1
|
Collins TJC, Morgan PK, Man K, Lancaster GI, Murphy AJ. The influence of metabolic disorders on adaptive immunity. Cell Mol Immunol 2024; 21:1109-1119. [PMID: 39134802 PMCID: PMC11442657 DOI: 10.1038/s41423-024-01206-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 07/19/2024] [Indexed: 10/02/2024] Open
Abstract
The immune system plays a crucial role in protecting the body from invading pathogens and maintaining tissue homoeostasis. Maintaining homoeostatic lipid metabolism is an important aspect of efficient immune cell function and when disrupted immune cell function is impaired. There are numerous metabolic diseases whereby systemic lipid metabolism and cellular function is impaired. In the context of metabolic disorders, chronic inflammation is suggested to be a major contributor to disease progression. A major contributor to tissue dysfunction in metabolic disease is ectopic lipid deposition, which is generally caused by diet and genetic factors. Thus, we propose the idea, that similar to tissue and organ damage in metabolic disorders, excessive accumulation of lipid in immune cells promotes a dysfunctional immune system (beyond the classical foam cell) and contributes to disease pathology. Herein, we review the evidence that lipid accumulation through diet can modulate the production and function of immune cells by altering cellular lipid content. This can impact immune cell signalling, activation, migration, and death, ultimately affecting key aspects of the immune system such as neutralising pathogens, antigen presentation, effector cell activation and resolving inflammation.
Collapse
Affiliation(s)
- Thomas J C Collins
- Division of Immunometabolism, Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia
- Department of Immunology, Monash University, Melbourne, VIC, 3004, Australia
| | - Pooranee K Morgan
- Division of Immunometabolism, Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia
- Department of Immunology, Monash University, Melbourne, VIC, 3004, Australia
| | - Kevin Man
- Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, 3000, Australia
- Department of Microbiology and Immunology, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Graeme I Lancaster
- Division of Immunometabolism, Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia
- Department of Immunology, Monash University, Melbourne, VIC, 3004, Australia
| | - Andrew J Murphy
- Division of Immunometabolism, Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia.
- Department of Immunology, Monash University, Melbourne, VIC, 3004, Australia.
| |
Collapse
|
2
|
Gjøen T, Ruyter B, Østbye TK. Effects of eicosapentaneoic acid on innate immune responses in an Atlantic salmon kidney cell line in vitro. PLoS One 2024; 19:e0302286. [PMID: 38805503 PMCID: PMC11132502 DOI: 10.1371/journal.pone.0302286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 04/02/2024] [Indexed: 05/30/2024] Open
Abstract
Studies of the interplay between metabolism and immunity, known as immunometabolism, is steadily transforming immunological research into new understandings of how environmental cues like diet are affecting innate and adaptive immune responses. The aim of this study was to explore antiviral transcriptomic responses under various levels of polyunsaturated fatty acid. Atlantic salmon kidney cells (ASK cell line) were incubated for one week in different levels of the unsaturated n-3 eicosapentaneoic acid (EPA) resulting in cellular levels ranging from 2-20% of total fatty acid. These cells were then stimulated with the viral mimic and interferon inducer poly I:C (30 ug/ml) for 24 hours before total RNA was isolated and sequenced for transcriptomic analyses. Up to 200 uM EPA had no detrimental effects on cell viability and induced very few transcriptional changes in these cells. However, in combination with poly I:C, our results shows that the level of EPA in the cellular membranes exert profound dose dependent effects of the transcriptional profiles induced by this treatment. Metabolic pathways like autophagy, apelin and VEGF signaling were attenuated by EPA whereas transcripts related to fatty acid metabolism, ferroptosis and the PPAR signaling pathways were upregulated. These results suggests that innate antiviral responses are heavily influenced by the fatty acid profile of salmonid cells and constitute another example of the strong linkage between general metabolic pathways and inflammatory responses.
Collapse
Affiliation(s)
- Tor Gjøen
- Department of Pharmacy, Section for Pharmacology and Pharmaceutical Biosciences, University of Oslo, Oslo, Norway
| | - Bente Ruyter
- Nofima (Norwegian Institute of Food, Fisheries and Aquaculture Research), Ås, Norway
| | - Tone Kari Østbye
- Nofima (Norwegian Institute of Food, Fisheries and Aquaculture Research), Ås, Norway
| |
Collapse
|
3
|
Amr M, Farid A. Impact of cow, buffalo, goat or camel milk consumption on oxidative stress, inflammation and immune response post weaning time. Sci Rep 2024; 14:9967. [PMID: 38693190 PMCID: PMC11063178 DOI: 10.1038/s41598-024-59959-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 04/17/2024] [Indexed: 05/03/2024] Open
Abstract
Milk is a whitish liquid that is secreted from mammary glands; and considered as the primary source of nutrition for newborns since they are not able to digest solid food. However, it contains primary nutrients, as well as growth and immune factors. Early weaning is a critical issue that face women and their babies in developing countries. To avoid infant malnutrition, they tend to use other milk types instead of baby formula. Therefore, the present study aimed to evaluate the impact of cow, buffalo, goat or camel milk consumption on oxidative stress, inflammation and immune response in male and female Sprague Dawley rats post weaning time. The amino acids, fatty acids, minerals and vitamins in the tested milk types were evaluated. Animals were divided into 5 groups (control, cow, buffalo, goat and camel milk administrated groups) (10 rats/group); each animal was administrated by 3.4 ml/day. Rats were administered with milk for 6 weeks; at the end of the 5th week, five animals of each group were isolated and the remaining five animals were immunized with sheep red blood cells (SRBCs) and kept for another week to mount immune response. The effect of different milk types on rats' immune response towards SRBCs was evaluated through pro-inflammatory cytokines, antioxidants, ESR and CRP measurement; together, with the histopathological examination of spleen samples and hemagglutination assay. Camel milk consumption reduced oxidative stress and inflammation in spleen that resulted from SRBCs immunization; in addition to, B cell stimulation that was apparent from the high level of anti-SRBCs antibodies. Camel milk is recommended for newborn consumption, due to its high-water content, unsaturated fatty acids, and vitamin C, as well as low lactose and fat content.
Collapse
Affiliation(s)
- Maryam Amr
- Zoology Department, Faculty of Science, Cairo University, Cairo, Egypt
| | - Alyaa Farid
- Zoology Department, Faculty of Science, Cairo University, Cairo, Egypt.
| |
Collapse
|
4
|
Ji X, Wu L, Marion T, Luo Y. Lipid metabolism in regulation of B cell development and autoimmunity. Cytokine Growth Factor Rev 2023; 73:40-51. [PMID: 37419766 DOI: 10.1016/j.cytogfr.2023.06.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Accepted: 06/29/2023] [Indexed: 07/09/2023]
Abstract
B cells play an important role in adaptive immunity and participate in the process of humoral immunity mainly by secreting antibodies. The entire development and differentiation process of B cells occurs in multiple microenvironments and is regulated by a variety of environmental factors and immune signals. Differentiation biases or disfunction of B cells participate in the process of many autoimmune diseases. Emerging studies report the impact of altered metabolism in B cell biology, including lipid metabolism. Here, we discuss how extracellular lipid environment and metabolites, membrane lipid-related components, and lipid synthesis and catabolism programs coordinate B cell biology and describe the crosstalk of lipid metabolic programs with signal transduction pathways and transcription factors. We conclude with a summary of therapeutic targets for B cell lipid metabolism and signaling in autoimmune diseases and discuss important future directions.
Collapse
Affiliation(s)
- Xing Ji
- Laboratory of Rheumatology and Immunology, Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Liang Wu
- Laboratory of Rheumatology and Immunology, Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Tony Marion
- Department of Microbiology, Immunology, and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Yubin Luo
- Laboratory of Rheumatology and Immunology, Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
5
|
Patel D, Goruk S, Richard C, Field CJ. Combined Supplementation with Arachidonic and Docosahexaenoic Acids in T Helper Type-2 Skewed Brown Norway Rat Offspring is Beneficial in the Induction of Oral Tolerance toward Ovalbumin and Immune System Development. J Nutr 2022; 152:2165-2178. [PMID: 35648474 DOI: 10.1093/jn/nxac118] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 04/08/2022] [Accepted: 05/26/2022] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND A T helper type-2 (Th2) skewed immune response is associated with food allergies. DHA and arachidonic acid (ARA) have been shown to promote oral tolerance (OT) in healthy rodents. OBJECTIVES We studied the effect of combined ARA + DHA supplementation during the suckling and weaning periods on OT and immune system development in Th2-skewed Brown Norway rat offspring. METHODS Dams were fed ARA + DHA (0.45% ARA, 0.8% DHA wt/wt of total fat; n = 10) as a suckling period diet (SPD) or control SPD (0% ARA, 0% DHA, n = 8). At 3 wk, offspring from each SPD group received ARA + DHA (0.5% ARA, 0.5% DHA wt/wt of total fat) weaning diet (WD), or control until 8 wk. For OT, offspring were orally exposed to either ovalbumin (OVA) or placebo between 21 and 25 d, followed by systemic immunization with OVA + adjuvant at 7 wk. Primary outcomes, ex vivo cytokine production by splenocytes and plasma OVA-specific Igs, were analyzed using a 3-way ANOVA. RESULTS At 8 wk, despite no lasting effect of SPD on splenocytes fatty acids, ARA + DHA WD resulted in 2× higher DHA in splenocyte phospholipid compositions without affecting ARA. OT development was observed in OVA-exposed groups with 15% lower plasma OVA-IgE (P = 0.04) and 35% lower OVA-IgG1 (P = 0.01) than placebo. ARA + DHA SPD resulted in 35% lower OVA-IgG1 and iIL-6 (P = 0.04) when stimulated with LPS, and a higher proportion of mature B cells (OX12+, P = 0.0004, and IgG+, P = 0.008). ARA + DHA WD resulted in 20% higher Th1 cytokines (TNF-α and IFN-γ) production to lymphocyte stimulant and higher splenocyte proportion of CD45RA+ (pan-B cells) and OX6+ (dendritic cells) than control WD (P values < 0.05). CONCLUSIONS Combined supplementation of ARA and DHA is beneficial for OT development, especially in the suckling period. Further, ARA + DHA supplementation can also counteract the Th2-skewed immunity of Brown Norway rat offspring through higher Th1 cytokine production by lymphocytes.
Collapse
Affiliation(s)
- Dhruvesh Patel
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Susan Goruk
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Caroline Richard
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Catherine J Field
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
6
|
Lee J, Lee JK, Lee JJ, Park S, Jung S, Lee HJ, Ha JH. Partial Replacement of High-Fat Diet with Beef Tallow Attenuates Dyslipidemia and Endoplasmic Reticulum Stress in db/ db Mice. J Med Food 2022; 25:660-674. [PMID: 35617705 DOI: 10.1089/jmf.2022.k.0019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
High-fat diet (HFD) consumption is closely associated with an increased risk of metabolic syndromes (MetS), such as obesity, type 2 diabetes, and cardiovascular diseases (CVDs). Therefore, the consumption of alternative and functional fatty acids to replace saturated fatty acids and/or trans-fatty acids with polyunsaturated fatty acids has become an important dietary strategy for the prevention of MetS. Consumption of omega-3 fatty acids (n-3) reduces various physiological complications, including CVDs, nonalcoholic fatty liver disease, and insulin resistance, related to inflammatory responses. In this study, we investigated the partial replacement effects of HFD with beef tallow (BT) on dyslipidemia and endoplasmic reticulum (ER) stress in male db/db mice. The animals were grouped to one of four dietary intervention groups (n = 16 per group): (1) normal diet, (2) HFD, (3) HFD partially replaced with regular beef tallow (HFD+BT1), or (4) HFD partially replaced with beef tallow containing a relatively reduced omega-6 fatty acid (n-6)/n-3 ratio (HFD+BT2) than HFD+BT1. After 6 weeks of dietary intervention, 1 mg/kg of phosphate-buffered saline or tunicamycin (TM) was injected intraperitoneally. HFD+BT2 significantly suppressed the serum total cholesterol and non-high-density lipoprotein cholesterol levels more than HFD and HFD+BT1, and triglyceride levels in the epididymal adipose tissue (EAT) were remarkably decreased. Mice that received HFD+BT2 had elevated protein expressions of phospho-AMP-activated protein kinase (p-AMPK). Moreover, HFD+BT2 effectively inhibited ER stress in the liver and EAT. Consistent with our hypothesis, HFD+BT2 remarkably alleviated dyslipidemia and TM-inducible ER stress, while activating p-AMPK.
Collapse
Affiliation(s)
- Jisu Lee
- Department of Food Science and Nutrition, Dankook University, Cheonan, Korea
| | - Jennifer K Lee
- Food Science and Human Nutrition Department, University of Florida, Gainesville, Florida, USA
| | - Jae-Joon Lee
- Department of Food and Nutrition, Chosun University, Gwangju, Korea
| | - Seohyun Park
- Department of Food Science and Nutrition, Dankook University, Cheonan, Korea
| | - Sunyoon Jung
- Department of Food Science and Nutrition, Dankook University, Cheonan, Korea
| | - Hyun-Joo Lee
- Department of Nutrition and Culinary Science, Hankyong National University, Ansung, Korea
| | - Jung-Heun Ha
- Department of Food Science and Nutrition, Dankook University, Cheonan, Korea.,Research Center for Industrialization of Natural Neutralization, Dankook University, Yongin, Korea
| |
Collapse
|
7
|
Son HK, Xiang H, Park S, Lee J, Lee JJ, Jung S, Ha JH. Partial Replacement of Dietary Fat with Polyunsaturated Fatty Acids Attenuates the Lipopolysaccharide-Induced Hepatic Inflammation in Sprague-Dawley Rats Fed a High-Fat Diet. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph182010986. [PMID: 34682732 PMCID: PMC8535618 DOI: 10.3390/ijerph182010986] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 10/13/2021] [Accepted: 10/16/2021] [Indexed: 12/17/2022]
Abstract
In this study, we investigated whether the partial replacement of dietary fat with polyunsaturated fatty acids (PUFAs) ameliorated the lipopolysaccharide (LPS)-induced hepatic inflammation in rats fed a high-fat diet. Male Sprague-Dawley rats were divided into three groups and provided each of the following diets: (1) high-fat diet (HFD), (2) HFD with perilla oil (PO), and (3) HFD with corn oil (CO). After 12 weeks of dietary intervention, the rats were intraperitoneally injected with LPS (5 mg/kg) from Escherichia coli O55:B5 or phosphate-buffered saline (PBS). Following LPS stimulation, serum insulin levels were increased, while PO and CO lowered the serum levels of glucose and insulin. In the liver, LPS increased the triglyceride levels, while PO and CO alleviated the LPS-induced hepatic triglyceride accumulation. In the LPS injected rats, the mRNA expression of genes related to inflammation and endoplasmic reticulum (ER) stress was attenuated by PO and CO in the liver. Furthermore, hepatic levels of proteins involved in the nuclear factor kappa-light-chain-enhancer of activated B cells/mitogen-activated protein kinase pathways, antioxidant response, and ER stress were lowered by PO- and CO-replacement. Therefore, the partial replacement of dietary fat with PUFAs alleviates LPS-induced hepatic inflammation during HFD consumption, which may decrease metabolic abnormalities.
Collapse
Affiliation(s)
- Hee-Kyoung Son
- Research Center for Industrialization of Natural Neutralization, Dankook University, Cheonan 31116, Korea; (H.-K.S.); (H.X.); (S.P.); (J.L.)
| | - Huo Xiang
- Research Center for Industrialization of Natural Neutralization, Dankook University, Cheonan 31116, Korea; (H.-K.S.); (H.X.); (S.P.); (J.L.)
- Department of Food Science and Nutrition, Dankook University, Cheonan 31116, Korea
| | - Seohyun Park
- Research Center for Industrialization of Natural Neutralization, Dankook University, Cheonan 31116, Korea; (H.-K.S.); (H.X.); (S.P.); (J.L.)
- Department of Food Science and Nutrition, Dankook University, Cheonan 31116, Korea
| | - Jisu Lee
- Research Center for Industrialization of Natural Neutralization, Dankook University, Cheonan 31116, Korea; (H.-K.S.); (H.X.); (S.P.); (J.L.)
- Department of Food Science and Nutrition, Dankook University, Cheonan 31116, Korea
| | - Jae-Joon Lee
- Department of Food and Nutrition, Chosun University, Gwangju 61452, Korea;
| | - Sunyoon Jung
- Research Center for Industrialization of Natural Neutralization, Dankook University, Cheonan 31116, Korea; (H.-K.S.); (H.X.); (S.P.); (J.L.)
- Department of Food Science and Nutrition, Dankook University, Cheonan 31116, Korea
- Correspondence: (S.J.); (J.-H.H.); Tel.: +82-31-8005-3186 (S.J.); +82-41-550-3479 (J.-H.H.)
| | - Jung-Heun Ha
- Research Center for Industrialization of Natural Neutralization, Dankook University, Cheonan 31116, Korea; (H.-K.S.); (H.X.); (S.P.); (J.L.)
- Department of Food Science and Nutrition, Dankook University, Cheonan 31116, Korea
- Correspondence: (S.J.); (J.-H.H.); Tel.: +82-31-8005-3186 (S.J.); +82-41-550-3479 (J.-H.H.)
| |
Collapse
|
8
|
Kobayashi A, Ito A, Shirakawa I, Tamura A, Tomono S, Shindou H, Hedde PN, Tanaka M, Tsuboi N, Ishimoto T, Akashi-Takamura S, Maruyama S, Suganami T. Dietary Supplementation With Eicosapentaenoic Acid Inhibits Plasma Cell Differentiation and Attenuates Lupus Autoimmunity. Front Immunol 2021; 12:650856. [PMID: 34211460 PMCID: PMC8240640 DOI: 10.3389/fimmu.2021.650856] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 05/26/2021] [Indexed: 12/31/2022] Open
Abstract
Accumulating evidence suggests that cholesterol accumulation in leukocytes is causally associated with the development of autoimmune diseases. However, the mechanism by which fatty acid composition influences autoimmune responses remains unclear. To determine whether the fatty acid composition of diet modulates leukocyte function and the development of systemic lupus erythematosus, we examined the effect of eicosapentaenoic acid (EPA) on the pathology of lupus in drug-induced and spontaneous mouse models. We found that dietary EPA supplementation ameliorated representative lupus manifestations, including autoantibody production and immunocomplex deposition in the kidneys. A combination of lipidomic and membrane dynamics analyses revealed that EPA remodels the lipid composition and fluidity of B cell membranes, thereby preventing B cell differentiation into autoantibody-producing plasma cells. These results highlight a previously unrecognized mechanism by which fatty acid composition affects B cell differentiation into autoantibody-producing plasma cells during autoimmunity, and imply that EPA supplementation may be beneficial for therapy of lupus.
Collapse
Affiliation(s)
- Azusa Kobayashi
- Department of Molecular Medicine and Metabolism, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Ayaka Ito
- Department of Molecular Medicine and Metabolism, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
- Department of Immunometabolism, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Ibuki Shirakawa
- Department of Molecular Medicine and Metabolism, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
- Department of Immunometabolism, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Atsushi Tamura
- Department of Organic Biomaterials, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Susumu Tomono
- Department of Microbiology and Immunology, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Hideo Shindou
- Department of Lipid Signaling, National Center for Global Health and Medicine, Tokyo, Japan
- Department of Medical Lipid Science, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Per Niklas Hedde
- Laboratory for Fluorescence Dynamics, Beckman Laser Institute and Medical Clinic, Department of Pharmaceutical Sciences, University of California Irvine, Irvine, CA, United States
| | - Miyako Tanaka
- Department of Molecular Medicine and Metabolism, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
- Department of Immunometabolism, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Naotake Tsuboi
- Department of Nephrology, Fujita Health University Graduate School of Medicine, Toyoake, Japan
| | - Takuji Ishimoto
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Sachiko Akashi-Takamura
- Department of Microbiology and Immunology, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Shoichi Maruyama
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takayoshi Suganami
- Department of Molecular Medicine and Metabolism, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
- Department of Immunometabolism, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
9
|
Tourkochristou E, Triantos C, Mouzaki A. The Influence of Nutritional Factors on Immunological Outcomes. Front Immunol 2021; 12:665968. [PMID: 34135894 PMCID: PMC8201077 DOI: 10.3389/fimmu.2021.665968] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 05/05/2021] [Indexed: 12/19/2022] Open
Abstract
Through food intake, humans obtain a variety of nutrients that are essential for growth, cellular function, tissue development, energy, and immune defense. A special interaction between nutrients and gut-associated lymphoid tissue occurs in the intestinal tract. Enterocytes of the intestinal barrier act as sensors for antigens from nutrients and the intestinal microbiota, which they deliver to the underlying immune system of the lamina propria, triggering an immune response. Studies investigating the mechanism of influence of nutrition on immunological outcomes have highlighted an important role of macronutrients (proteins, carbohydrates, fatty acids) and micronutrients (vitamins, minerals, phytochemicals, antioxidants, probiotics) in modulating immune homeostasis. Nutrients exert their role in innate immunity and inflammation by regulating the expression of TLRs, pro- and anti-inflammatory cytokines, thus interfering with immune cell crosstalk and signaling. Chemical substrates derived from nutrient metabolism may act as cofactors or blockers of enzymatic activity, influencing molecular pathways and chemical reactions associated with microbial killing, inflammation, and oxidative stress. Immune cell function appears to be influenced by certain nutrients that form parts of the cell membrane structure and are involved in energy production and prevention of cytotoxicity. Nutrients also contribute to the initiation and regulation of adaptive immune responses by modulating B and T lymphocyte differentiation, proliferation and activation, and antibody production. The purpose of this review is to present the available data from the field of nutritional immunology to elucidate the complex and dynamic relationship between nutrients and the immune system, the delineation of which will lead to optimized nutritional regimens for disease prevention and patient care.
Collapse
Affiliation(s)
- Evanthia Tourkochristou
- Division of Hematology, Department of Internal Medicine, Medical School, University of Patras, Patras, Greece
| | - Christos Triantos
- Division of Gastroenterology, Department of Internal Medicine, Medical School, University of Patras, Patras, Greece
| | - Athanasia Mouzaki
- Division of Hematology, Department of Internal Medicine, Medical School, University of Patras, Patras, Greece
| |
Collapse
|
10
|
Feng C, Li L, Li Q, Switzer K, Liu M, Han S, Zheng B. Docosahexaenoic acid ameliorates autoimmune inflammation by activating GPR120 signaling pathway in dendritic cells. Int Immunopharmacol 2021; 97:107698. [PMID: 33932699 DOI: 10.1016/j.intimp.2021.107698] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 04/13/2021] [Accepted: 04/18/2021] [Indexed: 12/19/2022]
Abstract
Although the phenomenon that omega-3 polyunsaturated fatty acids (n-3 PUFAs) shows to have a beneficial effect in patients suffering from multiple sclerosis (MS) and other autoimmune diseases has been empirically well-documented, the molecular mechanisms that underline the anti-inflammatory effects of n-3 PUFAs are yet to be understood. In experimental autoimmune encephalomyelitis (EAE), a model for MS, we show that one of the underlying mechanisms by which dietary docosahexaenoic acid (DHA) exerts its anti-inflammatory effect is regulating the functional activities of dendritic cells (DCs). In DHA-treated EAE mice, DCs acquire a regulatory phenotype characterized by low expression of co-stimulatory molecules, decreased production of pro-inflammatory cytokines, and enhanced capability of regulatory T-cell induction. The effect of DHA on DCs is mediated by the lipid-sensing receptor, G protein-coupled receptor 120 (GPR120). A GPR120-specific small-molecule agonist could ameliorate the autoimmune inflammation by regulating DCs, while silencing GPR120 in DCs strongly increased the immunogenicity of DCs. Stimulation of GPR120 induces suppressor of cytokine signaling 3 (SOCS3) expression and down-regulates signal transducer and activator of transcription 3 (STAT3) phosphorylation, explaining the molecular mechanism for regulatory DC induction.
Collapse
Affiliation(s)
- Chunlei Feng
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Lingyun Li
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Qing Li
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Kirsten Switzer
- Department of Pathology & Immunology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
| | - Mingyao Liu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Shuhua Han
- Department of Pathology & Immunology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
| | - Biao Zheng
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China; Department of Pathology & Immunology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States.
| |
Collapse
|
11
|
Darwesh AM, Bassiouni W, Sosnowski DK, Seubert JM. Can N-3 polyunsaturated fatty acids be considered a potential adjuvant therapy for COVID-19-associated cardiovascular complications? Pharmacol Ther 2021; 219:107703. [PMID: 33031856 PMCID: PMC7534795 DOI: 10.1016/j.pharmthera.2020.107703] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 09/28/2020] [Indexed: 02/06/2023]
Abstract
Coronavirus disease 2019 (COVID-19), caused by the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), has currently led to a global pandemic with millions of confirmed and increasing cases around the world. The novel SARS-CoV-2 not only affects the lungs causing severe acute respiratory dysfunction but also leads to significant dysfunction in multiple organs and physiological systems including the cardiovascular system. A plethora of studies have shown the viral infection triggers an exaggerated immune response, hypercoagulation and oxidative stress, which contribute significantly to poor cardiovascular outcomes observed in COVID-19 patients. To date, there are no approved vaccines or therapies for COVID-19. Accordingly, cardiovascular protective and supportive therapies are urgent and necessary to the overall prognosis of COVID-19 patients. Accumulating literature has demonstrated the beneficial effects of n-3 polyunsaturated fatty acids (n-3 PUFA) toward the cardiovascular system, which include ameliorating uncontrolled inflammatory reactions, reduced oxidative stress and mitigating coagulopathy. Moreover, it has been demonstrated the n-3 PUFAs, eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), are precursors to a group of potent bioactive lipid mediators, generated endogenously, which mediate many of the beneficial effects attributed to their parent compounds. Considering the favorable safety profile for n-3 PUFAs and their metabolites, it is reasonable to consider n-3 PUFAs as potential adjuvant therapies for the clinical management of COVID-19 patients. In this article, we provide an overview of the pathogenesis of cardiovascular complications secondary to COVID-19 and focus on the mechanisms that may contribute to the likely benefits of n-3 PUFAs and their metabolites.
Collapse
Affiliation(s)
- Ahmed M Darwesh
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - Wesam Bassiouni
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Deanna K Sosnowski
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - John M Seubert
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada; Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
12
|
Kobayashi S, Phung HT, Tayama S, Kagawa Y, Miyazaki H, Yamamoto Y, Maruyama T, Ishii N, Owada Y. Fatty acid-binding protein 3 regulates differentiation of IgM-producing plasma cells. FEBS J 2021; 288:1130-1141. [PMID: 32578350 DOI: 10.1111/febs.15460] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 05/26/2020] [Accepted: 06/09/2020] [Indexed: 01/02/2023]
Abstract
Plasma cells (PCs), which aim to protect host health, produce various subsets of immunoglobulin (Ig) in response to extracellular pathogens. Blimp-1 (encoded by Prdm1)-a protein that is highly expressed by PCs-is important for PC functions, including the generation of Igs. Fatty acid-binding protein 3 (FABP3) is a carrier protein of polyunsaturated fatty acids (PUFAs) and participates in multiple cellular functions. Although the functions of FABP3 in neurons and cardiac myocytes are well-noted, their roles in immune cells remain to be fully elucidated. In this study, we demonstrate that FABP3 is expressed in activated B cells and that FABP3 promotes PC development and IgM secretion. Moreover, we provide the first evidence that FABP3 is necessary for Blimp-1 expression, by regulating the histone modification of its promoter region. Taken together, our findings reveal that FABP3 acts as a positive regulator of B-cell activation by controlling histone acetylation of the Blimp-1 gene, thereby playing a role in host defense against pathogens.
Collapse
Affiliation(s)
- Shuhei Kobayashi
- Department of Organ Anatomy, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hai The Phung
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Shunichi Tayama
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yoshiteru Kagawa
- Department of Organ Anatomy, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hirofumi Miyazaki
- Department of Organ Anatomy, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yui Yamamoto
- Department of Organ Anatomy, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takashi Maruyama
- Mucosal Immunology Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Naoto Ishii
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yuji Owada
- Department of Organ Anatomy, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
13
|
Wu Y, Zhao J, Xu C, Ma N, He T, Zhao J, Ma X, Thacker PA. Progress towards pig nutrition in the last 27 years. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2020; 100:5102-5110. [PMID: 29691867 DOI: 10.1002/jsfa.9095] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 04/05/2018] [Accepted: 04/19/2018] [Indexed: 06/08/2023]
Abstract
Over the last 27 years (1990-2017), based on the revolutionary progresses of basic nutrition research, novel methods and techniques have been developed which bring a profound technological revolution to pig production from free-range system to intensive farming all over the world. Basic theoretical innovations and feed production studies have provided vital advancements in pig nutrition by developing formula feed, utilizing balanced diets, determining feed energy value, dividing pig physiological stages, enhancing gut health, and improving feed processing technique. Formula feed is the primary contributor of the rise of the mechanized farming industry, and meets comprehensive nutritional needs of the pig. The focuses of the development of a balanced diet by optimizing nutrient levels are the amino acids balance, the balance between amino acids and energy, the balance between calcium and phosphorus. Multiple-site-production and targeted feeding program have been applied extensively. Early weaning of piglets improves production efficiency, but piglets that have not yet fully developed their intestine are prone to diarrhea. Therefore, intestinal health has received special attention in recent years. Feed processing technologies, such as granulation, puffing, fermentation and enzymatic hydrolysis, can improve the utilization of feed nutrients and reduce production cost. However, increasing a sow's potential for production, seeking alternatives to antibiotics, reducing drug treatment in piglets, developing functional additives and improving meat quality remain future challenges. Herein, we outline the important progresses of pig nutrition in the past 27 years, which will shed light on the basic nutrition rules of pig production, and help to push forward its future development. © 2018 Society of Chemical Industry.
Collapse
Affiliation(s)
- Yi Wu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Jianfei Zhao
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Chenchen Xu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Ning Ma
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Ting He
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Jinshan Zhao
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao, China
| | - Xi Ma
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
- Department of Internal Medicine and Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, USA
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao, China
| | - Phil A Thacker
- Department of Animal and Poultry Science, University of Saskatchewan, Saskatoon, Canada
| |
Collapse
|
14
|
Bachmann MC, Bellalta S, Basoalto R, Gómez-Valenzuela F, Jalil Y, Lépez M, Matamoros A, von Bernhardi R. The Challenge by Multiple Environmental and Biological Factors Induce Inflammation in Aging: Their Role in the Promotion of Chronic Disease. Front Immunol 2020; 11:570083. [PMID: 33162985 PMCID: PMC7591463 DOI: 10.3389/fimmu.2020.570083] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Accepted: 09/24/2020] [Indexed: 12/13/2022] Open
Abstract
The aging process is driven by multiple mechanisms that lead to changes in energy production, oxidative stress, homeostatic dysregulation and eventually to loss of functionality and increased disease susceptibility. Most aged individuals develop chronic low-grade inflammation, which is an important risk factor for morbidity, physical and cognitive impairment, frailty, and death. At any age, chronic inflammatory diseases are major causes of morbimortality, affecting up to 5-8% of the population of industrialized countries. Several environmental factors can play an important role for modifying the inflammatory state. Genetics accounts for only a small fraction of chronic-inflammatory diseases, whereas environmental factors appear to participate, either with a causative or a promotional role in 50% to 75% of patients. Several of those changes depend on epigenetic changes that will further modify the individual response to additional stimuli. The interaction between inflammation and the environment offers important insights on aging and health. These conditions, often depending on the individual's sex, appear to lead to decreased longevity and physical and cognitive decline. In addition to biological factors, the environment is also involved in the generation of psychological and social context leading to stress. Poor psychological environments and other sources of stress also result in increased inflammation. However, the mechanisms underlying the role of environmental and psychosocial factors and nutrition on the regulation of inflammation, and how the response elicited for those factors interact among them, are poorly understood. Whereas certain deleterious environmental factors result in the generation of oxidative stress driven by an increased production of reactive oxygen and nitrogen species, endoplasmic reticulum stress, and inflammation, other factors, including nutrition (polyunsaturated fatty acids) and behavioral factors (exercise) confer protection against inflammation, oxidative and endoplasmic reticulum stress, and thus ameliorate their deleterious effect. Here, we discuss processes and mechanisms of inflammation associated with environmental factors and behavior, their links to sex and gender, and their overall impact on aging.
Collapse
Affiliation(s)
| | - Sofía Bellalta
- School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Roque Basoalto
- School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | | | - Yorschua Jalil
- School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Macarena Lépez
- School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Anibal Matamoros
- School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile.,Institute of Biological Sciences (ICB), Federal University of Pará, Belem, Brazil
| | - Rommy von Bernhardi
- School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
15
|
Radzikowska U, Rinaldi AO, Çelebi Sözener Z, Karaguzel D, Wojcik M, Cypryk K, Akdis M, Akdis CA, Sokolowska M. The Influence of Dietary Fatty Acids on Immune Responses. Nutrients 2019; 11:E2990. [PMID: 31817726 PMCID: PMC6950146 DOI: 10.3390/nu11122990] [Citation(s) in RCA: 185] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 11/25/2019] [Accepted: 12/02/2019] [Indexed: 12/16/2022] Open
Abstract
Diet-derived fatty acids (FAs) are essential sources of energy and fundamental structural components of cells. They also play important roles in the modulation of immune responses in health and disease. Saturated and unsaturated FAs influence the effector and regulatory functions of innate and adaptive immune cells by changing membrane composition and fluidity and by acting through specific receptors. Impaired balance of saturated/unsaturated FAs, as well as n-6/n-3 polyunsaturated FAs has significant consequences on immune system homeostasis, contributing to the development of many allergic, autoimmune, and metabolic diseases. In this paper, we discuss up-to-date knowledge and the clinical relevance of the influence of dietary FAs on the biology, homeostasis, and functions of epithelial cells, macrophages, dendritic cells, neutrophils, innate lymphoid cells, T cells and B cells. Additionally, we review the effects of dietary FAs on the pathogenesis of many diseases, including asthma, allergic rhinitis, food allergy, atopic dermatitis, rheumatoid arthritis, multiple sclerosis as well as type 1 and 2 diabetes.
Collapse
Affiliation(s)
- Urszula Radzikowska
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, 7265 Davos Wolfgang, Switzerland
- Christine Kühne-Center for Allergy Research and Education, 7265 Davos Wolfgang, Switzerland
- Department of Regenerative Medicine and Immune Regulation, Medical University of Bialystok, 15-269 Bialystok, Poland
| | - Arturo O Rinaldi
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, 7265 Davos Wolfgang, Switzerland
- Christine Kühne-Center for Allergy Research and Education, 7265 Davos Wolfgang, Switzerland
| | - Zeynep Çelebi Sözener
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, 7265 Davos Wolfgang, Switzerland
- Department of Chest Disease, Division of Allergy and Clinical Immunology, Ankara University School of Medicine, 06100 Ankara, Turkey
| | - Dilara Karaguzel
- Department of Biology, Faculty of Science, Hacettepe University, 06800 Ankara, Turkey
| | - Marzena Wojcik
- Department of Structural Biology, Medical University of Lodz, 90-752 Lodz, Poland
| | - Katarzyna Cypryk
- Department of Internal Medicine and Diabetology, Medical University of Lodz, 90-549 Lodz, Poland
| | - Mübeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, 7265 Davos Wolfgang, Switzerland
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, 7265 Davos Wolfgang, Switzerland
- Christine Kühne-Center for Allergy Research and Education, 7265 Davos Wolfgang, Switzerland
| | - Milena Sokolowska
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, 7265 Davos Wolfgang, Switzerland
- Christine Kühne-Center for Allergy Research and Education, 7265 Davos Wolfgang, Switzerland
| |
Collapse
|
16
|
Ma C, Vasu R, Zhang H. The Role of Long-Chain Fatty Acids in Inflammatory Bowel Disease. Mediators Inflamm 2019; 2019:8495913. [PMID: 31780872 PMCID: PMC6874876 DOI: 10.1155/2019/8495913] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 10/03/2019] [Indexed: 02/05/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a complicated disease involving multiple pathogenic factors. The complex relationships between long-chain fatty acids (LCFAs) and the morbidity of IBD drive numerous studies to unravel the underlying mechanisms. A better understanding of the role of LCFAs in IBD will substitute or boost the current IBD therapies, thereby obtaining mucosal healing. In this review, we focused on the roles of LCFAs on the important links of inflammatory regulation in IBD, including in the pathogen recognition phase and in the inflammatory resolving phase, and the effects of LCFAs on immune cells in IBD.
Collapse
Affiliation(s)
- Chunxiang Ma
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
- Centre for Inflammatory Bowel Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Reshma Vasu
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Hu Zhang
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
- Centre for Inflammatory Bowel Disease, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
17
|
Gutiérrez S, Svahn SL, Johansson ME. Effects of Omega-3 Fatty Acids on Immune Cells. Int J Mol Sci 2019; 20:ijms20205028. [PMID: 31614433 PMCID: PMC6834330 DOI: 10.3390/ijms20205028] [Citation(s) in RCA: 320] [Impact Index Per Article: 53.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 09/27/2019] [Accepted: 10/02/2019] [Indexed: 12/14/2022] Open
Abstract
Alterations on the immune system caused by omega-3 fatty acids have been described for 30 years. This family of polyunsaturated fatty acids exerts major alterations on the activation of cells from both the innate and the adaptive immune system, although the mechanisms for such regulation are diverse. First, as a constitutive part of the cellular membrane, omega-3 fatty acids can regulate cellular membrane properties, such as membrane fluidity or complex assembly in lipid rafts. In recent years, however, a new role for omega-3 fatty acids and their derivatives as signaling molecules has emerged. In this review, we describe the latest findings describing the effects of omega-3 fatty acids on different cells from the immune system and their possible molecular mechanisms.
Collapse
Affiliation(s)
- Saray Gutiérrez
- Department of Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden.
| | - Sara L Svahn
- Department of Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden.
| | - Maria E Johansson
- Department of Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden.
| |
Collapse
|
18
|
Crouch MJ, Kosaraju R, Guesdon W, Armstrong M, Reisdorph N, Jain R, Fenton J, Shaikh SR. Frontline Science: A reduction in DHA-derived mediators in male obesity contributes toward defects in select B cell subsets and circulating antibody. J Leukoc Biol 2019; 106:241-257. [PMID: 30576001 PMCID: PMC10020993 DOI: 10.1002/jlb.3hi1017-405rr] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 11/20/2018] [Accepted: 12/11/2018] [Indexed: 01/25/2023] Open
Abstract
Obesity dysregulates B cell populations, which contributes toward poor immunological outcomes. We previously reported that differing B cell subsets are lowered in the bone marrow of obese male mice. Here, we focused on how lipid metabolites synthesized from docosahexaenoic acid (DHA) known as specialized pro-resolving lipid mediators (SPMs) influence specific B cell populations in obese male mice. Metabololipidomics revealed that splenic SPM precursors 14-hydroxydocosahexaenoic acid (14-HDHA), 17-hydroxydocosahexaenoic acid (17-HDHA), and downstream protectin DX (PDX) were decreased in obese male C57BL/6J mice. Simultaneous administration of these mediators to obese mice rescued major decrements in bone marrow B cells, modest impairments in the spleen, and circulating IgG2c, which is pro-inflammatory in obesity. In vitro studies with B cells, flow cytometry experiments with ALOX5-/- mice, and lipidomic analyses revealed the lowering of 14-HDHA/17-HDHA/PDX and dysregulation of B cell populations in obesity was driven indirectly via B cell extrinsic mechanisms. Notably, the lowering of lipid mediators was associated with an increase in the abundance of n-6 polyunsaturated fatty acids, which have a high affinity for SPM-generating enzymes. Subsequent experiments revealed female obese mice generally maintained the levels of SPM precursors, B cell subsets, and antibody levels. Finally, obese human females had increased circulating plasma cells accompanied by ex vivo B cell TNFα and IL-10 secretion. Collectively, the data demonstrate that DHA-derived mediators of the SPM pathway control the number of B cell subsets and pro-inflammatory antibody levels in obese male but not female mice through a defect that is extrinsic to B cells.
Collapse
Affiliation(s)
- Miranda J Crouch
- Department of Biochemistry & Molecular Biology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, USA.,East Carolina Diabetes & Obesity Institute, East Carolina University, Greenville, North Carolina, USA.,Department of Nutrition, Gillings School of Global Public Health and School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Rasagna Kosaraju
- Department of Biochemistry & Molecular Biology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, USA.,East Carolina Diabetes & Obesity Institute, East Carolina University, Greenville, North Carolina, USA
| | - William Guesdon
- Department of Biochemistry & Molecular Biology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, USA.,East Carolina Diabetes & Obesity Institute, East Carolina University, Greenville, North Carolina, USA
| | - Michael Armstrong
- Department of Pharmaceutical Sciences, University of Colorado, Denver, Colorado, USA
| | - Nichole Reisdorph
- Department of Pharmaceutical Sciences, University of Colorado, Denver, Colorado, USA
| | - Raghav Jain
- The College of Osteopathic Medicine, Michigan State University, East Lansing, Michigan, USA
| | - Jenifer Fenton
- The College of Osteopathic Medicine, Michigan State University, East Lansing, Michigan, USA
| | - Saame Raza Shaikh
- Department of Nutrition, Gillings School of Global Public Health and School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
19
|
Darwesh AM, Sosnowski DK, Lee TYT, Keshavarz-Bahaghighat H, Seubert JM. Insights into the cardioprotective properties of n-3 PUFAs against ischemic heart disease via modulation of the innate immune system. Chem Biol Interact 2019; 308:20-44. [DOI: 10.1016/j.cbi.2019.04.037] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 04/17/2019] [Accepted: 04/30/2019] [Indexed: 12/19/2022]
|
20
|
Petta I, Fraussen J, Somers V, Kleinewietfeld M. Interrelation of Diet, Gut Microbiome, and Autoantibody Production. Front Immunol 2018; 9:439. [PMID: 29559977 PMCID: PMC5845559 DOI: 10.3389/fimmu.2018.00439] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 02/19/2018] [Indexed: 12/12/2022] Open
Abstract
B cells possess a predominant role in adaptive immune responses via antibody-dependent and -independent functions. The microbiome of the gastrointestinal tract is currently being intensively investigated due to its profound impact on various immune responses, including B cell maturation, activation, and IgA antibody responses. Recent findings have demonstrated the interplay between dietary components, gut microbiome, and autoantibody production. "Western" dietary patterns, such as high fat and high salt diets, can induce alterations in the gut microbiome that in turn affects IgA responses and the production of autoantibodies. This could contribute to multiple pathologies including autoimmune and inflammatory diseases. Here, we summarize current knowledge on the influence of various dietary components on B cell function and (auto)antibody production in relation to the gut microbiota, with a particular focus on the gut-brain axis in the pathogenesis of multiple sclerosis.
Collapse
Affiliation(s)
- Ioanna Petta
- VIB Laboratory of Translational Immunomodulation, Center for Inflammation Research, Hasselt University, Diepenbeek, Belgium.,Biomedical Research Institute, Hasselt University, and School of Life Sciences, Transnationale Universiteit Limburg, Hasselt, Belgium
| | - Judith Fraussen
- Biomedical Research Institute, Hasselt University, and School of Life Sciences, Transnationale Universiteit Limburg, Hasselt, Belgium
| | - Veerle Somers
- Biomedical Research Institute, Hasselt University, and School of Life Sciences, Transnationale Universiteit Limburg, Hasselt, Belgium
| | - Markus Kleinewietfeld
- VIB Laboratory of Translational Immunomodulation, Center for Inflammation Research, Hasselt University, Diepenbeek, Belgium.,Biomedical Research Institute, Hasselt University, and School of Life Sciences, Transnationale Universiteit Limburg, Hasselt, Belgium
| |
Collapse
|
21
|
RostamiRad A, Ebrahimi SSS, Sadeghi A, Taghikhani M, Meshkani R. Palmitate-induced impairment of autophagy turnover leads to increased apoptosis and inflammation in peripheral blood mononuclear cells. Immunobiology 2018; 223:269-278. [DOI: 10.1016/j.imbio.2017.10.041] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 10/14/2017] [Indexed: 12/20/2022]
|
22
|
Jones GJB, Roper RL. The effects of diets enriched in omega-3 polyunsaturated fatty acids on systemic vaccinia virus infection. Sci Rep 2017; 7:15999. [PMID: 29167527 PMCID: PMC5700085 DOI: 10.1038/s41598-017-16098-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 11/03/2017] [Indexed: 01/01/2023] Open
Abstract
Omega-3 polyunsaturated fatty acids (PUFA, n-3 fatty acids), the key components of fish and flaxseed oils, are increasingly consumed by the public because of their potential health benefits and are available by prescription for hypertriglyceridemia. However, numerous studies have shown that these compounds are immunoregulatory and immunosuppressive and thus may increase susceptibility to infection. In this study, we tested the effects of the amount of fat and the types of fatty acid in the diet on infection by vaccinia virus, an acute infection that begins in the respiratory tract and spreads by viremia to internal organs. Male C57Bl6 mice (~5 week old) were fed for 3 weeks prior to infection and continuing during infection and recovery one of the following: 1) a normal low fat (13% kcal) diet, 2) a low fat diet containing n-3 PUFAs, 3) a high fat (41% kcal) diet rich in n-3 PUFAs, 4) a high fat n-6 PUFA diet, or 5) a high fat monounsaturated diet. We found no statistically significant differences in the susceptibility of mice to viral infection, morbidity, viral organ titers, recovery time, or mortality with these diets, indicating that, over this approximately 6-week time period, dietary fats did not substantially affect responses to poxviral infection.
Collapse
Affiliation(s)
- Gwendolyn J B Jones
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC, 27834, USA
| | - Rachel L Roper
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC, 27834, USA. .,East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University 600 Moye Blvd, Greenville, NC, 27834, United States of America.
| |
Collapse
|
23
|
Guesdon W, Kosaraju R, Brophy P, Clark A, Dillingham S, Aziz S, Moyer F, Willson K, Dick JR, Patil SP, Balestrieri N, Armstrong M, Reisdroph N, Shaikh SR. Effects of fish oils on ex vivo B-cell responses of obese subjects upon BCR/TLR stimulation: a pilot study. J Nutr Biochem 2017; 53:72-80. [PMID: 29195133 DOI: 10.1016/j.jnutbio.2017.10.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 10/06/2017] [Accepted: 10/16/2017] [Indexed: 12/29/2022]
Abstract
The long-chain n-3 polyunsaturated fatty acids (LC-PUFAs) eicosapentaenoic (EPA) and docosahexaenoic acid (DHA) in fish oil have immunomodulatory properties. B cells are a poorly studied target of EPA/DHA in humans. Therefore, in this pilot study, we tested how n-3 LC-PUFAs influence B-cell responses of obese humans. Obese men and women were assigned to consume four 1-g capsules per day of olive oil (OO, n=12), fish oil (FO, n=12) concentrate or high-DHA-FO concentrate (n=10) for 12 weeks in a parallel design. Relative to baseline, FO (n=9) lowered the percentage of circulating memory and plasma B cells, whereas the other supplements had no effect. There were no postintervention differences between the three supplements. Next, ex vivo B-cell cytokines were assayed after stimulation of Toll-like receptors (TLRs) and/or the B-cell receptor (BCR) to determine if the effects of n-3 LC-PUFAs were pathway-dependent. B-cell IL-10 and TNFα secretion was respectively increased with high DHA-FO (n=10), relative to baseline, with respective TLR9 and TLR9+BCR stimulation. OO (n=12) and FO (n=12) had no influence on B-cell cytokines compared to baseline, and there were no differences in postintervention cytokine levels between treatment groups. Finally, ex vivo antibody levels were assayed with FO (n=7) after TLR9+BCR stimulation. Compared to baseline, FO lowered IgM but not IgG levels accompanied by select modifications to the plasma lipidome. Altogether, the results suggest that n-3 LC-PUFAs could modulate B-cell activity in humans, which will require further testing in a larger cohort.
Collapse
Affiliation(s)
- William Guesdon
- Department of Biochemistry & Molecular Biology, Brody School of Medicine, East Carolina University; East Carolina Diabetes & Obesity Institute, East Carolina University
| | - Rasagna Kosaraju
- Department of Biochemistry & Molecular Biology, Brody School of Medicine, East Carolina University; East Carolina Diabetes & Obesity Institute, East Carolina University
| | - Patricia Brophy
- East Carolina Diabetes & Obesity Institute, East Carolina University
| | - Angela Clark
- East Carolina Diabetes & Obesity Institute, East Carolina University
| | | | - Shahnaz Aziz
- Department of Psychology, East Carolina University
| | - Fiona Moyer
- Department of Psychology, East Carolina University
| | - Kate Willson
- Department of Nutrition Science, East Carolina University
| | - James R Dick
- Institute of Aquaculture, University of Stirling, UK
| | | | - Nicholas Balestrieri
- Department of Biochemistry & Molecular Biology, Brody School of Medicine, East Carolina University
| | - Michael Armstrong
- Department of Pharmaceutical Sciences, University of Colorado, Denver, CO
| | - Nichole Reisdroph
- Department of Pharmaceutical Sciences, University of Colorado, Denver, CO
| | - Saame Raza Shaikh
- Department of Biochemistry & Molecular Biology, Brody School of Medicine, East Carolina University; East Carolina Diabetes & Obesity Institute, East Carolina University; Department of Nutrition, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill.
| |
Collapse
|
24
|
Arnemo M, Kavaliauskis A, Andresen AMS, Bou M, Berge GM, Ruyter B, Gjøen T. Effects of dietary n-3 fatty acids on Toll-like receptor activation in primary leucocytes from Atlantic salmon (Salmo salar). FISH PHYSIOLOGY AND BIOCHEMISTRY 2017; 43:1065-1080. [PMID: 28280951 DOI: 10.1007/s10695-017-0353-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 02/05/2017] [Indexed: 06/06/2023]
Abstract
The shortage of the n-3 fatty acids eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) on the international markets has led to increasing substitution of fish oil by plant oils in Atlantic salmon (Salmo salar) feed and thereby reducing the EPA and DHA content in salmon. However, the minimum required levels of these fatty acids in fish diets for securing fish health are unknown. Fish were fed with 0, 1 or 2% EPA or DHA alone or in combination of both over a period, growing from 50 to 400 g. Primary head kidney leucocytes were isolated and stimulated with Toll-like receptor (TLR) ligands to determine if EPA and DHA deficiency can affect expression of important immune genes and eicosanoid production. Several genes related to viral immune response did not vary between groups. However, there was a tendency that the high-level EPA and DHA groups expressed lower levels of IL-1β in non-stimulated leucocytes. These leucocytes were also more responsive to the TLR ligands, inducing higher expression levels of IL-1β and Mx1 after stimulation. The levels of prostaglandin E2 and leukotriene B4 in serum and media from stimulated leucocytes were lower in both low and high EPA and DHA groups. In conclusion, leucocytes from low EPA and DHA groups seemed to be less responsive towards immunostimulants, like TLR ligands, indicating that low levels or absence of dietary EPA and DHA may have immunosuppressive effects.
Collapse
Affiliation(s)
- Marianne Arnemo
- Department of Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, PO Box 1068, Blindern, 0316, Oslo, Norway
| | - Arturas Kavaliauskis
- Department of Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, PO Box 1068, Blindern, 0316, Oslo, Norway
| | | | - Marta Bou
- Nofima, P. O. Box 210, 1431, Ås, Norway
| | | | | | - Tor Gjøen
- Department of Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, PO Box 1068, Blindern, 0316, Oslo, Norway.
| |
Collapse
|
25
|
Mehdizadeh A, Bonyadi M, Darabi M, Rahbarghazi R, Montazersaheb S, Velaei K, Shaaker M, Somi MH. Common chemotherapeutic agents modulate fatty acid distribution in human hepatocellular carcinoma and colorectal cancer cells. ACTA ACUST UNITED AC 2017; 7:31-39. [PMID: 28546951 PMCID: PMC5439387 DOI: 10.15171/bi.2017.05] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 01/18/2017] [Accepted: 02/01/2017] [Indexed: 12/17/2022]
Abstract
![]()
Introduction: Cancer cells are critically correlated with lipid molecules, particularly fatty acids, as structural blocks for membrane building, energy sources, and related signaling molecules. Therefore, cancer progression is in direct correlation with fatty acid metabolism. The aim of this study was to investigate the potential effects of common chemotherapeutic agents on the lipid metabolism of hepatocellular carcinoma (HCC) and colorectal cancer (CRC) cells, with a focus on alterations in cellular fatty acid contents.
Methods: Human HepG2 and SW480 cell lines as HCC and CRC cells were respectively cultured in RPMI-1640 medium supplemented with non-toxic doses of 5-fluorouracil and doxorubicin for 72 hours. Oil Red O dye was used to estimate intracellular lipid vacuole intensity. Fatty acid analysis of isolated membrane phospholipids and cytoplasmic triglycerides (TG) was performed by gas-liquid chromatography (GLC) technique.
Results: Oil red O staining represented significantly higher lipid accumulation and density in cancer cells after exposure to the chemotherapeutic agents as compared to non-treated control cells. Doxorubicin and 5-fluorouracil treatment promoted the channeling of saturated fatty acids (SFAs) from phospholipids to triglyceride pool in both HepG2 (+5.91% and +8.50%, P < 0.05, respectively) and SW480 (+37.41% and +5.73%, P < 0.05, respectively) cell lines. However, total polyunsaturated fatty acid content was inversely shifted from TG to phospholipid fraction after doxorubicin and 5-fluorouracil incubation of HepG2 (+58.89% and +29.13%, P < 0.05, respectively) and SW480 (+19.20% and +14.65%, P < 0.05, respectively) cells.
Conclusion: Our data showed that common chemotherapeutic agents of HCC and CRC can induce significant changes in cellular lipid accumulation and distribution of fatty acids through producing highly saturated and unsaturated lipid droplets and membrane lipids, respectively. These metabolic side effects may be associated with gastrointestinal cancers treatment failure.
Collapse
Affiliation(s)
- Amir Mehdizadeh
- Endocrine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Morteza Bonyadi
- Liver and Gastrointestinal Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Center of Excellence for Biodiversity, Faculty of Natural Sciences, University of Tabriz, Iran
| | - Masoud Darabi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Kobra Velaei
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maghsood Shaaker
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad-Hossein Somi
- Liver and Gastrointestinal Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
26
|
Boddicker RL, Koltes JE, Fritz‐Waters ER, Koesterke L, Weeks N, Yin T, Mani V, Nettleton D, Reecy JM, Baumgard LH, Spencer JD, Gabler NK, Ross JW. Genome‐wide methylation profile following prenatal and postnatal dietary omega‐3 fatty acid supplementation in pigs. Anim Genet 2016; 47:658-671. [DOI: 10.1111/age.12468] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/06/2016] [Indexed: 02/06/2023]
Affiliation(s)
- R. L. Boddicker
- Department of Animal Science Iowa State University Ames IA 50011 USA
| | - J. E. Koltes
- Department of Animal Science Iowa State University Ames IA 50011 USA
| | | | - L. Koesterke
- Texas Advanced Computing Center University of Texas Austin TX 78758‐4497 USA
| | - N. Weeks
- Department of Mathematics Iowa State University Ames IA 50011 USA
| | - T. Yin
- Department of Statistics Iowa State University Ames IA 50011 USA
| | - V. Mani
- Department of Animal Science Iowa State University Ames IA 50011 USA
| | - D. Nettleton
- Department of Statistics Iowa State University Ames IA 50011 USA
| | - J. M. Reecy
- Department of Animal Science Iowa State University Ames IA 50011 USA
| | - L. H. Baumgard
- Department of Animal Science Iowa State University Ames IA 50011 USA
| | | | - N. K. Gabler
- Department of Animal Science Iowa State University Ames IA 50011 USA
| | - J. W. Ross
- Department of Animal Science Iowa State University Ames IA 50011 USA
| |
Collapse
|
27
|
Harris M, Kinnun JJ, Kosaraju R, Leng X, Wassall SR, Shaikh SR. Membrane Disordering by Eicosapentaenoic Acid in B Lymphomas Is Reduced by Elongation to Docosapentaenoic Acid as Revealed with Solid-State Nuclear Magnetic Resonance Spectroscopy of Model Membranes. J Nutr 2016; 146:1283-9. [PMID: 27306897 PMCID: PMC4926856 DOI: 10.3945/jn.116.231639] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 04/29/2016] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Plasma membrane organization is a mechanistic target of n-3 (ω-3) polyunsaturated fatty acids. Previous studies show that eicosapentaenoic acid (EPA; 20:5n-3) and docosahexaenoic acid (DHA; 22:6n-3) differentially disrupt plasma membrane molecular order to enhance the frequency and function of B lymphocytes. However, it is not known whether EPA and DHA affect the plasma membrane organization of B lymphomas differently to influence their function. OBJECTIVE We tested whether EPA and DHA had different effects on membrane order in B lymphomas and liposomes and studied their effects on B-lymphoma growth. METHODS B lymphomas were treated with 25 μmol EPA, DHA, or serum albumin control/L for 24 h. Membrane order was measured with fluorescence polarization, and cellular fatty acids (FAs) were analyzed with GC. Growth was quantified with a viability assay. (2)H nuclear magnetic resonance (NMR) studies were conducted on deuterated phospholipid bilayers. RESULTS Treating Raji, Ramos, and RPMI lymphomas for 24 h with 25 μmol EPA or DHA/L lowered plasma membrane order by 10-40% relative to the control. There were no differences between EPA and DHA on membrane order for the 3 cell lines. FA analyses revealed complex changes in response to EPA or DHA treatment and a large fraction of EPA was converted to docosapentaenoic acid (DPA; 22:5n-3). NMR studies, which were used to understand why EPA and DHA had similiar membrane effects, showed that phospholipids containing DPA, similar to DHA, were more ordered than those containing EPA. Finally, treating B lymphomas with 25 μmol EPA or DHA/L did not increase the frequency of B lymphomas compared with controls. CONCLUSIONS The results establish that 25 μmol EPA and DHA/L equally disrupt membrane order and do not promote B lymphoma growth. The data open a new area of investigation, which is how EPA's conversion to DPA substantially moderates its influence on membrane properties.
Collapse
Affiliation(s)
- Mitchell Harris
- Department of Biochemistry and Molecular Biology,,East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University, Greenville, NC; and
| | - Jacob J Kinnun
- Department of Physics, Indiana University–Purdue University Indianapolis, Indianapolis, IN
| | - Rasagna Kosaraju
- Department of Biochemistry and Molecular Biology,,East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University, Greenville, NC; and
| | - Xiaoling Leng
- Department of Physics, Indiana University–Purdue University Indianapolis, Indianapolis, IN
| | - Stephen R Wassall
- Department of Physics, Indiana University–Purdue University Indianapolis, Indianapolis, IN
| | - Saame Raza Shaikh
- Department of Biochemistry and Molecular Biology, East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University, Greenville, NC; and
| |
Collapse
|
28
|
Jia HJ, Zhang PJ, Liu YL, Jiang CG, Zhu X, Tian YP. Relationship of serum polyunsaturated fatty acids with cytokines in colorectal cancer. World J Gastroenterol 2016; 22:2524-2532. [PMID: 26937140 PMCID: PMC4768198 DOI: 10.3748/wjg.v22.i8.2524] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 02/03/2016] [Accepted: 02/20/2016] [Indexed: 02/06/2023] Open
Abstract
AIM To investigate the relationship of serum levels of polyunsaturated fatty acid (PUFA) with kinds of cytokines in colorectal cancer (CRC). METHODS Serum samples of 100 CRC patients were collected. The concentration of total n-3 PUFA which included C18:3 n-3, C20:5 n-3, C22:5 n-3, C22:6 n-3 and the total n-6 PUFA included C18:2 n-6, C18:3 n-6, C20:3 n-6, C20:4 n-6, and C22:5 n-6 were detected on GC-2010 Plus Gas Chromatograph with a OmegawaxTM 250 column. Cytokines were detected by MagPlexTM-C microspheres. P values for the trend were estimated by creating a continuous variable using the median value within quartiles. RESULTS Interleukin-6 (IL-6) showed significantly positive association with the C20:4 n-6 (P for trend = 0.004). Interferon gamma (IFN-γ) showed significant positive association with the C22:5 n-3 (P for trend = 0.035). IL-8 and matrix metalloproteinase-9 (MMP-9) showed significant inverse association with the C22:6 n-3 (P for trend = 0.049, and 0.021). MMP-2 showed significant inverse association with the C20:5 n-3 (P for trend = 0.008). MMP-7 showed significantly positive association with the ratio of n-6 PUFA and n-3 PUFA (P for trend = 0.008). MMP-7 also showed significantly inverse association with the ratio of C20:4 n-6 and (n-6 PUFA + n-3 PUFA) (P for trend = 0.024). IL-10 (P for trend = 0.023) and IL-6 (P for trend = 0.036) showed significantly positive association with the ratio of C20:4 n-6 and C20:5 n-3. CONCLUSION Our data suggested that serum levels of PUFA is related to the inflammation of CRC, and also play different role in regulation of immune response.
Collapse
|
29
|
Teague H, Harris M, Whelan J, Comstock SS, Fenton JI, Shaikh SR. Short-term consumption of n-3 PUFAs increases murine IL-5 levels, but IL-5 is not the mechanistic link between n-3 fatty acids and changes in B-cell populations. J Nutr Biochem 2015; 28:30-6. [PMID: 26878780 DOI: 10.1016/j.jnutbio.2015.09.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Revised: 09/02/2015] [Accepted: 09/09/2015] [Indexed: 12/31/2022]
Abstract
N-3 polyunsaturated fatty acids (PUFAs) exert immunomodulatory effects on B cells. We previously demonstrated that n-3 PUFAs enhanced the relative percentage and/or frequency of select B2 cell subsets. The objectives here were to determine if n-3 PUFAs (a) could boost cytokines that target B-cell frequency, (b) enhance the frequency of the B1 population and (c) to identify the mechanism by which n-3 PUFAs modify the proportion of B cells. Administration of n-3 PUFAs as fish oil to C57BL/6 mice enhanced secretion of the Th2 cytokine IL-5 but not IL-9 or IL-13. N-3 PUFAs had no influence on the percentage or frequency of peritoneal B1 or B2 cells. Subsequent experiments with IL-5(-/-) knockout mice showed n-3 PUFAs decreased the percentage of bone marrow B220(lo)IgM(hi) cells and increased the proportion and number of splenic IgM(+)IgD(lo)CD21(lo) cells compared to the control. These results, when compared with our previous findings with wild-type mice, suggested IL-5 had no role in mediating the effect of n-3 PUFAs on B-cell populations. To confirm this conclusion, we assayed IL-5 secretion in a diet-induced obesity model in which n-3 PUFAs enhanced the frequency of select B-cell subsets. N-3 PUFA supplementation as ethyl esters to obesogenic diets did not alter circulating IL-5 levels. Altogether, the data establish that n-3 PUFAs as fish oil can increase circulating IL-5 in lean mice, which has implications for several disease end points, but this increase in IL-5 is not the mechanistic link between n-3 PUFAs and changes in B-cell populations.
Collapse
Affiliation(s)
- Heather Teague
- Department of Biochemistry & Molecular Biology, East Carolina University; East Carolina Diabetes & Obesity Institute, East Carolina University
| | - Mitchel Harris
- Department of Biochemistry & Molecular Biology, East Carolina University; East Carolina Diabetes & Obesity Institute, East Carolina University
| | - Jarrett Whelan
- Department of Biochemistry & Molecular Biology, East Carolina University; East Carolina Diabetes & Obesity Institute, East Carolina University
| | - Sarah S Comstock
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI
| | - Jenifer I Fenton
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI; College of Osteopathic Medicine, Michigan State University, East Lansing, MI
| | - Saame Raza Shaikh
- Department of Biochemistry & Molecular Biology, East Carolina University; East Carolina Diabetes & Obesity Institute, East Carolina University; Department of Microbiology & Immunology, East Carolina University.
| |
Collapse
|
30
|
Shaikh SR, Boyle S, Edidin M. A high fat diet containing saturated but not unsaturated fatty acids enhances T cell receptor clustering on the nanoscale. Prostaglandins Leukot Essent Fatty Acids 2015; 100:1-4. [PMID: 26143085 PMCID: PMC4554807 DOI: 10.1016/j.plefa.2015.05.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 05/07/2015] [Indexed: 01/16/2023]
Abstract
Cell culture studies show that the nanoscale lateral organization of surface receptors, their clustering or dispersion, can be altered by changing the lipid composition of the membrane bilayer. However, little is known about similar changes in vivo, which can be effected by changing dietary lipids. We describe the use of a newly developed method, k-space image correlation spectroscopy, kICS, for analysis of quantum dot fluorescence to show that a high fat diet can alter the nanometer-scale clustering of the murine T cell receptor, TCR, on the surface of naive CD4(+) T cells. We found that diets enriched primarily in saturated fatty acids increased TCR nanoscale clustering to a level usually seen only on activated cells. Diets enriched in monounsaturated or n-3 polyunsaturated fatty acids had no effect on TCR clustering. Also none of the high fat diets affected TCR clustering on the micrometer scale. Furthermore, the effect of the diets was similar in young and middle aged mice. Our data establish proof-of-principle that TCR nanoscale clustering is sensitive to the composition of dietary fat.
Collapse
MESH Headings
- Animals
- CD4-Positive T-Lymphocytes/metabolism
- Diet, High-Fat
- Fatty Acids/metabolism
- Fatty Acids, Monounsaturated/metabolism
- Fatty Acids, Omega-3/metabolism
- Mice
- Mice, Transgenic
- Protein Multimerization
- Receptors, Antigen, T-Cell/chemistry
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/metabolism
- Spectrum Analysis/methods
Collapse
Affiliation(s)
- Saame Raza Shaikh
- Department of Biochemistry & Molecular Biology, East Carolina Diabetes & Obesity Institute, East Carolina University, 600 Moye Blvd, Greenville, NC 27834, USA.
| | - Sarah Boyle
- Department of Biology, The Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA
| | - Michael Edidin
- Department of Biology, The Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA
| |
Collapse
|
31
|
Duriancik DM, Comstock SS, Langohr IM, Fenton JI. High levels of fish oil enhance neutrophil development and activation and influence colon mucus barrier function in a genetically susceptible mouse model. J Nutr Biochem 2015; 26:1261-72. [PMID: 26297475 DOI: 10.1016/j.jnutbio.2015.06.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Revised: 05/28/2015] [Accepted: 06/04/2015] [Indexed: 12/23/2022]
Abstract
Dietary fatty acids influence immunologic homeostasis, but their effect on initiation of colitis, an immune-mediated disease, is not well established. Previously, our laboratory demonstrated that high doses of dietary fish oil (FO) increased colon inflammation and dysplasia in a model of infection-induced colitis. In the current study, we assessed the effects of high-dose dietary FO, 6% by weight, on colon inflammation, neutrophil recruitment and function, and mucus layer integrity in a genetically susceptible, colitis-prone mouse model in the absence of infection. FO-fed SMAD3(-/-) mice had increased colon inflammation evidenced by increased numbers of systemic and local neutrophils and increased neutrophil chemoattractant and inflammatory cytokine gene expression in the colon. Mucus layer thickness in the cecum and goblet cell numbers in the cecum and colon in FO-fed mice were reduced compared to control. FO consumption affected colitis in male and female mice differently. Compared to female control mice, neutrophils from FO-fed female mice had reduced reactive oxygen species (ROS) upon ex vivo stimulation with phorbol myristate acetate while FO-fed male mice produced increased ROS compared to control-fed male mice. In summary, dietary FO impaired mucus layer integrity and was associated with colon inflammation characterized by increased neutrophil numbers and altered neutrophil function. High-dose FO may have detrimental effects in populations genetically susceptible for inflammatory bowel disease and these effects may differ between males and females.
Collapse
Affiliation(s)
- David M Duriancik
- Department of Food Science & Human Nutrition, Michigan State University East Lansing, MI, 48824
| | - Sarah S Comstock
- Department of Food Science & Human Nutrition, Michigan State University East Lansing, MI, 48824
| | - Ingeborg M Langohr
- Department of Pathobiological Sciences Louisiana State University, Baton Rouge, LA, 70803
| | - Jenifer I Fenton
- Department of Food Science & Human Nutrition, Michigan State University East Lansing, MI, 48824.
| |
Collapse
|
32
|
Whelan J, Gowdy KM, Shaikh SR. N-3 polyunsaturated fatty acids modulate B cell activity in pre-clinical models: Implications for the immune response to infections. Eur J Pharmacol 2015; 785:10-17. [PMID: 26022530 DOI: 10.1016/j.ejphar.2015.03.100] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Revised: 01/15/2015] [Accepted: 03/05/2015] [Indexed: 12/12/2022]
Abstract
B cell antigen presentation, cytokine production, and antibody production are targets of pharmacological intervention in inflammatory and infectious diseases. Here we review recent pre-clinical evidence demonstrating that pharmacologically relevant levels of n-3 polyunsaturated fatty acids (PUFA) derived from marine fish oils influence key aspects of B cell function through multiple mechanisms. N-3 PUFAs modestly diminish B cell mediated stimulation of classically defined naïve CD4(+) Th1 cells through the major histocompatibility complex (MHC) class II pathway. This is consistent with existing data showing that n-3 PUFAs suppress the activation of Th1/Th17 cells through direct effects on helper T cells and indirect effects on antigen presenting cells. Mechanistically, n-3 PUFAs lower antigen presentation and T cell signaling by disrupting the formation of lipid microdomains within the immunological synapse. We then review data to show that n-3 PUFAs boost B cell activation and antibody production in the absence and presence of antigen stimulation. This has potential benefits for several clinical populations such as the aged and obese that have poor humoral immunity. The mode of action by which n-3 PUFA boost B cell activation and antibody production remains unclear, but may involve Th2 cytokines, enhanced production of specialized proresolving lipid mediators, and targeting of protein lateral organization in lipid microdomains. Finally, we highlight evidence to show that different n-3 PUFAs are not biologically equivalent, which has implications for the development of future interventions to target B cell activity.
Collapse
Affiliation(s)
- Jarrett Whelan
- Department of Biochemistry & Molecular Biology, East Carolina Diabetes & Obesity Institute, East Carolina Heart Institute, Brody School of Medicine, East Carolina University, Greenville, NC 27834, United States
| | - Kymberly M Gowdy
- Department of Pharmacology & Toxicology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, United States
| | - Saame Raza Shaikh
- Department of Biochemistry & Molecular Biology, East Carolina Diabetes & Obesity Institute, East Carolina Heart Institute, Brody School of Medicine, East Carolina University, Greenville, NC 27834, United States; Department of Microbiology & Immunology, East Carolina Diabetes & Obesity Institute, East Carolina Heart Institute, Brody School of Medicine, East Carolina University, Greenville, NC 27834, United States
| |
Collapse
|
33
|
Liu L, Feng R, Guo F, Li Y, Jiao J, Sun C. Targeted metabolomic analysis reveals the association between the postprandial change in palmitic acid, branched-chain amino acids and insulin resistance in young obese subjects. Diabetes Res Clin Pract 2015; 108:84-93. [PMID: 25700627 DOI: 10.1016/j.diabres.2015.01.014] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Revised: 01/09/2015] [Accepted: 01/13/2015] [Indexed: 01/13/2023]
Abstract
Obesity is the result of a positive energy balance and often leads to difficulties in maintaining normal postprandial metabolism. The changes in postprandial metabolites after an oral glucose tolerance test (OGTT) in young obese Chinese men are unclear. In this work, the aim is to investigate the complex metabolic alterations in obesity provoked by an OGTT using targeted metabolomics. We used gas chromatography-mass spectrometry and ultra high performance liquid chromatography-triple quadrupole mass spectrometry to analyze serum fatty acids, amino acids and biogenic amines profiles from 15 control and 15 obese subjects at 0, 30, 60, 90 and 120 min during an OGTT. Metabolite profiles from 30 obese subjects as independent samples were detected in order to validate the change of metabolites. There were the decreased levels of fatty acid, amino acids and biogenic amines after OGTT in obesity. At 120 min, percent change of 20 metabolites in obesity has statistical significance when comparing with the controls. The obese parameters was positively associated with changes in arginine and histidine (P<0.05) and the postprandial change in palmitic acid (PA), branched-chain amino acids (BCAAs) and phenylalanine between 1 and 120 min were positively associated with fasting insulin and HOMA-IR (all P<0.05) in the obese group. The postprandial metabolite of PA and BCAAs may play important role in the development and onset of insulin resistance in obesity. Our findings offer new insights in the complex physiological regulation of the metabolism during an OGTT in obesity.
Collapse
Affiliation(s)
- Liyan Liu
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, PR China
| | - Rennan Feng
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, PR China
| | - Fuchuan Guo
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, PR China
| | - Ying Li
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, PR China
| | - Jundong Jiao
- The Second Affiliated Hospital of Harbin Medical University, Harbin, PR China.
| | - Changhao Sun
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, PR China.
| |
Collapse
|
34
|
Shaikh SR, Haas KM, Beck MA, Teague H. The effects of diet-induced obesity on B cell function. Clin Exp Immunol 2015; 179:90-9. [PMID: 25169121 DOI: 10.1111/cei.12444] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/20/2014] [Indexed: 12/12/2022] Open
Abstract
B-1 and B-2 B cell subsets carry out a diverse array of functions that range broadly from responding to innate stimuli, antigen presentation, cytokine secretion and antibody production. In this review, we first cover the functional roles of the major murine B cell subsets. We then highlight emerging evidence, primarily in preclinical rodent studies, to show that select B cell subsets are a therapeutic target in obesity and its associated co-morbidities. High fat diets promote accumulation of select murine B cell phenotypes in visceral adipose tissue. As a consequence, B cells exacerbate inflammation and thereby insulin sensitivity through the production of autoantibodies and via cross-talk with select adipose resident macrophages, CD4(+) and CD8(+) T cells. In contrast, interleukin (IL)-10-secreting regulatory B cells counteract the proinflammatory profile and improve glucose sensitivity. We subsequently review data from rodent studies that show pharmacological supplementation of obesogenic diets with long chain n-3 polyunsaturated fatty acids or specialized pro-resolving lipid mediators synthesized from endogenous n-3 polyunsaturated fatty acids boost B cell activation and antibody production. This may have potential benefits for improving inflammation in addition to combating the increased risk of viral infection that is an associated complication of obesity and type II diabetes. Finally, we propose potential underlying mechanisms throughout the review by which B cell activity could be differentially regulated in response to high fat diets.
Collapse
Affiliation(s)
- S R Shaikh
- Department of Biochemistry and Molecular Biology, East Carolina Diabetes and Obesity Institute, East Carolina Heart Institute, Brody School of Medicine, East Carolina University, Greenville, NC, USA; Department of Microbiology and Immunology, East Carolina Diabetes and Obesity Institute, East Carolina Heart Institute, Brody School of Medicine, East Carolina University, Greenville, NC, USA
| | | | | | | |
Collapse
|
35
|
Gurzell EA, Teague H, Duriancik D, Clinthorne J, Harris M, Shaikh SR, Fenton JI. Marine fish oils are not equivalent with respect to B-cell membrane organization and activation. J Nutr Biochem 2014; 26:369-77. [PMID: 25616447 DOI: 10.1016/j.jnutbio.2014.11.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2014] [Revised: 10/29/2014] [Accepted: 11/05/2014] [Indexed: 12/29/2022]
Abstract
We previously reported that docosahexaenoic-acid (DHA)-enriched fish oil (DFO) feeding altered B-cell membrane organization and enhanced B-cell function. The purpose of this study was to evaluate whether menhaden oil (MO) and eicosapentaenoic-acid (EPA)-enriched fish oil (EFO) alters B-cell function/phenotype similarly. Mice were fed control (CON), MO, EFO or DFO diets for 5weeks. We evaluated the fatty acid composition of B-cell phospholipids, membrane microdomain organization, ex vivo B-cell functionality and in vivo B-cell subsets. Red blood cells and B cells were found to be strongly (r>0.85) and significantly (P<.001) correlated for major n-3 and n-6 long-chain polyunsaturated fatty acids (LCPUFAs). Compared to CON, MO and DFO resulted in decreased clustering of membrane microdomains, whereas EFO increased clustering. All fish oil treatments had 1.12-1.60 times higher CD40 expression following stimulation; however, we observed 0.86 times lower major histocompatibility complex class II expression and 0.7 times lower interleukin (IL)-6 production from EFO, but 3.25 times higher interferon-γ from MO and 1.5 times higher IL-6 from DFO. By 90min of incubation, MO had 1.11 times higher antigen uptake compared to CON, whereas EFO was 0.86 times lower. All fish oil treatments resulted in decreasingly mature splenic and bone marrow B-cell subsets. We conclude that diets high in n-3 LCPUFAs may elicit similar B-cell phenotypes but different organizational and functional outcomes. More specifically, these data suggest that the EPA and DHA content of a diet influences immunological outcomes, highlighting the importance of understanding how specific n-3 LCPUFAs modulate B-cell development and function.
Collapse
Affiliation(s)
- Eric A Gurzell
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI 48824
| | - Heather Teague
- Department of Biochemistry & Molecular Biology, Department of Microbiology & Immunology, East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC 27834
| | - David Duriancik
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI 48824
| | - Jonathan Clinthorne
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI 48824
| | - Mitchel Harris
- Department of Biochemistry & Molecular Biology, Department of Microbiology & Immunology, East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC 27834
| | - Saame Raza Shaikh
- Department of Biochemistry & Molecular Biology, Department of Microbiology & Immunology, East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC 27834
| | - Jenifer I Fenton
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI 48824; College of Osteopathic Medicine, Michigan State University, East Lansing, MI 48824.
| |
Collapse
|
36
|
Agbor LN, Wiest EF, Rothe M, Schunck WH, Walker MK. Role of CYP1A1 in modulating the vascular and blood pressure benefits of omega-3 polyunsaturated fatty acids. J Pharmacol Exp Ther 2014; 351:688-98. [PMID: 25316121 PMCID: PMC4244579 DOI: 10.1124/jpet.114.219535] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Accepted: 10/10/2014] [Indexed: 01/08/2023] Open
Abstract
The mechanisms that mediate the cardiovascular protective effects of omega 3 (n-3) polyunsaturated fatty acids (PUFAs) have not been fully elucidated. Cytochrome P450 1A1 efficiently metabolizes n-3 PUFAs to potent vasodilators. Thus, we hypothesized that dietary n-3 PUFAs increase nitric oxide (NO)-dependent blood pressure regulation and vasodilation in a CYP1A1-dependent manner. CYP1A1 wild-type (WT) and knockout (KO) mice were fed an n-3 or n-6 PUFA-enriched diet for 8 weeks and were analyzed for tissue fatty acids and metabolites, NO-dependent blood pressure regulation, NO-dependent vasodilation of acetylcholine (ACh) in mesenteric resistance arterioles, and endothelial NO synthase (eNOS) and phospho-Ser1177-eNOS expression in the aorta. All mice fed the n-3 PUFA diet showed significantly higher levels of n-3 PUFAs and their metabolites, and significantly lower levels of n-6 PUFAs and their metabolites. In addition, KO mice on the n-3 PUFA diet accumulated significantly higher levels of n-3 PUFAs in the aorta and kidney without a parallel increase in the levels of their metabolites. Moreover, KO mice exhibited significantly less NO-dependent regulation of blood pressure on the n-3 PUFA diet and significantly less NO-dependent, ACh-mediated vasodilation in mesenteric arterioles on both diets. Finally, the n-3 PUFA diet significantly increased aortic phospho-Ser1177-eNOS/eNOS ratio in the WT compared with KO mice. These data demonstrate that CYP1A1 contributes to eNOS activation, NO bioavailability, and NO-dependent blood pressure regulation mediated by dietary n-3 PUFAs.
Collapse
Affiliation(s)
- Larry N Agbor
- Department of Pharmaceutical Sciences, University of New Mexico, Albuquerque, New Mexico (L.N.A., E.F.W., M.K.W.); Lipidomix GmbH, Berlin, Germany (M.R.); and Max-Delbrück Center for Molecular Medicine, Berlin, Germany (W.-H.S.)
| | - Elani F Wiest
- Department of Pharmaceutical Sciences, University of New Mexico, Albuquerque, New Mexico (L.N.A., E.F.W., M.K.W.); Lipidomix GmbH, Berlin, Germany (M.R.); and Max-Delbrück Center for Molecular Medicine, Berlin, Germany (W.-H.S.)
| | - Michael Rothe
- Department of Pharmaceutical Sciences, University of New Mexico, Albuquerque, New Mexico (L.N.A., E.F.W., M.K.W.); Lipidomix GmbH, Berlin, Germany (M.R.); and Max-Delbrück Center for Molecular Medicine, Berlin, Germany (W.-H.S.)
| | - Wolf-Hagen Schunck
- Department of Pharmaceutical Sciences, University of New Mexico, Albuquerque, New Mexico (L.N.A., E.F.W., M.K.W.); Lipidomix GmbH, Berlin, Germany (M.R.); and Max-Delbrück Center for Molecular Medicine, Berlin, Germany (W.-H.S.)
| | - Mary K Walker
- Department of Pharmaceutical Sciences, University of New Mexico, Albuquerque, New Mexico (L.N.A., E.F.W., M.K.W.); Lipidomix GmbH, Berlin, Germany (M.R.); and Max-Delbrück Center for Molecular Medicine, Berlin, Germany (W.-H.S.)
| |
Collapse
|
37
|
Teague H, Harris M, Fenton J, Lallemand P, Shewchuk BM, Shaikh SR. Eicosapentaenoic and docosahexaenoic acid ethyl esters differentially enhance B-cell activity in murine obesity. J Lipid Res 2014; 55:1420-33. [PMID: 24837990 DOI: 10.1194/jlr.m049809] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Indexed: 01/06/2023] Open
Abstract
EPA and DHA are not biologically equivalent; however, their individual activity on B cells is unknown. We previously reported fish oil enhanced murine B-cell activity in obesity. To distinguish between the effects of EPA and DHA, we studied the ethyl esters of EPA and DHA on murine B-cell function as a function of time. We first demonstrate that EPA and DHA maintained the obese phenotype, with no improvements in fat mass, adipose inflammatory cytokines, fasting insulin, or glucose clearance. We then tested the hypothesis that EPA and DHA would increase the frequency of splenic B cells. EPA and DHA differentially enhanced the frequency and/or percentage of select B-cell subsets, correlating with increased natural serum IgM and cecal IgA. We next determined the activities of EPA and DHA on ex vivo production of cytokines upon lipopolysaccharide stimulation of B cells. EPA and DHA, in a time-dependent manner, enhanced B-cell cytokines with DHA notably increasing IL-10. At the molecular level, EPA and DHA differentially enhanced the formation of ordered microdomains but had no effect on Toll-like receptor 4 mobility. Overall, the results establish differential effects of EPA and DHA in a time-dependent manner on B-cell activity in obesity, which has implications for future clinical studies.
Collapse
Affiliation(s)
- Heather Teague
- Department of Biochemistry and Molecular Biology, Brody School of Medicine, East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC
| | - Mitchel Harris
- Department of Biochemistry and Molecular Biology, Brody School of Medicine, East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC
| | - Jenifer Fenton
- Department of Food Science and Nutrition, Michigan State University, East Lansing, MI
| | - Perrine Lallemand
- Department of Biochemistry and Molecular Biology, Brody School of Medicine, East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC
| | - Brian M Shewchuk
- Department of Biochemistry and Molecular Biology, Brody School of Medicine, East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC
| | - Saame Raza Shaikh
- Department of Biochemistry and Molecular Biology, Brody School of Medicine, East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC
| |
Collapse
|
38
|
Teng KT, Chang CY, Chang LF, Nesaretnam K. Modulation of obesity-induced inflammation by dietary fats: mechanisms and clinical evidence. Nutr J 2014; 13:12. [PMID: 24476102 PMCID: PMC3922162 DOI: 10.1186/1475-2891-13-12] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Accepted: 01/13/2014] [Indexed: 01/16/2023] Open
Abstract
Obesity plays a pivotal role in the development of low-grade inflammation. Dietary fatty acids are important modulators of inflammatory responses. Saturated fatty acids (SFA) and n-6 polyunsaturated fatty acids (PUFA) have been reported to exert pro-inflammatory effects. n-3 PUFA in particular, possess anti-inflammatory properties. Numerous clinical studies have been conducted over decades to investigate the impact of dietary fatty acids on inflammatory response in obese individuals, however the findings remained uncertain. High fat meals have been reported to increase pro-inflammatory responses, however there is limited evidence to support the role of individual dietary fatty acids in a postprandial state. Evidence in chronic studies is contradictory, the effects of individual dietary fatty acids deserves further attention. Weight loss rather than n-3 PUFA supplementation may play a more prominent role in alleviating low grade inflammation. In this context, the present review provides an update on the mechanistic insight and the influence of dietary fats on low grade inflammation, based on clinical evidence from acute and chronic clinical studies in obese and overweight individuals.
Collapse
Affiliation(s)
- Kim-Tiu Teng
- Product Development and Advisory Services, Malaysian Palm Oil Board (MPOB), 6 Persiaran Institusi, Bandar Baru Bangi, 43000 Kajang, Selangor, Malaysia.
| | | | | | | |
Collapse
|
39
|
Fenton JI, Hord NG, Ghosh S, Gurzell EA. Immunomodulation by dietary long chain omega-3 fatty acids and the potential for adverse health outcomes. Prostaglandins Leukot Essent Fatty Acids 2013; 89:379-90. [PMID: 24183073 PMCID: PMC3912985 DOI: 10.1016/j.plefa.2013.09.011] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Revised: 09/18/2013] [Accepted: 09/21/2013] [Indexed: 01/07/2023]
Abstract
Recommendations to consume fish for prevention of cardiovascular disease (CVD), along with the U.S. Food and Drug Administration-approved generally recognized as safe (GRAS) status for long chain omega-3 fatty acids, may have had the unanticipated consequence of encouraging long-chain omega-3 (ω-3) fatty acid [(eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA)] supplementation and fortification practices. While there is evidence supporting a protective role for EPA/DHA supplementation in reducing sudden cardiac events, the safety and efficacy of supplementation with LCω-3PUFA in the context of other disease outcomes is unclear. Recent studies of bacterial, viral, and fungal infections in animal models of infectious disease demonstrate that LCω-3PUFA intake dampens immunity and alters pathogen clearance and can result in reduced survival. The same physiological properties of EPA/DHA that are responsible for the amelioration of inflammation associated with chronic cardiovascular pathology or autoimmune states, may impair pathogen clearance during acute infections by decreasing host resistance or interfere with tumor surveillance resulting in adverse health outcomes. Recent observations that high serum LCω-3PUFA levels are associated with higher risk of prostate cancer and atrial fibrillation raise concern for adverse outcomes. Given the widespread use of supplements and fortification of common food items with LCω-3PUFA, this review focuses on the immunomodulatory effects of the dietary LCω-3PUFAs, EPA and DHA, the mechanistic basis for potential negative health outcomes, and calls for biomarker development and validation as rational first steps towards setting recommended dietary intake levels.
Collapse
Affiliation(s)
- Jenifer I Fenton
- Department of Food Science and Human, College of Agriculture and Natural Resources, Michigan State University, East Lansing, MI, United States; College of Osteopathic Medicine, Michigan State University, East Lansing, MI 48824, United States.
| | | | | | | |
Collapse
|
40
|
Tomasdottir V, Thorleifsdottir S, Vikingsson A, Hardardottir I, Freysdottir J. Dietary omega-3 fatty acids enhance the B1 but not the B2 cell immune response in mice with antigen-induced peritonitis. J Nutr Biochem 2013; 25:111-7. [PMID: 24332949 DOI: 10.1016/j.jnutbio.2013.09.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Revised: 09/06/2013] [Accepted: 09/10/2013] [Indexed: 12/14/2022]
Abstract
The effects of omega-3 fatty acids on the adaptive immune response have mainly been analysed in vitro with varying results. How omega-3 fatty acids affect the adaptive immune response in vivo is largely unknown. This study examined the effects of dietary fish oil on the adaptive immune response in antigen-induced inflammation in mice, focusing on its effects on B cells and B cell subsets. Mice were fed a control diet with or without 2.8% fish oil, immunized twice with methylated BSA (mBSA) and peritonitis induced by intraperitoneal injection of mBSA. Serum, spleen and peritoneal exudate were collected prior to and at different time points after induction of peritonitis. Serum levels of mBSA-specific antibodies were determined by ELISA and the number of peritoneal and splenic lymphocytes by flow cytometry. The levels of germinal center B cells and IgM(+), IgG(+) and CD138(+) cells in spleen were evaluated by immunoenzyme staining. Mice fed the fish oil diet had more peritoneal B1 cells, more IgM(+) cells in spleen and higher levels of serum mBSA-specific IgM antibodies compared with that in mice fed the control diet. However, dietary fish oil did not affect the number of peritoneal B2 cells, splenic IgG(+) or CD138(+) cells or serum levels of mBSA-specific IgG antibodies in mice with mBSA-induced peritonitis. These results indicate that dietary fish oil can enhance the adaptive immune response, specifically the B1 cell response, which may lead to better protection against secondary infection as well as improvement in reaching homeostasis following antigenic challenge.
Collapse
Affiliation(s)
- Valgerdur Tomasdottir
- Center of Rheumatology Research, Landspitali - The University Hospital of Iceland, Reykjavik, Iceland; Department of Immunology, Landspitali - The University Hospital of Iceland, Reykjavik, Iceland; Department of Biochemistry and Molecular Biology, Faculty of Medicine, Biomedical Center, University of Iceland, 101 Reykjavik, Iceland
| | - Sigrun Thorleifsdottir
- Center of Rheumatology Research, Landspitali - The University Hospital of Iceland, Reykjavik, Iceland; Department of Immunology, Landspitali - The University Hospital of Iceland, Reykjavik, Iceland; Department of Biochemistry and Molecular Biology, Faculty of Medicine, Biomedical Center, University of Iceland, 101 Reykjavik, Iceland
| | - Arnor Vikingsson
- Center of Rheumatology Research, Landspitali - The University Hospital of Iceland, Reykjavik, Iceland
| | - Ingibjorg Hardardottir
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Biomedical Center, University of Iceland, 101 Reykjavik, Iceland
| | - Jona Freysdottir
- Center of Rheumatology Research, Landspitali - The University Hospital of Iceland, Reykjavik, Iceland; Department of Immunology, Landspitali - The University Hospital of Iceland, Reykjavik, Iceland; Department of Immunology, Faculty of Medicine, Biomedical Center, University of Iceland, 101 Reykjavik, Iceland.
| |
Collapse
|
41
|
Teague H, Fhaner CJ, Harris M, Duriancik DM, Reid GE, Shaikh SR. n-3 PUFAs enhance the frequency of murine B-cell subsets and restore the impairment of antibody production to a T-independent antigen in obesity. J Lipid Res 2013; 54:3130-8. [PMID: 23986558 DOI: 10.1194/jlr.m042457] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The role of n-3 polyunsaturated fatty acids (PUFA) on in vivo B-cell immunity is unknown. We first investigated how n-3 PUFAs impacted in vivo B-cell phenotypes and antibody production in the absence and presence of antigen compared with a control diet. Lean mice consuming n-3 PUFAs for 4 weeks displayed increased percentage and frequency of splenic transitional 1 B cells. Upon stimulation with trinitrophenylated-lipopolysaccharide, n-3 PUFAs increased the number of splenic transitional 1/2, follicular, premarginal, and marginal zone B cells. n-3 PUFAs also increased surface, but not circulating, IgM. We next tested the effects of n-3 PUFAs in a model of obesity that is associated with suppressed humoral immunity. An obesogenic diet after ten weeks of feeding, relative to a lean control, had no effect on the frequency of B cells but lowered circulating IgM upon antigen stimulation. Administration of n-3 PUFAs to lean and obese mice increased the percentage and/or frequency of transitional 1 and marginal zone B cells. Furthermore, n-3 PUFAs in lean and obese mice increased circulating IgM relative to controls. Altogether, the data show n-3 PUFAs enhance B cell-mediated immunity in vivo, which has implications for immunocompromised populations, such as the obese.
Collapse
Affiliation(s)
- Heather Teague
- Department of Biochemistry and Molecular Biology, Brody School of Medicine, East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC
| | | | | | | | | | | |
Collapse
|
42
|
Teague H, Rockett BD, Harris M, Brown DA, Shaikh SR. Dendritic cell activation, phagocytosis and CD69 expression on cognate T cells are suppressed by n-3 long-chain polyunsaturated fatty acids. Immunology 2013; 139:386-94. [PMID: 23373457 DOI: 10.1111/imm.12088] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Revised: 01/09/2013] [Accepted: 01/30/2013] [Indexed: 12/14/2022] Open
Abstract
Docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA) are bioactive n-3 long-chain polyunsaturated fatty acids (LCPUFAs) in fish oil that exert immunosuppressive effects. A significant amount of literature shows that n-3 LCPUFAs suppress dendritic cell (DC) function in vitro; however, few studies have determined if the effects are emulated at the animal level. In this study, we first focused on the functional consequences of 5% (weight/weight) fish oil on splenic CD11c(+) DCs. Administration of n-3 LCPUFAs, modelling human pharmacological intake (2% of total kcal from EPA,1·3% from DHA), to C57BL/6 mice for 3 weeks reduced DC surface expression of CD80 by 14% and tumour necrosis factor-α secretion by 29% upon lipopolysaccharide stimulation relative to a control diet. The n-3 LCPUFAs also significantly decreased CD11c(+) surface expression and phagocytosis by 12% compared with the control diet. Antigen presentation studies revealed a 22% decrease in CD69 surface expression on transgenic CD4(+) T lymphocytes activated by DCs from mice fed fish oil. We then determined if the functional changes were mechanistically associated with changes in lipid microdomain clustering or plasma membrane microviscosity with n-3 LCPUFAs, as reported for B and T lymphocytes. Fish oil administration to mice did not influence cholera-toxin induced lipid microdomain clustering or microviscosity, even though EPA and DHA levels were significantly elevated relative to the control diet. Overall, our data show that n-3 LCPUFAs exert immunosuppressive effects on DCs, validating in vitro studies. The results also show that DC microdomain clustering and microviscosity were not changed by the n-3 LCPUFA intervention used in this study.
Collapse
Affiliation(s)
- Heather Teague
- Department of Biochemistry and Molecular Biology, East Carolina University, Greenville, NC, USA
| | | | | | | | | |
Collapse
|
43
|
Abstract
Numerous effects of n-3 fatty acids EPA and DHA on functional responses of cells involved in inflammation and immunity have been described. Fatty acid-induced modifications in membrane order and in the availability of substrates for eicosanoid synthesis are long-standing mechanisms that are considered important in explaining the effects observed. More recently, effects on signal transduction pathways and on gene expression profiles have been identified. Over the last 10 years or so, significant advances in understanding the mechanisms of action of n-3 fatty acids have been made. These include the identification of new actions of lipid mediators that were already described and of novel interactions among those mediators and the description of an entirely new family of lipid mediators, resolvins and protectins that have anti-inflammatory actions and are critical to the resolution of inflammation. It is also recognised that EPA and DHA can inhibit activation of the prototypical inflammatory transcription factor NF-κB. Recent studies suggest three alternative mechanisms by which n-3 fatty acids might have this effect. Within T-cells, as well as other cells of relevance to immune and inflammatory responses, EPA and DHA act to disrupt very early events involving formation of the structures termed lipid rafts which bring together various proteins to form an effective signalling platform. In summary, recent research has identified a number of new mechanisms of action that help to explain previously identified effects of n-3 fatty acids on inflammation and immunity.
Collapse
|
44
|
|
45
|
Fenton JI, McCaskey SJ. Curcumin and docosahexaenoic acid block insulin-induced colon carcinoma cell proliferation. Prostaglandins Leukot Essent Fatty Acids 2013; 88:219-26. [PMID: 23266210 DOI: 10.1016/j.plefa.2012.11.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2012] [Revised: 11/27/2012] [Accepted: 11/29/2012] [Indexed: 12/19/2022]
Abstract
Diets high in fish and curcumin are associated with a decreased risk of CRC. Insulin resistance and obesity are associated with increased CRC risk and higher reoccurrence rates. We utilized cell culture to determine if dietary compounds could reduce insulin-induced cell proliferation comparing the response in normal and metastatic colon epithelial cells. We treated model normal murine colon epithelial cells (YAMC) and adenocarcinoma cells (MC38) with docosahexaenoic acid (DHA) or curcumin alone and then co-treatments of the diet-derived compound and insulin were combined. Cell proliferation was stimulated with insulin (1 ug/mL) to model insulin resistance in obesity. Despite the presence of insulin, proliferation was reduced in the MC38 cells treated with 10 μM curcumin (p<0.001) and 50 μM DHA (p<0.001). Insulin stimulated MAPK and MEK phosphorylation was inhibited by DHA and curcumin in MC38 cancer cells. Here we show that curcumin and DHA can block insulin-induced colon cancer cell proliferation in vitro via a MEK mediated mechanism.
Collapse
Affiliation(s)
- Jenifer I Fenton
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, USA.
| | | |
Collapse
|
46
|
Chu X, Liu L, Na L, Lu H, Li S, Li Y, Sun C. Sterol regulatory element-binding protein-1c mediates increase of postprandial stearic acid, a potential target for improving insulin resistance, in hyperlipidemia. Diabetes 2013; 62:561-571. [PMID: 22966071 PMCID: PMC3554356 DOI: 10.2337/db12-0139] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2012] [Accepted: 07/18/2012] [Indexed: 12/25/2022]
Abstract
Elevated serum free fatty acids (FFAs) levels play an important role in the development of insulin resistance (IR) and diabetes. We investigated the dynamic changes and the underlying regulatory mechanism of postprandial FFA profile in hyperlipidemia (HLP) and their relation with insulin sensitivity in both humans and mice. We found that serum stearic acid (SA) is the only fatty acid that is increased dramatically in the postprandial state. The elevation of SA is due to increased insulin-stimulated de novo synthesis mediated by sterol regulatory element-binding protein-1c (SREBP-1c)/acetyl-CoA carboxylase/fatty acid synthase/elongation of long-chain fatty acid family member 6 (ELOVL6) and the elongation of palmitic acid (PA) catalyzed by ELOVL6. Downregulation of SREBP-1c or ELOVL6 by small interfering RNA can reduce SA synthesis in liver and serum SA level, followed by amelioration of IR in HLP mice. However, inhibition of SREBP-1c is more effective in improving IR than suppression of ELOVL6, which resulted in accumulation of PA. In summary, increased postprandial SA is caused by the insulin-stimulated SREBP-1c pathway and elongation of PA in HLP. Reduction of postprandial SA is a good candidate for improving IR, and SREBP-1c is potentially a better target to prevent IR and diabetes by decreasing SA.
Collapse
Affiliation(s)
- Xia Chu
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, Hei Longjiang Province, People’s Republic of China
| | - Liyan Liu
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, Hei Longjiang Province, People’s Republic of China
| | - Lixin Na
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, Hei Longjiang Province, People’s Republic of China
| | - Huimin Lu
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, Hei Longjiang Province, People’s Republic of China
| | - Songtao Li
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, Hei Longjiang Province, People’s Republic of China
| | - Ying Li
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, Hei Longjiang Province, People’s Republic of China
| | - Changhao Sun
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, Hei Longjiang Province, People’s Republic of China
| |
Collapse
|
47
|
Shaikh SR, Teague H. N-3 fatty acids and membrane microdomains: from model membranes to lymphocyte function. Prostaglandins Leukot Essent Fatty Acids 2012; 87:205-8. [PMID: 23107229 PMCID: PMC3573853 DOI: 10.1016/j.plefa.2012.09.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Revised: 09/24/2012] [Accepted: 09/26/2012] [Indexed: 10/27/2022]
Abstract
This article summarizes the author's research on fish oil derived n-3 fatty acids, plasma membrane organization and B cell function. We first cover basic model membrane studies that investigated how docosahexaenoic acid (DHA) targeted the organization of sphingolipid-cholesterol enriched lipid microdomains. A key finding here was that DHA had a relatively poor affinity for cholesterol. This work led to a model that predicted DHA acyl chains in cells would manipulate lipid-protein microdomain organization and thereby function. We then review how the predictions of the model were tested with B cells in vitro followed by experiments using mice fed fish oil. These studies reveal a highly complex picture on how n-3 fatty acids target lipid-protein organization and B cell function. Key findings are as follows: (1) n-3 fatty acids target not just the plasma membrane but also endomembrane organization; (2) DHA, but not eicosapentaenoic acid (EPA), disrupts microdomain spatial distribution (i.e. clustering), (3) DHA alters protein lateral organization and (4) changes in membrane organization are accompanied by functional effects on both innate and adaptive B cell function. Altogether, the research over the past 10 years has led to an evolution of the original model on how DHA reorganizes membrane microdomains. The work raises the intriguing possibility of testing the model at the human level to target health and disease.
Collapse
Affiliation(s)
- Saame Raza Shaikh
- Department of Biochemistry and Molecular Biology, East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University 600, Moye Blvd, Greenville, NC 27834, USA.
| | | |
Collapse
|
48
|
Gurzell EA, Teague H, Harris M, Clinthorne J, Shaikh SR, Fenton JI. DHA-enriched fish oil targets B cell lipid microdomains and enhances ex vivo and in vivo B cell function. J Leukoc Biol 2012. [PMID: 23180828 DOI: 10.1189/jlb.0812394] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
DHA is a n-3 LCPUFA in fish oil that generally suppresses T lymphocyte function. However, the effect of fish oil on B cell function remains relatively understudied. Given the important role of B cells in gut immunity and increasing human fish oil supplementation, we sought to determine whether DFO leads to enhanced B cell activation in the SMAD-/- colitis-prone mouse model, similar to that observed with C57BL/6 mice. This study tested the hypothesis that DHA from fish oil is incorporated into the B cell membrane to alter lipid microdomain clustering and enhance B cell function. Purified, splenic B cells from DFO-fed mice displayed increased DHA levels and diminished GM1 microdomain clustering. DFO enhanced LPS-induced B cell secretion of IL-6 and TNF-α and increased CD40 expression ex vivo compared with CON. Despite increased MHCII expression in the unstimulated ex vivo B cells from DFO-fed mice, we observed no difference in ex vivo OVA-FITC uptake in B cells from DFO or CON mice. In vivo, DFO increased lymphoid tissue B cell populations and surface markers of activation compared with CON. Finally, we investigated whether these ex vivo and in vivo observations were consistent with systemic changes. Indeed, DFO-fed mice had significantly higher plasma IL-5, IL-13, and IL-9 (Th2-biasing cytokines) and cecal IgA compared with CON. These results support the hypothesis and an emerging concept that fish oil enhances B cell function in vivo.
Collapse
Affiliation(s)
- Eric A Gurzell
- Michigan State University, East Lansing, MI 48824-1224, USA.
| | | | | | | | | | | |
Collapse
|
49
|
Whelan JT, Chen J, Miller J, Morrow RL, Lingo JD, Merrell K, Shaikh SR, Bridges LC. 9-cis-retinoic acid promotes cell adhesion through integrin dependent and independent mechanisms across immune lineages. J Nutr Biochem 2012; 24:832-41. [PMID: 22925918 DOI: 10.1016/j.jnutbio.2012.05.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2012] [Revised: 04/30/2012] [Accepted: 05/02/2012] [Indexed: 12/13/2022]
Abstract
Retinoids are essential in the proper establishment and maintenance of immunity. Although retinoids are implicated in immune related processes, their role in immune cell adhesion has not been well established. In this study, the effect of 9-cis-retinoic acid (9-cis-RA) on human hematopoietic cell adhesion was investigated. 9-cis-RA treatment specifically induced cell adhesion of the human immune cell lines HuT-78, NB4, RPMI 8866 and U937. Due to the prominent role of integrin receptors in mediating immune cell adhesion, we sought to evaluate if cell adhesion was integrin-dependent. By employing a variety of integrin antagonist including function-blocking antibodies and EDTA, we establish that 9-cis-RA prompts immune cell adhesion through established integrin receptors in addition to a novel integrin-independent process. The novel integrin-independent adhesion required the presence of retinoid and was attenuated by treatment with synthetic corticosteroids. Finally, we demonstrate that 9-cis-RA treatment of primary murine B-cells induces ex vivo adhesion that persists in the absence of integrin function. Our study is the first to demonstrate that 9-cis-RA influences immune cell adhesion through at least two functionally distinct mechanisms.
Collapse
Affiliation(s)
- Jarrett T Whelan
- Department of Biochemistry and Molecular Biology, The Brody School of Medicine at East Carolina University, Greenville, NC 27834, USA
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Williams JA, Batten SE, Harris M, Rockett BD, Shaikh SR, Stillwell W, Wassall SR. Docosahexaenoic and eicosapentaenoic acids segregate differently between raft and nonraft domains. Biophys J 2012; 103:228-37. [PMID: 22853900 DOI: 10.1016/j.bpj.2012.06.016] [Citation(s) in RCA: 142] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2011] [Revised: 06/06/2012] [Accepted: 06/12/2012] [Indexed: 01/01/2023] Open
Abstract
Omega-3 polyunsaturated fatty acids (n-3 PUFA), enriched in fish oils, are increasingly recognized to have potential benefits for treating many human afflictions. Despite the importance of PUFA, their molecular mechanism of action remains unclear. One emerging hypothesis is that phospholipids containing n-3 PUFA acyl chains modify the structure and composition of membrane rafts, thus affecting cell signaling. In this study the two major n-3 PUFA found in fish oils, eicosapentaenoic (EPA) and docosahexaenoic (DHA) acids, are compared. Using solid-state (2)H NMR spectroscopy we explored the molecular organization of 1-[(2)H(31)]palmitoyl-2-eicosapentaenoylphosphatidylcholine (PEPC-d(31)) and 1-[(2)H(31)]palmitoyl-2-docosahexaenoylphosphatidylcholine (PDPC-d(31)) in mixtures with sphingomyelin (SM) and cholesterol (chol). Our results indicate that whereas both PEPC-d(31) and PDPC-d(31) can accumulate into SM-rich/chol-rich raftlike domains, the tendency for DHA to incorporate into rafts is more than twice as great as for EPA. We propose that DHA may be the more bioactive component of fish oil that serves to disrupt lipid raft domain organization. This mechanism represents an evolution in the view of how PUFA remodel membrane architecture.
Collapse
Affiliation(s)
- Justin A Williams
- Department of Physics, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana, USA
| | | | | | | | | | | | | |
Collapse
|