Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 199
Filter
1.
Food Funct ; 12(14): 6588-6602, 2021 Jul 21.
Article in English | MEDLINE | ID: mdl-34100498

ABSTRACT

The natural structure of whole food plays an important role in the physiological impact of bioactive compounds present within the food, also known as the "matrix effect". Long-chain omega-3 polyunsaturated fatty acids (LCn-3PUFAs) are one example of a food-derived nutrient, mostly found in fish, that is believed to be influenced by the food matrix. However, most previous studies have compared only the long-term bioavailability of fish versus fish oil and have used commercial sources of fish oil. The present study aimed to investigate whether fish (salmon) matrix influences the transit of LCn-3PUFAs during in vitro digestion and affects bioavailability in healthy females. Meals containing intact salmon (intact structure), minced salmon (some structure) and defatted salmon + oil (no structure) with identical macronutrient compositions were developed. Healthy female participants (n = 13) consumed the meals in a postprandial crossover study and blood was collected at regular time points for 6 h post meal consumption. In parallel, in vitro digestion of the meals was performed using a human gastric simulator (HGS) and digesta samples were collected at regular time points for 6 h. Results: showed that plasma concentration of eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) were significantly higher after participants consumed intact salmon compared to the other meals (covariate analysis p < 0.001). The in vitro digestion results showed defatted salmon + oil meal had a faster decrease in pH and faster fat emptying from the HGS than the other two meals. The defatted salmon + oil meal more closely followed fat emptying of a homogeneous unstructured meal, whereas the other meals exhibited phase separation with a delay in fat emptying. Conclusion: The fish matrix (salmon) plays an important role in the bioaccessibility and bioavailability of EPA and DHA. The differences are partly explained by fat digestion and emptying from the stomach. This study suggests that the natural structure of fish has a functional effect on the absorption and bioavailability of fish oil.


Subject(s)
Digestion , Fatty Acids, Omega-3/pharmacokinetics , Meals , Salmon , Adult , Animals , Biological Availability , Cross-Over Studies , Docosahexaenoic Acids/blood , Docosahexaenoic Acids/pharmacokinetics , Eicosapentaenoic Acid/blood , Eicosapentaenoic Acid/pharmacokinetics , Female , Fish Oils/pharmacokinetics , Humans , Postprandial Period , Seafood , Young Adult
2.
Biochemistry ; 60(22): 1741-1754, 2021 06 08.
Article in English | MEDLINE | ID: mdl-34029049

ABSTRACT

In this paper, human platelet 12-lipoxygenase [h12-LOX (ALOX12)], human reticulocyte 15-lipoxygenase-1 [h15-LOX-1 (ALOX15)], and human epithelial 15-lipoxygenase-2 [h15-LOX-2 (ALOX15B)] were observed to react with docosahexaenoic acid (DHA) and produce 17S-hydroperoxy-4Z,7Z,10Z,13Z,15E,19Z-docosahexaenoic acid (17S-HpDHA). The kcat/KM values with DHA for h12-LOX, h15-LOX-1, and h15-LOX-2 were 12, 0.35, and 0.43 s-1 µM-1, respectively, which demonstrate h12-LOX as the most efficient of the three. These values are comparable to their counterpart kcat/KM values with arachidonic acid (AA), 14, 0.98, and 0.24 s-1 µM-1, respectively. Comparison of their product profiles with DHA demonstrates that the three LOX isozymes produce 11S-HpDHA, 14S-HpDHA, and 17S-HpDHA, to varying degrees, with 17S-HpDHA being the majority product only for the 15-LOX isozymes. The effective kcat/KM values (kcat/KM × percent product formation) for 17S-HpDHA of the three isozymes indicate that the in vitro value of h12-LOX was 2.8-fold greater than that of h15-LOX-1 and 1.3-fold greater than that of h15-LOX-2. 17S-HpDHA was an effective substrate for h12-LOX and h15-LOX-1, with four products being observed under reducing conditions: protectin DX (PDX), 16S,17S-epoxy-4Z,7Z,10Z,12E,14E,19Z-docosahexaenoic acid (16S,17S-epoxyDHA), the key intermediate in neuroprotection D1 biosynthesis [NPD1, also known as protectin D1 (PD1)], 11,17S-diHDHA, and 16,17S-diHDHA. However, h15-LOX-2 did not react with 17-HpDHA. With respect to their effective kcat/KM values, h12-LOX was markedly less effective than h15-LOX-1 in reacting with 17S-HpDHA, with a 55-fold lower effective kcat/KM in producing 16S,17S-epoxyDHA and a 27-fold lower effective kcat/KM in generating PDX. This is the first direct demonstration of h15-LOX-1 catalyzing this reaction and reveals an in vitro pathway for PDX and NPD1 intermediate biosynthesis. In addition, epoxide formation from 17S-HpDHA and h15-LOX-1 was negatively affected via allosteric regulation by 17S-HpDHA (Kd = 5.9 µM), 12S-hydroxy-5Z,8Z,10E,14Z-eicosatetraenoic acid (12S-HETE) (Kd = 2.5 µM), and 17S-hydroxy-13Z,15E,19Z-docosatrienoic acid (17S-HDTA) (Kd = 1.4 µM), suggesting a possible regulatory pathway in reducing epoxide formation. Finally, 17S-HpDHA and PDX inhibited platelet aggregation, with EC50 values of approximately 1 and 3 µM, respectively. The in vitro results presented here may help advise in vivo PDX and NPD1 intermediate (i.e., 16S,17S-epoxyDHA) biosynthetic investigations and support the benefits of DHA rich diets.


Subject(s)
Docosahexaenoic Acids/metabolism , Lipoxygenases/metabolism , Allosteric Regulation , Arachidonate 12-Lipoxygenase/metabolism , Arachidonate 15-Lipoxygenase/metabolism , Arachidonic Acid/metabolism , Arachidonic Acids/metabolism , Biosynthetic Pathways , Blood Platelets/metabolism , Docosahexaenoic Acids/pharmacokinetics , Docosahexaenoic Acids/pharmacology , Humans , Lipoxygenase/metabolism , Lipoxygenases/biosynthesis
3.
Article in English | MEDLINE | ID: mdl-33360684

ABSTRACT

INTRODUCTION: Supplements with eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) are generally oil-based formulations containing their triacylglycerols, phospholipids or ethyl-esters (EE). Recently, a l-lysine salt of carboxylic EPA and DHA became available (Lys-FFA), which necessitated to study its oral absorption and plasma kinetics in humans. OBJECTIVES: The in vitro dissolution characteristics, oral bioavailability and 48 h plasma profiles of EPA and DHA (as triacylglycerides) of Lys-FFA, relative to a commercially available oil-based EE supplement. METHODS: Dissociation of the lysine from the FFAs was studied in vitro applying simulated gastric (12 h) and intestinal (3 h) conditions. In an open label, randomized, two-way cross-over design, oral administration of Lys-FFA (500 mg EPA plus 302 mg DHA) versus EE (504 mg EPA plus 378 mg DHA) was studied over 48 h, in eight female volunteers. Plasma profiles of EPA and DHA were described by Area Under the Curve (AUC; 0-12 h), Cmax and Tmax. RESULTS: Dissolution studies with Lys-FFA showed complete dissociation under both conditions. In volunteers Lys-FFA showed rapid absorption and high bioavailability indicated by significant differences in both the AUC0-12hr and Cmax when compared to the EE comparator (p<0.001), with AUC0-12hr which was for EPA 5 times higher with Lys-FFA than with the EE formulation. CONCLUSION: This first-in-man study of Lys-FFA demonstrated rapid absorption of EPA and DHA and a considerably higher bioavailability compared to an EE supplement under fasting conditions. The release and absorption characteristics from this solid form offer several new options in terms of formulation technology and dosing.


Subject(s)
Docosahexaenoic Acids , Eicosapentaenoic Acid/analogs & derivatives , Lysine , Biological Availability , Cross-Over Studies , Docosahexaenoic Acids/administration & dosage , Docosahexaenoic Acids/pharmacokinetics , Eicosapentaenoic Acid/administration & dosage , Eicosapentaenoic Acid/pharmacokinetics , Female , Humans , Lysine/administration & dosage , Lysine/pharmacokinetics
4.
J Neurosci Res ; 99(2): 561-572, 2021 02.
Article in English | MEDLINE | ID: mdl-32964457

ABSTRACT

Stroke severely impairs quality of life and has a high mortality rate. On the other hand, dietary docosahexaenoic acid (DHA) prevents neuronal damage. In this review, we describe the effects of dietary DHA on ischemic stroke-associated neuronal damage and its role in stroke prevention. Recent epidemiological studies have been conducted to analyze stroke prevention through DHA intake. The effects of dietary intake and supply of DHA to neuronal cells, DHA-mediated inhibition of neuronal damage, and its mechanism, including the effects of the DHA metabolite, neuroprotectin D1 (NPD1), were investigated. These studies revealed that DHA intake was associated with a reduced risk of stroke. Moreover, studies have shown that DHA intake may reduce stroke mortality rates. DHA, which is abundant in fish oil, passes through the blood-brain barrier to accumulate as a constituent of phospholipids in the cell membranes of neuronal cells and astrocytes. Astrocytes supply DHA to neuronal cells, and neuronal DHA, in turn, activates Akt and Raf-1 to prevent neuronal death or damage. Therefore, DHA indirectly prevents neuronal damage. Furthermore, NDP1 blocks neuronal apoptosis. DHA, together with NPD1, may block neuronal damage and prevent stroke. The inhibitory effect on neuronal damage is achieved through the antioxidant (via inducing the Nrf2/HO-1 system) and anti-inflammatory effects (via promoting JNK/AP-1 signaling) of DHA.


Subject(s)
Brain Damage, Chronic/prevention & control , Docosahexaenoic Acids/therapeutic use , Ischemic Stroke/diet therapy , Nerve Degeneration/prevention & control , Stroke/prevention & control , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/pharmacokinetics , Anti-Inflammatory Agents/therapeutic use , Antioxidants/administration & dosage , Antioxidants/pharmacokinetics , Antioxidants/therapeutic use , Apoptosis/drug effects , Biological Availability , Biological Transport , Blood-Brain Barrier , Brain Damage, Chronic/etiology , Dietary Fats/administration & dosage , Dietary Fats/pharmacokinetics , Dietary Fats/therapeutic use , Docosahexaenoic Acids/administration & dosage , Docosahexaenoic Acids/metabolism , Docosahexaenoic Acids/pharmacokinetics , Docosahexaenoic Acids/pharmacology , Fatty Acid-Binding Proteins/physiology , Fish Oils/administration & dosage , Fish Oils/pharmacokinetics , Humans , Incidence , Ischemic Stroke/complications , Ischemic Stroke/epidemiology , Membrane Lipids/metabolism , Mice , Neoplasm Proteins/physiology , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Plant Oils/administration & dosage , Plant Oils/pharmacokinetics , Signal Transduction/drug effects , Symporters/deficiency , Symporters/physiology , alpha-Linolenic Acid/pharmacokinetics
5.
Eur J Pharm Biopharm ; 158: 273-283, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33242579

ABSTRACT

Hepatic-arterial infusion (HAI) of low-density lipoprotein (LDL) nanoparticles reconstituted with docosahexaenoic acid (DHA) (LDL-DHA) has been shown in a rat hepatoma model to be a promising treatment for hepatocellular carcinoma. To date, little is known regarding the safety of HAI of LDL-DHA to the liver. Therefore, we aimed to investigate the deposition, metabolism and safety of HAI of LDL-DHA (2, 4 or 8 mg/kg) in the rat. Following HAI, fluorescent labeled LDL nanoparticles displayed a biexponential plasma concentration time curve as the particles were rapidly extracted by the liver. Overall, increasing doses of HAI of LDL-DHA was well tolerated in the rat. Body weight, plasma biochemistry and histology were all unremarkable and molecular markers of inflammation did not increase with treatment. Lipidomics analyses showed that LDL-DHA was preferentially oxidized to the anti-inflammatory mediator, protectin DX. We conclude that HAI of LDL-DHA nanoparticles is not only safe, but provides potential hepatoprotective benefits.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Docosahexaenoic Acids/administration & dosage , Drug Carriers/chemistry , Liver Neoplasms, Experimental/drug therapy , Liver Neoplasms/drug therapy , Animals , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Docosahexaenoic Acids/adverse effects , Docosahexaenoic Acids/pharmacokinetics , Dose-Response Relationship, Drug , Drug Carriers/adverse effects , Humans , Infusions, Intra-Arterial , Lipoproteins, LDL/adverse effects , Lipoproteins, LDL/chemistry , Liver/blood supply , Liver/pathology , Liver Neoplasms/pathology , Liver Neoplasms, Experimental/pathology , Male , Nanoparticles/chemistry , Rats , Tissue Distribution
6.
Clin Transl Sci ; 14(2): 683-691, 2021 03.
Article in English | MEDLINE | ID: mdl-33202089

ABSTRACT

Sjögren's syndrome (SS) is an autoimmune disease with no effective treatment options. Resolvin D1 (RvD1) belongs to a class of lipid-based specialized pro-resolving mediators that showed efficacy in preclinical models of SS. We developed a physiologically-based pharmacokinetic (PBPK) model of RvD1 in mice and optimized the model using plasma and salivary gland pharmacokinetic (PK) studies performed in NOD/ShiLtJ mice with SS-like features. The predictive performance of the PBPK model was also evaluated with two external datasets from the literature reporting RvD1 PKs. The PBPK model adequately captured the observed concentrations of RvD1 administered at different doses and in different species. The PKs of RvD1 in virtual humans were predicted using the verified PBPK model at various doses (0.01-10 mg/kg). The first-in-human predictions of RvD1 will be useful for the clinical trial design and translation of RvD1 as an effective treatment strategy for SS.


Subject(s)
Docosahexaenoic Acids/pharmacokinetics , Models, Biological , Animals , Datasets as Topic , Docosahexaenoic Acids/administration & dosage , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Female , Humans , Male , Mice , Models, Animal , Salivary Glands/metabolism , Sjogren's Syndrome/drug therapy , Tissue Distribution
7.
Article in English | MEDLINE | ID: mdl-33221700

ABSTRACT

Pre-clinical evidence suggests that omega-3 (n-3) polyunsaturated fatty acids (PUFAs), in particular, docosahexaenoic acid (DHA) have been shown to affect testosterone synthesis in males. This study is a secondary analysis of a randomized controlled trial which determined the effect of a DHA-enriched fish oil supplement on insulin resistance. The aim of the current study was to determine whether testosterone levels change in response to a DHA-enriched fish oil intervention. Overweight and obese men and women without diabetes were recruited to the study. Participants were stratified by sex and randomly allocated to intervention (860 mg DHA + 120 g EPA/day; FO) or an isocaloric control (corn oil; CO) for 12 weeks. A fasted blood sample was collected pre- and post-intervention. Fatty acid composition of erythrocyte membranes was measured using gas chromatography. Total testosterone and metabolic parameters were measured by an accredited commercial pathology laboratory. Sixty-one participants (CO/FO: n = 29/32) were included in the current analysis (male: n = 22, 36.07%).  DHA-enriched fish oil supplementation increased total testosterone levels in males after adjusting for baseline levels, age and BMI. There was no treatment effect in females. Changes in testosterone levels in males were positively associated with changes to omega-3 PUFAs EPA and DHA and inversely correlated with omega-6 PUFA, arachidonic acid and dihomo-gamma-linolenic acid content in erythrocyte membranes, and was associated with beneficial changes to fasting insulin and HOMA-IR across the course of the study. DHA-enriched fish oil supplementation increases testosterone levels in overweight and obese men. Further research is warranted to substantiate these findings with a larger sample size and a longer follow-up period.


Subject(s)
Dietary Supplements , Docosahexaenoic Acids/administration & dosage , Obesity/blood , Obesity/diet therapy , Testosterone/blood , Adolescent , Adult , Aged , Docosahexaenoic Acids/pharmacokinetics , Double-Blind Method , Humans , Male , Middle Aged
8.
Article in English | MEDLINE | ID: mdl-33038830

ABSTRACT

We recently reported that acute injection of docosahexaenoic acid (DHA) triglyceride emulsions (tri-DHA) conferred neuroprotection after hypoxic-ischemic (HI) injury in a neonatal mouse stroke model. We showed that exogenous DHA increased concentrations of DHA in brain mitochondria as well as DHA-derived specialized pro-resolving mediator (SPM) levels in the brain. The objective of the present study was to investigate the distribution of emulsion particles and changes in plasma lipid profiles after tri-DHA injection in naïve mice and in animals subjected to HI injury. We also examined whether tri-DHA injection would change DHA- and eicosapentaenoic acid (EPA)-derived SPM levels in the brain. To address this, neonatal (10-day-old) naïve and HI mice were injected with radiolabeled tri-DHA emulsion (0.375 g tri-DHA/kg bw), and blood clearance and tissue distribution were analyzed. Among all the organs assayed, the lowest uptake of emulsion particles was in the brain (<0.4% recovered dose) in both naïve and HI mice, while the liver had the highest uptake. Tri-DHA administration increased DHA concentrations in plasma lysophosphatidylcholine and non-esterified fatty acids. Additionally, treatment with tri-DHA after HI injury significantly elevated the levels of DHA-derived SPMs and monohydroxy-containing DHA-derived products in the brain. Further, tri-DHA administration increased resolvin E2 (RvE2, 5S,18R-dihydroxy-eicosa-6E,8Z,11Z,14Z,16E-pentaenoic acid) and monohydroxy-containing EPA-derived products in the brain. These results suggest that the transfer of DHA through plasma lipid pools plays an important role in DHA brain transport in neonatal mice subjected to HI injury. Furthermore, increases in EPA and EPA-derived SPMs following tri-DHA injection demonstrate interlinked metabolism of these two fatty acids. Hence, changes in both EPA and DHA profile patterns need to be considered when studying the protective effects of DHA after HI brain injury. Our results highlight the need for further investigation to differentiate the effects of DHA from EPA on neuroprotective pathways following HI damage. Such information could contribute to the development of specific DHA-EPA formulations to improve clinical endpoints and modulate potential biomarkers in ischemic brain injury.


Subject(s)
Brain Injuries , Brain/metabolism , Docosahexaenoic Acids , Eicosapentaenoic Acid/blood , Hypoxia-Ischemia, Brain , Triglycerides , Animals , Brain Injuries/drug therapy , Brain Injuries/metabolism , Brain Injuries/pathology , Docosahexaenoic Acids/pharmacokinetics , Docosahexaenoic Acids/pharmacology , Emulsions , Hypoxia-Ischemia, Brain/drug therapy , Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/pathology , Mice , Triglycerides/pharmacokinetics , Triglycerides/pharmacology
9.
Food Funct ; 11(10): 8637-8647, 2020 Oct 21.
Article in English | MEDLINE | ID: mdl-32936172

ABSTRACT

Fish oil-loaded hollow solid lipid micro- and nanoparticles were prepared by atomization of the CO2-expanded lipid mixture. The obtained particles were spherical and free-flowing with an average particle size of 6.9 µm. Fish oil loading efficiency was achieved at 92.3% (w/w). The in vitro digestive stability, lipid digestibility and EPA and DHA bioaccessibility of the fish oil-loaded particles were examined using an in vitro sequential digestion model. The mean particle diameter increased markedly after oral (15.2 µm) and gastric (32.4 µm) digestion and then decreased after the small intestinal stage (24.0 µm). Fish oil-loaded particles remained spherical and intact but mainly agglomerated on the top phase throughout the oral and gastric digestion. However, a mixed digesta was formed after the small intestinal digestion, which contained digested broken particle pieces, undigested fish oil-loaded particles, free fatty acids, monoacylglycerols and micelles. The extent of lipolysis was significantly increased for the 30% fish oil-loaded particles as compared to physical mixtures of empty hollow solid lipid particles or bulk FHSO and fish oil (p < 0.05). Moreover, EPA and DHA bioaccessibility was significantly improved from 9.7 to 18.2% with the 30% fish oil-loaded particles (p < 0.05).


Subject(s)
Digestion , Fish Oils , Nanoparticles , Biological Availability , Docosahexaenoic Acids/pharmacokinetics , Eicosapentaenoic Acid/pharmacokinetics , Fish Oils/chemistry , Fish Oils/pharmacokinetics , In Vitro Techniques , Lipolysis , Particle Size
10.
Clin Pharmacol Drug Dev ; 9(8): 985-994, 2020 11.
Article in English | MEDLINE | ID: mdl-32567203

ABSTRACT

In patients with coronary heart disease undergoing primary prevention, hypertriglyceridemia is a residual risk for cardiovascular events. Omega-3 carboxylic acid (OM3-CA), a mixture of the free fatty acid forms of eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), may be beneficial in reducing triglyceride levels. As part of the clinical development program of OM3-CA in China, this phase I study evaluated the pharmacokinetics, safety, and tolerability profile of OM3-CA in healthy subjects. The pharmacokinetic results of this study were also compared with those of available data for Western populations. Fourteen healthy Chinese subjects (aged 18-45 years) received once-daily oral OM3-CA 4 g for 14 consecutive days. Pharmacokinetic parameters were assessed from both baseline-uncorrected and baseline-corrected plasma concentrations vs time profile of EPA, DHA, and EPA plus DHA. Following single and multiple oral doses of OM3-CA, the absorption of EPA, DHA, and EPA plus DHA was steady with median tmax occurring at 5.5-6 hours after both single and multiple dosing. Close to steady-state concentrations in plasma were reached after 14 days of continuous once-daily dosing, and accumulation was confirmed for EPA, DHA, and EPA plus DHA. Of the 14 subjects treated with OM3-CA, 6 (42.9%) reported at least 1 adverse event (diarrhea) during the study, which was determined as mild and treatment emergent. No serious adverse events were reported. In summary, the pharmacokinetic profile of oral OM3-CA 4 g after single and multiple dosing in healthy Chinese subjects is consistent with that observed in other ethnic populations.


Subject(s)
Carboxylic Acids/pharmacokinetics , Docosahexaenoic Acids/pharmacokinetics , Eicosapentaenoic Acid/pharmacokinetics , Fatty Acids, Nonesterified/pharmacokinetics , Fatty Acids, Omega-3/pharmacokinetics , Healthy Volunteers/statistics & numerical data , Hypertriglyceridemia/drug therapy , Administration, Oral , Adult , Area Under Curve , Asian People/ethnology , Carboxylic Acids/administration & dosage , Carboxylic Acids/adverse effects , Carboxylic Acids/blood , Docosahexaenoic Acids/administration & dosage , Docosahexaenoic Acids/blood , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Tolerance , Eicosapentaenoic Acid/administration & dosage , Eicosapentaenoic Acid/blood , Fatty Acids, Nonesterified/administration & dosage , Fatty Acids, Nonesterified/blood , Fatty Acids, Omega-3/administration & dosage , Fatty Acids, Omega-3/adverse effects , Fatty Acids, Omega-3/blood , Female , Humans , Hypertriglyceridemia/prevention & control , Male , Middle Aged , Safety
11.
Toxicol Appl Pharmacol ; 400: 115037, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32417438

ABSTRACT

BACKGROUND: In recent years, small animal arterial port-catheter systems have been implemented in rodents with reasonable success. The aim of the current study is to employ the small animal port-catheter system to evaluate the safety of multiple hepatic-artery infusions (HAI) of low-density lipoprotein-docosahexaenoic acid (LDL-DHA) nanoparticles to the rat liver. METHODS: Wistar rats underwent surgical placement of indwelling HAI ports. Repeated administrations of PBS or LDL-DHA nanoparticles were performed through the port at baseline and days 3 and 6. Rats were sacrificed on day 9 at which point blood and various organs were collected for histopathology and biochemical analyses. RESULTS: The port-catheter systems were implanted successfully and repeated infusions of PBS or LDL-DHA nanoparticles were tolerated well by all animals over the duration of the study. Measurements of serum liver/renal function tests, glucose and lipid levels did not differ between control and LDL-DHA treated rats. The liver histology was unremarkable in the LDL-DHA treated rats and the expression of hepatic inflammatory regulators (NF-κß, IL-6 and CRP) were similar to control rats. Repeated infusions of LDL-DHA nanoparticles did not alter liver glutathione content or the lipid profile in the treated rats. The DHA extracted by the liver was preferentially metabolized to the anti-inflammatory DHA-derived mediator, protectin DX. CONCLUSION: Our findings indicate that repeated HAI of LDL-DHA nanoparticles is not only well tolerated and safe in the rat, but may also be protective to the liver.


Subject(s)
Catheters, Indwelling/adverse effects , Docosahexaenoic Acids/administration & dosage , Hepatic Artery , Infusions, Intra-Arterial/adverse effects , Lipoproteins, LDL/administration & dosage , Liver/metabolism , Nanoparticles/administration & dosage , Animals , Blood Glucose/analysis , Docosahexaenoic Acids/pharmacokinetics , Infusions, Intra-Arterial/methods , Kidney Function Tests , Lipids/blood , Lipoproteins, LDL/pharmacokinetics , Liver/blood supply , Liver Function Tests , Male , Rats, Wistar , Tissue Distribution
12.
Biomed Chromatogr ; 34(10): e4905, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32449539

ABSTRACT

A simple and specific LC-MS/MS method was developed and validated for the determination of ethyl ester of eicosapentaenoic acid (EPAEE) and ethyl ester of docosahexaenoic acid (DHAEE). After deproteinized with acetonitrile, the plasma samples were separated on a C18 column using a gradient elution system consisted of methanol and 1.0 mM ammonium acetate in water. The detection used an atmospheric-pressure chemical ionization ion source in positive mode with multiple reaction monitoring for the quantitation of EPAEE and DHAEE. The acceptable linearity was achieved over the concentration ranges of 1.00~1000 ng/mL for EPAEE and 2.50~2500 ng/mL for DHAEE. The method was successfully applied to a pharmacokinetic study of EPAEE and DHAEE in healthy Chinese volunteers after the oral administration of 4 g omega-3-acid ethyl esters 90 soft capsule. The pharmacokinetic profiles of EPAEE and DHAEE were observed for the first time in Chinese volunteers, which reached a maximum concentration of 499 ± 243 ng/mL and 1596 ± 476 ng/mL for EPAEE and DHAEE, respectively. The areas under the plasma concentration-time curve were 1290 ± 765 ng/mL·h for EPAEE and 4369 ± 1680 ng/mL·h for DHAEE, respectively.


Subject(s)
Chromatography, Liquid/methods , Docosahexaenoic Acids/blood , Eicosapentaenoic Acid/analogs & derivatives , Tandem Mass Spectrometry/methods , Administration, Oral , Docosahexaenoic Acids/chemistry , Docosahexaenoic Acids/pharmacokinetics , Eicosapentaenoic Acid/blood , Eicosapentaenoic Acid/chemistry , Eicosapentaenoic Acid/pharmacokinetics , Fatty Acids, Omega-3/administration & dosage , Humans , Limit of Detection , Linear Models , Reproducibility of Results
13.
Mol Nutr Food Res ; 64(11): e1901238, 2020 06.
Article in English | MEDLINE | ID: mdl-32277573

ABSTRACT

SCOPE: The docosahexaenoic acid ester of hydroxy linoleic acid (13-DHAHLA) is a bioactive lipid with anti-inflammatory properties from the family of fatty acid esters of hydroxy fatty acids (FAHFA). METHODS AND RESULTS: To explore the biosynthesis of 13-DHAHLA from dietary oils, C57BL/6N mice are gavaged for 8 days with various corn oil/marine oil mixtures containing the same amount of DHA. Plasma levels of omega-3 FAHFAs are influenced by the lipid composition of the mixtures but do not reflect the changes in bioavailability of polyunsaturated fatty acids in plasma. Triacylglycerol-bound DHA and linoleic acid serve as more effective precursors for 13-DHAHLA synthesis than DHA bound in phospholipids or wax esters. Both 13(S)- and 13(R)-DHAHLA inhibit antigen and PGE2 -induced chemotaxis and degranulation of mast cells to a comparable extent and 13(S)-DHAHLA is identified as the predominant isomer in mouse adipose tissue. CONCLUSION: Here, the optimal nutritional source of DHA is identified, which supports production of anti-inflammatory FAHFAs, as triacylglycerol-based marine oil and also reveals a possible role of triacylglycerols in the synthesis of FAHFA lipokines.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Docosahexaenoic Acids/pharmacokinetics , Oils/chemistry , Oils/pharmacokinetics , Animals , Anti-Inflammatory Agents, Non-Steroidal/blood , Aquatic Organisms , Biological Availability , Chemotaxis/drug effects , Fatty Acids, Omega-3/pharmacokinetics , Fatty Acids, Omega-3/pharmacology , Female , Linoleic Acids/chemistry , Male , Mast Cells/drug effects , Mice, Inbred C57BL , Stereoisomerism , Triglycerides/chemistry
14.
Clin Exp Dent Res ; 6(2): 225-235, 2020 04.
Article in English | MEDLINE | ID: mdl-32250566

ABSTRACT

OBJECTIVES: Sjögren's syndrome (SS) is an autoimmune disease that causes chronic inflammation of the salivary glands leading to secretory dysfunction. Previous studies demonstrated that aspirin-triggered resolvin D1 (AT-RvD1) reduces inflammation and restores tissue integrity in salivary glands. Specifically, progression of SS-like features in NOD/ShiLtJ mice can be systemically halted using AT-RvD1 prior or after disease onset to downregulate proinflammatory cytokines, upregulate anti-inflammatory molecules, and restore saliva production. Therefore, the goal of this paper was to create a physiologically based pharmacokinetic (PBPK) model to offer a reasonable starting point for required total AT-RvD1 dosage to be administered in future mice and humans thereby eliminating the need for excessive use of animals and humans in preclinical and clinical trials, respectively. Likewise, PBPK modeling was employed to increase the range of testable scenarios for elucidating the mechanisms under consideration. MATERIALS AND METHODS: Pharmacokinetics following intravenous administration of a 0.1 mg/kg dose of AT-RvD1 in NOD/ShiLtJ were predicted in both plasma and saliva using PBPK modeling with PK-Sim® and MoBi® Version 7.4 software. RESULTS: The model provides high-value pathways for future validation via in vivo studies in NOD/ShiLtJ to corroborate the findings themselves while also establishing this method as a means to better target drug development and clinical study design. CONCLUSIONS: Clinical and basic research would benefit from knowledge of the potential offered by computer modeling. Specifically, short-term utility of these pharmacokinetic modeling findings involves improved targeting of in vivo studies as well as longer term prospects for drug development and/or better designs for clinical trials.


Subject(s)
Docosahexaenoic Acids/pharmacokinetics , Models, Biological , Sjogren's Syndrome/drug therapy , Administration, Intravenous , Animals , Aspirin/pharmacology , Clinical Trials as Topic/methods , Computer Simulation , Disease Models, Animal , Docosahexaenoic Acids/administration & dosage , Drug Dosage Calculations , Drug Evaluation, Preclinical/methods , Humans , Mice , Saliva/chemistry , Salivary Glands/metabolism , Sjogren's Syndrome/blood , Tissue Distribution
15.
Mol Nutr Food Res ; 64(12): e2000059, 2020 06.
Article in English | MEDLINE | ID: mdl-32304625

ABSTRACT

SCOPE: Currently available omega-3 fatty acid supplements do not enrich the docosahexaenoic acid (DHA) of the adult brain because they are absorbed as triacylglycerol, whereas the transporter at the blood brain barrier requires lysophosphatidylcholine (LPC)-DHA. The hypothesis that treatment of krill oil (KO), which contains DHA/eicosapentaenoic acid (EPA) at the SN2 position of phosphatidylcholine, with SN1-specific lipase will generate LPC-DHA/EPA and which can be absorbed intact and transported into the brain, is tested. METHODS: KO and fish oil (FO) are treated with Mucor meihei lipase, incorporated into AIN 93G diet, and fed to 2-month-old mice for 30 days. Fatty acid composition is analyzed by gas chromatography/mass spectroscopy. Brain derived neurotrophic factor (BDNF) is measured by ELISA. RESULTS: Lipase-treated (LT) KO increases brain DHA and EPA, respectively, 5-and 70-fold better than untreated (UT) KO. FO, whether lipase-treated or not, has no effect on brain DHA/EPA. LTKO is also more efficient in enriching liver DHA/EPA, but less efficient than UTKO and FO in enriching adipose tissue and heart. Brain BDNF is significantly increased by LTKO, but only marginally by other preparations. CONCLUSIONS: Pretreatment of dietary KO with lipase enables it to efficiently increase brain DHA/EPA because of the generation of LPC-DHA/EPA.


Subject(s)
Brain/metabolism , Docosahexaenoic Acids/pharmacokinetics , Eicosapentaenoic Acid/pharmacokinetics , Euphausiacea/chemistry , Oils/pharmacokinetics , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animals , Brain/drug effects , Brain-Derived Neurotrophic Factor/metabolism , Dietary Supplements , Docosahexaenoic Acids/blood , Eicosapentaenoic Acid/blood , Fatty Acids, Omega-3/metabolism , Fish Oils/chemistry , Fish Oils/pharmacokinetics , Heart/drug effects , Lipase/chemistry , Liver/drug effects , Liver/metabolism , Male , Mice, Inbred C57BL , Oils/chemistry , Tissue Distribution
16.
Drug Dev Ind Pharm ; 46(4): 597-605, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32162980

ABSTRACT

Development of self-nanoemulsifying drug delivery systems (SNEDDS) of docosahexaenoic acid (DHA) is reported with the aim to achieve enhanced dissolution rate. The optimized composition of liquid SNEDDS (L-SNEDDS) formulation was Labrafil M1944 CS, 47% v/v Tween 80, 27% v/v Transcutol P, and 0.1% v/v DHA. L-SNEDDS were solidified using Syloid XDP 3150 as solid porous carrier. The droplet size, polydispersity index, zeta potential, percentage drug loading, and cloud point for L-SNEDDS were found to be 43.51 ± 1.36 nm, 0.186 ± 0.053, -19.20 ± 1.21 mV, 93.23 ± 1.71, and 88.60 ± 2.54 °C, respectively. Similarly, for solid SNEDDS (S-SNEDDS) the above parameters were found to be 57.32 ± 1.87 nm, 0.261 ± 0.043, -16.60 ± 2.18 mV, 91.23 ± 1.88, and 89.50 ± 1.18 °C, respectively. The formulations (L-SNEDDS, S-SNEDDS powder, and S-SNEDDS tablet) showed significant (p<.05) improvement in dissolution rate of drug in 0.1 N HCl (pH 1.2) and phosphate buffer (pH 6.8) as compared to unprocessed DHA. In both the dissolution media, the dissolution rate was found more that 85% in 90 min. Absence of drug precipitation, phase separation, and turbidity during thermodynamic stability studies indicated that the developed SNEDDS were stable. Hence, it was concluded that SNEDDS have offered sufficient stability as well as dissolution rate of DHA.


Subject(s)
Docosahexaenoic Acids/administration & dosage , Drug Delivery Systems/methods , Nanoparticles/chemistry , Administration, Oral , Biological Availability , Docosahexaenoic Acids/pharmacokinetics , Drug Liberation , Drug Stability , Emulsions/chemistry , Particle Size , Silicon Dioxide/chemistry , Solubility , Surface-Active Agents , Tablets
17.
Article in English | MEDLINE | ID: mdl-31931275

ABSTRACT

Oxidized lipids derived from omega-6 (n-6) and omega-3 (n-3) polyunsaturated fatty acids, collectively known as oxylipins, are bioactive signaling molecules that play diverse roles in human health and disease. Supplementation with n-3 docosahexaenoic acid (DHA) during pregnancy has been reported to decrease the risk of preterm birth in singleton pregnancies, which may be due to effects of DHA supplementation on oxylipins or their precursor n-6 and n-3 fatty acids. There is only limited understanding of the levels and trajectory of changes in plasma oxylipins during pregnancy, effects of DHA supplementation on oxylipins and unesterified fatty acids, and whether and how oxylipins and their unesterified precursor fatty acids influence preterm birth. In the present study we used liquid chromatography-tandem mass spectrometry to profile oxylipins and their precursor fatty acids in the unesterified pool using plasma samples collected from a subset of pregnant Australian women who participated in the ORIP (Omega-3 fats to Reduce the Incidence of Prematurity) study. ORIP is a large randomized controlled trial testing whether daily supplementation with n-3 DHA can reduce the incidence of early preterm birth compared to control. Plasma was collected at study entry (≈pregnancy week 14) and again at ≈week 24, in a subgroup of 48 ORIP participants-12 cases with spontaneous preterm (<37 weeks) birth and 36 matched controls with spontaneous term (≥40 weeks) birth. In the combined preterm and term pregnancies, we observed that in the control group without DHA supplementation unesterified AA and AA-derived oxylipins 12-HETE, 15-HETE and TXB2 declined between weeks 14-24 of pregnancy. Compared to control, DHA supplementation increased unesterified DHA, EPA, and AA, DHA-derived 4-HDHA, 10-HDHA and 19,20-EpDPA, and AA-derived 12-HETE at 24 weeks. In exploratory analysis independent of DHA supplementation, participants with concentrations above the median for 5-lipoxygenase derivatives of AA (5-HETE, Odds Ratio (OR) 8.2; p = 0.014) or DHA (4-HDHA, OR 8.0; p = 0.015) at 14 weeks, or unesterified AA (OR 5.1; p = 0.038) at 24 weeks had higher risk of spontaneous preterm birth. The hypothesis that 5-lipoxygenase-derived oxylipins and unesterified AA could serve as mechanism-based biomarkers predicting spontaneous preterm birth should be evaluated in larger, adequately powered studies.


Subject(s)
Docosahexaenoic Acids/administration & dosage , Fatty Acids, Unsaturated/blood , Oxylipins/blood , Premature Birth/prevention & control , Adult , Australia , Chromatography, Liquid , Dietary Supplements , Docosahexaenoic Acids/pharmacokinetics , Fatty Acids, Omega-3/blood , Fatty Acids, Omega-6/blood , Female , Gestational Age , Humans , Pregnancy , Tandem Mass Spectrometry
18.
Mol Nutr Food Res ; 64(4): e1900824, 2020 02.
Article in English | MEDLINE | ID: mdl-31797565

ABSTRACT

Omega-3 fatty acid consumption has been suggested to be beneficial for the prevention of type 2 diabetes mellitus (T2DM). Its effects have been attributed to anti-inflammatory activity, with the inhibition of arachidonic acid metabolism playing a central role. However, a more recent view is that omega-3 fatty acids play an active role as the precursors of potent, specialized pro-resolving mediators (SPMs), such as resolvins, protectins, and maresins. Docosahexaenoic acid (DHA)- and eicosapentaenoic-acid-derived SPMs are identified in the adipose tissue but the levels of certain SPMs (e.g., protectin D1) are markedly reduced with obesity, suggesting adipose SPM deficiency, potentially resulting in unresolved inflammation. Supplementation of the biosynthetic intermediates of SPM (e.g., 17-hydroxy-DHA) or omega-3 fatty acids increases the level of adipose SPMs, reduces adipose inflammation (decrease in macrophage accumulation and change to less inflammatory macrophages), and enhances insulin sensitivity. The findings from studies using rodent obesity models must be translated to humans. It will be important to further elucidate the underlying mechanisms by which obesity reduces the levels of and the sensitivity to SPM in adipose tissues. This will enable the development of nutrition therapy to enhance the effects of omega-3 fatty acids in the prevention and/or treatment of T2DM.


Subject(s)
CD59 Antigens/immunology , Docosahexaenoic Acids/pharmacokinetics , Fatty Acids, Omega-3/pharmacokinetics , Metabolic Syndrome/immunology , Animals , CD59 Antigens/metabolism , CD59 Antigens/pharmacokinetics , Diabetes Mellitus, Type 2/prevention & control , Docosahexaenoic Acids/immunology , Docosahexaenoic Acids/metabolism , Eicosapentaenoic Acid/immunology , Eicosapentaenoic Acid/metabolism , Eicosapentaenoic Acid/pharmacokinetics , Fatty Acids, Omega-3/metabolism , Fish Oils/chemistry , Fish Oils/pharmacokinetics , Humans , Inflammation/diet therapy , Inflammation/prevention & control , Metabolic Syndrome/metabolism , Metabolic Syndrome/prevention & control , Obesity/complications , Obesity/diet therapy
19.
J Nutr Biochem ; 74: 108231, 2019 12.
Article in English | MEDLINE | ID: mdl-31665653

ABSTRACT

Docosahexaenoic acid (DHA) is highly concentrated in the brain, and its deficiency is associated with several neurological disorders including Alzheimer's disease. However, the currently used supplements do not appreciably enrich brain DHA, although they enrich most other tissues. We tested the hypothesis that the ability of the dietary carrier to augment brain DHA depends upon the generation of DHA-lysophosphatidylcholine (LPC), the preferred carrier of DHA across the blood brain barrier. We compared the efficacy of DHA-triacylglycerol (TAG), di-DHA phosphatidylcholine (PC) and DHA-LPC to enrich brain DHA following their gavage to normal rats for 30 days, all at a dose of 10 mg DHA/day. The results show that DHA from TAG, which is released as free DHA or monoacylglycerol during digestion and is absorbed as TAG in chylomicrons, was incorporated preferentially into adipose tissue and heart but not into brain. In contrast, LPC-DHA increased brain DHA by up to 100% but had no effect on adipose tissue. Di-DHA PC, which generates both free DHA and LPC-DHA during the digestion, enriched DHA in brain, as well as in heart and liver. Brain-derived neurotrophic factor was increased by di-DHA PC and DHA-LPC, but not by TAG-DHA, showing that enrichment of brain DHA correlated with its functional effect. We conclude that dietary DHA from TAG or from natural PC (sn-2 position) is not suitable for brain enrichment, whereas DHA from LPC (at either sn-1 or sn-2 position) or from sn-1 position of PC efficiently enriches the brain and is functionally effective.


Subject(s)
Brain/metabolism , Docosahexaenoic Acids/administration & dosage , Drug Carriers/administration & dosage , Lysophosphatidylcholines/administration & dosage , Phosphatidylcholines/administration & dosage , Triglycerides/administration & dosage , Animals , Arachidonic Acid/metabolism , Arachidonic Acid/pharmacokinetics , Behavior, Animal/drug effects , Brain/drug effects , Brain-Derived Neurotrophic Factor/metabolism , Dietary Supplements , Docosahexaenoic Acids/metabolism , Docosahexaenoic Acids/pharmacokinetics , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Lysophosphatidylcholines/blood , Male , Maze Learning , Rats, Sprague-Dawley , Tissue Distribution
20.
Prog Lipid Res ; 75: 100997, 2019 07.
Article in English | MEDLINE | ID: mdl-31442526

ABSTRACT

A typical feature of marine foods is that they are rich in docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA), which have formed a large-scale global industry. DHA/EPA phospholipids (PLs) are ubiquitous in marine foods and are the main DHA/EPA molecular forms in fish roe, shrimp and shellfish. Much attention has been focused on the bioavailability and health benefits that are influenced by the type and esterified form of dietary fatty acids. Recently, numerous findings have suggested that dietary DHA/EPA-PLs are superior to the triacylglycerol (TAG) or ethyl ester forms in exerting their functional properties through specific mechanisms of action. However, there is no comprehensive review covering the health benefits of dietary marine DHA/EPA-enriched PLs. In this paper, we review publications on the nutritional functions of DHA/EPA-enriched glycerophospholipids, including the effects on brain function, antitumor activity, lipid metabolism, and glucose metabolism. The current research status regarding the active ingredients, sources, models, treatment, duration, and mechanisms are presented. In addition, the way in which the structure-activity relationship of DHA/EPA-PLs is affected by ester-bond structure at the sn-1 position, fatty acid at the sn-2 position and polar head group at the sn-3 position is also reviewed. DHA/EPA-PLs are one of the major n-3 long-chain polyunsaturated fatty acid dietary forms in our diet, and we should maximize the ability to fully exploit the nutritional properties of DHA/EPA.


Subject(s)
Aquatic Organisms/chemistry , Docosahexaenoic Acids/pharmacokinetics , Eicosapentaenoic Acid/pharmacokinetics , Animals , Antineoplastic Agents/pharmacokinetics , Biological Availability , Blood Glucose/drug effects , Dietary Fats, Unsaturated , Humans , Lipid Metabolism/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...