Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 43
1.
Nutrients ; 15(16)2023 Aug 11.
Article En | MEDLINE | ID: mdl-37630732

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.


Flax , Gastrointestinal Microbiome , Male , Animals , Mice , Linseed Oil/pharmacology , Lipopolysaccharides , Interleukin-10 , Brain-Gut Axis , Tumor Necrosis Factor-alpha , Diet
2.
Obes Rev ; 24(9): e13598, 2023 09.
Article En | MEDLINE | ID: mdl-37395146

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.


Gastrointestinal Microbiome , Humans , Appetite Regulation , Appetite , Obesity/etiology , Inflammation/complications
3.
J Gerontol A Biol Sci Med Sci ; 78(2): 195-205, 2023 02 24.
Article En | MEDLINE | ID: mdl-35977540

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.


Alzheimer Disease , Gastrointestinal Microbiome , Humans , Aged , Gastrointestinal Microbiome/physiology , Diet , Cognition/physiology , Alzheimer Disease/prevention & control , Inflammation , Brain
4.
Data Brief ; 38: 107409, 2021 Oct.
Article En | MEDLINE | ID: mdl-34632012

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.

5.
J Nutr Biochem ; 98: 108818, 2021 12.
Article En | MEDLINE | ID: mdl-34271098

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.


Flax/chemistry , Gastrointestinal Microbiome/drug effects , Lignans/pharmacology , Linseed Oil/pharmacology , Animals , Butylene Glycols/pharmacology , Cecum/metabolism , Cecum/microbiology , Diet/methods , Dietary Fiber/pharmacology , Fatty Acids, Omega-3/pharmacology , Feces/microbiology , Female , Glucosides/pharmacology , Mice , Mice, Inbred C57BL
6.
Nutrition ; 91-92: 111388, 2021.
Article En | MEDLINE | ID: mdl-34298481

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.


Fatty Acids, Omega-3 , Inflammasomes , Adipocytes , Adipose Tissue , Animals , Coculture Techniques , Diet, High-Fat , Fatty Acids, Omega-3/pharmacology , Mice , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein , Obesity/drug therapy , Th1 Cells
7.
J Nutr Biochem ; 95: 108763, 2021 09.
Article En | MEDLINE | ID: mdl-33965532

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.


Apoptosis/drug effects , Fish Oils/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Inflammation/drug therapy , Mammary Neoplasms, Experimental/drug therapy , Obesity/chemically induced , Adipose Tissue/drug effects , Animals , Body Weight/drug effects , Breast Neoplasms , Cell Line, Tumor , Dietary Supplements , Fatty Acids/chemistry , Female , Fish Oils/administration & dosage , Humans , Mammary Glands, Animal/chemistry , Mice , RNA, Messenger/genetics , RNA, Messenger/metabolism , Random Allocation , Receptor, ErbB-2
8.
Nutrients ; 13(3)2021 Feb 26.
Article En | MEDLINE | ID: mdl-33652785

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.


Diet, High-Fat/methods , Diet, Reducing/methods , Dietary Supplements , Obesity/diet therapy , Phaseolus , Adipose Tissue/metabolism , Animals , Biomarkers/metabolism , Body Weight/drug effects , Diet, High-Fat/adverse effects , Feces/microbiology , Gastrointestinal Microbiome , Inflammation , Intestinal Mucosa/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/metabolism , Phenotype , Powders , Severity of Illness Index
9.
J Nutr Biochem ; 72: 108216, 2019 10.
Article En | MEDLINE | ID: mdl-31476608

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.


Diet, High-Fat/adverse effects , Fish Oils/pharmacology , Gastrointestinal Microbiome/drug effects , Intestines/drug effects , Obesity/diet therapy , Adipokines/blood , Animals , Body Weight/drug effects , Colon/drug effects , Colon/physiology , Dietary Supplements , Eating/drug effects , Fatty Acids, Omega-3/metabolism , Feces/microbiology , Gastrointestinal Microbiome/genetics , Glucose Tolerance Test , Ileum/drug effects , Ileum/physiology , Intestines/physiology , Intra-Abdominal Fat/drug effects , Intra-Abdominal Fat/pathology , Male , Mice, Inbred C57BL , Obesity/etiology , Panniculitis/etiology , Panniculitis/prevention & control
10.
Adv Food Nutr Res ; 90: 135-181, 2019.
Article En | MEDLINE | ID: mdl-31445595

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.


Functional Food/analysis , Phenols/chemistry , Polysaccharides/chemistry , Polysaccharides/physiology , Digestion , Fermentation , Food Technology , Gastrointestinal Microbiome/physiology , Health Status , Humans , Nutritive Value , Phenols/analysis , Polyphenols/analysis , Polyphenols/chemistry , Polysaccharides/analysis , Prebiotics
11.
Nutrients ; 11(8)2019 Aug 09.
Article En | MEDLINE | ID: mdl-31405019

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.


Bacteria/metabolism , Colon/microbiology , Cooking , Dietary Fiber/metabolism , Gastrointestinal Microbiome , Lens Plant/metabolism , Seeds/metabolism , Animals , Bacteria/genetics , Cadherins/genetics , Cadherins/metabolism , Colon/metabolism , Diet , Dietary Fiber/administration & dosage , Fatty Acids/metabolism , Feces/microbiology , Hot Temperature , Male , Mice, Inbred C57BL , Mucins/genetics , Mucins/metabolism , Nutritive Value , Permeability , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Tight Junction Proteins/genetics , Tight Junction Proteins/metabolism
12.
J Nutr Biochem ; 70: 91-104, 2019 08.
Article En | MEDLINE | ID: mdl-31195365

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.


Diet, High-Fat , Inflammation/physiopathology , Intestinal Mucosa/physiopathology , Phaseolus , Adipokines/metabolism , Adipose Tissue/metabolism , Akkermansia , Animal Feed , Animals , Body Weight , Carbohydrate Metabolism , Dietary Fiber , Dietary Supplements , Epithelial Cells/metabolism , Epithelial Cells/pathology , Feces , Fermentation , Fluorescein-5-isothiocyanate , Gastrointestinal Microbiome , Intestinal Mucosa/metabolism , Male , Mice , Mice, Inbred C57BL , Obesity/metabolism , Permeability , Phenotype , Prevotella , RNA, Ribosomal, 16S/metabolism , Verrucomicrobia
13.
Am J Physiol Gastrointest Liver Physiol ; 315(5): G788-G798, 2018 11 01.
Article En | MEDLINE | ID: mdl-30095298

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.


Colitis, Ulcerative/diet therapy , Diet, Fat-Restricted , Enterobacteriaceae Infections/diet therapy , Fatty Acids, Unsaturated/adverse effects , Flax/chemistry , Animals , Citrobacter rodentium/drug effects , Colitis, Ulcerative/microbiology , Enterobacteriaceae Infections/microbiology , Fatty Acids, Unsaturated/pharmacology , Gastrointestinal Microbiome/drug effects , Male , Mice , Mice, Inbred C57BL
14.
J Nutr Biochem ; 56: 215-223, 2018 06.
Article En | MEDLINE | ID: mdl-29631142

The enriched levels of nondigestible fermentable carbohydrates and phenolic compounds found in common beans can exert immunomodulatory effects within the colon that improve gut health and mitigate the severity of colitis-associated inflammatory pathology. Prior to acute colitis onset, C57Bl/6 mice were prefed isocaloric 20% cooked navy bean (NB) or black bean (BB) diets for 3 weeks and switched to control basal diet (BD) 24 h prior to colitis induction via 5-day exposure to dextran sodium sulfate (2% w/v in drinking water)+3 days of fresh water. The severity of the acute colitis phenotype was attenuated by bean prefeeding, evidenced by reduced colon tissue inflammatory transcription factor activation (NFκB, STAT3) and inflammatory mediator levels in the colon (IL-1ß, IL-6, IL-18 and MCP-1) and serum (TNFα, IL-6, IL-1ß, MCP-1) versus BD (P≤.05). Additionally, biomarkers of enhanced wound repair responses were increased by bean prefeeding including colon tissue protein levels of IL-22, IL-27 and activated (i.e., GTP-bound) Cdc42 and Rac1 versus BD (P≤.05). mRNA expressions of genes involved in normal colonic epithelial function and the promotion of epithelial barrier integrity, defense and/or restitution and wound closure including MUC1, RELMß, IgA and REG3γ were all increased in NB and BB prefed mice versus BD (P≤.05). Collectively, bean supplementation prior to colitis induction (i.e., mimicking disease relapse) primes the colonic microenvironment to attenuate the severity of the colitis inflammatory phenotype and maintain aspects of epithelial barrier function.


Colitis/diet therapy , Colonic Diseases/diet therapy , Dietary Supplements , Epithelium/metabolism , Inflammation/diet therapy , Phaseolus , Animals , Biomarkers/metabolism , Colitis/pathology , Colon/pathology , Colonic Diseases/pathology , Cytokines/metabolism , Dextran Sulfate , Diet , Disease Models, Animal , Fermentation , Intestinal Mucosa/metabolism , Male , Mice , Mice, Inbred C57BL , Phenotype
15.
Appl Physiol Nutr Metab ; 43(9): 893-901, 2018 Sep.
Article En | MEDLINE | ID: mdl-29522694

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.


Cicer , Colitis/diet therapy , Inflammation/diet therapy , Animals , Biomarkers/metabolism , Colitis/chemically induced , Dextran Sulfate , Diet , Disease Models, Animal , Flour , Inflammation/chemically induced , Interleukin-18/metabolism , Interleukins/metabolism , Intestinal Mucosa/metabolism , Male , Mice, Inbred C57BL , NF-kappa B/metabolism , Tumor Necrosis Factor-alpha/metabolism , Interleukin-22
16.
Nutrients ; 9(12)2017 Nov 27.
Article En | MEDLINE | ID: mdl-29186929

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


Adipose Tissue/drug effects , Fatty Acids, Omega-3/pharmacology , Immunomodulation , Obesity/drug therapy , Adipokines/blood , Adipose Tissue/metabolism , Adiposity/drug effects , Animals , Disease Models, Animal , Docosahexaenoic Acids/pharmacology , Eicosapentaenoic Acid/pharmacology , Humans , Immunity, Cellular , Inflammation/blood , Inflammation/drug therapy , Obesity/blood , Signal Transduction
17.
J Nutr Biochem ; 49: 89-100, 2017 11.
Article En | MEDLINE | ID: mdl-28915390

Common beans (Phaseolus vulgaris L.) are enriched in non-digestible fermentable carbohydrates and phenolic compounds that can modulate the colonic microenvironment (microbiota and host epithelial barrier) to improve gut health. In a comprehensive assessment of the impact of two commonly consumed bean varieties (differing in levels and types of phenolic compounds) within the colonic microenvironment, C57Bl/6 mice were fed diets supplemented with 20% cooked navy bean (NB) or black bean (BB) flours or an isocaloric basal diet control (BD) for 3 weeks. NB and BB similarly altered the fecal microbiota community structure (16S rRNA sequencing) notably by increasing the abundance of carbohydrate fermenting bacteria such as Prevotella, S24-7 and Ruminococcus flavefaciens, which coincided with enhanced short chain fatty acid (SCFA) production (microbial-derived carbohydrate fermentation products) and colonic expression of the SCFA receptors GPR-41/-43/-109a. Both NB and BB enhanced multiple aspects of mucus and epithelial barrier integrity vs. BD including: (i) goblet cell number, crypt mucus content and mucin mRNA expression, (ii) anti-microbial defenses (Reg3γ), (iii) crypt length and epithelial cell proliferation, (iv) apical junctional complex components (occludin, JAM-A, ZO-1 and E-cadherin) mRNA expression and (v) reduced serum endotoxin concentrations. Interestingly, biomarkers of colon barrier integrity (crypt height, mucus content, cell proliferation and goblet cell number) were enhanced in BB vs. NB-fed mice, suggesting added benefits attributable to unique BB components (e.g., phenolics). Overall, NB and BB improved baseline colonic microenvironment function by altering the microbial community structure and activity and promoting colon barrier integrity and function; effects which may prove beneficial in attenuating gut-associated diseases.


Colon/metabolism , Dysbiosis/prevention & control , Functional Food , Gastrointestinal Microbiome/physiology , Intestinal Mucosa/metabolism , Phytohemagglutinins , Seeds , Animals , Biomarkers/metabolism , Cell Proliferation , Cellular Microenvironment , Colon/cytology , Colon/microbiology , Colon/pathology , Dietary Carbohydrates/administration & dosage , Dietary Carbohydrates/metabolism , Dietary Fiber/administration & dosage , Dietary Fiber/metabolism , Dysbiosis/metabolism , Dysbiosis/microbiology , Dysbiosis/pathology , Feces/microbiology , Fermentation , Gene Expression Regulation, Developmental , Intestinal Mucosa/cytology , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Male , Mice, Inbred C57BL , Molecular Typing , Prevotella/classification , Prevotella/growth & development , Prevotella/isolation & purification , Random Allocation , Ruminococcus/chemistry , Ruminococcus/growth & development , Ruminococcus/isolation & purification
18.
Mol Nutr Food Res ; 60(11): 2396-2412, 2016 11.
Article En | MEDLINE | ID: mdl-27349947

SCOPE: This study investigated the effects of cooked whole asparagus (ASP) versus its equivalent level of purified flavonoid glycoside, rutin (RUT), on dextran sodium sulfate (DSS)-induced colitis and subsequent colitis recovery in mice. METHODS AND RESULTS: C57BL/6 male mice were fed an AIN-93G basal diet (BD), or BD supplemented with 2% cooked ASP or 0.025% RUT for 2 wks prior to and during colitis induction with 2% DSS in water for 7 days, followed by 5 days colitis recovery. In colitic mice, both ASP and RUT upregulated mediators of improved barrier integrity and enhanced mucosal injury repair (e.g. Muc1, IL-22, Rho-A, Rac1, and Reg3γ), increased the proportion of mouse survival, and improved disease activity index. RUT had the greatest effect in attenuating DSS-induced colonic damage indicated by increased crypt and goblet cell restitution, reduced colonic myeloperoxidase, as well as attenuated DSS-induced microbial dysbiosis (reduced Enterobacteriaceae and Bacteroides, and increased unassigned Clostridales, Oscillospira, Lactobacillus, and Bifidobacterium). CONCLUSION: These findings demonstrate that dietary cooked ASP and its flavonoid glycoside, RUT, may be useful in attenuating colitis severity by modulating the colonic microenvironment resulting in reduced colonic inflammation, promotion of colonic mucosal injury repair, and attenuation of colitis-associated microbial dysbiosis.


Colitis/chemically induced , Dextran Sulfate/adverse effects , Rutin/pharmacology , Sulfates/pharmacology , Animals , Colon/metabolism , Cytokines/metabolism , Dietary Supplements , Disease Models, Animal , Inflammation , Interleukins/metabolism , Intestinal Mucosa/drug effects , Male , Mice , Mice, Inbred C57BL , Rutin/analysis , Interleukin-22
19.
J Nutr Biochem ; 32: 29-38, 2016 06.
Article En | MEDLINE | ID: mdl-27142734

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


Dietary Supplements , Fatty Acid Desaturases/metabolism , Fatty Acids, Omega-3/administration & dosage , Fatty Acids, Omega-6/administration & dosage , Immunomodulation , Leukocytes, Mononuclear/immunology , Spleen/immunology , Animals , Cells, Cultured , Crosses, Genetic , Cytokines/metabolism , Fatty Acid Desaturases/genetics , Fatty Acids, Omega-3/metabolism , Fatty Acids, Omega-6/metabolism , Female , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Lipopolysaccharides/toxicity , Male , Mice , Mice, Knockout , Spleen/cytology , Spleen/drug effects , Spleen/metabolism
20.
J Nutr Biochem ; 34: 61-72, 2016 08.
Article En | MEDLINE | ID: mdl-27208584

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.


Adipocytes, White/metabolism , Dietary Supplements , Fatty Acids, Omega-3/therapeutic use , Inflammasomes/metabolism , Macrophages/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Obesity/diet therapy , 3T3-L1 Cells , Adipocytes, White/immunology , Adipocytes, White/pathology , Animals , Anti-Inflammatory Agents, Non-Steroidal/analysis , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , CD11b Antigen/metabolism , Cell Communication , Cells, Cultured , Coculture Techniques , Diet, High-Fat/adverse effects , Dietary Supplements/analysis , Fatty Acids, Omega-3/analysis , Fatty Acids, Omega-3/metabolism , Female , Fish Oils/chemistry , Fish Oils/therapeutic use , Gene Expression Regulation , Inflammasomes/immunology , Macrophages/immunology , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Obesity/immunology , Obesity/metabolism , Obesity/pathology , Spleen/immunology , Spleen/metabolism , Spleen/pathology
...