Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2015 Apr 15.
Published in final edited form as: Biochem Pharmacol. 2014 Jan 13;88(4):565–572. doi: 10.1016/j.bcp.2013.12.014

Therapeutic Implications of the Prostaglandin Pathway in Alzheimer’s Disease

Eiron Cudaback 1, Nikolas L Jorstad 1, Yue Yang 1, Thomas J Montine 1, C Dirk Keene 1,*
PMCID: PMC3972296  NIHMSID: NIHMS555966  PMID: 24434190

Abstract

An important pathologic hallmark of Alzheimer’s disease (AD) is neuroinflammation, a process characterized in AD by disproportionate activation of cells (microglia and astrocytes, primarily) of the non-specific innate immune system within the CNS. While inflammation itself is not intrinsically detrimental, a delicate balance of pro- and anti-inflammatory signals must be maintained to ensure that long-term exaggerated responses do not damage the brain over time. Non-steroidal anti-inflammatory drugs (NSAIDs) represent a broad class of powerful therapeutics that temper inflammation by inhibiting cyclooxygenase-mediated signaling pathways including prostaglandins, which are the principal mediators of CNS neuroinflammation. While historically used to treat discrete or systemic inflammatory conditions, epidemiologic evidence suggests that protracted NSAID use may delay AD onset, as well as decrease disease severity and rate of progression. Unfortunately, clinical trials with NSAIDs have thus far yielded disappointing results, including premature discontinuation of a large-scale prevention trial due to unexpected cardiovascular side effects. Here we review the literature and make the argument that more targeted exploitation of downstream prostaglandin signaling pathways may offer significant therapeutic benefits for AD while minimizing adverse side effects. Directed strategies such as these may ultimately help to delay the deleterious consequences of brain aging and might someday lead to new therapies for AD and other chronic neurodegenerative diseases.

Keywords: Alzheimer’s disease, prostaglandin, neuroinflammation, cyclooxygenase, NSAID

1. Neuroinflammation in AD

Alzheimer’s disease (AD) is an incurable neurodegenerative disorder affecting tens of millions of Americans and their families. Sporadic, or late onset, AD (LOAD) is slowly progressive, with age representing the single greatest risk factor. While the neuropathologic hallmarks of the disease include both intra- and extracellular aggregation of neurotoxic peptides within discrete brain regions, widespread neuroinflammation prominently accompanies these lesions [14]. Indeed, inflammatory molecules, including prostaglandins and cytokines, increase in cerebral spinal fluid (CSF) and brain parenchyma with age [58] and are associated with age-dependent cognitive impairment [911], suggesting that exacerbated or persistent neuroinflammation is a potentially significant driver of pathogenesis and disease progression in age-related neurodegenerative diseases [12], and especially in AD [13]. Furthermore, this inflammatory milieu is present in both the initial asymptomatic latent disease phase [14] in addition to more advanced stages of AD, supporting the development of targeted anti-inflammatory pharmacotherapy approaches aimed at prevention as well as treatment [15].

2. NSAIDs and AD: Epidemiology

Anti-inflammatory drugs are widely used for a variety of conditions, from simple headaches and fevers to serious autoimmune disorders such as rheumatoid arthritis (RA). Noting that various molecular signatures of inflammation are markedly increased in the brains of AD subjects [1, 16, 17], more than twenty years ago McGeer, Rogers, and colleagues observed that coincidence of AD and RA was significantly below the levels expected in the general population, implicating anti-inflammatory drugs as potentially neuroprotective. Specifically, the authors calculated general AD prevalence to be 2.7% in patients older than 64 years, and according to hospital discharge statistics, found that a diagnosis of AD and RA in the same patient existed in 29 of 13,246 individuals (0.2%) [18]. Furthermore, only 2 of 169 autopsied putative AD cases were documented to have preexisting RA, and the coincidence of both disorders was less than 0.5% (6 of 1332) in RA and AD clinics [18]. They inferred from this association that chronic anti-inflammatory treatment may provide a significant preventive or therapeutic intervention for AD.

3. NSAIDs and AD: Clinical trials

Although epidemiological findings were ultimately met with skepticism because of the unusually low AD prevalence cited compared to other reported observations [19], Rogers et al conducted a short-term double-blind placebo-controlled clinical trial using the non-steroidal anti-inflammatory drug (NSAID) indomethacin [20]. They reported reduced cognitive impairment in patients prescribed 100 to 150 mg/d of indomethacin after six months of treatment compared to controls [20]. Unfortunately, the study was hampered by poor tolerance of drug side effects and significant participant dropout, and did not address the effects of NSAIDs during the prodromal asymptomatic phase of AD since inclusion criteria for study subjects included previous AD diagnosis. Several clinical trials followed mostly in patients with clinical AD with varying limited success [2125]. Even so, administration of non-selective [2631] and selective [26, 32] COX inhibitors in various murine AD models results in significant reduction of amyloid pathology and associated neuroinflammation, as well as improved performance in cognitive testing. Combined with existing epidemiological evidence [1, 1618] supporting this protective role for NSAIDs in the development of AD, the ADAPT study tested the preventive efficacy of selective and non-selective COX inhibitors in AD[33, 34], but it was abruptly discontinued due to significant cardiovascular side effects [35, 36]. Noteworthy, a subset of participants in that trial who did not develop AD early in the trial were shown to have a lower incidence of AD with long-term follow up, suggesting a subgroup in the study in whom NSAID administration may have provided a preventive benefit [36, 37]. However, for others in the trial, NSAID administration was poorly tolerated and in others may have unmasked latent AD [35, 36]. The study authors acknowledge that the therapeutic effect of NSAIDs likely varies as a function of AD stage and progression [33, 34], confirming that increased efficacy might be achieved through prophylactic NSAID administration years in advance of clinical onset. Thus, while NSAIDs demonstrated neuroprotection in numerous experimental studies, there appeared to be limited clinical benefit for patients with AD (Table 1). Zahs and Ashe offer a possible explanation for this unfortunate contradiction, proposing that current animal models of AD more accurately reflect early latent phases of disease, failing to accurately capture the more clinically observed time course[38]. If true, this suggests that NSAID clinical trials should focus on prevention over intervention, a perspective supported by the original observation in RA subjects where NSAID regimens were likely initiated during latent disease phase in predisposed individuals. An additional consideration is the deleterious side effects associated with chronic NSAID use, especially selective COX-2 inhibitors like celecoxib and rofecoxib, in elderly patients [35, 39]. Thus, a more effective, less toxic alternative is needed.

Table 1.

Results from randomized, double-blinded, placebo-controlled clinical trials using currently prescribed NSAIDs.

Drugs No. of subjects Primary outcome measure Results Pre-treatment state References
Celecoxib 425 aADAS-cog and bCIBIC+ Did not slow the progression Mild-to-moderate AD [126]
Celecoxib or Naproxen 2528 Incidence of AD Reduction in AD incidence among asymptomatic enrollees who were given naproxen. High incidence of adverse side effects for both drugs Symptomatic AD [37]
Ibuprofren 132 aADAS-cog Did not slow the progression Mild-to-moderate AD [127]
Prednisone 138 aADAS-cog Did not slow the progression Probable AD [128]
Rofecoxib 692 aADAS-cog and bCIBIC+ Did not slow the progression Mild-to-moderate AD [129]
Rofecoxib 1457 Percentage of patients with a clinical diagnosis of AD Did not slow the progression MCI [130]
Rofecoxib or Naproxen 351 aADAS-cog Did not slow the progression Mild-to-moderate AD [131]
Triflusal 257 aADAS-cog Triflusal therapy was associated with a significant lower rate of conversion to dementia MCI [132]

ADAS-cog (Alzheimer Disease Assessment Scale- Cognitive); CIBIC+ (Clinician’s Interview Based Impression of Change with caregiver input); AD (Alzheimer’s Disease); MCI (mild cognitive impairment).

4. Cyclooxygenase and AD

NSAIDs inhibit cyclooxygenase (COX) enzymes, the rate-limiting mediators of prostanoid biosynthesis. Prostanoids are a broad class of arachidonic acid-derived paracrine signaling molecules that include thromboxanes, prostaglandins, and prostacyclins. Under normal conditions, a constitutive pool of prostanoids ubiquitously regulates a host of diverse physiological processes, including vasomotor tone, platelet aggregation, ovulation, and neonatal development [4043]. However, a second highly responsive and inducible prostanoid pool of primarily prostaglandin E2 (PGE2) mediates the initiation and propagation of inflammation, and is thought to contribute to the disproportionate inflammatory response in AD, and to many other pathological processes such as carcinogenesis and metastasis [44, 45]. As endogenous lipid signaling molecules, the prostanoids are not proactively synthesized and stored within cells for future release, but are rather generated on demand in response to a variety of inflammatory triggers. One of these is amyloid β (Aβ), a pleotropic neurotoxin and neuropathologic hallmark of AD [46]. In addition to direct neurotoxicity, Aβ mediates increased cellular phospholipase activity [47, 48], the first step in prostanoid biosynthesis, in addition to stimulation of pro-inflammatory cytokine secretion.

Phospholipase A2 (PLA2) family members liberate fatty acid substrates, especially arachidonic acid, via hydrolysis of cellular membrane phospholipids, and thus initiate prostanoid signaling cascades (Figure 1). Interestingly, PLA2-independent sources of arachidonic acid also appear to contribute to the pool of readily available precursors for prostanoid biosynthesis [49, 50], especially in brain [51], suggesting a more complicated upstream signaling scheme in vivo. Free arachidonic acid is then converted to PGH2 by COX enzymes in a two-step reaction that transiently forms the labile prostanoid intermediate PGG2 via oxygenation, followed by the peroxidation of PGG2 to yield more stable PGH2 (Figure 1). In addition to COX enzymes, lipoxygenases and cytochrome P450s readily metabolize arachidonic acid, yielding leukotrienes and epoxyeicosatrienoic acids, respectively [52]. Two important COX isoforms exist, COX-1 and COX-2, each having distinct but overlapping tissue distributions and activity profiles. COX-1 is expressed in most tissues and cell types and is constitutively active, and as such has been historically considered a crucial player in the maintenance of general cell physiology and homeostasis. In contrast, COX-2 expression shows greater restriction and is uniquely inducible, collectively limiting its activity and delineating its role as a respondent to stress and insult. A variety of stimuli, including cell proliferation and differentiation, neoplasia, environmental stress, and inflammation, can induce COX-2 expression and increase prostanoid levels. COX-2 expression in all tissues is tightly controlled and is important in housekeeping roles, including regulation of systemic energy homeostasis [53], postnatal renal development [54], female fertility[55], and maintenance of cardiovascular homeostasis[56], as evidenced by the cardiovascular risks associated with chronic use of COX-2 selective inhibitors such as rofecoxib and celecoxib [39, 57]. COX-2 expression in brain is normally very low but is markedly increased in AD [58, 59]. COX-3, a novel splice variant of COX-1, has been identified and shows significant enrichment in human cerebral cortex and heart [60]. While a link may exist between COX-3 and AD [61, 62], additional studies are needed to understand this relationship, especially given the fact that multiple COX-1 splice variants have been described and controversy remains as to the expression of COX-3 in humans [63].

Figure 1.

Figure 1

Generalized prostanoid signaling scheme. PLA2-dependent hydrolysis of membrane phospholipids liberates AA, providing substrate for COX-1 and COX-2. Stepwise oxygenation and peroxidation reactions by COX enzymes yield PGH2, which is then selectively converted to the various prostanoids by specific synthases. PGT then transports newly synthesized prostanoids to the extracellular compartment where they are free to act as autocrine or paracrine activators of cell-surface G protein-coupled prostanoid receptor subtypes. Preferred Gα subunit coupling is also denoted for each prostanoid receptor subtype. PLA2 (phospholipase A2); AA (arachidonic acid); COX (cyclooxygenase); PGG2 (prostaglandin G2); PGH2 (prostaglandin H2); PGE2 (prostaglandin E2); PGD2 (prostaglandin D2); PGF (prostaglandin F); PGI2 (prostaglandin I2); TXA2 (thromboxane A2); mPGES (microsomal PGE synthase); cPGES (cytosolic PGE synthase); PGDS (PGD synthase); PGFS (PGF synthase); PGIS (PGI synthase); TXS (TXA synthase); PGT (prostaglandin transporter); EP1 – 4 (PGE receptor 1 – 4); DP (PGD receptor); FP (PGF receptor); IP (PGI receptor); TP (TXA receptor).

COX activity has been repeatedly implicated in neuroinflammation associated with normal aging [6466] and in the pathophysiology of experimental AD [29, 30, 6773]. Isoform-specific COX expression changes are well documented in patients with AD[74, 75]. Thus, the epidemiological evidence supporting a therapeutic benefit of chronic NSAID administration in AD is presumed to derive from COX inhibition and modulation of neuroinflammation, although the effects may be more direct. For instance, many NSAIDs and structurally related compounds directly target gamma secretase [7678], a membrane-associated multimeric protein complex that includes the aspartyl protease presenilin. Mutations in presenilin 1 and 2 are a well-known cause of familial AD. The gamma secretase complex differentially cleaves the transmembrane protein substrate amyloid precursor protein (APP), subsequently releasing Aβ. Select NSAIDs, in addition to several structural enantiomers, inhibit gamma secretase activity, and therefore reduce formation and accumulation of Aβ in animal models [76, 79, 80] and human trials [81, 82]. Nevertheless, NSAIDs appear to reduce risk of AD-related dementia independent of direct or indirect modulation of Aβ levels based on studies by Szekely [83], and identification of downstream effectors that specifically mediate this function has led to increased study of prostaglandin pathways as therapeutic targets for AD.

5. Prostaglandins

Transgenic animal models continue to support the therapeutic efficacy of NSAIDs for AD [2632], and, while clinical trials have been variable, if not disappointing, some are working to identify patient subpopulations who might benefit from NSAID therapy, such as those in the ADAPT trial. However, in order to treat a broader AD population, a more targeted and efficacious pharmacotherapy is needed with a limited side effect profile. COX activation results in a cascade of prostanoid signaling with an abundance of potential downstream therapeutic targets, and COX inhibitors result in blanket suppression of prostanoid pathways. Teasing apart the COX-dependent neuroprotective pathways from the ones involved in cardiovascular toxicity or even neurotoxicity is an active area of research, since epidemiological and experimental studies continue to support the presence of a molecular target of NSAIDS in the development or progression of AD. COX activation through specific synthases results in the production of a number of biologically relevant prostaglandins (Figure 1): Prostaglandin I2 (PGI2), prostaglandin F2 alpha (PGF2a), prostaglandin D2 (PGD2), thromboxane (TXA2), and prostaglandin E2 (PGE2). Each of these molecules has distinctive functional pathways and physiological consequences, such as immuno- or vaso-modulation, that can be beneficial or deleterious depending on the tissue substrate and clinical context.

6. Prostaglandin Pathways and AD

NSAIDs represent more of a pharmacological sledgehammer than a silver bullet in the therapeutic battle against AD. Yet their repeated efficacy in animal models and remarkable tale of epidemiology remain undeniable. While the answer may lie in appropriate temporal administration, it remains noteworthy that the site of NSAID anti-inflammatory action exists far upstream of a diverse prostaglandin signaling scheme. The question of therapeutic value may be better answered through directed targeting of the diverse downstream players, taking advantage of the high degree of receptor selectivity and responsiveness exhibited by this system.

In response to inflammatory stimuli, the prostaglandins are synthesized and then freely diffuse from cells into the extracellular compartment[84]. Clearance and recycling of extracellular prostaglandins is achieved by a family of organic anion-transporting polypeptide transporters[85, 86]. Prostaglandins act on cognate G protein-coupled receptors (GPCRs) to induce an inflammatory response (Figure 1), and numerous studies are under way targeting individual GPCRs in experimental AD as an effort to tease apart the neurotoxic and neuroprotective portions of the prostaglandin pathway. The GPCRs utilized by prostaglandin receptors subserve diverse cellular functions and their modulation may be associated with diverse cytotoxicities, so a more directed approach utilizing specific receptors is needed.

Of the five prostaglandins, PGE2 is the major effector in the CNS based on synthase expression data and direct measurements [87], and is the most studied with regard to neuroinflammation. PGE2 is synthesized in either the membrane by microsomal prostaglandin E synthase (mPGES) or in the cytosol by cytosolic PGES (cPGES). As the inducible form, much attention has been paid to the value of mPGES as a potential therapeutic target, and the efficacy of a variety of inhibitors has been investigated in relevant disease models [88]. However, it is important to note that the precision gained through targeted mPGES inhibition is only one level more selective than COX inhibitors. Instead, we propose that greater therapeutic value can be achieved by targeting prostaglandin receptors, the next level down. Indeed, PGE2 can effect a multitude of functional outcomes, owing its diverse signaling repertoire to the activation of four cell-surface GPCR subtypes, EP1, EP2, EP3, and EP4 (Figure 1), in addition to several functionally relevant EP3 splice variants [89]. Of particular interest to this review, each of these receptors has been studied in relation to experimental AD [46, 9094].

To initially test the relevance of various EP receptors in neurodegenerative disease, several groups developed genetic mouse models of AD that lacked specific PGE2 receptor subtypes and then measured outcomes of cognition, amyloid plaque burden, proinflammatory cytokine expression, and oxidative stress [90, 92]. Amyloid plaque burden was significantly reduced in EP1 knockout mice expressing both the Swedish amyloid precursor protein (APP) and PS1 mutations, which were identified in human familial AD [90]. These mice, when exposed to either LPS or ischemia, were shown to have less neuronal injury and lower secretion of proinflammatory cytokines, TNFα and IL-6, than their WT counterparts [95, 96]. As with any genetic model, potential developmental differences temper interpretation of the data. WT mice exposed to ischemia by carotid occlusion and then treated with SC-51089, a highly specific EP1 antagonist, were shown to have similar neuroprotective phenotypes to EP1 knockout mice [96, 97]. These experiments have yet to be replicated in experimental AD in vivo, but SC-51089 showed highly reduced Aβ neurotoxicity in a neuroblastoma cell line (MC65) and in primary neuron cultures [98, 99]. These data support EP1 as a potential therapeutic target for AD, and a highly selective antagonist such as compound SC-51089, as a potential effector. However, to our knowledge, this drug or those like it have not been clinically tested in AD.

The other three PGE2 receptors have been evaluated in a similar manner to EP1. EP2 receptor knockout AD mice showed reduced oxidative damage in comparison with controls, and, when given a bone marrow transplant of wild type macrophages, showed reduced Aβ plaque size [91, 100]. Interestingly, EP2 receptor knockout mice have cognitive deficits at baseline suggesting an important role for EP2 receptor signaling for neurodevelopment or normal cognitive functioning, and dampening enthusiasm for EP2 receptor clinical trials until the mechanism of this effect is better understood and can be effectively addressed [101]. EP3 receptor deficient AD mice demonstrated reduced proinflammatory gene expression, cytokine production, oxidative stress, and plaque production, as did EP4 receptor knockouts, which additionally demonstrated reduced atherosclerosis [46, 93, 102]. EP4 antagonist AE30208 acted to improve cognitive function and reduce Aβ plaques in an AD model [93]. All PGE2 receptors show positive correlations with Aβ toxicity in culture and in vivo, and therefore represent a class of potential therapeutic targets with the potential to circumvent some of the toxic side effects of NSAIDs. In experimental AD, compounds SC-51089 and AE30208, which target EP1 and EP4, respectively, appear to have some efficacy. Availability of selective EP2 and EP3 receptor antagonists has been limited until recently, and therefore pharmacologic manipulation in experimental AD is less studied, but EP2 and EP3 receptor agonism has been shown to increase APP holoprotein concentrations and infarct size in ischemic insults in mice [103, 104].

7. Potential Prostaglandin Pathway Therapeutics

Understanding which receptor driven pathways are responsible for the neuroinflammation seen in AD requires additional PGE2 receptor compounds to be tested in AD models. These drugs are available, have been used for diverse other studies, and may ultimately have therapeutic value for AD (Table 2). ONO-8713, ONO-8711, and SC-51089 are the most specific and favored EP1 antagonists and have been used effectively in models of colon/intestinal and urinary bladder inflammation [105107]. SC-51809 was shown to be neuroprotective in AD [96, 97], while ONO-8713 and ONO-8711 have not been tested. EP1 agonists such as Iloprost and ONO-D1-004 might be expected to cause neurotoxicity in light of the neuroprotective phenotype of EP1 knockout mice, but these studies have not been done. PF-04418948 is a recently developed, highly selective EP2 antagonist created by Pfizer [108]. While the less specific AH-6809 (EP2 antagonist) has been shown to reduce levels of APP holoprotein in experimental AD, treatment with PF-04418948 has not been reported. Conversely, butaprost, the only EP2 agonist tested so far in experimental AD, was shown to increase levels of APP holoprotein [103]. Compounds AH-13205 and ONO-AE1-259 have been used in bronchodilation and airway smooth muscle research and remain strong candidates for testing in experimental AD [109, 110]. EP3 deficient mice were shown to have reduced proinflammatory gene expression, cytokine production, oxidative stress, and plaque production. ONO-AE3-240 and DG041 are widely used EP3 antagonists used in urinary system research and stroke models, but there is no in vivo testing reported to date with these drugs that corroborates results of EP3 deficiency in experimental AD [111, 112]. Of the EP3 effectors, the most specific agonists are ONO-AE-248 and SC-46275 and are currently primarily used for arterial vessel research [113, 114]. Treatment of AD mice with the highly specific EP4 antagonist, ONO-AE3-208, resulted in an increase in cognitive function and decrease in Aβ plaque [93]. In addition to ONO-AE3-208, GW627368 and L-161, 982 are highly specific EP4 antagonists that need to be evaluated. None of these drugs have been tested clinically.

Table 2.

Effect of prostanoid receptor subtype-specific agonists and antagonists on Alzheimer’s disease-related outcomes in vitro and in vivo.

Receptor Effect Drug AD outcome Reference
EP1a Antagonist SC-51089 ↓Neuronal Injury [96]
↑ Neuroprotective [97]
↓ Aβ toxicity [98]

EP2 Agonist Butaprost ↑ APP holoprotein [103]

EP2 Antagonist AH-6809 ↓ APP holoprotein [103]

EP4a Antagonist ONO-AE3-208 ↑ Cognitive function [93]
↓ Aβ plaque

DPa Agonist BW245C ↑ Neuronal damage [121]

DPa Agonist SQ-27986 ↑ Neuronal damage [121]

DPa Antagonist BWA868C ↑ Neuroprotective [121]

TP Agonist I-BOP ↑ Aβ production [124]

TPa Antagonist Daltroban ↓ Aβ production [124]
↓ APP production [125]

TPa Antagonist S18886 ↓ Aβ production [124]
↓ APP production [125]

Aβ (amyloid beta); APP (amyloid precursor protein);

a

Highly specific effector.

Though PGE2 is the principal prostaglandin in the CNS and is of primary interest, prostaglandin receptors to PGD2, PGF2a, and TXA2 have all been shown to have biological relevance in AD (Table 2) [115117]. Frontal cortical brain samples from later diagnosed AD patients had decreased microsomal PGD2 and PGF2a production [118]. In LOAD brain samples, PGD2 synthase was found to be highly associated with Aβ plaques[119]. Additionally, PGD synthase/transthyretin complex levels were found to be six times higher in LOAD and MCI patients than in controls[120]. Taken together, observational data indicates that PGD2 is associated with AD clinical and neuropathological phenotype. Experimental studies suggest that Aβ plaques may modulate PGD2 synthesis by interfering with PGD2 synthase. In microglia/neuron co-cultures, BW245C and SQ27986, both DP (PGD2 receptor) agonists, were shown to result in microglia-mediated neuronal toxicity. Conversely, neuroprotective properties have been ascribed to DP antagonist BWA868C, presenting a possible pathway for therapeutic drug intervention [121]. The role of PGF2a and TXA2 in AD is less clear. Both molecules are elevated in urine samples from probable AD patients [122, 123], and administration of thromboxane receptor (TP) antagonists Daltroban and S18886 result in a reduction of Aβ. Conversely, TP receptor agonist I-BOP resulted in increased Aβ levels, further demonstrating the therapeutic potential of modulation of individual prostaglandin receptor effectors [124, 125]. Other non-PGE2 receptors and drugs can be found in Table 2. It should be noted that there are currently no specific FP receptor antagonists.

8. Conclusion

Over 30 epidemiological studies have been reported on NSAIDs in AD, most indicating NSAID administration was associated with reduced incidence of AD [13], but NSAID toxicity has complicated efforts for long term prevention trials. Further studies are needed to identify downstream targets of COX cascades to deliver this therapeutic benefit whilst avoiding intolerable side effects. Chief among these COX-dependent pathways are the prostaglandins and PGE2. While NSAIDS result in broad suppression of prostaglandin signaling through COX inhibition, selective prostaglandin receptor effectors have the potential advantage of targeting mechanisms specific to pathogenesis whilst sparing pathways critical for other aspects of normal neuro- and peripheral immunoregulation. There is much to be learned, but studies demonstrating improved cognitive function, reduced plaque burden, and suppressed free radical injury through antagonism of EP1, EP4, DP, and TP receptors hold promise for future targeted drug therapies. There are currently an abundance of prostaglandin receptor agonists and antagonists that are readily available and have been tested in diverse models of inflammatory disease. However, only a scant few of these effectors have ever been tested for efficacy in experimental AD. Future studies are needed to determine which molecular components of the prostaglandin pathway can be targeted for as preventive or therapeutic interventions for AD.

Acknowledgments

Supported by grants from the National Institutes of Health (ES16754 and P50AG05136) and the Nancy and Buster Alvord Endowment

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.McGeer PL, Itagaki S, Tago H, McGeer EG. Reactive microglia in patients with senile dementia of the Alzheimer type are positive for the histocompatibility glycoprotein HLA-DR. Neurosci Lett. 1987;79:195–200. doi: 10.1016/0304-3940(87)90696-3. [DOI] [PubMed] [Google Scholar]
  • 2.Rogers J, Luber-Narod J, Styren SD, Civin WH. Expression of immune system-associated antigens by cells of the human central nervous system: relationship to the pathology of Alzheimer’s disease. Neurobiol Aging. 1988;9:339–49. doi: 10.1016/s0197-4580(88)80079-4. [DOI] [PubMed] [Google Scholar]
  • 3.Itagaki S, McGeer PL, Akiyama H. Presence of T-cytotoxic suppressor and leucocyte common antigen positive cells in Alzheimer’s disease brain tissue. Neurosci Lett. 1988;91:259–64. doi: 10.1016/0304-3940(88)90690-8. [DOI] [PubMed] [Google Scholar]
  • 4.Eikelenboom P, Hack CE, Rozemuller JM, Stam FC. Complement activation in amyloid plaques in Alzheimer’s dementia. Virchows Arch B Cell Pathol Incl Mol Pathol. 1989;56:259–62. doi: 10.1007/BF02890024. [DOI] [PubMed] [Google Scholar]
  • 5.Cribbs DH, Berchtold NC, Perreau V, Coleman PD, Rogers J, Tenner AJ, et al. Extensive innate immune gene activation accompanies brain aging, increasing vulnerability to cognitive decline and neurodegeneration: a microarray study. J Neuroinflammation. 2012;9:179. doi: 10.1186/1742-2094-9-179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Bruunsgaard H, Pedersen BK. Age-related inflammatory cytokines and disease. Immunol Allergy Clin North Am. 2003;23:15–39. doi: 10.1016/s0889-8561(02)00056-5. [DOI] [PubMed] [Google Scholar]
  • 7.Starr ME, Evers BM, Saito H. Age-associated increase in cytokine production during systemic inflammation: adipose tissue as a major source of IL-6. J Gerontol A Biol Sci Med Sci. 2009;64:723–30. doi: 10.1093/gerona/glp046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Casolini P, Catalani A, Zuena AR, Angelucci L. Inhibition of COX-2 reduces the age-dependent increase of hippocampal inflammatory markers, corticosterone secretion, and behavioral impairments in the rat. J Neurosci Res. 2002;68:337–43. doi: 10.1002/jnr.10192. [DOI] [PubMed] [Google Scholar]
  • 9.Simen AA, Bordner KA, Martin MP, Moy LA, Barry LC. Cognitive dysfunction with aging and the role of inflammation. Ther Adv Chronic Dis. 2011;2:175–95. doi: 10.1177/2040622311399145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Holmes C, Cunningham C, Zotova E, Woolford J, Dean C, Kerr S, et al. Systemic inflammation and disease progression in Alzheimer disease. Neurology. 2009;73:768–74. doi: 10.1212/WNL.0b013e3181b6bb95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Noble JM, Manly JJ, Schupf N, Tang MX, Mayeux R, Luchsinger JA. Association of C-reactive protein with cognitive impairment. Arch Neurol. 2010;67:87–92. doi: 10.1001/archneurol.2009.308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Cooper-Knock J, Kirby J, Ferraiuolo L, Heath PR, Rattray M, Shaw PJ. Gene expression profiling in human neurodegenerative disease. Nat Rev Neurol. 2012;8:518–30. doi: 10.1038/nrneurol.2012.156. [DOI] [PubMed] [Google Scholar]
  • 13.Wyss-Coray T, Rogers J. Inflammation in Alzheimer disease-a brief review of the basic science and clinical literature. Cold Spring Harb Perspect Med. 2012;2:a006346. doi: 10.1101/cshperspect.a006346. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Michaud M, Balardy L, Moulis G, Gaudin C, Peyrot C, Vellas B, et al. Proinflammatory Cytokines, Aging, and Age-Related Diseases. J Am Med Dir Assoc. 2013 doi: 10.1016/j.jamda.2013.05.009. [DOI] [PubMed] [Google Scholar]
  • 15.Szekely CA, Zandi PP. Non-steroidal anti-inflammatory drugs and Alzheimer’s disease: the epidemiological evidence. CNS Neurol Disord Drug Targets. 2010;9:132–9. doi: 10.2174/187152710791012026. [DOI] [PubMed] [Google Scholar]
  • 16.McGeer PL, Itagaki S, Tago H, McGeer EG. Occurrence of HLA-DR reactive microglia in Alzheimer’s disease. Ann N Y Acad Sci. 1988;540:319–23. doi: 10.1111/j.1749-6632.1988.tb27086.x. [DOI] [PubMed] [Google Scholar]
  • 17.McGeer PL, Akiyama H, Itagaki S, McGeer EG. Activation of the classical complement pathway in brain tissue of Alzheimer patients. Neurosci Lett. 1989;107:341–6. doi: 10.1016/0304-3940(89)90843-4. [DOI] [PubMed] [Google Scholar]
  • 18.McGeer PL, McGeer E, Rogers J, Sibley J. Anti-inflammatory drugs and Alzheimer disease. Lancet. 1990;335:1037. doi: 10.1016/0140-6736(90)91101-f. [DOI] [PubMed] [Google Scholar]
  • 19.Beard CM, Kokman E, Kurland LT. Rheumatoid arthritis and susceptibility to Alzheimer’s disease. Lancet. 1991;337:1426. doi: 10.1016/0140-6736(91)93122-p. [DOI] [PubMed] [Google Scholar]
  • 20.Rogers J, Kirby LC, Hempelman SR, Berry DL, McGeer PL, Kaszniak AW, et al. Clinical trial of indomethacin in Alzheimer’s disease. Neurology. 1993;43:1609–11. doi: 10.1212/wnl.43.8.1609. [DOI] [PubMed] [Google Scholar]
  • 21.Breitner JC, Gau BA, Welsh KA, Plassman BL, McDonald WM, Helms MJ, et al. Inverse association of anti-inflammatory treatments and Alzheimer’s disease: initial results of a co-twin control study. Neurology. 1994;44:227–32. doi: 10.1212/wnl.44.2.227. [DOI] [PubMed] [Google Scholar]
  • 22.Rich JB, Rasmusson DX, Folstein MF, Carson KA, Kawas C, Brandt J. Nonsteroidal anti-inflammatory drugs in Alzheimer’s disease. Neurology. 1995;45:51–5. doi: 10.1212/wnl.45.1.51. [DOI] [PubMed] [Google Scholar]
  • 23.Stewart WF, Kawas C, Corrada M, Metter EJ. Risk of Alzheimer’s disease and duration of NSAID use. Neurology. 1997;48:626–32. doi: 10.1212/wnl.48.3.626. [DOI] [PubMed] [Google Scholar]
  • 24.The Canadian Study of Health and Aging: risk factors for Alzheimer’s disease in Canada. Neurology. 1994;44:2073–80. doi: 10.1212/wnl.44.11.2073. [DOI] [PubMed] [Google Scholar]
  • 25.Beard CM, Waring SC, O’Brien PC, Kurland LT, Kokmen E. Nonsteroidal anti-inflammatory drug use and Alzheimer’s disease: a case-control study in Rochester, Minnesota, 1980 through 1984. Mayo Clin Proc. 1998;73:951–5. doi: 10.4065/73.10.951. [DOI] [PubMed] [Google Scholar]
  • 26.Choi JK, Jenkins BG, Carreras I, Kaymakcalan S, Cormier K, Kowall NW, et al. Anti-inflammatory treatment in AD mice protects against neuronal pathology. Exp Neurol. 2010;223:377–84. doi: 10.1016/j.expneurol.2009.07.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Varvel NH, Bhaskar K, Kounnas MZ, Wagner SL, Yang Y, Lamb BT, et al. NSAIDs prevent, but do not reverse, neuronal cell cycle reentry in a mouse model of Alzheimer disease. J Clin Invest. 2009;119:3692–702. doi: 10.1172/JCI39716. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Sung S, Yang H, Uryu K, Lee EB, Zhao L, Shineman D, et al. Modulation of nuclear factor-kappa B activity by indomethacin influences A beta levels but not A beta precursor protein metabolism in a model of Alzheimer’s disease. Am J Pathol. 2004;165:2197–206. doi: 10.1016/s0002-9440(10)63269-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Lim GP, Yang F, Chu T, Chen P, Beech W, Teter B, et al. Ibuprofen suppresses plaque pathology and inflammation in a mouse model for Alzheimer’s disease. J Neurosci. 2000;20:5709–14. doi: 10.1523/JNEUROSCI.20-15-05709.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Lim GP, Yang F, Chu T, Gahtan E, Ubeda O, Beech W, et al. Ibuprofen effects on Alzheimer pathology and open field activity in APPsw transgenic mice. Neurobiol Aging. 2001;22:983–91. doi: 10.1016/s0197-4580(01)00299-8. [DOI] [PubMed] [Google Scholar]
  • 31.McKee AC, Carreras I, Hossain L, Ryu H, Klein WL, Oddo S, et al. Ibuprofen reduces Abeta, hyperphosphorylated tau and memory deficits in Alzheimer mice. Brain Res. 2008;1207:225–36. doi: 10.1016/j.brainres.2008.01.095. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Choi SH, Aid S, Caracciolo L, Minami SS, Niikura T, Matsuoka Y, et al. Cyclooxygenase-1 inhibition reduces amyloid pathology and improves memory deficits in a mouse model of Alzheimer’s disease. J Neurochem. 2013;124:59–68. doi: 10.1111/jnc.12059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Lyketsos CG, Breitner JC, Green RC, Martin BK, Meinert C, Piantadosi S, et al. Naproxen and celecoxib do not prevent AD in early results from a randomized controlled trial. Neurology. 2007;68:1800–8. doi: 10.1212/01.wnl.0000260269.93245.d2. [DOI] [PubMed] [Google Scholar]
  • 34.Martin BK, Szekely C, Brandt J, Piantadosi S, Breitner JC, Craft S, et al. Cognitive function over time in the Alzheimer’s Disease Anti-inflammatory Prevention Trial (ADAPT): results of a randomized, controlled trial of naproxen and celecoxib. Arch Neurol. 2008;65:896–905. doi: 10.1001/archneur.2008.65.7.nct70006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Breitner JC, Martin BK, Meinert CL. The suspension of treatments in ADAPT: concerns beyond the cardiovascular safety of celecoxib or naproxen. PLoS Clin Trials. 2006;1:e41. doi: 10.1371/journal.pctr.0010041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Leoutsakos JM, Muthen BO, Breitner JC, Lyketsos CG. Effects of non-steroidal anti-inflammatory drug treatments on cognitive decline vary by phase of pre-clinical Alzheimer disease: findings from the randomized controlled Alzheimer’s Disease Anti-inflammatory Prevention Trial. Int J Geriatr Psychiatry. 2012;27:364–74. doi: 10.1002/gps.2723. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Breitner JC, Baker LD, Montine TJ, Meinert CL, Lyketsos CG, Ashe KH, et al. Extended results of the Alzheimer’s disease anti-inflammatory prevention trial. Alzheimers Dement. 2011;7:402–11. doi: 10.1016/j.jalz.2010.12.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Zahs KR, Ashe KH. ‘Too much good news’ - are Alzheimer mouse models trying to tell us how to prevent, not cure, Alzheimer’s disease? Trends Neurosci. 2010;33:381–9. doi: 10.1016/j.tins.2010.05.004. [DOI] [PubMed] [Google Scholar]
  • 39.McGettigan P, Henry D. Cardiovascular risk with non-steroidal anti-inflammatory drugs: systematic review of population-based controlled observational studies. PLoS Med. 2011;8:e1001098. doi: 10.1371/journal.pmed.1001098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Liu B, Zhang Y, Zhu N, Li H, Luo W, Zhou Y. A vasoconstrictor role for cyclooxygenase-1-mediated prostacyclin synthesis in mouse renal arteries. Am J Physiol Renal Physiol. 2013 doi: 10.1152/ajprenal.00332.2013. [DOI] [PubMed] [Google Scholar]
  • 41.Vitale P, Tacconelli S, Perrone MG, Malerba P, Simone L, Scilimati A, et al. Synthesis, pharmacological characterization, and docking analysis of a novel family of diarylisoxazoles as highly selective cyclooxygenase-1 (COX-1) inhibitors. J Med Chem. 2013;56:4277–99. doi: 10.1021/jm301905a. [DOI] [PubMed] [Google Scholar]
  • 42.Edelman AB, Jensen JT, Doom C, Hennebold JD. Impact of the prostaglandin synthase-2 inhibitor celecoxib on ovulation and luteal events in women. Contraception. 2013;87:352–7. doi: 10.1016/j.contraception.2012.07.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Vogel JP, West HM, Dowswell T. Titrated oral misoprostol for augmenting labour to improve maternal and neonatal outcomes. Cochrane Database Syst Rev. 2013;9:CD010648. doi: 10.1002/14651858.CD010648.pub2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Al-Kharusi MR, Smartt HJ, Greenhough A, Collard TJ, Emery ED, Williams AC, et al. LGR5 promotes survival in human colorectal adenoma cells and is upregulated by PGE2: implications for targeting adenoma stem cells with NSAIDs. Carcinogenesis. 2013;34:1150–7. doi: 10.1093/carcin/bgt020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Dai ZJ, Ma XB, Kang HF, Gao J, Min WL, Guan HT, et al. Antitumor activity of the selective cyclooxygenase-2 inhibitor, celecoxib, on breast cancer in Vitro and in Vivo. Cancer Cell Int. 2012;12:53. doi: 10.1186/1475-2867-12-53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Shi J, Wang Q, Johansson JU, Liang X, Woodling NS, Priyam P, et al. Inflammatory prostaglandin E2 signaling in a mouse model of Alzheimer disease. Ann Neurol. 2012;72:788–98. doi: 10.1002/ana.23677. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Desbene C, Malaplate-Armand C, Youssef I, Garcia P, Stenger C, Sauvee M, et al. Critical role of cPLA2 in Abeta oligomer-induced neurodegeneration and memory deficit. Neurobiol Aging. 2012;33:1123, e17–29. doi: 10.1016/j.neurobiolaging.2011.11.008. [DOI] [PubMed] [Google Scholar]
  • 48.Sagy-Bross C, Hadad N, Levy R. Cytosolic phospholipase Aalpha upregulation mediates apoptotic neuronal death induced by aggregated amyloid-beta peptide. Neurochem Int. 2013;63:541–550. doi: 10.1016/j.neuint.2013.09.007. [DOI] [PubMed] [Google Scholar]
  • 49.Rosenberger TA, Villacreses NE, Contreras MA, Bonventre JV, Rapoport SI. Brain lipid metabolism in the cPLA2 knockout mouse. J Lipid Res. 2003;44:109–17. doi: 10.1194/jlr.m200298-jlr200. [DOI] [PubMed] [Google Scholar]
  • 50.Reisenberg M, Singh PK, Williams G, Doherty P. The diacylglycerol lipases: structure, regulation and roles in and beyond endocannabinoid signalling. Philos Trans R Soc Lond B Biol Sci. 2012;367:3264–75. doi: 10.1098/rstb.2011.0387. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Nomura DK, Morrison BE, Blankman JL, Long JZ, Kinsey SG, Marcondes MC, et al. Endocannabinoid hydrolysis generates brain prostaglandins that promote neuroinflammation. Science. 2011;334:809–13. doi: 10.1126/science.1209200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Greene ER, Huang S, Serhan CN, Panigrahy D. Regulation of inflammation in cancer by eicosanoids. Prostaglandins Other Lipid Mediat. 2011;96:27–36. doi: 10.1016/j.prostaglandins.2011.08.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Vegiopoulos A, Muller-Decker K, Strzoda D, Schmitt I, Chichelnitskiy E, Ostertag A, et al. Cyclooxygenase-2 controls energy homeostasis in mice by de novo recruitment of brown adipocytes. Science. 2010;328:1158–61. doi: 10.1126/science.1186034. [DOI] [PubMed] [Google Scholar]
  • 54.Dinchuk JE, Car BD, Focht RJ, Johnston JJ, Jaffee BD, Covington MB, et al. Renal abnormalities and an altered inflammatory response in mice lacking cyclooxygenase II. Nature. 1995;378:406–9. doi: 10.1038/378406a0. [DOI] [PubMed] [Google Scholar]
  • 55.Lim H, Paria BC, Das SK, Dinchuk JE, Langenbach R, Trzaskos JM, et al. Multiple female reproductive failures in cyclooxygenase 2-deficient mice. Cell. 1997;91:197–208. doi: 10.1016/s0092-8674(00)80402-x. [DOI] [PubMed] [Google Scholar]
  • 56.Bresalier RS, Sandler RS, Quan H, Bolognese JA, Oxenius B, Horgan K, et al. Cardiovascular events associated with rofecoxib in a colorectal adenoma chemoprevention trial. N Engl J Med. 2005;352:1092–102. doi: 10.1056/NEJMoa050493. [DOI] [PubMed] [Google Scholar]
  • 57.Trelle S, Reichenbach S, Wandel S, Hildebrand P, Tschannen B, Villiger PM, et al. Cardiovascular safety of non-steroidal anti-inflammatory drugs: network meta-analysis. Bmj. 2011;342:c7086. doi: 10.1136/bmj.c7086. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Hoozemans JJ, Rozemuller JM, van Haastert ES, Veerhuis R, Eikelenboom P. Cyclooxygenase-1 and -2 in the different stages of Alzheimer’s disease pathology. Curr Pharm Des. 2008;14:1419–27. doi: 10.2174/138161208784480171. [DOI] [PubMed] [Google Scholar]
  • 59.Kitamura Y, Shimohama S, Koike H, Kakimura J, Matsuoka Y, Nomura Y, et al. Increased expression of cyclooxygenases and peroxisome proliferator-activated receptor-gamma in Alzheimer’s disease brains. Biochem Biophys Res Commun. 1999;254:582–6. doi: 10.1006/bbrc.1998.9981. [DOI] [PubMed] [Google Scholar]
  • 60.Chandrasekharan NV, Dai H, Roos KL, Evanson NK, Tomsik J, Elton TS, et al. COX-3, a cyclooxygenase-1 variant inhibited by acetaminophen and other analgesic/antipyretic drugs: cloning, structure, and expression. Proc Natl Acad Sci U S A. 2002;99:13926–31. doi: 10.1073/pnas.162468699. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Cui JG, Kuroda H, Chandrasekharan NV, Pelaez RP, Simmons DL, Bazan NG, et al. Cyclooxygenase-3 gene expression in Alzheimer hippocampus and in stressed human neural cells. Neurochem Res. 2004;29:1731–7. doi: 10.1023/b:nere.0000035809.70905.8a. [DOI] [PubMed] [Google Scholar]
  • 62.Dhull DK, Bhateja D, Dhull RK, Padi SS. Differential role of cyclooxygenase isozymes on neuronal density in hippocampus CA1 region of intracerebroventricular streptozotocin treated rat brain. J Chem Neuroanat. 2012;43:48–51. doi: 10.1016/j.jchemneu.2011.10.001. [DOI] [PubMed] [Google Scholar]
  • 63.Chandrasekharan NV, Simmons DL. The cyclooxygenases. Genome Biol. 2004;5:241. doi: 10.1186/gb-2004-5-9-241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Aid S, Bosetti F. Gene expression of cyclooxygenase-1 and Ca(2+)-independent phospholipase A(2) is altered in rat hippocampus during normal aging. Brain Res Bull. 2007;73:108–13. doi: 10.1016/j.brainresbull.2007.02.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Weerasinghe GR, Coon SL, Bhattacharjee AK, Harry GJ, Bosetti F. Regional protein levels of cytosolic phospholipase A2 and cyclooxygenase-2 in Rhesus monkey brain as a function of age. Brain Res Bull. 2006;69:614–21. doi: 10.1016/j.brainresbull.2006.03.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Lukiw WJ. Gene expression profiling in fetal, aged, and Alzheimer hippocampus: a continuum of stress-related signaling. Neurochem Res. 2004;29:1287–97. doi: 10.1023/b:nere.0000023615.89699.63. [DOI] [PubMed] [Google Scholar]
  • 67.Heneka MT, Sastre M, Dumitrescu-Ozimek L, Hanke A, Dewachter I, Kuiperi C, et al. Acute treatment with the PPARgamma agonist pioglitazone and ibuprofen reduces glial inflammation and Abeta1–42 levels in APPV717I transgenic mice. Brain. 2005;128:1442–53. doi: 10.1093/brain/awh452. [DOI] [PubMed] [Google Scholar]
  • 68.Jantzen PT, Connor KE, DiCarlo G, Wenk GL, Wallace JL, Rojiani AM, et al. Microglial activation and beta -amyloid deposit reduction caused by a nitric oxide-releasing nonsteroidal anti-inflammatory drug in amyloid precursor protein plus presenilin-1 transgenic mice. J Neurosci. 2002;22:2246–54. doi: 10.1523/JNEUROSCI.22-06-02246.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Choi SH, Aid S, Caracciolo L, Minami SS, Niikura T, Matsuoka Y, et al. Cyclooxygenase-1 inhibition reduces amyloid pathology and improves memory deficits in a mouse model of Alzheimer’s disease. J Neurochem. 2012;124:59–68. doi: 10.1111/jnc.12059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Wilkinson BL, Cramer PE, Varvel NH, Reed-Geaghan E, Jiang Q, Szabo A, et al. Ibuprofen attenuates oxidative damage through NOX2 inhibition in Alzheimer’s disease. Neurobiol Aging. 2010;33:197, e21–32. doi: 10.1016/j.neurobiolaging.2010.06.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Choi SH, Bosetti F. Cyclooxygenase-1 null mice show reduced neuroinflammation in response to beta-amyloid. Aging (Albany NY) 2009;1:234–44. doi: 10.18632/aging.100021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Coma M, Sereno L, Da Rocha-Souto B, Scotton TC, Espana J, Sanchez MB, et al. Triflusal reduces dense-core plaque load, associated axonal alterations and inflammatory changes, and rescues cognition in a transgenic mouse model of Alzheimer’s disease. Neurobiol Dis. 2010;38:482–91. doi: 10.1016/j.nbd.2010.01.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Kukar T, Murphy MP, Eriksen JL, Sagi SA, Weggen S, Smith TE, et al. Diverse compounds mimic Alzheimer disease-causing mutations by augmenting Abeta42 production. Nat Med. 2005;11:545–50. doi: 10.1038/nm1235. [DOI] [PubMed] [Google Scholar]
  • 74.Yermakova AV, Rollins J, Callahan LM, Rogers J, O’Banion MK. Cyclooxygenase-1 in human Alzheimer and control brain: quantitative analysis of expression by microglia and CA3 hippocampal neurons. J Neuropathol Exp Neurol. 1999;58:1135–46. doi: 10.1097/00005072-199911000-00003. [DOI] [PubMed] [Google Scholar]
  • 75.Hoozemans JJ, Rozemuller AJ, Janssen I, De Groot CJ, Veerhuis R, Eikelenboom P. Cyclooxygenase expression in microglia and neurons in Alzheimer’s disease and control brain. Acta Neuropathol. 2001;101:2–8. doi: 10.1007/s004010000251. [DOI] [PubMed] [Google Scholar]
  • 76.Eriksen JL, Sagi SA, Smith TE, Weggen S, Das P, McLendon DC, et al. NSAIDs and enantiomers of flurbiprofen target gamma-secretase and lower Abeta 42 in vivo. J Clin Invest. 2003;112:440–9. doi: 10.1172/JCI18162. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Weggen S, Eriksen JL, Sagi SA, Pietrzik CU, Ozols V, Fauq A, et al. Evidence that nonsteroidal anti-inflammatory drugs decrease amyloid beta 42 production by direct modulation of gamma-secretase activity. J Biol Chem. 2003;278:31831–7. doi: 10.1074/jbc.M303592200. [DOI] [PubMed] [Google Scholar]
  • 78.Borgegard T, Jureus A, Olsson F, Rosqvist S, Sabirsh A, Rotticci D, et al. First and second generation gamma-secretase modulators (GSMs) modulate amyloid-beta (Abeta) peptide production through different mechanisms. J Biol Chem. 2012;287:11810–9. doi: 10.1074/jbc.M111.305227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Weggen S, Eriksen JL, Das P, Sagi SA, Wang R, Pietrzik CU, et al. A subset of NSAIDs lower amyloidogenic Abeta42 independently of cyclooxygenase activity. Nature. 2001;414:212–6. doi: 10.1038/35102591. [DOI] [PubMed] [Google Scholar]
  • 80.Kukar TL, Ladd TB, Bann MA, Fraering PC, Narlawar R, Maharvi GM, et al. Substrate-targeting gamma-secretase modulators. Nature. 2008;453:925–9. doi: 10.1038/nature07055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Green RC, Schneider LS, Amato DA, Beelen AP, Wilcock G, Swabb EA, et al. Effect of tarenflurbil on cognitive decline and activities of daily living in patients with mild Alzheimer disease: a randomized controlled trial. Jama. 2009;302:2557–64. doi: 10.1001/jama.2009.1866. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Wilcock GK, Black SE, Hendrix SB, Zavitz KH, Swabb EA, Laughlin MA. Efficacy and safety of tarenflurbil in mild to moderate Alzheimer’s disease: a randomised phase II trial. Lancet Neurol. 2008;7:483–93. doi: 10.1016/S1474-4422(08)70090-5. [DOI] [PubMed] [Google Scholar]
  • 83.Szekely CA, Green RC, Breitner JC, Ostbye T, Beiser AS, Corrada MM, et al. No advantage of A beta 42-lowering NSAIDs for prevention of Alzheimer dementia in six pooled cohort studies. Neurology. 2008;70:2291–8. doi: 10.1212/01.wnl.0000313933.17796.f6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Chan BS, Satriano JA, Pucci M, Schuster VL. Mechanism of prostaglandin E2 transport across the plasma membrane of HeLa cells and Xenopus oocytes expressing the prostaglandin transporter “PGT”. J Biol Chem. 1998;273:6689–97. doi: 10.1074/jbc.273.12.6689. [DOI] [PubMed] [Google Scholar]
  • 85.Kanai N, Lu R, Satriano JA, Bao Y, Wolkoff AW, Schuster VL. Identification and characterization of a prostaglandin transporter. Science. 1995;268:866–9. doi: 10.1126/science.7754369. [DOI] [PubMed] [Google Scholar]
  • 86.Lu R, Kanai N, Bao Y, Schuster VL. Cloning, in vitro expression, and tissue distribution of a human prostaglandin transporter cDNA(hPGT) J Clin Invest. 1996;98:1142–9. doi: 10.1172/JCI118897. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Milatovic D, Montine TJ, Aschner M. Prostanoid signaling: dual role for prostaglandin E2 in neurotoxicity. Neurotoxicology. 2011;32:312–9. doi: 10.1016/j.neuro.2011.02.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Chang HH, Meuillet EJ. Identification and development of mPGES-1 inhibitors: where we are at? Future Med Chem. 2011;3:1909–34. doi: 10.4155/fmc.11.136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Israel DD, Regan JW. EP(3) prostanoid receptor isoforms utilize distinct mechanisms to regulate ERK 1/2 activation. Biochim Biophys Acta. 2009;1791:238–45. doi: 10.1016/j.bbalip.2009.01.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Zhen G, Kim YT, Li RC, Yocum J, Kapoor N, Langer J, et al. PGE2 EP1 receptor exacerbated neurotoxicity in a mouse model of cerebral ischemia and Alzheimer’s disease. Neurobiol Aging. 2012;33:2215–9. doi: 10.1016/j.neurobiolaging.2011.09.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Keene CD, Chang RC, Lopez-Yglesias AH, Shalloway BR, Sokal I, Li X, et al. Suppressed accumulation of cerebral amyloid {beta} peptides in aged transgenic Alzheimer’s disease mice by transplantation with wild-type or prostaglandin E2 receptor subtype 2-null bone marrow. Am J Pathol. 2010;177:346–54. doi: 10.2353/ajpath.2010.090840. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Liang X, Wang Q, Hand T, Wu L, Breyer RM, Montine TJ, et al. Deletion of the prostaglandin E2 EP2 receptor reduces oxidative damage and amyloid burden in a model of Alzheimer’s disease. J Neurosci. 2005;25:10180–7. doi: 10.1523/JNEUROSCI.3591-05.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Hoshino T, Namba T, Takehara M, Murao N, Matsushima T, Sugimoto Y, et al. Improvement of cognitive function in Alzheimer’s disease model mice by genetic and pharmacological inhibition of the EP(4) receptor. J Neurochem. 2012;120:795–805. doi: 10.1111/j.1471-4159.2011.07567.x. [DOI] [PubMed] [Google Scholar]
  • 94.Hoshino T, Namba T, Takehara M, Nakaya T, Sugimoto Y, Araki W, et al. Prostaglandin E2 stimulates the production of amyloid-beta peptides through internalization of the EP4 receptor. J Biol Chem. 2009;284:18493–502. doi: 10.1074/jbc.M109.003269. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Keene CD, Chang R, Stephen C, Nivison M, Nutt SE, Look A, et al. Protection of hippocampal neurogenesis from toll-like receptor 4-dependent innate immune activation by ablation of prostaglandin E2 receptor subtype EP1 or EP2. Am J Pathol. 2009;174:2300–9. doi: 10.2353/ajpath.2009.081153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Shimamura M, Zhou P, Casolla B, Qian L, Capone C, Kurinami H, et al. Prostaglandin E2 type 1 receptors contribute to neuronal apoptosis after transient forebrain ischemia. J Cereb Blood Flow Metab. 2013;33:1207–14. doi: 10.1038/jcbfm.2013.69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Abe T, Kunz A, Shimamura M, Zhou P, Anrather J, Iadecola C. The neuroprotective effect of prostaglandin E2 EP1 receptor inhibition has a wide therapeutic window, is sustained in time and is not sexually dimorphic. J Cereb Blood Flow Metab. 2009;29:66–72. doi: 10.1038/jcbfm.2008.88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Li X, Rose SE, Montine KS, Keene CD, Montine TJ. Antagonism of neuronal prostaglandin E(2) receptor subtype 1 mitigates amyloid beta neurotoxicity in vitro. J Neuroimmune Pharmacol. 2013;8:87–93. doi: 10.1007/s11481-012-9380-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Rohrbeck CA, Twentyman CT. Multimodal assessment of impulsiveness in abusing, neglecting, and nonmaltreating mothers and their preschool children. J Consult Clin Psychol. 1986;54:231–6. doi: 10.1037//0022-006x.54.2.231. [DOI] [PubMed] [Google Scholar]
  • 100.Cimino PJ, Yang Y, Li X, Hemingway JF, Cherne MK, Khademi SB, et al. Ablation of the microglial protein DOCK2 reduces amyloid burden in a mouse model of Alzheimer’s disease. Exp Mol Pathol. 2013;94:366–71. doi: 10.1016/j.yexmp.2013.01.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Savonenko A, Munoz P, Melnikova T, Wang Q, Liang X, Breyer RM, et al. Impaired cognition, sensorimotor gating, and hippocampal long-term depression in mice lacking the prostaglandin E2 EP2 receptor. Exp Neurol. 2009;217:63–73. doi: 10.1016/j.expneurol.2009.01.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Shi J, Johansson J, Woodling NS, Wang Q, Montine TJ, Andreasson K. The prostaglandin E2 E-prostanoid 4 receptor exerts anti-inflammatory effects in brain innate immunity. J Immunol. 2010;184:7207–18. doi: 10.4049/jimmunol.0903487. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Pooler AM, Arjona AA, Lee RK, Wurtman RJ. Prostaglandin E2 regulates amyloid precursor protein expression via the EP2 receptor in cultured rat microglia. Neurosci Lett. 2004;362:127–30. doi: 10.1016/j.neulet.2004.03.013. [DOI] [PubMed] [Google Scholar]
  • 104.Ahmad M, Ahmad AS, Zhuang H, Maruyama T, Narumiya S, Dore S. Stimulation of prostaglandin E2-EP3 receptors exacerbates stroke and excitotoxic injury. J Neuroimmunol. 2007;184:172–9. doi: 10.1016/j.jneuroim.2006.12.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.O’Callaghan G, Ryan A, Neary P, O’Mahony C, Shanahan F, Houston A. Targeting the EP1 receptor reduces Fas ligand expression and increases the antitumor immune response in an in vivo model of colon cancer. Int J Cancer. 2013;133:825–34. doi: 10.1002/ijc.28076. [DOI] [PubMed] [Google Scholar]
  • 106.Ikeda M, Kawatani M, Maruyama T, Ishihama H. Prostaglandin facilitates afferent nerve activity via EP1 receptors during urinary bladder inflammation in rats. Biomed Res. 2006;27:49–54. doi: 10.2220/biomedres.27.49. [DOI] [PubMed] [Google Scholar]
  • 107.Shahbazian A, Heinemann A, Peskar BA, Holzer P. Differential peristaltic motor effects of prostanoid (DP, EP, IP, TP) and leukotriene receptor agonists in the guinea-pig isolated small intestine. Br J Pharmacol. 2002;137:1047–54. doi: 10.1038/sj.bjp.0704958. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Birrell MA, Maher SA, Buckley J, Dale N, Bonvini S, Raemdonck K, et al. Selectivity profiling of the novel EP2 receptor antagonist, PF-04418948, in functional bioassay systems: atypical affinity at the guinea pig EP2 receptor. Br J Pharmacol. 2013;168:129–38. doi: 10.1111/j.1476-5381.2012.02088.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Aso H, Ito S, Mori A, Suganuma N, Morioka M, Takahara N, et al. Differential regulation of airway smooth muscle cell migration by E-prostanoid receptor subtypes. Am J Respir Cell Mol Biol. 2013;48:322–9. doi: 10.1165/rcmb.2012-0158OC. [DOI] [PubMed] [Google Scholar]
  • 110.Buckley J, Birrell MA, Maher SA, Nials AT, Clarke DL, Belvisi MG. EP4 receptor as a new target for bronchodilator therapy. Thorax. 2011;66:1029–35. doi: 10.1136/thx.2010.158568. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Jugus MJ, Jaworski JP, Patra PB, Jin J, Morrow DM, Laping NJ, et al. Dual modulation of urinary bladder activity and urine flow by prostanoid EP3 receptors in the conscious rat. Br J Pharmacol. 2009;158:372–81. doi: 10.1111/j.1476-5381.2009.00275.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Ikeda-Matsuo Y, Tanji H, Narumiya S, Sasaki Y. Inhibition of prostaglandin E2 EP3 receptors improves stroke injury via anti-inflammatory and anti-apoptotic mechanisms. J Neuroimmunol. 2011;238:34–43. doi: 10.1016/j.jneuroim.2011.06.014. [DOI] [PubMed] [Google Scholar]
  • 113.Hung GH, Jones RL, Lam FF, Chan KM, Hidaka H, Suzuki M, et al. Investigation of the pronounced synergism between prostaglandin E2 and other constrictor agents on rat femoral artery. Prostaglandins Leukot Essent Fatty Acids. 2006;74:401–15. doi: 10.1016/j.plefa.2006.04.002. [DOI] [PubMed] [Google Scholar]
  • 114.Longrois D, Gomez I, Foudi N, Topal G, Dhaouadi M, Kotelevets L, et al. Prostaglandin E(2) induced contraction of human intercostal arteries is mediated by the EP(3) receptor. Eur J Pharmacol. 2012;681:55–9. doi: 10.1016/j.ejphar.2012.01.041. [DOI] [PubMed] [Google Scholar]
  • 115.Mohri I, Kadoyama K, Kanekiyo T, Sato Y, Kagitani-Shimono K, Saito Y, et al. Hematopoietic prostaglandin D synthase and DP1 receptor are selectively upregulated in microglia and astrocytes within senile plaques from human patients and in a mouse model of Alzheimer disease. J Neuropathol Exp Neurol. 2007;66:469–80. doi: 10.1097/01.jnen.0000240472.43038.27. [DOI] [PubMed] [Google Scholar]
  • 116.Ciabattoni G, Porreca E, Di Febbo C, Di Iorio A, Paganelli R, Bucciarelli T, et al. Determinants of platelet activation in Alzheimer’s disease. Neurobiol Aging. 2007;28:336–42. doi: 10.1016/j.neurobiolaging.2005.12.011. [DOI] [PubMed] [Google Scholar]
  • 117.Amtul Z, Uhrig M, Wang L, Rozmahel RF, Beyreuther K. Detrimental effects of arachidonic acid and its metabolites in cellular and mouse models of Alzheimer’s disease: structural insight. Neurobiol Aging. 2012;33:831, e21–31. doi: 10.1016/j.neurobiolaging.2011.07.014. [DOI] [PubMed] [Google Scholar]
  • 118.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]
  • 119.Kanekiyo T, Ban T, Aritake K, Huang ZL, Qu WM, Okazaki I, et al. Lipocalin-type prostaglandin D synthase/beta-trace is a major amyloid beta-chaperone in human cerebrospinal fluid. Proc Natl Acad Sci U S A. 2007;104:6412–7. doi: 10.1073/pnas.0701585104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Lovell MA, Lynn BC, Xiong S, Quinn JF, Kaye J, Markesbery WR. An aberrant protein complex in CSF as a biomarker of Alzheimer disease. Neurology. 2008;70:2212–8. doi: 10.1212/01.wnl.0000312383.39973.88. [DOI] [PubMed] [Google Scholar]
  • 121.Bate C, Kempster S, Williams A. Prostaglandin D2 mediates neuronal damage by amyloid-beta or prions which activates microglial cells. Neuropharmacology. 2006;50:229–37. doi: 10.1016/j.neuropharm.2005.09.008. [DOI] [PubMed] [Google Scholar]
  • 122.Kim KM, Jung BH, Paeng KJ, Kim I, Chung BC. Increased urinary F(2)-isoprostanes levels in the patients with Alzheimer’s disease. Brain Res Bull. 2004;64:47–51. doi: 10.1016/j.brainresbull.2004.04.016. [DOI] [PubMed] [Google Scholar]
  • 123.Yuan L, Lester P. Oxidative stress and age-related neurodegeneration. Boca Raton: CRC Press; 2006. [Google Scholar]
  • 124.Soper JH, Sugiyama S, Herbst-Robinson K, James MJ, Wang X, Trojanowski JQ, et al. Brain-penetrant tetrahydronaphthalene thromboxane A2-prostanoid (TP) receptor antagonists as prototype therapeutics for Alzheimer’s disease. ACS Chem Neurosci. 2012;3:928–40. doi: 10.1021/cn3000795. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Shineman DW, Zhang B, Leight SN, Pratico D, Lee VM. Thromboxane receptor activation mediates isoprostane-induced increases in amyloid pathology in Tg2576 mice. J Neurosci. 2008;28:4785–94. doi: 10.1523/JNEUROSCI.0684-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Soininen H, West C, Robbins J, Niculescu L. Long-term efficacy and safety of celecoxib in Alzheimer’s disease. Dement Geriatr Cogn Disord. 2007;23:8–21. doi: 10.1159/000096588. [DOI] [PubMed] [Google Scholar]
  • 127.Pasqualetti P, Bonomini C, Dal Forno G, Paulon L, Sinforiani E, Marra C, et al. A randomized controlled study on effects of ibuprofen on cognitive progression of Alzheimer’s disease. Aging Clin Exp Res. 2009;21:102–10. doi: 10.1007/BF03325217. [DOI] [PubMed] [Google Scholar]
  • 128.Aisen PS, Davis KL, Berg JD, Schafer K, Campbell K, Thomas RG, et al. A randomized controlled trial of prednisone in Alzheimer’s disease. Alzheimer’s Disease Cooperative Study. Neurology. 2000;54:588–93. doi: 10.1212/wnl.54.3.588. [DOI] [PubMed] [Google Scholar]
  • 129.Reines SA, Block GA, Morris JC, Liu G, Nessly ML, Lines CR, et al. Rofecoxib: no effect on Alzheimer’s disease in a 1-year, randomized, blinded, controlled study. Neurology. 2004;62:66–71. doi: 10.1212/wnl.62.1.66. [DOI] [PubMed] [Google Scholar]
  • 130.Thal LJ, Ferris SH, Kirby L, Block GA, Lines CR, Yuen E, et al. A randomized, double-blind, study of rofecoxib in patients with mild cognitive impairment. Neuropsychopharmacology. 2005;30:1204–15. doi: 10.1038/sj.npp.1300690. [DOI] [PubMed] [Google Scholar]
  • 131.Aisen PS, Schafer KA, Grundman M, Pfeiffer E, Sano M, Davis KL, et al. Effects of rofecoxib or naproxen vs placebo on Alzheimer disease progression: a randomized controlled trial. Jama. 2003;289:2819–26. doi: 10.1001/jama.289.21.2819. [DOI] [PubMed] [Google Scholar]
  • 132.Gomez-Isla T, Blesa R, Boada M, Clarimon J, Del Ser T, Domenech G, et al. A randomized, double-blind, placebo controlled-trial of triflusal in mild cognitive impairment: the TRIMCI study. Alzheimer Dis Assoc Disord. 2008;22:21–9. doi: 10.1097/WAD.0b013e3181611024. [DOI] [PubMed] [Google Scholar]

RESOURCES