U.S. flag

An official website of the United States government

NCBI Bookshelf. A service of the National Library of Medicine, National Institutes of Health.

Kobeissy FH, editor. Brain Neurotrauma: Molecular, Neuropsychological, and Rehabilitation Aspects. Boca Raton (FL): CRC Press/Taylor & Francis; 2015.

Cover of Brain Neurotrauma

Brain Neurotrauma: Molecular, Neuropsychological, and Rehabilitation Aspects.

Show details

Chapter 10Cytoprotective Role of Prostaglandin D2 DP1 Receptor against Neuronal Injury Following Acute Excitotoxicity and Cerebral Ischemia

and .

10.1. INTRODUCTION

Each year, hundreds of thousands of people experience a new or recurrent stroke. This devastating acute neurologic condition is broadly categorized into ischemic (84%) and hemorrhagic (16%) stroke. Stroke is a leading cause of death and long-term disability around the world. Immediately after an ischemic episode, there is a significant rise in glutamate levels at the synapse, leading to a significant increase in intracellular calcium levels. This leads to a cascade of events ending in acute excitotoxicity and cell death. This process also stimulates the activation of various enzymes, notably calcium-dependent cPLA, which liberates arachidonic acid from membrane phospholipids, making it a substrate to cyclooxygenase enzymes. At that point, through a coordinated system, all prostaglandin (PG) syntheses generate specific prostanoids, which are members of fatty acids that include PGD2, PGE2, PGF, PGI2, and TxA2. These PGs exert their actions through specific high-affinity G protein-coupled receptors named DP1-DP2 (PGD2 receptor 1-PGD2 receptor 2), EP1-EP4 (PGE2 receptor 1-4), FP (PGF receptor), IP (PGI2 receptor or prostacyclin receptor), and TP (thromboxane receptor) receptors, respectively. The focus here is on PGD2, as it is the most abundant PG in the brain. Although it has been well studied for its role in sleep modulation, recently, the role of PGD2 and its receptor DP1 has been implicated in acute neurologic conditions. Using genetic and pharmacologic tools, these new results reveal that PGD2 and its receptor pathway have beneficial effects and protect the brain against acute neurologic conditions such as excitotoxicity and ischemic stroke. Additional studies using compounds that selectively activate the DP1 receptor (PGD2 receptor 1) might pave the way for therapeutic testing of this target in clinical settings.

This chapter briefly reviews the pathophysiology of ischemic stroke and treatments proposed in preclinical models, and especially focus on PGD2 and the role of its DP1 receptor in acute ischemic injury. With the understanding that blood flow plays a critical function in ischemic outcomes, a better appreciation of the role of PGD2/PGD2 receptor 1 (DP1) in cerebral blood flow will be presented. Finally, there will be a discussion to review whether the PGD2 DP1 receptor can be used as a target to attenuate brain damage initiated by ischemic stroke or excitotoxicity.

10.2. STROKE PATHOPHYSIOLOGY

Stroke is a medical emergency stemming from acute functional or anatomical neurologic dysfunction caused by a disturbance in the blood supply to discrete areas in the brain. In general, stroke is broken down into two categories: ischemic and hemorrhagic, accounting for approximately 84% and 16% of the total stroke incidents, respectively (Go et al., 2013; Roger et al., 2012). Ischemic stroke is a condition in which the blood supply to a particular region of the brain is permanently or transiently restricted because of the obstruction of a blood vessel by a blood clot or by arterial stenosis, thus depriving the brain cells of the glucose, oxygen, and energy required for their function (Andersen et al., 2009; Chambers et al., 1987; Garcia, 1975; Sutherland et al., 2012). Ischemic stroke is further categorized into thrombotic or embolic stroke. Thrombosis is a condition in which a blood clot is formed at the site of the occlusion, whereas embolism is a condition in which a blood clot is formed in a distant major artery and travels to the occlusion site. Thrombi are formed in the intracranial arteries when the basal lamina is damaged and plaques are formed along the injured site. This process is further facilitated by the activation and aggregation of platelets at the injured site, further facilitating the coagulation cascade. This process by itself can lead to hypoperfusion or occlusion of the blood supply, and certain factors can lead to an increase in shear stress and can dislodge the plaque, which can then lead to occlusion of the blood vessels, causing a significant decrease in the blood flow and leading to an acute ischemic condition. The ischemic episode is also facilitated by atherosclerosis, a condition in which the arterial wall is thickened for various reasons, including the deposition of fatty materials, inflammation, accumulation of macrophages, and formation of multiple thrombi (Aoki et al., 2007; Becker, 1998; Chamorro, 2004; Duvall and Vorchheimer, 2004; Eltzschig and Eckle, 2011; Fenton et al., 1998; Kanematsu et al., 2011; Kargman et al., 1998; Kogure et al., 1996; Mendez et al., 1987). Ultimately, all of this results in reduced blood flow or rupture of the inner arterial wall and the release of the blood clot into the blood stream. This is in contrast to an embolic stroke, in which a blood clot formed at a distant site travels and lodges in a cerebral artery. Some prominent causes of emboli include microemboli breaking away from a plaque in the carotid artery, atrial fibrillation, patent foramen ovale, hypokinetic left ventricle, and vascular or cardiac surgical procedures (Jaillard et al., 1999; Wardlaw et al., 2001; Wolf et al., 1987).

10.3. HYPOXIA, FREE RADICAL DAMAGE, ENERGY DEPLETION, AND NEURONAL DEPOLARIZATION

The brain has a relatively high demand for oxygen and glucose, and similar to the heart, it does not have a high antioxidant system, making it most susceptible to various neurologic conditions, including stroke. As a result, in ischemic/hypoxic conditions, the focal impairment of cerebral blood flow restricts the delivery of vital substrates such as oxygen and glucose. This subsequently imbalances ionic gradients and membrane potentials are lost. Consequently, presynaptic channels are activated and excitatory amino acids such as glutamate are released into the synaptic cleft. Because of energy depletion, the presynaptic reuptake of excitatory amino acids is attenuated, resulting in the accumulation of a significant amount of glutamate in the extracellular space (Ardizzone et al., 2004; Dirnagl et al., 1999; Kemp and McKernan, 2002; Lipton, 1999; Lipton and Rosenberg, 1994; Sutherland et al., 2011; Wang, 2011). As a result, N-methyl-D-aspartate (NMDA) and metabotropic glutamate receptors are activated and an influx of Ca2+ and Na+ causes excitotoxicity and edema in the postsynaptic neurons (Figure 10.1). Excitotoxicity is one of the primary events propagating immediate cell death, whereas edema is one of the major determinants of the final outcome after stroke (Asano et al., 1985; Bhardwaj et al., 2000; Bounds et al., 1981; Choi, 1992; Dirnagl et al., 1999; Lynch and Guttmann, 2002; Macdonald and Stoodley, 1998; Manley et al., 2000; Urushitani et al., 2001).

FIGURE 10.1. Some of the major cellular pathophysiologic cascades associated with ischemic stroke.

FIGURE 10.1

Some of the major cellular pathophysiologic cascades associated with ischemic stroke. Occlusion of the blood vessel leads to cerebral ischemia, which subsequently leads to energy failure and presynaptic neuronal membrane depolarization. This causes massive (more...)

10.4. EXCITOTOXICITY, CALCIUM SIGNALING, AND ACTIVATION OF PROSTANOID SYNTHESIS

Neurons in an ischemic brain usually die by means of necrotic or delayed cell death mechanisms. An important insight into the mechanism of penumbral infarction was revealed by the pioneering work of Dr. Brian Meldrum’s laboratory showing that the preemptive blockade of the glutamate receptor reduced infarct size (Simon et al., 1984). During ischemic stroke, levels of excitatory amino acids, especially glutamate, increase rapidly within the affected area because of their synaptic release from core neurons undergoing ischemic depolarization. Glutamate regulates the activity of various excitatory metabotropic receptors, including the NMDA receptor, which is a Ca2+ membrane channel. Glutamate acts as an NMDA receptor agonist and increases Ca2+ influx to the postsynaptic neurons up to a toxic level, causing immediate injury to these neurons (Lipton, 1999; Smith, 2004). The use of a competitive and/or noncompetitive antagonist of the NMDA receptor can reduce experimental infarct volume to a significant level by antagonizing the effects of glutamate (Lipton, 1999). Based on these preclinical reports, a similar approach was further extended in clinical stroke settings; however, no appreciable beneficial effects were observed—most likely from the limitation for timely application of such drugs after the first symptoms of stroke onset.

An increase in the intracellular levels of Ca2+ also initiates a cascade of cytoplasmic and nuclear events that dictate the pathophysiology of the ailment. Some of these events include activation of proteolytic enzymes that degrade cytoskeletal and extracellular matrix proteins and activation of phospholipase A2—notably cytosolic phospholipase A2 (cPLA), which initiates a proinflammatory cascade (Adams et al., 1996; Bazan, 2005; Bonventre, 1997; Kishimoto et al., 2010; Phillis et al., 2006; Phillis and O’Regan, 2004; Tanaka et al., 2003). PLA2 acts on membrane-bound phospholipids and generates arachidonic acid (AA), which is subsequently converted into hydroxyeicosatetraenoic acid (HETE) and leukotriene through lipoxygenase (LOX) pathways, epoxyeicosatrienoic acid (EETs), and HETE through cytochrome P450 (CYP) pathways, or into prostanoids and thromboxanes through cyclooxygenase (COX) and prostaglandin (PG)/thromboxane synthase pathways (Bazan et al., 2002; Funk, 2001; Khanapure et al., 2007; Kudo and Murakami, 2002; Phillis et al., 2006; Wymann and Schneiter, 2008) (Figure 10.2). Thus, the initial insult after ischemic stroke causes a hostile environment with resilient excitotoxicity, the generation of oxygen- and nitrogen-free radicals and proteolytic enzymes, and the activation of an inflammatory cascade that initiates cellular, molecular, and hemodynamic events converting the center of the damage into an ischemic core. These events destroy brain cells in the ischemic core by lipolysis, proteolysis, disaggregation of microtubules, bioenergetic failure, and the breakdown of ion homeostasis (del Zoppo et al., 2000; Dirnagl et al., 1999; Iadecola and Alexander, 2001; Lo, 2008; Lo et al., 2003). This ischemic core is surrounded by a partially affected area often referred to as the ischemic penumbra, the peri-infarct area, or the area at risk. Either because of incomplete occlusion, hypoperfusion, or partial compensatory blood flow through collateral circulation, the ischemic penumbra has partially preserved energy metabolism (Bang et al., 2011; Frykholm et al., 2005; Liebeskind, 2005; Lo, 2008). This is the salvageable area; however, if left unattended and without treatment, this area can be overtaken by the advancing ischemic core through ongoing excitotoxicity and/or deleterious secondary cascades such as spreading membrane depolarization, edema, postischemic inflammation, and delayed cell death (del Zoppo et al., 2000; Dirnagl et al., 1999; Iadecola and Alexander, 2001; Lo, 2008; Lo et al., 2003). Although the primary cascade of excitotoxicity has been a target to treat stroke, because of the complex pathophysiology of stroke, many of the poststroke neuroprotective efforts are focused on salvaging the “penumbra” by minimizing secondary events.

FIGURE 10.2. Simplified schematic illustration of PG syntheses and their action on their high-affinity specific receptors.

FIGURE 10.2

Simplified schematic illustration of PG syntheses and their action on their high-affinity specific receptors. AAs are generated from membrane phospholipids, and then COX acts on AA to generate intermediate PG endoperoxide H (PGH2). The tissue-specific (more...)

10.5. TREATMENTS AVAILABLE FOR STROKE

Although stroke is one of the leading causes of morbidity and mortality in developed nations, effective therapies are still elusive. Essentially, the only approved pharmacologic therapy against stroke is the use of thrombolytic agents such as tissue plasminogen activator (tPA) (del Zoppo and Koziol, 2007; Yamamoto et al., 1991; Zivin et al., 1985). However, because the use of tPA is highly time-dependent, the most beneficial effect of tPA treatment can only be obtained within ~4.5 hours after the onset of ischemia. If given beyond the stroke-onset therapeutic window of ~4.5 hours, tPA can lead to vascular rupture; thereby, transforming an ischemic stroke into a hemorrhagic stroke, a phenomenon known as hemorrhagic transformation (Deb et al., 2010; Rosell et al., 2008; Wang, 2011; Xing et al., 2011). Consequently, a significant amount of research money and effort has been devoted to actively pursuing cutting-edge research and subsequently translating any findings into clinical trials to elicit treatment options or to using other therapies that can potentially minimize tPA side effects and/or extend the therapeutic window of tPA. Some of these approaches include the use of NMDA receptor antagonists, free radical scavengers, vasodilators, matrix metalloproteinase inhibitors, thrombolytics and antithrombotics, platelet inhibitors, COX inhibitors, and many others (Chan, 2001; Cheng et al., 2006; Clark et al., 2001; Cunningham et al., 2005; Fang et al., 2006; Gerriets et al., 2003; Green and Ashwood, 2005; Grossetete and Rosenberg, 2008; Hinkle and Bowman, 2003; Hurley et al., 2002; Maeda et al.; Morita-Fujimura et al., 2001; Rao and Balachandran, 2002; Saleem et al., 2008; Serteser et al., 2002; Vartiainen et al., 2001; Weinberger, 2006; Wilson and Gelb, 2002; Zhao et al., 2006). Although some of these later compounds have had success at certain stages of the preclinical and clinical trials; unfortunately, none of them have been successful in resolving ischemic brain damage (Green, 2002; Ikonomidou and Turski, 2002). It is clear that there is an urgent need to pursue an effective therapeutic option or a combination of drugs that can target the various steps of the stroke cascade over time.

10.6. AA CASCADE

Essential fatty acids (EFAs), a subset of unsaturated fatty acids, play various physiologic and pathophysiologic roles in neural cells by regulating intracellular signaling. Because of the lack of the necessary enzymes that are required for their synthesis, humans obtain EFAs from external sources and then incorporate them into the phospholipids. In the brain, EFAs are released by the action of PLA2 enzymes and they modulate various physiologic functions, including synaptic function, neuronal toxicity, cerebrovascular tone, neuroinflammation, and oxidative stress (Bazan, 2005; Doré et al., 2003; Kishimoto et al., 2010; Phillis and O’Regan, 2003; Sanchez-Mejia et al., 2008). The PLA2 enzymes are a family of lipases that are categorized into 12 groups based on various factors such as biological activity, substrate specificity, activating factors, and localization (Burke and Dennis, 2009a, 2009b; Kudo and Murakami, 2002). In the brain, AA and docosahexaenoic acid are present in bilipid layers of brain cells and appear to be among the most important fatty acids because of their action in generating more free fatty acids and their biological activity. AA acts as a substrate for at least three well-studied pathways, which are the LOX, CYP, and COX/PG synthase pathways. These pathways are tissue specific, and often cell specific, and the final outcomes are highly dependent on the nature and site of injury.

10.6.1. LOX Pathway

Briefly, in this pathway, AA is converted into leukotrienes by the action of LOX. Leukotrienes act through specific receptors and play important physiologic and pathophysiologic roles. Recently, some selective and effective compounds targeting the LOX pathway have been developed and are being tested to treat asthma and seasonal allergies (Capra et al., 2007; Ribeiro et al., 2006). These pathways are actively being further explored as novel therapeutic targets, and their roles in various ailments are being defined.

10.6.2. CYP Pathway

The AA in this pathway is acted upon by two different enzymes, namely CYP hydroxylase and CYP epoxygenase, leading to two distinct pathways. The CYP hydroxylase enzymes convert AA into HETE. The main metabolite of this pathway is 20-HETE, which is considered to be proinflammatory, and plays an important vascular function (Ishizuka et al., 2008; Roman, 2002). Various metabolites generated in this pathway are being tested for their efficacy in regulating the outcomes of various disease conditions, including stroke (Miyata et al., 2005; Renic et al., 2009; Roman, 2002). The CYP epoxygenase enzymes generate various EET from AA. It has been proposed that EETs are endothelium-derived hyperpolarizing factors and have an anti-inflammatory action in various animal models of diseases (Campbell et al., 1996; Fleming, 2007; Gross et al., 2008; Imig, 2005).

10.6.3. COX/PG Synthase Pathway

COX isoforms COX-1 and COX-2 are the two most studied and recognized oxygenases (Smith et al., 2000). Once cells are activated by trauma, stress, growth factor, cytokines, etc., a 20-carbon unsaturated fatty acid is released from the lipid bilayer of the plasma membrane and is mobilized by different types of phospholipases such as secretory phospholipase A2, cPLA2, and calcium-independent phospholipase A2. The AA generated as a result of PLA2 action is converted into prostanoids by the sequential action of COX and PG synthase enzymes (Figure 10.2). COXs are considered to be proinflammatory, therefore, drugs such as COX inhibitors, aspirin, nonsteroidal anti-inflammatory drugs (NSAIDs), and other compounds that inhibit COX activity are effective drugs for treating pain, inflammation, and cardiovascular disease (Asanuma et al., 2004; Ballou et al., 2000; Grosser et al., 2006; Hoffmann, 2000; Minghetti, 2004; Park et al., 2007; Simmons et al., 2004; Szekely et al., 2004). The COX inhibitors are usually used acutely due to the side effects. Based on their anti-inflammatory effects, COX inhibitors were promoted as promising candidates for treating stroke and other neurologic conditions; however, this enthusiasm ceased when several clinical trials revealed an increased incidence of acute renal failure, myocardial infarction, and thrombotic stroke in patients treated with COX-2 inhibitors (FitzGerald, 2004; Grosser et al., 2006).

10.7. COX AND NEURODEGENERATION

COX enzymes are rate-limiting enzymes associated with the conversion of AA into PGE2, PGD2, PGF2 α, PGI2, and TxA2, collectively known as prostanoids. Two types of COX (COX-1 and COX-2) have been well studied. They are also known as PG endoperoxide H synthases-1 and -2. The two isozymes are quite similar and share ~60% homology in their amino acids. The COX-2 active site has a larger pocket, thus making it more accommodating for using fatty acids (Rouzer and Marnett, 2009; Smith et al., 2000). COX-1 is responsible for normal physiologic or “housekeeping” functions, whereas COX-2 is an inducible isoform involved in inflammation, mitogenesis, and signal transduction (Kam and See, 2000). Although COX-1 is generally considered to be constitutively expressed, depending on which “inducible” PG synthases it is coupled to, it can have effects on various inflammatory and other cellular and organelle pathways.

COX enzymes have catalytic and regulatory subunits and are bound to the endoplasmic reticulum and nuclear membrane. COX-2 immunoreactivity in neurons shows that it is localized to dendritic spines, which are the site of NMDA receptor–mediated neurotransmission (Kaufmann et al., 1996). COX-2 is expressed in the brain at a high basal level selectively in pyramidal neurons of hippocampal and cortical circuits in neurons of the amygdala. COX-2 is an important mediator of inflammation, including neuroinflammation (Feng et al., 1993; Gobbo and O’Mara, 2004). It has been reported that coordinated upregulation of cPLA2 and COX-2 activities contribute to brain damage after cerebral ischemia and neuroinflammation (Bosetti and Weerasinghe, 2003; Sandhya et al., 1998). COX-2 plays an important role in brain injury caused by ischemia (Doré et al., 2003), kainic acid–induced seizures (Baik et al., 1999; Kelley et al., 1999), NMDA toxicity (Hewitt, 2000; Iadecola et al., 2001), Alzheimer disease (Xiang et al., 2002), and Parkinson disease (Teismann et al., 2003), along with other neurologic disorders (Consilvio et al., 2004; Madrigal et al., 2003). The effects of genetic manipulation of COX-2 on focal ischemia have been reported by several groups, including ours (Doré et al., 2003; Iadecola et al., 2001). In COX-2 knockout mice (COX-2-/-), Iadecola and coworkers (Iadecola et al., 2001) found reduced cerebral infarction after middle cerebral artery occlusion (MCAO), whereas in our previous study, we found that overexpression of COX-2 enhanced cerebral infarction (Doré et al., 2003). Consistent with these results, we and others (Doré et al., 2003; Iadecola and Ross, 1997; Nakayama et al., 1998; Nogawa et al., 1997; Resnick et al., 1998) have shown that the use of specific COX-2 inhibitors significantly reduced the infarction in wild-type mice. However, we, for the first time, unveiled that COX-2–specific inhibitors were unable to significantly attenuate the infarction (Doré et al., 2003), notably in conditions in which COX-2 was already overexpressed. These conflicting results suggest that the basal level of COX-1 and other PG synthase enzymes are important factors to consider in addition to COX-2. Moreover, the unique and different roles of various PGs and their receptors in regulation of neuroinflammation and neuronal cell death should not be neglected.

The NSAIDs have been widely used to nonselectively target COX-induced inflammation. Since their discovery (Vane, 1971), NSAIDs have been used to inhibit COX and consequently the synthesis of PGs, and to play a vital role in the physiology and pathophysiology of the cell. Given the antiinflammatory effects of COX inhibitors, these compounds were proposed and successfully tested to be neuroprotective in diverse neurologic conditions. However, this promising arena essentially ceased when several clinical trials revealed adverse side-effects associated with COX inhibitors. For example, detrimental outcomes were observed in Merck’s Adenomatous Polyp PRevention On Vioxx (i.e., APPROVe) study, which resulted in the termination of this clinical trial and withdrawal of the COX-2 inhibitor Vioxx on September 30, 2004, and there was a similar end for Bextra, manufactured and marketed by Pfizer (Davies and Jamali, 2004; Lenzer, 2005; Singh, 2004; Topol, 2004). Essentially, only Celebrex is currently used for various other ailments. This opened a new debate on the pharmacological effects of COX inhibitors and their uses, and the focus of argument shifted toward PGs. Therefore, our laboratory and that of others have been especially interested in the various PG receptors, knowing that the majority of all drugs are targeting G protein–coupled receptors (GPCRs). Therefore, it is imperative to understand the importance of PG syntheses, their relationships, and the role of PGs and their receptors, notably in the cardiovascular and neuronal systems.

10.8. PG SYNTHESES

PGs are bioactive lipids involved in a myriad of diverse and essential homeostatic physiological and pathological functions. PGs are formed by most of the body’s cells and can act as autocrine or paracrine agent, or both. Unlike most of the other hormones, PGs are not stored but are synthesized de novo from AA, and are rapidly processed because of their short half-lives. As mentioned previously in the AA cascade section, PGH2 serves as a common substrate and can be converted into the relatively stable PGs and thromboxanes by the action of specific synthases/isomerases. PGE synthase (cytosolic: cPGES-1; membrane-associated: mPGES-1 and mPGES-2), PG I synthase (PGIS), PG D synthase (hematopoietic-type: H-PGDS, and lipocalin-type: LPGDS), PG F synthase (PGFS), and thromboxane synthase (TxS) into PGE2, PGI2, PGD2, PGF, and TxA, respectively (Figure 10.2). Given the important roles that PGs play, PG synthase inhibitors have been tested in various disease conditions to better understand the role of their respective PGs (Kanekiyo et al., 2007; Murakami and Kudo, 2006; Qu et al., 2006; Santovito et al., 2009; Takemiya et al., 2010).

10.9. PGs AND THEIR HIGH-AFFINITY RECEPTORS

Each PG binds with different affinities to various GPCR designated as DP1-DP2 (PGD2 receptor 1, PGD2 receptor 2), EP1-EP4 (PGE2 receptor 1-4), FP (PGF2 α receptor), IP (prostacyclin receptor), and TP (thromboxane receptor) for PGD2, PGE2, PGF2 a, PGI2, and TxA2, respectively, and exerts its physiologic and pathophysiologic effects through these receptors (Breyer et al., 2001; Coleman et al., 1995; Jones et al., 2009; Narumiya, 2009; Narumiya et al., 1999; Sharif et al., 1998; Woodward et al., 2011). Although the literature associates these prostanoids as essentially binding only with their receptor family members, in most cases, these receptors cross-react with other receptors. However, this has not been discussed and elaborated on well enough in literature.

In general, EP2, EP4, DP1, and IP are known to cause an increase of intracellular cyclic adenosine 3’,5’-monophosphate (cAMP), whereas EP1, FP, and TP cause an increase of intracellular free Ca2+ (Wright et al., 2001 and see Mohan et al. for a comprehensive review [Mohan et al., 2012]). These receptors share approximately 20% to 30% of their sequences with each other. We and others have previosuly exposed the “genealogy” of these receptors, which further supports the proposed classification of these receptors and their downstream cell signaling (Hirai et al., 2001; Mohan et al., 2012; Toh et al., 1995). However, caution should be used when impying it as a general rule because there are always exceptions to such rules. Most of the prostanoid receptors interact with a wide range of intracellular signaling pathways and facilitate receptor activation and functions. PGs play significant physiologic and pathophysiologic roles in circulation, vascular regulation, hypertension, sleep regulation, temperature regulation, parturition, ocular pressure, skin inflammation, airway maintenance, bowel syndromes, vasodilation, vasoconstriction, excitotoxicity, and stroke (Chemtob et al., 1990a, 1990b, 1995; Faraci and Heistad, 1998; Wright et al., 2001). In general, it has been shown that the receptors that activate the cAMP pathway lead to better outcomes after ischemic stroke, whereas those that activate the Ca2+ pathway lead to worse outcomes. However, this general perception is not true in all conditions, and exceptions have also been reported. Because PGD2 is the most abundant and often neglected prostanoid, we will briefly expose additional focused information about this prostanoid and its main receptor, DP1. The other PGD2 receptor was named DP2 once it was found that PGD2 has some affinity toward this orphan receptor, previously known as CRTH2 receptor (Nagata et al., 1999a). Moreover, this receptor did not evolve from the same phylogenetic tree as the other PG receptors (Mohan et al., 2012).

10.10. PGD2 AND ITS METABOLISM

PGD2 is generated from PGH2 by the action of two distinct PGD synthases: L-PGDS and H-PGDS. PGD2 was first identified in the late 1960s (Granstrom et al., 1968; Hayashi and Tanouchi, 1973), and a later study provided evidence that this PG is a potent inhibitor of platelet aggregation (Smith et al., 1974). Since then, the role of PGD2 has been investigated in a wide range of pharmacological activities. The diverse activities of PGD2 in regulating local blood flow, as a bronchial airway caliber, in leukocyte function, and so on, are mediated by its high-affinity interactions with its GPCRs. However, the half-life of PGD2 is very short (about 1.5 minutes in blood); therefore, under certain conditions, it is rapidly metabolized. The main products that have been detected in vivo are Δ12PGJ2 and 9α11βPGF2 (Heinemann et al., 2003; Sandig et al., 2006b). Other putative metabolites are 13,14-dihydro-15-keto-PGD2 16, Δ12PGD2, 15-deoxy-Δ12,14PGD2, and 15-deoxy-Δ12,14PGJ2 (Gazi et al., 2005; Monneret et al., 2002) (Figure 10.3). Although the exact mechanism of PGD2 metabolism is still elusive, it has been postulated that 13,14-dihydro-15-keto-PGD2, Δ12PGD2, and 15-deoxy-Δ12,14PGJ2 are formed locally at the site of inflammation. It is important to note that all of these metabolites confine their activity to DP2 receptor, and their activity on the DP1 receptor has not been reported in the literature. Other metabolites of PGD2 such as Δ12PGJ2 and 15-deoxy- Δ12,13PGJ2 have effects on the resolution of inflammation by inhibiting cytokine production and the induction of apoptosis. These effects are mediated by the peroxisome proliferator-activated receptor-γ (PPARγ)-dependent or PPARγ-independent mechanisms (Forman et al., 1995; Rossi et al., 2000). Previous work from our laboratory also shows that J series PG (dPGJ2 and 15-deoxy-Δ12,14PGJ2) treatments upregulate heme oxygenase and attenuate neuroinflammation (Zhuang et al., 2003a, 2003b).

FIGURE 10.3. PGD2 metabolism and its effects.

FIGURE 10.3

PGD2 metabolism and its effects. PGD2 can act as autocrine or paracrine agent, or both, and exerts its effects mainly through its GPCR DP1 receptor, although some reports on peripheral systems suggest the action of PGD2 through CRTH2 (or DP2) receptors. (more...)

10.11. PGD2 AND DP RECEPTORS

PGD2 has peripheral and central physiologic effects. Some of the peripheral effects of PGD2 include vasodilatation, bronchoconstriction, inhibition of platelet aggregation, glycogenolysis, vasoconstriction, allergic reaction mediation, and intraocular pressure reduction (Angeli et al., 2004; Casteleijn et al., 1988; Darius et al., 1994; Matsugi et al., 1995; Matsuoka et al., 2000; Narumiya and Toda, 1985; Sturzebecher et al., 1989; Whittle et al., 1983). Among all PGs, PGD2 is the most abundant in the brain. In the central nervous system, under normal physiologic conditions, PGD2 contributes to sleep induction, modulation of body temperature, olfactory function, hormone release, nociception, and neuromodulation (Eguchi et al., 1999; Gelir et al., 2005; Hayaishi, 2002; Hayaishi and Urade, 2002; Mizoguchi et al., 2001; Urade and Hayaishi, 1999). PGD2 levels are significantly increased under pathologic conditions (Hata and Breyer, 2004; Hatoum et al., 2005; Luster and Tager, 2004; Naffah-Mazzacoratti et al., 1995; Seregi et al., 1990; Tegtmeier et al., 1990); however, whether this increase is harmful or beneficial is controversial. PGD2 protects astrocyte cultures by increasing the production of neurotrophins (Angeli et al., 2004). It has also been shown that PGD2 acts as an inflammatory mediator of allergic reactions, such as asthma and conjunctivitis, which are initiated by the immunoglobulin E–mediated type 1 response. Moreover, there are reports showing some deleterious effects of PGD2, mainly regulated through the DP2 receptor or through PPARγ pathways (Almishri et al., 2005; Gazi et al., 2005; Kagawa et al., 2011; Nagata and Hirai, 2003; Nagata et al., 1999a).

10.12. DP1 RECEPTOR

PGD2 exerts its biological action by binding and activating through distinct and specific cell receptors. Recent findings suggest that there are two types of the receptor for PGD2 : the first is known as the DP1 or DP receptor and the second has recently been discovered and is known as chemoattractant receptor homologous molecule expressed on T helper type 2 cells (CRTH2). CRTH2 is also now known as the DP2 receptor; however, it has a different “genealogy” and a different homology compared with other prostanoid receptors (Abe et al., 1999; Hata et al., 2003; Mohan et al., 2012; Toh et al., 1995). CRTH2 signaling is independent of PGD2 production (Bohm et al., 2004; Sandig et al., 2006a) and has different biological functions and cellular distribution than the DP1 receptor (Hirai et al., 2001; Sawyer et al., 2002). It is located on the 11q12-q13.3, 19A, and 1q43 chromosomes in the human, mouse, and rat, respectively, and is encoded by the genes PTGDR2, Gpr44, and Ptgdr2 in the human, mouse, and rat, respectively. It contains 382 to 403 amino acids (Abe et al., 1999; Hata et al., 2003; Nagata et al., 1999b; Shichijo et al., 2003) and its structure closely resembles that of the formyl peptide receptor (formyl-Met-Leu-Phe) and the B leukotriene receptor. It is expressed on Th2-type lymphocytes, basophils, and eosinophils. DP2 is widely expressed in the brain, heart, stomach, ovary, testes, and skin, and it is biologically involved in eosinophil activation, chemotaxis, degranulation, and cytokine production.

Primarily, the anti-inflammatory and vasoregulatory properties of PGD2 are associated with DP1 receptors; although, the proinflammatory effects of DP1 have also been reported (Angeli et al., 2004; Hammad et al., 2003; Matsuoka et al., 2000; Spik et al., 2005). DP1 is a GPCR coupled with Gαs and is located on the 14q22.1, 14B, and 15p14 chromosomes in the human, mouse, and rat, respectively. The DP1 receptor (encoded by the gene PTGDR) was first cloned in 1994, and it contains 357 to 359 amino acids (Boie et al., 1995; Hirata et al., 1994; Ishikawa et al., 1996; Wright et al., 1999). On the phylogenetic tree, this receptor shows significant sequence homology with IP and EP2 receptors of PGI2 and PGE2, respectively, and is distributed throughout the body, including in ciliary epithelial cells, the retinal choroid, the iris, eosinophils, platelets, the colon, and the brain. Physiologically, DP1 receptors are involved in inhibiting platelet aggregation and histamine release, and in the relaxation of myometrium and smooth muscles. Furthermore, any alteration in DP1 receptor results in an asthmatic response, reduced nicotinic-induced vasodilation, and sleep alteration. The human DP1 receptor binds strongly to PGD2, followed by PGE2, PGF2, and PGI2, and least to thromboxane A2. These receptors are expressed in various tissues and cells, including bone marrow, small intestine, lung, stomach, mast cells, and brain, and are involved in various pathophysiological functions such as platelet activation, vasodilation (niacin flush), sleep regulation, and immune cell activation. The DP1 receptor stimulates adenylyl cyclase, leading to increased levels of cAMP and decreased platelet aggregation (Hata and Breyer, 2004). Recently, several selective agonists such as BW245C, AS702224, and TS-022, and antagonists such as MK-0524 (or Laropiprant) and BWA868C of the DP1 receptor are being used in various animal models (Cheng et al., 2006; Crider et al., 1999; Hirano et al., 2007; Kabashima and Narumiya, 2003; Van Hecken et al., 2007). The use of these pharmacologic agents and genetic manipulations is extremely helpful in providing insight into the mechanism of action of PGD2 and the DP1 receptor (Table 10.1). The in vivo role of DP1 in the periphery has been well investigated (Angeli et al., 2004; Hata and Breyer, 2004; Koch et al., 2005), whereas until recently, its role in the brain was limited to studies related to sleep and eye movement (Campbell and Feinberg, 1996; Hayaishi and Urade, 2002; Obal and Krueger, 2003; Urade and Hayaishi, 1999).

TABLE 10.1

TABLE 10.1

List of DP1 Receptor Agonists and Antagonists That Have Been Tested in Various Preclinical and Clinical Studies

10.13. THE ROLE OF PGD2 AND DP1 RECEPTOR IN STROKE

PGD2 has been studied extensively for its role in sleep-awake and thermoregulation studies. However, its importance in regulating brain injuries has recently been recognized. Based on the abundance of PGD2 in the brain, we wanted to determine the role of PGD2 and its DP1 receptor in ischemic stroke and excitotoxicity. Our data suggest that genetic deletion of DP1 receptors make mice more susceptible to ischemic brain injury (Saleem et al., 2007). There are several animal models reported to induce cerebral ischemia. In our studies, we used the filament model of transient cerebral ischemia. To induce cerebral ischemia or stroke, mice were anesthetized and a nylon monofilament coated with silicon was inserted into the internal carotid artery and forwarded up to the origin of the middle cerebral artery. With the help of laser Doppler flowmetry, cerebral blood flow was monitored. MCAO by the filament results in a significant (>80%) decrease in cerebral blood flow, confirming the occlusion. After the occlusion, the incision was temporarily sealed and the mice were placed in humidity- and temperature-regulated recovery units. To achieve the reperfusion, the mice were anesthetized, the incision was opened, and the nylon filament was retracted, which resulted in recirculation of the blood. In our study, we found that the neurologic deficit and brain infarction was significantly higher in DP1-/- mice compared with wild-type mice. To confirm that this was through the direct effect of the DP1 receptor, we thoroughly investigated the cerebral vascular anatomy of wild-type and DP1-/- mice. We found that the effect observed in our study was not an artifact from a difference in vascular anatomy.

Thereafter, in a follow-up study, we tested the effect of pharmacologic activation of the DP1 receptor by using a selective DP1 receptor agonist. We found that NMDA-induced excitotoxicity and cerebral ischemic brain damage was significantly attenuated by the DP1 receptor-selective agonist BW245C (Ahmad et al., 2010). In this study, we first showed that intrastriatal microinjection of NMDA resulted in significant brain damage that was significantly aggravated in DP1-/- mice, whereas the DP1 receptor-selective agonist BW245C had a dose-dependent neuroprotective effect. Moreover, we also showed that the minimum effective dose (10 nmol) observed in the NMDA-induced toxicity paradigm was also significantly efficient in minimizing MCAO-induced brain damage. Similarly, the effect of the DP1 agonist BW245C was tested in an in vitro model of ischemia by using the dispersed neuronal culture and hippocampal slice cultures from wild-type mice (Liang et al., 2005). In that study, it was found that the DP1 agonist BW245C attenuated the glutamate-induced cell death and oxygen–glucose deprivation induced hippocampal slice culture degeneration. Recently, in a report investigating a new compound, GIF-0173, it was found that this compound had beneficial effects against cerebral ischemia by regulating the DP1 receptor (Thura et al., 2009). In an animal model of neonatal hypoxic-ischemic encephalopathy (HIE), one of the leading causes of acute mortality and chronic disability in infants and children, a group led by Dr. Masako Taniike found that PGD2 protects brain against HIE (Taniguchi et al., 2007). Furthermore, they show that the level of PGD2 is significantly upregulated in the HIE mouse brain. Their data suggest that the beneficial effect of the DP1 receptor could be from its role in endothelial cells and the vasculature.

As we have mentioned in the preceding sections, some of the PG receptors activate calcium signaling, whereas some activate the cAMP pathway. Activation of the cAMP pathway has been reported to have a protective effect and restores normal cellular process (Hanson et al., 1998; Ryde and Greene, 1988; Walton et al., 1999; Walton and Dragunow, 2000). Some of the direct intracellular targets for cAMP are protein kinase A (PKA), the exchange protein activated by cAMP (Epac), and cyclic nucleotide-gated ion channels (Bos, 2006; Craven and Zagotta, 2006; de Rooij et al., 1998; Murray and Shewan, 2008). However, cAMP-mediated activation of PKA is the most widely studied aspect of cAMP signaling (Murray, 2008; Tasken and Aandahl, 2004). There are numerous studies that propose that the activation of cAMP/PKA has a neuroprotective effect through the activation of various transcription factors, including cAMP-responsive element-binding protein (Mantamadiotis et al., 2002; Walton et al., 1999; Walton and Dragunow, 2000). Similarly, it has been reported that DP1 activation significantly augments cAMP levels, which then activate various pathways leading to neuroprotection. Moreover, PGD2 is substantially produced in blood, and DP1 receptors are significantly present in vascular lining and endothelial cells. Previous reports suggest that DP1 activation by BW245C regulates hemodynamics in rats (Koch et al., 2005). Moreover, DP1 receptor activation has also been implicated in a clinical condition known as niacin flush, where use of niacin increases PGD2, which then acts on DP1 and induces vasodilation, resulting in hot flushes on a patient’s skin. Activation of DP1 by the selective agonist BW245C decreases inflammation in murine models of dermatitis (Angeli et al., 2004). Interestingly, in clinical settings, BW245C was found to increase intraocular pressure (Shah et al., 1984). To further extend our findings on the role of the PGD2 DP1 receptor in stroke, we tested the effect of the DP1 receptor agonist BW245C on cerebral blood flow before, during, and after ischemic stroke. It was astonishing that this compound was able to improve cerebral blood flow during and after stroke. Moreover, because imbalance hemostasis is also an important trigger of stroke onset or propagation, we tested the effect of BW245C on hemostasis (tail bleeding time and coagulation). To our amazement, we found that BW245C increased tail bleeding in vivo, and inhibited ex vivo blood coagulation. These novel works have been presented at various international scientific meetings and conferences (Ahmad et al., 2013a, 2013b).

Although the potential neuroprotective effect of the DP1 receptor is being reported, the signaling cascade leading to this neuroprotective effect is still unclear. Thus far, by using DP1 selective agents, various researchers are able to show that cAMP/PKA is one of the important pathways thorough which DP1 exerts its effect (Liang et al., 2005). This study also shows that PKA inhibitors were able to inhibit PGD2 levels as well as PGD2 -mediated neuroprotection. In another study that elegantly used DP1-/- mice, it was shown that wild-type mice, after hypoxic ischemic brain injury, had better angiogenesis compared with DP1-/- mice. From this, the authors suggest that the DP1 receptor–mediated beneficial effect after hypoxic ischemic brain injury could possibly be through its role in the vasculature. Based on these reports and reports from our laboratory, it is evident that activation of the PGD2 DP1 receptor activates the cAMP/PKA pathway, resulting in the activation of transcription factor(s), cerebral blood flow improvement, and maintenance of hemostasis; however, more experiments and insights are required to fully exploit the role of DP1 in stroke. Therefore, more targeted preclinical studies are required to translate the potential of this treatment in clinical stroke.

10.14. CONCLUSION

The accumulating data thus far show that PGD2 is substantially produced in the brain, blood, and vascular linings and that the DP1 receptor is expressed in endothelial and neural cells. Under acute or chronic neurodegeneration, this receptor has been shown to have beneficial effects that are achieved by regulating the downstream signaling cascade of cAMP or through regulating physiologic pathways such as blood flow and hemostasis. Although these are preliminary studies, they nevertheless propose the potential of the DP1 receptor as a novel therapeutic target to rescue the brain from neurodegeneration. The availability of selective ligands further facilitates this process and additional rigorous testing in comorbid conditions may pave the way for this target to be tested in clinical settings.

ACKNOWLEDGMENTS

This work was supported by grants from the American Heart Association (0830172N; to A.S.A.) and NIH (R01 NS046400-01; to S.D.). The authors would like to thank Rebecca Astrom for editing the manuscript, and all lab members for constructive feedback on the chapter.

REFERENCES

  1. Abe H, Takeshita T, Nagata K, Arita T, Endo Y, Fujita T. et al. Molecular cloning, chromosome mapping and characterization of the mouse CRTH2 gene, a putative member of the leukocyte chemoattractant receptor family. Gene. 1999;227:71–7. [PubMed: 9931443]
  2. Adams J, Collaco-Moraes Y, de Belleroche J. Cyclooxygenase-2 induction in cerebral cortex: An intracellular response to synaptic excitation. J Neurochem. 1996;66:6–13. [PubMed: 8522990]
  3. Ahmad A.S, Ahmad M, Maruyama T, Narumiya S, Dore S. Prostaglandin D2 DP1 receptor is beneficial in ischemic stroke and in acute exicitotoxicity in young and old mice. Age (Dordr). 2010;32:271–82. [PMC free article: PMC2926852] [PubMed: 20640551]
  4. Ahmad A.S, Maruyama T, Narumiya S, Dore S. Correlation between prostaglandin D2 DP1 receptor effects on cerebral blood flow and stroke outcomes. Stroke. 2013a;44 International Stroke Conference Poster Abstracts.
  5. Ahmad A.S, Narumiya S, Dore S. Cerebral blood flow is a major contributing factor towards the beneficial effects of PGD2 DP1 activation. Society for Neuroscience. 2013b 2013 Annual meeting.
  6. Almishri W, Cossette C, Rokach J, Martin J.G, Hamid Q, Powell W.S. Effects of prostaglandin D2, 15-deoxy-Delta12,14-prostaglandin J2, and selective DP1 and DP2 receptor agonists on pulmonary infiltration of eosinophils in Brown Norway rats. J Pharmacol Exp Ther. 2005;313:64–9. [PubMed: 15590767]
  7. Andersen K.K, Olsen T.S, Dehlendorff C, Kammersgaard L.P. Hemorrhagic and ischemic strokes compared: Stroke severity, mortality, and risk ractors. Stroke. 2009;40:2068–2072. [PubMed: 19359645]
  8. Angeli V, Staumont D, Charbonnier A.S, Hammad H, Gosset P, Pichavant M. et al. Activation of the D prostanoid receptor 1 regulates immune and skin allergic responses. J Immunol. 2004;172:3822–9. [PubMed: 15004188]
  9. Aoki T, Kataoka H, Morimoto M, Nozaki K, Hashimoto N. Macrophage-derived matrix metalloproteinase-2 and -9 promote the progression of cerebral aneurysms in rats. Stroke. 2007;38:162–169. [PubMed: 17122420]
  10. Arai I, Takaoka A, Hashimoto Y, Honma Y, Koizumi C, Futaki N. et al. Effects of TS-022, a newly developed prostanoid DP1 receptor agonist, on experimental pruritus, cutaneous barrier disruptions and atopic dermatitis in mice. Eur J Pharmacol. 2007;556:207–14. [PubMed: 17141215]
  11. Ardizzone T.D, Lu A, Wagner K.R, Tang Y, Ran R, Sharp F.R. Glutamate receptor blockade attenuates glucose hypermetabolism in perihematomal brain after experimental intracerebral hemorrhage in rat. Stroke. 2004;35:2587–91. [PubMed: 15375303]
  12. Asano T, Gotoh O, Koide T, Takakura K. Ischemic brain edema following occlusion of the middle cerebral artery in the rat. II: Alteration of the eicosanoid synthesis profile of brain microvessels. Stroke. 1985;16:110–3. [PubMed: 3966253]
  13. Asanuma M, Miyazaki I, Ogawa N. Neuroprotective effects of nonsteroidal anti-inflammatory drugs on neurodegenerative diseases. Curr Pharm Des. 2004;10:695–700. [PubMed: 14965332]
  14. Baik E.J, Kim E.J, Lee S.H, Moon C. Cyclooxygenase-2 selective inhibitors aggravate kainic acid induced seizure and neuronal cell death in the hippocampus. Brain Res. 1999;843:118–29. [PubMed: 10528118]
  15. Ballou L.R, Botting R.M, Goorha S, Zhang J, Vane J.R. Nociception in cyclooxygenase isozyme-deficient mice. Proc Natl Acad Sci U S A. 2000;97:10272–6. [PMC free article: PMC27868] [PubMed: 10954756]
  16. Bang O.Y, Saver J.L, Kim S.J, Kim G.M, Chung C.S, Ovbiagele B. et al. Collateral flow predicts response to endovascular therapy for acute ischemic stroke. Stroke. 2011;42:693–9. [PMC free article: PMC3051344] [PubMed: 21233472]
  17. Bazan N.G. Lipid signaling in neural plasticity, brain repair, and neuroprotection. Mol Neurobiol. 2005;32:89–103. [PubMed: 16077186]
  18. Bazan N.G, Colangelo V, Lukiw W.J. Prostaglandins and other lipid mediators in Alzheimer’s disease. Prostaglandins Other Lipid Mediat. 2002;68–69:197–210. [PubMed: 12432919]
  19. Becker K.J. Inflammation and acute stroke. Curr Opin Neurol. 1998;11:45–9. [PubMed: 9484616]
  20. Bhardwaj A, Harukuni I, Murphy S.J, Alkayed N.J, Crain B.J, Koehler R.C. et al. Hypertonic saline worsens infarct volume after transient focal ischemia in rats. Stroke. 2000;31:1694–701. [PubMed: 10884475]
  21. Bohm E, Sturm G.J, Weiglhofer I, Sandig H, Shichijo M, McNamee A. et al. 11-Dehydro-thromboxane B2, a stable thromboxane metabolite, is a full agonist of chemoattractant receptor-homologous molecule expressed on TH2 cells (CRTH2) in human eosinophils and basophils. J Biol Chem. 2004;279:7663–70. [PubMed: 14668348]
  22. Boie Y, Sawyer N, Slipetz D.M, Metters K.M, Abramovitz M. Molecular cloning and characterization of the human prostanoid DP receptor. J Biol Chem. 1995;270:18910–6. [PubMed: 7642548]
  23. Bonventre J.V. Roles of phospholipases A2 in brain cell and tissue injury associated with ischemia and excitotoxicity. J Lipid Mediat Cell Signal. 1997;17:71–9. [PubMed: 9302656]
  24. Bos J.L. Epac proteins: Multi-purpose cAMP targets. Trends Biochem Sci. 2006;31:680–6. [PubMed: 17084085]
  25. Bosetti F, Weerasinghe G.R. The expression of brain cyclooxygenase-2 is down-regulated in the cytosolic phospholipase A2 knockout mouse. J Neurochem. 2003;87:1471–7. [PubMed: 14713302]
  26. Bounds J.V, Wiebers D.O, Whisnant J.P, Okazaki H. Mechanisms and timing of deaths from cerebral infarction. Stroke. 1981;12:474–7. [PubMed: 7314170]
  27. Bregar U, Jug B, Keber I, Cevc M, Sebestjen M. Extended-release niacin/laropiprant improves endothelial function in patients after myocardial infarction. Heart and Vessels. 2013:1–7. [PubMed: 23712600]
  28. Breyer R.M, Bagdassarian C.K, Myers S.A, Breyer M.D. Prostanoid receptors: Subtypes and signaling. Annu Rev Pharmacol Toxicol. 2001;41:661–90. [PubMed: 11264472]
  29. Burke J.E, Dennis E.A. Phospholipase A2 biochemistry. Cardiovasc Drugs Ther. 2009a;23:49–59. [PMC free article: PMC2823292] [PubMed: 18931897]
  30. Burke J.E, Dennis E.A. Phospholipase A2 structure/function, mechanism, and signaling. J Lipid Res 50 Suppl. 2009b:S237–S242. [PMC free article: PMC2674709] [PubMed: 19011112]
  31. Campbell I.G, Feinberg I. Noncompetitive NMDA channel blockade during waking intensely stimulates NREM delta. J Pharmacol Exp Ther. 1996;276:737–42. [PubMed: 8632344]
  32. Campbell W.B, Gebremedhin D, Pratt P.F, Harder D.R. Identification of epoxyeicosatrienoic acids as endothelium-derived hyperpolarizing factors. Circ Res. 1996;78:415–23. [PubMed: 8593700]
  33. Capra V, Thompson M.D, Sala A, Cole D.E, Folco G, Rovati G.E. Cysteinyl-leukotrienes and their receptors in asthma and other inflammatory diseases: Critical update and emerging trends. Med Res Rev. 2007;27:469–527. [PubMed: 16894531]
  34. Casteleijn E, Kuiper J, Van Rooij H.C, Kamps J.A, Koster J.F, Van Berkel T.J. Prostaglandin D2 mediates the stimulation of glycogenolysis in the liver by phorbol ester. Biochem J. 1988;250:77–80. [PMC free article: PMC1148817] [PubMed: 3162673]
  35. Chambers B.R, Norris J.W, Shurvell B.L, Hachinski V.C. Prognosis of acute stroke. Neurology. 1987;37:221–5. [PubMed: 3808302]
  36. Chamorro A. Role of inflammation in stroke and atherothrombosis. Cerebrovasc Dis 17 Suppl. 2004;3:1–5. [PubMed: 14730251]
  37. Chan P.H. Reactive oxygen radicals in signaling and damage in the ischemic brain. J Cereb Blood Flow Metab. 2001;21:2–14. [PubMed: 11149664]
  38. Chemtob S, Beharry K, Rex J, Varma D.R, Aranda J.V. Changes in cerebrovascular prostaglandins and thromboxane as a function of systemic blood pressure. Cerebral blood flow autoregulation of the newborn. Circ Res. 1990a;67:674–82. [PubMed: 2397575]
  39. Chemtob S, Beharry K, Rex J, Varma D.R, Aranda J.V. Prostanoids determine the range of cerebral blood flow autoregulation of newborn piglets. Stroke. 1990b;21:777–84. [PubMed: 2339458]
  40. Chemtob S, Hardy P, Abran D, Li D.Y, Peri K, Cuzzani O, Varma D.R. Peroxide-cyclooxygenase interactions in postasphyxial changes in retinal and choroidal hemodynamics. J Appl Physiol. 1995;78:2039–46. [PubMed: 7665397]
  41. Cheng K, Wu T.J, Wu K.K, Sturino C, Metters K, Gottesdiener K. et al. Antagonism of the prostaglandin D2 receptor 1 suppresses nicotinic acid-induced vasodilation in mice and humans. Proc Natl Acad Sci U S A. 2006;103:6682–7. [PMC free article: PMC1458941] [PubMed: 16617107]
  42. Choi D.W. Excitotoxic cell death. J Neurobiol. 1992;23:1261–76. [PubMed: 1361523]
  43. Clark W.M, Rinker L.G, Lessov N.S, Lowery S.L, Cipolla M.J. Efficacy of antioxidant therapies in transient focal ischemia in mice. Stroke. 2001;32:1000–4. [PubMed: 11283403]
  44. Coleman R.A, Eglen R.M, Jones R.L, Narumiya S, Shimizu T, Smith W.L. et al. Prostanoid and leukotriene receptors: A progress report from the IUPHAR working parties on classification and nomenclature. Adv Prostaglandin Thromboxane Leukot Res. 1995;23:283–5. [PubMed: 7732854]
  45. Consilvio C, Vincent A.M, Feldman E.L. Neuroinflammation, COX-2, and ALS—A dual role? Exp Neurol. 2004;187:1–10. [PubMed: 15081582]
  46. Craven K.B, Zagotta W.N. CNG and HCN channels: Two peas, one pod. Annu Rev Physiol. 2006;68:375–401. [PubMed: 16460277]
  47. Crider J.Y, Griffin B.W, Sharif N.A. Prostaglandin DP receptors positively coupled to adenylyl cyclase in embryonic bovine tracheal (EBTr) cells: Pharmacological characterization using agonists and antagonists. Br J Pharmacol. 1999;127:204–10. [PMC free article: PMC1565977] [PubMed: 10369474]
  48. Cunningham L.A, Wetzel M, Rosenberg G.A. Multiple roles for MMPs and TIMPs in cerebral ischemia. Glia. 2005;50:329–39. [PubMed: 15846802]
  49. Darius H, Michael-Hepp J, Thierauch K.H, Fisch A. Inhibition of human platelets and polymorphonuclear neutrophils by the potent and metabolically stable prostaglandin D2 analog ZK 118.182. Eur J Pharmacol. 1994;258:207–13. [PubMed: 7522176]
  50. Davies N.M, Jamali F. COX-2 selective inhibitors cardiac toxicity: Getting to the heart of the matter. J Pharm Pharm Sci. 2004;7:332–6. [PubMed: 15576013]
  51. de Rooij J, Zwartkruis F.J, Verheijen M.H, Cool R.H, Nijman S.M, Wittinghofer A. et al. Epac is a Rap1 guanine-nucleotide-exchange factor directly activated by cyclic AMP. Nature. 1998;396:474–7. [PubMed: 9853756]
  52. Deb P, Sharma S, Hassan K.M. Pathophysiologic mechanisms of acute ischemic stroke: An overview with emphasis on therapeutic significance beyond thrombolysis. Pathophysiology. 2010;17:197–218. [PubMed: 20074922]
  53. del Zoppo G, Ginis I, Hallenbeck J.M, Iadecola C, Wang X, Feuerstein G.Z. Inflammation and stroke: Putative role for cytokines, adhesion molecules and iNOS in brain response to ischemia. Brain Pathol. 2000;10:95–112. [PMC free article: PMC8098633] [PubMed: 10668900]
  54. del Zoppo G.J, Koziol J.A. Recanalization and stroke outcome. Circulation. 2007;115:2602–5. [PubMed: 17515478]
  55. Dirnagl U, Iadecola C, Moskowitz M.A. Pathobiology of ischaemic stroke: An integrated view. Trends Neurosci. 1999;22:391–7. [PubMed: 10441299]
  56. Dishy V, Liu F, Ebel D.L, Atiee G.J, Royalty J, Reilley S. et al. Effects of aspirin when added to the prostaglandin D2 receptor antagonist laropiprant on niacin-induced flushing symptoms. J Clin Pharmacol. 2009;49:416–22. [PubMed: 19246721]
  57. Doré S, Otsuka T, Mito T, Sugo N, Hand T, Wu L. et al. Neuronal overexpression of cyclooxygenase-2 increases cerebral infarction. Ann Neurol. 2003;54:155–62. [PubMed: 12891667]
  58. Duvall W.L, Vorchheimer D.A. Multi-bed vascular disease and atherothrombosis: Scope of the problem. J Thromb Thrombolysis. 2004;17:51–61. [PubMed: 15277788]
  59. Eguchi N, Minami T, Shirafuji N, Kanaoka Y, Tanaka T, Nagata A. et al. Lack of tactile pain (allodynia) in lipocalin-type prostaglandin D synthase-deficient mice. Proc Natl Acad Sci U S A. 1999;96:726–30. [PMC free article: PMC15204] [PubMed: 9892701]
  60. Eltzschig H.K, Eckle T. Ischemia and reperfusion—From mechanism to translation. Nat Med. 2011;17:1391–401. [PMC free article: PMC3886192] [PubMed: 22064429]
  61. Fang Y.C, Wu J.S, Chen J.J, Cheung W.M, Tseng P.H, Tam K.B. et al. Induction of prostacyclin/PGI2 synthase expression after cerebral ischemia-reperfusion. J Cereb Blood Flow Metab. 2006;26:491–501. [PubMed: 16094316]
  62. Faraci F.M, Heistad D.D. Regulation of the cerebral circulation: Role of endothelium and potassium channels. Physiol Rev. 1998;78:53–97. [PubMed: 9457169]
  63. Feng L, Sun W, Xia Y, Tang W.W, Chanmugam P, Soyoola E. et al. Cloning two isoforms of rat cyclooxygenase: Differential regulation of their expression. Arch Biochem Biophys. 1993;307:361–8. [PubMed: 8274023]
  64. Fenton J.n, Ofosu F.A, Brezniak D.V, Hassouna H.I. Thrombin and antithrombotics [review] Semin Thromb Hemost. 1998;24:87–91. [PubMed: 9579630]
  65. Fitzgerald G.A. COX-2 and beyond: Approaches to prostaglandin inhibition in human disease. Nat Rev Drug Discov. 2003;2:879–90. [PubMed: 14668809]
  66. Fitzgerald G.A. Coxibs and cardiovascular disease. N Engl J Med. 2004;351:1709–11. [PubMed: 15470192]
  67. Fleming I. DiscrEET regulators of homeostasis: Epoxyeicosatrienoic acids, cytochrome P450 epoxygenases and vascular inflammation. Trends Pharmacol Sci. 2007;28:448–52. [PubMed: 17764757]
  68. Forman B.M, Tontonoz P, Chen J, Brun R.P, Spiegelman B.M, Evans R.M. 15-Deoxy-Δ12,14-Prostaglandin J2 is a ligand for the adipocyte determination factor PPARγ Cell. 1995;83:803–812. [PubMed: 8521497]
  69. Frykholm P, Hillered L, Langstrom B, Persson L, Valtysson J, Enblad P. Relationship between cerebral blood flow and oxygen metabolism, and extracellular glucose and lactate concentrations during middle cerebral artery occlusion and reperfusion: A microdialysis and positron emission tomography study in nonhuman primates. J Neurosurg. 2005;102:1076–84. [PubMed: 16028767]
  70. Funk C.D. Prostaglandins and leukotrienes: Advances in eicosanoid biology. Science. 2001;294:1871–5. [PubMed: 11729303]
  71. Garcia J.H. The neuropathology of stroke. Hum Pathol. 1975;6:583–98. [PubMed: 809342]
  72. Gazi L, Gyles S, Rose J, Lees S, Allan C, Xue L. et al. Delta12-prostaglandin D2 is a potent and selective CRTH2 receptor agonist and causes activation of human eosinophils and Th2 lymphocytes. Prostaglandins Other Lipid Mediat. 2005;75:153–67. [PubMed: 15789622]
  73. Gelir E, Arslan S.O, Sayan H, Pinar L. Effect of rapid-eye-movement sleep deprivation on rat hypothalamic prostaglandins. Prostaglandins Leukot Essent Fatty Acids. 2005;73:391–6. [PubMed: 16039105]
  74. Gerriets T, Stolz E, Walberer M, Kaps M, Bachmann G, Fisher M. Neuroprotective effects of MK-801 in different rat stroke models for permanent middle cerebral artery occlusion: Adverse effects of hypothalamic damage and strategies for its avoidance. Stroke. 2003;34:2234–9. [PubMed: 12920258]
  75. Go A.S, Mozaffarian D, Roger V.L, Benjamin E.J, Berry J.D, Borden W.B. et al. Heart disease and stroke statistics—2013 update: A report from the American Heart Association. Circulation. 2013;127:e6–e245. [PMC free article: PMC5408511] [PubMed: 23239837]
  76. Gobbo O.L, O’Mara S.M. Post-treatment, but not pre-treatment, with the selective cyclooxygenase-2 inhibitor celecoxib markedly enhances functional recovery from kainic acid-induced neurodegeneration. Neuroscience. 2004;125:317–27. [PubMed: 15062975]
  77. Goh Y, Nakajima M, Azuma I, Hayaishi O. Effects of prostaglandin D2 and its analogues on intraocular pressure in rabbits. Jpn J Ophthalmol. 1988;32:471–80. [PubMed: 3236567]
  78. Granstrom E, Lands W.E, Samuelsson B. Biosynthesis of 9-alpha,15-dihydroxy-11-ketoprost-13-enoic acid. J Biol Chem. 1968;243:4104–8. [PubMed: 5666951]
  79. Gray S.J, Giles H, Posner J. The effect of a prostaglandin DP-receptor partial agonist (192C86) on platelet aggregation and the cardiovascular system in healthy volunteers. Br J Clin Pharmacol. 1992;34:344–51. [PMC free article: PMC1381418] [PubMed: 1457268]
  80. Green A.R. Why do neuroprotective drugs that are so promising in animals fail in the clinic? An industry perspective. Clin Exp Pharmacol Physiol. 2002;29:1030–4. [PubMed: 12366397]
  81. Green A.R, Ashwood T. Free radical trapping as a therapeutic approach to neuroprotection in stroke: Experimental and clinical studies with NXY-059 and free radical scavengers. Curr Drug Targets CNS Neurol Disord. 2005;4:109–18. [PubMed: 15857295]
  82. Gross G.J, Gauthier K.M, Moore J, Falck J.R, Hammock B.D, Campbell W.B, Nithipatikom K. Effects of the selective EET antagonist, 14,15-EEZE, on cardioprotection produced by exogenous or endogenous EETs in the canine heart. American J physiol. 2008;294:H2838–44. [PMC free article: PMC2863006] [PubMed: 18441205]
  83. Grosser T, Fries S, Fitzgerald G.A. Biological basis for the cardiovascular consequences of COX-2 inhibition: Therapeutic challenges and opportunities. J Clin Invest. 2006;116:4–15. [PMC free article: PMC1323269] [PubMed: 16395396]
  84. Grossetete M, Rosenberg G.A. Matrix metalloproteinase inhibition facilitates cell death in intracerebral hemorrhage in mouse. J Cereb Blood Flow Metab. 2008;28:752–63. [PubMed: 17971790]
  85. Hamid-Bloomfield S, Payne A.N, Petrovic A.A, Whittle B.J.R. The role of prostanoid TP- and DP-receptors in the bronchoconstrictor effect of inhaled PGD2 in anaesthetized guinea-pigs: Effect of the DP-antagonist BW A868C. Br J Pharmacol. 1990;100:761–766. [PMC free article: PMC1917592] [PubMed: 2119842]
  86. Hammad H, de Heer H.J, Soullie T, Hoogsteden H.C, Trottein F, Lambrecht B.N. Prostaglandin D2 inhibits airway dendritic cell migration and function in steady state conditions by selective activation of the D prostanoid receptor 1. J Immunol. 2003;171:3936–40. [PubMed: 14530310]
  87. Hammad H, Kool M, Soullie T, Narumiya S, Trottein F, Hoogsteden H.C. et al. Activation of the D prostanoid 1 receptor suppresses asthma by modulation of lung dendritic cell function and induction of regulatory T cells. J Exp Med. 2007;204:357–67. [PMC free article: PMC2118726] [PubMed: 17283205]
  88. Hanson M.G Jr, Shen S, Wiemelt A.P, McMorris F.A, Barres B.A. Cyclic AMP elevation is sufficient to promote the survival of spinal motor neurons in vitro. J Neurosci. 1998;18:7361–71. [PMC free article: PMC6793263] [PubMed: 9736656]
  89. Hata A.N, Breyer R.M. Pharmacology and signaling of prostaglandin receptors: Multiple roles in inflammation and immune modulation. Pharmacol Ther. 2004;103:147–66. [PubMed: 15369681]
  90. Hata A.N, Zent R, Breyer M.D, Breyer R.M. Expression and molecular pharmacology of the mouse CRTH2 receptor. J Pharmacol Exp Therapeutics. 2003;306:463–470. [PubMed: 12721327]
  91. Hatoum O.A, Gauthier K.M, Binion D.G, Miura H, Telford G, Otterson M.F. et al. Novel mechanism of vasodilation in inflammatory bowel disease. Arterioscler Thromb Vasc Biol. 2005;25:2355–61. [PubMed: 16141408]
  92. Hayaishi O. Molecular genetic studies on sleep-wake regulation, with special emphasis on the prostaglandin D2 system. J Appl Physiol. 2002;92:863–8. [PubMed: 11796702]
  93. Hayaishi O, Urade Y. Prostaglandin D2 in sleep-wake regulation: Recent progress and perspectives. Neuroscientist. 2002;8:12–5. [PubMed: 11843094]
  94. Hayashi M, Tanouchi T. Synthesis of 11-dehydro-13,14-dihydro-PGE1 [prostaglandin E1] and -PGD2 [prostaglandin D2] J Organic Chem. 1973;38:2115–2116.
  95. Heinemann A, Schuligoi R, Sabroe I, Hartnell A, Peskar B.A. Δ12-Prostaglandin J2, a plasma metabolite of prostaglandin D2, causes eosinophil mobilization from the bone marrow and primes eosinophils for chemotaxis. J Immunol. 2003;170:4752–4758. [PubMed: 12707356]
  96. Hewitt D.J. The use of NMDA-receptor antagonists in the treatment of chronic pain. Clin J Pain. 2000;16:S73–9. [PubMed: 10870744]
  97. Hinkle J.L, Bowman L. Neuroprotection for ischemic stroke. J Neurosci Nurs. 2003;35:114–8. [PubMed: 12795039]
  98. Hirai H, Tanaka K, Yoshie O, Ogawa K, Kenmotsu K, Takamori Y. et al. Prostaglandin D2 selectively induces chemotaxis in T helper type 2 cells, eosinophils, and basophils via seven-transmembrane receptor CRTH2. J Exp Med. 2001;193:255–61. [PMC free article: PMC2193345] [PubMed: 11208866]
  99. Hirano Y, Shichijo M, Deguchi M, Nagira M, Suzuki N, Nishitani Y. et al. Synergistic effect of PGD2 via prostanoid DP receptor on TNF-[alpha]-induced production of MCP-1 and IL-8 in human monocytic THP-1 cells. Eur J Pharmacol. 2007;560:81–88. [PubMed: 17307163]
  100. 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 U S A. 1994;91:11192–6. [PMC free article: PMC45193] [PubMed: 7972033]
  101. Hoffmann C. COX-2 in brain and spinal cord implications for therapeutic use. Curr Med Chem. 2000;7:1113–20. [PubMed: 11032961]
  102. Armitage J, Jiang L. HPS2-THRIVE randomized placebo-controlled trial in 25 673 high-risk patients of ER niacin/laropiprant: Trial design, pre-specified muscle and liver outcomes, and reasons for stopping study treatment. Eur Heart J. 2013;34:1279–1291. HPS2-THRIVE-Collaborative-Group. [PMC free article: PMC3640201] [PubMed: 23444397]
  103. Hurley S.D, Olschowka J.A, O’Banion M.K. Cyclooxygenase inhibition as a strategy to ameliorate brain injury. J Neurotrauma. 2002;19:1–15. [PubMed: 11852973]
  104. Iadecola C, Alexander M. Cerebral ischemia and inflammation. Curr Opin Neurol. 2001;14:89–94. [PubMed: 11176223]
  105. Iadecola C, Niwa K, Nogawa S, Zhao X, Nagayama M, Araki E. et al. Reduced susceptibility to ischemic brain injury and N-methyl-D-aspartate-mediated neurotoxicity in cyclooxygenase-2-deficient mice. Proc Natl Acad Sci USA. 2001;98:1294–9. [PMC free article: PMC14748] [PubMed: 11158633]
  106. Iadecola C, Ross M.E. Molecular pathology of cerebral ischemia: Delayed gene expression and strategies for neuroprotection. Ann N Y Acad Sci. 1997;835:203–17. [PubMed: 9616775]
  107. Ikonomidou C, Turski L. Why did NMDA receptor antagonists fail clinical trials for stroke and traumatic brain injury? Lancet Neurol. 2002;1:383–6. [PubMed: 12849400]
  108. Imig J.D. Epoxide hydrolase and epoxygenase metabolites as therapeutic targets for renal diseases. Am J Physiol Renal Physiol. 2005;289:F496–503. [PubMed: 16093425]
  109. Ishikawa T.O, Tamai Y, Rochelle J.M, Hirata M, Namba T, Sugimoto Y. et al. Mapping of the genes encoding mouse prostaglandin D, E, and F and prostacyclin receptors. Genomics. 1996;32:285–8. [PubMed: 8833158]
  110. Ishizuka T, Cheng J, Singh H, Vitto M.D, Manthati V.L, Falck J.R. et al. 20-Hydroxyeicosatetraenoic acid stimulates nuclear factor-kappaB activation and the production of inflammatory cytokines in human endothelial cells. J Pharmacol Exp Ther. 2008;324:103–10. [PubMed: 17947496]
  111. Jaillard A, Cornu C, Durieux A, Moulin T, Boutitie F, Lees K.R, Hommel M. Hemorrhagic transformation in acute ischemic stroke. The MAST-E study. MAST-E Group. Stroke. 1999;30:1326–32. [PubMed: 10390303]
  112. Jones R.L, Giembycz M.A, Woodward D.F. Prostanoid receptor antagonists: Development strategies and therapeutic applications. Br J Pharmacol. 2009;158:104–45. [PMC free article: PMC2795261] [PubMed: 19624532]
  113. Kabashima K, Narumiya S. The DP receptor, allergic inflammation and asthma. Prostaglandins Leukot Essent Fatty Acids. 2003;69:187–94. [PubMed: 12895602]
  114. Kagawa S, Fukunaga K, Oguma T, Suzuki Y, Shiomi T, Sayama K. et al. Role of prostaglandin D2 receptor CRTH2 in sustained eosinophil accumulation in the airways of mice with chronic asthma. Int Arch Allergy Immunol 155 Suppl. 2011;1:6–11. [PubMed: 21646789]
  115. Kam P.C, See A.U. Cyclo-oxygenase isoenzymes: Physiological and pharmacological role. Anaesthesia. 2000;55:442–9. [PubMed: 10792135]
  116. Kanekiyo T, Ban T, Aritake K, Huang Z.L, Qu W.M, 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. [PMC free article: PMC1851035] [PubMed: 17404210]
  117. Kanematsu Y, Kanematsu M, Kurihara C, Tada Y, Tsou T.-L, van Rooijen N. et al. Critical roles of macrophages in the formation of intracranial aneurysm. Stroke. 2011;42:173–178. [PMC free article: PMC3021554] [PubMed: 21106959]
  118. Kargman D.E, Tuck C, Berglund L, Lin I.F, Mukherjee R.S, Thompson E.V. et al. Lipid and lipoprotein levels remain stable in acute ischemic stroke: The Northern Manhattan Stroke Study. Atherosclerosis. 1998;139:391–9. [PubMed: 9712347]
  119. Kaufmann W.E, Worley P.F, 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 U S A. 1996;93:2317–21. [PMC free article: PMC39793] [PubMed: 8637870]
  120. Kelley K.A, Ho L, Winger D, Freire-Moar J, Borelli C.B, Aisen P.S, Pasinetti G.M. Potentiation of excitotoxicity in transgenic mice overexpressing neuronal cyclooxygenase-2. Am J Pathol. 1999;155:995–1004. [PMC free article: PMC1866889] [PubMed: 10487857]
  121. Kemp J.A, McKernan R.M. NMDA receptor pathways as drug targets. Nat Neurosci. 2002;5 Suppl:1039–42. [PubMed: 12403981]
  122. Khanapure S.P, Garvey D.S, Janero D.R, Letts L.G. Eicosanoids in inflammation: Biosynthesis, pharmacology, and therapeutic frontiers. Curr Top Med Chem. 2007;7:311–40. [PubMed: 17305573]
  123. Kishimoto K, Li R.C, Zhang J, Klaus J.A, Kibler K.K, Doré S. et al. Cytosolic phospholipase A2 alpha amplifies early cyclooxygenase-2 expression, oxidative stress and MAP kinase phosphorylation after cerebral ischemia in mice. J Neuroinflammation. 2010;7:42. [PMC free article: PMC2923122] [PubMed: 20673332]
  124. Koch K.A, Wessale J.L, Moreland R, Reinhart G.A, Cox B.F. Effects of BW245C, a prostaglandin DP receptor agonist, on systemic and regional haemodynamics in the anaesthetized rat. Clin Exp Pharmacol Physiol. 2005;32:931–5. [PubMed: 16405449]
  125. Kogure K, Yamasaki Y, Matsuo Y, Kato H, Onodera H. Inflammation of the brain after ischemia. Acta Neurochir Suppl. 1996;66:40–3. [PubMed: 8780795]
  126. Kudo I, Murakami M. Phospholipase A2 enzymes. Prostaglandins Other Lipid Mediat. 2002;68–69:3–58. [PubMed: 12432908]
  127. Lenzer J. Pfizer is asked to suspend sales of painkiller. BMJ. 2005:330–862. [PMC free article: PMC556183] [PubMed: 15831855]
  128. Liang X, Wu L, Hand T, Andreasson K. Prostaglandin D2 mediates neuronal protection via the DP1 receptor. J Neurochem. 2005;92:477–86. [PubMed: 15659218]
  129. Liebeskind D.S. Neuroprotection from the collateral perspective. IDrugs. 2005;8:222–8. [PubMed: 15772894]
  130. Lipton P. Ischemic cell death in brain neurons. Physiol Rev. 1999;79:1431–568. [PubMed: 10508238]
  131. Lipton S.A, Rosenberg P.A. Excitatory amino acids as a final common pathway for neurologic disorders. N Engl J Med. 1994;330:613–22. [PubMed: 7905600]
  132. Lo E.H. A new penumbra: Transitioning from injury into repair after stroke. Nat Med. 2008;14:497–500. [PubMed: 18463660]
  133. Lo E.H, Dalkara T, Moskowitz M.A. Mechanisms, challenges and opportunities in stroke. Nat Rev Neurosci. 2003;4:399–415. [PubMed: 12728267]
  134. Luster A.D, Tager A.M. T-cell trafficking in asthma: Lipid mediators grease the way. Nat Rev Immunol. 2004;4:711–24. [PubMed: 15343370]
  135. Lynch D.R, Guttmann R.P. Excitotoxicity: Perspectives based on N-methyl-D-aspartate receptor subtypes. J Pharmacol Exp Ther. 2002;300:717–23. [PubMed: 11861773]
  136. Macdonald R.L, Stoodley M. Pathophysiology of cerebral ischemia. Neurol Med Chir (Tokyo). 1998;38:1–11. [PubMed: 9540326]
  137. Madrigal J.L, Moro M.A, Lizasoain I, Lorenzo P, Fernandez A.P, Rodrigo J. et al. Induction of cyclooxygenase-2 accounts for restraint stress-induced oxidative status in rat brain. Neuropsychopharmacology. 2003;28:1579–88. [PubMed: 12784118]
  138. Maeda K, Hata R, Hossmann K.A. Regional metabolic disturbances and cerebrovascular anatomy after permanent middle cerebral artery occlusion in C57black/6 and SV129 mice. Neurobiol Dis. 1999;6:101–8. [PubMed: 10343325]
  139. Manley G.T, Fujimura M, Ma T, Noshita N, Filiz F, Bollen A.W, Chan P, Verkman A.S. Aquaporin-4 deletion in mice reduces brain edema after acute water intoxication and ischemic stroke. Nat Med. 2000;6:159–63. [PubMed: 10655103]
  140. Mantamadiotis T, Lemberger T, Bleckmann S.C, Kern H, Kretz O, Martin Villalba A. et al. Disruption of CREB function in brain leads to neurodegeneration. Nat Genet. 2002;31:47–54. [PubMed: 11967539]
  141. Matsugi T, Kageyama M, Nishimura K, Giles H, Shirasawa E. Selective prostaglandin D2 receptor stimulation elicits ocular hypotensive effects in rabbits and cats. Eur J Pharmacol. 1995;275:245–50. [PubMed: 7768291]
  142. Matsuoka T, Hirata M, Tanaka H, Takahashi Y, Murata T, Kabashima K. et al. Prostaglandin D2 as a mediator of allergic asthma. Science. 2000;287:2013–7. [PubMed: 10720327]
  143. Mendez I, Hachinski V, Wolfe B. Serum lipids after stroke. Neurology. 1987;37:507–11. [PubMed: 3822149]
  144. Minami T, Okuda-Ashitaka E, Nishizawa M, Mori H, Ito S. Inhibition of nociceptin-induced allodynia in conscious mice by prostaglandin D2. Br J Pharmacol. 1997;122:605–10. [PMC free article: PMC1564979] [PubMed: 9375954]
  145. Minghetti L. Cyclooxygenase-2 (COX-2) in inflammatory and degenerative brain diseases. J Neuropathol Exp Neurol. 2004;63:901–10. [PubMed: 15453089]
  146. Miyata N, Seki T, Tanaka Y, Omura T, Taniguchi K, Doi M. et al. Beneficial effects of a new 20-hydroxyeicosatetraenoic acid synthesis inhibitor, TS-011 [N-(3-chloro-4-morpholin-4-yl) phenyl-N’-hydroxyimido formamide], on hemorrhagic and ischemic stroke. J Pharmacol Exp Ther. 2005;314:77–85. [PubMed: 15831442]
  147. Mizoguchi A, Eguchi N, Kimura K, Kiyohara Y, Qu W.M, Huang Z.L. et al. Dominant localization of prostaglandin D receptors on arachnoid trabecular cells in mouse basal forebrain and their involvement in the regulation of non-rapid eye movement sleep. Proc Natl Acad Sci U S A. 2001;98:11674–9. [PMC free article: PMC58788] [PubMed: 11562489]
  148. Mohan S, Ahmad A.S, Glushakov A, Chambers C, Dore S. Putative role of prostaglandin receptor in intracerebral hemorrhage. Front Neurol. 2012;3:1–17. [PMC free article: PMC3477820] [PubMed: 23097645]
  149. Monneret G, Li H, Vasilescu J, Rokach J, Powell W.S. 15-Deoxy-Δ12,1412,14-prostaglandins D2 and J2 are potent activators of human eosinophils. J Immunol. 2002;168:3563–3569. [PubMed: 11907120]
  150. Morita-Fujimura Y, Fujimura M, Yoshimoto T, Chan P.H. Superoxide during reperfusion contributes to caspase-8 expression and apoptosis after transient focal stroke. Stroke. 2001;32:2356–61. [PubMed: 11588326]
  151. Murakami M, Kudo I. Prostaglandin e synthase: A novel drug target for inflammation and cancer. Curr Pharm Des. 2006;12:943–54. [PubMed: 16533161]
  152. Murray A.J. Pharmacological PKA inhibition: All may not be what it seems. Sci Signal. 2008;1 re4. [PubMed: 18523239]
  153. Murray A.J, Shewan D.A. Epac mediates cyclic AMP-dependent axon growth, guidance and regeneration. Mol Cell Neurosci. 2008;38:578–88. [PubMed: 18583150]
  154. Naffah-Mazzacoratti M.G, Bellissimo M.I, Cavalheiro E.A. Profile of prostaglandin levels in the rat hippocampus in pilocarpine model of epilepsy. Neurochem Int. 1995;27:461–6. [PubMed: 8574174]
  155. Nagata K, Hirai H. The second PGD(2) receptor CRTH2: Structure, properties, and functions in leukocytes. Prostaglandins Leukot Essent Fatty Acids. 2003;69:169–77. [PubMed: 12895600]
  156. Nagata K, Hirai H, Tanaka K, Ogawa K, Aso T, Sugamura K. et al. CRTH2, an orphan receptor of T-helper-2-cells, is expressed on basophils and eosinophils and responds to mast cell-derived factor(s). FEBS Lett. 1999a;459:195–9. [PubMed: 10518017]
  157. Nagata K, Tanaka K, Ogawa K, Kemmotsu K, Imai T, Yoshie O. et al. Selective expression of a novel surface molecule by human Th2 cells in vivo. J Immunol. 1999b;162:1278–86. [PubMed: 9973380]
  158. Nakayama M, Uchimura K, Zhu R.L, Nagayama T, Rose M.E, Stetler R.A. et al. Cyclooxygenase-2 inhibition prevents delayed death of CA1 hippocampal neurons following global ischemia. Proc Natl Acad Sci U S A. 1998;95:10954–9. [PMC free article: PMC28002] [PubMed: 9724811]
  159. Narumiya S. Prostanoids and inflammation: A new concept arising from receptor knockout mice. J Mol Med. 2009;87:1015–22. [PubMed: 19609495]
  160. Narumiya S, Sugimoto Y, Ushikubi F. Prostanoid receptors: Structures, properties, and functions. Physiol Rev. 1999;79:1193–226. [PubMed: 10508233]
  161. Narumiya S, Toda N. Different responsiveness of prostaglandin D2-sensitive systems to prostaglandin D2 and its analogues. Br J Pharmacol. 1985;85:367–75. [PMC free article: PMC1916609] [PubMed: 2992661]
  162. Nogawa S, Zhang F, Ross M.E, Iadecola C. Cyclo-oxygenase-2 gene expression in neurons contributes to ischemic brain damage. J Neurosci. 1997;17:2746–55. [PMC free article: PMC6573095] [PubMed: 9092596]
  163. Obal F Jr, Krueger J.M. Biochemical regulation of non-rapid-eye-movement sleep. Front Biosci. 2003;8:d520–50. [PubMed: 12700031]
  164. Park S.Y, Kim T.H, Kim H.I, Shin Y.K, Lee C.S, Park M, Song J.H. Celecoxib inhibits Na+ currents in rat dorsal root ganglion neurons. Brain Res. 2007;1148:53–61. [PubMed: 17359944]
  165. Phillis J.W, Horrocks L.A, Farooqui A.A. Cyclooxygenases, lipoxygenases, and epoxygenases in CNS: Their role and involvement in neurological disorders. Brain Res Rev. 2006;52:201–43. [PubMed: 16647138]
  166. Phillis J.W, O’Regan M.H. The role of phospholipases, cyclooxygenases, and lipoxygenases in cerebral ischemic/traumatic injuries. Crit Rev Neurobiol. 2003;15:61–90. [PubMed: 14513863]
  167. Phillis J.W, O’Regan M.H. A potentially critical role of phospholipases in central nervous system ischemic, traumatic, and neurodegenerative disorders. Brain Res Brain Res Rev. 2004;44:13–47. [PubMed: 14739001]
  168. Qu W.M, Huang Z.L, Xu X.H, Aritake K, Eguchi N, Nambu F. et al. Lipocalin-type prostaglandin D synthase produces prostaglandin D2 involved in regulation of physiological sleep. Proc Natl Acad Sci U S A. 2006;103:17949–54. [PMC free article: PMC1693853] [PubMed: 17093043]
  169. Rao A.V, Balachandran B. Role of oxidative stress and antioxidants in neurodegenerative diseases. Nutr Neurosci. 2002;5:291–309. [PubMed: 12385592]
  170. Renic M, Klaus J.A, Omura T, Kawashima N, Onishi M, Miyata N. et al. Effect of 20-HETE inhibition on infarct volume and cerebral blood flow after transient middle cerebral artery occlusion. J Cereb Blood Flow Metab. 2009;29:629–39. [PMC free article: PMC2821901] [PubMed: 19107134]
  171. Resnick D.K, Graham S.H, Dixon C.E, Marion D.W. Role of cyclooxygenase 2 in acute spinal cord injury. J Neurotrauma. 1998;15:1005–13. [PubMed: 9872457]
  172. Ribeiro J.D, Toro A.A, Baracat E.C. Antileukotrienes in the treatment of asthma and allergic rhinitis. J Pediatr (Rio J). 2006;82:S213–21. [PubMed: 17136298]
  173. Roger V.L, Go A.S, Lloyd-Jones D.M, Benjamin E.J, Berry J.D, Borden W.B. et al. Heart disease and stroke statistics— 2012 update: A report from the American Heart Association. Circulation. 2012;125:e2–e220. [PMC free article: PMC4440543] [PubMed: 22179539]
  174. Roman R.J. P-450 metabolites of arachidonic acid in the control of cardiovascular function. Physiol Rev. 2002;82:131–85. [PubMed: 11773611]
  175. Rosell A, Foerch C, Murata Y, Lo E.H. Mechanisms and markers for hemorrhagic transformation after stroke. Acta Neurochir Suppl. 2008;105:173–8. [PubMed: 19066105]
  176. Rossi A, Kapahi P, Natoli G, Takahashi T, Chen Y, Karin M, Santoro M.G. Anti-inflammatory cyclopentenone prostaglandins are direct inhibitors of IkappaB kinase. Nature. 2000;403:103–8. [PubMed: 10638762]
  177. Rouzer C.A, Marnett L.J. Cyclooxygenases: Structural and functional insights. J Lipid Res. 2009;50 Suppl:S29, 34. [PMC free article: PMC2674713] [PubMed: 18952571]
  178. Rydel R.E, Greene L.A. cAMP analogs promote survival and neurite outgrowth in cultures of rat sympathetic and sensory neurons independently of nerve growth factor. Proc Natl Acad Sci U S A. 1988;85:1257–61. [PMC free article: PMC279746] [PubMed: 2829221]
  179. Saleem S, Zhuang H, Biswal S, Christen Y, Dore S. Ginkgo biloba extract neuroprotective action is dependent on heme oxygenase 1 in ischemic reperfusion brain injury. Stroke. 2008;39:3389–96. [PMC free article: PMC6010172] [PubMed: 18845796]
  180. Saleem S, Zhuang H, de Brum-Fernandes A.J, Maruyama T, Narumiya S, Doré S. PGD2 DP1 receptor protects brain from ischemia-reperfusion injury. Eur J Neurosci. 2007;26:73–78. [PMC free article: PMC2386988] [PubMed: 17573924]
  181. Sanchez-Mejia R.O, Newman J.W, Toh S, Yu G.Q, Zhou Y, Halabisky B. et al. Phospholipase A2 reduction ameliorates cognitive deficits in a mouse model of Alzheimer’s disease. Nat Neurosci. 2008;11:1311–8. [PMC free article: PMC2597064] [PubMed: 18931664]
  182. Sandhya T.L, Ong W.Y, Horrocks L.A, Farooqui A.A. A light and electron microscopic study of cytoplasmic phospholipase A2 and cyclooxygenase-2 in the hippocampus after kainate lesions. Brain Res. 1998;788:223–31. [PubMed: 9555027]
  183. Sandig H, Andrew D, Barnes A.A, Sabroe I, Pease J. 9a,11b-PGF2 and its stereoisomer PGF2a are novel agonists of the chemoattractant receptor, CRTH2. FEBS Lett. 2006a;580:373–9. [PubMed: 16378605]
  184. Sandig H, Andrew D, Barnes A.A, Sabroe I, Pease J. 9α,11β-PGF2 and its stereoisomer PGF2α novel agonists of the chemoattractant receptor, CRTH2. FEBS Letters. 2006b;580:373–379. [PubMed: 16378605]
  185. Santovito D, Mezzetti A, Cipollone F. Cyclooxygenase and prostaglandin synthases: Roles in plaque stability and instability in humans. Curr Opin Lipidol. 2009;20:402–8. [PubMed: 19741338]
  186. Sawyer N, Cauchon E, Chateauneuf A, Cruz R.P.G, Nicholson D.W, Metters K.M. et al. Molecular pharmacology of the human prostaglandin D2 receptor, CRTH2. Br J Pharmacol. 2002;137:1163–72. [PMC free article: PMC1573602] [PubMed: 12466225]
  187. Seregi A, Folly G, Heldt R, Vizi E.S, Hertting G. Differential prostaglandin formation induced by convulsions in the brain of mice susceptible (DBA/2J) and resistant (CFLP) to acoustic stimulation. Epilepsy Res. 1990;5:131–6. [PubMed: 2328714]
  188. Serteser M, Ozben T, Gumuslu S, Balkan S, Balkan E. The effects of NMDA receptor antagonist MK-801 on lipid peroxidation during focal cerebral ischemia in rats. Prog Neuropsychopharmacol Biol Psychiatry. 2002;26:871–7. [PubMed: 12369259]
  189. Shah A, Pickles H, Joshi M, Webster A, O’Grady J. Effects of single oral dose administration of a hydantoin prostaglandin analogue BW 245C in man. Life Sci. 1984;34:2281–6. [PubMed: 6727564]
  190. Sharif N.A, Xu S.X, Williams G.W, Crider J.Y, Griffin B.W, Davis T.L. Pharmacology of [3H]prostaglandin E1/[3H]prostaglandin E2 and [3H]prostaglandin F2a binding to EP3 and FP prostaglandin receptor binding sites in bovine corpus luteum: Characterization and correlation with functional data. J Pharmacol Exp Ther. 1998;286:1094–102. [PubMed: 9694973]
  191. Shichijo M, Arimura A, Hirano Y, Yasui K, Suzuki N, Deguchi M, Abraham W.M. A prostaglandin D2 receptor antagonist modifies experimental asthma in sheep. Clin Exp Allergy. 2009;39:1404–14. [PubMed: 19486034]
  192. Shichijo M, Sugimoto H, Nagao K, Inbe H, Encinas J.A, Takeshita K. et al. Chemoattractant receptor-homologous molecule expressed on Th2 cells activation in vivo increases blood leukocyte counts and its blockade abrogates 13,14-dihydro-15-keto-prostaglandin D2-induced eosinophilia in rats. J Pharmacol Exp Ther. 2003;307:518–25. [PubMed: 12975488]
  193. Simmons D.L, Botting R.M, Hla T. Cyclooxygenase isozymes: The biology of prostaglandin synthesis and inhibition. Pharmacol Rev. 2004;56:387–437. [PubMed: 15317910]
  194. Simon R.P, Swan J.H, Griffiths T, Meldrum B.S. Blockade of N-methyl-D-aspartate receptors may protect against ischemic damage in the brain. Science. 1984;226:850–2. [PubMed: 6093256]
  195. Singh D. Merck withdraws arthritis drug worldwide. BMJ. 2004:329–816. [PMC free article: PMC521598] [PubMed: 15472245]
  196. Smith J.B, Silver M.J, Ingerman C.M, Kocsis J.J. Prostaglandin D2 inhibits the aggregation of human platelets. Thromb Res. 1974;5:291–9. [PubMed: 4439372]
  197. Smith W.L, DeWitt D.L, Garavito R.M. Cyclooxygenases: Structural, cellular, and molecular biology. Annu Rev Biochem. 2000;69:145–82. [PubMed: 10966456]
  198. Smith W.S. Pathophysiology of focal cerebral ischemia: A therapeutic perspective. J Vasc Interv Radiol. 2004;15:S3–12. [PubMed: 15101511]
  199. Spik I, Brenuchon C, Angeli V, Staumont D, Fleury S, Capron M. et al. Activation of the prostaglandin D2 receptor DP2/CRTH2 increases allergic inflammation in mouse. J Immunol. 2005;174:3703–8. [PubMed: 15749909]
  200. Sturzebecher S, Nieuweboer B, Matthes S, Schillinger E. Effects of PGD2, PGE1, and PGI2-analogues on PGDF-release and aggregation of human gelfiltered platelets. Prog Clin Biol Res. 1989;301:365–9. [PubMed: 2477855]
  201. Sugimoto M, Arai I, Futaki N, Hashimoto Y, Sakurai T, Honma Y, Nakaike S. The anti-pruritic efficacy of TS-022, a prostanoid DP1 receptor agonist, is dependent on the endogenous prostaglandin D2 level in the skin of NC/Nga mice. Eur J Pharmacol. 2007;564:196–203. [PubMed: 17328887]
  202. Sutherland B.A, Minnerup J, Balami J.S, Arba F, Buchan A.M, Kleinschnitz C. Neuroprotection for ischaemic stroke: Translation from the bench to the bedside. Int J Stroke. 2012;7:407–18. [PubMed: 22394615]
  203. Sutherland B.A, Papadakis M, Chen R.L, Buchan A.M. Cerebral blood flow alteration in neuroprotection following cerebral ischaemia. J Physiol. 2011;589:4105–14. [PMC free article: PMC3180571] [PubMed: 21708904]
  204. Szekely C.A, Thorne J.E, Zandi P.P, Ek M, Messias E, Breitner J.C, Goodman S.N. Nonsteroidal anti-inflammatory drugs for the prevention of Alzheimer’s disease: A systematic review. Neuroepidemiology. 2004;23:159–69. [PubMed: 15279021]
  205. Takemiya T, Matsumura K, Sugiura H, Maehara M, Yasuda S, Uematsu S. et al. Endothelial microsomal prostaglandin E synthase-1 exacerbates neuronal loss induced by kainate. J Neurosci Res. 2010;88:381–90. [PubMed: 19658194]
  206. Tanaka E, Niiyama S, Sato S, Yamada A, Higashi H. Arachidonic acid metabolites contribute to the irreversible depolarization induced by in vitro ischemia. J Neurophysiol. 2003;90:3213–23. [PubMed: 12917387]
  207. Taniguchi H, Mohri I, Okabe-Arahori H, Aritake K, Wada K, Kanekiyo T. et al. Prostaglandin D2 protects neonatal mouse brain from hypoxic ischemic injury. J Neurosci. 2007;27:4303–12. [PMC free article: PMC6672304] [PubMed: 17442814]
  208. Tasken K, Aandahl E.M. Localized effects of cAMP mediated by distinct routes of protein kinase A. Physiol Rev. 2004;84:137–67. [PubMed: 14715913]
  209. Tegtmeier F, Weber C, Heister U, Haker I, Scheller D, Nikolov R, Holler M. Eicosanoids in rat brain during ischemia and reperfusion—Correlation to DC depolarization. J Cereb Blood Flow Metab. 1990;10:358–64. [PubMed: 2329122]
  210. Teismann P, Vila M, Choi D.K, Tieu K, Wu D.C, Jackson-Lewis V. et al. COX-2 and neurodegeneration in Parkinson’s disease. Ann N Y Acad Sci. 2003;991:272–7. [PubMed: 12846993]
  211. Thura M, Hokamura K, Yamamoto S, Maeda M, Furuta K, Suzuki M. et al. GIF-0173 protects against cerebral infarction through DP1 receptor activation. Exp Neurol. 2009;219:481–91. [PubMed: 19576888]
  212. Toh H, Ichikawa A, Narumiya S. Molecular evolution of receptors for eicosanoids. FEBS Lett. 1995;361:17–21. [PubMed: 7890033]
  213. Topol E.J. Failing the public health—Rofecoxib, Merck, and the FDA. N Engl J Med. 2004;351:1707–9. [PubMed: 15470193]
  214. Toris C.B, Zhan G.L, Feilmeier M.R, Camras C.B, McLaughlin M.A. Effects of a prostaglandin DP receptor agonist, AL-6598, on aqueous humor dynamics in a nonhuman primate model of glaucoma. J Ocul Pharmacol Ther. 2006;22:86–92. [PubMed: 16722794]
  215. Urade Y, Hayaishi O. Prostaglandin D2 and sleep regulation. Biochim Biophys Acta. 1999;1436:606–15. [PubMed: 9989291]
  216. Urushitani M, Nakamizo T, Inoue R, Sawada H, Kihara T, Honda K. et al. N-methyl-D-aspartate receptor-mediated mitochondrial Ca2+ overload in acute excitotoxic motor neuron death: A mechanism distinct from chronic neurotoxicity after Ca2+ influx. J Neurosci Res. 2001;63:377–387. [PubMed: 11223912]
  217. Van Hecken A, Depré M, De Lepeleire I, Thach C, Oeyen M, Van Effen J. et al. The effect of MK-0524, a prostaglandin D2 receptor antagonist, on prostaglandin D2-induced nasal airway obstruction in healthy volunteers. Eur J Clin Pharmacol. 2007;63:135–141. [PubMed: 17200838]
  218. Vane J.R. Inhibition of prostaglandin synthesis as a mechanism of action for aspirin-like drugs. Nat New Biol. 1971;231:232–5. [PubMed: 5284360]
  219. Vartiainen N, Huang C.Y, Salminen A, Goldsteins G, Chan P.H, Koistinaho J. Piroxicam and NS-398 rescue neurones from hypoxia/reoxygenation damage by a mechanism independent of cyclo-oxygenase inhibition. J Neurochem. 2001;76:480–9. [PubMed: 11208911]
  220. Walton M, Woodgate A.M, Muravlev A, Xu R, During M.J, Dragunow M. CREB phosphorylation promotes nerve cell survival. J Neurochem. 1999;73:1836–42. [PubMed: 10537041]
  221. Walton M.R, Dragunow I. Is CREB a key to neuronal survival? Trends Neurosci. 2000;23:48–53. [PubMed: 10652539]
  222. Wang X. Recent advances in stroke: Molecular mechanisms, approaches, and treatments. Cent Nerv Syst Agents Med Chem. 2011;11:80. [PubMed: 21521173]
  223. Wardlaw J.M, Dennis M.S, Warlow C.P, Sandercock P.A. Imaging appearance of the symptomatic perforating artery in patients with lacunar infarction: Occlusion or other vascular pathology? Ann Neurol. 2001;50:208–15. [PubMed: 11506404]
  224. Weinberger J.M. Evolving therapeutic approaches to treating acute ischemic stroke. J Neurol Sci. 2006;249:101–9. [PubMed: 17005205]
  225. Whittle B.J, Moncada S, Mullane K, Vane J.R. Platelet and cardiovascular activity of the hydantoin BW245C, a potent prostaglandin analogue. Prostaglandins. 1983;25:205–23. [PubMed: 6344147]
  226. Wilson J.X, Gelb A.W. Free radicals, antioxidants, and neurologic injury: Possible relationship to cerebral protection by anesthetics. J Neurosurg Anesthesiol. 2002;14:66–79. [PubMed: 11773828]
  227. Wolf P.A, Abbott R.D, Kannel W.B. Atrial fibrillation: A major contributor to stroke in the elderly. The Framingham Study. Arch Intern Med. 1987;147:1561–4. [PubMed: 3632164]
  228. Woodward D.F, Hawley S.B, Williams L.S, Ralston T.R, Protzman C.E, Spada C.S. et al. Studies on the ocular pharmacology of prostaglandin D2. Invest Ophthalmol Vis Sci. 1990;31:138–46. [PubMed: 2298534]
  229. Woodward D.F, Jones R.L, Narumiya S. International Union of Basic and Clinical Pharmacology. LXXXIII: Classification of prostanoid receptors, updating 15 years of progress. Pharmacol Rev. 2011;63:471–538. [PubMed: 21752876]
  230. Woodward D.F, Lawrence R.A, Fairbairn C.E, Shan T, Williams L.S. Intraocular pressure effects of selective prostanoid receptor agonists involve different receptor subtypes according to radioligand binding studies. J Lipid Mediat. 1993;6:545–53. [PubMed: 8358015]
  231. Wright D.H, Abran D, Bhattacharya M, Hou X, Bernier S.G, Bouayad A. et al. Prostanoid receptors: Ontogeny and implications in vascular physiology. Am J Physiol Regul Integr Comp Physiol. 2001;281:R1343–60. [PubMed: 11641101]
  232. Wright D.H, Nantel F, Metters K.M, Ford-Hutchinson A.W. A novel biological role for prostaglandin D2 is suggested by distribution studies of the rat DP prostanoid receptor. Eur J Pharmacol. 1999;377:101–15. [PubMed: 10448933]
  233. Wymann M.P, Schneiter R. Lipid signalling in disease. Nat Rev Mol Cell Biol. 2008;9:162–76. [PubMed: 18216772]
  234. Xiang Z, Ho L, Yemul S, Zhao Z, Qing W, Pompl P, Kelley K. et al. Cyclooxygenase-2 promotes amyloid plaque deposition in a mouse model of Alzheimer’s disease neuropathology. Gene Expr. 2002;10:271–8. [PMC free article: PMC5977525] [PubMed: 12450219]
  235. Xing Y, Jiang X, Yang Y, Xi G. Hemorrhagic transformation induced by acute hyperglycemia in a rat model of transient focal ischemia. Acta Neurochir Suppl. 2011;111:49–54. [PubMed: 21725731]
  236. Yamamoto Y, Clower B.R, Haining J.L, Smith R.R. Effect of tissue plasminogen activator on intimal platelet accumulation in cerebral arteries after subarachnoid hemorrhage in cats. Stroke. 1991;22:780–4. [PubMed: 1905429]
  237. Zhao B.Q, Wang S, Kim H.Y, Storrie H, Rosen B.R, Mooney D.J. et al. Role of matrix metalloproteinases in delayed cortical responses after stroke. Nat Med. 2006;12:441–445. [PubMed: 16565723]
  238. Zhao H, Ohinata K, Yoshikawa M. Central prostaglandin D(2) exhibits anxiolytic-like activity via the DP(1) receptor in mice. Prostaglandins Other Lipid Mediat. 2009;88:68–72. [PubMed: 19007903]
  239. Zhuang H, Kim Y.S, Namiranian K, Doré S. Prostaglandins of J series control heme oxygenase expression: Potential significance in modulating neuroinflammation. Ann N Y Acad Sci. 2003a;993:208–16. [PubMed: 12853315]
  240. Zhuang H, Pin S, Li X, Doré S. Regulation of heme oxygenase expression by cyclopentenone prostaglandins. Exp Biol Med. 2003b;228:499–505. [PubMed: 12709576]
  241. Zivin J.A, Fisher M, DeGirolami U, Hemenway C.C, Stashak J.A. Tissue plasminogen activator reduces neurological damage after cerebral embolism. Science. 1985;230:1289–92. [PubMed: 3934754]
© 2015 by Taylor & Francis Group, LLC.
Bookshelf ID: NBK299187PMID: 26269890

Views

  • PubReader
  • Print View
  • Cite this Page

Other titles in this collection

Related information

  • PMC
    PubMed Central citations
  • PubMed
    Links to PubMed

Similar articles in PubMed

See reviews...See all...

Recent Activity

Your browsing activity is empty.

Activity recording is turned off.

Turn recording back on

See more...