Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Methods Mol Biol ; 2662: 135-145, 2023.
Article in English | MEDLINE | ID: mdl-37076677

ABSTRACT

In response to cold induction, brown adipose tissues (BAT) and emerged brown-like adipocytes (beige adipocytes) in subcutaneous white adipose tissues (WAT browning/beiging) are activated. Thermogenesis is increased during glucose and fatty acid uptake and metabolism in adult humans and mice. This activation of BAT or WAT beiging to generate heat helps to counteract diet-induced obesity. This protocol applies the glucose analog radiotracer 18F-fluorodeoxyglucose (FDG), coupled with positron emission tomography and computed tomography (PET/CT) scanning to evaluate cold-induced thermogenesis in the active BAT (interscapular region) and browned/beiged WAT (subcutaneous adipose region) in mice. The PET/CT scanning technique not only can quantify cold-induced glucose uptake in well-known BAT and beige-fat depots but also helps to visualize the anatomical location of novel uncharacterized mouse BAT and beige fat where cold-induced glucose uptake is high. Histological analysis is further employed to validate signals of delineated anatomical regions in PET/CT images as bona fide mouse BAT or beiged WAT fat depots.


Subject(s)
Adipose Tissue, Beige , Positron Emission Tomography Computed Tomography , Humans , Adult , Mice , Animals , Adipose Tissue, Beige/metabolism , Adipose Tissue, Beige/pathology , Adipose Tissue, White/diagnostic imaging , Adipose Tissue, White/metabolism , Adipose Tissue, Brown/diagnostic imaging , Adipose Tissue, Brown/metabolism , Obesity/metabolism , Uncoupling Protein 1/metabolism
2.
Biochimie ; 184: 26-39, 2021 May.
Article in English | MEDLINE | ID: mdl-33548390

ABSTRACT

Brown and beige adipose tissues are the primary sites for adaptive non-shivering thermogenesis. Although they have been known principally for their thermogenic effects, in recent years, it has emerged that, just like white adipose tissue (WAT), brown and beige adipose tissues also play an important role in the regulation of metabolic health through secretion of various brown adipokines (batokines) in response to various physiological cues. These secreted batokines target distant organs and tissues such as the liver, heart, skeletal muscles, brain, WAT, and perform various local and systemic functions in an autocrine, paracrine, or endocrine manner. Brown and beige adipose tissues are therefore now receiving increasing levels of attention with respect to their effects on various other organs and tissues. Identification of novel secreted factors by these tissues may help in the discovery of drug candidates for the treatment of various metabolic disorders such as obesity, type-2 diabetes, skeletal deformities, cardiovascular diseases, dyslipidemia. In this review, we comprehensively describe the emerging secretory role of brown/beige adipose tissues and the metabolic effects of various brown/beige adipose tissues secreted factors on other organs and tissues in endocrine/paracrine manners, and as well as on brown/beige adipose tissue itself in an autocrine manner. This will provide insights into understanding the potential secretory role of brown/beige adipose tissues in improving metabolic health.


Subject(s)
Adipokines/metabolism , Adipose Tissue, Beige/metabolism , Adipose Tissue, Brown/metabolism , Diabetes Mellitus, Type 2/metabolism , Obesity/metabolism , Thermogenesis , Adipose Tissue, Beige/pathology , Adipose Tissue, Brown/pathology , Animals , Diabetes Mellitus, Type 2/pathology , Humans , Obesity/pathology
3.
Med Sci Monit ; 26: e926789, 2020 Nov 23.
Article in English | MEDLINE | ID: mdl-33223514

ABSTRACT

BACKGROUND The mechanism of how intermittent fasting (IF) improves metabolism is not fully understood. Our study aimed to explore the effect of IF on lipid metabolism in obese mice, specifically on the intestinal flora. MATERIAL AND METHODS Diet-induced obese (DIO) mice were subjected to ad libitum (AL) feeding or IF (alternate-day fasting) for 30 days. We examined the lipid metabolism, fat distribution, gene expression of lipid metabolism, and intestinal flora in the mice. RESULTS Despite having access to the same high-fat diet as the AL-fed groups, IF mice displayed pronounced weight loss, and their lipid metabolism significantly improved, mainly reflected in lower serum lipid levels and ameliorated liver steatosis. IF also reduced metabolic endotoxemia in DIO mice. The 16S ribosomal deoxyribonucleic acid gene amplicon sequencing suggested that IF did not change the community richness but had a tendency to increase community diversity in the intestinal flora. In addition, IF significantly reduced the ratio of Firmicutes to Bacteroidetes and increased the relative abundance of Allobaculum in the intestinal flora. CONCLUSIONS IF can improve fat metabolism, reduce fat accumulation, promote white fat conversion to beige, and improve gut microbiota.


Subject(s)
Fasting , Gastrointestinal Microbiome , Lipid Metabolism , Adipose Tissue, Beige/pathology , Adipose Tissue, White/pathology , Animals , Body Weight , Diet, High-Fat , Discriminant Analysis , Dysbiosis/blood , Dysbiosis/microbiology , Energy Intake , Feeding Behavior , Lipopolysaccharides/blood , Male , Mice, Inbred C57BL , Mice, Obese
4.
Cardiovasc Pathol ; 49: 107259, 2020.
Article in English | MEDLINE | ID: mdl-32692664

ABSTRACT

Perivascular adipose tissue (PVAT) is a fat tissue deposit that encircles the vasculature. PVAT is traditionally known to protect the vasculature from external stimuli that could cause biological stress. In addition to the protective role of PVAT, it secretes certain biologically active substances known as adipokines that induce paracrine effects on proximate blood vessels. These adipokines influence vascular tones. There are different types of PVAT and they are phenotypically and functionally distinct. These are the white and brown PVATs. Under certain conditions, white PVAT could undergo phenotypic switch to attain a brown PVAT-like phenotype. This type of PVAT is referred to as Beige PVAT. The morphology of adipose tissue is influenced by species, age, and sex. These factors play significant roles in adipose tissue mass, functionality, paracrine activity, and predisposition to vascular diseases. The difficulty that is currently experienced in extrapolating animal models to human physiology could be traceable to these factors. Up till now, the involvement of PVAT in the development of vascular pathology is still not well understood. Brown and white PVAT contribute differently to vascular pathology. Thus, the PVAT could be a therapeutic target in curbing certain vascular diseases. In this review, knowledge would be updated on the multifaceted involvement of PVAT in vascular pathology and also explore its vascular therapeutic potential.


Subject(s)
Adipose Tissue, Beige/pathology , Adipose Tissue, Brown/pathology , Adipose Tissue, White/pathology , Arteries/pathology , Vascular Diseases/pathology , Adipokines/metabolism , Adipose Tissue, Beige/drug effects , Adipose Tissue, Beige/metabolism , Adipose Tissue, Beige/physiopathology , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/metabolism , Adipose Tissue, Brown/physiopathology , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Adipose Tissue, White/physiopathology , Adiposity , Animals , Arteries/drug effects , Arteries/metabolism , Arteries/physiopathology , Cardiovascular Agents/therapeutic use , Hemodynamics , Humans , Inflammation Mediators/metabolism , Paracrine Communication , Signal Transduction , Vascular Diseases/drug therapy , Vascular Diseases/metabolism , Vascular Diseases/physiopathology
5.
Clin Sci (Lond) ; 134(12): 1537-1553, 2020 06 26.
Article in English | MEDLINE | ID: mdl-32556103

ABSTRACT

Hyperuricaemia (HUA) significantly increases the risk of metabolic syndrome and is strongly associated with the increased prevalence of high serum free fatty acids (FFAs) and insulin resistance. However, the underlying mechanisms are not well established, especially the effect of uric acid (UA) on adipose tissue, a vital organ in regulating whole-body energy and FFA homeostasis. In the present study, we noticed that adipocytes from the white adipose tissue of patients with HUA were hypertrophied and had decreased UCP1 expression. To test the effects of UA on adipose tissue, we built both in vitro and in vivo HUA models and elucidated that a high level of UA could induce hypertrophy of adipocytes, inhibit their hyperplasia and reduce their beige-like characteristics. According to mRNA-sequencing analysis, UA significantly decreased the expression of leptin in adipocytes, which was closely related to fatty acid metabolism and the AMPK signalling pathway, as indicated by KEGG pathway analysis. Moreover, lowering UA using benzbromarone (a uricosuric agent) or metformin-induced activation of AMPK expression significantly attenuated UA-induced FFA metabolism impairment and adipose beiging suppression, which subsequently alleviated serum FFA elevation and insulin resistance in HUA mice. Taken together, these observations confirm that UA is involved in the aetiology of metabolic abnormalities in adipose tissue by regulating leptin-AMPK pathway, and metformin could lessen HUA-induced serum FFA elevation and insulin resistance by improving adipose tissue function via AMPK activation. Therefore, metformin could represent a novel treatment strategy for HUA-related metabolic disorders.


Subject(s)
Adipocytes/pathology , Adipose Tissue, Beige/pathology , Adipose Tissue, White/pathology , Fatty Acids, Nonesterified/blood , Hyperuricemia/blood , Hyperuricemia/drug therapy , Insulin Resistance , Metformin/therapeutic use , 3T3-L1 Cells , Adenylate Kinase/metabolism , Adipocytes/drug effects , Adipocytes/metabolism , Adipose Tissue, Beige/drug effects , Adipose Tissue, White/drug effects , Adult , Animals , Enzyme Activation , Female , Humans , Hypertrophy , Leptin/metabolism , Lipogenesis , Lipolysis , Male , Metformin/pharmacology , Mice , Mice, Inbred C57BL , Middle Aged , Signal Transduction , Triglycerides/metabolism , Uric Acid/blood
6.
Article in English | MEDLINE | ID: mdl-32425889

ABSTRACT

Bariatric surgeries induce marked and durable weight loss in individuals with morbid obesity through powerful effects on both food intake and energy expenditure. While alterations in gut-brain communication are increasingly implicated in the improved eating behavior following bariatric surgeries, less is known about the mechanistic basis for energy expenditure changes. Brown adipose tissue (BAT) and beige adipose tissue (BeAT) have emerged as major regulators of whole-body energy metabolism in humans as well as in rodents due to their ability to convert the chemical energy in circulating glucose and fatty acids into heat. In this Review, we critically discuss the steadily growing evidence from preclinical and clinical studies suggesting that Roux-en-Y gastric bypass (RYGB) and vertical sleeve gastrectomy (VSG), the two most commonly performed bariatric surgeries, enhance BAT/BeAT thermogenesis. We address the documented mechanisms, highlight study limitations and finish by outlining unanswered questions in the subject. Further understanding how and to what extent bariatric surgeries enhance BAT/BeAT thermogenesis may not only aid in the development of improved obesity pharmacotherapies that safely and optimally target both sides of the energy balance equation, but also in the development of novel hyperglycemia and/or hyperlipidemia pharmacotherapies.


Subject(s)
Adipose Tissue, Beige/pathology , Adipose Tissue, Brown/pathology , Bariatric Surgery/adverse effects , Thermogenesis , Animals , Humans
7.
J Clin Invest ; 130(5): 2319-2331, 2020 05 01.
Article in English | MEDLINE | ID: mdl-31961829

ABSTRACT

BACKGROUNDBeige adipose tissue is associated with improved glucose homeostasis in mice. Adipose tissue contains ß3-adrenergic receptors (ß3-ARs), and this study was intended to determine whether the treatment of obese, insulin-resistant humans with the ß3-AR agonist mirabegron, which stimulates beige adipose formation in subcutaneous white adipose tissue (SC WAT), would induce other beneficial changes in fat and muscle and improve metabolic homeostasis.METHODSBefore and after ß3-AR agonist treatment, oral glucose tolerance tests and euglycemic clamps were performed, and histochemical analysis and gene expression profiling were performed on fat and muscle biopsies. PET-CT scans quantified brown adipose tissue volume and activity, and we conducted in vitro studies with primary cultures of differentiated human adipocytes and muscle.RESULTSThe clinical effects of mirabegron treatment included improved oral glucose tolerance (P < 0.01), reduced hemoglobin A1c levels (P = 0.01), and improved insulin sensitivity (P = 0.03) and ß cell function (P = 0.01). In SC WAT, mirabegron treatment stimulated lipolysis, reduced fibrotic gene expression, and increased alternatively activated macrophages. Subjects with the most SC WAT beiging showed the greatest improvement in ß cell function. In skeletal muscle, mirabegron reduced triglycerides, increased the expression of PPARγ coactivator 1 α (PGC1A) (P < 0.05), and increased type I fibers (P < 0.01). Conditioned media from adipocytes treated with mirabegron stimulated muscle fiber PGC1A expression in vitro (P < 0.001).CONCLUSIONMirabegron treatment substantially improved multiple measures of glucose homeostasis in obese, insulin-resistant humans. Since ß cells and skeletal muscle do not express ß3-ARs, these data suggest that the beiging of SC WAT by mirabegron reduces adipose tissue dysfunction, which enhances muscle oxidative capacity and improves ß cell function.TRIAL REGISTRATIONClinicaltrials.gov NCT02919176.FUNDINGNIH: DK112282, P30GM127211, DK 71349, and Clinical and Translational science Awards (CTSA) grant UL1TR001998.


Subject(s)
Acetanilides/administration & dosage , Adrenergic beta-3 Receptor Agonists/administration & dosage , Blood Glucose/metabolism , Obesity/blood , Thiazoles/administration & dosage , Adipocytes/metabolism , Adipocytes/pathology , Adipose Tissue, Beige/metabolism , Adipose Tissue, Beige/pathology , Adult , Aged , Biopsy , Female , Gene Expression Regulation/drug effects , Humans , Male , Middle Aged , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Obesity/drug therapy , Obesity/pathology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/biosynthesis
8.
J Physiol Biochem ; 76(2): 213-226, 2020 May.
Article in English | MEDLINE | ID: mdl-31811543

ABSTRACT

Since the rediscovery of active brown and beige adipose tissues in humans a decade ago, great efforts have been made to identify the mechanisms underlying the activation and inactivation of these tissues, with the hope of designing potential strategies to fight against obesity and associated metabolic disorders such as type 2 diabetes. Active brown/beige fat increases the energy expenditure and is associated with reduced hyperglycemia and hyperlipidemia, whereas its atrophy and inactivation have been associated with obesity and aging. Autophagy, which is the process by which intracellular components are degraded within the lysosomes, has recently emerged as an important regulatory mechanism of brown/beige fat plasticity. Studies have shown that autophagy participates in the intracellular remodeling events that occur during brown/beige adipogenesis, thermogenic activation, and inactivation. The autophagic degradation of mitochondria appears to be important for the inactivation of brown fat and the transition from beige-to-white adipose tissue. Moreover, autophagic dysregulation in adipose tissues has been associated with obesity. Thus, understanding the regulatory mechanisms that control autophagy in the physiology and pathophysiology of adipose tissues might suggest novel treatments against obesity and its associated metabolic diseases.


Subject(s)
Adipose Tissue, Beige/metabolism , Adipose Tissue, Brown/metabolism , Autophagy , Metabolic Diseases/metabolism , Obesity/metabolism , Adipogenesis , Adipose Tissue, Beige/cytology , Adipose Tissue, Beige/pathology , Adipose Tissue, Brown/cytology , Adipose Tissue, Brown/pathology , Animals , Humans , Mitochondria/metabolism , Thermogenesis
9.
Sci Rep ; 9(1): 8658, 2019 06 17.
Article in English | MEDLINE | ID: mdl-31209239

ABSTRACT

In a recent study, repeated cold application induced beiging in subcutaneous white adipose tissue (SC WAT) of humans independent of body mass index. To identify factors that promote or inhibit beiging, we performed multiplex analysis of gene expression with the Nanostring nCounter system (the probe set contained genes for specific immune cell markers, cytokines, and chemokines) on the SC WAT from lean subjects. Multiple correlations analysis identified mast cell tryptase and CCL26, a chemokine for mast cells, as genes whose change correlated positively with the change in UCP1 in SC WAT, leading to the hypothesis that mast cells promote SC WAT beiging in response to cold. We quantified mast cell recruitment into SC WAT and degranulation. Mast cells increased in number in SC WAT in lean subjects, and there was an increase in the number of degranulated mast cells in both lean subjects and subjects with obesity. We determined that norepinephrine stimulated mast cell degranulation and histamine release in vitro. In conclusion, cold stimulated adipose tissue mast cell recruitment in lean subjects and mast cell degranulation in SC WAT of all research participants independent of baseline body mass index, suggesting that mast cells promote adipose beiging through the release of histamine or other products.


Subject(s)
Adipose Tissue, Beige/metabolism , Chemokine CCL26/genetics , Mast Cells/metabolism , Obesity/genetics , Subcutaneous Fat/metabolism , Thermogenesis/genetics , Tryptases/genetics , Adipose Tissue, Beige/pathology , Adult , Case-Control Studies , Cell Count , Cell Degranulation/drug effects , Cell Proliferation/drug effects , Chemokine CCL26/metabolism , Cold Temperature , Cytokines/genetics , Cytokines/metabolism , Energy Metabolism/drug effects , Energy Metabolism/genetics , Female , Gene Expression Profiling , Gene Expression Regulation , Histamine/biosynthesis , Humans , Male , Mast Cells/drug effects , Mast Cells/pathology , Norepinephrine/pharmacology , Obesity/metabolism , Obesity/pathology , Subcutaneous Fat/pathology , Tryptases/metabolism , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism
10.
J Clin Invest ; 129(6): 2305-2317, 2019 03 19.
Article in English | MEDLINE | ID: mdl-30888335

ABSTRACT

Chronic alcohol consumption causes liver injury, inflammation and fibrosis, thereby increasing morbidity and mortality. Paradoxically, modest drinking is believed to confer metabolic improvement, but the underlying mechanism remains elusive. Here, we have identified a novel hepatoprotective brain/brown adipose tissue (BAT)/liver axis. Alcohol consumption or direct alcohol administration into the brain stimulated hypothalamic neural circuits and sympathetic nerves innervating BAT, and dramatically increased BAT uncoupling protein 1 (Ucp1) expression and activity in a BAT sympathetic nerve-dependent manner. BAT and beige fat oxidized fatty acids to fuel Ucp1-mediated thermogenesis, thereby inhibiting lipid trafficking into the liver. BAT also secreted several adipokines, including adiponectin that suppressed hepatocyte injury and death. Genetic deletion of Ucp1 profoundly augmented alcohol-induced liver steatosis, injury, inflammation and fibrosis in male and female mice. Conversely, activation of BAT and beige fat through cold exposure suppressed alcoholic liver disease development. Our results unravel an unrecognized brain alcohol-sensing/sympathetic nerve/BAT/liver axis that counteracts liver steatosis and injury.


Subject(s)
Adipose Tissue, Brown/metabolism , Ethanol/adverse effects , Fatty Liver, Alcoholic/metabolism , Liver/metabolism , Adipose Tissue, Beige/metabolism , Adipose Tissue, Beige/pathology , Adipose Tissue, Brown/pathology , Animals , Cold Temperature , Ethanol/pharmacology , Fatty Liver, Alcoholic/genetics , Fatty Liver, Alcoholic/pathology , Female , Hypothalamus/metabolism , Hypothalamus/pathology , Liver/pathology , Male , Mice , Mice, Knockout , Sympathetic Nervous System/metabolism , Sympathetic Nervous System/pathology , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism
11.
J Physiol Biochem ; 75(1): 1-10, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30506389

ABSTRACT

Mammalian adipose tissue is traditionally categorized into white and brown relating to their function and morphology: while white serves as an energy storage, brown adipose tissue acts as the heat generator maintaining the core body temperature. The most recently identified type of fat, beige adipocyte tissue, resembles brown fat by morphology and function but is developmentally more related to white. The synthesis of beige fat, so-called browning of white fat, has developed into a topical issue in diabetes and metabolism research. This is due to its favorable effect on whole-body energy metabolism and the fact that it can be recruited during adult life. Indeed, brown and beige adipose tissues have been demonstrated to play a role in glucose homeostasis, insulin sensitivity, and lipid metabolism-all factors related to pathogenesis of type 2 diabetes. Many agents capable of initiating browning have been identified so far and tested widely in humans and animal models including in vitro and in vivo experiments. Interestingly, several agents demonstrated to have browning activity are in fact secreted as adipokines from brown and beige fat tissue, suggesting a physiological relevance both in beige adipocyte recruitment processes and in maintenance of metabolic homeostasis. The newest findings on agents driving beige fat recruitment, their mechanisms, and implications on type 2 diabetes are discussed in this review.


Subject(s)
Adipose Tissue, Beige/drug effects , Adipose Tissue, Brown/drug effects , Adipose Tissue, White/drug effects , Diabetes Mellitus, Type 2/drug therapy , Lipotropic Agents/pharmacology , Adipose Tissue, Beige/metabolism , Adipose Tissue, Beige/pathology , Adipose Tissue, Brown/metabolism , Adipose Tissue, Brown/pathology , Adipose Tissue, White/metabolism , Adipose Tissue, White/pathology , Animals , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Energy Metabolism/drug effects , Energy Metabolism/genetics , Glucagon-Like Peptide 1/pharmacology , Glucose/metabolism , Humans , Insulin Resistance , Leptin/pharmacology , Lipid Metabolism/drug effects , Lipid Metabolism/genetics , Melatonin/pharmacology , Natriuretic Peptides/pharmacology , Thermogenesis/drug effects , Thermogenesis/genetics , Tretinoin/pharmacology
12.
JCI Insight ; 3(15)2018 08 09.
Article in English | MEDLINE | ID: mdl-30089732

ABSTRACT

BACKGROUND: The induction of beige adipocytes in s.c. white adipose tissue (WAT) depots of humans is postulated to improve glucose and lipid metabolism in obesity. The ability of obese, insulin-resistant humans to induce beige adipose tissue is unknown. METHODS: We exposed lean and obese research participants to cold (30-minute ice pack application each day for 10 days of the upper thigh) or treated them with the ß3 agonist mirabegron. We determined beige adipose marker expression by IHC and quantitative PCR, and we analyzed mitochondrial bioenergetics and UCP activity with an Oxytherm system. RESULTS: Cold significantly induced UCP1 and TMEM26 protein in both lean and obese subjects, and this response was not associated with age. Interestingly, these proteins increased to the same extent in s.c. WAT of the noniced contralateral leg, indicating a crossover effect. We further analyzed the bioenergetics of purified mitochondria from the abdominal s.c. WAT of cold-treated subjects and determined that repeat ice application significantly increased uncoupled respiration, consistent with the UCP1 protein induction and subsequent activation. Cold also increased State 3 and maximal respiration, and this effect on mitochondrial bioenergetics was stronger in summer than winter. Chronic treatment (10 weeks; 50 mg/day) with the ß3 receptor agonist mirabegron induces UCP1, TMEM26, CIDEA, and phosphorylation of HSL on serine660 in obese subjects. CONCLUSION: Cold or ß3 agonists cause the induction of beige adipose tissue in human s.c. WAT; this phenomenon may be exploited to increase beige adipose in older, insulin-resistant, obese individuals. TRIAL REGISTRATION: Clinicaltrials.gov NCT02596776, NCT02919176. FUNDING: NIH (DK107646, DK112282, P20GM103527, and by CTSA grant UL1TR001998).


Subject(s)
Acetanilides/pharmacology , Adipose Tissue, Beige/metabolism , Adrenergic beta-3 Receptor Agonists/pharmacology , Obesity/metabolism , Subcutaneous Fat/metabolism , Thiazoles/pharmacology , Acetanilides/therapeutic use , Adipose Tissue, Beige/drug effects , Adipose Tissue, Beige/pathology , Adrenergic beta-3 Receptor Agonists/therapeutic use , Adult , Biopsy , Cold Temperature/adverse effects , Energy Metabolism/drug effects , Energy Metabolism/physiology , Female , Humans , Lipid Metabolism/drug effects , Lipid Metabolism/physiology , Male , Membrane Proteins/metabolism , Middle Aged , Obesity/drug therapy , Obesity/pathology , Subcutaneous Fat/drug effects , Subcutaneous Fat/pathology , Thermogenesis/physiology , Thiazoles/therapeutic use , Uncoupling Protein 1/metabolism
13.
Curr Diab Rep ; 18(10): 80, 2018 08 17.
Article in English | MEDLINE | ID: mdl-30120579

ABSTRACT

PURPOSE OF REVIEW: The global prevalence of type 2 diabetes (T2D) is escalating at alarming rates, demanding the development of additional classes of therapeutics to further reduce the burden of disease. Recent studies have indicated that increasing the metabolic activity of brown and beige adipose tissue may represent a novel means to reduce circulating glucose and lipids in people with T2D. The AMP-activated protein kinase (AMPK) is a cellular energy sensor that has recently been demonstrated to be important in potentially regulating the metabolic activity of brown and beige adipose tissue. The goal of this review is to summarize recent work describing the role of AMPK in brown and beige adipose tissue, focusing on its role in adipogenesis and non-shivering thermogenesis. RECENT FINDINGS: Ablation of AMPK in mouse adipocytes results in cold intolerance, a reduction in non-shivering thermogenesis in brown adipose tissue (BAT), and the development of non-alcoholic fatty liver disease (NAFLD) and insulin resistance; effects associated with a defect in mitochondrial specific autophagy (mitophagy) within BAT. The effects of a ß3-adrenergic agonist on the induction of BAT thermogenesis and the browning of white adipose tissue (WAT) are also blunted in mice lacking adipose tissue AMPK. A specific AMPK activator, A-769662, also results in the activation of BAT and the browning of WAT, effects which may involve demethylation of the PR domain containing 16 (Prdm16) promoter region, which is important for BAT development. AMPK plays an important role in the development and maintenance of brown and beige adipose tissue. Adipose tissue AMPK is reduced in people with insulin resistance, consistent with findings that mice lacking adipocyte AMPK develop greater NAFLD and insulin resistance. These data suggest that pharmacologically targeting adipose tissue AMPK may represent a promising strategy to enhance energy expenditure and reduce circulating glucose and lipids, which may be effective for the treatment of NAFLD and T2D.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Adipose Tissue, Beige/enzymology , Adipose Tissue, Brown/enzymology , Diabetes Mellitus, Type 2/enzymology , Insulin Resistance , Obesity/enzymology , Adipose Tissue, Beige/pathology , Adipose Tissue, Brown/pathology , Animals , Humans
14.
PLoS One ; 13(8): e0201661, 2018.
Article in English | MEDLINE | ID: mdl-30071087

ABSTRACT

BACKGROUND: Teff is a staple food in Ethiopia that is rich in dietary fiber. Although gaining popularity in Western countries because it is gluten-free, the effects of teff on glucose metabolism remain unknown. AIM: To evaluate the effects of teff on body weight and glucose metabolism compared with an isocaloric diet containing wheat. RESULTS: Mice fed teff weighed approximately 13% less than mice fed wheat (p < 0.05). The teff-based diet improved glucose tolerance compared with the wheat group with normal chow but not with a high-fat diet. Reduced adipose inflammation characterized by lower expression of TNFα, Mcp1, and CD11c, together with higher levels of cecal short chain fatty acids such as acetate, compared with the control diet containing wheat after 14 weeks of dietary treatment. In addition, beige adipocyte formation, characterized by increased expression of Ucp-1 (~7-fold) and Cidea (~3-fold), was observed in the teff groups compared with the wheat group. Moreover, a body-weight matched experiment revealed that teff improved glucose tolerance in a manner independent of body weight reduction after 6 weeks of dietary treatment. Enhanced beige adipocyte formation without improved adipose inflammation in a body-weight matched experiment suggests that the improved glucose metabolism was a consequence of beige adipocyte formation, but not solely through adipose inflammation. However, these differences between teff- and wheat-containing diets were not observed in the high-fat diet group. CONCLUSIONS: Teff improved glucose tolerance likely by promoting beige adipocyte formation and improved adipose inflammation.


Subject(s)
Adipose Tissue, Beige/metabolism , Carbohydrate Metabolism/drug effects , Dietary Fiber/pharmacology , Eragrostis/metabolism , Adipose Tissue, Beige/pathology , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Blood Glucose/analysis , Body Temperature , CD11c Antigen/genetics , CD11c Antigen/metabolism , Fatty Acids, Volatile/analysis , Fatty Acids, Volatile/chemistry , Feces/chemistry , Glucose Tolerance Test , Inflammation/metabolism , Inflammation/prevention & control , Male , Mice , Mice, Inbred C57BL , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism
15.
Ann Anat ; 219: 102-120, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30049662

ABSTRACT

Firstly identified by anatomists, the fat tissue is nowadays an area of intense research due to increased global prevalence of obesity and its associated diseases. Histologically, there are four types of fat tissue cells which are currently recognized (white, brown, beige, and perivascular adipocytes). Therefore, in this study we are reviewing the most recent data regarding the origin, structure, and molecular mechanisms involved in the development of adipocytes. White adipocytes can store triglycerides as a consequence of lipogenesis, under the regulation of growth hormone or leptin and adiponectin, and release fatty acids resulted from lipolysis, under the regulation of the sympathetic nervous system, glucocorticoids, TNF-α, insulin, and natriuretic peptides. Brown adipocytes possess a mitochondrial transmembrane protein thermogenin or UCP1 which allows heat generation. Recently, thermogenic, UCP positive adipocytes have been identified in the subcutaneous white adipose tissue and have been named beige adipocytes. The nature of these cells is still controversial, as current theories are suggesting their origin either by transdifferentiation of white adipocytes, or by differentiation from an own precursor cell. Perivascular adipocytes surround most of the arteries, exhibiting a supportive role and being involved in the maintenance of intravascular temperature. Thoracic perivascular adipocytes resemble brown adipocytes, while abdominal ones are more similar to white adipocytes and, consequently, are involved in obesity-induced inflammatory reactions. The factors involved in the regulation of adipose stem cells differentiation may represent potential pathways to inhibit or to divert adipogenesis. Several molecules, such as pro-adipogenic factors (FGF21, BMP7, BMP8b, and Cox-2), cell surface proteins or receptors (Asc-1, PAT2, P2RX5), and hypothalamic receptors (MC4R) have been identified as the most promising targets for the development of future therapies. Further investigations are necessary to complete the knowledge about adipose tissue and the development of a new generation of therapeutic tools based on molecular targets.


Subject(s)
Adipocytes/cytology , Adipose Tissue, Beige/pathology , Adipose Tissue, Brown/pathology , Adipose Tissue, White/pathology , Lipodystrophy/pathology , Obesity/pathology , Adipocytes/pathology , Adipocytes/ultrastructure , Adipogenesis/physiology , Adipose Tissue, Beige/metabolism , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Humans
16.
J Intern Med ; 284(5): 492-504, 2018 11.
Article in English | MEDLINE | ID: mdl-29923291

ABSTRACT

Many of the comorbidities of obesity, including type 2 diabetes and cardiovascular diseases, are related to the low-grade chronic inflammation of white adipose tissue. Under white adipocyte stress, local infiltration of immune cells and enhanced production of pro-inflammatory cytokines together reduce metabolic flexibility and lead to insulin resistance in obesity. Whereas white adipocytes act in energy storage, brown and beige adipocytes specialize in energy expenditure. Brown and beige activity protects against obesity and associated metabolic disorders, such as hyperglycaemia and hyperlipidaemia. Compared to white fat, brown adipose tissue depots are less susceptible to developing local inflammation in response to obesity; however, strong obesogenic insults ultimately induce a locally pro-inflammatory environment in brown fat. This condition directly alters the thermogenic activity of brown fat by impairing its energy expenditure mechanism and uptake of glucose for use as a fuel substrate. Pro-inflammatory cytokines also impair beige adipogenesis, which occurs mainly in subcutaneous adipose tissue. There is evidence that inflammatory processes occurring in perivascular adipose tissues alter their brown-versus-white plasticity, impair the extent of browning in these depots and favour the local release of vasculature damaging signals. In summary, the targeting of brown and beige adipose tissues by pro-inflammatory signals and the subsequent impairment of their thermogenic and metabolite draining activities appears to represent obesity-driven disturbances that contribute to metabolic syndrome and cardiovascular alterations in obesity.


Subject(s)
Adipose Tissue, Beige/pathology , Adipose Tissue, Brown/pathology , Inflammation/pathology , Metabolic Diseases/pathology , Obesity/pathology , Animals , Humans , Inflammation/etiology , Metabolic Diseases/complications , Obesity/complications
17.
Diabetes ; 67(2): 169-179, 2018 02.
Article in English | MEDLINE | ID: mdl-29358486

ABSTRACT

The Outstanding Scientific Achievement Award recognizes distinguished scientific achievement in the field of diabetes, taking into consideration independence of thought and originality. Gregory R. Steinberg, PhD, professor of medicine, Canada Research Chair, J. Bruce Duncan Endowed Chair in Metabolic Diseases, and codirector of the Metabolism and Childhood Obesity Research Program at McMaster University, Hamilton, Ontario, Canada, received the prestigious award at the American Diabetes Association's 77th Scientific Sessions, 9-13 June 2017, in San Diego, CA. He presented the Outstanding Scientific Achievement Award Lecture, "Cellular Energy Sensing and Metabolism-Implications for Treating Diabetes," on Monday, 12 June 2017.The survival of all cells is dependent on the constant challenge to match energetic demands with nutrient availability, a task that is mediated through a highly conserved network of metabolic fuel sensors that orchestrate both cellular and whole-organism energy balance. A mismatch between cellular energy demand and nutrient availability is a key factor contributing to the development of type 2 diabetes; thus, understanding the fundamental mechanisms by which cells sense nutrient availability and demand may lead to the development of new treatments. Glucose-lowering therapies, such as caloric restriction, exercise, and metformin, all induce an energetic challenge that results in the activation of the cellular energy sensor AMP-activated protein kinase (AMPK). Activation of AMPK in turn suppresses lipid synthesis and inflammation while increasing glucose uptake, fatty acid oxidation, and mitochondrial function. In contrast, high levels of nutrient availability suppress AMPK activity while also increasing the production of peripheral serotonin, a gut-derived endocrine factor that suppresses ß-adrenergic-induced activation of brown adipose tissue. Identifying new ways to manipulate these two ancient fuel gauges by activating AMPK and inhibiting peripheral serotonin may lead to the development of new therapies for treating type 2 diabetes.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Diabetes Mellitus, Type 2/metabolism , Energy Metabolism , Feedback, Physiological , Insulin Resistance , Models, Biological , Serotonin/metabolism , Adipose Tissue, Beige/drug effects , Adipose Tissue, Beige/metabolism , Adipose Tissue, Beige/pathology , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/metabolism , Adipose Tissue, Brown/pathology , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Adipose Tissue, White/pathology , Animals , Awards and Prizes , Caloric Restriction , Cell Survival/drug effects , Combined Modality Therapy , Diabetes Mellitus, Type 2/pathology , Diabetes Mellitus, Type 2/prevention & control , Diabetes Mellitus, Type 2/therapy , Endocrinology , Energy Intake/drug effects , Energy Metabolism/drug effects , Enzyme Activation/drug effects , Exercise , Feedback, Physiological/drug effects , Humans , Hypoglycemic Agents/therapeutic use , Liver/drug effects , Liver/metabolism , Liver/pathology , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Serotonin/blood
18.
Endocrinology ; 159(1): 356-367, 2018 01 01.
Article in English | MEDLINE | ID: mdl-28973559

ABSTRACT

Laminin α4 (LAMA4) is located in the extracellular basement membrane that surrounds each individual adipocyte. Here we show that LAMA4 null (Lama4-/-) mice exhibit significantly higher energy expenditure (EE) relative to wild-type (WT) mice at room temperature and when exposed to a cold challenge, despite similar levels of food intake and locomotor activity. The Lama4-/- mice are resistant to age- and diet-induced obesity. Expression of uncoupling protein 1 is higher in subcutaneous white adipose tissue of Lama4-/- mice relative to WT animals on either a chow diet or a high-fat diet. In contrast, uncoupling protein 1 expression was not increased in brown adipose tissue. Lama4-/- mice exhibit significantly improved insulin sensitivity compared with WT mice, suggesting improved metabolic function. Overall, these data provide critical evidence for a role of the basement membrane in EE, weight gain, and systemic insulin sensitivity.


Subject(s)
Adipose Tissue, Beige/metabolism , Adiposity , Energy Metabolism , Insulin Resistance , Laminin/metabolism , Obesity/metabolism , Subcutaneous Fat/metabolism , Adipose Tissue, Beige/pathology , Adipose Tissue, Brown/metabolism , Adipose Tissue, Brown/pathology , Adult Stem Cells/metabolism , Adult Stem Cells/pathology , Animals , Basement Membrane/metabolism , Basement Membrane/pathology , Cells, Cultured , Cold Temperature/adverse effects , Diet, High-Fat/adverse effects , Energy Intake , Gene Expression Regulation, Developmental , Laminin/genetics , Male , Mice, Knockout , Obesity/etiology , Obesity/pathology , Organ Specificity , Stress, Physiological , Subcutaneous Fat/pathology , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism , Weight Gain
19.
Endocrinology ; 159(1): 323-340, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29040448

ABSTRACT

It is unknown how the lack of insulin receptor (IR)/insulinlike growth factor I receptor (IGFIR) in a tissue-specific manner affects brown fat development and mitochondrial integrity and function, as well as its effect on the redistribution of the adipose organ and the metabolic status. To address this important issue, we developed IR/IGFIR double-knockout (DKO) in a brown adipose tissue-specific manner. Lack of those receptors caused severe brown fat atrophy, enhanced beige cell clusters in inguinal fat; loss of mitochondrial mass; mitochondrial damage related to cristae disruption; and the loss of proteins involved in autophagosome formation, mitophagy, mitochondrial quality control, and dynamics and thermogenesis. More important, DKO mice showed an impaired thermogenesis upon cold exposure, based on a failure in the mitochondrial fission mechanisms and a much lower uncoupling protein 1 transcription rate and content. As a result, DKO mice under normal conditions showed an obesity susceptibility, revealed by increased body fat mass and insulin resistance. Upon consumption of a high-fat diet, DKO mice displayed frank obesity, as shown by increased body weight, increased adiposity, insulin resistance, hyperinsulinemia, and hypertriglyceridemia, all consistent with a metabolic syndrome. Collectively, our data suggest a cause-and-effect relationship between failure in brown fat thermogenesis and increased adiposity and obesity.


Subject(s)
Adipose Tissue, Brown/metabolism , Metabolic Syndrome/metabolism , Mitochondrial Dynamics , Obesity/metabolism , Receptor, IGF Type 1/metabolism , Receptor, Insulin/metabolism , Thermogenesis , Adipose Tissue, Beige/metabolism , Adipose Tissue, Beige/pathology , Adipose Tissue, Beige/ultrastructure , Adipose Tissue, Brown/pathology , Adipose Tissue, Brown/ultrastructure , Adiposity , Animals , Atrophy , Diet, High-Fat/adverse effects , Hyperinsulinism/etiology , Hypertriglyceridemia/etiology , Insulin Resistance , Male , Metabolic Syndrome/etiology , Metabolic Syndrome/pathology , Metabolic Syndrome/physiopathology , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission , Mitochondria/metabolism , Mitochondria/pathology , Mitochondria/ultrastructure , Obesity/etiology , Obesity/pathology , Obesity/physiopathology , Organ Specificity , Receptor, IGF Type 1/genetics , Receptor, Insulin/genetics , Weight Gain
20.
J Nutr Biochem ; 49: 123-132, 2017 11.
Article in English | MEDLINE | ID: mdl-28945993

ABSTRACT

Caloric restriction (CR) is one of the most promising strategies for weight loss but is associated with loss of lean mass, whereas compounds such as trans-10,cis-12 conjugated linoleic acid (t10-c12 CLA) have been promoted as antiobesity agents. To compare the mechanisms of weight reduction by CR and t10-c12 CLA, body composition, glucose control, and characteristics of adipose tissue with respect to cell turnover (stem cells and preadipocytes, apoptosis and autophagy) and Tbx-1 localization were examined in obese db/db mice and lean C57BL/6J mice undergoing CR or fed CLA isomers (0.4% w/w c9-t11 or t10-c12) for 4 weeks. Our findings show that the t10-c12 CLA reduced whole-body fat mass by decreasing all fat depots (visceral, inguinal, brown/interscapular), while CR lowered both whole-body fat and lean mass in obese mice. t10-c12 CLA elevated blood glucose in both obese and lean mice, while glycemia was not altered by CR. The adipocyte stem cell population remained unchanged; however, t10-c12 CLA reduced and CR elevated the proportion of immature adipocytes in obese mice, suggesting differential effects on adipocyte maturation. t10-c12 CLA reduced apoptosis (activated caspase-3) in both obese and lean mice but did not alter autophagy (LC3II/LC3I). Nuclear Tbx-1, a marker of metabolically active beige adipocytes, was greater in the adipose of t10-c12 CLA-fed animals. Thus, weight loss achieved via t10-c12 CLA primarily involves fat loss and more cells with Tbx-1 localized to the nucleus, while CR operates through a mechanism that reduces both lean and fat mass and blocks adipocyte differentiation.


Subject(s)
Adipogenesis , Adiposity , Anti-Obesity Agents/therapeutic use , Caloric Restriction , Dietary Supplements , Linoleic Acids, Conjugated/therapeutic use , Obesity/diet therapy , Adipose Tissue, Beige/metabolism , Adipose Tissue, Beige/pathology , Adipose Tissue, Brown/metabolism , Adipose Tissue, Brown/pathology , Adipose Tissue, White/metabolism , Adipose Tissue, White/pathology , Adult Stem Cells/metabolism , Adult Stem Cells/pathology , Animals , Anti-Obesity Agents/adverse effects , Apoptosis , Biomarkers/metabolism , Caloric Restriction/adverse effects , Dietary Supplements/adverse effects , Linoleic Acids, Conjugated/adverse effects , Male , Mice, Inbred C57BL , Mice, Mutant Strains , Obesity/metabolism , Obesity/pathology , Random Allocation , T-Box Domain Proteins/metabolism , Weight Loss
SELECTION OF CITATIONS
SEARCH DETAIL
...