Literature
首页医源资料库在线期刊美国临床营养学杂志2004年79卷第6期

Dietary long-chain n–3 fatty acids for the prevention of cancer: a review of potential mechanisms

来源:《美国临床营养学杂志》
摘要:3fattyacids,especiallythelong-chainpolyunsaturatedfattyacidseicosapentaenoicacidanddocosahexaenoicacid,presentinfattyfishandfishoilsinhibitcarcinogenesis。3fattyacidintake,andcancerriskare,however,somewhatlessconsistent。3fattyacids。3fattyacidintakeandcanc......

点击显示 收起

Susanna C Larsson, Maria Kumlin, Magnus Ingelman-Sundberg and Alicja Wolk

1 From the Divisions of Nutritional Epidemiology (SCL and AW), Experimental Asthma and Allergy Research (MK), and Molecular Toxicology (MI-S), The National Institute of Environmental Medicine, Karolinska Institutet, Stockholm.

2 Supported by grants from the Swedish Cancer Society (to AW).

3 Reprints not available. Address correspondence to SC Larsson, Division of Nutritional Epidemiology, The National Institute of Environmental Medicine, Karolinska Institutet, Box 210, SE-171 77 Stockholm, Sweden. E-mail: susanna.larsson{at}imm.ki.se.


ABSTRACT  
Increasing evidence from animal and in vitro studies indicates that n–3 fatty acids, especially the long-chain polyunsaturated fatty acids eicosapentaenoic acid and docosahexaenoic acid, present in fatty fish and fish oils inhibit carcinogenesis. The epidemiologic data on the association between fish consumption, as a surrogate marker for n–3 fatty acid intake, and cancer risk are, however, somewhat less consistent. This review highlights current knowledge of the potential mechanisms of the anticarcinogenic actions of n–3 fatty acids. Moreover, a possible explanation of why some epidemiologic studies failed to find an association between n–3 fatty acid intake and cancer risk is provided. Several molecular mechanisms whereby n–3 fatty acids may modify the carcinogenic process have been proposed. These include suppression of arachidonic acid-derived eicosanoid biosynthesis; influences on transcription factor activity, gene expression, and signal transduction pathways; alteration of estrogen metabolism; increased or decreased production of free radicals and reactive oxygen species; and mechanisms involving insulin sensitivity and membrane fluidity. Further studies are needed to evaluate and verify these mechanisms in humans to gain more understanding of the effects of n–3 fatty acid intake on cancer risk.

Key Words: n–3 Fatty acids • eicosapentaenoic acid • docosahexaenoic acid • -linolenic acid • arachidonic acid • carcinogenesis • eicosanoids • gene expression • epidemiology


INTRODUCTION  
We recently reviewed epidemiologic studies on the relation between intakes of fish and marine fatty acids and the risks of breast and prostate cancers and of other hormone-related cancers (1). In brief, ecologic studies have shown that high per capita fish consumption is correlated with a lower incidence of cancer in the population (2–5). Additionally, the decreased consumption of fish and increased intake of vegetable oils rich in n–6 fatty acids among Japanese women during the past decades have been accompanied by increased breast cancer rates (6). Nevertheless, analytic epidemiologic studies having a case-control or cohort design have not yielded clear conclusions concerning the protective effect of fish consumption or n–3 fatty acid intake against cancer; although some studies showed an inverse association between the intake of n–3 fatty acids (7, 8) or fish (9–16) and cancer risk, most did not (17–25).

The role that the long-chain, marine n–3 polyunsaturated fatty acids (PUFAs) eicosapentaenoic acid (EPA, 20:5n–3) and docosahexaenoic acid (DHA, 22:6n–3), which are present in fatty cold-water fish and fish oils, play in the etiology of cancer has been highlighted by animal experiments and in vitro studies showing that these PUFAs suppress the development of major cancers (26–31). These experimental findings are supported by results from clinical studies showing a reduction in intestinal hyperproliferation after consumption of fish oil-derived n–3 PUFAs in subjects at elevated risk of colon cancer due to sporadic colonic adenomas (32, 33). Although a few previous reviews have described some selected actions through which long-chain n–3 fatty acids may play a role in carcinogenesis, such as biosynthesis of eicosanoids (34, 35), lipid peroxidation (36–38), and some signal transduction pathways (34, 36), to our knowledge, no comprehensive review that puts all these pieces and further evidence together is available.

The present review focuses on several putative mechanisms whereby long-chain n–3 fatty acids may modulate the carcinogenic process. Furthermore, a potential explanation of why several case-control studies and large cohort studies failed to confirm a protective effect of long-chain n–3 fatty acids against cancer development is briefly discussed. Moreover, we discuss how knowledge of the mechanisms of action of PUFAs should be taken into account in epidemiologic analyses.


MECHANISMS OF POTENTIAL CHEMOPREVENTIVE EFFECTS OF n–3 FATTY ACIDS ON CARCINOGENESIS  
Mounting evidence shows that dietary n–3 PUFAs inhibit the promotion and progression stages of carcinogenesis. Several molecular mechanisms whereby n–3 PUFAs potentially affect carcinogenesis have been proposed. These mechanisms include 1) suppression of arachidonic acid (AA, 20:4n–6)-derived eicosanoid biosynthesis, which results in altered immune response to cancer cells and modulation of inflammation, cell proliferation, apoptosis, metastasis, and angiogenesis; 2) influences on transcription factor activity, gene expression, and signal transduction, which leads to changes in metabolism, cell growth, and differentiation; 3) alteration of estrogen metabolism, which leads to reduced estrogen-stimulated cell growth; 4) increased or decreased production of free radicals and reactive oxygen species; and 5) mechanisms involving insulin sensitivity and membrane fluidity.

Inhibition of arachidonic acid-derived eicosanoid biosynthesis
One of the more important functions of PUFAs (n–3 and n–6 fatty acids) is related to their enzymatic conversion into eicosanoids (Figure 1), which are short-lived, hormone-like lipids with chain lengths of 20 carbon atoms (eicosa = 20). Eicosanoids are biologically potent and have a wide array of activities: they modulate inflammatory and immune responses and play a critical role in platelet aggregation, cellular growth, and cell differentiation. The precursor fatty acids for the formation of eicosanoids are dihomo--linolenic acid (DGLA, 20:3n–6), AA, and EPA. Linoleic acid (LA, 18:2n–6) and -linolenic acid (-LNA, 18:3n–3) are the predominant plant-derived dietary PUFAs and are the precursors of DGLA and AA and of EPA, respectively. The production of eicosanoids begins with the liberation of PUFAs from membrane phospholipids by the action of various phospholipases. Thereafter, these PUFAs serve as substrates for cyclooxygenases (COX-1, which is a constitutive enzyme, and COX-2, which is an inducible enzyme), lipoxygenases (5-, 12-, and 15-lipoxygenase), or cytochrome P450 monooxygenases. The cyclooxygenases give rise to prostaglandins and thromboxanes, whereas the lipoxygenases produce leukotrienes, hydroxy fatty acids, and lipoxins. Cytochrome P450 monooxygenase-mediated oxidation of PUFAs generates hydroxyfatty acids, dihydroxyfatty acids, and epoxy fatty acids. The relative proportions of PUFAs in cell membranes, as well as cell type, are the primary factors in regulating which eicosanoid will be generated. Hydrolytic release of PUFAs from phospholipids appears to occur indiscriminately with n–3 and n–6 PUFAs. Because the major PUFA in cell membranes is AA, most eicosanoids produced will be of the 2-series prostanoids (prostaglandins and thromboxanes) and the 4-series leukotrienes, with 2 and 4 double bonds, respectively, in the products. EPA is a substrate for 3-series prostanoids and 5-series leukotrienes. In general, AA-derived eicosanoids have proinflammatory effects (39–41)—although prostaglandin E2 (PGE2) has been suggested to also have antiinflammatory properties (42)—whereas EPA-derived eicosanoids have antiinflammatory effects. Eicosanoids generated from AA, such as PGE2, leukotriene B4, thromboxane A2, and 12-hydroxyeicosatetraenoic acid, have been positively linked to carcinogenesis (34). For example, PGE2 promotes tumor cell survival and is found at higher concentrations in cancer cells than in normal cells (43). The mechanisms whereby PGE2 promotes tumor survival include inhibition of apoptosis and stimulation of cell proliferation (44–46). It has also been reported that PGE2 increases tumor progression by promoting tumor angiogenesis (47–49). 12-Hydroxyeicosatetraenoic acid has been shown to suppress apoptosis (50, 51) and promote tumor angiogenesis (52) and tumor cell adhesion to endothelial cells (53, 54); the latter is an essential and early event in the initiation of the metastatic cascade. Some lipoxygenase products generated from AA, such as leukotriene B4 and 5-hydroxyeicosatetraenoic acid, also play a role in tumor cell adhesion (55) and thus may augment metastatic potential. Leukotriene B4 further enhances generation of reactive oxygen species (40), which may attack DNA and lead to cancer initiation. AA-derived eicosanoids synthesized by the action of cytochrome P450 monooxygenase were recently shown to influence several biological processes, including cell proliferation, apoptosis, and inflammation (56). For example, 14,15-epoxyeicosatrienoic acid inhibits apoptosis (57) and increases cell proliferation (58). Although several AA-derived eicosanoids have been suggested to promote carcinogenesis, some of them, such as PGI2 (59), 15d-PGJ2 (metabolite of PGD2) (60), and 15(S)-hydroxyeicosatetraenoic acid (61), as well as the LA-derived 13(S)-hydroxyoctadecadienoic acid (62, 63), have been found to suppress cell proliferation and induce apoptosis.


View larger version (18K):
FIGURE 1.. Overview of the metabolism of n–6 and n–3 polyunsaturated fatty acids (PUFAs) into eicosanoids involved in inflammation and carcinogenesis. The names of these eicosanoids are shown in bold. LA, linoleic acid (18:2n–6); -LNA, -linolenic acid (18:3n–3); GLA, -linolenic acid (18:3n–6); DGLA, dihomo--linolenic acid (20:3n–6); AA, arachidonic acid (20:4n–6); EPA, eicosapentaenoic acid (20:5n–3); DHA, docosahexaenoic acid (22:6n–3); PLA2, phospholipase A2; LOX, lipoxygenase; COXs, cyclooxygenases (COX-1 and COX-2); 15-HETE, 15(S)-hydroxyeicosatetraenoic acid; 12-HETE, 12-hydroxyeicosatetraenoic acid; 5-HETE, 5-hydroxyeicosatetraenoic acid; HEPE, hydroxyeicosapentaenoic acid; HPETE, hydroperoxyeicosatetraenoic acid; HPEPE, hydroperoxyeicosapentaenoic acid; LT, leukotriene; HODE, hydroxyoctadecadienoic acid; PG, prostaglandin; TX, thromboxane.

 
The most salient mechanism by which n–3 fatty acids may lower the risk of cancer is through their suppressing effect on the biosynthesis of AA-derived eicosanoids. This effect is achieved at several levels. First, high intakes of n–3 fatty acids result in their incorporation into membrane phospholipids, where they partially replace AA (64). By decreasing the availability of AA precursors, this substitution suppresses the biosynthesis of AA-derived eicosanoids in favor of EPA-derived 3-series prostanoids and 5-series leukotrienes. Second, n–3 PUFAs compete with n–6 PUFAs for desaturases and elongases, and n–3 PUFAs have greater affinities for the enzymes than do n–6 PUFAs. Thus, a higher intake of n–3 PUFAs reduces the desaturation and elongation of LA to AA (34) and thus the production of AA-derived eicosanoids. Third, n–3 fatty acids suppress COX-2 (65–67) and compete with n–6 fatty acids for cyclooxygenases to form eicosanoids (68–70). Compared with AA, EPA is the preferential substrate for lipoxygenase; hence an increased EPA intake leads to higher formation of EPA-derived lipoxygenase products at the expense of AA-derived lipoxygenase products when both fatty acids are simultaneously available (71). Dietary n–6 PUFAs, in contrast with n–3 PUFAs, have been reported to up-regulate the expression of rat COX-2 and, to some extent, COX-1 (72) and thus increase the production of prostanoids. Finally, n–3 PUFAs enhance eicosanoid catabolism, which is postulated to be mediated through induction of peroxisomal enzymes (73). The formation of AA-derived eicosanoids is decreased not only by n–3 PUFAs but also by eicosanoids derived from them, and some of these eicosanoids (eg, 15-hydroperoxyeicosapentaenoic acid) have an even more inhibitory effect than does EPA (74). Taken together, these effects at different levels dramatically reduce the AA-derived eicosanoids that are linked to inflammation and carcinogenesis.

Note that the potency of dietary EPA and DHA is estimated to be approximately five-fold that of -LNA for the suppression of AA-derived eicosanoids (35). Similarly, the activities of 5- and 6-desaturase are considerably lower in rats fed a fish-oil (rich in EPA and DHA) diet than in those fed a flaxseed-oil (rich in -LNA) diet (75, 76).

Influence on transcription factor activity, gene expression, and signal transduction
Dietary PUFAs and their metabolites may exert some of their antitumor effects by affecting gene expression or the activities of signal transduction molecules involved in the control of cell growth, differentiation apoptosis, angiogenesis, and metastasis.

Peroxisome proliferator-activated receptor
The first transcription factor that was identified as being regulated by fatty acids was the peroxisome proliferator-activated receptor- (PPAR) (77), a member of the PPAR family, which also comprises PPAR (also referred to as PPARß) and PPAR (3 isoforms: 1, 2, and 3). These ligand-activated transcription factors were first found to be implicated in the regulation of lipid metabolism and homeostasis but have recently appeared to be involved in cell proliferation, cell differentiation, and inflammatory responses (78, 79). The preferred natural ligands of PPAR are PUFAs, including LA, -LNA, AA, and EPA (80, 81). Endogenous ligands include 15d-PGJ2, 9(S)-hydroxyoctadecadienoic acid, 13(S)-hydroxyoctadecadienoic acid, and 15-hydroxyeicosatetraenoic acid (80, 82, 83). In addition to being a PPAR agonist, EPA, but not other fatty acids (-LNA, DHA, and n–6 PUFAs), has been shown to significantly increase PPAR1 messenger RNA concentrations in isolated adipocytes (84). PPAR can be activated by fibrates (hypolipidemic drugs) (79) and by various saturated and unsaturated fatty acids, including palmitic acid, oleic acid, LA, AA (85), conjugated LA (86), and EPA (77). Known activators of PPAR are DGLA, EPA, AA, palmitic acid, and the prostaglandins PGA1 (derived from DGLA) and PGD3 (80). PPAR is expressed in several epithelial tissues that are important in human cancers (83). Agonists of PPAR have been found to have antiproliferative effects both in vitro (87–92) and in vivo (93, 94). For instance, in a phase II clinical study in patients with advanced prostate cancer, the PPAR agonist troglitazone blocked or reversed tumor progression, which led to a prolonged stabilization of or decrease in prostate-specific antigen in 50% of the patients (93, 95). Furthermore, reduced concentrations of 15-hydroxyeicosatetraenoic acid, an endogenous ligand for PPAR in the prostate, contribute to increased proliferation of and reduced differentiation in prostate carcinoma (96). DHA was found to induce apoptosis in vascular smooth muscle cells by activation of PPAR, p38 mitogen-activated protein kinases, bax, and cytochrome c (94). Murata et al (97) reported that EPA decreases the activity of mitogen-activated protein kinase and inhibits cell proliferation in HepG2 cells. Both PPAR and PPAR have antiinflammatory properties and may thereby contribute to suppression of carcinogenesis (79). PPAR has been suggested to act as an inducer of cell proliferation and as a promoter of the progression of certain types of cancer (80). PPAR antagonists may have a role in decreasing colon cancer risk (80) although this has not been conclusively shown.

Nuclear transcription factor B
The nuclear transcription factor B (NF-B) family of transcription factors is involved in cytokine gene expression, cellular adhesion, cell cycle activation, apoptosis, and carcinogenesis (98). Constitutive NF-B activation in cancer appears to play a role in tumor growth (98). In an experimental study, n–3 fatty acids significantly decreased NF-B activation in murine macrophages (99). Furthermore, cells treated with n–3 fatty acids showed a significant decrease in both messenger RNA and protein expression of tumor necrosis factor (decreases of 47% and 46%, respectively) (99).

ras and protein kinase C
Collett et al (100) showed that, compared with LA, DHA lowers the activation of ras oncogenes, which are frequently activated in tumors, in mouse colon cells. ras activation by point mutation or overexpression is associated with elevated concentrations of cellular diacylglycerol and thus down-regulation of protein kinase C (PKC) (101). Feeding rats dietary fish oil has been shown to block the azoxymethane (a carcinogen)-induced decrease in steady-state concentrations of PKC and - isozymes, both of which have tumor suppressor functions (102). Unlike PKC and -, PKC ß2, which is induced early during colon carcinogenesis (103), promotes colon cancer (104, 105). Murray et al (103) showed a significant decrease in the concentration of membrane-associated PKC ß2 in the colonic epithelium of rats fed fish oil. Furthermore, the fish-oil diet blocks PKC ß2-mediated hyperproliferation and enhances carcinogenesis in intestinal epithelial cells (103).

Ornithine decarboxylase
Ornithine decarboxylase (ODC), the rate-limiting enzyme in polyamine biosynthesis, is intimately involved in normal cellular proliferation. Both ODC activity and polyamine content are significantly higher in most colorectal neoplasms than in normal, adjacent, healthy control tissues (106, 107). Rao and Reddy (108) investigated the modulating effect of high-fat diets rich in n–3, n–6, and n–9 fatty acids on ODC activity in the liver, colon, and small intestinal mucosa. The authors showed that high amounts of corn oil (rich in n–6 fatty acids) in the diet increase the activities of ODC and tyrosine-specific protein kinase in the colon and liver of male F344 rats, whereas high dietary amounts of fish oil and olive oil (rich in n–9 fatty acids) suppress these activities (108). These results were supported by those of Bartram et al (109), who showed that, compared with corn oil, dietary fish oil suppresses ODC activity in healthy humans.

3-Hydroxy-3-methylglutaryl coenzyme-A reductase
Several studies in rats showed that the long-chain n–3 fatty acids reduce the activity and concentration of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase (110–113), which catalyzes the biosynthesis of mevalonate. In addition to being essential for the biosynthesis of cholesterol and coenzyme Q, mevalonate is required for DNA synthesis and cell proliferation (114). HMG-CoA reductase inhibitors (statins) have been shown to have antiangiogenic properties (115), which suggests that HMG-CoA is involved in angiogenesis. However, unlike statins, long-chain n–3 fatty acids have generally not been shown to decrease cholesterol concentrations in humans (116). Thus, a potential effect of long-chain fatty acids on HMG-CoA reductase activity in humans remains speculative.

Cyclooxygenase-2 and lipoxygenases
Several studies indicate that although n–6 PUFAs promote colon and mammary carcinogenesis by up-regulating expression of p21ras and COX-2, n–3 PUFAs may exert one of their antitumor effects by suppressing the expression of p21ras and COX-2 (65, 117, 118). COX-2 expression has been shown to down-regulate the apoptotic pathway (34). Overexpression of COX-2 has been detected in many types of cancer, including cancer of the breast, colon, and prostate (119, 120). Numerous epidemiologic studies found that long-term use of COX-2 inhibitors (nonsteroidal antiinflammatory drugs) is associated with a lower risk of colorectal cancer, adenomatous polyps, and perhaps other types of cancer (121). COX-2 catalyzes the conversion of procarcinogens to carcinogens, and significant amounts of xenobiotics could be oxidized to mutagens by COX-2. Moreover, metabolic turnover of AA is sufficient to produce mutagens. For example, malondialdehyde, a byproduct of the oxidation of AA, is highly reactive and forms adducts with DNA (122).

Nitric oxide
Nitric oxide (NO) and reactive products derived from it, such as reactive nitrogen species, are mutagenic and have the potential to produce nitration, nitrosation, and deamination reactions on DNA bases (123, 124). Excessive production of NO during chronic inflammation is believed to cause DNA damage and impaired DNA repair (eg, mutation of the p53 tumor suppressor gene) and, in the long term, cancer (124–126). Tumor-derived NO promotes tumor growth and metastasis by enhancing the invasive, angiogenic, and migratory abilities of tumor cells (124, 126, 127), which may also be triggered by activation of COX-2 (124). Another mechanism whereby NO may stimulate tumor growth is by increasing the production of PGE2 (128), which is implicated in tumor progression. NO production in a macrophage cell line was found to be suppressed by the n–3 PUFAs -LNA, EPA, and DHA in a dose-dependent fashion (129). Several other studies provide additional evidence for a suppressing effect of DHA on NO production (130–132).

Alteration of estrogen metabolism
It is well known that estrogen has proliferative effects on estrogen-sensitive tissues and that high estrogen concentrations may increase the risk of breast cancer and of some other hormone-dependent cancers. The AA-derived eicosanoid PGE2 has been shown to stimulate the activity of aromatase P450, which converts 19-carbon steroids to estrogens (133). In contrast, PGE3, a product of EPA metabolism, does not activate aromatase P450. Hence, an increased intake of EPA, which leads to increased production of PGE3 and decreased production of PGE2, is expected to decrease estrogen production and thus reduce estrogen-stimulated cell growth. Although a high intake of n–3 PUFAs relative to that of n–6 PUFAs may decrease endogenous estrogen production, no studies have yet directly examined this issue in humans.

Increased or decreased production of free radicals and reactive oxygen species
Free radicals and reactive oxygen species produced in cells may attack PUFAs to form lipid hydroperoxides, which decompose in chain reactions to form more free radicals and reactive aldehydes such as trans-4-hydroxy-2-nonenal and malondialdehyde. These metabolites potentially generate promutagenic exocyclic DNA adducts in human cells, which lead to cancer (134, 135). Generally, the autooxidizability of various fatty acids in an air atmosphere is roughly proportional to the number of double bounds in the molecule. The long-chain, highly unsaturated n–3 fatty acids are therefore believed to promote lipid peroxidation and thus carcinogenesis. These assumptions are based on the results of investigations of the in vitro oxidation of unsaturated fatty acids in homogeneous systems (136). However, there is evidence that, compared with the intake of n–6 fatty acids, the intake of n–3 fatty acids suppresses so-called free radical diseases, such as cancer, ageing, and atherosclerosis, which suggests that lipid peroxidation in vivo may not correspond with that in vitro (35). For instance, several studies found that increasing the dietary intakes of EPA and DHA does not increase the oxidative susceptibility of LDL cholesterol (137–139). Moreover, Takahashi et al (140) reported that genes coding for some antioxidant enzymes (eg, glutathione transferases and manganese-superoxide dismutase) were up-regulated in mice fed a fish-oil diet, which suggests a protective effect against the production of reactive oxygen species and thus against cancer initiation. Studies in healthy humans also showed that consumption of a diet providing >2.3 g EPA plus DHA/d decreases superoxide production (40). Inflammation has been hypothesized to increase the production of free radicals and reactive oxygen species, which leads to carcinogenesis. Although n–6 fatty acids augment these events through the overproduction of AA-derived proinflammatory eicosanoids, the n–3 fatty acids suppress inflammation and thus the overproduction of free radicals and carcinogenesis (Figure 2).


View larger version (33K):
FIGURE 2.. Hypothetical scheme showing potential mechanisms whereby n–6 polyunsaturated fatty acids (PUFAs) and n–3 PUFAs may promote and suppress carcinogenesis, respectively. In initiated tumor cells, phospolipase A2, cyclooxygenase 2, and lipoxygenases are often overexpressed, which leads to overproduction of arachidonic acid (AA, 20:4n–6)-derived eicosanoids that augment inflammation. Nitric oxide, which is elevated in inflammation, is implicated in both the initiation and the progression stages of carcinogenesis. Nitric oxide may stimulate tumor growth and metastasis by enhancing the angiogenic and migratory abilities of tumor cells. Dietary n–3 PUFAs reduce the desaturation and elongation of linoleic acid (18:2n–6) to AA, the incorporation of AA into membranes, and the biosynthesis of AA-derived eicosanoids; suppress inflammation; stimulate apoptosis; up-regulate the expression of genes coding for antioxidant enzymes; and thus inhibit tumor growth and metastasis. + and solid arrows, stimulation; – and dashed arrows, suppression; , increase. EPA, eicosapentaenoic acid (20:5n–3); DHA, docosahexaenoic acid (22:6n–3); ROS, reactive oxygen species; RNS, reactive nitrogen species.

 
In this context, note that some researchers found that the inhibitory effects of fish oil on the growth of tumors in vitro are abolished by the concurrent addition of vitamin E (37, 141–143) or vitamin C (143), which suggests that oxidized products of n–3 PUFAs suppress cell growth. Generation of oxygen radicals appears to be involved in the initiation of apoptosis and in the natural defense against transformed or foreign cells (144). Thus, the inhibitory effects of long-chain n–3 PUFAs on cell growth may, at least partly, be explained by their formation of oxidation products, which leads to apoptosis and cell growth arrest. To date, however, there is little direct evidence that n–3 PUFAs influence the carcinogenic process by alteration of free radical production in humans. Further studies on the role of lipid hydroperoxides in the modulation of tumor growth in vivo are needed to elucidate their role in carcinogenesis.

Other potential mechanisms
In addition to the potential mechanisms described above, dietary n–3 PUFAs may also modulate carcinogenesis through effects on insulin sensitivity and cell membrane fluidity, although these mechanisms have been less well studied. The n–3 fatty acid EPA has been found to improve insulin sensitivity in rats (145–147) and patients with type 2 diabetes (148). The effect has been proposed as being mediated through PPAR and PPAR (149) but may also involve modification of the phospholipid components of skeletal muscle membranes (145, 150). Iigo et al (151) showed that treatment of colon carcinoma cells with DHA resulted in altered tumor cell membrane characteristics and a decreased ability to metastasize.


DISCUSSION  
Substantial evidence from experimental and animal studies indicates that long-chain n–3 fatty acids in fish and fish oils inhibit carcinogenesis. Epidemiologic studies examining the associations of fish and marine n–3 fatty acids with the risk of development of cancer have, however, been inconclusive (1). About one-third to one-half of the studies that examined the relation between the intake of long-chain n–3 fatty acids or fish and cancers of the breast, prostate, endometrium, or ovary reported a statistically significant reduction in the risks of these cancers. The remaining studies either found an inverse association that was not statistically significant or failed to find any association.

There are several possible explanations for these null findings. First, in comparison with the consistent protective effect of long-chain n–3 fatty acids in animal and in vitro studies, the inconsistent associations observed in analytic epidemiologic studies may partly be due to the fact that the intake of long-chain n–3 fatty acids in some of the studied populations was too low to produce a protective effect. Other possible explanations include low within-population variability in the intake of fish or n–3 fatty acids (which limits the statistical power to detect an association) and nondifferential misclassification of estimated n–3 fatty acid intake. Although most of the potential mechanisms by which long-chain n–3 fatty acids may inhibit carcinogenesis are at the promotion and progression stages, the critical period for dietary n–3 fatty acid exposure may be during childhood or early adulthood. Thus, if exposure information is obtained at middle or old age when cancer is diagnosed, the association between n–3 fatty acid intake and cancer might be missed.

Another aspect that has to be taken into account when evaluating results from epidemiologic studies is that most studies examined the association between cancer risk and total fish intake rather than fatty fish intake, which may better mirror total intake of marine n–3 fatty acids. Total fat content in fish varies widely between species, from 0.6–0.7 g/100 g in halibut and cod to 16.0–18.5 g/100 g in mackerel and herring from the Pacific (Table 1). The composition of the fat depends on the geographic area in which the fish live, the fish's diet, and seasonal variations (34) and on environmental factors, such as temperature, salinity, and the depth at which the fish live, with the highest content of EPA and DHA in cold-water fish (152). In the future, the farming industry may also have important influences on the fat composition of the fish. The n–3 fatty acid -LNA, which is found in dark green leafy vegetables, rapeseed oil (canola oil), flaxseed, some nuts (especially walnuts), and soybeans, may also bias results if only fish consumption is taken into account. Nevertheless, although humans can convert -LNA to EPA, which can be further elongated and desaturated to DHA, this conversion is not very efficient. The extent of the conversion of -LNA to EPA has not been fully characterized and may depend on intakes of total fat, -LNA, EPA, DHA, and LA (153–157). It has been reported that when the intake of LA is held constant at 15 g/d, the total percentage of conversion of -LNA to EPA and DHA is 11–18.5%, but when the intake of LA is increased from 15 to 30 g/d, this conversion is reduced to 5–11% (155). A recent study showed that 2.8% of the dietary -LNA consumed was converted to EPA and that this conversion was down-regulated (2-fold) in subjects who consumed a diet high in EPA and DHA (154). Pawlosky et al (157) reported an even more limited conversion of -LNA to EPA in humans: only 0.2% of plasma -LNA was converted to EPA. Low-fat diets result in increased 5- and 6-desaturation (156), which may increase the conversion of -LNA to EPA.


View this table:
TABLE 1. Amounts of total fat (fatty acids), -linolenic acid (-LNA), eicosapentaenoic acid (EPA), docosahexaenoic acid (DHA), linoleic acid (LA), and arachidonic acid (AA) and ratios of n–3 to n–6 fatty acids in selected species of fish and in meat1

 
Another drawback is that most epidemiologic studies largely analyzed the intake of n–3 PUFAs without taking into account the intake of n–6 PUFAs. Given the above-described mechanisms through which EPA and DHA may decrease the risk of cancer development, the ratio of n–3 to n–6 PUFAs seems to be more important than is the absolute intake of n–3 PUFAs. Indeed, ratios of n–3 to n–6 PUFAs, but not absolute concentrations of these fatty acids, in adipose tissue biopsy specimens were found to be inversely associated with breast cancer risk in a multinational epidemiologic study (158). Experimental data indicate that a ratio of n–3 to n–6 PUFAs of 1:1 or 1:2 is needed for protection against the development of cancer (159). In most Western countries, the ratio is 1:10–1:20 (159); hence, no effect on carcinogenesis would be expected. Although dietary LA intake of up to 2–3% of energy intake increases tissue AA concentrations, LA intake of >3% of energy intake is poorly correlated with tissue AA concentrations (160, 161). Because the average LA intake in the United States and Western Europe is 6–7% of energy intake (162), a moderate change in dietary LA intake would not be expected to modulate tissue AA concentrations. However, LA intakes of >12% of energy intake may actually decrease tissue AA concentrations because of inhibition of 6-desaturase activity (156). On the other hand, dietary preformed AA, which is found in meat and fish (Table 1), is much more effective in enriching tissue phospholipid membranes than is LA (163). Thus, a low LA intake and a high n–3 fatty acid intake seems to be needed to suppress AA-derived eicosanoids, and such a diet is not very common in Western societies. Because tissue concentrations of AA, in contrast with those of LA, are strongly influenced by dietary intake, epidemiologic studies of the relation between the ratio of AA to n–3 PUFAs and cancer risk may be warranted.

The absence of an association between dietary long-chain fatty acids and cancer risk in some epidemiologic studies may not exclude the possibility of different effects in subgroups. The potential protective effect of dietary long-chain n–3 fatty acids may be modified by intakes of antioxidants, such as vitamins E and C; such modification has been observed in experimental studies but has not been taken into account in epidemiologic analyses to date.

An important issue of concern is that the fish oils and marine n–3 fatty acids used in experimental settings may differ from those normally consumed by humans in their content of contaminated substances. Thus, a possible beneficial effect of marine n–3 fatty acids may be offset by potential carcinogenic substances, such as some pesticides and heavy metals (eg, mercury), that accumulate in fatty fish. Furthermore, heterocyclic amines formed during the cooking of fish at high temperatures (164) have been shown to produce cancer in various organs in animals (165).

Another possible explanation for the discrepancy between animal and epidemiologic studies involves differences in doses and the stage of tumor development. In animal studies, large doses of n–3 PUFAs were usually used, and tumors were artificially induced. In addition, most of these studies did not address the initiation phase of carcinogenesis. Hence, high doses of n–3 PUFAs applied during the promotion and progression stages of tumor development may indeed inhibit carcinogenesis in animal models, whereas long-term exposures to relatively low doses of long-chain n–3 PUFAs may not be as effective against cancer development in humans. Alternatively, the inconsistencies in results between animal and epidemiologic studies may be due to publication bias. Small animal studies, which take relatively little effort and money, are more likely to suffer from publication bias than are large, well-designed epidemiologic studies. Consequently, the overall picture may be biased toward protective effects in animal studies.

In the light of the above-mentioned methodologic difficulties and limitations of observational epidemiologic studies, it is not surprising that the results reported from these studies to date on the association between long-chain n–3 fatty acids and cancer risk are inconsistent. Future epidemiologic studies have to take into account more aspects, as mentioned above, in the collection and analysis of data. In epidemiologic analyses, the biological interplay—observed in experimental studies—between n–3 and n–6 fatty acids and other factors (eg, vitamin E and antiinflammatory drugs) should be taken into account in appropriate statistical analyses to address these issues.

In summary, several mechanisms whereby n–3 fatty acids may modify the carcinogenic process were described. These fatty acids can suppress AA-derived eicosanoid biosynthesis; influence transcription factor activity, gene expression, and signal transduction pathways; modulate estrogen metabolism; increase or decrease the production of free radicals and reactive oxygen species; and influence insulin sensitivity and membrane fluidity. On the basis of these multiple mechanisms, n–3 PUFAs may have an important influence on carcinogenesis. Further studies are needed to identify new mechanisms and to evaluate and verify these mechanisms in humans to gain more understanding of the effects of marine n–3 fatty acid intake on cancer risk in real-life situations. Epidemiologic studies with more detailed information about n–3 and n–6 fatty acid exposures and improved analytic approaches that take into account the biological interplay between several nutritional factors in cancer development are needed.


ACKNOWLEDGMENTS  
The manuscript was drafted by SCL. MK and MI-S critically reviewed the manuscript. AW made intellectual contributions throughout the whole process of manuscript preparation. All the authors reviewed the final version. None of the authors had any conflicts of interest.


REFERENCES  

  1. Terry PD, Rohan TE, Wolk A. Intakes of fish and marine fatty acids and the risks of cancers of the breast and prostate and of other hormone-related cancers: a review of the epidemiologic evidence. Am J Clin Nutr 2003;77:532–43.
  2. Kaizer L, Boyd NF, Kriukov V, Tritchler D. Fish consumption and breast cancer risk: an ecological study. Nutr Cancer 1989;12:61–8.
  3. Hursting SD, Thornquist M, Henderson MM. Types of dietary fat and the incidence of cancer at five sites. Prev Med 1990;19:242–53.
  4. Caygill CP, Charlett A, Hill MJ. Fat, fish, fish oil and cancer. Br J Cancer 1996;74:159–64.
  5. Sasaki S, Horacsek M, Kesteloot H. An ecological study of the relationship between dietary fat intake and breast cancer mortality. Prev Med 1993;22:187–202.
  6. Lands WE, Hamazaki T, Yamazaki K, et al. Changing dietary patterns. Am J Clin Nutr 1990;51:991–3.
  7. Maillard V, Bougnoux P, Ferrari P, et al. n–3 And n–6 fatty acids in breast adipose tissue and relative risk of breast cancer in a case-control study in Tours, France. Int J Cancer 2002;98:78–83.
  8. Norrish AE, Skeaff CM, Arribas GL, Sharpe SJ, Jackson RT. Prostate cancer risk and consumption of fish oils: a dietary biomarker-based case-control study. Br J Cancer 1999;81:1238–42.
  9. Terry P, Lichtenstein P, Feychting M, Ahlbom A, Wolk A. Fatty fish consumption and risk of prostate cancer. Lancet 2001;357:1764–6.
  10. Terry P, Wolk A, Vainio H, Weiderpass E. Fatty fish consumption lowers the risk of endometrial cancer: a nationwide case-control study in Sweden. Cancer Epidemiol Biomarkers Prev 2002;11:143–5.
  11. Willett WC, Stampfer MJ, Colditz GA, Rosner BA, Speizer FE. Relation of meat, fat, and fiber intake to the risk of colon cancer in a prospective study among women. N Engl J Med 1990;323:1664–72.
  12. Favero A, Parpinel M, Franceschi S. Diet and risk of breast cancer: major findings from an Italian case-control study. Biomed Pharmacother 1998;52:109–15.
  13. Franceschi S, Favero A, La Vecchia C, et al. Influence of food groups and food diversity on breast cancer risk in Italy. Int J Cancer 1995;63:785–9.
  14. Braga C, La Vecchia C, Negri E, Franceschi S, Parpinel M. Intake of selected foods and nutrients and breast cancer risk: an age- and menopause-specific analysis. Nutr Cancer 1997;28:258–63.
  15. Bosetti C, Negri E, Franceschi S, et al. Diet and ovarian cancer risk: a case-control study in Italy. Int J Cancer 2001;93:911–5.
  16. Augustsson K, Michaud DS, Rimm EB, et al. A prospective study of intake of fish and marine fatty acids and prostate cancer. Cancer Epidemiol Biomarkers Prev 2003;12:64–7.
  17. Chajes V, Hulten K, Van Kappel AL, et al. Fatty-acid composition in serum phospholipids and risk of breast cancer: an incident case-control study in Sweden. Int J Cancer 1999;83:585–90.
  18. Holmes MD, Hunter DJ, Colditz GA, et al. Association of dietary intake of fat and fatty acids with risk of breast cancer. JAMA 1999;281:914–20.
  19. Schuurman AG, van den Brandt PA, Dorant E, Goldbohm RA. Animal products, calcium and protein and prostate cancer risk in The Netherlands Cohort Study. Br J Cancer 1999;80:1107–13.
  20. Severson RK, Nomura AM, Grove JS, Stemmermann GN. A prospective study of demographics, diet, and prostate cancer among men of Japanese ancestry in Hawaii. Cancer Res 1989;49:1857–60.
  21. Toniolo P, Riboli E, Shore RE, Pasternack BS. Consumption of meat, animal products, protein, and fat and risk of breast cancer: a prospective cohort study in New York. Epidemiology 1994;5:391–7.
  22. Vatten LJ, Solvoll K, Loken EB. Frequency of meat and fish intake and risk of breast cancer in a prospective study of 14,500 Norwegian women. Int J Cancer 1990;46:12–5.
  23. Gertig DM, Hankinson SE, Hough H, et al. N-acetyl transferase 2 genotypes, meat intake and breast cancer risk. Int J Cancer 1999;80:13–7.
  24. Giovannucci E, Rimm EB, Stampfer MJ, Colditz GA, Ascherio A, Willett WC. Intake of fat, meat, and fiber in relation to risk of colon cancer in men. Cancer Res 1994;54:2390–7.
  25. Kampman E, Slattery ML, Bigler J, et al. Meat consumption, genetic susceptibility, and colon cancer risk: a United States multicenter case-control study. Cancer Epidemiol Biomarkers Prev 1999;8:15–24.
  26. Karmali RA, Marsh J, Fuchs C. Effect of omega-3 fatty acids on growth of a rat mammary tumor. J Natl Cancer Inst 1984;73:457–61.
  27. Lindner MA. A fish oil diet inhibits colon cancer in mice. Nutr Cancer 1991;15:1–11.
  28. Rose DP, Connolly JM, Meschter CL. Effect of dietary fat on human breast cancer growth and lung metastasis in nude mice. J Natl Cancer Inst 1991;83:1491–5.
  29. Tsai WS, Nagawa H, Kaizaki S, Tsuruo T, Muto T. Inhibitory effects of n–3 polyunsaturated fatty acids on sigmoid colon cancer transformants. J Gastroenterol 1998;33:206–12.
  30. Boudreau MD, Sohn KH, Rhee SH, Lee SW, Hunt JD, Hwang DH. 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;61:1386–91.
  31. Narayanan BA, Narayanan NK, Reddy BS. Docosahexaenoic acid regulated genes and transcription factors inducing apoptosis in human colon cancer cells. Int J Oncol 2001;19:1255–62.
  32. Anti M, Armelao F, Marra G, et al. Effects of different doses of fish oil on rectal cell proliferation in patients with sporadic colonic adenomas. Gastroenterology 1994;107:1709–18.
  33. Anti M, Marra G, Armelao F, et al. Effect of omega-3 fatty acids on rectal mucosal cell proliferation in subjects at risk for colon cancer. Gastroenterology 1992;103:883–91.
  34. Rose DP, Connolly JM. Omega-3 fatty acids as cancer chemopreventive agents. Pharmacol Ther 1999;83:217–44.
  35. Okuyama H, Kobayashi T, Watanabe S. Dietary fatty acids–the n–6/n–3 balance and chronic elderly diseases. Excess linoleic acid and relative n–3 deficiency syndrome seen in Japan. Prog Lipid Res 1996;35:409–57.
  36. Bartsch H, Nair J, Owen RW. Dietary polyunsaturated fatty acids and cancers of the breast and colorectum: emerging evidence for their role as risk modifiers. Carcinogenesis 1999;20:2209–18.
  37. Bougnoux P. n–3 Polyunsaturated fatty acids and cancer. Curr Opin Clin Nutr Metab Care 1999;2:121–6.
  38. Stoll BA. n–3 Fatty acids and lipid peroxidation in breast cancer inhibition. Br J Nutr 2002;87:193–8.
  39. Cowing BE, Saker KE. Polyunsaturated fatty acids and epidermal growth factor receptor/mitogen-activated protein kinase signaling in mammary cancer. J Nutr 2001;131:1125–8.
  40. Calder PC, Grimble RF. Polyunsaturated fatty acids, inflammation and immunity. Eur J Clin Nutr 2002;56(suppl):S14–9.
  41. Calder PC, Yaqoob P, Thies F, Wallace FA, Miles EA. Fatty acids and lymphocyte functions. Br J Nutr 2002;87(suppl):S31–48.
  42. Raud J, Dahlen SE, Sydbom A, Lindbom L, Hedqvist P. Enhancement of acute allergic inflammation by indomethacin is reversed by prostaglandin E2: apparent correlation with in vivo modulation of mediator release. Proc Natl Acad Sci U S A 1988;85:2315–9.
  43. Chulada PC, Thompson MB, Mahler JF, et al. Genetic disruption of Ptgs-1, as well as Ptgs-2, reduces intestinal tumorigenesis in Min mice. Cancer Res 2000;60:4705–8.
  44. Cheuk BL, Chew SB, Fiscus RR, Wong PY. Cyclooxygenase-2 regulates apoptosis in rat epididymis through prostaglandin D2. Biol Reprod 2002;66:374–80.
  45. Tsutsumi S, Haruna R, Tomisato W, et al. Effects of prostaglandins on spontaneous apoptosis in gastric mucosal cells. Dig Dis Sci 2002;47:84–9.
  46. Leahy KM, Ornberg RL, Wang Y, Zweifel BS, Koki AT, Masferrer JL. Cyclooxygenase-2 inhibition by celecoxib reduces proliferation and induces apoptosis in angiogenic endothelial cells in vivo. Cancer Res 2002;62:625–31.
  47. Rose DP, Connolly JM. Antiangiogenicity of docosahexaenoic acid and its role in the suppression of breast cancer cell growth in nude mice. Int J Oncol 1999;15:1011–5.
  48. 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;121:1339–47.
  49. Pai R, Szabo IL, Soreghan BA, Atay S, Kawanaka H, Tarnawski AS. PGE(2) stimulates VEGF expression in endothelial cells via ERK2/JNK1 signaling pathways. Biochem Biophys Res Commun 2001;286:923–8.
  50. Pidgeon GP, Kandouz M, Meram A, Honn KV. Mechanisms controlling cell cycle arrest and induction of apoptosis after 12-lipoxygenase inhibition in prostate cancer cells. Cancer Res 2002;62:2721–7.
  51. Ding XZ, Tong WG, Adrian TE. 12-Lipoxygenase metabolite 12(S)-HETE stimulates human pancreatic cancer cell proliferation via protein tyrosine phosphorylation and ERK activation. Int J Cancer 2001;94:630–6.
  52. Rose DP, Connolly JM. Regulation of tumor angiogenesis by dietary fatty acids and eicosanoids. Nutr Cancer 2000;37:119–27.
  53. Honn KV, Nelson KK, Renaud C, Bazaz R, Diglio CA, Timar J. Fatty acid modulation of tumor cell adhesion to microvessel endothelium and experimental metastasis. Prostaglandins 1992;44:413–29.
  54. Lipkin M, Reddy B, Newmark H, Lamprecht SA. Dietary factors in human colorectal cancer. Annu Rev Nutr 1999;19:545–86.
  55. Damtew B, Spagnuolo PJ. Tumor cell-endothelial cell interactions: evidence for roles for lipoxygenase products of arachidonic acid in metastasis. Prostaglandins Leukot Essent Fatty Acids 1997;56:295–300.
  56. Kroetz DL, Zeldin DC. Cytochrome P450 pathways of arachidonic acid metabolism. Curr Opin Lipidol 2002;13:273–83.
  57. Chen JK, Capdevila J, Harris RC. Cytochrome p450 epoxygenase metabolism of arachidonic acid inhibits apoptosis. Mol Cell Biol 2001;21:6322–31.
  58. Chen JK, Falck JR, Reddy KM, Capdevila J, Harris RC. Epoxyeicosatrienoic acids and their sulfonimide derivatives stimulate tyrosine phosphorylation and induce mitogenesis in renal epithelial cells. J Biol Chem 1998;273:29254–61.
  59. Schneider MR, Tang DG, Schirner M, Honn KV. Prostacyclin and its analogues: antimetastatic effects and mechanisms of action. Cancer Metastasis Rev 1994;13:349–64.
  60. Bishop-Bailey D, Calatayud S, Warner TD, Hla T, Mitchell JA. Prostaglandins and the regulation of tumor growth. J Environ Pathol Toxicol Oncol 2002;21:93–101.
  61. Tang S, Bhatia B, Maldonado CJ, et al. Evidence that arachidonate 15-lipoxygenase 2 is a negative cell cycle regulator in normal prostate epithelial cells. J Biol Chem 2002;277:16189–201.
  62. Shureiqi I, Chen D, Lee JJ, et al. 15-LOX-1: a novel molecular target of nonsteroidal anti-inflammatory drug-induced apoptosis in colorectal cancer cells. J Natl Cancer Inst 2000;92:1136–42.
  63. Shureiqi I, Wojno KJ, Poore JA, et al. Decreased 13-S-hydroxyoctadecadienoic acid levels and 15-lipoxygenase-1 expression in human colon cancers. Carcinogenesis 1999;20:1985–95.
  64. Crawford M, Galli C, Visioli F, Renaud S, Simopoulos AP, Spector AA. Role of plant-derived omega-3 fatty acids in human nutrition. Ann Nutr Metab 2000;44:263–5.
  65. Hamid R, Singh J, Reddy BS, Cohen LA. Inhibition by dietary menhaden oil of cyclooxygenase-1 and -2 in N-nitrosomethylurea-induced rat mammary tumors. Int J Oncol 1999;14:523–8.
  66. Ringbom T, Huss U, Stenholm A, et al. Cox-2 inhibitory effects of naturally occurring and modified fatty acids. J Nat Prod 2001;64:745–9.
  67. Singh J, Hamid R, Reddy BS. Dietary fat and colon cancer: modulation of cyclooxygenase-2 by types and amount of dietary fat during the postinitiation stage of colon carcinogenesis. Cancer Res 1997;57:3465–70.
  68. Culp BR, Titus BG, Lands WE. Inhibition of prostaglandin biosynthesis by eicosapentaenoic acid. Prostaglandins Med 1979;3:269–78.
  69. Marshall LA, Johnston PV. Modulation of tissue prostaglandin synthesizing capacity by increased ratios of dietary alpha-linolenic acid to linoleic acid. Lipids 1982;17:905–13.
  70. Corey EJ, Shih C, Cashman JR. Docosahexaenoic acid is a strong inhibitor of prostaglandin but not leukotriene biosynthesis. Proc Natl Acad Sci U S A 1983;80:3581–4.
  71. Grimm H, Mayer K, Mayser P, Eigenbrodt E. Regulatory potential of n–3 fatty acids in immunological and inflammatory processes. Br J Nutr 2002;87(suppl):S59–67.
  72. Badawi AF, Archer MC. Effect of hormonal status on the expression of the cyclooxygenase 1 and 2 genes and prostaglandin synthesis in rat mammary glands. Prostaglandins Other Lipid Mediat 1998;56:167–81.
  73. von Schacky C, Kiefl R, Marcus AJ, Broekman MJ, Kaminski WE. Dietary n–3 fatty acids accelerate catabolism of leukotriene B4 in human granulocytes. Biochim Biophys Acta 1993;1166:20–4.
  74. Tsunomori M, Fujimoto Y, Muta E, Nishida H, Sakuma S, Fujita T. 15-Hydroperoxyeicosapentaenoic acid inhibits arachidonic acid metabolism in rabbit platelets more potently than eicosapentaenoic acid. Biochim Biophys Acta 1996;1300:171–6.
  75. Garg ML, Sebokova E, Wierzbicki A, Thomson AB, Clandinin MT. Differential effects of dietary linoleic and alpha-linolenic acid on lipid metabolism in rat tissues. Lipids 1988;23:847–52.
  76. Christiansen EN, Lund JS, Rortveit T, Rustan AC. Effect of dietary n–3 and n–6 fatty acids on fatty acid desaturation in rat liver. Biochim Biophys Acta 1991;1082:57–62.
  77. Jump DB. The biochemistry of n–3 polyunsaturated fatty acids. J Biol Chem 2002;277:8755–8.
  78. Grimaldi PA. Fatty acid regulation of gene expression. Curr Opin Clin Nutr Metab Care 2001;4:433–7.
  79. Vamecq J, Latruffe N. Medical significance of peroxisome proliferator-activated receptors. Lancet 1999;354:141–8.
  80. Berger J, Moller DE. The mechanisms of action of PPARs. Annu Rev Med 2002;53:409–35.
  81. Houseknecht KL, Cole BM, Steele PJ. Peroxisome proliferator-activated receptor gamma (PPARgamma) and its ligands: a review. Domest Anim Endocrinol 2002;22:1–23.
  82. Schild RL, Schaiff WT, Carlson MG, Cronbach EJ, Nelson DM, Sadovsky Y. The activity of PPAR gamma in primary human trophoblasts is enhanced by oxidized lipids. J Clin Endocrinol Metab 2002;87:1105–10.
  83. Rosen ED, Spiegelman BM. PPARgamma: a nuclear regulator of metabolism, differentiation, and cell growth. J Biol Chem 2001;276:37731–4.
  84. Chambrier C, Bastard JP, Rieusset J, et al. Eicosapentaenoic acid induces mRNA expression of peroxisome proliferator-activated receptor gamma. Obes Res 2002;10:518–25.
  85. Gottlicher M, Widmark E, Li Q, Gustafsson JA. Fatty acids activate a chimera of the clofibric acid-activated receptor and the glucocorticoid receptor. Proc Natl Acad Sci U S A 1992;89:4653–7.
  86. Price PT, Nelson CM, Clarke SD. Omega-3 polyunsaturated fatty acid regulation of gene expression. Curr Opin Lipidol 2000;11:3–7.
  87. Mueller E, Sarraf P, Tontonoz P, et al. Terminal differentiation of human breast cancer through PPAR gamma. Mol Cell 1998;1:465–70.
  88. Sarraf P, Mueller E, Jones D, et al. Differentiation and reversal of malignant changes in colon cancer through PPARgamma. Nat Med 1998;4:1046–52.
  89. Kubota T, Koshizuka K, Williamson EA, et al. Ligand for peroxisome proliferator-activated receptor gamma (troglitazone) has potent antitumor effect against human prostate cancer both in vitro and in vivo. Cancer Res 1998;58:3344–52.
  90. Sarraf P, Mueller E, Smith WM, et al. Loss-of-function mutations in PPAR gamma associated with human colon cancer. Mol Cell 1999;3:799–804.
  91. Clay CE, Namen AM, Atsumi G, et al. Influence of J series prostaglandins on apoptosis and tumorigenesis of breast cancer cells. Carcinogenesis 1999;20:1905–11.
  92. Chang TH, Szabo E. Enhanced growth inhibition by combination differentiation therapy with ligands of peroxisome proliferator-activated receptor-gamma and inhibitors of histone deacetylase in adenocarcinoma of the lung. Clin Cancer Res 2002;8:1206–12.
  93. Mueller E, Smith M, Sarraf P, et al. Effects of ligand activation of peroxisome proliferator-activated receptor gamma in human prostate cancer. Proc Natl Acad Sci U S A 2000;97:10990–5.
  94. Diep QN, Touyz RM, Schiffrin EL. Docosahexaenoic acid, a peroxisome proliferator-activated receptor-alpha ligand, induces apoptosis in vascular smooth muscle cells by stimulation of p38 mitogen-activated protein kinase. Hypertension 2000;36:851–5.
  95. Hisatake JI, Ikezoe T, Carey M, Holden S, Tomoyasu S, Koeffler HP. Down-regulation of prostate-specific antigen expression by ligands for peroxisome proliferator-activated receptor gamma in human prostate cancer. Cancer Res 2000;60:5494–8.
  96. Shappell SB, Gupta RA, Manning S, et al. 15S-Hydroxyeicosatetraenoic acid activates peroxisome proliferator-activated receptor gamma and inhibits proliferation in PC3 prostate carcinoma cells. Cancer Res 2001;61:497–503.
  97. Murata M, Kaji H, Iida K, Okimura Y, Chihara K. Dual action of eicosapentaenoic acid in hepatoma cells: up-regulation of metabolic action of insulin and inhibition of cell proliferation. J Biol Chem 2001;276:31422–8.
  98. Schwartz SA, Hernandez A, Mark Evers B. The role of NF-kappaB/IkappaB proteins in cancer: implications for novel treatment strategies. Surg Oncol 1999;8:143–53.
  99. Novak TE, Babcock TA, Jho DH, Helton WS, Espat NJ. NF-kappa B inhibition by omega-3 fatty acids modulates LPS-stimulated macrophage TNF-alpha transcription. Am J Physiol 2003;284:L84–9.
  100. Collett ED, Davidson LA, Fan YY, Lupton JR, Chapkin RS. n–6 And n–3 polyunsaturated fatty acids differentially modulate oncogenic Ras activation in colonocytes. Am J Physiol 2001;280:C1066–75.
  101. Chang WL, Chapkin RS, Lupton JR. Fish oil blocks azoxymethane-induced rat colon tumorigenesis by increasing cell differentiation and apoptosis rather than decreasing cell proliferation. J Nutr 1998;128:491–7.
  102. Jiang YH, Lupton JR, Chapkin RS. Dietary fish oil blocks carcinogen-induced down-regulation of colonic protein kinase C isozymes. Carcinogenesis 1997;18:351–7.
  103. Murray NR, Weems C, Chen L, et al. Protein kinase C betaII and TGFbetaRII in omega-3 fatty acid-mediated inhibition of colon carcinogenesis. J Cell Biol 2002;157:915–20.
  104. Murray NR, Davidson LA, Chapkin RS, Clay Gustafson W, Schattenberg DG, Fields AP. Overexpression of protein kinase C betaII induces colonic hyperproliferation and increased sensitivity to colon carcinogenesis. J Cell Biol 1999;145:699–711.
  105. Gokmen-Polar Y, Murray NR, Velasco MA, Gatalica Z, Fields AP. Elevated protein kinase C betaII is an early promotive event in colon carcinogenesis. Cancer Res 2001;61:1375–81.
  106. Giardiello FM, Hamilton SR, Hylind LM, Yang VW, Tamez P, Casero RA Jr. Ornithine decarboxylase and polyamines in familial adenomatous polyposis. Cancer Res 1997;57:199–201.
  107. Hixson LJ, Garewal HS, McGee DL, et al. Ornithine decarboxylase and polyamines in colorectal neoplasia and mucosa. Cancer Epidemiol Biomarkers Prev 1993;2:369–74.
  108. Rao CV, Reddy BS. Modulating effect of amount and types of dietary fat on ornithine decarboxylase, tyrosine protein kinase and prostaglandins production during colon carcinogenesis in male F344 rats. Carcinogenesis 1993;14:1327–33.
  109. Bartram HP, Gostner A, Scheppach W, et al. Effects of fish oil on rectal cell proliferation, mucosal fatty acids, and prostaglandin E2 release in healthy subjects. Gastroenterology 1993;105:1317–22.
  110. El-Sohemy A, Archer MC. Regulation of mevalonate synthesis in rat mammary glands by dietary n–3 and n–6 polyunsaturated fatty acids. Cancer Res 1997;57:3685–7.
  111. Froyland L, Vaagenes H, Asiedu DK, Garras A, Lie O, Berge RK. Chronic administration of eicosapentaenoic acid and docosahexaenoic acid as ethyl esters reduced plasma cholesterol and changed the fatty acid composition in rat blood and organs. Lipids 1996;31:169–78.
  112. Hromadova M, Sebokova E, Klimes I. HMG-CoA reductase activity in the liver of rats with hereditary hypertriglyceridemia: effect of dietary fish oil. Endocr Regul 1994;28:211–4.
  113. Choi YS, Goto S, Ikeda I, Sugano M. Effect of dietary n–3 polyunsaturated fatty acids on cholesterol synthesis and degradation in rats of different ages. Lipids 1989;24:45–50.
  114. El-Sohemy A, Archer MC. Regulation of mevalonate synthesis in low density lipoprotein receptor knockout mice fed n–3 or n–6 polyunsaturated fatty acids. Lipids 1999;34:1037–43.
  115. Park HJ, Kong D, Iruela-Arispe L, Begley U, Tang D, Galper JB. 3-Hydroxy-3-methylglutaryl coenzyme A reductase inhibitors interfere with angiogenesis by inhibiting the geranylgeranylation of RhoA. Circ Res 2002;91:143–50.
  116. Harris WS. n–3 Fatty acids and serum lipoproteins: human studies. Am J Clin Nutr 1997;65(suppl):1645S–54S.
  117. Singh J, Hamid R, Reddy BS. Dietary fish oil inhibits the expression of farnesyl protein transferase and colon tumor development in rodents. Carcinogenesis 1998;19:985–9.
  118. Badawi AF, El-Sohemy A, Stephen LL, Ghoshal AK, Archer MC. The effect of dietary n–3 and n–6 polyunsaturated fatty acids on the expression of cyclooxygenase 1 and 2 and levels of p21ras in rat mammary glands. Carcinogenesis 1998;19:905–10.
  119. Williams CS, Mann M, DuBois RN. The role of cyclooxygenases in inflammation, cancer, and development. Oncogene 1999;18:7908–16.
  120. Dempke W, Rie C, Grothey A, Schmoll HJ. Cyclooxygenase-2: a novel target for cancer chemotherapy? J Cancer Res Clin Oncol 2001;127:411–7.
  121. Thun MJ, Henley SJ, Patrono C. Nonsteroidal anti-inflammatory drugs as anticancer agents: mechanistic, pharmacologic, and clinical issues. J Natl Cancer Inst 2002;94:252–66.
  122. Dannenberg AJ, Zakim D. Chemoprevention of colorectal cancer through inhibition of cyclooxygenase-2. Semin Oncol 1999;26:499–504.
  123. Wiseman H, Halliwell B. Damage to DNA by reactive oxygen and nitrogen species: role in inflammatory disease and progression to cancer. Biochem Genet 1996;313:17–29.
  124. Lala PK, Chakraborty C. Role of nitric oxide in carcinogenesis and tumour progression. Lancet Oncol 2001;2:149–56.
  125. Ambs S, Hussain SP, Harris CC. Interactive effects of nitric oxide and the p53 tumor suppressor gene in carcinogenesis and tumor progression. FASEB J 1997;11:443–8.
  126. Jadeski LC, Chakraborty C, Lala PK. Role of nitric oxide in tumour progression with special reference to a murine breast cancer model. Can J Physiol Pharmacol 2002;80:125–35.
  127. Cooke JP, Losordo DW. Nitric oxide and angiogenesis. Circulation 2002;105:2133–5.
  128. Wink DA, Vodovotz Y, Laval J, Laval F, Dewhirst MW, Mitchell JB. The multifaceted roles of nitric oxide in cancer. Carcinogenesis 1998;19:711–21.
  129. Ohata T, Fukuda K, Takahashi M, Sugimura T, Wakabayashi K. Suppression of nitric oxide production in lipopolysaccharide-stimulated macrophage cells by omega 3 polyunsaturated fatty acids. Jpn J Cancer Res 1997;88:234–7.
  130. Khair-El-Din T, Sicher SC, Vazquez MA, et al. Transcription of the murine iNOS gene is inhibited by docosahexaenoic acid, a major constituent of fetal and neonatal sera as well as fish oils. J Exp Med 1996;183:1241–6.
  131. Jeyarajah DR, Kielar M, Penfield J, Lu CY. Docosahexaenoic acid, a component of fish oil, inhibits nitric oxide production in vitro. J Surg Res 1999;83:147–50.
  132. Lu CY, Penfield JG, Khair-el-Din TA, et al. Docosahexaenoic acid, a constituent of fetal and neonatal serum, inhibits nitric oxide production by murine macrophages stimulated by IFN gamma plus LPS, or by IFN gamma plus Listeria monocytogenes. J Reprod Immunol 1998;38:31–53.
  133. Noble LS, Takayama K, Zeitoun KM, et al. Prostaglandin E2 stimulates aromatase expression in endometriosis-derived stromal cells. J Clin Endocrinol Metab 1997;82:600–6.
  134. Fang JL, Vaca CE, Valsta LM, Mutanen M. Determination of DNA adducts of malonaldehyde in humans: effects of dietary fatty acid composition. Carcinogenesis 1996;17:1035–40.
  135. Nair J, Vaca CE, Velic I, Mutanen M, Valsta LM, Bartsch H. High dietary omega-6 polyunsaturated fatty acids drastically increase the formation of etheno-DNA base adducts in white blood cells of female subjects. Cancer Epidemiol Biomarkers Prev 1997;6:597–601.
  136. Cosgrove JP, Church DF, Pryor WA. The kinetics of the autoxidation of polyunsaturated fatty acids. Lipids 1987;22:299–304.
  137. Higdon JV, Du SH, Lee YS, Wu T, Wander RC. Supplementation of postmenopausal women with fish oil does not increase overall oxidation of LDL ex vivo compared to dietary oils rich in oleate and linoleate. J Lipid Res 2001;42:407–18.
  138. Bonanome A, Biasia F, De Luca M, et al. n–3 Fatty acids do not enhance LDL susceptibility to oxidation in hypertriacylglycerolemic hemodialyzed subjects. Am J Clin Nutr 1996;63:261–6.
  139. Wander RC, Du SH, Ketchum SO, Rowe KE. Effects of interaction of RRR--tocopheryl acetate and fish oil on low-density-lipoprotein oxidation in postmenopausal women with and without hormone-replacement therapy. Am J Clin Nutr 1996;63:184–93.
  140. Takahashi M, Tsuboyama-Kasaoka N, Nakatani T, et al. Fish oil feeding alters liver gene expressions to defend against PPARalpha activation and ROS production. Am J Physiol 2002;282:G338–48.
  141. Chajes V, Sattler W, Stranzl A, Kostner GM. Influence of n–3 fatty acids on the growth of human breast cancer cells in vitro: relationship to peroxides and vitamin-E. Breast Cancer Res Treat 1995;34:199–212.
  142. Lhuillery C, Cognault S, Germain E, Jourdan ML, Bougnoux P. Suppression of the promoter effect of polyunsaturated fatty acids by the absence of dietary vitamin E in experimental mammary carcinoma. Cancer Lett 1997;114:233–4.
  143. Dommels YE, Haring MM, Keestra NG, Alink GM, van Bladeren PJ, van Ommen B. The role of cyclooxygenase in n–6 and n–3 polyunsaturated fatty acid mediated effects on cell proliferation, PGE(2) synthesis and cytotoxicity in human colorectal carcinoma cell lines. Carcinogenesis 2003;24:385–92.
  144. Jenkins DJ, Kendall CW, Vidgen E, et al. Health aspects of partially defatted flaxseed, including effects on serum lipids, oxidative measures, and ex vivo androgen and progestin activity: a controlled crossover trial. Am J Clin Nutr 1999;69:395–402.
  145. Mori Y, Murakawa Y, Katoh S, et al. Influence of highly purified eicosapentaenoic acid ethyl ester on insulin resistance in the Otsuka Long-Evans Tokushima Fatty rat, a model of spontaneous non-insulin-dependent diabetes mellitus. Metabolism 1997;46:1458–64.
  146. Mori Y, Murakawa Y, Yokoyama J, et al. Effect of highly purified eicosapentaenoic acid ethyl ester on insulin resistance and hypertension in Dahl salt-sensitive rats. Metabolism 1999;48:1089–95.
  147. Minami A, Ishimura N, Sakamoto S, et al. Effect of eicosapentaenoic acid ethyl ester v. oleic acid–rich safflower oil on insulin resistance in type 2 diabetic model rats with hypertriacylglycerolaemia. Br J Nutr 2002;87:157–62.
  148. Popp-Snijders C, Schouten JA, Heine RJ, van der Meer J, van der Veen EA. Dietary supplementation of omega-3 polyunsaturated fatty acids improves insulin sensitivity in non-insulin-dependent diabetes. Diabetes Res 1987;4:141–7.
  149. Picard F, Auwerx J. PPAR gamma and glucose homeostasis. Annu Rev Nutr 2002;22:167–97.
  150. Storlien LH, Kriketos AD, Calvert GD, Baur LA, Jenkins AB. Fatty acids, triglycerides and syndromes of insulin resistance. Prostaglandins Leukot Essent Fatty Acids 1997;57:379–85.
  151. Iigo M, Nakagawa T, Ishikawa C, et al. Inhibitory effects of docosahexaenoic acid on colon carcinoma 26 metastasis to the lung. Br J Cancer 1997;75:650–5.
  152. Livsmedelsverket. [The National Food Administration.] Livsmedelstabell—fettsyror. [Food chart-fatty acids.] Jönköping, Sweden: Tryckeri AB Småland, 1998 (in Swedish).
  153. Vermunt SH, Mensink RP, Simonis MM, Hornstra G. Effects of dietary alpha-linolenic acid on the conversion and oxidation of 13C-alpha-linolenic acid. Lipids 2000;35:137–42.
  154. Burdge GC, Finnegan YE, Minihane AM, Williams CM, Wootton SA. Effect of altered dietary n–3 fatty acid intake upon plasma lipid fatty acid composition, conversion of [13C]alpha-linolenic acid to longer-chain fatty acids and partitioning towards beta-oxidation in older men. Br J Nutr 2003;90:311–21.
  155. Emken EA, Adlof RO, Gulley RM. Dietary linoleic acid influences desaturation and acylation of deuterium-labeled linoleic and linolenic acids in young adult males. Biochim Biophys Acta 1994;1213:277–88.
  156. Jones PJH, Kubow S. Lipids, sterols, and their metabolites. In: Shiels ME, Olson JA, Shike M, Ross CA, eds. Modern nutrition in health and disease. Baltimore: Williams and Wilkins, 1999:67–94.
  157. Pawlosky RJ, Hibbeln JR, Novotny JA, Salem N Jr. Physiological compartmental analysis of alpha-linolenic acid metabolism in adult humans. J Lipid Res 2001;42:1257–65.
  158. Simonsen N, van't Veer P, Strain JJ, et al. Adipose tissue omega-3 and omega-6 fatty acid content and breast cancer in the EURAMIC study. European Community Multicenter Study on Antioxidants, Myocardial Infarction, and Breast Cancer. Am J Epidemiol 1998;147:342–52.
  159. Simopoulos AP. The Mediterranean diets: what is so special about the diet of Greece? The scientific evidence. J Nutr 2001;131(suppl):3065S–73S.
  160. James MJ, Gibson RA, D'Angelo M, Neumann MA, Cleland LG. Simple relationships exist between dietary linoleate and the n–6 fatty acids of human neutrophils and plasma. Am J Clin Nutr 1993;58:497–500.
  161. Caughey GE, Mantzioris E, Gibson RA, Cleland LG, James MJ. The effect on human tumor necrosis factor alpha and interleukin 1 beta production of diets enriched in n–3 fatty acids from vegetable oil or fish oil. Am J Clin Nutr 1996;63:116–22.
  162. Zock PL, Katan MB. Linoleic acid intake and cancer risk: a review and meta-analysis. Am J Clin Nutr 1998;68:142–53.
  163. Whelan J, Surette ME, Hardardottir I, et al. Dietary arachidonate enhances tissue arachidonate levels and eicosanoid production in Syrian hamsters. J Nutr 1993;123:2174–85.
  164. Keating GA, Bogen KT. Methods for estimating heterocyclic amine concentrations in cooked meats in the US diet. Food Chem Toxicol 2001;39:29–43.
  165. Sugimura T. Nutrition and dietary carcinogens. Carcinogenesis 2000;21:387–95.
Received for publication September 22, 2003. Accepted for publication December 2, 2003.


作者: Susanna C Larsson
医学百科App—中西医基础知识学习工具
  • 相关内容
  • 近期更新
  • 热文榜
  • 医学百科App—健康测试工具