Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 511
Filter
1.
Nat Commun ; 15(1): 7957, 2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39261467

ABSTRACT

Postprandial IL-1ß surges are predominant in the white adipose tissue (WAT), but its consequences are unknown. Here, we investigate the role of IL-1ß in WAT energy storage and show that adipocyte-specific deletion of IL-1 receptor 1 (IL1R1) has no metabolic consequences, whereas ubiquitous lack of IL1R1 reduces body weight, WAT mass, and adipocyte formation in mice. Among all major WAT-resident cell types, progenitors express the highest IL1R1 levels. In vitro, IL-1ß potently promotes adipogenesis in murine and human adipose-derived stem cells. This effect is exclusive to early-differentiation-stage cells, in which the adipogenic transcription factors C/EBPδ and C/EBPß are rapidly upregulated by IL-1ß and enriched near important adipogenic genes. The pro-adipogenic, but not pro-inflammatory effect of IL-1ß is potentiated by acute treatment and blocked by chronic exposure. Thus, we propose that transient postprandial IL-1ß surges regulate WAT remodeling by promoting adipogenesis, whereas chronically elevated IL-1ß levels in obesity blunts this physiological function.


Subject(s)
Adipocytes , Adipogenesis , Adipose Tissue, White , CCAAT-Enhancer-Binding Protein-beta , Interleukin-1beta , Receptors, Interleukin-1 Type I , Adipogenesis/drug effects , Adipogenesis/genetics , Animals , Interleukin-1beta/metabolism , Humans , Adipocytes/metabolism , Adipocytes/cytology , Receptors, Interleukin-1 Type I/metabolism , Receptors, Interleukin-1 Type I/genetics , Mice , CCAAT-Enhancer-Binding Protein-beta/metabolism , CCAAT-Enhancer-Binding Protein-beta/genetics , Adipose Tissue, White/metabolism , Adipose Tissue, White/cytology , CCAAT-Enhancer-Binding Protein-delta/metabolism , CCAAT-Enhancer-Binding Protein-delta/genetics , Male , Mice, Knockout , Stem Cells/metabolism , Stem Cells/drug effects , Mice, Inbred C57BL , Cell Differentiation/drug effects
2.
Nat Commun ; 15(1): 6622, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39103342

ABSTRACT

Sex steroids modulate the distribution of mammalian white adipose tissues. Moreover, WAT remodeling requires adipocyte progenitor cells. Nevertheless, the sex-dependent mechanisms regulating adipocyte progenitors remain undetermined. Here, we uncover Cxcr4 acting in a sexually dimorphic manner to affect a pool of proliferating cells leading to restriction of female fat mass. We find that deletion of Cxcr4 in Pparγ-expressing cells results in female, not male, lipodystrophy, which cannot be restored by high-fat diet consumption. Additionally, Cxcr4 deletion is associated with a loss of a pool of proliferating adipocyte progenitors. Cxcr4 loss is accompanied by the upregulation of estrogen receptor alpha in adipose-derived PPARγ-labelled cells related to estradiol hypersensitivity and stalled adipogenesis. Estrogen removal or administration of antiestrogens restores WAT accumulation and dynamics of adipose-derived cells in Cxcr4-deficient mice. These findings implicate Cxcr4 as a female adipogenic rheostat, which may inform strategies to target female adiposity.


Subject(s)
Adipocytes , Adipogenesis , Adiposity , PPAR gamma , Receptors, CXCR4 , Stem Cells , Animals , Receptors, CXCR4/metabolism , Receptors, CXCR4/genetics , Female , Male , Mice , Adipocytes/metabolism , Adipocytes/cytology , Stem Cells/metabolism , Stem Cells/cytology , PPAR gamma/metabolism , PPAR gamma/genetics , Mice, Knockout , Adipose Tissue, White/metabolism , Adipose Tissue, White/cytology , Diet, High-Fat/adverse effects , Estrogen Receptor alpha/metabolism , Estrogen Receptor alpha/genetics , Mice, Inbred C57BL , Estradiol/pharmacology , Estradiol/metabolism , Cell Proliferation , Sex Factors , Sex Characteristics
3.
Genes (Basel) ; 15(8)2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39202377

ABSTRACT

White adipose tissue (WAT) makes up about 20-25% of total body mass in healthy individuals and is crucial for regulating various metabolic processes, including energy metabolism, endocrine function, immunity, and reproduction. In adipose tissue research, "adipogenesis" is commonly used to refer to the process of adipocyte formation, spanning from stem cell commitment to the development of mature, functional adipocytes. Although, this term should encompass a wide range of processes beyond commitment and differentiation, to also include other stages of adipose tissue development such as hypertrophy, hyperplasia, angiogenesis, macrophage infiltration, polarization, etc.… collectively, referred to herein as the adipogenic cycle. The term "differentiation", conversely, should only be used to refer to the process by which committed stem cells progress through distinct phases of subsequent differentiation. Recognizing this distinction is essential for accurately interpreting research findings on the mechanisms and stages of adipose tissue development and function. In this review, we focus on the molecular regulation of white adipose tissue development, from commitment to terminal differentiation, and examine key functional aspects of WAT that are crucial for normal physiology and systemic metabolic homeostasis.


Subject(s)
Adipogenesis , Adipose Tissue, White , Cell Differentiation , Stem Cells , Humans , Adipose Tissue, White/metabolism , Adipose Tissue, White/cytology , Animals , Cell Differentiation/genetics , Stem Cells/metabolism , Stem Cells/cytology , Adipogenesis/genetics , Adipocytes/metabolism , Adipocytes/cytology , Energy Metabolism
4.
Front Endocrinol (Lausanne) ; 15: 1396965, 2024.
Article in English | MEDLINE | ID: mdl-38982992

ABSTRACT

Adipose tissues, particularly beige and brown adipose tissue, play crucial roles in energy metabolism. Brown adipose tissues' thermogenic capacity and the appearance of beige cells within white adipose tissue have spurred interest in their metabolic impact and therapeutic potential. Brown and beige fat cells, activated by environmental factors like cold exposure or by pharmacology, share metabolic mechanisms that drive non-shivering thermogenesis. Understanding these two cell types requires advanced, yet broadly applicable in vitro models that reflect the complex microenvironment and vasculature of adipose tissues. Here we present mouse vascularized adipose spheroids of the stromal vascular microenvironment from inguinal white adipose tissue, a tissue with 'beiging' capacity in mice and humans. We show that adding a scaffold improves vascular sprouting, enhances spheroid growth, and upregulates adipogenic markers, thus reflecting increased adipocyte maturity. Transcriptional profiling via RNA sequencing revealed distinct metabolic pathways upregulated in our vascularized adipose spheroids, with increased expression of genes involved in glucose metabolism, lipid metabolism, and thermogenesis. Functional assessment demonstrated increased oxygen consumption in vascularized adipose spheroids compared to classical 2D cultures, which was enhanced by ß-adrenergic receptor stimulation correlating with elevated ß-adrenergic receptor expression. Moreover, stimulation with the naturally occurring adipokine, FGF21, induced Ucp1 mRNA expression in the vascularized adipose spheroids. In conclusion, vascularized inguinal white adipose tissue spheroids provide a physiologically relevant platform to study how the stromal vascular microenvironment shapes adipocyte responses and influence activated thermogenesis in beige adipocytes.


Subject(s)
Spheroids, Cellular , Thermogenesis , Animals , Mice , Spheroids, Cellular/metabolism , Adipose Tissue, White/metabolism , Adipose Tissue, White/cytology , Mice, Inbred C57BL , Male , Adipocytes/metabolism , Adipocytes/cytology , Adipose Tissue, Brown/metabolism , Adipose Tissue, Brown/cytology , Cells, Cultured , Adipocytes, Beige/metabolism , Adipocytes, Beige/cytology , Energy Metabolism , Adipogenesis/physiology , Microphysiological Systems
5.
Commun Biol ; 7(1): 787, 2024 Jun 29.
Article in English | MEDLINE | ID: mdl-38951550

ABSTRACT

Adipose tissue development begins in the fetal period, and continues to expand after birth. Dysregulation of adipose tissue during weaning may predispose individuals to lifelong metabolic disorders. However, the developmental remodeling of adipose tissue during weaning remains largely unexplored. Here we comprehensively compare the changes in mouse subcutaneous white adipose tissue from 7 days after birth to 7 days after weaning using single-cell RNA sequencing along with other molecular and histologic assays. We characterize the developmental trajectory of preadipocytes and indicate the commitment of preadipocytes with beige potential during weaning. Meanwhile, we find immune cells unique to weaning period, whose expression of extracellular matrix proteins implies potential regulation on preadipocyte. Finally, the strongest cell-cell interaction during weaning determined by the TGFß ligand-receptor pairs is between preadipocytes and endotheliocytes. Our results provide a detailed and unbiased cellular landscape and offer insights into the potential regulation of adipose tissue remodeling during weaning.


Subject(s)
Adipose Tissue, White , Single-Cell Analysis , Subcutaneous Fat , Weaning , Animals , Mice , Adipose Tissue, White/metabolism , Adipose Tissue, White/cytology , Subcutaneous Fat/metabolism , Subcutaneous Fat/cytology , Mice, Inbred C57BL , Adipocytes/metabolism , Adipocytes/cytology , Male , Female
7.
Int J Biochem Cell Biol ; 171: 106583, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38657899

ABSTRACT

Protein crotonylation plays a role in regulating cellular metabolism, gene expression, and other biological processes. NDUFA9 (NADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit 9) is closely associated with the activity and function of mitochondrial respiratory chain complex I. Mitochondrial function and respiratory chain are closely related to browning of white adipocytes, it's speculated that NDUFA9 and its crotonylation are associated with browning of white adipocytes. Firstly, the effect of NDUFA9 on white adipose tissue was verified in white fat browning model mice, and it was found that NDUFA9 promoted mitochondrial respiration, thermogenesis, and browning of white adipose tissue. Secondly, in cellular studies, it was discovered that NDUFA9 facilitated browning of white adipocytes by enhancing mitochondrial function, mitochondrial complex I activity, ATP synthesis, and mitochondrial respiration. Again, the level of NDUFA9 crotonylation was increased by treating cells with vorinostat (SAHA)+sodium crotonate (NaCr) and overexpressing NDUFA9, it was found that NDUFA9 crotonylation promoted browning of white adipocytes. Meanwhile, the acetylation level of NDUFA9 was increased by treating cells with SAHA+sodium acetate (NaAc) and overexpressing NDUFA9, the assay revealed that NDUFA9 acetylation inhibited white adipocytes browning. Finally, combined with the competitive relationship between acetylation and crotonylation, it was also demonstrated that NDUFA9 crotonylation promoted browning of white adipocytes. Above results indicate that NDUFA9 and its crotonylation modification promote mitochondrial function, which in turn promotes browning of white adipocytes. This study establishes a theoretical foundation for the management and intervention of obesity, which is crucial in addressing obesity and related medical conditions in the future.


Subject(s)
Adipocytes, White , Mitochondria , Animals , Mice , Mitochondria/metabolism , Mitochondria/drug effects , Adipocytes, White/metabolism , Adipocytes, White/drug effects , Adipocytes, White/cytology , Male , Mice, Inbred C57BL , Thermogenesis/drug effects , Adipocytes, Brown/metabolism , Adipocytes, Brown/drug effects , 3T3-L1 Cells , Electron Transport Complex I/metabolism , Electron Transport Complex I/genetics , Adipose Tissue, White/metabolism , Adipose Tissue, White/cytology , Acetylation/drug effects
8.
Nature ; 620(7972): 192-199, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37495690

ABSTRACT

Sympathetic activation during cold exposure increases adipocyte thermogenesis via the expression of mitochondrial protein uncoupling protein 1 (UCP1)1. The propensity of adipocytes to express UCP1 is under a critical influence of the adipose microenvironment and varies between sexes and among various fat depots2-7. Here we report that mammary gland ductal epithelial cells in the adipose niche regulate cold-induced adipocyte UCP1 expression in female mouse subcutaneous white adipose tissue (scWAT). Single-cell RNA sequencing shows that glandular luminal epithelium subtypes express transcripts that encode secretory factors controlling adipocyte UCP1 expression under cold conditions. We term these luminal epithelium secretory factors 'mammokines'. Using 3D visualization of whole-tissue immunofluorescence, we reveal sympathetic nerve-ductal contact points. We show that mammary ducts activated by sympathetic nerves limit adipocyte UCP1 expression via the mammokine lipocalin 2. In vivo and ex vivo ablation of mammary duct epithelium enhance the cold-induced adipocyte thermogenic gene programme in scWAT. Since the mammary duct network extends throughout most of the scWAT in female mice, females show markedly less scWAT UCP1 expression, fat oxidation, energy expenditure and subcutaneous fat mass loss compared with male mice, implicating sex-specific roles of mammokines in adipose thermogenesis. These results reveal a role of sympathetic nerve-activated glandular epithelium in adipocyte UCP1 expression and suggest that mammary duct luminal epithelium has an important role in controlling glandular adiposity.


Subject(s)
Adipocytes , Adipose Tissue, White , Epithelium , Mammary Glands, Animal , Thermogenesis , Animals , Female , Male , Mice , Adipocytes/metabolism , Adipose Tissue, White/cytology , Adipose Tissue, White/metabolism , Epithelium/innervation , Epithelium/metabolism , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism , Mammary Glands, Animal/cytology , Mammary Glands, Animal/innervation , Mammary Glands, Animal/physiology , Cold Temperature , Sympathetic Nervous System/physiology , Energy Metabolism , Oxidation-Reduction , Sex Characteristics
9.
J Nutr Biochem ; 100: 108898, 2022 02.
Article in English | MEDLINE | ID: mdl-34748921

ABSTRACT

The adipocytes play an important role in driving the obese-state-white adipose tissue (WAT) stores the excess energy as fat, wherein brown adipose tissue (BAT) is responsible for energy expenditure via the thermoregulatory function of uncoupling protein 1 (UCP1)-the imbalance between these two onsets obesity. Moreover, the anti-obesity effects of brown-like-adipocytes (beige) in WAT are well documented. Browning, the process of transformation of energy-storing into energy-dissipating adipocytes, is a potential preventive strategy against obesity and its related diseases. In the present study, to explore an alternative source of natural products in the regulation of adipocyte transformation, we assessed the potential of theobromine (TB), a bitter alkaloid of the cacao plant, inducing browning in mice (in vivo) and primary adipocytes (in vitro). Dietary supplementation of TB significantly increased skin temperature of the inguinal region in mice and induced the expression of UCP1 protein. It also increased the expression levels of mitochondrial marker proteins in subcutaneous adipose tissues but not in visceral adipose tissues. The microarray analysis showed that TB supplementation upregulated multiple thermogenic and beige adipocyte marker genes in subcutaneous adipose tissue. Furthermore, in mouse-derived primary adipocytes, TB upregulated the expression of the UCP1 protein and mitochondrial mass in a PPARγ ligand-dependent manner. It also increased the phosphorylation levels of PPARγ coactivator 1α without affecting its protein expression. These results indicate that dietary supplementation of TB induces browning in subcutaneous WAT and enhances PPARγ-induced UCP1 expression in vitro, suggesting its potential to treat obesity.


Subject(s)
Adipocytes, Beige/physiology , Adipocytes, White/physiology , Dietary Supplements , PPAR gamma/metabolism , Theobromine/administration & dosage , Adipocytes, White/drug effects , Adipose Tissue, White/cytology , Adipose Tissue, White/metabolism , Animals , Male , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Mitophagy , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Phosphorylation , Protons , Signal Transduction , Skin Temperature , Theobromine/pharmacology , Thermogenesis , Transcriptome , Uncoupling Protein 1/metabolism , Weight Gain
10.
Sci Rep ; 11(1): 24105, 2021 12 16.
Article in English | MEDLINE | ID: mdl-34916557

ABSTRACT

Thyroid hormone (TH) and thyroid hormone receptor (THR) regulate stem cell proliferation and differentiation during development, as well as during tissue renewal and repair in the adult. THR undergoes posttranslational modification by small ubiquitin-like modifier (SUMO). We generated the THRA (K283Q/K288R)-/- mouse model for in vivo studies and used human primary preadipocytes expressing the THRA sumoylation mutant (K283R/K288R) and isolated preadipocytes from mutant mice for in vitro studies. THRA mutant mice had reduced white adipose stores and reduced adipocyte cell diameter on a chow diet, compared to wild-type, and these differences were further enhanced after a high fat diet. Reduced preadipocyte proliferation in mutant mice, compared to wt, was shown after in vivo labeling of preadipocytes with EdU and in preadipocytes isolated from mice fat stores and studied in vitro. Mice with the desumoylated THRA had disruptions in cell cycle G1/S transition and this was associated with a reduction in the availability of cyclin D2 and cyclin-dependent kinase 2. The genes coding for cyclin D1, cyclin D2, cyclin-dependent kinase 2 and Culin3 are stimulated by cAMP Response Element Binding Protein (CREB) and contain CREB Response Elements (CREs) in their regulatory regions. We demonstrate, by Chromatin Immunoprecipitation (ChIP) assay, that in mice with the THRA K283Q/K288R mutant there was reduced CREB binding to the CRE. Mice with a THRA sumoylation mutant had reduced fat stores on chow and high fat diets and reduced adipocyte diameter.


Subject(s)
Adipose Tissue, White/metabolism , Sumoylation/physiology , Thyroid Hormone Receptors alpha/metabolism , Thyroid Hormone Receptors alpha/physiology , Adipocytes/pathology , Adipocytes/physiology , Adipose Tissue, White/cytology , Animals , CREB-Binding Protein/metabolism , Cell Proliferation , Diet, High-Fat/adverse effects , Humans , Mice , Mice, Mutant Strains , Small Ubiquitin-Related Modifier Proteins/physiology
11.
FASEB J ; 35(12): e22018, 2021 12.
Article in English | MEDLINE | ID: mdl-34731499

ABSTRACT

Adipose tissue is the primary site of energy storage, playing important roles in health. While adipose research largely focuses on obesity, fat also has other critical functions, producing adipocytokines and contributing to normal nutrient metabolism, which in turn play important roles in satiety and total energy homeostasis. SMAD2/3 proteins are downstream mediators of activin signaling, which regulate critical preadipocyte and mature adipocyte functions. Smad2 global knockout mice exhibit embryonic lethality, whereas global loss of Smad3 protects mice against diet-induced obesity. The direct contributions of Smad2 and Smad3 in adipose tissues, however, are unknown. Here, we sought to determine the primary effects of adipocyte-selective reduction of Smad2 or Smad3 on diet-induced adiposity using Smad2 or Smad3 "floxed" mice intercrossed with Adiponectin-Cre mice. Additionally, we examined visceral and subcutaneous preadipocyte differentiation efficiency in vitro. Almost all wild type subcutaneous preadipocytes differentiated into mature adipocytes. In contrast, visceral preadipocytes differentiated poorly. Exogenous activin A suppressed differentiation of preadipocytes from both depots. Smad2 conditional knockout (Smad2cKO) mice did not exhibit significant effects on weight gain, irrespective of diet, whereas Smad3 conditional knockout (Smad3cKO) male mice displayed a trend of reduced body weight on high-fat diet. On both diets, Smad3cKO mice displayed an adipose depot-selective phenotype, with a significant reduction in subcutaneous fat mass but not visceral fat mass. Our data suggest that Smad3 is an important contributor to the maintenance of subcutaneous white adipose tissue in a sex-selective fashion. These findings have implications for understanding SMAD-mediated, depot selective regulation of adipocyte growth and differentiation.


Subject(s)
Adipogenesis , Adipose Tissue, White/cytology , Adiposity , Intra-Abdominal Fat/cytology , Smad2 Protein/physiology , Smad3 Protein/physiology , Subcutaneous Fat/cytology , Activins/genetics , Activins/metabolism , Adipose Tissue, White/metabolism , Animals , Cell Differentiation , Diet, High-Fat , Female , Intra-Abdominal Fat/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Subcutaneous Fat/metabolism
12.
Cells ; 10(11)2021 11 08.
Article in English | MEDLINE | ID: mdl-34831295

ABSTRACT

Adipose tissue has been classified based on its morphology and function as white, brown, or beige/brite. It plays an essential role as a regulator of systemic metabolism through paracrine and endocrine signals. Recently, multiple adipocyte subtypes have been revealed using RNA sequencing technology, going beyond simply defined morphology but also by their cellular origin, adaptation to metabolic stress, and plasticity. Here, we performed an in-depth analysis of publicly available single-nuclei RNAseq from adipose tissue and utilized a workflow template to characterize adipocyte plasticity, heterogeneity, and secretome profiles. The reanalyzed dataset led to the identification of different subtypes of adipocytes including three subpopulations of thermogenic adipocytes, and provided a characterization of distinct transcriptional profiles along the adipocyte trajectory under thermogenic challenges. This study provides a useful resource for further investigations regarding mechanisms related to adipocyte plasticity and trans-differentiation.


Subject(s)
Adipocytes, White/cytology , Adipose Tissue, White/cytology , Cell Nucleus/metabolism , Cell Plasticity , RNA-Seq , Thermogenesis/physiology , Animals , Mice , Temperature , Uncoupling Protein 1/metabolism
13.
STAR Protoc ; 2(4): 100937, 2021 12 17.
Article in English | MEDLINE | ID: mdl-34778849

ABSTRACT

Isolation of viable immune cells from human tissues is critical for the characterization of cellular and molecular processes underlying disease pathogenesis. Here, we describe protocols for the isolation of highly viable immune cells from liver wedges and mesenteric white adipose tissue resections from obese persons. Notably, characterization of the isolated single-immune cell suspensions, via utility of basic immunological interrogations and genetic approaches, promises to generate an improved understanding of altered immunological pathways in obese individuals with or without metabolic diseases. For complete details on the use and execution of this protocol, please refer to Moreno-Fernandez et al. (2021).


Subject(s)
Cell Separation/methods , Immune System/cytology , Liver , Mesentery , Single-Cell Analysis/methods , Adipose Tissue, White/cytology , Adipose Tissue, White/pathology , Adolescent , Biopsy , Cells, Cultured , Female , Humans , Liver/cytology , Liver/pathology , Male , Mesentery/cytology , Mesentery/pathology , Pediatric Obesity
14.
Biochem Biophys Res Commun ; 585: 155-161, 2021 12 31.
Article in English | MEDLINE | ID: mdl-34801935

ABSTRACT

Glutamine is the most abundant amino acid in the body, and adipose tissue is one of the glutamine-producing organs. Glutamine has important and unique metabolic functions; however, its effects in adipocytes are still unclear. 3T3-L1 adipocytes produced and secreted glutamine dependent on glutamine synthetase, but preadipocytes did not. The inhibition of glutamine synthetase by l-methionine sulfoximine (MSO) impaired the differentiation of preadipocytes to mature adipocytes, and this inhibitory effect of MSO was rescued by exogenous glutamine supplementation. Glutamine concentrations were low, and Atgl gene expression was high in epididymal white adipose tissues of fasting mice in vivo. In 3T3-L1 adipocytes, glutamine deprivation induced Atgl expression and increased glycerol concentration in culture medium. Atgl expression is regulated by FoxO1, and glutamine deprivation reduced FoxO1 phosphorylation (Ser256), indicating the activation of FoxO1. These results demonstrate that glutamine is necessary for the differentiation of preadipocytes and regulates lipolysis through FoxO1 in mature adipocytes.


Subject(s)
Adipocytes/metabolism , Cell Differentiation/physiology , Glutamine/deficiency , Lipolysis/physiology , 3T3-L1 Cells , Adipocytes/cytology , Adipose Tissue, White/cytology , Adipose Tissue, White/metabolism , Animals , Blotting, Western , Cell Differentiation/genetics , Cells, Cultured , Forkhead Box Protein O1/genetics , Forkhead Box Protein O1/metabolism , Gene Expression Regulation , Glutamate-Ammonia Ligase/genetics , Glutamate-Ammonia Ligase/metabolism , Glutamine/metabolism , Lipase/genetics , Lipase/metabolism , Lipolysis/genetics , Male , Mice , Mice, Inbred C57BL , Reverse Transcriptase Polymerase Chain Reaction
15.
FASEB J ; 35(11): e21966, 2021 11.
Article in English | MEDLINE | ID: mdl-34624148

ABSTRACT

Adipose tissue is central to the regulation of energy balance. While white adipose tissue (WAT) is responsible for triglyceride storage, brown adipose tissue specializes in energy expenditure. Deterioration of brown adipocyte function contributes to the development of metabolic complications like obesity and diabetes. These disorders are also leading symptoms of the Bardet-Biedl syndrome (BBS), a hereditary disorder in humans which is caused by dysfunctions of the primary cilium and which therefore belongs to the group of ciliopathies. The cilium is a hair-like organelle involved in cellular signal transduction. The BBSome, a supercomplex of several Bbs gene products, localizes to the basal body of cilia and is thought to be involved in protein sorting to and from the ciliary membrane. The effects of a functional BBSome on energy metabolism and lipid mobilization in brown and white adipocytes were tested in whole-body Bbs4 knockout mice that were subjected to metabolic challenges. Chronic cold exposure reveals cold-intolerance of knockout mice but also ameliorates the markers of metabolic pathology detected in knockouts prior to cold. Hepatic triglyceride content is markedly reduced in knockout mice while circulating lipids are elevated, altogether suggesting that defective lipid metabolism in adipose tissue creates increased demand for systemic lipid mobilization to meet energetic demands of reduced body temperatures. These findings taken together suggest that Bbs4 is essential for the regulation of adipose tissue lipid metabolism, representing a potential target to treat metabolic disorders.


Subject(s)
Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Lipid Metabolism , Microtubule-Associated Proteins/physiology , Adipose Tissue, Brown/cytology , Adipose Tissue, White/cytology , Animals , Energy Metabolism , Male , Mesenchymal Stem Cells , Mice , Mice, Inbred C57BL , Thermogenesis
16.
Food Funct ; 12(19): 9151-9164, 2021 Oct 04.
Article in English | MEDLINE | ID: mdl-34606532

ABSTRACT

Metabolic syndrome caused obesity has long been recognized as a risk of health. Celery and celery extracts have various medicinal properties, such as anti-diabetes and anti-inflammatory properties and blood glucose and serum lipid reduction. However, the effect of probiotic fermentation on celery juice and the association between fermented celery juice (FCJ) and obesity were unclear. This study aimed to evaluate the beneficial effects of FCJ on high-fat diet (HFD) induced obesity and related metabolic syndromes. C57BL/6 mice were randomly divided into six groups (n = 15 per group) fed either a normal diet (ND) or HFD with or without CJ/FCJ (10 g kg-1 day-1) by oral gavage for 12 weeks. Here we demonstrated that the probiotic fermentation of celery juice (CJ) could enhance the active ingredients in celery, such as total polyphenols, flavonoids, vitamin C and SOD. Compared to the slight improvement induced by CJ ingestion, FCJ intake significantly inhibited body weight gain, prevented dyslipidemia and hyperglycemia, and suppressed visceral fat accumulation. Furthermore, 16S rRNA sequencing analysis revealed that FCJ intake altered the composition of gut microbiota, increasing the ratio of Firmicutes/Bacteroidetes and the relative abundance of beneficial bacteria (Lactobacillus, Ruminococcaceae_UCG-014, Faecalibaculum and Blautia), and decreasing the relative abundance of harmful bacteria (Alloprevotella and Helicobacter). These findings suggest that FCJ can prevent HFD-induced obesity and become a novel gut microbiota modulator to prevent HFD-induced gut dysbiosis and obesity-related metabolic disorders.


Subject(s)
Apium , Diet, High-Fat , Dietary Supplements , Fermented Beverages , Gastrointestinal Microbiome , Obesity/prevention & control , Adipocytes/cytology , Adipocytes/physiology , Adipose Tissue, Brown/cytology , Adipose Tissue, White/cytology , Animals , Diabetes Mellitus, Type 2/prevention & control , Dyslipidemias/prevention & control , Fermented Beverages/analysis , Hyperglycemia/prevention & control , Intra-Abdominal Fat/anatomy & histology , Male , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/prevention & control
17.
Int J Mol Sci ; 22(18)2021 Sep 09.
Article in English | MEDLINE | ID: mdl-34575926

ABSTRACT

Epigenetic signals and chromatin-modifying proteins play critical roles in adipogenesis, which determines the risk of obesity and which has recently attracted increasing interest. Histone demethylase 2A (KDM2A) is an important component of histone demethylase; however, its direct effect on fat deposition remains unclear. Here, a KDM2A loss of function was performed using two unbiased methods, small interfering RNA (siRNA) and Cre-Loxp recombinase systems, to reveal its function in adipogenesis. The results show that the knockdown of KDM2A by siRNAs inhibited the proliferation capacity of 3T3-L1 preadipocytes. Furthermore, the promotion of preadipocyte differentiation was observed in siRNA-treated cells, manifested by the increasing content of lipid droplets and the expression level of adipogenic-related genes. Consistently, the genetic deletion of KDM2A by Adipoq-Cre in primary adipocytes exhibited similar phenotypes to those of 3T3-L1 preadipocytes. Interestingly, the knockdown of KDM2A upregulates the expression level of Transportin 1(TNPO1), which in turn may induce the nuclear translocation of PPARγ and the accumulation of lipid droplets. In conclusion, the ablation of KDM2A inhibits preadipocyte proliferation and promotes its adipogenic differentiation. This work provides direct evidence of the exact role of KDM2A in fat deposition and provides theoretical support for obesity therapy that targets KDM2A.


Subject(s)
Adipocytes/cytology , Adipocytes/metabolism , Adipogenesis/genetics , Cell Differentiation/genetics , Jumonji Domain-Containing Histone Demethylases/genetics , 3T3-L1 Cells , Adipose Tissue, White/cytology , Adipose Tissue, White/metabolism , Animals , Cell Proliferation , Gene Expression Regulation , Gene Knockout Techniques , Jumonji Domain-Containing Histone Demethylases/metabolism , Mice , Protein Transport
18.
Int J Mol Sci ; 22(17)2021 Aug 27.
Article in English | MEDLINE | ID: mdl-34502211

ABSTRACT

Obesity is a condition characterized by uncontrolled expansion of adipose tissue mass resulting in pathological weight gain. Histone deacetylases (HDACs) have emerged as crucial players in epigenetic regulation of adipocyte metabolism. Previously, we demonstrated that selective inhibition of class I HDACs improves white adipocyte functionality and promotes the browning phenotype of murine mesenchymal stem cells (MSCs) C3H/10T1/2 differentiated to adipocytes. These effects were also observed in db/db and diet induced obesity mouse models and in mice with adipose-selective inactivation of HDAC3, a member of class I HDACs. The molecular basis of class I HDACs action in adipose tissue is not deeply characterized and it is not known whether the effects of their inhibition are exerted on adipocyte precursors or mature adipocytes. Therefore, the aim of the present work was to explore the molecular mechanism of class I HDAC action in adipocytes by evaluating the effects of HDAC3-specific silencing at different stages of differentiation. HDAC3 was silenced in C3H/10T1/2 MSCs at different stages of differentiation to adipocytes. shRNA targeting HDAC3 was used to generate the knock-down model. Proper HDAC3 silencing was assessed by measuring both mRNA and protein levels of mouse HDAC3 via qPCR and western blot, respectively. Mitochondrial DNA content and gene expression were quantified via qPCR. HDAC3 silencing at the beginning of differentiation enhanced adipocyte functionality by amplifying the expression of genes regulating differentiation, oxidative metabolism, browning and mitochondrial activity, starting from 72 h after induction of differentiation and silencing. Insulin signaling was enhanced as demonstrated by increased AKT phosphorylation following HDAC3 silencing. Mitochondrial content/density did not change, while the increased expression of the transcriptional co-activator Ppargc1b suggests the observed phenotype was related to enhanced mitochondrial activity, which was confirmed by increased maximal respiration and proton leak linked to reduced coupling efficiency. Moreover, the expression of pro-inflammatory markers increased with HDAC3 early silencing. To the contrary, no differences in terms of gene expression were found when HDAC3 silencing occurred in terminally differentiated adipocyte. Our data demonstrated that early epigenetic events mediated by class I HDAC inhibition/silencing are crucial to commit adipocyte precursors towards the above-mentioned metabolic phenotype. Moreover, our data suggest that these effects are exerted on adipocyte precursors.


Subject(s)
Adipose Tissue, Brown/physiology , Adipose Tissue, White/physiology , Cell Differentiation , Gene Expression Regulation , Histone Deacetylases/metabolism , Mitochondria/metabolism , Phenotype , Adipose Tissue, Brown/cytology , Adipose Tissue, White/cytology , Animals , Histone Deacetylases/genetics , Mice , Mice, Inbred C3H
19.
Biochem Biophys Res Commun ; 578: 1-6, 2021 11 12.
Article in English | MEDLINE | ID: mdl-34520979

ABSTRACT

Developmentally regulated GTP-binding protein 2 (DRG2) participates in the regulation of proliferation and differentiation of multiple cells. However, whether DRG2 regulates adipocyte differentiation and related metabolic control remains elusive. This study revealed increases in body weight and adiposity in DRG2 transgenic (Tg) mice overexpressing DRG2. Consistent with these results, DRG2 Tg mice showed increased expression of genes involved in adipogenesis and lipid metabolism in the white adipose tissue. DRG2 was also identified to control adipogenesis by cooperating with peroxisome proliferator activated receptor-γ (PPAR-γ) in cultured adipocytes. Overall, the findings of the current study suggest that DRG2 plays an active role in regulating adipocyte differentiation, and thus participates in the development of obesity during exposure to a fat-rich diet.


Subject(s)
Adipose Tissue, White/cytology , GTP-Binding Proteins/metabolism , PPAR gamma/metabolism , Adipogenesis , Adipose Tissue, White/metabolism , Animals , Body Weight , Cell Differentiation , Disease Models, Animal , GTP-Binding Proteins/genetics , Lipid Metabolism , Mice , Mice, Transgenic
20.
Biol Open ; 10(9)2021 09 15.
Article in English | MEDLINE | ID: mdl-34409430

ABSTRACT

White adipose tissue hyperplasia has been shown to be crucial for handling excess energy in healthy ways. Though adipogenesis mechanisms have been underscored in vitro, we lack information on how tissue and systemic factors influence the differentiation of new adipocytes. While this could be studied in zebrafish, adipocyte identification currently relies on neutral lipid labeling, thus precluding access to cells in early stages of differentiation. Here we report the generation and analysis of a zebrafish line with the transgene fabp4a(-2.7):EGFPcaax. In vivo confocal microscopy of the pancreatic and abdominal visceral depots of transgenic larvae, revealed the presence of labeled mature adipocytes as well as immature cells in earlier stages of differentiation. Through co-labeling for blood vessels, we observed a close interaction of differentiating adipocytes with endothelial cells through cell protrusions. Finally, we implemented hyperspectral imaging and spectral phasor analysis in Nile Red-labeled transgenic larvae and revealed the lipid metabolic transition towards neutral lipid accumulation of differentiating adipocytes. Altogether our work presents the characterization of a novel adipocyte-specific label in zebrafish and uncovers previously unknown aspects of in vivo adipogenesis. This article has an associated First Person interview with the first author of the paper.


Subject(s)
Adipocytes/physiology , Adipogenesis/genetics , Adipose Tissue, White/cytology , Cell Differentiation/genetics , Zebrafish/embryology , Adiponectin/metabolism , Animals , Animals, Genetically Modified , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Line , Complement Factor D/metabolism , Endothelial Cells/physiology , Fatty Acid-Binding Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL