1
|
Drumm BT, Gupta N, Mircea A, Griffin CS. Cells and ionic conductances contributing to spontaneous activity in bladder and urethral smooth muscle. J Physiol 2024. [PMID: 39323077 DOI: 10.1113/jp284744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 09/02/2024] [Indexed: 09/27/2024] Open
Abstract
Smooth muscle organs of the lower urinary tract comprise the bladder detrusor and urethral wall, which have a reciprocal contractile relationship during urine storage and micturition. As the bladder fills with urine, detrusor smooth muscle cells (DSMCs) remain relaxed to accommodate increases in intravesical pressure while urethral smooth muscle cells (USMCs) sustain tone to occlude the urethral orifice, preventing leakage. While neither organ displays coordinated regular contractions as occurs in small intestine, lymphatics or renal pelvis, they do exhibit patterns of rhythmicity at cellular and tissue levels. In rabbit and guinea-pig urethra, electrical slow waves are recorded from USMCs. This activity is linked to cells expressing vimentin, c-kit and Ca2+-activated Cl- channels, like interstitial cells of Cajal in the gastrointestinal tract. In mouse, USMCs are rhythmically active (firing propagating Ca2+ waves linked to contraction), and this cellular rhythmicity is asynchronous across tissues and summates to form tone. Experiments in mice have failed to demonstrate a voltage-dependent mechanism for regulating this rhythmicity or contractions in vitro, suggesting that urethral tone results from an intrinsic ability of USMCs to 'pace' their own Ca2+ mobilization pathways required for contraction. DSMCs exhibit spontaneous transient contractions, increases in intracellular Ca2+ and action potentials. Consistent across numerous species, including humans, this activity relies on voltage-dependent Ca2+ influx in DSMCs. While interstitial cells are present in the bladder, they do not 'pace' the organ in an excitatory manner. Instead, specialized cells (PDGFRα+ interstitial cells) may 'negatively pace' DSMCs to prevent bladder overexcitability.
Collapse
Affiliation(s)
- Bernard T Drumm
- Smooth Muscle Research Centre, Department of Life & Health Science, Dundalk Institute of Technology, Dundalk, Ireland
| | - Neha Gupta
- Smooth Muscle Research Centre, Department of Life & Health Science, Dundalk Institute of Technology, Dundalk, Ireland
| | - Alexandru Mircea
- Smooth Muscle Research Centre, Department of Life & Health Science, Dundalk Institute of Technology, Dundalk, Ireland
| | - Caoimhin S Griffin
- Smooth Muscle Research Centre, Department of Life & Health Science, Dundalk Institute of Technology, Dundalk, Ireland
| |
Collapse
|
2
|
Gupta A, Manchanda R. Computational modeling of inhibitory signal transduction in urinary bladder PDGFRα+ cells. Comput Methods Biomech Biomed Engin 2024; 27:1161-1170. [PMID: 37424292 DOI: 10.1080/10255842.2023.2234063] [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: 02/24/2023] [Accepted: 06/30/2023] [Indexed: 07/11/2023]
Abstract
A crucial aspect of bladder function is the maintenance of a normo-active detrusor during bladder filling. The physiological mechanisms and pathways underlying this function are yet to be fully elucidated. Premature detrusor contractions are a key phenotype in detrusor overactivity, a common pathophysiological condition of the urinary bladder. Recent literature has identified PDFGRα+ cells as mediators in transducing inhibitory signals to detrusor smooth muscle cells via gap junctions. We employ computational modeling to study transduction pathways via which inhibitory signals are generated in PDFGRα+ cells in response to purinergic, nitrergic and mechanical stimuli. The key focus of our study here is to explore the effect of ATP, stretch and NO on the membrane potential of PDFGRα+ cells, which is driven to hyperpolarized potentials via the activation of SK3 channels. Our results indicate that purinergic, mechanical and nitrergic inputs can induce significant membrane hyperpolarizations of 20-35 mV relative to the resting membrane potential. Given the interconnections between PDFGRα+ cells and detrusor SMCs through gap junctions, these hyperpolarizations can have significant functional implications in the maintenance of a normo-active detrusor as also in departures from this state as seen in detrusor overactivity.
Collapse
Affiliation(s)
- Amritanshu Gupta
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Rohit Manchanda
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| |
Collapse
|
3
|
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
|
4
|
Li X, Hu J, Yin P, Liu L, Chen Y. Mechanotransduction in the urothelium: ATP signalling and mechanoreceptors. Heliyon 2023; 9:e19427. [PMID: 37674847 PMCID: PMC10477517 DOI: 10.1016/j.heliyon.2023.e19427] [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: 04/23/2023] [Revised: 08/10/2023] [Accepted: 08/22/2023] [Indexed: 09/08/2023] Open
Abstract
The urothelium, which covers the inner surface of the bladder, is continuously exposed to a complex physical environment where it is stimulated by, and responds to, a wide range of mechanical cues. Mechanically activated ion channels endow the urothelium with functioning in the conversion of mechanical stimuli into biochemical events that influence the surface of the urothelium itself as well as suburothelial tissues, including afferent nerve fibres, interstitial cells of Cajal and detrusor smooth muscle cells, to ensure normal urinary function during the cycle of filling and voiding. However, under prolonged and abnormal loading conditions, the urothelial sensory system can become maladaptive, leading to the development of bladder dysfunction. In this review, we summarize developments in the understanding of urothelial mechanotransduction from two perspectives: first, with regard to the functions of urothelial mechanotransduction, particularly stretch-mediated ATP signalling and the regulation of urothelial surface area; and secondly, with regard to the mechanoreceptors present in the urothelium, primarily transient receptor potential channels and mechanosensitive Piezo channels, and the potential pathophysiological role of these channels in the bladder. A more thorough understanding of urothelial mechanotransduction function may inspire the development of new therapeutic strategies for lower urinary tract diseases.
Collapse
Affiliation(s)
| | | | - Ping Yin
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Lumin Liu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Yuelai Chen
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| |
Collapse
|
5
|
Zhao M, Ding N, Wang H, Zu S, Liu H, Wen J, Liu J, Ge N, Wang W, Zhang X. Activation of TRPA1 in Bladder Suburothelial Myofibroblasts Counteracts TGF-β1-Induced Fibrotic Changes. Int J Mol Sci 2023; 24:ijms24119501. [PMID: 37298451 DOI: 10.3390/ijms24119501] [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: 02/23/2023] [Revised: 05/15/2023] [Accepted: 05/19/2023] [Indexed: 06/12/2023] Open
Abstract
The activation of the transient receptor potential ankyrin 1 (TRPA1) channel has anti-fibrotic effects in the lung and intestine. Suburothelial myofibroblasts (subu-MyoFBs), a specialized subset of fibroblasts in the bladder, are known to express TRPA1. However, the role of the TRPA1 in the development of bladder fibrosis remains elusive. In this study, we use the transforming growth factor-β1 (TGF-β1) to induce fibrotic changes in subu-MyoFBs and assess the consequences of TRPA1 activation utilizing RT-qPCR, western blotting, and immunocytochemistry. TGF-β1 stimulation increased α-SMA, collagen type I alpha 1 chain(col1A1), collagen type III (col III), and fibronectin expression, while simultaneously suppressing TRPA1 in cultured human subu-MyoFBs. The activation of TRPA1, with its specific agonist allylisothiocyanate (AITC), inhibited TGF-β1-induced fibrotic changes, and part of these inhibition effects could be reversed by the TRPA1 antagonist, HC030031, or by reducing TRPA1 expression via RNA interference. Furthermore, AITC reduced spinal cord injury-induced fibrotic bladder changes in a rat model. The increased expression of TGF-β1, α-SMA, col1A1 and col III, and fibronectin, and the downregulation of TRPA1, were also detected in the mucosa of fibrotic human bladders. These findings suggest that TRPA1 plays a pivotal role in bladder fibrosis, and the negative cross talk between TRPA1 and TGF-β1 signaling may represent one of the mechanisms underlying fibrotic bladder lesions.
Collapse
Affiliation(s)
- Mengmeng Zhao
- Department of Urology, The Second Hospital of Shandong University, Jinan 250033, China
| | - Ning Ding
- Department of Urology, The Second Hospital of Shandong University, Jinan 250033, China
| | - Haoyu Wang
- Department of Urology, The Second Hospital of Shandong University, Jinan 250033, China
| | - Shulu Zu
- Department of Urology, The Second Hospital of Shandong University, Jinan 250033, China
| | - Hanwen Liu
- Department of Urology, The Second Hospital of Shandong University, Jinan 250033, China
| | - Jiliang Wen
- Department of Urology, The Second Hospital of Shandong University, Jinan 250033, China
| | - Jiaxin Liu
- Department of Urology, The Second Hospital of Shandong University, Jinan 250033, China
| | - Nan Ge
- Department of Urology, The Second Hospital of Shandong University, Jinan 250033, China
| | - Wenzhen Wang
- Department of Urology, The Second Hospital of Shandong University, Jinan 250033, China
| | - Xiulin Zhang
- Department of Urology, The Second Hospital of Shandong University, Jinan 250033, China
| |
Collapse
|
6
|
Athavale ON, Avci R, Cheng LK, Du P. Computational models of autonomic regulation in gastric motility: Progress, challenges, and future directions. Front Neurosci 2023; 17:1146097. [PMID: 37008202 PMCID: PMC10050371 DOI: 10.3389/fnins.2023.1146097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 02/27/2023] [Indexed: 03/17/2023] Open
Abstract
The stomach is extensively innervated by the vagus nerve and the enteric nervous system. The mechanisms through which this innervation affects gastric motility are being unraveled, motivating the first concerted steps towards the incorporation autonomic regulation into computational models of gastric motility. Computational modeling has been valuable in advancing clinical treatment of other organs, such as the heart. However, to date, computational models of gastric motility have made simplifying assumptions about the link between gastric electrophysiology and motility. Advances in experimental neuroscience mean that these assumptions can be reviewed, and detailed models of autonomic regulation can be incorporated into computational models. This review covers these advances, as well as a vision for the utility of computational models of gastric motility. Diseases of the nervous system, such as Parkinson’s disease, can originate from the brain-gut axis and result in pathological gastric motility. Computational models are a valuable tool for understanding the mechanisms of disease and how treatment may affect gastric motility. This review also covers recent advances in experimental neuroscience that are fundamental to the development of physiology-driven computational models. A vision for the future of computational modeling of gastric motility is proposed and modeling approaches employed for existing mathematical models of autonomic regulation of other gastrointestinal organs and other organ systems are discussed.
Collapse
|
7
|
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
|
8
|
Perkins ME, Vizzard MA. Transient receptor potential vanilloid type 4 (TRPV4) in urinary bladder structure and function. CURRENT TOPICS IN MEMBRANES 2022; 89:95-138. [PMID: 36210154 PMCID: PMC10486315 DOI: 10.1016/bs.ctm.2022.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Bladder pain syndrome (BPS)/interstitial cystitis (IC) is a urologic, chronic pelvic pain syndrome characterized by pelvic pain, pressure, or discomfort with urinary symptoms. Symptom exacerbation (flare) is common with multiple, perceived triggers including stress. Multiple transient receptor potential (TRP) channels (TRPA1, TRPV1, TRPV4) expressed in the bladder have specific tissue distributions in the lower urinary tract (LUT) and are implicated in bladder disorders including overactive bladder (OAB) and BPS/IC. TRPV4 channels are strong candidates for mechanosensors in the urinary bladder and TRPV4 antagonists are promising therapeutic agents for OAB. In this perspective piece, we address the current knowledge of TRPV4 distribution and function in the LUT and its plasticity with injury or disease with an emphasis on BPS/IC. We review our studies that extend the knowledge of TRPV4 in urinary bladder function by focusing on (i) TRPV4 involvement in voiding dysfunction, pelvic pain, and non-voiding bladder contractions in NGF-OE mice; (ii) distention-induced luminal ATP release mechanisms and (iii) involvement of TRPV4 and vesicular release mechanisms. Finally, we review our lamina propria studies in postnatal rat studies that demonstrate: (i) the predominance of the TRPV4+ and PDGFRα+ lamina propria cellular network in early postnatal rats; (ii) the ability of exogenous mediators (i.e., ATP, TRPV4 agonist) to activate and increase the number of lamina propria cells exhibiting active Ca2+ events; and (iii) the ability of ATP and TRPV4 agonist to increase the rate of integrated Ca2+ activity corresponding to coupled lamina propria network events and the formation of propagating wavefronts.
Collapse
Affiliation(s)
- Megan Elizabeth Perkins
- Department of Neurological Sciences, The Larner College of Medicine, The University of Vermont, Burlington, VT, United States
| | - Margaret A Vizzard
- Department of Neurological Sciences, The Larner College of Medicine, The University of Vermont, Burlington, VT, United States.
| |
Collapse
|
9
|
Hiroshige T, Uemura KI, Hirashima S, Togo A, Ohta K, Nakamura KI, Igawa T. Three-dimensional analysis of interstitial cells in the lamina propria of the murine vas deferens by confocal laser scanning microscopy and FIB/SEM. Sci Rep 2022; 12:9484. [PMID: 35676513 PMCID: PMC9177838 DOI: 10.1038/s41598-022-13245-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 05/23/2022] [Indexed: 11/09/2022] Open
Abstract
The present study aimed to explore the three-dimensional (3D) ultrastructure of interstitial cells (ICs) within the lamina propria of the murine vas deferens and the spatial relationships between epithelial cells and surrounding cells. Focused ion beam scanning electron microscopy and confocal laser scanning microscopy were performed. ICs within the lamina propria had a flat, sheet-like structure of cytoplasm with multiple cellular processes. In addition, two types of 3D structures that comprised cell processes of flat, sheet-like ICs were observed: one was an accordion fold-like structure and the other was a rod-shaped structure. ICs were located parallel to the epithelium and were connected to each other via gap junctions or adherens junctions. Moreover, multiple sphere-shaped extracellular vesicle-like structures were frequently observed around the ICs. The ICs formed a complex 3D network comprising sheet-like cytoplasm and multiple cell processes with different 3D structures. From this morphological study, we noted that ICs within the lamina propria of murine vas deferens may be involved in signal transmission between the epithelium and smooth muscle cells by physical interaction and by exchanging extracellular vesicles.
Collapse
Affiliation(s)
- Tasuku Hiroshige
- Department of Urology, Kurume University School of Medicine, Kurume, 830-0011, Japan.
| | - Kei-Ichiro Uemura
- Department of Urology, Kurume University School of Medicine, Kurume, 830-0011, Japan
| | - Shingo Hirashima
- Division Microscopic and Development Anatomy, Department of Anatomy, Kurume University School of Medicine, Kurume, 830-0011, Japan
| | - Akinobu Togo
- Advanced Imaging Research Center, Kurume University School of Medicine, Kurume, 830-0011, Japan
| | - Keisuke Ohta
- Division Microscopic and Development Anatomy, Department of Anatomy, Kurume University School of Medicine, Kurume, 830-0011, Japan.,Advanced Imaging Research Center, Kurume University School of Medicine, Kurume, 830-0011, Japan
| | - Kei-Ichiro Nakamura
- Cognitive and Molecular Research Institute of Brain Diseases, Kurume University School of Medicine, Kurume, 830-0011, Japan
| | - Tsukasa Igawa
- Department of Urology, Kurume University School of Medicine, Kurume, 830-0011, Japan
| |
Collapse
|
10
|
Perkins ME, Girard BM, Campbell SE, Vizzard MA. Imatinib Mesylate Reduces Voiding Frequency in Female Mice With Acute Cyclophosphamide-Induced Cystitis. Front Syst Neurosci 2022; 16:867875. [PMID: 35645740 PMCID: PMC9135974 DOI: 10.3389/fnsys.2022.867875] [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: 02/01/2022] [Accepted: 04/19/2022] [Indexed: 01/28/2023] Open
Abstract
Lamina propria interstitial cells that express the tyrosine kinase receptor, platelet-derived growth factor receptor alpha (PDGFRα) may play a role in urinary sensory signaling. Imatinib mesylate, also referred to as imatinib, is a tyrosine kinase inhibitor that can inhibit PDGFRα and has been widely used in urological research. We evaluated the functional effects of imatinib administration (via oral gavage or intravesical infusion) with two different experimental designs (prevention and treatment), in a cyclophosphamide (CYP)-induced cystitis (acute, intermediate, and chronic), male and female rodent model using conscious cystometry and somatic sensitivity testing. Imatinib significantly (0.0001 ≤ p ≤ 0.05) decreased voiding frequency and increased bladder capacity in acute CYP-induced cystitis, by the prevention (females) and treatment (females and males) designs. Imatinib was not effective in preventing or treating intermediate or chronic CYP-induced cystitis in either sex. Interestingly, in the prevention experiments, imatinib administration increased (0.0001 ≤ p ≤ 0.01) voiding frequency and decreased bladder capacity in control mice. However, in the treatment experiments, imatinib administration decreased (0.01 ≤ p ≤ 0.05) voiding frequency and increased bladder capacity in control mice. Bladder function improvements observed with imatinib treatment in acute CYP-induced cystitis mice remained and additionally improved with a second dose of imatinib 24 hours after CYP treatment. Imatinib administration did not affect pelvic somatic sensitivity in female mice with acute CYP-induced cystitis. Our studies suggest that (1) imatinib improves bladder function in mice with acute CYP-induced cystitis with a prevention and treatment design and (2) interstitial cells may be a useful target to improve bladder function in cystitis.
Collapse
|
11
|
Perkins M, Girard BM, Campbell SE, Hennig GW, Vizzard MA. Imatinib Mesylate Reduces Neurotrophic Factors and pERK and pAKT Expression in Urinary Bladder of Female Mice With Cyclophosphamide-Induced Cystitis. Front Syst Neurosci 2022; 16:884260. [PMID: 35528149 PMCID: PMC9072830 DOI: 10.3389/fnsys.2022.884260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 03/28/2022] [Indexed: 01/28/2023] Open
Abstract
Imatinib mesylate is a tyrosine kinase inhibitor that inhibits platelet-derived growth factor receptor (PDGFR)-α, -β, stem cell factor receptor (c-KIT), and BCR-ABL. PDGFRα is expressed in a subset of interstitial cells in the lamina propria (LP) and detrusor muscle of the urinary bladder. PDGFRα + interstitial cells may contribute to bladder dysfunction conditions such as interstitial cystitis/bladder pain syndrome (IC/BPS) or overactive bladder (OAB). We have previously demonstrated that imatinib prevention via oral gavage or treatment via intravesical infusion improves urinary bladder function in mice with acute (4 hour, h) cyclophosphamide (CYP)-induced cystitis. Here, we investigate potential underlying mechanisms mediating the bladder functional improvement by imatinib using a prevention or treatment experimental design. Using qRT-PCR and ELISAs, we examined inflammatory mediators (NGF, VEGF, BDNF, CCL2, IL-6) previously shown to affect bladder function in CYP-induced cystitis. We also examined the distribution of phosphorylated (p) ERK and pAKT expression in the LP with immunohistochemistry. Imatinib prevention significantly (0.0001 ≤ p ≤ 0.05) reduced expression for all mediators examined except NGF, whereas imatinib treatment was without effect. Imatinib prevention and treatment significantly (0.0001 ≤ p ≤ 0.05) reduced pERK and pAKT expression in the upper LP (U. LP) and deeper LP (D. LP) in female mice with 4 h CYP-induced cystitis. Although we have previously demonstrated that imatinib prevention or treatment improves bladder function in mice with cystitis, the current studies suggest that reductions in inflammatory mediators contribute to prevention benefits of imatinib but not the treatment benefits of imatinib. Differential effects of imatinib prevention or treatment on inflammatory mediators may be influenced by the route and frequency of imatinib administration and may also suggest other mechanisms (e.g., changes in transepithelial resistance of the urothelium) through which imatinib may affect urinary bladder function following CYP-induced cystitis.
Collapse
Affiliation(s)
- Megan Perkins
- Department of Neurological Sciences, The Larner College of Medicine, The University of Vermont, Burlington, VT, United States
| | - Beatrice M. Girard
- Department of Neurological Sciences, The Larner College of Medicine, The University of Vermont, Burlington, VT, United States
| | - Susan E. Campbell
- Department of Neurological Sciences, The Larner College of Medicine, The University of Vermont, Burlington, VT, United States
| | - Grant W. Hennig
- Department of Pharmacology, The Larner College of Medicine, The University of Vermont, Burlington, VT, United States
| | - Margaret A. Vizzard
- Department of Neurological Sciences, The Larner College of Medicine, The University of Vermont, Burlington, VT, United States
| |
Collapse
|
12
|
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
|
13
|
Hiroshige T, Uemura KI, Hirashima S, Hino K, Togo A, Ohta K, Igawa T, Nakamura KI. Three-Dimensional Analysis of Interstitial Cells in the Smooth Muscle Layer of Murine Vas Deferens Using Confocal Laser Scanning Microscopy and FIB/SEM. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2022; 28:1-9. [PMID: 35078549 DOI: 10.1017/s1431927622000058] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The smooth muscle contraction of the vas deferens has the important function of transporting sperm. Interstitial cells (ICs) play a critical role in the pacing and modulation of various smooth muscle organs by interactions with nerves and smooth muscle. Elucidating the three-dimensional (3D) architecture of ICs is important for understanding their spatial relationship on the mesoscale between ICs, smooth muscle cells (SMCs), and nerves. In this study, the 3D ultrastructure of ICs in the smooth muscle layer of murine vas deferens and the spatial relationships between ICs, nerves, and smooth muscles were observed using confocal laser scanning microscopy and focused ion beam/scanning electron microscopy. ICs have sheet-like structures as demonstrated by 3D observation using modern analytical techniques. Sheet-like ICs have two types of 3D structures, one flattened and the other curled. Multiple extracellular vesicle (EV)-like structures were frequently observed in ICs. Various spatial relations were observed in areas between ICs, nerves, and SMCs, which formed a complex 3D network with each other. These results suggest that ICs in the smooth muscle layer of murine vas deferens may have two subtypes with different sheet-like structures and may be involved in neuromuscular signal transmission via physical interaction and EVs.
Collapse
Affiliation(s)
- Tasuku Hiroshige
- Department of Urology, Kurume University School of Medicine, Kurume830-0011, Japan
- Division of Microscopic and Development Anatomy, Department of Anatomy, Kurume University School of Medicine, Kurume830-0011, Japan
| | - Kei-Ichiro Uemura
- Department of Urology, Kurume University School of Medicine, Kurume830-0011, Japan
| | - Shingo Hirashima
- Division of Microscopic and Development Anatomy, Department of Anatomy, Kurume University School of Medicine, Kurume830-0011, Japan
| | - Kiyosato Hino
- Division of Microscopic and Development Anatomy, Department of Anatomy, Kurume University School of Medicine, Kurume830-0011, Japan
| | - Akinobu Togo
- Advanced Imaging Research Center, Kurume University School of Medicine, Kurume830-0011, Japan
| | - Keisuke Ohta
- Division of Microscopic and Development Anatomy, Department of Anatomy, Kurume University School of Medicine, Kurume830-0011, Japan
- Advanced Imaging Research Center, Kurume University School of Medicine, Kurume830-0011, Japan
| | - Tsukasa Igawa
- Department of Urology, Kurume University School of Medicine, Kurume830-0011, Japan
| | - Kei-Ichiro Nakamura
- Division of Microscopic and Development Anatomy, Department of Anatomy, Kurume University School of Medicine, Kurume830-0011, Japan
- Cognitive and Molecular Research Institute of Brain Diseases, Kurume University School of Medicine, Kurume830-0011, Japan
| |
Collapse
|
14
|
Zhao M, Chen Z, Liu L, Ding N, Wen J, Liu J, Wang W, Ge N, Zu S, Song W, Chen G, Zhang X. Functional Expression of Transient Receptor Potential and Piezo1 Channels in Cultured Interstitial Cells of Human-Bladder Lamina Propria. Front Physiol 2022; 12:762847. [PMID: 35069237 PMCID: PMC8774296 DOI: 10.3389/fphys.2021.762847] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 12/03/2021] [Indexed: 01/25/2023] Open
Abstract
The interstitial cells in bladder lamina propria (LP-ICs) are believed to be involved in sensing/afferent signaling in bladder mucosa. Transient receptor potential (TRP) cation channels act as mechano- or chemo-sensors and may underlie some of the sensing function of bladder LP-ICs. We aimed to investigate the molecular and functional expression of TRP channels implicated in bladder sensory function and Piezo1/Piezo2 channels in cultured LP-ICs of the human bladder. Bladder tissues were obtained from patients undergoing cystectomy. LP-ICs were isolated and cultured, and used for real-time reverse transcription-quantitative polymerase chain reaction, immunocytochemistry, and calcium-imaging experiments. At the mRNA level, TRPA1, TRPV2, and Piezo1 were expressed most abundantly. Immunocytochemical staining showed protein expression of TRPA1, TRPV1, TRPV2, TRPV4, TRPM8, as well as Piezo1 and Piezo2. Calcium imaging using channel agonists/antagonists provided evidence for functional expression of TRPA1, TRPV2, TRPV4, Piezo1, but not of TRPV1 or TRPM8. Activation of these channels with their agonist resulted in release of adenosine triphosphate (ATP) from LP-ICs. Inhibition of TRPV2, TRPV4 and Piezo1 blocked the stretch induced intracellular Ca2+ increase. Whereas inhibition of TRPA1 blocked H2O2 evoked response in LP-ICs. Our results suggest LP-ICs of the bladder can perceive stretch or chemical stimuli via activation of TRPV2, TRPV4, Piezo1 and TRPA1 channels. LP-ICs may work together with urothelial cells for perception and transduction of mechanical or chemical signals in human-bladder mucosa.
Collapse
Affiliation(s)
- MengMeng Zhao
- Department of Urology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zhenghao Chen
- Department of Urology, Friendship Hospital, Capital Medical University, Beijing, China
| | - Lei Liu
- Department of Urology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Ning Ding
- Department of Urology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jiliang Wen
- Department of Urology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jiaxin Liu
- Department of Urology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - WenZhen Wang
- Department of Urology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Nan Ge
- Department of Urology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Shulu Zu
- Department of Urology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wei Song
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Guoqing Chen
- Department of Urology, China Rehabilitation Research Center, School of Rehabilitation, Capital Medical University, Beijing, China
| | - Xiulin Zhang
- Department of Urology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
15
|
Shi B, Wu Y, Chen H, Ding J, Qi J. Understanding of mouse and human bladder at single-cell resolution: integrated analysis of trajectory and cell-cell interactive networks based on multiple scRNA-seq datasets. Cell Prolif 2021; 55:e13170. [PMID: 34951074 PMCID: PMC8780900 DOI: 10.1111/cpr.13170] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 11/11/2021] [Accepted: 11/30/2021] [Indexed: 11/29/2022] Open
Abstract
Objectives To elaborately decipher the mouse and human bladders at single‐cell levels. Materials and Methods We collected more than 50,000 cells from multiple datasets and created, up to date, the largest integrated bladder datasets. Pseudotime trajectory of urothelium and interstitial cells, as well as dynamic cell‐cell interactions, was investigated. Biological activity scores and different roles of signaling pathways between certain cell clusters were also identified. Results The glucose score was significantly high in most urothelial cells, while the score of H3 acetylation was roughly equally distributed across all cell types. Several genes via a pseudotime pattern in mouse (Car3, Dkk2, Tnc, etc.) and human (FBLN1, S100A10, etc.) were discovered. S100A6, TMSB4X, and typical uroplakin genes seemed as shared pseudotime genes for urothelial cells in both human and mouse datasets. In combinational mouse (n = 16,688) and human (n = 22,080) bladders, we verified 1,330 and 1,449 interactive ligand‐receptor pairs, respectively. The distinct incoming and outgoing signaling was significantly associated with specific cell types. Collagen was the strongest signal from fibroblasts to urothelial basal cells in mouse, while laminin pathway for urothelial basal cells to smooth muscle cells (SMCs) in human. Fibronectin 1 pathway was intensely sent by myofibroblasts, received by urothelial cells, and almost exclusively mediated by SMCs in mouse bladder. Interestingly, the cell cluster of SMCs 2 was the dominant sender and mediator for Notch signaling in the human bladder, while SMCs 1 was not. The expression of integrin superfamily (the most common communicative pairs) was depicted, and their co‐expression patterns were located in certain cell types (eg, Itgb1 and Itgb4 in mouse and human basal cells). Conclusions This study provides a complete interpretation of the normal bladder at single‐cell levels, offering an in‐depth resource and foundation for future research.
Collapse
Affiliation(s)
- Bowen Shi
- Department of Urology, School of Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Yanyuan Wu
- Department of Urology, School of Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Haojie Chen
- Department of Urology, School of Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Jie Ding
- Department of Urology, School of Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Jun Qi
- Department of Urology, School of Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
16
|
Çakıcı ÖU, Dinçer S. The effect of amino acids on the bladder cycle: a concise review. Amino Acids 2021; 54:13-31. [PMID: 34853916 DOI: 10.1007/s00726-021-03113-5] [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: 06/17/2021] [Accepted: 11/25/2021] [Indexed: 11/26/2022]
Abstract
The human bladder maintains a cycle of filling, storing, and micturating throughout an individual's lifespan. The cycle relies on the ability of the bladder to expand without increasing the intravesical pressure, which is only possible with the controlled relaxation of well-complaint muscles and the congruously organized construction of the bladder wall. A competent bladder outlet, which functions in a synchronous fashion with the bladder, is also necessary for this cycle to be completed successfully without deterioration. In this paper, we aimed to review the contemporary physiological findings on bladder physiology and examine the effects of amino acids on clinical conditions affecting the bladder, with special emphasis on the available therapeutic evidence and possible future roles of the amino acids in the treatment of the bladder-related disorders.
Collapse
Affiliation(s)
- Özer Ural Çakıcı
- Attending Urologist, Private Practice, Ankara, Turkey.
- PhD Candidate in Physiology, Department of Physiology, Gazi University, Ankara, Turkey.
| | - Sibel Dinçer
- Professor in Physiology, Department of Physiology, Gazi University, Ankara, Turkey
| |
Collapse
|
17
|
Lee K, Park SO, Choi PC, Ryoo SB, Lee H, Peri LE, Zhou T, Corrigan RD, Yanez AC, Moon SB, Perrino BA, Sanders KM, Koh SD. Molecular and functional characterization of detrusor PDGFRα positive cells in spinal cord injury-induced detrusor overactivity. Sci Rep 2021; 11:16268. [PMID: 34381120 PMCID: PMC8357952 DOI: 10.1038/s41598-021-95781-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 07/23/2021] [Indexed: 12/02/2022] Open
Abstract
Volume accommodation occurs via a novel mechanism involving interstitial cells in detrusor muscles. The interstitial cells in the bladder are PDGFRα+, and they restrain the excitability of smooth muscle at low levels and prevents the development of transient contractions (TCs). A common clinical manifestation of spinal cord injury (SCI)-induced bladder dysfunction is detrusor overactivity (DO). Although a myogenic origin of DO after SCI has been suggested, a mechanism for development of SCI-induced DO has not been determined. In this study we hypothesized that SCI-induced DO is related to loss of function in the regulatory mechanism provided by PDGFRα+ cells. Our results showed that transcriptional expression of Pdgfra and Kcnn3 was decreased after SCI. Proteins encoded by these genes also decreased after SCI, and a reduction in PDGFRα+ cell density was also documented. Loss of PDGFRα+ cells was due to apoptosis. TCs in ex vivo bladders during filling increased dramatically after SCI, and this was related to the loss of regulation provided by SK channels, as we observed decreased sensitivity to apamin. These findings show that damage to the mechanism restraining muscle contraction during bladder filling that is provided by PDGFRα+ cells is causative in the development of DO after SCI.
Collapse
Affiliation(s)
- Ken Lee
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, 89557, USA
| | - Sang O Park
- Department of Emergency Medicine, Konkuk University School of Medicine, Seoul, South Korea
| | - Pil-Cho Choi
- Department of Emergency Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University, Seoul, South Korea
| | - Seung-Bum Ryoo
- Department of Surgery, Seoul National University Hospital, College of Medicine, Seoul National University, Seoul, South Korea
| | - Haeyeong Lee
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, 89557, USA
| | - Lauren E Peri
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, 89557, USA
| | - Tong Zhou
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, 89557, USA
| | - Robert D Corrigan
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, 89557, USA
| | - Andrew C Yanez
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, 89557, USA
| | - Suk B Moon
- Department of Surgery, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Brian A Perrino
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, 89557, USA
| | - Kenton M Sanders
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, 89557, USA
| | - Sang Don Koh
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, 89557, USA.
| |
Collapse
|
18
|
Drumm BT, Thornbury KD, Hollywood MA, Sergeant GP. Role of Ano1 Ca 2+-activated Cl - channels in generating urethral tone. Am J Physiol Renal Physiol 2021; 320:F525-F536. [PMID: 33554780 DOI: 10.1152/ajprenal.00520.2020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Urinary continence is maintained in the lower urinary tract by the contracture of urethral sphincters, including smooth muscle of the internal urethral sphincter. These contractions occlude the urethral lumen, preventing urine leakage from the bladder to the exterior. Over the past 20 years, research on the ionic conductances that contribute to urethral smooth muscle contractility has greatly accelerated. A debate has emerged over the role of interstitial cell of Cajal (ICC)-like cells in the urethra and their expression of Ca2+-activated Cl- channels encoded by anoctamin-1 [Ano1; transmembrane member 16 A (Tmem16a) gene]. It has been proposed that Ano1 channels expressed in urethral ICC serve as a source of depolarization for smooth muscle cells, increasing their excitability and contributing to tone. Although a clear role for Ano1 channels expressed in ICC is evident in other smooth muscle organs, such as the gastrointestinal tract, the role of these channels in the urethra is unclear, owing to differences in the species (rabbit, rat, guinea pig, sheep, and mouse) examined and experimental approaches by different groups. The importance of clarifying this situation is evident as effective targeting of Ano1 channels may lead to new treatments for urinary incontinence. In this review, we summarize the key findings from different species on the role of ICC and Ano1 channels in urethral contractility. Finally, we outline proposals for clarifying this controversial and important topic by addressing how cell-specific optogenetic and inducible cell-specific genetic deletion strategies coupled with advances in Ano1 channel pharmacology may clarify this area in future studies.NEW & NOTEWORTHY Studies from the rabbit have shown that anoctamin-1 (Ano1) channels expressed in urethral interstitial cells of Cajal (ICC) serve as a source of depolarization for smooth muscle cells, increasing excitability and tone. However, the role of urethral Ano1 channels is unclear, owing to differences in the species examined and experimental approaches. We summarize findings from different species on the role of urethral ICC and Ano1 channels in urethral contractility and outline proposals for clarifying this topic using cell-specific optogenetic approaches.
Collapse
Affiliation(s)
- Bernard T Drumm
- Smooth Muscle Research Centre, Dundalk Institute of Technology, Dundalk, Ireland
| | - Keith D Thornbury
- Smooth Muscle Research Centre, Dundalk Institute of Technology, Dundalk, Ireland
| | - Mark A Hollywood
- Smooth Muscle Research Centre, Dundalk Institute of Technology, Dundalk, Ireland
| | - Gerard P Sergeant
- Smooth Muscle Research Centre, Dundalk Institute of Technology, Dundalk, Ireland
| |
Collapse
|
19
|
Dalghi MG, Montalbetti N, Carattino MD, Apodaca G. The Urothelium: Life in a Liquid Environment. Physiol Rev 2020; 100:1621-1705. [PMID: 32191559 PMCID: PMC7717127 DOI: 10.1152/physrev.00041.2019] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 03/02/2020] [Accepted: 03/14/2020] [Indexed: 02/08/2023] Open
Abstract
The urothelium, which lines the renal pelvis, ureters, urinary bladder, and proximal urethra, forms a high-resistance but adaptable barrier that surveils its mechanochemical environment and communicates changes to underlying tissues including afferent nerve fibers and the smooth muscle. The goal of this review is to summarize new insights into urothelial biology and function that have occurred in the past decade. After familiarizing the reader with key aspects of urothelial histology, we describe new insights into urothelial development and regeneration. This is followed by an extended discussion of urothelial barrier function, including information about the roles of the glycocalyx, ion and water transport, tight junctions, and the cellular and tissue shape changes and other adaptations that accompany expansion and contraction of the lower urinary tract. We also explore evidence that the urothelium can alter the water and solute composition of urine during normal physiology and in response to overdistension. We complete the review by providing an overview of our current knowledge about the urothelial environment, discussing the sensor and transducer functions of the urothelium, exploring the role of circadian rhythms in urothelial gene expression, and describing novel research tools that are likely to further advance our understanding of urothelial biology.
Collapse
Affiliation(s)
- Marianela G Dalghi
- Department of Medicine, Renal-Electrolyte Division, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Nicolas Montalbetti
- Department of Medicine, Renal-Electrolyte Division, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Marcelo D Carattino
- Department of Medicine, Renal-Electrolyte Division, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Gerard Apodaca
- Department of Medicine, Renal-Electrolyte Division, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
20
|
Gabella G. Muscle cells, nerves, fibroblasts and vessels in the detrusor of the rat urinary bladder. J Smooth Muscle Res 2020; 55:34-67. [PMID: 31708509 PMCID: PMC6851244 DOI: 10.1540/jsmr.55.34] [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] [Indexed: 01/05/2023] Open
Abstract
All the cells of rat detrusor muscle fall into one of five ultrastructural types: muscle
cells, fibroblasts, axons and glia, and vascular cells (endothelial cells and pericytes).
The tissue is ~79% cellular and 21% non-cellular. Muscle cells occupy 72%, nerves ~4% (1/3
axons, 2/3 glia), and fibroblast >3% of space. Muscle cells (up to 6 µm across and ~600
µm long, packed to almost 100,000 per mm2) have surface-to-volume ratio of 2.4
µm2/µm3 ~93% of cell volume is contractile apparatus, 3.1%
mitochondria and 2.5% nucleus. Cell profiles are irregular but sectional area decreases
regularly towards either end of the cell. Muscle cells are gathered into bundles (the
mechanical units of detrusor), variable in length and size, but of constant width. The
musculature is highly compact (without fascia or capsule) with smooth outer surfaces and
extensive association and adhesion between its cells. Among many types of intercellular
contact and junction, digitations are very common, each muscle cell issuing minute
finger-like processes that abut on adjacent cells. Sealed apposition are wide areas of
specialized contact, possibly forming a chamber between two muscle cells, distinct from
the extracellular space at large (stromal space). The innervation is very dense, virtually
all intramuscular axons being varicose (including afferent ones). There are identifiable
neuro-muscular junctions on each muscle cell, often several junctions on a single cell.
There are also unattached terminals. Fibroblasts (involved in the production of collagen),
~1% of the total number of cells, do not make specialized contacts.
Collapse
|
21
|
Yu Z, Liao J, Chen Y, Zou C, Zhang H, Cheng J, Liu D, Li T, Zhang Q, Li J, Yang X, Ye Y, Huang Z, Long X, Yang R, Mo Z. Single-Cell Transcriptomic Map of the Human and Mouse Bladders. J Am Soc Nephrol 2019; 30:2159-2176. [PMID: 31462402 DOI: 10.1681/asn.2019040335] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 07/21/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Having a comprehensive map of the cellular anatomy of the normal human bladder is vital to understanding the cellular origins of benign bladder disease and bladder cancer. METHODS We used single-cell RNA sequencing (scRNA-seq) of 12,423 cells from healthy human bladder tissue samples taken from patients with bladder cancer and 12,884 cells from mouse bladders to classify bladder cell types and their underlying functions. RESULTS We created a single-cell transcriptomic map of human and mouse bladders, including 16 clusters of human bladder cells and 15 clusters of mouse bladder cells. The homology and heterogeneity of human and mouse bladder cell types were compared and both conservative and heterogeneous aspects of human and mouse bladder evolution were identified. We also discovered two novel types of human bladder cells. One type is ADRA2A + and HRH2 + interstitial cells which may be associated with nerve conduction and allergic reactions. The other type is TNNT1 + epithelial cells that may be involved with bladder emptying. We verify these TNNT1 + epithelial cells also occur in rat and mouse bladders. CONCLUSIONS This transcriptomic map provides a resource for studying bladder cell types, specific cell markers, signaling receptors, and genes that will help us to learn more about the relationship between bladder cell types and diseases.
Collapse
Affiliation(s)
- Zhenyuan Yu
- Institute of Urology and Nephrology.,Center for Genomic and Personalized Medicine.,Departments of Urology and.,Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, China.,Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Key Laboratory of Colleges and Universities, Nanning, China
| | - Jinling Liao
- Center for Genomic and Personalized Medicine.,Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, China.,Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Key Laboratory of Colleges and Universities, Nanning, China
| | - Yang Chen
- Institute of Urology and Nephrology.,Center for Genomic and Personalized Medicine.,Departments of Urology and.,Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, China.,Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Key Laboratory of Colleges and Universities, Nanning, China
| | - Chunlin Zou
- Key Laboratory of Longevity and Ageing-related Diseases, Ministry of Education, Nanning, China.,Center for Translational Medicine, Guangxi Medical University, Nanning, China
| | - Haiying Zhang
- Center for Genomic and Personalized Medicine.,Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, China.,Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Key Laboratory of Colleges and Universities, Nanning, China
| | - Jiwen Cheng
- Institute of Urology and Nephrology.,Center for Genomic and Personalized Medicine.,Departments of Urology and.,Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, China.,Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Key Laboratory of Colleges and Universities, Nanning, China
| | - Deyun Liu
- Institute of Urology and Nephrology.,Departments of Urology and
| | - Tianyu Li
- Institute of Urology and Nephrology.,Departments of Urology and
| | - Qingyun Zhang
- Institute of Urology and Nephrology.,Center for Genomic and Personalized Medicine.,Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, China.,Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Key Laboratory of Colleges and Universities, Nanning, China.,Department of Urology, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, China
| | - Jiaping Li
- Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory of Precision Medicine in Cardio-cerebrovascular Diseases Control and Prevention, Nanning, China.,Guangxi Clinical Research Center for Cardio-cerebrovascular Diseases, Nanning, China; and
| | - Xiaobo Yang
- Center for Genomic and Personalized Medicine.,Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, China.,Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Key Laboratory of Colleges and Universities, Nanning, China
| | - Yu Ye
- Institute of Urology and Nephrology.,Center for Genomic and Personalized Medicine.,Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, China.,Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Key Laboratory of Colleges and Universities, Nanning, China.,Scientific Research Department, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Zhiguang Huang
- Center for Genomic and Personalized Medicine.,Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, China.,Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Key Laboratory of Colleges and Universities, Nanning, China
| | - Xinyang Long
- Center for Genomic and Personalized Medicine.,Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, China.,Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Key Laboratory of Colleges and Universities, Nanning, China
| | - Rirong Yang
- Center for Genomic and Personalized Medicine, .,Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, China.,Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Key Laboratory of Colleges and Universities, Nanning, China.,Department of Immunology, School of Preclinical Medicine, Guangxi Medical University, Nanning, China
| | - Zengnan Mo
- Institute of Urology and Nephrology, .,Center for Genomic and Personalized Medicine.,Departments of Urology and.,Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, China.,Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Key Laboratory of Colleges and Universities, Nanning, China
| |
Collapse
|
22
|
Hao Y, Wang L, Chen H, Hill WG, Robson SC, Zeidel ML, Yu W. Targetable purinergic receptors P2Y12 and A2b antagonistically regulate bladder function. JCI Insight 2019; 4:122112. [PMID: 31434806 DOI: 10.1172/jci.insight.122112] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 07/26/2019] [Indexed: 12/21/2022] Open
Abstract
Abnormalities in purine availability or purinergic receptor density are commonly seen in patients with lower urinary tract symptoms (LUTS), but the underlying mechanisms relating altered receptor function to LUTS are unknown. Here we provide extensive evidence for the reciprocal interplay of multiple receptors responding to ATP, ADP (adenosine diphosphate), and adenosine, agonists that regulate bladder function significantly. ADP stimulated P2Y12 receptors, causing bladder smooth muscle (BSM) contraction, whereas adenosine signaling through potentially newly defined A2b receptors, actively inhibited BSM purinergic contractility. The modulation of adenylyl cyclase-cAMP signaling via A2b and P2Y12 interaction actively regulated bladder contractility by modulating intracellular calcium levels. KO mice lacking the receptors display diametrically opposed bladder phenotypes, with P2Y12-KO mice exhibiting an underactive bladder (UAB) phenotype with increased bladder capacity and reduced voiding frequency, whereas A2b-KO mice have an overactive bladder (OAB), with decreased capacity and increased voiding frequency. The opposing phenotypes in P2Y12-KO and A2b-KO mice not only resulted from dysregulated BSM contractility, but also from abnormal BSM cell growth. Finally, we demonstrate that i.p. administration of drugs targeting P2Y12 or A2b receptor rescues these abnormal phenotypes in both KO mice. These findings strongly indicate that P2Y12 and A2b receptors are attractive therapeutic targets for human patients with LUTS.
Collapse
Affiliation(s)
- Yuan Hao
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Lu Wang
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA.,Chongqing University, Chongqing, China
| | - Huan Chen
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Warren G Hill
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Simon C Robson
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Mark L Zeidel
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Weiqun Yu
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
23
|
Durnin L, Kwok B, Kukadia P, McAvera R, Corrigan RD, Ward SM, Zhang Y, Chen Q, Koh SD, Sanders KM, Mutafova-Yambolieva VN. An ex vivo bladder model with detrusor smooth muscle removed to analyse biologically active mediators released from the suburothelium. J Physiol 2018; 597:1467-1485. [PMID: 30289177 DOI: 10.1113/jp276924] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 10/02/2018] [Indexed: 12/15/2022] Open
Abstract
KEY POINTS Studies of urothelial cells, bladder sheets or lumens of filled bladders have suggested that mediators released from urothelium into suburothelium (SubU)/lamina propria (LP) activate mechanisms controlling detrusor excitability. None of these approaches, however, has enabled direct assessment of availability of mediators at SubU/LP during filling. We developed an ex vivo mouse bladder preparation with intact urothelium and SubU/LP but no detrusor, which allows direct access to the SubU/LP surface of urothelium during filling. Pressure-volume measurements during filling demonstrated that bladder compliance is governed primarily by the urothelium. Measurements of purine mediators in this preparation demonstrated asymmetrical availability of purines in lumen and SubU/LP, suggesting that interpretations based solely on intraluminal measurements of mediators may be inaccurate. The preparations are suitable for assessments of release, degradation and transport of mediators in SubU/LP during bladder filling, and are superior to experimental approaches previously used for urothelium research. ABSTRACT The purpose of this study was to develop a decentralized (ex vivo) detrusor smooth muscle (DSM)-denuded mouse bladder preparation, a novel model that enables studies on availability of urothelium-derived mediators at the luminal and anti-luminal aspects of the urothelium during filling. Urinary bladders were excised from C57BL6/J mice and the DSM was removed by fine-scissor dissection without touching the mucosa. Morphology and cell composition of the preparation wall, pressure-volume relationships during filling, and fluorescent dye permeability of control, protamine sulfate- and lipopolysaccharide-treated denuded bladders were characterized. The preparation wall contained intact urothelium and suburothelium (SubU)/lamina propria (LP) and lacked the DSM and the serosa. The utility of the model for physiological research was validated by measuring release, metabolism and transport of purine mediators at SubU/LP and in bladder lumen during filling. We determined asymmetrical availability of purines (e.g. ATP, ADP, AMP and adenosine) in lumen and at SubU/LP during filling, suggesting differential mechanisms of release, degradation and bilateral transurothelial transport of purines during filling. Some observations were validated in DSM-denuded bladder of the cynomolgus monkey (Macaca fascicularis). The novel model was superior to current models utilized to study properties of the urothelium (e.g. cultured urothelial cells, bladder mucosa sheets mounted in Ussing chambers or isolated bladder strips in organ baths) in that it enabled direct access to the vicinity of SubU/LP during authentic bladder filling. The model is particularly suitable for understanding local mechanisms of urothelium-DSM connectivity and for broad understanding of the role of urothelium in regulating continence and voiding.
Collapse
Affiliation(s)
- Leonie Durnin
- Department of Physiology and Cell Biology, University of Nevada Reno School of Medicine, Reno, NV, 89557-0575, USA
| | - Benjamin Kwok
- Department of Physiology and Cell Biology, University of Nevada Reno School of Medicine, Reno, NV, 89557-0575, USA
| | - Priya Kukadia
- Department of Physiology and Cell Biology, University of Nevada Reno School of Medicine, Reno, NV, 89557-0575, USA
| | - Roisin McAvera
- Department of Physiology and Cell Biology, University of Nevada Reno School of Medicine, Reno, NV, 89557-0575, USA
| | - Robert D Corrigan
- Department of Physiology and Cell Biology, University of Nevada Reno School of Medicine, Reno, NV, 89557-0575, USA
| | - Sean M Ward
- Department of Physiology and Cell Biology, University of Nevada Reno School of Medicine, Reno, NV, 89557-0575, USA
| | - Ying Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Qi Chen
- Department of Physiology and Cell Biology, University of Nevada Reno School of Medicine, Reno, NV, 89557-0575, USA
| | - Sang Don Koh
- Department of Physiology and Cell Biology, University of Nevada Reno School of Medicine, Reno, NV, 89557-0575, USA
| | - Kenton M Sanders
- Department of Physiology and Cell Biology, University of Nevada Reno School of Medicine, Reno, NV, 89557-0575, USA
| | | |
Collapse
|
24
|
Giglio D, Podmolíková L, Tobin G. Changes in the Neuronal Control of the Urinary Bladder in a Model of Radiation Cystitis. J Pharmacol Exp Ther 2018; 365:327-335. [PMID: 29530925 DOI: 10.1124/jpet.117.246371] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 02/20/2018] [Indexed: 03/08/2025] Open
Abstract
Currently, we have assessed the neuronal control of the urinary bladder in radiation cystitis and whether interstitial cells contribute to the condition. Fourteen days after bladder irradiation (20 Gy), rats were sedated and the urinary bladder was cut into two sagittal halves where electrical field stimulation (EFS; 5-20 Hz) was applied on the pelvic nerve afferents or stretch (80 mN) on one-half of the bladder, while contractions were registered on the contralateral half of the bladder in the absence and presence of increasing doses of imatinib (1-10 mg/kg; inhibitor of c-kit-positive interstitial cells), atropine (1 mg/kg; to block muscarinic M3 receptors), or pyridoxalphosphate-6-azophenyl-2',4'-disulfonic acid (2 mg/kg; P2X1 purinoceptor antagonist). Urinary bladders were also excised for organ bath experiments, Western blot, quantitative polymerase chain reaction, and immunohistochemistry. In vivo, EFS contractions were enhanced after irradiation, and imatinib (1-10 mg/mg) inhibited contractions by EFS and stretched dose-dependently in controls but not in irradiated bladders. In the irradiated bladder in vitro, atropine resistance was increased and imatinib (100 µM) inhibited contractions by EFS and agonists (ATP, methacholine) in irradiated bladders and controls. The urinary bladder expressions of P2X1 purinoceptors, muscarinic M3 receptor, choline acetyltransferase, c-kit, and the agonist of c-kit, stem cell factor, were not changed by irradiation. In conclusion, bladder irradiation affects several levels of neuronal control of the urinary bladder. Interstitial cells may contribute to some of the symptoms associated with radiation cystitis.
Collapse
Affiliation(s)
- Daniel Giglio
- Department of Pharmacology, Institution of Neuroscience and Physiology (D.G., L.P., G.T.) and Department of Oncology, Institution of Clinical Sciences, University of Gothenburg, Gothenburg, Sweden (D.G.); and Department of Medical Biochemistry, Faculty of Medicine, Charles University, Hradec Králové, Czech Republic (L.P.)
| | - Lucie Podmolíková
- Department of Pharmacology, Institution of Neuroscience and Physiology (D.G., L.P., G.T.) and Department of Oncology, Institution of Clinical Sciences, University of Gothenburg, Gothenburg, Sweden (D.G.); and Department of Medical Biochemistry, Faculty of Medicine, Charles University, Hradec Králové, Czech Republic (L.P.)
| | - Gunnar Tobin
- Department of Pharmacology, Institution of Neuroscience and Physiology (D.G., L.P., G.T.) and Department of Oncology, Institution of Clinical Sciences, University of Gothenburg, Gothenburg, Sweden (D.G.); and Department of Medical Biochemistry, Faculty of Medicine, Charles University, Hradec Králové, Czech Republic (L.P.)
| |
Collapse
|
25
|
Comparative immunohistochemical characterization of interstitial cells in the urinary bladder of human, guinea pig and pig. Histochem Cell Biol 2018; 149:491-501. [DOI: 10.1007/s00418-018-1655-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/15/2018] [Indexed: 01/20/2023]
|