Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Nutrients ; 16(13)2024 Jun 29.
Article in English | MEDLINE | ID: mdl-38999834

ABSTRACT

In obesity, circulating saturated fatty acids (SFAs) and inflammatory cytokines interfere with skeletal muscle insulin signaling, leading to whole body insulin resistance. Further, obese skeletal muscle is characterized by macrophage infiltration and polarization to the inflammatory M1 phenotype, which is central to the development of local inflammation and insulin resistance. While skeletal muscle-infiltrated macrophage-myocyte crosstalk is exacerbated by SFA, the effects of other fatty acids, such as n-3 and n-6 polyunsaturated fatty acids (PUFAs), are less studied. Thus, the objective of this study was to determine the effects of long-chain n-3 and n-6 PUFAs on macrophage M1 polarization and subsequent effects on myocyte inflammation and metabolic function compared to SFA. Using an in vitro model recapitulating obese skeletal muscle cells, differentiated L6 myocytes were cultured for 24 h with RAW 264.7 macrophage-conditioned media (MCM), followed by insulin stimulation (100 nM, 20 min). MCM was generated by pre-treating macrophages for 24 h with 100 µM palmitic acid (16:0, PA-control), arachidonic acid (20:4n-6, AA), or docosahexaenoic acid (22:6n-3, DHA). Next, macrophage cultures were stimulated with a physiological dose (10 ng/mL) of lipopolysaccharide for an additional 12 h to mimic in vivo obese endotoxin levels. Compared to PA, both AA and DHA reduced mRNA expression and/or secreted protein levels of markers for M1 (TNFα, IL-6, iNOS; p < 0.05) and increased those for M2 (IL-10, TGF-ß; p < 0.05) macrophage polarization. In turn, AA- and DHA-derived MCM reduced L6 myocyte-secreted cytokines (TNFα, IL-6; p < 0.05) and chemokines (MCP-1, MIP-1ß; p < 0.05). Only AA-derived MCM increased L6-myocyte phosphorylation of Akt (p < 0.05), yet this was inconsistent with improved insulin signaling, as only DHA-derived MCM improved L6 myocyte glucose uptake (p < 0.05). In conclusion, dietary n-3 and n-6 PUFAs may be a useful strategy to modulate macrophage-myocyte inflammatory crosstalk and improve myocyte insulin sensitivity in obesity.


Subject(s)
Fatty Acids, Omega-3 , Inflammation , Insulin Resistance , Macrophages , Animals , Macrophages/metabolism , Macrophages/drug effects , Mice , Fatty Acids, Omega-3/pharmacology , Inflammation/metabolism , RAW 264.7 Cells , Fatty Acids, Omega-6/pharmacology , Insulin/metabolism , Cytokines/metabolism , Obesity/metabolism , Signal Transduction/drug effects , Rats , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/drug effects
2.
Nutrition ; 91-92: 111388, 2021.
Article in English | MEDLINE | ID: mdl-34298481

ABSTRACT

OBJECTIVES: Chronic low-grade inflammation in obesity is partly driven by inflammatory cross talk between adipocytes and interferon-γ-secreting CD4+ T-helper (Th)1 cells, a process we have shown may be mitigated by long-chain (LC) ω-3 polyunsaturated fatty acids (PUFAs). Our objective was to study pivotal mediators of interactions between Th1 cells and adipocytes as potential mechanisms underlying the antiinflammatory effects of LC ω-3 PUFAs. METHODS: Using an in vitro model, 3T3-L1 adipocytes were cocultured with purified splenic CD4+ T cells from C57BL/6 mice consuming one of two isocaloric high-fat (HF) diets (60% kcal fat), containing either 41.2% kcal from lard + 18.7% kcal from corn oil (control, HF) or 41.2% kcal from lard + 13.4% kcal from corn oil + 5.3% kcal from fish oil (HF+FO). Cocultures were stimulated for 48 h with lipopolysaccharide (10 ng/mL). RESULTS: Compared with HF cocultures, HF+FO reduced Th1-cell markers (including secreted interferon-γ) and increased Th2-cell markers, consistent with reduced expression of genes related to major histocompatibility complex II (P < 0.05). HF+FO also blunted markers of priming and activity of the NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome (P < 0.05). In confirmatory work, 3T3-L1 adipocyte pretreatment with the LC ω-3 PUFA docosahexaenoic acid (100 µM, 24 h) blunted interferon-γ-induced (5 ng/mL, 24 h) expression of genes related to major histocompatibility complex II and priming and activity markers of the NLRP3 inflammasome compared with control (P < 0.05). CONCLUSIONS: Inflammatory interactions between CD4+ T cells and adipocytes may provide a target for LC ω-3 PUFAs to mitigate obesity-associated inflammation.


Subject(s)
Fatty Acids, Omega-3 , Inflammasomes , Adipocytes , Adipose Tissue , Animals , Coculture Techniques , Diet, High-Fat , Fatty Acids, Omega-3/pharmacology , Mice , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein , Obesity/drug therapy , Th1 Cells
3.
Food Funct ; 12(14): 6348-6362, 2021 Jul 21.
Article in English | MEDLINE | ID: mdl-34105575

ABSTRACT

High fat meal-induced postprandial inflammation is exacerbated in overweight and obesity and may contribute to cardiovascular disease (CVD) risk. This study aimed to determine the effects of apples, rich in anti-inflammatory polyphenols, on biomarkers of postprandial inflammation in individuals with overweight and obesity. A randomized, crossover trial was conducted with n = 26 participants (17 female/9 male; mean age 45.5 ± 3.12 years; mean BMI 34.1 ± 1.18 kg m-2) to assess the effects of 3 whole Gala apples (∼200 g) on the 2, 4 and 6 h postprandial response to a high fat meal providing 1 g fat per kg body weight. Changes in plasma biomarkers of inflammation (as the primary outcome) and endotoxin exposure, non-esterified fatty acids (NEFA) and total antioxidant capacity (TAC) were measured. Fasting (0 h) and 4 h peripheral blood mononuclear cells (PBMC) were also isolated from whole blood and stimulated with or without a physiological dose (10 ng mL-1) of lipopolysaccharide (LPS) to measure secreted cytokines. Apples modulated postprandial plasma IFN-γ and reduced its peak concentration (-12.8%), and increased both 4 h (14.4%) and peak (10.5%) TAC (P < 0.05). In unstimulated and LPS-stimulated PBMC, apples reduced secreted IL-6 (-49.3% and -17.1%) and TNF-α (-43.3% and -14.7%) and increased IL-4 (93.1% and 15.8%) in both the unstimulated and LPS-stimulated conditions, as well as decreased GM-CSF (-26.0%) and IL-17 (-47.9%) in unstimulated PBMC and G-CSF (-19.8%) in LPS-stimulated PBMC (P < 0.05). These data suggest acute whole Gala apple consumption may be an effective dietary strategy to mitigate high fat meal-induced postprandial inflammation that exacerbates CVD risk in overweight and obesity. This study was registered at clinicaltrials.gov, NCT03523403, The Apple Study: Investigating the Effects of Whole Apple Consumption on Risk Factors for Chronic Metabolic Diseases in Overweight and Obese Adults.


Subject(s)
Diet, High-Fat/adverse effects , Inflammation/diet therapy , Malus , Obesity/diet therapy , Overweight/diet therapy , Adult , Biomarkers/blood , Cross-Over Studies , Cytokines/blood , Female , Fruit , Humans , Inflammation/blood , Interleukin-6/blood , Leukocytes, Mononuclear/metabolism , Male , Metabolic Diseases/epidemiology , Middle Aged , Postprandial Period , Risk Factors , Treatment Outcome
4.
J Nutr Biochem ; 95: 108763, 2021 09.
Article in English | MEDLINE | ID: mdl-33965532

ABSTRACT

Obesity is associated with inflammation and has been shown to increase breast cancer severity. The objective of this study was to examine the effect of fish oil (FO) supplementation in obesity-associated mammary tumorigenesis in the MMTV-neu(ndl)-YD5 mouse model of human epidermal growth factor receptor-2 positive BC. Female mice were fed one of three diets for 16 weeks: i) high fat diet [HF, % kacl: 41.2% lard, 18.7% corn oil (CO)], ii) an isocaloric HF plus menhaden FO diet (HF+FO, % kcal: 41.2 lard, 13.4% CO, 5.3% FO), iii) low fat diet (LF, % kcal: 4.7% lard, 6% CO). HF mice had increased body weight, visceral adipose weight and serum hormone concentrations (increased leptin and resistin; decreased adiponectin) versus LF, which was attenuated in the HF+FO group versus HF (P<.05). Compared to HF, tumor onset was delayed in HF+FO and LF mice (P<0.05). Compared to HF, HF+FO reduced mammary tumor multiplicity (-27%), tumor weight (-46%) and total tumor volume (-50%) (P<0.05). Additionally, HF+FO reduced mammary tumor multiplicity (-33%), tumor weight (-39%) and total tumor volume (-60%) versus LF. HF+FO improved mammary tumor apoptosis status with increased expression of pro-apoptotic Bad and decreased expression of anti-apoptotic Bcl-xLmediators versus HF (P<0.05). Additionally, HF+FO decreased tumor protein expression of activated Akt, NFκB p65 and STAT3, versus HF (P<0.05). Tumor mRNA expression of inflammatory mediators TNFα, IL-6 and leptin were reduced in HF+FO, whereas IL-10 expression was increased compared to HF (P<0.05). Collectively these results demonstrate the efficacy of FO supplementation for improving obesity-associated breast cancer outcomes.


Subject(s)
Apoptosis/drug effects , Fish Oils/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Inflammation/drug therapy , Mammary Neoplasms, Experimental/drug therapy , Obesity/chemically induced , Adipose Tissue/drug effects , Animals , Body Weight/drug effects , Breast Neoplasms , Cell Line, Tumor , Dietary Supplements , Fatty Acids/chemistry , Female , Fish Oils/administration & dosage , Humans , Mammary Glands, Animal/chemistry , Mice , RNA, Messenger/genetics , RNA, Messenger/metabolism , Random Allocation , Receptor, ErbB-2
5.
Am J Clin Nutr ; 114(2): 752-763, 2021 08 02.
Article in English | MEDLINE | ID: mdl-33964852

ABSTRACT

BACKGROUND: Obesity-associated low-grade inflammation contributes to the development of cardiovascular disease (CVD). Apples are rich in anti-inflammatory bioactives including polyphenols and fiber. OBJECTIVES: We aimed to determine the effects of regular apple consumption on fasting plasma biomarkers of inflammation (primary outcome), endotoxemia, carbohydrate and lipid metabolism (glucose, insulin, triacylglycerol; secondary outcomes), and peripheral blood mononuclear cell (PBMC)-secreted cytokines (secondary outcome) in individuals with overweight and obesity. METHODS: A randomized, controlled, parallel-arm trial was conducted with n = 46 participants. After avoiding foods and beverages rich in polyphenols and fiber for 2 wk, participants consumed 3 whole Gala apples (∼200 g edible parts)/d as part of their habitual diet (n = 23) or avoided apples (control, n = 23) for 6 wk. All participants limited consumption of polyphenols and fiber during the 6-wk trial. Fasting blood samples were collected before and after 6 wk for analysis of plasma biomarkers and isolation of PBMCs, which were cultured for 24 h unstimulated or stimulated with LPS (10 ng/mL). RESULTS: Forty-four participants completed the trial (30 female, 14 male; mean ± SEM age: 45.4 ± 2.2 y; BMI: 33.4 ± 0.9 kg/m2). After ANCOVA and correcting for multiple comparisons, apples decreased fasting plasma C-reactive protein by 17.0% (range: 14.3%-19.6%, P = 0.005), IL-6 by 12.4% (range: 6.7%-17.5%, P < 0.001), and LPS-binding protein by 20.7% (range: 14.1%-26.4%, P < 0.001) compared with control. Apples also decreased PBMC-secreted IL-6 by 28.3% (range: 22.4%-33.5%, P < 0.001) and IL-17 by 11.0% (range 5.8-15.6%, P = 0.003) in the unstimulated condition compared with control. Exploratory analysis showed apples also increased plasma total antioxidant capacity by 9.6% (range: 1.7-18.9%, P = 0.002) compared with control. However, apples had no effect on anthropometric or other CVD risk markers. CONCLUSIONS: Six-week daily whole Gala apple consumption may be an effective dietary strategy to mitigate the obesity-associated inflammation that exacerbates CVD risk, without weight loss. This trial was registered at clinicaltrials.gov as NCT03523403.


Subject(s)
Inflammation/metabolism , Leukocytes, Mononuclear/metabolism , Malus , Overweight , Adult , Biomarkers/metabolism , Humans
6.
Methods Mol Biol ; 2184: 111-130, 2020.
Article in English | MEDLINE | ID: mdl-32808222

ABSTRACT

The co-culture of adipocytes and immune cells, such as macrophages or T cells (CD4+ or CD8+ subsets), is a novel experimental approach used to study paracrine interactions (or the cross talk) between cultured cell types in isolation, in order to understand their role in obese adipose tissue (AT) inflammation and dysfunction. Here we describe the general methodologies required for the co-culture of mature adipocytes (differentiated 3T3-L1 pre-adipocyte cell line) with primary immune cell subsets purified from mouse splenic mononuclear cells using a magnetic MicroBead positive selection, wherein multiple immune cell populations can be purified sequentially from a single mouse spleen, thereby providing diversity in the types of immune cells that can be co-cultured with adipocytes. Additionally, we describe experimental procedures for co-culturing adipocytes and immune cells in two different co-culture systems, including a cell contact-dependent co-culture system, wherein the cells are in direct physical contact, and a cell contact-independent, soluble mediator-driven co-culture system wherein the cells are physically separated by a trans-well semipermeable membrane. Finally, we discuss how these co-culture models can be utilized to recapitulate the AT microenvironment in obesity by utilizing physiologically relevant ratios of adipocytes:immune cells (specifically CDllb+ macrophages, CD4+ T cells, or CD8+ T cells) and lipopolysaccharide stimulation that mimics endotoxin concentrations observed in obesity.


Subject(s)
CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/cytology , Coculture Techniques/methods , Macrophages/cytology , 3T3-L1 Cells , Adipocytes , Adipose Tissue/cytology , Animals , Cell Differentiation/physiology , Cells, Cultured , Cellular Microenvironment/physiology , Inflammation/pathology , Mice , Obesity/pathology , Spleen/cytology
7.
J Nutr Biochem ; 86: 108488, 2020 12.
Article in English | MEDLINE | ID: mdl-32827664

ABSTRACT

Obese adipose tissue (AT) inflammation is partly driven by accumulation of CD4+ T helper (Th)1 cells and reduced Th2 and T regulatory subsets, which promotes macrophage chemotaxis and ensuing AT metabolic dysfunction. This study investigated CD4+ T cell/adipocyte cytokine-mediated paracrine interactions (cross talk) as a target for dietary intervention to mitigate obese AT inflammation. Using an in vitro co-culture model designed to recapitulate CD4+ T cell accumulation in obese AT (5% of stromal vascular cellular fraction), 3T3-L1 adipocytes were co-cultured with purified splenic CD4+ T cells from C57Bl/6 mice consuming one of two isocaloric diets containing either 10% w/w safflower oil (control, CON) or 7% w/w safflower oil+3% w/w fish oil (FO) for 4 weeks (n=8-11/diet). The FO diet provided 1.9% kcal from the long-chain (LC) n-3 polyunsaturated fatty acids (PUFAs) eicosapentaenoic acid and docosahexaenoic acid, a dose that can be achieved by supplementation. Co-cultures were stimulated for 48 h with lipopolysaccharide (LPS) to mimic in vivo obese endotoxin levels or with conditioned media collected from LPS-stimulated visceral AT isolated from CON-fed mice. In both stimulation conditions, FO reduced mRNA expression and/or secreted protein levels of Th1 markers (T-bet, IFN-γ) and increased Th2 markers (GATA3, IL-4), concomitant with reduced inflammatory cytokines (IL-1ß, IL-6, IL-12p70, TNF-α), macrophage chemokines (MCP-1, MCP-3, MIP-1α, MIP-2) and levels of activated central regulators of inflammatory signaling (NF-κB, STAT-1, STAT-3) (P<.05). Therefore, CD4+ T cell/adipocyte cross talk represents a potential target for LC n-3 PUFAs to mitigate obese AT inflammation.


Subject(s)
Adipocytes/immunology , CD4-Positive T-Lymphocytes/cytology , Fatty Acids, Omega-3/metabolism , Inflammation/drug therapy , Obesity/immunology , 3T3-L1 Cells , Adipose Tissue/immunology , Animals , Chemokines/metabolism , Coculture Techniques , Diet , Disease Models, Animal , Female , Fish Oils/metabolism , Inflammation/blood , Inflammation/immunology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Obese , NF-kappa B p50 Subunit/metabolism , Obesity/blood , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction
8.
Nutrients ; 12(5)2020 May 12.
Article in English | MEDLINE | ID: mdl-32408695

ABSTRACT

Adipose tissue (AT) expansion induces local hypoxia, a key contributor to the chronic low-grade inflammation that drives obesity-associated disease. Apple flavonols phloretin (PT) and phlorizin (PZ) are suggested anti-inflammatory molecules but their effectiveness in obese AT is inadequately understood. Using in vitro models designed to reproduce the obese AT microenvironment, 3T3-L1 adipocytes were cultured for 24 h with PT or PZ (100 µM) concurrent with the inflammatory stimulus lipopolysaccharide (LPS; 10 ng/mL) and/or the hypoxia mimetic cobalt chloride (CoCl2; 100 µM). Within each condition, PT was more potent than PZ and its effects were partially mediated by peroxisome proliferator-activated receptor (PPAR)-γ (p < 0.05), as tested using the PPAR-γ antagonist bisphenol A diglycidyl ether (BADGE). In LPS-, CoCl2-, or LPS + CoCl2-stimulated adipocytes, PT reduced mRNA expression and/or secreted protein levels of inflammatory and macrophage chemotactic adipokines, and increased that of anti-inflammatory and angiogenic adipokines, which was consistent with reduced mRNA expression of M1 polarization markers and increased M2 markers in RAW 264.7 macrophages cultured in media collected from LPS + CoCl2-simulated adipocytes (p < 0.05). Further, within LPS + CoCl2-stimulated adipocytes, PT reduced reactive oxygen species accumulation, nuclear factor-κB activation, and apoptotic protein expression (p < 0.05). Overall, apple flavonols attenuate critical aspects of the obese AT phenotype.


Subject(s)
Adipocytes/drug effects , Angiogenesis Inducing Agents/metabolism , Anti-Inflammatory Agents/pharmacology , Flavonols/pharmacology , Malus/chemistry , 3T3-L1 Cells , Adipose Tissue/drug effects , Animals , Cobalt , Inflammation , Lipopolysaccharides , Mice , Obesity , PPAR gamma , Signal Transduction/drug effects
9.
Br J Nutr ; 123(7): 807-817, 2020 04 14.
Article in English | MEDLINE | ID: mdl-31902373

ABSTRACT

Whole apples are a source of pectin and polyphenols, both of which show potential to modulate postprandial lipaemia (PPL). The present study aimed to explore the effects of whole apple consumption on PPL, as a risk factor for CVD, in generally healthy but overweight and obese adults. A randomised, crossover acute meal trial was conducted with seventeen women and nine men (mean BMI of 34·1 (sem 0·2) kg/m2). Blood samples were collected for 6 h after participants consumed an oral fat tolerance test meal that provided 1 g fat/kg body weight and 1500 mg acetaminophen per meal for estimating gastric emptying, with and without three whole raw Gala apples (approximately 200 g). Plasma TAG (with peak postprandial concentration as the primary outcome), apoB48, chylomicron-rich fraction particle size and fatty acid composition, glucose, insulin and acetaminophen were analysed. Differences between with and without apples were identified by ANCOVA. Apple consumption did not alter postprandial TAG response, chylomicron properties, glucose or acetaminophen (P > 0·05), but did lead to a higher apoB48 peak concentration and exaggerated insulin between 20 and 180 min (P < 0·05). Overall, as a complex food matrix, apples did not modulate postprandial TAG when consumed with a high-fat meal in overweight and obese adults, but did stimulate insulin secretion, potentially contributing to an increased TAG-rich lipoprotein production.


Subject(s)
Apolipoprotein B-48/blood , Fatty Acids/blood , Fruit , Malus , Triglycerides/blood , Adult , Aged , Blood Glucose , Cross-Over Studies , Diet , Female , Humans , Insulin/blood , Male , Meals , Middle Aged , Postprandial Period , Young Adult
10.
J Nutr Biochem ; 76: 108243, 2020 02.
Article in English | MEDLINE | ID: mdl-31760229

ABSTRACT

Obese visceral adipose tissue (AT) inflammation is driven by adipokine-mediated cross talk between CD8+ T cells and adipocytes, a process mitigated by long-chain (LC) n-3 polyunsaturated fatty acids (PUFA) but underlying mechanisms and ensuing effects on macrophage polarization status are unknown. Using an in vitro co-culture model that recapitulates the degree of CD8+ T cell infiltration reported in obese AT, 3T3-L1 adipocytes were co-cultured for 24 h with purified splenic CD8+ T cells from C57Bl/6 mice consuming either a 10% w/w safflower oil (control, CON) or 7% w/w safflower oil + 3% w/w fish oil (FO) diet for 4 weeks (n=8-10/diet). Co-cultured cells were in direct contact or in a contact-independent condition separated by a Transwell permeable membrane and stimulated with lipopolysaccharide (10 ng/ml) to mimic in vivo obese endotoxin levels. In contact-dependent co-cultures, FO reduced inflammatory (IL-6, TNFα, IFN-γ) and macrophage chemotactic (CCL2, CCL7, CCL3) mRNA expression and/or secreted protein, NF-κB p65 activation, ROS accumulation, NLRP3 inflammasome priming (Nlrp3, Il1ß mRNA) and activation (caspase-1 activity) compared to CON (P<.05). The anti-inflammatory action of FO was reproduced by the addition of a TNF-α neutralizing antibody (1 µg/ml) to CON co-cultures (CON/anti-TNF-α), albeit to a lesser degree. Conditioned media from FO and CON/anti-TNF-α co-cultures, in turn, reduced RAW 264.7 macrophage mRNA expression of M1 polarization markers (iNos, Cd11c, Ccr2) and associated inflammatory cytokines (Il6, Tnfα, Il1ß) compared to CON. These data suggest that inflammatory CD8+ T cell/adipocyte cross talk is partially attributable to TNF-α signaling, which can be mitigated by LC n-3 PUFA.


Subject(s)
Adipocytes/metabolism , CD8-Positive T-Lymphocytes/cytology , Fatty Acids, Omega-3/metabolism , Fish Oils/metabolism , Inflammation/metabolism , Macrophages/metabolism , Tumor Necrosis Factor-alpha/metabolism , 3T3-L1 Cells , Animals , Body Weight , Coculture Techniques , Female , Male , Mice , Mice, Inbred C57BL , RAW 264.7 Cells/cytology , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction
11.
J Nutr Biochem ; 72: 108216, 2019 10.
Article in English | MEDLINE | ID: mdl-31476608

ABSTRACT

Impaired intestinal health characterized by a dysbiotic microbial community and a dysfunctional epithelial barrier contributes to host inflammation and metabolic dysfunction in obesity. Fish oil (FO)-derived n-3 polyunsaturated fatty acids have been shown to improve aspects of the obese phenotype; however, their effect on obese intestinal health is unknown. This study aimed to determine the effect of dietary FO on the intestinal microenvironment, including the microbial community and epithelial barrier, in a mouse model of high-fat diet induced obesity and metabolic dysfunction. Male C57BL/6 mice were fed (12 weeks) either a high-fat diet (HF, 60% fat as kcal) or an isocaloric HF supplemented with Menhaden FO (5.3% kcal, HF + FO). 16S rRNA sequencing was used to determine changes in fecal microbiota. Intestinal (ileum and colon) and epididymal adipose tissue RNA was used to assess biomarkers of barrier integrity and inflammatory status, respectively. Serum was used to assess adipokine concentrations and insulin resistance. HF + FO diet altered the fecal microbiota by decreasing the abundance of Firmicutes and increasing the abundance of members of the Bacteroidetes phyla, as well as increasing the abundance of antiobesogenic Akkermansia muciniphila, compared to HF. Intestinal epithelial barrier functions were improved by HF + FO evidenced by increased mRNA expression of tight junction components, antimicrobial defenses and mucus barrier components. HF + FO-fed mice exhibited improvements in homeostatic model assessment of insulin resistance, oral glucose tolerance and serum adipokine concentrations and epididymal mRNA expression (increased adiponectin and decreased leptin) versus HF. HF + FO improved obese intestinal health and attenuated metabolic dysfunction associated with obesity.


Subject(s)
Diet, High-Fat/adverse effects , Fish Oils/pharmacology , Gastrointestinal Microbiome/drug effects , Intestines/drug effects , Obesity/diet therapy , Adipokines/blood , Animals , Body Weight/drug effects , Colon/drug effects , Colon/physiology , Dietary Supplements , Eating/drug effects , Fatty Acids, Omega-3/metabolism , Feces/microbiology , Gastrointestinal Microbiome/genetics , Glucose Tolerance Test , Ileum/drug effects , Ileum/physiology , Intestines/physiology , Intra-Abdominal Fat/drug effects , Intra-Abdominal Fat/pathology , Male , Mice, Inbred C57BL , Obesity/etiology , Panniculitis/etiology , Panniculitis/prevention & control
12.
J Nutr Biochem ; 70: 91-104, 2019 08.
Article in English | MEDLINE | ID: mdl-31195365

ABSTRACT

Obesity is associated with impaired intestinal epithelial barrier function and an altered microbiota community structure, which contribute to host systemic inflammation and metabolic dysfunction. Fiber-rich common beans (Phaseolus vulgaris) promote intestinal health (microbiota and host epithelial barrier integrity) in lean mice. The objective was to assess the intestinal health promoting effects of navy bean supplementation during high-fat (HF)diet-induced obesity. Male C57BL/6 mice were fed either a high-fat (HF) diet (60% of kcal from fat) or an isocaloric HF diet supplemented with 15.7% (by weight) cooked navy bean powder (HF+B) for 12 weeks. Compared to HF, the HF+B diet altered the fecal microbiota community structure (16S rRNA gene sequencing), most notably increasing abundance of Akkermansia muciniphila (+19-fold), whose abundance typically decreases in obese humans and rodents. Additionally, HF+B fecal abundance of carbohydrate fermenting, short chain fatty acid (SCFA) producing Prevotella (+332-fold) and S24-7 (+1.6-fold) and fecal SCFA levels were increased. HF+B improved intestinal health and epithelial barrier integrity versus HF, evidenced by reduced serum fluorescein isothiocyanate (FITC)-dextran concentration in an in vivo gut permeability test, and increased intestinal mRNA expression of tight junction components (ZO-1, occludin), anti-microbial defenses (Reg3γ, IgA, Defα5, Defß2) and mucins (Muc2). Additionally, HF+B improved the systemic obese phenotype via reduced serum HOMA-IR and leptin:adiponectin ratio, and locally via attenuation of epididymal adipose tissue crown-like structure formation, adipocyte size, and inflammatory transcription factor (NFκBp65 and STAT3) activation. Therefore, navy bean supplementation improved obese intestinal health (microbiota and epithelial barrier integrity) and attenuated the severity of the obese phenotype.


Subject(s)
Diet, High-Fat , Inflammation/physiopathology , Intestinal Mucosa/physiopathology , Phaseolus , Adipokines/metabolism , Adipose Tissue/metabolism , Akkermansia , Animal Feed , Animals , Body Weight , Carbohydrate Metabolism , Dietary Fiber , Dietary Supplements , Epithelial Cells/metabolism , Epithelial Cells/pathology , Feces , Fermentation , Fluorescein-5-isothiocyanate , Gastrointestinal Microbiome , Intestinal Mucosa/metabolism , Male , Mice , Mice, Inbred C57BL , Obesity/metabolism , Permeability , Phenotype , Prevotella , RNA, Ribosomal, 16S/metabolism , Verrucomicrobia
13.
Appl Physiol Nutr Metab ; 43(9): 893-901, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29522694

ABSTRACT

The potential for a chickpea-supplemented diet (rich in fermentable nondigestible carbohydrates and phenolic compounds) to modify the colonic microenvironment and attenuate the severity of acute colonic inflammation was investigated. C57Bl/6 male mice were fed a control basal diet or basal diet supplemented with 20% cooked chickpea flour for 3 weeks prior to acute colitis onset induced by 7-day exposure to dextran sodium sulfate (DSS; 2% w/v in drinking water) and colon and serum levels of inflammatory mediators were assessed. Despite an equal degree of DSS-induced epithelial barrier histological damage and clinical symptoms between dietary groups, biomarkers of the ensuing inflammatory response were attenuated by chickpea pre-feeding, including reduced colon tissue activation of nuclear factor kappa B and inflammatory cytokine production (tumor necrosis factor alpha and interleukin (IL)-18). Additionally, colon protein expression of anti-inflammatory (IL-10) and epithelial repair (IL-22 and IL-27) cytokines were increased by chickpea pre-feeding. Furthermore, during acute colitis, chickpea pre-feeding increased markers of enhanced colonic function, including Relmß and IgA gene expression. Collectively, chickpea pre-feeding modulated the baseline function of the colonic microenvironment, whereby upon induction of acute colitis, the severity of the inflammatory response was attenuated.


Subject(s)
Cicer , Colitis/diet therapy , Inflammation/diet therapy , Animals , Biomarkers/metabolism , Colitis/chemically induced , Dextran Sulfate , Diet , Disease Models, Animal , Flour , Inflammation/chemically induced , Interleukin-18/metabolism , Interleukins/metabolism , Intestinal Mucosa/metabolism , Male , Mice, Inbred C57BL , NF-kappa B/metabolism , Tumor Necrosis Factor-alpha/metabolism , Interleukin-22
14.
J Nutr Biochem ; 55: 41-52, 2018 05.
Article in English | MEDLINE | ID: mdl-29413488

ABSTRACT

Marine-derived n-3 polyunsaturated fatty acids (PUFAs), such as eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), have been shown to inhibit mammary carcinogenesis. However, evidence regarding plant-based α-linolenic acid (ALA), the major n-3 PUFA in the Western diet, remains equivocal. The objective of this study was to examine the effect of lifelong exposure to plant- or marine-derived n-3 PUFAs on pubertal mammary gland and tumor development in MMTV-neu(ndl)-YD5 mice. It is hypothesized that lifelong exposure to n-3 PUFA reduces terminal end buds during puberty leading to delayed tumor onset, volume and multiplicity. It is further hypothesized that plant-derived n-3 PUFAs will exert dose-dependent effects. Harems of MMTV-FVB males were bred with wild-type females and fed either a (1) 10% safflower (10% SF, n-6 PUFA, control), (2) 10% flaxseed (10% FS), (3) 7% safflower plus 3% flaxseed (3% FS) or (4) 7% safflower plus 3% menhaden (3% FO) diet. Female offspring were maintained on parental diets. Compared to SF, 10% FS and 3% FO reduced (P<.05) terminal end buds at 6 weeks and tumor volume and multiplicity at 20 weeks. A dose-dependent reduction of tumor volume and multiplicity was observed in mice fed 3% and 10% FS. Antitumorigenic effects were associated with altered HER2, pHER-2, pAkt and Ki-67 protein expression. Compared to 10% SF, 3% FO significantly down-regulated expression of genes involved in eicosanoid synthesis and inflammation. From this, it can be estimated that ALA was 1/8 as potent as EPA+DHA. Thus, marine-derived n-3 PUFAs have greater potency versus plant-based n-3 PUFAs.


Subject(s)
Fatty Acids, Omega-3/pharmacology , Fish Oils/pharmacology , Mammary Neoplasms, Experimental/prevention & control , Animals , Body Weight/drug effects , Eating/drug effects , Fatty Acids/analysis , Fatty Acids, Omega-3/chemistry , Female , Gene Expression Regulation, Neoplastic/drug effects , Linseed Oil/chemistry , Male , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice, Inbred Strains , Proto-Oncogene Proteins c-akt/metabolism , Puberty/drug effects , Receptor, ErbB-2/metabolism , Safflower Oil/chemistry
15.
Nutrients ; 9(12)2017 Nov 27.
Article in English | MEDLINE | ID: mdl-29186929

ABSTRACT

Obesity is a global health concern with rising prevalence that increases the risk of developing other chronic diseases. A causal link connecting overnutrition, the development of obesity and obesity-associated co-morbidities is visceral adipose tissue (AT) dysfunction, characterized by changes in the cellularity of various immune cell populations, altered production of inflammatory adipokines that sustain a chronic state of low-grade inflammation and, ultimately, dysregulated AT metabolic function. Therefore, dietary intervention strategies aimed to halt the progression of obese AT dysfunction through any of the aforementioned processes represent an important active area of research. In this connection, fish oil-derived dietary long-chain n-3 polyunsaturated fatty acids (PUFA) in the form of eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) have been demonstrated to attenuate obese AT dysfunction through multiple mechanisms, ultimately affecting AT immune cellularity and function, adipokine production, and metabolic signaling pathways, all of which will be discussed herein.


Subject(s)
Adipose Tissue/drug effects , Fatty Acids, Omega-3/pharmacology , Immunomodulation , Obesity/drug therapy , Adipokines/blood , Adipose Tissue/metabolism , Adiposity/drug effects , Animals , Disease Models, Animal , Docosahexaenoic Acids/pharmacology , Eicosapentaenoic Acid/pharmacology , Humans , Immunity, Cellular , Inflammation/blood , Inflammation/drug therapy , Obesity/blood , Signal Transduction
16.
J Nutr Biochem ; 32: 29-38, 2016 06.
Article in English | MEDLINE | ID: mdl-27142734

ABSTRACT

Typically fatty acids (FA) exert differential immunomodulatory effects with n-3 [α-linolenic acid (ALA), eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA)] and n-6 [linoleic acid (LA) and arachidonic acid (AA)] exerting anti- and pro-inflammatory effects, respectively. This over-simplified interpretation is confounded by a failure to account for conversion of the parent FA (LA and ALA) to longer-chain bioactive products (AA and EPA/DHA, respectively), thereby precluding discernment of the immunomodulatory potential of specific FA. Therefore, we utilized the Δ6-desaturase model, wherein knockout mice (D6KO) lack the Fads2 gene encoding for the rate-limiting enzyme that initiates FA metabolism, thereby providing a model to determine specific FA immunomodulatory effects. Wild-type (WT) and D6KO mice were fed one of four isocaloric diets differing in FA source (9weeks): corn oil (LA-enriched), arachidonic acid single cell oil (AA-enriched), flaxseed oil (ALA-enriched) or menhaden fish oil (EPA/DHA-enriched). Splenic mononuclear cell cytokine production in response to lipopolysaccharide (LPS), T-cell receptor (TCR) and anti-CD40 stimulation was determined. Following LPS stimulation, AA was more bioactive compared to LA, by increasing inflammatory cytokine production of IL-6 (1.2-fold) and TNFα (1.3-fold). Further, LPS-stimulated IFNγ production in LA-fed D6KO mice was reduced 5-fold compared to LA-fed WT mice, indicating that conversion of LA to AA was necessary for cytokine production. Conversely, ALA exerted an independent immunomodulatory effect from EPA/DHA and all n-3 FA increased LPS-stimulated IL-10 production versus LA and AA. These data definitively identify specific immunomodulatory effects of individual FA and challenge the simplified view of the immunomodulatory effects of n-3 and n-6 FA.


Subject(s)
Dietary Supplements , Fatty Acid Desaturases/metabolism , Fatty Acids, Omega-3/administration & dosage , Fatty Acids, Omega-6/administration & dosage , Immunomodulation , Leukocytes, Mononuclear/immunology , Spleen/immunology , Animals , Cells, Cultured , Crosses, Genetic , Cytokines/metabolism , Fatty Acid Desaturases/genetics , Fatty Acids, Omega-3/metabolism , Fatty Acids, Omega-6/metabolism , Female , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Lipopolysaccharides/toxicity , Male , Mice , Mice, Knockout , Spleen/cytology , Spleen/drug effects , Spleen/metabolism
17.
J Nutr Biochem ; 34: 61-72, 2016 08.
Article in English | MEDLINE | ID: mdl-27208584

ABSTRACT

Adipocyte-macrophage cross-talk propagates immune responses in obese adipose tissue (AT). Long-chain n-3 polyunsaturated fatty acids (LC n-3 PUFA) mitigate inflammation, partly through up-regulation of adiponectin; however, specific mechanisms are unclear. We determined if adipocyte-macrophage cross-talk could be mitigated by dietary LC n-3 PUFA and if this was dependent on adiponectin-mediated signaling. We utilized an in vitro co-culture model mimicking the ratio of adipocytes:macrophages in obese AT, whereby 3T3-L1 adipocytes were co-cultured with splenic CD11b(+) macrophages from C57BL/6 mice fed high-fat control (HF-CON; 34% w/w fat) or fish oil diets (HF-FO; 34% w/w fat containing 7.6% w/w FO), as well as mice fed low-fat control (LF-CON; 10% w/w fat) or FO diets (LF-FO; 10% w/w fat containing 3% w/w FO). Co-culture conditions tested effects of soluble mediator-driven mechanisms (trans-well system), cell contact and low-dose lipopolysaccharide (LPS) mimicking acute or chronic inflammatory conditions. HF-FO macrophages from acute LPS-stimulated trans-well co-cultures had decreased mRNA expression of Casp1, Il1ß and Il18, as well as cellular caspase-1 activity compared to HF-CON macrophages (P≤.05). Moreover, adipocytes from acute LPS-stimulated HF-FO co-cultures had decreased caspase-1 activity and decreased IL-1ß/IL-18 levels following chronic LPS pretreatment compared to HF-CON co-cultures (P≤.05). Additionally, in contact co-cultures with adiponectin-neutralizing antibody, the FO-mediated modulation of NFκB activity and decrease in phosphorylated p65 NFκB, expression of NLRP3 inflammasome genes, M1 macrophage marker genes and inflammatory cytokine/chemokine secretion were controlled partly through adiponectin, while cellular caspase-1 activity and IL-1ß/1L-18 levels were decreased independently of adiponectin (P≤.05). LC n-3 PUFA may decrease the intensity of adipocyte-macrophage cross-talk to mitigate obesity-associated pathologies.


Subject(s)
Adipocytes, White/metabolism , Dietary Supplements , Fatty Acids, Omega-3/therapeutic use , Inflammasomes/metabolism , Macrophages/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Obesity/diet therapy , 3T3-L1 Cells , Adipocytes, White/immunology , Adipocytes, White/pathology , Animals , Anti-Inflammatory Agents, Non-Steroidal/analysis , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , CD11b Antigen/metabolism , Cell Communication , Cells, Cultured , Coculture Techniques , Diet, High-Fat/adverse effects , Dietary Supplements/analysis , Fatty Acids, Omega-3/analysis , Fatty Acids, Omega-3/metabolism , Female , Fish Oils/chemistry , Fish Oils/therapeutic use , Gene Expression Regulation , Inflammasomes/immunology , Macrophages/immunology , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Obesity/immunology , Obesity/metabolism , Obesity/pathology , Spleen/immunology , Spleen/metabolism , Spleen/pathology
18.
Mol Nutr Food Res ; 60(3): 621-30, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26616354

ABSTRACT

SCOPE: CD8(+) T cell/adipocyte paracrine interactions represent a critical step in the development of the obese inflammatory phenotype that is disrupted by long-chain n-3 PUFA. Our objective was to determine the effect of flaxseed-derived n-3 PUFA (α-linolenic acid) on these paracrine interactions. METHODS AND RESULTS: C57BL/6 mice were fed 3.5% flaxseed oil (FX) + 3.5% corn oil diet w/w or an isocaloric 7% corn oil w/w control diet (CON) for 3 wk. 3T3-L1 adipocytes and purified primary splenic CD8(+) T cells were cocultured at an obese cellular ratio (10% CD8(+) T cells) and LPS-stimulated (10 ng/mL mimicking obese circulating endotoxin levels) for 24 h. FX cocultures reduced (i) secreted IL-6, tumor necrosis factor α (TNF-α), macrophage chemoattractant protein 1 (MCP-1), macrophage inflammatory protein 1α (MIP-1α), and RANTES (regulated on activation, normal T cell expressed and secreted) levels; (ii) activation of inflammatory transcription factors NFκB (nuclear factor kappa-light-chain-enhancer of activated B cell) p65 and signal transducer and activator of transcription-3 (STAT3); and (iii) RAW264.7 macrophage chemotaxis versus CON (p ≤ 0.05). Coculture of pre-inflamed adipocytes (10 ng/mL LPS, 24 h prior to CD8(+) T-cell addition) resulted in reduced secretion of IL-6, IL-1ß, MCP-1, MCP-3, MIP-1ß, and RANTES in FX cocultures versus CON (p ≤ 0.05). CONCLUSION: FX exerts an anti-chemotactic and anti-inflammatory effect on CD8(+) T cell/adipocyte paracrine interactions (cross-talk), which has the potential to mitigate macrophage chemotaxis which drives components of the obese phenotype.


Subject(s)
Adipocytes/drug effects , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , CD8-Positive T-Lymphocytes/drug effects , Chemotaxis/drug effects , Linseed Oil/pharmacology , Adipocytes/metabolism , Animals , CD8-Positive T-Lymphocytes/metabolism , Cell Communication/drug effects , Coculture Techniques , Dietary Fats, Unsaturated/pharmacology , Fatty Acids/metabolism , Fatty Acids, Omega-3/pharmacology , Gene Expression Regulation/drug effects , Lipopolysaccharides/pharmacology , Male , Mice, Inbred C57BL
19.
Front Nutr ; 2: 31, 2015.
Article in English | MEDLINE | ID: mdl-26528480

ABSTRACT

Adipose tissue (AT) macrophages (ATM) play a key role in obesity-associated pathologies, and their phenotype can be influenced by the local tissue microenvironment. Interestingly, long-chain n-3 polyunsaturated fatty acids (LC n-3 PUFA) and the LC n-3 PUFA-upregulated adipokine, adiponectin (Ad), may mitigate excessive ATM inflammatory M1-polarization responses. However, to what extent LC n-3 PUFA and Ad work in concert to affect macrophage phenotype has not been examined. Thus, we used an established ex vivo AT organ culture model using visceral AT from mice fed a control (CON; 10% w/w safflower oil) n-6 PUFA-rich diet or an isocaloric fish oil (FO; 3% w/w menhaden oil + 7% w/w safflower oil)-derived LC n-3 PUFA-rich diet to generate AT conditioned media (ACM). We then evaluated if CON or FO ACM affected macrophage polarization markers in a model designed to mimic acute [18 h ACM plus lipopolysaccharide (LPS) for the last 6 h] or chronic (macrophages treated with LPS-challenged CON or FO ACM for 24 h) inflammation ± Ad-neutralizing antibody and the LPS-neutralizing agent, polymyxin B. In the acute inflammation model, macrophages treated with FO ACM had decreased lipid uptake and mRNA expression of M1 markers (Nos2, Nfκb, Il6, Il18, Ccl2, and Ccl5) compared with CON ACM (p ≤ 0.05); however, these effects were largely attenuated when Ad was neutralized (p > 0.05). Furthermore, in the chronic inflammation model, macrophages treated with FO ACM had decreased mRNA expression of M1 markers (Nos2, Tnfα, Ccl2, and Il1ß) and IL-6 and CCL2 secretion (p ≤ 0.05); however, some of these effects were lost when Ad was neutralized, and were further exacerbated when both Ad and LPS were neutralized. Taken together, this work shows that LC n-3 PUFA and Ad work in concert to suppress certain M1 macrophage responses. Thus, future strategies to modulate the ATM phenotype should consider the role of both LC n-3 PUFA and Ad in mitigating obese AT inflammation.

20.
Lipids Health Dis ; 14: 53, 2015 Jun 06.
Article in English | MEDLINE | ID: mdl-26048287

ABSTRACT

Circulating levels of triacylglycerol (TG) is a recognized risk factor for developing cardiovascular disease, a leading cause of death worldwide. The Institute of Medicine and the American Heart Association both recommend the consumption of n-3 polyunsaturated fatty acids (PUFA), specifically eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), to reduce serum TG in hyperlipidemic individuals. Additionally, a number of systematic reviews have shown that individuals with any degree of dyslipidemia, elevated serum TG and/or cholesterol, may benefit from a 20-30% reduction in serum TG after consuming n-3 PUFA derived from marine sources. Given that individuals with serum lipid levels ranging from healthy to borderline dyslipidemic constitute a large portion of the population, the focus of this review was to assess the potential for n-3 PUFA consumption to reduce serum TG in such individuals. A total of 1341 studies were retrieved and 38 clinical intervention studies, assessing 2270 individuals, were identified for inclusion in the current review. In summary, a 9-26% reduction in circulating TG was demonstrated in studies where ≥ 4 g/day of n-3 PUFA were consumed from either marine or EPA/DHA-enriched food sources, while a 4-51% reduction was found in studies where 1-5 g/day of EPA and/or DHA was consumed through supplements. Overall, this review summarizes the current evidence with regards to the beneficial effect of n-3 PUFA on circulating TG levels in normolipidemic to borderline hyperlipidemic, otherwise healthy, individuals. Thus demonstrating that n-3 PUFA may play an important role in the maintenance of cardiovascular health and disease prevention.


Subject(s)
Fatty Acids, Omega-3/therapeutic use , Hyperlipidemias/drug therapy , Triglycerides/blood , Adult , Aged , Diet , Dietary Supplements , Female , Humans , Hyperlipidemias/blood , Male , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL
...