SciELO - Scientific Electronic Library Online

 
vol.27 número4Relación entre los niveles séricos de la proteína C reactiva y medidas antropométricas: una revisión sistemática de los estudios realizados en SuraméricaMecanismos moleculares posibles mediados por la soja en la prevención y el tratamiento de la hepatopatía grasa no alcohólica índice de autoresíndice de materiabúsqueda de artículos
Home Pagelista alfabética de revistas  

Servicios Personalizados

Revista

Articulo

Indicadores

Links relacionados

  • En proceso de indezaciónCitado por Google
  • No hay articulos similaresSimilares en SciELO
  • En proceso de indezaciónSimilares en Google

Compartir


Nutrición Hospitalaria

versión On-line ISSN 1699-5198versión impresa ISSN 0212-1611

Nutr. Hosp. vol.27 no.4 Madrid jul./ago. 2012

https://dx.doi.org/10.3305/nh.2012.27.4.5783 

REVISIÓN

 

Role of oleic acid in immune system; mechanism of action; a review

Papel del ácido oleico en el sistema inmune; mecanismo de acción; revisión científica

 

 

C. Carrillo, M.a del M. Cavia and S. Alonso-Torre

Área de Nutrición y Bromatología, Facultad de Ciencias. Universidad de Burgos. Burgos. Spain.

Correspondence

 

 


ABSTRACT

Introduction: Although n-3 polyunsaturated fatty acids have been widely described as anti-inflammatory fats, little is known about the role of oleic acid in immune system.
Aim: The aim of the present review is to join all the reports available in order to analyze where exactly the knowledge concerning this topic is and what the causes of the controversial data could be.
Methods: We searched electronic databases and bibliographies of selected articles were inspected for further reference.
Results: Diets rich in oleic acid have beneficial effects in inflammatory-related diseases. In addition, a wide range of studies evaluate the effect of oleic acid in different cellular functions thus reporting a potential mechanism for the biological effect of such a fat. However, some controversial data can be found in literature, maybe related to the kind of study or even the dose of the reagent added.
Conclusion: In conclusion, oleic acid could be reported as an anti-inflammatory fatty acid playing a role in the activation of different pathways of immune competent cells.

Key words: Oleic acid. Immune system. Intracellular signaling.


RESUMEN

Introducción: Los ácidos grasos poliinsaturados de la familia n-3 han sido ampliamente caracterizados por su potencial antiinflamatorio. Sin embargo, las evidencias relativas al papel del ácido oleico en el sistema inmune son escasas.
Objetivo: El objetivo de la presente revisión bibliográfica es hacer una recopilación de todos y cada uno de los trabajos publicados a este respecto, al objeto de evaluar dónde se encuentra el conocimiento relativo a esta área y cuáles pueden ser las causas de los resultados contradictorios.
Métodos: Se ha realizado una búsqueda bibliográfica a través de bases de datos electrónicas y las referencias de los artículos de interés han sido utilizadas como fuente de búsquedas más avanzadas.
Resultados: Las dietas ricas en ácido oleico parecen estar asociadas con un beneficio en determinadas patologías de base inflamatoria. Además, un gran número de estudios se han centrado en evaluar el papel que juega tal ácido graso en distintas funciones celulares, argumentando posibles mecanismos que sustentarían los efectos biológicos que se atribuyen a su consumo. Sin embargo, en algunos casos se observan resultados contradictorios que quizá puedan deberse al tipo de estudio desarrollado o incluso a la dosis de ácido con la que se experimenta.
Conclusión: En conclusión, el ácido oleico podría ser presentado como una grasa anti-inflamatoria dado el papel que juega en la activación de distintos mecanismos de señalización de células inmunocompetentes.

Palabras clave: Acido oleico. Sistema inmune. Señalización celular.


Abbreviations
PUFA: Polyunsaturated fatty acid.
OA: Oleic acid.
MUFAs: Monounsaturated fatty acids.
VCAM-1: Vascular cell adhesion molecule-1.
ICAM-1: Intercellular adhesion molecule-1.
NF-kb: Nuclear factor-kappa B.
AA: Arachidonic acid.
EPA: Eicosapentanoic acid.
ROS: Reactive oxygen species.
O2': Superoxide.
PL: Phospholipase.
IP3: Inositol,1,4,5 triphosphate.
DAG: Diacylglycerol.
[Ca2+]i: Intracellular Ca2+ concentration.
PKC: Protein kinase C.
HOSO: Sunflower oil.
NK: Natural killer.
TCR: T cell receptor.
AICD: Activation-induced cell death.
PA: Phosphatidic acid.
DHA: Docosahexaenoic acid.
PIP2: Phosphatidilinositol 4,5 biphosphate.
PI-PLC: Phosphatidylinositol-specific PLC.
polyDAGs: Polyunsaturated DAGs.
monoDAGs: Mono-unsaturated DAGs.
satDAGs: Saturated DAGs.
PI: Phosphatidylinositol.
PS: Phosphatidylserine.
PC: Phosphatidylcholine.
PE: Phosphatidylethanolamine.
SOG: 1-stearoyl-2-oleyl-sn-glycerol.
Stromal Interaction Molecule 1: STIM1.

 

Introduction

The first evidence of the beneficial effect of fatty acids in the human immune system and in inflammatory processes comes from epidemiological studies on Greenland Eskimos, who presented a low rate of several inflammatory-related diseases, which was directly linked to their high levels of n-3 polyunsaturated fatty acid (PUFA) intake. By contrast, the normal diet in various parts of the world contains other unsaturated fatty acids; an example is the widespread use of olive oil, rich in oleic acid (OA), in Mediterranean countries. OA is not only abundant in certain diets but is also the most important constituent of plasma free fatty acids. However, all the scientific effort has been mainly focused on the study of n-3 PUFA, letting the research concerning the effect of the monounsaturated OA be overshadowed. In general, the n-6 PUFA are believed to enhance immune function whereas the n-3 PUFA suppress it. Olive oil has classically been used as a placebo treatment in studies investigating the effects of fish oils on immune function, because monounsaturated fatty acids (MUFAs) were typically regarded as being neutral fatty acids. Nevertheless, there is evidence that MUFA-rich oils have effects which are similar to the effects of fish oils on animals. The studies focusing on the immunomodulatory properties of MUFA have, therefore, reported controversial results, thus the aim of the present review is to join them all in order to analyze where exactly the knowledge concerning this topic is and what the causes of the controversial data cou

Biological effects of oleic acid in the immune system

The Mediterranean Diet appears to be effective in reducing coronary atherosclerosis and the risk of fatal complications like sudden cardiac death.1 Both fish oil and olive oil have been reported to lower plasma fibrinogen in women with high baseline fibrinogen concentrations in a double-blind crossover study.2 Other studies have demonstrated no significant difference between fish-oil supplements and olive oil in preventing restenosis after coronary angioplasty.3 It has been also suggested that the consumption of olive oil may have beneficial effects on rheumatoid arthritis and it has been proposed that the suppressive effect of olive oil on the development of these pathologies may be exerted via an effect on the immune system.4-6

As described above, diets rich in MUFA have been linked with a low prevalence of inflammatory disease. As cells of the immune system are an inherent part of the inflammatory events involved in the development and progression of inflammatory disease, we will describe the influence of OA in different aspects of immune cells.

 

Materials and methods

Search strategy

We consulted studies published in electronic databases such as Pubmed or Medline. The bibliographies of selected articles were inspected for any further reference.

Firstly, we studied the title and abstract of all kind of papers (regular or review papers) with a potential interest to understand the role of oleic acid in immune system. We mostly focused on those related to the mechanisms of action of such a fatty acid at the cellular level. Thus, the main key words used in the search were: "oleic acid", "immune system", "neutrophils", "lymphocytes", "intracellular signaling". Then, the text of the main trials that met the criteria previously mentioned was fully examined to extract the specific data included in the review.

 

Results and discussion

The innate immune system: key role of neutrophils

Effect of oleic acid on neutrophil effector functions

ADHESION, MIGRATION AND PHAGOCITOSIS

Neutrophils play a pivotal role in the defense of the human body against infections. However, overwhelming activation of neutrophils is known to elicit tissue damage. Numerous studies in literature evaluate the effects of OA on leukocyte adhesion. In vitro studies have shown that treatment with micromolar amounts of oleate inhibited the endothelial expression of the vascular cell adhesion molecule-1 (VCAM-1), E-selectin, and the intercellular adhesion molecule-1 (ICAM-1) in several endothelial cells.7-11 Other authors found that OA had no effects on human endothelial cells or on the leukocyte adhesion molecule.12 In any case, a lack of proinflamatory effects of OA has been reported. The previous findings are supported by studies in vivo demonstrating that a meal rich in OA beneficially modulates postprandial soluble ICAM-1 and VCAM-1.13 These data constitute an additional explanation for the beneficial effects of OA, exerted through the inhibition of the very early phenomena in atherogenesis, by modulating endothelial activation through the expression of the gene products involved in leukocyte recruitment, thus confirming the link between OA and cardiovascular disease complications.

In addition, OA has the ability to reduce the inflammatory effects of long-chain saturated fatty acids in human aortic endothelial cells. In this sense, this fatty acid inhibited the stearic acid-induced increase in ICAM-1 expression as well as stearic acid-induced phosphorylation of nuclear factor-kappa B (NF-kB), a transcriptional regulator of ICAM-1.14

Studies in other cell lines have also corroborated the findings above stated. Thus, ICAM-1 expression of non-stimulated and cytokine stimulated Caco-2 cells cultured for 22 days with arachidonic acid (AA), was significantly higher as compared to eicosapentanoic acid (EPA) and OA, suggesting that the replacement of AA by EPA or OA in the colon mucosa might have beneficial effects for inflammatory bowel disease patients.15

By contrast, OA has been also reported to increase the cell surface expression of CD11b and induce the high affinity state of this integrin. This MUFA, through a CD11b-mediated mechanism, induces neutrophil aggregation and neutrophil-endothelial cell attachment.16

Only a few studies are available concerning the effect of OA on leukocyte migration. Ferrante et al. demonstrated that OA was able to inhibit leukocyte-migration but its effect was far from the one exhibite by PUFA.17

Conflicting results can be found concerning other leukocyte functions. Whereas some researchers have reported that OA enhanced neutrophil phagocytic capacity and candidacidal activity,18,19 others show that this fatty acid caused no changes in bactericidal activity and only moderated decreases in phagocytosis and chemotaxis in very high concentrations.20

REACTIVE OXYGEN SPECIES (ROS) PRODUCTION

Once they have arrived at the damaged tissue and engulfed the pathogen, activated neutrophils secrete several cytotoxins, such as superoxide (O2·-), the precursor of other ROS, granule proteases and bioactive lipids.21,22 ROS production is related to the killing of invading microorganisms, but it can also directly or indirectly cause damage by destroying surrounding tissues. Thus, it is of utmost importance to clarify the effect of fatty acids on neutrophils respiratory burst.

Numerous studies have reported the ability of unsaturated fatty acids to influence ROS production in neutrophils.18 23-32 However, depending on the experimental conditions, fatty acids can inhibit, enhance or even synergize neutrophil activation. For example, some studies reported that C18 fatty acids inhibit ROS generation.31 By contrast, others demonstrated an important interaction between fatty acids and cytokines, showing a markedly augmented amount of superoxide produced in response to fatty acids in TNF-pretreated neutrophils.28 In any case, most of the studies in literature have reported an increased ROS generation by unsaturated fatty acids-stimulated neutrophils. Within these, all reports establish differences between fatty acids in their ability to induce ROS generation in unstimulated neutrophils, suggesting that fatty acids distinctively influence neutrophil function depending on the fatty acid structure. Nevertheless, contradictory observations can be found in this respect, which may be in part the result of the different methods used in the determinations.26

Mechanisms undergoing OA effects

As described in the previous section, numerous studies have shown that unsaturated fatty acids stimulate O2·- release from neutrophils and macrophages. However, the mechanism by which these agents exert its effect is complex and has been the subject of several studies.

Perturbation of neutrophils membrane, either by phagocytosis or by different agents stimulates a number of responses which includes activation of an NADPH oxidase. Stimulation of this oxidase leads to the production of large quantities of ROS. Free fatty acids are known to activate NADPH-oxidase in the cell-free O2·- generating system.32,33 Robinson et al. proposed a mechanism for the fatty acid-induced respiratory burst related, at least in part, to its ability to activate phospholipase A2 (PLA2).34 The endogenous AA liberated by PLA2 action on membrane phospholipids may be in a compartment that effectively activates the NADPH oxidase which is responsible for the respiratory burst. These findings (the direct activation of PLA2 by fatty acids) are consistent with previous reports demonstrating that fatty acid-induced ROS production, and various other biological responses, in leukocytes, are due to the fatty acids themselves rather than to cyclooxygenase or lipoxygenase metabolites.17,35-38 By contrast, the latter reports argue against those suggesting that fatty acids can be metabolized by lipoxygenase to hydroperoxides which are intermediates in the release of O2·-.39,40

Some researchers have detected the expression of a GPCR40 in bovine neutrophils. The same group, hypothesized that OA could modulate bovine neutrophil responses and these responses could be induced by GPR40 activation.41 By contrast, others have suggested that unsaturated fatty acids do not act as other stimulating agents which bind to neutrophils at specific receptors. The latter proposed that they may exert their effects by intercalating into and disordering regions of membranes,42 43 thus affecting proteins involved in O2. production that are associated with those regions.24 According to these authors, a component involved in O2·- production appears to be sensitive to transitions in the bilayer. A potential component of this pathway could be phospholipase C which may be activated by phase transitions in the membrane induced by cis-unsaturated fatty acids.24,44 Inositol,1,4,5 triphosphate (IP3) and diacylglycerol (DAG) are two products of this lipase. The former leads to the increase in intracellular Ca2+ concentration ([Ca2+]i) whereas the latter is the physiological activator of protein kinase C (PKC).45 Both mobilization of intracellular Ca2+ and PKC activation have been postulated as potential mechanisms behind the ROS generation event induced by fatty acids. However, although numerous studies can be found in this respect, contradictory are their results. Table 1

Fatty acids, herein OA, have been reported to alter Ca2+ homeostasis in different immunocompetent cells.46-51 However, the relationship between OA-induced ROS production and Ca2+ mobilization is complex and the precise mechanism of action is still unclear. Some authors have reported a Ca2+ -independent mechanism behind the ROS production.52 By contrast, other studies have shown a link between these two pathways.25,41 Other studies available focus on the role of Ca2+ ion in the extracellular medium, suggesting an inhibitory effect of Ca2+ on the fatty acid-induced O2·- generation, attributed to the ionic interaction between the carboxyl group of the fatty acid and the Ca2+.27

In addition, the involvement of a PKC is also controversial.18,25,52 A role for PKC in the stimulation of neutrophils has been proposed on the basis of the ability of various activators of this enzyme to elicit different cellular responses in neutrophils.53-55 As unsaturated fatty acids stimulate PKC in vitro, a role for fatty acids as "second messengers" in the regulation of O2.- production has been thus proposed.53 By contrast, several reports failed to demonstrate a link between PKC activation and ROS production.18,29 Moreover, there are also findings on the PKC-mediated capacity of some fatty acids to enhance the PMA-stimulated O2.- production at nanomolar concentrations and to depress it at micromolar concentrations.56 Table2

Adaptative immune response: key role of lymphocytes

Effects of OA on lymphocyte functions

OLEIC AND IN T CELL PROFILERATION

Several studies have reported a potential role of MUFA-rich diets on immunomodulatory processes based on its ability to influence the proliferation of immune cells.

Verlengia et al. have reported an in vitro inhibiting effect of OA on the proliferation of Jurkat T cells and a reduction in the production of IL-2 and INF-gamma.57 These findings are further corroborated by minerval, an OA synthetic analogue- which also inhibited proliferation of Jurkat cells.58

Animal studies have reported an inhibition of lymphocyte proliferation in response to a T-cell mitogen. Thus, rats fed for weeks on olive oil diets showed a consistent inhibition of spleen and lymph node lymphocyte proliferation.59,60 Similarly, feeding cashew kernel oil, which is rich in OA, also resulted in an inhibition of rat spleen lymphocyte proliferation compared with feeding them the coconut oil diet.61

Olive oil contains a number of antioxidants, sterols, hydrocarbons and alcohols. In order to elucidate whether the effects reported above were due to OA or to some other component of the oil, Jeffery et al. fed rats for 6 weeks on diets containing 20% by weight of olive oil, safflower oil, or high OA sunflower oil (HOSO), using a low-fat diet containing 2.5% by weight of lipid as a control. The results showed a significantly lower mitogen-stimulated spleen lymphocyte proliferation following olive oil or HOSO feeding as compared with low-fat or safflower oil feeding, these observations indicating that the effects of olive oil feeding are most likely due to OA rather than to other components of olive oil.62

Moreover, the use of such oils results in variation in the levels of several fatty acids together and not only the one under investigation. In this sense, to obtain further information about the immunomodulatory effects of specific dietary fatty acids some investigators performed a controlled study in which one fatty acid was exchanged for another, without altering the levels of other fatty acids in the diet. The nine diets used differed in their contents of palmitic, oleic, linoleic and α-linolenic acids. The proliferation of spleen lymphocytes decreased as the level of OA in the diet increased.63

Other studies on the proliferation induced by fatty acids "per se", showed an enhanced lymphocyte proliferation, IL-2 and TNF-α production, induced by a Rice bran oil diet (rich in linoleic acid) compared to a HOSO.64

Studies in humans are lacking and the results available differ from the findings obtained in laboratory animals above reported. Consumption of a MUFA-rich diet did not affect the proliferative response of cells, either in whole-blood cultures or in peripheral blood mononuclear cells, to the T cell mitogen concanavalin A.65 Such controversial results could be related to the doses tested.

The mechanisms by which OA influences T lymphocytes functions are not clear but this fatty acid seems to be involved in the regulation of cell cycle. Epithelial growth factor receptor (a critical crossroad of multiple receptor pathways which is potentially implicated in the regulation of proliferation and possibly involved in atherogenesis) could be considered a target for unsaturated fatty acids.66 Mata et al. have demonstrated that MUFA diets produce the lowest induction of smooth-muscle-cell entry in the cell cycle when compared with other fatty acid-enriched diets although these authors did not investigate whether or not this effect influences cell division and proliferation.67

OLEIC ACID AND NK CELLS

Natural killer (NK) cells are part of natural rather than specific immunity, since they are not activated by a specific antigen. However, these cells are a subset of lymphocytes, found mainly in the blood and spleen, and that is the reason why they are included in this section. They are derived from the bone marrow but they do not undergo thymic maturation.

Rats fed for weeks on a diet rich in olive oil resulted in a significant suppression of NK cell activity, compared with feeding a low-fat diet or diets containing hydrogenated coconut oil or safflower oil, although it was not as great as that resulting from feeding a diet containing fish oil.68 Jeffery et al. demonstrated once again that the inhibitor effect of olive oil feeding on NK cell proliferation is most likely due to OA rather than to other components of olive oil.62 In addition, other reports have focus on studying the single effect attributable to OA itself finding a significant negative linear relationship between the OA content of the diet and NK cell activity, suggesting that dietary OA causes diminished NK cell activity.63

However, as happened in lymphocytes proliferation, studies developed in middle-aged men, have given controversial results. These subjects increased their OA intake at the expense of saturated fatty acids and after a month there was a non-significant reduction in NK cell activity.65 Once again, the differences observed between animal and human studies are likely to be due to the extreme doses added in laboratory research.

APOPTOTIC EFFECTS OF OA IN T CELLS

Programmed cell death or apoptosis is a physiological process to get rid of non-functional or surplus cells. With regard to T lymphocytes, this process make possible to eliminate those lymphocytes having a T cell receptor (TCR) that recognize their own antigens (self tolerance) during thymic maduration. Equally, once the attack has been neutralized, the immune system eliminates the excess of T lymphocytes activated in order to restore "the pool" of these cells. Such two pathways for the removal of T lymphocytes, are developed trough a kind of apoptosis known as "AICD" (activation-induced cell death).69-71 Both autoimmunity, caused by recognition of self antigens, and inmunopatology, caused by a senseless presence of excessive amount of T lymphocytes activated, can be prevented by AICD.

Human lymphocytes, Jurkat (T lymphocyte) and Raji (B lymphocyte) cells, show morphological features of apoptosis after exposure to OA.72,73 In addition, Llado et al. reported for the first time that minerval, an OA synthetic analog, also induced apoptosis in Jurkat T cells.58 Thymocytes are also affected by PUFAs treatment, but no studies have been developed with OA.74

The mechanism of cell death induced by these fatty acids seems to be related to mitochondrial depolarization and ROS production or caspase 3 and 6 activities production. Moreover, evidence is presented that OA is less toxic to human lymphocytes than linoleic acid so OA may offer an immunologically less harmful alternative to linoleic acid for parenteral and enteral diets preparation.72,73,75,76

As previously reviewed, OA could be considered a potential immunomodulatory fat. However, the mechanisms by which this fatty acid exerts its effects are still unclear and available evidences will be described in next section.

Mechanism involved in the immunomodulatory effects of OA

The main target of unsaturated fatty acids is the membrane where they are going to be incorporated into the phospholipids. Once incorporated, these fatty acids could affect cellular function directly or indirectly. In fact, several hypotheses concerning the mechanisms of action of unsaturated fatty acids have been postulated:

- Increase in membrane fluidity which improves membrane-protein interaction and modulates signal transduction.77

- Decrease in AA content in membrane phospholipids and therefore, arachidonic-derived eicosa-noids, known as proinflammatory agents.78,79

- Improvements of oxidant status as MUFAs are less sensitive to lipid peroxidation and ROS production.80

- Modulation of intracellular pathways that play a central role in cell activation.81-85

- Modulation of gene expression involved in cytokine production.57

LIPID SIGNALING MODULATION

Membrane phospholipids are involved in the signal transduction pathways started out of the cell. Dietary unsaturated fatty acids or cells treatment with fatty acids, leads to their esterification in sn-2 position of membrane phospholipids. Once incorporated in the phospholipids, they can be hydrolyzed by several phospholipases. Phospholipases A1 and A2 (PLA1 and PLA2) lead to the release of those fatty acids present in sn-1 and sn-2 position of the glycerol, respectively, and to the subsequent lysophospholipides generation. Phospholipase C (PLC) hydrolyzes PIP2 releasing also two second messengers: DAG and IP3. Phospholipase D (PLD) acts mainly on phosphatidilcoline (PC) and leads to the production of phosphatidic acid (PA), which is rapidly hydrolyzed to form DAG. Additionally, PA can also activate PLC and increase the affinity of the enzyme for the subtract.86,87 As PA is the result of the hydrolysis of PC by PLD, the PLD activation can also lead to the activation of PLC.

Stearic acid and palmitic acid are almost exclusively located at the sn-1 position of the different glycerophospholipids, with the majority of OA and other unsaturated fatty acids found at the sn-2 position.88,89 Thus, n-9 MUFA taken by the diet will result in its incorporation at sn-2 position of membrane phospholipids instead of AA. Depending on the action of different phospholipases, it will affect intracellular signaling pathways both as a free OA (under the action of PLA2)90,91 or as a DAG-containing OA (under the action of PLC and PLD).92

PHOSPHOLIPASE A2

Phospholipase A2 enzymes catalyze the hydrolysis of ester bonds at the sn-2 position of membrane phospholipids and simultaneously release fatty acids, such as AA and lysophospholipids.93,94 The hydrolysis of phospholipids by PLA2 is a key phase in the regulation of inflammatory processes as AA release leads to the production of eicosanoids.95 Stimulus activation of PLA2 can release OA as well, a non-proinflammatory-mediator.96

Several studies have demonstrated that unsaturated fatty acids, including OA, can inhibit both secretion and activity of PLA297,98 results suggesting anti-inflammatory roles for OA during the inflammatory reaction, as AA release and thus subsequent products from its metabolism are inhibited. By contrast, other studies claim a non-significant enhancement of PLA2 activity in the presence of OA, compared with a suppression effect of EPA or docosahexaenoic acid (DHA) or an enhanced activity induced by linoleic acid.99

Furthermore, unsaturated fatty acids are reported to be capable of regulating the enzymatic system involved in their own release. In this sense, Kambe et al. have reported that cells incubated with exogenous unsaturated fatty acids in the presence of IL-1 and serum, showed a significant increase in AA release. OA proved to be the one which induced AA release in a lower amount; AA and linoleic acid were more potent.79

PHOSPHOLIPASE C

Phosphatidylinositol-specific PLC (PI-PLC) plays an important role in cell signal transduction. PLC can hydrolyse phosphatidilinositol 4,5 biphosphate (PIP2) into two second messengers: IP3 and DAG. These messengers then promote the release of Ca2+ from intracellular stores, and the activation of PKC, respectively.

A number of studies have reported the effect of fatty acids on PLC activity. Unsaturated fatty acids, such as OA, but not saturated ones, block signal transduction by interfering with receptor-PLC or PLC-substrate interaction without preventing ligand binding.100 Sanderson and Calder developed a study with rats feeding them on diets with different oils and they showed that the level of PIP2 in spleen lymphocytes was unaffected by diet. However, an olive oil diet (as well as a fish oil one) significantly decreased the concentration of IP3 in stimulated lymphocytes. However, in stimulated lymphocytes, the phosphorylation state of the enzyme, as well as that of a range of other proteins, was decreased following feeding on olive oil diets (and fish oil ones).101

By contrast, some researchers showed that addition of OA to rat microsomal fractions stimulates specifically the hydrolysis of membrane PI.102 However, these unsaturated fatty acids do not directly affect PIP2-hydrolysis activities of various PLC isozymes. Hwang et al. demonstrated that unsaturated fatty acids, such as OA, enhance the activation of PLC by tau, a protein known to activate phospholipase.103 In addition, AHNAK, another protein, in combination with unsaturated fatty acids, such as OA, can activate PLC. These authors did not find an activator effect of the fatty acids alone either.104

Other researchers have focused on studying the effect of the incorporation of different fatty acids in inositol lipids. These results showed that fatty acids induced modifications in PLC activity. PIP2-PLC activities reached a maximum when inositol lipids containing OA became more abundant than normal.105

PHOSPHOLIPASE D

Phospholipase D is widely distributed in mammalian cells and is implicated in a variety of physiological processes that reveal it to be a member of the signal transducing phospholipases. PLD is an enzyme which is located in the plasma membrane, mainly in caveoles, membrane specialized domains rich in sphingolipids and cholesterol "lipid rafts".106 It catalyzes the hydrolysis of PC to form PA, releasing the soluble choline head-group into the cytosol. PA is extremely short lived and is rapidly hydrolysed by the enzyme PA phosphohydrolase to form DAG.

Two families of PLD can be distinguished depending on their sensitivity to PIP2. Some PLD can be activated only in the presence of such a lipid (PLD PIP2-sensitives), while others do not need it (PLD-PIP2-insensitives).

Whereas PLD PIP2-sensitives are activated by several proteins,107-110 the PLD PIP2 -insensitives are activated by OA111 and therefore known as PLD-oleate dependent.112 This type of PLD activity is drastically increased during apoptosis of Jurkat T cells. In fact, PLD activation in lymphocytes T is related to antiproli-ferative and apoptotic signals. Such results suggest the possibility that PLD plays roles in differentiation, survival and apoptosis in mammalian cells.112,113

These findings could be of physiological relevance when studying the mechanisms of OA acid induced apoptosis in T cells.

DLACYLGLYCEROL

A diglyceride, or a diacylglycerol, is a glyceride consisting of two fatty acid chains covalently bonded to a glycerol molecule through ester linkages. In mammalian cells there are, at least, 50 structurally distinct molecular species of sn-1,2-diacylglycerol,114,115 whose fatty acyl groups can be polyunsaturated, mono-unsaturated or saturated. When cells are stimulated by appropriate agonists, PIP2 hydrolysis catalysed by PIP2-PLC becomes an important source of new DAG that are able to activate PKC. Cell stimulation also commonly activates PLD-catalysed PC hydrolysis. The PA generated by PLD is thought to be an intracellular signal whose action also results in DAG production.

Phosphatidilinositol 4,5 biphosphate and PC (the precursors of DAG) typically have very different fatty-acyl complements and their hydrolysis yields very different DAG. Inositol lipids, which make up 5-10% of the total phospholipid content of most mammalian cells, are mainly polyunsaturated: 30-80% of total phosphoinositide is typically the sn-1-stearoyl-2-arachidonyl species.114-118 PC is much more abundant, making up around 30-50% of total mammalian cell phospholipids. In many cells, PC predominantly contains saturated and mono-unsaturated fatty acids; relatively few PC species are polyunsaturated.114-118

Dyacylglycerol production in stimulated cells is often biphasic: there is an initial rapid rise in DAG concentration and then, a slower accumulation that can be sustained for an hour or more. Polyunsaturated DAGs (polyDAGs) -with a saturated or mono-unsaturated 1-acyl group and a polyunsaturated 2-acyl group- predominate during the initial phase; however, they are largely restricted to a few polyDAG species, notably 1-stearoyl-2-arachidonyl-DAG. During the sustained phase, the concentrations of a broad range of mono-unsaturated DAGs (monoDAGs) and saturated DAGs (satDAGs) rise, and these are accompanied by smaller amounts of polyDAGs. The initial polyDAGs are mainly products of PIP2 hydrolysis, whereas the mono/satDAGs generated in the sustained phase are derived predominantly (through dephosphorylation of PLD generated PA) from PC.114,115,119

Moreover, fatty acid composition of membrane phospholipids can be further influenced by the diet. Previous studies have shown that in T-cells exposed to an OA-rich medium, the total monounsaturated fatty acyl content was increased by 130% in PI, 100% in phosphatidylserine (PS), 160% in phosphatidylcholine and 180% in phosp-hatidylethanolamine (PE).120 Other researchers showed that palmitic and oleic acids were found preferentially incorporated into PC and the majority of the highly unsaturated fatty acid, AA, was incorporated into both PE and the PI-PS group.121 In any case, during the dietary intake of n-9 fatty acids, the cells will be enriched with this fatty acid and a cell activation via PLC- or PLD-pathway may give rise to the production of DAG in the conjugated form of 1-stearoyl-2-oleyl-sn-glycerol (SOG), as it has been previously reported in human Jurkat T-cells.122

Several reports have proposed that the fatty acids that make up the phosphoinositides, function as intracellular modulators of the activity of certain enzymes.105 The best characterized function of receptor-stimulated DAG production is activation of PKC, although not all DAG species are capable of activating this enzyme.123 In this sense, Madani et al. have demonstrated differences in modulating the activity of several isoforms of PKC by two polyDAG: SEG (DAG-containing EPA) and SDG (DAG-containing DHA).124 Additionally, some channels in the membrane that can be directly activated by DAG have been recently identified (see below).

ELCOSANOIDS PRODUCTION

Oleic acid could exert its antiinflamatory effect by influencing AA metabolism. This PUFA is in turn the precursor of the eicosanoids proinflamatory agents. Thus, competitive substitution of membrane arachidonate by n-9 fats could be associated with modifications in the rate of arachidonic-derived metabolites release, suggesting that as a mechanism for the OA induced-immune modulatory pathways. In this sense, evidences show that the n-9 diet significantly decreased both mucosal arachidonate concentrations and AA:EPA ratio as compared with the n-6 diet, which may in part account for the observed beneficial effect of olive oil.125

SIGNALING PATHWAYS MODULATED BY OLEIC ACID

Fatty acids can modulate several signaling pathways, from the antigen presentation to the T cell proliferation. In the present review we will focus on the role of OA in Ca2+ signals.

Ca2+ SIGNALING

Most of the studies available in immunocompetent cells evaluate the effect of polyunsaturated n-6 and n-3 on Ca2+ signaling. Although OA has been reported to alter CA2+ homeostasis in several cell lines,126,127 little is known about its effects concerning T cells.

Oleic acid has been demonstrated to induce increases in [Ca2+]i in several cell lines.91,126-128 By contrast, other studies have demonstrated that OA inhibit the agonist-induced extracellular Ca2+ influx in T cells,47,129,130 and other cell types.131,132 With regards to the mechanisms behind these alterations in Ca2+ homeostasis, Gamberuci et al. have demonstrated that OA discharges almost all the thapsi-gargin-sensitive Ca2+ pool. In addition, neither pretreatment of the cell with the G-protein interfering agent pertussis toxin nor with a phospholipase inhibitor prevents the induction of Ca2+ mobilization by OA.84 Researching further the mechanisms by which such Ca2+ -mobilizing effects occur, these authors suggest that an increased Ca2+ efflux from thapsigargine-sensitive pool is more likely to occur than an inhibition of active Ca2+ transport into the same pool. However, although these data supported those reported by Chow and Jondal for PUFAs, these researchers did not find similar results concerning OA.46

In addition, Ca2+ channels such as TRPC3, TRPC6 and TRPC7 have been reported to be activated by the exogenous addition of DAG.133-136 Aires et al. have studied the effect of a DAG-containing DHA in the activation of TRPC channels.137 Results from our group have recently reported a role of a DAG containing OA in the activation of TRPC6 and TRPC3 channels in human Jurkat T cells.138

OTHER MECHANISMS

One emerging view is that the potential mechanism by which PUFAs influence immune cell function could be related to its ability to modulate lipid rafts. Lipid rafts are postulated to be specialized lipid microdomains rich in sphingolipids and cholesterol. They are involved in serving as platforms for cellular signaling events.139-142 For instance, the ER Ca2+ sensor STIM1 (Stromal Interaction Molecule 1) has been reported to interact with Orai and TRPC in these specific parts of the membrane, leading to the extracellular Ca2+ entry.143,144 It has been recently described that the disruption of lipid rafts avoids the interaction between STIM1 and Orai1.145,146

The supposed stability of lipid rafts is attributed to the favorable interaction between the amide of the sphingosine backbone and the hydroxyl of an adjacent sphingolipid as well as hydrogen bonding between the 3-OH of cholesterol and the sphingosine amide. Saturated acyl chains are thought to promote the formation of rafts since they are more extended than unsaturated chains and packed well amongst themselves and with cholesterol. Previous reports have shown that PUFA can modify the composition of lipid rafts.147-151 In this sense, several researchers have demonstrated that T cells submitted to PUFA treatments, results in an inhibition of different signal transduction pathways.152-156 However, most of the studies in literature involved PUFA and OA disruption of lipid rafts is still unclear.

Oleic acid in clinical nutrition

As we have reported above, diets containing a high amount of olive oil in experimental animals, produce a suppression of lymphocyte proliferation, an inhibition of cytokine production and a reduction in NK cell activity.57,59,60,68 Despite these alterations in immune functions, it has been reported that olive oil-rich diets are not as immunosuppressive as fish oil diets. An important aspect in inmunonutrition is focused on the relationship between fats, the immune system and host resistance to infection, particularly when these nutrients are supplied to patients at risk of sepsis. Different studies have determined that olive oil-rich diets do not impair the host resistance to infection.157,158 Therefore, olive oil constitutes a suitable fat that may be applied in clinical nutrition and administered to critically ill patients.

The current knowledge of these aspects and the beneficial effect attributed to an olive oil-rich parenteral emulsion (Clinoleic) have been recently reported elsewhere.157-160

 

References

1. De Lorgeril M. Traditional Mediterranean diet in prevention of myocardial infartion and cancers. Medecine et Longevite 2010; 2 (1): 40-5.         [ Links ]

2. Oosthuizen W, Vorster HH, Jerling JC, Barnard HC, Smuts CM, Silvis N et al. Both fish oil and olive oil lowered plasma fibrinogen in women with high baseline fibrinogen levels. Thromb Haemost 1994; 72 (4): 557-62.         [ Links ]

3. Dehmer GJ, Popma JJ, van den Berg EK, Eichhorn EJ, Prewitt JB, Campbell WB et al. Reduction in the rate of early restenosis after coronary angioplasty by a diet supplemented with n-3 fatty acids. N Engl J Med 1988; 319 (12): 733-40.         [ Links ]

4. Kremer JM, Lawrence DA, Jubiz W, DiGiacomo R, Rynes R, Bartholomew LE et al. Dietary fish oil and olive oil supplementation in patients with rheumatoid arthritis. Clinical and immunologic effects. Arthritis Rheum 1990; 33 (6): 810-20.         [ Links ]

5. Linos A, Kaklamanis E, Kontomerkos A, Koumantaki Y, Gazi S, Vaiopoulos G et al. The effect of olive oil and fish consumption on rheumatoid arthritis - a case control study. Scand J Rheumatol 1991; 20 (6): 419-26.         [ Links ]

6. Berbert AA, Kondo CR, Almendra CL, Matsuo T, Dichi I. Supplementation of fish oil and olive oil in patients with rheumatoid arthritis. Nutrition 2005; 21 (2): 131-6.         [ Links ]

7. Carluccio MA, Massaro M, Bonfrate C, Siculella L, Maffia M, Nicolardi G et al. Oleic acid inhibits endothelial activation: A direct vascular antiatherogenic mechanism of a nutritional component in the mediterranean diet. Arterioscler Thromb Vasc Biol 1999; 19 (2): 220-8.         [ Links ]

8. Massaro M, Carluccio MA, De Caterina R. Direct vascular antiatherogenic effects of oleic acid: a clue to the cardioprotective effects of the Mediterranean diet. Cardiologia 1999; 44 (6):507-13.         [ Links ]

9. Sanadgol N, Mostafaie A, Bahrami G, Mansouri K, Ghanbari F, Bidmeshkipour A. Elaidic acid sustains LPS and TNF-alpha induced ICAM-1 and VCAM-I expression on human bone marrow endothelial cells (HBMEC). Clin Biochem 2010; 43(12): 968-72.         [ Links ]

10. Christon RA. Mechanisms of action of dietary fatty acids in regulating the activation of vascular endothelial cells during atherogenesis. Nutr Rev 2003; 61 (8): 272-9.         [ Links ]

11. De Caterina R, Libby P. Control of endothelial leukocyte adhesion molecules by fatty acids. Lipids 1996; 31: S57-63.         [ Links ]

12. Holthe MR, Andersson Y, Lyberg T. Lack of proinflammatory effects of free fatty acids on human umbilical cord vein endothelial cells and leukocytes. Acta Obstet Gynecol Scand 2005; 84 (7): 672-8.         [ Links ]

13. Pacheco YM, López S, Bermúdez B, Abia R, Villar J, Muriana FJ. A meal rich in oleic acid beneficially modulates postprandial sICAM-1 and sVCAM-1 in normotensive and hypertensive hyper-triglyceridemic subjects. J Nutr Biochem 2008; 19 (3): 200-5.         [ Links ]

14. Harvey KA, Walker CL, Xu Z, Whitley P, Pavlina TM, Hise M et al. Oleic acid inhibits stearic acid-induced inhibition of cell growth and pro-inflammatory responses in human aortic endothelial cells. J Lipid Res 2010; 51 (12): 3470-80.         [ Links ]

15. Ramakers JD, Mensink RP, Schaart G, Plat J. Arachidonic acid but not eicosapentaenoic acid (EPA) and oleic acid activates NF-kappaB and elevates ICAM-1 expression in Caco-2 cells. Lipids 2007; 42 (8): 687-98.         [ Links ]

16. Mastrangelo AM, Jeitner TM, Eaton JW. Oleic acid increases cell surface expression and activity of CD11b on human neutrophils. J Immunol 1998; 161 (8): 4268-75.         [ Links ]

17. Ferrante A, Goh D, Harvey DP, Robinson BS, Hii CS, Bates EJ et al. Neutrophil migration inhibitory properties of polyunsaturated fatty acids. The role of fatty acid structure, metabolism, and possible second messenger systems. J Clin Invest 1994; 93 (3): 1063-70.         [ Links ]

18. Padovese R, Curi R. Modulation of rat neutrophil function in vitro by cis- and trans-MUFA. Br J Nutr 2009; 101 (9): 1351-9.         [ Links ]

19. Martins de Lima-Salgado T, Coccuzzo Sampaio S, Cury-Boaventura MF, Curi R. Modulatory effect of fatty acids on fungicidal activity, respiratory burst and TNF-alpha and IL-6 production in J774 murine macrophages. Br J Nutr 2011; 105 (8):1173-9.         [ Links ]

20. Hawley HP, Gordon GB. The effects of long chain free fatty acids on human neutrophil function and structure. Lab Invest 1976; 34 (2): 216-22.         [ Links ]

21. Hampton MB, Winterbourn CC. Modification of neutrophil oxidant production with diphenyleneiodonium and its effect on bacterial killing. Free Radic Biol Med 1995; 18 (4): 633-9.         [ Links ]

22. Nathan C. Neutrophils and immunity: challenges and opportunities. Nat Rev Immunol 2006; 6 (3): 173-82.         [ Links ]

23. Badwey JA, Curnutte JT, Karnovsky ML. cis-Polyunsaturated fatty acids induce high levels of superoxide production by human neutrophils. J Biol Chem 1981; 256 (24): 12640-3.         [ Links ]

24. Badwey JA, Curnutte JT, Robinson JM, Berde CB, Karnovsky MJ, Karnovsky ML. Effects of free fatty acids on release of superoxide and on change of shape by human neutrophils. Reversibility by albumin. J Biol Chem 1984; 259 (12): 7870-7.         [ Links ]

25. Morimoto YM, Sato E, Nobori K, Takahashi R, Utsumi K. Effect of calcium ion on fatty acid-induced generation of superoxide in guinea pig neutrophils. Cell Struct Funct 1986; 11 (2): 143-55.         [ Links ]

26. Hatanaka E, Levada-Pires AC, Pithon-Curi TC, Curi R. Systematic study on ROS production induced by oleic, linoleic, and gamma-linolenic acids in human and rat neutrophils. Free Radic Biol Med 2006; 41 (7): 1124-32.         [ Links ]

27. Yamaguchi T, Kaneda M, Kakinuma K. Effect of saturated and unsaturated fatty acids on the oxidative metabolism of human neutrophils. The role of calcium ion in the extracellular medium. Biochim Biophys Acta 1986; 861 (3): 440-6.         [ Links ]

28. Li Y, Ferrante A, Poulos A, Harvey DP. Neutrophil oxygen radical generation. Synergistic responses to tumor necrosis factor and mono/polyunsaturated fatty acids. J Clin Invest 1996; 97 (7): 1605-9.         [ Links ]

29. Hardy SJ, Robinson BS, Ferrante A, Hii CS, Johnson DW, Poulos A et al. Polyenoic very-long-chain fatty acids mobilize intracellular calcium from a thapsigargin-insensitive pool in human neutrophils. The relationship between Ca2+ mobilization and superoxide production induced by long- and very-long-chain fatty acids. Biochem J 1995; 311: 689-97.         [ Links ]

30. Juttner B, Kroplin J, Coldewey SM, Witt L, Osthaus WA, Weilbach C et al. Unsaturated long-chain fatty acids induce the respiratory burst of human neutrophils and monocytes in whole blood. Nutr Metab (Lond) 2008; 5: 19.         [ Links ]

31. Hwang TL, Su YC, Chang HL, Leu YL, Chung PJ, Kuo LM et al. Suppression of superoxide anion and elastase release by C18 unsaturated fatty acids in human neutrophils. J Lipid Res 2009; 50 (7): 1395-408.         [ Links ]

32. Tanaka T, Makino R, Iizuka T, Ishimura Y, Kanegasaki S. Activation by saturated and monounsaturated fatty acids of the O2- -generating system in a cell-free preparation from neutrophils. J Biol Chem 1988; 263 (27): 13670-6.         [ Links ]

33. Seifert R, Schultz G. Fatty-acid-induced activation of NADPH oxidase in plasma membranes of human neutrophils depends on neutrophil cytosol and is potentiated by stable guanine nucleotides. Eur J Biochem 1987; 162 (3): 563-9.         [ Links ]

34. Robinson BS, Hii CS, Ferrante A. Activation of phospholipase A2 in human neutrophils by polyunsaturated fatty acids and its role in stimulation of superoxide production. Biochem J 1998; 336: 611-7.         [ Links ]

35. McPhail LC, Shirley PS, Clayton CC, Snyderman R. Activation of the respiratory burst enzyme from human neutrophils in a cell-free system. Evidence for a soluble cofactor. J Clin Invest 1985; 75 (5): 1735-9.         [ Links ]

36. Smith RJ, Sam LM, Justen JM, Leach KL, Epps DE. Human polymorphonuclear neutrophil activation with arachidonic acid. Br J Pharmacol 1987; 91 (3): 641-9.         [ Links ]

37. Bates EJ, Ferrante A, Harvey DP, Nandoskar M, Poulos A. Docosahexanoic acid (22:6, n-3) but not eicosapentaenoic acid (20:5, n-3) can induce neutrophil-mediated injury of cultured endothelial cells: involvement of neutrophil elastase. J Leukoc Biol 1993; 54 (6): 590-8.         [ Links ]

38. Bromberg Y, Pick E. Unsaturated fatty acids as second messengers of superoxide generation by macrophages. Cell Immunol 1983; 79 (2): 240-52.         [ Links ]

39. Smith RL, Weidemann MJ. Reactive oxygen production associated with arachidonic acid metabolism by peritoneal macrophages. Biochem Biophys Res Commun 1980; 97 (3): 973-80.         [ Links ]

40. Yoshimoto S, Yoshimoto T, Tsubura E. Arachidonic acid-induced chemiluminescence of human polymorphonuclear leukocytes. Biochem Biophys Res Commun 1982; 107 (3): 779-84.         [ Links ]

41. Hidalgo MA, Nahuelpan C, Manosalva C, Jara E, Carretta MD, Conejeros I et al. Oleic acid induces intracellular calcium mobilization, MAPK phosphorylation, superoxide production and granule release in bovine neutrophils. Biochem Biophys Res Commun 2011; 409 (2): 280-6.         [ Links ]

42. Usher JR, Epand RM, Papahadjopoulos D. The effect of free fatty acids on the thermotropic phase transition of dimyristoyl glycerophosphocholine. Chem Phys Lipids 1978; 22 (3): 245-53.         [ Links ]

43. Klausner RD, Kleinfeld AM, Hoover RL, Karnovsky MJ. Lipid domains in membranes. Evidence derived from structural perturbations induced by free fatty acids and lifetime heterogeneity analysis. J Biol Chem 1980; 255 (4): 1286-95.         [ Links ]

44. Takenawa T, Nagai Y. Effect of unsaturated fatty acids and Ca2+ on phosphatidylinositol synthesis and breakdown. J Biochem 1982; 91 (3): 793-9.         [ Links ]

45. Kishimoto A, Takai Y, Mori T, Kikkawa U, Nishizuka Y. Activation of calcium and phospholipid-dependent protein kinase by diacylglycerol, its possible relation to phosphatidylinositol turnover. J Biol Chem 1980; 255 (6): 2273-6.         [ Links ]

46. Chow SC, Jondal M. Polyunsaturated free fatty acids stimulate an increase in cytosolic Ca2+ by mobilizing the inositol 1,4,5-trisphosphate-sensitive Ca2+ pool in T cells through a mechanism independent of phosphoinositide turnover. J Biol Chem 1990; 265 (2): 902-7.         [ Links ]

47. Chow SC, Ansotegui IJ, Jondal M. Inhibition of receptor-mediated calcium influx in T cells by unsaturated non-esterified fatty acids. Biochem J 1990; 267 (3): 727-32.         [ Links ]

48. Randriamampita C, Trautmann A. Arachidonic acid activates Ca2+ extrusion in macrophages. J Biol Chem 1990; 265 (30):18059-62.         [ Links ]

49. Breittmayer JP, Pelassy C, Cousin JL, Bernard A, Aussel C. The inhibition by fatty acids of receptor-mediated calcium movements in Jurkat T-cells is due to increased calcium extrusion. J Biol Chem 1993; 268 (28): 20812-7.         [ Links ]

50. Carrillo C, Cavia MM, Alonso-Torre SR. Oleic Acid Versus Linoleic and -Linolenic Acid. Different Effects on Ca2+ Signaling in Rat Thymocytes. Cell Physiol Biochem 2011; 27: 373-80.         [ Links ]

51. Carrillo C, Cavia MM, Roelofs HM, Wanten G, Alonso-Torre SR. Activation of human neutrophils by oleic acid involves the production of reactive oxygen species and a rise in cytosolic calcium concentration: a comparison with n-6 polyunsaturated fatty acids. Cell Physiol Biochem 2011; 28 (2): 329-38.         [ Links ]

52. Hardy SJ, Robinson BS, Ferrante A, Hii CS, Johnson DW, Poulos A et al. Polyenoic very-long-chain fatty acids mobilize intracellular calcium from a thapsigargin-insensitive pool in human neutrophils. The relationship between Ca2+ mobilization and superoxide production induced by long- and very-long-chain fatty acids. Biochem J 1995; 311: 689-97.         [ Links ]

53. McPhail LC, Clayton CC, Snyderman R. A potential second messenger role for unsaturated fatty acids: activation of Ca2+ -dependent protein kinase. Science 1984; 224 (4649): 622-5.         [ Links ]

54. Helfman DM, Appelbaum BD, Vogler WR, Kuo JF. Phospholipid-sensitive Ca2+ -dependent protein kinase and its substrates in human neutrophils. Biochem Biophys Res Commun 1983; 111(3): 847-53.         [ Links ]

55. Fujita I, Irita K, Takeshige K, Minakami S. Diacylglycerol, 1-oleoyl-2-acetyl-glycerol, stimulates superoxide-generation from human neutrophils. Biochem Biophys Res Commun 1984; 120 (2): 318-24.         [ Links ]

56. Kadri-Hassani N, Leger CL, Descomps B. The fatty acid bimodal action on superoxide anion production by human adherent monocytes under phorbol 12-myristate 13-acetate or diacylglycerol activation can be explained by the modulation of protein kinase C and p47phox translocation. J Biol Chem 1995; 270 (25): 15111-8.         [ Links ]

57. Verlengia R, Gorjao R, Kanunfre CC, Bordin S, de Lima TM, Curi R. Effect of arachidonic acid on proliferation, cytokines production and pleiotropic genes expression in Jurkat cells - a comparison with oleic acid. Life Sci 2003; 73 (23): 2939-51.         [ Links ]

58. Llado V, Gutiérrez A, Martínez J, Casas J, Teres S, Higuera M et al. Minerval induces apoptosis in Jurkat and other cancer cells. J Cell Mol Med 2010; 14 (3): 659-70.         [ Links ]

59. Yaqoob P, Newsholme EA, Calder PC. The effect of dietary lipid manipulation on rat lymphocyte subsets and proliferation. Immunology 1994; 82 (4): 603-10.         [ Links ]

60. Yaqoob P, Newsholme EA, Calder PC. The effect of fatty acids on leukocyte subsets and proliferation in whole blood. Nutr Res 1995;15:279-87.         [ Links ]

61. Calder PC, Costa-Rosa LF, Curi R. Effects of feeding lipids of different fatty acid compositions upon rat lymphocyte proliferation. Life Sci 1995; 56 (6): 455-63.         [ Links ]

62. Jeffery NM, Yaqoob P, Newsholme EA, Calder PC. The effects of olive oil upon rat serum lipid levels and lymphocyte functions appear to be due to oleic acid. Ann Nutr Metab 1996; 40 (2): 71-80.         [ Links ]

63. Jeffery NM, Cortina M, Newsholme EA, Calder PC. Effects of variations in the proportions of saturated, monounsaturated and polyunsaturated fatty acids in the rat diet on spleen lymphocyte functions. Br J Nutr 1997; 77 (5): 805-23.         [ Links ]

64. Sierra S, Lara-Villoslada F, Olivares M, Jiménez J, Boza J, Xaus J. Increased immune response in mice consuming rice bran oil. Eur J Nutr 2005; 44 (8): 509-16.         [ Links ]

65. Yaqoob P, Knapper JA, Webb DH, Williams CM, Newsholme EA, Calder PC. Effect of olive oil on immune function in middle-aged men. Am J Clin Nutr 1998; 67 (1): 129-35.         [ Links ]

66. Vacaresse N, Lajoie-Mazenc I, Auge N, Suc I, Frisach MF, Salvayre R et al. Activation of epithelial growth factor receptor pathway by unsaturated fatty acids. Circ Res 1999; 85 (10): 892-9.         [ Links ]

67. Mata P, Varela O, Alonso R, Lahoz C, de Oya M, Badimon L. Monounsaturated and polyunsaturated n-6 fatty acid-enriched diets modify LDL oxidation and decrease human coronary smooth muscle cell DNA synthesis. Arterioscler Thromb Vasc Biol 1997; 17 (10): 2088-95.         [ Links ]

68. Yaqoob P, Newsholme EA, Calder PC. Inhibition of natural killer cell activity by dietary lipids. Immunol Lett 1994; 41 (23): 241-7.         [ Links ]

69. Bouillet P, Strasser A. Bax and Bak: back-bone of T cell death. Nat Immunol 2002; 3 (10): 893-4.         [ Links ]

70. Green DR, Droin N, Pinkoski M. Activation-induced cell death in T cells. Immunol Rev 2003; 193: 70-81.         [ Links ]

71. Opferman JT, Korsmeyer SJ. Apoptosis in the development and maintenance of the immune system. Nat Immunol 2003; 4(5): 410-5.         [ Links ]

72. Cury-Boaventura MF, Pompeia C, Curi R. Comparative toxicity of oleic acid and linoleic acid on Jurkat cells. Clin Nutr 2004; 23 (4): 721-32.         [ Links ]

73. Cury-Boaventura MF, Pompeia C, Curi R. Comparative toxicity of oleic acid and linoleic acid on Raji cells. Nutrition 2005; 21 (3): 395-405.         [ Links ]

74. Prasad A, Bloom MS, Carpenter DO. Role of calcium and ROS in cell death induced by polyunsaturated fatty acids in murine thymocytes. J Cell Physiol 2010; 225 (3): 829-36.         [ Links ]

75. Fernanda Cury-Boaventura M, Cristine Kanunfre C, Gorjao R, Martins de Lima T, Curi R. Mechanisms involved in Jurkat cell death induced by oleic and linoleic acids. Clin Nutr 2006; 25(6): 1004-14.         [ Links ]

76. Cury-Boaventura MF, Gorjao R, de Lima TM, Newsholme P, Curi R. Comparative toxicity of oleic and linoleic acid on human lymphocytes. Life Sci 2006; 78 (13): 1448-56.         [ Links ]

77. De Jonge HW, Dekkers DH, Bastiaanse EM, Bezstarosti K, van der Laarse A, Lamers JM. Eicosapentaenoic acid incorporation in membrane phospholipids modulates receptor-mediated phospholipase C and membrane fluidity in rat ventricular myocytes in culture. J Mol Cell Cardiol 1996; 28 (5): 1097-108.         [ Links ]

78. Corsetto PA, Montorfano G, Zava S, Jovenitti IE, Cremona A, Berra B et al. Effects of n-3 PUFAs on breast cancer cells through their incorporation in plasma membrane. Lipids Health Dis 2011; 10: 73.         [ Links ]

79. Kambe T, Murakami M, Kudo I. Polyunsaturated fatty acids potentiate interleukin-1-stimulated arachidonic acid release by cells overexpressing type IIA secretory phospholipase A2. FEBS Lett 1999; 453 (1-2): 81-4.         [ Links ]

80. Nakbi A, Tayeb W, Dabbou S, Issaoui M, Grissa AK, Attia N et al. Dietary olive oil effect on antioxidant status and fatty acid profile in the erythrocyte of 2,4-D- exposed rats. Lipids Health Dis 9: 89.         [ Links ]

81. Aires V, Hichami A, Filomenko R, Ple A, Rebe C, Bettaieb A et al. Docosahexaenoic acid induces increases in [Ca2+]i via inositol 1,4,5-triphosphate production and activates protein kinase C gamma and -delta via phosphatidylserine binding site: implication in apoptosis in U937 cells. Mol Pharmacol 2007; 72 (6):1545-56.         [ Links ]

82. Bonin A, Khan NA. Regulation of calcium signalling by doco-sahexaenoic acid in human T-cells. Implication of CRAC channels. J Lipid Res 2000; 41 (2): 277-84.         [ Links ]

83. De Jonge HW, Dekkers DH, Lamers JM. Polyunsaturated fatty acids and signalling via phospholipase C-beta and A2 in myocardium. Mol Cell Biochem 1996; 157 (1-2): 199-210.         [ Links ]

84. Gamberucci A, Fulceri R, Bygrave FL, Benedetti A. Unsaturated fatty acids mobilize intracellular calcium independent of IP3 generation and VIA insertion at the plasma membrane. Biochem Biophys Res Commun 1997; 241 (2): 312-6.         [ Links ]

85. Yaqoob P. Fatty acids as gatekeepers of immune cell regulation. Trends Immunol 2003; 24 (12): 639-45.         [ Links ]

86. Jones GA, Carpenter G. The regulation of phospholipase C-gamma 1 by phosphatidic acid. Assessment of kinetic parameters. J Biol Chem 1993; 268 (28): 20845-50.         [ Links ]

87. Zhou C, Horstman D, Carpenter G, Roberts MF. Action of phosphatidylinositol-specific phospholipase Cgamma1 on soluble and micellar substrates. Separating effects on catalysis from modulation of the surface. J Biol Chem 1999; 274 (5): 2786-93.         [ Links ]

88. Mahadevappa VG, Holub BJ. The molecular species composition of individual diacyl phospholipids in human platelets. Biochim Biophys Acta 1982; 713 (1): 73-9.         [ Links ]

89. MacDonald JI, Sprecher H. Phospholipid fatty acid remodeling in mammalian cells. Biochim Biophys Acta 1991; 1084 (2): 105-21.         [ Links ]

90. Mircheva K, Minkov I, Ivanova T, Panaiotov I, Proust JE, Verger R. Comparative study of lipolysis by PLA2 of DOPC substrates organized as monolayers, bilayer vesicles and nanocapsules. Colloids Surf B Biointerfaces 2008; 67 (1): 107-14.         [ Links ]

91. Siafaka-Kapadai A, Hanahan DJ, Javors MA. Oleic acid-induced Ca2+ mobilization in human platelets: is oleic acid an intracellular messenger? J Lipid Mediat Cell Signal 1997; 15 (3): 215-32.         [ Links ]

92. Pettitt TR, Martin A, Horton T, Liossis C, Lord JM, Wakelam MJ. Diacylglycerol and phosphatidate generated by phospholipases C and D, respectively, have distinct fatty acid compositions and functions. Phospholipase D-derived diacylglycerol does not activate protein kinase C in porcine aortic endothelial cells. J Biol Chem 1997; 272 (28): 17354-9.         [ Links ]

93. Van den Bosch H. Intracellular phospholipases A. Biochim Biophys Acta 1980; 604 (2): 191-246.         [ Links ]

94. Braquet P, Touqui L, Shen TY, Vargaftig BB. Perspectives in platelet-activating factor research. Pharmacol Rev 1987; 39 (2): 97-145.         [ Links ]

95. Serhan CN, Haeggstrom JZ, Leslie CC. Lipid mediator networks in cell signaling: update and impact of cytokines. Faseb J 1996; 10 (10): 1147-58.         [ Links ]

96. Smith JB, Dangelmaier C, Mauco G. Measurement of arachi-donic acid liberation in thrombin-stimulated human platelets. Use of agents that inhibit both the cyclooxygenase and lipoxygenase enzymes. Biochim Biophys Acta 1985; 835 (2): 344-51.         [ Links ]

97. Franson R, Raghupathi R, Fry M, Saal J, Vishwanath B, Ghosh SS et al. Inhibition of human phospholipases A2 by cis-unsaturated fatty acids and oligomers of prostaglandin B1. Adv Exp Med Biol 1990; 279: 219-30.         [ Links ]

98. Alaoui El Azher M, Havet N, Singer M, Dumarey C, Touqui L. Inhibition by unsaturated fatty acids of type II secretory phosp-holipase A2 synthesis in guinea-pig alveolar macrophages evidence for the eicosanoid-independent pathway. Eur J Biochem 2000; 267 (12): 3633-9.         [ Links ]

99. Tappia PS, Man WJ, Grimble RF. Influence of unsaturated fatty acids on the production of tumour necrosis factor and interleukin-6 by rat peritoneal macrophages. Mol Cell Biochem 1995; 143 (2): 89-98.         [ Links ]

100. Casabiell X, Pandiella A, Casanueva FF. Regulation of epidermal-growth-factor-receptor signal transduction by cis-unsaturated fatty acids. Evidence for a protein kinase C-independent mechanism. Biochem J 1991; 278: 679-87.         [ Links ]

101. Sanderson P, Calder PC. Dietary fish oil appears to prevent the activation of phospholipase C-gamma in lymphocytes. Biochim Biophys Acta 1998; 1392 (2-3): 300-8.         [ Links ]

102. Irvine RF, Letcher AJ, Dawson RM. Fatty acid stimulation of membrane phosphatidylinositol hydrolysis by brain phosphatidylinositol phosphodiesterase. Biochem J 1979; 178 (2): 497-500.         [ Links ]

103. Hwang SC, Jhon DY, Bae YS, Kim JH, Rhee SG. Activation of phospholipase C-gamma by the concerted action of tau proteins and arachidonic acid. J Biol Chem 1996; 271 (31): 18342-9.         [ Links ]

104. Sekiya F, Bae YS, Jhon DY, Hwang SC, Rhee SG. AHNAK, a protein that binds and activates phospholipase C-gamma1 in the presence of arachidonic acid. J Biol Chem 1999; 274 (20):13900-7.         [ Links ]

105. Carricaburu V, Fournier B. Phosphoinositide fatty acids regulate phosphatidylinositol 5-kinase, phospholipase C and protein kinase C activities. Eur J Biochem 2001; 268 (5): 1238-49.         [ Links ]

106. McDermott M, Wakelam MJ, Morris AJ. Phospholipase D. Biochem Cell Biol 2004; 82 (1): 225-53.         [ Links ]

107. Hammond SM, Altshuller YM, Sung TC, Rudge SA, Rose K, Engebrecht J, et al. Human ADP-ribosylation factor-activated phosphatidylcholine-specific phospholipase D defines a new and highly conserved gene family. J Biol Chem 1995; 270(50): 29640-3.         [ Links ]

108. Colley WC, Sung TC, Roll R, Jenco J, Hammond SM, Altshuller Y et al. Phospholipase D2, a distinct phospholipase D isoform with novel regulatory properties that provokes cytoskele-tal reorganization. Curr Biol 1997; 7 (3): 191-201.         [ Links ]

109. Kodaki T, Yamashita S. Cloning, expression, and characterization of a novel phospholipase D complementary DNA from rat brain. J Biol Chem 1997; 272 (17): 11408-13.         [ Links ]

110. López I, Arnold RS, Lambeth JD. Cloning and initial characterization of a human phospholipase D2 (hPLD2). ADP-ribosylation factor regulates hPLD2. J Biol Chem 1998; 273 (21):12846-52.         [ Links ]

111. Frohman MA, Sung TC, Morris AJ. Mammalian phospholipase D structure and regulation. Biochim Biophys Acta 1999; 1439 (2): 175-86.         [ Links ]

112. Kasai T, Ohguchi K, Nakashima S, Ito Y, Naganawa T, Kondo N et al. Increased activity of oleate-dependent type phospholipase D during actinomycin D-induced apoptosis in Jurkat T cells. J Immunol 1998; 161 (12): 6469-74.         [ Links ]

113. Nakashima S, Nozawa Y. Possible role of phospholipase D in cellular differentiation and apoptosis. Chem Phys Lipids 1999; 98 (1-2): 153-64.         [ Links ]

114. Pessin MS, Raben DM. Molecular species analysis of 1,2-diglycerides stimulated by alpha-thrombin in cultured fibroblasts. J Biol Chem 1989; 264 (15): 8729-38.         [ Links ]

115. Pettitt TR, Wakelam MJ. Bombesin stimulates distinct time-dependent changes in the sn-1,2-diradylglycerol molecular species profile from Swiss 3T3 fibroblasts as analysed by 3,5-dinitrobenzoyl derivatization and HPLC separation. Biochem J 1993; 289: 487-95.         [ Links ]

116. Holbrook PG, Pannell LK, Murata Y, Daly JW. Molecular species analysis of a product of phospholipase D activation. Phosphatidylethanol is formed from phosphatidylcholine in phorbol ester- and bradykinin-stimulated PC12 cells. J Biol Chem 1992; 267 (24): 16834-40.         [ Links ]

117. Hermans SW, Engelmann B, Reinhardt U, Bartholomeus-Van Nooij IG, De Pont JJ, Willems PH. Diradylglycerol formation in cholecystokinin-stimulated rabbit pancreatic acini. Assessment of precursor phospholipids by means of molecular species analysis. Eur J Biochem 1996; 235 (1-2): 73-81.         [ Links ]

118. Lee C, Fisher SK, Agranoff BW, Hajra AK. Quantitative analysis of molecular species of diacylglycerol and phosphatidate formed upon muscarinic receptor activation of human SK-N-SH neuroblastoma cells. J Biol Chem 1991; 266 (34):22837-46.         [ Links ]

119. Sebaldt RJ, Adams DO, Uhing RJ. Quantification of contributions of phospholipid precursors to diradylglycerols in stimulated mononuclear phagocytes. Biochem J 1992; 284: 367-75.         [ Links ]

120. Chow SC, Sisfontes L, Jondal M, Bjorkhem I. Modification of membrane phospholipid fatty acyl composition in a leukemic T cell line: effects on receptor mediated intracellular Ca2+ increase. Biochim Biophys Acta 1991; 1092 (3): 358-66.         [ Links ]

121. Pelassy C, Mary D, Aussel C. Diacylglycerol production in Jurkat T-cells: differences between CD3, CD2 and PHA activation pathways. Cell Signal 1991; 3 (1): 35-40.         [ Links ]

122. Singh BB, Lockwich TP, Bandyopadhyay BC, Liu X, Bollimuntha S, Brazer SC et al. VAMP2-dependent exocytosis regulates plasma membrane insertion of TRPC3 channels and contributes to agonist-stimulated Ca2+ influx. Mol Cell 2004; 15 (4): 635-46.         [ Links ]

123. Marignani PA, Epand RM, Sebaldt RJ. Acyl chain dependence of diacylglycerol activation of protein kinase C activity in vitro. Biochem Biophys Res Commun 1996; 225 (2): 469-73.         [ Links ]

124. Madani S, Hichami A, Legrand A, Belleville J, Khan NA. Implication of acyl chain of diacylglycerols in activation of different isoforms of protein kinase C. Faseb J 2001; 15 (14):2595-601.         [ Links ]

125. Bartoli R, Fernandez-Banares F, Navarro E, Castella E, Mane J, Alvarez M, et al. Effect of olive oil on early and late events of colon carcinogenesis in rats: modulation of arachidonic acid metabolism and local prostaglandin E2 synthesis. Gut 2000; 46(2): 191-9.         [ Links ]

126. Fujiwara K, Maekawa F, Yada T. Oleic acid interacts with GPR40 to induce Ca2+ signaling in rat islet beta-cells: mediation by PLC and L-type Ca2+ channel and link to insulin release. Am J Physiol Endocrinol Metab 2005; 289 (4): E670-7.         [ Links ]

127. Hardy S, St-Onge GG, Joly E, Langelier Y, Prentki M. Oleate promotes the proliferation of breast cancer cells via the G protein-coupled receptor GPR40. J Biol Chem 2005; 280 (14): 13285-91.         [ Links ]

128. Katsuta Y, Iida T, Hasegawa K, Inomata S, Denda M. Function of oleic acid on epidermal barrier and calcium influx into keratinocytes is associated with N-methyl D-aspartate-type glutamate receptors. Br J Dermatol 2009; 160 (1): 69-74.         [ Links ]

129. Richieri GV, Kleinfeld AM. Free fatty acid perturbation of transmembrane signaling in cytotoxic T lymphocytes. J Immunol 1989; 143 (7): 2302-10.         [ Links ]

130. Gamberucci A, Fulceri R, Benedetti A. Inhibition of store-dependent capacitative Ca2+ influx by unsaturated fatty acids. Cell Calcium 1997; 21 (5): 375-85.         [ Links ]

131. Zugaza JL, Casabiell XA, Bokser L, Casanueva FF. Oleic acid blocks EGF-induced [Ca2+]i release without altering cellular metabolism in fibroblast EGFR T17. Biochem Biophys Res Commun 1995; 207 (1): 105-10.         [ Links ]

132. Ekokoski E, Forss L, Tornquist K. Inhibitory action of fatty acids on calcium fluxes in thyroid FRTL-5 cells. Mol Cell Endocrinol 1994; 103 (1-2): 125-32.         [ Links ]

133. Okada T, Inoue R, Yamazaki K, Maeda A, Kurosaki T, Yamakuni T et al. Molecular and functional characterization of a novel mouse transient receptor potential protein homologue TRP7. Ca2+ -permeable cation channel that is constitutively activated and enhanced by stimulation of G protein-coupled receptor. J Biol Chem 1999; 274 (39): 27359-70.         [ Links ]

134. Hofmann T, Obukhov AG, Schaefer M, Harteneck C, Gudermann T, Schultz G. Direct activation of human TRPC6 and TRPC3 channels by diacylglycerol. Nature 1999; 397 (6716): 259-63.         [ Links ]

135. Gamberucci A, Giurisato E, Pizzo P, Tassi M, Giunti R, McIntosh DP et al. Diacylglycerol activates the influx of extracellular cations in T-lymphocytes independently of intracellular calcium-store depletion and possibly involving endogenous TRP6 gene products. Biochem J 2002; 364 (Pt 1): 245-54.         [ Links ]

136. Tesfai Y, Brereton HM, Barritt GJ. A diacylglycerol-activated Ca2+ channel in PC12 cells (an adrenal chromaffin cell line) correlates with expression of the TRP-6 (transient receptor potential) protein. Biochem J 2001; 358: 717-26.         [ Links ]

137. Aires V, Hichami A, Boulay G, Khan NA. Activation of TRPC6 calcium channels by diacylglycerol (DAG)-containing arachidonic acid: a comparative study with DAG-containing docosahexaenoic acid. Biochimie 2007; 89 (8): 926-37.         [ Links ]

138. Carrillo C, Hichami A, Andreoletti P, Cherkaoui-Malki M, Cavia MM, Abdoul-Azize S et al. Diacylglycerol-containing oleic acid induces increases in [Ca2+]i via TRPC3/6 channels in human T-cells. BBA - Molecular and Cell Biology of Lipids 2012; 1821 (4): 618-26.         [ Links ]

139. Ahmed SN, Brown DA, London E. On the origin of sphingolipid/cholesterol-rich detergent-insoluble cell membranes: physiological concentrations of cholesterol and sphingolipid induce formation of a detergent-insoluble, liquid-ordered lipid phase in model membranes. Biochemistry 1997; 36 (36):10944-53.         [ Links ]

140. Edidin M. Patches and fences: probing for plasma membrane domains. J Cell Sci Suppl 1993; 17: 165-9.         [ Links ]

141. Edidin M. Lipids on the frontier: a century of cell-membrane bilayers. Nat Rev Mol Cell Biol 2003; 4 (5): 414-8.         [ Links ]

142. Hooper NM. Detergent-insoluble glycosphingolipid/cholesterol-rich membrane domains, lipid rafts and caveolae (review). Mol Membr Biol 1999; 16 (2): 145-56.         [ Links ]

143. Pani B, Ong HL, Liu X, Rauser K, Ambudkar IS, Singh BB. Lipid rafts determine clustering of STIM1 in endoplasmic reticulum-plasma membrane junctions and regulation of store-operated Ca2+ entry (SOCE). J Biol Chem 2008; 283 (25):17333-40.         [ Links ]

144. Pani B, Singh BB. Lipid rafts/caveolae as microdomains of calcium signaling. Cell Calcium 2009; 45 (6): 625-33.         [ Links ]

145. Galan C, Woodard GE, Dionisio N, Salido GM, Rosado JA. Lipid rafts modulate the activation but not the maintenance of store-operated Ca2+ entry. Biochim Biophys Acta 2010; 1803 (9): 1083-93.         [ Links ]

146. Jardin I, Salido GM, Rosado JA. Role of lipid rafts in the interaction between hTRPC1, Orai1 and STIM1. Channels (Austin) 2008; 2 (6): 401-3.         [ Links ]

147. Rogers KR, Kikawa KD, Mouradian M, Hernandez K, McKinnon KM, Ahwah SM et al. Docosahexaenoic acid alters epidermal growth factor receptor-related signaling by disrupting its lipid raft association. Carcinogenesis 2010; 31 (9):1523-30.         [ Links ]

148. Schley PD, Brindley DN, Field CJ. (n-3) PUFA alter raft lipid composition and decrease epidermal growth factor receptor levels in lipid rafts of human breast cancer cells. J Nutr 2007; 137 (3): 548-53.         [ Links ]

149. Shaikh SR, Edidin M. Polyunsaturated fatty acids, membrane organization, T cells, and antigen presentation. Am J Clin Nutr 2006; 84 (6): 1277-89.         [ Links ]

150. Siddiqui RA, Harvey KA, Zaloga GP, Stillwell W. Modulation of lipid rafts by Omega-3 fatty acids in inflammation and cancer: implications for use of lipids during nutrition support. Nutr Clin Pract 2007; 22 (1): 74-88.         [ Links ]

151. Ye S, Tan L, Ma J, Shi Q, Li J. Polyunsaturated docosahexae-noic acid suppresses oxidative stress induced endothelial cell calcium influx by altering lipid composition in membrane caveolar rafts. Prostaglandins Leukot Essent Fatty Acids 2010; 83 (1): 37-43.         [ Links ]

152. Stulnig TM, Berger M, Sigmund T, Raederstorff D, Stockinger H, Waldhausl W. Polyunsaturated fatty acids inhibit T cell signal transduction by modification of detergent-insoluble membrane domains. J Cell Biol 1998; 143 (3): 637-44.         [ Links ]

153. Stulnig TM, Huber J, Leitinger N, Imre EM, Angelisova P, Nowotny P et al. Polyunsaturated eicosapentaenoic acid displaces proteins from membrane rafts by altering raft lipid composition. J Biol Chem 2001; 276 (40): 37335-40.         [ Links ]

154. Li Q, Wang M, Tan L, Wang C, Ma J, Li N et al. Docosahexaenoic acid changes lipid composition and interleukin-2 receptor signaling in membrane rafts. J Lipid Res 2005; 46 (9): 1904-13.         [ Links ]

155. Webb Y, Hermida-Matsumoto L, Resh MD. Inhibition of protein palmitoylation, raft localization, and T cell signaling by 2-bromopalmitate and polyunsaturated fatty acids. J Biol Chem 2000; 275 (1): 261-70.         [ Links ]

156. Fan YY, McMurray DN, Ly LH, Chapkin RS. Dietary (n-3) polyunsaturated fatty acids remodel mouse T-cell lipid rafts. J Nutr 2003; 133 (6): 1913-20.         [ Links ]

157. Puertollano MA, Puertollano E, Alvarez de Cienfuegos G, de Pablo MA. Significance of olive oil in the host immune resistance to infection. Br J Nutr 2007; 98 (Suppl. 1): S54-8.         [ Links ]

158. Puertollano MA, Puertollano E, Alvarez de Cienfuegos G, de Pablo Martinez MA. Olive oil, immune system and infection. Nutr Hosp 2010; 25 (1): 1-8.         [ Links ]

159. Sala-Vila A, Barbosa VM, Calder PC. Olive oil in parenteral nutrition. Curr Opin Clin Nutr Metab Care 2007; 10 (2): 165-74.         [ Links ]

160. Pontes-Arruda A. Biological benefits of an oleic acid-rich lipid emulsion for parenteral nutrition. Clinical Nutrition 2009;4:19-23.         [ Links ]

 

 

Correspondence:
Sara Alonso-Torre.
Pl. Misael Bañuelos, s/n.
09001 Burgos (Spain).
E-mail: salonso@ubu.es

Recibido: 7-II-2012.
Aceptado: 2-III-2012.

Creative Commons License Todo el contenido de esta revista, excepto dónde está identificado, está bajo una Licencia Creative Commons