Abstract
Phospholipase A2 (PLA2), cyclooxygenase (COX) and prostaglandin (PG) synthase are enzymes involved in arachidonate cascade. PLA2 liberates arachidonic acid (AA) from cell membrane lipids. COX oxidizes AA to PGG2 followed by an endoperoxidase reaction that converts PGG2 into PGH2. PGs are generated from astrocytes, microglial cells and neurons in the central nervous system, and are altered in the brain of demented patients. Dementia is principally diagnosed into Alzheimer’s disease (AD) and vascular dementia (VaD). In older patients, the brain lesions associated with each pathological process often occur together. Regional brain microvascular abnormalities appear before cognitive decline and neurodegeneration. The coexistence of AD and VaD pathology is often termed mixed dementia. AD and VaD brain lesions interact in important ways to decline cognition, suggesting common pathways of the two neurological diseases. Arachidonate cascade is one of the converged intracellular signal transductions between AD and VaD. PLA2 from mammalian sources are classified as secreted (sPLA2), Ca2+-dependent, cytosolic (cPLA2) and Ca2+-independent cytosolic PLA2 (iPLA2). PLA2 activity can be regulated by calcium, by phosphorylation, and by agonists binding to G-protein-coupled receptors. cPLA2 is upregulalted in AD, but iPLA2 is downregulated. On the other hand, sPLA2 is increased in animal models for VaD. COX-2 is induced and PGD2 are elevated in both AD and VaD. This review presents evidences for central roles of PLA2s, COXs and PGs in the dementia.
Key Words: Alzheimer’s disease; arachidonic acid; cyclooxygenase; mixed dementia; phospholipase A2; prostaglandin; vascular dementia; PGD2; 15d-Δ12,14-PGJ2
INTRODUCTION
Dementia is currently defined by DSM-IV [3] as a loss of intellectual abilities of sufficient severity to interfere with social or occupational functioning, always accompanied by memory impairment and at least one of the following: impairment of abstract thinking, judgment or other disturbance of higher cortical function in the absence of delirium. Alzheimer’s type is the commonest form of dementia, the other forms being vascular dementia (VaD) and mixed dementia. For more than 30 years, Alzheimer’s disease (AD) has been classified and managed as a neurodegenerative disease characterized by extracellular amyloid plaques and intracellular neurofibrillary tangles (NFTs) [8] (Table 1). On the other hand, VaD is caused by vascular lesions, cerebral infarctions, multiple lacunar infarctions and ischemic periventricular leukoencephalopathy (Table 1).
Table 1.
Symptoms, Lesions and Risk Factors of Alzheimer’s Disease and Vascular Dementia
Alzheimer’s dementia | Vascular dementia | |
---|---|---|
Symptoms | Cognitive decline Extrapyramidal motor dysfunction |
Impairment of short-term memory, Visual disturbances Brain stem abnormalities, Sensory or motor symopoms |
Lesions | Extracellular amyloid plaques Intracellular neurofibrillary tangles |
Cerebral infacrction, Multiple lacunar infarctions Ischemic periventricular leukoencephalopathy |
Risk factors | Aging, Stroke, Hypertension, Diabetes, Smoking High serum homocystein, Apolipoprotein E4 Amyloid precursor protein, Presenilin 1, Presenilin2 |
Aging, Stroke, Hypertension, Diabetes, Smoking Coronary artery disease, Hyperlipidemia |
AD is the major form of dementia affecting older people and accounts for 60% to 65% of dementia cases. Autopsy series from dementia clinics report that coexisting vascular pathology occurs in 24% to 28% of AD cases [75]. There is emerging evidence that the cascade of events leading to the development of AD brain plaques and tangles may be due to ischemia resulting from cerebrovascular disease (CVD) [22]. The association of the apolipoprotein E (APOE) ε4 genotype with an increased risk for both AD and CVD further suggests a potential link between CVD and AD [39] (Table 1). Conversely, amyloid deposition in cerebral blood vessels due to AD increases the risk for hemorrhagic strokes and subsequent VaD [12]. These common pathways leading from CVD to both AD and VaD support the notion that when there is evidence of both CVD and a gradual progressive dementia, the illness should be conceptualized as the coexistence of interacting pathological processes resulting in mixed dementia.
Stroke or symptoms of transient brain ischemia have long been viewed as the hallmark expression of CVD (Table 1). A considerable number of cerebral infarcts are clinically silent although they share the same risk factors as clinically apparent stroke [10]. The fact that white matter hyper-intensities (WMH) significantly predict future stroke [83] and mortality [131] and are associated with evidence of disease to the carotids [76] and other organ systems [20] lends further support to the notion that WMH are part of a spectrum of vascular related brain injury [20]. Finally, vascular risk factors are also significantly associated with cerebral atrophy in humans [118] and animal models [35], and at least one hypothesis suggests that atrophy results from an ischemic process [81]. In conclusion, it appears that vascular factors lead to a spectrum of a symptomatic brain injury. These various forms of brain injury also affect behavior in a variety of ways, further expanding the clinical relevance of vascular factors and the potential interaction between AD and VaD. In this review, arachidonate cascade in the central nervous system (CNS) is focused as one of the common pathological processes in AD and VaD.
1. CLASSIFICATION OF DEMENTIA
1.1. Alzheimer’s Disease
AD is the fourth largest cause of death for people over 65 years of age. AD is a neurodegenerative disease characterized primarily by cognitive impairment and secondarily by motor dysfunction (Table 1). AD is the most common cause of late-life dementia [152]. AD can be divided into an early-onset form (onset<60 years) and the more common late-onset (>60 years) form. Although most cases of AD are thought to be sporadic, the risk factors for AD include stroke [11], hypertension [11], diabetes [104], smoking [105] and high serum homocysteine [58]. All these conditions have a vascular involvement and are known to reduce cerebral perfusion. ApoE ε4 is associated with the late-onset AD [17]. Three genes are linked to early-onset AD including the β-amyloid precursor protein (APP) on chromosome 21 [68], presenilin-1 (PS1) on chromosome 14 [124], and presenilin-2 (PS2) on chromosome 1 [69]. AD is defined pathologically by extracellular neuritic plaques comprised of fibrillar deposits of a 4 kDa hydrophobic polypeptide known as β-amyloid (Aβ) and NFTs consisting of paired helical filaments (PHF) of hyperphosphorylated tau (HP-Tau) (Table 1). Other pathologic hallmarks of AD activated microglia, reactive astrocytes, and neuronal cell loss [126].
Aβ deposition is an invariant feature of AD, and there is a strong correlation between the amyloid burden in the cortex and the degree of cognitive impairment in the patient [127]. In most AD cases, Aβ peptides also form some deposits in the cerebrovasculature, leading to cerebral amyloid angiopathy and hemorrhagic stroke. Plaques consist of aggregated Aβ peptides that are produced by proteolytic cleavages of the amyloid precursor protein (APP) by the action of proteases called β-and γ-secretases (Fig. 1). As a result of APP processing, neurons produce Aβ peptides of different sizes, the most prominent being 40 and 42 amino acids long (Aβ40 and Aβ42, respectively). The majority of all Aβ isoforms produced is Aβ40, but about 10% is Aβ42 [172]. Cleavage of the APP may occur via two pathways: (1) the normal pathway involves α-secretase and does not lead to formation of Aβ and (2) an alternative pathway, which involves β- and γ-secretases and liberates Aβ, a peptide associated with the neurodegeneration seen in AD [99]. γ-Secretase is a membrane protease complex that possesses presenilin as a catalytic subunit. Nicastrin (NCT) [71] and APH-1 [67] initially form a subcomplex to bind and stabilize presenilin. Then, Pen-2 confers the γ-secretase activity [67] and facilitates endoproteolysis of presenilin. There are both secretory and intracellular pools of Aβ. Soluble extracellular Aβ40/42 forms aggregated Aβ40/42 or is uptaken. Aβ produced intraneuronally and reuptaJien contributes to the intracellular Aβ accumulation.
Fig. (1).
The amyloid β protein cascade in AD. Scheme showing the Aβ hypothesis in AD, which is based on biochemical, neuropathological, molecular-biological and genetic evidences. Aβ: amyloid β protein, APP: amyloid precursor protein, fAβ: fibrillar amyloid β protein, GSK: glycogen synthase kinase, HP-Tau: hyperphosphorylated tau, MAPK: mitogen-activated protein kinase, PHF: paired helical filaments, NCT: nicastrin, PS: presenilin.
The intracellular Aβ staining is most evident in AD-vulnerable brain regions, pyramidal neurons of the hippocampus and entorhinal cortex [37]. Intracellular Aβ deposition is detected prior to the appearance of PHF-positive structures, indicating that it precedes both NFT and Aβ plaque deposition [26] (Fig. 1). Furthermore, Aβ42 oligomers are observed within abnormal processes and synaptic compartments in human AD brains [134]. Aβ42 oligomers turn out to be potent neurotoxins in neuronal organotypic cultures at nanomolar concentrations and can inhibit hippocampal long-term potentiation (LTP) and disrupt synaptic plasticity [64]. The activity-dependent synaptic plasticity such as LTP is a critical component of the neural mechanisms underlying learning and memory [86].
Transgenic (Tg) mice are used as animal models for AD amyloidosis. For example, Tg2576 mice develop characteristic AD-like Aβ brain deposits because of overexpression of a human APP transgene with a double mutation found in a Swedish family with early onset AD [48]. Urine, plasma, and CNS levels of lipid peroxidation are increased as early as 8 months of age in Tg2576 mice compared with WT mice, preceding the onset of Aβ deposition and a surge in CNS Aβ40/42 in the Tg2576 mice. On the other hand, the development of extrapyramidal motor dysfunction in AD has been associated with coexistent Parkinson’s disease [23] and the Lewy body variant of AD [41], and is related to concomitant synucleinopathy (Fig. 1).
The primary genetic risk factor for AD is the inherited complement of alleles for ApoE (Table 1). In comparison to the risk level of the most common ε3/ε3 genotype, the ε4 and ε2 alleles of the human ApoE gene, respectively, increase and decrease sporadic AD risk with 65% of AD attributable to the presence of ε4 and an additional 23% attributable to the absence of an ε2 [17]. Using hippocampal slices from ApoE knockout (ApoE-KO) and human ApoE2, E3, and E4 targeted replacement (ApoE-TR) mice, we found that oligomeric Aβ42 inhibited LTP with a hierarchy of susceptibility mirroring clinical AD risk (ApoE4-TR > ApoE3-TR = ApoE-KO > ApoE2-TR), and that comparable doses of unaggregated Aβ42 did not affect LTP. These data provide a novel link among ApoE isoform, Aβ42, and a functional cellular model of memory. In this model, ApoE4 confers again of negative function synergistic with Aβ42, ApoE2 is protective, and the ApoE Aβ interaction is specific to oligomeric Aβ42 [139].
Fig. 2 shows one of mechanisms involved in the neurotoxicity of Aβ. Although soluble Aβ exhibits low toxicity, fibrillar Aβ (fAβ), an Aβ conformation similar to that found in the AD brain, greatly increase Aβ toxicity in neuronal cultures [140]. fAβ generates reactive oxygen species (ROS) [142], which cause membrane lipid peroxidation and disturb the integrity of neuronal membranes.
Fig. (2).
Steps involved in neurodegeneration of AD. Scheme showing the Aβ hypothesis in AD, which is based on novel findings on the role of arachidonate cascade. AA: arachidonic acid, AMPA/KAR: α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid/ kainate receptor, APP: amyloid precursor protein, COX-2: cyclooxygenase-2, cPLA2, cytosolic phospholipase A2, fAβ: fibrillar amyloid β protein, GSK: glycogen synthase kinase, HP-Tau: hyperphosphorylated tau, MAPK: mitogen activated protein kinase, mAβ: amyloid β protein monomer, Na+/GluT: Na+-dependent glutamate transporter, NFTs: neurofibrillary tangles, NMDAR: N-methyl-D-aspartate receptor, oAβ: amyloid β protein oligomers, PL: phospholipids, ROS: reactive oxygen species, sAβ: soluble amyloid β protein, Glu: glutamate, L-VSCC: L-type voltage-sensitive Ca2+ channel.
These free radicals stimulate L-type voltage-sensitive Ca2+ channel (L-VSCC), and potentiate the influx of Ca2+ into neurons [142]. Increased oxidative stress is an early event in AD that decreases with disease progression and formation of characteristic lesions [100]. Products of lipid peroxidation show a significant increase of intracellular Aβ production [107].
Glutamate transporters on the glial membrane are Na+-dependent and transmembrane gradient of Na+ and K+ is the driving force for the transport (Fig. 2). Aβ-induced elevation of [Ca2+]i causes further Ca2+ release from endoplasmic reticulum by IP3-signalling. These changes in glial membrane conductance reduces glial glutamate uptake by blockade of Na+-dependent glutamate transporters (Na+/Glu transporter) [43]. Glutamate acts at three types of ionotropic receptors on post-synaptic membrane. These receptors have been named after their agonists as N-methyl-D-aspartate (NMDA), α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) and kainate (KA) receptors. NMDA receptors are present mainly in neurons and their activation results into Ca2+ influx [45], whereas AMPA and KA receptors are present in neurons as well as in glia and cause Na+-influx and K+-efflux on activation [80]. This increases glutamate concentration in synaptic cleft beyond physiological levels [130], causing calcium excitotoxicity-induced neuronal death by activating NMDA receptors on post-synaptic neuronal membrane.
The increment in intracellular Ca2+ concentration ([Ca2+]i) affects Ca2+-dependent enzymes including cytosolic phospholipases A2 (cPLA2s), nucleases, kinases and proteases (Fig. 2). These enzymes are involved in fAβ-induced neuronal cell death, which is typified by several features characteristic of apoptosis [141, 157–161]. Besides Ca2+-dependent protein kinases, fAβ stimulates various kinases including mitogen-activated protein kinase (MAPK) and glycogen synthase kinase (GSK) [136]. MAPK phosphorylates and activates cPLA2 [73], up-regulates cyclooxygenase-2 (COX-2) via NF-κB [56]. cPLA2 mediates apoptosis by Aβ [63]. Tau protein is phosphorylated by MAPK [114] and GSK [109]. The hyperphosphorylated tau (HP-Tau) protein form NFTs resulting in the impaired axonal transport (Fig. 2). The NFTs of microtubules and HP-Tau proteins are the major pathological lesions in AD brains [8].
1.2. Vascular Dementia
There are several features of VaD distinguished from AD (Table 1). In the initial stages of VaD, major impairments of short-term memory are not always observed unlike in AD. The cholinergic system is involved in VaD similar to that seen in AD, however. Stroke-prone spontaneously hypertensive rats, an animal model of VaD, have impaired cholinergic function that appears to correlate with deficits in learning and memory function [60]. VaD is the second most common form of dementia, ranking after AD, and is likely to have an increasing impact in an aging population [29, 103]. In the Rotterdam study [103], investigators found that 6.3% of study participants aged 55 to 106 years met the criteria for dementia, but the frequency increased exponentially with age to 43.2% at age 95 years and greater. While AD is the predominant diagnosis in that oldest population (AD: 72%, vascular dementia: 16%), [103] studies of eastern populations, such as those in Japan, China, and Russia, have suggested that vascular dementia may be more common in these populations than AD [29]. In other surveys, prevalence of dementia varied from 0.3% to 1% in persons aged 60 to 64 years and doubled every 5 years, to about 50% in persons older than 95 years [78].
VaD can result from ischemic or hemorrhagic brain damage. The three most common mechanisms causing this disease are single, strategically placed infarcts; multiple cortical infarcts; and subcortical small-vessel disease [7]. These may occur individually or in combination and result in neuropathology that comprises combinations of multiple large infarcts, single strategic infarcts, lacunes, and white-matter lesions. Single strategic infarcts are of particular note because damage in critical locations such as the anteromedial thalamus can cause significant disability in the absence of sensory and motor problems. Because of the variety of pathogenic mechanisms involved in vascular dementia, clinical manifestations can be heterogeneous. Clinical deficits are determined by the size, location, and type of cerebral damage. Common focal neurologic signs include hemiparesis, visual field defects, pseudobulbar palsy, and hemisensory loss. Neurologic symptoms include visual disturbances, brainstem abnormalities, and sensory or motor symptoms (Table 1). The degree of loss of cognition and other function is the sum of various single deficits caused by individual infarcts and hemorrhages. Cognitive and behavioral abnormalities may progress in a stepwise or fluctuating manner and are often accompanied by motor and sensory abnormalities.
Stroke is the potent risk factor for VaD identified in epidemiologic data [54] (Table 1). Stroke is caused by a critical alteration of blood flow to a region of the brain. An acute obstruction of an artery results in ischemia i.e., insufficient blood flow to the tissue [122]. The ischemic brain suffers a mismatch between its cellular energy demands and the ability of the vascular system to supply substrate, most importantly oxygen. Subsequently, neurologic malfunctions and neuronal cell death are caused by increased intracellular calcium, excessive extracellular glutamate, free radicals, and inflammation (Fig. 3). At the beginning of the stroke, there is a definite gradation of injury-a central area or core, with low blood flow already showing signs of massive cell death, and an outer area, the penumbra, that is still alive, but will malfunction after several days. A rat with the middle cerebral artery (MCA) occlusion has been established as an animal model for stroke [144]. MCA occlusion causes irreversible necrosis and infarction in the core [40]. On the other hand, cell death is induced not only via necrosis, but also via apoptosis, and cells remain viable for several hours in the penumbra [70]. Therefore, interventions designed to terminate the reversible proapoptotic state are expected to reduce the ischemic damage and lead to successful treatment of stroke.
Fig. (3).
Steps involved in neurodegeneration of stroke. Scheme showing the sPLA2 hypothesis in ischemia, which is based on novel findings on the role of arachidonate cascade. AA: arachidonic acid, AMPA/KAR: α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid/ kainate receptor, COX-2: cyclooxygenase-2, cPLA2, cytosolic Phospholipase A2, Glu: glutamate, IL-1: interleukin-1, MAPK: mitogen activated protein kinase, Na+/GluT: Na+-dependent glutamate transporter, NMDAR: N-methyl-D-aspartate receptor, PL: phospholipids, ROS: reactive oxygen species, sPLA2: secretory phospholipase A2, sPLA2R: secretory Phospholipase A2 receptor, TNP-α: tumor necrosis factor- α, L-VSCC: L-type voltage-sensitive Ca2+ channel.
2. PHOSPHOLIPASE A2
PLA2 (EC3.1.1.4.) belongs to a family of enzymes that catalyze the cleavage of fatty acids from the sn-2 position of glycerophospholipids to produce free fatty acids and lysophospholipids [5,145]. PLA2s participate in a wide variety of physiological processes, including phospholipid digestion, remodeling of cell membranes and host defense. They also take part in other processes by producing precursors of various types of biologically active lipid mediators (Fig. 4), such as prostaglandins (PGs), leukotrienes (LTs), thromboxanes (TXs) and platelet-activating factor (PAF) [5]. In the mammalian system, more than 19 different isoforms of PLA2 have been identified, and different PLA2s have been shown to participate in pathophysiological events related to cell injury, inflammation, and apoptosis [87,110]. According to their biochemical features such as cellular localization, requirement of Ca2+, substrate specificity and the primary structure, these PLA2s are classified into several families, including low molecular weight secretory PLA2 (sPLA2), Ca2+-sensitive arachidonoyl-specific 85-kDa cytosolic PLA2 (cPLA2), Ca2+-independent PLA2 (iPLA2), and PAF-acetylhydrolase [129]. In the mammalian CNS, five sPLA2 (IIA, IIC, IIF, V and XII), cPLA2 (IV) and iPLA2 (VI) have been detected [32, 84, 146] (Table 2).
Fig. (4).
Schematic pathway of prostanoids formation and actions. PGs protect neurons via receptors coupled to Gs in a cAMP-dependent manner. AA: arachidonic acid, AC: adenylate cyclase, COX: cyclooxygenase, DP: PGD2 receptor, EP: PGE2α receptor, FP: PGP2α receptor, Gi: inhibitory GTP-binding protein, Gq: GTP-binding protein stimulating PLC, Gs: stimulatory GTP-binding protein, IP: PGI2 receptor, PG: prostaglandin, PGDS: PGD2α synthase, PGES: PGE2 synthase, PGFS: PGF2α synthase, PGIS: PGI2 synthase, PL: phsopholipid, PLA2: Phospholipase A2, PLC: phospholipase C, TP: TXA2 receptor, TXA2: thromboxane A2, TXAS: TXA2 synthase.
Table 2.
Phsysiochemical Properties of Brain PLA2
sPLA2 | cPLA2 | iPLA2 | |
---|---|---|---|
Brain | IIA, IIC, IIF, XII | IVA, IVB, IVC | VIA-1,VIA-2,VIB |
Molecular mass | 14kDa | 85kDa | SOkDa |
localization | Extracellular | Cytosol | Cytosol |
Calcium | Stimulation | Translo cation | None |
Substrate | Pho sphatidy lcho line | Pho sphati dy lcho 1 ine | Phosphatidylcholine |
Fatty acid | None | Arachidonic acid | Linoleic acid |
2.1. Secretory Phospholipase A2
PLA2s IIA, IIC, IV and VI mRNA appear to be ubiquitously expressed in rat brain, with relatively similar levels in all brain regions (Table 2). PLA2-IIF is induced in brains of mice injected with lipopolysaccharide intra-peritoneally, to stimulate an inflammatory reaction [88]. PLA2-V mRNA is found at high levels in the hippocampus and at low levels in most other areas. Measurement of PLA2s activity levels demonstrates that PLA2-VI constitutes the dominant form of activity in adult rat brain, with the highest levels in the hippocampus and striatum.
Mammalian sPLA2s possess a consensus domain structure consisting of a Ca2+-binding loop and an active center including the His-Asp pair, and are classified into different groups according to the number and positions of cysteine residues as well as their characteristic domain structures. sPLA2-IIA possesses an intramolecular disulfide bond between Cys50 and Cys137, as well as an amino acid C-terminal extension, and sPLA2-IIA does not have a propeptide. sPLA2-IIA is found in many cells and tissues and its expression is modulated by various inflammatory cytokines, such as interleukin 1β (IL-1β) and tumor necrosis factor-α (TNF-α) in astrocytes [55] (Fig. 3). These inflammatory cytokines are increased by cerebral ischemia, leading to the expression of sPLA2-IIA in the ischemic brain [102]. The cortical sPLA2 activity is significantly elevated in response to focal cerebral ischemia [102]. sPLA2-IIA is induced in the cortex, in which the ischemic core and the penumbra coexist [74]. The sPLA2 inhibitor, indoxam, reduces the elevated PLA2 activity completely and the infarct volume significantly in the cortex [102]. Indoxam ameliorated occlusion-induced inflammation and neurodegeneration in the penumbra, suggesting that sPLA2 might play an important role in apoptosis in the penumbra [102]. As shown in Fig. 3, sPLA2-IIA potentiates Ca2+ influx through L-VSCC [163] and glutamate receptors [62], and induces neuronal cell death via apoptosis [74]. Although sPLA2-IB has not yet been detected in the brain, high-affinity binding sites of sPLA2-IB exist on the plasma membrane of cortical neurons [164]. sPLA2-IB also induces the excess influx of Ca2+ into neurons via L-VSCC [165], and subsequently neuronal cell death via apoptosis [166]. It is unclear whether the high-affinity binding sites of sPLA2-IB are sPLA2 receptor or not (Fig. 3).
2.2. Cytosolic Phospholipase A2
cPLA2 belongs to the group IV PLA2s (Table 2). Although four isoforms, i.e., cPLA2α, -β, -γ and -δ, have been identified, the 85 kDa cPLA2α has been studied most extensively. cPLA2 requires micromolar Ca2+ for activity; however unlike sPLA2, calcium is necessary for binding cPLA2 to membrane or phospholipids vesicles rather than for catalysis. An increase in intracellular Ca2+ concentration promotes binding of Ca2+ ions to the C2 domain and then allows cPLA2α to translocate from the cytosol to the perinuclear region including the Golgi apparatus, endoplasmic reticulum and nuclear envelope [36,123]. Activation of cPLA2 proceeds primarily through phosphorylation [73,91]. This protein is comprised of a C2 domain and multiple phosphorylation sites, including two consensus sites (Ser505 and Ser727) for phosphorylation by mitogen-activated protein kinases (MAPKs) [73] and a Ser515 site for Ca2+/calmodulin-dependent protein kinase [91].
In neurons, cPLA2 is known to control fundamental processes, including neurotransmitter release and long-term potentiation. Both functions are altered in the early stages of AD [24]. Using hippocampal organotypic cultures, a study has shown that cPLA2 mediates apoptosis induced by ischemic insults [4]. Following a transient ischemic episode, cPLA2 immunostaining has been observed in astrocytes or reactive glial cells of the rat brain [16], and an increased expression of the gene for cPLA2 was observed in neurons of the hippocampus [106]. cPLA2 immunoreactive astrocytes are detected in regions of the cortex that contained numerous amyloid β deposits [132].
2.3. Ca2+-independent Phospholipase A2
The iPLA2 family is comprised of group VIA and VIB (Table 2). Group VIA enzyme has at least five splice variants, all with ankyrin repeats, whereas group VIB iPLA2 lacks ankyrin repeats but consists of a signal motif for peroxisome localization [89]. In normal rat brain, >70% of PLA2 activity can be attributed to iPLA2s. iPLA2 is the dominant PLA2 in the cytosolic fraction, and generally regarded as house keeping enzymes for the maintenance of membrane phospholipids [169]. iPLA2 display a substrate preference toward the fatty acid chain in the sn-2 position of phosphatidylcholine of linoleyl > palmitoyl > oleoyl > arachidonyl, and a head group preference for choline >= ethanolamine » inositol. iPLA2 activity is significantly decreased in the dorsolateral prefrontal, but not the lateral temporal cortex of the frontal AD case, a variant of AD [137]. The frontal cortex is not typically the most damaged brain in AD. In typical AD cases, the density of NFTs is greatest in the entorhinal cortex and adjoining hippocampal formation [6]. In frontal AD, however, the density of NFTs in the prefrontal cortex is as high as or higher than in the entorhinal cortex [57].
3. CYCLOOXYGENASE
COX is classified into three forms, a constitutive form, COX-1, a mitogen-inducible form, COX-2 and a variant of COX-1, COX-3 [25]. COX-1 is expressed intensely in microglia in rat brain and in control human brains as well as in hippocampal and cerebral cortical neurons [21]. A COX-3 mRNA is abundant in the human cerebral cortex. In contrast, COX-2 is absent in the control human brains but is induced robustly in neuronal cell bodies and dendrites in peri-infarct regions during the acute phase of focal ischemic injury [138]. The COX-2 gene is expressed throughout the rat forebrain, with higher levels in the cerebral cortex and hippocampus in discrete populations of neurons, but not in glia [170]. COX-2 is localized in neuronal dendritic spines where active synapses are present [59]. In the brain, basal expression of COX-2 has been shown to be regulated by synaptic activity, and its expression is upregulated by a high frequency stimulation that is associated with LTP [170]. COX-2 inhibitors significantly reduce postsynaptic membrane excitability, back-propagating dendritic action potential-associated Ca2+ influx and LTP induction in hippocampus dentate granule neurons, while a COX-1 inhibitor is ineffective [15]. These studies suggest that COX-2 participates in synaptic modification.
Epidemiology has pointed to nonsteroidal anti-inflammatory drugs (NSAIDs) reducing the risk of developing AD [65]. The beneficial actions of NSAIDs are linked to inhibition of the inducible COX-2 activity at inflammatory sites. On the other hand, their side effects (e.g., gastric damage) are ascribed to suppression of the constitutive COX-1 activity. Clinical trials of AD patients with a COX inhibitor, indomethacin, indicated a beneficial effect [79,115]. COX-2 is up-regulated in the brain of AD patients [108] and transgenic mouse models [86,87]. COX-2 is induced in Aβ-treated SH-SY5Y neuroblastoma cells [108]. COX-2 selective inhibitors, e.g. S-2474, prevent cerebral cortical neurons from undergoing Aβ-induced apoptosis [157]. Slowing of cognitive decline in patients with mild to moderate AD over 12 months have not yet been detected in the trial of COX-2 inhibitors, rofecoxive and naproxen, however [49]. COX-3 is elevated in AD and in the primary culture of IL-1β/Aβ1-42-treated human neural (HN) cells [19]. The inflammation in AD and in stressed HN cells, COX-3 may play ancillary roles in membrane-based COX signaling or when basal levels of COX-1 or COX-2 expression persist.
Infarcts in the MCA up-regulate COX-2 in the brain of stroke patients [50,117]. In the animal model, gene expression of COX-2 is induced in the ischemic brain [98]. Resting cerebral blood flow is decreased [97], and brain ischemia/reperfusion injury is increased [51] in COX-1-deficient mice. In contrast, COX-2 deficient mice display reduced susceptibility to ischemic injury and NMDA neurotoxicity [52]. The COX-2 selective inhibitors reduce infarct size following focal ischemia in rats [98] and effectively limit hippocampal damage following forebrain ischemia in the gerbil [14]. In the primary culture, another COX-2 selective inhibitor, S-2474, rescues cortical neurons from undergoing sPLA2-IIA-induced apoptosis [157]. Thus, the pathway through PLA2 and COX-2 plays an important role in neurodegeneration after ischemia.
4. PROSTAGLANDIN
Eicosanoids are divided into two groups according to their mechanism of action: the conventional eicosanoids, e.g., PGD2 and PGE2, the cyclopentenone-type PGs (Fig. 4), e.g., PGJ2 and PGA2 (Fig. 5). PGD2 and PGE2 are further dehydrated to produce PGJ2 and PGA2, respectively [101] (Fig. 5). Once AA has been supplied, both isoforms of COX form PGG2 and PGH2 via identical enzymatic processes (Fig. 4). Following these steps, PGH2 can be metabolized by different enzyme pathways to a range of products with potent biological effects. The profile of products made by cells expressing COX-1/COX-3 or COX-2 is therefore determined by the presence of different downstream enzymes. Eicosanoids modulate cellular function during a variety of physiologic and pathologic processes [94].
Fig. (5).
Actions of PGD2 and PGE2 metabolites to neurons. 15d-Δ12,14-PGJ2 mediates the neurotoxicity of PGD2 independently from PPAR-γ, probably via the binding sites of 15d-Δ12,14-PGJ2 on plasma membranes. AC: adenylate cyclase, DP1: PGD2 receptor coupled to Gs, DP2: CRTH2 receptors, Gi: inhibitory GTP-binding protein, Gs: stimulatory GTP-binding protein, PG: prostaglandin, PKA: cAMP-dependent protein kinase, 15d-Δ12,14-PGJ2: 15-deoxy-Δ12,14-PGJ2, PPARγ: peroxisome proliferator-activated receptorγ, JBS: binding site of 15-deoxy-Δ12,14-PGJ2 on plasma membranes.
4.1. Prostaglandin E2
Earlier it appeared that when cells expressed large amounts of COX-2, the PGH2 formed could be saturating for the PG synthase enzymes resulting in the formation of proportionately larger amounts of PGE2 [82], possibly by non-enzymatic conversion (Fig. 4). In addition to the cytosolic PGE synthase, there is an inducible microsomal or membrane-associated perinuclear PGE synthase (mPGES) regulated, for instance, by proinflammatory cytokines and glucocorticoids. While the cytosolic PGE synthase is principally coupled with COX-1, this inducible mPGES appears coupled with COX-2 [90]. PGES is induced by Aβ in astrocytes [121]. PGE2 is elevated in the cerebrospinal fluids of living AD patients [85] and in the postischemic brain [171]. PGE2 synthesis is reduced in the postmortem cerebral cortices of AD patients [153]. In in vitro models for AD, PGE2 rescues neurons from the toxicity of Aβ [160] and TNFα [66]. Intracerebroventricular injection of naturally secreted human Aβ inhibited LTP [148], a correlate of learning and memory [86]. PGE2 reverses COX-2 inhibitor-induced suppression of postsynaptic membrane excitability, back-propagating dendritic action potential-associated Ca2+ influx and LTP in hippocampal dentate granule neurons [15], indicating that PGE2 also participates in synaptic modification.
Four PGE2 receptor subtypes, i.e., EP1, EP2, EP3, and EP4, have been identified [92]. Activation of EP1 produces phosphatidylinositol and increases intracellular Ca2+ concentration ([Ca2+]i) [96]. EP2 and EP4 are coupled to Gs, adenylate cyclase (AC) activation and cAMP generation [96]. There are at least three variants of EP3, and these variants can cause increased [Ca2+]i or the activation or inhibition of AC [92]. PGE2 is suggested to be involved in the NMDA-mediated component of oxygen-glucose deprivation toxicity via the activation of the prostanoids EP1 and/or EP3 [34]. In rodent in vivo and in vitro models for stroke, PGE2 exhibits neuroprotective function against cerebral ischemia via its receptor, EP2 [77] and glutamate toxicity [2]. In addition to neurons and astrocytes, EP2 is expressed in the microglia [13]. The role of microglial cells in the AD brain is controversial, as it remains unclear if the microglial cells form the amyloid fibrils of plaques or react to them in a macrophage-phagocytic role. Although activated microglia can secrete neurotoxic factors to surrounding neurons, PGE2 reduces microglial-induced neurotoxicity via EP2 [13]. On the other hand, microglial cells specifically lacking EP2 increases Aβ phagocytosis and decrease Ap-induced damage to neurons [127]. Thus, microglia play dual roles in secretion of neurotoxins and phagocytosis of amyloid.
4.2. Prostaglandin F2α
PGF2α is synthesized via three pathways from PGE2, PGD2, or PGH2 by PGE 9-ketoreductase, PGD 11-ketoreductase, or PGH 9-, 11-endoperoxide reductase, respectively [149] (Fig. 4). Although there are no reports of PGF synthase being inducible, its levels in the uterus may increase during pregnancy associated with the peak of PGF2α production that accompanies parturition [151]. Oxidative damage to the CNS predominantly manifests as lipid peroxidation (LPO) because of the high content of polyunsaturated fatty acids that are particularly susceptible to oxidation. 8-iso-PGF2α is an isoprostane (iP). Isoprostanes are specific and sensitive markers of in vivo LPO [111]. Levels of a major isoprostane, 8, 12-iso-iPF2α-V I, are increased not only in specific AD brain regions [112] but also in the urine, plasma, and CSF of patients with a clinical diagnosis of AD [113]. Plasma levels of isoprostanes were significantly higher among stroke patients than control [119]. PGF2α receptor FP is coupled with Gq, stimulates phospholipase C (PLC), and increases [Ca2+]i [93]. Although PGF2α exhibits no neurotoxicity in the primary culture of rat cortical neurons [74], it is unclear whether PGF2α is a neurotoxic or neuroprotective factor.
4.3. Prostaglandin I2
PGI2 is produced from PGH2 by the action of PGI synthase, a member of the P450 superfamily [42] (Fig. 4). Plasma levels of PGI2 were significantly higher among stroke patients than control [119]. PGI2 receptor (IP) is coupled to AC via Gs [43]. PGI2 is a potent vasodilator, and has an antithrombotic effect via IP1. PGI2 exhibits protective effects for ischemic neuronal damage because it improves cerebral circulation [38]. Another type of PGI2 receptor, IP2, is in the CNS [135]. In the rat brain, IP2 receptor is expressed in the hippocampus, cerebral cortex, thalamus, striatum, and septum, and the IP1 receptor is expressed in the caudal medulla such as the nucleus of the solitary tract. IP receptor ligands prevent the death of hippocampal neurons in vitro and protect CA1 pyramidal neurons against ischemic damage in vivo [120].
4.4. Thromboxane A2
TXA2 is metabolized from PGH2 by tromboxane synthase [95] (Fig. 4). An elevated level of TXA2 has been found in the postmortem brains of AD patients [55]. TXA2 is suggested to be involved in the extrapyramidal motor dysfunction of patients with AD [167]. Patients with poststroke dementia more often show increased thromboxane biosynthesis than nondemented stroke patients [147]. In the brain, TXA2 receptors (TP) are expressed in both neuronal cells and glial cells [30]. TP stimulates PLC via Gq, and elevates [Ca2+]i [93]. Neurotoxicity of TXA2 has not yet been detected in in vitro [162] and in vivo [167] models.
4.5. Prostaglandin D2
PGD synthase (PGDS) exists in both lipocalin-type and hematopoietic forms [143]. The lipocalin type is a major constituent of the human cerebrospinal fluid (CSF), representing 3% of total CSF protein [156]. The hematopoietic form produces PGD2 in cells such as mast cells, basophils, and Th2 cells. PGD2 is increased significantly in the postmortem cerebral cortex of Alzheimer-type dementia patients [55] and an animal model for stroke, gerbil brain during reperfusion after bilateral common carotid artery occlusion [31]. PGD2 elicits its downstream effects by activating two G protein-coupled receptors, the DP and chemoattractant receptor-homologous molecule expressed on Th2 cells (CRTH2) receptors (hereafter termed DP1 and DP2) [1,46,47]. The DP1 and DP2 receptors have divergent effects on cAMP production, and are coupled to Gs and Gi respectively [18,44]. At physiological concentrations from 1 nM to 1 μM, PGD2 protects hippocampal neurons from glutamate toxicity via DP1 in a cAMP-dependent manner [72]. The shared neuroprotection of the DP1, EP2 and IP2 receptors, which are coupled to cAMP, would suggest that other prostaglandin receptors similarly positively coupled to cAMP production, such as the PGE2, EP4 receptor, may also promote neuroprotection.
In contrast, at high concentrations (> 1 μM), PGD2 itself causes apoptosis in the primary culture of cortical neurons [168]. BWA868C, a selective DP1 antagonist, does not prevent neurons from PGD2-induced apoptosis. mRNA of DP2 receptor is detected in the forebrain such as cortex and hippocampus [72]. 15-deoxy-Δ12,14-prostaglandin J2 (15d-Δ12,14-PGJ2), a selective agonist for DP2 receptor, is neurotoxic [116], suggesting a possible involvement of DP2 in the neurotoxicity of PGD2. However, several evidences do not support the possibility. First, few specific binding sites of [3H]PGD2 are detected in plasma membranes from rat cortices [168]. Second, the extent of specific [3H]PGD2 in total binding is very low (30-40%), although binding sites of PGD2 have been reported in synaptosomes of rat [128] and human brains [150]. Third, another selective agonist for DP2 receptor, 15d-Δ12,14-PGD2, exhibits no neurotoxicity [168]. Fourth, PGD2 requires a latent time to cause neuronal apoptosis [168]. Finally, the ED50 value (8.2 μM) of PGD2 is much higher than the affinity for DP2 receptor (binding affinities (Ki) = 24 nM) [94]. These discrepancies suggest that the neurotoxicity of PGD2 is mediated by other pathways besides DPs.
4.6. Prostaglandin J2 and Prostaglandin A2
PGD2 is non-enzymatic ally dehydrated to produce PGJ2, Δ12-PGJ2, and 15d-Δ12,14-PGJ2 [27,168] (Fig. 5). As well as PGD2, 15d-Δ12,14-PGJ2 induced neuronal cell death via apoptosis [116,168]. 15d-Δ12,14-PGJ2 is a ligand for the nuclear receptor, peroxysome proliferators-activated receptory (PPARγ) [28,61]. Activation of PPARγ protects the brain from cerebral ischemia [133] and Aβ-dependent neurodegeneration [53]. Thus, l5d-Δ12,14-PGJ2 causes neuronal cell death via the pathway independent from PPARγ. Novel binding sites of 15d-Δ12,14-PGJ2 are found in the neuronal plasma membranes of the cerebral cortex [168], and are tentatively termed JBS, here (Fig. 5). The specific binding sites of [3H]15d-Δ12,14-PGJ2 (>80%) in total binding is much higher than that of [3H]PGD2 (30-40%). The affinity for JBS is 15d-Δ12,14-PGJ2 > Δ12-PGJ2 > PGJ2 » PGD2, in the same sequence as the neurotoxic potency. Another neurotoxic cyclopentenone PG, PGA2, is metabolized from PGE2, and also binds JBS [168]. None of the other eicosanoids, PPAR-agonists, or a PGD2 receptor blocker exhibits any affinity for the binding site and the neurotoxicity. There are close correlation between the affinity of eicosanoids for JBS and their neurotoxicity, indicating the involvement of JBS in neuronal apoptosis.
CONCLUSION
Dementia is principally diagnosed to AD, VaD and the mixed dementia, suggesting common causative mechanisms (Fig. 6). In these diseases, free radicals and glutamate are released and stimulate L-VSCCs and glutamate receptors, respectively. Excess influx of Ca2+ disrupts intracellular calcium homeostasis. Arachidonate cascade is one of the downstreams of the second messenger, calcium ion, and is the important pathway in common with AD and VaD. Among various enzymes in the cerebral arachidonate cascade, cPLA2, COX-2 and PGDS are the key enzymes involved in the development of the two neurologic diseases. Among PGs, conventional types, PGD2 and PGE2, mediate neuronal protection via their receptors coupled to Gs in a cAMP-dependent manner. On the other hand, PGD2 and PGE2 are metabolized to neurotoxic cyclopentenone types, PGJ2 and PGA2, respectively. 15d-Δ12,14’ -PGJ2 mediates the COX-2 neurotoxicity independent from PPARγ, probably via the binding sites of 15d-Δ12,14-PGJ2 on the plasma membrane. Thus, one of causative mechanisms of AD and VaD are merged into cerebral arachidonate cascade. Further studies are required to clear the mechanism in which 15d-Δ12,14-PGJ2 induces neuronal apoptosis. The present review sheds light on the agonists and antagonists for enzymes and receptors in the arachidonate cascade as targets for therapy in AD and VaD.
Fig. (6).
Common causative mechanisms of AD and VaD. Scheme showing Aβ and sPLA2 hypothesis in AD and VaD, respectively, which is based on novel findings on the role of arachidonate cascade. Aβ: amyloid β protein, AMPA/KAR: α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid/ kainate receptor, COX-2: cyclooxygenase-2, cPLA2, cytosolic phospholipase A2, 15d-Δ12,14-PGJ2: 15dexoy-Δ12,14-prostaglandin J2, L-VSCC: L-type voltage-sensitive Ca2+ channel, NMDAR: N-methyl-D-aspartate receptor, PGD2:prostaglandin D2, sPLA2: secretory phospholipase A2.
REFERENCES
- 1.Abe H, Takeshita T, Nagata K, Arita T, Endo Y, Fujita T, Takayama H, Kubo M, Sugamura K. Molecular cloning, chromosome mapping and characterization of the mouse CRTH2 gene, a putative member of the leukocyte chemoattractant receptor family. Gene. 1999;227:71–77. doi: 10.1016/s0378-1119(98)00599-x. [DOI] [PubMed] [Google Scholar]
- 2.Akaike A, Kaneko S, Tamura Y, Nakata N, Shiomi H, Ushikubi F, Narumiya S. Prostaglandin E2 protects cultured cortical neurons against N-methyl-D-aspartate receptor-mediated glutamate cytotoxicity. Brain Res. 1994;663:237–243. doi: 10.1016/0006-8993(94)91268-8. [DOI] [PubMed] [Google Scholar]
- 3.American Psychiatric Association. Diagnostic and statistical manual of mental disorders. fourth. Washington D.C.: American Psychiatric Association; 1994. pp. 138–142. [Google Scholar]
- 4.Arai K, Ikegaya Y, Nakatani Y, Kudo I, Nishiyama N, Matsuki N. Phospholipase A2 mediates ischemic injury in the hippocampus: a regional difference of neuronal vulnerability. Eur J Neurosci. 2001;13:2319–2323. doi: 10.1046/j.0953-816x.2001.01623.x. [DOI] [PubMed] [Google Scholar]
- 5.Arita H, Nakano T, Hanasaki K. Thromboxane A2: its generation and role in platelet activation. Prog Lipid Res. 1989;28:273–301. doi: 10.1016/0163-7827(89)90002-7. [DOI] [PubMed] [Google Scholar]
- 6.Arnold SE, Hyman BT, Flory J, Damasio AR, Van Hoesen GW. The topographical and neuroanatomical distribution of neurofibrillary tangles and neuritic plaques in the cerebral cortex of patients with Alzheimer’s disease. Cereb Cortex. 1991;1:103–116. doi: 10.1093/cercor/1.1.103. [DOI] [PubMed] [Google Scholar]
- 7.Black SE. Vascular dementia. Postgrad Med. 2005;117:15–24. doi: 10.3810/pgm.2005.01.1563. [DOI] [PubMed] [Google Scholar]
- 8.Blessed G, Tomlinson BE, Roth M. The association between quantitative measures of dementia and of senile change in the cerebral grey matter of elderly subjects. Br J Psychiatry. 1968;114:797–811. doi: 10.1192/bjp.114.512.797. [DOI] [PubMed] [Google Scholar]
- 9.Boie Y, Sawyer N, Slipetz DM, Metters KM, Abramovitz M. Molecular cloning and characterization of the human prostanoid DP receptor. J Biol Chem. 1995;270:18910–18916. doi: 10.1074/jbc.270.32.18910. [DOI] [PubMed] [Google Scholar]
- 10.Boon A, Lodder J, Heuts-van Raak L, Kessels F. Silent brain infarcts in 755 consecutive patients with a first-ever supratentorial ischemic stroke. Relationship with index-stroke subtype, vascular risk factors, and mortality. Stroke. 1994;25:2384–2390. doi: 10.1161/01.str.25.12.2384. [DOI] [PubMed] [Google Scholar]
- 11.Breteler MM. Vascular risk factors for Alzheimer’s disease: an epidemiologic perspective. Neurobiol Aging. 2000;21:153–160. doi: 10.1016/s0197-4580(99)00110-4. [DOI] [PubMed] [Google Scholar]
- 12.Bugiani O. Aβ-related cerebral amyloid angiopathy. Neurol Sci. 2004;25(1):S1–2. doi: 10.1007/s10072-004-0204-9. [DOI] [PubMed] [Google Scholar]
- 13.Caggiano AO, Kraig RP. Prostaglandin E receptor subtypes in cultured rat microglia and their role in reducing lipopolysaccharide-induced interleukin-1β production. J Neurochem. 1999;72:565–575. doi: 10.1046/j.1471-4159.1999.0720565.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Candelario-Jalil E, Alvarez D, Gonzalez-Falcon A, Garcia-Cabrera M, Martinez-Sancez G, Merino N, Giuliani A, Leon OS. Neuroprotective efficacy of nimesulide against hippocampal neuronal damate following transient ischemia. Eur J Pharmacol. 2002;453:189–195. doi: 10.1016/s0014-2999(02)02422-6. [DOI] [PubMed] [Google Scholar]
- 15.Chen C, Bazan NG. Lipid signaling: Sleep, synaptic plasticity, and neuroprotection. Prostaglandins Other Lipid Med. 2005;77:65–76. doi: 10.1016/j.prostaglandins.2005.07.001. [DOI] [PubMed] [Google Scholar]
- 16.Clements JA, Stephenson DT, Smalstig EB, Roberts EF, Johnstone EM, Sharp JD. Reactive glia express cytosolic Phospholipase A2 after transient global forebrain ischemia in the rat. Stroke. 1996;27:527–535. doi: 10.1161/01.str.27.3.527. [DOI] [PubMed] [Google Scholar]
- 17.Corder EH, Saunders AM, Strittmatter WJ, Schmechel DE, Gaskell PC, Small GW, Roses AD, Haines JL, Pericak-Vance MA. Gene dose of apolipoprotein E type 4 allele and the risk of Alzheimer’s disease in late onset families. Science. 1993;261:921–923. doi: 10.1126/science.8346443. [DOI] [PubMed] [Google Scholar]
- 18.Crider JY, Griffin BW, Sharif NA. Prostaglandin DP receptors positively coupled to adenylyl cyclase in embryonic bovine tracheal (EBTr) cells: pharmacological characterization using agonists and antagonists. Br J Pharmacol. 1999;127:204–210. doi: 10.1038/sj.bjp.0702490. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Cui JG, Kuroda H, Chandrasekharan NV, Pelaez RP, Simmons DL, Bazan NG, Lukiw WJ. Cyclooxygenase-3 gene expression in Alzheimer hippocampus and in stressed human neural cells. Neurochem Res. 2004;29:1731–1737. doi: 10.1023/b:nere.0000035809.70905.8a. [DOI] [PubMed] [Google Scholar]
- 20.DeCarli C, Miller BL, Swan GE, Reed T, Wolf PA, Garner J, Jack L, Carmelli D. Predictors of brain morphology for the men of the NHLBI twins tudy. Stroke. 1999;30:529–536. doi: 10.1161/01.str.30.3.529. [DOI] [PubMed] [Google Scholar]
- 21.Deininger MH, Schluesenger HJ. Cyclooxygenase-1 and -2 are differentially localized to microglia and endothelium in rate EAE and glioma. J Neuroimmunol. 1999;95:202–208. doi: 10.1016/s0165-5728(98)00257-4. [DOI] [PubMed] [Google Scholar]
- 22.de la Torre JC. Vascular basis of Alzheimer’s pathogenesis. Ann N Y Acad Sci. 2002;977:196–215. doi: 10.1111/j.1749-6632.2002.tb04817.x. [DOI] [PubMed] [Google Scholar]
- 23.Ditter SM, Mirra SS. Neuropathologic and clinical features of Parkinson’s disease in Alzheimer’s disease patients. Neurology. 1987;37:754–760. doi: 10.1212/wnl.37.5.754. [DOI] [PubMed] [Google Scholar]
- 24.Farooqui AH, Yang HC, Horrocks L. Involvement of Phospholipase A2 in neurodegeneration. Neurochem Int. 1997;30:517–522. doi: 10.1016/s0197-0186(96)00122-2. [DOI] [PubMed] [Google Scholar]
- 25.Feng L, Sun W, Xia Y, Tang WW, Chanmugam P, Soyoola E, Wilson CB, Hwang D. Cloning two isoforms of rat cyclooxygenase: differential regulation of their expression. Arch Biochem Biophys. 1993;307:361–368. doi: 10.1006/abbi.1993.1601. [DOI] [PubMed] [Google Scholar]
- 26.Fernandez-Vizarra P, Fernandez AP, Castro-Blanco S, Serrano J, Bentura ML, Martinez-Murillo R, Martinez A, Rodrigo J. Intra- and extracellular Aβ and PHF in clinically evaluated cases of Alzheimer’s disease. Histol Histopathol. 2004;19:823–844. doi: 10.14670/HH-19.823. [DOI] [PubMed] [Google Scholar]
- 27.Fitzpatrick FA, Wynalda MA. Albumin-catalyzed metabolism of prostaglandin D2. J Biol Chem. 1983;258:11713–11718. [PubMed] [Google Scholar]
- 28.Forman BM, Tontonoz P, Chen J, Brun RP, Spiegelman BM, Evans RM. 15-deoxy-Δ12,14-prostaglandin J2 is a ligand for the adipocyte determination factor PPARγ. Cell. 1995;83:803–812. doi: 10.1016/0092-8674(95)90193-0. [DOI] [PubMed] [Google Scholar]
- 29.Fratiglioni L, De Ronchi D, Aguero-Torres H. Worldwide prevalence and incidence of dementia. Drugs Aging. 1999;5:365–375. doi: 10.2165/00002512-199915050-00004. [DOI] [PubMed] [Google Scholar]
- 30.Gao H, Peng B, Welch WJ, Wilcox C. Central thromboxane receptors: mRNA expression and mediation of pressor responses. Am J Physiol. 1997;272:R1493–R1500. doi: 10.1152/ajpregu.1997.272.5.R1493. [DOI] [PubMed] [Google Scholar]
- 31.Gaudet RJ, Alam I, Levine L. Accumulation of cyclooxygenase products of arachidonic acid metabolism in gerbil brain during reperfusion after bilateral common carotid artery occlusion. J Neurochem. 1980;35:653–658. doi: 10.1111/j.1471-4159.1980.tb03704.x. [DOI] [PubMed] [Google Scholar]
- 32.Gelb MH, Valentin E, Ghomashchi F, Lazdunski M, Lambeau G. Cloning and recombinant expression of a structurally novel human secreted Phospholipase A2. J Biol Chem. 2000;275:39823–39826. doi: 10.1074/jbc.C000671200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Gerashchenko D, Beuckmann CT, Kanaoka Y, Eguchi N, Gordon WC, Urade Y, Bazan NG, Hayaishi O. Dominant expression of rat prostanoid DP receptor mRNA in leptomeninges, inner segments of photoreceptor cells, iris epithelium, and ciliary processes. J Neurochem. 1998;71:937–945. doi: 10.1046/j.1471-4159.1998.71030937.x. [DOI] [PubMed] [Google Scholar]
- 34.Gendron TF, Brunette E, Tauskela JS, Morley P. The dual role of prostaglandin E2 in excitotoxicity and preconditioning-induced neuroprotection. Eur J Pharmacol. 2005;517:17–27. doi: 10.1016/j.ejphar.2005.05.031. [DOI] [PubMed] [Google Scholar]
- 35.Gesztelyi G, Finnegan W, DeMaro JA, Wang JY, Chen JL, Fenstermacher J. Parenchymal microvascular systems and cerebral atrophy in spontaneously hypertensive rats. Brain Res. 1993;611:249–257. doi: 10.1016/0006-8993(93)90510-t. [DOI] [PubMed] [Google Scholar]
- 36.Glover S, de Carvalho MS, Bayburt T, Jonas M, Chi E, Leslie CC, Gelb MH. Translocation of the 85-kDa Phospholipase A2 from cytosol to the nuclear envelope in rat basophilic leukemia cells stimulated with calcium ionophore or IgE/antigen. J Biol Chem. 1995;270:15359–15367. doi: 10.1074/jbc.270.25.15359. [DOI] [PubMed] [Google Scholar]
- 37.Gouras GK, Tsai J, Naslund J, Vincent B, Edgar M, Checler F, Greenfield JP, Haroutunian V, Buxbaum JD, Xu H, Greengard P, Relkin NR. Intraneuronal Aβ42 accumulation in human brain. Am J Pathol. 2000;156:15–20. doi: 10.1016/s0002-9440(10)64700-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Gryglewski RJ. Prostacyclin as a circulatory hormone. Biochem Pharmacol. 1979;28:3161–3166. doi: 10.1016/0006-2952(79)90055-8. [DOI] [PubMed] [Google Scholar]
- 39.Haan MN, Shemanski L, Jagust WJ, Manolio TA, Kuller L. The role of APOE ε4 in modulating effects of other risk factors for cognitive decline in elderly persons. J Am Med Assoc. 1999;282:40–46. doi: 10.1001/jama.282.1.40. [DOI] [PubMed] [Google Scholar]
- 40.Hallenbeck JM. Blood-damaged tissue interaction in experimental brain ischemia. Acta Neurochir. 1994;60:233–237. doi: 10.1007/978-3-7091-9334-1_62. [DOI] [PubMed] [Google Scholar]
- 41.Hansen L, Salmon D, Galasko D, Masliah E, Katzman R, DeTeresa R, Thal L, Pay MM, Hofstetter R, Klauber M. The Lewy body variant of Alzheimer’s disease: a clinical and pathologic entity. Neurology. 1990;40:1–8. doi: 10.1212/wnl.40.1.1. [DOI] [PubMed] [Google Scholar]
- 42.Hara S, Miyata A, Yokoyama C, Inoue H, Brugger R, Lottspeich F, Ullrich V, Tanabe T. Isolation and molecular cloning of prostacyclin synthase from bovine endothelial cells. J Biol Chem. 1994;269:19897–19903. [PubMed] [Google Scholar]
- 43.Harris ME, Wang Y, Pedigo NW, Jr., Hensley K, Butterfield DA, Carney JM. Amyloid β peptide (25-35) inhibits Na+-dependent glutamate uptake in rat hippocampal astrocyte cultures. J Neurochem. 1996;67:277–286. doi: 10.1046/j.1471-4159.1996.67010277.x. [DOI] [PubMed] [Google Scholar]
- 44.Hata AN, Zent R, Breyer MD, Breyer RM. Expression and molecular pharmacology of the mouse CRTH2 receptor. J Pharmacol Exp Ther. 2003;306:463–470. doi: 10.1124/jpet.103.050955. [DOI] [PubMed] [Google Scholar]
- 45.Headley PM, Grillner S. Excitatory amino acids and synaptic transmission: the evidence for a physiological function. Trends Pharmacol Sci. 1990;11:205–211. doi: 10.1016/0165-6147(90)90116-p. [DOI] [PubMed] [Google Scholar]
- 46.Hirai H, Tanaka K, Yoshie O, Ogawa K, Kenmotsu K, Takamori Y, Ichimasa M, Sugamura K, Nakamura M, Takano S, Nagata K. Prostaglandin D2 selectively induces chemotaxis in T helper type 2 cells, eosinophils, and basophils via seven-transmembrane receptor CRTH2. J Exp Med. 2001;193:255–261. doi: 10.1084/jem.193.2.255. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Hirata M, Kakizuka A, Aizawa M, Ushikubi F, Narumiya S. Molecular characterization of a mouse prostaglandin D receptor and functional expression of the cloned gene. Proc Natl Acad Sci USA. 1994;91:11192–11196. doi: 10.1073/pnas.91.23.11192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Hsiao K, Chapman P, Nilsen S, Eckman C, Harigaya Y, Younkin S, Yang F, Cole G. Correlative memory deficits, Aβ elevation, and amyloid plaques in transgenic mice. Science. 1996;274:99–102. doi: 10.1126/science.274.5284.99. [DOI] [PubMed] [Google Scholar]
- 49.Hwang DY, Chae KR, Kang TS, Hwang JH, Lim CH, Kang HK, Goo JS, Lee MR, Lim HJ, Min SH, Cho JY, Hong JT, Song CW, Paik SG, Cho JS, Kim YK. Alterations in behavior, amyloid β-42, caspase-3, and Cox-2 in mutant PS2 transgenic mouse model of Alzheimer’s disease. FASEB J. 2002;16:805–813. doi: 10.1096/fj.01-0732com. [DOI] [PubMed] [Google Scholar]
- 50.Iadecola C, Forster C, Nogawa S, Clark HB, Ross ME. Cyclooxygenase-2 immunoreactivity in the human brain following cerebral ischemia. Acta Neuropathol. 1999;98:9–14. doi: 10.1007/s004010051045. [DOI] [PubMed] [Google Scholar]
- 51.Iadecola C, Sugimoto K, Niwa K, Kazama K, Ross ME. Increased susceptibility to ischemic brain injury in cyclooxygenase-1 deficient mice. J Cereb Blood Flow Metab. 2001;21:1436–1441. doi: 10.1097/00004647-200112000-00008. [DOI] [PubMed] [Google Scholar]
- 52.Iadecola C, Niwa K, Nogawa S, Zhao M, Nagayama M, Araki E, Morham S, Ross ME. Reduced susceptibility to ischemic brain injury and N-methyl-D-aspartate-mediated neurotoxicity in cyclooxygenase-2-deficient mice. Proc Natl Acad Sci USA. 2001;98:1294–1299. doi: 10.1073/pnas.98.3.1294. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Inestrosa NC, Godoy JA, Quintanilla RA, Koenig CS, Bronfman M. Peroxisome proliferator-activated receptor γ is expressed in hippocampal neurons and its activation prevents βamyloid neurodegeneration: role of Wnt signaling. Exp Cell Res. 2005;304:91–104. doi: 10.1016/j.yexcr.2004.09.032. [DOI] [PubMed] [Google Scholar]
- 54.in’t Veld BA, Ruitenberg A, Hofman A, Stricker BH, Breteler MM. Antihypertensive drugs and incidence of dementia: the Rotterdam Study. Neurobiol Aging. 2001;22:407–412. doi: 10.1016/s0197-4580(00)00241-4. [DOI] [PubMed] [Google Scholar]
- 55.Iwamoto N, Kobayashi K, Kosaka K. The formation of prostaglandins in the postmortem cerebral cortex of Alzheimer-type dementia patients. J Neurol. 1989;236:80–84. doi: 10.1007/BF00314401. [DOI] [PubMed] [Google Scholar]
- 56.Jang JH, Surh YJ. β-amyloid-induced apoptosis is associated with cyclooxygenase-2 up-regulation via the mitogen-activated protein kinase-NF-κ B signaling pathway. Free Radic Biol Med. 2005;38:1604–1613. doi: 10.1016/j.freeradbiomed.2005.02.023. [DOI] [PubMed] [Google Scholar]
- 57.Johnson JK, Head E, Kim R, Starr A, Cotman CW. Clinical and pathological evidence for a frontal variant of Alzheimer disease. Arch Neurol. 1999;56:1233–1239. doi: 10.1001/archneur.56.10.1233. [DOI] [PubMed] [Google Scholar]
- 58.Kalmijn S, Launer LJ, Lindemans J, Bots ML, Hofman A, Breteler MM. Total homocysteine and cognitive decline in a community-based sample of elderly subjects: the Rotterdam Study. Am J Epidemiol. 1999;150:283–289. doi: 10.1093/oxfordjournals.aje.a010000. [DOI] [PubMed] [Google Scholar]
- 59.Kaufmann WE, Worley PF, Pegg J, Bremer M, Isakson P. COX-2, a synaptically induced enzyme, is expressed by excitatory neurons at postsynaptic sites in rat cerebral cortex. Proc Natl Acad Sci USA. 1996;93:2317–2321. doi: 10.1073/pnas.93.6.2317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Kimura S, Saito H, Minami M, Togashi H, Nakamura N, Nemoto M, Parvez HS. Pathogenesis of vascular dementia in stroke-prone spontaneously hypertensive rats. Toxicology. 2000;153:167–78. doi: 10.1016/s0300-483x(00)00312-7. [DOI] [PubMed] [Google Scholar]
- 61.Kliewer SA, Lenhard JM, Willson TM, Patel I, Morris DC, Lehmann JM. A prostaglandin J metabolite binds peroxisome proliferator-activated receptor γ and promotes adipocyte differentiation. Cell. 1998;83:813–819. doi: 10.1016/0092-8674(95)90194-9. [DOI] [PubMed] [Google Scholar]
- 62.Kolko M, DeCoster MA, Rodriguez de Turco EB, Bazan NG. Synergy by secretory Phospholipase A2 and glutamate on inducing cell death and sustained arachidonic acid metabolic changes in primary cortical cultures. J Biol Chem. 1996;271:32722–32726. doi: 10.1074/jbc.271.51.32722. [DOI] [PubMed] [Google Scholar]
- 63.Kriem B, Sponne I, Fifre A, Malaplate-Armand C, Lozac’h-Pillot K, Koziel V, Yen-Potin FT, Bihain B, Oster T, Olivier JL, Pillot T. Cytosolic Phospholipase A2 mediates neuronal apoptosis induced by soluble oligomers of the amyloid-β peptide. FASEB J. 2005;19:85–87. doi: 10.1096/fj.04-1807fje. [DOI] [PubMed] [Google Scholar]
- 64.Lambert MP, Barlow AK, Chromy BA, Edwards C, Freed R, Liosatos M, Morgan TE, Rozovsky I, Trommer B, Viola KL, Wals P, Zhang C, Finch CE, Krafft GA, Klein WL. Diffusible, nonfibrillar ligands derived from Aβ1-42 are potent central nervous system neurotoxins. Proc Natl Acad Sci USA. 1998;95:6448–6453. doi: 10.1073/pnas.95.11.6448. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Launer L. Nonsteroidal anti-infiammatory drug use and the risk for Alzheimer’s disease: dissecting the epidemiological evidence. Drugs. 2003;63:731–739. doi: 10.2165/00003495-200363080-00001. [DOI] [PubMed] [Google Scholar]
- 66.Lee EO, Shin YJ, Chong YH. Mechanisms involved in prostaglandin E2-mediated neuroprotection against TNF-β: possible involvement of multiple signal transduction and β catenin/T-cell factor. J Neuroimmunol. 2004;155:21–31. doi: 10.1016/j.jneuroim.2004.05.012. [DOI] [PubMed] [Google Scholar]
- 67.Lee SF, Shah S, Yu C, Wigley WC, Li H, Lim M, Pedersen K, Han W, Thomas P, Lundkvist J, Hao YH, Yu G. A conserved GXXXG motif in APH-1 is critical for assembly and activity of the γ-secretase complex. J Biol Chem. 2004;279:4144–4152. doi: 10.1074/jbc.M309745200. [DOI] [PubMed] [Google Scholar]
- 68.Levy E, Carman MD, Fernandez-Madrid IJ, Power MD, Lieberburg I, van Duinen SG, Bots GT, Luyendijk W, Frangione B. Mutation of the Alzheimer’s disease amyloid gene in hereditary cerebral hemorrhage, Dutch type. Science. 1990;248:1124–1126. doi: 10.1126/science.2111584. [DOI] [PubMed] [Google Scholar]
- 69.Levy-Lahad E, Wasco W, Poorkaj P, Romano DM, Oshima J, Pettingell WH, Yu CE, Jondro PD, Schmidt SD, Wang K, Crowley AC, Fu YH, Guenette SY, Galas D, Nemens E, Wijisman EM, Bird TD, Schellenberg GD, Tanzi RE. Candidate gene for the chromosome 1 familial Alzheimer’s disease locus. Science. 1995;269:973–977. doi: 10.1126/science.7638622. [DOI] [PubMed] [Google Scholar]
- 70.Li Y, Sharov VG, Jiang N, Zaloga C, Sabbah HN, Chopp M. Ultrastructural and light microscopic evidence of apoptosis after middle cerebral artery occlusion in the rat. Am J Pathol. 1995;146:1045–1051. [PMC free article] [PubMed] [Google Scholar]
- 71.Li YM, Xu M, Lai MT, Huang Q, Castro JL, DiMuzio-Mower J, Harrison T, Lellis C, Nadin A, Neduvelil JG, Register RB, Sardana MK, Shearman MS, Smith AL, Shi XP, Yin KC, Shafer JA, Gardell SJ. Photoactivated gamma-secretase inhibitors directed to the active site covalently label presenilin 1. Nature. 2000;405:689–694. doi: 10.1038/35015085. [DOI] [PubMed] [Google Scholar]
- 72.Liang X, Wu L, Hand T, Andreasson K. Prostaglandin D2 mediates neuronal protection via the DP1 receptor. J Neurochem. 2005;92:477–986. doi: 10.1111/j.1471-4159.2004.02870.x. [DOI] [PubMed] [Google Scholar]
- 73.Lin LL, Wartmann M, Lin AY, Knopf JL, Seth A, Davis RJ. cPLA2 is phosphorylated and activated by MAP kinase. Cell. 1993;72:269–278. doi: 10.1016/0092-8674(93)90666-e. [DOI] [PubMed] [Google Scholar]
- 74.Lin TN, Wang Q, Simonyi A, Chen JJ, Cheung WM, He YY, Xu J, Sun AY, Hsu CY, Sun GY. Induction of secretory Phospholipase A2 in reactive astrocytes in response to transient focal cerebral ischemia in the rat brain. J Neurochem. 2004;90:637–645. doi: 10.1111/j.1471-4159.2004.02540.x. [DOI] [PubMed] [Google Scholar]
- 75.Massoud F, Devi G, Stern Y, Lawton A, Goldman JE, Liu Y, Chin SS, Mayeux R. A clinicopathological comparison of community-based and clinic-based cohorts of patients with dementia. Arch Neurol. 1999;56:1368–1373. doi: 10.1001/archneur.56.11.1368. [DOI] [PubMed] [Google Scholar]
- 76.Manolio TA, Burke GL, O’Leary DH, Evans G, Beauchamp N, Knepper L, Ward B. Relationships of cerebral MRI findings to ultrasonographic carotid atherosclerosis in older adults: the Cardiovascular Health Study. CHS Collaborative Research Group. Arterioscler Thromb Vasc Biol. 1999;19:356–365. doi: 10.1161/01.atv.19.2.356. [DOI] [PubMed] [Google Scholar]
- 77.McCullough L, Wu L, Haughey N, Liang X, Hand T, Wang Q, Breyer RM, Andreasson K. Neuroprotective function of the PGE2 EP2 receptor in cerebral ischemia. J Neurosci. 2004;24:257–268. doi: 10.1523/JNEUROSCI.4485-03.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.McDowell I. Alzheimer’s disease: insights from epidemiology. Aging. 2001;13(Milano):143–162. doi: 10.1007/BF03351474. [DOI] [PubMed] [Google Scholar]
- 79.McGeer PL. Cyclooxygenase-2 inhibitors relational and therapeutic potential for Alzheimer’s disease. Drugs Aging. 2000;17:1–11. doi: 10.2165/00002512-200017010-00001. [DOI] [PubMed] [Google Scholar]
- 80.Meldrum BS. Glutamate as a neurotransmitter in the brain: review of physiology and pathology. J Nutr. 2000;130:1007S–1015S. doi: 10.1093/jn/130.4.1007S. [DOI] [PubMed] [Google Scholar]
- 81.Mentis MJ, Salerno J, Horwitz B, Grady C, Schapiro MB, Murphy DG, Rapoport SI. Reduction of functional neuronal connectivity in long-term treated hypertension. Stroke. 1994;25:601–607. doi: 10.1161/01.str.25.3.601. [DOI] [PubMed] [Google Scholar]
- 82.Mitchell JA, Warner TD. Cyclooxygenase-2: pharmacology, physiology, biochemistry and relevance to NSAID therapy. Br J Pharmacol. 1999;128:1121–1132. doi: 10.1038/sj.bjp.0702897. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Miyao S, Takano A, Teramoto J, Takahashi A. Leukoaraiosis in relation to prognosis for patients with lacunar infarction. Stroke. 1992;23:1434–1438. doi: 10.1161/01.str.23.10.1434. [DOI] [PubMed] [Google Scholar]
- 84.Molloy GY, Rattray M, Williams RJ. Genes encoding multiple forms of Phospholipase A2 are expressed in rat brain. Neurosci Lett. 1998;258:139–142. doi: 10.1016/s0304-3940(98)00838-6. [DOI] [PubMed] [Google Scholar]
- 85.Montine TJ, Sidell KR, Crews BC, Markesbery WR, Marnett LJ, Roberts LJ, Morrow JD. Elevated CSF prostaglandin E2 levels in patients with probable AD. Neurology. 1999;53:1495–1498. doi: 10.1212/wnl.53.7.1495. [DOI] [PubMed] [Google Scholar]
- 86.Morris RG, Moser EI, Riedel G, Martin SJ, Sandin J, Day M, O’Carroll C. Elements of a neurobiological theory of the hippocampus: the role of activity-dependent synaptic plasticity in memory. Philos Transact: Biol Sci. 2003;358:773–786. doi: 10.1098/rstb.2002.1264. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Murakami M, Nakatani Y, Atsumi G, Inoue K, Kudo I. Regulatory functions of Phospholipase A2. Crit Rev Immunol. 1997;17:225–283. doi: 10.1615/critrevimmunol.v17.i3-4.10. [DOI] [PubMed] [Google Scholar]
- 88.Murakami M, Yoshihara K, Shimbara S, Lambeau G, Gelb MH, Singer AG, Sawada M, Inagaki N, Nagai H, Ishihara M, Ishikawa Y, Ishii T, Kudo I. Cellular arachidonatereleasing function and inflammation-associated expression of group IIF secretory Phospholipase A2. J Biol Chem. 2002;277:19145–19155. doi: 10.1074/jbc.M112385200. [DOI] [PubMed] [Google Scholar]
- 89.Murakami M, Kudo I. Phospholipase A2. J Biochem. 2002;131(Tokyo):285–292. doi: 10.1093/oxfordjournals.jbchem.a003101. [DOI] [PubMed] [Google Scholar]
- 90.Murakami M, Kambe T, Shimbara S, Kudo I. Functional coupling between various Phospholipase A2s and cyclooxygenases in immediate and delayed prostanoid biosynthetic pathways. J Biol Chem. 1999;274:3103–3115. doi: 10.1074/jbc.274.5.3103. [DOI] [PubMed] [Google Scholar]
- 91.Muthalif MM, Hefner Y, Canaan S, Harper J, Zhou H, Parmentier JH, Aebersold R, Gelb MH, Malik KU. Functional interaction of calcium-/calmodulin-dependent protein kinase II and cytosolic Phospholipase A2. J Biol Chem. 2001;276:39653–39660. doi: 10.1074/jbc.M103136200. [DOI] [PubMed] [Google Scholar]
- 92.Narumiya S. Prostanoid receptors and signal transduction. Prog Brain Res. 1996;113:231–241. doi: 10.1016/s0079-6123(08)61091-2. [DOI] [PubMed] [Google Scholar]
- 93.Narumiya S, FitzGerald GA. Genetic and pharmacological analysis of prostanoid receptor function. J Clin Invest. 2001;108:25–30. doi: 10.1172/JCI13455. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Needleman P, Turk J, Jakschik BA, Morrison AR, Lefkowith JB. Arachidonic acid metabolism. Ann Rev Biochem. 1986;55:69–102. doi: 10.1146/annurev.bi.55.070186.000441. [DOI] [PubMed] [Google Scholar]
- 95.Needleman P, Moncada S, Bunting S, Vane JR, Hamberg M, Samuelsson B. Identification of an enzyme in platelet microsomes which generates thromboxane A2 from prostaglandin endoperoxides. Nature. 1976;261:558–560. doi: 10.1038/261558a0. [DOI] [PubMed] [Google Scholar]
- 96.Negishi M, Sugimoto Y, Ichikawa A. Prostaglandin E receptors. J Lipid Med Cell Signal. 1995;12:379–391. doi: 10.1016/0929-7855(95)00024-k. [DOI] [PubMed] [Google Scholar]
- 97.Niwa K, Haensel C, Ross ME, Iadecola C. Cyclooxygenase-1 participants in selected vasodilator responses of the cerebral circulation. Circ Res. 2001;88:600–608. doi: 10.1161/01.res.88.6.600. [DOI] [PubMed] [Google Scholar]
- 98.Nogawa S, Zhang F, Ross ME, Iadecola C. Cyclooxygenase-2 gene expression in neurons contributes to ischemic brain damage. J Neurosci. 1997;17:2746–2755. doi: 10.1523/JNEUROSCI.17-08-02746.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Nunan J, Small DH. Proteolytic processing of the amyloid-beta protein precursor of Alzheimer’s disease. Essays Biochem. 2002;38:37–49. doi: 10.1042/bse0380037. [DOI] [PubMed] [Google Scholar]
- 100.Nunomura A, Perry G, Aliev G, Hirai K, Takeda A, Balraj EK, Jones PK, Ghanbari H, Wataya T, Shimohama S, Chiba S, Atwood CS, Petersen RB, Smith MA. Oxidative damage is the earliest event in Alzheimer disease. J Neuropathol Exp Neurol. 2001;60:759–767. doi: 10.1093/jnen/60.8.759. [DOI] [PubMed] [Google Scholar]
- 101.Ohno K, Sakai T, Fukushima M, Narumiya S, Fujiwara M. Site and mechanisms of growth inhibition by prostaglandins IV. Effect of cyclopentenone prostaglandins on cell progression of G1-enriched Hela S3 cells. J Pharmacol Exp Ther. 1988;245:294–299. [PubMed] [Google Scholar]
- 102.Oka S, Arita H. Inflammatory factors stimulate expression of group II Phospholipase A2 in rat cultured astrocytes. J Biol Chem. 1991;266:9956–9960. [PubMed] [Google Scholar]
- 103.Ott A, Breteler MM, van Harskamp F, Claus JJ, van der Cammen TJ, Grobbee DE, Hofman A. Prevalence of Alzheimer’s disease and vascular dementia: association with education. The Rotterdam study. Br Med J. 1995;310:970–973. doi: 10.1136/bmj.310.6985.970. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Ott A, Stolk RP, Hofman A, van Harskamp F, Grobbee DE, Breteler MM. Association of diabetes mellitus and dementia: the Rotterdam Study. Diabetologia. 1996;39:1392–1397. doi: 10.1007/s001250050588. [DOI] [PubMed] [Google Scholar]
- 105.Ott A, Slooter AJ, Hofman A, van Harskamp F, Witteman JC, Van Broeckhoven C, van Duijn CM, Breteler MM. Smoking and risk of dementia and Alzheimer’s disease in a population-based cohort study: the Rotterdam Study. Lancet. 1998;351:1840–1843. doi: 10.1016/s0140-6736(97)07541-7. [DOI] [PubMed] [Google Scholar]
- 106.Owada Y, Tominaga T, Yoshimoto T, Kondo H. Molecular cloning of rat cDNA for cytosolic Phospholipase A2 and the increased gene expression in the dentete gyrus following transient forebrain ischemia. Mol Brain Res. 1994;25:364–368. doi: 10.1016/0169-328x(94)90174-0. [DOI] [PubMed] [Google Scholar]
- 107.Paola D, Domenicotti C, Nitti M, Vitali A, Borghi R, Cottalasso D, Zaccheo D, Odetti P, Strocchi P, Marinari UM, Tabaton M, Pronzato MA. Oxidative stress induces increase in intracellular amyloid β-protein production and selective activation of βI and βII PKCs in NT2 cells. Biochem Biophys Res Commun. 2000;268:642–646. doi: 10.1006/bbrc.2000.2164. [DOI] [PubMed] [Google Scholar]
- 108.Pasinetti GM, Aisen PS. Cyclooxygenase-2 expression is increased in frontal cortex of Alzheimer’s disease brain. Neuroscience. 1998;87:319–324. doi: 10.1016/s0306-4522(98)00218-8. [DOI] [PubMed] [Google Scholar]
- 109.Pei JJ, Tanaka T, Tung YC, Braak E, Iqbal K, Grundke-Iqbal I. Distribution, levels, and activity of glycogen synthase kinase-3 in the Alzheimer disease brain. J Neuropathol Exp Neurol. 1997;56:70–78. doi: 10.1097/00005072-199701000-00007. [DOI] [PubMed] [Google Scholar]
- 110.Phillis JW, O’Regan MH. The role of phospholipases, cyclooxygenases, and lipoxygenases in cerebral ischemic/traumatic injuries. Crit Rev Neurobiol. 2003;15:61–90. doi: 10.1615/critrevneurobiol.v15.i1.30. [DOI] [PubMed] [Google Scholar]
- 111.Pratico D, Rokach J, Tangirala RK. Brains of aged apolipoprotein E-deficient mice have increased levels of F2-isoprostanes, in vivo markers of lipid peroxidation. J Neurochem. 1999;73:736–741. doi: 10.1046/j.1471-4159.1999.0730736.x. [DOI] [PubMed] [Google Scholar]
- 112.Pratico D, Lee VMY, Trojanowski JQ, Rokach J, FitzGerald GA. Increased F2-isoprostanes in Alzheimer’s disease: evidence of enhanced lipid peroxidation in vivo. FASEB J. 1998;12:1777–1783. doi: 10.1096/fasebj.12.15.1777. [DOI] [PubMed] [Google Scholar]
- 113.Pratico D, Delanty N. Oxidative injury in diseases of the central nervous system: focus on Alzheimer’s disease. Am J Med. 2000;109:577–585. doi: 10.1016/s0002-9343(00)00547-7. [DOI] [PubMed] [Google Scholar]
- 114.Rapoport M, Ferreira A. PD98059 prevents neurite degeneration induced by fibrillar beta-amyloid in mature hippocampal neurons. J Neurochem. 2000;74:125–133. doi: 10.1046/j.1471-4159.2000.0740125.x. [DOI] [PubMed] [Google Scholar]
- 115.Rogers J, Kirby LC, Hempelman SR, Berry DL, Mcgeer PL, Kaszniak AW, Zalinski J, Cofield M, Mansukhani L, Willson P, Kogan F. Clinical Trial of indomethacin in Alzheimer’s disease. Neurology. 1993;43:1609–1611. doi: 10.1212/wnl.43.8.1609. [DOI] [PubMed] [Google Scholar]
- 116.Rohn TT, Wong SM, Cotman CW, Cribbs DH. 15-deoxy-Δ12,14-prostaglandin J2, a specific ligand for peroxisome proliferator-activated receptor-g, induces neuronal apoptosis. Neuroreport. 2001;12:839–843. doi: 10.1097/00001756-200103260-00043. [DOI] [PubMed] [Google Scholar]
- 117.Sairanen T, Ristimäki A, Karjalainen-Lindsberg ML, Paetau A, Kaste M, Lindsberg P. Cyclooxygenase-2 is induced globally in infarcted human brain. Annu Neurol. 1998;43:738–747. doi: 10.1002/ana.410430608. [DOI] [PubMed] [Google Scholar]
- 118.Salerno JA, Murphy DG, Horwitz B, DeCarli C, Haxby JV, Rapoport SI, Schapiro MB. Brain atrophy in hypertension. A volumetric magnetic resonance imaging study. Hypertension. 1992;20:340–348. doi: 10.1161/01.hyp.20.3.340. [DOI] [PubMed] [Google Scholar]
- 119.Sanchez-Moreno C, Dashe JF, Scott T, Thaler D, Folstein MF, Martin A. Decreased levels of plasma vitamin C and increased concentrations of inflammatory and oxidative stress markers after stroke. Stroke. 2004;35:163–168. doi: 10.1161/01.STR.0000105391.62306.2E. [DOI] [PubMed] [Google Scholar]
- 120.Satoh T, Ishikawa Y, Kataoka Y, Cui Y, Yanase H, Kato K, Watanabe Y, Nakadate K, Matsumura K, Hatanaka H, Kataoka K, Noyori R, Suzuki M. CNS-specific prostacyclin ligands as neuronal survival-promoting factors in the brain. Eur J Neurosci. 1999;113:3115–3124. doi: 10.1046/j.1460-9568.1999.00791.x. [DOI] [PubMed] [Google Scholar]
- 121.Satoh K, Nagano Y, Shimomura C, Suzuki N, Saeki Y, Yokota H. Expression of prostaglandin E synthase mRNA is induced in β-amyloid treated rat astrocytes. Neurosci Lett. 2000;283:221–223. doi: 10.1016/s0304-3940(00)00926-5. [DOI] [PubMed] [Google Scholar]
- 122.Schehr RS. New treatments for acute stroke. Nat Biotechnol. 1996;14:1549–1554. doi: 10.1038/nbt1196-1549. [DOI] [PubMed] [Google Scholar]
- 123.Schievella AR, Regier MK, Smith WL, Lin WL. Calcium-mediated translocation of cytosolic Phospholipase A2 to the nuclear envelope and endoplasmic reticulum. J Biol Chem. 1995;270:30749–30754. doi: 10.1074/jbc.270.51.30749. [DOI] [PubMed] [Google Scholar]
- 124.Schellenberg GD, Bird TD, Wijsman EM, Orr HT, Anderson L, Nemens E, White JA, Bonnycastle L, Weber JL, Alonso ME, Potter H, Heston LL, Martin GM. Genetic linkage evidence for a familial Alzheimer’s disease locus on chromosome 14. Science. 1992;258:668–71. doi: 10.1126/science.1411576. [DOI] [PubMed] [Google Scholar]
- 125.Selkoe DJ. The molecular pathology of Alzheimer’s disease. Neuron. 1991;6:487–498. doi: 10.1016/0896-6273(91)90052-2. [DOI] [PubMed] [Google Scholar]
- 126.Selkoe DJ. Alzheimer’s disease: a central role for amyloid. J Neuropathol Exp Neurol. 1994;53:438–447. doi: 10.1097/00005072-199409000-00003. [DOI] [PubMed] [Google Scholar]
- 127.Shie FS, Montine KS, Breyer RM, Montine TJ. Microglial EP2 as a new target to increase amyloid β phagocytosis and decrease amyloid β -induced damage to neurons. Brain Pathol. 2005;15:134–138. doi: 10.1111/j.1750-3639.2005.tb00509.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Shimizu T, Yamamoto A, Hayaishi O. Specific binding of prostaglandin D2 to rat brain synaptic membrane. J Biol Chem. 1982;257:13570–13575. [PubMed] [Google Scholar]
- 129.Six DA, Dennis EA. The expanding superfamily of Phospholipase A2 enzymes: classification and characterization. Biochim Biophys Acta. 2000;1488:1–19. doi: 10.1016/s1388-1981(00)00105-0. [DOI] [PubMed] [Google Scholar]
- 130.Sonkusare SK, Kaul CL, Ramarao P. Dementia of Alzheimer’s disease and other neurodegenerative disordersmemantine, a new hope. Pharmacol Res. 2005;51:1–17. doi: 10.1016/j.phrs.2004.05.005. [DOI] [PubMed] [Google Scholar]
- 131.Streifler JY, Eliasziw M, Benavente OR, Alamowitch S, Fox AJ, Hachinski VC, Barnett HJ. North American Symptomatic Carotid Endarterectomy Trial Group.Prognostic importance of leukoaraiosis in patients with symptomatic internal carotid artery stenosis. Stroke. 2002;33:1651–1655. doi: 10.1161/01.str.0000018010.38749.08. [DOI] [PubMed] [Google Scholar]
- 132.Stephenson DT, Lemere CA, Selkoe DJ, Clemens JA. Cytosolic Phospholipase A2 (cPLA2) immunoreactivity is elevated in Alzheimer’s disease brain. NeuroBiol Dis. 1996;3:51–63. doi: 10.1006/nbdi.1996.0005. [DOI] [PubMed] [Google Scholar]
- 133.Sundararajan S, Gamboa JL, Victor NA, Wanderi EW, Lust WD, Landreth GE. Peroxisome proliferator-activated receptor-γ ligands reduce inflammation and infarction size in transient focal ischemia. Neuroscience. 2005;130:685–696. doi: 10.1016/j.neuroscience.2004.10.021. [DOI] [PubMed] [Google Scholar]
- 134.Takahashi RH, Almeida CG, Kearney PF, Yu F, Lin MT, Milner TA, Gouras GK. Oligomerization of Alzheimer’s β-amyloid within processes and synapses of cultured neurons and brain. J Neurosci. 2004;24:3592–3599. doi: 10.1523/JNEUROSCI.5167-03.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Takechi H, Matsumura K, Watanabe Y, Kato K, Noyori R, Suzuki M. A novel subtype of the prostacyclin receptor expressed in the central nervous system. J Biol Chem. 1996;271:5901–5906. doi: 10.1074/jbc.271.10.5901. [DOI] [PubMed] [Google Scholar]
- 136.Takashima A, Noguchi K, Sato K, Hoshino T, Imahori K. Tau protein kinase I is essential for amyloid β-proteininduced neurotoxicity. Proc Natl Acad Sci USA. 1993;90:7789–7793. doi: 10.1073/pnas.90.16.7789. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Talbot K, Young RA, Jolly-Tornetta C, Lee VM, Trojanowski JQ, Wolf BA. A frontal variant of Alzheimer’s disease exhibits decreased calcium-independent Phospholipase A2 activity in the prefrontal cortex. Neurochem Int. 2000;37:17–31. doi: 10.1016/s0197-0186(00)00006-1. [DOI] [PubMed] [Google Scholar]
- 138.Tomimoto H, Shibata M, Ihara M, Akiguchi I, Ohtani R, Bucka H. A comparative study on the expression of cyclooxygenase and 5-lipoxygenaseduring cerebral ischemia in humans. Acta Neuropathol. 2002;104(Berl):601–607. doi: 10.1007/s00401-002-0590-0. [DOI] [PubMed] [Google Scholar]
- 139.Trommer BL, Shah C, Yun SH, Gamkrelidze G, Pasternak ES, Blaine , Stine W, Manelli A, Sullivan P, Pasternak JF, LaDu MJ. ApoE isoform-specific effects on LTP: blockade by oligomeric amyloid-β1-42. NeuroBiol Dis. 2005;18:75–82. doi: 10.1016/j.nbd.2004.08.011. [DOI] [PubMed] [Google Scholar]
- 140.Ueda K, Fukui Y, Kageyama H. Amyloid β protein-induced neuronal cell death: neurotoxic properties of aggregated amyloid β protein. Brain Res. 1994;639:240–244. doi: 10.1016/0006-8993(94)91736-1. [DOI] [PubMed] [Google Scholar]
- 141.Ueda K, Yagami T, Kageyama H, Kawasaki K. Protein kinase inhibitor attenuates apoptotic cell death induced by amyloid β protein in culture of the rat cerebral cortex. Neurosci Lett. 1996;203:175–178. doi: 10.1016/0304-3940(95)12288-5. [DOI] [PubMed] [Google Scholar]
- 142.Ueda K, Shinohara S, Yagami T, Asakura K, Kawasaki K. Amyloid β protein potentiates Ca2+ influx through L-type voltage-sensitive Ca2+ channels: A possible involvement of free radicals. J Neurochem. 1997;68:265–271. doi: 10.1046/j.1471-4159.1997.68010265.x. [DOI] [PubMed] [Google Scholar]
- 143.Urade Y, Eguchi N. Lipocalin-type and hematopoietic prostaglandin D synthases as a novel example of functional convergence. Prostaglandins Other Lipid Mediat. 2002;68-69:375–382. doi: 10.1016/s0090-6980(02)00042-4. [DOI] [PubMed] [Google Scholar]
- 144.Umemura K, Kawai H, Ishihara H, Nakashima M. Inhibitory effect of clopidogrel, vapiprost and argatroban on the middle cerebral artery thrombosis in the rat. Jpn J Pharmacol. 1995;67:253–258. doi: 10.1254/jjp.67.253. [DOI] [PubMed] [Google Scholar]
- 145.Vadas P, Pruzanski W. Role of secretory phospholipases A2 in the pathobiology of disease. Lab Invest. 1986;55:391–404. [PubMed] [Google Scholar]
- 146.Valentin E, Ghomashchi F, Gelb MH, Lazdunski M, Lambeau G. On the diversity of secreted phospholipases A2. Cloning, tissue distribution, and functional expression of two novel mouse group II enzymes. J Biol Chem. 1999;274:31195–31202. doi: 10.1074/jbc.274.44.31195. [DOI] [PubMed] [Google Scholar]
- 147.van Kooten F, Ciabattoni G, Koudstaal PJ, Grobbee DE, Kluft C, Patrono C. Increased thromboxane biosynthesis is associated with poststroke dementia. Stroke. 1999;30:1542–1547. doi: 10.1161/01.str.30.8.1542. [DOI] [PubMed] [Google Scholar]
- 148.Walshm DM, Klyubin I, Fadeeva JV, Cullen WK, Anwyl R, Wolfe MS, Rowan MJ, Selkoe DJ. Naturally secreted oligomers of amyloid beta protein potently inhibit hippocampal long-term potentiation in vivo. Nature. 2002;416:535–539. doi: 10.1038/416535a. [DOI] [PubMed] [Google Scholar]
- 149.Watanabe K. Prostaglandin F synthase. Prostaglandins Other Lipid Mediat. 2002;68-69:401–407. doi: 10.1016/s0090-6980(02)00044-8. [DOI] [PubMed] [Google Scholar]
- 150.Watanabe Y, Tokumoto H, Yamashita A, Narumiya S, Mizuno N, Hayaishi O. Specific binding of prostaglandin D2, E2, F2α in postmortem human brain. Brain Res. 1985;342:110–116. doi: 10.1016/0006-8993(85)91358-7. [DOI] [PubMed] [Google Scholar]
- 151.Winchester SK, Imamura T, Gross GA, Muglia LM, Vogt SK, Wright J, Watanabe K, Tai HH, Muglia LJ. Coordinate regulation of prostaglandin metabolism for induction of parturition in mice. Endocrinology. 2002;143:2593–2598. doi: 10.1210/endo.143.7.8926. [DOI] [PubMed] [Google Scholar]
- 152.Wisniewski T, Frangione B. Molecular biology of brain aging and neurodegenerative disorders. Acta Neurobiol Exp. 1996;56(Wars):267–279. doi: 10.55782/ane-1996-1132. [DOI] [PubMed] [Google Scholar]
- 153.Wong PT, McGeer PL, McGeer EG. Decreased prostaglandin synthesis in postmortem cerebral cortex from patients with Alzheimer’s disease. Neurochem Int. 1992;21:197–202. doi: 10.1016/0197-0186(92)90147-j. [DOI] [PubMed] [Google Scholar]
- 154.Wright DH, Nantel F, Metters KM, Ford-Hutchinson AW. A novel biological role for prostaglandin D2 is suggested by distribution studies of the rat DP prostanoid receptor. Eur J Pharmacol. 1999;377:101–115. doi: 10.1016/s0014-2999(99)00358-1. [DOI] [PubMed] [Google Scholar]
- 155.Xiang Z, Ho L, Valdellon J, Borchelt D, Kelley K, Spielman L, Aisen PS, Pasinetti GM. Cyclooxygenase (COX)-2 and cell cycle activity in a transgenic mouse model of Alzheimer’s disease neuropathology. Neurobiol Aging. 2002;23:327–334. doi: 10.1016/s0197-4580(01)00282-2. [DOI] [PubMed] [Google Scholar]
- 156.Xu S, Venge P. Lipocalins as biochemical markers of disease. Biochim Biophys Acta. 2000;1482:298–307. doi: 10.1016/s0167-4838(00)00163-1. [DOI] [PubMed] [Google Scholar]
- 157.Yagami T, Ueda K, Asakura K, Sakaeda T, Kuroda T, Hata S, Kambayashi Y, Fujimoto M. Effects of S-2474, a novel nonsteroidal anti-inflammatory drug, on amyloid β protein-induced neuronal cell death. Br J Pharmacol. 2001;134:673–681. doi: 10.1038/sj.bjp.0704261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Yagami T, Ueda K, Asakura K, Kuroda T, Hata S, Sakaeda T, Kambayashi Y, Fujimoto M. Effects of endothelin B receptor agonists on amyloid β protein-induced neuronal cell death. Brain Res. 2002;948:72–81. doi: 10.1016/s0006-8993(02)02951-7. [DOI] [PubMed] [Google Scholar]
- 159.Yagami T, Ueda K, Asakura K, Sakaeda T, Nakazato H, Kuroda T, Hata S, Sakaguchi G, Itoh N, Kambayashi Y, Tsuzuki H. Gas6 rescues cortical neurons from amyloid β protein-induced apoptosis. Neuropharmacology. 2002;43:1289–1296. doi: 10.1016/s0028-3908(02)00333-7. [DOI] [PubMed] [Google Scholar]
- 160.Yagami T, Nakazato H, Ueda K, Asakura K, Kuroda T, Hata S, Sakaeda T, Sakaguchi G, Itoh N, Hashimoto Y, Hiroshige T, Kambayashi Y. Prostaglandin E2 rescues cortical neurons from amyloid beta protein-induced apoptosis. Brain Res. 2003;959:328–335. doi: 10.1016/s0006-8993(02)03773-3. [DOI] [PubMed] [Google Scholar]
- 161.Yagami T, Ueda K, Sakaeda T, Itoh N, Sakaguchi G, Noboru O, Hori Y, Fujimoto M. Protective effects of a selective L-type voltage-sensitive calcium channel blocker, S-312d, on neuronal cell death. Biochem Pharmacol. 2004;67:1153–1165. doi: 10.1016/j.bcp.2003.11.005. [DOI] [PubMed] [Google Scholar]
- 162.Yagami T, Ueda K, Asakura K, Hata S, Kuroda T, Sakaeda T, Takasu N, Tanaka K, Gemba T, Hori Y. Human group IIA secretory Phospholipase A2 induces neuronal cell death via apoptosis. Mol Pharmacol. 2002;61:114–126. doi: 10.1124/mol.61.1.114. [DOI] [PubMed] [Google Scholar]
- 163.Yagami T, Ueda K, Asakura K, Nakazato H, Hata S, Kuroda T, Sakaeda T, Sakaguchi G, Itoh N, Hashimoto Y, Hori Y. Human group IIA secretory Phospholipase A2 potentiates Ca2+ influx through L-type voltage-sensitive Ca2+ channel. J Neurochem. 2003;85:749–758. doi: 10.1046/j.1471-4159.2003.01712.x. [DOI] [PubMed] [Google Scholar]
- 164.Yagami T, Ueda K, Asakura K, Hata S, Kuroda T, Kishino J, Sakaeda T, Sakaguchi G, Itoh H, Hori Y. Group IB secretory Phospholipase A2 induces cell death in cultured cortical neurons: A possible involvement of its binding sites. Brain Res. 2002;949:197–201. doi: 10.1016/s0006-8993(02)03144-x. [DOI] [PubMed] [Google Scholar]
- 165.Yagami T, Ueda K, Asakura K, Sakaeda S, Hata S, Kuroda T, Sakaguchi G, Itoh N, Hashimoto Y, Hori Y. Porcine pancreatic group IB secretory Phospholipase A2 potentiates Ca2+ influx through L-type voltage-sensitive Ca2+ channel. Brain Res. 2003;960:71–80. doi: 10.1016/s0006-8993(02)03775-7. [DOI] [PubMed] [Google Scholar]
- 166.Yagami T, Ueda K, Asakura K, Hayasaki-Kajiwara Y, Nakazato H, Sakaeda T, Hata S, Kuroda T, Takasu N, Hori Y. Group IB secretory Phospholipase A2 induces neuronal cell death via apoptosis. J Neurochem. 2002;81:449–461. doi: 10.1046/j.1471-4159.2002.00800.x. [DOI] [PubMed] [Google Scholar]
- 167.Yagami T, Takahara Y, Ishibashi C, Sakaguchi G, Itoh N, Ueda K, Nakazato H, Okamura N, Hiramatsu Y, Honma T, Arimura A, Sakaeda T, Katsuura G. Amyloid β protein impairs motor function via thromboxane A2 in the rat striatum. NeuroBiol Dis. 2004;16:481–489. doi: 10.1016/j.nbd.2004.04.013. [DOI] [PubMed] [Google Scholar]
- 168.Yagami T, Ueda K, Asakura K, Takasu N, Sakaeda T, Itoh N, Sakaguchi G, Kishino J, Nakazato H, Katsuyama Y, Nagasaki T, Okamura N, Hori Y, Hanasaki K, Arimura A, Fujimoto M. Novel binding sites of 15-deoxy-Δ 12,14prostaglandin J2, in plasma membranes from primary rat cortical neurons. Exp Cell Res. 2003;291:212–227. doi: 10.1016/s0014-4827(03)00369-0. [DOI] [PubMed] [Google Scholar]
- 169.Yang HC, Mosior M, Ni B, Dennis EA. Regional distribution, ontogeny, purification, and characterization of the Ca2+-independent Phospholipase A2 from rat brain. J Neurochem. 1999;73:1278–1287. doi: 10.1046/j.1471-4159.1999.0731278.x. [DOI] [PubMed] [Google Scholar]
- 170.Yamagata K, Andreasson KI, Kaufmann WE, Barnes CA, Worley PF. Expression of a mitogen-inducible cyclooxygenase in brain neurons: Regulation by synaptic activity and glucocorticoids. Neuron. 1993;11:371–386. doi: 10.1016/0896-6273(93)90192-t. [DOI] [PubMed] [Google Scholar]
- 171.Yokota C, Kaji T, Kuge Y, Inoue H, Tamaki N, Minematsu K. Temporal and topographic profiles of cyclooxygenase-2 expression during 24 h of focal brain ishemia in rats. Neurosci Lett. 2004;357:219–222. doi: 10.1016/j.neulet.2003.12.109. [DOI] [PubMed] [Google Scholar]
- 172.Younkin SG. Evidence that Aβ42 is the real culprit in Alzheimer’s disease. Ann Neurol. 1995;37:287–288. doi: 10.1002/ana.410370303. [DOI] [PubMed] [Google Scholar]