Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
World J Clin Cases ; 9(31): 9333-9349, 2021 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-34877270

RESUMO

Inflammation is one of the primary factors associated with the causation and/or progression of several lifestyle disorders, including obesity, type 2 diabetes and non-alcoholic fatty liver disease (NAFLD). NAFLD is a spectrum of disorders, and starts with simple steatosis, progresses to non-alcoholic steatohepatitis, and then advances to fibrosis, cirrhosis and finally, hepatocellular carcinoma, due to perpetual cycles of insults caused by inflammation and other cellular stress. Emerging evidence has documented that patients with NAFLD have severe coronavirus disease 2019 (COVID-19), and patients with COVID-19 have a higher liver injury and mortality. Although the exact cause or mechanism is not known, inflammatory cytokine storm is a characteristic feature of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and is known to be associated with higher mortality among COVID-19 patients. Therefore, the COVID-19 pandemic seems to be a major concern in NAFLD patients, who have contracted SARS-CoV-2 infection and develop COVID-19. This is evident in patients at any stage of the NAFLD spectrum, as the inflammatory cytokine storm may cause and/or aggravate the progression or severity of NAFLD. Thus, there is a need for resolution of the inflammatory cytokine storm in these patients. A large body of evidence has demonstrated the efficacy of omega-3 long-chain polyunsaturated fatty acids (ω-3 LCPUFA) in NAFLD conditions, due to their anti-inflammatory, immunomodulatory and anti-viral properties. Therefore, intervention with ω-3 LCPUFA, an effective pharmaconutrient along with the standard treatment for COVID-19 may be useful in the management of the NAFLD spectrum in COVID-19 patients with pre-existing NAFLD conditions by resolving the inflammatory cytokine storm and thereby attenuating its progression. Although there are challenges in implementation, optimistically they can be circumvented and the pharmaconutrition strategy may be potentially helpful in tackling both the pandemics; NAFLD and COVID-19 at least in this subset of patients.

2.
Cell Biochem Funct ; 37(8): 578-590, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31495961

RESUMO

Here, we tested a hypothesis that vitamin A and/or its metabolic pathways are involved in the high-fructose-mediated alteration in adipose tissue biology. For this purpose, weanling male Wistar rats were provided with one of the following diets: control (C), control with vitamin A deficiency (C-VAD), high fructose (HFr), and HFr with VAD (HFr-VAD) for 16 weeks, except that half of the C-VAD diet-fed rats were shifted to HFr diet (C-VAD(s)HFr), after 8-week period. Compared with control, feeding of HFr diet significantly increased the triglyceride content (P ≤ .01) and thus adipocyte size (hypertrophy) (P ≤ .001) in visceral adipose depot; retroperitoneal white adipose tissue (RPWAT) and these changes were corroborated with de novo lipogenesis, as evidenced by the increased glycerol-3-phosphate dehydrogenase activity (P ≤ .01) and up-regulation of lipogenic pathway transcripts, fructose transporter, and aldehyde dehydrogenase 1 A1. On the contrary, the absence of vitamin A in the HFr diet (HFr-VAD) failed to exert these changes; however, it induced adipocyte hyperplasia. Further, vitamin A deficiency-mediated changes were reversed by replenishment, as evident from the group that was shifted from C-VAD to HFr diet. In conclusion, vitamin A and its metabolic pathway play a key determinant role in the high-fructose-induced triglyceride accumulation and adipocyte hypertrophy of visceral white adipose depot. SIGNIFICANCE OF THE STUDY: Here, we report the metabolic impact of high-fructose feeding under vitamin A-sufficient and vitamin A-deficient conditions. Feeding of high-fructose diet induced triglyceride accumulation and adipocyte hypertrophy of the visceral white adipose depots. These changes corroborated with augmented expression of vitamin A and lipid metabolic pathway genes. Contrarily, absence of vitamin A in the high-fructose diet did not elicit such responses, while vitamin A replenishment reversed the changes exerted by vitamin A deficiency. To our knowledge, this is the first study to report the role of vitamin A and its metabolic pathway in the high-fructose-induced triglyceride synthesis and its accumulation in visceral adipose depot and thus provide a new insight and scope to understand these nutrients interaction in clinical conditions.


Assuntos
Frutose/farmacologia , Gordura Intra-Abdominal/efeitos dos fármacos , Triglicerídeos/metabolismo , Vitamina A/administração & dosagem , Adiponectina/análise , Adiponectina/sangue , Animais , Diferenciação Celular/efeitos dos fármacos , Dieta , Ácidos Graxos/análise , Ácidos Graxos/química , Gordura Intra-Abdominal/citologia , Gordura Intra-Abdominal/metabolismo , Leptina/análise , Leptina/sangue , Lipogênese/efeitos dos fármacos , Masculino , Ratos , Ratos Wistar , Vitamina A/metabolismo , Deficiência de Vitamina A/metabolismo , Deficiência de Vitamina A/patologia , Deficiência de Vitamina A/veterinária
3.
Int J Vitam Nutr Res ; 89(1-2): 62-72, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-30957704

RESUMO

The liver is the main site of lipid metabolism and vitamin A storage. Dietary factors are known to affect liver function, thereby leading to metabolic abnormalities. Here, we assessed the impact of long-term feeding of a high-fat diet on hepatic vitamin A status and lipid metabolism. For this purpose, 14 male and 14 female 35-day-old mice (strain C57BL/6J) were each divided into 2 groups of 7 animals and fed either a stock diet or a high-fat (HF) diet for 26 weeks. In addition to increased body weight/weight gain, the HF diet induced hypertriglyceridemia in both (p < 0.01). However, liver triglyceride levels were comparable among groups, which could be partly explained by unaltered expression of various lipogenic pathway proteins such as sterol regulatory element binding protein 1 (SREBP1), fatty acid synthase (FAS), microsomal triglyceride transfer protein (MTTP), and glycerol 3-phosphate acyl transferase (GPAT). On the other hand, hepatic retinol stores increased significantly in both sexes, whereas males displayed elevated circulatory retinol levels. Notably, long-term feeding of a HF diet elevated n-3 polyunsaturated fatty acid (PUFA) and docosahexaenoic acid (DHA, C22:6) levels in the liver (p ≤ 0.001), which is in line with the over-expression of very long-chain fatty acid elongase 2 (ELOVL2) protein in both sexes of mice (p < 0.01). In conclusion, very long-term feeding of a HF diet increased hepatic retinol stores and induced hypertriglyceridemia. However, it had no effect on hepatic triglyceride accumulation, possibly due to increased DHA levels arising from the ELOVL2-mediated elongation pathway.


Assuntos
Acetiltransferases/metabolismo , Dieta Hiperlipídica , Ácidos Docosa-Hexaenoicos , Fígado/fisiologia , Acetiltransferases/química , Animais , Elongases de Ácidos Graxos , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL
4.
Prev Nutr Food Sci ; 22(3): 172-183, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-29043214

RESUMO

Vitamin A and its metabolites modulate insulin resistance and regulate stearoyl-CoA desaturase 1 (SCD1), which are also known to affect insulin resistance. Here, we tested, whether vitamin A-mediated changes in insulin resistance markers are associated with SCD1 regulation or not. For this purpose, 30-week old male lean and glucose-intolerant obese rats of WNIN/GR-Ob strain were given either a stock or vitamin A-enriched diet, i.e. 2.6 mg or 129 mg vitamin A/kg diet, for 14 weeks. Compared to the stock diet, vitamin A-enriched diet feeding improved hyperglycemia and glucose-clearance rate in obese rats and no such changes were seen in lean rats receiving identical diets. These changes were corroborated with concomitant increase in circulatory insulin and glycogen levels of liver and muscle (whose insulin signaling pathway genes were up-regulated) in obese rats. Further, the observed increase in muscle glycogen content in these obese rats could be explained by increased levels of the active form of glycogen synthase, the key regulator of glycogen synthesis pathway, possibly inactivated through increased phosphorylation of its upstream inhibitor, glycogen synthase kinase. However, the unaltered hepatic SCD1 protein expression (despite decreased mRNA level) and increased muscle-SCD1 expression (both at gene and protein levels) suggest that vitamin A-mediated changes on glucose metabolism are not associated with SCD1 regulation. Chronic consumption of vitamin A-enriched diet improved hyperglycemia and glucose-intolerance, possibly, through the regulation of intracellular signaling and glycogen synthesis pathways of muscle and liver, but not associated with SCD1.

5.
J Sci Food Agric ; 97(5): 1582-1591, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27417700

RESUMO

BACKGROUND: Adipose tissue, an endocrine organ, plays a vital role not only in energy homeostasis, but also in the development and/or progression of various metabolic diseases, such as insulin resistance, type 2 diabetes and non-alcoholic fatty liver disease (NAFLD), via several factors and mechanisms, including inflammation. This study tested, whether carrot juice administration affected the adipose tissue development and its inflammatory status in a high fructose diet-induced rat model. For this purpose, male weanling Wistar rats were divided into four groups and fed either control or high fructose diet of AIN-93G composition with or without carrot juice ingestion for an 8 week period. RESULTS: Administration of carrot juice did not affect the adiposity and cell size of visceral fat depot; retroperitoneal white adipose tissue (RPWAT), which was corroborated with unaltered expression of genes involved in adipogenic and lipogenic pathways. However, it significantly reduced the high fructose diet-induced elevation of plasma free fatty acid (FFA) (P ≤ 0.05), macrophage chemoattractant protein 1 (MCP1) (P ≤ 0.01) and high sensitive C-reactive protein (hsCRP) (P ≤ 0.05) levels. CONCLUSION: Carrot juice administration attenuated the high fructose diet-induced elevation of levels of circulatory FFA and pro-inflammatory mediators; MCP1 and hsCRP without affecting the adiposity and cell size of visceral fat depot; RPWAT. © 2016 Society of Chemical Industry.


Assuntos
Daucus carota , Frutose/efeitos adversos , Sucos de Frutas e Vegetais , Adiposidade/efeitos dos fármacos , Animais , Proteína C-Reativa/efeitos dos fármacos , Fatores Quimiotáticos/efeitos adversos , Dieta , Ácidos Graxos não Esterificados/sangue , Mediadores da Inflamação/efeitos adversos , Gordura Intra-Abdominal/citologia , Gordura Intra-Abdominal/efeitos dos fármacos , Masculino , Ratos Wistar
6.
Indian J Med Res ; 144(2): 238-244, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27934803

RESUMO

BACKGROUND & OBJECTIVES: Hepatic scavenger receptor class B1 (SR-B1), a high-density lipoprotein (HDL) receptor, is involved in the selective uptake of HDL-associated esterified cholesterol (EC), thereby regulates cholesterol homoeostasis and improves reverse cholesterol transport. Previously, we reported in euglycaemic obese rats (WNIN/Ob strain) that feeding of vitamin A-enriched diet normalized hypercholesterolaemia, possibly through hepatic SR-B1-mediated pathway. This study was aimed to test whether it would be possible to normalize hypercholesterolaemia in glucose-intolerant obese rat model (WNIN/GR/Ob) through similar mechanism by feeding identical vitamin A-enriched diet. METHODS: In this study, 30 wk old male lean and obese rats of WNIN/GR-Ob strain were divided into two groups and received either stock diet or vitamin A-enriched diet (2.6 mg or 129 mg vitamin A/kg diet) for 14 wk. Blood and other tissues were collected for various biochemical analyses. RESULTS: Chronic vitamin A-enriched diet feeding decreased hypercholesterolaemia and normalized abnormally elevated plasma HDL-cholesterol (HDL-C) levels in obese rats as compared to stock diet-fed obese groups. Further, decreased free cholesterol (FC) and increased esterified cholesterol (EC) contents of plasma cholesterol were observed, which were reflected in higher EC to FC ratio of vitamin A-enriched diet-fed obese rats. However, neither lecithin-cholesterol acyltransferase (LCAT) activity of plasma nor its expression (both gene and protein) in the liver were altered. On the contrary, hepatic cholesterol levels significantly increased in vitamin A-enriched diet fed obese rats. Hepatic SR-B1 expression (both mRNA and protein) remained unaltered among groups. Vitamin A-enriched diet fed obese rats showed a significant increase in hepatic low-density lipoprotein receptor mRNA levels, while the expression of genes involved in HDL synthesis, namely, ATP-binding cassette protein 1 (ABCA1) and apolipoprotein A-I, were downregulated. No such response was seen in vitamin A-supplemented lean rats as compared with their stock diet-fed lean counterparts. INTERPRETATION & CONCLUSIONS: Chronic vitamin A-enriched diet feeding decreased hypercholesterolaemia and normalized HDL-C levels, possibly by regulating pathways involved in HDL synthesis and degradation, independent of hepatic SR-B1 in this glucose-intolerant obese rat model.


Assuntos
Colesterol/sangue , Hipercolesterolemia/sangue , Obesidade/sangue , Receptores Depuradores Classe B/biossíntese , Vitamina A/administração & dosagem , Transportador 1 de Cassete de Ligação de ATP/biossíntese , Animais , Apolipoproteína A-I/biossíntese , Transporte Biológico/genética , Colesterol/genética , HDL-Colesterol/biossíntese , HDL-Colesterol/sangue , Dieta , Modelos Animais de Doenças , Regulação da Expressão Gênica , Humanos , Hipercolesterolemia/dietoterapia , Hipercolesterolemia/genética , Fígado/metabolismo , Masculino , Redes e Vias Metabólicas/genética , Obesidade/dietoterapia , Obesidade/genética , Fosfatidilcolina-Esterol O-Aciltransferase/sangue , Ratos , Receptores Depuradores Classe B/genética , Vitamina A/metabolismo
7.
Prev Nutr Food Sci ; 21(3): 171-180, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27752492

RESUMO

Non-alcoholic fatty liver disease (NAFLD) is one of the most prevalent liver diseases associated with an altered lifestyle, besides genetic factors. The control and management of NAFLD mostly depend on lifestyle modifications, due to the lack of a specific therapeutic approach. In this context, we assessed the effect of carrot juice on the development of high fructose-induced hepatic steatosis. For this purpose, male weanling Wistar rats were divided into 4 groups, fed either a control (Con) or high fructose (HFr) diet of AIN93G composition, with or without carrot juice (CJ) for 8 weeks. At the end of the experimental period, plasma biochemical markers, such as triglycerides, alanine aminotransferase, and ß-hydroxy butyrate levels were comparable among the 4 groups. Although, the liver injury marker, aspartate aminotransferase, levels in plasma showed a reduction, hepatic triglycerides levels were not significantly reduced by carrot juice ingestion in the HFr diet-fed rats (HFr-CJ). On the other hand, the key triglyceride synthesis pathway enzyme, hepatic stearoyl-CoA desaturase 1 (SCD1), expression at mRNA level was augmented by carrot juice ingestion, while their protein levels showed a significant reduction, which corroborated with decreased monounsaturated fatty acids (MUFA), particularly palmitoleic (C16:1) and oleic (C18:1) acids. Notably, it also improved the long chain n-3 polyunsaturated fatty acid, docosahexaenoic acid (DHA; C22:6) content of the liver in HFr-CJ. In conclusion, carrot juice ingestion decreased the SCD1-mediated production of MUFA and improved DHA levels in liver, under high fructose diet-fed conditions. However, these changes did not significantly lower the hepatic triglyceride levels.

8.
Data Brief ; 6: 625-9, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26909377

RESUMO

Here, we present the expression data on various metabolic pathways of liver with special emphasize on lipid and carbohydrate metabolism and long chain polyunsaturated fatty acid (PUFA) synthesis, both at gene and protein levels. The data were obtained to understand the effect of vitamin A deficiency on the expression status (both gene and protein levels) of some of the key factors involved in lipogenesis, fatty acid oxidation, triglyceride secretion, long chain PUFA, resolvin D1 synthesis, glucose transport and glycogen synthesis of liver, using modern biology tools, such as quantitative real-time PCR (RT-PCR) and immunoblotting techniques. This data article provides the supporting evidence to the article "Vitamin A deficiency suppresses high fructose-induced triglyceride synthesis and elevates resolvin D1 levels" [1] and therefore, these data may be referred back, for comprehensive understanding and interpretations and for future studies.

9.
Biochim Biophys Acta ; 1861(3): 156-65, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26597784

RESUMO

BACKGROUND/AIMS: Vitamin A and its metabolites are known to regulate lipid metabolism. However so far, no study has assessed, whether vitamin A deficiency per se aggravates or attenuates the development of non-alcoholic fatty liver disease (NAFLD). Therefore, here, we tested the impact of vitamin A deficiency on the development of NAFLD. METHODS: Male weanling Wistar rats were fed one of the following diets; control, vitamin A-deficient (VAD), high fructose (HFr) and VAD with HFr (VADHFr) of AIN93G composition, for 16weeks, except half of the VAD diet-fed rats were shifted to HFr diet (VAD(s)HFr), at the end of 8(th) week. RESULTS: Animals fed on VAD diet with HFr displayed hypotriglyceridemia (33.5mg/dL) with attenuated hepatic triglyceride accumulation (8.2mg/g), compared with HFr diet (89.5mg/dL and 20.6mg/g respectively). These changes could be partly explained by the decreased activity of glycerol 3-phosphate dehydrogenase (GPDH) and the down-regulation of stearoyl CoA desaturase 1 (SCD1), both at gene and protein levels, the key determinants of triglyceride biosynthesis. On the other hand, n-3 long chain polyunsaturated fatty acid, docosahexaenoic acid and its active metabolite; resolvin D1 (RvD1) levels were elevated in the liver and plasma of VAD diet-fed groups, which was negatively associated with triglyceride levels. All these factors confer vitamin A deficiency-mediated protection against the development of hepatic steatosis, which was also evident from the group shifted from VAD to HFr diet. CONCLUSIONS: Vitamin A deficiency attenuates high fructose-induced hepatic steatosis, by regulating triglyceride synthesis, possibly through GPDH, SCD1 and RvD1.


Assuntos
Ácidos Docosa-Hexaenoicos/metabolismo , Frutose , Fígado/metabolismo , Hepatopatia Gordurosa não Alcoólica/prevenção & controle , Triglicerídeos/biossíntese , Deficiência de Vitamina A/metabolismo , Adiposidade , Animais , Modelos Animais de Doenças , Regulação para Baixo , Glicerolfosfato Desidrogenase/genética , Glicerolfosfato Desidrogenase/metabolismo , Gordura Intra-Abdominal/metabolismo , Gordura Intra-Abdominal/fisiopatologia , Fígado/patologia , Masculino , Hepatopatia Gordurosa não Alcoólica/induzido quimicamente , Hepatopatia Gordurosa não Alcoólica/genética , Hepatopatia Gordurosa não Alcoólica/metabolismo , Hepatopatia Gordurosa não Alcoólica/fisiopatologia , Ratos Wistar , Estearoil-CoA Dessaturase/genética , Estearoil-CoA Dessaturase/metabolismo , Regulação para Cima , Deficiência de Vitamina A/genética , Deficiência de Vitamina A/fisiopatologia , Redução de Peso
10.
Indian J Med Res ; 141(3): 275-84, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25963488

RESUMO

During the last century, vitamin A has evolved from its classical role as a fat-soluble vitamin and attained the status of para-/autocrine hormone. Besides its well-established role in embryogenesis, growth and development, reproduction and vision, vitamin A has also been implicated in several other physiological processes. Emerging experimental evidences emphasize adipose tissue as an active endocrine organ with great propensity to continuous growth (throughout life). Due to various genetic and lifestyle factors, excess energy accumulates in adipose tissue as fat, resulting in obesity and other complications such as type 2 diabetes, hypertension, and cardiovascular disease. Recent in vitro and in vivo studies have shed light on vitamin A metabolites; retinaldehyde and retinoic acid and participation of their pathway proteins in the regulation of adipose tissue metabolism and thus, obesity. In this context, we discuss here some of our important findings, which establish the role of vitamin A (supplementation) in obesity and its associated disorders by employing an obese rat model; WNIN/Ob strain.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Hipertensão/metabolismo , Obesidade/metabolismo , Vitamina A/metabolismo , Tecido Adiposo/metabolismo , Tecido Adiposo/patologia , Animais , Diabetes Mellitus Tipo 2/patologia , Suplementos Nutricionais , Humanos , Hipertensão/patologia , Inflamação/metabolismo , Inflamação/patologia , Resistência à Insulina , Obesidade/patologia , Ratos , Vitamina A/uso terapêutico
11.
Exp Physiol ; 100(11): 1352-61, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25900735

RESUMO

NEW FINDINGS: What is the central question of this study? Previously, we reported that chronic feeding of a vitamin A-enriched diet to euglycaemic obese rats (WNIN/Ob) ameliorated obesity. Does this diet exert similar effects even with a different genetic background, i.e. obese rats of the WNIN/GR-Ob strain with impaired glucose tolerance? What is the main finding and its importance? Vitamin A-enriched diet aggravated weight gain and adiposity/obesity in both lean and glucose-intolerant obese rats of the WNIN/GR-Ob strain. Therefore, the role of genetic factors and their regulation by nutrients in determining health and disease conditions assumes greater significance in experimental and clinical research. Vitamin A and its metabolites are key regulators of the development of adipose tissue and its associated metabolic complications. Here, we tested, in a glucose-intolerant obese rat model (the WNIN/GR-Ob stain), whether feeding a vitamin A-enriched diet alters adiposity and its associated changes. For this purpose, 30-week-old male lean and obese rats were divided into two groups and received either stock diet or vitamin A-enriched diet [2.6 or 129 mg vitamin A (kg diet)(-1) , respectively] for 14 weeks. At the end, feeding of the vitamin A-enriched diet resulted in increased body weight gain/obesity and retroperitoneal white adipose tissue (RPWAT) in both lean and obese rats of the WNIN/GR-Ob strain, when compared with their respective control animals receiving stock diet, without affecting food intake. An improvement in hypertriglyceridaemia and circulatory non-esterified fatty acid levels and unaltered hepatic fatty acid oxidative and triglyceride secretory pathway proteins with vitamin A-enriched diet feeding are suggestive of enhanced hepatic clearance of circulatory lipids, resulting in increased hepatic triglyceride accumulation. Transcriptional analysis of RPWAT showed that feeding the vitamin A-enriched diet augmented the expression of adipogenic/adipose tissue-specific genes; peroxisome proliferator-activated receptor-γ, stearoyl CoA desaturase 1, retinol saturase, leptin and lipoprotein lipase and vitamin A metabolic pathway genes; retinoic acid receptors, retinoid X receptors and cytochrome P450 26B1. Besides, RPWAT-lipoprotein lipase-mediated clearance of triglyceride could also have contributed to increased adiposity and improved hypertriglyceridaemia. In conclusion, chronic feeding of vitamin A-enriched diet induces weight gain and adiposity in both lean and obese rats of the WNIN/GR-Ob strain, possibly through transcriptional regulation of key adipogenic pathway genes of RPWAT, but improves dyslipidaemia.


Assuntos
Adiposidade/efeitos dos fármacos , Dieta , Obesidade/patologia , Vitamina A/administração & dosagem , Aumento de Peso/efeitos dos fármacos , Adipogenia/genética , Animais , Regulação da Expressão Gênica , Glucose , Hipertrigliceridemia/patologia , Fígado/química , Masculino , Ratos , Ratos Endogâmicos , Triglicerídeos/química
12.
Nutr Metab (Lond) ; 11(1): 45, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25302071

RESUMO

BACKGROUND: Previously, we reported that vitamin A-enriched diet (129 mg/kg diet) intake reduces the adiposity development in obese rats of WNIN/Ob strain. Here, we hypothesize that dose lesser than 129 mg of vitamin A/kg diet would also be effective in ameliorating the development of obesity in these rats. METHODS: Five-month-old male lean and obese rats designated as A & B were divided into four subgroups (I, II, III and IV) consisting of 8 rats from each phenotype and received diets containing 2.6 mg (control group), 26 mg, 52 mg and 129 mg vitamin A/kg diet as retinyl palmitate for 20 weeks. Body composition and morphological analysis of brown adipose tissue (BAT) was analyzed. Expression of uncoupling protein 1 (UCP1), retinoic acid receptor α (RARα) and retinoid X receptor α (RXRα) in BAT and levels of Bcl2 and Bax in epididymal white adipose tissue (eWAT) were determined by immunoblotting. RESULTS: Vitamin A supplementation to obese rats at doses of 52 and 129 mg/kg diet showed reduced body weight gain and adiposity compared to control diet-fed obese rats receiving 2.6 mg of vitamin A/kg diet. In BAT of obese rats, vitamin A supplementation at doses of 26 and 52 mg of vitamin A/kg diet resulted in increased UCP1 expression with concomitant decrease in RARα and RXRα levels compared to control diet-fed obese rats. Further, transmission electron microscopy study revealed an increase in number of BAT mitochondria of obese rats supplemented with 26 and 52 mg of vitamin A/kg diet. Also, obese rats fed on 52 mg/kg diet resulted in increased apoptosis by altering the ratio of Bcl2 to Bax protein levels in eWAT. Notably, most of these changes were not observed in lean rats fed vitamin A-enriched diets. CONCLUSION: In conclusion, chronic consumption of 52 mg of vitamin A/kg diet seems to be an effective dose in ameliorating obesity possibly through mitochondriogenesis, UCP1-mediated thermogenesis in BAT and apoptosis in eWAT of obese rats. Therefore, the role of dietary vitamin A in correcting human obesity would be of unquestionable relevance and can only be addressed by future studies.

13.
J Atheroscler Thromb ; 21(11): 1197-207, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25100235

RESUMO

AIM: Vitamin A plays a major role in lipid metabolism. Previously, we reported that chronic vitamin A feeding (129 mg/kg) for two months normalized the abnormally high plasma HDL-cholesterol (HDL-C) levels in hypercholesterolemic obese rats by upregulating the hepatic scavenger receptor class B type 1 (SR-BI) expression. In this report, we hypothesize that the administration of a dose less than 129 mg of vitamin A/kg would also be effective in lowering the plasma HDL-C levels in these rats. METHODS: Changes in the activity and expression of proteins related to RCT were analyzed together with blood parameters in five-month-old male lean and obese rats supplemented with 2.6 (control group), 26, 52 and 129 mg of vitamin A/kg as retinyl palmitate for 20 weeks. RESULTS: Vitamin A supplementation in the obese rats decreased the plasma HDL-C levels with a concomitant increase in the hepatic SR-BI expression and lipase activity compared to that observed in the control diet-fed obese rats treated with 2.6 mg of vitamin A/kg diet. Furthermore, vitamin A supplementation at doses of 52 and 129 mg/kg diet reduced the plasma lecithin cholesterol acyltransferase activity and increased the hepatic ATP-binding cassette transporter protein A1 expression in the obese rats. Interestingly, most of these changes were not observed in the lean rats fed a vitamin A-enriched diet. CONCLUSIONS: Chronic feeding of a vitamin A-enriched diet in hypercholesterolemic obese rats normalizes the plasma HDL-C level and presumably improves RCT, with an effective dose of 52 mg/kg diet. Further studies should focus on the pharmacological potential of vitamin A supplementation to correct an abnormal human obesity-associated lipoprotein metabolism.


Assuntos
Colesterol/metabolismo , Dieta , Hipercolesterolemia/metabolismo , Fígado/metabolismo , Obesidade/complicações , Magreza/complicações , Vitamina A/análogos & derivados , Animais , Antioxidantes/administração & dosagem , Transporte Biológico , Diterpenos , Humanos , Hipercolesterolemia/etiologia , Hipercolesterolemia/patologia , Masculino , Ratos , Ratos Mutantes , Ésteres de Retinil , Vitamina A/administração & dosagem
14.
Biochim Biophys Acta ; 1821(1): 190-7, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21704731

RESUMO

Morbidity and mortality associated with increased white fat accumulation in visceral fat depots have focused attention on the pathways regulating the development of this tissue during embryogenesis, in adulthood, and while under the influence of obesogenic diets. Adipocytes undergo clonal expansion, differentiation (adipogenesis) and maturation through a complex network of transcriptional factors, most of which are expressed at similar levels in visceral and subcutaneous fat. Rigorous research attempts to unfold the pathways regulating expression and activity of adipogenic transcription factors that act in a fat-depot-specific manner. Peroxisome proliferator-activated receptor-γ (PPARγ) is the master regulator of adipogenesis, and is expressed at higher levels in subcutaneous than in visceral depots. PPARγ expression in adipogenesis is mediated by CCAAT/enhancer binding proteins (C/EBPs) and several transcription factors acting in conjunction with C/EBPs, although alternative pathways through zinc-finger protein-423 (ZFP423) transcription factor are sufficient to induce PPARγ expression and adipogenesis. Vitamin A and its metabolites, retinaldehyde and retinoic acid, are transcriptionally-active molecules. Retinoic acid is generated from retinaldehyde in adipose tissue by the aldehyde dehydrogenase-1 family of enzymes (Aldh1). In this review, we discuss the role of Aldh1 enzymes in the generation of retinoic acid during adipogenesis, in the regulation of the transcriptional network of PPARγ in a fat-depot-specific manner, and the important contribution of this autocrine pathway in the development of visceral obesity. This article is part of a Special Issue entitled Retinoid and Lipid Metabolism.


Assuntos
Adipogenia , Tecido Adiposo Branco/metabolismo , Tretinoína/metabolismo , Tecido Adiposo Branco/enzimologia , Família Aldeído Desidrogenase 1 , Animais , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Proteínas de Ligação a DNA , Humanos , Isoenzimas/metabolismo , Camundongos , Camundongos Knockout , Obesidade/metabolismo , PPAR gama/biossíntese , PPAR gama/metabolismo , Ratos , Retinal Desidrogenase/metabolismo , Fatores de Transcrição/metabolismo
15.
Ann Nutr Metab ; 58(4): 272-80, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21912103

RESUMO

BACKGROUND: The impact of industrial trans fatty acids (TFAs) on lipid metabolism and health remains elusive. METHODS: We compared the effect of long-term (52 weeks) ingestion of 10% partially hydrogenated vegetable oil, providing 4.2% of total energy from TFAs, on hepatic lipid metabolism and muscle insulin sensitivity in weanling female Fischer rats with that of palmolein (monounsaturated fatty acid, MUFA), sunflower (n-6 polyunsaturated fatty acid, PUFA), and a blend of sunflower and fish oil (n-3 PUFA). RESULTS: The proportion of plasma high-density lipoprotein cholesterol in total cholesterol and reverse cholesterol transport-associated protein expressions were similar in all the groups. Despite higher lipogenic-pathway protein levels, steatosis or hypertriglyceridemia was not observed in TFA-fed rats. Though TFA ingestion had no effect on fasting plasma glucose, insulin levels or oral glucose tolerance, it significantly decreased muscle insulin-stimulated glucose uptake as compared to PUFAs. Further, TFA ingestion increased adipose tissue retinol-binding protein 4 mRNA as compared to PUFAs (n-6 or n-3). The effects of MUFA (oleic acid) on all these parameters were comparable to those observed for TFAs. CONCLUSIONS: Compared to PUFA-rich diets, chronic consumption of a TFA-rich diet did not lead to steatosis or hypertriglyceridemia; however, it significantly impaired muscle insulin sensitivity, while no changes were found in the oral glucose tolerance test.


Assuntos
Colesterol/metabolismo , Dieta/efeitos adversos , Resistência à Insulina , Fígado/metabolismo , Músculos/metabolismo , Ácidos Graxos trans/efeitos adversos , Animais , Colesterol/sangue , Diafragma/metabolismo , Fígado Gorduroso/etiologia , Feminino , Manipulação de Alimentos , Regulação da Expressão Gênica , Intolerância à Glucose/etiologia , Hidrogenação , Hipertrigliceridemia/etiologia , RNA Mensageiro/metabolismo , Ratos , Ratos Endogâmicos F344 , Proteínas Plasmáticas de Ligação ao Retinol/genética , Proteínas Plasmáticas de Ligação ao Retinol/metabolismo , Ácidos Graxos trans/administração & dosagem , Desmame
16.
Mol Endocrinol ; 25(5): 799-809, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21436255

RESUMO

Vitamin A metabolite retinoic acid (RA) regulates life-sustaining differentiation processes and metabolic homeostasis. The aldehyde dehydrogenase-1 (Aldh1) family of enzymes (Aldh1a1, a2, and a3) catalyzes RA production from retinaldehyde and thereby controls concentrations of this transcriptionally active metabolite. The hierarchy of Aldh1 functions in adipose tissue has not been elucidated. We hypothesized that Aldh1 enzymes produce endogenous RA and regulate adipogenesis and fat formation in a fat depot-specific manner. We demonstrate that adipogenesis in vitro is accompanied by RA production generated primarily by Aldh1a1. In Aldh1a1-deficient adipocytes, adipogenesis is impaired compared with wild-type adipocytes due to markedly reduced expression of PPARγ regulated through zinc-finger protein 423 (ZFP423)-dependent mechanisms. These effects were recovered to some extent either by RA stimulation or overexpression of any of the Aldh1 enzymes in Aldh1a1(-/-) cells arguing that Aldh1a1 plays a dominant role in autocrine RA production. In vivo studies in C57/BL6 and Aldh1a1(-/-) mice on a regular diet revealed that multiple Aldh1 enzymes regulate differences in the formation of sc and visceral fat. In Aldh1a1(-/-) mice, visceral fat essentially lacked all Aldh1 expression. This loss of RA-producing enzymes was accompanied by 70% decreased expression of ZFP423, PPARγ, and Fabp4 in visceral fat of Aldh1a1(-/-) vs. wild-type mice and by the predominant loss of visceral fat. Subcutaneous fat of Aldh1a1(-/-) mice expressed Aldh1a3 for RA production that was sufficient to maintain expression of ZFP423 and PPARγ and sc fat mass. Our data suggest a paradigm for regulation of fat depots through the concerted action of Aldh1 enzymes that establish RA-dependent tandem regulation of transcription factors ZFP423 and PPARγ in a depot-specific manner.


Assuntos
Adipogenia , Isoenzimas/metabolismo , Retinal Desidrogenase/metabolismo , Células 3T3-L1 , Adipócitos/citologia , Adipócitos/enzimologia , Adulto , Família Aldeído Desidrogenase 1 , Animais , Distribuição da Gordura Corporal , Fator de Ligação a CCAAT/metabolismo , Proteínas de Ligação ao Cálcio , Diferenciação Celular , Citocinas/metabolismo , Proteínas de Ligação a DNA/metabolismo , Feminino , Genes Reporter , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Gordura Intra-Abdominal/metabolismo , Luciferases/biossíntese , Camundongos , Camundongos Endogâmicos C57BL , Nicotinamida Fosforribosiltransferase/metabolismo , PPAR gama/genética , PPAR gama/metabolismo , Elementos de Resposta , Gordura Subcutânea/metabolismo , Fatores de Transcrição/metabolismo , Transcrição Gênica , Tretinoína/metabolismo
17.
Nutr Metab (Lond) ; 6: 27, 2009 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-19519902

RESUMO

BACKGROUND: Microsomal stearoyl-CoA desaturase1 (SCD1) is the rate limiting enzyme involved in the biosynthesis of monounsaturated fatty acids (MUFAs); palmitoleic (16:1) and oleic (18:1) acid from their respective substrates palmitic (16:0) and stearic (18:0) acids. The ratio of 18:1 to 18:0 has been implicated in the regulation membrane fluidity and function. SCD1 is abundantly expressed in obese humans as well as rodent models. However, no studies have correlated the fatty acid desaturation index (16:1/16:0 and 18:1/18:0), an indicator of SCD1 activity with the markers of obesity in terms of body mass index (BMI) and adiposity index (AI). Therefore, here, we attempted to relate the fatty acid desaturation index with BMI and AI in Wistar NIN-obese mutant rat strains namely, WNIN/Ob and WNIN/GR-Ob (with impaired glucose tolerance). METHODS: For this purpose, 200 days old male 6 lean and 6 obese rats of both strains were taken. Fatty acid composition was analyzed in plasma, various tissues such as liver, white adipose tissues (retroperitoneal, epididymal, omental, and subcutaneous) and brown adipose tissue. RESULTS: Fatty acid composition data showed significant increase in palmitoleic (16:1) and oleic (18:1) acid levels, which were reflected in increased desaturation index (16:1/16:0 and 18:1/18:0) in plasma and all the tissues of obese rats of both strains, when compared with their respective age and sex-matched lean rats. Further, we found a strong positive correlation between desaturation index, BMI and AI in plasma and most of the tissues analyzed. CONCLUSION: So far, plasma Δ9 desaturation index has been well correlated with hypertriglyceridemia and we, by employing two models of obesity namely, WNIN/Ob and WNIN/GR-Ob, have shown Δ9 desaturation index of plasma correlated with physical markers of obesity such as BMI and AI. In conclusion, Δ9 desaturation index may serve as a potential sensitive biochemical marker to assess the degree of obesity and impact of therapeutic/nutritional interventions to combat obesity, along with other indicators.

18.
Atherosclerosis ; 204(1): 136-40, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-18848326

RESUMO

Dietary fatty acids are known to play an important role in the development as well as prevention of dyslipidemia. In this study, we evaluated the impact of feeding polyunsaturated fatty acids (PUFAs) for a period of 4 months on various aspects of cholesterol metabolism in genetically obese mutant rats of WNIN/GR-Ob strain. Based on their phenotype, lean and obese rats were divided into two groups, A and B respectively, and further subdivided depending on the type of dietary fat. Control groups of rats (AI and BI), were fed on 4% groundnut oil, which was replaced by safflower oil; n-6 PUFA diet (AII and BII) or oil blend of safflower and soybean oil, n-6 and n-3 PUFA diet (AIII and BIII) in the experimental groups. It was observed that feeding of diets with n-6 PUFA or a combination of n-6 and n-3 PUFAs resulted in marked elevation of plasma levels of total as well as HDL cholesterol and triglycerides in obese rats (BII and BIII), as compared to the control group (BI). Further, plasma HDL fraction of obese rats had elevated apolipoprotein E (apo E), while apo A1 levels remained unaltered. Increased lecithin: cholesterol acyltransferase (LCAT) activity and cholesteryl ester (CE) levels in the plasma and enhanced expression of hepatic scavenger receptor class B type1 (SR-B1) were also observed in PUFA-fed obese rats (BII and BIII). However, there was no change in hepatic ATP-binding cassette transporter protein A1 (ABCA1) levels in the obese rats fed on PUFA rich diets. Intriguingly, though these changes favor efficient removal of cholesterol from peripheral tissues, its esterification and enhanced clearance through reverse cholesterol transport (RCT); plasma HDL-C remained higher in these genetically dyslipidemic obese rats, thereby pointing at yet unknown mechanisms, involved in cholesterol homeostasis, which need to be studied.


Assuntos
Colesterol/metabolismo , Gorduras na Dieta/administração & dosagem , Dislipidemias/metabolismo , Ácidos Graxos Ômega-3/administração & dosagem , Ácidos Graxos Ômega-6/administração & dosagem , Fígado/metabolismo , Obesidade/metabolismo , Transportador 1 de Cassete de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/metabolismo , Animais , Apolipoproteínas/sangue , Colesterol/sangue , Ésteres do Colesterol/sangue , HDL-Colesterol/sangue , Gorduras na Dieta/metabolismo , Modelos Animais de Doenças , Ácidos Graxos Ômega-3/metabolismo , Ácidos Graxos Ômega-6/metabolismo , Masculino , Óleo de Amendoim , Fosfatidilcolina-Esterol O-Aciltransferase/sangue , Óleos de Plantas/administração & dosagem , Ratos , Ratos Mutantes , Óleo de Cártamo/administração & dosagem , Receptores Depuradores Classe B/metabolismo , Óleo de Soja/administração & dosagem , Triglicerídeos/sangue
19.
Obesity (Silver Spring) ; 15(2): 322-9, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17299104

RESUMO

OBJECTIVE: Scavenger receptor class BI (SR-BI), authentic high-density lipoprotein (HDL) receptors expressed in liver, are known to play an important role in HDL-cholesterol (C) metabolism and reverse cholesterol transport. Interestingly, obese rats of WNIN/Ob strain have abnormally elevated levels of serum HDL-C compared with their lean counterparts. Based on the well-established role of SR-B1 in HDL-C metabolism, it was hypothesized that these obese rats may have an underexpression of hepatic SR-B1 receptors. In view of the significant role of vitamin A in energy expenditure and obesity, we also tested whether vitamin A supplementation can correct abnormal HDL-C metabolism. RESEARCH METHODS AND PROCEDURES: To test this hypothesis, 7-month-old male lean and obese rats of WNIN/Ob strain were divided into two groups; each group was subdivided into two subgroups consisting of six lean and six obese rats and received diets containing either 2.6 or 129 mg vitamin A/kg diet for 2 months. RESULTS: At the end, obese rats receiving normal levels of vitamin A diet showed high serum HDL-C and lower hepatic SR-BI expression levels compared with lean counterparts. Furthermore, chronic dietary vitamin A supplementation resulted in overexpression of hepatic SR-BI receptors (protein and gene) with concomitant reduction in serum HDL-C levels in obese rats. DISCUSSION: Thus, our observations highlight the role of vitamin A in reverse cholesterol transport through up-regulation of hepatic SR-BI receptors and, thereby, HDL-C homeostasis in obese rats of WNIN/Ob strain.


Assuntos
HDL-Colesterol/metabolismo , Obesidade/metabolismo , Receptores Depuradores Classe B/metabolismo , Vitamina A/farmacologia , Animais , HDL-Colesterol/sangue , Dieta , Lipídeos/análise , Lipídeos/sangue , Fígado/química , Fígado/efeitos dos fármacos , Masculino , Obesidade/sangue , Obesidade/patologia , RNA Mensageiro/metabolismo , Ratos , Ratos Endogâmicos , Receptores Depuradores Classe B/sangue
20.
Obesity (Silver Spring) ; 14(1): 52-9, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16493122

RESUMO

OBJECTIVE: To understand the possible role of chronic dietary high vitamin A supplementation in body weight regulation and obesity using a novel WNIN/Ob obese rat model developed at the National Centre for Laboratory Animal Sciences of National Institute of Nutrition, India. RESEARCH METHODS AND PROCEDURES: Thirty-six 7-month-old male rats of lean, carrier, and obese phenotypes were broadly divided into two groups; each group was subdivided into three subgroups consisting of six lean, six carrier, and six obese rats and received diets containing either 2.6 or 129 mg vitamin A/kg of diet for 2 months. Body weight gain, food intake, and weights of various organs were recorded. Adiposity index and BMI were calculated. Serum and liver retinol and brown adipose tissue (BAT)-uncoupling protein1 (UCP1) mRNA expression levels were quantified. RESULTS: Chronic feeding of high but non-toxic doses of vitamin A through diet significantly reduced (P < or = 0.05) body weight gain, adiposity index, and retroperitoneal white adipose tissue mass (without affecting food intake) in obese rats compared with their lean and carrier counterparts. In general, vitamin A treatment significantly improved hepatic retinol stores (P < or = 0.05) in all phenotypes without affecting serum free retinol levels. However, augmented BAT-UCP1 expression was observed only in carrier and obese rats (whose basal expression was low). DISCUSSION: Our data suggest that chronic dietary vitamin A supplementation at high doses effectively regulates obesity in obese phenotype of the WNIN/Ob strain, possibly through up-regulation of the BAT-UCP1 gene and associated adipose tissue loss. However, in vitamin A-supplemented lean and carrier rats, changes in adiposity could not be related to BAT-UCP1 expression levels.


Assuntos
Ingestão de Energia/efeitos dos fármacos , Obesidade/metabolismo , Vitamina A/farmacologia , Vitaminas/farmacologia , Aumento de Peso/efeitos dos fármacos , Tecido Adiposo/metabolismo , Animais , Proteínas de Transporte/metabolismo , Suplementos Nutricionais , Relação Dose-Resposta a Droga , Canais Iônicos , Fígado/metabolismo , Masculino , Proteínas de Membrana/metabolismo , Proteínas Mitocondriais , Obesidade/genética , Fenótipo , Distribuição Aleatória , Ratos , Ratos Endogâmicos , Proteína Desacopladora 1 , Vitamina A/administração & dosagem , Vitamina A/metabolismo , Vitaminas/administração & dosagem , Vitaminas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...