Skip to main content
American Journal of Translational Research logoLink to American Journal of Translational Research
. 2024 Dec 15;16(12):7248–7261. doi: 10.62347/YZDZ1428

Adenosine signaling: a potential therapeutic target for psychogenic erectile dysfunction

Kai-Yang Xue 1, Ming-Xi Yan 1, Zhou Zhu 1, Jin Cui 1,2
PMCID: PMC11733318  PMID: 39822506

Abstract

Therapeutic modalities for psychogenic erectile dysfunction (PED) are poorly targeted because of the lack of specific pathological features. The common symptoms of PED include psychological stress-related negative emotions and erectile dysfunction. Exploring their common therapeutic targets is helpful in the development of effective PED treatment strategies. Adenosine locally acts as a vasodilator or neuromodulator in the penis and promotes erection. Recent studies have demonstrated that adenosine (ADO) signaling is also involved in psychological stress. Herein, we review the pathogenesis of PED and the interaction between ADO and the erection regulator nitric oxide (NO) in brain and penile tissues. In addition, we summarize the regulatory role of ADO signal transduction in penile erection, psychological stress and negative emotions. Through our study, we found that ADO is involved in psychological stress and erectile events by combining adenosine A1 receptors (A1R) and adenosine A2A receptors (A2AR). The application of A1R selective agonists may promote erection and improve psychological state.

Keywords: Psychogenic erectile dysfunction, psychological stress, purinergic signaling, adenosine

Introduction

Erectile dysfunction (ED), which refers to the inability of men to maintain penile erection to achieve satisfactory sexual performance [1], is a global health problem. According to statistics, this condition affects approximately 150 million men worldwide, and its prevalence rate increases with age. It is predicted that by 2025, up to 322 million patients will have ED worldwide [2]. An epidemiological survey conducted on almost 100,000 people in 8 countries showed that Italy (48.6%) had the highest prevalence of ED, followed by China (41.6%) and then Brazil (37.2%) [3]. This condition not only negatively affects marriage and family harmony but may also be an early symptom and a risk factor for cardiovascular and peripheral vascular diseases [4]. Therefore, active early diagnosis and treatment are particularly important.

Clinically, ED can be categorized into three subtypes based on its etiology: organic, psychogenic, and mixed [5]. With the rapid pace of society, psychogenic ED (PED) has gradually become the focus of social attention owing to its high incidence and complex pathogenesis [6]. Unlike organic ED (OED), PED lacks specific pathological features and is generally associated with several factors such as psychological stress, spousal relationship, and lack of sexual knowledge. Most patients experience negative emotions such as anxiety, depression, and fear [7,8]. PED has been found to account for 13%-85.2% of ED patients aged <40 years old [9,10]. Since 2014, the number of adolescent patients with ED has increased by 31-fold, mostly owing to psychological factors [11]. Notably, OED can be used as a source of stress to enable the patient to simulate the aforementioned negative emotions to transform into mixed ED. Thus, psychological factors are considered to be involved in the pathogenesis of ED [12].

Penile erection is mainly regulated by the cyclic nucleotide pathway, of which the NO-cyclic guanosine monophosphate (cGMP) signaling pathway is the main regulatory pathway. NO regulates cGMP synthesis through guanylate cyclase and induces cavernous smooth muscle diastole to promote penile erection, whereas phosphodiesterase type 5 (PDE5) in penile tissues can hydrolyze cGMP and cause penile weakness [13,14]. Phosphodiesterase type 5 inhibitors (PDE5I), such as tadalafil and sildenafil citrate, are the first-line treatment for ED owing to their ability to inhibit cGMP hydrolysis and improve erectile function [15]. Although PDE5I has been reported to rarely cause adverse reactions, such as headache, dyspepsia, and hypotension, its overall efficacy for patients with ED is satisfactory [16,17]. Previous studies have confirmed that PDE5I has a therapeutic effect on PED [18]; however, psychological stressors and stress-related negative emotions cannot be alleviated and treated with oral PDE5I. Long-term use of PDE5I may overlook the psychological issues of patients with ED. In 2013, the Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition (DSM-5) [19], emphasized the importance of psychotherapy for ED treatment. Therefore, at this stage, the clinical treatment plan for PED mostly involves combination therapy with oral PDE5I and psychotherapy [18]. However, adherence to this therapy is significantly affected by the long duration of psychotherapy, high costs, and the fact that most patients with PED are not psychotherapeutically aware [20-22]. Therefore, it is necessary to identify drugs that can regulate psychological states and promote penile erection.

In addition to NO/cGMP signaling pathway-related molecules, other molecules released by neurons, endothelial cells, and smooth muscle cells are involved in penile erection. Purinergic signaling is an important extracellular signal that is involved in physiological and pathological processes and has recently gained extensive attention. Studies have demonstrated that adenosine (ADO) and adenosine receptors (ARs) are closely associated with penile erection, psychological stress, emotion regulation, and other processes [23-25] and can be used to treat PED. This review summarizes the role of ADO signaling transduction in the aforementioned processes to determine the candidate drugs for PED.

Physiological mechanism of penile erection

Penile erection is a psychophysiological process that is initiated and dominated by the brain [26]. Previous studies have reported many brain regions related to penile erection, such as the prefrontal, parietal, and cingulate cortices; brain regions inhibiting erection, such as the caudate nucleus, amygdala, and hypothalamus; and brain regions that promote erection. The paraventricular nucleus of the hypothalamus is currently considered the control center of erection [27,28]. The brain releases glutamate (Glu), dopamine (DA), 5-hydroxytryptamine (5-HT), gamma-amino-butyric acid (GABA), and other neurotransmitters to the spinal cord after processing and integrating the signals of visual, auditory, and tactile stimuli. Neurotransmitters form nerve impulses that inhibit the T10-L2 lateral horn sympathetic nerve centers and the hypogastric nerve, excite the S2-S4 lateral horn parasympathetic nerve centers and pelvic splanchnic nerve, and mediate the synthesis and release of NO by neuronal nitric oxide synthase in the nonadrenergic-noncholinergic (NANC) nerve endings of the corpus cavernosum (CC) and endothelial nitric oxide synthase (eNOS) in the endothelial cells of the vascular endothelium. NO activates guanylate cyclase to produce cGMP, which activates protein kinase G (PKG), inducing hyperpolarization of smooth muscle cell membranes and relaxation of CC smooth muscles to promote erection [16,29,30].

Pathogenesis of PED

Previous studies have confirmed that patients with PED have anomalous brain structure and function [31,32]. As shown in Figure 1, the brain exposed to psychological stress initiates a series of neuroendocrine responses to block sexual activity. Emotion-related brain regions, such as the prefrontal cortex, amygdala, and hippocampus, are involved in psychological stress sensing and the generation and transmission of nerve impulses to the paraventricular nucleus of the hypothalamus. The paraventricular nucleus triggers sequential stress responses mediated by the autonomic nervous system and hypothalamic-pituitary-adrenal (HPA) axis [33]. On the one hand, the tension of the sympathetic nervous system (SNS) rapidly increases under stress, resulting in the release of a large amount of norepinephrine (NE). At this time, the expression of NO/nitric oxide synthase (NOS) is downregulated in the NANC and vascular endothelium, and cGMP production is reduced, resulting in penile weakness [34,35]. However, the release of large amounts of Glu during stress causes the accumulation of NO in the brain and slow activation of the HPA axis, which is characterized by increased secretion of corticotropin-releasing and adrenocorticotropic hormones. Eventually, this results in the release of a large amount of cortisol (COR). COR exerts a strong negative effect on brain areas related to emotion regulation, which may be the basis of negative emotion. Abnormal secretion of COR causes circadian rhythm disorders, further promoting the tension of SNS and the release of NE, resulting in penile weakness [36]. Furthermore, the body preferentially synthesizes COR during stress, leading to a compensatory decrease in the metabolic levels of dehydroepiandrosterone (DHEA), a precursor of testosterone (T). T regulates the formation and degradation of cGMP; thus, the obstruction of T production can directly cause ED. A persistently low T expression leads to the suppression of hypothalamic-pituitary-testicular axis function, which is characterized by decreased release of gonadotropin-releasing hormone and gonadotropin. This eventually results in hypothalamic damage and the formation of a malignant stress circuit [37-39].

Figure 1.

Figure 1

Pathogenesis of PED under psychological stress. “→” represent HPA/HPG axes, “…” represent ANS. When the body feels stress, the hypothalamus initiates a stress response, and the ANS is activated rapidly, which is manifested as SNS excitation and PSNS inhibition. HPA and HPG axes are activated slowly, which is manifested as HPA axis hyperexcitability and HPG axis inhibition, ultimately leading to ED and negative emotions. Abbreviations: ANS, Autonomic Nervous System; ATCH, adrenocor ticotropic hormore; CRH, corticotropin releasing hormone; Gn, gonadotropins; GnRH, gonadotropin-releasing hormone; HPG, hypothalamic-pituitary-gonadal; PSNS, parasympathetic nervous system.

Overview of ADO signaling

ADO is a precursor and metabolite of adenine nucleotides that is widely expressed in various systems of the body. It is mainly released by nerve endings and glial cells but is also generated by adenosine triphosphate (ATP) through the double hydrolysis of extracellular nucleosidases, such as ectonucleide triphosphate diphosphohydrolase-1 (CD39) and ecto-5’-nucleotidase (CD73). Simultaneously, intracellular ADO can also be transported outside the cell through nucleoside transporters on the cell membrane. Figure 2 shows the source and metabolism of ADO. The physiological role of ADO was first described by Drury and Szent-Gyorgyi [40], and it has since been shown to be involved in multiple physiological and pathological processes in vivo by binding to its specific receptor [41]. ARs currently have four subtypes, A1R, A2AR, adenosine A2B receptors (A2BR), and adenosine A3 receptors (A3R), which belong to the G protein-coupled receptor superfamily and have different distributions, transduction mechanisms, and functions [42]. In the physiological state, the concentration of ADO is maintained at a low level (1-2 μmol/L), and it mainly binds to high-affinity A1R and A2AR, whereas in pathological states, such as ischemia, inflammation, and stress, the extracellular concentration of ADO rapidly increases to 100-1,000 μmol/L, and it stimulates low-affinity A2BR and A3R [43].

Figure 2.

Figure 2

Overview of ADO Signaling. Intracellularly, ADO is mainly generated by AMP under the catalysis of Cyto 5’NT, and can also be produced by the hydrolysis of AdoHcy by SAHH. Most of the generated ADO is phosphorylated to AMP by ADK, thus participating in ATP synthesis and completing the recirculation of ADO, and a small portion of ADO is catalyzed by ADA to inosine, or transported to the extracellular space through nucleoside transport proteins on the cell membrane. Extracellularly, ATP becomes AMP after dephosphorylation catalyzed by CD39, and AMP becomes ADO after dephosphorylation catalyzed by CD73, which binds to specific receptors A1R, A2AR, A2BR, and A3R to participate in the physiological and pathological processes of the organism. In addition, ADO regulates cAMP synthesis through AC. Abbreviations: AC, adenylate cyclase; ADA, adenosine deaminase; ADK, adenosine kinase; AdoHcy, S-adenosyl-homocysteine; AMP, adenosine monophosphate; Cyto 5’NT, cytosolic 5’-nucleotidase; SAHH, S-adenosyl-homocysteine hydrolase.

Distribution of ARs in brain and penile tissues

Four subtypes of ARs have different regions and density distributions in the brain. A1R has a high-density distribution in the cerebral cortex, hippocampus, cerebellum, thalamus, brain stem, and other regions and is concentrated in the synaptic part of neurons [44]. A2AR is mainly located in DA-enriched regions, including the striatum, nucleus ambiguus, pallidum, and olfactory bulb; its mRNAs are also distributed in the hippocampus, hypothalamus, amygdala, and choroid plexus epithelial cells, as detected via Reverse transcription-Polymerase chain reaction (RT-PCR) [45-47]. In addition, a previous study confirmed that A2AR exists in the whole nerve axis in rats via immunohistochemistry [48,49]. Meanwhile, A2BR is mainly distributed in the Cornu ammonis 1 (CA1) and Cornu ammonis 3 (CA3) regions of the hippocampus, with a few in the thalamus, lateral ventricles, and striatum [43]. Finally, A3R has a high-density distribution in the striatum, olfactory bulb, auditory nerve, hippocampus, hypothalamus, thalamus, and cerebellum but a low-density distribution in the cortex and amygdala [44]. Furthermore, it is mainly expressed in the synaptic terminals of neurons, and its distribution in the presynaptic membrane of hippocampal neurons is more abundant than that in the postsynaptic membrane, suggesting that A3R plays a pivotal role in the release of presynaptic neurotransmitters [50-52]. Glial cells are higher in proportion than neuronal cells in the brain, and the four AR subtypes have been detected in microglia and astrocytes [53,54]. Compared with brain tissues, few studies have examined the expression profile of ARs in penile tissues. It is currently believed that A1R is mainly expressed in the cavernous and dorsal nerves of the penis. A2BR is the main subtype expressed in the smooth muscle cells of the cavernous body of the penis, whereas A2AR has low expression in these cells. Notably, the A3R expression in penile tissues is difficult to detect [55-57]. The aforementioned distribution of ARs provides a material basis for the biological role of ADO signaling in the pathological process of PED.

Interaction between ADO and NO in brain and penile tissues

The production mechanism of NO is as follows: Glu in the synaptic gap binds to N-methyl-D-aspartate receptors in the postsynaptic membrane, resulting in the inward flow of Ca2+. Ca2+ and calmodulin activate NOS and catalyze the conversion of L-arginine to NO and citrulline [58]. Interestingly, ADO and NO share many similarities [59,60], for example, 1) both are recognized as vasodilators and neurotransmitters, 2) both have extremely short half-lives (<10 s), and 3) both exert their physiological effects through nucleotide second messengers. Because both ADO and NO have broad and important biological importance and the latter is considered to be the major regulator of penile erection [13], it is imperative to focus on their interactions between them.

NO regulates cGMP synthesis through guanylate cyclase, ADO regulates cyclic adenosine monophosphate (cAMP) synthesis through adenylate cyclase, and cAMP and cGMP activate downstream protein kinase A (PKA) and PKG, respectively, and then phosphorylate downstream targets to jointly regulate ion channels [55]. Thus, the cascade signaling pathways of ADO/cAMP and NO/cGMP restrict each other, which is the basis of the interaction between ADO and NO [61,62]. ADO and NO in the brain interact during neural and vascular regulations. During sustained stress, the brain releases large amounts of Glu, which results in NOS overactivation and a dramatic increase in NO synthesis [63,64]. Moderate amounts of NO exert neuroprotective effects. However, an excessive amount of NO induces neurotoxicity, which can activate the HPA axis and increase the expression of COR, directly damaging the erectile center and emotion-related brain regions [65]. Therefore, the inhibition of NO accumulation in brain regions is expected to improve mental state and erectile function. The inhibitory and facilitatory effects of ADO on neurotransmission are mainly mediated by A1R and A2AR [66,67]. Studies have found that the brain releases endogenous ADO during stress while reducing NO levels and eNOS activities [68-70]. NO in the basal ganglia, striatum, and hippocampus can promote ADO release and antagonize A1R activity, which may help counteract NO neurotoxicity [43]. In terms of vascular regulation, several dynamic exercise experiments have shown that NO and ADO exert common effects on vascular blood supply and oxygen delivery. When NO is sufficient, the vasodilatory effect of ADO is weakened; otherwise, the vasodilatory effect is enhanced [71-73].

ADO in penile tissues can also indirectly regulate cGMP synthesis by activating ARs to promote NO production [74]. Using genetic and pharmacological tools, it has been shown that ADO can induce cGMP production in mouse CC via A2BR. However, it cannot promote cGMP production in CC smooth muscle cells, suggesting that ADO-induced NO originates from endothelial cells rather than muscle cells [55]. Electrophysiological experiments have shown that cGMP produced by electrically stimulated CC is partially dependent on the A2BR-mediated NO pathway, suggesting that endogenous ADO is an important molecule in penile erection [56]. Further experiments have confirmed that blood flow shear stress induces endogenous ADO production in endothelial cells and increases eNOS phosphorylation via A2BR activation of the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) signaling pathway, which promotes NO production. The AKT and eNOS phosphorylation levels in the penises of A2BR-knockout mice are both downregulated in wild-type mice, which exhibit impaired erectile function, suggesting that endogenous ADO induces NO production by activating A2BR [75]. Several in vitro experiments confirmed the aforementioned results indicating that the NO signaling pathway in endothelial cells is also involved in the exogenous ADO-mediated CC diastolic process [76-79].

ADO signaling and CC diastole

Both endogenous and exogenous ADO can regulate cAMP synthesis and promote penile erection. CD73-deficient mice exhibit reduced endogenous ADO expression, leading to impaired erectile function [80]. Contrarily, ADA-deficient mice demonstrate excessive accumulation of endogenous ADO, inducing abnormal erections, defined as persistent erection lasting at least 4 h in the absence of sexual stimulation [81]. As a phosphodiesterase inhibitor (PDEI), caffeine intake can upregulate cGMP and cAMP levels in the CC and promote penile erection [82]. The diastolic effect of exogenous ADO on CC has been observed in humans, rats, dogs, and rabbits [83-85], with varying degrees of diastolic effect among individuals. After ADO treatment, the diastolic effect of CC in patients with ED and diabetes was better than that in patients with ED and no diabetes, and the erectile function of both groups were better than ED rats [84]. Furthermore, the diastolic effect of ADO on CC in 24-month-old rabbits was superior to that in 3- and 7-month-old rabbits [85].

As shown in Figure 3, ADO signaling promotes erection by acting as a vasodilator and neuromodulator [86]. ADO acts as a negative feedback regulator of sympathetic neurotransmitters through A1R, which binds to A1R enriched on neuronal cells and inhibits NE release dose-dependent manner. High expression of NE following SNS excitation elicits CC smooth muscle contraction, whereas A1R activation inhibits the NE-induced contractile response of CC smooth muscles in mice, which in turn promotes erection [55,87]. Meanwhile, A2AR activation induces CC smooth muscle diastole, and it has been suggested that the CC diastolic effect exerted by ADO binding to A2AR is completely independent of the NO/cGMP pathway [88]. The ADO analog 5’-NN-ethylcarboxamide adenosine (NECA) is known to be a nonselective agonist of ARs, and drugs that excite A2AR can exert approximately 50% of the diastolic effect compared with NECA. Further in vitro experiments have confirmed that the diastolic effect exerted by NECA in preconstriction-treated human CC is significantly inhibited by the A2BR inhibitor, but this inhibitor does not significantly influence the diastolic effect exerted by drugs that previously excited A2AR [89]. Therefore, both A2AR and A2BR are involved in ADO-induced erections, which may be associated with the regulatory effect of ADO on penile arterial vascular tension after A2AR and A2BR activation [74].

Figure 3.

Figure 3

The role and mechanism of ARs in psychological stress and penile erection. +, relieve psychological stress and negative emotions or promote erection; -, promote psychological stress and negative emotions.

A2BR plays a pivotal role in ADO-mediated erection [81,90-94]. The NO/cGMP signaling pathway for erection is T-dependent. Low T levels inhibit the AKT/eNOS/cGMP signaling pathway and cAMP generation in rat CC by downregulating A2BR expression, ultimately leading to ED [95]. Excessive ADO in CC has also been suggested to induce abnormal penile erections via A2BR activation [56,96,97]. Therefore, strategies to promote or block A2BR activation may be beneficial for treating ED or abnormal erection. Furthermore, ADO combined with A2BR can exert a neuromodulatory effect similar to that of A1R (Figure 3). It can relax CC by inhibiting the nerve transmission of excitatory SNS to the rabbit penis. This effect is more pronounced in CC with intact endothelium [98]. Few studies have examined the involvement of A3R in erection, and the active site on A3R may not be involved in the ADO-induced erectile process. Notably, local or oral administration of A3R allosteric regulator has improved erectile function of ED rats [99]. A new ligand acting on the A3R allosteric regulatory site may exert special effects that differ from those of orthosteric ligands. Future studies focusing on this ligand are warranted.

ADO signaling, psychological stress, and negative emotions

Previous studies established myocardial ischemia models in patients with heart failure induced by psychological stress and ADO administration. The results indicated that the biological indicators of the two groups were highly consistent, suggesting that ADO administration simulates psychological stress responses [100]. Early findings in rodents indicated that psychological stress responses induced abnormal ADO signaling and that acute and chronic psychological stress responses induced by restraints in rats contributed to increased ATP hydrolysis and elevated ADO levels [101-103]. To simulate changes in ADO during human stress, continued exploration was conducted using zebrafish, which are genetically highly similar to humans. A zebrafish model of acute psychological stress was constructed by restricting activities. It was observed that the changes in ATP hydrolysis and ADO levels in zebrafish were consistent with those in rodents [104]. The same result was obtained in a zebrafish model of chronic psychological stress [105]. Psychological stress can also affect AR activity. For example, the activity of astrocyte A1R in the prefrontal cortex and hippocampus of a mouse model of social pressure-induced chronic stress was significantly inhibited, and the mouse exhibited a depression-like behavior [106]. Mouse model of chronic stress established by exposing it to continuous unpredictable stress exhibited anxiety-like behaviors, accompanied by decreased synaptic plasticity and increased A2AR expression at the end of allergic neurons in the hippocampus [107]. In conclusion, psychological stress can cause high ADO and A2AR expressions and low A1R expression in brain regions. This change is considered to be a strategy for the body to rebuild balance after stress, but the specific mechanism remains unclear [104].

ADO, A1R, and A2AR can also mediate psychological stress and stress-related negative emotions. Studies have found that A1R-knockout mice exhibit aggravated anxiety- and depression-like behaviors and are resistant to the antidepressant effect of sleep deprivation [108]; however, these effects are relieved after A1R activation [109-111]. Astrocyte activation can upregulate the expression of extracellular ADO and activate A1R to mediate the elimination of fear, which can be blocked by A1R antagonists [112]. Therefore, A1R activation can alleviate stress-related negative emotions, which may be associated with neural regulation. A1R can also inhibit the release of presynaptic Glu and increase the permeability of postsynaptic K+ channels, thus inhibiting nerve excitation to regulate mental state [45,113]. Furthermore, A1R activation can upregulate the expression of synaptophysin Homer1a in the prefrontal cortex and hippocampus, which is a common target of antidepressant therapy drugs, such as fluoxetine and ketamine [108,114,115]. A1R agonists are currently used to treat stress-related negative emotions; however, due to some complex reactions, the development of A1R-positive allosteric modifiers as powerful anti-anxiety drugs is being considered [116].

Knockout or inhibition of A2AR in hippocampal neurons can prevent the recovery of fear memory following stress [117]. The A2AR antagonist reversed the depression-like behavior induced by chronic psychological stress [107] and was proven to be effective against stress injury caused by maternal separation [118]. As a nonspecific AR antagonist, caffeine can reduce A2AR mRNA expression in caudate nucleus neurons and regulate stress-related anxiety-like behaviors [119]. Contrarily, the A2AR agonists can aggravate anxiety in mice [120], and an abnormally high A2AR expression in the lateral septum can lead to depression-like behaviors [121]. Therefore, inhibiting A2AR activity is another strategy to improve the mental state and relieve negative emotions, and its mechanism may be associated with the following neuroinflammation control and neuroendocrine regulation [122]. First, A2AR activation can upregulate the expression of cysteinyl aspartate-specific proteinase-1 (caspase-1) and interleukin-1β (IL-1β), leading to a neuroinflammatory response [123]. Second, A2AR induces neurotoxicity by promoting Glu release and inhibiting 5-HT release [124]. In addition, A2AR mediates the ADO and CD73 regulation of amygdala neuronal excitability and synaptic plasticity [125]. When the activity of A2AR is inhibited by caffeine, the expression of brain-driven neurotrophic factor in the hippocampus is upregulated, suggesting that inhibiting A2AR activity can exert a neuroprotective effect [119]. Furthermore, the inhibition of A2AR activity is associated with the recovery of HPA axis activity. Plasma corticosterone levels returned to the levels present in normal circadian rhythm after blocking A2AR, indicating that A2AR is directly involved in the stress response [118].

Conclusion

Although the previous two reviews [24,126] have clearly reported that ADO signaling is an important target for the treatment of ED, only the local effects of ADO in penile tissues have been highlighted. Penile erection is a complex event initiated by the brain, and PED is caused by psychological stress. In summary, the role of ADO signaling transduction in brain and penile tissues is important for the treatment of PED.

Our review found that the ADO/cAMP signaling pathway interacts with the dominant pathway of erection, namely, NO/cGMP, in both brain and penile tissues. In the brain, ADO and A2AR are expressed at high levels under psychological stress, whereas A1R is expressed at low levels. A1R signaling activation improves negative emotions via neuromodulation, whereas A2AR signaling inhibition improves negative emotions via neuroendocrine regulation and neuroinflammatory control. In penile tissues, ADO, A1R, A2AR, and A2BR were expressed at low levels in the ED state, A1R and A2BR activation negatively regulated sympathetic neurotransmitters via feedback, and A2AR and A2BR activation relaxed CC vascular smooth muscle cells. ADO combined with A1R and A2AR participating in psychological stress and erectile events is expected to be a candidate target for PED activation. However, the expression levels of ADO and A2AR in the brain and penis tissues showed a reverse trend, and the specific mechanism needs to be further explored. There is a similar expression trend of A1R in brain and penile tissues, and the use of A1R selective agonists may improve both erectile function and psychological status. As a PDEI and A2AR antagonist, caffeine may be beneficial for PED treatment. Notably, caffeine can also antagonize A1R and A2BR activities, and further research is warranted to support this conclusion. The A1R and A3R allosteric regulatory sites also demonstrate the potential for PED treatment and are worthy of further research in the future.

Acknowledgements

This work was supported by the Natural Science Foundation of China [NSFC 82360971].

Disclosure of conflict of interest

None.

References

  • 1.Shamloul R, Ghanem H. Erectile dysfunction. Lancet. 2013;381:153–165. doi: 10.1016/S0140-6736(12)60520-0. [DOI] [PubMed] [Google Scholar]
  • 2.Burnett AL, Nehra A, Breau RH, Culkin DJ, Faraday MM, Hakim LS, Heidelbaugh J, Khera M, McVary KT, Miner MM, Nelson CJ, Sadeghi-Nejad H, Seftel AD, Shindel AW. Erectile dysfunction: AUA guideline. J Urol. 2018;200:633–641. doi: 10.1016/j.juro.2018.05.004. [DOI] [PubMed] [Google Scholar]
  • 3.Goldstein I, Goren A, Li VW, Tang WY, Hassan TA. Epidemiology update of erectile dysfunction in eight countries with high burden. Sex Med Rev. 2020;8:48–58. doi: 10.1016/j.sxmr.2019.06.008. [DOI] [PubMed] [Google Scholar]
  • 4.Baccino D, Amico AF, Di Fusco SA, Nardi F, Colivicchi F. Erectile dysfunction and cardiovascular risk. G Ital Cardiol (Rome) 2023;24:628–635. doi: 10.1714/4068.40531. [DOI] [PubMed] [Google Scholar]
  • 5.MacDonald SM, Burnett AL. Physiology of erection and pathophysiology of erectile dysfunction. Urol Clin North Am. 2021;48:513–525. doi: 10.1016/j.ucl.2021.06.009. [DOI] [PubMed] [Google Scholar]
  • 6.Bodie JA, Beeman WW, Monga M. Psychogenic erectile dysfunction. Int J Psychiatry Med. 2003;33:273–293. doi: 10.2190/NHV6-3DYB-X51G-4BVM. [DOI] [PubMed] [Google Scholar]
  • 7.Ciaccio V, Di Giacomo D. Psychological factors related to impotence as a sexual dysfunction in young men: a literature scan for noteworthy research frameworks. Clin Pract. 2022;12:501–512. doi: 10.3390/clinpract12040054. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Noh Y, Kim M, Hong SH. Identification of emotional spectrums of patients taking an erectile dysfunction medication: ontology-based emotion analysis of patient medication reviews on social media. J Med Internet Res. 2023;25:e50152. doi: 10.2196/50152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Zou Z, Lin H, Zhang Y, Wang R. The role of nocturnal penile tumescence and rigidity (NPTR) monitoring in the diagnosis of psychogenic erectile dysfunction: a review. Sex Med Rev. 2019;7:442–454. doi: 10.1016/j.sxmr.2018.10.005. [DOI] [PubMed] [Google Scholar]
  • 10.Pozzi E, Fallara G, Capogrosso P, Boeri L, Belladelli F, Corsini C, Costa A, Candela L, Cignoli D, Cazzaniga W, Schifano N, Ventimiglia E, d’Arma A, Montorsi F, Salonia A. Primary organic versus primary psychogenic erectile dysfunction: findings from a real-life cross-sectional study. Andrology. 2022;10:1302–1309. doi: 10.1111/andr.13212. [DOI] [PubMed] [Google Scholar]
  • 11.Pantazis A, Franco I, Gitlin J. Erectile dysfunction in adolescents and young adults. Curr Urol Rep. 2024;25:225–232. doi: 10.1007/s11934-024-01213-9. [DOI] [PubMed] [Google Scholar]
  • 12.Corona G, Petrone L, Mannucci E, Mansani R, Balercia G, Krausz C, Giommi R, Forti G, Maggi M. Difficulties in achieving vs maintaining erection: organic, psychogenic and relational determinants. Int J Impot Res. 2005;17:252–258. doi: 10.1038/sj.ijir.3901298. [DOI] [PubMed] [Google Scholar]
  • 13.Uckert S, Hedlund P, Waldkirch E, Sohn M, Jonas U, Andersson KE, Stief CG. Interactions between cGMP- and cAMP-pathways are involved in the regulation of penile smooth muscle tone. World J Urol. 2004;22:261–266. doi: 10.1007/s00345-003-0394-4. [DOI] [PubMed] [Google Scholar]
  • 14.Lin CS, Lin G, Lue TF. Cyclic nucleotide signaling in cavernous smooth muscle. J Sex Med. 2005;2:478–491. doi: 10.1111/j.1743-6109.2005.00080.x. [DOI] [PubMed] [Google Scholar]
  • 15.ElHady AK, El-Gamil DS, Abdel-Halim M, Abadi AH. Advancements in phosphodiesterase 5 inhibitors: unveiling present and future perspectives. Pharmaceuticals (Basel) 2023;16:1266. doi: 10.3390/ph16091266. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Argiolas A, Argiolas FM, Argiolas G, Melis MR. Erectile dysfunction: treatments, advances and new therapeutic strategies. Brain Sci. 2023;13:802. doi: 10.3390/brainsci13050802. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Berner MM, Kriston L, Harms A. Efficacy of PDE-5-inhibitors for erectile dysfunction. A comparative meta-analysis of fixed-dose regimen randomized controlled trials administering the international index of erectile ffunction in broad-spectrum populations. Int J Impot Res. 2006;18:229–235. doi: 10.1038/sj.ijir.3901395. [DOI] [PubMed] [Google Scholar]
  • 18.Atallah S, Haydar A, Jabbour T, Kfoury P, Sader G. The effectiveness of psychological interventions alone, or in combination with phosphodiesterase-5 inhibitors, for the treatment of erectile dysfunction: a systematic review. Arab J Urol. 2021;19:310–322. doi: 10.1080/2090598X.2021.1926763. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders, 5th ed. In: American Psychiatric. Arlington ineligible companies; 2023. [Google Scholar]
  • 20.Han M, Wang X, Yang H, Wang X, Zhu H, Song M. Efficacy of online cognitive behavioral therapy for nonorganic erectile dysfunction in reproductive-age males during the COVID-19 pandemic: a randomized wait list-controlled trial. J Sex Med. 2023;20:1325–1332. doi: 10.1093/jsxmed/qdad117. [DOI] [PubMed] [Google Scholar]
  • 21.Dewitte M, Bettocchi C, Carvalho J, Corona G, Flink I, Limoncin E, Pascoal P, Reisman Y, Van Lankveld J. A psychosocial approach to erectile dysfunction: position statements from the European society of sexual medicine (ESSM) Sex Med. 2021;9:100434. doi: 10.1016/j.esxm.2021.100434. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Huang CC, Liang JH, Li GY, Liang SK, Song WR, Zhang X, Wei GQ, Zhu CH, Wei P, Chen YB. Low-dose daily de-escalatory administration of tadalafil for psychological erectile dysfunction. Zhonghua Nan Ke Xue. 2013;19:241–246. [PubMed] [Google Scholar]
  • 23.Krugel U. Purinergic receptors in psychiatric disorders. Neuropharmacology. 2016;104:212–225. doi: 10.1016/j.neuropharm.2015.10.032. [DOI] [PubMed] [Google Scholar]
  • 24.Phatarpekar PV, Wen J, Xia Y. Role of adenosine signaling in penile erection and erectile disorders. J Sex Med. 2010;7:3553–3564. doi: 10.1111/j.1743-6109.2009.01555.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Ren WJ, Tang Y. A review of the state of purinergic signaling and psychological stress. Sichuan Da Xue Xue Bao Yi Xue Ban. 2021;52:33–38. doi: 10.12182/20210160102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Panchatsharam PK, Durland J, Zito PM. Physiology, Erection. In: StatPearls. Treasure Island (FL) ineligible companies. Disclosure: Pranau Panchatsharam declares no relevant financial relationships with ineligible companies; 2023. [Google Scholar]
  • 27.Temel Y, Hafizi S, Tan S, Visser-Vandewalle V. Role of the brain in the control of erection. Asian J Androl. 2006;8:259–264. doi: 10.1111/j.1745-7262.2006.00110.x. [DOI] [PubMed] [Google Scholar]
  • 28.Argiolas A, Melis MR. Central control of penile erection: role of the paraventricular nucleus of the hypothalamus. Prog Neurobiol. 2005;76:1–21. doi: 10.1016/j.pneurobio.2005.06.002. [DOI] [PubMed] [Google Scholar]
  • 29.Oti T, Ueda R, Kumagai R, Nagafuchi J, Ito T, Sakamoto T, Kondo Y, Sakamoto H. Sexual experience induces the expression of gastrin-releasing peptide and oxytocin receptors in the spinal ejaculation generator in rats. Int J Mol Sci. 2021;22:10362. doi: 10.3390/ijms221910362. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Oti T, Satoh K, Uta D, Nagafuchi J, Tateishi S, Ueda R, Takanami K, Young LJ, Galione A, Morris JF, Sakamoto T, Sakamoto H. Oxytocin influences male sexual activity via non-synaptic axonal release in the spinal cord. Curr Biol. 2021;31:103–114. e5. doi: 10.1016/j.cub.2020.09.089. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Feng S, Dong L, Yan B, Zheng S, Feng Z, Li X, Li J, Sun N, Ning Y, Jia H. Altered functional connectivity of large-scale brain networks in psychogenic erectile dysfunction associated with cognitive impairments. Neuropsychiatr Dis Treat. 2023;19:1925–1933. doi: 10.2147/NDT.S426213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Yang Y, Qu L, Mu L, Yao J, Su C, Zheng Q, Zheng H, Zhang P, Li Y. Electroacupuncture for psychogenic erectile dysfunction: a resting-state functional magnetic resonance imaging study exploring the alteration of fractional amplitude of low frequency fluctuation. Front Hum Neurosci. 2023;17:1116202. doi: 10.3389/fnhum.2023.1116202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Xu J, Chen Y, Gu L, Liu X, Yang J, Li M, Rao K, Dong X, Yang S, Huang B, Jin L, Wang T, Liu J, Wang S, Bai J. Hypothalamic-pituitary-adrenal axis activity and its relationship to the autonomic nervous system in patients with psychogenic erectile dysfunction. Front Endocrinol (Lausanne) 2023;14:1103621. doi: 10.3389/fendo.2023.1103621. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Chan KL, Poller WC, Swirski FK, Russo SJ. Central regulation of stress-evoked peripheral immune responses. Nat Rev Neurosci. 2023;24:591–604. doi: 10.1038/s41583-023-00729-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Chen CJ, Kuo TB, Tseng YJ, Yang CC. Combined cardiac sympathetic excitation and vagal impairment in patients with non-organic erectile dysfunction. Clin Neurophysiol. 2009;120:348–352. doi: 10.1016/j.clinph.2008.10.155. [DOI] [PubMed] [Google Scholar]
  • 36.Nguyen L, Kakeda S, Watanabe K, Katsuki A, Sugimoto K, Igata N, Shinkai T, Abe O, Korogi Y, Ikenouchi A, Yoshimura R. Brain structural network alterations related to serum cortisol levels in drug-naive, first-episode major depressive disorder patients: a source-based morphometric study. Sci Rep. 2020;10:22096. doi: 10.1038/s41598-020-79220-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Knezevic E, Nenic K, Milanovic V, Knezevic NN. The role of cortisol in chronic stress, neurodegenerative diseases, and psychological disorders. Cells. 2023;12:2726. doi: 10.3390/cells12232726. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Koelsch S, Boehlig A, Hohenadel M, Nitsche I, Bauer K, Sack U. The impact of acute stress on hormones and cytokines, and how their recovery is affected by music-evoked positive mood. Sci Rep. 2016;6:23008. doi: 10.1038/srep23008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Van den Broeck T, Soebadi MA, Falter A, Raets L, Duponselle J, Lootsma J, Heintz A, Philtjens U, Hofkens L, Gonzalez-Viedma A, Driesen K, Sandner P, Albersen M, Brône B, Van Renterghem K. Testosterone induces relaxation of human corpus cavernosum tissue of patients with erectile dysfunction. Sex Med. 2020;8:114–119. doi: 10.1016/j.esxm.2019.10.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Drury AN, Szent-Gyorgyi A. The physiological activity of adenine compounds with especial reference to their action upon the mammalian heart. J Physiol. 1929;68:213–237. doi: 10.1113/jphysiol.1929.sp002608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Degubareff T, Sleator W Jr. Effects of caffeine on mammalian atrial muscle, and its interaction with adenosine and calcium. J Pharmacol Exp Ther. 1965;148:202–214. [PubMed] [Google Scholar]
  • 42.Franco R, Navarro G, Martinez-Pinilla E. The adenosine A(2A) receptor in the basal ganglia: expression, heteromerization, functional selectivity and signalling. Int Rev Neurobiol. 2023;170:49–71. doi: 10.1016/bs.irn.2023.04.008. [DOI] [PubMed] [Google Scholar]
  • 43.Liang S. Purinergic signaling in functions and diseases. In: People’s Health. Beijing ineligible companies. Disclosure: Shangdong Liang declares no relevant financial relationships with ineligible companies; 2009. [Google Scholar]
  • 44.Dixon AK, Gubitz AK, Sirinathsinghji DJ, Richardson PJ, Freeman TC. Tissue distribution of adenosine receptor mRNAs in the rat. Br J Pharmacol. 1996;118:1461–1468. doi: 10.1111/j.1476-5381.1996.tb15561.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Liu YJ, Chen J, Li X, Zhou X, Hu YM, Chu SF, Peng Y, Chen NH. Research progress on adenosine in central nervous system diseases. CNS Neurosci Ther. 2019;25:899–910. doi: 10.1111/cns.13190. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Fredholm BB, IJzerman AP, Jacobson KA, Linden J, Müller CE. International union of basic and clinical pharmacology. LXXXI. Nomenclature and classification of adenosine receptors--an update. Pharmacol Rev. 2011;63:1–34. doi: 10.1124/pr.110.003285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Wang M, Li Z, Song Y, Sun Q, Deng L, Lin Z, Zeng Y, Qiu C, Lin J, Guo H, Chen J, Guo W. Genetic tagging of the adenosine A2A receptor reveals its heterogeneous expression in brain regions. Front Neuroanat. 2022;16:978641. doi: 10.3389/fnana.2022.978641. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Mills JH, Kim DG, Krenz A, Chen JF, Bynoe MS. A2A adenosine receptor signaling in lymphocytes and the central nervous system regulates inflammation during experimental autoimmune encephalomyelitis. J Immunol. 2012;188:5713–5722. doi: 10.4049/jimmunol.1200545. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Rajasundaram S. Adenosine A2A receptor signaling in the immunopathogenesis of experimental autoimmune encephalomyelitis. Front Immunol. 2018;9:402. doi: 10.3389/fimmu.2018.00402. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Lopes LV, Rebola N, Pinheiro PC, Richardson PJ, Oliveira CR, Cunha RA. Adenosine A3 receptors are located in neurons of the rat hippocampus. Neuroreport. 2003;14:1645–1648. doi: 10.1097/00001756-200308260-00021. [DOI] [PubMed] [Google Scholar]
  • 51.Song X, Wu W, Warner M, Gustafsson JÅ. Liver X receptor regulation of glial cell functions in the CNS. Biomedicines. 2022;10:2165. doi: 10.3390/biomedicines10092165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Daré E, Schulte G, Karovic O, Hammarberg C, Fredholm BB. Modulation of glial cell functions by adenosine receptors. Physiol Behav. 2007;92:15–20. doi: 10.1016/j.physbeh.2007.05.031. [DOI] [PubMed] [Google Scholar]
  • 53.Madeira D, Domingues J, Lopes CR, Canas PM, Cunha RA, Agostinho P. Modification of astrocytic Cx43 hemichannel activity in animal models of AD: modulation by adenosine A(2A) receptors. Cell Mol Life Sci. 2023;80:340. doi: 10.1007/s00018-023-04983-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Gao Z. Adenosine A(2A) receptor and glia. Int Rev Neurobiol. 2023;170:29–48. doi: 10.1016/bs.irn.2023.08.002. [DOI] [PubMed] [Google Scholar]
  • 55.Ning C. Dissertation, Central South University of China. 2012. The role and mechanism of adenosine A(2B) receptor in abnormal penile erection and fibrosis. [Google Scholar]
  • 56.Mi T, Abbasi S, Zhang H, Uray K, Chunn JL, Xia LW, Molina JG, Weisbrodt NW, Kellems RE, Blackburn MR, Xia Y. Excess adenosine in murine penile erectile tissues contributes to priapism via A2B adenosine receptor signaling. J Clin Invest. 2008;118:1491–1501. doi: 10.1172/JCI33467. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Filippi S, Mancini M, Amerini S, Bartolini M, Natali A, Mancina R, Forti G, Ledda F, Maggi M. Functional adenosine receptors in human corpora cavernosa. Int J Androl. 2000;23:210–217. doi: 10.1046/j.1365-2605.2000.00232.x. [DOI] [PubMed] [Google Scholar]
  • 58.Ignarro LJ. Nitric oxide. A novel signal transduction mechanism for transcellular communication. Hypertension. 1990;16:477–483. doi: 10.1161/01.hyp.16.5.477. [DOI] [PubMed] [Google Scholar]
  • 59.Passos GR, de Oliveira MG, Ghezzi AC, Mello GC, Levi D’Ancona CA, Teixeira SA, Muscará MN, Grespan Bottoli CB, Vilela de Melo L, de Oliveira E, Antunes E, Mónica FZ. Periprostatic adipose tissue (PPAT) supernatant from obese mice releases anticontractile substances and increases human prostate epithelial cell proliferation: the role of nitric oxide and adenosine. Front Pharmacol. 2023;14:1145860. doi: 10.3389/fphar.2023.1145860. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Wilson C, Lee MD, Buckley C, Zhang X, McCarron JG. Mitochondrial ATP production is required for endothelial cell control of vascular tone. Function (Oxf) 2022;4:zqac063. doi: 10.1093/function/zqac063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Fu Q, Wang Y, Yan C, Xiang YK. Phosphodiesterases in heart and vessels: from physiology to diseases. Physiol Rev. 2024;104:765–834. doi: 10.1152/physrev.00015.2023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Zhu Z, Tang W, Qiu X, Xin X, Zhang J. Advances in targeting phosphodiesterase 1: from mechanisms to potential therapeutics. Eur J Med Chem. 2024;263:115967. doi: 10.1016/j.ejmech.2023.115967. [DOI] [PubMed] [Google Scholar]
  • 63.Crema LM, Pettenuzzo LF, Schlabitz M, Diehl L, Hoppe J, Mestriner R, Laureano D, Salbego C, Dalmaz C, Vendite D. The effect of unpredictable chronic mild stress on depressive-like behavior and on hippocampal A1 and striatal A2A adenosine receptors. Physiol Behav. 2013;109:1–7. doi: 10.1016/j.physbeh.2012.11.001. [DOI] [PubMed] [Google Scholar]
  • 64.Liebenberg N, Joca S, Wegener G. Nitric oxide involvement in the antidepressant-like effect of ketamine in the Flinders sensitive line rat model of depression. Acta Neuropsychiatr. 2015;27:90–96. doi: 10.1017/neu.2014.39. [DOI] [PubMed] [Google Scholar]
  • 65.Zhang J, Snyder SH. Nitric oxide in the nervous system. Annu Rev Pharmacol Toxicol. 1995;35:213–233. doi: 10.1146/annurev.pa.35.040195.001241. [DOI] [PubMed] [Google Scholar]
  • 66.Boison D. Adenosine kinase: exploitation for therapeutic gain. Pharmacol Rev. 2013;65:906–943. doi: 10.1124/pr.112.006361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Moreira-de-Sa A, Lourenço VS, Canas PM, Cunha RA. Adenosine A(2A) receptors as biomarkers of brain diseases. Front Neurosci. 2021;15:702581. doi: 10.3389/fnins.2021.702581. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Celebi G, Gocmez SS, Ozer C, Duruksu G, Yazır Y, Utkan T. Propolis prevents vascular endothelial dysfunction by attenuating inflammation and oxidative damage in the chronic unpredictable stress model of depression in rats. J Pharm Pharmacol. 2023;75:1418–1429. doi: 10.1093/jpp/rgad071. [DOI] [PubMed] [Google Scholar]
  • 69.Yin CY, Huang SY, Gao L, Lin YH, Chang L, Wu HY, Zhu DY, Luo CX. Neuronal nitric oxide synthase in nucleus accumbens specifically mediates susceptibility to social defeat stress through cyclin-dependent kinase 5. J Neurosci. 2021;41:2523–2539. doi: 10.1523/JNEUROSCI.0422-20.2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Vignjević Petrinović S, Budeč M, Marković D, Mitrović Ajtić O, Jovčić G, Milošević M, Momčilović S, Čokić V. Nitric oxide-dependent expansion of erythroid progenitors in a murine model of chronic psychological stress. Histochem Cell Biol. 2020;153:457–468. doi: 10.1007/s00418-020-01856-y. [DOI] [PubMed] [Google Scholar]
  • 71.Furchgott RF. The 1996 albert lasker medical research awards. The discovery of endothelium-derived relaxing factor and its importance in the identification of nitric oxide. JAMA. 1996;276:1186–1188. [PubMed] [Google Scholar]
  • 72.Frandsenn U, Bangsbo J, Sander M, Höffner L, Betak A, Saltin B, Hellsten Y. Exercise-induced hyperaemia and leg oxygen uptake are not altered during effective inhibition of nitric oxide synthase with N(G)-nitro-L-arginine methyl ester in humans. J Physiol. 2001;531:257–264. doi: 10.1111/j.1469-7793.2001.0257j.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Casey DP, Mohamed EA, Joyner MJ. Role of nitric oxide and adenosine in the onset of vasodilation during dynamic forearm exercise. Eur J Appl Physiol. 2013;113:295–303. doi: 10.1007/s00421-012-2439-0. [DOI] [PubMed] [Google Scholar]
  • 74.Labazi H, Tilley SL, Ledent C, Mustafa SJ. Role of adenosine receptor(s) in the control of vascular tone in the mouse pudendal artery. J Pharmacol Exp Ther. 2016;356:673–680. doi: 10.1124/jpet.115.230144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Wen J, Grenz A, Zhang Y, Dai Y, Kellems RE, Blackburn MR, Eltzschig HK, Xia Y. A2B adenosine receptor contributes to penile erection via PI3K/AKT signaling cascade-mediated eNOS activation. FASEB J. 2011;25:2823–2830. doi: 10.1096/fj.11-181057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Wyatt AW, Steinert JR, Wheeler-Jones CP, Morgan AJ, Sugden D, Pearson JD, Sobrevia L, Mann GE. Early activation of the p42/p44MAPK pathway mediates adenosine-induced nitric oxide production in human endothelial cells: a novel calcium-insensitive mechanism. FASEB J. 2002;16:1584–1594. doi: 10.1096/fj.01-0125com. [DOI] [PubMed] [Google Scholar]
  • 77.Vásquez G, Sanhueza F, Vásquez R, González M, San Martín R, Casanello P, Sobrevia L. Role of adenosine transport in gestational diabetes-induced L-arginine transport and nitric oxide synthesis in human umbilical vein endothelium. J Physiol. 2004;560:111–122. doi: 10.1113/jphysiol.2004.068288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Vignozzi L, Filippi S, Comeglio P, Cellai I, Morelli A, Rastrelli G, Maneschi E, Mannucci E, Maggi M. Metformin in vitro and in vivo increases adenosine signaling in rabbit corpora cavernosa. J Sex Med. 2014;11:1694–1708. doi: 10.1111/jsm.12572. [DOI] [PubMed] [Google Scholar]
  • 79.Li JM, Fenton RA, Wheeler HB, Powell CC, Peyton BD, Cutler BS, Dobson JG Jr. Adenosine A2a receptors increase arterial endothelial cell nitric oxide. J Surg Res. 1998;80:357–364. doi: 10.1006/jsre.1998.5439. [DOI] [PubMed] [Google Scholar]
  • 80.Wen J, Dai Y, Zhang Y, Zhang W, Kellems RE, Xia Y. Impaired erectile function in CD73-deficient mice with reduced endogenous penile adenosine production. J Sex Med. 2011;8:2172–2180. doi: 10.1111/j.1743-6109.2011.02316.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Dai Y, Zhang Y, Phatarpekar P, Mi T, Zhang H, Blackburn MR, Xia Y. Adenosine signaling, priapism and novel therapies. J Sex Med. 2009;6(Suppl 3):292–301. doi: 10.1111/j.1743-6109.2008.01187.x. [DOI] [PubMed] [Google Scholar]
  • 82.Yang R, Wang J, Chen Y, Sun Z, Wang R, Dai Y. Effect of caffeine on erectile function via up-regulating cavernous cyclic guanosine monophosphate in diabetic rats. J Androl. 2008;29:586–591. doi: 10.2164/jandrol.107.004721. [DOI] [PubMed] [Google Scholar]
  • 83.Noto T, Inoue H, Mochida H, Kikkawa K. Role of adenosine and P2 receptors in the penile tumescence in anesthetized dogs. Eur J Pharmacol. 2001;425:51–55. doi: 10.1016/s0014-2999(01)01167-0. [DOI] [PubMed] [Google Scholar]
  • 84.Gur S, Ozturk B. Altered relaxant responses to adenosine and adenosine 5’-triphosphate in the corpus cavernosum from men and rats with diabetes. Pharmacology. 2000;60:105–112. doi: 10.1159/000028354. [DOI] [PubMed] [Google Scholar]
  • 85.Ragazzi E, Chinellato A, Italiano G, Pagano F, Calabrò A. Characterization of in vitro relaxant mechanisms in erectile tissue from rabbits of different ages. Urol Res. 1996;24:317–322. doi: 10.1007/BF00389786. [DOI] [PubMed] [Google Scholar]
  • 86.Burnstock G. Purinergic signalling in the reproductive system in health and disease. Purinergic Signal. 2014;10:157–187. doi: 10.1007/s11302-013-9399-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Tostes RC, Giachini FR, Carneiro FS, Leite R, Inscho EW, Webb RC. Determination of adenosine effects and adenosine receptors in murine corpus cavernosum. J Pharmacol Exp Ther. 2007;322:678–685. doi: 10.1124/jpet.107.122705. [DOI] [PubMed] [Google Scholar]
  • 88.Mantelli L, Amerini S, Ledda F, Forti G, Maggi M. The potent relaxant effect of adenosine in rabbit corpora cavernosa is nitric oxide independent and mediated by A2 receptors. J Androl. 1995;16:312–317. [PubMed] [Google Scholar]
  • 89.Faria M, Magalhaes-Cardoso T, Lafuente-de-Carvalho JM, Correia-de-Sa P. Corpus cavernosum from men with vasculogenic impotence is partially resistant to adenosine relaxation due to endothelial A(2B) receptor dysfunction. J Pharmacol Exp Ther. 2006;319:405–413. doi: 10.1124/jpet.106.107821. [DOI] [PubMed] [Google Scholar]
  • 90.Moura VJG, Alencar AKN, Calasans-Maia JA, da Silva JS, Fraga CAM, Zapata-Sudo G, Barreiro EJ, Sudo RT. Novel agonist of adenosine receptor induces relaxation of corpus cavernosum in guinea pigs: an in vitro and in vivo study. Urology. 2015;85:1214.e17–1214.e21. doi: 10.1016/j.urology.2015.02.006. [DOI] [PubMed] [Google Scholar]
  • 91.Wen J, Du C, Bai F, Zhang Y, Feng Z, Yang X. Study on the therapeutic effect of adenosine deaminase on abnormal penile erection. The annual conference of urology in six provinces and one city in East China and the 2011 annual conference of urology and andrology in Zhejiang Province.2011. [Google Scholar]
  • 92.Kataoka K, Furukawa K, Nagao K, Ishii N, Tsuru H. The participation of adenosine receptors in the adenosine 5’-triphosphate-induced relaxation in the isolated rabbit corpus cavernosum penis. Int J Urol. 2007;14:764–768. doi: 10.1111/j.1442-2042.2007.01803.x. [DOI] [PubMed] [Google Scholar]
  • 93.Wen J, Wang B, Du C, Xu G, Zhang Z, Li Y, Zhang N. A2B adenosine receptor agonist improves erectile function in diabetic rats. Tohoku J Exp Med. 2015;237:141–148. doi: 10.1620/tjem.237.141. [DOI] [PubMed] [Google Scholar]
  • 94.Xiong Y, Qin F, Wei S, Yang X, Li J, Wu C, Zhang F, Yuan J. Targeting adenosine A2b receptor promotes penile rehabilitation of refractory erectile dysfunction. Adv Sci (Weinh) 2024;11:e2306514. doi: 10.1002/advs.202306514. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Kong X, Jiang J, Cheng B, Jiang R. Effect of low androgen status on the expression of adenosine A(2A) and A(2B) receptors in rat penile corpus cavernosum. Andrologia. 2019;51:e13344. doi: 10.1111/and.13344. [DOI] [PubMed] [Google Scholar]
  • 96.Wen J, Jiang X, Dai Y, Zhang Y, Tang Y, Sun H, Mi T, Phatarpekar PV, Kellems RE, Blackburn MR, Xia Y. Increased adenosine contributes to penile fibrosis, a dangerous feature of priapism, via A2B adenosine receptor signaling. FASEB J. 2010;24:740–749. doi: 10.1096/fj.09-144147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Ning C, Wen J, Zhang Y, Dai Y, Wang W, Zhang W, Qi L, Grenz A, Eltzschig HK, Blackburn MR, Kellems RE, Xia Y. Excess adenosine A2B receptor signaling contributes to priapism through HIF-1alpha mediated reduction of PDE5 gene expression. FASEB J. 2014;28:2725–2735. doi: 10.1096/fj.13-247833. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Chiang PH, Wu SN, Tsai EM, Wu CC, Shen MR, Huang CH, Chiang CP. Adenosine modulation of neurotransmission in penile erection. Br J Clin Pharmacol. 1994;38:357–362. doi: 10.1111/j.1365-2125.1994.tb04366.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Itzhak I, Cohen S, Fishman S, Fishman P. A3 adenosine receptor allosteric modulator CF602 reverses erectile dysfunction in a diabetic rat model. Andrologia. 2022;54:e14498. doi: 10.1111/and.14498. [DOI] [PubMed] [Google Scholar]
  • 100.Wawrzyniak AJ, Dilsizian V, Krantz DS, Harris KM, Smith MF, Shankovich A, Whittaker KS, Rodriguez GA, Gottdiener J, Li S, Kop W, Gottlieb SS. High concordance between mental stress-induced and adenosine-induced myocardial ischemia assessed using SPECT in heart failure patients: hemodynamic and biomarker correlates. J Nucl Med. 2015;56:1527–1533. doi: 10.2967/jnumed.115.157990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Fontella FU, Bruno AN, Crema LM, Battastini AM, Sarkis JJ, Netto CA, Dalmaz C. Acute and chronic stress alter ecto-nucleotidase activities in synaptosomes from the rat hippocampus. Pharmacol Biochem Behav. 2004;78:341–347. doi: 10.1016/j.pbb.2004.04.005. [DOI] [PubMed] [Google Scholar]
  • 102.Torres IL, Buffon A, Dantas G, Fürstenau CR, Böhmer AE, Battastini AM, Sarkis JJ, Dalmaz C, Ferreira MB. Chronic stress effects on adenine nucleotide hydrolysis in the blood serum and brain structures of rats. Pharmacol Biochem Behav. 2002;74:181–186. doi: 10.1016/s0091-3057(02)00994-2. [DOI] [PubMed] [Google Scholar]
  • 103.Böhmer AE, Fürstenau CR, Torres IL, Crema L, Battastini AM, Dalmaz C, Ferreira MB, Sarkis JJ. The effect of stress upon hydrolysis adenine nucleotides in blood serum of rats. Pharmacol Biochem Behav. 2003;75:467–471. doi: 10.1016/s0091-3057(03)00143-6. [DOI] [PubMed] [Google Scholar]
  • 104.Piato AL, Rosemberg DB, Capiotti KM, Siebel AM, Herrmann AP, Ghisleni G, Vianna MR, Bogo MR, Lara DR, Bonan CD. Acute restraint stress in zebrafish: behavioral parameters and purinergic signaling. Neurochem Res. 2011;36:1876–1886. doi: 10.1007/s11064-011-0509-z. [DOI] [PubMed] [Google Scholar]
  • 105.Zimmermann FF, Altenhofen S, Kist LW, Leite CE, Bogo MR, Cognato GP, Bonan CD. Unpredictable chronic stress alters adenosine metabolism in zebrafish brain. Mol Neurobiol. 2016;53:2518–2528. doi: 10.1007/s12035-015-9270-7. [DOI] [PubMed] [Google Scholar]
  • 106.Hao T, Du X, Yang S, Zhang Y, Liang F. Astrocytes-induced neuronal inhibition contributes to depressive-like behaviors during chronic stress. Life Sci. 2020;258:118099. doi: 10.1016/j.lfs.2020.118099. [DOI] [PubMed] [Google Scholar]
  • 107.Kaster MP, Machado NJ, Silva HB, Nunes A, Ardais AP, Santana M, Baqi Y, Müller CE, Rodrigues AL, Porciúncula LO, Chen JF, Tomé ÂR, Agostinho P, Canas PM, Cunha RA. Caffeine acts through neuronal adenosine A2A receptors to prevent mood and memory dysfunction triggered by chronic stress. Proc Natl Acad Sci U S A. 2015;112:7833–7838. doi: 10.1073/pnas.1423088112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Serchov T, Clement HW, Schwarz MK, Iasevoli F, Tosh DK, Idzko M, Jacobson KA, de Bartolomeis A, Normann C, Biber K, van Calker D. Increased signaling via adenosine A1 receptors, sleep deprivation, imipramine, and ketamine inhibit depressive-like behavior via induction of homer1a. Neuron. 2015;87:549–562. doi: 10.1016/j.neuron.2015.07.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Nam HW, Bruner RC, Choi DS. Adenosine signaling in striatal circuits and alcohol use disorders. Mol Cells. 2013;36:195–202. doi: 10.1007/s10059-013-0192-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.van Calker D, Biber K, Domschke K, Serchov T. The role of adenosine receptors in mood and anxiety disorders. J Neurochem. 2019;151:11–27. doi: 10.1111/jnc.14841. [DOI] [PubMed] [Google Scholar]
  • 111.Bozorgi H, Rashidy-Pour A, Moradikor N, Zamani M, Motaghi E. Neurobehavioral protective effects of Japanese sake yeast supplement against chronic stress-induced anxiety and depression-like symptoms in mice: possible role of central adenosine receptors. Psychopharmacology (Berl) 2024;241:401–416. doi: 10.1007/s00213-023-06496-3. [DOI] [PubMed] [Google Scholar]
  • 112.Li Y, Li L, Wu J, Zhu Z, Feng X, Qin L, Zhu Y, Sun L, Liu Y, Qiu Z, Duan S, Yu YQ. Activation of astrocytes in hippocampus decreases fear memory through adenosine A(1) receptors. Elife. 2020;9:e57155. doi: 10.7554/eLife.57155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Almeida RF, Comasseto DD, Ramos DB, Hansel G, Zimmer ER, Loureiro SO, Ganzella M, Souza DO. Guanosine anxiolytic-like effect involves adenosinergic and glutamatergic neurotransmitter systems. Mol Neurobiol. 2017;54:423–436. doi: 10.1007/s12035-015-9660-x. [DOI] [PubMed] [Google Scholar]
  • 114.Serchov T, Heumann R, van Calker D, Biber K. Signaling pathways regulating Homer1a expression: implications for antidepressant therapy. Biol Chem. 2016;397:207–214. doi: 10.1515/hsz-2015-0267. [DOI] [PubMed] [Google Scholar]
  • 115.Serchov T, Schwarz I, Theiss A, Sun L, Holz A, Döbrössy MD, Schwarz MK, Normann C, Biber K, van Calker D. Enhanced adenosine A(1) receptor and Homer1a expression in hippocampus modulates the resilience to stress-induced depression-like behavior. Neuropharmacology. 2020;162:107834. doi: 10.1016/j.neuropharm.2019.107834. [DOI] [PubMed] [Google Scholar]
  • 116.Vincenzi F, Ravani A, Pasquini S, Merighi S, Gessi S, Romagnoli R, Baraldi PG, Borea PA, Varani K. Positive allosteric modulation of A(1) adenosine receptors as a novel and promising therapeutic strategy for anxiety. Neuropharmacology. 2016;111:283–292. doi: 10.1016/j.neuropharm.2016.09.015. [DOI] [PubMed] [Google Scholar]
  • 117.Cen XQ, Li P, Wang B, Chen X, Zhao Y, Yang N, Peng Y, Li CH, Ning YL, Zhou YG. Knockdown of adenosine A2A receptors in hippocampal neurons prevents post-TBI fear memory retrieval. Exp Neurol. 2023;364:114378. doi: 10.1016/j.expneurol.2023.114378. [DOI] [PubMed] [Google Scholar]
  • 118.Batalha VL, Pego JM, Fontinha BM, Costenla AR, Valadas JS, Baqi Y, Radjainia H, Müller CE, Sebastião AM, Lopes LV. Adenosine A(2A) receptor blockade reverts hippocampal stress-induced deficits and restores corticosterone circadian oscillation. Mol Psychiatry. 2013;18:320–331. doi: 10.1038/mp.2012.8. [DOI] [PubMed] [Google Scholar]
  • 119.Florén Lind S, Stam F, Zelleroth S, Meurling E, Frick A, Grönbladh A. Acute caffeine differently affects risk-taking and the expression of BDNF and of adenosine and opioid receptors in rats with high or low anxiety-like behavior. Pharmacol Biochem Behav. 2023;227-228:173573. doi: 10.1016/j.pbb.2023.173573. [DOI] [PubMed] [Google Scholar]
  • 120.Jiang L, Ran H, Duan W, Zheng J. Regulatory effects of adenosine A2A receptors on psychomotor ability and mood behavior of mice. Med J Chin PLA. 2011;36:706–710. [Google Scholar]
  • 121.Wang M, Li P, Li Z, da Silva BS, Zheng W, Xiang Z, He Y, Xu T, Cordeiro C, Deng L, Dai Y, Ye M, Lin Z, Zhou J, Zhou X, Ye F, Cunha RA, Chen J, Guo W. Lateral septum adenosine A(2A) receptors control stress-induced depressive-like behaviors via signaling to the hypothalamus and habenula. Nat Commun. 2023;14:1880. doi: 10.1038/s41467-023-37601-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Zhao YF, Verkhratsky A, Tang Y, Illes P. Astrocytes and major depression: the purinergic avenue. Neuropharmacology. 2022;220:109252. doi: 10.1016/j.neuropharm.2022.109252. [DOI] [PubMed] [Google Scholar]
  • 123.Chiu GS, Darmody PT, Walsh JP, Moon ML, Kwakwa KA, Bray JK, McCusker RH, Freund GG. Adenosine through the A2A adenosine receptor increases IL-1beta in the brain contributing to anxiety. Brain Behav Immun. 2014;41:218–231. doi: 10.1016/j.bbi.2014.05.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Jacobsen JP, Medvedev IO, Caron MG. The 5-HT deficiency theory of depression: perspectives from a naturalistic 5-HT deficiency model, the tryptophan hydroxylase 2Arg439His knockin mouse. Philos Trans R Soc Lond B Biol Sci. 2012;367:2444–2459. doi: 10.1098/rstb.2012.0109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Simões AP, Gonçalves FQ, Rial D, Ferreira SG, Lopes JP, Canas PM, Cunha RA. CD73-mediated formation of extracellular adenosine is responsible for adenosine A(2A) receptor-mediated control of fear memory and amygdala plasticity. Int J Mol Sci. 2022;23:12826. doi: 10.3390/ijms232112826. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Wen J, Xia Y. Adenosine signaling: good or bad in erectile function? Arterioscler Thromb Vasc Biol. 2012;32:845–850. doi: 10.1161/ATVBAHA.111.226803. [DOI] [PubMed] [Google Scholar]

Articles from American Journal of Translational Research are provided here courtesy of e-Century Publishing Corporation

RESOURCES