Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 403
Filter
1.
Int J Sport Nutr Exerc Metab ; 34(4): 218-222, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38648883

ABSTRACT

Optimal omega-3 status, influenced by increased intake of eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), is vital for physiological health. This study investigated the impact of ad libitum fish oil supplementation on the omega-3 status of female athletes in a professional rugby league team during a competitive season. Twenty-four (n = 24) athletes participated, and their omega-3 status was assessed using the Omega-3 Index (O3I) and arachidonic acid (AA) to EPA ratio through finger-prick blood samples taken at the start and end of the season. They were given access to a fish oil supplement (PILLAR Performance, Australia) with a recommended daily dose of four capsules per day (2,160 mg EPA and 1,440 mg docosahexaenoic acid). At the beginning of the season, the group mean O3I was 4.77% (95% confidence interval [CI: 4.50, 5.04]) and the AA to EPA ratio was 14.89 (95% CI [13.22, 16.55]). None of the athletes had an O3I exceeding 8%. By the season's end, the O3I was a significantly increased to 7.28% (95% CI [6.64, 7.93], p < .0001) and AA to EPA ratio significantly decreased to a mean of 6.67 (95% CI [5.02, 8.31], p < .0001), driven primarily by the significant increase in EPA of +1.14% (95% CI [0.77, 1.51], p < .0001). However, these changes were varied between the athletes and most likely due to compliance. This study has demonstrated that using the objective O3I feedback scale is possible with elite female rugby athletes, but individual strategies will be required to achieve daily intake targets of EPA + DHA.


Subject(s)
Athletes , Dietary Supplements , Docosahexaenoic Acids , Eicosapentaenoic Acid , Fatty Acids, Omega-3 , Fish Oils , Football , Humans , Female , Fish Oils/administration & dosage , Australia , Fatty Acids, Omega-3/blood , Fatty Acids, Omega-3/administration & dosage , Eicosapentaenoic Acid/blood , Eicosapentaenoic Acid/administration & dosage , Young Adult , Docosahexaenoic Acids/blood , Docosahexaenoic Acids/administration & dosage , Adult , Sports Nutritional Physiological Phenomena , Arachidonic Acid/blood , Arachidonic Acid/administration & dosage , Nutritional Status
2.
Article in English | MEDLINE | ID: mdl-38377640

ABSTRACT

Enteral supplementation with arachidonic acid (AA) and docosahexaenoic acid (DHA) in extremely preterm infants has shown beneficial effects on retinopathy of prematurity and pulmonary outcome whereas exclusive DHA supplementation has been associated with increased pulmonary morbidity. This secondary analysis evaluates pulmonary outcome in 204 extremely preterm infants, randomized to receive AA (100 mg/kg/day) and DHA (50 mg/kg/day) enterally from birth until term age or standard care. Pulmonary morbidity was primarily assessed based on severity of bronchopulmonary dysplasia (BPD). Serum levels of AA and DHA during the first 28 days were analysed in relation to BPD. Supplementation with AA:DHA was not associated with increased BPD severity, adjusted OR 1.48 (95 % CI 0.85-2.61), nor with increased need for respiratory support at post menstrual age 36 weeks or duration of oxygen supplementation. Every 1 % increase in AA was associated with a reduction of BPD severity, adjusted OR 0.73 (95 % CI 0.58-0.92). In conclusion, in this study, with limited statistical power, enteral supplementation with AA:DHA was not associated with an increased risk of pulmonary morbidity, but higher levels of AA were associated with less severe BPD. Whether AA or the combination of AA and DHA have beneficial roles in the immature lung needs further research.


Subject(s)
Arachidonic Acid , Bronchopulmonary Dysplasia , Dietary Supplements , Docosahexaenoic Acids , Infant, Extremely Premature , Humans , Docosahexaenoic Acids/administration & dosage , Arachidonic Acid/administration & dosage , Arachidonic Acid/blood , Infant, Newborn , Female , Bronchopulmonary Dysplasia/prevention & control , Male , Enteral Nutrition , Lung/drug effects , Treatment Outcome
3.
Nutrients ; 13(5)2021 Apr 27.
Article in English | MEDLINE | ID: mdl-33925724

ABSTRACT

Arachidonic acid (ARA; 20:4n6) and docosahexaenoic acid (DHA; 22:6n3) are polyunsaturated fatty acids (FA) naturally present in breast milk and added to most North American infant formulas (IF). We investigated the safety and efficacy of novel sodium and potassium salts of arachidonic acid as bioequivalent to support tissue levels of ARA comparable to the parent oil; M. alpina oil (Na-ARA and K-ARA) and including a Na-DHA group. Pigs of both sexes were randomized to one of five dietary treatments (n = 16 per treatment; 8 male and 8 female) from postnatal day 2 to 23. ARA and DHA were included as either triglyceride (TG) or salt. Target dietary ARA/DHA concentrations as percent of total FA by weight were as follows: TT (0.47 TG/0.32 TG), NaT (0.47 Na-salt/0.32 TG), KT (0.47 K-salt/0.32 TG), and Na0 (0.47 Na-salt/0.00), NaNa (0.47 Na-salt/0.32 Na-salt). The primary outcome in this study was bioequivalence of ARA brain accretion. Growth performance; blood and tissue fatty acid levels; liver histology; complete blood cell counts; and serum chemistries were all evaluated. Overall, diets containing test sources of ARA and DHA did not affect growth performance; liver histology; or substantially influence hematological outcomes as compared with TT. The results confirm that the use of Na and K salt forms of ARA yield bioequivalent ARA accretion in the cerebral cortex and retinal tissue compared to TG-ARA. These findings confirm that use of Na-ARA and K-ARA salts in the young pig was safe and nutritionally bioequivalent to TG-ARA for critical neural tissues.


Subject(s)
Arachidonic Acid/administration & dosage , Arachidonic Acid/blood , Diet/methods , Potassium/administration & dosage , Sodium/administration & dosage , Animals , Female , Male , Models, Animal , Potassium/blood , Salts/administration & dosage , Salts/blood , Sodium/blood , Swine
4.
Food Funct ; 12(2): 802-814, 2021 Jan 21.
Article in English | MEDLINE | ID: mdl-33393955

ABSTRACT

Alibertia edulis leaf extract is commonly used in folk medicine, with rutin caffeic and vanillic acids being its major compounds. The Alibertia edulis leaf extract was investigated for its pharmacological effects via platelet aggregation, calcium mobilization, cyclic nucleotides levels, vasodilator-stimulated phosphoprotein Ser157 and Ser239 and protein kinase Cß2 phosphorylation, thromboxane B2, cyclooxygenases 1 and 2, docking and molecular dynamics. Alibertia edulis leaf extract significantly inhibited (100-1000 µg mL-1) platelet aggregation induced by different agonists. Arachidonic acid increased levels of calcium and thromboxane B2, phosphorylation of vasodilator-stimulated phosphoprotein Ser157 and Ser239, and protein kinase Cß, which were significantly reduced by Alibertia edulis leaf extract, rutin, and caffeic acid as well mixtures of rutin/caffeic acid. Cyclooxygenase 1 activity was inhibited for Alibertia edulis leaf extract, rutin and caffeic acid. These inhibitions were firsrtly explored by specific stabilization of rutin and caffeic acid compared to diclofenac at the catalytic site from docking score and free-energy dissociation profiles. Then, simulations detailed the rutin interactions close to the heme group and Tyr385, responsible for catalyzing the conversion of arachidonic acid to its products. Our results reveal the antiplatelet aggregation properties of Alibertia edulis leaf extract, rutin and caffeic acid providing pharmacological information about its origin from cyclooxygenase 1 inhibition and its downstream pathway.


Subject(s)
Gene Expression Regulation/drug effects , Plant Extracts/pharmacology , Platelet Aggregation/drug effects , Prostaglandin-Endoperoxide Synthases/metabolism , Rubiaceae/chemistry , Thromboxanes/antagonists & inhibitors , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/administration & dosage , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Adenosine Diphosphate/administration & dosage , Adenosine Diphosphate/pharmacology , Animals , Arachidonic Acid/administration & dosage , Arachidonic Acid/pharmacology , Calcium/metabolism , Collagen/administration & dosage , Collagen/pharmacology , Cyclooxygenase Inhibitors , Humans , Plant Extracts/chemistry , Plant Leaves/chemistry , Thromboxanes/genetics , Thromboxanes/metabolism , Zebrafish
5.
J Pediatr ; 232: 23-30.e1, 2021 05.
Article in English | MEDLINE | ID: mdl-33358843

ABSTRACT

OBJECTIVE: To determine feasibility of providing a concentrated emulsified long-chain polyunsaturated fatty acids (LCPUFA) supplement to very low birth weight infants, and to evaluate blood LCPUFA concentrations at 2 and 8 weeks of study supplementation. STUDY DESIGN: This prospective, randomized, double-blind, placebo-controlled trial randomized infants to receive (1) LCPUFA-120 (a supplement of 40 mg/kg/day docosahexaenoic acid [DHA] and 80 mg/kg/day arachidonic acid [ARA]; DHA:ARA at 1:2 ratio), (2) LCPUFA-360 (a supplement of 120 mg/kg/day DHA and 240 mg/kg/day ARA), or (3) sunflower oil (placebo control). Infants received supplement daily for 8 weeks or until discharge, whichever came first. Whole blood LCPUFA levels (wt%; g/100 g) were measured at baseline, 2 weeks, and 8 weeks. RESULTS: Infants were 28 weeks of gestation (IQR, 27-30 weeks of gestation) and weighed 1040 g (IQR, 910-1245 g). At 2 weeks, the change in blood DHA (wt%) from baseline differed significantly among groups (sunflower oil, n = 6; -0.63 [IQR, -0.96 to -0.55]; LCPUFA-120: n = 12; -0.14 [IQR, -0.72 to -0.26]; LCPUFA-360, n = 12; 0.46 [IQR, 0.17-0.81]; P = .002 across groups). Change in blood ARA (wt%) also differed by group (sunflower oil: -2.2 [IQR, -3.9 to -1.7]; LCPUFA-120: 0.1 [IQR, -2.1 to 1.1] vs LCPUFA-360: 2.9 IQR, 1.5 to 4.5]; P = .0002). Change from baseline to 8 weeks significantly differed between groups for DHA (P = .02) and ARA (P = .003). CONCLUSIONS: Enteral LCPUFA supplementation supported higher blood DHA by 2 weeks. LCPUFA supplementation at 360 mg of combined DHA and ARA is likely necessary to reduce declines as well as allow increases in whole blood concentrations in the first 8 weeks of life. TRIAL REGISTRATION: Clinicaltrials.gov: NCT03192839.


Subject(s)
Arachidonic Acid/administration & dosage , Dietary Supplements , Docosahexaenoic Acids/administration & dosage , Enteral Nutrition , Infant, Very Low Birth Weight , Arachidonic Acid/blood , Docosahexaenoic Acids/blood , Double-Blind Method , Female , Humans , Infant , Infant, Newborn , Infant, Premature , Male , Prospective Studies
6.
Arch Med Res ; 52(1): 107-120, 2021 01.
Article in English | MEDLINE | ID: mdl-32981754

ABSTRACT

Previously, I suggested that arachidonic acid (AA, 20:4 n-6) and similar bioactive lipids (BALs) inactivate SARS-CoV-2 and thus, may be of benefit in the prevention and treatment of COVID-19. This proposal is supported by the observation that (i) macrophages and T cells (including NK cells, cytotoxic killer cells and other immunocytes) release AA and other BALs especially in the lungs to inactivate various microbes; (ii) pro-inflammatory metabolites prostaglandin E2 (PGE2) and leukotrienes (LTs) and anti-inflammatory lipoxin A4 (LXA4) derived from AA (similarly, resolvins, protectins and maresins derived from eicosapentaenoic acid: EPA and docosahexaenoic acid: DHA) facilitate the generation of M1 (pro-inflammatory) and M2 (anti-inflammatory) macrophages respectively; (iii) AA, PGE2, LXA4 and other BALs inhibit interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) synthesis; (iv) mesenchymal stem cells (MSCs) that are of benefit in COVID-19 elaborate LXA4 to bring about their beneficial actions and (v) subjects with insulin resistance, obesity, type 2 diabetes mellitus, hypertension, coronary heart disease and the elderly have significantly low plasma concentrations of AA and LXA4 that may render them more susceptible to SARS-CoV-2 infection and cytokine storm that is associated with increased mortality seen in COVID-19. Statins, colchicine, and corticosteroids that appear to be of benefit in COVID-19 can influence BALs metabolism. AA, and other BALs influence cell membrane fluidity and thus, regulate ACE-2 (angiotensin converting enzyme-2) receptors (the ligand through which SARS-CoV2 enters the cell) receptors. These observations lend support to the contention that administration of BALs especially, AA could be of significant benefit in prevention and management of COVI-19 and other enveloped viruses.


Subject(s)
COVID-19 Drug Treatment , COVID-19/blood , Lipids/administration & dosage , Lipids/blood , Animals , Arachidonic Acid/administration & dosage , Arachidonic Acid/blood , COVID-19/immunology , Cytokines/immunology , Humans , Rats , SARS-CoV-2/isolation & purification
7.
Pediatrics ; 146(4)2020 10.
Article in English | MEDLINE | ID: mdl-32887793

ABSTRACT

BACKGROUND AND OBJECTIVES: Children born preterm experience socioemotional difficulties, including increased risk of autism spectrum disorder (ASD). In this secondary analysis, we tested the effect of combined docosahexaenoic acid (DHA) and arachidonic acid (AA) supplementation during toddlerhood on caregiver-reported socioemotional outcomes of children born preterm. We hypothesized that children randomly assigned to DHA + AA would display better socioemotional outcomes compared with those randomly assigned to a placebo. METHODS: Omega Tots was a single-site randomized, fully masked, parallel-group, placebo-controlled trial. Children (N = 377) were 10 to 16 months at enrollment, born at <35 weeks' gestation, and assigned to 180 days of daily 200-mg DHA + 200-mg AA supplementation or a placebo (400 mg corn oil). Caregivers completed the Brief Infant-Toddler Social and Emotional Assessment and the Pervasive Developmental Disorders Screening Test-II, Stage 2 at the end of the trial. Liner mixed models and log-binomial regression compared socioemotional outcomes between the DHA + AA and placebo groups. RESULTS: Outcome data were available for 83% of children (n treatment = 161; n placebo = 153). Differences between DHA + AA and placebo groups on Brief Infant-Toddler Social and Emotional Assessment scores were of small magnitude (Cohen's d ≤ 0.15) and not statistically significant. Children randomly assigned to DHA + AA had a decreased risk of scoring at-risk for ASD on the Pervasive Developmental Disorders Screening Test-II, Stage 2 (21% vs 32%; risk ratio = 0.66 [95% confidence interval: 0.45 to 0.97]; risk difference = -0.11 [95% confidence interval: -0.21 to -0.01]) compared with children randomly assigned to a placebo. CONCLUSIONS: No evidence of benefit of DHA + AA supplementation on caregiver-reported outcomes of broad socioemotional development was observed. Supplementation resulted in decreased risk of clinical concern for ASD. Further exploration in larger samples of preterm children and continued follow-up of children who received DHA + AA supplementation as they approach school age is warranted.


Subject(s)
Arachidonic Acid/administration & dosage , Autism Spectrum Disorder/prevention & control , Child Development/drug effects , Dietary Supplements , Docosahexaenoic Acids/administration & dosage , Confidence Intervals , Female , Gestational Age , Humans , Infant , Infant, Premature , Male , Medication Adherence , Placebos/administration & dosage , Sex Factors , Treatment Outcome
8.
Sci Rep ; 10(1): 13824, 2020 08 14.
Article in English | MEDLINE | ID: mdl-32796880

ABSTRACT

The present work assessed the effects of dietary ratios of essential fatty acids, arachidonic (ARA), eicosapentaenoic (EPA) and docosahexaenoic acid (DHA), on liver and intestine oxidative status, intestinal histomorphology and gut microbiota of gilthead sea bream. Four isoproteic and isolipidic plant-based diets were formulated containing a vegetable oil blend as the main lipid source. Diets were supplemented with ARA/EPA/DHA levels (%DM) equivalent to: 2%:0.2%:0.1% (Diet A); 1.0%:0.4%:0.4% (Diet B); 0%:0.6%:0.6% (Diet C); 0%:0.3%:1.5% (Diet D) and tested in triplicate groups for 56 days. Lipid peroxidation was higher in fish fed diets C and D while no differences were reported between diets regarding total, oxidized, and reduced glutathione, and oxidative stress index. Glutathione reductase was higher in fish fed diet A than diets C and D. No histological alterations were observed in the distal intestine. Lower microbiota diversity was observed in intestinal mucosa of fish fed diet C than A, while diets C and D enabled the proliferation of health-promoting bacteria from Bacteroidetes phylum (Asinibacterium sp.) and the absence of pathogenic species like Edwardsiella tarda. Overall, results suggest that a balance between dietary ARA/EPA + DHA promotes gilthead sea bream juveniles' health however higher dietary content of n-3 LC-PUFA might limited the presence of microbial pathogens in intestinal mucosa.


Subject(s)
Animal Feed , Animal Nutritional Physiological Phenomena/physiology , Arachidonic Acid/administration & dosage , Diet/veterinary , Dietary Supplements , Docosahexaenoic Acids/administration & dosage , Eicosapentaenoic Acid/administration & dosage , Gastrointestinal Microbiome , Oxidative Stress , Sea Bream/metabolism , Sea Bream/microbiology , Animals , Glutathione Reductase/metabolism , Intestinal Mucosa , Intestines/microbiology , Lipid Peroxidation , Liver/metabolism
9.
FASEB J ; 34(8): 10027-10040, 2020 08.
Article in English | MEDLINE | ID: mdl-32592197

ABSTRACT

Aspirin prevents thrombosis by inhibiting platelet cyclooxygenase (COX)-1 activity and the production of thromboxane (Tx)A2 , a pro-thrombotic eicosanoid. However, the non-platelet actions of aspirin limit its antithrombotic effects. Here, we used platelet-COX-1-ko mice to define the platelet and non-platelet eicosanoids affected by aspirin. Mass-spectrometry analysis demonstrated blood from platelet-COX-1-ko and global-COX-1-ko mice produced similar eicosanoid profiles in vitro: for example, formation of TxA2 , prostaglandin (PG) F2α , 11-hydroxyeicosatraenoic acid (HETE), and 15-HETE was absent in both platelet- and global-COX-1-ko mice. Conversely, in vivo, platelet-COX-1-ko mice had a distinctly different profile from global-COX-1-ko or aspirin-treated control mice, notably significantly higher levels of PGI2 metabolite. Ingenuity Pathway Analysis (IPA) predicted that platelet-COX-1-ko mice would be protected from thrombosis, forming less pro-thrombotic TxA2 and PGE2 . Conversely, aspirin or lack of systemic COX-1 activity decreased the synthesis of anti-aggregatory PGI2 and PGD2 at non-platelet sites leading to predicted thrombosis increase. In vitro and in vivo thrombosis studies proved these predictions. Overall, we have established the eicosanoid profiles linked to inhibition of COX-1 in platelets and in the remainder of the cardiovascular system and linked them to anti- and pro-thrombotic effects of aspirin. These results explain why increasing aspirin dosage or aspirin addition to other drugs may lessen antithrombotic protection.


Subject(s)
Aspirin/pharmacology , Blood Platelets/metabolism , Cyclooxygenase 1/physiology , Cyclooxygenase Inhibitors/pharmacology , Eicosanoids/metabolism , Membrane Proteins/physiology , Thrombosis/metabolism , Animals , Arachidonic Acid/administration & dosage , Blood Platelets/drug effects , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Thrombosis/drug therapy , Thrombosis/pathology
10.
Eur Rev Med Pharmacol Sci ; 24(4): 2110-2119, 2020 02.
Article in English | MEDLINE | ID: mdl-32141581

ABSTRACT

OBJECTIVE: The aim of this study is to understand whether the responsiveness of the hypothalamic-pituitary-adrenal (HPA) axis to stress increases excessively with aging in senescence-accelerated mice-prone 10 (SAMP10) and to investigate the role of arachidonic acid (ARA) in this process. MATERIALS AND METHODS: The area under the curve of CORT concentration (CORT-AUC), an index of the HPA axis responsiveness to stress, was assessed in SAMP10 subjected to a 30-minute restraint stress up to 120 minutes after the restraint stress onset. Furthermore, the HPA axis responsiveness was evaluated in aged SAMP10 fed 0.4% ARA-containing diet (ARA group) or control diet (CON group) for 4 weeks. Three weeks later, these mice were divided into a group with a 30-minute restraint stress (CON-S or ARA-S group) and a group without restraint stress (CON-NS or ARA-NS group). Hippocampi were collected after stress release and fatty acid and glucocorticoid receptor (GR) protein levels were evaluated in the nucleus and cytosol. RESULTS: The CORT-AUC of aged SAMP10 was 21% significantly higher than that of young SAMP10. In the ARA group, hippocampal ARA was 0.5% significantly higher than that in the CON group. CORT-AUC in the ARA group was 24% significantly lower than that in the CON group. The ratio of GR protein levels in the nucleus and cytosol in the ARA-S group was 1.72 times significantly higher than that in the ARA-NS group but no difference was observed between the CON-S and CON-NS groups. CONCLUSIONS: Dietary ARA seems to suppress age-related excessive enhancement of the HPA axis responsiveness via attenuation of age-related decline in hippocampal GR translocation into the nucleus after stress loading, which may contribute to an improvement of mental health.


Subject(s)
Aging/drug effects , Arachidonic Acid/pharmacology , Hypothalamo-Hypophyseal System/drug effects , Pituitary-Adrenal System/drug effects , Stress, Physiological/drug effects , Aging/metabolism , Animals , Arachidonic Acid/administration & dosage , Dietary Supplements , Hypothalamo-Hypophyseal System/metabolism , Mice , Pituitary-Adrenal System/metabolism
11.
Pharmazie ; 75(1): 23-26, 2020 01 02.
Article in English | MEDLINE | ID: mdl-32033629

ABSTRACT

Early administration of antiplatelet agents in acute ischemic stroke patients (AIS) receiving intravenous thrombolysis (IVT) is a potential therapeutic strategy, however, safety and efficacy are not well established. We hypothesize that antiplatelet pretreatment (AP) before IVT have a similarly role as initiation of AP within the first 24 hours following IVT. We aimed to explore the effect of AP on platelet aggregation and clinical outcomes in thrombolysis-treated AIS patients. We enrolled AIS patients treated with IVT at the Neurology Department of the Nanjing First Hospital from January 2016 to June 2018. Prior use of antiplatelet agent was recorded. Light transmittance aggregometry was used to estimate the maximum platelet aggregation (MPA). Linear regression model was performed to investigate the factors associated with MPA. Multivariate logistic regression was used to analyse the association between AP and clinical outcomes. A total of 59 patients were included; 23 (38.9 %) were taking antiplatelet agent before stroke. Prior AP (ß = -20.209, SE mean=6.574; P=0.004) was significantly lower the arachidonic acid-induced MPA at the time point of 3h after thrombolysis. AP did not increase of the risk for sICH (OR=3.41, 95%CI 0.16-7.20, p=0.436) or mortality (OR=3.55, 95%CI 0.39-8.52, p=0.260). There were no associations between AP and improved clinical outcomes (all P>0.05). In thrombolysis-treated AIS patients, AP was associated with lower MPA after thrombolysis. AP is safe in these patients, however, further studies are required to confirm the efficacy.


Subject(s)
Fibrinolytic Agents/administration & dosage , Ischemic Stroke/drug therapy , Platelet Aggregation Inhibitors/administration & dosage , Tissue Plasminogen Activator/administration & dosage , Administration, Intravenous , Aged , Aged, 80 and over , Arachidonic Acid/administration & dosage , Arachidonic Acid/pharmacology , Cohort Studies , Female , Fibrinolytic Agents/therapeutic use , Humans , Ischemic Stroke/physiopathology , Male , Middle Aged , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/adverse effects , Platelet Function Tests , Prospective Studies , Time Factors , Tissue Plasminogen Activator/therapeutic use , Treatment Outcome
12.
Nutrients ; 12(1)2020 Jan 01.
Article in English | MEDLINE | ID: mdl-31906339

ABSTRACT

After birth, preterm infants are deficient in arachidonic acid (ARA), docosahexaenoic acid (DHA), and antioxidants, increasing their risk of oxidative stress-related pathologies. The principal aim was to evaluate if supplementation with long-chain polyunsaturated fatty acids (LCPUFAs) improves antioxidant defenses. In total, 21 preterm infants were supplemented with ARA and DHA in a 2:1 ratio (ARA:DHA-S) or with medium-chain triglycerides (MCT-S). Plasma n-3 and n-6 LCPUFAs were measured at birth, postnatal day 28, and 36 weeks of postmenstrual age (36 WPA) by gas chromatography-mass spectroscopy. Plasma antioxidants (glutathione (GSH), catalase, and thiols) and oxidative damage biomarkers (malondialdehyde (MDA), carbonyls) were analyzed at the same time points by spectrophotometry, and scores of antioxidant status (Antiox-S) and oxidative damage (Proxy-S) were calculated. At 36 WPA, linoleic acid (LA) and dihomo--linolenic acid (DGLA) were decreased in ARA:DHA-S compared to the MCT-S group (LA: ARA:DHA-S = 18.54 1.68, MCT-S = 22.80 1.41; p = 0.018; DGLA: ARA:DHA-S = 1.68 0.38, MCT-S = 2.32 0.58; p = 0.018). Furthermore, α-linolenic acid (ALA) was increased in ARA:DHA-S (ARA:DHA-S = 0.52 0.33, MCT-S = 0.22 0.10; p = 0.018). Additionally, LA:DHA ratio was decreased in the ARA:DHA-S compared to control group (ARA:DHA-S = 6.26 2.35, MCT-S = 8.21 2.65; p = 0.045). By the end of supplementation (36 WPA), catalase, thiol groups, and Antiox-S were significantly higher in neonates receiving ARA:DHA-S compared to those receiving MCT-S, with no differences in oxidative stress biomarkers. In conclusion, ARA:DHA supplementation in preterm neonates resulted in an overall improvement in antioxidant to oxidant balance and a decrease in early fatty acid precursors of the n-6 relative to the n-3 pathway. These effects may reduce oxidative stress and inflammation.


Subject(s)
Antioxidants/metabolism , Dietary Supplements , Fatty Acids, Unsaturated/administration & dosage , Infant, Premature/blood , Oxidative Stress/physiology , Arachidonic Acid/administration & dosage , Docosahexaenoic Acids/administration & dosage , Female , Humans , Infant, Newborn , Male , Pilot Projects , Triglycerides/administration & dosage , alpha-Linolenic Acid/administration & dosage
13.
J Exp Biol ; 223(Pt 3)2020 02 05.
Article in English | MEDLINE | ID: mdl-31953363

ABSTRACT

The present study investigated the effect of arachidonic acid (AA) and selected prostaglandins on the regulation of vitellogenesis, ecdysteroidogenesis and methyl farnesoate (MF) synthesis in the freshwater crab Oziotelphusa senex senex and the giant mud crab, Scylla serrata Administration of AA and prostaglandin F2α (PGF2α) and prostaglandin E2 (PGE2) significantly increased ovarian index, oocyte diameter and ovarian vitellogenin levels and ecdysteroid and MF levels in the hemolymph of crabs. Secretions of MF and ecdysteroids from in vitro cultured mandibular organs (MO) and Y-organs (YO) isolated from intermolt crabs injected with AA, PGF2α and PGE2 were greater when compared with controls. In contrast, injection of prostaglandin D2 (PGD2) had no effect on vitellogenesis, ecdysteroid and MF levels in circulation. In vitro secretion of MF from MO explants isolated from avitellogenic crabs incubated with AA, PGF2α and PGE2 increased in a time-dependent manner. Conversely, incubation of YOs isolated from avitellogenic crabs with AA, PGF2α and PGE2 had no effect on secretion of ecdsyteroids. These results implicate prostaglandins in the regulation of reproduction by inducing the synthesis of MF and consequent ecdysteroid synthesis in brachyuran crabs, and provide an alternative molecular intervention mechanism to the traditional eyestalk ablation methodology to induce vitellogenesis and ovarian maturation in crustaceans.


Subject(s)
Arachidonic Acid/administration & dosage , Brachyura/physiology , Ecdysteroids/metabolism , Fatty Acids, Unsaturated/metabolism , Prostaglandins/administration & dosage , Vitellogenesis/physiology , Animals , Brachyura/drug effects , Female , Species Specificity , Vitellogenesis/drug effects
14.
Nutr Neurosci ; 23(12): 931-945, 2020 Dec.
Article in English | MEDLINE | ID: mdl-30806182

ABSTRACT

During the development of the central nervous system, oligodendrocytes (OLs) are responsible for myelination, the formation of the myelin sheath around axons. This process enhances neuronal connectivity and supports the maturation of emerging cognitive functions. In humans, recent evidence suggests that early life nutrition may affect myelination. In the present study, we investigated the impact of a blend containing docosahexaenoic acid, arachidonic acid, vitamin B12, vitamin B9, iron and sphingomyelin, or each of these nutrients individually, on oligodendrocyte precursor cells (OPCs) proliferation and maturation into OLs as well as their myelinating properties. By using an in vitro model, developed to study each step of myelination, we found that the nutrient blend increased the number of OPCs and promoted their differentiation and maturation into OLs, as measured by quantifying A2B5 positive cells, myelin-associated glycoprotein (MAG) positive cells and area, myelin binding protein (MBP) positive cells and area, respectively. Moreover, measuring myelination by quantifying the overlapping signal between neurofilament and either MAG or MBP revealed a positive effect of the blend on OLs myelinating properties. In contrast, treatment with each individual nutrient resulted in differential effects on the various readouts. This work suggests that dietary intake of these nutrients during early life, might be beneficial for myelination.


Subject(s)
Arachidonic Acid/administration & dosage , Docosahexaenoic Acids/administration & dosage , Folic Acid/administration & dosage , Iron/administration & dosage , Myelin Sheath/drug effects , Neurons/drug effects , Sphingomyelins/administration & dosage , Vitamin B 12/administration & dosage , Animals , Cells, Cultured , Myelin Sheath/physiology , Neurons/physiology , Oligodendrocyte Precursor Cells/drug effects , Oligodendrocyte Precursor Cells/physiology , Rats, Wistar
15.
Nutr Diet ; 77(1): 131-138, 2020 02.
Article in English | MEDLINE | ID: mdl-30338904

ABSTRACT

AIM: To validate the polyunsaturated food frequency questionnaire (PUFA FFQ) and test for reproducibility in people with end stage renal disease on dialysis treatment. METHODS: Participants (n = 32) completed the PUFA FFQ and three 24-hour recalls. Erythrocyte samples (n = 29) were used for erythrocyte fatty acid analysis. The triangular relationship between the PUFA FFQ, 24-hour recalls and the biomarker was assessed using the method of triads. Agreement between the two dietary methods was also assessed using Bland-Altman plots and classification by quintiles. Reproducibility was tested on a subset of the group (n = 8). RESULTS: The PUFA FFQ was a valid measure of all PUFA except for docosapentaenoic acid (DPA) and arachidonic acid (AA). Strong validity coefficients were found for n-3 long-chain PUFA (LCPUFA), eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) of 0.914 (95% CI: 0.665, 0.997) and 0.889 (95% CI: 0.706, 0.994), respectively. In the Bland-Altman plots 91-100% of observations fell between the limits of agreement for all PUFA. There were significant correlations between the initial FFQ and the repeat FFQ for all PUFA except DPA and AA. CONCLUSIONS: The PUFA FFQ is a valid tool for assessing PUFA intake in people with end stage renal disease.


Subject(s)
Fatty Acids, Unsaturated/blood , Kidney Failure, Chronic/blood , Renal Dialysis , Surveys and Questionnaires , Aged , Aged, 80 and over , Arachidonic Acid/administration & dosage , Arachidonic Acid/blood , Body Mass Index , Cohort Studies , Diet , Diet Records , Eicosapentaenoic Acid/administration & dosage , Eicosapentaenoic Acid/blood , Erythrocytes/metabolism , Fatty Acids, Omega-3/administration & dosage , Fatty Acids, Omega-3/blood , Fatty Acids, Unsaturated/administration & dosage , Female , Humans , Kidney Failure, Chronic/drug therapy , Male , Mental Recall , Middle Aged , Reproducibility of Results
16.
Article in English | MEDLINE | ID: mdl-31672574

ABSTRACT

A high consumption of polyunsaturated fatty acids (PUFAs), particularly n-3 PUFAs, is atheroprotective. PUFAs incorporation into membrane phospholipids alters the functionality of membrane proteins. We studied the consequences of the in vitro supplementation of several PUFAs on the FA profiles and on ABCA1-dependent cholesterol efflux capacities from cholesterol-loaded macrophages. Arachidonic acid (AA, C20:4 n-6) and, to a lesser extent, eicosapentaenoic acid (EPA, C20:5 n-3), dose-dependently impaired cholesterol efflux from cholesterol-loaded J774 mouse macrophages without alterations in ABCA1 expression, whereas docosahexaenoic acid (DHA, C22:6 n-3) had no impact. AA cells exhibited higher proportions of arachidonic acid and adrenic acid (C22:4 n-6), its elongation product. EPA cells exhibited slightly higher proportions of EPA associated with much higher proportions of docosapentaenoic acid (C22:5 n-3), its elongation product and with lower proportions of AA. Conversely, both EPA and DHA and, to a lesser extent, AA decreased cholesterol efflux from cholesterol-loaded primary human macrophages (HMDM). The differences observed in FA profiles after PUFA supplementations were different from those observed for the J774 cells. In conclusion, we are the first to report that AA and EPA, but not DHA, have deleterious effects on the cardioprotective ABCA1 cholesterol efflux pathway from J774 foam cells. Moreover, the membrane incorporation of PUFAs does not have the same impact on cholesterol efflux from murine (J774) or human (HMDM) cholesterol-loaded macrophages. This finding emphasizes the key role of the cellular model in cholesterol efflux studies and may partly explain the heterogeneous literature data on the impact of PUFAs on cholesterol efflux.


Subject(s)
Arachidonic Acid/administration & dosage , Cell Membrane/drug effects , Cholesterol/metabolism , Docosahexaenoic Acids/administration & dosage , Eicosapentaenoic Acid/administration & dosage , Foam Cells/drug effects , ATP Binding Cassette Transporter 1/metabolism , Animals , Atherosclerosis/etiology , Atherosclerosis/prevention & control , Cell Line, Tumor , Cell Membrane/metabolism , Cholesterol/administration & dosage , Cholesterol/adverse effects , Dietary Supplements , Foam Cells/cytology , Foam Cells/metabolism , Healthy Volunteers , Humans , Mice , Phospholipids/metabolism , Primary Cell Culture
17.
J Nutr Biochem ; 73: 108226, 2019 11.
Article in English | MEDLINE | ID: mdl-31520815

ABSTRACT

Long-chain polyunsaturated fatty acids modulate bone mass and adipocyte metabolism. Arachidonic acid (AA, C20:4 n-6) is elevated in obesity and postulated to stimulate bone resorption. This study aimed to determine the effect of AA on bone mass, quality, and adiposity in diet-induced obesity during growth. Male Sprague-Dawley rats (n=42, 4-week) were randomized into groups fed a control diet (CTRL, AIN-93G), high-fat diet (HFD, 35% kcal fat) or HFD + AA (1% w/w diet) for 6 weeks. Body composition, bone mineral density and microarchitecture were measured using dual-energy X-ray absorptiometry and micro-computed tomography. Red blood cell fatty acid profile was measured with gas chromatography. Group differences were evaluated using repeated measures two-way analysis of variance with Tukey-Kramer post hoc testing. Total energy intake did not differ among diet groups. At week 6, HFD + AA had significantly greater body fat % (12%), body weight (6%) and serum leptin concentrations (125%) than CTRL, whereas visceral fat (mass and %, assessed with micro-computed tomography) was increased in both HFD and HFD + AA groups. HFD + AA showed reduced whole body bone mineral content and femur mid-diaphyseal cortical bone cross-sectional area than HFD and CTRL, without impairment in bone strength. Contrarily, HFD + AA had greater femur metaphyseal trabecular vBMD (35%) and bone volume fraction (5%) compared to controls. Inclusion of AA elevated leptin concentrations in male rats. The early manifestations of diet-induced obesity on bone mass were accelerated with AA. Studies of longer duration are needed to clarify the effect of AA on peak bone mass following growth cessation.


Subject(s)
Arachidonic Acid/adverse effects , Bone Density/drug effects , Bone and Bones/physiopathology , Diet, High-Fat/adverse effects , Obesity/etiology , 3T3-L1 Cells , Adipocytes/drug effects , Adiposity/drug effects , Animals , Arachidonic Acid/administration & dosage , Biomechanical Phenomena , Body Composition/drug effects , Bone Resorption/etiology , Bone and Bones/drug effects , Energy Intake , Fatty Acids/pharmacology , Femur/drug effects , Femur/pathology , Leptin/blood , Male , Mice , Obesity/pathology , Obesity/physiopathology , Rats , Rats, Sprague-Dawley
18.
Nutrients ; 11(9)2019 Sep 09.
Article in English | MEDLINE | ID: mdl-31505819

ABSTRACT

Vitamin D and omega 3 fatty acid (ω-3) co-supplementation potentially improves type 1 diabetes (T1D) by attenuating autoimmunity and counteracting inflammation. This cohort study, preliminary to a randomized control trial (RCT), is aimed at evaluating, in a series of T1D children assuming Mediterranean diet and an intake of cholecalciferol of 1000U/day from T1D onset, if ω-3 co-supplementation preserves the residual endogen insulin secretion (REIS). Therefore, the cohort of 22 "new onsets" of 2017 received ω-3 (eicosapentenoic acid (EPA) plus docosahexaenoic acid (DHA), 60 mg/kg/day), and were compared retrospectively vs. the 37 "previous onsets" without ω-3 supplementation. Glicosilated hemoglobin (HbA1c%), the daily insulin demand (IU/Kg/day) and IDAA1c, a composite index (calculated as IU/Kg/day × 4 + HbA1c%), as surrogates of REIS, were evaluated at recruitment (T0) and 12 months later (T12). In the ω-3 supplemented group, dietary intakes were evaluated at T0 and T12. As an outcome, a decreased insulin demand (p < 0.01), particularly as pre-meal boluses (p < 0.01), and IDAA1c (p < 0.05), were found in the ω-3 supplemented group, while HbA1c% was not significantly different. Diet analysis in the ω-3 supplemented group, at T12 vs. T0, highlighted that the intake of arachidonic acid (AA) decreased (p < 0.01). At T0, the AA intake was inversely correlated with HbA1c% (p < 0.05; r;. 0.411). In conclusion, the results suggest that vitamin D plus ω-3 co-supplementation as well as AA reduction in the Mediterranean diet display benefits for T1D children at onset and deserve further investigation.


Subject(s)
Diabetes Mellitus, Type 1/therapy , Diet, Mediterranean , Dietary Supplements , Fatty Acids, Omega-3/administration & dosage , Vitamin D/administration & dosage , Arachidonic Acid/administration & dosage , Child , Cholecalciferol/administration & dosage , Diabetes Mellitus, Type 1/blood , Female , Glycated Hemoglobin/analysis , Humans , Insulin/therapeutic use , Insulin Secretion/drug effects , Male , Randomized Controlled Trials as Topic , Retrospective Studies
19.
Br J Nutr ; 122(3): 252-261, 2019 08 14.
Article in English | MEDLINE | ID: mdl-31405389

ABSTRACT

Oxylipins are bioactive lipid mediators synthesised from PUFA. The most well-known oxylipins are the eicosanoids derived from arachidonic acid (ARA), and many of them influence cardiac physiology in health and disease. Oxylipins are also formed from other n-3 and n-6 PUFA such as α-linolenic acid (ALA), EPA, DHA and linoleic acid (LA), but fundamental data on the heart oxylipin profile, and the effect of diet and sex on this profile, are lacking. Therefore, weanling female and male Sprague-Dawley rats were given American Institute of Nutrition (AIN)-93G-based diets modified in oil composition to provide higher levels of ALA, EPA, DHA, LA and LA + ALA, compared with control diets. After 6 weeks, free oxylipins in rat hearts were increased primarily by their precursor PUFA, except for EPA oxylipins, which were increased not only by dietary EPA but also by dietary ALA or DHA. Dietary DHA had a greater effect than ALA or EPA on reducing ARA oxylipins. An exception to the dietary n-3 PUFA-lowering effects on ARA oxylipins was observed for several ARA-derived PG metabolites that were higher in rats given EPA diets. Higher dietary LA increased LA oxylipins, but it had no effect on ARA oxylipins. Overall, heart oxylipins were higher in female rats, but this depended on dietary treatment: the female oxylipin:male oxylipin ratio was higher in rats provided the ALA compared with the DHA diet, with other diet groups having ratios in between. In conclusion, individual PUFA and sex have unique and interactive effects on the rat heart free oxylipin profile.


Subject(s)
Animal Feed , Fatty Acids, Omega-3/administration & dosage , Fatty Acids, Omega-6/administration & dosage , Myocardium/metabolism , Oxylipins/metabolism , Animals , Arachidonic Acid/administration & dosage , Docosahexaenoic Acids/administration & dosage , Eicosapentaenoic Acid/administration & dosage , Female , Heart/physiology , Linoleic Acid/administration & dosage , Lipid Metabolism , Male , Rats , Rats, Sprague-Dawley , Sex Factors , alpha-Linolenic Acid/administration & dosage
20.
Nutrients ; 11(7)2019 Jul 06.
Article in English | MEDLINE | ID: mdl-31284588

ABSTRACT

Evidence suggests a role of long chain polyunsaturated fatty acids (LC-PUFA), in which animal foods are especially rich, in optimal neural development. The LC-PUFAs docosahexaenoic acid (DHA) and arachidonic acid, found in high concentrations in the brain and retina, have potential beneficial effects on cognition, and motor and visual functions. Phenylketonuria (PKU) is the most common inborn error of amino acid metabolism. The treatment of PKU consists of a phenylalanine-free diet, which limits the intake of natural proteins of high biological value. In this systematic review, we summarize the available evidence supporting a role for LC-PUFA supplementation as an effective means of increasing LC-PUFA levels and improving visual and neurocognitive functions in PKU patients. Data from controlled trials of children and adults (up to 47 years of age) were obtained by searching the MEDLINE and SCOPUS databases following Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. For each selected study, the risk of bias was assessed applying the methodology of the Cochrane Collaboration. The findings indicate that DHA supplementation in PKU patients from 2 weeks to 47 years of age improves DHA status and decreases visual evoked potential P100 wave latency in PKU children from 1 to 11 years old. Neurocognitive data are inconclusive.


Subject(s)
Arachidonic Acid/administration & dosage , Dietary Supplements , Docosahexaenoic Acids/administration & dosage , Phenylketonurias/diet therapy , Adolescent , Adult , Arachidonic Acid/adverse effects , Child , Child, Preschool , Cognition , Diet, Protein-Restricted , Dietary Supplements/adverse effects , Docosahexaenoic Acids/adverse effects , Evoked Potentials, Motor , Evoked Potentials, Visual , Female , Humans , Infant , Infant, Newborn , Male , Middle Aged , Phenylketonurias/diagnosis , Phenylketonurias/physiopathology , Phenylketonurias/psychology , Randomized Controlled Trials as Topic , Treatment Outcome , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...