Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
Am J Physiol Gastrointest Liver Physiol ; 315(5): G788-G798, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30095298

RESUMO

Flaxseed is high in ω-3 polyunsaturated fatty acids, fiber, and lignans known to lower cholesterol levels. However, its use for prevention or treatment of inflammatory bowel diseases has yielded mixed results, perhaps related to dietary interactions. In this study, we evaluated the impact of ground flaxseed supplementation on the severity of Citrobacter rodentium-induced colitis in the setting of either a high-fat (HF, ~36%kcal) or reduced-fat (RF, ~12%kcal) diet. After weaning, C57BL/6 mice ( n = 8-15/treatment) were fed ground flaxseed (7 g/100 g diet) with either HF (HF Flx) or RF (RF Flx) diets for 4 wk before infection with C. rodentium or sham gavage. Weight changes, mucosal inflammation, pathogen burden, gut microbiota composition, tissue polyunsaturated fatty acids, and cecal short-chain fatty acids were compared over a 14-day infection period. The RF diet protected against C. rodentium-induced colitis, whereas the RF Flx diet increased pathogen burden, exacerbated gut inflammation, and promoted gut dysbiosis. When compared with the RF diet, both HF and HF Flx diets resulted in more severe pathology in response to C. rodentium infection. Our findings demonstrate that although an RF diet protected against C. rodentium-induced colitis and associated gut dysbiosis in mice, beneficial effects were diminished with ground flaxseed supplementation. NEW & NOTEWORTHY Our results demonstrate a strong protective effect of a reduced-fat diet against intestinal inflammation, dysbiosis, and pathogen burden during Citrobacter rodentium-induced colitis. However, ground flaxseed supplementation in the setting of a reduced-fat diet exacerbated colitis despite higher levels of intestinal n-3 polyunsaturated fatty acids and cecal short-chain fatty acids.


Assuntos
Colite Ulcerativa/dietoterapia , Dieta com Restrição de Gorduras , Infecções por Enterobacteriaceae/dietoterapia , Ácidos Graxos Insaturados/efeitos adversos , Linho/química , Animais , Citrobacter rodentium/efeitos dos fármacos , Colite Ulcerativa/microbiologia , Infecções por Enterobacteriaceae/microbiologia , Ácidos Graxos Insaturados/farmacologia , Microbioma Gastrointestinal/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL
2.
J Nutr ; 145(4): 829-38, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25833786

RESUMO

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.


Assuntos
Adipocinas/metabolismo , Linfócitos T CD8-Positivos/efeitos dos fármacos , Ácidos Graxos Ômega-3/administração & dosagem , Óleos de Peixe/administração & dosagem , Células 3T3-L1 , Adipócitos/efeitos dos fármacos , Tecido Adiposo/efeitos dos fármacos , Tecido Adiposo/metabolismo , Animais , Linfócitos T CD8-Positivos/metabolismo , Diferenciação Celular/efeitos dos 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/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Baço/citologia , Baço/efeitos dos fármacos , Baço/metabolismo , Fator de Transcrição RelA/genética , Fator de Transcrição RelA/metabolismo
3.
Am J Physiol Gastrointest Liver Physiol ; 306(12): G1042-55, 2014 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-24763556

RESUMO

Flaxseed (FS), a dietary oilseed, contains a variety of anti-inflammatory bioactives, including fermentable fiber, phenolic compounds (lignans), and the n-3 polyunsaturated fatty acid (PUFA) α-linolenic acid. The objective of this study was to determine the effects of FS and its n-3 PUFA-rich kernel or lignan- and soluble fiber-rich hull on colitis severity in a mouse model of acute colonic inflammation. C57BL/6 male mice were fed a basal diet (negative control) or a basal diet supplemented with 10% FS, 6% kernel, or 4% hull for 3 wk prior to and during colitis induction via 5 days of 2% (wt/vol) dextran sodium sulfate (DSS) in their drinking water (n = 12/group). An increase in anti-inflammatory metabolites (hepatic n-3 PUFAs, serum mammalian lignans, and cecal short-chain fatty acids) was associated with consumption of all FS-based diets, but not with anti-inflammatory effects in DSS-exposed mice. Dietary FS exacerbated DSS-induced acute colitis, as indicated by a heightened disease activity index and an increase in colonic injury and inflammatory biomarkers [histological damage, apoptosis, myeloperoxidase, inflammatory cytokines (IL-6 and IL-1ß), and NF-κB signaling-related genes (Nfkb1, Ccl5, Bcl2a1a, Egfr, Relb, Birc3, and Atf1)]. Additionally, the adverse effect of the FS diet was extended systemically, as serum cytokines (IL-6, IFNγ, and IL-1ß) and hepatic cholesterol levels were increased. The adverse effects of FS were not associated with alterations in fecal microbial load or systemic bacterial translocation (endotoxemia). Collectively, this study demonstrates that although consumption of a 10% FS diet enhanced the levels of n-3 PUFAs, short-chain polyunsaturated fatty acids, and lignans in mice, it exacerbated DSS-induced colonic injury and inflammation.


Assuntos
Colite/metabolismo , Colo/lesões , Linho/toxicidade , Mucosa Intestinal/metabolismo , Doença Aguda , Animais , Colite/induzido quimicamente , Colite/patologia , Colo/metabolismo , Colo/patologia , Sulfato de Dextrana , Suplementos Nutricionais/toxicidade , Modelos Animais de Doenças , Ácidos Graxos Ômega-3/metabolismo , Mucosa Intestinal/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL
4.
Br J Nutr ; 111(9): 1549-63, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24521520

RESUMO

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.


Assuntos
Colite/prevenção & controle , Colo/imunologia , Modelos Animais de Doenças , Alimento Funcional , Mucosa Intestinal/imunologia , Phaseolus , Sementes , Animais , Antioxidantes/análise , Antioxidantes/uso terapêutico , Biomarcadores/análise , Biomarcadores/sangue , Biomarcadores/metabolismo , Caspase 3/metabolismo , Ceco/imunologia , Ceco/metabolismo , Ceco/patologia , Colite/imunologia , Colite/metabolismo , Colite/patologia , Colo/metabolismo , Colo/patologia , Culinária , Citocinas/sangue , Citocinas/genética , Citocinas/metabolismo , Proteína Ligante Fas/biossíntese , Proteína Ligante Fas/genética , Proteína Ligante Fas/metabolismo , Ácidos Graxos Voláteis/metabolismo , Fezes/química , Alimento Funcional/efeitos adversos , Alimento Funcional/análise , Regulação da Expressão Gênica , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Estresse Oxidativo , Phaseolus/efeitos adversos , Phaseolus/química , Sementes/efeitos adversos , Sementes/química
5.
Compr Rev Food Sci Food Saf ; 13(4): 400-412, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33412703

RESUMO

The properties of cereals products, bread, pasta, muffins, cookies, cakes, and bars, enriched with flaxseed, were reviewed to highlight suitable processing conditions for the production of high-quality flaxseed-enriched products with the desired health attributes. The review highlights the contrasting effect of flaxseed enrichment on the mechanical and physical properties of cereal products according to product type, flaxseed enrichment level, and processing history. Flaxseed lipids remain stable for most processing and storage conditions, presumably due to the significant antioxidant properties of lignans, but information is lacking on the impact of home-handling, such as bread toasting, on lipid oxidation. Cereal products enriched with flaxseed generally exhibit similar or improved shelf life compared to equivalent products with no flaxseed enrichment, suggesting that flaxseed may limit starch retrogradation, maintain moisture content, and delay microbial growth. Sensory analysis shows lower organoleptic properties of most cereal products containing flaxseed, but similar consumer acceptance for cereal products without and with flaxseed enrichment up to 15% is reported in the literature. This review indicates the need to better understand the impact of flaxseed enrichment on product microstructure and to conduct an extensive assessment of the health effects of flaxseed-enriched products, since very few studies have focused on the quantification of the bioaccessibility, bioavailability, and activity of flaxseed bioactive compounds for a variety of processing conditions and product formulation.

6.
Obes Rev ; 24(9): e13598, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37395146

RESUMO

A dysbiotic intestinal microbiome has been linked to chronic diseases such as obesity, which may suggest that interventions that target the microbiome may be useful in treating obesity and its complications. Appetite dysregulation and chronic systemic low-grade inflammation, such as that observed in obesity, are possibly linked with the intestinal microbiome and are potential therapeutic targets for the treatment of obesity via the microbiome. Dietary pulses (e.g., common beans) are composed of nutrients and compounds that possess the potential to modulate the gut microbiota composition and function which can in turn improve appetite regulation and chronic inflammation in obesity. This narrative review summarizes the current state of knowledge regarding the connection between the gut microbiome and obesity, appetite regulation, and systemic and adipose tissue inflammation. More specifically, it highlights the efficacy of interventions employing dietary common beans as a means to improve gut microbiota composition and/or function, appetite regulation, and inflammation in both rodent obesity and in humans. Collectively, results presented and discussed herein provide insight on the gaps in knowledge necessary for a comprehensive understanding of the potential of beans as a treatment for obesity while highlighting what further research is required to gain this understanding.


Assuntos
Microbioma Gastrointestinal , Humanos , Regulação do Apetite , Apetite , Obesidade/etiologia , Inflamação/complicações
7.
J Gerontol A Biol Sci Med Sci ; 78(2): 195-205, 2023 02 24.
Artigo em Inglês | MEDLINE | ID: mdl-35977540

RESUMO

Interest in the gut-brain axis and its implications for neurodegenerative diseases, such as Alzheimer's disease and related dementias, is growing. Microbial imbalances in the gastrointestinal tract, which are associated with impaired cognition, may represent a therapeutic target for lowering dementia risk. Multicomponent lifestyle interventions are a promising dementia risk reduction strategy and most often include diet and exercise, behaviors that are also known to modulate the gut microbiome. A better understanding of the role of the gut microbiome in diet and exercise effects on cognition may help to optimize these lifestyle interventions. The purpose of this review is to summarize findings from diet and exercise interventions that have investigated cognitive changes via effects on the microbiome. We aim to discuss the underlying mechanisms, highlight current gaps in the field, and provide new research directions. There is evidence mainly from rodent studies supporting the notion that microbiota changes mediate the effects of diet and exercise on cognition, with potential mechanisms including end-product metabolites and regulation of local and systemic inflammation. The field lacks whole diet and exercise interventions, especially those involving human participants. It is further limited by heterogeneous rodent models, outcome assessments, and the absence of proper mediation analyses. Trials including older adults with dementia risk factors, factorial designs of diet and exercise, and pre and post measures of microbiota, end-product metabolites, and inflammation would help to elucidate and potentially leverage the role of the microbiome in lowering dementia risk through lifestyle modification.


Assuntos
Doença de Alzheimer , Microbioma Gastrointestinal , Humanos , Idoso , Microbioma Gastrointestinal/fisiologia , Dieta , Cognição/fisiologia , Doença de Alzheimer/prevenção & controle , Inflamação , Encéfalo
8.
Nutrients ; 15(16)2023 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-37630732

RESUMO

The microbiota gut-brain axis (mGBA) is an important contributor to mental health and neurological and mood disorders. Lipopolysaccharides (LPS) are endotoxins that are components of Gram-negative bacteria cell walls and have been widely shown to induce both systemic and neuro-inflammation. Flaxseed (Linum usitatissimum) is an oilseed rich in fibre, n3-poly-unsaturated fatty acid (alpha-linolenic acid (ALA)), and lignan, secoisolariciresinol diglucoside, which all can induce beneficial effects across varying aspects of the mGBA. The objective of this study was to determine the potential for dietary supplementation with flaxseed or flaxseed oil to attenuate LPS-induced inflammation through modulation of the mGBA. In this study, 72 5-week-old male C57Bl/6 mice were fed one of three isocaloric diets for 3 weeks: (1) AIN-93G basal diet (BD), (2) BD + 10% flaxseed (FS), or (3) BD + 4% FS oil (FO). Mice were then injected with LPS (1 mg/kg i.p) or saline (n = 12/group) and samples were collected 24 h post-injection. Dietary supplementation with FS, but not FO, partially attenuated LPS-induced systemic (serum TNF-α and IL-10) and neuro-inflammation (hippocampal and/or medial prefrontal cortex IL-10, TNF-α, IL-1ß mRNA expression), but had no effect on sickness and nest-building behaviours. FS-fed mice had enhanced fecal microbial diversity with increased relative abundance of beneficial microbial groups (i.e., Lachnospiraceae, Bifidobacterium, Coriobacteriaceae), reduced Akkermansia muciniphila, and increased production of short-chain fatty acids (SCFAs), which may play a role in its anti-inflammatory response. Overall, this study highlights the potential for flaxseed to attenuate LPS-induced inflammation, in part through modulation of the intestinal microbiota, an effect which may not be solely driven by its ALA-rich oil component.


Assuntos
Linho , Microbioma Gastrointestinal , Masculino , Animais , Camundongos , Óleo de Semente do Linho/farmacologia , Lipopolissacarídeos , Interleucina-10 , Eixo Encéfalo-Intestino , Fator de Necrose Tumoral alfa , Dieta
9.
Nutrition ; 91-92: 111388, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34298481

RESUMO

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.


Assuntos
Ácidos Graxos Ômega-3 , Inflamassomos , Adipócitos , Tecido Adiposo , Animais , Técnicas de Cocultura , Dieta Hiperlipídica , Ácidos Graxos Ômega-3/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Proteína 3 que Contém Domínio de Pirina da Família NLR , Obesidade/tratamento farmacológico , Células Th1
10.
Nutrients ; 13(3)2021 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-33652785

RESUMO

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.


Assuntos
Dieta Hiperlipídica/métodos , Dieta Redutora/métodos , Suplementos Nutricionais , Obesidade/dietoterapia , Phaseolus , Tecido Adiposo/metabolismo , Animais , Biomarcadores/metabolismo , Peso Corporal/efeitos dos fármacos , Dieta Hiperlipídica/efeitos adversos , Fezes/microbiologia , Microbioma Gastrointestinal , Inflamação , Mucosa Intestinal/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Obesidade/metabolismo , Fenótipo , Pós , Índice de Gravidade de Doença
11.
Data Brief ; 38: 107409, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34632012

RESUMO

Dietary flaxseed (FS) and its components including FS oil (FSO), secoisolariciresinol diglucoside (SDG) and fiber, are processed by the gut microbiota. These data are in support of the article entitled "Discriminatory and cooperative effects within the mouse gut microbiota in response to flaxseed and its oil and lignan components", Journal of Nutritional Biochemistry [1]. Here we describe data generated by 16S rRNA sequencing of DNA obtained from cecum contents and feces of C57BL/6 female mice fed either a basal diet (BD, AIN93G), or isocaloric diets containing 10% FS, or 10% FS-equivalent amounts of FSO or SDG for 21 days. These include bacterial community composition and inferred KEGG pathways; the raw data are publicly available at the NCBI SRA database (BioProject ID PRJNA683934). Furthermore, this work includes detailed experimentation procedures, total bacterial counts (qPCR) in the cecum content and feces, and correlation analysis between a selected bacterial genus, Bacteroides and a predicted metabolic pathway. FS is utilized worldwide, especially for the prevention and/or treatment of diseases including cardiovascular diseases, diabetes and cancer. These data will be valuable as a reference to study different FS cultivars and SDG- or FSO- enriched products on the gut microbiota, to study gut microbial responses to FS and its components in different mouse strains and mammalian hosts to elucidate individualized effects, and to understand the importance of the gut microbiota for FS benefits.

12.
J Nutr Biochem ; 98: 108818, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34271098

RESUMO

Gut microbial processing of dietary flaxseed (FS) contributes to its health benefits, but the relative effects of its bioactive components (lignans, omega-3 fatty acids, fiber) on the microbiota are unclear. We investigated the gut microbial compositional and functional responses to whole FS and its isolated components, FS oil (FSO) and secoisolariciresinol diglucoside (SDG) (precursor to microbial-derived enterolignans) to help understand their contribution to whole FS benefits. Cecum content and fecal samples were collected from C57BL/6 female mice fed a basal diet (AIN93G) or isocaloric diets containing 10% FS or 10% FS-equivalent amounts of FSO or SDG for 21 days. Cecal and fecal microbiota composition and predicted genomic functions, and their relationship with serum enterolignans were evaluated. Only FS modified the community structure. Shared- and diet-specific enriched taxa and functions were identified. Carbohydrate and protein processing functions were enriched in FS mice, and there was a positive correlation between select enriched taxa, encompassing fiber degraders and SDG metabolizers, and serum enterolignans. This was not observed in mice receiving isolated FSO and SDG, suggesting that FS fiber supports SDG microbial metabolism. In conclusion, the cooperative activities of a diverse microbiota are necessary to process FS components and, when administered at the amount present in FS, these components may act together to affect SDG-derived enterolignans production. This has implications for the use of FS, FSO and SDG in clinical practice.


Assuntos
Linho/química , Microbioma Gastrointestinal/efeitos dos fármacos , Lignanas/farmacologia , Óleo de Semente do Linho/farmacologia , Animais , Butileno Glicóis/farmacologia , Ceco/metabolismo , Ceco/microbiologia , Dieta/métodos , Fibras na Dieta/farmacologia , Ácidos Graxos Ômega-3/farmacologia , Fezes/microbiologia , Feminino , Glucosídeos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL
13.
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
14.
Cell Mol Life Sci ; 66(24): 3873-94, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19669093

RESUMO

Together with the estrogen receptor (ER) alpha, estrogen receptor beta (ER beta ) mediates many of the physiological effects of estrogens. As ER beta is crucially involved in a variety of important physiological processes, its activity should be tightly regulated. ER beta regulation is achieved by hormone binding as well as by posttranslational modifications of the receptor. Furthermore, ER beta expression levels are under circadian control and can be regulated by DNA methylation of the ER beta promoter region. There are also a number of factors that can interfere with ER beta activity, such as phytoestrogens, endocrine disruptive chemicals, and growth factors. In this article, we outline different mechanisms of ER beta regulation and how they are implicated in various diseases. We also discuss how these insights might help to specifically target ER beta in drug design.


Assuntos
Receptor beta de Estrogênio/genética , Receptor beta de Estrogênio/metabolismo , Neoplasias/patologia , Processamento Alternativo , Metilação de DNA , Regulação Neoplásica da Expressão Gênica , Humanos , Modelos Biológicos , Neoplasias/genética , Neoplasias/metabolismo , Fitoestrógenos/metabolismo , Ligação Proteica , Processamento de Proteína Pós-Traducional
15.
Adv Food Nutr Res ; 90: 135-181, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31445595

RESUMO

Commercial trends based of the emergence of plant-based functional foods lead to investigate the structure-function relationship of their main bioactive constituents and their interactions in the food matrix and throughout the gastro-intestinal tract. Among these bioactive constituents, dietary polysaccharides and polyphenols have shown to interact at the molecular level and these interactions may have consequences on the polysaccharides physical and nutritional properties. The methods of investigation and mechanisms of interactions between polysaccharides and polyphenols are reviewed in light of their respective technological and nutritional functionalities. Finally, the potential impact of the co-occurrence or co-ingestion of polyphenols and polysaccharides on the technological and nutritional functionality of the polysaccharides are investigated.


Assuntos
Alimento Funcional/análise , Fenóis/química , Polissacarídeos/química , Polissacarídeos/fisiologia , Digestão , Fermentação , Tecnologia de Alimentos , Microbioma Gastrointestinal/fisiologia , Nível de Saúde , Humanos , Valor Nutritivo , Fenóis/análise , Polifenóis/análise , Polifenóis/química , Polissacarídeos/análise , Prebióticos
16.
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
17.
Nutrients ; 11(8)2019 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-31405019

RESUMO

Dietary pulses, including lentils, are protein-rich plant foods that are enriched in intestinal health-promoting bioactives, such as non-digestible carbohydrates and phenolic compounds. The aim of this study was to investigate the effect of diets supplemented with cooked red lentils on the colonic microenvironment (microbiota composition and activity and epithelial barrier integrity and function). C57Bl/6 male mice were fed one of five diets: a control basal diet (BD), a BD-supplemented diet with 5, 10 or 20% cooked red lentils (by weight), or a BD-supplemented diet with 0.7% pectin (equivalent soluble fiber level as found in the 20% lentil diet). Red lentil supplementation resulted in increased: (1) fecal microbiota α-diversity; (2) abundance of short-chain fatty acid (SCFA)-producing bacteria (e.g., Prevotella, Roseburia and Dorea spp.); (3) concentrations of fecal SCFAs; (4) mRNA expression of SCFA receptors (G-protein-coupled receptors (GPR 41 and 43) and tight/adherens junction proteins (Zona Occulden-1 (ZO-1), Claudin-2, E-cadherin). Overall, 20% lentil had the greatest impact on colon health outcomes, which were in part explained by a change in the soluble and insoluble fiber profile of the diet. These results support recent public health recommendations to increase consumption of plant-based protein foods for improved health, in particular intestinal health.


Assuntos
Bactérias/metabolismo , Colo/microbiologia , Culinária , Fibras na Dieta/metabolismo , Microbioma Gastrointestinal , Lens (Planta)/metabolismo , Sementes/metabolismo , Animais , Bactérias/genética , Caderinas/genética , Caderinas/metabolismo , Colo/metabolismo , Dieta , Fibras na Dieta/administração & dosagem , Ácidos Graxos/metabolismo , Fezes/microbiologia , Temperatura Alta , Masculino , Camundongos Endogâmicos C57BL , Mucinas/genética , Mucinas/metabolismo , Valor Nutritivo , Permeabilidade , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Proteínas de Junções Íntimas/genética , Proteínas de Junções Íntimas/metabolismo
18.
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
19.
Menopause ; 15(1): 171-9, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-17545920

RESUMO

OBJECTIVE: Flaxseed, the richest source of mammalian lignan precursors, enhances the tumor growth-inhibitory effect of tamoxifen while exerting no adverse effects on other estrogen-responsive tissues such as bone. Ingestion of sesame seed produces mammalian lignans comparable with flaxseed, but its anticancer potential is unknown. This study determined the interactive effects of sesame seed and tamoxifen on established MCF-7 tumor growth and bone health in ovariectomized athymic mice simulating a postmenopausal condition. DESIGN: Mice with established MCF-7 tumors were treated for 8 weeks with (1) basal diet (negative control), (2) 10% sesame seed, (3) basal diet + tamoxifen implant, (4) 10% sesame seed + tamoxifen implant, or (5) basal diet + estrogen implant (positive control). Weekly palpable tumor size, final tumor weight, cell proliferation, and apoptosis were measured. Bone mineral content, bone mineral density, and biomechanical strength testing were performed on the femur and lumbar vertebrae. RESULTS: Sesame seed induced regression of established tumor size similar to the negative control but tended to negate the tumor-inhibitory effect of tamoxifen, in part by reducing apoptosis. Sesame seed combined with tamoxifen induced higher bone mineral content, bone mineral density, and biomechanical strength in the femur and lumbar vertebrae than either treatment alone. A significant positive relationship was found between final tumor weight and bone strength parameters. CONCLUSIONS: Sesame seed is not protective and negatively interferes with tamoxifen in inducing regression of established MCF-7 tumor size but beneficially interacts with tamoxifen on bone in ovariectomized athymic mice.


Assuntos
Conservadores da Densidade Óssea/farmacologia , Densidade Óssea/efeitos dos fármacos , Neoplasias da Mama/tratamento farmacológico , Interações Alimento-Droga , Lignanas/farmacologia , Sesamum , Tamoxifeno/antagonistas & inibidores , Animais , Antineoplásicos Hormonais/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Terapia Combinada , Camundongos , Camundongos Nus , Ovariectomia , Pré-Menopausa , Sementes , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
20.
J Steroid Biochem Mol Biol ; 112(1-3): 13-9, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18703142

RESUMO

Previously we have shown that MCF-7 human breast tumor growth is stimulated after prolonged treatment with dietary soy protein isolate (SPI). However, the effects are attenuated when SPI is combined with flaxseed (FS). This study determined the changes that occur in tumor growth biomarkers, after both short- and long-term treatment with SPI, FS or their combination, to help identify signaling pathways potentially involved in SPI-stimulated tumor growth. Ovariectomized mice with established MCF-7 tumors were fed basal diet (control), 20%SPI, 10%FS, or SPI+FS for 2 or 25 weeks. After 2 weeks, there were no differences in tumor size, however, compared with control, SPI-treated tumors had higher IGF-IR and cyclin D1 while FS and SPI+FS-fed mice had lower pMAPK expression. After 25 weeks, SPI-treated tumors were larger, had higher proliferation, ERalpha, cyclin D1, IGF-IR, and pMAPK and lower ERbeta and HER2 levels. When combined with FS, however, the effects on these tumor biomarkers induced by SPI were attenuated. This study demonstrates that SPI and FS differently modulate tumor biomarkers of estrogen and growth factor signaling pathways, after both short- and long-term treatment, which may indicate a role of these pathways in the tumor stimulatory effects of SPI and the tumor inhibitory effects of FS.


Assuntos
Biomarcadores Tumorais/metabolismo , Linho/química , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Neoplasias Experimentais/metabolismo , Preparações de Plantas/farmacologia , Receptores de Estrogênio/metabolismo , Proteínas de Soja/efeitos adversos , Animais , Linhagem Celular Tumoral , Proliferação de Células , Ciclina D1/metabolismo , Feminino , Humanos , Sistema de Sinalização das MAP Quinases/fisiologia , Camundongos , Camundongos Nus , Transplante de Neoplasias , Neoplasias Experimentais/induzido quimicamente , Neoplasias Experimentais/patologia , Ovariectomia , Preparações de Plantas/efeitos adversos , Receptor IGF Tipo 1/metabolismo , Transdução de Sinais , Proteínas de Soja/administração & dosagem , Transplante Heterólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA