Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
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
2.
Front Endocrinol (Lausanne) ; 12: 780300, 2021.
Article in English | MEDLINE | ID: mdl-35111136

ABSTRACT

The risk of obesity in adulthood is subject to programming in the womb. Maternal obesity contributes to programming of obesity and metabolic disease risk in the adult offspring. With the increasing prevalence of obesity in women of reproductive age there is a need to understand the ramifications of maternal high-fat diet (HFD) during pregnancy on offspring's metabolic heath trajectory. In the present study, we determined the long-term metabolic outcomes on adult male and female offspring of dams fed with HFD during pregnancy. C57BL/6J dams were fed either Ctrl or 60% Kcal HFD for 4 weeks before and throughout pregnancy, and we tested glucose homeostasis in the adult offspring. Both Ctrl and HFD-dams displayed increased weight during pregnancy, but HFD-dams gained more weight than Ctrl-dams. Litter size and offspring birthweight were not different between HFD-dams or Ctrl-dams. A significant reduction in random blood glucose was evident in newborns from HFD-dams compared to Ctrl-dams. Islet morphology and alpha-cell fraction were normal but a reduction in beta-cell fraction was observed in newborns from HFD-dams compared to Ctrl-dams. During adulthood, male offspring of HFD-dams displayed comparable glucose tolerance under normal chow. Male offspring re-challenged with HFD displayed glucose intolerance transiently. Adult female offspring of HFD-dams demonstrated normal glucose tolerance but displayed increased insulin resistance relative to controls under normal chow diet. Moreover, adult female offspring of HFD-dams displayed increased insulin secretion in response to high-glucose treatment, but beta-cell mass were comparable between groups. Together, these data show that maternal HFD at pre-conception and during gestation predisposes the female offspring to insulin resistance in adulthood.


Subject(s)
Blood Glucose/metabolism , Diet, High-Fat , Glucose Intolerance/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Maternal Exposure , Obesity, Maternal/metabolism , Prenatal Exposure Delayed Effects/metabolism , Animals , Animals, Newborn , Birth Weight , Female , Insulin-Secreting Cells/pathology , Litter Size , Male , Mice , Organ Size , Pregnancy
3.
Mol Metab ; 42: 101078, 2020 12.
Article in English | MEDLINE | ID: mdl-32919095

ABSTRACT

OBJECTIVE: Canonical Wnt/ß-catenin signaling is a well-studied endogenous regulator of mesenchymal cell fate determination, promoting osteoblastogenesis and inhibiting adipogenesis. However, emerging genetic evidence in humans links a number of Wnt pathway members to body fat distribution, obesity, and metabolic dysfunction, suggesting that this pathway also functions in adipocytes. Recent studies in mice have uncovered compelling evidence that the Wnt signaling pathway plays important roles in adipocyte metabolism, particularly under obesogenic conditions. However, complexities in Wnt signaling and differences in experimental models and approaches have thus far limited our understanding of its specific roles in this context. METHODS: To investigate roles of the canonical Wnt pathway in the regulation of adipocyte metabolism, we generated adipocyte-specific ß-catenin (ß-cat) knockout mouse and cultured cell models. We used RNA sequencing, ChIP sequencing, and molecular approaches to assess expression of Wnt targets and lipogenic genes. We then used functional assays to evaluate effects of ß-catenin deficiency on adipocyte metabolism, including lipid and carbohydrate handling. In mice maintained on normal chow and high-fat diets, we assessed the cellular and functional consequences of adipocyte-specific ß-catenin deletion on adipose tissues and systemic metabolism. RESULTS: We report that in adipocytes, the canonical Wnt/ß-catenin pathway regulates de novo lipogenesis (DNL) and fatty acid monounsaturation. Further, ß-catenin mediates effects of Wnt signaling on lipid metabolism in part by transcriptional regulation of Mlxipl and Srebf1. Intriguingly, adipocyte-specific loss of ß-catenin is sensed and defended by CD45-/CD31- stromal cells to maintain tissue-wide Wnt signaling homeostasis in chow-fed mice. With long-term high-fat diet, this compensatory mechanism is overridden, revealing that ß-catenin deletion promotes resistance to diet-induced obesity and adipocyte hypertrophy and subsequent protection from metabolic dysfunction. CONCLUSIONS: Taken together, our studies demonstrate that Wnt signaling in adipocytes is required for lipogenic gene expression, de novo lipogenesis, and lipid desaturation. In addition, adipose tissues rigorously defend Wnt signaling homeostasis under standard nutritional conditions, such that stromal-vascular cells sense and compensate for adipocyte-specific loss. These findings underscore the critical importance of this pathway in adipocyte lipid metabolism and adipose tissue function.


Subject(s)
Adipocytes/metabolism , Adipose Tissue/metabolism , Wnt Signaling Pathway/physiology , Adipocytes/physiology , Adipogenesis/physiology , Adipose Tissue/physiology , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Cell Differentiation , Cells, Cultured , Gene Expression/genetics , Gene Expression Regulation/genetics , Lipid Metabolism , Lipogenesis/physiology , Mice , Mice, Knockout , Obesity , Sterol Regulatory Element Binding Protein 1 , Stromal Cells/metabolism , Wnt Signaling Pathway/drug effects , Wnt Signaling Pathway/genetics , Wnt1 Protein/metabolism , beta Catenin/genetics , beta Catenin/metabolism
4.
Biotechnol Bioeng ; 117(12): 3891-3901, 2020 12.
Article in English | MEDLINE | ID: mdl-32729936

ABSTRACT

Dysfunctional adipose tissue plays a central role in the pathogenesis of the obesity-related metabolic disease, including type 2 diabetes. Targeting adipose tissue using biopolymer implants is a novel therapeutic approach for metabolic disease. We transplanted porous poly(lactide-co-glycolide) (PLG) implants coated with human interleukin-4 (hIL-4)-expressing lentivirus into epididymal white adipose tissue (eWAT) of mice fed a high-fat diet. Tissue and systemic inflammation and metabolism were studied with flow cytometry, immunohistochemistry, quantitative real-time polymerase chain reaction, adipose tissue histology, and in vivo glucose tolerance testing at 2 and 10 weeks of a high-fat diet. PLG implants carrying hIL-4-expressing lentivirus implanted into epididymal white adipose tissue of mice-regulated adipose tissue inflammation, including increased CD3+ CD4+ T-cell frequency, increased eWAT adipocyte hypertrophy, and decreased FASN and ATGL expression, along with reduced fasting blood glucose levels. These effects were observed in early obesity but were not maintained in established obesity. Local delivery of bioimplants loaded with cytokine-expressing lentivirus vectors to adipose tissue influences tissue inflammation and systemic metabolism in early obesity. Further study will be required to show more durable metabolic effects. These data demonstrate that polymer biomaterials implanted into adipose tissue have the potential to modulate local tissue and systemic inflammation and metabolism.


Subject(s)
Adipose Tissue/metabolism , Implants, Experimental , Interleukin-4 , Lentivirus , Obesity/metabolism , Transduction, Genetic , Animals , Disease Models, Animal , Humans , Inflammation/genetics , Inflammation/metabolism , Interleukin-4/biosynthesis , Interleukin-4/genetics , Male , Mice , Obesity/genetics
5.
Obesity (Silver Spring) ; 28(6): 1086-1097, 2020 06.
Article in English | MEDLINE | ID: mdl-32281747

ABSTRACT

OBJECTIVE: Weight regain after weight loss is common, and there is evidence to suggest negative effects on health because of weight cycling. This study sought to investigate the impact of weight regain in formerly obese mice on adipose tissue architecture and stromal cell function. METHODS: A diet-switch model was employed for obesity induction, weight loss, and weight regain in mice. Flow cytometry quantified adipose tissue leukocytes in adipose tissue. Liver and adipose tissue depots were compared to determine tissue-specific effects of weight cycling. RESULTS: Epididymal white adipose tissue of formerly obese mice failed to expand in response to repeat exposure to high-fat diet and retained elevated numbers of macrophages and T cells. Weight regain was associated with disproportionally elevated liver mass, hepatic triglyceride content, serum insulin concentration, and serum transaminase concentration. These effects occurred despite an extended 6-month weight loss cycle and they demonstrate that formerly obese mice maintain durable alterations in their physiological response to weight regain. Conditioned media from epididymal adipose tissue of formerly obese mice inhibited adipogenesis of 3T3-L1 preadipocytes, suggesting a potential mechanism to explain failed epididymal adipose tissue expansion during weight regain. CONCLUSIONS: Metabolic abnormalities related to defects in adipose tissue expansion and ongoing dysfunction manifest in formerly obese mice during weight regain.


Subject(s)
Adipose Tissue/metabolism , Fatty Liver/metabolism , Obesity/metabolism , Weight Gain/physiology , Animals , Diet, High-Fat , Gene Expression , Male , Mice , Mice, Inbred C57BL , Mice, Obese
6.
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
7.
Mol Cell Endocrinol ; 505: 110740, 2020 04 05.
Article in English | MEDLINE | ID: mdl-31987897

ABSTRACT

Adipose tissue derived chronic inflammation is a critical component of obesity induced type II diabetes. Major histocompatibility complex II (MHCII) mediated T cell activation within adipose tissue is one mechanism that contributes to this phenotype. However, the contribution of dendritic cells as professional antigen presenting cells in adipose issue has not previously been explored. Using ItgaxCre x MHCIIfl/fl (M11cKO) mice we observed adipose tissue specific changes in adipose tissue leukocytes. While there was a complete knockout of MHCII in dendritic cells, MHCII was also absent on the majority of macrophages. This resulted in reduction of TCR expression in CD4+ T cells in obese adipose tissue, and an increase in CD8+ and CD4+ CD8+ double positive T cells with decreased CD4+ T cells independent of diet type. Increased CD8+ cells were not observed in the spleen, suggesting adipose tissue T cell regulation is tissue specific. In vitro studies demonstrated more potent antigen presentation function in adipose tissue dendritic cells compared to macrophages. Obese M11cKO mice had decreased CD11c+ adipose tissue macrophages. Despite the changes of immune cellularity in adipose tissue, M11cKO largely did not change inflammatory gene expression in adipose tissue and did not demonstrate differences in glucose and insulin intolerance. Overall MHCII expression on CD11c+ cells is important for maintaining CD4+ and CD8+ adipose tissue T cells, but these cellular changes fail to alter inflammatory output and systemic metabolism.


Subject(s)
Adipose Tissue/pathology , Dendritic Cells/pathology , Homeostasis , Obesity/immunology , Signal Transduction , T-Lymphocytes/immunology , Animals , Antigen Presentation/immunology , CD11 Antigens/metabolism , Cell Proliferation , Gene Expression Regulation , Glucose/metabolism , Histocompatibility Antigens Class II/metabolism , Inflammation/genetics , Inflammation/pathology , Insulin Resistance , Macrophages/pathology , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Spleen/pathology
8.
Mol Metab ; 14: 60-70, 2018 08.
Article in English | MEDLINE | ID: mdl-29934059

ABSTRACT

OBJECTIVE: Long noncoding RNAs (lncRNAs) are emerging as powerful regulators of adipocyte differentiation and gene expression. However, their significance in adipose tissue metabolism and physiology has not been demonstrated in vivo. We previously identified Blnc1 as a conserved lncRNA regulator of brown and beige adipocyte differentiation. In this study, we investigated the physiological role of Blnc1 in thermogenesis, adipose remodeling and systemic metabolism. METHODS: We generated fat-specific Blnc1 transgenic and conditional knockout mouse strains and investigated how adipocyte Blnc1 levels are causally linked to key aspects of metabolic health following diet-induced obesity. We performed studies using cultured adipocytes to establish cell-autonomous role of Blnc1 in regulating adipocyte gene programs. RESULTS: Blnc1 is highly induced in both brown and white fats from obese mice. Fat-specific inactivation of Blnc1 impairs cold-induced thermogenesis and browning and exacerbates obesity-associated brown fat whitening, adipose tissue inflammation and fibrosis, leading to more severe insulin resistance and hepatic steatosis. On the contrary, transgenic expression of Blnc1 in adipose tissue elicits the opposite and beneficial metabolic effects, supporting a critical role of Blnc1 in driving adipose adaptation and homeostatic remodeling during obesity. Mechanistically, Blnc1 cell-autonomously attenuates proinflammatory cytokine signaling and promotes fuel storage in adipocytes through its protein partner Zbtb7b. CONCLUSIONS: This study illustrates a surprisingly pleiotropic and dominant role of lncRNA in driving adaptive adipose tissue remodeling and preserving metabolic health.


Subject(s)
Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Homeostasis , Obesity/genetics , RNA, Long Noncoding/genetics , Adipose Tissue, Brown/cytology , Adipose Tissue, White/cytology , Animals , Cell Line, Tumor , Cells, Cultured , Cytokines/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Diet, High-Fat/adverse effects , Energy Metabolism , Mice , Mice, Inbred C57BL , Obesity/etiology , RNA, Long Noncoding/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
9.
J Leukoc Biol ; 103(4): 615-628, 2018 04.
Article in English | MEDLINE | ID: mdl-29493813

ABSTRACT

Obesity-related changes in adipose tissue leukocytes, in particular adipose tissue macrophages (ATMs) and dendritic cells (ATDCs), are implicated in metabolic inflammation, insulin resistance, and altered regulation of adipocyte function. We evaluated stromal cell and white adipose tissue (WAT) expansion dynamics with high fat diet (HFD) feeding for 3-56 days, quantifying ATMs, ATDCs, endothelial cells (ECs), and preadipocytes (PAs) in visceral epididymal WAT and subcutaneous inguinal WAT. To better understand mechanisms of the early response to obesity, we evaluated ATM proliferation and lipid accumulation. ATMs, ATDCs, and ECs increased with rapid WAT expansion, with ATMs derived primarily from a CCR2-independent resident population. WAT expansion stimulated proliferation in resident ATMs and ECs, but not CD11c+ ATMs or ATDCs. ATM proliferation was unperturbed in Csf2- and Rag1-deficient mice with WAT expansion. Additionally, ATM apoptosis decreased with WAT expansion, and proliferation and apoptosis reverted to baseline with weight loss. Adipocytes reached maximal hypertrophy at 28 days of HFD, coinciding with a plateau in resident ATM accumulation and the appearance of lipid-laden CD11c+ ATMs in visceral epididymal WAT. ATM increases were proportional to tissue expansion and adipocyte hypertrophy, supporting adipocyte-mediated regulation of resident ATMs. The appearance of lipid-laden CD11c+ ATMs at peak adipocyte size supports a role in responding to ectopic lipid accumulation within adipose tissue. In contrast, ATDCs increase independently of proliferation and may be derived from circulating precursors. These changes precede and establish the setting in which large-scale adipose tissue infiltration of CD11c+ ATMs, inflammation, and adipose tissue dysfunction contributes to insulin resistance.


Subject(s)
Adipose Tissue, White/cytology , Cell Proliferation , Dendritic Cells/cytology , Endothelium, Vascular/cytology , Lipids/analysis , Macrophages/cytology , Obesity/physiopathology , Adipose Tissue, White/metabolism , Animals , Dendritic Cells/metabolism , Diet, High-Fat/adverse effects , Endothelium, Vascular/metabolism , Female , Humans , Inflammation/physiopathology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL
10.
Adipocyte ; 6(2): 134-140, 2017 04 03.
Article in English | MEDLINE | ID: mdl-28425841

ABSTRACT

Predictors of weight loss responses are not well-defined. We hypothesized that adipose tissue phenotypic features related to remodeling would be associated with bariatric surgery weight loss responses. Visceral and subcutaneous adipose tissues collected from patients during bariatric surgery were studied with flow cytometry, immunohistochemistry, and QRTPCR, and results correlated with weight loss outcomes. Age, male sex, and a diagnosis of type 2 diabetes were associated with less weight loss. Adipocyte size was increased and preadipocyte frequency was decreased in visceral adipose tissue from diabetic subjects. Decreased adipose tissue preadipocyte frequency was associated with less weight loss in women but not men. These data suggest that phenotypic features of adipose tissue remodeling may predict responses to weight loss interventions.


Subject(s)
Adipose Tissue/metabolism , Adipose Tissue/pathology , Adipocytes/physiology , Adiposity , Bariatric Surgery/methods , Female , Humans , Hyperplasia/metabolism , Hypertrophy/metabolism , Intra-Abdominal Fat , Male , Obesity/metabolism , Subcutaneous Fat/metabolism , Treatment Outcome , Weight Loss/physiology
11.
Diabetes ; 66(2): 392-406, 2017 02.
Article in English | MEDLINE | ID: mdl-28108608

ABSTRACT

Obesity causes dramatic proinflammatory changes in the adipose tissue immune environment, but relatively little is known regarding how this inflammation responds to weight loss (WL). To understand the mechanisms by which meta-inflammation resolves during WL, we examined adipose tissue leukocytes in mice after withdrawal of a high-fat diet. After 8 weeks of WL, mice achieved similar weights and glucose tolerance values as age-matched lean controls but showed abnormal insulin tolerance. Despite fat mass normalization, total and CD11c+ adipose tissue macrophage (ATM) content remained elevated in WL mice for up to 6 months and was associated with persistent fibrosis in adipose tissue. ATMs in formerly obese mice demonstrated a proinflammatory profile, including elevated expression of interferon-γ, tumor necrosis factor-α, and interleukin-1ß. T-cell-deficient Rag1-/- mice showed a degree of ATM persistence similar to that in WT mice, but with reduced inflammatory gene expression. ATM proliferation was identified as the predominant mechanism by which ATMs are retained in adipose tissue with WL. Our study suggests that WL does not completely resolve obesity-induced ATM activation, which may contribute to the persistent adipose tissue damage and reduced insulin sensitivity observed in formerly obese mice.


Subject(s)
Adipose Tissue/immunology , Cell Proliferation , Macrophages/immunology , Obesity/immunology , Weight Loss/immunology , Adipose Tissue/cytology , Animals , Body Weight , Diet, High-Fat , Flow Cytometry , Fluorescent Antibody Technique , Gene Expression Profiling , Glucose Tolerance Test , Homeodomain Proteins/genetics , Immunoblotting , Immunohistochemistry , Inflammation/immunology , Insulin/metabolism , Interferon-gamma/immunology , Interleukin-1beta/immunology , Macrophages/cytology , Male , Mice , Mice, Knockout , Mice, Obese , T-Lymphocytes , Tumor Necrosis Factor-alpha/immunology
12.
J Immunol ; 197(9): 3650-3661, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27683748

ABSTRACT

Dynamic changes of adipose tissue leukocytes, including adipose tissue macrophage (ATM) and adipose tissue dendritic cells (ATDCs), contribute to obesity-induced inflammation and metabolic disease. However, clear discrimination between ATDC and ATM in adipose tissue has limited progress in the field of immunometabolism. In this study, we use CD64 to distinguish ATM and ATDC, and investigated the temporal and functional changes in these myeloid populations during obesity. Flow cytometry and immunostaining demonstrated that the definition of ATM as F4/80+CD11b+ cells overlaps with other leukocytes and that CD45+CD64+ is specific for ATM. The expression of core dendritic cell genes was enriched in CD11c+CD64- cells (ATDC), whereas core macrophage genes were enriched in CD45+CD64+ cells (ATM). CD11c+CD64- ATDCs expressed MHC class II and costimulatory receptors, and had similar capacity to stimulate CD4+ T cell proliferation as ATMs. ATDCs were predominantly CD11b+ conventional dendritic cells and made up the bulk of CD11c+ cells in adipose tissue with moderate high-fat diet exposure. Mixed chimeric experiments with Ccr2-/- mice demonstrated that high-fat diet-induced ATM accumulation from monocytes was dependent on CCR2, whereas ATDC accumulation was less CCR2 dependent. ATDC accumulation during obesity was attenuated in Ccr7-/- mice and was associated with decreased adipose tissue inflammation and insulin resistance. CD45+CD64+ ATM and CD45+CD64-CD11c+ ATDCs were identified in human obese adipose tissue and ATDCs were increased in s.c. adipose tissue compared with omental adipose tissue. These results support a revised strategy for unambiguous delineation of ATM and ATDC, and suggest that ATDCs are independent contributors to adipose tissue inflammation during obesity.


Subject(s)
Adipose Tissue/immunology , Dendritic Cells/immunology , Inflammation/immunology , Macrophages/immunology , Obesity/immunology , Animals , Cells, Cultured , Diet, High-Fat , Gene Expression Profiling , Humans , Immunophenotyping , Insulin Resistance , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, CCR2/genetics , Receptors, CCR7/genetics , Receptors, IgG/metabolism
13.
Obesity (Silver Spring) ; 24(3): 597-605, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26916240

ABSTRACT

OBJECTIVE: The relationship between adipose tissue fibrosis, adipocyte hypertrophy, and preadipocyte hyperplasia in the context of obesity and the correlation of these tissue-based phenomena with systemic metabolic disease are poorly defined. The goal of this study was to clarify the relationship between adipose tissue fibrosis, adipocyte hypertrophy, and preadipocyte hyperplasia in human obesity and determine the correlation of these adipose-tissue based phenomena with diabetes. METHODS: Visceral and subcutaneous adipose tissues from humans with obesity collected during bariatric surgery were studied with QRTPCR, immunohistochemistry, and flow cytometry for expression of collagens and fibrosis-related proteins, adipocyte size, and preadipocyte frequency. Results were correlated with clinical characteristics including diabetes status. RESULTS: Fibrosis was decreased, hypertrophy was increased, and preadipocyte frequency and fibrotic gene expression were decreased in adipose tissues from diabetic subjects compared to non-diabetic subjects. These differences were greater in visceral compared to subcutaneous adipose tissue. CONCLUSIONS: These data are consistent with the hypothesis that adipose tissue fibrosis in the context of human obesity limits adipocyte hypertrophy and is associated with a reciprocal increase in adipocyte hyperplasia, with beneficial effects on systemic metabolism. These findings suggest adipose tissue fibrosis as a potential target for manipulation of adipocyte metabolism.


Subject(s)
Adipocytes/metabolism , Adipose Tissue/metabolism , Diabetes Mellitus, Type 2/metabolism , Hyperplasia/metabolism , Obesity/metabolism , Bariatric Surgery , Female , Fibrosis , Humans , Hypertrophy/metabolism , Male , Middle Aged , Subcutaneous Fat/metabolism
14.
J Leukoc Biol ; 99(6): 1107-19, 2016 06.
Article in English | MEDLINE | ID: mdl-26658005

ABSTRACT

Obesity activates both innate and adaptive immune responses in adipose tissue, but the mechanisms critical for regulating these responses remain unknown. CD40/CD40L signaling provides bidirectional costimulatory signals between antigen-presenting cells and CD4(+) T cells, and CD40L expression is increased in obese humans. Therefore, we examined the contribution of CD40 to the progression of obesity-induced inflammation in mice. CD40 was highly expressed on adipose tissue macrophages in mice, and CD40/CD40L signaling promoted the expression of antigen-presenting cell markers in adipose tissue macrophages. When fed a high fat diet, Cd40-deficient mice had reduced accumulation of conventional CD4(+) T cells (Tconv: CD3(+)CD4(+)Foxp3(-)) in visceral fat compared with wild-type mice. By contrast, the number of regulatory CD4(+) T cells (Treg: CD3(+)CD4(+)Foxp3(+)) in lean and obese fat was similar between wild-type and knockout mice. Adipose tissue macrophage content and inflammatory gene expression in fat did not differ between obese wild-type and knockout mice; however, major histocompatibility complex class II and CD86 expression on adipose tissue macrophages was reduced in visceral fat from knockout mice. Similar results were observed in chimeric mice with hematopoietic Cd40-deficiency. Nonetheless, neither whole body nor hematopoietic disruption of CD40 ameliorated obesity-induced insulin resistance in mice. In human adipose tissue, CD40 expression was positively correlated with CD80 and CD86 expression in obese patients with type 2 diabetes. These findings indicate that CD40 signaling in adipose tissue macrophages regulates major histocompatibility complex class II and CD86 expression to control the expansion of CD4(+) T cells; however, this is largely dispensable for the development of obesity-induced inflammation and insulin resistance in mice.


Subject(s)
Adipose Tissue/pathology , CD4-Positive T-Lymphocytes/metabolism , CD40 Antigens/metabolism , Histocompatibility Antigens Class II/metabolism , Macrophages/metabolism , Obesity/immunology , Adiposity/drug effects , Animals , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/metabolism , B7-1 Antigen/metabolism , B7-2 Antigen/metabolism , CD4-Positive T-Lymphocytes/drug effects , CD40 Ligand/metabolism , Diet, High-Fat , Hematopoiesis/drug effects , Humans , Insulin/pharmacology , Macrophages/drug effects , Male , Mice, Inbred C57BL , Obesity/pathology , Omentum/drug effects , Omentum/metabolism , Omentum/pathology , Signal Transduction/drug effects
15.
Adipocyte ; 4(4): 264-72, 2015.
Article in English | MEDLINE | ID: mdl-26451282

ABSTRACT

The extracellular matrix (ECM) plays important roles in maintaining adequate adipose tissue function and in metabolic regulation. Here we have examined the organization of a relatively unexplored adipose tissue ECM component, elastin and its response to diet induced obesity in mice. Additionally, we have explored the regulation and requirement of macrophage metalloelastase, MMP-12, in adipose tissue ECM remodeling in obesity. In visceral fat depots, elastin fibers form a mesh-like net that becomes denser with diet-induced obesity. In contrast, the elastin fibers in subcutaneous adipose depots are more linear in organization, and are tightly associated with adipose tissue macrophages (ATMs). We found that Mmp12 is produced predominantly by ATMs and can be induced with both short- and long-term high fat diet challenge and rapid remodeling induced by lipolysis. This contrasts with Mmp14 and Timp1 which are further induced only after chronic obesity in non-ATM populations. We examined obese transgenic Mmp12 (-/-) mice and found an increase in gene expression of ECM genes with diet-induced obesity, but showed few significant differences in metabolic parameters, elastin matrix density, or in adipose tissue inflammation. Together, these studies reveal the architecture and diet-induced regulation of the elastin matrix and suggest that MMP-12 is not required for elastin matrix remodeling or for the metabolic dysfunction that occurs with obesity.

16.
J Biol Chem ; 290(21): 13250-62, 2015 May 22.
Article in English | MEDLINE | ID: mdl-25869128

ABSTRACT

Women of reproductive age are protected from metabolic disease relative to postmenopausal women and men. Most preclinical rodent studies are skewed toward the use of male mice to study obesity-induced metabolic dysfunction because of a similar protection observed in female mice. How sex differences in obesity-induced inflammatory responses contribute to these observations is unknown. We have compared and contrasted the effects of high fat diet-induced obesity on glucose metabolism and leukocyte activation in multiple depots in male and female C57Bl/6 mice. With both short term and long term high fat diet, male mice demonstrated increased weight gain and CD11c(+) adipose tissue macrophage content compared with female mice despite similar degrees of adipocyte hypertrophy. Competitive bone marrow transplant studies demonstrated that obesity induced a preferential contribution of male hematopoietic cells to circulating leukocytes and adipose tissue macrophages compared with female cells independent of the sex of the recipient. Sex differences in macrophage and hematopoietic cell in vitro activation in response to obesogenic cues were observed to explain these results. In summary, this report demonstrates that male and female leukocytes and hematopoietic stem cells have cell-autonomous differences in their response to obesity that contribute to an amplified response in males compared with females.


Subject(s)
Blood Glucose/metabolism , Diet, High-Fat/adverse effects , Hematopoietic Stem Cells/cytology , Inflammation/immunology , Obesity/etiology , Adipose Tissue/cytology , Adipose Tissue/immunology , Adipose Tissue/metabolism , Animals , Biomarkers/analysis , Cells, Cultured , Colony-Forming Units Assay , Female , Flow Cytometry , Glucose Tolerance Test , Hematopoietic Stem Cells/metabolism , Immunohistochemistry , Inflammation/complications , Inflammation/pathology , Lipids/analysis , Male , Mice , Mice, Inbred C57BL , Myelopoiesis/physiology , Obesity/metabolism , Obesity/pathology , Sex Factors , Weight Gain
17.
Cell Rep ; 9(2): 605-17, 2014 Oct 23.
Article in English | MEDLINE | ID: mdl-25310975

ABSTRACT

An adaptive immune response triggered by obesity is characterized by the activation of adipose tissue CD4(+) T cells by unclear mechanisms. We have examined whether interactions between adipose tissue macrophages (ATMs) and CD4(+) T cells contribute to adipose tissue metainflammation. Intravital microscopy identifies dynamic antigen-dependent interactions between ATMs and T cells in visceral fat. Mice deficient in major histocompatibility complex class II (MHC II) showed protection from diet-induced obesity. Deletion of MHC II expression in macrophages led to an adipose tissue-specific decrease in the effector/memory CD4(+) T cells, attenuation of CD11c(+) ATM accumulation, and improvement in glucose intolerance by increasing adipose tissue insulin sensitivity. Ablation experiments demonstrated that the maintenance of proliferating conventional T cells is dependent on signals from CD11c(+) ATMs in obese mice. These studies demonstrate the importance of MHCII-restricted signals from ATMs that regulate adipose tissue T cell maturation and metainflammation.


Subject(s)
Adipose Tissue/immunology , CD4-Positive T-Lymphocytes/immunology , Genes, MHC Class II , Lymphocyte Activation , Macrophages/immunology , Obesity/immunology , Adipose Tissue/cytology , Animals , CD11c Antigen/metabolism , CD4-Positive T-Lymphocytes/metabolism , Cells, Cultured , Gene Deletion , HLA-DR Antigens/genetics , HLA-DR Antigens/metabolism , Humans , Macrophages/metabolism , Mice , Mice, Inbred C57BL
18.
Mol Metab ; 3(6): 664-75, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25161889

ABSTRACT

Obesity is associated with an activated macrophage phenotype in multiple tissues that contributes to tissue inflammation and metabolic disease. To evaluate the mechanisms by which obesity potentiates myeloid activation, we evaluated the hypothesis that obesity activates myeloid cell production from bone marrow progenitors to potentiate inflammatory responses in metabolic tissues. High fat diet-induced obesity generated both quantitative increases in myeloid progenitors as well as a potentiation of inflammation in macrophages derived from these progenitors. In vivo, hematopoietic stem cells from obese mice demonstrated the sustained capacity to preferentially generate inflammatory CD11c(+) adipose tissue macrophages after serial bone marrow transplantation. We identified that hematopoietic MyD88 was important for the accumulation of CD11c(+) adipose tissue macrophage accumulation by regulating the generation of myeloid progenitors from HSCs. These findings demonstrate that obesity and metabolic signals potentiate leukocyte production and that dietary priming of hematopoietic progenitors contributes to adipose tissue inflammation.

19.
Diabetes ; 62(8): 2762-72, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23493569

ABSTRACT

The proinflammatory activation of leukocytes in adipose tissue contributes to metabolic disease. How crosstalk between immune cells initiates and sustains adipose tissue inflammation remains an unresolved question. We have examined the hypothesis that adipose tissue macrophages (ATMs) interact with and regulate the function of T cells. Dietary obesity was shown to activate the proliferation of effector memory CD4(+) T cells in adipose tissue. Our studies further demonstrate that ATMs are functional antigen-presenting cells that promote the proliferation of interferon-γ-producing CD4(+) T cells in adipose tissue. ATMs from lean and obese visceral fat process and present major histocompatibility complex (MHC) class II-restricted antigens. ATMs were sufficient to promote proliferation and interferon-γ production from antigen-specific CD4(+) T cells in vitro and in vivo. Diet-induced obesity increased the expression of MHC II and T-cell costimulatory molecules on ATMs in visceral fat, which correlated with an induction of T-cell proliferation in that depot. Collectively, these data indicate that ATMs provide a functional link between the innate and adaptive immune systems within visceral fat in mice.


Subject(s)
Adipose Tissue/immunology , Antigen-Presenting Cells/immunology , CD4-Positive T-Lymphocytes/immunology , Macrophages/immunology , Obesity/immunology , Adipose Tissue/metabolism , Animals , Antigen-Presenting Cells/metabolism , CD4-Positive T-Lymphocytes/metabolism , Diet , Glucose Tolerance Test , Inflammation/immunology , Insulin/blood , Insulin Resistance/immunology , Lymphocyte Activation/immunology , Macrophages/metabolism , Mice , Obesity/metabolism , Phagocytosis/immunology
20.
PLoS One ; 8(3): e57929, 2013.
Article in English | MEDLINE | ID: mdl-23472120

ABSTRACT

Neuropeptide Y (NPY) is induced in peripheral tissues such as adipose tissue with obesity. The mechanism and function of NPY induction in fat are unclear. Given the evidence that NPY can modulate inflammation, we examined the hypothesis that NPY regulates the function of adipose tissue macrophages (ATMs) in response to dietary obesity in mice. NPY was induced by dietary obesity in the stromal vascular cells of visceral fat depots from mice. Surprisingly, the induction of Npy was limited to purified ATMs from obese mice. Significant basal production of NPY was observed in cultured bone marrow derived macrophage and dendritic cells (DCs) and was increased with LPS stimulation. In vitro, addition of NPY to myeloid cells had minimal effects on their activation profiles. NPY receptor inhibition promoted DC maturation and the production of IL-6 and TNFα suggesting an anti-inflammatory function for NPY signaling in DCs. Consistent with this, NPY injection into lean mice decreased the quantity of M1-like CD11c(+) ATMs and suppressed Ly6c(hi) monocytes. BM chimeras generated from Npy(-/-) donors demonstrated that hematopoietic NPY contributes to the obesity-induced induction of Npy in fat. In addition, loss of Npy expression from hematopoietic cells led to an increase in CD11c(+) ATMs in visceral fat with high fat diet feeding. Overall, our studies suggest that NPY is produced by a range of myeloid cells and that obesity activates the production of NPY in adipose tissue macrophages with autocrine and paracrine effects.


Subject(s)
Adipose Tissue/cytology , Gene Expression Regulation , Inflammation/metabolism , Macrophages/metabolism , Neuropeptide Y/metabolism , Obesity/metabolism , Animals , Dendritic Cells/cytology , Hematopoietic Stem Cells/cytology , Insulin Resistance , Interleukin-6/metabolism , Macrophages/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Monocytes/cytology , Receptors, Neuropeptide Y/metabolism , Triglycerides/metabolism , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL