Abstract
A significant number of women experience stress urinary incontinence (SUI), which greatly affects their quality of life. Recent research investigating utilization of stem cells and their derivatives for the prevention and treatment of SUI has been performed to test the effect of cell source and method of administration in several animal models of SUI. The type of stem cell, timing of optimal dose or doses after injury, mechanism of action of stem cells, and route of administration must be investigated both preclinically and clinically before stem cell therapy becomes a possible treatment for SUI, although the future of this therapy looks promising. This article reviews the progress in stem cell research for incontinence and describes areas of future work as suggested by research in other fields.
Keywords: Stem cell, Incontinence, Regenerative, Voiding dysfunction, Stress urinary incontinence, Women’s health, Gynecology, Noninvasive therapy, Muscle-derived stem cells, MDSCs, Bone marrow–derived stem cells, BMSCs, Adipose-derived stem cells, ADCSCs, Human umbilical-cord blood cells, HUCBs
Introduction
Urinary incontinence and bladder dysfunction affect a large number of people, who experience a decreased quality of life due to social and sexual isolation as well as feelings of shame, anxiety, and depression [1]. As the elderly population increases, both the number of people who have a decreased quality of life due to these conditions and the economic burden of treating these dysfunctions are expected to increase dramatically [2].
Stress urinary incontinence (SUI) is typically treated with pelvic floor exercises, a bulking agent, or surgery. Duloxetine, a pharmaceutical agent, is approved for SUI treatment in some countries, but is not approved for this indication in the United States. Surgery remains the gold standard for bothersome cases and options include a Burch colposuspension procedure, an autologous fascial sling, and a midurethral synthetic sling. The latter has become the most common operation for SUI due to its minimally invasive nature, ease of use, and good long-term efficacy with up to 11 years of follow-up [3]. Nonetheless, up to a third of women undergo a second anti-SUI surgery due to recurrent SUI during their lifetime [4, 5]. In some patients, slings show no efficacy immediately after surgery and success rates decline steadily after surgery [6, 7].
There is a lack of noninvasive therapy successful at treating SUI over the long term, suggesting the potential for development of innovative procedures such as stem cell therapy. Various techniques using cells derived from various tissues (e.g., adipose and muscle tissues) utilizing different application techniques have been tested in several different animal models of incontinence [8]. Myoblasts have been tested for application to urology; however, in cardiology, myoblasts have been shown to have slower growth and poorer regeneration capabilities than stem cells [9]. Therefore, several studies have tested whether to use stem cells directly or to differentiate them into myoblasts or along other lineages before implantation [10–12].
Early clinical trials testing stem cells as a treatment for incontinence have used autologous muscle-derived stem cells (MDSCs) that have been injected directly either transurethrally or periurethrally [8]. A recent randomized blinded trial showed almost 50% improvement in patients 1 year after injection, although the study size was small [13].
Development of stem cell therapy for treatment of nonurological disorders, such as those in cardiology, is more advanced. Preclinical studies investigating safety, paracrine effects, source of cell, and efficacy have been completed [14–16, 17••, 18–24] and several randomized, double-blind, placebo-controlled clinical trials investigating both direct and intravenous infusion as well as dosage already have been done [25••, 26, 27]. An intracoronary infusion of autologous bone marrow or MDSC has been demonstrated to be safe in humans by the task force of the European Heart Association [28]. Because of the importance of delivery timing after injury due to the expression of cytokines and chemokines in the heart and blood, there currently are large, multicenter, randomized, double-blind, placebo-controlled clinical trials recruiting patients to evaluate optimal time points to deliver bone-marrow mesenchymal stem cells (BMSCs) after moderate to large myocardial infarction, particularly in high-risk patients [29, 30].
To reach this stage in the field of urology, or more specifically, incontinence, more basic science research comparing type of cells, dosages, timing of optimal dose or doses after injury, mechanism of action of stem cells, and route of administration must be performed. This will enable large multicenter clinical trials to focus on functional improvements, different treatments for stress, urge, or mixed incontinence, and potential adverse events. This article reviews the progress in stem cell research to treat or prevent incontinence and describes areas of future work as suggested by research in other fields.
Overview of Stem Cells
Stem cells are the current basis for tissue engineering and regenerative medicine. They are located in various parts of the body and replenish tissues, bone, and blood throughout life as well as enable healing after injury [31]. Stem cells are described by their differentiation potential: embryonic stem cells are totipotent, while adult stem cells are pluripotent or multipotent and cannot be differentiated into as many different types of tissues as embryonic cells [32]. Adult stem cells are, for the most part, tissue- or niche-specific, although the unique and defining markers for each stem cell population have not been found in vivo [31]. It is unknown whether adult stem cells are the same population of cells residing in different niches or separate populations with distinct characteristics and/or lineages [31]. These various questions arise because stem cells do not exhibit consistent signaling profiles or surface markers; rather, they act based on the cells and niche around them [31]. Because of this, various efforts to answer these questions have emerged. One study found mesenchymal stem cells with similar characteristics in many organs, including heart, kidney, spleen, and brain, and suggested that there is a common perivascular origin [33]. Other groups have compared stem cells isolated from adipose tissue, bone marrow, and umbilical cord blood and have found many similarities in morphology, phenotype, and gene expression [34, 35]. Further studies are needed to identify distinct characteristics and capabilities of each stem cell population, or if they are the same, how stem cells behave in different niches and what combination is the most effective.
Currently, a barrier to tissue engineering for clinical therapy is the lack of an adequate vascular supply for larger-volume implants [36]. Cells need to be within a tenth of a millimeter of a capillary to get the required oxygen for their processes as well as to remove waste products [37]. With the potential to create organs from stem cells, this problem can be overcome because it has been shown that stem cells induce neovascularization [9]. Although low cell retention and survival rate after transplantation are major problems with cell-based therapy, there is functional improvement despite the low number of engrafted cells in multiple injury models such as spinal cord injury, stroke, and myocardial infarction [22, 24, 38, 39].
Various mechanisms have been proposed to explain the reparative effects of stem cells because they are able to self-proliferate, differentiate, suppress immune function, release paracrine factors, and home to sites of injury [40]. Some researchers believe that direct engraftment and/or stem cell fusion or differentiation is responsible for improvement, while others believe trophic or paracrine factors influence the cells and microenvironment of the damaged tissue or organ. There is support for both arguments across fields, with some studies showing evidence of transdifferentiation or fusion in the target organ after direct injection to the injured area [41, 42] and others specifically showing that no fusion occurred [43]. Functional improvement also occurs when stem cells are injected into a distant uninjured muscle, indicating that factors released by stem cells distant to the injury may enable the reparative process [44••]. Injection of stem cell–produced factors or homing cytokines alone, without the actual stem cells, also has demonstrated functional improvement [16, 17, 45]. Specifically for incontinence, it has been shown that stem cell homing cytokines specific to mesenchymal stem cells, chemokine (C-C motif) ligand 7 (previously monocyte chemotactic protein 3) and its receptor are upregulated in the urethra and vagina after a simulated childbirth injury in rats, suggesting that homing after injury is possible [46, 47]. In contrast, stromal cell–derived factor 1, the stem cell homing cytokine specific to hematopoietic stem cells, is not upregulated after this injury [46, 47].
It is currently uncertain whether stem cells should be injected as is or differentiated first into myoblasts, since myoblasts will only fill the defect as needed and then stop multiplying, controlled presumably by cytokines and other factors [48]. However, it has been shown that stem cells generate larger grafts, have higher survival rates, and more effectively remodel injured cardiac tissue compared with myoblasts [9]. Overall, more is unknown than known in terms of optimal cell type, route of administration, mechanism of action, and the importance, or lack thereof, of engraftment and long-term survival.
Embryonic Stem Cells
Embryonic stem cells are totipotent and have the capability to differentiate into the three embryonic germ cell layers (endoderm, mesoderm, and ectoderm), making them attractive for use in regenerative medicine [40]. Although embryonic stem cells have not been used for incontinence, they have been utilized to investigate the possible creation of renal tubules or generation of functional bioartificial renal units for patients with renal failure [49, 50]. They also were used to create sperm in previously infertile male mice, although there has not yet been successful gamete creation [51, 52]. However, there are serious concerns about tumorigenicity [53]. Furthermore, the political and ethical debates concerning obtaining and using embryonic stem cells limit their current potential therapeutic use [54].
Bone Marrow–Derived Mesenchymal Stem Cells
Bone marrow contains heterogenous populations of cells, including erythrocytes, macrophages, endothelial cells, fibroblasts, and adipocytes, as well as hematopoietic and mesenchymal stem cells [40]. BMSCs currently are the principally used cells both preclinically and clinically [40]; however, they are less commonly utilized in urology or incontinence research [8]. This may be due to the fact that although BMSCs are pluripotent cells characterized as capable of differentiating easily into osteogenic, adipogenic, and chondrogenic lineages [55, 56], the procedure for bone marrow removal/aspiration is painful, requiring general or spinal anesthesia, and may be more tolerable for cardiac injury as opposed to the treatment of incontinence [57, 58].
BMSCs injected in two sites periurethrally 2 weeks after a bilateral pudendal nerve transection were found to improve function as assessed by measurement of leak point pressure (LPP) and urethral closure pressure 1 month after injection [59•]. The injected cells stained positive for smooth muscle actin, vimentin, and desmin and formed dorsolateral muscle masses at the injection sites, effectively compressing the urethral lumen and acting as a bulking agent [59•]. Similar results have been observed in experiments in vitro when BMSCs stained positive for desmin and smooth muscle actin and expressed myosin heavy chain [11, 60], indicating differentiation of BMSCs into smooth and striated muscle under appropriate conditions. When BMSCs were implanted periurethrally into atrophic urethras (damaged by pudendal nerve transection), the sites of injection and newly formed tissue stained positive for desmin and myosin heavy chain, indicating the presence of striated skeletal muscle as confirmed by histological analysis and improved LPP [60].
These results differ somewhat from that of Kinebuchi et al. [61•], who found no difference in LPP with a urethrolysis injury between control and BMSCs, but did note that BMSCs differentiated into striated muscle and peripheral nerve cells but not smooth muscle. It is possible these differences exist because of the differences in injury models, time point of injection after injury, and functional test time point after injury.
In one interesting study, BMSCs were combined with a sling model in rats after bilateral sciatic nerve transection [62•]. While both a sling with and without BMSCs demonstrated good functional outcomes using LPP, the BMSC-seeded sling had a better ligament-like formation after 12 weeks, higher force to failure, and more collagen matrix deposition [62•]. A combination stem cell sling possibly is a promising integration method that may provide long-term success for both structural and functional support of the deficient urethral sphincter in humans.
Muscle-Derived Stem Cells
Muscle tissue represents a valuable and easily accessible source of adult stem cells. Their procurement entails minimal morbidity and can be done under local anesthesia [63]. Though in the past there were technical difficulties associated with growing these cells and selecting for stem cells, this technical difficulty seems to have been overcome by various preplating techniques [64–66]. In short, to obtain MDSCs, muscle tissue is enzymatically digested, then cultured for a short period of time to allow fibroblasts to attach to the plate or flask. The supernatant that now contains the myogenic fraction, and therefore, the MDSC precursor, is then transferred to a new flask, thereby enriching for the desired cells which are collected several days after seeding [66]. Several studies have shown that MDSCs are capable of proliferation, multipotent differentiation, and self-renewal [66, 67], indicating their potential for tissue regeneration.
When compared with saline treatment after transection of the sciatic nerve in rats, both fibroblasts and MDSCs injected intraurethrally increased LPP, but muscle of MDSC-injected urethras contracted with 27% greater amplitude, while the fibroblast-injected urethras eventually became obstructed [68]. Furuta et al. [58] also investigated LPP and contractile responses of the proximal urethra 6 weeks after an injection of human MDSCs into the proximal urethras of female nude rats that had previously received bilateral pudendal nerve transection [58]. There was no significant difference between the LPP in sham-operated saline-treated rats and nerve transected rats injected with MDSCs, indicating a return to normal in the stem cell–treated animals. However, after application of hexamethonium, which blocks autonomic efferent nerves, the LPP in nerve-transected rats with MDSCs was significantly decreased compared with the sham-operated saline-treated group. The contractile responses to phenylephrine, but not to carbachol, in nerve-transected rats with MDSCs were significantly increased compared with sham-operated, saline-treated and nerve-transected, saline-treated groups [58]. These results suggest that the functional improvements mediated by MDSCs are due to improvements in smooth muscle innervated by autonomic nerves [58].
In a pudendal nerve denervation model in rats, Lee et al. [69] showed that a periurethral injection of MDSCs increases LPP and urethral closure pressure 4 and 12 weeks after injury, and was superior to bovine collagen 12 weeks after injury. There was no immunogenic reaction or severe lymphocyte reactions observed at the site of injection [69]. The investigators observed an increase in skeletal muscle mass at the sites of injection, suggesting conditions different from that of the experiments of Furuta et al. [58], who demonstrated that functional improvements were due to changes in smooth muscle.
When MDSCs were mixed with fibrin glue and injected into the urethra after pudendal nerve transection in rats, the number of surviving MDSCs was significantly increased [70]. There was an increased muscle-to-collagen ratio and microvessel density, demonstrating that fibrin glue may potentially promote cell attachment and proliferation [70]. Chancellor et al. [71] demonstrated that MDSCs are capable of proliferation to form myoblasts in vivo, which then will fuse, when mature, in postmitotic (i.e., no longer dividing) myotubes, limiting growth and avoiding the risk of a potential obstruction of the urinary tract [71]. Kajbafzadeh et al. [72] injected autologous myoblasts transurethrally followed by postoperative pelvic floor electrical stimulation, which demonstrated an improvement of urinary incontinence in children with bladder exstrophy, indicating the potential to combine the myoblasts or MDSCs with traditional treatment.
All clinical trials on incontinence have used autologous MDSCs and injected them directly into the urethra either transurethrally or periurethrally. The first was done by Strasser et al. [73] with improvements in quality of life and thickness and contractility of the urethral sphincter, with a success rate of over 90% for women and over 50% for men [73]. However, this study was later retracted due to breaches in ethical guidelines and protocol irregularities [74]. In North America, a small clinical trial with eight women showed more modest and realistic improvements and changes, with one patient achieving complete continence and four indicating improvement from baseline 1 year after the initial injection [75]. A larger, blinded, and randomized trial by the same group had an almost 50% cure rate in patients 1 year after two injections and other slightly different techniques as evidenced by the increased dry percentage [13].
Preliminary clinical data from a multicenter study demonstrated that intrasphincteric injection of autologous MDSCs at various doses reduced the amount of leakage during a 24-hour pad test and lowered the incidence of diary-reported stress leaks over 3 days. These improvements started as early as 1 month after injection and lasted through the study’s end point of 6 months, with some evidence suggesting that the higher doses of MDSCs are associated with greater and faster improvement of incontinence symptoms, although not all patients improved (even at the highest dose or the longest time point) and no cure or “dry” rates were reported [76]. All minor complications associated with treatment occurred at low rates and either resolved independently or were easily treated, suggesting the feasibility of utilizing MDSCs as a therapy for SUI. Because most preclinical studies are done in acute injury models, this is an important result demonstrating stem cell efficacy long after the injury has occurred. Although the authors did not speculate on the mechanism of action of the cells, it is interesting to note that the effect of treatment occurred as late as 1 to 6 months after the actual injection.
Adipose-Derived Stem Cells
Adipose-derived stem cells (ADSCs) are plentiful and can be obtained by minimally invasive lipoaspiration of the abundant adipose tissue in humans [77]. Several studies have shown that ADSCs are multipotent cells and are capable of differentiating into adipogenic, chondrogenic, myogenic, and osteogenic cells [57, 77]. Rodriguez et al. [78] showed that ADSCs can be obtained from processed lipoaspirate and are able to differentiate into functional smooth muscle cells capable of contraction with cholinergic stimulation, followed by relaxation similar to normal muscle tissue.
In a rat postpartum vaginal balloon dilation and bilateral ovariectomy model of simulated childbirth, ADSCs were derived from periovarian fat and then reinjected either via the tail vein or directly into the urethra [79••]. Both injection routes resulted in a higher concentration of elastin and smooth muscle in the urethra as well as an improvement in voiding function 4 weeks after injection. ADSCs were detected in the connective tissue along and underneath the urothelium [79••].
ADSCs combined with nerve growth factor (NGF) encapsulated within poly(lactic-co-glycolic acid), or PLGA, microspheres were injected directly into the urethra by Zhao et al. [80•]. NGF-PLGA microspheres increased cell survival and retention in the urethra, and 8 weeks after treatment, rats with ADSCs and NGF-PLGA microspheres were recovered significantly more than all the other groups (saline-treated [phosphate-buffered saline], ADSC and NGF only, ADSC and PLGA only, and ADSC-only treated groups). Both LPP and retrograde urethral perfusion pressure, returned to levels comparative to that of an uninjured rat [80•].
ADSCs also have undergone induction into myoblasts and appear to be typical smooth cells expressing sarcomeric and desmin proteins both in vitro [12] and in vivo after direct injection into the urethra [10]. Fu et al. [10] used an injury model combining ovariectomy followed by a weighted vaginal distention intended to model a difficult childbirth and menopause Myoblasts (induced from ADSCs) or ADSCs (as a control) were injected 3 months after the injury, and there was no difference between injections 1 month after injection; however, at 3 months after treatment, the rats that received a myoblast injection had fully restored LPP and functional bladder capacity [10]. Animals with a myoblast injection had thicker smooth muscle and more longitudinal muscle fibers explaining the increased function as compared with ADSC-treated rats. This is an interesting result that conflicts with other researchers who found myoblasts did not have a positive effect on left ventricular function after infarction in a blinded randomized trial [81] and that myoblasts are inferior when compared to MDSCs in grafting and surviving oxidative stress, among other cases [9]. Furthermore, other researchers have had success with ADSCs, although Zhao et al. [80•] found that ADSCs alone did not demonstrate as much recovery as ADSCs mixed with NGF-PLGA microspheres. Perhaps the differences are due to differences in processing and isolation, because in the study by Fu et al. [10], neither the ADSCs nor myoblasts were selected for cell surface markers, so the populations are potentially heterogenous and mixed.
In a clinical study, ADSCs harvested and purified from lipoaspiration were injected periurethrally in two male patients after radical prostatectomy [82]. Urinary incontinence progressively improved after 2 weeks of injection and persisted up to 12 weeks after treatment, with decreases in leakage volume and increases in quality of life, maximal urethral closure pressure, and functional urethral length. A bulking effect was demonstrated on magnetic resonance imaging 12 weeks after treatment, indicating that ADSCs may have contributed to the improvement of sphincteric function [82]. Interestingly, there was a sequential increase in blood flow to the region after ADSC injection that persisted for at least 12 weeks, potentially indicating the effect of cytokines secreted from stem cells causing neovascularization [83]. Unfortunately, this work was later retracted due to a breach of ethical research guidelines, and therefore, needs to be repeated and validated under appropriate ethical research approval [84].
Human Umbilical Cord Blood Stem Cell
Human umbilical-cord blood cells (HUCBs) are derived from the trophoblast and are not considered embryonic cells. Therefore, these cells potentially have an advantage over progenitor cells from other sources because of their young chronologic age [85]. Because stem cells from older patients divide fewer times and have less differentiation potential than stem cells from young patients [86], HUCBs may represent a useful source of cells for transplantation to restore sphincter function. In addition, using stem cells obtained from cord blood avoids invasive procedures [87].
Preclinically, Lim et al. [88] demonstrated that rats that received HUCB after electrocauterization of periurethral tissue had significantly improved LPP, and that histologically, the urethral sphincter of rats that received HUCBs was identical to that of an uninjured rat. In this study, most HUCBs were found in the lamina propria beneath the urethral mucosa, with a few cells found between the muscle layers of the urethral sphincter. Although the authors suggest that the functional improvement was not due to changes in the sphincter [88], paracrine effects from this nearby area are still possible, especially because after the injury and HUCB treatment, the sphincter was not disrupted as it was in the untreated animals.
HUCBs also have been tested in female patients with incontinence stemming from intrinsic sphincter deficiency, mixed incontinence, or urethral hypermobility [89]. At 12 months after treatment, 72% of patients had more than 50% improvement of their incontinence symptoms [89].
Other Sources
Potentially, there are stem cells sources for treatment of incontinence and urological reconstruction that involve less-invasive procedures than those described above. Hair follicle cells have been found to contain adult stem cells that are multipotent and differentiate into adipogenic, osteogenic, and epithelial lineages depending on isolation and culture procedures [90, 91]. Additionally, they were found to contain p63, a protein necessary for formation of urothelium [92]. An even less–invasive method for collecting cells would be from urine: two to seven multipotent stem cells have been isolated per 100 mL of urine, along with many other stem/progenitor cells that did not expand in culture, and cells that were partially or fully differentiated that washed off with the first media change [93]. Urine progenitor cells (about 0.2% of the cellular population in urine) stained positive for CD73, CD90 (Thy-1), and CD105 (endoglin), markers that are characteristic of mesenchymal cells, and stained negative for CD45, CD31, CD34, and other hematopoietic and endothelial lineage markers [93].
Conclusions
Although there has been a fair amount of preclinical research on utilization of stem cells and their derivatives as treatments for incontinence, much research still needs to be done before these therapies can be introduced into practice. Larger multicenter clinical trials that are double-blinded and placebo-controlled must be done; however, there is still much preclinical investigation to be done before we reach that point.
It is unknown which cell type or injection site is more effective in treatment, or if different cell types may be superior for different types of incontinence. All of these can be elucidated once various cell types and methods of administration are tested in the same animal injury model. Additionally, different stem cells may respond to injury in different ways, and stem cells used to treat incontinence may have a unique response due to the compound neuromuscular injury of childbirth. Thus, it may be difficult to extrapolate information from other fields into the field of incontinence. Therefore, the optimal cell type cannot be determined until these preclinical investigations occur.
It also is not known if the primary action of stem cells is by differentiation into smooth or striated muscle, by paracrine action of trophic factors produced by injected stem cells on innate tissues, by a combination of paracrine effects and immunosuppression, or by another mechanism as yet undelineated. It is important to note that the mechanism of action for stem cells is still not fully determined in any field, but there is some evidence for paracrine effects, as well as transdifferentiation [41–45].
Once the mechanistic action of each type of stem cell with therapeutic potential is determined, this information then can be used to determine what combination of stem cell source and method of administration is likely to be most effective in treating or preventing incontinence of various types. In addition, it ought to drive choices regarding the appropriate dose of cells and number of treatments. If retention of stem cells in the target organ is shown to be important for enhancing the mechanism of action of stem cells, substances that enhance cell retention and survival should be further investigated.
Aside from injecting cells for treatment of incontinence, stem cells may be combined with existing treatment options, such as slings, as evidenced by Zhou et al. [62•]. This potentially could improve the long-term efficacy of slings and reduce complications such as erosion or extrusion of synthetic slings, although this needs to be studied further both in preclinical animal models and in clinical trials.
In summary, although much work needs to be done both preclinically and clinically before stem cell therapy can replace surgery as the mainstay of treatment of stress urinary incontinence, the future looks promising.
Acknowledgements
Partial support for this publication provided by NIH R01 HD059859 and NIH R01 HD038679. This publication also was made possible by the Case Western Reserve University/Cleveland Clinic CTSA Grant Number UL1 RR024989 from NIH/National Center for Research Resources (NCRR), the Cleveland Clinic Glickman Urological and Kidney Institute: Section of Female Pelvic Medicine and Reconstructive Surgery, and the Rehabilitation Research & Development Service of the Department of Veterans Affairs.
Springer would like to thank Dr. Howard B. Goldman, Section Editor of the Voiding Dysfunction section, for his proposal and review of this article.
Footnotes
Disclosures No potential conflicts of interest relevant to this article were reported.
Contributor Information
Charuspong Dissaranan, Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH 44195, USA.
Michelle A. Cruz, Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH 44195, USA Department of Biomedical Engineering, Cleveland Clinic, 9500 Euclid Avenue, ND20, Cleveland, OH 44195, USA.
Bruna M. Couri, Department of Obstetrics and Gynecology, Cleveland Clinic, Cleveland, OH 44105, USA
Howard B. Goldman, Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH 44195, USA
Margot S. Damaser, Email: damasem@ccf.org, Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Department of Biomedical Engineering, Cleveland Clinic, 9500 Euclid Avenue, ND20, Cleveland, OH 44195, USA; Louis Stokes Cleveland Department of Veterans Administration Medical Center, Advanced Platform Technology Center, Cleveland, OH 44106, USA.
References
Papers of particular interest, published recently, have been highlighted as:
• Of importance
•• Of major importance
- 1.Charalambous S, Trantafylidis A. Impact of urinary incontinence on quality of life. Pelviperineology. 2009;28(2):51–53. [Google Scholar]
- 2.Milsom I. Lower urinary tract symptoms in women. Curr Opin Urol. 2009;19(4):337–341. doi: 10.1097/MOU.0b013e32832b659d. [DOI] [PubMed] [Google Scholar]
- 3.Nilsson CG, Palva K, Rezapour M, Falconer C. Eleven years prospective follow-up of the tension-free vaginal tape procedure for treatment of stress urinary incontinence. Int Urogynecol J. 2008;19(8):1043–1047. doi: 10.1007/s00192-008-0666-z. [DOI] [PubMed] [Google Scholar]
- 4.Olsen AL, Smith VJ, Bergstrom JO, Colling JC, Clark AL. Epidemiology of surgically managed pelvic organ prolapse and urinary incontinence. Obstet Gynecol. 1997;89:501–506. doi: 10.1016/S0029-7844(97)00058-6. [DOI] [PubMed] [Google Scholar]
- 5.Rogers RG. What’s best in the treatment of stress urinary incontinece? New Engl J Med. 2010;362(22):2124–2125. doi: 10.1056/NEJMe1005367. [DOI] [PubMed] [Google Scholar]
- 6.Albo ME, Richter HE, Brubaker L, Norton P, Kraus SR, Zimmern PE, et al. Burch colposuspension versus fascial sling to reduce urinary stress incontinence. New Engl J Med. 2007;356(21):2143–2155. doi: 10.1056/NEJMoa070416. [DOI] [PubMed] [Google Scholar]
- 7.Gungorduk K, Celebi I, Ark C, Celikkol O, Yildirim G. Which type of mid-urethral sling procedure should be chosen for treatment of stress urinary incontinance with intrinsic sphincter deficiency? tension-free vaginal tape or transobturator tape. Acta Obstet Gynecol Scand. 2009;88(8):920–926. doi: 10.1080/00016340903100354. [DOI] [PubMed] [Google Scholar]
- 8.Staack A, Rodriguez LV. Stem cells for the treatment of urinary incontinence. Curr Urol Rep. 2011;12(1):41–46. doi: 10.1007/s11934-010-0155-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Oshima H, Payne TR, Urish KL, Sakai T, Ling YQ, Gharaibeh B, et al. Differential myocardial infarct repair with muscle stem cells compared to myoblasts. Mol Ther. 2005;12(6):1130–1141. doi: 10.1016/j.ymthe.2005.07.686. [DOI] [PubMed] [Google Scholar]
- 10.Fu Q, Song XF, Liao GL, Deng CL, Cui L. Myoblasts differentiated from adipose-derived stem cells to treat stress urinary incontinence. Urology. 2010;75(3):718–723. doi: 10.1016/j.urology.2009.10.003. [DOI] [PubMed] [Google Scholar]
- 11.Drost AC, Weng S, Feil G, Schafer 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. Hematopoietic Stem Cells Vii. 2009;1176:135–143. doi: 10.1111/j.1749-6632.2009.04610.x. [DOI] [PubMed] [Google Scholar]
- 12.Wu GZ, Zheng X, Jiang ZQ, Wang JH, Song YF. Induced differentiation of adipose-derived stromal cells into myoblasts. J Huazhong Univ Sci Technol Med Sci. 2010;30(3):285–290. doi: 10.1007/s11596-010-0344-5. [DOI] [PubMed] [Google Scholar]
- 13.Herschorn S, Carr L, Birch C, Murphy M, Robert M, Jankowski R, et al. Autologous muscle-derived cells as therapy for stress urinary incontinence: a randomized, blinded trial. Neurourol Urodyn. 2010;29(2):307. [Google Scholar]
- 14.Tang XL, Rokosh G, Sanganalmath SK, Pang Y, Yuan FP, Dai SJ, et al. Beneficial effects of cardiac progenitor cells on LV function 1 year after treatment in rats with myocardial infarction. Circulation. 2008;118(18):S289–S290. [Google Scholar]
- 15.Amado LC, Schuleri KH, Saliaris AP, Boyle AJ, Helm R, Oskouei B, et al. Multimodality noninvasive imaging demonstrates in vivo cardiac regeneration after mesenchymal stem cell therapy. J Am Coll Cardiol. 2006;48(10):2116–2124. doi: 10.1016/j.jacc.2006.06.073. [DOI] [PubMed] [Google Scholar]
- 16.Kuliszewski MA, Kobulnik J, Lindner JR, Stewart DJ, Leong-Poi H. Vascular gene transfer of SDF-1 promotes endothelial progenitor cell engraftment and enhances angiogenesis in ischemic muscle. Mol Ther. 2011;19(5):895–902. doi: 10.1038/mt.2011.18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Sundararaman S, Miller TJ, Pastore JM, Kiedrowski M, Aras R, Penn MS. Plasmid-based transient human stromal cell-derived factor-1 gene transfer improves cardiac function in chronic heart failure. Gene Ther. 2011 doi: 10.1038/gt.2011.18. In this paper, the authors demonstrate that increasing expression of stromal cell–derived factor-1 promoted angiogenesis and improved cardiac function in rats with ischemic heart failure, presumably by attracting stem cells to the region to facilitate recovery from the injury. These data demonstrate that stand-alone nonviral gene transfer is a strategy for improving cardiac function, and could possibly work for the lower urinary tract as well.
- 18.Gandia C, Arminan A, Garcia-Verdugo JM, Lledo E, Ruiz A, Minana MD, et al. Human dental pulp stem cells improve left ventricular function, induce angiogenesis, and reduce infarct size in rats with acute myocardial infarction. Stem Cells. 2008;26(3):638–645. doi: 10.1634/stemcells.2007-0484. [DOI] [PubMed] [Google Scholar]
- 19.Nagaya N, Fujii T, Iwase T, Ohgushi H, Itoh T, Uematsu M, et al. Intravenous administration of mesenchymal stem cells improves cardiac function in rats with acute myocardial infarction through angiogenesis and myogenesis. Am J Physiol Heart Circ Physiol. 2004;287(6):H2670–H2676. doi: 10.1152/ajpheart.01071.2003. [DOI] [PubMed] [Google Scholar]
- 20.Tang XL, Rokosh G, Guo YR, Bolli R. Cardiac progenitor cells and bone marrow-derived very small embryonic-like stem cells for cardiac repair after myocardial infarction. Circ J. 2010;74(3):390–404. doi: 10.1253/circj.cj-09-0923. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Hashemi SM, Ghods S, Kolodgie FD, Parcham-Azad K, Keane M, Hamamdzic D, et al. A placebo controlled, dose-ranging, safety study of allogenic mesenchymal stem cells injected by endomyocardial delivery after an acute myocardial infarction. Eur Hear J. 2008;29(2):251–259. doi: 10.1093/eurheartj/ehm559. [DOI] [PubMed] [Google Scholar]
- 22.Gnecchi M, He HM, Liang OD, Melo LG, Morello F, Mu H, et al. Paracrine action accounts for marked protection of ischemic heart by Akt-modified mesenchymal stem cells. Nat Med. 2005;11(4):367–368. doi: 10.1038/nm0405-367. [DOI] [PubMed] [Google Scholar]
- 23.Uemura R, Xu M, Ahmad N, Ashraf M. Bone marrow stem cells prevent left ventricular remodeling of ischemic heart through paracrine signaling. Circ Res. 2006;98(11):1414–1421. doi: 10.1161/01.RES.0000225952.61196.39. [DOI] [PubMed] [Google Scholar]
- 24.Gnecchi M, Zhang ZP, Ni AG, Dzau VJ. Paracrine mechanisms in adult stem cell signaling and therapy. Circ Res. 2008;103(11):1204–1219. doi: 10.1161/CIRCRESAHA.108.176826. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Hare JM, Traverse JH, Henry TD, Dib N, Strumpf RK, Schulman SP, et al. A randomized, double-blind, placebo-controlled, dose-escalation study of intravenous adult human mesenchymal stem cells (prochymal) after acute myocardial infarction. J Am Coll Cardiol. 2009;54(24):2277–2286. doi: 10.1016/j.jacc.2009.06.055. The authors report results that demonstrate that intravenous allogeneic human mesenchymal stem cells are safe to administer to patients with myocardial infarction. This clinical study provides pivotal safety and provisional efficacy data for allogeneic BMSCs in post-infarction patients and suggests this approach could be useful in therapy for other conditions.
- 26.Janssens S, Dubois C, Bogaert J, Theunissen K, Deroose C, Desmet W, et al. Autologous bone marrow-derived stem-cell transfer in patients with ST-segment elevation myocardial infarction: double-blind, randomised controlled trial. Lancet. 2006;367(9505):113–121. doi: 10.1016/S0140-6736(05)67861-0. [DOI] [PubMed] [Google Scholar]
- 27.Assmus B, Rolf A, Erbs S, Elsasser A, Haberbosch W, Hambrecht R, et al. Clinical outcome 2 years after intracoronary administration of bone marrow-derived progenitor cells in acute myocardial infarction. Circ Heart Fail. 2010;3(1):89–96. doi: 10.1161/CIRCHEARTFAILURE.108.843243. [DOI] [PubMed] [Google Scholar]
- 28.Bartunek J, Dimmeler S, Drexler H, Fernandez-Aviles F, Galinanes M, Janssens S, et al. The consensus of the task force of the European Society of Cardiology concerning the clinical investigation of the use of autologous adult stem cells for repair of the heart. Eur Hear J. 2006;27(11):1338–1340. doi: 10.1093/eurheartj/ehi793. [DOI] [PubMed] [Google Scholar]
- 29.Traverse JH, Henry TD, Vaughn DE, Ellis SG, Pepine CJ, Willerson JT, et al. Rationale and design for TIME: a phase II, randomized, double-blind, placebo-controlled pilot trial evaluating the safety and effect of timing of administration of bone marrow mononuclear cells after acute myocardial infarction. Am Hear J. 2009;158(3):356–363. doi: 10.1016/j.ahj.2009.06.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Traverse JH, Henry TD, Vaughan DE, Ellis SG, Pepine CJ, Willerson JT, et al. LateTIME a phase-ii, randomized, double-blinded, placebo-controlled, pilot trial evaluating the safety and effect of administration of bone marrow mononuclear cells 2 to 3 weeks after acute myocardial infarction. Tex Hear Inst J. 2010;37(4):412–420. [PMC free article] [PubMed] [Google Scholar]
- 31.Morrison SJ, Spradling AC. Stem cells and niches: mechanisms that promote stem cell maintenance throughout life. Cell. 2008;132(4):598–611. doi: 10.1016/j.cell.2008.01.038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Shokeir AA, Harraz AM, El-Din ABS. Tissue engineering and stem cells: basic principles and applications in urology. Int J Urol. 2010;17(12):964–973. doi: 10.1111/j.1442-2042.2010.02643.x. [DOI] [PubMed] [Google Scholar]
- 33.Meirelles LDS, Chagastelles PC, Nardi NB. Mesenchymal stem cells reside in virtually all post-natal organs and tissues. J Cell Sci. 2006;119(11):2204–2213. doi: 10.1242/jcs.02932. [DOI] [PubMed] [Google Scholar]
- 34.Lee RH, Kim B, Choi I, Kim H, Choi HS, Suh K, et al. Characterization and expression analysis of mesenchymal stem cells from human bone marrow and adipose tissue. Cell Physiol Biochem. 2004;14(4–6):311–324. doi: 10.1159/000080341. [DOI] [PubMed] [Google Scholar]
- 35.Kern S, Eichler H, Stoeve J, Kluter H, Bieback K. Comparative analysis of mesenchymal stem cells from bone marrow, umbilical cord blood, or adipose tissue. Stem Cells. 2006;24(5):1294–1301. doi: 10.1634/stemcells.2005-0342. [DOI] [PubMed] [Google Scholar]
- 36.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(7–8):1034–1044. doi: 10.1089/ten.2005.11.1034. [DOI] [PubMed] [Google Scholar]
- 37.Goodsell DS. The molecular perspective: VEGF and angiogenesis. Stem Cells. 2003;21(1):118–119. doi: 10.1634/stemcells.21-1-118. [DOI] [PubMed] [Google Scholar]
- 38.Prockop DJ, Gregory CA, Spees JL. One strategy for cell and gene therapy: harnessing the power of adult stem cells to repair tissues. Proc Natl Acad Sci U S A. 2003;100:11917–11923. doi: 10.1073/pnas.1834138100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Zhao LR, Duan WM, Reyes M, Keene CD, Verfaillie CM, Low WC. Human bone marrow stem cells exhibit neural phenotypes and ameliorate neurological deficits after grafting into the ischemic brain of rats. Exp Neurol. 2002;174(1):11–20. doi: 10.1006/exnr.2001.7853. [DOI] [PubMed] [Google Scholar]
- 40.Salem HK, Thiemermann C. Mesenchymal stromal cells: current understanding and clinical status. Stem Cells. 2010;28(3):585–596. doi: 10.1002/stem.269. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Amado LC, Saliaris AP, Schuleri KH, St John M, Xie JS, Cattaneo S, et al. Cardiac repair with intramyocardial injection of allogeneic mesenchymal stem cells after myocardial infarction. Proc Natl Acad Sci U S A. 2005;102(32):11474–11479. doi: 10.1073/pnas.0504388102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Sasaki M, Abe R, Fujita Y, Ando S, Inokuma D, Shimizu H. Mesenchymal stem cells are recruited into wounded skin and contribute to wound repair by transdifferentiation into multiple skin cell type. J Immunol. 2008;180(4):2581–2587. doi: 10.4049/jimmunol.180.4.2581. [DOI] [PubMed] [Google Scholar]
- 43.Duffield JS, Bonventre JV. Kidney tubular epithelium is restored without replacement with bone marrow-derived cells during repair after ischemic injury. Kidney Int. 2005;68(5):1956–1961. doi: 10.1111/j.1523-1755.2005.00629.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Shabbir A, Zisa D, Suzuki G, Lee T. Heart failure therapy mediated by the trophic activities of bone marrow mesenchymal stem cells: a noninvasive therapeutic regimen. Am J Physiol Heart Circ Physiol. 2009;296(6):H1888–H1897. doi: 10.1152/ajpheart.00186.2009. The authors demonstrated that intramuscularly injected mesenchymal stem cells (MSCs) and MSC-conditioned medium each significantly improved ventricular function in a hamster heart failure model, suggesting that cell therapy does not need to be local to be effective. Because multiple tissues are damaged during delivery, this suggests that a less invasive approach could be utilized to deliver a cell-based therapy.
- 45.Dissaranan C, Gill B, Cruz M, Salcedo L, Brian B, Kiedrowski M, et al. Intravenous mesenchymal stem cells facilitate recovery from stress urinary incontinence after childbirth injury. Neurourol Urodyn. 2011;30(2):235. [Google Scholar]
- 46.Woo LL, Hijaz A, Kuang M, Penn MS, Damaser MS, Rackley RR. Overexpression of stem cell homing cytokines in rat pelvic organs following vaginal distension. J Urol. 2007;177:1568–1572. doi: 10.1016/j.juro.2006.11.047. [DOI] [PubMed] [Google Scholar]
- 47.Wood HM, Kuang M, Woo L, Hijaz A, Butler RS, Penn M, et al. Cytokine expression after vaginal distention of different durations in virgin Sprague-Dawley rats. J Urol. 2008;180(2):753–759. doi: 10.1016/j.juro.2008.03.182. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.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(9):1259–1265. doi: 10.1016/j.exger.2004.07.003. [DOI] [PubMed] [Google Scholar]
- 49.Lanza RP, Chung HY, Yoo JJ, Wettstein PJ, Blackwell C, Borson N, et al. Generation of histocompatible tissues using nuclear transplantation. Nat Biotechnol. 2002;20(7):689–696. doi: 10.1038/nbt703. [DOI] [PubMed] [Google Scholar]
- 50.Minuth WW, Sorokin L, Schumacher K. Generation of renal tubules at the interface of an artificial interstitium. Cell Physiol Biochem. 2004;14(4–6):387–394. doi: 10.1159/000080348. [DOI] [PubMed] [Google Scholar]
- 51.Geijsen N, Horoschak M, Kim K, Gribnau J, Eggan K, Daley GQ. In vitro generation of male germ cells from embryonic stem cells. Biol Reprod. 2004;87 doi: 10.1038/nature02247. [DOI] [PubMed] [Google Scholar]
- 52.Toyooka Y, Tsunekawa N, Akasu R, Noce T. Embryonic stem cells can form germ cells in vitro. Proc Natl Acad Sci U S A. 2003;100(20):11457–11462. doi: 10.1073/pnas.1932826100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Przyborski SA. Differentiation of human embryonic stem cells after transplantation in immune-deficient mice. Stem Cells. 2005;23(9):1242–1250. doi: 10.1634/stemcells.2005-0014. [DOI] [PubMed] [Google Scholar]
- 54.Jain KK. Ethical and regulatory aspects of embryonic stem cell research. Expert Opin Biol Ther. 2005;5(2):153–162. doi: 10.1517/14712598.5.2.153. [DOI] [PubMed] [Google Scholar]
- 55.Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca JD, et al. Multilineage potential of adult human mesenchymal stem cells. Science. 1999;284(5411):143–147. doi: 10.1126/science.284.5411.143. [DOI] [PubMed] [Google Scholar]
- 56.Delorme B, Charbord P. Culture and characterization of human bone marrow mesenchymal stem cells. Tissue Eng Meth Mol Med. 2007;140:67–81. doi: 10.1007/978-1-59745-443-8_4. [DOI] [PubMed] [Google Scholar]
- 57.Nikolavsky D, Chancellor MB. Stem cell therapy for stress urinary incontinence. Neurourol Urodyn. 2010;29:S36–S41. doi: 10.1002/nau.20833. [DOI] [PubMed] [Google Scholar]
- 58.Furuta A, Jankowski RJ, Pruchnic R, Egawa S, Yoshimura N, Chancellor MB. Physiological effects of human muscle-derived stem cell implantation on urethral smooth muscle function. Int Urogynecol J. 2008;19(9):1229–1234. doi: 10.1007/s00192-008-0608-9. [DOI] [PubMed] [Google Scholar]
- 59. Kim SO, Na HS, Kwon D, Joo SY, Kim HS, Ahn Y. Bone-marrow-derived mesenchymal stem cell transplantation enhances closing pressure and leak point pressure in a female urinary incontinence rat model. Urol Int. 2011;86(1):110–116. doi: 10.1159/000317322. The authors demonstrated that allogenic MSCs can increase LPP in a rat model, of, SUI, providing preliminary data for clinical trials in this area.
- 60.Corcos J, Ghoniem G, Comiter C, Westney OL, Herschorn S. Durability of macroplastioue (R) injection for female stress urinary incontinence: 2 years experience. Neurourol Urodyn. 2010;29(2):275. [Google Scholar]
- 61. Kinebuchi Y, Aizawa N, Imamura T, Ishizuka O, Igawa Y, Nishizawa O. Autologous bone-marrow-derived mesenchymal stem cell transplantation into injured rat urethral sphincter. Int J Urol. 2010;17(4):359–368. doi: 10.1111/j.1442-2042.2010.02471.x. The authors treated rats that had undergone urethrolysis and periuethral cardiotoxin injection with autologous BMSCs. They found that the proportions of skeletal muscle cells and peripheral nerves in the urethra were significantly greater in the BMSC group compared to the control group, suggesting that stem cells can facilitate reinnervation of the urethra after a denervation injury.
- 62. Zou XH, Zhi YL, Chen X, Jin HM, Wang LL, Jiang YZ, et al. Mesenchymal stem cell seeded knitted silk sling for the treatment of stress urinary incontinence. Biomater. 2010;31(18):4872–4879. doi: 10.1016/j.biomaterials.2010.02.056. The authors developed a tissue-engineered sling with bone marrow–derived mesenchymal stem cell–seeded degradable silk scaffold. They found that both the silk sling and tissue-engineered sling showed convincing functional effects for the treatment of SUI in a rat model. The better ligament-like tissue formation in the tissue-engineered sling suggested the potential for improved long-term function with less failure.
- 63.Usas A, Huard J. Muscle-derived stem cells for tissue engineering and regenerative therapy. Biomaterials. 2007;28(36):5401–5406. doi: 10.1016/j.biomaterials.2007.09.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Li Y. Isolating stem cells from soft musculoskeletal tissues. J Vis Exp. 2010;5(41) doi: 10.3791/2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Qu ZQ, Balkir L, van Deutekom JCT, Robbins PD, Pruchnic R, Huard J. Development of approaches to improve cell survival in myoblast transfer therapy. J Cell Biol. 1998;142(5):1257–1267. doi: 10.1083/jcb.142.5.1257. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Wu XY, Wang SL, Chen BL, An XL. Muscle-derived stem cells: isolation, characterization, differentiation, and application in cell and gene therapy. Cell Tissue Res. 2010;340(3):549–667. doi: 10.1007/s00441-010-0978-4. [DOI] [PubMed] [Google Scholar]
- 67.Deasy BM, Jankowski RJ, Huard J. Muscle-derived stem cells: characterization and potential for cell-mediated therapy. Blood Cell Mol Dis. 2001;27(5):924–933. doi: 10.1006/bcmd.2001.0463. [DOI] [PubMed] [Google Scholar]
- 68.Kwon D, Kim Y, Pruchnic R, Jankowski R, Usiene I, De Miguel F, et al. Periurethral cellular injection: comparison of muscle-derived progenitor cells and fibroblasts with regard to efficacy and tissue contractility in an animal model of stress urinary incontinence. Urology. 2006;68(2):449–454. doi: 10.1016/j.urology.2006.03.040. [DOI] [PubMed] [Google Scholar]
- 69.Lee JY, Paik SY, Yuk SH, Lee JH, Ghil SH, Lee SS. Long term effects of muscle-derived stem cells on leak point pressure and closing pressure in rats with transected pudendal nerves. Mol Cell. 2004;18(3):309–313. [PubMed] [Google Scholar]
- 70.Xu Y, Song Y. Transplantation of muscle-derived stem cell plus fibrin glue restores urethral function in a pudendal nerve-transected rat model. Neurourol Urodyn. 2009;28(7):822–823. doi: 10.1590/s0100-879x2010007500112. [DOI] [PubMed] [Google Scholar]
- 71.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(3):279–287. doi: 10.1002/(sici)1520-6777(2000)19:3<279::aid-nau9>3.0.co;2-m. [DOI] [PubMed] [Google Scholar]
- 72.Kajbafzadeh AM, Elmi A, Payabvash S, Salmasi AH, Saeedi P, Mohamadkhani A, et al. Transurethral autologous myoblast injection for treatment of urinary incontinence in children with classic bladder exstrophy. J Urol. 2008;180(3):1098–1105. doi: 10.1016/j.juro.2008.05.057. [DOI] [PubMed] [Google Scholar]
- 73.Strasser H, Marksteiner R, Margreiter E, Mitterberger M, Pinggera GM, Frauscher F, et al. Transurethral ultrasonography-guided injection of adult autologous stem cells versus transurethral endoscopic injection of collagen in treatment of urinary incontinence. World J Urol. 2007;25(4):385–392. doi: 10.1007/s00345-007-0190-7. [DOI] [PubMed] [Google Scholar]
- 74.Kleinert S, Horton R. Retraction-autologous myoblasts and fibroblasts for treatment of stress urinary incontinence: a randomised controlled trial. Lancet. 2008;372(9641):789–790. doi: 10.1016/S0140-6736(08)61320-3. [DOI] [PubMed] [Google Scholar]
- 75.Carr LK, Steele D, Steele S, Wagner D, Pruchnic R, Jankowski R, et al. 1-year follow-up of autologous muscle-derived stem cell injection pilot study to treat stress urinary incontinence. Int Urogynecol J. 2008;19(6):881–883. doi: 10.1007/s00192-007-0553-z. [DOI] [PubMed] [Google Scholar]
- 76.Peters K, Kaufman M, Dmochowski R, Carr L, Hershorn S, Fischer M, et al. Autologous muscle derived cell therapy for the treatment of female stress urinary incontinence: a multi-center experience. J Urol. 2011;185(4):e535–e536. [Google Scholar]
- 77.Roche R, Festy F, Fritel X. Stem cells for stress urinary incontinence: the adipose promise. J Cell Mol Med. 2010;14(1–2):135–142. doi: 10.1111/j.1582-4934.2009.00915.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Rodriguez LV, Alfonso Z, Zhang R, Leung J, Wu B, Ignarro LJ. Clonogenic multipotent stem cells in human adipose tissue differentiate into functional smooth muscle cells. Proc Natl Acad Sci U S A. 2006;103(32):12167–12172. doi: 10.1073/pnas.0604850103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Lin GT, Wang GF, Banie L, Ning HX, Shindel AW, Fandel TM, et al. Treatment of stress urinary incontinence with adipose tissue-derived stem cells. Cytotherapy. 2010;12(1):88–95. doi: 10.3109/14653240903350265. The authors investigated whether transplantation of ADSCs can treat SUI in a rat model. They found that ADSC-treated groups had significantly higher elastin content than the control group, and within the ADSC-treated groups, rats with normal voiding pattern also had significantly higher elastin content than rats with voiding dysfunction. This suggests that proper elastin repair is important to restoration of function in treating SUI.
- 80. Zhao W, Zhang C, Jin C, Zhang Z, Kong L, Xu W, et al. Periurethral injection of autologous adipose-derived stem cells with controlled-release nerve growth factor for the treatment of stress urinary incontinence in a rat model. Eur Urol. 2011;59(1):155–163. doi: 10.1016/j.eururo.2010.10.038. The authors investigated if nerve growth factor facilitated the effect of ADSCs delivered periurethrally. They found significant improvements in LPP as well as in the amount of muscle and ganglia in the group that received nerve growth factor, suggesting that this and other growth factors could be utilized to facilitate recovery from SUI.
- 81.Menasch P, Alfieri O, Janssens S, McKenna W, Reichenspurner H, Trinquart L, et al. The myoblast autologous grafting in ischemic cardiomyopathy (MAGIC) trial—first randomized placebo-controlled study of myoblast transplantation. Circulation. 2008;117(9):1189–1200. doi: 10.1161/CIRCULATIONAHA.107.734103. [DOI] [PubMed] [Google Scholar]
- 82.Yamamoto T, Gotoh M, Hattori R, Toriyama K, Kamei Y, Iwaguro H, et al. Periurethral injection of autologous adipose-derived stem cells for the treatment of stress urinary incontinence in patients undergoing radical prostatectomy: report of two initial cases (Retracted article. See vol. 17, pg. 896, 2010) Int J Urol. 2010;17(1):75–82. doi: 10.1111/j.1442-2042.2009.02429.x. [DOI] [PubMed] [Google Scholar]
- 83.Rehman J, Traktuev D, Li JL, Merfeld-Clauss S, Temm-Grove CJ, Bovenkerk JE, et al. Secretion of angiogenic and antiapoptotic factors by human adipose stromal cells. Circulation. 2004;109(10):1292–1298. doi: 10.1161/01.CIR.0000121425.42966.F1. [DOI] [PubMed] [Google Scholar]
- 84.Retraction. Periurethral injection of autologous adipose-derived stem cells for the treatment of stress urinary incontinence in patients undergoing radical prostatectomy: report of two initial cases. Int J Urol. 2010;17(10):896. [PubMed] [Google Scholar]
- 85.Flynn A, Barry F, O'Brien T. UC blood-derived mesenchyrnal stromal cells: an overview. Cytotherapy. 2007;9(8):717–726. doi: 10.1080/14653240701584578. [DOI] [PubMed] [Google Scholar]
- 86.Roobrouck VD, Ulloa-Montoya F, Verfaillie CM. Self-renewal and differentiation capacity of young and aged stem cells. Exp Cell Res. 2008;314(9):1937–1944. doi: 10.1016/j.yexcr.2008.03.006. [DOI] [PubMed] [Google Scholar]
- 87.Kim JY, Jeon HB, Jang YS, Oh W, Jhang JW. Application of human umbilical cord blood-derived mesenchymal stem cells in disease models. World J Stem Cells. 2010;2(2):34–38. doi: 10.4252/wjsc.v2.i2.34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Lim JJ, Jang JB, Kim JY, Moon SH, Lee CN, Lee KJ. Human umbilical cord blood mononuclear cell transplantation in rats with intrinsic sphincter deficiency. J Kor Med Sci. 2010;25(5):663–670. doi: 10.3346/jkms.2010.25.5.663. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Lee CN, Jang JB, Kim JY, Koh C, Baek JY, Lee KJ. Human cord blood stem cell therapy for treatment of stress urinary incontinence. J Kor Med Sci. 2010;25(6):813–816. doi: 10.3346/jkms.2010.25.6.813. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Jahoda CAB, Whitehouse CJ, Reynolds AJ, Hole N. Hair follicle dermal cells differentiate into adipogenic and osteogenic lineages. Exp Dermatol. 2003;12(6):849–859. doi: 10.1111/j.0906-6705.2003.00161.x. [DOI] [PubMed] [Google Scholar]
- 91.Lavker RM, Sun TT, Oshima H, Barrandon Y, Akiyama M, Ferraris C, et al. Hair follicle stem cells. J Investig Dermatol Symp Proc. 2003;8(1):28–38. doi: 10.1046/j.1523-1747.2003.12169.x. [DOI] [PubMed] [Google Scholar]
- 92.Drewa T, Joachimiak R, Kaznica A, Sarafian V, Sir J. Primary cultures from rat vibrissae as a potential cell source for in vitro construction of urinary bladder wall grafts. Transplant Proc. 2009;41(5):1932–1935. doi: 10.1016/j.transproceed.2009.02.091. [DOI] [PubMed] [Google Scholar]
- 93.Zhang YY, McNeill E, Tian H, Soker S, Andersson KE, Yoo JJ, et al. Urine derived cells are a potential source for urological tissue reconstruction. J Urol. 2008;180(5):2226–2233. doi: 10.1016/j.juro.2008.07.023. [DOI] [PubMed] [Google Scholar]