Abstract
Overactive bladder (OAB) syndrome is a prevalent condition of the lower urinary tract that causes symptoms, such as urinary frequency, urinary urgency, urge incontinence, and nocturia, and disproportionately affects women and the elderly. Current medications for OAB merely provide symptomatic relief with considerable limitations, as they are no more than moderately effective, not to mention that they may cause substantial adverse effects. Identifying novel molecular targets to facilitate the development of new medical therapies with higher efficacy and safety for OAB is in an urgent unmet need. Although the molecular mechanisms underlying the pathophysiology of OAB largely remain elusive and are likely multifactorial, mounting evidence from preclinical studies over the past decade reveals that the pro-inflammatory pathways engaging cyclooxygenases and their prostanoid products, particularly the prostaglandin E2 (PGE2), may play essential roles in the progression of OAB. The goals of this review are to summarize recent progresses in our knowledge on the pathogenic roles of PGE2 in the OAB and to provide new mechanistic insights into the signaling pathways transduced by its four G-protein-coupled receptors (GPCRs), i.e., EP1–EP4, in the overactive detrusor smooth muscle. We also discuss the feasibility of targeting these GPCRs as an emerging strategy to treat OAB with better therapeutic specificity than the current medications.
Keywords: Detrusor overactivity, EP, GPCRs, Inflammation, OAB, PGE2
Graphical Abstract

1. Introduction
The overactive bladder (OAB) syndrome is commonly featured by the detrusor overactivity and constitutes the most prevalent type of the lower urinary tract dysfunction (LUTD). According to the International Urogynecology Association (IUGA) and the International Continence Society (ICS), OAB syndrome is defined as the urinary urgency that is accompanied by frequency and nocturia, either with (wet-OAB) or without (dry-OAB) urgency urinary incontinence, and in the absence of urinary tract infection or any other palpable urinary pathology [1]. As a chronic medical condition, OAB causes a sudden and frequent desire to urinate that is often difficult to control, imposing a major impact on the quality of life in a large proportion of the population. Depending on the study, the OAB condition has been diagnosed in up to 27% of men and 43% of women worldwide [2], and women are more likely to suffer an urgent urinary incontinence [3]. Moreover, the older populations are more prone to OAB symptoms, and the number of patients with OAB dramatically increases with age. More than 40% of patients with OAB have incontinence, and most urinary incontinence is solely caused by OAB. Though it is not life-threatening, OAB negatively affects people’s personal life and causes enormous economic burdens, as the average per capital cost and the total national cost for OAB per year are projected to be $1969 and $82.6 billion, respectively, by 2020 in the US alone [4].
The recommended first-line nonpharmacologic therapies for OAB symptoms are various behavioral interventions, such as lifestyle modification, training in voiding habits, and pelvic floor muscle exercise. Pharmacotherapy is often initiated as an adjuvant to optimize the overall outcomes when conservative management fails to achieve adequate improvement [2]. A number of antimuscarinic drugs and β3 adrenergic receptor agonists are currently available to treat OAB as the second-line treatment. In particularly, the β3 agonist drugs are increasingly prescribed largely owing to their fewer side effects [5], although their primary mechanisms of action are not fully understood yet [6]. Intravesical injection of onabotulinumtoxinA is considered to be safer and more effective as a third-line treatment for adults with refractory OAB who do not sufficiently respond to or cannot tolerate the second-line medications [2]. However, very few OAB patients can gain complete relief with these current medications, as they are no more than moderately effective in the vast majority of patients. In addition, all these anti-OAB drugs can have moderate to severe side effects: antimuscarinic drugs commonly cause dry mouth, constipation, and dyspepsia; β3 adrenergic receptor agonists lead to hypertension; the most common complications after onabotulinumtoxinA injection are urinary tract infection and retention [7]. Given the significant economic impacts of the disease and the substantial limitations of current medications, it is an urgent unmet need to identify new molecular targets and develop safer and more efficient therapies for OAB.
2. COX/PGE2 cascade in OAB
The etiology of OAB symptoms is not fully understood but is believed be associated with overactivity of the detrusor urinae muscle, which might relate to a variety of contributory factors, such as female sex, neurologic diseases, aging, bladder outlet obstruction, metabolic diseases [8, 9]. Several recent studies have identified signs of chronic inflammation in urine specimens and biopsied bladder tissues from the OAB patients without urinary tract infection [10, 11], insinuating a pathogenic role for inflammation in OAB. The upregulated inflammatory molecules found in patients with OAB include C-reactive protein (CRP), various cytokines, chemokines, and prostanoids [12, 13]. As a key pro-inflammatory mediator, prostaglandin E2 (PGE2) in the bladder is primarily synthesized in the urothelial and muscle layers and exerts local actions such as the micturition reflex under normal physiological and pathophysiological conditions [14]. Elevated PGE2 levels have been found in experimental animals with overactive detrusor [15, 16] as well as in patients with OAB [17, 18]. Intravesical administration of PGE2 led to urinary symptoms and detrusor overactivity in human patients and experimental animals [19–21], indicative of the involvement of PGE2 in the regulation of urinary smooth muscle function.
PGE2 is synthesized through the oxidization of arachidonic acid in a two-step reaction. During the first step, arachidonic acid from the membrane-bound phospholipids is converted by cyclooxygenase (COX) to intermediate prostaglandin G2 (PGG2) and then PGH2 (Fig. 1). As the second step, the short-lived PGH2 is rapidly converted to five different prostanoids, consisting of prostaglandins PGE2, PGD2, and PGF2α, thromboxane TXA2, and prostacyclin PGI2 by tissue-specific prostanoid synthases. Prostanoids exert their physiological and pathological functions through acting on a panel of G protein-coupled receptors (GPCRs), among which are four receptors bound and activated by PGE2, namely, EP1, EP2, EP3, and EP4 (Fig. 1). Receptor-coupled G proteins are heterotrimers consisting of α, β, and γ subunits, among which the Gα subunit plays a dominant role in the hydrolytic function of G proteins [22]. In bladder detrusor muscle, EP2, EP4 and β3-adrenergic receptors are coupled to Gαs subunit that stimulates the cAMP signaling; M3 muscarinic and EP1 receptors are associated with Gαq that can lead to the calcium-calmodulin and protein kinase C (PKC)-mediated pathways; EP3 mainly links with Gαi to exert an inhibitory effect on the cAMP generation (Fig. 1).
Figure 1. PGE2 biosynthesis and signaling.

The biosynthesis of PGE2 begins from the cleavage of membrane-bound phospholipid into arachidonic acid, which is catalyzed by phospholipase A2 (PLA2). Subsequently, the arachidonic acid is oxidized by cyclooxygenase-1 or 2 (COX-1/2) to intermediate prostaglandin G2 (PGG2) and then PGH2. The short-lived PGH2 is rapidly converted to PGE2 by tissue-specific PGE synthase (PGES). PGE2 exerts its physiological and pathological functions through acting on a panel of G protein-coupled receptors (GPCRs), EP1, EP2, EP3, and EP4. In bladder detrusor muscle, the activation of EP1 and EP3 are likely to induce muscle contraction, while the activation of EP4 is likely to cause muscle relaxation. The function of EP2 in the urinary bladder needs further investigation.
COX has two isozymes: constitutive COX-1 and inducible COX-2, both of which are found in human urothelium and detrusor muscle of the bladder (COX-1: https://www.ncbi.nlm.nih.gov/gene/5742; COX-2: https://www.ncbi.nlm.nih.gov/gene/5743) [23, 24]. The constitutional expression of COX-1 is responsible for the biosynthesis of prostanoids to maintain the homeostasis within the normal bladder, whereas the COX-2 can be quickly induced by various detrimental or pro-inflammatory stimuli under pathophysiological conditions [25]. Because the COX-2 induction can trigger long-lasting pro-inflammatory processes that might facilitate the disease progression, a number of selective and non-selective COX inhibitors have been tested to determine whether blocking the COX enzymes can decrease the spontaneous contractile activities of the bladder in various animal models of OAB.
Intravesical instillation of aspirin in rabbit bladders that were partially obstructed attenuated the contractility caused by the cholinergic stimulation with KCl [26]. Several other non-selective COX inhibitors, such as dexketoprofen, indomethacin, meloxicam, naproxen, paracetamol, flurbiprofen, and nimesulide, as well as COX-2 selective inhibitors including NS-398, celecoxib, rofecoxib, and L-745337, were investigated in rat urodynamic models. The results from these studies overall indicated that the COX-1 may be involved in modulating the activation threshold of the micturition reflex under normal conditions, whereas the COX-2 inhibition can prevent or reverse the urodynamic alterations triggered by bladder inflammation [27, 28]. In a clinical study involving 32 patients with urinary symptoms due to detrusor instability, the administration of indomethacin for one month was demonstrated to achieve a highly significant improvement in diurnal and nocturnal frequency [29]. Likewise, intravesical administration of ketoprofen, another non-selective COX inhibitor, for four weeks has been shown to improve the OAB symptom in 18 out of 30 female patients without any observatory side effects [30]. The success in these preclinical and clinical studies suggest that the COX inhibition might represent a promising strategy to treat OAB. However, the well-recognized severe adverse effects on gastrointestinal and cardiovascular systems by the chronic use of COX inhibitors indicate that certain COX prostanoid products might mediate some beneficial effects [31]. As such, targeting a key pro-inflammatory prostanoid receptor might represent a more specific therapeutic strategy compared to generic block of the entire COX cascade [32, 33].
3. PGE2 receptors and OAB
COX enzymes execute pro-inflammatory effects mainly through synthesizing PGE2. Upon COX-2 induction, the elevated PGE2 binds and activates four receptors EP1–EP4, which have divergent engagement in phosphoinositol and cAMP-dependent pathways (Fig. 2) [34, 35]. These membrane-bound receptors are differentially expressed in the lower urinary system (EP1: https://gtexportal.org/home/gene/PTGER1; EP2: https://gtexportal.org/home/gene/PTGER2; EP3: https://gtexportal.org/home/gene/PTGER3; EP4: https://gtexportal.org/home/gene/PTGER4), and play important roles in the maintenance of normal bladder function as well as the pathogenesis of bladder dysfunctions [36]. Given that modulating PGE2 signaling might provide better therapeutic specificity than blocking PGE2 synthesis by COX inhibition, over the past decade these four EP receptors have been under intense investigation as potential molecular targets for new treatments of OAB. In the rest of this review, we discuss their contributions to the pathophysiology of detrusor overactivity and examine the feasibility of pharmacologically targeting these GPCRs for new pharmacotherapies of OAB (Tables 1–4).
Figure 2. PGE2 signaling that regulates the activities of detrusor smooth muscle.

EP1 receptor couples to Gαq and its activation stimulates phospholipase C (PLC) that hydrolyzes phosphatidylinositol 4,5-bisphosphate (PIP2) into diacylglycerol (DAG) and inositol 1,4,5-trisphosphate (IP3). DAG further activates protein kinase C (PKC) which induces the Ca2+ influx current through the voltage dependent Ca2+ channel (VDCC). Meanwhile, PKC may also cause a direct inhibition of the ryanodine receptor (RyR) or a reduction in the sarcoplasmic reticulum (SR) Ca2+ load, resulting in an inhibition of the big potassium channel (BK channel) of the detrusor smooth muscle. IP3 elevates the intracellular Ca2+ levels through the interaction with IP3 sensitive Ca2+ channel at the SR. The intracellular Ca2+ of the detrusor smooth muscle binds to the calmodulin to form a calcium-calmodulin complex that activates myosin light chain kinase (MLCK). MLCK phosphorylates myosin to enable its interaction with the actin for the initiation of cross-bridge cycling and muscle contraction. Both EP2 and EP4 receptors couple to Gαs subunit which activates the adenylate cyclase (AC) to synthesize cAMP. cAMP in turn activates protein kinase A (PKA) that subsequently inhibits the VDCC but activates Na+-Ca2+ exchanger (NCX) on the cell membrane. Activated PKA also phosphorylates both IP3 sensitive Ca2+ channel and Ca2+-ATPase on the SR to cause Ca2+ sequestration, and thus downregulates the intracellular Ca2+ levels. PKA might also activate RyR, leading to the Ca2+ releasing to form Ca2+ sparks, which in turn activate the BK channel. Upon activation, the transient BK current leads to the hyperpolarization of the cell membrane which closes the VDCC. EP3 receptor mainly couples to Gαi for an inhibitory effect on adenylate cyclase, thereby downregulating the intracellular cAMP levels to increase Ca2+ levels and facilitate the muscle contraction. Only the major pathways are indicated.
Table 1.
Recent studies on the EP1 receptor in OAB models
| Drug | Drug action | Experimental model | Drug dose and route | Main therapeutic outcomes | Reference |
|---|---|---|---|---|---|
| N/A | N/A | Moderate BOO induced in female adult DBA/1LacJ mice | Genetic ablation of EP1 receptor | • Did not change the urodynamic parameters or bladder hypertrophy. • Prevented the intravesical PGE2-induced detrusor overactivity. |
Schröder et al., 2004 |
| ONO-8711 | Antagonist | Acetic acid (0.1%)-induced inflammation of the urinary bladders in adult female Wistar rats | 1 and 3 mg/kg, i.v. | • Reduced the inflammation-associated bladder afferent neuronal activity. | Ikeda et al., 2006 |
| PF-2907617-02 | Antagonist | Normal adult female Sprague-Dawley rats | 0.1 and 1 mg/kg, i.v. | • PF-2907617-02 (1 mg/kg) increased bladder capacity, micturition volume and micturition interval but had no effect on other urodynamic parameters in normal rats. | Lee et al., 2007 |
| PF-2907617-02 | Antagonist | Moderate BOO induced in adult female Sprague-Dawley rats | 1 mg/kg, i.v. | • Increased micturition interval and decreased the frequency and amplitude of nonvoiding contraction in rats with BOO. • Had no effect on bladder capacity or residual volume in BOO rats. |
Lee et al., 2007 |
| PF-2907617-02 | Antagonist | Intravesical PGE2 (50 µM)-induced detrusor overactivity in adult female Sprague-Dawley rats | 0.1 and 1 mg/kg, i.v. | • Decreased the intravesical PGE2 stimulated detrusor overactivity in normal rats. | Lee et al., 2007 |
| PF-2907617 | Antagonist | Bladder strips from female adult Sprague Dawley rats, male cynomolgus macaque monkeys | 300 nM, bath | • Caused an inhibitory effect on the PGE2-induced contraction of bladder strips from rats, but not of those from monkeys or humans. | Root et al., 2015 |
| ONO-8713 | Antagonist | Male guinea pig urinary bladder | 500 nM, bath | • Decreased the frequency of muscarinic agonist arecaidine-induced bladder contraction. | Hohnen et al., 2016 |
Abbreviations: BOO, bladder outlet obstruction; i.v., intravenous; OAB, overactive bladder; PGE2, prostaglandin E2.
Table 4.
Recent OAB studies on the PGE2 receptors in humans
| Drug | Drug action | Experimental model | Drug dose and route | Main therapeutic outcomes | Reference |
|---|---|---|---|---|---|
| ONO-8539 | EP1 Antagonist | 435 OAB patients in a 12-week, randomized, double-blind, placebo controlled, parallel group study | 30, 100 or 300 mg, twice daily | • No significant improvement compared to the placebo. | Chapple et al., 2014 |
| PF-2907617 | EP1 Antagonist | Bladder strips from male and female humans (age: 47–77 years) | 300 nM, bath | • Caused an inhibitory effect on the PGE2-induced contraction of bladder strips from rats, but not of those from monkeys or humans. | Root et al., 2015 |
| Sulprostone | EP3 Agonist | Bladder strips from male and female humans (age: 47–77 years) | 300 nM, bath | • Induced potent contraction of rat bladder muscles. • Caused weak contraction in monkey and human bladder strips. |
Root et al., 2015 |
| CJ24979 | EP3 Antagonist | Bladder strips from male and female humans (age: 47–77 years) | 1 µM, bath | • Inhibited the PGE2-induced contraction of bladder strips from rats and monkeys, but not those from humans. | Root et al., 2015 |
Abbreviations: OAB, overactive bladder; PGE2, prostaglandin E2.
3.1. EP1 receptor
Among the four EP receptors, the expression of EP1 in the mouse bladder is the highest (https://www.ncbi.nlm.nih.gov/gene/19216), suggesting some physiological functions in the lower urinary system. EP1 receptor couples to Gαq – upon activation by PGE2 – to stimulate phospholipase C (PLC) that hydrolyzes phosphatidylinositol 4,5-bisphosphate (PIP2) into diacylglycerol (DAG) and inositol 1,4,5-trisphosphate (IP3) [37]. DAG in turn directly activates some isoforms of protein kinase C (PKC) that can induce the Ca2+ influx current through the voltage dependent Ca2+ channels (VDCC) (Fig. 2) [38, 39]. In the meantime, PKC may also cause a direct inhibition of the ryanodine receptors (RyRs) or a reduction in the sarcoplasmic reticulum Ca2+ load, resulting in an inhibition of the large conductance calcium-activated potassium channel (or big potassium, BK) channels of the detrusor smooth muscle [39]. In fact, it has been suggested that the outward spontaneous transient BK currents (TBKCs) from the activated BK channels can induce the hyperpolarization of the cell membrane and detrusor muscle relaxation [40], where a direct action of estrogen on the BK channels might be involved [41]. It is worth noting that the involvement of female gonadal hormone such as estrogen and progesterone in the regulation of BK channels might contribute to sex difference in the prevalent of urgent urinary incontinence [42–44]. As the other second messenger of Gαq, IP3 elevates the cytoplasmic Ca2+ level via activating its receptors in the sarcoplasmic reticulum that function as Ca2+ channels [45]. The cytoplasmic Ca2+ of the detrusor smooth muscle binds to the calmodulin to form a Ca2+-calmodulin complex that activates myosin light chain kinase (MLCK). The latter then phosphorylates myosin to enable its interaction with the actin for the initiation of cross-bridge cycling and muscle contraction (Fig. 2) [46]. In addition to its presence in the detrusor muscle and urothelium, EP1 receptor has been shown in the intramural ganglia cells of the bladder [47], suggesting its involvement of neurologic regulation of detrusor muscle contraction [48].
The genetic deletion of EP1 receptor prevented detrusor overactivity caused by PGE2 stimulation or outlet obstruction in mice (Table 1) [49]. ONO-8711, a selective EP1 antagonist, was shown to decrease the afferent nerve activity in the rat bladder, indicating that the EP1 receptor might play an important role in the inflammation-induced primary afferent never activity that elicits the micturition reflex [50]. Similarly, intravenous administration of EP1 antagonist PF-2907617–02 increased bladder capacity, micturition volume, and micturition interval, and decreased the frequency and amplitude of non-voiding contraction in rats with partial bladder outlet obstruction (BOO) or PGE2-induced OAB [20]. Ex vivo studies using bladder stripes also suggested that the EP1 inhibition by PF-2907617-02 or ONO-8713 was able to reduce the bladder contraction frequency in rats and guinea pigs [48, 51]. Despite the success in preclinical studies, a randomized, double-blind, placebo controlled phase II clinical trial showed that daily administration of an EP1 antagonist ONO-8539 for 12 weeks did not significantly improve the micturition behavior in human patients with nonneurogenic overactive bladder syndrome (https://clinicaltrials.gov/ct2/show/NCT00876421) (Table 4) [52].
3.2. EP2 receptor
Both EP2 and EP4 receptors couple to Gαs subunit that activates the adenylate cyclase to generate cAMP, which has been shown to play essential roles in human detrusor smooth muscle contraction via regulating the calcium sensitization [53]. However, it has been suggested that the functional coupling to cAMP is more efficient for EP2 receptor than EP4 [35]. cAMP directly activates protein kinase A (PKA), which subsequently inhibits the voltage-dependent Ca2+ channel but activates the Na+-Ca2+ exchanger (NCX) on the cell membrane. Furthermore, the activated PKA also lead to the phosphorylation of both IP3 sensitive Ca2+ channel and Ca2+-ATPase on the SR of the detrusor smooth muscle cells to cause Ca2+ sequestration. These combined effects by PKA cause a downregulation of the intracellular Ca2+ levels (Fig. 2) [54, 55]. In addition, PKA presumably can stimulate the RyRs on the sarcoplasmic reticulum to release Ca2+ sparks and activate the BK channels, thereby inducing the hyperpolarization of urinary bladder smooth muscle cells, which facilitates detrusor muscle relaxation [40, 56].
However, there is very little information about the role of EP2 in the physiological process of micturition, even though the expression of the receptor has been demonstrated on the urethra, detrusor muscle and interstitial ganglia cells in the bladder of guinea pigs [47, 57, 58] as well as in the adult mouse bladder (https://www.ncbi.nlm.nih.gov/gene/19217). The oral administration of ONO-8055, a dual EP2/EP3 agonist, decreased post-void residual urine, voiding pressure, bladder capacity, and urethral pressure in rats with neurogenic underactive bladder (UAB) caused by the lumbar spinal canal stenosis [59], but did not augment the urethral relaxation or bladder contractility in normal rats [60]. Interestingly, ONO-8055 increased the voided volume, average flow rate, and maximum flow rate in monkeys with underactive bladder induced by radical hysterectomy [61]. These studies together demonstrated the therapeutic potential of ONO-8055 to treat neurogenic underactive bladder syndrome. However, whether EP2 or EP3 is the therapeutic target of the compound remains to be determined, as the compound shows similar potency on EP2 and EP3 (EC50: 0.67 vs. 0.7 nM) [62]. Up to date, no selective EP2 antagonist has been tested in animal models of OAB.
3.3. EP3 receptor
Although the EP3 receptor primarily couples to Gi for an inhibitory effect on adenylate cyclase to downregulate the intracellular cAMP level, it has several alternative splice variants that differ in their intracellular C-terminus and are associated with multiple different G proteins [63]. In humans, in addition to Gi, the EP3-II and EP3-IV isoforms also couple to Gs, whereas EP3-I and EP3-II couple to Gq. Likewise, there are three EP3 isoforms in mice – α, β, and γ. The EP3γ isoform appears to also couple to Gs, and all these three isoforms couple to G12/13 to activate Rho pathway for smooth muscle contraction. In consequence, the G protein complexes associate with EP3, upon the receptor activation by PGE2, dissociate into Gαi, Gα12/13, Gαs, and Gβγ components that proceed to activate multiple signaling pathways depending on the cellular context. Thus, a differential isoform expression pattern of the receptor can provide another level of regulation of the EP3-mediated signaling in different types of cells and tissues. The expression of EP3 in the bladder is relatively low when compared to other organs and tissues, such as adrenal gland, colon, kidney, mammary gland, ovary, placenta, small intestine, and fat pad (https://www.ncbi.nlm.nih.gov/gene/19218).
The genetic ablation of EP3 in mice improved the bladder capacity by approximately 185% and largely blunted the PGE2-induced bladder hyperactivity, whereas intravesical infusion of the selective EP3 receptor agonist GR63799X considerably reduced the bladder capacity in wild-type mice (Table 2) [64]. In line, EP3 receptor activation by sulprostone induced potent contraction of rat bladder muscles, whereas selective EP3 antagonist CJ24979 antagonized the PGE2-induced contraction of bladder strips (Tables 2 and 4) [48]. In addition to its expression in the urothelium and bladder detrusor muscle, EP3 has been found in the interstitial cells of Cajal (ICC) of the bladder, where its activation by PGE2 might contribute to the bladder detrusor contraction via activating the hyperpolarization-activated cyclic nucleotide-gated (HCN) channels and Ca2+ influx [65]. It is likely that EP3 receptor activates the HCN channels via altering the intracellular cAMP levels, generating an inward Na+ current that leads to the membrane depolarization and activation of T-type voltage-dependent Calcium channels [65, 66]. Intravenous administration of DG-041, a peripherally restricted EP3-selective antagonist, decreased the frequency of bladder rhythmic contraction and prevented the visceromotor reflex (VMR) response to bladder distension in rats [67]. Intraduodenal administration of selective EP3 agonist GR63799X reduced, while EP3 antagonists DG-041 and CM-9 enhanced, the bladder capacity in conscious rats, evaluated by assessing the micturition interval and volume of urine per void [68]. Similar to the EP4, the protein expression of EP3 in the bladder was induced by nearly three-fold in encephalomyelitis mice with OAB symptoms, and the intravenous injection of EP3 antagonist DG-041 was shown to decrease the micturition frequency per day and increase the void weight per void in these animals [69].
Table 2.
Recent studies on the EP3 receptor in OAB models
| Drug | Drug action | Experimental model | Drug dose and route | Main therapeutic outcomes | Reference |
|---|---|---|---|---|---|
| N/A | N/A | PGE2-induced OAB in adult 129P3/J or B6 mice | Genetic ablation of EP3 receptor | • Enhanced bladder capacity under control conditions. • Blunted the intravesical PGE2 induced bladder overactivity. |
McCafferty et al., 2008 |
| GR63799X | Agonist | PGE2-induced OAB in adult 129P3/J or B6 mice | 10 µM, intravesical infusion for 120 min | • Reduced bladder capacity under control conditions in wild-type but not in EP3 knock-out mice. | McCafferty et al., 2008 |
| DG-041 | Antagonist | Adult female Sprague-Dawley rat bladder rhythmic contraction model and bladder pain model | 10 mg/kg, i.v. | • Reduced the frequency of bladder rhythmic contraction. • Inhibited the VMR response to bladder distension. |
Su et al., 2008 |
| GR63799X | Agonist | Female spontaneously hypertensive rats | 0.001–1 mg/kg, i.d. | • Reduced bladder capacity. • Reduced micturition interval and void volume. |
Jugus et al., 2009 |
| CM-9 | Antagonist | Female spontaneously hypertensive rats | 10 and 30 mg/kg, i.d. | • Enhanced bladder capacity. • Increased micturition interval and void volume. |
Jugus et al., 2009 |
| DG-041 | Antagonist | Female spontaneously hypertensive rats | 30 mg/kg, i.d. | • Enhanced bladder capacity. • Increased micturition interval and void volume. |
Jugus et al., 2009 |
| Sulprostone | Agonist | Neurologic bladder model induced by encephalomyelitis in adult female SWXJ mice | 100 µg/kg, i.v. | • Increased micturition frequency and decreased urine output per micturition over a 24-hour test period. | Xue et al., 2013 |
| DG-041 | Antagonist | Neurologic bladder model induced by encephalomyelitis in adult female SWXJ mice | 100 µg/kg, i.v. | • Decreased micturition frequency and increased urine output per micturition over a 24-hr test period. | Xue et al., 2013 |
| Sulprostone | Agonist | Bladder strips from female adult Sprague Dawley rats, male cynomolgus macaque monkeys | 300 nM, bath | • Induced potent contraction of rat bladder muscles. • Caused weak contraction in monkey and human bladder strips. |
Root et al., 2015 |
| CJ24979 | Antagonist | Bladder strips from female adult Sprague Dawley rats, male cynomolgus macaque monkeys | 1 µM, bath | • Inhibited the PGE2-induced contraction of bladder strips from rats and monkeys, but not those from humans. | Root et al., 2015 |
| Sulprostone | Agonist | Bladder detrusor strips from adult female C57BL/6 mice and Sprague-Dawley rats. | 10 μM, bath | • Increased the calcium concentration of the ICCs. • Enhanced the contraction amplitude of the bladder detrusor. |
Wu et al., 2017 |
Abbreviations: ICCs, interstitial cells of Cajal; i.d., intraduodenal; i.v., intravenous; OAB, overactive bladder; PGE2, prostaglandin E2; VMR, visceromotor reflex.
3.4. EP4 receptor
As the other PGE2 receptor coupled to Gαs, EP4 is also expressed in the normal adult bladder at a relatively low basal lever (https://www.ncbi.nlm.nih.gov/gene/19219) but dramatically increases in obstructed bladder detrusor smooth muscle and epithelium in rats (Table 3) [70]. A selective EP4 agonist ONO-AE1–329 was able to relax KCl-induced contraction of bladder strips from rats with bladder outlet obstruction (BOO). Further, the intravesical infusion of ONO-AE1–329 significantly increased bladder capacity without altering the micturition pressure in rats with bladder outlet obstruction, whereas it had no effect in control animals [70]. In another interesting study, EP4 expression was elevated by nearly 100% in the detrusor muscle of OAB rats induced by cyclophosphamide (CYP). However, AH23848, a selective EP4 antagonist – when given intravenously – significantly extended the intercontraction interval (ICI) in CYP-treated rats without affecting the normal cohort [71]. Similarly, intravenous injection of AH23848 was also shown to decrease the micturition frequency and improve the OAB symptoms in encephalomyelitis mice, where EP4 expression was also elevated in the OAB bladder [69]. Intravenous or intravesical administration of MF-191, a more potent and selective EP4 antagonist, suppressed the CYP-induced OAB and inflammatory cell infiltration in the detrusor muscle of rats [72]. Interestingly, formalin-induced prostatic inflammation also elevated the PGE2 level and the expression of EP4, but not other EP receptors, in both the bladder mucosa and detrusor layers. Further, intravesical application of selective EP4 antagonist ONO-AE3–208 increased the intercontraction interval in rats with prostatic inflammation but did not affect the control animals [73]. Taken together, these findings suggest that the EP4 expression is generally induced in OAB detrusor muscle and epithelium, and the EP4 activation overall contributes to the inflammation-associated OAB but might counteract against the BOO-induced OAB. The conflicting outcomes might be explained by the specificity of each OAB model and different compounds (agonists vs. antagonists) used in these studies. However, more study using genetic approach is required to fully understand the roles of EP4 receptor in OAB and might help to clarify this contradiction.
Table 3.
Recent studies on the EP4 receptor in OAB models
| Drug | Drug action | Experimental model | Drug dose and route | Main therapeutic outcomes | Reference |
|---|---|---|---|---|---|
| AH-23848 | Antagonist | CYP-induced OAB in adult female Sprague-Dawley rats | 0.01 and 0.1 mg/kg, i.v. | • Extended intercontraction interval. • Had no effect on other urodynamic variables such as baseline pressure, pressure threshold, and contraction amplitude. • Had no significant effect in control animals. |
Chuang et al., 2010 |
| OAO-AE1–329 | Agonist | Bladder strips from rats with BOO | , bath | • Relaxed KCl-induced contraction. | Beppu et al., 2011 |
| OAO-AE1–329 | Agonist | Rats with BOO | 10 µM, continuously intravesical infusion | • Increased bladder capacity without altering micturition pressure. • No effect in control animals. |
Beppu et al., 2011 |
| MF-191 | Antagonist | CYP-induced OAB in adult female Sprague-Dawley rats | 0.1 and 1 mg/kg, i.v. | • MF-191 (only 1 mg/kg) improved intercontraction interval and reduced inflammatory cell infiltration. | Chuang et al., 2012 |
| MF-191 | Antagonist | CYP-induced OAB in adult female Sprague-Dawley rats | 10 and 100 nM, continuously intravesical infusion | • MF-191 (100 nM) suppressed CYP-induced bladder overactivity. | Chuang et al., 2012 |
| MF-191 | Antagonist | Intravesical PGE2-induced bladder overactivity in rats | 0.1 and 1 mg/kg, i.v. | • MF-191 (1 mg/kg) improved intercontraction interval but had no effect on baseline pressure, pressure threshold, and contraction amplitude. | Chuang et al., 2012 |
| CAY-10598 | Agonist | Neurologic bladder model induced by encephalomyelitis in adult female SWXJ mice | 100 µg/kg, i.v. | • Increased micturition frequency and decreased urine output per micturition over a 24-hour test period. | Xue et al., 2013 |
| AH-23848 | Antagonist | Neurologic bladder model induced by encephalomyelitis in adult female SWXJ mice | 100 µg/kg, i.v. | • Decreased micturition frequency and increased urine output per micturition over a 24-hour test period. | Xue et al., 2013 |
| ONO-AE3–208 | Antagonist | Prostatitis induced OAB in male adult Sprague-Dawley rats | 30 µM, intravesical infusion | • Increased intercontraction interval. • Did not have any significant effect in control rats. |
Mizoguchi et al., 2019 |
Abbreviations: BOO, bladder outlet obstruction; CYP, cyclophosphamide; i.v., intravenous; KCl, potassium chloride; OAB, overactive bladder; PGE2, prostaglandin E2.
4. Conclusive remarks and perspectives
Though widely considered as a benign condition, OAB negatively affects people’s personal life and imposes enormous economic burdens on individuals, families, and the society. The current behavioral treatment combined with pharmacotherapy have considerable limitations, as they are merely able to provide moderate efficacy for only a small proportion of patients. Daily use of β3 adrenergic receptor agonists and antimuscarinic drugs for OAB substantially increases the risk of moderate to severe side effects such as dry mouth, constipation, hypertension, and even cardiovascular abnormalities, which further complicate the urinary conditions. Recent evidence from animal studies suggests that PGE2 via its four GPCRs plays essential roles during the pathogenesis of OAB in various models (Tables 1–4). With interest waning in the use of selective and non-selective COX inhibitors for inflammation-associated diseases due to their well-recognized toxicities to the gastrointestinal and cardiovascular systems, it has been widely proposed that targeting these PGE2 receptors might provide novel therapeutic strategies to treat OAB [74]. Given the fact that the majority of the EP-based OAB therapies were only tested in animal models, the current knowledge about their safety in humans is very limited. A recent phase II clinical study on EP1 selective antagonist ONO-8539 for OAB treatment suggests that it can cause slightly lower incidence of adverse effects when compared to antimuscarinic drug tolterodine (43% vs. 46.6%) [52]. Nevertheless, future studies are required to determine the safety of these PGE2 receptors-targeting compounds in comparison with the current antimuscarinic drugs and β3 adrenergic receptor agonists.
4.1. The case of EP2
Among the four PGE2 receptor subtypes, the Gαs-coupled EP2 has been understudied in animal models of bladder dysfunction largely in that the selective antagonists for this receptor were not available until recently [75]. Using the poorly selective EP antagonist AH-6809 or EP2/EP3 dual agonist ONO-8055 simply cannot provide conclusive evidence to determine whether the EP2 receptor is involved in the pathophysiology of either underactive or overactive bladders [59, 62, 69]. Selective EP2 agonists such as butaprost and ONO-AE1–259 are also not much useful for in vivo studies due to the lack of stability of their prostanoid-like structures [75]. Considering its role in a number of chronic inflammation-associated conditions both in the CNS and periphery [32] as well as its nonnegligent expression in multiple sites of adult bladders from diverse species (https://www.ncbi.nlm.nih.gov/gene/19217) [47, 57, 58], the Gs-coupled EP2 might also contribute to induced PGE2-promoted OAB, where elevated cAMP engaging PKA signaling mediates excitatory cellular response and facilitates the detrusor contraction (Fig. 2). Future studies using highly potent bioavailable selective EP2 antagonists, such as TG4-155, TG6-10-1, and TG6-129 [76, 77], in preclinical models of OAB are needed to determine this possibility (Fig. 3). In addition, the EP2-selective positive allosteric modulators including TG3-95-1 and TG6-270 might also provide useful tools to determine the role of EP2 in detrusor muscle; they enhance the potency of PGE2 on EP2 receptor in various pathological inflamed tissues but usually do not alter the receptor activity in the absence of PGE2 under normal physiological conditions [78, 79]. These chemical probes have diverse pharmacokinetic profiles for in vivo uses [80, 81], and were proven very useful tools to determine the pathophysiological roles of EP2 in a number of inflammation-associated conditions, such as seizure [82–86], Parkinson’s disease [87], retinitis pigmentosa [88], microglial activation and death [89, 90], glioma [34, 91–93], hyperalgesia [94], and brain ischemia [33, 95, 96].
Figure 3. Chemical structures of potential EP-targeting compounds for treatment of OAB.

(A) EP1 antagonists. (B) EP2 antagonists and positive allosteric modulators. (C) EP3 agonists and antagonists. (D) EP4 agonists and antagonists.
4.2. Where do we go from here?
The status (relaxation vs. contraction) of the smooth detrusor muscle of the urinary bladder fundamentally determines the storage and voiding of the urine, in which several families of K+ channels play central roles [40, 56]. It thus has been proposed that modulating these K+ channels directly or indirectly via targeting their upstream regulatory mechanisms might hold a therapeutic potential to control the urinary bladder functions. Using the guinea pig bladder strips, Parajuli et al. provided experimental evidence that PGE2 might increase the spontaneous phasic contraction of the detrusor smooth muscle via the large conductance voltage- and Ca2+-activated K+ (BK) channels, accompanied by an elevation of intracellular Ca2+ levels [39]. Identifying the EP receptor subtype that is directly involved in the PGE2-regulated spontaneous transient BK currents and the subsequent increase of intracellular Ca2+ would be a very interesting topic for the future studies and might provide a molecular target for novel pharmacological manipulation and therapeutic intervention for the underactivity or overactivity of the urinary detrusor.
Given the general concerns about the selectivity of pharmacological compounds, future studies should also be directed to use genetic strategies to validate the findings from pharmacological studies. Answering the question that whether the agonists or antagonists have any effects in OAB animals that lack the potentially targeted EP receptors would help to validate their target engagement, which is an essential step to their development into therapeutic agents for OAB. Moreover, all these EP receptors are also expressed in many other tissues and organ systems, where they may play some important roles under normal physiological conditions. Therefore, developing PGE2 receptors-targeted therapeutics with higher efficacy and safety for the bladder overactivity would require a particular consideration of the undesirable effects on other important normal functions. For instance, the peripherally restricted antagonists TG6–129 for EP2 and DG-041 for EP3 might specifically modify the bladder micturition and bladder nociception via modulating the receptors on peripheral neurons or detrusor muscle cells without affecting the normal functions of the receptors in the CNS.
ACKNOWLEDGEMENTS
We thank Dr. Georgi V. Petkov for insightful suggestions. This work was supported by the National Institutes of Health (NIH) grants R01NS100947 (J.J.) and R21NS109687 (J.J.).
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
CONFLICT OF INTEREST
The authors declare no competing financial interest.
REFERENCES
- [1].Haylen BT, de Ridder D, Freeman RM, Swift SE, Berghmans B, Lee J, Monga A, Petri E, Rizk DE, Sand PK, Schaer GN, An International Urogynecological Association (IUGA)/International Continence Society (ICS) joint report on the terminology for female pelvic floor dysfunction, Int Urogynecol J 21(1) (2010) 5–26. [DOI] [PubMed] [Google Scholar]
- [2].Lightner DJ, Gomelsky A, Souter L, Vasavada SP, Diagnosis and Treatment of Overactive Bladder (Non-Neurogenic) in Adults: AUA/SUFU Guideline Amendment 2019, J Urol 202(3) (2019) 558–563. [DOI] [PubMed] [Google Scholar]
- [3].Irwin DE, Milsom I, Hunskaar S, Reilly K, Kopp Z, Herschorn S, Coyne K, Kelleher C, Hampel C, Artibani W, Abrams P, Population-based survey of urinary incontinence, overactive bladder, and other lower urinary tract symptoms in five countries: results of the EPIC study, Eur Urol 50(6) (2006) 1306–14; discussion 1314–5. [DOI] [PubMed] [Google Scholar]
- [4].Ganz ML, Smalarz AM, Krupski TL, Anger JT, Hu JC, Wittrup-Jensen KU, Pashos CL, Economic costs of overactive bladder in the United States, Urology 75(3) (2010) 526–32, 532 e1–18. [DOI] [PubMed] [Google Scholar]
- [5].Silva I, Magalhaes-Cardoso MT, Ferreirinha F, Moreira S, Costa AF, Silva D, Vieira C, Silva-Ramos M, Correia-de-Sa P, beta3 Adrenoceptor-induced cholinergic inhibition in human and rat urinary bladders involves the exchange protein directly activated by cyclic AMP 1 favoring adenosine release, Br J Pharmacol 177(7) (2020) 1589–1608. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [6].Silva I, Costa AF, Moreira S, Ferreirinha F, Magalhaes-Cardoso MT, Calejo I, Silva-Ramos M, Correia-de-Sa P, Inhibition of cholinergic neurotransmission by beta3-adrenoceptors depends on adenosine release and A1-receptor activation in human and rat urinary bladders, Am J Physiol Renal Physiol 313(2) (2017) F388–F403. [DOI] [PubMed] [Google Scholar]
- [7].Cox L, Cameron AP, OnabotulinumtoxinA for the treatment of overactive bladder, Research and reports in urology 6 (2014) 79–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [8].Drake M, Overactive Bladder, in: L.R.K. Wein Alan J., Partin Alan W., Peters Craig A. (Ed.), Campbell-Walsh Urology, Elsevier, Philadelphia, PA, U.S., 2015, p. 1799. [Google Scholar]
- [9].White N, Iglesia CB, Overactive Bladder, Obstet Gynecol Clin North Am 43(1) (2016) 59–68. [DOI] [PubMed] [Google Scholar]
- [10].Apostolidis A, Jacques TS, Freeman A, Kalsi V, Popat R, Gonzales G, Datta SN, Ghazi-Noori S, Elneil S, Dasgupta P, Fowler CJ, Histological changes in the urothelium and suburothelium of human overactive bladder following intradetrusor injections of botulinum neurotoxin type A for the treatment of neurogenic or idiopathic detrusor overactivity, Eur Urol 53(6) (2008) 1245–53. [DOI] [PubMed] [Google Scholar]
- [11].Tyagi P, Barclay D, Zamora R, Yoshimura N, Peters K, Vodovotz Y, Chancellor M, Urine cytokines suggest an inflammatory response in the overactive bladder: a pilot study, International urology and nephrology 42(3) (2010) 629–35. [DOI] [PubMed] [Google Scholar]
- [12].Wrobel AF, Kluz T, Surkont G, Wlazlak E, Skorupski P, Filipczak A, Rechberger T, Novel biomarkers of overactive bladder syndrome, Ginekol Pol 88(10) (2017) 568–573. [DOI] [PubMed] [Google Scholar]
- [13].Furuta A, Yamamoto T, Suzuki Y, Gotoh M, Egawa S, Yoshimura N, Comparison of inflammatory urine markers in patients with interstitial cystitis and overactive bladder, Int Urogynecol J 29(7) (2018) 961–966. [DOI] [PubMed] [Google Scholar]
- [14].Andersson KE, Wein AJ, Pharmacology of the lower urinary tract: basis for current and future treatments of urinary incontinence, Pharmacol Rev 56(4) (2004) 581–631. [DOI] [PubMed] [Google Scholar]
- [15].Park JM, Yang T, Arend LJ, Schnermann JB, Peters CA, Freeman MR, Briggs JP, Obstruction stimulates COX-2 expression in bladder smooth muscle cells via increased mechanical stretch, Am J Physiol 276(1) (1999) F129–36. [DOI] [PubMed] [Google Scholar]
- [16].Hu VY, Malley S, Dattilio A, Folsom JB, Zvara P, Vizzard MA, COX-2 and prostanoid expression in micturition pathways after cyclophosphamide-induced cystitis in the rat, Am J Physiol Regul Integr Comp Physiol 284(2) (2003) R574–85. [DOI] [PubMed] [Google Scholar]
- [17].Kim JC, Park EY, Seo SI, Park YH, Hwang TK, Nerve growth factor and prostaglandins in the urine of female patients with overactive bladder, J Urol 175(5) (2006) 1773–6; discussion 1776. [DOI] [PubMed] [Google Scholar]
- [18].Kim JC, Park EY, Hong SH, Seo SI, Park YH, Hwang TK, Changes of urinary nerve growth factor and prostaglandins in male patients with overactive bladder symptom, Int J Urol 12(10) (2005) 875–80. [DOI] [PubMed] [Google Scholar]
- [19].Schussler B, Comparison of the mode of action of prostaglandin E2 (PGE2) and sulprostone, a PGE2-derivative, on the lower urinary tract in healthy women. A urodynamic study, Urological research 18(5) (1990) 349–52. [DOI] [PubMed] [Google Scholar]
- [20].Lee T, Hedlund P, Newgreen D, Andersson KE, Urodynamic effects of a novel EP(1) receptor antagonist in normal rats and rats with bladder outlet obstruction, J Urol 177(4) (2007) 1562–7. [DOI] [PubMed] [Google Scholar]
- [21].Hokanson JA, Langdale CL, Sridhar A, Grill WM, OAB without an overactive bladder in the acute prostaglandin E2 rat model, Am J Physiol Renal Physiol 313(5) (2017) F1169–F1177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [22].Yu Y, Nguyen DT, Jiang J, G protein-coupled receptors in acquired epilepsy: Druggability and translatability, Progress in Neurobiology 183 (2019) 101682. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [23].de Jongh R, Grol S, van Koeveringe GA, van Kerrebroeck PE, de Vente J, Gillespie JI, The localization of cyclo-oxygenase immuno-reactivity (COX I-IR) to the urothelium and to interstitial cells in the bladder wall, J Cell Mol Med 13(9B) (2009) 3069–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [24].Park JM, Yang T, Arend LJ, Smart AM, Schnermann JB, Briggs JP, Cyclooxygenase-2 is expressed in bladder during fetal development and stimulated by outlet obstruction, Am J Physiol 273(4) (1997) F538–44. [DOI] [PubMed] [Google Scholar]
- [25].Rouzer CA, Marnett LJ, Cyclooxygenases: structural and functional insights, J Lipid Res 50 Suppl (2009) S29–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [26].Kibar Y, Irkilata HC, Yaman H, Onguru O, Coguplugil AE, Ergin G, Seyrek M, Yildiz O, Dayanc M, The effect of intravesical acetylsalicylic acid instillation on tissue prostaglandin levels after partial bladder outlet obstruction in rabbits, Neurourol Urodyn 30(8) (2011) 1646–51. [DOI] [PubMed] [Google Scholar]
- [27].Lecci A, Birder LA, Meini S, Catalioto RM, Tramontana M, Giuliani S, Criscuoli M, Maggi CA, Pharmacological evaluation of the role of cyclooxygenase isoenzymes on the micturition reflex following experimental cystitis in rats, Br J Pharmacol 130(2) (2000) 331–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [28].Angelico P, Guarneri L, Velasco C, Cova R, Leonardi A, Clarke DE, Testa R, Effect of cyclooxygenase inhibitors on the micturition reflex in rats: correlation with inhibition of cyclooxygenase isozymes, BJU Int 97(4) (2006) 837–46. [DOI] [PubMed] [Google Scholar]
- [29].Cardozo LD, Stanton SL, A comparison between bromocriptine and indomethacin in the treatment of detrusor instability, J Urol 123(3) (1980) 399–401. [DOI] [PubMed] [Google Scholar]
- [30].Sprem M, Milicic D, Oreskovic S, Ljubojevic N, Kalafatic D, Intravesically administered ketoprofen in treatment of detrusor instability: cross-over study, Croat Med J 41(4) (2000) 423–7. [PubMed] [Google Scholar]
- [31].Grosser T, Fries S, FitzGerald GA, Biological basis for the cardiovascular consequences of COX-2 inhibition: therapeutic challenges and opportunities, The Journal of clinical investigation 116(1) (2006) 4–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [32].Jiang J, Dingledine R, Prostaglandin receptor EP2 in the crosshairs of anti-inflammation, anti-cancer, and neuroprotection, Trends Pharmacol Sci 34(7) (2013) 413–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [33].Jiang J, Yu Y, Small molecules targeting cyclooxygenase/prostanoid cascade in experimental brain ischemia: Do they translate?, Med Res Rev n/a(n/a) (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [34].Jiang J, Qiu J, Li Q, Shi Z, Prostaglandin E2 Signaling: Alternative Target for Glioblastoma?, Trends Cancer 3(2) (2017) 75–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [35].Woodward DF, Jones RL, Narumiya S, International Union of Basic and Clinical Pharmacology. LXXXIII: classification of prostanoid receptors, updating 15 years of progress, Pharmacol Rev 63(3) (2011) 471–538. [DOI] [PubMed] [Google Scholar]
- [36].Rahnama’i MS, van Kerrebroeck PE, de Wachter SG, van Koeveringe GA, The role of prostanoids in urinary bladder physiology, Nat Rev Urol 9(5) (2012) 283–90. [DOI] [PubMed] [Google Scholar]
- [37].Tang CH, Yang RS, Fu WM, Prostaglandin E2 stimulates fibronectin expression through EP1 receptor, phospholipase C, protein kinase Calpha, and c-Src pathway in primary cultured rat osteoblasts., J Biol Chem 280(24) (2005) 22907–16. [DOI] [PubMed] [Google Scholar]
- [38].Kamp TJ, Hell JW, Regulation of cardiac L-type calcium channels by protein kinase A and protein kinase C, Circ Res 87(12) (2000) 1095–102. [DOI] [PubMed] [Google Scholar]
- [39].Parajuli SP, Provence A, Petkov GV, Prostaglandin E2 excitatory effects on guinea pig urinary bladder smooth muscle: a novel regulatory mechanism mediated by large-conductance voltage- and Ca2+-activated K+ channels, Eur J Pharmacol 738 (2014) 179–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [40].Petkov GV, Role of potassium ion channels in detrusor smooth muscle function and dysfunction, Nat Rev Urol 9(1) (2011) 30–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [41].Hanna-Mitchell AT, Robinson D, Cardozo L, Everaert K, Petkov GV, Do we need to know more about the effects of hormones on lower urinary tract dysfunction? ICI-RS 2014, Neurourol Urodyn 35(2) (2016) 299–303. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [42].Lorca RA, Prabagaran M, England SK, Functional insights into modulation of BKCa channel activity to alter myometrial contractility, Front Physiol 5 (2014) 289. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [43].Eapen RS, Radomski SB, Gender differences in overactive bladder, Can J Urol 23(Suppl 1) (2016) 2–9. [PubMed] [Google Scholar]
- [44].Wong CM, Tsang SY, Yao X, Chan FL, Huang Y, Differential effects of estrogen and progesterone on potassium channels expressed in Xenopus oocytes, Steroids 73(3) (2008) 272–9. [DOI] [PubMed] [Google Scholar]
- [45].Dawson AP, Calcium signalling: how do IP3 receptors work?, Curr Biol 7(9) (1997) R544–7. [DOI] [PubMed] [Google Scholar]
- [46].Deng M, Ding W, Min X, Xia Y, MLCK-independent phosphorylation of MLC20 and its regulation by MAP kinase pathway in human bladder smooth muscle cells, Cytoskeleton (Hoboken) 68(3) (2011) 139–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [47].Rahnama’i MS, Hohnen R, van Kerrebroeck PE, van Koeveringe GA, Evidence for prostaglandin E2 receptor expression in the intramural ganglia of the guinea pig urinary bladder, J Chem Neuroanat 64–65 (2015) 43–7. [DOI] [PubMed] [Google Scholar]
- [48].Root JA, Davey DA, Af Forselles KJ, Prostanoid receptors mediating contraction in rat, macaque and human bladder smooth muscle in vitro, Eur J Pharmacol 769 (2015) 274–9. [DOI] [PubMed] [Google Scholar]
- [49].Schroder A, Newgreen D, Andersson KE, Detrusor responses to prostaglandin E2 and bladder outlet obstruction in wild-type and Ep1 receptor knockout mice, J Urol 172(3) (2004) 1166–70. [DOI] [PubMed] [Google Scholar]
- [50].Ikeda M, Kawatani M, Maruyama T, Ishihama H, Prostaglandin facilitates afferent nerve activity via EP1 receptors during urinary bladder inflammation in rats, Biomed Res 27(2) (2006) 49–54. [DOI] [PubMed] [Google Scholar]
- [51].Hohnen R, Rahnama’i MS, Van Gink M, Meriaux C, Van Koeveringe GA, Ex vivo modulation of muscarinically induced contractions by PGE2 and an EP1 receptor antagonist in the guinea pig urinary bladder, Bladder 3(2) (2016) e25. [Google Scholar]
- [52].Chapple CR, Abrams P, Andersson KE, Radziszewski P, Masuda T, Small M, Kuwayama T, Deacon S, Phase II study on the efficacy and safety of the EP1 receptor antagonist ONO-8539 for nonneurogenic overactive bladder syndrome, J Urol 191(1) (2014) 253–60. [DOI] [PubMed] [Google Scholar]
- [53].Hayashi M, Kajioka S, Itsumi M, Takahashi R, Shahab N, Ishigami T, Takeda M, Masuda N, Yamaguchi A, Naito S, Actions of cAMP on calcium sensitization in human detrusor smooth muscle contraction, BJU Int 117(1) (2016) 179–91. [DOI] [PubMed] [Google Scholar]
- [54].Morgado M, Cairrao E, Santos-Silva AJ, Verde I, Cyclic nucleotide-dependent relaxation pathways in vascular smooth muscle, Cell Mol Life Sci 69(2) (2012) 247–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [55].Yamamura H, Cole WC, Kita S, Hotta S, Murata H, Suzuki Y, Ohya S, Iwamoto T, Imaizumi Y, Overactive bladder mediated by accelerated Ca2+ influx mode of Na+/Ca2+ exchanger in smooth muscle, Am J Physiol Cell Physiol 305(3) (2013) C299–308. [DOI] [PubMed] [Google Scholar]
- [56].Petkov GV, Central role of the BK channel in urinary bladder smooth muscle physiology and pathophysiology, Am J Physiol Regul Integr Comp Physiol 307(6) (2014) R571–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [57].Rahnama’i MS, van Koeveringe GA, Essers PB, de Wachter SG, de Vente J, van Kerrebroeck PE, Gillespie JI, Prostaglandin receptor EP1 and EP2 site in guinea pig bladder urothelium and lamina propria, J Urol 183(3) (2010) 1241–7. [DOI] [PubMed] [Google Scholar]
- [58].Rahnama’i MS, Biallosterski BT, de Wachter SG, Van Kerrebroeck PE, van Koeveringe GA, The distribution of the prostaglandin E receptor type 2 (EP2) in the detrusor of the guinea pig, Prostaglandins Other Lipid Mediat 99(3–4) (2012) 107–15. [DOI] [PubMed] [Google Scholar]
- [59].Sekido N, Kida J, Mashimo H, Wakamatsu D, Okada H, Matsuya H, Promising Effects of a Novel EP2 and EP3 Receptor Dual Agonist, ONO-8055, on Neurogenic Underactive Bladder in a Rat Lumbar Canal Stenosis Model, J Urol 196(2) (2016) 609–16. [DOI] [PubMed] [Google Scholar]
- [60].Sekido N, Kida J, Otsuki T, Mashimo H, Matsuya H, Okada H, Further characterization of a novel EP2 and EP3 receptor dual agonist, ONO-8055, on lower urinary tract function in normal and lumbar canal stenosis rats, Low Urin Tract Symptoms 12(1) (2020) 99–106. [DOI] [PubMed] [Google Scholar]
- [61].Matsuya H, Sekido N, Kida J, Mashimo H, Wakamatsu D, Okada H, Effects of an EP2 and EP3 Receptor Dual Agonist, ONO-8055, on a Radical Hysterectomy-Induced Underactive Bladder Model in Monkeys, Low Urin Tract Symptoms 10(2) (2018) 204–211. [DOI] [PubMed] [Google Scholar]
- [62].Kinoshita A, Higashino M, Yoshida K, Aratani Y, Kakuuchi A, Hanada K, Takeda H, Naganawa A, Matsuya H, Ohmoto K, Synthesis and evaluation of a potent, well-balanced EP2/EP3 dual agonist, Bioorganic & medicinal chemistry 26(1) (2018) 200–214. [DOI] [PubMed] [Google Scholar]
- [63].Hirata T, Narumiya S, Prostanoid receptors, Chem Rev 111(10) (2011) 6209–30. [DOI] [PubMed] [Google Scholar]
- [64].McCafferty GP, Misajet BA, Laping NJ, Edwards RM, Thorneloe KS, Enhanced bladder capacity and reduced prostaglandin E2-mediated bladder hyperactivity in EP3 receptor knockout mice, Am J Physiol Renal Physiol 295(2) (2008) F507–14. [DOI] [PubMed] [Google Scholar]
- [65].Wu C, Dong X, Liu Q, Zhu J, Zhang T, Wang Q, Hu X, Yang Z, Li L, EP3 activation facilitates bladder excitability via HCN channels on ICCs, Biochem Biophys Res Commun 485(2) (2017) 535–541. [DOI] [PubMed] [Google Scholar]
- [66].Bernard M, Dejos C, Berges T, Regnacq M, Voisin P, Activation of rhodopsin gene transcription in cultured retinal precursors of chicken embryo: role of Ca(2+) signaling and hyperpolarization-activated cation channels, J Neurochem 129(1) (2014) 85–98. [DOI] [PubMed] [Google Scholar]
- [67].Su X, Lashinger ES, Leon LA, Hoffman BE, Hieble JP, Gardner SD, Fries HE, Edwards RM, Li J, Laping NJ, An excitatory role for peripheral EP3 receptors in bladder afferent function, Am J Physiol Renal Physiol 295(2) (2008) F585–94. [DOI] [PubMed] [Google Scholar]
- [68].Jugus MJ, Jaworski JP, Patra PB, Jin J, Morrow DM, Laping NJ, Edwards RM, Thorneloe KS, Dual modulation of urinary bladder activity and urine flow by prostanoid EP3 receptors in the conscious rat, Br J Pharmacol 158(1) (2009) 372–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [69].Xue R, Jia Z, Kong X, Pi G, Ma S, Yang J, Effects of PGE2 EP3/EP4 receptors on bladder dysfunction in mice with experimental autoimmune encephalomyelitis, Am J Physiol Renal Physiol 305(12) (2013) F1656–62. [DOI] [PubMed] [Google Scholar]
- [70].Beppu M, Araki I, Yoshiyama M, Du S, Kobayashi H, Zakoji H, Takeda M, Bladder outlet obstruction induced expression of prostaglandin E2 receptor subtype EP4 in the rat bladder: a possible counteractive mechanism against detrusor overactivity, J Urol 186(6) (2011) 2463–9. [DOI] [PubMed] [Google Scholar]
- [71].Chuang YC, Yoshimura N, Huang CC, Wu M, Tyagi P, Chancellor MB, Expression of E-series prostaglandin (EP) receptors and urodynamic effects of an EP4 receptor antagonist on cyclophosphamide-induced overactive bladder in rats, BJU Int 106(11) (2010) 1782–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [72].Chuang YC, Tyagi P, Huang CC, Chancellor MB, Yoshimura N, Mechanisms and urodynamic effects of a potent and selective EP4 receptor antagonist, MF191, on cyclophosphamide and prostaglandin E2-induced bladder overactivity in rats, BJU Int 110(10) (2012) 1558–64. [DOI] [PubMed] [Google Scholar]
- [73].Mizoguchi S, Wolf-Johnson AS, Ni J, Mori K, Suzuki T, Takaoka E, Mimata H, DeFranco DB, Wang Z, Birder LA, Yoshimura N, The role of prostaglandin and E series prostaglandin receptor type 4 receptors in the development of bladder overactivity in a rat model of chemically induced prostatic inflammation, BJU Int 124(5) (2019) 883–891. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [74].Dobrek Łukasz TPJ, The role of prostanoids in the urinary bladder function and a potential use of prostanoid-targeting pharmacological agents in bladder overactivity treatment., Acta Pol Pharm 72(1) (2015) 13–19. [PubMed] [Google Scholar]
- [75].Markovic T, Jakopin Z, Dolenc MS, Mlinaric-Rascan I, Structural features of subtype-selective EP receptor modulators, Drug discovery today 22(1) (2017) 57–71. [DOI] [PubMed] [Google Scholar]
- [76].Jiang J, Ganesh T, Du Y, Quan Y, Serrano G, Qui M, Speigel I, Rojas A, Lelutiu N, Dingledine R, Small molecule antagonist reveals seizure-induced mediation of neuronal injury by prostaglandin E2 receptor subtype EP2, Proc Natl Acad Sci U S A 109(8) (2012) 3149–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [77].Ganesh T, Jiang J, Shashidharamurthy R, Dingledine R, Discovery and characterization of carbamothioylacrylamides as EP2 selective antagonists, ACS Med Chem Lett 4(7) (2013) 616–621. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [78].Jiang J, Ganesh T, Du Y, Thepchatri P, Rojas A, Lewis I, Kurtkaya S, Li L, Qui M, Serrano G, Shaw R, Sun A, Dingledine R, Neuroprotection by selective allosteric potentiators of the EP2 prostaglandin receptor, Proc Natl Acad Sci U S A 107(5) (2010) 2307–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [79].Jiang J, Van TM, Ganesh T, Dingledine R, Discovery of 2-Piperidinyl Phenyl Benzamides and Trisubstituted Pyrimidines as Positive Allosteric Modulators of the Prostaglandin Receptor EP2, ACS chemical neuroscience 9(4) (2018) 699–707. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [80].Ganesh T, Jiang J, Yang MS, Dingledine R, Lead optimization studies of cinnamic amide EP2 antagonists, J Med Chem 57(10) (2014) 4173–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [81].Ganesh T, Jiang J, Dingledine R, Development of second generation EP2 antagonists with high selectivity, European journal of medicinal chemistry 82 (2014) 521–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [82].Jiang J, Yang MS, Quan Y, Gueorguieva P, Ganesh T, Dingledine R, Therapeutic window for cyclooxygenase-2 related anti-inflammatory therapy after status epilepticus, Neurobiol Dis 76 (2015) 126–136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [83].Du Y, Kemper T, Qiu J, Jiang J, Defining the therapeutic time window for suppressing the inflammatory prostaglandin E2 signaling after status epilepticus, Expert Rev Neurother 16(2) (2016) 123–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [84].Jiang J, Yu Y, Kinjo ER, Du Y, Nguyen HP, Dingledine R, Suppressing pro-inflammatory prostaglandin signaling attenuates excitotoxicity-associated neuronal inflammation and injury, Neuropharmacology 149 (2019) 149–160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [85].Nagib MM, Yu Y, Jiang J, Targeting prostaglandin receptor EP2 for adjunctive treatment of status epilepticus, Pharmacol Ther 209 (2020) 107504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [86].Yu Y, Jiang J, COX-2/PGE2 axis regulates hippocampal BDNF/TrkB signaling via EP2 receptor after prolonged seizures, Epilepsia Open 5(3) (2020) 418–431. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [87].Kang X, Qiu J, Li Q, Bell KA, Du Y, Jung DW, Lee JY, Hao J, Jiang J, Cyclooxygenase-2 contributes to oxidopamine-mediated neuronal inflammation and injury via the prostaglandin E2 receptor EP2 subtype, Sci Rep 7(1) (2017) 9459. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [88].Yang W, Xiong G, Lin B, Cyclooxygenase-1 mediates neuroinflammation and neurotoxicity in a mouse model of retinitis pigmentosa, Journal of neuroinflammation 17(1) (2020) 306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [89].Fu Y, Yang MS, Jiang J, Ganesh T, Joe E, Dingledine R, EP2 Receptor Signaling Regulates Microglia Death, Mol Pharmacol 88(1) (2015) 161–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [90].Quan Y, Jiang J, Dingledine R, EP2 receptor signaling pathways regulate classical activation of microglia, J Biol Chem 288(13) (2013) 9293–302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [91].Jiang J, Dingledine R, Role of prostaglandin receptor EP2 in the regulations of cancer cell proliferation, invasion, and inflammation, J Pharmacol Exp Ther 344(2) (2013) 360–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [92].Qiu J, Li Q, Bell KA, Yao X, Du Y, Zhang E, Yu JJ, Yu Y, Shi Z, Jiang J, Small-molecule inhibition of prostaglandin E receptor 2 impairs cyclooxygenase-associated malignant glioma growth, Br J Pharmacol 176(11) (2019) 1680–1699. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [93].Jiang J, Methods and compositions for the treatment of gliomas, University of Cincinnati, US patent 10758516, 2020.
- [94].Greaves E, Horne AW, Jerina H, Mikolajczak M, Hilferty L, Mitchell R, Fleetwood-Walker SM, Saunders PT, EP2 receptor antagonism reduces peripheral and central hyperalgesia in a preclinical mouse model of endometriosis, Sci Rep 7(1) (2017) 44169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [95].Li L, Yu Y, Hou R, Hao J, Jiang J, Inhibiting the PGE2 Receptor EP2 Mitigates Excitotoxicity and Ischemic Injury, ACS Pharmacol Transl Sci 3(4) (2020) 635–643. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [96].Li L, Sluter MN, Yu Y, Jiang J, Prostaglandin E receptors as targets for ischemic stroke: Novel evidence and molecular mechanisms of efficacy, Pharmacol Res (2020) 105238. [DOI] [PMC free article] [PubMed]
