1
|
Pastuszka A, Tobor S, Łoniewski I, Wierzbicka-Woś A, Sielatycka K, Styburski D, Cembrowska-Lech D, Koszutski T, Kurowicz M, Korlacka K, Podkówka A, Lemiński A, Brodkiewicz A, Hyla-Klekot L, Skonieczna-Żydecka K. Rewriting the urinary tract paradigm: the urobiome as a gatekeeper of host defense. Mol Biol Rep 2025; 52:497. [PMID: 40407923 PMCID: PMC12102141 DOI: 10.1007/s11033-025-10609-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 05/13/2025] [Indexed: 05/26/2025]
Abstract
The urobiome, or urinary tract microbiome, has emerged as a crucial component in maintaining urinary health and defending against infections. Recent advances in next-generation sequencing (NGS) have debunked the long-held belief that the urinary tract is sterile, revealing a unique ecosystem of microorganisms. The urobiome interacts with the urothelium and mucosa-associated lymphoid tissue (MALT) to support local immunity, playing an integral role in defending the urinary tract against pathogens. Through complex communication processes like quorum sensing, the urobiome regulates microbial behavior and controls interactions with host tissues, helping to prevent pathogen colonization and infection. However, dysbiosis in the urobiome can disrupt this balance, making the urinary tract more susceptible to infections, including urinary tract infections (UTIs). Studies have highlighted specific microbial compositions associated with both healthy and disease states, suggesting that shifts in the urobiome may correlate with various urological diseases. Furthermore, microbial diversity within the urinary tract differs by factors such as age and gender, reflecting the dynamic nature of the urobiome. Future research focusing on the interplay between the urobiome, host immune defenses, and pathogenic mechanisms may lead to innovative diagnostic and therapeutic approaches. Understanding how microbial composition changes during disease states could enable targeted treatments, potentially reducing reliance on antibiotics and minimizing resistance issues. The urobiome thus represents a promising frontier in urology, with implications for enhancing urinary health and treating infections more effectively.
Collapse
Affiliation(s)
- Agnieszka Pastuszka
- Chair and Department of Descriptive and Topographic Anatomy, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Katowice, Poland
| | - Szymon Tobor
- Department of Pediatric Surgery and Urology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Igor Łoniewski
- Department of Biochemical Science, Faculty of Health Sciences, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | | | - Katarzyna Sielatycka
- Sanprobi Sp. z. o. o sp. k., Szczecin, Poland
- Institute of Biology, Faculty of Exact and Natural Sciences, University of Szczecin, Szczecin, Poland
| | | | | | - Tomasz Koszutski
- Department of Pediatric Surgery and Urology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Marek Kurowicz
- Chair and Department of Descriptive and Topographic Anatomy, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Katowice, Poland
| | - Klaudia Korlacka
- Department of Pediatric Surgery and Urology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Albert Podkówka
- Department of Biochemical Science, Faculty of Health Sciences, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Artur Lemiński
- Department of Biochemical Science, Faculty of Health Sciences, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Andrzej Brodkiewicz
- Department of Pediatrics, Child Nephrology, Dialysotherapy and Management of Acute Poisoning, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Lidia Hyla-Klekot
- Department of Pediatric Surgery and Urology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Karolina Skonieczna-Żydecka
- Department of Biochemical Science, Faculty of Health Sciences, Pomeranian Medical University in Szczecin, Szczecin, Poland.
| |
Collapse
|
2
|
Wille S, Peukert N, Haak R, Riedel J, Mayer S, Kluth D, Lacher M, Gosemann JH, Markel M. Development of the Urinary Tract in Fetal Rats: A Micro-CT Study. Eur J Pediatr Surg 2023; 33:53-60. [PMID: 36395789 DOI: 10.1055/s-0042-1758681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Micro-computed tomography (micro-CT) is an established tool to study fetal development in rodents. This study aimed to use micro-CT imaging to visualize the development of the urinary tract in fetal rats. MATERIALS AND METHODS Fetal rats from embryonic day (ED) 15, ED17, ED19, ED21, and N0 (newborn) (n = 6 per group; 3 males) were fixed and desiccated using the "critical point" technique. We utilized the micro-CT system (SkyScan) and analyzed the resulting scans with CTAn, DataViewer, and ImageJ to visualize the morphology and quantify the volumes of kidney, bladder, adrenal gland, as well as length of the ureter. RESULTS High-resolution micro-CT showed continuous growth of both kidneys from ED15 to N0, with the highest increase between ED19 and ED21. The length of the ureter increased from ED15 to ED21 and remained stable until birth. The volume of the bladder steadily increased from ED15 to N0.In females, a statistically higher volume of the adrenal gland on ED21 was observed, whereas no sex-specific differences were seen for kidney, ureter, and bladder development. CONCLUSION Micro-CT depicts an excellent tool to study urinary tract development in the fetal and neonatal rat. It enables the metric quantification of longitudinal anatomic changes in high definition without previous destructive tissue preparation. The present study revealed sex-specific differences of the adrenal gland development and provides comprehensive data for the understanding of fetal urinary tract development, inspiring future research on congenital urological malformations.
Collapse
Affiliation(s)
- Stephanie Wille
- Department of Pediatric Surgery, University of Leipzig, Leipzig, Germany.,Department of Interdisciplinary Medical Intensive Care, University of Leipzig, Leipzig, Germany
| | - Nicole Peukert
- Department of Pediatric Surgery, University of Leipzig, Leipzig, Germany
| | - Rainer Haak
- Department of Cariology, Endodontology, and Periodontology, University of Leipzig, Leipzig, Germany
| | - Jan Riedel
- Department of Pediatric Surgery, University of Leipzig, Leipzig, Germany
| | - Steffi Mayer
- Department of Pediatric Surgery, University of Leipzig, Leipzig, Germany
| | - Dietrich Kluth
- Department of Pediatric Surgery, University of Leipzig, Leipzig, Germany
| | - Martin Lacher
- Department of Pediatric Surgery, University of Leipzig, Leipzig, Germany
| | | | - Moritz Markel
- Department of Pediatric Surgery, University of Leipzig, Leipzig, Germany
| |
Collapse
|
3
|
Yu Q, Wu C, Chen Y, Li B, Wang R, Huang R, Li X, Gu D, Wang X, Duan X, Li S, Liu Y, Wu W, Hennenberg M, Zeng G. Inhibition of LIM kinase reduces contraction and proliferation in bladder smooth muscle. Acta Pharm Sin B 2021; 11:1914-1930. [PMID: 34386328 PMCID: PMC8343115 DOI: 10.1016/j.apsb.2021.01.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 10/17/2020] [Accepted: 11/03/2020] [Indexed: 01/03/2023] Open
Abstract
Overactive bladder (OAB) is the most bothersome symptom in lower urinary tract symptoms (LUTS). Current pharmacologic treatment aims to inhibit detrusor contraction; however, shows unsatisfied efficacy and high discontinuation rate. LIM kinases (LIMKs) promote smooth muscle contraction in the prostate; however, their function in the bladder smooth muscle remains unclear. Here, we studied effects of the LIMK inhibitors on bladder smooth muscle contraction and proliferation both in vitro and in vivo experiments. Bladder expressions of LIMKs are elevated in OAB rat detrusor tissues. Two LIMK inhibitors, SR7826 and LIMKi3, inhibit contraction of human detrusor strip, and cause actin filament breakdown, as well as cell proliferation reduction in cultured human bladder smooth muscle cells (HBSMCs), paralleled by reduced cofilin phosphorylation. Silencing of LIMK1 and LIMK2 in HBSMCs resulted in breakdown of actin filaments and decreased cell proliferation. Treatment with SR7826 or LIMKi3 decreased micturition frequency and bladder detrusor hypertrophy in rats with bladder outlet obstruction. Our study suggests that LIMKs may promote contraction and proliferation in the bladder smooth muscle, which could be inhibited by small molecule LIMK inhibitors. LIMK inhibitors could be a potential therapeutic strategy for OAB- related LUTS.
Collapse
Key Words
- 4E-BP1, 4E-binding protein 1
- ADF, actin depolymerizing factors
- BOO, bladder outlet obstruction
- BPH, benign prostatic hyperplasia
- Bladder smooth muscle contraction
- CCK-8, Cell Counting Kit-8
- Cofilin phosphorylation
- Ct, number of cycles
- DMSO, dimethyl sulfoxide
- EdU, 5-ethynyl-2′-deoxyuridine
- GAPDH, glyceraldehyde 3-phosphate dehydrogenase
- H&E, hematoxylin and eosin
- HBSMCs, human bladder smooth muscle cells
- HRP, horseradish peroxidase
- LIMK
- LIMKs, LIM kinases
- LUTS, lower urinary tract symptoms
- Lower urinary tract symptoms (LUTS)
- MLC, myosin light chain
- MW, molecular weight
- MYPT1, myosin-binding subunit
- OAB, overactive bladder
- Overactive bladder (OAB)
- PCNA, proliferating cell nuclear antigen
- RT-qPCR, reverse transcription and quantitative polymerase chain reaction
- STK16, serine/threonine kinase 16
- TESK1, testicular protein kinase 1
- TXA2, thromboxane A2
- WST-8, 2-(2-methoxy-4-nitrophenyl)-3-(4-nitrophenyl)-5-(2,4-disulfophenyl)-2H-tetrazolium monosodium salt
- siRNA, small interfering RNA
Collapse
Affiliation(s)
- Qingfeng Yu
- Department of Urology and Guangdong Key Laboratory of Urology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510230, China
| | - Chengjie Wu
- Department of Urology and Guangdong Key Laboratory of Urology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510230, China
| | - Yeda Chen
- Department of Urology and Guangdong Key Laboratory of Urology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510230, China
| | - Bingsheng Li
- Department of Urology, University Hospital, LMU Munich, Munich 81377, Germany
| | - Ruixiao Wang
- Department of Urology, University Hospital, LMU Munich, Munich 81377, Germany
| | - Ru Huang
- Department of Urology, University Hospital, LMU Munich, Munich 81377, Germany
| | - Xuechun Li
- Department of Urology and Guangdong Key Laboratory of Urology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510230, China
| | - Di Gu
- Department of Urology and Guangdong Key Laboratory of Urology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510230, China
| | - Xiaolong Wang
- Department of Urology, University Hospital, LMU Munich, Munich 81377, Germany
| | - Xiaolu Duan
- Department of Urology and Guangdong Key Laboratory of Urology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510230, China
| | - Shujue Li
- Department of Urology and Guangdong Key Laboratory of Urology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510230, China
| | - Yang Liu
- Department of Urology and Guangdong Key Laboratory of Urology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510230, China
| | - Wenqi Wu
- Department of Urology and Guangdong Key Laboratory of Urology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510230, China
| | - Martin Hennenberg
- Department of Urology, University Hospital, LMU Munich, Munich 81377, Germany
| | - Guohua Zeng
- Department of Urology and Guangdong Key Laboratory of Urology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510230, China
- Corresponding author. Tel.: +86 20 34294165.
| |
Collapse
|
4
|
Nakajima T, Sakai N, Nogimura M, Tomooka Y. Developmental mechanisms regulating the formation of smooth muscle layers in the mouse uterus†. Biol Reprod 2020; 103:750-759. [DOI: 10.1093/biolre/ioaa104] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 05/20/2020] [Accepted: 06/12/2020] [Indexed: 11/13/2022] Open
Abstract
Abstract
Uterine smooth muscle cells differentiate from mesenchymal cells, and gap junctions connect the muscle cells in the myometrium. At the neonatal stage, a uterine smooth muscle layer is situated away from the epithelium when smooth muscle cells are grafted near the epithelium, suggesting that the epithelium plays an important role in differentiation, proliferation, and/or migration of smooth muscle cells. In this study, developmental mechanisms regulating the formation of the smooth muscle layers in the mouse uterus were analyzed using an in vitro culture model. Differentiation of smooth muscle cells occurs at a neonatal stage because ACTA2 gene expression was increased at the outer layer, and GJA1 was not expressed in cellular membranes of uterine smooth muscle cells by postnatal day 15. To analyze the effects of the epithelium on the differentiation of smooth muscle cells, a bulk uterine mesenchymal cell line was established from p53−/− mice at postnatal day 3 (P3US cells). Co-culture with Müllerian ductal epithelial cells (E1 cells) induced repulsive migration of ACTA2-positive cells among bulk P3US cells from E1 cells, but it had no effects on the migration of any of 100% ACTA2-positive or negative smooth muscle cell lines cloned from P3US cells. Thus, uterine epithelial cells indirectly affected the repulsive migration of smooth muscle cells via mesenchymal cells. Conditioned medium by E1 cells inhibited differentiation into smooth muscle cells of clonal cells established from P3US cells. Therefore, the uterine epithelium inhibits the differentiation of stem-like progenitor mesenchymal cells adjacent to the epithelium into smooth muscle cells.
Collapse
Affiliation(s)
- Tadaaki Nakajima
- Department of Biological Science and Technology, Faculty of Industrial Science and Technology, Tokyo University of Science, Tokyo, Japan
- Institute of Industrial Science, The University of Tokyo, Tokyo, Japan
| | - Naoto Sakai
- Department of Biological Science and Technology, Faculty of Industrial Science and Technology, Tokyo University of Science, Tokyo, Japan
| | - Miho Nogimura
- Department of Biological Science and Technology, Faculty of Industrial Science and Technology, Tokyo University of Science, Tokyo, Japan
| | - Yasuhiro Tomooka
- Department of Biological Science and Technology, Faculty of Industrial Science and Technology, Tokyo University of Science, Tokyo, Japan
| |
Collapse
|
5
|
Chamorro CI, Reinfeldt Engberg G, Fossum M. Molecular and histological studies of bladder wound healing in a rodent model. Wound Repair Regen 2020; 28:293-306. [PMID: 32011053 DOI: 10.1111/wrr.12797] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 11/27/2019] [Accepted: 01/16/2020] [Indexed: 01/01/2023]
Abstract
The field of regenerative medicine encounters different challenges. The success of tissue-engineered implants is dependent on proper wound healing. Today, the process of normal urinary bladder wound healing is poorly characterized. We aspired to explore and elucidate the natural response to injury in an in vivo model in order to further optimize tissue regeneration in future studies. In this study, we aimed to characterize histological and molecular changes during normal healing in a rat model by performing a standardized incisional wound followed by surgical closure. We used a rodent model (n = 40) to follow the healing process in the urinary bladder for 28 days. Surgical exposure of the bladder without incision (n = 40) was performed in controls. Histological characterization and western blot analyses of proteins was carried out using specific staining and markers for inflammation, proliferation, angiogenesis, and tissue maturation. For the molecular characterization of gene expression total RNA was collected for RT2 -PCR in wound healing pathway arrays. Analysis of histology revealed distinct, but overlapping, phases of healing with a local inflammatory response (days 1-8) simultaneous with a rapid formation of granulation tissue and proliferation (days 2-8). We also identified significant changes in gene expression related to inflammation, proliferation, and extracellular matrix formation. Healing of an incisional wound in a rodent urinary bladder demonstrated that all the classical phases of wound healing: hemostasis, inflammation, proliferation followed by tissue maturation were present. Our data suggest that the bladder and the skin share similar molecular signaling during wound healing, although we noted differences in the duration of each phase compared to previous studies in rat skin. Further studies will address whether our findings can be extrapolated to the human bladder.
Collapse
Affiliation(s)
- Clara I Chamorro
- Department of Women's and Children's Health, Center of Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,Department of Pediatric Surgery, Surgical Clinic C, Copenhagen University Hospital Rigshospitalet, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Gisela Reinfeldt Engberg
- Department of Women's and Children's Health, Center of Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,Department of Pediatric Surgery, Uppsala University Children's Hospital, Uppsala, Sweden
| | - Magdalena Fossum
- Department of Women's and Children's Health, Center of Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,Department of Highly Specialized Pediatric Surgery and Medicine, Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden.,Department of Pediatric Surgery, Surgical Clinic C, Copenhagen University Hospital Rigshospitalet, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| |
Collapse
|
6
|
Bouhout S, Chabaud S, Bolduc S. Collagen hollow structure for bladder tissue engineering. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 102:228-237. [DOI: 10.1016/j.msec.2019.04.052] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 04/13/2019] [Accepted: 04/16/2019] [Indexed: 01/03/2023]
|
7
|
Sun S, Yang H. Tissue-Specific Localization NUCB2/nesfatin-1 in the Liver and Heart of Mouse Fetus. Dev Reprod 2018; 22:331-339. [PMID: 30680332 PMCID: PMC6344366 DOI: 10.12717/dr.2018.22.4.331] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 11/17/2018] [Accepted: 12/05/2018] [Indexed: 02/06/2023]
Abstract
NUCB2/nesfatin-1 is first known to be expressed in the hypothalamus while controlling appetite and energy metabolism. However, recent studies have shown that NUCB2/nesfatin-1 was expressed in the various organs as well as the hypothalamus. Our previous reports also demonstrated that NUCB2/nesfatin-1 was expressed in the ovary, testis, pituitary gland, lung, kidney, and stomach of fetal and adult mice. However, the role of NUCB2/nesfatin-1 in mouse fetus remains unknown. Thus, the aim of this study was to investigate whether NUCB2/nestatin-1 is expressed in mouse fetus at the developmental stage in which organogenesis begins. To do this, we performed in situ hybridization (ISH) and immunohistochemistry (IHC) staining to examine the distribution of NUCB2 mRNA and nesfatin-1 protein in the mouse fetal organs during early developmental stages, especially at embryonic day (E) 10.5. As a result of ISH, NUCB2 mRNA positive signals were more frequent in the liver, but there were relatively few positive signals in heart. On the other hand, no positive signals were detected in other organs. These ISH results were validated by IHC staining and qRT-PCR analysis. Expression of nesfatin-1 protein detected by IHC staining was similar to that of NUCB2 mRNA detected by ISH in the liver and heart. In addition, the levels of NUCB2 mRNA expression analyzed by qRT-PCR were significantly increased in the liver and heart compared to other organs of the mouse fetus at E13.5, whereas its level was extensively decreased in the liver, but increased in the lung, stomach, and kidney of the mouse fetus at E17.5. These results suggest that NUCB2/nesfatin-1 may play an important role in liver and heart development and physiological functions in the developmental process of mouse fetus. Further studies are needed on the function of NUCB2/nesfatin-1, which is highly expressed in the various organs, including liver and heart during mouse development.
Collapse
Affiliation(s)
- Sojung Sun
- Dept. of Bioenvironmental Technology, College of Natural Sciences, Seoul Women's University, Seoul 01797, Korea
| | - Hyunwon Yang
- Dept. of Bioenvironmental Technology, College of Natural Sciences, Seoul Women's University, Seoul 01797, Korea
| |
Collapse
|
8
|
Liaw A, Cunha GR, Shen J, Cao M, Liu G, Sinclair A, Baskin L. Development of the human bladder and ureterovesical junction. Differentiation 2018; 103:66-73. [PMID: 30236462 DOI: 10.1016/j.diff.2018.08.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 08/22/2018] [Accepted: 08/24/2018] [Indexed: 11/13/2022]
Abstract
The urinary bladder collects urine from the kidneys and stores it until the appropriate moment for voiding. The trigone and ureterovesical junctions are key to bladder function, by allowing one-way passage of urine into the bladder without obstruction. Embryological development of these structures has been studied in multiple animal models as well as humans. In this report we review the existing literature on bladder development and cellular signalling with particular focus on bladder development in humans. The bladder and ureterovesical junction form primarily during the fourth to eighth weeks of gestation, and arise from the primitive urogenital sinus following subdivision of the cloaca. The bladder develops through mesenchymal-epithelial interactions between the endoderm of the urogenital sinus and mesodermal mesenchyme. Key signalling factors in bladder development include shh, TGF-β, Bmp4, and Fgfr2. A concentration gradient of shh is particularly important in development of bladder musculature, which is vital to bladder function. The ureterovesical junction forms from the interaction between the Wolffian duct and the bladder. The ureteric bud arises from the Wolffian duct and is incorporated into the developing bladder at the trigone. It was previously thought that the trigonal musculature developed primarily from the Wolffian duct, but it has been shown to develop primarily from bladder mesenchyme. Following emergence of the ureters from the Wolffian ducts, extensive epithelial remodelling brings the ureters to their final trigonal positions via vitamin A-induced apoptosis. Perturbation of this process is implicated in clinical obstruction or urine reflux. Congenital malformations include ureteric duplication and bladder exstrophy.
Collapse
Affiliation(s)
- Aron Liaw
- Department of Urology, University of California, San Francisco, San Francisco, CA Division of Pediatric Urology, University of California San Francisco Benioff Children's Hospital, San Francisco, CA 94143, United States
| | - Gerald R Cunha
- Department of Urology, University of California, San Francisco, San Francisco, CA Division of Pediatric Urology, University of California San Francisco Benioff Children's Hospital, San Francisco, CA 94143, United States
| | - Joel Shen
- Department of Urology, University of California, San Francisco, San Francisco, CA Division of Pediatric Urology, University of California San Francisco Benioff Children's Hospital, San Francisco, CA 94143, United States
| | - Mei Cao
- Department of Urology, University of California, San Francisco, San Francisco, CA Division of Pediatric Urology, University of California San Francisco Benioff Children's Hospital, San Francisco, CA 94143, United States
| | - Ge Liu
- Department of Urology, University of California, San Francisco, San Francisco, CA Division of Pediatric Urology, University of California San Francisco Benioff Children's Hospital, San Francisco, CA 94143, United States
| | - Adriane Sinclair
- Department of Urology, University of California, San Francisco, San Francisco, CA Division of Pediatric Urology, University of California San Francisco Benioff Children's Hospital, San Francisco, CA 94143, United States
| | - Laurence Baskin
- Department of Urology, University of California, San Francisco, San Francisco, CA Division of Pediatric Urology, University of California San Francisco Benioff Children's Hospital, San Francisco, CA 94143, United States.
| |
Collapse
|
9
|
Dong J, Hu Y, Fan X, Wu X, Mao Y, Hu B, Guo H, Wen L, Tang F. Single-cell RNA-seq analysis unveils a prevalent epithelial/mesenchymal hybrid state during mouse organogenesis. Genome Biol 2018; 19:31. [PMID: 29540203 PMCID: PMC5853091 DOI: 10.1186/s13059-018-1416-2] [Citation(s) in RCA: 129] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 02/28/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Organogenesis is crucial for proper organ formation during mammalian embryonic development. However, the similarities and shared features between different organs and the cellular heterogeneity during this process at single-cell resolution remain elusive. RESULTS We perform single-cell RNA sequencing analysis of 1916 individual cells from eight organs and tissues of E9.5 to E11.5 mouse embryos, namely, the forebrain, hindbrain, skin, heart, somite, lung, liver, and intestine. Based on the regulatory activities rather than the expression patterns, all cells analyzed can be well classified into four major groups with epithelial, mesodermal, hematopoietic, and neuronal identities. For different organs within the same group, the similarities and differences of their features and developmental paths are revealed and reconstructed. CONCLUSIONS We identify mutual interactions between epithelial and mesenchymal cells and detect epithelial cells with prevalent mesenchymal features during organogenesis, which are similar to the features of intermediate epithelial/mesenchymal cells during tumorigenesis. The comprehensive transcriptome at single-cell resolution profiled in our study paves the way for future mechanistic studies of the gene-regulatory networks governing mammalian organogenesis.
Collapse
Affiliation(s)
- Ji Dong
- Beijing Advanced Innovation Center for Genomics (ICG), Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking University, Beijing, 100871, People's Republic of China
- Biomedical Institute for Pioneering Investigation via Convergence, College of Life Sciences, Peking University, Beijing, 100871, People's Republic of China
| | - Yuqiong Hu
- Beijing Advanced Innovation Center for Genomics (ICG), Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking University, Beijing, 100871, People's Republic of China
- Biomedical Institute for Pioneering Investigation via Convergence, College of Life Sciences, Peking University, Beijing, 100871, People's Republic of China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, People's Republic of China
| | - Xiaoying Fan
- Beijing Advanced Innovation Center for Genomics (ICG), Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking University, Beijing, 100871, People's Republic of China
- Biomedical Institute for Pioneering Investigation via Convergence, College of Life Sciences, Peking University, Beijing, 100871, People's Republic of China
| | - Xinglong Wu
- Beijing Advanced Innovation Center for Genomics (ICG), Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking University, Beijing, 100871, People's Republic of China
- Biomedical Institute for Pioneering Investigation via Convergence, College of Life Sciences, Peking University, Beijing, 100871, People's Republic of China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, People's Republic of China
| | - Yunuo Mao
- Beijing Advanced Innovation Center for Genomics (ICG), Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking University, Beijing, 100871, People's Republic of China
- Biomedical Institute for Pioneering Investigation via Convergence, College of Life Sciences, Peking University, Beijing, 100871, People's Republic of China
| | - Boqiang Hu
- Beijing Advanced Innovation Center for Genomics (ICG), Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking University, Beijing, 100871, People's Republic of China
- Biomedical Institute for Pioneering Investigation via Convergence, College of Life Sciences, Peking University, Beijing, 100871, People's Republic of China
| | - Hongshan Guo
- Beijing Advanced Innovation Center for Genomics (ICG), Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking University, Beijing, 100871, People's Republic of China
- Biomedical Institute for Pioneering Investigation via Convergence, College of Life Sciences, Peking University, Beijing, 100871, People's Republic of China
| | - Lu Wen
- Beijing Advanced Innovation Center for Genomics (ICG), Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking University, Beijing, 100871, People's Republic of China
- Biomedical Institute for Pioneering Investigation via Convergence, College of Life Sciences, Peking University, Beijing, 100871, People's Republic of China
| | - Fuchou Tang
- Beijing Advanced Innovation Center for Genomics (ICG), Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking University, Beijing, 100871, People's Republic of China.
- Biomedical Institute for Pioneering Investigation via Convergence, College of Life Sciences, Peking University, Beijing, 100871, People's Republic of China.
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, People's Republic of China.
| |
Collapse
|
10
|
Cunha GR, Baskin L. Mesenchymal-epithelial interaction techniques. Differentiation 2016; 91:20-7. [PMID: 26610327 PMCID: PMC4874915 DOI: 10.1016/j.diff.2015.10.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Accepted: 10/24/2015] [Indexed: 11/18/2022]
Abstract
This paper reviews the importance of mesenchymal-epithelial interactions in development and gives detailed technical protocols for investigating these interactions. Successful analysis of mesenchymal-epithelial interactions requires knowing the ages in which embryonic, neonatal and adult organs can be separated into mesenchymal and epithelial tissues. Methods for separation of mesenchymal and epithelial tissues and preparation of tissue recombinants are described.
Collapse
Affiliation(s)
- Gerald R Cunha
- Department of Urology, University of California, Box 0738, San Francisco, CA 94143, United States.
| | - Lawrence Baskin
- Department of Urology, University of California, Box 0738, San Francisco, CA 94143, United States
| |
Collapse
|
11
|
Bouhout S, Tremblay J, Bolduc S. Maintenance of bladder urothelia integrity and successful urothelialization of various tissue-engineered mesenchymes in vitro. In Vitro Cell Dev Biol Anim 2015; 51:922-31. [PMID: 26091628 DOI: 10.1007/s11626-015-9923-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 05/15/2015] [Indexed: 11/29/2022]
Abstract
Tissue-engineering offers the opportunity to produce hybrid tissues in vitro. The induction of bladder urothelial cells (BUCs) differentiation in vitro has been assessed by several research groups to build bladder models for fundamental studies and clinical applications. However, BUC induction of advanced differentiation in culture remains a challenging task. To reach this goal, optimal culture conditions are required, notably the use of specific additives as well as proper mesenchymal support. The best positive control for BUCs functional state monitoring is native urothelium collected from healthy bladder samples. In order to establish the best culture conditions to maintain and promote BUC differentiated state, native urothelia were cultured on various mesenchymes. Native bladder mesenchymes were used as controls for the maintenance of native urothelia. Histological and ultrastructural analyses showed the necessity to have a cellularized mesenchyme for rapid formation of a pseudostratified urothelium, allowing apical membrane rearrangement of the superficial cells in culture. Taken together, the results strongly suggest that it is possible to conserve the integrity of urothelia in vitro and, thus, potentially use them for eventual clinical applications and pharmacological investigations.
Collapse
Affiliation(s)
- Sara Bouhout
- Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEX Faculté de médecine, Université Laval, Québec, Canada. .,Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEX, Faculté de médecine, Centre de Recherche du CHU de Québec, Université Laval, Québec, Canada.
| | - Julie Tremblay
- Service d'anatomopathologie du CHU de Québec, Québec, Canada
| | - Stephane Bolduc
- Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEX, Faculté de médecine, Centre de Recherche du CHU de Québec, Université Laval, Québec, Canada
| |
Collapse
|
12
|
Mahfuz I, Darling T, Wilkins S, White S, Cheng W. New insights into the pathogenesis of bladder exstrophy-epispadias complex. J Pediatr Urol 2013; 9:996-1005. [PMID: 23743131 DOI: 10.1016/j.jpurol.2013.05.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2012] [Accepted: 05/01/2013] [Indexed: 02/02/2023]
Abstract
Bladder exstrophy-epispadias complex (BEEC) is a complex and debilitating congenital disease. Familial and twin studies suggest a possible genetic component in BEEC pathogenesis. Bladder mesenchyme (detrusor) development requires induction by a signal from bladder urothelium, and we and others have shown the Shh-Gli-Bmp4 signalling pathway is likely to be involved. P63 is a master regulator in epithelial stratification and is expressed in urothelium. We have shown that p63 knock-out mice undergo excessive urothelial apoptosis. Failure of mesenchymal induction by epithelium leads to BEEC. We further demonstrated that insertion/deletion (in/del) polymorphisms (1 base pair (bp) ins and 4 bp ins., and 12 bp del) in the ΔNP63 promoter reduce transcriptional efficiency, and are associated with a statistically significant increase in the risk of BEEC in humans. Furthermore, a Genome-Wide Expression Profiling (GWEP) study suggests possible involvement of PERP in human BEEC. Intriguingly, PERP is a direct target of p63 during development, and is also involved in epithelial stratification. PERP co-localizes with desmosome, and both PERP and desmosome are essential for maintaining tissue integrity by cellular adhesion and epithelial stratification. A recent study showed that PERP and desmosome expression levels are abnormal in human BEEC patients. This review describes the role of the P63 > PERP > desmosome pathway in the development of human bladder during embryogenesis. We hypothesize that disruption of this pathway may increase the risk of BEEC.
Collapse
Affiliation(s)
- Istiak Mahfuz
- Monash Institute of Medical Research, Faculty of Medicine, Nursing and Health Sciences, Monash University, Australia
| | | | | | | | | |
Collapse
|
13
|
Islam SS, Mokhtari RB, Kumar S, Maalouf J, Arab S, Yeger H, Farhat WA. Spatio-temporal distribution of Smads and role of Smads/TGF-β/BMP-4 in the regulation of mouse bladder organogenesis. PLoS One 2013; 8:e61340. [PMID: 23620745 PMCID: PMC3631207 DOI: 10.1371/journal.pone.0061340] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Accepted: 03/07/2013] [Indexed: 11/24/2022] Open
Abstract
Although Shh, TGF-β and BMP-4 regulate radial patterning of the bladder mesenchyme and smooth muscle differentiation, it is not known what transcription factors, local environmental cues or signaling cascades mediate bladder smooth muscle differentiation. We investigated the expression patterns of signaling mediated by Smad2 and Smad3 in the mouse embryonic bladder from E12.5 to E16.5 by using qRT-PCR, in situ hybridization and antibodies specifically recognizing individual Smad proteins. The role of Smad2 and Smad3 during smooth muscle formation was examined by disrupting the Smad2/3 signaling pathway using TβR1 inhibitor SB-431542 in organ culture system. qRT-PCR results showed that R-Smads, Co-Smad and I-Smads were all expressed during bladder development. RNA ISH for BMP-4 and immunostaining of TGF-β1 showed that BMP-4 and TGF-β1 were expressed in the transitional epithelium, lamina propia and muscularis mucosa. Smad1, Smad5 and Smad8 were first expressed in the bladder epithelium and continued to be expressed in the transitional epithelium, muscularis mesenchyme and lamina propia as the bladder developed. Smad2, Smad3 and Smad4 were first detected in the bladder epithelium and subsequently were expressed in the muscularis mesenchyme and lamina propia. Smad6 and Smad7 showed overlapping expression with R-Smads, which are critical for bladder development. In bladder explants (E12.5 to E16.5) culture, Smad2 and Smad3 were found localized within the nuclei, suggesting critical transcriptional regulatory effects during bladder development. E12.5 to E16.5 bladders were cultured with and without TβR1 inhibitor SB-431542 and assessed by qRT-PCR and immunofluorescence. After three days in culture in SB-431542, α-SMA, Smad2 and Smad3 expressions were significantly decreased compared with controls, however, with no significant changes in the expression of smooth muscle myosin heavy chain (SM-Myh. Based on the Smad expression patterns, we suggest that individual or combinations of Smads may be necessary during mouse bladder organogenesis and may be critical mediators for bladder smooth muscle differentiation.
Collapse
Affiliation(s)
- Syed S. Islam
- Developmental and Stem Cell Biology, The Hospital for Sick Children Research Institute, Toronto, ON, Canada
| | - Reza Bayat Mokhtari
- Department of Pediatric Laboratory Medicine, The Hospital for Sick Children Research Institute, Toronto, ON, Canada
| | - Sushil Kumar
- Physiology and Experimental Medicine, The Hospital for Sick Children Research Institute, Toronto, ON, Canada
| | | | - Sara Arab
- University of Toronto, Toronto, ON, Canada
| | - Herman Yeger
- Department of Pediatric Laboratory Medicine, The Hospital for Sick Children Research Institute, Toronto, ON, Canada
| | - Walid A. Farhat
- Developmental and Stem Cell Biology, The Hospital for Sick Children Research Institute, Toronto, ON, Canada
| |
Collapse
|
14
|
Hicks AN, Campeau L, Burmeister D, Bishop CE, Andersson KE. Lack of nicotinamide mononucleotide adenylyltransferase 2 (Nmnat2): consequences for mouse bladder development and function. Neurourol Urodyn 2013; 32:1130-6. [PMID: 23371862 DOI: 10.1002/nau.22372] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2012] [Accepted: 12/12/2012] [Indexed: 11/09/2022]
Abstract
AIMS To describe the morphological and functional consequences for bladder development and function when nicotinamide mononucleotide adenylyltransferase 2 (Nmnat2) is lacking or reduced. METHODS The Bloated Bladder (Blad) mouse, lacking Nmnat2, and heterozygotes were utilized for this investigation. Morphology and development of the bladder were studied using immunohistochemistry against urothelial, smooth muscle, and nerve markers. Functional effects were assessed by organ bath experiments and cystometry. RESULTS Homozygote mutants were malformed and died at birth, whereas heterozygotes survived and morphologically did not differ from wild-type controls. Morphological bladder changes appeared in the Blad mutants as early as embryonic day 15.5 (E15.5) with an extremely distended bladder at E18.5. Staining revealed that all the bladder layers were present and expressed mature markers in all three genotypes. No nerves could be demonstrated by immunohistochemistry in the Blad mutant bladder at E18.5. Organ bath analysis showed that bladders from Blad mutant showed signs of denervation supersensitivity in response to carbachol, and no response to electrical stimulation of nerves at E18.5. Adult heterozygotes, which have a reduced expression of Nmnat2 at E18.5, showed decreased responses to carbachol and electrical stimulation compared to wild-type controls. The latter also retained their ability to empty their bladders, but showed increased micturition pressures compared to controls. CONCLUSIONS Complete loss of Nmnat2 leads to a mature but distended bladder in utero and is not compatible with survival. Moderate loss of Nmnat2 has no effect on bladder development, survival, and has only modest effects on bladder function later in life.
Collapse
Affiliation(s)
- Amy N Hicks
- Wake Forest Institute for Regenerative Medicine, Winston Salem, North Carolina
| | | | | | | | | |
Collapse
|
15
|
DeSouza KR, Saha M, Carpenter AR, Scott M, McHugh KM. Analysis of the Sonic Hedgehog signaling pathway in normal and abnormal bladder development. PLoS One 2013; 8:e53675. [PMID: 23308271 PMCID: PMC3538723 DOI: 10.1371/journal.pone.0053675] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2012] [Accepted: 12/03/2012] [Indexed: 11/18/2022] Open
Abstract
In this study, we examined the expression of Sonic Hedgehog, Patched, Gli1, Gli2, Gli3 and Myocardin in the developing bladders of male and female normal and megabladder (mgb-/-) mutant mice at embryonic days 12 through 16 by in situ hybridization. This analysis indicated that each member of the Sonic Hedgehog signaling pathway as well as Myocardin displayed distinct temporal and spatial patterns of expression during normal bladder development. In contrast, mgb-/- bladders showed both temporal and spatial changes in the expression of Patched, Gli1 and Gli3 as well as a complete lack of Myocardin expression. These changes occurred primarily in the outer mesenchyme of developing mgb-/- bladders consistent with the development of an amuscular bladder phenotype in these animals. These results provide the first comprehensive analysis of the Sonic Hedgehog signaling pathway during normal bladder development and provide strong evidence that this key signaling cascade is critical in establishing radial patterning in the developing bladder. In addition, the lack of detrusor smooth muscle development observed in mgb-/- mice is associated with bladder-specific temporospatial changes in Sonic Hedgehog signaling coupled with a lack of Myocardin expression that appears to result in altered patterning of the outer mesenchyme and poor initiation and differentiation of smooth muscle cells within this region of the developing bladder.
Collapse
Affiliation(s)
- Kristin R DeSouza
- Center for Molecular and Human Genetics, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, United States of America.
| | | | | | | | | |
Collapse
|
16
|
Yoshio Y, Ishii K, Arase S, Hori Y, Nishikawa K, Soga N, Kise H, Arima K, Sugimura Y. Effect of transforming growth factor α overexpression on urogenital organ development in mouse. Differentiation 2010; 80:82-8. [PMID: 20638776 DOI: 10.1016/j.diff.2010.06.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2010] [Revised: 06/25/2010] [Accepted: 06/29/2010] [Indexed: 10/19/2022]
Abstract
Transforming growth factor-α (TGFα) promotes cell proliferation by binding to the epidermal growth factor receptor (EGFR). TGFα and EGFR overexpression have been reported in various human cancers. However, whether TGFα induces cancer by itself is unknown in urogenital organs. To investigate whether TGFα overexpression induces carcinogenesis in urogenital organs, we analyzed the phenotypes of urogenital organs in male TGFα transgenic (TG) mice of the CD1 strain. Urogenital organs including the kidney, bladder, prostate, seminal vesicles, testes, and epididymis were isolated from 4- to 48-week-old TGFα TG and wild-type (WT) CD1 mice. Prostates were separated into anterior prostate (AP), dorsolateral prostate (DLP), and ventral prostate (VP). Neither tumor formation nor epithelial hyperplasia was observed in the TGFα TG mouse urogenital organs that we have investigated. Histopathologically, in prostate, we found an increased number of p63-positive basal epithelial cells in the TGFα TG mice AP and DLP. There was no morphological change in the stromal component, such as hypercellular stroma or fibrosis. However, bladder weight was greater in TGFα TG mice than that in WT mice, and distended bladders were observed macroscopically in 19 of 20 TGFα TG mice over 20 weeks of age. Ki67 labeling index was increased significantly in the TGFα TG mouse urethral epithelium, whereas neither epithelial hyperplasia nor hypertrophy was observed. In conclusion, our results suggest that TGFα overexpression in mouse urogenital organs alone may not be responsible for tumor formation and epithelial hyperplasia, but is involved in bladder outlet obstruction.
Collapse
Affiliation(s)
- Yuko Yoshio
- Department of Nephro-Urologic Surgery and Andrology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie 514-8507, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Özel SK, Emir H, Dervişoğlu S, Akpolat N, Şenel B, Kazez A, Söylet Y, Çetin G, Danişmend N, Büyükünal SNC. The roles of extracellular matrix proteins, apoptosis and c-kit positive cells in the pathogenesis of ureteropelvic junction obstruction. J Pediatr Urol 2010; 6:125-9. [PMID: 19716769 DOI: 10.1016/j.jpurol.2009.07.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2009] [Accepted: 07/23/2009] [Indexed: 01/12/2023]
Abstract
AIM To investigate histopathological changes in ureteropelvic junction obstruction (UPJO) from an etiological perspective. PATIENTS AND METHODS Medical records of patients with UPJO were reviewed and pathological specimens collected. Nephrectomy materials from forensic autopsies were taken as controls. Specimens were assessed with light microscopy. Fibronectin, type 4 collagen, laminin, Bax and Bcl-2 expression for apoptosis, together with interstitial cells of Cajal determination with c-kit were determined immunohistochemically. Staining scores were evaluated semiquantitatively. Results were evaluated using Mann-Whitney U-test. RESULTS Control group comprised 14 children (median age, 3.5 years; 6 months-17 years). Study group comprised 22 children with UPJO (median age, 9 months; 1 month-10 years). Light microscopy revealed non-specific inflammation, epithelial proliferation and atrophy with fibrosis in the smooth muscle of the UPJ in all patients. Fibronectin, type 4 collagen and laminin were found to be significantly increased in UPJO at the intrafascicular space of smooth muscle and the matrix of stroma. Bcl-2 expression was increased in UPJO. c-Kit was unable to stain interstitial cells of Cajal, but staining for mast cells was significant. CONCLUSIONS High expression of fibronectin, laminin and type 4 collagen may indicate a relation to the pathogenesis of UPJO. Defective kidney morphogenesis, during branching and tubulogenesis of ureteric bud, may be responsible for this congenital pathology.
Collapse
Affiliation(s)
- S K Özel
- Fırat University Faculty of Medicine, Department of Pediatric Surgery, Elazığ, Turkey.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Urothelium-derived Sonic hedgehog promotes mesenchymal proliferation and induces bladder smooth muscle differentiation. Differentiation 2010; 79:244-50. [PMID: 20227816 DOI: 10.1016/j.diff.2010.02.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2009] [Revised: 02/12/2010] [Accepted: 02/18/2010] [Indexed: 10/19/2022]
Abstract
Induction of smooth muscle differentiation from bladder mesenchyme depends on signals that originate from the urothelium. We hypothesize Sonic hedgehog (Shh) is the urothelial signal that promotes bladder mesenchymal proliferation and induces bladder smooth muscle differentiation. Pregnant FVB mice were euthanized on embryonic day (E) 12.5 and fetal bladders were harvested. Two experimental protocols were utilized: Specimens were sized by serial sectioning. Cell counts were performed after trypsin digestion. Immunohistochemistry was performed to detect smooth muscle-specific protein expression. alpha-Actin expression was quantified using Western blot. All specimens were viable at 72h. BLM cultured without Shh survived but did not grow or undergo smooth muscle differentiation. IB cultured without Shh and BLM cultured with Shh grew and expressed smooth muscle proteins at 72h. IB cultured with Shh were larger and contained more cells than IB cultured without Shh (all p<0.05). Increasing Shh concentration from 48 to 480nM did not change bladder size, cell counts, or the level of alpha-actin expression. Prior to culture, IB did not express alpha-actin. After culture of IB in Shh-deficient media, alpha-actin was detected throughout the mesenchyme except in the submucosal layer. The IB submucosa was thinner after culture with 48nM Shh and smooth muscle completely obliterated the submucosa after culture with 480nM Shh. In fetal mouse bladders, urothelium-derived Shh is necessary for mesenchymal proliferation and smooth muscle differentiation. Shh concentration affects mesenchymal proliferation and patterning of bladder smooth muscle.
Collapse
|
19
|
Yang R, Amir J, Liu H, Chaqour B. Mechanical strain activates a program of genes functionally involved in paracrine signaling of angiogenesis. Physiol Genomics 2008; 36:1-14. [PMID: 18854370 PMCID: PMC2604786 DOI: 10.1152/physiolgenomics.90291.2008] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2008] [Accepted: 10/07/2008] [Indexed: 12/16/2022] Open
Abstract
Studies were performed to examine the extent to which mechanical stimuli mediate control of angiogenesis in bladder cells both in vitro and in vivo. Differential gene expression between control nonstretched and cyclically stretched bladder smooth muscle cells was assessed using oligonucleotide microarrays and pathway analysis by the web tool Fast Assignment and Transference of Information (FatiGO). Data showed that a substantial proportion (33 of 86) of mechanically responsive genes were angiogenesis-related and include cytokines, growth-related factors, adhesion proteins, and matricellular, signal transduction, extracellular matrix (ECM), and inflammatory molecules. Integrative knowledge of protein-protein interactions revealed that 12 mechano-sensitive gene-encoded proteins have interacting partner(s) in the vascular system confirming their potential role in paracrine regulation of angiogenesis. Angiogenic genes include matricellular proteins such as Cyr61/CCN1, CTGF/CCN2 and tenascin C, components of the VEGF and IGF systems, ECM proteins such as type I collagen and proteoglycans, and matrix metalloproteinases. In an in vivo model of bladder overdistension, 5 of 11 mechano-responsive angiogenic genes, independently tested by real-time PCR, were upregulated as a result of pressure overload including Cyr61/CCN1, CTGF/CCN2, MCP-1, VEGF-A, MMP-1, and midkine. Meanwhile, the molecular anatomy of angiogenic gene promoters reveals the presence of GA box-binding for the myc-associated zinc finger protein, MAZ, often found adjacent to binding sites for mechano-responsive transcription factors (e.g., NF-kappaB), suggesting that the coordinated activity of these factors may induce selective angiogenic gene transcription. These data suggest that mechanical control of angiogenic genes is an integral part of the adaptive and plasticity responses to mechanical overload.
Collapse
Affiliation(s)
- Ru Yang
- Department of Anatomy and Cell Biology, State University of New York Downstate Medical Center, Brooklyn, New York 11203-2098, USA
| | | | | | | |
Collapse
|
20
|
Anumanthan G, Makari JH, Honea L, Thomas JC, Wills ML, Bhowmick NA, Adams MC, Hayward SW, Matusik RJ, Brock JW, Pope JC. Directed differentiation of bone marrow derived mesenchymal stem cells into bladder urothelium. J Urol 2008; 180:1778-83. [PMID: 18721942 PMCID: PMC4802964 DOI: 10.1016/j.juro.2008.04.076] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2007] [Indexed: 12/21/2022]
Abstract
PURPOSE We have previously reported that embryonic rat bladder mesenchyma has the appropriate inductive signals to direct pluripotent mouse embryonic stem cells toward endodermal derived urothelium and develop mature bladder tissue. We determined whether nonembryonic stem cells, specifically bone marrow derived mesenchymal stem cells, could serve as a source of pluripotent or multipotent progenitor cells. MATERIALS AND METHODS Epithelium was separated from the mesenchymal shells of embryonic day 14 rat bladders. Mesenchymal stem cells were isolated from mouse femoral and tibial bone marrow. Heterospecific recombinant xenografts were created by combining the embryonic rat bladder mesenchyma shells with mesenchymal stem cells and grafting them into the renal subcapsular space of athymic nude mice. Grafts were harvested at time points of up to 42 days and stained for urothelial and stromal differentiation. RESULTS Histological examination of xenografts comprising mouse mesenchymal stem cells and rat embryonic rat bladder mesenchyma yielded mature bladder structures showing normal microscopic architecture as well as proteins confirming functional characteristics. Specifically the induced urothelium expressed uroplakin, a highly selective marker of urothelial differentiation. These differentiated bladder structures demonstrated appropriate alpha-smooth muscle actin staining. Finally, Hoechst staining of the xenografts revealed nuclear architecture consistent with a mouse mesenchymal stem cell origin of the urothelium, supporting differentiated development of these cells. CONCLUSIONS In the appropriate signaling environment bone marrow derived mesenchymal stem cells can undergo directed differentiation toward endodermal derived urothelium and develop into mature bladder tissue in a tissue recombination model. This model serves as an important tool for the study of bladder development with long-term application toward cell replacement therapies in the future.
Collapse
Affiliation(s)
- Govindaraj Anumanthan
- Departments of Urologic Surgery (GA, JHM, LH, JCT, NAB, MCA, SWH, RJM, JWB, JCP), Pathology (MLW) and Cancer Biology (NAB, SWH, JRM) and Vanderbilt-Ingram Cancer Center (NAB, SWH, JM), Vanderbilt University Medical Center and Division of Pediatric Urology, Vanderbilt Children’s Hospital (GA, JHM, LH, HCT, NAB, MCA, JWB, JCP), Nashville, Tennessee
| | - John H. Makari
- Departments of Urologic Surgery (GA, JHM, LH, JCT, NAB, MCA, SWH, RJM, JWB, JCP), Pathology (MLW) and Cancer Biology (NAB, SWH, JRM) and Vanderbilt-Ingram Cancer Center (NAB, SWH, JM), Vanderbilt University Medical Center and Division of Pediatric Urology, Vanderbilt Children’s Hospital (GA, JHM, LH, HCT, NAB, MCA, JWB, JCP), Nashville, Tennessee
| | - Lindsey Honea
- Departments of Urologic Surgery (GA, JHM, LH, JCT, NAB, MCA, SWH, RJM, JWB, JCP), Pathology (MLW) and Cancer Biology (NAB, SWH, JRM) and Vanderbilt-Ingram Cancer Center (NAB, SWH, JM), Vanderbilt University Medical Center and Division of Pediatric Urology, Vanderbilt Children’s Hospital (GA, JHM, LH, HCT, NAB, MCA, JWB, JCP), Nashville, Tennessee
| | - John C. Thomas
- Departments of Urologic Surgery (GA, JHM, LH, JCT, NAB, MCA, SWH, RJM, JWB, JCP), Pathology (MLW) and Cancer Biology (NAB, SWH, JRM) and Vanderbilt-Ingram Cancer Center (NAB, SWH, JM), Vanderbilt University Medical Center and Division of Pediatric Urology, Vanderbilt Children’s Hospital (GA, JHM, LH, HCT, NAB, MCA, JWB, JCP), Nashville, Tennessee
| | - Marcia L. Wills
- Departments of Urologic Surgery (GA, JHM, LH, JCT, NAB, MCA, SWH, RJM, JWB, JCP), Pathology (MLW) and Cancer Biology (NAB, SWH, JRM) and Vanderbilt-Ingram Cancer Center (NAB, SWH, JM), Vanderbilt University Medical Center and Division of Pediatric Urology, Vanderbilt Children’s Hospital (GA, JHM, LH, HCT, NAB, MCA, JWB, JCP), Nashville, Tennessee
| | - Neil A. Bhowmick
- Departments of Urologic Surgery (GA, JHM, LH, JCT, NAB, MCA, SWH, RJM, JWB, JCP), Pathology (MLW) and Cancer Biology (NAB, SWH, JRM) and Vanderbilt-Ingram Cancer Center (NAB, SWH, JM), Vanderbilt University Medical Center and Division of Pediatric Urology, Vanderbilt Children’s Hospital (GA, JHM, LH, HCT, NAB, MCA, JWB, JCP), Nashville, Tennessee
| | - Mark C. Adams
- Departments of Urologic Surgery (GA, JHM, LH, JCT, NAB, MCA, SWH, RJM, JWB, JCP), Pathology (MLW) and Cancer Biology (NAB, SWH, JRM) and Vanderbilt-Ingram Cancer Center (NAB, SWH, JM), Vanderbilt University Medical Center and Division of Pediatric Urology, Vanderbilt Children’s Hospital (GA, JHM, LH, HCT, NAB, MCA, JWB, JCP), Nashville, Tennessee
| | - Simon W. Hayward
- Departments of Urologic Surgery (GA, JHM, LH, JCT, NAB, MCA, SWH, RJM, JWB, JCP), Pathology (MLW) and Cancer Biology (NAB, SWH, JRM) and Vanderbilt-Ingram Cancer Center (NAB, SWH, JM), Vanderbilt University Medical Center and Division of Pediatric Urology, Vanderbilt Children’s Hospital (GA, JHM, LH, HCT, NAB, MCA, JWB, JCP), Nashville, Tennessee
| | - Robert J. Matusik
- Departments of Urologic Surgery (GA, JHM, LH, JCT, NAB, MCA, SWH, RJM, JWB, JCP), Pathology (MLW) and Cancer Biology (NAB, SWH, JRM) and Vanderbilt-Ingram Cancer Center (NAB, SWH, JM), Vanderbilt University Medical Center and Division of Pediatric Urology, Vanderbilt Children’s Hospital (GA, JHM, LH, HCT, NAB, MCA, JWB, JCP), Nashville, Tennessee
| | - John W. Brock
- Departments of Urologic Surgery (GA, JHM, LH, JCT, NAB, MCA, SWH, RJM, JWB, JCP), Pathology (MLW) and Cancer Biology (NAB, SWH, JRM) and Vanderbilt-Ingram Cancer Center (NAB, SWH, JM), Vanderbilt University Medical Center and Division of Pediatric Urology, Vanderbilt Children’s Hospital (GA, JHM, LH, HCT, NAB, MCA, JWB, JCP), Nashville, Tennessee
| | - John C. Pope
- Departments of Urologic Surgery (GA, JHM, LH, JCT, NAB, MCA, SWH, RJM, JWB, JCP), Pathology (MLW) and Cancer Biology (NAB, SWH, JRM) and Vanderbilt-Ingram Cancer Center (NAB, SWH, JM), Vanderbilt University Medical Center and Division of Pediatric Urology, Vanderbilt Children’s Hospital (GA, JHM, LH, HCT, NAB, MCA, JWB, JCP), Nashville, Tennessee
| |
Collapse
|
21
|
Sonic Hedgehog Mediator Gli2 Regulates Bladder Mesenchymal Patterning. J Urol 2008; 180:1543-50. [DOI: 10.1016/j.juro.2008.06.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2007] [Indexed: 01/24/2023]
|
22
|
Abstract
Smooth muscle differentiation is induced in the embryonic bladder by the centrally located urothelium in the undifferentiated mesenchyme in the periphery adjacent to the serosa. We hypothesize that under the appropriate signal the entire undifferentiated bladder mesenchyme is capable of smooth muscle differentiation and that the urothelium patterns fibromuscular development. Embryonic bladders of wild-type and Green Fluorescent Protein mice were separated into urothelial and mesenchymal components before smooth muscle differentiation (E12.5-E13). The urothelial layer green fluorescent protein was recombined and grafted with the mesenchyme (wild-type) in an orthotopic position, heterotopic position and ectopic position. In all cases, a zone of smooth muscle inhibition was observed adjacent to the epithelium whether the urothelium was in an orthotopic or heterotypic position. Bladder mesenchyme and bladder epithelium grafted alone did not grow. In conclusion, the full thickness of bladder mesenchyme is capable of smooth muscle differentiation dependent on the location of urothelium. These experiments support the hypothesis that urothelium secretes a diffusible factor that at high concentrations inhibits smooth muscle and at low concentrations induces smooth muscle, thus patterning mesenchymal cell differentiation across the full thickness of the fibromuscular bladder wall.
Collapse
Affiliation(s)
- Mei Cao
- Department of Urology, University of California, San Francisco, California 94143, USA
| | | | | | | |
Collapse
|
23
|
Oottamasathien S, Williams K, Franco OE, Wills ML, Thomas JC, Sharif-Afshar AR, DeMarco RT, Brock JW, Bhowmick NA, Hayward SW, Pope JC. Urothelial Inhibition of Transforming Growth Factor-β in a Bladder Tissue Recombination Model. J Urol 2007; 178:1643-9. [PMID: 17707033 DOI: 10.1016/j.juro.2007.03.163] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2006] [Indexed: 11/21/2022]
Abstract
PURPOSE We examined the role of transforming growth factor-beta in urothelial and bladder development. Transforming growth factor-beta signaling was attenuated in the urothelial compartment and the subsequent effects were examined in a tissue recombination model. MATERIALS AND METHODS Urothelium was cultured from adult rat bladders and transfected with control vector C7Delta or mutant DNIIR (dominant negative transforming growth factor-beta receptor II). Grafts were created by recombining transfected urothelium plus embryonic day 18 bladder mesenchyma and placed beneath the renal capsule of athymic mouse hosts. Grafts were harvested at 21 and 42 days. Final tissues were evaluated with staining and immunohistochemistry using hematoxylin and eosin, Gomori's trichrome strain, broad-spectrum uroplakin, smooth muscle actin-alpha, phosphorylated SMAD2 and Ki67 antigen. Bladder structures were defined as having smooth muscle, suburothelial connective tissue and mature urothelium expressing uroplakin. Urothelial compartment diameters were measured and subcategorized as small--0.10 to 0.40, medium--0.41 to 1.0 and large--greater than 1.1 mm. RESULTS At 21 days 14 C7Delta control and 15 DNIIR grafts were evaluated. No bladder tissue was seen in the C7Delta grafts vs 49 in DNIIR tissue, including 30 small, 9 medium and 10 large tissues. At 42 days 14 C7Delta and 12 DNIIR grafts were evaluated. Six bladder structures (5 small and 1 medium) were seen in the C7Delta cohort vs 27 (14 small, 7 medium and 6 large) in the DNIIR group. Immunohistochemical detection of phosphorylated-SMAD2 was significantly attenuated in DNIIR tissue. In addition, Ki67 proliferative indexes were 4.0-fold higher in the DNIIR cohort compared to those in C7Delta tissues. CONCLUSIONS We successfully observed that primary urothelium cultures can be genetically manipulated and recombined with undifferentiated mesenchyma to grow bladder tissue. By attenuating transforming growth factor-beta signaling in the urothelium superior bladder tissue growth occurred, suggesting that transforming growth factor-beta is a growth inhibitor in this organ system.
Collapse
Affiliation(s)
- Siam Oottamasathien
- Division of Pediatric Urology, Department of Urologic Surgery, Vanderbilt Children's Hospital, Nashville, Tennessee 37232-2765, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Lendvay TS, Sweet R, Han CH, Soygur T, Cheng JF, Plaire JC, Charleston JS, Charleston LB, Bagai S, Cochrane K, Rubio E, Bassuk JA. Compensatory paracrine mechanisms that define the urothelial response to injury in partial bladder outlet obstruction. Am J Physiol Renal Physiol 2007; 293:F1147-56. [PMID: 17609292 DOI: 10.1152/ajprenal.00006.2007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Diseases and conditions affecting the lower urinary tract are a leading cause of dysfunctional sexual health, incontinence, infection, and kidney failure. The growth, differentiation, and repair of the bladder's epithelial lining are regulated, in part, by fibroblast growth factor (FGF)-7 and -10 via a paracrine cascade originating in the mesenchyme (lamina propria) and targeting the receptor for FGF-7 and -10 within the transitional epithelium (urothelium). The FGF-7 gene is located at the 15q15-q21.1 locus on chromosome 15 and four exons generate a 3.852-kb mRNA. Five duplicated FGF-7 gene sequences that localized to chromosome 9 were predicted not to generate functional protein products, thus validating the use of FGF-7-null mice as an experimental model. Recombinant FGF-7 and -10 induced proliferation of human urothelial cells in vitro and transitional epithelium of wild-type and FGF-7-null mice in vivo. To determine the extent that induction of urothelial cell proliferation during the bladder response to injury is dependent on FGF-7, an animal model of partial bladder outlet obstruction was developed. Unbiased stereology was used to measure the percentage of proliferating urothelial cells between obstructed groups of wild-type and FGF-7-null mice. The stereological analysis indicated that a statistical significant difference did not exist between the two groups, suggesting that FGF-7 is not essential for urothelial cell proliferation in response to partial outlet obstruction. In contrast, a significant increase in FGF-10 expression was observed in the obstructed FGF-7-null group, indicating that the compensatory pathway that functions in this model results in urothelial repair.
Collapse
Affiliation(s)
- Thomas S Lendvay
- Program in Human Urothelial Biology, Seattle Children's Hospital Research Institute, Seattle, WA 98105, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Oottamasathien S, Williams K, Franco OE, Thomas JC, Saba K, Bhowmick NA, Staack A, Demarco RT, Brock JW, Hayward SW, Pope JC. Bladder tissue formation from cultured bladder urothelium. Dev Dyn 2007; 235:2795-801. [PMID: 16804891 DOI: 10.1002/dvdy.20886] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Tissue recombination is a powerful method to evaluate the paracrine-signaling events that orchestrate the development of organs using the in vivo environment of a host rodent. Studies have reported the successful generation of primary cultures of rodent bladder urothelium, but none have reported their use to recapitulate bladder tissue with tissue recombination. We propose that primary cultured bladder urothelium, when recombined with inductive embryonic bladder mesenchyme, will form bladder tissue in a recombination model. Adult rat bladders were isolated and urothelium obtained. Sheets of bladder urothelium were re-suspended in collagen and maintained in tissue culture. After expansion (>20 passages), the urothelium was recombined with embryonic day-14 mouse bladder mesenchyme, then grafted beneath the renal capsule of immunocompromised mouse hosts. Grafts were harvested after 28 days. Control grafts were performed with bladder mesenchyme alone, cultured bladder urothelium alone, and collagen matrix alone. Final tissues were evaluated with staining and immunohistochemistry (H&E, Gomori's trichrome, broad-spectrum uroplakin, and smooth muscle actin alpha and gamma). Immunocytochemistry on cultured urothelium for broad-spectrum keratin, vimentin, and broad-spectrum uroplakin confirmed pure populations, void of mesenchymal contaminants. Staining of recombinant grafts demonstrated bladder tissue with mature urothelium and stromal differentiation. Control tissues were void of bladder tissue formation. We have successfully demonstrated that a chimeric bladder is formed from primary cultured bladder urothelium recombined with embryonic bladder mesenchyme. This is a powerful new tool for investigating the molecular mechanisms of bladder development and disease. Future applications may include the in vitro genetic manipulation of urothelium and examining those effects on growth and development in an in vivo environment.
Collapse
Affiliation(s)
- Siam Oottamasathien
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Ersoy Y, Ercan F, Cetinel S. A comparative ontogenic study of urinary bladder: impact of the epithelial differentiation in embryonic and newborn rats. Anat Histol Embryol 2006; 35:365-74. [PMID: 17156089 DOI: 10.1111/j.1439-0264.2006.00699.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The present study aimed to show the cellular and subcellular distribution of glycogen content during the differentiation of urothelial cells from simple cuboidal to stratified transitional epithelium. Bladder samples were taken from rat embryos on the 15th to 19th days and newborn at 21st day. During the development of the bladder, the formation of fusiform vesicles, asymmetric unit membrane (AUM) and microridges were examined with staining with haematoxylin-eosin and periodic acid Schiff for light microscope and periodic acid-thiocharbohydrazide-silver proteinate for transmission electron microscope. The topographical changes of luminal differentiation were examined with the scanning electron microscope. The urothelium was simple cuboidal from 15th till the 17th days of gestation. Glycogen content was present in the cytoplasm till the 18th day of gestation. At the early stage (16th day) of gestation, the apical surface contains microvilli that points the undifferentiated cells. The density of microvilli decreased and ropy microridges appeared at the 17th day of gestation. The small discoid vesicles lined with AUM developed at the apical cytoplasm of the surface cells at the 17th day of gestation. After this stage, both the density of microridges and large and elongated fusiform vesicles increased. The differentiation of the urothelium begins with the formation of the round and small vesicles, continues with the formation of the AUM and at the final stage there is a decrease in both glycogen content and the appearance of the microridges at the luminal surface of the urothelial cells.
Collapse
Affiliation(s)
- Y Ersoy
- Department of Histology-Embryology, School of Medicine, Marmara University, 34668 Haydarpaşa, Istanbul, Turkey
| | | | | |
Collapse
|
27
|
Cheng W, Jacobs WB, Zhang JJR, Moro A, Park JH, Kushida M, Qiu W, Mills AA, Kim PCW. DeltaNp63 plays an anti-apoptotic role in ventral bladder development. Development 2006; 133:4783-92. [PMID: 17079275 DOI: 10.1242/dev.02621] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The bladder, the largest smooth-muscle organ in the human body, is responsible for urine storage and micturition. P63, a homolog of the p53 tumor-suppressor gene, is essential for the development of all stratified epithelia, including the bladder urothelium. The N-terminal truncated isoform of p63, DeltaNp63, is known to have anti-apoptotic characteristics. We have established that DeltaNp63 is not only the predominant isoform expressed throughout the bladder, but is also preferentially expressed in the ventral bladder urothelium during early development. We observed a host of ventral defects in p63-/- embryos, including the absence of the abdominal and ventral bladder walls. This number of ventral defects is identical to bladder exstrophy, a congenital anomaly exhibited in human neonates. In the absence of p63, the ventral urothelium was neither committed nor differentiated, whereas the dorsal urothelium was both committed and differentiated. Furthermore, in p63-/- bladders, apoptosis in the ventral urothelium was significantly increased. This was accompanied by the upregulation of mitochondrial apoptotic mediators Bax and Apaf1, and concurrent upregulation of p53. Overexpression of DeltaNp63gamma and DeltaNp63beta in p63-/- bladder primary cell cultures resulted in a rescue, evidenced by significantly reduced expressions of Bax and Apaf1. We conclude that DeltaNp63 plays a crucial anti-apoptotic role in normal bladder development.
Collapse
Affiliation(s)
- Wei Cheng
- Department of Surgery, Hospital for Sick Children, Toronto, M5G 1X8, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Bush KT, Vaughn DA, Li X, Rosenfeld MG, Rose DW, Mendoza SA, Nigam SK. Development and differentiation of the ureteric bud into the ureter in the absence of a kidney collecting system. Dev Biol 2006; 298:571-84. [PMID: 16934795 DOI: 10.1016/j.ydbio.2006.07.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2006] [Revised: 06/30/2006] [Accepted: 07/10/2006] [Indexed: 02/06/2023]
Abstract
Six1-/- mice were found to have apparently normal ureters in the absence of a kidney, suggesting that the growth and development of the unbranched ureter is largely independent of the more proximal portions of the UB which differentiates into the highly branched renal collecting system. Culture of isolated urinary tracts (from normal and mutant mice) on Transwell filters was employed to study the morphogenesis of this portion of the urogenital system. Examination of the ureters revealed the presence of a multi-cell layered tubule with a lumen lined by cells expressing uroplakin (a protein exclusively expressed in the epithelium of the lower urinary tract). Cultured ureters of both the wild-type and Six1 mutant become contractile and undergo peristalsis, an activity preceded by the expression of alpha-smooth muscle actin (alphaSMA). Treatment with a number of inhibitors of signaling molecules revealed that inhibition of PI3 kinase dissociates the developmental expression of alphaSMA from ureter growth and elongation. Epidermal growth factor also perturbed smooth muscle differentiation in culture. Moreover, the peristalsis of the ureter in the absence of the kidney in the Six1-/- mouse indicates that the development of this clinically important function of ureter (peristaltic movement of urine) is not dependent on fluid flow through the ureter. In keeping with this, isolated ureters cultured in the absence of surrounding tissues elongate, differentiate and undergo peristalsis when cultured on a filter and undergo branching morphogenesis when cultured in 3-dimensional extracellular matrix gels in the presence of a conditioned medium derived from a metanephric mesenchyme (MM) cell line. In addition, ureters of Six1-/- urinary tracts (i.e., lacking a kidney) displayed budding structures from their proximal ends when cultured in the presence of GDNF and FGFs reminiscent of UB budding from the wolffian duct. Taken together with the above data, this indicates that, although the distal ureter (at least early in its development) retains some of the characteristics of the more proximal UB, the growth and differentiation (i.e., development of smooth muscle actin, peristalsis and uroplakin expression) of the distal non-branching ureter are inherent properties of this portion of the UB, occurring independently of detectable influences of either the undifferentiated MM (unlike the upper portion of the ureteric bud) or more differentiated metanephric kidney. Thus, the developing distal ureter appears to be a unique anatomical structure which should no longer be considered as simply the non-branching portion of the ureteric bud. In future studies, the ability to independently analyze and study the portion of the UB that becomes the renal collecting system and that which becomes the ureter should facilitate distinguishing the developmental nephrome (renal ontogenome) from the ureterome.
Collapse
Affiliation(s)
- Kevin T Bush
- Department of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0693, and Urological Diseases Research Center, Department of Urology, Children's Hospital, Boston, MA 02115, USA
| | | | | | | | | | | | | |
Collapse
|
29
|
Howard PS, Kucich U, Coplen DE, He Y. Transforming growth factor-beta1–induced hypertrophy and matrix expression in human bladder smooth muscle cells. Urology 2005; 66:1349-53. [PMID: 16360482 DOI: 10.1016/j.urology.2005.06.124] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2005] [Revised: 06/01/2005] [Accepted: 06/24/2005] [Indexed: 12/01/2022]
Abstract
OBJECTIVES To determine whether transforming growth factor beta (TGF-beta) could activate hyperplasia, hypertrophy, and altered collagen expression in human detrusor smooth muscle cells (SMCs). METHODS Human bladder SMCs were treated in vitro with TGF-beta1 and analyzed for changes in both proliferative and hypertrophic responses by cell number and volume measurements, as well as for alterations in extracellular matrix gene and protein expression by Northern blot and enzyme-linked immunosorbent assay. RESULTS Proliferation of bladder SMCs was refractory to TGF-beta1, whereas the cells became hypertrophic upon TGF-beta1 treatment. The interstitial collagens, types I and III, were increased significantly in TGF-beta1-treated cultures in a dose-dependent manner. These increases were blocked in the presence of TGF-beta1 neutralizing antibody and also when cultures were treated with the protein synthesis inhibitor cycloheximide, indicating that new protein synthesis is necessary for upregulation of the interstitial collagens. Messenger ribonucleic acid transcripts for both the COL1A1 and COL3A1 genes were elevated at 4, 6, and 24 hours in TGF-beta1-treated cultures, preceding the expression of the collagenous protein, showing that TGF-beta1 effects on bladder smooth muscle occur, at least in part, at the transcriptional level. CONCLUSIONS These results indicate that human bladder SMCs have the potential to mediate both a hypertrophic and fibrotic response upon TGF-beta1 stimulation.
Collapse
Affiliation(s)
- Pamela S Howard
- Department of Anatomy and Cell Biology, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6030, USA.
| | | | | | | |
Collapse
|
30
|
Poladia DP, Bauer JA. Early cell-specific changes in nitric oxide synthases, reactive nitrogen species formation, and ubiquitinylation during diabetes-related bladder remodeling. Diabetes Metab Res Rev 2003; 19:313-9. [PMID: 12879409 DOI: 10.1002/dmrr.385] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
BACKGROUND Urinary bladder dysfunction in diabetes is a well-recognized phenomenon but the mechanisms involved and initiating events are not clear. The physiological production of nitric oxide (NO) plays an important role in bladder tone and local immune defense, and recent studies have shown that NO-derived reactive nitrogen species occur in many settings of chronic disease, including other diabetic complications. Here, we investigated the early time-dependent and cell-specific changes in the nitric oxide synthase isoforms (NOS1, 2, and 3), peroxynitrite, and ubiquitinylation in the well-documented streptozotocin-induced rat model of diabetes. METHODS Immunohistochemical methods and automated digital imaging were used for the measurement of morphometric and histochemical analysis of the bladder tissue regions. Region-specific 3-nitrotyrosine (a biomarker of NO dysregulation and reactive nitrogen species formation) and ubiquitinylated protein prevalence (marker of proteasomal activity) were also investigated. RESULTS Immunohistochemistry revealed early, time-dependent, and cell-specific alterations in the three isoforms of NOS. We also observed region-specific increases in protein nitration, demonstrating first-time evidence of reactive nitrogen species formation in this setting. The changes in nitration did not pattern changes in NOS2 induction or tissue ubiquitinylation, and these alterations preceded any detectable changes in bladder structure (3 days vs 2 weeks) in this same animal preparation. CONCLUSIONS These data demonstrate that selective and regionally distinct changes in nitric oxide production and impaired nitric oxide control are early events during diabetic cystopathy and that mechanisms leading to increased oxidative stress and proteasomal activation may be key participants leading to organ dysfunction in this setting.
Collapse
Affiliation(s)
- Deepali Pitre Poladia
- Division of Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio, USA
| | | |
Collapse
|
31
|
Brown AL, Farhat W, Merguerian PA, Wilson GJ, Khoury AE, Woodhouse KA. 22 week assessment of bladder acellular matrix as a bladder augmentation material in a porcine model. Biomaterials 2002; 23:2179-90. [PMID: 11962659 DOI: 10.1016/s0142-9612(01)00350-7] [Citation(s) in RCA: 109] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Previous studies on the reconstruction of porcine bladder using bladder acellular matrix allograft (BAMA) have indicated positive preliminary results with respect to graft shrinkage and cellular repopulation. The current study was conducted to investigate the feasibility of using BAMA in a similar model of bladder reconstruction out to longer time frames (22 weeks). At predetermined time points, the macroscopic, histological and mechanical properties of explanted native and BAMA tissues were evaluated and compared. Macroscopically, contracture of the BAMA was observed. The peripheral regions of the grafts experienced extensive cellular repopulation. Towards the centre however, all grafts were consistently devoid of organized smooth muscle bundles and a well-developed urothelium. An alteration in both the amount and organization of collagen was also observed within this region. Significant differences (p < 0.05) in the rupture strain and the elastic modulus of the BAMA compared to native bladder tissue appear to correlate with macroscopic graft contracture as well as the fibroproliferative tissue response of the matrix.
Collapse
Affiliation(s)
- A L Brown
- Department of Chemical Engineering and Applied Chemistry, Institute for Biomaterials and Biomedical Engineering, University of Toronto, Ont., Canada
| | | | | | | | | | | |
Collapse
|
32
|
Abstract
OBJECTIVES To determine the anatomical steps leading to bladder agenesis in rats prenatally exposed as fetuses on gestational days (GD) 6-9 to adriamycin. MATERIALS AND METHODS Timed-pregnant Sprague-Dawley rats were injected intraperitoneally with adriamycin at 2 mg/kg (n = 28) on GD 6-9 (vaginal plug = day 0). The control group (n = 21) received saline. Fetuses were harvested on GD 10, 11, 12, 13, 14, 15 and 16. Serial paraffin sections were prepared from a minimum of 10 experimental and five control fetuses at each gestational age, and stained with either trichrome or haematoxylin and eosin, and examined by light microscopy. RESULTS In the control group the urorectal septum first became visible and the mesonephric ducts apparently abutting the anterior cloaca on GD 12. The presumptive urinary bladder was clearly defined on GD 14. On GD 15, the common excretory ducts became incorporated into the newly formed urogenital sinus and the ureters opened into the bladder. In the treated animals, beginning on GD 11, the undivided cloaca was noticeably smaller and by GD 13-14, the vesical extension of the urogenital sinus was conspicuously absent. Instead, opposite ureters joined to drain directly into the proximal blind-ending urethra or the persistent distal urogenital sinus. CONCLUSIONS Prenatal exposure of rat fetuses to adriamycin resulted in primary agenesis rather than secondary resorption of the bladder. The ontogeny showed that the mechanism underpinning bladder development is unique and is under the influence of factors that can be targeted by adriamycin. Further work will elucidate the unique nature of bladder organogenesis and should have important applications in future research into artificial bladders.
Collapse
Affiliation(s)
- M I Liu
- F. Douglas Stephens Surgical Research Laboratory, Murdoch Children's Hospital Research Institute, Melbourne, Australia
| | | |
Collapse
|
33
|
Dahms SE, Piechota HJ, Dahiya R, Gleason CA, Hohenfellner M, Tanagho EA. Bladder acellular matrix graft in rats: its neurophysiologic properties and mRNA expression of growth factors TGF-alpha and TGF-beta. Neurourol Urodyn 2000; 17:37-54. [PMID: 9453691 DOI: 10.1002/(sici)1520-6777(1998)17:1<37::aid-nau7>3.0.co;2-e] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
To assess the neurophysiologic properties and molecular mechanisms of the bladder acellular matrix graft (BAMG), we performed cystometric and neurophysiologic studies in male Sprague-Dawley rats (n = 46) at varying intervals. The animals were assigned to 3 groups: 1) normal, 2) partial cystectomy (>50%), and 3) partial cystectomy (>50%) and grafting with a BAMG of equal size. Additionally, matrix-grafted and host bladders were processed for analysis of mRNA expression of transforming growth factor (TGF)-alpha, TGF-beta1, TGF-beta2, and TGF-beta3 by reverse transcriptase polymerase chain reaction. Matrix-grafted bladders showed a significantly higher bladder capacity at 3 and 6 weeks and 4 months than those with partial cystectomy alone, and a significantly higher bladder capacity at 4 months than in normal controls (P < or = 0.01). Residual urine volume was significantly increased at 4 months. Electrostimulation of the pelvic nerve provoked generalized bladder contractions, a response that was reduced by atropine and hexamethonium. Variable induction of TGF-alpha, TGF-beta1, TGF-beta2, and TGF-beta3 gene transcription was evident in the BAMG, with prominent mRNA expression of TGF-alpha and TGF-beta1 6 months after surgery. These cystometric results and detrusor responses to stimulation provide further evidence that graft components do not interfere with host components. Matrix-grafted rat bladders generate, although not increased over time, adequate intravesical pressure responses to produce sustained voiding. Gene expression of different growth factors may be significant in understanding their role in the development and differentiation of the BAMG for partial bladder replacement.
Collapse
Affiliation(s)
- S E Dahms
- Department of Urology, University of California School of Medicine, San Francisco, USA
| | | | | | | | | | | |
Collapse
|
34
|
Adam RM, Borer JG, Williams J, Eastham JA, Loughlin KR, Freeman MR. Amphiregulin is coordinately expressed with heparin-binding epidermal growth factor-like growth factor in the interstitial smooth muscle of the human prostate. Endocrinology 1999; 140:5866-75. [PMID: 10579352 DOI: 10.1210/endo.140.12.7221] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Peptide growth factors have been proposed as mediators of smooth muscle-epithelial cell interactions in the human prostate; however, the identity of these molecules has not been established. In this study, we compared expression levels of messenger RNAs (mRNAs) encoding the epidermal growth factor (EGF) receptor-related receptor tyrosine kinases (ErbB1 through 4), the six EGF receptor ligands, EGF, transforming growth factor (TGF)-alpha, amphiregulin (ARG), HB-EGF, betacellulin, and epiregulin, and the related molecule heregulin-alpha, in a series of 10 prostate tissue specimens. Only EGF showed a disease-specific association, with increased mRNA levels in four of five PCa specimens in comparison to matched normal tissue from the same subject. In contrast, ARG and HB-EGF mRNAs showed a coordinate pattern of expression in 7/10 specimens that was distinct from all other growth factor or receptor genes examined and from mRNAs for prostate specific antigen, the androgen receptor and GAPDH, a house-keeping enzyme. Analysis of an additional series of benign prostatic hyperplasia and prostate cancer specimens from 60 individuals confirmed that ARG and HB-EGF mRNA levels varied in a highly coordinate manner (r = 0.93; P < 0.0001) but showed no association with disease. ARG was immunolocalized largely to interstitial smooth muscle cells (SMC), previously identified as the site of synthesis of HB-EGF in the prostate, while the cognate ARG and HB-EGF receptor, ErbB1, was localized exclusively to ductal epithelial cells and carcinoma cells. Although ARG was a relatively poor mitogen for Balb/c3T3 cells in comparison to HB-EGF, it was similar in potency to HB-EGF in stimulating human prostate epithelial cell growth, suggesting that prostate epithelia may be a physiologic target for ARG in vivo. Expression of both ARG and HB-EGF mRNAs was induced in cultured prostate SMC by fibroblast growth factor-2, a human prostate SMC mitogen linked to prostate disease. These findings indicate that ARG and HB-EGF are likely to be key mediators of directional signaling between SMC and epithelial cells in the human prostate and appear to be coordinately regulated.
Collapse
Affiliation(s)
- R M Adam
- Department of Urology, Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | | | | | |
Collapse
|
35
|
|
36
|
|
37
|
|
38
|
Abstract
It took more than half of a century for urologists to recognize that hydronephrosis is not necessarily equivalent to obstruction. Keeping this important truism in mind, particularly when dealing with antenatal hydronephrosis, one must also remember that hydronephrosis is not a normal condition. It is conceivable that although the initial intrinsic stenosis or ureterovascular obstruction may not be clinically significant in terms of renal functional damage, as compensatory renal pelvic dilatation develops, secondary obstructive elements may be recruited to create an insertional anomaly and peripelvic fibrosis. The individual types of UPJ obstruction that are seen in diagnostic studies or on the operating table may represent isolated "snapshots" of evolving pathophysiologic processes. If this is true, patients with asymptomatic congenital hydronephrosis, although lacking obvious renal function loss, require long-term follow-up.
Collapse
Affiliation(s)
- J M Park
- Department of Urology, University of Michigan, School of Medicine, Ann Arbor, USA
| | | |
Collapse
|
39
|
Parrott JA, Skinner MK. Developmental and hormonal regulation of keratinocyte growth factor expression and action in the ovarian follicle. Endocrinology 1998; 139:228-35. [PMID: 9421419 DOI: 10.1210/endo.139.1.5680] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The developing ovarian follicle is one of the most rapidly proliferating normal tissues in vivo. Mesenchymal-epithelial cell interactions between theca cells and granulosa cells are essential for this follicular expansion. Ovarian hormones (i.e. estrogen and LH) may promote follicular development by regulating the local production of mesenchymal inducer proteins that mediate theca cell-granulosa cell interactions. Recently, theca cells were shown to produce keratinocyte growth factor (KGF) that can act in a paracrine manner to stimulate granulosa cell growth. In this study, the developmental and hormonal regulation of KGF was examined during follicular development in the bovine ovary. Expression of KGF in theca cells and the KGF receptor (KGFR, or splice variant of the fibroblast growth factor family receptor family, FGFR-2) in granulosa cells was examined using RT-PCR. Both KGF and KGFR were detected throughout follicular development in small (<5 mm), medium (5-10 mm), and large (>10 mm) follicles. Quantitative RT-PCR assays were used to determine steady-state levels of KGF and KGFR messenger RNAs. Developmental regulation of KGF and KGFR was analyzed in freshly isolated theca cells and granulosa cells from small, medium, and large follicles. Observations demonstrated that expression of KGF (in theca cells) and KGFR (in granulosa cells) was highest in large follicles. These results suggest that KGF actions are important for the rapid proliferation of granulosa cells in large follicles. Estrogen and LH are the primary endocrine hormones that regulate theca cell function in vivo. Therefore, hormonal regulation of KGF was analyzed by treating serum-free theca cell cultures with estrogen and human CG (hCG, an LH agonist). Results showed that both estrogen and hCG stimulated KGF gene expression in theca cells. These results suggest that estrogen and LH may promote follicular growth (i.e. granulosa cell proliferation), in part, by stimulating the local production of KGF. Effects of KGF on granulosa cell differentiated functions were examined. Treatment with KGF reduced basal levels and FSH-stimulated levels of aromatase activity in bovine and rat granulosa cells. In addition, KGF inhibited the ability of hCG to stimulate progesterone production by granulosa cells. The inhibition of granulosa cell steroid production by KGF was likely the indirect effect of promoting cellular proliferation. Therefore, KGF directly stimulates granulosa cell proliferation and indirectly inhibits granulosa cell differentiated functions. Combined results suggest that theca cell production of KGF may be important for ovarian folliculogenesis. This is the first report of the regulation of KGF expression in the ovary. The developmental and hormonal regulation of KGF and KGFR during folliculogenesis provides evidence that KGF may be important for hormone-induced granulosa cell proliferation. As a result, KGF may be essential for establishing the microenvironment required for oocyte maturation in the ovary.
Collapse
Affiliation(s)
- J A Parrott
- Reproductive Endocrinology Center, University of California, San Francisco 94143-0556, USA
| | | |
Collapse
|