Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
2.
Nat Immunol ; 24(5): 757-766, 2023 05.
Article in English | MEDLINE | ID: mdl-37012544

ABSTRACT

Obesity-related metabolic organ inflammation contributes to cardiometabolic disorders. In obese individuals, changes in lipid fluxes and storage elicit immune responses in the adipose tissue (AT), including expansion of immune cell populations and qualitative changes in the function of these cells. Although traditional models of metabolic inflammation posit that these immune responses disturb metabolic organ function, studies now suggest that immune cells, especially AT macrophages (ATMs), also have important adaptive functions in lipid homeostasis in states in which the metabolic function of adipocytes is taxed. Adverse consequences of AT metabolic inflammation might result from failure to maintain local lipid homeostasis and long-term effects on immune cells beyond the AT. Here we review the complex function of ATMs in AT homeostasis and metabolic inflammation. Additionally, we hypothesize that trained immunity, which involves long-term functional adaptations of myeloid cells and their bone marrow progenitors, can provide a model by which metabolic perturbations trigger chronic systemic inflammation.


Subject(s)
Adipose Tissue , Macrophages , Humans , Homeostasis , Obesity , Lipids , Inflammation
3.
JCI Insight ; 8(3)2023 02 08.
Article in English | MEDLINE | ID: mdl-36752206

ABSTRACT

Patients with nonalcoholic steatohepatitis (NASH) have increased expression of liver monocyte chemoattractant protein-1 (MCP-1), but its cellular source and contribution to various aspects of NASH pathophysiology remain debated. We demonstrated increased liver CCL2 (which encodes MCP-1) expression in patients with NASH, and commensurately, a 100-fold increase in hepatocyte Ccl2 expression in a mouse model of NASH, accompanied by increased liver monocyte-derived macrophage (MoMF) infiltrate and liver fibrosis. To test repercussions of increased hepatocyte-derived MCP-1, we generated hepatocyte-specific Ccl2-knockout mice, which showed reduced liver MoMF infiltrate as well as decreased liver fibrosis. Forced hepatocyte MCP-1 expression provoked the opposite phenotype in chow-fed wild-type mice. Consistent with increased hepatocyte Notch signaling in NASH, we observed a close correlation between markers of Notch activation and CCL2 expression in patients with NASH. We found that an evolutionarily conserved Notch/recombination signal binding protein for immunoglobulin kappa J region binding site in the Ccl2 promoter mediated transactivation of the Ccl2 promoter in NASH diet-fed mice. Increased liver MoMF infiltrate and liver fibrosis seen in opposite gain-of-function mice was ameliorated with concomitant hepatocyte Ccl2 knockout or CCR2 inhibitor treatment. Hepatocyte Notch activation prompts MCP-1-dependent increase in liver MoMF infiltration and fibrosis.


Subject(s)
Chemokine CCL2 , Non-alcoholic Fatty Liver Disease , Animals , Mice , Chemokine CCL2/metabolism , Hepatocytes/metabolism , Liver Cirrhosis/pathology , Non-alcoholic Fatty Liver Disease/metabolism
4.
Diabetes ; 71(11): 2395-2401, 2022 11 01.
Article in English | MEDLINE | ID: mdl-35944274

ABSTRACT

The increase of functional ß-cell mass is paramount to maintaining glucose homeostasis in the setting of systemic insulin resistance and/or augmented metabolic load. Understanding compensatory mechanisms that allow ß-cell mass adaptation may allow for the discovery of therapeutically actionable control nodes. In this study, we report the rapid and robust ß-cell hyperplasic effect in a mouse model of overfeeding-induced obesity (OIO) based on direct gastric caloric infusion. By performing RNA sequencing in islets isolated from OIO mice, we identified Sin3a as a novel transcriptional regulator of ß-cell mass adaptation. ß-Cell-specific Sin3a knockout animals showed profound diabetes due to defective acquisition of postnatal ß-cell mass. These findings reveal a novel regulatory pathway in ß-cell proliferation and validate OIO as a model for discovery of other mechanistic determinants of ß-cell adaptation.


Subject(s)
Insulin Resistance , Insulin-Secreting Cells , Mice , Animals , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Obesity/genetics , Obesity/metabolism , Disease Models, Animal , Glucose/metabolism
5.
Cell Rep ; 37(3): 109845, 2021 10 19.
Article in English | MEDLINE | ID: mdl-34686319

ABSTRACT

In mammals, changes in weight elicit responses that favor a return to one's previous weight and promote weight stability. It has been hypothesized that palatable sweet and high-fat foods disturb the defense of body weight, leading to weight gain. We find that increasing sweetness or percent calories from fat increases diet palatability but that only increases in nutritive fat content increase caloric intake and body weight. In a mouse model of overfeeding that activates weight defense, high-fat diets, but not sweetened diets, attenuate the defense of body weight, leading to weight gain. The ability of a palatable, high-fat diet to increase food intake does not require tasting or smelling the food. Instead, the direct infusion of a high-fat diet into the stomach increases the ad libitum intake of less palatable, low-fat food. Post-oral sensing of percent calories from fat modulates feeding behavior to alter weight stability.


Subject(s)
Dietary Fats/administration & dosage , Dietary Sugars/administration & dosage , Eating , Energy Intake , Feeding Behavior , Taste , Weight Gain , Animal Feed , Animals , Food Preferences , Male , Mice, Inbred C57BL , Smell , Time Factors
7.
J Vis Exp ; (169)2021 03 28.
Article in English | MEDLINE | ID: mdl-33843938

ABSTRACT

The physiological and pathophysiological roles of extracellular vesicles (EVs) have become increasingly recognized, making the EV field a quickly evolving area of research. There are many different methods for EV isolation, each with distinct advantages and disadvantages that affect the downstream yield and purity of EVs. Thus, characterizing the EV prep isolated from a given source by a chosen method is important for interpretation of downstream results and comparison of results across laboratories. Various methods exist for determining the size and quantity of EVs, which can be altered by disease states or in response to external conditions. Nanoparticle tracking analysis (NTA) is one of the prominent technologies used for high-throughput analysis of individual EVs. Here, we present a detailed protocol for quantification and size determination of EVs isolated from mouse perigonadal adipose tissue and human plasma using a breakthrough technology for NTA representing major advances in the field. The results demonstrate that this method can deliver reproducible and valid total particle concentration and size distribution data for EVs isolated from different sources using different methods, as confirmed by transmission electron microscopy. The adaptation of this instrument for NTA will address the need for standardization in NTA methods to increase rigor and reproducibility in EV research.


Subject(s)
Extracellular Vesicles/metabolism , Humans , Nanoparticles , Reproducibility of Results
8.
Obesity (Silver Spring) ; 29 Suppl 1: S31-S38, 2021 04.
Article in English | MEDLINE | ID: mdl-33759394

ABSTRACT

Although many individuals achieve weight loss of 10% or more, the ability to maintain a reduced body mass over months and years is much rarer. Unfortunately, our understanding of the adverse consequences of having overweight and obesity argues that long-term maintenance of a reduced weight provides the greatest health benefit. However, to achieve long-term weight reduction requires overcoming neuroendocrine systems that favor restoration of one's initial weight. Identifying and characterizing the components of these systems will be important if we are to develop therapies and strategies to reduce the rates of obesity and its complications in our modern society. During this session, Eric Ravussin and Steven R. Smith, respectively, discussed the physiology of the weight-reduced state that favors weight regain and a molecular component that contributes to this response.


Subject(s)
Energy Metabolism/physiology , Weight Loss/physiology , Humans , National Institute of Diabetes and Digestive and Kidney Diseases (U.S.)/organization & administration , Obesity/metabolism , Obesity/physiopathology , Obesity/therapy , Overweight/metabolism , Overweight/physiopathology , Overweight/therapy , United States
9.
Cell ; 183(7): 1739-1741, 2020 12 23.
Article in English | MEDLINE | ID: mdl-33357398

ABSTRACT

In this issue of Cell, Ringel et al. reveal a link between lipid utilization in the tumor microenvironment and anti-tumor immunity in obese mice. These findings provide one explanation for how obesity worsens cancer outcomes and may point to a new metabolic approach to treating some cancers.


Subject(s)
Neoplasms , Tumor Microenvironment , Animals , Mice , Mice, Obese , Obesity , T-Lymphocytes
10.
Ann Intern Med ; 173(10): 782-790, 2020 11 17.
Article in English | MEDLINE | ID: mdl-32726151

ABSTRACT

BACKGROUND: Obesity is a risk factor for pneumonia and acute respiratory distress syndrome. OBJECTIVE: To determine whether obesity is associated with intubation or death, inflammation, cardiac injury, or fibrinolysis in coronavirus disease 2019 (COVID-19). DESIGN: Retrospective cohort study. SETTING: A quaternary academic medical center and community hospital in New York City. PARTICIPANTS: 2466 adults hospitalized with laboratory-confirmed severe acute respiratory syndrome coronavirus 2 infection over a 45-day period with at least 47 days of in-hospital observation. MEASUREMENTS: Body mass index (BMI), admission biomarkers of inflammation (C-reactive protein [CRP] level and erythrocyte sedimentation rate [ESR]), cardiac injury (troponin level), and fibrinolysis (D-dimer level). The primary end point was a composite of intubation or death in time-to-event analysis. RESULTS: Over a median hospital length of stay of 7 days (interquartile range, 3 to 14 days), 533 patients (22%) were intubated, 627 (25%) died, and 59 (2%) remained hospitalized. Compared with overweight patients, patients with obesity had higher risk for intubation or death, with the highest risk among those with class 3 obesity (hazard ratio, 1.6 [95% CI, 1.1 to 2.1]). This association was primarily observed among patients younger than 65 years and not in older patients (P for interaction by age = 0.042). Body mass index was not associated with admission levels of biomarkers of inflammation, cardiac injury, or fibrinolysis. LIMITATIONS: Body mass index was missing for 28% of patients. The primary analyses were conducted with multiple imputation for missing BMI. Upper bounding factor analysis suggested that the results are robust to possible selection bias. CONCLUSION: Obesity is associated with increased risk for intubation or death from COVID-19 in adults younger than 65 years, but not in adults aged 65 years or older. PRIMARY FUNDING SOURCE: National Institutes of Health.


Subject(s)
Betacoronavirus , Body Mass Index , Coronavirus Infections/epidemiology , Intubation, Intratracheal/statistics & numerical data , Obesity/epidemiology , Pneumonia, Viral/epidemiology , Academic Medical Centers , Age Factors , Aged , Biomarkers/blood , Blood Sedimentation , C-Reactive Protein/analysis , COVID-19 , Cohort Studies , Female , Fibrin Fibrinogen Degradation Products/analysis , Hospitalization , Hospitals, Community , Humans , Length of Stay/statistics & numerical data , Male , Middle Aged , New York City/epidemiology , Pandemics , Proportional Hazards Models , Retrospective Studies , SARS-CoV-2 , Troponin/blood
11.
Mol Metab ; 34: 146-156, 2020 04.
Article in English | MEDLINE | ID: mdl-32180554

ABSTRACT

OBJECTIVE: In mouse models, deficiency of TTC39B (T39) decreases hepatic lipogenic gene expression and protects against diet-induced steatohepatitis. While assessing the therapeutic potential of antisense oligonucleotides (ASOs) targeting T39, we discovered an unexpected weight loss phenotype. The objective of this study was to determine the mechanism of the resistance to diet-induced obesity. METHODS: To assess therapeutic potential, we used antisense oligonucleotides (ASO) to knock down T39 expression in a Western or high-fat, high-cholesterol, high-sucrose-diet-fed Ldlr-/- or wild-type mice. RESULTS: T39 ASO treatment led to decreased hepatic lipogenic gene expression and decreased hepatic triglycerides. Unexpectedly, T39 ASO treatment protected against diet-induced obesity. The reduced weight gain was seen with two different ASOs that decreased T39 mRNA in adipose tissue macrophages (ATMs), but not with a liver-targeted GalNac-ASO. Mice treated with the T39 ASO displayed increased browning of gonadal white adipose tissue (gWAT) and evidence of increased lipolysis. However, T39 knockout mice displayed a similar weight loss response when treated with T39 ASO, indicating an off-target effect. RNA-seq analysis of gWAT showed a widespread increase in type I interferon (IFN)-responsive genes, and knockout of the IFN receptor abolished the weight loss phenotype induced by the T39 ASO. Some human T39 ASOs and ASOs with different modifications targeting LDLR also induced a type I IFN response in THP1 macrophages. CONCLUSION: Our data suggest that extrahepatic targeting of T39 by ASOs in ATMs produced an off-target type 1 IFN response, leading to activation of lipolysis, browning of WAT, and weight loss. While our findings suggest that ASOs may induce off-target type 1 IFN response more commonly than previously thought, they also suggest that therapeutic induction of type 1 IFN selectively in ATMs could potentially represent a novel approach to the treatment of obesity.


Subject(s)
Diet, High-Fat/adverse effects , Interferon Type I/biosynthesis , Obesity/chemically induced , Obesity/drug therapy , Oligonucleotides, Antisense/pharmacology , Animals , Female , Injections, Subcutaneous , Interferon Type I/deficiency , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/metabolism , Obesity/prevention & control , Oligonucleotides, Antisense/administration & dosage
12.
Cell Stem Cell ; 24(4): 654-669.e6, 2019 04 04.
Article in English | MEDLINE | ID: mdl-30930146

ABSTRACT

Hair growth can be induced from resting mouse hair follicles by topical application of JAK inhibitors, suggesting that JAK-STAT signaling is required for maintaining hair follicle stem cells (HFSCs) in a quiescent state. Here, we show that Oncostatin M (OSM), an IL-6 family cytokine, negatively regulates hair growth by signaling through JAK-STAT5 to maintain HFSC quiescence. Genetic deletion of the OSM receptor or STAT5 can induce premature HFSC activation, suggesting that the resting telogen stage is actively maintained by the hair follicle niche. Single-cell RNA sequencing revealed that the OSM source is not intrinsic to the hair follicle itself and is instead a subset of TREM2+ macrophages that is enriched within the resting follicle and deceases immediately prior to HFSC activation. In vivo inhibition of macrophage function was sufficient to induce HFSC proliferation and hair cycle induction. Together these results clarify how JAK-STAT signaling actively inhibits hair growth.


Subject(s)
Hair Follicle/cytology , Hair/growth & development , Macrophages/metabolism , Oncostatin M/metabolism , Stem Cells/cytology , Animals , Cell Cycle , Cell Proliferation , Dermis/cytology , Dermis/metabolism , Female , Hair Follicle/metabolism , Humans , Janus Kinase 2/metabolism , Male , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Immunologic/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction , Stem Cells/metabolism
13.
Science ; 363(6430): 989-993, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30819964

ABSTRACT

To meet systemic metabolic needs, adipocytes release fatty acids and glycerol through the action of neutral lipases. Here, we describe a secondary pathway of lipid release from adipocytes that is independent of canonical lipolysis. We found that adipocytes release exosome-sized, lipid-filled vesicles (AdExos) that become a source of lipid for local macrophages. Adipose tissue from lean mice released ~1% of its lipid content per day via exosomes ex vivo, a rate that more than doubles in obese animals. AdExos and associated factors were sufficient to induce in vitro differentiation of bone marrow precursors into adipose tissue macrophage-like cells. Thus, AdExos are both an alternative pathway of local lipid release and a mechanism by which parenchymal cells can modulate tissue macrophage differentiation and function.


Subject(s)
Adipocytes/metabolism , Adipose Tissue/immunology , Exosomes/metabolism , Lipid Metabolism , Macrophages/metabolism , Adipose Tissue/cytology , Animals , Bone Marrow Cells/metabolism , Cell Differentiation , Cells, Cultured , Lipase/metabolism , Lipolysis , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/metabolism
14.
Diabetol Metab Syndr ; 11: 14, 2019.
Article in English | MEDLINE | ID: mdl-30774722

ABSTRACT

BACKGROUND: Plasma albumin is reduced during inflammation. Obesity, a strong risk factor for type 2 diabetes (T2D), is associated with adipose tissue inflammation. However, whether albumin is associated with adipose tissue inflammation and whether it predicts T2D are unclear. METHODS: Adults (predominantly American Indian) from a longitudinal study were included. Macrophage content and gene expression related to recruitment/activation were measured from subcutaneous adipose tissue (n = 51). The relationship between plasma albumin and adiposity (dual-energy X-ray absorptiometry or hydrodensitometry), glucose (oral glucose tolerance test), insulin action (hyperinsulinemic-euglycemic clamp), and insulin secretion (intravenous glucose tolerance test) were evaluated (n = 422). Progression to T2D was evaluated by Cox regression (median follow-up 8.8 years; 102 progressors). RESULTS: Albumin was associated with macrophage markers including C1QB (r = - 0.30, p = 0.04), CSF1R (r = - 0.30, p = 0.03), and CD11b (r = - 0.36, p = 0.01). Albumin was inversely associated with body fat percentage (r = - 0.14, p = 0.003), fasting plasma glucose (r = - 0.17, p = 0.0003), and 2 h plasma glucose (r = - 0.11, p = 0.03), and was reduced in impaired glucose regulation compared with normal glucose regulation (mean ± SD: 39.4 ± 3.6 g/l and 40.1 ± 3.9 g/l, respectively; p = 0.049). Albumin predicted T2D, even after adjustment for confounders (HR, 0.75; 95% CI 0.58-0.96; p = 0.02; per one SD difference in albumin). CONCLUSIONS: Reduced albumin is associated with an unfavorable metabolic profile, characterized by increased adipose tissue inflammation, adiposity, and glucose, and with an increased risk for T2D.

15.
Cell Metab ; 28(2): 289-299.e5, 2018 08 07.
Article in English | MEDLINE | ID: mdl-29937378

ABSTRACT

Weight is defended so that increases or decreases in body mass elicit responses that favor restoration of one's previous weight. While much is known about the signals that respond to weight loss and the central role that leptin plays, the lack of experimental systems studying the overfed state has meant little is known about pathways defending against weight gain. We developed a system to study this physiology and found that overfed mice defend against increased weight gain with graded anorexia but, unlike weight loss, this response is independent of circulating leptin concentration. In overfed mice that are unresponsive to orexigenic stimuli, adipose tissue is transcriptionally and immunologically distinct from fat of ad libitum-fed obese animals. These findings provide evidence that overfeeding-induced obesity alters adipose tissue and central responses in ways that are distinct from ad libitum obesity and activates a non-leptin system to defend against weight gain.


Subject(s)
Adipose Tissue/metabolism , Leptin/physiology , Obesity/metabolism , Weight Gain , Weight Loss , Adipose Tissue/immunology , Animals , Anorexia , Hyperphagia , Mice , Mice, Inbred C57BL , Mice, Obese
16.
Diabetes ; 65(4): 967-80, 2016 04.
Article in English | MEDLINE | ID: mdl-26868294

ABSTRACT

Adipose tissue (AT) macrophages (ATMs) contribute to obesity-induced inflammation and metabolic dysfunction, but also play critical roles in maintaining tissue homeostasis. ATMs catabolize lipid in a lysosomal-dependent manner required for the maintenance of AT; deficiency in lysosomal acid lipase (Lipa), the enzyme required for lysosome lipid catabolism, leads to AT atrophy and severe hepatic steatosis, phenotypes rescued by macrophage-specific expression of Lipa Autophagy delivers cellular products, including lipid droplets, to lysosomes. Given that obesity increases autophagy in AT and contributes to lipid catabolism in other cells, it was proposed that autophagy delivers lipid to lysosomes in ATMs and is required for AT homeostasis. We found that obesity does increase autophagy in ATMs. However, genetic or pharmacological inhibition of autophagy does not alter the lipid balance of ATMs in vitro or in vivo. In contrast to the deficiency of lysosomal lipid hydrolysis, the ablation of autophagy in macrophages does not lead to AT atrophy or alter metabolic phenotypes in lean or obese animals. Although the lysosomal catabolism of lipid is necessary for normal ATM function and AT homeostasis, delivery of lipid to lysosomes is not autophagy dependent and strongly suggests the existence of another lipid delivery pathway critical to lysosome triglyceride hydrolysis in ATMs.


Subject(s)
Adipose Tissue/metabolism , Autophagy/physiology , Lipid Metabolism , Macrophages/metabolism , Adipose Tissue/ultrastructure , Animals , Autophagy/genetics , Autophagy-Related Protein 7/genetics , Cells, Cultured , Homeostasis/physiology , Leptin/deficiency , Leptin/genetics , Lipid Metabolism/genetics , Lipolysis/genetics , Lysosomes/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Sterol Esterase/genetics , Sterol Esterase/metabolism
17.
Obesity (Silver Spring) ; 24(1): 172-83, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26663512

ABSTRACT

OBJECTIVE: Insulin-like growth factor-1 (IGF1) regulates differentiation and growth of tissues and reduces stress and injury. IGF1 also in a tissue-specific manner modulates the differentiation and lipid storage capacity of adipocytes in vitro, but its roles in adipose tissue development and response to stress are not known. METHODS: To study IGF1 in vivo, the cellular sources of adipose tissue Igf1 expression were identified and mice were generated with targeted deletion in adipocytes and macrophages. The effects of adipocyte and macrophage deficiency of IGF1 on adipose tissue development and the response to chronic (high-fat feeding) and acute (cold challenge) stress were studied. RESULTS: The expression of Igf1 by adipose tissue was derived from multiple cell types including adipocytes and macrophages. In lean animals, adipocytes were the primary source of IGF1, but in obesity expression by adipocytes was reduced and by macrophages increased, so as to maintain overall adipose tissue Igf1 expression. Genetic deletion studies revealed that adipocyte-derived IGF1 regulated perigonadal but not subcutaneous adipose tissue mass during high-fat feeding and the development of obesity. Conversely, macrophage-derived IGF1 acutely modulated perigonadal adipose tissue mass during thermogenic challenges. CONCLUSIONS: Local IGF1 is not required in lean adipose tissue development but is required to maintain homeostasis during both chronic and acute metabolic stresses.


Subject(s)
Adipocytes/metabolism , Adipocytes/physiology , Adipose Tissue/metabolism , Homeostasis/physiology , Insulin-Like Growth Factor I/physiology , Macrophages/physiology , Obesity/physiopathology , Stress, Physiological/physiology , Adipogenesis/physiology , Animals , Male , Mice , Mice, Inbred C57BL , Thermogenesis/physiology
18.
Immunity ; 43(4): 624-6, 2015 Oct 20.
Article in English | MEDLINE | ID: mdl-26488811

ABSTRACT

Long studied as modulators of insulin sensitivity, adipose tissue immune cells have recently been implicated in regulating fat mass and weight gain. In this issue of Immunity, Reisner and colleagues (2015) report that ablation of perforin-expressing dendritic cells induces T cell expansion, worsening autoimmunity and surprisingly increasing adiposity.


Subject(s)
Autoimmunity/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Inflammation/immunology , Metabolic Syndrome/immunology , Pore Forming Cytotoxic Proteins/analysis , Animals , Female , Male
19.
PLoS One ; 10(8): e0135842, 2015.
Article in English | MEDLINE | ID: mdl-26317499

ABSTRACT

Obesity-induced inflammation in visceral adipose tissue (VAT) is a major contributor to insulin resistance and type 2 diabetes. Whereas innate immune cells, notably macrophages, contribute to visceral adipose tissue (VAT) inflammation and insulin resistance, the role of adaptive immunity is less well defined. To address this critical gap, we used a model in which endogenous activation of T cells was suppressed in obese mice by blocking MyD88-mediated maturation of CD11c+ antigen-presenting cells. VAT CD11c+ cells from Cd11cCre+Myd88fl/fl vs. control Myd88fl/fl mice were defective in activating T cells in vitro, and VAT T and B cell activation was markedly reduced in Cd11cCre+Myd88fl/fl obese mice. However, neither macrophage-mediated VAT inflammation nor systemic inflammation were altered in Cd11cCre+Myd88fl/fl mice, thereby enabling a focused analysis on adaptive immunity. Unexpectedly, fasting blood glucose, plasma insulin, and the glucose response to glucose and insulin were completely unaltered in Cd11cCre+Myd88fl/fl vs. control obese mice. Thus, CD11c+ cells activate VAT T and B cells in obese mice, but suppression of this process does not have a discernible effect on macrophage-mediated VAT inflammation or systemic glucose homeostasis.


Subject(s)
Immunity, Innate , Insulin Resistance , Intra-Abdominal Fat/immunology , Lymphocyte Activation , Macrophages/immunology , Obesity/immunology , Animals , CD11 Antigens/genetics , CD11 Antigens/metabolism , Intra-Abdominal Fat/cytology , Lymphocyte Subsets/immunology , Male , Mice , Mice, Inbred C57BL , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Obesity/metabolism
20.
Cell Metab ; 20(4): 565-72, 2014 Oct 07.
Article in English | MEDLINE | ID: mdl-25295786

ABSTRACT

Mammals regulate fat mass so that increases or reductions in adipose tissue mass activate responses that favor return to one's previous weight. A reduction in fat mass activates a system that increases food intake and reduces energy expenditure; conversely, overfeeding and rapid adipose tissue expansion reduces food intake and increases energy expenditure. With the identification of leptin nearly two decades ago, the central circuit that defends against reductions in body fat was revealed. However, the systems that defend against rapid expansion of fat mass remain largely unknown. Here we review the physiology of the overfed state and evidence for a distinct regulatory system, which unlike the leptin-mediated system, we propose primarily measures a functional aspect of adipose tissue and not total mass per se.


Subject(s)
Adipose Tissue/metabolism , Signal Transduction , Animals , Body Weight , Eating , Energy Metabolism , Humans , Leptin/metabolism , Receptors, Leptin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL