Journal List > Korean J Urol > v.51(1) > 1005431

Bae and Yoo: Cell-Based Therapy for Urinary Incontinence

Abstract

Urinary incontinence has become a societal problem that affects millions of people worldwide. Although numerous therapeutic modalities are available, none has been shown to be entirely satisfactory. Consequently, cell-based approaches using regenerative medicine technology have emerged as a potential solution that would provide a means of correcting anatomical deficiencies and restoring normal function. As such, numerous cell-based investigations have been performed to develop systems that are focused on addressing clinical needs. While most of these attempts remain in the experimental stages, several clinical trials are being designed or are in progress. This article provides an overview of the cell-based approaches that utilize various cell sources to develop effective treatment modalities for urinary incontinence.

INTRODUCTION

Urinary incontinence, which is characterized as any involuntary leakage of urine, is a social and distressing problem that affects approximately 25 percent of women and 9 percent of men [1,2]. Although urinary incontinence is classified by a variety of types, the underlying pathology can be grouped into two major categories: urethral hypermobility and intrinsic sphincter deficiency [3]. However, most cases of urinary incontinence exist between the extremes of these two categories, and most patients have elements of both disorders [4]. Although conservative management, such as dietary and lifestyle changes, pelvic floor muscle training, and pharmacologic agents, has been somewhat helpful in alleviating symptoms in mild conditions, these measures do not correct the underlying pathology [5-9]. In contrast, surgical and interventional procedures seek to increase the coaptation pressure of the urethral sphincter by means of sling procedures, artificial sphincter placement, and injectable bulking agents [10]. Sling procedures are considered the gold standard in correcting the underlying anatomical deformities, with success rates ranging from 80% to 94% [11-13]. However, complications associated with these procedures and long-term consequences of urethral and vaginal erosions of the sling materials remain as unsolved challenges. Injectable bulking agents, such as polytetrafluoroethylene, bovine collagen, silicone particles, and carbon beads, have also been applied clinically as a treatment modality for incontinence. These agents are designed to augment urethral and bladder neck tissues by increasing resistance to urine flow, but they have minimal effects on urethral mobility. As such, approximately 25% of patients with incontinence show correction of urinary incontinence, whereas 50% demonstrate improvement and the remaining 25% experience treatment failure [14,15]. More importantly, untoward effects associated with the bulking agents, including long-term biocompatibility, recurrence, inflammatory responses, migration, and erosion, remain a problem [11,16].
Due to the limitations of the current treatment modalities for urinary incontinence, investigators have sought alternative approaches to treating urinary incontinence. Regenerative medicine has emerged as an innovative scientific field that focuses on the development of new approaches to repairing cells, tissues, and organs for clinical applications. Recent advances in cell-based technology using regenerative medicine techniques suggest that this approach holds enormous potential to improve human conditions by encompassing alteration of the current biological state of a targeted tissue, augmentation of depleted function, or absolute functional tissue replacement [17-19]. Consequently, efforts in cell-based technology have focused on developing systems that would restore and maintain normal tissue function. To that end, numerous cell-based investigations have been performed to address urinary incontinence. This article aims to provide an overview of the cell-based investigations that utilize a wide range of cell sources to overcome the limitations of current treatment modalities for urinary incontinence.

FUNDAMENTALS OF CELL-BASED THERAPY FOR URINARY INCONTINENCE

The basic components required to achieve functional tissues and organs are cells, scaffolds, and the in vivo environment [17,18]. Regenerative medicine strategies that have been demonstrated to be successful involve the use of biocompatible matrices either with or without cells. The matrices are used either as supporting scaffolds to promote and facilitate tissue regeneration over smaller gaps or as cell delivery vehicles for larger defects. When cells are used, donor tissue is dissociated into individual cells, which are expanded in culture, combined with a support matrix, and introduced into the body to form mature and functional tissues. The cells can be delivered into the target region surgically or through a needle injection, depending on the type of tissue applications. Cells for tissue reconstitution can be derived from the native organ to be replaced, thus avoiding rejection. In instances where normal tissues are not available, different cell sources may be explored. Stem and progenitor cells offer numerous opportunities in regenerative medicine. Cells derived from various stages of development can be either implanted directly in the target tissues or guided into specific cell lineages in vitro followed by implantation in vivo.
Cells are an essential component required to augment tissue function for urinary incontinence. Cells derived from various sources have been used for urinary incontinence, including chondrocytes, smooth muscle cells, muscle precursor cells (MPCs), adipose-derived stem cells, and bone marrow stromal cells [20]. The use of these cells is aimed at achieving coaptation of the bladder neck region by augmenting tissue mass or restoring sphincter function. To use cells for application in urinary incontinence, a cell expansion system that permits recovery of large quantities of target cells must be developed. This has been one of the challenges that is being continuously investigated. Even when some organs, such as the liver, have a high regenerative capacity in vivo, in vitro expansion of cells derived from these organs has proved to be difficult. However, the discovery of privileged sites for committed precursor cells in specific organs and extensive study of the conditions that promote precursor cell maintenance and differentiation within these sites have begun to overcome some of the limitations associated with cell expansion in vitro. Urothelial cells, for instance, have been grown in culture in the past, but with only limited success. However, several novel culture protocols have been developed over the past two decades that allow for the maintenance of precursor cells in an undifferentiated state. Because these cells can remain in the growth phase, the ability to expand urothelial cultures is vastly improved [21-24]. These studies indicate that it may be possible to collect autologous cells from patient's tissues, expand them in culture, and return them to the donor in sufficient quantities for reconstructive purposes [23,25-30].
Biomaterials for various cell-based therapies are usually designed to replicate the biological and mechanical functions of native tissue structures and their extracellular matrix (ECM). They provide three-dimensional architecture for cells to reconstitute into new tissues and allow for the delivery of cells and bioactive factors (e.g., peptides, growth factors) to desired sites in the body to enhance functionality [31]. Because most mammalian cell types are anchorage-dependent, biomaterials serve as a cell-adhesion substrate that can deliver cells to specific regions of the body with a high loading efficiency. An ideal biomaterial for cell therapy should be biodegradable and absorbable without eliciting inflammatory responses that interfere with cellular function and tissue formation. Incompatible materials are destined for an inflammatory or foreign-body response that eventually leads to rejection or necrosis. Because biomaterials for cell therapy provide temporary mechanical support while the cells undergo spatial tissue organization, a suitable biomaterial should maintain adequate mechanical integrity to support tissue formation during early stages of development. To develop a suitable biomaterial for the cell-based treatment of urinary incontinence, various factors need to be considered, including biocompatibility, biodegradation, and structural and mechanical properties. These materials should be able to 1) facilitate the delivery of cells to target sites in the urethra and bladder neck region, 2) maintain the three-dimensional architecture that permits formation of new tissues, and 3) guide the development of new tissues with appropriate function [31]. As such, numerous biomaterials, both synthetic and naturally derived, have been developed and used as substrates for urinary incontinence, including collagen and alginate [32-37].
To achieve functional tissue in vivo, implanted cells must maintain viability by obtaining an adequate supply of nutrients and oxygen. One of the continued challenges in engineering clinically relevant tissues is the establishment of vascularization for implanted tissues and organs. This is especially true for large tissue masses consisting of cell-based implants that require a sufficient blood supply to maintain viability. It is well known that cellular implants that are more than a few hundred microns away from a blood vessel will not survive because of diffusion limitations. Consequently, tissue implants that are unable to establish vascularization in time result in cell death and tissue necrosis [38]. This has been a critical limiting factor for developing functional tissues for human applications [39]. An approach to maintaining tissue viability in vivo is to place the engineered tissue adjacent to a heavily vascularized tissue such as the omentum to achieve adequate vascularization [40]. However, this approach may not always be feasible because the target implantation site may not be in close proximity. To overcome this limitation, several strategies have been used to facilitate the delivery of oxygen and improve the survival of implanted cells. Angiogenic growth factors such as vascular endothelial growth factor (VEGF), which is a potent endothelial cell-specific mitogen, have been used to promote neo-vascularization [41,42]. Although the effectiveness of an enhanced angiogenic response has been demonstrated in many tissue systems, the rate of angiogenesis cannot be accelerated, thus limiting the size of implantable tissue masses [43]. Another approach to overcoming diffusion limitation is to configure biomaterials that facilitate vascularization to implanted cells and tissues by modifying properties and porosity [44,45]. Recently, efforts have been placed in developing strategies to prolong cell survival until host neovascularization is achieved by increasing oxygen tension or incorporating oxygen-generating biomaterials [46,47].

CELL-BASED APPROACHES FOR URINARY INCONTINENCE

1. Injectable chondrocytes

One of the early investigations of cell therapy for urinary incontinence involved the use of autologous chondrocytes (cartilage cells). Chondrocytes were chosen as a tissue bulking agent because this cell type possesses an inherent ability to produce extracellular matrix and maintain structural integrity in vivo, thus providing ideal tissue bulking for the treatment of urinary incontinence [32]. In addition, chondrocytes can be easily isolated, grown, and expanded in culture conditions. Alginate, a liquid solution of gluronic and mannuronic acid, serves as a substrate for the delivery of chondrocytes through a needle. In a preclinical study demonstrating the feasibility of using chondrocytes as a bulking agent, autologous chondrocytes from porcine ear cartilage were grown and expanded in culture. The chondrocytes, suspended in alginate, were then injected endoscopically to correct anatomical deformities of the vesicoureteral junction. This study showed that chondrocytes combined with alginate in vitro can be easily injected cystoscopically, and that the elastic cartilage tissue formed within the injection region can correct vesicoureteral reflux without any evidence of obstruction [48].
Based on the results of multiple experimental studies, two multicenter clinical trials were conducted using the engineered chondrocyte technology. In one study, patients with vesicoureteral reflux were treated at 10 centers across the United States by following the same strategy that involved the use of autologous chondrocytes combined with alginate. The patients had a success rate similar to that with other injectable substances in terms of cure [49]. In the second study, patients with urinary incontinence were treated endoscopically with injected chondrocytes at three different medical centers in the United States. Thirty-two patients received a single outpatient injection just distal to the bladder neck. The investigators concluded that this treatment was safe, effective, and durable, with 50% of patients remaining completely dry 12 months after a single injection. Twenty-six of 32 patients reported at least some improvements at 3 months after the injection that continued until at least the 12-month follow-up visit [32].

2. Injectable muscle cells

The potential use of injectable cultured myoblasts for the treatment of stress urinary incontinence has been investigated [50,51]. Labeled myoblasts were directly injected into the proximal urethra and lateral bladder walls of athymic mice with a micro-syringe in an open surgical procedure. Tissue harvested up to 35 days after injection contained the labeled myoblasts, as well as evidence of differentiation into regenerative myofibers. This study showed that a significant portion of the injected myoblast population persisted in vivo. Similar techniques of sphincter-derived muscle cells have been used for the treatment of urinary incontinence in a pig model [52]. The fact that myoblasts can be labeled and survive after injection and begin the process of myogenic differentiation supports the feasibility of using cultured cells of muscular origin as an injectable bioimplant.
The use of injectable MPCs has also been investigated for use in the treatment of urinary incontinence due to irreversible urethral sphincter injury or developmental defects. MPCs are the quiescent satellite cells found in each myofiber that can proliferate to form myoblasts and eventually myotubes and new muscle tissue. The standard MPC grafting technique consists of an injection of cells that were originally harvested from a muscle biopsy by enzymatic digestion and then cultured under specific conditions to eliminate nonmyogenic cells. However, there is now increasing evidence that these successive steps of MPC preparation alter the myogenic potential of these cells in vivo [53,54].
MPCs have been shown to play an active role in the regeneration of injured striated urethral sphincter [55]. In a subsequent study, autologous MPCs were injected into a rat model of urethral sphincter injury, and replacement of mature myotubes as well as restoration of functional motor units were noted in the regenerating sphincter muscle tissue [56]. This was the first demonstration of the replacement of both sphincter muscle tissue and its innervation by the injection of MPCs, which suggests that MPCs may be a minimally invasive solution for urinary incontinence in patients with irreversible urinary sphincter muscle insufficiency. Another study investigated a method of intraurethral MPC transplantation consisting of implanting freshly isolated myofibers with their satellite cells. The investigators hypothesized that myofiber death after implantation would induce activation of satellite cells in vivo, thus avoiding the necessity for MPC extraction and in vitro expansion [57]. Histological studies showed that the myogenic process, consisting of myofiber degeneration and satellite cell activation, was observed at 7 days after transplantation, followed by myotube formation replacing parental myofibers at 30 days. In the subsequent experiments, implantation of myofiber strips to endoscopic sphincter injured urethra generated a pressure peak that decreased after endoscopic injury and reappeared 60 minutes later, indicating that this action was tonic and under neural control. Histologically, the myotubes were oriented in the same direction as the parental myofibers, suggesting that satellite cell fusion was guided by the tubes of extracellular matrix surrounding each myofiber. There was evidence that new nerve fibers developed in the vicinity of the implanted myotubules, and neural tissue was present in greater density than in surrounding tissue. These findings suggest the presence of concomitant neuronal development and innervation of the transplanted myofibers. In contrast, the experimental results of another animal study indicated that the improvement of muscle regeneration was not better than that with muscle-derived stem cells [58]. The reasons for this discrepancy remain to be explored.
Cultured myoblasts are one of the most studied cell types for the treatment of incontinence [59,60]. The fact that MPCs survive after injection and initiate the process of myogenic differentiation further supports the feasibility of using cultured cells of muscular origin as an injectable bioimplant [56]. Although many studies have been performed in laboratory settings using protocols developed for animal cells to demonstrate the principle and feasibility of using cultured cells of muscular origin, culture systems compatible with clinical application are needed [61,62]. One problem in these established protocols is the use of extracellular matrix proteins derived from cancer cells (Matrigel®, BD Biosciences, San Jose, USA), which contain a high concentration of various growth factors and are commonly used to promote the proliferation and differentiation of muscle precursors. In order to achieve rapid clinical translation of MPC therapy, culture conditions were optimized for human application. A human MPC culture system that uses FDA-approved substrates with defined media for expansion and differentiation was developed [62].
An interesting clinical trial that used striated muscle-derived myoblast transplantation was reported by Strasser et al [63]. Using transurethral ultrasound-guided injections of autologous myoblasts and fibroblasts, 42 patients (29 women, 13 men) with urinary stress incontinence were treated. In 35 patients, urinary incontinence was completely cured. In seven patients who had undergone multiple surgical procedures and radiotherapy, urinary incontinence improved but was not eliminated. No side effects or complications were reported postoperatively. In this study, the authors concluded that urinary incontinence can be treated effectively with autologous stem cells. It is noteworthy that this represents the first attempt to use an autologous stem cell strategy in clinical urology. The same investigators conducted a randomized controlled trial comparing ultrasound-guided transurethral rhabdosphincter injection of autologous myoblasts and fibroblasts vs. transurethral collagen injection for incontinence in 63 eligible women [64]. At the follow-up examination at 12 months, 38 of the 42 women injected with autologous cells were completely continent, compared with 2 of the 21 patients given conventional treatment with collagen. However, this report was later retracted by the Lancet because of several controversial issues [65]. Despite the retraction of the report on this clinical trial, several other FDA-approved cell therapy clinical trials targeting incontinence are on the horizon (http://www.clinicaltrials.gov).

3. Stem cells

One of the objectives of regenerative medicine therapy for incontinence is reconstruction of the sphincter muscle itself. Stem cells have been proposed as a promising cell source to replace, repair, or enhance the biological functions of damaged sphincter [66]. Stem cells are defined by their ability to self-renew and differentiate into a variety of cell types. They are further classified by the breadth of cell lineages into which they may potentially differentiate [67]. Research that explores the possible applications of stem cells in the field of urology has been increasing. The current strategy for the cell-based approach to build functional tissues uses autologous cells from the diseased organs. However, in instances where normal cells cannot be obtained because of extensive end-stage disease, stem cells are envisioned as an ideal source of cells because they can differentiate into desired cell types if guided appropriately [68].
The pluripotency of human embryonic stem cells is highlighted by their ability to form embryoid bodies in vitro, which are cell aggregations that contain all three germ layers. The use of embryonic stem cells as a primary nonimmunogenic tissue source for seeding of decellularized scaffolds has been heavily investigated [69]. However, clinical applications using embryonic stem cells are faced with several challenges, including the propensity of these cells to form teratomas in vivo and ethical concerns about the destruction of human embryos [70]. Recently, fetal stem cells derived from amniotic fluid and placenta have been described and represent a novel source of stem cells [71,72]. These cells express markers consistent with human embryonic stem cells, such as OCT4 and SSEA-4, but they do not form teratomas. The cells are multipotent and can differentiate into cells from all three germ layers. In addition, the cells have high replicative potential and can be stored for future self-use, without the risks for rejection and without ethical concerns [73]. Although these cells do differentiate into the myogenic lineage, further studies are necessary to determine their utility in urinary incontinence applications.
Multipotent stem cells have become an attractive option for stem cell therapies in many organ systems, including the urologic system. The use of these cells avoids the controversy surrounding human embryonic stem cells, and unlike embryonic cells, they do not trans-differentiate into a malignant phenotype. Thus, there is a diminished risk for teratoma formation when implanted in vivo and they can be extracted from many different tissues, including bone marrow, striated muscle, fat, skin, testicle, and synovial membrane [74]. Adult-derived stem cells, such as adipose-derived stem cells and bone marrow stromal cells or mesenchymal stem cells (MSCs), are gaining popularity because they have a more extensive differentiation potential than previously reported [75-77]. One limitation that hampers rapid clinical translation is the expandability of the cells. Strategies to overcome this limitation are being developed, such as adding growth factors to the injected cells to stimulate proliferation or culturing cells on biodegradable matrices composed of natural extracellular matrix proteins or synthetic polymers [78-80].
The use of adipose-derived stem cells has been explored for the treatment of urinary incontinence [81]. Processed lipoaspirate (PLA) cells are abundant in the adipose tissue, and they are amenable to harvesting under local anesthesia. These cells are phenotypically similar to mesenchymal stem cells and have been shown to differentiate into adipogenic, chondrogenic, osteogenic, neurogenic, and myogenic lineages [82]. In one study, lipoaspirates from female patients undergoing liposuction were processed to yield a pluripotent population of PLA cells that were injected into the bladder and urethra. Eight weeks after injection, PLA cells demonstrated the expression of alpha-smooth muscle actin, an early marker of smooth muscle differentiation. This study suggests that PLA cells are an easily accessible source of pluripotent cells, making them ideal for tissue regeneration. Human PLA cells injected into the urinary tract show morphological and phenotypic evidence of smooth muscle incorporation and differentiation over time. In another study, the effects of adipose-derived stem cells isolated from the peri-ovary fat were implanted in a rat model of incontinence [83]. This study showed that the treated animals showed a normal voiding pattern and possessed higher elastin and smooth muscle content than did untreated incontinent animals.
Bone marrow derived MSCs have been used as a cell source for the treatment of urinary incontinence [84]. MSCs were exposed to 5-azacytidine to induce myogenic differentiation and were culture expanded. Subsequently, these cells were implanted into the bladder neck region of athymic rats. The results of this study suggested that the use of MSCs for the treatment of urinary incontinence may be a feasible option [84].

CONCLUSIONS

Rapid advances in cell-based technologies have provided innovative therapeutic opportunities for numerous human conditions. This article presents cell-based approaches that are designed to address the challenges involved with the current treatment modalities for urinary incontinence. These include the use of a wide spectrum of cell sources ranging from nonurologic autologous cells to stem cells at various stages of development. Although the current status of research in this area remains mostly in the laboratory, several technologies are currently being developed and validated clinically. These innovative developments have generated enormous hope and anticipation that the new modalities will provide effective treatments and improve the quality of life of millions of patients in the near future.

ACKNOWLEDGEMENT

The authors thank Dr. Jennifer Olson for editorial assistance.

References

1. Hannestad YS, Rortveit G, Sandvik H, Hunskaar S. A community-based epidemiological survey of female urinary incontinence: the Norwegian EPINCONT study. Epidemiology of Incontinence in the County of Nord-Trondelag. J Clin Epidemiol. 2000; 53:1150–1157. PMID: 11106889.
2. Malmsten UG, Milsom I, Molander U, Norlen LJ. Urinary incontinence and lower urinary tract symptoms: an epidemiological study of men aged 45 to 99 years. J Urol. 1997; 158:1733–1737. PMID: 9334589.
crossref
3. Dalpiaz O, Curti P. Role of perineal ultrasound in the evaluation of urinary stress incontinence and pelvic organ prolapse: a systematic review. Neurourol Urodyn. 2006; 25:301–306. PMID: 16688711.
crossref
4. Walsh LP, Zimmern PE, Pope N, Shariat SF. Comparison of the Q-tip test and voiding cystourethrogram to assess urethral hypermobility among women enrolled in a randomized clinical trial of surgery for stress urinary incontinence. J Urol. 2006; 176:646–649. PMID: 16813912.
crossref
5. Alhasso A, Glazener CM, Pickard R, N'Dow J. Adrenergic drugs for urinary incontinence in adults. Cochrane Database Syst Rev. 2003; 2:CD001842. PMID: 12804414.
crossref
6. Bo K, Talseth T, Holme I. Single blind, randomised controlled trial of pelvic floor exercises, electrical stimulation, vaginal cones, and no treatment in management of genuine stress incontinence in women. BMJ. 1999; 318:487–493. PMID: 10024253.
crossref
7. Burns PA, Pranikoff K, Nochajski TH, Hadley EC, Levy KJ, Ory MG. A comparison of effectiveness of biofeedback and pelvic muscle exercise treatment of stress incontinence in older community-dwelling women. J Gerontol. 1993; 48:M167–M174. PMID: 8315230.
8. Glazener CM, Herbison GP, MacArthur C, Grant A, Wilson PD. Randomised controlled trial of conservative management of postnatal urinary and faecal incontinence: six year follow up. BMJ. 2005; 330:337. PMID: 15615766.
crossref
9. Goode PS, Burgio KL, Locher JL, Roth DL, Umlauf MG, Richter HE, et al. Effect of behavioral training with or without pelvic floor electrical stimulation on stress incontinence in women: a randomized controlled trial. JAMA. 2003; 290:345–352. PMID: 12865375.
10. Sharifi-Aghdas F. Surgical management of stress urinary incontinence. Urol J. 2005; 2:175–182. PMID: 17602425.
11. Kiilholma P, Mäkinen J, Chancellor MB, Pitkänen Y, Hirvonen T. Modified Burch colposuspension for stress urinary incontinence in females. Surg Gynecol Obstet. 1993; 176:111–115. PMID: 8421796.
12. Nilsson CG, Kuuva N, Falconer C, Rezapour M, Ulmsten U. Long-term results of the tension-free vaginal tape (TVT) procedure for surgical treatment of female stress urinary incontinence. Int Urogynecol J Pelvic Floor Dysfunct. 2001; 12(Suppl 2):S5–S8. PMID: 11450979.
crossref
13. Ulmsten U, Falconer C, Johnson P, Jomaa M, Lanner L, Nilsson CG, et al. A multicenter study of tension-free vaginal tape (TVT) for surgical treatment of stress urinary incontinence. Int Urogynecol J Pelvic Floor Dysfunct. 1998; 9:210–213. PMID: 9795826.
crossref
14. Herschorn S, Steele DJ, Radomski SB. Followup of intraurethral collagen for female stress urinary incontinence. J Urol. 1996; 156:1305–1309. PMID: 8808860.
crossref
15. Corcos J, Fournier C. Periurethral collagen injection for the treatment of female stress urinary incontinence: 4-year follow-up results. Urology. 1999; 54:815–818. PMID: 10565739.
crossref
16. Smaldone MC, Chancellor MB. Muscle derived stem cell therapy for stress urinary incontinence. World J Urol. 2008; 26:327–332. PMID: 18470515.
crossref
17. Atala A. Tissue engineering and regenerative medicine: concepts for clinical application. Rejuvenation Res. 2004; 7:15–31. PMID: 15256042.
crossref
18. Atala A. Recent developments in tissue engineering and regenerative medicine. Curr Opin Pediatr. 2006; 18:167–171. PMID: 16601497.
crossref
19. Koh CJ, Atala A. Tissue engineering, stem cells, and cloning: opportunities for regenerative medicine. J Am Soc Nephrol. 2004; 15:1113–1125. PMID: 15100351.
crossref
20. Kuhn A, Gelman W, Kuhn P. Injectable therapy for urinary incontinence: a review. Praxis. 2004; 93:188–192. PMID: 15008014.
21. Cilento BG, Freeman MR, Schneck FX, Retik AB, Atala A. Phenotypic and cytogenetic characterization of human bladder urothelia expanded in vitro. J Urol. 1994; 152:665–670. PMID: 7517459.
crossref
22. Scriven SD, Booth C, Thomas DF, Trejdosiewicz LK, Southgate J. Reconstitution of human urothelium from monolayer cultures. J Urol. 1997; 158:1147–1152. PMID: 9258159.
crossref
23. Liebert M, Hubbel A, Chung M, Wedemeyer G, Lomax MI, Hegeman A, et al. Expression of mal is associated with urothelial differentiation in vitro: identification by differential display reverse-transcriptase polymerase chain reaction. Differentiation. 1997; 61:177–185. PMID: 9084136.
crossref
24. Puthenveettil JA, Burger MS, Reznikoff CA. Replicative senescence in human uroepithelial cells. Adv Exp Med Biol. 1999; 462:83–91. PMID: 10599415.
crossref
25. Cilento BG, Freeman MR, Schneck FX, Retik AB, Atala A. Phenotypic and cytogenetic characterization of human bladder urothelia expanded in vitro. J Urol. 1994; 152:665–670. PMID: 7517459.
crossref
26. Freeman MR, Yoo JJ, Raab G, Soker S, Adam RM, Schneck FX, et al. Heparin-binding EGF-like growth factor is an autocrine growth factor for human urothelial cells and is synthesized by epithelial and smooth muscle cells in the human bladder. J Clin Invest. 1997; 99:1028–1036. PMID: 9062361.
crossref
27. Liebert M, Wedemeyer G, Abruzzo LV, Kunkel SL, Hammerberg C, Cooper KD, et al. Stimulated urothelial cells produce cytokines and express an activated cell surface antigenic phenotype. Semin Urol. 1991; 9:124–130. PMID: 1853009.
28. Nguyen HT, Park JM, Peters CA, Adam RM, Orsola A, Atala A, et al. Cell-specific activation of the HB-EGF and ErbB1 genes by stretch in primary human bladder cells. In Vitro Cell Dev Biol Anim. 1999; 35:371–375. PMID: 10462199.
crossref
29. Oberpenning F, Meng J, Yoo JJ, Atala A. De novo reconstitution of a functional mammalian urinary bladder by tissue engineering. Nat Biotechnol. 1999; 17:149–155. PMID: 10052350.
crossref
30. Puthenveettil JA, Burger MS, Reznikoff CA. Replicative senescence in human uroepithelial cells. Adv Exp Med Biol. 1999; 462:83–91. PMID: 10599415.
crossref
31. Kim BS, Mooney DJ. Development of biocompatible synthetic extracellular matrices for tissue engineering. Trends Biotechnol. 1998; 16:224–230. PMID: 9621462.
crossref
32. Bent AE, Tutrone RT, McLennan MT, Lloyd LK, Kennelly MJ, Badlani G. Treatment of intrinsic sphincter deficiency using autologous ear chondrocytes as a bulking agent. Neurourol Urodyn. 2001; 20:157–165. PMID: 11170190.
crossref
33. Noth U, Tuli R, Osyczka AM, Danielson KG, Tuan RS. In vitro engineered cartilage constructs produced by press-coating biodegradable polymer with human mesenchymal stem cells. Tissue Eng. 2002; 8:131–144. PMID: 11886661.
34. Wang Y, Blasioli DJ, Kim HJ, Kim HS, Kaplan DL. Cartilage tissue engineering with silk scaffolds and human articular chondrocytes. Biomaterials. 2006; 27:4434–4442. PMID: 16677707.
crossref
35. Keegan PE, Atiemo K, Cody J, McClinton S, Pickard R. Periurethral injection therapy for urinary incontinence in women. Cochrane Database Syst Rev. 2007; 3:CD003881. PMID: 17636740.
crossref
36. Trabucco E, Soderberg M, Cobellis L, Torella M, Bystrom B, Ekman-Ordeberg G, et al. Role of proteoglycans in the organization of periurethral connective tissue in women with stress urinary incontinence. Maturitas. 2007; 58:395–405. PMID: 18022774.
crossref
37. Isom-Batz G, Zimmern PE. Collagen injection for female urinary incontinence after urethral or periurethral surgery. J Urol. 2009; 181:701–704. PMID: 19091340.
crossref
38. Folkman J, Hochberg M. Self-regulation of growth in three dimensions. J Exp Med. 1973; 138:745–753. PMID: 4744009.
crossref
39. Khademhosseini A, Langer R, Borenstein J, Vacanti JP. Microscale technologies for tissue engineering and biology. Proc Natl Acad Sci U S A. 2006; 103:2480–2487. PMID: 16477028.
crossref
40. Atala A, Bauer SB, Soker S, Yoo JJ, Retik AB. Tissue-engineered autologous bladders for patients needing cystoplasty. Lancet. 2006; 367:1241–1246. PMID: 16631879.
crossref
41. Ahrendt G, Chickering DE, Pranieri JP. Angiogenic growth factors: a review for tissue engineering. Tissue Eng. 1998; 4:117–130.
crossref
42. De Coppi P, Delo D, Farrugia L, Udompanyanan K, Yoo JJ, Nomi M, et al. Angiogenic gene-modified muscle cells for enhancement of tissue formation. Tissue Eng. 2005; 11:1034–1044. PMID: 16144439.
43. Smith MK, Peters MC, Richardson TP, Garbern JC, Mooney DJ. Locally enhanced angiogenesis promotes transplanted cell survival. Tissue Eng. 2004; 10:63–71. PMID: 15009931.
crossref
44. Tilkorn D, Messina A, Bedogni A, Keramidaris E, Han X, Palmer J, et al. Implanted myoblast survival is dependent on the degree of vascularization in a novel delayed implantation/prevascularization tissue engineering model. Tissue Eng Part A. 2009.
crossref
45. Seach N, Mattesich M, Abberton K, Matsuda K, Tilkorn DJ, Rophael J, et al. Vascularized tissue engineering mouse chamber model supports thymopoiesis of ectopic thymus tissue grafts. Tissue Eng Part C Methods. 2009; Epub ahead of print.
crossref
46. Harrison BS, Eberli D, Lee SJ, Atala A, Yoo JJ. Oxygen generating biomaterials for tissue regeneration. Biomaterials. 2007; 28:4628–4634. PMID: 17681597.
47. Oh SH, Ward CL, Atala A, Yoo JJ, Harrison BS. Oxygen generating scaffolds for enhancing engineered tissue survival. Biomaterials. 2009; 30:757–762. PMID: 19019425.
crossref
48. Atala A, Kim W, Paige KT, Vacanti CA, Retik AB. Endoscopic treatment of vesicoureteral reflux with a chondrocyte-alginate suspension. J Urol. 1994; 152:641–643. PMID: 8021988.
crossref
49. Diamond DA, Caldamone AA. Endoscopic correction of vesicoureteral reflux in children using autologous chondrocytes: preliminary results. J Urol. 1999; 162:1185–1188. PMID: 10458462.
crossref
50. Yokoyama T, Huard J, Chancellor MB. Myoblast therapy for stress urinary incontinence and bladder dysfunction. World J Urol. 2000; 18:56–61. PMID: 10766045.
crossref
51. Chancellor MB, Yokoyama T, Tirney S, Mattes CE, Ozawa H, Yoshimura N, et al. Preliminary results of myoblast injection into the urethra and bladder wall: a possible method for the treatment of stress urinary incontinence and impaired detrusor contractility. Neurourol Urodyn. 2000; 19:279–287. PMID: 10797585.
crossref
52. Strasser H, Berjukow S, Marksteiner R, Margreiter E, Hering S, Bartsch G, et al. Stem cell therapy for urinary stress incontinence. Exp Gerontol. 2004; 39:1259–1265. PMID: 15489048.
crossref
53. Collins CA, Olsen I, Zammit PS, Heslop L, Petrie A, Partridge TA, et al. Stem cell function, self-renewal, and behavioral heterogeneity of cells from the adult muscle satellite cell niche. Cell. 2005; 122:289–301. PMID: 16051152.
crossref
54. Smythe GM, Grounds MD. Exposure to tissue culture conditions can adversely affect myoblast behavior in vivo in whole muscle grafts: implications for myoblast transfer therapy. Cell Transplant. 2000; 9:379–393. PMID: 10972337.
crossref
55. Yiou R, Lefaucheur JP, Atala A. The regeneration process of the striated urethral sphincter involves activation of intrinsic satellite cells. Anat Embryol. 2003; 206:429–435. PMID: 12728313.
crossref
56. Yiou R, Yoo JJ, Atala A. Restoration of functional motor units in a rat model of sphincter injury by muscle precursor cell autografts. Transplantation. 2003; 76:1053–1060. PMID: 14557752.
57. Lecoeur C, Swieb S, Zini L, Riviere C, Combrisson H, Gherardi R, et al. Intraurethral transfer of satellite cells by myofiber implants results in the formation of innervated myotubes exerting tonic contractions. J Urol. 2007; 178:332–337. PMID: 17507041.
crossref
58. Jankowski RJ, Deasy BM, Huard J. Muscle-derived stem cells. Gene Ther. 2002; 9:642–647. PMID: 12032710.
crossref
59. Chancellor MB, Yokoyama T, Tirney S, Mattes CE, Ozawa H, Yoshimura N, et al. Preliminary results of myoblast injection into the urethra and bladder wall: a possible method for the treatment of stress urinary incontinence and impaired detrusor contractility. Neurourol Urodyn. 2000; 19:279–287. PMID: 10797585.
crossref
60. Yokoyama T, Huard J, Chancellor MB. Myoblast therapy for stress urinary incontinence and bladder dysfunction. World J Urol. 2000; 18:56–61. PMID: 10766045.
crossref
61. Bonavaud S, Agbulut O, D'Honneur G, Nizard R, Mouly V, Butler-Browne G. Preparation of isolated human muscle fibers: a technical report. in vitro Cell Dev Biol Anim. 2002; 38:66–72. PMID: 11928997.
crossref
62. Eberli D, Soker S, Atala A, Yoo JJ. Optimization of human skeletal muscle precursor cell culture and myofiber formation in vitro. Methods. 2009; 47:98–103. PMID: 18952174.
crossref
63. Strasser H, Marksteiner R, Margreiter E, Pinggera GM, Mitterberger M, Fritsch H, et al. Stem cell therapy for urinary incontinence. Urologe A. 2004; 43:1237–1241. PMID: 15549161.
64. Strasser H, Marksteiner R, Margreiter E, Pinggera GM, Mitterberger M, Frauscher F, et al. Autologous myoblasts and fibroblasts versus collagen for treatment of stress urinary incontinence in women: a randomised controlled trial. Lancet. 2007; 369:2179–2186. PMID: 17604800.
65. Kleinert S, Horton R. Retraction--autologous myoblasts and fibroblasts versus collagen [corrected] for treatment of stress urinary incontinence in women: a [corrected] randomised controlled trial. Lancet. 2008; 372:789–790. PMID: 18774408.
66. Feki A, Faltin DL, Lei T, Dubuisson JB, Jacob S, Irion O. Sphincter incontinence: is regenerative medicine the best alternative to restore urinary or anal sphincter function? Int J Biochem Cell Biol. 2007; 39:678–684. PMID: 17208507.
crossref
67. Keller G. Embryonic stem cell differentiation: emergence of a new era in biology and medicine. Genes Dev. 2005; 19:1129–1155. PMID: 15905405.
crossref
68. Becker C, Jakse G. Stem cells for regeneration of urological structures. Eur Urol. 2007; 51:1217–1228. PMID: 17254699.
crossref
69. Lewis JM, Cheng EY. Non-traditional management of the neurogenic bladder: tissue engineering and neuromodulation. ScientificWorldJournal. 2007; 7:1230–1241. PMID: 17704856.
crossref
70. Reubinoff BE, Pera MF, Fong CY, Trounson A, Bongso A. Embryonic stem cell lines from human blastocysts: somatic differentiation in vitro. Nat Biotechnol. 2000; 18:399–404. PMID: 10748519.
crossref
71. De Coppi P, Bartsch G Jr, Siddiqui MM, Xu T, Santos CC, Perin L, et al. Isolation of amniotic stem cell lines with potential for therapy. Nat Biotechnol. 2007; 25:100–106. PMID: 17206138.
crossref
72. De Coppi P, Callegari A, Chiavegato A, Gasparotto L, Piccoli M, Taiani J, et al. Amniotic fluid and bone marrow derived mesenchymal stem cells can be converted to smooth muscle cells in the cryo-injured rat bladder and prevent compensatory hypertrophy of surviving smooth muscle cells. J Urol. 2007; 177:369–376. PMID: 17162093.
crossref
73. Atala A. Regenerative medicine and tissue engineering in urology. Urol Clin North Am. 2009; 36:199–209. PMID: 19406321.
crossref
74. Baker SC, Rohman G, Southgate J, Cameron NR. The relationship between the mechanical properties and cell behaviour on PLGA and PCL scaffolds for bladder tissue engineering. Biomaterials. 2009; 30:1321–1328. PMID: 19091399.
crossref
75. Fierabracci A, Caione P, Di Giovine M, Zavaglia D, Bottazzo GF. Identification and characterization of adult stem/progenitor cells in the human bladder (bladder spheroids): perspectives of application in pediatric surgery. Pediatr Surg Int. 2007; 23:837–839. PMID: 17619197.
crossref
76. Lane TA, Ho AD, Bashey A, Peterson S, Young D, Law P. Mobilization of blood-derived stem and progenitor cells in normal subjects by granulocyte-macrophage- and granulocyte-colony-stimulating factors. Transfusion. 1999; 39:39–47. PMID: 9920165.
crossref
77. Vrana KE, Hipp JD, Goss AM, McCool BA, Riddle DR, Walker SJ, et al. Nonhuman primate parthenogenetic stem cells. Proc Natl Acad Sci U S A. 2003; 100(Suppl 1):11911–11916. PMID: 14504386.
crossref
78. Brazel CY, Ducceschi MH, Pytowski B, Levison SW. The FLT3 tyrosine kinase receptor inhibits neural stem/progenitor cell proliferation and collaborates with NGF to promote neuronal survival. Mol Cell Neurosci. 2001; 18:381–393. PMID: 11640895.
crossref
79. Inanc B, Elcin AE, Elcin YM. Human embryonic stem cell differentiation on tissue engineering scaffolds: effects of NGF and retinoic acid induction. Tissue Eng Part A. 2008; 14:955–964. PMID: 19230122.
80. Nakajima M, Ishimuro T, Kato K, Ko IK, Hirata I, Arima Y, et al. Combinatorial protein display for the cell-based screening of biomaterials that direct neural stem cell differentiation. Biomaterials. 2007; 28:1048–1060. PMID: 17081602.
crossref
81. Jack GS, Almeida FG, Zhang R, Alfonso ZC, Zuk PA, Rodriguez LV. Processed lipoaspirate cells for tissue engineering of the lower urinary tract: implications for the treatment of stress urinary incontinence and bladder reconstruction. J Urol. 2005; 174:2041–2045. PMID: 16217390.
crossref
82. Zuk PA, Zhu M, Mizuno H, Huang J, Futrell JW, Katz AJ, et al. Multilineage cells from human adipose tissue: implications for cell-based therapies. Tissue Eng. 2001; 7:211–228. PMID: 11304456.
crossref
83. Lin G, Wang G, Banie L, Ning H, Shindel AW, Fandel TM, et al. Treatment of stress urinary incontinence with adipose tissue-derived stem cells. Cytotherapy. 2009; Epub ahead of print.
crossref
84. Drost AC, Weng S, Feil G, Schäfer J, Baumann S, Kanz L, et al. In vitro myogenic differentiation of human bone marrow-derived mesenchymal stem cells as a potential treatment for urethral sphincter muscle repair. Ann N Y Acad Sci. 2009; 1176:135–143. PMID: 19796241.
crossref
TOOLS
Similar articles