Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 9(11): e113369, 2014.
Article in English | MEDLINE | ID: mdl-25412423

ABSTRACT

Macrophage infiltration is a critical determinant of high-fat diet induced adipose tissue inflammation and insulin resistance. The precise mechanisms underpinning the initiation of macrophage recruitment and activation are unclear. Macrophage migration inhibitory factor (MIF), a pro-inflammatory cytokine, displays chemokine-like properties. Circulating MIF levels are elevated during obesity however its role in high-fat diet induced adipose inflammation and insulin resistance remains elusive. Wildtype and MIF-/- C57Bl\6J mice were fed chow or high-fat diet. Body weight and food intake was assessed. Glucose homeostasis was monitored by glucose and insulin tolerance tests. Adipose tissue macrophage recruitment and adipose tissue insulin sensitivity was evaluated. Cytokine secretion from stromal vascular fraction, adipose explants and bone marrow macrophages was measured. Inflammatory signature and insulin sensitivity of 3T3-L1-adipocytes co-cultured with wildtype and MIF-/- macrophage was quantified. Hepatic triacylglyceride levels were assessed. MIF-/- exhibited reduced weight gain. Age and weight-matched obese MIF-/- mice exhibited improved glucose homeostasis coincident with reduced adipose tissue M1 macrophage infiltration. Obese MIF-/- stromal vascular fraction secreted less TNFα and greater IL-10 compared to wildtype. Activation of JNK was impaired in obese MIF-/-adipose, concomitant with pAKT expression. 3T3-L1-adipocytes cultured with MIF-/- macrophages had reduced pro-inflammatory cytokine secretion and improved insulin sensitivity, effects which were also attained with MIF inhibitor ISO-1. MIF-/- liver exhibited reduced hepatic triacyglyceride accumulation, enhanced pAKT expression and reduced NFκB activation. MIF deficiency partially protects from high-fat diet induced insulin resistance by attenuating macrophage infiltration, ameliorating adipose inflammation, which improved adipocyte insulin resistance ex vivo. MIF represents a potential therapeutic target for treatment of high-fat diet induced insulin resistance.


Subject(s)
Diet, High-Fat/adverse effects , Fatty Liver/immunology , Inflammation/immunology , Intramolecular Oxidoreductases/deficiency , Macrophage Migration-Inhibitory Factors/deficiency , Macrophages/immunology , Obesity/immunology , 3T3 Cells , Adipose Tissue/immunology , Adipose Tissue/metabolism , Animals , Body Weight , Eating , Fatty Liver/prevention & control , Insulin Resistance , Macrophages/cytology , Mice , Obesity/chemically induced , Obesity/metabolism
2.
Am J Physiol Endocrinol Metab ; 305(7): E834-44, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23921145

ABSTRACT

Emerging evidence has demonstrated that saturated fatty acids prime pro-IL-1ß production and inflammasome-mediated IL-1ß activation is critical in obesity-associated insulin resistance (IR). Nonetheless, IL-1 receptor I-deficient (IL-1RI(-/-)) mice develop mature-onset obesity despite consuming a low-fat diet (LFD). With this apparent contradiction, the present study evaluated whether IL-1RI(-/-) mice were protected against long-term (6 mo) high-fat diet (HFD)-induced IR. Male wild-type and IL-1RI(-/-) mice were fed LFD or HFD for 3 or 6 mo, and glucose and insulin tolerance tests were performed. Adipose insulin sensitivity, cytokine profiles, and adipocyte morphology were assessed. The adipogenic potential of stromal vascular fraction was determined. Hepatic lipid accumulation and insulin sensitivity were characterized. IL-1RI(-/-) mice developed glucose intolerance and IR after 6 mo HFD compared with 3 mo HFD, coincident with enhanced weight gain, hyperinsulinemia, and hyperleptinemia. The aggravated IR phenotype was associated with loss of adipose functionality, switch from adipocyte hyperplasia to hypertrophy and hepatosteatosis. Induction of adipogenic genes was reduced in IL-1RI(-/-) preadipocytes after 6 mo HFD compared with 3 mo HFD. Obese LFD-IL-1RI(-/-) mice exhibited preserved metabolic health. IL-1RI(-/-) mice develop glucose intolerance and IR after 6 mo HFD intervention. While mature-onset obesity is evident in LFD-IL-1RI(-/-) mice, the additional metabolic insult of HFD was required to drive adipose inflammation and systemic IR. These findings indicate an important interaction between dietary fat and IL-1, relevant to optimal metabolic health.


Subject(s)
Diet, High-Fat/adverse effects , Glucose Intolerance/etiology , Insulin Resistance/physiology , Receptors, Interleukin-1 Type I/metabolism , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Adiposity/drug effects , Animals , Dietary Fats/metabolism , Fatty Liver/etiology , Fatty Liver/genetics , Fatty Liver/metabolism , Glucose Intolerance/genetics , Glucose Intolerance/metabolism , Glucose Tolerance Test , Inflammation/genetics , Inflammation/metabolism , Insulin/blood , Insulin/pharmacology , Leptin/blood , Male , Mice , Mice, Knockout , Receptors, Interleukin-1 Type I/genetics
3.
Mol Nutr Food Res ; 56(8): 1212-22, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22700321

ABSTRACT

SCOPE: Inflammasome-mediated inflammation is a critical regulator of obesity-induced insulin resistance (IR). We hypothesized that saturated fatty acids (SFA) directly prime the NLRP3 inflammasome via TLR4 concurrent with IR. We focused on dendritic cells (DCs) (CD11c(+) CD11b(+) F4/80(-) ), which are recruited into obese adipose tissue following high-fat diet (HFD) challenge and are a key cell in inflammasome biology. METHODS AND RESULTS: C57BL/6 mice were fed HFD for 16 weeks (45% kcal palm oil), glucose homeostasis was monitored by glucose and insulin tolerance tests. Stromal vascular fraction (SVF) cells were isolated from adipose and analyzed for CD11c(+) CD11b(+) F480(-) DC. Following coculture with bone marrow derived DC (BMDC) insulin-stimulated (3) H-glucose transport into adipocytes, IL-1ß secretion and caspase-1 activation was monitored. BMDCs primed with LPS (100 ng/mL), linoleic acid (LA; 200 µM), or palmitic acid (PA; 200 µM) were used to monitor inflammasome activation. We demonstrated significant infiltration of DCs into adipose after HFD. HFD-derived DCs reduce adipocyte insulin sensitivity upon coculture co-incident with enhanced adipocyte caspase-1 activation/IL-1ß secretion. HFD-derived DCs are skewed toward a pro-inflammatory phenotype with increased IL-1ß secretion, IL-1R1, TLR4, and caspase-1 expression. Complementary in vitro experiments demonstrate that TLR4 is critical in propagating SFA-mediated inflammasome activation. CONCLUSION: SFA represent metabolic triggers priming the inflammasome, promoting adipocyte inflammation/IR, suggesting direct effects of SFA on inflammasome activation via TLR4.


Subject(s)
Carrier Proteins/metabolism , Dendritic Cells/drug effects , Fatty Acids/pharmacology , Inflammasomes/drug effects , Insulin Resistance/physiology , Toll-Like Receptor 4/metabolism , 3T3-L1 Cells , Adipose Tissue/cytology , Adipose Tissue/metabolism , Animals , Caspase 1/metabolism , Coculture Techniques , Cytokines/metabolism , Dendritic Cells/metabolism , Diet, High-Fat/adverse effects , Dietary Fats/pharmacology , Inflammasomes/metabolism , Interleukin-1beta/metabolism , Linoleic Acid/pharmacology , Male , Mice , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein
4.
FASEB J ; 26(10): 4287-94, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22700871

ABSTRACT

Aging and adiposity are associated with chronic low-grade inflammation, which underlies the development of obesity-associated complications, including type 2 diabetes mellitus (T2DM). The mechanisms underlying adipose inflammation may include macrophage infiltration and activation, which, in turn, affect insulin sensitivity of adipocytes. There is a growing appreciation that specific lipid mediators (including lipoxins, resolvins, and protectins) can promote the resolution of inflammation. Here, we investigated the effect of lipoxin A4 (LXA4), the predominant endogenously generated lipoxin, on adipose tissue inflammation. Using adipose tissue explants from perigonadal depots of aging female C57BL/6J mice (Animalia, Chordata, Mus musculus) as a model of age-associated adipose inflammation, we report that LXA4 (1 nM) attenuates adipose inflammation, decreasing IL-6 and increasing IL-10 expression (P<0.05). The altered cytokine milieu correlated with increased GLUT-4 and IRS-1 expression, suggesting improved insulin sensitivity. Further investigations revealed the ability of LXA4 to rescue macrophage-induced desensitization to insulin-stimulated signaling and glucose uptake in cultured adipocytes, using vehicle-stimulated cells as controls. This was associated with preservation of Akt activation and reduced secretion of proinflammatory cytokines, including TNF-α. We therefore propose that LXA4 may represent a potentially useful and novel therapeutic strategy to subvert adipose inflammation and insulin resistance, key components of T2DM.


Subject(s)
Adipose Tissue/drug effects , Adipose Tissue/immunology , Inflammation/drug therapy , Lipoxins/therapeutic use , Adipose Tissue/metabolism , Animals , Female , Glucose Transporter Type 4/metabolism , Inflammation/metabolism , Insulin Receptor Substrate Proteins/metabolism , Insulin Resistance/physiology , Interleukin-10/metabolism , Interleukin-6/metabolism , Mice , Mice, Inbred C57BL , Obesity/drug therapy , Obesity/metabolism , Tumor Necrosis Factor-alpha/metabolism
5.
J Nutr Biochem ; 23(9): 1192-200, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22137266

ABSTRACT

OBJECTIVE: Adipose tissue inflammation with immune cell recruitment plays a key role in obesity-induced insulin resistance (IR). Long-chain (LC) n-3 polyunsaturated fatty acids (PUFA) eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) have anti-inflammatory potential; however, their individual effects on adipose IR are ill defined. We hypothesized that EPA and DHA may differentially affect macrophage-induced IR in adipocytes. METHODS: J774.2 macrophages pretreated with EPA or DHA (50 µM for 5 days) were stimulated with lipopolysaccharide (LPS, 100 ng/ml for 30 min-48 h). Cytokine secretion profiles and activation status of macrophages were assessed by enzyme-linked immunosorbent assay and flow cytometry. Pretreated macrophages were seeded onto transwell inserts and placed over 3T3-L1 adipocytes for 24-72 h; effects on adipocyte-macrophage cytokine cross-talk and insulin-stimulated ³H-glucose transport into adipocytes were monitored. RESULTS: DHA had more potent anti-inflammatory effects relative to EPA, with marked attenuation of LPS-induced nuclear factor (NF)κB activation and tumor necrosis factor (TNF)α secretion in macrophages. DHA specifically enhanced anti-inflammatory interleukin (IL)-10 secretion and reduced the expression of proinflammatory M1 (F4/80⁺/CD11⁺) macrophages. Co-culture of DHA-enriched macrophages with adipocytes attenuated IL-6 and TNFα secretion while enhancing IL-10 secretion. Conditioned media (CM) from DHA-enriched macrophages attenuated adipocyte NFκB activation. Adipocytes co-cultured with DHA-enriched macrophages maintained insulin sensitivity with enhanced insulin-stimulated ³H-glucose transport, GLUT4 translocation and preservation of insulin-receptor substrate-1 expression compared to co-culture with untreated macrophages. We confirmed that IL-10 expressed by DHA-enriched macrophages attenuates the CM-induced proinflammatory IR phenotype in adipocytes. CONCLUSIONS: We demonstrate an attenuated proinflammatory phenotype of DHA-pretreated macrophages, which when co-cultured with adipocytes partially preserved insulin sensitivity.


Subject(s)
Adipocytes, White/metabolism , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Docosahexaenoic Acids/metabolism , Insulin Resistance , Interleukin-10/metabolism , Macrophage Activation , Macrophages/immunology , 3T3-L1 Cells , Adipocytes, White/immunology , Animals , Biological Transport , Cell Communication , Cell Line, Transformed , Coculture Techniques , Culture Media, Conditioned , Cytokines/metabolism , Eicosapentaenoic Acid/metabolism , Glucose/metabolism , Glucose Transporter Type 4/metabolism , Insulin/metabolism , Insulin Receptor Substrate Proteins/metabolism , Macrophages/metabolism , Mice , Protein Transport
6.
Proc Nutr Soc ; 70(4): 408-17, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21835098

ABSTRACT

High-fat diet-induced obesity is associated with a chronic state of low-grade inflammation, which pre-disposes to insulin resistance (IR), which can subsequently lead to type 2 diabetes mellitus. Macrophages represent a heterogeneous population of cells that are instrumental in initiating the innate immune response. Recent studies have shown that macrophages are key mediators of obesity-induced IR, with a progressive infiltration of macrophages into obese adipose tissue. These adipose tissue macrophages are referred to as classically activated (M1) macrophages. They release cytokines such as IL-1ß, IL-6 and TNFα creating a pro-inflammatory environment that blocks adipocyte insulin action, contributing to the development of IR and type 2 diabetes mellitus. In lean individuals macrophages are in an alternatively activated (M2) state. M2 macrophages are involved in wound healing and immunoregulation. Wound-healing macrophages play a major role in tissue repair and homoeostasis, while immunoregulatory macrophages produce IL-10, an anti-inflammatory cytokine, which may protect against inflammation. The functional role of T-cell accumulation has recently been characterised in adipose tissue. Cytotoxic T-cells are effector T-cells and have been implicated in macrophage differentiation, activation and migration. Infiltration of cytotoxic T-cells into obese adipose tissue is thought to precede macrophage accumulation. T-cell-derived cytokines such as interferon γ promote the recruitment and activation of M1 macrophages augmenting adipose tissue inflammation and IR. Manipulating adipose tissue macrophages/T-cell activity and accumulation in vivo through dietary fat modification may attenuate adipose tissue inflammation, representing a therapeutic target for ameliorating obesity-induced IR.


Subject(s)
Adipose Tissue/immunology , Dietary Fats/immunology , Inflammation/immunology , Insulin Resistance/immunology , Macrophages/metabolism , Obesity/immunology , T-Lymphocytes/metabolism , Cytokines/metabolism , Diet, High-Fat/adverse effects , Dietary Fats/adverse effects , Humans , Inflammation/etiology , Obesity/complications
7.
Diabetes ; 60(6): 1688-98, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21515850

ABSTRACT

OBJECTIVE: High-fat diet (HFD)-induced adipose tissue inflammation is a critical feature of diet-induced insulin resistance (IR); however, the contribution of interleukin-1 receptor I (IL-1RI)-mediated signals to this phenotype has not been defined. We hypothesized that lack of IL-1RI may ameliorate HFD-induced IR by attenuating adipose tissue inflammation. RESEARCH DESIGN AND METHODS: Glucose homeostasis was monitored in chow- and HFD-fed wild-type (WT) and IL-1RI(-/-) mice by glucose tolerance and insulin tolerance tests. Macrophage recruitment and cytokine signature of adipose tissue macrophages was evaluated. Insulin sensitivity and cytokine secretion from adipose explants was quantified. Cytokine secretion and adipocyte insulin sensitivity was measured in cocultures of WT or IL-1RI(-/-) macrophages with 3T3L1 adipocytes. Synergistic effects of IL-1ß with tumor necrosis factor (TNF)-α on inflammation was monitored in WT and IL-1RI(-/-) bone-marrow macrophages and adipose explants. RESULTS: Lean and obese IL-1RI(-/-) animals exhibited enhanced glucose homeostasis by glucose tolerance test and insulin tolerance test. M1/M2 macrophage number in adipose tissue was comparable between genotypes; however, TNF-α and IL-6 secretion was lower from IL-1RI(-/-) adipose tissue macrophages. IL-1RI(-/-) adipose exhibited enhanced insulin sensitivity, elevated pAKT, lower cytokine secretion, and attenuated induction of phosphorylated signal transducer and activator of transcription 3 and suppressor of cytokine signaling molecule 3 after HFD. Coculture of WT, but not IL-1RI(-/-) macrophages, with 3T3L1 adipocytes enhanced IL-6 and TNF-α secretion, reduced adiponectin secretion, and impaired adipocyte insulin sensitivity. TNF-α and IL-1ß potently synergized to enhance inflammation in WT macrophages and adipose, an effect lost in the absence of IL-1RI. CONCLUSIONS: Lack of IL-1RI protects against HFD-induced IR coincident with reduced local adipose tissue inflammation, despite equivalent immune cell recruitment.


Subject(s)
Adipose Tissue/drug effects , Adipose Tissue/immunology , Dietary Fats/adverse effects , Glucose/metabolism , Inflammation/immunology , Inflammation/prevention & control , Insulin Resistance/physiology , Receptors, Interleukin-1 Type I/deficiency , 3T3-L1 Cells , Animals , Female , Genotype , Inflammation/genetics , Insulin/pharmacology , Interleukin-1beta/metabolism , Interleukin-1beta/pharmacology , Interleukin-6/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NIH 3T3 Cells , Obesity/immunology , Obesity/metabolism , Receptors, Interleukin-1 Type I/genetics , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...