Next Article in Journal
Radiation-Induced Overexpression of TGFβ and PODXL Contributes to Colorectal Cancer Cell Radioresistance through Enhanced Motility
Next Article in Special Issue
Stem Cells and Exosomes: New Therapies for Intervertebral Disc Degeneration
Previous Article in Journal
Gut Microbiota and Neuroplasticity
Previous Article in Special Issue
Mesenchymal Stem Cells, Bioactive Factors, and Scaffolds in Bone Repair: From Research Perspectives to Clinical Practice
 
 
Correction published on 18 December 2023, see Cells 2023, 12(24), 2857.
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Cell Therapy for Anal Sphincter Incontinence: Where Do We Stand?

1
Division of Digestive Surgery, University Hospitals of Geneva, 1205 Geneva, Switzerland
2
Department of Surgery, Geneva Medical School, University of Geneva, 1205 Geneva, Switzerland
3
Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
4
Faculty of Science and Medicine, University of Fribourg, 1700 Fribourg, Switzerland
*
Author to whom correspondence should be addressed.
Cells 2021, 10(8), 2086; https://doi.org/10.3390/cells10082086
Submission received: 30 June 2021 / Revised: 5 August 2021 / Accepted: 6 August 2021 / Published: 13 August 2021 / Corrected: 18 December 2023
(This article belongs to the Collection Stem Cells in Tissue Engineering and Regeneration)

Abstract

:
Anal sphincter incontinence is a chronic disease, which dramatically impairs quality of life and induces high costs for the society. Surgery, considered as the best curative option, shows a disappointing success rate. Stem/progenitor cell therapy is pledging, for anal sphincter incontinence, a substitute to surgery with higher efficacy. However, the published literature is disparate. Our aim was to perform a review on the development of cell therapy for anal sphincter incontinence with critical analyses of its pitfalls. Animal models for anal sphincter incontinence were varied and tried to reproduce distinct clinical situations (acute injury or healed injury with or without surgical reconstruction) but were limited by anatomical considerations. Cell preparations used for treatment, originated, in order of frequency, from skeletal muscle, bone marrow or fat tissue. The characterization of these preparations was often incomplete and stemness not always addressed. Despite a lack of understanding of sphincter healing processes and the exact mechanism of action of cell preparations, this treatment was evaluated in 83 incontinent patients, reporting encouraging results. However, further development is necessary to establish the correct indications, to determine the most-suited cell type, to standardize the cell preparation method and to validate the route and number of cell delivery.

1. Introduction

Anal sphincter incontinence (ASI) is defined by an involuntary loss of gases, solids and/or liquid materials. The prevalence of ASI is widely underestimated and ranges from 0.4 to 24% [1]. ASI is an underrated pathology in regards to its morbidity and its impact on the society and is associated with impaired quality of life and psychopathologies such as depression, anxiety disorder and impaired sexual function [2]. In the USA, ASI’s global cost has been estimated to be around 11 billion dollars per year [2].
The physiopathology of ASI is not completely understood but results from a functional impairment of one or more of the four anatomic components that allow a normal continence: The internal anal sphincter (IAS), the external anal sphincter (EAS), the pelvic floor muscles and the sensory-motor apparatus [3]. ASI is primarily managed conservatively but sphincter repair surgery is proposed for large defects of either EAS, IAS or both with conservative treatment failure [4,5]. However, long-term results of anal sphincter repair are unsatisfactory, notably regarding IAS repair [6,7,8,9]. Several strategies have been developed to improve sphincter repair surgery or to replace it without outstanding successes [10,11,12]. Since more than a decade, the transplantation of stem cells (multipotent cells with limited proliferation ability) or progenitor cells (multipotent cells with limited proliferation ability) has been proposed as an adjunct or direct therapy for ASI following the hypothesis that transplanted cells will either commit differentiation into EAS or IAS muscle or produce growth factors that will favor sphincter healing (Table S1). A consensus among researchers was never reached, and various source cells and protocols have been proposed and investigated with preclinical and clinical studies. We present herein a critical analysis of cell therapy for ASI. Of note, as transplanted cells were heterogenous regarding their origin and phenotype, the generic term “multipotent cell” will be used to describe the transplanted cells.

2. Physiopathology of Anal Sphincter Injuries, Models and Cell Therapy-Based Strategies

2.1. Mechanisms of Sphincter Injuries

In patients with ASI, there are at least two mechanisms leading to a modification in the pelvic floor structure or function. First, sphincters can be injured by perineal tearing, stretching and/or ischemia; obstetric trauma being the most common cause [13]. The episiotomy procedure is also associated with anal incontinence [14]. A second mechanism involves the degeneration of pelvic floor muscles, which appears with ageing, but rapidly after menopause as pelvic floor structures are hormone responsive [13]. Proposed animal models of anal incontinence tried to simulate distinct clinical situations where continence is impaired by an acute or chronic mechanism. In these models, various methods were used to harm these structures, including muscle section or excision, thermal injury and even nerve crushing [15] (Table 1). However, these experimental models of ASI do not consider the stretching effect of pelvic floor that can occur during childbirth. Thus, some authors have proposed a more realistic rodent model of childbirth injury by means of a balloon inflated in the vagina for a prolonged time [16,17]. This model was, however, never used to assess the potential of cell therapy to cure ASI.

2.2. Acute Anal Sphincter Injury and Healing

The classical clinical situation of acute anal sphincter injury is well illustrated by childbirth trauma, which still occurs in 11% of vaginal deliveries and can extend up to the IAS and sometimes the rectum (starting from posterior wall of vagina) [49] (Figure 1). In the case of extending traumatism to the level of rectal wall, dedicated stem cells of the anal canal transition zone, positive for cytokeratin 17, participate in the healing process of the mucosa of the rectum and the anal canal [50]. On the other hand, the healing process of anal sphincters has not been thoroughly studied and it is supposed to be very similar to other muscle-healing processes [51]. Under optimal conditions, healing ultimately leads to the generation of new myofibers/smooth muscle cells from muscle satellite cells/progenitor smooth muscle cells or the reparation of damaged myofibers after fusion with muscle satellite cells [52,53]. After anal sphincter injury, it is thought that inflammatory cells’ cross-talk produces cytokines and growth factors that will recruit stem cells and progenitor cells [51,53]. These cells could be mobilized from bone marrow or the surrounding tissues and might favor healing, be incorporated into the wound or further recruit stem cells and progenitor cells [51,53,54]. Especially, skeletal muscle satellite cells, which are localized in periphery of myofibers, near vascular or nerve structures, are a source of myoblasts and, further, new myocytes after their activation [55,56]. However, Lorenzi et al. reported in rats, after direct anal sphincter injury and repair, the persistence of fibrous tissue with dilated blood vessels and muscle cell degeneration patterns [19].
Progenitor cells produce cytokines like stromal-derived factor 1 (SDF-1) that are both chemoattractive for progenitor cells (including myoblasts and smooth muscle progenitor cells) but also contribute to cell proliferation, migration and survival [51,57,58]. SDF-1 seems to be a prominent cytokine for anal sphincter healing. Salcedo et al. reported a rapid local burst of SDF-1 and monocyte chemotactic protein-3 (MCP-3) expression in rats one hour after sphincter injury and up to 21 days after injury [59]. Moreover, an injection of plasmids with SDF-1 directly into EAS muscle or transplantation of SDF-1 transfected progenitor cells both improved continence in rodents after partial sphincterectomy with the same extend [35,36].
In theory, progenitor cells could be transplanted soon after injury and this is considered to be the best option to maximize the effect of transplantation (Figure 2) [51]. Indeed, it is supposed that progenitor cells might increase the natural healing process notably through the local release of cytokines such as SDF-1 [51,59]. This strategy was evaluated in several preclinical studies where progenitor cells were either directly injected after injury with surgical reparation, or not, to mimic the situation of a direct repair of sphincters [18,19,20,21,22,23,24,25,32]. Of note, some authors delayed this intervention by 24 h [15,31,32,38]. To the best of our knowledge, progenitor cells have never been injected in a patient with acute anal sphincter injury.

2.3. Unrepaired Anal Sphincter Injury

Most often, an anal sphincter tear is usually identified after delivery and surgically repaired [49]. Sometimes, the diagnosis can be missed, resulting in an occult anal sphincter injury [60]. As for an acute anal injury, little is known about the long-term healing and remodeling of damaged muscle, and an analogy can be made with the repair processes of skeletal and smooth muscles from other localizations (cf. Section 2.2). Probably, some unrepaired anal sphincter lesions could spontaneously heal. There exists evidence that a clear cut through anal sphincters in rats can heal spontaneously without inducing ASI [31,39]. As a result, almost all models of ASI in rats imply the partial resection of anal sphincters. In rat models where simple anal sphincters section were performed, acute and chronic inflammation was seen at the site of injury, characterized by neutrophils, monocytes macrophages infiltrations and fibrous tissue [20,22], disorganization of striated fibers of the EAS [35,37], but also mucin pool inclusions with histiocytes [34]. In rabbits, Rajasekaran investigated the effects of an EAS clear-cut section over time. The authors reported early collagen deposits from one week after injury, but also extensive fibrosis appearing three weeks after injury, at the site of myotomy but also beyond [61]. The presence of fibrosis was confirmed by other authors in rabbits [40,43]. In dogs, three weeks after IAS and EAS partial excision, Kang et al. reported focal interstitial inflammation, fibrosis and atrophy of smooth and striated muscles [46].
Anal sphincters encircle the anal canal, and the loss of this circular shape, as a result of injury, directly impairs the continence function [62]. However, during sphincter repair surgery, one can observe that the retracted muscle edges are held together with fibrous tissue, which bridges the defect [63]. Thus, unrepaired damage to IAS or EAS does not evolve into a hole in the sphincter ring (EAS and/or IAS), but rather into altered tissue, which fills the breach. In rats, this tissue contained mast cells and other inflammatory cells [18]. To the best of our knowledge, the importance of this tissue was never investigated in humans but could be of importance, especially if the muscle gap is the target of cell therapy.
In the majority of preclinical studies, researchers investigated the direct injection of mesenchymal stem cells (MSC) in the area of muscle injury (Figure 2). This approach could be wrong as recent evidence stated that the transplantation of stem cells of mesenchymal origin during the late phase of healing can be deleterious and increase fibrosis [64]. Surprisingly, in the majority of fundamental studies modeling anal injury, the recovery time before cell therapy was very short (days) whereas after sphincter injury, ASI appears years or even decades after the initial trauma [65]. Therefore, future models should also consider a realistic healing time after anal sphincter injury before applying cell therapy. Indeed, trials with patients suffering from long-lasting ASI with EAS lesions have investigated the direct injection of multipotent cells into EAS lesions [66], muscle edges [66,67] or entire muscle [68,69].
Another strategy to treat patients with ASI is the transplantation of a biosphincter. Bitar’s group, from the Wake Forest Institute for Regenerative Medicine, has been working for decades on the implantation of bioengineered anal sphincters composed of innervated smooth muscle cells (Table S2). The results are promising and the group recently implanted the construct in large animals, including non-human primates [47,48]. This strategy is interesting as it provides a complete and functional IAS substitute and might be part of a reconstruction strategy for patients in whom IAS has been totally or partially removed such as for ultra-low rectal cancer resections. However, IAS engineering has some hurdles to overcome. First, this approach uses IAS or digestive smooth muscle cells and intestinal neuronal cells. For translation to the clinic, tissue might be procured from organ donors, requiring further immunosuppression and exposing patients to its associated risks. Those risks need to be balanced with the fact that ASI is a non-life-threatening condition. Moreover, Araki et al. recently demonstrated the feasibility of anorectal transplantation in a dog model, with this approach being serious concurrent to allogenic biosphincters [70]. Further, the tissue culture of such constructs requires 6 to 8 weeks of culture, which increases the risk of microbial contamination. Finally, the cost of the creation of an organic construct seems to be higher than cell isolation and expansion.

2.4. Secondary Repaired Anal Sphincter Injury

Patients with visible EAS or IAS sphincter lesions are good candidates for surgical repair. Park’s sphincteroplasty is the most common procedure where the two edges of the damaged sphincters are brought together with an overlapping suture [5,63]. Usually, the surrounding fibrous tissue, which also connects the retracted muscle, is not dissected as it offers a firm support for knotting [63]. To the best of our knowledge, the healing of such a delayed repair has never been investigated. Due to the poor long-term results of sphincteroplasty, the idea to strengthen the reparation with multipotent cells has emerged. As mentioned above, in preclinical studies, some authors injected multipotent cells during sphincter repair surgery but this was never done after secondary repair in animals [20,21,24,25,33]. However, Sarveazad et al. successfully and safely injected adipose tissue-derived multipotent cells after EAS sphincteroplasty in five women and two men (Table 2) [71].

2.5. Chronic Anal Sphincter Impairment

In some patients with ASI, a clear injury event cannot be found, and patients have either weak anal sphincter tonus without an anomaly on imaging or hypotrophy of anal sphincters on imaging. This could be the result of aging, denervation or degenerative mechanisms inducing a chronic injury to the anal sphincters [39]. IAS is responsible for the anal sphincter basal tone and contributes to up to 85% of the pressure necessary to keep the anal canal closed [75]. Indeed, IAS basal tonus is maintained without neural stimulus by the high expression of proteins inducing contractility in the IAS smooth muscle cells [76,77]. A high content of smooth muscle actin and myosin isoforms has been reported in IAS [77,78,79], but spontaneous contraction property is driven by the continuous phosphorylation of myosin light chain by protein kinase C (PKC) and Rho kinase systems; indeed proteins of these cascades, such as PKCα, RhoA, CPI-17 and HSP27 were prominent in human IAS smooth muscle cells compared to smooth muscle cells sampled from a human colon used as a control [79]. Specifically, it was demonstrated in humans that the RhoA/ROCK system is the main effector of the IAS basal tone and is constitutively activated by the renin-angiotensin system and arachidonic acid products [80]. On the other hand, IAS non-adrenergic non-cholinergic nerves negatively regulate the basal tone, essentially through nitric oxide (NO), which is the main neurotransmitter negatively affecting IAS [81]. Moreover, it was recently demonstrated in rats that RhoA/ROCK system proteins were the target of microRNA, which were reducing the IAS basal tone by a diminution of the expression of components necessary for signal transduction [82,83]. Particularly, micro-RNA-133a was increased with aging in rats and might partially explain the degeneration of IAS [82]. Other effects of aging, observed in colon smooth muscle cells, could also be present in IAS, including a diminution of RhoA translocation, a diminution in the association of the pairs actin/myosin and tropomyosin/HSP27 and a decrease in the phosphorylation of HSP27 [84]. These elements could partially explain the weakness of anal sphincters seen in older people.
Diabetes has been associated with ASI [85] and may affect anal sphincters from the cellular to the nervous level [77,82,83]. However, the precise mechanism of diabetes on anorectal function has been poorly investigated. At the cellular level, the expression of miRNA-133, linked to IAS degeneration, was also increased in diabetic rodents [82]. Diabetic neuropathy affects gastrointestinal motility [86] and measures of anal function in diabetic patients showed a reduction of maximal squeeze pressure (which is a reflection of EAS function) in diabetic patients compared to healthy controls [87]. Furthermore, patients suffering from inflammatory bowel disease are more prone to report ASI. There is probably an overlap between the alteration in bowel motility, the decrease in rectal compliance due to rectum inflammation and the sequel of repeated anal fistula surgery [88,89]. However, further investigation of muscle alteration during inflammatory bowel disease has never been carried out.
To date, there is unfortunately no model mimicking a degeneration of the muscles of EAS or IAS. Hosokawa et al. proposed a model of EAS and IAS injury using cardiotoxin but the injury was finally similar to sphincter resection [90]. This model was never used to assess cell therapy efficacity/efficiency.

3. Multipotent Cell Origin and Isolation

3.1. Multipotent Cell Origins

Multipotent cells proposed for ASI cell therapy can be derived from various tissues. The most frequent sites were skeletal muscle, bone marrow or adipose tissue. Cell preparations were either syngeneic or autologous, and allogeneic or xenogeneic transplantation was marginal (four preclinical studies and one clinical study). Cells from a muscular origin were used in the majority of the identified publications: In 17 studies, they originated from skeletal muscle or EAS [24,25,28,29,41,42,44,45,46,66,67,68,69,74,91,92,93] and in 11 studies, from smooth muscle or IAS [48,79,94,95,96,97,98,99,100,101,102] (Table 3). Four out of six clinical trials used skeletal muscle multipotent cells whereas the other used adipose tissue multipotent cells [66,67,68,69,74]. Two in vivo publications reported the use of commercial H9c2 rat heart myoblasts [20,34].
Cells of bone marrow origin were used in nine studies [15,18,19,22,31,33,36,38,40] and only six tested cells originating from adipose tissue [23,26,27,30,71,72]. Neural cells were used for bioengineered constructs in eight publications [95,96,97,98,99,102]. Bioengineered constructs used smooth muscle seeded with neuronal cells from different origins. Finally, only one publication evaluated the potential of human umbilical cord matrix cells [40].
Skeletal muscle-derived cells seem to be an interesting source for ASI therapy. Different multipotent cells can be extracted from skeletal muscle: Satellite cells and other resident multipotent cells, the former having the ability to become new satellite cells or myoblasts, the precursors of myocytes (Figure 1) [104]. Thurner et al. demonstrated that smooth muscle cells can eventually be derived from myogenic progenitors [105]. The function of other muscle resident multipotent cells is not completely understood and this category encompasses different kinds of multipotent cells such as adult pericytes, PW1+ interstitial cells and fibro-adipogenic progenitors [106].
Satellite cells have the ability to reform muscle fiber [104] and have been proposed to treat conditions such as Duchenne disease [107], coronary artery disease [108] and also urinary incontinence [109]. It is supposed that skeletal muscle multipotent cells differentiated into new myocyte and could resupply EAS with new fibers (Figure 1). Indeed, the apparition of new fibers in EAS and the expression of muscle proteins have been observed by several authors [28,42,44,45,91,92]. However, the appearance of new muscle fibers does not necessarily indicate that anal function is improved. In this regard, several in vivo studies [20,29,46] using myogenic cells were inconclusive concerning anal function recovery despite cell engraftment confirmation.
Cells derived from adipose tissue constitute an interesting option, as subcutaneous fat tissue is easily accessible. The cells used in the reported studies [23,26,27,71] had the characteristics of MSC including the ability to differentiate into various tissues. MSC are multipotent cells that have been evaluated over the last years to treat various conditions including spinal cord injury, corneal or uvea injury, lung injury, cerebral injury, colitis, alopecia, muscular degenerative disease, myocardial infarction, liver injury, multiple sclerosis, Parkinson disease, cancer and to improve wound healing [110]. Indeed, allogeneic adipose tissue MSCs became popular because of their poor immunogenicity and their availability after liposuction surgery. However, recent evidence demonstrated the development of donor-specific antibodies, and MSC rejection has been documented [111]. Besides MSC’s ability to differentiate into various cells, their paracrine action have been proposed to mediate most of their effects [110,112]. MSC produce a large amount of growth factors and extracellular vesicles [113]. Strategies using encapsulated MSC conserve the effects of MSC, notably on liver fibrosis, confirming the efficacy of paracrine action and treatment [114]. Thus, MSC can be seen as in situ bioreactors delivering growth factors to neighboring cells. MSC have the ability to induce smooth muscle regeneration from the gut and the bladder [115,116] and also skeletal muscle regeneration [117,118]. Thus, MSC therapy may promote the healing and regeneration of both IAS and EAS.

3.2. Methods for Multipotent Cell Isolation and Processing

Stem cell and progenitor cells were retrieved from rats, mice, rabbits, non-human primate or humans (Table S1). Different harvest methods were used according to the origin of the collected tissue, but protocols were similar to existing standard, with the first step of washing and decontamination followed by the digestion of tissue and, finally, the purification of the cell suspension before plating. In the majority of studies, isolation procedures were sufficiently detailed, but some studies lack essential information concerning the isolation procedures. Skeletal muscle was digested with collagenase I [41,42,98], collagenase type II [93], collagenase type IV [92] collagenase type XI [29,46,91], collagenase NB6 [69], trypsin [91] and and/or dispase II [44,45,91]. Intestinal smooth muscle was digested using collagenase I [94] or collagenase II [79,93,95,97,99,100,101,102,103,119]. Enteric neurons were isolated after the digestion of tissue with collagenase II and dispase II [15,95,97,102,103]. For bone-marrow MSC, bones were flushed, and bone marrow collected, washed, sometimes fractionated with density gradient and plated [18,31,32,35,36,37,38]. For fat multipotent cells, adipose tissue was digested with collagenase I [23,26,27,71].

3.3. Methods for Multipotent Cell Characterization

The characterization of isolated multipotent cells is mandatory for reproducibility but also for quality purpose, especially when a clinical application for ASI treatment is foreseen. Among publications using muscle multipotent cells, only 13 studies reported or referred to a proper characterization of the cells and were heterogeneous for markers (Table 3) [28,29,41,42,44,45,46,48,66,67,68,69,93]. Indeed, international criteria for MSC definition were not always applied/fulfilled/verified [120].
Cells of bone marrow origin were used in nine publications [15,18,19,22,31,33,36,40,118] and were well characterized only in seven [15,18,19,31,33,35,38]. Cells originating from adipose tissue were more often precisely characterized and were at least CD90+ and CD45 [23,26,27,30,71]. Satellite cells have typical features such as the expression of the transcription factor PAX7 (Figure 1) [104] but there is currently a lack of standardization in the nomenclature and characterization of other myogenic cells [121]. Thus, multipotent cells from a skeletal origin were widely used to treat in vivo models of ASI, but their efficacy, as well as their identification, remained elusive. As the cell types used were insufficiently characterized, it cannot be excluded that the beneficial effect on ASI was partially mediated by co-isolated contaminant multipotent cells from connective tissue. Moreover, if we assume that satellite cells were responsible for the positive effects of skeletal muscle cell preparation on ASI, this effect was thus induced by their action on the skeletal muscle of EAS and not on the smooth muscle of IAS.

4. Multipotent Cell Transplantation

4.1. Practical Considerations

Before transplantation, cells were cultivated on plastic dishes and the number of passages before injection in the identified clinical trials ranged from three to ten. The number of injected cells, clearly reported by identified in vivo reports, ranged from 10,000 up to 90 million. In clinical trials, this number ranged from 200,000 up to 2 billion. However, the minimum number of cells required to obtain a beneficial effect on ASI remains elusive and only a few authors performed a real titration [28,92]. Thus, an excess of cells was used to compensate stem cell and progenitor cell death. Transplanted cell survival is a main concern in the field of cell therapy and it can be impaired by several factors related to the mechanical force applied during cell application, detachment from cell substrate and receiving site with inflammation and/or local hypoxia [122]. Indeed, forces generated during injection with a syringe needle are sufficient to induce up to 40% of cell death [122]. Thus, strategies have been developed to improve cell engraftment and transplantation success.

4.2. Adjuvant Therapy

As a strategy to limit cellular stress due to transplantation, some authors proposed to protect cells with biomaterials [44,45,46,110]. Indeed, it is known that preserving cell-extracellular substrate interactions can limit stem cell apoptosis [122]. Biomaterials were typically scaffolds of decellularized matrices or hydrogel polymers [23,33,34,35,43]. Deserving the same purpose, multipotent cells were also transplanted as sheets of cells instead of individual cells [26]. Alternatively, Trébol et al. seeded suture thread with MSC to be used for sphincter reconstruction [27] whereas Ding et al. reinforced reconstruction with a patch of an acellular dermal matrix also seeded with MSC [33].
As mentioned earlier, in some studies, authors performed immediate injection of cells along with sphincter repair, confronting cells with an acute inflammatory environment that presumably precludes cell survival [122]. On the other hand, inflammation can also enhance cell settling and homing, and the injection of myoblasts in healthy regions of EAS did not restore continence compared to the injection of cells into injured parts [28,122]. Other known strategies to improve cell survival implicated pre-conditioning of cells with either thermal preconditioning, hypoxic preconditioning, acidic preconditioning or nutrient deprivation preconditioning [122]. The goal of these strategies is to induce anti-apoptotic protein expression [122]. Injection sites were also prepared, and electrical stimulation was used in two in vivo studies [37,38] and two clinical studies to promote the homing of cells into anal sphincters [66,68]. Moreover, laser beam stimulation along with cell therapy was used in one study to promote muscle proliferation [30].
Further, growth factors (SDF-1, FGF) described to promote stem cell implantation were delivered in situ by bioscaffolds or through osmotic pumps [35,37,99,100,103]. Two studies transfected cells with SDF-1 plasmids before implantation [35,36]. As an alternative to the use of growth factors, platelet rich plasma, which is known to contain numerous growth factors, might be transplanted conjointly with stem cells [123].

4.3. Measure of Outcomes and Results

For in vitro studies, physiological functional evaluation was carried out in almost all identified studies to assess the contractility potential of constructed sphincters. For in vivo studies, outcomes were highly variable: The most common methods for outcome assessment were histology, anorectal manometry, physiological functionality evaluation and electromyography or electrophysiology. Some authors tracked cells using magnetic resonance imaging [42] or labeled them with fluorescent proteins (GFP) [91].
The determination of the outcome in a clinical study on anal incontinence is challenging. Different definitions of ASI exists, including incontinence to gases or not [1]. Further, in order to be comparable to the literature, authors are choosing outcomes that appear to be a gold standard in the medical literature. In almost all published clinical studies, one incontinence score was used as the primary outcome. However, a recent analysis of different incontinence scores pointed out that no single score reaches relevant psychometric soundness and recommended the use of at least two scores to evaluate ASI [124]. However, the utilization of objective outcomes may be more reliable such as high-resolution anal manometry or contact EMG (For e.g., MAPLe® device, Medtronic, Dublin, Ireland [125]).

4.4. Results

A review of the literature identified a total of 52 original publications. Seven publications reported in vitro results (Table S2) [79,93,94,95,97,98,119], with six on bioengineered constructs among them [79,93,94,95,97,98]. One publication reported the isolation of cells from human IAS and EAS and assessed their viability [93]. In vivo experiments were reported in 38 publications (Table S1) [15,18,19,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,37,38,40,41,42,43,44,45,46,48,91,92,96,99,100,101,102,103], including five articles on heterotopic sphincter bioconstruct implantation [96,99,100,101,103]. Seven human studies were identified (Table 2) [66,67,68,69,71,72,74], including three randomized controlled trials [69,70,71,72] and one case report [74]. A total of 83 patients received cell therapy for ASI treatment.
Almost all patients included in clinical trials exhibited EAS injury. Four out of seven studies used the Wexner score as the primary outcome. The FIQL score was used in two studies. Other variables measured were anorectal manometry (5/7), endoanal ultrasonography (3/7) and electromyography (EMG) (3/7). The longest follow-up was reported by the group of Frudinger et al. who injected autologous myoblasts into EAS in 10 voluntary women with EAS defect or atrophy [66]. After a follow-up of 5 years, the mean Wexner score decreased from 15.3 (SD-2.4) before intervention to 0.7 (SD 1.3) (p > 0.001). In addition, anal manometry demonstrated an improvement of median resting and squeeze pressures (20 (IQR 17–28) to 32 (25–43) and 23 (IQR 20–34) to 33 (IQR 31–66), respectively). The same group started a second trial including 34 females and 5 males and found a reduction of Wexner score of −16.2 (SD-3.66) for women and -18.8 (SD-1.30) for male at one year. These results were better than sphincteroplasty, which typically induces a long-term reduction of −1 to −5.2 of median/mean Wexner score (mean follow-up between seven and eight years) [126,127]. In a similar study (muscle tissue-derived multipotent cells injected into EAS), Boyer et al. reported a reduction of median Wexner score of −6.4 (range −12 to 2) (p = 0.006) for the intervention group and a reduction of −1 (range −8 to 6) (p = 0.35) for the placebo [69]. However, using adipose tissue-derived multipotent cells in a randomized triple blinded placebo-controlled trial, De la Portilla et al. failed to demonstrate any effect on Wexner score of cell transplantation into EAS defect [72]. Globally, all seven reports described encouraging results regarding at least one of the measured outcomes (which was not necessary the primary outcome).
Until now, in vivo experiments have demonstrated a relative oncological safety of such a strategy. One in vivo study described focal cell growth at the injection site but without malignant characteristics [25]. Recent evidence confirmed that pure stem cell cultures, particularly MSC, did not develop malignant cells [128,129]. However, MSC have opposite effects on tumor cells and can promote or suppress tumor growth in vitro and in vivo [130]. Published clinical trials, which used either skeletal muscle tissue or adipose tissue multipotent cells, demonstrated that the procedures were safe.

5. Conclusions

Preclinical and clinical studies have demonstrated the safety of multipotent cell utilization for ASI. Published clinical studies have shown promising results, but only three of them were controlled with placebo injection [69,70,71,72]. These controlled studies had relative short follow-ups considering that ASI is a disease evolving over time. Thus, all the presented strategies for cell therapy in the context of ASI deserve further randomized controlled trials with improved methodology regarding outcome measurement and follow-up.
The ideal therapy for ASI should be cost-effective with a long-lasting effect. Apart from research and development costs, good manufacturing practices, GMP certifications and implementation charges, routine use of cell therapy appears to be highly costly [131]. Trébol et al. estimated the maximal production costs in Spain to be 7400 USD for 40 million autologous fat-derived cells or 8500 € for 100 million allogeneic fat-derived cells [130]. In our hospital, the cost for a sphincteroplasty, with a typical length of stay of two days, is 5000 USD. Recently, Gräs et al. proposed a cost-effective alternative to cell transplantation for anal sphincter regeneration. Following promising results for urinary incontinence, the authors discussed the possibility to inject fragmented muscle fibers, instead of expanded cells, into injured anal sphincters [121].
After transplantation, progenitor cells and stem cells might act by paracrine effects and/or by differentiation into functional muscular cells. It should be pointed out that the exact underlying mechanism remains poorly understood, and that basic research on this topic is still required to understand which factors and conditions are leading to cell engraftment, differentiation and finally tissue regeneration [123,132]. Moreover, the natural history of sphincter lesion/repaired sphincter healing should be better understood in order to select appropriate cell preparations and transplantation techniques. Some groups reported a different approach, considering the use of cell therapy as an add-on to sphincter repair, either directly after lesion, to simulate the primary repair of an acute obstetrical tear or at distance [18,19,20,21,22,23,24,25]. Thus, it remains to be determined how stem cells and progenitor cells should be used for ASI: As a substitution to surgery or along with surgery.

Supplementary Materials

The following are available online at https://www.mdpi.com/2073-4409/10/8/2086/s1, Table S1. List and description of identified in vivo publications investigating cell therapy for the treatment of anal sphincter incontinence, Table S2. List of in vitro publications.

Author Contributions

Conceptualization, A.B., B.R. and J.M.; methodology, A.B. and J.M.; software, A.B.; validation, A.B., J.M., resources, A.B., L.H.B., E.L., C.G.-G. and C.T.; writing—original draft preparation, A.B., J.M., R.P.H.M. and C.G.-G.; writing—review and editing, A.B., J.M., R.P.H.M., E.L., N.C.B., B.R., C.T., L.H.B., C.G.-G., F.R.; visualization, A.B.; supervision, L.H.B., C.T., C.T. and F.R.; project administration, A.B.; funding acquisition, A.B., F.R. and C.T. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by University Hospital of Geneva “Projet Recherche&Développement”, PRD 19-I-2020.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.

References

  1. Chatoor, D.R.; Taylor, S.J.; Cohen, C.R.G.; Emmanuel, A.V. Faecal Incontinence. Br. J. Surg. 2007, 94, 134–144. [Google Scholar] [CrossRef] [PubMed]
  2. Meyer, I.; Richter, H.E. Impact of Fecal Incontinence and Its Treatment on Quality of Life in Women. Womens Health Lond. Engl. 2015, 11, 225–238. [Google Scholar] [CrossRef]
  3. Williams, K.S.; Shalom, D.F.; Winkler, H.A. Faecal Incontinence: A Narrative Review of Clinic-Based Management for the General Gynaecologist. J. Obstet. Gynaecol. J. Inst. Obstet. Gynaecol. 2018, 38, 1–9. [Google Scholar] [CrossRef]
  4. Nandivada, P.; Nagle, D. Surgical Therapies for Fecal Incontinence. Curr. Opin. Gastroenterol. 2014, 30, 69–74. [Google Scholar] [CrossRef] [PubMed]
  5. Wexner, S.D.; Bleier, J. Current Surgical Strategies to Treat Fecal Incontinence. Expert Rev. Gastroenterol. Hepatol. 2015, 9, 1577–1589. [Google Scholar] [CrossRef] [PubMed]
  6. Zorcolo, L.; Covotta, L.; Bartolo, D.C.C. Outcome of Anterior Sphincter Repair for Obstetric Injury: Comparison of Early and Late Results. Dis. Colon Rectum 2005, 48, 524–531. [Google Scholar] [CrossRef] [PubMed]
  7. Gronewold, M.; Kroencke, T.; Hagedorn, A.; Tunn, R.; Gauruder-Burmester, A. [External anal sphincter repair using the overlapping technique in patients with anal incontinence and concomitant pudendal nerve damage]. Zentralblatt Für Chir. 2008, 133, 129–134. [Google Scholar] [CrossRef]
  8. Demirbas, S.; Atay, V.; Sucullu, I.; Filiz, A.I. Overlapping Repair in Patients with Anal Sphincter Injury. Med. Princ. Pract. Int. J. Kuwait Univ. Health Sci. Cent. 2008, 17, 56–60. [Google Scholar] [CrossRef]
  9. Brown, S.R.; Wadhawan, H.; Nelson, R.L. Surgery for Faecal Incontinence in Adults. Cochrane Database Syst. Rev. 2013, CD001757. [Google Scholar] [CrossRef]
  10. Zutshi, M.; Hull, T.; Gurland, B. Anal Encirclement with Sphincter Repair (AESR Procedure) Using a Biological Graft for Anal Sphincter Damage Involving the Entire Circumference. Colorectal Dis. Off. J. Assoc. Coloproctology G. B. Irel. 2012, 14, 592–595. [Google Scholar] [CrossRef]
  11. De la Portilla, F. Internal Anal Sphincter Augmentation and Substitution. Gastroenterol. Rep. 2014, 2, 106–111. [Google Scholar] [CrossRef]
  12. Alam, N.N.; Narang, S.K.; Köckerling, F.; Daniels, I.R.; Smart, N.J. Anal Sphincter Augmentation Using Biological Material. Front. Surg. 2015, 2. [Google Scholar] [CrossRef] [PubMed]
  13. Rao, S.S.C. Pathophysiology of Adult Fecal Incontinence. Gastroenterology 2004, 126, S14–S22. [Google Scholar] [CrossRef] [PubMed]
  14. LaCross, A.; Groff, M.; Smaldone, A. Obstetric Anal Sphincter Injury and Anal Incontinence Following Vaginal Birth: A Systematic Review and Meta-Analysis. J. Midwifery Womens Health 2015, 60, 37–47. [Google Scholar] [CrossRef] [PubMed]
  15. Salcedo, L.; Mayorga, M.; Damaser, M.; Balog, B.; Butler, R.; Penn, M.; Zutshi, M. Mesenchymal Stem Cells Can Improve Anal Pressures after Anal Sphincter Injury. Stem Cell Res. 2013, 10, 95–102. [Google Scholar] [CrossRef] [PubMed]
  16. Healy, C.F.; O’Herlihy, C.; O’Brien, C.; O’Connell, P.R.; Jones, J.F.X. Experimental Models of Neuropathic Fecal Incontinence: An Animal Model of Childbirth Injury to the Pudendal Nerve and External Anal Sphincter. Dis. Colon Rectum 2008, 51, 1619–1626, discussion 1626. [Google Scholar] [CrossRef] [PubMed]
  17. Wai, C.Y.; Rahn, D.D.; White, A.B.; Word, R.A. Recovery of External Anal Sphincter Contractile Function after Prolonged Vaginal Distention or Sphincter Transection in an Animal Model. Obstet. Gynecol. 2008, 111, 1426–1434. [Google Scholar] [CrossRef] [PubMed]
  18. Mazzanti, B.; Lorenzi, B.; Borghini, A.; Boieri, M.; Ballerini, L.; Saccardi, R.; Weber, E.; Pessina, F. Local Injection of Bone Marrow Progenitor Cells for the Treatment of Anal Sphincter Injury: In-Vitro Expanded versus Minimally-Manipulated Cells. Stem Cell Res. Ther. 2016, 7, 85. [Google Scholar] [CrossRef]
  19. Lorenzi, B.; Pessina, F.; Lorenzoni, P.; Urbani, S.; Vernillo, R.; Sgaragli, G.; Gerli, R.; Mazzanti, B.; Bosi, A.; Saccardi, R.; et al. Treatment of Experimental Injury of Anal Sphincters with Primary Surgical Repair and Injection of Bone Marrow-Derived Mesenchymal Stem Cells. Dis. Colon Rectum 2008, 51, 411–420. [Google Scholar] [CrossRef]
  20. Fitzwater, J.L.; Grande, K.B.; Sailors, J.L.; Acevedo, J.F.; Word, R.A.; Wai, C.Y. Effect of Myogenic Stem Cells on the Integrity and Histomorphology of Repaired Transected External Anal Sphincter. Int. Urogynecol. J. 2015, 26, 251–256. [Google Scholar] [CrossRef]
  21. White, A.B.; Keller, P.W.; Acevedo, J.F.; Word, R.A.; Wai, C.Y. Effect of Myogenic Stem Cells on Contractile Properties of the Repaired and Unrepaired Transected External Anal Sphincter in an Animal Model. Obstet. Gynecol. 2010, 115, 815–823. [Google Scholar] [CrossRef] [PubMed]
  22. Pathi, S.D.; Acevedo, J.F.; Keller, P.W.; Kishore, A.H.; Miller, R.T.; Wai, C.Y.; Word, R.A. Recovery of the Injured External Anal Sphincter after Injection of Local or Intravenous Mesenchymal Stem Cells. Obstet. Gynecol. 2012, 119, 134–144. [Google Scholar] [CrossRef]
  23. Kuismanen, K.; Juntunen, M.; Narra Girish, N.; Tuominen, H.; Huhtala, H.; Nieminen, K.; Hyttinen, J.; Miettinen, S. Functional Outcome of Human Adipose Stem Cell Injections in Rat Anal Sphincter Acute Injury Model. Stem Cells Transl. Med. 2018, 7, 295–304. [Google Scholar] [CrossRef]
  24. Lane, F.L.; Jacobs, S.A.; Craig, J.B.; Nistor, G.; Markle, D.; Noblett, K.L.; Osann, K.; Keirstead, H. In Vivo Recovery of the Injured Anal Sphincter after Repair and Injection of Myogenic Stem Cells: An Experimental Model. Dis. Colon Rectum 2013, 56, 1290–1297. [Google Scholar] [CrossRef] [PubMed]
  25. Jacobs, S.A.; Lane, F.L.; Pham, Q.-A.; Nistor, G.; Robles, R.; Chua, C.; Boubion, B.; Osann, K.; Keirstead, H. Safety Assessment of Myogenic Stem Cell Transplantation and Resulting Tumor Formation. Female Pelvic Med. Reconstr. Surg. 2013, 19, 362–368. [Google Scholar] [CrossRef]
  26. Inoue, Y.; Fujita, F.; Yamaguchi, I.; Kinoe, H.; Kawahara, D.; Sakai, Y.; Kuroki, T.; Eguchi, S. Improvement of Anal Function by Adipose-Derived Stem Cell Sheets. Dig. Surg. 2018, 35, 64–69. [Google Scholar] [CrossRef] [PubMed]
  27. Trébol, J.; Georgiev-Hristov, T.; Vega-Clemente, L.; García-Gómez, I.; Carabias-Orgaz, A.; García-Arranz, M.; García-Olmo, D. Rat Model of Anal Sphincter Injury and Two Approaches for Stem Cell Administration. World J. Stem Cells 2018, 10, 1–14. [Google Scholar] [CrossRef]
  28. Bisson, A.; Fréret, M.; Drouot, L.; Jean, L.; Le Corre, S.; Gourcerol, G.; Doucet, C.; Michot, F.; Boyer, O.; Lamacz, M. Restoration of Anal Sphincter Function after Myoblast Cell Therapy in Incontinent Rats. Cell Transplant. 2015, 24, 277–286. [Google Scholar] [CrossRef]
  29. Kang, S.-B.; Lee, H.N.; Lee, J.Y.; Park, J.-S.; Lee, H.S.; Lee, J.Y. Sphincter Contractility after Muscle-Derived Stem Cells Autograft into the Cryoinjured Anal Sphincters of Rats. Dis. Colon Rectum 2008, 51, 1367–1373. [Google Scholar] [CrossRef]
  30. Sarveazad, A.; Babahajian, A.; Yari, A.; Rayner, C.K.; Mokhtare, M.; Babaei-Ghazani, A.; Agah, S.; Mahjoubi, B.; Shamseddin, J.; Yousefifard, M. Combination of Laser and Human Adipose-Derived Stem Cells in Repair of Rabbit Anal Sphincter Injury: A New Therapeutic Approach. Stem Cell Res. Ther. 2019, 10, 367. [Google Scholar] [CrossRef]
  31. Salcedo, L.; Penn, M.; Damaser, M.; Balog, B.; Zutshi, M. Functional Outcome after Anal Sphincter Injury and Treatment with Mesenchymal Stem Cells. Stem Cells Transl. Med. 2014, 3, 760–767. [Google Scholar] [CrossRef] [PubMed]
  32. Li, P.; Ma, X.; Jin, W.; Li, X.; Hu, J.; Jiang, X.; Guo, X. Effects of Local Injection and Intravenous Injection of Allogeneic Bone Marrow Mesenchymal Stem Cells on the Structure and Function of Damaged Anal Sphincter in Rats. J. Tissue Eng. Regen. Med. 2020, 14, 989–1000. [Google Scholar] [CrossRef]
  33. Ding, Z.; Liu, X.; Ren, X.; Zhang, Q.; Zhang, T.; Qian, Q.; Liu, W.; Jiang, C. Galectin-1-Induced Skeletal Muscle Cell Differentiation of Mesenchymal Stem Cells Seeded on an Acellular Dermal Matrix Improves Injured Anal Sphincter. Discov. Med. 2016, 21, 331–340. [Google Scholar] [PubMed]
  34. Montoya, T.I.; Acevedo, J.F.; Smith, B.; Keller, P.W.; Sailors, J.L.; Tang, L.; Word, R.A.; Wai, C.Y. Myogenic Stem Cell-Laden Hydrogel Scaffold in Wound Healing of the Disrupted External Anal Sphincter. Int. Urogynecol. J. 2015, 26, 893–904. [Google Scholar] [CrossRef]
  35. Sun, L.; Xie, Z.; Kuang, M.; Penn, M.; Damaser, M.S.; Zutshi, M. Regenerating the Anal Sphincter: Cytokines, Stem Cells, or Both? Dis. Colon Rectum 2017, 60, 416–425. [Google Scholar] [CrossRef] [PubMed]
  36. Sun, L.; Kuang, M.; Penn, M.; Damaser, M.S.; Zutshi, M. Stromal Cell-Derived Factor 1 Plasmid Regenerates Both Smooth and Skeletal Muscle After Anal Sphincter Injury in the Long Term. Dis. Colon Rectum 2017, 60, 1320–1328. [Google Scholar] [CrossRef] [PubMed]
  37. Sun, L.; Yeh, J.; Xie, Z.; Kuang, M.; Damaser, M.S.; Zutshi, M. Electrical Stimulation Followed by Mesenchymal Stem Cells Improves Anal Sphincter Anatomy and Function in a Rat Model at a Time Remote From Injury. Dis. Colon Rectum 2016, 59, 434–442. [Google Scholar] [CrossRef]
  38. Li, X.; Guo, X.; Jin, W.; Lu, J. Effects of Electroacupuncture Combined with Stem Cell Transplantation on Anal Sphincter Injury-Induced Faecal Incontinence in a Rat Model. Acupunct. Med. J. Br. Med. Acupunct. Soc. 2018, 36, 254–260. [Google Scholar] [CrossRef] [PubMed]
  39. Yamaguchi, I.; Fujita, F.; Yamanouchi, K.; Mishima, T.; Kawahara, D.; Sakai, Y.; Ito, S.; Kanetaka, K.; Takatsuki, M.; Kuroki, T.; et al. A Novel Animal Model of Long-Term Sustainable Anal Sphincter Dysfunction. J. Surg. Res. 2013, 184, 813–818. [Google Scholar] [CrossRef]
  40. Aghaee-Afshar, M.; Rezazadehkermani, M.; Asadi, A.; Malekpour-Afshar, R.; Shahesmaeili, A.; Nematollahi-mahani, S.N. Potential of Human Umbilical Cord Matrix and Rabbit Bone Marrow-Derived Mesenchymal Stem Cells in Repair of Surgically Incised Rabbit External Anal Sphincter. Dis. Colon Rectum 2009, 52, 1753–1761. [Google Scholar] [CrossRef]
  41. Kajbafzadeh, A.-M.; Kajbafzadeh, M.; Sabetkish, S.; Sabetkish, N.; Tavangar, S.M. Tissue-Engineered External Anal Sphincter Using Autologous Myogenic Satellite Cells and Extracellular Matrix: Functional and Histological Studies. Ann. Biomed. Eng. 2016, 44, 1773–1784. [Google Scholar] [CrossRef]
  42. Elmi, A.; Kajbafzadeh, A.-M.; Oghabian, M.A.; Talab, S.S.; Tourchi, A.; Khoei, S.; Rafie, B.; Esfahani, S.A. Anal Sphincter Repair with Muscle Progenitor Cell Transplantation: Serial Assessment with Iron Oxide-Enhanced MRI. AJR Am. J. Roentgenol. 2014, 202, 619–625. [Google Scholar] [CrossRef]
  43. Kajbafzadeh, A.-M.; Elmi, A.; Talab, S.S.; Esfahani, S.A.; Tourchi, A. Functional External Anal Sphincter Reconstruction for Treatment of Anal Incontinence Using Muscle Progenitor Cell Auto Grafting. Dis. Colon Rectum 2010, 53, 1415–1421. [Google Scholar] [CrossRef]
  44. Oh, H.-K.; Lee, H.S.; Lee, J.H.; Oh, S.H.; Lim, J.-Y.; Ahn, S.; Hwang, J.-Y.; Kang, S.-B. Functional and Histological Evidence for the Targeted Therapy Using Biocompatible Polycaprolactone Beads and Autologous Myoblasts in a Dog Model of Fecal Incontinence. Dis. Colon Rectum 2015, 58, 517–525. [Google Scholar] [CrossRef] [PubMed]
  45. Oh, H.-K.; Lee, H.S.; Lee, J.H.; Oh, S.H.; Lim, J.-Y.; Ahn, S.; Kang, S.-B. Coadministration of Basic Fibroblast Growth Factor-Loaded Polycaprolactone Beads and Autologous Myoblasts in a Dog Model of Fecal Incontinence. Int. J. Colorectal Dis. 2015, 30, 549–557. [Google Scholar] [CrossRef] [PubMed]
  46. Kang, S.-B.; Lee, H.S.; Lim, J.-Y.; Oh, S.H.; Kim, S.J.; Hong, S.-M.; Jang, J.-H.; Cho, J.-E.; Lee, S.-M.; Lee, J.H. Injection of Porous Polycaprolactone Beads Containing Autologous Myoblasts in a Dog Model of Fecal Incontinence. J. Korean Surg. Soc. 2013, 84, 216–224. [Google Scholar] [CrossRef] [PubMed]
  47. Bitar, K.N.; Zakhem, E.; Bohl, J.L.; Tamburini, R.; Dadhich, P.; Scott, C.; Knutson, D.T.; Giliam, J.H. Implantation of Autologous Biosphincters in a Non-Human Primate (NHP) Model of Fecal Incontinence. Gastroenterology 2017, 152, S15. [Google Scholar] [CrossRef]
  48. Dadhich, P.; Bohl, J.L.; Tamburrini, R.; Zakhem, E.; Scott, C.; Kock, N.; Mitchell, E.; Gilliam, J.; Bitar, K.N. BioSphincters to Treat Fecal Incontinence in Nonhuman Primates. Sci. Rep. 2019, 9, 18096. [Google Scholar] [CrossRef]
  49. Meister, M.R.; Rosenbloom, J.I.; Lowder, J.L.; Cahill, A.G. Techniques for Repair of Obstetric Anal Sphincter Injuries. Obstet. Gynecol. Surv. 2018, 73, 33–39. [Google Scholar] [CrossRef]
  50. Mitoyan, L.; Chevrier, V.; Hernandez-Vargas, H.; Ollivier, A.; Homayed, Z.; Pannequin, J.; Poizat, F.; De Biasi-Cador, C.; Charafe-Jauffret, E.; Ginestier, C.; et al. A Stem Cell Population at the Anorectal Junction Maintains Homeostasis and Participates in Tissue Regeneration. Nat. Commun. 2021, 12, 2761. [Google Scholar] [CrossRef]
  51. Plair, A.; Bennington, J.; Williams, J.K.; Parker-Autry, C.; Matthews, C.A.; Badlani, G. Regenerative Medicine for Anal Incontinence: A Review of Regenerative Therapies beyond Cells. Int. Urogynecol. J. 2020. [Google Scholar] [CrossRef]
  52. Sass, F.A.; Fuchs, M.; Pumberger, M.; Geissler, S.; Duda, G.N.; Perka, C.; Schmidt-Bleek, K. Immunology Guides Skeletal Muscle Regeneration. Int. J. Mol. Sci. 2018, 19, 835. [Google Scholar] [CrossRef]
  53. Schmidt, M.; Schüler, S.C.; Hüttner, S.S.; von Eyss, B.; von Maltzahn, J. Adult Stem Cells at Work: Regenerating Skeletal Muscle. Cell. Mol. Life Sci. CMLS 2019, 76, 2559–2570. [Google Scholar] [CrossRef]
  54. Yin, H.; Price, F.; Rudnicki, M.A. Satellite Cells and the Muscle Stem Cell Niche. Physiol. Rev. 2013, 93, 23–67. [Google Scholar] [CrossRef] [PubMed]
  55. Dumont, N.A.; Wang, Y.X.; Rudnicki, M.A. Intrinsic and Extrinsic Mechanisms Regulating Satellite Cell Function. Dev. Camb. Engl. 2015, 142, 1572–1581. [Google Scholar] [CrossRef] [PubMed]
  56. Chellini, F.; Tani, A.; Zecchi-Orlandini, S.; Sassoli, C. Influence of Platelet-Rich and Platelet-Poor Plasma on Endogenous Mechanisms of Skeletal Muscle Repair/Regeneration. Int. J. Mol. Sci. 2019, 20, 683. [Google Scholar] [CrossRef]
  57. Chatterjee, M.; Huang, Z.; Zhang, W.; Jiang, L.; Hultenby, K.; Zhu, L.; Hu, H.; Nilsson, G.P.; Li, N. Distinct Platelet Packaging, Release, and Surface Expression of Proangiogenic and Antiangiogenic Factors on Different Platelet Stimuli. Blood 2011, 117, 3907–3911. [Google Scholar] [CrossRef] [PubMed]
  58. Brzoska, E.; Kowalewska, M.; Markowska-Zagrajek, A.; Kowalski, K.; Archacka, K.; Zimowska, M.; Grabowska, I.; Czerwińska, A.M.; Czarnecka-Góra, M.; Stremińska, W.; et al. Sdf-1 (CXCL12) Improves Skeletal Muscle Regeneration via the Mobilisation of Cxcr4 and CD34 Expressing Cells. Biol. Cell 2012, 104, 722–737. [Google Scholar] [CrossRef] [PubMed]
  59. Salcedo, L.; Sopko, N.; Jiang, H.-H.; Damaser, M.; Penn, M.; Zutshi, M. Chemokine Upregulation in Response to Anal Sphincter and Pudendal Nerve Injury: Potential Signals for Stem Cell Homing. Int. J. Colorectal Dis. 2011, 26, 1577–1581. [Google Scholar] [CrossRef]
  60. Andrews, V.; Sultan, A.H.; Thakar, R.; Jones, P.W. Occult Anal Sphincter Injuries--Myth or Reality? BJOG Int. J. Obstet. Gynaecol. 2006, 113, 195–200. [Google Scholar] [CrossRef]
  61. Rajasekaran, M.R.; Sinha, S.; Seo, Y.; Salehi, M.; Bhargava, V.; Mittal, R.K. Myoarchitectural and Functional Alterations in Rabbit External Anal Sphincter Muscle Following Experimental Surgical Trauma. Am. J. Physiol. Gastrointest. Liver Physiol. 2014, 307, G445–G451. [Google Scholar] [CrossRef] [PubMed]
  62. Cherry, D.A.; Rothenberger, D.A. Pelvic Floor Physiology. Surg. Clin. North Am. 1988, 68, 1217–1230. [Google Scholar] [CrossRef] [PubMed]
  63. Browning, G.G.; Motson, R.W. Anal Sphincter Injury. Management and Results of Parks Sphincter Repair. Ann. Surg. 1984, 199, 351–357. [Google Scholar] [CrossRef] [PubMed]
  64. Walraven, M.; Hinz, B. Therapeutic Approaches to Control Tissue Repair and Fibrosis: Extracellular Matrix as a Game Changer. Matrix Biol. J. Int. Soc. Matrix Biol. 2018, 71–72, 205–224. [Google Scholar] [CrossRef] [PubMed]
  65. Saldana Ruiz, N.; Kaiser, A.M. Fecal Incontinence—Challenges and Solutions. World J. Gastroenterol. 2017, 23, 11–24. [Google Scholar] [CrossRef] [PubMed]
  66. Frudinger, A.; Pfeifer, J.; Paede, J.; Kolovetsiou-Kreiner, V.; Marksteiner, R.; Halligan, S. Autologous Skeletal Muscle-Derived Cell Injection for Anal Incontinence Due to Obstetric Trauma: A Five-Year Follow-up of an Initial Study of Ten Patients. Colorectal Dis. Off. J. Assoc. Coloproctology G. B. Irel. 2015. [Google Scholar] [CrossRef] [PubMed]
  67. Romaniszyn, M.; Rozwadowska, N.; Malcher, A.; Kolanowski, T.; Walega, P.; Kurpisz, M. Implantation of Autologous Muscle-Derived Stem Cells in Treatment of Fecal Incontinence: Results of an Experimental Pilot Study. Tech. Coloproctology 2015, 19, 685–696. [Google Scholar] [CrossRef]
  68. Frudinger, A.; Marksteiner, R.; Pfeifer, J.; Margreiter, E.; Paede, J.; Thurner, M. Skeletal Muscle-Derived Cell Implantation for the Treatment of Sphincter-Related Faecal Incontinence. Stem Cell Res. Ther. 2018, 9, 233. [Google Scholar] [CrossRef]
  69. Boyer, O.; Bridoux, V.; Giverne, C.; Bisson, A.; Koning, E.; Leroi, A.-M.; Chambon, P.; Déhayes, J.; Le Corre, S.; Jacquot, S.; et al. Autologous Myoblasts for the Treatment of Fecal Incontinence: Results of a Phase 2 Randomized Placebo-Controlled Study (MIAS). Ann. Surg. 2018, 267, 443–450. [Google Scholar] [CrossRef]
  70. Araki, J.; Nishizawa, Y.; Fujita, N.; Sato, T.; Iizuka, T.; Kamata, M.; Hatayama, N.; Yakura, T.; Hirai, S.; Tashiro, K.; et al. Anorectal Transplantation: The First Long-Term Success in a Canine Model. Ann. Surg. 2021. [Google Scholar] [CrossRef]
  71. Sarveazad, A.; Newstead, G.L.; Mirzaei, R.; Joghataei, M.T.; Bakhtiari, M.; Babahajian, A.; Mahjoubi, B. A New Method for Treating Fecal Incontinence by Implanting Stem Cells Derived from Human Adipose Tissue: Preliminary Findings of a Randomized Double-Blind Clinical Trial. Stem Cell Res. Ther. 2017, 8, 40. [Google Scholar] [CrossRef] [PubMed]
  72. De la Portilla, F.; Guerrero, J.L.; Maestre, M.V.; Leyva, L.; Mera, S.; García-Olmo, D.; Rodríguez, A.; Mata, R.; Lora, F. Treatment of Fecal Incontinence with Autologous Expanded Mesenchymal Stem Cells: Results of a Pilot Study. Colorectal Dis. Off. J. Assoc. Coloproctology G. B. Irel. 2020. [Google Scholar] [CrossRef]
  73. Frudinger, A.; Kölle, D.; Schwaiger, W.; Pfeifer, J.; Paede, J.; Halligan, S. Muscle-Derived Cell Injection to Treat Anal Incontinence Due to Obstetric Trauma: Pilot Study with 1 Year Follow-Up. Gut 2010, 59, 55–61. [Google Scholar] [CrossRef]
  74. Romaniszyn, M.; Michal, R.; Rozwadowska, N.; Natalia, R.; Nowak, M.; Marcin, N.; Malcher, A.; Agnieszka, M.; Kolanowski, T.; Tomasz, K.; et al. Successful Implantation of Autologous Muscle-Derived Stem Cells in Treatment of Faecal Incontinence Due to External Sphincter Rupture. Int. J. Colorectal Dis. 2013, 28, 1035–1036. [Google Scholar] [CrossRef] [PubMed]
  75. Frenckner, B.; Euler, C.V. Influence of Pudendal Block on the Function of the Anal Sphincters. Gut 1975, 16, 482–489. [Google Scholar] [CrossRef] [PubMed]
  76. Burleigh, D.E.; D’Mello, A. Neural and Pharmacologic Factors Affecting Motility of the Internal Anal Sphincter. Gastroenterology 1983, 84, 409–417. [Google Scholar] [CrossRef] [PubMed]
  77. Bitar, K.N.; Raghavan, S. Intestinal Tissue Engineering: Current Concepts and Future Vision of Regenerative Medicine in the Gut. Neurogastroenterol. Motil. Off. J. Eur. Gastrointest. Motil. Soc. 2012, 24, 7–19. [Google Scholar] [CrossRef] [PubMed]
  78. Szymanski, P.T.; Chacko, T.K.; Rovner, A.S.; Goyal, R.K. Differences in Contractile Protein Content and Isoforms in Phasic and Tonic Smooth Muscles. Am. J. Physiol. 1998, 275, C684–C692. [Google Scholar] [CrossRef] [PubMed]
  79. Somara, S.; Gilmont, R.R.; Dennis, R.G.; Bitar, K.N. Bioengineered Internal Anal Sphincter Derived from Isolated Human Internal Anal Sphincter Smooth Muscle Cells. Gastroenterology 2009, 137, 53–61. [Google Scholar] [CrossRef]
  80. Rattan, S.; Singh, J.; Kumar, S.; Phillips, B. Nature of Extracellular Signal That Triggers RhoA/ROCK Activation for the Basal Internal Anal Sphincter Tone in Humans. Am. J. Physiol.-Gastrointest. Liver Physiol. 2015, 308, G924–G933. [Google Scholar] [CrossRef]
  81. Rattan, S. The Internal Anal Sphincter: Regulation of Smooth Muscle Tone and Relaxation. Neurogastroenterol. Motil. Off. J. Eur. Gastrointest. Motil. Soc. 2005, 17 (Suppl. S1), 50–59. [Google Scholar] [CrossRef] [PubMed]
  82. Singh, J.; Boopathi, E.; Addya, S.; Phillips, B.; Rigoutsos, I.; Penn, R.B.; Rattan, S. Aging-Associated Changes in MicroRNA Expression Profile of Internal Anal Sphincter Smooth Muscle: Role of MicroRNA-133a. Am. J. Physiol.-Gastrointest. Liver Physiol. 2016, 311, G964–G973. [Google Scholar] [CrossRef]
  83. Singh, J.; Mohanty, I.; Addya, S.; Phillips, B.; Mee Yong, H.; An, S.S.; Penn, R.B.; Rattan, S. Role of Differentially Expressed MicroRNA-139-5p in the Regulation of Phenotypic Internal Anal Sphincter Smooth Muscle Tone. Sci. Rep. 2017, 7, 1477. [Google Scholar] [CrossRef] [PubMed]
  84. Bitar, K.N. Aging and Gi Smooth Muscle Fecal Incontinence: Is Bioengineering an Option. Exp. Gerontol. 2005, 40, 643–649. [Google Scholar] [CrossRef] [PubMed]
  85. Townsend, M.K.; Matthews, C.A.; Whitehead, W.E.; Grodstein, F. Risk Factors for Fecal Incontinence in Older Women. Am. J. Gastroenterol. 2013, 108, 113–119. [Google Scholar] [CrossRef]
  86. Yarandi, S.S.; Srinivasan, S. Diabetic Gastrointestinal Motility Disorders and the Role of Enteric Nervous System: Current Status and Future Directions. Neurogastroenterol. Motil. 2014, 26, 611–624. [Google Scholar] [CrossRef]
  87. Reszczyńska, M.; Kempiński, R. The Prevalence of Enteropathy Symptoms from the Lower Gastrointestinal Tract and the Evaluation of Anorectal Function in Diabetes Mellitus Patients. J. Clin. Med. 2021, 10, 415. [Google Scholar] [CrossRef]
  88. Papathanasopoulos, A.; Van Oudenhove, L.; Katsanos, K.; Christodoulou, D.; Tack, J.; Tsianos, E.V. Severity of Fecal Urgency and Incontinence in Inflammatory Bowel Disease: Clinical, Manometric and Sonographic Predictors. Inflamm. Bowel Dis. 2013, 19, 2450–2456. [Google Scholar] [CrossRef]
  89. Bassotti, G.; Antonelli, E.; Villanacci, V.; Nascimbeni, R.; Dore, M.P.; Pes, G.M.; Maconi, G. Abnormal Gut Motility in Inflammatory Bowel Disease: An Update. Tech. Coloproctology 2020, 24, 275–282. [Google Scholar] [CrossRef]
  90. Hosokawa, T.; Konuma, N.; Ikeda, T.; Hashimoto, M.; Kaneda, H.; Ohashi, K.; Matsumoto, T.; Koshinaga, T. Establishment of a New Anal Sphincter Injury Model in Rats Based on Cardiotoxin. J. Pediatr. Surg. 2015, 50, 1352–1358. [Google Scholar] [CrossRef]
  91. Saihara, R.; Komuro, H.; Urita, Y.; Hagiwara, K.; Kaneko, M. Myoblast Transplantation to Defecation Muscles in a Rat Model: A Possible Treatment Strategy for Fecal Incontinence after the Repair of Imperforate Anus. Pediatr. Surg. Int. 2009, 25, 981–986. [Google Scholar] [CrossRef]
  92. Craig, J.B.; Lane, F.L.; Nistor, G.; Motakef, S.; Pham, Q.-A.; Keirstead, H. Allogenic Myoblast Transplantation in the Rat Anal Sphincter. Female Pelvic Med. Reconstr. Surg. 2010, 16, 205–208. [Google Scholar] [CrossRef]
  93. Son, I.T.; Lee, H.S.; Ihn, M.H.; Lee, K.H.; Kim, D.-W.; Lee, K.-W.; Kim, J.-S.; Kang, S.-B. Isolation of Internal and External Sphincter Progenitor Cells from the Human Anal Sphincter with or without Radiotherapy. Colorectal Dis. Off. J. Assoc. Coloproctology G. B. Irel. 2019, 21, 38–47. [Google Scholar] [CrossRef] [PubMed]
  94. Singh, J.; Rattan, S. Bioengineered Human IAS Reconstructs with Functional and Molecular Properties Similar to Intact IAS. Am. J. Physiol. Gastrointest. Liver Physiol. 2012, 303, G713–G722. [Google Scholar] [CrossRef] [PubMed]
  95. Gilmont, R.R.; Raghavan, S.; Somara, S.; Bitar, K.N. Bioengineering of Physiologically Functional Intrinsically Innervated Human Internal Anal Sphincter Constructs. Tissue Eng. Part A 2014, 20, 1603–1611. [Google Scholar] [CrossRef] [PubMed]
  96. Raghavan, S.; Miyasaka, E.A.; Gilmont, R.R.; Somara, S.; Teitelbaum, D.H.; Bitar, K.N. Perianal Implantation of Bioengineered Human Internal Anal Sphincter Constructs Intrinsically Innervated with Human Neural Progenitor Cells. Surgery 2014, 155, 668–674. [Google Scholar] [CrossRef] [PubMed]
  97. Zakhem, E.; Rego, S.L.; Raghavan, S.; Bitar, K.N. The Appendix as a Viable Source of Neural Progenitor Cells to Functionally Innervate Bioengineered Gastrointestinal Smooth Muscle Tissues. Stem Cells Transl. Med. 2015, 4, 548–554. [Google Scholar] [CrossRef] [PubMed]
  98. Rego, S.L.; Raghavan, S.; Zakhem, E.; Bitar, K.N. Enteric Neural Differentiation in Innervated, Physiologically Functional, Smooth Muscle Constructs Is Modulated by Bone Morphogenic Protein 2 Secreted by Sphincteric Smooth Muscle Cells. J. Tissue Eng. Regen. Med. 2017, 11, 1251–1261. [Google Scholar] [CrossRef]
  99. Raghavan, S.; Miyasaka, E.A.; Hashish, M.; Somara, S.; Gilmont, R.R.; Teitelbaum, D.H.; Bitar, K.N. Successful Implantation of Physiologically Functional Bioengineered Mouse Internal Anal Sphincter. Am. J. Physiol. - Gastrointest. Liver Physiol. 2010, 299, G430–G439. [Google Scholar] [CrossRef]
  100. Hashish, M.; Raghavan, S.; Somara, S.; Gilmont, R.R.; Miyasaka, E.; Bitar, K.N.; Teitelbaum, D.H. Surgical Implantation of a Bioengineered Internal Anal Sphincter. J. Pediatr. Surg. 2010, 45, 52–58. [Google Scholar] [CrossRef]
  101. Miyasaka, E.A.; Raghavan, S.; Gilmont, R.R.; Mittal, K.; Somara, S.; Bitar, K.N.; Teitelbaum, D.H. In Vivo Growth of a Bioengineered Internal Anal Sphincter: Comparison of Growth Factors for Optimization of Growth and Survival. Pediatr. Surg. Int. 2011, 27, 137–143. [Google Scholar] [CrossRef] [PubMed]
  102. Bohl, J.L.; Zakhem, E.; Bitar, K.N. Successful Treatment of Passive Fecal Incontinence in an Animal Model Using Engineered Biosphincters: A 3-Month Follow-Up Study. Stem Cells Transl. Med. 2017, 6, 1795–1802. [Google Scholar] [CrossRef] [PubMed]
  103. Raghavan, S.; Gilmont, R.R.; Miyasaka, E.A.; Somara, S.; Srinivasan, S.; Teitelbaum, D.H.; Bitar, K.N. Successful Implantation of Bioengineered, Intrinsically Innervated, Human Internal Anal Sphincter. Gastroenterology 2011, 141, 310–319. [Google Scholar] [CrossRef] [PubMed]
  104. Tierney, M.T.; Sacco, A. Satellite Cell Heterogeneity in Skeletal Muscle Homeostasis. Trends Cell Biol. 2016, 26, 434–444. [Google Scholar] [CrossRef]
  105. Thurner, M.; Deutsch, M.; Janke, K.; Messner, F.; Kreutzer, C.; Beyl, S.; Couillard-Després, S.; Hering, S.; Troppmair, J.; Marksteiner, R. Generation of Myogenic Progenitor Cell-Derived Smooth Muscle Cells for Sphincter Regeneration. Stem Cell Res. Ther. 2020, 11, 233. [Google Scholar] [CrossRef] [PubMed]
  106. Tedesco, F.S.; Moyle, L.A.; Perdiguero, E. Muscle Interstitial Cells: A Brief Field Guide to Non-Satellite Cell Populations in Skeletal Muscle. Methods Mol. Biol. Clifton NJ 2017, 1556, 129–147. [Google Scholar] [CrossRef]
  107. Torrente, Y.; Belicchi, M.; Marchesi, C.; D’Antona, G.; Cogiamanian, F.; Pisati, F.; Gavina, M.; Giordano, R.; Tonlorenzi, R.; Fagiolari, G.; et al. Autologous Transplantation of Muscle-Derived CD133+ Stem Cells in Duchenne Muscle Patients. Cell Transplant. 2007, 16, 563–577. [Google Scholar] [CrossRef]
  108. Zhang, D.; Wang, L.; Zhang, F.; Li, C.; Zhu, T.; Cao, K.; Ma, W.; Yang, Z. Nine-Year Follow-up of Local Implantation of Autologous Skeletal Myoblasts in a Patient with Coronary Heart Disease. Am. J. Case Rep. 2013, 14, 139–142. [Google Scholar] [CrossRef]
  109. Lecoeur, C.; Swieb, S.; Zini, L.; Rivière, C.; Combrisson, H.; Ghérardi, R.; Abbou, C.; Yiou, R. Intraurethral Transfer of Satellite Cells by Myofiber Implants Results in the Formation of Innervated Myotubes Exerting Tonic Contractions. J. Urol. 2007, 178, 332–337. [Google Scholar] [CrossRef]
  110. Abbasi-Malati, Z.; Roushandeh, A.M.; Kuwahara, Y.; Roudkenar, M.H. Mesenchymal Stem Cells on Horizon: A New Arsenal of Therapeutic Agents. Stem Cell Rev. 2018, 14, 484–499. [Google Scholar] [CrossRef]
  111. Sanabria-de la Torre, R.; Quiñones-Vico, M.I.; Fernández-González, A.; Sánchez-Díaz, M.; Montero-Vílchez, T.; Sierra-Sánchez, Á.; Arias-Santiago, S. Alloreactive Immune Response Associated to Human Mesenchymal Stromal Cells Treatment: A Systematic Review. J. Clin. Med. 2021, 10, 2991. [Google Scholar] [CrossRef]
  112. Meier, R.P.H.; Müller, Y.D.; Morel, P.; Gonelle-Gispert, C.; Bühler, L.H. Transplantation of Mesenchymal Stem Cells for the Treatment of Liver Diseases, Is There Enough Evidence? Stem Cell Res. 2013, 11, 1348–1364. [Google Scholar] [CrossRef]
  113. Balaphas, A.; Meyer, J.; Sadoul, R.; Morel, P.; Gonelle-Gispert, C.; Bühler, L.H. Extracellular Vesicles: Future Diagnostic and Therapeutic Tools for Liver Disease and Regeneration. Liver Int. Off. J. Int. Assoc. Study Liver 2019. [Google Scholar] [CrossRef] [PubMed]
  114. Meier, R.P.H.; Mahou, R.; Morel, P.; Meyer, J.; Montanari, E.; Muller, Y.D.; Christofilopoulos, P.; Wandrey, C.; Gonelle-Gispert, C.; Bühler, L.H. Microencapsulated Human Mesenchymal Stem Cells Decrease Liver Fibrosis in Mice. J. Hepatol. 2015, 62, 634–641. [Google Scholar] [CrossRef]
  115. Pokrywczynska, M.; Jundzill, A.; Warda, K.; Buchholz, L.; Rasmus, M.; Adamowicz, J.; Bodnar, M.; Marszalek, A.; Helmin-Basa, A.; Michalkiewicz, J.; et al. Does the Mesenchymal Stem Cell Source Influence Smooth Muscle Regeneration in Tissue-Engineered Urinary Bladders? Cell Transplant. 2017, 26, 1780–1791. [Google Scholar] [CrossRef] [PubMed]
  116. Zhang, N.; Qin, X.; Zhang, J.; Zhang, Z.; Li, Y.; Xie, Y.; Kong, D.; Du, R.; Huang, X.; Xu, Y. Bone Marrow Mesenchymal Stem Cells Accelerate the Morphological and Functional Recovery of Neovaginas. Artif. Organs 2018, 42, 1206–1215. [Google Scholar] [CrossRef] [PubMed]
  117. Nakamura, Y.; Miyaki, S.; Ishitobi, H.; Matsuyama, S.; Nakasa, T.; Kamei, N.; Akimoto, T.; Higashi, Y.; Ochi, M. Mesenchymal-Stem-Cell-Derived Exosomes Accelerate Skeletal Muscle Regeneration. FEBS Lett. 2015, 589, 1257–1265. [Google Scholar] [CrossRef]
  118. Linard, C.; Brachet, M.; L’homme, B.; Strup-Perrot, C.; Busson, E.; Bonneau, M.; Lataillade, J.-J.; Bey, E.; Benderitter, M. Long-Term Effectiveness of Local BM-MSCs for Skeletal Muscle Regeneration: A Proof of Concept Obtained on a Pig Model of Severe Radiation Burn. Stem Cell Res. Ther. 2018, 9, 299. [Google Scholar] [CrossRef]
  119. Hecker, L.; Baar, K.; Dennis, R.G.; Bitar, K.N. Development of a Three-Dimensional Physiological Model of the Internal Anal Sphincter Bioengineered in Vitro from Isolated Smooth Muscle Cells. Am. J. Physiol. Gastrointest. Liver Physiol. 2005, 289, G188–G196. [Google Scholar] [CrossRef]
  120. Dominici, M.; Le Blanc, K.; Mueller, I.; Slaper-Cortenbach, I.; Marini, F.; Krause, D.; Deans, R.; Keating, A.; Prockop, D.; Horwitz, E. Minimal Criteria for Defining Multipotent Mesenchymal Stromal Cells. The International Society for Cellular Therapy Position Statement. Cytotherapy 2006, 8, 315–317. [Google Scholar] [CrossRef]
  121. Gräs, S.; Tolstrup, C.K.; Lose, G. Regenerative Medicine Provides Alternative Strategies for the Treatment of Anal Incontinence. Int. Urogynecol. J. 2017, 28, 341–350. [Google Scholar] [CrossRef]
  122. Baldari, S.; Di Rocco, G.; Piccoli, M.; Pozzobon, M.; Muraca, M.; Toietta, G. Challenges and Strategies for Improving the Regenerative Effects of Mesenchymal Stromal Cell-Based Therapies. Int. J. Mol. Sci. 2017, 18, 2087. [Google Scholar] [CrossRef] [PubMed]
  123. Balaphas, A.; Schiltz, B.; Liot, E.; Robert-Yap, J.; Ris, F. What Is the Role of Stem Cell Therapy in the Treatment of Anal Incontinence? Colorectal Dis. Off. J. Assoc. Coloproctology G. B. Irel. 2021, 23, 551–552. [Google Scholar] [CrossRef] [PubMed]
  124. Bols, E.M.J.; Hendriks, H.J.M.; Berghmans, L.C.M.; Baeten, C.G.M.I.; de Bie, R.A. Responsiveness and Interpretability of Incontinence Severity Scores and FIQL in Patients with Fecal Incontinence: A Secondary Analysis from a Randomized Controlled Trial. Int. Urogynecol. J. 2013, 24, 469–478. [Google Scholar] [CrossRef]
  125. Voorham-van der Zalm, P.J.; Voorham, J.C.; van den Bos, T.W.L.; Ouwerkerk, T.J.; Putter, H.; Wasser, M.N.J.M.; Webb, A.; DeRuiter, M.C.; Pelger, R.C.M. Reliability and Differentiation of Pelvic Floor Muscle Electromyography Measurements in Healthy Volunteers Using a New Device: The Multiple Array Probe Leiden (MAPLe). Neurourol. Urodyn. 2013, 32, 341–348. [Google Scholar] [CrossRef] [PubMed]
  126. Johnson, E.; Carlsen, E.; Steen, T.B.; Backer Hjorthaug, J.O.; Eriksen, M.T.; Johannessen, H.-O. Short- and Long-Term Results of Secondary Anterior Sphincteroplasty in 33 Patients with Obstetric Injury. Acta Obstet. Gynecol. Scand. 2010, 89, 1466–1472. [Google Scholar] [CrossRef] [PubMed]
  127. Lamblin, G.; Bouvier, P.; Damon, H.; Chabert, P.; Moret, S.; Chene, G.; Mellier, G. Long-Term Outcome after Overlapping Anterior Anal Sphincter Repair for Fecal Incontinence. Int. J. Colorectal Dis. 2014, 29, 1377–1383. [Google Scholar] [CrossRef] [PubMed]
  128. Garcia, S.; Bernad, A.; Martín, M.C.; Cigudosa, J.C.; Garcia-Castro, J.; de la Fuente, R. Pitfalls in Spontaneous in Vitro Transformation of Human Mesenchymal Stem Cells. Exp. Cell Res. 2010, 316, 1648–1650. [Google Scholar] [CrossRef]
  129. Torsvik, A.; Røsland, G.V.; Svendsen, A.; Molven, A.; Immervoll, H.; McCormack, E.; Lønning, P.E.; Primon, M.; Sobala, E.; Tonn, J.-C.; et al. Spontaneous Malignant Transformation of Human Mesenchymal Stem Cells Reflects Cross-Contamination: Putting the Research Field on Track - Letter. Cancer Res. 2010, 70, 6393–6396. [Google Scholar] [CrossRef] [PubMed]
  130. Trébol, J.; Carabias-Orgaz, A.; García-Arranz, M.; García-Olmo, D. Stem Cell Therapy for Faecal Incontinence: Current State and Future Perspectives. World J. Stem Cells 2018, 10, 82–105. [Google Scholar] [CrossRef]
  131. Lipsitz, Y.Y.; Milligan, W.D.; Fitzpatrick, I.; Stalmeijer, E.; Farid, S.S.; Tan, K.Y.; Smith, D.; Perry, R.; Carmen, J.; Chen, A.; et al. A Roadmap for Cost-of-Goods Planning to Guide Economic Production of Cell Therapy Products. Cytotherapy 2017, 19, 1383–1391. [Google Scholar] [CrossRef] [PubMed]
  132. De la Portilla, F. Reply to Balaphas et Al. Colorectal Dis. Off. J. Assoc. Coloproctology G. B. Irel. 2021, 23, 1002–1003. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic representation of events occurring after delivery-related acute anal sphincters injury [51,53]. (A): The tear will induce a phase of inflammation where polymorphonuclear cells are attracted in the first hour and are responsible to maintain the function of macrophages that will arrive later. At the same time, platelets recruitment following the rupture of small vessels induces the releasee of SDF-1 following their degranulation. SDF-1 is primordial for anal sphincter regeneration and acts by attracting non-resident progenitor cells, myoblasts and smooth-muscle progenitors to the wound and by promoting the survival, migration and proliferation of these cells. (B): Macrophages of M1 (CD68+) polarity arrives after the polymorphonuclear cells and largely contribute to the acute inflammation phase. (C): During the healing phase, activated satellite cells contribute to new muscle formation (black arrows) and each step is characterized by a specific hallmark protein expression (blue boxes). Then, macrophage polarity switches to M2 (CD163+) and they produce cytokines such as Il-10 helping proliferation and differentiation of satellite cells. (D): During the healing phase, different kinds of non-myogenic local progenitor cells are activated, and some of them, like the fibro-adipogenic progenitors, are conditioned by the presence of IL-4 secreted by eosinophils, with this pathway being necessary for skeletal muscle regeneration. Non-myogenic local progenitor cells interplay indirectly by the production of cytokines like FGF-2 and IGF-1 but can also fuse (camel arrows) with other non-myogenic local progenitor cells, satellite cells, myoblasts or even myofibers, inducing hypertrophy. Finally, in case of damage to the rectal wall mucosa, local stem cells of the anal transition zone contribute to the reparation of the rectum and anal transition zone mucosa. qSC: Quiescent satellite cell, aSC: Activated satellite cell, SMC: Smooth muscle cell, pSMC: Progenitor smooth muscle cell. ATZ: Anal transition zone.
Figure 1. Schematic representation of events occurring after delivery-related acute anal sphincters injury [51,53]. (A): The tear will induce a phase of inflammation where polymorphonuclear cells are attracted in the first hour and are responsible to maintain the function of macrophages that will arrive later. At the same time, platelets recruitment following the rupture of small vessels induces the releasee of SDF-1 following their degranulation. SDF-1 is primordial for anal sphincter regeneration and acts by attracting non-resident progenitor cells, myoblasts and smooth-muscle progenitors to the wound and by promoting the survival, migration and proliferation of these cells. (B): Macrophages of M1 (CD68+) polarity arrives after the polymorphonuclear cells and largely contribute to the acute inflammation phase. (C): During the healing phase, activated satellite cells contribute to new muscle formation (black arrows) and each step is characterized by a specific hallmark protein expression (blue boxes). Then, macrophage polarity switches to M2 (CD163+) and they produce cytokines such as Il-10 helping proliferation and differentiation of satellite cells. (D): During the healing phase, different kinds of non-myogenic local progenitor cells are activated, and some of them, like the fibro-adipogenic progenitors, are conditioned by the presence of IL-4 secreted by eosinophils, with this pathway being necessary for skeletal muscle regeneration. Non-myogenic local progenitor cells interplay indirectly by the production of cytokines like FGF-2 and IGF-1 but can also fuse (camel arrows) with other non-myogenic local progenitor cells, satellite cells, myoblasts or even myofibers, inducing hypertrophy. Finally, in case of damage to the rectal wall mucosa, local stem cells of the anal transition zone contribute to the reparation of the rectum and anal transition zone mucosa. qSC: Quiescent satellite cell, aSC: Activated satellite cell, SMC: Smooth muscle cell, pSMC: Progenitor smooth muscle cell. ATZ: Anal transition zone.
Cells 10 02086 g001
Figure 2. Cell therapy-based strategies for anal sphincter incontinence treatment. (A,B) Multipotent cells can be transplanted directly after injury with or without simultaneous reparation. (C) After injury, healing progresses but sometimes the muscle tissue is altered and responsible for ASI. Multipotent cells can be transplanted directly into the scar or in the neighborhood. (D) The best scenario could be to reinforce the surgical reparation of a sphincter defect with multipotent cells.
Figure 2. Cell therapy-based strategies for anal sphincter incontinence treatment. (A,B) Multipotent cells can be transplanted directly after injury with or without simultaneous reparation. (C) After injury, healing progresses but sometimes the muscle tissue is altered and responsible for ASI. Multipotent cells can be transplanted directly into the scar or in the neighborhood. (D) The best scenario could be to reinforce the surgical reparation of a sphincter defect with multipotent cells.
Cells 10 02086 g002
Table 1. Models for anal sphincter incontinence. NHP: non-human primate.
Table 1. Models for anal sphincter incontinence. NHP: non-human primate.
Designation of ModelTime Lapse between Injury and InterventionPublication Reporting the ModelSpeciesProcedureSphincter
Acute Anal Sphincter InjurySphincterotomy and repair0Mazzanti et al., 2016 [18], Lorenzi et al., 2008 [19] RatSphincterotomy and primary repair of sphinctersIAS and EAS
Repaired sphincterotomy0Fitzwater et al., 2015 [20], White et al 2010 [21], Pathi et al., 2012 [22]RatFull thickness 7 mm incision of sphincters followed by repair IAS and EAS
Anal sphincter injury0Kuismanen et al., 2018 [23]RatIncision of full thickness sphincter with mucosa followed by mucosa and IAS repairIAS and EAS
Proctoepisiotomy0Lane et al., 2013 [24], Jacobs et al., 2013 [25]RatProctoepisiotomy with repairEAS
Sphincterotomy0Inoue et al., 2018 [26]RatRemoval of a left semicircle of sphincterIAS and EAS
Extra-mucosal myotomy0Trébol et al., 2018 [27]Rat1 cm-long incision preserving the mucosaIAS and EAS
Anal sphincter cryoinjury0Bisson et al., 2013 [28] RatTwo cryoinjuries of sphincters at 24 h interval with liquid nitrogen on a 90° sectorIAS and EAS
Anal sphincter cryoinjury0Kang et al., 2008 [29]RatCryoinjury of right hemi-sphincters IAS and EAS
Sphincterotomy0Sarveazad et al., 2019 [30]RabbitLeft lateral sphincterotomyIAS and EAS
Sphincterotomy24 hSalcedo et al., 2013 [15]Rat2–3 mm-thick transection of sphinctersIAS and EAS
Pudendal nerve crush24 hSalcedo et al., 2013 [15]RatPosterior incision of sacro-coccygeal area and 30 s crushing of the nerves on both sidesna
Unrepaired Anal Sphincter InjuryPartial anal sphincter excision24 h and 3 weeksSalcedo et al., 2014 [31], Li et al., 2020 [32]RatExcision of 1/3 of ventral anal sphinctersIAS and EAS
Anal sphincter injury2 weeksDing et al.,2016 [33]Rat0.2 cm-long sphincters incisionIAS and EAS
Unrepaired
sphincterotomy
2 weeksMontoya et al., 2015 [34]RatFull thickness 7 mm incision of sphincters IAS and EAS
Chronic large anal sphincter defect3 weeksSun et al., 2017 [35], Sun et al., 2017 [36], Sun et al., 2016 [37]Rat50% excision of ventral portion of anal sphinctersIAS and EAS
Anal sphincter damage ndLi et al., 2018 [38]Rat3 mm-long incision in the right posterolateral sphincterIAS and EAS
Intersphincteric resection modelnaYamaguchi et al., 2013 [39]Rat50% excision of IAS and a part of EASIAS and EAS
Sphincterotomy2 weeksAghaee-Afshar et al., 2009 [40]RabbitRight lateral sphincterotomyEAS
Excision of external anal sphincter3 to 24 weeksKajbafzadeh et al., 2016 [41], Elmi et al., 2014 [42], Kajbafzadeh et al., 2010 [43]RabbitSubtotal to total excision of posterior sphincter EAS
Sphincter injury4 weeksOh et al., 2015 [44], Oh et al., 2015 [45], Kang et al., 2013 [46]DogResection of 25% of posterior anal sphinctersIAS and EAS
Internal sphincter hemi-sphincterectomy6–8 weeksBohl et al., 2017 [47], Dadhich et al., 2019 [48]Rabbit, NHP50% excision of ventral portion of anal sphincterIAS
Table 2. List of clinical studies on cell therapy for anal incontinence treatments. bn: billion, mio: million, FIQL: Fecal Incontinence Quality of Life.
Table 2. List of clinical studies on cell therapy for anal incontinence treatments. bn: billion, mio: million, FIQL: Fecal Incontinence Quality of Life.
PublicationDesignWomenMenConcerned SphincterCellular TherapyPassages nNumber of Injected CellOutcomesFollow-Up
De la Portilla et al. 2020 [72]Randomized controlled trial62EASAutologous adipose tissue derived multipotent cells injected into EAS lesionUp to 74 bnWexner incontinence score, FIQL, safety and feasibility, manometry 12 months
Frudinger et al. 2018 [68]Explorative baseline controlled study345EASAnal electrical stimulation followed by 12 injections of autologous myoblasts from pectoralis muscle into the entire EAS.325 mioWexner incontinence score and FIQL score, anorectal manometry12 months
Boyer et al. 2018 [69] Randomized controlled trial120EASAutologous myoblasts derived from quadriceps injected at 8 points into the EAS under endoanal ultrasonography guindance30.2 to 2 bnWexner incontinence score12 months
Sarvezaad et al. 2017 [71]Randomized controlled trial72EASAllogenic hADSC injection along with external sphincteroplasty56 mioEndoanal ultrasonography and EMG2 months
Romaniszyn et al. 2015 [67]Pilot study101EASAutologous myoblasts injected into EAS under ultrasound guidance7–10 passages100 mioAnorectal manometry, EMG, endoanal ultrasonography12 months
Frudinger et al. 2010/2015 [73,66] Pilot study100EASAnal electrical stimulation followed by 12–14 injections of autologous myoblasts from pectoralis muscle into the defect and the edges of EAS-20.16 mioWexner incontinence score, anorectal manometry (contractile tone), endoanal ultrasonography60 months
Romaniszyn et al. 2013 [74]Case report01IAS and EASInjection of autologous myoblast from quadriceps into the scar tissue of the EAS.6–7600 mioAnorectal manometry, EMG12 months
Table 3. Characteristics of primary cells proposed for cell therapy of anal sphincter incontinence. MHC: myosin heavy chain, Sma: α-smooth-muscle-actin, and MyoG: moygenin.
Table 3. Characteristics of primary cells proposed for cell therapy of anal sphincter incontinence. MHC: myosin heavy chain, Sma: α-smooth-muscle-actin, and MyoG: moygenin.
PublicationCells OriginSpeciesSurface Antigens ExpressedSurface Antigens Not ExpressedGENE ExpressionIntracellular Protein ExpressedIntracellular Protein Not ExpressedDifferentiation Test
Muscle-Derived CellsBisson et al., 2015 [28]Skeletal muscleRatCD56-DES, MYOD1, MYF5,---
Lane et al., 2013 [24], Jacobs et al., 2013 [25], Craig et al., 2010 [92] Skeletal muscleRat------
Saihara et al., 2009 [91] Skeletal muscleRat-----Myotubes
Kang et al., 2008 [29] Skeletal muscleRatCD34CD45-Desmin--
Kajbafzadeh et al., 2016 [41]Skeletal muscleRabbit---Pax7, Desmin-Myotubes
Elmi et al., 2014 [42] Skeletal muscleRabbit---Desmin, MyoD-
Oh et al., 2015 [44], Oh et al., 2015 [45] Skeletal muscleDog---Pax7, SmaMHC, MyoGMyotubes
Kang et al., 2013 [46] Skeletal muscleDog---Pax7MHCα-SMA
Boyer et al., 2018 [69]Skeletal muscleHumanCD90, HLA-ICD34, CD45, CD133DES, MYOD1, MYF5, PAX7---
Frudinger et al., 2015 [66], Frudinger et al., 2018 [68] Skeletal muscleHumanSSEA3, SSEA4, CD56, CD90-NANOG1, NACAM1, MYOD1, PAX7, PAX3, MYF5, DES, MYOGDesmin, UTF1, Pax7, Myf5-Myotubes
Romaniszyn et al., 2015 [67]Skeletal muscleHumanCD56-DES, MYOD1, MYOG--Myocyte
Romaniszyn et al., 2013 [74]Skeletal muscleHuman------
Son et al., 2019 [93]EAS HumanCD34, NG2--Pax7-MyoG, MyHC
Bohl et al., 2017 [102], Rego et al., 2017 [98] Smooth muscle Rabbit
Raghavan et al., 2010 [99], Hashish et al., 2010 [100], Miyasaka et al., 2011 [101]IASMouse------
Zakhem et al., 2015 [97], Rego et al. 2017 [98] IASRabbit------
Dadhich et al., 2019 [48]IASNHP--SMTN-Sma, and smoothelin-
Gilmont et al., 2014 [95] IASHuman------
Singh and Rattan 2012 [94] IASHuman------
Raghavan et al., 2014 [96], Somara et al., 2009 [79]IASHuman------
Bone Marrow-Derived CellsLi et al., 2018 [38] Bone marrow Rat-CD34, CD45----
Ding et al., 2016 [33]Bone marrow, transfected with galectin-1RatCD90CD45----
Sun et al., 2017 [36]Bone marrow Rat------
Mazzanti et al., 2016 [18], Lorenzi et al., 2008 [19]Bone marrow RatCD44, CD54, CD73, CD90, CD106CD11b, CD11c, CD45---Osteogenic and adipogenic
Salcedo et al., 2014 [31], Salcedo et al., 2013 [15] Bone marrow Rat-CD34, CD45----
Pathi et al., 2012 [22]Bone marrow Rat------
Aghaee-Afshar et al., 2009 [40]Bone marrow Rabbit------
Adipose Tissue-Derived CellsTrébol et al., 2018 [27]Adipose tissueRatCD29, CD90CD11n, CD45----
Inoue et al., 2018 [26]Adipose tissueRatCD90CD31, CD45---Adipogenic and myogenic
Sarveazad et al., 2019 [30]Adipose tissueHumanCD29, CD73, CD105CD34, CD45
Sarveazad et al., 2017 [71]Adipose tissue HumanCD44, CD73, CD90CD31, CD45----
Kuismanen et al., 2018 [23] Adipose tissue HumanCD73, CD90, CD105CD14, CD19, CD34, CD45RO, CD54, HLA-DR----
Neural Tissue-Derived CellsBohl et al., 2017 [102]Enteric Neural SystemRabbit------
Zakhem et al., 2015 [97]Appendix neuronal systemRabbitP75(NTR)--Sox2, Nestin Neurospheres
Rego et al., 2017 [98]Enteric neuronal systemRabbit-----Neurospheres
Dadhich et al., 2019 [48]Enteric neuronal systemNHPP75(NTR) smoothelin, oct4
Gilmont et al., 2014 [95]Enteric neuronal systemHumanP75(NTR)----Neurospheres
Raghavan et al., 2014 [96], Raghavan et al., 2011 [103]Enteric neuronal systemHuman------
MiscellaneousAghaee-Afshar et al., 2009 [40]Umbilical cord matrix Human------
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Balaphas, A.; Meyer, J.; Meier, R.P.H.; Liot, E.; Buchs, N.C.; Roche, B.; Toso, C.; Bühler, L.H.; Gonelle-Gispert, C.; Ris, F. Cell Therapy for Anal Sphincter Incontinence: Where Do We Stand? Cells 2021, 10, 2086. https://doi.org/10.3390/cells10082086

AMA Style

Balaphas A, Meyer J, Meier RPH, Liot E, Buchs NC, Roche B, Toso C, Bühler LH, Gonelle-Gispert C, Ris F. Cell Therapy for Anal Sphincter Incontinence: Where Do We Stand? Cells. 2021; 10(8):2086. https://doi.org/10.3390/cells10082086

Chicago/Turabian Style

Balaphas, Alexandre, Jeremy Meyer, Raphael P. H. Meier, Emilie Liot, Nicolas C. Buchs, Bruno Roche, Christian Toso, Leo H. Bühler, Carmen Gonelle-Gispert, and Frédéric Ris. 2021. "Cell Therapy for Anal Sphincter Incontinence: Where Do We Stand?" Cells 10, no. 8: 2086. https://doi.org/10.3390/cells10082086

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop