14

Bladder tissue regeneration

F. Wezel and J. Southgate,    University of York, UK

Abstract:

This chapter reviews recent developments for novel regenerative medicine approaches for urinary bladder reconstruction. The chapter introduces clinical requirements for functional tissue replacement and discusses the use of synthetic and natural matrices for bladder reconstruction. It then describes the application of cell-seeded bio-matrices using adult progenitor and stem cells and provides an outlook for future directions in bladder tissue engineering, such as the use of multi- or pluri-potent stem cell sources.

Key words

urinary bladder; tissue engineering; cystoplasty; natural biomaterials; synthetic polymers; stem cells

14.1 Introduction

14.1.1 The bladder: structure and function

The bladder is a complex organ whose primary function is to store variable volumes of urine for extended periods of time. By retaining urine at safe, physiological pressures, the bladder protects the kidneys from damage (Thomas, 1997). The remarkable capacity and compliance of the bladder are dependent on the structural, biomechanical and biological properties of the smooth muscle wall and the highly specialised urothelial lining, which provides both urinary barrier and mechanosensory functions (Birder et al., 2012). In common with all tissue engineering, the ability to deliver successful (i.e. safe and functional) engineering of partial or whole bladder organ constructs requires fit-for-purpose biomaterials and a comprehensive understanding of bladder structure, cell/tissue biology and physiology. Given that until recently the urinary bladder was considered to be a passive urine storage organ, it is unsurprising that past attempts to reconstruct it with unsuitable materials have resulted in failure.

The mammalian bladder is composed of four distinct layers, with an outer serosal layer surrounding the detrusor muscle compartment, which is made up of three loosely arranged layers of smooth muscle (Fig. 14.1). Concentrically within this, the lamina propria is a viscoelastic collagenous connective tissue supporting a variety of cellular structures, including blood vessels, sensory and motor neurons. A basal lamina separates the lamina propria from the urothelium. The urothelium itself is a transitional epithelium comprising three stratified zones: a single row of basal cells attached to the basement membrane, several layers of intermediate cells and a single, overlying row of superficial ‘umbrella’ cells that abuts onto the luminal space. The function of the urothelium as a urinary barrier occurs primarily at the level of the superficial cells, with the paracellular barrier maintained by intercellular tight junctions (Acharya et al., 2004; Varley et al., 2006) and the transcellular barrier provided by multiple thickened plaques of asymmetric unit membrane (AUM) embedded in the outer leaflet of the apical membrane (Hicks, 1965). The AUM plaques are constituted by the interactions of four uroplakin (‘urothelium-plaque’) proteins and are a unique feature of urothelium (Wu et al., 1990; Yu et al., 1994; Olsburgh et al., 2003). The AUM plaques are formed in the Golgi apparatus and transported to the apical membrane as fusiform vesicles (Tu et al., 2002), thereby providing a source of membrane for accommodating changes in surface area and maintaining a low pressure environment during bladder filling.

image
14.1 Transverse section through the urinary bladder stained with haematoxylin and eosin to show tissue features. Scale bar 200 μm. (Acknowledgement for micrograph: Arianna Hustler and Edward Bowen.)

Such is the relationship between urothelial structure and function that loss of one component of the AUM can have devastating effects on urothelial structure and transcellular barrier properties (Hu et al., 2000, 2002). Thus, urothelial differentiation antigens not only provide objective markers of urothelial cytodifferentiation but, by virtue of their role in the urothelium, may also be regarded as surrogate markers of urinary barrier function. Unfortunately, expression of these markers is not invariably reported in bladder tissue-engineering reports, leading to discrepancy in the interpretation of some studies.

An important feature of the urothelium is its high regenerative capacity. Thus, although the urothelium is regarded as a ‘stable’ or quiescent tissue with an extremely slow rate of cell turnover, which may be as long as 200 days (Hicks, 1975), it is able to undergo rapid proliferation in response to acute injury (Peyton et al., 2012). Lavelle and colleagues performed a controlled study of selective urothelial damage in rats, which showed that recovery of transcellular and paracellular components of the urinary barrier occurred within 72 hours, with the intermediate cells undergoing rapid maturation to form differentiated umbrella cells (Lavelle et al., 2002). The excellent regenerative and differentiation capacity of urothelium is critical to maintaining the urine-proofing properties of the bladder and has positive implications for developing tissue-engineering strategies.

14.1.2 The clinical need for bladder reconstruction

With over 120 000 new cases reported in Europe (International Association for Cancer Research) and 150 000 in the USA (National Cancer Institute) in 2012, bladder cancer is the ninth most common cancer diagnosis worldwide. Those patients requiring cystectomy (mainly for muscle invasive bladder cancer) represent the largest group requiring surgical reconstruction of the lower urinary tract. Current approaches for urinary diversions following cystectomy commonly involve reconfiguring bowel in the form of orthotopic ileal neobladders, ileal conduit stomas or continent pouches (Studer et al., 2004; Stein and Skinner, 2006).

Benign disease processes or surgical interventions that render the bladder unstable, under high pressure, or lacking in capacity or compliance can result in a range of clinical problems ranging from mild to severe chronic urinary incontinence to irreversible kidney damage caused by raised upper tract pressures. Examples include neuropathic bladder (e.g. secondary to myelomeningocele, multiple sclerosis or spinal cord injury), severe detrusor instability and end-stage interstitial cystitis. There have been recent improvements in the medical management of these conditions using anticholinergics and especially, the intravesical injection of Botulinum Toxin-A (‘Botox’) or sacral neuromodulation/neurostimulation in selected cases (Fowler et al., 2012). However, a more permanent surgical augmentation remains the clinical need for those patients who develop a small-capacity, poorly compliant bladder, where intractable incontinence or pain destroys quality of life, or where serious kidney damage is imminent (Cain and Rink, 2010; Biers et al., 2012). As discussed in detail below (Section 14.3.1), although the surgical autoaugmentation of the bladder using bowel is considered the ‘gold standard’ treatment for end stage bladder disease, it is associated with significant clinical complications that are driving research to find alternative approaches.

14.2 Concepts, strategies and biomaterials for bladder reconstruction and tissue engineering

14.2.1 Concepts and strategies

An ideal tissue-engineered urinary bladder would mimic the range of functions fulfilled by the normal healthy bladder. During filling and voiding, the bladder undergoes dramatic changes in volume and is exposed to considerable mechanical forces. Adequate compliance is critical to accomplish the low pressure storage of urine and protection of the kidneys in the upper urinary tract. The development of sensory self-voiding function is outside current objectives and in all current and proposed bladder reconstruction strategies, it is anticipated that voluntary emptying will be aided by clean intermittent self-catherisation (CISC), either via the urethra or via an ileal conduit stoma or a vesicotomy, such as described by Mitrofanoff (1980).

Novel approaches for bladder reconstruction can be categorised as biomaterials-based, cell-based or combined (i.e. tissue engineering) strategies. The former, involving implantation of a biomaterial, is a passive approach that relies on the regenerative capacity of the host for full integration, whereby the material becomes cellularised and is eventually resorbed and replaced. The alternative is to harvest and expand cells from an appropriate host tissue in vitro, prior to transplanting the cells back into the body, with or without a biomaterial ‘scaffold’ to provide structure. The theoretical advantage of this latter approach is that clinically useful numbers of autologous cells are generated by propagation in the controlled environment of the laboratory, prior to surgical reimplantation into the host as a nascent or functional tissue.

A major challenge with all approaches is that where the underlying pathology of the host is unresolved, biomaterial integration and/or sourcing of fully functional, autologous cells for ex vivo tissue-engineering approaches may prove impossible. In this context, it is noteworthy that most experimental tissue-engineering models use healthy animals and problems can emerge when promising approaches are transferred to a clinical ‘disease setting’ (Bhargava et al., 2008).

14.2.2 Biomaterials

An ideal biomaterial scaffold should provide both structural support and adequate access to cells and nutrients to enable cells to engraft, survive, interact and be maintained. A more ambitious aim is that biomaterials provide instructive or ‘niche’ environments to support specific tissue development and differentiation. It is well-established that biomechanical properties such as stiffness and bioactive features that modulate cell-matrix interactions may influence cell phenotype and tissue function (Li and Xie, 2005; Rizvi and Wong, 2005; Rohman et al., 2007; Baker et al., 2009; Engelhardt et al., 2010). For example, human mesenchymal stem cells may be directed along neuronal, muscle or bone lineages by varying the stiffness of the scaffold (Engler et al., 2006). However, it is important to realise that there is no blueprint and tissue structure is not preformed in nature, but is an emergent property of development.

Several categories of biomaterial scaffold have been described for soft tissue applications:

• decellularised natural matrices produced from a variety of tissues, including small intestine submucosa (SIS) (Badylak et al., 1989; Zhang et al., 2000), bladder (Bolland et al., 2007) (Fig. 14.2), pericardium (Mirsadraee et al., 2007; da Costa et al., 2009) and dermis (Eberli et al., 2010);

• matrices produced from natural polymers (e.g. collagen (Gilbert, 2008), alginate (Rowley et al., 1999), chitosan (Drewa et al., 2008) and hyaluronan (Arimura et al., 2005));

• synthetic polymers including poly(ethylene glycol) (PEG) (Adelow et al., 2008), poly(lactic-co-glycolic acid) (PLGA) and poly(ε-caprolactone) (PCL) (Baker et al., 2009, 2011; reviewed by Aitken and Bagli, 2009; Wiesmann and Lammers, 2009).

image
14.2 Macroscopic appearance of a natural matrix derived from porcine bladder by decellularisation (Bolland et al., 2007). Decellularisation of the full thickness wall was achieved after distension and immersion of the intact bladder in a sequential series of sterile extraction buffers, including detergents and DNAse to lyse and remove cell components and render the tissue acellular. At the end of the procedure, the decellularised bladder is dissected open to present the biomaterial as a flattened sheet. The biomaterial retains many useful properties of the bladder wall including strength and compliance – as shown in these different fields. Actual size of material shown in relaxed state is 49 × 92 mm. (Acknowledgement for micrograph: Anna Radford.)

Scaffolds may be generated from extracted biological or synthetic polymers using a variety of processing techniques, including electrospinning (Baker et al., 2006), phase separation (Rowlands et al., 2007), gas foaming (Mooney et al., 1996), particulate leaching (McGlohorn et al., 2004; Baker et al., 2011), inkjet-printing (Roth et al., 2004) and chemical cross-linking (Park et al., 2002) (reviewed in Wiesmann and Lammers, 2009). These techniques have been used to create scaffolds of different shapes and porosity to facilitate cell engraftment. Scaffolds may be further functionalised by incorporation, surface adsorption or chemical attachment of growth and other bioactive factors, for example, to enhance angiogenesis and encourage vascularisation (Mikos et al., 1993; Wang et al., 2008; Rohman et al., 2009; Lee et al., 2010; reviewed by Chen et al., 2010; Kaully et al., 2009).

A particular advantage of animal-derived natural matrices is that following decellularisation, they retain tissue-specific architectures and extracellular matrix (ECM) proteins (Bolland et al., 2007), thus providing a wide range of biological and physical material properties specified by the nature of the originating tissue (reviewed by Gilbert et al., 2006; Davis et al., 2010). The high degree of conservation of matrix proteins between species (collagens, laminins and fibronectins) means that these matrices tend to be non-immunogenic and represent natural substrates for influencing cellular repopulation and tissue integration (Marcal et al., 2012).

14.3 Review of past and current strategies in bladder reconstruction

14.3.1 Vascularised tissue grafts

The use of reconfigured vascularised or pedicled host tissue grafts to augment the bladder has a long history. Currently, the most commonly performed procedure for end-stage bladder disease involves replacing or augmenting the bladder with a vascularised segment of the patient’s own bowel. This procedure of enterocystoplasty involves isolating a full thickness segment of bowel on its vascular pedicle, detubularising it along the antimesenteric border and incorporating it into the bi-valved bladder as an augmentation cytoplasty or, after cystectomy, as an orthotopic neo-bladder or conduit (Greenwell et al., 2001; Beier-Holgersen et al., 1994).

Enterocystoplasty was first described in a canine model in 1888 and then in man a year later, but it was not until the mid-twentieth century that the technique became popular for the treatment of the contracted, tuberculous bladder (Tizzoni and Foggi, 1888; Von-Mikulicz, 1889; Couvelaire, 1950). Stomach (gastrocystoplasty), small intestine (ileocystoplasty) and large intestine (colocystoplasty) have all been used as the reconstructing segment, but in the UK, ileocystoplasty is the most commonly performed procedure (Thomas, 1997).

Despite many patients experiencing the benefits of improved continence, improved urodynamic parameters and greater control over voiding, enterocystoplasty carries with it the potential for a number of serious complications. These are mainly attributable to the fact that bowel mucosa is structurally and physiologically unsuited to exposure to urine and include both early complications associated with all major abdominal surgery and specific, longer-term complications of enterocystoplasty, including spontaneous perforation of the bladder, mucus production by the bowel epithelium, bladder stones, bacteriuria, metabolic disturbances and malignancy (reviewed by Thomas, 1997; Greenwell et al., 2001).

Given that the side effects of enterocystoplasty are related to the long-term interaction of urine with the bowel mucosa, the logical progression would be to remove the bowel epithelium to leave the raw muscle surface facing the lumen – so-called seromuscular enterocystoplasty. Experimentally, in rabbit, canine, porcine and bovine surgical models, this approach has resulted in graft fibrosis and shrinkage, attributable to severe inflammation secondary to urinary exposure and irritation or infection of the graft and to ischaemia or damage to the intestine during dissection (Motley et al., 1990; Salle et al., 1990; Aktug et al., 2001; Fraser et al., 2004; Hafez et al., 2005). This phenomenon is independent of which side of the bowel wall faced the lumen. Severe fibrosis was also observed when a non-seeded vascularised capsule-flap of abdominal wall or gracilis muscle was incorporated into the rat bladder (Schoeller et al., 2004). However, in a series of 129 human bladder augmentations using demucosalised intestine, Lima and colleagues showed that fibrosis and shrinkage was prevented by the use of a silicon balloon left in the bladder for 2 weeks post-augmentation (Lima et al., 2004). Pedicled omental flaps to repair or augment the bladder (omentocystoplasty) have been largely successful both clinically and in animal models, particularly when used to close defects associated with vesico-vaginal fistula (Kiricuta and Goldstein, 1972).

Although demucosalisation of the bowel prior to incorporation into the bladder has inevitably resulted in graft fibrosis and shrinkage, when urothelium has been allowed to cover the augmenting graft, shrinkage occurred minimally or not at all (Aktug et al., 2001; Schoeller et al., 2004; Hafez et al., 2005). Hafez and colleagues (2005) developed an aerosol transfer technique in a porcine model using urothelial and bladder smooth muscle cell suspensions in fibrin glue. Autologous urothelial cells with or without smooth muscle cells, isolated at hemicystectomy, were sprayed onto demucosalised colon and then incorporated into the remaining bladder. After 6 weeks, this led to the development of a stratified, multilayered uroplakin-positive urothelium atop of a bladder or colonic smooth muscle submucosa, respectively, and no inflammation was described. Although the procedure did not involve propagation of urothelial cells in culture, it feasibly could do, the point of interest being whether in vitro-generated cells would remain capable of developing into a morphologically differentiated urothelial tissue after transplanting back in vivo.

A cell-engineering adaptation of enterocystoplasty has been described in a pig model wherein in vitro-propagated autologous urothelial cell sheets were implanted onto a vascularised, de-epithelialised host smooth muscle segment used to augment the bladder (Fraser et al., 2004). The urothelium was transplanted from cell culture to the surgical site using a Vicryl™ mesh carrier. The advantage of this ‘composite cystoplasty’ strategy over a full tissue-engineered approach is that the in vitro component of the procedure is confined to propagation of a single, highly regenerative cell type, the urothelium, which is combined with a preformed, innervated and vascularised smooth muscle host tissue.

In the most recent surgical series (Turner et al., 2011), the technique of extra-luminal dissection described by Hafez and colleagues (Hafez et al., 2003) was adapted to produce the de-epithelialised segment of bowel, and this was combined with an in vitro-generated functionally differentiated autologous urothelium (Turner et al., 2008) at the time of the composite cystoplasty reconstruction (Plate IX, between pages 354 and 355). Seven pigs underwent successful bladder augmentation using this technique and when sacrificed at 3 months, the bladder augments were found to be viable with no evidence of fibrosis or contraction. When examined histologically, all the augmented segments were completely covered with urothelium. Importantly, there was no evidence of colonic mucosal or crypt regrowth and unlike the initial study (Fraser et al., 2004), only minimal inflammatory changes were observed (Turner et al., 2011). This approach appears promising and is at the point of translation to clinic.

14.3.2 Free tissue grafts

Shortly after the first colocystoplasty was described in 1912, attempts were made to incorporate free biological tissue into the bladder. First, Neuhof (1917) used a free fascial patch in dogs and since then split skin grafts, placenta, peritoneum and dural membrane have all been used as patches (Draper et al., 1952; Kelami et al., 1970; Hutschenreiter et al., 1978; Fishman et al., 1987). There have been mixed results reported, with complications often arising as a result of normal functioning of the donor tissue (such as hair growth on skin grafts), alongside more general problems such as graft contraction and stone formation.

Nevertheless, the appeal of using free biological tissue persisted. Stenzl et al. (2000) performed detrusor myectomy using free latissimus dorsi (LD) grafts in four dogs. This approach was based on Carpentier’s LD cardiac wrap for patients with severe cardiomyopathy, which was the first recorded case of substituting non-skeletal muscle with skeletal muscle (Carpentier and Chachques, 1985). The procedure has been transferred to the clinical setting and initial clinical experiences were reported in 24 patients with bladder acontractility who required clean intermittent catheterisation (Gakis et al., 2011). Although the first clinical results appear promising, this procedure is still considered experimental.

14.3.3 Acellular matrices

The decellularisation of an allogeneic or xenogeneic tissue can potentially provide a bio- and tissue-compatible polymeric scaffold or matrix for recellularisation by the recipient’s own cells. Decellularised matrices retain the tissue-specific architecture with potential for tissue-specific cell–matrix interaction and differentiation cues. However, they also carry the potential risk of contamination by xenogeneic factors and for immunological reaction in the case of incomplete decellularisation. Another potential problem is the inherent biological variability of the source.

The two most common preparations described for potential use in bladder reconstruction are porcine SIS (Zhang et al., 2000; Kropp et al., 2004) and bladder acellular matrix graft (BAMG) (Dahms et al., 1998). In their natural tissue states, these matrices are heavily populated with cells and hence must undergo extensive decellularisation to remove all potentially immunogenic material. Non-cross-linked tissue matrices have been described as slow release vehicles of naturally occurring growth factors because, once implanted, they slowly degrade, acting as a scaffold for new ECM proteins produced by the in-growing cells (Kim et al., 2000; Badylak, 2002).

SIS

SIS has been used as a reconstructive tool in musculoskeletal, vascular and urological specialties with promising results. Its preparation entails the removal of the major cellular components of the bowel wall to leave the collagen- and elastin-rich submucosal layer. When incorporated into a bladder reconstruction, SIS degrades rapidly and completely and the breakdown products enter the circulation and can be detected in the urine (Badylak et al., 1998; Record et al., 2001). In its place, cellular encroachment and infiltration occurs rapidly, with the resultant tissue resembling that of the surrounding native organ.

One in vivo study reported that SIS has good biocompatibility when implanted in rats, as there was less inflammatory response compared to synthetic (PLGA) or combined (SIS-PLGA) scaffold materials (Kim et al., 2007). Early biocompatibility studies of macerated SIS periureteric injection and bladder patch grafts in pigs demonstrated the potential for smooth muscle and vascular in-growth (Knapp et al., 1994). Analysis of SIS patches 11 months after incorporation into rat bladders not only showed replacement by normal bladder tissue, but also vascularisation and re-innervation (Vaught et al., 1996). Furthermore, although of a lower magnitude, appropriate contractile and relaxatory responses were elicited on chemical stimulation of the patch, suggesting expression of neurotransmitter receptors. Similar results were obtained using SIS patches implanted in dogs, which confirmed that the regenerated grafts had similar viscoelastic properties to native bladder, despite having a reduced muscle: collagen ratio (Kropp et al., 1996a). This last fact may explain the decrease in magnitude of contraction observed. In addition, extensive neovascularisation had occurred in the submucosa and it was suggested that afferent nerves had re-innervated the segment (Kropp et al., 1996a, 1996b).

A limiting factor for functional outcome is the size of the implanted graft. In a canine subtotal cystectomy model (90% partial cystectomy), Zhang and his colleagues (2006) found graft shrinkage and severe inflammation, adhesion and stone formation when the bladders were augmented using seeded or unseeded SIS. Because of the inferior outcome compared to a 40% partial cystectomy model, the authors questioned SIS as an adequate graft material for (sub-) complete tissue-engineered bladder substitution.

SIS appears to offer some potential as a candidate for future clinical studies of bladder reconstruction. However, there is evidence that not all SIS is created equal, with the regenerative potential and incidence of complications being dependent upon the age of the donor pig and the region of the bowel from which the SIS matrices are derived (Kropp et al., 2004; Ashley et al., 2010). Furthermore, some reports have cast doubt on the biological safety of commercially-available SIS which, in vitro, has demonstrated cytotoxic effects on urothelial cells and has been found to retain porcine nuclear residues (Feil et al., 2006).

BAMG

The isolation of the bladder submucosa by dissection to leave a cell-depleted tissue, similar to SIS, has been described (Chen et al., 1999). More commonly, split- (urothelium-free) or full-thickness bladder is decellularised (Sutherland et al., 1996; Probst et al., 1997, 2000; Dahms et al., 1998; Piechota et al., 1998; Merguerian et al., 2000; Reddy et al., 2000; Brown et al., 2002; Bolland et al., 2007; Marcal et al., 2012). Implanting BAMG in the bladders of rats, dogs and pigs has shown regeneration of urothelial and muscle layers, with innervation and vascularisation of the graft (Sutherland et al., 1996; Probst et al., 1997, 2000; Piechota et al., 1998; Reddy et al., 2000). Contraction and relaxation has been described in BAMG-reconstructed bladders, but as with SIS, at reduced magnitude relative to normal bladder. A key functional difference between BAMG and SIS relates to the compliance of the material prior to implantation, with the SIS material 30 times less compliant than either native bladder or regenerated SIS (Kropp et al., 1996b). Conversely, non-regenerated split-thickness and full-thickness BAMG exhibited similar biomechanical properties to native bladder from the outset (Dahms et al., 1998). Whether this constitutes a critical advantage or not is perhaps moot when the ultimate aim is to produce a regenerated patch that is functionally equivalent to native bladder tissue.

Potential problems associated with the approach of using decellularised matrices include lithogenesis, graft shrinkage and incomplete/disorganised smooth muscle infiltration. Graft shrinkage due to fibroproliferative change has been shown to increase with time, with up to a 48% reduction in graft size (Brown et al., 2002). It should be considered that although regenerated smooth muscle within the grafts is often disorganised (i.e. does not form bundles), in surgical models, the extent and speed of cell incorporation is dependent upon the size of the grafts. In rat models, graft size is small (~ 0.5 cm2) whereas it is much greater in large animals (e.g. incorporation of 4 × 4 cm2 acellular dermal biomatrix patches into pig bladder; Akbal et al., 2006) and hence, it is unsurprising that smooth muscle bundles have been reported to be scanty at the centres of larger patches (Piechota et al., 1998; Brown et al., 2002). In practical terms, the surface area of bladder augmentation in man is an order of magnitude greater than many described experimentally in vivo and this represents a severe limitation to the translation of much reconstructive bladder research, particularly where rodent models have been used. Lithogenesis too is a particular problem in rat models, with up to 75% and 80% of animals found to have bladder stones in SIS and BAMG reconstructions, respectively (Vaught et al., 1996; Piechota et al., 1998). The problem is not confined to rodents: one group treated pigs with alendronate, an osteoclast inhibitor, to reduce urinary calcium concentrations following the discovery of microcalcification in the suburothelial zone of BAMG (Reddy et al., 2000), but such treatment does not allow for accurate determination of the risks involved.

Decellularised biomaterials may retain biological activity and this may encourage the in-growth of tissue. Furthermore, given that the composition and structure of the ECM is exclusive to individual tissues, there may be advantages in using orthotopic-derived matrices. For example, BAMG may be predicted to contain more appropriate growth factors for bladder tissue engineering than SIS (Badylak, 2004; Bolland et al., 2007; Marcal et al., 2012). Indeed, BAMG has been shown to be capable of sustained release of exogenous basic fibroblast growth factor and was demonstrated in a dose-dependent manner to significantly reduce graft shrinkage in a rat model of bladder augmentation (Kanematsu et al., 2003). Potentially, infiltration and organisation of smooth muscle bundles in both SIS and BAM grafts may be enhanced by the incorporation of growth factors and other bioactive substances (reviewed in Chen et al., 2010).

Natural matrices that undergo chemical or non-chemical cross-linking and terminal sterilisation to enhance the physical attributes and stability of the material are invariably rendered inert and may engender cytotoxic responses, thus ultimately inhibiting cellular incorporation (Badylak, 2002; Kimuli et al., 2004; Feil et al., 2006). Although some processed biomaterials have shown comparable results to SIS and BAMG, further development is necessary to realise the full potential of processed biomatrices (Nuininga et al., 2004). One such cross-linked material is Pelvicol™ (Permacol™ in the UK). This decellularised porcine dermis is used clinically in genitourinary reconstruction, for example as a corporal patch in Peyronie’s disease, a pubovaginal sling (Santucci and Barber, 2005), and recently for hypospadias repair (Springer and Subramaniam, 2012). However, early in vitro and in vivo assessments of this material for bladder reconstruction have been less promising. In the laboratory, Kimuli and colleagues (2004) reported poor smooth muscle cell infiltration of the material, possibly as a result of chemical cross-linking. Furthermore, an experimental study of bladder augmentation in rabbits using Pelvicol™ concluded that it was an unsuitable material for the procedure (Ayyildiz et al., 2006).

Akbal and colleagues (2006) used a 4 × 4 cm2 AlloDerm® acellular dermis graft to augment porcine bladders. The results were disappointing when the material was used in poorly compliant bladders in an experimental model of bladder outlet obstruction, whereas good results were achieved in healthy control animals. The authors concluded that the procedure was not recommended in low compliant bladders. The results from another in vivo study has supported the importance of post-operative mechanical distension of the neobladder (‘mechanical loading’) to facilitate the development and maintenance of adequate capacity and compliance (Boruch et al., 2010).

The limited success from incorporating passive natural tissue matrices into the bladder has led support to studies where biomaterials are pre-seeded with urothelial and smooth muscle cells ex vivo with the aim of enhancing tissue integration following implantation. Of underlying relevance to this approach is the pioneering work of Baskin and colleagues, who first showed that bladder smooth muscle development from the fetal mesenchyme was dependent upon paracrine interactions with the urothelium (Baskin et al., 1996; DiSandro et al., 1998). The potential for reciprocal interactions between urothelial and smooth muscle compartments during bladder tissue engineering has been investigated both in vitro (Fujiyama et al., 1995; Zhang et al., 2000; Ram-Liebig et al., 2004, 2006; Brown et al., 2005) and in vivo (Yoo and Meng, 1998; Master et al., 2003; Zhang et al., 2004). There is some controversy about the precise mechanisms underlying these interactions that is outside the scope of the present review. However, it is clear from recent research (Shin et al., 2011) that heterotypic cell–cell interactions are likely to play a critical role in the development of functional tissue-engineered bladders.

14.3.4 Natural ECM

The ECM has been used extensively as a xenogeneic and allogeneic biomaterial for cells of many types, reflecting its natural evolution as a tissue scaffold. Collagen, the most abundant protein within the ECM and the major structural protein in the body, is largely responsible for the strength and conformability of natural materials. Collagen has been shown to encourage cell growth, have minimal immunogenicity and can be readily purified and moulded into the desired form, making it an ideal tool for tissue-engineering applications (Elbahnasy et al., 1998; Hattori et al., 2006). Purified collagen, however, when used for reconstruction in the urinary tract, has been shown to lose its tensile strength and to be susceptible to tearing during suturing (Elbahnasy et al., 1998).

To overcome these problems, collagen has been reinforced with synthetic materials (see below), and with natural tissues, including a pedicled omental flap (Hattori et al., 2006). The latter investigators employed a porcine in vivo model to demonstrate that collagen sponge became vascularised when combined with omentum for 7 days in vivo and that only when pre-conditioned in this way was the collagen sponge able to support passive engineering of the bladder. This approach has important implications for other natural or synthetic biomaterials, as it provides a strategy for in vivo-preintegration of a scaffold for subsequent use in passive tissue engineering.

14.3.5 Synthetic grafts

The obvious advantages of synthetic materials can be seen in the usage of standardised raw materials and processing reproducibility, resulting in lower production costs compared to biological matrices.

The incorporation of synthetic materials alone into the bladder has largely been met with failure, primarily as a result of biological and mechanical incompatibility. Plastics, polyvinyl sponge, polytetrafluoroethylene (Teflon™) and Japanese paper have all been used to reconstruct the bladder with variable results, but none has been pursued to the present day and the use of such materials is considered obsolete (Bohne and Urwiller, 1957; Kudish, 1957; Kelami et al., 1970; Fujita, 1978). Perhaps the most promising report was the experimental bladder reconstruction in rabbits with a 6.25 cm2 poly(epsilon-benzyloxycarbonyl-L-lysine) membrane (Koiso et al., 1983). By 6 months it was reported that the resorbable membrane was completely replaced with normal urothelium and smooth muscle and there were no recorded complications. Despite such a positive study, no follow-on or clinical studies have ensued.

Bladder wall constructs comprising scaffolds seeded with urothelial and smooth muscle cells have been the most extensively researched strategy for bladder reconstruction, with a consensus that seeded constructs are superior over non-seeded scaffolds in terms of limiting graft shrinkage and loss of function (Yoo and Meng, 1998; Oberpenning et al., 1999; Atala et al., 2006; Zhang et al., 2006; Jayo et al., 2008; Tanaka et al., 2010). In a subtotal cystectomy canine model (about 80% removal of the bladder) with a follow-up of 2 years, a PLGA-based biodegradable synthetic polymer matrix seeded with autologous urothelial and smooth muscle cells was reported to result in tissue formation similar to the native bladder, including a three-layered detrusor muscle. Urodynamic studies revealed similar viscoelastic characteristics compared to a control group in which the native bladder was re-implanted and the dogs were able to void by increasing their abdominal tone. Moreover, the constructs were reported to grow during skeletal maturation of the young animals (Jayo et al., 2008).

Atala and colleagues were the first to demonstrate the feasibility of seeding cells onto a purely synthetic matrix for implantation in vivo (Oberpenning et al., 1999). PLGA is a well-characterised biomaterial with predictable biodegradability properties and is widely used as Vicryl™ sutures and meshes within the field of surgery. It is non-toxic and biocompatible with both normal human urothelial and bladder smooth muscle cells (Pariente et al., 2001, 2002; Scriven et al., 2001). These qualities make Vicryl™ an attractive candidate for combination with natural materials to form implantable constructs for bladder reconstruction. Oberpenning et al. (1999) used a polyglycolic acid (PGA) mesh, moulded into the shape of a bladder and coated with PLGA, and seeded the outer and inner surfaces of the biomaterial with autologous smooth muscle and urothelial cells, respectively. The constructs were then implanted in vivo onto a bladder base remaining after trigone-sparing cystectomy in dogs. Once coated with fibrin glue, the construct was wrapped with omentum and the animals were monitored for up to 11 months. There were no reported complications and mean capacity and compliance of the neobladders were similar to measurements pre-operatively. At 3 months, the polymer had degraded, leaving a vascularised, innervated tissue composed of organised smooth muscle bundles and a stratified urothelium, which was positive with antibodies against AUM. It is unlikely that the same degree of regeneration would have occurred without an omental wrap, such is its ability to induce neovascularisation.

Methods to improve cell attachment and proliferation on synthetic materials have also been explored. One solution is to coat the synthetic material with a biological substance or to use a surface modification procedure prior to seeding to encourage attachment. For example, in vitro, smooth muscle cells have been shown to attach and proliferate on a biodegradable polyesterurethane foam pre-treated with fetal bovine serum (Danielsson et al., 2006) and on plasma-coated, electrospun polystyrene (Baker et al., 2006). Alternatively, the material can be combined with one of the aforementioned natural materials to act as a biodegradable scaffold, giving strength and conformability to the structure. It is perhaps surprising, given the results described by Oberpenning et al. that when combined with PLGA, collagen hybrid matrices have shown mixed results in vitro. In one study, smooth muscle cells were able to proliferate and retain expression of differentiation markers on a gel-based construct, but not on a sponge, the opposite being the case for urothelial cells, which stratified on a sponge but not a gel, although unequivocal markers of urothelial differentiation were not shown (Nakanishi et al., 2003).

As biomaterial properties can have differential effects upon proliferation, migration and differentiation of different cell types, this must be taken into consideration when developing the ideal synthetic material. For instance, smooth muscle cells adopted a more natural organisation when grown on electrospun polystyrene scaffolds where fibres were aligned rather than showing a random distribution (Baker et al., 2006). Similarly, urothelial and smooth muscle cells showed improved growth properties on materials where the elastic modulus most closely matched that of the bladder (Rohman et al., 2007).

Meanwhile, Atala and colleagues (2006) have made the transition from canine model to clinical trials. Collagen-only and collagen–PGA hybrid scaffolds were seeded with autologous smooth muscle and urothelial cells and implanted into nine patients with severely neuropathic bladders. Three had a collagen-only implant, one had collagen-only implant with an omental wrap and three patients had the collagen–PGA hybrid scaffold with an omental wrap. Two patients were lost to follow-up and one patient with a collagen-only implant underwent conventional augmentation because of progressively rising intravesical pressures. For the remaining patients, although followed up annually for up to 5 years, not all results were available at each time point and only four had investigations in the fifth year. There were minimal or modest increases in capacity and compliance of the bladders, with the best outcome in patients receiving cell-seeded collagen-coated PGA scaffolds that were wrapped in omentum as a vascular bed. The new bladder tissue was described as having a normal structure, with smooth muscle and stratified urothelium; however, the differentiation status of the urothelium was not reported.

More recent developments in the field include the incorporation of polymers, such as PLGA nanoparticles into decellularised matrices, e.g. SIS or BAMG grafts (Mondalek et al., 2008, 2010; Geng et al., 2011; Roth et al., 2011). With the aim of improving the consistency and biocompatility of the biomaterial, the incorporation of hyaluronic acid PLGA nanoparticles into SIS was reported to enhance angiogenesis and smooth muscle cell regeneration in a canine partial cystectomy model (Mondalek et al., 2010; Roth et al., 2011). Another group has incorporated vascular endothelial growth factor (VEGF)-loaded PLGA nanoparticles into BAMG and showed good biocompatibility in vitro and in vivo (Geng et al., 2011), although no results have yet been reported for its use in bladder reconstruction.

14.4 Cell conditioning in an external bioreactor

14.4.1 Static conditioning

It is widely accepted that cells lose functional and differentiated characteristics when isolated from their host tissue and propagated in cell culture. For all active tissue-engineering strategies involving cell-seeded approaches, the critical question is whether this loss is reversible and thus whether (and how) cultured cells can be induced to differentiate and form functional tissue equivalents.

For example, normal human urothelial (NHU) cells grown in monoculture adopt a proliferative, highly regenerative, but non-differentiated state (Southgate et al., 1994). It has been shown possible to switch these in vitro-propagated cells to a stratified, differentiated and functional barrier urothelium following manipulation of the growth medium (Cross et al., 2005). The resultant urothelium had functional barrier properties, as assessed by a high transepithelial electrical resistance (TER) of > 3000 Ω cm2 and low diffusive permeability to urea, water and dextran. This technique was adapted and applied to porcine urothelial cells in vitro (Turner et al., 2008) prior to application in a surgical model of composite cystoplasty (described in Section 14.3.1) (Turner et al., 2011). Progress has also been made in the identification of the molecular pathways involved in inducing urothelial differentiation, with the nuclear receptor, peroxisome proliferator activated receptor gamma (PPARγ), identified as a key regulator of urothelial differentiation (Varley et al., 2004, 2006, 2009). Notably, although a urothelial stem cell is purported to reside basally in situ (Gaisa et al., 2011), it has been shown that both basal and suprabasal-derived urothelial cells demonstrate equal proliferation and differentiation potential in vitro, thus negating the need to isolate specific progenitor populations for tissueengineering purposes (Wezel et al., 2014).

14.4.2 Biomechanical conditioning

Further improvement of the biomimetic properties of in vitro-generated bladder tissue may be achieved by simulating the physical environment of the bladder (Korossis et al., 2006). So-called ‘functional tissue engineering’ employs an external bioreactor to condition cells seeded onto a scaffold by controlling nutrition and providing appropriate mechanostimulation. Mechanosensitivity is a requirement in all cells and allows them to respond appropriately to physiological signals, as well as to insults such as physical stress and osmotic pressure gradients (Hamill and Martinac, 2001); for example, in vitro studies of myofibroblasts showed that proliferation and biosynthetic activity changed with the degree of mechanical stress (Grinnell, 1994). The bladder fills with urine passively and undergoes several fill–void cycles daily, and despite the large and often rapid changes in volume, the urinary barrier remains intact. The urothelium manages this by being exquisitely sensitive to stretch, with the superficial cells mobilising the AUM-containing fusiform vesicles to open onto the luminal membrane in response to filling, thus maintaining an appropriate surface area (Truschel et al., 2002). Replication of the fill–void cycle would seem to be an appropriate measure when generating a biomimetic tissue in vivo and may have significant consequences for tissue functionalisation.

14.5 Future trends

Adult autologous cells have several advantages over allogeneic stem cells in tissue-engineering approaches as the perfect genetic match excludes immunological conflicts and the need for immunosuppression. However, where the damage to the bladder reflects an underlying irreversible disease process, a biopsy may not provide sufficient autologous healthy cells to propagate clinically useful quantities for tissue engineering (Subramaniam et al., 2011). Similarly, in patients suffering from urothelial cancer, the use of autologous cells from the urinary tract may not be safe. In such patients, alternative cell sources may be required for tissue-engineered approaches to urinary tract reconstruction.

One possibility is to use an alternative epithelium, such as buccal mucosa, which has a history of transplant use in the urinary tract (Bhargava and Chapple, 2004) and which can be cultured successfully in vitro (Southgate et al., 1987; Lauer et al., 2001; Bhargava et al., 2008). Alternatively, the use of differentiation-directed stem cells may be an option to form stratified epithelial tissue or smooth muscle cells for integration with scaffold matrices. To date, only very early experimental data exist using multipotent stem cells (including rodent hair-follicle derived (Drewa et al., 2009), adipose-derived (Rodriguez et al., 2006; Jack et al., 2009), mesenchymal stem cells (Shukla et al., 2008; Tian et al., 2010) and pluripotent embryonic stem cells (Oottamasathien et al., 2007; Kinebuchi et al., 2008; Thomas et al., 2008)). Differentiation has been induced using defined cues (Oottamasathien et al., 2007) or by placing them into a bladder-specific microenvironment (e.g. fetal bladder mesenchyme; Thomas et al., 2008). These approaches are a long way from clinical use, with insufficient objective characterisation of differentiated function or safety.

A major disadvantage of allogeneic versus autologous stem cells is the need for a genetically matched donor and the risk of contamination. In 2006, Takahashi reported the reversion of a somatic fibroblast to a pluripotent stem cell phenotype by overexpression of four transcription factors Oct4, Sox2, Myc and Klf4, thus generating patient-specific so-called ‘induced pluripotent stem cells’ (Takahashi and Yamanaka, 2006). The challenge remains to unlock the potential of pluripotent cells to form urothelial cells in a controlled manner, which has not yet been achieved, although early work has begun to identify the key transcriptional regulators required to define urothelial lineage commitment (Oottamasathien et al., 2007; Thomas et al., 2008; Varley et al., 2009).

14.6 Conclusions

Reconstruction of the urinary bladder is carried out when conservative and medical therapies have failed to alleviate the debilitating symptoms of a small, non-compliant or diseased bladder. Enterocystoplasty has provided relief for many patients, but is recognised to carry the price of serious complications resulting from the long-term interactions between the bowel epithelium and urine. Despite some high profile reports, the current reality is that active tissue engineering of full bladder wall equivalents is overambitious given the complexity of the tissue and lack of progress in bladder smooth muscle cell biology. By contrast, advances in urothelial cell biology support the simpler strategy of composite cystoplasty, where the bladder is reconstructed with host bowel segments lined by an in vitro-engineered autologous urothelium.

Clinical practice would favour a cell-free passive bladder engineering approach incorporating biomaterials alone. This relies on identifying suitable scaffold materials that both harness a tissue integration response and match the physical requirements of the bladder for compliance. At present these properties are best realised by natural decellularised matrices, particularly those from an orthotopic but xenogeneic source, and this highlights a need for the development of new, fit-for-purpose biocompatible synthetic polymeric materials.

Taking a passive engineering approach forward requires consideration of the clinical relevance of the experimental model used for proof of principle testing, particularly in terms of the size of patch incorporated and the lack of underlying pathology. Remembering lessons already learnt, for instance exploiting the omentum as a vascularisation bed, are likely to be part of the key to future success.

14.7 References

1. Acharya P, Beckel J, Ruiz WG, et al. Distribution of the tight junction proteins ZO-1, occludin, and claudin-4, -8, and -12 in bladder epithelium. Am J Physiol Renal Physiol. 2004;287:F305–F318.

2. Adelow C, Segura T, Hubbell JA, Frey P. The effect of enzymatically degradable poly(ethylene glycol) hydrogels on smooth muscle cell phenotype. Biomaterials. 2008;29:314–326.

3. Aitken KJ, Bagli DJ. The bladder extracellular matrix Part II: regenerative applications. Nat Rev Urol. 2009;6:612–621.

4. Akbal C, Lee SD, Packer SC, Davis MM, Rink RC, Kaefer M. Bladder augmentation with acellular dermal biomatrix in a diseased animal model. J Urol. 2006;176:1706–1711.

5. Aktug T, Ozdemir T, Agartan C, Ozer E, Olguner M, Akgur FM. Experimentally prefabricated bladder. J Urol. 2001;165:2055–2058.

6. Arimura H, Ouchi T, Kishida A, Ohya Y. Preparation of a hyaluronic acid hydrogel through polyion complex formation using cationic polylactide-based microspheres as a biodegradable crosslinking agent. J Biomater Sci Polym Ed. 2005;16:1347–1358.

7. Ashley RA, Roth CC, Palmer BW, et al. Regional variations in small intestinal submucosa evoke differences in inflammation with subsequent impact on tissue regeneration in the rat bladder augmentation model. BJU Int. 2010;105:1462–1468.

8. Atala A, Bauer SB, Soker S, Yoo JJ, Retik AB. Tissue-engineered autologous bladders for patients needing cystoplasty. Lancet. 2006;367:1241–1246.

9. Ayyildiz A, Nuhoglu B, Huri E, Ozer E, Gurdal M, Germiyanoglu C. Using porcine acellular collagen matrix (Pelvicol) in bladder augmentation: experimental study. Int Braz J Urol. 2006;32:88–92 discussion 92–3.

10. Badylak SF. The extracellular matrix as a scaffold for tissue reconstruction. Semin Cell Dev Biol. 2002;13:377–383.

11. Badylak SF. Xenogeneic extracellular matrix as a scaffold for tissue reconstruction. Transpl Immunol. 2004;12:367–377.

12. Badylak SF, Lantz GC, Coffey A, Geddes LA. Small intestinal submucosa as a large diameter vascular graft in the dog. J Surg Res. 1989;47:74–80.

13. Badylak SF, Kropp B, McPherson T, Liang H, Snyder PW. Small intestional submucosa: a rapidly resorbed bioscaffold for augmentation cystoplasty in a dog model. Tissue Eng. 1998;4:379–387.

14. Baker SC, Atkin N, Gunning PA, et al. Characterisation of electrospun polystyrene scaffolds for three-dimensional in vitro biological studies. Biomaterials. 2006;27:3136–3146.

15. 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.

16. Baker SC, Rohman G, Hinley J, Stahlschmidt J, Cameron NR, Southgate J. Cellular integration and vascularisation promoted by a resorbable, particulate-leached, cross-linked poly(epsilon-caprolactone) scaffold. Macromol Biosci. 2011;11:618–627.

17. Baskin LS, Hayward SW, Young P, Cunha GR. Role of mesenchymalepithelial interactions in normal bladder development. J Urol. 1996;156:1820–1827.

18. Beier-Holgersen R, Kirkeby LT, Nordling J. ‘Clam’ ileocystoplasty. Scand J Urol Nephrol. 1994;28:55–58.

19. Bhargava S, Chapple CR. Buccal mucosal urethroplasty: is it the new gold standard? BJU Int. 2004;93:1191–1193.

20. Bhargava S, Patterson JM, Inman RD, MacNeil S, Chapple CR. Tissue-engineered buccal mucosa urethroplasty – clinical outcomes. Eur Urol. 2008;53:1263–1269.

21. Biers SM, Venn SN, Greenwell TJ. The past, present and future of augmentation cystoplasty. BJU Int. 2012;109:1280–1293.

22. Birder LA, Ruggieri M, Takeda M, et al. How does the urothelium affect bladder function in health and disease? ICI-RS 2011. Neurourol Urodyn. 2012;31:293–299.

23. Bohne AW, Urwiller KL. Experience with urinary bladder regeneration. J Urol. 1957;77:725–732.

24. Bolland F, Korossis S, Wilshaw SP, et al. Development and characterisation of a full-thickness acellular porcine bladder matrix for tissue engineering. Biomaterials. 2007;28:1061–1070.

25. Boruch AV, Nieponice A, Qureshi IR, Gilbert TW, Badylak SF. Constructive remodeling of biologic scaffolds is dependent on early exposure to physiologic bladder filling in a canine partial cystectomy model. J Surg Res. 2010;161:217–225.

26. Brown AL, Farhat W, Merguerian PA, Wilson GJ, Khoury AE, Woodhouse KA. 22 week assessment of bladder acellular matrix as a bladder augmentation material in a porcine model. Biomaterials. 2002;23:2179–2190.

27. Brown AL, Brook-Allred TT, Waddell JE, et al. Bladder acellular matrix as a substrate for studying in vitro bladder smooth muscle–urothelial cell interactions. Biomaterials. 2005;26:529–543.

28. Cain MP, Rink RC. Augmentation for neuropathic bladder dysfunction – a thing of the past? J Urol. 2010;183:2124–2125.

29. Carpentier A, Chachques JC. Myocardial substitution with a stimulated skeletal muscle: first successful clinical case. Lancet. 1985;1:1267.

30. Chen F, Yoo JJ, Atala A. Acellular collagen matrix as a possible ‘off the shelf’ biomaterial for urethral repair. Urology. 1999;54:407–410.

31. Chen FM, Zhang M, Wu ZF. Toward delivery of multiple growth factors in tissue engineering. Biomaterials. 2010;31:6279–6308.

32. Couvelaire R. The ‘little bladder’ of genito-urinary tuberculosis; classification, site and variants of bladder-intestine transplants. J Urol Medicale Chir. 1950;56:381–434.

33. Cross WR, Eardley I, Leese HJ, Southgate J. A biomimetic tissue from cultured normal human urothelial cells: analysis of physiological function. Am J Physiol Renal Physiol. 2005;289:F459–F468.

34. da Costa FD, Santos LR, Collatusso C, et al. Thirteen years’ experience with the Ross Operation. J Heart Valve Dis. 2009;18:84–94.

35. Dahms SE, Piechota HJ, Dahiya R, Lue TF, Tanagho EA. Composition and biomechanical properties of the bladder acellular matrix graft: comparative analysis in rat, pig and human. Br J Urol. 1998;82:411–419.

36. Danielsson C, Ruault S, Simonet M, Neuenschwander P, Frey P. Polyesterurethane foam scaffold for smooth muscle cell tissue engineering. Biomaterials. 2006;27:1410–1415.

37. Davis NF, McGuire BB, Callanan A, Flood HD, McGloughlin TM. Xenogenic extracellular matrices as potential biomaterials for interposition grafting in urological surgery. J Urol. 2010;184:2246–2253.

38. DiSandro MJ, Li Y, Baskin LS, Hayward S, Cunha G. Mesenchymalepithelial interactions in bladder smooth muscle development: epithelial specificity. J Urol. 1998;160:1040–1046 discussion 1079.

39. Draper JW, Stark RB, Lau MW. Replacement of mucous membrane of urinary bladder with thick-split grafts of skin: experimental observations. Plast Reconstr Surg. 1952;10:252–259.

40. Drewa T, Adamowicz J, Lysik J, Polaczek J, Pielichowski J. Chitosan scaffold enhances nerve regeneration within the in vitro reconstructed bladder wall: an animal study. Urol Int. 2008;81:330–334.

41. Drewa T, Joachimiak R, Kaznica A, Sarafian V, Pokrywczynska M. Hair stem cells for bladder regeneration in rats: preliminary results. Transplant Proc. 2009;41:4345–4351.

42. Eberli D, Rodriguez S, Atala A, Yoo JJ. In vivo evaluation of acellular human dermis for abdominal wall repair. J Biomed Mater Res A. 2010;93:1527–1538.

43. Elbahnasy AM, Shalhav A, Hoenig DM, Figenshau R, Clayman RV. Bladder wall substitution with synthetic and non-intestinal organic materials. J Urol. 1998;159:628–637.

44. Engelhardt EM, Stegberg E, Brown RA, et al. Compressed collagen gel: a novel scaffold for human bladder cells. J Tissue Eng Regen Med. 2010;4:123–130.

45. Engler AJ, Sen S, Sweeney HL, Discher DE. Matrix elasticity directs stem cell lineage specification. Cell. 2006;126:677–689.

46. Feil G, Christ-Adler M, Maurer S, et al. Investigations of urothelial cells seeded on commercially available small intestine submucosa. Eur Urol. 2006;50:1330–1337.

47. Fishman IJ, Flores FN, Scott FB, Spjut HJ, Morrow B. Use of fresh placental membranes for bladder reconstruction. J Urol. 1987;138:1291–1294.

48. Fowler CJ, Auerbach S, Ginsberg D, et al. OnabotulinumtoxinA improves health-related quality of life in patients with urinary incontinence due to idiopathic overactive bladder: a 36-week, double-blind, placebo-controlled, randomized, dose-ranging trial. Eur Urol. 2012;62:148–157.

49. Fraser M, Thomas DF, Pitt E, Harnden P, Trejdosiewicz LK, Southgate J. A surgical model of composite cystoplasty with cultured urothelial cells: a controlled study of gross outcome and urothelial phenotype. BJU Int. 2004;93:609–616.

50. Fujita K. The use of resin-sprayed thin paper for urinary bladder regeneration. Invest Urol. 1978;15:355–357.

51. Fujiyama C, Masaki Z, Sugihara H. Reconstruction of the urinary bladder mucosa in three-dimensional collagen gel culture: fibroblast-extracellular matrix interactions on the differentiation of transitional epithelial cells. J Urol. 1995;153:2060–2067.

52. Gakis G, Ninkovic M, van Koeveringe GA, et al. Functional detrusor myoplasty for bladder acontractility: long-term results. J Urol. 2011;185:593–599.

53. Gaisa NT, Graham TA, McDonald SA, et al. The human urothelium consists of multiple clonal units, each maintained by a stem cell. J Pathol. 2011;255:163–171.

54. Geng H, Song H, Qi J, Cui D. Sustained release of VEGF from PLGA nanoparticles embedded thermo-sensitive hydrogel in full-thickness porcine bladder acellular matrix. Nanoscale Res Lett. 2011;6:312.

55. Gilbert TW. Collagen fiber alignment and biaxial mechanical behavior of porcine urinary bladder derived extracellular matrix. Biomaterials. 2008;29:4775–4782.

56. Gilbert TW, Sellaro TL, Badylak SF. Decellularization of tissues and organs. Biomaterials. 2006;27:3675–3683.

57. Greenwell TJ, Venn SN, Mundy AR. Augmentation cystoplasty. BJU Int. 2001;88:511–525.

58. Grinnell F. Fibroblasts, myofibroblasts, and wound contraction. J Cell Biol. 1994;124:401–404.

59. Hafez AT, Bagli DJ, Bahoric A, et al. Aerosol transfer of bladder urothelial and smooth muscle cells onto demucosalized colonic segments: a pilot study. J Urol. 2003;169:2316–2319 discussion 2320.

60. Hafez AT, Afshar K, Bagli DJ, et al. Aerosol transfer of bladder urothelial and smooth muscle cells onto demucosalized colonic segments for porcine bladder augmentation in vivo: a 6-week experimental study. J Urol. 2005;174:1663–1667 discussion 1667–8.

61. Hamill OP, Martinac B. Molecular basis of mechanotransduction in living cells. Physiol Rev. 2001;81:685–740.

62. Hattori K, Joraku A, Miyagawa T, Kawai K, Oyasu R, Akaza H. Bladder reconstruction using a collagen patch prefabricated within the omentum. Int J Urol. 2006;13:529–537.

63. Hicks RM. The fine structure of the transitional epithelium of rat ureter. J Cell Biol. 1965;26:25–48.

64. Hicks RM. The mammalian urinary bladder: an accommodating organ. Biol Rev Camb Philos Soc. 1975;50:215–246.

65. Hu P, Deng FM, Liang FX, et al. Ablation of uroplakin III gene results in small urothelial plaques, urothelial leakage, and vesicoureteral reflux. J Cell Biol. 2000;151:961–972.

66. Hu P, Meyers S, Liang FX, et al. Role of membrane proteins in permeability barrier function: uroplakin ablation elevates urothelial permeability. Am J Physiol Renal Physiol. 2002;283:F1200–F1207.

67. Hutschenreiter G, Rumpelt HJ, Klippel KF, Hohenfellner R. The free peritoneal transplant as substitute for the urinary bladder wall. Invest Urol. 1978;15:375–379.

68. Jack GS, Zhang R, Lee M, Xu Y, Wu BM, Rodriguez LV. Urinary bladder smooth muscle engineered from adipose stem cells and a three dimensional synthetic composite. Biomaterials. 2009;30:3259–3270.

69. Jayo MJ, Jain D, Ludlow JW, et al. Long-term durability, tissue regeneration and neo-organ growth during skeletal maturation with a neo-bladder augmentation construct. Regen Med. 2008;3:671–682.

70. Kanematsu A, Yamamoto S, Noguchi T, Ozeki M, Tabata Y, Ogawa O. Bladder regeneration by bladder acellular matrix combined with sustained release of exogenous growth factor. J Urol. 2003;170:1633–1638.

71. Kaully T, Kaufman-Francis K, Lesman A, Levenberg S. Vascularization – the conduit to viable engineered tissues. Tissue Eng Part B Rev. 2009;15:159–169.

72. Kelami A, Dustmann HO, Ludtke-Handjery A, Carcamo V, Herlld G. Experimental investigations of bladder regeneration using teflon-felt as a bladder wall substitute. J Urol. 1970;104:693–698.

73. Kim BS, Baez CE, Atala A. Biomaterials for tissue engineering. World J Urol. 2000;18:2–9.

74. Kim MS, Ahn HH, Shin YN, Cho MH, Khang G, Lee HB. An in vivo study of the host tissue response to subcutaneous implantation of PLGA-and/or porcine small intestinal submucosa-based scaffolds. Biomaterials. 2007;28:5137–5143.

75. Kimuli M, Eardley I, Southgate J. In vitro assessment of decellularized porcine dermis as a matrix for urinary tract reconstruction. BJU Int. 2004;94:859–866.

76. Kinebuchi Y, Johkura K, Sasaki K, Imamura T, Mimura Y, Nishizawa O. Direct induction of layered tissues from mouse embryonic stem cells: potential for differentiation into urinary tract tissue. Cell Tissue Res. 2008;331:605–615.

77. Kiricuta I, Goldstein AM. The repair of extensive vesicovaginal fistulas with pedicled omentum: a review of 27 cases. J Urol. 1972;108:724–727.

78. Knapp PM, Lingeman JE, Siegel YI, Badylak SF, Demeter RJ. Biocompatibility of small-intestinal submucosa in urinary tract as augmentation cystoplasty graft and injectable suspension. J Endourol. 1994;8:125–130.

79. Koiso K, Komai T, Niijima T. Experimental urinary bladder reconstruction using a synthetic poly(alpha-amino acids) membrane. Artif Organs. 1983;7:232–237.

80. Korossis S, Bolland F, Ingham E, Fisher J, Kearney J, Southgate J. Review: tissue engineering of the urinary bladder: considering structure-function relationships and the role of mechanotransduction. Tissue Eng. 2006;12:635–644.

81. Kropp BP, Rippy MK, Badylak SF, et al. Regenerative urinary bladder augmentation using small intestinal submucosa: urodynamic and histopathologic assessment in long-term canine bladder augmentations. J Urol. 1996a;155:2098–2104.

82. Kropp BP, Sawyer BD, Shannon HE, et al. Characterization of small intestinal submucosa regenerated canine detrusor: assessment of reinnervation, in vitro compliance and contractility. J Urol. 1996b;156:599–607.

83. Kropp BP, Cheng EY, Lin HK, Zhang Y. Reliable and reproducible bladder regeneration using unseeded distal small intestinal submucosa. J Urol. 2004;172:1710–1713.

84. Kudish HG. The use of polyvinyl sponge for experimental cystoplasty. J Urol. 1957;78:232–235.

85. Lauer G, Schimming R, Frankenschmidt A. Intraoral wound closure with tissue-engineered mucosa: new perspectives for urethra reconstruction with buccal mucosa grafts. Plast Reconstr Surg. 2001;107:25–33.

86. Lavelle J, Meyers S, Ramage R, et al. Bladder permeability barrier: recovery from selective injury of surface epithelial cells. Am J Physiol Renal Physiol. 2002;283:F242–F253.

87. Lee CH, Cook JL, Mendelson A, Moioli EK, Yao H, Mao JJ. Regeneration of the articular surface of the rabbit synovial joint by cell homing: a proof of concept study. Lancet. 2010;376:440–448.

88. Li L, Xie T. Stem cell niche: structure and function. Annu Rev Cell Dev Biol. 2005;21:605–631.

89. Lima SV, Araujo LA, Vilar FO. Nonsecretory intestinocystoplasty: a 10-year experience. J Urol. 2004;171:2636–2639 discussion 2639–40.

90. Marcal H, Ahmed T, Badylak SF, Tottey S, Foster LJ. A comprehensive protein expression profile of extracellular matrix biomaterial derived from porcine urinary bladder. Regen Med. 2012;7:159–166.

91. Master VA, Wei G, Liu W, Baskin LS. Urothlelium facilitates the recruitment and trans-differentiation of fibroblasts into smooth muscle in acellular matrix. J Urol. 2003;170:1628–1632.

92. McGlohorn JB, Holder Jr WD, Grimes LW, Thomas CB, Burg KJ. Evaluation of smooth muscle cell response using two types of porous polylactide scaffolds with differing pore topography. Tissue Eng. 2004;10:505–514.

93. Merguerian PA, Reddy PP, Barrieras DJ, et al. Acellular bladder matrix allografts in the regeneration of functional bladders: evaluation of large-segment (> 24 cm) substitution in a porcine model. BJU Int. 2000;85:894–898.

94. Mikos AG, Sarakinos G, Lyman MD, Ingber DE, Vacanti JP, Langer R. Prevascularization of porous biodegradable polymers. Biotechnol Bioeng. 1993;42:716–723.

95. Mirsadraee S, Wilcox HE, Watterson KG, et al. Biocompatibility of acellular human pericardium. J Surg Res. 2007;143:407–414.

96. Mitrofanoff P. Trans-appendicular continent cystostomy in the management of the neurogenic bladder. Chir Pediatr. 1980;21:297–305.

97. Mondalek FG, Lawrence BJ, Kropp BP, et al. The incorporation of poly(lactic-co-glycolic) acid nanoparticles into porcine small intestinal submucosa biomaterials. Biomaterials. 2008;29:1159–1166.

98. Mondalek FG, Ashley RA, Roth CC, et al. Enhanced angiogenesis of modified porcine small intestinal submucosa with hyaluronic acid-poly(lactide-co-glycolide) nanoparticles: from fabrication to preclinical validation. J Biomed Mater Res A. 2010;94:712–719.

99. Mooney DJ, Baldwin DF, Suh NP, Vacanti JP, Langer R. Novel approach to fabricate porous sponges of poly(d, l-lactic-co-glycolic acid) without the use of organic solvents. Biomaterials. 1996;17:1417–1422.

100. Motley RC, Montgomery BT, Zollman PE, Holley KE, Kramer SA. Augmentation cystoplasty utilizing de-epithelialized sigmoid colon: a preliminary study. J Urol. 1990;143:1257–1260.

101. Nakanishi Y, Chen G, Komuro H, et al. Tissue-engineered urinary bladder wall using PLGA mesh–collagen hybrid scaffolds: a comparison study of collagen sponge and gel as a scaffold. J Pediatr Surg. 2003;38:1781–1784.

102. Neuhof H. Fascial transplantation into visceral defects: an experimental and clinical study. Surg, Gynec and Obst. 1917;25:383.

103. Nuininga JE, van Moerkerk H, Hanssen A, et al. A rabbit model to tissue engineer the bladder. Biomaterials. 2004;25:1657–1661.

104. 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.

105. Olsburgh J, Harnden P, Weeks R, et al. Uroplakin gene expression in normal human tissues and locally advanced bladder cancer. J Pathol. 2003;199:41–49.

106. Oottamasathien S, Wang Y, Williams K, et al. Directed differentiation of embryonic stem cells into bladder tissue. Dev Biol. 2007;304:556–566.

107. Pariente JL, Kim BS, Atala A. In vitro biocompatibility assessment of naturally derived and synthetic biomaterials using normal human urothelial cells. J Biomed Mater Res. 2001;55:33–39.

108. Pariente JL, Kim BS, Atala A. In vitro biocompatibility evaluation of naturally derived and synthetic biomaterials using normal human bladder smooth muscle cells. J Urol. 2002;167:1867–1871.

109. Park SN, Park JC, Kim HO, Song MJ, Suh H. Characterization of porous collagen/hyaluronic acid scaffold modified by 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide cross-linking. Biomaterials. 2002;23:1205–1212.

110. Peyton CC, Burmeister D, Petersen B, Andersson KE, Christ G. Characterization of the early proliferative response of the rodent bladder to subtotal cystectomy: a unique model of mammalian organ regeneration. PLoS One. 2012;7:e47414.

111. Piechota HJ, Dahms SE, Nunes LS, Dahiya R, Lue TF, Tanagho EA. In vitro functional properties of the rat bladder regenerated by the bladder acellular matrix graft. J Urol. 1998;159:1717–1724.

112. Probst M, Dahiya R, Carrier S, Tanagho EA. Reproduction of functional smooth muscle tissue and partial bladder replacement. Br J Urol. 1997;79:505–515.

113. Probst M, Piechota HJ, Dahiya R, Tanagho EA. Homologous bladder augmentation in dog with the bladder acellular matrix graft. BJU Int. 2000;85:362–371.

114. Ram-Liebig G, Meye A, Hakenberg OW, Haase M, Baretton G, Wirth MP. Induction of proliferation and differentiation of cultured urothelial cells on acellular biomaterials. BJU Int. 2004;94:922–927.

115. Ram-Liebig G, Ravens U, Balana B, Haase M, Baretton G, Wirth MP. New approaches in the modulation of bladder smooth muscle cells on viable detrusor constructs. World J Urol. 2006;24:429–437.

116. Record RD, Hillegonds D, Simmons C, et al. In vivo degradation of 14C-labeled small intestinal submucosa (SIS) when used for urinary bladder repair. Biomaterials. 2001;22:2653–2659.

117. Reddy PP, Barrieras DJ, Wilson G, et al. Regeneration of functional bladder substitutes using large segment acellular matrix allografts in a porcine model. J Urol. 2000;164:936–941.

118. Rizvi AZ, Wong MH. Epithelial stem cells and their niche: there’s no place like home. Stem Cells. 2005;23:150–165.

119. 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 USA. 2006;103:12167–12172.

120. Rohman G, Pettit JJ, Isaure F, Cameron NR, Southgate J. Influence of the physical properties of two-dimensional polyester substrates on the growth of normal human urothelial and urinary smooth muscle cells in vitro. Biomaterials. 2007;28:2264–2274.

121. Rohman G, Baker SC, Southgate J, Cameron NR. Heparin functionalisation of porous PLGA scaffolds for controlled, biologically relevant delivery of growth factors for soft tissue engineering. J Mater Chem. 2009;19:9265–9273.

122. Roth CC, Mondalek FG, Kibar Y, et al. Bladder regeneration in a canine model using hyaluronic acid-poly(lactic-co-glycolic-acid) nanoparticle modified porcine small intestinal submucosa. BJU Int. 2011;108:148–155.

123. Roth EA, Xu T, Das M, Gregory C, Hickman JJ, Boland T. Inkjet printing for high-throughput cell patterning. Biomaterials. 2004;25:3707–3715.

124. Rowlands AS, Lim SA, Martin D, Cooper-White JJ. Polyurethane/poly(lactic-co-glycolic) acid composite scaffolds fabricated by thermally induced phase separation. Biomaterials. 2007;28:2109–2121.

125. Rowley JA, Madlambayan G, Mooney DJ. Alginate hydrogels as synthetic extracellular matrix materials. Biomaterials. 1999;20:45–53.

126. Salle JL, Fraga JC, Lucib A, Lampertz M, Jobim G, Putten A. Seromuscular enterocystoplasty in dogs. J Urol. 1990;144:454–456 discussion 460.

127. Santucci RA, Barber TD. Resorbable extracellular matrix grafts in urologic reconstruction. Int Braz J Urol. 2005;31:192–203.

128. Schoeller T, Neumeister MW, Huemer GM, et al. Capsule induction technique in a rat model for bladder wall replacement: an overview. Biomaterials. 2004;25:1663–1673.

129. Scriven SD, Trejdosiewicz LK, Thomas DFM, Southgate J. Urothelial cell transplantation using biodegradable synthetic scaffolds. J Mat Sci Mat Med. 2001;12:991–996.

130. Shin K, Lee J, Guo N, et al. Hedgehog/Wnt feedback supports regenerative proliferation of epithelial stem cells in bladder. Nature. 2011;472:110–114.

131. Shukla D, Box GN, Edwards RA, Tyson DR. Bone marrow stem cells for urologic tissue engineering. World J Urol. 2008;26:341–349.

132. Southgate J, Williams HK, Trejdosiewicz LK, Hodges GM. Primary culture of human oral epithelial cells Growth requirements and expression of differentiated characteristics. Lab Invest. 1987;56:211–223.

133. Southgate J, Hutton KA, Thomas DF, Trejdosiewicz LK. Normal human urothelial cells in vitro: proliferation and induction of stratification. Lab Invest. 1994;71:583–594.

134. Springer A, Subramaniam R. Preliminary experience with the use of acellular collagen matrix in redo surgery for urethrocutaneous fistula. Urology. 2012;80:1156–1160.

135. Stein JP, Skinner DG. Surgical atlas: the orthotopic T-pouch ileal neobladder. BJU Int. 2006;98:469–482.

136. Stenzl A, Strasser H, Klima G, et al. Reconstruction of the lower urinary tract using autologous muscle transfer and cell seeding: current status and future perspectives. World J Urol. 2000;18:44–50.

137. Studer UE, Varol C, Danuser H. Orthotopic ileal neobladder. BJU Int. 2004;93:183–193.

138. Subramaniam R, Hinley J, Stahlschmidt J, Southgate J. Tissue engineering potential of urothelial cells from diseased bladders. J Urol. 2011;186:2014–2020.

139. Sutherland RS, Baskin LS, Hayward SW, Cunha GR. Regeneration of bladder urothelium, smooth muscle, blood vessels and nerves into an acellular tissue matrix. J Urol. 1996;156:571–577.

140. Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006;126:663–676.

141. Tanaka ST, Thangappan R, Eandi JA, Leung KN, Kurzrock EA. Bladder wall transplantation – long-term survival of cells: implications for bioengineering and clinical application. Tissue Eng Part A. 2010;16:2121–2127.

142. Thomas DF. Surgical treatment of urinary incontinence. Arch Dis Child. 1997;76:377–380.

143. Thomas JC, Oottamasathien S, Makari JH, et al. Temporal-spatial protein expression in bladder tissue derived from embryonic stem cells. J Urol. 2008;180:1784–1789.

144. Tian H, Bharadwaj S, Liu Y, et al. Myogenic differentiation of human bone marrow mesenchymal stem cells on a 3D nano fibrous scaffold for bladder tissue engineering. Biomaterials. 2010;31:870–877.

145. Tizzoni G, Foggi A. Die Weiderhestellung der Harnblase. Centralbl F Chir. 1888;15:921–923.

146. Truschel ST, Wang E, Ruiz WG, et al. Stretch-regulated exocytosis/endocytosis in bladder umbrella cells. Mol Biol Cell. 2002;13:830–846.

147. Tu L, Sun TT, Kreibich G. Specific heterodimer formation is a prerequisite for uroplakins to exit from the endoplasmic reticulum. Mol Biol Cell. 2002;13:4221–4230.

148. Turner A, Subramanian R, Thomas DF, et al. Transplantation of autologous differentiated urothelium in an experimental model of composite cystoplasty. Eur Urol. 2011;59:447–454.

149. Turner AM, Subramaniam R, Thomas DF, Southgate J. Generation of a functional, differentiated porcine urothelial tissue in vitro. Eur Urol. 2008;54:1423–1432.

150. Varley CL, Stahlschmidt J, Lee WC, et al. Role of PPARgamma and EGFR signalling in the urothelial terminal differentiation programme. J Cell Sci. 2004;117:2029–2036.

151. Varley CL, Garthwaite MA, Cross W, Hinley J, Trejdosiewicz LK, Southgate J. PPARgamma-regulated tight junction development during human urothelial cytodifferentiation. J Cell Physiol. 2006;208:407–417.

152. Varley CL, Bacon EJ, Holder JC, Southgate J. FOXA1 and IRF-1 intermediary transcriptional regulators of PPARgamma-induced urothelial cytodifferentiation. Cell Death Differ. 2009;16:103–114.

153. Vaught JD, Kropp BP, Sawyer BD, et al. Detrusor regeneration in the rat using porcine small intestinal submucosal grafts: functional innervation and receptor expression. J Urol. 1996;155:374–378.

154. Von-Mikulicz J. Zur operation der angebarenen blaben-Spalte. Zentralbl Chir. 1889;20:641–643.

155. Wang AY, Leong S, Liang YC, Huang RC, Chen CS, Yu SM. Immobilization of growth factors on collagen scaffolds mediated by polyanionic collagen mimetic peptides and its effect on endothelial cell morphogenesis. Biomacromolecules. 2008;9:2929–2936.

156. Wezel F, Pearson J, Southgate J. Plasticity of in vitro-generated urothelial cells for functional tissue formation. Tissue Eng Part A 2014; (in press: PMID24350594).

157. Wiesmann HP, Lammers L. Scaffold structure and fabrication. In: Meyer U, Meyer T, Handschel J, Wiesmann HP, eds. Fundamentals of tissue engineering and regenerative medicine. Springer 2009.

158. Wu XR, Manabe M, Yu J, Sun TT. Large scale purification and immunolocalization of bovine uroplakins I, II, and III Molecular markers of urothelial differentiation. J Biol Chem. 1990;265:19170–19179.

159. Yoo JJ, Meng J. Bladder augmentation using allogenic bladder submucosa seeded with cells. Urology. 1998;51:221–225.

160. Yu J, Lin JH, Wu XR, Sun TT. Uroplakins Ia and Ib, two major differentiation products of bladder epithelium, belong to a family of four transmembrane domain (4TM) proteins. J Cell Biol. 1994;125:171–182.

161. Zhang Y, Kropp BP, Moore P, et al. Coculture of bladder urothelial and smooth muscle cells on small intestinal submucosa: potential applications for tissue engineering technology. J Urol. 2000;164:928–934 discussion 934–5.

162. Zhang Y, Kropp BP, Lin HK, Cowan R, Cheng EY. Bladder regeneration with cell-seeded small intestinal submucosa. Tissue Eng. 2004;10:181–187.

163. Zhang Y, Frimberger D, Cheng EY, Lin HK, Kropp BP. Challenges in a larger bladder replacement with cell-seeded and unseeded small intestinal submucosa grafts in a subtotal cystectomy model. BJU Int. 2006;98:1100–1105.

..................Content has been hidden....................

You can't read the all page of ebook, please click here login for view all page.
Reset