Low-Intensity Extracorporeal Shock Wave Therapy Promotes Bladder Regeneration and Improves Overactive Bladder Induced by Ovarian Hormone Deficiency from Rat Animal Model to Human Clinical Trial

Kun-Ling Lin, Jian-He Lu, Kuang-Shun Chueh, Tai-Jui Juan, Bin-Nan Wu, Shu-Mien Chuang, Yung-Chin Lee, Mei-Chen Shen, Cheng-Yu Long, Yung-Shun Juan, Kun-Ling Lin, Jian-He Lu, Kuang-Shun Chueh, Tai-Jui Juan, Bin-Nan Wu, Shu-Mien Chuang, Yung-Chin Lee, Mei-Chen Shen, Cheng-Yu Long, Yung-Shun Juan

Abstract

Postmenopausal women with ovary hormone deficiency (OHD) are subject to overactive bladder (OAB) symptoms. The present study attempted to elucidate whether low-intensity extracorporeal shock wave therapy (LiESWT) alters bladder angiogenesis, decreases inflammatory response, and ameliorates bladder hyperactivity to influence bladder function in OHD-induced OAB in human clinical trial and rat model. The ovariectomized (OVX) for 12 months Sprague-Dawley rat model mimicking the physiological condition of menopause was utilized to induce OAB and assess the potential therapeutic mechanism of LiESWT (0.12 mJ/mm2, 300 pulses, and 3 pulses/second). The randomized, single-blinded clinical trial was enrolled 58 participants to investigate the therapeutic efficacy of LiESWT (0.25 mJ/mm2, 3000 pulses, 3 pulses/second) on postmenopausal women with OAB. The results revealed that 8 weeks' LiESWT inhibited interstitial fibrosis, promoted cell proliferation, enhanced angiogenesis protein expression, and elevated the protein phosphorylation of ErK1/2, P38, and Akt, leading to decreased urinary frequency, nocturia, urgency, urgency incontinence, and post-voided residual urine volume, but increased voided urine volume and the maximal flow rate of postmenopausal participants. In conclusion, LiESWT attenuated inflammatory responses, increased angiogenesis, and promoted proliferation and differentiation, thereby improved OAB symptoms, thereafter promoting social activity and the quality of life of postmenopausal participants.

Keywords: bladder angiogenesis; low-intensity extracorporeal shock wave therapy; ovarian hormone deficiency; overactive bladder.

Conflict of interest statement

The authors declare no conflict of interest.

Figures

Figure 1
Figure 1
Urodynamic analysis of bladder cystometric parameters and voiding behavior shown in an OVX induced OAB of rat model. (A) Cystometry recordings of micturition pressure, voiding volume, and frequency, including voiding contraction (arrows) and non-voiding contraction (asterisks). (B) Tracing analysis of 24-h voiding behavior by metabolic cage. The OVX group significantly increased bladder maturation pressure, voiding contraction, non-voiding contraction, and micturition frequency, whereas LiESWT treatment significantly improved bladder voiding pattern and capacity.
Figure 2
Figure 2
Histopathological examination for bladder damage was shown by Masson’s trichrome staining, immunostaining, and Western blots. (AD, A′D′) The bladder pathological features of the sham group (A,A′), the OVX group (B,B′), the OVX + SW4 group (C,C′), and the OVX + SW8 group (D,D′). Masson’s trichrome stain showed the navy blue-stained nucleus, green-stained collagen, and red-stained smooth muscle were highlighted. In the sham group (A,A′), there were three to five layers of UL and only sparse collagen (yellow arrow) distributed in the SL. In the OVX group (B,B′), the morphology was characterized by thinner layer of UL (black arrows), and much collagen accumulation and interstitial fibrosis in the SL (yellow arrows). In contrast, the pathological features of the OVX + SW4 group (C,C′) and the OVX + SW8 group (D,D′) showed improved OVX-associated bladder damages by increasing a thicker layer of urothelium (black arrows) and reducing interstitial fibrosis (yellow arrows) compared with the OVX group. Particularly, there are many mononuclear cell groups gathered into sphere (yellow arrowheads) and much collagen accumulation (yellow arrows) around the sphere in the SL of the OVX + SW8 group (D,D′). (EH) The distribution of cell-adhesion marker E-cadherin by immunostaining was shown. In the sham group (E), the E-cadherin staining was found in intercellular junctions of urothelium. On the contrary, there was less E-cadherin staining expression in the thin urothelium of the OVX group (F), but the immunostaining of the OVX + SW4 group (G) and the OVX + SW8 group (H) was enhanced the staining in the UL. (I,J) Western blot was evaluated the protein levels of bladder inflammation (TGF-ß1 and COX-2), interstitial fibrosis (fibronectin and type I collagen), and urothelial structure (E-cadherin and uroplakin III). Both the inflammatory and fibrosis markers (TGF-ß1, fibronectin, type I collagen, and COX-2) were significantly increased in the OVX group compared to the sham group. However, the proteins noticeably decreased in the OVX + SW4 group and the OVX + SW8 group compared to the OVX group except TGF-ß1 expression in the OVX + SW4 group. Note: CK, cytokeratin; UL, urothelial layer; UBL, urothelial basal layer; SL, suburothelial layer; UPKIII, uroplakin III; TGF-ß1, transforming growth factor ß 1. Data were expressed as mean ± SD for n = 6, * p < 0.05; ** p < 0.01 versus the sham group. †p < 0.05; ††p < 0.01 versus the OVX group.
Figure 3
Figure 3
LiESWT improved OVX-induced pathological alteration and urothelial junction-associated protein expression. The expressions of proliferating and differential markers (Ki67, CK14 and CD44) and urothelial tight junction proteins (Claudin-4 and ZO-1) were assessed by immunofluorescence evaluation (AH) and Western blot (I,J). (AD) The staining of proliferation marker Ki67 was less distribution in the bladder tissues of the sham group (A), the OVX group (B) and the OVX + SW4 group (C). On the contrary, the Ki67 immunostaining was obviously expressed in the UBL and the sphere of SL in the OVX + SW8 group (D). (EH) Double-labeled analysis of Claudin-4 (fluorescein isothiocyanate; green, upper panels) and CK14 (rhodamine; red, lower panels) was widely distributed in the UL of the sham group (E). The co-staining was restricted to the thin and disrupted urothelium in OVX group (F). However, the labeling of the OVX + SW8 group (H) was markedly expressed in the UBL and the sphere of SL compared to the OVX group (F) and the OVX + SW4 group (G). Nuclear DNA was labeled with DAPI (blue). (I, J) The protein levels of Ki67, CK14, CD44, Claudin-4, and ZO-1 were investigated by Western blotting analysis. The protein levels noticeably enhanced in the OVX + SW8 group compared to the OVX group. Note: CK, cytokeratin; UL, urothelial layer; UBL, urothelial basal layer; SL, suburothelial layer. Values are means ± SD for n = 6. * p < 0.05; ** p < 0.01 versus the sham group. ††p < 0.01 versus the OVX group.
Figure 4
Figure 4
Proposed potential mechanism of regulating angiogenic remodeling triggered by LiESWT. The angiogenesis markers were analyzed by immunostaining (AD) and Western blots (EH). (AD) The α-SMA immunostaining widely distributed in smooth muscle of microvessels beneath UBL and vessels in the SL and muscular layer in the sham group (A). In the OVX group (B), the α-SMA expression was reduced in the SL compared to the sham group. However, the expression levels in the OVX + SW4 group (C) and the OVX + SW8 group (D) were increased in the SL and beneath UBL. (E,G) The levels of angiogenesis-associated proteins, including Laminin, Integrin-α6 (Laminin receptor), α-SMA, CD31, VEGF, VEGF-R1, and VEGF-R2 (VEGF receptor), were quantified by Western blots. Furthermore, the protein level in the OVX group was much lower than the sham group, except Integrin-α6. However, the expressions of angiogenesis markers were obviously increased in the OVX + SW4 group and the OVX + SW8 group compared with the OVX group. Therefore, LiESWT altered bladder angiogenic remodeling. Note: UL, urothelial layer; UBL, urothelial basal layer; SL, suburothelial layer; VEGF, vascular endothelial growth factor. (F,H): Proposed potential mechanism involved in regulating angiogenic remodeling triggered by LiESWT. The signaling-related kinases for angiogenic response, including Erk1/2, p-Erk1/2, P38, p-P38, C-Jun, p-C-Jun, Akt, and p-Akt, were quantified by Western blots. The expression levels of both Erk1/2 and Akt proteins were significantly declined in the OVX group, but the expression level of C-Jun was promoted as compared with the sham group. Besides, LiESWT treatment significantly promoted the phosphorylation of Erk1/2, P38, and Akt in the bladder as compared with the OVX group, whereas it reduced the phosphorylation of C-Jun. Therefore, LiESWT enhanced angiogenesis through the phosphorylation of P38, Erk1/2, and Akt. Values are means ± SD for n = 6. * p < 0.05; ** p < 0.01 versus the sham group. †p < 0.05; ††p < 0.01 versus the OVX group.
Figure 5
Figure 5
Analysis of postmenopausal participants with OAB symptoms. (A) The percentage of postmenopausal participants with OAB symptoms. OAB consisted of daytime frequency, nocturia urgency with or without urgency incontinence. (B) The therapeutic efficacy of LiESWT improved the OAB symptoms evaluated by 3-day urinary diary. The changes in daytime frequency, nocturia and urgency by 3-day urinary diary record at W4, W8, F1, F3, and F6 compared with W0. The mean times of daytime frequency, nocturia and urgency were noticeably decreased after LiESWT treatment. W0: the baseline, W4: 4-week of LiESWT treatment, W8: 8-week of LiESWT treatment, F1: 1-month follow-up, F3: 3-month follow-up, F6: 6-month follow-up. * p < 0.05; ** p < 0.01 as compared with W0.
Figure 6
Figure 6
Improvement in OAB symptoms and life bothersome questionnaire scores after LiESWT treatment. (A,C) The therapeutic effect of LiESWT was analyzed by the OAB symptoms and life bothersome questionnaires, including OABSS, ICIQ-SF, UDI-6, and IIQ-7. LiESWT treatment significantly reduced in the scores of OABSS, ICIQ-SF, UDI-6, and IIQ-7 as compared with the sham group. (B,D) Improvement of OABSS sub-scores for OAB symptoms after LiESWT treatment, including daytime frequency, nocturia, urgency, and urgency incontinence. LiESWT improved OAB symptoms and the QoL. Note: OABSS, overactive bladder symptom scores. ICIQ-SF: international consultation on incontinence questionnaire-short form, UDI-6: urogenital distress inventory-6, IIQ-7: incontinence impact questionnaire-7, W0: the baseline, W4: 4 weeks of LiESWT treatment, W8: 8 weeks of LiESWT treatment, F1: 1-month follow-up, F3: 3-month follow-up. The blue or orange font denotes the p-value before and after 4 weeks treatment in the sham group or in the LiESWT-treated group, respectively. The purple font indicates the p-value between the sham group and the LiESWT-treated group at the W0 and W4. * p < 0.05; ** p < 0.01 as compared with W0.
Figure 7
Figure 7
A study diagram proposed for the potential effect of LiESWT on improving the OAB symptoms. Part I: Rat animal experiment: LiESWT (0.12 mJ/mm2, 300 impulses, 3 Hz, and once/week) for OVX-induced OAB rat was placed on the lower abdomen. The effect of LiESWT enhanced anti-inflammation, promoted cell proliferation, and altered angiogenesis through VEGF/VEGF-R/MAPK (P38 and Erk1/2) pathways and Laminin/Integrin-α6/Akt pathways to improve OAB-induced by OHD. Part II: Human clinical trial: LiESWT (0.25 mJ/mm2, 3000 pulses, 3 Hz, and once/week) for postmenopausal OAB participants was placed on the lower abdomen with two fingers apart from the pubis. The results revealed that LiESWT improved bladder function and ameliorated the OAB symptoms after 8-week LiESWT. Note: OVX, ovariectomy; OHD, ovary hormone deficiency; OAB, overactive bladder; LiESWT, low-intensity extracorporeal shock wave therapy; CK, cytokeratin; VEGF, vascular endothelial growth factor; TGF-ß1, transforming growth factor ß 1.

References

    1. Hunskaar S., Burgio K., Diokno A., Herzog A.R., Hjalmas K., Lapitan M.C. Epidemiology and natural history of urinary incontinence in women. Urology. 2003;62:16–23. doi: 10.1016/S0090-4295(03)00755-6.
    1. Goldstein I., Dicks B., Kim N.N., Hartzell R. Multidisciplinary overview of vaginal atrophy and associated genitourinary symptoms in postmenopausal women. Sex Med. 2013;1:44–53. doi: 10.1002/sm2.17.
    1. Nappi R.E., Kokot-Kierepa M. Women’s voices in the menopause: Results from an international survey on vaginal atrophy. Maturitas. 2010;67:233–238. doi: 10.1016/j.maturitas.2010.08.001.
    1. Nappi R.E., Kokot-Kierepa M. Vaginal Health: Insights, Views & Attitudes (VIVA)—Results from an international survey. Climacteric. 2012;15:36–44. doi: 10.3109/13697137.2011.647840.
    1. Iosif C.S., Bekassy Z. Prevalence of genito-urinary symptoms in the late menopause. Acta Obs. Gynecol Scand. 1984;63:257–260. doi: 10.3109/00016348409155509.
    1. Bodner-Adler B., Alarab M., Ruiz-Zapata A.M., Latthe P. Effectiveness of hormones in postmenopausal pelvic floor dysfunction-International Urogynecological Association research and development-committee opinion. Int. Urogynecol. J. 2020;31:1577–1582. doi: 10.1007/s00192-019-04070-0.
    1. Cheng C.L., Li J.R., Lin C.H., de Groat W.C. Positive association of female overactive bladder symptoms and estrogen deprivation: A nationwide population-based cohort study in Taiwan. Medicine. 2016;95:e4107. doi: 10.1097/MD.0000000000004107.
    1. Weiderpass E., Baron J.A., Adami H.O., Magnusson C., Lindgren A., Bergström R., Correia N., Persson I. Low-potency oestrogen and risk of endometrial cancer: A case-control study. Lancet. 1999;353:1824–1828. doi: 10.1016/S0140-6736(98)10233-7.
    1. Robinson D., Toozs-Hobson P., Cardozo L. The effect of hormones on the lower urinary tract. Menopause Int. 2013;19:155–162. doi: 10.1177/1754045313511398.
    1. Teng J., Wang Z.Y., Jarrard D.F., Bjorling D.E. Roles of estrogen receptor alpha and beta in modulating urothelial cell proliferation. Endocr. Relat. Cancer. 2008;15:351–364. doi: 10.1677/erc.1.01255.
    1. Imamov O., Yakimchuk K., Morani A., Schwend T., Wada-Hiraike O., Razumov S., Warner M., Gustafsson J.-A. Gustafsson, Estrogen receptor beta-deficient female mice develop a bladder phenotype resembling human interstitial cystitis. Proc. Natl. Acad. Sci. USA. 2007;104:9806–9809. doi: 10.1073/pnas.0703410104.
    1. Long C.Y., Liu C.M., Hsu S.C., Wu C.H., Wang C.L., Tsai E.M. A randomized comparative study of the effects of oral and topical estrogen therapy on the vaginal vascularization and sexual function in hysterectomized postmenopausal women. Menopause. 2006;13:737–743. doi: 10.1097/01.gme.0000227401.98933.0b.
    1. Juan Y.S., Chuang S.M., Long C.Y., Chen C.H., Levin R.M., Liu K.M., Huang C.H. Neuroprotection of green tea catechins on surgical menopause-induced overactive bladder in a rat model. Menopause. 2012;19:346–354. doi: 10.1097/gme.0b013e31822d652d.
    1. Juan Y.S., Chuang S.M., Lee Y.L., Long C.Y., Wu T.H., Chang W.C., Levin R.M., Liu K.M., Huang C.H. Green tea catechins decrease oxidative stress in surgical menopause-induced overactive bladder in a rat model. BJU Int. 2012;110:e236–e244. doi: 10.1111/j.1464-410X.2012.11258.x.
    1. Hass M.A., Nichol P., Lee L., Levin R.M. Estrogen modulates permeability and prostaglandin levels in the rabbit urinary bladder. Prostaglandins Leukot Essent Fat. Acids. 2009;80:125–129. doi: 10.1016/j.plefa.2008.11.010.
    1. Lin A.D., Mannikarottu A., Kogan B.A., Whitbeck C., Chichester P., Leggett R.E., Levin R.M. Estrogen induces angiogenesis of the female rabbit bladder. J. Endocrinol. 2006;190:241–246. doi: 10.1677/joe.1.06701.
    1. Lin A.D., Levin R., Kogan B., Whitbeck C., Chichester P., Sokol R., Mannikarottu A. Estrogen induced functional hypertrophy and increased force generation of the female rabbit bladder. Neurourol Urodyn. 2006;25:473–479. doi: 10.1002/nau.20258.
    1. Lin A.D., Levin R.M., Kogan B.A., Whitbeck C., Leggett R.E., Kearns C., Mannikarottu A. Alteration of contractile and regulatory proteins in estrogen-induced hypertrophy of female rabbit bladder. Urology. 2006;68:1139–1143. doi: 10.1016/j.urology.2006.08.1094.
    1. Lee Y.L., Lin K.L., Wu B.N., Chuang S.M., Wu W.J., Lee Y.C., Ho W.T., Juan Y.S. Epigallocatechin-3-gallate alleviates bladder overactivity in a rat model with metabolic syndrome and ovarian hormone deficiency through mitochondria apoptosis pathways. Sci. Rep. 2018;8:5358. doi: 10.1038/s41598-018-23800-w.
    1. Aikawa K., Sugino T., Matsumoto S., Chichester P., Whitbeck C., Levin R.M. The effect of ovariectomy and estradiol on rabbit bladder smooth muscle contraction and morphology. J. Urol. 2003;170:634–637. doi: 10.1097/.
    1. Losordo D.W., Isner J.M. Estrogen and angiogenesis: A review. Arter. Thromb Vasc. Biol. 2001;21:6–12. doi: 10.1161/01.ATV.21.1.6.
    1. Przydacz M., Golabek T., Dudek P., Lipinski M., Chlosta P. Prevalence and bother of lower urinary tract symptoms and overactive bladder in Poland, an Eastern European Study. Sci. Rep. 2020;10:19819. doi: 10.1038/s41598-020-76846-0.
    1. Haylen B.T., de Ridder D., Freeman R.M., Swift S.E., Berghmans B., Lee J., Monga A., Petri E., Rizk D.E., Sand P.K., et al. International Urogynecological, S. International Continence, An International Urogynecological Association (IUGA)/International Continence Society (ICS) joint report on the terminology for female pelvic floor dysfunction. Neurourol. Urodyn. 2010;29:4–20. doi: 10.1002/nau.20798.
    1. Lapitan M.C., Chye P.L. The epidemiology of overactive bladder among females in Asia: A questionnaire survey. Int. Urogynecol. J. Pelvic Floor Dysfunct. 2001;12:226–231. doi: 10.1007/s001920170043.
    1. Soliman Y., Meyer R. Baum, Falls in the Elderly Secondary to Urinary Symptoms. Rev. Urol. 2016;18:28–32.
    1. Krause M., Wheeler T.L., 2nd, Snyder T.E., Richter H.E. Local Effects of Vaginally Administered Estrogen Therapy: A Review. J. Pelvic Med. Surg. 2009;15:105–114. doi: 10.1097/SPV.0b013e3181ab4804.
    1. Zimmermann R., Cumpanas A., Miclea F., Janetschek G. Extracorporeal shock wave therapy for the treatment of chronic pelvic pain syndrome in males: A randomised, double-blind, placebo-controlled study. Eur. Urol. 2009;56:418–424. doi: 10.1016/j.eururo.2009.03.043.
    1. Moayednia A., Haghdani S., Khosrawi S., Yousefi E., Vahdatpour B. Long-term effect of extracorporeal shock wave therapy on the treatment of chronic pelvic pain syndrome due to non bacterial prostatitis. J. Res. Med. Sci. 2014;19:293–296.
    1. Guu S.J., Geng J.H., Chao I.T., Lin H.T., Lee Y.C., Juan Y.S., Liu C.C., Wang C.J., Tsai C.C. Efficacy of Low-Intensity Extracorporeal Shock Wave Therapy on Men With Chronic Pelvic Pain Syndrome Refractory to 3-As Therapy. Am. J. Mens Health. 2018;12:441–452. doi: 10.1177/1557988317736585.
    1. Yuan P., Ma D., Zhang Y., Gao X., Liu Z., Li R., Wang T., Wang S., Liu J., Liu X. Efficacy of low-intensity extracorporeal shock wave therapy for the treatment of chronic prostatitis/chronic pelvic pain syndrome: A systematic review and meta-analysis. Neurourol. Urodyn. 2019;38:1457–1466. doi: 10.1002/nau.24017.
    1. Guu S.J., Liu C.C., Juan Y.S., Li C.C., Tsai C.C. The 12-month follow-up of the low-intensity extracorporeal shockwave therapy in the treatment of patients with chronic pelvic pain syndrome refractory to 3-As medications. Aging Male. 2020;23:793–800. doi: 10.1080/13685538.2019.1597341.
    1. Chung E., Wang J. A state-of-art review of low intensity extracorporeal shock wave therapy and lithotripter machines for the treatment of erectile dysfunction. Expert Rev. Med. Dev. 2017;14:929–934. doi: 10.1080/17434440.2017.1403897.
    1. Chung E., Cartmill R. Evaluation of clinical efficacy, safety and patient satisfaction rate after low-intensity extracorporeal shockwave therapy for the treatment of male erectile dysfunction: An Australian first open-label single-arm prospective clinical trial. BJU Int. 2015;115(Suppl. 5):46–49. doi: 10.1111/bju.13035.
    1. Clavijo R.I., Kohn T.P., Kohn J.R., Ramasamy R. Effects of Low-Intensity Extracorporeal Shockwave Therapy on Erectile Dysfunction: A Systematic Review and Meta-Analysis. J. Sex. Med. 2017;14:27–35. doi: 10.1016/j.jsxm.2016.11.001.
    1. Dong L., Chang D., Zhang X., Li J., Yang F., Tan K., Yang Y., Yong S., Yu X. Effect of Low-Intensity Extracorporeal Shock Wave on the Treatment of Erectile Dysfunction: A Systematic Review and Meta-Analysis. Am. J. Mens Health. 2019;13:1557988319846749. doi: 10.1177/1557988319846749.
    1. Sokolakis I., Hatzichristodoulou G. Clinical studies on low intensity extracorporeal shockwave therapy for erectile dysfunction: A systematic review and meta-analysis of randomised controlled trials. Int. J. Impot. Res. 2019;31:177–194. doi: 10.1038/s41443-019-0117-z.
    1. Myojo M., Ando J., Uehara M., Daimon M., Watanabe M., Komuro I. Feasibility of Extracorporeal Shock Wave Myocardial Revascularization Therapy for Post-Acute Myocardial Infarction Patients and Refractory Angina Pectoris Patients. Int. Heart J. 2017;58:185–190. doi: 10.1536/ihj.16-289.
    1. Lee Y.C., Chuang S.M., Lin K.L., Chen W.C., Lu J.H., Chueh K.S., Shen M.C., Liu L.W., Long C.Y., Juan Y.S. Low-Intensity Extracorporeal Shock Wave Therapy Ameliorates the Overactive Bladder: A Prospective Pilot Study. Biomed. Res. Int. 2020;2020:9175676. doi: 10.1155/2020/9175676.
    1. Long C.Y., Lin K.L., Lee Y.C., Chuang S.M., Lu J.H., Wu B.N., Chueh K.S., Ker C.R., Shen M.C., Juan Y.S. Therapeutic effects of Low intensity extracorporeal low energy shock wave therapy (LiESWT) on stress urinary incontinence. Sci. Rep. 2020;10:5818. doi: 10.1038/s41598-020-62471-4.
    1. Chuang Y.C., Meng E., Chancellor M., Kuo H.C. Pain reduction realized with extracorporeal shock wave therapy for the treatment of symptoms associated with interstitial cystitis/bladder pain syndrome-A prospective, multicenter, randomized, double-blind, placebo-controlled study. Neurourol. Urodyn. 2020;39:1505–1514. doi: 10.1002/nau.24382.
    1. Chen Y.T., Yang C.C., Sun C.K., Chiang H.J., Chen Y.L., Sung P.H., Zhen Y.Y., Huang T.H., Chang C.L., Chen H.H., et al. Extracorporeal shock wave therapy ameliorates cyclophosphamide-induced rat acute interstitial cystitis though inhibiting inflammation and oxidative stress-in vitro and in vivo experiment studies. Am. J. Transl. Res. 2014;6:631–648.
    1. Wang H.S., Oh B.S., Wang B., Ruan Y., Zhou J., Banie L., Lee Y.C., Tamaddon A., Zhou T., Wang G., et al. Low-intensity extracorporeal shockwave therapy ameliorates diabetic underactive bladder in streptozotocin-induced diabetic rats. BJU Int. 2018;122:490–500. doi: 10.1111/bju.14216.
    1. Lee Y.C., Hsieh T.J., Tang F.H., Jhan J.H., Lin K.L., Juan Y.S., Wang H.S., Long C.Y. Therapeutic effect of Low intensity Extracorporeal Shock Wave Therapy (Li-ESWT) on diabetic bladder dysfunction in a rat model. Int. J. Med. Sci. 2021;18:1423–1431. doi: 10.7150/ijms.55274.
    1. Gruenwald I., Appel B., Vardi Y. Low-intensity extracorporeal shock wave therapy—A novel effective treatment for erectile dysfunction in severe ED patients who respond poorly to PDE5 inhibitor therapy. J. Sex. Med. 2012;9:259–264. doi: 10.1111/j.1743-6109.2011.02498.x.
    1. Cohen J.M., Fagin A.P., Hariton E., Niska J.R., Pierce M.W., Kuriyama A., Whelan J.S., Jackson J.L., Dimitrakoff J.D. Therapeutic intervention for chronic prostatitis/chronic pelvic pain syndrome (CP/CPPS): A systematic review and meta-analysis. PLoS ONE. 2012;7:e41941. doi: 10.1371/journal.pone.0041941.
    1. Van Voorhis B.J. Genitourinary symptoms in the menopausal transition. Am. J. Med. 2005;118(Suppl. 12B):47–53. doi: 10.1016/j.amjmed.2005.10.004.
    1. Blakeman P.J., Hilton P., Bulmer J.N. Oestrogen and progesterone receptor expression in the female lower urinary tract, with reference to oestrogen status. BJU Int. 2000;86:32–38. doi: 10.1046/j.1464-410x.2000.00724.x.
    1. Ning N., Lin G., Lue T.F., Lin C.S. Effects of estrogen, raloxifene, and levormeloxifene on the expression of Rho-kinase signaling molecules in urethral smooth muscle cells. Urology. 2010;76:1517.e6–1517.e11. doi: 10.1016/j.urology.2010.07.470.
    1. Dobberfuhl A.D., Schuler C., Leggett R.E., De E.J.B., Levin R.M. Estrogen replacement is protective to the effect of in vitro hypoxia on female rabbit bladder and pelvic floor contractile response. Investig. Clin. Urol. 2020;61:432–440. doi: 10.4111/icu.2020.61.4.432.
    1. Gee E., Milkiewicz M., Haas T.L. p38 MAPK activity is stimulated by vascular endothelial growth factor receptor 2 activation and is essential for shear stress-induced angiogenesis. J. Cell Physiol. 2010;222:120–126. doi: 10.1002/jcp.21924.
    1. Fan N., Zhang M., Xu K., Ke X., Ding Y., Wang D., Liu Y., Ning Y., Deng X., He H. Serum level of vascular endothelial growth factor decreased in chronic ketamine abusers. Drug Alcohol Depend. 2015;152:57–61. doi: 10.1016/j.drugalcdep.2015.04.022.
    1. Zhu G.Q., Jeon S.H., Bae W.J., Choi S.W., Jeong H.C., Kim K.S., Kim S.J., Cho H.J., Ha U.S., Hong S.H., et al. Efficient Promotion of Autophagy and Angiogenesis Using Mesenchymal Stem Cell Therapy Enhanced by the Low-Energy Shock Waves in the Treatment of Erectile Dysfunction. Stem Cells Int. 2018;2018:1302672. doi: 10.1155/2018/1302672.
    1. Hassanpour M., Rezabakhsh A., Pezeshkian M., Rahbarghazi R., Nouri M. Distinct role of autophagy on angiogenesis: Highlights on the effect of autophagy in endothelial lineage and progenitor cells. Stem Cell Res. Ther. 2018;9:305. doi: 10.1186/s13287-018-1060-5.
    1. Hayashi D., Kawakami K., Ito K., Ishii K., Tanno H., Imai Y., Kanno E., Maruyama R., Shimokawa H., Tachi M. Low-energy extracorporeal shock wave therapy enhances skin wound healing in diabetic mice: A critical role of endothelial nitric oxide synthase. Wound Repair Regen. 2012;20:887–895. doi: 10.1111/j.1524-475X.2012.00851.x.
    1. Kuo Y.R., Wang C.T., Wang F.S., Chiang Y.C., Wang C.J. Extracorporeal shock-wave therapy enhanced wound healing via increasing topical blood perfusion and tissue regeneration in a rat model of STZ-induced diabetes. Wound Repair Regen. 2009;17:522–530. doi: 10.1111/j.1524-475X.2009.00504.x.
    1. Dumfarth J., Zimpfer D., Vogele-Kadletz M., Holfeld J., Sihorsch F., Schaden W., Czerny M., Aharinejad S., Wolner E., Grimm M. Prophylactic low-energy shock wave therapy improves wound healing after vein harvesting for coronary artery bypass graft surgery: A prospective, randomized trial. Ann. Thorac. Surg. 2008;86:1909–1913. doi: 10.1016/j.athoracsur.2008.07.117.
    1. Kuo Y.R., Wang C.T., Wang F.S., Yang K.D., Chiang Y.C., Wang C.J. Extracorporeal shock wave treatment modulates skin fibroblast recruitment and leukocyte infiltration for enhancing extended skin-flap survival. Wound Repair Regen. 2009;17:80–87. doi: 10.1111/j.1524-475X.2008.00444.x.
    1. Huang T.H., Sun C.K., Chen Y.L., Wang C.J., Yin T.C., Lee M.S., Yip H.K. Shock Wave Enhances Angiogenesis through VEGFR2 Activation and Recycling. Mol. Med. 2017;22:850–862. doi: 10.2119/molmed.2016.00108.
    1. Weihs A.M., Fuchs C., Teuschl A.H., Hartinger J., Slezak P., Mittermayr R., Redl H., Junger W.G., Sitte H.H., Runzler D. Shock wave treatment enhances cell proliferation and improves wound healing by ATP release-coupled extracellular signal-regulated kinase (ERK) activation. J. Biol. Chem. 2014;289:27090–27104. doi: 10.1074/jbc.M114.580936.
    1. Wang B., Zhou J., Banie L., Reed-Maldonado A.B., Ning H., Lu Z., Ruan Y., Zhou T., Wang H.S., Oh B.S., et al. Low-intensity extracorporeal shock wave therapy promotes myogenesis through PERK/ATF4 pathway. Neurourol. Urodyn. 2018;37:699–707. doi: 10.1002/nau.23380.
    1. Wang H.J., Lee W.C., Tyagi P., Huang C.C., Chuang Y.C. Effects of low energy shock wave therapy on inflammatory moleculars, bladder pain, and bladder function in a rat cystitis model. Neurourol. Urodyn. 2017;36:1440–1447. doi: 10.1002/nau.23141.
    1. Mariotto S., de Prati A.C., Cavalieri E., Amelio E., Marlinghaus E., Suzuki H. Extracorporeal shock wave therapy in inflammatory diseases: Molecular mechanism that triggers anti-inflammatory action. Curr. Med. Chem. 2009;16:2366–2372. doi: 10.2174/092986709788682119.
    1. Jiang D., Liang J., Fan J., Yu S., Chen S., Luo Y., Prestwich G.D., Mascarenhas M.M., Garg H.G., Quinn D.A., et al. Regulation of lung injury and repair by Toll-like receptors and hyaluronan. Nat. Med. 2005;11:1173–1179. doi: 10.1038/nm1315.
    1. Mondalek F.G., Fung K.M., Yang Q., Wu W., Lu W., Palmer B.W., Frimberger D.C., Greenwood-Van Meerveld B., Hurst R.E., Kropp B.P., et al. Temporal expression of hyaluronic acid and hyaluronic acid receptors in a porcine small intestinal submucosa-augmented rat bladder regeneration model. World J. Urol. 2015;33:1119–1128. doi: 10.1007/s00345-014-1403-5.
    1. Kaya G., Rodriguez I., Jorcano J.L., Vassalli P., Stamenkovic I. Selective suppression of CD44 in keratinocytes of mice bearing an antisense CD44 transgene driven by a tissue-specific promoter disrupts hyaluronate metabolism in the skin and impairs keratinocyte proliferation. Genes Dev. 1997;11:996–1007. doi: 10.1101/gad.11.8.996.
    1. Zhang X., Yan X., Wang C., Tang T., Chai Y. The dose-effect relationship in extracorporeal shock wave therapy: The optimal parameter for extracorporeal shock wave therapy. J. Surg. Res. 2014;186:484–492. doi: 10.1016/j.jss.2013.08.013.
    1. Wu A.K., Zhang X., Wang J., Ning H., Zaid U., Villalta J.D., Wang G., Banie L., Lin G., Lue T.F. Treatment of stress urinary incontinence with low-intensity extracorporeal shock wave therapy in a vaginal balloon dilation induced rat model. Transl. Androl. Urol. 2018;7:S7–S16. doi: 10.21037/tau.2017.12.36.
    1. Chuang Y.C., Huang T.L., Tyagi P., Huang C.C. Urodynamic and Immunohistochemical Evaluation of Intravesical Botulinum Toxin A Delivery Using Low Energy Shock Waves. J. Urol. 2016;196:599–608. doi: 10.1016/j.juro.2015.12.078.
    1. Juan Y.S., Lee Y.L., Long C.Y., Wong J.H., Jang M.Y., Lu J.H., Wu W.J., Huang Y.S., Chang W.C., Chuang S.M. Translocation of NF-kappaB and expression of cyclooxygenase-2 are enhanced by ketamine-induced ulcerative cystitis in rat bladder. Am. J. Pathol. 2015;185:2269–2285. doi: 10.1016/j.ajpath.2015.04.020.
    1. Liu K.M., Chuang S.M., Long C.Y., Lee Y.L., Wang C.C., Lu M.C., Lin R.J., Lu J.H., Jang M.Y., Wu W.J., et al. Ketamine-induced ulcerative cystitis and bladder apoptosis involve oxidative stress mediated by mitochondria and the endoplasmic reticulum. Am. J. Physiol. Renal Physiol. 2015;309:F318–F331. doi: 10.1152/ajprenal.00607.2014.
    1. Chuang S.M., Liu K.M., Li Y.L., Jang M.Y., Lee H.H., Wu W.J., Chang W.C., Levin R.M., Juan Y.S. Dual involvements of cyclooxygenase and nitric oxide synthase expressions in ketamine-induced ulcerative cystitis in rat bladder. Neurourol. Urodyn. 2013;32:1137–1143. doi: 10.1002/nau.22367.
    1. Homma Y., Yoshida M., Seki N., Yokoyama O., Kakizaki H., Gotoh M., Yamanishi T., Yamaguchi O., Takeda M., Nishizawa O. Symptom assessment tool for overactive bladder syndrome—Overactive bladder symptom score. Urology. 2006;68:318–323. doi: 10.1016/j.urology.2006.02.042.
    1. Ozberk S.S., Gundogar H., Ozkaya M., Taner I.L., Erciyas K. The effect of photobiomodulation therapy on nonsurgical periodontal treatment in patients with type 2 diabetes mellitus: A randomized controlled, single-blind, split-mouth clinical trial. Lasers Med. Sci. 2019;35:497–504. doi: 10.1007/s10103-019-02897-z.
    1. Rosario J.L. Relief from Back Pain Through Postural Adjustment: A Controlled Clinical Trial of the Immediate Effects of Muscular Chains Therapy (MCT) Int. J. Ther. Massage Bodyw. 2014;7:2–6. doi: 10.3822/ijtmb.v7i3.214.

Source: PubMed

3
Subscribe