Next Article in Journal
Monocytic Cell Adhesion to Oxidised Ligands: Relevance to Cardiovascular Disease
Next Article in Special Issue
Structural and Biomechanical Properties of Supramolecular Nanofiber-Based Hydrogels in Biomedicine
Previous Article in Journal
Lipedema: Insights into Morphology, Pathophysiology, and Challenges
Previous Article in Special Issue
De Novo Self-Assembling Peptides Mediate the Conversion of Temozolomide and Delivery of a Model Drug into Glioblastoma Multiforme Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Tissue Engineering as a Promising Treatment for Glottic Insufficiency: A Review on Biomolecules and Cell-Laden Hydrogel

1
Department of Otorhinolaryngology-Head and Neck Surgery, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
2
Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
3
Department of Obstetrics and Gynaecology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
*
Author to whom correspondence should be addressed.
Biomedicines 2022, 10(12), 3082; https://doi.org/10.3390/biomedicines10123082
Submission received: 29 September 2022 / Revised: 11 November 2022 / Accepted: 16 November 2022 / Published: 30 November 2022

Abstract

:
Glottic insufficiency is widespread in the elderly population and occurs as a result of secondary damage or systemic disease. Tissue engineering is a viable treatment for glottic insufficiency since it aims to restore damaged nerve tissue and revitalize aging muscle. After injection into the biological system, injectable biomaterial delivers cost- and time-effectiveness while acting as a protective shield for cells and biomolecules. This article focuses on injectable biomaterials that transport cells and biomolecules in regenerated tissue, particularly adipose, muscle, and nerve tissue. We propose Wharton’s Jelly mesenchymal stem cells (WJMSCs), induced pluripotent stem cells (IP-SCs), basic fibroblast growth factor (bFGF), vascular endothelial growth factor (VEGF), hepatocyte growth factor (HGF), insulin growth factor-1 (IGF-1) and extracellular vesicle (EV) as potential cells and macromolecules to be included into biomaterials, with some particular testing to support them as a promising translational medicine for vocal fold regeneration.

1. Introduction

Voice disorders affect 16.9% of the adult population (aged 18 and more) and 13.1% more of the elderly population (aged 85 and more) [1,2,3]. Glottic insufficiency is diagnosed when the vocal fold does not entirely close during phonation [4]. It impairs voice production and the protection of the lower airway, resulting in impaired social function, decreased work performance, and an increased risk of aspiration. Due to the unique characteristics of the human vocal fold and the numerous causes of glottic insufficiency, it is difficult to recommend the optimal treatment for this illness. Tissue engineering has advantages in this field since it strives to enhance regeneration and provides longer-lasting or even permanent vocal fold augmentation [5,6]. Tissue engineering has been researched extensively in several regenerative techniques, including cartilage, neuron, cardiac, and bone regeneration [7,8,9,10]. Nonetheless, previous studies [11,12,13,14] have identified vocal fold fibroblasts, muscle progenitor cells, embryonic stem cells (ESCs), bone marrow mesenchymal stem cells (BMMSCs), and adipose stem cells (ASCs) with or without the use of a scaffold as a delivery vehicle for vocal fold regeneration. This study seeks to outline the most recent advancements in injectable biomaterials that transport biomolecules and cells for regeneration purposes and to identify future directions for tissue engineering–based treatment of glottic insufficiency.

1.1. Structure of Vocal Fold

Three layers—cover, transition, and body—are thought to make up a human vocal fold [15]. Collagen, elastin, hyaluronic acid (HA), decorin, and fibronectin make up the majority of the extracellular matrix (ECM) proteins found in the lamina propria [16]. The superficial layers of lamina propria and epithelium in the cover have vibratory qualities which are crucial for phonation. Superficial lamina propria comprises loosely packed connective tissue [17]. The transition layer is made up of an intermediate layer primarily of elastin and deep layer of collagen. Collagen provides mechanical support to the vocal fold while elastin maintains the elasticity of the vocal fold [18,19]. The vocalis muscle makes up the body layer, which forms the base of this intricate three-dimensional structure [15]. Interestingly, cell junctions hold stratified squamous epithelium, which serves as a protective layer around membranous vocal folds [20]. Compared to newborn epithelial cells, adult epithelial cells displayed more significant intercellular gaps, greater mechanical strength, and more excellent elasticity [21]. In the lamina propria, the vascular network disperses differently. Only capillaries are seen in the superficial lamina propria; arterioles and venules are located in the intermediate and deep lamina propria. In muscular tissue, bigger vessels are more prevalent. Pericytes have been seen on capillaries, and it is thought that pericytes can shield capillaries from lamina propria vibration. Additionally, pericytes are found to be crucial for angiogenesis [22].
The paraglottic area, which houses intrinsic laryngeal muscle, nerve connections, and adipose tissue, is connected laterally to the vocal fold [23]. Intrinsic laryngeal muscles include thyroarytenoid (TA), lateral-cricoarytenoid (LCA) and interarytenoid (IA) muscles for adduction, and the posterior cricoarytenoid (PCA) muscle for abduction [24]. It is suggested that the TA, cricothyroid (CT), and LCA function as a single muscle stimulated by a motor unit [25]. Recurrent laryngeal nerves (RLN) and superior laryngeal nerves (SLN) provide intrinsic and extrinsic impulses, respectively [26]. RLN perform adduction and abduction functions [27]. The inability to change vocal pitch due to CT motor loss is directly related to SLN dysfunction [28]. The RLN innervates the thyroarytenoid-lateral cricoarytenoid (TA-LCA) for adduction, whereas the SLN innervates the CT. It can be summarized that the composition of the vocal fold includes elastic components which enable its phonation properties, mechanically strong components to support its structure, blood capillaries to provide nutrients to cells, and nerves to control its movement. The structure of the human vocal fold is briefly demonstrated in Figure 1.

1.2. Etiologies of Glottic Insufficiency

It is vital for the clinician to differentiate the causes of glottic insufficiency in opting for suitable treatments. The main symptoms of glottic insufficiency include being unable to generate an effective voice and being unable to protect the lower airway during swallowing. The most common cause of glottic insufficiency is vocal fold paralysis/paresis. Simply put, dysfunctional nerves or muscles are the primary causes of the vocal fold’s inadequate closure. Figure 2 explains the physiological characteristics of the normal condition and glottic insufficiency.
Vocal fold paralysis/paresis is identified when the RLN or SLN are damaged, causing the inability of the intrinsic laryngeal muscles to contract [24,29,30], contributing to the inability to move the vocal fold. Vocal fold paresis is defined when the nerve is partially damaged, causing incomplete signaling or abnormal signaling of nerve; paralysis is diagnosed when the vocal fold is not able to move completely [26]. Vocal fold paresis/paralysis has many etiologies, including scarring, iatrogenic disorders, malignancy, central nervous system pathology, and systemic illnesses [31]. An idiopathic cause is searched for when an aetiology cannot be determined after a comprehensive study.
Vocal fold atrophy is characterized when there is dissipation of muscle and loss of intonation even though the TA-LCA complex is mobile within a certain range [32]. Presbyphonia, child/adolescent, and inborn vocal fold scar are the three distinct types of vocal fold atrophy [33,34,35,36]. Commonly, the decreased sensitivity or malfunction of the contractile components inside the TA muscle is linked to the pathophysiology of vocal fold atrophy [1]. One of the possible causes of an ageing voice is structural changes in the vocal fold’s lamina propria [37]. The lamina propria became stiffer from increased collagen density and decreased elastin and HA density. Additionally, it was discovered that the activity of collagenase decreased in ageing vocal folds [16,38].

2. Current Treatment for Glottic Insufficiency and Limitations

2.1. Surgery and Complementary Treatment

Traditional intervention for glottic insufficiency involves improving glottic closure. As such, type 1 medialization thyroplasty is one of the treatments for glottic insufficiency. It involves implantation of a foreign material into the paraglottic space. It is durable but has some drawbacks, such as a difficult technique and the possibility of vocal fold vibratory function loss over time [39]. Gore-Tex ribbon (polytetrafluoroethylene) and Silastic (silicone rubber) are examples of inert materials used in this surgical procedure [40]. Nonselective laryngeal reinnervation (NSLR) is performed by connecting the ansa cervicalis to the RLN. In observations lasting up to 12 months, NSLR has been proven to improve the voice result of patients considerably [41]. However, NSLR is a technically challenging operation requiring a lengthy process performed under general anesthesia. In patients with bilateral vocal fold immobility, treatment focuses on improving respiration rather than the voice where carbon dioxide laser posterior cordectomy has been described with favorable outcomes [42].
Voice therapy is also widely suggested by practitioners. Previous research assessed voice therapy’s efficacy in RLN and SLN activation in canine vocal fold paresis and paralysis [29]. It was determined that phonation is superior during RLN activation but contracting during SLN activation. Voice therapy and surgical intervention were examined in a separate clinical investigation, demonstrating positive effects on phonation for both arms. However, surgery was performed on patients with more severe symptoms [43].

2.2. In-Office Injection: Injectate Type

Vocal fold injection is less expensive and time-consuming than other procedures. Among materials that have been used for injection are paraffin, Teflon, autologous fat, bovine collagen, carboxymethylcellulose (CMC), calcium hydroxyapatite (CaHA) and HA [44,45]. CaHA could withstand more prolonged periods of augmentation than CMC, and practitioners stated that CaHA injections required more force. Despite their distinct qualities, the voice results are comparable [46].
Fat injection was examined as a treatment for individuals with unilateral vocal fold paralysis (UVFP) and vocal atrophy/scar [47]. This study concluded that fat augmentation is suitable for UVFP since it is autologous and durable but not for vocal fold atrophy/scar because it reduces the vocal range. Nonetheless, fat augmentation in conjunction with platelet-rich plasma (PRP) reduced the recovery period and allergic response in type II sulcus vocalis compared to fat augmentation alone [48]. In adipose tissue engineering, autologous fat was injected into vocal muscle and paraglottic spaces to enhance neovascularization and prevent bulk injection at one site [49]. PRP is rich in various growth factors that can assist in regenerating local tissue and provide anti-inflammatory properties [50,51]. However, direction injection of PRP is not sustainable over time, and it requires re-injection to obtain a sustainable outcome.

3. Tissue Engineering as a Promising Treatment for Glottic Insufficiency

3.1. Tissue Engineering in Vocal Fold Injection

Regenerative medicine is the approach of reinstating human cells, tissue or organs to their usual role [52]. Tissue engineering is application of biomaterial with or without cell transplantation to encourage endogenous regeneration and regain functional tissues or organs [53]. Fillers such as Teflon, polydimethysilicone and calcium hydroxyapatite are commonly injected into the vocal folds to improve glottal closure but are linked with the risk of inflammation, migration, and granuloma development [54]. Moreover, current clinical trial research for vocal fold injection focuses primarily on biomaterial alone (ClinicalTrials.gov number: NCT04700566, NCT03790956, NCT02163772) or direct injection of biomolecules or cells (ClinicalTrials.gov number: NCT05354544, NCT05385159, NCT03749863, NCT02622464, NCT02120781, NCT02904824, NCT04839276). It is suggested that combining biomaterials with biomolecules such as growth factors can improve the efficacy [55]. The combination of biomolecules, cells, and a scaffold serves as a unique delivery system. As cells proliferate to generate new tissue, biomolecules promote the growth of new tissue, and the scaffold serves as an environment for the regeneration of new tissue [56,57]. The comparison between injection of cells and biomolecules without and with a scaffold is shown in Figure 3.
Metals, ceramics, and polymers are the three most common types of scaffold biomaterials [58]. Polymeric hydrogel is often utilized for injecting vocal folds due to its ease of usage, biomimicry, and capacity to produce irregular shapes in confined spaces [59]. Polymeric hydrogel can be fabricated using synthetic or natural molecules [60], as shown in Supplementary Figure S1. It can be crosslinked by either physical or chemical processes. As a result, chemically crosslinked hydrogels exhibited a rapid gelation reaction and may be suited for injection into the vocal folds [61,62]. Besides chemical crosslinking, this study utilized click chemistry to generate a hydrogel capable of reducing the glottal gap in a rabbit model [63]. Thermosensitive hydrogel is also suitable for vocal fold injection since it solidifies at body temperature [64]. Direct injection of cells is a concern, as this delivery method causes ineffective cell survival and retention [65]. Figure 4 demonstrates that integrating cells into hydrogel reduces this issue and prolongs biomolecules’ transport, enhancing the regeneration impact in scarred vocal folds. Hydrogel is excellent for medication delivery due to its longer-lasting release and environmental sensitivity [66]. Hydrogel has a high water content and mimics the milieu of living organisms [67]. Cell encapsulation can be performed either during or after scaffold creation, provided that the process does not harm the cells [68].
Another benefit of the hydrogel as a delivery vehicle is the precise control of its re-release period. The hydrogel can be engineered to degrade at a rate that is commensurate with the time required for tissue regeneration [69]. Cell fate in the hydrogel is closely associated with degradation rate and substrate modification. As hydrogel degrades, stem cells differentiate into mature parent cells such as chondrocytes and osteocytes [67]. Moreover, a biodegradable gelatin hydrogel microsphere was able to progressively release bFGF in a rabbit model with wounded vocal folds [70]. Nevertheless, natural polymeric hydrogel is subject to fast biodegradation due to its low mechanical properties. The mechanical properties of the hydrogel can be finetuned via different crosslinking strategies such as click chemistry, interpenetrating networks, nanocomposites and more [71]. During finetuning of hydrogel, the microenvironment changes attributable to the crosslinking mechanism will impact the reactions of macrophages and fibroblasts [72]. It is very important to develop suitable mechanical properties of hydrogel, which has similar stiffness to that in body tissue and will help to regulate native fibroblast expression [73]. The interaction between an encapsulated cell and hydrogel is insufficiently evidenced; however, cell encapsulation in the hydrogel was able to modify the breakdown rate of hydrogel, as this study demonstrated that hydrogel containing Schwann cells degraded quicker than hydrogel alone [74]. Cell-hydrogel interactions, such as cell adhesion and movement, must be elucidated to prove the viability of hydrogel applications [75]. Cell adhesion in the hydrogel guarantees its capacity to integrate with native tissue throughout the healing process. Having a connection with this, cell adhesion receptors include the tripeptide arginine-glycine-aspartic acid (RGD) by facilitating cell communication via integrin dimers. Other receptors that stimulate cell adhesion include ligands derived from laminin (YIGSR) and collagen (GFOGER) [76]. Cell-hydrogel interaction is also affected by the hydrogel’s physicomechanical properties, such as its rigidity, composition, viscoelasticity, and microenvironment [76,77].
Numerous preliminary studies of hydrogel use for various regenerative reasons have been done. Examples include neurons, wound, tendons, bone, and muscle [78,79,80,81,82,83]. In a 3D hydrogel, Schwann cells express laminin and collagen IV, which may enhance axonal development [74]. A literature research was undertaken to outline the types of cells and macromolecules used to encapsulate in an injectable hydrogel, as shown in Supplementary Figure S2, and to indicate their possible application in vocal fold regeneration. The subject is separated into two subsections: cell encapsulation and biomolecule encapsulation. Combinations of the keywords “hydrogel”, “stem cell”, “growth factor”, “secretome”, “fibroblast”, “cell”, “incorporation”, “encapsulation”, “delivery”, and “cell-laden” were used to search Web of Science (WoS) for relevant publications. There were a total of 17,799 articles found, and irrelevant articles were omitted; 151 articles on injectable biomaterials for biomolecule and cell encapsulation in adipose, angiogenesis, muscle, and nerve regenerative medicine were included.

3.2. Injectable Hydrogel as Cell Delivery Vehicle

The majority of research develops encapsulation techniques for broad regeneration objectives. Muscle regeneration is followed by angiogenesis, nerve regeneration, and adipose tissue engineering in terms of the number of relevant studies. Comparatively, more research tried to encapsulate cells alone in biomaterials, followed by encapsulation of growth factor, MSC-extracellular vehicle (EV), and siRNA (siRNA). As illustrated in Supplementary Figure S3, three major categories of cells were examined.
In clinical studies, direct injection of adipose-derived stem cells (ASCs) improved voice outcomes in patients with vocal fold scarring and glottic insufficiency [84,85]. Numerous in vivo investigations involving direct injection of ASCs have shown that ASCs can upregulate HA while downregulating collagen type I, type III, matrix metalloproteinase (Mmp1), and Mmp8 expression [86,87,88]. ASCs were also intimately linked to the secretion of FGF2, HGF, and basic fibroblast growth factor (bFGF). However, with direct injection, ASCs were only able to survive for 14 days; encapsulation helps to circumvent this problem [89,90]. It is uncertain whether ASCs or BMMSCs are more effective for augmenting vocal folds. Hiwatashi and colleagues recommended ASCs because they would increase HA control more effectively than BMMSCs [91]. Bartlett and colleagues recommended otherwise [92]. By stimulating BMMSCs with transforming growth factor beta (TGF-β), differentiation of vocal fold fibroblast into myofibroblast is inhibited [93]. Few investigations demonstrated that the qualities of ASCs are superior to those of BMMSCs because they are more stable, anti-inflammatory, proliferative, and have the same capacity to differentiate into various lineages [94,95,96,97]. The anti-fibrosis function of ASCs is depicted in Figure 5.
Human umbilical cord-mesenchymal stem cells are also known as Wharton’s Jelly-mesenchymal stem cells (WJMSCs). A WJMSC is a mucous connective tissue in the umbilical cord, which is usually discarded [98]. It is able to differentiate into different types of cells, namely mesoderm (adipocyte, osteocyte, chondrocyte), ectoderm (nerve) and endoderm (islet and liver cells) [99]. The addition of nerve growth factor (NGF) to WJMSCs in a collagen scaffold has been shown to enhance the differentiation and healing of a wounded RLN in rabbits [100]. In addition, WJMSCs have low immunogenicity with respect to T cells, B cells, dendritic cells, natural killer (NK) cells, neutrophils, and mast cells [101]. Insufficient evidence supports the use of WJMSCs in vocal fold augmentation. As it can potentially regenerate myocyte [102,103], nerve cell [104,105,106] and adipocytes, which are abundant in vocal folds, it has a strong potential to repair atrophied or damaged tissue in the vocal fold. With its superior immunomodulatory capabilities, it can prevent inflammation and minimise vocal fold fibrosis. Figure 6 depicts the role of WJMSCs in neuron and muscle regeneration, while Figure 7 summarises their immunomodulatory features.
iPSCs are a relatively novel use for augmenting vocal folds. Previous research indicated its ability to rebuild injured muscle in rat model vocal folds [107]. iPSCs have the potential to develop into epithelial cells and alleviate fibrosis when incorporated into hydrogel [108,109]. iPSCs have been shown to develop into endothelial cells when exposed to growth factors such as activin A, bone morphogenetic protein 4 (BMP4), and bFGF. However, the application of iPSCs in vocal fold augmentation has yielded limited results. Additionally, it can repair skeletal muscle and Schwann cells [110,111]. However, it is known that iPSCs are tumorigenic and carry a high risk for clinical application [112]. In order to resolve this problem, additional investigation and comprehension of its underlying mechanics are required.
Native tissue serves a crucial role in facilitating wound healing. Myofibroblast stimulation from fibroblast is known to enhance the creation of ECM proteins. TGF-1 facilitates the procedure [113]. Epithelial cells, fibroblasts, macrophages, and platelets produce TGF-β. It has activities in both promoting and inhibiting wound healing; for example, it inhibits airway epithelial growth but promotes mucosal remodelling. The vocal fold wound healing process is regulated by epidermal growth factor (EGF) and transforming growth factor beta 1 (TGF-1) [114]. Following injury, epithelial cells increased in thickness and permeability [115]. Branco and his colleague discovered that the lamina propria and epithelial layers of ageing vocal folds tend to atrophy [116]. Therefore, the structural alterations of the vocal fold’s native tissue are essential for maintaining its efficient vibrational state.

3.3. Injectable Hydrogel as Biomolecule Delivery Vehicle

As hydrogel has a special affinity for water, it can be used as a hydrophilic growth factor delivery system. Based on the application (slow and extended or fast and short release), protein retention and delivery can be modified [117]. A hydrogel with low crosslinking, small particle size and susceptibility to enzymatic degradation will result in quicker growth factor release [118]. Growth factor incorporation will increase the bioactivity of the hydrogel [119]. For instance, Walters and colleagues demonstrated [120] that combining platelet-derived growth factor AB (PDGF-AB) and TGF-1 in collagen hydrogel promotes the differentiation of ASCs into smooth muscle cells. The thermosensitive heparin-poloxamer hydrogel containing bFGF and NGF enhances Schwann cell proliferation via the PI3K/Akt, JAK/STAT and MAPK/ERK signalling pathways [121]. To provide multiple regenerative aims, three different types of growth factors, namely VEGF, PDGF and BMP2, were released from a collagen hydrogel over a 28-day period in order to stimulate angiogenesis in a rat model [122].
As described in Supplementary Figure S4, the biomolecules employed in biomaterials can be categorised into four categories: neurotrophic growth factor, growth factor, proteins, and extracellular vesicles. In a rabbit model, a collagen scaffold containing NGF and human umbilical MSC was administered. After eight weeks, a positive response was obtained in the RLN injury model [100]. In vocal fold regeneration, brain-derived neurotrophic factor (BDNF), ciliary neurotrophic factor (CNF), and stromal cell-derived factor-1 (SDF-1) have not been researched. The SDF-1/CXCR4-mediated FAK/PI3K/Akt pathway [123] is thought to protect neuron tissue by avoiding cell death and inflammation. Following nerve damage, BDNF and SDF-1 increase in order to repair and modulate cells [124]. Ciliary neurotrophic factor enhances axon regeneration by binding to ciliary neurotrophic factor receptor α and then activating STAT3 [125]. Glottic insufficiency may have various causes, including muscle atrophy, nerve degeneration or damage, and anatomical alterations to the lamina propria. Consequently, treatment with just nerve growth factor may not yield optimal results.
As the vocal fold is composed of epithelium and fibroblasts, EGF can reconstitute functional mucosa by promoting epithelium regeneration and HA synthesis. Several clinical trials [126,127,128] have demonstrated that direct injection of bFGF into the vocal fold has a beneficial effect on functional voice results. The most effective therapeutic impact on vocal fold atrophy was produced by this treatment. As the majority of instances were caused by ageing, bFGF use was shown to increase fibroblast synthesis of HA, hence lowering collagen deposition It was believed that bFGF treatment was more effective than biomaterial implantation because it altered the vibratory characteristics of the vocal fold and increased its volume [129]. bFGF was not only able to restore the flexibility of the vocal fold but also increased the density of the thyroarytenoid muscle in aged vocal folds [130].
Hepatocyte growth factor (HGF) has anti-fibrotic and angiogenesis properties and is produced by mesenchymal cells such as fibroblasts, macrophages, renal mesangium, etc. A clinical investigation demonstrated the efficacy and regeneration potential of HGF in patients with vocal fold scarring and sulcus. By repeatedly injecting the vocal fold with HGF, the patients exhibited a considerably improved outcome while maintaining a high level of safety. An earlier pre-clinical investigation demonstrated that HGF could boost fibroblast synthesis of HA and decrease collagen deposition [131]. In a second in-vitro study of a damaged vocal fold in rabbits, HGF delivered in a hyaluronic/alginate hydrogel was more effective than HGF injection alone [132]. With immediate treatment of HGF after vocal fold injury, collagen synthesis was decreased and angiogenesis was stimulated [133].
There is paucity of data on the effect of PDGF-BB on vocal fold regeneration. One study, however, showed that PDGF-BB can stimulate vessel development in rat models [134]. Similar to PDGF-BB, angiogenin has not been researched in the regeneration of vocal folds. By blocking the TGF-1/Smad pathway, it has been shown to alter fibroblast scar formation [135]. VEGF stimulates the development of blood vessels, which is essential for tissue regeneration. Then, numerous investigations on VEGF in various areas, including skeletal, peripheral nerve, dental pulp, and heart regeneration, were conducted [136,137,138,139,140]. VEGF promotes the development of HUVEC and neurite cells through the Erk/Akt pathway [141]. IGF-1 is one of the key proteins that regulate skeletal muscle metabolism pathways such as PI3K/Akt/mTOR and PI3K/Akt/GSK3β. IGF-1 suppresses cytokines that produce muscle atrophy and myostatin via these signalling pathways by suppressing nuclear factor-kappa beta (NF-ĸB) and Smad pathways. It can also promote skeletal muscle stem cells for the regeneration of skeletal muscle [142]. Insulin growth factor-1 (IGF-1) was induced by myokine/cytokine Meteorin-like that promotes myogenesis [143].
Researchers are currently interested in the exosome from umbilical cord MSC, since it can prevent tumorigenic concerns. It has great promise for use in a variety of conditions, including wounds, type 2 diabetes, inflammatory bowel disease, Alzheimer’s disease, spinal cord injury, myocardial ischemia injury, and graft-versus-host disease [144]. Extracellular vesicles (EVs) can be subdivided into numerous categories. Microvesicles and exosomes are among the subgroups. Microvesicles transport phosphatidylserine-containing proteins, mRNAs, miRNAs, and lipids, while exosomes transport DNA, lipids, RNAs, and proteins [145]. Human umbilical cord EVs have been demonstrated to exert anti-inflammatory qualities (interleukin-10, IL-10) and reduced pro-inflammatory responses (IL-1β, IL-6), followed by improved motor, axon, and Schwann cell regeneration [146]. Myoblasts release EVs containing HGF, IGF-1, TGF-3, VEGF, fibroblast growth factor-β3 (TGF-β3) and fibroblast growth factor 2 (FGF2). These secretions promote cell interaction between myoblasts for proliferation [147]. EVs enhance cross-talk in skeletal muscle to induce glucose release and adipose dissociation [148]. Interestingly, EVs that are secreted by astrocyte support a variety of tasks that range from delivering angiogenic factors such as FGF2, and VEGF to heat shock proteins, synapsin 1 and apolipoprotein-D, which aid in neuronal protection in hazardous settings [149]. EVs play a role in neuronal cell communication and in response to external stimuli such as inflammatory responses and the central nervous system [150]. Although EV can be created from a variety of cell sources, EV derived from MSC is persuasive in its ability to regenerate the central nervous system [151]. Figure 8 summarises the regenerative properties of EVs in neuronal and muscular tissue.
Recent studies [152] on patients with vocal fold atrophy, scarring, and sulcus have examined PRP. PRP is a combination of autologous growth factors that stimulate EGFR secretion to promote wound healing. It includes PDGF, TGF-β, VEGF, EGF and IGF [153]. However, this treatment demonstrated a short augmentation period, which is typically between three to six months [152]. PRP has been demonstrated to increase ECM remodelling and decrease collagen density during vocal fold wound healing [153]. Platelets release cytokines and growth factors that stimulate cell proliferation, angiogenesis, and cell migration during vocal fold injury [154].
Gene therapy aims to transmit genetic material, such as deoxyribonucleic acid (DNA) or ribonucleic acid (RNA), in order to modify the genetic outcome [155]. Nonetheless, gene therapy has created safety concerns, as it may generate negative side effects such as inflammatory reactions [156]. A study sought to increase the effectiveness of silencing prolyl hydroxylase domain-containing protein 2 (PHD2) by delivering siRNA through biodegradable biomaterial, to regulate the inflammatory response of the host and promote angiogenesis upon implantation of biomaterials [157]. PHD2 is one of the essential genes that affect endothelial cell inflammatory properties. By silencing PHD2, it is possible to reduce over-inflammation, which has applications in acute inflammatory diseases [158]. By incorporating biomaterial application into gene therapy, the biomaterial can delay the release of gene materials through progressive degradation and reduce the risk of inflammation [159,160]. Figure 8 indicates the overall regenerative capacity of biomolecules in the vocal fold.

4. Future Study and Limitation

Previous research has proposed two types of cell encapsulation, including stem cells and native cells. The majority of previous research demonstrated the role of native cells in maintaining the vibratory function of vocal folds but not their regenerative capacity. Therefore, the current review suggests that encapsulating stem cells in biomaterials is a more promising technique for augmenting vocal folds. This is because glottic insufficiency may continue, necessitating the use of regenerating biomaterials. Moreover, immunomodulatory characteristics of MSCs can decrease fibrosis during injury [101,161]. In a clinical experiment, cell treatment for vocal fold regeneration utilizing autologous BMMSCs yielded promising results; however, other MSCs sources should be examined to minimize invasive cell harvesting techniques [162]. Future research should focus on incorporating stem cells into biomaterials, particularly WJMSCs and iPSCs, to improve regeneration outcomes. Although WJMSCs and iPSCs have been explored in a variety of applications, including in neuronal tissue, cardiac tissue, skin, cartilage, muscle, and bone, there are few preliminary studies that indicate their potential for vocal fold regeneration [163]. Although both ESCs and iPSCs exhibited the ability to differentiate, the cultivation of ESCs involved ethical problems and the possibility of host rejection, whereas the cultivation of iPSCs needed costly technology and a metagenesis risk [164]. Moreover, some nations restricted the use of ESCs owing to religious practices [165]. Previous research on the application of MSCs to vocal fold regeneration focused exclusively on BMMSCs and ASCs [166]. BMMSCs were the most extensively investigated adult stem cells, but practical translation remained difficult, and ASCs were favored because of their superior stability and differentiation capacity [167]. WJMSCs were found to have superior cell proliferation and immunophenotypic indicators compared to ASCs [168]. Future research must give empirical data on WJMSCs and iPSCs in vocal fold regeneration as a basis for translational research. Future research should examine the efficacy of restoring native vocal folds by direct injection of WJMSCs and with hydrogel, the concentration of WJMSCs, and the sustainability of the treatment. Additional research is required to achieve genomic stability, low tumorigenesis, low toxicity, and low immunogenicity in iPSCs for use in vocal fold regeneration [169]. Future research must also address [170] the type of cell sources to be induced, the method of gene change, and the efficiency of induction.
As stated previously, the lamina propria capillaries are lined with pericytes, and research has shown that pericytes can differentiate from human pluripotent stem cells and are involved in fibrogenesis, angiogenesis, immunomodulation, and differentiation [171,172]. Pericyte-like differentiated ASCs have been found to promote endothelial cells, which aid in retinal vascularization [173]. Although previous studies suggested that pericytes were multipotent and capable of regenerating damaged tissue and promoting healing, a single study [174] contradicted this notion. Therefore, the interaction between injected stem cells and native pericyte is a topic worthy of investigation, since it may be one of the mechanisms underlying angiogenesis.
bFGF and HGF have been clinically studied, and their efficacy has been established [126,127,128,131]. However, the direct injection administration route was utilized. There are currently studies incorporating bFGF into biomaterials that indicate a positive in vivo outcome [70,175]. Future study should determine whether the integration of these two growth factors into biomaterials is effective in prolonging the release period and enhancing regeneration in a shorter time frame. As vocal folds are composed of extensive muscle, nerve, and adipose tissue, this review also proposes that VEGF and IGF-1 may have potential applications in vocal fold regeneration, as VEGF increases the proliferation of epithelial and neuronal cells and IGF-1 promotes muscle regeneration. However, research should investigate the regenerative efficiency, structural changes following administration, and the VEGF and IGF-1 pathways implicated in the vocal fold.
Previous research has suggested that the MSC’s regenerative properties may be due to the release of EVs [176]. EV delivery provides cell-free therapy, eliminating drawbacks such as carcinogenesis, graft-versus-host disease, and instability due to storage and senescence [177,178]. It is debatable in multiple applications, including nervous, cardiac, bone, cartilage, kidney, liver, muscle, and wound healing, but not vocal fold regeneration [179,180]. With that, this review suggests that EVs are a potential biomolecule for use in vocal fold regeneration. As a result of its anti-immunogenic and regenerative properties, it is capable of restoring nerve and muscle function in the vocal fold, as nerve and muscle degeneration or dysfunction are the most common causes of vocal fold paralysis. There are studies in using different sources of EVs, from BMMSCs, WJMSCs or ASCs and more, and the claim for their effectiveness was affirmative. However, the efficacy of EVs derived from different types of cell sources, as well as the comparison between direct injection of EVs and incorporation in an injectable hydrogel, should be elucidated. Nevertheless, isolation procedures, storage conditions, and injection concentrations need to be fully optimized. Only with sufficient pre-clinical data can the clinical application of EVs for individuals with glottic insufficiency be translated.
Moreover, this review also suggests that future study should explore incorporation of cells and biomolecules together in hydrogel, to obtain better results. The synergetic effect of cells and biomolecules will provide better regenerative outcomes for the native tissue. In short, current tissue engineering in glottic insufficiency has received inadequate study, whereby current progress focuses on direct injection of cells or biomolecules and encapsulation of cells or biomolecules alone; the sources of cells might not be relevant for expansion in clinical studies (for example, the use of BMMSCs). This review proposes that future studies should look into encapsulation of cells and biomolecules together in hydrogel and application of relevant cell sources such as WJMSCs and iPSCs. Table 1 and Table 2 provide a summary of current research on the injection of biomolecules or cells into the vocal folds for regenerative purposes, as well as a list of cells or biomolecules with the potential for hydrogel encapsulation for vocal fold injection.

5. Conclusions

Most current tissue engineering in glottic insufficiency focuses on direct injection, biomaterials, cells, or biomolecules independently. Compared to other disease models, the encapsulation of cells and biomolecules in hydrogel offers better synergetic effects. Therefore, this review suggests the potential use of WJMSCs as comparison to ASCs and BMMSCs, due to the ease of obtaining it and its good proliferation. Application of iPSCs should have more studies to yield mature processing techniques and robust outcomes. Encapsulation of VEGF, IGF-1, and EVs in the regeneration of vocal folds should also be elucidate, as that of PRP, bFGF and HGF have been. Based on a review of the literature, the tissue engineering of these stem cells and biomolecules has promising potential in the regeneration of vocal fold muscles and nerves. The recommendation is based on a review of the relevant literature; therefore, additional work and research should be conducted on the suggested cells and biomolecules to provide preliminary evidence for translational application.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/biomedicines10123082/s1, Figure S1: Type of hydrogel; Figure S2: Type of delivery of various regenerative target; Figure S3: Type of cell delivery via hydrogel; Figure S4: Type of biomolecule delivery via hydrogel.

Author Contributions

Conceptualization, W.-C.N., M.M.B., M.B.F., Y.L. and M.A.; methodology, W.-C.N., M.A. and M.B.F.; validation, M.M.B., M.B.F., Y.L. and M.A.; formal analysis, W.-C.N.; investigation, W.-C.N., M.A. and Y.L.; data curation, W.-C.N., M.A., A.A.Z. and Y.L.; writing—original draft preparation, W.-C.N.; writing—review and editing, M.M.B., M.B.F., Y.L., A.A.Z. and M.A.; visualization, W.-C.N., M.M.B., M.B.F., Y.L., A.A.Z. and M.A; supervision, M.M.B., M.B.F., Y.L., A.A.Z. and M.A.; project administration, M.B.F. and M.A.; funding acquisition, Y.L. and M.A. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Minister of Higher Education (MOHE) Fundamental Research Grant Scheme (FGRS), grant number FRGS/1/2020/SKK06/UKM/01/1.

Institutional Review Board Statement

The study was conducted according to the ethical guidelines underpinned by right-based theories, whereby it adheres to the principles of beneficence, non-maleficence, justice, honesty and gratitude. The study was approved by the Institutional Review Board (or Ethics Committee) of UNIVERSITI KEBANGSAAN MALAYSIA (protocol code FF-2020-818 on 3 February 2020).

Informed Consent Statement

Not applicable.

Data Availability Statement

Data sharing is not applicable to this article as no new data were created or analyzed in this study.

Acknowledgments

We would like to thank the Otorhinolaryngology-Head and Neck Surgery department, Centre of Tissue Engineering and Regenerative Medicine, Obstetrics and Gynaecology department and Faculty of Medicine UKM for providing resources to complete this review.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

ASCsAdipose stem cells
BDNFBrain-derived neurotrophic factor
bFGFBasic fibroblast growth factor
BMMSCsBone marrow mesenchymal stem cells
BMP4Bone morphogenetic protein 4
CaHACalcium hydroxyapatite
CMCCarboxymethylcellulose
CNFCiliary neurotrophic factor
CTCricothyroid
DNADeoxyribonucleic acid
ECFCsEndothelial colony-forming cells
ECMExtracellular matrix
EGFEpidermal growth factor
ESCEmbryonic stem cell
EVExtracellular vesicle
FGF2Fibroblast growth factor 2
GAGGlycosaminoglycans
GFOGERCollagen-derived ligands
HAHyaluronic acid
HGFHepatocyte growth factor
HUVECHuman umbilical vein endothelial cell
IAInterarytenoid
IGF-1Insulin growth factor-1
IL Interleukin
iPSCsInduced pluripotent stem cells
LCALateral-cricoarytenoid
MMPMatrix metalloproteinase
MSCMesenchymal stem cell
NF-ĸBNuclear factor-kappa beta
NGFNerve growth factor
NKNatural killer
NSLRNon-selective laryngeal reinnervation
PCAPosterior cricoarytenoid
PDGF-ABPlatelet-derived growth factor AB
PGProteoglycans
PHD2prolyl hydroxylase domain-containing protein 2
PRPPlatelet-rich plasma
PSPProgressive supranuclear palsy
RGDRipeptide arginine-glycine-aspartic acid
RLNRecurrent laryngeal nerve
RNARibonucleic acid
SDF-1Stromal cell-derived factor-1
SLNSuperior laryngeal nerve
TAThyroarytenoid
TA-LCAThyroarytenoid-lateral cricoarytenoid
TGF-βTransforming growth factor beta
UVFPUnilateral vocal fold paralysis
VEGFVascular endothelial growth factor
VHIVoice Handicap Index
WJMSCsWharton’s Jelly mesenchymal stem cells
YIGSRLaminin-derived ligands

References

  1. Rosow, D.E.; Pan, D.R. Presbyphonia and Minimal Glottic Insufficiency. Otolaryngol. Clin. N. Am. 2019, 52, 617–625. [Google Scholar] [CrossRef] [PubMed]
  2. De Araújo Pernambuco, L.; Espelt, A.; Balata, P.M.M.; de Lima, K.C. Prevalence of voice disorders in the elderly: A systematic review of population-based studies. Eur. Arch. Oto-Rhino-Laryngol. 2015, 272, 2601–2609. [Google Scholar] [CrossRef] [PubMed]
  3. Lyberg-Åhlander, V.; Rydell, R.; Fredlund, P.; Magnusson, C.; Wilén, S. Prevalence of Voice Disorders in the General Population, Based on the Stockholm Public Health Cohort. J. Voice 2019, 33, 900–905. [Google Scholar] [CrossRef] [PubMed]
  4. Onwordi, L.N.; Al Yaghchi, C. Airway Glottic Insufficiency; StatPearls Publishing: Treasure Island, FL, USA, 2022. [Google Scholar]
  5. Lee, S.W.; Park, K.N. A long-term comparative prospective study between reinnervation and injection laryngoplasty. Laryngoscope 2018, 128, 1893–1897. [Google Scholar] [CrossRef] [PubMed]
  6. Ban, M.J.; Lee, S.C.; Park, J.H.; Park, K.N.; Kim, H.K.; Lee, S.W. Regenerative efficacy of fibroblast growth factor for the treatment of aged vocal fold: From animal model to clinical application. Clin. Otolaryngol. 2021, 46, 131–137. [Google Scholar] [CrossRef] [PubMed]
  7. Sharma, V.; Dash, S.K.; Govarthanan, K.; Gahtori, R.; Negi, N.; Barani, M.; Tomar, R.; Chakraborty, S.; Mathapati, S.; Bishi, D.K.; et al. Recent Advances in Cardiac Tissue Engineering for the Management of Myocardium Infarction. Cells 2021, 10, 2538. [Google Scholar] [CrossRef] [PubMed]
  8. Walczak, P.A.; Perez-Esteban, P.; Bassett, D.C.; Hill, E.J. Modelling the central nervous system: Tissue engineering of the cellular microenvironment. Emerg. Top. life Sci. 2021, 5, 507–517. [Google Scholar] [CrossRef] [PubMed]
  9. Pedrero, S.G.; Llamas-Sillero, P.; Serrano-López, J. A Multidisciplinary Journey towards Bone Tissue Engineering. Materials 2021, 14, 4896. [Google Scholar] [CrossRef] [PubMed]
  10. Stampoultzis, T.; Karami, P.; Pioletti, D.P. Thoughts on cartilage tissue engineering: A 21st century perspective. Curr. Res. Transl. Med. 2021, 69, 103299. [Google Scholar] [CrossRef]
  11. Fishman, J.M.; Long, J.; Gugatschka, M.; De Coppi, P.; Hirano, S.; Hertegard, S.; Thibeault, S.L.; Birchall, M.A. Stem cell approaches for vocal fold regeneration. Laryngoscope 2016, 126, 1865–1870. [Google Scholar] [CrossRef]
  12. Kumai, Y. Pathophysiology of Fibrosis in the Vocal Fold: Current Research, Future Treatment Strategies, and Obstacles to Restoring Vocal Fold Pliability. Int. J. Mol. Sci. 2019, 20, 2551. [Google Scholar] [CrossRef] [Green Version]
  13. Tian, H.; Pan, J.; Chen, L.; Wu, Y. A narrative review of current therapies in unilateral recurrent laryngeal nerve injury caused by thyroid surgery. Gland Surg. 2022, 11, 270–278. [Google Scholar] [CrossRef] [PubMed]
  14. Mattei, A.; Magalon, J.; Bertrand, B.; Philandrianos, C.; Veran, J.; Giovanni, A. Cell therapy and vocal fold scarring. Eur. Ann. Otorhinolaryngol. Head Neck Dis. 2017, 134, 339–345. [Google Scholar] [CrossRef] [PubMed]
  15. Benboujja, F.; Greenberg, M.; Nourmahnad, A.; Rath, N.; Hartnick, C. Evaluation of the Human Vocal Fold Lamina Propria Development Using Optical Coherence Tomography. Laryngoscope 2021, 131, E2558–E2565. [Google Scholar] [CrossRef] [PubMed]
  16. Wrona, E.A.; Peng, R.; Amin, M.R.; Branski, R.C.; Freytes, D.O. Extracellular Matrix for Vocal Fold Lamina Propria Replacement: A Review. Tissue Eng. Part B Rev. 2016, 22, 421–429. [Google Scholar] [CrossRef] [PubMed]
  17. Saran, M.; Georgakopoulos, B.; Bordoni, B. Anatomy, Head and Neck, Larynx Vocal Cords; StatPearls Publishing: Treasure Island, FL, USA, 2022. [Google Scholar]
  18. Tang, S.S.; Mohad, V.; Gowda, M.; Thibeault, S.L. Insights Into the Role of Collagen in Vocal Fold Health and Disease. J. Voice 2017, 31, 520–527. [Google Scholar] [CrossRef]
  19. Moore, J.; Thibeault, S. Insights into the role of elastin in vocal fold health and disease. J. Voice 2012, 26, 269–275. [Google Scholar] [CrossRef] [Green Version]
  20. Levendoski, E.E.; Leydon, C.; Thibeault, S.L. Vocal fold epithelial barrier in health and injury: A research review. J. Speech Lang. Hear. Res. 2014, 57, 1679–1691. [Google Scholar] [CrossRef] [Green Version]
  21. Sato, K.; Chitose, S.-I.; Sato, K.; Sato, F.; Ono, T.; Umeno, H. Epithelium of the human vocal fold as a vibrating tissue. Auris Nasus Larynx 2021, 48, 704–709. [Google Scholar] [CrossRef] [PubMed]
  22. Sato, K. Pericytes in the Human Vocal Fold Mucosa. Adv. Exp. Med. Biol. 2018, 1109, 79–93. [Google Scholar] [CrossRef]
  23. Jones, C.L.; Achuthan, A.; Erath, B.D. Modal response of a computational vocal fold model with a substrate layer of adipose tissue. J. Acoust. Soc. Am. 2015, 137, EL158–EL164. [Google Scholar] [CrossRef]
  24. Chhetri, D.K.; Neubauer, J.; Sofer, E. Posterior cricoarytenoid muscle dynamics in canines and humans. Laryngoscope 2014, 124, 2363–2367. [Google Scholar] [CrossRef] [Green Version]
  25. Manriquez, R.; Peterson, S.D.; Prado, P.; Orio, P.; Galindo, G.E.; Zanartu, M. Neurophysiological Muscle Activation Scheme for Controlling Vocal Fold Models. IEEE Trans. Neural Syst. Rehabil. Eng. 2019, 27, 1043–1052. [Google Scholar] [CrossRef] [PubMed]
  26. Ivey, C.M. Vocal Fold Paresis. Otolaryngol. Clin. N. Am. 2019, 52, 637–648. [Google Scholar] [CrossRef] [PubMed]
  27. Weissbrod, P.; Pitman, M.J.; Sharma, S.; Bender, A.; Schaefer, S.D. Quantity and three-dimensional position of the recurrent and superior laryngeal nerve lower motor neurons in a rat model. Ann. Otol. Rhinol. Laryngol. 2011, 120, 761–768. [Google Scholar] [CrossRef] [PubMed]
  28. Dekhou, A.S.; Morrison, R.J.; Gemechu, J.M. The Superior Laryngeal Nerve and Its Vulnerability in Surgeries of the Neck. Diagnostics 2021, 11, 1243. [Google Scholar] [CrossRef] [PubMed]
  29. Dewan, K.; Vahabzadeh-Hagh, A.; Soofer, D.; Chhetri, D.K. Neuromuscular compensation mechanisms in vocal fold paralysis and paresis. Laryngoscope 2017, 127, 1633–1638. [Google Scholar] [CrossRef] [PubMed]
  30. Marina, M.B.; Marie, J.-P.; Birchall, M.A. Laryngeal reinnervation for bilateral vocal fold paralysis. Curr. Opin. Otolaryngol. Head Neck Surg. 2011, 19, 434–438. [Google Scholar] [CrossRef] [PubMed]
  31. Salik, I.; Winters, R. Bilateral Vocal Cord Paralysis; StatPearls Publishing: Treasure Island, FL, USA, 2022. [Google Scholar]
  32. Carroll, T.L.; Rosen, C.A. Trial vocal fold injection. J. Voice 2010, 24, 494–498. [Google Scholar] [CrossRef] [PubMed]
  33. Martins, R.H.G.; Gonçalvez, T.M.; Pessin, A.B.B.; Branco, A. Aging voice: Presbyphonia. Aging Clin. Exp. Res. 2014, 26, 1–5. [Google Scholar] [CrossRef] [PubMed]
  34. Samlan, R.A.; Kunduk, M.; Ikuma, T.; Black, M.; Lane, C. Vocal Fold Vibration in Older Adults With and Without Age-Related Dysphonia. Am. J. Speech-Lang. Pathol. 2018, 27, 1039–1050. [Google Scholar] [CrossRef] [PubMed]
  35. Bouhabel, S.; Hartnick, C.J. Current trends in practices in the treatment of pediatric unilateral vocal fold immobility: A survey on injections, thyroplasty and nerve reinnervation. Int. J. Pediatr. Otorhinolaryngol. 2018, 109, 115–118. [Google Scholar] [CrossRef] [PubMed]
  36. Padia, R.; Smith, M.E. Posterior Glottic Insufficiency in Children. Ann. Otol. Rhinol. Laryngol. 2017, 126, 268–273. [Google Scholar] [CrossRef] [PubMed]
  37. Bruzzi, C.; Salsi, D.; Minghetti, D.; Negri, M.; Casolino, D.; Sessa, M. Presbiphonya. Acta Biomed. 2017, 88, 6–10. [Google Scholar] [CrossRef] [PubMed]
  38. Chen, X.; Thibeault, S.L. Characteristics of age-related changes in cultured human vocal fold fibroblasts. Laryngoscope 2008, 118, 1700–1704. [Google Scholar] [CrossRef] [Green Version]
  39. Dayangku, N.P.; Marina, M.B.; Mawaddah, A.; WP, S.E.; Abdullah, S. Gore-Tex Medialisation Thyroplasty for Unilateral Vocal Cord Palsy: A Tertiary Centre 7 Years Experience. IIUM Med. J. Malaysia 2016, 15, 13–17. [Google Scholar] [CrossRef]
  40. Wilson, A.; Kimball, E.E.; Sayce, L.; Luo, H.; Khosla, S.M.; Rousseau, B. Medialization Laryngoplasty: A Review for Speech-Language Pathologists. J. Speech. Lang. Hear. Res. 2021, 64, 481–490. [Google Scholar] [CrossRef]
  41. Ab Rani, A.; Azman, M.; Ubaidah, M.A.; Mohamad Yunus, M.R.; Sani, A.; Mat Baki, M. Nonselective Laryngeal Reinnervation versus Type 1 Thyroplasty in Patients with Unilateral Vocal Fold Paralysis: A Single Tertiary Centre Experience. J. Voice 2021, 35, 487–492. [Google Scholar] [CrossRef] [PubMed]
  42. Mawaddah, A.; Marina, M.B.; Halimuddin, S.; Mohd Razif, M.Y.; Abdullah, S. A Ten-Year Kuala Lumpur Review on Laser Posterior Cordectomy for Bilateral Vocal Fold Immobility. Malays. J. Med. Sci. 2016, 23, 65–70. [Google Scholar] [CrossRef] [PubMed]
  43. Bick, E.; Dumberger, L.D.; Farquhar, D.R.; Davis, H.; Ramsey, E.; Buckmire, R.A.; Shah, R.N. Does Voice Therapy Improve Vocal Outcomes in Vocal Fold Atrophy? Ann. Otol. Rhinol. Laryngol. 2021, 130, 602–608. [Google Scholar] [CrossRef] [PubMed]
  44. Dion, G.R.; Nielsen, S.W. In-Office Laryngology Injections. Otolaryngol. Clin. N. Am. 2019, 52, 521–536. [Google Scholar] [CrossRef] [PubMed]
  45. Chow, X.H.; Johari, S.F.; Rosla, L.; Wahab, A.F.A.; Azman, M.; Baki, M.M. Early Transthyrohyoid Injection Laryngoplasty Under Local Anaesthesia in a Single Tertiary Center of Southeast Asia: Multidimensional Voice Outcomes. Turkish Arch. Otorhinolaryngol. 2021, 59, 271–281. [Google Scholar] [CrossRef]
  46. Cohen, J.T.; Benyamini, L. Voice outcome after vocal fold injection augmentation with carboxymethyl cellulose versus calcium hydroxyapatite. J. Laryngol. Otol. 2020, 134, 263–269. [Google Scholar] [CrossRef] [PubMed]
  47. Lahav, Y.; Malka-Yosef, L.; Shapira-Galitz, Y.; Cohen, O.; Halperin, D.; Shoffel-Havakuk, H. Vocal Fold Fat Augmentation for Atrophy, Scarring, and Unilateral Paralysis: Long-term Functional Outcomes. Otolaryngol. Head Neck Surg. 2021, 164, 631–638. [Google Scholar] [CrossRef]
  48. Tsou, Y.-A.; Tien, V.H.; Chen, S.-H.; Shih, L.-C.; Lin, T.-C.; Chiu, C.-J.; Chang, W.-D. Autologous Fat Plus Platelet-Rich Plasma versus Autologous Fat Alone on Sulcus Vocalis. J. Clin. Med. 2022, 11, 725. [Google Scholar] [CrossRef] [PubMed]
  49. Cantarella, G.; Mazzola, R.F.; Gaffuri, M.; Iofrida, E.; Biondetti, P.; Forzenigo, L.V.; Pignataro, L.; Torretta, S. Structural Fat Grafting to Improve Outcomes of Vocal Folds’ Fat Augmentation: Long-term Results. Otolaryngol. Head Neck Surg. 2018, 158, 135–143. [Google Scholar] [CrossRef] [PubMed]
  50. Seria, E.; Galea, G.; Borg, J.; Schembri, K.; Grech, G.; Tagliaferro, S.S.; Felice, A. Novel leukocyte-depleted platelet-rich plasma-based skin equivalent as an in vitro model of chronic wounds: A preliminary study. BMC Mol. Cell Biol. 2021, 22, 1–13. [Google Scholar] [CrossRef] [PubMed]
  51. Zamani, M.; Yaghoubi, Y.; Movassaghpour, A.; Shakouri, K.; Mehdizadeh, A.; Pishgahi, A.; Yousefi, M. Novel therapeutic approaches in utilizing platelet lysate in regenerative medicine: Are we ready for clinical use? J. Cell. Physiol. 2019, 234, 17172–17186. [Google Scholar] [CrossRef] [PubMed]
  52. Baptista, P.M.; Atala, A. Chapter 1—Regenerative Medicine: The Hurdles and Hopes; Academic Press: Boston, MA, USA, 2016; pp. 3–7. ISBN 978-0-12-800548-4. [Google Scholar]
  53. Furth, M.E.; Atala, A. Chapter 6—Tissue Engineering: Future Perspectives; Academic Press: Boston, MA, USA, 2014; pp. 83–123. ISBN 978-0-12-398358-9. [Google Scholar]
  54. Walimbe, T.; Panitch, A.; Sivasankar, P.M. A Review of Hyaluronic Acid and Hyaluronic Acid-based Hydrogels for Vocal Fold Tissue Engineering. J. Voice 2017, 31, 416–423. [Google Scholar] [CrossRef] [PubMed]
  55. Bakhshandeh, B.; Zarrintaj, P.; Oftadeh, M.O.; Keramati, F.; Fouladiha, H.; Sohrabi-Jahromi, S.; Ziraksaz, Z. Tissue engineering; strategies, tissues, and biomaterials. Biotechnol. Genet. Eng. Rev. 2017, 33, 144–172. [Google Scholar] [CrossRef] [PubMed]
  56. Akter, F. Chapter 2—Principles of Tissue Engineering; Academic Press: Cambridge, MA, USA, 2016; pp. 3–16. ISBN 978-0-12-805361-4. [Google Scholar]
  57. Koh, B.; Sulaiman, N.; Fauzi, M.B.; Law, J.X.; Ng, M.H.; Idrus, R.B.H.; Yazid, M.D. Three dimensional microcarrier system in mesenchymal stem cell culture: A systematic review. Cell Biosci. 2020, 10, 1–16. [Google Scholar] [CrossRef]
  58. Sheikh, I.; Dahman, Y. Chapter 2—Applications of Nanobiomaterials in Hard Tissue Engineering; William Andrew Publishing: Norwich, NY, USA, 2016; pp. 33–62. ISBN 978-0-323-42862-0. [Google Scholar]
  59. Li, L.; Stiadle, J.M.; Lau, H.K.; Zerdoum, A.B.; Jia, X.; Thibeault, S.L.; Kiick, K.L. Tissue engineering-based therapeutic strategies for vocal fold repair and regeneration. Biomaterials 2016, 108, 91–110. [Google Scholar] [CrossRef] [Green Version]
  60. Kannan, R.; Wei, G.; Ma, P.X. Chapter 2—Synthetic polymeric biomaterials for tissue engineering. In Woodhead Publishing Series in Biomaterials; Woodhead Publishing: Sawston, UK, 2022; pp. 41–74. ISBN 978-0-12-820508-2. [Google Scholar]
  61. Heris, H.K.; Latifi, N.; Vali, H.; Li, N.; Mongeau, L. Investigation of Chitosan-glycol/glyoxal as an Injectable Biomaterial for Vocal Fold Tissue Engineering. Procedia Eng. 2015, 110, 143–150. [Google Scholar] [CrossRef] [Green Version]
  62. Li, L.; Stiadle, J.M.; Levendoski, E.E.; Lau, H.K.; Susan, L.; Kiick, K.L.; Surgery, N. Biocompatibility of Injectable Resilin-based Hydrogels. J. Biomed. Mater. Res. B Appl. Biomater. 2018, 106, 2229–2242. [Google Scholar] [CrossRef] [PubMed]
  63. Kwon, S.; Choi, H.; Park, C.; Choi, S.; Kim, E.; Kim, S.W.; Kim, C.-S.; Koo, H. In vivo vocal fold augmentation using an injectable polyethylene glycol hydrogel based on click chemistry. Biomater. Sci. 2021, 9, 108–115. [Google Scholar] [CrossRef] [PubMed]
  64. Yap, L.-S.; Yang, M.-C. Thermo-reversible injectable hydrogel composing of pluronic F127 and carboxymethyl hexanoyl chitosan for cell-encapsulation. Colloids Surf. B Biointerfaces 2020, 185, 110606. [Google Scholar] [CrossRef] [PubMed]
  65. Tong, X.; Yang, F. Recent Progress in Developing Injectable Matrices for Enhancing Cell Delivery and Tissue Regeneration. Adv. Healthc. Mater. 2018, 7, e1701065. [Google Scholar] [CrossRef]
  66. Oliva, N.; Conde, J.; Wang, K.; Artzi, N. Designing Hydrogels for On-Demand Therapy. Acc. Chem. Res. 2017, 50, 669–679. [Google Scholar] [CrossRef]
  67. Ma, J.; Huang, C. Composition and Mechanism of Three-Dimensional Hydrogel System in Regulating Stem Cell Fate. Tissue Eng. Part B Rev. 2020, 26, 498–518. [Google Scholar] [CrossRef]
  68. Hamilton, M.; Harrington, S.; Dhar, P.; Stehno-Bittel, L. Hyaluronic Acid Hydrogel Microspheres for Slow Release Stem Cell Delivery. ACS Biomater. Sci. Eng. 2021, 7, 3754–3763. [Google Scholar] [CrossRef]
  69. Young, S.A.; Riahinezhad, H.; Amsden, B.G. In situ-forming, mechanically resilient hydrogels for cell delivery. J. Mater. Chem. B 2019, 7, 5742–5761. [Google Scholar] [CrossRef]
  70. Imaizumi, M.; Nakamura, R.; Nakaegawa, Y.; Dirja, B.T.; Tada, Y.; Tani, A.; Sugino, T.; Tabata, Y.; Omori, K. Regenerative potential of basic fibroblast growth factor contained in biodegradable gelatin hydrogel microspheres applied following vocal fold injury: Early effect on tissue repair in a rabbit model. Braz. J. Otorhinolaryngol. 2021, 87, 274–282. [Google Scholar] [CrossRef] [PubMed]
  71. Fuchs, S.; Shariati, K.; Ma, M. Specialty Tough Hydrogels and Their Biomedical Applications. Adv. Healthc. Mater. 2020, 9, 1901396. [Google Scholar] [CrossRef] [PubMed]
  72. Boddupalli, A.; Zhu, L.; Bratlie, K.M. Methods for Implant Acceptance and Wound Healing: Material Selection and Implant Location Modulate Macrophage and Fibroblast Phenotypes. Adv. Healthc. Mater. 2016, 5, 2575–2594. [Google Scholar] [CrossRef] [PubMed]
  73. Hinz, B. Matrix mechanics and regulation of the fibroblast phenotype. Periodontology 2000 2013, 63, 14–28. [Google Scholar] [CrossRef] [PubMed]
  74. Aregueta-Robles, U.A.; Martens, P.J.; Poole-Warren, L.A.; Green, R.A. Tissue engineered hydrogels supporting 3D neural networks. Acta Biomater. 2019, 95, 269–284. [Google Scholar] [CrossRef]
  75. Dang, L.H.; Doan, P.; Nhi, T.T.Y.; Nguyen, D.T.; Nguyen, B.T.; Nguyen, T.P.; Tran, N.Q. Multifunctional injectable pluronic-cystamine-alginate-based hydrogel as a novel cellular delivery system towards tissue regeneration. Int. J. Biol. Macromol. 2021, 185, 592–603. [Google Scholar] [CrossRef]
  76. Madl, C.M.; Heilshorn, S.C. Engineering Hydrogel Microenvironments to Recapitulate the Stem Cell Niche. Annu. Rev. Biomed. Eng. 2018, 20, 21–47. [Google Scholar] [CrossRef]
  77. Vernerey, F.J.; Lalitha Sridhar, S.; Muralidharan, A.; Bryant, S.J. Mechanics of 3D Cell-Hydrogel Interactions: Experiments, Models, and Mechanisms. Chem. Rev. 2021, 121, 11085–11148. [Google Scholar] [CrossRef]
  78. Ho, M.T.; Teal, C.J.; Shoichet, M.S. A hyaluronan/methylcellulose-based hydrogel for local cell and biomolecule delivery to the central nervous system. Brain Res. Bull. 2019, 148, 46–54. [Google Scholar] [CrossRef]
  79. Xu, Q.; Sigen, A.; Gao, Y.; Guo, L.; Creagh-Flynn, J.; Zhou, D.; Greiser, U.; Dong, Y.; Wang, F.; Tai, H.; et al. A hybrid injectable hydrogel from hyperbranched PEG macromer as a stem cell delivery and retention platform for diabetic wound healing. Acta Biomater. 2018, 75, 63–74. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Yin, H.; Yan, Z.; Bauer, R.J.; Peng, J.; Schieker, M.; Nerlich, M.; Docheva, D. Functionalized thermosensitive hydrogel combined with tendon stem/progenitor cells as injectable cell delivery carrier for tendon tissue engineering. Biomed. Mater. 2018, 13, 34107. [Google Scholar] [CrossRef] [PubMed]
  81. Datta, S.; Rameshbabu, A.P.; Bankoti, K.; Jana, S.; Roy, S.; Sen, R.; Dhara, S. Microsphere embedded hydrogel construct—Binary delivery of alendronate and BMP-2 for superior bone regeneration. J. Mater. Chem. B 2021, 9, 6856–6869. [Google Scholar] [CrossRef] [PubMed]
  82. Yuan, X.; Yuan, W.; Ding, L.; Shi, M.; Luo, L.; Wan, Y.; Oh, J.; Zhou, Y.; Bian, L.; Deng, D.Y.B. Cell-adaptable dynamic hydrogel reinforced with stem cells improves the functional repair of spinal cord injury by alleviating neuroinflammation. Biomaterials 2021, 279, 121190. [Google Scholar] [CrossRef]
  83. Lev, R.; Seliktar, D. Hydrogel biomaterials and their therapeutic potential for muscle injuries and muscular dystrophies. J. R. Soc. Interface 2018, 15, 20170380. [Google Scholar] [CrossRef]
  84. Mattei, A.; Bertrand, B.; Jouve, E.; Blaise, T.; Philandrianos, C.; Grimaud, F.; Giraudo, L.; Aboudou, H.; Dumoulin, C.; Arnaud, L.; et al. Feasibility of First Injection of Autologous Adipose Tissue-Derived Stromal Vascular Fraction in Human Scarred Vocal Folds: A Nonrandomized Controlled Trial. JAMA Otolaryngol. Head Neck Surg. 2020, 146, 355–363. [Google Scholar] [CrossRef]
  85. Lasso, J.M.; Poletti, D.; Scola, B.; Gómez-Vilda, P.; García-Martín, A.I.; Fernández-Santos, M.E. Injection Laryngoplasty Using Autologous Fat Enriched with Adipose-Derived Regenerative Stem Cells: A Safe Therapeutic Option for the Functional Reconstruction of the Glottal Gap after Unilateral Vocal Fold Paralysis. Stem Cells Int. 2018, 2018, 8917913. [Google Scholar] [CrossRef] [Green Version]
  86. Li, X.; Wang, H.; Xu, W. HGF and bFGF Secreted by Adipose-Derived Mesenchymal Stem Cells Revert the Fibroblast Phenotype Caused by Vocal Fold Injury in a Rat Model. J. Voice 2020, 36, 622–629. [Google Scholar] [CrossRef]
  87. Valerie, A.; Vassiliki, K.; Irini, M.; Nikolaos, P.; Karampela, E.; Apostolos, P. Adipose-Derived Mesenchymal Stem Cells in the Regeneration of Vocal Folds: A Study on a Chronic Vocal Fold Scar. Stem Cells Int. 2016, 2016, 9010279. [Google Scholar] [CrossRef] [Green Version]
  88. Morisaki, T.; Kishimoto, Y.; Tateya, I.; Kawai, Y.; Suzuki, R.; Tsuji, T.; Hiwatashi, N.; Nakamura, T.; Omori, K.; Kitano, H.; et al. Adipose-derived mesenchymal stromal cells prevented rat vocal fold scarring. Laryngoscope 2018, 128, E33–E40. [Google Scholar] [CrossRef]
  89. Huang, D.; Wang, R.; Yang, S. Cogels of Hyaluronic Acid and Acellular Matrix for Cultivation of Adipose-Derived Stem Cells: Potential Application for Vocal Fold Tissue Engineering. BioMed Res. Int. 2016, 2016, 6584054. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Goel, A.N.; Gowda, B.S.; Veena, M.S.; Shiba, T.L.; Long, J.L. Adipose-Derived Mesenchymal Stromal Cells Persist in Tissue-Engineered Vocal Fold Replacement in Rabbits. Ann. Otol. Rhinol. Laryngol. 2018, 127, 962–968. [Google Scholar] [CrossRef]
  91. Hiwatashi, N.; Hirano, S.; Suzuki, R.; Kawai, Y.; Mizuta, M.; Kishimoto, Y.; Tateya, I.; Kanemaru, S.-I.; Nakamura, T.; Dezawa, M.; et al. Comparison of ASCs and BMSCs combined with atelocollagen for vocal fold scar regeneration. Laryngoscope 2016, 126, 1143–1150. [Google Scholar] [CrossRef] [PubMed]
  92. Bartlett, R.S.; Gaston, J.D.; Ye, S.; Kendziorski, C.; Thibeault, S.L. Mechanotransduction of vocal fold fibroblasts and mesenchymal stromal cells in the context of the vocal fold mechanome. J. Biomech. 2019, 83, 227–234. [Google Scholar] [CrossRef]
  93. Hiwatashi, N.; Bing, R.; Kraja, I.; Branski, R.C. Mesenchymal stem cells have antifibrotic effects on transforming growth factor-β1-stimulated vocal fold fibroblasts. Laryngoscope 2017, 127, E35–E41. [Google Scholar] [CrossRef] [Green Version]
  94. Strioga, M.; Viswanathan, S.; Darinskas, A.; Slaby, O.; Michalek, J. Same or not the same? Comparison of adipose tissue-derived versus bone marrow-derived mesenchymal stem and stromal cells. Stem Cells Dev. 2012, 21, 2724–2752. [Google Scholar] [CrossRef] [PubMed]
  95. Zhou, W.; Lin, J.; Zhao, K.; Jin, K.; He, Q.; Hu, Y.; Feng, G.; Cai, Y.; Xia, C.; Liu, H.; et al. Single-Cell Profiles and Clinically Useful Properties of Human Mesenchymal Stem Cells of Adipose and Bone Marrow Origin. Am. J. Sports Med. 2019, 47, 1722–1733. [Google Scholar] [CrossRef]
  96. Bacakova, L.; Zarubova, J.; Travnickova, M.; Musilkova, J.; Pajorova, J.; Slepicka, P.; Kasalkova, N.S.; Svorcik, V.; Kolska, Z.; Motarjemi, H.; et al. Stem cells: Their source, potency and use in regenerative therapies with focus on adipose-derived stem cells—A review. Biotechnol. Adv. 2018, 36, 1111–1126. [Google Scholar] [CrossRef]
  97. Si, Z.; Wang, X.; Sun, C.; Kang, Y.; Xu, J.; Wang, X.; Hui, Y. Adipose-derived stem cells: Sources, potency, and implications for regenerative therapies. Biomed. Pharmacother. 2019, 114, 108765. [Google Scholar] [CrossRef]
  98. Nagamura-Inoue, T.; He, H. Umbilical cord-derived mesenchymal stem cells: Their advantages and potential clinical utility. World J. Stem Cells 2014, 6, 195–202. [Google Scholar] [CrossRef]
  99. Ding, D.-C.; Chang, Y.-H.; Shyu, W.-C.; Lin, S.-Z. Human umbilical cord mesenchymal stem cells: A new era for stem cell therapy. Cell Transplant. 2015, 24, 339–347. [Google Scholar] [CrossRef]
  100. Pan, Y.; Jiao, G.; Yang, J.; Guo, R.; Li, J.; Wang, C. Insights into the Therapeutic Potential of Heparinized Collagen Scaffolds Loading Human Umbilical Cord Mesenchymal Stem Cells and Nerve Growth Factor for the Repair of Recurrent Laryngeal Nerve Injury. Tissue Eng. Regen. Med. 2017, 14, 317–326. [Google Scholar] [CrossRef]
  101. Li, N.; Hua, J. Interactions between mesenchymal stem cells and the immune system. Cell. Mol. Life Sci. 2017, 74, 2345–2360. [Google Scholar] [CrossRef]
  102. Han, K.-H.; Kim, A.-K.; Kim, M.-H.; Kim, D.-H.; Go, H.-N.; Kim, D.-I. Enhancement of angiogenic effects by hypoxia-preconditioned human umbilical cord-derived mesenchymal stem cells in a mouse model of hindlimb ischemia. Cell Biol. Int. 2016, 40, 27–35. [Google Scholar] [CrossRef]
  103. 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]
  104. Li, J.; Gao, F.; Ma, S.; Zhang, Y.; Zhang, J.; Guan, F.; Yao, M. Control the fate of human umbilical cord mesenchymal stem cells with dual-enzymatically cross-linked gelatin hydrogels for potential applications in nerve regeneration. J. Tissue Eng. Regen. Med. 2020, 14, 1261–1271. [Google Scholar] [CrossRef] [PubMed]
  105. Li, Y.; Yang, J.; Fu, G.; Zhou, P.; Liu, Y.; Li, Z.; Jiao, G. Human umbilical cord mesenchymal stem cells differentiate into neuron-like cells after induction with B27-supplemented serum-free medium. J. South. Med. Univ. 2020, 40, 1340–1345. [Google Scholar] [CrossRef]
  106. Guo, Z.-Y.; Sun, X.; Xu, X.-L.; Zhao, Q.; Peng, J.; Wang, Y. Human umbilical cord mesenchymal stem cells promote peripheral nerve repair via paracrine mechanisms. Neural Regen. Res. 2015, 10, 651–658. [Google Scholar] [CrossRef] [PubMed]
  107. DIrja, B.T.; Yoshie, S.; Ikeda, M.; Imaizumi, M.; Nakamura, R.; Otsuki, K.; Nomoto, Y.; Wada, I.; Hazama, A.; Omori, K. Potential of laryngeal muscle regeneration using induced pluripotent stem cell-derived skeletal muscle cells. Acta Otolaryngol. 2016, 136, 391–396. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Imaizumi, M.; Li-Jessen, N.Y.K.; Sato, Y.; Yang, D.T.; Thibeault, S.L. Retention of Human-Induced Pluripotent Stem Cells (hiPS) With Injectable HA Hydrogels for Vocal Fold Engineering. Ann. Otol. Rhinol. Laryngol. 2017, 126, 304–314. [Google Scholar] [CrossRef]
  109. Imaizumi, M.; Sato, Y.; Yang, D.T.; Thibeault, S.L. In vitro epithelial differentiation of human induced pluripotent stem cells for vocal fold tissue engineering. Ann. Otol. Rhinol. Laryngol. 2013, 122, 737–747. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. Huang, Z.; Powell, R.; Phillips, J.B.; Haastert-Talini, K. Perspective on Schwann Cells Derived from Induced Pluripotent Stem Cells in Peripheral Nerve Tissue Engineering. Cells 2020, 9, 2497. [Google Scholar] [CrossRef] [PubMed]
  111. Rao, L.; Qian, Y.; Khodabukus, A.; Ribar, T.; Bursac, N. Engineering human pluripotent stem cells into a functional skeletal muscle tissue. Nat. Commun. 2018, 9, 126. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Aboul-Soud, M.A.M.; Alzahrani, A.J.; Mahmoud, A. Induced Pluripotent Stem Cells (iPSCs)-Roles in Regenerative Therapies, Disease Modelling and Drug Screening. Cells 2021, 10, 2319. [Google Scholar] [CrossRef]
  113. Mora-Navarro, C.; Badileanu, A.; Gracioso Martins, A.M.; Ozpinar, E.W.; Gaffney, L.; Huntress, I.; Harrell, E.; Enders, J.R.; Peng, X.; Branski, R.C.; et al. Porcine Vocal Fold Lamina Propria-Derived Biomaterials Modulate TGF-β1-Mediated Fibroblast Activation in Vitro. ACS Biomater. Sci. Eng. 2020, 6, 1690–1703. [Google Scholar] [CrossRef] [PubMed]
  114. Leydon, C.; Imaizumi, M.; Bartlett, R.S.; Wang, S.F.; Thibeault, S.L. Epithelial cells are active participants in vocal fold wound healing: An in vivo animal model of injury. PLoS ONE 2014, 9, e115389. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Suzuki, R.; Katsuno, T.; Kishimoto, Y.; Nakamura, R.; Mizuta, M.; Suehiro, A.; Yamashita, M.; Nakamura, T.; Tateya, I.; Omori, K. Process of tight junction recovery in the injured vocal fold epithelium: Morphological and paracellular permeability analysis. Laryngoscope 2018, 128, E150–E156. [Google Scholar] [CrossRef]
  116. Branco, A.; Todorovic Fabro, A.; Gonçalves, T.M.; Garcia Martins, R.H. Alterations in extracellular matrix composition in the aging larynx. Otolaryngol. Head Neck Surg. 2015, 152, 302–307. [Google Scholar] [CrossRef]
  117. Caballero Aguilar, L.M.; Silva, S.M.; Moulton, S.E. Growth factor delivery: Defining the next generation platforms for tissue engineering. J. Control. Release 2019, 306, 40–58. [Google Scholar] [CrossRef]
  118. Buie, T.; McCune, J.; Cosgriff-Hernandez, E. Gelatin Matrices for Growth Factor Sequestration. Trends Biotechnol. 2020, 38, 546–557. [Google Scholar] [CrossRef]
  119. Sarrigiannidis, S.O.; Rey, J.M.; Dobre, O.; González-García, C.; Dalby, M.J.; Salmeron-Sanchez, M. A tough act to follow: Collagen hydrogel modifications to improve mechanical and growth factor loading capabilities. Mater. Today Bio 2021, 10, 100098. [Google Scholar] [CrossRef] [PubMed]
  120. 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] [PubMed]
  121. Li, R.; Li, Y.; Wu, Y.; Zhao, Y.; Chen, H.; Yuan, Y.; Xu, K.; Zhang, H.; Lu, Y.; Wang, J.; et al. Heparin-Poloxamer Thermosensitive Hydrogel Loaded with bFGF and NGF Enhances Peripheral Nerve Regeneration in Diabetic Rats. Biomaterials 2018, 168, 24–37. [Google Scholar] [CrossRef] [PubMed]
  122. Subbiah, R.; Ruehle, M.A.; Klosterhoff, B.S.; Lin, A.S.P.; Hettiaratchi, M.H.; Willett, N.J.; Bertassoni, L.E.; García, A.J.; Guldberg, R.E. Triple growth factor delivery promotes functional bone regeneration following composite musculoskeletal trauma. Acta Biomater. 2021, 127, 180–192. [Google Scholar] [CrossRef]
  123. Ma, S.; Zhou, J.; Huang, T.; Zhang, Z.; Xing, Q.; Zhou, X.; Zhang, K.; Yao, M.; Cheng, T.; Wang, X.; et al. Sodium alginate/collagen/stromal cell-derived factor-1 neural scaffold loaded with BMSCs promotes neurological function recovery after traumatic brain injury. Acta Biomater. 2021, 131, 185–197. [Google Scholar] [CrossRef] [PubMed]
  124. Said, M.F.; Islam, A.A.; Massi, M.N. Prihantono Effect of erythropoietin administration on the expression of brain-derived neurotrophic factor, stromal cell-derived Factor-1, and neuron-specific enolase in traumatic brain injury: A literature review. Ann. Med. Surg. 2021, 69, 102666. [Google Scholar] [CrossRef] [PubMed]
  125. Lee, N.; Spearry, R.P.; Rydyznski, C.E.; MacLennan, A.J. Muscle ciliary neurotrophic factor receptor α contributes to motor neuron STAT3 activation following peripheral nerve lesion. Eur. J. Neurosci. 2019, 49, 1084–1090. [Google Scholar] [CrossRef] [PubMed]
  126. Takeharu, K.; Kurakami, K.; Konomi, U.; Komazawa, D.; Misawa, K.; Imayoshi, S.; Watanabe, Y. Safety and short-term outcomes of basic fibroblast growth factor injection for sulcus vocalis. Acta Otolaryngol. 2018, 138, 1014–1019. [Google Scholar] [CrossRef]
  127. Hirano, S.; Tateya, I.; Kishimoto, Y.; Kanemaru, S.; Ito, J. Clinical trial of regeneration of aged vocal folds with growth factor therapy. Laryngoscope 2012, 122, 327–331. [Google Scholar] [CrossRef]
  128. Kanazawa, T.; Komazawa, D.; Indo, K.; Akagi, Y.; Lee, Y.; Nakamura, K.; Matsushima, K.; Kunieda, C.; Misawa, K.; Nishino, H.; et al. Single injection of basic fibroblast growth factor to treat severe vocal fold lesions and vocal fold paralysis. Laryngoscope 2015, 125, E338–E344. [Google Scholar] [CrossRef]
  129. Sueyoshi, S.; Umeno, H.; Kurita, T.; Fukahori, M.; Chitose, S.-I. Long-term outcomes of basic fibroblast growth factor treatments in patients with vocal fold scarring, aged vocal fold, and sulcus vocalis. Auris Nasus Larynx 2021, 48, 949–955. [Google Scholar] [CrossRef] [PubMed]
  130. Okui, A.; Konomi, U.; Kanazawa, T.; Komazawa, D.; Nakamura, K.; Matsushima, K.; Watanabe, Y. Therapeutic Efficacy of Basic Fibroblast Growth Factor in Patients With Vocal Fold Atrophy. Laryngoscope 2020, 130, 2847–2852. [Google Scholar] [CrossRef] [PubMed]
  131. Hirano, S.; Kawamoto, A.; Tateya, I.; Mizuta, M.; Kishimoto, Y.; Hiwatashi, N.; Kawai, Y.; Tsuji, T.; Suzuki, R.; Kaneko, M.; et al. A phase I/II exploratory clinical trial for intracordal injection of recombinant hepatocyte growth factor for vocal fold scar and sulcus. J. Tissue Eng. Regen. Med. 2018, 12, 1031–1038. [Google Scholar] [CrossRef] [PubMed]
  132. Choi, J.-S.; Heang Oh, S.; Kim, Y.-M.; Lim, J.-Y. Hyaluronic Acid/Alginate Hydrogel Containing Hepatocyte Growth Factor and Promotion of Vocal Fold Wound Healing. Tissue Eng. Regen. Med. 2020, 17, 651–658. [Google Scholar] [CrossRef]
  133. Dias Garcia, R.I.; Tsuji, D.H.; Imamura, R.; Mauad, T.; Ferraz da Silva, L.F. Effects of hepatocyte growth factor injection and reinjection on healing in the rabbit vocal fold. J. Voice 2012, 26, e7–e12. [Google Scholar] [CrossRef]
  134. Minardi, S.; Pandolfi, L.; Taraballi, F.; Wang, X.; De Rosa, E.; Mills, Z.D.; Liu, X.; Ferrari, M.; Tasciotti, E. Enhancing Vascularization through the Controlled Release of Platelet-Derived Growth Factor-BB. ACS Appl. Mater. Interfaces 2017, 9, 14566–14575. [Google Scholar] [CrossRef]
  135. Pan, S.-C.; Lee, C.-H.; Chen, C.-L.; Fang, W.-Y.; Wu, L.-W. Angiogenin Attenuates Scar Formation in Burn Patients by Reducing Fibroblast Proliferation and Transforming Growth Factor β1 Secretion. Ann. Plast. Surg. 2018, 80, S79–S83. [Google Scholar] [CrossRef] [PubMed]
  136. Li, X.W.; Sun, H.C.; Liu, X.H. Vascular endothelial growth factor-loaded microspheres promote dental pulp regeneration and vascularization. Zhonghua Kou Qiang Yi Xue Za Zhi 2018, 53, 42–48. [Google Scholar] [CrossRef] [PubMed]
  137. Zhu, L.; Dissanayaka, W.L.; Zhang, C. Dental pulp stem cells overexpressing stromal-derived factor-1α and vascular endothelial growth factor in dental pulp regeneration. Clin. Oral Investig. 2019, 23, 2497–2509. [Google Scholar] [CrossRef] [PubMed]
  138. Räsänen, M.; Sultan, I.; Paech, J.; Hemanthakumar, K.A.; Yu, W.; He, L.; Tang, J.; Sun, Y.; Hlushchuk, R.; Huan, X.; et al. VEGF-B Promotes Endocardium-Derived Coronary Vessel Development and Cardiac Regeneration. Circulation 2021, 143, 65–77. [Google Scholar] [CrossRef] [PubMed]
  139. Rao, F.; Wang, Y.; Zhang, D.; Lu, C.; Cao, Z.; Sui, J.; Wu, M.; Zhang, Y.; Pi, W.; Wang, B.; et al. Aligned chitosan nanofiber hydrogel grafted with peptides mimicking bioactive brain-derived neurotrophic factor and vascular endothelial growth factor repair long-distance sciatic nerve defects in rats. Theranostics 2020, 10, 1590–1603. [Google Scholar] [CrossRef]
  140. Hu, K.; Olsen, B.R. Vascular endothelial growth factor control mechanisms in skeletal growth and repair. Dev. Dyn. 2017, 246, 227–234. [Google Scholar] [CrossRef] [Green Version]
  141. Gnavi, S.; di Blasio, L.; Tonda-Turo, C.; Mancardi, A.; Primo, L.; Ciardelli, G.; Gambarotta, G.; Geuna, S.; Perroteau, I. Gelatin-based hydrogel for vascular endothelial growth factor release in peripheral nerve tissue engineering. J. Tissue Eng. Regen. Med. 2017, 11, 459–470. [Google Scholar] [CrossRef] [Green Version]
  142. Yoshida, T.; Delafontaine, P. Mechanisms of IGF-1-Mediated Regulation of Skeletal Muscle Hypertrophy and Atrophy. Cells 2020, 9, 1970. [Google Scholar] [CrossRef]
  143. Baht, G.S.; Bareja, A.; Lee, D.E.; Rao, R.R.; Huang, R.; Huebner, J.L.; Bartlett, D.B.; Hart, C.R.; Gibson, J.R.; Lanza, I.R.; et al. Meteorin-like facilitates skeletal muscle repair through a Stat3/IGF-1 mechanism. Nat. Metab. 2020, 2, 278–289. [Google Scholar] [CrossRef] [Green Version]
  144. Yaghoubi, Y.; Movassaghpour, A.; Zamani, M.; Talebi, M.; Mehdizadeh, A.; Yousefi, M. Human umbilical cord mesenchymal stem cells derived-exosomes in diseases treatment. Life Sci. 2019, 233, 116733. [Google Scholar] [CrossRef] [PubMed]
  145. Abbaszadeh, H.; Ghorbani, F.; Derakhshani, M.; Movassaghpour, A.; Yousefi, M. Human umbilical cord mesenchymal stem cell-derived extracellular vesicles: A novel therapeutic paradigm. J. Cell. Physiol. 2020, 235, 706–717. [Google Scholar] [CrossRef]
  146. Ma, Y.; Dong, L.; Zhou, D.; Li, L.; Zhang, W.; Zhen, Y.; Wang, T.; Su, J.; Chen, D.; Mao, C.; et al. Extracellular vesicles from human umbilical cord mesenchymal stem cells improve nerve regeneration after sciatic nerve transection in rats. J. Cell. Mol. Med. 2019, 23, 2822–2835. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Wang, W.; Li, M.; Chen, Z.; Xu, L.; Chang, M.; Wang, K.; Deng, C.; Gu, Y.; Zhou, S.; Shen, Y.; et al. Biogenesis and function of extracellular vesicles in pathophysiological processes of skeletal muscle atrophy. Biochem. Pharmacol. 2022, 198, 114954. [Google Scholar] [CrossRef] [PubMed]
  148. Vechetti, I.J.J.; Valentino, T.; Mobley, C.B.; McCarthy, J.J. The role of extracellular vesicles in skeletal muscle and systematic adaptation to exercise. J. Physiol. 2021, 599, 845–861. [Google Scholar] [CrossRef]
  149. Upadhya, R.; Zingg, W.; Shetty, S.; Shetty, A.K. Astrocyte-derived extracellular vesicles: Neuroreparative properties and role in the pathogenesis of neurodegenerative disorders. J. Control. Release 2020, 323, 225–239. [Google Scholar] [CrossRef]
  150. Delpech, J.-C.; Herron, S.; Botros, M.B.; Ikezu, T. Neuroimmune Crosstalk through Extracellular Vesicles in Health and Disease. Trends Neurosci. 2019, 42, 361–372. [Google Scholar] [CrossRef]
  151. Andjus, P.; Kosanović, M.; Milićević, K.; Gautam, M.; Vainio, S.J.; Jagečić, D.; Kozlova, E.N.; Pivoriūnas, A.; Chachques, J.-C.; Sakaj, M.; et al. Extracellular Vesicles as Innovative Tool for Diagnosis, Regeneration and Protection against Neurological Damage. Int. J. Mol. Sci. 2020, 21, 6859. [Google Scholar] [CrossRef] [PubMed]
  152. Woo, P.; Murry, T. Short-Term Voice Improvement after Repeated Office-Based Platelet-Rich Plasma PRP Injection in Patients with Vocal Fold Scar, Sulcus, and Atrophy. J. Voice 2021. [Google Scholar] [CrossRef]
  153. Woo, S.H.; Jeong, H.-S.; Kim, J.P.; Koh, E.-H.; Lee, S.U.; Jin, S.M.; Kim, D.H.; Sohn, J.H.; Lee, S.H. Favorable vocal fold wound healing induced by platelet-rich plasma injection. Clin. Exp. Otorhinolaryngol. 2014, 7, 47–52. [Google Scholar] [CrossRef] [PubMed]
  154. Cobden, S.B.; Oztürk, K.; Duman, S.; Esen, H.; Aktan, T.M.; Avunduk, M.C.; Elsurer, C. Treatment of Acute Vocal Fold Injury With Platelet-Rich Plasma. J. Voice 2016, 30, 731–735. [Google Scholar] [CrossRef] [PubMed]
  155. Tang, R.; Xu, Z. Gene therapy: A double-edged sword with great powers. Mol. Cell. Biochem. 2020, 474, 73–81. [Google Scholar] [CrossRef] [PubMed]
  156. Anguela, X.M.; High, K.A. Entering the Modern Era of Gene Therapy. Annu. Rev. Med. 2019, 70, 273–288. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Nelson, C.E.; Kim, A.J.; Adolph, E.J.; Gupta, M.K.; Yu, F.; Hocking, K.M.; Davidson, J.M.; Guelcher, S.A.; Duvall, C.L. Tunable delivery of siRNA from a biodegradable scaffold to promote angiogenesis in vivo. Adv. Mater. 2014, 26, 607–614. [Google Scholar] [CrossRef] [Green Version]
  158. Fan, Q.; Mao, H.; Xie, L.; Pi, X. Prolyl Hydroxylase Domain-2 Protein Regulates Lipopolysaccharide-Induced Vascular Inflammation. Am. J. Pathol. 2019, 189, 200–213. [Google Scholar] [CrossRef] [PubMed]
  159. Liu, Y.; Zhao, N.; Xu, F.-J. pH-Responsive Degradable Dextran-Quantum Dot Nanohybrids for Enhanced Gene Delivery. ACS Appl. Mater. Interfaces 2019, 11, 34707–34716. [Google Scholar] [CrossRef]
  160. Lara-Velazquez, M.; Alkharboosh, R.; Norton, E.S.; Ramirez-Loera, C.; Freeman, W.D.; Guerrero-Cazares, H.; Forte, A.J.; Quiñones-Hinojosa, A.; Sarabia-Estrada, R. Chitosan-Based Non-viral Gene and Drug Delivery Systems for Brain Cancer. Front. Neurol. 2020, 11, 740. [Google Scholar] [CrossRef] [PubMed]
  161. Kolosova, K.; Gao, Q.; Tuznik, M.; Bouhabel, S.; Kost, K.M.; Wang, H.; Li-Jessen, N.Y.K.; Mongeau, L.; Wiseman, P.W. Characterizing Vocal Fold Injury Recovery in a Rabbit Model With Three-Dimensional Virtual Histology. Laryngoscope 2021, 131, 1578–1587. [Google Scholar] [CrossRef] [PubMed]
  162. Mattei, A.; Magalon, J.; Velier, M.; Dignat-George, F.; Giovanni, A.; Sabatier, F. Commentary about mesenchymal stem cells and scarred vocal folds. Stem Cell Res. Ther. 2020, 11, 173. [Google Scholar] [CrossRef] [PubMed]
  163. Abbaszadeh, H.; Ghorbani, F.; Derakhshani, M.; Movassaghpour, A.A.; Yousefi, M.; Talebi, M.; Shamsasenjan, K. Regenerative potential of Wharton’s jelly-derived mesenchymal stem cells: A new horizon of stem cell therapy. J. Cell. Physiol. 2020, 235, 9230–9240. [Google Scholar] [CrossRef]
  164. Pappas, J.J.; Yang, P.C. Human ESC vs. iPSC—Pros and Cons. J. Cardiovasc. Transl. Res. 2008, 1, 96–99. [Google Scholar] [CrossRef] [PubMed]
  165. Halevy, T.; Urbach, A. Comparing ESC and iPSC-Based Models for Human Genetic Disorders. J. Clin. Med. 2014, 3, 1146–1162. [Google Scholar] [CrossRef] [Green Version]
  166. Kaboodkhani, R.; Mehrabani, D.; Karimi-Busheri, F. Achievements and Challenges in Transplantation of Mesenchymal Stem Cells in Otorhinolaryngology. J. Clin. Med. 2021, 10, 2940. [Google Scholar] [CrossRef] [PubMed]
  167. Ntege, E.H.; Sunami, H.; Shimizu, Y. Advances in regenerative therapy: A review of the literature and future directions. Regen. Ther. 2020, 14, 136–153. [Google Scholar] [CrossRef] [PubMed]
  168. Christodoulou, I.; Kolisis, F.N.; Papaevangeliou, D.; Zoumpourlis, V. Comparative Evaluation of Human Mesenchymal Stem Cells of Fetal (Wharton’s Jelly) and Adult (Adipose Tissue) Origin during Prolonged In Vitro Expansion: Considerations for Cytotherapy. Stem Cells Int. 2013, 2013, 246134. [Google Scholar] [CrossRef]
  169. Suman, S.; Domingues, A.; Ratajczak, J.; Ratajczak, M.Z. Potential Clinical Applications of Stem Cells in Regenerative Medicine. Adv. Exp. Med. Biol. 2019, 1201, 1–22. [Google Scholar] [CrossRef] [PubMed]
  170. Shi, Y.; Inoue, H.; Wu, J.C.; Yamanaka, S. Induced pluripotent stem cell technology: A decade of progress. Nat. Rev. Drug Discov. 2017, 16, 115–130. [Google Scholar] [CrossRef]
  171. Su, H.; Cantrell, A.C.; Zeng, H.; Zhu, S.-H.; Chen, J.-X. Emerging Role of Pericytes and Their Secretome in the Heart. Cells 2021, 10, 548. [Google Scholar] [CrossRef] [PubMed]
  172. Shimizu, F.; Kanda, T. Pericytes of the Nervous System: Physiological and Pathological Role. Brain Nerve 2020, 72, 151–158. [Google Scholar] [CrossRef]
  173. Mannino, G.; Gennuso, F.; Giurdanella, G.; Conti, F.; Drago, F.; Salomone, S.; Furno, D.L.; Bucolo, C.; Giuffrida, R. Pericyte-like differentiation of human adipose-derived mesenchymal stem cells: An in vitro study. World J. Stem Cells 2020, 12, 1152–1170. [Google Scholar] [CrossRef]
  174. Guimarães-Camboa, N.; Cattaneo, P.; Sun, Y.; Moore-Morris, T.; Gu, Y.; Dalton, N.D.; Rockenstein, E.; Masliah, E.; Peterson, K.L.; Stallcup, W.B.; et al. Pericytes of Multiple Organs Do Not Behave as Mesenchymal Stem Cells In Vivo. Cell Stem Cell 2017, 20, 345–359.e5. [Google Scholar] [CrossRef] [Green Version]
  175. Choi, Y.H.; Kim, S.H.; Kim, I.G.; Lee, J.H.; Kwon, S.K. Injectable basic fibroblast growth factor-loaded alginate/hyaluronic acid hydrogel for rejuvenation of geriatric larynx. Acta Biomater. 2019, 89, 104–114. [Google Scholar] [CrossRef] [PubMed]
  176. Wiklander, O.P.B.; Brennan, M.Á.; Lötvall, J.; Breakefield, X.O.; El Andaloussi, S. Advances in therapeutic applications of extracellular vesicles. Sci. Transl. Med. 2019, 11, 492. [Google Scholar] [CrossRef]
  177. Tang, Y.; Zhou, Y.; Li, H.-J. Advances in mesenchymal stem cell exosomes: A review. Stem Cell Res. Ther. 2021, 12, 71. [Google Scholar] [CrossRef]
  178. Zhao, A.G.; Shah, K.; Cromer, B.; Sumer, H. Mesenchymal Stem Cell-Derived Extracellular Vesicles and Their Therapeutic Potential. Stem Cells Int. 2020, 2020, 8825771. [Google Scholar] [CrossRef]
  179. Tsiapalis, D.; O’Driscoll, L. Mesenchymal Stem Cell Derived Extracellular Vesicles for Tissue Engineering and Regenerative Medicine Applications. Cells 2020, 9, 991. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  180. Alqurashi, H.; Ortega Asencio, I.; Lambert, D.W. The Emerging Potential of Extracellular Vesicles in Cell-Free Tissue Engineering and Regenerative Medicine. Tissue Eng. Part B Rev. 2021, 27, 530–538. [Google Scholar] [CrossRef]
  181. Hirano, S.; Sugiyama, Y.; Kaneko, M.; Mukudai, S.; Fuse, S.; Hashimoto, K. Intracordal Injection of Basic Fibroblast Growth Factor in 100 Cases of Vocal Fold Atrophy and Scar. Laryngoscope 2021, 131, 2059–2064. [Google Scholar] [CrossRef] [PubMed]
  182. Kim, I.G.; Park, M.R.; Choi, Y.H.; Choi, J.S.; Ahn, H.J.; Kwon, S.K.; Lee, J.H. Regeneration of Paralyzed Vocal Fold by the Injection of Plasmid DNA Complex-Loaded Hydrogel Bulking Agent. ACS Biomater. Sci. Eng. 2019, 5, 1497–1508. [Google Scholar] [CrossRef] [PubMed]
  183. Choi, Y.H.; Ahn, H.J.; Park, M.R.; Han, M.J.; Lee, J.H.; Kwon, S.K. Dual growth factor-immobilized bioactive injection material for enhanced treatment of glottal insufficiency. Acta Biomater. 2019, 86, 269–279. [Google Scholar] [CrossRef] [PubMed]
  184. Wang, F.; Li, Z.; Khan, M.; Tamama, K.; Kuppusamy, P.; Wagner, W.R.; Sen, C.K.; Guan, J. Injectable, rapid gelling and highly flexible hydrogel composites as growth factor and cell carriers. Acta Biomater. 2010, 6, 1978–1991. [Google Scholar] [CrossRef] [PubMed]
  185. Raimondo, T.M.; Li, H.; Kwee, B.J.; Kinsley, S.; Budina, E.; Anderson, E.M.; Doherty, E.J.; Talbot, S.G.; Mooney, D.J. Combined delivery of VEGF and IGF-1 promotes functional innervation in mice and improves muscle transplantation in rabbits. Biomaterials 2019, 216, 119246. [Google Scholar] [CrossRef] [PubMed]
  186. Zhou, Y.; Liu, S.; Zhao, M.; Wang, C.; Li, L.; Yuan, Y.; Li, L.; Liao, G.; Bresette, W.; Zhang, J.; et al. Injectable extracellular vesicle-released self-assembling peptide nanofiber hydrogel as an enhanced cell-free therapy for tissue regeneration. J. Control. Release 2019, 316, 93–104. [Google Scholar] [CrossRef]
Figure 1. Structure of human vocal fold. Created with BioRender.com (accessed on 24 October 2022).
Figure 1. Structure of human vocal fold. Created with BioRender.com (accessed on 24 October 2022).
Biomedicines 10 03082 g001
Figure 2. Coronal section showing a comparison between normal condition and glottic insufficiency.
Figure 2. Coronal section showing a comparison between normal condition and glottic insufficiency.
Biomedicines 10 03082 g002
Figure 3. A comparison between direct injection of cells and biomolecules versus injection of cells and biomolecules encapsulated in a hydrogel.
Figure 3. A comparison between direct injection of cells and biomolecules versus injection of cells and biomolecules encapsulated in a hydrogel.
Biomedicines 10 03082 g003
Figure 4. Hydrogel protects cells during injection.
Figure 4. Hydrogel protects cells during injection.
Biomedicines 10 03082 g004
Figure 5. The anti-fibrosis function of ASCs.
Figure 5. The anti-fibrosis function of ASCs.
Biomedicines 10 03082 g005
Figure 6. WJMSC functions. (a) The function of WJMSCs in neuronal and muscular regeneration. (b) Immunomodulatory properties of WJMSCs in preventing fibrosis.
Figure 6. WJMSC functions. (a) The function of WJMSCs in neuronal and muscular regeneration. (b) Immunomodulatory properties of WJMSCs in preventing fibrosis.
Biomedicines 10 03082 g006
Figure 7. Function of EVs in neuron and muscle regeneration.
Figure 7. Function of EVs in neuron and muscle regeneration.
Biomedicines 10 03082 g007
Figure 8. Therapeutic effect of biomolecules on vocal fold regeneration. Created with Biorender.com (accessed on 24 October 2022).
Figure 8. Therapeutic effect of biomolecules on vocal fold regeneration. Created with Biorender.com (accessed on 24 October 2022).
Biomedicines 10 03082 g008
Table 1. Study outcome and limitation of recent studies on vocal fold injection for regeneration purposes.
Table 1. Study outcome and limitation of recent studies on vocal fold injection for regeneration purposes.
No.Type of EncapsulationType of TreatmentStudy Design/Study OutcomeClinicalTrials.gov Number (accessed on 23 October 2022)/Reference
1Autologous BMMSCsEncapsulation with hyaluronan gelPilot study.
Scarred vocal fold is improved.
NCT01981330
Direct injectionClinical trial phase 1/2.NCT04290182
Limitation: Difficult to source and expand for clinical application.
2Autologous ASCsEncapsulated with injectable collagen scaffoldClinical trial phase 2.NCT04164485
Direct injectionClinical trial phase 1/2.NCT02904824
Direct injectionImproved ECM regeneration in rat model.[88]
Direct injectionClinical trial.
Overall voice outcome was improved.
[84,85]
Limitation: Direct injection yielded short cell retention in providing regenerative effect. It had slower cell proliferation and lesser immunophenotypic indicators than WJMSCs.
3bFGFDirect injectionClinical trial.
Overall voice outcome was improved.
[126,127,128,181]
Limitation: Single injection was insufficient to obtain satisfactory improvement.
4HGFDirect injectionClinical trial.
Overall voice outcome was improved.
[131]
Encapsulated with injectable HA/ALG scaffoldHGF in HA/ALG had greater sustained release than direct injection in rabbit model.[132]
Direct injectionRe-injection of HGF in rabbit with injured vocal fold reduced collagen expression more significantly.[133]
Limitation: Direct injection of HGF had limited retention time for regenerative effect.
4PRP & autologous fatDirect injectionClinical trial phase 4NCT04839276
PRPDirect injectionN/ANCT03749863
Limitation: Applied in short augmentation purpose.
5Autologous fibroblastDirect injectionClinical trial phase 2NCT02120781
Limitation: Difficulty in sourcing available dermal fibroblast for treatment and possible delayed treatment.
6Plasmic DNA (pDNA)Encapsulated in injectable ALG/HA with PCL microspheres Collagen and HA composition were improved in rabbit with injured vocal fold.[182]
Limitation: Complicated components in building suitable hydrogel for pDNA.
Table 2. Cells or biomolecules which had potential for hydrogel encapsulation for vocal fold injection (based on literature review).
Table 2. Cells or biomolecules which had potential for hydrogel encapsulation for vocal fold injection (based on literature review).
No.Type of EncapsulationType of TreatmentStudy Design/Study OutcomeReference
1Human umbilical cord WJMSCs with NGFEncapsulated in heparinized collagen scaffoldDamaged RLN regenerated in in vivo (rabbit)
Scaffold with WJMSCs/NGF had better histomorphological outcome than no
WJMSCs/NGF or alone.
[100]
2iPSCsDirect injectioniPSCs able to differentiate into skeletal muscle tissue and implanted in thyroarytenoid muscle of rat model.
More work needed to ensure safety of iPSCs.
[111]
Encapsulated in HA hydrogel with EGFHydrogel with iPSCs and EGF had less fibrosis in injured vocal fold cells in vitro & rat model.[108,109]
3bFGF & HGFEncapsulated in polycaprolactone (PCL)/pluronic F127 microspheresSustained release of bFGF and HGF reduced muscle degeneration and increased muscle regeneration in injured vocal fold of rabbit model.[183]
4bFGFEncapsulated in gelatin microsphereScarred formation was reduced in rabbit model.
Long term study and inflammation study needed for future study.
[70]
5VEGFEncapsulated in microsphereImproved dental pulp regeneration in mice model.[136]
6BDNF & VEGFEncapsulated in chitosan nanofiber hydrogelHydrogel with VEGF provided microenvironment and improved nerve regeneration in rat model.[139]
7IGF-1 & MSCEncapsulated in thermosensitive type 1 collagenRelease of IGF-1 was sustained (2 weeks) and improved MSCs cell proliferation in the hydrogel.[184]
8IGF-1 & VEGFEncapsulated in alginate hydrogelRelease of IGF-1 and VEGF were sustained and improved muscle function in mice and rabbit models.[185]
9EVsSource: WJMSCs
Direct injection
Improved nerve regeneration in rat model.[146]
Source: BMMSCs
Encapsulated in matrix metalloproteinase-2 sensitive self- assembling peptide
Sustained release of EVs in hydrogel and had better outcome of renal function in mice model than direct injection.[186]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Ng, W.-C.; Lokanathan, Y.; Baki, M.M.; Fauzi, M.B.; Zainuddin, A.A.; Azman, M. Tissue Engineering as a Promising Treatment for Glottic Insufficiency: A Review on Biomolecules and Cell-Laden Hydrogel. Biomedicines 2022, 10, 3082. https://doi.org/10.3390/biomedicines10123082

AMA Style

Ng W-C, Lokanathan Y, Baki MM, Fauzi MB, Zainuddin AA, Azman M. Tissue Engineering as a Promising Treatment for Glottic Insufficiency: A Review on Biomolecules and Cell-Laden Hydrogel. Biomedicines. 2022; 10(12):3082. https://doi.org/10.3390/biomedicines10123082

Chicago/Turabian Style

Ng, Wan-Chiew, Yogeswaran Lokanathan, Marina Mat Baki, Mh Busra Fauzi, Ani Amelia Zainuddin, and Mawaddah Azman. 2022. "Tissue Engineering as a Promising Treatment for Glottic Insufficiency: A Review on Biomolecules and Cell-Laden Hydrogel" Biomedicines 10, no. 12: 3082. https://doi.org/10.3390/biomedicines10123082

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