Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
J Nutr ; 151(7): 1854-1878, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33982105

ABSTRACT

BACKGROUND: Many nutrients have powerful immunomodulatory actions with the potential to alter susceptibility to coronavirus disease 2019 (COVID-19) infection, progression to symptoms, likelihood of severe disease, and survival. OBJECTIVE: The aim was to review the latest evidence on how malnutrition across all its forms (under- and overnutrition and micronutrient status) may influence both susceptibility to, and progression of, COVID-19. METHODS: We synthesized information on 13 nutrition-related components and their potential interactions with COVID-19: overweight, obesity, and diabetes; protein-energy malnutrition; anemia; vitamins A, C, D, and E; PUFAs; iron; selenium; zinc; antioxidants; and nutritional support. For each section we provide: 1) a landscape review of pertinent material; 2) a systematic search of the literature in PubMed and EMBASE databases, including a wide range of preprint servers; and 3) a screen of 6 clinical trial registries. All original research was considered, without restriction to study design, and included if it covered: 1) severe acute respiratory syndrome coronavirus (CoV) 2 (SARS-CoV-2), Middle East respiratory syndrome CoV (MERS-CoV), or SARS-CoV viruses and 2) disease susceptibility or 3) disease progression, and 4) the nutritional component of interest. Searches took place between 16 May and 11 August 2020. RESULTS: Across the 13 searches, 2732 articles from PubMed and EMBASE, 4164 articles from the preprint servers, and 433 trials were returned. In the final narrative synthesis, we include 22 published articles, 38 preprint articles, and 79 trials. CONCLUSIONS: Currently there is limited evidence that high-dose supplements of micronutrients will either prevent severe disease or speed up recovery. However, results of clinical trials are eagerly awaited. Given the known impacts of all forms of malnutrition on the immune system, public health strategies to reduce micronutrient deficiencies and undernutrition remain of critical importance. Furthermore, there is strong evidence that prevention of obesity and type 2 diabetes will reduce the risk of serious COVID-19 outcomes. This review is registered at PROSPERO as CRD42020186194.


Subject(s)
Anemia/epidemiology , COVID-19/epidemiology , COVID-19/immunology , Diabetes Mellitus/epidemiology , Nutritional Status , Obesity/epidemiology , Protein-Energy Malnutrition/epidemiology , Antioxidants/metabolism , COVID-19/prevention & control , COVID-19/therapy , Comorbidity , Dietary Supplements , Disease Progression , Fatty Acids, Omega-3/immunology , Fatty Acids, Omega-6/immunology , Humans , Iron/immunology , Nutritional Support , SARS-CoV-2 , Selenium/immunology , Severity of Illness Index , Vitamins/immunology , Zinc/immunology
2.
Int J Mol Sci ; 20(10)2019 May 16.
Article in English | MEDLINE | ID: mdl-31100828

ABSTRACT

In spite of therapeutic improvements in the treatment of different hematologic malignancies, the prognosis of acute myeloid leukemia (AML) treated solely with conventional induction and consolidation chemotherapy remains poor, especially in association with high risk chromosomal or molecular aberrations. Recent discoveries describe the complex interaction of immune effector cells, as well as the role of the bone marrow microenvironment in the development, maintenance and progression of AML. Lipids, and in particular omega-3 as well as omega-6 polyunsaturated fatty acids (PUFAs) have been shown to play a vital role as signaling molecules of immune processes in numerous benign and malignant conditions. While the majority of research in cancer has been focused on the role of lipid mediators in solid tumors, some data are showing their involvement also in hematologic malignancies. There is a considerable amount of evidence that AML cells are targetable by innate and adaptive immune mechanisms, paving the way for immune therapy approaches in AML. In this article we review the current data showing the lipid mediator and lipidome patterns in AML and their potential links to immune mechanisms.


Subject(s)
Leukemia, Myeloid, Acute/drug therapy , Lipids/therapeutic use , Adaptive Immunity/drug effects , Bone Marrow , Disease Progression , Fatty Acids, Omega-3/immunology , Fatty Acids, Omega-3/therapeutic use , Fatty Acids, Omega-6/immunology , Fatty Acids, Omega-6/therapeutic use , Fatty Acids, Unsaturated , Hematologic Neoplasms/drug therapy , Hematopoiesis , Humans , Immunity, Innate/drug effects , Immunotherapy , Inflammation , Leukemia, Myeloid, Acute/immunology , Lipids/immunology , Neoplasms/drug therapy , Prognosis , Tumor Microenvironment
3.
Trends Mol Med ; 25(2): 124-135, 2019 02.
Article in English | MEDLINE | ID: mdl-30642681

ABSTRACT

The intestinal mucosa serves as a highly selective barrier that allows the absorption of nutrients and water while restricting microbiota access to tissues. This barrier is compromised in inflammatory conditions such as infectious colitis and inflammatory bowel disease (IBD). In response to mucosal injury, there is a temporal recruitment of leukocytes that crosstalk with epithelial cells to orchestrate repair. Specialized pro-resolving mediators (SPMs) play an important role in the resolution of inflammation and epithelial repair. SPMs actively promote resolution of inflammation by contributing to the clearance of neutrophils, stimulating efferocytosis, and promoting epithelial repair. SPMs have potential to serve as targeted therapeutic agents to be used in adjuvant therapy to promote resolution of inflammation and epithelial repair in chronic inflammatory diseases.


Subject(s)
Colitis/pathology , Inflammatory Bowel Diseases/pathology , Intestinal Mucosa/pathology , Animals , Colitis/immunology , Fatty Acids, Omega-3/analysis , Fatty Acids, Omega-3/immunology , Fatty Acids, Omega-6/analysis , Fatty Acids, Omega-6/immunology , Humans , Inflammatory Bowel Diseases/immunology , Intestinal Mucosa/immunology
4.
Clin Nutr ; 37(3): 1075-1081, 2018 06.
Article in English | MEDLINE | ID: mdl-28747247

ABSTRACT

The purpose of this review is to explain the historical and clinical background for intravenous fish oil administration, to evaluate its results by using a product specific metaanalysis, and to stimulate further research in the immune-modulatory potential of fish oil. Concerning the immune-modulatory effects of fatty acids, a study revealed that ω-3 as well as ω-6 fatty acids would prolong transplant survival, and only a mixture with an ω-6:ω-3 ratio of 2.1:1 would give immune-neutral results. In 1998, the label of a newly registered fish oil emulsion also acknowledged this immune-neutral ratio in conjunction with ω-6 lipids. Also, two fish oil-supplemented fat emulsions, registered in 2004, used a similar ω-6:ω-3 ratio. Such an immune-neutral ω-6:ω-3 ratio denoted progress for most patients compared to pure ω-6 lipid emulsions. However, this immune-neutrality might on the other hand be responsible for the limited positive clinical results gained so far in critically ill and surgical patients where in most cases significance could only be shown for the pooled effect of numerous trials. Our product specific metaanalysis also did not reveal any differences, neither in infections rates nor in ICU or hospital length of stay. To evaluate the immune-modulatory effect of fish oil administered alone, new dose finding studies, reporting relevant clinical outcome parameters, are required. Precise mechanistic or physiological biomarkers for the indication of such a therapy should also be developed and validated.


Subject(s)
Critical Care/methods , Fat Emulsions, Intravenous/therapeutic use , Immunologic Factors/therapeutic use , Parenteral Nutrition/methods , Postoperative Complications/prevention & control , Administration, Intravenous , Critical Illness , Fat Emulsions, Intravenous/administration & dosage , Fatty Acids, Omega-3/immunology , Fatty Acids, Omega-3/therapeutic use , Fatty Acids, Omega-6/immunology , Fatty Acids, Omega-6/therapeutic use , Fish Oils , Humans , Immunologic Factors/administration & dosage , Immunologic Factors/immunology , Postoperative Complications/immunology , Surgical Procedures, Operative
5.
Lipids ; 51(5): 571-82, 2016 05.
Article in English | MEDLINE | ID: mdl-26645280

ABSTRACT

Dysregulation of adipokines is a hallmark of obesity. Polyunsaturated fatty acids in fish oil may exert anti-inflammatory effects on adipose tissue mitigating the dysregulation of adipokines thereby preventing obesity. This study investigated the effects of high-fat diets containing different amounts of n3 polyunsaturated fatty acids (PUFA) on adiposity and adipokine production in mice. Mice were fed a low-fat or a high-fat diet with 16 or 45 % of energy from corn oil (low n3 PUFA) in comparison with a high-fat diet containing soybean or high-oleic sunflower oil (adequate n3 PUFA) or flaxseed or fish oil (high n3 PUFA) for 11 weeks. High-fat diets, regardless of types of oils, significantly increased body fat mass and body weights compared to the low-fat diet. Adipose fatty acid composition and contents reflected dietary fatty acid profiles. The high-fat fish oil diet significantly increased adiponectin and reduced leptin concentrations in both plasma and adipose tissue; it did not elevate plasma insulin concentration compared to the high-fat corn oil diet. All high-fat diets elevated concentrations of plasminogen activator inhibitor-1 (PAI-1) and monocyte chemoattractant protein-1 (MCP-1) but lowered resistin concentrations in both plasma and adipose tissue. In conclusion, fish oil may be beneficial in improving insulin sensitivity by upregulation of adiponectin and downregulation of leptin production; n3 and n6 PUFA do not play a role at the dietary levels tested in reducing adiposity and production of pro-inflammatory cytokines (leptin, PAI-1, MCP-1 and resistin) and anti-inflammatory cytokine adiponectin.


Subject(s)
Adipokines/immunology , Adiposity , Diet, High-Fat , Dietary Fats, Unsaturated/immunology , Fatty Acids, Omega-3/immunology , Fatty Acids, Omega-6/immunology , Adipokines/analysis , Adipokines/blood , Adipose Tissue/chemistry , Adipose Tissue/immunology , Animals , Diet, High-Fat/adverse effects , Dietary Fats, Unsaturated/analysis , Energy Intake , Fatty Acids, Omega-3/analysis , Fatty Acids, Omega-6/analysis , Insulin/analysis , Insulin/blood , Insulin/immunology , Male , Mice , Mice, Inbred C57BL , Obesity/immunology
6.
Br J Nutr ; 114(4): 577-85, 2015 Aug 28.
Article in English | MEDLINE | ID: mdl-26179875

ABSTRACT

Increased intake of vegetable oils rich in n-6 PUFA, including soyabean oil, has been associated with an increase in allergic disease. The present study aimed to determine the effect of an increasing dose of dietary vegetable oil on allergic outcomes in mice. To study this, mice received a 7 v. 10 % soyabean oil diet before and during oral sensitisation with whey or whey hyperimmune serum transfer. Another group of mice received partial whey hydrolysate (pWH) while being fed the diets before oral sensitisation. The acute allergic skin response, serum Ig level, mouse mast cell protease-1 (mMCP-1) concentration and/or splenic T-cell percentages were determined upon whey challenge. When the diets were provided before and during oral sensitisation, the acute allergic skin response was increased in mice fed the 10 % soyabean oil diet compared with the 7 % soyabean oil diet. Whey IgE and IgG1 levels remained unaltered, whereas mMCP-1 levels increased in mice fed the 10 % soyabean oil diet. Furthermore, allergic symptoms were increased in naive mice fed the 10 % soyabean oil diet and sensitised with whey hyperimmune serum. In addition to enhancing the mast cell response, the 10 % soyabean oil diet increased the percentage of activated Th1 and Th2 cells as well as increased the ratios of Th2:regulatory T cells and Th2:Th1 when compared with the 7 % soyabean oil diet. Oral tolerance induction by pWH was abrogated in mice fed the 10 % soyabean oil diet compared with those fed the 7 % soyabean oil diet during pretreatment with pWH. In conclusion, increased intake of soyabean oil rich in n-6 PUFA suppresses tolerance induction by pWH and enhances the severity of the allergic effector response in whey-allergic mice. Dietary vegetable oils rich in n-6 PUFA may enhance the susceptibility to develop or sustain food allergy.


Subject(s)
Diet/adverse effects , Fatty Acids, Omega-6/immunology , Immunity/drug effects , Milk Hypersensitivity , Milk Proteins/immunology , Soybean Oil/immunology , T-Lymphocyte Subsets/metabolism , Allergens , Animals , Chymases/blood , Dietary Fats/administration & dosage , Dietary Fats/adverse effects , Dietary Fats/immunology , Disease Models, Animal , Fatty Acids, Omega-6/administration & dosage , Fatty Acids, Omega-6/adverse effects , Feeding Behavior , Female , Immunoglobulins/blood , Mast Cells/metabolism , Mice , Milk Hypersensitivity/etiology , Soybean Oil/administration & dosage , Soybean Oil/adverse effects , Spleen/metabolism , T-Lymphocytes, Regulatory/metabolism , Th1 Cells/metabolism , Th1-Th2 Balance/drug effects , Th2 Cells/metabolism , Whey Proteins
7.
Mol Med Rep ; 11(6): 4149-57, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25672643

ABSTRACT

Omega­3 and ­6 polyunsaturated fatty acids (PUFAs) can directly or indirectly regulate immune homeostasis via inflammatory pathways, and components of these pathways are crucial targets of microRNAs (miRNAs). However, no study has examined the changes in the miRNA transcriptome during PUFA­regulated inflammatory processes. Here, we established PUFA diet­induced autoimmune­prone (AP) and autoimmune­averse (AA) rat models, and studied their physical characteristics and immune status. Additionally, miRNA expression patterns in the rat models were compared using microarray assays and bioinformatic methods. A total of 54 miRNAs were differentially expressed in common between the AP and the AA rats, and the changes in rno­miR­19b­3p, ­146b­5p and ­183­5p expression were validated using stem­loop reverse transcription­quantitative polymerase chain reaction. To better understand the mechanisms underlying PUFA­regulated miRNA changes during inflammation, computational algorithms and biological databases were used to identify the target genes of the three validated miRNAs. Furthermore, Gene Ontology (GO) term annotation and KEGG pathway analyses of the miRNA targets further allowed to explore the potential implication of the miRNAs in inflammatory pathways. The predicted PUFA­regulated inflammatory pathways included the Toll­like receptor (TLR), T cell receptor (TCR), NOD­like receptor (NLR), RIG­I­like receptor (RLR), mitogen­activated protein kinase (MAPK) and the transforming growth factor­ß (TGF­ß) pathway. This study is the first report, to the best of our knowledge, on in vivo comparative profiling of miRNA transcriptomes in PUFA diet­induced inflammatory rat models using a microarray approach. The results provide a useful resource for future investigation of the role of PUFA­regulated miRNAs in immune homeostasis.


Subject(s)
Fatty Acids, Omega-3/immunology , Fatty Acids, Omega-6/immunology , Gene Expression Profiling , Inflammation/genetics , MicroRNAs/genetics , Animals , Diet , Inflammation/immunology , MicroRNAs/immunology , Rats, Wistar , Transcriptome
8.
Curr Pharm Des ; 20(6): 946-53, 2014.
Article in English | MEDLINE | ID: mdl-23701554

ABSTRACT

There may be a causal relationship between intake of n-6 polyunsaturated fatty acids (PUFAs) and childhood allergic diseases. This can be explained by plausible biological mechanisms involving eicosanoid mediators produced from the n-6 PUFA arachidonic acid. Long chain n-3 PUFAs are found in fish and fish oils. These fatty acids act to oppose the actions of n-6 PUFAs. Thus, it is considered that n-3 PUFAs will lower the risk of developing allergic diseases. In support of this, protective associations have been reported between maternal fish intake during pregnancy and allergic outcomes in infants and children from those pregnancies. However, studies of fish intake during infancy and childhood and allergic outcomes in those infants or children are inconsistent, although some reported a protective association. Supplementing pregnant women with fish oil can induce immunologic changes in cord blood. This supplementation has been reported in some studies to decrease sensitisation to common food allergens and to lower the prevalence and severity of atopic dermatitis in the first year of life. The protective effect of maternal n-3 PUFAs may last until adolescence of the offspring. Fish oil supplementation in infancy may decrease the risk of developing some manifestations of allergic disease, although this benefit may not persist. Whether fish oil is a useful therapy in children with asthma receiving standard therapy is not clear from studies performed to date and this requires further exploration.


Subject(s)
Fatty Acids, Omega-3/immunology , Fatty Acids, Omega-6/immunology , Hypersensitivity/immunology , Adolescent , Adult , Age Factors , Animals , Child , Dietary Supplements , Fatty Acids, Omega-3/administration & dosage , Fatty Acids, Omega-6/administration & dosage , Female , Fish Oils/administration & dosage , Fish Oils/immunology , Food Hypersensitivity/immunology , Humans , Infant , Pregnancy
9.
Inflamm Bowel Dis ; 19(3): 650-61, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23328774

ABSTRACT

Recent epidemiological studies highlight the key role of the type of consumed unsaturated fatty acid and the development of ulcerative colitis (UC). We aimed to review the potential mechanisms behind the antiinflammatory effects of unsaturated fatty acids on intestinal inflammation, to discuss their potential limitations, and to propose a new reappraisal of polyunsaturated fatty acids (PUFAs) in the pathophysiology of inflammatory bowel disease (IBD). A literature search using PubMed was carried out to identify relevant studies (basic science, epidemiological studies, or clinical trials) with unsaturated fatty acids and IBD. Only articles published in English were included. IBD patients exhibit an altered lipid metabolism. While in vitro and in vivo studies have demonstrated the antiinflammatory properties of n-3 polyunsaturated fatty acids in experimental models IBD, results of clinical trials have been disappointing. In addition, the impact of fatty acid on innate immunity as an alternative therapeutic approach is explored. This may offer insight into therapeutic avenues for designing n-3 PUFA diet therapy for IBD.


Subject(s)
Crohn Disease , Fatty Acids, Omega-3 , Fatty Acids, Omega-6 , Crohn Disease/diet therapy , Crohn Disease/etiology , Crohn Disease/immunology , Crohn Disease/metabolism , Diet , Fatty Acids, Omega-3/immunology , Fatty Acids, Omega-3/metabolism , Fatty Acids, Omega-3/therapeutic use , Fatty Acids, Omega-6/immunology , Fatty Acids, Omega-6/metabolism , Fatty Acids, Omega-6/therapeutic use , Humans , Immunity, Innate , Intestines/immunology , Intestines/microbiology
10.
Surgery ; 151(5): 745-55, 2012 May.
Article in English | MEDLINE | ID: mdl-22244177

ABSTRACT

BACKGROUND: Parenteral nutrition (PN) is indispensable for meeting caloric and substrate needs of patients who cannot receive adequate amounts of enteral nutrition; however, PN impairs hepatic immunity. We examined the effects of ω-3 and -6 polyunsaturated fatty acids, added individually to fat-free PN, on hepatic immunity in a murine model. We focused on serum liver enzymes, cytokine production, histopathology, and the outcomes after intraportal bacterial challenge. METHODS: Male Institute of Cancer Research mice were randomized into 4 groups; ad libitum chow (CHOW), fat-free PN (FF-PN), PN + fish oil (FO-PN), or PN + safflower oil (SO-PN). After the mice had been fed for 5 days, hepatic mononuclear cells (MNCs) were isolated. The number of MNCs was counted and cytokine production (tumor necrosis factor [TNF]-α and interleukin [IL]-10) by hepatic MNCs in response to lipopolysaccharide (LPS) was measured. Blood samples were analyzed for hepatobiliary biochemical parameters. Moreover, 1.0 × 10(7) pseudomonas aeruginosa were delivered by intraportal injection. Survival and histology were examined. RESULTS: Hepatic MNC numbers were significantly less in the FO-PN and FF-PN than in the CHOW group, whereas the SO-PN group showed moderate recovery of hepatic MNC numbers. The CHOW, FO-PN, and SO-PN groups showed LPS dose-dependent increases in TNF-α levels. These increases were blunted in the FF-PN group. IL-10 levels were increased LPS dose-dependently in the CHOW and FO-PN groups, but no marked changes were observed with LPS stimulation in the SO-PN and FF-PN groups. Plasma levels of aspartate aminotransferase, alanine aminotransferase, and alkaline phosphatase were significantly greater in the FF-PN than in the FO- and SO-PN and CHOW groups. The FO-PN group showed significantly improved survival compared with the SO-PN and FF-PN groups, showing essentially no morphologic hepatic abnormalities. CONCLUSION: Addition of fish oil to PN was advantageous in terms of reversing PN-induced deterioration of hepatic immunity, as reflected by altered cytokine production. Fish oil administration was also useful for preventing PN-induced hepatobiliary dysfunction. These changes seem to result in better survival and to protect against severe tissue damage after intraportal bacterial challenge. This therapy may have the potential to ameliorate PN-induced impairment of host immunity and thereby decrease morbidity and mortality.


Subject(s)
Fatty Acids, Omega-3/immunology , Fatty Acids, Omega-6/immunology , Fish Oils/immunology , Leukocytes, Mononuclear/metabolism , Liver/immunology , Parenteral Nutrition/methods , Safflower Oil/immunology , Animals , Biomarkers/metabolism , Cytokines/metabolism , Fatty Acids, Omega-3/administration & dosage , Fatty Acids, Omega-6/administration & dosage , Fish Oils/administration & dosage , Lipopolysaccharides/administration & dosage , Lipopolysaccharides/immunology , Liver/cytology , Liver/microbiology , Liver/pathology , Male , Mice , Mice, Inbred ICR , Parenteral Nutrition/adverse effects , Pseudomonas Infections/immunology , Pseudomonas Infections/prevention & control , Pseudomonas aeruginosa/immunology , Random Allocation , Safflower Oil/administration & dosage
11.
Clin Exp Allergy ; 41(3): 407-16, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21255139

ABSTRACT

BACKGROUND: Prevalence of atopic disorders has increased rapidly, but aetiological factors responsible for this increase are still largely unknown. Prenatal exposure to a pro-inflammatory fatty acid status is hypothesized although little research has been carried out. OBJECTIVE: To investigate whether prenatal fatty acid exposures are associated with atopy in childhood. METHODS: In the KOALA Birth Cohort Study, maternal blood samples (n=1275) at 34-36 weeks of pregnancy were assayed for n-6 and n-3 long-chain polyunsaturated fatty acids (LCPs). The full spectrum of offspring atopic manifestations (wheeze, asthma, allergic rhinoconjunctivitis, eczema, atopic dermatitis, allergic sensitization, and high total IgE) until the age of 6-7 years was assessed by repeated parental questionnaires and measurements of total and specific IgE. Associations of maternal fatty acid status with child atopic outcomes were analysed using multivariable logistic regression and generalized estimating equations for repeated measurements. RESULTS: High ratio of maternal n-6 vs. n-3 LCPs was associated with a lower risk of eczema in the child (P for trend 0.012). More specifically, we found a decreased risk of eczema in the first 7 months of life with increasing arachidonic acid levels (P for trend 0.013). No associations were found between maternal fatty acids and offspring airway-related atopic manifestations, sensitization, or high total IgE. CONCLUSION AND CLINICAL RELEVANCE: The development of atopic disorders in early childhood is associated with prenatal exposure to n-6 vs. n-3 fatty acids, but with inconsistencies between different manifestations. Further exploration of associations with maternal diet and genetic variants in genes regulating fatty acid metabolism are required. This study shows that the influence of prenatal exposure to fatty acids on the risk of eczema in the child is limited to the first year of life.


Subject(s)
Diet/adverse effects , Fatty Acids, Omega-3/blood , Fatty Acids, Omega-6/blood , Hypersensitivity, Immediate/epidemiology , Child , Child, Preschool , Cohort Studies , Fatty Acids, Omega-3/immunology , Fatty Acids, Omega-6/immunology , Female , Humans , Hypersensitivity, Immediate/etiology , Hypersensitivity, Immediate/immunology , Infant , Infant, Newborn , Male , Maternal Exposure , Pregnancy , Prevalence , Surveys and Questionnaires
12.
Clin Nutr ; 29(1): 5-12, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19931231

ABSTRACT

Many chronic conditions involve excessive inflammation that is damaging to host tissues. Excessive or inappropriate inflammation and immunosuppression are components of the response to surgery, trauma, injury and infection in some individuals and these can lead, progressively, to sepsis and septic shock. Hyperinflammation is characterised by the production of inflammatory cytokines, eicosanoids and other inflammatory mediators, while the immunosuppression is characterised by impairment of antigen presentation and of certain T cell responses. N-6 fatty acids may contribute to the hyperinflamed and immunosuppressed states. N-3 fatty acids from fish oil decrease the production of inflammatory cytokines and eicosanoids. They act both directly (by replacing arachidonic acid as an eicosanoid precursor) and indirectly (by altering the expression of inflammatory genes through effects on transcription factor activation). Thus, these fatty acids are potentially useful anti-inflammatory agents and may be of benefit in patients with chronic inflammatory diseases or at risk of hyperinflammation and sepsis. An emerging application of n-3 fatty acids is in surgical or critically ill patients where they may be added to parenteral or enteral formulas. Studies to date are suggestive of clinical benefits from these approaches, although more robust data are needed especially in critically ill patients.


Subject(s)
Fatty Acids/immunology , Inflammation/immunology , Nutritional Physiological Phenomena/immunology , Animals , Anti-Inflammatory Agents/therapeutic use , Cell Nucleus/immunology , Critical Illness , Cytokines/immunology , Eicosanoids/immunology , Europe , Fatty Acids/pharmacology , Fatty Acids/therapeutic use , Fatty Acids, Omega-3/immunology , Fatty Acids, Omega-3/pharmacology , Fatty Acids, Omega-3/therapeutic use , Fatty Acids, Omega-6/immunology , Fish Oils/immunology , Fish Oils/pharmacology , Fish Oils/therapeutic use , Humans , Immune Tolerance/immunology , Inflammation/prevention & control , Membranes/immunology , Parenteral Nutrition/methods , Postoperative Complications/prevention & control , Societies, Medical
13.
Am J Clin Nutr ; 86(3): 682-9, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17823433

ABSTRACT

BACKGROUND: Breastfeeding accounts for a sizable proportion of infant HIV infections. Some fatty acids (FAs) are potent immunomodulators with virucidal activity, and their primary source in breastfed children is breast milk. OBJECTIVES: The aims of the study were to examine whether the percentage weight concentration of FAs in breast milk was associated with the risk of mother-to-child transmission (MTCT) of HIV by breastfeeding and with shedding of cell-free virus (CFV) or cell-associated virus (CAV) in breast milk. DESIGN: We conducted a case-control study nested within a cohort of HIV-infected Tanzanian women and children. We matched 59 incident breastfeeding MTCT cases to 59 nontransmitting controls based on the child's age at sample collection. We quantified FAs, CFV, and CAV in a breast milk sample collected before the infant's first positive HIV test. RESULTS: After adjustment for indicators of maternal HIV disease stage, the risk of MTCT was inversely related to 11c,14c-eicosadienoic acid [odds ratio (OR) for quartile 4 compared with quartile 1: 0.21; P for trend = 0.04], arachidonic acid (OR: 0.21; P for trend = 0.03), and dihomo-gamma-linolenic acid (OR: 0.24; P for trend = 0.03); the latter 2 were also linearly, inversely related to virus shedding in breast milk. Lauric acid and pentadecanoic acid were associated with increased MTCT, whereas trans FAs were related to higher CAV and CFV. CONCLUSION: Increasing concentrations of long-chain n-6 polyunsaturated FAs in breast milk might reduce the risk of MTCT.


Subject(s)
Fatty Acids, Omega-6/analysis , HIV Infections/transmission , HIV-1 , Milk, Human/chemistry , Milk, Human/virology , Virus Shedding , Adult , Arachidonic Acids/analysis , Breast Feeding/adverse effects , Case-Control Studies , Cell-Free System , Cohort Studies , Eicosapentaenoic Acid/analysis , Fatty Acids/analysis , Fatty Acids, Omega-6/immunology , Female , HIV Infections/prevention & control , HIV-1/isolation & purification , Humans , Infant , Infant, Newborn , Infectious Disease Transmission, Vertical/prevention & control , Lauric Acids/analysis , Tanzania/epidemiology , gamma-Linolenic Acid/analysis
14.
Am J Clin Nutr ; 85(5): 1171-84, 2007 May.
Article in English | MEDLINE | ID: mdl-17490951

ABSTRACT

Total parenteral nutrition is the final option for nutritional support of patients with severe intestinal failure. Lipid emulsions constitute the main source of fuel calories and fatty acids (FAs) in parenteral nutrition formulations. However, adverse effects on patient outcomes have been attributed to the use of lipids, mostly in relation to impaired immune defenses and altered inflammatory responses. Over the years, this issue has remained in the limelight, also because technical advances have provided no safeguard against the most daunting problems, ie, infectious complications. Nevertheless, numerous investigations have failed to produce a clear picture of the immunologic characteristics of the most commonly used soybean oil-derived lipid emulsions, although their high content of n-6 polyunsaturated FAs (PUFAs) has been considered a drawback because of their proinflammatory potential. This concern initiated the development of emulsions in which part of the n-6 FA component is replaced by less bioactive FAs, such as coconut oil (rich in medium-chain saturated FAs) or olive oil (rich in the n-9 monounsaturated FA oleic acid). Another approach has been to use fish oil (rich in n-3 PUFA), the FAs of which have biological activities different from those of n-6 PUFAs. Recent studies on the modulation of host defenses and inflammation by fish-oil emulsions have yielded consistent data, which indicate that these emulsions may provide a tool to beneficially alter the course of immune-mediated conditions. Although most of these lipids have not yet become available on the US market, this review synthesizes available information on immunologic characteristics of the different lipids that currently can be applied via parenteral nutrition support.


Subject(s)
Dietary Fats, Unsaturated/immunology , Fat Emulsions, Intravenous/adverse effects , Immune System/drug effects , Lipid Metabolism/drug effects , Parenteral Nutrition, Total/methods , Coconut Oil , Dietary Fats, Unsaturated/administration & dosage , Fat Emulsions, Intravenous/chemistry , Fat Emulsions, Intravenous/metabolism , Fatty Acids, Omega-3/administration & dosage , Fatty Acids, Omega-3/immunology , Fatty Acids, Omega-6/administration & dosage , Fatty Acids, Omega-6/immunology , Fish Oils/administration & dosage , Fish Oils/chemistry , Fish Oils/immunology , Humans , Lipid Metabolism/physiology , Membrane Lipids/metabolism , Olive Oil , Plant Oils , Soybean Oil/administration & dosage , Soybean Oil/chemistry , Soybean Oil/immunology
15.
Annu Rev Nutr ; 26: 45-73, 2006.
Article in English | MEDLINE | ID: mdl-16848700

ABSTRACT

Research describing fatty acids as modulators of inflammation and immune responses abounds. Many of these studies have focused on one particular group of fatty acids, omega-3. The data from animal studies have shown that these fatty acids can have powerful anti-inflammatory and immunomodulatory activities in a wide array of diseases (e.g., autoimmunity, arthritis, and infection). However, the evidence from human trials is more equivocal. In this review, a historical framework for understanding how and why fatty acids may affect the immune system is provided. Second, highlights of two recent landmark reports from the Agency for Healthcare Research and Quality are presented. These reports critically evaluate the evidence from human clinical trials of omega-3 fatty acids and rheumatoid arthritis, asthma, and a few other immune-mediated diseases. Third, the data from human clinical trials investigating the impact of various bioactive fatty acids on ex vivo and in vivo immune response are reviewed. Limitations in experimental design and immune assays commonly used are discussed. The discordance between expectation and evidence in this field has been a disappointment. Recommendations for improving both animal-based and human studies are provided.


Subject(s)
Dietary Fats, Unsaturated/immunology , Fatty Acids, Omega-3/immunology , Fatty Acids, Unsaturated/immunology , Inflammation/metabolism , Animals , Arthritis, Rheumatoid/diet therapy , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/metabolism , Asthma/diet therapy , Asthma/immunology , Asthma/metabolism , Dietary Fats, Unsaturated/metabolism , Disease Models, Animal , Evidence-Based Medicine , Fatty Acids, Omega-3/metabolism , Fatty Acids, Omega-6/immunology , Fatty Acids, Omega-6/metabolism , Fatty Acids, Unsaturated/metabolism , Humans , Inflammation/diet therapy , Inflammation/immunology
17.
Lipids ; 38(4): 343-52, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12848278

ABSTRACT

The immune system is involved in host defense against infectious agents, tumor cells, and environmental insults. Inflammation is an important component of the early immunologic response. Inappropriate or dysfunctional immune responses underlie acute and chronic inflammatory diseases. The n-6 PUFA arachidonic acid (AA) is the precursor of prostaglandins, leukotrienes, and related compounds that have important roles in inflammation and in the regulation of immunity. Feeding fish oil results in partial replacement of AA in cell membranes by EPA. This leads to decreased production of AA-derived mediators, through several mechanisms, including decreased availability of AA, competition for cyclooxygenase (COX) and lipoxygenase (LOX) enzymes, and decreased expression of COX-2 and 5-LOX. This alone is a potentially beneficial anti-inflammatory effect of n-3 FA. However, n-3 FA have a number of other effects that might occur downstream of altered eicosanoid production or might be independent of this effect. For example, dietary fish oil results in suppressed production of proinflammatory cytokines and can modulate adhesion molecule expression. These effects occur at the level of altered gene expression. Fish oil feeding has been shown to ameliorate the symptoms of some animal models of autoimmune disease and to protect against the effects of endotoxin. Clinical studies have reported that oral fish oil supplementation has beneficial effects in rheumatoid arthritis and among some asthmatics, supporting the idea that the n-3 FA in fish oil are anti-inflammatory. There are indications that the inclusion of fish oil in enteral and parenteral formulae is beneficial to patients.


Subject(s)
Fatty Acids, Unsaturated/immunology , Fatty Acids, Unsaturated/metabolism , Inflammation/immunology , Triglycerides/immunology , Triglycerides/metabolism , Animals , Anti-Inflammatory Agents/immunology , Anti-Inflammatory Agents/metabolism , Anti-Inflammatory Agents/pharmacology , Cell Adhesion Molecules/biosynthesis , Cell Adhesion Molecules/genetics , Cytokines/biosynthesis , Cytokines/immunology , Eicosanoids/biosynthesis , Fatty Acids, Omega-3 , Fatty Acids, Omega-6/immunology , Fatty Acids, Omega-6/metabolism , Fatty Acids, Unsaturated/pharmacology , Humans , Inflammation/metabolism , Triglycerides/pharmacology , Up-Regulation/drug effects
18.
Lipids ; 38(4): 323-41, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12848277

ABSTRACT

The essentiality of n-6 polyunsaturated fatty acids (PUFA) is described in relation to a thymus/thymocyte accretion of arachidonic acid (20:4n-6, AA) in early development, and the high requirement of lymphoid and other cells of the immune system for AA and linoleic acid (1 8:2n-6, LA) for membrane phospholipids. Low n-6 PUFA intakes enhance whereas high intakes decrease certain immune functions. Evidence from in vitro and in vivo studies for a role of AA metabolites in immune cell development and functions shows that they can limit or regulate cellular immune reactions and can induce deviation toward a T helper (Th)2-like immune response. In contrast to the effects of the oxidative metabolites of AA, the longer-chain n-6 PUFA produced by gamma-linolenic acid (18:3n-6, GLA) feeding decreases the Th2 cytokine and immunoglobulin (Ig)G1 antibody response. The n-6 PUFA, GLA, dihomo-gamma-linolenic acid (20:3n-6, DHLA) and AA, and certain oxidative metabolites of AA can also induce T-regulatory cell activity, e.g., transforming growth factor (TGF)-beta-producing T cells; GLA feeding studies also demonstrate reduced proinflammatory interleukin (IL)-1 and tumor necrosis factor (TNF)-alpha production. Low intakes of long-chain n-3 fatty acids (fish oils) enhance certain immune functions, whereas high intakes are inhibitory on a wide range of functions, e.g., antigen presentation, adhesion molecule expression, Th1 and Th2 responses, proinflammatory cytokine and eicosanoid production, and they induce lymphocyte apoptosis. Vitamin E has a demonstrable critical role in long-chain n-3 PUFA interactions with immune functions, often reversing the effects of fish oil. The effect of dietary fatty acids on animal autoimmune disease models depends on both the autoimmune model and the amount and type of fatty acids fed. Diets low in fat, essential fatty acid deficient (EFAD), or high in long-chain n-3 PUFA from fish oils increase survival and reduce disease severity in spontaneous autoantibody-mediated disease, whereas high-fat LA-rich diets increase disease severity. In experimentally induced T cell-mediated autoimmune disease, EFAD diets or diets supplemented with long-chain n-3 PUFA augment disease, whereas n-6 PUFA prevent or reduce the severity. In contrast, in both T cell- and antibody-mediated autoimmune disease, the desaturated/elongated metabolites of LA are protective. PUFA of both the n-6 and n-3 families are clinically useful in human autoimmune-inflammatory disorders, but the precise mechanisms by which these fatty acids exert their clinical effects are not well understood. Finally, the view that all n-6 PUFA are proinflammatory requires revision, in part, and their essential regulatory and developmental role in the immune system warrants appreciation.


Subject(s)
Autoimmunity/physiology , Fatty Acids, Omega-6/immunology , Fatty Acids, Unsaturated/immunology , Triglycerides/immunology , Animals , Dietary Fats, Unsaturated/metabolism , Dietary Fats, Unsaturated/pharmacology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Fatty Acids, Omega-3 , Fatty Acids, Omega-6/metabolism , Fatty Acids, Unsaturated/metabolism , Humans , Immune System/immunology , Immune System/metabolism , Inflammation/immunology , Inflammation/metabolism , Triglycerides/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...