1
|
Jasmine, Baraiya DH, Kavya TT, Mandal A, Chakraborty S, Sathish N, Francis CMR, Binoy Joseph D. Epithelial and mesenchymal compartments of the developing bladder and urethra display spatially distinct gene expression patterns. Dev Biol 2025; 520:155-170. [PMID: 39798644 DOI: 10.1016/j.ydbio.2025.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 01/02/2025] [Accepted: 01/07/2025] [Indexed: 01/15/2025]
Abstract
The lower urinary tract is comprised of the bladder and urethra and develops from the cloaca, a transient endoderm-derived structure formed from the caudal hindgut. After cloacal septation to form the urogenital sinus and anorectal tract, the bladder gradually develops from the anterior portion of the urogenital sinus while the urethra elongates distally into the genital tubercle. The bladder is a target for regenerative and reconstructive therapies but engineering an impermeable bladder epithelial lining has proven challenging. Urethral epithelial function, including its role as an active immune barrier, is poorly studied and neglected in regenerative therapy. A deeper understanding of epithelial patterning of the urogenital sinus by the surrounding mesenchyme, also accounting for sex-specific differences, can inform regenerative therapies. In this study, we identified spatially distinct genes in the epithelial and mesenchymal compartments of the developing mouse bladder and urethra that could be potential drivers of patterning in the lower urinary tract. Our data revealed spatially restricted domains of transcription factor expression in the epithelium that corresponded with bladder or urethra-specific differentiation. Additionally, we identified the genes Wnt2, Klf4 and Pitx2 that localize to the mesenchyme of the developing bladder and could be potential drivers of bladder differentiation. Our data revealed an increase in the expression of several chemokine genes including Cx3cl1 and Cxcl14 in the developing urethral epithelium that correlated with an increase in epithelial-associated macrophages in the urethra. A survey of sex-specific differences in epithelial and mesenchymal compartments revealed several differentially expressed genes between the male and female urethra but few sex-specific differences in bladder. By comparing spatially distinct gene expression in the developing lower urinary tract, our study provides insights into the divergent differentiation trajectories of the fetal bladder and urethra that establish their adult functions.
Collapse
Affiliation(s)
- Jasmine
- Institute for Stem Cell Science and Regenerative Medicine (iBRIC-inStem), GKVK-Post, Bellary Road, Bengaluru, Karnataka, 560065, India
| | - Divyeksha H Baraiya
- Institute for Stem Cell Science and Regenerative Medicine (iBRIC-inStem), GKVK-Post, Bellary Road, Bengaluru, Karnataka, 560065, India
| | - T T Kavya
- Institute for Stem Cell Science and Regenerative Medicine (iBRIC-inStem), GKVK-Post, Bellary Road, Bengaluru, Karnataka, 560065, India
| | - Aparna Mandal
- Institute for Stem Cell Science and Regenerative Medicine (iBRIC-inStem), GKVK-Post, Bellary Road, Bengaluru, Karnataka, 560065, India
| | - Shreya Chakraborty
- Institute for Stem Cell Science and Regenerative Medicine (iBRIC-inStem), GKVK-Post, Bellary Road, Bengaluru, Karnataka, 560065, India
| | - Neha Sathish
- Institute for Stem Cell Science and Regenerative Medicine (iBRIC-inStem), GKVK-Post, Bellary Road, Bengaluru, Karnataka, 560065, India
| | - Cynthia Marian Rebecca Francis
- Institute for Stem Cell Science and Regenerative Medicine (iBRIC-inStem), GKVK-Post, Bellary Road, Bengaluru, Karnataka, 560065, India
| | - Diya Binoy Joseph
- Institute for Stem Cell Science and Regenerative Medicine (iBRIC-inStem), GKVK-Post, Bellary Road, Bengaluru, Karnataka, 560065, India.
| |
Collapse
|
2
|
Sun JT, Pan CL, Mao YH, Wang Z, Sun JL, Zhang XX, Yang Y, Wei ZT, Xu YD. Exploring the protective effect and mechanism of icariside II on the bladder in a rat model of radiation cystitis based on transcriptome sequencing. Int J Radiat Biol 2024; 100:1493-1504. [PMID: 39166981 DOI: 10.1080/09553002.2024.2386982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 06/20/2024] [Accepted: 07/18/2024] [Indexed: 08/23/2024]
Abstract
PURPOSE Radiation cystitis (RC) is a complex and common complication after radiotherapy for pelvic cancer. Icariside II (ICAII) is a flavonoid compound extracted from Epimedium, a traditional Chinese medicine, with various pharmacological activities. The aim of the present study was to investigate the cysto-protective effects of ICAII in RC rats and its possible mechanisms. MATERIALS AND METHODS A rat model of induced radiation cystitis using pelvic X-ray irradiation was used, and bladder function was assessed by bladder volume and bladder leakage point pressure (LPP) after ICAII treatment. HE and Masson stains were used to assess the histopathological changes in the bladder. IL-6, TNF-α, IL-10, IL-4 and IL-1β were measured by ELISA to assess the level of inflammation. The gene-level changes in ICAII-treated RC were observed by transcriptome sequencing, and then the potential targets of action and biological mechanisms were explored by PPI, GO and KEGG enrichment analysis of the differentially expressed genes. Finally, the predicted targets of action were experimentally validated using immunohistochemistry, RT-qPCR, molecular docking and CETSA. RESULTS ICAII significantly increased bladder volume and the LPP, ameliorated pathological damage to bladder tissues, decreased the levels of IL-6, TNF-α, and IL-1β, and increased the levels of IL-10 and IL-4 in radiation-injured rats. A total of 90 differentially expressed genes were obtained by transcriptome sequencing, and PPI analysis identified H3F3C, ISG15, SPP1, and LCN2 as possible potential targets of action. GO and KEGG analyses revealed that these differentially expressed genes were mainly enriched in the pathways metabolism of xenobiotics by cytochrome P450, arachidonic acid metabolism, Staphylococcus aureus infection and chemical carcinogenesis - reactive oxygen species. Experimental validation showed that ICAII could significantly increase the expression of H3F3C and ISG15 and inhibit the expression of SPP1 and LCN2. ICAII binds well to H3F3C, ISG15, SPP1 and LCN2, with the best binding ability to H3F3C. Furthermore, ICAII inhibited the protein degradation of H3F3C in bladder epithelial cells. CONCLUSIONS ICAII may alleviate the bladder inflammatory response and inhibit the fibrosis process of bladder tissues through the regulation of H3F3C, ISG15, SPP1, and LCN2 targets and has a protective effect on the bladder of radioinjured rats. In particular, H3F3C may be one of the most promising therapeutic targets.
Collapse
Affiliation(s)
- Jun-Tao Sun
- Changchun University of Chinese Medicine, Changchun, China
| | - Chen-Li Pan
- Changchun University of Chinese Medicine, Changchun, China
| | - Yin-Hui Mao
- Changchun University of Chinese Medicine, Changchun, China
| | - Zhuo Wang
- Changchun University of Chinese Medicine, Changchun, China
| | - Ji-Lei Sun
- Department of Urology, The Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | | | - Yong Yang
- Department of Urology, The Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Zhi-Tao Wei
- Department of Urology, The Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Yong-De Xu
- Department of Urology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
3
|
McCloskey KD, Kanai A, Panicker JN, Hashitani H, Fry CH. What do we really know about the external urethral sphincter? CONTINENCE (AMSTERDAM, NETHERLANDS) 2024; 10:None. [PMID: 39669761 PMCID: PMC11636974 DOI: 10.1016/j.cont.2024.101223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2024]
Abstract
The external urethral sphincter (EUS), composed of skeletal muscle, along with a smooth muscle-lined internal urethral sphincter (IUS), have crucial roles in maintaining continence during bladder filling and facilitating urine flow during voiding. Disruption of this complex activity has profound consequences on normal lower urinary tract function during the micturition cycle. However, relatively little is known about the normal and pathological functions of these particular muscle types, how activity can be manipulated and regulated and why, for example, loss of EUS function and sarcopenia is associated with ageing. Here we discuss the unique physiological, biochemical and metabolic properties of striated and smooth muscle components of the urethral sphincter, which have distinct roles in maintaining continence during bladder filling. Relevant in vivo models for investigation of pathophysiological mechanisms, and for pre-clinical evaluation of therapeutic approaches are reviewed. Electromyography and Urethral Pressure Profile recordings are pivotal to understanding the function and dysfunction of the EUS and for clinical evaluation of e.g. urinary retention. Pre-clinical and clinical studies have revealed that age- or disease-related tissue remodelling that lead to filling/voiding disorders may be mitigated with emerging therapeutic approaches.
Collapse
Affiliation(s)
- Karen D. McCloskey
- Patrick G Johnston Centre for Cancer Research, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, 97 Lisburn Road, Belfast, Northern Ireland, UK
| | - Anthony Kanai
- Department of Medicine, Renal-Electrolyte Division, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jalesh N. Panicker
- Department of Uro-Neurology, The National Hospital for Neurology and Neurosurgery, Queen Square, London, UK
- UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London, London, UK
| | - Hikaru Hashitani
- Department of Cell Physiology, Graduate School of Medical Sciences, Nagoya City University, Nagoya 467-8601, Japan
| | - Christopher H. Fry
- University of Bristol, School of Physiology, Pharmacology and Neuroscience, Faculty of Life Sciences, Bristol BS8 1TD, UK
| |
Collapse
|
4
|
Apodaca G. Defining the molecular fingerprint of bladder and kidney fibroblasts. Am J Physiol Renal Physiol 2023; 325:F826-F856. [PMID: 37823192 PMCID: PMC10886799 DOI: 10.1152/ajprenal.00284.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 10/04/2023] [Accepted: 10/06/2023] [Indexed: 10/13/2023] Open
Abstract
Fibroblasts are integral to the organization and function of all organs and play critical roles in pathologies such as fibrosis; however, we have limited understanding of the fibroblasts that populate the bladder and kidney. In this review, I describe how transcriptomics is leading to a revolution in our understanding of fibroblast biology by defining the molecular fingerprint (i.e., transcriptome) of universal and specialized fibroblast types, revealing gene signatures that allows one to resolve fibroblasts from other mesenchymal cell types, and providing a new comprehension of the fibroblast lineage. In the kidney, transcriptomics is giving us new insights into the molecular fingerprint of kidney fibroblasts, including those for cortical fibroblasts, medullary fibroblasts, and erythropoietin (EPO)-producing Norn fibroblasts, as well as new information about the gene signatures of kidney myofibroblasts and the transition of kidney fibroblasts into myofibroblasts. Transcriptomics has also revealed that the major cell type in the bladder interstitium is the fibroblast, and that multiple fibroblast types, each with their own molecular fingerprint, are found in the bladder wall. Interleaved throughout is a discussion of how transcriptomics can drive our future understanding of fibroblast identification, diversity, function, and their roles in bladder and kidney biology and physiology in health and in disease states.
Collapse
Affiliation(s)
- Gerard Apodaca
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| |
Collapse
|
5
|
Clayton DR, Ruiz WG, Dalghi MG, Montalbetti N, Carattino MD, Apodaca G. Studies of ultrastructure, gene expression, and marker analysis reveal that mouse bladder PDGFRA + interstitial cells are fibroblasts. Am J Physiol Renal Physiol 2022; 323:F299-F321. [PMID: 35834272 PMCID: PMC9394772 DOI: 10.1152/ajprenal.00135.2022] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/07/2022] [Accepted: 07/06/2022] [Indexed: 11/22/2022] Open
Abstract
Fibroblasts are crucial to normal and abnormal organ and tissue biology, yet we lack basic insights into the fibroblasts that populate the bladder wall. Candidates may include bladder interstitial cells (also referred to as myofibroblasts, telocytes, and interstitial cells of Cajal-like cells), which express the fibroblast-associated marker PDGFRA along with VIM and CD34 but whose form and function remain enigmatic. By applying the latest insights in fibroblast transcriptomics, coupled with studies of gene expression, ultrastructure, and marker analysis, we observe the following: 1) that mouse bladder PDGFRA+ cells exhibit all of the ultrastructural hallmarks of fibroblasts including spindle shape, lack of basement membrane, abundant endoplasmic reticulum and Golgi, and formation of homotypic cell-cell contacts (but not heterotypic ones); 2) that they express multiple canonical fibroblast markers (including Col1a2, CD34, LY6A, and PDGFRA) along with the universal fibroblast genes Col15a1 and Pi16 but they do not express Kit; and 3) that PDGFRA+ fibroblasts include suburothelial ones (which express ACTA2, CAR3, LY6A, MYH10, TNC, VIM, Col1a2, and Col15a1), outer lamina propria ones (which express CD34, LY6A, PI16, VIM, Col1a2, Col15a1, and Pi16), intermuscular ones (which express CD34, VIM, Col1a2, Col15a1, and Pi16), and serosal ones (which express CD34, PI16, VIM, Col1a2, Col15a1, and Pi16). Collectively, our study revealed that the ultrastructure of PDFRA+ interstitial cells combined with their expression of multiple canonical and universal fibroblast-associated gene products indicates that they are fibroblasts. We further propose that there are four regionally distinct populations of fibroblasts in the bladder wall, which likely contribute to bladder function and dysfunction.NEW & NOTEWORTHY We currently lack basic insights into the fibroblasts that populate the bladder wall. By exploring the ultrastructure of mouse bladder connective tissue cells, combined with analyses of their gene and protein expression, our study revealed that PDGRA+ interstitial cells (also referred to as myofibroblasts, telocytes, and interstitial cells of Cajal-like cells) are fibroblasts and that the bladder wall contains multiple, regionally distinct populations of these cells.
Collapse
Affiliation(s)
- Dennis R Clayton
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Wily G Ruiz
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Marianela G Dalghi
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | | - Marcelo D Carattino
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Gerard Apodaca
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
6
|
Molecular Mechanisms and Key Processes in Interstitial, Hemorrhagic and Radiation Cystitis. BIOLOGY 2022; 11:biology11070972. [PMID: 36101353 PMCID: PMC9311586 DOI: 10.3390/biology11070972] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 06/17/2022] [Accepted: 06/23/2022] [Indexed: 11/22/2022]
Abstract
Simple Summary Pathologies of the bladder are called cystitis. They cause discomfort for the patient. Due to persistent pain, bleeding, urinary incontinence, and uncontrolled urination, the chronic forms cause considerable degradation to patient quality of life. Currently, there is no curative treatment for the most severe forms. This is both an economic and a societal problem. Although the different forms of cystitis have different causes, they share common mechanisms. We propose to describe in detail the key processes and the associated mechanisms involved in abacterial cystitis. Abstract Cystitis is a bladder disease with a high rate of prevalence in the world population. This report focuses on Interstitial Cystitis (IC), Hemorrhagic Cystitis (HC) and Chronic Radiation Cystitis. These pathologies have different etiologies, but they share common symptoms, for instance, pain, bleeding, and a contracted bladder. Overall, treatments are quite similar for abacterial cystitis, and include bladder epithelium protective or anti-inflammatory agents, alleviating pain and reducing bleeding. This review summarizes the mechanisms that the pathologies have in common, for instance, bladder dysfunction and inflammation. Conversely, some mechanisms have been described as present in only one pathology, such as neural regulation. Based on these specificities, we propose identifying a mechanism that could be common to all the above-mentioned pathologies.
Collapse
|
7
|
Zwaans BMM, Grobbel M, Carabulea AL, Lamb LE, Roccabianca S. Increased extracellular matrix stiffness accompanies compromised bladder function in a murine model of radiation cystitis. Acta Biomater 2022; 144:221-229. [PMID: 35301146 PMCID: PMC9100859 DOI: 10.1016/j.actbio.2022.03.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 02/14/2022] [Accepted: 03/08/2022] [Indexed: 11/30/2022]
Abstract
Radiation cystitis, a long-term bladder defect due to pelvic radiation therapy, results in lower urinary tract symptoms, such as urinary frequency and nocturia, suggestive of compromised bladder compliance. The goal of this study was to identify alterations to the mechanical behavior of the urinary bladder extracellular matrix of a murine model of radiation cystitis, at 3 and 6 months after radiation exposure. The results of this study demonstrated that the extracellular matrix of irradiated bladders was significantly less distensible when compared to age matching controls. These findings coincided with functional bladder changes, including increased number of voids and decreased voided volume. Both mechanical and functional changes were apparent at 3 months post-irradiation and were statistically significant at 6 months, demonstrating the progressive nature of radiation cystitis. Overall, the results of this study indicate that irradiation exposure changes both the mechanical and physiological properties of the bladder. STATEMENT OF SIGNIFICANCE: In humans, radiation cystitis results in lower urinary tract symptoms, such as urinary frequency and nocturia, suggestive of compromised bladder compliance. This pathology can significantly affect recovery and quality of life for cancer survivors. Gaining knowledge about how alterations to the mechanical behavior of the urinary bladder extracellular matrix can affect urinary function will have a significant impact on this population. The results of this study demonstrated that the extracellular matrix of irradiated bladders was significantly less distensible when compared to age matching controls, in a mouse model of radiation cystitis. These findings were accompanied by functional voiding changes, including increased number of voids and decreased voided volume. The results of this study uncovered that irradiation exposure changes the mechanical and physiological properties of the bladder.
Collapse
Affiliation(s)
- Bernadette M M Zwaans
- Department of Urology, Beaumont Health System, Royal Oak, MI, United States; Oakland University William Beaumont School of Medicine, Rochester, MI, United States
| | - Marissa Grobbel
- Department of Mechanical Engineering, Michigan State University, East Lansing, MI, United States
| | | | - Laura E Lamb
- Department of Urology, Beaumont Health System, Royal Oak, MI, United States; Oakland University William Beaumont School of Medicine, Rochester, MI, United States
| | - Sara Roccabianca
- Department of Mechanical Engineering, Michigan State University, East Lansing, MI, United States.
| |
Collapse
|
8
|
Zabbarova IV, Ikeda Y, Kozlowski MG, Tyagi P, Birder L, Chakrabarty B, Perera S, Dhir R, Straub AC, Sandner P, Andersson KE, Drake M, Fry CH, Kanai A. Benign prostatic hyperplasia/obstruction ameliorated using a soluble guanylate cyclase activator. J Pathol 2022; 256:442-454. [PMID: 34936088 PMCID: PMC8930559 DOI: 10.1002/path.5859] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 12/09/2021] [Accepted: 12/20/2021] [Indexed: 09/22/2023]
Abstract
Benign prostatic hyperplasia (BPH) is a feature of ageing males. Up to half demonstrate bladder outlet obstruction (BOO) with associated lower urinary tract symptoms (LUTS) including bladder overactivity. Current therapies to reduce obstruction, such as α1-adrenoceptor antagonists and 5α-reductase inhibitors, are not effective in all patients. The phosphodiesterase-5 inhibitor (PDE5I) tadalafil is also approved to treat BPH and LUTS, suggesting a role for nitric oxide (NO• ), soluble guanylate cyclase (sGC), and cGMP signalling pathways. However, PDE5I refractoriness can develop for reasons including nitrergic nerve damage and decreased NO• production, or inflammation-related oxidation of the sGC haem group, normally maintained in a reduced state by the cofactor cytochrome-b5-reductase 3 (CYB5R3). sGC activators, such as cinaciguat (BAY 58-2667), have been developed to enhance sGC activity in the absence of NO• or when sGC is oxidised. Accordingly, their effects on the prostate and LUT function of aged mice were evaluated. Aged mice (≥24 months) demonstrated a functional BPH/BOO phenotype, compared with adult animals (2-12 months), with low, delayed voiding responses and elevated intravesical pressures as measured by telemetric cystometry. This was consistent with outflow tract histological and molecular data that showed urethral constriction, increased prostate weight, greater collagen deposition, and cellular hyperplasia. All changes in aged animals were attenuated by daily oral treatment with cinaciguat for 2 weeks, without effect on serum testosterone levels. Cinaciguat had only transient (1 h) cardiovascular effects with oral gavage, suggesting a positive safety profile. The benefit of cinaciguat was suggested by its reversal of an overactive cystometric profile in CYB5R3 smooth muscle knockout mice that mirrors a profile of oxidative dysfunction where PDE5I may not be effective. Thus, the aged male mouse is a suitable model for BPH-induced BOO and cinaciguat has a demonstrated ability to reduce prostate-induced obstruction and consequent effects on bladder function. © 2021 The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Irina V. Zabbarova
- University of Pittsburgh, Department of Medicine, Renal-Electrolyte Division, Pittsburgh, PA, USA
| | - Youko Ikeda
- University of Pittsburgh, Department of Medicine, Renal-Electrolyte Division, Pittsburgh, PA, USA
| | - Mark G. Kozlowski
- University of Pittsburgh, Department of Medicine, Renal-Electrolyte Division, Pittsburgh, PA, USA
| | - Pradeep Tyagi
- University of Pittsburgh, Department of Urology, Pittsburgh, PA, USA
| | - Lori Birder
- University of Pittsburgh, Department of Medicine, Renal-Electrolyte Division, Pittsburgh, PA, USA
- University of Pittsburgh, Department of Pharmacology and Chemical Biology, Pittsburgh, PA, USA
| | - Basu Chakrabarty
- University of Bristol, School of Physiology, Pharmacology and Neuroscience, Bristol, UK
| | - Subashan Perera
- University of Pittsburgh, Department of Medicine, Geriatrics Division, Pittsburgh, PA, USA
| | - Rajiv Dhir
- University of Pittsburgh, Department of Pathology, Pittsburgh, PA, USA
| | - Adam C. Straub
- University of Pittsburgh, Department of Pharmacology and Chemical Biology, Pittsburgh, PA, USA
- Heart, Lung, Blood and Vascular Medicine Institute, Pittsburgh, PA, USA
| | | | - Karl-Erik Andersson
- Lund University, Division of Clinical Chemistry and Pharmacology, Lund, Sweden
| | - Marcus Drake
- University of Bristol, School of Physiology, Pharmacology and Neuroscience, Bristol, UK
| | - Christopher H. Fry
- University of Bristol, School of Physiology, Pharmacology and Neuroscience, Bristol, UK
| | - Anthony Kanai
- University of Pittsburgh, Department of Medicine, Renal-Electrolyte Division, Pittsburgh, PA, USA
- University of Pittsburgh, Department of Pharmacology and Chemical Biology, Pittsburgh, PA, USA
| |
Collapse
|
9
|
Ikeda Y, Zabbarova I, Tyagi P, Hitchens TK, Wolf-Johnston A, Wipf P, Kanai A. Targeting neurotrophin and nitric oxide signaling to treat spinal cord injury and associated neurogenic bladder overactivity. CONTINENCE (AMSTERDAM, NETHERLANDS) 2022; 1:100014. [PMID: 37207253 PMCID: PMC10194419 DOI: 10.1016/j.cont.2022.100014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Purpose or the research Nearly 300,000 people are affected by spinal cord injury (SCI) with approximately 18,000 new cases annually, according to the National SCI Statistics Center. SCI affects physical mobility and impairs the function of multiple internal organs to cause lower urinary tract (LUT) dysfunctions manifesting as detrusor sphincter dyssynergia (DSD) and neurogenic detrusor overactivity (NDO) with detrimental consequences to the quality of life and increased morbidity. Multiple lines of evidence now support time dependent evolution of the complex SCI pathology which requires a multipronged treatment approach of immediate, specialized care after spinal cord trauma bookended by physical rehabilitation to improve the clinical outcomes. Instead of one size fits all treatment approach, we propose adaptive drug treatment to counter the time dependent evolution of SCI pathology, with three small molecule drugs with distinctive sites of action for the recovery of multiple functions. Principal results Our findings demonstrate the improvement in the recovery of hindlimb mobility and bladder function of spinal cord contused mice following administration of small molecules targeting neurotrophin receptors, LM11A-31 and LM22B-10. While LM11A-31 reduced the cell death in the spinal cord, LM22B-10 promoted cell survival and axonal growth. Moreover, the soluble guanylate cyclase (sGC) activator, cinaciguat, enhanced the revascularization of the SCI injury site to promote vessel formation, dilation, and increased perfusion. Major conclusions Our adaptive three drug cocktail targets different stages of SCI and LUTD pathology: neuroprotective effect of LM11A-31 retards the cell death that occurs in the early stages of SCI; and LM22B-10 and cinaciguat promote neural remodeling and reperfusion at later stages to repair spinal cord scarring, DSD and NDO. LM11A-31 and cinaciguat have passed phase I and IIa clinical trials and possess significant potential for accelerated clinical testing in SCI/LUTD patients.
Collapse
Affiliation(s)
- Youko Ikeda
- University of Pittsburgh, School of Medicine, Department of Medicine, Renal-Electrolyte Division, USA
- University of Pittsburgh, School of Medicine, Department of Pharmacology & Chemical Biology, USA
| | - Irina Zabbarova
- University of Pittsburgh, School of Medicine, Department of Medicine, Renal-Electrolyte Division, USA
| | - Pradeep Tyagi
- University of Pittsburgh, School of Medicine, Department of Urology, USA
| | - T. Kevin Hitchens
- University of Pittsburgh, School of Medicine, Animal Imaging Center, USA
| | - Amanda Wolf-Johnston
- University of Pittsburgh, School of Medicine, Department of Medicine, Renal-Electrolyte Division, USA
| | - Peter Wipf
- University of Pittsburgh, Dietrich School of Arts and Sciences, Department of Chemistry, USA
| | - Anthony Kanai
- University of Pittsburgh, School of Medicine, Department of Medicine, Renal-Electrolyte Division, USA
- University of Pittsburgh, School of Medicine, Department of Pharmacology & Chemical Biology, USA
- Correspondence to: University of Pittsburgh, School of Medicine, Department of Medicine, A1224 Scaife Hall, 3550 Terrace Street, Pittsburgh, PA, 15261, USA. (A. Kanai)
| |
Collapse
|
10
|
Liu Q, Wang R, Ma N, Wang C, Chen W. Telmisartan inhibits bladder smooth muscle fibrosis in neurogenic bladder rats. Exp Ther Med 2022; 23:216. [PMID: 35126719 PMCID: PMC8796288 DOI: 10.3892/etm.2022.11140] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 12/07/2021] [Indexed: 12/15/2022] Open
Abstract
Hypertension is associated with bladder symptoms. The present study investigated whether an angiotensin receptor blocker could improve the symptoms and pathological changes associated with a neurogenic bladder (NB). A Sprague-Dawley rat model of NB was constructed. Rats in the sham and model groups were gavaged with saline, and rats in the treatment group were gavaged with telmisartan. Urodynamic parameters, including maximum cystometric capacity, residual urine volume, bladder wet weight, bladder compliance and detrusor pressure, were detected. Masson and H&E staining were performed to assess bladder fibrosis and histopathological changes. The expression levels of basic fibroblast growth factor (bFGF), TGF-β1, Collagen I, Collagen III, and α-smooth muscle actin (α-SMA) were also measured by reverse transcription-quantitative PCR, western blotting and immunohistochemistry. The model rats exhibited symptoms and pathological changes associated with NB. Treatment with telmisartan reduced maximum cystometric capacity, residual urine volume, bladder compliance and bladder wet weight, and increased detrusor pressure in model rats. The tissue staining results showed that telmisartan exerted an antifibrotic effect. In addition, telmisartan inhibited the expression of bFGF, TGF-β1, Collagen I, Collagen III and α-SMA in model rats. Therefore, the results of the present study indicated that telmisartan may serve as a potential therapeutic agent for NB.
Collapse
Affiliation(s)
- Qian Liu
- Department of Pediatric Surgery, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Ruoyi Wang
- Department of Pediatric Surgery, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Nan Ma
- Department of Pediatric Surgery, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Chuntian Wang
- Department of Pediatric Surgery, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Weixiu Chen
- Department of Pediatric Surgery, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| |
Collapse
|
11
|
Understanding Molecular Mechanisms and Identifying Key Processes in Chronic Radiation Cystitis. Int J Mol Sci 2022; 23:ijms23031836. [PMID: 35163758 PMCID: PMC8836784 DOI: 10.3390/ijms23031836] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 01/20/2022] [Accepted: 01/26/2022] [Indexed: 02/01/2023] Open
Abstract
Chronic radiation cystitis (CRC) is a consequence of pelvic radiotherapy and affects 5–10% of patients. The pathology of CRC is without curative treatment and is characterized by incontinence, pelvic pain and hematuria, which severely degrades patients’ quality of life. Current management strategies rely primarily on symptomatic measures and have certain limitations. Thanks to a better understanding of the pathophysiology of radiation cystitis, studies targeting key manifestations such as inflammation, neovascularization and cell atrophy have emerged and are promising avenues for future treatment. However, the mechanisms of CRC are still better described in animal models than in human models. Preclinical studies conducted to elucidate the pathophysiology of CRC use distinct models and are most often limited to specific processes, such as fibrosis, vascular damage and inflammation. This review presents a synthesis of experimental studies aimed at improving our understanding of the molecular mechanisms at play and identifying key processes in CRC.
Collapse
|
12
|
Samuel CS, Bennett RG. Relaxin as an anti-fibrotic treatment: Perspectives, challenges and future directions. Biochem Pharmacol 2021; 197:114884. [PMID: 34968489 DOI: 10.1016/j.bcp.2021.114884] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/13/2021] [Accepted: 12/15/2021] [Indexed: 02/07/2023]
Abstract
Fibrosis refers to the scarring and hardening of tissues, which results from a failed immune system-coordinated wound healing response to chronic organ injury and which manifests from the aberrant accumulation of various extracellular matrix components (ECM), primarily collagen. Despite being a hallmark of prolonged tissue damage and related dysfunction, and commonly associated with high morbidity and mortality, there are currently no effective cures for its regression. An emerging therapy that meets several criteria of an effective anti-fibrotic treatment, is the recombinant drug-based form of the human hormone, relaxin (also referred to as serelaxin, which is bioactive in several other species). This review outlines the broad anti-fibrotic and related organ-protective roles of relaxin, mainly from studies conducted in preclinical models of ageing and fibrotic disease, including its ability to ameliorate several aspects of fibrosis progression and maturation, from immune cell infiltration, pro-inflammatory and pro-fibrotic cytokine secretion, oxidative stress, organ hypertrophy, cell apoptosis, myofibroblast differentiation and ECM production, to its ability to facilitate established ECM degradation. Studies that have compared and/or combined these therapeutic effects of relaxin with current standard of care medication have also been discussed, along with the main challenges that have hindered the translation of the anti-fibrotic efficacy of relaxin to the clinic. The review then outlines the future directions as to where scientists and several pharmaceutical companies that have recognized the therapeutic potential of relaxin are working towards, to progress its development as a treatment for human patients suffering from various fibrotic diseases.
Collapse
Affiliation(s)
- Chrishan S Samuel
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria 3800, Australia; Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria 3052, Australia.
| | - Robert G Bennett
- Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA; Department of Internal Medicine, Division of Diabetes, Endocrinology & Metabolism, University of Nebraska Medical Center, Omaha, NE 68198-4130, USA.
| |
Collapse
|
13
|
Li Y, Chakraborty A, Broughton BRS, Ferens D, Widdop RE, Ricardo SD, Samuel CS. Comparing the renoprotective effects of BM-MSCs versus BM-MSC-exosomes, when combined with an anti-fibrotic drug, in hypertensive mice. Biomed Pharmacother 2021; 144:112256. [PMID: 34607108 DOI: 10.1016/j.biopha.2021.112256] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 09/15/2021] [Accepted: 09/26/2021] [Indexed: 12/29/2022] Open
Abstract
Fibrosis, a hallmark of chronic kidney disease (CKD), impairs the viability of human bone marrow derived-mesenchymal stromal cells (BM-MSCs) post-transplantation. To address this, we demonstrated that combining BM-MSCs with the anti-fibrotic drug, serelaxin (RLX), enhanced BM-MSC-induced renoprotection in preclinical CKD models. Given the increased interest and manufacturing advantages to using stem cell-derived exosomes (EXO) as therapeutics, this study determined whether RLX could enhance the therapeutic efficacy of BM-MSC-EXO, and compared the renoprotective effects of RLX and BM-MSC-EXO versus RLX and BM-MSCs in mice with hypertensive CKD. Adult male C57BL/6 mice were uninephrectomised, received deoxycorticosterone acetate and given saline to drink (1K/DOCA/salt) for 21 days. Control mice were uninephrectomised and given normal drinking water for the same time-period. Subgroups of 1K/DOCA/salt-hypertensive mice were then treated with either RLX (0.5 mg/kg/day) or BM-MSC-EXO (25 μg/mouse; equivalent to 1-2 × 106 BM-MSCs/mouse) alone; combinations of RLX and BM-MSC-EXO or BM-MSCs (1 × 106/mouse); or the mineralocorticoid receptor antagonist, spironolactone (20 mg/kg/day), from days 14-21. 1K/DOCA/salt-hypertensive mice developed kidney tubular damage, inflammation and fibrosis, and impaired kidney function 21 days post-injury. Whilst RLX alone attenuated the 1K/DOCA/salt-induced fibrosis, BM-MSC-EXO alone only diminished measures of tissue inflammation post-treatment. Comparatively, the combined effects of RLX and BM-MSC-EXO or BM-MSCs demonstrated similar anti-fibrotic efficacy, but RLX and BM-MSCs offered broader renoprotection over RLX and/or BM-MSC-EXO, and comparable effects to spironolactone. Only RLX and BM-MSCs, but not RLX and/or BM-MSC-EXO, also attenuated the 1K/DOCA/salt-induced hypertension. Hence, although RLX improved the renoprotective effects of BM-MSC-EXO, combining RLX with BM-MSCs provided a better therapeutic option for hypertensive CKD.
Collapse
Affiliation(s)
- Yifang Li
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria 3800, Australia
| | - Amlan Chakraborty
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria 3800, Australia
| | - Brad R S Broughton
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria 3800, Australia
| | - Dorota Ferens
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria 3800, Australia
| | - Robert E Widdop
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria 3800, Australia
| | - Sharon D Ricardo
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria 3800, Australia; Stem Cells and Development Program, Monash Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria 3800, Australia
| | - Chrishan S Samuel
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria 3800, Australia; Stem Cells and Development Program, Monash Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria 3800, Australia; Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria 3052, Australia.
| |
Collapse
|
14
|
Gao F, Dong W, Liu P, Narayanan J, Fish BL, Jacobs ER, Medhora M. Molecular Changes in miRNA in Irradiated Rat Kidneys: Role of miR-34a and its Vascular Targets in the Notch Pathway. Radiat Res 2021; 196:611-622. [PMID: 34330145 PMCID: PMC10416360 DOI: 10.1667/rade-20-00078.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 07/19/2021] [Indexed: 11/03/2022]
Abstract
The mechanism(s) of vascular regression in adult organs remains an unexplored gap. Irradiation to the kidney results in vascular regression and renal failure. The goal of this work was to determine molecular mechanism(s) of radiation-induced vascular regression and its mitigation by the drug lisinopril. Female WAG/RijCmcr rats received either 13 Gy X-ray irradiation, sparing one leg, or no irradiation, the latter serving as age-matched controls. Some irradiated animals received lisinopril. Kidney miRNA-seq was performed 35 days postirradiation, before symptoms of nephropathy. MicroRNA expression profiles were compared with data from humans. MicroRNA targets were predicted using TargetScan and confirmed by qRT-PCR and Western blot. Renal vascular endothelial cell density was evaluated at 100 days to confirm vascular regression. The normal rat kidney microRNA profile resembled that of humans. MiR-34a was increased >7-fold and emerged as the predominant rat microRNA altered by radiation. Expression of Jagged1, a ligand in the Notch pathway of vascular development and a target of miR-34a-5p was decreased by radiation but not in irradiated rats receiving lisinopril. Radiation decreased endothelial cells in the kidneys at 100 days, confirming vascular regression. In conclusion, the results of this study showed that radiation greatly increased miRNA34-a in rat kidneys, while lisinopril mitigated radiation-induced decrease of the Notch ligand, Jagged1, a molecular target of miRNA34-a.
Collapse
Affiliation(s)
- Feng Gao
- Department of Radiation Oncology Medical College of Wisconsin, Wauwatosa, Wisconsin
- Department of College of Dental Medicine - Illinois, Midwestern University, Downers Grove, Illinois
| | - Wei Dong
- Department of Radiation Oncology Medical College of Wisconsin, Wauwatosa, Wisconsin
| | - Pengyuan Liu
- Department of Physiology Medical College of Wisconsin, Wauwatosa, Wisconsin
| | - Jayashree Narayanan
- Department of Radiation Oncology Medical College of Wisconsin, Wauwatosa, Wisconsin
| | - Brian L. Fish
- Department of Radiation Oncology Medical College of Wisconsin, Wauwatosa, Wisconsin
| | - Elizabeth R. Jacobs
- Department of Physiology Medical College of Wisconsin, Wauwatosa, Wisconsin
- Department of Pulmonary Medicine Medical College of Wisconsin, Wauwatosa, Wisconsin
- Department of Cardiovascular Center, Medical College of Wisconsin, Wauwatosa, Wisconsin
- Research Service, Department of Veterans Affairs, Zablocki VAMC, Milwaukee, Wisconsin
| | - Meetha Medhora
- Department of Radiation Oncology Medical College of Wisconsin, Wauwatosa, Wisconsin
- Department of Physiology Medical College of Wisconsin, Wauwatosa, Wisconsin
- Department of Pulmonary Medicine Medical College of Wisconsin, Wauwatosa, Wisconsin
- Department of Cardiovascular Center, Medical College of Wisconsin, Wauwatosa, Wisconsin
- Research Service, Department of Veterans Affairs, Zablocki VAMC, Milwaukee, Wisconsin
| |
Collapse
|
15
|
Voiding defects in acute radiation cystitis driven by urothelial barrier defect through loss of E-cadherin, ZO-1 and Uroplakin III. Sci Rep 2021; 11:19277. [PMID: 34588475 PMCID: PMC8481534 DOI: 10.1038/s41598-021-98303-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 08/30/2021] [Indexed: 11/22/2022] Open
Abstract
Long term-side effects from cancer therapies are a growing health care concern as life expectancy among cancer survivors increases. Damage to the bladder is common in patients treated with radiation therapy for pelvic cancers and can result in radiation (hemorrhagic) cystitis (RC). The disease progression of RC consists of an acute and chronic phase, separated by a symptom-free period. Gaining insight in tissue changes associated with these phases is necessary to develop appropriate interventions. Using a mouse preclinical model, we have previously shown that fibrosis and vascular damage are the predominant pathological features of chronic RC. The goal of this study was to determine the pathological changes during acute RC. We identified that radiation treatment results in a temporary increase in micturition frequency and decrease in void volume 4–8 weeks after irradiation. Histologically, the micturition defect is associated with thinning of the urothelium, loss of urothelial cell–cell adhesion and tight junction proteins and decrease in uroplakin III expression. By 12 weeks, the urothelium had regenerated and micturition patterns were similar to littermate controls. No inflammation or fibrosis were detected in bladder tissues after irradiation. We conclude that functional bladder defects during acute RC are driven primarily by a urothelial defect.
Collapse
|
16
|
Johal N, Cao KX, Xie B, Millar M, Davda R, Ahmed A, Kanai AJ, Wood DN, Jabr RI, Fry CH. Contractile and Structural Properties of Detrusor from Children with Neurogenic Lower Urinary Tract Dysfunction. BIOLOGY 2021; 10:biology10090863. [PMID: 34571740 PMCID: PMC8471516 DOI: 10.3390/biology10090863] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/25/2021] [Accepted: 08/29/2021] [Indexed: 12/11/2022]
Abstract
Simple Summary Disorders of bladder function can result from congenital spinal cord developmental defects and can remain in a significant number of patients despite surgical improvements to repair the primary defect. We studied the ability of bladder wall muscle from such patients to contract, a function essential to void collected urine and avoid urinary tract infections and potential damage to the kidneys. Tissue was taken when patients were several years old, at the time of surgical operations to improve bladder function. This tissue would otherwise have been discarded and was collected with the full ethical approval and consent of parents or guardians. We found that the ability of the bladder wall samples to contract was impaired and was generally stiffer; both of which would make it more difficult for the bladder to void urine. These functional changes were associated with a replacement of muscle with connective tissue (fibrosis). The experiments provide a pathway to devise strategies that might improve bladder function in these patients through reversal of the intrinsic tissue pathways that increase fibrosis. Abstract Neurogenic lower urinary tract (NLUT) dysfunction in paediatric patients can arise after congenital or acquired conditions that affect bladder innervation. With some patients, urinary tract dysfunction remains and is more difficult to treat without understanding the pathophysiology. We measured in vitro detrusor smooth muscle function of samples from such bladders and any association with altered Wnt-signalling pathways that contribute to both foetal development and connective tissue deposition. A comparator group was tissue from children with normally functioning bladders. Nerve-mediated and agonist-induced contractile responses and passive stiffness were measured. Histology measured smooth muscle and connective tissue proportions, and multiplex immunohistochemistry recorded expression of protein targets associated with Wnt-signalling pathways. Detrusor from the NLUT group had reduced contractility and greater stiffness, associated with increased connective tissue content. Immunohistochemistry showed no major changes to Wnt-signalling components except down-regulation of c-Myc, a multifunctional regulator of gene transcription. NLUT is a diverse term for several diagnoses that disrupt bladder innervation. While we cannot speculate about the reasons for these pathophysiological changes, their recognition should guide research to understand their ultimate causes and develop strategies to attenuate and even reverse them. The role of changes to the Wnt-signalling pathways was minor.
Collapse
Affiliation(s)
- Navroop Johal
- Department of Urology, Great Ormond St Hospital for Children NHS Foundation Trust, London WC1N 3JH, UK; (N.J.); (K.X.C.)
| | - Kevin X. Cao
- Department of Urology, Great Ormond St Hospital for Children NHS Foundation Trust, London WC1N 3JH, UK; (N.J.); (K.X.C.)
| | - Boyu Xie
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol BS8 1TD, UK; (B.X.); (R.I.J.)
| | - Michael Millar
- Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK;
| | - Reena Davda
- Departments of Oncology and Urology, University College London Hospital, London W1G 8PH, UK; (R.D.); (D.N.W.)
| | - Aamir Ahmed
- Centre for Stem Cell Regeneration, King’s College London, London WC2R 2LS, UK;
| | - Anthony J. Kanai
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15260, USA;
| | - Dan N. Wood
- Departments of Oncology and Urology, University College London Hospital, London W1G 8PH, UK; (R.D.); (D.N.W.)
| | - Rita I. Jabr
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol BS8 1TD, UK; (B.X.); (R.I.J.)
| | - Christopher H. Fry
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol BS8 1TD, UK; (B.X.); (R.I.J.)
- Correspondence:
| |
Collapse
|
17
|
Abstract
Underactive bladder and impaired bladder compliance are irreversible problems associated with bladder fibrosis. Remodeling of the extracellular matrix is regarded as an important mechanism associated with bladder fibrosis. However, various risk factors and conditions contribute to the functional impairment of the bladder associated with fibrosis, and there is limited knowledge about bladder fibrosis-associated problems in the field of neurourology. Further studies are thus necessary to elucidate the underlying mechanism of bladder fibrosis and to identify effective treatment.
Collapse
|
18
|
Turner AC, Powers SA, Odom MR, Pak ES, Ashcraft KA, Koontz BF, Hannan JL. Impact of prostatic radiation therapy on bladder contractility and innervation. Neurourol Urodyn 2021; 40:1470-1478. [PMID: 34015163 DOI: 10.1002/nau.24705] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/03/2021] [Accepted: 05/06/2021] [Indexed: 11/10/2022]
Abstract
AIMS To determine the effect of prostatic radiation therapy (RT) on bladder contractility and morphology, and axon, or neuron profiles within the detrusor and major pelvic ganglia (MPG) in male rats. METHODS Male Sprague-Dawley rats (8 weeks) received a single dose of prostatic RT (0 or 22 Gy). Bladders and MPG were collected 2- and 10-weeks post-RT. Detrusor contractile responses to carbachol and electrical field stimulation (EFS) were measured. Bladders were stained with Masson's trichrome, and antibodies for nonspecific neuronal marker, cholinergic nerve marker choline acetyltransferase (ChAT), and alpha-smooth muscle actin. MPG gene expression was assessed by quantitative polymerase chain reaction for ubiquitin carboxy-terminal hydrolase L1 (Uchl1) and Chat. RESULTS At 2 weeks post-RT, bladder smooth muscle, detrusor cholinergic axon profiles, and MPG Chat gene expression were increased (p < .05), while carbachol and EFS-mediated contractions were decreased (p < .05). In contrast, at 10 weeks post-RT, nerve-mediated contractions were increased compared with control (p < .05), while bladder smooth muscle, detrusor cholinergic axon profiles, MPG Chat expression, and carbachol contractions had normalized. At both 2- and 10-weeks post-RT, there was no change in detrusor nonspecific axon profiles and MPG Uchl1 expression. CONCLUSION In a rat model, RT of the prostate and MPG was associated with early changes in MPG Chat gene expression, and bladder cholinergic axon profiles and smooth muscle content which resolved over time. After RT recovery, bladder contractility decreased early and increased by 10 weeks. Long-term changes to the MPG and increased bladder cholinergic axons may contribute to RT-induced bladder dysfunction in prostate cancer survivors.
Collapse
Affiliation(s)
- Alexander C Turner
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, USA
| | - Shelby A Powers
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, USA.,Department of Psychiatry & Behavioral Sciences, Duke University, Durham, North Carolina, USA
| | - Michael R Odom
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, USA.,Division of Urology, Department of Surgery, Duke University, Durham, North Carolina, USA
| | - Elena S Pak
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, USA
| | - Kathleen A Ashcraft
- Department of Radiation Oncology, Duke University, Durham, North Carolina, USA
| | - Bridget F Koontz
- Department of Radiation Oncology, Duke University, Durham, North Carolina, USA
| | - Johanna L Hannan
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, USA
| |
Collapse
|
19
|
Johal N, Cao K, Arthurs C, Millar M, Thrasivoulou C, Ahmed A, Jabr RI, Wood D, Cuckow P, Fry CH. Contractile function of detrusor smooth muscle from children with posterior urethral valves - The role of fibrosis. J Pediatr Urol 2021; 17:100.e1-100.e10. [PMID: 33214068 PMCID: PMC9099076 DOI: 10.1016/j.jpurol.2020.11.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 10/08/2020] [Accepted: 11/01/2020] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Posterior urethral valves (PUV) is the most common cause of congenital bladder outflow obstruction with persistent lower urinary tract and renal morbidities. There is a spectrum of functional bladder disorders ranging from hypertonia to bladder underactivity, but the aetiology of these clinical conditions remains unclear. AIMS AND OBJECTIVES We tested the hypothesis that replacement of detrusor muscle with non-muscle cells and excessive deposition of connective tissue is an important factor in bladder dysfunction with PUV. We used isolated detrusor samples from children with PUV and undergoing primary or secondary procedures in comparison to age-matched data from children with functionally normal bladders. In vitro contractile properties, as well as passive stiffness, were measured and matched to histological assessment of muscle and connective tissue. We examined if a major pathway for fibrosis was altered in PUV tissue samples. METHODS Isometric contractions were measured in vitro in response to either stimulation of motor nerves to detrusor or exposure to cholinergic and purinergic receptor agonists. Passive mechanical stiffness was measured by rapid stretching of the tissue and recording changes to muscle tension. Histology measured the relative amounts of detrusor muscle and connective tissue. Multiplex quantitative immunofluorescence labelling using five epitope markers was designed to determine cellular pathways, in particular the Wnt-signalling pathway, responsible for any changes to excessive deposition of connective tissue. RESULTS AND DISCUSSION PUV tissue showed equally reduced contractile function to efferent nerve stimulation or exposure to contractile agonists. Passive muscle stiffness was increased in PUV tissue samples. The smooth muscle:connective tissue ratio was also diminished and mirrored the reduction of contractile function and the increase of passive stiffness. Immunofluorescence labelling showed in PUV samples increased expression of the matrix metalloproteinase, MMP-7; as well as cyclin-D1 expression suggesting cellular remodelling. However, elements of a fibrosis pathway associated with Wnt-signalling were either reduced (β-catenin) or unchanged (c-Myc). The accumulation of extracellular matrix, containing collagen, will contribute to the reduced contractile performance of the bladder wall. It will also increase tissue stiffness that in vivo would lead to reduced filling compliance. CONCLUSIONS Replacement of smooth muscle with fibrosis is a major contributory factor in contractile dysfunction in the hypertonic PUV bladder. This suggests that a potential strategy to restore normal contractile and filling properties is development of the effective use of antifibrotic agents.
Collapse
Affiliation(s)
- Navroop Johal
- Department of Urology, Great Ormond St Hospital for Sick Children and Institute of Child Health, UCL, London, UK
| | - Kevin Cao
- Department of Urology, Great Ormond St Hospital for Sick Children and Institute of Child Health, UCL, London, UK
| | - Callum Arthurs
- Centre for Stem Cell and Regenerative Medicine, King's College London, UK
| | - Michael Millar
- Queen's Medical Research Institute, University of Edinburgh, UK
| | | | - Aamir Ahmed
- Centre for Stem Cell and Regenerative Medicine, King's College London, UK
| | - Rita I Jabr
- School of Biochemistry and Medical Sciences, University of Surrey, Guildford, UK
| | - Dan Wood
- Department of Urology, University College London Hospitals, London, UK
| | - Peter Cuckow
- Department of Urology, Great Ormond St Hospital for Sick Children and Institute of Child Health, UCL, London, UK
| | - Christopher H Fry
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, UK.
| |
Collapse
|
20
|
Bosch R, McCloskey K, Bahl A, Arlandis S, Ockrim J, Weiss J, Greenwell T. Can radiation-induced lower urinary tract disease be ameliorated in patients treated for pelvic organ cancer: ICI-RS 2019? Neurourol Urodyn 2020; 39 Suppl 3:S148-S155. [PMID: 32662556 PMCID: PMC7496485 DOI: 10.1002/nau.24380] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 04/18/2020] [Indexed: 12/15/2022]
Abstract
Aims This article reviews the clinical outcomes and basic science related to negative effects of radiotherapy (RT) on the lower urinary tract (LUT) when used to treat pelvic malignancies. Methods The topic was discussed at the 2019 meeting of the International Consultation on Incontinence―Research Society during a “think tank” session and is summarized in the present article. Results RT is associated with adverse effects on the LUT, which may occur during treatment or which can develop over decades posttreatment. Here, we summarize the incidence and extent of clinical symptoms associated with several modes of delivery of RT. RT impact on normal tissues including urethra, bladder, and ureters is discussed, and the underlying biology is examined. We discuss innovative in vivo methodologies to mimic RT in the laboratory and their potential use in the elucidation of mechanisms underlying radiation‐associated pathophysiology. Finally, emerging questions that need to be addressed through further research are proposed. Conclusions We conclude that RT‐induced negative effects on the LUT represent a significant clinical problem. Although this has been reduced with improved methods of delivery to spare normal tissue, we need to (a) discover better approaches to protect normal tissue and (b) develop effective treatments to reverse radiation damage.
Collapse
Affiliation(s)
- Ruud Bosch
- Department of Urologic Oncology, UMC Utrecht Cancer Center, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Karen McCloskey
- Centre for Cancer Research and Cell Biology, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, UK
| | - Amit Bahl
- Bristol Cancer Institute, University Hospitals Bristol NHS Foundation Trust, Bristol, UK
| | - Salvador Arlandis
- Functional and Reconstructive Urology Section, Hospital Universitari Politècnic La Fe, Valencia, Spain
| | - Jeremy Ockrim
- Female, Functional and Restorative Urology Unit, University College London Hospitals, London, UK
| | - Jeffrey Weiss
- Department of Urology, SUNY Downstate Health Sciences University, Brooklyn, New York
| | - Tamsin Greenwell
- Female, Functional and Restorative Urology Unit, University College London Hospitals, London, UK
| |
Collapse
|
21
|
Zwaans BMM, Nicolai HE, Chancellor MB, Lamb LE. Prostate cancer survivors with symptoms of radiation cystitis have elevated fibrotic and vascular proteins in urine. PLoS One 2020; 15:e0241388. [PMID: 33119677 PMCID: PMC7595289 DOI: 10.1371/journal.pone.0241388] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 10/13/2020] [Indexed: 01/22/2023] Open
Abstract
Radiation for pelvic cancers can result in severe bladder damage and radiation cystitis (RC), which is characterized by chronic inflammation, fibrosis, and vascular damage. RC development is poorly understood because bladder biopsies are difficult to obtain. The goal of this study is to gain understanding of molecular changes that drive radiation-induced cystitis in cancer survivors using urine samples from prostate cancer survivors with history of radiation therapy. 94 urine samples were collected from prostate cancer survivors with (n = 85) and without (n = 9) history of radiation therapy. 15 patients with radiation history were officially diagnosed with radiation cystitis. Levels of 47 different proteins were measured using Multiplex Luminex. Comparisons were made between non-irradiated and irradiated samples, and within irradiated samples based on radiation cystitis diagnosis, symptom scores or hematuria. Statistical analysis was performed using Welch’s t-test. In prostate cancer survivors with history of radiation therapy, elevated levels of PAI 1, TIMP1, TIMP2, HGF and VEGF-A were detected in patients that received a radiation cystitis diagnosis. These proteins were also increased in patients suffering from hematuria or high symptom scores. No inflammatory proteins were detected in the urine, except in patients with gross hematuria and end stage radiation cystitis. Active fibrosis and vascular distress is detectable in the urine through elevated levels of associated proteins. Inflammation is only detected in urine of patients with end-stage radiation cystitis disease. These results suggest that fibrosis and vascular damage drive the development of radiation cystitis and could lead to the development of more targeted treatments.
Collapse
Affiliation(s)
- Bernadette M. M. Zwaans
- Department of Urology, William Beaumont Hospital, Royal Oak, Michigan, United States of America
- Oakland University William Beaumont School of Medicine, Rochester, Michigan, United States of America
| | - Heinz E. Nicolai
- Departamento de Urología, Universidad de Chile, Santiago, Chile
- Hospital Clínico San Borja Arriarán, Santiago, Chile
| | - Michael B. Chancellor
- Department of Urology, William Beaumont Hospital, Royal Oak, Michigan, United States of America
- Oakland University William Beaumont School of Medicine, Rochester, Michigan, United States of America
| | - Laura E. Lamb
- Department of Urology, William Beaumont Hospital, Royal Oak, Michigan, United States of America
- Oakland University William Beaumont School of Medicine, Rochester, Michigan, United States of America
- * E-mail:
| |
Collapse
|
22
|
Zuppone S, Bresolin A, Spinelli AE, Fallara G, Lucianò R, Scarfò F, Benigni F, Di Muzio N, Fiorino C, Briganti A, Salonia A, Montorsi F, Vago R, Cozzarini C. Pre-clinical Research on Bladder Toxicity After Radiotherapy for Pelvic Cancers: State-of-the Art and Challenges. Front Oncol 2020; 10:527121. [PMID: 33194587 PMCID: PMC7642999 DOI: 10.3389/fonc.2020.527121] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 08/31/2020] [Indexed: 01/01/2023] Open
Abstract
Despite the dramatic advancements in pelvic radiotherapy, urinary toxicity remains a significant side-effect. The assessment of clinico-dosimetric predictors of radiation cystitis (RC) based on clinical data has improved substantially over the last decade; however, a thorough understanding of the physiopathogenetic mechanisms underlying the onset of RC, with its variegated acute and late urinary symptoms, is still largely lacking, and data from pre-clinical research is still limited. The aim of this review is to provide an overview of the main open issues and, ideally, to help investigators in orienting future research. First, anatomy and physiology of bladder, as well as the current knowledge of dose and dose-volume effects in humans, are briefly summarized. Subsequently, pre-clinical radiobiology aspects of RC are discussed. The findings suggest that pre-clinical research on RC in animal models is a lively field of research with growing interest in the development of new radioprotective agents. The availability of new high precision micro-irradiators and the rapid advances in small animal imaging might lead to big improvement into this field. In particular, studies focusing on the definition of dose and fractionation are warranted, especially considering the growing interest in hypo-fractionation and ablative therapies for prostate cancer treatment. Moreover, improvement in radiotherapy plans optimization by selectively reducing radiation dose to more radiosensitive substructures close to the bladder would be of paramount importance. Finally, thanks to new pre-clinical imaging platforms, reliable and reproducible methods to assess the severity of RC in animal models are expected to be developed.
Collapse
Affiliation(s)
- Stefania Zuppone
- Division of Experimental Oncology, Urological Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Fondazione Centro San Raffaele, Milan, Italy
| | - Andrea Bresolin
- Fondazione Centro San Raffaele, Milan, Italy.,Department of Medical Physics, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Antonello E Spinelli
- Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giuseppe Fallara
- Division of Experimental Oncology, Urological Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Roberta Lucianò
- Unit of Pathology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Federico Scarfò
- Unit of Pathology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Fabio Benigni
- Division of Experimental Oncology, Urological Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Nadia Di Muzio
- Department of Radiotherapy, IRCCS San Raffaele Scientific Institute, Milan, Italy.,University Vita-Salute San Raffaele, Milan, Italy
| | - Claudio Fiorino
- Department of Medical Physics, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Alberto Briganti
- Division of Experimental Oncology, Urological Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy.,University Vita-Salute San Raffaele, Milan, Italy
| | - Andrea Salonia
- Division of Experimental Oncology, Urological Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy.,University Vita-Salute San Raffaele, Milan, Italy
| | - Francesco Montorsi
- Division of Experimental Oncology, Urological Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy.,University Vita-Salute San Raffaele, Milan, Italy
| | - Riccardo Vago
- Division of Experimental Oncology, Urological Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy.,University Vita-Salute San Raffaele, Milan, Italy
| | - Cesare Cozzarini
- Department of Radiotherapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
23
|
Effects of an alpha-1d adrenoreceptor antagonist (naftopidil) on bladder dysfunction after radiotherapy in female rats. Int Urogynecol J 2020; 32:2747-2755. [PMID: 32761376 DOI: 10.1007/s00192-020-04472-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 07/23/2020] [Indexed: 10/23/2022]
Abstract
PURPOSE Storage-phase bladder dysfunction can develop after pelvic radiotherapy. As the alpha-1d adrenoreceptor (a1d-AR) is dominant in the human detrusor, we aimed to investigate the effect of an a1d-AR antagonist on bladder dysfunction after pelvic radiotherapy in a rat model. MATERIALS AND METHODS Twenty-four female Wistar rats were used. Eight rats (14-15 weeks, 250-300 g) were randomized to three groups (normal reference group, radiation alone group and radiation plus naftopidil group). An 18-Gy dose of radiotherapy was applied to the radiation alone and radiation plus naftopidil groups. Naftopidil (20 mg/kg) was administered daily to the radiation plus naftopidil group. Four weeks after radiation, all rats underwent cystometry and were killed for reverse transcription polymerase chain reaction to detect mRNAs [a1d-AR, brain-derived neurotrophic factor (BDNF) and vascular endothelial growth factor (VEGF)], Western blot to detect proteins (a1d-AR, extracellular-signal-regulated kinase, BDNF and VEGF) and immunohistochemistry. RESULTS Compared to the radiation alone group, (1) the decrease in the mRNA and protein expression of a1d-AR and VEGF was ameliorated, (2) the increase in the expression of BDNF mRNA and proteins such as extracellular-signal-regulated kinase and BDNF was suppressed, (3) submucosal thickness and vascularity on immunohistochemistry were improved, and (4) the baseline intravesical pressure and intercontraction interval in cystometry were ameliorated in the radiation plus naftopidil group. CONCLUSION Administration of an a1d-AR antagonist could improve storage-phase bladder dysfunction after radiotherapy not only by upregulating a1d-AR, which might decrease bladder compliance, but also by enhancing vascularity, which might protect the urinary bladder from chronic ischemic inflammation.
Collapse
|
24
|
Fry CH, Chakrabarty B, Hashitani H, Andersson KE, McCloskey K, Jabr RI, Drake MJ. New targets for overactive bladder-ICI-RS 2109. Neurourol Urodyn 2020; 39 Suppl 3:S113-S121. [PMID: 31737931 PMCID: PMC8114459 DOI: 10.1002/nau.24228] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Accepted: 10/31/2019] [Indexed: 12/16/2022]
Abstract
AIM To review evidence for novel drug targets that can manage overactive bladder (OAB) symptoms. METHODS A think tank considered evidence from the literature and their own research experience to propose new drug targets in the urinary bladder to characterize their use to treat OAB. RESULTS Five classes of agents or cellular pathways were considered. (a) Cyclic nucleotide-dependent (cyclic adenosine monophosphate and cyclic guanosine monophosphate) pathways that modulate adenosine triphosphate release from motor nerves and urothelium. (b) Novel targets for β3 agonists, including the bladder wall vasculature and muscularis mucosa. (c) Several TRP channels (TRPV1 , TRPV4 , TRPA1 , and TRPM4 ) and their modulators in affecting detrusor overactivity. (d) Small conductance Ca2+ -activated K+ channels and their influence on spontaneous contractions. (e) Antifibrosis agents that act to modulate directly or indirectly the TGF-β pathway-the canonical fibrosis pathway. CONCLUSIONS The specificity of action remains a consideration if particular classes of agents can be considered for future development as receptors or pathways that mediate actions of the above mentioned potential agents are distributed among most organ systems. The tasks are to determine more detail of the pathological changes that occur in the OAB and how the specificity of potential drugs may be directed to bladder pathological changes. An important conclusion was that the storage, not the voiding, phase in the micturition cycle should be investigated and potential targets lie in the whole range of tissue in the bladder wall and not just detrusor.
Collapse
Affiliation(s)
- Christopher Henry Fry
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - Basu Chakrabarty
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - Hikaru Hashitani
- Department of Cell Physiology, Nagoya City University, Nagoya, Japan
| | - Karl-Erik Andersson
- Institute of Laboratory Medicine, Lund University, Lund, Sweden
- Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - Karen McCloskey
- School of Medicine, Dentistry and Biomedical Sciences, Queens University Belfast, Belfast, UK
| | - Rita I. Jabr
- Division of Biochemical Sciences, Faculty of Health and Biomedical Sciences, University of Surrey, Guildford, UK
| | | |
Collapse
|
25
|
Nishikawa N, Chakrabarty B, Kitney D, Jabr R, Kanai A, Fry C. Stretch- and carbachol-induced ATP release from bladder wall preparations of young and aged mice. Neurourol Urodyn 2020; 39:1644-1652. [PMID: 32531080 DOI: 10.1002/nau.24426] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 05/28/2020] [Accepted: 05/31/2020] [Indexed: 12/19/2022]
Abstract
AIMS Bladder wall stretch increases tissue tension and releases adenosine 5'-triphosphate (ATP) as part of a transduction process to sense bladder filling. Aging is associated with bladder fibrosis to produce a stiffer bladder wall: this may augment ATP release and contribute to age-dependent urgency. Muscarinic agonists also release ATP and present a potential target for antimuscarinic agents, but its age-dependency is unknown. This study aimed, in young and old mice, to: (a) quantify the relationship between bladder wall stiffness and stretch-dependent ATP release and; (b) characterize muscarinic agonist-dependent release. METHODS ATP release from young (9-12 weeks) and aged (24 months) mouse bladder wall was measured in vitro, with a luciferin-luciferase assay, after stretch or carbachol exposure. Bladder wall stiffness, measured simultaneously during stretch, was compared to histological proportions of connective tissue and detrusor muscle. RESULTS With young mice, stretch-activated ATP release required an intact mucosa and was positively associated with wall stiffness. ATP release by carbachol was about four-fold greater compared to stretch. With aged mice: ATP release varied a hundred-fold and no association with stiffness; carbachol release diminished; connective tissue and mucosa thickness increased. CONCLUSIONS With young mice, stretch, or muscarinic agonists potently induce bladder wall ATP release. Stretch-dependent release is proportional to bladder wall stiffness, independent of the extent of stretch. With aged mice dependence of stretch-activated ATP release with stiffness was lost. The huge variability of release suggests that aged mice do not form a homogenous cohort and may underlie the heterogeneity in bladder filling sensations.
Collapse
Affiliation(s)
- Nobuyuki Nishikawa
- The School of Physiology, Pharmacology & Neuroscience, University of Bristol, Bristol, UK.,School of Biosciences and Medicine, University of Surrey, Guildford, UK
| | - Basu Chakrabarty
- The School of Physiology, Pharmacology & Neuroscience, University of Bristol, Bristol, UK
| | - Darryl Kitney
- The School of Physiology, Pharmacology & Neuroscience, University of Bristol, Bristol, UK.,School of Biosciences and Medicine, University of Surrey, Guildford, UK
| | - Rita Jabr
- School of Biosciences and Medicine, University of Surrey, Guildford, UK
| | - Anthony Kanai
- School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Christopher Fry
- The School of Physiology, Pharmacology & Neuroscience, University of Bristol, Bristol, UK
| |
Collapse
|
26
|
Diaz EC, Briggs M, Wen Y, Zhuang G, Wallace SL, Dobberfuhl AD, Kao CS, Chen BC. Characterizing relaxin receptor expression and exploring relaxin's effect on tissue remodeling/fibrosis in the human bladder. BMC Urol 2020; 20:44. [PMID: 32321501 PMCID: PMC7178754 DOI: 10.1186/s12894-020-00607-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 03/30/2020] [Indexed: 02/06/2023] Open
Abstract
Background Relaxin is an endogenous protein that has been shown to have antifibrotic properties in various organ systems. There has been no characterization of relaxin’s role in the human bladder. Our objective was to characterize relaxin receptor expression in the human bladder and assess relaxin’s effect on tissue remodeling/fibrosis pathways in bladder smooth muscle cells. Methods Relaxin family peptide receptor 1 (RXFP1) and RXFP2 expression was assessed using quantitative reverse transcriptase-PCR (qRT-PCR) and immunohistochemistry (IHC) on primary bladder tissue. Primary human smooth muscle bladder cells were cultured and stimulated with various concentrations of relaxin. Western blot, qRTPCR, ELISA, and zymogram assays were used to analyze fibrosis/tissue remodeling pathway proteins. Results There was universal mRNA transcript detection and protein expression of relaxin receptors in primary bladder specimens. Immunohistochemistry demonstrated RXFP1 and RXFP2 localizing to both urothelial and smooth muscle cell layers of the bladder. 24 h of in vitro relaxin stimulation did not affect mRNA expression of selected proteins in human bladder smooth muscle cells. However, 48 h of in vitro relaxin stimulation resulted in upregulation of active (p = 0.004) and latent (p = 0.027) MMP-2 in cell lysate, and upregulation of active MMP-2 in supernatant (p = 0.04). There was a dose dependent relationship with increasing expression of MMP-2 with increasing relaxin concentration. Relaxin stimulation resulted in decreased levels of active and total TGF-β1 in supernatant and extracellular matrix (p < 0.005 with 100 ng/mL relaxin stimulation). Conclusions In the human bladder, relaxin receptors are expressed at the dome and trigone and localize to the urothelium and smooth muscle cell layers. Stimulation of human bladder SMCs with relaxin in vitro affects expression of MMP-2 and TGF-β1.
Collapse
Affiliation(s)
- Edward C Diaz
- Department of Urology, Stanford University Medical Center, 300 Pasteur Drive, Grant S-287, Stanford, CA, 94305, USA. .,Present Address: Division of Pediatric Urology, Advocate Children's Hospital, 8901 West Golf Road, Suite 301, Des Plaines, IL, 60016, USA.
| | - Mason Briggs
- Department of Urology, Stanford University Medical Center, 300 Pasteur Drive, Grant S-287, Stanford, CA, 94305, USA.,Department of Obstetrics and Gynecology, Stanford University Medical Center, 300 Pasteur Drive, Rm A370, MC 5317, Stanford, CA, 94305, USA
| | - Yan Wen
- Department of Obstetrics and Gynecology, Stanford University Medical Center, 300 Pasteur Drive, Rm A370, MC 5317, Stanford, CA, 94305, USA
| | - Guobing Zhuang
- Department of Obstetrics and Gynecology, Stanford University Medical Center, 300 Pasteur Drive, Rm A370, MC 5317, Stanford, CA, 94305, USA
| | - Shannon L Wallace
- Department of Obstetrics and Gynecology, Stanford University Medical Center, 300 Pasteur Drive, Rm A370, MC 5317, Stanford, CA, 94305, USA
| | - Amy D Dobberfuhl
- Department of Urology, Stanford University Medical Center, 300 Pasteur Drive, Grant S-287, Stanford, CA, 94305, USA
| | - Chia-Sui Kao
- Department of Pathology, Stanford University Medical Center, 300 Pasteur Drive, Rm L235, Stanford, CA, 94305, USA
| | - Bertha C Chen
- Department of Obstetrics and Gynecology, Stanford University Medical Center, 300 Pasteur Drive, Rm A370, MC 5317, Stanford, CA, 94305, USA
| |
Collapse
|
27
|
Zwaans BMM, Wegner KA, Bartolone SN, Vezina CM, Chancellor MB, Lamb LE. Radiation cystitis modeling: A comparative study of bladder fibrosis radio-sensitivity in C57BL/6, C3H, and BALB/c mice. Physiol Rep 2020; 8:e14377. [PMID: 32109348 PMCID: PMC7048381 DOI: 10.14814/phy2.14377] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 01/29/2020] [Accepted: 01/30/2020] [Indexed: 12/12/2022] Open
Abstract
A subset of patients receiving radiation therapy for pelvic cancer develop radiation cystitis, a complication characterized by mucosal cell death, inflammation, hematuria, and bladder fibrosis. Radiation cystitis can reduce bladder capacity, cause incontinence, and impair voiding function so severely that patients require surgical intervention. Factors influencing onset and severity of radiation cystitis are not fully known. We tested the hypothesis that genetic background is a contributing factor. We irradiated bladders of female C57BL/6, C3H, and BALB/c mice and evaluated urinary voiding function, bladder shape, histology, collagen composition, and distribution of collagen-producing cells. We found that the genetic background profoundly affects the severity of radiation-induced bladder fibrosis and urinary voiding dysfunction. C57BL/6 mice are most susceptible and C3H mice are most resistant. Irradiated C57BL/6 mouse bladders are misshapen and express more abundant collagen I and III proteins than irradiated C3H and BALB/c bladders. We localized Col1a1 and Col3a1 mRNAs to FSP1-negative stromal cells in the bladder lamina propria and detrusor. The number of collagen I and collagen III-producing cells can predict the average voided volume of a mouse. Collectively, we show that genetic factors confer sensitivity to radiation cystitis, establish C57BL/6 mice as a sensitive preclinical model, and identify a potential role for FSP1-negative stromal cells in radiation-induced bladder fibrosis.
Collapse
Affiliation(s)
- Bernadette M. M. Zwaans
- Department of UrologyWilliam Beaumont HospitalRoyal OakMIUSA
- Oakland University William Beaumont School of MedicineRoyal OakMIUSA
| | - Kyle A. Wegner
- Molecular and Environmental Toxicology CenterSchool of Medicine and Public HealthUniversity of Wisconsin‐MadisonMadisonWIUSA
- Department of Comparative BiosciencesSchool of Veterinary MedicineUniversity of Wisconsin‐MadisonMadisonWIUSA
| | | | - Chad M. Vezina
- Molecular and Environmental Toxicology CenterSchool of Medicine and Public HealthUniversity of Wisconsin‐MadisonMadisonWIUSA
- Department of Comparative BiosciencesSchool of Veterinary MedicineUniversity of Wisconsin‐MadisonMadisonWIUSA
| | - Michael B. Chancellor
- Department of UrologyWilliam Beaumont HospitalRoyal OakMIUSA
- Oakland University William Beaumont School of MedicineRoyal OakMIUSA
| | - Laura E. Lamb
- Department of UrologyWilliam Beaumont HospitalRoyal OakMIUSA
- Oakland University William Beaumont School of MedicineRoyal OakMIUSA
| |
Collapse
|
28
|
Fry CH, McCloskey KD. Spontaneous Activity and the Urinary Bladder. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1124:121-147. [PMID: 31183825 DOI: 10.1007/978-981-13-5895-1_5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The urinary bladder has two functions: to store urine, when it is relaxed and highly compliant; and void its contents, when intravesical pressure rises due to co-ordinated contraction of detrusor smooth muscle in the bladder wall. Superimposed on this description are two observations: (1) the normal, relaxed bladder develops small transient increases of intravesical pressure, mirrored by local bladder wall movements; (2) pathological, larger pressure variations (detrusor overactivity) can occur that may cause involuntary urine loss and/or detrusor overactivity. Characterisation of these spontaneous contractions is important to understand: how normal bladder compliance is maintained during filling; and the pathophysiology of detrusor overactivity. Consideration of how spontaneous contractions originate should include the structural complexity of the bladder wall. Detrusor smooth muscle layer is overlain by a mucosa, itself a complex structure of urothelium and a lamina propria containing sensory nerves, micro-vasculature, interstitial cells and diffuse muscular elements.Several theories, not mutually exclusive, have been advanced for the origin of spontaneous contractions. These include intrinsic rhythmicity of detrusor muscle; modulation by non-muscular pacemaking cells in the bladder wall; motor input to detrusor by autonomic nerves; regulation of detrusor muscle excitability and contractility by the adjacent mucosa and spontaneous contraction of elements of the lamina propria. This chapter will consider evidence for each theory in both normal and overactive bladder and how their significance may vary during ageing and development. Further understanding of these mechanisms may also identify novel drug targets to ameliorate the clinical consequences of large contractions associated with detrusor overactivity.
Collapse
Affiliation(s)
- Christopher H Fry
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK.
| | - Karen D McCloskey
- School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, UK
| |
Collapse
|
29
|
Kanai AJ, Konieczko EM, Bennett RG, Samuel CS, Royce SG. Relaxin and fibrosis: Emerging targets, challenges, and future directions. Mol Cell Endocrinol 2019; 487:66-74. [PMID: 30772373 PMCID: PMC6475456 DOI: 10.1016/j.mce.2019.02.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 02/04/2019] [Accepted: 02/04/2019] [Indexed: 01/15/2023]
Abstract
The peptide hormone relaxin is well-known for its anti-fibrotic actions in several organs, particularly from numerous studies conducted in animals. Acting through its cognate G protein-coupled receptor, relaxin family peptide receptor 1 (RXFP1), serelaxin (recombinant human relaxin) has been shown to consistently inhibit the excessive extracellular matrix production (fibrosis) that results from the aberrant wound-healing response to tissue injury and/or chronic inflammation, and at multiple levels. Furthermore, it can reduce established scarring by promoting the degradation of aberrant extracellular matrix components. Following on from the review that describes the mechanisms and signaling pathways associated with the extracellular matrix remodeling effects of serelaxin (Ng et al., 2019), this review focuses on newly identified tissue targets of serelaxin therapy in fibrosis, and the limitations associated with (se)relaxin research.
Collapse
Affiliation(s)
- Anthony J Kanai
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Elisa M Konieczko
- Biology Department, Morosky College of Health Professions and Sciences, Gannon University, Erie, PA, USA.
| | - Robert G Bennett
- Research Service, VA Nebraska-Western Iowa Health Care System, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA; Research Service, VA Nebraska-Western Iowa Health Care System, Department of Biochemistry & Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA.
| | - Chrishan S Samuel
- Cardiovascular Disease Theme, Monash Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, VIC, Australia.
| | - Simon G Royce
- Cardiovascular Disease Theme, Monash Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, VIC, Australia; Central Clinical School, Monash University, Prahran, VIC, Australia.
| |
Collapse
|
30
|
Johal NS, Arthurs C, Cuckow P, Cao K, Wood DN, Ahmed A, Fry CH. Functional, histological and molecular characteristics of human exstrophy detrusor. J Pediatr Urol 2019; 15:154.e1-154.e9. [PMID: 30745011 DOI: 10.1016/j.jpurol.2018.12.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 11/07/2018] [Accepted: 12/20/2018] [Indexed: 11/18/2022]
Abstract
INTRODUCTION Bladder exstrophy is a congenital anomaly involving foetal exposure and protrusion of the open bladder through an incomplete lower abdominal wall. Techniques to surgically correct exstrophy after birth have greatly improved, but it still presents a major challenge to achieve continence and a good quality of life for patients and their families as the pathophysiology of bladder dysfunction is unknown. OBJECTIVES A multimodal approach was used to characterise the histological and biomechanical properties of exstrophy detrusor. These were correlated with myocyte responses to agonists and an evaluation of developmental signalling pathways to evaluate the cause of bladder dysfunction in exstrophy. STUDY DESIGN Detrusor muscle specimens were obtained during corrective surgery from four exstrophy groups: neonatal (1-3 days, n = 8), younger children (7 months-5 years, n = 13) and older children (8-14 years, n = 11) undergoing secondary procedures and cloacal exstrophy (16 days-9 years, n = 9); control specimens were obtained from children (3 months-9 years, n = 14) undergoing surgery for other pathologies but with normal bladder function. Five lines of experiments were undertaken: measurement of connective tissue to detrusor muscle ratio, contractile responses to electrical and agonist stimulation; in vitro biomechanical stiffness, intracellular Ca2+ responses to contractile agonists and immunohistochemistry for proteins (MMP-7, cyclinD1, β-catenin and c-myc) involved in fibrosis generation. Exstrophy data were compared with those from the control group. RESULTS Exstrophy tissue demonstrated reduced smooth muscle compared with connective tissue, reduced contractile responses and greater mechanical stiffness. However, intracellular Ca2+ responses to agonists were maintained. These changes were greatest in neonatal and cloacal exstrophy samples and least in those from older paediatric bladders. Immunolabelled MMP-7, β-catenin and c-myc were reduced in exstrophy samples. DISCUSSION These results highlight the reality that newborns with exstrophy have significantly reduced compliance and bladder underactivity, which may persist or return to normal values with surgery and age. The primary cause of underactivity is increased connective tissue in relation to detrusor muscle; however, detrusor myocyte function remains normal. Finally, the increase of the smooth muscle content in the paediatric bladder group indicates a remodelling response of the bladder to surgical correction and time. Excess gestational fibrosis is associated with changed expression of key proteins in the Wnt-signalling pathway, a potential aetiological factor and therapeutic target. CONCLUSION Results point to connective tissue deposition as the primary pathological process that determines bladder function with normal myocyte function. Future research that reduces connective tissue deposition may lead to improvement in outcomes for these children.
Collapse
Affiliation(s)
- N S Johal
- Department of Urology, Great Ormond St Hospital for Sick Children, London, UK
| | - C Arthurs
- School of Physiology, Pharmacology & Neuroscience, University of Bristol, UK; Centre for Stem Cells and Regenerative Medicine, King's College London, UK
| | - P Cuckow
- Department of Urology, Great Ormond St Hospital for Sick Children, London, UK
| | - K Cao
- School of Physiology, Pharmacology & Neuroscience, University of Bristol, UK; Centre for Stem Cells and Regenerative Medicine, King's College London, UK
| | - D N Wood
- Department of Urology, University College Hospitals, London, UK
| | - A Ahmed
- Centre for Stem Cells and Regenerative Medicine, King's College London, UK
| | - C H Fry
- School of Physiology, Pharmacology & Neuroscience, University of Bristol, UK.
| |
Collapse
|