Sleeping beauty: awakening urothelium from its slumber REVIEW Sleeping beauty: awakening urothelium from its slumber Zarine R. Balsara1,2 and Xue Li1,2 1Department of Urology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts; and 2Department of Surgery, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts Submitted 17 June 2016; accepted in final form 22 January 2017 Balsara ZR, Li X. Sleeping beauty: awakening urothelium from its slumber. Am J Physiol Renal Physiol 312: F732–F743, 2017. First published January 25, 2017; doi:10.1152/ajprenal.00337.2016.—The bladder urothelium is essentially quiescent but regenerates readily upon injury. The process of urothelial regeneration harkens back to the process of urothelial development whereby urothelial stem/progenitor cells must proliferate and terminally differentiate to establish all three urothelial layers. How the urothelium regulates the level of proliferation and the timing of differentiation to ensure the precise degree of regeneration is of significant interest in the field. Without a carefully-orchestrated process, urothelial regeneration may be inadequate, thereby exposing the host to toxins or pathogens. Alternatively, regeneration may be excessive, thereby setting the stage for tumor development. This review describes our current understanding of urothelial regeneration. The current controversies surrounding the identity and location of urothelial progenitor cells that mediate urothelial regeneration are discussed and evidence for each model is provided. We emphasize the factors that have been shown to be crucial for urothelial regeneration, including local growth factors that stimulate repair, and epithelial-mesenchymal cross talk, which ensures feedback regulation. Also high- lighted is the emerging concept of epigenetic regulation of urothelial regeneration, which additionally fine tunes the process through transcriptional regulation of cell cycle genes and growth and differentiation factors. Finally, we emphasize how several of these pathways and/or programs are often dysregulated during malignant transformation, further corroborating their importance in directing normal urothe- lial regeneration. Together, evidence in the field suggests that any attempt to exploit regenerative programs for the purposes of enhanced urothelial repair or replace- ment must take into account this delicate balance. urothelium; regeneration; progenitor cells; superficial cells; label retention; lineage tracing; epithelial-mesenchymal cross talk; epigenetics THE UROTHELIUM IS A UNIQUE EPITHELIAL SURFACE that lines most of the genitourinary tract, including the renal pelvis, ureters, bladder, and proximal urethra. Urothelium consists of multiple layers of epithelial cells that can change size and shape to accommodate fluctuating volumes of urine. This mucosal ep- ithelial surface also serves as a barrier to prevent absorption of toxic substances like acid and urea from the urine and to defend against entry of pathogens from the external environment (30). Implicit in this latter function is the ability of urothelium to “sense” and respond to the presence of pathogens within the genitourinary tract (88). Coordinating other cues from the external environment, such as chemical, thermal, and mechan- ical stimuli, requires an additional layer of sophistication (38). Besides direct expression of neuronal sensory receptors and ion channels on urothelial cells and their ability to release chemi- cals and neurotransmitters, afferent nerves also innervate the detrusor muscle and extend into the urothelial layer to help the bladder respond to external stimuli (8, 9, 21, 64, 107). Urothelium Comprises Three Major Cell Types Despite this wide range of functions, the urothelium has a relatively simple structure comprising three main cell types that are distinguished by their location, size, and expression of molecular markers. Directly facing the luminal surface are large (50 –120 �m in diameter) multinucleated hexagonal cells known as superficial or umbrella cells, which are principally responsible for the barrier function of the urothelium (103). Adjacent superficial cells are connected by tight junction pro- teins including claudin-8 and zona occludens 1 (ZO-1) that restrict exchange of ions and solutes between cells and between urine and blood (1, 32, 75). Superficial cells are covered by a crystalline lattice comprising four major uroplakin proteins that together form asymmetric unit membrane (AUM) plaques. These plaques further restrict permeability to water, solutes, and toxins (37, 90, 105, 106). Superficial cells also contribute Address for reprint requests and other correspondence: X. Li, Depts. of Urology and Surgery, Boston Children’s Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115 (e-mail: sean.li@childrens.harvard. edu). Am J Physiol Renal Physiol 312: F732–F743, 2017. First published January 25, 2017; doi:10.1152/ajprenal.00337.2016. 1931-857X/17 Copyright © 2017 the American Physiological Society http://www.ajprenal.orgF732 Downloaded from journals.physiology.org/journal/ajprenal at Carnegie Mellon Univ (128.182.081.034) on April 5, 2021. http://doi.org/10.1152/ajprenal.00337.2016. mailto:sean.li@childrens.harvard.edu mailto:sean.li@childrens.harvard.edu to the plasticity in urothelial cell surface area through a regulated process of endocytosis or exocytosis of discoidal/ fusiform-shaped vesicles (DFVs) containing uroplakins (47, 50, 109). Underlying the superficial cell layer is a layer of intermediate cells that are significantly smaller (20 �m in diameter) than superficial cells. Finally, along the basement membrane is a layer of basal cells. Despite being the smallest population in size (5–10 �m in diameter), basal cells constitute the most abundant population of cells in adult urothelium. Given the substantial size discrepancy between superficial and either basal or intermediate cells, it is no surprise that histo- logical analysis of whole-mount adult mouse bladders has revealed that one superficial cell spans the area of ~40 under- lying intermediate/basal cells (36). Depending on the species, there can be as few as three discrete layers of urothelial cells in the mouse bladder and up to seven layers in the human bladder, with the additional layers contributed by interme- diate cells (11). In addition to the size discrepancy among urothelial cells in the different layers, urothelial cells can also be distinguished by molecular differentiation markers, which begin to be ex- pressed at different stages of embryogenesis (Fig. 1). Superfi- cial cells represent terminally differentiated cells and are the only cell layer in the bladder to express the low-molecular- weight cytokeratin 20 (Krt20) (29, 68, 82). Superficial cells also express several uroplakins (Upk) but lack expression of the high-molecular-weight cytokeratin Krt5, the transcriptional factor p63, and signaling molecule Sonic Hedgehog (Shh) (25). Similar to superficial cells, most intermediate cells are Upk� and Krt5�, but in contrast, intermediate cells also express p63. Diverging from superficial and intermediate cells, basal cells distinguish themselves by expression of high levels of Krt5 and p63 but are negative for Upk and Krt20. Notably, despite the previous assumption that each of the three urothelial layers comprises a homogenous population of cells based on these five markers, our recent findings suggest that there is, in fact, significant urothelial cell heterogeneity. For example, variable levels of histone H3 lysine 27 trimethylation (H3K27me3), an epigenetic modification often associated with gene silencing, are apparent among urothelial cells within the Krt5� basal as well as Krt5� intermediate and superficial cells (26a). In addition, ~14% of Krt5� basal cells also express Krt14. While the significance of urothelial cell heterogeneity remains to be determined, the Krt5�/Krt14� subpopulation of basal cells may play an important role in urothelial regeneration and tumorigenesis (to be discussed in subsequent sections) (55, 77, 81). Emerging techniques like single-cell RNA sequencing (scRNA-seq) may further stratify cells within each discrete layer and potentially identify functional differences among cells in each layer. Normally Quiescent Urothelium Rapidly Regenerates in Response to Injury Unlike the epithelium of the skin and intestine, mature urothelium has a very low mitotic index and turnover rate. Pulse-labeling of unstimulated rat bladders with tritiated thy- midine revealed a labeling index of ~0.2– 0.5% (14, 30, 61, 65) while uninjured mouse bladders had an even lower labeling index of 0.11% (36, 58). Similarly, bladder biopsies from normal human patients that were cultured with tritiated thymi- dine in vitro had a labeling index of 0.12% (28). Based on these low labeling indexes, turnover rates of quiescent rodent urothe- lium have been estimated to be approximately once every 200 days (14, 30). The prevailing quiescence of the urothelium makes its ability to awaken rapidly in response to damage even more remarkable. Within hours of chemical injury with cyclo- phosphamide (CPP) or protamine sulfate (PS) or biological insult with uropathogenic Escherichia coli (UPEC), the urothe- lium begins to proliferate and initiate the process of regener- ation (Fig. 2) (25, 71, 84). One can imagine that urothelial regeneration needs to be carefully controlled. Incomplete re- generation results in potential breaches in barrier function (Fig. 3) whereby toxic substances or pathogens in the urine can gain access to the bloodstream, stimulate local tissue inflammation, and/or depolarize afferent nerve fibers. In fact, this last process has been hypothesized as being a potential cause of bladder pain syndrome or interstitial cystitis (44, 83, 94). Conversely, unrestrained regeneration can lead to urothelial hyperplasia and possible malignant transformation (Fig. 3). An understanding of the molecular mechanisms responsible for maintaining the delicate balance between urothelial quiescence and regenera- tion is critical for devising new clinical strategies to prevent or treat diseases of the urothelium. Given the priority of maintaining a protective barrier, it is not surprising that one of the first steps in urothelial regener- SC IC BC (Shh- p63- Upk+ Krt5- Krt14- Krt20+) (Shh+ p63+ Upk+ Krt5- Krt14- Krt20-) (Shh+ p63+ Upk- Krt5+ Krt14-/+ Krt20-) A B