Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 160
Filter
1.
J Cell Mol Med ; 25(20): 9837-9850, 2021 10.
Article in English | MEDLINE | ID: mdl-34528389

ABSTRACT

The inflammation of adipose tissue is one of the most common secondary pathological changes in atherosclerosis, which in turn influences the process of atherosclerosis. Natriuretic peptides have been revealed important effect in regulating adipose metabolism. However, the relationship between natriuretic peptide receptor C and inflammation of adipose tissue in atherosclerosis remains unknown. This study aims to explore the effect natriuretic peptide receptor C exerts on the regulation of the adipose inflammation in atherosclerotic mice induced by western-type diet and its overlying mechanisms. To clarify the importance of NPRC of adipose inflammation in atherosclerotic mice, NPRC expression was measured in mice fed with chow diet and western-type diet for 12 weeks and we found a considerable increase in adipose tissue of atherosclerotic mice. Global NPRC knockout in mice was bred onto ApoE-/- mice to generate NPRC-/- ApoE-/- mice, which displayed remarked increase in browning of white adipose tissue and lipolysis of adipose tissue and decrease in adipose inflammation manifested by decreased macrophage invasion to form less CLS (crown-like structure), reduced oxidative stress and alleviated expression of TNFα, IL-6, IL-1ß and MCP1, but increased expression of adiponectin in adipose tissue. Moreover, our study showed that white adipose tissue browning in NPRC-/- ApoE-/- atherosclerotic mice was associated with decreased inflammatory response through cAMP/PKA signalling activation. These results identify NPRC as a novel regulator for adipose inflammation in atherosclerotic mice by modulating white adipose tissue browning.


Subject(s)
Apolipoproteins E/deficiency , Hypercholesterolemia/complications , Panniculitis/etiology , Panniculitis/metabolism , Receptors, Atrial Natriuretic Factor/deficiency , Animals , Biomarkers , Cyclic AMP , Cyclic AMP-Dependent Protein Kinases/metabolism , Disease Models, Animal , Disease Susceptibility , Gene Expression Regulation , Hypercholesterolemia/genetics , Hypercholesterolemia/metabolism , Immunohistochemistry , Inflammasomes/metabolism , Mice , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Oxidative Stress , Panniculitis/pathology , Signal Transduction
2.
Arterioscler Thromb Vasc Biol ; 41(10): 2563-2574, 2021 10.
Article in English | MEDLINE | ID: mdl-34348490

ABSTRACT

Objective: The accumulation of inflammatory leukocytes is a prerequisite of adipose tissue inflammation during cardiometabolic disease. We previously reported that a genetic deficiency of the intracellular signaling adaptor TRAF5 (TNF [tumor necrosis factor] receptor-associated factor 5) accelerates atherosclerosis in mice by increasing inflammatory cell recruitment. Here, we tested the hypothesis that an impairment of TRAF5 signaling modulates adipose tissue inflammation and its metabolic complications in a model of diet-induced obesity in mice. Approach and Results: To induce diet-induced obesity and adipose tissue inflammation, wild-type or Traf5-/- mice consumed a high-fat diet for 18 weeks. Traf5-/- mice showed an increased weight gain, impaired insulin tolerance, and increased fasting blood glucose. Weight of livers and peripheral fat pads was increased in Traf5-/- mice, whereas lean tissue weight and growth were not affected. Flow cytometry of the stromal vascular fraction of visceral adipose tissue from Traf5-/- mice revealed an increase in cytotoxic T cells, CD11c+ macrophages, and increased gene expression of proinflammatory cytokines and chemokines. At the level of cell types, expression of TNF[alpha], MIP (macrophage inflammatory protein)-1[alpha], MCP (monocyte chemoattractant protein)-1, and RANTES (regulated on activation, normal T-cell expressed and secreted) was significantly upregulated in Traf5-deficient adipocytes but not in Traf5-deficient leukocytes from visceral adipose tissue. Finally, Traf5 expression was lower in adipocytes from obese patients and mice and recovered in adipose tissue of obese patients one year after bariatric surgery. Conclusions: We show that a genetic deficiency of TRAF5 in mice aggravates diet-induced obesity and its metabolic derangements by a proinflammatory response in adipocytes. Our data indicate that TRAF5 may promote anti-inflammatory and obesity-preventing signaling events in adipose tissue.


Subject(s)
Adipocytes/metabolism , Adipose Tissue/metabolism , Cytokines/metabolism , Inflammation Mediators/metabolism , Lymphocytes/metabolism , Obesity/metabolism , Panniculitis/metabolism , TNF Receptor-Associated Factor 5/deficiency , Adipocytes/immunology , Adipocytes/pathology , Adipose Tissue/immunology , Adipose Tissue/pathology , Adiposity , Adult , Aged , Animals , Diet, High-Fat , Disease Models, Animal , Female , Humans , Lymphocytes/immunology , Macrophages/immunology , Macrophages/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Obesity/genetics , Obesity/immunology , Obesity/pathology , Panniculitis/genetics , Panniculitis/immunology , Panniculitis/pathology , Signal Transduction , TNF Receptor-Associated Factor 5/genetics
3.
J Mol Endocrinol ; 67(3): 55-69, 2021 07 26.
Article in English | MEDLINE | ID: mdl-34213423

ABSTRACT

Pentraxin 3 (PTX3) is a soluble pattern recognition receptor playing an important role in immune response and inflammation. Lipopolysaccharide (LPS) stimulation can significantly induce PTX3 expression and secretion in adipocytes. Appropriate regulation of PTX3 secretion is critical for inflammatory homeostasis. Using chemical inhibitors of conventional and unconventional protein secretion, we explored the mechanisms that control LPS-stimulated PTX3 secretion in 3T3-L1 adipocytes. Inhibiting the conventional protein secretion blocked LPS-stimulated PTX3 secretion, resulting in cellular PTX3 accumulation in adipocytes. We also detected PTX3 in exosomes from LPS-treated adipocytes; inhibiting exosome trafficking attenuated PTX3 secretion. However, only 4.3% of secreted PTX3 was detected in exosomes compared to 95.7% in the non-exosomal fractions. The fractionation of isolated exosomes by the iodixanol density gradient centrifugation confirmed that a small portion of secreted PTX3 overlapped with exosomal markers in small extracellular-vesicle fractions. We conclude that PTX3 is secreted mainly through conventional protein secretion, and a small percentage of PTX3 is released in exosomes from LPS-stimulated adipocytes.


Subject(s)
Adipocytes/metabolism , Biomarkers , C-Reactive Protein/biosynthesis , Panniculitis/metabolism , Serum Amyloid P-Component/biosynthesis , 3T3-L1 Cells , Adipocytes/drug effects , Animals , C-Reactive Protein/chemistry , C-Reactive Protein/genetics , Cells, Cultured , Endosomal Sorting Complexes Required for Transport/metabolism , Extracellular Vesicles/metabolism , Gene Expression Regulation , Lipopolysaccharides/immunology , Lipopolysaccharides/pharmacology , Macrophages/immunology , Macrophages/metabolism , Mice , Mitochondria/genetics , Mitochondria/metabolism , Panniculitis/etiology , Protein Interaction Domains and Motifs , Protein Sorting Signals , Serum Amyloid P-Component/chemistry , Serum Amyloid P-Component/genetics
4.
Molecules ; 26(5)2021 Mar 05.
Article in English | MEDLINE | ID: mdl-33807712

ABSTRACT

BACKGROUND: Tart cherries (Prunus cerasus L.) are a rich source of anthocyanins. They are phytochemical flavonoids found in red and blue fruits, and vegetables that can reduce hyperlipidemia. Visceral Adipose Tissue (VAT) has emerged as a major player in driving obesity-related inflammatory response. METHODS: This study has investigated the potential positive effects of tart cherries on rats with Diet-Induced Obesity (DIO). In particular, the inflammatory status in retroperitoneal (RPW) and perigonadal (PGW) adipose tissue were studied. Rats were fed ad libitum for 17 weeks with a hypercaloric diet with the supplementation of tart cherries seeds powder (DS) and seeds powder plus tart cherries juice containing 1mg of anthocyanins (DJS). In RPW and PGW, expression of CRP, IL-1 ß, TNF-α, CCL2 and CD36, were measured by qRT-PCR, Western blot and immunohistochemistry techniques. RESULTS: No differences in the weight of RPW and PGW animals were found between DS and DJS groups compared to DIO rats. However, an increase of inflammatory markers was observed in DIO group in comparison with control lean rats. A modulation of these markers was evident upon tart cherry supplementation. CONCLUSION: Study results suggest that tart cherry enriched-diet did not modify the accumulation of visceral fat, but it decreased inflammatory markers in both tissues. Therefore, this supplementation could be useful, in combination with healthy lifestyles, to modify adipose tissue cell metabolism limiting-obesity related organ damage.


Subject(s)
Biomarkers/metabolism , Fruit and Vegetable Juices , Intra-Abdominal Fat/metabolism , Obesity/diet therapy , Prunus avium/chemistry , Animals , CD36 Antigens/genetics , CD36 Antigens/metabolism , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Diet, High-Fat/adverse effects , Dietary Supplements , Gene Expression Regulation , Intra-Abdominal Fat/drug effects , Macrophages/drug effects , Macrophages/pathology , Male , Obesity/etiology , Panniculitis/diet therapy , Panniculitis/genetics , Panniculitis/metabolism , Rats, Wistar , Seeds
5.
Mol Metab ; 48: 101220, 2021 06.
Article in English | MEDLINE | ID: mdl-33774223

ABSTRACT

OBJECTIVE: Neuroimmune interactions between the sympathetic nervous system (SNS) and macrophages are required for the homeostasis of multiple tissues, including the adipose tissue. It has been proposed that the SNS maintains adipose tissue macrophages (ATMs) in an anti-inflammatory state via direct norepinephrine (NE) signaling to macrophages. This study aimed to investigate the physiological importance of this paradigm by utilizing a mouse model in which the adrenergic signaling from the SNS to macrophages, but not to other adipose tissue cells, was disrupted. METHODS: We generated a macrophage-specific B2AR knockout mouse (Adrb2ΔLyz2) by crossing Adrb2fl/fl and Lyz2Cre/+ mice. We have previously shown that macrophages isolated from Adrb2ΔLyz2 animals do not respond to NE stimulation in vitro. Herein we performed a metabolic phenotyping of Adrb2ΔLyz2 mice on either chow or high-fat diet (HFD). We also assessed the adipose tissue function of Adrb2ΔLyz2 animals during fasting and cold exposure. Finally, we transplanted Adrb2ΔLyz2 bone marrow to low-density lipoprotein receptor (LDLR) knockout mice and investigated the development of atherosclerosis during Western diet feeding. RESULTS: We demonstrated that SNS-associated ATMs have a transcriptional profile indicative of activated beta-2 adrenergic receptor (B2AR), the main adrenergic receptor isoform in myeloid cells. However, Adrb2ΔLyz2 mice have unaltered energy balance on a chow or HFD. Furthermore, Adrb2ΔLyz2 mice show similar levels of adipose tissue inflammation and function during feeding, fasting, or cold exposure, and develop insulin resistance during HFD at the same rate as controls. Finally, macrophage-specific B2AR deletion does not affect the development of atherosclerosis on an LDL receptor-null genetic background. CONCLUSIONS: Overall, our data suggest that the SNS does not directly modulate the phenotype of adipose tissue macrophages in either lean mice or mouse models of cardiometabolic disease. Instead, sympathetic nerve activity exerts an indirect effect on adipose tissue macrophages through the modulation of adipocyte function.


Subject(s)
Atherosclerosis/complications , Atherosclerosis/metabolism , Insulin Resistance/genetics , Macrophages/metabolism , Obesity/complications , Obesity/metabolism , Panniculitis/metabolism , Receptors, Adrenergic, beta-2/metabolism , Signal Transduction/genetics , Adipocytes/metabolism , Adipose Tissue, White/metabolism , Animals , Atherosclerosis/genetics , Bone Marrow Transplantation/methods , Cells, Cultured , Diet, High-Fat/adverse effects , Diet, Western/adverse effects , Disease Models, Animal , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/genetics , Panniculitis/genetics , Phenotype , Receptors, Adrenergic, beta-2/genetics , Sympathetic Nervous System/metabolism
6.
Cell Death Dis ; 12(2): 212, 2021 02 26.
Article in English | MEDLINE | ID: mdl-33637697

ABSTRACT

Obesity is caused by a long-term imbalance between energy intake and consumption and is regulated by multiple signals. This study investigated the effect of signaling scaffolding protein Gab2 on obesity and its relevant regulation mechanism. Gab2 knockout (KO) and wild-type (WT) mice were fed with a standard diet (SD) or high-fat diet (HFD) for 12 weeks. The results showed that the a high-fat diet-induced Gab2 expression in adipose tissues, but deletion of Gab2 attenuated weight gain and improved glucose tolerance in mice fed with a high-fat diet. White adipose tissue and systemic inflammations were reduced in HFD-fed Gab2 deficiency mice. Gab2 deficiency increased the expression of Ucp1 and other thermogenic genes in brown adipose tissue. Furthermore, the regulation of Gab2 on the mature differentiation and function of adipocytes was investigated in vitro using primary or immortalized brown preadipocytes. The expression of brown fat-selective genes was found to be elevated in differentiated adipocytes without Gab2. The mechanism of Gab2 regulating Ucp1 expression in brown adipocytes involved with its downstream PI3K (p85)-Akt-FoxO1 signaling pathway. Our research suggests that deletion of Gab2 suppresses diet-induced obesity by multiple pathways and Gab2 may be a novel therapeutic target for the treatment of obesity and associated complications.


Subject(s)
Adaptor Proteins, Signal Transducing/deficiency , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Energy Metabolism , Obesity/prevention & control , Panniculitis/prevention & control , Adaptor Proteins, Signal Transducing/genetics , Adipose Tissue, Brown/physiopathology , Adipose Tissue, White/physiopathology , Adiposity , Animals , Blood Glucose/metabolism , Cell Line , Class Ia Phosphatidylinositol 3-Kinase/metabolism , Diet, High-Fat , Disease Models, Animal , Forkhead Box Protein O1/metabolism , Insulin Resistance , Male , Mice, Inbred C57BL , Mice, Knockout , Obesity/genetics , Obesity/metabolism , Obesity/physiopathology , Panniculitis/genetics , Panniculitis/metabolism , Panniculitis/physiopathology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Uncoupling Protein 1/metabolism , Weight Gain
7.
Arterioscler Thromb Vasc Biol ; 41(1): 284-301, 2021 01.
Article in English | MEDLINE | ID: mdl-33054397

ABSTRACT

OBJECTIVE: Smooth muscle cells and pericytes display remarkable plasticity during injury and disease progression. Here, we tested the hypothesis that perivascular cells give rise to Klf4-dependent macrophage-like cells that augment adipose tissue (AT) inflammation and metabolic dysfunction associated with diet-induced obesity (DIO). Approach and Results: Using Myh11-CreERT2 eYFP (enhanced yellow fluorescent protein) mice and flow cytometry of the stromovascular fraction of epididymal AT, we observed a large fraction of smooth muscle cells and pericytes lineage traced eYFP+ cells expressing macrophage markers. Subsequent single-cell RNA sequencing, however, showed that the majority of these cells had no detectable eYFP transcript. Further exploration revealed that intraperitoneal injection of tamoxifen in peanut oil, used for generating conditional knockout or reporter mice in thousands of previous studies, resulted in large increase in the autofluorescence and false identification of macrophages within epididymal AT as being eYFP+; and unintended proinflammatory consequences. Using newly generated Myh11-DreERT2tdTomato mice given oral tamoxifen, we virtually eliminated the problem with autofluorescence and identified 8 perivascular cell dominated clusters, half of which were altered upon DIO. Given that perivascular cell KLF4 (kruppel-like factor 4) can have beneficial or detrimental effects, we tested its role in obesity-associated AT inflammation. While smooth muscle cells and pericytes-specific Klf4 knockout (smooth muscle cells and pericytes Klf4Δ/Δ) mice were not protected from DIO, they displayed improved glucose tolerance upon DIO, and showed marked decreases in proinflammatory macrophages and increases in LYVE1+ lymphatic endothelial cells in the epididymal AT. CONCLUSIONS: Perivascular cells within the AT microvasculature dynamically respond to DIO and modulate tissue inflammation and metabolism in a KLF4-dependent manner.


Subject(s)
Adipose Tissue/metabolism , Cell Plasticity , Kruppel-Like Transcription Factors/metabolism , Myocytes, Smooth Muscle/metabolism , Obesity/metabolism , Panniculitis/metabolism , Pericytes/metabolism , Adipose Tissue/pathology , Animals , Blood Glucose/metabolism , Cell Lineage , Diet, High-Fat , Disease Models, Animal , Endothelial Cells/metabolism , Endothelial Cells/pathology , Inflammation Mediators/metabolism , Insulin Resistance , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/deficiency , Kruppel-Like Transcription Factors/genetics , Macrophages/metabolism , Macrophages/pathology , Male , Mice, Knockout , Myocytes, Smooth Muscle/pathology , Obesity/etiology , Obesity/genetics , Obesity/pathology , Panniculitis/etiology , Panniculitis/genetics , Panniculitis/pathology , Pericytes/pathology
8.
Arterioscler Thromb Vasc Biol ; 41(2): 796-807, 2021 02.
Article in English | MEDLINE | ID: mdl-33380173

ABSTRACT

OBJECTIVE: The aim of this study was to unravel mechanisms whereby deficiency of the transcription factor Id3 (inhibitor of differentiation 3) leads to metabolic dysfunction in visceral obesity. We investigated the impact of loss of Id3 on hyaluronic acid (HA) production by the 3 HAS isoenzymes (HA synthases; -1, -2, and -3) and on obesity-induced adipose tissue (AT) accumulation of proinflammatory B cells. Approach and Results: Male Id3-/- mice and respective wild-type littermate controls were fed a 60% high-fat diet for 4 weeks. An increase in inflammatory B2 cells was detected in Id3-/- epididymal AT. HA accumulated in epididymal AT of high-fat diet-fed Id3-/- mice and circulating levels of HA were elevated. Has2 mRNA expression was increased in epididymal AT of Id3-/- mice. Luciferase promoter assays showed that Id3 suppressed Has2 promoter activity, while loss of Id3 stimulated Has2 promoter activity. Functionally, HA strongly promoted B2 cell adhesion in the AT and on cultured vascular smooth muscle cells of Id3-/- mice, an effect sensitive to hyaluronidase. CONCLUSIONS: Our data demonstrate that loss of Id3 increases Has2 expression in the epididymal AT, thereby promoting HA accumulation. In turn, elevated HA content promotes HA-dependent binding of B2 cells and an increase in the B2 cells in the AT, which contributes to AT inflammation.


Subject(s)
Adipose Tissue/metabolism , B-Lymphocytes/metabolism , Hyaluronan Synthases/metabolism , Hyaluronic Acid/biosynthesis , Inhibitor of Differentiation Proteins/metabolism , Panniculitis/metabolism , Adipose Tissue/immunology , Animals , B-Lymphocytes/immunology , Cell Adhesion , Cells, Cultured , Coculture Techniques , Diet, High-Fat , Disease Models, Animal , Hyaluronan Synthases/genetics , Inhibitor of Differentiation Proteins/genetics , Macrophages/immunology , Macrophages/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/immunology , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/immunology , Myocytes, Smooth Muscle/metabolism , Panniculitis/genetics , Panniculitis/immunology , Phenotype , Signal Transduction , Up-Regulation
9.
Oxid Med Cell Longev ; 2020: 8882763, 2020.
Article in English | MEDLINE | ID: mdl-33312340

ABSTRACT

This study was conducted to assess the protective effect of extract of match (EM) on high-fat diet- (HFD-) induced cognitive deficits in male C57BL/6 mice. It was found that EM improved glucose tolerance status by measuring OGTT and IPGTT with HFD-induced mice. EM protected behavioral and memory dysfunction in Y-maze, passive avoidance, and Morris water maze tests. Consumption of EM reduced fat mass, dyslipidemia, and inflammation in adipose tissue. Also, EM ameliorated hepatic and cerebral antioxidant systems. EM improved the cerebral cholinergic system by regulating ACh contents and expression of AChE and ChAT. Also, EM restored mitochondrial function in liver and brain tissue. EM attenuated hepatic inflammatory effect, lipid synthesis, and cholesterol metabolism by regulating the protein expression of TNF-α, TNFR1, p-IRS-1, p-JNK, IL-1ß, iNOS, COX-2, HMGCR, PPARγ, and FAS. Finally, EM regulated cognitive function and neuroinflammation in the whole brain, hippocampus, and cerebral cortex by regulating the protein expression of p-JNK, p-Akt, p-tau, Aß, BDNF, IDE, COX-2, and IL-1ß. These findings suggest that EM might be a potential source of functional food to improve metabolic disorder-associated cognitive dysfunction.


Subject(s)
Cognitive Dysfunction , Diet, High-Fat/adverse effects , Dyslipidemias , Memory Disorders , Panniculitis , Tea , Adipose Tissue/metabolism , Adipose Tissue/pathology , Animals , Cognitive Dysfunction/chemically induced , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/pathology , Cognitive Dysfunction/therapy , Dyslipidemias/chemically induced , Dyslipidemias/metabolism , Dyslipidemias/pathology , Dyslipidemias/therapy , Gene Expression Regulation , Inflammation/chemically induced , Inflammation/metabolism , Inflammation/pathology , Inflammation/therapy , Male , Memory Disorders/chemically induced , Memory Disorders/metabolism , Memory Disorders/pathology , Memory Disorders/therapy , Mice , Panniculitis/chemically induced , Panniculitis/metabolism , Panniculitis/pathology , Panniculitis/therapy
10.
Int J Mol Sci ; 21(17)2020 Aug 23.
Article in English | MEDLINE | ID: mdl-32842547

ABSTRACT

Endometrial cancer (EC) is one of the most common malignancies of the female reproductive organs. The most characteristic feature of EC is the frequent association with metabolic disorders. However, the components of these disorders that are involved in carcinogenesis remain unclear. Accumulating epidemiological studies have clearly revealed that hyperinsulinemia, which accompanies these disorders, plays central roles in the development of EC via the insulin-phosphoinositide 3 kinase (PI3K) signaling pathway as a metabolic driver. Recent comprehensive genomic analyses showed that over 90% of ECs have genomic alterations in this pathway, resulting in enhanced insulin signaling and production of optimal tumor microenvironments (TMEs). Targeting PI3K signaling is therefore an attractive treatment strategy. Several clinical trials for recurrent or advanced ECs have been attempted using PI3K-serine/threonine kinase (AKT) inhibitors. However, these agents exhibited far lower efficacy than expected, possibly due to activation of alternative pathways that compensate for the PIK3-AKT pathway and allow tumor growth, or due to adaptive mechanisms including the insulin feedback pathway that limits the efficacy of agents. Overcoming these responses with careful management of insulin levels is key to successful treatment. Further interest in specific TMEs via the insulin PI3K-pathway in obese women will provide insight into not only novel therapeutic strategies but also preventive strategies against EC.


Subject(s)
Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/metabolism , Glucose/metabolism , Obesity/complications , Phosphatidylinositol 3-Kinase/metabolism , Diabetes Complications/etiology , Drug Resistance, Neoplasm/drug effects , Endometrial Neoplasms/genetics , Estrogens/metabolism , Female , Humans , Insulin Resistance , Metformin/pharmacology , Obesity/metabolism , Panniculitis/complications , Panniculitis/metabolism , Phosphatidylinositol 3-Kinase/genetics , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Risk Factors , Signal Transduction , Tumor Microenvironment
11.
Adipocyte ; 9(1): 325-334, 2020 12.
Article in English | MEDLINE | ID: mdl-32603641

ABSTRACT

Adipocyte-mediated inflammatory signalling has been proposed to alter adipose physiology in obesity and Type 2 diabetes mellitus. Novel targets for alteration of inflammatory signalling are needed to improve obesity-related outcomes. The γ-secretase enzyme complex has been suggested to play a role both in adipocyte function as well as in immune regulation. We hypothesized that adipocyte-specific γ-secretase inhibition could alter the inflammatory makeup of adipose tissue. We found that genetic blockade of γ-secretase in adipocytes leads to a decrease in EMR1 (F4/80) expression, as a marker of macrophage presence, in adipose tissue without changes in expression of markers of other inflammatory cell types. To explore the mechanism by which adipocytes can alter macrophage function in vitro, fully differentiated 3T3-L1 adipocytes were treated with a γ-secretase inhibitor in the presence of lipopolysaccharide (LPS) and transcription of IL6 and ccl2 (MCP1) were quantified. IL-6 expression and secretion were significantly inhibited by γ-secretase blockade, with little effect on MCP1. Preconditioned media from 3T3-L1 adipocytes treated with a γ-secretase inhibitor also alters macrophage activation but did not affect macrophage translocation in vitro. Therefore, γ-secretase inhibition in fully differentiated adipocytes can alter IL-6 signalling to macrophages, consistent with our hypothesis that that γ-secretase is involved in adipocyte-initiated inflammatory signalling cascades.


Subject(s)
Adipocytes/metabolism , Adipose Tissue/metabolism , Amyloid Precursor Protein Secretases/metabolism , Interleukin-6/biosynthesis , Panniculitis/metabolism , 3T3-L1 Cells , Adipose Tissue/pathology , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Animals , Biomarkers , Cytokines/metabolism , Inflammation Mediators/metabolism , Lipopolysaccharides/immunology , Macrophage Activation/immunology , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Mice , Panniculitis/etiology , Panniculitis/pathology , Protease Inhibitors/pharmacology , Signal Transduction
12.
Int J Mol Sci ; 21(11)2020 Jun 11.
Article in English | MEDLINE | ID: mdl-32545355

ABSTRACT

Adipose tissue is an active endocrine and immune organ that controls systemic immunometabolism via multiple pathways. Diverse immune cell populations reside in adipose tissue, and their composition and immune responses vary with nutritional and environmental conditions. Adipose tissue dysfunction, characterized by sterile low-grade chronic inflammation and excessive immune cell infiltration, is a hallmark of obesity, as well as an important link to cardiometabolic diseases. Amongst the pro-inflammatory factors secreted by the dysfunctional adipose tissue, interleukin (IL)-1ß, induced by the NLR family pyrin domain-containing 3 (NLRP3) inflammasome, not only impairs peripheral insulin sensitivity, but it also interferes with the endocrine and immune functions of adipose tissue in a paracrine manner. Human studies indicated that NLRP3 activity in adipose tissues positively correlates with obesity and its metabolic complications, and treatment with the IL-1ß antibody improves glycaemia control in type 2 diabetic patients. In mouse models, genetic or pharmacological inhibition of NLRP3 activation pathways or IL-1ß prevents adipose tissue dysfunction, including inflammation, fibrosis, defective lipid handling and adipogenesis, which in turn alleviates obesity and its related metabolic disorders. In this review, we summarize both the negative and positive regulators of NLRP3 inflammasome activation, and its pathophysiological consequences on immunometabolism. We also discuss the potential therapeutic approaches to targeting adipose tissue inflammasome for the treatment of obesity and its related metabolic disorders.


Subject(s)
Adipose Tissue/metabolism , Inflammasomes/metabolism , Metabolic Diseases/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Adipokines/metabolism , Adipose Tissue/drug effects , Adipose Tissue/pathology , Animals , Autophagy , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Humans , Insulin Resistance , Lipid Metabolism , Lipopolysaccharides/pharmacology , Metabolic Diseases/drug therapy , Metabolic Diseases/pathology , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Obesity/metabolism , Obesity/pathology , Panniculitis/metabolism , Panniculitis/pathology
13.
Mol Metab ; 39: 101008, 2020 09.
Article in English | MEDLINE | ID: mdl-32389828

ABSTRACT

OBJECTIVE: Glucose-dependent insulinotropic polypeptide (GIP) conveys information from ingested nutrients to peripheral tissues, signaling energy availability. The GIP Receptor (GIPR) is also expressed in the bone marrow, notably in cells of the myeloid lineage. However, the importance of gain and loss of GIPR signaling for diverse hematopoietic responses remains unclear. METHODS: We assessed the expression of the Gipr in bone marrow (BM) lineages and examined functional roles for the GIPR in control of hematopoiesis. Bone marrow responses were studied in (i) mice fed regular or energy-rich diets, (ii) mice treated with hematopoietic stressors including acute 5-fluorouracil (5-FU), pamsaccharide (LPS), and Pam3CysSerLys4 (Pam3CSK4), with or without pharmacological administration of a GIPR agonist, and (iii) mice with global (Gipr-/-) or selective deletion of the GIPR (GiprTie2-/-) with and without bone marrow transplantation (BMT). RESULTS: Gipr is expressed within T cells, myeloid cells, and myeloid precursors; however, these cell populations were not different in peripheral blood, spleen, or BM of Gipr-/- and GiprTie2-/- mice. Nevertheless, gain and loss of function studies revealed that GIPR signaling controls the expression of BM Toll-like receptor (TLR) and Notch-related genes regulating hematopoiesis. Loss of the BM GIPR attenuates the extent of adipose tissue inflammation and dysregulates the hematopoietic response to BMT. GIPR agonism modified BM gene expression profiles following 5-FU and Pam3CSK4 whereas loss of the Gipr altered the hematopoietic responses to energy excess, two TLR ligands, and 5-FU. However, the magnitude of the cellular changes in hematopoiesis in response to gain or loss of GIPR signaling was relatively modest. CONCLUSION: These studies identify a functional gut hormone-BM axis positioned for the transduction of signals linking nutrient availability to the control of TLR and Notch genes regulating hematopoiesis. Nevertheless, stimulation or loss of GIPR signaling has minimal impact on basal hematopoiesis or the physiological response to hematopoietic stress.


Subject(s)
Energy Metabolism/genetics , Hematopoiesis/genetics , Receptors, Gastrointestinal Hormone/genetics , Adipose Tissue/metabolism , Animals , Biomarkers , Body Composition , Bone Marrow Cells/metabolism , Fluorouracil/pharmacology , Gene Expression , Gene Expression Regulation , Lipopolysaccharides/immunology , Mice , Mice, Knockout , Panniculitis/etiology , Panniculitis/metabolism , Panniculitis/pathology , Receptors, Gastrointestinal Hormone/agonists , Receptors, Gastrointestinal Hormone/metabolism , Receptors, Notch/genetics , Receptors, Notch/metabolism , Signal Transduction , Toll-Like Receptors/genetics , Toll-Like Receptors/metabolism
14.
Pediatr Blood Cancer ; 67(6): e28302, 2020 06.
Article in English | MEDLINE | ID: mdl-32285995

ABSTRACT

This report offers novel clinical and diagnostic aspects of the association between germline mutations in HAVCR2 and subcutaneous panniculitis-like T-cell lymphoma (SPTCL). The patient presented with panniculitis-like T-cell lymphoma involving mesenteric fatty tissue associated with hemophagocytic lymphohistiocytosis (HLH). Five years later, he developed a clonally unrelated SPTCL and underwent hematopoietic stem cell transplantation. Retrospectively, he was found to carry germline mutations in HAVCR2 associated with reduced T-cell immunoglobulin mucin-3 (TIM-3) expression. We show that mesenteric fatty tissue localization of SPTCL can be the presenting manifestation of TIM-3 deficiency, that this condition predisposes to recurrent lymphoma, and that flow cytometry is a possible screening tool.


Subject(s)
Germ-Line Mutation , Hepatitis A Virus Cellular Receptor 2/deficiency , Hepatitis A Virus Cellular Receptor 2/genetics , Lymphohistiocytosis, Hemophagocytic/pathology , Lymphoma, T-Cell/pathology , Mesentery/pathology , Panniculitis/pathology , Adolescent , Humans , Lymphohistiocytosis, Hemophagocytic/complications , Lymphohistiocytosis, Hemophagocytic/genetics , Lymphohistiocytosis, Hemophagocytic/metabolism , Lymphoma, T-Cell/complications , Lymphoma, T-Cell/genetics , Lymphoma, T-Cell/metabolism , Male , Mesentery/metabolism , Panniculitis/complications , Panniculitis/genetics , Panniculitis/metabolism , Prognosis
15.
Mol Nutr Food Res ; 64(11): e1901166, 2020 06.
Article in English | MEDLINE | ID: mdl-32281732

ABSTRACT

SCOPE: Low-calorie sweetener (LCS) consumption is associated with metabolic disease in observational studies. However, physiologic mechanisms underlying LCS-induced metabolic impairments in humans are unclear. This study is aimed at identifying molecular pathways in adipose impacted by LCSs. METHODS AND RESULTS: Seven females with overweight or obesity, who did not report LCS use, consumed 12 ounces of diet soda containing sucralose and acesulfame-potassium (Ace-K) three times daily for 8 weeks. A subcutaneous adipose biopsy from the left abdomen and a fasting blood sample were collected at baseline and post-intervention. Global gene expression were assessed using RNA-sequencing followed by functional pathway analysis. No differences in circulating metabolic or inflammatory biomarkers were observed. However, ANOVA detected 828 differentially expressed annotated genes after diet soda consumption (p < 0.05), including transcripts for inflammatory cytokines. Fifty-eight of 140 canonical pathways represented in pathway analyses regulated inflammation, and several key upstream regulators of inflammation (e.g., TNF-alpha) were also represented. CONCLUSION: Consumption of diet soda with sucralose and Ace-K alters inflammatory transcriptomic pathways (e.g., NF-κB signaling) in subcutaneous adipose tissue but does not significantly alter circulating biomarkers. Findings highlight the need to examine molecular and metabolic effects of LCS exposure in a larger randomized control trial for a longer duration.


Subject(s)
Adipose Tissue/drug effects , Artificially Sweetened Beverages/adverse effects , Sucrose/analogs & derivatives , Thiazines/adverse effects , Adipose Tissue/physiology , Female , Gene Expression Regulation/drug effects , Humans , Obesity/metabolism , Obesity/physiopathology , Panniculitis/chemically induced , Panniculitis/immunology , Panniculitis/metabolism , Sucrose/adverse effects , Sweetening Agents/adverse effects , Young Adult
16.
Mol Metab ; 39: 100983, 2020 09.
Article in English | MEDLINE | ID: mdl-32229247

ABSTRACT

OBJECTIVE: Expansion of visceral adipose tissue (VAT) and metabolic inflammation are consequences of obesity and associated with type 2 diabetes (T2DM). Metabolically activated adipose tissue macrophages (ATMs) undergo qualitative and quantitative changes that influence their inflammatory responses. How these cells contribute to insulin resistance (IR) in humans is not well understood. Cholesterol 25-Hydroxylase (CH25H) converts cholesterol into 25-Hydroxycholesterol (25-HC), an oxysterol that modulates immune responses. Using human and murine models, we investigated the role of CH25H in metabolic inflammation. METHODS: We performed transcriptomic (RNASeq) analysis on the human whole AT biopsies and sorted ATMs from obese non-diabetic (NDM) and obese diabetic (DM) subjects to inquire if CH25H was increased in DM. We challenged mice lacking Ch25h with a high-fat diet (HFD) to characterize their metabolic and immunologic profiling. Ch25h KO mice and human adipose tissue biopsies from NDM and DM subjects were analyzed. LC-MS was conducted to measure 25-HC level in AT. In vitro analysis permitted us to investigate the effect of 25-HC on cytokine expression. RESULTS: In our RNASeq analysis of human visceral and subcutaneous biopsies, gene pathways related to inflammation were increased in obese DM vs. non-DM subjects that included CH25H. CH25H was enriched in the stromal vascular fraction of human adipose tissue and highly expressed in CD206+ human ATMs by flow cytometry analysis. We measured the levels of the oxysterols, 25-HC and 7α25diHC, in human visceral adipose tissue samples and showed a correlation between BMI and 25-HC. Using mouse models of diet-induced obesity (DIO), we found that HFD-induced Ch25h expression in eWAT and increased levels of 25-HC in AT. On HFD, Ch25h KO mice became obese but exhibited reduced plasma insulin levels, improved insulin action, and decreased ectopic lipid deposit. Improved insulin sensitivity in Ch25h KO mice was due to attenuation of CD11c+ adipose tissue macrophage infiltration in eWAT. Finally, by testing AT explants, bone marrow-derived macrophages (BMDMs) and SVF cells from Ch25h deficient mice, we observed that 25-HC is required for the expression of pro-inflammatory genes. 25-HC was also able to induce inflammatory genes in preadipocytes. CONCLUSIONS: Our data suggest a critical role for CH25H/25-HC in the progression of meta-inflammation and insulin resistance in obese humans and mouse models of obesity. In response to obesogenic stimuli, CH25H/25-HC could exert a pro-inflammatory role.


Subject(s)
Adipose Tissue/metabolism , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Obesity/complications , Obesity/metabolism , Panniculitis/etiology , Steroid Hydroxylases/metabolism , 3T3-L1 Cells , Adult , Animals , Biomarkers , Cytokines/metabolism , Diabetes Mellitus, Type 2/diagnosis , Disease Models, Animal , Disease Susceptibility , Female , Gene Expression Profiling , Gene Expression Regulation , Humans , Insulin Resistance/genetics , Macrophages/immunology , Macrophages/metabolism , Male , Metabolome , Mice , Mice, Knockout , Middle Aged , Obesity/diagnosis , Panniculitis/metabolism , Panniculitis/pathology , Sequence Analysis, RNA , Signal Transduction , Steroid Hydroxylases/genetics
17.
Arterioscler Thromb Vasc Biol ; 40(5): 1110-1122, 2020 05.
Article in English | MEDLINE | ID: mdl-32131612

ABSTRACT

The immune system plays an important role in obesity-induced adipose tissue inflammation and the resultant metabolic dysfunction, which can lead to hypertension, dyslipidemia, and insulin resistance and their downstream sequelae of type 2 diabetes mellitus and cardiovascular disease. While macrophages are the most abundant immune cell type in adipose tissue, other immune cells are also present, such as B cells, which play important roles in regulating adipose tissue inflammation. This brief review will overview B-cell subsets, describe their localization in various adipose depots and summarize our knowledge about the function of these B-cell subsets in regulating adipose tissue inflammation, obesity-induced metabolic dysfunction and atherosclerosis.


Subject(s)
Adipose Tissue/immunology , Atherosclerosis/immunology , B-Lymphocyte Subsets/immunology , Panniculitis/immunology , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Adipose Tissue/pathology , Animals , Anti-Inflammatory Agents/therapeutic use , Atherosclerosis/diagnosis , Atherosclerosis/metabolism , Atherosclerosis/therapy , Autoimmunity , B-Lymphocyte Subsets/drug effects , B-Lymphocyte Subsets/metabolism , B-Lymphocyte Subsets/pathology , Cell Communication , Cytokines/immunology , Cytokines/metabolism , Humans , Immunotherapy , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Panniculitis/diagnosis , Panniculitis/metabolism , Panniculitis/therapy , Phenotype , Signal Transduction
18.
Metabolism ; 106: 154194, 2020 05.
Article in English | MEDLINE | ID: mdl-32135161

ABSTRACT

BACKGROUND: Low-grade inflammation and metabolic dysregulation are common comorbidities of obesity, both of which are associated with alterations in iRhom2-regulated pro-inflammatory cytokine and epidermal growth factor receptor (EGFR) ligand signaling. OBJECTIVE: Our objective was to determine the role of iRhom2 in the regulation of low-grade inflammation and metabolic dysregulation in a murine model of diet-induced obesity. METHODS: Wild type (WT) and iRhom2-deficient mice were fed normal chow (NC) or a high-fat diet (HFD) starting at 5 weeks of age for up to 33 weeks. Body composition, glucose and insulin tolerance, feeding behavior, and indirect calorimetry were measured at defined time points. Adipose tissue cytokine expression and inflammatory lesions known as crown-like structures (CLS) were analyzed at the end-point of the study. RESULTS: iRhom2-deficient mice show accelerated fat gain on a HFD, accompanied by insulin resistance. Indirect calorimetry did not demonstrate changes in energy expenditure or food intake, but locomotor activity was significantly reduced in HFD iRhom2-deficient mice. Interestingly, CLS, macrophage infiltration, and tumor necrosis factor (TNF) production were decreased in adipose tissue from HFD iRhom2-deficient mice, but circulating cytokines were unchanged. In inguinal and perigonadal fat, the EGFR ligand amphiregulin was markedly induced in HFD controls but completely prevented in iRhom2-deficient mice, suggesting a potentially dominant role of EGFR-dependent mechanisms over TNF in the modulation of insulin sensitivity. CONCLUSIONS: This study elucidates a novel role for iRhom2 as an immuno-metabolic regulator that affects adipose tissue inflammation independent of insulin resistance.


Subject(s)
Adipose Tissue/metabolism , Carrier Proteins/physiology , Diet, High-Fat , Inflammation/pathology , Insulin Resistance/genetics , Obesity/etiology , Weight Gain/genetics , Adipose Tissue/pathology , Animals , Carrier Proteins/genetics , Cells, Cultured , Diet, High-Fat/adverse effects , Disease Progression , Down-Regulation/genetics , Glucose Intolerance/genetics , Glucose Intolerance/metabolism , Glucose Intolerance/pathology , Inflammation/genetics , Inflammation/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/genetics , Obesity/metabolism , Obesity/pathology , Panniculitis/genetics , Panniculitis/metabolism , Panniculitis/pathology
19.
Front Immunol ; 11: 598566, 2020.
Article in English | MEDLINE | ID: mdl-33584664

ABSTRACT

Accumulating evidence reveals that adipose tissue is an immunologically active organ that exerts multiple impacts on the regulation of systemic energy metabolism. Adipose tissue immunity is modulated by the interactions between adipocytes and various immune cells. Nevertheless, the underlying mechanisms that control inter-cellular interactions between adipocytes and immune cells in adipose tissue have not been thoroughly elucidated. Recently, it has been demonstrated that adipocytes utilize lipid metabolites as a key mediator to initiate and mediate diverse adipose tissue immune responses. Adipocytes present lipid antigens and secrete lipid metabolites to determine adipose immune tones. In addition, the interactions between adipocytes and adipose immune cells are engaged in the control of adipocyte fate and functions upon metabolic stimuli. In this review, we discuss an integrated view of how adipocytes communicate with adipose immune cells using lipid metabolites. Also, we briefly discuss the newly discovered roles of adipose stem cells in the regulation of adipose tissue immunity.


Subject(s)
Adipocytes/metabolism , Adipose Tissue/immunology , Adipose Tissue/metabolism , Lipid Metabolism , Animals , Antigen Presentation , Biomarkers , Disease Susceptibility , Energy Metabolism , Humans , Immunity, Innate , Immunomodulation , Lipids/immunology , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , Panniculitis/etiology , Panniculitis/metabolism , Panniculitis/pathology , Stem Cells/metabolism
20.
Front Immunol ; 11: 612013, 2020.
Article in English | MEDLINE | ID: mdl-33854494

ABSTRACT

Adipocytes are the largest cell type in terms of volume, but not number, in adipose tissue. Adipocytes are prominent contributors to systemic metabolic health. Obesity, defined by excess adipose tissue (AT), is recognized as a low-grade chronic inflammatory state. Cytokines are inflammatory mediators that are produced in adipose tissue (AT) and function in both AT homeostatic as well as pathological conditions. AT inflammation is associated with systemic metabolic dysfunction and obesity-associated infiltration and proliferation of immune cells occurs in a variety of fat depots in mice and humans. AT immune cells secrete a variety of chemokines and cytokines that act in a paracrine manner on adjacent adipocytes. TNFα, IL-6, and MCP-1, are well studied mediators of AT inflammation. Oncostatin M (OSM) is another proinflammatory cytokine that is elevated in AT in human obesity, and its specific receptor (OSMRß) is also induced in conditions of obesity and insulin resistance. OSM production and paracrine signaling in AT regulates adipogenesis and the functions of AT. This review summarizes the roles of the oncostatin M receptor (OSMRß) as a modulator of adipocyte development and function its contributions to immunological adaptations in AT in metabolic disease states.


Subject(s)
Adipocytes/metabolism , Adipose Tissue/metabolism , Inflammation Mediators/metabolism , Obesity/metabolism , Oncostatin M Receptor beta Subunit/metabolism , Oncostatin M/metabolism , Panniculitis/metabolism , Adipocytes/immunology , Adipose Tissue/immunology , Adipose Tissue/physiopathology , Animals , Energy Metabolism , Homeostasis , Humans , Insulin Resistance , Obesity/immunology , Obesity/physiopathology , Panniculitis/immunology , Panniculitis/physiopathology , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL