Open Access

A high salt diet impairs the bladder epithelial barrier and activates the NLRP3 and NF‑κB signaling pathways to induce an overactive bladder in vivo

  • Authors:
    • Jingwen Xue
    • Zhipeng Zhou
    • Zhangrui Zhu
    • Qi Sun
    • Yuexuan Zhu
    • Peng Wu
  • View Affiliations

  • Published online on: July 12, 2024     https://doi.org/10.3892/etm.2024.12651
  • Article Number: 362
  • Copyright : © Xue et al. This is an open access article distributed under the terms of Creative Commons Attribution License [CC BY 4.0].

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Overactive bladder (OAB) is a condition characterized by an urgency to urinate, which is associated with the urodynamic observation of detrusor overexcitation. Although the etiology of OAB is currently unclear, it has been suggested that in patients with OAB, disruption of bladder epithelial barrier integrity can disturb the normal contractile function of the detrusor. Additionally, dietary preferences have been suggested to influence the severity of OAB. Therefore, the aim of the present study was to investigate the effect of a high salt diet (HSD) on the development of OAB in a murine model. Mice were fed either a HSD or standard diet for 8 weeks, following which voiding characteristics and bladder barrier function were assessed. The present study demonstrated that a HSD in mice was associated with OAB‑like symptoms such as increased urinary frequency and non‑voiding bladder contractions. The HSD group demonstrated a thinner bladder mucus layer and decreased expression of bladder barrier markers, tight junction protein‑1 and claudin‑1, which may be potentially indicative of induced bladder damage. A HSD for 8 weeks in mice and a high salt treatment at the uroepithelium cellular (SV‑HUC‑1s) level resulted in increased uroepithelial oxidative stress and inflammatory cell infiltration, as indicated by increased expression levels of TNF‑α and IL‑1β, as well as activation of the nucleotide‑binding domain leucine‑rich‑containing family pyrin domain‑containing 3 (NLRP3) and NF‑κB signaling pathways in vivo and in vitro. Therefore, the present study indicated that a HSD could be a potentially important risk factor for the development of OAB, as it may be associated with overactivation of contractile function of the bladder by impairing the integrity of the bladder epithelial barrier and activation of the NLRP3 and NF‑κB signaling pathways. Remodeling of the bladder barrier and reduction of the inflammatory response may be potential targets for the treatment of OAB in the future.

Related Articles

Journal Cover

September-2024
Volume 28 Issue 3

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Xue J, Zhou Z, Zhu Z, Sun Q, Zhu Y and Wu P: A high salt diet impairs the bladder epithelial barrier and activates the NLRP3 and NF‑&kappa;B signaling pathways to induce an overactive bladder <em>in</em> <em>vivo</em>. Exp Ther Med 28: 362, 2024
APA
Xue, J., Zhou, Z., Zhu, Z., Sun, Q., Zhu, Y., & Wu, P. (2024). A high salt diet impairs the bladder epithelial barrier and activates the NLRP3 and NF‑&kappa;B signaling pathways to induce an overactive bladder <em>in</em> <em>vivo</em>. Experimental and Therapeutic Medicine, 28, 362. https://doi.org/10.3892/etm.2024.12651
MLA
Xue, J., Zhou, Z., Zhu, Z., Sun, Q., Zhu, Y., Wu, P."A high salt diet impairs the bladder epithelial barrier and activates the NLRP3 and NF‑&kappa;B signaling pathways to induce an overactive bladder <em>in</em> <em>vivo</em>". Experimental and Therapeutic Medicine 28.3 (2024): 362.
Chicago
Xue, J., Zhou, Z., Zhu, Z., Sun, Q., Zhu, Y., Wu, P."A high salt diet impairs the bladder epithelial barrier and activates the NLRP3 and NF‑&kappa;B signaling pathways to induce an overactive bladder <em>in</em> <em>vivo</em>". Experimental and Therapeutic Medicine 28, no. 3 (2024): 362. https://doi.org/10.3892/etm.2024.12651