Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
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
11.
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
12.
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
13.
J Nutr ; 145(4): 829-38, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25833786

ABSTRACT

BACKGROUND: Obese adipose tissue (AT) inflammation is characterized by dysregulated adipokine production and immune cell accumulation. Cluster of differentiation (CD) 8+ T cell AT infiltration represents a critical step that precedes macrophage infiltration. n-3 (ω-3) Polyunsaturated fatty acids (PUFAs) exert anti-inflammatory effects in obese AT, thereby disrupting AT inflammatory paracrine signaling. OBJECTIVE: We assessed the effect of n-3 PUFAs on paracrine interactions between adipocytes and primary CD8+ T cells co-cultured at the cellular ratio observed in obese AT. METHODS: C57BL/6 mice were fed either a 3% menhaden fish-oil + 7% safflower oil (FO) diet (wt:wt) or an isocaloric 10% safflower oil (wt:wt) control (CON) for 3 wk, and splenic CD8+ T cells were isolated by positive selection (via magnetic microbeads) and co-cultured with 3T3-L1 adipocytes. Co-cultures were unstimulated (cells alone), T cell receptor stimulated, or lipopolysaccharide (LPS) stimulated for 24 h. RESULTS: In LPS-stimulated co-cultures, FO reduced secreted protein concentrations of interleukin (IL)-6 (-42.6%), tumor necrosis factor α (-67%), macrophage inflammatory protein (MIP) 1α (-52%), MIP-1ß (-62%), monocyte chemotactic protein (MCP) 1 (-23%), and MCP-3 (-19%) vs. CON, which coincided with a 74% reduction in macrophage chemotaxis toward secreted chemotaxins in LPS-stimulated FO-enriched co-culture-conditioned media. FO increased mRNA expression of the inflammatory signaling negative regulators monocyte chemoattractant 1-induced protein (Mcpip; +9.3-fold) and suppressor of cytokine signaling 3 (Socs3; +1.7-fold), whereas FO reduced activation of inflammatory transcription factors nuclear transcription factor κB (NF-κB) p65 and signal transducer and activator of transcription 3 (STAT3) by 27% and 33%, respectively. Finally, mRNA expression of the inflammasome components Caspase1 (-36.4%), Nod-like receptor family pyrin domain containing 3 (Nlrp3; -99%), and Il1b (-68.8%) were decreased by FO compared with CON (P ≤ 0.05). CONCLUSION: FO exerted an anti-inflammatory and antichemotactic effect on the cross-talk between CD8+ T cells and adipocytes and has implications in mitigating macrophage-centered AT-driven components of the obese phenotype.


Subject(s)
Adipokines/metabolism , CD8-Positive T-Lymphocytes/drug effects , Fatty Acids, Omega-3/administration & dosage , Fish Oils/administration & dosage , 3T3-L1 Cells , Adipocytes/drug effects , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animals , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation/drug effects , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Chemokine CCL3/genetics , Chemokine CCL3/metabolism , Chemokine CCL4/genetics , Chemokine CCL4/metabolism , Chemokine CCL7/genetics , Chemokine CCL7/metabolism , Interleukin-6/metabolism , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Obesity/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Spleen/cytology , Spleen/drug effects , Spleen/metabolism , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism
14.
Nutrients ; 6(11): 4760-93, 2014 Oct 30.
Article in English | MEDLINE | ID: mdl-25360510

ABSTRACT

Globally, the prevalence of obesity is increasing which subsequently increases the risk of the development of obesity-related chronic diseases. Low-grade chronic inflammation and dysregulated adipose tissue inflammatory mediator/adipokine secretion are well-established in obesity, and these factors increase the risk of developing inflammation-associated cancer. Breast cancer is of particular interest given that increased inflammation within the subcutaneous mammary adipose tissue depot can alter the local tissue inflammatory microenvironment such that it resembles that of obese visceral adipose tissue. Therefore, in obese women with breast cancer, increased inflammatory mediators both locally and systemically can perpetuate inflammation-associated pro-carcinogenic signaling pathways, thereby increasing disease severity. Herein, we discuss some of these inflammation-associated pro-carcinogenic mechanisms of the combined obese breast cancer phenotype and offer evidence that dietary long chain n-3 polyunsaturated fatty acids (PUFA) may have utility in mitigating the severity of obesity-associated inflammation and breast cancer.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/epidemiology , Fatty Acids, Omega-3/administration & dosage , Obesity/drug therapy , Obesity/epidemiology , Paracrine Communication/drug effects , Adiponectin/metabolism , Aromatase/metabolism , Breast Neoplasms/complications , Cytokines/metabolism , Estrogens/metabolism , Fatty Acids, Omega-6/administration & dosage , Female , Humans , Inflammation/complications , Inflammation/drug therapy , Intra-Abdominal Fat/drug effects , Intra-Abdominal Fat/metabolism , Leptin/metabolism , Obesity/complications , Prevalence
SELECTION OF CITATIONS
SEARCH DETAIL