Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2011 Sep 1.
Published in final edited form as: Curr Opin Clin Nutr Metab Care. 2010 Sep;13(5):569–573. doi: 10.1097/MCO.0b013e32833b648e

Dietary CLA and n-3 PUFA in inflammatory bowel disease

Josep Bassaganya-Riera 1, Raquel Hontecillas 1
PMCID: PMC2947030  NIHMSID: NIHMS237437  PMID: 20508519

Abstract

Purpose of review

Inflammatory bowel disease (IBD) is a debilitating and widespread immune-mediated illness of unknown etiology. Current treatments are modestly successful and with significant side effects. The purpose of this review is to summarize the current understanding of mechanisms of action underlying the anti-inflammatory actions of CLA and n-3 PUFA in IBD.

Recent findings

Nutrition-based interventions that target peroxisome proliferator-activated receptors (PPARs) such as dietary conjugated linoleic acid (CLA) or n-3 PUFA have demonstrated anti-inflammatory efficacy in animal models of IBD. Clinical data on n-3 PUFA in IBD remains generally unimpressive, although results of a recent human study demonstrate that IBD remission can be maintained by maintaining the n-3/n-6 ratio over 0.65 via n-3 PUFA intervention. In mice, CLA prevented inflammation-driven colorectal cancer by activating PPAR γ and modulating regulatory T cells and macrophages. CLA is the subject of an ongoing clinical study in Crohn’s disease patients.

Summary

Compelling evidence demonstrates that n-3 PUFA and CLA prevent or ameliorate IBD in animal models. However this basic knowledge has not been translated into novel nutrition-based clinical interventions. For both compounds there is an urgent need for placebo-controlled, large-scale, multi-center clinical trials.

Keywords: conjugated linoleic acid, omega-3 PUFA, immune regulation, PPARs, inflammatory bowel disease

Introduction

Inflammatory bowel disease (IBD) is a chronic, recurring immunoinflammatory illness afflicting over 1.4 million people in the United States and 0.1–24.5/100,000 people worldwide with two clinical manifestations -- Crohn’s disease (CD) and ulcerative colitis (UC) (1, 2). Even though IBD therapies have improved (3, 4), they are modestly successful for the long-term management of the disease and result in significant side effects, including immune suppression (5). The incidence of complementary and alternative medicine use was 49.5% for IBD patients (6). Hence, developing nutritional interventions against IBD such as CLA and n-3 PUFA remains important.

Peroxisome proliferator-activated receptors: molecular targets for dietary lipid-based intervention

A promising avenue for developing such immunonutrition-based treatments for IBD is by targeting peroxisome proliferator-activated receptors (PPARs). PPARs are nuclear receptors for endogenous lipid molecules [i.e., prostaglandins or hydroxy-containing PUFA such as 12/15-hydroxyeicosatetraenoic (HETE), 13-hydroxyoctadecadienoic (HODE)] and molecular targets for drugs against type II diabetes (79). They belong to the superfamily of nuclear hormone receptors with 48 members identified in the human genome. There are three known PPAR isoforms; α, β or δ, and γ, which differ in their tissue distribution and functional activity (10). PPAR activation and expression is controlled by a diverse set of natural and synthetic molecules, including nutrients, non-nutrient endogenous ligands and drugs [i.e., thiazolidinediones (TZDs) and fibrates]. In this regard, 5-ASA, a drug broadly described as an anti-inflammatory treatment for IBD, is a PPAR γ agonist (11). Some of the ligands are pan-PPAR agonists, whereas others are either isoform-specific or target two PPAR isoforms (10). PPARs have been studied primarily as drug targets. However, their main biological function is the sensing of intracellular nutrient concentrations –i.e., polyunsaturated fatty acids (PUFA), triglycerides, free fatty acids and lipoproteins- and regulation of gene expression involved in maintaining both metabolic and tissue homeostasis. Of note, results of a recent clinical study in UC patients demonstrate that rosiglitazone (Avandia™), a TZD agonist of PPAR γ and a U.S. Food and Drug Administration (FDA)-approved drug for treating type II diabetes, is also efficacious in the treatment of mild to moderately active UC (12). In spite of its efficacy, rosiglitazone is unlikely to be adopted for treating IBD due to its significant side effects (9, 13) and a FDA-issued ‘Black box’ warning. Thus, there is a need to discover novel naturally occurring agonists of PPARs that exert therapeutic and prophylactic actions against IBD with limited or no adverse side effects. There is also a need to elucidate their mechanisms of action since the limited understanding about the mechanisms of action has halted further progress in the development of nutrition-based approaches for preventing IBD.

Mechanisms of Action of Conjugated Linoleic Acid (CLA) in IBD

CLA is a mixture of positional and geometric isomers of octadecadienoic acid (predominantly at position 9 and 11, or 10 and 12) appear in a conjugated double bond system (two double bonds separated by a single bond). Several CLA isomers, including cis-9, trans-11 CLA, are naturally found in milk, cheese and ruminant products (14). In 2002, we found that dietary CLA-supplementation suppresses colonic inflammation and upregulates colonic PPAR γ expression in pigs with bacterial-induced colitis (15). Since that original report we have used mice with a targeted deletion of PPAR γ in immune and epithelial cells to demonstrate that activation of colonic PPAR γ by CLA mediates protection from experimental IBD in mice (16). In addition, n-3 PUFA appear to antagonize the effects of CLA on PPAR γ in a pig model of dextran sodium sulfate (DSS) colitis (17). Several reports by others confirm increases of PPAR γ expression and activity in adipocytes (18), skeletal muscle (19), colonic mucosa (20) and macrophages (21, 22) with CLA treatment or CLA-rich diets. However, a reduction in PPAR γ expression in adipocytes by CLA has also been reported (23, 24). This may be due to cell-type specificity of the response to CLA or isomer specificity since the t10, c12 was able to reduce PPAR γ expression in adipocytes but the c9, t11 failed to show the same suppressive effect on PPAR γ activity (25). In RAW 264.7 mouse macrophage (RAW) cells, CLA isomers decreased the expression of COX2, TNF-α, nitric oxide, IL-1β and IL-6 mRNA via PPAR γ (21). In HT-29 and Caco-2 intestinal epithelial cell lines the generation of CLA by probiotic bacteria induced apoptosis via an increase in PPAR γ expression (26). Thus, the protective effect of CLA against IBD appears to be mediated though PPAR γ activation. However, since CLA can also modulate the production of arachidonic acid metabolites (27, 28), it is also likely that the reduced production of inflammatory lipid mediators may contribute to CLA’s beneficial actions in IBD.

Evans and colleagues recently demonstrated that dietary CLA-supplementation ameliorated inflammation-driven colorectal cancer in mice (29). CLA also increased the percentages of regulatory CD4+ T cells (Treg) in mesenteric lymph nodes (MLN) of PPAR γ-expressing, but not immune cell-specific PPAR γ null mice, suggesting that CLA modulates the Treg compartment by activating PPAR γ. The results of this project are in line with the findings of previous IBD studies showing that this effect of CLA was mediated, in part, through a PPAR γ-dependent mechanism (16) as well as reports demonstrating that PPAR γ is required for Treg function (30, 31).

Unlike TZDs, CLA has been shown to be safe (32). Accordingly, the FDA granted CLA generally regarded as safe (GRAS) status (GRN 00232) in July 11, 2008. In contrast to corticosteroids, CLA suppresses gut inflammatory responses while enhancing antigen-specific responsiveness of T cells against viral and bacterial pathogens (3336). Open label clinical studies are currently underway to determine the anti-inflammatory efficacy of CLA in human patients with IBD. As a follow up of these ongoing studies, placebo-controlled, large-scale, multi-center studies should investigate the interplay between dietary CLA-supplementation, mucosal immunity and the gut microbiota.

N-3 PUFA in IBD Prevention

N-3 PUFA [i.e., linolenic acid (LN), docosahexaenoic (DHA) and eicosapentaenoic (EPA)] elicit potent anti-inflammatory and immunoregulatory properties either directly (37, 38) or following transcellular processing that results in the generation of hydroxy-containing n-3 PUFA metabolites (39). In similar action to dietary CLA, n-3 PUFA have been reported to ameliorate intestinal inflammation in animal models of IBD (40). However, previous IBD pre-clinical studies examined the efficacy of doses over 4 g of n-3 PUFA per 100 g diet, which would be unattainable in a human clinical setting. Thus, while the pre-clinical story in animal models is strong, the translational value of the doses of n-3 PUFA utilized is questionable.

In this regard, in 2005 MacLean and colleagues (41) reviewed 13 controlled trials that investigated the effects of n-3 PUFA on clinical, sigmoidoscopic or histologic scores. Only 3 studies found that n-3 PUFA administration decreased the corticosteroid requirement. Based on these findings, it was concluded that the available data are insufficient to recommend n-3 PUFA for the management of IBD in humans (e.g., GRAS Notice No. GRN 000105).

In 2006 we used a pig model of experimental IBD to examine the clinical activity of two fatty acid mixtures (i.e., n-3 PUFA and CLA) that have been proposed to prevent or ameliorate IBD (17). Our data demonstrate that during a 7-day challenge with DSS, n-3 PUFA failed to ameliorate clinical disease but favored a faster remission. Mechanistically, the effects of n-3 PUFA on epithelial regeneration appeared to be dependent on activation of PPAR δ (17).

A more recent study in human patients with CD and UC published by Uchiyama and colleagues published in 2010 (42) suggests a relationship between the accumulation of n-3 PUFA in the cell membranes and remission from IBD. Interestingly, in this study 2,250–3000 mg/day 5-ASA was administered to all patients. Since 5-ASA is a known PPAR γ agonist, it is possible that 5-ASA may have interacted with n-3 PUFA to favor remission.

Brix et al provided evidence demonstrating that CD4+ T cell activation is differentially modulated by bacteria-primed dendritic cells, but is down-regulated by n-3 PUFA (43). However, the underlying mechanisms mediating this effect have not been fully characterized. Since PPARs can modulate the function of effector and regulatory T cells (30, 31, 44) and given the established role of PUFA as PPAR agonists (45), it is puzzling that PPAR-dependent mechanisms were not investigated in this study. Nonetheless, the ability of n-3 PUFA to modulate immune responses to bacterial antigens is novel and it should be further explored. It is also important to investigate the direct effect of nutritional compounds on gut microbial populations.

Diet-Microbiome Interactions in IBD

The pathogenesis of IBD involves an interaction among the genetic predisposition of the individual, the environment and the gut microbiota. Understanding how n-3 PUFA and CLA favorably modulate the interface between diet, mucosal immunity and gut microbiota is of crucial importance for gaining useful insights regarding both clinical efficacy and mechanism of action. This will require a comprehensive assessment of the effects of the compounds on mucosal immune responses and gut microbial populations and host-bacterial interaction networks.

Sequence-based metagenomics technology has enabled the study of uncultured bacteria and archea by culture-independent identification and characterization of microorganisms using direct DNA sequencing. Recent advances in sequencing technology have made large characterization of complete metagenomic sequencing more accessible (4648). For instance, pyrosequencing has been applied to study characterization of microbial biodiversity and/or relative abundance of proteins specific to ecological niches such as seas (49), drainage (50) and waste water (51), soil and carcasses (52), worms (53), human skin (54) and gut (55). Analysis of 16S rRNA genes provides insight into the phylogenetic structure, composition and community dynamics of microbiota in their host environments. These new technologies will facilitate high-throughput and comprehensive analyses of the effect of dietary compounds in the gut microbiota.

Progress is underway in investigating the relationship between obesity and gut microbiota. For instance, at higher taxonomic classification two bacterial phyla that dominate this ecosystem belong to Firmicutes (Gram-positive bacteria) and Bacteroidetes (Gram-negative bacteria) (56) with Firmicutes dominating the guts of the obese individuals. Recent studies have also shown that diet and obesity influence the composition of gut microbial populations (5761). Thus, it is novel and important to examine how dietary components and IBD interact to modulate resident gut microbial populations. The gut microbiota may be manipulated by dietary components such as n-3 PUFA and CLA to favorably influence inflammatory disease risk by reducing endotoxin load via shifts in the composition and metabolic activity of the microbial community. There is some evidence suggesting that probiotic bacteria (i.e., VSL3) can produce CLA (26). However, very little is known about the extent to which the gut microbial environment mediates the associations between specific dietary components and IBD. Given that Bacteroidetes are the main source of lipopolysaccharide (LPS) and the established role of LPS in mediating low-grade chronic inflammation, the characterization of gut microbial communities during IBD as well as the modulation by dietary CLA, n-3 PUFA and other PPAR γ and δ agonists such as abscisic acid (ABA) (62) will provide novel and important insights on how to suppress intestinal inflammation in CD and UC patients.

Modeling Nutritional Interventions in IBD

Since PPAR γ and other molecular targets for IBD therapeutics are ubiquitously expressed in the gut, tracing clinical improvements from nutritional interventions back to concrete PPAR γ-initiated immunological mechanisms has proven extremely challenging. PPAR γ activity delineates the susceptibility to intestinal inflammation ranging from highly pro-inflammatory (low expression or activation) to anti-inflammatory (high expression or activation) states. In addition to testing the efficacy of such compounds in animal models and in patients with IBD, there is a need to build predictive, multiscale models of the intestine for understanding how important therapeutic targets modulate the immune response dynamics, gut pathology and anti-inflammatory responses.

Highly relevant to the discovery of novel PPAR γ-based therapeutic and prophylactic strategies for IBD is the identification of the most critical immunoregulatory mechanisms of the mucosal immune system that prevent inflammation and the subsequent destruction of the gut mucosa. For instance, computational/mathematical models of the gut will facilitate performing in silico deletions of putative regulatory mechanisms (i.e., regulatory T cells, M2 macrophages, tolerogenic dendritic cells) during an inflammatory challenge and investigate the consequences on indicators of enteric pathology such as epithelial erosion, immune cell infiltration or increased concentrations of pro-inflammatory cytokines in the mucosa. We constructed for the first time a model with an initial level of granularity at the cellular (immune and epithelial cells), with multiple tissues and compartments such as lumen, colonic lamina propria (LP) and MLN (63). We next asked which immune cell subsets would be more greatly associated with the recovery from disease. The model predicted that allowing the reversion rate from M1 macrophages to M2 macrophages facilitates recovery from colitis, indicating that M2 macrophages suppress gut inflammation. Validation of this prediction in a mouse model of experimental IBD demonstrated that pharmacologically inducing differentiation of macrophages into an M2 phenotype (64) by activating PPAR γ with pioglitazone, a TZD, facilitated recovery from colonic inflammation, as measured by a disease activity index. Moreover, the deletion of PPAR γ in macrophages (but not in other immune or epithelial cells) in Lysozyme M-Cre+ mice abrogated the ability of PIA to induce recovery from colitis. Further studies are required to track clinical and pathological improvements caused by naturally occurring PPAR γ agonists back to specific cellular and molecular interactions within the mucosal immune system network.

Conclusions

Insufficient progress has been made in translating the basic knowledge on the prevention of IBD by n-3 PUFA and CLA into actionable information for implementing nutrition-based interventions in the clinic. There is an urgent need for data from placebo-controlled, large-scale, multi-center clinical trials designed to perform a comprehensive assessment of the effects of such interventions on mucosal immune responses and gut microbial populations. A better mechanistic understanding in combination with more comprehensive data from well-designed clinical studies will lay the groundwork for accelerating the development of safer and more efficacious nutrition-based therapies against IBD.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Lakatos PL. Recent trends in the epidemiology of inflammatory bowel diseases: up or down? World J Gastroenterol. 2006;12:6102–6108. doi: 10.3748/wjg.v12.i38.6102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.CDC. Epidemiology of the inflammatory bowel disease [Google Scholar]
  • 3.Camilleri M. GI clinical research 2002–2003: The year in review. Clinical Gastroenterology and Hepatology. 2003;1:415–420. doi: 10.1016/s1542-3565(03)00220-9. [DOI] [PubMed] [Google Scholar]
  • 4.Lichtenstein GR, Abreu M, Present D. Recent advances in the treatment of Crohn's colitis. The center for health care education, LLC; 2003. [Google Scholar]
  • 5.Maconi G, Colombo E, Zerbi P, Sampietro GM, Fociani P, Bosani M, Cassinotti A, Casini V, Russo A, Ardizzone S, Porta M, Bianchi Porro G. Prevalence, detection rate and outcome of cytomegalovirus infection in ulcerative colitis patients requiring colonic resection. Dig Liver Dis. 2005;37:418–423. doi: 10.1016/j.dld.2005.01.011. [DOI] [PubMed] [Google Scholar]
  • 6.Kong SC, Hurlstone DP, Pocock CY, Walkington LA, Farquharson NR, Bramble MG, McAlindon ME, Sanders DS. The Incidence of self-prescribed oral complementary and alternative medicine use by patients with gastrointestinal diseases. J Clin Gastroenterol. 2005;39:138–141. [PubMed] [Google Scholar]
  • 7.Mokdad AH, Bowman BA, Ford ES, Vinicor F, Marks JS, Koplan JP. The continuing epidemics of obesity and diabetes in the United States. Jama. 2001;286:1195–1200. doi: 10.1001/jama.286.10.1195. [DOI] [PubMed] [Google Scholar]
  • 8.Desvergne B, Wahli W. Peroxisome proliferator-activated receptors: nuclear control of metabolism. Endocr Rev. 1999;20:649–688. doi: 10.1210/edrv.20.5.0380. [DOI] [PubMed] [Google Scholar]
  • 9.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. Circulation. 2003 October 7;108:2941–2948. doi: 10.1161/01.CIR.0000103683.99399.7E. 2003. [DOI] [PubMed] [Google Scholar]
  • 10.Mangelsdorf DJ, Thummel C, Beato M, Herrlich P, Schutz G, Umesono K, Blumberg B, Kastner P, Mark M, Chambon P, et al. The nuclear receptor superfamily: the second decade. Cell. 1995;83:835–839. doi: 10.1016/0092-8674(95)90199-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Rousseaux C, Lefebvre B, Dubuquoy L, Lefebvre P, Romano O, Auwerx J, Metzger D, Wahli W, Desvergne B, Naccari GC, Chavatte P, Farce A, Bulois P, Cortot A, Colombel JF, Desreumaux P. Intestinal antiinflammatory effect of 5-aminosalicylic acid is dependent on peroxisome proliferator-activated receptor-gamma. J Exp Med. 2005;201:1205–1215. doi: 10.1084/jem.20041948. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Lewis JD, Lichtenstein GR, Deren JJ, Sands BE, Hanauer SB, Katz JA, Lashner B, Present DH, Chuai S, Ellenberg JH, Nessel L, Wu GD. Rosiglitazone for active ulcerative colitis: a randomized placebo-controlled trial. Gastroenterology. 2008;134:688–695. doi: 10.1053/j.gastro.2007.12.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Marcy TR, Britton ML, Blevins SM. Second-generation thiazolidinediones and hepatotoxicity. Ann Pharmacother. 2004;38:1419–1423. doi: 10.1345/aph.1E072. [DOI] [PubMed] [Google Scholar]
  • 14.Bassaganya-Riera J, Hontecillas R, Beitz DC. Colonic anti-inflammatory mechanisms of conjugated linoleic acid. Clin Nutr. 2002;21:451–459. doi: 10.1054/clnu.2002.0594. [DOI] [PubMed] [Google Scholar]
  • 15.Hontecillas R, Wannemeulher MJ, Zimmerman DR, Hutto DL, Wilson JH, Ahn DU, Bassaganya-Riera J. Nutritional regulation of porcine bacterial-induced colitis by conjugated linoleic acid. J Nutr. 2002;132:2019–2027. doi: 10.1093/jn/132.7.2019. [DOI] [PubMed] [Google Scholar]
  • 16.Bassaganya-Riera J, Reynolds K, Martino-Catt S, Cui Y, Hennighausen L, Gonzalez F, Rohrer J, Benninghoff AU, Hontecillas R. Activation of PPAR gamma and delta by conjugated linoleic acid mediates protection from experimental inflammatory bowel disease. Gastroenterology. 2004;127:777–791. doi: 10.1053/j.gastro.2004.06.049. [DOI] [PubMed] [Google Scholar]
  • 17.Bassaganya-Riera J, Hontecillas R. CLA and n-3 PUFA differentially modulate clinical activity and colonic PPAR-responsive gene expression in a pig model of experimental IBD. Clin Nutr. 2006;25:454–465. doi: 10.1016/j.clnu.2005.12.008. [DOI] [PubMed] [Google Scholar]
  • 18.McNeel RL, Smith EO, Mersmann HJ. Isomers of conjugated linoleic acid modulate human preadipocyte differentiation. In Vitro Cell Dev Biol Anim. 2003;39:375–382. doi: 10.1290/1543-706X(2003)039<0375:IOCLAM>2.0.CO;2. [DOI] [PubMed] [Google Scholar]
  • 19.Meadus WJ, MacInnis R, Dugan ME. Prolonged dietary treatment with conjugated linoleic acid stimulates porcine muscle peroxisome proliferator activated receptor gamma and glutamine-fructose aminotransferase gene expression in vivo. J Mol Endocrinol. 2002;28:79–86. doi: 10.1677/jme.0.0280079. [DOI] [PubMed] [Google Scholar]
  • 20.Kohno H, Suzuki R, Noguchi R, Hosokawa M, Miyashita K, Tanaka T. Dietary anticancer effect of bitter gourd oil. 2002 AOCS Annual Meeting; Minneapolis, Minnesota: AOCS; 2002. [Google Scholar]
  • 21.Yu Y, Correll PH, Vanden Heuvel JP. Conjugated linoleic acid decreases production of pro-inflammatory products in macrophages: evidence for a PPAR gamma-dependent mechanism. Biochim Biophys Acta. 2002;1581:89–99. doi: 10.1016/s1388-1981(02)00126-9. [DOI] [PubMed] [Google Scholar]
  • 22.Yang M, Cook ME. Dietary conjugated linoleic acid decreased cachexia, macrophage tumor necrosis factor-alpha production, and modifies splenocyte cytokines production. Exp Biol Med (Maywood) 2003;228:51–58. doi: 10.1177/153537020322800107. [DOI] [PubMed] [Google Scholar]
  • 23.Kennedy A, Chung S, LaPoint K, Fabiyi O, McIntosh MK. Trans-10, cis-12 conjugated linoleic acid antagonizes ligand-dependent PPARgamma activity in primary cultures of human adipocytes. J Nutr. 2008;138:455–461. doi: 10.1093/jn/138.3.455. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Miller JR, Siripurkpong P, Hawes J, Majdalawieh A, Ro HS, McLeod RS. The trans-10, cis-12 isomer of conjugated linoleic acid decreases adiponectin assembly by PPARgamma-dependent and PPARgamma-independent mechanisms. J Lipid Res. 2008;49:550–562. doi: 10.1194/jlr.M700275-JLR200. [DOI] [PubMed] [Google Scholar]
  • 25.Brandebourg TD, Hu CY. Isomer-specific regulation of differentiating pig preadipocytes by conjugated linoleic acids. J Anim Sci. 2005;83:2096–2105. doi: 10.2527/2005.8392096x. [DOI] [PubMed] [Google Scholar]
  • 26.Ewaschuk JB, Walker JW, Diaz H, Madsen KL. Bioproduction of conjugated linoleic acid by probiotic bacteria occurs in vitro and in vivo in mice. J Nutr. 2006;136:1483–1487. doi: 10.1093/jn/136.6.1483. [DOI] [PubMed] [Google Scholar]
  • 27.Whigham LD, Cook EB, Stahl JL, Saban R, Bjorling DE, Pariza MW, Cook ME. CLA reduces antigen-induced histamine and PGE(2) release from sensitized guinea pig tracheae. Am J Physiol Regul Integr Comp Physiol. 2001;280:R908–R912. doi: 10.1152/ajpregu.2001.280.3.R908. [DOI] [PubMed] [Google Scholar]
  • 28.Whigham LD, Higbee A, Bjorling DE, Park Y, Pariza MW, Cook ME. Decreased antigen-induced eicosanoid release in conjugated linoleic acid-fed guinea pigs. Am J Physiol Regul Integr Comp Physiol. 2002;282:R1104–R1112. doi: 10.1152/ajpregu.00075.2001. [DOI] [PubMed] [Google Scholar]
  • 29. Evans NP, Misyak S, Schmelz EM, Guri AJ, Hontecillas R, Bassaganya-Riera J. CLA decreases inflammation-induced colorectal cancer in mice through activation of PPAR γ. J. Nutr. 2010 doi: 10.3945/jn.109.115642. In Press. * An important article demonstrating that CLA ameliorates IBD-related colorectal cancer possibly through a PPAR gamma-dependent mechanis
  • 30.Hontecillas R, Bassaganya-Riera J. Peroxisome proliferator-activated receptor gamma is required for regulatory CD4+ T cell-mediated protection against colitis. J Immunol. 2007;178:2940–2949. doi: 10.4049/jimmunol.178.5.2940. [DOI] [PubMed] [Google Scholar]
  • 31.Wohlfert EA, Nichols FC, Nevius E, Clark RB. Peroxisome proliferator-activated receptor gamma (PPARgamma) and immunoregulation: enhancement of regulatory T cells through PPARgamma-dependent and - independent mechanisms. J Immunol. 2007;178:4129–4135. doi: 10.4049/jimmunol.178.7.4129. [DOI] [PubMed] [Google Scholar]
  • 32.Whigham LD, O'Shea M, Mohede IC, Walaski HP, Atkinson RL. Safety profile of conjugated linoleic acid in a 12-month trial in obese humans. Food Chem Toxicol. 2004;42:1701–1709. doi: 10.1016/j.fct.2004.06.008. [DOI] [PubMed] [Google Scholar]
  • 33.Bassaganya-Riera J, Hontecillas R, Zimmerman DR, Wannemuehler MJ. Dietary conjugated linoleic acid modulates phenotype and effector functions of porcine cd8(+) lymphocytes. J Nutr. 2001;131:2370–2377. doi: 10.1093/jn/131.9.2370. [DOI] [PubMed] [Google Scholar]
  • 34.Bassaganya-Riera J, Hontecillas R, Zimmerman DR, Wannemuehler MJ. Long-term influence of lipid nutrition on the induction of CD8(+) responses to viral or bacterial antigens. Vaccine. 2002;20:1435–1444. doi: 10.1016/s0264-410x(01)00465-0. [DOI] [PubMed] [Google Scholar]
  • 35.Bassaganya-Riera J, Pogranichniy RM, Jobgen SC, Halbur PG, Yoon KJ, O'Shea M, Mohede I, Hontecillas R. Conjugated linoleic acid ameliorates viral infectivity in a pig model of virally induced immunosuppression. J Nutr. 2003;133:3204–3214. doi: 10.1093/jn/133.10.3204. [DOI] [PubMed] [Google Scholar]
  • 36.Albers R, Van Der Wielen RP, Brink EJ, Hendriks HF, Dorovska-Taran VN, Mohede IC. Effects of cis-9, trans-11 and trans-10, cis-12 conjugated linoleic acid (CLA) isomers on immune function in healthy men. Eur J Clin Nutr. 2003;57:595–603. doi: 10.1038/sj.ejcn.1601585. [DOI] [PubMed] [Google Scholar]
  • 37.Kelley DS, Taylor PC, Nelson GJ, Schmidt PC, Ferretti A, Erickson KL, Yu R, Chandra RK, Mackey BE. Docosahexaenoic acid ingestion inhibits natural killer cell activity and production of inflammatory mediators in young healthy men. Lipids. 1999;34:317–324. doi: 10.1007/s11745-999-0369-5. [DOI] [PubMed] [Google Scholar]
  • 38.Kew S, Gibbons ES, Thies F, McNeill GP, Quinlan PT, Calder PC. The effect of feeding structured triacylglycerols enriched in eicosapentaenoic or docosahexaenoic acids on murine splenocyte fatty acid composition and leucocyte phagocytosis. Br J Nutr. 2003;90:1071–1080. doi: 10.1079/bjn2003996. [DOI] [PubMed] [Google Scholar]
  • 39.Serhan CN, Clish CB, Brannon J, Colgan SP, Chiang N, Gronert K. Novel Functional Sets of Lipid-derived Mediators with Antiinflammatory Actions Generated from Omega-3 Fatty Acids via Cyclooxygenase 2-Nonsteroidal Antiinflammatory Drugs and Transcellular Processing. J Exp Med. 2000;192:1197–1204. doi: 10.1084/jem.192.8.1197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Nieto N, Torres MI, Rios A, Gil A. Dietary polyunsaturated fatty acids improve histological and biochemical alterations in rats with experimental ulcerative colitis. J Nutr. 2002;132:11–19. doi: 10.1093/jn/132.1.11. [DOI] [PubMed] [Google Scholar]
  • 41.MacLean CH, Mojica WA, Newberry SJ, Pencharz J, Garland RH, Tu W, Hilton LG, Gralnek IM, Rhodes S, Khanna P, Morton SC. Systematic review of the effects of n-3 fatty acids in inflammatory bowel disease. Am J Clin Nutr. 2005;82:611–619. doi: 10.1093/ajcn.82.3.611. [DOI] [PubMed] [Google Scholar]
  • 42. Uchiyama K, Nakamura M, Odahara S, Koido S, Katahira K, Shiraishi H, Ohkusa T, Fujise K, Tajiri H. N-3 polyunsaturated fatty acid diet therapy for patients with inflammatory bowel disease. Inflamm Bowel Dis. 2010 doi: 10.1002/ibd.21251. * An original research article demonstrating efficacy of n-3 PUFA in maintaining IBD remission
  • 43.Brix S, Lund P, Kjaer TM, Straarup EM, Hellgren LI, Frokiaer H. CD4(+) T-cell activation is differentially modulated by bacteria-primed dendritic cells, but is generally down-regulated by n-3 polyunsaturated fatty acids. Immunology. 2009 doi: 10.1111/j.1365-2567.2009.03163.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Clark RB, Bishop-Bailey D, Estrada-Hernandez T, Hla T, Puddington L, Padula SJ. The nuclear receptor PPAR gamma and immunoregulation: PPAR gamma mediates inhibition of helper T cell responses. J Immunol. 2000;164:1364–1371. doi: 10.4049/jimmunol.164.3.1364. [DOI] [PubMed] [Google Scholar]
  • 45.Jump DB, Clarke SD. Regulation of gene expression by dietary fat. Annu Rev Nutr. 1999;19:63–90. doi: 10.1146/annurev.nutr.19.1.63. [DOI] [PubMed] [Google Scholar]
  • 46.Stein JL, Marsh TL, Wu KY, Shizuya H, DeLong EF. Characterization of uncultivated prokaryotes: isolation and analysis of a 40-kilobase-pair genome fragment from a planktonic marine archaeon. J Bacteriol. 1996;178:591–599. doi: 10.1128/jb.178.3.591-599.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Rondon MR, August PR, Bettermann AD, Brady SF, Grossman TH, Liles MR, Loiacono KA, Lynch BA, MacNeil IA, Minor C, Tiong CL, Gilman M, Osburne MS, Clardy J, Handelsman J, Goodman RM. Cloning the soil metagenome: a strategy for accessing the genetic and functional diversity of uncultured microorganisms. Appl Environ Microbiol. 2000;66:2541–2547. doi: 10.1128/aem.66.6.2541-2547.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Schmidt TM, DeLong EF, Pace NR. Analysis of a marine picoplankton community by 16S rRNA gene cloning and sequencing. J Bacteriol. 1991;173:4371–4378. doi: 10.1128/jb.173.14.4371-4378.1991. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.DeLong EF, Preston CM, Mincer T, Rich V, Hallam SJ, Frigaard NU, Martinez A, Sullivan MB, Edwards R, Brito BR, Chisholm SW, Karl DM. Community genomics among stratified microbial assemblages in the ocean's interior. Science. 2006;311:496–503. doi: 10.1126/science.1120250. [DOI] [PubMed] [Google Scholar]
  • 50.Tyson GW, Chapman J, Hugenholtz P, Allen EE, Ram RJ, Richardson PM, Solovyev VV, Rubin EM, Rokhsar DS, Banfield JF. Community structure and metabolism through reconstruction of microbial genomes from the environment. Nature. 2004;428:37–43. doi: 10.1038/nature02340. [DOI] [PubMed] [Google Scholar]
  • 51.Strous M, Pelletier E, Mangenot S, Rattei T, Lehner A, Taylor MW, Horn M, Daims H, Bartol-Mavel D, Wincker P, Barbe V, Fonknechten N, Vallenet D, Segurens B, Schenowitz-Truong C, Medigue C, Collingro A, Snel B, Dutilh BE, Op den Camp HJ, van der Drift C, Cirpus I, van de Pas-Schoonen KT, Harhangi HR, van Niftrik L, Schmid M, Keltjens J, van de Vossenberg J, Kartal B, Meier H, Frishman D, Huynen MA, Mewes HW, Weissenbach J, Jetten MS, Wagner M, Le Paslier D. Deciphering the evolution and metabolism of an anammox bacterium from a community genome. Nature. 2006;440:790–794. doi: 10.1038/nature04647. [DOI] [PubMed] [Google Scholar]
  • 52.Tringe SG, von Mering C, Kobayashi A, Salamov AA, Chen K, Chang HW, Podar M, Short JM, Mathur EJ, Detter JC, Bork P, Hugenholtz P, Rubin EM. Comparative metagenomics of microbial communities. Science. 2005;308:554–557. doi: 10.1126/science.1107851. [DOI] [PubMed] [Google Scholar]
  • 53.Woyke T, Teeling H, Ivanova NN, Huntemann M, Richter M, Gloeckner FO, Boffelli D, Anderson IJ, Barry KW, Shapiro HJ, Szeto E, Kyrpides NC, Mussmann M, Amann R, Bergin C, Ruehland C, Rubin EM, Dubilier N. Symbiosis insights through metagenomic analysis of a microbial consortium. Nature. 2006;443:950–955. doi: 10.1038/nature05192. [DOI] [PubMed] [Google Scholar]
  • 54.Gao Z, Tseng CH, Pei Z, Blaser MJ. Molecular analysis of human forearm superficial skin bacterial biota. Proc Natl Acad Sci U S A. 2007;104:2927–2932. doi: 10.1073/pnas.0607077104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Gill SR, Pop M, Deboy RT, Eckburg PB, Turnbaugh PJ, Samuel BS, Gordon JI, Relman DA, Fraser-Liggett CM, Nelson KE. Metagenomic analysis of the human distal gut microbiome. Science. 2006;312:1355–1359. doi: 10.1126/science.1124234. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Mahowald MA, Rey FE, Seedorf H, Turnbaugh PJ, Fulton RS, Wollam A, Shah N, Wang C, Magrini V, Wilson RK, Cantarel BL, Coutinho PM, Henrissat B, Crock LW, Russell A, Verberkmoes NC, Hettich RL, Gordon JI. Characterizing a model human gut microbiota composed of members of its two dominant bacterial phyla. Proc Natl Acad Sci U S A. 2009;106:5859–5864. doi: 10.1073/pnas.0901529106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Turnbaugh PJ, Backhed F, Fulton L, Gordon JI. Diet-induced obesity is linked to marked but reversible alterations in the mouse distal gut microbiome. Cell Host Microbe. 2008;3:213–223. doi: 10.1016/j.chom.2008.02.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Turnbaugh PJ, Gordon JI. An invitation to the marriage of metagenomics and metabolomics. Cell. 2008;134:708–713. doi: 10.1016/j.cell.2008.08.025. [DOI] [PubMed] [Google Scholar]
  • 59.Ley RE, Backhed F, Turnbaugh P, Lozupone CA, Knight RD, Gordon JI. Obesity alters gut microbial ecology. Proc Natl Acad Sci U S A. 2005;102:11070–11075. doi: 10.1073/pnas.0504978102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Ley RE, Peterson DA, Gordon JI. Ecological and evolutionary forces shaping microbial diversity in the human intestine. Cell. 2006;124:837–848. doi: 10.1016/j.cell.2006.02.017. [DOI] [PubMed] [Google Scholar]
  • 61.Ley RE, Turnbaugh PJ, Klein S, Gordon JI. Microbial ecology: human gut microbes associated with obesity. Nature. 2006;444:1022–1023. doi: 10.1038/4441022a. [DOI] [PubMed] [Google Scholar]
  • 62.Guri AJ, Hontecillas R, Bassaganya-Riera J. Abscisic acid ameliorates experimental IBD by downregulating cellular adhesion molecule expression and suppressing immune cell infiltration. Clinical Nutrition. 2010 doi: 10.1016/j.clnu.2010.02.009. In Press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Wendelsdorf k, Bassaganya-Riera J, Hontecillas R, Eubank S. Model of colon inflammation: Immune Modulatory Mechanisms in IBD. Journal of Theoretical Biology. 2010 doi: 10.1016/j.jtbi.2010.03.027. In Press. * An original research publication showing the first mathematical model of IBD.
  • 64.Odegaard JI, Ricardo-Gonzalez RR, Goforth MH, Morel CR, Subramanian V, Mukundan L, Red Eagle A, Vats D, Brombacher F, Ferrante AW, Chawla A. Macrophage-specific PPARgamma controls alternative activation and improves insulin resistance. Nature. 2007;447:1116–1120. doi: 10.1038/nature05894. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES