Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
J Clin Invest ; 133(19)2023 10 02.
Article in English | MEDLINE | ID: mdl-37781916

ABSTRACT

The comprehensive assessment of long-term effects of reducing intake of energy (CALERIE-II; NCT00427193) clinical trial established that caloric restriction (CR) in humans lowers inflammation. The identity and mechanism of endogenous CR-mimetics that can be deployed to control obesity-associated inflammation and diseases are not well understood. Our studies have found that 2 years of 14% sustained CR in humans inhibits the expression of the matricellular protein, secreted protein acidic and rich in cysteine (SPARC), in adipose tissue. In mice, adipose tissue remodeling caused by weight loss through CR and low-protein diet feeding decreased, while high-fat diet-induced (HFD-induced) obesity increased SPARC expression in adipose tissue. Inducible SPARC downregulation in adult mice mimicked CR's effects on lowering adiposity by regulating energy expenditure. Deletion of SPARC in adipocytes was sufficient to protect mice against HFD-induced adiposity, chronic inflammation, and metabolic dysfunction. Mechanistically, SPARC activates the NLRP3 inflammasome at the priming step and downregulation of SPARC lowers macrophage inflammation in adipose tissue, while excess SPARC activated macrophages via JNK signaling. Collectively, reduction of adipocyte-derived SPARC confers CR-like metabolic and antiinflammatory benefits in obesity by serving as an immunometabolic checkpoint of inflammation.


Subject(s)
Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Animals , Humans , Mice , Adipose Tissue/metabolism , Diet, High-Fat/adverse effects , Inflammasomes/genetics , Inflammasomes/metabolism , Inflammation/metabolism , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Obesity/metabolism , Osteonectin/genetics , Osteonectin/metabolism
3.
Cell Metab ; 33(11): 2277-2287.e5, 2021 11 02.
Article in English | MEDLINE | ID: mdl-34473956

ABSTRACT

Aging impairs the integrated immunometabolic responses, which have evolved to maintain core body temperature in homeotherms to survive cold stress, infections, and dietary restriction. Adipose tissue inflammation regulates the thermogenic stress response, but how adipose tissue-resident cells instigate thermogenic failure in the aged are unknown. Here, we define alterations in the adipose-resident immune system and identify that type 2 innate lymphoid cells (ILC2s) are lost in aging. Restoration of ILC2 numbers in aged mice to levels seen in adults through IL-33 supplementation failed to rescue old mice from metabolic impairment and increased cold-induced lethality. Transcriptomic analyses revealed intrinsic defects in aged ILC2, and adoptive transfer of adult ILC2s are sufficient to protect old mice against cold. Thus, the functional defects in adipose ILC2s during aging drive thermogenic failure.


Subject(s)
Immunity, Innate , Interleukin-33 , Adipose Tissue , Aging , Animals , Lung , Lymphocytes , Mice , Mice, Inbred C57BL
4.
Elife ; 102021 06 21.
Article in English | MEDLINE | ID: mdl-34151773

ABSTRACT

Increasing age is the strongest predictor of risk of COVID-19 severity and mortality. Immunometabolic switch from glycolysis to ketolysis protects against inflammatory damage and influenza infection in adults. To investigate how age compromises defense against coronavirus infection, and whether a pro-longevity ketogenic diet (KD) impacts immune surveillance, we developed an aging model of natural murine beta coronavirus (mCoV) infection with mouse hepatitis virus strain-A59 (MHV-A59). When inoculated intranasally, mCoV is pneumotropic and recapitulates several clinical hallmarks of COVID-19 infection. Aged mCoV-A59-infected mice have increased mortality and higher systemic inflammation in the heart, adipose tissue, and hypothalamus, including neutrophilia and loss of γδ T cells in lungs. Activation of ketogenesis in aged mice expands tissue protective γδ T cells, deactivates the NLRP3 inflammasome, and decreases pathogenic monocytes in lungs of infected aged mice. These data establish harnessing of the ketogenic immunometabolic checkpoint as a potential treatment against coronavirus infection in the aged.


Subject(s)
Coronavirus Infections/diet therapy , Diet, Ketogenic/methods , Murine hepatitis virus/pathogenicity , Age Factors , Aging , Animals , COVID-19/diet therapy , Coronavirus Infections/metabolism , Coronavirus Infections/mortality , Disease Models, Animal , Glycolysis , Humans , Inflammasomes/metabolism , Ketone Bodies/metabolism , Male , Mice , Mice, Inbred C57BL , Murine hepatitis virus/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , SARS-CoV-2
5.
bioRxiv ; 2020 Sep 12.
Article in English | MEDLINE | ID: mdl-33236006

ABSTRACT

Increasing age is the strongest predictor of risk of COVID-19 severity. Unregulated cytokine storm together with impaired immunometabolic response leads to highest mortality in elderly infected with SARS-CoV-2. To investigate how aging compromises defense against COVID-19, we developed a model of natural murine beta coronavirus (mCoV) infection with mouse hepatitis virus strain MHV-A59 (mCoV-A59) that recapitulated majority of clinical hallmarks of COVID-19. Aged mCoV-A59-infected mice have increased mortality and higher systemic inflammation in the heart, adipose tissue and hypothalamus, including neutrophilia and loss of γδ T cells in lungs. Ketogenic diet increases beta-hydroxybutyrate, expands tissue protective γδ T cells, deactivates the inflammasome and decreases pathogenic monocytes in lungs of infected aged mice. These data underscore the value of mCoV-A59 model to test mechanism and establishes harnessing of the ketogenic immunometabolic checkpoint as a potential treatment against COVID-19 in the elderly. HIGHLIGHTS: - Natural MHV-A59 mouse coronavirus infection mimics COVID-19 in elderly.- Aged infected mice have systemic inflammation and inflammasome activation.- Murine beta coronavirus (mCoV) infection results in loss of pulmonary γδ T cells.- Ketones protect aged mice from infection by reducing inflammation. ETOC BLURB: Elderly have the greatest risk of death from COVID-19. Here, Ryu et al report an aging mouse model of coronavirus infection that recapitulates clinical hallmarks of COVID-19 seen in elderly. The increased severity of infection in aged animals involved increased inflammasome activation and loss of γδ T cells that was corrected by ketogenic diet.

6.
Cell Metab ; 30(6): 1024-1039.e6, 2019 12 03.
Article in English | MEDLINE | ID: mdl-31735593

ABSTRACT

During aging, visceral adiposity is often associated with alterations in adipose tissue (AT) leukocytes, inflammation, and metabolic dysfunction. However, the contribution of AT B cells in immunometabolism during aging is unexplored. Here, we show that aging is associated with an expansion of a unique population of resident non-senescent aged adipose B cells (AABs) found in fat-associated lymphoid clusters (FALCs). AABs are transcriptionally distinct from splenic age-associated B cells (ABCs) and show greater expansion in female mice. Functionally, whole-body B cell depletion restores proper lipolysis and core body temperature maintenance during cold stress. Mechanistically, the age-induced FALC formation, AAB, and splenic ABC expansion is dependent on the Nlrp3 inflammasome. Furthermore, AABs express IL-1R, and inhibition of IL-1 signaling reduces their proliferation and increases lipolysis in aging. These data reveal that inhibiting Nlrp3-dependent B cell accumulation can be targeted to reverse metabolic impairment in aging AT.


Subject(s)
Adipose Tissue , Aging/metabolism , B-Lymphocytes , Homeostasis , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/physiology , Adipose Tissue/metabolism , Adipose Tissue/pathology , Animals , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Body Temperature Regulation , Cold-Shock Response , Female , Interleukin-18/metabolism , Interleukin-1beta/metabolism , Lipolysis , Male , Mice , Receptors, Interleukin-1/metabolism
7.
Cell Rep ; 24(5): 1085-1092.e6, 2018 07 31.
Article in English | MEDLINE | ID: mdl-30067966

ABSTRACT

Inducers of satiety are drug targets for weight loss to mitigate obesity-associated diseases. Nucleobindin-2 (Nucb2) is thought to be post-translationally processed into bioactive nesfatin-1 peptide, which reportedly induces satiety, causes weight loss, and thus improves insulin sensitivity. Here, we show that deletion of Nucb2 did not affect food intake or adiposity and, instead, caused insulin resistance in mice fed a high-fat diet. In addition, ablation of Nucb2 in orexigenic hypothalamic Agrp neurons did not affect food intake, and nesfatin-1 was detectable in serum, despite global deletion of Nucb2 protein. Upon high-fat diet feeding, the loss of Nucb2 exacerbated metabolic inflammation in adipose tissue macrophages in an NFκB-dependent manner without inducing classical M1 or alternative M2-like macrophage polarization. Furthermore, the loss of Nucb2 in myeloid cells but not in adipocytes mediated the insulin resistance in response to a high-fat diet. Our study reveals that Nucb2 links metabolic inflammation to insulin resistance without affecting weight gain and food intake.


Subject(s)
Insulin Resistance , Nucleobindins/genetics , Obesity/metabolism , Adiposity , Animals , Diet, High-Fat/adverse effects , Eating , Macrophages/metabolism , Mice , NF-kappa B/metabolism , Obesity/etiology , Obesity/genetics , Satiation
8.
Nature ; 550(7674): 119-123, 2017 10 05.
Article in English | MEDLINE | ID: mdl-28953873

ABSTRACT

Catecholamine-induced lipolysis, the first step in the generation of energy substrates by the hydrolysis of triglycerides, declines with age. The defect in the mobilization of free fatty acids in the elderly is accompanied by increased visceral adiposity, lower exercise capacity, failure to maintain core body temperature during cold stress, and reduced ability to survive starvation. Although catecholamine signalling in adipocytes is normal in the elderly, how lipolysis is impaired in ageing remains unknown. Here we show that adipose tissue macrophages regulate the age-related reduction in adipocyte lipolysis in mice by lowering the bioavailability of noradrenaline. Unexpectedly, unbiased whole-transcriptome analyses of adipose macrophages revealed that ageing upregulates genes that control catecholamine degradation in an NLRP3 inflammasome-dependent manner. Deletion of NLRP3 in ageing restored catecholamine-induced lipolysis by downregulating growth differentiation factor-3 (GDF3) and monoamine oxidase A (MAOA) that is known to degrade noradrenaline. Consistent with this, deletion of GDF3 in inflammasome-activated macrophages improved lipolysis by decreasing levels of MAOA and caspase-1. Furthermore, inhibition of MAOA reversed the age-related reduction in noradrenaline concentration in adipose tissue, and restored lipolysis with increased levels of the key lipolytic enzymes adipose triglyceride lipase (ATGL) and hormone sensitive lipase (HSL). Our study reveals that targeting neuro-immunometabolic signalling between the sympathetic nervous system and macrophages may offer new approaches to mitigate chronic inflammation-induced metabolic impairment and functional decline.


Subject(s)
Adipocytes/metabolism , Adipose Tissue/metabolism , Aging/metabolism , Catecholamines/metabolism , Inflammasomes/metabolism , Lipolysis , Macrophages/metabolism , Adipose Tissue/cytology , Adipose Tissue/drug effects , Aging/drug effects , Aging/genetics , Animals , Caspase 1/metabolism , Catecholamines/pharmacology , Gene Expression Profiling , Gene Expression Regulation/drug effects , Growth Differentiation Factor 3/deficiency , Growth Differentiation Factor 3/genetics , Growth Differentiation Factor 3/metabolism , Lipase/metabolism , Lipolysis/drug effects , Lipolysis/genetics , Mice , Monoamine Oxidase/metabolism , Monoamine Oxidase Inhibitors/pharmacology , NLR Family, Pyrin Domain-Containing 3 Protein/deficiency , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Norepinephrine/metabolism , Sterol Esterase/metabolism
9.
Cell Rep ; 19(2): 225-234, 2017 04 11.
Article in English | MEDLINE | ID: mdl-28402847

ABSTRACT

In concert with their phagocytic activity, macrophages are thought to regulate the host immunometabolic responses primarily via their ability to produce specific cytokines and metabolites. Here, we show that IL-4-differentiated, M2-like macrophages secrete IGF1, a hormone previously thought to be exclusively produced from liver. Ablation of IGF1 receptors from myeloid cells reduced phagocytosis, increased macrophages in adipose tissue, elevated adiposity, lowered energy expenditure, and led to insulin resistance in mice fed a high-fat diet. The investigation of adipose macrophage phenotype in obese myeloid IGF1R knockout (MIKO) mice revealed a reduction in transcripts associated with M2-like macrophage activation. Furthermore, the MIKO mice infected with helminth Nippostrongylus brasiliensis displayed delayed resolution from infection with normal insulin sensitivity. Surprisingly, cold challenge did not trigger an overt M2-like state and failed to induce tyrosine hydroxylase expression in adipose tissue macrophages of control or MIKO mice. These results show that IGF1 signaling shapes the macrophage-activation phenotype.


Subject(s)
Insulin Resistance/genetics , Insulin-Like Growth Factor I/genetics , Macrophages/immunology , Strongylida Infections/immunology , Adipose Tissue/immunology , Adipose Tissue/metabolism , Adiposity , Animals , Cell Differentiation/immunology , Diet, High-Fat , Insulin Resistance/immunology , Insulin-Like Growth Factor I/immunology , Interleukin-4/immunology , Macrophages/metabolism , Mice , Mice, Knockout , Nippostrongylus/pathogenicity , Phagocytosis/genetics , Signal Transduction/immunology , Strongylida Infections/metabolism , Strongylida Infections/parasitology
10.
Cell Rep ; 14(7): 1571-1580, 2016 Feb 23.
Article in English | MEDLINE | ID: mdl-26876170

ABSTRACT

The hallmarks of age-related immune senescence are chronic inflammation, aberrant expansion of effector memory, and loss of naive T lymphocytes due in part to systemic activation of innate immune sensor NLRP3 inflammasome in myeloid lineage cells. The endogenous mechanisms that regulate inflammasome activation during aging are unknown. Here, we present evidence that growth hormone receptor (GH-R)-dependent downregulation of NLRP3 inflammasome in macrophages is linked to pro-longevity effects that maintain immune system homeostasis in aging. Deletion of GH-R prevented the macrophage-driven age-related activation of inflammasome in response to NLRP3 ligands and also increased the preservation of naive T cells, even in advanced age and with higher IFNγ secretion from effector cells. The mechanism of inflammasome inhibition is linked to autocrine somatotropic axis as ablation of IGF1R in macrophages lowered the NLRP3 inflammasome activation. Together, our findings show that functional somatotropic axis in macrophages controls inflammation, thus linking NLRP3-mediated innate immune signaling to health span and longevity.


Subject(s)
Aging/genetics , Carrier Proteins/genetics , Inflammasomes/genetics , Macrophages/immunology , Receptor, IGF Type 1/genetics , Receptors, Somatotropin/genetics , Aging/immunology , Animals , Autocrine Communication , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Carrier Proteins/immunology , Gene Expression Regulation , Homeostasis/immunology , Immunity, Innate , Immunologic Memory , Inflammasomes/immunology , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Longevity/genetics , Longevity/immunology , Macrophages/cytology , Mice , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Receptor, IGF Type 1/deficiency , Receptor, IGF Type 1/immunology , Receptors, Somatotropin/deficiency , Receptors, Somatotropin/immunology , Signal Transduction , Spleen/cytology , Spleen/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology
11.
Proc Natl Acad Sci U S A ; 113(4): 1026-31, 2016 Jan 26.
Article in English | MEDLINE | ID: mdl-26755598

ABSTRACT

Age-related thymic degeneration is associated with loss of naïve T cells, restriction of peripheral T-cell diversity, and reduced healthspan due to lower immune competence. The mechanistic basis of age-related thymic demise is unclear, but prior evidence suggests that caloric restriction (CR) can slow thymic aging by maintaining thymic epithelial cell integrity and reducing the generation of intrathymic lipid. Here we show that the prolongevity ketogenic hormone fibroblast growth factor 21 (FGF21), a member of the endocrine FGF subfamily, is expressed in thymic stromal cells along with FGF receptors and its obligate coreceptor, ßKlotho. We found that FGF21 expression in thymus declines with age and is induced by CR. Genetic gain of FGF21 function in mice protects against age-related thymic involution with an increase in earliest thymocyte progenitors and cortical thymic epithelial cells. Importantly, FGF21 overexpression reduced intrathymic lipid, increased perithymic brown adipose tissue, and elevated thymic T-cell export and naïve T-cell frequencies in old mice. Conversely, loss of FGF21 function in middle-aged mice accelerated thymic aging, increased lethality, and delayed T-cell reconstitution postirradiation and hematopoietic stem cell transplantation (HSCT). Collectively, FGF21 integrates metabolic and immune systems to prevent thymic injury and may aid in the reestablishment of a diverse T-cell repertoire in cancer patients following HSCT.


Subject(s)
Aging/immunology , Fibroblast Growth Factors/physiology , Immunosenescence , Thymus Gland/immunology , Animals , Mice , Mice, Inbred C57BL , Receptors, Antigen, T-Cell/physiology , T-Lymphocytes/immunology
12.
Nat Med ; 21(3): 263-9, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25686106

ABSTRACT

The ketone bodies ß-hydroxybutyrate (BHB) and acetoacetate (AcAc) support mammalian survival during states of energy deficit by serving as alternative sources of ATP. BHB levels are elevated by starvation, caloric restriction, high-intensity exercise, or the low-carbohydrate ketogenic diet. Prolonged fasting reduces inflammation; however, the impact that ketones and other alternative metabolic fuels produced during energy deficits have on the innate immune response is unknown. We report that BHB, but neither AcAc nor the structurally related short-chain fatty acids butyrate and acetate, suppresses activation of the NLRP3 inflammasome in response to urate crystals, ATP and lipotoxic fatty acids. BHB did not inhibit caspase-1 activation in response to pathogens that activate the NLR family, CARD domain containing 4 (NLRC4) or absent in melanoma 2 (AIM2) inflammasome and did not affect non-canonical caspase-11, inflammasome activation. Mechanistically, BHB inhibits the NLRP3 inflammasome by preventing K(+) efflux and reducing ASC oligomerization and speck formation. The inhibitory effects of BHB on NLRP3 are not dependent on chirality or starvation-regulated mechanisms like AMP-activated protein kinase (AMPK), reactive oxygen species (ROS), autophagy or glycolytic inhibition. BHB blocks the NLRP3 inflammasome without undergoing oxidation in the TCA cycle, and independently of uncoupling protein-2 (UCP2), sirtuin-2 (SIRT2), the G protein-coupled receptor GPR109A or hydrocaboxylic acid receptor 2 (HCAR2). BHB reduces NLRP3 inflammasome-mediated interleukin (IL)-1ß and IL-18 production in human monocytes. In vivo, BHB or a ketogenic diet attenuates caspase-1 activation and IL-1ß secretion in mouse models of NLRP3-mediated diseases such as Muckle-Wells syndrome, familial cold autoinflammatory syndrome and urate crystal-induced peritonitis. Our findings suggest that the anti-inflammatory effects of caloric restriction or ketogenic diets may be linked to BHB-mediated inhibition of the NLRP3 inflammasome.


Subject(s)
3-Hydroxybutyric Acid/pharmacology , Carrier Proteins/antagonists & inhibitors , Caspase 1/drug effects , Cryopyrin-Associated Periodic Syndromes , Inflammasomes/antagonists & inhibitors , Interleukin-1beta/drug effects , Monocytes/drug effects , Adult , Aged , Animals , Diet, Ketogenic , Disease Models, Animal , Female , Humans , Inflammation , Interleukin-18 , Interleukin-1beta/metabolism , Male , Mice , NLR Family, Pyrin Domain-Containing 3 Protein , Potassium/metabolism
13.
J Biol Chem ; 289(20): 14045-55, 2014 May 16.
Article in English | MEDLINE | ID: mdl-24662293

ABSTRACT

Successful adaptation to periods of chronic caloric excess is a highly coordinated event that is critical to the survival and propagation of species. Transcription factor C/ebp homologous protein (Chop) is thought to be an important molecular mediator that integrates nutrient signals to endoplasmic reticulum (ER) stress and innate immune activation. Given that aberrant ER stress response is implicated in inducing metabolic inflammation and insulin resistance, we hypothesized that ER stress target gene Chop integrates immune and metabolic systems to adapt to chronic positive energy balance. Here we report that inactivation of Chop in mice fed a high fat diet led to significant increase in obesity caused by a reduction in energy expenditure without any change in food intake. Importantly, ablation of Chop does not induce metabolically healthy obesity, because Chop-deficient mice fed a high fat diet had increased hepatic steatosis with significantly higher insulin resistance. Quantification of adipose tissue leukocytosis revealed that elimination of Chop during obesity led to substantial increase in number of adipose tissue T and B lymphocytes. In addition, deficiency of Chop led to increase in total number of myeloid subpopulations like neutrophils and F4/80(+) adipose tissue macrophages without any alterations in the frequency of M1- or M2-like adipose tissue macrophages. Further investigation of inflammatory mechanisms revealed that ablation of Chop increases the sensitivity of macrophages to inflammasome-induced activation of IL-ß in macrophages. Our findings indicate that regulated expression of Chop during obesity is critical for adaptation to chronic caloric excess and maintenance of energy homeostasis via integration of metabolic and immune systems.


Subject(s)
Adipose Tissue/immunology , Leukocytosis/immunology , Leukocytosis/metabolism , Obesity/immunology , Obesity/metabolism , Transcription Factor CHOP/metabolism , Animals , Energy Metabolism , Gene Deletion , Inflammasomes/metabolism , Macrophages/immunology , Male , Mice , T-Lymphocytes/immunology , Transcription Factor CHOP/deficiency , Transcription Factor CHOP/genetics
14.
Cell Metab ; 18(4): 519-32, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-24093676

ABSTRACT

Despite a wealth of clinical data showing an association between inflammation and degenerative disorders in the elderly, the immune sensors that causally link systemic inflammation to aging remain unclear. Here we detail a mechanism by which the Nlrp3 inflammasome controls systemic low-grade age-related "sterile" inflammation in both periphery and brain independently of the noncanonical caspase-11 inflammasome. Ablation of Nlrp3 inflammasome protected mice from age-related increases in the innate immune activation, alterations in CNS transcriptome, and astrogliosis. Consistent with the hypothesis that systemic low-grade inflammation promotes age-related degenerative changes, the deficient Nlrp3 inflammasome-mediated caspase-1 activity improved glycemic control and attenuated bone loss and thymic demise. Notably, IL-1 mediated only Nlrp3 inflammasome-dependent improvement in cognitive function and motor performance in aged mice. These studies reveal Nlrp3 inflammasome as an upstream target that controls age-related inflammation and offer an innovative therapeutic strategy to lower Nlrp3 activity to delay multiple age-related chronic diseases.


Subject(s)
Aging , Carrier Proteins/metabolism , Inflammasomes/metabolism , Animals , Apoptosis Regulatory Proteins , CARD Signaling Adaptor Proteins , Carrier Proteins/genetics , Caspase 1/metabolism , Caspases/deficiency , Caspases/genetics , Caspases/metabolism , Caspases, Initiator , Cognition/physiology , Cytoskeletal Proteins/deficiency , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Glucose Intolerance , Hippocampus/metabolism , Immunity, Innate , Interleukin-1/metabolism , Mice , Mice, Knockout , Motor Activity/physiology , NLR Family, Pyrin Domain-Containing 3 Protein , Receptors, Interleukin-1/deficiency , Receptors, Interleukin-1/genetics , Receptors, Interleukin-1/metabolism , Signal Transduction , Transcriptome
15.
Methods Mol Biol ; 1040: 195-209, 2013.
Article in English | MEDLINE | ID: mdl-23852606

ABSTRACT

The infiltration of immune cell subsets in adipose tissue termed "adipose tissue leukocytosis" is a critical event in the development of chronic inflammation and obesity-associated comorbidities. Given that a significant proportion of cells in adipose tissue of obese patients are of hematopoietic lineage, the distinct adipose depots represent an uncharacterized immunological organ that can impact metabolic functions. Here, we describe approaches to characterize and isolate leukocytes from the complex adipose tissue microenvironment, to aid mechanistic studies to better understand the role of specific pattern recognition receptors (PRRs) such as inflammasomes in adipose-immune cross talk.


Subject(s)
Adipose Tissue/pathology , Leukocytosis/diagnosis , Obesity/pathology , Animals , Flow Cytometry , Humans , Immunophenotyping , Intra-Abdominal Fat/pathology , Leukocytes/cytology , Leukocytes/metabolism , Mice , Obesity/immunology
16.
Age (Dordr) ; 35(4): 1091-104, 2013 Aug.
Article in English | MEDLINE | ID: mdl-22661299

ABSTRACT

Calorie restriction (CR) remains the most robust metabolic intervention to extend lifespan and improve healthspan in several species. Using global and targeted mass spectrometry-based metabolomics approaches, here we show that chronic CR prevents age-related changes in specific metabolic signatures. Global metabolomic analysis using ultra-performance liquid chromatography-tandem mass spectrometry detected more than 7,000 metabolites in sera from ad-libitum-fed young, aged, and aged C57BL/6 mice maintained on 40 % CR. Multivariate statistical analysis of mass spectrometry data revealed a clear separation among the young, aged, and aged-CR mice demonstrating the potential of this approach for producing reliable metabolic profiles that discriminate based on age and diet. We have identified 168 discriminating features with high statistical significance (p ≤ 0.001) and validated and quantified three of these metabolites using targeted metabolite analysis. Calorie restriction prevented the age-related alteration in specific metabolites, namely lysophosphatidylcholines (16:1 and 18:4), sphingomyelin (d18:1/12:0), tetracosahexaenoic acid, and 7α-dihydroxy-4-cholesten-3-one, in the serum. Pathway analysis revealed that CR impacted the age-related changes in metabolic byproducts of lipid metabolism, fatty acid metabolism, and bile acid biosynthesis. Our data suggest that metabolomics approach has the potential to elucidate the metabolic mechanism of CR's potential anti-aging effects in larger-scale investigations.


Subject(s)
Aging/blood , Caloric Restriction , Lipid Metabolism/physiology , Metabolome/physiology , Animals , Chromatography, High Pressure Liquid , Disease Models, Animal , Female , Mass Spectrometry , Mice , Mice, Inbred C57BL
17.
Cell Rep ; 1(1): 56-68, 2012 Jan 26.
Article in English | MEDLINE | ID: mdl-22832107

ABSTRACT

The collapse of thymic stromal cell microenvironment with age and resultant inability of the thymus to produce naive T cells contributes to lower immune-surveillance in the elderly. Here we show that age-related increase in 'lipotoxic danger signals' such as free cholesterol (FC) and ceramides, leads to thymic caspase-1 activation via the Nlrp3 inflammasome. Elimination of Nlrp3 and Asc, a critical adaptor required for inflammasome assembly, reduces age-related thymic atrophy and results in an increase in cortical thymic epithelial cells, T cell progenitors and maintenance of T cell repertoire diversity. Using a mouse model of irradiation and hematopoietic stem cell transplantation (HSCT), we show that deletion of the Nlrp3 inflammasome accelerates T cell reconstitution and immune recovery in middle-aged animals. Collectively, these data demonstrate that lowering inflammasome-dependent caspase-1 activation increases thymic lymphopoiesis and suggest that Nlrp3 inflammasome inhibitors may aid the re-establishment of a diverse T cell repertoire in middle-aged or elderly patients undergoing HSCT.


Subject(s)
Aging/immunology , Carrier Proteins/metabolism , Inflammasomes/metabolism , Thymus Gland/growth & development , Thymus Gland/immunology , Aging/drug effects , Animals , Apoptosis/drug effects , Apoptosis/immunology , Caspase 1/metabolism , Cellular Microenvironment , Cellular Senescence/drug effects , Cellular Senescence/immunology , Ceramides/metabolism , Cholesterol/metabolism , Enzyme Activation/drug effects , Epithelial Cells/drug effects , Epithelial Cells/immunology , Epithelial Cells/pathology , Gene Deletion , Hematopoietic Stem Cell Transplantation , Interleukin-18/metabolism , Interleukin-1beta/metabolism , Lipids/toxicity , Mice , NLR Family, Pyrin Domain-Containing 3 Protein , Organ Size/drug effects , Stem Cells/drug effects , Stem Cells/pathology , T-Lymphocytes/drug effects , T-Lymphocytes/pathology , Thymocytes/drug effects , Thymocytes/immunology , Thymocytes/pathology , Thymus Gland/enzymology , Thymus Gland/pathology
18.
Endocrinology ; 152(11): 4039-45, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21862613

ABSTRACT

Clinical evidence that the blockade of IL-1ß in type-2 diabetic patients improves glycemia is indicative of an autoinflammatory mechanism that may trigger adiposity-driven pancreatic damage. IL-1ß is a key contributor to the obesity-induced inflammation and subsequent insulin resistance, pancreatic ß-cell dysfunction, and the onset of type 2 diabetes. Our previous studies demonstrated that the ceramides activate the Nod-like receptor family, pyrin domain containing 3 (Nlrp3) inflammasome to cause the generation of mature IL-1ß and ablation of the Nlrp3 inflammasome in diet-induced obesity improves insulin signaling. However, it remains unclear whether the posttranslational processing of active IL-1ß in pancreas is regulated by the NLRP3 inflammasome or whether the alternate mechanisms play a dominant role in chronic obesity-induced pancreatic ß-cell exhaustion. Here we show that loss of ASC, a critical adaptor required for the assembly of the NLRP3 and absent in melanoma 2 inflammasome substantially improves the insulin action. Surprisingly, despite lower insulin resistance in the chronically obese NLRP3 and ASC knockout mice, the insulin levels were substantially higher when the inflammasome pathway was eliminated. The obesity-induced increase in maturation of pancreatic IL-1ß and pancreatic islet fibrosis was dependent on the NLRP3 inflammasome activation. Furthermore, elimination of NLRP3 inflammasome protected the pancreatic ß-cells from cell death caused by long-term high-fat feeding during obesity with significant increase in the size of the islets of Langerhans. Collectively, this study provides direct in vivo evidence that activation of the NLRP3 inflammasome in diet-induced obesity is a critical trigger in causing pancreatic damage and is an important mechanism of progression toward type 2 diabetes.


Subject(s)
Carrier Proteins/metabolism , Cytoskeletal Proteins/metabolism , Inflammasomes/metabolism , Obesity/metabolism , Pancreas/metabolism , Animals , Apoptosis Regulatory Proteins , CARD Signaling Adaptor Proteins , Carrier Proteins/genetics , Cytoskeletal Proteins/genetics , Fibrosis , Inflammasomes/genetics , Inflammation/metabolism , Inflammation/pathology , Insulin/blood , Insulin Resistance/physiology , Interleukin-1beta/metabolism , Leptin/blood , Mice , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Obesity/genetics , Obesity/pathology , Pancreas/pathology
19.
Nat Med ; 17(2): 179-88, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21217695

ABSTRACT

The emergence of chronic inflammation during obesity in the absence of overt infection or well-defined autoimmune processes is a puzzling phenomenon. The Nod-like receptor (NLR) family of innate immune cell sensors, such as the nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3 (Nlrp3, but also known as Nalp3 or cryopyrin) inflammasome are implicated in recognizing certain nonmicrobial originated 'danger signals' leading to caspase-1 activation and subsequent interleukin-1ß (IL-1ß) and IL-18 secretion. We show that calorie restriction and exercise-mediated weight loss in obese individuals with type 2 diabetes is associated with a reduction in adipose tissue expression of Nlrp3 as well as with decreased inflammation and improved insulin sensitivity. We further found that the Nlrp3 inflammasome senses lipotoxicity-associated increases in intracellular ceramide to induce caspase-1 cleavage in macrophages and adipose tissue. Ablation of Nlrp3 in mice prevents obesity-induced inflammasome activation in fat depots and liver as well as enhances insulin signaling. Furthermore, elimination of Nlrp3 in obese mice reduces IL-18 and adipose tissue interferon-γ (IFN-γ) expression, increases naive T cell numbers and reduces effector T cell numbers in adipose tissue. Collectively, these data establish that the Nlrp3 inflammasome senses obesity-associated danger signals and contributes to obesity-induced inflammation and insulin resistance.


Subject(s)
Carrier Proteins/physiology , Inflammasomes/physiology , Inflammation/physiopathology , Insulin Resistance/physiology , Obesity/physiopathology , Adipose Tissue/metabolism , Adipose Tissue/physiopathology , Animals , Caspase 1/physiology , Disease Models, Animal , Female , Glucose Tolerance Test , Humans , Inflammation/metabolism , Insulin/physiology , Interleukin-1beta/physiology , Macrophages/physiology , Male , Mice , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein , Obesity/metabolism , Reverse Transcriptase Polymerase Chain Reaction
20.
J Immunol ; 185(3): 1836-45, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20581149

ABSTRACT

Emerging evidence suggests that increases in activated T cell populations in adipose tissue may contribute toward obesity-associated metabolic syndrome. The present study investigates three unanswered questions: 1) Do adipose-resident T cells (ARTs) from lean and obese mice have altered cytokine production in response to TCR ligation?; 2) Do the extralymphoid ARTs possess a unique TCR repertoire compared with lymphoid-resident T cells and whether obesity alters the TCR diversity in specific adipose depots?; and 3) Does short-term elimination of T cells in epididymal fat pad without disturbing the systemic T cell homeostasis regulate inflammation and insulin-action during obesity? We found that obesity reduced the frequency of naive ART cells in s.c. fat and increased the effector-memory populations in visceral fat. The ARTs from diet-induced obese (DIO) mice had a higher frequency of IFN-gamma(+), granzyme B(+) cells, and upon TCR ligation, the ARTs from DIO mice produced increased levels of proinflammatory mediators. Importantly, compared with splenic T cells, ARTs exhibited markedly restricted TCR diversity, which was further compromised by obesity. Acute depletion of T cells from epididymal fat pads improved insulin action in young DIO mice but did not reverse obesity-associated feed forward cascade of chronic systemic inflammation and insulin resistance in middle-aged DIO mice. Collectively, these data establish that ARTs have a restricted TCR-Vbeta repertoire, and T cells contribute toward the complex proinflammatory microenvironment of adipose tissue in obesity. Development of future long-term T cell depletion protocols specific to visceral fat may represent an additional strategy to manage obesity-associated comorbidities.


Subject(s)
Adipose Tissue/immunology , Adipose Tissue/pathology , Inflammation Mediators/metabolism , Insulin Resistance/immunology , Obesity/immunology , Obesity/pathology , Receptors, Antigen, T-Cell/biosynthesis , T-Lymphocyte Subsets/immunology , Adipose Tissue/metabolism , Animals , CD4-CD8 Ratio , Cells, Cultured , Diet/adverse effects , Female , Glucose Tolerance Test/methods , Homeostasis/immunology , Humans , Immunologic Memory , Inflammation Mediators/physiology , Lymphocyte Depletion , Male , Mice , Mice, Inbred C57BL , Obesity/metabolism , Receptors, Antigen, T-Cell/antagonists & inhibitors , Subcutaneous Fat, Abdominal/immunology , Subcutaneous Fat, Abdominal/metabolism , Subcutaneous Fat, Abdominal/pathology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology , Up-Regulation/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...