Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 format correct + 3099 word(s) 3099 2020-11-09 04:36:54 |
2 format correct -2 word(s) 3097 2020-11-17 02:56:11 | |
3 format correct -6 word(s) 3093 2020-11-17 03:08:07 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Tu, M.; Wang, W.; Zhang, G.; Hammock, B.D. ω-3 PUFA on colon cancer. Encyclopedia. Available online: https://encyclopedia.pub/entry/3048 (accessed on 16 April 2024).
Tu M, Wang W, Zhang G, Hammock BD. ω-3 PUFA on colon cancer. Encyclopedia. Available at: https://encyclopedia.pub/entry/3048. Accessed April 16, 2024.
Tu, Maolin, Weicang Wang, Guodong Zhang, Bruce D. Hammock. "ω-3 PUFA on colon cancer" Encyclopedia, https://encyclopedia.pub/entry/3048 (accessed April 16, 2024).
Tu, M., Wang, W., Zhang, G., & Hammock, B.D. (2020, November 17). ω-3 PUFA on colon cancer. In Encyclopedia. https://encyclopedia.pub/entry/3048
Tu, Maolin, et al. "ω-3 PUFA on colon cancer." Encyclopedia. Web. 17 November, 2020.
ω-3 PUFA on colon cancer
Edit

Substantial human and animal studies support the beneficial effects of ω-3 polyunsaturated fatty acids (PUFAs) on colonic inflammation and colorectal cancer (CRC). However, there are inconsistent results, which have shown that ω-3 PUFAs have no effect or even detrimental effects, making it difficult to effectively implement ω-3 PUFAs for disease prevention. A better understanding of the molecular mechanisms for the anti-inflammatory and anticancer effects of ω-3 PUFAs will help to clarify their potential health-promoting effects, provide a scientific base for cautions for their use, and establish dietary recommendations.

ω-3 PUFAs colorectal cancer eicosanoids cytochrome P450 monooxygenases soluble epoxide hydrolase

1. Introduction

There are ~1.8 million new cases of and ~881,000 deaths from colorectal cancer (CRC) every year [1]. It is estimated that ~30% of cancers in developed countries are diet-related [2]. Therefore, it is important to develop effective diet-based prevention strategies to reduce CRC risks. Epidemiological and preclinical data support that ω-3 polyunsaturated fatty acids (PUFAs), such as plant-derived α-linolenic acid (ALA, 18:3ω-3) and marine fish-derived eicosapentaenoic acid (EPA, 20:5ω-3), docosapentaenoic acid (DPA, 22:5ω-3), and docosahexaenoic acid (DHA, 22:6ω-3), may reduce CRC risks, in part, through suppressing colonic inflammation. In contrast, ω-6 PUFAs, such as linoleic acid (LA, 18:2ω-6) and arachidonic acid (ARA, 20:4ω-6), are suggested to exaggerate the development of colonic inflammation and CRC [3][4][5][6][7][8]. This is important because the current Western diet has 30–50-times more ω-6 PUFAs than ω-3 PUFAs. The validation of the beneficial effects of ω-3 PUFAs on CRC will have a significant impact on public health. However, after decades of research, the anti-CRC efficacy of ω-3 PUFAs remains inconclusive, making it difficult to make dietary recommendations or guidelines of ω-3 PUFAs for CRC prevention [9]. The inconsistent results suggest that there could be more complex mechanisms, which may be subject to specific cellular and/or metabolic modulation, involved in the anticancer and anti-inflammatory effects of ω-3 PUFAs. Therefore, it is of critical importance to better understand the mechanisms behind the anticancer and anti-inflammatory activities of ω-3 PUFAs to optimize their use for CRC prevention.

A widely accepted molecular mechanism to explain the potential health-promoting effects of ω-3 PUFAs is that they can compete with ARA (a major ω-6 PUFA) for the enzymatic metabolism catalyzed by cyclooxygenase (COX), lipoxygenase (LOX), and cytochrome P450 (CYP) enzymes, leading to reduced levels of ω-6-series metabolites (termed eicosanoids) that are predominately proinflammatory and protumorigenic, and/or increased levels of ω-3-series metabolites, which have less detrimental or even beneficial effects [10][11][12][13]. A recent study showed that there is a high degree of interindividual variability in metabolizing ω-3 PUFAs to generate lipid metabolites [14]. Thus, it is feasible that polymorphisms in the genes encoding the ω-3 PUFA-metabolizing enzymes could affect the metabolism of ω-3 PUFAs, impacting the generation of bioactive lipid metabolites in tissues and contributing to observed mixed results in ω-3 PUFA studies [15]. A better understanding of the interactions of ω-3 PUFAs with their metabolizing enzymes could lead to targeted human studies to better understand the metabolic individuality and nutrition effects of ω-3 PUFAs [15][16].

In this review, we summarize recent studies of ω-3 PUFAs on CRC and colonic inflammation (inflammatory bowel disease (IBD)) and discuss the potential roles of ω-3 PUFA-metabolizing enzymes, notably the CYP enzymes, in mediating the actions of ω-3 PUFAs.

2. Effects of ω-3 PUFAs on CRC and IBD

2.1. Effects of ω-3 PUFAs on CRC

Epidemiological and preclinical studies support the preventive effects of ω-3 PUFAs on CRC. In Table 1, we focus on the recent human studies on ω-3 PUFAs, as well as previous studies that have shown the beneficial effect of the ω-3 PUFAs and have been discussed by other review articles. A meta-analysis demonstrated a small but significant ~12% reduction of CRC risk between the highest and lowest ω-3 PUFA consumption groups [17]. In the VITamins And Lifestyle (VITAL) cohort study, the individuals who routinely took fish oil supplements had lower risks of developing CRC compared with those who did not take supplements [18]. The European Prospective Investigation into Cancer and Nutrition (EPIC) study also showed that increased ω-3 PUFA consumption reduced CRC risks [19]. In a randomized, double-blind, placebo-controlled trial, EPA intake was associated with reduced polyp number and size in familial adenomatous polyposis (FAP) patients [20]. Increased intake of ω-3 PUFAs was also associated with improved disease-free survival in stage III CRC patients [21]. In a phase II double-blind, randomized, placebo-controlled trial, EPA intake increased overall survival in advanced CRC patients undergoing liver resection due to liver metastases (CRCLM) [22]. Together, these studies support the conclusion that ω-3 PUFAs reduce the risks of CRC.

Table 1. Recent epidemiological and clinical studies of ω-3 polyunsaturated fatty acid (PUFA) supplementation for the prevention/treatment of colorectal cancer (CRC).

Study

Individuals

N

ω-3 PUFA treatment

Dose

Duration

Control treatment

Results

Reference

VITAL prospective cohort

US adults

68,109

Fish oil supplements

N/A

4+days/week for 3+years

no use

↓ CRC risk

Kantor et al., 2014 [18]

EPIC prospective cohort

European adults

521,324

Highest ω-3 PUFAs intake

>470 mg/day

Median 14.9 years

lowest ω-3 PUFAs intake

↓ CRC risk

Aglago et al., 2020 [19]

Randomized, double-blind, placebo-controlled trial

FAP patients

EPA-FFA (n = 28)

EPA-FFA

2 g/day

6 months

Placebo

(n = 27)

↓ polyp diameters

West et al., 2010 [20]

CALGB adjuvant chemotherapy trial

stage III

colon

cancer patients

1011

Highest marine ω-3 PUFAs intake

0.33-0.57 g/day

>8 years

lowest marine ω-3 PUFAs intake

↑ disease-free survival

Blarigan et al., 2018 [21]

Double-blind, randomised, placebo-controlled trial

CRCLM patients

EPA-FFA (n = 43)

EPA-FFA

2 g/day

12–65 days

Placebo

(n = 45)

↑ overall survival;

no effect in disease burden and early CRC recurrence rates

Cockbain et al., 2014 [22]

HPFS and NHS cohort

US adults

123,529

Highest marine ω-3 PUFAs intake

≥ 0.30 g/d (women)

≥ 0.41 g/d (men)

24–26 years

lowest marine ω-3 PUFAs intake

No effect on overall CRC risk; ↑ distal colon cancer risk in men and women;

↓ rectal cancer risk in men

Song et al., 2014 [23]

Randomized, double-blind, placebo-controlled clinical trial

colon cancer patients

ω-3 PUFA (n = 21)

ω-3 PUFA intravenous infusion

0.2 g/ kg/day

night before and morning after resection surgery

Saline infusions

(n = 23)

↑ infectious complications

Bakker et al., 2020 [24]

Abbreviations: VITAL, VITamins And Lifestyle; EPIC, European Prospective Investigation into Cancer and Nutrition; EPA, eicosapentaenoic acid; FFA, free fatty acid; FAP, familial adenomatous polyposis; CALGB, Cancer and Leukemia Group B; CRCLM, colorectal cancer liver metastases; HPFS, Health Professionals Follow-Up Study; NHS, Nurses' Health Study.

Consistent with the human studies, recent animal studies also support the beneficial effects of ω-3 PUFAs on CRC (Table 2). Treatment with an ω-3 PUFA mixture or EPA reduced intestinal polyposis formation in a spontaneous intestinal cancer model (using ApcMin/+ mice) [25][26]. Dietary administration of EPA also decreased tumor incidence and multiplicity in a chemically induced colitis-associated colorectal cancer (CAC) model [27]. In addition, administration of fish oil suppressed the aberrant crypt foci number and adenoma incidence in 1,2-dimethylhydrazine (DMH) or azoxymethane (AOM)-induced CRC models in rats [28][29]. Besides dietary feeding studies using ω-3 PUFAs, previous studies also showed that fat-1 transgenic mice, which have higher tissue levels of ω-3 PUFAs, have reduced development of CRC in both Apc gene mutation-induced CRC model [30] and chemically induced CAC model [31][32].

In addition to the orthotropic CRC tumor models discussed above, ω-3 PUFAs have also been shown to inhibit CRC in xenograft and metastasis models. Our recent study showed that administration of an ω-3 PUFAs-enriched diet inhibited MC38 (murine colon adenocarcinoma cell) tumor growth in a murine xenograft model [33]. Consistent with our result, fish oil- or DHA-rich diets attenuated tumor burden and aggressivity in HCT-116 or SW620 (both are human colon cancer cells) xenograft tumor models in nude mice [34][35][36]. In a MC-26 colon cancer cell-induced CRC metastasis model, treatment of EPA suppressed liver metastases in BALB/c mice [37]. In a CC531 colon cancer cell-derived liver metastasis model in rats, administration of an ω-3 PUFAs-rich diet reduced hepatic tumor incidence and burden [38]. Moreover, ω-3 PUFAs could be used to enhance the actions and reduce the toxicity of anticancer drugs. The coadministration of oxaliplatin and DHA synergistically inhibited HCT-116 xenograft tumor growth in nude mice [35]. Overall, these results support the anti-CRC effects of ω-3 PUFAs.

Human and animal studies also support that the dietary intake of ω-3 PUFAs-rich foods, such as fish, flaxseed, and walnuts, reduces the risks of CRC. In the EPIC cohort study, the consumption of ω-3 PUFAs-rich fish was linked with lower risks of developing CRC . Stage III CRC patients who regularly consumed dark fish (≥1 time per week) had increased disease-free survival rates and lower cancer recurrence/motility risks compared to those who did not . Consistent with the human studies, the administration of a flaxseed-rich diet reduced aberrant crypt foci formation in both proximal and distal colon in an AOM-induced CRC model in rats [39]. The intake of a walnut-added diet also attenuated tumor growth in a HT29 cell-induced CRC xenograft model in mice [40]. ω-3 PUFAs could exhibit beneficial effects via regulating microbiota during CRC. The administration of EPA increased the abundance of Lactobacillus in a CAC cancer model in mice . The intake of EPA and DHA mixture could also increase the levels of Bifidobacterium, Roseburia, and Lactobacillus in humans [41]. Though more studies are needed to determine the extent to which food components, besides the ω-3 PUFAs, contribute to the observed anti-CRC effects, these results further support the beneficial effects of ω-3 PUFAs on CRC.

Though many studies support the beneficial effects of ω-3 PUFAs on CRC, there are inconsistent results from animal and human studies. Some reports, in fact, have shown that ω-3 PUFAs had no effect [42][43] or even detrimental effects on the development of CRC [44][45] (Table 1–2). The Health Professionals Follow-Up Study (HPFS) and Nurses' Health Study (NHS) cohort studies showed that ω-3 PUFA intake had no effect on overall CRC risks, and even increased distal colon cancer risk in certain individuals. The supplementation of ω-3 PUFAs had no effect on the recurrence or survival rate in stage III colon cancer patients [46]. Moreover, in a randomized, double-blind, placebo-controlled clinical trial, compared with saline infusion, intravenous infusions of ω-3 PUFAs worsened the infectious complications in CRC patients undergoing colon resection. Other postoperative complications were also reported in CRC patients who received ω-3 PUFAs after surgery [47]. Animal studies also showed that the treatment of fish oil exacerbated Helicobacter hepaticus-induced colitis and adenocarcinoma in SMAD3-deficient mice [45]. These inconsistent results make it difficult to effectively implement ω-3 PUFAs to reduce the risks of CRC.

Table 2. Preclinical studies of ω-3 PUFA supplementation for the prevention/treatment of CRC.

Model

Species

ω-3 PUFA treatment

Dose

Duration

Control treatment

Results

Reference

ApcMin/+ mouse

C57BL/6

mouse

Fish oil

12% in diet

10 weeks

Standard diet with soybean oil

↓ intestinal polyp growth

Notarnicola et al., 2017 [25]

ApcMin/+

mouse

C57BL/6

mouse

EPA-FFA

2.5% or 5% in diet

12 weeks

AIN-93G diet with soybean oil

↓ polyp number and load in both small intestine and colon.

Fini et al., 2010 [26]

ApcMin/+ mouse

C57BL/6

mouse

Endogenous ω-3 PUFA synthesis by transgene of fat-1

20 weeks

ApcMin/+ mice

on standard diet with safflower oil

↓ intestinal polyposis

Han et al., 2016 [30]

AOM/DSS-induced CRC model

C57BL/6

mouse

Endogenous ω-3 PUFA synthesis by transgene of fat-1

16 weeks

Wild‐type mice on standard diet

↓ Tumor number

Han et al., 2016 [31]

AOM/DSS-induced CRC model

C57BL/6

mouse

Endogenous ω-3 PUFA synthesis by transgene of fat-1

11 weeks

Wild‐type mice on AIN-93G diet with safflower oil

↓ incidence and growth rate

Nowak et al., 2007 [32]

AOM/DSS-induced CRC model

C57BL/6

mouse

EPA-FFA

1% in diet

15 weeks

AIN-93G diet with corn oil

↓ tumor multiplicity, incidence and maximum tumor size

Piazzi et al., 2014 [27]

DMH-induced CRC model

Wistar rat

Fish oil

18% in diet

36 weeks

AIN-93G diet with soybean oil

↓ number of aberrant crypt foci;

↓ incidence of adenoma

Moreira et al., 2009 [28]

AOM-induced CRC model

F344 rat

Fish oil

10% in diet

26 weeks

AIN-93G diet with mixed lipids

↓ colon tumor incidence and multiplicity

Reddy et al., 2005 [29]

MC38 cell-based xenograft

model

C57BL/6

mouse

DHASCO

Algae oil

8% in diet

5 weeks

AIN-93G diet with corn oil

↓ tumor volume and weight

Wang et al., 2016 [33]

SW620 cell-based xenograft

model

Nude mouse

Fish oil

12% by calorie

6 weeks

Standard diet

↓ tumor growth and less aggressive

Bathen et al., 2008 [34]

HCT116 cell-based xenograft

model

Nude mouse

DHA

10mg/kg

every other day for 13 days

Ethanol

↓ tumor size

Jeong et al., 2019 [35]

HCT116 cell-based xenograft

model

Nude mouse

DHA

3% in diet

14 days

Standard diet with sunflower oil

↓ tumor growth

Fluckiger et al., 2016 [36]

H. hepaticus-induced

CRC

model

SMAD3

deficiency

mouse

Fish oil

6% in diet

12 weeks

AIN-93G diet with corn oil

↑ adenocarcinoma formation

Woodworth et al., 2010 [45]

Abbreviations: AIN, American Institute of Nutrition; AOM, azoxymethane; DSS, dextran sodium sulfate; DMH, 1,2-Dimethylhydrazine; i.p. intraperitoneal; SMAD3, mothers against decapentaplegic homolog 3.

2.2. Effects of ω-3 PUFAs on IBD

IBD, which is characterized by chronic inflammation in intestinal tissues, severely impacts the quality of life of the patients. Symptoms include abdominal pain, vomiting, diarrhea, and rectal bleeding. The incidence and prevalence of IBD have risen dramatically in recent decades: In 2015, ~1.3% of US adults (3 million) were estimated to be diagnosed with IBD [48], representing a 50% increase from 1999 (2 million) [49]. To date, there is no cure for IBD, and the current anti-IBD treatments could lead to serious side effects, including infection, bone marrow dysfunction, and organ dysfunction [50]. Therefore, it is important to develop novel preventive strategies to reduce the risks of IBD.

Human and animal studies support the beneficial effects of ω-3 PUFAs on the development of IBD. The intake of fish oil reduced the abundance and activity of cytotoxic NK cells and improved the disease condition in IBD patients [51]. Fish oil also decreased disease activity index and reduced neutrophil infiltration in ulcerative colitis (UC, a subtype of IBD) patients [52][53]. In animal models, ω-3 PUFAs suppressed T cell-transplantation-induced colitis in severe combined immunodeficient (SCID) mice [54]. The treatment of a ω-3 PUFA (using linseed oil)-rich diet reduced the incidence of ovalbumin-induced allergic diarrhea in a food allergy mouse model [55]. The intake of ω-3 PUFAs, especially the EPA, reduced tissue damage and IBD-associated diarrhea, bloody stools, and weight loss in dextran sodium sulfate (DSS)-induced colitis models in mice and rats [56][57][58]. In ischemia-reperfusion (IR) rats, the intake of ω-3 PUFA-attenuated IR-induced mucosal injury in intestine [59]. In addition to the nutritional intervention of ω-3 PUFAs, fat-1 transgenic mice, which have higher tissue levels of ω-3 PUFAs, have been shown to exhibit reduced colonic inflammation in DSS- or 2,4,6-trinitrobenzenesulfonic acid (TNBS)-induced colitis . ω-3 PUFAs mainly exhibit beneficial effects via regulating immune cell infiltration during IBD. The administration of ω-3 PUFAs reduced the colonic infiltration of neutrophils [53][58], macrophages[62], T cells [54], and NK cells [51] in IBD mice and patients. Moreover, ω-3 PUFAs have been shown to decrease proinflammatory cytokines (TNF-α, IL-12, IL-1β, iNOS, and/or IL-6), enhance epithelial barrier function, upregulate antioxidative enzymes, and reduce lipid oxidation-derived compounds [54][57][58][59][60][61], and therefore inhibit the development of IBD in mice or rats.

There are also inconsistent reports, which have shown that ω-3 PUFAs have no effect or even adverse effects on IBD. In randomized, placebo-controlled trials, ω-3 PUFAs intake has had no effect in improving the recovery of colitis [63][64], and has even enhanced disease activity in UC patients [65]. Moreover, ω-3 PUFAs had no effect on either chemotherapy-induced enterocolitis in acute myeloid leukemia (AML) patients [66] or type 2 diabetes-induced duodenal inflammation in obesity patient [67]. In animal models, the treatment of fish oil has had little effect on DSS- or TNBS-induced colitis in rats [68][69], and has exacerbated the DSS-induced colitis in mice [70]. ω-3 PUFAs have also been shown to exaggerate chemotherapy (5-fluorouracil)-induced small intestine damage in rats [71].

2.3. Potential Reasons for the Mixed Results of ω-3 PUFAs

Overall, the effects of ω-3 PUFAs on CRC and IBD are controversial, making it difficult to effectively use ω-3 PUFAs for disease prevention. There are several possible reasons for the mixed results in ω-3 PUFA studies.

  1. Both CRC and IBD are highly heterogeneous diseases, and previous studies have shown that ω-3 PUFAs have varied effects on different types of diseases. The plasma level of ω-3 PUFAs was negatively associated with the risks of proximal colon cancer, but with not distal colon cancer or overall CRC risk . The consumption of ω-3 PUFAs decreased the risks of developing rectal cancer but increased the risks of developing distal colon cancer in men . The administration of fish oil reduced the aberrant crypt foci and adenoma incidence, but not the carcinoma incidence, in a DMH-induced CRC model in rats . It is feasible that ω-3 PUFAs target some specific types of colon carcinogenesis or inflammation, which remains to be better defined.
  2. Interindividual genetic variations could also influence the effects of ω-3 PUFAs on CRC and IBD. Many human studies have demonstrated significant interindividual variations in response to ω-3 PUFAs [72][73][74][75][76], which has made it difficult to confirm the efficacy of ω-3 PUFAs. The continuation of the current ω-3 PUFA research paradigms that neglect interindividual variation can be expected to keep generating mixed results and to fail to clarify their effects . Notably, recent research supports that ω-3 PUFA-metabolizing enzymes contribute to the biological actions of ω-3 PUFAs. A recent study showed that there is a high degree of interindividual variability in metabolizing ω-3 PUFAs to generate lipid metabolites . In addition, many studies support the critical roles of ω-3 lipid metabolizing enzymes in the activities of ω-3 PUFAs. For example, Dwyer et al.
  3. [75]showed that a diet rich in ω-3 PUFAs decreased, while a diet rich in ω-6 PUAFs increased, the risks of atherosclerosis in the subpopulation carrying a specific 5-LOX genotype but not in the general population. Other studies have also supported that polymorphism in genes encoding lipid-metabolizing genes affect the effects of ω-3 PUFAs on CRC. Notably, in a population-based case-control study, lower DHA consumption is linked to increased CRC risk in individuals with polymorphic variants in the PTGS1 gen[74]The ω-3 PUFAs consumption only increased disease-free survival rate in CRC patients with upregulation of the PTGS2 gene [77]. These results emphasize the need to better understand the roles of lipid metabolism in the actions of ω-3 PUFAs.
  4. Contamination and impurities in medication, supplements, and products can potentially compromise the protective effects of ω-3 PUFAs in clinical applications. ω-3 PUFAs are highly unstable and are easily oxidized. Oxidized ω-3 PUFAs release lipid peroxidation/oxidative products, which are cytotoxic and genotoxic to colonic cells [78][79]. Moreover, persistent organic pollutants (POPs) and foreign contaminations in fish oil supplements could exacerbate the colon carcinogenesis by stimulating aberrant crypt foci formation in rats [80]. The use of high-quality ω-3 PUFAs is critical in future human and animal studies to exclude the potential adverse effects from lipid oxidative products and contaminations. In addition, multiple studies have shown that the beneficial effects of ω-3 PUFAs, including anti-inflammation [81][82], anti-atherosclerosis [83], and anti-metastasis [84] effects, are dose-dependent. More studies are needed to determine the optimal dose and treatment time to maximize the beneficial effect of ω-3 PUFAs and to establish the official recommended daily intake for the general public and for CRC

References

  1. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2018, 68, 394-424, doi:10.3322/caac.21492.
  2. Key, T.J.; Allen, N.E.; Spencer, E.A.; Travis, R.C. The effect of diet on risk of cancer. Lancet (London, England) 2002, 360, 861-868, doi:10.1016/S0140-6736(02)09958-0.
  3. Sasazuki, S.; Inoue, M.; Iwasaki, M.; Sawada, N.; Shimazu, T.; Yamaji, T.; Takachi, R.; Tsugane, S. Intake of n-3 and n-6 polyunsaturated fatty acids and development of colorectal cancer by subsite: Japan Public Health Center-based prospective study. Int J Cancer 2011, 129, 1718-1729, doi:10.1002/ijc.25802.
  4. Murff, H.J.; Shrubsole, M.J.; Cai, Q.; Smalley, W.E.; Dai, Q.; Milne, G.L.; Ness, R.M.; Zheng, W. Dietary intake of PUFAs and colorectal polyp risk. Am J Clin Nutr 2012, 95, 703-712, doi:10.3945/ajcn.111.024000.
  5. Kim, S.; Sandler, D.P.; Galanko, J.; Martin, C.; Sandler, R.S. Intake of polyunsaturated fatty acids and distal large bowel cancer risk in whites and African Americans. Am J Epidemiol 2010, 171, 969-979, doi:10.1093/aje/kwq032.
  6. Hall, M.N.; Chavarro, J.E.; Lee, I.M.; Willett, W.C.; Ma, J. A 22-year prospective study of fish, n-3 fatty acid intake, and colorectal cancer risk in men. Cancer Epidemiol Biomarkers Prev 2008, 17, 1136-1143, doi:10.1158/1055-9965.EPI-07-2803.
  7. Schloss, I.; Kidd, M.S.; Tichelaar, H.Y.; Young, G.O.; O'Keefe, S.J. Dietary factors associated with a low risk of colon cancer in coloured west coast fishermen. S Afr Med J 1997, 87, 152-158.
  8. Pot, G.K.; Geelen, A.; van Heijningen, E.M.; Siezen, C.L.; van Kranen, H.J.; Kampman, E. Opposing associations of serum n-3 and n-6 polyunsaturated fatty acids with colorectal adenoma risk: an endoscopy-based case-control study. Int J Cancer 2008, 123, 1974-1977, doi:10.1002/ijc.23729.
  9. MacLean, C.H.; Newberry, S.J.; Mojica, W.A.; Khanna, P.; Issa, A.M.; Suttorp, M.J.; Lim, Y.W.; Traina, S.B.; Hilton, L.; Garland, R., et al. Effects of omega-3 fatty acids on cancer risk: a systematic review. JAMA 2006, 295, 403-415, doi:10.1001/jama.295.4.403.
  10. Rose, D.P.; Connolly, J.M. Omega-3 fatty acids as cancer chemopreventive agents. Pharmacol Ther 1999, 83, 217-244.
  11. Serhan, C.N.; Petasis, N.A. Resolvins and protectins in inflammation resolution. Chem Rev 2011, 111, 5922-5943, doi:10.1021/cr100396c.
  12. Sapieha, P.; Stahl, A.; Chen, J.; Seaward, M.R.; Willett, K.L.; Krah, N.M.; Dennison, R.J.; Connor, K.M.; Aderman, C.M.; Liclican, E., et al. 5-Lipoxygenase metabolite 4-HDHA is a mediator of the antiangiogenic effect of omega-3 polyunsaturated fatty acids. Sci Transl Med 2011, 3, 69ra12, doi:10.1126/scitranslmed.3001571.
  13. Bagga, D.; Wang, L.; Farias-Eisner, R.; Glaspy, J.A.; Reddy, S.T. Differential effects of prostaglandin derived from omega-6 and omega-3 polyunsaturated fatty acids on COX-2 expression and IL-6 secretion. Proc Natl Acad Sci U S A 2003, 100, 1751-1756, doi:10.1073/pnas.0334211100.
  14. Nording, M.L.; Yang, J.; Georgi, K.; Hegedus Karbowski, C.; German, J.B.; Weiss, R.H.; Hogg, R.J.; Trygg, J.; Hammock, B.D.; Zivkovic, A.M. Individual variation in lipidomic profiles of healthy subjects in response to omega-3 Fatty acids. PLoS One 2013, 8, e76575, doi:10.1371/journal.pone.0076575.
  15. Simopoulos, A.P. Genetic variants in the metabolism of omega-6 and omega-3 fatty acids: their role in the determination of nutritional requirements and chronic disease risk. Exp Biol Med (Maywood) 2010, 235, 785-795, doi:10.1258/ebm.2010.009298.
  16. Zeisel, S.H.; Waterland, R.A.; Ordovas, J.M.; Muoio, D.M.; Jia, W.; Fodor, A. Highlights of the 2012 Research Workshop: Using nutrigenomics and metabolomics in clinical nutrition research. JPEN J Parenter Enteral Nutr 2013, 37, 190-200, doi:10.1177/0148607112462401.
  17. Geelen, A.; Schouten, J.M.; Kamphuis, C.; Stam, B.E.; Burema, J.; Renkema, J.M.S.; Bakker, E.-J.; van't Veer, P.; Kampman, E. Fish Consumption, n-3 Fatty Acids, and Colorectal Cancer: A Meta-Analysis of Prospective Cohort Studies. American Journal of Epidemiology 2007, 166, 1116-1125, doi:10.1093/aje/kwm197.
  18. Kantor, E.D.; Lampe, J.W.; Peters, U.; Vaughan, T.L.; White, E. Long-chain omega-3 polyunsaturated fatty acid intake and risk of colorectal cancer. Nutr Cancer 2014, 66, 716-727, doi:10.1080/01635581.2013.804101.
  19. Aglago, E.K.; Huybrechts, I.; Murphy, N.; Casagrande, C.; Nicolas, G.; Pischon, T.; Fedirko, V.; Severi, G.; Boutron-Ruault, M.C.; Fournier, A., et al. Consumption of Fish and Long-chain n-3 Polyunsaturated Fatty Acids Is Associated With Reduced Risk of Colorectal Cancer in a Large European Cohort. Clin Gastroenterol Hepatol 2020, 18, 654-666 e656, doi:10.1016/j.cgh.2019.06.031.
  20. West, N.J.; Clark, S.K.; Phillips, R.K.; Hutchinson, J.M.; Leicester, R.J.; Belluzzi, A.; Hull, M.A. Eicosapentaenoic acid reduces rectal polyp number and size in familial adenomatous polyposis. Gut 2010, 59, 918-925, doi:10.1136/gut.2009.200642.
  21. Van Blarigan, E.L.; Fuchs, C.S.; Niedzwiecki, D.; Ye, X.; Zhang, S.; Song, M.; Saltz, L.B.; Mayer, R.J.; Mowat, R.B.; Whittom, R., et al. Marine omega-3 Polyunsaturated Fatty Acid and Fish Intake after Colon Cancer Diagnosis and Survival: CALGB 89803 (Alliance). Cancer Epidemiol Biomarkers Prev 2018, 27, 438-445, doi:10.1158/1055-9965.EPI-17-0689.
  22. Cockbain, A.J.; Volpato, M.; Race, A.D.; Munarini, A.; Fazio, C.; Belluzzi, A.; Loadman, P.M.; Toogood, G.J.; Hull, M.A. Anticolorectal cancer activity of the omega-3 polyunsaturated fatty acid eicosapentaenoic acid. Gut 2014, 63, 1760-1768, doi:10.1136/gutjnl-2013-306445.
  23. Song, M.; Chan, A.T.; Fuchs, C.S.; Ogino, S.; Hu, F.B.; Mozaffarian, D.; Ma, J.; Willett, W.C.; Giovannucci, E.L.; Wu, K. Dietary intake of fish, omega-3 and omega-6 fatty acids and risk of colorectal cancer: A prospective study in U.S. men and women. Int J Cancer 2014, 135, 2413-2423, doi:10.1002/ijc.28878.
  24. Bakker, N.; van den Helder, R.S.; Stoutjesdijk, E.; van Pelt, J.; Houdijk, A.P.J. Effects of perioperative intravenous omega-3 fatty acids in colon cancer patients: a randomized, double-blind, placebo-controlled clinical trial. Am J Clin Nutr 2020, 111, 385-395, doi:10.1093/ajcn/nqz281.
  25. Notarnicola, M.; Tutino, V.; De Nunzio, V.; Dituri, F.; Caruso, M.G.; Giannelli, G. Dietary omega-3 Polyunsaturated Fatty Acids Inhibit Tumor Growth in Transgenic Apc(Min/+) Mice, Correlating with CB1 Receptor Up-Regulation. Int J Mol Sci 2017, 18, doi:10.3390/ijms18030485.
  26. Fini, L.; Piazzi, G.; Ceccarelli, C.; Daoud, Y.; Belluzzi, A.; Munarini, A.; Graziani, G.; Fogliano, V.; Selgrad, M.; Garcia, M., et al. Highly purified eicosapentaenoic acid as free fatty acids strongly suppresses polyps in Apc(Min/+) mice. Clin Cancer Res 2010, 16, 5703-5711, doi:10.1158/1078-0432.CCR-10-1990.
  27. Piazzi, G.; D'Argenio, G.; Prossomariti, A.; Lembo, V.; Mazzone, G.; Candela, M.; Biagi, E.; Brigidi, P.; Vitaglione, P.; Fogliano, V., et al. Eicosapentaenoic acid free fatty acid prevents and suppresses colonic neoplasia in colitis-associated colorectal cancer acting on Notch signaling and gut microbiota. Int J Cancer 2014, 135, 2004-2013, doi:10.1002/ijc.28853.
  28. Moreira, A.P.; Sabarense, C.M.; Dias, C.M.; Lunz, W.; Natali, A.J.; Gloria, M.B.; Peluzio, M.C. Fish oil ingestion reduces the number of aberrant crypt foci and adenoma in 1,2-dimethylhydrazine-induced colon cancer in rats. Braz J Med Biol Res 2009, 42, 1167-1172, doi:10.1590/s0100-879x2009001200008.
  29. Reddy, B.S.; Patlolla, J.M.; Simi, B.; Wang, S.H.; Rao, C.V. Prevention of colon cancer by low doses of celecoxib, a cyclooxygenase inhibitor, administered in diet rich in omega-3 polyunsaturated fatty acids. Cancer Res 2005, 65, 8022-8027, doi:10.1158/0008-5472.CAN-05-0212.
  30. Han, Y.M.; Park, J.M.; Cha, J.Y.; Jeong, M.; Go, E.J.; Hahm, K.B. Endogenous conversion of omega-6 to omega-3 polyunsaturated fatty acids in fat-1 mice attenuated intestinal polyposis by either inhibiting COX-2/beta-catenin signaling or activating 15-PGDH/IL-18. Int J Cancer 2016, 138, 2247-2256, doi:10.1002/ijc.29956.
  31. Han, Y.M.; Jeong, M.; Park, J.M.; Kim, M.Y.; Go, E.J.; Cha, J.Y.; Kim, K.J.; Hahm, K.B. The omega-3 polyunsaturated fatty acids prevented colitis-associated carcinogenesis through blocking dissociation of beta-catenin complex, inhibiting COX-2 through repressing NF-kappaB, and inducing 15-prostaglandin dehydrogenase. Oncotarget 2016, 7, 63583-63595, doi:10.18632/oncotarget.11544.
  32. Nowak, J.; Weylandt, K.H.; Habbel, P.; Wang, J.; Dignass, A.; Glickman, J.N.; Kang, J.X. Colitis-associated colon tumorigenesis is suppressed in transgenic mice rich in endogenous n-3 fatty acids. Carcinogenesis 2007, 28, 1991-1995, doi:10.1093/carcin/bgm166.
  33. Wang, W.; Yang, J.; Nimiya, Y.; Lee, K.S.S.; Sanidad, K.; Qi, W.; Sukamtoh, E.; Park, Y.; Liu, Z.; Zhang, G. omega-3 Polyunsaturated fatty acids and their cytochrome P450-derived metabolites suppress colorectal tumor development in mice. J Nutr Biochem 2017, 48, 29-35, doi:10.1016/j.jnutbio.2017.06.006.
  34. Bathen, T.F.; Holmgren, K.; Lundemo, A.G.; Hjelstuen, M.H.; Krokan, H.E.; Gribbestad, I.S.; Schonberg, S.A. Omega-3 fatty acids suppress growth of SW620 human colon cancer xenografts in nude mice. Anticancer Res 2008, 28, 3717-3723.
  35. Jeong, S.; Kim, D.Y.; Kang, S.H.; Yun, H.K.; Kim, J.L.; Kim, B.R.; Park, S.H.; Na, Y.J.; Jo, M.J.; Jeong, Y.A., et al. Docosahexaenoic Acid Enhances Oxaliplatin-Induced Autophagic Cell Death via the ER Stress/Sesn2 Pathway in Colorectal Cancer. Cancers (Basel) 2019, 11, doi:10.3390/cancers11070982.
  36. Fluckiger, A.; Dumont, A.; Derangere, V.; Rebe, C.; de Rosny, C.; Causse, S.; Thomas, C.; Apetoh, L.; Hichami, A.; Ghiringhelli, F., et al. Inhibition of colon cancer growth by docosahexaenoic acid involves autocrine production of TNFalpha. Oncogene 2016, 35, 4611-4622, doi:10.1038/onc.2015.523.
  37. Hawcroft, G.; Volpato, M.; Marston, G.; Ingram, N.; Perry, S.L.; Cockbain, A.J.; Race, A.D.; Munarini, A.; Belluzzi, A.; Loadman, P.M., et al. The omega-3 polyunsaturated fatty acid eicosapentaenoic acid inhibits mouse MC-26 colorectal cancer cell liver metastasis via inhibition of PGE2-dependent cell motility. Br J Pharmacol 2012, 166, 1724-1737, doi:10.1111/j.1476-5381.2012.01882.x.
  38. Gutt, C.N.; Brinkmann, L.; Mehrabi, A.; Fonouni, H.; Muller-Stich, B.P.; Vetter, G.; Stein, J.M.; Schemmer, P.; Buchler, M.W. Dietary omega-3-polyunsaturated fatty acids prevent the development of metastases of colon carcinoma in rat liver. Eur J Nutr 2007, 46, 279-285, doi:10.1007/s00394-007-0662-y.
  39. Williams, D.; Verghese, M.; Walker, L.T.; Boateng, J.; Shackelford, L.; Chawan, C.B. Flax seed oil and flax seed meal reduce the formation of aberrant crypt foci (ACF) in azoxymethane-induced colon cancer in Fisher 344 male rats. Food Chem Toxicol 2007, 45, 153-159, doi:10.1016/j.fct.2006.08.014.
  40. Tsoukas, M.A.; Ko, B.J.; Witte, T.R.; Dincer, F.; Hardman, W.E.; Mantzoros, C.S. Dietary walnut suppression of colorectal cancer in mice: Mediation by miRNA patterns and fatty acid incorporation. J Nutr Biochem 2015, 26, 776-783, doi:10.1016/j.jnutbio.2015.02.009.
  41. Watson, H.; Mitra, S.; Croden, F.C.; Taylor, M.; Wood, H.M.; Perry, S.L.; Spencer, J.A.; Quirke, P.; Toogood, G.J.; Lawton, C.L., et al. A randomised trial of the effect of omega-3 polyunsaturated fatty acid supplements on the human intestinal microbiota. Gut 2018, 67, 1974-1983, doi:10.1136/gutjnl-2017-314968.
  42. Akedo, I.; Ishikawa, H.; Nakamura, T.; Kimura, K.; Takeyama, I.; Suzuki, T.; Kameyama, M.; Sato, S.; Nakamura, T.; Matsuzawa, Y., et al. Three cases with familial adenomatous polyposis diagnosed as having malignant lesions in the course of a long-term trial using docosahexanoic acid (DHA)-concentrated fish oil capsules. Jpn J Clin Oncol 1998, 28, 762-765, doi:DOI 10.1093/jjco/28.12.762.
  43. Kobayashi, M.; Tsubono, Y.; Otani, T.; Hanaoka, T.; Sobue, T.; Tsugane, S. Fish, long-chain n-3 polyunsaturated fatty acids, and risk of colorectal cancer in middle-aged Japanese: the JPHC study. Nutr Cancer 2004, 49, 32-40, doi:10.1207/s15327914nc4901_5.
  44. Stern, M.C.; Butler, L.M.; Corral, R.; Joshi, A.D.; Yuan, J.M.; Koh, W.P.; Yu, M.C. Polyunsaturated fatty acids, DNA repair single nucleotide polymorphisms and colorectal cancer in the Singapore Chinese Health Study. J Nutrigenet Nutrigenomics 2009, 2, 273-279, doi:10.1159/000308467.
  45. Woodworth, H.L.; McCaskey, S.J.; Duriancik, D.M.; Clinthorne, J.F.; Langohr, I.M.; Gardner, E.M.; Fenton, J.I. Dietary fish oil alters T lymphocyte cell populations and exacerbates disease in a mouse model of inflammatory colitis. Cancer Res 2010, 70, 7960-7969, doi:10.1158/0008-5472.CAN-10-1396.
  46. Lee, J.Y.; Sim, T.B.; Lee, J.E.; Na, H.K. Chemopreventive and Chemotherapeutic Effects of Fish Oil derived Omega-3 Polyunsaturated Fatty Acids on Colon Carcinogenesis. Clin Nutr Res 2017, 6, 147-160, doi:10.7762/cnr.2017.6.3.147.
  47. Chen, Y.; Liu, B.L.; Shang, B.; Chen, A.S.; Liu, S.Q.; Sun, W.; Yin, H.Z.; Yin, J.Q.; Su, Q. Nutrition support in surgical patients with colorectal cancer. World J Gastroenterol 2011, 17, 1779-1786, doi:10.3748/wjg.v17.i13.1779.
  48. Dahlhamer, J.M.; Zammitti, E.P.; Ward, B.W.; Wheaton, A.G.; Croft, J.B. Prevalence of Inflammatory Bowel Disease Among Adults Aged > 18 Years - United States, 2015. MMWR Morb Mortal Wkly Rep 2016, 65, 1166-1169, doi:10.15585/mmwr.mm6542a3.
  49. Nguyen, G.C.; Chong, C.A.; Chong, R.Y. National estimates of the burden of inflammatory bowel disease among racial and ethnic groups in the United States. J Crohns Colitis 2014, 8, 288-295, doi:10.1016/j.crohns.2013.09.001.
  50. Cohen, B.L.; Sachar, D.B. Update on anti-tumor necrosis factor agents and other new drugs for inflammatory bowel disease. BMJ (Clinical research ed.) 2017, 357, j2505, doi:10.1136/bmj.j2505.
  51. Almallah, Y.Z.; Richardson, S.; O'Hanrahan, T.; Mowat, N.A.; Brunt, P.W.; Sinclair, T.S.; Ewen, S.; Heys, S.D.; Eremin, O. Distal procto-colitis, natural cytotoxicity, and essential fatty acids. Am J Gastroenterol 1998, 93, 804-809, doi:10.1111/j.1572-0241.1998.229_a.x.
  52. Aslan, A.; Triadafilopoulos, G. Fish oil fatty acid supplementation in active ulcerative colitis: a double-blind, placebo-controlled, crossover study. Am J Gastroenterol 1992, 87, 432-437.
  53. McCall, T.B.; O'Leary, D.; Bloomfield, J.; O'Morain, C.A. Therapeutic potential of fish oil in the treatment of ulcerative colitis. Aliment Pharmacol Ther 1989, 3, 415-424, doi:10.1111/j.1365-2036.1989.tb00232.x.
  54. Whiting, C.V.; Bland, P.W.; Tarlton, J.F. Dietary n-3 polyunsaturated fatty acids reduce disease and colonic proinflammatory cytokines in a mouse model of colitis. Inflamm Bowel Dis 2005, 11, 340-349, doi:10.1097/01.mib.0000164016.98913.7c.
  55. Kunisawa, J.; Arita, M.; Hayasaka, T.; Harada, T.; Iwamoto, R.; Nagasawa, R.; Shikata, S.; Nagatake, T.; Suzuki, H.; Hashimoto, E., et al. Dietary omega3 fatty acid exerts anti-allergic effect through the conversion to 17,18-epoxyeicosatetraenoic acid in the gut. Sci Rep 2015, 5, 9750, doi:10.1038/srep09750.
  56. Sharma, M.; Kaur, R.; Kaushik, K.; Kaushal, N. Redox modulatory protective effects of omega-3 fatty acids rich fish oil against experimental colitis. Toxicol Mech Methods 2019, 29, 244-254, doi:10.1080/15376516.2018.1553220.
  57. Varnalidis, I.; Ioannidis, O.; Karamanavi, E.; Ampas, Z.; Poutahidis, T.; Taitzoglou, I.; Paraskevas, G.; Botsios, D. Omega 3 fatty acids supplementation has an ameliorative effect in experimental ulcerative colitis despite increased colonic neutrophil infiltration. Rev Esp Enferm Dig 2011, 103, 511-518.
  58. Morin, C.; Blier, P.U.; Fortin, S. MAG-EPA reduces severity of DSS-induced colitis in rats. Am J Physiol Gastrointest Liver Physiol 2016, 310, G808-821, doi:10.1152/ajpgi.00136.2015.
  59. Brahmbhatt, V.; Oliveira, M.; Briand, M.; Perrisseau, G.; Bastic Schmid, V.; Destaillats, F.; Pace-Asciak, C.; Benyacoub, J.; Bosco, N. Protective effects of dietary EPA and DHA on ischemia-reperfusion-induced intestinal stress. J Nutr Biochem 2013, 24, 104-111, doi:10.1016/j.jnutbio.2012.02.014.
  60. Hudert, C.A.; Weylandt, K.H.; Lu, Y.; Wang, J.; Hong, S.; Dignass, A.; Serhan, C.N.; Kang, J.X. Transgenic mice rich in endogenous omega-3 fatty acids are protected from colitis. Proc Natl Acad Sci U S A 2006, 103, 11276-11281, doi:10.1073/pnas.0601280103.
  61. Yum, H.W.; Kang, J.X.; Hahm, K.B.; Surh, Y.J. Constitutive omega-3 fatty acid production in fat-1 transgenic mice and docosahexaenoic acid administration to wild type mice protect against 2,4,6-trinitrobenzene sulfonic acid-induced colitis. Biochem Biophys Res Commun 2017, 487, 847-855, doi:10.1016/j.bbrc.2017.04.140.
  62. Matsunaga, H.; Hokari, R.; Kurihara, C.; Okada, Y.; Takebayashi, K.; Okudaira, K.; Watanabe, C.; Komoto, S.; Nakamura, M.; Tsuzuki, Y., et al. Omega-3 polyunsaturated fatty acids ameliorate the severity of ileitis in the senescence accelerated mice (SAM)P1/Yit mice model. Clin Exp Immunol 2009, 158, 325-333, doi:10.1111/j.1365-2249.2009.04020.x.
  63. Turner, D.; Steinhart, A.H.; Griffiths, A.M. Omega 3 fatty acids (fish oil) for maintenance of remission in ulcerative colitis. Cochrane Database Syst Rev 2007, 10.1002/14651858.CD006443.pub2, CD006443, doi:10.1002/14651858.CD006443.pub2.
  64. Barbosa, D.S.; Cecchini, R.; El Kadri, M.Z.; Rodriguez, M.A.; Burini, R.C.; Dichi, I. Decreased oxidative stress in patients with ulcerative colitis supplemented with fish oil omega-3 fatty acids. Nutrition 2003, 19, 837-842, doi:10.1016/s0899-9007(03)00162-x.
  65. Dichi, I.; Frenhane, P.; Dichi, J.B.; Correa, C.R.; Angeleli, A.Y.; Bicudo, M.H.; Rodrigues, M.A.; Victoria, C.R.; Burini, R.C. Comparison of omega-3 fatty acids and sulfasalazine in ulcerative colitis. Nutrition 2000, 16, 87-90, doi:10.1016/s0899-9007(99)00231-2.
  66. Bukki, J.; Stanga, Z.; Tellez, F.B.; Duclos, K.; Kolev, M.; Krahenmann, P.; Pabst, T.; Iff, S.; Juni, P. Omega-3 poly-unsaturated fatty acids for the prevention of severe neutropenic enterocolitis in patients with acute myeloid leukemia. Nutr Cancer 2013, 65, 834-842, doi:10.1080/01635581.2013.801998.
  67. Labonte, M.E.; Couture, P.; Tremblay, A.J.; Hogue, J.C.; Lemelin, V.; Lamarche, B. Eicosapentaenoic and docosahexaenoic acid supplementation and inflammatory gene expression in the duodenum of obese patients with type 2 diabetes. Nutr J 2013, 12, 98, doi:10.1186/1475-2891-12-98.
  68. Vieira de Barros, K.; Gomes de Abreu, G.; Xavier, R.A.; Real Martinez, C.A.; Ribeiro, M.L.; Gambero, A.; de Oliveira Carvalho, P.; Silveira, V.L. Effects of a high fat or a balanced omega 3/omega 6 diet on cytokines levels and DNA damage in experimental colitis. Nutrition 2011, 27, 221-226, doi:10.1016/j.nut.2009.11.014.
  69. Shoda, R.; Matsueda, K.; Yamato, S.; Umeda, N. Therapeutic efficacy of N-3 polyunsaturated fatty acid in experimental Crohn's disease. J Gastroenterol 1995, 30 Suppl 8, 98-101.
  70. Matsunaga, H.; Hokari, R.; Kurihara, C.; Okada, Y.; Takebayashi, K.; Okudaira, K.; Watanabe, C.; Komoto, S.; Nakamura, M.; Tsuzuki, Y., et al. Omega-3 fatty acids exacerbate DSS-induced colitis through decreased adiponectin in colonic subepithelial myofibroblasts. Inflamm Bowel Dis 2008, 14, 1348-1357, doi:10.1002/ibd.20491.
  71. Torres, D.M.; Tooley, K.L.; Butler, R.N.; Smith, C.L.; Geier, M.S.; Howarth, G.S. Lyprinol only partially improves indicators of small intestinal integrity in a rat model of 5-fluorouracil-induced mucositis. Cancer Biol Ther 2008, 7, 295-302, doi:10.4161/cbt.7.2.5332.
  72. Fradet, V.; Cheng, I.; Casey, G.; Witte, J.S. Dietary omega-3 fatty acids, cyclooxygenase-2 genetic variation, and aggressive prostate cancer risk. Clin Cancer Res 2009, 15, 2559-2566, doi:10.1158/1078-0432.ccr-08-2503.
  73. Wang, J.; John, E.; Ingles, S. 5-lipoxygenase and 5-lipoxygenase-activating protein gene polymorphisms, dietary linoleic acid, and risk for breast cancer. Cancer Epidemiol Biomarkers Prev 2008, 17, 2748 - 2754.
  74. Habermann, N.; Ulrich, C.M.; Lundgreen, A.; Makar, K.W.; Poole, E.M.; Caan, B.; Kulmacz, R.; Whitton, J.; Galbraith, R.; Potter, J.D., et al. PTGS1, PTGS2, ALOX5, ALOX12, ALOX15, and FLAP SNPs: interaction with fatty acids in colon cancer and rectal cancer. Genes Nutr 2013, 8, 115-126, doi:10.1007/s12263-012-0302-x.
  75. Dwyer, J.H.; Allayee, H.; Dwyer, K.M.; Fan, J.; Wu, H.; Mar, R.; Lusis, A.J.; Mehrabian, M. Arachidonate 5-lipoxygenase promoter genotype, dietary arachidonic acid, and atherosclerosis. N Engl J Med 2004, 350, 29-37, doi:10.1056/NEJMoa025079.
  76. Stephensen, C.B.; Armstrong, P.; Newman, J.W.; Pedersen, T.L.; Legault, J.; Schuster, G.U.; Kelley, D.; Vikman, S.; Hartiala, J.; Nassir, R., et al. ALOX5 gene variants affect eicosanoid production and response to fish oil supplementation. J Lipid Res 2011, 52, 991-1003, doi:10.1194/jlr.P012864.
  77. Song, M.; Ou, F.S.; Zemla, T.J.; Hull, M.A.; Shi, Q.; Limburg, P.J.; Alberts, S.R.; Sinicrope, F.A.; Giovannucci, E.L.; Van Blarigan, E.L., et al. Marine omega-3 fatty acid intake and survival of stage III colon cancer according to tumor molecular markers in NCCTG Phase III trial N0147 (Alliance). Int J Cancer 2019, 145, 380-389, doi:10.1002/ijc.32113.
  78. Wang, W.; Yang, H.; Johnson, D.; Gensler, C.; Decker, E.; Zhang, G. Chemistry and biology of ω-3 PUFA peroxidation-derived compounds. Prostaglandins & other lipid mediators 2017, 132, 84-91.
  79. Gueraud, F.; Tache, S.; Steghens, J.P.; Milkovic, L.; Borovic-Sunjic, S.; Zarkovic, N.; Gaultier, E.; Naud, N.; Helies-Toussaint, C.; Pierre, F., et al. Dietary polyunsaturated fatty acids and heme iron induce oxidative stress biomarkers and a cancer promoting environment in the colon of rats. Free Radic Biol Med 2015, 83, 192-200, doi:10.1016/j.freeradbiomed.2015.02.023.
  80. Hong, M.Y.; Hoh, E.; Kang, B.; DeHamer, R.; Kim, J.Y.; Lumibao, J. Fish Oil Contaminated with Persistent Organic Pollutants Induces Colonic Aberrant Crypt Foci Formation and Reduces Antioxidant Enzyme Gene Expression in Rats. J Nutr 2017, 147, 1524-1530, doi:10.3945/jn.117.251082.
  81. Ferguson, J.F.; Mulvey, C.K.; Patel, P.N.; Shah, R.Y.; Doveikis, J.; Zhang, W.; Tabita-Martinez, J.; Terembula, K.; Eiden, M.; Koulman, A., et al. Omega-3 PUFA supplementation and the response to evoked endotoxemia in healthy volunteers. Mol Nutr Food Res 2014, 58, 601-613, doi:10.1002/mnfr.201300368.
  82. Gupta, S.; Knight, A.G.; Gupta, S.; Keller, J.N.; Bruce-Keller, A.J. Saturated long-chain fatty acids activate inflammatory signaling in astrocytes. J Neurochem 2012, 120, 1060-1071, doi:10.1111/j.1471-4159.2012.07660.x.
  83. Gladine, C.; Newman, J.W.; Durand, T.; Pedersen, T.L.; Galano, J.M.; Demougeot, C.; Berdeaux, O.; Pujos-Guillot, E.; Mazur, A.; Comte, B. Lipid profiling following intake of the omega 3 fatty acid DHA identifies the peroxidized metabolites F4-neuroprostanes as the best predictors of atherosclerosis prevention. PLoS One 2014, 9, e89393, doi:10.1371/journal.pone.0089393.
  84. Rose, D.P.; Connolly, J.M.; Coleman, M. Effect of omega-3 fatty acids on the progression of metastases after the surgical excision of human breast cancer cell solid tumors growing in nude mice. Clin Cancer Res 1996, 2, 1751-1756.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , ,
View Times: 642
Revisions: 3 times (View History)
Update Date: 17 Nov 2020
1000/1000