Hostname: page-component-8448b6f56d-mp689 Total loading time: 0 Render date: 2024-04-23T08:16:45.770Z Has data issue: false hasContentIssue false

Bioactive dietary long-chain fatty acids: emerging mechanisms of action

Published online by Cambridge University Press:  01 December 2008

Robert S. Chapkin*
Affiliation:
Faculty of Nutrition, Texas A&M University, College Station, TX, USA Center for Environmental and Rural Health, Texas A&M University, College Station, TX, USA Vegetable and Fruit Improvement Center, Texas A&M University, College Station, TX, USA
David N. McMurray
Affiliation:
Faculty of Nutrition, Texas A&M University, College Station, TX, USA Center for Environmental and Rural Health, Texas A&M University, College Station, TX, USA Department of Microbial and Molecular Pathogenesis, Texas A&M University Health Science Center, College Station, TX, USA
Laurie A. Davidson
Affiliation:
Faculty of Nutrition, Texas A&M University, College Station, TX, USA Center for Environmental and Rural Health, Texas A&M University, College Station, TX, USA
Bhimanagouda S. Patil
Affiliation:
Faculty of Nutrition, Texas A&M University, College Station, TX, USA Vegetable and Fruit Improvement Center, Texas A&M University, College Station, TX, USA
Yang-Yi Fan
Affiliation:
Faculty of Nutrition, Texas A&M University, College Station, TX, USA
Joanne R. Lupton
Affiliation:
Faculty of Nutrition, Texas A&M University, College Station, TX, USA Center for Environmental and Rural Health, Texas A&M University, College Station, TX, USA
*
*Corresponding author: Dr Robert S. Chapkin, fax +1 979 862 2378, email r-chapkin@tamu.edu
Rights & Permissions [Opens in a new window]

Abstract

The plasma membranes of all eukaryotic cells contain heterogeneous self-organising intrinsically unstable liquid ordered domains or lipid assemblies in which key signal transduction proteins are localised. These assemblies are classified as ‘lipid rafts’ (10–200 nm), which are composed mostly of cholesterol and sphingolipid microdomains and therefore do not integrate well into the fluid phospholipid bilayers. In addition, caveolae represent a subtype of lipid raft macrodomain that form flask-shaped membrane invaginations containing structural proteins, i.e. caveolins. With respect to the diverse biological effects of long-chain PUFA, increasing evidence suggests that n-3 PUFA and perhaps conjugated fatty acids uniquely alter the basic properties of cell membranes. Because of its polyunsaturation, DHA and possibly conjugated linoleic acid are sterically incompatible with sphingolipid and cholesterol and, therefore, appear to alter lipid raft behaviour and protein function. The present review examines the evidence indicating that dietary sources of n-3 PUFA can profoundly alter the biochemical make up of lipid rafts/caveolae microdomains, thereby influencing cell signalling, protein trafficking and cell cytokinetics.

Type
Review Article
Copyright
Copyright © The Authors 2008

The balance between cell proliferation and apoptosis is critical to the maintenance of steady-state cell populations in the body. In general, dysregulation of this mechanism can disrupt homeostasis, resulting in clonal expansion, with the resultant overproduction of affected cells. The programmed induction of cell death also represents a mechanism by which inappropriately activated cells and cells possessing DNA damage can be deleted. It has now been clearly established that chronic inflammation can perturb cellular homeostasis and drive malignant transformation by progressively inhibiting apoptosis of target cell types, for example, T-cells and epithelial cells(Reference Karin1, Reference Rollins2). Hence, chemotherapeutic agents such as dietary long-chain n-3 PUFA and possibly conjugated fatty acid species, which restore the normal proliferative and apoptotic pathways, have the potential for effectively treating cancers that depend on aberrations of these pathways to stay alive(Reference Hong, Lupton, Morris, Wang, Carroll, Davidson, Elder and Chapkin3). The following sections describe a mechanistic membrane-based model which may in part explain the pleiotropic properties of bioactive PUFA.

n-3 Polyunsaturated fatty acids

Although beyond the scope of the present review, a number of investigators have recently addressed the role of n-3 PUFA in suppressing chronic inflammation and cancer(Reference Chapkin, McMurray and Lupton4Reference Serhan, Yacoubian and Yang6). With respect to mechanisms which functionally link the pleiotropic effects of bioactive dietary long-chain n-3 PUFA, inflammation and cancer, examples include (i) metabolic interconversion into novel bioactive eicosanoids(Reference Ariel and Serhan7, Reference Bazan8), (ii) modulation of nuclear receptor activation, gene transcription and translation(Reference deUrquiza, Liu, Sjoberg, Zetterström, Griffiths, Sjövall and Perlmann9Reference Jump, Botolin, Wang, Xu, Christian and Demeure12) (Fig. 1), (iii) alteration of membrane phospholipid composition and functionality of self-organising lipid domains(Reference Ma, Seo, Switzer, Fan, McMurray, Lupton and Chapkin13) (Fig. 2), (iv) effects on protein trafficking, including cytosol-to-membrane translocation(Reference Chapkin, McMurray and Lupton4, Reference Seo, Barhoumi, Johnson, Lupton and Chapkin14) and (v) interaction with SCFA to trigger lipid oxidation and intracellular Ca2+ compartmentalisation(Reference Kolar, Barhoumi, Lupton and Chapkin15, Reference Kolar, Barhoumi, Callaway, Fan, Wang, Lupton and Chapkin16).

Fig. 1 Nuclear receptor activation by conjugated linoleic acid (CLA). FABP, fatty acid-binding proteins (molecular chaperone); ER, endoplasmic reticulum; RXR, retinoid X receptors. CLA transactivates PPAR nuclear receptors. n-3 PUFA suppress NF-κB activation. All membranes incorporate EPA, DHA and conjugated PUFA to different degrees.

Fig. 2 Putative membrane microdomain-altering properties of n-3 PUFA and conjugated linoleic acid (CLA). Dietary DHA and CLA are incorporated into both the bulk phase of the plasma membrane as well as discrete heterogeneous cholesterol/sphingolipid-rich raft domains. This can alter plasma membrane organisation of inner leaflets and the dynamic partitioning of transduction proteins, thereby modulating their function.

The health benefits of long-chain PUFA are diverse and nutritional studies continue to demonstrate important benefits from the consumption of n-3 PUFA(Reference Chapkin, McMurray and Lupton4, Reference Ariel and Serhan7, Reference Bazan8, Reference Chapkin, Wang, Fan, Lupton and Prior17Reference Harris, Poston and Haddock19). Since the use of a health claim on labels for foods containing n-3 PUFA has been approved, food companies are now mobilising to incorporate these fatty acids into a range of novel commercial foods in order to provide for the wider public consumption of these bioactive compounds. Hence, it is now important to precisely determine how specific long-chain PUFA modulate cell phenotype and reduce the risk of developing cancer and inflammatory disorders.

Conjugated linoleic acid

There is growing interest with regard to the use and commercial availability of conjugated positional and geometric isomers of PUFA, particularly conjugated dienoic isomers of linoleic acid (conjugated linoleic acid; CLA). For example, in certain model systems, CLA is a powerful anti-cancer agent, capable of promoting growth arrest and apoptosis in tumour cells(Reference Ou, Ip, Lisafeld and Ip20Reference Song, Sneddon, Heys and Wahle22). In addition, CLA triggers adipose delipidation in rodent species(Reference Chung, Brown, Provo, Hopkins and McIntosh23Reference Pérez-Matute, Marti, Martínez, Fernández-Otero, Stanhope, Havel and Moreno-Aliaga25), and although there has been very little published clinical research(Reference Navarro, Fernandez-Quintela, Churruca and Portillo26, Reference Tricon and Yaqoob27), recent preliminary evidence suggests that mixed-isomer CLA supplementation can alter fat oxidation and energy expenditure in human subjects(Reference Close, Schoeller, Watras and Nora28, Reference Whigham, Watras and Schoeller29). However, the precise mechanism of action remains elusive. Since n-3 PUFA and CLA exhibit overlapping phenotypic properties, we propose a unifying molecular mechanism which may in part explain their protective effects.

Effect of bioactive polyunsaturated fatty acids on cell membranes

It is generally believed that the plasma membrane consists of a mosaic of functional microdomains that facilitate interactions between resident proteins and lipids(Reference Laude and Prior30, Reference Hancock31). Visible examples of these include caveolae, flask-shaped invaginations containing the structural protein caveolin-1 and many signal transduction proteins(Reference Anderson32). In addition, morphologically heterogeneous featureless microdomains, consisting mostly of cholesterol and sphingolipids, unable to integrate well into the fluid phospholipid bilayers, exist as ‘lipid rafts’(Reference Simons and Ikonen33). Although the existence of lipid rafts is still debated, new sophisticated imaging approaches have started to define cell surface nanoscale organisation(Reference Hancock31). Significantly, both cholesterol-dependent microdomains, analogous to lipid rafts, and non-raft signalling microdomains have been observed using electron microscopic imaging of two-dimensional plasma membrane sheets(Reference Prior, Muncke, Parton and Hancock34). These studies have provided a template for further investigation into the effects of dietary PUFA on cell surface organisation and cell cytokinetics, apoptosis and disease progression.

With respect to the biological effects of n-3 PUFA, increasing evidence suggests that DHA is a unique fatty acid because it significantly alters basic properties of cell membranes, including acyl (ester-linked fatty acid) chain order and fluidity, phase behaviour, elastic compressibility, ion permeability, fusion, rapid flip-flop and resident protein function(Reference Stillwell and Wassall35). In part, due to the number of cis double bonds, DHA is sterically incompatible with sphingolipid and cholesterol and, therefore, appears to alter lipid raft behaviour(Reference Stillwell and Wassall35). Interestingly, a number of studies have recently demonstrated that dietary n-3 PUFA are incorporated into diverse cell types and appear to uniquely modulate cell membrane microdomains(Reference Ma, Seo, Switzer, Fan, McMurray, Lupton and Chapkin13, Reference Fan, Ly, Barhoumi, McMurray and Chapkin36Reference Geyeregger, Zeyda, Zlabinger, Waldhäusl and Stulnig39). Indeed, we recently demonstrated that n-3 PUFA feeding can markedly alter lipid/protein composition of mouse colonic caveolae microdomains, thereby selectively modulating the localisation and function of caveolar proteins(Reference Ma, Seo, Switzer, Fan, McMurray, Lupton and Chapkin13, Reference Seo, Barhoumi, Johnson, Lupton and Chapkin14, Reference Ma, Seo, Davidson, Callaway, Fan, Lupton and Chapkin37). In addition, we demonstrated that H-Ras and endothelial NO synthase are displaced from caveolae in n-3 PUFA-fed mice, which was associated with the suppression of Ras-dependent signalling. In contrast, localisation of non-caveolae-resident proteins, K-ras and clathrin, was not affected, indicating selective displacement of acylated signalling proteins from caveolae by n-3 PUFA. Our findings highlight a novel modality by which n-3 PUFA influence membrane micro-organisation, thereby modulating biological responses.

Using T-cell-culture models, Stulnig et al. were the first to document the ability of PUFA enrichment to selectively modify the cytoplasmic layer of lipid rafts(Reference Stulnig, Berger, Sigmund, Raederstorff, Stockinner and Waldhäusl40, Reference Stulnig, Huber, Leitinger, Imre, Angelisová, Nowotny and Waldhäusl41). In complementary experiments, we investigated the effect of dietary n-3 PUFA on cholesterol/sphingolipid-rich plasma membrane microdomains (i.e. rafts) in mouse splenic T-cells(Reference Fowler, Chapkin and McMurray5, Reference Fowler, McMurray, Fan, Aukema and Chapkin42, Reference Switzer, McMurray, Morris and Chapkin43). A very novel and unexpected outcome from this effort was the demonstration that dietary n-3 PUFA reduced (by about 45 %) lipid raft sphingolipid content and altered raft fatty acid composition(Reference Fan, Ly, Barhoumi, McMurray and Chapkin36, Reference Fan, McMurray, Ly and Chapkin44, Reference Switzer, Fan, Wang, McMurray and Chapkin45). Therefore, we hypothesised that PUFA classes (n-6 v. n-3) differentially modulate T-cell membrane microdomains, which is supported by recent studies indicating that stimulation-induced protein kinase C θ translocation into T-cell lipid rafts is suppressed by dietary n-3 PUFA(Reference Fan, Ly, Barhoumi, McMurray and Chapkin36). In addition, in an attempt to further probe the effects of DHA on protein kinase C θ effector pathway signalling, we have recently demonstrated that the diet modification of lipid rafts is associated with the suppression of transcription factor NF-κB, AP-1 transcription site activation, IL-2 secretion and lymphoproliferation(Reference Fan, Ly, Barhoumi, McMurray and Chapkin36). With respect to lymphocyte subsets, recent studies indicate that the macromolecular complex organisation in lipid rafts is distinct in non-polarised, T helper cell, Th1 and Th2 polarised subsets(Reference Leitenberg, Balamuth and Bottomly46). This would suggest that these subsets, i.e. regulators of cell-mediated immunity (Th1) and humoral immunity (Th2), would respond differently to dietary PUFA-induced perturbation. However, the ability of DHA to influence membrane raft-mediated signalling in polarised T-cells has not been determined to date.

There is cogent evidence indicating that lipid raft integrity is a prerequisite for optimised signalling between T-cells and antigen-presenting cells(Reference Gaus, Chklovskaia, Fazekas de St Groth, Jessup and Harder47, Reference Harder, Rentero, Zech and Gaus48). In addition, recent studies suggest that long-chain PUFA can block antigen presentation by interfering with lipid raft-dependent formation of the immunological synapse(Reference Zeyda, Säemann, Stuhlmeier, Mascher, Nowotny, Zlabinger, Waldhäusl and Stulnig38, Reference Geyeregger, Zeyda, Zlabinger, Waldhäusl and Stulnig39, Reference Shaikh and Edidin49). Overall, these findings provide evidence indicating that dietary n-3 PUFA can profoundly alter the biochemical make up of cell membrane lipid rafts/caveolae microdomains, which may directly or indirectly influence membrane fusion and cell–cell signalling. Interestingly, only a single study to date has examined the effects of CLA with regard to lipid raft/caveolae composition. Huot & Ma(Reference Huot and Ma50) demonstrated that mixed CLA isomers are concentrated in caveolae phospholipids, resulting in the reduction of caveolae-resident proteins, caveolin-1 and Her-2/neu, in Michigan Cancer Foundation (MCF)-7 breast cancer cells. Unfortunately, few studies to date have assessed the physical properties of CLA isomers(Reference Yin, Kramer, Yurawecz, Eynard, Mossoba and Yu51). Therefore, future studies using purified CLA isomers are needed in order to elucidate how conjugated fatty acid structure affects membrane structure and function.

Docosahexaenoic acid alters the size and distribution of lipid rafts

In a proof of principle study, we sought to determine the effect of DHA on the size and distribution of lipid rafts in vivo (Reference Chapkin, Wang, Fan, Lupton and Prior17). Using immunogold electron microscopy of plasma membrane sheets coupled with spatial point analysis, morphologically featureless microdomains were visualised in HeLa cells. Cells were transfected with green fluorescent protein truncated H-ras (GFP-tH), which is located exclusively to inner leaflet rafts, and subsequently incubated with DHA and control fatty acid, for example, oleic acid (18 : 1n-9) for 48 h. Univariate K-function analysis of GFP-tH (5 nm gold) revealed that the interparticle distance was significantly reduced by DHA treatment compared with control fatty acid, indicating that select PUFA can increase clustering of proteins in cholesterol-dependent microdomains (GFP-tH), whereas non-raft microdomains are insensitive to DHA modulation. These novel findings suggest that the plasma membrane organisation of inner leaflets is fundamentally altered by DHA enrichment (Fig. 2).

‘Cholesterol-centric’ view of membranes

SFA compared with PUFA have a preferential affinity for cholesterol. This relationship provides the basis for a lipid-driven mechanism for the lateral segregation of membrane elements into cholesterol-rich and -poor microdomains(Reference Stillwell and Wassall35, Reference Huster, Arnold and Gawrisch52Reference Niu and Litman55). For example, unfavourable interaction between cholesterol and PUFA chains has been clearly demonstrated by the exclusion of cholesterol from dipolyunsaturated phosphatidylcholine membranes where it is forced to directly contact polyunsaturated chains. Studies using a variety of techniques including differential scanning calorimetry(Reference Zerouga, Jenski and Stillwell56), 1H NMR and nuclear Overhauser enhancement spectroscopy with magic angle spinning(Reference Huster, Arnold and Gawrisch52), determination of partition coefficients(Reference Kariel, Davidson and Keough57), measurements of lateral compressibility(Reference Needham and Nunn58) and fluorescence anisotropy(Reference Mitchell and Litman53, Reference Mitchell and Litman59) indicate that the poor affinity of DHA and perhaps CLA for cholesterol provides a lipid-driven mechanism for lateral phase separation of cholesterol-rich lipid microdomains from the surrounding bulk membrane. This could in principle alter the size, stability and distribution of cell surface lipid microdomains such as rafts. Indeed, growing evidence from model membrane studies suggest that the energetically less favourable interaction between cholesterol and PUFA, especially DHA, promotes lateral phase segregation into sterol-poor/PUFA-rich and sterol-rich/SFA-rich microdomains(Reference Huster, Arnold and Gawrisch52, Reference Mitchell and Litman53, Reference Kariel, Davidson and Keough57, Reference Brzustowicz, Cherezov, Caffrey, Stillwell and Wassall60Reference Pasenkiewicz-Gierula, Subczynski and Kusumi62).

Huang & Feigenson proposed the umbrella model to describe the solubility and condensing effect of cholesterol within membranes(Reference Huang and Feigenson63). According to this model, phospholipid head groups act as ‘umbrellas’ to prevent the energetically unfavourable contact of the non-polar part of cholesterol with interfacial water. This shielding will be less effective for DHA-containing phospholipids with a large molecular cross-sectional area, facilitating cholesterol precipitation at a lower concentration. In addition, this model allows for speculation that phosphatidylethanolamine with a smaller head group may enhance the DHA-associated reduction in shielding effects relative to phosphatidylcholine. Consistent with this notion, unlike phosphatidylcholine bilayers where a marked reduction in cholesterol solubility requires polyunsaturation at both sn-1 and sn-2 positions, DHA at the sn-2 position with a saturated sn-1 chain is sufficient in phosphatidylethanolamine to trigger cholesterol precipitation(Reference Shaikh, Cherezov, Caffrey, Stillwell and Wassall64).

Conjugated n-3 polyunsaturated fatty acids

There is growing evidence that the combination of conjugated double bonds and n-3 PUFA may have enhanced chemoprotective properties. Recent studies using a number of model systems suggest that conjugated EPA and conjugated DHA suppress tumour growth(Reference Danbara, Yuri, Tsujita-Kyutoku, Sato, Senzaki, Takada, Hada, Miyazawa, Okazaki and Tsubura65, Reference Kimura and Sumiyoshi66), suppress topoisomerases(Reference Yonezawa, Tsuzuki and Eitsuka67), induce apoptosis(Reference Tsuzuki, Tanaka, Kuwahara and Miyazawa68, Reference Tsuzuki, Kambe, Shibata, Kawakami, Nakagawa and Miyazawa69), inhibit lipid accumulation(Reference Tsuzuki, Kawakami, Nakagawa and Miyazawa70) and have potential use as therapeutic dietary supplements for minimising tumour angiogenesis(Reference Tsuzuki, Tanaka, Kuwahara and Miyazawa68, Reference Tsuzuki, Shibata, Kawakami, Nakagaya and Miyazawa71). Typically, conjugated EPA and conjugated DHA are generated by alkaline isomerisation, producing a mixture of isomers with conjugated double bonds, although small amounts are found in marine algae and seal oil. Since the exact structure of these novel fatty acid species has not been fully characterised, future studies are required to verify their safety and efficacy in humans. In addition, it remains to be determined whether conjugated EPA and conjugated DHA alter the size and distribution of cell surface microdomains.

Conclusion

A growing body of literature supports the contention that bioactive food components containing n-3 PUFA are important in suppressing chronic inflammation and cancer. Although the mechanism of EPA and DHA action is still not fully defined in molecular terms, it is becoming increasingly clear that n-3 PUFA alter cell membrane lipid microdomain composition, thereby favourably modulating the relay of extracellular signals from surface receptors to downstream signalling networks. Clearly, further studies are needed to clarify the nature of lipid rafts and the biological role of conjugated fatty acid species, including CLA, conjugated EPA and conjugated DHA families.

Acknowledgements

Financial support by National Institutes of Health grants CA59034, CA129444, DK071707 and P30ES09106, US Department of Agriculture 2005-34 402-16 401 Designing Foods for Health, and the American Institute for Cancer Research is gratefully acknowledged. There are no conflicts of interest associated with this manuscript. R. S. C., L. A. D., Y.-Y. F. and J. R. L. compiled data from collaborative experiments; D. N. M. evaluated the T-cell literature as well as contributions made from collaborative experiments. B. S. P. provided input related to novel fatty acid sources.

References

1Karin, M (2006) Nuclear factor-kB in cancer development and progression. Nature 441, 431436.CrossRefGoogle Scholar
2Rollins, BJ (2006) Inflammatory chemokines in cancer growth and progression. Eur J Cancer 42, 760767.CrossRefGoogle ScholarPubMed
3Hong, MY, Lupton, JR, Morris, JS, Wang, N, Carroll, RJ, Davidson, LA, Elder, RH & Chapkin, RS (2000) Dietary fish oil reduces O6-methylguanine DNA adduct levels in rat colon in part by increasing apoptosis during tumor initiation. Cancer Epidemiol Biomarkers Prev 9, 819826.Google Scholar
4Chapkin, RS, McMurray, DN & Lupton, JR (2007) Colon cancer, fatty acids and anti-inflammatory compounds. Curr Opin Gastroenterol 23, 4854.CrossRefGoogle ScholarPubMed
5Fowler, KH, Chapkin, RS & McMurray, DN (1993) Effects of purified dietary n-3 ethyl esters on murine T-lymphocyte function. J Immunol 151, 51865197.CrossRefGoogle Scholar
6Serhan, CN, Yacoubian, S & Yang, R (2008) Anti-inflammatory and proresolving lipid mediators. Annu Rev Pathol Mech Dis 3, 279312.CrossRefGoogle ScholarPubMed
7Ariel, A & Serhan, CN (2007) Resolvins and protectins in the termination program of acute inflammation. Trends Immunol 28, 176183.CrossRefGoogle ScholarPubMed
8Bazan, NG (2007) Omega-3 fatty acids, pro-inflammatory signaling and neuroprotection. Curr Opin Clin Nutr Metab Care 10, 136141.CrossRefGoogle ScholarPubMed
9deUrquiza, AM, Liu, S, Sjoberg, M, Zetterström, RH, Griffiths, W, Sjövall, J & Perlmann, T (2000) Docosahexaenoic acid, a ligand for the retinoid X receptor in mouse brain. Science 290, 21402144.CrossRefGoogle Scholar
10Fan, YY, Spencer, TE, Wang, N, Moyer, MP & Chapkin, RS (2003) Chemopreventive n-3 fatty acids activate RXRα in colonocytes. Carcinogenesis 4, 15411548.CrossRefGoogle Scholar
11Davidson, LA, Nguyen, DV, Hokanson, RM, et al. (2004) Chemopreventive n-3 polyunsaturated fatty acids reprogram genetic signatures during colon cancer initiation and progression in the rat. Cancer Res 64, 67976804.CrossRefGoogle ScholarPubMed
12Jump, DB, Botolin, D, Wang, Y, Xu, J, Christian, B & Demeure, O (2005) Fatty acid regulation of hepatic gene transcription. J Nutr 135, 25032506.CrossRefGoogle ScholarPubMed
13Ma, DW, Seo, J, Switzer, KC, Fan, YY, McMurray, DN, Lupton, JR & Chapkin, RS (2004) n-3 PUFA and membrane microdomains: a new frontier in bioactive lipid research. J Nutr Biochem 15, 700706.CrossRefGoogle ScholarPubMed
14Seo, J, Barhoumi, R, Johnson, AE, Lupton, JR & Chapkin, RS (2006) Docosahexaenoic acid selectively inhibits plasma membrane targeting of lipidated proteins. FASEB J 20, 770772.CrossRefGoogle ScholarPubMed
15Kolar, SS, Barhoumi, R, Lupton, JR & Chapkin, RS (2007) Docosahexaenoic acid and butyrate synergistically induce colonocyte apoptosis by enhancing mitochondrial Ca2+ accumulation. Cancer Res 67, 55615568.CrossRefGoogle ScholarPubMed
16Kolar, SS, Barhoumi, R, Callaway, ES, Fan, YY, Wang, N, Lupton, JR & Chapkin, RS (2007) Synergy between docosahexaenoic acid and butyrate elicits p53-independent apoptosis via mitochondrial Ca2+ accumulation in colonocytes. Am J Physiol Gastrointest Liver Physiol 293, G935G943.CrossRefGoogle ScholarPubMed
17Chapkin, RS, Wang, N, Fan, YY, Lupton, JR & Prior, IA (2008) Docosahexaenoic acid alters the size and distribution of cell surface microdomains. Biochim Biophys Acta 1778, 466471.CrossRefGoogle ScholarPubMed
18Biscione, F, Pignalberi, C, Totteri, A, Messina, F & Altamura, G (2007) Cardiovascular effects of omega-3 free fatty acids. Curr Vasc Pharmacol 5, 163172.CrossRefGoogle ScholarPubMed
19Harris, WS, Poston, WC & Haddock, CK (2007) Tissue n-3 and n-6 fatty acids and risk for coronary heart disease events. Atherosclerosis 193, 110.CrossRefGoogle ScholarPubMed
20Ou, L, Ip, C, Lisafeld, B & Ip, MM (2007) Conjugated linoleic acid induces apoptosis of murine mammary tumor cells via Bcl-2 loss. Biochem Biophys Res Commun 356, 10441049.CrossRefGoogle ScholarPubMed
21Bozzo, F, Bocca, C, Colombatto, S & Miglietta, A (2007) Antiproliferative effect of conjugated linoleic acid in Caco-2 cells: involvement of PPARγ and APC/β-catenin pathways. Chem Biol Interact 169, 110121.CrossRefGoogle ScholarPubMed
22Song, HJ, Sneddon, AA, Heys, SD & Wahle, KW (2006) Induction of apoptosis and inhibition of NF-κB activation in human prostate cancer cells by the cis-9, trans-11 but not the trans-10, cis-12 isomer of conjugated linoleic acid. Prostate 66, 839846.CrossRefGoogle Scholar
23Chung, S, Brown, JM, Provo, JN, Hopkins, R & McIntosh, MK (2005) Conjugated linoleic acid promotes human adipocyte insulin resistance through NF-κB dependent cytokine production. J Biol Chem 280, 3844538456.CrossRefGoogle ScholarPubMed
24Liu, LF, Purushotham, A, Wendel, AA & Belury, MA (2007) Combined effects of rosiglitazone and conjugated linoleic acid on adiposity, insulin sensitivity, and hepatic steatosis in high-fat-fed mice. Am J Physiol Liver Physiol 292, G1671G1682.Google ScholarPubMed
25Pérez-Matute, P, Marti, A, Martínez, JA, Fernández-Otero, MP, Stanhope, KL, Havel, PJ & Moreno-Aliaga, MJ (2007) Conjugated linoleic acid inhibits glucose metabolism, leptin and adiponectin-secretion in primary cultured rat adipocytes. Mol Cell Endocrinol 268, 5058.CrossRefGoogle ScholarPubMed
26Navarro, V, Fernandez-Quintela, A, Churruca, I & Portillo, MP (2006) The fat-lowering effect of conjugated linoleic acid: a comparison between animal and human studies. J Physiol Biochem 62, 137148.CrossRefGoogle ScholarPubMed
27Tricon, S & Yaqoob, P (2006) Conjugated linoleic acid and human health: a critical evaluation of the evidence. Curr Opin Nutr Metab Care 9, 105110.CrossRefGoogle Scholar
28Close, RN, Schoeller, DA, Watras, AC & Nora, EH (2007) Conjugated linoleic acid supplementation alters the 6-mo change in fat oxidation during sleep. Am J Clin Nutr 86, 797804.CrossRefGoogle ScholarPubMed
29Whigham, LD, Watras, AC & Schoeller, DA (2007) Efficacy of conjugated linoleic acid for reducing fat mass: a meta-analysis in humans. Am J Clin Nutr 85, 12031211.CrossRefGoogle Scholar
30Laude, AJ & Prior, IA (2004) Plasma membrane microdomains: organization, function and trafficking. Mol Membr Biol 21, 193205.CrossRefGoogle ScholarPubMed
31Hancock, JF (2006) Lipid rafts: contentious only from simplistic standpoints. Nat Rev Mol Cell Biol 7, 456462.CrossRefGoogle ScholarPubMed
32Anderson, RGW (1998) The caveolae membrane system. Annu Rev Biochem 67, 199225.CrossRefGoogle ScholarPubMed
33Simons, K & Ikonen, E (1997) Functional rafts in cell membranes. Nature 387, 569572.CrossRefGoogle ScholarPubMed
34Prior, IA, Muncke, C, Parton, RG & Hancock, JF (2003) Direct visualization of Ras proteins in spatially distinct cell surface microdomains. J Cell Biol 160, 165170.CrossRefGoogle ScholarPubMed
35Stillwell, W & Wassall, SR (2003) Docosahexaenoic acid: membrane properties of a unique fatty acid. Chem Phys Lipids 126, 127.CrossRefGoogle ScholarPubMed
36Fan, YY, Ly, LH, Barhoumi, R, McMurray, DN & Chapkin, RS (2004) Dietary docosahexaenoic acid suppresses T-cell protein kinase C-theta lipid raft recruitment and interleukin-2 production. J Immunol 173, 61516160.CrossRefGoogle Scholar
37Ma, DW, Seo, J, Davidson, LA, Callaway, ES, Fan, YY, Lupton, JR & Chapkin, RS (2004) n-3 PUFA alter caveolae lipid composition and resident protein localization in mouse colon. FASEB J 18, 10401042.CrossRefGoogle ScholarPubMed
38Zeyda, M, Säemann, MD, Stuhlmeier, KM, Mascher, DG, Nowotny, PN, Zlabinger, GJ, Waldhäusl, W & Stulnig, TM (2005) Polyunsaturated block dendritic cell activation and function independently of NF-κB activation. J Biol Chem 280, 1429314301.CrossRefGoogle ScholarPubMed
39Geyeregger, R, Zeyda, M, Zlabinger, GJ, Waldhäusl, W & Stulnig, TM (2005) Polyunsaturated fatty acids interfere with formation of the immunological synapse. J Leuk Biol 77, 680688.CrossRefGoogle ScholarPubMed
40Stulnig, TM, Berger, M, Sigmund, T, Raederstorff, D, Stockinner, H & Waldhäusl, W (1998) Polyunsaturated fatty acids inhibit T cell signal transduction by modification of detergent-insoluble membrane domains. J Cell Biol 143, 637644.CrossRefGoogle ScholarPubMed
41Stulnig, TM, Huber, J, Leitinger, N, Imre, E-M, Angelisová, P, Nowotny, P & Waldhäusl, W (2001) Polyunsaturated eicosapentaenoic acid displaces proteins from membrane rafts by altering raft lipid composition. J Biol Chem 276, 3733537340.CrossRefGoogle ScholarPubMed
42Fowler, KH, McMurray, DN, Fan, YY, Aukema, HM & Chapkin, RS (1993) Purified dietary n-3 polyunsaturated fatty acids alter diacylglycerol mass and molecular species composition in concanavalin A stimulated murine splenocytes. Biochim Biophys Acta 1210, 8996.CrossRefGoogle ScholarPubMed
43Switzer, KC, McMurray, DN, Morris, JS & Chapkin, RS (2003) Dietary n-3 polyunsaturated fatty acids selectively promote activation-induced cell death in T-lymphocytes. J Nutr 133, 496503.CrossRefGoogle ScholarPubMed
44Fan, YY, McMurray, DN, Ly, LH & Chapkin, RS (2003) Dietary (n-3) polyunsaturated fatty acids remodel mouse T-cell lipid rafts. J Nutr 133, 19131920.CrossRefGoogle ScholarPubMed
45Switzer, KC, Fan, YY, Wang, N, McMurray, DM & Chapkin, RS (2004) Dietary n-3 polyunsaturated fatty acids promote activation-induced cell death in the Th1-polarized murine CD4+T-cells. J Lipid Res 45, 14821492.CrossRefGoogle ScholarPubMed
46Leitenberg, D, Balamuth, F & Bottomly, K (2001) Changes in the T cell receptor macromolecular signaling complex and membrane microdomains during T cell development and activation. Sem Immunol 13, 129138.CrossRefGoogle Scholar
47Gaus, K, Chklovskaia, E, Fazekas de St Groth, B, Jessup, W & Harder, T (2005) Condensation of the plasma membrane at the site of T lymphocyte activation. J Cell Biol 171, 121131.CrossRefGoogle ScholarPubMed
48Harder, T, Rentero, C, Zech, T & Gaus, K (2007) Plasma membrane segregation during T cell activation: probing the order of domains. Curr Opin Immunol 19, 470475.CrossRefGoogle Scholar
49Shaikh, SR & Edidin, M (2007) Immunosuppressive effects of polyunsaturated fatty acids on antigen presentation by human leukocyte antigen class I molecules. J Lipid Res 48, 127138.CrossRefGoogle ScholarPubMed
50Huot, PS & Ma, WL (2007) CLA incorporates into caveolae phospholipids and reduces caveolin-1 expression in MCF-7 cells. Presented at the 98th American Oil Chemists' Society (AOCS) Annual Meeting, Quebec, Canada, May 2007, p. 81. Urbana, IL: AOCS Press..Google Scholar
51Yin, JJ, Kramer, JK, Yurawecz, MP, Eynard, AR, Mossoba, MM & Yu, L (2006) Effects of conjugated linoleic acid (CLA) isomers on oxygen diffusion-concentration products in liposomes and phospholipids solutions. J Agric Food Chem 54, 72877293.CrossRefGoogle ScholarPubMed
52Huster, D, Arnold, K & Gawrisch, K (1998) Influence of docosahexaenoic acid and cholesterol on lateral lipid organization in phospholipid mixtures. Biochemistry 37, 1729917308.CrossRefGoogle ScholarPubMed
53Mitchell, DC & Litman, BJ (1998) Effect of cholesterol on molecular order and dynamics in highly polyunsaturated phospholipid bilayers. Biophys J 75, 896908.CrossRefGoogle ScholarPubMed
54Pasenkiewicz-Gierula, M, Subczynski, WK & Kusumi, A (1991) Influence of phospholipid unsaturation on the cholesterol distribution in membranes. Biochimie 73, 13111316.CrossRefGoogle ScholarPubMed
55Niu, SL & Litman, BJ (2002) Determination of membrane cholesterol partition coefficient using a lipid vesicle-cyclodextrin binary system: effect of phospholipid acyl chain unsaturation and headgroup composition. Biophys J 83, 34083415.CrossRefGoogle ScholarPubMed
56Zerouga, M, Jenski, LJ & Stillwell, W (1995) Comparison of phosphatidylcholines containing one or two docosahexaenoic acyl chains on properties of phospholipid monolayers and bilayers. Biochim Biophys Acta 1236, 266272.CrossRefGoogle ScholarPubMed
57Kariel, N, Davidson, E & Keough, KM (1991) Cholesterol does not remove the gel-liquid crystalline phase transition of phosphatidylcholines containing two polyenoic acyl chains. Biochim Biophys Acta 1062, 7076.CrossRefGoogle Scholar
58Needham, D & Nunn, RS (1990) Elastic deformation and failure of lipid bilayer membranes containing cholesterol. Biophys J 58, 9971009.CrossRefGoogle ScholarPubMed
59Mitchell, DC & Litman, BJ (1998) Molecular order and dynamics in bilayers consisting of highly polyunsaturated phospholipids. Biophys J 74, 879891.CrossRefGoogle ScholarPubMed
60Brzustowicz, MR, Cherezov, V, Caffrey, M, Stillwell, W & Wassall, SR (2002) Molecular organization of cholesterol in polyunsaturated membranes: microdomain formation. Biophys J 82, 285298.CrossRefGoogle ScholarPubMed
61Brzustowicz, MR, Cherezov, V, Zerouga, M, Caffrey, M, Stillwell, W & Wassall, SR (2002) Controlling membrane cholesterol content. A role for polyunsaturated (docosahexaenoate) phospholipids. Biochemistry 41, 1250912519.CrossRefGoogle Scholar
62Pasenkiewicz-Gierula, M, Subczynski, WK & Kusumi, A (1990) Rotational diffusion of a steroid molecule in phosphatidylcholine-cholesterol membranes: fluid-phase microimmiscibility in unsaturated phosphatidylcholine-cholesterol membranes. Biochemistry 29, 40594069.CrossRefGoogle ScholarPubMed
63Huang, J & Feigenson, GW (1999) A microscopic interaction model of maximum solubility of cholesterol in lipid bilayers. Biophys J 76, 21422157.CrossRefGoogle ScholarPubMed
64Shaikh, SR, Cherezov, V, Caffrey, M, Stillwell, W & Wassall, SR (2003) Interaction of cholesterol with a docosahexaenoic acid-containing phosphatidylethanolamine: trigger for microdomain/raft formation? Biochemistry 42, 1202812037.CrossRefGoogle ScholarPubMed
65Danbara, N, Yuri, T, Tsujita-Kyutoku, M, Sato, M, Senzaki, H, Takada, H, Hada, T, Miyazawa, T, Okazaki, K & Tsubura, A (2004) 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 50, 7179.CrossRefGoogle ScholarPubMed
66Kimura, Y & Sumiyoshi, M (2005) Antitumor and antimetastatic actions of eicosapentaenoic acid ethylester and its by-products formed during accelerated stability testing. Cancer Sci 96, 441450.CrossRefGoogle ScholarPubMed
67Yonezawa, Y, Tsuzuki, T, Eitsuka, T, et al. (2005) Inhibitory effect of conjugated eicosapentaenoic acid on human DNA topoisomerases I and II. Arch Biochem Biophys 435, 197206.CrossRefGoogle ScholarPubMed
68Tsuzuki, T, Tanaka, K, Kuwahara, S & Miyazawa, T (2005) Synthesis of the conjugated trienes 5E,7E,9E,14Z,17Z-eiocospentaenoic acid and 5Z,7E,9E,14Z,17Z-eiocsapentaenoic acid and their induction of apoptosis in DLD-1 colorectal adenocarcinoma cells. Lipids 40, 147154.CrossRefGoogle Scholar
69Tsuzuki, T, Kambe, T, Shibata, A, Kawakami, Y, Nakagawa, K & Miyazawa, T (2007) Conjugated EPA activates mutant p53 via lipid peroxidation and induces p53-dependent apoptosis in DLD-1 colorectal adenocarcinoma human cells. Biochim Biophys Acta 1771, 2030.CrossRefGoogle ScholarPubMed
70Tsuzuki, T, Kawakami, Y, Nakagawa, K & Miyazawa, T (2006) Conjugated docosahexaenoic acid inhibits lipid accumulation in rats. J Nutr Biochem 17, 518524.CrossRefGoogle ScholarPubMed
71Tsuzuki, T, Shibata, A, Kawakami, Y, Nakagaya, K & Miyazawa, T (2007) Anti-angiogenic effects of conjugated docosahexaenoic acid in vitro and in vivo. Biosci Biotechnol Biochem 71, 19021910.CrossRefGoogle ScholarPubMed
Figure 0

Fig. 1 Nuclear receptor activation by conjugated linoleic acid (CLA). FABP, fatty acid-binding proteins (molecular chaperone); ER, endoplasmic reticulum; RXR, retinoid X receptors. CLA transactivates PPAR nuclear receptors. n-3 PUFA suppress NF-κB activation. All membranes incorporate EPA, DHA and conjugated PUFA to different degrees.

Figure 1

Fig. 2 Putative membrane microdomain-altering properties of n-3 PUFA and conjugated linoleic acid (CLA). Dietary DHA and CLA are incorporated into both the bulk phase of the plasma membrane as well as discrete heterogeneous cholesterol/sphingolipid-rich raft domains. This can alter plasma membrane organisation of inner leaflets and the dynamic partitioning of transduction proteins, thereby modulating their function.