Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 192
Filter
1.
Front Immunol ; 12: 757967, 2021.
Article in English | MEDLINE | ID: mdl-34759931

ABSTRACT

Innate lymphoid cells (ILCs) are the most recently described group of lymphoid subpopulations. These tissue-resident cells display a heterogeneity resembling that observed on different groups of T cells, hence their categorization as cytotoxic NK cells and helper ILCs type 1, 2 and 3. Each one of these groups is highly diverse and expresses different markers in a context-dependent manner. Type 2 innate lymphoid cells (ILC2s) are activated in response to helminth parasites and regulate the immune response. They are involved in the etiology of diseases associated with allergic responses as well as in the maintenance of tissue homeostasis. Markers associated with their identification differ depending on the tissue and model used, making the study and understanding of these cells a cumbersome task. This review compiles evidence for the heterogeneity of ILC2s as well as discussion and analyses of molecular markers associated with their identity, function, tissue-dependent expression, and how these markers contribute to the interaction of ILC2s with specific microenvironments to maintain homeostasis or respond to pathogenic challenges.


Subject(s)
Antigens, Differentiation/analysis , Lymphocyte Subsets/immunology , Adipose Tissue, White/immunology , Adipose Tissue, White/pathology , Animals , Cytokines/metabolism , Helminthiasis/immunology , Histocompatibility Antigens Class II/immunology , Homeostasis , Humans , Immunophenotyping , Inflammation , Intestines/immunology , Lung/immunology , Lymphocyte Subsets/chemistry , Mice , Nutrients , Organ Specificity , Proto-Oncogene Proteins c-kit/immunology , Receptors, Cell Surface/immunology , Skin/immunology , Stem Cell Factor/immunology
2.
Presse Med ; 50(3): 104073, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34547374

ABSTRACT

Lipodystrophy syndromes are rare, heterogeneous disorders characterized by the complete or partial deficiency of adipose tissue and are classified according to the extent of fat loss in generalized or partial subtypes, or based on the pathogenic mechanisms in genetic or acquired. While in most cases of congenital forms of lipodystrophy a genetic alteration can be identified, the pathogenic mechanisms responsible for the acquired diseases are not fully clarified. Based on the evidence of a positive association between most acquired lipodystrophies and autoimmune disorders including immune mediated alterations in the adipose tissue of patients affected by acquired lipodystrophy, a reaction against white adipose tissue antigens is postulated. Recent acquisitions have shed new light on the possible pathogenic mechanisms and identified novel forms of acquired lipodystrophy which are possibly immune-mediated. The aim of this review is to give an update on acquired lipodystrophies describing pathogenic mechanisms involved and the relationships between acquired lipodystrophies and other autoimmune disorders. Larger studies based on international disease registries are needed to collect accurate information on the prevalence, risk factors, genetic predisposition, natural history, disease markers and treatment efficacy of these ultrarare disorders.


Subject(s)
Adipose Tissue, White/immunology , Autoimmunity , Lipodystrophy/immunology , Adipose Tissue , Genetic Predisposition to Disease , Humans , Immune Checkpoint Inhibitors/adverse effects , Lipodystrophy/classification , Lipodystrophy/etiology , Lipodystrophy/therapy , Paraneoplastic Syndromes/etiology , Risk Factors , Syndrome
3.
Biochem Pharmacol ; 192: 114723, 2021 10.
Article in English | MEDLINE | ID: mdl-34364887

ABSTRACT

Both obesity and aging are associated with the development of metabolic diseases such as type 2 diabetes and cardiovascular disease. Chronic low-grade inflammation of adipose tissue is one of the mechanisms implicated in the progression of these diseases. Obesity and aging trigger adipose tissue alterations that ultimately lead to a pro-inflammatory phenotype of the adipose tissue-resident immune cells. Obesity and aging also share other features such as a higher visceral vs. subcutaneous adipose tissue ratio and a decreased lifespan. Here, we review the common characteristics of obesity and aging and the alterations in white adipose tissue and resident immune cells. We focus on the adipose tissue metabolic derangements in obesity and aging such as inflammation and adipose tissue remodeling.


Subject(s)
Adipocytes, White/immunology , Adipose Tissue, White/immunology , Aging/immunology , Body Fat Distribution/methods , Obesity/immunology , Adipocytes, White/metabolism , Adipocytes, White/pathology , Adipose Tissue, White/metabolism , Adipose Tissue, White/pathology , Aging/metabolism , Aging/pathology , Animals , Humans , Intra-Abdominal Fat/immunology , Intra-Abdominal Fat/metabolism , Intra-Abdominal Fat/pathology , Obesity/metabolism , Obesity/pathology
4.
Front Immunol ; 12: 696810, 2021.
Article in English | MEDLINE | ID: mdl-34335609

ABSTRACT

Changed dietary habits in Western countries such as reduced fiber intake represent an important lifestyle factor contributing to the increase in inflammatory immune-mediated diseases. The mode of action of beneficial fiber effects is not fully elucidated, but short-chain fatty acids (SCFA) and gut microbiota have been implicated. The aim of this study was to explore the impact of dietary fiber on lupus pathology and to understand underlying mechanisms. Here, we show that in lupus-prone NZB/WF1 mice low fiber intake deteriorates disease progression reflected in accelerated mortality, autoantibody production and immune dysregulation. In contrast to our original assumption, microbiota suppression by antibiotics or direct SCFA feeding did not influence the course of lupus-like disease. Mechanistically, our data rather indicate that in low fiber-fed mice, an increase in white adipose tissue mass, fat-inflammation and a disrupted intestinal homeostasis go along with systemic, low-grade inflammation driving autoimmunity. The links between obesity, intestinal leakage and low-grade inflammation were confirmed in human samples, while adaptive immune activation predominantly correlated with lupus activity. We further propose that an accelerated gastro-intestinal passage along with energy dilution underlies fiber-mediated weight regulation. Thus, our data highlight the often-overlooked effects of dietary fiber on energy homeostasis and obesity prevention. Further, they provide insight into how intricately the pathologies of inflammatory immune-mediated conditions, such as obesity and autoimmunity, might be interlinked, possibly sharing common pathways.


Subject(s)
Dietary Fiber/deficiency , Lupus Erythematosus, Systemic/etiology , Obesity/etiology , Adaptive Immunity , Adipose Tissue, White/immunology , Adipose Tissue, White/metabolism , Adipose Tissue, White/pathology , Adiposity , Adolescent , Adult , Aged , Aged, 80 and over , Animal Feed , Animals , Autoantibodies/blood , Autoimmunity , Case-Control Studies , Dietary Fiber/administration & dosage , Disease Models, Animal , Disease Progression , Energy Metabolism , Female , Humans , Inflammation Mediators/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/metabolism , Lupus Erythematosus, Systemic/pathology , Male , Mice, Inbred NZB , Middle Aged , Nutritive Value , Obesity/immunology , Obesity/metabolism , Obesity/pathology , Permeability , Young Adult
5.
Front Immunol ; 12: 719979, 2021.
Article in English | MEDLINE | ID: mdl-34381461

ABSTRACT

Adipose tissue macrophages (ATMs) regulate homeostasis and contribute to the metabolically harmful chronic inflammation in obese individuals. While evident heterogeneity of resident ATMs has been described previously, their phenotype, developmental origin, and functionality remain inconsistent. We analyzed white adipose tissue (WAT) during homeostasis and diet interventions using comprehensive and unbiased single-cell mass cytometry and genetic lineage tracking models. We now provide a uniform definition of individual subsets of resident ATMs. We show that in lean mice, WAT co-harbors eight kinetically evolving CD206+ macrophage subpopulations (defined by TIM4, CD163, and MHC II) and two CD206- macrophage subpopulations. TIM4-CD163+, TIM4-CD163- and CD206- macrophage populations are largely bone marrow-derived, while the proliferating TIM4+CD163+ subpopulation is of embryonic origin. All macrophage subtypes are active in phagocytosis, endocytosis, and antigen processing in vitro, whereas TIM4+CD163+ cells are superior in scavenging in vivo. A high-fat diet induces massive infiltration of CD206- macrophages and selective down-regulation of MHC II on TIM4+ macrophages. These changes are reversed by dietary intervention. Thus, the developmental origin and environment jointly regulate the functional malleability of resident ATMs.


Subject(s)
Adipose Tissue, White/metabolism , Macrophages/metabolism , Proteome/metabolism , Proteomics , Single-Cell Analysis , Adipose Tissue/immunology , Adipose Tissue/metabolism , Adipose Tissue, White/immunology , Animals , Biomarkers , Cell Differentiation , Cell Plasticity/genetics , Cell Plasticity/immunology , Cellular Reprogramming , Computational Biology , Energy Metabolism , Immunohistochemistry , Immunophenotyping , Macrophages/immunology , Male , Mice , Mice, Knockout , Models, Animal , Obesity/etiology , Obesity/metabolism , Obesity/pathology , Phagocytosis , Proteomics/methods , Single-Cell Analysis/methods
6.
Front Immunol ; 12: 670566, 2021.
Article in English | MEDLINE | ID: mdl-34220817

ABSTRACT

White adipose tissue (AT) contributes significantly to inflammation - especially in the context of obesity. Several of AT's intrinsic features favor its key role in local and systemic inflammation: (i) large distribution throughout the body, (ii) major endocrine activity, and (iii) presence of metabolic and immune cells in close proximity. In obesity, the concomitant pro-inflammatory signals produced by immune cells, adipocytes and adipose stem cells help to drive local inflammation in a vicious circle. Although the secretion of adipokines by AT is a prime contributor to systemic inflammation, the lipotoxicity associated with AT dysfunction might also be involved and could affect distant organs. In HIV-infected patients, the AT is targeted by both HIV infection and antiretroviral therapy (ART). During the primary phase of infection, the virus targets AT directly (by infecting AT CD4 T cells) and indirectly (via viral protein release, inflammatory signals, and gut disruption). The initiation of ART drastically changes the picture: ART reduces viral load, restores (at least partially) the CD4 T cell count, and dampens inflammatory processes on the whole-body level but also within the AT. However, ART induces AT dysfunction and metabolic side effects, which are highly dependent on the individual molecules and the combination used. First generation thymidine reverse transcriptase inhibitors predominantly target mitochondrial DNA and induce oxidative stress and adipocyte death. Protease inhibitors predominantly affect metabolic pathways (affecting adipogenesis and adipocyte homeostasis) resulting in insulin resistance. Recently marketed integrase strand transfer inhibitors induce both adipocyte adipogenesis, hypertrophy and fibrosis. It is challenging to distinguish between the respective effects of viral persistence, persistent immune defects and ART toxicity on the inflammatory profile present in ART-controlled HIV-infected patients. The host metabolic status, the size of the pre-established viral reservoir, the quality of the immune restoration, and the natural ageing with associated comorbidities may mitigate and/or reinforce the contribution of antiretrovirals (ARVs) toxicity to the development of low-grade inflammation in HIV-infected patients. Protecting AT functions appears highly relevant in ART-controlled HIV-infected patients. It requires lifestyle habits improvement in the absence of effective anti-inflammatory treatment. Besides, reducing ART toxicities remains a crucial therapeutic goal.


Subject(s)
Adipose Tissue, White/immunology , Adipose Tissue, White/metabolism , HIV Infections/immunology , HIV Infections/metabolism , Animals , Anti-HIV Agents/therapeutic use , HIV Infections/drug therapy , Humans , Inflammation/immunology , Inflammation/metabolism , Obesity/immunology , Obesity/metabolism
7.
Front Immunol ; 12: 629391, 2021.
Article in English | MEDLINE | ID: mdl-34122403

ABSTRACT

Little is known about the involvement of type 2 immune response-promoting intestinal tuft cells in metabolic regulation. We here examined the temporal changes in small intestinal tuft cell number and activity in response to high-fat diet-induced obesity in mice and investigated the relation to whole-body energy metabolism and the immune phenotype of the small intestine and epididymal white adipose tissue. Intake of high fat diet resulted in a reduction in overall numbers of small intestinal epithelial and tuft cells and reduced expression of the intestinal type 2 tuft cell markers Il25 and Tslp. Amongst >1,700 diet-regulated transcripts in tuft cells, we observed an early association between body mass expansion and increased expression of the gene encoding the serine protease inhibitor neuroserpin. By contrast, tuft cell expression of genes encoding gamma aminobutyric acid (GABA)-receptors was coupled to Tslp and Il25 and reduced body mass gain. Combined, our results point to a possible role for small intestinal tuft cells in energy metabolism via coupled regulation of tuft cell type 2 markers and GABA signaling receptors, while being independent of type 2 immune cell involvement. These results pave the way for further studies into interventions that elicit anti-obesogenic circuits via small intestinal tuft cells.


Subject(s)
Energy Metabolism , Epithelial Cells/metabolism , Intestine, Small/metabolism , Obesity/metabolism , Adipose Tissue, White/immunology , Adipose Tissue, White/metabolism , Animals , Cytokines/genetics , Cytokines/metabolism , Diet, High-Fat , Disease Models, Animal , Epithelial Cells/immunology , Gene Expression Regulation , Interleukins/genetics , Interleukins/metabolism , Intestine, Small/immunology , Male , Mice, Inbred C57BL , Neuropeptides/genetics , Neuropeptides/metabolism , Obesity/etiology , Obesity/genetics , Obesity/immunology , Phenotype , Receptors, GABA/genetics , Receptors, GABA/metabolism , Serpins/genetics , Serpins/metabolism , Signal Transduction , Time Factors , Weight Gain , Thymic Stromal Lymphopoietin , Neuroserpin
8.
Front Immunol ; 12: 643144, 2021.
Article in English | MEDLINE | ID: mdl-33717200

ABSTRACT

Glucose-dependent insulinotropic polypeptide (GIP) communicates information on energy availability from the gut to peripheral tissues. Disruption of its signaling in myeloid immune cells during high-fat diet (HFD)-induced obesity impairs energy homeostasis due to the unrestrained metabolically deleterious actions of S100A8/A9 alarmin. White adipose tissue (WAT) type 2 immune cell networks are important for maintaining metabolic and energy homeostasis and limiting obesity-induced inflammation. Nevertheless, the consequences of losing immune cell GIP receptor (GIPR) signaling on type 2 immunity in WAT remains unknown. Bone marrow (BM) chimerism was used to generate mice with GIPR (Gipr-/- BM) and GIPR/S100A8/A9 (Gipr-/- /S100a9-/- BM) deletion in immune cells. These mice were subjected to short (5 weeks) and progressive (14 weeks) HFD regimens. GIPR-deficiency was also targeted to myeloid cells by crossing Giprfl/fl mice and Lyz2cre/+ mice (LysMΔGipr ). Under both short and progressive HFD regimens, Gipr-/- BM mice exhibited altered expression of key type 2 immune cytokines in the epididymal visceral WAT (epiWAT), but not in subcutaneous inguinal WAT. This was further linked to declined representation of type 2 immune cells in epiWAT, such as group 2 innate lymphoid cells (ILC2), eosinophils, and FOXP3+ regulatory T cells (Tregs). Co-deletion of S100A8/A9 in Gipr-/- immune cells reversed the impairment of type 2 cytokine expression in epiWAT, suggesting a mechanistic role for this alarmin in type 2 immune suppression. LysMΔGipr mice on HFD also displayed altered expression of type 2 immune mediators, highlighting that GIPR-deficiency in myeloid immune cells is responsible for the impairment of type 2 immune networks. Finally, abrogated GIPR signaling in immune cells also affected adipocyte fraction cells, inducing their increased production of the beiging interfering cytokine IL-10 and stress- related type 2 cytokine IL-13. Collectively, these findings attribute an important role for GIPR in myeloid immune cells in supporting WAT type 2 immunity.


Subject(s)
Adipose Tissue, White/immunology , Lymphocytes/immunology , Obesity/immunology , Receptors, Gastrointestinal Hormone/physiology , Adipose Tissue, White/metabolism , Animals , Calgranulin A/physiology , Calgranulin B/physiology , Diet, High-Fat , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Signal Transduction/physiology , Thermogenesis
9.
JCI Insight ; 6(8)2021 04 22.
Article in English | MEDLINE | ID: mdl-33724954

ABSTRACT

Despite studies implicating adipose tissue T cells (ATT) in the initiation and persistence of adipose tissue inflammation, fundamental gaps in knowledge regarding ATT function impedes progress toward understanding how obesity influences adaptive immunity. We hypothesized that ATT activation and function would have tissue-resident-specific properties and that obesity would potentiate their inflammatory properties. We assessed ATT activation and inflammatory potential within mouse and human stromal vascular fraction (SVF). Surprisingly, murine and human ATTs from obese visceral white adipose tissue exhibited impaired inflammatory characteristics upon stimulation. Both environmental and cell-intrinsic factors are implicated in ATT dysfunction. Soluble factors from obese SVF inhibit ATT activation. Additionally, chronic signaling from macrophage major histocompatibility complex II (MHCII) is necessary for ATT impairment in obese adipose tissue but is independent of increased PD1 expression. To assess intracellular signaling mechanisms responsible for ATT inflammation impairments, single-cell RNA sequencing of ATTs was performed. ATTs in obese adipose tissue exhibit enrichment of genes characteristic of T cell exhaustion and increased expression of coinhibitory receptor Btla. In sum, this work suggests that obesity-induced ATTs have functional characteristics and gene expression resembling T cell exhaustion induced by local soluble factors and cell-to-cell interactions in adipose tissue.


Subject(s)
Adaptive Immunity/immunology , Adipose Tissue/immunology , Intra-Abdominal Fat/immunology , Obesity/immunology , T-Lymphocytes/immunology , Adipose Tissue, White/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Histocompatibility Antigens Class II/immunology , Humans , Macrophages/immunology , Male , Mice , Middle Aged , Programmed Cell Death 1 Receptor/immunology , Receptors, Immunologic/immunology , T-Lymphocyte Subsets/immunology
10.
Cancer Prev Res (Phila) ; 14(5): 541-550, 2021 05.
Article in English | MEDLINE | ID: mdl-33648942

ABSTRACT

Excess body fat and sedentary behavior are associated with increased breast cancer risk and mortality, including in normal weight women. To investigate underlying mechanisms, we examined whether adiposity and exercise impact the breast microenvironment (e.g., inflammation and aromatase expression) and circulating metabo-inflammatory factors. In a cross-sectional cohort study, breast white adipose tissue (WAT) and blood were collected from 100 women undergoing mastectomy for breast cancer risk reduction or treatment. Self-reported exercise behavior, body composition measured by dual-energy x-ray absorptiometry (DXA), and waist:hip ratio were obtained prior to surgery. Breast WAT inflammation (B-WATi) was assessed by IHC and aromatase expression was assessed by quantitative PCR. Metabolic and inflammatory blood biomarkers that are predictive of breast cancer risk and progression were measured. B-WATi was present in 56 of 100 patients and was associated with older age, elevated BMI, postmenopausal status, decreased exercise, hypertension and dyslipidemia (Ps < 0.001). Total body fat and trunk fat correlated with B-WATi and breast aromatase levels (Ps < 0.001). Circulating C-reactive protein, IL6, insulin, and leptin positively correlated with body fat and breast aromatase levels, while negative correlations were observed for adiponectin and sex hormone binding globulin (P < 0.001). Inverse relationships were observed with exercise (Ps < 0.05). In a subgroup of 39 women with normal BMI, body fat levels positively correlated with B-WATi and aromatase expression (Ps < 0.05). In conclusion, elevated body fat levels and decreased exercise are associated with protumorigenic micro- and host environments in normal, overweight, and obese individuals. These findings support the development of BMI-agnostic lifestyle interventions that target adiposity. PREVENTION RELEVANCE: We report that individuals with high body fat and low exercise levels have breast inflammation, higher breast aromatase expression, and levels of circulating metabo-inflammatory factors that have been associated with increased breast cancer risk. These findings support interventions to lower adiposity, even among normal weight individuals, to prevent tumor growth.


Subject(s)
Adipose Tissue, White/pathology , Adiposity/immunology , Breast Neoplasms/prevention & control , Breast/pathology , Exercise/immunology , Absorptiometry, Photon , Adipose Tissue, White/immunology , Adipose Tissue, White/surgery , Adult , Aged , Aged, 80 and over , Breast/immunology , Breast/surgery , Breast Neoplasms/immunology , Breast Neoplasms/metabolism , Breast Neoplasms/surgery , Cross-Sectional Studies , Exercise/statistics & numerical data , Female , Humans , Mastectomy , Middle Aged , Sedentary Behavior , Self Report/statistics & numerical data , Tumor Microenvironment/immunology
11.
Physiol Rep ; 9(3): e14721, 2021 02.
Article in English | MEDLINE | ID: mdl-33527668

ABSTRACT

BACKGROUND: LDL-cholesterol lowering variants that upregulate receptor uptake of LDL, such as in PCSK9 and HMGCR, are associated with diabetes via unclear mechanisms. Activation of the NLRP3 inflammasome/interleukin-1 beta (IL-1ß) pathway promotes white adipose tissue (WAT) dysfunction and type 2 diabetes (T2D) and is regulated by LDL receptors (LDLR and CD36). We hypothesized that: (a) normocholesterolemic subjects with lower plasma PCSK9, identifying those with higher WAT surface-expression of LDLR and CD36, have higher activation of WAT NLRP3 inflammasome and T2D risk factors, and; (b) LDL upregulate adipocyte NLRP3 inflammasome and inhibit adipocyte function. METHODOLOGY: Post hoc analysis was conducted in 27 overweight/ obese subjects with normal plasma LDL-C and measures of disposition index (DI during Botnia clamps) and postprandial fat metabolism. WAT was assessed for surface-expression of LDLR and CD36 (immunohistochemistry), protein expression (immunoblot), IL-1ß secretion (AlphaLISA), and function (3 H-triolein storage). RESULTS: Compared to subjects with higher than median plasma PCSK9, subjects with lower PCSK9 had higher WAT surface-expression of LDLR (+81%) and CD36 (+36%), WAT IL-1ß secretion (+284%), plasma IL-1 receptor-antagonist (+85%), and postprandial hypertriglyceridemia, and lower WAT pro-IL-1ß protein (-66%), WAT function (-62%), and DI (-28%), without group-differences in body composition, energy intake or expenditure. Adjusting for WAT LDLR or CD36 eliminated group-differences in WAT function, DI, and postprandial hypertriglyceridemia. Native LDL inhibited Simpson-Golabi Behmel-syndrome (SGBS) adipocyte differentiation and function and increased inflammation. CONCLUSION: Normocholesterolemic subjects with lower plasma PCSK9 and higher WAT surface-expression of LDLR and CD36 have higher WAT NLRP3 inflammasome activation and T2D risk factors. This may be due to LDL-induced inhibition of adipocyte function.


Subject(s)
Adipose Tissue, White/metabolism , CD36 Antigens/metabolism , Cholesterol/blood , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Obesity/blood , Proprotein Convertase 9/blood , Receptors, LDL/metabolism , Adipocytes, White/immunology , Adipocytes, White/metabolism , Adipogenesis , Adipose Tissue, White/immunology , Aged , Biomarkers/blood , Cells, Cultured , Diabetes Mellitus, Type 2/etiology , Down-Regulation , Female , Humans , Interleukin-1beta/metabolism , Male , Middle Aged , Obesity/complications , Obesity/enzymology , Obesity/immunology , Risk Assessment , Risk Factors
12.
JCI Insight ; 6(3)2021 02 08.
Article in English | MEDLINE | ID: mdl-33351782

ABSTRACT

Interleukin-10 (IL-10) is a critical cytokine used by immune cells to suppress inflammation. Paradoxically, immune cell-derived IL-10 can drive insulin resistance in obesity by suppressing adipocyte energy expenditure and thermogenesis. However, the source of IL-10 necessary for the suppression of adipocyte thermogenesis is unknown. We show here that CD4+Foxp3+ regulatory T cells (Tregs) are a substantial source of IL-10 and that Treg-derived IL-10 can suppress adipocyte beiging. Unexpectedly, Treg-specific loss of IL-10 resulted in increased insulin sensitivity and reduced obesity in high-fat diet-fed male mice. Mechanistically, we determined that Treg-specific loss of the transcription factor Blimp-1, a driver of IL-10 expression by Tregs, phenocopied the Treg-specific IL-10-deficient mice. Loss of Blimp-1 expression in Tregs resulted in reduced ST2+KLRG1+, IL-10-secreting Tregs, particularly in the white adipose tissue. Blimp-1-deficient mice were protected from glucose intolerance, insulin resistance, and diet-induced obesity, through increased white adipose tissue browning. Taken together, our data show that Blimp-1-regulated IL-10 secretion by Tregs represses white adipose tissue beiging to maintain adipose tissue homeostasis.


Subject(s)
Insulin Resistance/immunology , Insulin Resistance/physiology , Interleukin-10/immunology , Obesity/etiology , Positive Regulatory Domain I-Binding Factor 1/physiology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/physiology , Adipose Tissue, Beige/immunology , Adipose Tissue, Beige/metabolism , Adipose Tissue, White/immunology , Adipose Tissue, White/metabolism , Animals , Diet, High-Fat/adverse effects , Glucose Intolerance/immunology , Glucose Intolerance/metabolism , Interleukin-10/deficiency , Interleukin-10/genetics , Male , Mice , Mice, Knockout , Mice, Transgenic , Obesity/immunology , Obesity/physiopathology , Positive Regulatory Domain I-Binding Factor 1/deficiency , Positive Regulatory Domain I-Binding Factor 1/genetics , Thermogenesis/immunology , Thermogenesis/physiology
13.
Mol Med ; 26(1): 126, 2020 12 09.
Article in English | MEDLINE | ID: mdl-33297933

ABSTRACT

BACKGROUND: Innervation of adipose tissue is essential for the proper function of this critical metabolic organ. Numerous surgical and chemical denervation studies have demonstrated how maintenance of brain-adipose communication through both sympathetic efferent and sensory afferent nerves helps regulate adipocyte size, cell number, lipolysis, and 'browning' of white adipose tissue. Neurotrophic factors are growth factors that promote neuron survival, regeneration, and plasticity, including neurite outgrowth and synapse formation. Peripheral immune cells have been shown to be a source of neurotrophic factors in humans and mice. Although a number of immune cells reside in the adipose stromal vascular fraction (SVF), it has remained unclear what roles they play in adipose innervation. We previously demonstrated that adipose SVF secretes brain derived neurotrophic factor (BDNF). METHODS: We now show that deletion of this neurotrophic factor from the myeloid lineage of immune cells led to a 'genetic denervation' of inguinal subcutaneous white adipose tissue (scWAT), thereby causing decreased energy expenditure, increased adipose mass, and a blunted UCP1 response to cold stimulation. RESULTS: We and others have previously shown that noradrenergic stimulation via cold exposure increases adipose innervation in the inguinal depot. Here we have identified a subset of myeloid cells that home to scWAT upon cold exposure and are Ly6C+ CCR2+ Cx3CR1+ monocytes/macrophages that express noradrenergic receptors and BDNF. This subset of myeloid lineage cells also clearly interacted with peripheral nerves in the scWAT and were therefore considered neuroimmune cells. CONCLUSIONS: We propose that these myeloid lineage, cold induced neuroimmune cells (CINCs) are key players in maintaining adipose innervation as well as promoting adipose nerve remodeling under noradrenergic stimulation, such as cold exposure.


Subject(s)
Adipose Tissue/immunology , Adipose Tissue/innervation , Adipose Tissue/metabolism , Neuroimmunomodulation , Adipose Tissue, White/immunology , Adipose Tissue, White/innervation , Adipose Tissue, White/metabolism , Animals , Biomarkers , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Cold Temperature , Diet , Energy Metabolism , Female , Gene Expression , Male , Mice , Mice, Knockout , Neuroimmunomodulation/genetics , Phenotype , Stress, Physiological
14.
Front Immunol ; 11: 1800, 2020.
Article in English | MEDLINE | ID: mdl-32973755

ABSTRACT

White adipose tissue but recently also brown adipose tissue have emerged as endocrine organs. Age-associated obesity is accompanied by prolonged and elevated lipopolysaccharide (LPS)-induced sickness symptoms and increased cytokine and adipokine levels in the circulation partially originating from adipose tissue. In the present study, ex vivo fat explants were used to investigate how the exogenous pathogen-associated molecular pattern (PAMP) LPS or the endogenous danger-associated molecular patterns (DAMPs) high mobility group box-1 protein (HMGB1) and biglycan modulate the release of cytokines and adipokines/batokines and, thus, could influence systemic and/or local inflammation. The response of adipose tissue (epididymal, retroperitoneal, subcutaneous, and brown) was compared between young lean and old obese rats (2 vs. 24 months old). LPS induced a strong interleukin (IL)-6 and tumor necrosis factor (TNF) alpha release into the supernatant of all adipose tissue types investigated. HMGB1 (subcutaneous) and biglycan (retroperitoneal) led to an increased release of IL-6 and TNFalpha (HMGB1) and decreased visfatin and adiponectin (biglycan) secretion from epididymal adipose tissue (young rats). Visfatin was also decreased by HMGB1 in retroperitoneal adipose tissue of old rats. We found significantly higher leptin (all fat pads) and adiponectin (subcutaneous) levels in supernatants of adipose tissue from old compared to young rats, whereas visfatin secretion showed the opposite. The expression of the biglycan receptor Toll-like receptor (TLR) 2 as well as the LPS and HMGB1 receptors TLR4 and receptor for advanced glycation end products (RAGE) were reduced with age (TLR4/RAGE) and by stimulation with their ligands (subcutaneous). Overall, we revealed that adipokines/adipose-tissue released cytokines show some modulation of their release caused by mediators of septic (batokines) and sterile inflammation with potential implication for acute and chronic disease. Moreover, aging may increase or decrease the release of fat-derived mediators. These data show that DAMPS and LPS locally modulate cytokine secretion while only DAMPS but not LPS can locally alter adipokine secretion during inflammation.


Subject(s)
Adipokines/metabolism , Adipose Tissue, Brown/drug effects , Adipose Tissue, White/drug effects , Biglycan/pharmacology , Cytokines/metabolism , HMGB1 Protein/pharmacology , Lipopolysaccharides/pharmacology , Toll-Like Receptors/agonists , Adipose Tissue, Brown/immunology , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/immunology , Adipose Tissue, White/metabolism , Age Factors , Animals , Male , Rats, Wistar , Receptor for Advanced Glycation End Products/agonists , Receptor for Advanced Glycation End Products/metabolism , Secretory Pathway , Signal Transduction , Tissue Culture Techniques , Toll-Like Receptors/metabolism
15.
Front Immunol ; 11: 1953, 2020.
Article in English | MEDLINE | ID: mdl-32973799

ABSTRACT

Obesity is a major risk factor for the development of insulin resistance and type 2 diabetes. However, the mechanisms that trigger the underlying adipose tissues inflammation are not completely understood. Here, we show that the E3 ubiquitin ligase March1 controls the phenotypic and functional properties of CD8+ T cells in mice white adipose tissue. In a diet-induced obesity model, mice lacking March1 [March1 knockout (KO)] show increased insulin resistance compared to their WT counterparts. Also, in obese March1 KO mice, the proportions of effector/memory (Tem) and resident/memory (Trm) CD8+ T cells were higher in the visceral adipose tissue, but not in the spleen. The effect of March1 on insulin resistance and on the phenotype of adipose tissue CD8+ T cells was independent of major histocompatibility complex class II ubiquitination. Interestingly, we adoptively transferred either WT or March1 KO splenic CD8+ T cells into obese WT chimeras that had been reconstituted with Rag1-deficient bone marrow. We observed an enrichment of Tem and Trm cells and exacerbated insulin resistance in mice that received March1 KO CD8 T cells. Mechanistically, we found that March1 deficiency alters the metabolic activity of CD8+ T cells. Our results provide additional evidence of the involvement of CD8+ T cells in adipose tissue inflammation and suggest that March1 controls the metabolic reprogramming of these cells.


Subject(s)
Adipose Tissue, White/enzymology , CD8-Positive T-Lymphocytes/metabolism , Immunologic Memory , Insulin Resistance , Obesity/enzymology , Ubiquitin-Protein Ligases/deficiency , Adipose Tissue, White/immunology , Adoptive Transfer , Animals , Blood Glucose/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/transplantation , Diet, High-Fat , Disease Models, Animal , Energy Metabolism , Lymphocyte Activation , Mice, Inbred C57BL , Mice, Knockout , Obesity/genetics , Obesity/immunology , Phenotype , Spleen/enzymology , Spleen/immunology , Ubiquitin-Protein Ligases/genetics
16.
J Agric Food Chem ; 68(31): 8255-8262, 2020 Aug 05.
Article in English | MEDLINE | ID: mdl-32643946

ABSTRACT

Medium- and long-chain triacylglycerols (MLCTs) were synthesized from rapeseed oil (RO), one kind of commonly used edible long-chain triacylglycerols (TGs), and then delivered to high-fat diet (HFD)-induced obese rats. Compared with RO, MLCT consumption exhibited more potent effects on reducing body and tissue weight gains, plasma TG, and total cholesterol (TC) levels and on improving hepatic TG, TC, fatty acid synthase, acetyl-CoA carboxylase, and lipoprteinlipase contents. Meanwhile, lower amounts of tumor necrosis factor-α (TNF-α), monocyte chemoattractant protein-1, and endotoxin in plasma, lower levels of interleukin-6 and TNF-α, and higher levels of interleukin-10 in both livers and white adipose tissues were detected in MLCT-fed rats. MLCT intake also remarkably suppressed the size of adipocytes and the number of macrophages. In conclusion, our study suggested that the interesterified MLCT was more efficacious in improving the lipid metabolism and inflammation in HFD-induced obese rats than RO.


Subject(s)
Lipid Metabolism , Obesity/drug therapy , Triglycerides/chemistry , Triglycerides/metabolism , Adipose Tissue, White/immunology , Adipose Tissue, White/metabolism , Animals , Chemokine CCL2/genetics , Chemokine CCL2/immunology , Cholesterol/metabolism , Diet, High-Fat/adverse effects , Esterification , Humans , Liver/immunology , Liver/metabolism , Male , Obesity/etiology , Obesity/immunology , Obesity/metabolism , Rapeseed Oil/chemistry , Rapeseed Oil/metabolism , Rats , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
17.
Rejuvenation Res ; 23(4): 349-352, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32718231

ABSTRACT

Proinflammatory alterations of white adipose tissue (WAT) with increasing age play an important role in mammalian aging. WAT produced eotaxin-1 (CCL11-C-C motif chemokine ligand 11) and monocyte chemoattractant protein 1 (MCP-1) (CCL2) are elevated in old mammals. Obese and old adipose tissues produce excessive proinflammatory cytokines such as interleukin (IL)-6, CCL2, and IL-1-beta that contribute to inflammaging. WAT-based inflammaging involves an altered homeostatic equilibrium between proinflammatory cells such as activated type 1 macrophages, B cells (high IgJ) and T cells, and anti-inflammatory eosinophils and Tregs. Specifically, young and lean individuals exhibit a high eosinophil-to-macrophage ratio with an enrichment of alternative activated tissue macrophages that is reduced in the WAT of aging mice. Eosinophils from young animals adoptively transferred to old mice, home to WAT and reverse many of the immunoinflammatory signatures associated with aging. Whether eosinophil-based therapies for inflammaging could be created remains an open question.


Subject(s)
Adipose Tissue, White/immunology , Adipose Tissue/immunology , Anti-Inflammatory Agents/pharmacology , Antinematodal Agents/pharmacology , Eosinophils/immunology , Helminths/immunology , Macrophages/immunology , Animals , Anti-Inflammatory Agents/immunology , Antinematodal Agents/immunology , Cytokines/metabolism , Humans
18.
FASEB J ; 34(7): 9755-9770, 2020 07.
Article in English | MEDLINE | ID: mdl-32510702

ABSTRACT

Regulatory T cells (Tregs) play essential roles in obesity and diabetes. Here, we report a role of Tregs in enhancing ß3-adrenergic receptor agonist CL316243 (CL)-stimulated thermogenic program in subcutaneous adipose tissue (SAT), but not in visceral fat. CL treatment for 7 days increased SAT adipocyte beiging and thermogenic gene expression in male or female mice. Adoptive transfer of Tregs enhanced this CL activity. Such Treg activity lost in male epididymal white adipose tissue (eWAT) and female gonadal gWAT. Adipocyte culture yielded the same conclusion. Tregs enhanced the expression of CL-induced thermogenic genes in SAT from male and female mice. This activity of Tregs reduced or disappeared in adipocytes from eWAT or gWAT. Both CL and Tregs induced much higher UCP-1 (uncoupling protein-1) expression in SAT from females than that from males. A mechanistic study demonstrated a role of Tregs in suppressing the expression of M1 macrophage markers (Tnfa, Il6, iNos, Ip10) and promoting the expression of M2 macrophage markers (Mrc1, Arg1, Il10) in bone-marrow-derived macrophages or in SAT from male or female mice. In female mice with pre-established obesity, Treg adoptive transfer reduced the gWAT weight in 2 weeks. Together with CL treatment, Treg adoptive transfer reduced the SAT weight and further improved CL-induced glucose metabolism and insulin sensitivity in female obese mice, but did not affect CL-induced body weight loss in male or female obese mice. This study revealed a predominant role of Tregs in female mice in promoting adipocyte beiging and thermogenesis in SAT, in part by slanting M2 macrophage polarization.


Subject(s)
Adipose Tissue, Brown/pathology , Adipose Tissue, White/pathology , Obesity/etiology , Subcutaneous Fat/pathology , T-Lymphocytes, Regulatory/immunology , Thermogenesis , Adipose Tissue, Brown/immunology , Adipose Tissue, White/immunology , Animals , Energy Metabolism , Female , Insulin Resistance , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/pathology , Subcutaneous Fat/immunology , T-Lymphocytes, Regulatory/pathology
19.
Sci Rep ; 10(1): 9437, 2020 06 10.
Article in English | MEDLINE | ID: mdl-32523023

ABSTRACT

Adipocytes from white-adipose tissue are known to produce inflammatory cytokines, which play a major role in energy balance and metabolism. While they can respond to pathogen-associated molecular pattern (PAMPs) such as lipopolysaccharide (LPS) from bacteria, it is not known whether adipocytes can be stimulated by fungal cells. Previously, adipocytes were shown to produce toll-like receptor 2 (TLR2), a ß-glucan receptor, suggesting that they could respond to ß-glucan on the fungal cell wall. In this study, we show that heat-killed yeast induce an inflammatory response in adipocytes. Using fungal-like particles, namely laminarin-coated beads (LCB), we find that these particles trigger the expression of many key inflammatory genes in dose- and time-dependent fashions in adipocytes. These results suggest that ß-glucan on the fungal cell wall is sufficient to elicit an inflammatory response in adipocytes. In addition, we show that both LCB and LCB-treated conditioned medium from RAW 264.7 murine macrophages (LCB-RM) induce the expression of those inflammatory genes through IKKß-IκBα proteins. Together, we conclude that the fungal-like particles and the conditioned medium elicit an inflammatory response in adipocytes through the canonical or classical NF-κB pathway.


Subject(s)
Adipose Tissue, White/immunology , Adipose Tissue, White/metabolism , Glucans/metabolism , 3T3-L1 Cells , Adipocytes/metabolism , Adipose Tissue, White/physiology , Animals , Culture Media, Conditioned/pharmacology , Cytokines/metabolism , Glucans/pharmacology , Inflammation/metabolism , Lipopolysaccharides/pharmacology , Macrophages/metabolism , Mice , NF-KappaB Inhibitor alpha/metabolism , NF-kappa B/metabolism , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/metabolism , beta-Glucans/metabolism
20.
Circ Res ; 126(11): 1590-1612, 2020 05 22.
Article in English | MEDLINE | ID: mdl-32437300

ABSTRACT

Adipose tissue (AT) plays a central role in both metabolic health and pathophysiology. Its expansion in obesity results in increased mortality and morbidity, with contributions to cardiovascular disease, diabetes mellitus, fatty liver disease, and cancer. Obesity prevalence is at an all-time high and is projected to be 50% in the United States by 2030. AT is home to a large variety of immune cells, which are critical to maintain normal tissue functions. For example, γδ T cells are fundamental for AT innervation and thermogenesis, and macrophages are required for recycling of lipids released by adipocytes. The expansion of visceral white AT promotes dysregulation of its immune cell composition and likely promotes low-grade chronic inflammation, which has been proposed to be the underlying cause for the complications of obesity. Interestingly, weight loss after obesity alters the AT immune compartment, which may account for the decreased risk of developing these complications. Recent technological advancements that allow molecular investigation on a single-cell level have led to the discovery of previously unappreciated heterogeneity in many organs and tissues. In this review, we will explore the heterogeneity of immune cells within the visceral white AT and their contributions to homeostasis and pathology.


Subject(s)
Adipose Tissue, White/immunology , Leukocytes/immunology , Obesity/immunology , Adipose Tissue, White/cytology , Adipose Tissue, White/pathology , Animals , Humans , Obesity/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...