This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
1. Introduction
Muscle stem cells, also called satellite cells (SCs) and defined by the transcription factor paired box 7 (Pax7), are responsible for skeletal muscle maintenance and repair. Once activated after injury, SCs enter the cell cycle, commit to myogenic progenitor cells (MPCs), differentiate into myoblast cells, and finally fuse into multinucleated myotubes. A subset of SCs undergoes asymmetric division, renewing the SC pool [1]. However, endogenous SCs may be inefficient in responding to severe trauma or chronic degenerative diseases such as malnutrition, neuromuscular diseases, and sleep apnea [2–4]. When more than 20% of the muscle volume is missing, as in volumetric muscle loss (VML), endogenous self-repair is hindered [5]. Moreover, SCs are found in very limited numbers in adult muscles and exhibit undesirable amplification activity in vitro [6]; thus, these SCs are not the appropriate stem cell pool for muscle repair.
Dental pulp stem cells (DPSCs) are a type of mesenchymal stem cells (MSCs) that exhibit therapeutic potential for tissue regeneration [7]. These cells are easily established from dental pulp [8] and have several advantages compared to other types of MSCs: better proliferative potential and migratory capabilities [9], a simpler primary isolation method, a higher success rate in long-term in vitro culture [10], and long-term storage without losing their stemness and ability to differentiate [11, 12]. Kerkis et al. [13] found the therapeutic effect of human DPSCs on Golden Retrievers with muscular dystrophy. Recently, DPSCs were shown to undergo myogenic differentiation (MyoD), and these myogenic lineage cells could treat the dystrophic muscles of mdx/SCID mice [14]. However, the current myogenic efficiency is still quite low [15]. Here, we searched for novel factors to develop appropriate MyoD systems for DPSCs.
MyoD is regulated by genes such as MyoD1, myogenic factor 5 (Myf5), myogenic regulatory factor 4 (MRF4), Desmin, and myosin heavy chain (MyHC) to determine cell fate, including myoblast differentiation into myocytes, fusion into myotubes, and maintenance of myofibers [15, 16]. Factors, molecules, or stimuli can be applied to affect MSCs’ fate and commitment [17], and biocompatible scaffolds are provided for the natural environment in the use of MSCs [12]. A recent RNA-seq analysis identified that repressed bone morphogenetic protein (BMP) and activated Wnt signaling pathways are required for nascent somites during embryonic myogenesis [18]. Wnt activation alone can induce pluripotent stem cells (PSCs) committed to myogenic progenitors. It further requires BMP inhibition to maintain progenitor fate, followed by the myogenic program to generate myofibers and associated Pax7+ cells [19]. These results suggest that Wnt activation or BMP antagonism might be important mechanisms in myogenic regulation.
Noggin, a secreted homodimeric glycoprotein, is promoted by Wnt-1 in medial somites and is required for embryonic somite and skeletal patterning [20, 21]. Then, Noggin was found to inhibit the actions of BMP-2, -4, -5, -7, -13, and -14, thus blocking Smad-dependent signaling [22]. BMP signaling has been shown to play a role in controlling SC lineage progression during embryonic myogenesis [23], and these SC progeny secrete BMP antagonists, such as Noggin, for the differentiation of muscle progenitor cells [24]. Noggin-null mice display defective skeletal muscle fibers [25]. However, the effect of Noggin on the MyoD of DPSCs has not yet been reported.
In the present study, Noggin was applied to MyoD systems for DPSCs. Then, we implanted these pretreated DPSCs combined with Matrigel as scaffolds into the mouse tibialis anterior muscle with VML. Here, we explore the DPSCs repair capacity in muscle injury.
2. Materials and Methods
2.1. Cell Culture and Identification
DPSCs were acquired as previously described (Figure 1a,b, Table S1) [26] and cultured in α-MEM (Gibco, NY, USA) with 10% fetal bovine serum (FBS, Gibco, NY, USA) and 1% penicillin-streptomycin (Gibco, NY, USA) at 37°C in the presence of 5% CO2 and 95% air. Flow cytometric analysis was used to determine the cell surface markers present on DPSCs. DPSCs were identified with phycoerythrin (PE)-conjugated antibodies against human CD29 (#555443) and FITC-conjugated antibodies against human CD90 (#555595) from BD Biosciences (CA, USA), fluorescein isothiocyanate (FITC)-conjugated antibodies against human CD44 (#11-0441-85) from eBioscience (CA, USA), and PE-conjugated antibodies against human CD34 (#343605) and FITC-conjugated antibodies against human CD45 (#304005) from BioLegend (CA, USA) and analyzed using FlowJo software (FlowJo, OR, USA). Cell cycle analysis was also performed after staining for 24 h with propidium iodide (PI) (Beyotime, Shanghai, China) according to the manufacturer’s protocol using flow cytometry (ACEA NovoCyte, CA, USA).
[figure(s) omitted; refer to PDF]
2.2. MyoD Assay
DPSCs were seeded on 6-well plates at a cell density of 4000 cells/cm2 in an expansion medium consisting of α-MEM with 10% FBS for adherence. When the confluence reached 80%, the culture medium was replaced first with a myogenic induction medium containing the following for 24 h: IMDM (Gibco, NY, USA) + 2% FBS + 1 μM 5-Aza-2′-deoxycytidine (5-Aza) (an important trigger for myogenic commitment differentiation [14]) (Sigma-Aldrich, CA, USA) [27]. IMDM + 2% FBS without 5-Aza served as the base control medium. Subsequently, cells were rinsed three times in phosphate-buffered saline (PBS, HyClone, UT, USA) and then transferred to the following differentiation medium: IMDM with 10% FBS (defined as Day 0) (Figure 1a). The differentiation medium was supplemented with or without Noggin (100 ng/mL = 100N or 200 ng/mL = 200N) or 50 ng/mL BMP4. Noggin (#10267) from Sino Biological (Beijing, China) and BMP4 (HZ-1045) from HumanZyme (IL, USA) were used. Cells were cultured for 3 weeks, the differentiation medium was changed every 3 days, and the cells were observed under a microscope to confirm the formation of myotubes.
2.3. VML Defect Modeling and Stem Cell Transplantation
Animal protocols were approved by Cyagen IACUC (No. ACU20-A034). Male Balb/C mice (8 weeks old) were purchased from Shanghai Bikai Biotechnology. All animals underwent surgery to create a muscle defect by unilaterally resecting the tibialis anterior muscle under 2% pentobarbital sodium anesthesia. The thin layer of fascia covering the tibialis anterior muscle was dissected away, and a 2 × 5 mm area was stained with hematoxylin. After a blunt separation, a 2 mm-deep cut was made around the stained hematine, creating a 2 × 5 × 2 mm defect. This VML defect accounted for a loss of ~40% of the tibialis anterior muscle. DPSCs were cultured and differentiated by 5-aza and Noggin treatment, and Matrigel (Corning, NY, USA) was used to provide a scaffold for stem cell transplantation. Matrigel alone (n = 6) or Matrigel combined with Noggin-treated (n = 6) or untreated DPSCs (n = 6) was used to fill the dissected area. If no immediate transplantation was performed (n = 6), the incision was sutured closed. Mice were allowed to heal for 30 days.
2.4. Hematoxylin and Eosin Staining
The tibialis anterior muscles were excised from mice in the four groups and fixed in ice-cold 4% PFA. After dehydration in a graded alcohol series, the tissue was embedded in paraffin and cut into 5 µm-thick sections by using a Leica RM2255 rotary microtome (Leica Microsystems, Mannheim, Germany). After deparaffinization in xylene and hydration through a graded alcohol series to ddH2O, hematoxylin, and eosin (Servicebio, Wuhan, China), staining was performed according to the manufacturer’s protocols.
2.5. RNA Extraction and Real-Time Polymerase Chain Reaction (PCR)
Total RNA was extracted from the cells using TRIzol (Life Technologies, CA, USA). The RNA concentration was quantified using a spectrophotometer by measuring the OD260/280 ratio (1.80–1.95). DNA was removed by gDNase at 42°C for 3 min. cDNA was reverse transcribed using a FastQuant RT Kit (Tiangen, Beijing, China) at 42°C for 15 min and at 95°C for 3 min. Subsequently, SYBR Green Premix (Tiangen, Beijing, China) was used for the amplification of cDNA on a Light Cycler 96 system (Roche, Basel, Switzerland) with each primer at 0.6 mM and 150 ng of cDNA template. The thermocycling conditions were as follows: 95°C for 15 min, followed by 40 cycles at 95°C for 10 s and at 60°C for 32 s. Transcript levels were normalized using β-actin as a housekeeping gene and analyzed with the 2−∆∆Ct method. The primer sequences can be found in Table S2.
2.6. Western Blot Analysis
Total protein was extracted in RIPA lysis buffer (Thermo Fisher Scientific, MA, USA) plus protease/phosphatase inhibitor cocktail (#5872, CST, MA, USA). Lysate proteins (25 mg) were loaded onto SDS-polyacrylamide gels (7.5% or 10%) and subsequently transferred onto PVDF membranes. Target proteins on the membranes were incubated overnight at 4°C with primary antibodies diluted 1:1000–1:200 in 1 × TBS-Tween (TBS: 0.05 M Tris, 0.15 M NaCl [pH 7.5]; with 0.2% Tween-20). Antibodies against β-actin (abs830031; Absin Bioscience, Shanghai, China), Pax7 (ab187339), and Myf5 (ab125078) from Abcam; MyHC IIB (BF-F3) and MyHC IIA (SC-71) from Developmental Studies Hybridoma Bank (DSHB, ID, USA); p-Smad 1/5/9 (#13820) and Smad 1 (#6944) from Cell Signaling Technology (CST, MA, USA); and MyoD (#18943), Desmin (#60226), MRF4 (#11754), Pax3 (#21383), Six1 (#10709), Eya2 (#11314), ID1 (#18475), and MSX1 (#17678) from Proteintech (IL, USA) were used. After intensive washing, the membranes were incubated with HRP-conjugated secondary antibodies (#7076, anti-mouse IgG; #7074, antirabbit IgG, both from CST) for 1–2 h. The membranes were visualized using Super Signal West Dura substrate (Thermo Scientific, MA, USA), and the bands were detected with an AI600 imager (GE Healthcare, IL, USA).
2.7. Immunofluorescence Microscopy
Cells, myofibers, and sections were fixed with 4% paraformaldehyde and placed in PBS containing 0.25% Triton X-100 for 10 min. After the samples were blocked with 3% bovine serum albumin (BSA, Solarbio, Beijing, China) in PBS for 30 min at room temperature, they were then incubated with the primary antibody in PBS containing 2.5% BSA at 4°C overnight. In addition to the primary antibodies listed above, antibodies against Pax7 (PAX7, DSHB), CD34 (#14486, Proteintech), human Lamin A/C (MBS477941, MyBioSource), human nucleoli (hNu) (ab190710, Abcam), and Laminin (ab11575, Abcam) were used. After washing, the samples were incubated with anti-mouse Alexa-594 or Alexa-488, or antirabbit Alexa-594 or Alexa-488 (Jackson ImmunoResearch, PA, USA), and then with 4′,6-diamidino-2-phenylindole (DAPI). Images of the cells were captured using a microscope (Leica Microsystems, Mannheim, Germany) and analyzed with ImageJ.
2.8. Cell Cycle Analysis
The cells were harvested and fixed in 70% precooled ethanol overnight at 4°C. After washing with PBS the next day, the cells were incubated with a solution containing RNase A and 50 µg/mL of PI (C1052, Beyotime, Shanghai, China) for 30 min at 37°C. Then, the cell cycle analysis was examined using a Novocyte 2040R flow cytometer (ACEA Biosciences, Hangzhou, China) with a cell count of ~0.5 × 104 ~ 2.0 × 104 frequency of G1, S, and G2 phases using NovoExpress software version 1.4.1 (ACEA Novocyte). The results were graphed using GraphPad Prism software.
2.9. Statistical Analysis
We performed statistical analyses using GraphPad Prism 5.0. Data are expressed as the mean ± standard deviation. The protein quantities were analyzed using semi-quantitative analysis. Comparisons between groups were performed using the Mann‒Whitney U test or Kruskal‒Wallis H test with multiple comparisons. Statistical significance was defined as
3. Results
3.1. Noggin Promotes the Formation of Myotubes in DPSCs
We cultured and identified DPSCs (Figure S1) and found that 5-Aza can induce MyoD in DPSCs by increasing myogenic markers (Figure S2) but with low efficiency. To examine whether Noggin could affect the MyoD of DPSCs, we treated DPSCs with Noggin after 5-Aza induction (Figure 1a). First, we found that Noggin had no significant effect on the proliferation of DPSCs (Figure S3 a–c) but increased myotube formation in DPSCs (Figure 1b,c). A more pronounced myotube morphology was observed after treatment with the Noggin protein for 21 days, and the differential index of multinuclear myotubes increased to ~20% (Figure 1d). The protein expression levels of MyHC (Figure 1e), such as MyHCIIA (Figure 1f) and MyHCIIB (Figure 1g), were significantly increased compared to those in the control group. This finding implies that Noggin might facilitate the formation of myotubes in DPSCs.
3.2. Noggin Accelerates the Progression of MyoD in DPSCs
To evaluate the effect of Noggin treatment on the MyoD of DPSCs, we assessed the expression of myogenic genes such as MyoD, MRF4, and Desmin on Days 7, 14, and 21. Noggin increased the expression of MyoD and Desmin (Figure 2a,b) but had no effect on the mRNA expression of MRF4 on Day 7 (Figure 2c). Immunofluorescence staining for MyoD (Figure 2d) and Desmin (Figure 2e) revealed elevated levels of MyoD (Figure 2f) and an increased quantity of long, spindle-shaped myotube-like cells (Figure 2g) in the Noggin-treated groups. Moreover, the protein expression levels of Desmin (Figure 2j) and MRF4 (Figure 2k) significantly increased upon treatment with Noggin on Day 14 (Figure 2h). In our study, Noggin was shown to play an important regulatory role in accelerating the MyoD of DPSCs, which might be a novel factor in myogenesis.
[figure(s) omitted; refer to PDF]
3.3. Noggin Increases the Generation of Pax7+ Satellite-Like Cells in DPSCs
SCs are characterized by the expression of the paired box (Pax)3/7. Six1 and eya2 have been shown to activate SCs (Pax7+) and promote myoblast differentiation. Notably, we found that Noggin increased the protein levels of Pax7 in DPSCs cultured in a proliferation medium (Figure S3). To determine SCs generation through MyoD of DPSCs with Noggin, we measured the relative mRNA levels on Days 1, 3, and 7, as well as the protein expression levels of Six1, Pax3, Pax7, and Eya2 on Days 7, 14, and 21.
The mRNA expression of Pax7 increased in a concentration-dependent manner under Noggin treatment, reaching its peak on Day 3 and remaining higher than that of the control on Day 7 (Figure 3a). Consistent with the expression of Pax7, the mRNA expression of Pax3 increased on Day 3 and remained elevated on Day 7 (Figure 3b). Additionally, the mRNA expression of Six1 (Figure 3c) and Eya2 (Figure 3d) increased on Day 7. Western blotting (Figure 3e) and immunofluorescence staining (Figure 3f) for Pax7 revealed that Noggin upregulated the protein expression of Pax7 on Days 7, 14, and 21 (Figure 3g,h). The protein expression levels of Pax3 (Figure 3i), Six1 (Figure 3j), and Eya2 (Figure 3k) were also increased in the Noggin-treated groups on Day 14 and remained high on Day 21 (Figure 3e). These results suggest that Noggin promotes the myogenic process in DPSCs by upregulating the expression of Six1/Eya2 in addition to Pax3/Pax7.
[figure(s) omitted; refer to PDF]
3.4. Noggin Antagonizes BMP by Downregulating p-Smad 1/5/9, Thus Facilitating the MyoD of DPSCs
To determine whether Noggin regulates the MyoD of DPSCs by regulating Smad signaling, we first simulated 3D protein structures using SWISS-MODEL and visualized them with PyMOL software, which showed that Noggin competitively inhibits the binding of BMP4 to BMP-receptor I A (BMPRIA) (Figure 4a). Then, we blocked the effect of Noggin by adding the BMP protein. The protein levels of Pax7, Pax3, Eya2, and Desmin were observed (Figure 4b). Compared with that of the Noggin treatment group (200N), their expression decreased in the Noggin + BMP treatment group. Among them, Pax7 was most prominent when compared with that in the 5-Aza or control groups (Figure 4b).
[figure(s) omitted; refer to PDF]
To further explore the downstream regulation of BMP signaling by Noggin, we determined the phosphorylation levels of members of the BMP/p-Smad pathway (Figure 4c−e). The protein levels of p-Smad 1/5/9 increased following the activation of the BMP pathway by adding BMP4 (Figure 4c). Noggin persistently eliminated the phosphorylation levels even after subsequent stimulation with BMP4 (Figure 4c−e). Downstream effectors of the BMP/p-Smad pathway, such as inhibitors of DNA binding 1 (ID1) and msh homeobox 1 (MSX1), were also downregulated by Noggin treatment (Figure 4f,g). Therefore, our results indicate that Noggin facilitates the MyoD of DPSCs by inhibiting BMP/Smad activation.
3.5. Noggin-Pretreated DPSCs Combined With Matrigel Can Effectively Repair Muscle Injury in VML
To test the utility of DPSCs in repairing muscle injuries, we established a mouse VML injury model that exhibited a loss of ~40% of the tibialis anterior muscle (Figure 5a−c). In the untreated VML-injured group, the volumetric defect remained (Figure 5d). Shrinkage occurred due to the defect area. The muscle defect deformity exhibited the deposition of a thin layer of disorganized collagenous scar tissue (Figure 5d). The injury defect exceeded the threshold and could not be restored by the endogenous regenerative potential of the skeletal muscle. This result indicates the success of VML modeling.
[figure(s) omitted; refer to PDF]
The utilization of scaffolds can provide temporary mechanical support and the necessary growth environment for seed cell adhesion, growth, proliferation, and differentiation. Therefore, we used the thermosensitive hydrogel Matrigel to provide a scaffold for stem cell transplantation (Figure 5d). We observed that the Matrigel group exhibited a reduced shrinkage area and decreased muscle fibrosis (Figure 5d). In addition, we found some mononuclear cells infiltrating in the defect (Figure 5d).
To examine whether Noggin-pretreated DPSCs could better repair muscle injury than the controls, we transplanted DPSC bioconstructs into the defects of VML muscles (Figure 5a–c). We implanted untreated DPSCs (Figure 5d) or Noggin-pretreated DPSCs (Figure 5d) reconstituted in Matrigel into the defects. Morphometric analysis of muscle cross-sections revealed that the Noggin-treated DPSCs showed a 73% decrease in the size of the defect and a 69% decrease in scar tissue (Figure 5d–f). In contrast to the defects in the above two groups, which exhibited irreversible and robust fibrotic scars, the defects treated with DPSCs showed markedly reduced fibrotic tissue surrounded by cells of varying morphologies, such as fibrotic, inflammatory, and vessel-like cells, indicating the tissue repair process (Figure 5d). In contrast, Noggin-treated DPSCs might have accelerated this process and facilitated the improved formation of muscle tissue, leaving little cell infiltration (Figure 5d).
3.6. Noggin-Pretreated DPSCs Bioconstructs Can Benefit the Muscle SC Population and Promote Myogenic Repair
To explore the contribution of grafted cells to muscle injury, we immunostained muscle cross-sections for Pax7 (SC marker), MyoD (activated SCs/myoblasts), hNu, and human LaminA/C (specific antibody to trace human cells), and Laminin (to identify position within the sarcolemma). Pax7/MyoD costaining revealed that stem cell transplantation increased the proportion of activated SCs compared to the sham or Matrigel groups (Figure S4). We also observed that hNu was integrated into the nucleus of regenerated tissue and was located on the Laminin-stained muscle sarcolemma (Figure S4), a phenomenon more readily discernible in the Noggin-treated DPSCs groups than in the DPSCs groups. Increased numbers of hLaminAC+/Pax7+ (Figure 5g) and hNu+/MyoD+ (Figure 5g) cells were also found in the Noggin-treated DPSCs groups compared with the DPSCs groups, indicating an increase in donor-derived SCs (Figure 5h−k). These results suggest that Noggin-treated DPSCs partially benefited the SC population. This benefit might provide support for muscle regeneration in VML injury.
4. Discussion
In our study, we found that Noggin could promote the MyoD of DPSCs and increase the generation of satellite-like cells by regulating the BMP/Smad signaling pathway. Then, we treated VML model mice with Noggin-pretreated DPSCs bioconstructs and found that Noggin pretreatment improved the repair of muscle injury. Our work implies that Noggin can promote the MyoD of DPSCs, rendering DPSCs an important source of muscle stem cells for muscle repair.
Noggin, a secreted BMP antagonist, was promoted by Wnt-1 in medial somites and found to promote the expression of MyoD in embryonic tissues [21, 23]. Our results showed that Noggin could promote the MyoD of DPSCs (Figures 1 and 2) and increase the number of Pax3+/Pax7+ cells (Figure 3). Previous studies have concluded that Pax3-mediated myogenesis requires an environment where Six1 synergizes with Eya2 to activate the expression of MyoD [28]. Our results showed that Noggin promoted the expression of Six1 and Eya2, suggesting that these Pax3+/Pax7+ cells were in an environment conducive to subsequent MyoD. We also found that Noggin eliminated Smad phosphorylation, accompanied by decreased levels of MSX1 and ID1 (Figure 4). Consistent with our results, Noggin inhibited p-Smad1/5/8 and repressed ID1 and MSX1 expression, impeding myoblast differentiation [29–31]. Sustained release of BMP4 significantly decreased Pax7+ and MyoD+ SC densities [32]. When BMP4 was added to antagonize Noggin, we observed inhibition of the MyoD of DPSCs. Our data revealed that Pax3/7 might be related to the BMP/Smad pathway in the context of Noggin effects. Previous research has shown that activated Pax7+ cells coexpressed p-Smad1/5/8 in injured mouse skeletal muscle sections [31]. In the competition for MSX1, the expression of downstream effectors of the BMP/Smad pathway, Pax3 or Pax7, was increased towards the myogenic fate [33]. Knockout of Noggin leads to increased levels of p-Smad1/5/8, ID1, and MSX1, and a decreased population of mesenchymal Pax7+ muscle precursor cells [25].
Muscle regeneration is ensured by Pax7+ SCs; when activated, SCs are able to divide asymmetrically, enabling self-renewal or myoblast differentiation [1]. ESCs-derived Pax7+ cells can give rise to both muscle fibers and Pax7+ satellite-like cells when grafted in vivo, suggesting that these cells might behave like SCs [34]. In our study, we found that Noggin expanded Pax7+ cells in DPSCs. After 21 days of differentiation in vitro, the expression levels of late myogenic marker genes (MyHC) and SC marker (Pax7) were both higher with Noggin treatment (Figures 1 and 3). These results revealed that Noggin could promote the generation of Pax7+ satellite-like cells from DPSCs (Figure 6).
[figure(s) omitted; refer to PDF]
We then transplanted Noggin-pretreated DPSCs combined with Matrigel into defects of a VML injury model. In our study, Noggin-pretreated DPSCs bioconstructs decreased the size of the defect and increased the number of donor-derived hLaminAC+/Pax7+ and hNu+/MyoD+ cells in mouse muscles (Figure 5g). Some scholars transplanted ESC- or PSC-derived muscle progenitor cells into NSG-mdx4cv mice and found that donor-derived hLaminAC+/Pax7+ SCs contributed to the muscle stem cell pool in tissues [35]. The successful use of Pax7+ satellite-like cells derived from ESCs in muscle injury may be attributed to their self-renewal capacity within the SCs pool [36].
DPSCs are a type of MSCs from dental pulp, with easy access, long-term storage, and high proliferative and differentiation potential compared to other types of MSCs [9–12] and can serve as a proper stem cell source for muscle regeneration. SCs derived from MSCs were shown to give rise to differentiated muscle fibers [37]. However, the contribution of 5-Aza-induced DPSCs engraftment might only be a paracrine effect instead of MyoD [14]. In contrast to muscle-derived cells or ESCs, the transplant efficiency of MSCs was relatively low, with ~1%–2% as hybrid myofibers [38]. We speculate that the low myogenic efficiency may be attributed to fewer SCs. Our results indicated that Noggin expanded Pax7+ satellite-like cells in DPSCs, which increased the percentage of donor-derived cells to ~15%–20% (Figure 5i–k). This finding suggests that Noggin-treated DPSCs can partially benefit the muscle SC pool and promote myogenic repair in VML injury.
5. Conclusions
In this study, we demonstrate that Noggin, a secreted BMP antagonist, is an important protein that promotes MyoD and increases the generation of Pax7+ satellite-like cells by modulating BMP/Smad signaling. These satellite-like cells combined with Matrigel can effectively repair mouse tibialis anterior muscle injury in a VML model, leading to increased repair size and decreased scar tissue. Noggin-treated DPSCs bioconstructs can benefit the Pax7+ SC pool and promote muscle regeneration. This finding suggests the importance of producing Pax7+ SCs and highlights the potential of these SCs for improved muscle regeneration. Noggin might enhance the generation of satellite-like cells from DPSCs to provide potential transplantable stem cells for muscle regeneration. Further investigations are needed to study the relationship between Pax7 and Smad/its downstreams in order to improve myogenic induction conditions and enhance the transplantation of DPSCs into damaged muscle tissue.
Ethics Statement
Animal protocols were approved by Cyagen IACUC (No. ACU20-A034). The research proposal for the use of human samples was approved by the Medical Ethical Committee of Shanghai Stomatological Hospital, Fudan University.
Consent
We have also obtained informed consents to participate from the human donors.
Disclosure
A preprint has previously been published (Zhang, MH et al. 2021) [39].
Author Contributions
Yue-Hua Liu and Xin-Xin Han conceived and designed the experiments. Meng-Han Zhang, Li-Ming Yu, Wei-Hua Zhang, and Jia-Jia Deng performed the experiments. Li-Ming Yu, Bing-Jing Sun, Mei-Hua Chen, Wei Huang, and Hua He analyzed data and interpreted the results. Meng-Han Zhang and Wei-Hua Zhang assembled the figures. Yue-Hua Liu and Xin-Xin Han provided financial support. Meng-Han Zhang and Li-Ming Yu wrote and revised the manuscript. Yue-Hua Liu and Xin-Xin Han reviewed and edited the manuscript. Wei-Hua Zhang and Jiao Li assisted with the literature searches and revised the manuscript. All authors read and approved the final manuscript. Meng-Han Zhang and Li-Ming Yu contributed equally to this work.
[1] H. Yin, F. Price, M. A. Rudnicki, "Satellite Cells and the Muscle Stem Cell Niche," Physiological Reviews, vol. 93 no. 1, pp. 23-67, 2013.
[2] R. R. Kalyani, M. Corriere, L. Ferrucci, "Age-Related and Disease-Related Muscle Loss: The Effect of Diabetes, Obesity, and Other Diseases," The Lancet Diabetes & Endocrinology, vol. 2 no. 10, pp. 819-829, DOI: 10.1016/S2213-8587(14)70034-8, 2014.
[3] J. Dong, X. Niu, X. Chen, "“Injury and Apoptosis in the Palatopharyngeal Muscle in Patients With Obstructive Sleep Apnea-Hypopnea Syndrome," Medical Science Monitor, vol. 26, 2020.
[4] B. E. Pollot, B. T. Corona, "Volumetric Muscle Loss," Skeletal Muscle Regeneration in the Mouse, vol. 1460, pp. 19-31, 2016.
[5] K. Garg, C. L. Ward, B. J. Hurtgen, "“Volumetric Muscle Loss: Persistent Functional Deficits Beyond Frank Loss of Tissue," Journal of Orthopaedic Research, vol. 33 no. 1, pp. 40-46, 2015.
[6] C. Sun, C. Serra, G. Lee, K. R. Wagner, "“Stem Cell-Based Therapies for Duchenne Muscular Dystrophy," Experimental Neurology, vol. 323, 2020.
[7] M. La Noce, A. Stellavato, V. Vassallo, "Hyaluronan-Based Gel Promotes Human Dental Pulp Stem Cells Bone Differentiation by Activating YAP/TAZ Pathway," Cells, vol. 10 no. 11, 2021.
[8] P. Naddeo, L. Laino, M. La Noce, "Surface Biocompatibility of Differently Textured Titanium Implants With Mesenchymal Stem," Dental Materials, vol. 31 no. 3, pp. 235-243, DOI: 10.1016/j.dental.2014.12.015, 2015.
[9] V. Mattei, S. Martellucci, F. Pulcini, F. Santilli, M. Sorice, S. Delle Monache, "Regenerative Potential of DPSCs and Revascularization: Direct, Paracrine or Autocrine Effect?," Stem Cell Reviews and Reports, vol. 17 no. 5, pp. 1635-1646, DOI: 10.1007/s12015-021-10162-6, 2021.
[10] V. Tirino, F. Paino, R. d’Aquino, V. Desiderio, A. De Rosa, G. Papaccio, "Methods for the Identification, Characterization and Banking of Human DPSCs: Current Strategies and Perspectives," Stem Cell Reviews and Reports, vol. 7 no. 3, pp. 608-615, DOI: 10.1007/s12015-011-9235-9, 2011.
[11] W. Zhang, X. F. Walboomers, S. Shi, M. Fan, J. A. Jansen, "“Multilineage Differentiation Potential of Stem Cells Derived From Human Dental Pulp After Cryopreservation," Tissue Engineering, vol. 12 no. 10, pp. 2813-2823, 2006.
[12] A. La Gatta, V. Tirino, M. Cammarota, "Gelatin-Biofermentative Unsulfated Glycosaminoglycans Semi-Interpenetrating Hydrogels via Microbial-Transglutaminase Crosslinking Enhance Osteogenic Potential of Dental Pulp Stem Cells," Regenerative Biomaterials, vol. 8 no. 3,DOI: 10.1093/rb/rbaa052, 2021.
[13] I. Kerkis, C. E. Ambrosio, A. Kerkis, "“Early Transplantation of Human Immature Dental Pulp Stem Cells From Baby Teeth to Golden Retriever Muscular Dystrophy (GRMD) Dogs: Local or Systemic?," Journal of Translational Medicine, vol. 6, 2008.
[14] A. Pisciotta, M. Riccio, G. Carnevale, "Stem Cells Isolated From Human Dental Pulp and Amniotic Fluid Improve Skeletal Muscle Histopathology in mdx/SCID Mice," Stem Cell Research & Therapy, vol. 6 no. 1,DOI: 10.1186/s13287-015-0141-y, 2015.
[15] D. Yu, Z. Cai, D. Li, "Myogenic Differentiation of Stem Cells for Skeletal Muscle Regeneration," Stem Cells International, vol. 2021, 2021.
[16] W. Roman, E. R. Gomes, "Nuclear Positioning in Skeletal Muscle," Seminars in Cell & Developmental Biology, vol. 82, pp. 51-56, 2018.
[17] L. L. Muzio, M. Mascitti, M. La Noce, F. Posa, Y. Kudo, N. Cirillo, "Molecules and Biomaterial Technologies Affecting Stem Cell Differentiation," Stem Cells International, vol. 2022, 2022.
[18] H. Xi, W. Fujiwara, K. Gonzalez, "“In Vivo Human Somitogenesis Guides Somite Development From hPSCs," Cell Reports, vol. 18 no. 6, pp. 1573-1585, 2017.
[19] J. Chal, A. L. T. Al, M. Oginuma, "Recapitulating Early Development of Mouse Musculoskeletal Precursors of the Paraxial Mesoderm in Vitro," Development, vol. 145 no. 6, 2018.
[20] C. Marcelle, M. R. Stark, M. Bronner-Fraser, "Coordinate Actions of BMPs, Wnts, Shh and Noggin Mediate Patterning of the Dorsal Somite," Development, vol. 124 no. 20, pp. 3955-3963, 1997.
[21] E. Hirsinger, D. Duprez, C. Jouve, P. Malapert, J. Cooke, O. Pourquie, "Noggin Acts Downstream of Wnt and Sonic Hedgehog to Antagonize BMP4 in Avian Somite Patterning," Development, vol. 124 no. 22, pp. 4605-4614, 1997.
[22] D. P. Brazil, R. H. Church, S. Surae, C. Godson, F. Martin, "BMP Signalling: Agony and Antagony in the Family," Trends in Cell Biology, vol. 25 no. 5, pp. 249-264, 2015.
[23] A. Stantzou, E. Schirwis, S. Swist, "BMP Signaling Regulates Satellite Cell-Dependent Postnatal Muscle Growth," Development, vol. 144 no. 15, pp. 2737-2747, 2017.
[24] C. Linker, C. Lesbros, M. R. Stark, C. Marcelle, "Intrinsic Signals Regulate the Initial Steps of Myogenesis in Vertebrates," Development, vol. 130 no. 20, pp. 4797-4807, 2003.
[25] D. Costamagna, H. Mommaerts, M. Sampaolesi, P. Tylzanowski, "Noggin Inactivation Affects the Number and Differentiation Potential of Muscle Progenitor Cells in Vivo," Scientific Reports, vol. 6, 2016.
[26] W. Zhang, L. Yu, X. Han, "The Secretome of Human Dental Pulp Stem Cells Protects Myoblasts From Hypoxia-Induced Injury via the Wnt/Beta-Catenin Pathway," International Journal of Molecular Medicine, vol. 45 no. 5, pp. 1501-1513, 2020.
[27] R. Nakatsuka, T. Nozaki, Y. Uemura, "5-Aza-2′-Deoxycytidine Treatment Induces Skeletal Myogenic Differentiation of Mouse Dental Pulp Stem Cells," Archives of Oral Biology, vol. 55 no. 5, pp. 350-357, 2020.
[28] T. A. Heanue, R. Reshef, R. J. Davis, "Synergistic Regulation of Vertebrate Muscle Development by Dach2, Eya2, and Six1, Homologs of Genes Required for Drosophila Eye Formation," Genes & Development, vol. 13 no. 24, pp. 3231-3243, DOI: 10.1101/gad.13.24.3231, 1999.
[29] X. Leng, X. Ji, Y. Hou, R. Settlage, H. Jiang, "Roles of the Proteasome and Inhibitor of DNA Binding 1 Protein in Myoblast Differentiation," FASEB Journal, vol. 33 no. 6, pp. 7403-7416, 2019.
[30] X. Zhu, M. Li, X. Jia, "The Homeoprotein Msx1 Cooperates With Pkn1 to Prevent Terminal Differentiation in Myogenic Precursor Cells," Biochimie, vol. 162, pp. 55-65, 2019.
[31] J. L. Clever, Y. Sakai, R. A. Wang, D. B. Schneider, "Inefficient Skeletal Muscle Repair in Inhibitor of Differentiation Knockout Mice Suggests a Crucial Role for BMP Signaling During Adult Muscle Regeneration," American Journal of Physiology-Cell Physiology, vol. 298 no. 5, pp. C1087-C1099, DOI: 10.1152/ajpcell.00388.2009, 2010.
[32] B. C. Anderson, M. L. Daniel, J. D. Kendall, S. P. Christiansen, L. K. McLoon, "Sustained Release of Bone Morphogenetic Protein-4 in Adult Rabbit Extraocular Muscle Results in Decreased Force and Muscle Size: Potential for Strabismus Treatment," Investigative Opthalmology & Visual Science, vol. 52 no. 7, pp. 4021-4029, DOI: 10.1167/iovs.10-6878, 2011.
[33] P. Daubas, N. Duval, L. Bajard, "Fine-Tuning the Onset of Myogenesis by Homeobox Proteins that Interact with the Myf5 Limb enhancer," Biology Open, vol. 4 no. 12, pp. 1614-1624, 2015.
[34] J. Chal, M. Oginuma, Z. Al Tanoury, "Differentiation of Pluripotent Stem Cells to Muscle Fiber to Model Duchenne Muscular Dystrophy," Nature Biotechnology, vol. 33 no. 9, pp. 962-969, 2015.
[35] R. Darabi, R. Arpke, S. Irion, "Human ES- and iPS-Derived Myogenic Progenitors Restore Dystrophin and Improve Contractility Upon Transplantation in Dystrophic mice," Stem Cell, vol. 10, pp. 610-619, 2012.
[36] H. Chang, M. Yoshimoto, K. Umeda, "Generation of Transplantable, Functional Satellite-Like Cells From Mouse Embryonic Stem," FASEB Journal, vol. 23 no. 6, pp. 1907-1919, DOI: 10.1096/fj.08-123661, 2009.
[37] M. A. LaBarge, H. M. Blau, "Biological Progression From Adult Bone Marrow to Mononucleate Muscle Stem Cell to Multinucleate Muscle Fiber in Response to Injury," Cell, vol. 111 no. 4, pp. 589-601, 2002.
[38] F. Chretien, P. A. Dreyfus, C. Christov, "In Vivo Fusion of Circulating Fluorescent Cells With Dystrophin-Deficient Myofibers Results in Extensive Sarcoplasmic Fluorescence Expression but Limited Dystrophin Sarcolemmal Expression," The American Journal of Pathology, vol. 166 no. 6, pp. 1741-1748, DOI: 10.1016/S0002-9440(10)62484-4, 2005.
[39] M. H. Zhang, L. M. Yu, W. H. Zhang, "“Noggin Combined With Dental Pulp Stem Cells Repair Muscle Injury Through Smad/Pax7 Signaling Pathway," Research Square, 2021.
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer
Copyright © 2024 Meng-Han Zhang et al. This is an open access article distributed under the Creative Commons Attribution License (the “License”), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Notwithstanding the ProQuest Terms and Conditions, you may use this content in accordance with the terms of the License. https://creativecommons.org/licenses/by/4.0/
Abstract
A proper source of stem cells is key to muscle injury repair. Dental pulp stem cells (DPSCs) are an ideal source for the treatment of muscle injuries due to their high proliferative and differentiation capacities. However, the current myogenic induction efficiency of human DPSCs hinders their use in muscle regeneration due to the unknown induction mechanism. In this study, we treated human DPSCs with Noggin, a secreted antagonist of bone morphogenetic protein (BMP), and discovered that Noggin can effectively promote myotube formation. We also found that Noggin can accelerate the skeletal myogenic differentiation (MyoD) of DPSCs and promote the generation of Pax7+ satellite-like cells. Noggin increased the expression of myogenic markers and the transcriptional and translational abundance of satellite cell (SC) markers in DPSCs. Moreover, BMP4 inhibited Pax7 expression and activated p-Smad1/5/9, while Noggin eliminated BMP4-induced p-Smad1/5/9 in DPSCs. This finding suggests that Noggin antagonizes BMP by downregulating p-Smad and facilitates the MyoD of DPSCs. Then, we implanted Noggin-pretreated DPSCs combined with Matrigel into the mouse tibialis anterior muscle with volumetric muscle loss (VML) and observed a 73% reduction in the size of the defect and a 69% decrease in scar tissue. Noggin-treated DPSCs can benefit the Pax7+ SC pool and promote muscle regeneration. This work reveals that Noggin can enhance the production of satellite-like cells from the MyoD of DPSCs by regulating BMP/Smad signaling, and these satellite-like cell bioconstructs might possess a relatively fast capacity for muscle regeneration.
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer
Details











1 Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases Shanghai Stomatological Hospital and School of Stomatology Fudan University Shanghai China; School of Stomatology Affiliated to Medical College Zhejiang University Hangzhou China
2 Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases Shanghai Stomatological Hospital and School of Stomatology Fudan University Shanghai China
3 Department of Neurosurgery Third Affiliated Hospital of Second Military Medical University Shanghai China