycogenolysis and gluconeogenesis in the liver, and enhanced thermogenic activity of white and brown adipose tissue. Correlation analysis suggests a strong correlation between glucagon dependent changes and energy expenditure markers. Interestingly, in-vitro treatment of the serum of menthol treated mice increased energy expenditure markers in mature 3T3L1 adipocytes, which was prevented in the presence of non-competitive glucagon receptor antagonist, L-168,049, indicating that menthol-induced increase in serum glucagon is responsible for increase in energy expenditure phenotype. In conclusion, the present work provides evidence that glucagon plays an important role in the preventive effect of menthol against HFD-induced weight gain and related complications.

5.
Food Funct ; 9(2): 1254-1264, 2018 Feb 21.
Article in English | MEDLINE | ID: mdl-29393319

ABSTRACT

Probiotic lactic acid bacteria are known to modulate gut associated immune responses. Not many studies have reported on the role of Weissella species in preventing lipopolysaccharide (LPS) induced proinflammatory stress in murine macrophages as well as in human intestinal epithelial cells (Caco-2). Therefore, the present study was taken up to evaluate the probiotic attributes of four newly isolated Weissella strains (two each from fermented dosa batter and a human infant faecal sample); these attributes are cholesterol reduction, adhesion to Caco-2 cells and mucin and their ability to prevent LPS-induced nitric oxide and proinflammatory cytokine (IL-6, IL-1ß and TNFα) production by the murine macrophages and IL-8 production by the human epithelial cells. Reduction in LPS induced pro-inflammatory stress was compared with a well-studied probiotic bacterium Lactobacillus rhamnosus GG. The results suggested that the strains were tolerant to gastric conditions (pH 3.0) and bile salts. In addition, the strains exhibited moderate cell surface hydrophobicity, cholesterol reduction and adhesion to Caco-2 cells and gastric mucin. All the strains could prevent LPS-induced nitric oxide and IL-6 production in murine macrophages, while strain 28 alone prevented IL-1ß production. All the strains could prevent IL-8 production by the human epithelial cells. The present study led to the first line selection of W. cibaria 28 as a putative strain for future studies as it showed adhesion to Caco-2 cells and gastric mucin and cholesterol reduction besides preventing LPS-induced pro-inflammatory stress in macrophages and in human colonic epithelial cells.


Subject(s)
Epithelial Cells/drug effects , Intestines/drug effects , Macrophages/drug effects , Probiotics/pharmacology , Weissella/physiology , Animals , Bacterial Adhesion , Caco-2 Cells , Epithelial Cells/immunology , Epithelial Cells/microbiology , Humans , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Intestines/immunology , Intestines/microbiology , Lipopolysaccharides/adverse effects , Macrophages/immunology , Mice , Probiotics/isolation & purification , Weissella/genetics , Weissella/isolation & purification
6.
Int J Biol Macromol ; 106: 994-1003, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28837853

ABSTRACT

Arabinoxylan (AX), a non-starch polysaccharide extracted from cereals such as wheat, rice and millets, is known to impart various health promoting effects. Our earlier study suggested that finger millet (FM) could ameliorate high fat diet (HFD)-induced metabolic derangements. The present study is aimed to evaluate the effect of FM-AX supplementation, a key bioactive from finger millet, on HFD-induced metabolic and gut bacterial derangements. Male Swiss albino mice were fed with normal chow diet (NPD) or HFD (60%kcal from fat) for 10 weeks. FM-AX was orally supplemented at doses of 0.5 and 1.0g/kg bodyweight on every alternate day for 10 weeks. Glucose tolerance, serum hormones, hepatic lipid accumulation and inflammation, white adipose tissue marker gene expression, adipocyte size and inflammation; metagenomic alterations in cecal bacteria; cecal short chain fatty acids and colonic tight junction gene expressions were studied. FM-AX supplementation prevented HFD-induced weight gain, alerted glucose tolerance and serum lipid profile, hepatic lipid accumulation and inflammation. Hepatic and white adipose tissue gene expressions were beneficially modulated. Further, AX supplementation prevented metagenomic alterations in cecum; improved ileal and colonic health and overall prevented metabolic endotoxemia. Present work suggests that AX from finger millet can be developed as a nutraceutical for the management of HFD- induced obesity.


Subject(s)
Dysbiosis/diet therapy , Eleusine , Endotoxemia/diet therapy , Inflammation/diet therapy , Xylans/administration & dosage , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Animals , Body Weight , Diet, High-Fat/adverse effects , Dysbiosis/microbiology , Dysbiosis/pathology , Endotoxemia/metabolism , Endotoxemia/pathology , Gastrointestinal Microbiome/drug effects , Gene Expression Regulation/drug effects , Glucose Tolerance Test , Humans , Inflammation/metabolism , Inflammation/pathology , Lipid Metabolism/drug effects , Lipid Metabolism/genetics , Liver/drug effects , Liver/metabolism , Mice , Xylans/chemistry
7.
Eur J Nutr ; 57(8): 2897-2911, 2018 Dec.
Article in English | MEDLINE | ID: mdl-29127476

ABSTRACT

PURPOSE: Cranberries are a rich source of polyphenolic antioxidants. Purified sugars or artificial sweeteners are being added to cranberry-based food products to mask tartness. Refined sugar and artificial sweeteners intake modulate gut microbiota and result in metabolic complications. We evaluated effects of isomalto-oligosaccharides (IMOs; sweet tasting non-digestible oligosaccharides) with cranberry extract (CRX) on high fat diet (HFD)-induced metabolic alterations in mice. METHODS: Male Swiss albino mice were fed normal chow or HFD (58% fat kcal), and were administered either CRX (200 mg/kg) alone or in combination with IMOs (1 g/kg). Cecal short-chain fatty acids, abundances of selected (1) butyrate producing, (2) metabolically beneficial, and (3) selective lipopolysaccharides producing gram negative gut bacteria were studied. Further, gut-related histological, biochemical, genomic changes along with circulating pro-/anti-inflammatory markers and systemic obesity-associated metabolic changes were studied. RESULTS: Co-supplementation of CRX and IMOs significantly improved cecal SCFAs, especially butyrate levels, selected butyrate-producing bacteria (clostridial cluster XIVa bacteria) and butyrate kinase expression in HFD-fed mice. The combination also significantly improved gut beneficial bacterial abundance, gut histology and related changes (colon mucin production, gut permeability) as compared to individual agents. It also prevented HFD-induced systemic and tissue inflammation, glucose intolerance and systemic obesity-associated metabolic changes in adipose tissue and liver. The combination of CRX and IMOs appeared more effective in the prevention of HFD-induced gut derangements. CONCLUSION: Combination of CRX and IMOs could be advantageous for normalization of metabolic alterations seen in diet-induced obesity via beneficial modulation of gastrointestinal health.


Subject(s)
Butyrates/metabolism , Metabolic Syndrome/drug therapy , Oligosaccharides/pharmacology , Plant Extracts/pharmacology , Vaccinium macrocarpon/chemistry , Animals , Cecum/drug effects , Cecum/metabolism , Colon/drug effects , Colon/metabolism , Cytokines/blood , Diet, High-Fat/adverse effects , Dietary Supplements , Fatty Acids/metabolism , Fruit/chemistry , Gastrointestinal Microbiome/drug effects , Glucose Intolerance/metabolism , Inflammation/drug therapy , Lipopolysaccharides/metabolism , Male , Metabolic Syndrome/blood , Metabolic Syndrome/etiology , Mice , Obesity/drug therapy , Polyphenols/pharmacology
8.
Pharmacol Res ; 123: 103-113, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28668709

ABSTRACT

High fat diet (HFD)-induced alterations in gut microbiota and resultant 'leaky gut' phenomenon promotes metabolic endotoxemia, ectopic fat deposition, and low-grade systemic inflammation. Here we evaluated the effects of a combination of green tea extract (GTE) with isomalto-oligosaccharide (IMOs) on HFD-induced alterations in mice. Male Swiss albino mice were fed with HFD (58% fat kcal) for 12 weeks. Systemic adiposity, gut derangement parameters and V3-V4 region based 16S rRNA metagenomic sequencing, ectopic fat deposition, liver metabolome analysis, systemic and tissue inflammation, and energy homeostasis markers along with gene expression analysis in multiple tissues were done in mice supplemented with GTE, IMOs or their combination. The combination of GTE and IMOs effectively prevented HFD-induced adiposity and lipid accumulation in liver and muscle while normalizing fasting blood glucose, insulin, glucagon, and leptin levels. Co-administration of GTE with IMOs effectively modulated liver metabolome associated with lipid metabolism. It also prevented leaky gut phenotype and HFD-induced increase in circulating lipopolysaccharides and pro-inflammatory cytokines (e.g. resistin, TNF-α, and IL-1ß) and reduction in anti-inflammatory cytokines (e.g. adiponectin and IL-6). Gene expression analysis across multiple tissues further supported these functional outcomes. Most importantly, this combination improved beneficial gut microbiota (Lactobacillus sp., Bifidobacteria, Akkermansia muciniphila, Roseburia spp.) abundances, restored Firmicutes/Bacteriodetes and improved Prevotella/Bacteroides proportions. In particular, a combination of these two agents has shown improved beneficial effects on multiple parameters studied. Data presented herein suggests that strategically chosen food components might be highly effective in the prevention of HFD-induced alterations and may further be developed as functional foods.


Subject(s)
Camellia sinensis , Diet, High-Fat , Dysbiosis/prevention & control , Gastrointestinal Microbiome/drug effects , Oligosaccharides/pharmacology , Plant Extracts/pharmacology , Prebiotics , Adiposity/drug effects , Animals , Cytokines/blood , Liver/drug effects , Liver/metabolism , Male , Mice
9.
Food Funct ; 8(3): 1174-1183, 2017 Mar 22.
Article in English | MEDLINE | ID: mdl-28180212

ABSTRACT

The protective role of kodo millet whole grain and bran supplementation in diet induced obesity has not been investigated. Here we have studied the role of kodo millet supplementation in age matched Swiss albino mice that were randomly divided into groups and fed their respective diets for 16 weeks. A high fat diet increased weight gain, reduced glucose tolerance, increased serum lipids, altered hepatic and adipocyte gene expression and caused dysbiosis in the intestinal beneficial bacteria. Kodo millet supplementation did not affect weight gain but it improved glucose tolerance and prevented an increase in the serum cholesterol and lipid parameters (P ≤ 0.05), modulated adipogenesis related gene expression, decreased serum IL-6 and LPS levels (P ≤ 0.05), promoted selected beneficial gut bacterial abundances (Lactobacillus sp., Bifidobacteria, Akkermansia and Roseburia spp.) and improved the total short chain fatty acid production (P ≤ 0.05) and acetate levels (P ≤ 0.05) in cecal contents. This study provides evidence that kodo millet supplementation alleviates high-fat diet induced changes and hence can be incorporated as a functional ingredient for the management of obesity.


Subject(s)
Dietary Fiber/metabolism , Dietary Supplements/analysis , Gastrointestinal Microbiome , Obesity/diet therapy , Obesity/metabolism , Obesity/microbiology , Paspalum/metabolism , Adipogenesis , Animals , Bacteria/metabolism , Diet, High-Fat/adverse effects , Female , Humans , Interleukin-6/immunology , Lipid Metabolism , Male , Mice , Mice, Inbred C57BL , Obesity/immunology , Whole Grains/metabolism
10.
Biofactors ; 42(2): 201-11, 2016.
Article in English | MEDLINE | ID: mdl-26893251

ABSTRACT

Cinnamaldehyde, a bioactive component of cinnamon, is increasingly gaining interest for its preventive and therapeutic effects against metabolic complications like type-2 diabetes. This study is an attempt to understand the effect of cinnamaldehyde in high-fat diet (HFD)-associated increase in fasting-induced hyperphagia and related hormone levels, adipose tissue lipolysis and inflammation, and selected cecal microbial count in mice. Cinnamaldehyde, at 40 µM dose, prevented lipid accumulation and altered gene expression toward lipolytic phenotype in 3T3-L1 preadipocyte cell lines. In vivo, cinnamaldehyde coadministration prevented HFD-induced body weight gain, decreased fasting-induced hyperphagia, as well as circulating leptin and leptin/ghrelin ratio. In addition to that, cinnamaldehyde altered serum biochemical parameters related to lipolysis, that is, glycerol and free fatty acid levels. At transcriptional level, cinnamaldehyde increased anorectic gene expression in hypothalamus and lipolytic gene expression in visceral white adipose tissue. Furthermore, cinnamaldehyde also decreased serum IL-1ß and inflammatory gene expression in visceral white adipose tissue. However, cinnamaldehyde did not modulate the population of selected gut microbial (Lactobacillus, Bifidibaceria, and Roseburia) count in cecal content. In conclusion, cinnamaldehyde increased adipose tissue lipolysis, decreased fasting-induced hyperphagia, normalized circulating levels of leptin/ghrelin ratio, and reduced inflammation in HFD-fed mice, which augurs well for its antiobesity role.


Subject(s)
Acrolein/analogs & derivatives , Dietary Supplements , Hyperphagia/drug therapy , Inflammation/drug therapy , 3T3-L1 Cells , Acrolein/administration & dosage , Adipose Tissue/drug effects , Adipose Tissue/pathology , Animals , Diet, High-Fat , Fasting/adverse effects , Gene Expression Regulation/drug effects , Humans , Hyperphagia/metabolism , Hyperphagia/pathology , Inflammation/blood , Inflammation/genetics , Inflammation/pathology , Interleukin-1beta/blood , Lipid Metabolism/drug effects , Lipolysis/drug effects , Mice , Weight Gain/drug effects
12.
Int J Endocrinol ; 2015: 282375, 2015.
Article in English | MEDLINE | ID: mdl-26089880

ABSTRACT

Fibroblast growth factor 21 (FGF21) modulates a diverse range of biological functions, including glucose and lipid metabolism, adaptive starvation response, and energy homeostasis, but with limited mechanistic insight. FGF21 treatment has been shown to inhibit hepatic growth hormone (GH) intracellular signaling. To evaluate GH axis involvement in FGF21 actions, transgenic mice overexpressing bovine GH were used. Expectedly, in response to FGF21 treatment control littermates showed metabolic improvements whereas GH transgenic mice resisted most of the beneficial effects of FGF21, except an attenuation of the innate hyperinsulinemia. Since FGF21 is believed to exert its effects mostly at the transcriptional level, we analyzed and observed significant upregulation in expression of various genes involved in carbohydrate and lipid metabolism, energy homeostasis, and antioxidant defense in FGF21-treated controls, but not in GH transgenics. The resistance of GH transgenic mice to FGF21-induced changes underlines the necessity of normal GH signaling for the beneficial effects of FGF21.

13.
Age (Dordr) ; 37(3): 9765, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25953669

ABSTRACT

Longevity and aging are influenced by common intracellular signals of the insulin/insulin-like growth factor (IGF)-1 pathway. Abnormally high levels of bioactive IGF-1 increase the development of various cancers and may contribute to metabolic diseases such as insulin resistance. Enhanced availability of IGF-1 is promoted by cleavage of IGF binding proteins (IGFBPs) by proteases, including the pregnancy-associated plasma protein-A (PAPPA). In vitro, PAPP-A is regulated by pro-inflammatory cytokines (PICs) such as interleukin (IL)-6 and tumor necrosis factor (TNF). Mice born with deficiency of the Papp-a gene (PAPP-A knockout (KO) mice) live ~30-40 % longer than their normal littermates and have decreased bioactive IGF-1 on standard diets. Our objective was to elucidate how the effects of high-fat, high-sucrose diet (HFHS) promote obesity, induce metabolic dysfunction, and alter systemic cytokine expression in PAPP-A KO and normal mice. PAPP-A KO mice fed HFHS diet for 10 weeks were more glucose tolerant and had enhanced insulin sensitivity compared to normal mice fed HFHS diet. PAPP-A KO mice fed HFHS diet had lower levels of pro-inflammatory cytokines (IL-2, IL-6, and TNF-α) compared to normal mice fed the same diet. However, anti-inflammatory cytokine levels (IL-4 and adiponectin) were higher in PAPP-A KO mice fed HFHS diet compared to normal mice fed HFHS. Circulating PAPP-A levels were elevated in normal mice fed an HFHS diet compared to normal mice fed a standard, low-fat, low-sucrose (LFLS) diet. Indirect calorimetry showed, at 10 weeks of feeding HFHS diet, significantly increased oxygen consumption (VO2) in PAPP-A KO mice fed HFHS diet compared to normal mice fed the same diet. Furthermore, respiratory quotient (RQ) was significantly lower in PAPP-A KO mice fed HFHS diet compared to normal (N) mice fed HFHS diet indicating PAPP-A KO mice fed HFHS diet are able to rely on fat as their primary source of energy more so than normal controls. We conclude that PAPP-A KO mice are resistant to the HFHS diet induction of metabolic dysfunction associated with higher levels of anti-inflammatory cytokines and a remarkably metabolic flexible phenotype and that some of the effects of HFHS diet in normal animals may be due to increased levels of PAPP-A.


Subject(s)
Diet, High-Fat , Energy Metabolism/physiology , Pregnancy-Associated Plasma Protein-A/deficiency , Sucrose/administration & dosage , Adiponectin/blood , Animals , Calorimetry, Indirect , Insulin/blood , Insulin-Like Growth Factor I/metabolism , Interleukin-2/blood , Interleukin-4/blood , Interleukin-6/blood , Longevity , Mice , Mice, Knockout , Oxygen Consumption/physiology , Phenotype , Respiratory Rate/physiology , Tumor Necrosis Factor-alpha/blood
14.
PLoS One ; 10(5): e0127701, 2015.
Article in English | MEDLINE | ID: mdl-26010905

ABSTRACT

BACKGROUND: Two types of adipose tissues, white (WAT) and brown (BAT) are found in mammals. Increasingly novel strategies are being proposed for the treatment of obesity and its associated complications by altering amount and/or activity of BAT using mouse models. METHODOLOGY/PRINCIPLE FINDINGS: The present study was designed to: (a) investigate the differential expression of genes in LACA mice subcutaneous WAT (sWAT) and BAT using mouse DNA microarray, (b) to compare mouse differential gene expression with previously published human data; to understand any inter- species differences between the two and (c) to make a comparative assessment with C57BL/6 mouse strain. In mouse microarray studies, over 7003, 1176 and 401 probe sets showed more than two-fold, five-fold and ten-fold change respectively in differential expression between murine BAT and WAT. Microarray data was validated using quantitative RT-PCR of key genes showing high expression in BAT (Fabp3, Ucp1, Slc27a1) and sWAT (Ms4a1, H2-Ob, Bank1) or showing relatively low expression in BAT (Pgk1, Cox6b1) and sWAT (Slc20a1, Cd74). Multi-omic pathway analysis was employed to understand possible links between the organisms. When murine two fold data was compared with published human BAT and sWAT data, 90 genes showed parallel differential expression in both mouse and human. Out of these 90 genes, 46 showed same pattern of differential expression whereas the pattern was opposite for the remaining 44 genes. Based on our microarray results and its comparison with human data, we were able to identify genes (targets) (a) which can be studied in mouse model systems to extrapolate results to human (b) where caution should be exercised before extrapolation of murine data to human. CONCLUSION: Our study provides evidence for inter species (mouse vs human) differences in differential gene expression between sWAT and BAT. Critical understanding of this data may help in development of novel ways to engineer one form of adipose tissue to another using murine model with focus on human.


Subject(s)
Adipose Tissue, Brown/metabolism , Gene Expression Regulation/physiology , Subcutaneous Fat/metabolism , Animals , Gene Expression Profiling , Humans , Mice , Protein Array Analysis/methods , Real-Time Polymerase Chain Reaction , Species Specificity
15.
J Mol Endocrinol ; 54(2): 171-84, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25691498

ABSTRACT

GH/STAT5 signaling is desensitized in the liver in adult transgenic mice overexpressing GH; however, these animals present greater body size. To assess whether the STAT5 pathway is active during the growth period in the liver in these animals, and how signaling modulators participate in this process, growing transgenic mice and normal siblings were evaluated. STAT5 does not respond to an acute GH-stimulus, but displays higher basal phosphorylation in the livers of growing GH-overexpressing mice. GH receptor and the positive modulators glucocorticoid receptor and HNF1 display greater abundance in transgenic animals, supporting the activity of STAT5. The negative modulators cytokine-induced suppressor and PTP1B are increased in GH-overexpressing mice. The suppressors SOCS2 and SOCS3 exhibit higher mRNA levels in transgenic mice but lower protein content, indicating that they are being actively degraded. Therefore, STAT5 signaling is increased in the liver in GH-transgenic mice during the growth period, with a balance between positive and negative effectors resulting in accelerated but controlled growth.


Subject(s)
Growth Hormone/metabolism , Liver/growth & development , Liver/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction , Animals , Gene Expression Regulation, Developmental , Insulin-Like Growth Factor I/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Phosphorylation , Receptors, Somatotropin/genetics , Receptors, Somatotropin/metabolism , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/metabolism
16.
Aging Cell ; 13(6): 981-1000, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25244225

ABSTRACT

In addition to their extended lifespans, slow-aging growth hormone receptor/binding protein gene-disrupted (knockout) (GHR-KO) mice are hypoinsulinemic and highly sensitive to the action of insulin. It has been proposed that this insulin sensitivity is important for their longevity and increased healthspan. We tested whether this insulin sensitivity of the GHR-KO mouse is necessary for its retarded aging by abrogating that sensitivity with a transgenic alteration that improves development and secretory function of pancreatic ß-cells by expressing Igf-1 under the rat insulin promoter 1 (RIP::IGF-1). The RIP::IGF-1 transgene increased circulating insulin content in GHR-KO mice, and thusly fully normalized their insulin sensitivity, without affecting the proliferation of any non-ß-cell cell types. Multiple (nonsurvivorship) longevity-associated physiological and endocrinological characteristics of these mice (namely beneficial blood glucose regulatory control, altered metabolism, and preservation of memory capabilities) were partially or completely normalized, thus supporting the causal role of insulin sensitivity for the decelerated senescence of GHR-KO mice. We conclude that a delayed onset and/or decreased pace of aging can be hormonally regulated.


Subject(s)
Aging/metabolism , Insulin Resistance/physiology , Insulin-Like Growth Factor I/metabolism , Insulin/metabolism , Longevity/physiology , Receptors, Somatotropin/metabolism , Animals , Female , Male , Mice , Mice, Knockout , Phenotype
17.
Br J Nutr ; 112(9): 1447-58, 2014 Nov 14.
Article in English | MEDLINE | ID: mdl-25234097

ABSTRACT

Several epidemiological studies have shown that the consumption of finger millet (FM) alleviates diabetes-related complications. In the present study, the effect of finger millet whole grain (FM-WG) and bran (FM-BR) supplementation was evaluated in high-fat diet-fed LACA mice for 12 weeks. Mice were divided into four groups: control group fed a normal diet (10 % fat as energy); a group fed a high-fat diet; a group fed the same high-fat diet supplemented with FM-BR; a group fed the same high-fat diet supplemented with FM-WG. The inclusion of FM-BR at 10 % (w/w) in a high-fat diet had more beneficial effects than that of FM-WG. FM-BR supplementation prevented body weight gain, improved lipid profile and anti-inflammatory status, alleviated oxidative stress, regulated the expression levels of several obesity-related genes, increased the abundance of beneficial gut bacteria (Lactobacillus, Bifidobacteria and Roseburia) and suppressed the abundance of Enterobacter in caecal contents (P≤ 0·05). In conclusion, FM-BR supplementation could be an effective strategy for preventing high-fat diet-induced changes and developing FM-BR-enriched functional foods.


Subject(s)
Diet, High-Fat/adverse effects , Eleusine , Inflammation/prevention & control , Intestines/microbiology , Obesity/metabolism , Oxidative Stress , Adipose Tissue/metabolism , Animals , Bacterial Load , Blood Glucose/metabolism , Dietary Supplements , Gene Expression Regulation , Lipids/blood , Liver/metabolism , Mice , Mitochondria, Muscle/metabolism , Muscle, Skeletal/metabolism , Nutrigenomics , Obesity/genetics , Pancreas/metabolism , Seeds , Weight Gain
18.
PLoS One ; 9(7): e103093, 2014.
Article in English | MEDLINE | ID: mdl-25072597

ABSTRACT

OBJECTIVE: Targeting the energy storing white adipose tissue (WAT) by pharmacological and dietary means in order to promote its conversion to energy expending "brite" cell type holds promise as an anti-obesity approach. Present study was designed to investigate/revisit the effect of capsaicin on adipogenic differentiation with special reference to induction of "brite" phenotype during differentiation of 3T3-L1 preadipocytes. METHODS: Multiple techniques such as Ca2+ influx assay, Oil Red-O staining, nutrigenomic analysis in preadipocytes and matured adipocytes have been employed to understand the effect of capsaicin at different doses. In addition to in-vitro experiments, in-vivo studies were carried out in high-fat diet (HFD) fed rats treated with resiniferatoxin (RTX) (a TRPV1 agonist) and in mice administered capsaicin. RESULTS: TRPV1 channels are expressed in preadipocytes but not in adipocytes. In preadipocytes, both capsaicin and RTX stimulate Ca2+ influx in dose-dependent manner. This stimulation may be prevented by capsazepine, a TRPV1 antagonist. At lower doses, capsaicin inhibits lipid accumulation and stimulates TRPV1 gene expression, while at higher doses it enhances accumulation of lipids and suppresses expression of its receptor. In doses of 0.1-100 µM, capsaicin promotes expression of major pro-adipogenic factor PPARγ and some of its downstream targets. In concentrations of 1 µM, capsaicin up-regulates anti-adipogenic genes. Low-dose capsaicin treatment of 3T3-L1 preadipocytes differentiating into adipocytes results in increased expression of brown fat cell marker genes. In white adipose of mice, capsaicin administration leads to increase in browning-specific genes. Global TRPV1 ablation (i.p. by RTX administration) leads to increase in locomotor activity with no change in body weight. CONCLUSION: Our findings suggest the dual modulatory role of capsaicin in adipogenesis. Capsaicin inhibits adipogenesis in 3T3-L1 via TRPV1 activation and induces brown-like phenotype whereas higher doses.


Subject(s)
Adipocytes/cytology , Adipogenesis/drug effects , Capsaicin/pharmacology , Cell Differentiation/drug effects , Phenotype , 3T3-L1 Cells , Adipocytes/metabolism , Adipocytes, Brown/cytology , Adipocytes, Brown/drug effects , Adipocytes, Brown/metabolism , Adipogenesis/genetics , Adipokines/metabolism , Animals , Biomarkers/metabolism , Body Weight/drug effects , Capsaicin/analogs & derivatives , Cell Differentiation/genetics , Diet, High-Fat , Gene Expression Regulation/drug effects , Lipid Metabolism , Mice , Motor Activity/drug effects , PPAR gamma/metabolism , Pain Threshold/drug effects , TRPV Cation Channels/metabolism
19.
J Nutr Biochem ; 25(9): 893-902, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24917046

ABSTRACT

Obesity is a global health problem and recently it has been seen as a growing concern for developing countries. Several bioactive dietary molecules have been associated with amelioration of obesity and associated complications and capsaicin is one among them. The present work is an attempt to understand and provide evidence for the novel mechanisms of anti-obesity activity of capsaicin in high fat diet (HFD)-fed mice. Swiss albino mice divided in three groups (n=8-10) i.e. control, HFD fed and capsaicin (2mg/kg, po)+HFD fed were administered respective treatment for 3months. After measuring phenotypic and serum related biochemical changes, effect of capsaicin on HFD-induced transcriptional changes in hypothalamus, white adipose tissue (WAT) (visceral and subcutaneous), brown adipose tissue (BAT) and gut microbial alterations was studied and quantified. Our results suggest that, in addition to its well-known effects, oral administration of capsaicin (a) modulates hypothalamic satiety associated genotype, (b) alters gut microbial composition, (c) induces "browning" genotype (BAT associated genes) in subcutaneous WAT and (d) increases expression of thermogenesis and mitochondrial biogenesis genes in BAT. The present study provides evidence for novel and interesting mechanisms to explain the anti-obesity effect of capsaicin.


Subject(s)
Capsaicin/pharmacology , Diet, High-Fat , Hypothalamus/drug effects , Intestines/microbiology , Transcription, Genetic/drug effects , Animals , Bacteria/isolation & purification , Base Sequence , Blood Glucose/metabolism , Body Weight/drug effects , Colony Count, Microbial , DNA Primers , Feeding Behavior/drug effects , Gene Expression/drug effects , Hypothalamus/metabolism , Male , Mice , Weight Gain/drug effects
20.
Age (Dordr) ; 36(3): 9651, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24789008

ABSTRACT

Poor blood glucose homeostatic regulation is common, consequential, and costly for older and elderly populations, resulting in pleiotrophically adverse clinical outcomes. Somatotrophic signaling deficiency and dietary restriction have each been shown to delay the rate of senescence, resulting in salubrious phenotypes such as increased survivorship. Using two growth hormone (GH) signaling-related, slow-aging mouse mutants we tested, via longitudinal analyses, whether genetic perturbations that increase survivorship also improve blood glucose homeostatic regulation in senescing mammals. Furthermore, we institute a dietary restriction paradigm that also decelerates aging, an intermittent fasting (IF) feeding schedule, as either a short-term or a sustained intervention beginning at either middle or old age, and assess its effects on blood glucose control. We find that either of the two genetic alterations in GH signaling ameliorates fasting hyperglycemia; additionally, both longevity-inducing somatotrophic mutations improve insulin sensitivity into old age. Strikingly, we observe major and broad improvements in blood glucose homeostatic control by IF: IF improves ad libitum-fed hyperglycemia, glucose tolerance, and insulin sensitivity, and reduces hepatic gluconeogenesis, in aging mutant and normal mice. These results on correction of aging-resultant blood glucose dysregulation have potentially important clinical and public health implications for our ever-graying global population, and are consistent with the Longevity Dividend concept.


Subject(s)
Aging/physiology , Blood Glucose/metabolism , Fasting/blood , Growth Hormone/deficiency , Insulin Resistance/physiology , Insulin/blood , Longevity/physiology , Animals , Caloric Restriction , Energy Metabolism , Female , Male , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...