Skip to main content
  • Review
  • Published:

Diets involved in PPAR and PI3K/AKT/PTEN pathway may contribute toneuroprotection in a traumatic brain injury

Abstract

Traumatic encephalopathy has emerged as a significant public health problem. Itis believed that traumatic encephalopathy is caused by exposure to repetitivebrain trauma prior to the initial symptoms of neurodegenerative disease.Therefore, prevention is important for the disease. The PI3K/AKT/PTEN(phosphoinositide-3 kinase/AKT/phosphatase and tensin homologue deleted onchromosome 10) pathway has been shown to play a pivotal role in neuroprotection,enhancing cell survival by stimulating cell proliferation and inhibitingapoptosis. PTEN negatively regulates the PI3K/AKT pathways through its lipidphosphatase activity. Although PTEN has been discovered as a tumor suppressor,PTEN is also involved in several other diseases, including diabetes andAlzheimer’s disease. Dietary fish oil rich in polyunsaturated fatty acidsmay induce the PTEN expression by activation of peroxisomeproliferator-activated receptor. Supplementation of these natural compounds mayprovide a new therapeutic approach to the brain disorder. We review recentstudies on the features of several diets and the signaling pathways involved intraumatic encephalopathy.

Introduction

Traumatic brain injury is a major health problem throughout the world and is aleading cause of mortality and disability [1, 2]. The consequent encephalopathy is a complicated pathological process;however, the main cause of the deleterious cascades may be cell damage inmitochondria at the cellular level [3]. Reactive oxygen species (ROS), caspases, and apoptosis may be the mainparticipants in the mitochondrial cell damage. Traumatic brain injury is associatedwith permanent spatial learning dysfunction and motor deficits due to brain damage [4]. No pharmacological therapies have yet been approved for the treatment oftraumatic brain injury. The possibility of an effective treatment could be based onthe fact that the majority of traumatic neurodegeneration is due to apathophysiological cascade after the injury that exacerbates the damaging effects ofthe injury. One of the validated mechanisms revealed in experimental traumatic braininjury involves oxygen radical-induced oxidative damage to lipids, proteins, andnucleic acids [3, 5]. Developing new therapies for traumatic brain injury requires elucidationof the neuroprotective mechanisms [5]. The ROS are generated during mitochondrial oxidative metabolism as wellas in cellular response to pathogens, which act as signaling molecules and regulatevarious physiological processes, including proliferation, differentiation,apoptosis, and migration [6–8]. In addition, protein and lipid oxidation by ROS is proposed as a crucialdeterminant of brain health. Nicotinamide adenine dinucleotide phosphate (NADPH)oxidase is a complex that produces ROS during the ischemic period, which is also amajor source of endogenous ROS that comes from mitochondria during the process ofoxidative phosphorylation to produce energy in the form of ATP [9]. NADPH oxidase-generated ROS are also implicated in the development ofangiotensin II-dependent hypertension mediated through the hypothalamic neurons [10]. In addition, ROS are produced by intracellular membrane oxidases.Inflammation is a source of ROS at the sites of tissue. It is important for cells toneutralize ROS before they can damage cellular macromolecules. One mechanism bywhich ROS are thought to exert their effects is through the reversible regulation oftarget molecules such as protein kinase C, mitogen-activated protein kinase,phosphoinositide-3 kinase (PI3K), tyrosine phosphatase, and phosphatase and tensinhomolog deleted on chromosome 10 (PTEN) [11]. However, less is known about the initial regulation of signalingmolecules by ROS. Cellular ROS metabolism is tightly regulated by a variety ofproteins involved in the redox mechanism.

Traumatic brain injury is a devastating neurological injury associated withsignificant morbidity and mortality. The prevention of brain dysfunction intraumatic encephalopathy is a public health concern because of a lack of effectivetreatments. Several potential preventive factors, including modifiable lifestylefactors such as diet, have been suggested by epidemiological research [12]. It has been demonstrated that dietary choices can play a key role in theneuroprotection of traumatic encephalopathy [12]. However, the epidemiological analysis of the relations between nutrientconsumption and neroprotection is complex, and it is unlikely that a singlecomponent plays a major role. The complexity of human diet, especially the highsynergistic or antagonistic correlation among the effects of various nutrients andfoods, makes it difficult to examine their distinct effects. Because many factors inlife influence brain function, several interventions might be promising in theprevention of brain dysfunction in traumatic encephalopathy. The main objective ofthis article is to review the studies linking potential protective factors topathogenesis of traumatic encephalopathy, focusing on the roles of polyunsaturatedomega-3 fatty acids (PUFAs) and curcumin in the PI3K/AKT/PTEN pathway. We willsummarize the current research into mechanisms by which several diet factors bind tothe interaction partners to transduce signals downstream and the implications forthe disease-associated biology.

Reactive oxygen species involved in the PI3K/AKT/PTEN pathway and in neuronaldisorder

Studies indicate that prevention of traumatic brain injury-induced ROS productiondecreases blood–brain barrier disruption, neuronal death, and microglialactivation, which may have high therapeutic potential to reduce traumatic braininjury-induced neuronal death [13]. In addition, a number of studies have demonstrated an antioxidantrole for tumor-suppressor proteins, activating the expression of someantioxidant genes in response to oxidative stress. Tumor-suppressor genesregulate diverse cellular activities, including DNA damage repair, cell cyclearrest, cell proliferation, cell differentiation, migration, and apoptosis [14]. PTEN is a tumor-suppressor gene that is frequently deletedor mutated in a variety of human cancers. It has been demonstrated thatupregulation of PTEN causes modulation of PI3K/AKT signaling to reduce ROSgeneration in cells [15]. Phosphatidylinositol 3,4,5-triphosphate (PIP3) is the major secondmessenger of the PI3K pathway that mediates receptor tyrosine kinase signalingto the survival kinase AKT. PTEN negatively regulates the activity of PI3K/AKTsignaling by converting PIP3 to PIP2 (phosphatidylinositol 4,5-bisphosphate).Increased levels of PIP3 at the membrane cause PH domain-containing proteinssuch as AKT to co-localize, resulting in the kinase-mediated phosphorylation andactivation [16]. The activated AKT phosphorylates target proteins involved in cellsurvival, cell cycling, angiogenesis, and metabolism for neuroprotection(Figure 1). A role for PI3K/AKT signaling insynaptic scaling is suggested by the findings that inhibition of PI3K blockshomeostatic AMPA (α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid)receptor delivery to synapses [17]. A clear dissociation between AKT and ribosomal S6K signaling markerscould be involved in the brain pathological process [18]. The phosphorylation of presenilin 1 (PS1) downregulates itscell-surface expression, which leads to an impaired activation of the PI3K/AKTcell survival signaling. The PS1 also regulates induction of hypoxia-induciblefactor-1α [19]. Accordingly, the abnormal activation of glycogen synthasekinase-3-beta (GSK3β) can reduce neuronal viability [20]. In other words, selective downregulation of the AKT concurrent withelevated GSK3β activity may be linked to brain dysfunctional pathogenesis [21]. Recently, it has been shown that AKT activation may play atherapeutic role in neurodegenerative diseases [22, 23].

Figure 1
figure 1

Schematic representation and overview of PTEN/PI3K/AKT signaling and amodel of the mechanism of peroxisome proliferator-activated receptor(PPAR) action. Similar to other nuclear hormone receptors, PPARsact as a ligand-activated transcription factor. PPARs, in response toligand binding, hetero-dimerize with retinoid-X-receptor (RXR) and bindPPAR response element (PPRE) DNA sequences in the promoters of targetgenes, including PTEN. The uncontrolled generation of reactiveoxygen species (ROS) might contribute to cell proliferation byinhibiting PTEN function. Examples of molecules known to act on thePTEN/PI3K/AKT regulatory pathways are also shown; these molecules mayrelate to the function of presenilin. Hammerheads mean inhibition. Somecritical pathways have been omitted for clarity. GSK3, glycogen synthasekinase-3; HDM2, human homologue of murine mdm2; HIF-1α,hypoxia-inducible factor-1α; IKK, IκB kinase; MAPK,mitogen-activated protein kinase; mTOR, mammalian target of rapamycin;NF-κB, nuclear factor-kappa-B; NOS, nitric oxide synthase; PI3K,phosphoinositide-3 kinase; PTEN, phosphatase and tensin homologuedeleted on chromosome 10; PUFA, polyunsaturated fatty acid; RA, retinoicacid; TSC, tuberous sclerosis complex; TSP1, thrombospondin 1; VEGF,vascular endothelial growth factor.

The tumor suppressor PTEN, which antagonizes the PI3K/AKT pathway, has beenrecognized to play a key role in neural functions. Its level has been found tobe reduced in Alzheimer’s disease (AD) brains [23, 24]. PTEN negatively regulates the activity of PI3K/AKT signaling byconverting PIP3 to PIP2. The PIP3 is the principal second messenger of the PI3Kpathway that mediates receptor tyrosine kinase signaling to the survival kinaseAKT. Increased levels of PIP3 at the membrane cause PH domain-containingproteins such as AKT and PDK-1 to co-localize, resulting in the kinase-mediatedphosphorylation and activation [16]. Schematic structures of the AKT and PTEN protein are shown inFigure 2. The activated AKT phosphorylates targetproteins involved in cell survival, cell cycling, and metabolism. Cell cyclemediators affected by AKT and PTEN include the forkhead transcription factorsand GSK3 [25, 26]. PTEN acts as a regulator of maintaining basal levels of PIP3 below athreshold for those signaling activation. PTEN also plays an important role inthe induction of apoptotic cell death signals in cells when cells lose contactwith the extracellular matrix [27]. Presenilins may play an important role in signaling pathwaysinvolving PI3K/AKT and PTEN that are crucial for physiological functions and thepathogenesis of AD [28]. PTEN may also be involved in a disease state such asParkinson’s disease [29].

Figure 2
figure 2

Schematic structures of human PPAR, AKT1, and PTEN protein. Thepredicted consensual domain structures for each protein are depicted.The sizes of protein are modified for clarity. C2 domain, a structuraldomain involved in targeting proteins to cell membranes; PDZ, a commonstructural domain in signaling proteins (the abbreviation stands forPSD95, Dlg, ZO-1); PPAR, peroxisome proliferator-activated receptor;PTEN, phosphatase and tensin homologue deleted on chromosome 10.

Potential therapeutic approach for cellular protection via the modulation ofPI3K/AKT/PTEN pathway

A wide variety of compounds have been identified as peroxisomeproliferator-activated receptor (PPAR) ligands. The n-3 PUFAs have a beneficialeffect on most of the metabolic risk factors by regulating gene transcriptionfactors, including PPARα and PPARγ [30]. Treating cells with the insulin-sensitizing drug pioglitazone, aPPARγ agonist, attenuates the ROS signaling pathway [31]. Correcting insulin signal dysregulation in traumatic brain injurymay also offer a potential therapeutic approach. A schematic protein structureof the PPARs is shown in Figure 2. Ligand-activatedPPARs bind as heterodimers with the retinoid X receptor (RXR) on PPAR responseelements, which are present in the promoter regions of the responsive genes [32] (Figure 1). Retinoic acid also affects abroad spectrum of physiological processes, including cell growth,differentiation, morphogenesis, reproduction, and development [33], through the action of two types of receptors: the retinoic acidreceptors (RARs) and the RXRs. The transcriptional control by the PPAR/RXRheterodimer also requires interaction with co-regulator complexes [34]. Thus, selective action of PPARs in vivo results from theinterplay at a time point of each of the co-factors available. A number of PPARtarget genes have been characterized. Combined treatment with agonists for theheterodimeric binding partners of PPARγ and the RXRs shows additiveenhancement of the amyloid-beta (Aβ) uptake that is mediated by RXRαactivation [35]. Simultaneous activation of the PPARγ/RXRα heterodimer mayprove beneficial in prevention of traumatic brain injury. Furthermore,PPARγ represents a signaling system that can intercede to restore neuralnetworks [36]. It has been reported that oral administration of the RXR agonist,bexarotene, to a mouse model of AD results in enhanced clearance of solubleAβ [37]. Furthermore, bexarotene stimulated the rapid reversal of cognitivedeficits and improved neural circuit function. Accordingly, RXR activation maystimulate physiological Aβ clearance mechanisms.

Activated PPARs upregulate expression of PTEN (Figure 1). Type-2 diabetes is characterized by diminishedpancreatic β-cell function. Insulin signaling within the β-cells hasbeen shown to play an important role in maintaining the function of theβ-cells. Under basal conditions, enhanced insulin-PI3K signaling viadeletion of PTEN leads to increased β-cell mass [38]. Mice with PTEN deletion in pancreatic cells show an increase in theβ-cell mass because of both increased proliferation and reduced apoptosis.In particular, the relationship between PTEN function and adipocyte-specificfatty acid-binding protein FABP4 is of interest in β-cell signaling [39]. The interaction of PTEN to FABP4 suggests a role for thisphosphatase in the regulation of lipid metabolism and cell differentiation [40]. Tissue-targeted deletion of PTEN leads to improved insulinsensitivity in the insulin-responsive tissues and protects from diabetes [41]. On the other hand, ligands of PPARs are used as oral anti-diabetics [42]. The PTEN is ubiquitously expressed throughout earlyembryogenesis in mammals [43]. Interestingly, rosemary extract represses PTEN expressionin K562 leukemic culture cells [44]. The schematic structure of the PTEN protein is also shown inFigure 2. PTEN protein consists of N-terminalphosphatase, C-terminal C2, and PDZ (PSD-95, DLG1, and ZO-1) binding domains.The PTEN CX5R(S/T) motif resides within an active site that surrounds thecatalytic signature with three basic residues, which are critical for PTEN lipidphosphatase activity. The structure endows PTEN with its preference for acidicphospholipid substrates such as PIP3. Neuroprotection by inhibiting PTEN hasbeen reported by activating the anti-apoptotic PI3K/AKT pathway in primaryneurons [45–47].

Some diets may contribute to neuroprotective effects

Curcumin, a component of turmeric, potently lowers Aβ levels in adose-dependent manner. Furthermore, in vivo studies indicated thatcurcumin was able to reduce Aβ-related pathology in mouse models viaunknown molecular mechanisms [48]. In addition, curcumin can improve structure and plasticity ofsynapse and enhance their learning and memory abilities [49]. The protective effect of curcumin is associated with a significantattenuation in the expression of interleukin-1b, a pro-inflammatory cytokine [50]. Curcumin also reverses the induction of aquaporin-4, an astrocyticwater channel implicated in the development of cellular edema after brain trauma [50]. Curcumin blocks IL-1b-induced aquaporin-4 expression in culturedastrocytes by reduced activation of the p50 and p65 subunits of nuclearfactor-kappa-B. Interestingly, curcumin enhances synaptic plasticity andcognitive function after fluid percussion injury in rats [51], suggesting that curcumin may represent a potent therapeutic agentthat exerts multiple beneficial effects after traumatic brain injury. It issuggested that the neuroprotection of curcumin might be mediated via PI3K/AKTsignaling pathway [52]. Dietary treatment with curcumin, fish oil, or a combination of bothhas the potential to improve c-Jun N-terminal kinase signaling, phospho-taupathology, and cognitive deficits in AD [53].

Genistein, a phytoestrogen present in high concentrations in soy, alsodownregulates presenilin via the inhibition of ubiquilin 1 expression inlymphoid cells [54]. Genistein has potent anti-tumor activity in various cancer cells. Inaddition to the inhibition of tyrosine kinases, genistein has a strongestrogen-like effect, which is also beneficial for the plasticity of AD [55]. Genistein potentiates the anti-cancer effects of gemcitabine inhuman osteosarcoma via the downregulation of the Akt pathway [56]. Resveratrol also appears to be beneficial as an anti-AD agent [57–60]. Resveratrol treatment also prevented the pro-inflammatory effect offibrillar Aβ on macrophages by potently inhibiting the effect of Aβ [61]. The n-3 PUFAs are a family of biologically active fatty acids, whichhave a range of physiological roles that relate to optimal cellular functions.The simplest member of this family, α-linolenic acid, can be converted tothe biologically more active long-chain n-3 PUFAs such as eicosapentaenoic acidand docosahexaenoic acid. Several works have led to the identification ofvarious PPAR ligands that include the n-3 PUFAs [62, 63]. In addition, linoleic acid and γ-linolenic acid could bindPPARδ very well [64]. All distinct PPAR subtypes, PPARs (α, β, and γ),share a high degree of structural homology with other members of the superfamilyin the DNA-binding domain and ligand-binding domain. PPAR-ligands are emergingas potential therapeutics for inflammatory and other metabolic diseases. The useof n-3 PUFAs has been shown as a possible preventive measure for AD [65–68]. Retinoic acid affects a variety of physiological processes throughthe action of RAR and RXR. Stimulation of the RARα signaling pathway offerstherapeutic potential by clearing Aβ for the treatment of AD [69]. Retinoic acid plays a key role in the adult brain by participatingin the homeostatic control of synaptic plasticity and is essential for memoryfunction. Retinoids are vitamin A derivatives involved in cellular regulatoryprocesses, including cell differentiation and neurite outgrowth, which may alsoinfluence Aβ processing [70]. Thus, neuroprotecton could be performed by certain diets(Figure 3).

Figure 3
figure 3

Implication of certain diets in neuroprotection via modulation of thefunction of PPARs, PTEN, AKT, and presenilin. AD,Alzheimer’s disease; DHA, docosahexaenoic acid; EPA,eicosapentaenoic acid; PPAR, peroxisome proliferator-activated receptor;PTEN, phosphatase and tensin homologue deleted on chromosome 10.

Perspective

Increased ROS can enhance insulin signaling to attenuate the development ofinsulin resistance. The enhanced ROS-dependent insulin signaling is attributableto the oxidation and inhibition of PTEN. In patients with traumatic braininjury, nutritional status may result in changes in the biochemistry indicators.Curcumin, retinoic acids, and n-3 PUFAs are considered to exert the effects atseveral cellular levels. In addition, diet usually consists of complexcombinations of lipids or nutrients that might act synergistically orantagonistically. One of the pleiotropic properties of these foods could explaintheir disease-protective potentials, which could be mediated through modulationof PI3K/AKT/PTEN pathway. As PTEN is induced by the activated PPARs, this mayalso offer a potential therapeutic modality for the treatment of thosePTEN-related diseases. These key molecules may be regulated at multiple levels,including transcription, protein stability, and phosphorylation. So, preciseunderstanding of these regulations is crucial for therapeutic intervention andthe effective design of novel therapeutics. In addition to showing theantioxidant strategy of scavenging the initiating radicals in the injured braintissue, recent work has shown that carbonyl scavenging compounds can also act toprotect cellular proteins. Further mechanistic studies are needed in order toelucidate the precise molecular mechanisms and to determine whether an adequatedietary intake is related to improved brain function and to determine the roleit plays regarding the preservation of brain health. Long-term clinical studiesare obligatory to enlighten the effect of treatment in the management oftraumatic brain injury.

Note

This article is part of a series on Traumatic Brain Injury, edited byRobert Stern. Other articles in this series can be found athttp://alzres.com/series/traumaticbraininjury.

Abbreviations

AD:

Alzheimer’s disease

Aβ:

Amyloid-beta

FABP:

Fatty acid-bindingprotein

GSK3:

Glycogen synthase kinase-3

NADPH:

Nicotinamide adenine dinucleotidephosphate

PI3K:

Phosphoinositide-3 kinase

PIP2:

Phosphatidylinositol4,5-bisphosphate

PIP3:

Phosphatidylinositol 3,4,5-triphosphate

PPAR:

Peroxisomeproliferator-activated receptor

PS1:

Presenilin 1

PTEN:

Phosphatase and tensinhomologue deleted on chromosome 10

PUFA:

Polyunsaturated fatty acid

RAR:

Retinoicacid receptor

ROS:

Reactive oxygen species

RXR:

Retinoid-X-receptor.

References

  1. Xiong Y, Mahmood A, Chopp M: Animal models of traumatic brain injury. Nat Rev Neurosci. 2013, 14: 128-142.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  2. Levin H, Smith D: Traumatic brain injury: networks and neuropathology. Lancet Neurol. 2013, 12: 15-16. 10.1016/S1474-4422(12)70300-9.

    Article  PubMed Central  PubMed  Google Scholar 

  3. Cheng G, Kong RH, Zhang LM, Zhang JN: Mitochondria in traumatic brain injury and mitochondrial-targetedmultipotential therapeutic strategies. Br J Pharmacol. 2012, 16: 699-719.

    Article  Google Scholar 

  4. Song SX, Gao JL, Wang KJ, Li R, Tian YX, Wei JQ, Cui JZ: Attenuation of brain edema and spatial learning deficits by the inhibition ofNADPH oxidase activity using apocynin following diffuse traumatic braininjury in rats. Mol Med Rep. 2012, Oct 24: [Epub ahead of print],

    Google Scholar 

  5. Hellmich HL, Rojo DR, Micci MA, Sell SL, Boone DR, Crookshanks JM, DeWitt DS, Masel BE, Prough DS: Pathway analysis reveals common pro-survival mechanisms of metyrapone andcarbenoxolone after traumatic brain injury. PLoS One. 2013, 8: e53230-10.1371/journal.pone.0053230.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  6. Al-Gubory KH, Fowler PA, Garrel C: The roles of cellular reactive oxygen species, oxidative stress andantioxidants in pregnancy outcomes. Int J Biochem Cell Biol. 2010, 42: 1634-1650. 10.1016/j.biocel.2010.06.001.

    Article  CAS  PubMed  Google Scholar 

  7. Zhang Y, Du Y, Le W, Wang K, Kieffer N, Zhang J: Redox control of the survival of healthy and diseased cells. Antioxid Redox Signal. 2011, 15: 2867-2908. 10.1089/ars.2010.3685.

    Article  CAS  PubMed  Google Scholar 

  8. Scatena R: Mitochondria and cancer: a growing role in apoptosis, cancer cell metabolismand dedifferentiation. Adv Exp Med Biol. 2012, 942: 287-308. 10.1007/978-94-007-2869-1_13.

    Article  CAS  PubMed  Google Scholar 

  9. Ballard JW: Drosophila simulans as a novel model for studying mitochondrial metabolismand aging. Exp Gerontol. 2005, 40: 763-773. 10.1016/j.exger.2005.07.014.

    Article  CAS  PubMed  Google Scholar 

  10. Coleman CG, Wang G, Faraco G, Marques Lopes J, Waters EM, Milner TA, Iadecola C, Pickel VM: Membrane trafficking of NADPH oxidase p47(phox) in paraventricularhypothalamic neurons parallels local free radical production in angiotensinII slow-pressor hypertension. J Neurosci. 2013, 33: 4308-4316. 10.1523/JNEUROSCI.3061-12.2013.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  11. Li ZY, Yang Y, Ming M, Liu B: Mitochondrial ROS generation for regulation of autophagic pathways incancer. Biochem Biophys Res Commun. 2011, 414: 5-8. 10.1016/j.bbrc.2011.09.046.

    Article  CAS  PubMed  Google Scholar 

  12. Gillette-Guyonnet S, Secher M, Vellas B: Nutrition and neurodegeneration: epidemiological evidence and challenges forfuture research. Br J Clin Pharmacol. 2013, 75: 738-755.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  13. Choi BY, Jang BG, Kim JH, Lee BE, Sohn M, Song HK, Suh SW: Prevention of traumatic brain injury-induced neuronal death by inhibition ofNADPH oxidase activation. Brain Res. 2012, 1481: 49-58.

    Article  CAS  PubMed  Google Scholar 

  14. Asai T, Liu Y, Bae N, Nimer SD: The p53 tumor suppressor protein regulates hematopoietic stem cell fate. J Cell Physiol. 2011, 226: 2215-2221. 10.1002/jcp.22561.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  15. Xu J, Tian W, Ma X, Guo J, Shi Q, Jin Y, Xi J, Xu Z: The molecular mechanism underlying morphine-induced Akt activation: roles ofprotein phosphatases and reactive oxygen species. Cell Biochem Biophys. 2011, 61: 303-311. 10.1007/s12013-011-9213-5.

    Article  CAS  PubMed  Google Scholar 

  16. Howes AL, Arthur JF, Zhang T, Miyamoto S, Adams JW, Dorn GW, Woodcock EA, Brown JH: Akt-mediated cardiomyocyte survival pathways are compromised by G alphaq-induced phosphoinositide 4,5-bisphosphate depletion. J Biol Chem. 2003, 278: 40343-40351. 10.1074/jbc.M305964200.

    Article  CAS  PubMed  Google Scholar 

  17. Hou Q, Zhang D, Jarzylo L, Huganir RL, Man HY: Homeostatic regulation of AMPA receptor expression at single hippocampalsynapses. Proc Natl Acad Sci U S A. 2008, 105: 775-780. 10.1073/pnas.0706447105.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  18. Damjanac M, Rioux Bilan A, Paccalin M, Pontcharraud R, Fauconneau B, Hugon J, Page G: Dissociation of Akt/PKB and ribosomal S6 kinase signaling markers in atransgenic mouse model of Alzheimer’s disease. Neurobiol Dis. 2008, 29: 354-367. 10.1016/j.nbd.2007.09.008.

    Article  CAS  PubMed  Google Scholar 

  19. De Gasperi R, Sosa MA, Dracheva S, Elder GA: Presenilin-1 regulates induction of hypoxia inducible factor-1α: alteredactivation by a mutation associated with familial Alzheimer’sdisease. Mol Neurodegener. 2010, 5: 38-10.1186/1750-1326-5-38.

    Article  PubMed Central  PubMed  Google Scholar 

  20. Uemura K, Kuzuya A, Shimozono Y, Aoyagi N, Ando K, Shimohama S, Kinoshita A: GSK3beta activity modifies the localization and function of presenilin 1. J Biol Chem. 2007, 282: 15823-15832. 10.1074/jbc.M610708200.

    Article  CAS  PubMed  Google Scholar 

  21. Ryder J, Su Y, Ni B: Akt/GSK3beta serine/threonine kinases: evidence for a signalling pathwaymediated by familial Alzheimer’s disease mutations. Cell Signal. 2004, 16: 187-200. 10.1016/j.cellsig.2003.07.004.

    Article  CAS  PubMed  Google Scholar 

  22. Cheng B, Martinez AA, Morado J, Scofield V, Roberts JL, Maffi SK: Retinoic acid protects against proteasome inhibition associated cell death inSH-SY5Y cells via the AKT pathway. Neurochem Int. 2013, 62: 31-42. 10.1016/j.neuint.2012.10.014.

    Article  CAS  PubMed  Google Scholar 

  23. Haas-Kogan D, Stokoe D: PTEN in brain tumors. Expert Rev Neurother. 2008, 8: 599-610. 10.1586/14737175.8.4.599.

    Article  CAS  PubMed  Google Scholar 

  24. Gupta A, Dey CS: PTEN, a widely known negative regulator of insulin/PI3K signaling, positivelyregulates neuronal insulin resistance. Mol Biol Cell. 2012, 23: 3882-3898. 10.1091/mbc.E12-05-0337.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  25. Nakamura N, Ramaswamy S, Vazquez F, Signoretti S, Loda M, Sellers WR: Forkhead transcription factors are critical effectors of cell death and cellcycle arrest downstream of PTEN. Mol Cell Biol. 2000, 20: 8969-8982. 10.1128/MCB.20.23.8969-8982.2000.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  26. Mulholland DJ, Dedhar S, Wu H, Nelson CC: PTEN and GSK3beta: key regulators of progression to androgen-independentprostate cancer. Oncogene. 2006, 25: 329-337. 10.1038/sj.onc.1209020.

    Article  CAS  PubMed  Google Scholar 

  27. Fournier MV, Fata JE, Martin KJ, Yaswen P, Bissell MJ: Interaction of E-cadherin and PTEN regulates morphogenesis and growth arrestin human mammary epithelial cells. Cancer Res. 2009, 69: 4545-4552. 10.1158/0008-5472.CAN-08-1694.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  28. Zhang H, Liu R, Wang R, Hong S, Xu H, Zhang YW: Presenilins regulate the cellular level of the tumor suppressor PTEN. Neurobiol Aging. 2008, 29: 653-660. 10.1016/j.neurobiolaging.2006.11.020.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  29. Rochet JC, Hay BA, Guo M: Molecular insights into Parkinson’s disease. Prog Mol Biol Transl Sci. 2012, 107: 125-188.

    Article  CAS  PubMed  Google Scholar 

  30. Hajjar T, Meng GY, Rajion MA, Vidyadaran S, Othman F, Farjam AS, Li TA, Ebrahimi M: Omega 3 polyunsaturated fatty acid improves spatial learning and hippocampalperoxisome proliferator activated receptors (PPARα and PPARγ) geneexpression in rats. BMC Neurosci. 2012, 13: 109-10.1186/1471-2202-13-109.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  31. Yuan X, Zhang Z, Gong K, Zhao P, Qin J, Liu N: Inhibition of reactive oxygen species/extracellular signal-regulated kinasespathway by pioglitazone attenuates advanced glycation end products-inducedproliferation of vascular smooth muscle cells in rats. Biol Pharm Bull. 2011, 34: 618-623. 10.1248/bpb.34.618.

    Article  CAS  PubMed  Google Scholar 

  32. Chandra V, Huang P, Hamuro Y, Raghuram S, Wang Y, Burris TP, Rastinejad F: Structure of the intact PPAR-gamma-RXR- nuclear receptor complex on DNA. Nature. 2008, 456: 350-356.

    Article  PubMed Central  PubMed  Google Scholar 

  33. Tarrade A, Rochette-Egly C, Guibourdenche J, Evain-Brion D: The expression of nuclear retinoid receptors in human implantation. Placenta. 2000, 21: 703-710. 10.1053/plac.2000.0568.

    Article  CAS  PubMed  Google Scholar 

  34. Michalik L, Auwerx J, Berger JP, Chatterjee VK, Glass CK, Gonzalez FJ, Grimaldi PA, Kadowaki T, Lazar MA, O’Rahilly S, Palmer CN, Plutzky J, Reddy JK, Spiegelman BM, Staels B, Wahli W: International Union of Pharmacology. LXI. Peroxisome proliferator-activatedreceptors. Pharmacol Rev. 2006, 58: 726-741. 10.1124/pr.58.4.5.

    Article  CAS  PubMed  Google Scholar 

  35. Yamanaka M, Ishikawa T, Griep A, Axt D, Kummer MP, Heneka MT: PPARγ/RXRα-induced and CD36-mediated microglial amyloid-βphagocytosis results in cognitive improvement in amyloid precursorprotein/presenilin 1 mice. J Neurosci. 2012, 32: 17321-17331. 10.1523/JNEUROSCI.1569-12.2012.

    Article  CAS  PubMed  Google Scholar 

  36. Denner LA, Rodriguez-Rivera J, Haidacher SJ, Jahrling JB, Carmical JR, Hernandez CM, Zhao Y, Sadygov RG, Starkey JM, Spratt H, Luxon BA, Wood TG, Dineley KT: Cognitive enhancement with rosiglitazone links the hippocampal PPARγ andERK MAPK signaling pathways. J Neurosci. 2012, 32: 16725-16735a. 10.1523/JNEUROSCI.2153-12.2012.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  37. Cramer PE, Cirrito JR, Wesson DW, Lee CY, Karlo JC, Zinn AE, Casali BT, Restivo JL, Goebel WD, James MJ, Brunden KR, Wilson DA, Landreth GE: ApoE-directed therapeutics rapidly clear β-amyloid and reverse deficitsin AD mouse models. Science. 2012, 335: 1503-1506. 10.1126/science.1217697.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  38. Stiles BL, Kuralwalla-Martinez C, Guo W, Gregorian C, Wang Y, Tian J, Magnuson MA, Wu H: Selective deletion of Pten in pancreatic beta cells leads to increased isletmass and resistance to STZ-induced diabetes. Mol Cell Biol. 2006, 26: 2772-2781. 10.1128/MCB.26.7.2772-2781.2006.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  39. Gorbenko O, Panayotou G, Zhyvoloup A, Volkova D, Gout I, Filonenko V: Identification of novel PTEN-binding partners: PTEN interaction with fattyacid binding protein FABP4. Mol Cell Biochem. 2010, 337: 299-305. 10.1007/s11010-009-0312-1.

    Article  CAS  PubMed  Google Scholar 

  40. Tsuda M, Inoue-Narita T, Suzuki A, Itami S, Blumenberg M, Manabe M: Induction of gene encoding FABP4 in Pten-null keratinocytes. FEBS Lett. 2009, 583: 1319-1322. 10.1016/j.febslet.2009.03.030.

    Article  CAS  PubMed  Google Scholar 

  41. Stiles B, Wang Y, Stahl A, Bassilian S, Lee WP, Kim YJ, Sherwin R, Devaskar S, Lesche R, Magnuson MA, Wu H: Liver-specific deletion of negative regulator Pten results in fatty liver andinsulin hypersensitivity [corrected]. Proc Natl Acad Sci U S A. 2004, 101: 2082-2087. 10.1073/pnas.0308617100.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  42. Lecka-Czernik B: Aleglitazar, a dual PPARα and PPARγ agonist for the potential oraltreatment of type 2 diabetes mellitus. IDrugs. 2010, 13: 793-801.

    CAS  PubMed  Google Scholar 

  43. Knobbe CB, Lapin V, Suzuki A, Mak TW: The roles of PTEN in development, physiology and tumorigenesis in mousemodels: a tissue-by-tissue survey. Oncogene. 2008, 27: 5398-5415. 10.1038/onc.2008.238.

    Article  CAS  PubMed  Google Scholar 

  44. Yoshida H, Okumura N, Kitagishi Y, Nishimura Y, Matsuda S: Ethanol extract of rosemary repressed PTEN expression in K562 culturecells. Int J Appl Biol Pharm Technol. 2011, 2: 316-322.

    Google Scholar 

  45. Delgado-Esteban M, Martin-Zanca D, Andres-Martin L, Almeida A, Bolaños JP: Inhibition of PTEN by peroxynitrite activates thephosphoinositide-3-kinase/Akt neuroprotective signaling pathway. J Neurochem. 2007, 102: 194-205. 10.1111/j.1471-4159.2007.04450.x.

    Article  CAS  PubMed  Google Scholar 

  46. Fuentealba RA, Liu Q, Kanekiyo T, Zhang J, Bu G: Low density lipoprotein receptor-related protein 1 promotes anti-apoptoticsignaling in neurons by activating Akt survival pathway. J Biol Chem. 2009, 284: 34045-34053. 10.1074/jbc.M109.021030.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  47. Chen LM, Xiong YS, Kong FL, Qu M, Wang Q, Chen XQ, Wang JZ, Zhu LQ: Neuroglobin attenuates Alzheimer-like tau hyperphosphorylation by activatingAkt signaling. J Neurochem. 2012, 120: 157-164. 10.1111/j.1471-4159.2011.07275.x.

    Article  CAS  PubMed  Google Scholar 

  48. Zhang C, Browne A, Child D, Tanzi RE: Curcumin decreases amyloid-beta peptide levels by attenuating the maturationof amyloid-beta precursor protein. J Biol Chem. 2010, 285: 28472-28480. 10.1074/jbc.M110.133520.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  49. Sharma S, Ying Z, Gomez-Pinilla F: A pyrazole curcumin derivative restores membrane homeostasis disrupted afterbrain trauma. Exp Neurol. 2010, 226: 191-199. 10.1016/j.expneurol.2010.08.027.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  50. Laird MD, Sukumari-Ramesh S, Swift AE, Meiler SE, Vender JR, Dhandapani KM: Curcumin attenuates cerebral edema following traumatic brain injury in mice:a possible role for aquaporin-4?. J Neurochem. 2010, 113: 637-648. 10.1111/j.1471-4159.2010.06630.x.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  51. Wu Q, Chen Y, Li X: HDAC1 expression and effect of curcumin on proliferation of Raji cells. J Huazhong Univ Sci Technolog Med Sci. 2006, 26: 199-201. 10.1007/BF02895815.

    Article  PubMed  Google Scholar 

  52. Wang R, Li YH, Xu Y, Li YB, Wu HL, Guo H, Zhang JZ, Zhang JJ, Pan XY, Li XJ: Curcumin produces neuroprotective effects via activating brain-derivedneurotrophic factor/TrkB-dependent MAPK and PI-3K cascades in rodentcortical neurons. Prog Neuropsychopharmacol Biol Psychiatry. 2010, 34: 147-153. 10.1016/j.pnpbp.2009.10.016.

    Article  CAS  PubMed  Google Scholar 

  53. Ma QL, Yang F, Rosario ER, Ubeda OJ, Beech W, Gant DJ, Chen PP, Hudspeth B, Chen C, Zhao Y, Vinters HV, Frautschy SA, Cole GM: Beta-amyloid oligomers induce phosphorylation of tau and inactivation ofinsulin receptor substrate via c-Jun N-terminal kinase signaling:suppression by omega-3 fatty acids and curcumin. J Neurosci. 2009, 29: 9078-9089. 10.1523/JNEUROSCI.1071-09.2009.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  54. Yoshida H, Okumura N, Nishimura Y, Kitagishi Y, Matsuda S: Turmeric and curcumin suppress presenilin 1 protein expression in Jurkatcells. Exp Ther Med. 2011, 2: 629-632.

    PubMed Central  CAS  PubMed  Google Scholar 

  55. Kadish I, van Groen T: Lesion-induced hippocampal plasticity in transgenic Alzheimer’s diseasemouse models: influences of age, genotype, and estrogen. J Alzheimers Dis. 2009, 18: 429-445.

    CAS  PubMed  Google Scholar 

  56. Liang C, Li H, Shen C, Lai J, Shi Z, Liu B, Tao HM: Genistein potentiates the anti-cancer effects of gemcitabine in humanosteosarcoma via the downregulation of Akt and nuclear factor-κBpathway. Anticancer Agents Med Chem. 2012, 12: 554-563. 10.2174/187152012800617867.

    Article  CAS  PubMed  Google Scholar 

  57. Villaflores OB, Chen YJ, Chen CP, Yeh JM, Wu TY: Curcuminoids and resveratrol as anti-Alzheimer agents. Taiwan J Obstet Gynecol. 2012, 51: 515-525. 10.1016/j.tjog.2012.09.005.

    Article  PubMed  Google Scholar 

  58. Huang TC, Lu KT, Wo YY, Wu YJ, Yang YL: Resveratrol protects rats from Aβ-induced neurotoxicity by the reductionof iNOS expression and lipid peroxidation. PLoS One. 2011, 6: e29102-10.1371/journal.pone.0029102.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  59. Li F, Gong Q, Dong H, Shi J: Resveratrol, a neuroprotective supplement for Alzheimer’s disease. Curr Pharm Des. 2012, 18: 27-33. 10.2174/138161212798919075.

    Article  PubMed  Google Scholar 

  60. Karuppagounder SS, Pinto JT, Xu H, Chen HL, Beal MF, Gibson GE: Dietary supplementation with resveratrol reduces plaque pathology in atransgenic model of Alzheimer’s disease. Neurochem Int. 2009, 54: 111-118. 10.1016/j.neuint.2008.10.008.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  61. Capiralla H, Vingtdeux V, Zhao H, Sankowski R, Al-Abed Y, Davies P, Marambaud P: Resveratrol mitigates lipopolysaccharide- and Aβ-mediated microglialinflammation by inhibiting the TLR4/NF-κB/STAT signaling cascade. J Neurochem. 2012, 120: 461-472. 10.1111/j.1471-4159.2011.07594.x.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  62. Kouroumichakis I, Papanas N, Zarogoulidis P, Liakopoulos V, Maltezos E, Mikhailidis DP: Fibrates: therapeutic potential for diabetic nephropathy?. Eur J Intern Med. 2012, 23: 309-316. 10.1016/j.ejim.2011.12.007.

    Article  CAS  PubMed  Google Scholar 

  63. Friedland SN, Leong A, Filion KB, Genest J, Lega IC, Mottillo S, Poirier P, Reoch J, Eisenberg MJ: The cardiovascular effects of peroxisome proliferator-activated receptoragonists. Am J Med. 2012, 125: 126-133. 10.1016/j.amjmed.2011.08.025.

    Article  CAS  PubMed  Google Scholar 

  64. Fu J, Zhang XW, Liu K, Li QS, Zhang LR, Yang XH, Zhang ZM, Li CZ, Luo Y, He ZX, Zhu HL: Hypolipidemic activity in Sprague–Dawley rats and constituents of anovel natural vegetable oil from Cornus wilsoniana fruits. Food Sci. 2012, 77: H160-H169. 10.1111/j.1750-3841.2012.02786.x.

    Article  CAS  Google Scholar 

  65. Donohue MC, Aisen PS: Mixed model of repeated measures versus slope models in Alzheimer’sdisease clinical trials. J Nutr Health Aging. 2012, 16: 360-364. 10.1007/s12603-012-0047-7.

    Article  CAS  PubMed  Google Scholar 

  66. Shah R: The role of nutrition and diet in Alzheimer disease: a systematic review. J Am Med Dir Assoc. 2013, 14: 398-402. 10.1016/j.jamda.2013.01.014.

    Article  PubMed  Google Scholar 

  67. Swanson D, Block R, Mousa SA: Omega-3 fatty acids EPA and DHA: health benefits throughout life. Adv Nutr. 2012, 3: 1-7. 10.3945/an.111.000893.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  68. Calon F: Omega-3 polyunsaturated fatty acids in Alzheimer’s disease: keyquestions and partial answers. Curr Alzheimer Res. 2011, 8: 470-478. 10.2174/156720511796391881.

    Article  CAS  PubMed  Google Scholar 

  69. Goncalves MB, Clarke E, Hobbs C, Malmqvist T, Deacon R, Jack J, Corcoran JP: Amyloid β inhibits retinoic acid synthesis exacerbating Alzheimerdisease pathology which can be attenuated by an retinoic acid receptorα agonist. Eur J Neurosci. 2013, 37: 1182-1192. 10.1111/ejn.12142.

    Article  PubMed Central  PubMed  Google Scholar 

  70. Lerner AJ, Gustaw-Rothenberg K, Smyth S, Casadesus G: Retinoids for treatment of Alzheimer’s disease. Biofactors. 2012, 38: 84-89. 10.1002/biof.196.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgments

This work was supported by grants-in-aid from the Ministry of Education, Culture,Sports, Science and Technology in Japan. In addition, this work was supported inpart by the grant from Nakagawa Masashichi Shoten Co., Ltd.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Satoru Matsuda.

Additional information

Competing interests

The authors declare that they have no competing interests.

Yasuko Kitagishi and Satoru Matsuda contributed equally to this work.

Authors’ original submitted files for images

Rights and permissions

Reprints and permissions

About this article

Cite this article

Kitagishi, Y., Matsuda, S. Diets involved in PPAR and PI3K/AKT/PTEN pathway may contribute toneuroprotection in a traumatic brain injury. Alz Res Therapy 5, 42 (2013). https://doi.org/10.1186/alzrt208

Download citation

  • Published:

  • DOI: https://doi.org/10.1186/alzrt208

Keywords