Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 199
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Biochemistry ; 60(22): 1741-1754, 2021 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-34029049

RESUMEN

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.


Asunto(s)
Ácidos Docosahexaenoicos/metabolismo , Lipooxigenasas/metabolismo , Regulación Alostérica , Araquidonato 12-Lipooxigenasa/metabolismo , Araquidonato 15-Lipooxigenasa/metabolismo , Ácido Araquidónico/metabolismo , Ácidos Araquidónicos/metabolismo , Vías Biosintéticas , Plaquetas/metabolismo , Ácidos Docosahexaenoicos/farmacocinética , Ácidos Docosahexaenoicos/farmacología , Humanos , Lipooxigenasa/metabolismo , Lipooxigenasas/biosíntesis
2.
J Neurosci Res ; 99(2): 561-572, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32964457

RESUMEN

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.


Asunto(s)
Daño Encefálico Crónico/prevención & control , Ácidos Docosahexaenoicos/uso terapéutico , Accidente Cerebrovascular Isquémico/dietoterapia , Degeneración Nerviosa/prevención & control , Accidente Cerebrovascular/prevención & control , Animales , Antiinflamatorios/administración & dosificación , Antiinflamatorios/farmacocinética , Antiinflamatorios/uso terapéutico , Antioxidantes/administración & dosificación , Antioxidantes/farmacocinética , Antioxidantes/uso terapéutico , Apoptosis/efectos de los fármacos , Disponibilidad Biológica , Transporte Biológico , Barrera Hematoencefálica , Daño Encefálico Crónico/etiología , Grasas de la Dieta/administración & dosificación , Grasas de la Dieta/farmacocinética , Grasas de la Dieta/uso terapéutico , Ácidos Docosahexaenoicos/administración & dosificación , Ácidos Docosahexaenoicos/metabolismo , Ácidos Docosahexaenoicos/farmacocinética , Ácidos Docosahexaenoicos/farmacología , Proteínas de Unión a Ácidos Grasos/fisiología , Aceites de Pescado/administración & dosificación , Aceites de Pescado/farmacocinética , Humanos , Incidencia , Accidente Cerebrovascular Isquémico/complicaciones , Accidente Cerebrovascular Isquémico/epidemiología , Lípidos de la Membrana/metabolismo , Ratones , Proteínas de Neoplasias/fisiología , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Aceites de Plantas/administración & dosificación , Aceites de Plantas/farmacocinética , Transducción de Señal/efectos de los fármacos , Simportadores/deficiencia , Simportadores/fisiología , Ácido alfa-Linolénico/farmacocinética
3.
Toxicol Appl Pharmacol ; 400: 115037, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32417438

RESUMEN

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.


Asunto(s)
Catéteres de Permanencia/efectos adversos , Ácidos Docosahexaenoicos/administración & dosificación , Arteria Hepática , Infusiones Intraarteriales/efectos adversos , Lipoproteínas LDL/administración & dosificación , Hígado/metabolismo , Nanopartículas/administración & dosificación , Animales , Glucemia/análisis , Ácidos Docosahexaenoicos/farmacocinética , Infusiones Intraarteriales/métodos , Pruebas de Función Renal , Lípidos/sangre , Lipoproteínas LDL/farmacocinética , Hígado/irrigación sanguínea , Pruebas de Función Hepática , Masculino , Ratas Wistar , Distribución Tisular
4.
Biomed Chromatogr ; 34(10): e4905, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32449539

RESUMEN

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.


Asunto(s)
Cromatografía Liquida/métodos , Ácidos Docosahexaenoicos/sangre , Ácido Eicosapentaenoico/análogos & derivados , Espectrometría de Masas en Tándem/métodos , Administración Oral , Ácidos Docosahexaenoicos/química , Ácidos Docosahexaenoicos/farmacocinética , Ácido Eicosapentaenoico/sangre , Ácido Eicosapentaenoico/química , Ácido Eicosapentaenoico/farmacocinética , Ácidos Grasos Omega-3/administración & dosificación , Humanos , Límite de Detección , Modelos Lineales , Reproducibilidad de los Resultados
5.
Drug Dev Ind Pharm ; 46(4): 597-605, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32162980

RESUMEN

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.


Asunto(s)
Ácidos Docosahexaenoicos/administración & dosificación , Sistemas de Liberación de Medicamentos/métodos , Nanopartículas/química , Administración Oral , Disponibilidad Biológica , Ácidos Docosahexaenoicos/farmacocinética , Liberación de Fármacos , Estabilidad de Medicamentos , Emulsiones/química , Tamaño de la Partícula , Dióxido de Silicio/química , Solubilidad , Tensoactivos , Comprimidos
6.
Curr Opin Clin Nutr Metab Care ; 22(2): 116-123, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30550388

RESUMEN

PURPOSE OF REVIEW: To describe recent strategies that have been developed to enhance absorption of eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) from dietary supplements. RECENT FINDINGS: The long-chain omega-3 fatty acids EPA and DHA have important physiologic functions, and numerous potential health benefits have been suggested by results from observational studies and randomized, controlled trials. EPA and DHA intakes in the average American diet are substantially below recommended levels. Dietary supplements are available for consumers wishing to increase their intakes, but many of these are in ethyl ester formulations from which EPA and DHA are poorly absorbed when consumed without a meal containing dietary fat. Technologies have been developed to enhance EPA and DHA absorption through in-situ emulsification, which facilitates bioavailability, even in the absence of a fat-containing meal. Findings from randomized controlled trials of absorption enhancers incorporated into omega-3 fatty acid supplements demonstrate that they can markedly improve the bioavailability of EPA and DHA. SUMMARY: The development of absorption enhancement technology to increase bioavailability of long-chain omega-3 fatty acids has important implications for studies on the health effects of dietary supplement and pharmaceutical products containing EPA and/or DHA.


Asunto(s)
Suplementos Dietéticos , Ácidos Grasos Omega-3/farmacocinética , Disponibilidad Biológica , Ácidos Docosahexaenoicos/farmacocinética , Ácido Eicosapentaenoico/farmacocinética , Ésteres , Humanos
7.
J Nutr ; 149(1): 46-56, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30601995

RESUMEN

Background: Docosahexaenoic acid (DHA) has been shown to reduce growth of breast cancer cells in vitro and in vivo; it may also benefit the action of cytotoxic cancer drugs. The mechanisms for these observations are not completely understood. Objectives: We sought to explore how pretreatment of MDA-MB-231 breast cancer cells with DHA alters gene expression with doxorubicin (DOX) treatment and confirm that feeding DHA to tumor-bearing nu/nu mice improves the efficacy of DOX. Methods: MDA-MB-231 cells were subjected to 4 conditions: a control mixture of 40 µM linoleic and 40 µM oleic acid (OALA), DHA (60 µM plus OALA), OALA DOX (0.41 µM), or DHA DOX (plus OALA) and assessed for effects on viability and function. Female nu/nu mice (6 wk old) bearing MDA-MB-231 tumors were randomly assigned to a nutritionally complete diet (20 g ± 2.8 g DHA/100 g diet) containing a polyunsaturated:saturated fat ratio of 0.5, with or without injections 2 times/wk of 5 mg DOX/kg for 4 wk. Results: Microarray and protein analysis indicated that DHA DOX cells, compared with OALA DOX, had upregulated expression of apoptosis genes, Caspase-10 (1.3-fold), Caspase-9 (1.4-fold), and Receptor (TNFRSF)-interacting serine-threonine kinase 1 (RIPK1) (1.2-fold), while downregulating cell cycle genes, Cyclin B1 (-2.1-fold), WEE1 (-1.6-fold), and cell division cycle 25 homolog C (CDC25C) (-1.8-fold) (P < 0.05). DHA DOX-treated mice had 50% smaller tumors than control mice (P < 0.05). Analysis of proapoptotic proteins from tumors of DHA DOX mice showed increased Caspase-10 (by 68%) and BH3 interacting domain death agonist (Bid) (by 50%), decreased B-cell CLL/lymphoma 2 (BCL2) (by 24%), and decreased cell cycle proteins Cyclin B1 and Cdc25c (both by 42%), compared with control mice (P < 0.05). Conclusions: Supplementation with DHA facilitates the action of DOX in MDA-MB-231 cells and in nu/nu mice, which may occur via amplification of the effect of DOX on apoptosis and cell cycle genes.


Asunto(s)
Ácidos Docosahexaenoicos/farmacología , Ácidos Docosahexaenoicos/farmacocinética , Doxorrubicina/farmacología , Doxorrubicina/farmacocinética , Neoplasias Experimentales/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Neoplasias de la Mama , Puntos de Control del Ciclo Celular , Línea Celular Tumoral , Interacciones Farmacológicas , Femenino , Humanos , Ratones , Ratones Desnudos , Distribución Aleatoria
8.
J Neurochem ; 144(1): 81-92, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29105065

RESUMEN

Lower levels of the cognitively beneficial docosahexaenoic acid (DHA) are often observed in Alzheimer's disease (AD) brains. Brain DHA levels are regulated by the blood-brain barrier (BBB) transport of plasma-derived DHA, a process facilitated by fatty acid-binding protein 5 (FABP5). This study reports a 42.1 ± 12.6% decrease in the BBB transport of 14 C-DHA in 8-month-old AD transgenic mice (APPswe,PSEN1∆E9) relative to wild-type mice, associated with a 34.5 ± 6.7% reduction in FABP5 expression in isolated brain capillaries of AD mice. Furthermore, short-term spatial and recognition memory deficits were observed in AD mice on a 6-month n-3 fatty acid-depleted diet, but not in AD mice on control diet. This intervention led to a dramatic reduction (41.5 ± 11.9%) of brain DHA levels in AD mice. This study demonstrates FABP5 deficiency and impaired DHA transport at the BBB are associated with increased vulnerability to cognitive deficits in mice fed an n-3 fatty acid-depleted diet, in line with our previous studies demonstrating a crucial role of FABP5 in BBB transport of DHA and cognitive function.


Asunto(s)
Barrera Hematoencefálica , Trastornos del Conocimiento/etiología , Ácidos Docosahexaenoicos/farmacocinética , Proteínas de Unión a Ácidos Grasos/fisiología , Proteínas de Neoplasias/fisiología , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Química Encefálica , Trastornos del Conocimiento/genética , Trastornos del Conocimiento/metabolismo , Grasas de la Dieta/administración & dosificación , Ácidos Docosahexaenoicos/deficiencia , Proteínas de Escherichia coli , Proteínas de Unión a Ácidos Grasos/biosíntesis , Ácidos Grasos Omega-3/deficiencia , Femenino , Humanos , Masculino , Aprendizaje por Laberinto , Trastornos de la Memoria/etiología , Trastornos de la Memoria/genética , Trastornos de la Memoria/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación Missense , Proteínas de Neoplasias/biosíntesis , Polisacárido Liasas , Presenilina-1/genética , Presenilina-1/metabolismo , Reconocimiento en Psicología , Proteínas Recombinantes de Fusión/metabolismo
9.
Biochim Biophys Acta Biomembr ; 1860(5): 1135-1142, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29338975

RESUMEN

The understanding of lipid bilayer structure and function has been advanced by the application of molecular fluorophores. However, the effects of these probe molecules on the physicochemical properties of membranes being studied are poorly understood. A quartz crystal microbalance with dissipation monitoring instrument was used in this work to investigate the impact of two commonly used fluorescent probes, 1­palmitoyl­2­{12­[(7­nitro­2­1,3­benzoxadiazol­4­yl)amino]dodecanoyl}­sn­glycero­3­phosphocholine (NBD-PC) and 1,2­dipalmitoyl­sn­glycero­3­phosphoethanolamine­n­(lissamine rhodamine­B­sulfonyl) (Lis-Rhod PE), on the formation and physicochemical properties of a 1­palmitoyl­2­oleoyl­sn­glycero­3­phosphocholine supported lipid bilayer (POPC-SLB). The interaction of the POPC-SLB and fluorophore-modified POPC-SLB with docosahexaenoic acid, DHA, was evaluated. The incorporation of DHA into the POPC-SLB was observed to significantly decrease in the presence of the Lis-Rhod PE probe compared with the POPC-SLB. In addition, it was observed that the small concentration of DHA incorporated into the POPC:NBD-PC SLB can produce rearrangement processes followed by the lost not only of DHA but also of POPC or NBD-PC molecules or both during the washing step. This work has significant implications for the interpretation of data employing fluorescent reporter molecules within SLBs.


Asunto(s)
Ácidos Docosahexaenoicos/metabolismo , Colorantes Fluorescentes/farmacología , Membrana Dobles de Lípidos/metabolismo , Fosfatidilcolinas/metabolismo , 4-Cloro-7-nitrobenzofurazano/análogos & derivados , 4-Cloro-7-nitrobenzofurazano/química , 4-Cloro-7-nitrobenzofurazano/farmacología , Ácidos Docosahexaenoicos/farmacocinética , Colorantes Fluorescentes/química , Membrana Dobles de Lípidos/química , Conformación Molecular , Fosfatidilcolinas/química , Fosfatidilcolinas/farmacocinética , Fosfatidilcolinas/farmacología , Tecnicas de Microbalanza del Cristal de Cuarzo , Rodaminas/química , Rodaminas/farmacología
10.
Biochim Biophys Acta Biomembr ; 1860(5): 1125-1134, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29305832

RESUMEN

Eicosapentaenoic (EPA, 20:5), docosahexaenoic (DHA, 22:6) and docosapentaenoic (DPA, 22:5) acids are omega-3 polyunsaturated fatty acids (n-3 PUFA) obtained from dietary consumption of fish oils that potentially alleviate the symptoms of a range of chronic diseases. We focus here on the plasma membrane as a site of action and investigate how they affect molecular organization when taken up into a phospholipid. All atom MD simulations were performed to compare 1-stearoyl-2-eicosapentaenoylphosphatylcholine (EPA-PC, 18:0-20:5PC), 1-stearoyl-2-docosahexaenoylphosphatylcholine (DHA-PC, 18:0-22:6PC), 1-stearoyl-2-docosapentaenoylphosphatylcholine (DPA-PC, 18:0-22:5PC) and, as a monounsaturated control, 1-stearoyl-2-oleoylphosphatidylcholine (OA-PC, 18:0-18:1PC) bilayers. They were run in the absence and presence of 20mol% cholesterol. Multiple double bonds confer high disorder on all three n-3 PUFA. The different number of double bonds and chain length for each n-3 PUFA moderates the reduction in membrane order exerted (compared to OA-PC, S¯CD=0.152). EPA-PC (S¯CD=0.131) is most disordered, while DPA-PC (S¯CD=0.140) is least disordered. DHA-PC (S¯CD=0.139) is, within uncertainty, the same as DPA-PC. Following the addition of cholesterol, order in EPA-PC (S¯CD=0.169), DHA-PC (S¯CD=0.178) and DPA-PC (S¯CD=0.182) is increased less than in OA-PC (S¯CD=0.214). The high disorder of n-3 PUFA is responsible, preventing the n-3 PUFA-containing phospholipids from packing as close to the rigid sterol as the monounsaturated control. Our findings establish that EPA, DHA and DPA are not equivalent in their interactions within membranes, which possibly contributes to differences in clinical efficacy.


Asunto(s)
Membrana Celular/metabolismo , Ácidos Docosahexaenoicos/farmacocinética , Ácido Eicosapentaenoico/farmacocinética , Ácidos Grasos Omega-3/química , Ácidos Grasos Omega-3/farmacocinética , Ácidos Grasos Insaturados/farmacocinética , Membrana Celular/química , Colesterol/metabolismo , Ácidos Docosahexaenoicos/química , Ácido Eicosapentaenoico/química , Ácidos Grasos Omega-3/clasificación , Ácidos Grasos Omega-3/metabolismo , Ácidos Grasos Insaturados/química , Fluidez de la Membrana , Modelos Moleculares , Conformación Molecular , Simulación de Dinámica Molecular
11.
Curr Opin Clin Nutr Metab Care ; 21(2): 78-82, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29206689

RESUMEN

PURPOSE OF REVIEW: This review outlines recent advances in placental lipid transport in relation to maternal metabolic status and pregnancy outcome. A particular focus of this review will be on the way these findings may influence our understanding of placental transfer of the essential fatty acid docosahexaenoic acid (DHA) which is crucial for fetal neurodevelopment and of lipid transfer as a predisposing factor for childhood obesity. RECENT FINDINGS: Placental metabolism may determine the quantity and composition of fatty acids delivered to the fetus. Maternal factors, such as obesity, appear to regulate placental lipid metabolism and may influence fatty acids delivery to the fetus. Although the role of placental metabolism is now recognized, new evidence also suggests important roles for nontraditional fatty acid transporters such as Mfsd2a which facilitates transfer of DHA. SUMMARY: Placental lipid metabolism is likely to be a determinant of placental transfer of fatty acids to the fetus. Maternal conditions, such as obesity, have now been shown to regulate placental lipid metabolism and thus may influence fatty acid transfer and fetal development. However, it is not yet clear how regulation of placental lipid metabolism affects fatty acid delivery to the fetus and its long-term health.


Asunto(s)
Ácidos Docosahexaenoicos/farmacocinética , Ácidos Grasos/farmacocinética , Placenta/metabolismo , Transporte Biológico , Femenino , Desarrollo Fetal , Feto/metabolismo , Humanos , Metabolismo de los Lípidos , Intercambio Materno-Fetal , Obesidad/metabolismo , Embarazo , Simportadores , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
12.
Curr Opin Clin Nutr Metab Care ; 21(2): 71-77, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29206690

RESUMEN

PURPOSE OF REVIEW: To summarize recent advances pertaining to the mechanisms regulating brain docosahexaenoic acid (DHA) uptake. DHA is an omega-3 polyunsaturated fatty acid highly enriched in neuronal membranes and it is implicated in several important neurological processes. However, DHA synthesis is extremely limited within the brain. RECENT FINDINGS: There are two main plasma pools that supply the brain with DHA: the nonesterified pool and the lysophosphatidylcholine (lysoPtdCho) pool. Quantitatively, plasma nonesterified-DHA (NE-DHA) is the main contributor to brain DHA. Fatty acid transport protein 1 (FATP1) in addition to fatty acid-binding protein 5 (FABP5) are key players that regulate brain uptake of NE-DHA. However, the plasma half-life of lysoPtdCho-DHA and its brain partition coefficient are higher than those of NE-DHA after intravenous administration. SUMMARY: The mechanisms regulating brain DHA uptake are more complicated than once believed, but recent advances provide some clarity notably by suggesting that FATP1 and FABP5 are key contributors to cellular uptake of DHA at the blood-brain barrier. Elucidating how DHA enters the brain is important as we might be able to identify methods to better deliver DHA to the brain as a potential therapeutic.


Asunto(s)
Encéfalo/efectos de los fármacos , Ácidos Docosahexaenoicos/farmacocinética , Administración Intravenosa , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Línea Celular Tumoral , Ácidos Docosahexaenoicos/sangre , Proteínas de Transporte de Ácidos Grasos/genética , Proteínas de Transporte de Ácidos Grasos/metabolismo , Proteínas de Unión a Ácidos Grasos/genética , Proteínas de Unión a Ácidos Grasos/metabolismo , Humanos , Insulina/sangre , Neuronas/metabolismo
13.
Crit Rev Food Sci Nutr ; 58(15): 2617-2631, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-28665691

RESUMEN

Docosahexaenoic acid (DHA) is a key nutritional n-3 polyunsaturated fatty acid and needs to be supplied by the human diet. High levels of DHA intake appear to reduce the risk of depression, bipolar disorder, and mood disorders. On the basis of these connections between DHA and neurological health, this paper reviews what is currently known about DHA and children neurodevelopment as well as the benefits of DHA intake to prevention of autism and behavior disorders through a selective and representative revision of different papers ranging from pure observational studies to randomized controlled trials (RCTs). This review also highlights the issue of DHA bioaccessibility and its implications to the performance of studies. As main conclusions, it can be mentioned that high DHA intake may prevent autism disorder. However, more studies are required to strengthen the connection between autism and dietary DHA. Regarding behavioral disorders, the evidence is also contradictory, thereby raising the need of further studies. From all screened studies on autism, attention deficit/hyperactivity disorder, and other disorders, it can be concluded that study samples should be larger for greater statistical significance and RCTs should be more carefully designed.


Asunto(s)
Desarrollo Infantil/efectos de los fármacos , Fenómenos Fisiológicos Nutricionales Infantiles , Ácidos Docosahexaenoicos/farmacología , Ácidos Docosahexaenoicos/farmacocinética , Disponibilidad Biológica , Niño , Ácidos Docosahexaenoicos/administración & dosificación , Humanos
14.
Pharm Res ; 35(4): 91, 2018 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-29520477

RESUMEN

PURPOSE: The main purpose of this study was to formulate an oil-in-water nanoemulsion of a next generation taxoid DHA-SBT-1214 and evaluate its biodistribution and pharmacokinetics. METHODS: DHA-SBT-1214 was encapsulated in a fish oil containing nanoemulsion using a high pressure homogenization method. Following morphological characterization of the nanoemulsions, qualitative and quantitative biodistribution was evaluated in naïve and cancer stem cell-enriched PPT-2 human prostate tumor bearing mice. RESULTS: DHA-SBT-1214 was successfully encapsulated up to 20 mg/ml in the nanoemulsion formulation and had an average oil droplet size of 200 nm. Using a DiR near infra-red dye encapsulated nanoemulsion, we have shown the delivery of nanoemulsion to mouse tumor region. By quantitative analysis, DHA-SBT-1214 encapsulated nanoemulsion demonstrated improved pharmacokinetic properties in plasma and different tissues as compared to its solution form. Furthermore, the nanoemulsions were stable and had slower in vitro drug release compared to its solution form. CONCLUSIONS: The results from this study demonstrated effective encapsulation of the drug in a nanoemulsion and this nanoemulsion showed sustained plasma levels and enhanced tumor delivery relative to the solution form.


Asunto(s)
Antineoplásicos/farmacocinética , Ácidos Docosahexaenoicos/farmacocinética , Composición de Medicamentos/métodos , Neoplasias/tratamiento farmacológico , Taxoides/farmacocinética , Administración Intravenosa , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Línea Celular Tumoral , Ácidos Docosahexaenoicos/administración & dosificación , Ácidos Docosahexaenoicos/química , Liberación de Fármacos , Emulsiones , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Nanopartículas/química , Taxoides/administración & dosificación , Taxoides/química , Distribución Tisular , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1862(10 Pt A): 1092-1098, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28760485

RESUMEN

Recent studies suggest that at least two pools of plasma docosahexaenoic acid (DHA) can supply the brain: non-esterified DHA (NE-DHA) and lysophosphatidylcholine (lysoPtdCho)-DHA. In contrast to NE-DHA, brain uptake of lysoPtdCho-DHA appears to be mediated by a specific transporter, but whether both forms of DHA supply undergo the same metabolic fate, particularly with regards to enrichment of specific phospholipid (PL) subclasses, remains to be determined. This study aimed to evaluate brain uptake of NE-DHA and lysoPtdCho-DHA into brain PL classes. Fifteen-week-old rats were infused intravenously with radiolabelled NE-14C-DHA or lysoPtdCho-14C-DHA (n=4/group) over five mins to achieve a steady-state plasma level. PLs were extracted from the brain and separated by thin layer chromatography and radioactivity was quantified by liquid scintillation counting. The net rate of entry of lysoPtdCho-DHA into the brain was between 59% and 86% lower than the net rate of entry of NE-DHA, depending on the PL class. The proportion of total PL radioactivity in the lysoPtdCho-14C-DHA group compared to the NE-14C-DHA group was significantly higher in choline glycerophospholipids (ChoGpl) (48% vs 28%, respectively) but lower in ethanolamine glycerophospholipids (EtnGpl) (32% vs 46%, respectively). In both groups, radioactivity was disproportionally high in phosphatidylinositol and ChoGpl but low in phosphatidylserine and EtnGpl compared to the corresponding DHA pool size. This suggests that DHA undergoes extensive PL remodeling after entry into the brain.


Asunto(s)
Encéfalo/metabolismo , Ácidos Docosahexaenoicos , Glicerofosfolípidos/metabolismo , Lisofosfatidilcolinas , Animales , Ácidos Docosahexaenoicos/farmacocinética , Ácidos Docosahexaenoicos/farmacología , Lisofosfatidilcolinas/farmacocinética , Lisofosfatidilcolinas/farmacología , Masculino , Ratas , Ratas Sprague-Dawley
16.
Ophthalmology ; 124(5): 634-643, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28153441

RESUMEN

PURPOSE: To evaluate the associations between intakes of eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) and the intermediate and advanced stages of age-related macular degeneration (AMD). DESIGN: Prospective cohort study. PARTICIPANTS: We followed 75 889 women from the Nurses' Health Study and 38 961 men from the Health Professionals Follow-Up Study who were at least 50 years old, from 1984 to 2012 and 1986 to 2010, respectively. Cohort participants are mostly white (≥95%). METHODS: We assessed dietary intake by a validated food frequency questionnaire (FFQ) at baseline and every 4 years. We calculated cumulative average intakes of EPA and DHA from FFQs and also computed predicted erythrocyte and plasma scores directly from food intake using regression models. Cox proportional hazards models were used to compute the associations with AMD outcomes. MAIN OUTCOME MEASURES: We confirmed 1589 incident intermediate and 1356 advanced AMD cases (primarily neovascular AMD) with a visual acuity of 20/30 or worse, owing primarily to AMD, by medical record review. RESULTS: For intermediate AMD, the pooled hazard ratio (HR) between the 2 cohorts for DHA comparing the extreme quintiles of intake was 0.78 (95% confidence interval [CI], 0.66-0.92; P trend, 0.008) and for EPA + DHA was 0.83 (95% CI, 0.71-0.98; P trend, 0.03). The pooled HR for fatty fish, comparing ≥5 servings per week to almost never, was 0.61 (95% CI, 0.46-0.81; P trend, <0.001). For advanced AMD, the pooled HR for DHA was 1.01 (95% CI, 0.84-1.21; P trend, 0.75) and for fatty fish was 0.80 (95% CI, 0.59-1.08; P trend, 0.11). Secondary analyses using predicted erythrocyte and plasma scores of EPA and DHA yielded slightly stronger inverse associations for intermediate AMD and similar results for advanced AMD. CONCLUSIONS: Higher intakes of EPA and DHA may prevent or delay the occurrence of visually significant intermediate AMD. However, the totality of current evidence for EPA and DHA and advanced AMD is discordant, though there was no association with advanced AMD in the present study.


Asunto(s)
Suplementos Dietéticos , Ácidos Docosahexaenoicos/administración & dosificación , Ácido Eicosapentaenoico/administración & dosificación , Agudeza Visual , Degeneración Macular Húmeda/dietoterapia , Adulto , Anciano , Progresión de la Enfermedad , Ácidos Docosahexaenoicos/farmacocinética , Relación Dosis-Respuesta a Droga , Ácido Eicosapentaenoico/farmacocinética , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Factores de Riesgo , Factores de Tiempo , Resultado del Tratamiento , Degeneración Macular Húmeda/sangre , Degeneración Macular Húmeda/diagnóstico
17.
Crit Rev Food Sci Nutr ; 57(11): 2245-2262, 2017 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-25897862

RESUMEN

The potential antineoplastic effect of the long-chain n-3 polyunsaturated fatty acids (LC n-3 PUFA) eicosapentaenoic (EPA) and docosahexaenoic acid (DHA) remains a highly controversial issue. Numerous animal studies have supported the anticancer role of these dietary fatty acids, whereas conflicting results have been obtained in population studies, and only a few intervention human trials have been so far performed. In view of the possibility that the anticancer effects may be maximally observed within a defined range of EPA and DHA doses, herein we critically review the results and doses used in both animal studies and human clinical trials focusing on the possible n-3 PUFA protective effects against breast and prostate cancer. Our main aim is to identify the EPA and/or DHA ranges of doses needed to obtain clear anticancer effects. This may be of great help in designing future animal studies, and also in understanding the most appropriate dose for further human intervention studies. Moreover, since the healthy effects of these fatty acids have been strictly related to their increased incorporation in plasma and tissue lipids, we also examine and discuss the incorporation changes following the administration of the effective anticancer EPA and/or DHA doses in animals and humans.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Ácidos Docosahexaenoicos/administración & dosificación , Ácido Eicosapentaenoico/administración & dosificación , Neoplasias de la Próstata/tratamiento farmacológico , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/sangre , Neoplasias de la Mama/metabolismo , Modelos Animales de Enfermedad , Ácidos Docosahexaenoicos/farmacocinética , Relación Dosis-Respuesta a Droga , Ácido Eicosapentaenoico/farmacocinética , Femenino , Humanos , Masculino , Neoplasias de la Próstata/metabolismo , Ensayos Clínicos Controlados Aleatorios como Asunto , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Lipids Health Dis ; 16(1): 204, 2017 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-29037249

RESUMEN

BACKGROUND: Absorption of EPA and DHA from Omega-3-acid ethyl ester (EE) concentrate supplements occurs most efficiently when taken in context of a fatty meal; adequate fat intake is required to release bile salts that emulsify and pancreatic enzymes that digest omega-3-containing lipids in the intestine. Current guidelines recommend reduction in fat intake and therefore there is a need to optimize the absorption of Omega-3 in those consuming low-fat or no-fat meals. To this end, BASF has developed an Absorption Acceleration Technology, a novel self-micro-emulsifying delivery system (SMEDS) formulation of highly concentrated Omega-3-acid EE which enables rapid emulsification and microdroplet formation upon entering the aqueous environment of the gut therefore enhances the absorption. METHODS: Two separate single dose, crossover studies were conducted to determine the relative bioavailability of omega-3-acid EE concentrate, either as a novel SMEDS formulation (PRF-021) or as control, in healthy fasted male and female adults at two dose levels (Study 1 "low dose": 630 mg EPA + DHA in PRF-021 vs. 840 mg EPA + DHA in control; Study 2 "high dose": 1680 mg EPA + DHA in PRF-021 vs. 3360 mg EPA + DHA in control). Blood samples were collected immediately before supplementation and at defined time intervals for 48 h. Plasma concentration of total EPA and DHA were determined for pharmacokinetic analysis, area under the curve (AUC) and maximum observed concentration (Cmax) was determined. RESULTS: Total EPA plus DHA absorption from SMEDS formulation PRF-021 were 6.4 and 11.5 times higher compared to control in low- and high-dose studies respectively, determined as the ratio of baseline corrected, dose normalized AUC0-24h of PRF-021 over that of control. EPA and DHA individually showed differing levels of enhancement: the AUC0-24h ratio for EPA was 23.8 and 25.7 in low and high dose studies, respectively, and the AUC0-24h ratio for DHA was 3.6 and 5.6 in low and high dose studies, respectively. Cmax was also increased for both EPA and DHA 2.7- to 9.2-fold. CONCLUSION: PRF-021 is a novel SMEDS formulation of Omega-3-acid EE demonstrating a marked improvement in absorption of a single dose of EPA and DHA EE under fasted conditions. This allows adequate absorption of Omega-3 from the supplement without the requirement of a high-fat meal.


Asunto(s)
Suplementos Dietéticos , Ácidos Docosahexaenoicos/farmacocinética , Sistemas de Liberación de Medicamentos/métodos , Ácido Eicosapentaenoico/farmacocinética , Absorción Intestinal/fisiología , Adulto , Área Bajo la Curva , Disponibilidad Biológica , Estudios Cruzados , Ácidos Docosahexaenoicos/administración & dosificación , Ácido Eicosapentaenoico/administración & dosificación , Emulsiones , Ésteres/administración & dosificación , Ayuno/fisiología , Femenino , Humanos , Masculino , Persona de Mediana Edad
19.
Drug Dev Res ; 78(7): 332-339, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28771757

RESUMEN

Preclinical Research The aim of this work was to evaluate the effect of docosahexaenoic acid (DHA) on the pharmacokinetics and pharmacodynamics-nociception-of naproxen in rats, as well as to determine the gastric safety resulting from this combination versus naproxen alone. Female Wistar rats were orally administered DHA, naproxen or the DHA-naproxen mixture at fixed-ratio combination of 1:3. The antinociceptive effect was evaluated using the formalin test. The gastric injury was determined 3 h after naproxen administration. An isobolographic analysis was performed to characterize the antinociceptive interaction between DHA and naproxen. To determine the possibility of pharmacokinetic interactions, the oral bioavailability of naproxen was evaluated in presence and absence of oral DHA. The experimental effective dose ED30 values (Zexp) were decreased from theoretical additive dose values (Zadd; P < 0.05). The isobolographic analysis showed that the combination exhibited supra-additive interaction. The oral administration of DHA increased the pharmacokinetic parameter AUC0-t of naproxen (P < 0.05). Furthermore, the gastric damage induced by naproxen was abolished when this drug was combined with DHA. These data suggest that oral administration of DHA-naproxen combination induces gastric safety and supra-additive antinociceptive effect in the formalin test so that this combination could be useful to management of inflammatory pain. Drug Dev Res 78 : 332-339, 2017. © 2017 Wiley Periodicals, Inc.


Asunto(s)
Ácidos Docosahexaenoicos/administración & dosificación , Naproxeno/administración & dosificación , Dolor/tratamiento farmacológico , Estómago/efectos de los fármacos , Administración Oral , Animales , Ácidos Docosahexaenoicos/farmacocinética , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Quimioterapia Combinada , Femenino , Masculino , Naproxeno/farmacocinética , Dimensión del Dolor/efectos de los fármacos , Ratas , Ratas Wistar
20.
J Lipid Res ; 57(12): 2208-2216, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27707818

RESUMEN

It was hypothesized that under induced lipid malabsorption/maldigestion conditions, an enriched sn-1(3)-monoacylglycerol (MAG) oil may be a better carrier for n-3 long-chain PUFAs (LC-PUFAs) compared with triacylglycerol (TAG) from fish oil. This monocentric double blinded clinical trial examined the accretion of EPA (500 mg/day) and DHA (300 mg/day) when consumed as TAG or MAG, into the erythrocytes, plasma, and chylomicrons of 45 obese (BMI ≥30 kg/m2 and ≤40 kg/m2) volunteers who were and were not administered Orlistat, an inhibitor of pancreatic lipases. Intake of MAG-enriched oil resulted in higher accretion of LC-PUFAs than with TAG, the concentrations of EPA and DHA in erythrocytes being, respectively, 72 and 24% higher at 21 days (P < 0.001). In addition, MAG increased the plasma concentration of EPA by 56% (P < 0.001) as compared with TAG. In chylomicrons, MAG intake yielded higher levels of EPA with the area under the curve (0-10 h) of EPA being 55% greater (P = 0.012). In conclusion, in obese human subjects with Orlistat-induced lipid maldigestion/malabsorption conditions, LC-PUFA MAG oil increased LC-PUFA levels in erythrocytes, plasma, and chylomicrons to a greater extent than TAG. These results indicate that MAG oil might require minimal enzymatic digestion prior to intestinal uptake and transfer across the epithelial barrier.


Asunto(s)
Ácidos Docosahexaenoicos/farmacocinética , Ácido Eicosapentaenoico/farmacocinética , Trastornos del Metabolismo de los Lípidos/tratamiento farmacológico , Monoglicéridos/administración & dosificación , Adulto , Fármacos Antiobesidad/efectos adversos , Fármacos Antiobesidad/uso terapéutico , Membrana Celular/metabolismo , Quilomicrones , Ácidos Docosahexaenoicos/administración & dosificación , Método Doble Ciego , Ácido Eicosapentaenoico/administración & dosificación , Eritrocitos/metabolismo , Femenino , Aceites de Pescado/administración & dosificación , Aceites de Pescado/farmacocinética , Humanos , Lactonas/efectos adversos , Lactonas/uso terapéutico , Trastornos del Metabolismo de los Lípidos/inducido químicamente , Masculino , Persona de Mediana Edad , Obesidad/sangre , Obesidad/tratamiento farmacológico , Orlistat
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA