Skip to main content
Erschienen in: Cardiovascular Diabetology 1/2008

Open Access 01.12.2008 | Hypothesis

Are fish oil omega-3 long-chain fatty acids and their derivatives peroxisome proliferator-activated receptor agonists?

verfasst von: Osman ABSM Gani

Erschienen in: Cardiovascular Diabetology | Ausgabe 1/2008

Abstract

Background

Peroxisome proliferator-activated receptors (PPARα, PPARγ, and PPARδ) are physiological sensors for glucose and lipid homeostasis. They are also the targets of synthetic drugs; such as fibrates as PPARα agonists which lower lipid level, and glitazones as PPARγ agonists which lower glucose level. As diabetes and metabolic diseases are often associated with high blood glucose and lipid levels, drugs that activate both PPARα/γ would be a logical approach. But synthetically developed PPARα/γ dual agonists and glitazones are showing side effects such as weight gain and edema. Therefore, natural compounds and their close derivatives are focused as future drugs against metabolic diseases.

Presentation of hypothesis

Docosahexaenoic acid and eicosapentaenoic acid, which are the fatty acids abundant in fish oil, are traditionally used against metabolic diseases. These fatty acids act as PPAR agonists that transcript the genes involved in glucose and lipid homeostasis. Present hypothesis suggests that the derivatives of these fatty acids are stronger PPAR agonists than the parent compounds. X-ray structures of PPARs indicate that α or β derivatives of fatty acids would fit into PPARα/γ binding cavity. Therefore, the derivatives will exhibit stronger affinities and activities than the parent compounds.

Testing of the hypothesis

Ligand binding assays and gene transactivation assays should be performed to test the hypothesis. Fluorescence-based methods are advantageous in binding assays, because they were found more suitable for fatty acid binding assays. In transactivation assays, care should be taken to remove contaminants from recombinant proteins.

Implications of the hypothesis

Present hypothesis is framed on the basis of molecular structure of natural PPAR agonists. Small structural changes in the molecular structure of fatty acids have a great influence on activating different PPARs. Therefore, this hypothesis bridges the concept of natural PPAR agonists and the use of structural information in designing new drugs against diabetes and metabolic syndrome. The derivatives may also be used as anti-inflammatory and anticancer agents.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1475-2840-7-6) contains supplementary material, which is available to authorized users.

Competing interests

The author declares no competing interests.
Abkürzungen
PPARs
Peroxisome proliferator-activated receptors
DHA
Docosahexaenoic acid
EPA
Eicosapentaenoic acid
LBD
Ligand binding domain

Background

Peroxisome proliferator-activated receptors (PPARs) are members of nuclear receptor superfamily. Three isotypes of PPARs (PPARα, PPARγ, and PPARδ) have distinct tissue distributions, distinct physiological roles, and distinct ligand specificities [1]. Activation of PPARα lowers lipid levels, PPARγ increases insulin sensitivity, and PPARδ regulates cholesterol and glucose levels [13]. PPAR ligands comprise natural or synthetic compounds, including fatty acids and eicosanoids [4, 5]. As synthetic compounds, fibrates are known as PPARα agonists and glitazones act as PPARγ agonists [2] (Fig. 1). PPARα/γ dual agonists have also been developed, because type 2 diabetes and metabolic syndrome are often associated with high lipid and glucose level in blood.
Antidiabetic glitazones exhibit side effects such as weight gain, edema, and increased risk of myocardial infarctions [6, 7], which have limited the use of these drugs in diabetic patients with high lipid levels. The PPARα/γ dual agonists, which reached clinical trials, have been suspended for safety issues [8]. Although much information is not available, these dual agonists are believed to exhibit similar side effects as with glitazones [2, 9]. Bezafibrate, a traditional PPARα agonist, has recently been identified as a safe and synthetic pan agonist for all PPAR isotypes [10, 11], although with relatively low potency and low affinity (Kd ~5 μM for PPARδ [5]).
Development of drugs from natural origin against chronic diseases, such as metabolic syndrome and diabetes, has gain focus recently. Because metabolic system can easily excrete biomolecules and their derivatives, thereby avoid undesirable effects. Fish oil, which contains docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA), is traditionally used as functional food against metabolic diseases. These beneficial health effects of DHA and EPA are thought to arise from their binding and activating PPARs [12, 13]. Phytanic acid, a natural PPAR agonist from human diet, has also been shown to enhance glucose uptake and thereby increases insulin sensitivity, however with less capacity to differentiate adipocytes [14]. A recent study has reported an extract of Fuligo candida, Fuligocandin B, to induce 15-deoxy-Δ12,14 prostaglandin J2 (15d-PGJ2), which is the most potent endogenous PPARγ ligand [15, 16].
The hypothesis of this article is drawn on the assumption of PPARα/γ dual agonists from naturally originated molecules, that the derivatives of DHA and EPA would show stronger affinities for PPARs than the parent compounds. Therefore, these compounds may be used as putative drugs against diabetes and metabolic syndrome.

Presentation of hypothesis

The hypothesis is based on (a) functional roles of PPARs – PPAR agonists reduce blood glucose and lipid levels, and (b) structural details of ligand binding cavity of PPARs, which suggest how a ligand fits into the cavity.

Functional roles of PPARs

PPARs perform their activities by endogenous ligands produced by metabolism of fatty acids. Unmetabolized fatty acids can also act as PPAR ligands. Activities of these ligands vary according to their binding specificities for different PPARs, and on distributions of these ligands in different organs [17]. Because of this diversity, not all endogenous PPAR ligands are characterized yet. However, PPAR activities can be summarized as lipid sensors by their complementary actions. PPARα is expressed mainly in the tissues with high capacity for fatty acid oxidations, e.g. liver, heart, skeletal muscle etc. On the other hand, PPARγ is expressed predominantly in the adipose tissue but is also expressed in immune and inflammatory cells, mucosa of the colon and placenta. If the fatty acid concentrations are increased, PPARα uptakes and oxidizes fatty acids and their metabolites [18], and PPARγ enhances storage of fatty acids in the adipose tissue [19]. These combined activities of PPARα and PPARγ cause increased utilization of glucose than fatty acids in the skeletal muscles, and cause enhanced insulin sensitivity (Fig. 2). Although PPARδ's role is not well defined, it is also implicated for fatty acid oxidation [20].

Structure of PPARs

Like other nuclear receptors, 3D structure PPARs consists of a DNA binding domain in the N-terminus and a ligand binding domain (LBD) in the C-terminus [21]. In canonical mechanism, ligand binding to PPARs causes conformational changes in the receptor, which release corepressor and recruit coactivator. Then the receptors form compulsory heterodimers with another nuclear receptor, named retinoid X-receptor (RXR), and the resulting complex finally bind to DNA of target genes [22] (Fig. 3).
To date, 37 X-ray crystallographically determined structures of PPAR-LBDs are deposited in the Protein Data Bank (PDB) in different apo- and holo forms. These structures shows that the LBD is composed of 13 helices and a small four-stranded β-sheet (Fig. 3B). The lower part of LBD contains a Y-shaped cavity with the volume of 1300–1400 Å3 for ligand binding. This cavity includes 34 amino acids; each arm of the cavity is ~12 Å in length. One of the arms is hydrophilic but the other two are hydrophobic. While the amino acids in the polar arm form H-bonds with the ligand polar atoms, those in the hydrophobic arms form non-specific interactions with hydrophobic part of the ligands. Across PPARs, ~80% amino acids in the cavity are conserved, and the overall size of the cavity is also similar [23]. However, the topology is different inside the cavities of different PPARs. These differences influence ligand specificities. Ligand binding stabilizes the LBD in the active conformation; as such coactivator and RXR can bind to the activated LBD [24].

Fatty acids and their derivatives as PPAR agonists

For centuries, consumption of fatty fish is considered to protect against metabolic diseases. During early 1970s, Danish physicians discovered that Greenland Eskimos consuming fatty fishes exhibited low incidence of heart diseases and arthritis despite high-fat diet. This finding suggested beneficial effects of DHA and EPA. Fish oil supplementation successfully passed clinical trials and is now effectively used for treating metabolic syndrome [25].
Usually, PPAR ligands have three essential parts for optimal binding: (a) polar head group, (b) linker region, and (c) hydrophobic tail. DHA and EPA have a carboxylic group which serves as the polar head group, and their long chains form the required linker and hydrophobic regions. In addition, X-ray structures of PPARs show some free spaces proximal to the polar arm of binding cavity [26], and substitutions at α or β positions on DHA and EPA would fit into these spaces. Specifically, hydrophobic substituents that complement the size of free spaces would be a choice. However, substituents may not fit into PPARδ because of its narrow polar regions [27]. Size and hydrophobic/hydrophilic ratio of substituents may influence the ratio of binding between PPARα and PPARγ [28]. A high PPARα/PPARγ ratio of affinities of putative ligands would be safer dual agonists, because the side effects are thought to arise from high PPARγ affinity [2].

Limitation of the hypothesis

EPA and DHA have many beneficial health effects which are not typical for PPAR ligands. For example, their capacity to reduce coronary heart disease, blood pressure, primary heart attack and rheumatoid arthritis [2931] are not observed with typical PPAR ligands. Also, various developmental problems including attention-deficit/hyperactivity disorder have been linked to biological deficiencies in polyunsaturated fatty acids [32]. But these fatty acids do not show many PPAR effects such as reversing insulin resistance [33, 34], which may occur due to dissociation between n-3 polyunsaturated fatty acid and lipid metabolism and insulin action in insulin resistant state [35]. Therefore, an interesting observation will be whether substitutions at α or β positions on these fatty acids impart such typical PPAR effects.

Testing of hypothesis

Ligand binding assays and gene transactivation assays would confirm the hypothesis. Ligand binding assays quantify the binding affinities such as Kd or IC50 values. Previously published experiments with fatty acids showed inconsistent binding affinities. Krey et al.[5] reported binding affinities in 5–10 μM range, while Lin et al[36] reported this range of 5–17 nM. The latter range corresponds to the intracellular free fatty acid concentrations which are in the range of 7–50 nM [37]. Until now, no data are available about the affinities of DHA, DHA derivatives, and EPA derivatives. Therefore, fluorescence-based methods as used by Lin et al. may provide correct binding affinities. In transactivation assays of PPARα/γ genes, precautions should be taken to remove unwanted fatty acids in the recombinant proteins [38]. In both ligand binding and transactivation assays, ligands with high ratio of PPARα/γ affinity and activity should be chosen, which mean that compounds showing less PPARγ affinities and activities would be ideal.

Implication of the hypothesis

The ligand binding cavity of PPARs is 3–4 times larger than the other nuclear receptors, indicating their capability to accommodate and bind variety of natural and synthetic lipophilic acids. Many previous studies have revealed the roles of natural PPAR agonists against specific diseases. For example, natural PPAR agonists such as 15d-PGJ2 are emerging as important regulators of immunity and inflammation [3941]. 15d-PGJ2 has also been implicated for antitumor activities [42]. PPARα mediates the anti-inflammatory actions of palmitoylethanolamide, the naturally occurring amide of palmitic acid and ethanolamine [43]. Synthetic PPARγ agonists glitazones have been reported and used in phase I-II human clinical trials as anticancer agents [44].
Present hypothesis is framed on the basis of using natural PPAR agonists that small structural changes in the molecular structure of fatty acids have a great influence on activating different PPARs [45]. Therefore, this hypothesis bridges the concept of natural PPAR agonists and the use of structural information in designing new drugs against diabetes and metabolic syndrome. The derivatives may also be used as anti-inflammatory and anticancer agents.

Acknowledgements

I thank Professor Ingebrigt Sylte for revising the manuscript critically.
This article is published under license to BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://​creativecommons.​org/​licenses/​by/​2.​0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Competing interests

The author declares no competing interests.
Anhänge

Authors’ original submitted files for images

Literatur
1.
Zurück zum Zitat Willson TM, Brown PJ, Sternbach DD, Henke BR: The PPARs: from orphan receptors to drug discovery. J Med Chem. 2000, 43 (4): 527-550. 10.1021/jm990554g.CrossRefPubMed Willson TM, Brown PJ, Sternbach DD, Henke BR: The PPARs: from orphan receptors to drug discovery. J Med Chem. 2000, 43 (4): 527-550. 10.1021/jm990554g.CrossRefPubMed
2.
Zurück zum Zitat Henke BR: Peroxisome proliferator-activated receptor alpha/gamma dual agonists for the treatment of type 2 diabetes. J Med Chem. 2004, 47 (17): 4118-4127. 10.1021/jm030631e.CrossRefPubMed Henke BR: Peroxisome proliferator-activated receptor alpha/gamma dual agonists for the treatment of type 2 diabetes. J Med Chem. 2004, 47 (17): 4118-4127. 10.1021/jm030631e.CrossRefPubMed
3.
Zurück zum Zitat Barish GD, Narkar VA, Evans RM: PPAR delta: a dagger in the heart of the metabolic syndrome. J Clin Invest. 2006, 116 (3): 590-597. 10.1172/JCI27955.PubMedCentralCrossRefPubMed Barish GD, Narkar VA, Evans RM: PPAR delta: a dagger in the heart of the metabolic syndrome. J Clin Invest. 2006, 116 (3): 590-597. 10.1172/JCI27955.PubMedCentralCrossRefPubMed
4.
Zurück zum Zitat Kliewer SA, Sundseth SS, Jones SA, Brown PJ, Wisely GB, Koble CS, Devchand P, Wahli W, Willson TM, Lenhard JM, Lehmann JM: Fatty acids and eicosanoids regulate gene expression through direct interactions with peroxisome proliferator-activated receptors alpha and gamma. Proc Natl Acad Sci U S A. 1997, 94 (9): 4318-4323. 10.1073/pnas.94.9.4318.PubMedCentralCrossRefPubMed Kliewer SA, Sundseth SS, Jones SA, Brown PJ, Wisely GB, Koble CS, Devchand P, Wahli W, Willson TM, Lenhard JM, Lehmann JM: Fatty acids and eicosanoids regulate gene expression through direct interactions with peroxisome proliferator-activated receptors alpha and gamma. Proc Natl Acad Sci U S A. 1997, 94 (9): 4318-4323. 10.1073/pnas.94.9.4318.PubMedCentralCrossRefPubMed
5.
Zurück zum Zitat Krey G, Braissant O, L'Horset F, Kalkhoven E, Perroud M, Parker MG, Wahli W: Fatty acids, eicosanoids, and hypolipidemic agents identified as ligands of peroxisome proliferator-activated receptors by coactivator-dependent receptor ligand assay. Mol Endocrinol. 1997, 11 (6): 779-791. 10.1210/me.11.6.779.CrossRefPubMed Krey G, Braissant O, L'Horset F, Kalkhoven E, Perroud M, Parker MG, Wahli W: Fatty acids, eicosanoids, and hypolipidemic agents identified as ligands of peroxisome proliferator-activated receptors by coactivator-dependent receptor ligand assay. Mol Endocrinol. 1997, 11 (6): 779-791. 10.1210/me.11.6.779.CrossRefPubMed
6.
Zurück zum Zitat Nesto RW, Bell D, Bonow RO, Fonseca V, Grundy SM, Horton ES, Le Winter M, Porte D, Semenkovich CF, Smith S, Young LH, Kahn R: Thiazolidinedione use, fluid retention, and congestive heart failure: a consensus statement from the American Heart Association and American Diabetes Association. October 7, 2003. Circulation. 2003, 108 (23): 2941-2948. 10.1161/01.CIR.0000103683.99399.7E.CrossRefPubMed Nesto RW, Bell D, Bonow RO, Fonseca V, Grundy SM, Horton ES, Le Winter M, Porte D, Semenkovich CF, Smith S, Young LH, Kahn R: Thiazolidinedione use, fluid retention, and congestive heart failure: a consensus statement from the American Heart Association and American Diabetes Association. October 7, 2003. Circulation. 2003, 108 (23): 2941-2948. 10.1161/01.CIR.0000103683.99399.7E.CrossRefPubMed
7.
Zurück zum Zitat Nissen SE, Wolski K: Effect of rosiglitazone on the risk of myocardial infarction and death from cardiovascular causes. N Engl J Med. 2007, 356 (24): 2457-2471. 10.1056/NEJMoa072761.CrossRefPubMed Nissen SE, Wolski K: Effect of rosiglitazone on the risk of myocardial infarction and death from cardiovascular causes. N Engl J Med. 2007, 356 (24): 2457-2471. 10.1056/NEJMoa072761.CrossRefPubMed
8.
Zurück zum Zitat Fievet C, Fruchart JC, Staels B: PPARalpha and PPARgamma dual agonists for the treatment of type 2 diabetes and the metabolic syndrome. Curr Opin Pharmacol. 2006, 6 (6): 606-614. 10.1016/j.coph.2006.06.009.CrossRefPubMed Fievet C, Fruchart JC, Staels B: PPARalpha and PPARgamma dual agonists for the treatment of type 2 diabetes and the metabolic syndrome. Curr Opin Pharmacol. 2006, 6 (6): 606-614. 10.1016/j.coph.2006.06.009.CrossRefPubMed
9.
Zurück zum Zitat Ferrara P, Gohlke H, Price DJ, Klebe G, Brooks CL: Assessing scoring functions for protein-ligand interactions. J Med Chem. 2004, 47 (12): 3032-3047. 10.1021/jm030489h.CrossRefPubMed Ferrara P, Gohlke H, Price DJ, Klebe G, Brooks CL: Assessing scoring functions for protein-ligand interactions. J Med Chem. 2004, 47 (12): 3032-3047. 10.1021/jm030489h.CrossRefPubMed
10.
Zurück zum Zitat Peters JM, Aoyama T, Burns AM, Gonzalez FJ: Bezafibrate is a dual ligand for PPARalpha and PPARbeta: studies using null mice. Biochim Biophys Acta. 2003, 1632 (1-3): 80-89.CrossRefPubMed Peters JM, Aoyama T, Burns AM, Gonzalez FJ: Bezafibrate is a dual ligand for PPARalpha and PPARbeta: studies using null mice. Biochim Biophys Acta. 2003, 1632 (1-3): 80-89.CrossRefPubMed
11.
Zurück zum Zitat Tenenbaum A, Motro M, Fisman EZ: Dual and pan-peroxisome proliferator-activated receptors (PPAR) co-agonism: the bezafibrate lessons. Cardiovasc Diabetol. 2005, 4: 14-10.1186/1475-2840-4-14.PubMedCentralCrossRefPubMed Tenenbaum A, Motro M, Fisman EZ: Dual and pan-peroxisome proliferator-activated receptors (PPAR) co-agonism: the bezafibrate lessons. Cardiovasc Diabetol. 2005, 4: 14-10.1186/1475-2840-4-14.PubMedCentralCrossRefPubMed
12.
Zurück zum Zitat Neschen S, Morino K, Dong J, Wang-Fischer Y, Cline GW, Romanelli AJ, Rossbacher JC, Moore IK, Regittnig W, Munoz DS, Kim JH, Shulman GI: n-3 Fatty acids preserve insulin sensitivity in vivo in a peroxisome proliferator-activated receptor-alpha-dependent manner. Diabetes. 2007, 56 (4): 1034-1041. 10.2337/db06-1206.CrossRefPubMed Neschen S, Morino K, Dong J, Wang-Fischer Y, Cline GW, Romanelli AJ, Rossbacher JC, Moore IK, Regittnig W, Munoz DS, Kim JH, Shulman GI: n-3 Fatty acids preserve insulin sensitivity in vivo in a peroxisome proliferator-activated receptor-alpha-dependent manner. Diabetes. 2007, 56 (4): 1034-1041. 10.2337/db06-1206.CrossRefPubMed
13.
Zurück zum Zitat von Schacky C: n-3 PUFA in CVD: influence of cytokine polymorphism. Proc Nutr Soc. 2007, 66 (2): 166-170. 10.1017/S0029665107005411.CrossRefPubMed von Schacky C: n-3 PUFA in CVD: influence of cytokine polymorphism. Proc Nutr Soc. 2007, 66 (2): 166-170. 10.1017/S0029665107005411.CrossRefPubMed
14.
Zurück zum Zitat Heim M, Johnson J, Boess F, Bendik I, Weber P, Hunziker W, Fluhmann B: Phytanic acid, a natural peroxisome proliferator-activated receptor (PPAR) agonist, regulates glucose metabolism in rat primary hepatocytes. FASEB J. 2002, 16 (7): 718-720.PubMed Heim M, Johnson J, Boess F, Bendik I, Weber P, Hunziker W, Fluhmann B: Phytanic acid, a natural peroxisome proliferator-activated receptor (PPAR) agonist, regulates glucose metabolism in rat primary hepatocytes. FASEB J. 2002, 16 (7): 718-720.PubMed
15.
Zurück zum Zitat Forman BM, Tontonoz P, Chen J, Brun RP, Spiegelman BM, Evans RM: 15-Deoxy-delta 12, 14-prostaglandin J2 is a ligand for the adipocyte determination factor PPAR gamma. Cell. 1995, 83 (5): 803-812. 10.1016/0092-8674(95)90193-0.CrossRefPubMed Forman BM, Tontonoz P, Chen J, Brun RP, Spiegelman BM, Evans RM: 15-Deoxy-delta 12, 14-prostaglandin J2 is a ligand for the adipocyte determination factor PPAR gamma. Cell. 1995, 83 (5): 803-812. 10.1016/0092-8674(95)90193-0.CrossRefPubMed
16.
Zurück zum Zitat Hasegawa H, Yamada Y, Komiyama K, Hayashi M, Ishibashi M, Sunazuka T, Izuhara T, Sugahara K, Tsuruda K, Masuda M, Takasu N, Tsukasaki K, Tomonaga M, Kamihira S: A novel natural compound, a cycloanthranilylproline derivative (Fuligocandin B), sensitizes leukemia cells to apoptosis induced by tumor necrosis factor related apoptosis-inducing ligand (TRAIL) through 15-deoxy-Delta 12, 14 prostaglandin J2 production. Blood. 2007, 110 (5): 1664-1674. 10.1182/blood-2007-01-068981.CrossRefPubMed Hasegawa H, Yamada Y, Komiyama K, Hayashi M, Ishibashi M, Sunazuka T, Izuhara T, Sugahara K, Tsuruda K, Masuda M, Takasu N, Tsukasaki K, Tomonaga M, Kamihira S: A novel natural compound, a cycloanthranilylproline derivative (Fuligocandin B), sensitizes leukemia cells to apoptosis induced by tumor necrosis factor related apoptosis-inducing ligand (TRAIL) through 15-deoxy-Delta 12, 14 prostaglandin J2 production. Blood. 2007, 110 (5): 1664-1674. 10.1182/blood-2007-01-068981.CrossRefPubMed
17.
Zurück zum Zitat Evans RM, Barish GD, Wang YX: PPARs and the complex journey to obesity. Nat Med. 2004, 10 (4): 355-361. 10.1038/nm1025.CrossRefPubMed Evans RM, Barish GD, Wang YX: PPARs and the complex journey to obesity. Nat Med. 2004, 10 (4): 355-361. 10.1038/nm1025.CrossRefPubMed
18.
Zurück zum Zitat Vu-Dac N, Schoonjans K, Kosykh V, Dallongeville J, Fruchart JC, Staels B, Auwerx J: Fibrates increase human apolipoprotein A-II expression through activation of the peroxisome proliferator-activated receptor. J Clin Invest. 1995, 96 (2): 741-750. 10.1172/JCI118118.PubMedCentralCrossRefPubMed Vu-Dac N, Schoonjans K, Kosykh V, Dallongeville J, Fruchart JC, Staels B, Auwerx J: Fibrates increase human apolipoprotein A-II expression through activation of the peroxisome proliferator-activated receptor. J Clin Invest. 1995, 96 (2): 741-750. 10.1172/JCI118118.PubMedCentralCrossRefPubMed
19.
Zurück zum Zitat Rosen ED, Walkey CJ, Puigserver P, Spiegelman BM: Transcriptional regulation of adipogenesis. Genes Dev. 2000, 14 (11): 1293-1307.PubMed Rosen ED, Walkey CJ, Puigserver P, Spiegelman BM: Transcriptional regulation of adipogenesis. Genes Dev. 2000, 14 (11): 1293-1307.PubMed
20.
Zurück zum Zitat Wang YX, Lee CH, Tiep S, Yu RT, Ham J, Kang H, Evans RM: Peroxisome-proliferator-activated receptor delta activates fat metabolism to prevent obesity. Cell. 2003, 113 (2): 159-170. 10.1016/S0092-8674(03)00269-1.CrossRefPubMed Wang YX, Lee CH, Tiep S, Yu RT, Ham J, Kang H, Evans RM: Peroxisome-proliferator-activated receptor delta activates fat metabolism to prevent obesity. Cell. 2003, 113 (2): 159-170. 10.1016/S0092-8674(03)00269-1.CrossRefPubMed
21.
Zurück zum Zitat Mangelsdorf DJ, Thummel C, Beato M, Herrlich P, Schutz G, Umesono K, Blumberg B, Kastner P, Mark M, Chambon P, Evans RM: The nuclear receptor superfamily: the second decade. Cell. 1995, 83 (6): 835-839. 10.1016/0092-8674(95)90199-X.CrossRefPubMed Mangelsdorf DJ, Thummel C, Beato M, Herrlich P, Schutz G, Umesono K, Blumberg B, Kastner P, Mark M, Chambon P, Evans RM: The nuclear receptor superfamily: the second decade. Cell. 1995, 83 (6): 835-839. 10.1016/0092-8674(95)90199-X.CrossRefPubMed
22.
Zurück zum Zitat Keller H, Dreyer C, Medin J, Mahfoudi A, Ozato K, Wahli W: Fatty acids and retinoids control lipid metabolism through activation of peroxisome proliferator-activated receptor-retinoid X receptor heterodimers. Proc Natl Acad Sci U S A. 1993, 90 (6): 2160-2164. 10.1073/pnas.90.6.2160.PubMedCentralCrossRefPubMed Keller H, Dreyer C, Medin J, Mahfoudi A, Ozato K, Wahli W: Fatty acids and retinoids control lipid metabolism through activation of peroxisome proliferator-activated receptor-retinoid X receptor heterodimers. Proc Natl Acad Sci U S A. 1993, 90 (6): 2160-2164. 10.1073/pnas.90.6.2160.PubMedCentralCrossRefPubMed
23.
Zurück zum Zitat Zoete V, Grosdidier A, Michielin O: Peroxisome proliferator-activated receptor structures: Ligand specificity, molecular switch and interactions with regulators. Biochim Biophys Acta. 2007 Zoete V, Grosdidier A, Michielin O: Peroxisome proliferator-activated receptor structures: Ligand specificity, molecular switch and interactions with regulators. Biochim Biophys Acta. 2007
24.
Zurück zum Zitat Glass CK, Rosenfeld MG: The coregulator exchange in transcriptional functions of nuclear receptors. Genes Dev. 2000, 14 (2): 121-141.PubMed Glass CK, Rosenfeld MG: The coregulator exchange in transcriptional functions of nuclear receptors. Genes Dev. 2000, 14 (2): 121-141.PubMed
25.
Zurück zum Zitat Connor WE: Importance of n-3 fatty acids in health and disease. Am J Clin Nutr. 2000, 71 (1 Suppl): 171S-5S.PubMed Connor WE: Importance of n-3 fatty acids in health and disease. Am J Clin Nutr. 2000, 71 (1 Suppl): 171S-5S.PubMed
26.
Zurück zum Zitat Sheu SH, Kaya T, Waxman DJ, Vajda S: Exploring the binding site structure of the PPAR gamma ligand-binding domain by computational solvent mapping. Biochemistry (Mosc). 2005, 44 (4): 1193-1209. 10.1021/bi048032c.CrossRef Sheu SH, Kaya T, Waxman DJ, Vajda S: Exploring the binding site structure of the PPAR gamma ligand-binding domain by computational solvent mapping. Biochemistry (Mosc). 2005, 44 (4): 1193-1209. 10.1021/bi048032c.CrossRef
27.
Zurück zum Zitat Nolte RT, Wisely GB, Westin S, Cobb JE, Lambert MH, Kurokawa R, Rosenfeld MG, Willson TM, Glass CK, Milburn MV: Ligand binding and co-activator assembly of the peroxisome proliferator-activated receptor-gamma. Nature. 1998, 395 (6698): 137-143. 10.1038/25931.CrossRefPubMed Nolte RT, Wisely GB, Westin S, Cobb JE, Lambert MH, Kurokawa R, Rosenfeld MG, Willson TM, Glass CK, Milburn MV: Ligand binding and co-activator assembly of the peroxisome proliferator-activated receptor-gamma. Nature. 1998, 395 (6698): 137-143. 10.1038/25931.CrossRefPubMed
28.
Zurück zum Zitat Cronet P, Petersen JF, Folmer R, Blomberg N, Sjoblom K, Karlsson U, Lindstedt EL, Bamberg K: Structure of the PPARalpha and -gamma ligand binding domain in complex with AZ 242; ligand selectivity and agonist activation in the PPAR family. Structure. 2001, 9 (8): 699-706. 10.1016/S0969-2126(01)00634-7.CrossRefPubMed Cronet P, Petersen JF, Folmer R, Blomberg N, Sjoblom K, Karlsson U, Lindstedt EL, Bamberg K: Structure of the PPARalpha and -gamma ligand binding domain in complex with AZ 242; ligand selectivity and agonist activation in the PPAR family. Structure. 2001, 9 (8): 699-706. 10.1016/S0969-2126(01)00634-7.CrossRefPubMed
29.
Zurück zum Zitat Bucher HC, Hengstler P, Schindler C, Meier G: N-3 polyunsaturated fatty acids in coronary heart disease: a meta-analysis of randomized controlled trials. Am J Med. 2002, 112 (4): 298-304. 10.1016/S0002-9343(01)01114-7.CrossRefPubMed Bucher HC, Hengstler P, Schindler C, Meier G: N-3 polyunsaturated fatty acids in coronary heart disease: a meta-analysis of randomized controlled trials. Am J Med. 2002, 112 (4): 298-304. 10.1016/S0002-9343(01)01114-7.CrossRefPubMed
30.
Zurück zum Zitat Fortin PR, Lew RA, Liang MH, Wright EA, Beckett LA, Chalmers TC, Sperling RI: Validation of a meta-analysis: the effects of fish oil in rheumatoid arthritis. J Clin Epidemiol. 1995, 48 (11): 1379-1390. 10.1016/0895-4356(95)00028-3.CrossRefPubMed Fortin PR, Lew RA, Liang MH, Wright EA, Beckett LA, Chalmers TC, Sperling RI: Validation of a meta-analysis: the effects of fish oil in rheumatoid arthritis. J Clin Epidemiol. 1995, 48 (11): 1379-1390. 10.1016/0895-4356(95)00028-3.CrossRefPubMed
31.
Zurück zum Zitat Morris MC, Sacks F, Rosner B: Does fish oil lower blood pressure? A meta-analysis of controlled trials. Circulation. 1993, 88 (2): 523-533.CrossRefPubMed Morris MC, Sacks F, Rosner B: Does fish oil lower blood pressure? A meta-analysis of controlled trials. Circulation. 1993, 88 (2): 523-533.CrossRefPubMed
32.
Zurück zum Zitat Sinn N, Bryan J: Effect of supplementation with polyunsaturated fatty acids and micronutrients on learning and behavior problems associated with child ADHD. J Dev Behav Pediatr. 2007, 28 (2): 82-91. 10.1097/01.DBP.0000267558.88457.a5.CrossRefPubMed Sinn N, Bryan J: Effect of supplementation with polyunsaturated fatty acids and micronutrients on learning and behavior problems associated with child ADHD. J Dev Behav Pediatr. 2007, 28 (2): 82-91. 10.1097/01.DBP.0000267558.88457.a5.CrossRefPubMed
33.
Zurück zum Zitat Manco M, Calvani M, Mingrone G: Effects of dietary fatty acids on insulin sensitivity and secretion. Diabetes Obes Metab. 2004, 6 (6): 402-413. 10.1111/j.1462-8902.2004.00356.x.CrossRefPubMed Manco M, Calvani M, Mingrone G: Effects of dietary fatty acids on insulin sensitivity and secretion. Diabetes Obes Metab. 2004, 6 (6): 402-413. 10.1111/j.1462-8902.2004.00356.x.CrossRefPubMed
34.
Zurück zum Zitat Mustad VA, Demichele S, Huang YS, Mika A, Lubbers N, Berthiaume N, Polakowski J, Zinker B: Differential effects of n-3 polyunsaturated fatty acids on metabolic control and vascular reactivity in the type 2 diabetic ob/ob mouse. Metabolism. 2006, 55 (10): 1365-1374. 10.1016/j.metabol.2006.06.007.CrossRefPubMed Mustad VA, Demichele S, Huang YS, Mika A, Lubbers N, Berthiaume N, Polakowski J, Zinker B: Differential effects of n-3 polyunsaturated fatty acids on metabolic control and vascular reactivity in the type 2 diabetic ob/ob mouse. Metabolism. 2006, 55 (10): 1365-1374. 10.1016/j.metabol.2006.06.007.CrossRefPubMed
35.
Zurück zum Zitat Podolin DA, Gayles EC, Wei Y, Thresher JS, Pagliassotti MJ: Menhaden oil prevents but does not reverse sucrose-induced insulin resistance in rats. Am J Physiol. 1998, 274 (3 Pt 2): R840-8.PubMed Podolin DA, Gayles EC, Wei Y, Thresher JS, Pagliassotti MJ: Menhaden oil prevents but does not reverse sucrose-induced insulin resistance in rats. Am J Physiol. 1998, 274 (3 Pt 2): R840-8.PubMed
36.
Zurück zum Zitat Lin Q, Ruuska SE, Shaw NS, Dong D, Noy N: Ligand selectivity of the peroxisome proliferator-activated receptor alpha. Biochemistry (Mosc). 1999, 38 (1): 185-190. 10.1021/bi9816094.CrossRef Lin Q, Ruuska SE, Shaw NS, Dong D, Noy N: Ligand selectivity of the peroxisome proliferator-activated receptor alpha. Biochemistry (Mosc). 1999, 38 (1): 185-190. 10.1021/bi9816094.CrossRef
37.
Zurück zum Zitat Hamilton JA: Fatty acid transport: difficult or easy?. J Lipid Res. 1998, 39 (3): 467-481.PubMed Hamilton JA: Fatty acid transport: difficult or easy?. J Lipid Res. 1998, 39 (3): 467-481.PubMed
38.
Zurück zum Zitat Fyffe SA, Alphey MS, Buetow L, Smith TK, Ferguson MA, Sorensen MD, Bjorkling F, Hunter WN: Recombinant human PPAR-beta/delta ligand-binding domain is locked in an activated conformation by endogenous fatty acids. J Mol Biol. 2006, 356 (4): 1005-1013. 10.1016/j.jmb.2005.12.047.CrossRefPubMed Fyffe SA, Alphey MS, Buetow L, Smith TK, Ferguson MA, Sorensen MD, Bjorkling F, Hunter WN: Recombinant human PPAR-beta/delta ligand-binding domain is locked in an activated conformation by endogenous fatty acids. J Mol Biol. 2006, 356 (4): 1005-1013. 10.1016/j.jmb.2005.12.047.CrossRefPubMed
39.
Zurück zum Zitat Jiang C, Ting AT, Seed B: PPAR-gamma agonists inhibit production of monocyte inflammatory cytokines. Nature. 1998, 391 (6662): 82-86. 10.1038/34184.CrossRefPubMed Jiang C, Ting AT, Seed B: PPAR-gamma agonists inhibit production of monocyte inflammatory cytokines. Nature. 1998, 391 (6662): 82-86. 10.1038/34184.CrossRefPubMed
40.
Zurück zum Zitat Ray DM, Akbiyik F, Bernstein SH, Phipps RP: CD40 engagement prevents peroxisome proliferator-activated receptor gamma agonist-induced apoptosis of B lymphocytes and B lymphoma cells by an NF-kappaB-dependent mechanism. J Immunol. 2005, 174 (7): 4060-4069.CrossRefPubMed Ray DM, Akbiyik F, Bernstein SH, Phipps RP: CD40 engagement prevents peroxisome proliferator-activated receptor gamma agonist-induced apoptosis of B lymphocytes and B lymphoma cells by an NF-kappaB-dependent mechanism. J Immunol. 2005, 174 (7): 4060-4069.CrossRefPubMed
41.
Zurück zum Zitat Ricote M, Li AC, Willson TM, Kelly CJ, Glass CK: The peroxisome proliferator-activated receptor-gamma is a negative regulator of macrophage activation. Nature. 1998, 391 (6662): 79-82. 10.1038/34178.CrossRefPubMed Ricote M, Li AC, Willson TM, Kelly CJ, Glass CK: The peroxisome proliferator-activated receptor-gamma is a negative regulator of macrophage activation. Nature. 1998, 391 (6662): 79-82. 10.1038/34178.CrossRefPubMed
42.
Zurück zum Zitat Piva R, Gianferretti P, Ciucci A, Taulli R, Belardo G, Santoro MG: 15-Deoxy-delta 12,14-prostaglandin J2 induces apoptosis in human malignant B cells: an effect associated with inhibition of NF-kappa B activity and down-regulation of antiapoptotic proteins. Blood. 2005, 105 (4): 1750-1758. 10.1182/blood-2004-04-1360.CrossRefPubMed Piva R, Gianferretti P, Ciucci A, Taulli R, Belardo G, Santoro MG: 15-Deoxy-delta 12,14-prostaglandin J2 induces apoptosis in human malignant B cells: an effect associated with inhibition of NF-kappa B activity and down-regulation of antiapoptotic proteins. Blood. 2005, 105 (4): 1750-1758. 10.1182/blood-2004-04-1360.CrossRefPubMed
43.
Zurück zum Zitat Lo Verme J, Fu J, Astarita G, La Rana G, Russo R, Calignano A, Piomelli D: The nuclear receptor peroxisome proliferator-activated receptor-alpha mediates the anti-inflammatory actions of palmitoylethanolamide. Mol Pharmacol. 2005, 67 (1): 15-19. 10.1124/mol.104.006353.CrossRefPubMed Lo Verme J, Fu J, Astarita G, La Rana G, Russo R, Calignano A, Piomelli D: The nuclear receptor peroxisome proliferator-activated receptor-alpha mediates the anti-inflammatory actions of palmitoylethanolamide. Mol Pharmacol. 2005, 67 (1): 15-19. 10.1124/mol.104.006353.CrossRefPubMed
44.
Zurück zum Zitat Grommes C, Landreth GE, Heneka MT: Antineoplastic effects of peroxisome proliferator-activated receptor gamma agonists. Lancet Oncol. 2004, 5 (7): 419-429. 10.1016/S1470-2045(04)01509-8.CrossRefPubMed Grommes C, Landreth GE, Heneka MT: Antineoplastic effects of peroxisome proliferator-activated receptor gamma agonists. Lancet Oncol. 2004, 5 (7): 419-429. 10.1016/S1470-2045(04)01509-8.CrossRefPubMed
45.
Zurück zum Zitat Granlund L, Larsen LN, Nebb HI, Pedersen JI: Effects of structural changes of fatty acids on lipid accumulation in adipocytes and primary hepatocytes. Biochim Biophys Acta. 2005, 1687 (1-3): 23-30.CrossRefPubMed Granlund L, Larsen LN, Nebb HI, Pedersen JI: Effects of structural changes of fatty acids on lipid accumulation in adipocytes and primary hepatocytes. Biochim Biophys Acta. 2005, 1687 (1-3): 23-30.CrossRefPubMed
Metadaten
Titel
Are fish oil omega-3 long-chain fatty acids and their derivatives peroxisome proliferator-activated receptor agonists?
verfasst von
Osman ABSM Gani
Publikationsdatum
01.12.2008
Verlag
BioMed Central
Erschienen in
Cardiovascular Diabetology / Ausgabe 1/2008
Elektronische ISSN: 1475-2840
DOI
https://doi.org/10.1186/1475-2840-7-6

Weitere Artikel der Ausgabe 1/2008

Cardiovascular Diabetology 1/2008 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Erhebliches Risiko für Kehlkopfkrebs bei mäßiger Dysplasie

29.05.2024 Larynxkarzinom Nachrichten

Fast ein Viertel der Personen mit mäßig dysplastischen Stimmlippenläsionen entwickelt einen Kehlkopftumor. Solche Personen benötigen daher eine besonders enge ärztliche Überwachung.

Nach Herzinfarkt mit Typ-1-Diabetes schlechtere Karten als mit Typ 2?

29.05.2024 Herzinfarkt Nachrichten

Bei Menschen mit Typ-2-Diabetes sind die Chancen, einen Myokardinfarkt zu überleben, in den letzten 15 Jahren deutlich gestiegen – nicht jedoch bei Betroffenen mit Typ 1.

15% bedauern gewählte Blasenkrebs-Therapie

29.05.2024 Urothelkarzinom Nachrichten

Ob Patienten und Patientinnen mit neu diagnostiziertem Blasenkrebs ein Jahr später Bedauern über die Therapieentscheidung empfinden, wird einer Studie aus England zufolge von der Radikalität und dem Erfolg des Eingriffs beeinflusst.

Costims – das nächste heiße Ding in der Krebstherapie?

28.05.2024 Onkologische Immuntherapie Nachrichten

„Kalte“ Tumoren werden heiß – CD28-kostimulatorische Antikörper sollen dies ermöglichen. Am besten könnten diese in Kombination mit BiTEs und Checkpointhemmern wirken. Erste klinische Studien laufen bereits.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.