1
|
Iguchi N, Teimouri A, Wilcox DT, Malykhina AP, Cost NG. Lower urinary dysfunction as a long-term effect of childhood vincristine treatment, with potential influences by sex and dose. Sci Rep 2024; 14:15049. [PMID: 38951167 PMCID: PMC11217273 DOI: 10.1038/s41598-024-65313-9] [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: 03/05/2024] [Accepted: 06/19/2024] [Indexed: 07/03/2024] Open
Abstract
Vincristine (VCR) is one of the most widely used chemotherapy agents in treating pediatric cancer. Nonetheless, it is known to cause dose-dependent neurotoxicity which can impact virtually every organ system. Despite its widespread use, the precise impact of VCR on the lower urinary tract (LUT) remains inadequately elucidated. Our initial clinical and translational investigations suggest a sex-specific influence of childhood VCR exposure on LUT function. Thus, the current study aimed to investigate the late effects of systemic VCR exposure on LUT physiology and the underlying mechanisms, focusing on dosage and male-sex, employing juvenile CD-1 mice as a model. Male mice subjected to VCR exhibited augmented functional bladder capacity accompanied by frequent non-void contractions during awake cystometry, alongside mast cell accumulation within the bladder, compared to the saline-treated control group. Noteworthy functional changes were observed in bladder strips from the VCR group, including decreased nerve-mediated contraction, heightened contractile responses to cholinergic and purinergic agonists, enhanced responsiveness to histamine-primarily via histamine receptor 1 (Hrh1)-and an augmented relaxation effect with compound 48/80 (a mast cell degranulator), relative to the control group. Significant changes in gene expression levels associated with neuroinflammation and nociception were observed in both the bladder and lumbosacral dorsal root ganglia (Ls-DRG) of the VCR group. These findings suggest that VCR exposure during childhood, particularly in males, triggers neuroimmune responses in the bladder and Ls-DRG, amplifying responsiveness to neurotransmitters in the bladder, thereby contributing to LUT dysfunction characterized by a mixed bladder phenotype as a late effect during survivorship.
Collapse
Affiliation(s)
- Nao Iguchi
- Division of Urology, Department of Surgery, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Ali Teimouri
- Division of Urology, Department of Surgery, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Duncan T Wilcox
- Division of Urology, Department of Surgery, University of Colorado School of Medicine, Aurora, CO, 80045, USA
- Department of Urology, Children's Hospital Colorado, 13123 E. 16th Avenue, Aurora, CO, 80045, USA
| | - Anna P Malykhina
- Division of Urology, Department of Surgery, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Nicholas G Cost
- Division of Urology, Department of Surgery, University of Colorado School of Medicine, Aurora, CO, 80045, USA.
- Department of Urology, Children's Hospital Colorado, 13123 E. 16th Avenue, Aurora, CO, 80045, USA.
- The Surgical Oncology Program, Children's Hospital Colorado, Aurora, CO, 80045, USA.
| |
Collapse
|
2
|
Xie AX, Iguchi N, Malykhina AP. Long-term follow-up of TREK-1 KO mice reveals the development of bladder hypertrophy and impaired bladder smooth muscle contractility with age. Am J Physiol Renal Physiol 2024; 326:F957-F970. [PMID: 38601986 PMCID: PMC11386977 DOI: 10.1152/ajprenal.00382.2023] [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: 11/30/2023] [Revised: 03/04/2024] [Accepted: 03/27/2024] [Indexed: 04/12/2024] Open
Abstract
Stretch-activated two-pore domain K+ (K2P) channels play important roles in many visceral organs, including the urinary bladder. The TWIK-related K+ channel TREK-1 is the predominantly expressed K2P channel in the urinary bladder of humans and rodents. Downregulation of TREK-1 channels was observed in the urinary bladder of patients with detrusor overactivity, suggesting their involvement in the pathogenesis of voiding dysfunction. This study aimed to characterize the long-term effects of TREK-1 on bladder function with global and smooth muscle-specific TREK-1 knockout (KO) mice. Bladder morphology, bladder smooth muscle (BSM) contractility, and voiding patterns were evaluated up to 12 mo of age. Both sexes were included in this study to probe the potential sex differences. Smooth muscle-specific TREK-1 KO mice were used to distinguish the effects of TREK-1 downregulation in BSM from the neural pathways involved in the control of bladder contraction and relaxation. TREK-1 KO mice developed enlarged urinary bladders (by 60.0% for males and by 45.1% for females at 6 mo; P < 0.001 compared with the age-matched control group) and had a significantly increased bladder capacity (by 137.7% at 12 mo; P < 0.0001) and compliance (by 73.4% at 12 mo; P < 0.0001). Bladder strips isolated from TREK-1 KO mice exhibited decreased contractility (peak force after KCl at 6 mo was 1.6 ± 0.7 N/g compared with 3.4 ± 2.0 N/g in the control group; P = 0.0005). The lack of TREK-1 channels exclusively in BSM did not replicate the bladder phenotype observed in TREK-1 KO mice, suggesting a strong neurogenic origin of TREK-1-related bladder dysfunction.NEW & NOTEWORTHY This study compared voiding function and bladder phenotypes in global and smooth muscle-specific TREK-1 KO mice. We found significant age-related changes in bladder contractility, suggesting that the lack of TREK-1 channel activity might contribute to age-related changes in bladder smooth muscle physiology.
Collapse
Affiliation(s)
- Alison Xiaoqiao Xie
- Division of Urology, Department of SurgeryUniversity of Colorado Anschutz Medical CampusAuroraColoradoUnited States
| | - Nao Iguchi
- Division of Urology, Department of SurgeryUniversity of Colorado Anschutz Medical CampusAuroraColoradoUnited States
| | - Anna P Malykhina
- Division of Urology, Department of SurgeryUniversity of Colorado Anschutz Medical CampusAuroraColoradoUnited States
| |
Collapse
|
3
|
Shimizu N, Saito T, Wada N, Hashimoto M, Shimizu T, Kwon J, Cho KJ, Saito M, Karnup S, de Groat WC, Yoshimura N. Molecular Mechanisms of Neurogenic Lower Urinary Tract Dysfunction after Spinal Cord Injury. Int J Mol Sci 2023; 24:7885. [PMID: 37175592 PMCID: PMC10177842 DOI: 10.3390/ijms24097885] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 04/21/2023] [Accepted: 04/22/2023] [Indexed: 05/15/2023] Open
Abstract
This article provides a synopsis of current progress made in fundamental studies of lower urinary tract dysfunction (LUTD) after spinal cord injury (SCI) above the sacral level. Animal models of SCI allowed us to examine the effects of SCI on the micturition control and the underlying neurophysiological processes of SCI-induced LUTD. Urine storage and elimination are the two primary functions of the LUT, which are governed by complicated regulatory mechanisms in the central and peripheral nervous systems. These neural systems control the action of two functional units in the LUT: the urinary bladder and an outlet consisting of the bladder neck, urethral sphincters, and pelvic-floor striated muscles. During the storage phase, the outlet is closed, and the bladder is inactive to maintain a low intravenous pressure and continence. In contrast, during the voiding phase, the outlet relaxes, and the bladder contracts to facilitate adequate urine flow and bladder emptying. SCI disrupts the normal reflex circuits that regulate co-ordinated bladder and urethral sphincter function, leading to involuntary and inefficient voiding. Following SCI, a spinal micturition reflex pathway develops to induce an overactive bladder condition following the initial areflexic phase. In addition, without proper bladder-urethral-sphincter coordination after SCI, the bladder is not emptied as effectively as in the normal condition. Previous studies using animal models of SCI have shown that hyperexcitability of C-fiber bladder afferent pathways is a fundamental pathophysiological mechanism, inducing neurogenic LUTD, especially detrusor overactivity during the storage phase. SCI also induces neurogenic LUTD during the voiding phase, known as detrusor sphincter dyssynergia, likely due to hyperexcitability of Aδ-fiber bladder afferent pathways rather than C-fiber afferents. The molecular mechanisms underlying SCI-induced LUTD are multifactorial; previous studies have identified significant changes in the expression of various molecules in the peripheral organs and afferent nerves projecting to the spinal cord, including growth factors, ion channels, receptors and neurotransmitters. These findings in animal models of SCI and neurogenic LUTD should increase our understanding of pathophysiological mechanisms of LUTD after SCI for the future development of novel therapies for SCI patients with LUTD.
Collapse
Affiliation(s)
- Nobutaka Shimizu
- Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (N.S.)
- Pelvic Floor Center, Kochi Medical School, Kochi University, Nankoku 783-8505, Japan
| | - Tetsuichi Saito
- Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (N.S.)
| | - Naoki Wada
- Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (N.S.)
| | - Mamoru Hashimoto
- Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (N.S.)
| | - Takahiro Shimizu
- Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (N.S.)
- Department of Pharmacology, Kochi Medical School, Kochi University, Nankoku 783-8505, Japan
| | - Joonbeom Kwon
- Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (N.S.)
| | - Kang Jun Cho
- Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (N.S.)
| | - Motoaki Saito
- Department of Pharmacology, Kochi Medical School, Kochi University, Nankoku 783-8505, Japan
| | - Sergei Karnup
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - William C. de Groat
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Naoki Yoshimura
- Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (N.S.)
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| |
Collapse
|
4
|
Iguchi N, Dönmez Mİ, Malykhina AP, Wilcox DT. Anti-fibrotic effect of tocotrienols for bladder dysfunction due to partial bladder outlet obstruction. Investig Clin Urol 2023; 64:189-196. [PMID: 36882179 PMCID: PMC9995959 DOI: 10.4111/icu.20220328] [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: 09/28/2022] [Revised: 12/20/2022] [Accepted: 01/11/2023] [Indexed: 02/16/2023] Open
Abstract
PURPOSE To investigate potential beneficial effects of tocotrienols which have been suggested to inhibit hypoxia-inducible factor (HIF) pathway, on partial bladder outlet obstruction (PBOO)-induced bladder pathology. MATERIALS AND METHODS PBOO was surgically created in juvenile male mice. Sham-operated mice were used as controls. Animals received daily oral administration of either tocotrienols (T3) or soybean oil (SBO, vehicle) from day 0 to 13 post-surgery. Bladder function was examined in vivo by void spot assay. At 2 weeks post-surgery, the bladders were subjected to physiological evaluation of detrusor contractility in vitro using bladder strips, histology by H&E staining and collagen imaging, and gene expression analyses by quantitative PCR. RESULTS A significant increase in the number of small voids was observed after 1 week of PBOO compared to the control groups. At 2 weeks post-surgery, PBOO+SBO mice showed a further increase in the number of small voids, which was not observed in PBOO+T3 group. PBOO-induced decrease in detrusor contractility was similar between two treatments. PBOO induced bladder hypertrophy to the same degree in both SBO and T3 treatment groups, however, fibrosis in the bladder was significantly less prominent in the T3 group than the SBO group following PBOO (1.8- vs. 3.0-fold increase in collagen content compared to the control). Enhanced levels of HIF target genes in the bladders were observed in PBOO+SBO group, but not in PBOO+T3 group compared to the control. CONCLUSIONS Oral tocotrienol treatment reduced the progression of urinary frequency and bladder fibrosis by suppressing HIF pathways triggered by PBOO.
Collapse
Affiliation(s)
- Nao Iguchi
- Division of Urology, Department of Surgery, University of Colorado Denver School of Medicine, Aurora, CO, USA
| | - M İrfan Dönmez
- Division of Urology, Department of Surgery, University of Colorado Denver School of Medicine, Aurora, CO, USA.,Division of Pediatric Urology, Department of Urology, Istanbul University Istanbul Faculty of Medicine, Istanbul, Turkey
| | - Anna P Malykhina
- Division of Urology, Department of Surgery, University of Colorado Denver School of Medicine, Aurora, CO, USA
| | - Duncan T Wilcox
- Division of Urology, Department of Surgery, University of Colorado Denver School of Medicine, Aurora, CO, USA.,Department of Pediatric Urology, Children's Hospital Colorado, Aurora, CO, USA.
| |
Collapse
|
5
|
Clayton DB, Tong CMC, Li B, Taylor AS, De S, Mason MD, Dudley AG, Davidoff O, Kobayashi H, Haase VH. Inhibition of hypoxia-inducible factor-prolyl hydroxylation protects from cyclophosphamide-induced bladder injury and urinary dysfunction. Am J Physiol Renal Physiol 2022; 323:F81-F91. [PMID: 35499237 PMCID: PMC9236868 DOI: 10.1152/ajprenal.00344.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 04/25/2022] [Accepted: 04/25/2022] [Indexed: 11/22/2022] Open
Abstract
Disruption of the blood-urine barrier can result in acute or chronic inflammatory bladder injury. Activation of the oxygen-regulated hypoxia-inducible factor (HIF) pathway has been shown to protect mucosal membranes by increasing the expression of cytoprotective genes and by suppressing inflammation. The activity of HIF is controlled by prolyl hydroxylase domain (PHD) dioxygenases, which have been exploited as therapeutic targets for the treatment of anemia of chronic kidney disease. Here, we established a mouse model of acute cyclophosphamide (CYP)-induced blood-urine barrier disruption associated with inflammation and severe urinary dysfunction to investigate the HIF-PHD axis in inflammatory bladder injury. We found that systemic administration of dimethyloxalylglycine or molidustat, two small-molecule inhibitors of HIF-prolyl hydroxylases, profoundly mitigated CYP-induced bladder injury and inflammation as assessed by morphological analysis of transmural edema and urothelial integrity and by measuring tissue cytokine expression. Void spot analysis to examine bladder function quantitatively demonstrated that HIF-prolyl hydroxylase inhibitor administration normalized micturition patterns and protected against CYP-induced alteration of urinary frequency and micturition patterns. Our study highlights the therapeutic potential of HIF-activating small-molecule compounds for the prevention or therapy of bladder injury and urinary dysfunction due to blood-urine barrier disruption.NEW & NOTEWORTHY Disruption of the blood-urine barrier can result in acute or chronic inflammatory bladder injury. Here, we demonstrate that pharmacological inhibition of hypoxia-inducible factor (HIF)-prolyl hydroxylation prevented bladder injury and protected from urinary dysfunction in a mouse model of cyclophosphamide-induced disruption of the blood-urine barrier. Our study highlights a potential role for HIF-activating small-molecule compounds in the prevention or therapy of bladder injury and urinary dysfunction and provides a rationale for future clinical studies.
Collapse
Affiliation(s)
- Douglass B Clayton
- Division of Pediatric Urology, Department of Urology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Ching Man Carmen Tong
- Division of Pediatric Urology, Department of Urology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Belinda Li
- Division of Pediatric Urology, Department of Urology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Abby S Taylor
- Division of Pediatric Urology, Department of Urology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Shuvro De
- Division of Pediatric Urology, Department of Urology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Matthew D Mason
- Division of Pediatric Urology, Department of Urology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Anne G Dudley
- Division of Pediatric Urology, Department of Urology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Olena Davidoff
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Medical and Research Services, Department of Veterans Affairs Hospital, Tennessee Valley Healthcare System, Nashville, Tennessee
| | - Hanako Kobayashi
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Medical and Research Services, Department of Veterans Affairs Hospital, Tennessee Valley Healthcare System, Nashville, Tennessee
| | - Volker H Haase
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Medical and Research Services, Department of Veterans Affairs Hospital, Tennessee Valley Healthcare System, Nashville, Tennessee
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee
| |
Collapse
|
6
|
Sexual dimorphic impacts of systemic vincristine on lower urinary tract function. Sci Rep 2022; 12:5113. [PMID: 35332157 PMCID: PMC8948262 DOI: 10.1038/s41598-022-08585-3] [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: 11/11/2021] [Accepted: 03/09/2022] [Indexed: 11/20/2022] Open
Abstract
Vincristine (VCR) is one of the most common chemotherapy agents used in pediatric oncology. Despite the well-known VCR-induced peripheral neuropathy, potential impacts of VCR on lower urinary tract (LUT) function remain poorly defined. We investigated the effects of systemic VCR exposure in childhood on LUT function by using juvenile mice treated with VCR (4 mg/kg) or saline and evaluated at 5 weeks later. VCR induced a decreased urinary frequency with increased functional bladder capacity and non-void contractions. There were no changes in detrusor contractility between the groups. VCR exposure caused sexual dimorphic changes; in females, increased intravesical pressure at micturition and downregulations of a major player in bladder afferent firing, Htr3b, in the bladders, and Cav1.2 in the lumbosacral dorsal root ganglia (Ls-DRG), while male mice displayed increases in bladder compliance and detrusor activity, upregulations of IL-2, Trpa1 and Itga1 in the bladders and neuroinflammation-related genes, P2×4, P2×7, IL-2 and CD68 in the Ls-DRG. These results suggest that that systemic VCR exposure caused sensory neuropathy via sex-dimorphic mechanisms, leading to altered LUT function. These changes might clinically present as gender-specific signs or symptoms of LUT dysfunction, and follow-up urological assessment may be of benefit for pediatric cancer patients treated with VCR.
Collapse
|
7
|
Xie AX, Iguchi N, Clarkson TC, Malykhina AP. Pharmacogenetic inhibition of lumbosacral sensory neurons alleviates visceral hypersensitivity in a mouse model of chronic pelvic pain. PLoS One 2022; 17:e0262769. [PMID: 35077502 PMCID: PMC8789164 DOI: 10.1371/journal.pone.0262769] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 01/04/2022] [Indexed: 12/12/2022] Open
Abstract
The study investigated the cellular and molecular mechanisms in the peripheral nervous system (PNS) underlying the symptoms of urologic chronic pelvic pain syndrome (UCPPS) in mice. This work also aimed to test the feasibility of reversing peripheral sensitization in vivo in alleviating UCPPS symptoms. Intravesical instillation of vascular endothelial growth factor A (VEGFA) was used to induce UCPPS-like symptoms in mice. Spontaneous voiding spot assays and manual Von Frey tests were used to evaluate the severity of lower urinary tract symptoms (LUTS) and visceral hypersensitivity in VEGFA-instilled mice. Bladder smooth muscle strip contractility recordings (BSMSC) were used to identify the potential changes in myogenic and neurogenic detrusor muscle contractility at the tissue-level. Quantitative real-time PCR (qPCR) and fluorescent immunohistochemistry were performed to compare the expression levels of VEGF receptors and nociceptors in lumbosacral dorsal root ganglia (DRG) between VEGFA-instilled mice and saline-instilled controls. To manipulate primary afferent activity, Gi-coupled Designer Receptors Exclusively Activated by Designer Drugs (Gi-DREADD) were expressed in lumbosacral DRG neurons of TRPV1-Cre-ZGreen mice via targeted adeno-associated viral vector (AAVs) injections. A small molecule agonist of Gi-DREADD, clozapine-N-oxide (CNO), was injected into the peritoneum (i. p.) in awake animals to silence TRPV1 expressing sensory neurons in vivo during physiological and behavioral recordings of bladder function. Intravesical instillation of VEGFA in the urinary bladders increased visceral mechanical sensitivity and enhanced RTX-sensitive detrusor contractility. Sex differences were identified in the baseline detrusor contractility responses and VEGF-induced visceral hypersensitivity. VEGFA instillations in the urinary bladder led to significant increases in the mRNA and protein expression of transient receptor potential cation channel subfamily A member 1 (TRPA1) in lumbosacral DRG, whereas the expression levels of transient receptor potential cation channel subfamily V member 1 (TRPV1) and VEGF receptors (VEGFR1 and VEGFR2) remained unchanged when compared to saline-instilled animals. Importantly, the VEGFA-induced visceral hypersensitivity was reversed by Gi-DREADD-mediated neuronal silencing in lumbosacral sensory neurons. Activation of bladder VEGF signaling causes sensory neural plasticity and visceral hypersensitivity in mice, confirming its role of an UCPPS biomarker as identified by the Multidisciplinary Approach to the Study of Chronic Pelvic Pain (MAPP) research studies. Pharmacogenetic inhibition of lumbosacral sensory neurons in vivo completely reversed VEGFA-induced pelvic hypersensitivity in mice, suggesting the strong therapeutic potential for decreasing primary afferent activity in the treatment of pain severity in UCPPS patients.
Collapse
Affiliation(s)
- Alison Xiaoqiao Xie
- Department of Surgery, School of Medicine, Anschutz Medical Campus, University of Colorado, Denver, Colorado, United States of America
| | - Nao Iguchi
- Department of Surgery, School of Medicine, Anschutz Medical Campus, University of Colorado, Denver, Colorado, United States of America
| | - Taylor C. Clarkson
- Department of Surgery, School of Medicine, Anschutz Medical Campus, University of Colorado, Denver, Colorado, United States of America
| | - Anna P. Malykhina
- Department of Surgery, School of Medicine, Anschutz Medical Campus, University of Colorado, Denver, Colorado, United States of America
| |
Collapse
|
8
|
Wada N, Karnup S, Kadekawa K, Shimizu N, Kwon J, Shimizu T, Gotoh D, Kakizaki H, de Groat W, Yoshimura N. Current knowledge and novel frontiers in lower urinary tract dysfunction after spinal cord injury: Basic research perspectives. UROLOGICAL SCIENCE 2022; 33:101-113. [PMID: 36177249 PMCID: PMC9518811 DOI: 10.4103/uros.uros_31_22] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
This review article aims to summarize the recent advancement in basic research on lower urinary tract dysfunction (LUTD) following spinal cord injury (SCI) above the sacral level. We particularly focused on the neurophysiologic mechanisms controlling the lower urinary tract (LUT) function and the SCI-induced changes in micturition control in animal models of SCI. The LUT has two main functions, the storage and voiding of urine, that are regulated by a complex neural control system. This neural system coordinates the activity of two functional units in the LUT: the urinary bladder and an outlet including bladder neck, urethra, and striated muscles of the pelvic floor. During the storage phase, the outlet is closed and the bladder is quiescent to maintain a low intravesical pressure and continence, and during the voiding phase, the outlet relaxes and the bladder contracts to promote efficient release of urine. SCI impairs voluntary control of voiding as well as the normal reflex pathways that coordinate bladder and sphincter function. Following SCI, the bladder is initially areflexic but then becomes hyperreflexic due to the emergence of a spinal micturition reflex pathway. However, the bladder does not empty efficiently because coordination between the bladder and urethral sphincter is lost. In animal models of SCI, hyperexcitability of silent C-fiber bladder afferents is a major pathophysiological basis of neurogenic LUTD, especially detrusor overactivity. Reflex plasticity is associated with changes in the properties of neuropeptides, neurotrophic factors, or chemical receptors of afferent neurons. Not only C-fiber but also Aδ-fiber could be involved in the emergence of neurogenic LUTD such as detrusor sphincter dyssynergia following SCI. Animal research using disease models helps us to detect the different contributing factors for LUTD due to SCI and to find potential targets for new treatments.
Collapse
|
9
|
Li T, Xing Y, Zhang G, Wang Y, Wei Y, Cui L, Zhang S, Wang Q. Circular RNA Plasmacytoma Variant Translocation 1 (CircPVT1) knockdown ameliorates hypoxia-induced bladder fibrosis by regulating the miR-203/Suppressor of Cytokine Signaling 3 (SOCS3) signaling axis. Bioengineered 2022; 13:1288-1303. [PMID: 35000524 PMCID: PMC8805914 DOI: 10.1080/21655979.2021.2001221] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 10/29/2021] [Indexed: 11/02/2022] Open
Abstract
The effects of circular RNAs (circRNAs) on bladder outlet obstruction (BOO)-induced hypertrophy and fibrogenesis in rats and hypoxia-induced bladder smooth muscle cell (BSMC) fibrosis remain unclear. This study aimed to determine the regulatory role of circRNAs in the phenotypic changes in BSMCs in BOO-induced rats.circRNAmicroarray and real-time PCR were used to explore differentiated expressed circRNAs. Bioinformatics analyses and dual-luciferase reporter were performed to identify the targets for circRNA PVT1 (circPVT1). BOO was performed to establish a bladder fibrosis animal model. The circPVT1 and suppressor of cytokine signaling 3 (SOCS3) expression levels were upregulated (p = 0.0061 and 0.0328, respectively), whereas the microRNA-203a (miR-203) level was downregulated in rats with bladder remodeling (p=0.0085). Bioinformatics analyses and dual-luciferase reporter assay results confirmed that circPVT1 sponges miR-203 and that the latter targets the 3'-untranslated region of SOCS3. Additionally, circPVT1 knockdown alleviated BOO-induced bladder hypertrophy and fibrogenesis. Furthermore, hypoxia was induced in BSMCs to establish a cell model of bladder fibrosis. Hypoxia induction in BSMCs resulted in upregulated circPVT1 and SOCS3 levels (p = 0.0052) and downregulated miR-203 levels. Transfection with circPVT1 and SOCS3 shRNA ameliorated hypoxia-induced transforming growth factor-β (TGF-β1), TGFβR1, α-smooth muscle actin, fibrotic growth factor, extracellular matrix subtypes, BSMC proliferation, and apoptosis-associated cell injury, whereas co-transfection with miR-203 inhibitor counteracted the effect of circPVT1 shRNA on these phenotypes.These findings revealed a novel circRNA regulator of BOO-associated bladder wall remodeling and hypoxia-induced phenotypic changes in BMSCs by targeting the miR-203-SOCS3 signaling axis.
Collapse
Affiliation(s)
- Teng Li
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yi Xing
- Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Guoxian Zhang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yan Wang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yinsheng Wei
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lingang Cui
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shaojin Zhang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qingwei Wang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
10
|
Differentially Expressed Genes Correlated with Fibrosis in a Rat Model of Chronic Partial Bladder Outlet Obstruction. Processes (Basel) 2021. [DOI: 10.3390/pr9122219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Chronic partial bladder outlet obstruction (PBOO) is a prevalent clinical problem that may result from multiple etiologies. PBOO may be a secondary condition to various anatomical and functional abnormalities. Bladder fibrosis is the worst outcome of PBOO. However, gene alterations and the mechanism of fibrosis development after PBOO onset are not clear. Therefore, we aimed to investigate gene expression alterations during chronic PBOO. A rat model of PBOO was established and validated by a significant increase in rat bladder weight. The bladder samples were further analyzed by microarray, and differentially expressed genes (DEGs) that are more related to PBOO compared with the control genes were selected. The data showed that 16 significantly upregulated mRNAs and 3 significantly downregulated mRNAs are involved in fibrosis. Moreover, 13 significantly upregulated mRNAs and 12 significantly downregulated mRNAs are related to TGFB signaling. Twenty-two significantly upregulated mRNAs and nine significantly downregulated mRNAs are related to the extracellular matrix. The genes with differential expressions greater than four-fold included Grem1, Thbs1, Col8a1, Itga5, Tnc, Lox, Timp1, Col4a1, Col4a2, Bhlhe40, Itga1, Tgfb3, and Gadd45b. The gene with a differential expression less than a quarter-fold was Thbs2. These findings show the potential roles of these genes in the physiology of PBOO.
Collapse
|
11
|
Dalghi MG, Ruiz WG, Clayton DR, Montalbetti N, Daugherty SL, Beckel JM, Carattino MD, Apodaca G. Functional roles for PIEZO1 and PIEZO2 in urothelial mechanotransduction and lower urinary tract interoception. JCI Insight 2021; 6:e152984. [PMID: 34464353 PMCID: PMC8525643 DOI: 10.1172/jci.insight.152984] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 08/12/2021] [Indexed: 11/17/2022] Open
Abstract
The mechanisms that link visceral mechanosensation to the perception of internal organ status (i.e., interoception) remain elusive. In response to bladder filling, the urothelium releases ATP, which is hypothesized to stimulate voiding function by communicating the degree of bladder fullness to subjacent tissues, including afferent nerve fibers. To determine if PIEZO channels function as mechanosensors in these events, we generated conditional urothelial Piezo1-, Piezo2-, and dual Piezo1/2-knockout (KO) mice. While functional PIEZO1 channels were expressed in all urothelial cell layers, Piezo1-KO mice had a limited phenotype. Piezo2 expression was limited to a small subset of superficial umbrella cells, yet male Piezo2-KO mice exhibited incontinence (i.e., leakage) when their voiding behavior was monitored during their active dark phase. Dual Piezo1/2-KO mice had the most affected phenotype, characterized by decreased urothelial responses to mechanical stimulation, diminished ATP release, bladder hypoactivity in anesthetized Piezo1/2-KO females but not males, and urinary incontinence in both male and female Piezo1/2-KO mice during their dark phase but not inactive light one. Our studies reveal that the urothelium functions in a sex- and circadian rhythm–dependent manner to link urothelial PIEZO1/2 channel–driven mechanotransduction to normal voiding function and behavior, and in the absence of these signals, bladder dysfunction ensues.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Marcelo D Carattino
- Department of Medicine.,Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Gerard Apodaca
- Department of Medicine.,Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
12
|
Kwon J, Lee EJ, Cho HJ, Jang JA, Han MS, Kwak E, Kim H, An J, Park D, Han S, Shimizu N, Suzuki T, Takaoka EI, Yoshimura N. Antifibrosis treatment by inhibition of VEGF, FGF, and PDGF receptors improves bladder wall remodeling and detrusor overactivity in association with modulation of C-fiber afferent activity in mice with spinal cord injury. Neurourol Urodyn 2021; 40:1460-1469. [PMID: 34015154 DOI: 10.1002/nau.24704] [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: 01/22/2021] [Revised: 04/28/2021] [Accepted: 05/03/2021] [Indexed: 11/08/2022]
Abstract
AIMS Spinal cord injury (SCI) above the sacral level causes bladder dysfunction and remodeling with fibrosis. This study examined the antifibrotic effects using nintedanib, an inhibitor of vascular endothelial growth factor, fibroblast growth factor, and platelet-derived growth factor receptors, on detrusor overactivity (DO) and bladder fibrosis, as well as the modulation mechanisms of C-fiber afferent pathways. METHODS Thirty female C57BL/6 mice were divided into group A (spinal intact), group B (SCI with vehicle), and group C (SCI with nintedanib). At 2 weeks after SCI, vehicle or 50 mg/kg nintedanib was administered subcutaneously for 2 weeks. Then, cystometry was conducted, followed by RT-PCR measurements of fibrosis-related molecules, muscarinic, β-adrenergic, TRP and purinergic receptors in the bladder or L6-S1 dorsal root ganglia (DRG). Trichrome stain and Western blot analysis of transforming growth factor-beta and fibronectin were performed in the bladder. TRPV1 expression in L6 DRG was measured by immunohistochemistry. RESULTS In cystometry, intercontraction intervals, nonvoiding contractions, voided volume, and voiding efficiency were significantly improved in group C versus group B. RT-PCR, Western blotting, and trichrome staining revealed the fibrotic changes in the bladder of group B, which was improved in group C. Increased messenger RNA levels of TRPV1, TRPA1, P2X2 , and P2X3 in DRG of group B were significantly decreased in group C. TRPV1 immunoreactivity in DRG was increased in group B, but decreased in group C. CONCLUSIONS Nintedanib improves storage and voiding dysfunctions and bladder fibrosis in SCI mice. Also, nintedanib-induced improvement of DO is associated with reduced expression of C-fiber afferent markers, suggesting the modulation of bladder C-fiber afferent activity.
Collapse
Affiliation(s)
- Joonbeom Kwon
- Department of Urology, Daegu Fatima Hospital, Daegu, South Korea.,Research Institute, Daegu Fatima Hospital, Daegu, South Korea.,Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Eun-Ju Lee
- Research Institute, Daegu Fatima Hospital, Daegu, South Korea
| | - Hyun-Jung Cho
- Research Institute, Daegu Fatima Hospital, Daegu, South Korea
| | - Ji-Ae Jang
- Research Institute, Daegu Fatima Hospital, Daegu, South Korea
| | - Min-Su Han
- Research Institute, Daegu Fatima Hospital, Daegu, South Korea
| | - Eunkyoung Kwak
- Department of Pathology, Daegu Fatima Hospital, Daegu, South Korea
| | - Haesoo Kim
- Department of Anesthesiology, Daegu Fatima Hospital, Daegu, South Korea
| | - Jihyun An
- Department of Anesthesiology, Daegu Fatima Hospital, Daegu, South Korea
| | - Donghwi Park
- Department of Physical Medicine and Rehabilitation, University of Ulsan College of Medicine, Ulsan, South Korea
| | - Seungwoo Han
- Department of Internal Medicine, Kyungpook National University School of Medicine, Daegu, South Korea
| | - Nobutaka Shimizu
- Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Takahisa Suzuki
- Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Ei-Ichiro Takaoka
- Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Naoki Yoshimura
- Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
13
|
Iguchi N, Carrasco A, Xie AX, Pineda RH, Malykhina AP, Wilcox DT. Functional constipation induces bladder overactivity associated with upregulations of Htr2 and Trpv2 pathways. Sci Rep 2021; 11:1149. [PMID: 33441874 PMCID: PMC7806916 DOI: 10.1038/s41598-020-80794-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 12/23/2020] [Indexed: 01/23/2023] Open
Abstract
Bladder and bowel dysfunction (BBD) is a common yet underdiagnosed paediatric entity that describes lower urinary tract symptoms (LUTS) accompanied by abnormal bowel patterns manifested as constipation and/or encopresis. LUTS usually manifest as urgency, urinary frequency, incontinence, and urinary tract infections (UTI). Despite increasing recognition of BBD as a risk factor for long-term urinary tract problems including recurrent UTI, vesicoureteral reflux, and renal scarring, the mechanisms underlying BBD have been unclear, and treatment remains empirical. We investigated how constipation affects the lower urinary tract function using a juvenile murine model of functional constipation. Following four days of functional constipation, animals developed LUTS including urinary frequency and detrusor overactivity evaluated by awake cystometry. Physiological examination of detrusor function in vitro using isolated bladder strips, demonstrated a significant increase in spontaneous contractions without affecting contractile force in response to electrical field stimulation, carbachol, and KCl. A significant upregulation of serotonin receptors, Htr2a and Htr2c, was observed in the bladders from mice with constipation, paralleled with augmented spontaneous contractions after pre-incubation of the bladder strips with 0.5 µM of serotonin. These results suggest that constipation induced detrusor overactivity and increased excitatory serotonin receptor activation in the urinary bladder, which contributes to the development of BBD.
Collapse
Affiliation(s)
- Nao Iguchi
- Division of Urology, Department of Surgery, University of Colorado Denver School of Medicine, 12700 E 19th Avenue, Aurora, CO, 80045, USA
| | - Alonso Carrasco
- Children's Hospital Colorado, 13123 E 16th Avenue, B463, Aurora, CO, 80045, USA
- Children's Mercy Kansas City, 2401 Gillham Rd, Kansas City, MO, 64108, USA
| | - Alison X Xie
- Division of Urology, Department of Surgery, University of Colorado Denver School of Medicine, 12700 E 19th Avenue, Aurora, CO, 80045, USA
| | - Ricardo H Pineda
- Division of Urology, Department of Surgery, University of Colorado Denver School of Medicine, 12700 E 19th Avenue, Aurora, CO, 80045, USA
| | - Anna P Malykhina
- Division of Urology, Department of Surgery, University of Colorado Denver School of Medicine, 12700 E 19th Avenue, Aurora, CO, 80045, USA
| | - Duncan T Wilcox
- Division of Urology, Department of Surgery, University of Colorado Denver School of Medicine, 12700 E 19th Avenue, Aurora, CO, 80045, USA.
- Children's Hospital Colorado, 13123 E 16th Avenue, B463, Aurora, CO, 80045, USA.
| |
Collapse
|
14
|
Valve Bladder Syndrome Associated with Posterior Urethral Valves: Natural History, Work-up, and Management. CURRENT BLADDER DYSFUNCTION REPORTS 2020. [DOI: 10.1007/s11884-020-00577-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
15
|
Kaya-Sezginer E, Yilmaz-Oral D, Gur S. Administration of human umbilical cord blood mononuclear cells restores bladder dysfunction in streptozotocin-induced diabetic rats. Low Urin Tract Symptoms 2019; 11:232-240. [PMID: 31207098 DOI: 10.1111/luts.12268] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 02/01/2019] [Accepted: 04/24/2019] [Indexed: 12/17/2022]
Abstract
OBJECTIVE This study evaluated the effect of human umbilical cord blood mononuclear cells (HUCB-MNCs) on bladder dysfunction in streptozotocin (STZ; 35 mg/kg, i.v.)-induced diabetic rats. METHODS Adult male Sprague-Dawley rats (n = 30) were equally divided into three groups: control group, STZ-diabetic group, and HUCB-MNC-treated group (1 × 106 cells). HUCB-MNCs were isolated by density gradient centrifugation from eight healthy donors and injected into the corpus cavenosum in STZ-diabetic rats 4 weeks after the induction of diabetes. Studies were performed 4 weeks after HUCB-MNC or vehicle injection. In vitro organ bath studies were performed on bladder strips, whereas protein expression of hypoxia-inducible factor (HIF)-1α, vascular endothelial growth factor (VEGF), and α-smooth muscle actin (SMA) in the bladder and the ratio of smooth muscle cells (SMCs) to collagen were determined using western blotting and Masson trichrome staining. RESULTS Neurogenic contractions of detrusor smooth muscle strips were 55% smaller in the diabetic group than control group (P < 0.05); these contractions were normalized by HUCB-MNC treatment. In addition, HUCB-MNC treatment restored the impaired maximal carbachol-induced contractile response in detrusor strips in the diabetic group (29%; P < 0.05). HUCB-MNC treatment improved the KCl-induced contractile response in the diabetic bladder (68%; P < 0.05), but had no effect on ATP-induced contractile responses. Increased expression of HIF-1α and VEGF protein and decreased expression of α-SMA protein and the SMC/collagen ratio in diabetic rats were reversed by HUCB-MNC. CONCLUSION Administration of HUCB-MNCs facilitates bladder function recovery, which is likely related to downregulation of HIF-1α expression and attenuation of fibrosis in STZ-diabetic rats.
Collapse
Affiliation(s)
- Ecem Kaya-Sezginer
- Department of Biochemistry, Faculty of Pharmacy, Ankara University, Ankara, Turkey
| | - Didem Yilmaz-Oral
- Department of Pharmacology, Faculty of Pharmacy, Ankara University, Ankara, Turkey.,Department of Pharmacology, Faculty of Pharmacy, Cukurova University, Adana, Turkey
| | - Serap Gur
- Department of Pharmacology, Faculty of Pharmacy, Ankara University, Ankara, Turkey
| |
Collapse
|
16
|
Iguchi N, Dönmez Mİ, Carrasco A, Wilcox DT, Pineda RH, Malykhina AP, Cost NG. Doxorubicin induces detrusor smooth muscle impairments through myosin dysregulation, leading to a risk of lower urinary tract dysfunction. Am J Physiol Renal Physiol 2019; 317:F197-F206. [PMID: 31066574 DOI: 10.1152/ajprenal.00090.2019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Cytotoxic chemotherapy is the foundation for the treatment of the wide variety of childhood malignancies; however, these therapies are known to have a variety of deleterious side effects. One common chemotherapy used in children, doxorubicin (DOX), is well known to cause cardiotoxicity and cardiomyopathy. Recent studies have revealed that DOX impairs skeletal and smooth muscle function and contributes to fatigue and abnormal intestinal motility in patients. In this study, we tested the hypothesis that systemic DOX administration also affects detrusor smooth muscle (DSM) function in the urinary bladder, especially when administered at a young age. The effects on the DSM and bladder function were assessed in BALB/cJ mice that received six weekly intravenous injections of DOX (3 mg·kg-1·wk-1) or saline for the control group. Systemic DOX administration resulted in DSM hypertrophy, increased voiding frequency, and a significant attenuation of DSM contractility, followed by a slower relaxation compared with the control group. Gene expression analyses revealed that unlike DOX-induced cardiotoxicity, the bladders from DOX-administered animals showed no changes in oxidative stress markers; instead, downregulation of large-conductance Ca2+-activated K+ channels and altered expression of myosin light-chain kinase coincided with reduced myosin light-chain phosphorylation. These results indicate that in vivo DOX exposure caused DSM dysfunction by dysregulation of molecules involved in the detrusor contractile-relaxation mechanisms. Collectively, our findings suggest that survivors of childhood cancer treated with DOX may be at increased risk of bladder dysfunction and benefit from followup surveillance of bladder function.
Collapse
Affiliation(s)
- Nao Iguchi
- Division of Urology, Department of Surgery, University of Colorado Denver School of Medicine , Aurora, Colorado
| | - M İrfan Dönmez
- Division of Urology, Department of Surgery, University of Colorado Denver School of Medicine , Aurora, Colorado.,Children's Hospital Colorado , Aurora, Colorado
| | - Alonso Carrasco
- Children's Hospital Colorado , Aurora, Colorado.,Children's Mercy Kansas City, Kansas City, Missouri
| | - Duncan T Wilcox
- Division of Urology, Department of Surgery, University of Colorado Denver School of Medicine , Aurora, Colorado.,Children's Hospital Colorado , Aurora, Colorado
| | - Ricardo H Pineda
- Division of Urology, Department of Surgery, University of Colorado Denver School of Medicine , Aurora, Colorado
| | - Anna P Malykhina
- Division of Urology, Department of Surgery, University of Colorado Denver School of Medicine , Aurora, Colorado
| | - Nicholas G Cost
- Division of Urology, Department of Surgery, University of Colorado Denver School of Medicine , Aurora, Colorado.,Children's Hospital Colorado , Aurora, Colorado
| |
Collapse
|
17
|
Patel MS, Bowen DK, Tassone NM, Gould AD, Kochan KS, Firmiss PR, Kukulka NA, Devine MY, Li B, Gong EM, Dettman RW. The Homeodomain Transcription Factor NKX3.1 Modulates Bladder Outlet Obstruction Induced Fibrosis in Mice. Front Pediatr 2019; 7:446. [PMID: 31781523 PMCID: PMC6861332 DOI: 10.3389/fped.2019.00446] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 10/15/2019] [Indexed: 12/29/2022] Open
Abstract
Fibrosis is an irreversible remodeling process characterized by the deposition of collagen in the extracellular matrix of various organs through a variety of pathologies in children, leading to the stiffening of healthy tissues and organ dysfunction. Despite the prevalence of fibrotic disease in children, large gaps exist in our understanding of the mechanisms that lead to fibrosis, and there are currently no therapies to treat or reverse it. We previously observed that castration significantly reduces fibrosis in the bladders of male mice that have been partially obstructed. Here, we investigated if the expression of androgen response genes were altered in mouse bladders after partial bladder outlet obstruction (PO). Using a QPCR microarray and QRTPCR we found that PO was sufficient to increase expression of the androgen response gene Nkx3.1. Consistent with this was an increase in the expression of NKX3.1 protein. Immunofluorescent antibody localization demonstrated nuclear NKX3.1 in most bladder cells after PO. We tested if genetic deletion of Nkx3.1 alters remodeling of the bladder wall after PO. After PO, Nkx3.1 KO/KO bladders underwent remodeling, demonstrating smaller bladder area, thickness, and bladder: body weight ratios than obstructed, wild type controls. Remarkably, Nkx3.1 KO/KO specifically affected histological parameters of fibrosis, including reduced collagen to muscle ratio. Loss of Nkx3.1 altered collagen and smooth muscle cytoskeletal gene expression following PO which supported our histologic findings. Together these findings indicated that after PO, Nkx3.1 expression is induced in the bladder and that it mediates important pathways that lead to tissue fibrosis. As Nkx3.1 is an androgen response gene, our data suggest a possible mechanism by which fibrosis is mediated in male mice and opens the possibility of a molecular pathway mediated by NKX3.1 that could explain sexual dimorphism in bladder fibrosis.
Collapse
Affiliation(s)
- Mehul S Patel
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Diana K Bowen
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States.,Gong Laboratory, Division of Pediatric Urology, Stanley Manne Children's Research Institute, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, United States
| | - Nicholas M Tassone
- Gong Laboratory, Division of Pediatric Urology, Stanley Manne Children's Research Institute, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, United States
| | - Andrew D Gould
- Gong Laboratory, Division of Pediatric Urology, Stanley Manne Children's Research Institute, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, United States
| | - Kirsten S Kochan
- Gong Laboratory, Division of Pediatric Urology, Stanley Manne Children's Research Institute, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, United States
| | - Paula R Firmiss
- Gong Laboratory, Division of Pediatric Urology, Stanley Manne Children's Research Institute, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, United States
| | - Natalie A Kukulka
- Gong Laboratory, Division of Pediatric Urology, Stanley Manne Children's Research Institute, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, United States
| | - Megan Y Devine
- Gong Laboratory, Division of Pediatric Urology, Stanley Manne Children's Research Institute, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, United States
| | - Belinda Li
- Department of Urology, Loyola University Medical Center, Maywood, IL, United States
| | - Edward M Gong
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States.,Gong Laboratory, Division of Pediatric Urology, Stanley Manne Children's Research Institute, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, United States
| | - Robert W Dettman
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States.,Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| |
Collapse
|
18
|
Wang N, Duan L, Ding J, Cao Q, Qian S, Shen H, Qi J. MicroRNA-101 protects bladder of BOO from hypoxia-induced fibrosis by attenuating TGF-β-smad2/3 signaling. IUBMB Life 2018; 71:235-243. [PMID: 30549198 DOI: 10.1002/iub.1968] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 10/01/2018] [Indexed: 11/07/2022]
Abstract
Bladder outlet obstruction is a common disease, which always evokes urinary bladder wall remodeling significantly. It has been suggested that bladder outlet obstruction can make the bladder progression from inflammation to fibrosis, and hypoxia may play a vital role. It has been found the expression of microRNA-101 varied in bladder after BOO. But what role microRNA-101 and hypoxia play in bladder is not well known. This study is to investigate the mechanism of microRNA-101 and hypoxia in fibrosis of bladder after BOO. We found the expression of microRNA-101 and hif-1α increased in bladder after BOO. Hypoxia could promote the expression of extracellular matrix subtypes and microRNA-101 in BSMCs. When microRNA-101b was translated into BSMCs, the smad2/3 signaling pathway was found to repress. Dual luciferase reporter detected that microRNA-101b attenuated the TGF-β signaling pathway by inhibiting the expression of TGFβR1. Then, we conclude microRNA-101b is induced by hypoxia and represses fibrosis of BSMCs by inhibiting the expression of TGFβR1 through TGF-β signaling pathway, and it may be an anti-fibrotic miRNA for therapy. © 2018 IUBMB Life, 71(1):235-243, 2019.
Collapse
Affiliation(s)
- Ning Wang
- Department of Urology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liujian Duan
- Department of Urology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Ding
- Department of Urology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qifeng Cao
- Department of Urology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Subo Qian
- Department of Urology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haibo Shen
- Department of Urology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Qi
- Department of Urology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
19
|
Imamura T, Shimamura M, Ogawa T, Minagawa T, Nagai T, Silwal Gautam S, Ishizuka O. Biofabricated Structures Reconstruct Functional Urinary Bladders in Radiation-Injured Rat Bladders. Tissue Eng Part A 2018; 24:1574-1587. [DOI: 10.1089/ten.tea.2017.0533] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Affiliation(s)
- Tetsuya Imamura
- Department of Urology, Shinshu University School of Medicine, Nagano, Japan
| | | | - Teruyuki Ogawa
- Department of Urology, Shinshu University School of Medicine, Nagano, Japan
| | - Tomonori Minagawa
- Department of Urology, Shinshu University School of Medicine, Nagano, Japan
| | - Takashi Nagai
- Department of Urology, Shinshu University School of Medicine, Nagano, Japan
| | | | - Osamu Ishizuka
- Department of Urology, Shinshu University School of Medicine, Nagano, Japan
| |
Collapse
|
20
|
Tassone NM, Li B, Devine MY, Hausner PM, Patel MS, Gould AD, Kochan KS, Dettman RW, Gong EM. Voided volumes predict degree of partial bladder outlet obstruction in a murine model. AMERICAN JOURNAL OF CLINICAL AND EXPERIMENTAL UROLOGY 2018; 6:189-196. [PMID: 30510971 PMCID: PMC6261872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 10/08/2018] [Indexed: 06/09/2023]
Abstract
The partial bladder outlet obstruction animal model (pBOO) is commonly used as a model for obstructive uropathy. Unfortunately, pBOO demonstrates variable degrees of obstruction requiring bladder weight (BW) or urodynamic studies to determine true obstruction. Our objective is to identify extent of obstruction by correlating early post-operative Void Stains on Paper (VSOP) assays with ultimate BW in mice. pBOO was performed on 32 mice 1- and 4-week VSOPs were quantified for mean voided volume (mVV). At 4 weeks, bladders were harvested and weighed. Correlation was evaluated through bivariate kernel density estimation and a Pearson correlation coefficient (SAS). Single variable histogram of the data established groups based on BWs and mVV. mVV's and bladder weights within group pairings were averaged and plotted to render a non-linear regression model. A significant correlation was found between 1-week mVVs and 4-week BWs upon bivariate analysis with a correlation coefficient of -0.758 (p = 0.0294). A non-linear regression of plotted data defined a statistically significant fit equation correlating 1-week mVV to 4-week BW. We demonstrate a novel method for forecasting degree of obstruction in pBOO based on 1-week post-operative VSOP mVV.
Collapse
Affiliation(s)
- Nicholas M Tassone
- Department of Pediatric Urology, Ann & Robert H. Lurie Children’s Hospital of Chicago225 E. Chicago Ave., Chicago, IL 60611, USA
| | - Belinda Li
- Department of Pediatric Urology, Ann & Robert H. Lurie Children’s Hospital of Chicago225 E. Chicago Ave., Chicago, IL 60611, USA
- Department of Urology, Loyola University Health System2160 S. First Ave., Maywood, IL 60153, USA
| | - Megan Y Devine
- Department of Pediatric Urology, Ann & Robert H. Lurie Children’s Hospital of Chicago225 E. Chicago Ave., Chicago, IL 60611, USA
| | - Paulette M Hausner
- Department of Pediatric Urology, Ann & Robert H. Lurie Children’s Hospital of Chicago225 E. Chicago Ave., Chicago, IL 60611, USA
- Department of Biology, Loyola University1032 W. Sheridan Rd., Chicago, IL 60660, USA
| | - Mehul S Patel
- Department of Urology, Feinberg School of Medicine, Northwestern University303 E. Chicago Ave., 16-703, Chicago, IL 60611, USA
| | - Andrew D Gould
- Department of Pediatric Urology, Ann & Robert H. Lurie Children’s Hospital of Chicago225 E. Chicago Ave., Chicago, IL 60611, USA
| | - Kirsten S Kochan
- Department of Pediatric Urology, Ann & Robert H. Lurie Children’s Hospital of Chicago225 E. Chicago Ave., Chicago, IL 60611, USA
- Department of Biology, DePaul University1110 W. Belden Ave., Chicago, IL 60614, USA
| | - Robert W Dettman
- Department of Pediatric Urology, Ann & Robert H. Lurie Children’s Hospital of Chicago225 E. Chicago Ave., Chicago, IL 60611, USA
- Department of Urology, Feinberg School of Medicine, Northwestern University303 E. Chicago Ave., 16-703, Chicago, IL 60611, USA
| | - Edward M Gong
- Department of Pediatric Urology, Ann & Robert H. Lurie Children’s Hospital of Chicago225 E. Chicago Ave., Chicago, IL 60611, USA
- Department of Urology, Feinberg School of Medicine, Northwestern University303 E. Chicago Ave., 16-703, Chicago, IL 60611, USA
| |
Collapse
|
21
|
Hill WG, Zeidel ML, Bjorling DE, Vezina CM. Void spot assay: recommendations on the use of a simple micturition assay for mice. Am J Physiol Renal Physiol 2018; 315:F1422-F1429. [PMID: 30156116 DOI: 10.1152/ajprenal.00350.2018] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Investigators have for decades used mouse voiding patterns as end points for studying behavioral biology. It is only recently that mouse voiding patterns were adopted for study of lower urinary tract physiology. The spontaneous void spot assay (VSA), a popular micturition assessment tool, involves placing a mouse in an enclosure lined by filter paper and quantifying the resulting urine spot pattern. The VSA has advantages of being inexpensive and noninvasive, but some investigators challenge its ability to distinguish lower urinary tract function from behavioral voiding. A consensus group of investigators who regularly use the VSA was established by the National Institutes of Health in 2015 to address the strengths and weaknesses of the assay, determine whether it can be standardized across laboratories, and determine whether it can be used as a surrogate for evaluating urinary function. Here we leverage experience from the consensus group to review the history of the VSA and its uses, summarize experiments to optimize assay design for urinary physiology assessment, and make best practice recommendations for performing the assay and analyzing its results.
Collapse
Affiliation(s)
- Warren G Hill
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School , Boston, Massachusetts
| | - Mark L Zeidel
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School , Boston, Massachusetts
| | - Dale E Bjorling
- Department of Surgical Sciences, University of Wisconsin-Madison , Madison, Wisconsin.,University of Wisconsin-Madison/University of Massachusetts-Boston, George M. O'Brien Center for Benign Urologic Research, Madison, Wisconsin and Boston, Massachusetts
| | - Chad M Vezina
- University of Wisconsin-Madison/University of Massachusetts-Boston, George M. O'Brien Center for Benign Urologic Research, Madison, Wisconsin and Boston, Massachusetts.,Department of Comparative Biosciences, University of Wisconsin-Madison , Madison, Wisconsin
| |
Collapse
|
22
|
Iguchi N, Malykhina AP, Wilcox DT. Early life voiding dysfunction leads to lower urinary tract dysfunction through alteration of muscarinic and purinergic signaling in the bladder. Am J Physiol Renal Physiol 2018; 315:F1320-F1328. [PMID: 30089034 DOI: 10.1152/ajprenal.00154.2018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Lower urinary tract dysfunction (LUTD) is a common problem in children and constitutes up to 40% of pediatric urology clinic visits. Improved diagnosis and interventions have been leading to better outcomes in many patients, whereas some children are left untreated or do not respond to the treatment successfully. In addition, many of these patients are lost by the pediatric urologists during their teenage years, and the outcome in later life largely remains unidentified. Studies suggest childhood LUTD is associated with subsequent adult urinary tract symptoms. However, whether and how early life LUTD attributes to urinary symptoms in those patients later in life remains to be elucidated. In the current study, we investigated the effects of early life voiding perturbation on bladder function using a neonatal maternal separation (NMS) protocol in mice. The NMS group displayed a delayed development of voluntary voiding behavior, a significant reduction of functional bladder capacity, and bladder overactivity compared with control mice later in life. In vitro evaluation of detrusor smooth muscle and molecular study showed a decrease in muscarinic contribution alongside an increase in purinergic contribution in detrusor contractility in NMS mice compared with control group. These results suggest that early life bladder dysfunction interfered with the normal maturation of the voluntary micturition control and facilitated LUTD in a later stage, which is at least partly attributed to an alteration of muscarinic and purinergic signaling in the urinary bladder.
Collapse
Affiliation(s)
- Nao Iguchi
- Division of Urology, Department of Surgery, University of Colorado Denver School of Medicine , Aurora, Colorado
| | - Anna P Malykhina
- Division of Urology, Department of Surgery, University of Colorado Denver School of Medicine , Aurora, Colorado
| | - Duncan T Wilcox
- Division of Urology, Department of Surgery, University of Colorado Denver School of Medicine , Aurora, Colorado.,Children's Hospital Colorado , Aurora, Colorado
| |
Collapse
|