Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Food Funct ; 11(1): 297-304, 2020 Jan 29.
Article in English | MEDLINE | ID: mdl-31915782

ABSTRACT

In recent years, the conversion of white adipocytes to brown-like adipocytes by pharmacological and dietary compounds has gained attention as an effective strategy to fight obesity. Strawberry bioactive compounds present several biological activities including antioxidant, anti-inflammatory, anti-cancer, anti-atherosclerotic and antiadipogenic properties. However, to the best of our knowledge, the possible role of strawberry bioactive compounds in white adipose tissue (WAT) browning has never been explored. Our results demonstrated that a strawberry methanolic extract (SE) significantly reduced 3T3-L1 pre-adipocytes differentiation, and down-regulated the mRNA expression of the adipogenic transcription factors CCAAT/enhancer-binding protein (C/REB- α) and peroxisome proliferation-activated receptor (PPAR-γ). It also down-regulated the mRNA expression of resistin and angiotensinogen, two genes considered as markers of white adipocytes, while increased the mRNA expression of pyruvate dehydrogenase lipoamide kinase isozyme 4 (PDK4) and uncoupling protein 1 (UCP1) which, conversely, are brown adipocyte-specific markers. Likewise, SE stimulated AMP-activated protein kinase (AMPKα), sirtuin 1 (Sirt1) and the peroxisome proliferator activated receptor gamma coactivator 1-alpha (PGC-1α), suggesting a possible increase in mitochondrial biogenesis. It also stimulated oxygen consumption rate and uncoupled respiration. Taken together, all these results suggest that SE induces brown fat-like phenotype in 3T3-L1 cells and may have potential therapeutic implications for treatment and/or prevention of obesity.


Subject(s)
Adipocytes, Brown/cytology , Adipocytes, White/drug effects , Adipogenesis/drug effects , Fragaria/chemistry , Plant Extracts/pharmacology , 3T3-L1 Cells , AMP-Activated Protein Kinases/metabolism , Adipocytes, White/cytology , Animals , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Cell Differentiation , Methanol , Mice , PPAR gamma/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/metabolism , Sirtuin 1/metabolism , Transcription Factors/metabolism , Uncoupling Protein 1/metabolism
2.
Planta Med ; 86(1): 45-54, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31663108

ABSTRACT

A previous study showed that the meroterpenoid-rich fraction of an ethanolic extract of Sargassum serratifolium (MES) stimulated adipose tissue browning and inhibited diet-induced obesity and metabolic syndrome. Sargaquinoic acid (SQA) is a major component in MES. We investigated the effects of SQA on the differentiation of preadipocytes to the beige adipocytes. SQA was treated in 3T3-L1 adipocytes differentiated under a special condition that has been reported to induce the browning of adipocytes. SQA at 10 µM reduced lipid accumulation by approximately 23%. SQA at 2.5 - 10 µM induced the differentiation of white adipocytes to beige adipocytes partially by increasing the mitochondrial density and the expression of beige/brown adipocyte markers. In addition, SQA activated lipid catabolic pathways, evidenced by the increased expression levels of perilipin, carnitine palmitoyltransferase 1, and acyl-CoA synthetase long-chain family member 1. As a partial mechanism, biochemical and in silico analyses indicate that SQA activated AMP-activated protein kinase signaling in adipocytes.


Subject(s)
Adipocytes, Brown/drug effects , Adipogenesis/drug effects , Alkenes/pharmacology , Benzoquinones/pharmacology , Sargassum/chemistry , 3T3-L1 Cells , AMP-Activated Protein Kinases/metabolism , Adipocytes, Brown/cytology , Alkenes/isolation & purification , Alkenes/toxicity , Animals , Benzoquinones/isolation & purification , Benzoquinones/toxicity , Mice , Signal Transduction/drug effects
3.
J Physiol Biochem ; 76(2): 193-211, 2020 May.
Article in English | MEDLINE | ID: mdl-31845114

ABSTRACT

The hypothalamus is a brain region in charge of many vital functions. Among them, BAT thermogenesis represents an essential physiological function to maintain body temperature. In the metabolic context, it has now been established that energy expenditure attributed to BAT function can contribute to the energy balance in a substantial extent. Thus, therapeutic interest in this regard has increased in the last years and some studies have shown that BAT function in humans can make a real contribution to improve diabetes and obesity-associated diseases. Nevertheless, how the hypothalamus controls BAT activity is still not fully understood. Despite the fact that much has been known about the mechanisms that regulate BAT activity in recent years, and that the central regulation of thermogenesis offers a very promising target, many questions remain still unsolved. Among them, the possible human application of knowledge obtained from rodent studies, and drug administration strategies able to specifically target the hypothalamus. Here, we review the current knowledge of homeostatic regulation of BAT, including the molecular insights of brown adipocytes, its central control, and its implication in the development of obesity.


Subject(s)
Adipocytes, Brown/metabolism , Hypothalamus/metabolism , Obesity/metabolism , Thermogenesis , Adipocytes, Brown/cytology , Animals , Energy Metabolism , Humans
4.
Int J Obes (Lond) ; 44(3): 715-726, 2020 03.
Article in English | MEDLINE | ID: mdl-31467421

ABSTRACT

OBJECTIVES: High-esterified pectin (HEP) is a prebiotic able to modulate gut microbiota, associated with health-promoting metabolic effects in glucose and lipid metabolism and adipostatic hormone sensitivity. Possible effects regulating adaptive thermogenesis and energy waste are poorly known. Therefore, we aimed to study how physiological supplementation with HEP is able to affect microbiota, energy metabolism and adaptive thermogenic capacity, and to contribute to the healthier phenotype promoted by HEP supplementation, as previously shown. We also attempted to decipher some of the mechanisms involved in the HEP effects, including in vitro experiments. SUBJECTS AND EXPERIMENTAL DESIGN: We used a model of metabolic malprogramming consisting of the progeny of rats with mild calorie restriction during pregnancy, both under control diet and an obesogenic (high-sucrose) diet, supplemented with HEP, combined with in vitro experiments in primary cultured brown and white adipocytes treated with the postbiotic acetate. RESULTS: Our main findings suggest that chronic HEP supplementation induces markers of brown and white adipose tissue thermogenic capacity, accompanied by a decrease in energy efficiency, and prevention of weight gain under an obesogenic diet. We also show that HEP promotes an increase in beneficial bacteria in the gut and peripheral levels of acetate. Moreover, in vitro acetate can improve adipokine production, and increase thermogenic capacity and browning in brown and white adipocytes, respectively, which could be part of the protection mechanism against excess weight gain observed in vivo. CONCLUSION: HEP and acetate stand out as prebiotic/postbiotic active compounds able to modulate both brown-adipocyte metabolism and browning and protect against obesity.


Subject(s)
Adipocytes, Brown/drug effects , Adipocytes, White/drug effects , Pectins/pharmacology , Prebiotics , Thermogenesis/drug effects , Acetates/metabolism , Acetates/pharmacology , Adipocytes, Brown/cytology , Adipocytes, Brown/metabolism , Adipocytes, White/cytology , Adipocytes, White/metabolism , Animals , Caloric Restriction , Dietary Supplements , Female , Gastrointestinal Microbiome/drug effects , Male , Pectins/administration & dosage , Pectins/metabolism , Pregnancy , Rats , Rats, Wistar
5.
Cell Death Differ ; 24(12): 2077-2088, 2017 12.
Article in English | MEDLINE | ID: mdl-28885620

ABSTRACT

Muscles of sarcopenic people show hypotrophic myofibers and infiltration with adipose and, at later stages, fibrotic tissue. The origin of infiltrating adipocytes resides in fibro-adipogenic precursors and nonmyogenic mesenchymal progenitor cells, and in satellite cells, the adult stem cells of skeletal muscles. Myoblasts and brown adipocytes share a common Myf5+ progenitor cell: the cell fate depends on levels of bone morphogenetic protein 7 (BMP-7), a TGF-ß family member. S100B, a Ca2+-binding protein of the EF-hand type, is expressed at relatively high levels in myoblasts from sarcopenic humans and exerts anti-myogenic effects via NF-κB-dependent inhibition of MyoD, a myogenic transcription factor acting upstream of the essential myogenic factor, myogenin. Adipogenesis requires high levels of ROS, and myoblasts of sarcopenic subjects show elevated ROS levels. Here we show that: (1) ROS overproduction in myoblasts results in upregulation of S100B levels via NF-κB activation; and (2) ROS/NF-κB-induced accumulation of S100B causes myoblast transition into brown adipocytes. S100B activates an NF-κB/Ying Yang 1 axis that negatively regulates the promyogenic and anti-adipogenic miR-133 with resultant accumulation of the brown adipogenic transcription regulator, PRDM-16. S100B also upregulates BMP-7 via NF-κB/Ying Yang 1 with resultant BMP-7 autocrine activity. Interestingly, myoblasts from sarcopenic humans show features of brown adipocytes. We also show that S100B levels and NF-κB activity are elevated in brown adipocytes obtained by culturing myoblasts in adipocyte differentiation medium and that S100B knockdown or NF-κB inhibition in myoblast-derived brown adipocytes reconverts them into fusion-competent myoblasts. At last, interstitial cells and, unexpectedly, a subpopulation of myofibers in muscles of geriatric but not young mice co-express S100B and the brown adipocyte marker, uncoupling protein-1. These results suggest that S100B is an important intracellular molecular signal regulating Myf5+ progenitor cell differentiation into fusion-competent myoblasts or brown adipocytes depending on its levels.


Subject(s)
Adipocytes, Brown/metabolism , MicroRNAs/metabolism , Myoblasts/metabolism , NF-kappa B/metabolism , Oxidative Stress/physiology , S100 Calcium Binding Protein beta Subunit/metabolism , Adipocytes, Brown/cytology , Animals , Bone Morphogenetic Protein 7/genetics , Bone Morphogenetic Protein 7/metabolism , Humans , Male , Mice , MicroRNAs/genetics , Myoblasts/cytology , Reactive Oxygen Species/metabolism , S100 Calcium Binding Protein beta Subunit/genetics , Transfection , YY1 Transcription Factor/metabolism
6.
Amino Acids ; 49(5): 957-964, 2017 05.
Article in English | MEDLINE | ID: mdl-28260165

ABSTRACT

L-Arginine has been reported to enhance brown adipose tissue developments in fetal lambs of obese ewes, but the underlying mechanism is unknown. The present study tested the hypothesis that L-arginine stimulates growth and development of brown adipocyte precursor cells (BAPCs) through activation of mammalian target of rapamycin cell signaling. BAPCs isolated from fetal lambs at day 90 of gestation were incubated   for 6 h in arginine-free DMEM, and then cultured in DMEM with concentrations of 50, 100, 200, 500 or 1000 µmol L-arginine/L for 24-96 h. Cell proliferation, protein turnover, the mammalian target of rapamycin (mTOR) signaling pathway and pre-adipocyte differentiation markers were determined. L-arginine treatment enhanced (P < 0.05) BAPC growth and protein synthesis, while inhibiting proteolysis in a dose-dependent manner. Compared with 50 and 100 µmol/L (the concentrations of arginine in the maternal plasma of obese ewes), 200 µmol L-arginine/L (the concentrations of arginine in the maternal plasma of obese ewes receiving arginine supplementation) increased (P < 0.05) the abundances of phosphorylated mTOR, P70S6K and 4EBP1, as well as the abundances of PGC1α, UCP1, BMP7 and PRDM16. These novel findings indicate that increasing extra-cellular arginine concentration from 50 to 200 µmol/L activates mTOR cell signaling in BAPCs and enhances their growth and development in a dose-dependent manner. Our results provide a mechanism for arginine supplementation to enhance the development of brown adipose tissue in fetal lambs.


Subject(s)
Adipocytes, Brown/drug effects , Arginine/pharmacology , Gene Expression Regulation, Developmental , Obesity/genetics , TOR Serine-Threonine Kinases/genetics , Adipocytes, Brown/cytology , Adipocytes, Brown/metabolism , Animals , Bone Morphogenetic Protein 7/genetics , Bone Morphogenetic Protein 7/metabolism , Cell Differentiation , Cell Line , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Eukaryotic Initiation Factors/genetics , Eukaryotic Initiation Factors/metabolism , Female , Fetus , Obesity/metabolism , Obesity/pathology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/genetics , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Sheep, Domestic , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism , Zinc Fingers/genetics
7.
J Agric Food Chem ; 63(35): 7721-30, 2015 Sep 09.
Article in English | MEDLINE | ID: mdl-26244589

ABSTRACT

This study was designed to evaluate the effects of Platycodon grandiflorum A. DC. ethanolic extract (PG) on obesity in brown/white preadipocytes. The effect of PG on the differentiation and mitochondrial biogenesis of brown adipocytes is still not examined. An in vivo study showed that PG induced weight loss in mice with high-fat-diet-induced obesity. PG successfully suppressed the differentiation of 3T3-L1 cells by down-regulating cellular induction of the peroxisome proliferators activated receptor γ (PPARγ), CCAAT enhancer binding protein α (C/EBPα), lipin-1, and adiponectin but increasing expression of silent mating type information regulation 2 homologue 1 (SIRT1) and the phosphorylation of AMP-activated protein kinase α (AMPKα). The effect of PG on the adipogenic factors was compared with that of its bioactive compound platycodin D. In addition, PG increased expressions of mitochondria-related genes, including uncoupling protein 1 (UCP1), peroxisome proliferator activated receptor-coactivator 1 α (PGC1α), PR domain containing 16 (PRDM16), SIRT3, nuclear respiratory factor (NRF), and cytochrome C (CytC) in primary brown adipocytes. These results indicate that PG stimulates the differentiation of brown adipocytes through modulation of mitochondria-related genes and could offer clinical benefits as a supplement to treat obesity.


Subject(s)
Adipocytes, Brown/drug effects , Adipogenesis/drug effects , Mitochondria/drug effects , Obesity/physiopathology , Plant Extracts/pharmacology , Platycodon/chemistry , 3T3-L1 Cells , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Adipocytes, Brown/cytology , Adipocytes, Brown/metabolism , Animals , CCAAT-Enhancer-Binding Protein-alpha/genetics , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Humans , Male , Mice , Mitochondria/genetics , Mitochondria/metabolism , Obesity/drug therapy , Obesity/genetics , Obesity/metabolism , PPAR gamma/genetics , PPAR gamma/metabolism , Plant Extracts/administration & dosage , Plant Extracts/isolation & purification
8.
J Transl Med ; 13: 135, 2015 Apr 30.
Article in English | MEDLINE | ID: mdl-25925588

ABSTRACT

BACKGROUND: Functional brown adipose tissue (BAT), involved in energy expenditure, has recently been detected in substantial amounts in adults. Formerly overlooked BAT has now become an attractive anti-obesity target. METHODS AND RESULTS: Molecular characterization of human brown and white adipocytes, using a myriad of techniques including high-throughput RNA sequencing and functional assays, showed that PAZ6 and SW872 cells exhibit classical molecular and phenotypic markers of brown and white adipocytes, respectively. However, the pre-adipocyte cell line SGBS presents a versatile phenotype. A transit expression of classical brown markers such as UCP1 and PPARγ peaked and declined at day 28 post-differentiation initiation. Conversely, white adipocyte markers, including Tcf21, showed reciprocal behavior. Interestingly, leptin levels peaked at day 28 whereas the highest adiponectin mRNA levels were detected at day 14 of differentiation. Phenotypic analysis of the abundance and shape of lipid droplets were consistent with the molecular patterns. Accordingly, the oxidative capacity of SGBS adipocytes peaked on differentiation day 14 and declined progressively towards differentiation day 28. CONCLUSIONS: Our studies have unveiled a new phenotype of human adipocytes, providing a tool to identify molecular gene expression patterns and pathways involved in the conversion between white and brown adipocytes.


Subject(s)
Adipocytes, Brown/metabolism , Adipocytes, White/metabolism , Adipogenesis , Adipocytes/cytology , Adipocytes, Brown/cytology , Adipocytes, White/cytology , Adiponectin/metabolism , Cell Differentiation , Cell Line , Cells, Cultured , DNA, Complementary/metabolism , Electron Transport Complex I/metabolism , Gene Expression Profiling , Gene Expression Regulation , High-Throughput Nucleotide Sequencing , Humans , Ion Channels/metabolism , Leptin/metabolism , Microscopy, Fluorescence , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Oxygen/chemistry , Phenotype , RNA, Messenger/metabolism , Sequence Analysis, RNA , Uncoupling Protein 1
9.
Genes Dev ; 29(3): 308-21, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25644605

ABSTRACT

PR domain-containing 16 (PRDM16) induces expression of brown fat-specific genes in brown and beige adipocytes, although the underlying transcription-related mechanisms remain largely unknown. Here, in vitro studies show that PRDM16, through its zinc finger domains, directly interacts with the MED1 subunit of the Mediator complex, is recruited to the enhancer of the brown fat-specific uncoupling protein 1 (Ucp1) gene through this interaction, and enhances thyroid hormone receptor (TR)-driven transcription in a biochemically defined system in a Mediator-dependent manner, thus providing a direct link to the general transcription machinery. Complementary cell-based studies show that upon forskolin treatment, PRDM16 induces Ucp1 expression in undifferentiated murine embryonic fibroblasts, that this induction depends on MED1 and TR, and, consistent with a direct effect, that PRDM16 is recruited to the Ucp1 enhancer. Related studies have defined MED1 and PRDM16 interaction domains important for Ucp1 versus Ppargc1a induction by PRDM16. These results reveal novel mechanisms for PRDM16 function through the Mediator complex.


Subject(s)
Adipocytes, Brown/cytology , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Developmental , Ion Channels/genetics , Mediator Complex Subunit 1/metabolism , Mitochondrial Proteins/genetics , Transcription Factors/metabolism , Adipocytes, Brown/metabolism , Animals , Cell Line , Colforsin/pharmacology , Enhancer Elements, Genetic/physiology , Gene Expression Regulation, Developmental/drug effects , HEK293 Cells , Humans , Mice , Protein Binding , Protein Structure, Tertiary/genetics , Uncoupling Protein 1
10.
Endocrinology ; 152(10): 3680-9, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21862615

ABSTRACT

Brown adipose tissue plays an important role in obesity, insulin resistance, and diabetes. We have previously shown that the transition from brown preadipocytes to mature adipocytes is mediated in part by insulin receptor substrate (IRS)-1 and the cell cycle regulator protein necdin. In this study, we used pharmacological inhibitors and adenoviral dominant negative constructs to demonstrate that this transition involves IRS-1 activation of Ras and ERK1/2, resulting in phosphorylation of cAMP response element-binding protein (CREB) and suppression of necdin expression. This signaling did not include an elevation of intracellular calcium. A constitutively active form of CREB expressed in IRS-1 knockout cells decreased necdin promoter activity, necdin mRNA, and necdin protein levels, leading to a partial restoration of differentiation. By contrast, forkhead box protein (Fox)O1, which is regulated by the phosphoinositide 3 kinase-Akt pathway, increased necdin promoter activity. Based on reporter gene assays using truncations of the necdin promoter and chromatin immunoprecipitation studies, we demonstrated that CREB and FoxO1 are recruited to the necdin promoter, likely interacting with specific consensus sequences in the proximal region. Based on these results, we propose that insulin/IGF-I act through IRS-1 phosphorylation to stimulate differentiation of brown preadipocytes via two complementary pathways: 1) the Ras-ERK1/2 pathway to activate CREB and 2) the phosphoinositide 3 kinase-Akt pathway to deactivate FoxO1. These two pathways combine to decrease necdin levels and permit the clonal expansion and coordinated gene expression necessary to complete brown adipocyte differentiation.


Subject(s)
Adipocytes, Brown/cytology , Cyclic AMP Response Element-Binding Protein/physiology , Forkhead Transcription Factors/physiology , Insulin-Like Growth Factor I/physiology , Insulin/physiology , Nerve Tissue Proteins/physiology , Nuclear Proteins/physiology , Adipogenesis , Animals , Cell Differentiation , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/metabolism , Forkhead Box Protein O1 , Insulin Receptor Substrate Proteins/physiology , Mice , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Phosphorylation , Promoter Regions, Genetic , Signal Transduction
11.
Cell Metab ; 13(5): 573-83, 2011 May 04.
Article in English | MEDLINE | ID: mdl-21531339

ABSTRACT

Hypothalamic neuropeptide Y (NPY) has been implicated in control of energy balance, but the physiological importance of NPY in the dorsomedial hypothalamus (DMH) remains unclear. Here we report that knockdown of NPY expression in the DMH by adeno-associated virus-mediated RNAi reduced fat depots in rats fed regular chow and ameliorated high-fat diet-induced hyperphagia and obesity. DMH NPY knockdown resulted in development of brown adipocytes in inguinal white adipose tissue through the sympathetic nervous system. This knockdown increased uncoupling protein 1 expression in both inguinal fat and interscapular brown adipose tissue (BAT). Consistent with the activation of BAT, DMH NPY knockdown increased energy expenditure and enhanced the thermogenic response to a cold environment. This knockdown also increased locomotor activity, improved glucose homeostasis, and enhanced insulin sensitivity. Together, these results demonstrate critical roles of DMH NPY in body weight regulation through affecting food intake, body adiposity, thermogenesis, energy expenditure, and physical activity.


Subject(s)
Adipocytes, Brown/cytology , Body Weight , Diet, Atherogenic , Hypothalamus/metabolism , Neuropeptide Y/physiology , Obesity/prevention & control , Adipocytes, Brown/metabolism , Adipose Tissue, White/cytology , Adipose Tissue, White/metabolism , Animals , Cells, Cultured , Dependovirus/genetics , Down-Regulation , Eating , Energy Metabolism , Glucose/metabolism , Glucose Tolerance Test , Homeostasis , Immunoblotting , Insulin/pharmacology , Ion Channels/genetics , Ion Channels/metabolism , Male , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Obesity/etiology , Obesity/metabolism , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Uncoupling Protein 1
12.
Front Biosci (Landmark Ed) ; 16(4): 1589-608, 2011 01 01.
Article in English | MEDLINE | ID: mdl-21196250

ABSTRACT

The last decade has witnessed a profound resurgence in brown adipose tissue (BAT) research. The need for such a dramatic increase stems from the ever-growing trend toward global obesity. Indeed, it is currently estimated that rates of obesity in developed countries such as the United States exceed 35% of the population (1). The higher incidence of obesity is associated with increased prevalence of the metabolic syndrome including diabetes, hypertension, and coronary heart disease, among others (1, 2). BAT holds great promise in combating obesity given its unprecedented metabolic capacity. Leading the way has been recent studies, which conclusively demonstrate significant quantities of functional BAT in adult humans (3-7). These findings have been complimented by elegant studies elucidating the developmental origin of the brown adipocyte and the transcriptional regulation involved in its differentiation. This review will attempt to meld the wealth of new information regarding BAT development with established literature to provide an up to date synopsis of what is known and thus a framework for future research directions.


Subject(s)
Adipose Tissue, Brown/physiology , Body Temperature Regulation/physiology , Adipocytes, Brown/cytology , Adipocytes, Brown/drug effects , Adipocytes, Brown/physiology , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/growth & development , Adult , Aged , Animals , Animals, Newborn , Arginine/administration & dosage , Body Temperature Regulation/drug effects , Bone Morphogenetic Proteins/physiology , Catecholamines/physiology , Cell Differentiation/drug effects , Cell Lineage , DNA-Binding Proteins/physiology , Diet/adverse effects , Dietary Supplements , Gene Expression Regulation , Heat-Shock Proteins/physiology , Humans , Infant, Newborn , Ion Channels/physiology , Lipolysis/drug effects , Middle Aged , Mitochondrial Proteins/physiology , Obesity/etiology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Receptors, Adrenergic, beta/physiology , Sheep , Thyroid Hormones/physiology , Transcription Factors/physiology , Uncoupling Protein 1
SELECTION OF CITATIONS
SEARCH DETAIL