Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.745
Filter
2.
Sci Rep ; 14(1): 10789, 2024 05 11.
Article in English | MEDLINE | ID: mdl-38734719

ABSTRACT

Brown adipocytes are potential therapeutic targets for the prevention of obesity-associated metabolic diseases because they consume circulating glucose and fatty acids for heat production. Angiotensin II (Ang II) peptide is involved in the pathogenesis of obesity- and cold-induced hypertension; however, the mechanism underlying the direct effects of Ang II on human brown adipocytes remains unclear. Our transcriptome analysis of chemical compound-induced brown adipocytes (ciBAs) showed that the Ang II type 1 receptor (AGTR1), but not AGTR2 and MAS1 receptors, was expressed. The Ang II/AGTR1 axis downregulated the expression of mitochondrial uncoupling protein 1 (UCP1). The simultaneous treatment with ß-adrenergic receptor agonists and Ang II attenuated UCP1 expression, triglyceride lipolysis, and cAMP levels, although cAMP response element-binding protein (CREB) phosphorylation was enhanced by Ang II mainly through the protein kinase C pathway. Despite reduced lipolysis, both coupled and uncoupled mitochondrial respiration was enhanced in Ang II-treated ciBAs. Instead, glycolysis and glucose uptake were robustly activated upon treatment with Ang II without a comprehensive transcriptional change in glucose metabolic genes. Elevated mitochondrial energy status induced by Ang II was likely associated with UCP1 repression. Our findings suggest that the Ang II/AGTR1 axis participates in mitochondrial thermogenic functions via glycolysis.


Subject(s)
Adipocytes, Brown , Angiotensin II , Glycolysis , Mitochondria , Thermogenesis , Uncoupling Protein 1 , Humans , Adipocytes, Brown/metabolism , Adipocytes, Brown/drug effects , Glycolysis/drug effects , Angiotensin II/pharmacology , Angiotensin II/metabolism , Mitochondria/metabolism , Mitochondria/drug effects , Thermogenesis/drug effects , Uncoupling Protein 1/metabolism , Uncoupling Protein 1/genetics , Lipolysis/drug effects , Receptor, Angiotensin, Type 1/metabolism , Receptor, Angiotensin, Type 1/genetics , Glucose/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism
3.
Commun Biol ; 7(1): 632, 2024 May 25.
Article in English | MEDLINE | ID: mdl-38796563

ABSTRACT

The stomach-derived hormone ghrelin regulates essential physiological functions. The ghrelin receptor (GHSR) has ligand-independent actions; therefore, GHSR gene deletion may be a reasonable approach to investigate the role of this system in feeding behaviors and diet-induced obesity (DIO). Here, we investigate the effects of a long-term (12-month) high-fat (HFD) versus regular diet on obesity-related measures in global GHSR-KO and wild-type (WT) Wistar male and female rats. Our main findings are that the GHSR gene deletion protects against DIO and decreases food intake during HFD in male but not in female rats. GHSR gene deletion increases thermogenesis and brain glucose uptake in male rats and modifies the effects of HFD on brain glucose metabolism in a sex-specific manner, as assessed with small animal positron emission tomography. We use RNA-sequencing to show that GHSR-KO rats have upregulated expression of genes responsible for fat oxidation in brown adipose tissue. Central administration of a novel GHSR inverse agonist, PF-5190457, attenuates ghrelin-induced food intake, but only in male, not in female mice. HFD-induced binge-like eating is reduced by inverse agonism in both sexes. Our results support GHSR as a promising target for new pharmacotherapies for obesity.


Subject(s)
Diet, High-Fat , Obesity , Rats, Wistar , Receptors, Ghrelin , Sex Characteristics , Animals , Receptors, Ghrelin/genetics , Receptors, Ghrelin/metabolism , Diet, High-Fat/adverse effects , Male , Female , Rats , Obesity/metabolism , Obesity/genetics , Ghrelin/metabolism , Thermogenesis/drug effects , Eating/drug effects , Adipose Tissue, Brown/metabolism , Adipose Tissue, Brown/drug effects
4.
Sci Rep ; 14(1): 12430, 2024 05 30.
Article in English | MEDLINE | ID: mdl-38816541

ABSTRACT

Dietary trans 10, cis 12-conjugated linoleic acid (t10c12-CLA) is a potential candidate in anti-obesity trials. A transgenic mouse was previously successfully established to determine the anti-obesity properties of t10c12-CLA in male mice that could produce endogenous t10c12-CLA. To test whether there is a different impact of t10c12-CLA on lipid metabolism in both sexes, this study investigated the adiposity and metabolic profiles of female Pai mice that exhibited a dose-dependent expression of foreign Pai gene and a shift of t10c12-CLA content in tested tissues. Compared to their gender-match wild-type littermates, Pai mice had no fat reduction but exhibited enhanced lipolysis and thermogenesis by phosphorylated hormone-sensitive lipase and up-regulating uncoupling proteins in brown adipose tissue. Simultaneously, Pai mice showed hepatic steatosis and hypertriglyceridemia by decreasing gene expression involved in lipid and glucose metabolism. Further investigations revealed that t10c10-CLA induced excessive prostaglandin E2, adrenaline, corticosterone, glucagon and inflammatory factors in a dose-dependent manner, resulting in less heat release and oxygen consumption in Pai mice. Moreover, fibroblast growth factor 21 overproduction only in monoallelic Pai/wt mice indicates that it was sensitive to low doses of t10c12-CLA. These results suggest that chronic t10c12-CLA has system-wide effects on female health via synergistic actions of various hormones.


Subject(s)
Corticosterone , Dinoprostone , Epinephrine , Fibroblast Growth Factors , Glucagon , Linoleic Acids, Conjugated , Mice, Transgenic , Animals , Female , Fibroblast Growth Factors/metabolism , Fibroblast Growth Factors/genetics , Mice , Linoleic Acids, Conjugated/pharmacology , Linoleic Acids, Conjugated/metabolism , Corticosterone/metabolism , Dinoprostone/metabolism , Glucagon/metabolism , Epinephrine/metabolism , Thermogenesis/drug effects , Thermogenesis/genetics , Male , Lipid Metabolism/drug effects , Adipose Tissue, Brown/metabolism , Adipose Tissue, Brown/drug effects , Fatty Liver/metabolism , Fatty Liver/genetics , Lipolysis/drug effects , Hypertriglyceridemia/metabolism , Hypertriglyceridemia/genetics , Adiposity/drug effects
5.
Cell Metab ; 36(5): 891-892, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38718756

ABSTRACT

Brown adipose tissue has long been functionally characterized as an organ that regulates thermogenesis, body weight set point, and glucose homeostasis. In the May 9, 2024, issue of Cell, Verkerke et al. discover a novel function for brown adipose tissue in processing branched-chain amino acids into antioxidant metabolites that enter the circulation and regulate insulin signaling in the liver.


Subject(s)
Adipocytes, Brown , Adipocytes, Brown/metabolism , Animals , Humans , Adipose Tissue, Brown/metabolism , Thermogenesis , Amino Acids, Branched-Chain/metabolism , Insulin/metabolism , Signal Transduction , Liver/metabolism
6.
Nat Commun ; 15(1): 3473, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38724563

ABSTRACT

Neuronal differentiation-the development of neurons from neural stem cells-involves neurite outgrowth and is a key process during the development and regeneration of neural functions. In addition to various chemical signaling mechanisms, it has been suggested that thermal stimuli induce neuronal differentiation. However, the function of physiological subcellular thermogenesis during neuronal differentiation remains unknown. Here we create methods to manipulate and observe local intracellular temperature, and investigate the effects of noninvasive temperature changes on neuronal differentiation using neuron-like PC12 cells. Using quantitative heating with an infrared laser, we find an increase in local temperature (especially in the nucleus) facilitates neurite outgrowth. Intracellular thermometry reveals that neuronal differentiation is accompanied by intracellular thermogenesis associated with transcription and translation. Suppression of intracellular temperature increase during neuronal differentiation inhibits neurite outgrowth. Furthermore, spontaneous intracellular temperature elevation is involved in neurite outgrowth of primary mouse cortical neurons. These results offer a model for understanding neuronal differentiation induced by intracellular thermal signaling.


Subject(s)
Cell Differentiation , Neurons , Signal Transduction , Temperature , Animals , PC12 Cells , Neurons/physiology , Neurons/cytology , Mice , Rats , Neuronal Outgrowth , Neurogenesis/physiology , Neurites/metabolism , Neurites/physiology , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neural Stem Cells/physiology , Thermometry/methods , Thermogenesis/physiology
7.
Int J Mol Sci ; 25(9)2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38732127

ABSTRACT

The process of adipocyte browning has recently emerged as a novel therapeutic target for combating obesity and obesity-related diseases. Non-shivering thermogenesis is the process of biological heat production in mammals and is primarily mediated via brown adipose tissue (BAT). The recruitment and activation of BAT can be induced through chemical drugs and nutrients, with subsequent beneficial health effects through the utilization of carbohydrates and fats to generate heat to maintain body temperature. However, since potent drugs may show adverse side effects, nutritional or natural substances could be safe and effective as potential adipocyte browning agents. This review aims to provide an extensive overview of the natural food compounds that have been shown to activate brown adipocytes in humans, animals, and in cultured cells. In addition, some key genetic and molecular targets and the mechanisms of action of these natural compounds reported to have therapeutic potential to combat obesity are discussed.


Subject(s)
Adipose Tissue, Brown , Biological Products , Obesity , Thermogenesis , Thermogenesis/drug effects , Humans , Animals , Adipose Tissue, Brown/metabolism , Adipose Tissue, Brown/drug effects , Biological Products/pharmacology , Biological Products/therapeutic use , Obesity/drug therapy , Obesity/metabolism , Adipocytes, Brown/metabolism , Adipocytes, Brown/drug effects
8.
Front Endocrinol (Lausanne) ; 15: 1385811, 2024.
Article in English | MEDLINE | ID: mdl-38765953

ABSTRACT

Background: Thermogenic beige adipocytes, which dissipate energy as heat, are found in neonates and adults. Recent studies show that neonatal beige adipocytes are highly plastic and contribute to >50% of beige adipocytes in adults. Neonatal beige adipocytes are distinct from recruited beige adipocytes in that they develop independently of temperature and sympathetic innervation through poorly defined mechanisms. Methods: We characterized the neonatal beige adipocytes in the inguinal white adipose tissue (iWAT) of C57BL6 postnatal day 3 and 20 mice (P3 and P20) by imaging, genome-wide RNA-seq analysis, ChIP-seq analysis, qRT-PCR validation, and biochemical assays. Results: We found an increase in acetylated histone 3 lysine 27 (H3K27ac) on the promoter and enhancer regions of beige-specific gene UCP1 in iWAT of P20 mice. Furthermore, H3K27ac ChIP-seq analysis in the iWAT of P3 and P20 mice revealed strong H3K27ac signals at beige adipocyte-associated genes in the iWAT of P20 mice. The integration of H3K27ac ChIP-seq and RNA-seq analysis in the iWAT of P20 mice reveal epigenetically active signatures of beige adipocytes, including oxidative phosphorylation and mitochondrial metabolism. We identify the enrichment of GA-binding protein alpha (GABPα) binding regions in the epigenetically active chromatin regions of the P20 iWAT, particularly on beige genes, and demonstrate that GABPα is required for beige adipocyte differentiation. Moreover, transcriptomic analysis and glucose oxidation assays revealed increased glycolytic activity in the neonatal iWAT from P20. Conclusions: Our findings demonstrate that epigenetic mechanisms regulate the development of peri-weaning beige adipocytes via GABPα. Further studies to better understand the upstream mechanisms that regulate epigenetic activation of GABPα and characterization of the metabolic identity of neonatal beige adipocytes will help us harness their therapeutic potential in metabolic diseases.


Subject(s)
Adipocytes, Beige , Adipogenesis , Adipose Tissue, White , Animals, Newborn , Chromatin , Epigenesis, Genetic , GA-Binding Protein Transcription Factor , Mice, Inbred C57BL , Animals , Mice , Adipocytes, Beige/metabolism , Chromatin/metabolism , Chromatin/genetics , Adipogenesis/genetics , Adipose Tissue, White/metabolism , GA-Binding Protein Transcription Factor/metabolism , GA-Binding Protein Transcription Factor/genetics , Male , Thermogenesis/genetics , Histones/metabolism , Histones/genetics
9.
Mol Metab ; 84: 101951, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38729241

ABSTRACT

OBJECTIVE: Hypothalamic signals potently stimulate energy expenditure by engaging peripheral mechanisms to restore energy homeostasis. Previous studies have identified several critical hypothalamic sites (e.g. preoptic area (POA) and ventromedial hypothalamic nucleus (VMN)) that could be part of an interconnected neurocircuit that controls tissue thermogenesis and essential for body weight control. However, the key neurocircuit that can stimulate energy expenditure has not yet been established. METHODS: Here, we investigated the downstream mechanisms by which VMN neurons stimulate adipose tissue thermogenesis. We manipulated subsets of VMN neurons acutely as well as chronically and studied its effect on tissue thermogenesis and body weight control, using Sf1Cre and Adcyap1Cre mice and measured physiological parameters under both high-fat diet and standard chow diet conditions. To determine the node efferent to these VMN neurons, that is involved in modulating energy expenditure, we employed electrophysiology and optogenetics experiments combined with measurements using tissue-implantable temperature microchips. RESULTS: Activation of the VMN neurons that express the steroidogenic factor 1 (Sf1; VMNSf1 neurons) reduced body weight, adiposity and increased energy expenditure in diet-induced obese mice. This function is likely mediated, at least in part, by the release of the pituitary adenylate cyclase-activating polypeptide (PACAP; encoded by the Adcyap1 gene) by the VMN neurons, since we previously demonstrated that PACAP, at the VMN, plays a key role in energy expenditure control. Thus, we then shifted focus to the subpopulation of VMNSf1 neurons that contain the neuropeptide PACAP (VMNPACAP neurons). Since the VMN neurons do not directly project to the peripheral tissues, we traced the location of the VMNPACAP neurons' efferents. We identified that VMNPACAP neurons project to and activate neurons in the caudal regions of the POA whereby these projections stimulate tissue thermogenesis in brown and beige adipose tissue. We demonstrated that selective activation of caudal POA projections from VMNPACAP neurons induces tissue thermogenesis, most potently in negative energy balance and activating these projections lead to some similar, but mostly unique, patterns of gene expression in brown and beige tissue. Finally, we demonstrated that the activation of the VMNPACAP neurons' efferents that lie at the caudal POA are necessary for inducing tissue thermogenesis in brown and beige adipose tissue. CONCLUSIONS: These data indicate that VMNPACAP connections with the caudal POA neurons impact adipose tissue function and are important for induction of tissue thermogenesis. Our data suggests that the VMNPACAP → caudal POA neurocircuit and its components are critical for controlling energy balance by activating energy expenditure and body weight control.


Subject(s)
Energy Metabolism , Neurons , Preoptic Area , Thermogenesis , Ventromedial Hypothalamic Nucleus , Animals , Ventromedial Hypothalamic Nucleus/metabolism , Thermogenesis/physiology , Preoptic Area/metabolism , Mice , Neurons/metabolism , Male , Steroidogenic Factor 1/metabolism , Steroidogenic Factor 1/genetics , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Pituitary Adenylate Cyclase-Activating Polypeptide/genetics , Diet, High-Fat , Mice, Inbred C57BL , Body Weight , Adipose Tissue, Brown/metabolism
10.
Front Immunol ; 15: 1375138, 2024.
Article in English | MEDLINE | ID: mdl-38812501

ABSTRACT

Objectives: The effects of cold exposure on whole-body metabolism in humans have gained increasing attention. Brown or beige adipose tissues are crucial in cold-induced thermogenesis to dissipate energy and thus have the potential to combat metabolic disorders. Despite the immune regulation of thermogenic adipose tissues, the overall changes in vital immune cells during distinct cold periods remain elusive. This study aimed to discuss the overall changes in immune cells under different cold exposure periods and to screen several potential immune cell subpopulations on thermogenic regulation. Methods: Cibersort and mMCP-counter algorithms were employed to analyze immune infiltration in two (brown and beige) thermogenic adipose tissues under distinct cold periods. Changes in some crucial immune cell populations were validated by reanalyzing the single-cell sequencing dataset (GSE207706). Flow cytometry, immunofluorescence, and quantitative real-time PCR assays were performed to detect the proportion or expression changes in mouse immune cells of thermogenic adipose tissues under cold challenge. Results: The proportion of monocytes, naïve, and memory T cells increased, while the proportion of NK cells decreased under cold exposure in brown adipose tissues. Conclusion: Our study revealed dynamic changes in immune cell profiles in thermogenic adipose tissues and identified several novel immune cell subpopulations, which may contribute to thermogenic activation of adipose tissues under cold exposure.


Subject(s)
Adipose Tissue, Brown , Cold Temperature , Thermogenesis , Thermogenesis/immunology , Animals , Mice , Adipose Tissue, Brown/immunology , Adipose Tissue, Brown/metabolism , Mice, Inbred C57BL , Male , Adipose Tissue, Beige/metabolism , Adipose Tissue, Beige/immunology , Adipose Tissue/immunology , Adipose Tissue/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Monocytes/immunology , Monocytes/metabolism
11.
Cell Rep ; 43(5): 114142, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38691458

ABSTRACT

Despite medical advances, there remains an unmet need for better treatment of obesity. Itaconate, a product of the decarboxylation of the tricarboxylic acid cycle intermediate cis-aconitate, plays a regulatory role in both metabolism and immunity. Here, we show that itaconate, as an endogenous compound, counteracts high-fat-diet (HFD)-induced obesity through leptin-independent mechanisms in three mouse models. Specifically, itaconate reduces weight gain, reverses hyperlipidemia, and improves glucose tolerance in HFD-fed mice. Additionally, itaconate enhances energy expenditure and the thermogenic capacity of brown adipose tissue (BAT). Unbiased proteomic analysis reveals that itaconate upregulates key proteins involved in fatty acid oxidation and represses the expression of lipogenic genes. Itaconate may provoke a major metabolic reprogramming by inducing fatty acid oxidation and suppression of fatty acid synthesis in BAT. These findings highlight itaconate as a potential activator of BAT-mediated thermogenesis and a promising candidate for anti-obesity therapy.


Subject(s)
Adipocytes, Brown , Diet, High-Fat , Mice, Inbred C57BL , Obesity , Succinates , Thermogenesis , Animals , Thermogenesis/drug effects , Obesity/metabolism , Obesity/drug therapy , Succinates/pharmacology , Diet, High-Fat/adverse effects , Mice , Male , Adipocytes, Brown/metabolism , Adipocytes, Brown/drug effects , Adipose Tissue, Brown/metabolism , Adipose Tissue, Brown/drug effects , Energy Metabolism/drug effects
12.
Food Funct ; 15(8): 4627-4641, 2024 Apr 22.
Article in English | MEDLINE | ID: mdl-38592736

ABSTRACT

Diet-induced thermogenesis (DIT) is crucial for maintaining body weight homeostasis, and the role of dietary fatty acids in modulating DIT is essential. However, the underlying mechanism of fatty acid regulated diet-induced thermogenesis remains elusive. Utilizing the diet- and genetic ablation-induced obese mice models, we found that the C16 unsaturated fatty acids, trans-palmitoleic acid (TPA) and cis-palmitoleic acid (CPA), significantly increased the energy expenditure by promoting the thermogenesis of brown adipose tissues and the production of beige cells in white adipose. As a result, there is a significant reduction in the occurrence of obesity, associated hepatic steatosis and hyperglycemia. Notably, TPA exhibited more potent effects on promoting DIT and alleviating obesity than CPA did. Using inhibitor and gene deletion mice models, we unveiled that TPA acted as a signaling molecule to play a biological function, which could be sensed by the hypothalamic FFAR1 to activate the sympathetic nervous system in promoting adipose tissue thermogenesis. Together, these results demonstrate the underlying mechanism of free fatty acids associated-DIT and will provide fresh insights into the roles of trans-fatty acids in the development of obesity.


Subject(s)
Fatty Acids, Monounsaturated , Hypothalamus , Mice, Inbred C57BL , Obesity , Receptors, G-Protein-Coupled , Signal Transduction , Thermogenesis , Animals , Thermogenesis/drug effects , Mice , Obesity/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/genetics , Fatty Acids, Monounsaturated/pharmacology , Hypothalamus/metabolism , Hypothalamus/drug effects , Male , Signal Transduction/drug effects , Energy Metabolism/drug effects , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Adipose Tissue, White/drug effects , Diet, High-Fat
13.
Life Sci ; 345: 122607, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38583857

ABSTRACT

Diabetes mellitus is a disorder characterised metabolic dysfunction that results in elevated glucose level in the bloodstream. Diabetes is of two types, type1 and type 2 diabetes. Obesity is considered as one of the major reasons intended for incidence of diabetes hence it turns out to be essential to study about the adipose tissue which is responsible for fat storage in body. Adipose tissues play significant role in maintaining the balance between energy stabilization and homeostasis. The three forms of adipose tissue are - White adipose tissue (WAT), Brown adipose tissue (BAT) and Beige adipose tissue (intermediate form). The amount of BAT gets reduced, and WAT starts to increase with the age. WAT when exposed to certain stimuli gets converted to BAT by the help of certain transcriptional regulators. The browning of WAT has been a matter of study to treat the metabolic disorders and to initiate the expenditure of energy. The three main regulators responsible for the browning of WAT are PRDM16, PPARγ and PGC-1α via various cellular and molecular mechanism. Presented review article includes the detailed elaborative aspect of genes and proteins involved in conversion of WAT to BAT.


Subject(s)
Adipose Tissue, Brown , Diabetes Mellitus, Type 2 , Humans , Adipose Tissue, Brown/metabolism , Diabetes Mellitus, Type 2/metabolism , Obesity/metabolism , Adiposity , Transcription Factors/metabolism , Adipose Tissue, White/metabolism , Thermogenesis/genetics
14.
Nan Fang Yi Ke Da Xue Xue Bao ; 44(3): 499-506, 2024 Mar 20.
Article in Chinese | MEDLINE | ID: mdl-38597441

ABSTRACT

OBJECTIVE: To investigate the effects of α7 nicotinic acetylcholine receptor (nAChR) agonist on ß3-adrenoceptor agonist-induced impairment of white fat homeostasis and beige adipose formation and heat production in obese mice. METHODS: Forty obese C57BL/6J mice were randomized into high-fat feeding group, ß3-adrenoceptor agonist-treated model group, α7 nAChR agonist group, and α7 nAChR inhibitor group (n=10), with another 10 mice with normal feeding as the blank control group. White adipose tissue from the epididymis of the mice were sampled for HE staining of the adipocytes. The expression levels of TNF-α, IL-1ß, IL-10 and TGF-ß in the white adipose tissue were determined by ELISA, and the mRNA levels of iNOS, Arg1, UCP-1, PRDM-16 and PGC-1α were detected using RT-qPCR. Western blotting was performed to detect the expression levels of NF-κB P65, p-JAK2, p-STAT3 in the white adipose tissue. RESULTS: Compared with those in the blank control group, the mice with high-fat feeding showed significantly increased body weight, more fat vacuoles in the white adipose tissue, increased volume of lipid droplets in the adipocytes, upregulated iNOS mRNA expression and protein expression of TNF-α and IL-1ß, and lowered expression of Arg-1 mRNA and IL-10 and TGF-ß proteins (P < 0.01). Treatment with α7 nAChR significantly reduced mRNA levels of PRDM-16, PGC-1α and UCP-1, lowered TNF-α and IL-1ß expressions, increased IL-10 and TGF-ß expressions, and reduced M1/M2 macrophage ratio in the white adipose tissues (P < 0.05 or 0.01). CONCLUSION: Activation of α7 nAchR improves white adipose tissue homeostasis impairment induced by ß3 agonist, promotes transformation of M1 to M2 macrophages, reduces inflammatory response in white adipose tissue, and promote beige adipogenesis and thermogenesis in obese mice.


Subject(s)
Interleukin-10 , alpha7 Nicotinic Acetylcholine Receptor , Animals , Male , Mice , Adipogenesis , Adipose Tissue, White/metabolism , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Homeostasis , Mice, Inbred C57BL , Mice, Obese , Receptors, Adrenergic/metabolism , RNA, Messenger/metabolism , Thermogenesis , Transforming Growth Factor beta/metabolism , Tumor Necrosis Factor-alpha/metabolism
15.
Proc Natl Acad Sci U S A ; 121(16): e2318935121, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38588421

ABSTRACT

Glucose is required for generating heat during cold-induced nonshivering thermogenesis in adipose tissue, but the regulatory mechanism is largely unknown. CREBZF has emerged as a critical mechanism for metabolic dysfunction-associated steatotic liver disease (MASLD), formerly known as nonalcoholic fatty liver disease (NAFLD). We investigated the roles of CREBZF in the control of thermogenesis and energy metabolism. Glucose induces CREBZF in human white adipose tissue (WAT) and inguinal WAT (iWAT) in mice. Lys208 acetylation modulated by transacetylase CREB-binding protein/p300 and deacetylase HDAC3 is required for glucose-induced reduction of proteasomal degradation and augmentation of protein stability of CREBZF. Glucose induces rectal temperature and thermogenesis in white adipose of control mice, which is further potentiated in adipose-specific CREBZF knockout (CREBZF FKO) mice. During cold exposure, CREBZF FKO mice display enhanced thermogenic gene expression, browning of iWAT, and adaptive thermogenesis. CREBZF associates with PGC-1α to repress thermogenic gene expression. Expression levels of CREBZF are negatively correlated with UCP1 in human adipose tissues and increased in WAT of obese ob/ob mice, which may underscore the potential role of CREBZF in the development of compromised thermogenic capability under hyperglycemic conditions. Our results reveal an important mechanism of glucose sensing and thermogenic inactivation through reversible acetylation.


Subject(s)
Adipose Tissue, Brown , Glucose , Mice , Humans , Animals , Glucose/metabolism , Adipose Tissue, Brown/metabolism , Acetylation , Adipose Tissue, White/metabolism , Energy Metabolism , Obesity/genetics , Obesity/metabolism , Thermogenesis/genetics , Mice, Inbred C57BL , Basic-Leucine Zipper Transcription Factors/metabolism
16.
Sci Rep ; 14(1): 9157, 2024 04 22.
Article in English | MEDLINE | ID: mdl-38644456

ABSTRACT

Brown adipose tissue (BAT) which is a critical regulator of energy homeostasis, and its activity is inhibited by obesity and low-grade chronic inflammation. Ginsenoside Rg3, the primary constituent of Korean red ginseng (steamed Panax ginseng CA Meyer), has shown therapeutic potential in combating inflammatory and metabolic diseases. However, it remains unclear whether Rg3 can protect against the suppression of browning or activation of BAT induced by inflammation. In this study, we conducted a screening of ginsenoside composition in red ginseng extract (RGE) and explored the anti-adipogenic effects of both RGE and Rg3. We observed that RGE (exist 0.25 mg/mL of Rg3) exhibited significant lipid-lowering effects in adipocytes during adipogenesis. Moreover, treatment with Rg3 (60 µM) led to the inhibition of triglyceride accumulation, subsequently promoting enhanced fatty acid oxidation, as evidenced by the conversion of radiolabeled 3H-fatty acids into 3H-H2O with mitochondrial activation. Rg3 alleviated the attenuation of browning in lipopolysaccharide (LPS)-treated beige adipocytes and primary brown adipocytes by recovered by uncoupling protein 1 (UCP1) and the oxygen consumption rate compared to the LPS-treated group. These protective effects of Rg3 on inflammation-induced inhibition of beige and BAT-derived thermogenesis were confirmed in vivo by treating with CL316,243 (a beta-adrenergic receptor agonist) and LPS to induce browning and inflammation, respectively. Consistent with the in vitro data, treatment with Rg3 (2.5 mg/kg, 8 weeks) effectively reversed the LPS-induced inhibition of brown adipocyte features in C57BL/6 mice. Our findings confirm that Rg3-rich foods are potential browning agents that counteract chronic inflammation and metabolic complications.


Subject(s)
Adipose Tissue, Brown , Ginsenosides , Lipopolysaccharides , Mitochondria , Panax , Plant Extracts , Thermogenesis , Ginsenosides/pharmacology , Animals , Thermogenesis/drug effects , Panax/chemistry , Mitochondria/metabolism , Mitochondria/drug effects , Mice , Plant Extracts/pharmacology , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/metabolism , Adipose Tissue, Beige/metabolism , Adipose Tissue, Beige/drug effects , Mice, Inbred C57BL , Male , Adipogenesis/drug effects
17.
Nat Commun ; 15(1): 2856, 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38565851

ABSTRACT

Aging, chronic high-fat diet feeding, or housing at thermoneutrality induces brown adipose tissue (BAT) involution, a process characterized by reduction of BAT mass and function with increased lipid droplet size. Single nuclei RNA sequencing of aged mice identifies a specific brown adipocyte population of Ucp1-low cells that are pyroptotic and display a reduction in the longevity gene syntaxin 4 (Stx4a). Similar to aged brown adipocytes, Ucp1-STX4KO mice display loss of brown adipose tissue mass and thermogenic dysfunction concomitant with increased pyroptosis. Restoration of STX4 expression or suppression of pyroptosis activation protects against the decline in both mass and thermogenic activity in the aged and Ucp1-STX4KO mice. Mechanistically, STX4 deficiency reduces oxidative phosphorylation, glucose uptake, and glycolysis leading to reduced ATP levels, a known triggering signal for pyroptosis. Together, these data demonstrate an understanding of rapid brown adipocyte involution and that physiologic aging and thermogenic dysfunction result from pyroptotic signaling activation.


Subject(s)
Adipose Tissue, Brown , Pyroptosis , Animals , Mice , Adipocytes, Brown/metabolism , Adipose Tissue, Brown/metabolism , Signal Transduction , Thermogenesis/physiology , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism
18.
Epigenetics Chromatin ; 17(1): 12, 2024 Apr 27.
Article in English | MEDLINE | ID: mdl-38678237

ABSTRACT

BACKGROUND: Regulation of the thermogenic response by brown adipose tissue (BAT) is an important component of energy homeostasis with implications for the treatment of obesity and diabetes. Our preliminary analyses of RNA-Seq data uncovered many nodes representing epigenetic modifiers that are altered in BAT in response to chronic thermogenic activation. Thus, we hypothesized that chronic thermogenic activation broadly alters epigenetic modifications of DNA and histones in BAT. RESULTS: Motivated to understand how BAT function is regulated epigenetically, we developed a novel method for the first-ever unbiased top-down proteomic quantitation of histone modifications in BAT and validated our results with a multi-omic approach. To test our hypothesis, wildtype male C57BL/6J mice were housed under chronic conditions of thermoneutral temperature (TN, 28°C), mild cold/room temperature (RT, 22°C), or severe cold (SC, 8°C) and BAT was analyzed for DNA methylation and histone modifications. Methylation of promoters and intragenic regions in genomic DNA decrease in response to chronic cold exposure. Integration of DNA methylation and RNA expression datasets suggest a role for epigenetic modification of DNA in regulation of gene expression in response to cold. In response to cold housing, we observe increased bulk acetylation of histones H3.2 and H4, increased histone H3.2 proteoforms with di- and trimethylation of lysine 9 (K9me2 and K9me3), and increased histone H4 proteoforms with acetylation of lysine 16 (K16ac) in BAT. CONCLUSIONS: Our results reveal global epigenetically-regulated transcriptional "on" and "off" signals in murine BAT in response to varying degrees of chronic cold stimuli and establish a novel methodology to quantitatively study histones in BAT, allowing for direct comparisons to decipher mechanistic changes during the thermogenic response. Additionally, we make histone PTM and proteoform quantitation, RNA splicing, RRBS, and transcriptional footprint datasets available as a resource for future research.


Subject(s)
Adipose Tissue, Brown , Cold-Shock Response , DNA Methylation , Epigenesis, Genetic , Histones , Mice, Inbred C57BL , Animals , Adipose Tissue, Brown/metabolism , Mice , Male , Histones/metabolism , Histone Code , Thermogenesis , Cold Temperature
19.
Cells ; 13(8)2024 Apr 19.
Article in English | MEDLINE | ID: mdl-38667324

ABSTRACT

After exposure to cold stress, animals enhance the production of beige adipocytes and expedite thermogenesis, leading to improved metabolic health. Although brown adipose tissue in rodents is primarily induced by ß3-adrenergic receptor (ADRB3) stimulation, the activation of major ß-adrenergic receptors (ADRBs) in pigs has been a topic of debate. To address this, we developed overexpression vectors for ADRB1, ADRB2, and ADRB3 and silenced the expression of these receptors to observe their effects on the adipogenic differentiation stages of porcine preadipocytes. Our investigation revealed that cold stress triggers the transformation of subcutaneous white adipose tissue to beige adipose tissue in pigs by modulating adrenergic receptor levels. Meanwhile, we found that ADRB3 promotes the transformation of white adipocytes into beige adipocytes. Notably, ADRB3 enhances the expression of beige adipose tissue marker genes, consequently influencing cellular respiration and metabolism by regulating lipolysis and mitochondrial expression. Therefore, ADRB3 may serve as a pivotal gene in animal husbandry and contribute to the improvement of cold intolerance in piglets.


Subject(s)
Adipocytes, Beige , Cold Temperature , Receptors, Adrenergic, beta-3 , Animals , Receptors, Adrenergic, beta-3/metabolism , Receptors, Adrenergic, beta-3/genetics , Adipocytes, Beige/metabolism , Swine , Adipogenesis/genetics , Lipolysis , Thermogenesis/genetics , Cell Differentiation , Mitochondria/metabolism
20.
Int J Mol Sci ; 25(8)2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38673954

ABSTRACT

The objective was to assess whether low-protein (LP) diets regulate food intake (FI) and thermogenesis differently during thermoneutral (TN) and heat stress (HS) conditions. Two-hundred-day-old male broiler chicks were weight-matched and assigned to 36 pens with 5-6 chicks/pen. After 2 weeks of acclimation, birds were subjected into four groups (9 pens/group) including (1) a normal-protein diet under TN (ambient temperature), (2) an LP diet under TN, (3) a normal-protein diet under HS (35 °C for 7 h/day), and (4) an LP diet under HS, for 4 weeks. During HS, but not TN, LP tended to decrease FI, which might be associated with a lower mRNA abundance of duodenal ghrelin and higher GIP during HS. The LP group had a higher thermal radiation than NP under TN, but during HS, the LP group had a lower thermal radiation than NP. This was linked with higher a transcript of muscle ß1AR and AMPKα1 during TN, but not HS. Further, LP increased the gene expression of COX IV during TN but reduced COX IV and the sirtuin 1 abundance during HS. The dietary protein content differentially impacted plasma metabolome during TN and HS with divergent changes in amino acids such as tyrosine and tryptophan. Compared to NP, LP had increased abundances of p_Tenericutes, c_Mollicutes, c_Mollicutes_RF9, and f_tachnospiraceae under HS. Overall, LP diets may mitigate the negative outcome of heat stress on the survivability of birds by reducing FI and heat production. The differential effect of an LP diet on energy balance during TN and HS is likely regulated by gut and skeletal muscle and alterations in plasma metabolites and cecal microbiota.


Subject(s)
Chickens , Diet, Protein-Restricted , Energy Metabolism , Heat-Shock Response , Animals , Chickens/metabolism , Male , Thermogenesis , Animal Feed , Eating
SELECTION OF CITATIONS
SEARCH DETAIL
...