Elsevier

Life Sciences

Volume 238, 1 December 2019, 116922
Life Sciences

Dysregulated NO/PDE5 signaling in the sickle cell mouse lower urinary tract: Reversal by oral nitrate therapy

https://doi.org/10.1016/j.lfs.2019.116922Get rights and content

Abstract

Aims

Nitric oxide (NO) has a critical, but not well understood, influence in the physiology of the lower urinary tract. We evaluated the effect of NO/phosphodiesterase (PDE)5 signaling in voiding dysfunction in the sickle cell disease (SCD) mouse, characterized by low NO bioavailability.

Main methods

Adult SCD (Sickle) and wild-type (WT) male mice were treated daily with sodium nitrate (10 mM) or vehicle. After 18 days, blood was obtained for nitrite measurement, urethra was collected for organ bath study, and bladder and urethra were collected for Western blot analysis of PDE5 phosphorylation (Ser-92) (activated form). Non-anesthetized mice underwent evaluation of urine volume by void spot assay. eNOS phosphorylation (Ser-1177) and nNOS phosphorylation (Ser-1412) (positive regulatory sites) were evaluated in the bladder and urethra of untreated mice.

Key findings

Sickle mice exhibited decreased eNOS, nNOS, and PDE5 phosphorylation in the bladder and urethra, decreased plasma nitrite levels, increased relaxation of phenylephrine-contracted urethral tissue to an NO donor sodium nitroprusside, and increased total urine volume, compared with WT mice. Nitrate treatment normalized plasma nitrite levels, relaxation of urethra to sodium nitroprusside, PDE5 phosphorylation in the urethra and bladder, and urine volume in Sickle mice.

Significance

Derangement in PDE5 activity associated with basally low NO bioavailability in the bladder and urethra contributes to the molecular basis for voiding abnormalities in Sickle mice. Inorganic nitrate supplementation normalized voiding in Sickle mice through mechanisms likely involving upregulation of PDE5 activity. These findings suggest that interventions targeting dysregulatory NO/PDE5 signaling may ameliorate overactive bladder in SCD.

Introduction

Micturition depends on the coordination between the urinary bladder and the urethra. Storage of urine in the bladder requires bladder relaxation during the filling phase, while bladder emptying requires coordinated contraction of the bladder and relaxation of the urethra [1]. Increased contractions of bladder detrusor muscle during the storage phase result in a hypercontractile voiding disorder, the most common cause of overactive bladder syndrome (OAB) [2]. OAB is clinically characterized by urinary urgency with or without urinary urge incontinence and is usually accompanied by urinary frequency and nocturia. The estimated prevalence of OAB is between 11.8 and 24.7% with similar rates in men and women [3].

Detrusor overactivity may result from impairment in detrusor smooth muscle tone, neuronal input, and/or sensory signals originating in the bladder [2]. A wide range of neurotransmitters control urine storage and voiding, including nitric oxide (NO). NO mediates relaxation of urethral and bladder neck smooth muscle [[4], [5], [6], [7]]. NO also contributes to regulation of detrusor tone. Systemic NO synthase (NOS) inhibition, intravesical NO scavenging, and knockout of neuronal NOS (nNOS) and cGMP-dependent protein kinase result in bladder overactivity in rats and mice [[8], [9], [10], [11], [12]], while intravesically or systemically supplied NO donors reduce bladder contraction frequency [13,14]. However, NO donors and cGMP analogues have also been shown to evoke a complex response (relaxant, contractile, or biphasic in isolated human detrusor muscle) [15] or have only a modest relaxing effect on isolated rat and mouse detrusor muscle [[16], [17], [18]]. Furthermore, both NO-mediated cGMP-dependent relaxation [19] and cGMP‐independent contraction of the detrusor [20,21] have been reported, further complicating the understanding of NO's role in bladder physiology and pathophysiology.

OAB is common in the sickle cell disease (SCD) population, with rates that exceed twice that of age-matched control subjects [22]. Symptoms of OAB in SCD include urinary frequency, urgency and nocturia [22,23]. SCD is caused by a single point mutation in the β-globin gene of hemoglobin leading to polymerization of hemoglobin under hypoxic conditions, red blood cell fragility, hemolysis, vaso‐occlusive crises, and inflammation [24]. SCD is also characterized by a chronic state of reduced NO bioavailability [25]. We have shown that homozygous SCD mice, a humanized mouse model of SCD, have decreased endothelial NO bioavailability in the penis associated with decreased phosphodiesterase (PDE)5 function, such that cGMP accumulation occurs upon neurostimulation, which results in excessive cavernosal vasorelaxation and priapism [[26], [27], [28]]. We have also demonstrated that SCD mice exhibit enhanced voiding and non‐voiding bladder contractions and produce greater volumes of urine [29]. These basic science findings correlate with clinical observations that both priapism and OAB are common disorders among the SCD population [22].

We hypothesized that chronically aberrant NO regulatory signaling in the bladder and urethra contributes to voiding dysfunction in SCD. We evaluated the effects of NO deficiency in lower urinary tract function using the SCD mouse at baseline and after long-term dietary inorganic nitrate supplementation.

Section snippets

Animals

Berkeley transgenic sickle cell mice (Sickle) and wild type (WT) male mice, 6–8 months old, were used. Berkeley Sickle mice have deletions of murine α and β globins and a transgene containing human α and β globins [30]. WT C57BL/6 mice were chosen as controls because this represents the predominant background strain for Sickle mice. In some experiments, hemizygous littermate mice were used as an additional control. WT mice and breeding pairs for Sickle mice (strain number 3342) were obtained

Protein expressions of P-eNOS (Ser-1177) and P-nNOS (Ser-1412) were decreased in the sickle mouse urethra and bladder

Activated (phosphorylated) forms of eNOS (Ser-1177) and nNOS (Ser-1412) were significantly decreased in the Sickle mouse urethra compared to that of WT mice, and did not differ from that of hemizygous mice (Fig. 1A and B). HEK293 cells transfected with eNOS and stimulated with insulin-like growth factor-1 [34], and major pelvic ganglia (MPG), were used as controls for P-eNOS (Ser-1177) and nNOS, respectively.

Activated (phosphorylated) eNOS (Ser-1177) was significantly decreased in the Sickle

Discussion

In this study, we demonstrate an association between PDE5 dysregulation and basally low NO bioavailability in the bladder and urethra, and impaired lower urinary tract function, in a mouse model of SCD. Long-term oral treatment with inorganic nitrate reversed aberrant signaling of the NO/PDE5 axis and normalized the amount of voided volume, a surrogate for urinary function, in Sickle mice. These results fit with our previous findings that low NO/PDE5 bioavailability in the Sickle mouse penis

Conclusion

Derangements in the PDE5 regulatory pathway and basally low NO bioavailability in the bladder and urethra are molecular conditions associated with voiding dysfunction in Sickle mice. Inorganic nitrate supplementation normalized voiding function in Sickle mice through mechanisms likely involving the upregulation of PDE5 activity in the bladder and urethra. These findings suggest that interventions targeting dysregulatory NO/PDE5 signaling may ameliorate OAB in SCD.

Declaration of competing interest

The authors declare that there are no conflicts of interest.

Acknowledgement

Funding:This work was supported by the National Institutes of Health [grant number NIH/NIDDK, R56DK114095]; and the São Paulo Research Foundation, Brazil [grant numbers FAPESP, 2017/08122–9, 2018/06243–6]. The funders had no involvement in study design, data collection, analysis and interpretation of data, writing of the report, or in the decision to submit the article for publication.

References (64)

  • R. Johansson et al.

    Activity and expression of nitric oxide synthase in the hypertrophied rat bladder and the effect of nitric oxide on bladder smooth muscle growth

    J. Urol.

    (2002)
  • C.S. Lin et al.

    Phosphodiesterase-5 expression and function in the lower urinary tract: a critical review

    Urology

    (2013)
  • C.S. Lin et al.

    Identification and regulation of human PDE5A gene promoter

    Biochem. Biophys. Res. Commun.

    (2001)
  • C.D. Koch et al.

    Enterosalivary nitrate metabolism and the microbiome: intersection of microbial metabolism, nitric oxide and diet in cardiac and pulmonary vascular health

    Free Radic. Biol. Med.

    (2017)
  • M. Oka et al.

    Suppression of bladder oxidative stress and inflammation by a phytotherapeutic agent in a rat model of partial bladder outlet obstruction

    J. Urol.

    (2009)
  • M.W. Witthaus et al.

    Bladder oxidative stress in sleep apnea contributes to detrusor instability and nocturia

    J. Urol.

    (2015)
  • E.A. Manci et al.

    Pathology of Berkeley sickle cell mice: similarities and differences with human sickle cell disease

    Blood

    (2006)
  • A. Ridha et al.

    Acute compartment syndrome secondary to rhabdomyolysis in a sickle cell trait patient

    Lancet

    (2014)
  • K.E. Andersson et al.

    Pharmacology of the lower urinary tract: basis for current and future treatments of urinary incontinence

    Pharmacol. Rev.

    (2004)
  • M.C. Michel et al.

    Basic mechanisms of urgency: preclinical and clinical evidence

    Eur. Urol.

    (2009)
  • R.S. Eapen et al.

    Review of the epidemiology of overactive bladder

    Res. Rep. Urol.

    (2016)
  • P. Hedlund

    Nitric oxide/cGMP‐mediated effects in the outflow region of the lower urinary tract‐is there a basis for pharmacological targeting of cGMP?

    World J. Urol.

    (2005)
  • F.Z. Mónica et al.

    Long-term nitric oxide deficiency causes muscarinic supersensitivity and reduces beta(3)-adrenoceptor-mediated relaxation, causing rat detrusor overactivity

    Br. J. Pharmacol.

    (2008)
  • A.L. Burnett et al.

    Urinary bladder‐urethral sphincter dysfunction in mice with targeted disruption of neuronal nitric oxide synthase models idiopathic voiding disorders in humans

    Nat. Med.

    (1997)
  • Y. Kamiyama et al.

    Inhibitory effects of nicorandil, a K ATP channel opener and a nitric oxide donor, on overactive bladder in animal models

    BJU Int.

    (2008)
  • K. Persson et al.

    Functional characteristics of urinary tract smooth muscles in mice lacking cGMP protein kinase type I

    Am. J. Physiol. Regul. Integr. Comp. Physiol.

    (2000)
  • A. Moon

    Influence of nitric oxide signalling pathways on pre‐contracted human detrusor smooth muscle in vitro

    BJU Int.

    (2002)
  • K. Persson et al.

    Effects of inhibition of the L-arginine/nitric oxide pathway in the rat lower urinary tract in vivo and in vitro

    Br. J. Pharmacol.

    (1992)
  • Y. Satake et al.

    Crucial roles of nitric oxide synthases in β-adrenoceptor-mediated bladder relaxation in mice

    Am. J. Physiol. Renal. Physiol.

    (2017)
  • E. Meng et al.

    Neuronal-derived nitric oxide modulates the activity of mouse detrusor smooth muscle

    Neurourol. Urodyn.

    (2012)
  • Y. Yanai et al.

    Role of nitric oxide/cyclic GMP pathway in regulating spontaneous excitations in detrusor smooth muscle of the Guinea‐pig bladder

    Neurourol. Urodyn.

    (2008)
  • U.A. Anele et al.

    Overactive bladder in adults with sickle cell disease

    Neurourol. Urodyn.

    (2016)
  • Cited by (4)

    • Blockading a new NSCLC immunosuppressive target by pluripotent autologous tumor vaccines magnifies sequential immunotherapy

      2022, Bioactive Materials
      Citation Excerpt :

      Adequate experimental results confirmed that PDE5i exhibited inhibitory efficiency against lung cancer in both in vitro and in vivo assays. PDE5 singling pathways are closely associated with NO and NO can serve as an immune enhancement agent for boosting immunotherapy [24,31–33]. Regarding this, LA as the endogenous NO donor is justifiably believed to be qualified for enhancing PDE5i-activated immune responses via generating abundant NO or cGMP [34].

    View full text