151
|
Bassaganya-Riera J, Guri AJ, Lu P, Climent M, Carbo A, Sobral BW, Horne WT, Lewis SN, Bevan DR, Hontecillas R. Abscisic acid regulates inflammation via ligand-binding domain-independent activation of peroxisome proliferator-activated receptor gamma. J Biol Chem 2011; 286:2504-16. [PMID: 21088297 PMCID: PMC3024745 DOI: 10.1074/jbc.m110.160077] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2010] [Revised: 10/16/2010] [Indexed: 01/01/2023] Open
Abstract
Abscisic acid (ABA) has shown efficacy in the treatment of diabetes and inflammation; however, its molecular targets and the mechanisms of action underlying its immunomodulatory effects remain unclear. This study investigates the role of peroxisome proliferator-activated receptor γ (PPAR γ) and lanthionine synthetase C-like 2 (LANCL2) as molecular targets for ABA. We demonstrate that ABA increases PPAR γ reporter activity in RAW 264.7 macrophages and increases ppar γ expression in vivo, although it does not bind to the ligand-binding domain of PPAR γ. LANCL2 knockdown studies provide evidence that ABA-mediated activation of macrophage PPAR γ is dependent on lancl2 expression. Consistent with the association of LANCL2 with G proteins, we provide evidence that ABA increases cAMP accumulation in immune cells. ABA suppresses LPS-induced prostaglandin E(2) and MCP-1 production via a PPAR γ-dependent mechanism possibly involving activation of PPAR γ and suppression of NF-κB and nuclear factor of activated T cells. LPS challenge studies in PPAR γ-expressing and immune cell-specific PPAR γ null mice demonstrate that ABA down-regulates toll-like receptor 4 expression in macrophages and T cells in vivo through a PPAR γ-dependent mechanism. Global transcriptomic profiling and confirmatory quantitative RT-PCR suggest novel candidate targets and demonstrate that ABA treatment mitigates the effect of LPS on the expression of genes involved in inflammation, metabolism, and cell signaling, in part, through PPAR γ. In conclusion, ABA decreases LPS-mediated inflammation and regulates innate immune responses through a bifurcating pathway involving LANCL2 and an alternative, ligand-binding domain-independent mechanism of PPAR γ activation.
Collapse
Affiliation(s)
- Josep Bassaganya-Riera
- From the Nutritional Immunology and Molecular Medicine Group, Virginia Bioinformatics Institute and
| | - Amir J. Guri
- From the Nutritional Immunology and Molecular Medicine Group, Virginia Bioinformatics Institute and
| | - Pinyi Lu
- From the Nutritional Immunology and Molecular Medicine Group, Virginia Bioinformatics Institute and
| | - Montse Climent
- From the Nutritional Immunology and Molecular Medicine Group, Virginia Bioinformatics Institute and
| | - Adria Carbo
- From the Nutritional Immunology and Molecular Medicine Group, Virginia Bioinformatics Institute and
| | - Bruno W. Sobral
- From the Nutritional Immunology and Molecular Medicine Group, Virginia Bioinformatics Institute and
| | - William T. Horne
- From the Nutritional Immunology and Molecular Medicine Group, Virginia Bioinformatics Institute and
| | - Stephanie N. Lewis
- From the Nutritional Immunology and Molecular Medicine Group, Virginia Bioinformatics Institute and
- Department of Biochemistry, Virginia Polytechnic Institute and State University, Blacksburg, Virginia 24061
| | - David R. Bevan
- Department of Biochemistry, Virginia Polytechnic Institute and State University, Blacksburg, Virginia 24061
| | - Raquel Hontecillas
- From the Nutritional Immunology and Molecular Medicine Group, Virginia Bioinformatics Institute and
| |
Collapse
|
152
|
Jacques C, Levy E, Muckle G, Jacobson SW, Bastien C, Dewailly É, Ayotte P, Jacobson JL, Saint-Amour D. Long-term effects of prenatal omega-3 fatty acid intake on visual function in school-age children. J Pediatr 2011; 158:83-90, 90.e1. [PMID: 20797725 PMCID: PMC2992831 DOI: 10.1016/j.jpeds.2010.06.056] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2010] [Revised: 05/26/2010] [Accepted: 06/29/2010] [Indexed: 10/19/2022]
Abstract
OBJECTIVE To assess the long-term effect on visual development of omega-3 polyunsaturated fatty acid (n-3 PUFA) intake during gestation. STUDY DESIGN Using visual evoked potentials (VEPs), the long-term effects on visual development were evaluated in 136 school-age Inuit children exposed to high levels of n-3 PUFAs during gestation. VEP protocols using color and motion stimuli were used to assess parvocellular and magnocellular responses. Concentrations of the two major n-3 PUFAs (docosahexaenoic acid [DHA] and eicosapentaenoic acid [EPA]) were measured in umbilical cord and child plasma phospholipids, reflecting prenatal and postnatal exposure, respectively. RESULTS After adjustment for confounders, cord plasma DHA level was found to be associated with shorter latencies of the N1 and P1 components of the color VEPs. No effects were found for current n-3 PUFA body burden or motion-onset VEPs. CONCLUSION This study demonstrates beneficial effects of DHA intake during gestation on visual system function at school age. DHA is particularly important for the early development and long-term function of the visual parvocellular pathway.
Collapse
Affiliation(s)
- Caroline Jacques
- Centre de recherche, CHU Sainte-Justine, 3175, Chemin de la Côte Sainte-Catherine, Montréal, Québec, Canada, H3T 1C5
- Département de nutrition, Université de Montréal, Pavillon Liliane-de-Stewart, 2405, Chemin de la Côte-Sainte-Catherine, Montréal, Québec, Canada, 2405, H3T 1A8
| | - Emile Levy
- Centre de recherche, CHU Sainte-Justine, 3175, Chemin de la Côte Sainte-Catherine, Montréal, Québec, Canada, H3T 1C5
- Département de nutrition, Université de Montréal, Pavillon Liliane-de-Stewart, 2405, Chemin de la Côte-Sainte-Catherine, Montréal, Québec, Canada, 2405, H3T 1A8
| | - Gina Muckle
- École de psychologie, Université Laval, Québec, Canada, G1K 7P4
- Axe de recherche en santé des populations et environnementale, Centre de recherche du Centre Hospitalier Universitaire de Québec, 2875 boul. Laurier, Québec, Canada, G1V 2M2
| | - Sandra W. Jacobson
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, 2751 E. Jefferson, Suite 460, Detroit, Michigan, MI 48207, United States
| | - Célyne Bastien
- École de psychologie, Université Laval, Québec, Canada, G1K 7P4
- Laboratoire de neurosciences comportementales humaines, Centre de recherche Université Laval Robert-Giffard (CRULRG), Beauport, Québec, Canada
| | - Éric Dewailly
- Axe de recherche en santé des populations et environnementale, Centre de recherche du Centre Hospitalier Universitaire de Québec, 2875 boul. Laurier, Québec, Canada, G1V 2M2
| | - Pierre Ayotte
- Axe de recherche en santé des populations et environnementale, Centre de recherche du Centre Hospitalier Universitaire de Québec, 2875 boul. Laurier, Québec, Canada, G1V 2M2
| | - Joseph L. Jacobson
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, 2751 E. Jefferson, Suite 460, Detroit, Michigan, MI 48207, United States
| | - Dave Saint-Amour
- Centre de recherche, CHU Sainte-Justine, 3175, Chemin de la Côte Sainte-Catherine, Montréal, Québec, Canada, H3T 1C5
- Département d’ophtalmologie, CHU Sainte-Justine, 3175, Côte Sainte-Catherine, Montréal, Québec, Canada, H3T 1C5
- Département de psychologie, Université du Québec à Montréal, Case postale 8888, succursale Centre-ville, Montréal, Québec, H3C 3P8
| |
Collapse
|
153
|
Oster RT, Tishinsky JM, Yuan Z, Robinson LE. Docosahexaenoic acid increases cellular adiponectin mRNA and secreted adiponectin protein, as well as PPARγ mRNA, in 3T3-L1 adipocytes. Appl Physiol Nutr Metab 2010; 35:783-9. [DOI: 10.1139/h10-076] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Adiponectin, a protein secreted from adipose tissue, has been shown to have anti-diabetic and anti-inflammatory effects, but its regulation is not completely understood. Long-chain n-3 fatty acids eicosapentaenoic acid (20:5n-3; EPA) and docosahexaenoic acid (22:6n-3; DHA) may be involved in adiponectin regulation as they are potential ligands for peroxisome proliferator-activated receptor-γ (PPARγ), a key transcription factor for the adiponectin gene. To examine this, 3T3-L1 adipocytes were incubated with 125 µmol·L–1 EPA, DHA, palmitic, or oleic acids complexed to albumin, or with albumin alone (control) for 24 h. Adipocytes were also incubated for 24 h with EPA and DHA plus bisphenol-A-diglycidyl ether (BADGE), a PPARγ antagonist. Both EPA and DHA increased (p < 0.05) secreted adiponectin concentration compared with the control (44% and 102%, respectively), but did not affect cellular adiponectin protein content. Incubation with BADGE and DHA inhibited increases in secreted adiponectin protein, suggesting that DHA may act through a PPARγ-dependent mechanism. However, BADGE had no effect on EPA-induced increases in secreted adiponectin protein. Only DHA enhanced (p < 0.05) PPARγ and adiponectin mRNA expression compared wtih the control. Our results demonstrate that DHA increases cellular adiponectin mRNA and secreted adiponectin protein in 3T3-L1 adipocytes, possibly by a mechanism involving PPARγ. Moreover, DHA increased adiponectin concentration to a greater extent (40% more, p < 0.05) compared with EPA, emphasizing the need to consider the independent actions of EPA and DHA in adipocytes.
Collapse
Affiliation(s)
- Richard T. Oster
- Department of Human Health and Nutritional Sciences, Animal Science and Nutrition Building, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Justine M. Tishinsky
- Department of Human Health and Nutritional Sciences, Animal Science and Nutrition Building, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Zongfei Yuan
- Department of Human Health and Nutritional Sciences, Animal Science and Nutrition Building, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Lindsay E. Robinson
- Department of Human Health and Nutritional Sciences, Animal Science and Nutrition Building, University of Guelph, Guelph, ON N1G 2W1, Canada
| |
Collapse
|
154
|
García-Arcos I, González-Kother P, Aspichueta P, Rueda Y, Ochoa B, Fresnedo O. Lipid Analysis Reveals Quiescent and Regenerating Liver-Specific Populations of Lipid Droplets. Lipids 2010; 45:1101-8. [DOI: 10.1007/s11745-010-3492-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2010] [Accepted: 10/19/2010] [Indexed: 12/29/2022]
|
155
|
Vara Prasad SSS, Jeya Kumar SS, Kumar PU, Qadri SSYH, Vajreswari A. Dietary fatty acid composition alters 11β-hydroxysteroid dehydrogenase type 1 gene expression in rat retroperitoneal white adipose tissue. Lipids Health Dis 2010; 9:111. [PMID: 20932307 PMCID: PMC2959202 DOI: 10.1186/1476-511x-9-111] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2010] [Accepted: 10/08/2010] [Indexed: 11/26/2022] Open
Abstract
The enzyme 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) amplifies intracellular glucocorticoid action by converting inactive glucocorticoids to their active forms in vivo. Adipose-specific overexpression of 11β-HSD1 induces metabolic syndrome in mice, whereas 11β-HSD1 null mice are resistant to it. Dietary trans and saturated fatty acids (TFAs and SFAs) are involved in the development of metabolic syndrome, whereas polyunsaturated fatty acids (PUFA) offer protection against this. Here, we report the effects of chronic feeding of different diets containing vanaspati (TFA rich), palm oil (SFA rich) and sunflower oil (PUFA rich) at 10%level on 11β-HSD1 gene expression in rat retroperitoneal adipose tissue. 11β-HSD1 gene expression was significantly higher in TFA rich diet-fed rats compared to SFA rich diet-fed rats, which in turn was significantly higher than PUFA rich diet-fed rats. Similar trend was observed in the expression of CCAAT-enhancer binding protein-α (C/EBP-α), the main transcription factor required for the expression of 11β-HSD1. We propose that TFAs and SFAs increase local amplification of glucocorticoid action in adipose tissue by upregulating 11β-HSD1 by altering C/EBP-α-gene expression. The increased levels of glucocorticoids in adipose tissue may lead to development of obesity and insulin resistance, thereby increasing the risk of developing metabolic syndrome.
Collapse
Affiliation(s)
- Sakamuri S S Vara Prasad
- Department of Biochemistry, National Institute of Nutrition, Indian Council of Medical Research, Jamai Osmania PO, Hyderabad-500 604, Andhra Pradesh, India
| | | | | | | | | |
Collapse
|
156
|
Dietary Lecithin Source Affects Growth Potential and Gene Expression in Sparus aurata Larvae. Lipids 2010; 45:1011-23. [DOI: 10.1007/s11745-010-3471-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2010] [Accepted: 08/25/2010] [Indexed: 10/19/2022]
|
157
|
Bassaganya-Riera J, Hontecillas R. Dietary conjugated linoleic acid and n-3 polyunsaturated fatty acids in inflammatory bowel disease. Curr Opin Clin Nutr Metab Care 2010; 13:569-73. [PMID: 20508519 PMCID: PMC2947030 DOI: 10.1097/mco.0b013e32833b648e] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
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 conjugated linoleic acid (CLA) and n-3 polyunsaturated fatty acids (PUFAs) in IBD. RECENT FINDINGS Nutrition-based interventions that target peroxisome proliferator-activated receptors (PPARs) such as dietary CLA and 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 more than 0.65 via n-3 PUFA intervention. In mice, CLA prevented inflammation-driven colorectal cancer by activating PPAR gamma 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, multicenter clinical trials.
Collapse
Affiliation(s)
- Josep Bassaganya-Riera
- Laboratory of Nutritional Immunology and Molecular Nutrition, Virginia Bioinformatics Institute, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA.
| | | |
Collapse
|
158
|
Tu WC, Cook-Johnson RJ, James MJ, Mühlhäusler BS, Gibson RA. Omega-3 long chain fatty acid synthesis is regulated more by substrate levels than gene expression. Prostaglandins Leukot Essent Fatty Acids 2010; 83:61-8. [PMID: 20573490 DOI: 10.1016/j.plefa.2010.04.001] [Citation(s) in RCA: 126] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2010] [Revised: 04/28/2010] [Accepted: 04/28/2010] [Indexed: 10/19/2022]
Abstract
The conversion of linoleic acid (LA) and alpha-linolenic acid (ALA) to long chain polyunsaturated fatty acids (LCPUFA) is known to involve desaturation and elongation steps. Although there is evidence that genes for these steps can be regulated by extremes of dietary PUFA, the degree to which there is meaningful regulation of LCPUFA levels in tissues by diet as a result of changes in expression of desaturase and elongase genes is unclear. In this study, we tested the effect of increasing ALA levels in diets of rats from 0.2% to 2.9% energy (en) against a constant LA level (1%en) on plasma and liver phospholipid LCPUFA content together with the expression of hepatic genes involved in PUFA metabolism, the desaturases FADS1 and FADS2, the elongases ELOV2 and ELOV5, and the transcription factors sterol regulatory element-binding protein-1c (SREBP-1c) and peroxisome proliferator-activated receptor alpha (PPARalpha). The levels of plasma and liver eicosapentaenoic acid (EPA) and docosapentaenoic acid (DPA) increased in proportion to dietary ALA whereas docosahexaenoic acid (DHA) increased only up to 1%en ALA. A low PUFA (0.4%en) reference diet stimulated the expression of delta 6 desaturase (FADS2) and elongase 2 (ELOVL2) when compared to higher PUFA diets. There was, however, no difference in the expression of any of the genes in rats, which were fed diets containing between 0.2%en and 2.9%en ALA and mRNA expression was unrelated to tissue/plasma LCPUFA content. These data suggest that the endogenous synthesis of n-3 LCPUFA from the precursor ALA is regulated independently of changes in the expression of the synthetic enzymes or regulatory transcription factor, and provides evidence that n-3 LCPUFA synthesis is regulated more by substrate competition for existing enzymes than by an increase in their mRNA expression.
Collapse
Affiliation(s)
- W C Tu
- Foodplus Research Centre, School of Agriculture, Food and Wine, Waite Campus, University of Adelaide, Adelaide, South Australia, Australia
| | | | | | | | | |
Collapse
|
159
|
Perales S, Alejandre MJ, Morales RP, Torres C, Linares A. Fish oil supplementation reverses the effect of cholesterol on apoptotic gene expression in smooth muscle cells. Lipids Health Dis 2010; 9:70. [PMID: 20630092 PMCID: PMC2914009 DOI: 10.1186/1476-511x-9-70] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2010] [Accepted: 07/14/2010] [Indexed: 01/17/2023] Open
Abstract
Background Nutritional control of gene regulation guides the transformation of smooth muscle cells (SMC) into foam cells in atherosclerosis. Oxidative stress has been reported in areas of lipid accumulation, activating proliferation genes. Suppression of oxidative stress by antioxidant administration reduces this activation and the progression of lesions. We hypothesized that fish oil consumption may protect against atherosclerotic vascular disease. The study objective was to determine the effects of dietary cholesterol and fish-oil intake on the apoptotic pathways induced by 25-hydroxycholesterol (25-HC) in SMC cultures. Methods An in vivo/in vitro cell model was used, culturing SMC isolated from chicks exposed to an atherogenic cholesterol-rich diet with 5% of cholesterol (SMC-Ch) alone or followed by an anti-atherogenic fish oil-rich diet with 10% of menhaden oil (SMC-Ch-FO) and from chicks on standard diet (SMC-C). Cells were exposed to 25-HC, studying apoptosis levels by flow cytometry (Annexin V) and expressions of caspase-3, c-myc, and p53 genes by quantitative real-time reverse transcriptase-polymerase chain reaction. Results: Exposure to 25-HC produced apoptosis in all three SMC cultures, which was mediated by increases in caspase-3, c-myc, and p53 gene expression. Changes were more marked in SMC-Ch than in SMC-C, indicating that dietary cholesterol makes SMC more susceptible to 25-HC-mediated apoptosis. Expression of p53 gene was elevated in SMC-Ch-FO. This supports the proposition that endogenous levels of p53 protect SMC against apoptosis and possibly against the development of atherosclerosis. Fish oil attenuated the increase in c-myc levels observed in SMC-C and SMC-Ch, possibly through its influence on the expression of antioxidant genes. Conclusion Replacement of a cholesterol-rich diet with a fish oil-rich diet produces some reversal of the cholesterol-induced changes, increasing the resistance of SMC to apoptosis.
Collapse
Affiliation(s)
- Sonia Perales
- Department of Biochemistry and Molecular Biology I, Faculty of Sciences, Campus Universitario de Fuentenueva Avenida Severo Ochoa s/n 18071 University of Granada, Spain
| | | | | | | | | |
Collapse
|
160
|
Ramsden C, Gagnon C, Graciosa J, Faurot K, David R, Bralley JA, Harden RN. Do omega-6 and trans fatty acids play a role in complex regional pain syndrome? A pilot study. PAIN MEDICINE (MALDEN, MASS.) 2010; 11:1115-25. [PMID: 20545870 PMCID: PMC9442504 DOI: 10.1111/j.1526-4637.2010.00882.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
OBJECTIVES The study aims to compare the omega-6 (n-6) and omega-3 (n-3) highly unsaturated fatty acids (HUFA), and trans fatty acid (trans FA) status of Complex Regional Pain Syndrome (CRPS) patients to pain-free controls. DESIGN Case control study. Setting. The setting was at a multidisciplinary rehabilitation center. PATIENTS Twenty patients that met the Budapest research diagnostic criteria for CRPS and 15 pain-free control subjects were included in this study. Outcome Measures. Fasting plasma fatty acids were collected from all participants. In CRPS patients, pain was assessed using the McGill Pain Questionnaire-Short Form. In addition, results from the perceived disability (Pain Disability Index), pain-related anxiety (Pain Anxiety Symptom Scale Short Form), depression (Center for Epidemiologic Studies Depression Scale Short Form), and quality of life (Short Form-36 [SF-36]) were evaluated. RESULTS Compared with controls, CRPS patients demonstrated elevated concentrations of n-6 HUFA and trans FA. No differences in n-3 HUFA concentrations were observed. Plasma concentrations of the n-6 HUFA docosatetraenoic acid were inversely correlated with the "vitality" section of the SF-36. Trans FA concentrations positively correlated with pain-related disability and anxiety. CONCLUSION These pilot data suggest that elevated n-6 HUFA and trans FA may play a role in CRPS pathogenesis. These findings should be replicated, and more research is needed to explore the clinical significance of low n-6 and trans FA diets with or without concurrent n-3 HUFA supplementation, for the management of CRPS.
Collapse
Affiliation(s)
- Christopher Ramsden
- Rehabilitation Institute of Chicago, Department of Physical Medicine and Rehabilitation, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
| | | | | | | | | | | | | |
Collapse
|
161
|
Abstract
Obesity leads to several chronic morbidities including type 2 diabetes, dyslipidaemia, atherosclerosis and hypertension, which are major components of the metabolic syndrome. White adipose tissue (WAT) metabolism and WAT-derived factors (fatty acids and adipokines) play an important role in the development of these metabolic disturbances. In fact, dysregulated adipokine secretion from the expanded WAT of obese individuals contributes to the development of systemic low-grade inflammation, insulin resistance and metabolic syndrome. The n-3 PUFA EPA and DHA have been widely reported to have protective effects in a range of chronic inflammatory conditions including obesity. In fact, n-3 PUFA have been shown to ameliorate low-grade inflammation in adipose tissue associated with obesity and up-regulate mitochondrial biogenesis and induce beta-oxidation in WAT in mice. Moreover, the ability of n-3 PUFA to regulate adipokine gene expression and secretion has been observed both in vitro and in vivo in rodents and human subjects. The present article reviews: (1) the physiological role of adiponectin, leptin and pre-B cell colony-enhancer factor/visfatin, three adipokines with immune-modulatory properties involved in the regulation of metabolism and insulin sensitivity and (2) the actions of n-3 PUFA on these adipokines focusing on the underlying mechanisms and the potential relationship with the beneficial effects of these fatty acids on obesity-associated metabolic disorders. It can be concluded that the ability of n-3 PUFA to improve obesity and insulin resistance conditions partially results from the modulation of WAT metabolism and the secretion of bioactive adipokines including leptin, adiponectin and visfatin.
Collapse
|
162
|
Chutkow WA, Birkenfeld AL, Brown JD, Lee HY, Frederick DW, Yoshioka J, Patwari P, Kursawe R, Cushman SW, Plutzky J, Shulman GI, Samuel VT, Lee RT. Deletion of the alpha-arrestin protein Txnip in mice promotes adiposity and adipogenesis while preserving insulin sensitivity. Diabetes 2010; 59:1424-34. [PMID: 20299477 PMCID: PMC2874703 DOI: 10.2337/db09-1212] [Citation(s) in RCA: 116] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
OBJECTIVE Thioredoxin interacting protein (Txnip), a regulator of cellular oxidative stress, is induced by hyperglycemia and inhibits glucose uptake into fat and muscle, suggesting a role for Txnip in type 2 diabetes pathogenesis. Here, we tested the hypothesis that Txnip-null (knockout) mice are protected from insulin resistance induced by a high-fat diet. RESEARCH DESIGN AND METHODS Txnip gene-deleted (knockout) mice and age-matched wild-type littermate control mice were maintained on a standard chow diet or subjected to 4 weeks of high-fat feeding. Mice were assessed for body composition, fat development, energy balance, and insulin responsiveness. Adipogenesis was measured from ex vivo fat preparations, and in mouse embryonic fibroblasts (MEFs) and 3T3-L1 preadipocytes after forced manipulation of Txnip expression. RESULTS Txnip knockout mice gained significantly more adipose mass than controls due to a primary increase in both calorie consumption and adipogenesis. Despite increased fat mass, Txnip knockout mice were markedly more insulin sensitive than controls, and augmented glucose transport was identified in both adipose and skeletal muscle. RNA interference gene-silenced preadipocytes and Txnip(-/-) MEFs were markedly adipogenic, whereas Txnip overexpression impaired adipocyte differentiation. As increased adipogenesis and insulin sensitivity suggested aspects of augmented peroxisome proliferator-activated receptor-gamma (PPARgamma) response, we investigated Txnip's regulation of PPARgamma function; manipulation of Txnip expression directly regulated PPARgamma expression and activity. CONCLUSIONS Txnip deletion promotes adiposity in the face of high-fat caloric excess; however, loss of this alpha-arrestin protein simultaneously enhances insulin responsiveness in fat and skeletal muscle, revealing Txnip as a novel mediator of insulin resistance and a regulator of adipogenesis.
Collapse
Affiliation(s)
- William A. Chutkow
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Cambridge, Massachusetts
- Veterans Administration Medical Center, West Roxbury, Massachusetts
| | - Andreas L. Birkenfeld
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Jonathan D. Brown
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Cambridge, Massachusetts
- Veterans Administration Medical Center, West Roxbury, Massachusetts
| | - Hui-Young Lee
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - David W. Frederick
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Jun Yoshioka
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Cambridge, Massachusetts
| | - Parth Patwari
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Cambridge, Massachusetts
| | - Romy Kursawe
- Department of Pediatrics, Yale University School of Medicine, New Haven, Connecticut
| | - Samuel W. Cushman
- Experimental Diabetes, Metabolism, and Nutrition Section, National Institute of Diabetes and Digestive and Kidney Disease, Bethesda, Maryland
| | - Jorge Plutzky
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Cambridge, Massachusetts
| | - Gerald I. Shulman
- Howard Hughes Medical Institute and Departments of Internal Medicine and Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut; and
| | - Varman T. Samuel
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
- Veterans Administration Medical Center, West Haven, Connecticut
- Corresponding author: Varman T. Samuel,
| | - Richard T. Lee
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Cambridge, Massachusetts
| |
Collapse
|
163
|
Lee JY, Zhao L, Hwang DH. Modulation of pattern recognition receptor-mediated inflammation and risk of chronic diseases by dietary fatty acids. Nutr Rev 2010; 68:38-61. [PMID: 20041999 DOI: 10.1111/j.1753-4887.2009.00259.x] [Citation(s) in RCA: 106] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Chronic inflammation is known to promote the development of many chronic diseases. Pattern recognition receptors (PRRs), Toll-like receptors (TLRs), and nucleotide-binding oligomerization domain proteins (NODs) mediate both infection-induced inflammation and sterile inflammation by recognizing pathogen- associated molecular patterns and endogenous molecules, respectively. PRR-mediated inflammation is an important determinant in altering the risk of many chronic diseases. Saturated fatty acids (SFAs) can activate PRRs, leading to enhanced expression of pro-inflammatory target gene products. However, n-3 polyunsaturated fatty acids (PUFAs) inhibit agonist-induced activation of PRRs. These results suggest that SFAs and n-3 PUFAs can reciprocally modulate PRR-mediated inflammation, and that PRRs and their downstream signaling components are molecular targets for dietary strategies to reduce chronic inflammation and subsequent risk of chronic diseases. This advancement in knowledge provides a new paradigm for understanding the mechanism by which different dietary fatty acids modify risk of chronic diseases including insulin resistance, atherosclerosis, and cancer.
Collapse
Affiliation(s)
- Joo Y Lee
- Western Human Nutrition Research Center, ARS, USDA and Department of Nutrition, University of California, Davis, CA, USA
| | | | | |
Collapse
|
164
|
The role of inflammation and macrophage accumulation in the development of obesity-induced type 2 diabetes mellitus and the possible therapeutic effects of long-chain n-3 PUFA. Proc Nutr Soc 2010; 69:232-43. [PMID: 20158940 DOI: 10.1017/s0029665110000042] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The WHO estimate that >1 x 10(6) deaths in Europe annually can be attributed to diseases related to excess body weight, and with the rising global obesity levels this death rate is set to drastically increase. Obesity plays a central role in the metabolic syndrome, a state of insulin resistance that predisposes patients to the development of CVD and type 2 diabetes mellitus. Obesity is associated with low-grade chronic inflammation characterised by inflamed adipose tissue with increased macrophage infiltration. This inflammation is now widely believed to be the key link between obesity and development of insulin resistance. In recent years it has been established that activation of pro-inflammatory pathways can cross talk with insulin signalling pathways via a number of mechanisms including (a) down-regulation of insulin signalling pathway proteins (e.g. GLUT4 and insulin receptor substrate (IRS)-1), (b) serine phosphorylation of IRS-1 blocking its tyrosine phosphorylation in response to insulin and (c) induction of cytokine signalling molecules that sterically hinder insulin signalling by blocking coupling of the insulin receptor to IRS-1. Long-chain (LC) n-3 PUFA regulate gene expression (a) through transcription factors such as PPAR and NF-kappaB and (b) via eicosanoid production, reducing pro-inflammatory cytokine production from many different cells including the macrophage. LC n-3 PUFA may therefore offer a useful anti-inflammatory strategy to decrease obesity-induced insulin resistance, which will be examined in the present review.
Collapse
|
165
|
A high-fat diet induces lower expression of retinoid receptors and their target genes GAP-43/neuromodulin and RC3/neurogranin in the rat brain. Br J Nutr 2010; 103:1720-9. [PMID: 20102671 DOI: 10.1017/s0007114509993886] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Numerous studies have reported an association between cognitive impairment in old age and nutritional factors, including dietary fat. Retinoic acid (RA) plays a central role in the maintenance of cognitive processes via its nuclear receptors (NR), retinoic acid receptor (RAR) and retinoid X receptor (RXR), and the control of target genes, e.g. the synaptic plasticity markers GAP-43/neuromodulin and RC3/neurogranin. Given the relationship between RA and the fatty acid signalling pathways mediated by their respective NR (RAR/RXR and PPAR), we investigated the effect of a high-fat diet (HFD) on (1) PUFA status in the plasma and brain, and (2) the expression of RA and fatty acid NR (RARbeta, RXRbetagamma and PPARdelta), and synaptic plasticity genes (GAP-43 and RC3), in young male Wistar rats. In the striatum of rats given a HFD for 8 weeks, real-time PCR (RT-PCR) revealed a decrease in mRNA levels of RARbeta ( - 14 %) and PPARdelta ( - 13 %) along with an increase in RXRbetagamma (+52 %). Concomitantly, RT-PCR and Western blot analysis revealed (1) a clear reduction in striatal mRNA and protein levels of RC3 ( - 24 and - 26 %, respectively) and GAP-43 ( - 10 and - 42 %, respectively), which was confirmed by in situ hybridisation, and (2) decreased hippocampal RC3 and GAP-43 protein levels (approximately 25 %). Additionally, HFD rats exhibited a significant decrease in plasma ( - 59 %) and brain ( - 6 %) n-3 PUFA content, mainly due to the loss of DHA. These results suggest that dietary fat induces neurobiological alterations by modulating the brain RA signalling pathway and n-3 PUFA content, which have been previously correlated with cognitive impairment.
Collapse
|
166
|
Effect of dietary fatty acids on expression of lipogenic enzymes and fatty acid profile in tissues of bulls. Animal 2010; 4:755-62. [DOI: 10.1017/s1751731110000431] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
|
167
|
Perales S, Alejandre MJ, Palomino-Morales R, Torres C, Linares A. Influence of cholesterol and fish oil dietary intake on nitric oxide-induced apoptosis in vascular smooth muscle cells. Nitric Oxide 2009; 22:205-12. [PMID: 20040380 DOI: 10.1016/j.niox.2009.12.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2009] [Revised: 12/12/2009] [Accepted: 12/20/2009] [Indexed: 01/04/2023]
Abstract
Apoptosis of vascular smooth muscle cells (SMC) is critically involved in the progression of atherosclerosis. We previously reported that dietary cholesterol intake induces changes in SMC at molecular and gene expression levels. The objectives of the present study were to investigate the differential response to nitric oxide of vascular SMC obtained from chicks after cholesterol and fish oil dietary intake and to examine effects on the main pro-apoptotic and anti-apoptotic genes. Dietary cholesterol intake reduced the Bcl-2/Bax (anti-apoptotic/pro-apoptotic) protein ratio in SMC, making them more susceptible to apoptosis. When cholesterol was withdrawn and replaced with a fish oil-enriched diet, the Bcl-xl/Bax protein ratio significantly increased, reversing the changes induced by cholesterol. The decrease in c-myc gene expression after apoptotic stimuli and the increase in Bcl-xl/Bax ratio indicate that fish oil has a protective role against apoptosis in SMC. Nitroprussiate-like nitric oxide donors exerted an intensive action on vascular SMC cultures. However, SMC-C (isolated from animals fed with control diet) and SMC-Ch (isolated from animals fed with cholesterol-enriched diet) responded differently to nitric oxide, especially in their bcl-2 and bcl-xl gene expression. SMC isolated from animals fed with cholesterol-enriched and then fish oil-enriched diet (SMC-Ch-FO cultures) showed an intermediate apoptosis level (Bcl-2/Bax ratio) between SMC-C and SMC-Ch, induction of c-myc expression and elevated p53 expression. These findings indicate that fish oil protects SMC against apoptosis.
Collapse
Affiliation(s)
- Sonia Perales
- Department of Biochemistry and Molecular Biology, Faculty of Sciences, University of Granada, Granada, Spain
| | | | | | | | | |
Collapse
|
168
|
|
169
|
Tulk HMF, Robinson LE. Modifying the n-6/n-3 polyunsaturated fatty acid ratio of a high-saturated fat challenge does not acutely attenuate postprandial changes in inflammatory markers in men with metabolic syndrome. Metabolism 2009; 58:1709-16. [PMID: 19625064 DOI: 10.1016/j.metabol.2009.05.031] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2008] [Accepted: 05/20/2009] [Indexed: 01/01/2023]
Abstract
Metabolic syndrome (MetS) features chronic inflammation and exaggerated postprandial triacylglyceride (TAG) responses. Fasting concentrations of interleukin-6 (IL-6) and C-reactive protein (CRP), key inflammatory mediators, decrease after sustained n-3 polyunsaturated fatty acid (PUFA) intake; however, the ability of n-3 PUFA to attenuate postprandial inflammatory responses is not well studied. Thus, we examined the acute effect of modifying the n-6/n-3 PUFA ratio of a high-saturated fatty acid (SFA) oral fat tolerance test (OFTT) on postprandial TAG and inflammatory responses in men with MetS. Men (n = 8, > or = 45 years old) with MetS ingested 2 high-SFA OFTTs (1 g fat per kilogram body weight), with either a 20:1 (low n-3) or 2:1 (high n-3) n-6/n-3 PUFA ratio, and a water control in a randomized crossover design. Blood samples were collected for 8 hours after treatment to measure postprandial TAG, free fatty acids, IL-6, soluble IL-6 receptor, and CRP. Postprandial TAG increased at the same rate after ingestion of the low-n-3 and high-n-3 OFTTs; however, both OFTTs were significantly different from the water control. There were no differences in the rate at which IL-6 concentrations increased after ingestion of either of the OFTTs compared with water. Furthermore, neither time nor treatment affected circulating soluble IL-6 receptor or CRP concentrations. Thus, increasing the n-3 PUFA content of a high-SFA OFTT does not acutely change postprandial TAG or inflammatory responses in men with MetS.
Collapse
Affiliation(s)
- Hilary M F Tulk
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada N1G 2W1
| | | |
Collapse
|
170
|
Comparison of seal oil to tuna oil on plasma lipid levels and blood pressure in hypertriglyceridaemic subjects. Lipids 2009; 44:827-35. [PMID: 19727884 DOI: 10.1007/s11745-009-3333-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2009] [Accepted: 07/20/2009] [Indexed: 01/16/2023]
Abstract
As meat is a rich source of the omega-3 fatty acid docosapentaenoic acid (DPA) and Australians consume six times more meat than fish, investigation of the potential health benefit of DPA is warranted. The aims were to compare the effects of seal oil supplementation with fish oil, on measures of plasma lipids and blood pressure in hypertriglyceridaemic subjects. Forty-eight volunteers were recruited from the Wollongong community and were randomly allocated to one of three groups either receiving 1 g/day of long-chain omega-3 polyunsaturated fatty acids (LC n-3 PUFA) using one of three oils: seal oil capsules (340 mg eicosapentaenoic acid (EPA), 230 mg DPA, 450 mg DHA), fish oil capsules (210 mg EPA, 30 mg DPA, 810 mg DHA) or placebo capsules (containing sunola oil) for 6 weeks. Plasma triglycerides remained unchanged in the placebo group, whilst reductions of 7 and 14% (P < 0.05) were seen in the fish oil and seal oil groups respectively. Systolic blood pressure improved by 8 and 5 mmHg with seal oil and fish oil respectively (P < 0.05). The mean arterial pressure was significantly lower after seal oil supplementation (P < 0.005) compared with the placebo group. These results indicate that seal oil is as effective as fish oil in lowering plasma triglycerides and blood pressure.
Collapse
|
171
|
Trøseid M, Arnesen H, Hjerkinn EM, Seljeflot I. Serum levels of interleukin-18 are reduced by diet and n-3 fatty acid intervention in elderly high-risk men. Metabolism 2009; 58:1543-9. [PMID: 19595382 DOI: 10.1016/j.metabol.2009.04.031] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2008] [Accepted: 04/24/2009] [Indexed: 11/16/2022]
Abstract
Inflammation plays a central role in the development and progression of atherosclerosis, and inflammatory markers have been reported to predict cardiovascular events. Mediterranean-like diet and very long chain omega-3 polyunsaturated fatty acid (n-3 PUFA) supplementation have been reported to reduce the risk of cardiovascular mortality and morbidity, but the mechanisms are not fully clarified. The aims of the present study were to investigate the effect of such interventions on serum levels of inflammatory markers, and potential associations with changes in serum fatty acids and anthropometric measures. This was a randomized 2 x 2 factorial-designed trial comparing the effect of 3 years of dietary counseling, n-3 PUFA supplementation (2.4 g/d), or both on different measures of atherosclerosis in elderly high-risk men (N = 563). Levels of interleukin-18 (IL-18) were decreased by diet (-10.5% vs baseline, P = .012 compared with no diet) and by n-3 PUFA supplementation (-9.9% vs baseline, P = .008 compared with placebo). Other measured inflammatory markers were not affected. Changes in IL-18 were significantly correlated to changes in triglycerides (r = 0.20, P < .001), eicosapentaenoic acid (r = -0.14, P = .030), docosahexaenoic acid (r = -0.14, P = .034), body mass index (r = 0.16, P < .001), and waist circumference (r = 0.12, P = .007). In conclusion, levels of IL-18 were significantly reduced by Mediterranean-like diet and n-3 PUFA supplementation. However, the changes correlated only weakly to changes in triglycerides, serum fatty acids, and anthropometric measures. The cardioprotective effects of both interventions might thus in part be explained by reduced levels of IL-18, but probably beyond changes in serum fatty acids and body composition.
Collapse
Affiliation(s)
- Marius Trøseid
- Center for Clinical Heart Research, Ullevål Department of Cardiology, Oslo University Hospital, University of Oslo, N-0407 Oslo, Norway
| | | | | | | |
Collapse
|
172
|
Siddique RA, Tandon M, Ambwani T, Rai SN, Atreja SK. Nutrigenomics: Nutrient-Gene Interactions. FOOD REVIEWS INTERNATIONAL 2009. [DOI: 10.1080/87559120903155883] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
173
|
Fasting triacylglycerol status, but not polyunsaturated/saturated fatty acid ratio, influences the postprandial response to a series of oral fat tolerance tests. J Nutr Biochem 2009; 20:694-704. [DOI: 10.1016/j.jnutbio.2008.06.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2008] [Revised: 06/10/2008] [Accepted: 06/17/2008] [Indexed: 12/28/2022]
|
174
|
Cheng IS, Liao SF, Liu KL, Liu HY, Wu CL, Huang CY, Mallikarjuna K, Smith RW, Kuo CH. Effect of dietary glycemic index on substrate transporter gene expression in human skeletal muscle after exercise. Eur J Clin Nutr 2009; 63:1404-10. [DOI: 10.1038/ejcn.2009.100] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
175
|
Abstract
PURPOSE OF REVIEW To summarize recent findings relating to the impact of dietary fat composition on whole body lipid metabolism, and common gene variants on the blood lipid response to dietary fat change. RECENT FINDINGS In recent years a more comprehensive understanding of the impact of polyunsaturated fat (PUFA) intake on the regulation of transcription factors involved in lipogenesis and fatty acid and lipoprotein metabolism has emerged. The evidence is suggestive of a greater potency of the long chain n-3 PUFA, eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), and in particular their oxidative products, relative to n-6 PUFA. In the area of nutrigenetics a number of common gene variants have been identified which may be important determinants of the blood lipid response to altered dietary fat composition. However, confirmation of associations in independent cohorts, and an understanding of the size effect of individual or combinations of genotypes, is often lacking. SUMMARY Although in the future, genotyping holds the potential as a public health tool to target and personalize dietary advice, nutrigenetics is a relatively new science, and further research is needed to address the existing inconsistencies and knowledge gaps.
Collapse
Affiliation(s)
- Anne Marie Minihane
- School of Chemistry, Food Biosciences and Pharmacy, University of Reading, Berkshire, UK.
| |
Collapse
|
176
|
Medvedovic M, Gear R, Freudenberg JM, Schneider J, Bornschein R, Yan M, Mistry MJ, Hendrix H, Karyala S, Halbleib D, Heffelfinger S, Clegg DJ, Anderson MW. Influence of fatty acid diets on gene expression in rat mammary epithelial cells. Physiol Genomics 2009; 38:80-8. [PMID: 19351911 PMCID: PMC2696152 DOI: 10.1152/physiolgenomics.00007.2009] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2009] [Accepted: 04/01/2009] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND This study examines the impact of dietary fatty acids on regulation of gene expression in mammary epithelial cells before and during puberty. METHODS Diets primarily consisted of n-9 monounsaturated fatty acids (olive oil), n-6 polyunsaturated fatty acids (safflower), saturated acids (butter), and the reference AIN-93G diet (soy oil). The dietary regimen mimics the repetitive nature of fatty acid exposure in Western diets. Diet-induced changes in gene expression were examined in laser capture microdissected mammary ductal epithelial cells at day of weaning and end of puberty. PCNA immunohistochemistry analysis compared proliferation rates between diets. RESULTS Genes differentially expressed between each test diets and the reference diet were significantly enriched by cell cycle genes. Some of these genes were involved in activation of the cell cycle pathway or the G2/M check point pathway. Although there were some differences in the level of differential expression, all diets showed qualitatively the same pattern of differential expression compared to the reference diet. Cluster analysis identified an expanded set of cell cycle as well as immunity and sterol metabolism related clusters of differentially expressed genes. CONCLUSION Fatty acid-enriched diets significantly upregulated proliferation above normal physiological levels during puberty. Higher cellular proliferation during puberty caused by enriched fatty acid diets poses a potential increase risk of mammary cancer in later life. The human homologs of 27 of 62 cell cycle rat genes are included in a human breast cancer cluster of 45 cell cycle genes, further emphasizing the importance of our findings in the rat model.
Collapse
Affiliation(s)
- M Medvedovic
- Department of Cancer and Cell Biology, University of Cincinnati, Cincinnati, Ohio 45267-0521, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
177
|
de Roos B, Mavrommatis Y, Brouwer IA. Long-chain n-3 polyunsaturated fatty acids: new insights into mechanisms relating to inflammation and coronary heart disease. Br J Pharmacol 2009; 158:413-28. [PMID: 19422375 DOI: 10.1111/j.1476-5381.2009.00189.x] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Evidence from observational studies, prospective cohort studies and randomized clinical intervention studies indicate that moderate doses of long-chain n-3 polyunsaturated fatty acids (LC n-3 PUFA) significantly decrease risk of fatal coronary heart disease (CHD). Higher doses and longer duration of intervention may also protect from non-fatal CHD events. The exact mechanisms through which LC n-3 PUFA has an effect on CHD are not well established but may include a decrease in fasting and postprandial triacylglycerol levels, a decrease in arrhythmias, modulation of platelet aggregation and decreased synthesis of pro-inflammatory agents. The mechanistic relation between LC n-3 PUFA and inflammation has attracted great interest, and in vitro studies have revealed that these fatty acids decrease endothelial activation, affect eicosanoid metabolism (including epoxygenation pathways) and induce inflammatory resolution. However, the effects of LC n-3 PUFA on established biomarkers of inflammation and endothelial activation in vivo are not strong. Consequently we need new and more sensitive and systemic biomarkers to reveal the effects of LC n-3 PUFA on localized inflammatory processes.
Collapse
Affiliation(s)
- Baukje de Roos
- University of Aberdeen, Rowett Institute of Nutrition & Health, Aberdeen, UK.
| | | | | |
Collapse
|
178
|
Pan J, Keffer J, Emami A, Ma X, Lan R, Goldman R, Chung FL. Acrolein-derived DNA adduct formation in human colon cancer cells: its role in apoptosis induction by docosahexaenoic acid. Chem Res Toxicol 2009; 22:798-806. [PMID: 19341237 PMCID: PMC2683896 DOI: 10.1021/tx800355k] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The apoptotic effects of docosahexaenoic acid (DHA) and other omega-3 polyunsaturated fatty acids (PUFAs) have been documented in cell and animal studies. The molecular mechanism by which DHA induces apoptosis is unclear. Although there is no direct evidence, some studies have suggested that DNA damage generated through lipid peroxidation may be involved. Our previous studies showed that DHA, because it has a high degree of unsaturation, can give rise to the acrolein-derived 1,N(2)-propanodeoxyguanosine (Acr-dG) as a major class of DNA adducts via lipid oxidation. As a first step to investigate the possible role of oxidative DNA damage in apoptosis induced by DHA, we examined the relationships between oxidative DNA damage and apoptosis caused by DHA in human colon cancer HT-29 cells. Apoptosis and oxidative DNA damage, including Acr-dG and 8-oxo-deoxyguanosine (8-oxo-dG) formation, in cells treated with DHA and omega-6 PUFAs, including arachidonic acid (AA) and linoleic acid (LA), were measured. DHA induced apoptosis in a dose- and time-dependent manner with a concentration range from 0 to 300 microM as indicated by increased caspase-3 activity and PARP cleavage. In contrast, AA and LA had little or no effect at these concentrations. The Acr-dG levels were increased in HT-29 cells treated with DHA at 240 and 300 microM, and the increases were correlated with the induction of apoptosis at these concentrations, while no significant changes were observed for 8-oxo-dG. Because proteins may compete with DNA to react with acrolein, we then examined the effects of BSA on DHA-induced apoptosis and oxidative DNA damage. The addition of BSA to HT-29 cell culture media significantly decreases Acr-dG levels with a concomitant decrease in the apoptosis induced by DHA. The reduced Acr-dG formation is attributed to the reaction of BSA with acrolein as indicated by increased levels of total protein carbonyls. Similar correlations between Acr-dG formation and apoptosis were observed in HT-29 cells directly incubated with 0-200 microM acrolein. Additionally, DHA treatment increased the level of DNA strand breaks and caused cell cycle arrested at G1 phase. Taken together, these results demonstrate the parallel relationships between Acr-dG level and apoptosis in HT-29 cells, suggesting that the formation of Acr-dG in cellular DNA may contribute to apoptosis induced by DHA.
Collapse
Affiliation(s)
- Jishen Pan
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057
| | - Jessica Keffer
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD
| | - Armaghan Emami
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057
| | - Xiaoyue Ma
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057
| | - Renny Lan
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057
| | - Radoslav Goldman
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057
| | - Fung-Lung Chung
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057
| |
Collapse
|
179
|
Shi Y, Pestka JJ. Mechanisms for suppression of interleukin-6 expression in peritoneal macrophages from docosahexaenoic acid-fed mice. J Nutr Biochem 2009; 20:358-68. [PMID: 18602807 PMCID: PMC2727136 DOI: 10.1016/j.jnutbio.2008.04.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2007] [Revised: 04/02/2008] [Accepted: 04/14/2008] [Indexed: 10/21/2022]
Abstract
Consumption of the trichothecene mycotoxin deoxynivalenol (DON) induces interleukin-6 (IL-6)-dependent IgA nephropathy (IgAN) in mice. This effect can be prevented by feeding long-chain n-3 polyunsaturated fatty acids (PUFAs) found in fish oil. The purpose of this study was to identify the signal transduction pathways by which DON up-regulates IL-6 in the peritoneal macrophage and how consumption of fish oil enriched with the n-3 PUFA docosahexaenoic acid (DHA) suppresses these processes. Incubation with DON induced IL-6 expression in naïve macrophages maximally at 3 h. Knockdown of the transcription factor cAMP response element-binding protein (CREB) or pharmacologic inhibition of the CREB kinases Akt1/2, MSK1 and RSK1 down-regulated this expression. Inhibition of double-stranded RNA-activated protein kinase (PKR) suppressed not only IL-6 expression but also phosphorylation of CREB and its upstream kinases, Akt1, MSK1 and RSK1. Phosphorylations of PKR, CREB kinases and CREB were markedly impaired in peritoneal macrophages isolated from mice that consumed DHA-enriched fish oil for 6 to 8 weeks. DHA's effects were not explainable by increased activity of protein phosphatase 1 and 2A since both were suppressed in mice consuming the DHA diet. Although cells cultured directly with DHA expressed less IL-6 compared to cells cultured with arachidonic acid (AA), neither fatty acid treatment affected DON-induced protein phosphorylation. Furthermore, DHA and AA similarly inhibited cell-free protein kinase activity. These data suggest that DON-induced IL-6 expression is CREB mediated and PKR dependent, and that requisite kinase activities for these pathways were suppressed in macrophages from mice fed DHA for an extended period.
Collapse
Affiliation(s)
- Yuhui Shi
- Dept. of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, USA
- Center for Integrative Toxicology, Michigan State University, East Lansing, MI, USA
| | - James J. Pestka
- Dept. of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, USA
- Center for Integrative Toxicology, Michigan State University, East Lansing, MI, USA
- Dept. of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
180
|
Hontecillas R, O'Shea M, Einerhand A, Diguardo M, Bassaganya-Riera J. Activation of PPAR γ and α by Punicic Acid Ameliorates Glucose Tolerance and Suppresses Obesity-Related Inflammation. J Am Coll Nutr 2009; 28:184-95. [DOI: 10.1080/07315724.2009.10719770] [Citation(s) in RCA: 115] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
181
|
Roumen C, Blaak EE, Corpeleijn E. Lifestyle intervention for prevention of diabetes: determinants of success for future implementation. Nutr Rev 2009; 67:132-46. [DOI: 10.1111/j.1753-4887.2009.00181.x] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
182
|
Corpeleijn E, Saris WHM, Blaak EE. Metabolic flexibility in the development of insulin resistance and type 2 diabetes: effects of lifestyle. Obes Rev 2009; 10:178-93. [PMID: 19207879 DOI: 10.1111/j.1467-789x.2008.00544.x] [Citation(s) in RCA: 164] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Lipotoxicity in skeletal muscle plays a critical role in the aetiology of insulin resistance and type 2 diabetes mellitus by interference of lipid metabolites with insulin signalling and action. The dynamics of lipid oxidation and fine tuning with fatty acid uptake and intramyocellular triacylglycerol turnover may be very important to limit the accumulation of lipid intermediates. The use of metabolic inflexibility, defined as the impaired capacity to increase fat oxidation upon increased fatty acid availability and to switch between fat and glucose as the primary fuel source after a meal, does more justice to the complexity of changes in fuel oxidation during the day. Fatty acid availability, uptake and oxidation all play a role in metabolic flexibility and insulin resistance. During high fatty acid availability, fatty acid transporters may limit cellular and mitochondrial fatty acid uptake and thus limit fat oxidation. After a meal, when the demand for fatty acids as fuel is low, an increased fractional extraction of lipids from plasma may promote intramyocellular lipid accumulation and insulin resistance. Furthermore, defects in fuel switching cluster together with impaired mitochondrial content and/or function. Lifestyle changes in dietary fat intake, physical activity and weight loss may improve metabolic flexibility in skeletal muscle, and thereby contribute to the prevention of type 2 diabetes.
Collapse
Affiliation(s)
- E Corpeleijn
- Department of Human Biology, The Nutrition and Toxicology Research Institute (NUTRIM), Maastricht University, Maastricht, The Netherlands.
| | | | | |
Collapse
|
183
|
Drozdowski LA, Clandinin MT, Thomson ABR. Morphological, kinetic, membrane biochemical and genetic aspects of intestinal enteroplasticity. World J Gastroenterol 2009; 15:774-87. [PMID: 19230039 PMCID: PMC2653378 DOI: 10.3748/wjg.15.774] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The process of intestinal adaptation (“enteroplasticity”) is complex and multifaceted. Although a number of trophic nutrients and non-nutritive factors have been identified in animal studies, successful, reproducible clinical trials in humans are awaited. Understanding mechanisms underlying this adaptive process may direct research toward strategies that maximize intestinal function and impart a true clinical benefit to patients with short bowel syndrome, or to persons in whom nutrient absorption needs to be maximized. In this review, we consider the morphological, kinetic and membrane biochemical aspects of enteroplasticity, focus on the importance of nutritional factors, provide an overview of the many hormones that may alter the adaptive process, and consider some of the possible molecular profiles. While most of the data is derived from rodent studies, wherever possible, the results of human studies of intestinal enteroplasticity are provided.
Collapse
|
184
|
Ballou MA, Gomes RC, Juchem SO, DePeters EJ. Effects of dietary supplemental fish oil during the peripartum period on blood metabolites and hepatic fatty acid compositions and total triacylglycerol concentrations of multiparous Holstein cows. J Dairy Sci 2009; 92:657-69. [PMID: 19164678 DOI: 10.3168/jds.2008-1196] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The objectives were to evaluate the effects of dietary fish oil on plasma metabolite, hepatic fatty acid composition, and total triacylglycerol concentrations. Multiparous Holstein cows (n = 42) were completely randomized to 1 of 3 treatments at 3 wk prepartum. Treatments were no supplemental lipid or supplemental lipid from either Energy Booster (Milk Specialties Co., Dundee, IL) or fish oil. Treatment diets were fed from -21 d relative to expected date of parturition until 10 d postpartum. Treatments were fed as a bolus before the a.m. feeding. The dose of lipid fed during the prepartum period was 250 g, whereas approximately 0.92% of the previous day's dry matter intake was supplemented postpartum. Blood was collected 3 times weekly for determination of plasma metabolites. Liver biopsies were performed at 21 and 10 d before expected date of parturition and 1 and 14 d after parturition to determine fatty acid compositions and total triacylglycerol concentrations. Dry matter intake, milk yield, and loss of body weight or body condition score were not affected by supplementing the diet with lipid or by the source of lipid. Supplemental lipid tended to increase plasma glucose and decrease nonesterified fatty acids during the postpartum period. Furthermore, plasma beta-hydroxybutyrate was reduced during the postpartum period in the lipid-supplemented treatments. However, source of supplemental lipid had no influence on any blood metabolite. Supplemental fish oil altered the fatty acid composition of liver phospholipids and triacylglycerols, decreasing total saturated fatty acids and increasing total n-3 and long-chain polyunsaturated fatty acids (>20 carbon fatty acids). Despite the altered fatty acid composition, hepatic total triacylglycerol concentrations were unaffected by supplemental fish oil. Furthermore, the improved metabolic profile following lipid supplementation did not decrease hepatic total triacylglycerol concentrations.
Collapse
Affiliation(s)
- M A Ballou
- Department of Animal Science and Nutritional Biology Graduate Group, University of California, Davis 95616, USA.
| | | | | | | |
Collapse
|
185
|
Mochizuki K, Suzuki T, Goda T. PPAR alpha and PPAR delta transactivity and p300 binding activity induced by arachidonic acid in colorectal cancer cell line Caco-2. J Nutr Sci Vitaminol (Tokyo) 2009; 54:298-302. [PMID: 18797151 DOI: 10.3177/jnsv.54.298] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
It is reported that arachidonic acid strongly induces the conformational change in vitro and transactivity of PPAR alpha in colorectal cancer cell line Caco-2. In this study, we demonstrated that the induction of conformational change and transactivity of PPAR delta by arachidonic acid, as well as other polyunsaturated fatty acids, was considerably lower than that of PPAR alpha. Mammalian two-hybrid assay showed that arachidonic acid enhanced binding of one of the coactivators, p300, to PPAR alpha but not to PPAR delta. Additionally, arachidonic acid induced in vitro binding of both PPAR alpha-RXR alpha and PPAR delta-RXR alpha heterodimers to several PPREs on CRBPII, L-FABP and ACO genes. Our results suggest that the lower transactivity of PPAR delta for arachidonic acid in Caco-2 cells, compared with PPAR alpha, is associated with the binding activity of p300 to the receptor.
Collapse
Affiliation(s)
- Kazuki Mochizuki
- Department of Nutrition, School and Nutritional Sciences, The University of Shizuoka, 51-1 Yada, Suruga, Shizuoka 422-8526, Japan
| | | | | |
Collapse
|
186
|
Nascimento FAM, Barbosa-da-Silva S, Fernandes-Santos C, Mandarim-de-Lacerda CA, Aguila MB. Adipose tissue, liver and pancreas structural alterations in C57BL/6 mice fed high-fat-high-sucrose diet supplemented with fish oil (n-3 fatty acid rich oil). ACTA ACUST UNITED AC 2009; 62:17-25. [PMID: 19186042 DOI: 10.1016/j.etp.2008.12.008] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2008] [Revised: 11/27/2008] [Accepted: 12/23/2008] [Indexed: 01/09/2023]
Abstract
Fish oil treatment was used in reversing the morphological and metabolic changes of C57BL/6 mice fed high-fat-high-sucrose (HFHS) diet. Two-month-old male C57BL/6 mice were fed HFHS chow or standard chow (SC). At 3 months of age, HFHS mice were separated into an untreated group (HFHS) and a group treated with fish oil (HFHS-Fo, 1.5g/kg/day). At 4 months of age, HFHS fed mice had an increase in body mass (BM) and total body fat, when the animals were sacrificed. Both parameters were lower in HFHS-Fo than in HFHS mice. Plasma glucose and insulin levels were not affected among the groups, but HFHS and HFHS-Fo animals had higher homeostasis model assessment for insulin resistance HOMA-IR ratio. HFHS and HFHS-FO mice had increased plasma total cholesterol and LDL-C, HFHS-Fo increased plasma HDL-C and decreased triglycerides levels. The liver mass (LM) and the adipocytes' size were larger in HFHS mice, while HFHS-Fo mice had a lower LM and smaller adipocytes. The liver steatosis and hepatocyte binucleation were increased in HFHS mice, while HFHS-Fo mice had reduced liver steatosis and hepatocyte binucleation. HFHS-Fo mice had a lower pancreas mass, while HFHS animals had higher islet pancreatic diameter. The SC group showed strong expression for insulin, glucagon and a glucose transporter type 2 GLUT-2 in all pancreatic islets, while in HFHS mice there was less expression for GLUT-2. However, HFHS-Fo mice showed an increase of GLUT-2 expression. In conclusion, dietary fish oil treatment reduces body mass and fat pad adiposity, and also by reducing plasma TG and pancreatic islet hypertrophy in mice fed high-fat-high-sucrose diet. Furthermore, fish oil improves glucagon and GLUT-2 expressions when it is decreased in insulin, but in hepatocyte binucleation and hepatic steatosis where the effect is reduced.
Collapse
Affiliation(s)
- Fernanda A M Nascimento
- Laboratório de Morfometria e Morfologia Cardiovascular, Centro Biomédico, Instituto de Biologia, Universidade do Estado do Rio de Janeiro, Av. 28 de Setembro 87 (fds) 20551-030 Rio de Janeiro, RJ, Brasil
| | | | | | | | | |
Collapse
|
187
|
McCurdy CE, Bishop JM, Williams SM, Grayson BE, Smith MS, Friedman JE, Grove KL. Maternal high-fat diet triggers lipotoxicity in the fetal livers of nonhuman primates. J Clin Invest 2009; 119:323-35. [PMID: 19147984 DOI: 10.1172/jci32661] [Citation(s) in RCA: 321] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2007] [Accepted: 11/26/2008] [Indexed: 12/11/2022] Open
Abstract
Maternal obesity is thought to increase the offspring's risk of juvenile obesity and metabolic diseases; however, the mechanism(s) whereby excess maternal nutrition affects fetal development remain poorly understood. Here, we investigated in nonhuman primates the effect of chronic high-fat diet (HFD) on the development of fetal metabolic systems. We found that fetal offspring from both lean and obese mothers chronically consuming a HFD had a 3-fold increase in liver triglycerides (TGs). In addition, fetal offspring from HFD-fed mothers (O-HFD) showed increased evidence of hepatic oxidative stress early in the third trimester, consistent with the development of nonalcoholic fatty liver disease (NAFLD). O-HFD animals also exhibited elevated hepatic expression of gluconeogenic enzymes and transcription factors. Furthermore, fetal glycerol levels were 2-fold higher in O-HFD animals than in control fetal offspring and correlated with maternal levels. The increased fetal hepatic TG levels persisted at P180, concurrent with a 2-fold increase in percent body fat. Importantly, reversing the maternal HFD to a low-fat diet during a subsequent pregnancy improved fetal hepatic TG levels and partially normalized gluconeogenic enzyme expression, without changing maternal body weight. These results suggest that a developing fetus is highly vulnerable to excess lipids, independent of maternal diabetes and/or obesity, and that exposure to this may increase the risk of pediatric NAFLD.
Collapse
Affiliation(s)
- Carrie E McCurdy
- Department of Pediatrics, University of Colorado Denver, Aurora, Colorado 80045, USA
| | | | | | | | | | | | | |
Collapse
|
188
|
Papadopoulos GA, Erkens T, Maes DGD, Peelman LJ, van Kempen TATG, Buyse J, Janssens GPJ. Peripartal feeding strategy with different n-6:n-3 ratios in sows: effect on gene expression in backfat white adipose tissue postpartum. Br J Nutr 2009; 101:197-205. [PMID: 18498673 DOI: 10.1017/s0007114508994782] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The aim of this study was to describe the effects of two diets differing in n-6:n-3 ratio and prepartal feeding regime on gene expression of PPARgamma1a/1b, PPARgamma1c/1d, PPARgamma2, PPARgamma coactivator 1A (PPARGC1A), GLUT4, TNFalpha, adiponectin, leptin, leptin receptor (LEPR), fatty acid binding protein 4 (FABP4), lipoprotein lipase (LPL) in sows' white adipose tissue on the first day of lactation. The relationship between mRNA expression of these genes and circulating insulin, leptin and thyroid hormones was also considered. Diets contained a low (supplemented with fish oil; f group) or a high (supplemented with sunflower oil; s group) n-6:n-3 ratio and were provided from 8 (f8, s8) or 3d (f3, s3) before parturition (onset day 8 or 3). A low n-6:n-3 ratio reduced the 1d postpartum expression of PPARgamma2 and PPARGC1A but only when applied from 3 d before parturition. Circulating leptin was negatively correlated with mRNA expression of adiponectin, LEPR and LPL, whereas thyroxine was positively correlated with levels of PPARGC1A. In conclusion, the effect of dietary treatments, e.g. altering the n-6:n-3 ratio, around parturition on the expression of crucial genes in nutrient metabolism can be modulated by the duration of application before parturition.
Collapse
Affiliation(s)
- Georgios A Papadopoulos
- Laboratory of Animal Nutrition, Faculty of Veterinary Medicine, Ghent University, Heidestraat 19, B-9820 Merelbeke, Belgium.
| | | | | | | | | | | | | |
Collapse
|
189
|
Vanschoonbeek K, Wouters K, van der Meijden PE, van Gorp PJ, Feijge MA, Herfs M, Schurgers LJ, Hofker MH, de Maat MP, Heemskerk JW. Anticoagulant Effect of Dietary Fish Oil in Hyperlipidemia. Arterioscler Thromb Vasc Biol 2008; 28:2023-9. [DOI: 10.1161/atvbaha.107.156992] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective—
In hyperlipidemia, dietary fish oil containing n-3 polyunsaturated fatty acids (PUFA) provokes plasma triacylglycerol lowering and hypocoagulant activity. Using APOE2 knock-in mice, the relation of these fish-oil effects with altered gene expression was investigated.
Methods and Results—
Male APOE2 knock-in mice, fed regular low-fat diet, had elevated plasma levels of triacylglycerol and coagulation factors. Plasma lipids and (anti)coagulant factors reduced on feeding the mice with fish oil (n-3 PUFA) or, to a lesser degree, with sunflowerseed oil (n-6 PUFA). The fish-oil diet provoked a 40% reduction in thrombin generation. Microarray (Affymetrix) and single-gene expression analysis of mouse livers showed that fish oil induced: (1) upregulation of genes contributing to lipid degradation and oxidation; (2) downregulation of genes of γ-glutamyl carboxylase and of transcription factors implicated in lipid synthesis; (3) unchanged expression of coagulation factor genes. After fish-oil diet, vitamin K–dependent coagulation factors accumulated in periportal areas of the liver; prothrombin was partly retained in uncarboxylated form. Only part of the changes in gene expression were different from the effects of sunflowerseed oil diet.
Conclusions—
The hypocoagulant effect of n-3 PUFA is not caused by reduced hepatic synthesis of coagulation factors, but rather results from retention of uncarboxylated coagulation factors. In contrast, the lipid-lowering effect of n-3 PUFA links to altered expression of genes that regulate transcription and fatty acid metabolism.
Collapse
Affiliation(s)
- Kristof Vanschoonbeek
- From the Departments of Biochemistry (K.V., P.E.J.v.M., M.A.H.F., L.J.S., J.W.M.H.), Human Biology (K.V.), and Molecular Genetics (K.W., P.J.v.G., M.H.H.), CARIM and NUTRIM, and VitaK (M.H., L.J.S.), Maastricht University, The Netherlands; the Department of Pathology and Laboratory Medicine (M.H.H.), University Medical Center Groningen, The Netherlands; and the Department of Hematology (M.d.M.), Erasmus Medical Centre Rotterdam, The Netherlands
| | - Kristiaan Wouters
- From the Departments of Biochemistry (K.V., P.E.J.v.M., M.A.H.F., L.J.S., J.W.M.H.), Human Biology (K.V.), and Molecular Genetics (K.W., P.J.v.G., M.H.H.), CARIM and NUTRIM, and VitaK (M.H., L.J.S.), Maastricht University, The Netherlands; the Department of Pathology and Laboratory Medicine (M.H.H.), University Medical Center Groningen, The Netherlands; and the Department of Hematology (M.d.M.), Erasmus Medical Centre Rotterdam, The Netherlands
| | - Paola E.J. van der Meijden
- From the Departments of Biochemistry (K.V., P.E.J.v.M., M.A.H.F., L.J.S., J.W.M.H.), Human Biology (K.V.), and Molecular Genetics (K.W., P.J.v.G., M.H.H.), CARIM and NUTRIM, and VitaK (M.H., L.J.S.), Maastricht University, The Netherlands; the Department of Pathology and Laboratory Medicine (M.H.H.), University Medical Center Groningen, The Netherlands; and the Department of Hematology (M.d.M.), Erasmus Medical Centre Rotterdam, The Netherlands
| | - Patrick J. van Gorp
- From the Departments of Biochemistry (K.V., P.E.J.v.M., M.A.H.F., L.J.S., J.W.M.H.), Human Biology (K.V.), and Molecular Genetics (K.W., P.J.v.G., M.H.H.), CARIM and NUTRIM, and VitaK (M.H., L.J.S.), Maastricht University, The Netherlands; the Department of Pathology and Laboratory Medicine (M.H.H.), University Medical Center Groningen, The Netherlands; and the Department of Hematology (M.d.M.), Erasmus Medical Centre Rotterdam, The Netherlands
| | - Marion A.H. Feijge
- From the Departments of Biochemistry (K.V., P.E.J.v.M., M.A.H.F., L.J.S., J.W.M.H.), Human Biology (K.V.), and Molecular Genetics (K.W., P.J.v.G., M.H.H.), CARIM and NUTRIM, and VitaK (M.H., L.J.S.), Maastricht University, The Netherlands; the Department of Pathology and Laboratory Medicine (M.H.H.), University Medical Center Groningen, The Netherlands; and the Department of Hematology (M.d.M.), Erasmus Medical Centre Rotterdam, The Netherlands
| | - Marjolein Herfs
- From the Departments of Biochemistry (K.V., P.E.J.v.M., M.A.H.F., L.J.S., J.W.M.H.), Human Biology (K.V.), and Molecular Genetics (K.W., P.J.v.G., M.H.H.), CARIM and NUTRIM, and VitaK (M.H., L.J.S.), Maastricht University, The Netherlands; the Department of Pathology and Laboratory Medicine (M.H.H.), University Medical Center Groningen, The Netherlands; and the Department of Hematology (M.d.M.), Erasmus Medical Centre Rotterdam, The Netherlands
| | - Leon J. Schurgers
- From the Departments of Biochemistry (K.V., P.E.J.v.M., M.A.H.F., L.J.S., J.W.M.H.), Human Biology (K.V.), and Molecular Genetics (K.W., P.J.v.G., M.H.H.), CARIM and NUTRIM, and VitaK (M.H., L.J.S.), Maastricht University, The Netherlands; the Department of Pathology and Laboratory Medicine (M.H.H.), University Medical Center Groningen, The Netherlands; and the Department of Hematology (M.d.M.), Erasmus Medical Centre Rotterdam, The Netherlands
| | - Marten H. Hofker
- From the Departments of Biochemistry (K.V., P.E.J.v.M., M.A.H.F., L.J.S., J.W.M.H.), Human Biology (K.V.), and Molecular Genetics (K.W., P.J.v.G., M.H.H.), CARIM and NUTRIM, and VitaK (M.H., L.J.S.), Maastricht University, The Netherlands; the Department of Pathology and Laboratory Medicine (M.H.H.), University Medical Center Groningen, The Netherlands; and the Department of Hematology (M.d.M.), Erasmus Medical Centre Rotterdam, The Netherlands
| | - Moniek P.M. de Maat
- From the Departments of Biochemistry (K.V., P.E.J.v.M., M.A.H.F., L.J.S., J.W.M.H.), Human Biology (K.V.), and Molecular Genetics (K.W., P.J.v.G., M.H.H.), CARIM and NUTRIM, and VitaK (M.H., L.J.S.), Maastricht University, The Netherlands; the Department of Pathology and Laboratory Medicine (M.H.H.), University Medical Center Groningen, The Netherlands; and the Department of Hematology (M.d.M.), Erasmus Medical Centre Rotterdam, The Netherlands
| | - Johan W.M. Heemskerk
- From the Departments of Biochemistry (K.V., P.E.J.v.M., M.A.H.F., L.J.S., J.W.M.H.), Human Biology (K.V.), and Molecular Genetics (K.W., P.J.v.G., M.H.H.), CARIM and NUTRIM, and VitaK (M.H., L.J.S.), Maastricht University, The Netherlands; the Department of Pathology and Laboratory Medicine (M.H.H.), University Medical Center Groningen, The Netherlands; and the Department of Hematology (M.d.M.), Erasmus Medical Centre Rotterdam, The Netherlands
| |
Collapse
|
190
|
Dietary n-6 and n-3 polyunsaturated fatty acids: from biochemistry to clinical implications in cardiovascular prevention. Biochem Pharmacol 2008; 77:937-46. [PMID: 19022225 DOI: 10.1016/j.bcp.2008.10.020] [Citation(s) in RCA: 514] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2008] [Revised: 10/20/2008] [Accepted: 10/21/2008] [Indexed: 11/20/2022]
Abstract
Linoleic acid (LA) and alpha linolenic acid (ALA) belong to the n-6 (omega-6) and n-3 (omega-3) series of polyunsaturated fatty acids (PUFA), respectively. They are defined "essential" fatty acids since they are not synthesized in the human body and are mostly obtained from the diet. Food sources of ALA and LA are most vegetable oils, cereals and walnuts. This review critically revises the most significant epidemiological and interventional studies on the cardioprotective activity of PUFAs, linking their biological functions to biochemistry and metabolism. In fact, a complex series of desaturation and elongation reactions acting in concert transform LA and ALA to their higher unsaturated derivatives: arachidonic acid (AA) from LA, eicosapentaenoic (EPA) and docosahexaenoic acids (DHA) from ALA. EPA and DHA are abundantly present in fish and fish oil. AA and EPA are precursors of different classes of pro-inflammatory or anti-inflammatory eicosanoids, respectively, whose biological activities have been evoked to justify risks and benefits of PUFA consumption. The controversial origin and clinical role of the n-6/n-3 ratio as a potential risk factor in cardiovascular diseases is also examined. This review highlights the important cardioprotective effect of n-3 in the secondary prevention of sudden cardiac death due to arrhythmias, but suggests caution to recommend dietary supplementation of PUFAs to the general population, without considering, at the individual level, the intake of total energy and fats.
Collapse
|
191
|
Kim HJ, Lee KT, Park YB, Jeon SM, Choi MS. Dietary docosahexaenoic acid-rich diacylglycerols ameliorate hepatic steatosis and alter hepatic gene expressions in C57BL/6J-Lep(ob/ob) mice. Mol Nutr Food Res 2008; 52:965-73. [PMID: 18481331 DOI: 10.1002/mnfr.200700315] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
We examined the effect of docosahexaenoic acid (DHA)-enriched structured lipids-diacylglycerol (SL-DG), which were synthesized using soybean oil (SO) and algae oil (AO), on hepatic lipid metabolism and the mRNA expression of genes involved in hepatic steatosis of C57BL/6J-Lep(ob/ob) compared to the SL-triacylglycerol (TG). The animals were fed a high-fat (10% lard and 10% test oils) and high-cholesterol (0.2% cholesterol) diet for 12 weeks. Mice fed SL-DG showed a lower total white adipose tissue weight and plasma triglyceride concentration than the SO group. Reduction of hepatic triglyceride content in the SL-DG group was related with the suppression of hepatic enzyme activities for fatty acid and triglyceride synthesis along with fecal triglyceride excretion compared to the SL-TG. SL-DG also lowered hepatic cholesterol levels by suppressing cholesterol regulating enzyme activity compared to the SO group. Moreover, SL-DG lowered the mRNA expressions of sterol regulatory element binding protein-1 and its target genes than TG-form oils (SO, AO and SL-TG) in the liver. Thus, the current results suggest that DHA-enriched SL-DG oil used in this study is beneficial for ameliorating hepatic steatosis in obese animal model by improving hepatic fatty acid and cholesterol metabolic enzyme activity and their gene expression.
Collapse
Affiliation(s)
- Hye-Jin Kim
- Department of Food Science and Nutrition, Kyungpook National University, Daegu, Korea
| | | | | | | | | |
Collapse
|
192
|
Moon YA, Hammer RE, Horton JD. Deletion of ELOVL5 leads to fatty liver through activation of SREBP-1c in mice. J Lipid Res 2008; 50:412-423. [PMID: 18838740 DOI: 10.1194/jlr.m800383-jlr200] [Citation(s) in RCA: 175] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Elongation of very long chain fatty acids (ELOVL)5 is one of seven mammalian fatty acid condensing enzymes involved in microsomal fatty acid elongation. To determine the in vivo substrates and function of ELOVL5, we generated Elovl5(-/-) mice. Studies using liver microsomal protein from wild-type and knockout mice demonstrated that the elongation of gamma-linolenic (C18:3, n-6) to dihomo-gamma-linolenic (C20:3, n-6) and stearidonic (C18:4, n-3) to omega3-arachidonic acid (C20:4, n-3) required ELOVL5 activity. Tissues of Elovl5(-/-) mice accumulated the C18 substrates of ELOVL5 and the levels of the downstream products, arachidonic acid (C20:4, n-6) and docosahexaenoic acid (DHA, C22:6, n-3), were decreased. A consequence of decreased cellular arachidonic acid and DHA concentrations was the activation of sterol regulatory element-binding protein (SREBP)-1c and its target genes involved in fatty acid and triglyceride synthesis, which culminated in the development of hepatic steatosis in Elovl5(-/-) mice. The molecular and metabolic changes in fatty acid metabolism in Elovl5(-/-) mice were reversed by dietary supplementation with arachidonic acid and DHA. These studies demonstrate that reduced ELOVL5 activity leads to hepatic steatosis, and endogenously synthesized PUFAs are key regulators of SREBP-1c activation and fatty acid synthesis in livers of mice.
Collapse
Affiliation(s)
- Young-Ah Moon
- From the Department of Molecular Genetics, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390-9046
| | - Robert E Hammer
- Department of Biochemistry, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390-9046
| | - Jay D Horton
- From the Department of Molecular Genetics, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390-9046; Department of Internal Medicine, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390-9046.
| |
Collapse
|
193
|
Vardar-Sengul S, Buduneli E, Turkoglu O, Buduneli N, Atilla G, Wahlgren J, Sorsa T, Baylas H. The Effects of Selective COX-2 Inhibitor/Celecoxib and Omega-3 Fatty Acid on Matrix Metalloproteinases, TIMP-1, and Laminin-5γ2-Chain Immunolocalization in Experimental Periodontitis. J Periodontol 2008; 79:1934-41. [DOI: 10.1902/jop.2008.080001] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
194
|
Yeo WK, Lessard SJ, Chen ZP, Garnham AP, Burke LM, Rivas DA, Kemp BE, Hawley JA. Fat adaptation followed by carbohydrate restoration increases AMPK activity in skeletal muscle from trained humans. J Appl Physiol (1985) 2008; 105:1519-26. [PMID: 18801964 DOI: 10.1152/japplphysiol.90540.2008] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We have previously reported that 5 days of a high-fat diet followed by 1 day of high-carbohydrate intake (Fat-adapt) increased rates of fat oxidation and decreased rates of muscle glycogenolysis during submaximal cycling compared with consumption of an isoenergetic high-carbohydrate diet (HCHO) for 6 days (Burke et al. J Appl Physiol 89: 2413-2421, 2000; Stellingwerff et al. Am J Physiol Endocrinol Metab 290: E380-E388, 2006). To determine potential mechanisms underlying shifts in substrate selection, eight trained subjects performed Fat-adapt and HCHO. On day 7, subjects performed 1-h cycling at 70% peak O2 uptake. Muscle biopsies were taken immediately before and after exercise. Resting muscle glycogen content was similar between treatments, but muscle triglyceride levels were higher after Fat-adapt (P < 0.05). Resting AMPK-alpha1 and -alpha2 activity was higher after Fat-adapt (P = 0.02 and P = 0.05, respectively), while the phosphorylation of AMPK's downstream target, acetyl-CoA carboxylase (pACC at Ser221), tended to be elevated after Fat-adapt (P = 0.09). Both the respiratory exchange ratio (P < 0.01) and muscle glycogen utilization (P < 0.05) were lower during exercise after Fat-adapt. Exercise increased AMPK-alpha1 activity after HCHO (P = 0.03) but not Fat-adapt. Exercise was associated with an increase in pACC at Ser221 for both dietary treatments (P < 0.05), with postexercise pACC Ser221 higher after Fat-adapt (P = 0.02). In conclusion, compared with HCHO, Fat-adapt increased resting muscle triglyceride stores and resting AMPK-alpha1 and -alpha2 activity. Fat-adapt also resulted in higher rates of whole body fat oxidation, reduced muscle glycogenolysis, and attenuated the exercise-induced rise in AMPK-alpha1 and AMPK-alpha2 activity compared with HCHO. Our results demonstrate that AMPK-alpha1 and AMPK-alpha2 activity and fuel selection in skeletal muscle in response to exercise can be manipulated by diet and/or the interactive effects of diet and exercise training.
Collapse
Affiliation(s)
- Wee Kian Yeo
- Exercise Metabolism Group, School of Medical Sciences, RMIT University, Victoria, Australia
| | | | | | | | | | | | | | | |
Collapse
|
195
|
Massaro M, Scoditti E, Carluccio MA, De Caterina R. Basic mechanisms behind the effects of n-3 fatty acids on cardiovascular disease. Prostaglandins Leukot Essent Fatty Acids 2008; 79:109-15. [PMID: 18951002 DOI: 10.1016/j.plefa.2008.09.009] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The epidemiological association between high intakes of n-3 fatty acids (FA) and decreased morbidity and mortality from cardiovascular disease (CVD) can be explained by two main basic mechanisms: (a) an effect on atherothrombosis, and (b) an effect on cardiac arrhythmias. These mechanisms probably reflect different beneficial influences of n-3 FA on cardiovascular biology. Effects on atherothrombosis include the modulation of the expression of pro-atherogenic genes (e.g., endothelial leukocyte adhesion molecules, inflammatory cytokines and cyclooxygenase (COX)-2) and the hepatic synthesis of very low density lipoproteins (VLDL), and are slow in onset, requiring incorporation into cell membrane phospholipids, and usually doses in humans in the order of 3g/day or higher. Effects on cardiac arrhythmias include complex interactions with ion channels (sodium, potassium and calcium channels), typically requiring the presence of free FA in extracellular fluids and usually occurring with lower doses (around 1g/day) of nutritional or pharmacological intake. We have focused most of our research effort in unraveling the pathophysiological background of protection by n-3 FA from atherothrombosis. As the result of incorporation of n-3 FA in the sn-2 position predominantly of the phosphatidyl ethanolamine pool in the inner leaflet of the plasma membrane, n-3 FA appear on the one hand to increase the production of bioactive lipid mediators (protectins and resolvins) affecting cytokine-induced signal transduction; and on the other hand to directly interfere with the generation of reactive oxygen species (mostly hydrogen peroxide), directly responsible for the activation of the transcription factor nuclear factor (NF)-kappaB, which controls the expression of a variety of pro-inflammatory and pro-atherogenic genes, including those encoding for interleukin (IL)-1, IL-6, IL-8, tumor necrosis factor (TNF)alpha, vascular cell adhesion molecule-1 (VCAM-1), E-selectin, and COX-2. The upstream-direct or indirect-inhibition of cytokine- and other atherogenic trigger-induced signaling pathway may involve interference with the activation of protein kinase (PK) C isoforms and NADP(H) oxidase. Such interference may also explain the blunt anti-inflammatory effect of n-3 FA in many experimental models and clinical conditions of inflammation. All together, these mechanisms may provide an integrated view of how n-3 FA may affect CVD.
Collapse
Affiliation(s)
- Marika Massaro
- C.N.R. Institute of Clinical Physiology, Pisa and Lecce, Italy; University of Lecce, Ecotekne, Lecce, Italy
| | | | | | | |
Collapse
|
196
|
Yu YH, Lin EC, Wu SC, Cheng WTK, Mersmann HJ, Wang PH, Ding ST. Docosahexaenoic acid regulates adipogenic genes in myoblasts via porcine peroxisome proliferator-activated receptor gamma. J Anim Sci 2008; 86:3385-92. [PMID: 18676720 DOI: 10.2527/jas.2008-1051] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
The nuclear transcription factor peroxisome proliferator-activated receptor gamma (PPARgamma) triggers adipocyte differentiation by regulating lipogenic genes. A ligand for PPARgamma is necessary to activate PPARgamma function. Fatty acids are potential ligands for PPARgamma activation. The current experiment was designed to determine the potential for individual fatty acids to activate porcine PPARgamma ectopically expressed in myoblasts. The expression of adipocyte fatty acid binding protein (aP2) and adiponectin in myoblasts stably expressing porcine PPARgamma was increased when docosahexaenoic acid (DHA) was added to the adipogenic medium. The response was positively related to DHA concentration and suggests that DHA may bind to and activate porcine PPARgamma, leading to increased expression of aP2 and adiponectin. The conditioned media collected from myoblasts expressing PPARgamma between d 3 and 6 or between d 6 and 9, but not DHA itself, activated the aP2 gene promoter-driven luciferase activity. These results suggest that a metabolite of DHA is the ligand binding to and activating porcine PPARgamma. The metabolite and pathway for its production are currently unknown.
Collapse
Affiliation(s)
- Y H Yu
- Department of Animal Science and Technology, Institute of Biotechnology, National Taiwan University, Taipei 106, Taiwan
| | | | | | | | | | | | | |
Collapse
|
197
|
Mochizuki K, Mochizuki H, Kawai H, Ogura Y, Shimada M, Takase S, Goda T. Possible role of fatty acids in milk as the regulator of the expression of cytosolic binding proteins for fatty acids and vitamin A through PPARalpha in developing rats. J Nutr Sci Vitaminol (Tokyo) 2008; 53:515-21. [PMID: 18202540 DOI: 10.3177/jnsv.53.515] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Fatty acids in milk are thought to play an important role in intestinal maturation and gene expression in the postnatal small intestine. In this study, we determined the jejunal mRNA levels, in rats, of peroxisome proliferator-activated receptor alpha (PPARalpha) and PPARdelta which are nuclear receptors for fatty acids. We also measured expression of their target genes during the postnatal period, namely liver type fatty acid-binding protein (L-FABP) and cellular retinol-binding protein, type II (CRBPII). The mRNA levels of PPARalpha, L-FABP and CRBPII, but not PPARdelta, gradually increased during the suckling period and then sharply declined to a low level at the end of the weaning period. Rat pups at 17 d of age, weaned to a high-fat diet, showed significantly greater mRNA levels of PPARalpha, L-FABP and CRBPII than those weaned to a low-fat diet. Oral administration of PPARalpha ligand, WY14,643 during four consecutive days of the weanling period caused a parallel increase in the mRNA levels of PPARalpha, L-FABP and CRBPII genes. Furthermore, caprylic acid and oleic acid, which are major components of fatty acids in milk, induced jejunal PPARalpha, L-FABP and CRBPII gene expression. Our results suggest that fatty acids in milk may play a pivotal role in maintaining an enhanced level of expression of L-FABP and CRBPII genes in the small intestine, presumably by acting as inducers of PPARalpha gene expression.
Collapse
Affiliation(s)
- Kazuki Mochizuki
- School of Food and Nutritional Sciences, The University of Shizuoka, Shizuoka, Japan
| | | | | | | | | | | | | |
Collapse
|
198
|
Gawecka A, Michalkiewicz J, Kornacka MK, Luckiewicz B, Kubiszewska I. Immunologic Properties Differ in Preterm Infants Fed Olive Oil vs Soy-Based Lipid Emulsions During Parenteral Nutrition. JPEN J Parenter Enteral Nutr 2008; 32:448-53. [DOI: 10.1177/0148607108319802] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Agnieszka Gawecka
- From the Department of Neonatology, Medical Academy, Warsaw, Poland; the Pediatric Intensive Care Unit, Medical Academy Children's Hospital, Warsaw, Poland; the Immunology and Microbiology Department, The Children's Memorial Health Institute, Warsaw, Poland; the Hospital Pharmacy of Princess Anna Mazowiecka Hospital, Warsaw, Poland; and the Immunology Department, Rydygiers's Medical Academy, Bydgoszcz, Poland
| | - Jacek Michalkiewicz
- From the Department of Neonatology, Medical Academy, Warsaw, Poland; the Pediatric Intensive Care Unit, Medical Academy Children's Hospital, Warsaw, Poland; the Immunology and Microbiology Department, The Children's Memorial Health Institute, Warsaw, Poland; the Hospital Pharmacy of Princess Anna Mazowiecka Hospital, Warsaw, Poland; and the Immunology Department, Rydygiers's Medical Academy, Bydgoszcz, Poland
| | - Maria Katarzyna Kornacka
- From the Department of Neonatology, Medical Academy, Warsaw, Poland; the Pediatric Intensive Care Unit, Medical Academy Children's Hospital, Warsaw, Poland; the Immunology and Microbiology Department, The Children's Memorial Health Institute, Warsaw, Poland; the Hospital Pharmacy of Princess Anna Mazowiecka Hospital, Warsaw, Poland; and the Immunology Department, Rydygiers's Medical Academy, Bydgoszcz, Poland
| | - Barbara Luckiewicz
- From the Department of Neonatology, Medical Academy, Warsaw, Poland; the Pediatric Intensive Care Unit, Medical Academy Children's Hospital, Warsaw, Poland; the Immunology and Microbiology Department, The Children's Memorial Health Institute, Warsaw, Poland; the Hospital Pharmacy of Princess Anna Mazowiecka Hospital, Warsaw, Poland; and the Immunology Department, Rydygiers's Medical Academy, Bydgoszcz, Poland
| | - Iza Kubiszewska
- From the Department of Neonatology, Medical Academy, Warsaw, Poland; the Pediatric Intensive Care Unit, Medical Academy Children's Hospital, Warsaw, Poland; the Immunology and Microbiology Department, The Children's Memorial Health Institute, Warsaw, Poland; the Hospital Pharmacy of Princess Anna Mazowiecka Hospital, Warsaw, Poland; and the Immunology Department, Rydygiers's Medical Academy, Bydgoszcz, Poland
| |
Collapse
|
199
|
Abstract
The type and quantity of dietary fat ingested contributes to the onset and progression of chronic diseases, like diabetes and atherosclerosis. The liver plays a central role in whole body lipid metabolism and responds rapidly to changes in dietary fat composition. Polyunsaturated fatty acids (PUFA) play a key role in membrane composition and function, metabolism and the control of gene expression. Certain PUFA, like the n-3 PUFA, enhance hepatic fatty acid oxidation and inhibit fatty acid synthesis and VLDL secretion, in part, by regulating gene expression. Our studies have established that key transcription factors, like PPARalpha, SREBP-1, ChREBP and MLX, are regulated by n-3 PUFA, which in turn control levels of proteins involved in lipid and carbohydrate metabolism. Of the n-3 PUFA, 22:6,n-3 has recently been established as a key controller of hepatic lipid synthesis. 22:6,n-3 controls the 26S proteasomal degradation of the nuclear form of SREBP-1. SREBP-1 is a major transcription factor that controls the expression of multiple genes involved fatty acid synthesis and desaturation. 22:6,n-3 suppresses nuclear SREBP-1, which in turn suppresses lipogenesis. This mechanism is achieved, in part, through control of the phosphorylation status of protein kinases. This review will examine both the general features of PUFA-regulated hepatic gene transcription and highlight the unique mechanisms by which 22:6,n-3 impacts gene expression. The outcome of this analysis will reveal that changes in hepatic 22:6,n-3 content has a major impact on hepatic lipid and carbohydrate metabolism. Moreover, the mechanisms involve 22:6,n-3 control of several well-known signaling pathways, such as Akt, Erk1/2, Gsk3beta and PKC (novel or atypical). 22:6,n-3 control of these same signaling pathways in non-hepatic tissues may help to explain the diverse actions of n-3 PUFA on such complex physiological processes as visual acuity and learning.
Collapse
Affiliation(s)
- Donald B Jump
- Department of Physiology, Michigan State University, East Lansing, MI 48824, United States.
| | | | | | | | | | | |
Collapse
|
200
|
Sasaki N, Egashira Y, Sanada H. Down-regulation of alpha-amino-beta-carboxymuconate-epsilon-semialdehyde decarboxylase by polyunsaturated fatty acids in hepatocytes is not mediated by PPARalpha. Eur J Nutr 2008; 47:80-6. [PMID: 18320257 DOI: 10.1007/s00394-008-0699-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2007] [Accepted: 02/13/2008] [Indexed: 01/11/2023]
Abstract
BACKGROUND alpha-Amino-beta-carboxymuconate-epsilon-semialdehyde decarboxylase (ACMSD) is a key enzyme in NAD biosynthesis from tryptophan. Dietary polyunsaturated fatty acids (PUFA) have been shown to suppress hepatic ACMSD activity and its mRNA level in rat. However the mechanism of the suppressive action has not been clarified yet. Although the phenomena that fatty acids suppress the expression of ACMSD in rat liver have been established in vivo experiment, it is still obscure whether the effect of fatty acids on the expression of the enzyme is caused by its direct or indirect action, because there have been very few investigations performed in vitro. AIM OF THE STUDY In this study, to examine whether down-regulation of ACMSD mRNA by PUFA involves peroxisome proliferator-activated receptor (PPAR) alpha mediated mechanism or not, we investigated the effect of PUFA on the ACMSD expression by using primary cultured rat hepatocytes. METHODS For this purpose we investigated the effect of PUFA (linoleic acid and eicosapentanoic acid) on the ACMSD mRNA level in primary-cultured rat hepatocytes and compared its effect with that of WY-14,643 (a PPARalpha agonist). After the incubation of hepatocytes with fatty acids, WY-14,643 and/or MK886 (a PPARalpha antagonist), mRNA levels of ACMSD and a peroxisome marker enzyme acyl-CoA oxidase (ACO) were determined by competitive reverse transcription-polymerase chain reaction (RT-PCR) method. RESULTS ACMSD mRNA level in primary hepatocytes were decreased by the incubation with high concentrations of linoleic acid, eicosapentaenoic acid (EPA) and WY-14,643. The appearance of ACO mRNA by WY-14,643 was remarkably increased, and those by linoleic acid and EPA were increased less than that by WY-14,643. Moreover, the suppression of ACMSD mRNA and the augmentation of ACO mRNA by WY-14,643 were inhibited by MK886, but the suppression by PUFA was not substantially affected by MK886. CONCLUSIONS The present study suggesting that the mechanism of decrease in ACMSD mRNA level by PUFA was different from that by WY-14,643, and that there would be any pathway other than PPARalpha mediated one for PUFA to regulate ACMSD expression.
Collapse
Affiliation(s)
- Naho Sasaki
- Graduate School of Science and Technology, Chiba University, 648 Mastudo, Mastudo-shi, Chiba 271 8510, Japan.
| | | | | |
Collapse
|