Int J Stem Cells.  2025 Feb;18(1):72-86. 10.15283/ijsc23147.

Assessment of the Therapeutic Effectiveness of Glutathione-Enhanced Mesenchymal Stem Cells in Rat Models of Chronic Bladder Ischemia-Induced Overactive Bladder and Detrusor Underactivity

Affiliations
  • 1Urology Institute, Ewha Womans University Mokdong Hospital, Seoul, Korea
  • 2UroGyn Efficacy Evaluation Center, Institute of Convergence Medicine, Ewha Womans University Mokdong Hospital, Seoul, Korea
  • 3Department of Cell and Genetic Engineering, Asan Medical Center, Brain Korea 21 Project,University of Ulsan College of Medicine, Seoul, Korea
  • 4Center for Cell Therapy, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
  • 5Department of Urology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea

Abstract

Overactive bladder (OAB) and detrusor underactivity (DUA) are representative voiding dysfunctions with a chronic nature and limited treatment modalities, and are ideal targets for stem cell therapy. In the present study, we investigated the therapeutic efficacy of human mesenchymal stem cells (MSCs) with a high antioxidant capacity generated by the Primed Fresh OCT4 (PFO) procedure in chronic bladder ischemia (CBI)-induced OAB and DUA rat models. Sixteen-week-old male Sprague-Dawley rats were divided into three groups (sham, OAB or DUA, and stem cell groups; n=10, respectively). CBI was induced by bilateral iliac arterial injury (OAB, 10 times; DUA, 30 times) followed by a 1.25% cholesterol diet for 8 weeks. Seven weeks after injury, rats in the stem cell and other groups were injected with 1×10 6 PFO-MSCs and phosphate buffer, respectively. One week later, bladder function was analyzed by awake cystometry and bladders were harvested for histological analysis. CBI with a high-fat diet resulted in atrophy of smooth muscle and increased collagen deposits correlating with reduced detrusor contractility in both rat models. Arterial injury 10 and 30 times induced OAB (increased number of non-voiding contractions and shortened micturition interval) and DUA (prolonged micturition interval and increased residual volume), respectively. Injection of PFO-MSCs with the enhanced glutathione dynamics reversed both functional and histological changes; it restored the contractility, micturition interval, residual volume, and muscle layer, with reduced fibrosis. CBI followed by a high-fat diet with varying degrees of arterial injury induced OAB and DUA in rats. In addition, PFO-MSCs alleviated functional and histological changes in both rat models.

Keyword

Urinary bladder; Overactive; Underactive; Adult stem cell; Glutathoine

Figure

  • Fig. 1 The Primed Fresh OCT4 (PFO) procedure enhances the core functions of human umbilical cord derived-mesenchymal stem cells (hUC-MSCs). (A) Microscopic analysis of normal (naïve) hUC-MSCs and PFO-MSCs. Scale bar=1 mm. (B) Real-time monitoring of the glutathione (GSH)-recovering capacity (GRC) and basal GSH levels in naïve MSCs and PFO-MSCs upon exposure to 200 μM diamide (arrow). The GSH dynamics of each sample were quantified based on the initial F510/F580 fluorescence ratio (FR) (for baseline total GSH) and slope after diamide treatment (for GRC), as previously described (11). (C) Flow cytometry analyses of expression of MSC surface proteins (CD29 and CD90) and hematopoietic lineage markers (CD34 and CD45) in naïve MSCs and PFO-MSCs. An IgG isotype antibody was used as a control. (D) Differentiation into osteogenic (left panel, magnification ×200, scale bar=200 μm), chondrogenic (center panel, magnification ×100, scale bar=200 μm), and adipogenic (right panel, magnification ×400, scale bar=50 μm) lineages, determined using Alizarin Red S, Alcian Blue, and Oil Red O staining, respectively. (E-G) The beneficial effects of the PFO procedure in hUC-MSCs on cell proliferation (n=3, E), the colony forming unit-fibroblast (CFU-F) potency (n=3, F), and chemotaxis (n=7, G) in response to treatment with 10 ng/mL PDGF-AA. Representative results for each assay are shown on the left (magnification ×200, scale bar=100 μm in G). Cell proliferation was determined by the MTT assay. Quantitative data are presented as ratios relative to the naïve control group and are expressed as the mean±SEM. *p<0.05, **p<0.01, ***p<0.001 compared with the naïve group. Statistical analyses were performed by unpaired t-test (F, G) or two-way ANOVA (B, E).

  • Fig. 2 Mesenchymal stem cell enriched by the Primed Fresh OCT4 procedure (PFO-MSC) injection ameliorates bladder function in the overactive bladder (OAB) rat model. (A) Representative awake cystometry results and (B) quantitative analysis of bladder voiding parameters at 1 week post-injection of 1×106 PFO-MSCs into rat bladders. Sham: sham-operated. Quantitative results from ten independent animals per group are presented as the mean±SEM. **p<0.01, ***p<0.001 compared with the OAB group. A one-way ANOVA with the Bonferroni post-hoc test was used for statistical analysis. IAP: intraabdominal pressure, IVP: intravesical pressure.

  • Fig. 3 Histological analysis of the effects of mesenchymal stem cell enriched by the Primed Fresh OCT4 procedure (PFO-MSC) injection on overactive bladder (OAB) bladder injury. (A-C) Representative images of H&E staining (magnification ×100, scale bar=100 μm, A), immunohistochemical staining of α-smooth muscle actin (magnification ×100, scale bar=100 μm, B), and Masson’s trichrome staining (magnification ×200, scale bar=100 μm, C) in bladder tissues at 1 week after injection of PFO-MSCs. (D) Representative images of terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) staining to detect apoptotic cells (magnification ×400, scale bar=100 μm) in bladder tissue from the indicated groups. Nuclei were stained with 4’6-diamino-2-phenylindole (DAPI). The percentage of apoptotic cells (green) was derived by calculating the ratio of apoptotic cells (TUNEL positive) to total cells (DAPI-stained nuclei). All quantitative data are presented as the mean±SEM (n=10). **p<0.01, ***p<0.001 compared with the OAB group. A one-way ANOVA with the Bonferroni post-hoc test was used for statistical analysis.

  • Fig. 4 Mesenchymal stem cell enriched by the Primed Fresh OCT4 procedure (PFO-MSC) injection ameliorates bladder function in the detrusor underactivity (DUA) rat model. (A) Representative awake cystometry results and (B) quantitative analysis of bladder voiding parameters at 1 week post-injection of 1×106 PFO-MSCs into rat bladders. Quantitative results from 10 independent animals per group are presented as the mean±SEM. *p<0.05, **p<0.01, ***p<0.001 compared with the DUA group. A one-way ANOVA with the Bonferroni post-hoc test was used for statistical analysis. IAP: intraabdominal pressure, IVP: intravesical pressure.

  • Fig. 5 Histological analysis of the effects of mesenchymal stem cell enriched by the Primed Fresh OCT4 procedure (PFO-MSC) injection on detrusor underactivity (DUA) bladder injury. (A-C) Representative images of H&E staining (magnification ×100, scale bar=100 μm, A), immunohistochemical staining of α-smooth muscle actin (magnification ×100, scale bar=100 μm, B), and Masson’s trichrome staining (magnification ×200, scale bar=100 μm, C) in bladder tissues at 1 week after injection of PFO-MSCs. (D) Representative images (left panel) and quantitation right panel) of terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) staining to detect apoptotic cells (green, magnification ×400, scale bar=100 μm) in bladder tissue from the indicated groups. Nuclei were stained with 4’6-diamino-2-phenylindole (DAPI). All quantitative data are presented as the mean±SEM (n=10). **p<0.01, ***p<0.001 compared with the DUA group. A one-way ANOVA with the Bonferroni post-hoc test was used for statistical analysis.

  • Fig. 6 Engraftment of mesenchymal stem cells enriched by the Primed Fresh OCT4 procedure (PFO-MSCs) in chronic bladder ischemia (CBI) induced detrusor underactivity (DUA) bladders. (A, B) Representative images (magnification ×400, scale bar= 100 μm) showing immunofluorescence staining of human B2M (hB2M, green) in overactive bladder (OAB) (A) or DUA (B) bladder tissues from the indicated groups at 1 week post- injection of PFO-MSCs. Nuclei were stained with 4’6-diamino-2-phenylindole (DAPI, blue). It is noteworthy that hB2M+ cells engrafted primarily as pericytes and located near muscle fibers within the DUA bladders.

  • Fig. 7 Injection of Primed Fresh OCT4 (PFO) induces angiogenesis in bladders with chronic bladder ischemia (CBI). (A, B) Representative images showing immunohistochemical staining of CD31 (magnification ×100, scale bar=100 μm) in overactive bladder (OAB) (A) or detrusor underactivity (DUA) (B) bladder tissues 1 week post-transplantation of mesenchymal stem cells enriched by the PFO procedure (PFO-MSCs). Nuclei were stained with Mayer’s hematoxylin. The number of CD31+ vessels in the right panel is presented as the mean±SEM (n=5). (C) Changes in the expression of gene transcripts related to angiogenesis in naïve or PFO-MSCs. Expression is presented as the % of GAPDH (n=3). *p<0.05, **p<0.01, ***p<0.001, compared with CBI groups (one-way ANOVA with Bonferroni post-hoc test, C), or compared with naïve cells (unpaired t-test, D). TEK: TEK receptor tyrosine kinase, HGF: hepatocyte growth factor.


Reference

References

1. Bartoli S, Aguzzi G, Tarricone R. 2010; Impact on quality of life of urinary incontinence and overactive bladder: a systematic literature review. Urology. 75:491–500. DOI: 10.1016/j.urology.2009.07.1325. PMID: 19962738.
2. Peyronnet B, Mironska E, Chapple C, et al. 2019; A comprehensive review of overactive bladder pathophysiology: on the way to tailored treatment. Eur Urol. 75:988–1000. DOI: 10.1016/j.eururo.2019.02.038. PMID: 30922690.
3. Aldamanhori R, Chapple CR. 2017; Underactive bladder, detrusor underactivity, definition, symptoms, epidemiology, etiopathogenesis, and risk factors. Curr Opin Urol. 27:293–299. DOI: 10.1097/MOU.0000000000000381. PMID: 28221218.
4. Raju R, Linder BJ. 2020; Evaluation and treatment of overactive bladder in women. Mayo Clin Proc. 95:370–377. DOI: 10.1016/j.mayocp.2019.11.024. PMID: 32029089.
5. Aggarwal H, Zimmern PE. 2016; Underactive bladder. Curr Urol Rep. 17:17. DOI: 10.1007/s11934-016-0582-6. PMID: 26874529.
6. Zakrzewski W, Dobrzyński M, Szymonowicz M, Rybak Z. 2019; Stem cells: past, present, and future. Stem Cell Res Ther. 10:68. DOI: 10.1186/s13287-019-1165-5. PMID: 30808416. PMCID: PMC6390367.
7. Kim M, Yu HY, Ju H, et al. 2019; Induction of detrusor underactivity by extensive vascular endothelial damages of iliac arteries in a rat model and its pathophysiology in the genetic levels. Sci Rep. 9:16328. DOI: 10.1038/s41598-019-52811-4. PMID: 31705030. PMCID: PMC6841737.
8. Yu HY, Shin JH, Yun H, et al. 2021; A preclinical study of human embryonic stem cell-derived mesenchymal stem cells for treating detrusor underactivity by chronic bladder ischemia. Stem Cell Rev Rep. 17:2139–2152. DOI: 10.1007/s12015-021-10204-z. PMID: 34189670. PMCID: PMC8599399.
9. Kim Y, Jin HJ, Heo J, et al. 2018; Small hypoxia-primed mesenchymal stem cells attenuate graft-versus-host disease. Leukemia. 32:2672–2684. DOI: 10.1038/s41375-018-0151-8. PMID: 29789652. PMCID: PMC6286327.
10. Jeong EM, Yoon JH, Lim J, et al. 2018; Real-time monitoring of glutathione in living cells reveals that high glutathione levels are required to maintain stem cell function. Stem Cell Reports. 10:600–614. DOI: 10.1016/j.stemcr.2017.12.007. PMID: 29307581. PMCID: PMC5830891.
11. Lim J, Heo J, Ju H, et al. 2020; Glutathione dynamics determine the therapeutic efficacy of mesenchymal stem cells for graft-versus-host disease via CREB1-NRF2 pathway. Sci Adv. 6:eaba1334. DOI: 10.1126/sciadv.aba1334. PMID: 32490200. PMCID: PMC7239701.
12. Lim J, Lee S, Ju H, et al. 2017; Valproic acid enforces the priming effect of sphingosine-1 phosphate on human mesenchymal stem cells. Int J Mol Med. 40:739–747. DOI: 10.3892/ijmm.2017.3053. PMID: 28677769. PMCID: PMC5547989.
13. Lim J, Heo J, Yu HY, et al. 2021; Small-sized mesenchymal stem cells with high glutathione dynamics show improved therapeutic potency in graft-versus-host disease. Clin Transl Med. 11:e476. DOI: 10.1002/ctm2.476. PMID: 34323414. PMCID: PMC8255063.
14. Ju H, Yun H, Kim Y, et al. 2023; Activating transcription factor-2 supports the antioxidant capacity and ability of human mesenchymal stem cells to prevent asthmatic airway inflammation. Exp Mol Med. 55:413–425. DOI: 10.1038/s12276-023-00943-z. PMID: 36765266. PMCID: PMC9981582.
15. Kim J, Gong YX, Jeong EM. 2023; Measuring glutathione regeneration capacity in stem cells. Int J Stem Cells. 16:356–362. DOI: 10.15283/ijsc23047. PMID: 37385637. PMCID: PMC10465335.
16. Jeong EM, Shin JW, Lim J, et al. 2019; Monitoring glutathione dynamics and heterogeneity in living stem cells. Int J Stem Cells. 12:367–379. DOI: 10.15283/ijsc18151. PMID: 30836726. PMCID: PMC6657947.
17. Mushahary D, Spittler A, Kasper C, Weber V, Charwat V. 2018; Isolation, cultivation, and characterization of human mesenchymal stem cells. Cytometry A. 93:19–31. DOI: 10.1002/cyto.a.23242. PMID: 29072818.
18. Lee S, Lim J, Lee JH, et al. 2020; Ascorbic acid 2-glucoside stably promotes the primitiveness of embryonic and mesenchymal stem cells through ten-eleven translocation- and cAMP-responsive element-binding protein-1-dependent mechanisms. Antioxid Redox Signal. 32:35–59. DOI: 10.1089/ars.2019.7743. PMID: 31656084.
19. Shin JH, Ryu CM, Yu HY, et al. 2022; Safety of human embryonic stem cell-derived mesenchymal stem cells for treating interstitial cystitis: a phase I study. Stem Cells Transl Med. 11:1010–1020. DOI: 10.1093/stcltm/szac065. PMID: 36069837. PMCID: PMC9585946.
20. Yu HY, Lee S, Ju H, et al. 2022; Intravital imaging and single cell transcriptomic analysis for engraftment of mesenchymal stem cells in an animal model of interstitial cystitis/bladder pain syndrome. Biomaterials. 280:121277. DOI: 10.1016/j.biomaterials.2021.121277. PMID: 34861510.
21. Kim Y, Ju H, Yoo SY, et al. 2023; Glutathione dynamics is a potential predictive and therapeutic trait for neoadjuvant chemotherapy response in bladder cancer. Cell Rep Med. 4:101224. DOI: 10.1016/j.xcrm.2023.101224. PMID: 37797616. PMCID: PMC10591055.
22. Heo J, Noh BJ, Lee S, et al. 2020; Phosphorylation of TFCP2L1 by CDK1 is required for stem cell pluripotency and bladder carcinogenesis. EMBO Mol Med. 12:e10880. DOI: 10.15252/emmm.201910880. PMID: 31709755. PMCID: PMC6949511.
23. Heo J, Lee J, Nam YJ, et al. 2022; The CDK1/TFCP2L1/ID2 cascade offers a novel combination therapy strategy in a preclinical model of bladder cancer. Exp Mol Med. 54:801–811. DOI: 10.1038/s12276-022-00786-0. PMID: 35729325. PMCID: PMC9256744.
24. Ryu CM, Yu HY, Lee HY, et al. 2018; Longitudinal intravital imaging of transplanted mesenchymal stem cells elucidates their functional integration and therapeutic potency in an animal model of interstitial cystitis/bladder pain syndrome. Theranostics. 8:5610–5624. DOI: 10.7150/thno.27559. PMID: 30555567. PMCID: PMC6276303.
25. Heo J, Lim J, Lee S, et al. 2017; Sirt1 regulates DNA methylation and differentiation potential of embryonic stem cells by antagonizing Dnmt3l. Cell Rep. 18:1930–1945. DOI: 10.1016/j.celrep.2017.01.074. PMID: 28228259.
26. Li C, Hu Y. 2023; Extracellular vesicles derived from mesenchymal stem cells as cell-free therapy for intrauterine adhesion. Int J Stem Cells. 16:260–268. DOI: 10.15283/ijsc21177. PMID: 37385632. PMCID: PMC10465336.
27. Nambiar AK, Arlandis S, Bø K, et al. 2022; European Association of Urology Guidelines on the diagnosis and management of female non-neurogenic lower urinary tract symptoms. Part 1: diagnostics, overactive bladder, stress urinary incontinence, and mixed urinary incontinence. Eur Urol. 82:49–59. DOI: 10.1016/j.eururo.2022.01.045. PMID: 35216856.
28. Arlandis S, Bø K, Cobussen-Boekhorst H, et al. 2022; European Association of Urology Guidelines on the management of female non-neurogenic lower urinary tract symptoms. Part 2: underactive bladder, bladder outlet obstruction, and nocturia. Eur Urol. 82:60–70. DOI: 10.1016/j.eururo.2022.01.044. PMID: 35181193.
29. Maher RL, Hanlon J, Hajjar ER. 2014; Clinical consequences of polypharmacy in elderly. Expert Opin Drug Saf. 13:57–65. DOI: 10.1517/14740338.2013.827660. PMID: 24073682. PMCID: PMC3864987.
30. Shin JH, Ryu CM, Yu HY, Shin DM, Choo MS. 2020; Current and future directions of stem cell therapy for bladder dysfunction. Stem Cell Rev Rep. 16:82–93. DOI: 10.1007/s12015-019-09922-2. PMID: 31758372. PMCID: PMC6987049.
31. Han S, Yim HW, Jeong H, Choi S, Han S. 2023; Establishing rationale for the clinical development of cell therapy products: consensus between risk and benefit. Int J Stem Cells. 16:16–26. DOI: 10.15283/ijsc21189. PMID: 36581365. PMCID: PMC9978837.
32. Azadzoi KM, Chen BG, Radisavljevic ZM, Siroky MB. 2011; Molecular reactions and ultrastructural damage in the chronically ischemic bladder. J Urol. 186:2115–2122. DOI: 10.1016/j.juro.2011.06.047. PMID: 21944111.
33. Bosch R, Abrams P, Averbeck MA, et al. 2019; Do functional changes occur in the bladder due to bladder outlet obstruction? - ICI-RS 2018. Neurourol Urodyn. 38(Suppl 5):S56–S65. DOI: 10.1002/nau.24076. PMCID: PMC6915908.
34. Fusco F, Creta M, De Nunzio C, et al. 2018; Progressive bladder remodeling due to bladder outlet obstruction: a systematic review of morphological and molecular evidences in humans. BMC Urol. 18:15. DOI: 10.1186/s12894-018-0329-4. PMID: 29519236. PMCID: PMC5844070.
35. Aizawa N, Igawa Y. 2017; Pathophysiology of the underactive bladder. Investig Clin Urol. 58(Suppl 2):S82–S89. DOI: 10.4111/icu.2017.58.S2.S82. PMID: 29279880. PMCID: PMC5740034.
Full Text Links
  • IJSC
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2025 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr