1
|
Falsetti I, Palmini G, Iantomasi T, Brandi ML, Tonelli F. Mechanisms of Action of Phytoestrogens and Their Role in Familial Adenomatous Polyposis. Pharmaceutics 2024; 16:640. [PMID: 38794302 PMCID: PMC11125335 DOI: 10.3390/pharmaceutics16050640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 04/30/2024] [Accepted: 05/02/2024] [Indexed: 05/26/2024] Open
Abstract
Familial adenomatous polyposis (FAP) is a rare disease characterized by the development of adenomatous polyps in the colon and rectum already in adolescence. If left untreated, patients develop colorectal cancer (CRC) with a 100% probability. To date, the gold standard of FAP management is surgery, which is associated with morbidity and mortality. A chemopreventive agent capable of delaying, preventing and reversing the development of CRC has been sought. Several classes of drugs have been used but to date no chemopreventive drug has been found for the management of this disease. In recent years, the importance of estrogen receptors in FAP and CRC, particularly the β subtype, has emerged. Indeed, the expression of the latter is strongly reduced in adenomatous polyps and CRC and is inversely correlated with the aggressiveness of the disease. Since phytoestrogens have a high affinity for this receptor, they have been suggested for use as chemopreventive agents in FAP and CRC. A combination of phytoestrogens and insoluble fibres has proved particularly effective. In this review, the various mechanisms of action of phytoestrogens were analyzed and the effectiveness of using phytoestrogens as an effective chemopreventive strategy was discussed.
Collapse
Affiliation(s)
- Irene Falsetti
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50139 Florence, Italy; (I.F.); (T.I.)
| | - Gaia Palmini
- Fondazione Italiana Ricerca sulle Malattie dell’Osso (FIRMO Onlus), 50129 Florence, Italy; (G.P.); (M.L.B.)
| | - Teresa Iantomasi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50139 Florence, Italy; (I.F.); (T.I.)
| | - Maria Luisa Brandi
- Fondazione Italiana Ricerca sulle Malattie dell’Osso (FIRMO Onlus), 50129 Florence, Italy; (G.P.); (M.L.B.)
| | - Francesco Tonelli
- Fondazione Italiana Ricerca sulle Malattie dell’Osso (FIRMO Onlus), 50129 Florence, Italy; (G.P.); (M.L.B.)
| |
Collapse
|
2
|
Wang T, Brown NM, McCoy AN, Sandler RS, Keku TO. Omega-3 Polyunsaturated Fatty Acids, Gut Microbiota, Microbial Metabolites, and Risk of Colorectal Adenomas. Cancers (Basel) 2022; 14:4443. [PMID: 36139601 PMCID: PMC9496906 DOI: 10.3390/cancers14184443] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/02/2022] [Accepted: 09/08/2022] [Indexed: 11/17/2022] Open
Abstract
Omega-3 polyunsaturated fatty acids (ω-3 PUFAs) are thought to protect against colorectal adenoma (CRA) development. We aimed to further understand the underlying mechanisms by examining the relationships between ω-3 PUFAs and the gut microbiota on CRAs. We assessed the mucosal microbiota via bacterial 16S rRNA sequencing among 217 CRA cases and 218 controls who completed PUFA intake questionnaires. The overall microbial composition was assessed by α-diversity measurements (diversity, richness, and evenness). Global metabolomics was conducted using a random subset of case−control pairs (n = 50). We compared microbiota and metabolite signatures between cases and controls according to fold change (FC). Odds ratios (OR) and confidence intervals (CI) were estimated from logistic regression for associations of ω-3 PUFAs and the microbiota with CRAs. We observed an inverse association between overall ω-3 PUFA intake and CRAs, especially for short-chain ω -3 PUFAs (OR = 0.45, 95% CI: 0.21, 0.97). Such inverse associations were modified by bacterial evenness (p-interaction = 0.03). Participants with higher levels (FC > 2) of bile acid-relevant metabolites were more likely to have CRAs than the controls, and the correlation between bile acids and bacterial diversity differed by case−control status. Our findings suggest that ω-3 PUFAs are inversely associated with CRA development, and the association may be modified by gut microbiota profiles.
Collapse
Affiliation(s)
- Tengteng Wang
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Nicole M. Brown
- Center for Gastrointestinal Disease and Biology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Amber N. McCoy
- Center for Gastrointestinal Disease and Biology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Robert S. Sandler
- Center for Gastrointestinal Disease and Biology, University of North Carolina, Chapel Hill, NC 27599, USA
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Temitope O. Keku
- Center for Gastrointestinal Disease and Biology, University of North Carolina, Chapel Hill, NC 27599, USA
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
3
|
Supplementation with phytoestrogens and insoluble fibers reduces intestinal carcinogenesis and restores ER-β expression in Apc-driven colorectal carcinogenesis. Eur J Cancer Prev 2019; 29:27-35. [PMID: 31651566 DOI: 10.1097/cej.0000000000000542] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Supplementation with phytoestrogens and insoluble fibers has been reported to reduce duodenal polyps in colectomized familial adenomatous polyposis patients, with a mechanism involving, at least in part, upregulation of estrogen receptor-β subtype, whose expression is lowered during intestinal tumorigenesis. These data suggest a protective effect also in the colon, the main target organ for tumorigenesis in familial adenomatous polyposis and a major cancer type in non-familial (sporadic) cancers. Therefore, we tested whether a similar preparation might reduce tumorigenesis in the colon of Pirc rats (F344/NTac-Apc) mutated in the Apc gene and thus, like familial adenomatous polyposis patients, spontaneously developing multiple tumors in the colon. We first demonstrate that estrogen receptor-β expression in Pirc rat colon is significantly down-regulated compared to age-matched wt rats. Then, Pirc rats aged 1 month were treated for 3 months with Adipol (Adi), a patented preparation containing phytoestrogens and insoluble fibers. Colon tumorigenesis was significantly reduced by Adi treatment (colon tumors/rat were 5.3 ± 0.8 and 2.9 ± 0.3, Mucin Depleted Foci/rat 127 ± 6.6 and 97.1 ± 8.6 in Controls and Adi-treated rats, respectively, means ± SE, P < 0.01). The treatment also normalized colon proliferation pattern along the crypt and significantly increased apoptosis in colon tumors. Estrogen receptor-β expression was increased by Adi treatment, especially in the tumors. These positive effects suggest that Adipol may be exploited as a chemopreventive agent to reduce cancer risk in familial adenomatous polyposis patients and to postpone prophylactic colectomy. Moreover, given the similarities between familial adenomatous polyposis and sporadic colorectal cancer, it might also be used as chemopreventive agent in colorectal cancer patients at risk.
Collapse
|
4
|
DeLuca JAA, Garcia-Villatoro EL, Allred CD. Flaxseed Bioactive Compounds and Colorectal Cancer Prevention. Curr Oncol Rep 2018; 20:59. [DOI: 10.1007/s11912-018-0704-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
5
|
Effect of α-linolenic acid-rich diacylglycerol oil on protein kinase C activation in the rat digestive tract and lingual mucosa. Food Chem Toxicol 2017; 103:168-173. [DOI: 10.1016/j.fct.2017.02.033] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 12/27/2016] [Accepted: 02/23/2017] [Indexed: 11/21/2022]
|
6
|
Alpha-linolenic acid-enriched diacylglycerol oil does not promote tumor development in tongue and gastrointestinal tract tissues in a medium-term multi-organ carcinogenesis bioassay using male F344 rat. Food Chem Toxicol 2017; 106:185-192. [PMID: 28465188 DOI: 10.1016/j.fct.2017.04.040] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 04/19/2017] [Accepted: 04/28/2017] [Indexed: 11/23/2022]
Abstract
Alpha-linolenic acid (ALA)-enriched diacylglycerol (DAG) oil is an edible oil enriched with DAG (>80%) and ALA (>50%). The present study investigated whether ALA-DAG oil promotes tumorigenesis in the tongue and gastrointestinal tract, using a rat medium-term multi-organ carcinogenesis bioassay model. Rats were treated with five genotoxic carcinogens to induce multi-organ tumorigenesis until week 4, and from 1 week after withdrawal, fed a semi-synthetic diet (AIN-93G) containing ALA-DAG oil at concentrations of 0, 13,750, 27,500, and 55,000 ppm. Rats fed AIN-93G containing 55,000 ppm ALA-triacylglycerol or a standard basal diet served as reference and negative control groups, respectively. Animals were euthanized at week 30. ALA-DAG oil was shown to have no effects on survival, general condition, body weight, food consumption, or organ weight. More discolored spots were observed in the stomachs of the 13,750- and 55,000-ppm ALA-DAG groups than in those of the control groups; however, there were no differences in the frequency of histopathological findings across groups. There were no meaningful increases in the incidence of pre-neoplastic and neoplastic lesions in the tongue and gastrointestinal tract among the groups. We therefore conclude that ALA-DAG oil does not promote tumor development in the digestive system.
Collapse
|
7
|
Wang Y, DiSalvo M, Gunasekara DB, Dutton J, Proctor A, Lebhar MS, Williamson IA, Speer J, Howard RL, Smiddy NM, Bultman SJ, Sims CE, Magness ST, Allbritton NL. Self-renewing Monolayer of Primary Colonic or Rectal Epithelial Cells. Cell Mol Gastroenterol Hepatol 2017; 4:165-182.e7. [PMID: 29204504 PMCID: PMC5710741 DOI: 10.1016/j.jcmgh.2017.02.011] [Citation(s) in RCA: 149] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 02/15/2017] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS Three-dimensional organoid culture has fundamentally changed the in vitro study of intestinal biology enabling novel assays; however, its use is limited because of an inaccessible luminal compartment and challenges to data gathering in a three-dimensional hydrogel matrix. Long-lived, self-renewing 2-dimensional (2-D) tissue cultured from primary colon cells has not been accomplished. METHODS The surface matrix and chemical factors that sustain 2-D mouse colonic and human rectal epithelial cell monolayers with cell repertoires comparable to that in vivo were identified. RESULTS The monolayers formed organoids or colonoids when placed in standard Matrigel culture. As with the colonoids, the monolayers exhibited compartmentalization of proliferative and differentiated cells, with proliferative cells located near the peripheral edges of growing monolayers and differentiated cells predominated in the central regions. Screening of 77 dietary compounds and metabolites revealed altered proliferation or differentiation of the murine colonic epithelium. When exposed to a subset of the compound library, murine organoids exhibited similar responses to that of the monolayer but with differences that were likely attributable to the inaccessible organoid lumen. The response of the human primary epithelium to a compound subset was distinct from that of both the murine primary epithelium and human tumor cells. CONCLUSIONS This study demonstrates that a self-renewing 2-D murine and human monolayer derived from primary cells can serve as a physiologically relevant assay system for study of stem cell renewal and differentiation and for compound screening. The platform holds transformative potential for personalized and precision medicine and can be applied to emerging areas of disease modeling and microbiome studies.
Collapse
Key Words
- 2-D, two-dimensional
- 3-D, three-dimensional
- ALP, alkaline phosphatase
- CAG, cytomegalovirus enhancer plus chicken actin promoter
- CI, confidence interval
- Colonic Epithelial Cells
- Compound Screening
- ECM, extracellular matrix
- EDU, 5-ethynyl-2′-deoxyuridine
- EGF, epidermal growth factor
- ENR-W, cell medium with [Wnt-3A] of 30 ng/mL
- ENR-w, cell medium with [Wnt-3A] of 10 ng/mL
- HISC, human intestinal stem cell medium
- IACUC, Institutional Animal Care and Use Committee
- ISC, intestinal stem cell
- Monolayer
- Organoids
- PBS, phosphate-buffered saline
- PDMS, polydimethylsiloxane
- RFP, red fluorescent protein
- SEM, scanning electron microscope
- SSMD, strictly standardized mean difference
- UNC, University of North Carolina
- α-ChgA, anti-chromogranin A
- α-Muc2, anti-mucin2
Collapse
Affiliation(s)
- Yuli Wang
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina
| | - Matthew DiSalvo
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill, and North Carolina State University, Raleigh, North Carolina
| | - Dulan B. Gunasekara
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina
| | - Johanna Dutton
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill, and North Carolina State University, Raleigh, North Carolina
| | - Angela Proctor
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina
| | - Michael S. Lebhar
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill, and North Carolina State University, Raleigh, North Carolina
| | - Ian A. Williamson
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill, and North Carolina State University, Raleigh, North Carolina
| | - Jennifer Speer
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina
| | - Riley L. Howard
- Department of Applied Physical Sciences, University of North Carolina, Chapel Hill, North Carolina
| | - Nicole M. Smiddy
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina
| | - Scott J. Bultman
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina
| | - Christopher E. Sims
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina
| | - Scott T. Magness
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill, and North Carolina State University, Raleigh, North Carolina
| | - Nancy L. Allbritton
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina,Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill, and North Carolina State University, Raleigh, North Carolina,Department of Applied Physical Sciences, University of North Carolina, Chapel Hill, North Carolina,Correspondence Address correspondence to: Nancy L. Allbritton, MD, PhD, Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina 27599. fax: (919) 962-2388.Department of ChemistryUniversity of North CarolinaChapel HillNorth Carolina 27599
| |
Collapse
|
8
|
Power KA, Lepp D, Zarepoor L, Monk JM, Wu W, Tsao R, Liu R. Dietary flaxseed modulates the colonic microenvironment in healthy C57Bl/6 male mice which may alter susceptibility to gut-associated diseases. J Nutr Biochem 2015; 28:61-9. [PMID: 26878783 DOI: 10.1016/j.jnutbio.2015.09.028] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 09/28/2015] [Accepted: 09/30/2015] [Indexed: 12/25/2022]
Abstract
Understanding how dietary components alter the healthy baseline colonic microenvironment is important in determining their roles in influencing gut health and gut-associated diseases. Dietary flaxseed (FS) has demonstrated anti-colon cancer effects in numerous rodent models, however, exacerbated acute colonic mucosal injury and inflammation in a colitis model. This study investigates whether FS alters critical aspects of gut health in healthy unchallenged mice, which may help explain some of the divergent effects observed following different gut-associated disease challenges. Four-week-old C57Bl/6 male mice were fed an AIN-93G basal diet (BD) or an isocaloric BD+10% ground FS diet for 3 weeks. FS enhanced colon goblet cell density, mucus production, MUC2 mRNA expression, and cecal short chain fatty acid levels, indicative of beneficial intestinal barrier integrity responses. Additionally, FS enhanced colonic regenerating islet-derived protein 3 gamma (RegIIIγ) and reduced MUC1 and resistin-like molecule beta (RELMβ) mRNA expression which may indicate altered responses in regulating microbial defense and injury repair responses. FS diet altered the fecal microbial community structure (16S rRNA gene profiling), including a 20-fold increase in Prevotella spp. and a 30-fold reduction in Akkermansia muciniphila abundance. A 10-fold reduction in A. muciniphila abundance by FS was also demonstrated in the colon tissue-associated microbiota (quantitative PCR). Furthermore, fecal branched chain fatty acids were increased by FS, indicative of increased microbial-derived putrefactive compounds. In conclusion, consumption of a FS-supplemented diet alters the baseline colonic microenvironment of healthy mice which may modify subsequent mucosal microbial defense and injury-repair responses leading to altered susceptibility to different gut-associated diseases.
Collapse
Affiliation(s)
- Krista A Power
- Guelph Food Research Centre, Agriculture and Agri-Food Canada, AAFC, Guelph, Ontario.
| | - Dion Lepp
- Guelph Food Research Centre, Agriculture and Agri-Food Canada, AAFC, Guelph, Ontario
| | - Leila Zarepoor
- Guelph Food Research Centre, Agriculture and Agri-Food Canada, AAFC, Guelph, Ontario
| | - Jennifer M Monk
- Guelph Food Research Centre, Agriculture and Agri-Food Canada, AAFC, Guelph, Ontario
| | - Wenqing Wu
- Guelph Food Research Centre, Agriculture and Agri-Food Canada, AAFC, Guelph, Ontario
| | - Rong Tsao
- Guelph Food Research Centre, Agriculture and Agri-Food Canada, AAFC, Guelph, Ontario
| | - Ronghua Liu
- Guelph Food Research Centre, Agriculture and Agri-Food Canada, AAFC, Guelph, Ontario
| |
Collapse
|
9
|
Schiessel DL, Yamazaki RK, Kryczyk M, Coelho I, Yamaguchi AA, Pequito DCT, Brito GAP, Borghetti G, Fernandes LC. α-Linolenic Fatty Acid Supplementation Decreases Tumor Growth and Cachexia Parameters in Walker 256 Tumor-Bearing Rats. Nutr Cancer 2015; 67:839-46. [DOI: 10.1080/01635581.2015.1043021] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
10
|
Gurzell EA, Wiesinger JA, Morkam C, Hemmrich S, Harris WS, Fenton JI. Is the omega-3 index a valid marker of intestinal membrane phospholipid EPA+DHA content? Prostaglandins Leukot Essent Fatty Acids 2014; 91:87-96. [PMID: 24913088 PMCID: PMC4127132 DOI: 10.1016/j.plefa.2014.04.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Revised: 03/24/2014] [Accepted: 04/04/2014] [Indexed: 01/07/2023]
Abstract
Despite numerous studies investigating n-3 long chain polyunsaturated fatty acid (LCPUFA) supplementation and inflammatory bowel diseases (IBD), the extent to which dietary n-3 LCPUFAs incorporate in gastrointestinal (GI) tissues and correlate with red blood cell (RBC) n-3 LCPUFA content is unknown. In this study, mice were fed three diets with increasing percent of energy (%en) derived from eicosapentaenoic acid (EPA)+docosahexaenoic acid (DHA). Dietary levels reflected recommended intakes of fish/fish oil by the American Heart Association. We analyzed the FA composition of phospholipids extracted from RBCs, plasma, and GI tissues. We observed that the 0.1%en EPA+DHA diet was sufficient to significantly increase the omega-3 index (RBC EPA+DHA) after 5 week feeding. The baseline EPA levels were 0.2-0.6% across all tissues increasing to 1.6-4.3% in the highest EPA+DHA diet; these changes resulted in absolute increases of 1.4-3.9% EPA across tissues. The baseline DHA levels were 2.2-5.9% across all tissues increasing to 5.8-10.5% in the highest EPA+DHA diet; these changes resulted in absolute increases of 3.2-5.7% DHA across tissues. These increases in EPA and DHA across all tissues resulted in strong (r>0.91) and significant (P<0.001) linear correlations between the omega-3 index and plasma/GI tissue EPA+DHA content, suggesting that the omega-3 index reflects the relative amounts of EPA+DHA in GI tissues. These data demonstrate that the GI tissues are highly responsive to dietary LCPUFA supplementation and that the omega-3 index can serve as a valid biomarker for assessing dietary EPA+DHA incorporation into GI tissues.
Collapse
Affiliation(s)
- Eric A Gurzell
- Department of Food Science and Human Nutrition, Michigan State University, MI, United States
| | - Jason A Wiesinger
- Department of Food Science and Human Nutrition, Michigan State University, MI, United States
| | - Christina Morkam
- Department of Food Science and Human Nutrition, Michigan State University, MI, United States
| | - Sophia Hemmrich
- Department of Food Science and Human Nutrition, Michigan State University, MI, United States
| | - William S Harris
- Sanford School of Medicine, The University of South Dakota, Sioux Falls, SD (WSH), United States
| | - Jenifer I Fenton
- Department of Food Science and Human Nutrition, Michigan State University, MI, United States; College of Osteopathic Medicine, East Lansing, MI, United States.
| |
Collapse
|
11
|
Goyal A, Sharma V, Upadhyay N, Gill S, Sihag M. Flax and flaxseed oil: an ancient medicine & modern functional food. JOURNAL OF FOOD SCIENCE AND TECHNOLOGY 2014; 51:1633-53. [PMID: 25190822 PMCID: PMC4152533 DOI: 10.1007/s13197-013-1247-9] [Citation(s) in RCA: 306] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Revised: 12/11/2013] [Accepted: 12/26/2013] [Indexed: 02/05/2023]
Abstract
Flaxseed is emerging as an important functional food ingredient because of its rich contents of α-linolenic acid (ALA, omega-3 fatty acid), lignans, and fiber. Flaxseed oil, fibers and flax lignans have potential health benefits such as in reduction of cardiovascular disease, atherosclerosis, diabetes, cancer, arthritis, osteoporosis, autoimmune and neurological disorders. Flax protein helps in the prevention and treatment of heart disease and in supporting the immune system. As a functional food ingredient, flax or flaxseed oil has been incorporated into baked foods, juices, milk and dairy products, muffins, dry pasta products, macaroni and meat products. The present review focuses on the evidences of the potential health benefits of flaxseed through human and animals' recent studies and commercial use in various food products.
Collapse
Affiliation(s)
- Ankit Goyal
- />Dairy Chemistry Division, National Dairy Research Institute, Karnal, Haryana India 132001
| | - Vivek Sharma
- />Dairy Chemistry Division, National Dairy Research Institute, Karnal, Haryana India 132001
| | - Neelam Upadhyay
- />Dairy Chemistry Division, National Dairy Research Institute, Karnal, Haryana India 132001
| | - Sandeep Gill
- />BITS Pilani, Hyderabad Campus Shameerpet Mandal Rangareddy District, Hyderabad, Andhra Pradesh India 500078
| | - Manvesh Sihag
- />Dairy Chemistry Division, National Dairy Research Institute, Karnal, Haryana India 132001
| |
Collapse
|
12
|
Zarepoor L, Lu JT, Zhang C, Wu W, Lepp D, Robinson L, Wanasundara J, Cui S, Villeneuve S, Fofana B, Tsao R, Wood GA, Power KA. Dietary flaxseed intake exacerbates acute colonic mucosal injury and inflammation induced by dextran sodium sulfate. Am J Physiol Gastrointest Liver Physiol 2014; 306:G1042-55. [PMID: 24763556 DOI: 10.1152/ajpgi.00253.2013] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
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-1β), 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-1β) 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.
Collapse
Affiliation(s)
- Leila Zarepoor
- Guelph Food Research Centre, Agriculture and Agri-Food Canada (AAFC), Guelph, Ontario, Canada; Department of Human Health and Nutritional Science, University of Guelph, Guelph, Ontario, Canada
| | - Jenifer T Lu
- Guelph Food Research Centre, Agriculture and Agri-Food Canada (AAFC), Guelph, Ontario, Canada; Department of Human Health and Nutritional Science, University of Guelph, Guelph, Ontario, Canada
| | - Claire Zhang
- Guelph Food Research Centre, Agriculture and Agri-Food Canada (AAFC), Guelph, Ontario, Canada; Department of Human Health and Nutritional Science, University of Guelph, Guelph, Ontario, Canada
| | - Wenqing Wu
- Guelph Food Research Centre, Agriculture and Agri-Food Canada (AAFC), Guelph, Ontario, Canada
| | - Dion Lepp
- Guelph Food Research Centre, Agriculture and Agri-Food Canada (AAFC), Guelph, Ontario, Canada
| | - Lindsay Robinson
- Department of Human Health and Nutritional Science, University of Guelph, Guelph, Ontario, Canada
| | | | - Steve Cui
- Guelph Food Research Centre, Agriculture and Agri-Food Canada (AAFC), Guelph, Ontario, Canada
| | | | - Bourlaye Fofana
- Crops and Livestock Research Centre, AAFC, Charlottetown, Prince Edward Island, Canada; and
| | - Rong Tsao
- Guelph Food Research Centre, Agriculture and Agri-Food Canada (AAFC), Guelph, Ontario, Canada
| | - Geoffrey A Wood
- Department of Pathobiology, University of Guelph, Guelph, Ontario, Canada
| | - Krista A Power
- Guelph Food Research Centre, Agriculture and Agri-Food Canada (AAFC), Guelph, Ontario, Canada; Department of Human Health and Nutritional Science, University of Guelph, Guelph, Ontario, Canada;
| |
Collapse
|
13
|
Abstract
Protein kinase C (PKC) is a family of phospholipid-dependent serine/threonine kinases, which can be further classified into three PKC isozymes subfamilies: conventional or classic, novel or nonclassic, and atypical. PKC isozymes are known to be involved in cell proliferation, survival, invasion, migration, apoptosis, angiogenesis, and drug resistance. Because of their key roles in cell signaling, PKC isozymes also have the potential to be promising therapeutic targets for several diseases, such as cardiovascular diseases, immune and inflammatory diseases, neurological diseases, metabolic disorders, and multiple types of cancer. This review primarily focuses on the activation, mechanism, and function of PKC isozymes during cancer development and progression.
Collapse
|
14
|
|
15
|
Murff HJ, Shrubsole MJ, Cai Q, Smalley WE, Dai Q, Milne GL, Ness RM, Zheng W. Dietary intake of PUFAs and colorectal polyp risk. Am J Clin Nutr 2012; 95:703-12. [PMID: 22277551 PMCID: PMC3278245 DOI: 10.3945/ajcn.111.024000] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Marine-derived n-3 (omega-3) PUFAs may reduce risk of developing colorectal cancer; however, few studies have investigated the association of n-3 PUFA intakes on colorectal polyp risk. OBJECTIVE The objective of this study was to examine the associations of dietary PUFA intake on risk of colorectal adenomatous and hyperplastic polyps. DESIGN This was a colonoscopy-based case-control study that included 3166 polyp-free control subjects, 1597 adenomatous polyp cases, and 544 hyperplastic polyp cases. Dietary PUFA intake was calculated from food-frequency questionnaires and tested for association by using unconditional logistic regression. The urinary prostaglandin E(2) metabolite, which is a biomarker of prostaglandin E(2) production, was measured in 896 participants by using liquid chromatography and tandem mass spectrometry. RESULTS n-6 PUFAs were not associated with adenomatous or hyperplastic polyps in either men or women. Marine-derived n-3 PUFAs were associated with reduced risk of colorectal adenomas in women only, with an adjusted OR of 0.67 (95% CI: 0.47, 0.97) for the highest quintile of intake compared with the lowest quintile of intake (P-trend = 0.01). Dietary intake of α-linolenic acid was associated with an increased risk of hyperplastic polyps in men (P-trend = 0.03), which was not seen in women. In women, but not in men, dietary intake of marine-derived n-3 PUFAs was negatively correlated with urinary prostaglandin E(2) production (r = -0.18; P = 0.002). CONCLUSION Higher intakes of marine-derived n-3 PUFAs are associated with lower risk of adenomatous polyps in women, and the association may be mediated in part through a reduction in the production of prostaglandin E(2). This trial was registered at clinicaltrials.gov as NCT00625066.
Collapse
Affiliation(s)
- Harvey J Murff
- Division of General Internal Medicine, Vanderbilt University School of Medicine, Nashville, TN 37203, USA.
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Galbraith RL, Poole EM, Duggan D, Muehling J, Hsu L, Makar K, Xiao L, Potter JD, Ulrich CM. Polymorphisms in WNT6 and WNT10A and colorectal adenoma risk. Nutr Cancer 2011; 63:558-64. [PMID: 21547848 DOI: 10.1080/01635581.2011.542539] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The WNT/β-catenin signaling pathway upregulates transcription of genes involved in cell proliferation and cancer progression; it has been implicated in colorectal adenoma formation. To date, no studies have examined polymorphisms in WNT genes or WNT gene-environment interactions in relation to adenoma risk. Within a colonoscopy-based case-control study of 628 adenoma cases and 516 polyp-free controls, we analyzed two tagSNPs in WNT6 (rs6747776 G > C, rs6754599 G > C) and WNT10A (rs7349332 G > A, rs10177996 A > G). The WNT6 rs6747776 homozygous minor allele (CC) was associated with increased risk of colorectal adenoma (OR = 2.75, 95% CI: 1.03-7.31). We observed a statistically significant interaction between WNT6 rs6747776 and the proportion of calories from total fat (P-int = 0.02), where the highest risk was observed among those with minor alleles and lowest fat intake. We also detected a marginally significant (0.05 < P ≤ 0.10) interaction with fish intake (P-int = 0.09). Additionally, a marginally significant interaction was observed between proportion of calories from saturated fat and the WNT10A rs7349332 polymorphism. Our results suggest that genetic variability in the WNT pathway may play a role in colorectal adenoma formation or may partly mediate the increased risk of colorectal cancer associated with fat intake.
Collapse
|
17
|
The health promoting properties of the conjugated isomers of α-linolenic acid. Lipids 2010; 46:105-19. [PMID: 21161605 DOI: 10.1007/s11745-010-3501-5] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2010] [Accepted: 11/03/2010] [Indexed: 12/17/2022]
Abstract
The bioactive properties of the conjugated linoleic acid (CLA) isomers have long been recognised and are the subject of a number of excellent reviews. However, despite this prominence the CLA isomers are not the only group of naturally occurring dietary conjugated fatty acids which have shown potent bioactivity. In a large number of in vitro and in vivo studies, conjugated α-linolenic acid (CLNA) isomers have displayed potent anti-inflammatory, immunomodulatory, anti-obese and anti-carcinogenic activity, along with the ability to improve biomarkers of cardio-vascular health. CLNA isomers are naturally present in high concentrations in a large variety of seed oils but can also be produced in vitro by strains of lactobacilli and bifidobactena through the activity of the enzyme linoleic acid isomerase on α-linolenic acid. In this review, we will address the possible therapeutic roles that CLNA may play in a number of conditions afflicting Western society and the mechanisms through which this activity is mediated.
Collapse
|
18
|
Tammariello AE, Milner JA. Mouse models for unraveling the importance of diet in colon cancer prevention. J Nutr Biochem 2010; 21:77-88. [PMID: 20122631 DOI: 10.1016/j.jnutbio.2009.09.014] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2009] [Revised: 07/30/2009] [Accepted: 09/21/2009] [Indexed: 01/28/2023]
Abstract
Diet and genetics are both considered important risk determinants for colorectal cancer, a leading cause of death worldwide. Several genetically engineered mouse models have been created, including the ApcMin mouse, to aid in the identification of key cancer related processes and to assist with the characterization of environmental factors, including the diet, which influence risk. Current research using these models provides evidence that several bioactive food components can inhibit genetically predisposed colorectal cancer, while others increase risk. Specifically, calorie restriction or increased exposure to n-3 fatty acids, sulforaphane, chafuroside, curcumin and dibenzoylmethane were reported protective. Total fat, calories and all-trans retinoic acid are associated with an increased risk. Unraveling the importance of specific dietary components in these models is complicated by the basal diet used, the quantity of test components provided and interactions among food components. Newer models are increasingly available to evaluate fundamental cellular processes, including DNA mismatch repair, immune function and inflammation as markers for colon cancer risk. Unfortunately, these models have been used infrequently to examine the influence of specific dietary components. The enhanced use of these models can shed mechanistic insights about the involvement of specific bioactive food and components and energy as determinants of colon cancer risk. However, the use of available mouse models to exactly represent processes important to human gastrointestinal cancers will remain a continued scientific challenge.
Collapse
|
19
|
Boudry G, Douard V, Mourot J, Lallès JP, Le Huërou-Luron I. Linseed oil in the maternal diet during gestation and lactation modifies fatty acid composition, mucosal architecture, and mast cell regulation of the ileal barrier in piglets. J Nutr 2009; 139:1110-7. [PMID: 19403717 DOI: 10.3945/jn.108.102640] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
In this study, we investigated the effect of supplementation of the maternal diet with linseed oil [rich in 18:3(n-3)] on fatty acid composition, mucosal architecture, and mast cell regulation of barrier function in piglet ileum. Sixteen sows were fed a lard (LAR)- or a linseed oil (LSO)-based diet during gestation and lactation. Fatty acid composition of maternal RBC at parturition and of milk at d 14 of lactation were determined. Fatty acid composition, villous-crypt structure, and permeability to horseradish peroxidase in Ussing chambers after mast cell degranulation were determined in the ileum of piglets at d 0, 7, and 28. At d 0, 18:3(n-3) and 20:5(n-3) levels were higher, but 22:6(n-3) and 20:4(n-6) levels were lower in both maternal RBC and piglet ileum of the LSO group. Levels of 18:3(n-3) were also higher in the milk of LSO sows. Levels of 18:3(n-3) were higher in LSO piglet ileum at d 7 and 28. Moreover, at d 28, 20:4(n-6) ileal levels tended (P = 0.09) to be lower in LSO than in LAR piglets, in parallel with a lower mRNA expression of Delta5 desaturase. LSO piglets had shorter villi at d 0 and shorter crypts at d 7 compared with LAR piglets. The effect of mast cell degranulation on ileal permeability decreased with age in both groups but reached a minimum sooner in the LSO group (d 7) than in the LAR group (d 28). In conclusion, linseed oil supplementation of the maternal diet profoundly modifies the fatty acid composition, structure, and physiology of the offspring ileum.
Collapse
Affiliation(s)
- Gaëlle Boudry
- INRA, UMR1079, Systèmes d'Elevage, Nutrition Animale et Humaine, F-35000 Rennes, France.
| | | | | | | | | |
Collapse
|
20
|
Davis PA, Jenab M, Vanden Heuvel JP, Furlong T, Taylor S. Tree nut and peanut consumption in relation to chronic and metabolic diseases including allergy. J Nutr 2008; 138:1757S-1762S. [PMID: 18716182 DOI: 10.1093/jn/138.9.1757s] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
The New and Emerging Research session highlighted the emerging understanding of both the positive and negative effects of nuts consumption on health. The limited nature of both experimental and epidemiological evidence for positive relationship(s) between nut intake and health were noted. Study inconsistency and limitations, particularly survey methodology, were explored. Recent results from epidemiologic studies indicating a potential negative association between nut and seed intake and cancer risk were reviewed. The ability of walnuts to reduce endothelin suggests an interesting biochemical mechanism of nut action that may affect other endothelin-associated diseases, which should be further explored. The effects of nuts and their constituents on a nuclear receptor screen (PPARalpha, beta/delta, gamma, LXRalpha, beta, RXRalpha, beta, gamma, PXR, and FXR) have been explored. Nut allergenicity and approaches necessary to minimize this effect were also described. In contrast to the positive effects, nut allergies present tree nut-allergic consumers with health challenges. The Food Allergy and Anaphylaxis Network stressed the importance of ensuring that consumers with food allergies have legible, accurate food labels. The Food Allergen Labeling and Consumer Protection Act has engendered precautionary, worst-case allergen scenario labeling statements with unknown benefits to consumer health. Issues of cross-contamination due to shared equipment and shared facilities highlighted the need to rely on allergen control programs that use ELISA technology and have increased understanding of nut allergens. Ultimately, to maximize the positive benefits of nuts, the consumer must be provided with all the information required to make an informed choice.
Collapse
Affiliation(s)
- Paul A Davis
- Department of Nutrition, University of California, Davis, CA 95616, USA.
| | | | | | | | | |
Collapse
|
21
|
Abstract
Epidemiologic studies have suggested for decades an association between dietary fat and cancer risk. A large body of work performed in tissue culture and xenograft models of cancer supports an important role of various types of fat in modulating the cancer phenotype. Yet, the molecular mechanisms underlining the effects of fat on cancer initiation and progression are largely unknown. The relationships between saturated fat, polyunsaturated fat, cholesterol or phytanic acid with cancer have been reviewed respectively. However, few have considered the relationship between all of these fats and cancer. The purpose of this review is to present a more cohesive view of dietary fat-gene interactions, and outline a working hypothesis of the intricate connection between fat, genes and cancer.
Collapse
Affiliation(s)
- Yong Q Chen
- Cancer Biology, Wake Forest University School of Medicine, Medical Center Blvd., Winston-Salem, NC 27157, USA.
| | | | | | | | | |
Collapse
|
22
|
Peñalvo JL, Nurmi T. Application of coulometric electrode array detection to the analysis of isoflavonoids and lignans. J Pharm Biomed Anal 2006; 41:1497-507. [PMID: 16644172 DOI: 10.1016/j.jpba.2006.03.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2006] [Accepted: 03/15/2006] [Indexed: 11/20/2022]
Abstract
A comprehensive review of the available methods for the analysis of isoflavonoids and lignans in biological matrices based on coulometric electrode array detection (CEAD) is presented. Different aspects such as sample extraction and purification procedures as well as instrumental settings and method validation are discussed. Comparisons with other available protocols using different detection techniques such as mass spectrometry or immunoassay are also reported to underline the versatility and reliability of the detector. Practical notes and tips for scientists working with CEAD are also provided.
Collapse
Affiliation(s)
- José L Peñalvo
- Institute for Preventive Medicine, Nutrition and Cancer, Folkhälsan Research Center, and Division of Clinical Chemistry, University of Helsinki, Biomedicum, P.O. Box 63, 00014 Helsinki, Finland.
| | | |
Collapse
|
23
|
Affiliation(s)
- Clifford Hall
- Department of Cereal and Food Sciences, North Dakota State University, Fargo, North Dakota 58105, USA
| | | | | |
Collapse
|