Next Article in Journal
Ultra-Low Intensity Post-Pulse Affects Cellular Responses Caused by Nanosecond Pulsed Electric Fields
Previous Article in Journal
Milk Casein Inhibits Effect of Black Tea Galloylated Theaflavins to Inactivate SARS-CoV-2 In Vitro
Previous Article in Special Issue
Model-Based Characterization of E. coli Strains with Impaired Glucose Uptake
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Bioprocessing Considerations towards the Manufacturing of Therapeutic Skeletal and Smooth Muscle Cells

by
Teresa Franchi-Mendes
1,2,†,
Marília Silva
1,2,†,
Ana Luísa Cartaxo
1,2,
Ana Fernandes-Platzgummer
1,2,
Joaquim M. S. Cabral
1,2 and
Cláudia L. da Silva
1,2,*
1
Department of Bioengineering, iBB—Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
2
Associate Laboratory, i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Bioengineering 2023, 10(9), 1067; https://doi.org/10.3390/bioengineering10091067
Submission received: 30 June 2023 / Revised: 31 August 2023 / Accepted: 4 September 2023 / Published: 9 September 2023
(This article belongs to the Special Issue Design, Optimization and Scale-Up of Industrial Bioprocess)

Abstract

:
Tissue engineering approaches within the muscle context represent a promising emerging field to address the current therapeutic challenges related with multiple pathological conditions affecting the muscle compartments, either skeletal muscle or smooth muscle, responsible for involuntary and voluntary contraction, respectively. In this review, several features and parameters involved in the bioprocessing of muscle cells are addressed. The cell isolation process is depicted, depending on the type of tissue (smooth or skeletal muscle), followed by the description of the challenges involving the use of adult donor tissue and the strategies to overcome the hurdles of reaching relevant cell numbers towards a clinical application. Specifically, the use of stem/progenitor cells is highlighted as a source for smooth and skeletal muscle cells towards the development of a cellular product able to maintain the target cell’s identity and functionality. Moreover, taking into account the need for a robust and cost-effective bioprocess for cell manufacturing, the combination of muscle cells with biomaterials and the need for scale-up envisioning clinical applications are also approached.

Graphical Abstract

1. Introduction

Even though a considerable progress has been made in the cell therapy field for muscular disorders, there is still significant clinical demand for tissue-engineered muscle for transplantation or replacement therapy [1]. Indeed, tissue engineering approaches hold a promising future in treating diseases that affect skeletal muscle (SkM) and smooth muscle (SM), including cases of muscular dystrophies and volumetric muscle loss (VML) after cancer or trauma [2]. Moreover, muscle tissues that rely on contractile activity, such as the sphincters, bladder, intestine, diaphragm, face, hand, tongue, pharynx, larynx and esophagus, could potentially also be restored, or replaced using tissue engineering approaches [3]. Epidemiology of disorders related to SkM and SM are difficult to truly ascertain, as they can be multisystemic. For instance, reports on Duchenne muscular dystrophy (DMD), a SkM genetic disease, point to 3–7 cases per 100.000 population [4]. On the other hand, reports on SM-related disorders, such as stress urinary incontinence, suggest a prevalence of 30–40% in women [5,6], but these numbers may be underestimated due to the social stigma associated with them.
Tissue engineering approaches can include the use of stem/progenitor cells, combined with appropriate biomaterials, to generate the suitable microenvironment to functionally repair, replace and regenerate the damaged or lost organ [7,8]. From this perspective, to provide clinically relevant engineered muscle tissues, there is a demand for an in-depth optimization of the manufacturing of their functional building blocks, either skeletal muscle cells (SkMCs) and/or smooth muscle cells (SMCs). Importantly, a critical point in what concerns the engineering of these tissues is the need to generate functional cells with a contractile phenotype [9].
SM is derived from both mesoderm and neural crest cells, and it can have a local common progenitor origin in adult tissue (for example, vascular progenitors) [10]. SM tissue is located throughout the body and is crucial, from a functional standpoint, in a variety of tissues [11]. For instance, in the gastrointestinal tract, SM is essential for motility; therefore, any damage to the SM of the gastrointestinal system may have severe effects on digestion and nutrient absorption. In the urinary system, at the level of the kidneys, vascular SM dysfunction is associated with chronic kidney disease and can lead to end-stage renal disease [12]. In the cardiovascular system, SM is present in vessels to maintain blood pressure and flow, whereas in the respiratory tract, it is responsible for opening and closing airways. Overall, SM serves a purpose in almost every other organ system [11]. At a cellular level, SM is described as a nonstriated muscle, with neural innervations from the autonomic nervous system, and it differs from SkM in many ways, possibly the most functionally significant being its ability to be contracted and controlled involuntarily [11]. SMCs are usually characterized by identification of multiple markers namely smooth muscle actin (α-SMA), smoothelin, calponin, smooth muscle 22 (SM22α), and smooth muscle myosin heavy chain (MYH11) [10,13]. These proteins can also be transiently detected in other cell types, such as α-SMA in activated fibroblasts or myofibroblasts [10]. Therefore, in vitro manufacturing of SMCs needs to take into account the multitude of markers for SMC identity, as well as the ability for contractility.
When it comes to SkM, it represents about 40% of the human body mass and it is composed of bundles of voluntary contractile multinucleated muscle fibers, resulting from the fusion of myoblasts [2]. SkM is one of the adult tissues that still holds a remarkable ability to regenerate itself in response to injury (as well as exercise), despite the post-mitotic nature of its myofibers, due to the presence of a primitive cell population, defined as satellite cells [14]. This population is recognized as SkM-resident stem cells, located between the plasma membrane of myofibers and the basal lamina, providing a homeostatic microenvironment for tissue regeneration [14]. However, in cases of severe injury consisting of VML, the damage cannot be repaired naturally, affecting the patients’ quality of life by seriously limiting musculoskeletal functionality [15]. Causes of SkM damage are traumatic injuries; tumor resections and degenerative genetic diseases, namely DMD; amyotrophic lateral sclerosis (ALS); and pediatric Charcot–Marie–Tooth disease [15]. Mitotically quiescent satellite cells are activated once a disruption in the myofiber occurs as a response to muscle lesion [16,17]. Once activated, satellite cells undergo asymmetric division [18,19]. This maintains a satellite cell pool and generates myoblasts, which in turn proliferate and differentiate, giving rise to multinucleated myotubes [19,20]. During the myogenic process, there is a tight temporal regulation by specific transcription factors [21]. Particularly, satellite cells express paired box transcription factor 7 (Pax7), while myogenic differentiation factor 1 (MyoD) is expressed during myoblast proliferation, and myogenin is highly expressed once the differentiation into myocytes occurs [17,21]. Muscle-specific proteins, such as Desmin, a type III intermediate filament; sarcomeric α-actinin; and contractile proteins myosin heavy-chain 1 and 2, are typical identifiers of the terminal differentiation stage for SkMCs [17].
Although the myogenic process has defined hallmarks, it involves complex temporal dynamics and a mix of cell populations [22]. Firstly, satellite cells are considered a heterogeneous population [23] and consequences on their functionality are not yet fully understood. For instance, Pax7 transcription factor is the classic marker for satellite cells, but a population of Pax7-negative human muscle-derived cells able to regenerate muscle after transplantation in mouse models of muscle damage was identified [24]. On the other hand, the muscle milieu comprises not only myogenic cells, but also nonmyogenic players that have an essential role in homeostasis and during muscle lesion and repair [25]. Upon muscle injury, for example, immune cell infiltration (macrophages and neutrophils) occurs, creating firstly a proinflammatory environment that sustains satellite cell proliferation, followed by a balance of anti-inflammatory factors, which in turn favor a microenvironment towards differentiation [22,26,27]. Moreover, fibroadipogenic progenitors have been identified within the muscle milieu and cause crosstalk during muscle injury [28]. In this situation, they support myoblast differentiation and can also differentiate into myofibroblasts, which secrete the extracellular matrix (ECM) that surrounds the new myofibers [28]. Fibroadipogenic progenitors can also interact with immune cells, with a reciprocal regulation [28]. Furthermore, a population of denominated muscle-derived stem cells (MDSCs) has been reported as co-expressing myogenic and endothelial cells (ECs) markers, exhibiting myogenic differentiation potential in vitro and in vivo [29,30]. However, it is not clear if they represent an intermediate state during myogenesis or if they refer to the same population, only varying due to differences in isolation method [17].
As highlighted above, tissue engineering approaches for SM and SkM regeneration are of utmost clinical significance due to the broad functional relevance of these tissues throughout the body. As such, it is imperative to explore bioengineering strategies towards the development of advanced regenerative therapies to restore muscle structure and function.

2. Isolation and Ex Vivo Expansion of Smooth and Skeletal Muscle Cells

2.1. Smooth Muscle Cells (SMCs)

SMCs are an essential cell type found in several organs, including the respiratory tract, gastrointestinal tract, urinary bladder, uterus, male and female reproductive tracts and the vascular system [31]. Methodologies for SMC isolation described in the literature are based on explant and enzymatic digestion techniques (as depicted in Figure 1), mainly from human, porcine and rodent bladder tissues [32]. In the explant method, cells are allowed to adhere and migrate from the explant onto the culture surface followed by proliferation. The second method involves enzymatic digestion of the tissue sample followed by plating the dispersed cells onto a surface for adherent cell culture [32]. Contamination of primary cultures of SMCs with fibroblasts constitutes a major drawback due to their potential to outgrow the target cells. Both SMC isolation protocols were compared in terms of robustness and efficiency envisaging tissue engineering applications. In a study by Pokrywczynska et al., the most homogenous culture (98% purity) was obtained when porcine SMCs from bladder tissue were isolated with collagenase and dispase digestion [32]. Moreover, the enzymatic methods utilizing collagenase and dispase, and collagenase alone, enabled the isolation of a significantly higher number of viable cells compared to explant techniques [32]. Studies have been published outlining the procedures for isolating vascular SMCs, employing both explant and enzymatic digestion methods [33]. Successful culture of vascular SMCs using enzymatic digestion relies on critical factors such as the specific enzymatic composition and the duration of digestion. These factors may vary depending on the source of vascular tissue and the specific vascular bed under investigation, as the type and amount of connective tissue can differ across samples. The enzymatic cocktail described by Ray et al. is advantageous, as it allows for the rapid and reproducible isolation of vascular SMCs from murine aorta where large amounts of starting tissue might not be readily available [34]. Other studies also described the use of enzymatic cocktails based on collagenase only or collagenase and elastase for isolating vascular SMCs from rat model tissues [35,36]. Moreover, magnetic forces were employed to facilitate tissue digestion and, importantly, SMC phenotype was validated through the identification of multiple markers (α-SMA, smoothelin, calponin, SM22α and MYH11) and functional assays [35]. In contrast, using explants, McMurray et al. reported a standardized method for culturing aortic explants to study factors affecting phenotypic modulation of cells in culture [36]. These authors suggested that explant cultures provided a system for studying the growth of vascular SMCs without fully digesting the tissue, thus circumventing the variability issues associated with enzymatic digestion [37].
Overall, the challenges associated with SMC isolation methods rely on the type of protocol (enzymatic- or explant-based), which can impact the cell number obtained. Additionally, this also depends on the amount and quality of the tissue source (vascular, nonvascular). Therefore, alternative approaches comprising the use of induced pluripotent stem cells (iPSCs), as well as mesenchymal stromal cells (MSCs), are also being explored to achieve clinically significant cell numbers, which is covered in Section 3.1. Importantly, regardless of the cell isolation procedure and tissue source, SMC identity and functionality need to be carefully addressed throughout the culture process.

2.2. Skeletal Muscle Cells (SkMCs)

Cell isolation from different anatomic SkM groups has been attempted, including deltoid, triceps, quadriceps, sternocleidomastoid or vastus lateralis. Satellite cells represent about 3–5% of the nucleated cells in adult human muscle, and this number tends to decrease with aging [38]. Muscle progenitors (i.e., myoblasts) have been isolated through tissue mincing and enzymatic digestion using collagenase and dispase [39,40], as schematically depicted in Figure 1. This method displays limited efficiency not only due to the low content of primitive cells in adult muscles, but also due to the need to pass the resultant suspension through cell strainers to remove nondigested tissue [40]. Furthermore, as it occurs with SMCs isolation, fibroblast contamination can occur along culture time. Besides enzymatic digestion, explant or single myofiber isolation methods have also been attempted for several years, although these display a high inefficiency for the isolation of SkMCs (and subsequent ex vivo expansion) and have been mainly purposed towards pathophysiology models, namely to study muscle electrophysiology [41,42]. Nonetheless, isolated muscle progenitors can usually be plated onto collagen- [43,44] or Matrigel- (a basement membrane extract from mouse sarcoma) coated surfaces [45].
In terms of absolute cell numbers retrieved after isolation based on enzymatic protocols, approximately 1–2 × 105 myogenic progenitors could be collected from a single murine muscle [46], while combined protocols involving tissue digestion and explant outgrowth onto Matrigel have resulted in 1–2 × 107 myoblasts [40]. Moreover, in terms of culture medium conditions, it has been observed that isolated satellite cells of murine origin seem to retain their quiescent state when cultured under low serum conditions (2%) and proliferate when using high serum-content medium (20%) [47], which can constitute an artificial stimulus of muscle injury. These primitive cells can proliferate in vitro and preserve levels of myogenic differentiation potential, with identification of myotube formation capacity [45,47].
Cell enrichment using fluorescence/magnetic-activated cell sorting (FACS/MACS) has been described, for example, in murine models, through which muscle stem cells have been purified using VCAM+CD31CD45Sca1 sorting [48]. Sacco et al. reported the isolation of murine satellite cells from tibialis anterior muscle using enzymatic digestion and FACS enrichment based on a combination of markers: lack of expression of CD45, CD11b, CD31, Sca1 and positivity for CD34/integrin-α7 [23]. This enriched cell fraction has shown potential for transplantation, as cells proliferated and integrated into myofibers in recipient mouse muscle upon injury, as verified by tracking of Pax7+ mononucleated cells using luciferase and bioluminescence techniques [23]. In another study, a myosphere culture of human SkM-derived stem cells was established, with cells isolated through enzymatic digestion (from omohyoid muscle) without the need for Matrigel [49]. Despite the advanced age of the muscle biopsy donors (60 years old), the cells exhibited proliferative capacity over several passages and were able to differentiate into multinucleated myofibers [49]. In the comprehensive study conducted by Garcia et al., successful isolation of satellite cells from different human muscle biopsies (gastrocnemius, latissimus, vastus lateralis, rectus abdominis, among others) was reported [50]. Although there was a variable degree on the cell yield obtained per muscle, the authors typically achieved 104 highly purified satellite cells per 1 g of adult SkM. It is worth noting that the number of cells per gram presented a decreasing trend towards donors aged over 80 years old, which is in line with other reports of satellite cell deregulation during aging [51]. The complex isolation protocol established by Garcia et al. involved enzymatic digestion, MACS and FACS. Briefly, a negative selection using magnetic beads was performed to remove CD31 and CD45 positive cells, followed by flow cytometry purification of the CD31/CD34/CD45/CXCR4+/CD29+/CD56+ population. Interestingly, the therapeutic potential of these isolated cells was assessed by injection into immunocompromised mice with muscle injury and mouse models of DMD, being observed that human-derived Pax7 cells generated myofibers in the damaged muscles. Importantly, the isolated human cells retained their skeletal phenotype even after cryopreservation and thawing [50]. By using the explants technique, followed by MACS enrichment for CD56+ cells and subsequent culture in Matrigel and collagen constructs, it was observed that the CD56+-enriched fraction generated more myotubes compared to the unsorted counterparts [52]. The CD56-negative fraction mainly consisted of interstitial fibroblasts, along with smaller percentages of MSCs, immune cells, fibroadipogenic progenitors and ECs. The authors also tested an in vitro chemical muscle injury, which resulted in decreased hydrogel deformation as a measure of myofiber functionality [52]. This observation was then followed by proliferation of Pax7+ and MyoD+ cells, which identify satellite cells and committed progenitors, respectively [53].
Taken together, expansion of tissue-derived SkM stem/progenitor cells can be extremely time-consuming, and cost-effective approaches need to be established. Challenges in this field include (i) limited cell source (low percentage of satellite cells in adult SkM); (ii) lack of robust cell isolation protocols that allow for the collection of sufficient cell numbers; (iii) limited expertise in implementing scalable expansion platforms; (iv) in vitro expanded satellite cells potentially exhibiting impaired ability for muscle engraftment in vivo [54]. More specifically, satellite cells and myoblasts exhibit restricted proliferation ex vivo, tend towards spontaneous differentiation or enter a senescent phenotype, limiting their expansion potential even more [16,24,55].
Figure 1. Schematic representation and comparison of isolation methods for primary SkMCs and SMCs: enzymatic digestion and explant-based approaches. Enzymatic-based methods for SkM and SM processing involve tissue sample collection, followed by tissue mincing, digestion with proteases (e.g., collagenase and/or dispase enzymes), filtering through a cell strainer, resulting in a cell suspension that is usually plated on coated surfaces for SkMCs (such as Matrigel) and on plastic surfaces for SMCs. Another cell isolation method is the explant-based protocol, which is mainly applied for SMCs, while for SkMCs, it is highly limited and has been mainly purposed towards tissue and disease modelling. The explant technique involves fragmentation of the tissue sample into approximately 1–2 mm diameter explants, followed by cell adhesion, migration and proliferation from the explants onto the plastic surface. Each isolation approach is compared in different categories: scale-up is easier when using enzymatic approaches, while explant-based can facilitate good manufacturing practice (GMP) compliance and simpler protocol optimization. As enzymatic approaches comprise the selection of enzyme(s) composition, concentration and digestion duration, there is a need for a balance between milder digestion protocols and insufficient cell retrieval in contrast to more harsh protocols that can result in higher cell numbers, but with limited viability. Although the resulting cell yield from both explant and enzyme-based methods is described as limited, enzymatic methods can be advantageous when having a reduced amount of starting sample. Adapted from [17,56,57]. GMP: good manufacturing practice; SkMCs: skeletal muscle cells; SMCs: smooth muscle cells.
Figure 1. Schematic representation and comparison of isolation methods for primary SkMCs and SMCs: enzymatic digestion and explant-based approaches. Enzymatic-based methods for SkM and SM processing involve tissue sample collection, followed by tissue mincing, digestion with proteases (e.g., collagenase and/or dispase enzymes), filtering through a cell strainer, resulting in a cell suspension that is usually plated on coated surfaces for SkMCs (such as Matrigel) and on plastic surfaces for SMCs. Another cell isolation method is the explant-based protocol, which is mainly applied for SMCs, while for SkMCs, it is highly limited and has been mainly purposed towards tissue and disease modelling. The explant technique involves fragmentation of the tissue sample into approximately 1–2 mm diameter explants, followed by cell adhesion, migration and proliferation from the explants onto the plastic surface. Each isolation approach is compared in different categories: scale-up is easier when using enzymatic approaches, while explant-based can facilitate good manufacturing practice (GMP) compliance and simpler protocol optimization. As enzymatic approaches comprise the selection of enzyme(s) composition, concentration and digestion duration, there is a need for a balance between milder digestion protocols and insufficient cell retrieval in contrast to more harsh protocols that can result in higher cell numbers, but with limited viability. Although the resulting cell yield from both explant and enzyme-based methods is described as limited, enzymatic methods can be advantageous when having a reduced amount of starting sample. Adapted from [17,56,57]. GMP: good manufacturing practice; SkMCs: skeletal muscle cells; SMCs: smooth muscle cells.
Bioengineering 10 01067 g001

3. Strategies for Advancing SMC and SkMC Manufacturing

The production of engineered tissues and organs requires the use of a large number of cells. However, a major challenge in what concerns the therapeutic application of SMCs and SkMCs relies on their low amount in the tissue sources, resulting in insufficient quantities of isolated cells for clinical use. Even though autologous treatments may have a lower risk of rejection, it may not be feasible for all patients due to limitations related to cell availability and impaired cell fitness. In this context, allogeneic cell manufacturing, in large-scale setups, holds great potential in the muscle regeneration field. As such, the identification of effective expansion techniques is the first crucial step to obtain the required number of functional cells in a time- and cost-effective manner. Section 3.1 and Section 3.2 describe strategies for improving the in vitro/ex vivo expansion of SMCs and SkMCs, testing multiple cell sources and different culture settings (e.g., culture medium composition, biomaterial). These are also summarized in Table 1 and Table 2 for SMCs and SkMCs, respectively.

3.1. SMCs

As aforementioned, one of the major limitations of cell-based regenerative therapy targeting SM is the lack of donor tissue suitable for cell harvesting. For this reason, the use of stem cells, namely iPSCs, as a source of therapeutic SMCs has generated increased interest in the field. In particular, human-induced pluripotent stem cell (hiPSC)-derived vascular smooth muscle cells (VSMCs) are of great value for disease modeling, drug screening, cell therapies, and tissue engineering, offering an innovative approach to replacing or bypassing diseased blood vessels [58]. Considering the great challenge of retrieving enough primary VSMCs from fetal or adult human tissues, hiPSCs are considered as a valuable resource due to their easy accessibility, expandability and ability to give rise to almost any desired cell type [59]. Also, patient-specific derived iPSCs retain the patient’s genetic information, allowing these cells to trigger little to no immune response after transplantation [59]. Still, protocols to efficiently produce high quantity of hiPSC-derived VSMCs need further optimization. From this perspective, a scalable method for manufacturing this cell type was developed using alginate hydrogel microtubes, which resulted in high viability, purity (>80%) and yield (~5.0 × 108 cells/mL) [60]. The alginate hydrogel offers protection from hydrodynamic stress and limits cell mass to less than 400 µm, ensuring efficient nutrient diffusion while also reducing cell agglomeration [60]. Moreover, bioreactor-expanded VSMCs contributed to blood vessel formation in vivo, while also retaining similar expression levels of VSMCs markers compared with 2D cultured VSMCs [60]. Also aiming to improve hiPSC-derived VSMC production, Fang et al. proposed a hypoxic (5% O2 ) treatment during differentiation, effectively inducing proliferative hiPSC-derived VSMCs, via embryoid body-based differentiation [61]. The hypoxic conditions enhanced the formation, adhesion and amplification rates of embryoid bodies, and upon directed differentiation, hiPSC-VSMCs exhibited increased cell viability compared to culture under atmospheric air [61]. Envisaging the application of cell or tissue-based products in clinical practice, it is imperative to adapt protocols towards xeno(geneic)-free conditions, since animal-derived reagents may carry zoonoses and trigger immune responses of cell or tissue derivatives, which could lead to graft failure, besides being associated with batch-to-batch variability and lack of standardization [62]. From this perspective, a combination of human serum and human platelet lysate demonstrated effectiveness in replacing fetal bovine serum (FBS) to generate VSMCs from hiPSCs [58]. Functional xeno-free hiPSC-derived VSMCs were successfully obtained, suitable for scaffold-assisted vascular tissue engineering, which exhibited comparable mechanical strength to those developed from xenogeneic hiPSC-derived VSMCs [58]. This finding is consistent with the application of both human serum and human platelet lysate as substitutes for FBS in expansion strategies for cell therapies and tissue engineering [62,63,64]. To further improve cell purification from iPSCs, Li et al. used sorting (MACS or FACS) for CD34-positive cells to enrich for common vascular progenitors, followed by differentiation towards SMCs using platelet-derived growth factor (PDGF-BB) as medium supplement [65]. The obtained SMC population was injected into mouse models of urinary incontinence and the authors observed tissue remodeling with higher detection of elastin in the bladder [65].
Another cell type investigated as a source to obtain differentiated SMCs are MSCs, particularly those derived from adipose tissue (also referred to as adipose-derived stem cells (ASCs)). Chemical, physical and biological cues can be used to drive stem cell fate in vitro [17]. The suitability of three-dimensional scaffolds for culture and differentiation of MSCs can be influenced by the physical properties of the scaffold. This includes surface topography, microstructure and mechanical specifications, which affect cell adhesion, proliferation and differentiation [66]. Moreover, the use of microcarriers combined with stirred bioreactors is a widely applied technique for expanding anchorage-dependent cells [67,68]. The microcarriers provide a large surface area for cell adhesion and growth in a homogeneous and controlled environment. Envisioning a protocol to expand and differentiate ASCs into SM-like cells, and exploiting a format that requires minimal manipulation before clinical delivery, Parmar et al. prepared a microcarrier formulation composed of a biocompatible and degradable material, poly (D,L lactic-co-glycolic acid) (PLGA), using a thermally induced phase separation (TIPS) technique [69]. This resulted in a highly porous structure that facilitated controlled degradation compared with solid microcarriers. The authors observed that ASCs readily attached to the surface of TIPS microcarriers, differentiating into an SM-like phenotype. Moreover, expansion of SMCs on the surface of the microcarriers did not alter the integrity of the polymer microspheres, making them suitable as a potential cell delivery vehicle [69]. Using a similar experimental design, Ahmadi et al. also observed that SMCs can attach to PLGA microcarriers in suspension culture and exhibited enhanced cell growth combined with increased cell release capacity at the sites of delivery [70]. By using ASCs combined with collagen in a microsphere format, Walters et al. observed that SMC morphology and identity markers were highly detected under the presence of growth factors, namely PDGF-AB and transforming growth factor (TGF-β1), and under mechanical stretch [71]. In another study, ASCs were also used as a source for SMCs, without any scaffolds, under differentiation culture conditions using a low percentage of serum (1% FBS) [72]. SMC markers were identified at the end of 3 and 6 weeks in culture, by detection of α-SMA, MYH11 and smoothelin at protein and mRNA levels. Still, further elucidation is needed regarding the cell numbers obtained, for example, per gram of initial sample of adipose tissue, as well as the functionality of the generated SMCs [72].
When targeting the development of more robust methods for muscle tissue manufacturing, it is not only crucial to design effective platforms for cell expansion, but one should also consider that engineered SM tissues should be constructed with well-differentiated and aligned SMCs for proper functioning, mimicking native tissue. Keeping this in mind, organized cell/scaffold hybrids were employed as functional SM constructs using a bioreactor system [73]. Briefly, prior to bioreactor expansion, cells were seeded into porous sheet-type scaffolds, fabricated with polyurethane, and then subjected to cyclic mechanical strain with a self-designed stretching chamber. It was possible to conclude that cells had proliferated effectively after a 7-day culture period in the bioreactor, retaining their native premodulated contractile characteristics [73]. Moreover, mechanical stimulation is particularly critical for engineered vascular tissue, where in vivo shear forces at the blood interface help maintain the function of the endothelium [74]. In fact, various studies have shown that mechanical stresses are essential for growing tissue-engineered vascular constructs [75]. In this context, Mun et al. associated a pulse bioreactor system with poly (lactide-co-caprolactone) electrospun scaffolds to generate a 3D tubular-shaped graft, which exhibited mechanical properties similar to native blood vessels [76]. The SMCs expanded in this platform retained their basic characteristics, namely presence of α-SMA, and presented higher proliferation rates under pulsatile flow than when compared to static culture. Moreover, the physical stimuli comprising medium flow and pressure through the lumen of the construct enabled mimicking aspects of the native physiological environment [76]. In an attempt to mimic a vascular tunica media graft, electrospun gelatin fiber scaffolds were developed to grow human umbilical vein SMCs, making it possible to obtain SM-like cells with high viability upon expansion in a bioreactor system [77]. In fact, the dynamic setup almost doubled the rate of cell proliferation through the scaffold compared to static conditions, forming a full tissue-like structure throughout a scaffold of 250–300 μm thickness, 6 days post-seeding [77]. Additionally, other studies have also investigated VSMCs seeding on naturally derived vascular scaffolds, such as decellularized matrices [78]. Knox et al. combined a decellularization approach of ovine arteries in a pulsatile flow bioreactor with cardiovascular progenitors to generate a biomimetic vascular graft [79]. In this setup, differentiated SMCs were observed by detection of calponin and MYH11 at the end of 3 weeks in culture [79]. Even though decellularized scaffolds possess many desirable characteristics for vascular replacements, such as niche-like architecture, mechanical properties and biochemical signals, they also entail some degree of structural weaknesses, such as low porosity and watertightness, hampering the possibility of direct VSMCs injection into the vessel wall [80]. In an attempt to develop bioengineered vessels with a more physiologically relevant SMC layer, a modular bioreactor and perfusion system were established to specifically allow for the proliferation of vascular-like constructs over extended culture periods [78]. Using a decellularized porcine artery as a model scaffold, the authors improved bioreactor design, including multiple contiguous functions contained within a single system, where tissue processing, cell seeding, and cell culture were developed to achieve clinically relevant constructs. However, in this study, VSMCs were unable to penetrate beyond the adventitial–medial boundary [78]. In this context, aiming to improve this intrinsic problem, Yazdani et al. designed a strategy consisting of cyclic bioreactor preconditioning (i.e., flow and pressure), combining it with surface modification (removal of the adventitial layer) of decellularized porcine carotid arteries [80]. This prompted an increase in cellular seeding efficiency and proliferation, thus promoting a more uniform deposition and density of mature VSMCs in the engineered vessel [80].
Table 1. Overview of relevant studies using different cell sources for SMC engineering.
Table 1. Overview of relevant studies using different cell sources for SMC engineering.
Cell SourceCulture Setup
(Scaffold; Cell Sorting;
Growth Factors)
Main Outcome(s)Reference
iPSCsAlginateHigh VSMC purity (>80%) and
yield (~5.0 × 108 cells/mL)
[60]
Embryoid-based differentiation
Normoxia vs. hypoxia (5%O2)
Higher VSMC viability under hypoxia[61]
Human platelet lysate and human serum vs. FBS culture medium supplementationMechanical strength comparable to hiPSC-derived VSMCs under FBS-supplemented medium[58]
Cell sorting for CD34+ cells
PDGF-BB for SMCs differentiation
In vivo injection of SMCs into mouse models of urinary incontinence: tissue remodeling with higher detection of elastin[65]
MSCsASCsLow FBS supplementationIdentification of SMCs markers[72]
PLGA microcarriersSMC-like phenotype, with cell attachment and proliferation on the microcarriers[69,70]
Collagen microspheres
PDGF-AB and TGF-β1
Mechanical stretch
Identification of SMCs markers[71]
ASCs spheroids
Bioprinting on gelatin–alginate
TGF-β for SMCs induction
Assessment of viability, proliferation and SMC differentiation post-bioprinting[81]
Primary SMCsPolyurethane porous sheet-type scaffold
Cyclic mechanical strain
Observation of SMCs contractile capacity[73]
Poly (lactide-co-caprolactone) electrospun scaffold and collagen coating
Pulse bioreactor
Identification of α-SMA, and SMCs presented higher proliferation rates under pulsatile flow than when compared to static culture[76]
Electrospun gelatin fiber scaffold in bioreactorDynamic setup led to higher cell proliferation than under static conditions[77]
Decellularized porcine artery in bioreactorHigh VSMC viability, variable levels of cell seeding in the decellularized matrices[78,80]
Nanofibrous gelatin–PLLA scaffoldIn vivo model of urethral reconstruction exhibited SMC remodeling upon transplantation[82]
Bilayer silk scaffoldSMC alignment and proliferation in the scaffold[83]
Electrospun PCL and GelMAHigh cell viability and proliferation[84]
Bioprinting on GelMA and hyaluronic acidBilayer of outer SMCs and inner ECs, with 20 mm length and 4 mm lumen diameter[85]
Cardiovascular progenitor cellsDecellularized ovine arteries in a pulsatile flow bioreactorVSMCs by detection of calponin and MYH11[79]
ASCs: adipose-derived stem cells; ECs: endothelial cells; FBS: fetal bovine serum; GelMA: gelatin methacrylate; iPSCs: induced pluripotent stem cells; MSCs: mesenchymal stromal cells; MYH11: myosin heavy chain-11; PCL: poly(ε-caprolactone); PDGF: platelet-derived growth factor; PLGA: poly (D,L lactic-co-glycolic acid); PLLA: poly(l-lactic acid); SMCs: smooth muscle cells; TGF: transforming growth factor; VSMCs: vascular smooth muscle cells; α-SMA: alpha-smooth muscle actin.
When considering heterotypic culture configurations, it has been reported that in vitro vascular ECs–SMCs cocultures have an effect on SMC proliferation, migration, phenotypic expression and ECM production [86]. ECs can modulate SMC expression profile by increasing the expression of vascular endothelial cell growth factor (VEGF), PDGF-AA, PDGF-BB and TGF-β genes, and decreasing basic fibroblast growth factor (bFGF or FGF-2) gene expression, compared to SMC culture alone [87]. As such, these insights can be applied to improve tissue engineering strategies. Also, in the context of SMCs–ECs cocultures, in a study by Williams et al., a vascular construct perfusion bioreactor was developed, allowing for direct contact between SMCs and ECs, while providing a biomechanical environment that mimics features of the in vivo hemodynamics [88]. In this design, the bioreactor contained tubular poly (glycolic acid) (PGA) nonwoven felts, which were seeded sequentially with SMCs and ECs under dynamic flow conditions. SMCs populated the porous biomaterial and ECs were seeded onto the lumen surface, allowing the two cell types to interact. The lumen was perfused with culture medium, and ECs were exposed to pulsatile shear forces, mimicking the native aortic vessel wall tissue conditions. Results showed significant increase in SMC proliferation, more uniform cell distribution, more contractile SMC phenotype and downregulation of ECM deposition in the long term, compared to short-term coculture constructs [88].
Taken together, there is increasing interest in engineering SMCs in vascular grafts, using different types of natural-derived (silk protein) or synthetic scaffolds (polycaprolactone). In several studies, which differ in multiple parameters, such as cell origin, culture reagents, and scaffold design, mechanical stimuli seem to be a critical feature towards an SMC phenotype [71,89,90].

3.2. SkMCs

Different tissue sources have been explored to obtain SkMCs, not only by direct isolation of satellite and/or progenitor cells, but also by myogenic induction of MSCs (isolated from adipose tissue—ASCs, bone marrow and umbilical cord), and iPSCs [17]. To the best of our knowledge, in what concerns clinical translation, only cells obtained from SkM and adipose tissue have been applied in human clinical studies, which is covered in Section 4.
Concerning MSCs, several animal models (mouse, rat or larger models, like pig) have been employed as recipient models of muscle damage to study labelled donor MSCs [91,92]. In these studies, it was shown that bone marrow-derived MSCs can be recruited to areas of muscle injury and directly participate in myogenic regeneration by differentiation into myoblasts [91,92]. MSCs derived from other sources, namely umbilical cord, synovial membrane, adipose tissue, or tonsil, have been also shown to support muscle regeneration in vivo [93,94,95]. Particularly, concerning the umbilical cord, MSCs from Wharton’s jelly exhibited higher myogenic differentiation potential compared to cells isolated from umbilical cord blood under in vitro differentiation conditions, which seem to be related to CD90 basal expression by MSCs [96]. Under static culture conditions, MSCs can differentiate into SkM through stimulation with growth factors, typically FGF-2 and PDGF-AA [91], and others not so commonly reported, such as hepatocyte growth factor (HGF) and insulin-like growth facor-1 (IGF-1) [96]. On the other hand, in more complex in vitro 3D models, growth factor supplementation with HGF and IGF-1 was not sufficient to induce MSC myogenic differentiation [97]. MSCs are also extensively recognized by their paracrine secretion, potentially supporting regenerative features in a therapeutic context. For instance, in vivo models have described the proangiogenic potential of MSCs, providing a permissive niche for muscle regeneration [98,99]. Nonetheless, the potential of MSCs as a source of SkMCs for tissue engineering strategies is not well established. In the preliminary work by Testa et al., MSC differentiation towards SkMCs was tested using human serum counterparts instead of FBS [100]. Increased levels of myogenesis markers were observed when using high serum- or platelet-rich plasma concentrations, namely features restricted to the detection of CD56 and myosin proteins [100].
ASCs, in particular, have demonstrated the ability to differentiate into the myocyte lineage [101]. This was shown not only by identification of MyoD and other myogenic transcription factors, but also morphologically, by formation of multinucleated myofibers by myogenic induction of ASCs. ASC differentiation into myogenic lineage can be achieved through culture medium supplementation (e.g., FGF-2 and GSK3 inhibitor), and increased using biophysical cues, namely cyclic strain [101,102]. In animal models, ASCs seemed to accelerate muscle repair despite the fact that donor ASCs were not detected in recipient damaged muscle, pointing towards a paracrine action [103]. Moreover, a long-term effect on muscle regeneration was not observed [103]. On the other hand, in vivo local or systemic administration of myogenic progenitors derived from ASCs resulted in long-term engraftment (12 weeks post-transplantation) in a mouse model of DMD [101]. In another study, MyoD expression was induced in ASCs through lentiviral transduction, and engineered cells were administered into murine models of injured muscle, which effectively incorporated into the multinucleated fibers in the newly repaired muscle [104].
The use of hiPSCs to obtain SkMCs has been challenging [16,105] and the field was recently reviewed by Iberite et al. [106]. Protocols towards SkM differentiation have been reported based on gene transfer and external factors medium supplementation (growth factors and small molecules) [16]. hiPSC reprograming through gene transfer, for instance, based on viral vectors, aims to modulate overexpression of essential myogenic transcription factors previously mentioned, namely Pax [107] and MyoD [108]. This type of strategy may be able to produce cells at high yields, although the requirement of viral vectors can hinder cell therapy and tissue engineering prospects due to potential safety issues and differentiated cells might display an immature phenotype [16].
Differentiation of hiPSCs towards SkMCs has also been reported through modulation of culture conditions, especially through GSK3 β inhibitors and FGF-2 supplementation [109,110,111], and also the use of ascorbic acid, BMP-4, IGF-1, insulin and PDGF [112]. Indeed, modulation of multiple signaling pathways (Wnt, TGF- β, Notch, BMP, PI3K, hedgehog and retinoic acid) has been described, adding extra challenges towards robust and consistent platforms to obtain SkMCs [105,112,113]. The common feature between the multiple protocols reported is the production of Pax7+ myogenic progenitors, although at different efficiencies [110,112,114]. Moreover, in order to enrich for myogenic progenitors, FACS has been applied, based on different surface markers according to the protocols used, which in turn also differ and are influenced by culture parameters [16,115]. In fact, the surface markers reported varied between ERBB3+NGFR+ [109,115]; CD24-CD10+ [116]; CD57ACHR+cMET+ [117]; and CD57 NCAM1+ [118]. To facilitate the FACS protocol, a Pax7-based reporter system was tested [119]. It was observed that transplantation of the enriched population of Pax7 transgene expression myogenic progenitors resulted in cell engraftment into the murine muscle, while injection of a mixed population without previous FACS enrichment did not produce myofibers into the damaged muscle [119]. Although this type of knowledge acquired from murine models is valuable, its application in a human regenerative medicine context is limited, as more studies are needed to clarify and validate these markers in human counterparts. Overall, the purification protocols for target cell populations seem to enrich for muscle precursors that can exhibit enhanced myotube formation in vitro or muscle engraftment in vivo. Moreover, although iPSC differentiation protocols can allow for the generation of high numbers of myogenic progenitors (e.g., 2 × 1016 cells upon 43 days of culture [120]), there is still a need to understand the role and representativeness of the surface markers employed when using cell purification strategies and its correlation with cell function and therapeutic potential [16].
Table 2. Overview of relevant studies using different cell sources for SkMC engineering.
Table 2. Overview of relevant studies using different cell sources for SkMC engineering.
Cell SourceCulture Setup
(Scaffold; Cell Sorting; Growth Factors)
Main Outcome(s)Reference
iPSCsGSK3 β inhibitor, FGF-2, ascorbic acid, BMP-4, IGF-1, insulin and PDGF [112]Generation of Pax7+ myogenic progenitors at varying efficiencies[112,114]
GSK3 β inhibitor, FGF-2 and ITS 2 × 1016 myogenic progenitors upon 43 days of culture[120]
FACS based on different surface markers: ERBB3+NGFR+;
CD24CD10+; CD57ACHR+cMET+;
CD57 NCAM1+
Enhanced in vitro myotube formation and/or enhanced muscle engraftment in vivo[115,116,117,118]
PCL scaffold with decellularized skeletal ECM motifsIn vivo cell integration in murine model of VML[121]
MSCsAdipose tissueGSK3 β inhibitor and FGF-2 supplementation
Cyclic strain
Identification of myogenic transcription factors and multinucleated myofibers[101,102]
BMFGF-2 and PDGF-AAObserved differentiation towards myoblasts[91]
HGF and IGF-1HGF and IGF-1 may not be sufficient for myogenic differentiation[97]
Umbilical cordHorse serum-supplemented medium or HGF, IGF-1 and FGF-2Enhanced myogenic differentiation potential of MSCs from umbilical cord tissue compared to MSCs from umbilical cord blood[96]
Primary muscleHuman serum vs. FBSEnhanced detection of CD56 and myosin proteins under high serum or platelet-rich plasma[100]
Primary SkMCsFACS depletion for CD45, CD31, CD11b, Sca1 and enrichment for CD34 and/or integrinα7
Fibrin vs. Matrigel
Higher cell expansion (threefold) on fibrin gel than Matrigel[122]
Electrospun nanofiber of PMMALaminin-coated PMMA facilitated myoblast proliferation in comparison to collagen-coated PMMA [123]
Collagen and Matrigel replating stepsShort-time protocol with cell isolation from low amounts of skeletal muscle (0.02 g) [124]
Fibrin-based hydrogelElectrical stimulation led to improved contractility and mature phenotype[125]
BMP: bone morphogenetic protein; ECM: extracellular matrix; FACS: fluorescence-activated cell sorting; FBS: fetal bovine serum; HGF: hepatocyte growth factor; IGF-1: insulin-like growth facor-1; iPSCs: induced pluripotent stem cells; ITS: insulin–transferrin–selenium–ethanolamine; MSCs: mesenchymal stromal cells; PCL: poly(ε-caprolactone); PDGF: platelet-derived growth factor; PMMA: poly(methyl methacrylate); SkMCs: skeletal muscle cells; VML: volumetric muscle loss.
Coculture strategies have been reported to recapitulate more closely the native SkM environment. Cocultures of isolated myoblasts with fibroblasts seem to improve the migration of the myogenic cells, but no effects on proliferation or myotube formation were explored [44]. Other authors reported cocultures of primary SKMCs with MSCs to improve myogenic differentiation and did not observe a significant effect on muscle cell proliferation [126,127]. Particularly, in a study by Cai et al., cocultures of primary rat myoblasts with MSCs from bone marrow or adipose tissue were performed on electrospun polycaprolactone and collagen scaffolds [127]. Under differentiation conditions for myoblasts (a low serum concentration (2%) and a serum-free formulation), it was possible to observe an upregulation of myogenic markers such as myosin heavy chain 2 and α-actinin 2 on the coculture settings in comparison to monocultures [127]. On the other hand, in a study by Juhas et al., an immune cell population, macrophages, was included into engineered constructs of murine myoblasts within Matrigel and fibrin [128]. Promotion of myogenesis and less myofiber apoptosis were observed after induced chemical muscle injury [128]. Interestingly, other authors reported the use of inflammation-related cytokines to improve in vitro satellite cell culture and in vivo muscle engraftment in a recipient mouse model [129]. Moreover, the use of conditioned medium from other immune cells, T lymphocytes, seemed to sustain satellite cell proliferation to higher cell passages [130].
Attempts to expand satellite cells ex vivo have been described in multiple studies, whether by inhibiting the production of differentiation factors or by promoting their proliferative capacity [55,131]. Of note, culture parameter specifications required to promote human myoblast proliferation and prevent spontaneous differentiation during the expansion phase may include close monitoring of cell confluence levels and specific coatings, such as those laminin-based [132,133].
Besides the modulation of biochemical conditions through different culture medium cocktails, physical cues are also critical to SkMCs. For example, soft hydrogels with elastic modulus similar to muscle tissue were shown to sustain self-renewal of satellite cells better than plastic surfaces [134]. Importantly, two frequently used biomaterials, collagen, usually derived from rat tail, and Matrigel, represent xenogeneic options to be used in this context [17]. Zhu et al. also used xenogeneic fibrin in comparison to Matrigel to culture murine SkMCs after tissue digestion with collagenase and dispase, and FACS enrichment [122]. Cells negative for CD45, CD31, CD11b, Sca1 markers, and positive for CD34 and/or integrin-α7 were considered muscle stem cells and a threefold higher cell expansion factor was reported when using fibrin gel compared to Matrigel. The use of biomaterials in SkM engineering has been extensively reviewed [135] and there is a huge search for matrices that can mimic the native ECM and provide structural architecture with the mechanical signals offered by the scaffold supporting satellite cell maintenance [136].
Overall, there is still an unmet need towards the development of culture systems amenable to recapitulate the native tissue microenvironment ex vivo in order to support the self-renewal of satellite cells and the maintenance of their regenerative ability through the generation of differentiated cells with a contractile function [17].

4. Clinical Studies with Expanded SMCs and SkMCs

Despite the multiple efforts performed towards muscle regenerative approaches, there is still a limited number of human clinical trials studying the administration of SMCs or SkMCs. Importantly, upon transplantation into damaged tissues, the administered cells (SMCs or SkMCs) face unfavorable environment signals (e.g., limiting oxygen concentrations), which hinders their maintenance, proliferation and function [137]. Concerning SkM, stem/progenitor cells have shown the potential to restore injured muscle, although it has been observed that their capacity ex vivo is lost, as aforementioned.
Table 3 summarizes the main clinical trials employing distinct types of muscle cells in the autologous context and for different pathological settings, with functional incontinence being one of the major applications. The required cell number of SMCs or SkMCs varied among the studies analyzed, but included multiple doses with a range from 5 to 200 million cells per administration [138]. Of notice, several clinical trials employed ASCs as a therapeutic approach for stress urinary incontinence [139,140,141]. Despite the fact that the use of ASCs could potentially overcome the limited efficiency of the ex vivo expansion of SkMCs and SMCs, a trial with five female patients did not show any effective improvement [139]. Overall, ASCs hold great promise as a cell source for muscle repair and regeneration, but further studies are needed to establish their effectiveness. In general, most of the trials showed safety (phase I) and preliminary efficacy (phase I/II) but are limited in terms of objective measurements of successful outcome.

5. Conclusions

The wide distribution of SM and SkM throughout the body, along with their unique properties, highlights the importance of in-depth knowledge of their anatomy, physiology and function. This understanding is crucial to addressing clinical needs in cases of organ or tissue damage.
A critical aspect in the context of muscle tissue engineering is the limited availability and suitability of donor tissues for cell harvesting. Autologous cells, for instance, might display impaired regenerative potential due to a specific pathological setting and/or aging. To overcome this challenge, alternative cell sources, such as iPSCs or adult stem/progenitor cells like MSCs, have been proposed as a starting source for myogenic differentiation, particularly for SMC bioengineering [58,59,69]. Similar considerations apply to SkMC engineering, in which tissue resident stem cells (or satellite cells) represent a potential cell source, though with challenges, especially when considering elderly patients [17,51].
Bioreactors are crucial tools for the robust development of standardized and high-quality engineered tissue products, with great potential to advance therapies targeting muscle repair and regeneration. Indeed, bioreactor technologies can be used to (i) expand the target cell types to generate clinically relevant cell numbers and/or (ii) mimic the in vivo microenvironment by exposing cells to relevant physical and biochemical stimuli allowing for the generation of cells and tissues with the desired identity and function. Of note, the development of engineered muscle tissues also relies on the use of biomaterials able to support tissue function, and more complex approaches, such as bioprinting, are being explored [153,154].
Overall, the translation of engineered muscle tissue-based strategies into clinics depends on biotechnology advances involving the combination of innovative cell culture technologies and biomaterial scaffold fabrication, followed by the optimization of large-scale manufacturing processes (Figure 2). Importantly, tissue engineering-based approaches are classified as advanced therapy medicinal cell products (ATMPs) by the European Medicines Agency (EMA). Thus, both biomaterial and cellular components must undergo rigorous quality control and detailed certification to meet good manufacturing practice (GMP) criteria. This entails the need for directing strategies from the very beginning of the experimental setup towards translation in order to establish muscle tissue engineering grafts as the future gold standard for muscle repair and regeneration.

Author Contributions

Conceptualization, T.F.-M., M.S. and C.L.d.S.; writing—original draft preparation, T.F.-M. and M.S.; figure preparation, A.L.C. and T.F.-M.; writing—review and editing, T.F.-M., M.S., A.L.C., A.F.-P., J.M.S.C. and C.L.d.S. All authors have read and agreed to the published version of the manuscript.

Funding

Projects UROPRINT (Urinary bladder bioprinting for fully autologous transplantation) and AMELIE (Anchored Muscle Cells for Incontinence) are funded by the European Union’s Horizon 2020 Research and Innovation Program (grant agreements no 964883 and no 874807, respectively).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Xu, X.; Yi, F.; Pan, H.; Duan, S.; Ding, Z.; Yuan, G.; Qu, J.; Zhang, H. Progress and prospects in stem cell therapy. Nat. Publ. Gr. 2013, 34, 741–746. [Google Scholar] [CrossRef] [PubMed]
  2. Dumont, N.A.; Bentzinger, C.F.; Sincennes, M.; Rudnicki, M.A. Satellite Cells and Skeletal Muscle Regeneration. Compr. Physiol. 2015, 5, 1027–1059. [Google Scholar] [PubMed]
  3. Musar, A. Muscle Homeostasis and Regeneration: From Molecular Mechanisms to Therapeutic Opportunities. Cells 2020, 9, 2033. [Google Scholar] [CrossRef] [PubMed]
  4. Crisafulli, S.; Sultana, J.; Fontana, A.; Salvo, F.; Messina, S.; Trifirò, G. Global epidemiology of Duchenne muscular dystrophy: An updated systematic review and meta-analysis. Orphanet J. Rare Dis. 2020, 15, 141. [Google Scholar] [CrossRef]
  5. Edenfield, A.; Patnam, R.; Swift, S. A narrative review of the epidemiology, diagnosis, and treatment of latent stress urinary incontinence. Neurourol. Urodyn. 2019, 38, S7–S11. [Google Scholar] [CrossRef]
  6. Zhang, R.Q.; Xia, M.C.; Cui, F.; Chen, J.W.; Bian, X.D.; Xie, H.J.; Shuang, W.B. Epidemiological survey of adult female stress urinary incontinence. BMC Women’s Health 2021, 21, 172. [Google Scholar] [CrossRef]
  7. Nguyen, J.H.; Chung, J.D.; Lynch, G.S.; Ryall, J.G. The Microenvironment Is a Critical Regulator of Muscle Stem Cell Activation and Proliferation. Front. Cell Dev. Biol. 2019, 7, 254. [Google Scholar] [CrossRef]
  8. Qazi, T.H.; Duda, G.N.; Ort, M.J.; Perka, C.; Geissler, S.; Winkler, T. Cell therapy to improve regeneration of skeletal muscle injuries. J. Cachexia Sarcopenia Muscle 2019, 49, 501–516. [Google Scholar] [CrossRef]
  9. Touyz, R.M.; Alves-lopes, R.; Rios, F.J.; Camargo, L.L.; Anagnostopoulou, A.; Arner, A.; Montezano, A.C. Vascular smooth muscle contraction in hypertension. Cardiovasc. Res. 2018, 15, 529–539. [Google Scholar] [CrossRef]
  10. Sinha, S.; Iyer, D.; Granata, A. Embryonic origins of human vascular smooth muscle cells: Implications for in vitro modeling and clinical application. Cell. Mol. Life Sci. 2014, 71, 2271. [Google Scholar] [CrossRef]
  11. Donadon, M.; Santoro, M.M. The Origin and Mechanisms of Smooth Muscle Cell Development in Vertebrates, The Company of Biologists Ltd. Development. 2021, 148, dev197384. [Google Scholar] [CrossRef] [PubMed]
  12. Iwamoto, Y.; Maruhashi, T.; Kajikawa, M.; Oda, N.; Kishimoto, S.; Matsui, S.; Hashimoto, H.; Aibara, Y.; Mohamad, F.; Hidaka, T.; et al. Chronic kidney disease is associated with vascular smooth muscle dysfunction but not with endothelial dysfunction. Int. J. Cardiol. 2018, 254, 284–290. [Google Scholar] [CrossRef] [PubMed]
  13. Zhang, J.C.L.; Kim, S.; Helmke, B.P.; Yu, W.W.; Du, K.L.; Lu, M.M.; Strobeck, M.; Yu, Q.-C.; Parmacek, M.S. Analysis of SM22α-Deficient Mice Reveals Unanticipated Insights into Smooth Muscle Cell Differentiation and Function. Mol. Cell. Biol. 2001, 21, 1336. [Google Scholar] [CrossRef]
  14. Scharner, J.; Zammit, P.S. The muscle satellite cell at 50: The formative years. Skelet. Muscle 2011, 1, 28. [Google Scholar] [CrossRef] [PubMed]
  15. Liu, J.; Saul, D.; Böker, K.O.; Ernst, J.; Lehman, W.; Schilling, A.F. Current Methods for Skeletal Muscle Tissue Repair and Regeneration. Biomed Res. Int. 2018, 16, 1984879. [Google Scholar] [CrossRef] [PubMed]
  16. Judson, R.N.; Rossi, F.M.V. Towards stem cell therapies for skeletal muscle repair. NPJ Regen. Med. 2020, 5, 10. [Google Scholar] [CrossRef]
  17. Salemi, S.; Prange, J.A.; Baumgartner, V.; Mohr-Haralampieva, D.; Eberli, D. Adult stem cell sources for skeletal and smooth muscle tissue engineering. Stem Cell Res. Ther. 2022, 13, 156. [Google Scholar] [CrossRef]
  18. Handschin, C.; Mortezavi, A.; Plock, J.; Eberli, D. External physical and biochemical stimulation to enhance skeletal muscle bioengineering. Adv. Drug Deliv. Rev. 2015, 82–83, 168–175. [Google Scholar] [CrossRef]
  19. Kuang, S.; Kuroda, K.; Le Grand, F.; Rudnicki, M.A. Asymmetric self-renewal and commitment of satellite stem cells in muscle. Cell 2007, 129, 999–1010. [Google Scholar] [CrossRef]
  20. 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. 2019, 76, 2559–2570. [Google Scholar] [CrossRef]
  21. Yablonka-Reuveni, Z.; Day, K.; Vine, A.; Shefer, G. Defining the transcriptional signature of skeletal muscle stem cells. J. Anim. Sci. 2008, 86, E207–E216. [Google Scholar] [CrossRef]
  22. Oprescu, S.N.; Yue, F.; Qiu, J.; Brito, L.F.; Kuang, S. Temporal Dynamics and Heterogeneity of Cell Populations during Skeletal Muscle Regeneration. iScience 2020, 23, 100993. [Google Scholar] [CrossRef]
  23. Sacco, A.; Doyonnas, R.; Kraft, P.; Vitorovic, S.; Blau, H.M. Self-renewal and expansion of single transplanted muscle stem cells. Nature 2008, 456, 502–506. [Google Scholar] [CrossRef]
  24. Marg, A.; Escobar, H.; Karaiskos, N.; Grunwald, S.A.; Metzler, E.; Kieshauer, J.; Sauer, S.; Pasemann, D.; Malfatti, E.; Mompoint, D.; et al. Human muscle-derived CLEC14A-positive cells regenerate muscle independent of PAX7. Nat. Commun. 2019, 10, 5776. [Google Scholar] [CrossRef] [PubMed]
  25. Lecourt, S.; Marolleau, J.P.; Fromigué, O.; Vauchez, K.; Andriamanalijaona, R.; Ternaux, B.; Lacassagne, M.N.; Robert, I.; Boumédiene, K.; Chéreau, F.; et al. Characterization of distinct mesenchymal-like cell populations from human skeletal muscle in situ and in vitro. Exp. Cell Res. 2010, 316, 2513–2526. [Google Scholar] [CrossRef] [PubMed]
  26. Lu, H.; Huang, D.; Ransohoff, R.M.; Zhou, L. Acute skeletal muscle injury: CCL2 expression by both monocytes and injured muscle is required for repair. FASEB J. 2011, 25, 3344. [Google Scholar] [CrossRef] [PubMed]
  27. Deng, B.; Wehling-Henricks, M.; Villalta, S.A.; Wang, Y.; Tidball, J.G. IL-10 triggers changes in macrophage phenotype that promote muscle growth and regeneration. J. Immunol. 2012, 189, 3669–3680. [Google Scholar] [CrossRef] [PubMed]
  28. Koike, H.; Manabe, I.; Oishi, Y. Mechanisms of cooperative cell-cell interactions in skeletal muscle regeneration. Inflamm. Regen. 2022, 42, 48. [Google Scholar] [CrossRef]
  29. Huard, J.; Cao, B.; Qu-Petersen, Z. Muscle-derived stem cells: Potential for muscle regeneration. Birth Defects Res. Part C Embryo Today 2003, 69, 230–237. [Google Scholar] [CrossRef]
  30. Matthias, N.; Hunt, S.D.; Wu, J.; Lo, J.; Smith Callahan, L.A.; Li, Y.; Huard, J.; Darabi, R. Volumetric muscle loss injury repair using in situ fibrin gel cast seeded with muscle-derived stem cells (MDSCs). Stem Cell Res. 2018, 27, 65–73. [Google Scholar] [CrossRef]
  31. Liu, M.; Gomez, D. Smooth Muscle Cell phenotypic diversity: At the crossroads of lineage tracing and single-cell transcriptomics. Arterioscler. Thromb. Vasc. Biol. 2020, 39, 1715–1723. [Google Scholar] [CrossRef]
  32. Pokrywczynska, M.; Balcerczyk, D.; Jundzill, A.; Gagat, M.; Czapiewska, M.; Kloskowski, T.; Nowacki, M.; Gastecka, A.M.; Bodnar, M.; Grzanka, A.; et al. Isolation, expansion and characterization of porcine urinary bladder smooth muscle cells for tissue engineering. Biol. Proced. Online 2016, 18, 17. [Google Scholar] [CrossRef] [PubMed]
  33. Adhikari, N.; Shekar, K.C.; Staggs, R.; Win, Z.; Steucke, K.; Lin, Y.-W.; Wei, L.-N.; Alford, P.; Hall, J.L. Guidelines for the Isolation and Characterization of Murine Vascular Smooth Muscle Cells. A Report from the International Society of Cardiovascular Translational Research. J. Cardiovasc. Transl. Res. 2016, 8, 158–163. [Google Scholar] [CrossRef] [PubMed]
  34. Ray, J.; Leach, R.; Herbert, J.; Benson, M. Isolation of vascular smooth msucle cells from a single murine aorta. Methods Cell Sci. 2001, 23, 185–188. [Google Scholar] [CrossRef] [PubMed]
  35. He, C.; Wang, M.; Yan, Z.; Zhang, S.; Liu, H. Isolation and culture of vascular smooth muscle cells from rat placenta. J. Cell. Physiol. 2019, 234, 7675–7682. [Google Scholar] [CrossRef] [PubMed]
  36. Weber, S.C.; Gratopp, A.; Akanbi, S.; Rheinlaender, C.; Sallmon, H.; Barikbin, P.; Koehne, P.S. Isolation and Culture of Fibroblasts, Vascular Smooth Muscle, and Endothelial Cells From the Fetal Rat Ductus Arteriosus. Pediatr. Res. 2011, 70, 236–241. [Google Scholar] [CrossRef] [PubMed]
  37. Metz, R.P.; Patterson, J.L.; Wilson, E. Vascular Smooth Muscle Cells: Isolation, Culture, and Characterization. Cardiovasc. Dev. Methods Protoc. 2012, 843, 169–176. [Google Scholar] [CrossRef]
  38. Day, K.; Shefer, G.; Shearer, A.; Yablonka-Reuveni, Z. The depletion of skeletal muscle satellite cells with age is concomitant with reduced capacity of single progenitors to produce reserve progeny. Dev. Biol. 2010, 340, 330–343. [Google Scholar] [CrossRef]
  39. Hindi, L.; McMillan, J.; Afroze, D.; Hindi, S.; Kumar, A. Isolation, Culturing, and Differentiation of Primary Myoblasts from Skeletal Muscle of Adult Mice. Bio-Protocol 2017, 7, e2248. [Google Scholar] [CrossRef]
  40. Shahini, A.; Vydiam, K.; Choudhury, D.; Rajabian, N.; Nguyen, T.; Lei, P.; Andreadis, S.T. Efficient and high yield isolation of myoblasts from skeletal muscle. Stem Cell Res. 2018, 30, 122–129. [Google Scholar] [CrossRef]
  41. Ceusters, J.; Lejeune, J.P.; Sandersen, C.; Niesten, A.; Lagneaux, L.; Serteyn, D. From skeletal muscle to stem cells: An innovative and minimally-invasive process for multiple species. Sci. Rep. 2017, 7, 696. [Google Scholar] [CrossRef]
  42. Smith, L.R.; Meyer, G.A. Skeletal muscle explants: Ex-vivo models to study cellular behavior in a complex tissue environment. Connect. Tissue Res. 2020, 61, 248–261. [Google Scholar] [CrossRef] [PubMed]
  43. Calve, S.; Odelberg, S.J.; Simon, H.G. A transitional extracellular matrix instructs cell behavior during muscle regeneration. Dev. Biol. 2010, 344, 259–271. [Google Scholar] [CrossRef] [PubMed]
  44. Goetsch, K.P.; Snyman, C.; Myburgh, K.H.; Niesler, C.U. Simultaneous isolation of enriched myoblasts and fibroblasts for migration analysis within a novel co-culture assay. Biotechniques 2015, 58, 25–32. [Google Scholar] [CrossRef]
  45. Wang, Z.; Cheung, D.; Zhou, Y.; Han, C.; Fennelly, C.; Criswell, T.; Soker, S. An in vitro culture system that supports robust expansion and maintenance of in vivo engraftment capabilities for myogenic progenitor cells from adult mice. Biores. Open Access 2014, 3, 79–87. [Google Scholar] [CrossRef]
  46. Motohashi, N.; Asakura, Y.; Asakura, A. Isolation, Culture, and Transplantation of Muscle Satellite Cells. J. Vis. Exp. 2014, 86, 50846. [Google Scholar] [CrossRef]
  47. Pasut, A.; Jones, A.E.; Rudnicki, M.A. Isolation and culture of individual myofibers and their satellite cells from adult skeletal muscle. J. Vis. Exp. 2013, 73, e50074. [Google Scholar] [CrossRef]
  48. Liu, L.; Cheung, T.H.; Charville, G.W.; Rando, T.A. Isolation of skeletal muscle stem cells by fluorescence-activated cell sorting. Nat. Protoc. 2015, 10, 1612–1624. [Google Scholar] [CrossRef]
  49. Wei, Y.; Li, Y.; Chen, C.; Stoelzel, K.; Kaufmann, A.M.; Albers, A.E. Human skeletal muscle-derived stem cells retain stem cell properties after expansion in myosphere culture. Exp. Cell Res. 2011, 317, 1016–1027. [Google Scholar] [CrossRef]
  50. Garcia, S.M.; Tamaki, S.; Lee, S.; Wong, A.; Jose, A.; Dreux, J.; Kouklis, G.; Sbitany, H.; Seth, R.; Knott, P.D.; et al. High-Yield Purification, Preservation, and Serial Transplantation of Human Satellite Cells. Stem Cell Rep. 2018, 10, 1160–1174. [Google Scholar] [CrossRef] [PubMed]
  51. Sousa-Victor, P.; García-Prat, L.; Muñoz-Cánoves, P. Control of satellite cell function in muscle regeneration and its disruption in ageing. Nat. Rev. Mol. Cell Biol. 2022, 23, 204–226. [Google Scholar] [CrossRef] [PubMed]
  52. Fleming, J.W.; Capel, A.J.; Rimington, R.P.; Wheeler, P.; Leonard, A.N.; Bishop, N.C.; Davies, O.G.; Lewis, M.P. Bioengineered human skeletal muscle capable of functional regeneration. BMC Biol. 2020, 18, 145. [Google Scholar] [CrossRef] [PubMed]
  53. Zammit, P.S. Function of the myogenic regulatory factors Myf5, MyoD, Myogenin and MRF4 in skeletal muscle, satellite cells and regenerative myogenesis. Semin. Cell Dev. Biol. 2017, 72, 19–32. [Google Scholar] [CrossRef]
  54. Du, X.; Li, W.; Du, G.; Cho, H.; Yu, M.; Fang, Q.; Lee, L.P.; Fang, J. Droplet Array-Based 3D Coculture System for High-Throughput Tumor Angiogenesis Assay. Anal. Chem. 2018, 90, 3253–3261. [Google Scholar] [CrossRef] [PubMed]
  55. Lean, G.; Halloran, M.; Mariscal, O.; Jamet, S.; Lumb, J.-P.; Crist, C. Ex vivo expansion of skeletal muscle stem cells with a novel small compound inhibitor of eIF2α dephosphorylation. Regen Med. Front. 2019, 1, e190003. [Google Scholar] [CrossRef]
  56. Pantelic, M.N.; Larkin, L.M. Stem Cells for Skeletal Muscle Tissue Engineering. Tissue Eng. Part B Rev. 2018, 24, 373–391. [Google Scholar] [CrossRef]
  57. Jonah, D.; Lee, B.C.S.; Lisa, M.; Larkin, K.W.V. Isolation and Purification of Satellite Cells for Skeletal Muscle Tissue Engineering. J. Regen. Med. 2015, 3, 117. [Google Scholar] [CrossRef]
  58. Luo, J.; Lin, Y.; Shi, X.; Li, G.; Kural, M.H.; Anderson, C.W.; Ellis, M.W.; Riaz, M.; Tellides, G.; Niklason, L.E.; et al. Xenogeneic-free Generation of Vascular Smooth Muscle Cells from Human Induced Pluripotent Stem Cells for Vascular Tissue Engineering. Acta Biomater. 2022, 119, 155–168. [Google Scholar] [CrossRef]
  59. Hockemeyer, D.; Jaenisch, R.; Biology, C. Induced pluripotent stem cells meet genome editing. Cell Stem Cell 2017, 18, 573–586. [Google Scholar] [CrossRef]
  60. Lin, H.; Qiu, X.; Du, Q.; Li, Q.; Wang, O.; Akert, L.; Wang, Z.; Anderson, D.; Liu, K.; Gu, L.; et al. Engineered Microenvironment for Manufacturing Human Pluripotent Stem Cell-Derived Vascular Smooth Muscle Cells Haishuang. Stem Cell Rep. 2019, 12, 84–97. [Google Scholar] [CrossRef]
  61. Fang, L.; Mei, J.; Yao, B.; Liu, J.; Liu, P.; Wang, X.; Zhou, J.; Lin, Z. Hypoxia facilitates proliferation of smooth muscle cells derived from pluripotent stem cells for vascular tissue engineering. J. Tissue Eng. Regen. Med. 2022, 16, 744–756. [Google Scholar] [CrossRef] [PubMed]
  62. Astori, G.; Amati, E.; Bambi, F.; Bernardi, M.; Chieregato, K.; Schäfer, R.; Sella, S.; Rodeghiero, F. Platelet lysate as a substitute for animal serum for the ex-vivo expansion of mesenchymal stem / stromal cells: Present and future. Stem Cell Res. Ther. 2016, 7, 93. [Google Scholar] [CrossRef] [PubMed]
  63. Henschler, R.; Gabriel, C.; Schallmoser, K.; Burnouf, T.; Koh, M.B.C. Human platelet lysate current standards and future developments. Transfusion 2019, 59, 1407–1413. [Google Scholar] [CrossRef]
  64. Pérez-simon, J.A.; López-villar, O.; Andreu, E.J.; Rifón, J.; Muntion, S.; Campelo, M.D.; Sánchez-guijo, F.M.; Martinez, C.; Valcarcel, D.; Cañizo, C. Mesenchymal stem cells expanded in vitro with human serum for the treatment of acute and chronic graft- versus -host disease: Results of a phase I/II clinical trial. Haematologica 2011, 96, 1072–1076. [Google Scholar] [CrossRef]
  65. Li, Y.; Green, M.; Wen, Y.; Wei, Y.; Wani, P.; Wang, Z.; Pera, R.R.; Chen, B. Efficacy and Safety of Immuno-Magnetically Sorted Smooth Muscle Progenitor Cells Derived from Human-Induced Pluripotent Stem Cells for Restoring Urethral Sphincter Function. Stem Cells Transl. Med. 2017, 6, 1158–1167. [Google Scholar] [CrossRef]
  66. Li, J.; Liu, Y.; Zhang, Y.; Yao, B.; Enhejirigala, B.; Li, Z.; Song, W.; Wang, Y.; Duan, X.; Yuan, X.; et al. Biophysical and Biochemical Cues of Biomaterials Guide Mesenchymal Stem Cell Behaviors. Front. Cell Dev. Biol. 2021, 9, 640388. [Google Scholar] [CrossRef] [PubMed]
  67. Knight, T.; Basu, J.; Rivera, E.A.; Spencer, T.; Jain, D.; Payne, R. Fabrication of a multi-layer three-dimensional scaffold with controlled porous micro-architecture for application in small intestine tissue engineering. Cell Adhes. Migr. 2013, 7, 267–274. [Google Scholar] [CrossRef]
  68. Schop, D.; Janssen, F.W.; Borgart, E.; Bruijn, J.D. De Expansion of mesenchymal stem cells using a microcarrier-based cultivation system: Growth and metabolism. J. Tissue Eng. Regen. Med. 2008, 2, 126–135. [Google Scholar] [CrossRef]
  69. Parmar, N.; Ahmadi, R.; Day, R.M. A Novel Method for Differentiation of Human Mesenchymal Stem Cells into Smooth Muscle-Like Cells on Clinically Deliverable Thermally Induced. Tissue Eng. 2015, 21, 404–412. [Google Scholar] [CrossRef]
  70. Ahmadi, R.; Mordan, N.; Forbes, A.; Day, R.M. Acta Biomaterialia Enhanced attachment, growth and migration of smooth muscle cells on microcarriers produced using thermally induced phase separation. Acta Biomater. 2011, 7, 1542–1549. [Google Scholar] [CrossRef]
  71. Walters, B.; Turner, P.A.; Rolauffs, B.; Hart, M.L.; Stegemann, J.P. Controlled Growth Factor Delivery and Cyclic Stretch Induces a Smooth Muscle Cell-like Phenotype in Adipose-Derived Stem Cells. Cells 2021, 10, 3123. [Google Scholar] [CrossRef]
  72. Salem, S.A.; Hwie, A.N.M.; Saim, A.; Kong, C.H.C.; Sagap, I.; Singh, R.; Yusof, M.R.; Zainuddin, Z.M.; Idrus, R.H. Human Adipose Tissue Derived Stem Cells as a Source of Smooth Muscle Cells in the Regeneration of Muscular Layer of Urinary Bladder Wall. MJMS 2013, 20, 80. [Google Scholar] [PubMed]
  73. Cha, J.M.; Park, S.N.; Park, G.O.; Kim, J.K.; Suh, H. Construction of functional soft tissues from premodulated smooth muscle cells using a bioreactor system. Artif. Organs 2006, 30, 704–707. [Google Scholar] [CrossRef] [PubMed]
  74. Meza, D.; Musmacker, B.; Steadman, E.; Stransky, T.; Rubenstein, D.A.; Yin, W. Endothelial Cell Biomechanical Responses are Dependent on Both Fluid Shear Stress and Tensile Strain. Cell. Mol. Bioeng. 2019, 12, 311–325. [Google Scholar] [CrossRef]
  75. Rouwkema, J.; Koopman, B.F.; Van Blitterswijk, C.A.; Dhert, W.J.; Malda, J. Supply of Nutrients to Cells in Engineered Tissues Supply of Nutrients to Cells in Engineered. Biotechnol. Genet. Eng. Rev. 2013, 8725, 2046–5556. [Google Scholar] [CrossRef]
  76. Mun, C.H.; Jung, Y.; Kim, S.; Kim, H.C.; Kim, S.H. Effects of Pulsatile Bioreactor Culture on Vascular Smooth Muscle Cells Seeded on Electrospun Poly (lactide-co-ε-caprolactone) Scaffold. Artif. Organs 2013, 8, 256–261. [Google Scholar] [CrossRef] [PubMed]
  77. Elsayed, Y.; Lekakou, C.; Labeed, F.; Tomlins, P. Smooth muscle tissue engineering in crosslinked electrospun gelatin scaffolds. Soc. Biomater. 2015, 8, 313–321. [Google Scholar] [CrossRef]
  78. McFetridge, P.S.; Daniel, J.W.; Bodamyali, T.; Horrocks, M.; Chaudhuri, J.B. Preparation of porcine carotid arteries for vascular tissue engineering applications. J. Biomed. Mater. Res.—Part A 2004, 70, 224–234. [Google Scholar] [CrossRef]
  79. Knox, C.; Garcia, K.; Tran, J.; Wilson, S.M.; Blood, A.B.; Kearns-Jonker, M.; Martens, T.P. A Biomimetic Approach Utilizing Pulsatile Perfusion Generates Contractile Vascular Grafts. Tissue Eng. Part A 2023, 29, 358–371. [Google Scholar] [CrossRef]
  80. Yazdani, S.K.; Watts, B.; Machingal, M.; Jarajapu, Y.P.; Van Dyke, M.E.; Christ, G.J.; Amiel, G.E.; Komura, M.; Shapira, O.; Yoo, J.J.; et al. Smooth Muscle Cell Seeding of Decellularized Scaffolds: The Importance of Bioreactor Preconditioning to Development of a More Native Architecture for Tissue-Engineered Blood Vessels. Tissue Eng. 2009, 15, 827–840. [Google Scholar] [CrossRef]
  81. Yipeng, J.; Yongde, X.; Yuanyi, W.; Jilei, S.; Jiaxiang, G.; Jiangping, G.; Yong, Y. Microtissues enhance smooth muscle differentiation and cell viability of hADSCs for three dimensional bioprinting. Front. Physiol. 2017, 8, 534. [Google Scholar] [CrossRef]
  82. Liu, G.; Fu, M.; Li, F.; Fu, W.; Zhao, Z.; Xia, H.; Niu, Y. Tissue-engineered PLLA/gelatine nanofibrous scaffold promoting the phenotypic expression of epithelial and smooth muscle cells for urethral reconstruction. Mater. Sci. Eng. C 2020, 111, 110810. [Google Scholar] [CrossRef]
  83. Chen, Y.; Guo, C.; Manousiouthakis, E.; Wang, X.; Cairns, D.M.; Roh, T.T.; Du, C.; Kaplan, D.L. Bi-Layered Tubular Microfiber Scaffolds as Functional Templates for Engineering Human Intestinal Smooth Muscle Tissue. Adv. Funct. Mater. 2020, 30, 2000543. [Google Scholar] [CrossRef] [PubMed]
  84. Jin, Q.; Fu, Y.; Zhang, G.; Xu, L.; Jin, G.; Tang, L.; Ju, J.; Zhao, W.; Hou, R. Nanofiber electrospinning combined with rotary bioprinting for fabricating small-diameter vessels with endothelium and smooth muscle. Compos. Part B Eng. 2022, 234, 109691. [Google Scholar] [CrossRef]
  85. Xu, L.; Varkey, M.; Jorgensen, A.; Ju, J.; Jin, Q.; Park, J.H.; Fu, Y.; Zhang, G.; Ke, D.; Zhao, W.; et al. Bioprinting small diameter blood vessel constructs with an endothelial and smooth muscle cell bilayer in a single step. Biofabrication 2020, 12, 045012. [Google Scholar] [CrossRef] [PubMed]
  86. Méndez-barbero, N.; Gutiérrez-muñoz, C.; Blanco-Colio, L.M. Cellular crosstalk between endothelial and smooth muscle cells in vascular wall remodeling. Int. J. Mol. Sci. 2021, 22, 7284. [Google Scholar] [CrossRef]
  87. Heydarkhan-Hagvall, S.; Helenius, G.; Johansson, B.R.; Li, J.Y.; Mattsson, E.; Risberg, B. Co-Culture of Endothelial Cells and Smooth Muscle Cells Affects Gene Expression of Angiogenic Factors. J. Cell. Biochem. 2003, 1259, 1250–1259. [Google Scholar] [CrossRef]
  88. Williams, C.; Wick, T. Endothelial Cell—Smooth Muscle Cell Co-Culture in a Perfusion Bioreactor System. Ann. Biomed. Eng. 2005, 33, 920–928. [Google Scholar] [CrossRef]
  89. Saunders, S.K.; Cole, S.Y.; Sierra, V.A.; Bracamonte, J.H.; Toldo, S.; Soares, J.S. Evaluation of perfusion-driven cell seeding of small diameter engineered tissue vascular grafts with a custom-designed seed-and-culture bioreactor. PLoS ONE 2022, 17, e0269499. [Google Scholar] [CrossRef]
  90. Sun, L.; Li, X.; Yang, T.; Lu, T.; Du, P.; Jing, C.; Chen, Z.; Lin, F.; Zhao, G.; Zhao, L. Construction of spider silk protein small-caliber tissue engineering vascular grafts based on dynamic culture and its performance evaluation. J. Biomed. Mater. Res. Part A 2023, 111, 71–87. [Google Scholar] [CrossRef]
  91. Dezawa, M.; Ishikawa, H.; Itokazu, Y.; Yoshihara, T.; Hoshino, M.; Takeda, S.I.; Ide, C.; Nabeshima, Y.I. Bone marrow stromal cells generate muscle cells and repair muscle degeneration. Science 2005, 309, 314–317. [Google Scholar] [CrossRef] [PubMed]
  92. de la Garza-Rodea, A.S.; van der Velde, I.; Boersma, H.; Gonçalves, M.A.F.V.; van Bekkum, D.W.; de Vries, A.A.F.; Knaän-Shanzer, S. Long-term contribution of human bone marrow mesenchymal stromal cells to skeletal muscle regeneration in mice. Cell Transplant. 2011, 20, 217–231. [Google Scholar] [CrossRef] [PubMed]
  93. De Bari, C.; Dell’Accio, F.; Vandenabeele, F.; Vermeesch, J.R.; Raymackers, J.M.; Luyten, F.P. Skeletal muscle repair by adult human mesenchymal stem cells from synovial membrane. J. Cell Biol. 2003, 160, 909–918. [Google Scholar] [CrossRef]
  94. Rodriguez, A.M.; Pisani, D.; Dechesne, C.A.; Turc-Carel, C.; Kurzenne, J.Y.; Wdziekonski, B.; Villageois, A.; Bagnis, C.; Breittmayer, J.P.; Groux, H.; et al. Transplantation of a multipotent cell population from human adipose tissue induces dystrophin expression in the immunocompetent mdx mouse. J. Exp. Med. 2005, 201, 1397–1405. [Google Scholar] [CrossRef]
  95. Meligy, F.Y.; Shigemura, K.; Behnsawy, H.M.; Fujisawa, M.; Kawabata, M.; Shirakawa, T. The efficiency of in vitro isolation and myogenic differentiation of MSCs derived from adipose connective tissue, bone marrow, and skeletal muscle tissue. In Vitro Cell. Dev. Biol. Anim. 2012, 48, 203–215. [Google Scholar] [CrossRef] [PubMed]
  96. Mishra, S.; Sevak, J.K.; Das, A.; Arimbasseri, G.A.; Bhatnagar, S.; Gopinath, S.D. Umbilical cord tissue is a robust source for mesenchymal stem cells with enhanced myogenic differentiation potential compared to cord blood. Sci. Rep. 2020, 10, 18978. [Google Scholar] [CrossRef]
  97. Witt, R.; Weigand, A.; Boos, A.M.; Cai, A.; Dippold, D.; Boccaccini, A.R.; Schubert, D.W.; Hardt, M.; Lange, C.; Arkudas, A.; et al. Mesenchymal stem cells and myoblast differentiation under HGF and IGF-1 stimulation for 3D skeletal muscle tissue engineering. BMC Cell Biol. 2017, 18, 15. [Google Scholar] [CrossRef]
  98. 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]
  99. Pereira, A.R.S.; Mendes, T.F.; Ministro, A.; Teixeira, M.; Filipe, M.; Santos, J.M.; Bárcia, R.N.; Goyri-O’Neill, J.; Pinto, F.; Cruz, P.E.; et al. Therapeutic angiogenesis induced by human umbilical cord tissue-derived mesenchymal stromal cells in a murine model of hindlimb ischemia. Stem Cell Res. Ther. 2016, 7, 145. [Google Scholar] [CrossRef]
  100. Testa, S.; Riera, C.S.; Fornetti, E.; Riccio, F.; Fuoco, C.; Bernardini, S.; Baldi, J.; Costantini, M.; Foddai, M.L.; Cannata, S.; et al. Skeletal Muscle-Derived Human Mesenchymal Stem Cells: Influence of Different Culture Conditions on Proliferative and Myogenic Capabilities. Front. Physiol. 2020, 11, 553198. [Google Scholar] [CrossRef]
  101. Zhang, Y.; Zhu, Y.; Li, Y.; Cao, J.; Zhang, H.; Chen, M.; Wang, L.; Zhang, C. Long-term engraftment of myogenic progenitors from adipose-derived stem cells and muscle regeneration in dystrophic mice. Hum. Mol. Genet. 2015, 24, 6029–6040. [Google Scholar] [CrossRef] [PubMed]
  102. Yilgor Huri, P.; Cook, C.A.; Hutton, D.L.; Goh, B.C.; Gimble, J.M.; DiGirolamo, D.J.; Grayson, W.L. Biophysical cues enhance myogenesis of human adipose derived stem/stromal cells. Biochem. Biophys. Res. Commun. 2013, 438, 180–185. [Google Scholar] [CrossRef] [PubMed]
  103. Peçanha, R.; Bagno, L.D.L.E.S.; Ribeiro, M.B.; Ferreira, A.B.R.; Moraes, M.O.; Zapata-Sudo, G.; Kasai-Brunswick, T.H.; Campos-de-Carvalho, A.C.; Goldenberg, R.C.D.S.; Werneck-de-Castro, J.P.S. Adipose-derived stem-cell treatment of skeletal muscle injury. J. Bone Jt. Surg. Am. 2012, 94, 609–617. [Google Scholar] [CrossRef] [PubMed]
  104. Goudenege, S.; Pisani, D.F.; Wdziekonski, B.; Di Santo, J.P.; Bagnis, C.; Dani, C.; Dechesne, C.A. Enhancement of myogenic and muscle repair capacities of human adipose-derived stem cells with forced expression of MyoD. Mol. Ther. 2009, 17, 1064–1072. [Google Scholar] [CrossRef]
  105. Pourquié, O.; Al Tanoury, Z.; Chal, J. The Long Road to Making Muscle In Vitro. Curr. Top. Dev. Biol. 2018, 129, 123–142. [Google Scholar] [CrossRef]
  106. Iberite, F.; Gruppioni, E.; Ricotti, L. Skeletal muscle differentiation of human iPSCs meets bioengineering strategies: Perspectives and challenges. NPJ Regen. Med. 2022, 7, 23. [Google Scholar] [CrossRef]
  107. Darabi, R.; Santos, F.N.C.; Filareto, A.; Pan, W.; Koene, R.; Rudnicki, M.A.; Kyba, M.; Perlingeiro, R.C.R. Assessment of the myogenic stem cell compartment following transplantation of Pax3/Pax7-induced embryonic stem cell-derived progenitors. Stem Cells 2011, 29, 777–790. [Google Scholar] [CrossRef]
  108. Young, C.S.; Hicks, M.R.; Ermolova, N.V.; Nakano, H.; Jan, M.; Younesi, S.; Karumbayaram, S.; Kumagai-Cresse, C.; Wang, D.; Zack, J.A.; et al. A Single CRISPR-Cas9 Deletion Strategy that Targets the Majority of DMD Patients Restores Dystrophin Function in hiPSC-Derived Muscle Cells. Cell Stem Cell 2016, 18, 533–540. [Google Scholar] [CrossRef]
  109. Shelton, M.; Kocharyan, A.; Liu, J.; Skerjanc, I.S.; Stanford, W.L. Robust generation and expansion of skeletal muscle progenitors and myocytes from human pluripotent stem cells. Methods 2016, 101, 73–84. [Google Scholar] [CrossRef]
  110. Hicks, M.; Pyle, A. The Path from Pluripotency to Skeletal Muscle: Developmental Myogenesis Guides the Way. Cell Stem Cell 2015, 17, 255–257. [Google Scholar] [CrossRef]
  111. Chal, J.; Al Tanoury, Z.; Hestin, M.; Gobert, B.; Aivio, S.; Hick, A.; Cherrier, T.; Nesmith, A.P.; Parker, K.K.; Pourquié, O. Generation of human muscle fibers and satellite-like cells from human pluripotent stem cells in vitro. Nat. Protoc. 2016, 11, 1833–1850. [Google Scholar] [CrossRef]
  112. Fang, J.; Sia, J.; Soto, J.; Wang, P.; Li, L.A.K.; Hsueh, Y.Y.; Sun, R.; Faull, K.F.; Tidball, J.G.; Li, S. Skeletal muscle regeneration via the chemical induction and expansion of myogenic stem cells in situ or in vitro. Nat. Biomed. Eng. 2021, 5, 864–879. [Google Scholar] [CrossRef]
  113. Chal, J.; Pourquié, O. Making muscle: Skeletal myogenesis in vivo and in vitro. Development 2017, 144, 2104–2122. [Google Scholar] [CrossRef] [PubMed]
  114. Chal, J.; Oginuma, M.; Al Tanoury, Z.; Gobert, B.; Sumara, O.; Hick, A.; Bousson, F.; Zidouni, Y.; Mursch, C.; Moncuquet, P.; et al. Differentiation of pluripotent stem cells to muscle fiber to model Duchenne muscular dystrophy. Nat. Biotechnol. 2015, 33, 962–969. [Google Scholar] [CrossRef] [PubMed]
  115. Hicks, M.R.; Hiserodt, J.; Paras, K.; Fujiwara, W.; Eskin, A.; Jan, M.; Xi, H.; Young, C.S.; Evseenko, D.; Nelson, S.F.; et al. ERBB3 and NGFR mark a distinct skeletal muscle progenitor cell in human development and hPSCs. Nat. Cell Biol. 2017, 20, 46–57. [Google Scholar] [CrossRef] [PubMed]
  116. Wu, J.; Matthias, N.; Lo, J.; Ortiz-Vitali, J.L.; Shieh, A.W.; Wang, S.H.; Darabi, R. A Myogenic Double-Reporter Human Pluripotent Stem Cell Line Allows Prospective Isolation of Skeletal Muscle Progenitors. Cell Rep. 2018, 25, 1966–1981. [Google Scholar] [CrossRef]
  117. Borchin, B.; Chen, J.; Barberi, T. Derivation and FACS-mediated purification of PAX3+/PAX7+ skeletal muscle precursors from human pluripotent stem cells. Stem Cell Rep. 2013, 1, 620–631. [Google Scholar] [CrossRef] [PubMed]
  118. Choi, I.Y.; Lim, H.T.; Estrellas, K.; Mula, J.; Cohen, T.V.; Zhang, Y.; Donnelly, C.J.; Richard, J.P.; Kim, Y.J.; Kim, H.; et al. Concordant but Varied Phenotypes among Duchenne Muscular Dystrophy Patient-Specific Myoblasts Derived using a Human iPSC-Based Model. Cell Rep. 2016, 15, 2301–2312. [Google Scholar] [CrossRef] [PubMed]
  119. Kim, J.; Magli, A.; Chan, S.S.K.; Oliveira, V.K.P.; Wu, J.; Darabi, R.; Kyba, M.; Perlingeiro, R.C.R. Expansion and Purification Are Critical for the Therapeutic Application of Pluripotent Stem Cell-Derived Myogenic Progenitors. Stem Cell Rep. 2017, 9, 12–22. [Google Scholar] [CrossRef]
  120. van der Wal, E.; Herrero-Hernandez, P.; Wan, R.; Broeders, M.; in ’t Groen, S.L.M.; van Gestel, T.J.M.; van IJcken, W.F.J.; Cheung, T.H.; van der Ploeg, A.T.; Schaaf, G.J.; et al. Large-Scale Expansion of Human iPSC-Derived Skeletal Muscle Cells for Disease Modeling and Cell-Based Therapeutic Strategies. Stem Cell Rep. 2018, 10, 1975–1990. [Google Scholar] [CrossRef]
  121. Jin, Y.; Shahriari, D.; Jeon, E.J.; Park, S.; Choi, Y.S.; Back, J.; Lee, H.; Anikeeva, P.; Cho, S.W. Functional Skeletal Muscle Regeneration with Thermally Drawn Porous Fibers and Reprogrammed Muscle Progenitors for Volumetric Muscle Injury. Adv. Mater. 2021, 33, 2007946. [Google Scholar] [CrossRef] [PubMed]
  122. Zhu, P.; Zhou, Y.; Wu, F.; Hong, Y.; Wang, X.; Shekhawat, G.; Mosenson, J.; Wu, W.S. Selective Expansion of Skeletal Muscle Stem Cells from Bulk Muscle Cells in Soft Three-Dimensional Fibrin Gel. Stem Cells Transl. Med. 2017, 6, 1412. [Google Scholar] [CrossRef] [PubMed]
  123. Zahari, N.K.; Idrus, R.B.H.; Chowdhury, S.R. Laminin-coated poly(Methyl methacrylate) (PMMA) nanofiber scaffold facilitates the enrichment of skeletal muscle myoblast population. Int. J. Mol. Sci. 2017, 18, 2242. [Google Scholar] [CrossRef]
  124. Yoshioka, K.; Kitajima, Y.; Okazaki, N.; Chiba, K.; Yonekura, A.; Ono, Y. A Modified Pre-plating Method for High-Yield and High-Purity Muscle Stem Cell Isolation From Human/Mouse Skeletal Muscle Tissues. Front. Cell Dev. Biol. 2020, 8, 793. [Google Scholar] [CrossRef]
  125. Khodabukus, A.; Madden, L.; Prabhu, N.K.; Koves, T.R.; Jackman, C.P.; Muoio, D.M.; Bursac, N. Electrical stimulation increases hypertrophy and metabolic flux in tissue-engineered human skeletal muscle. Biomaterials 2019, 198, 259–269. [Google Scholar] [CrossRef] [PubMed]
  126. Kulesza, A.; Burdzinska, A.; Szczepanska, I.; Zarychta-Wisniewska, W.; Pajak, B.; Bojarczuk, K.; Dybowski, B.; Paczek, L. The Mutual Interactions between Mesenchymal Stem Cells and Myoblasts in an Autologous Co-Culture Model. PLoS ONE 2016, 11, e0161693. [Google Scholar] [CrossRef]
  127. Cai, A.; Hardt, M.; Schneider, P.; Schmid, R.; Lange, C.; Dippold, D.; Schubert, D.W.; Boos, A.M.; Weigand, A.; Arkudas, A.; et al. Myogenic differentiation of primary myoblasts and mesenchymal stromal cells under serum-free conditions on PCL-collagen I-nanoscaffolds. BMC Biotechnol. 2018, 18, 75. [Google Scholar] [CrossRef]
  128. Juhas, M.; Abutaleb, N.; Wang, J.T.; Ye, J.; Shaikh, Z.; Sriworarat, C.; Qian, Y.; Bursac, N. Incorporation of macrophages into engineered skeletal muscle enables enhanced muscle regeneration. Nat. Biomed. Eng. 2018, 2, 942–954. [Google Scholar] [CrossRef] [PubMed]
  129. Ho, A.T.V.; Palla, A.R.; Blake, M.R.; Yucel, N.D.; Wang, Y.X.; Magnusson, K.E.G.; Holbrook, C.A.; Kraft, P.E.; Delp, S.L.; Blau, H.M. Prostaglandin E2 is essential for efficacious skeletal muscle stem-cell function, augmenting regeneration and strength. Proc. Natl. Acad. Sci. USA 2017, 114, 6675–6684. [Google Scholar] [CrossRef]
  130. Fu, X.; Xiao, J.; Wei, Y.; Li, S.; Liu, Y.; Yin, J.; Sun, K.; Sun, H.; Wang, H.; Zhang, Z.; et al. Combination of inflammation-related cytokines promotes long-term muscle stem cell expansion. Cell Res. 2015, 25, 655–673. [Google Scholar] [CrossRef]
  131. Le Grand, F.; Jones, A.E.; Seale, V.; Scimè, A.; Rudnicki, M.A. Wnt7a activates the planar cell polarity pathway to drive the symmetric expansion of satellite stem cells. Cell Stem Cell 2009, 4, 535–547. [Google Scholar] [CrossRef] [PubMed]
  132. Kino-Oka, M.; Chowdhury, S.R.; Muneyuki, Y.; Manabe, M.; Saito, A.; Sawa, Y.; Taya, M. Automating the Expansion Process of Human Skeletal Muscle Myoblasts with Suppression of Myotube Formation. Tissue Eng. Part C Methods 2009, 15, 717–728. [Google Scholar] [CrossRef] [PubMed]
  133. Ishii, K.; Sakurai, H.; Suzuki, N.; Mabuchi, Y.; Sekiya, I.; Sekiguchi, K.; Akazawa, C. Recapitulation of Extracellular LAMININ Environment Maintains Stemness of Satellite Cells In Vitro. Stem Cell Rep. 2018, 10, 568–582. [Google Scholar] [CrossRef] [PubMed]
  134. Gilbert, P.M.; Havenstrite, K.L.; Magnusson, K.E.G.; Sacco, A.; Leonardi, N.A.; Kraft, P.; Nguyen, N.K.; Thrun, S.; Lutolf, M.P.; Blau, H.M. Substrate elasticity regulates skeletal muscle stem cell self-renewal in culture. Science 2010, 329, 1078. [Google Scholar] [CrossRef]
  135. Wolf, M.T.; Dearth, C.L.; Sonnenberg, S.B.; Loboa, E.G.; Badylak, S.F. Naturally derived and synthetic scaffolds for skeletal muscle reconstruction. Adv. Drug Deliv. Rev. 2015, 84, 208–221. [Google Scholar] [CrossRef]
  136. Han, W.M.; Anderson, S.E.; Mohiuddin, M.; Barros, D.; Nakhai, S.A.; Shin, E.; Amaral, I.F.; Pêgo, A.P.; García, A.J.; Jang, Y.C. Synthetic matrix enhances transplanted satellite cell engraftment in dystrophic and aged skeletal muscle with comorbid trauma. Sci. Adv. 2018, 4, eaar4008. [Google Scholar] [CrossRef]
  137. Bouchentouf, M.; Skuk, D.; Tremblay, J.P. Early and massive death of myoblasts transplanted into skeletal muscle: Responsible factors and potential solutions. Curr. Opin. Organ Transplant. 2007, 12, 664–667. [Google Scholar] [CrossRef]
  138. Peters, K.M.; Dmochowski, R.R.; Carr, L.K.; Robert, M.; Kaufman, M.R.; Sirls, L.T.; Herschorn, S.; Birch, C.; Kultgen, P.L.; Chancellor, M.B. Autologous muscle derived cells for treatment of stress urinary incontinence in women. J. Urol. 2014, 192, 469–476. [Google Scholar] [CrossRef]
  139. Kuismanen, K.; Sartoneva, R.; Haimi, S.; Mannerström, B.; Tomás, E.; Miettinen, S.; Nieminen, K. Autologous Adipose Stem Cells in Treatment of Female Stress Urinary Incontinence: Results of a Pilot Study. Stem Cells Transl. Med. 2014, 3, 936. [Google Scholar] [CrossRef]
  140. Gotoh, M.; Yamamoto, T.; Kato, M.; Majima, T.; Toriyama, K.; Kamei, Y.; Matsukawa, Y.; Hirakawa, A.; Funahashi, Y. Regenerative treatment of male stress urinary incontinence by periurethral injection of autologous adipose-derived regenerative cells: 1-year outcomes in 11 patients. Int. J. Urol. 2014, 21, 294–300. [Google Scholar] [CrossRef]
  141. Yamamoto, T.; Gotoh, M.; Kato, M.; Majima, T.; Toriyama, K.; Kamei, Y.; Iwaguro, H.; Matsukawa, Y.; Funahashi, Y. Periurethral injection of autologous adipose-derived regenerative cells for the treatment of male stress urinary incontinence: Report of three initial cases. Int. J. Urol. 2012, 19, 652–659. [Google Scholar] [CrossRef]
  142. Gussoni, E.; Pavlath, G.K.; Lanctot, A.M.; Sharma, K.R.; Miller, R.G.; Steinman, L.; Blau, H.M. Normal dystrophin transcripts detected in Duchenne muscular dystrophy patients after myoblast transplantation. Nature 1992, 356, 435–438. [Google Scholar] [CrossRef] [PubMed]
  143. Law, P.K.; Goodwin, T.G.; Fang, Q.; Duggirala, V.; Larkin, C.; Florendo, J.A.; Kirby, D.S.; Deering, M.B.; Li, H.J.; Chen, M.; et al. Feasibility, safety, and efficacy of myoblast transfer therapy on Duchenne muscular dystrophy boys. Cell Transplant. 1992, 1, 235–244. [Google Scholar] [CrossRef] [PubMed]
  144. Périé, S.; Trollet, C.; Mouly, V.; Vanneaux, V.; Mamchaoui, K.; Bouazza, B.; Marolleau, J.P.; Laforêt, P.; Chapon, F.; Eymard, B.; et al. Autologous Myoblast Transplantation for Oculopharyngeal Muscular Dystrophy: A Phase I/Iia Clinical Study. Mol. Ther. 2014, 22, 219. [Google Scholar] [CrossRef] [PubMed]
  145. Carr, L.K.; Robert, M.; Kultgen, P.L.; Herschorn, S.; Birch, C.; Murphy, M.; Chancellor, M.B. Autologous muscle derived cell therapy for stress urinary incontinence: A prospective, dose ranging study. J. Urol. 2013, 189, 595–601. [Google Scholar] [CrossRef]
  146. Sharifiaghdas, F.; Zohrabi, F.; Moghadasali, R.; Shekarchian, S.; Jaroughi, N.; Bolurieh, T.; Baharvand, H.; Aghdami, N. Autologous Muscle-derived Cell Injection for Treatment of Female Stress Urinary Incontinence: A Single- Arm Clinical Trial with 24-months Follow-Up. Urol. J. 2019, 16, 482–487. [Google Scholar] [CrossRef] [PubMed]
  147. Gräs, S.; Klarskov, N.; Lose, G. Intraurethral injection of autologous minced skeletal muscle: A simple surgical treatment for stress urinary incontinence. J. Urol. 2014, 192, 850–855. [Google Scholar] [CrossRef]
  148. Gerullis, H.; Eimer, C.; Georgas, E.; Homburger, M.; El-Baz, A.G.; Wishahi, M.; Borós, M.; Ecke, T.H.; Otto, T. Muscle-derived cells for treatment of iatrogenic sphincter damage and urinary incontinence in men. Sci. World J. 2012, 2012, 898535. [Google Scholar] [CrossRef] [PubMed]
  149. Mitterberger, M.; Marksteiner, R.; Margreiter, E.; Pinggera, G.M.; Frauscher, F.; Ulmer, H.; Fussenegger, M.; Bartsch, G.; Strasser, H. Myoblast and fibroblast therapy for post-prostatectomy urinary incontinence: 1-year followup of 63 patients. J. Urol. 2008, 179, 226–231. [Google Scholar] [CrossRef] [PubMed]
  150. Sèbe, P.; Doucet, C.; Cornu, J.N.; Ciofu, C.; Costa, P.; De Medina, S.G.D.; Pinset, C.; Haab, F. Intrasphincteric injections of autologous muscular cells in women with refractory stress urinary incontinence: A prospective study. Int. Urogynecol. J. 2011, 22, 183–189. [Google Scholar] [CrossRef]
  151. Atala, A.; Bauer, S.B.; Soker, S.; Yoo, J.J.; Retik, A.B. Tissue-engineered autologous bladders for patients needing cystoplasty. Lancet 2006, 367, 1241–1246. [Google Scholar] [CrossRef]
  152. Raya-Rivera, A.; Esquiliano, D.R.; Yoo, J.J.; Lopez-Bayghen, E.; Soker, S.; Atala, A. Tissue-engineered autologous urethras for patients who need reconstruction: An observational study. Lancet 2011, 377, 1175–1182. [Google Scholar] [CrossRef]
  153. Kang, M.S.; Lee, S.H.; Park, W.J.; Lee, J.E.; Kim, B.; Han, D.W. Advanced techniques for skeletal muscle tissue engineering and regeneration. Bioengineering 2020, 7, 99. [Google Scholar] [CrossRef] [PubMed]
  154. Volpi, M.; Paradiso, A.; Costantini, M.; Świȩszkowski, W. Hydrogel-Based Fiber Biofabrication Techniques for Skeletal Muscle Tissue Engineering. ACS Biomater. Sci. Eng. 2022, 8, 379–405. [Google Scholar] [CrossRef] [PubMed]
  155. Cai, Z.; Liu, D.; Yang, Y.; Xie, W.; He, M.; Yu, D.; Wu, Y.; Wang, X.; Xiao, W.; Li, Y. The role and therapeutic potential of stem cells in skeletal muscle in sarcopenia. Stem Cell Res. Ther. 2022, 13, 28. [Google Scholar] [CrossRef] [PubMed]
  156. Jovic, D.; Yu, Y.; Wang, D.; Wang, K.; Li, H.; Xu, F.; Liu, C.; Liu, J.; Luo, Y. A Brief Overview of Global Trends in MSC-Based Cell Therapy. Stem Cell Rev. Rep. 2022, 18, 1525–1545. [Google Scholar] [CrossRef] [PubMed]
  157. Eugenis, I.; Wu, D.; Rando, T.A. Cells, scaffolds, and bioactive factors: Engineering strategies for improving regeneration following volumetric muscle loss. Biomaterials 2021, 278, 121173. [Google Scholar] [CrossRef]
Figure 2. Considerations on the key points for SMC and SkMC manufacturing. From selection of cell source to culture setup, with potential combination of biomaterials. In terms of cell source, MSCs and iPSCs present broader availability and higher expansion potential, though with a limited maturation phenotype of the target cell type. Due to their low immunogenic profile, MSCs can be used in an allogeneic therapeutic setting, in contrast to primary isolated muscle cells. Concerning culture systems, a balance between complexity (e.g., cocultures that need to accommodate more than one cell type) and feasibility must be considered, with mechanical stimulus representing a critical aspect in muscle tissue engineering. Scaffold design and selection should take into account multiple characteristics such as its composition and stiffness, and if it targets in vitro/ex vivo use only, or if it is intended to be used as a cell delivery vehicle in vivo or as an architectural implanted scaffold. Adapted from [155,156,157]. ECs: endothelial cells; iPSCs: induced pluripotent stem cells; MSCs: mesenchymal stromal cells; SkMCs: skeletal muscle cells; SMCs: smooth muscle cells.
Figure 2. Considerations on the key points for SMC and SkMC manufacturing. From selection of cell source to culture setup, with potential combination of biomaterials. In terms of cell source, MSCs and iPSCs present broader availability and higher expansion potential, though with a limited maturation phenotype of the target cell type. Due to their low immunogenic profile, MSCs can be used in an allogeneic therapeutic setting, in contrast to primary isolated muscle cells. Concerning culture systems, a balance between complexity (e.g., cocultures that need to accommodate more than one cell type) and feasibility must be considered, with mechanical stimulus representing a critical aspect in muscle tissue engineering. Scaffold design and selection should take into account multiple characteristics such as its composition and stiffness, and if it targets in vitro/ex vivo use only, or if it is intended to be used as a cell delivery vehicle in vivo or as an architectural implanted scaffold. Adapted from [155,156,157]. ECs: endothelial cells; iPSCs: induced pluripotent stem cells; MSCs: mesenchymal stromal cells; SkMCs: skeletal muscle cells; SMCs: smooth muscle cells.
Bioengineering 10 01067 g002
Table 3. Summary of cell types used in major clinical trials targeting muscle repair and regeneration.
Table 3. Summary of cell types used in major clinical trials targeting muscle repair and regeneration.
Cell TypeCulture ConditionsDisease/ApplicationNumber of PatientsMain ObservationsYearReference
Myoblasts-Duchenne muscular dystrophy8No dystrophin expression restored;
No significant strength recovery.
1992[142]
MyoblastsDMEM with 10% horse serumDuchenne muscular dystrophy21No significant strength recovery.1992[143]
MyoblastsFBS and FGF-2Oculopharyngeal muscular dystrophy12Safety tested;
Quality of life improved.
2014[144]
SkM-derived cellsProprietary methodUrinary incontinence38Safety tested;
No major adverse events.
2013[145]
SkM-derived cellsProprietary methodUrinary incontinence80Safety profile tested;
Improvement in stress incontinence symptoms.
2014[138]
SkM-derived cellsHam’s F10 with 20% FBSUrinary incontinence20Partial response;
Symptoms relapse after 2 years in 50% of the initial responders.
2019[146]
SkM (minced tissue)Without ex vivo cultureUrinary incontinence35Symptoms improved in 25–63% of patients;
Minor adverse events.
2014[147]
SkM-derived cellsDMEM/F12 and 10% fetal calf serumDamaged urethral sphincter222After 1 year, in 46% patients, no therapeutic effect;
42% reported improvement of symptoms and in 12% urinary continence was restored.
2012[148]
Myoblasts (and fibroblasts)DMEM/F12 with 20% autologous serumUrinary incontinence after prostatectomy631 year follow-up shows restored continence in 41 patients;
5 patients did not show improvement.
2008[149]
Progenitor SkM cells-Urinary incontinence12Quality of life improvement reported.2010[150]
SMCs (and urothelial cells)DMEM with 10% fetal calf serumCystoplasty756% of patients showed signs of enhanced bladder function.2006[151]
SMCs (and epithelial cells)DMEM with EGFUrethral reconstruction5Histology showed that engineered constructs integrated patient’s tissue.2011[152]
DMEM: Dulbecco’s modified Eagle’s medium; FBS: fetal bovine serum; EGF: epidermal growth factor; FGF: fibroblast growth factor; SMCs: smooth muscle cells; SkM: skeletal muscle.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Franchi-Mendes, T.; Silva, M.; Cartaxo, A.L.; Fernandes-Platzgummer, A.; Cabral, J.M.S.; da Silva, C.L. Bioprocessing Considerations towards the Manufacturing of Therapeutic Skeletal and Smooth Muscle Cells. Bioengineering 2023, 10, 1067. https://doi.org/10.3390/bioengineering10091067

AMA Style

Franchi-Mendes T, Silva M, Cartaxo AL, Fernandes-Platzgummer A, Cabral JMS, da Silva CL. Bioprocessing Considerations towards the Manufacturing of Therapeutic Skeletal and Smooth Muscle Cells. Bioengineering. 2023; 10(9):1067. https://doi.org/10.3390/bioengineering10091067

Chicago/Turabian Style

Franchi-Mendes, Teresa, Marília Silva, Ana Luísa Cartaxo, Ana Fernandes-Platzgummer, Joaquim M. S. Cabral, and Cláudia L. da Silva. 2023. "Bioprocessing Considerations towards the Manufacturing of Therapeutic Skeletal and Smooth Muscle Cells" Bioengineering 10, no. 9: 1067. https://doi.org/10.3390/bioengineering10091067

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