Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Nutrients ; 16(13)2024 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-38999834

RESUMO

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.


Assuntos
Ácidos Graxos Ômega-3 , Inflamação , Resistência à Insulina , Macrófagos , Animais , Macrófagos/metabolismo , Macrófagos/efeitos dos fármacos , Camundongos , Ácidos Graxos Ômega-3/farmacologia , Inflamação/metabolismo , Células RAW 264.7 , Ácidos Graxos Ômega-6/farmacologia , Insulina/metabolismo , Citocinas/metabolismo , Obesidade/metabolismo , Transdução de Sinais/efeitos dos fármacos , Ratos , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/efeitos dos fármacos
2.
Nutrients ; 14(14)2022 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-35889783

RESUMO

Short-chain fatty acids (SCFA) produced from dietary non-digestible carbohydrate fermentation have metabolic effects in skeletal muscle; however, their effect on inflammatory mediator production is unknown. In this study, L6 myotubes were cultured with individual SCFA (acetate, propionate, and butyrate) at 0.5 mM and 2.5 mM ± 10 ng/mL lipopolysaccharide (LPS) or ± 500 µM palmitic acid (PA) for 24 h. In response to LPS, only butyrate had an effect at the lower concentration (0.5 mM), whereas at the higher concentration (2.5 mM) both propionate and butyrate reduced MCP-1, MIP-1α, and RANTES secretion (p < 0.05), and only butyrate reduced IL-6 secretion and intracellular protein levels of phospho-STAT3 (p < 0.05). In response to PA, 0.5 mM butyrate reduced protein expression of phospho-NFκB p65 and the secretion of IL-6, MIP-1α, and MCP-1, whereas all three SCFA reduced RANTES secretion (p < 0.05). At the 2.5 mM SCFA concentration combined with PA stimulation, all three SCFA reduced intracellular protein expression of phospho-NFκB p65 and phospho-STAT3 and secreted protein levels of MCP-1, IL-6, and RANTES, whereas only butyrate reduced secretion of MIP-1α (p < 0.05). Thus, SCFA exhibit differential effects on inflammatory mediator expression in response to LPS and PA stimulation, which has implications for their individual impacts on inflammation-mediated skeletal muscle dysfunction.


Assuntos
Lipopolissacarídeos , Propionatos , Butiratos/metabolismo , Quimiocina CCL3 , Quimiocina CCL5 , Carboidratos da Dieta , Ácidos Graxos Voláteis/metabolismo , Ácidos Graxos Voláteis/farmacologia , Interleucina-6 , Lipopolissacarídeos/farmacologia , Fibras Musculares Esqueléticas/metabolismo , Ácido Palmítico/farmacologia , Propionatos/metabolismo
3.
Food Funct ; 12(14): 6348-6362, 2021 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-34105575

RESUMO

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.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Inflamação/dietoterapia , Malus , Obesidade/dietoterapia , Sobrepeso/dietoterapia , Adulto , Biomarcadores/sangue , Estudos Cross-Over , Citocinas/sangue , Feminino , Frutas , Humanos , Inflamação/sangue , Interleucina-6/sangue , Leucócitos Mononucleares/metabolismo , Masculino , Doenças Metabólicas/epidemiologia , Pessoa de Meia-Idade , Período Pós-Prandial , Fatores de Risco , Resultado do Tratamento
4.
J Nutr Biochem ; 95: 108763, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33965532

RESUMO

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.


Assuntos
Apoptose/efeitos dos fármacos , Óleos de Peixe/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Inflamação/tratamento farmacológico , Neoplasias Mamárias Experimentais/tratamento farmacológico , Obesidade/induzido quimicamente , Tecido Adiposo/efeitos dos fármacos , Animais , Peso Corporal/efeitos dos fármacos , Neoplasias da Mama , Linhagem Celular Tumoral , Suplementos Nutricionais , Ácidos Graxos/química , Feminino , Óleos de Peixe/administração & dosagem , Humanos , Glândulas Mamárias Animais/química , Camundongos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Distribuição Aleatória , Receptor ErbB-2
5.
Am J Clin Nutr ; 114(2): 752-763, 2021 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-33964852

RESUMO

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.


Assuntos
Inflamação/metabolismo , Leucócitos Mononucleares/metabolismo , Malus , Sobrepeso , Adulto , Biomarcadores/metabolismo , Humanos
6.
PLoS One ; 15(9): e0238522, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32946478

RESUMO

The effects of feeding frequency on postprandial response of circulating appetite-regulating hormones, insulin, glucose and amino acids, and on physical activity, energy expenditure, and respiratory quotient were studied in healthy adult cats. Two experiments were designed as a 2 x 3 replicated incomplete Latin square design. Eight cats, with an average body weight (BW) of 4.34 kg ± 0.04 and body condition score (BCS) of 5.4 ± 1.4 (9 point scale), were fed isocaloric amounts of a commercial adult maintenance canned cat food either once (0800 h) or four times daily (0800 h, 1130 h, 1500 h, 1830 h). Study 1 consisted of three 21-d periods. On day 14, two fasted and 11 postprandial blood samples were collected over 24 hours to measure plasma concentrations of ghrelin, GLP-1, GIP, leptin, PYY, insulin and amino acids, and whole blood glucose. Physical activity was monitored from day 15 to 21 of each period. In Study 2 indirect calorimetry was performed on the last day of each period. Body weight was measured weekly and feed intake recorded daily in both experiments. No effect of feeding regimen on BW was detected. Cats eating four times daily had lesser plasma concentrations of GIP and GLP-1 (P<0.05) and tended to have lesser plasma PYY concentrations (P<0.1). Plasma leptin and whole blood glucose concentrations did not differ between regimens (P>0.1). Cats fed once daily had a greater postprandial plasma amino acid response, and greater plasma ghrelin and insulin concentrations (P<0.05). Physical activity was greater in cats fed four times (P<0.05), though energy expenditure was similar between treatments at fasting and in postprandial phases. Finally, cats eating one meal had a lower fasting respiratory quotient (P<0.05). Overall, these data indicate that feeding once a day may be a beneficial feeding management strategy for indoor cats to promote satiation and lean body mass.


Assuntos
Aminoácidos/metabolismo , Regulação do Apetite , Gatos/fisiologia , Comportamento Alimentar , Hormônios/metabolismo , Aminoácidos/sangue , Ração Animal/análise , Animais , Apetite , Glicemia/análise , Glicemia/metabolismo , Gatos/sangue , Metabolismo Energético , Feminino , Grelina/sangue , Grelina/metabolismo , Hormônios/sangue , Insulina/sangue , Insulina/metabolismo , Masculino , Fotoperíodo , Condicionamento Físico Animal , Respiração
7.
Methods Mol Biol ; 2184: 111-130, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32808222

RESUMO

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.


Assuntos
Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/citologia , Técnicas de Cocultura/métodos , Macrófagos/citologia , Células 3T3-L1 , Adipócitos , Tecido Adiposo/citologia , Animais , Diferenciação Celular/fisiologia , Células Cultivadas , Microambiente Celular/fisiologia , Inflamação/patologia , Camundongos , Obesidade/patologia , Baço/citologia
8.
J Nutr Biochem ; 86: 108488, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32827664

RESUMO

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.


Assuntos
Adipócitos/imunologia , Linfócitos T CD4-Positivos/citologia , Ácidos Graxos Ômega-3/metabolismo , Inflamação/tratamento farmacológico , Obesidade/imunologia , Células 3T3-L1 , Tecido Adiposo/imunologia , Animais , Quimiocinas/metabolismo , Técnicas de Cocultura , Dieta , Modelos Animais de Doenças , Feminino , Óleos de Peixe/metabolismo , Inflamação/sangue , Inflamação/imunologia , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Subunidade p50 de NF-kappa B/metabolismo , Obesidade/sangue , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais
9.
Nutrients ; 12(5)2020 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-32408695

RESUMO

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.


Assuntos
Adipócitos/efeitos dos fármacos , Indutores da Angiogênese/metabolismo , Anti-Inflamatórios/farmacologia , Flavonóis/farmacologia , Malus/química , Células 3T3-L1 , Tecido Adiposo/efeitos dos fármacos , Animais , Cobalto , Inflamação , Lipopolissacarídeos , Camundongos , Obesidade , PPAR gama , Transdução de Sinais/efeitos dos fármacos
10.
Br J Nutr ; 123(7): 807-817, 2020 04 14.
Artigo em Inglês | MEDLINE | ID: mdl-31902373

RESUMO

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.


Assuntos
Apolipoproteína B-48/sangue , Ácidos Graxos/sangue , Frutas , Malus , Triglicerídeos/sangue , Adulto , Idoso , Glicemia , Estudos Cross-Over , Dieta , Feminino , Humanos , Insulina/sangue , Masculino , Refeições , Pessoa de Meia-Idade , Período Pós-Prandial , Adulto Jovem
11.
Nutrients ; 12(12)2020 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-33419361

RESUMO

Lifestyle habits, such as the consumption of a healthy diet, may prevent up to 30-50% of breast cancer (BC) cases. Dietary fats are of specific interest, as research provides strong evidence regarding the association of dietary fats and BC. However, there is limited research on the role of different types of fats including polyunsaturated (PUFA), monounsaturated (MUFA), and saturated fatty acids (SFA). The objective of this study was to determine the effects of lifelong exposure to various dietary fats on mammary tumour development over a 20-week period. Female heterozygous MMTV-neu (ndl) YD5 mouse models were fed five maternal diets containing (1) 10% safflower oil (n-6 PUFA, control), (2) 3% menhaden oil + 7% safflower oil (marine n-3 PUFA, control), (3) 3% flaxseed + 7% safflower oil (plant-based n-3 PUFA), (4) 10% olive oil (MUFA), or (5) 10% lard (SFA). The primary measures, tumour latency, volume, and multiplicity differed by diet treatment in the following general order, n-6 PUFA > plant n-3 PUFA, SFA, MUFA > marine n-3 PUFA. Overall, these findings show that the quality of the diet plays a significant role influencing mammary tumour outcomes.


Assuntos
Dieta/veterinária , Ácidos Graxos/administração & dosagem , Genes erbB-2/genética , Neoplasias Mamárias Animais/patologia , Ração Animal , Animais , Ácidos Graxos/classificação , Feminino , Neoplasias Mamárias Animais/dietoterapia , Neoplasias Mamárias Animais/genética , Camundongos , Camundongos Transgênicos , Resultado do Tratamento
12.
J Nutr Biochem ; 76: 108243, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31760229

RESUMO

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.


Assuntos
Adipócitos/metabolismo , Linfócitos T CD8-Positivos/citologia , Ácidos Graxos Ômega-3/metabolismo , Óleos de Peixe/metabolismo , Inflamação/metabolismo , Macrófagos/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Células 3T3-L1 , Animais , Peso Corporal , Técnicas de Cocultura , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Células RAW 264.7/citologia , RNA Mensageiro/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais
13.
J Nutr Biochem ; 72: 108216, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31476608

RESUMO

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.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Óleos de Peixe/farmacologia , Microbioma Gastrointestinal/efeitos dos fármacos , Intestinos/efeitos dos fármacos , Obesidade/dietoterapia , Adipocinas/sangue , Animais , Peso Corporal/efeitos dos fármacos , Colo/efeitos dos fármacos , Colo/fisiologia , Suplementos Nutricionais , Ingestão de Alimentos/efeitos dos fármacos , Ácidos Graxos Ômega-3/metabolismo , Fezes/microbiologia , Microbioma Gastrointestinal/genética , Teste de Tolerância a Glucose , Íleo/efeitos dos fármacos , Íleo/fisiologia , Intestinos/fisiologia , Gordura Intra-Abdominal/efeitos dos fármacos , Gordura Intra-Abdominal/patologia , Masculino , Camundongos Endogâmicos C57BL , Obesidade/etiologia , Paniculite/etiologia , Paniculite/prevenção & controle
14.
J Nutr Biochem ; 70: 91-104, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31195365

RESUMO

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.


Assuntos
Dieta Hiperlipídica , Inflamação/fisiopatologia , Mucosa Intestinal/fisiopatologia , Phaseolus , Adipocinas/metabolismo , Tecido Adiposo/metabolismo , Akkermansia , Ração Animal , Animais , Peso Corporal , Metabolismo dos Carboidratos , Fibras na Dieta , Suplementos Nutricionais , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Fezes , Fermentação , Fluoresceína-5-Isotiocianato , Microbioma Gastrointestinal , Mucosa Intestinal/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/metabolismo , Permeabilidade , Fenótipo , Prevotella , RNA Ribossômico 16S/metabolismo , Verrucomicrobia
15.
Appl Physiol Nutr Metab ; 44(5): 455-460, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30794435

RESUMO

This Horizons is part of a series that identifies key, forward-thinking research questions and challenges that need to be addressed. Specifically, this Horizons paper discusses research in nutritional supplements and nutraceuticals for health, physical activity, and performance, and is the product of a discussion by an expert panel that took place in January 2018 prior to the Canadian Nutrition Society Thematic Conference "Advances in Sport Nutrition from Daily Living to High Performance Sport". The objective of this Horizons paper was to identify core considerations for future studies for this research area, and how scientists can be leaders in the field to ensure the best quality science is available for decision makers. It is strongly recommended that the various elements highlighted throughout this Horizons paper will increase the awareness of the significant before-, during-, and after-research due-diligence required to produce research of the highest quality. While it is recognized that many scientists will not be able to meet all of these aspects, it is nonetheless important to consider the points outlined and to recognize that those elements that are not met in studies may be significant limitations. Highlights Research questions that are hypothesis-driven are the strongest, and when combined with careful planning of the study, the result will often be of the best quality. Studies with a strong experimental design help discern between evidence-based findings and those that have not been substantiated.


Assuntos
Suplementos Nutricionais , Exercício Físico , Fenômenos Fisiológicos da Nutrição Esportiva , Ciências da Nutrição e do Esporte/tendências , Animais , Canadá , Humanos
16.
Appl Physiol Nutr Metab ; 43(9): 893-901, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-29522694

RESUMO

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.


Assuntos
Cicer , Colite/dietoterapia , Inflamação/dietoterapia , Animais , Biomarcadores/metabolismo , Colite/induzido quimicamente , Sulfato de Dextrana , Dieta , Modelos Animais de Doenças , Farinha , Inflamação/induzido quimicamente , Interleucina-18/metabolismo , Interleucinas/metabolismo , Mucosa Intestinal/metabolismo , Masculino , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Interleucina 22
17.
J Nutr Biochem ; 55: 41-52, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29413488

RESUMO

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.


Assuntos
Ácidos Graxos Ômega-3/farmacologia , Óleos de Peixe/farmacologia , Neoplasias Mamárias Experimentais/prevenção & controle , Animais , Peso Corporal/efeitos dos fármacos , Ingestão de Alimentos/efeitos dos fármacos , Ácidos Graxos/análise , Ácidos Graxos Ômega-3/química , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Óleo de Semente do Linho/química , Masculino , Neoplasias Mamárias Experimentais/metabolismo , Neoplasias Mamárias Experimentais/patologia , Camundongos Endogâmicos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Puberdade/efeitos dos fármacos , Receptor ErbB-2/metabolismo , Óleo de Cártamo/química
18.
J Nutr Biochem ; 34: 61-72, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27208584

RESUMO

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.


Assuntos
Adipócitos Brancos/metabolismo , Suplementos Nutricionais , Ácidos Graxos Ômega-3/uso terapêutico , Inflamassomos/metabolismo , Macrófagos/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/antagonistas & inibidores , Obesidade/dietoterapia , Células 3T3-L1 , Adipócitos Brancos/imunologia , Adipócitos Brancos/patologia , Animais , Anti-Inflamatórios não Esteroides/análise , Anti-Inflamatórios não Esteroides/química , Anti-Inflamatórios não Esteroides/metabolismo , Anti-Inflamatórios não Esteroides/uso terapêutico , Antígeno CD11b/metabolismo , Comunicação Celular , Células Cultivadas , Técnicas de Cocultura , Dieta Hiperlipídica/efeitos adversos , Suplementos Nutricionais/análise , Ácidos Graxos Ômega-3/análise , Ácidos Graxos Ômega-3/metabolismo , Feminino , Óleos de Peixe/química , Óleos de Peixe/uso terapêutico , Regulação da Expressão Gênica , Inflamassomos/imunologia , Macrófagos/imunologia , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Obesidade/imunologia , Obesidade/metabolismo , Obesidade/patologia , Baço/imunologia , Baço/metabolismo , Baço/patologia
19.
Clin Nutr ; 35(6): 1347-1353, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-26972089

RESUMO

BACKGROUND & AIM: During cancer development, fat loss occurs in most cancer patients. Characterization of the behavior of fat loss from visceral (VAT) and subcutaneous adipose tissue (SAT) depots has not been established. The first objective of this study was to assess the intensity and time course of changes in VAT and SAT depots of advanced cancer patients in the year preceding death. Secondly, this study explored the differences in adipokine content and fatty acid composition between VAT and SAT depots and in relation to changes in fat mass. METHODS: Longitudinal quantitative analyses of computed tomography images was conducted to define changes in adipose tissue cross sectional areas in fat depots in advanced colorectal and cholangiocarcinoma cancer patients (n = 46) at mean time points corresponding to 9, 6, 3 and 1 month before death. Proportions of adipose tissue fatty acid and adipokine content were characterized in a second cohort of advanced colorectal cancer patients (n = 16). RESULTS: On average, loss of total adipose tissue (TAT) happens at all time intervals but there is an elevation in the intensity of loss close to death. Nine months from death, 42% of patients were losing fat (Mean TAT cross sectional area change = -0.2 ± 13 cm2) whereas within one month from death, fat wasting was observed in 78% of patients (-60.1 ± 9.2 cm2, P = 0.001). However, loss of TAT did not reflect changes in VAT and SAT in the same direction or intensity. Intensity of VAT loss remains constant throughout the disease progression whereas SAT is more likely to be gained further way from death. Nine month prior to death, mean change in cross sectional area of VAT was -7.9 ± 6.8 cm2 whereas, mean change in CSA of SAT was 7.4 ± 7.7 cm2 (p = 0.03). One month before death, mean VAT and SAT absolute changes were -24.5 ± 4.9 cm2 and -34.5 ± 5.2 cm2, respectively (p = 0.05). Moreover, fat losing patients had higher proportions of polyunsaturated fatty acids, especially n-6 fatty acids, in VAT compared to patients who were gaining fat (mean = 15.4% in losing group vs. 13.4% in gaining group; p = 0.03). VAT contained more monocyte chemoattractant protein-1 than SAT, whereas leptin levels were higher in SAT. CONCLUSIONS: Further from death, VAT and SAT behave differently whereas close to death, accelerated loss occurs in both depots. These differences are further characterized by differences in fatty acid composition and adipokine levels.


Assuntos
Ácidos Graxos Ômega-6/análise , Gordura Intra-Abdominal/química , Gordura Intra-Abdominal/patologia , Neoplasias/patologia , Gordura Subcutânea/química , Gordura Subcutânea/patologia , Adipocinas/análise , Adulto , Idoso , Composição Corporal , Colangiocarcinoma/mortalidade , Colangiocarcinoma/patologia , Neoplasias Colorretais/mortalidade , Neoplasias Colorretais/patologia , Ácidos Graxos/análise , Feminino , Humanos , Estudos Longitudinais , Masculino , Pessoa de Meia-Idade , Neoplasias/mortalidade , Estudos Retrospectivos , Tomografia Computadorizada por Raios X
20.
Mol Nutr Food Res ; 60(3): 621-30, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26616354

RESUMO

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.


Assuntos
Adipócitos/efeitos dos fármacos , Anti-Inflamatórios não Esteroides/farmacologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Quimiotaxia/efeitos dos fármacos , Óleo de Semente do Linho/farmacologia , Adipócitos/metabolismo , Animais , Linfócitos T CD8-Positivos/metabolismo , Comunicação Celular/efeitos dos fármacos , Técnicas de Cocultura , Gorduras Insaturadas na Dieta/farmacologia , Ácidos Graxos/metabolismo , Ácidos Graxos Ômega-3/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Lipopolissacarídeos/farmacologia , Masculino , Camundongos Endogâmicos C57BL
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA