Abstract
Colorectal cancer (CRC) is the third leading cause of cancer-related death in the world. Multiple evidence suggests that there is an association between excess fat consumption and the risk of CRC. The long chain n-3 polyunsaturated fatty acids (LC n-3 PUFA), especially eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), are essential for human health, and both in vitro and in vivo studies have shown that these fatty acids can prevent CRC development through various molecular mechanisms. These include the modulation of arachidonic acid (AA) derived prostaglandin synthesis, alteration of growth signaling pathways, arrest of the cell cycle, induction of cell apoptosis, suppression of angiogenesis and modulation of inflammatory response. Human clinical studies found that LC n-3 PUFA combined with chemotherapeutic agents can improve the efficacy of treatment and reduce the dosage of chemotherapy and associated side effects. In this review, we discuss comprehensively the anti-cancer effects of LC n-3 PUFA on CRC, with a main focus on the underlying molecular mechanisms.
Keywords: colorectal cancer, LC n-3 PUFA, EPA, DHA, molecular mechanisms
Background
Colorectal cancer (CRC) is the third most common cancer after lung and breast cancer and the fourth leading cause of cancer death worldwide.1,2 CRC is a multifactorial disease caused by the interaction of genetic and environmental factors 3 and it presents in 1 of 3 patterns: sporadic, inherited, and familial. The majority of CRC cases are sporadic and approximately 70% to 80% are derived from somatic mutations without any family history. 3 The inherited and familial causes (about 35%) of CRC are derived from germline mutations. 4 Age, poor diet, and sedentary lifestyles are considered the main environmental contributors to the disease. 5
There is considerable evidence indicating that red and processed meats, and alcoholic beverages are risk factors for CRC. However, grains, vegetables and fruits, dairy products, and fish and other seafood are linked with a decreased risk of CRC.6,7 Studies have also shown that high intakes of energy, saturated fatty acids (SFA) and sucrose are associated with increased risk of CRC; while high intakes of dietary fibers, calcium and long chain n-3 polyunsaturated fatty acids (LC n-3 PUFA) contributed to low incidence of CRC.8 -11
The onset of CRC is a complex process, and, in most cases, CRC starts with polyps occurring on the epithelial layer of the colon or rectum. These polyps may be benign (for example hyperplastic polyp), pre-malignant (eg, tubular adenoma), or malignant (eg, colorectal adenocarcinoma). 12 Several genetic, molecular, cellular, and histological changes were found to be associated with the transformation of normal epithelium to adenoma and invasive metastatic adenocarcinoma.13,14 This usually takes 10 to 15 years due to the incremental accumulation of genetic alterations.3,15 Three main causes of colorectal carcinogenesis have been identified. 16 The first one is the suppressor pathway of chromosomal instability (CIN) involving the accumulation of molecular alterations that influence oncogene activation (KRAS) and inactivation of tumor suppressor genes (DCC, APC, SMAD4, and TP53). 17 The second mechanism is the accumulation of errors in DNA replication as a result of mutations of the genes that are responsible for DNA repair (MSH2, MLH1, MSH6, PMS2, MLH3, MSH3, PISI, and Exol). This is known as the microsatellite instability pathway (MSI). 18 The third mechanism is the CpG island methylator phenotype (CIMP) pathway. This is due to the vast hyper-methylation of promoter CpG island sites that silence the tumor suppressor genes. 19 The progression of CRC is commonly associated with multiple steps such as hyperplasia of colonic crypts, colonic crypt dysplasia, adenoma, adenocarcinoma, invasion, and distant metastasis. 20
Currently available treatments for CRC are surgery, chemotherapy, radiotherapy, and molecular–targeted therapy.3,21 Surgery might be curative only when the disease is diagnosed at its early stages. Treatments for patients diagnosed at advanced stages are chemotherapy, radiotherapy, or a combination of both. However, these therapies are associated with numerous side effects leading to a significant impact on patients’ quality of life. 22 In recent years, a significant focus of cancer therapeutic research has been shifted to marine sources as they serve as a province for a range of bioactive compounds.23,24 Nutraceuticals that have little or no side effects have received considerable attention for the prevention and management of CRC and other invasive metastatic carcinomas.25,26
There is growing evidence showing that LC n-3 PUFA originated from marine oils (eg, fish oil and krill oil) have anti-CRC properties. These fatty acids have been reported to exhibit multiple anti-cancer effects on various stages of CRC from primary to tertiary, including advanced metastasis.26,27 Epidemiological studies have found that populations consuming LC n-3 PUFA-rich diets (such as fish and other seafood) have fewer cases of CRC compared to populations that consume diets containing less LC n-3 PUFA.28,29 A meta-analysis reported that a 50 g increment in the daily consumption of fish was associated with a statistically significant 4% reduction in CRC risk. 30 A recent European Prospective Investigation showed that CRC incidence decreases with increasing proportions of red blood cell membrane n-3 PUFA, particularly EPA. 31 Conversely, a diet containing a relatively high proportion of n-6 PUFA (eg, typical western style) was associated with an increased risk of inflammatory bowel disease (IBD) and colon carcinogenesis.32,33 This review discusses the beneficial effects of LC n-3 PUFA, mainly eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) and their two main marine oil sources, fish oil and krill oil, on the initiation, progression and apoptosis of CRC. A particular focus is on the molecular mechanisms underlying the anti-cancer properties of these fatty acids and their marine sources.
Methods
The databases of PubMed, PubMed Central, MEDLINE, Springer Link, and Wiley Online Library were searched using the key words of n-3 PUFA, EPA, DHA, fish oil, krill oil, colorectal cancer, molecular mechanism, signaling pathway, cell apoptosis, cell cycle, and anti-inflammatory. The inclusion criteria are defined molecular targets of LC-n3 PUFA in colorectal cancer. As there are a large number of studies available, the title and abstract were used for initial screening. Full text screening was then applied for original research and their relevant and key citations. Exclusion criteria include the preprints, conference proceedings, articles with only abstract available, and articles were not written in English.
Polyunsaturated Fatty Acids and Their Effects on Colorectal Cancer
There are two major families of polyunsaturated fatty acids (PUFA) including omega 3 (n-3 PUFA) and omega 6 (n-6 PUFA); and they are not metabolically interchangeable. 34 The members of n-3 and n-6 families are considered essential fatty acids for humans, since they cannot be synthesized de novo in the body and, therefore, must be obtained from dietary sources.27,34 Plant-based α-linolenic acid (ALA, C18:3, n-3) is known as the parent form of the LC n-3 PUFA, eicosapentaenoic acid (EPA, C20:5, n-3), docosapentaenoic acid (DPA, C22:5, n-3), and docosahexaenoic acid (DHA, C22:6, n-3), and it is an essential fatty acid. 35 ALA is commonly found in green leafy vegetables, nuts, especially in walnuts, and oils, such as flaxseed oil, soybean oil, and canola oil.36,37 After consumption, ALA can be utilized to synthesize LC n-3 PUFA by enzymes desaturases (delta-6 and 5) and elongases (2 and 5) mainly in the liver. But this process can also take place in other organs such as brain, kidney, and testicles.38,39 However, the body cannot synthesize EPA and DHA from ALA in sufficient quantities. 40 Only about 5% of the consumed ALA is converted to EPA in the body due to a lack of delta desaturases to catalyze the addition of double bonds.41,42 Therefore, these fatty acids are obtained mainly from fish (especially those from cold-water fatty fish, such as mackerel and salmon), fish products, and other seafood including oysters, mussels, and shrimps, as well as from dietary supplements.43,44 Fish oil and krill oil are the two major commercially available sources of EPA and DHA supplements.
The n-6 PUFA, linoleic acid (C18:2, n-6) is another essential fatty acid and the parental form of arachidonic acid (AA). Linoleic acid is mainly found in plant oils such as sunflower and corn oils, grains, and animal fats. Humans can readily metabolize linoleic acid to obtain AA via desaturation and elongation by the same set of enzymes as in the synthesis of EPA.27,39 In addition, AA can be obtained directly from beef, pork and eggs. 39 The LC n-3 PUFA and AA are biologically vital for the human body, because of their important roles in the phospholipid cell membrane structure, modulation of cellular signaling, membrane fluidity, cellular interaction, and lipid metabolism.27,45 EPA and AA are released from the cell membrane by the action of phospholipase enzymes, especially phospholipase A2 (PLA-2) and C (PLC) and metabolized through three main pathways of cyclo-oxygenase (COX), lipoxygenase (LOX), and cytochrome P450 (CYP450) . 46 The main metabolites are prostaglandins (PGs) and thromboxanes by COX-1 and COX-2, leukotrienes (LTs) and lipoxins by LOX, and hydroxyeicosatetraenoic acids (HETEs) and dihydroxyeicosatetraenoic acids (DHETEs) by cytochrome P450. In general, eicosanoids derived from AA are typically involved directly in the development of inflammation. Moreover, they are also involved in a range of biological process and modulate diverse physiological responses.46 -48 Prostaglandins have received more attention for their roles in modulating inflammation. It has been reported that prostaglandin-E2 (PGE-2), derived from the metabolism of AA, as an important pro-inflammatory agent, is associated with the onset and progression of several cancers through cell proliferation, angiogenesis, migration, and invasion while inhibiting apoptosis.49,50 In addition, it plays a significant role in the early stages of colorectal carcinogenesis. 51 LTB4, another metabolite of AA, apart from its pro-inflammatory action, has also been reported to stimulate cancer cell growth52,53
In contrast, prostaglandin-E3 (PGE-3), derived from EPA, is generally known for its anti-inflammatory and anti-cancer properties54,55. It has been found that PGE-3 can antagonize the effects of PGE-2, although few studies have shown discrepant results that PGE-3 causes the similar deleterious effects as PGE-2 on epithelial barrier function or promotes the proliferation of cancer cell line.56,57
In the cells, there is a competition between n-3 and n-6 PUFA for their metabolic process via COX enzymes. However, these enzymes have a higher affinity for EPA of n-3 PUFA rather than AA of n-6 PUFA, especially COX-2. 58 This leads to the formation of anti-tumorigenic PGE-3 and the reduction in the synthesis of PGE-2. In addition, LTB5 produced by LOX from EPA has shown anti-inflammatory and anti-cancer properties.59 -61 Furthermore, EPA and DHA can also produce a family of pro-resolving anti-inflammatory mediators including resolvins, protectins and maresins.62,63 The food sources, metabolism and functions of major metabolites of PUFA are summarized in Figure 1.
Figure 1.
Schematic overview of biosynthesis of long chain PUFA and actions of main metabolites from arachidonic acid verse that from EPA. High metabolism of linoleic acid to arachidonic acid contributes to cancer risk and progression through the synthesis of pro-inflammatory and pro-tumor lipid metabolites. High metabolism of α-linolenic acid to EPA and DHA reduces cancer risk and progression through the synthesis of anti-inflammatory and anti-tumor lipid metabolites.
Abbreviations: COX1/2, cyclooxygenase 1/2; LOX, lipoxygenase; PGE2/3, prostaglandin E2/E3; LTB4/5, leukotrienes B4/B5; Ras, retrovirus-associated DNA sequences; MEK, mitogen-activated protein kinase; ERK, extracellular signal-regulated kinase; Wnt, Wingless-related integration site; BLT1/BLT2, Leukotriene B4 receptor, NF-ĸB, nuclear factor kappa-light-chian-enhancer of activated B cells; FADS1/2, Fatty acid desaturase 1/2; ELOV5, fatty acid elongase 5.
The main anti-cancer properties of LC n-3 PUFA involve the modulation of COX-2 enzymatic activity, alteration of the functions of cell surface receptors and membrane characteristics, enhancement of cellular oxidative stress, and production of anti-inflammatory mediators including resolvins, protectins, and maresins.27,62 It has been reported that LC n-3 PUFA can inhibit cancer cell proliferation and reduce tumor growth through various mechanisms, including the alteration of signaling pathways involved in carcinogenesis such as angiogenesis and cell metastasis64 -67; regulation of cell cycle; as well as induction of cell apoptosis.68 -70 The characteristic functions of those proposed molecular mechanisms are summarized in Figure 2.
Figure 2.
Targeted signaling pathways by LC n-3 PUFA in relation to the death of cancer cells. (A) represents the inhibition of EGFR activation and the inhibition of its downstream signaling pathways. (B) represents the suppression of phosphorylation of the RAS/MEK pathway. (C) shows the inhibition of the PI3K/AKT/mTOR signaling cascade. Phosphorylated AKT activates mTOR, which further activates the transcription factors necessary for the transcription of genes essential for cell proliferation, metastasis, angiogenesis. (D) shows an intrinsic apoptosis pathway that activates through the up-regulation of pro-apoptotic Bcl-2 protein and down-regulation of anti-apoptotic Bcl-2 protein that leads to alteration of MMP to release cytochrome c. Cytochrome c forms an apoptosome with the binding of Apaf and caspase-9 to further activate caspase-3 to induce apoptosis. (E) represents the intrinsic apoptosis pathway through the endoplasmic reticulum. (F) represents the extrinsic pathway of apoptosis showing Eas/FasL interaction and DISC formation leading to caspase-8 activation. (G) represents the suppression of NF-қB that upregulates the transcription of genes involved in inflammation, angiogenesis, and metastasis. (H) represents all phases of the cell cycle together with its cyclin and CDKs. The LC n-3 PUFA suppress the cell cycle by alteration of cyclin and CDKs. Abbreviations: EGFR, epidermal growth factor receptor; EGF, epidermal growth factor; P, phosphorylation; RAS, retrovirus-associated DNA sequences; RAF, rapidly accelerated fibrosarcoma; MEK, mitogen-activated protein kinase; ERK, extracellular signal-regulated kinase; PI3K, Phosphatidylinositol-3-kinase; AKT, threonine-protein kinase; mTOR, Mammalian target of rapamycin; Bcl-2, B-cell lymphoma-2; BAX, Bcl-2 Associated X-protein; BAK, Bcl-2 Antagonist/Killer; tBid, truncated Bid; Puma, p53 upregulated modulator of apoptosis; MCL-1, myeloid cell leukemia-1; Bcl-xL, B-cell lymphoma-extra-large; BIM, Proapoptotic Bcl-2 homology 3-only protein; FADD, Fas-associated death domain protein; TRAIL-1 /2, Tumor necrosis factor-related apoptosis-inducing ligand receptor 1 /2; FAS, Fas Cell Surface Death Receptor; DISC, death-inducting signaling complex; CDK, cyclin-dependent kinase; G1, gap 1/growth 1; G2, gap-2/growth 2; M, mitosis; S, Synthesis Phase; DNA, deoxyribonucleic acid; JNK, Jun N-terminal kinase; NF-ĸB, nuclear factor kappa-light-chain-enhancer of activated B cells; COX-2, cyclooxygenase-2; PGE-2, prostaglandin E2; iNOS, inducible nitric oxide synthase; NO, nitric oxide; BH3, canonical mitochondrial apoptosis.
LC n-3 PUFA Alter CRC Growth Signaling Pathways
The cellular behavior in healthy individuals is tightly controlled by a complex network of signaling pathways involving growth factors, which ensure the proliferation of cells only when required. Dysregulation of these growth factor-dependent cell proliferation signaling pathways is one of the hallmarks of cancer development and progression. One of the identified dysregulations of the cell signaling pathway in CRC is the overexpression and activation of the epidermal growth factor receptor (EGFR). The EGFR is a multifunctional member of the ErbB family of tyrosine kinase receptors that transmits a growth-inducing signal to the cell.71,72 The higher expression of EGFR has been recognized as an important player in CRC initiation and progression. The EGFR is activated through interaction with its ligands, epidermal growth factor (EGF), and transforming growth factor-alpha (TGF-α). Overexpression of the EGFR-ligand complex plays a crucial role in cell proliferation, differentiation, survival, adhesion, migration, tumorigenesis, as well as resistance to cancer therapy.72,73 The binding of EGFR with its corresponding ligand triggers the catalytic activity of its intrinsic kinase that leads to the activation of several downstream intracellular signaling pathways, including Ras/Raf/Mitogen Erk1/2, the Phosphatidylinositol 3-Kinase (PI3K)/A Serine/Threonine-Protein Kinase (Akt)/mammalian target of rapamycin (mTOR) and signal transducer and activator of transcription (STAT-3), PLC-gamma-1, and c-Jun N-terminal kinase (JNK). 74 The overexpression of these signaling pathways causes the up-regulation of cell growth and carcinogenesis. 75 The inhibition or inactivation of the EGFR complex involves signaling pathways that are associated with an anti-cancer mechanism.65,66,70,76 Studies have shown that LC n-3 PUFA treatment can inhibit the activation of EGFR and its downstream intracellular signaling pathways Ras/Erk and AKT.66,72 Free fatty acid extract of krill oil treatment was also found to reduce the expression of EGFR/pEGFR and their downstream signaling, pERK1/2 and pAKT along with the downregulation of programed death-ligand 1 (PD-L1). 77
It has also been found that increased expression of vascular endothelial growth factor (VEGF) is associated with the progression of CRC; and the enhancement of VEGF signaling pathways could increase cancer cell proliferation, metastasis, and influences survival rates of CRC patients. 78 Activation of the VEGF receptor could also lead to transphosphorylation, an increase in intrinsic catalytic activity, and the creation of receptor binding sites on tyrosine kinases (RTK) to recruit cytoplasmic signaling proteins that activate mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase 1/2 (ERK 1/2) and mTOR/AKT pathways. 79 Continuous activation of MAPK and mTOR due to VEGF signaling stimulates the inhibition of cellular apoptosis and increases cell survival through the up-regulation of anti-apoptotic B cell lymphoma (Bcl) regulatory proteins, Bcl-2 and Bcl-xL.80,81 Furthermore, COX-2 is also involved in the expression of VEGF, because COX-2 and PGE-2 are well-established upstream regulators of VEGF during angiogenesis.82,83 Some studies have observed that COX-2-derived PGE-2 signaling through the prostaglandin (EP) 4 receptor stimulates cell proliferation, angiogenesis, and resistance to anti-tumor immune response and apoptosis.84,85 Furthermore, experimental results have shown that nitric oxide (NO) can promote cell survival, proliferation, inhibiting apoptosis,86,87 and regulate VEGF-mediated angiogenesis. 88 Studies found that LC n-3 PUFA treatment can reduce the expression of COX-2 and the synthesis of PGE-2.82,89 Furthermore, LC n-3 PUFA induce apoptosis through the modulation of Bcl-2 family proteins90,91 and decrease of VEGF signaling pathways65,92 involved in cell migration, blood vessel formation, and NO production.93,94
The peroxisome proliferator-activated-receptors (PPARs) are a group of the nuclear receptor proteins. 95 The LC n-3 PUFA are thought to play a crucial role in upregulating the transcription factor PPAR-alpha. This regulation is important to reduce the activity of the transcription factor NF-ĸB,96 -98 which is essential to mitigate inflammatory responses. 99 It has been reported that these beneficial effects are associated withthe mediators of LC n-3 PUFA including resolvins, protectins and maresins 99 In addition, the enhanced expression of PPARs was reported to be associated with the suppression of cellular proliferation64,100 and angiogenesis through the downregulation of VEGF. 65 It was found that EPA modulates PPARs to reduce cell viability by inducing cellular apoptosis. 64 Table 1 summarizes the in vitro studies that examined the effects of LC n-3 PUFA on the modulation of different survival signaling pathways involved in colorectal carcinogenesis.
Table 1.
Summary of Studies Investigating the Effects of LC n-3 PUFA on the Modulation of Survival Signaling Pathways in CRC Cells.
| Fatty Acids | Cell Lines | Effects | Molecular targets | References |
|---|---|---|---|---|
| DHA 2.5-10 μM |
SW-480, HCT-116 | Induces apoptosis | Reduces the β-catenin level by alternating the expression of T cell factor (TCF)-β catenin target genes, reduces survivin, and activates caspase-3 | Calviello et al 101 |
| DHA 5-50 μM |
SW-480, HCT-116, HT-29 and Caco-2 | Decreases cell viability and induces apoptosis |
Suppresses AKT phosphorylation, modulates p38 MARK pathway | Toit-Kohn et al 102 |
| DHA 30 µM | HT-29, HCT-116, SW-480 | Reduces cell proliferation and induces apoptosis | Inhibits the phosphorylation of pERK. Decreases the expression of GRP78. Activates caspase 4 |
Fasano et al 70 |
| DHA 100 µM | WiDr | Decreases cell proliferation | Decreases EGFR protein, and Ras and Erk activation | Rogers et al 66 |
| EPA/DHA 100 μM | HCT-116, HT-29/219, Caco-2, SW-742 and LS-180 |
Reduce cell proliferation | Down regulate VEGF protein, Modulate cellular miR-126 DNA methylation |
Moradi Sarabi et al 103 |
| EPA/DHA 10-30 µM | HT-29 | Reduce cell proliferation | Reduce VEGF and COX-2 expression, inhibit ERK 1/2 phosphorylation, and Hypoxia-inducible factor -1α protein overexpression | Calviello et al 65 |
| EPA 150 µM DHA 150 µM |
LoVo and RKO | Reduce cell proliferation and change cellular morphology | Decrease synthesis of PGE-2 and Leukotriene B4. Suppress expression of COX-2 |
Zhang et al 89 |
| EPA 40 µM | SW-480, HCT-116, HT-29 and Caco-2 | Reduces cell viability | Upregulates miR-378 | Weng et al 104 |
| EPA 1-150 µM |
HT-29 | Decreases proliferation | Directly involved with activation of PPARγ1 | Allred et al 64 |
| EPA/DHA 25-200 µM |
Human chemo-sensitive colon cancer HT-29, The chemo-resistant counterpart HT-29 dx cells | Reduce chemoresistance in HT-29 dx cells | Down regulation the endogenous cholesterol synthesis by promoting hydroxymethylglutaryl-coenzyme A reductase ubiquitination via Trc8 E3 ligase | Gelsomino et al 105 |
| EPA 12.5-200 μM |
Human HCA-7, HT-29 & Mouse MC-26 |
Induces apoptosis | Inhibits EP4 receptor signaling by PGE-3 derived from EPA. Reduces synthesis of PGE-2 | Hawcroft et al 106 |
| EPA 0-150 µM |
HCT-116 and SW-480 | Reduces cell proliferation and induce apoptosis | Inhibits mTOR signaling pathway. Induces cell cycle arrest in G0/G1 |
D’Angelo et al 107 |
| EPA 150 µM | SW-480 | Reduces cell viability | Up regulate miR-101 and downregulates Cox-2 | Cai et al 108 |
| Free fatty acid extract of krill oil | DLD-1 and HT-29 | Inhibits cell migration | Reduces expression of EGFR/pEGFR and their downstream signaling pathways pERK/ERK1/2 and pAKT/AKT; reduces expression of PD-L1 | Jayathilake et al 77 |
| Monoglyceride-EPA (MAG-EPA) 1-100 μM |
HCT-116 | Reduces cell proliferation and induces apoptosis | Decreases EGFR, VEGFR, and AKT activation pathway | Morin et al 109 |
| Ethyl esters of n-3 PUFA | SW 620 and HCT-116 | Reduces cell proliferation and invasion | Decrease Akt phosphorylation | Pfister et al 110 |
LC n-3 PUFA Induce CRC Death via Apoptosis
The onset of CRC associates with uncontrolled cell proliferation and a reduction in cell apoptosis. Apoptosis is a programed process of cell death and caspases are fundamental for this mechanism. There are three distinct pathways involved in the apoptotic mechanism, including the intrinsic (mitochondrial), the extrinsic (through the death receptors), and intrinsic endoplasmic reticulum (ER) pathways.111 -113
The intrinsic apoptosis pathway responds to diverse stress signals such as growth factor deprivation, DNA damage, and reactive oxygen species (ROS). Mitochondria are central to this pathway, which involves pro- and anti-apoptotic members of Bcl-2 family proteins.114,115 The pro-apoptotic Bcl-2 proteins include Bax, Bak, Diva, Bcl-Xs, Bik, Bim, Bad, and cBid; and the anti-apoptotic Bcl-2 proteins are Bcl-2, Bcl-XL, Mcl-1, CED-9, A1, Bfl-1. These proteins collectively regulate the mitochondrial membrane potential (MMP). An imbalance in pro- and anti-apoptotic proteins triggers -apoptotic events.116 -118 and results in the release of pro-apoptotic regulators, cytochrome c, Smac/Diablo, endonuclease G, and apoptosis-inducing factor (AIF) from mitochondria into the cytosol.112,119 Cytochrome c then triggers the formation of apoptosome complex via interaction with apoptotic protease activating factor 1 (Apaf1). This complex is involved in the activation of the initiator caspase, caspase-9, then caspases 3 and 7, leading to cell apoptosis.112,120 Furthermore, Smac/Diablo promotes apoptosis through the inhibition of anti-apoptotic protein, endonuclease G, and translocation of AIF in the cell nucleus. This causes a large amount of DNA damage via a caspase-independent apoptotic pathway.121,122
Cell apoptosis through the extrinsic pathway involves death domain receptors, such as tumor necrosis factor (TNF)-α, CD95 (Fas or apoptosis antigen 1), and TNF-related apoptosis-inducing ligand (TRAIL) receptors, after interaction with their corresponding ligands. The activation of these receptor-ligand complexes causes the formation of death-inducing signaling complexes (DISCs), including TNF receptor-associated death domain (TRADD), Fas-associated death domain (FADD), and procaspase-8/FLICE receptor-interacting protein kinase 1 (RIPK1). 112 These complexes activate caspases 8 and 10, and then caspase-7 leading to apoptotic cell death.112,123
Apart from those two caspase pathways, the intrinsic endoplasmic reticulum (ER) pathway is also related to apoptosis. The ER is a site for the synthesis, folding, modification of the cell surface proteins, and intracellular calcium storage compartment. The stress of ER could be induced by the accumulation of unfolded and/or misfolded proteins in the ER lumen. This could then activate the unfolded protein response (UPR) signaling pathway associated with a wide variety of human diseases. Furthermore, the unfolded protein response and continuous ER stress activate the apoptosis through caspases 3 and 7, or via a p53-dependent pathway.124,125
Numerous studies in the last two decades suggest that LC-n-3 PUFA treatment prompts colorectal cancer cell apoptosis through intrinsic or extrinsic pathways, as summarized in Table 2. LC n-3 PUFA elevate pro-apoptotic proteins, and suppress anti-apoptotic proteins, in the Bcl-2 family.126 -132 Furthermore, studies also showed that LC n-3 PUFA induce apoptosis via the activation of extrinsic pathways involving caspases 9 and 8.133,134 Giros et al found that LC n-3 PUFA induce apoptosis through both intrinsic and extrinsic pathways, via the release of Smac/Diablo and cytochrome c into the cytosol and the activation of caspase-8. Their results demonstrated that extrinsic apoptosis is independent of death receptor activation but influenced by LC n-3 PUFA on FLICE-like inhibitory proteins. 127
Table 2.
Summary of Studies Investigating the Effects of LC n-3 PUFA on Apoptosis of CRC Cells.
| Fatty acids/marine oil | Cell lines | Effects | Molecular targets | References |
|---|---|---|---|---|
| DHA 100 µM | HT-29, SW-480, SW-620, Caco-2, LoVo, HCT-116, HCT-8 | Reduces cell proliferation | Inhibits GrB expression, EMT, and cancer cell invasion | D’Eliseo et al 135 |
| DHA 70 µM | SW-620 | Reduces cell proliferation | Elevates phosphorylation of eIF2α, induces ER stress | Jakobsen et al 136 |
| DHA 70 µM | SW-620 | Reduces cell proliferation | Induces cell cycle arrest in both G1 and G2 phase, increases the C/EBP Homologous protein (CHOP) level | Slagsvold et al 131 |
| DHA 50-100 µM | HT-29 | Decreases cell proliferation and induces apoptosis | Decreases survivin expression, induces caspase-3 activation | Hosseini et al 137 |
| DHA 2.5-10 µM | SW-480 and HCT-116 | Induces apoptosis | Reduces β-catenin level, anti-apoptotic protein survivin, TCF-β-catenin target gene | Calviello et al 101 |
| DHA 20 µM | HT-29 | Reduces proliferation and induces apoptosis | Increases ROS production and lipid peroxidation, increases TNF-α mediated apoptosis |
Hofmanová et al 133 |
| DHA 50 µM | SW-480 | Induces apoptosis | Enhances TRAIL, activates Bax/Bak, reduces MMP, releases cytochrome c, upregulates CHOP, decreases X-linked inhibiter of apoptosis protein (XIAP) |
Skender et al 130 |
| DHA 100-200 µM | HCT-116 | Reduces cell proliferation and induces apoptosis | Reduces survivin, increases Wt-p53 level, increases caspase-3 activation | Sam et al 68 |
| DHA 50-150 µM | LS-174T | Reduces proliferation, induces apoptosis | Inhibits survivin expression, activates caspase-3 | Ahangar et al 138 |
| Conjugated docosahexaenoic acid (CDHA) | Colo- 201 | Inhibits cell proliferation and induced apoptosis. | Upregulation of the apoptosis enhancing proteins (Bak and Bcl-xS) and downregulation of the apoptosis inhibition proteins Bcl-xL and Bcl-2 | Danbara et al 126 |
| EPA 24 µM & DHA 20.5 µM |
HT-29, LS-174 |
Induce pro-apoptotic effects | Depolarize mitochondrial membrane | Granci et al 128 |
| EPA/DHA or combination 50, 100 & 150 µM |
LS-174T | Both fatty acid & combination reduce cell viability and induce apoptosis | Decrease survivin mRNA expression and activate caspase-3 | Sam et al 139 |
| EPA/DHA 60 µM | Caco-2, HT-29, HCT-116, LOVO and SW-480 | Both fatty acid activate intrinsic and extrinsic death pathway | Release cytochrome c, Smac/Diablo, and activate caspase-9. Inhibition of FLIP, XIAP |
Giros et al 127 |
| EPA/DHA more than 120 µM | LoVo and RKO | Induce apoptosis | Decrease MMP, the formation of ROS, activation of caspase-9 and 3, increase in Bax/Bcl2 expression | Zhang et al 132 |
| Fish oil | Caco-2 and HT-29 | Induces apoptosis | Downregulation of Cox-2 and decrease in Bcl-2 expression | Llor et al 129 |
| EPA/DHA 100 μM |
Caco-2 | Reduce CRC cell proliferation, induce apoptosis (opposite results observed at 10 μM) |
Induce cell death via mitochondrial apoptosis pathway. | Storniolo et al 57 |
| Free fatty acid extract of krill oil. EPA 100 & 200 μM |
HCT-15, SW-480 and Caco-2 | Inhibit cell proliferation and induce apoptosis | Induce cell death via potentially mitochondrial mediated pathway | Jayathilake et al 140 |
| Free fatty acid extract of krill oil. EPA and DHA | DLD-1, HT-29 and LIM-2405, and one mouse CRC cell line, CT-26 | Supress cell proliferation (comparable with chemotherapeutic agent Oxaliplatin). and increase ROS formation | Induce mitochondrial apoptosis pathway. Increases expression of caspase-3 and caspase-9 leading to DNA damage | Jayathilake et al 141 |
LC n-3 PUFA Alter CRC Cell Cycle
The cell cycle orchestrates precise molecular events, ensuring the generation of identical cell copies. Healthy cells regulate growth signals activated by factors such as growth factors, cell-to-cell interaction molecules, and extracellular matrix components that affect cell growth and maintain the cell cycle. This helps to control the total number of cells, and the structure and function of normal tissues. 116 However, some cells are progressively transferred into a neoplastic state (cancer) when they escape from the normal cell cycle and produce their own growth factors independently to proliferate infinitely. 116
The cell cycle consists of four sequential phases: G1 (gap 1), S (DNA synthesis), G2 (gap 2), and M (mitosis). Cyclin-dependent kinase (CDK) plays a pivotal role in regulating the progression of cell cycle and preventing transitions between phases.142,143 CDK activation occurs through the interaction with cyclins, forming a cyclin-CDK complex143,144 In response to mitogen signals, CDK4 and CDK6 activate the D-type cyclins, facilitating G1 progression and activation of the transcription factors.144,145 CDK2 is subsequently activated by cyclin E and cyclin A, initiating DNA replication. The activation of cyclin B-CDK1 complex leads to mitosis, while cyclin-dependent kinase inhibitors (CKI) including INK4 and Cip/Kip families monitor and deactivate CDK-cyclin complexes that arrest the cell cycle. The INK family has four members, p15, p16, p18, and p19; and the Cip/Kip family comprises p21, p27, p57, p107, and p130.144,146,147
Several studies reported that LC n-3 PUFA inhibit cancer cell proliferation through the alteration of cell cycle progression. Table 3 summarizes the results of in vitro studies elucidating the impacts of LC n-3 PUFA on the CRC cell cycle. Treatment with DHA and/or EPA arrests the cell cycle progression in the G1 phase in CRC cells.69,126,131 Molecular components crucial for CRC cell cycle progression, such as Cdc25c, Cdc25b, Cdc20, CDK1, CDK2, and cyclin D, A, and B are downregulated by LC n-3 PUFA.69,126,131,136,148 Studies found that DHA treatment increases the level of p21 and reduces the level of cyclin D1, inducing cell cycle arrest.126,131 LC n-3 PUFA also activate the p53 pathway, causing DNA damage and modulating the p21 signaling pathway. 149
Table 3.
Summary of Studies Investigating the Effects of LC n-3 PUFA on the Cell Cycle of CRC.
| LC n-3 PUFA | Cell lines | Effects | Molecular targets | References |
|---|---|---|---|---|
| Conjugated docosahexaenoic acid (CDHA) 1% | Colo-201 Colo-201 in Nude mice |
Inhibits cell cycle progression. Significantly decreases transplanted Colo-201 cells in mice |
Accumulates cells in the G1 phase, increases p21 Cip1/Waf1, decreases cyclin D1, cyclin E, and nuclear cell proliferating antigen | Danbara et al 126 |
| DHA 20 µM | HT-29 | Reduces cell viability | Arrests cell cycle in G1/G2 phases | Hofmanová et al 133 |
| DHA 70 µM | SW-620 | Arrests cell cycle | Increases P21 and stratifin, decreases Cdc25, CDK1(Cdc2), arrests cell cycle in both G1 and G2 phases | Slagsvold et al 131 |
| DHA 50 µM | Caco-2 and HT-29 | Reduces proliferation | Arrests cell cycle at G0/G1 phase, reduces cyclin D1 expression and phosphorylation of GSK3β | Murad et al 69 |
| DHA 70 µM | SW-620 | Reduces cell proliferation, induces apoptosis | Reduces cyclin D1 and arrests cell cycle at G1phase | Jakobsen et al 136 |
| DHA 150 µM | HT-29 | Reduces cell proliferation | Arrests cell cycle at G1 phase, reduces cyclin D1 and E | Chen and Istfan 148 |
| DHA/EPA 125 µM |
COLO-205, WiDr | Reduces cell proliferation | Arrests cell cycle at G0/G1 phase | Kato et al 150 |
| DHA/EPA 35-70 µM |
SW-480, SW-620 |
Reduce cell proliferation | Arrest cell cycle at G2/M phase, downregulate nSREBP1 | Schønberg et al 151 |
| EPA 10-100 µM DHA 7.5-75 µM |
Caco-2 | Reduce cell proliferation and induce apoptosis | Arrest cell cycle at G0/G1 and S phases | Jordan and Stein 152 |
Overall, a large number of in vitro studies have confirmed the positive impacts of LC n-3 PUFA on CRC cells, and several molecular signaling pathways have been suggested. However, the comparative roles of EPA versus DHA versus total LC n-3 PUFA in the modulation of these molecular pathways are not clear, given that different doses, treatment durations as well as cell lines were used in different studies.
The Anti-inflammatory Effects of LC n-3 PUFA on CRC Cells
The relationship between chronic inflammation and cancer initiation and progression has been well recognized and documented for several types of cancer, including CRC.153 -155 The risk of CRC could be increased by 10 times in patients with a history of inflammatory bowel disease. 156 The cyclic auto-activation process of the inflammatory signaling pathways increases the production of immune cells, and release of several pro-inflammatory cytokines (IL-1, IL-2, IL-4, IL-6, IL-12, IFNɤ, and TNFα), chemokines (IL-8, monocyte chemo-attraction protein-1 (MCP-1/CCL2)), growth factors, reactive oxygen and nitrogen species, and lipid molecules (saturated fatty acids)157,158 These mediators stimulate signaling processes within the cells that play an important role in the growth and development of cancer. 157
Furthermore, the soluble mediators generated by cancer cells also contribute to the recruitment and activation of immune cells and the excessive production of pro-inflammatory mediators. The inflammatory reaction generated by immune cells activates and maintains several signaling processes. These processes continue to stimulate proliferative signaling, the survival of cancer cells, and extracellular matrix (ECM), which facilitate tumor growth by modifying the enzymes that enable angiogenesis and metastasis.116,159 -163 In addition, some tumor and myeloid cells, including macrophages, polymorphonuclear neutrophils (PMN), and myeloid-derived suppressor cells (MDSCs) express mediators of the immunosuppressive checkpoint, such as programed death-ligand 1 (PD-L1) and ligand 2 (PD-L2) resulting in cellular interactions that suppress T-cell proliferation and function.164,165 Studies have found that LC n-3 PUFA have an ability to down-regulate inflammation and inflammatory cell infiltration of tumors. 166 Furthermore, LC n-3 PUFA have pro-resolving effects on both innate and adaptive immunity through multiple mechanisms, including influence on various cellular phenotypes that coordinate the host response against tumors. Resolvins that are the metabolites from LC n-3 PUFA have an endogenic pro-resolution activity that protects against abnormal/uncontrolled innate inflammatory responses. 165 Altogether the inflammatory responses play a crucial role in tumor development at different stages including initiation, promotion, malignant transformation, invasion, and metastasis. 161 Recently, the proliferative effects of pro-resolvin mediators on Caco-2 cell line have been reported. 167 The proliferation of intestinal epithelial cells stimulated by pro-resolvin mediators was related to wound closure. More studies using other cell lines are warranted to elucidate the effect of pro-resolvin mediators on CRC growth. LC n-3 PUFA have shown useful anti-inflammatory properties, due to the immunomodulatory effects of substances derived from these fatty acids through eicosanoid metabolism. 168 The main anti-tumor effect of LC n-3 PUFA can be mediated through the downregulation of the synthesis of pro-inflammatory eicosanoids from n-6 PUFA of AA. 169
The phospholipid membrane normally contains more AA than other 20-carbon PUFA. 169 AA is the molecular substrate causing the over-activation of some enzymatic pathways in CRC, such as COX and LOX.170,171 The PGE-2 produced by COX from the metabolism of AA is typically pro-inflammatory,27,172 and excess PGE-2 has been found to be linked with the onset and progression of colorectal carcinogenesis through increased cell proliferation, angiogenesis, cell migration and invasion, as well as the inhibition of apoptosis.173,174
Dietary supplementation with LC n-3 PUFA significantly decrease the concentration of AA in the cell membrane, as well as its ability to displace AA as a molecular substrate in the COX and LOX pathways.27,175 The actions of metabolized derivatives of LC n-3 PUFA, such as prostaglandin E3 (PGE-3), on the substrate of COX and LOX are typically anti-inflammatory. 176 Therefore, it has been found to reduce angiogenesis,177,178 inflammation,27,179 and exert anti-cancer properties.27,180 The studies on the anti-CRC effects of LC n-3 PUFA via modulation of the inflammatory response are summarized in Table 4.
Table 4.
Summary of Studies on Anti-Inflammatory Effects of LC n-3 PUFA in CRC Cells.
| LC n-3 PUFA | Cell lines | Effects | Molecular targets | References |
|---|---|---|---|---|
| DHA 5 µg/mL | Caco-2 | Reduces cell proliferation, induces apoptosis | Up-regulates p21 and p27 Downregulates iNOS, IFN, cyclic GMP and NFĸB |
Narayanan et al 94 |
| DHA 0-100 µM | LS-174T | Reduces cell viability, suppresses AA-induced cell viability | Significantly lowers PGE-2 formation, downregulates COX-2 expression | Habbel et al 172 |
| DHA 30-100 μM | HCT-116 | Induces apoptosis | Decreases Bcl-2, CDK-2 and CDK-4, decreases β- catenin expression. Inhibits COX-2 and activation of NF-kB |
Han et al 181 |
| DHA 50-100 μM | HCT-116 and HCT-8 CRC cells | Induces apoptosis | Reduces IL-6 and IL-8 expression. Reduces TNF-α synthesis through decreasing mir-21. |
Fluckiger et al 182 |
| EPA 10-30 µM DHA 10-30 µM |
HT-29 | Inhibit cell growth (both EPA and DHA) | Inhibit VEGF expression and reduce COX-2/PGE-2 level | Calviello et al 65 |
| EPA-FFA 12.5-200 μM |
Human HCA-7 CRC, and Mouse MC-26 CRC cells | Induces apoptosis | Reduces COX-2 dependent PGE-2 synthesis and increases PGE-3. Inactivates PGE-2-EP4 receptor | Hawcroft et al 82 |
LC n-3 PUFA can downregulate the Bcl-2 and inhibit the Nuclear factor-kB (NF-κB) pathway. 181 A similar effect has been observed in other studies, showing that LC n-3 PUFA can cause the suppression of genes involved in the NF-ĸB pathway.94,174,180 NF-ĸB is a transcription factor that regulates the expression of many genes involved with the upregulation of COX, production of inflammatory cytokines, the progression of the tumor cell cycle, and adhesion molecules. These activities play an important role in tumor invasiveness and provide tumors with the inflammatory microenvironment that supports tumor progression, invasion of surrounding tissue, angiogenesis, and metastasis.183 -186 In addition, the experimental evidence shows that LC n-3 PUFA can decrease the gene expression of inflammatory molecules such as the inflammatory cytokines interleukin IL-1β, IL-6 and TNF-α.187,188 The anti-inflammatory properties of LC n-3 PUFA include the reduction of anti-apoptotic genes, Bcl-2 and survivin and the increase of pro-apoptotic factors.172,181 More studies are warranted to further identify the specific interactions of LC n-3 PUFA and gut microbiota, and their impacts on the immune response in CRC.
The Anti-Cancer Effects of LC n-3 PUFA in Vivo
Animal studies have demonstrated the beneficial effects of LC n-3 PUFA on CRC at the early stages of carcinogenesis. EPA and DHA can reduce tumor growth, progression, and invasion by inhibiting cell proliferation and inducing cell apoptosis.109,189-193 The LC n-3 PUFA supplementation in immunosuppressed mice with human CRC cell xenografts significantly reduced tumor size compared to the control group.150,189,190,194,195 Several studies have also reported positive outcomes following supplementation with LC n-3 PUFA-rich marine oils including fish oil and krill oil.178,193,195,196 In addition, these studies have reported several molecular actions of LC n-3 PUFA in CRC tumor inhibition (Table 5). For example, modulation of COX metabolism and reduction of PGE-2 production in tumors, 180 inhibition of EGFR and VEGF signal transduction through the alteration of lipid raft composition and fluidity, 197 and downregulation of pathways associated with CRC promoting signals such as AKT90,195,198 and Wnt/β-catenin. 180
Table 5.
Summary of In Vivo Studies Investigating the Effects of LC n-3 PUFA and Their Marine Oil Sources on CRC and Associated Signaling Pathways.
| Treatments | Animal models | Effects | Molecular targets | References |
|---|---|---|---|---|
| 1) Control, 2) DHA diet, 3) High DHA diet 5 wk |
C57BL/6 mice. MC-38 cells n = 24 |
Both diets reduce tumor weight by 50% | Reduce the presence of endothelial cells (CD31+ and CD-45), reduce the expression of pro-oncogenic genes such as cmyc, Axin2, and C-jun in tumor tissues | Wang et al 192 |
| 1) Fish oil and pectin 2) Control - Corn oil + cellulose 35 wk |
Male Sprague Dawley rats (n = 40) AOM-injected |
Fish oil reduces tumor growth and induce apoptosis | Suppress COX (mPGE-2 and PGE-2) and Wnt/ β-catenin pathway. Elevate pro-apoptotic, eicosapentaenoic acid-derived COX metabolite, PGE-3 | Vanamala et al 180 |
| 1) Control 2) EPA ethyl ester 3) DHA ethyl ester 1 g/kg bodyweight for 5 wk |
Athymic Balb/c nude mice HT-29 cells (n = 45) |
Both EPA and DHA treatments reduce tumor growth | Reduce COX-2 and PGE-2 expression Reduce VEGF expression |
Calviello et al 65 |
| 1) Untreated control 2) MAG-EPA treated 3) Krill oil treated 4) EPA- ethyl ester treated 618 mg/kg/d 30 days |
Nude mice (n = 32) HCT-116 |
MAG-EPA reduces tumor growth more efficiently than krill oil and EPA-EE | Decreases activation of EGFR, VEGFR, and AKT pathways reduce VEGF and HIF1α expression | Morin et al 198 |
| 1) Corn oil 2) No EPA-FFA 3) 2.5% (w/w-1) EPA-FFA 4) 5% (w/w-1) EPA-FFA 28 days |
Balb/c MC-26 (n = 64) |
5% (w/w-1) EPA-FFA reduces tumor size | Reduces PGE-2 and ERK 1/2 levels, increase PGE-3 level | Hawcroft et al 106 |
| 1) 8% Mazola corn oil 2) 24% Mazola corn oil 3) 38% Mazola corn oil + 16% menhaden oil 4) 8% Mazola corn + 16% DHA rich single cell oil (DHASCO) 55 days |
Athymic mice COLO-205 (n = 24) | DHASCO reduces tumor growth, induce apoptosis | Reduces tumor growth via p53 dependent and independent pathway | Kato et al 150 |
| 1) 7.5% fish oil 2) 7.5% corn oil 8 wk |
Balb/c nude mice HCT-15 |
Fish oil reduces tumor growth | Downregulates COX-2, HIF-1α, VEGF-A, MMP-1, MMP-9, SCP2 and SDC3 | Zou et al 194 |
| 1) Control diet 2) Modified diet FO:CO (1:1) 3) Modified diet FO:CO (2.5:1) 16 wk |
Male Wistar rat | Both FO diets reduce tumor size, inhibit inflammation signals | Reducers, Raf, MEK1/2, Erk 1/2, Akt and c-fos | Kansal et al 199 |
| 1) 4% Olive oil 2) 4% Fish oil 3) 4% Flaxseed oil 4) Soybean oil (control) 9 wk |
Wistar rats (n = 40) 1,2-dimetilhidrazine (DMH)injected |
Fish oil reduces tumor growth | Reduces IL-8 | Rosa et al 200 |
| 1) Control 1 - control diet for 5 wk 2) Control 2 - control diet for 10 wk 3) Treatment group - 5 wk on standard diet + 5 wk on salmon oil diet. |
C57BL/6 J male mice (heterozygote mutation for the Apc gene) (n = 15) |
Salmon oil treatment supresses intestinal polyps and reverses polyp development | Down-regulates cell proliferation markers, induces estrogen receptor β and LDL receptor, and decreases p-STAT3 Ser protein expression |
Notarnicola et al 201 |
| 1) Control -(combination of oil 5% in normal diets) 2) DHA-enriched diet (3% of Omegavie DHA 90TG oil and 2% of sunflower oil) 3) DHA-enriched diet + 100µg CTRL IgG 4) DHA-enriched diet + 100µg anti-TNFα 16 days |
Male athymic nude mice (n = 32) HCT-116 cells |
DHA diet reduces tumor volume | Reduce human miR-21, increase human TNFα mRNA | Fluckiger et al 182 |
| 1) Control diet 2) 2% EPA diet 16 wk |
C57BL/6 J-Apc mutant mice (n = 10) | EPA reduces tumor size and number | Resolvin E3 inhibits Cox-2 expression via the miR-101- mediated pathway | Cai et al 108 |
| 1) Control (soy bean) 2) Fish oil 36 wk |
Wistar rats (male), DMH injection n = 20 |
Fish oil protects against the DMH induced preneoplastic colon lesions and adenoma development | Moreira et al 202 | |
| 1) Control 2) EPA-FFA 2.5% 3) EPA-FFA 5% 12 wk |
Male ApcMin/+ mice on a C57BL/6 J background and wild-type (wt) mice n = 48 |
Both EPA groups show reduction of polyp number, decrease cellular proliferation and increase in apoptosis | Reduce COX-2 expression, β-catenin nuclear translocation |
Fini et al,2010 203 |
| 1) Control diet 2) Fish oil diet 2 wk pre-treatment and 3 wk after treatment |
Athymic nude mice (female) n = 51 HCT-116 cells |
Fish oil reduces tumor growth | Inhibits prostanoid synthesis from AA | Boudreau et al, 2001 193 |
| 1) Control 2) Fish oil: Corn oil (1:1) 3) Fish oil: Corn oil (2.5:1) 4 wk |
Wistar rats | Fish oil and corn oil combination reduce the cell proliferation and induce apoptosis | Decrease Ras, Raf, MEK1/2, Erk1/2 and c-fos level | Kansal et al, 2012 191 |
| Diet modified with fish oil (12% calorie) 6 wk |
Balb/c nu/nu athymic mice SW-620 |
Fish oil reduces tumor growth | Reduces phosphocholine | Bathen et al, 2008 189 |
| 1) Control (corn oil + butyrate) 2) Fish oil) fish oil + butyrate) 7 wk |
Sprague-Dawley rats n = 80 AOM |
Fish oil presents chemoprevention properties and increases apoptosis. | Increases the expression of p21 | Crim et al,2008 204 |
| 1) Control (standard chow diet) 2) 5% krill oil supplementation 3) 10% krill oil supplementation 4) 15% krill oil supplementation. All krill oil 7 wk |
Male Balb/c mice CT-26 CRC cells orthotopic implantation | Krill oil reduces tumor growth in a dose dependent manner; inhibits tumor cell proliferation and micro-vessel density | Down-regulates EGFR signaling pathway; Activates caspase 7, PARP cleavage and induce DNA/RNA damage |
Jayathilake et al, 2022 195 |
| 1) Control (standard chow diet) 2) Krill oil group (150g/kg/d) 3) Corn group (150g/kg/d) 4) Krill oil with ½ dose of oxaliplatin group (150g/kg/dkrill oil + 1.5 mg/kg/d oxaliplatin) 5) Corn oil with ½ dose of oxaliplatin group (150g/kg/d + 1.5 mg/kg/d oxaliplatin) 6) Oxaliplatin group (oxaliplatin 3 mg/kg/d) 7 wk |
Male Balb/c mice CT-26 CRC cells orthotopic implantation (n = 36) | Krill oil reduces the tumor growth comparable with oxaliplatin treatment | Increases the expression of cytochrome c, cleave caspase-9, caspase-3 and DNA damage. Decreases the expression of PD-L1, PD-L2 and HSP-70 | Jayathilake et al, 2022 196 |
| 1) Control- canola oil 1 ml 2) Control + n-3 PUFA [300µg/kg/day (EPA 180 µg +DHA 120 µg)] 3) (methotrexate +canola oil (similar to group 1) 4) methotrexate +n-3 PUFA (similar to group 2) 6 days |
Sprague -Dawley rats (male) (n = 32) | n-3 PUFA Increase body weight, reduce MTX-induced intestinal mucositis and inflammatory cytokines in intestinal mucosa and serum, decrease intestinal epithelial programed cell death |
Reduce TNF-α, NF-қB and COX-2. Inhibit intestinal epithelial cell apoptosis | Koppelmann et al,2021 205 |
| 1) Control 2) n-3 PUFA, 2 g/ kg-1/d 4 wk |
Female Sprague Dawley rats (n = 60) CRC induced by N-methyl-N-nitrosourea |
n- 3 PUFA reduce tumor growth through inhibiting CRC cell proliferation and inducing apoptosis | Inhibits PI3K/AKT/Bcl-2 signaling | Huang et al, 2020 90 |
Kato et al reported that DHA inhibits tumor growth more efficiently than EPA in athymic mice with COLO 205 subcutaneous xenografts. The DHA treatment reduced tumorigenesis by inhibiting genes responsible for tumor angiogenesis, as well as inducing cellular apoptosis via p53 dependent and independent pathways. These positive effects were found to be correlated with the increased level of LC n-3 PUFA in tumor tissues and the reduction of phosphocholine (PC) in xenografts. 150 Zou et al reported that the molecular mechanisms associated with the anti-cancer properties of LC n-3 PUFA involve the inhibition of several genes, such as COX-2, HIF-1α, VEGF-A, COMP, MMP-1, MMP-9, SCP2, SDC3 in nude mice with HCT-15 subcutaneous xenografts. 194 Kansal et al showed that dietary supplementation with LC n-3 PUFA-rich fish oil significantly suppress CRC development and increased apoptosis by reducing the expression of the Ras-induced Raf/MEK/ Erk 1/2 and Akt signaling pathways in rats treated with a carcinogen 1,2-dimethylhydrazine(DMH). 191 Furthermore, Huang et al found that n-3 PUFA attenuated MNU-induced CRC in rats by inhibiting CRC cell proliferation and inducing CRC cell apoptosis via blocking PI3K/AKT and Bcl-2 signaling pathways. 90 The role of LC n-3 PUFA in decreasing cell proliferation in tumor tissues was also found to be associated with the modulation of inflammatory pathways. Rosa et al discovered that a fish oil-enriched diet reduced interleukin-8 (IL-8) expression and enhanced TGF-β expression in Wister rats treated with DMH. 200 In addition, fish oil was found to suppress the initiation of aberrant crypt foci (ACF) development in male Wister rats, 202 reduce TNF-α production in HCT-116 xenograft tumor-bearing nude mice, 182 reduce the expression of phosphorylated ERK 1/2, lower the PGE-2 and increase the PGE-3 levels in Balb/c mice. 106 Koppelmann et al found that n-3 PUFA inhibit NF-қB/COX-2 induced production of pro-inflammatory cytokines and inhibited cell apoptosis to prevent MTX-induced intestinal damage in male Sprague-Dawley rats. 205 Fish oil diet has also been shown to upregulate p21, induce cell cycle arrest, and promote apoptosis of cancer cells in rats with azoxymethane (AOM)-induced colon cancer. 204 LC n-3 PUFA rich diets were found to dramatically decrease cell proliferation and polyp formation and increase apoptosis of cancer cells through the downregulation of COX-2 and β catenin nuclear translocation in ApcMin/+ mice. 203 Recently, we reported that dietary supplementation with krill oil reduces CRC tumor growth and induces cancer cell death. 195 We also observed that the anti-cancer effects of krill oil are comparable with that of the clinical therapeutic agent, Oxaliplatin. These positive effects are associated with the downregulation of EGFR signaling pathways.195,196 and the activation of the intrinsic mitochondrial death pathway. 196 Available animal studies have confirmed some of reported molecular actions of LC n-3 PUFA in vitro. However, there are still gaps between in vitro and in vivo studies. Future in vivo studies focusing on those yet to be proven molecular signaling pathways are highly recommended.
LC n-3 PUFA as an Adjuvant Therapy for CRC
Several studies have investigated the action of LC n-3 PUFA as a single agent or in combination with chemotherapeutic agents in the treatment of cancer. 206 A recent review of current clinical evidence showed that LC n-3 PUFA supplementation delays cancer progression, maintains body weight, and improves appetite and overall quality of life in CRC patients. In the advanced cancer patients, supplementation also decreases pro-inflammatory cytokines and serious adverse events of chemotherapy. 207 The increased level of LC n-3 PUFA in tumor cell membrane and production of lipid mediators were found to be correlated with their anti-cancer properties. High levels of LC n-3 PUFA resulted in an alteration of lipid rafts leading to a range of beneficial events in tumor cells.105,208 The combined treatments of LC n-3 PUFA with chemotherapy have resulted in reducing the dose of chemotherapeutic agents and decreasing the side effects associated with higher doses of chemotherapy. The lower toxicity profile and nutritional benefits of LC n-3 PUFA provide a useful adjuvant therapy for CRC. 209 Fish oil emulsion rich in LC n-3 PUFA has shown anti-cancer effects in combined treatments with standard chemotherapeutic agents, 5-fluorouracil (5-FU), oxaliplatin (OX), or irinotecan (IRI) in CRC cell lines. 128 Fish oil emulsion improves the efficacy of chemotherapeutic agents through a Bax-dependent mitochondrial pathway. 128 Vasudevan et al have demonstrated that treatment with EPA in combination with 5-FU plus oxaliplatin (FuOx) reduces HT-29 and HCT-116 cancer cell growth. It also induces apoptosis through PARP cleavage, and downregulation of pAKT, β-catenin, and Wnt signaling pathways. 210 5-FU combined with fish oil emulsion inhibits the proliferation of Caco-2 CRC cells lines more significantly compared to cells treated by either agent alone. 152 Similar findings by Calviello et al showed that DHA combined with 5-FU resulted in a decrease of the expression of anti-apoptotic proteins Bcl-2 and Bcl-Xl and excessive expression of pro-apoptotic c-MYC in human CRC cells, with low toxicity. 173 De Carlo et al also observed that EPA combined with standard chemotherapies, 5-fluorouracil and oxaliplatin, synergistically reduces COLO 320 CRC cell proliferation and increases their sensitivity to chemotherapeutic drugs. 211
Similar results have also been observed in several animal models of CRC. Rani et al demonstrated that the administration of 5-FU combined with fish oil to mice with colon cancer induced by DMH and dextran sodium sulfate (DSS) enhances both DNA damage and an apoptotic index through the activation of cellular extrinsic and intrinsic apoptotic pathways while reducing the side effects associated with 5-FU treatment. 212 It was also reported that 5-FU combined with fish oil treatment inhibits tumor growth and arrests the cell cycle in mice with DMH/DSS-induced colon cancer. 213 Other studies have also highlighted the beneficial effect of LC n-3 PUFA as adjuvant therapy. Xue et al have found that LC n-3 PUFA supplementation combined with irinotecan (CPT-11) and a 5-FU cyclical regimen could synergistically increase chemo-sensitivity and reduce body weight loss, anorexia, and muscle wasting in rats bearing Ward colon tumor compared to the control animals. 214 Vasudevan et al have also observed that treatment with EPA combined with FuOx (5-FU + oxaliplatin) reduces the tumor size and pro-inflammatory mediators in SCID mice with HT-29 or HCT-116 cell-induced subcutaneous xenografts. 210 Jeong et al found that the oxaliplatin and DHA combination can further reduce oxaliplatin-induced cell viability and autophagy cell death both in vitro and in vivo. Moreover, the combination of oxaliplatin and DHA increased the expression of the stress-sensitive gene of SESN2 and increased ER stress. 215 Our recent study has demonstrated that krill oil combined with ½ dose of oxaliplatin can reduce tumor growth to a similar extent as oxaliplatin, without side effects. 196
Some human studies have also demonstrated the anti-cancer effects of LC n-3 PUFA combined with chemotherapy on CRC. It is found that fish oil supplementation positively modulates the nutritional status and reduces pro-inflammatory mediates in CRC patients undertaking chemotherapy. 207 Similarly, Mocellin et al observed that fish oil supplementation during chemotherapy improves C-reactive protein (CRP) values, CRP/albumin status, and prevents weight loss. 216 A study of patients receiving a combination of fish oil and chemotherapy (5-FU and leucovorin) showed that fish oil can prevent the loss of blood polymorphonuclear cells (PMNC), mainly neutrophils, and increase their phagocytosis and production of hydrogen peroxide as well as prevent body weight loss related to chemotherapy. 217 Read et al observed that EPA supplementation maintains the nutritional and inflammatory status in patients having chemotherapy at an advanced stage of CRC. 218
A more recent study by Koppelmann et al has also demonstrated the beneficial role of fish oil as an effective adjuvant therapy for colon cancer. 205 Their data showed that LC n-3 PUFA can prevent intestinal damage and stimulate intestinal recovery. The prospective study by Song et al has found that a higher intake of LC n 3-PUFA may be associated with better survival of patients with stage 3 colon cancer. 219
Taken together, these studies demonstrate that LC n-3 PUFA, either alone or in combination with currently used clinical chemotherapy, may be a useful therapy for CRC. This is attributed to the specific roles of these fatty acids in suppressing tumor growth and development via various molecular signaling pathways discussed in this review. The synergistic impact of LC n-3 PUFA and chemotherapies on CRC are beneficial to CRC patients. Two major sources of LC n-3 PUFA are fish oil and krill oil. The effects of fish oil on CRC have been reported. Preliminary studies have also shown the potential role of krill oil in CRC treatment. More in vivo studies and clinical trials are required to validate the therapeutic efficacy of krill oil.
Conclusion
The data presented in this review have shown the beneficial effects of LC n-3 PUFA on colorectal cancer. The results from several experimental studies using CRC cell lines and animal models provide strong evidence that LC n-3 PUFA supress CRC by modulating different molecular pathways associated with cancer development and progression. These include the action of LC n-3 PUFA on intracellular and extracellular receptors in various signaling pathways involved in cell proliferation, metastasis and apoptosis, as well as angiogenesis and inflammation. Clinical studies have also demonstrated that LC n-3 PUFA can enhance the efficacy and tolerability of chemotherapy by reducing the side effects and toxicity associated with conventional anti-cancer therapies.
Footnotes
Abbreviations: AKT, threonine-protein kinase; BAK, Bcl-2 antagonist killer; BAX, Bcl-2-associated X protein; Bax/Bak, nuclear encoded proteins; Bcl-2, B cell lymphoma protein-2; Bcl-xL, B cell lymphoma protein-xL; CDC25, cell division cycle 25 homolog; CDHA, conjugated docosahexaenoic acid; CDK-1, cyclin-dependent kinase 1; CDK-2, cyclin dependent kinases 2/4; c-fos, proto-oncogene; C/EBP, enhancer-binding proteins; CHOP, C/EBP homologous protein; CiPi/waf1, cell cycle inhibitor proteins; CO, corn oil; COMP, cartilage oligomeric matrix protein; COX-2, cyclooxygenase-2; cyclic GMP, cyclic guanosine monophosphate; DHA, docosahexaenoic acid; DMH, 1,2-dimetilhidrazine; DNA, deoxyribonucleic acid; EGFR, epidermal growth factor receptor; eIF2α, eukaryotic translation initiation factor 2-α; EMT, epithelial-to-mesenchymal transition; EP4, G protein–coupled prostaglandin receptor; EPA, eicosapentaenoic acid; EPA-EE, ethyl ester eicosapentaenoic acid; EPA-FFA, eicosapentaenoic free fatty acids; ER, endoplasmic reticulum; ERK 1/2, extracellular signal-regulated kinase; FFA, free fatty acid; FLIP, FLICE-inhibitory protein; GrB, granzyme B; GRP78, glucose related protein of 78 kDa; GSK3β, glycogen synthase kinase 3β; HIF-1α, hypoxia-inducible factor 1-alpha; IFN, interferon; IL-1-β, interleukin 1 beta; IL-6, interleukin 6; IL-8, interleukin 8; iNOS, inducible nitric oxide synthase; FO, fish oil; LTB4, leukotrienes B4; MAG-EPA, monoglyceride eicosapentaenoic acid; MAPK, mitogen-activated protein kinase; MEK 1/2, mitogen-activated protein kinase 1/2; mir-21, microRNA-21; MMP, mitochondrial membrane potential; MMP-1/9, matrix metalloproteinase-1/9; mPGE-2, messenger function for prostaglandin E2; mRNA, messenger ribonucleic acid; mTOR, mammalian target of rapamycin; NF-ĸB, nuclear factor kappa-light-chian-enhancer of activated B cells; n-SREBP1 nuclear sterol regulatory element-binding protein 1; P21/27, cyclin-dependent kinase inhibitor 21/27; PERK, PER-like ER kinase; PG, prostaglandin; PGE-2, prostaglandin E2; PGE-2/E-3, prostaglandin E2/E3; PGE-3, prostaglandin E3; PPAR-γ, peroxisome proliferator-activated receptor-γ; PUFA, polyunsaturated fatty acids; Raf, rapidly accelerated fibrosarcoma; Ras, retrovirus-associated DNA sequences; ROS, reactive oxygen species; SCD3, syndecan 3; SCP2, sterol carrier protein 2; TCF, T-cell factor; TNF-α, tumor necrosis factor; TRAIL, tumor necrosis factor-related apoptosis inducing ligand; VEGF, vascular endothelial growth factor; VEGFR, vascular endothelial growth factor receptor; Wnt, Wingless-related integration site; Wtp53, wild type p53; XIAP, X-chromosome-linked inhibitor of apoptosis protein.
Author Contributions: AJ, XS, KN conceived the idea for this article. AJ performed literature search, data analysis and drafted the manuscript. XS, KN and RL critically reviewed the manuscript. All authors read and approved the final manuscript.
The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.
Funding: The author(s) received no financial support for the research, authorship, and/or publication of this article.
ORCID iD: Xiao Qun Su
https://orcid.org/0000-0001-8178-406X
References
- 1. Gustavsson B, Carlsson G, Machover D, et al. A review of the evolution of systemic chemotherapy in the management of colorectal cancer. Clin Colorectal Cancer. 2015;14:1-10. doi: 10.1016/j.clcc.2014.11.002 [DOI] [PubMed] [Google Scholar]
- 2. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2020. CA Cancer J Clin. 2020;70:7-30. doi: 10.3322/caac.21590 [DOI] [PubMed] [Google Scholar]
- 3. Binefa G, Rodriguez-Moranta F, Teule A. Colorectal cancer: from prevention to personalized medicine. World J Gastroenterol. 2014;20:6786-6808. doi: 10.3748/wjg.v20.i22.6786 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Piñol V, Castells A, Andreu M, et al. Accuracy of revised Bethesda guidelines, microsatellite instability, and immunohistochemistry for the identification of patients with hereditary nonpolyposis colorectal cancer. JAMA. 2005;293:1986-1994. doi: 10.1001/jama.293.16.1986 [DOI] [PubMed] [Google Scholar]
- 5. Huxley RR, Ansary-Moghaddam A, Clifton P, et al. The impact of dietary and lifestyle risk factors on risk of colorectal cancer: a quantitative overview of the epidemiological evidence. Int J Cancer. 2009;125:171-180. doi: 10.1002/ijc.24343 [DOI] [PubMed] [Google Scholar]
- 6. Alegria-Lertxundi I, Bujanda L, Arroyo-Izaga M. Role of dairy foods, fish, white meat, and eggs in the prevention of colorectal cancer: a systematic review of observational studies in 2018-2022. Nutrients. 2022;14:3430. doi: 10.3390/nu14163430 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Kunzmann AT, Coleman HG, Huang WY, et al. Fruit and vegetable intakes and risk of colorectal cancer and incident and recurrent adenomas in the PLCO cancer screening trial. Int J Cancer. 2016;138:1851-1861. doi: 10.1002/ijc.29922 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Alegria-Lertxundi I, Aguirre C, Bujanda L, et al. Gene-diet interactions in colorectal cancer: survey design, instruments, participants and descriptive data of a case-control study in the Basque Country. Nutrients. 2020;12:2362. doi: 10.3390/nu12082362 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Veettil SK, Wong TY, Loo YS, et al. Role of Diet in colorectal cancer incidence: umbrella review of meta-analyses of prospective observational studies. JAMA Netw Open. 2021;4:e2037341. doi: 10.1001/jamanetworkopen.2020.37341 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Aglago E, Huybrechts I, Murphy N, 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;18:654-66.e6. doi: 10.1016/j.cgh.2019.06.031 [DOI] [PubMed] [Google Scholar]
- 11. Hu J, La Vecchia C, Negri E, Mery L. Nutrients and risk of colon cancer. Cancers. 2010;2:51-67. doi: 10.3390/cancers2010051 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Lee SWR, Rivadeneira DE, Steele SR, et al. Advanced Colonoscopy And Endoluminal Surgery. Springer International Publishing. 2017:115-132. doi:10,1007/978-3-319-48370-2 [Google Scholar]
- 13. Hamilton SD, Vogelstein B, Kudo S, et al. World Health Organization Classification of Tumours. Pathology and Genetics of Tumours of the Digestive System. IARC Press; 2000:69-92. [Google Scholar]
- 14. Pan SY, Morrison H. Epidemiology of cancer of the small intestine. World J Gastrointest Oncol. 2011;3:1-42. doi: 10.4251/wjgo.v3.i3.33 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Aarons CB, Shanmugan S, Bleier JI. Management of malignant colon polyps: current status and controversies. World J Gastroenterol. 2014;20:16178-16183. doi: 10.3748/wjg.v20.i43.16178 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. East JE, Vieth M, Rex DK. Serrated lesions in colorectal cancer screening: detection, resection, pathology and surveillance. Gut. 2015;64:991-1000. doi: 10.1136/gutjnl-2014-309041 [DOI] [PubMed] [Google Scholar]
- 17. Fearon ER, Vogelstein B. A genetic model for colorectal tumorigenesis. Cell. 1990;61:759-767. [DOI] [PubMed] [Google Scholar]
- 18. Boland CR, Sinicrope FA, Brenner DE, Carethers JM. Colorectal cancer prevention and treatment. Gastroenterology. 2000;118:S115-S128. doi: 10.1016/s0016-5085(00)70010-2 [DOI] [PubMed] [Google Scholar]
- 19. Weisenberger DJ, Siegmund KD, Campan M, et al. CpG island methylator phenotype underlies sporadic microsatellite instability and is tightly associated with BRAF mutation in colorectal cancer. Nat Genet. 2006;38:787-793. doi: 10.1038/ng1834 [DOI] [PubMed] [Google Scholar]
- 20. Katz SC, DeMatteo RP. Gastrointestinal stromal tumors and leiomyosarcomas. J Surg Oncol. 2008;97:350-359. doi: 10.1002/jso.20970 [DOI] [PubMed] [Google Scholar]
- 21. Xie Y-H, Chen Y-X, Fang J-Y. Comprehensive review of targeted therapy for colorectal cancer. Signal Transduct Target Ther. 2020;5:22. doi: 10.1038/s41392-020-0116-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Liu YQ, Wang XL, He DH, Cheng YX. Protection against chemotherapy- and radiotherapy-induced side effects: a review based on the mechanisms and therapeutic opportunities of phytochemicals. Phytomedicine. 2021;80:153402. doi: 10.1016/j.phymed.2020.153402 [DOI] [PubMed] [Google Scholar]
- 23. Karthikeyan A, Joseph A, Nair BG. Promising bioactive compounds from the marine environment and their potential effects on various diseases. J Genet Eng Biotechnol. 2022;20:14. doi: 10.1186/s43141-021-00290-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Manoharan S, Perumal E. Potential role of marine bioactive compounds in cancer signaling pathways: a review. Eur J Pharmacol. 2022;936:175330. doi: 10.1016/j.ejphar.2022.175330 [DOI] [PubMed] [Google Scholar]
- 25. Kuppusamy P, Yusoff MM, Maniam GP, et al. Nutraceuticals as potential therapeutic agents for colon cancer: a review. Acta Pharm Sin B. 2014;4:173-181. doi: 10.1016/j.apsb.2014.04.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Islam MR, Akash S, Rahman MM, et al. Colon cancer and colorectal cancer: prevention and treatment by potential natural products. Chem Biol Interact. 2022;368:110170. doi: 10.1016/j.cbi.2022.110170 [DOI] [PubMed] [Google Scholar]
- 27. Cockbain AJ, Toogood GJ, Hull MA. Omega-3 polyunsaturated fatty acids for the treatment and prevention of colorectal cancer. Gut. 2012;61:135-149. doi: 10.1136/gut.2010.233718 [DOI] [PubMed] [Google Scholar]
- 28. Tu K, Ma T, Zhou R, et al. Association between dietary fatty acid patterns and colorectal cancer risk: a large-scale case-control study in China. Nutrients. 2022;14:4375. doi: 10.3390/nu14204375 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Pericleous M, Mandair D, Caplin ME. Diet and supplements and their impact on colorectal cancer. J Gastrointest Oncol. 2013;4:409-423. doi: 10.3978/j.issn.2078-6891.2013.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Caini S, Chioccioli S, Pastore E, et al. Fish consumption and colorectal cancer risk: meta-analysis of prospective epidemiological studies and review of evidence from Animal Studies. Cancers. 2022;14:640. doi: 10.3390/cancers14030640 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Linseisen J, Grundmann N, Zoller D, et al. Red blood cell fatty acids and risk of colorectal cancer in the European Prospective Investigation into Cancer and Nutrition (EPIC). Cancer Epidemiol Biomarkers Prev. 2021;30:874-885. doi: 10.1158/1055-9965.Epi-20-1426 [DOI] [PubMed] [Google Scholar]
- 32. Aykan NF. Red Meat and colorectal cancer. Oncol Rev. 2015;9:288. doi: 10.4081/oncol.2015.288 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Haggar FA, Boushey RP. Colorectal cancer epidemiology: incidence, mortality, survival, and risk factors. Clin Colon Rectal Surg. 2009;22:191-197. doi: 10.1055/s-0029-1242458 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Goodnight Sh, Jr, Harris WS, Connor WE, Illingworth DR. Polyunsaturated fatty acids, hyperlipidemia, and thrombosis. Arteriosclerosis. 1982;2:87-113. doi: 10.1161/01.atv.2.2.87 [DOI] [PubMed] [Google Scholar]
- 35. Ellulu MS, Khaza’ai H, Abed Y, et al. Role of fish oil in human health and possible mechanism to reduce the inflammation. Inflammopharmacology. 2015;23:79-89. doi: 10.1007/s10787-015-0228-1 [DOI] [PubMed] [Google Scholar]
- 36. Gogus U, Smith C. n-3 omega fatty acids: a review of current knowledge. Int J Food Sci Technol. 2010;45:417-436. doi: 10.1111/j.1365-2621.2009.02151.x [DOI] [Google Scholar]
- 37. Stephenson JA, Al-Taan O, Arshad A, et al. The multifaceted effects of omega-3 polyunsaturated fatty acids on the hallmarks of cancer. Lipids. 2013;2013:261247. doi: 10.1155/2013/261247 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Valenzuela R, Metherel AH, Cisbani G, et al. Protein concentrations and activities of fatty acid desaturase and elongase enzymes in liver, brain, testicle, and kidney from mice: substrate dependency. Biofactors. 2024;50:89-100. doi: 10.1002/biof.1992 [DOI] [PubMed] [Google Scholar]
- 39. Videla LA, Hernandez-Rodas MC, Metherel AH, Valenzuela R. Influence of the nutritional status and oxidative stress in the desaturation and elongation of n-3 and n-6 polyunsaturated fatty acids: impact on non-alcoholic fatty liver disease. Prostaglandins Leukot Essent Fatty Acids. 2022;181:102441. doi: 10.1016/j.plefa.2022.102441 [DOI] [PubMed] [Google Scholar]
- 40. Kaur N, Chugh V, Gupta AK. Essential fatty acids as functional components of foods- a review. J Food Sci Technol. 2012;51:2289-2303. doi: 10.1007/s13197-012-0677-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Brenna JT. Efficiency of conversion of α-linolenic acid to long chain n-3 fatty acids in man. Curr Opin Clin Nutr Metab Care. 2002;5:127-132. doi: 10.1097/00075197-200203000-00002 [DOI] [PubMed] [Google Scholar]
- 42. Burdge G. ??-linolenic acid metabolism in men and women: nutritional and biological implications. Curr Opin Clin Nutr Metab Care. 2004;7:137-144. doi: 10.1097/00075197-200403000-00006 [DOI] [PubMed] [Google Scholar]
- 43. Freeman MP, Hibbeln JR, Wisner KL, et al. Omega-3 fatty acids: evidence basis for treatment and future research in psychiatry. J Clin Psychiatry. 2006;67:1954-1967. doi: 10.4088/jcp.v67n1217 [DOI] [PubMed] [Google Scholar]
- 44. Rodríguez M, G Rebollar P, Mattioli S, Castellini C. n-3 PUFA sources (Precursor/Products): a review of current knowledge on Rabbit. Animals. 2019;9:806. doi: 10.3390/ani9100806 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Roynette CE, Calder PC, Dupertuis YM, Pichard C. n-3 polyunsaturated fatty acids and colon cancer prevention. Clin Nutr. 2004;23:139-151. doi: 10.1016/j.clnu.2003.07.005 [DOI] [PubMed] [Google Scholar]
- 46. Nieves D, Moreno JJ. Effect of arachidonic and eicosapentaenoic acid metabolism on RAW 264.7 macrophage proliferation. J Cell Physiol. 2006;208:428-434. doi: 10.1002/jcp.20678 [DOI] [PubMed] [Google Scholar]
- 47. Cabral M, Martín-Venegas R, Moreno JJ. Leukotriene d4-induced caco-2 cell proliferation is mediated by prostaglandin E2 synthesis. Physiol Rep. 2015;3:e12417. doi: 10.14814/phy2.12417 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Cabral M, Martín-Venegas R, Moreno JJ. Role of arachidonic acid metabolites on the control of non-differentiated intestinal epithelial cell growth. Int J Biochem Cell Biol. 2013;45:1620-1628. doi: 10.1016/j.biocel.2013.05.009 [DOI] [PubMed] [Google Scholar]
- 49. Koontongkaew S, Monthanapisut P, Saensuk T. Inhibition of arachidonic acid metabolism decreases tumor cell invasion and matrix metalloproteinase expression. Prostaglandins Other Lipid Mediat. 2010;93:100-108. doi: 10.1016/j.prostaglandins.2010.07.002 [DOI] [PubMed] [Google Scholar]
- 50. Nakanishi M, Rosenberg DW. Multifaceted roles of PGE2 in inflammation and cancer. Semin Immunopathol. 2013;35:123-137. doi: 10.1007/s00281-012-0342-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Wang D, DuBois RN. An inflammatory mediator, prostaglandin E2, in colorectal cancer. Cancer. 2013;19:502-510. doi: 10.1097/PPO.0000000000000003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Wisastra R, Dekker FJ. Inflammation, cancer and oxidative lipoxygenase activity are intimately linked. Cancers. 2014;6:1500-1521. doi: 10.3390/cancers6031500 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Zhao Y, Wang W, Wang Q, Zhang X, Ye L. Lipid metabolism enzyme 5-LOX and its metabolite LTB4 are capable of activating transcription factor NF-κB in hepatoma cells. Biochem Biophys Res Commun. 2012;418:647-651. doi: 10.1016/j.bbrc.2012.01.068 [DOI] [PubMed] [Google Scholar]
- 54. Cui J, Shan K, Yang Q, et al. Prostaglandin E(3) attenuates macrophage-associated inflammation and prostate tumour growth by modulating polarization. J Cell Mol Med. 2021;25:5586-5601. doi: 10.1111/jcmm.16570 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Hull MA. Omega-3 polyunsaturated fatty acids. Best Pract Res Clin Gastroenterol. 2011;25:547-554. doi: 10.1016/j.bpg.2011.08.001 [DOI] [PubMed] [Google Scholar]
- 56. Rodríguez-Lagunas MJ, Ferrer R, Moreno JJ. Effect of eicosapentaenoic acid-derived prostaglandin E3 on intestinal epithelial barrier function. Prostaglandins Leukot Essent Fatty Acids. 2013;88:339-345. doi: 10.1016/j.plefa.2013.02.001 [DOI] [PubMed] [Google Scholar]
- 57. Storniolo CE, Cabral M, Busquets MA, Martín-Venegas R, Moreno JJ. Dual behavior of long-chain fatty acids and their cyclooxygenase/lipoxygenase metabolites on human intestinal caco-2 cell growth. Front Pharmacol. 2020;11:529976. doi: 10.3389/fphar.2020.529976 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Dong L, Zou H, Yuan C, et al. Different fatty acids compete with arachidonic acid for binding to the allosteric or catalytic subunits of cyclooxygenases to regulate prostanoid synthesis. J Biol Chem. 2016;291:4069-4078. doi: 10.1074/jbc.M115.698001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Arshad Z, Rezapour-Firouzi S, Mohammadian M, Ebrahimifar E. The sources of essential fatty acids for allergic and cancer patients; a connection with insight into mammalian target of rapamycin: a narrative review. Asian Pac J Cancer Prev. 2018;19:2391-2401. doi: 10.22034/apjcp.2018.19.9.2391 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Wang J, Yu JC, Kang WM, Ma ZQ. Superiority of a fish oil-enriched emulsion to medium-chain triacylglycerols/long-chain triacylglycerols in gastrointestinal surgery patients: a randomized clinical trial. Nutrition. 2012;28:623-629. doi: 10.1016/j.nut.2011.08.004 [DOI] [PubMed] [Google Scholar]
- 61. Sorensen LS, Thorlacius-Ussing O, Rasmussen HH, et al. Effects of perioperative supplementation with omega-3 fatty acids on leukotriene B4 and leukotriene B5 production by stimulated neutrophils in patients with colorectal cancer: a randomized, placebo-controlled intervention trial. Nutrients. 2014;6:4043-4057. doi: 10.3390/nu6104043 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Moro K, Nagahashi M, Ramanathan R, Takabe K, Wakai T. Resolvins and omega three polyunsaturated fatty acids: clinical implications in inflammatory diseases and cancer. World J Clin Cases. 2016;4:155-164. doi: 10.12998/wjcc.v4.i7.155 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Serhan CN, Dalli J, Colas RA, Winkler JW, Chiang N. Protectins and maresins: new pro-resolving families of mediators in acute inflammation and resolution bioactive metabolome. Biochim Biophys Acta. 2015;1851:397-413. doi: 10.1016/j.bbalip.2014.08.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Allred CD, Talbert DR, Southard RC, Wang X, Kilgore MW. PPARgamma1 as a molecular target of eicosapentaenoic acid in human colon cancer (HT-29) cells. J Nutr. 2008;138:250-256. doi: 10.1093/jn/138.2.250 [DOI] [PubMed] [Google Scholar]
- 65. Calviello G, Di Nicuolo F, Gragnoli S, et al. n-3 PUFAs reduce VEGF expression in human colon cancer cells modulating the COX-2/PGE2 induced ERK-1 and -2 and HIF-1alpha induction pathway. Carcinogenesis. 2004;25:2303-2310. doi: 10.1093/carcin/bgh265 [DOI] [PubMed] [Google Scholar]
- 66. Rogers KR, Kikawa KD, Mouradian M, et al. Docosahexaenoic acid alters epidermal growth factor receptor-related signaling by disrupting its lipid raft association. Carcinogenesis. 2010;31:1523-1530. doi: 10.1093/carcin/bgq111 [DOI] [PubMed] [Google Scholar]
- 67. Zhang K, Hu Z, Qi H, et al. G-protein-coupled receptors mediate ω-3 pufas-inhibited colorectal cancer by activating the Hippo pathway. Oncotarget. 2016;7:58315-58330. doi: 10.18632/oncotarget.11089 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Sam MR, Tavakoli-Mehr M, Safaralizadeh R. Omega-3 fatty acid DHA modulates p53, survivin, and microRNA-16-1 expression in KRAS-mutant colorectal cancer stem-like cells. Genes Nutr. 2018;13:8. doi: 10.1186/s12263-018-0596-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Murad LB, da Silva Nogueira P, de Araújo WM, et al. Docosahexaenoic acid promotes cell cycle arrest and decreases proliferation through WNT/β-catenin modulation in colorectal cancer cells exposed to γ-radiation. Biofactors. 2019;45:24-34. doi: 10.1002/biof.1455 [DOI] [PubMed] [Google Scholar]
- 70. Fasano E, Serini S, Piccioni E, et al. DHA induces apoptosis by altering the expression and cellular location of GRP78 in colon cancer cell lines. Biochim Biophys Acta. 2012;1822:1762-1772. doi: 10.1016/j.bbadis.2012.08.003 [DOI] [PubMed] [Google Scholar]
- 71. Sasaki T, Hiroki K, Yamashita Y. The role of epidermal growth factor receptor in cancer metastasis and microenvironment. Biomed Res Int. 2013;2013:546318. doi: 10.1155/2013/546318 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Turk HF, Barhoumi R, Chapkin RS. Alteration of EGFR spatiotemporal dynamics suppresses signal transduction. PLoS One. 2012;7:e39682. doi: 10.1371/journal.pone.0039682 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Herbst RS. Review of epidermal growth factor receptor biology. Int J Radiat Oncol Biol Phys. 2004;59:21-26. doi: 10.1016/j.ijrobp.2003.11.041 [DOI] [PubMed] [Google Scholar]
- 74. Wee P, Wang Z. Epidermal growth factor receptor cell proliferation signaling pathways. Cancers. 2017;9:52. doi: 10.3390/cancers9050052 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Seshacharyulu P, Ponnusamy MP, Haridas D, et al. Targeting the EGFR signaling pathway in cancer therapy. Expert Opin Ther Targets. 2012;16:15-31. doi: 10.1517/14728222.2011.648617 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. De Stefano A, Carlomagno C. Beyond KRAS: predictive factors of the efficacy of anti-EGFR monoclonal antibodies in the treatment of metastatic colorectal cancer. World J Gastroenterol. 2014;20:9732-9743. doi: 10.3748/wjg.v20.i29.9732 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Jayathilake AG, Veale MF, Luwor RB, Nurgali K, Su XQ. Krill oil extract inhibits the migration of human colorectal cancer cells and down-regulates EGFR signalling and PD-L1 expression. BMC Complement Med Ther. 2020;20:372. doi: 10.1186/s12906-020-03160-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Bendardaf R, El-Serafi A, Syrjänen K, Collan Y, Pyrhönen S. The effect of vascular endothelial growth factor-1 expression on survival of advanced colorectal cancer patients. Libyan J Med. 2017;12:1290741. doi: 10.1080/19932820.2017.1290741 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Karar J, Maity A. PI3K/AKT/mTOR pathway in angiogenesis. Front Mol Neurosci. 2011;4:51. doi: 10.3389/fnmol.2011.00051 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Nör JE, Christensen J, Mooney DJ, Polverini PJ. Vascular endothelial growth factor (VEGF)-mediated angiogenesis is associated with enhanced endothelial cell survival and induction of Bcl-2 expression. Am J Pathol. 1999;154:375-384. doi: 10.1016/s0002-9440(10)65284-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Pidgeon GP, Barr MP, Harmey JH, Foley DA, Bouchier-Hayes DJ. Vascular endothelial growth factor (VEGF) upregulates BCL-2 and inhibits apoptosis in human and murine mammary adenocarcinoma cells. Br J Cancer. 2001;85:273-278. doi: 10.1054/bjoc.2001.1876 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Hawcroft G, Loadman PM, Belluzzi A, Hull MA. Effect of eicosapentaenoic acid on E-type prostaglandin synthesis and EP4 receptor signaling in human colorectal cancer cells. Neoplasia. 2010;12:618-627. doi:10.1593%2Fneo.10388 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Kumar S, Sharma B, Sharma P, Agnihotri N. n-3 PUFAs: an Elixir in prevention of colorectal cancer. Curr Colorectal Cancer Rep. 2015;11:141-149. doi: 10.1007/s11888-015-0268-3 [DOI] [Google Scholar]
- 84. Chell SD, Witherden IR, Dobson RR, et al. Increased EP4 receptor expression in colorectal cancer progression promotes cell growth and anchorage independence. Cancer Res. 2006;66:3106-3113. doi: 10.1158/0008-5472.CAN-05-3702 [DOI] [PubMed] [Google Scholar]
- 85. Hawcroft G, Ko CW, Hull MA. Prostaglandin E2-EP4 receptor signalling promotes tumorigenic behaviour of HT-29 human colorectal cancer cells. Oncogene. 2007;26:3006-3019. doi: 10.1038/sj.onc.1210113 [DOI] [PubMed] [Google Scholar]
- 86. Dimmeler S, Hermann C, Galle J, Zeiher AM. Upregulation of superoxide dismutase and nitric oxide synthase mediates the apoptosis-suppressive effects of shear stress on endothelial cells. Arterioscler Thromb Vasc Biol. 1999;19:656-664. doi: 10.1161/01.ATV.19.3.656 [DOI] [PubMed] [Google Scholar]
- 87. Rössig L, Fichtlscherer B, Breitschopf K, et al. Nitric oxide inhibits caspase-3 by S-nitrosation in vivo. J Biol Chem. 1999;274:6823-6826. doi: 10.1074/jbc.274.11.6823 [DOI] [PubMed] [Google Scholar]
- 88. Cianchi F, Cortesini C, Bechi P, et al. Up-regulation of cyclooxygenase 2 gene expression correlates with tumor angiogenesis in human colorectal cancer. Gastroenterology. 2001;121:1339-1347. doi: 10.1053/gast.2001.29691 [DOI] [PubMed] [Google Scholar]
- 89. Zhang C, Yu H, Ni X, Shen S, Das UN. Growth inhibitory effect of polyunsaturated fatty acids (pufas) on colon cancer cells via their growth inhibitory metabolites and fatty acid composition changes. PLoS One. 2015;10:e0123256. doi: 10.1371/journal.pone.0123256 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Huang Z, Liu CA, Cai PZ, et al. Omega-3PUFA attenuates MNU-Induced colorectal cancer in rats by blocking PI3K/AKT/Bcl-2 signaling. Onco Targets Ther. 2020;13:1953-1965. doi: 10.2147/ott.S241298 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Park JM, Kwon SH, Han YM, Hahm KB, Kim EH. Omega-3 polyunsaturated fatty acids as potential chemopreventive agent for gastrointestinal cancer. J Cancer Prev. 2013;18:201-208. doi:10.15430%2FJCP.2013.18.3.201 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Matesanz N, Park G, McAllister H, et al. Docosahexaenoic acid improves the nitroso-redox balance and reduces VEGF-mediated angiogenic signaling in microvascular endothelial cells. Investig Ophthalmol Vis Sci. 2010;51:6815-6825. doi: 10.1167/iovs.10-5339 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. de Lima TM, de Sa Lima L, Scavone C, Curi R. Fatty acid control of nitric oxide production by macrophages. FEBS Lett. 2006;580:3287-3295. doi: 10.1016/j.febslet.2006.04.091 [DOI] [PubMed] [Google Scholar]
- 94. Narayanan BA, Narayanan NK, Simi B, Reddy BS. Modulation of inducible nitric oxide synthase and related proinflammatory genes by the omega-3 fatty acid docosahexaenoic acid in human colon cancer cells. Cancer Res. 2003;63:972-979. [PubMed] [Google Scholar]
- 95. Tyagi S, Gupta P, Saini AS, Kaushal C, Sharma S. The peroxisome proliferator-activated receptor: a family of nuclear receptors role in various diseases. J Adv Pharm Technol Res. 2011;2:236-240. doi: 10.4103/2231-4040.90879 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Hernández-Rodas MC, Valenzuela R, Echeverría F, et al. Supplementation with docosahexaenoic acid and extra virgin olive oil prevents liver steatosis induced by a High-Fat diet in mice through PPAR-α and nrf2 upregulation with concomitant SREBP-1c and NF-kB downregulation. Mol Nutr Food Res. 2017;61:1700479. doi: 10.1002/mnfr.201700479 [DOI] [PubMed] [Google Scholar]
- 97. Soto-Alarcón SA, Ortiz M, Orellana P, et al. Docosahexaenoic acid and hydroxytyrosol co-administration fully prevents liver steatosis and related parameters in mice subjected to high-fat diet: a molecular approach. Biofactors. 2019;45:930-943. doi: 10.1002/biof.1556 [DOI] [PubMed] [Google Scholar]
- 98. Echeverría F, Ortiz M, Valenzuela R, Videla LA. Long-chain polyunsaturated fatty acids regulation of PPARs, signaling: relationship to tissue development and aging. Prostaglandins Leukot Essent Fatty Acids. 2016;114:28-34. doi: 10.1016/j.plefa.2016.10.001 [DOI] [PubMed] [Google Scholar]
- 99. Beyer MP, Videla LA, Farías C, Valenzuela R. Potential clinical applications of pro-resolving lipids mediators from docosahexaenoic acid. Nutrients. 2023;15:3317. doi: 10.3390/nu15153317 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Chambrier C, Bastard JP, Rieusset J, et al. Eicosapentaenoic acid induces mRNA expression of peroxisome proliferator-activated receptor gamma. Obes Res. 2002;10:518-525. doi: 10.1038/oby.2002.70 [DOI] [PubMed] [Google Scholar]
- 101. Calviello G, Resci F, Serini S, et al. Docosahexaenoic acid induces proteasome-dependent degradation of beta-catenin, down-regulation of survivin and apoptosis in human colorectal cancer cells not expressing COX-2. Carcinogenesis. 2007;28:1202-1209. doi: 10.1093/carcin/bgl254 [DOI] [PubMed] [Google Scholar]
- 102. Toit-Kohn JL, Louw L, Engelbrecht AM. Docosahexaenoic acid induces apoptosis in colorectal carcinoma cells by modulating the PI3 kinase and p38 MAPK pathways. J Nutr Biochem. 2009;20:106-114. doi: 10.1016/j.jnutbio.2007.12.005 [DOI] [PubMed] [Google Scholar]
- 103. Moradi Sarabi M, Zahedi SA, Pajouhi N, et al. The effects of dietary polyunsaturated fatty acids on miR-126 promoter DNA methylation status and VEGF protein expression in the colorectal cancer cells. Genes Nutr. 2018;13:32. doi: 10.1186/s12263-018-0623-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Weng WH, Leung WH, Pang YJ, Kuo LW, Hsu HH. EPA significantly improves anti-EGFR targeted therapy by regulating miR-378 expression in colorectal cancer. Oncol Lett. 2018;16:6188-6194. doi: 10.3892/ol.2018.9408 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. Gelsomino G, Corsetto PA, Campia I, et al. Omega 3 fatty acids chemosensitize multidrug resistant colon cancer cells by down-regulating cholesterol synthesis and altering detergent resistant membranes composition. Mol Cancer. 2013;12:137. doi: 10.1186/1476-4598-12-137 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Hawcroft G, Volpato M, Marston G, 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 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107. D’Angelo L, Piazzi G, Pacilli A, et al. A combination of eicosapentaenoic acid-free fatty acid, epigallocatechin-3-gallate and proanthocyanidins has a strong effect on mTOR signaling in colorectal cancer cells. Carcinogenesis. 2014;35:2314-2320. doi: 10.1093/carcin/bgu173 [DOI] [PubMed] [Google Scholar]
- 108. Cai Y, Liu J, Cai SK, et al. Eicosapentaenoic acid’s metabolism of 15-LOX-1 promotes the expression of miR-101 thus inhibits Cox2 pathway in colon cancer. Onco Targets Ther. 2020;13:5605-5616. doi: 10.2147/ott.S237562 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Morin C, Rousseau Fortin S. Anti-proliferative effects of a new docosapentaenoic acid monoacylglyceride in colorectal carcinoma cells. Prostaglandins Leukot Essent Fatty Acids. 2013;89:203-213. doi: 10.1016/j.plefa.2013.07.004 [DOI] [PubMed] [Google Scholar]
- 110. Pfister E, Smith R, Lane MA. N-3 polyunsaturated fatty acid ethyl esters decrease the invasion, but not the proliferation, of human colorectal cancer cells via a PI3K-dependent mechanism in vitro. Prostaglandins Leukot Essent Fatty Acids. 2021;167:102273. doi: 10.1016/j.plefa.2021.102273 [DOI] [PubMed] [Google Scholar]
- 111. Cotter TG. Apoptosis and cancer: the genesis of a research field. Nat Rev Cancer. 2009;9:501-507. doi: 10.1038/nrc2663 [DOI] [PubMed] [Google Scholar]
- 112. D’Eliseo D, Velotti F. Omega-3 fatty acids and cancer cell cytotoxicity: implications for multi-targeted cancer therapy. J Clin Med. 2016;5:15. doi: 10.3390/jcm5020015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Logue SE, Cleary P, Saveljeva S, Samali A. New directions in ER stress-induced cell death. Apoptosis. 2013;18:537-546. doi: 10.1007/s10495-013-0818-6 [DOI] [PubMed] [Google Scholar]
- 114. Gupta S, Kass GE, Szegezdi E, Joseph B. The mitochondrial death pathway: a promising therapeutic target in diseases. J Cell Mol Med. 2009;13:1004-1033. doi: 10.1111/j.1582-4934.2009.00697.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Jin Z, El-Deiry WS. Overview of cell death signaling pathways. Cancer Biol Ther. 2005;4:139-163. doi: 10.4161/cbt.4.2.1508 [DOI] [PubMed] [Google Scholar]
- 116. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144:646-674. doi: 10.1016/j.cell.2011.02.013 [DOI] [PubMed] [Google Scholar]
- 117. Lowe SW, Cepero E, Evan G. Intrinsic tumour suppression. Nature. 2004;432:307-315. doi: 10.1038/nature03098 [DOI] [PubMed] [Google Scholar]
- 118. O’Brien MA, Kirby R. Apoptosis: a review of pro-apoptotic and anti-apoptotic pathways and dysregulation in disease. J Vet Emerg Crit Care. 2008;18:572-585. doi:org/10.1111/j.1476-4431.2008.00363.x [Google Scholar]
- 119. Giorgi C, Baldassari F, Bononi A, et al. Mitochondrial Ca(2+) and apoptosis. Cell Calcium. 2012;52:36-43. doi: 10.1016/j.ceca.2012.02.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. Li P, Nijhawan D, Budihardjo I, et al. Cytochrome c and datp-dependent formation of apaf-1/caspase-9 complex initiates an apoptotic protease cascade. Cell. 1997;91:479-489. doi: 10.1016/s0092-8674(00)80434-1 [DOI] [PubMed] [Google Scholar]
- 121. Galluzzi L, Vitale I, Abrams JM, et al. Molecular definitions of cell death subroutines: recommendations of the nomenclature committee on cell death 2012. Cell Death Differ. 2012;19:107-120. doi: 10.1038/cdd.2011.96 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122. Wang X. The expanding role of mitochondria in apoptosis. Genes Dev. 2001;15:2922-2933. http://genesdev.cshlp.org/content/15/22/2922.full [PubMed] [Google Scholar]
- 123. Elmore S. Apoptosis: a review of programmed cell death. Toxicol Pathol. 2007;35:495-516. doi: 10.1080/01926230701320337 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124. Li J, Lee B, Lee AS. Endoplasmic reticulum stress-induced apoptosis: multiple pathways and activation of p53-up-regulated modulator of apoptosis (PUMA) and NOXA by p53. J Biol Chem. 2006;281:7260-7270. doi: 10.1074/jbc.M509868200 [DOI] [PubMed] [Google Scholar]
- 125. Iurlaro R, Muñoz-Pinedo C. Cell death induced by endoplasmic reticulum stress. FEBS J. 2016;283:2640-2652. doi: 10.1111/febs.13598 [DOI] [PubMed] [Google Scholar]
- 126. Danbara N, Yuri T, Tsujita-Kyutoku M, et al. Conjugated docosahexaenoic acid is a potent inducer of cell cycle arrest and apoptosis and inhibits growth of colo 201 human colon cancer cells. Nutr Cancer. 2004;50:71-79. doi: 10.1207/s15327914nc5001_10 [DOI] [PubMed] [Google Scholar]
- 127. Giros A, Grzybowski M, Sohn VR, et al. Regulation of colorectal cancer cell apoptosis by the n-3 polyunsaturated fatty acids docosahexaenoic and Eicosapentaenoic. Cancer Prev Res. 2009;2:732-742. doi: 10.1158/1940-6207.CAPR-08-0197 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128. Granci V, Cai F, Lecumberri E, et al. Colon cancer cell chemosensitisation by fish oil emulsion involves apoptotic mitochondria pathway. Br J Nutr. 2013;109:1188-1195. doi: 10.1017/S000711451200308X [DOI] [PubMed] [Google Scholar]
- 129. Llor X, Pons E, Roca A, et al. The effects of fish oil, olive oil, oleic acid and linoleic acid on colorectal neoplastic processes. Clin Nutr. 2003;22:71-79. doi: 10.1054/clnu.2002.0627 [DOI] [PubMed] [Google Scholar]
- 130. Skender B, Hofmanová J, Slavík J, et al. DHA-mediated enhancement of TRAIL-induced apoptosis in colon cancer cells is associated with engagement of mitochondria and specific alterations in sphingolipid metabolism. BBA. 2014;1841:1308-1317. doi: 10.1016/j.bbalip.2014.06.005 [DOI] [PubMed] [Google Scholar]
- 131. Slagsvold JE, Pettersen CH, Størvold GL, et al. DHA alters expression of target proteins of cancer therapy in chemotherapy resistant SW620 colon cancer cells. Nutr Cancer. 2010;62:611-621. doi: 10.1080/01635580903532366 [DOI] [PubMed] [Google Scholar]
- 132. Zhang C, Yu H, Shen Y, et al. Polyunsaturated fatty acids trigger apoptosis of colon cancer cells through a mitochondrial pathway. Arch Med Sci. 2015;11:1081-1094. doi: 10.5114/aoms.2015.54865 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133. Hofmanová J, Vaculová A, Kozubík A. Polyunsaturated fatty acids sensitize human colon adenocarcinoma HT-29 cells to death receptor-mediated apoptosis. Cancer Lett. 2005;218:33-41. doi: 10.1016/j.canlet.2004.07.038 [DOI] [PubMed] [Google Scholar]
- 134. Narayanan BA, Narayanan NK, Reddy BS. Docosahexaenoic acid regulated genes and transcription factors inducing apoptosis in human colon cancer cells. Int J Oncol. 2001;19:1255-1262. doi: 10.3892/ijo.19.6.1255 [DOI] [PubMed] [Google Scholar]
- 135. D’Eliseo D, Di Rocco G, Loria R, et al. Epitelial-to-mesenchimal transition and invasion are upmodulated by tumor-expressed granzyme B and inhibited by docosahexaenoic acid in human colorectal cancer cells. J Exp Clin Cancer Res. 2016;35:24. doi: 10.1186/s13046-016-0302-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136. Jakobsen CH, Størvold GL, Bremseth H, et al. DHA induces ER stress and growth arrest in human colon cancer cells: associations with cholesterol and calcium homeostasis. J Lipids. 2008;49:2089-2100. doi: 10.1194/jlr.M700389-JLR200 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137. Hosseini F, Sam MR, Jabbari N, Mozdarani H. Modulating survivin as a radioresistant Factor, caspase-3, and apoptosis by Omega-3 docosahexaenoic acid sensitizes mutant-p53 colorectal cancer cells to γ-Irradiation. Cancer Biother Radiopharm. 2018;33:387-395. doi: 10.1089/cbr.2018.2445 [DOI] [PubMed] [Google Scholar]
- 138. Ahangar P, Sam M, Nejati V, et al. Treatment of undifferentiated colorectal cancer cells with fish-oil derived docosahexaenoic acid triggers caspase-3 activation and apoptosis. Cells with fish-oil derived docosahexaenoic acid triggers caspase-3 activation and apoptosis. J Cancer Res Ther. 2016;12:798-804. doi: 10.4103/0973-1482.157326 [DOI] [PubMed] [Google Scholar]
- 139. Sam MR, Ahangar P, Nejati V, Habibian R. Treatment of LS174T colorectal cancer stem-like cells with n-3 PUFAs induces growth suppression through inhibition of survivin expression and induction of caspase-3 activation. Cell Oncol. 2016;39:69-77. doi: 10.1007/s13402-015-0254-4 [DOI] [PubMed] [Google Scholar]
- 140. Jayathilake AG, Senior PV, Su XQ. Krill oil extract suppresses cell growth and induces apoptosis of human colorectal cancer cells. BMC Complement Altern Med. 2016;16:328. doi: 10.1186/s12906-016-1311-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141. Jayathilake AG, Kadife E, Luwor RB, Nurgali K, Su XQ. Krill oil extract suppresses the proliferation of colorectal cancer cells through activation of caspase 3/9. Nutr Metab. 2019;16:53. doi: 10.1186/s12986-019-0382-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142. Bruce A, Julian L, Martin R, et al. (eds.). The cell cycle and programmed cell death. In: Molecular Biology of the Cell, 5th ed. 2008;1060-1065. http://www.garlandscience.com/textbooks/0815341059.asp [Google Scholar]
- 143. Collins K, Jacks T, Pavletich NP. The cell cycle and cancer. Proc Natl Acad Sci USA. 1997;94:2776-2778. doi:10.1073%2Fpnas.94.7.2776 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144. Huret JL, Ahmad M, Arsaban M, et al. Atlas of genetics and cytogenetics in oncology and haematology in 2013. Nucleic Acids Res. 2013;41:D920-D924. doi: 10.1093/nar/gks1082 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145. Sherr CJ, Beach D, Shapiro GI. Targeting CDK4 and CDK6: from discovery to therapy. Cancer Discov. 2016;6:353-367. doi: 10.1158/2159-8290.CD-15-0894 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146. Cánepa ET, Scassa ME, Ceruti JM, et al. INK4 proteins, a family of mammalian CDK inhibitors with novel biological functions. IUBMB Life. 2007;59:419-426. doi: 10.1080/15216540701488358 [DOI] [PubMed] [Google Scholar]
- 147. Sherr CJ, Roberts JM. CDK inhibitors: positive and negative regulators of g1-phase progression. Genes Dev. 1999;13:1501-1512. doi: 10.1101/gad.13.12.1501 [DOI] [PubMed] [Google Scholar]
- 148. Chen ZY, Istfan NW. Docosahexaenoic acid, a major constituent of fish oil diets, prevents activation of cyclin-dependent kinases and S-phase entry by serum stimulation in HT-29 cells. Prostaglandins Leukot Essent Fatty Acids. 2001;64:67-73. doi: 10.1054/plef.2000.0239 [DOI] [PubMed] [Google Scholar]
- 149. Sarotra P, Kansal S, Sandhir R, Agnihotri N. Chemopreventive effect of different ratios of fish oil and corn oil on prognostic markers, DNA damage and cell cycle in colon carcinogenesis. Eur J Cancer Prev. 2012;21:147-154. doi: 10.1097/CEJ.0b013e32834c9bfb [DOI] [PubMed] [Google Scholar]
- 150. Kato T, Kolenic N, Pardini RS. Docosahexaenoic acid (DHA), a primary tumor suppressive omega-3 fatty acid, inhibits growth of colorectal cancer independent of p53 mutational status. Nutr Cancer. 2007;58:178-187. doi: 10.1080/01635580701328362 [DOI] [PubMed] [Google Scholar]
- 151. Schønberg SA, Lundemo AG, Fladvad T, et al. Closely related colon cancer cell lines display different sensitivity to polyunsaturated fatty acids, accumulate different lipid classes and downregulate sterol regulatory element-binding protein 1. FEBS J. 2006;273:2749-2765. doi: 10.1111/j.1742-4658.2006.05292.x [DOI] [PubMed] [Google Scholar]
- 152. Jordan A, Stein J. Effect of an omega-3 fatty acid containing lipid emulsion alone and in combination with 5-fluorouracil (5-FU) on growth of the colon cancer cell line caco-2. Eur J Nutr. 2003;42:324-331. doi: 10.1007/s00394-003-0427-1 [DOI] [PubMed] [Google Scholar]
- 153. Dubois R. Role of inflammation and inflammatory mediators in colorectal cancer. Am Clin Climatol. 2014;125:358-372. [PMC free article] [PubMed] [Google Scholar]
- 154. Maihöfner C, Charalambous MP, Bhambra U, et al. Expression of cyclooxygenase-2 parallels expression of interleukin-1beta, interleukin-6 and NF-kappab in human colorectal cancer. Carcinogenesis. 2003;24:665-671. doi:org/10.1093/carcin/bgg006 [DOI] [PubMed] [Google Scholar]
- 155. McConnell BB, Yang VW. The role of inflammation in the pathogenesis of colorectal cancer. Curr Colorectal Cancer Rep. 2009;5:69-74. doi: 10.1007/s11888-009-0011-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156. Lu H, Ouyang W, Huang C. Inflammation, a key event in cancer development. Mol Cancer. 2006;4:221-233. doi: 10.1158/1541-7786.MCR-05-0261 [DOI] [PubMed] [Google Scholar]
- 157. Diakos CI, Charles KA, McMillan DC, Clarke SJ. Cancer-related inflammation and treatment effectiveness. Lancet Oncol. 2014;15:e493-e503. doi: 10.1016/S1470-2045(14)70263-3 [DOI] [PubMed] [Google Scholar]
- 158. Glass CK, Olefsky JM. Inflammation and lipid signaling in the etiology of insulin resistance. Cell Metab. 2012;15:635-645. doi: 10.1016/j.cmet.2012.04.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159. Ben-Baruch A. Inflammation-associated immune suppression in cancer: the roles played by cytokines, chemokines and additional mediators. Semin Cancer Biol. 2006;16:38-52. doi: 10.1016/j.semcancer.2005.07.006 [DOI] [PubMed] [Google Scholar]
- 160. Coussens LM, Werb Z. Inflammation and cancer. Nature. 2002;420:860-867. doi: 10.1038/nature01322 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161. Grivennikov SI, Greten FR, Karin M. Immunity, inflammation, and cancer. Cell. 2010;140:883-899. doi: 10.1016/j.cell.2010.01.025 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162. Lin WW, Karin M. A cytokine-mediated link between innate immunity, inflammation, and cancer. J Clin Investig. 2007;117:1175-1183. doi: 10.1172/JCI31537 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163. Smyth MJ, Cretney E, Kershaw MH, Hayakawa Y. Cytokines in cancer immunity and immunotherapy. Immunol Rev. 2004;202:275-293. doi: 10.1111/j.0105-2896.2004.00199.x [DOI] [PubMed] [Google Scholar]
- 164. Görgün G, Samur MK, Cowens KB, et al. Lenalidomide enhances immune checkpoint blockade-induced immune response in multiple myeloma. Clin Cancer Res. 2015;21:4607-4618. doi: 10.1158/1078-0432.Ccr-15-0200 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165. Khadge S, Sharp JG, McGuire TR, et al. Immune regulation and anti-cancer activity by lipid inflammatory mediators. Int Immunopharmacol. 2018;65:580-592. doi: 10.1016/j.intimp.2018.10.026 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166. Mori TA, Beilin LJ. Omega-3 fatty acids and inflammation. Curr Atheroscler Rep. 2004;6:461-467. doi: 10.1007/s11883-004-0087-5 [DOI] [PubMed] [Google Scholar]
- 167. Storniolo CE, Pequera M, Vilariño A, Moreno JJ. Specialized pro-resolvin mediators induce cell growth and improve wound repair in intestinal epithelial caco-2 cell cultures. Prostaglandins Leukot Essent Fatty Acids. 2022;187:102520. doi: 10.1016/j.plefa.2022.102520 [DOI] [PubMed] [Google Scholar]
- 168. Weiss G, Meyer F, Matthies B, et al. Immunomodulation by perioperative administration of n-3 fatty acids. Br J Nutr. 2002;87 Suppl 1:S89-S94. doi: 10.1079/bjn2001461 [DOI] [PubMed] [Google Scholar]
- 169. Dupertuis YM, Meguid MM, Pichard C. Colon cancer therapy: new perspectives of nutritional manipulations using polyunsaturated fatty acids. Curr Opin Clin Nutr Metab Care. 2007;10:427-432. doi: 10.1097/MCO.0b013e3281e2c9d4 [DOI] [PubMed] [Google Scholar]
- 170. Fürstenberger G, Krieg P, Müller-Decker K, Habenicht AJ. What are cyclooxygenases and lipoxygenases doing in the driver’s seat of carcinogenesis? Int J Cancer. 2006;119:2247-2254. doi: 10.1002/ijc.22153 [DOI] [PubMed] [Google Scholar]
- 171. Roy J, Le Guennec JY, Galano JM, et al. Non-enzymatic cyclic oxygenated metabolites of omega-3 polyunsaturated fatty acid: bioactive drugs? Biochimie. 2016;120:56-61. doi: 10.1016/j.biochi.2015.06.010 [DOI] [PubMed] [Google Scholar]
- 172. Habbel P, Weylandt KH, Lichopoj K, et al. Docosahexaenoic acid suppresses arachidonic acid-induced proliferation of LS-174T human colon carcinoma cells. World J Gastroenterol. 2009;15:1079-1084. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173. Calviello G, Di Nicuolo F, Serini S, et al. Docosahexaenoic acid enhances the susceptibility of human colorectal cancer cells to 5-fluorouracil. Cancer Chemother Pharmacol. 2005;55:12-20. doi: 10.1007/s00280-004-0846-6 [DOI] [PubMed] [Google Scholar]
- 174. Chapkin RS, McMurray DN, Lupton JR. Colon cancer, fatty acids and anti-inflammatory compounds. Curr Opin Gastroenterol. 2007;23:48-54. doi: 10.1097/MOG.0b013e32801145d7 [DOI] [PubMed] [Google Scholar]
- 175. Tuncer S, Banerjee S. Eicosanoid pathway in colorectal cancer: recent updates. World J Gastroenterol. 2015;21:11748. doi: 10.3748/wjg.v21.i41.11748 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176. Calder PC. n−3 polyunsaturated fatty acids, inflammation, and inflammatory diseases. Am J Clin Nutr. 2006;83:1505s-1519S. doi: 10.1093/ajcn/83.6.1505S [DOI] [PubMed] [Google Scholar]
- 177. Zhuang W, Wang G, Li L, Lin G, Deng Z. Omega-3 polyunsaturated fatty acids reduce vascular endothelial growth factor production and suppress endothelial wound repair. J Cardiovasc Transl Res. 2013;6:287-293. doi: 10.1007/s12265-012-9409-0 [DOI] [PubMed] [Google Scholar]
- 178. Gu Z, Shan K, Chen H, Chen YQ. n-3 polyunsaturated fatty acids and their role in cancer chemoprevention. Curr Pharmacol Rep. 2015;1:283-294. doi: 10.1007/s40495-015-0043-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179. Irún P, Lanas A, Piazuelo E. Omega-3 polyunsaturated fatty acids and their bioactive metabolites in gastrointestinal malignancies related to unresolved inflammation. A Review. Front Pharmacol. 2019;10:852. doi: 10.3389/fphar.2019.00852 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180. Vanamala J, Glagolenko A, Yang P, et al. Dietary fish oil and pectin enhance colonocyte apoptosis in part through suppression of ppardelta/PGE2 and elevation of PGE3. Carcinogenesis. 2008;29:790-796. doi: 10.1093/carcin/bgm256 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181. Han YM, Jeong M, Park JM, et al. The ω-3 polyunsaturated fatty acids prevented colitis-associated carcinogenesis through blocking dissociation of β-catenin complex, inhibiting COX-2 through repressing NF-κB, and inducing 15-prostaglandin dehydrogenase. Oncotarget. 2016;7:63583-63595. doi: 10.18632/oncotarget.11544 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182. Fluckiger A, Dumont A, Derangère V, et al. Inhibition of colon cancer growth by docosahexaenoic acid involves autocrine production of tnfα. Oncogene. 2016;35:4611-4622. doi: 10.1038/onc.2015.523 [DOI] [PubMed] [Google Scholar]
- 183. Bentires-Alj M, Barbu V, Fillet M, et al. NF-kappab transcription factor induces drug resistance through MDR1 expression in cancer cells. Oncogene. 2003;22:90-97. doi: 10.1038/sj.onc.1206056 [DOI] [PubMed] [Google Scholar]
- 184. Burke JR, Pattoli MA, Gregor KR, et al. BMS-345541 is a highly selective inhibitor of I kappa B kinase that binds at an allosteric site of the enzyme and blocks NF-kappa B-dependent transcription in mice. J Biol Chem. 2003;278:1450-1456. doi: 10.1074/jbc.M209677200 [DOI] [PubMed] [Google Scholar]
- 185. Ghosh S, Hayden MS. New regulators of NF-kappab in inflammation. Nat Rev Immunol. 2008;8:837-848. doi: 10.1038/nri2423 [DOI] [PubMed] [Google Scholar]
- 186. Lee CH, Jeon YT, Kim SH, Song YS. NF-kappab as a potential molecular target for cancer therapy. Biofactors. 2007;29:19-35. doi: 10.1002/biof.5520290103 [DOI] [PubMed] [Google Scholar]
- 187. Calder PC. Omega-3 polyunsaturated fatty acids and inflammatory processes: nutrition or pharmacology? Br J Clin Pharmacol. 2013;75:645-662. doi: 10.1111/j.1365-2125.2012.04374.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188. Michalak A, Mosińska P, Fichna J. Polyunsaturated fatty acids and their derivatives: therapeutic value for inflammatory, functional gastrointestinal disorders, and colorectal cancer. Front Pharmacol. 2016;7:459. doi: 10.3389/fphar.2016.00459 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189. Bathen TF, Holmgren K, Lundemo AG, et al. Omega-3 fatty acids suppress growth of SW620 human colon cancer xenografts in nude mice. Anticancer Res. 2008;28:3717-3723. [PubMed] [Google Scholar]
- 190. Ichihara H, Zako K, Komizu Y, Goto K, Ueoka R. Therapeutic effects of hybrid liposomes composed of phosphatidylcholine and docosahexaenoic acid on the hepatic metastasis of colon carcinoma along with apoptosis in vivo. Biol Pharm Bull. 2011;34:901-905. doi: 10.1248/bpb.34.901 [DOI] [PubMed] [Google Scholar]
- 191. Kansal S, Negi AK, Bhatnagar A, Agnihotri N. Ras signaling pathway in the chemopreventive action of different ratios of fish oil and corn oil in experimentally induced colon carcinogenesis. Nutr Cancer. 2012;64:559-568. doi: 10.1080/01635581.2012.675619 [DOI] [PubMed] [Google Scholar]
- 192. Wang W, Yang J, Nimiya Y, et al. ω-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 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193. Boudreau MD, Sohn KH, Rhee SH, et al. Suppression of tumor cell growth both in nude mice and in culture by n-3 polyunsaturated fatty acids: mediation through cyclooxygenase-independent pathways. Cancer Res. 2001;61:1386-1391. doi: 10.1158/0008-5472.CAN-19-2362 [DOI] [PubMed] [Google Scholar]
- 194. Zou S, Meng X, Meng Y, et al. Microarray analysis of anti-cancer effects of docosahexaenoic acid on human colon cancer model in nude mice. International J. Clin. 2015;8:5075-5084. [PMC free article] [PubMed] [Google Scholar]
- 195. Jayathilake AG, Kadife E, Kuol N, et al. Krill oil supplementation reduces the growth of CT-26 orthotopic tumours in balb/c mice. BMC Complement Med Ther. 2022;22:34. doi: 10.1186/s12906-022-03521-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196. Jayathilake AG, Hassanzadeganroudsari M, Jovanovska V, et al. The comparative anti-cancer effects of krill oil and oxaliplatin in an orthotopic mouse model of colorectal cancer. Nutr Metab. 2022;19:12. doi: 10.1186/s12986-022-00646-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197. Chapkin RS, Seo J, McMurray DN, Lupton JR/ Mechanisms by which docosahexaenoic acid and related fatty acids reduce colon cancer risk and inflammatory disorders of the intestine. Chem Phys Lipids. 2008;153:14-23. doi: 10.1016/j.chemphyslip.2008.02.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198. Morin C, Rodríguez E, Blier PU, Fortin S. Potential application of eicosapentaenoic acid monoacylglyceride in the management of colorectal cancer. Mar Drugs. 2017;15:283. doi: 10.3390/md15090283 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199. Kansal S, Bhatnagar A, Agnihotri N. Fish oil suppresses cell growth and metastatic potential by regulating PTEN and NF-κB signaling in colorectal cancer. PLoS One. 2014;9:e84627. doi: 10.1371/journal.pone.0084627 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200. Rosa DD, Lourenço FC, da Fonseca AC, et al. Fish oil improves the lipid profile and reduces inflammatory cytokines in Wistar rats with precancerous colon lesions. Nutr Cancer. 2012;64:569-579. doi: 10.1080/01635581.2012.665563 [DOI] [PubMed] [Google Scholar]
- 201. Notarnicola M, Tutino V, Caruso MG, Francavilla A. n-3 polyunsaturated fatty acids reverse the development of polyps in Apc(Min/+) transgenic mice. Oncol Rep. 2016;35:504-510. doi: 10.3892/or.2015.4359 [DOI] [PubMed] [Google Scholar]
- 202. Moreira APB, Sabarense CM, Dias CMGC, et al. 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 [DOI] [PubMed] [Google Scholar]
- 203. Fini L, Piazzi G, Ceccarelli C, 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 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204. Crim KC, Sanders LM, Hong MY, et al. Upregulation of P21WAF1/CIP1 expression in vivo by butyrate administration can be chemoprotective or chemopromotive depending on the lipid component of the diet. Carcinogenesis. 2008;29:1415-1420. doi: 10.1093/carcin/bgn144 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205. Koppelmann T, Pollak Y, Ben-Shahar Y, Gorelik G, Sukhotnik I. The mechanisms of the anti-inflammatory and anti-apoptotic effects of Omega-3 polyunsaturated fatty acids during methotrexate-induced intestinal damage in cell line and in a rat model. Nutrients. 2021;13:888. doi: 10.3390/nu13030888 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206. Calviello G, Serini S, Piccioni E, Pessina G. Antineoplastic effects of n-3 polyunsaturated fatty acids in combination with drugs and radiotherapy: preventive and therapeutic strategies. Nutr Cancer. 2009;61:287-301. doi: 10.1080/01635580802582777 [DOI] [PubMed] [Google Scholar]
- 207. Silva Jde A, Trindade EB, Fabre ME, et al. Fish oil supplement alters markers of inflammatory and nutritional status in colorectal cancer patients. Nutr Cancer. 2012;64:267-273. doi: 10.1080/01635581.2012.643133 [DOI] [PubMed] [Google Scholar]
- 208. Lee JY, Sim TB, Lee JE, Na HK. 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 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209. Volpato M, Hull MA. Omega-3 polyunsaturated fatty acids as adjuvant therapy of colorectal cancer. Cancer Metastasis Rev. 2018;37:545-555. doi: 10.1007/s10555-018-9744-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210. Vasudevan A, Yu Y, Banerjee S, et al. Omega-3 fatty acid is a potential preventive agent for recurrent colon cancer. Cancer Prev Res. 2014;7:1138-1148. doi: 10.1158/1940-6207.CAPR-14-0177 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211. De Carlo F, Witte TR, Hardman WE, Claudio PP. Omega-3 eicosapentaenoic acid decreases CD133 colon cancer stem-like cell marker expression while increasing sensitivity to chemotherapy. PLoS One. 2013;8:e69760. doi: 10.1371/journal.pone.0069760 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212. Rani I, Sharma B, Kumar S, Kaur S, Agnihotri N. Apoptosis mediated chemosensitization of tumor cells to 5-fluorouracil on supplementation of fish oil in experimental colon carcinoma. Tumor Biol. 2017;39:1010428317695019. doi: 10.1177/1010428317695019 [DOI] [PubMed] [Google Scholar]
- 213. Rani I, Vaiphei K, Agnihotri N. Supplementation of fish oil augments efficacy and attenuates toxicity of 5-fluorouracil in 1,2-dimethylhydrazine dihydrochloride/dextran sulfate sodium-induced colon carcinogenesis. Cancer Chemother Pharmacol. 2014;74:309-322. doi: 10.1007/s00280-014-2497-6 [DOI] [PubMed] [Google Scholar]
- 214. Xue H, Le Roy S, Sawyer MB, et al. Single and combined supplementation of glutamine and n-3 polyunsaturated fatty acids on host tolerance and tumour response to 7-ethyl-10-[4-(1-piperidino)-1-piperidino]carbonyloxy-camptothecin (CPT-11)/5-fluorouracil chemotherapy in rats bearing ward colon tumour. Br J Nutr. 2009;102:434-442. doi: 10.1017/S0007114508199482 [DOI] [PubMed] [Google Scholar]
- 215. Jeong S, Kim DY, Kang SH, et al. Docosahexaenoic acid enhances oxaliplatin-induced autophagic cell death via the ER stress/sesn2 pathway in colorectal cancer. Cancers. 2019;11:982. doi: 10.3390/cancers11070982 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216. Mocellin MC, Pastore e Silva Jde A, Camargo Cde Q, et al. Fish oil decreases C-reactive protein/albumin ratio improving nutritional prognosis and plasma fatty acid profile in colorectal cancer patients. Lipids. 2013;48:879-888. doi: 10.1007/s11745-013-3816-0 [DOI] [PubMed] [Google Scholar]
- 217. Bonatto SJ, Oliveira HH, Nunes EA, et al. Fish oil supplementation improves neutrophil function during cancer chemotherapy. Lipids. 2012;47:383-389. doi: 10.1007/s11745-011-3643-0 [DOI] [PubMed] [Google Scholar]
- 218. Read JA, Beale PJ, Volker DH, et al. Nutrition intervention using an eicosapentaenoic acid (EPA)-containing supplement in patients with advanced colorectal cancer. Effects on nutritional and inflammatory status: a phase II trial. Support Care Cancer. 2007;15:301-307. doi: 10.1007/s00520-006-0153-3 [DOI] [PubMed] [Google Scholar]
- 219. Song M, Ou F-S, Zemla TJ, 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 [DOI] [PMC free article] [PubMed] [Google Scholar]


