Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Can J Diet Pract Res ; : 1-7, 2022 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-35014539

RESUMEN

Upon moving to a new country and new food environment, 2 important public health issues may be experienced by immigrants as they adapt to their new country of residence, namely a higher prevalence of food insecurity and/or a decline in overall health over time postimmigration. Therefore, improving the food environment experienced by new migrants may be an effective strategy to reduce long-term health complications and improve well-being postimmigration. The aim of this paper is to discuss the potential barriers experienced by new immigrants in the access, availability, and utilization of familiar culturally appropriate foods and the subsequent impact on their food security status. Culturally appropriate foods are foods commonly consumed as part of cultural food traditions and are often staples within the diet; however, limited availability of and/or access to these foods can reduce food security. By understanding the barriers to food security and challenges that may be faced by immigrants and refugees, dietitians will be better equipped to assist these individuals in accessing culturally familiar foods and improve quality of life. In this capacity, dietitians can play a critical public health nutrition role by serving as a conduit for new immigrants to access community resources and navigate a new food environment.

2.
Can J Diet Pract Res ; 83(3): 144-146, 2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-35503895

RESUMEN

Purpose: A pilot study to investigate the impact of the COVID-19 pandemic and shift to online learning and practicum training on dietetics students' perceptions of Partnership for Dietetic Education and Practice (PDEP) competency acquisition and mental health.Methods: Dietetics students (n = 19) at the University of Guelph (2020-2021) were invited to complete an anonymous online survey to assess self-reported online dietetics practicum training experiences including (i) benefits and challenges, (ii) PDEP competency acquisition, and (iii) impact on mental health.Results: The benefits of online dietetics training included schedule flexibility (42.1%), reduced commute time (31.6%), and acquiring virtual counselling experience (21.1%). Reported challenges were insufficient communication with preceptors (36.8%), increased project workload (57.9%), and technology (15.8%). In online practicum placements, 52.6% of dietetics students reported adequately acquiring all PDEP competencies, with Nutrition Care identified as the most challenging to obtain (63.2%). A negative impact on mental health and increased levels of stress/anxiety were reported in 94.7% of trainees. Notably, 63.2% of students favoured continuation of online dietetics training through a hybrid or entirely online format.Conclusion: Online dietetics training has the potential to complement the traditional in-person model; however, further adaptation is required to optimize PDEP competency acquisition and students' mental health.


Asunto(s)
COVID-19 , Dietética , Dietética/educación , Humanos , Salud Mental , Pandemias , Proyectos Piloto
3.
J Nutr ; 145(4): 829-38, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25833786

RESUMEN

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.


Asunto(s)
Adipoquinas/metabolismo , Linfocitos T CD8-positivos/efectos de los fármacos , Ácidos Grasos Omega-3/administración & dosificación , Aceites de Pescado/administración & dosificación , Células 3T3-L1 , Adipocitos/efectos de los fármacos , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Animales , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular/efectos de los fármacos , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Quimiocina CCL3/genética , Quimiocina CCL3/metabolismo , Quimiocina CCL4/genética , Quimiocina CCL4/metabolismo , Quimiocina CCL7/genética , Quimiocina CCL7/metabolismo , Interleucina-6/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Obesidad/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Bazo/citología , Bazo/efectos de los fármacos , Bazo/metabolismo , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIA/metabolismo
4.
J Nutr ; 144(8): 1306-13, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24944284

RESUMEN

Cluster of differentiation 4(+) (CD4(+)) effector T-cell subsets [e.g., T-helper (Th) 1 and Th17] are implicated in autoimmune and inflammatory disorders such as multiple sclerosis, psoriasis, and rheumatoid arthritis. Interleukin (IL)-6 is a pleiotropic cytokine that induces Th17 polarization via signaling through the membrane-bound transducer glycoprotein 130 (GP130). Previously, we demonstrated that n-3 (ω-3) polyunsaturated fatty acids (PUFAs) reduce CD4(+) T-cell activation and differentiation into pathogenic Th17 cells by 25-30%. Here we report that n-3 PUFAs alter the response of CD4(+) T cells to IL-6 in a lipid raft membrane-dependent manner. Naive splenic CD4(+) T cells from fat-1 transgenic mice exhibited 30% lower surface expression of the IL-6 receptor. This membrane-bound receptor is known to be shed during cellular activation, but the release of soluble IL-6 receptor after treatment with anti-CD3 and anti-CD28 was not changed in the CD4(+) T cells from fat-1 mice, suggesting that the decrease in surface expression was not due to ectodomain release. We observed a significant 20% decrease in the association of GP130 with lipid rafts in activated fat-1 CD4(+) T cells and a 35% reduction in GP130 homodimerization, an obligate requirement for downstream signaling. The phosphorylation of signal transducer and activator of transcription 3 (STAT3), a downstream target of IL-6-dependent signaling, was also decreased by 30% in response to exogenous IL-6 in fat-1 CD4(+) T cells. Our results suggest that n-3 PUFAs suppress Th17 cell differentiation in part by reducing membrane raft-dependent responsiveness to IL-6, an essential polarizing cytokine.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Ácidos Grasos Omega-3/farmacología , Interleucina-6/metabolismo , Células Th17/efectos de los fármacos , Animales , Recuento de Células , Diferenciación Celular/efectos de los fármacos , Receptor gp130 de Citocinas/genética , Receptor gp130 de Citocinas/metabolismo , Femenino , Activación de Linfocitos/efectos de los fármacos , Masculino , Ratones , Fosforilación , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Interleucina-6/genética , Receptores de Interleucina-6/metabolismo , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Bazo/efectos de los fármacos , Bazo/metabolismo
5.
Br J Nutr ; 111(9): 1549-63, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24521520

RESUMEN

Common beans contain non-digestible fermentable components (SCFA precursors) and phenolic compounds (phenolic acids, flavonoids and anthocyanins) with demonstrated antioxidant and anti-inflammatory potential. The objective of the present study was to assess the in vivo effect of cooked whole-bean flours, with differing phenolic compound levels and profiles, in a mouse model of acute colitis. C57BL/6 mice were fed a 20 % navy bean or black bean flour-containing diet or an isoenergetic basal diet (BD) for 2 weeks before the induction of experimental colitis via 7 d dextran sodium sulphate (DSS, 2 % (w/v) in the drinking-water) exposure. Compared with the BD, both bean diets increased caecal SCFA and faecal phenolic compound concentrations (P< 0·05), which coincided with both beneficial and adverse effects on colonic and systemic inflammation. On the one hand, bean diets reduced mRNA expression of colonic inflammatory cytokines (IL-6, IL-9, IFN-γ and IL-17A) and increased anti-inflammatory IL-10 (P< 0·05), while systemically reduced circulating cytokines (IL-1ß, TNFα, IFNγ, and IL-17A, P< 0·05) and DSS-induced oxidative stress. On the other hand, bean diets enhanced DSS-induced colonic damage as indicated by an increased histological injury score and apoptosis (cleaved caspase-3 and FasL mRNA expression) (P< 0·05). In conclusion, bean-containing diets exerted both beneficial and adverse effects during experimental colitis by reducing inflammatory biomarkers both locally and systemically while aggravating colonic mucosal damage. Further research is required to understand the mechanisms through which beans exert their effects on colonic inflammation and the impact on colitis severity in human subjects.


Asunto(s)
Colitis/prevención & control , Colon/inmunología , Modelos Animales de Enfermedad , Alimentos Funcionales , Mucosa Intestinal/inmunología , Phaseolus , Semillas , Animales , Antioxidantes/análisis , Antioxidantes/uso terapéutico , Biomarcadores/análisis , Biomarcadores/sangre , Biomarcadores/metabolismo , Caspasa 3/metabolismo , Ciego/inmunología , Ciego/metabolismo , Ciego/patología , Colitis/inmunología , Colitis/metabolismo , Colitis/patología , Colon/metabolismo , Colon/patología , Culinaria , Citocinas/sangre , Citocinas/genética , Citocinas/metabolismo , Proteína Ligando Fas/biosíntesis , Proteína Ligando Fas/genética , Proteína Ligando Fas/metabolismo , Ácidos Grasos Volátiles/metabolismo , Heces/química , Alimentos Funcionales/efectos adversos , Alimentos Funcionales/análisis , Regulación de la Expresión Génica , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Estrés Oxidativo , Phaseolus/efectos adversos , Phaseolus/química , Semillas/efectos adversos , Semillas/química
6.
Mediators Inflamm ; 2014: 917149, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25136149

RESUMEN

During colitis, activation of two inflammatory T cell subsets, Th17 and Th1 cells, promotes ongoing intestinal inflammatory responses. n-6 polyunsaturated fatty acid- (PUFA-) derived eicosanoids, such as prostaglandin E2 (PGE2), promote Th17 cell-mediated inflammation, while n-3 PUFA antagonize both Th17 and Th1 cells and suppress PGE2 levels. We utilized two genetic mouse models, which differentially antagonize PGE2 levels, to examine the effect on Th17 cells and disease outcomes in trinitrobenzene sulfonic acid- (TNBS-) induced colitis. Fat-1 mice contain the ω3 desaturase gene from C. elegans and synthesize n-3 PUFA de novo, thereby reducing the biosynthesis of n-6 PUFA-derived eicosanoids. In contrast, Fads1 Null mice contain a disrupted Δ5 desaturase gene and produce lower levels of n-6 PUFA-derived eicosanoids. Compared to Wt littermates, Fat-1 and Fads1 Null mice exhibited a similar colitic phenotype characterized by reduced colonic mucosal inflammatory eicosanoid levels and mRNA expression of Th17 cell markers (IL-17A, RORγτ, and IL-23), decreased percentages of Th17 cells and, improved colon injury scores (P ≤ 0.05). Thus, during colitis, similar outcomes were obtained in two genetically distinct models, both of which antagonize PGE2 levels via different mechanisms. Our data highlight the critical impact of n-6 PUFA-derived eicosanoids in the promotion of Th17 cell-mediated colonic inflammation.


Asunto(s)
Ácido Araquidónico/química , Colitis/tratamiento farmacológico , Colitis/inmunología , Eicosanoides/química , Eicosanoides/uso terapéutico , Inflamación/inmunología , Inflamación/metabolismo , Células TH1/metabolismo , Células Th17/metabolismo , Animales , Colitis/metabolismo , Femenino , Masculino , Ratones
7.
Nutrients ; 16(4)2024 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-38398822

RESUMEN

The fermentation of non-digestible carbohydrates produces short-chain fatty acids (SCFAs), which have been shown to impact both skeletal muscle metabolic and inflammatory function; however, their effects within the obese skeletal muscle microenvironment are unknown. In this study, we developed a skeletal muscle in vitro model to mimic the critical features of the obese skeletal muscle microenvironment using L6 myotubes co-treated with 10 ng/mL lipopolysaccharide (LPS) and 500 µM palmitic acid (PA) for 24 h ± individual SCFAs, namely acetate, propionate and butyrate at 0.5 mM and 2.5 mM. At the lower SCFA concentration (0.5 mM), all three SCFA reduced the secreted protein level of RANTES, and only butyrate reduced IL-6 protein secretion and the intracellular protein levels of activated (i.e., ratio of phosphorylated-total) NFκB p65 and STAT3 (p < 0.05). Conversely, at the higher SCFA concentration (2.5 mM), individual SCFAs exerted different effects on inflammatory mediator secretion. Specifically, butyrate reduced IL-6, MCP-1 and RANTES secretion, propionate reduced IL-6 and RANTES, and acetate only reduced RANTES secretion (p < 0.05). All three SCFAs reduced intracellular protein levels of activated NFκB p65 and STAT3 (p < 0.05). Importantly, only the 2.5 mM SCFA concentration resulted in all three SCFAs increasing insulin-stimulated glucose uptake compared to control L6 myotube cultures (p < 0.05). Therefore, SCFAs exert differential effects on inflammatory mediator secretion in a cell culture model, recapitulating the obese skeletal muscle microenvironment; however, all three SCFAs exerted a beneficial metabolic effect only at a higher concentration via increasing insulin-stimulated glucose uptake, collectively exerting differing degrees of a beneficial effect on obesity-associated skeletal muscle dysfunction.


Asunto(s)
Interleucina-6 , Propionatos , Humanos , Propionatos/farmacología , Interleucina-6/metabolismo , Ácidos Grasos Volátiles/metabolismo , Obesidad , Butiratos , Acetatos , Fibras Musculares Esqueléticas/metabolismo , Insulina/farmacología , Músculo Esquelético/metabolismo , Glucosa/metabolismo , Técnicas de Cultivo de Célula , Mediadores de Inflamación
8.
Nutrients ; 16(5)2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-38474847

RESUMEN

Altered intestinal health is also associated with the incidence and severity of many chronic inflammatory conditions, which could be attenuated via dietary n-3 PUFA interventions. However, little is known about the effect of lifelong exposure to n-3 PUFA from plant and marine sources (beginning in utero via the maternal diet) on early life biomarkers of intestinal health. Harems of C57Bl/6 mice were randomly assigned to one of three isocaloric AIN-93G modified diets differing in their fat sources consisting of the following: (i) 10% safflower oil (SO, enriched in n-6 PUFA), (ii) 3% flaxseed oil + 7% safflower oil (FX, plant-based n-3 PUFA-enriched diet), or (iii) 3% menhaden fish oil + 7% safflower oil (MO, marine-based n-3 PUFA-enriched diet). Mothers remained on these diets throughout pregnancy and offspring (n = 14/diet) continued on the same parental diet until termination at 3 weeks of age. In ileum, villi:crypt length ratios were increased in both the FX and MO dietary groups compared to SO (p < 0.05). Ileum mRNA expression of critical intestinal health biomarkers was increased by both n-3 PUFA-enriched diets including Relmß and REG3γ compared to SO (p < 0.05), whereas only the FX diet increased mRNA expression of TFF3 and Muc2 (p < 0.05) and only the MO diet increased mRNA expression of ZO-1 (p < 0.05). In the proximal colon, both the FX and MO diets increased crypt lengths compared to SO (p < 0.05), whereas only the MO diet increased goblet cell numbers compared to SO (p < 0.05). Further, the MO diet increased proximal colon mRNA expression of Relmß and REG3γ (p < 0.05) and both MO and FX increased mRNA expression of Muc2 compared to SO (p < 0.05). Collectively, these results demonstrate that lifelong exposure to dietary n-3 PUFA, beginning in utero, from both plant and marine sources, can support intestinal health development in early life. The differential effects between plant and marine sources warrants further investigation for optimizing health.


Asunto(s)
Ácidos Grasos Omega-3 , Ratones , Animales , Embarazo , Femenino , Aceite de Cártamo , Aceites de Pescado , Dieta , Ratones Endogámicos C57BL , Biomarcadores , Expresión Génica , ARN Mensajero , Ácidos Grasos
9.
Am J Physiol Cell Physiol ; 304(9): C905-17, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23426968

RESUMEN

Epidermal growth factor receptor (EGFR)-mediated signaling is required for optimal intestinal wound healing. Since n-3 polyunsaturated fatty acids (PUFA), specifically docosahexaenoic acid (DHA), alter EGFR signaling and suppress downstream activation of key signaling pathways, we hypothesized that DHA would be detrimental to the process of intestinal wound healing. Using a mouse immortalized colonocyte model, DHA uniquely reduced EGFR ligand-induced receptor activation, whereas DHA and its metabolic precursor eicosapentaenoic acid (EPA) reduced wound-induced EGFR transactivation compared with control (no fatty acid or linoleic acid). Under wounding conditions, the suppression of EGFR activation was associated with a reduction in downstream activation of cytoskeletal remodeling proteins (PLCγ1, Rac1, and Cdc42). Subsequently, DHA and EPA reduced cell migration in response to wounding. Mice were fed a corn oil-, DHA-, or EPA-enriched diet prior to intestinal wounding (2.5% dextran sodium sulfate for 5 days followed by termination after 0, 3, or 6 days of recovery). Mortality was increased in EPA-fed mice and colonic histological injury scores were increased in EPA- and DHA-fed mice compared with corn oil-fed (control) mice. Although kinetics of colonic EGFR activation and downstream signaling (PLCγ1, Rac1, and Cdc42) were delayed by both n-3 PUFA, colonic repair was increased in EPA- relative to DHA-fed mice. These results indicate that, during the early response to intestinal wounding, DHA and EPA uniquely delay the activation of key wound-healing processes in the colon. This effect is mediated, at least in part, via suppression of EGFR-mediated signaling and downstream cytoskeletal remodeling.


Asunto(s)
Ácidos Docosahexaenoicos/administración & dosificación , Ácido Eicosapentaenoico/administración & dosificación , Receptores ErbB/metabolismo , Procesamiento Proteico-Postraduccional , Cicatrización de Heridas , Animales , Ácido Araquidónico/metabolismo , Movimiento Celular , Células Cultivadas , Colitis/inducido químicamente , Colitis/tratamiento farmacológico , Colitis/patología , Colon/efectos de los fármacos , Colon/patología , Aceite de Maíz/administración & dosificación , Sulfato de Dextran , Suplementos Dietéticos , Ácidos Docosahexaenoicos/fisiología , Ácido Eicosapentaenoico/fisiología , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Neuropéptidos/metabolismo , Consumo de Oxígeno , Fosforilación , Transducción de Señal , Ácido alfa-Linolénico/farmacología , Ácido alfa-Linolénico/fisiología , Proteína de Unión al GTP cdc42/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Proteína de Unión al GTP rac1
10.
J Nutr ; 143(9): 1501-8, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23864512

RESUMEN

Little is known about the impact of n3 (ω3) PUFAs on polarization of CD4(+) T cells into effector subsets other than Th1 and Th2. We assessed the effects of dietary fat [corn oil (CO) vs. fish oil (FO)] and fermentable fiber [cellulose (C) vs. pectin (P)] (2 × 2 design) in male C57BL/6 mice fed CO-C, CO-P, FO-C, or FO-P diets for 3 wk on the ex vivo polarization of purified splenic CD4(+) T cells (using magnetic microbeads) into regulatory T cells [Tregs; forkhead box P3 (Foxp3(+)) cells] or Th17 cells [interleukin (IL)-17A(+) and retinoic acid receptor-related orphan receptor (ROR) γτ(+) cells] by flow cytometry. Treg polarization was unaffected by diet; however, FO independently reduced the percentage of both CD4(+) IL-17A(+) (P < 0.05) and CD4(+) RORγτ(+) cells (P < 0.05). Moreover, expression of another critical Th17-cell-related transcription factor, signal transducer and activator of transcription 3, was reduced by FO. Dietary FO reduced the surface expression of both IL-6R and IL-23R on polarized Th17 cells (P ≤ 0.05), thus interfering with the promotive effects of these critical cytokines on Th17 polarization. Additionally, C57BL/6 mice fed diets enriched in eicosapentaenoic acid (EPA), docosahexaenoic acid (DHA), or DHA + EPA similarly reduced Th17-cell polarization in comparison to CO by reducing expression of the Th17-cell signature cytokine (IL-17A; P = 0.0015) and transcription factor (RORγτ P = 0.02), whereas Treg polarization was unaffected. Collectively, these data show that n3 PUFAs exert a direct effect on the development of Th17 cells in healthy mice, implicating a novel n3 PUFA-dependent, anti-inflammatory mechanism of action via the suppression of the initial development of this inflammatory T-cell subset.


Asunto(s)
Antiinflamatorios/farmacología , Ácidos Grasos Insaturados/farmacología , Linfocitos T Reguladores/efectos de los fármacos , Células Th17/efectos de los fármacos , Animales , Diferenciación Celular/efectos de los fármacos , Aceite de Maíz/administración & dosificación , Aceite de Maíz/farmacología , Aceites de Pescado/administración & dosificación , Aceites de Pescado/farmacología , Interleucina-17/inmunología , Activación de Linfocitos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Linfocitos T Reguladores/inmunología , Células Th17/inmunología
11.
Biochem J ; 443(1): 27-37, 2012 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-22250985

RESUMEN

n-3 PUFA (polyunsaturated fatty acids), i.e. DHA (docosahexaenoic acid), found in fish oil, exhibit anti-inflammatory properties; however, the molecular mechanisms remain unclear. Since PtdIns(4,5)P2 resides in raft domains and DHA can alter the size of rafts, we hypothesized that PtdIns(4,5)P2 and downstream actin remodelling are perturbed by the incorporation of n-3 PUFA into membranes, resulting in suppressed T-cell activation. CD4+ T-cells isolated from Fat-1 transgenic mice (membranes enriched in n-3 PUFA) exhibited a 50% decrease in PtdIns(4,5)P2. Upon activation by plate-bound anti-CD3/anti-CD28 or PMA/ionomycin, Fat-1 CD4+ T-cells failed to metabolize PtdIns(4,5)P2. Furthermore, actin remodelling failed to initiate in Fat-1 CD4+ T-cells upon stimulation; however, the defect was reversed by incubation with exogenous PtdIns(4,5)P2. When Fat-1 CD4+ T-cells were stimulated with anti-CD3/anti-CD28-coated beads, WASP (Wiskott-Aldrich syndrome protein) failed to translocate to the immunological synapse. The suppressive phenotype, consisting of defects in PtdIns(4,5)P2 metabolism and actin remodelling, were recapitulated in CD4+ T-cells isolated from mice fed on a 4% DHA triacylglycerol-enriched diet. Collectively, these data demonstrate that n-3 PUFA, such as DHA, alter PtdIns(4,5)P2 in CD4+ T-cells, thereby suppressing the recruitment of WASP to the immunological synapse, and impairing actin remodelling in CD4+ T-cells.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Antiinflamatorios/farmacología , Linfocitos T CD4-Positivos/inmunología , Ácidos Grasos Omega-3/farmacología , Activación de Linfocitos , Fosfatidilinositol 4,5-Difosfato/fisiología , Animales , Linfocitos T CD4-Positivos/metabolismo , Cadherinas/genética , Células Cultivadas , Sinapsis Inmunológicas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosfatidilinositol 4,5-Difosfato/farmacología , Transporte de Proteínas , Bazo/citología , Proteína del Síndrome de Wiskott-Aldrich/metabolismo
12.
Nutrients ; 15(23)2023 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-38068780

RESUMEN

The tolerance model rests on the thesis of a physiologically regulated, albeit unsustainable, systemic attempt to adapt to the catabolic challenge posed by acute prepubescent malnutrition even in its severe forms. The model centers on the immunological component of the attempt, positing reorientation toward a non-inflammatory form of competence in place of the classic paradigm of immunological attrition and exhaustion. The foundation of the model was laid in 1990, and sixteen years later it was articulated formally on the basis of a body of evidence centered on T cell cytokines and interventions with cytokine and hormonal mediators. The benefit originally suggested was a reduced risk of autoimmune pathologies consequent to the catabolic release of self-antigens, hence the designation highlighting immune tolerance. Herein, the emergence of the tolerance model is traced from its roots in the recognition that acute malnutrition elicits an endocrine-based systemic adaptive attempt. Thereafter, the growth of the evidence base supporting the model is outlined, and its potential to shed new light on existing information is tested by application to the findings of a published clinical study of acutely malnourished children. Finally, some knowledge gaps pertinent to the model are identified and its potential for growth consonant with evolving perceptions of immunobiology is illustrated.


Asunto(s)
Desnutrición , Humanos , Niño , Citocinas , Tolerancia Inmunológica , Linfocitos T
13.
J Lipid Res ; 53(7): 1287-95, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22534642

RESUMEN

Arachidonic acid (20:4(Δ5,8,11,14), AA)-derived eicosanoids regulate inflammation and promote cancer development. Previous studies have targeted prostaglandin enzymes in an attempt to modulate AA metabolism. However, due to safety concerns surrounding the use of pharmaceutical agents designed to target Ptgs2 (cyclooxygenase 2) and its downstream targets, it is important to identify new targets upstream of Ptgs2. Therefore, we determined the utility of antagonizing tissue AA levels as a novel approach to suppressing AA-derived eicosanoids. Systemic disruption of the Fads1 (Δ5 desaturase) gene reciprocally altered the levels of dihomo-γ-linolenic acid (20:3(Δ8,11,14), DGLA) and AA in mouse tissues, resulting in a profound increase in 1-series-derived and a concurrent decrease in 2-series-derived prostaglandins. The lack of AA-derived eicosanoids, e.g., PGE2 was associated with perturbed intestinal crypt proliferation, immune cell homeostasis, and a heightened sensitivity to acute inflammatory challenge. In addition, null mice failed to thrive, dying off by 12 weeks of age. Dietary supplementation with AA extended the longevity of null mice to levels comparable to wild-type mice. We propose that this new mouse model will expand our understanding of how AA and its metabolites mediate inflammation and promote malignant transformation, with the eventual goal of identifying new drug targets upstream of Ptgs2.


Asunto(s)
Modelos Animales de Enfermedad , Eicosanoides/metabolismo , Ácido Graso Desaturasas/metabolismo , Animales , Ácido Araquidónico/administración & dosificación , delta-5 Desaturasa de Ácido Graso , Suplementos Dietéticos , Eicosanoides/deficiencia , Ácido Graso Desaturasas/deficiencia , Ácido Graso Desaturasas/genética , Inflamación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
14.
Am J Physiol Gastrointest Liver Physiol ; 302(1): G153-67, 2012 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-21940900

RESUMEN

The ligand-activated transcription factor peroxisome proliferator-activated receptor (PPAR)-δ is highly expressed in colonic epithelial cells; however, the role of PPARδ ligands, such as fatty acids, in mucosal inflammation and malignant transformation has not been clarified. Recent evidence suggests that the anti-inflammatory/chemoprotective properties of fish oil (FO)-derived n-3 polyunsaturated fatty acids (PUFAs) may be partly mediated by PPARδ. Therefore, we assessed the role of PPARδ in modulating the effects of dietary n-3 PUFAs by targeted deletion of intestinal epithelial cell PPARδ (PPARδ(ΔIEpC)). Subsequently, we documented changes in colon tumorigenesis and the inflammatory microenvironment, i.e., local [mesenteric lymph node (MLN)] and systemic (spleen) T cell activation. Animals were fed chemopromotive [corn oil (CO)] or chemoprotective (FO) diets during the induction of chronic inflammation/carcinogenesis. Tumor incidence was similar in control and PPARδ(ΔIEpC) mice. FO reduced mucosal injury, tumor incidence, colonic STAT3 activation, and inflammatory cytokine gene expression, independent of PPARδ genotype. CD8(+) T cell recruitment into MLNs was suppressed in PPARδ(ΔIEpC) mice. Similarly, FO reduced CD8(+) T cell numbers in the MLN. Dietary FO independently modulated MLN CD4(+) T cell activation status by decreasing CD44 expression. CD11a expression by MLN CD4(+) T cells was downregulated in PPARδ(ΔIEpC) mice. Lastly, splenic CD62L expression was downregulated in PPARδ(ΔIEpC) CD4(+) and CD8(+) T cells. These data demonstrate that expression of intestinal epithelial cell PPARδ does not influence azoxymethane/dextran sodium sulfate-induced colon tumor incidence. Moreover, we provide new evidence that dietary n-3 PUFAs attenuate intestinal inflammation in an intestinal epithelial cell PPARδ-independent manner.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Transformación Celular Neoplásica/efectos de los fármacos , Colitis/tratamiento farmacológico , Neoplasias del Colon/tratamiento farmacológico , Grasas Insaturadas en la Dieta/farmacología , Aceites de Pescado/farmacología , Eliminación de Gen , PPAR delta/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/inmunología , Animales , Antígeno CD11a/biosíntesis , Antígeno CD11a/inmunología , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/inmunología , Enfermedad Crónica , Colitis/genética , Colitis/inmunología , Neoplasias del Colon/genética , Neoplasias del Colon/inmunología , Citocinas/biosíntesis , Grasas Insaturadas en la Dieta/inmunología , Grasas Insaturadas en la Dieta/metabolismo , Femenino , Aceites de Pescado/inmunología , Receptores de Hialuranos/biosíntesis , Receptores de Hialuranos/inmunología , Mucosa Intestinal/inmunología , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , PPAR delta/genética , PPAR delta/inmunología , Factor de Transcripción STAT3/biosíntesis , Bazo/efectos de los fármacos , Bazo/inmunología
15.
J Nutr ; 142(1): 117-24, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22131549

RESUMEN

During colon inflammation, Th17 cells and immunosuppressive regulatory T cells (Treg) are thought to play promotive and preventative roles, respectively. Dietary (n-3) PUFA favorably modulate intestinal inflammation in part by downregulating T-cell activation and functionality. We used the Fat-1 mouse, a genetic model that synthesizes long-chain (n-3) PUFA de novo, to test the hypothesis that (n-3) PUFA protect against colonic inflammation by modulating the polarization of Treg and Th17 cells during colitis. Male and female wild-type (WT) and Fat-1 mice were administered dextran sodium sulfate (DSS) in the drinking water (2.5%) to induce acute (5 d DSS) or chronic (3 cycles DSS) colitis and the percentage of Treg and Th17 cells residing locally [colonic lamina propria (cLP)] and systemically (spleen) was determined by flow cytometry. The percentage of Treg in either tissue site was unaffected by genotype (P > 0.05); however, during chronic colitis, the percentage of Th17 cells residing in both the spleen and cLP was lower in Fat-1 mice compared to WT mice (P < 0.05). Colonic mucosal mRNA expression of critical Th17 cell cytokines and chemokine receptors (IL-17F, IL-21, and CCR6) were lower, whereas expression of the Th17 cell suppressive cytokine, IL-27, was greater in Fat-1 mice compared to WT mice during chronic colitis (P < 0.05). Moreover, colon histological scores were improved in Fat-1 mice (P < 0.05). Collectively, these results demonstrate for the first time, to our knowledge, that (n-3) PUFA can modulate the colonic mucosal microenvironment to suppress Th17 cell accumulation and inflammatory damage following the induction of chronic colitis.


Asunto(s)
Colitis/inmunología , Ácidos Grasos Omega-3/administración & dosificación , Células Th17/inmunología , Animales , Enfermedad Crónica , Colitis/metabolismo , Colitis/patología , Citocinas/metabolismo , Femenino , Citometría de Flujo , Masculino , Ratones , Ratones Mutantes
16.
Br J Nutr ; 107(9): 1249-53, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-21944220

RESUMEN

The declining inflammatory immune competence of acute (i.e. wasting) pre-pubescent protein-energy malnutrition has been regarded as reflecting an unregulated immunological disintegration. Recent evidence, however, suggests that malnutrition stimulates a regulated immunological reconfiguration to achieve a non-inflammatory form of competence, perhaps offering protection against autoimmune reactions - the 'Tolerance Model'. Our objective was to determine the influence of acute pre-pubescent malnutrition on the expression of genes critical to tolerogenic regulation. Male and female C57BL/6J mice, initially 19 d old, consumed a complete purified diet either ad libitum (age-matched controls) or in restricted daily quantities (mimicking marasmus), or consumed an isoenergetic low-protein diet ad libitum (mimicking incipient kwashiorkor) for 14 d (six animals per dietary group). Gene expression in the spleen, typically an inflammatory organ, and in the small intestine, a site designed for non-inflammatory defence, was assessed by real-time quantitative RT-PCR, and normalised to ß-actin. In the spleen of the malnourished groups, both IL-10 and transforming growth factor-ß1 mRNA expression increased compared with controls (P < 0.05), whereas mRNA expression of IL-12p40 decreased (P < 0.05). Conversely, malnutrition exerted no influence on the expression of mRNA for these cytokines in the small intestine (P>0.05). Moreover, forkhead box P3 mRNA expression, indicative of cell-based tolerogenic potential, was sustained in both the spleen and intestine of the malnourished groups (P>0.05). Thus, despite limited supplies of energy and substrates, the spleen shifted towards a non-inflammatory character and the intestine was sustained in this mode in advanced pre-pubescent weight loss. These findings provide the first support for the Tolerance Model at the level of mRNA transcript expression.


Asunto(s)
Regulación de la Expresión Génica , Tolerancia Inmunológica , Kwashiorkor/inmunología , Desnutrición Proteico-Calórica/inmunología , ARN Mensajero/metabolismo , Actinas/metabolismo , Fenómenos Fisiológicos Nutricionales de los Animales , Animales , Peso Corporal , Modelos Animales de Enfermedad , Femenino , Sistema Inmunológico , Inflamación , Interleucina-10/metabolismo , Subunidad p40 de la Interleucina-12/metabolismo , Mucosa Intestinal/metabolismo , Intestino Delgado/metabolismo , Kwashiorkor/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Desnutrición Proteico-Calórica/fisiopatología , Reacción en Cadena en Tiempo Real de la Polimerasa , Bazo/metabolismo , Factores de Tiempo , Factor de Crecimiento Transformador beta/metabolismo
17.
Nutrients ; 14(14)2022 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-35889783

RESUMEN

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.


Asunto(s)
Lipopolisacáridos , Propionatos , Butiratos/metabolismo , Quimiocina CCL3 , Quimiocina CCL5 , Carbohidratos de la Dieta , Ácidos Grasos Volátiles/metabolismo , Ácidos Grasos Volátiles/farmacología , Interleucina-6 , Lipopolisacáridos/farmacología , Fibras Musculares Esqueléticas/metabolismo , Ácido Palmítico/farmacología , Propionatos/metabolismo
18.
Nutrition ; 91-92: 111388, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34298481

RESUMEN

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.


Asunto(s)
Ácidos Grasos Omega-3 , Inflamasomas , Adipocitos , Tejido Adiposo , Animales , Técnicas de Cocultivo , Dieta Alta en Grasa , Ácidos Grasos Omega-3/farmacología , Ratones , Ratones Endogámicos C57BL , Proteína con Dominio Pirina 3 de la Familia NLR , Obesidad/tratamiento farmacológico , Células TH1
19.
Nutrients ; 13(3)2021 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-33652785

RESUMEN

Cooked common beans (Phaseolus vulgaris) improve intestinal health in lean mice and attenuate intestinal dysbiosis and inflammation when consumed concurrent with obesity development. We determined the effects of a high-fat (HF) bean supplemented diet in mice with established obesity (induced by 12 weeks of HF diet (60% fat as kcal)) compared to obese mice consuming a HF or low-fat (LF) weight loss control diet. Obese C57BL/6 male mice remained consuming HF for eight weeks or were randomly switched from HF to an isocaloric HF with 15.7% cooked navy bean powder diet (HFàHFB) or LF (11% fat as kcal; HFàLF) (n = 12/group). HFàHFB improved the obese phenotype, including (i) fecal microbiome (increased Prevotella, Akkermansia muciniphila, and short-chain fatty acid levels), (ii) intestinal health (increased ZO-1, claudin-2, Muc2, Relmß, and Reg3γ expression), and (iii) reduced adipose tissue (AT) inflammatory proteins (NFκBp65, STAT3, IL-6, MCP-1, and MIP-1α), versus HF (p < 0.05). Conversely, HFàLF reduced body weight and circulating hormones (leptin, resistin, and PAI-1) versus HF and HFàHFB (p < 0.05); however, AT inflammation and intestinal health markers were not improved to the same degree as HFàHFB (p < 0.05). Despite remaining on a HF obesogenic diet, introducing beans in established obesity improved the obese phenotype (intestinal health and adipose inflammation) more substantially than weight loss alone.


Asunto(s)
Dieta Alta en Grasa/métodos , Dieta Reductora/métodos , Suplementos Dietéticos , Obesidad/dietoterapia , Phaseolus , Tejido Adiposo/metabolismo , Animales , Biomarcadores/metabolismo , Peso Corporal/efectos de los fármacos , Dieta Alta en Grasa/efectos adversos , Heces/microbiología , Microbioma Gastrointestinal , Inflamación , Mucosa Intestinal/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Obesidad/metabolismo , Fenotipo , Polvos , Índice de Severidad de la Enfermedad
20.
J Nutr Biochem ; 95: 108763, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33965532

RESUMEN

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.


Asunto(s)
Apoptosis/efectos de los fármacos , Aceites de Pescado/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Inflamación/tratamiento farmacológico , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Obesidad/inducido químicamente , Tejido Adiposo/efectos de los fármacos , Animales , Peso Corporal/efectos de los fármacos , Neoplasias de la Mama , Línea Celular Tumoral , Suplementos Dietéticos , Ácidos Grasos/química , Femenino , Aceites de Pescado/administración & dosificación , Humanos , Glándulas Mamarias Animales/química , Ratones , ARN Mensajero/genética , ARN Mensajero/metabolismo , Distribución Aleatoria , Receptor ErbB-2
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA