1
|
Giampieri F, Cianciosi D, Ansary J, Elexpuru-Zabaleta M, Forbes-Hernandez TY, Battino M. Immunoinflammatory effects of dietary bioactive compounds. ADVANCES IN FOOD AND NUTRITION RESEARCH 2021; 95:295-336. [PMID: 33745515 DOI: 10.1016/bs.afnr.2020.12.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Inflammation is a key mechanism of the immune system that can be elicited by several factors, among them several chemical, physical and biological agents. Once stimulated, the inflammatory response activates a series of signaling pathways and a number of immune cells which promote, in a very coordinated manner, the neutralization of the infectious agent. However, if uncontrolled, the inflammatory status may become chronic leading, potentially, to tissue damage and disease onset. Several risk factors are associated with the development of chronic inflammation and, among these factors, diet plays an essential role. In this chapter the effects of some dietary bioactive compounds, including micronutrients, omega-3 fatty acids, nucleotides and polyphenols, on the immunoinflammatory responses in different cellular, animal and human studies have been summarized.
Collapse
Affiliation(s)
- Francesca Giampieri
- Dipartimento di Scienze Cliniche Specialistiche ed Odontostomatologiche, Sez. Biochimica, Università Politecnica delle Marche, Ancona, Italy; Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Danila Cianciosi
- Dipartimento di Scienze Cliniche Specialistiche ed Odontostomatologiche, Sez. Biochimica, Università Politecnica delle Marche, Ancona, Italy
| | - Johura Ansary
- Dipartimento di Scienze Cliniche Specialistiche ed Odontostomatologiche, Sez. Biochimica, Università Politecnica delle Marche, Ancona, Italy
| | | | - Tamara Y Forbes-Hernandez
- Nutrition and Food Science Group, Department of Analytical and Food Chemistry, CITACA, CACTI, University of Vigo, Vigo, Spain
| | - Maurizio Battino
- Dipartimento di Scienze Cliniche Specialistiche ed Odontostomatologiche, Sez. Biochimica, Università Politecnica delle Marche, Ancona, Italy; International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang, China.
| |
Collapse
|
2
|
Marcelino JM, Villas Boas GR, Cunha M, Deus Júnior R, Castro LH, Araújo FH, Traesel GK, Dos Santos AC, Souza RI, Paes M, Gubert P, Guterres ZDR, de Lima FF, Silva T, Silva RC, Cardoso CAL, Argandoña EJ, Macorini LF, Oesterreich SA. Determination of preclinical safety of oil obtained from Pachira aquatica Aublet (Malvaceae) seeds: histopathological, biochemical, hematological, and genetic toxicity studies in rats. Drug Chem Toxicol 2020; 45:1504-1521. [PMID: 33252270 DOI: 10.1080/01480545.2020.1845713] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Pachira aquatica is a species used for medicinal and food purposes and has numerous phytochemicals that may have systemic toxic effects and damage to genetic material. This study aimed to evaluate acute and short-term oral toxicity, as well as genotoxic and clastogenic effects of oil extracted from P. aquatica (PASO) seeds in rats and Drosophila melanogaster. The results obtained with biochemical and hematological analyses did not show significant changes in any evaluated parameters when compared with reference values for the species used in the study. Data from the histopathological analysis corroborated results found in this study. These findings indicate low acute and short-term toxicity following oral PASO exposure in rats under the experimental conditions tested. Tests performed in rats showed that PASO did not present significant genotoxic or clastogenic effects on the cells analyzed with the three doses tested. Treatment with PASO in the offspring of HB crossing, which showed high cytochrome P450 levels, did not exhibit genotoxic activity, as demonstrated by the SMART test. These results suggest that products from the hepatic oil metabolism did not show genotoxicity under the conditions tested. Together, the results indicate that, under the experimental conditions tested, PASO is safe for repeated intake. As PASO exhibited low potential to cause harmful effects on living organisms, our study encourages further research aimed at assessing its pharmacological activity, since it is a widely consumed plant.
Collapse
Affiliation(s)
| | - Gustavo Roberto Villas Boas
- Research Group on Development of Pharmaceutical Products (P&DProFar), Center for Biological and Health Sciences, Federal University of Western Bahia, Bahia, Brazil
| | - Mila Cunha
- Research Group on Development of Pharmaceutical Products (P&DProFar), Center for Biological and Health Sciences, Federal University of Western Bahia, Bahia, Brazil
| | - Ramão Deus Júnior
- Faculty of Health Sciences, Federal University of Grande Dourados, Dourados, Brazil
| | - Luis Henrique Castro
- Faculty of Health Sciences, Federal University of Grande Dourados, Dourados, Brazil
| | | | | | | | | | - Marina Paes
- Research Group on Development of Pharmaceutical Products (P&DProFar), Center for Biological and Health Sciences, Federal University of Western Bahia, Bahia, Brazil
| | - Priscila Gubert
- Research Group on Development of Pharmaceutical Products (P&DProFar), Center for Biological and Health Sciences, Federal University of Western Bahia, Bahia, Brazil.,Laboratory of Imunopathology KeizoAsami, Department of Biochemistry, Federal University of Pernambuco, Recife, Brazil
| | - Zaira da Rosa Guterres
- Laboratory of Phytogenesis and Mutagenesis, State University of Mato Grosso do Sul, Mato Grosso do Sul, Brazil
| | | | - Tania Silva
- Postgraduate Program in Environmental Science and Technology, Federal University of Grande Dourados, Mato Grosso do Sul, Brazil
| | - Rogério César Silva
- Chemistry Course, State University of Mato Grosso do Sul, Mato Grosso do Sul, Brazil
| | | | - Eliana Janet Argandoña
- Faculty of Engineering - FAEN/UFGD, Federal University of Grande Dourados, Mato Grosso do Sul, Brazil
| | - Luis Fernando Macorini
- Faculty of Biological Sciences and Health, University Center of Grande Dourados, Mato Grosso do Sul, Brazil
| | | |
Collapse
|
3
|
Long F, Yang X, Liu D, Guo Y, Wang Z. Involvement of the PKC–NF–κB signaling pathway in the regulation of T lymphocytes proliferation of chickens by conjugated linoleic acids. FOOD AGR IMMUNOL 2015. [DOI: 10.1080/09540105.2015.1079590] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
4
|
Calder PC. Marine omega-3 fatty acids and inflammatory processes: Effects, mechanisms and clinical relevance. Biochim Biophys Acta Mol Cell Biol Lipids 2014; 1851:469-84. [PMID: 25149823 DOI: 10.1016/j.bbalip.2014.08.010] [Citation(s) in RCA: 961] [Impact Index Per Article: 96.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Revised: 08/12/2014] [Accepted: 08/13/2014] [Indexed: 12/15/2022]
Abstract
Inflammation is a condition which contributes to a range of human diseases. It involves a multitude of cell types, chemical mediators, and interactions. Eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) are omega-3 (n-3) fatty acids found in oily fish and fish oil supplements. These fatty acids are able to partly inhibit a number of aspects of inflammation including leukocyte chemotaxis, adhesion molecule expression and leukocyte-endothelial adhesive interactions, production of eicosanoids like prostaglandins and leukotrienes from the n-6 fatty acid arachidonic acid, production of inflammatory cytokines, and T-helper 1 lymphocyte reactivity. In addition, EPA gives rise to eicosanoids that often have lower biological potency than those produced from arachidonic acid and EPA and DHA give rise to anti-inflammatory and inflammation resolving mediators called resolvins, protectins and maresins. Mechanisms underlying the anti-inflammatory actions of marine n-3 fatty acids include altered cell membrane phospholipid fatty acid composition, disruption of lipid rafts, inhibition of activation of the pro-inflammatory transcription factor nuclear factor kappa B so reducing expression of inflammatory genes, activation of the anti-inflammatory transcription factor peroxisome proliferator activated receptor γ and binding to the G protein coupled receptor GPR120. These mechanisms are interlinked, although the full extent of this is not yet elucidated. Animal experiments demonstrate benefit from marine n-3 fatty acids in models of rheumatoid arthritis (RA), inflammatory bowel disease (IBD) and asthma. Clinical trials of fish oil in RA demonstrate benefit, but clinical trials of fish oil in IBD and asthma are inconsistent with no overall clear evidence of efficacy. This article is part of a Special Issue entitled "Oxygenated metabolism of PUFA: analysis and biological relevance".
Collapse
Affiliation(s)
- Philip C Calder
- Human Development and Health Academic Unit, Faculty of Medicine, University of Southampton, Southampton, UK; NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust and University of Southampton, Southampton, UK; Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia.
| |
Collapse
|
5
|
Abstract
Numerous effects of n-3 fatty acids EPA and DHA on functional responses of cells involved in inflammation and immunity have been described. Fatty acid-induced modifications in membrane order and in the availability of substrates for eicosanoid synthesis are long-standing mechanisms that are considered important in explaining the effects observed. More recently, effects on signal transduction pathways and on gene expression profiles have been identified. Over the last 10 years or so, significant advances in understanding the mechanisms of action of n-3 fatty acids have been made. These include the identification of new actions of lipid mediators that were already described and of novel interactions among those mediators and the description of an entirely new family of lipid mediators, resolvins and protectins that have anti-inflammatory actions and are critical to the resolution of inflammation. It is also recognised that EPA and DHA can inhibit activation of the prototypical inflammatory transcription factor NF-κB. Recent studies suggest three alternative mechanisms by which n-3 fatty acids might have this effect. Within T-cells, as well as other cells of relevance to immune and inflammatory responses, EPA and DHA act to disrupt very early events involving formation of the structures termed lipid rafts which bring together various proteins to form an effective signalling platform. In summary, recent research has identified a number of new mechanisms of action that help to explain previously identified effects of n-3 fatty acids on inflammation and immunity.
Collapse
|
6
|
Abstract
Long chain omega-3 polyunsaturated fatty acids (LC n-3 PUFAs) are recommended for management of patients with wide-ranging chronic diseases, including coronary heart disease, rheumatoid arthritis, dementia, and depression. Increased consumption of eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) is recommended by many health authorities to prevent (up to 0.5 g/day) or treat chronic disease (1.0 g/day for coronary heart disease; 1.2–4 g/day for elevated triglyceride levels). Recommendations for dietary intake of LC n-3 PUFAs are often provided for α-linolenic acid, and for the combination of EPA and DHA. However, many studies have also reported differential effects of EPA, DHA and their metabolites in the clinic and at the laboratory bench. The aim of this article is to review studies that have identified divergent responses to EPA and DHA, and to explore reasons for these differences. In particular, we review potential contributing factors such as differential membrane incorporation, modulation of gene expression, activation of signaling pathways and metabolite formation. We suggest that there may be future opportunity to refine recommendations for intake of individual LC n-3 PUFAs.
Collapse
Affiliation(s)
- Fraser D Russell
- Faculty of Science, Health, Education and Engineering, University of the Sunshine Coast, Maroochydore, Queensland 4556, Australia.
| | | |
Collapse
|
7
|
Shaikh SR, Jolly CA, Chapkin RS. n-3 Polyunsaturated fatty acids exert immunomodulatory effects on lymphocytes by targeting plasma membrane molecular organization. Mol Aspects Med 2011; 33:46-54. [PMID: 22020145 DOI: 10.1016/j.mam.2011.10.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2011] [Revised: 10/04/2011] [Accepted: 10/09/2011] [Indexed: 01/26/2023]
Abstract
Fish oil, enriched in bioactive n-3 polyunsaturated fatty acids (PUFA), has therapeutic value for the treatment of inflammation-associated disorders. The effects of n-3 PUFAs are pleiotropic and complex; hence, an understanding of their cellular targets and molecular mechanisms of action remains incomplete. Here we focus on recent data indicating n-3 PUFAs exert immunosuppressive effects on the function of effector and regulatory CD4(+) T cells. In addition, we also present emerging evidence that n-3 PUFAs have immunomodulatory effects on B cells. We then focus on one multifaceted mechanism of n-3 PUFAs, which is the alteration of the biophysical and biochemical organization of the plasma membrane. This mechanism is central for downstream signaling, eicosanoid production, transcriptional regulation and cytokine secretion. We highlight recent work demonstrating n-3 PUFA acyl chains in the plasma membrane target the lateral organization of membrane signaling assemblies (i.e. lipid rafts or signaling networks) and de novo phospholipid biosynthesis. We conclude by proposing new functional and mechanistic questions in this area of research that will aid in the development of fish oil as adjuvant therapy for treating unresolved chronic inflammation.
Collapse
Affiliation(s)
- Saame Raza Shaikh
- Department of Biochemistry & Molecular Biology and East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC 27834, USA.
| | | | | |
Collapse
|
8
|
Akhtar Khan N. Polyunsaturated fatty acids in the modulation of T-cell signalling. Prostaglandins Leukot Essent Fatty Acids 2010; 82:179-87. [PMID: 20189788 DOI: 10.1016/j.plefa.2010.02.023] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2009] [Indexed: 11/25/2022]
Abstract
n-3 polyunsaturated fatty acids (PUFA) have been shown to modulate immune responses. These agents, being considered as adjuvant immunosuppressants, have been used in the treatment of various inflammatory and autoimmune diseases. However, the molecular mechanisms of action of n-3 PUFA-induced immunosuppressive effects are not well-understood. Since exogenous n-3 PUFA, under in vitro and in vivo conditions, are efficiently incorporated into T-cell plasma membranes, a number of recent studies have demonstrated that these agents may modulate T-cell signalling. In this review, the interactions of n-3 PUFA with the second messenger cascade initiated during early and late events of T-cell activation are discussed. We particularly focus on how these fatty acids can modulate the production of diacylglycerol and the activation of protein kinase C, mitogen activated protein kinase, calcium signalling and translocation of transcriptional factors, implicated in the regulation of gene transcription in T-cells.
Collapse
Affiliation(s)
- Naim Akhtar Khan
- University of Burgundy, Department of Physiology, UPRES EA4183 Lipids and Cell Signalling, Faculty of Life Sciences, Dijon, France.
| |
Collapse
|
9
|
Chapkin RS, Kim W, Lupton JR, McMurray DN. Dietary docosahexaenoic and eicosapentaenoic acid: emerging mediators of inflammation. Prostaglandins Leukot Essent Fatty Acids 2009; 81:187-91. [PMID: 19502020 PMCID: PMC2755221 DOI: 10.1016/j.plefa.2009.05.010] [Citation(s) in RCA: 205] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The inflammatory response is designed to help fight and clear infection, remove harmful chemicals, and repair damaged tissue and organ systems. Although this process, in general, is protective, the failure to resolve the inflammation and return the target tissue to homeostasis can result in disease, including the promotion of cancer. A plethora of published literature supports the contention that dietary n-3 polyunsaturated fatty acids (PUFA), and eicosapentaenoic (EPA, 20:5n-3) and docosahexaenoic acid (DHA, 22:6n-3) in particular, are important modulators of a host's inflammatory/immune responses. The following review describes a mechanistic model that may explain, in part, the pleiotropic anti-inflammatory and immunosuppressive properties of EPA and DHA. In this review, we focus on salient studies that address three overarching mechanisms of n-3 PUFA action: (i) modulation of nuclear receptor activation, i.e., nuclear factor-kappaB (NF-kappaB) suppression; (ii) suppression of arachidonic acid-cyclooxygenase-derived eicosanoids, primarily prostaglandin E(2)-dependent signaling; and (iii) alteration of the plasma membrane micro-organization (lipid rafts), particularly as it relates to the function of Toll-like receptors (TLRs), and T-lymphocyte signaling molecule recruitment to the immunological synapse (IS). We propose that lipid rafts may be targets for the development of n-3 PUFA-containing dietary bioactive agents to down-modulate inflammatory and immune responses and for the treatment of autoimmune and chronic inflammatory diseases.
Collapse
Affiliation(s)
- Robert S Chapkin
- Department of Nutrition and Food Science, Faculty of Nutrition, Kleberg Biotechnology Center, MS 2253, Texas A&M University, College Station, TX 77843-2253, USA.
| | | | | | | |
Collapse
|
10
|
Biondo PD, Brindley DN, Sawyer MB, Field CJ. The potential for treatment with dietary long-chain polyunsaturated n-3 fatty acids during chemotherapy. J Nutr Biochem 2008; 19:787-96. [PMID: 18602809 DOI: 10.1016/j.jnutbio.2008.02.003] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2007] [Revised: 02/08/2008] [Accepted: 02/15/2008] [Indexed: 12/19/2022]
Abstract
Dietary intake of long-chain omega-3 (or n-3) polyunsaturated fatty acids (PUFA), eicosapentaenoic acid (EPA), and docosahexaenoic acid (DHA) can affect numerous processes in the body, including cardiovascular, neurological and immune functions, as well as cancer. Studies on human cancer cell lines, animal models and preliminary trials with human subjects suggest that administration of EPA and DHA, found naturally in our diet in fatty fish, can alter toxicities and/or activity of many drugs used to treat cancer. Multiple mechanisms are proposed to explain how n-3 PUFA modulate the tumor cell response to chemotherapeutic drugs. n-3 PUFA are readily incorporated into cell membranes and lipid rafts, and their incorporation may affect membrane-associated signaling proteins such as Ras, Akt and Her-2/neu. Due to their high susceptibility to oxidation, it has also been proposed that n-3 PUFA may cause irreversible tumor cell damage through increased lipid peroxidation. n-3 PUFA may increase tumor cell susceptibility to apoptosis by altering expression or function of apoptotic proteins, or by modulating activity of survival-related transcription factors such as nuclear factor-kappaB. Some studies suggest n-3 PUFA may increase drug uptake or even enhance drug activation (e.g., in the case of some nucleoside analogue drugs). Further research is warranted to identify specific mechanisms by which n-3 PUFA increase chemotherapy efficacy and to determine the optimal cellular/membrane levels of n-3 PUFA required to promote these mechanisms, such that these fatty acids may be prescribed as adjuvants to chemotherapy.
Collapse
Affiliation(s)
- Patricia D Biondo
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada T6G 2P5
| | | | | | | |
Collapse
|
11
|
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.
Collapse
|
12
|
Aires V, Hichami A, Boulay G, Khan NA. Activation of TRPC6 calcium channels by diacylglycerol (DAG)-containing arachidonic acid: A comparative study with DAG-containing docosahexaenoic acid. Biochimie 2007; 89:926-37. [PMID: 17532549 DOI: 10.1016/j.biochi.2006.10.016] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2006] [Accepted: 10/27/2006] [Indexed: 11/29/2022]
Abstract
We synthesized a diacylglycerol (DAG)-containing arachidonic acid, i.e., 1-stearoyl-2-arachidonyl-sn-glycerol (SAG), and studied its implication in the modulation of canonical transient receptor potential sub-type 6 (TRPC6) channels in stably-transfected HEK-293 cells. SAG induced the influx of Ca(2+), and also of other bivalent cations like Ba(2+) and Sr(2+), in these cells. SAG-evoked Ca(2+) influx was not due to its metabolites as inhibitors of DAG-lipase (RHC80267) and DAG-kinase (R50922) failed to inhibit the response of the same. To emphasise that SAG exerts its action via its DAG configuration, but not due to the presence of stearic acid at sn-1 position, we synthesized 1-palmitoyl-2-arachidonyl-sn-glycerol (PAG). PAG-induced increases in [Ca(2+)](i) were not significantly different from those induced by SAG. For the comparative studies, we also synthesized the DAG-containing docosahexaenoic acid, i.e., 1-stearoyl-2-docosahexaenoyl-sn-glycerol (SDG). We observed that SDG and 1,2-dioctanoyl-sn-glycerol (DOG), a DAG analogue, also evoked increases in [Ca(2+)](i), which were lesser than those evoked by SAG. However, activation of TRPC6 channels by all the DAG molecular species (SAG, DOG and SDG) required Src kinases as the tyrosine kinase inhibitors, PP2 and SU6656, significantly attenuated the increases in [Ca(2+)](i) evoked by these agents. Moreover, disruption of lipid rafts with methyl-beta-cyclodextrin completely abolished SAG-, DOG- and SDG-induced increases in [Ca(2+)](i). The present study shows that SAG as well as SDG and DOG stimulate Ca(2+) influx through the activation of TRPC6 calcium channels which are regulated by Src kinases and intact lipid raft domains.
Collapse
Affiliation(s)
- Virginie Aires
- Département de Physiologie, UPRES Lipides and Nutrition, Université de Bourgogne, Faculté des Sciences de la Vie, 6 Boulevard Gabriel, 2100 Dijon, France
| | | | | | | |
Collapse
|
13
|
Chapkin RS, Davidson LA, Ly L, Weeks BR, Lupton JR, McMurray DN. Immunomodulatory effects of (n-3) fatty acids: putative link to inflammation and colon cancer. J Nutr 2007; 137:200S-204S. [PMID: 17182826 DOI: 10.1093/jn/137.1.200s] [Citation(s) in RCA: 108] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Chronic inflammation and colorectal cancer are closely linked. Although the overall mechanisms of inflammation-associated gastrointestinal carcinogenesis are complex, it is clear that antiinflammatory therapy is efficacious against neoplastic progression and malignant conversion. From a dietary perspective, fish oil containing (n-3) polyunsaturated fatty acids (PUFAs) has antiinflammatory properties, but for years the mechanism has remained obscure. Of relevance to the immune system in the intestine, we showed that (n-3) PUFA feeding alters the balance between CD4+ T-helper (Th1 and Th2) subsets by directly suppressing Th1 cell development (i.e., clonal expansion). This is noteworthy because Th1 cells mediate inflammatory diseases and resistance to intracellular pathogens or allergic hypersensitivity, and Th2 cells mediate resistance to extracellular pathogens. Therefore, any changes induced by (n-3) PUFAs in T-cell subset balance and function are important because the outcome is expected to suppress the development of autoimmune diseases and possibly the occurrence of colon cancer. Precisely how the immunomodulatory effects of (n-3) PUFAs influence inflammation-associated colonic tumor development is the subject of an ongoing investigation.
Collapse
Affiliation(s)
- Robert S Chapkin
- Faculty of Nutrition, Center for Environmental and Rural Health, Texas A&M University, College Station, TX 77843, USA.
| | | | | | | | | | | |
Collapse
|
14
|
Zhang P, Kim W, Zhou L, Wang N, Ly LH, McMurray DN, Chapkin RS. Dietary fish oil inhibits antigen-specific murine Th1 cell development by suppression of clonal expansion. J Nutr 2006; 136:2391-8. [PMID: 16920860 DOI: 10.1093/jn/136.9.2391] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
To determine the mechanisms by which dietary fish oil (FO) affects antigen-stimulated Th1 cell development, DO11.10 Rag 2(-/-) T cell receptor transgenic mice were fed a control diet (5% corn oil (CO) or a FO diet (1% CO + 4% FO, (n-3) PUFA) for 2 wk. CD4(+) T cells were cultured under neutral or Th1 polarizing conditions. FO feeding suppressed (P < 0.05) ovalbumin peptide-induced proliferation of nonpolarized CD4(+) T cells. Differentiation in vitro to Th1 cells was not affected by dietary FO, as evidenced by similar percentages of KJ1-26(+), IFN-gamma(+), IL-4(-) Th1 cells in cultures from CO-fed (99%) and FO-fed (97%) mice. However, the absolute number of viable Th1 cells in polarized cultures from FO-fed mice was less than half that observed in CO-fed mice (P < 0.05), indicating that FO inhibits in vitro Th1 clonal expansion. The reduced number of Th1 cells in FO cultures was not a result of increased apoptosis, because similar percentages of apoptotic Th1 cells were observed in cultures from FO- and CO-fed mice. IL-2-induced cell proliferation was significantly decreased in polarized Th1 cells from the FO group; however, the suppressed proliferation was not linked to reduced CD25 surface expression on antigen-stimulated CD4(+) T cells. Adoptively transferred CFSE-labeled DO11.10 CD4(+) cells into immunized mice (Th1 polarizing agents) showed that dietary FO reduced (P < 0.05) the number of cell divisions in vivo. These studies suggest that the attenuated inflammatory response which accompanies FO feeding may be explained, at least in part, by suppression of Th1 clonal expansion.
Collapse
Affiliation(s)
- Ping Zhang
- Department of Microbial and Molecular Pathogenesis, Texas A&M University System Health Science Center, College Station, TX, USA
| | | | | | | | | | | | | |
Collapse
|
15
|
Fan YY, Ly LH, Barhoumi R, McMurray DN, Chapkin RS. Dietary Docosahexaenoic Acid Suppresses T Cell Protein Kinase Cθ Lipid Raft Recruitment and IL-2 Production. THE JOURNAL OF IMMUNOLOGY 2004; 173:6151-60. [PMID: 15528352 DOI: 10.4049/jimmunol.173.10.6151] [Citation(s) in RCA: 195] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
To date, the proximal molecular targets through which dietary n-3 polyunsaturated fatty acids (PUFA) suppress the inflammatory process have not been elucidated. Because cholesterol and sphingolipid-enriched rafts have been proposed as platforms for compartmentalizing dynamically regulated signaling assemblies at the plasma membrane, we determined the in vivo effects of fish oil and highly purified docosahexaenoic acid (DHA; 22:6n-3) on T cell microdomain lipid composition and the membrane subdomain distribution of signal-transducing molecules (protein kinase C (PKC)theta;, linker for activation of T cells, and Fas/CD95), before and after stimulation. Mice were fed diets containing 5 g/100 g corn oil (control), 4 g/100 g fish oil (contains a mixture of n-3 PUFA) plus 1 g/100 g corn oil, or 4 g/100 g corn oil plus 1 g/100 g DHA ethyl ester for 14 days. Dietary n-3 PUFA were incorporated into splenic T cell lipid raft and soluble membrane phospholipids, resulting in a 30% reduction in raft sphingomyelin content. In addition, polyclonal activation-induced colocalization of PKCtheta; with lipid rafts was reduced by n-3 PUFA feeding. With respect to PKCtheta; effector pathway signaling, both AP-1 and NF-kappaB activation, IL-2 secretion, and lymphoproliferation were inhibited by fish oil feeding. Similar results were obtained when purified DHA was fed. These data demonstrate for the first time that dietary DHA alters T cell membrane microdomain composition and suppresses the PKCtheta; signaling axis.
Collapse
Affiliation(s)
- Yang-Yi Fan
- Molecular and Cell Biology Section, Faculty of Nutrition, Texas A&M University Health Science Center, College Station, TX 77843-2471, USA
| | | | | | | | | |
Collapse
|
16
|
Kew S, Wells S, Thies F, McNeill GP, Quinlan PT, Clark GT, Dombrowsky H, Postle AD, Calder PC. The Effect of Eicosapentaenoic Acid on Rat Lymphocyte Proliferation Depends Upon Its Position in Dietary Triacylglycerols. J Nutr 2003; 133:4230-8. [PMID: 14652377 DOI: 10.1093/jn/133.12.4230] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Animal and human studies have shown that greatly increasing the amount of fish oil [rich in long-chain (n-3) PUFA] in the diet can decrease lymphocyte functions. The effects of a more modest provision of long-chain (n-3) PUFA and whether eicosapentaenoic acid (20:5) and docosahexaenoic acid (22:6) have the same effects as one another are unclear. Whether the position of 20:5 or 22:6 in dietary triacylglycerols (TAG) influences their incorporation into immune cells and their subsequent functional effects is not known. In this study, male weanling rats were fed for 6 wk one of 9 diets that contained 178 g lipid/kg and that differed in the type of (n-3) PUFA and in the position of these in dietary TAG. The control diet contained 4.4 g alpha-linolenic acid (18:3)/100 g total fatty acids. In the other diets, 20:5 or 22:6 replaced a portion (50 or 100%) of 18:3, and were in the sn-2 or the sn-1(3) position of dietary TAG. There were significant dose-dependent increases in the proportion of 20:5 or 22:6 in spleen mononuclear cell phospholipids when 20:5 or 22:6 was fed. These increases were at the expense of arachidonic acid and were largely independent of the position of 20:5 or 22:6 in dietary TAG. Spleen lymphocyte proliferation increased dose dependently when 20:5 was fed in the sn-1(3) position of dietary TAG. There were no significant differences in interleukin-2, interferon-gamma or interleukin-10 production among spleen cells from rats fed the different diets. Prostaglandin E(2) production by spleen mononuclear cells was decreased by inclusion of either 20:5 or 22:6 in the diet in the sn-1(3) position. Thus, incorporation of 20:5 or 22:6 into spleen mononuclear cell phospholipids is not influenced by the position in dietary TAG. However, the pattern of incorporation may be influenced, and there are some differential functional effects of the position of long-chain (n-3) PUFA in dietary TAG. A moderate increase in the intake of 20:5 at the sn-1(3) position of dietary TAG increases lymphocyte proliferation.
Collapse
Affiliation(s)
- S Kew
- Institute of Human Nutrition, Division of Infection, Inflammation and Repair, University of Southampton, Southampton, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Fan YY, McMurray DN, Ly LH, Chapkin RS. Dietary (n-3) polyunsaturated fatty acids remodel mouse T-cell lipid rafts. J Nutr 2003; 133:1913-20. [PMID: 12771339 DOI: 10.1093/jn/133.6.1913] [Citation(s) in RCA: 178] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
In vitro evidence indicates that (n-3) polyunsaturated fatty acids (PUFA) suppress T-cell activation in part by displacing proteins from lipid rafts, specialized regions within the plasma membrane that play an important role in T-cell signal transduction. However, the ability of (n-3) PUFA to influence membrane microdomains in vivo has not been examined to date. Therefore, we compared the effect of dietary (n-3) PUFA on raft (liquid ordered) vs. soluble (liquid disordered) microdomain phospholipid composition in mouse T cells. Mice were fed diets containing either 5 g/100 g corn oil (control) or 4 g/100 g fish oil [contains (n-3) PUFA] + 1 g/100 g corn oil for 14 d. Splenic T-cell lipid rafts were isolated by density gradient centrifugation. Raft sphingomyelin content (mol/100 mol) was decreased (P < 0.05) in T cells isolated from (n-3) PUFA-fed mice. Dietary (n-3) PUFA were selectively incorporated into T-cell raft and soluble membrane phospholipids. Phosphatidylserine and glycerophosphoethanolamine, which are highly localized to the inner cytoplasmic leaflet, were enriched to a greater extent with unsaturated fatty acids compared with sphingomyelin, phosphatidylinositol and glycerophosphocholine. These data indicate for the first time that dietary (n-3) PUFA differentially modulate T-cell raft and soluble membrane phospholipid and fatty acyl composition.
Collapse
Affiliation(s)
- Yang-Yi Fan
- Faculty of Nutrition, Center for Environmental and Rural Health, Texas A&M University System, Health Science Center, College Station, TX, USA
| | | | | | | |
Collapse
|
18
|
Field CJ, Johnson IR, Schley PD. Nutrients and their role in host resistance to infection. J Leukoc Biol 2002. [DOI: 10.1189/jlb.71.1.16] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Affiliation(s)
- Catherine J. Field
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Canada
| | - Ian R. Johnson
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Canada
| | - Patricia D. Schley
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Canada
| |
Collapse
|
19
|
Field CJ, Clandinin MT, Van Aerde JE. Polyunsaturated fatty acids and T-cell function: implications for the neonate. Lipids 2001; 36:1025-32. [PMID: 11724454 DOI: 10.1007/s11745-001-0813-6] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Infant survival depends on the ability to respond effectively and appropriately to environmental challenges. Infants are born with a degree of immunological immaturity that renders them susceptible to infection and abnormal dietary responses (allergies). T-lymphocyte function is poorly developed at birth. The reduced ability of infants to respond to mitogens may be the result of the low number of CD45RO+ (memory/antigen-primed) T cells in the infant or the limited ability to produce cytokines [particularly interferon-y, interleukin (IL)-4, and IL-10. There have been many important changes in optimizing breast milk substitutes for infants; however, few have been directed at replacing factors in breast milk that convey immune benefits. Recent research has been directed at the neurological, retinal, and membrane benefits of adding 20:4n-6 (arachidonic acid; AA) and 22:6n-3 (docosahexaenoic acid; DHA) to infant formula. In adults and animals, feeding DHA affects T-cell function. However, the effect of these lipids on the development and function of the infant's immune system is not known. We recently reported the effect of adding DHA + AA to a standard infant formula on several functional indices of immune development. Compared with standard formula, feeding a formula containing DHA + AA increased the proportion of antigen mature (CD45RO+) CD4+ cells, improved IL-10 production, and reduced IL-2 production to levels not different from those of human milk-fed infants. This review will briefly describe T-cell development and the potential immune effect of feeding long-chain polyunsaturated fatty acids to the neonate.
Collapse
Affiliation(s)
- C J Field
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Canada.
| | | | | |
Collapse
|
20
|
Arrington JL, McMurray DN, Switzer KC, Fan YY, Chapkin RS. Docosahexaenoic acid suppresses function of the CD28 costimulatory membrane receptor in primary murine and Jurkat T cells. J Nutr 2001; 131:1147-53. [PMID: 11285317 DOI: 10.1093/jn/131.4.1147] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
(n-3) polyunsaturated fatty acids (PUFA) have been widely documented to reduce inflammation in diseases such as rheumatoid arthritis. This study sought to elucidate the mechanism whereby (n-3) PUFA downregulate T-cell proliferation. We hypothesized that membrane incorporation of dietary PUFA would alter membrane structure and consequently membrane receptor function. Female C57BL/6 mice were fed for 14 d one of three diets containing arachidonic acid (AA), fish oil or docosahexaenoic acid (DHA) that varied in lipid composition only. Spleens were harvested and T cells ( approximately 90% purity) were activated with agonists that stimulated proliferation at the receptor level [anti-CD3 (alphaCD3)/anti-CD28 (alphaCD28)], intracellularly [phorbol-12-myristate-13-acetate (PMA)/ionomycin] or with a combined receptor/intracellular agonist (alphaCD3/PMA). Although there was no significant difference (P > 0.05) in proliferative response across dietary groups within each agonist set, interleukin (IL)-2 secretion was significantly reduced (P = 0.05) in cells from DHA-fed mice stimulated with alphaCD3/alphaCD28. In parallel in vitro experiments, Jurkat T cells were incubated with 50 micromol/L linoleic acid, AA, or DHA. Similar agonists sets were employed, and cells incubated with DHA and AA had a significantly reduced (P < 0.05) IL-2 secretion in three of the agonist sets. However, only when the CD28 receptor was stimulated was there a significant difference (P < 0.05) between DHA and AA. The results of this study suggest the involvement of the CD28 receptor in reducing IL-2 secretion in DHA-fed mice and DHA-incubated Jurkat cells and that purified T cells from DHA-fed mice require accessory cells to modulate proliferative suppression.
Collapse
Affiliation(s)
- J L Arrington
- Faculty of Nutrition, Texas A&M University, College Station, TX, USA
| | | | | | | | | |
Collapse
|
21
|
Field CJ, Thomson CA, Van Aerde JE, Parrott A, Euler A, Lien E, Clandinin MT. Lower proportion of CD45R0+ cells and deficient interleukin-10 production by formula-fed infants, compared with human-fed, is corrected with supplementation of long-chain polyunsaturated fatty acids. J Pediatr Gastroenterol Nutr 2000; 31:291-9. [PMID: 10997375 DOI: 10.1097/00005176-200009000-00017] [Citation(s) in RCA: 118] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
BACKGROUND The immune consequences of adding 20:4n-6 and 22:6n-3 fatty acids to preterm infant formula are not known. METHODS The effect of feeding preterm infants (14-42 days of age) human milk (Human Milk group), infant formula (Formula group), or formula with added long-chain polyunsaturated fatty acids 20:4n-6 and 22:6n-3 (Formula + LCP group) on isolated peripheral blood lymphocytes (by flow cytometry) and lipid composition (by gas-liquid chromatography) was determined. Lymphocytes were stimulated in vitro with phytohemagglutinin to measure soluble interleukin (sIL)-2R and IL-10 production (by enzyme-linked immunosorbent assay). RESULTS With age, the percentage of CD3+ CD4+ T cells and the percentage of CD20+ cells increased in the Human Milk and Formula + LCP groups (P < 0.05), but not in the unsupplemented Formula group. Compared with the Formula group, CD4+ cells from the Formula + LCP and Human Milk groups expressed more CD45R0 (antigen mature) and less CD45RA (antigen naive) at 42 days of age (P < 0.05). At 42 days, IL-10 production was lower (P < 0.05) in cells of the Formula group than in cells of the Human Milk group. Production of IL-10 by the cells of the Formula + LCP group was not different from that produced by the Human Milk group cells. An age-related decrease (P < 0.05) in sIL-2R production by Formula + LCP lymphocytes was observed, but sIL-2R production at 42 days in the Formula + LCP group did not differ significantly from that in the Human Milk group. Compared with Formula alone, adding LCP to formula resulted in a lower C18:2n-6 and higher C20:4n-6 content in lymphocyte phospholipids (P < 0.05). CONCLUSIONS Adding LCP to a preterm infant formula resulted in lymphocyte populations, phospholipid composition, cytokine production, and antigen maturity that are more consistent with that in human milk-fed infants. This may affect the ability of the infant to respond to immune challenges.
Collapse
Affiliation(s)
- C J Field
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Canada
| | | | | | | | | | | | | |
Collapse
|
22
|
McMurray DN, Jolly CA, Chapkin RS. Effects of dietary n-3 fatty acids on T cell activation and T cell receptor-mediated signaling in a murine model. J Infect Dis 2000; 182 Suppl 1:S103-7. [PMID: 10944491 DOI: 10.1086/315909] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
A short-term feeding paradigm in mice, with diets enriched with eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), was used to study the modulation of T cell activation via the T cell receptor (TcR) and the downstream pathways of intracellular signaling. Diets enriched in EPA and DHA suppressed antigen-specific delayed hypersensitivity reactions and mitogen-induced proliferation of T cells. Cocultures of accessory cells and T cells from mice given different diets revealed that purified fatty acid ethyl esters acted directly on the T cell, rather than through the accessory cell. The loss of proliferative capacity was accompanied by reductions in interleukin (IL)-2 secretion and IL-2 receptor alpha chain mRNA transcription, suggesting that dietary EPA and DHA act, in part, by interrupting the autocrine IL-2 activation pathway. Dietary EPA and DHA blunted the production of intracellular second messengers, including diacylglycerol and ceramide, following mitogen stimulation in vitro. Dietary effects appear to vary with the agonist employed (i.e., anti-CD3 [TcR], anti-CD28, exogenous IL-2, or phorbol myristate acetate and ionomycin).
Collapse
Affiliation(s)
- D N McMurray
- Department of Medical Microbiology and Immunology, Texas A&M University System Health Science Center, College Station, TX 77843-1114, USA.
| | | | | |
Collapse
|
23
|
Moussa M, Tkaczuk J, Ragab J, Garcia J, Abbal M, Ohayon E, Ghisolfi J, Thouvenot JP. Relationship between the fatty acid composition of rat lymphocytes and immune functions. Br J Nutr 2000; 83:327-33. [PMID: 10884721 DOI: 10.1017/s0007114500000404] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The effects of dietary lipids on the fatty acid composition, activation and proliferation of lymphocytes were investigated. Weanling male Wistar rats were fed for 8 weeks on one of two low-fat diets which contained 50 g lipid/kg, or one of two high-fat diets containing 200 g lipid/kg, from either coconut oil or soyabean oil. The fatty acid composition of phospholipids from splenocyte membranes was affected by dietary lipid manipulation, and these differences influenced lymphocyte functions. Increased levels of linoleic acid in spleen lymphocytes correlated negatively with interleukin-2 receptor alpha-chain expression determined either by measuring the mean fluorescence or by the proportion of cells staining positive for CD25, and with the cell proliferation index. However, we found a positive correlation between interleukin-2 receptor alpha-chain expression determined by measuring the mean fluorescence and the cell proliferation index with the oleic acid concentration of spleen lymphocytes. Since phospholipid hydrolysis occurs early in lymphocyte activation, immunosuppressive effects induced by polyunsaturated fatty acids, described in the literature, could be due to an increase of linoleic acid or a decrease of oleic acid affecting many components of plasma-membrane-associated events involved in lymphocyte activation.
Collapse
Affiliation(s)
- M Moussa
- Group d'Etudes en Nutrition Infantile, Laboratoire de Biochimie, CHU Purpan, Toulouse, France.
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Furukawa K, Tashiro T, Yamamori H, Takagi K, Morishima Y, Sugiura T, Otsubo Y, Hayashi N, Itabashi T, Sano W, Toyoda Y, Nitta H, Nakajima N. Effects of soybean oil emulsion and eicosapentaenoic acid on stress response and immune function after a severely stressful operation. Ann Surg 1999; 229:255-61. [PMID: 10024108 PMCID: PMC1191639 DOI: 10.1097/00000658-199902000-00014] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVE To investigate the effects of soybean oil emulsion and oral or enteral administration of eicosapentaenoic acid (EPA) on stress response, cytokine production, protein metabolism, and immune function after surgery for esophageal cancer. SUMMARY BACKGROUND DATA It has been reported that safflower oil, rich in n-6 polyunsaturated fatty acid (n-6 PUFA), affects the survival rate of septic animals and decreases the immune function. It has also been reported that the administration of fish oil, in contrast, reduces these stress responses and stress-induced immunosuppression. In humans, the effects of soybean oil emulsion and the administration of EPA on stress response and immune function after surgery have not been established. METHODS Patients who underwent esophagectomy with thoracotomy were divided into three groups. Seven patients were fed by total parenteral nutrition (TPN) with soybean oil emulsion, which accounted for 20% of total calories. Seven patients were given oral or enteral administration of 1.8 g/day EPA, in addition to TPN with soybean oil emulsion. Nine patients served as the control group; these patients received fat-free TPN. Serum interleukin-6 (IL-6), C-reactive protein, concanavalin A (con A)- or phytohemagglutinin (PHA)-stimulated lymphocyte proliferation, natural killer cell activity, and stress hormones were measured. RESULTS The postoperative level of serum IL-6 was significantly higher in the group receiving soybean oil emulsion than in the fat-free group. Oral or enteral supplementation of EPA with soybean oil emulsion significantly reduced the level of serum IL-6 compared with the patients receiving soybean oil emulsion. Con A- or PHA-stimulated lymphocyte proliferation decreased significantly on postoperative day 7 in all groups of patients. The supplementation of EPA with soybean oil emulsion significantly improved the lymphocyte proliferation and natural killer cell activity on postoperative day 21 compared with the group receiving soybean oil emulsion. CONCLUSIONS Soybean oil emulsion amplifies, and the supplementation of EPA reduces, the stress response and stress-induced immunosuppression.
Collapse
Affiliation(s)
- K Furukawa
- First Department of Surgery, Chiba University School of Medicine, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Fritsche KL, Byrge M, Feng C. Dietary omega-3 polyunsaturated fatty acids from fish oil reduce interleukin-12 and interferon-gamma production in mice. Immunol Lett 1999; 65:167-73. [PMID: 10065739 DOI: 10.1016/s0165-2478(98)00109-6] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The objective of this study was to investigate the impact of feeding mice a diet rich in n-3 polyunsaturated fatty acids (PUFA) from fish oil on the interleukin-12 (IL-12) and interferon-gamma (IFNgamma) production during the early stage of an infectious challenge with Listeria monocytogenes. Weanling female C3H/HeN mice were fed AIN-93G experimental diets containing 20%, by weight one of three fat sources: lard (low PUFA), soybean oil (n-6 PUFA) or a mixture (9:1) of menhaden fish oil and corn oil (n-3 PUFA). After 4 weeks, mice were injected intraperitoneally with 10(5) Listeria monocytogenes and the concentration of IL-12(p70) and IFNgamma in serum was determined 24 h post-infection by ELISA. IL-12p35, IL-12p40 mRNA, and IFNgamma mRNA in the spleen were quantified by RNase protection assay. The number of IFNgamma-producing cells in the spleen was determined by flow cytometry using an intracellular staining procedure. We found that n-3 PUFA-fed mice had lower levels of circulating IL-12 at 24 h post-infection than n-6 PUFA- or low PUFA-fed mice (9.7+/-3.4 pg/ml vs. 61.6+/-10.6, and 44.4+/-12.5 pg/ml, respectively; P=0.002, n = 10/trt). The level of IL-12 p35 mRNA did not significantly differ among dietary treatment groups. However, IL-12p40 mRNA was significantly lower in n-3 PUFA- and n-6 PUFA-fed mice compared to low-PUFA-fed mice. Further, the n-3 PUFA group also had the lowest circulating IFNgamma (4.4+/-1.8 ng/ml vs. 9.1+/-1.0, and 9.7+/-2.1 ng/ml, respectively; P = 0.007. n = 8-10/trt). The n-3 PUFA-fed mice had significantly lower IFNgamma mRNA in their spleens compared to the mice fed the other fat sources. In agreement with having lower circulating IFNgamma and lower splenic IFNgamma mRNA, n-3 PUFA-fed mice had a significantly lower percentage of IFNgamma-producing cells in their spleens compared with the n-6 PUFA-fed group (2.1+/-0.6 vs. 4.2+/-0.7%; P = 0.037, n = 10/trt). In summary, feeding mice a diet rich in n-3 PUFA from fish oil significantly lowered the production of both IL-12 and IFNgamma during the early phase of a Listeria infection.
Collapse
Affiliation(s)
- K L Fritsche
- Department of Animal Sciences, University of Missouri, Columbia 65211, USA.
| | | | | |
Collapse
|
26
|
Sasaki T, Kanke Y, Kudoh K, Misawa Y, Shimizu J, Takita T. Effects of dietary docosahexaenoic acid on surface molecules involved in T cell proliferation. BIOCHIMICA ET BIOPHYSICA ACTA 1999; 1436:519-30. [PMID: 9989281 DOI: 10.1016/s0005-2760(98)00162-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
It is known that n-3 polyunsaturated fatty acids (PUFA) such as docosahexaenoic acid (DHA) suppress immunity as compared with n-6 PUFA such as linoleic acid (LA), but the mechanism involved in this phenomenon is still unclear. The present study was designed to assess the effect of dietary DHA on the surface molecules involved in T cell proliferation. Weanling male C57BL/6 mice were divided into four dietary groups that were fed a 10% fat diet for 4 weeks varying in amounts of DHA and LA. As the dietary DHA concentration increased, the surface expression of CD4 and CD8 on splenic T cells decreased, while that of CD28 increased. The surface expression of CD3, however, was invariable in all dietary groups. DNA synthesis of splenic T cells, induced by CD3 crosslinkage with anti-CD3 epsilon monoclonal antibody in the presence of CD28-mediated costimulation, increased as the DHA concentration was elevated. These observations suggest that diets rich in DHA exert some of their immunomodulatory effects by a downregulation of surface expression of CD4 and CD8 and by an upregulation of CD28-mediated costimulatory signal.
Collapse
Affiliation(s)
- T Sasaki
- Department of Bioregulation Studies, Graduate School of Agriculture, Tokyo University of Agriculture, Japan.
| | | | | | | | | | | |
Collapse
|
27
|
Fernandes G, Troyer DA, Jolly CA. The effects of dietary lipids on gene expression and apoptosis. Proc Nutr Soc 1998; 57:543-50. [PMID: 10096114 DOI: 10.1079/pns19980080] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The beneficial effects of dietary FO with respect to autoimmune disease, CVD and some types of cancer are well established. Studies conducted over the last 10-15 years have established the potent effects of FO on gene expression in the previously mentioned diseases. The effects of dietary FO appear to be selective in nature, with the expression of individual genes simultaneously being increased, decreased or completely unaffected. In order to elucidate the molecular mechanism(s) involved, recent studies have focused on analysing the effects of the long-chain polyunsaturated n-3 fatty acids EPA and DHA which are highly enriched in FO and thought to be the primary mediators of its biological activity. Indeed, it has been found that EPA and DHA appear to both directly and indirectly modulate gene expression in vivo, depending on the gene examined. The direct effects of EPA and DHA are most probably mediated by their ability to bind to positive and/or negative regulatory transcription factors, while the indirect effects appear to be mediated through alterations in the generation of intracellular lipid second messengers (e.g. diacylglycerol and ceramide). Future studies need to be focused on further elucidation of the inter- and intracellular signalling events mediated by dietary n-3 fatty acids. Understanding the molecular mechanism(s) modified by dietary FO will ultimately lead to improved dietary strategies to aid in the prevention of autoimmune disease, CVD and/or certain types of cancer.
Collapse
Affiliation(s)
- G Fernandes
- Department of Medicine, University of Texas Health Science Center at San Antonio 78284-7874, USA.
| | | | | |
Collapse
|
28
|
Jeffery NM, Cortina M, Newsholme EA, Calder PC. Effects of variations in the proportions of saturated, monounsaturated and polyunsaturated fatty acids in the rat diet on spleen lymphocyte functions. Br J Nutr 1997; 77:805-23. [PMID: 9175999 DOI: 10.1079/bjn19970077] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
To obtain further information about the immunomodulatory effects of specific dietary fatty acids, weanling male rats were fed for 6 weeks on high-fat (178 g/kg) diets which differed according to the principal fatty acids present. The nine diets used differed in their contents of palmitic, oleic, linoleic and alpha-linolenic acids; as a result the total polyunsaturated fatty acid (PUFA) content and the PUFA:saturated fatty acid ratio varied (from 17.8 to 58.5 g/100 g fatty acids and from 0.28 to 5.56 respectively). The n-6 PUFA:n-3 PUFA ratio was kept constant in all diets at approximately 7.0. The fatty acid composition of the serum and of spleen lymphocytes were significantly influenced by that of the diet fed. The ex vivo proliferation of spleen lymphocytes decreased as the level of oleic acid in the diet increased. Spleen natural killer cell activity decreased as the oleic acid content of the diet increased and increased as the palmitic acid content of the diet increased. The extent of the effects of these fatty acids on lymphocyte functions was modified by the nature of the background fatty acid composition of the diet.
Collapse
Affiliation(s)
- N M Jeffery
- Department of Biochemistry, University of Oxford
| | | | | | | |
Collapse
|
29
|
Jolly CA, Jiang YH, Chapkin RS, McMurray DN. Dietary (n-3) polyunsaturated fatty acids suppress murine lymphoproliferation, interleukin-2 secretion, and the formation of diacylglycerol and ceramide. J Nutr 1997; 127:37-43. [PMID: 9040541 DOI: 10.1093/jn/127.1.37] [Citation(s) in RCA: 190] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Elucidation of the mechanism(s) by which dietary fish oil, enriched in eicosapentaenoic acid (EPA, 20:5(n-3)] and docosahexaenoic acid [DHA, 22:6(n-3)], suppresses the inflammatory process is essential in maximizing this potentially therapeutic effect. Murine T-lymphocyte function and signal transduction were examined in response to a low fat, short term diet enriched in highly purified EPA or DHA ethyl esters. For 10 d, mice were fed comparable diets containing either 3% safflower oil ethyl esters (SAF), 2% SAF + 1% arachidonic acid triglyceride (AA), 2% SAF + 1% EPA, or 2% SAF + 1% DHA. Concanavalin A-induced T-lymphocyte proliferation in splenocyte cultures was significantly suppressed by dietary EPA and DHA while AA had no effect relative to the SAF control. The suppressed proliferative response in EPA- and DHA-fed mice was preceded temporally by a significant reduction in IL-2 secretion. Kinetics of mitogen-induced diacyl-sn-glycerol (DAG) and ceramide production did not differ significantly between SAF and AA diet groups. In contrast, DAG production was significantly suppressed in EP- and DHA-fed mice relative to the SAF and AA groups. The reduced DAG mass was paralleled by reduced ceramide mass following EPA and DHA feeding compared to the SAF and AA groups. Thus, low dose, short term dietary exposure to highly purified EPA or DHA appears to suppress mitogen-induced T-lymphocyte proliferation by inhibiting IL-2 secretion, and these events are accompanied by reductions in the production of essential lipid second messengers, DAG and ceramide.
Collapse
Affiliation(s)
- C A Jolly
- Faculty of Nutrition, Molecular and Cell Biology Group, College of Medicine, Texas A&M University, College Station 77843, USA
| | | | | | | |
Collapse
|
30
|
Laurenz JC, Gunn JM, Jolly CA, Chapkin RS. Alteration of glycerolipid and sphingolipid-derived second messenger kinetics in ras transformed 3T3 cells. BIOCHIMICA ET BIOPHYSICA ACTA 1996; 1299:146-54. [PMID: 8555247 DOI: 10.1016/0005-2760(95)00202-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The effect of ras transformation (rasB fibroblasts) on basal and serum-stimulated diacylglycerol (DAG) composition and mass was examined over time with respect to changes in membrane phospholipid composition and ceramide mass. RasB cells vs. nontransformed control cells (rasD and NR6) had chronically elevated DAG levels (up to 240 min) following serum stimulation, indicating a defect in the recovery phase of the intracellular DAG pulse. Ras transformation also had a dramatic effect on DAG composition. Molecular species analysis revealed that DAG from unstimulated rasB cells was enriched in the delta 9 desaturase fatty acyl species (monoenoate 18:1(n - 7) and 18:1(n - 9)), and depleted in arachidonic acid (20:4(n - 6)). With the exception of glycerophosphoinositol (GPI), DAG remodeling paralleled the compositional alterations in individual phospholipid classes. Importantly, ras transformation altered the fatty acyl composition of sphingomyelin, a precursor to the ceramide second messenger. With the addition of serum, control cells (rasD) had a progressive increase in ceramide mass with levels approximately 5-fold higher by 240 min. In contrast, ceramide levels did not increase in rasB cells at either 4 or 240 min. These results demonstrate that ras-oncogene, in addition to its effects on DAG metabolism, can also abolish the cellular increase in ceramide mass in response to serum stimulation. Since DAG and ceramide may have opposing biological functions, the prolonged elevation of DAG and the suppression of ceramide levels would be consistent with an enhanced proliferative capacity.
Collapse
Affiliation(s)
- J C Laurenz
- Faculty of Nutrition, Texas A&M University, College Station 77843-2471, USA
| | | | | | | |
Collapse
|
31
|
Jolly CA, Laurenz JC, McMurray DN, Chapkin RS. Diacylglycerol and ceramide kinetics in primary cultures of activated T-lymphocytes. Immunol Lett 1996; 49:43-8. [PMID: 8964608 DOI: 10.1016/0165-2478(95)02486-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
T cell activation results in the generation of diacylglycerol (DAG), the physiological activator of protein kinase C. Recently, ceramide, a bioactive lipid intracellular second messenger, has been shown to play a positive role in T cell proliferation. Most studies examining mitogen induction of DAG and ceramide in T cells have been conducted in cell lines over short periods of time (0-30 min) relative to the 2-3-h time frame required for commitment to proliferation. Therefore, we examined T cell mitogen-induced DAG and ceramide kinetics under physiologically relevant conditions during the initial 2 h of culture. Freshly isolated murine splenic lymphocytes were stimulated with the T cell-specific mitogen, concanavalin A (Con A). Our results show that Con A induced a multiphasic DAG response with significant peaks in DAG mass occurring at 2, 20 and 120 min. Concomitantly, ceramide mass was significantly increased 2 min following Con A addition and remained elevated until 120 min. Addition of C8-ceramide (10 microM) to lymphocyte cultures significantly enhanced mitogen-induced proliferation. These results demonstrate that DAG is continuously produced by activated T lymphocytes in a multiphasic fashion, and that ceramide is a positive effector molecule with respect to murine T cell proliferation. These results establish a foundation for further examination of the relationship between DAG, ceramide and T cell activation.
Collapse
Affiliation(s)
- C A Jolly
- Faculty of Nutrition, Texas A and M University, College Station 77843, USA
| | | | | | | |
Collapse
|