Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Nat Immunol ; 22(10): 1268-1279, 2021 10.
Article in English | MEDLINE | ID: mdl-34556885

ABSTRACT

Metabolic inflammation is closely linked to obesity, and is implicated in the pathogenesis of metabolic diseases. FTO harbors the strongest genetic association with polygenic obesity, and IRX3 mediates the effects of FTO on body weight. However, in what cells and how IRX3 carries out this control are poorly understood. Here we report that macrophage IRX3 promotes metabolic inflammation to accelerate the development of obesity and type 2 diabetes. Mice with myeloid-specific deletion of Irx3 were protected against diet-induced obesity and metabolic diseases via increasing adaptive thermogenesis. Mechanistically, macrophage IRX3 promoted proinflammatory cytokine transcription and thus repressed adipocyte adrenergic signaling, thereby inhibiting lipolysis and thermogenesis. JNK1/2 phosphorylated IRX3, leading to its dimerization and nuclear translocation for transcription. Further, lipopolysaccharide stimulation stabilized IRX3 by inhibiting its ubiquitination, which amplified the transcriptional capacity of IRX3. Together, our findings identify a new player, macrophage IRX3, in the control of body weight and metabolic inflammation, implicating IRX3 as a therapeutic target.


Subject(s)
Homeodomain Proteins/metabolism , Inflammation/metabolism , Macrophages/metabolism , Obesity/metabolism , Transcription Factors/metabolism , Adipocytes/metabolism , Adult , Animals , Body Weight/physiology , Cell Line , Diabetes Mellitus, Type 2/metabolism , Diet/methods , HEK293 Cells , Humans , Male , Metabolic Diseases/metabolism , Mice , RAW 264.7 Cells , THP-1 Cells , Thermogenesis/physiology , Transcription, Genetic/physiology , Young Adult
2.
Immunity ; 57(3): 513-527.e6, 2024 Mar 12.
Article in English | MEDLINE | ID: mdl-38262419

ABSTRACT

Accumulation of senescent cells in organs and tissues is a hallmark of aging and known to contribute to age-related diseases. Although aging-associated immune dysfunction, or immunosenescence, is known to contribute to this process, the underlying mechanism remains elusive. Here, we report that type 2 cytokine signaling deficiency accelerated aging and, conversely, that the interleukin-4 (IL-4)-STAT6 pathway protected macrophages from senescence. Mechanistically, activated STAT6 promoted the expression of genes involved in DNA repair both via homologous recombination and Fanconi anemia pathways. Conversely, STAT6 deficiency induced release of nuclear DNA into the cytoplasm to promote tissue inflammation and organismal aging. Importantly, we demonstrate that IL-4 treatment prevented macrophage senescence and improved the health span of aged mice to an extent comparable to senolytic treatment, with further additive effects when combined. Together, our findings support that type 2 cytokine signaling protects macrophages from immunosenescence and thus hold therapeutic potential for improving healthy aging.


Subject(s)
Cellular Senescence , Interleukin-4 , Animals , Mice , Interleukin-4/metabolism , Aging/genetics , Macrophages , Inflammation
3.
Cell ; 160(1-2): 74-87, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25543153

ABSTRACT

Type 2 innate lymphoid cells (ILC2s), an innate source of the type 2 cytokines interleukin (IL)-5 and -13, participate in the maintenance of tissue homeostasis. Although type 2 immunity is critically important for mediating metabolic adaptations to environmental cold, the functions of ILC2s in beige or brown fat development are poorly defined. We report here that activation of ILC2s by IL-33 is sufficient to promote the growth of functional beige fat in thermoneutral mice. Mechanistically, ILC2 activation results in the proliferation of bipotential adipocyte precursors (APs) and their subsequent commitment to the beige fat lineage. Loss- and gain-of-function studies reveal that ILC2- and eosinophil-derived type 2 cytokines stimulate signaling via the IL-4Rα in PDGFRα(+) APs to promote beige fat biogenesis. Together, our results highlight a critical role for ILC2s and type 2 cytokines in the regulation of adipocyte precursor numbers and fate, and as a consequence, adipose tissue homeostasis. PAPERCLIP:


Subject(s)
Adipose Tissue, Brown/metabolism , Lymphocytes/metabolism , Adipocytes/cytology , Adipocytes/metabolism , Animals , Cell Proliferation , Female , Interleukin-13/metabolism , Interleukin-33 , Interleukins/immunology , Lymphocytes/cytology , Male , Mice , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Receptors, Interleukin-4/metabolism , Signal Transduction , Stem Cells/metabolism
4.
Nat Immunol ; 18(5): 519-529, 2017 05.
Article in English | MEDLINE | ID: mdl-28346409

ABSTRACT

Obesity is associated with metabolic inflammation and endoplasmic reticulum (ER) stress, both of which promote metabolic disease progression. Adipose tissue macrophages (ATMs) are key players orchestrating metabolic inflammation, and ER stress enhances macrophage activation. However, whether ER stress pathways underlie ATM regulation of energy homeostasis remains unclear. Here, we identified inositol-requiring enzyme 1α (IRE1α) as a critical switch governing M1-M2 macrophage polarization and energy balance. Myeloid-specific IRE1α abrogation in Ern1f/f; Lyz2-Cre mice largely reversed high-fat diet (HFD)-induced M1-M2 imbalance in white adipose tissue (WAT) and blocked HFD-induced obesity, insulin resistance, hyperlipidemia and hepatic steatosis. Brown adipose tissue (BAT) activity, WAT browning and energy expenditure were significantly higher in Ern1f/f; Lyz2-Cre mice. Furthermore, IRE1α ablation augmented M2 polarization of macrophages in a cell-autonomous manner. Thus, IRE1α senses protein unfolding and metabolic and immunological states, and consequently guides ATM polarization. The macrophage IRE1α pathway drives obesity and metabolic syndrome through impairing BAT activity and WAT browning.


Subject(s)
Adipose Tissue, Brown/physiology , Adipose Tissue, White/pathology , Endoribonucleases/metabolism , Macrophages/physiology , Obesity/immunology , Protein Serine-Threonine Kinases/metabolism , Animals , Cell Differentiation/genetics , Diet, High-Fat , Disease Models, Animal , Endoplasmic Reticulum Stress , Endoribonucleases/genetics , Energy Metabolism/genetics , Humans , Macrophage Activation/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Serine-Threonine Kinases/genetics
5.
Cell ; 157(6): 1292-1308, 2014 Jun 05.
Article in English | MEDLINE | ID: mdl-24906148

ABSTRACT

Beige fat, which expresses the thermogenic protein UCP1, provides a defense against cold and obesity. Although a cold environment is the physiologic stimulus for inducing beige fat in mice and humans, the events that lead from the sensing of cold to the development of beige fat remain poorly understood. Here, we identify the efferent beige fat thermogenic circuit, consisting of eosinophils, type 2 cytokines interleukin (IL)-4/13, and alternatively activated macrophages. Genetic loss of eosinophils or IL-4/13 signaling impairs cold-induced biogenesis of beige fat. Mechanistically, macrophages recruited to cold-stressed subcutaneous white adipose tissue (scWAT) undergo alternative activation to induce tyrosine hydroxylase expression and catecholamine production, factors required for browning of scWAT. Conversely, administration of IL-4 to thermoneutral mice increases beige fat mass and thermogenic capacity to ameliorate pre-established obesity. Together, our findings have uncovered the efferent circuit controlling biogenesis of beige fat and provide support for its targeting to treat obesity.


Subject(s)
Adipose Tissue, Brown/metabolism , Eosinophils/metabolism , Interleukin-13/metabolism , Interleukin-4/metabolism , Macrophages/metabolism , Signal Transduction , Adipocytes, Brown/metabolism , Animals , Catecholamines/metabolism , Cold Temperature , Interleukin-13/genetics , Interleukin-4/genetics , Male , Mice , Mice, Inbred C57BL , Myeloid Cells/metabolism , Obesity/metabolism , Receptors, CCR2/metabolism , STAT6 Transcription Factor/metabolism , Thermogenesis
6.
Nature ; 480(7375): 104-8, 2011 Nov 20.
Article in English | MEDLINE | ID: mdl-22101429

ABSTRACT

All homeotherms use thermogenesis to maintain their core body temperature, ensuring that cellular functions and physiological processes can continue in cold environments. In the prevailing model of thermogenesis, when the hypothalamus senses cold temperatures it triggers sympathetic discharge, resulting in the release of noradrenaline in brown adipose tissue and white adipose tissue. Acting via the ß(3)-adrenergic receptors, noradrenaline induces lipolysis in white adipocytes, whereas it stimulates the expression of thermogenic genes, such as PPAR-γ coactivator 1a (Ppargc1a), uncoupling protein 1 (Ucp1) and acyl-CoA synthetase long-chain family member 1 (Acsl1), in brown adipocytes. However, the precise nature of all the cell types involved in this efferent loop is not well established. Here we report in mice an unexpected requirement for the interleukin-4 (IL-4)-stimulated program of alternative macrophage activation in adaptive thermogenesis. Exposure to cold temperature rapidly promoted alternative activation of adipose tissue macrophages, which secrete catecholamines to induce thermogenic gene expression in brown adipose tissue and lipolysis in white adipose tissue. Absence of alternatively activated macrophages impaired metabolic adaptations to cold, whereas administration of IL-4 increased thermogenic gene expression, fatty acid mobilization and energy expenditure, all in a macrophage-dependent manner. Thus, we have discovered a role for alternatively activated macrophages in the orchestration of an important mammalian stress response, the response to cold.


Subject(s)
Catecholamines/metabolism , Macrophage Activation , Macrophages/physiology , Stress, Physiological/physiology , Thermogenesis/physiology , Adipose Tissue/cytology , Adipose Tissue/metabolism , Animals , Body Temperature/genetics , Cells, Cultured , Cold Temperature , Energy Metabolism , Gene Expression Regulation , Humans , Interleukin-4 , Macrophages/metabolism , Male , Mice , Mice, Inbred BALB C , U937 Cells
7.
Blood ; 122(19): 3263-7, 2013 Nov 07.
Article in English | MEDLINE | ID: mdl-24065242

ABSTRACT

The rise of obesity and its attendant pathological sequelae, including type 2 diabetes and coronary artery disease, constitute an ongoing public health catastrophe in both the developed and, more recently, the developing world. Although the underlying pathophysiology is complex, chronic low-grade inflammation has emerged as a central driver of both primary metabolic dysfunction and subsequent tissue failure. Importantly, this inflammation has been shown to arise as a consequence of both the disruption of homeostatic tissue resident leukocytes and the recruitment of antagonistic effector cells from the circulation. In this review, we discuss the roles of visceral adipose tissue's salient leukocyte lineages in the transition to obesity and highlight key points at which this emerging immune axis may be manipulated for therapeutic effect.


Subject(s)
Coronary Artery Disease/pathology , Diabetes Mellitus, Type 2/pathology , Intra-Abdominal Fat/pathology , Leukocytes/pathology , Obesity/pathology , Cell Movement , Coronary Artery Disease/etiology , Coronary Artery Disease/immunology , Diabetes Mellitus, Type 2/etiology , Diabetes Mellitus, Type 2/immunology , Humans , Immunity, Innate , Inflammation/immunology , Inflammation/pathology , Insulin Resistance , Intra-Abdominal Fat/immunology , Leukocytes/immunology , Macrophages/immunology , Macrophages/pathology , Obesity/complications , Obesity/immunology
8.
Proc Natl Acad Sci U S A ; 108(38): 15852-7, 2011 Sep 20.
Article in English | MEDLINE | ID: mdl-21911379

ABSTRACT

The endoplasmic reticulum (ER)-resident protein kinase/endoribonuclease inositol-requiring enzyme 1 (IRE1) is activated through transautophosphorylation in response to protein folding overload in the ER lumen and maintains ER homeostasis by triggering a key branch of the unfolded protein response. Here we show that mammalian IRE1α in liver cells is also phosphorylated by a kinase other than itself in response to metabolic stimuli. Glucagon-stimulated protein kinase PKA, which in turn phosphorylated IRE1α at Ser(724), a highly conserved site within the kinase activation domain. Blocking Ser(724) phosphorylation impaired the ability of IRE1α to augment the up-regulation by glucagon signaling of the expression of gluconeogenic genes. Moreover, hepatic IRE1α was highly phosphorylated at Ser(724) by PKA in mice with obesity, and silencing hepatic IRE1α markedly reduced hyperglycemia and glucose intolerance. Hence, these results suggest that IRE1α integrates signals from both the ER lumen and the cytoplasm in the liver and is coupled to the glucagon signaling in the regulation of glucose metabolism.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Endoribonucleases/metabolism , Glucagon/pharmacology , Glucose/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Base Sequence , Cyclic AMP-Dependent Protein Kinases/genetics , Cytoplasm/metabolism , Endoplasmic Reticulum/metabolism , Endoribonucleases/genetics , Gene Expression Profiling , Hepatocytes/cytology , Hepatocytes/drug effects , Hepatocytes/metabolism , Immunoblotting , Liver/cytology , Liver/enzymology , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mutation , Obesity/genetics , Obesity/metabolism , Oligonucleotide Array Sequence Analysis , Phosphorylation/drug effects , Primary Cell Culture , Protein Serine-Threonine Kinases/genetics , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Serine/genetics , Serine/metabolism
9.
Nat Metab ; 6(4): 708-723, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38499763

ABSTRACT

Cachexia affects 50-80% of patients with cancer and accounts for 20% of cancer-related death, but the underlying mechanism driving cachexia remains elusive. Here we show that circulating lactate levels positively correlate with the degree of body weight loss in male and female patients suffering from cancer cachexia, as well as in clinically relevant mouse models. Lactate infusion per se is sufficient to trigger a cachectic phenotype in tumour-free mice in a dose-dependent manner. Furthermore, we demonstrate that adipose-specific G-protein-coupled receptor (GPR)81 ablation, similarly to global GPR81 deficiency, ameliorates lactate-induced or tumour-induced adipose and muscle wasting in male mice, revealing adipose GPR81 as the major mediator of the catabolic effects of lactate. Mechanistically, lactate/GPR81-induced cachexia occurs independently of the well-established protein kinase A catabolic pathway, but it is mediated by a signalling cascade sequentially activating Gi-Gßγ-RhoA/ROCK1-p38. These findings highlight the therapeutic potential of targeting GPR81 for the treatment of this life-threatening complication of cancer.


Subject(s)
Cachexia , Lactic Acid , Neoplasms , Receptors, G-Protein-Coupled , Cachexia/metabolism , Cachexia/etiology , Animals , Receptors, G-Protein-Coupled/metabolism , Mice , Humans , Lactic Acid/metabolism , Male , Female , Neoplasms/metabolism , Neoplasms/complications , Signal Transduction
10.
Trends Cell Biol ; 33(3): 182-184, 2023 03.
Article in English | MEDLINE | ID: mdl-36517314

ABSTRACT

The mitochondrial calcium uniporter (MCU) controls mitochondrial bioenergetics, and its activity varies greatly between tissues. Here, we highlight a recently identified MCU-EMRE-UCP1 complex, named thermoporter, in the adaptive thermogenesis of brown adipose tissue (BAT). The thermoporter enhances MCU activity to promote thermogenic metabolism, demonstrating a BAT-specific regulation for MCU activity.


Subject(s)
Calcium Channels , Mitochondria , Humans , Calcium Channels/metabolism , Mitochondria/metabolism , Cell Membrane/metabolism , Calcium/metabolism
11.
Nat Commun ; 14(1): 7102, 2023 11 04.
Article in English | MEDLINE | ID: mdl-37925548

ABSTRACT

Sympathetic innervation is essential for the development of functional beige fat that maintains body temperature and metabolic homeostasis, yet the molecular mechanisms controlling this innervation remain largely unknown. Here, we show that adipocyte YAP/TAZ inhibit sympathetic innervation of beige fat by transcriptional repression of neurotropic factor S100B. Adipocyte-specific loss of Yap/Taz induces S100b expression to stimulate sympathetic innervation and biogenesis of functional beige fat both in subcutaneous white adipose tissue (WAT) and browning-resistant visceral WAT. Mechanistically, YAP/TAZ compete with C/EBPß for binding to the zinc finger-2 domain of PRDM16 to suppress S100b transcription, which is released by adrenergic-stimulated YAP/TAZ phosphorylation and inactivation. Importantly, Yap/Taz loss in adipocytes or AAV-S100B overexpression in visceral WAT restricts both age-associated and diet-induced obesity, and improves metabolic homeostasis by enhancing energy expenditure of mice. Together, our data reveal that YAP/TAZ act as a brake on the beige fat innervation by blocking PRDM16-C/EBPß-mediated S100b expression.


Subject(s)
Adipose Tissue, Beige , Transcription Factors , Mice , Animals , Adipose Tissue, Beige/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Adipocytes/metabolism , Obesity/metabolism , Adipose Tissue, White/metabolism , Adipose Tissue, Brown/metabolism , Thermogenesis/genetics
12.
STAR Protoc ; 3(3): 101480, 2022 09 16.
Article in English | MEDLINE | ID: mdl-35755124

ABSTRACT

The communication between macrophage and adipocyte plays a critical role in the initiation and development of metabolic inflammation, which is difficult to study in vivo. Here, we provide a step-by-step protocol using differentiated cells to investigate the paracrine effects of classically activated macrophage on beige adipocyte metabolism in vitro. This protocol uses bone-marrow-derived macrophage and SVF-derived UCP1+ beige adipocyte in a culture model to study immune regulation of adipocyte metabolism by western blot analyses. For complete details on the use and execution of this protocol, please refer to Yao et al. (2021).


Subject(s)
Adipocytes, Beige , Adipocytes/metabolism , Animals , Cell Differentiation , Macrophages , Mice , Thermogenesis
13.
STAR Protoc ; 3(4): 101895, 2022 12 16.
Article in English | MEDLINE | ID: mdl-36595932

ABSTRACT

The classical Cre-LoxP system is time consuming. Here we detail a protocol that leverages Rosa26-LSL-Cas9;Adiponectin-Cre mice to restrict Cas9 expression in adipocytes. This enables specific deletion of target genes in brown adipocytes within 6 weeks by local injection of AAV-sgRNA into interscapular brown adipose tissue. We also describe an adiponectin-promoter-driven AAV vector to express sgRNA-resistant cDNA-encoded protein for subsequent rescue. This protocol thus provides an efficient means to specifically knockout and overexpress genes in brown adipocytes in vivo. For complete details on the use and execution of this protocol, please refer to Xue et al. (2022).1.


Subject(s)
Adipocytes, Brown , Adiponectin , Mice , Animals , Adipocytes, Brown/metabolism , Gene Knockout Techniques , Adiponectin/genetics , Adiponectin/metabolism , Adipose Tissue, Brown/metabolism , Promoter Regions, Genetic
14.
Front Immunol ; 13: 977485, 2022.
Article in English | MEDLINE | ID: mdl-36119080

ABSTRACT

Adipose tissue macrophage (ATM) has been appreciated for its critical contribution to obesity-associated metabolic diseases in recent years. Here, we discuss the regulation of ATM on both metabolic homeostatsis and dysfunction. In particular, the macrophage polarization and recruitment as well as the crosstalk between ATM and adipocyte in thermogenesis, obesity, insulin resistance and adipose tissue fibrosis have been reviewed. A better understanding of how ATM regulates adipose tissue remodeling may provide novel therapeutic strategies against obesity and associated metabolic diseases.


Subject(s)
Inflammation , Insulin Resistance , Adipose Tissue/metabolism , Humans , Inflammation/metabolism , Macrophages/metabolism , Obesity/metabolism
15.
Nat Commun ; 13(1): 6030, 2022 10 13.
Article in English | MEDLINE | ID: mdl-36229481

ABSTRACT

Fibrosis disrupts adipose tissue (AT) homeostasis and exacerbates metabolic dysfunction upon chronic caloric excess. The molecular mechanisms linking adipocyte plasticity to AT fibrosis are largely unknown. Here we show that the Hippo pathway is coupled with TGFß signaling to orchestrate a cellular and/or functional shift of adipocytes from energy storage to extracellular matrix (ECM) remodeling in AT fibrosis. We found that Lats1/2-knockout adipocytes could dedifferentiate into DPP4+ progenitor cells and convert to DPP4- myofibroblasts upon TGFß stimulation. On the other hand, Hippo pathway inhibition during obesity impaired adipocyte identity while promoted ECM remodeling activity of adipocytes. Macrophages recruited by CCL2 produced TGFß to accelerate AT fibrosis. YAP and TAZ, the Hippo downstream effectors, enhanced SMAD2 stability to promote fibrotic responses. Importantly, inhibition of YAP/TAZ activity in obese mice markedly relieved AT fibrosis and improved metabolic homeostasis. Together, our findings identify the Hippo pathway as a molecular switch in the initiation and development of AT fibrosis, implying it as a therapeutic target.


Subject(s)
Adaptor Proteins, Signal Transducing , Hippo Signaling Pathway , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adipocytes/metabolism , Adipose Tissue/metabolism , Animals , Dipeptidyl Peptidase 4/metabolism , Fibrosis , Mice , Protein Serine-Threonine Kinases/genetics , Transforming Growth Factor beta/metabolism
16.
Cell Metab ; 34(9): 1325-1341.e6, 2022 09 06.
Article in English | MEDLINE | ID: mdl-35977541

ABSTRACT

Uncoupling protein 1 (UCP1)-mediated adaptive thermogenesis protects mammals against hypothermia and metabolic dysregulation. Whether and how mitochondrial calcium regulates this process remains unclear. Here, we show that mitochondrial calcium uniporter (MCU) recruits UCP1 through essential MCU regulator (EMRE) to form an MCU-EMRE-UCP1 complex upon adrenergic stimulation. This complex formation increases mitochondrial calcium uptake to accelerate the tricarboxylic acid cycle and supply more protons that promote uncoupled respiration, functioning as a thermogenic uniporter. Mitochondrial calcium uptake 1 (MICU1) negatively regulates thermogenesis probably through inhibiting thermogenic uniporter formation. Accordingly, the deletion of Mcu or Emre in brown adipocytes markedly impairs thermogenesis and exacerbates obesity and metabolic dysfunction. Remarkably, the enhanced assembly of the thermogenic uniporter via Micu1 knockout or expressing linked EMRE-UCP1 results in opposite phenotypes. Thus, we have uncovered a "thermoporter" that provides a driving force for the UCP1 operation in thermogenesis, which could be leveraged to combat obesity and associated metabolic disorders.


Subject(s)
Adipose Tissue, Brown , Calcium , Adipose Tissue, Brown/metabolism , Animals , Calcium/metabolism , Calcium Channels , Mammals/metabolism , Mitochondrial Proteins/metabolism , Obesity/metabolism , Thermogenesis/physiology , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism
17.
Front Artif Intell ; 4: 749878, 2021.
Article in English | MEDLINE | ID: mdl-34778753

ABSTRACT

Reinforcement Learning (RL) based machine trading attracts a rich profusion of interest. However, in the existing research, RL in the day-trade task suffers from the noisy financial movement in the short time scale, difficulty in order settlement, and expensive action search in a continuous-value space. This paper introduced an end-to-end RL intraday trading agent, namely QF-TraderNet, based on the quantum finance theory (QFT) and deep reinforcement learning. We proposed a novel design for the intraday RL trader's action space, inspired by the Quantum Price Levels (QPLs). Our action space design also brings the model a learnable profit-and-loss control strategy. QF-TraderNet composes two neural networks: 1) A long short term memory networks for the feature learning of financial time series; 2) a policy generator network (PGN) for generating the distribution of actions. The profitability and robustness of QF-TraderNet have been verified in multi-type financial datasets, including FOREX, metals, crude oil, and financial indices. The experimental results demonstrate that QF-TraderNet outperforms other baselines in terms of cumulative price returns and Sharpe Ratio, and the robustness in the acceidential market shift.

18.
Environ Health Perspect ; 127(11): 117003, 2019 11.
Article in English | MEDLINE | ID: mdl-31724879

ABSTRACT

BACKGROUND: Atherosclerotic cardiovascular disease has become the leading cause of death worldwide, and environmental pollutants are increasingly recognized as risk factors for atherosclerosis. Liver X receptors (LXRs) play a central role in atherosclerosis; however, LXR activity of organic pollutants and associated potential risk of atherosclerosis have not yet been characterized. OBJECTIVES: This study aimed to explore whether LXR-antagonistic chemicals are present in indoor house dust and, if so, to characterize this activity in relation to changes in macrophages in vitro and cardiovascular disease indicators in vivo in an atherosclerosis ApoE-/- mouse model. METHODS: We used a His-LXRα-pull-down assay and a nontarget high-resolution mass spectrometry method to screen house dust collected from Chinese homes for LXRα- and LXRß-antagonist activity. A chemical identified in this manner was assessed for its ability to induce cholesterol efflux and foam cell formation in RAW264.7 macrophages, to down-regulate the expression of two LXR-dependent genes, ABCA1 and ABCG1, and finally to induce atherosclerotic lesions in vivo using an ApoE-/- mouse model. RESULTS: We identified the flame retardants triphenyl phosphate (TPHP) and 2-ethylhexyl diphenyl phosphate (EHDPP) in house dust samples and demonstrated their ability to antagonize LXRs. The potency of TPHP was similar to that of the LXR-antagonist SR9238. TPHP could also inhibit cholesterol efflux and promote foam cell formation in RAW264.7 macrophages and mouse peritoneal macrophages and significantly promoted atherosclerotic lesion formation in the ApoE-/- mouse model. CONCLUSIONS: We found LXR-antagonist chemicals in environmental samples of indoor dust from Chinese homes. One of the chemicals, TPHP, was able to promote the development of atherosclerotic lesions in the ApoE-/- mouse model. These results highlight the need to assess the LXR-antagonist activities of pollutants in future environmental management programs. https://doi.org/10.1289/EHP5039.


Subject(s)
Air Pollutants/adverse effects , Air Pollution, Indoor/analysis , Atherosclerosis/physiopathology , Dust/analysis , Animals , Atherosclerosis/chemically induced , China , Liver X Receptors/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout, ApoE , RAW 264.7 Cells
19.
Sci China Life Sci ; 59(12): 1232-1240, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27837402

ABSTRACT

The prevalence of obesity and type 2 diabetes is escalating to an epidemic proportion worldwide. Obesity is known to be associated with a state of chronic, low-grade inflammation. Emerging lines of evidence have shown that both innate and adaptive immune responses play crucial roles in the control of metabolic homeostasis. Macrophages in adipose tissues are the essential effector cells in orchestrating metabolic inflammation, which is thought to promote the pathogenic progression of obesity and obesity-related disorders. Here we discuss our current understanding of the distinct modes of activation of adipose tissue macrophages, which can sense the metabolic cues and exert profound effects upon adipose homeostasis. Targeting macrophages in adipose tissues may provide new avenues for developing immunomodulation-based therapeutics against obesity and obesity-associated metabolic diseases.


Subject(s)
Adipose Tissue/immunology , Homeostasis/immunology , Inflammation/immunology , Macrophages/immunology , Adipocytes/immunology , Adipose Tissue/metabolism , Animals , Humans , Inflammation/metabolism , Macrophage Activation/immunology , Models, Immunological , Obesity/immunology , Obesity/metabolism
20.
Cell Metab ; 23(1): 165-78, 2016 Jan 12.
Article in English | MEDLINE | ID: mdl-26549485

ABSTRACT

Chronic, low-grade inflammation triggered by excess intake of dietary lipids has been proposed to contribute to the pathogenesis of metabolic disorders, such as obesity, insulin resistance, type 2 diabetes, and atherosclerosis. Although considerable evidence supports a causal association between inflammation and metabolic diseases, most tests of this link have been performed in cold-stressed mice that are housed below their thermoneutral zone. We report here that thermoneutral housing of mice has a profound effect on the development of metabolic inflammation, insulin resistance, and atherosclerosis. Mice housed at thermoneutrality develop metabolic inflammation in adipose tissue and in the vasculature at an accelerated rate. Unexpectedly, this increased inflammatory response contributes to the progression of atherosclerosis but not insulin resistance. These findings not only suggest that metabolic inflammation can be uncoupled from obesity-associated insulin resistance, but also point to how thermal stress might limit our ability to faithfully model human diseases in mice.


Subject(s)
Atherosclerosis/immunology , Environmental Exposure , Insulin Resistance/immunology , Adaptive Immunity , Adipose Tissue, White/immunology , Adipose Tissue, White/pathology , Animals , Aorta, Thoracic/immunology , Aorta, Thoracic/pathology , Apolipoproteins E/genetics , Atherosclerosis/etiology , Diet, High-Fat/adverse effects , Housing, Animal , Immunity, Innate , Inflammation/immunology , Macrophages/immunology , Male , Mice, Inbred C57BL , Mice, Knockout
SELECTION OF CITATIONS
SEARCH DETAIL