We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine
Research ArticleOpen Accesscc iconby iconnc iconnd icon

Porous Se@SiO2 nanoparticles improve oxidative injury to promote muscle regeneration via modulating mitochondria

    Yu-Xia Yang‡

    Dalian Medical University, Dalian, 116044, People's Republic of China

    Sports Medicine Department, Northern Jiangsu People's Hospital, Clinical Medical College, Yangzhou University, Yangzhou, 225001, People's Republic of China

    ‡These authors contributed equally to this work

    Search for more papers by this author

    ,
    Ming-Sheng Liu‡

    Sports Medicine Department, Northern Jiangsu People's Hospital, Clinical Medical College, Yangzhou University, Yangzhou, 225001, People's Republic of China

    ‡These authors contributed equally to this work

    Search for more papers by this author

    ,
    Xi-Jian Liu

    School of Chemistry & Chemical Engineering, Shanghai University of Engineering Science, Shanghai, 201620, People's Republic of China

    ,
    Yu-Cheng Zhang

    Dalian Medical University, Dalian, 116044, People's Republic of China

    Sports Medicine Department, Northern Jiangsu People's Hospital, Clinical Medical College, Yangzhou University, Yangzhou, 225001, People's Republic of China

    ,
    Yang-Yang Hu

    Sports Medicine Department, Northern Jiangsu People's Hospital, Clinical Medical College, Yangzhou University, Yangzhou, 225001, People's Republic of China

    ,
    Rang-Shan Gao

    Dalian Medical University, Dalian, 116044, People's Republic of China

    Sports Medicine Department, Northern Jiangsu People's Hospital, Clinical Medical College, Yangzhou University, Yangzhou, 225001, People's Republic of China

    ,
    Er-Kai Pang

    Dalian Medical University, Dalian, 116044, People's Republic of China

    Sports Medicine Department, Northern Jiangsu People's Hospital, Clinical Medical College, Yangzhou University, Yangzhou, 225001, People's Republic of China

    ,
    Lei Hou

    Dalian Medical University, Dalian, 116044, People's Republic of China

    Sports Medicine Department, Northern Jiangsu People's Hospital, Clinical Medical College, Yangzhou University, Yangzhou, 225001, People's Republic of China

    ,
    Jing-Cheng Wang

    Sports Medicine Department, Northern Jiangsu People's Hospital, Clinical Medical College, Yangzhou University, Yangzhou, 225001, People's Republic of China

    &
    Wen-Yong Fei

    *Author for correspondence: Tel.: +86 180 5106 1779;

    E-mail Address: sbydyx105@163.com

    Sports Medicine Department, Northern Jiangsu People's Hospital, Clinical Medical College, Yangzhou University, Yangzhou, 225001, People's Republic of China

    Published Online:https://doi.org/10.2217/nnm-2022-0173

    Abstract

    Background: Acute skeletal muscle injuries are common among physical or sports traumas. The excessive oxidative stress at the site of injury impairs muscle regeneration. The authors have recently developed porous Se@SiO2 nanoparticles (NPs) with antioxidant properties. Methods: The protective effects were evaluated by cell proliferation, myogenic differentiation and mitochondrial activity. Then, the therapeutic effect was investigated in a cardiotoxin-induced muscle injury rat model. Results: Porous Se@SiO2 NPs significantly protected the morphological and functional stability of mitochondria, thus protecting satellite cells from H2O2-induced damage to cell proliferation and myogenic differentiation. In the rat model, intervention with porous Se@SiO2 NPs promoted muscle regeneration. Conclusion: This study reveals the application potential of porous Se@SiO2 NPs in skeletal muscle diseases related to mitochondrial dysfunction.

    Plain language summary

    Muscle injuries are very common in daily life and in sports. When a muscle is injured, the local response inhibits the regeneration and differentiation of stem cells inside the muscle, thus hindering muscle regeneration. The authors have recently developed a nanoparticle with the ability to protect muscle stem cell function, promote stem cell proliferation and differentiation and facilitate muscle regeneration after skeletal muscle injury in rats. Thus, this study reveals the potential of porous Se@SiO2 nanoparticles in skeletal muscle diseases associated with mitochondrial dysfunction.

    Acute skeletal muscle injuries are common among physical or sports traumas and are one of the most significant causes of disability [1]. Besides being prone to recurrence, this type of injury can also include pain, swelling and possibly osteofascial compartment syndrome [2]. Apart from mechanical damage to muscle cells, skeletal muscle injury also involves capillary rupture, hemorrhage, muscle tissue edema and inflammation, resulting in cytokines and inflammatory cells allowing direct access at the site of injury, induced mitochondrial dysfunction manifested as reactive oxygen species (ROS) overload, inhibition of mitochondrial respiration and disturbances in calcium homeostasis [3].

    Despite advances in sports medicine, effective treatments for skeletal muscle injury are still lacking. Surgical intervention, ice and rehabilitation exercises are the most common treatments for skeletal muscle injury, but the outcomes are unsatisfactory, and they prolong the functional recovery of the muscle, sometimes even predisposing it to repeated injury [4]. Though the main aim of the pharmacological intervention is to suppress the activation of inflammatory pathways, related reports have shown that excessive inhibition of inflammatory signaling pathways would affect the immune system and ultimately impair muscle regeneration [5]. Inefficient muscle regeneration often leads to poor contractility and progressive loss of muscle strength, resulting in loss of function [6]. Therefore, there is a great need to develop a new therapeutic strategy for musculoskeletal injuries.

    Satellite cells (SCs) are involved in repairing skeletal muscles after an injury [7]. They normally remain in a quiescent state and are characterized by the expression of PAX7 [8,9]. SCs initiate proliferation in response to specific stimuli such as direct trauma or extensive physical activity, and then they differentiate into new myofibers under the influence of specific factors, including MYOD and myogenin. They regulate the transcription of essential muscle-specific proteins such as MYHC [10]. However, in some cases, the injury is more severe than the compensatory ability of the skeletal muscle, and the number or function of SCs is restricted as a result of oxidative damage, which can lead to incomplete muscle regeneration and excessive fibrosis. Therefore, modulation of SCs at the site of injury has considerable therapeutic potential for promoting skeletal muscle regeneration.

    Mitochondria are essential organelles found in the cytoplasm of almost all eukaryotic cells. They are double-membrane-structure organelles with independent, self-replicating genomes and regulated cell metabolism. The oxidation/reduction O2-consuming metabolic pathways in mitochondria ensure survival at the cellular and organismal levels. In addition to their primary function, mitochondria also perform other essential functions, including biomolecule synthesis, ROS production, regulation of apoptosis and calcium homeostasis [11,12]. Mitochondrial dysfunction and oxidative stress play an important role in various metabolic disorders, including diabetes, obesity and hypertension [13]. As the largest metabolically active and most highly structured organelle, mitochondrial functions play vital roles in skeletal muscles. In addition to regulating the energy requirements of the cell, mitochondria are involved in many pathological processes [14].

    Overproduction of ROS is known to be caused in part by mitochondrial dysfunction. Although mitochondria are the major site of ROS production, the antioxidant enzyme system in mitochondria can scavenge excessive ROS [15]. When mitochondrial function is impaired, severe oxidative stress conditions limit the proliferation and differentiation of SCs, and the process of muscle regeneration is hindered. Because the application of antioxidants protects cells from death, it is evident that excessive oxidative stress results in cell death. The levels of antioxidants in SCs are a major factor in the outcome of cell-based therapies [16]. In addition, SCs regulate the regeneration of skeletal muscles by regulating the structure, metabolism and activity of mitochondria [12,17,18]. RNA sequencing of SCs activated after injury has revealed significant upregulation of mitochondrial genes, suggesting active mitochondrial metabolism [19]. However, few studies have examined the effects of antioxidant treatment on the mitochondria of SCs that are under oxidative stress, or even on the mechanisms of ROS in SCs [20]. Therefore, reducing oxidative damage to SCs by modulating mitochondrial activity may be a promising strategy for further promoting muscle regeneration after injury.

    Selenium (Se) is an essential micronutrient with a wide range of biological functions that can inhibit oxidative stress and scavenge excessive ROS [21]. However, the application of Se as a complementary therapy is narrow, and toxic effects due to excessive aggregation during Se administration in conventional or bulk form limit its application [22,23]. The development of nanotechnology appears to be a feasible solution. The authors designed porous Se@SiO2 nanoparticles (NPs), since they would slowly release Se from porous Se@SiO2 NPs and act as an ideal delivery system [24,25]. Porous Se@SiO2 NPs also have been shown to alleviate many diseases, including improving mitochondrial dysfunction in acute lung injury and alleviating oxidative damage. They are also effective in treating various diseases associated with oxidative damage or mitochondrial dysfunction [26].

    The authors of this study hypothesized that porous Se@SiO2 NPs could promote muscle regeneration and protect the myogenic differentiation of SCs. They created an oxidative stress environment using H2O2 in an in vitro cell culture. To induce myofiber necrosis and regeneration, they injected cardiotoxin (CTX) into the tibialis anterior (TA) muscle of rats to construct a model of acute injury. The potential mechanisms underlying the effect of porous Se@SiO2 NPs were investigated. This study provides preliminary evidence that porous Se@SiO2 NPs promote the proliferation and myogenic differentiation of SCs by scavenging mitochondrial ROS and improving mitochondrial activity under oxidative stress, ultimately promoting skeletal muscle regeneration in rats.

    Material & methods

    Preparation & characterization of porous Se@SiO2 NPs

    The porous Se@SiO2 NPs were synthesized following the procedure used in the authors' previous research [27]. A mixture of 2 ml of Cu2–x Se nanocrystals synthesized in nitrogen was mixed thoroughly with 20 ml of n-hexane, 20 ml of n-hexanol, 2 ml of Triton X-100, 0.6 ml of deionized water and 0.08 ml of ethyl orthosilicate. Subsequently, 0.1 ml of ammonia was added and orthosilicate was hydrolyzed to encapsulate the silica in the Se quantum dots prepared by oxidation to form solid Se@SiO2 nanospheres. Finally, they were dispersed in 10 mg/ml of polyvinylpyrrolidone solution and etched in hot water to form porous structures. The porous Se@SiO2 NPs were characterized by transmission electron microscopy (HT7700, HITACHI, Tokyo, Japan) and D/max2550 x-ray diffractometer (Cu K-α radiation; Rigaku, Tokyo, Japan).

    Isolation & culture of SCs

    All experiments were approved by the Animal Ethics Committee of Yangzhou University and conducted in accordance with the regulation and guidance. SCs were prepared as described previously [28]. Newborn male Sprague-Dawley suckling rats were euthanized, and the TA and gastrocnemius muscles were dissected. The muscle tissue was homogenized and enzymatically digested by adding type II collagenase. The enzymatically cleaved tissue homogenate was then centrifuged at 400–500 r.p.m. for 3 min. The supernatant was removed, and the pellet was resuspended in Collagenase/Dispase (Roche, Basel, Switzerland) and incubated in a water bath on a rocker. The homogenate was passed through a series of mesh cell strainers. The filtrate was centrifuged in a tabletop centrifuge at 1200 r.p.m. for 10 min. The pellet was resuspended gently in a growth medium containing DMEM (HyClone, UT, USA) supplemented with 20% fetal bovine serum (HyClone), 3% chicken embryo extract, 4 mM L-glutamine, 10 mM sodium pyruvate and 1% penicillin–streptomycin (Sangon Biotech, Shanghai, China). The solution was poured into Petri dishes and preplated in an incubator (5% CO2; 37°C) to remove fibroblasts. At about 3–5 days, the SC culture was shifted from growth medium to differentiation medium (DMEM supplemented with 5% horse serum, 1% penicillin/streptomycin, 4 mM L-glutamine and 10 mM sodium pyruvate). SCs from passages 3–4 were used for later experiments. The cells proliferated faster in about the first 3 days, then gradually started to differentiate, and differentiation was more obvious around day 7. Subsequent assays related to cell proliferation, death and senescence were selected on day 3, and assays related to myogenic differentiation and mitochondria were performed on day 7.

    Establishment of H2O2-induced damage models of SCs

    Based on the presence or absence of 20 μg/ml porous Se@SiO2 NPs and 100 μM H2O2, cells were divided into four groups: control group, in which cells were cultured in differentiation medium only; Se@SiO2 NPs group, in which cells were treated with differentiation medium and porous Se@SiO2 NPs; H2O2 group, in which cells were treated with differentiation medium and then with 100 μM H2O2 for 24 h; Se@SiO2 + H2O2 group, in which cells were treated with differentiation medium and porous Se@SiO2 NPs and then with 100 μM H2O2 for 24 h. To reduce the impact of H2O2 evaporation on the experiment, the authors increased the number of liquid changes in the medium within 24 h of adding H2O2. A day later, H2O2 solution was removed, and the cells were washed with phosphate-buffered saline (PBS) solution. All groups were cultured for 7 days.

    In vitro safety of porous Se@SiO2 NPs & cell proliferation assay

    First, the SCs were cultured with different concentration gradients of porous Se@SiO2 NPs (0–160 μg/ml), and cell samples were harvested after 1 day. In addition, the four groups of cell samples were harvested on days 1, 2 and 3. The effects of porous Se@SiO2 NPs and H2O2 on cell proliferation were assessed by MTT assay. Briefly, culture medium containing dimethyl sulfoxide (Invitrogen, MA, USA) served as control, and MTT solution (Sigma-Aldrich, MO, USA) was added to each well. Then the mixed liquid was sucked out, 150 μl/well of dimethyl sulfoxide was added and it was shaken for 10 min. The absorbance was measured by a microplate reader (MK3 type, Multiskan GO; Thermo Fisher Scientific, MA, USA) at 492 nm.

    Staining of live–dead cells & senescence-associated β-galactosidase (SA-β-gal) staining

    Cell samples were obtained after 3 days of induction. The diluted calcein AM/PI kit (Biovision, CA, USA) was added according to the instructions and incubated for 30 min, followed by washing. It was observed using a fluorescence microscope (Olympus IX71, Olympus, Tokyo, Japan). Red and green denoted dead cells and live cells. The ratio of living cells to dead cells was measured by ImageJ software, version 1.39.

    The presence of senescent cells was also evaluated through the activity of senescence-associated β-galactosidase (SA-β-gal). Cells collected at the same time were washed with PBS. SA-β-gal staining fixation solution (Beyotime, Shanghai, China) was added, followed by fixing at room temperature. The solution was then removed and the cells were washed with PBS. PBS was removed and dye solution was added per well according to the instructions, followed by overnight incubation at 37°C. The observations were recorded using an optical microscope.

    Detection of mitochondrial ROS

    On days 3 and 7, cell samples were taken. Dimethyl sulfoxide was added to MitoSoX Red Mitochondrial Superoxide Indicator (Yeasen, Shanghai, China) and mixed well to prepare storage solution and was diluted with Hanks' balanced salt solution; 1–2 ml probe working solution was added and the cells were fully covered, followed by incubation at 37°C in the dark for 10 min. Cells were then counterstained with Hoechst (Beyotime) for 5 min, and after sealing it was observed by confocal laser scanning microscopy (TCS SP5, Leica, Heidelberg, Germany).

    Measurement of mitochondrial morphology

    The mitochondrial morphology and actin filaments of SCs were observed under a confocal microscope on day 7. First, SCs in each group, as mentioned earlier, were plated on confocal Petri dishes. Then they were incubated with the MitoTracker Red CMXRos (Beyotime) in the ratio of 1:1000–1:10,000 and F-actin dye solution (Abcam, MA, USA) at 37°C for 30 min. The intracellular fluorescence was observed under a confocal fluorescence microscope (Leica). MitoTracker Red fluorescence staining of mitochondria in live cells was determined by ImageJ software.

    Evaluation of mitochondrial respiratory chain & catalase activities

    On day 7, the activities of the mitochondrial respiratory chain (MRC) were measured by the MRC Complex Activity Assay Kit (Solarbio, Peking, China) and the ATP Synthase Activity Assay Kit (YBio; Shanghai, China) according to the instructions. MRC activities were determined using a colorimetric assay at 340 nm (MRC complex I), 550 nm (MRC complex III) and 450 nm (ATP synthase). The unit enzyme activity was exhibited as nmol/min/mg protein. At the same time, the CAT Activity Detection Kit (Solarbio) was used to measure the activity of catalase (CAT). According to the instructions, the enzyme labeling instrument (ThermoMK3; Thermo Fisher Scientific) section wavelength was set to 405 nm for reading and calculating the absorbance value. The unit enzyme activity was exhibited as μmol/min/mg protein.

    Rat model of skeletal muscle injury

    All experimental procedures with rats were approved by Animal Ethics Committee of Yangzhou University. Common methods were adopted, for which 24 male, 8-week-old rats were placed in a supine position; the left limb was rinsed with 95% ethanol. Approximately 30 μl of saline solution dissolved with CTX (20 μM; WeiGa, Guangzhou, China) was injected into the middle belly of the left TA muscle. After the injection, analgesia and intramuscular injection of penicillin were given to prevent infection. Then the rats were randomly divided into four groups. In the CTX + Se@SiO2 group, porous Se@SiO2 NPs (1 mg/kg) were injected intramuscularly in the same place 2 h after CTX injection and porous Se@SiO2 NPs (1 mg/kg) were injected intraperitoneally per day; meanwhile, the right leg of the CTX + Se@SiO2 group of rats was labeled as the Se@SiO2 group. In the CTX group, normal saline was used in intramuscular injections 2 h after CTX injection and daily intraperitoneal injection of normal saline; at the same time, the right leg of the CTX group of rats was labeled as the control group. The concentration of porous Se@SiO2 NPs was chosen to be 1 mg/kg, according to the authors' previous studies; there was no significant toxic effect on the vital organs of the animals, and no significant aggregation was found in vivo [29]. According to previous studies, morphological changes occurred immediately after CTX injection, and over time the localization went from an inflammatory response to muscle regeneration, with maturation of regenerated muscle fibers in about 1 month [30,31]. The animals were sacrificed on days 1, 7, 14 and 28 after CTX injection. After euthanasia, TA muscles were dissected and snap-frozen in liquid nitrogen or preserved in formalin. Follow-up experiments were conducted by three different experimenters blinded to the study.

    Histological evaluation of muscle regeneration & fibrosis development

    The TA muscle samples preserved in formalin were then dehydrated, embedded in paraffin and cut into 4 μm sections. Subsequently, the sections were deparaffinized, rehydrated and subjected to histological staining. Hematoxylin and eosin staining was performed to evaluate injury and regeneration of myofibers; the Masson Modified Trichrome Staining Kit (Solarbio) was used to evaluate the deposition of collagen according to standard protocols.

    Western blot analysis

    For cell samples, whole-cell extracts in all four groups were prepared on day 7; for animal samples, rat TA muscle was collected on days 14 and 28. The samples were washed with PBS, followed by lysis in precooled RIPA lysate (P0013B, Beyotime), and then the protein was quantified by a BCA protein assay kit (KeyGEN, Shanghai, China). The protein samples were separated on a sodium dodecyl sulfate-polyacrylamide gel and then transferred to a polyvinylidene difluoride membrane. After blocking, the membrane was incubated with primary antibodies: SIRT1 (Proteintech, Wuhan, China), SOD1 (Proteintech), MYHC (Thermo Fisher Scientific), MYOD (Proteintech) and GAPDH (Proteintech). The polyvinylidene difluoride membrane was then treated with rabbit secondary antibody or rat secondary antibody (ZSGB-BIO, Peking, China). By employing enhanced chemiluminescence solution (Dingguo, Peking, China), the proteins were visualized, and the immunoreaction bands were quantified by ImageJ software. Protein expression was represented as the densitometric ratio of protein normalized to GAPDH.

    Immunofluorescence staining

    With immunofluorescence staining, the expression of markers was detected using antibodies against desmin (Proteintech), PAX7 (Proteintech), MYHC (Thermo Fisher Scientific), MYOD (Invitrogen), F-actin (Abcam) and dystrophin (Santa Cruz Biotechnology, TX, USA). Briefly, cell samples were washed thrice with PBS and then fixed with 4% paraformaldehyde for 15 min. After washing with PBS, the cells were permeabilized with 0.25% Triton X-100 (Rich Joint, Shanghai, China) for 10 min and incubated in 5% fetal bovine serum at 37°C for 1 h. Afterward, cells were incubated with primary antibodies overnight. After washing, secondary antibody was used to incubate the cells for 1 h at room temperature. The cells were then washed and treated with Hoechst (Beyotime) to stain the cell nuclei. In the end, samples were imaged under a fluorescence microscope (Olympus). Using ImageJ software according to the method described by Menconi et al. [32,33], quantitative image analysis was performed by analyzing five randomly selected injury fields per sample; fusion index was calculated as the number of nuclei in myotubes divided by the total number of nuclei counted. The average number of nuclei per myotube was determined by dividing the number of nuclei in myotubes by the total number of myotubes.

    Statistical analysis

    Qualitative data were representative of at least three independent experiments. Quantitative or semiquantitative data are expressed as the mean ± standard deviation. The data were analyzed using GraphPad Prism 6. Data were managed through one-way analysis of variance and Student's unpaired t-test. A difference was defined as statistically significant only when p < 0.05.

    Results

    Characterization of porous Se@SiO2 NPs & identification of SCs

    Figure 1A illustrates the fabrication process of porous Se@SiO2 NPs. Figure 1B shows the phase structure of Se@SiO2 NPs analyzed by x-ray diffractometer, and the characteristic peaks of Se@SiO2 NPs show the standard Se hexagonal phase except for the amorphous silica leading to an increase in the low-angle region.

    Figure 1. Characterization of the solid or porous Se@SiO2 NPs and identification of satellite cells.

    (A) Fabrication process of porous Se@SiO2 NPs. (B) x-ray diffractometer pattern of the solid Se@SiO2 NPs and the typical selenium nanocrystals (Joint Committee on Powder Diffraction Standards card no. 65–1,876). (C) Low and (D) high magnification of transmission electron microscopy images of the solid Se@SiO2 NPs. (E) Transmission electron microscopy images of porous Se@SiO2 NPs. (F)PAX7 and desmin were detected by immunofluorescence; cytoskeletal alterations were detected by staining with F-actin. The nuclei of satellite cells were stained with Hoechst. The scale bar represents 20 μm. (G) Cell proliferation in satellite cells in response to different porous Se@SiO2 NP concentrations.

    *p < 0.05; **p < 0.01; ***p < 0.001 compared with control (0 μg/ml).

    DIW: Deionized water; NP: Nanoparticle; PVP: Polyvinylpyrrolidone; TEOS: Tetraethyl orthosilicate.

    Transmission electron microscopy was used to measure the size and morphology of the porous Se@SiO2 NPs. Porous Se@SiO2 NPs had an average diameter of 55 nm; from the center to the surface, there were numerous irregular quantum dots (<5 nm; Figure 1C & D). The porous Se@SiO2 NPs exhibited a porous structure being treated with hot water (Figure 1E). The SCs were identified by immunofluorescence staining for PAX7 and desmin, which are specific markers, and cytoskeletal alterations were detected by staining for F-actin (Figure 1F). This result confirmed that the isolated cells were SCs because PAX7 was distributed in the nucleus and desmin was present in the cytoplasm. The safety of porous Se@SiO2 NPs was ascertained by determining the proliferation of cells in the presence of porous Se@SiO2 NPs. Porous Se@SiO2 NPs were added to the cells at concentrations ranging from 0 to 160 μg/ml. Compared with the control group, SCs showed different cell proliferation patterns, wherein the concentrations of 10 and 20 μg/ml significantly promoted cell proliferation, and concentrations above 40 μg/ml inhibited cell proliferation (Figure 1G). Therefore, for the porous Se@SiO2 NPs used in the subsequent experiments, the authors chose to conduct them at a concentration of 20 μg/ml, which has a positive effect on cell proliferation.

    Porous Se@SiO2 NPs protect SCs against H2O2-induced injury

    The MTT assay showed that porous Se@SiO2 NPs promoted cell proliferation (Figure 2A). Compared with the control group, cell proliferation was significantly enhanced in the Se@SiO2 group on days 1, 2 and 3. The cell proliferation was also higher in the Se@SiO2 + H2O2 group than in the H2O2 group. These results demonstrated that porous Se@SiO2 NPs remarkably enhanced the proliferation of SCs, and the inhibitory effect of H2O2 on cell growth was reduced.

    Figure 2. Porous Se@SiO2 nanoparticles protect satellite cells against H2O2-induced injury.

    (A) Absorbance response of the proliferation of different groups of cells on days 1, 2 and 3. (B) Senescence-associated β-galactosidase staining shows senescent cells as light blue. The scale bar represents 50 μm. (C) Staining of live–dead cells of satellite cells; live cells are shown in green, while dead cells are displayed in red. The scale bar represents 20 μm. (D) Analysis of the proportion of live–dead cells.

    *p < 0.05; **p < 0.01; ***p < 0.001.

    OD: Optical density; NP: Nanoparticle.

    In the staining of live–dead cells (Figure 2C & D), the green dye can permeate the cell membrane of live eukaryotic cells and stain the cell green, whereas the red dye stain can only label dead cells, since it is membrane impermeable. On day 3 of cell culture, H2O2 significantly increased cell death compared with the control group; however, the addition of porous Se@SiO2 NPs reduced cell death compared with that in the H2O2 group.

    On day 3, senescent cells were detected based on the expression of SA-β-gal, a prominent biomarker of aging. The addition of porous Se@SiO2 NPs did not affect cellular senescence (Figure 2B). Blue-stained SA-β-gal positivity was significantly elevated in the SCs after treatment with H2O2 compared with the control. Blue-stained SA-β-gal positivity was reduced significantly in the SCs treated with porous Se@SiO2 NPs. These results show that porous Se@SiO2 NPs significantly reduce the increase in cell senescence caused by oxidative stress.

    Porous Se@SiO2 NPs promote mitochondrial function under oxidative stress

    The authors examined the ability of porous Se@SiO2 NPs to promote the clearance of ROS in mitochondria on days 3 and 7. Porous Se@SiO2 NPs did not significantly affect the control group but elevated mitochondrial ROS levels after exposure to H2O2 (Figure 3A & B).

    Figure 3. Porous Se@SiO2 nanoparticles promote mitochondrial function under oxidative stress.

    (A & C) The representative confocal images illustrate the effect of Se@SiO2 nanoparticles on the formation of reactive oxygen species from mitochondria induced by H2O2 in satellite cells on days 3 (A) and 7 (B); MitoSOX Red Mitochondrial Superoxide Indicator was used to stain mitochondrial reactive oxygen species in red, and blue Hoechst dye stained the nucleus. Each image was captured at the same magnification. The scale bar represents 50 μm. (C) Western blot analysis of SIRT1, SOD1 and GAPDH expression in satellite cells from the four groups. Results were normalized to GAPDH expression and compared in (D) and (E).

    *p < 0.05; **p < 0.01; ***p < 0.001.

    To illustrate the underlying antioxidant mechanisms of porous Se@SiO2 NPs, the authors studied the protein levels of SIRT1. This regulates mitochondrial biogenesis by interacting with the transcription coactivator PGC-1α and the removal of damaged mitochondria. It is also associated with the self-renewal of SCs [16,34] and SOD1, which is an important antioxidant enzyme. Compared with the control group, H2O2 treatment considerably decreased SIRT1 and SOD1 expression. Compared with the H2O2 group, the addition of porous Se@SiO2 NPs enhanced the expression of SIRT1 and SOD1 in an oxidative stress environment (Figure 3C–E).

    Porous Se@SiO2 NPs enhance mitochondrial activity & dynamics under H2O2 stimulation in SCs

    Overaccumulation of H2O2 can cause oxidative damage, mitochondrial dysfunction and ultimately cell death. Therefore, the authors examined the most important H2O2 scavenging enzyme, CAT. Compared with the control group, the addition of porous Se@SiO2 NPs promoted CAT activity, but the activity of CAT decreased after treatment with H2O2. Compared with the H2O2 group, the activity of intracellular CAT increased after the addition of porous Se@SiO2 NPs (Figure 4A). To investigate the effects of porous Se@SiO2 NPs on mitochondrial respiration, the authors tested the activities of MRC complex I and III and ATP synthase in the respiratory chain (Figure 4B–D). Compared with the control group, the activity of MRC complex I and III, but not ATP synthase, was increased by adding porous Se@SiO2 NPs. The activity of MRC complex I and III and ATP synthase decreased after H2O2 treatment. Compared with the H2O2 group, the activity of MRC complex I and III and ATP synthase increased after the addition of porous Se@SiO2 NPs.

    Figure 4. Porous Se@SiO2 nanoparticles enhance mitochondrial activity and dynamics under H2O2 stimulation in satellite cells.

    (A) Catalase activity was affected by H2O2 and Se@SiO2 nanoparticles. Activities of mitochondrial respiratory chain complex I (B), mitochondrial respiratory chain complex III (C) and ATP synthase (D) in satellite cells. nanoparticles concentration = 20 μg/ml, H2O2 = 100 μM. (E) The mitochondrial morphology of satellite cells as represented by confocal images; MitoTracker Red CMXRos staining solution was used to label the mitochondria, which were displayed in red; cytoskeletal alterations were detected by staining with F-actin and are displayed in green; blue Hoechst dye stained the nucleus. Each image was captured at the same magnification. The scale bar represents 50 μm. (F) Quantification of the red fluorescence intensity in each group.

     *p < 0.05; **p < 0.01; ***p < 0.001.

    CAT: Catalase; MRC: Mitochondrial respiratory chain.

    The mitochondrial dynamics and morphology of SCs were studied by confocal laser microscopy. The general morphology of mitochondria was observed by labeling them with a red MitoTracker probe, and cytoskeletal alterations were detected by staining with F-actin to observe the overall shape of the cell and to make sure that the fluorescent probe correctly labeled the mitochondria in cells (Figure 4E & F). Both the control group and the Se@SiO2 group had numerous mitochondria distributed in the perinuclear areas with high density. Compared with the control group, mitochondrial fragmentation increased, and the density was decreased in H2O2-stimulated cells. Compared with the H2O2 group, the addition of the porous Se@SiO2 NPs increased the mitochondrial density, but the mitochondrial structure was still quite different from that in the control group.

    Porous Se@SiO2 NPs promote myogenic differentiation of SCs

    The authors further investigated the influence of porous Se@SiO2 NPs on the myogenic differentiation of SCs under oxidative stress on day 7. MYHC, which is responsible for muscle production in mammals, and MYOD, the early marker of myogenesis, were used to determine the extent of myogenic differentiation. Compared with the control group, the addition of porous Se@SiO2 NPs increased the expression of MYOD; however, the expression of MYHC and MYOD decreased significantly after H2O2 treatment. Compared with the H2O2 group, the addition of porous Se@SiO2 NPs increased the expression of both (Figure 5A–C). Immunofluorescence staining was used to observe myotubes by MYHC-specific antibody and the expression of MYOD (Figure 5D & F). The results were consistent with the results of the western blot analysis. Compared with the control group, the addition of porous Se@SiO2 NPs can increase the mean fluorescence intensity of MYOD, and exposure to H2O2 decreased the fusion index. It also reduced the mean fluorescence intensity of MYOD. Compared with the H2O2 group, the addition of porous Se@SiO2 NPs increased the fusion index and mean fluorescence intensity of MYOD (Figure 5E & G).

    Figure 5. Porous Se@SiO2 nanoparticles promote myogenic differentiation of satellite cells.

    (A) Western blot analysis of MYHC, MYOD and GAPDH expression in satellite cells after normalization to GAPDH; comparison shown in (B) and (C). (D & F) Images showing the effect of H2O2 and porous Se@SiO2 nanoparticles on the myogenic differentiation of satellite cells. MYHC in satellite cells myotubes and MYOD are shown in red, and Hoechst-stained nuclei appear blue. All images were captured at the same magnification. The scale bar represents 20 μm. (E) Myotube fusion index. (G) Mean fluorescence intensity of MYOD.

    *p < 0.05; **p < 0.01; ***p < 0.001.

    Porous Se@SiO2 NPs accelerate muscle regeneration & reduce collagen deposition in vivo

    The authors further verified the therapeutic effect of porous Se@SiO2 NPs on muscle regeneration after oxidative stress injury in rats. By intramuscularly injecting CTX into rats, the authors established a classical rat model of acute muscle injury (Figure 6A & B). Compared with the control group, porous Se@SiO2 NPs alone did not affect the normal skeletal muscles (Figure 6C). On day 1 after CTX injection, severe degeneration and necrosis of muscle fibers and exudation of numerous nuclei were seen in the damaged area, indicating successful model construction (Figure 6C). Compared with the control group, more newly formed muscle fibers with nuclei located in the center appeared 7 days after injury, accompanied by neovascularization in the CTX + Se@SiO2 group (Figure 6D). On day 14, many newly formed muscle cells fused into muscle tubes compared with the CTX group. The regenerated skeletal muscle fibers displayed a more orderly arrangement and had a larger diameter after porous Se@SiO2 NP treatment. On day 28, the muscle cells differentiated and matured. The transverse diameter of the muscle fibers was close to that of normal muscle, and the nucleus was located under the membrane. In the CTX group, the muscle cells were completely repaired, but they were still different from normal muscle cells because they had a disordered arrangement of muscle fibers. In the CTX + Se@SiO2 group, the muscle tissue was completely regenerated and showed almost no difference from normal muscle tissue.

    Figure 6. Porous Se@SiO2 nanoparticles accelerate muscle regeneration and reduce collagen deposition in vivo.

    (A) CTX injection into the tibialis anterior (TA) muscle and (B) isolation of the TA muscle. (C & D) The midbelly region of the rat TA muscles in the transverse paraffin section stained with hematoxylin and eosin. The scale bar represents 20 μm. (C) TA muscle morphology of the control group. Porous Se@SiO2 NP treatment had no significant effect on normal muscle. TA muscle morphology 1 day after CTX injection. (D) TA muscle morphology 7, 14 and 28 days after injection of CTX and CTX + porous Se@SiO2 NPs. (E & F) Masson trichrome staining of the midbelly region of the rat TA muscles in transverse paraffin section on days (E) 14 and (F) 28. The scale bar represents 20 μm.

    CTX: Cardiotoxin; NP: Nanoparticle.

    At 14 days after injury, images of Masson trichrome staining showed the presence of collagenous tissue, which indicated active collagen deposition in the CTX group compared with the CTX + Se@SiO2 group (Figure 6E). At 28 days after injury, less positive staining was observed in the gaps around regenerated myofibers in each group. Compared with the CTX group, porous Se@SiO2 NP treatment reduced collagen deposition in the later stages of muscle regeneration (Figure 6F).

    Porous Se@SiO2 NPs promote the formation of myotubes & increase the number of PAX7-positive cells

    The authors assessed skeletal muscle regeneration at protein levels on days 14 and 28. The changes were the same for both MYHC and MYOD. Thus, porous Se@SiO2 NP treatment can be considered to significantly increase the expression of MYHC and MYOD on days 14 and 28 after muscle injury (Figure 7A–C). This validated the previous results from in vitro experiments. The authors also used immunofluorescence analysis to observe muscle regeneration more intuitively (Figure 7D & E). Double immunostaining for MYHC and dystrophin with nuclear staining of sections indicated that the fluorescence intensity of MYHC increased with time. Local magnification of the image allowed clearer visualization of the double staining of MYHC and dystrophin, and the nucleus was located under the membrane. Treatment with porous Se@SiO2 NPs could accelerate skeletal muscle regeneration on days 14 and 28 after injury.

    Figure 7. Porous Se@SiO2 nanoparticles promote the formation of myotubes and increase the number of PAX7-positive cells.

    (A) Western blot analysis of MYHC, MYOD and GAPDH expression of satellite cells in each group after normalization to GAPDH expression; the comparison is shown in (B) and (C). (D) Immunofluorescence staining of MYHC was observed with dystrophin staining as background. Immunofluorescence analysis for MYHC (red) and dystrophin (green). Nuclei were stained with Hoechst, displayed in blue. The scale bar represents 20 μm. (E) Quantification of MYHC mean fluorescence intensity. (F) Representative images of PAX7 observed with dystrophin staining as background. Immunofluorescence analysis of PAX7 (red) and dystrophin (green). Nuclei were stained with Hoechst, displayed in blue. The scale bar represents 20 μm. (G) Quantification of PAX7 expression.

    *p < 0.05; **p < 0.01; ***p < 0.001.

    CTX: Cardiotoxin.

    PAX7 was specifically expressed in SCs. Double immunostaining for PAX7 and dystrophin with nuclear staining of sections was performed, and the number of PAX7-positive nuclei was determined in the muscle samples. The number of PAX7-positive cells was significantly higher in skeletal muscle after 14 days of action of porous Se@SiO2 NPs than in the CTX group. (Figure 7F & G).

    Discussion

    The proliferation and differentiation of SCs are vital for skeletal muscle regeneration after injury and cannot be replaced by unorthodox myogenic precursor cells [35]. To enhance the regenerative ability of a skeletal muscle, it is necessary to recognize the control mechanisms that determine the fate of SCs. Advancements in SCs have revealed that the dynamic metabolism, mitochondrial structure and activity are associated with SCs differentiation, and consequently drives muscle regeneration [12,17,18]. This study was based on exploring the effect of porous Se@SiO2 NPs on the proliferation and differentiation of SCs by modulating mitochondria under oxidative stress, and it examined the effect of these NPs on muscle regeneration after injury.

    Se is an essential micronutrient for the body and is incorporated into selenoproteins in the form of selenocysteine. Selenoproteins have oxidoreductase activity, thereby maintaining the physiological redox balance [36]. Se supplementation attenuates ischemia–reperfusion injury in the skeletal muscle and heart due to its antioxidant and anti-inflammatory properties [3739]. However, excessive aggregation of Se may have harmful effects on health. There is disagreement on the safe upper limit of Se. According to research, Se at the nanoscale level can reduce the risk of Se toxicity [27]; therefore, the authors of the present study developed porous Se@SiO2 NPs. The diameter of NPs made them suitable for absorption by cells. In a previous study, porous Se@SiO2 NPs labeled with fluorescein isothiocyanate were observed by confocal microscopy to enter the cytoplasm and elicit biological effects [26]. The release curve of porous Se@SiO2 NPs in vitro simulates the levels in the human body, indicating that NPs can achieve sustained release and get effectively distributed in the body [24]. Major organs are not affected by tolerable concentrations of porous Se@SiO2 NPs [29,40]. With the extension of time due to porous Se@SiO2 NPs, the problem of local excessive Se accumulation in the tissue is decreased dramatically, avoiding toxicity concerns [41]. The safety of porous Se@SiO2 NPs was confirmed in this study as well. The MTT assay showed that appropriate concentrations of porous Se@SiO2 NPs did not inhibit cell proliferation, indicating that porous Se@SiO2 NPs may be an ideal and safe material for further studies.

    When serious muscle trauma occurs, pathological stages, including the destruction phase, repair phase and remodeling phase, occur [29,30]. A large amount of ROS is produced after injury. ROS are a class of molecules that are continuously generated, converted and consumed in all living organisms, and they include H2O2, superoxide and hydroxyl radicals, which are generated mainly by electron leakage from the electron transport chain in mitochondria [6,42,43]. For SCs, the role of ROS is extremely complex. The production of ROS is related to the activation and subsequent differentiation of SCs. Moderate levels of ROS are necessary for myogenic differentiation as a signal molecule, while high levels of ROS lead to apoptosis or necrosis. High ROS levels also upregulate NF-κβ, which downregulates MYOD levels, thereby impairing myogenic differentiation [44,45]. In this study, a high concentration of H2O2 was used to induce oxidative stress injury in SCs. The porous Se@SiO2 NPs significantly decreased mitochondrial ROS levels under H2O2 exposure. Thus, porous Se@SiO2 NPs protected SCs from H2O2-induced cell damage and senescence. The fate of SCs in myogenic differentiation is regulated by MYOD at an early stage, and eventually cells exit the cell cycle and merge into the regenerated myotubes [46]. The level of myogenic differentiation was based on the expression of MYOD and MYHC; the expression of both these markers was reduced after H2O2 treatment. However, porous Se@SiO2 NPs suppressed the inhibitory effect of H2O2 on MYHC and MYOD. Thus, porous Se@SiO2 NPs could counteract the H2O2-induced inhibition of myogenic differentiation in SCs.

    Excessive levels of ROS are detrimental to mitochondria in SCs because they could target mitochondrial DNA and mitochondrial function, causing swelling and disruption of mitochondria [47,48]. Therefore, it is necessary to promote cellular homeostasis for protecting mitochondria. Mitochondrial damage and dysfunction can further lead to damage to the entire cell. To protect SCs from the underlying damage of excess ROS, mitochondria have their antioxidant systems, particularly superoxide dismutase and glutathione peroxidase, which scavenge ROS to sustain intracellular stability [45]. Thus, antioxidants are essential for mitigating cellular oxidative damage through mitochondria. Appropriate doses of antioxidants can maintain the level of ROS at an optimal level, so that ROS can act as physiological signaling molecules [25]. In this study, the porous Se@SiO2 NPs protected the morphological and functional stability of mitochondria by increasing the number of mitochondria and promoting mitochondrial respiration. To illustrate the potential antioxidant mechanism of porous Se@SiO2 NPs, the authors evaluated the expression of SOD1, which acts as an antioxidant enzyme, and SIRT1, which regulates mitochondrial genesis and removes damaged mitochondria. The results demonstrated that porous Se@SiO2 NPs upregulated the expression of SIRT1 and SOD1. This was consistent with previous studies that demonstrated that porous Se@SiO2 NPs reduce ROS levels by promoting the production of major endogenous antioxidants, including superoxide dismutase and glutathione [29].

    The effect of porous Se@SiO2 NPs was next investigated in a CTX-induced TA injury model. The results were consistent with the results of the in vitro experiments. Porous Se@SiO2 NPs upregulated the expression of MYHC and MYOD in the injured muscle on days 14 and 28. Morphological changes in muscle regeneration were observed in hematoxylin and eosin-stained sections, and treatment with porous Se@SiO2 NPs promoted more rapid muscle fiber regeneration and more regular arrangement between muscle fibers. Interestingly, porous Se@SiO2 NPs significantly increased the number of PAX7-positive cells in the injured region on day 14 compared with the CTX group, but there was no statistical significance on day 28. The authors speculate that this is because of the complete regeneration of the muscle on day 28 and because no additional SCs were required for skeletal muscle regeneration. Skeletal muscle regeneration is accompanied by fibrosis development, muscle fibrosis and oxidative stress, which are closely related to each other [49,50]. SCs may also contribute to fibrosis through the WNT signaling pathway [51]. In addition to a longer time for repair, fibrotic repair also disrupts the mechanical properties of the muscle and makes the affected muscle more vulnerable to reinjury [52]. The authors confirmed that porous Se@SiO2 NPs attenuated collagen deposition after injury, but its detailed mechanism is not known.

    Although this study confirmed that porous Se@SiO2 NPs promote skeletal muscle regeneration by modulating mitochondria to attenuate oxidative damage, additional studies are necessary to elucidate the potential impact of porous Se@SiO2 NPs on other mechanisms regulating mitochondrial homeostasis such as mitochondrial autophagy in SCs. Furthermore, the biological effects of NPs depend on their targeting efficiency, and further improvements in material properties will enable them to enter the mitochondria more effectively to exert their biological effects. In addition, acute degenerative necrosis of the skeletal muscle induced by CTX in this experiment is still somewhat different from skeletal muscle injuries seen in the clinic. Porous Se@SiO2 NPs should be further explored to investigate their potential therapeutic effects.

    Conclusion & future perspective

    This study provides initial evidence that porous Se@SiO2 NPs significantly promote the proliferation and myogenic differentiation of SCs under oxidative stress by scavenging mitochondrial ROS and improving mitochondrial activity (Figure 8). Treatment with porous Se@SiO2 NPs accelerated skeletal muscle regeneration and reduced fibrosis deposition after injury. Our results reveal the application potential of porous Se@SiO2 NPs in skeletal muscle diseases or other pathologies related to mitochondrial dysfunction. Further research is necessary in the future to improve the targeting efficiency of NPs; and safety studies in large animal models to facilitate better clinical translation.

    Figure 8. Porous Se@SiO2 nanoparticles promote skeletal muscle regeneration by inducing the stabilization of mitochondrial morphology and function and attenuating the oxidative damage of satellite cells.

    CTX: Cardiotoxin; NP: Nanoparticle; Oxphos: Oxidative phosphorylation; ROS: Reactive oxygen species.

    Summary points
    • Skeletal muscle injury produces an oxidative stress microenvironment that hinders skeletal muscle regeneration.

    • An oxidative stress microenvironment causes oxidative damage and dysfunction of mitochondria in skeletal muscle stem cells, resulting in impaired proliferation and differentiation.

    • Selenium (Se) is a trace element with antioxidant and other biological activities, but its application is currently very narrow.

    • The authors have developed porous Se@SiO2 nanoparticles (NPs) with slow and sustained release of Se.

    • Porous Se@SiO2 NPs have controlled slow release and better bioavailability than conventional forms of Se.

    • In vitro experiments demonstrated the role of Se NPs in regulating mitochondrial function and protecting satellite cell differentiation.

    • In vivo experiments demonstrated that porous Se@SiO2 NPs can promote the regeneration of skeletal muscle after acute injury.

    • This study suggests that porous Se@SiO2 NPs offer new therapeutic options for diseases related to mitochondrial dysfunction.

    Author contributions

    Substantial contributions to the conception or design of the work or the acquisition, analysis or interpretation of data for the work: Y-X Yang, M-S Liu, X-J Liu, E-K Pang, L Hou; drafting of the work or revising it critically for important intellectual content: Y-X Yang; final approval of the version to be published: Y-X Yang, M-S Liu, Y-C Zhang, Y-Y Hu, R-S Gao; agreement to be accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved: Y-X Yang, J-C Wang, W-Y Fei. Y-X Yang and M-S Liu contributed equally to this work and should be considered co-first authors.

    Acknowledgments

    The authors would like to express their special thanks to Q Kong for her assistance with writing and statistical analysis.

    Financial & competing interests disclosure

    This research was supported by the fund from the Clinical Application Oriented Medical Innovation through the National Orthopedics and Sports Rehabilitation Clinical Medical Research Center (contract number: 2021-NCRC-CXJJ-PY-07), the Scientific Research Project of Jiangsu Provincial Health and Health Commission (item number: M2021042) and the Yangzhou Social Development Project (item number: YZ2021084). The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

    No writing assistance was utilized in the production of this manuscript.

    Ethical conduct of research

    The authors state that they have obtained appropriate institutional review board approval or have followed the principles outlined in the Declaration of Helsinki for all human or animal experimental investigations. There are no investigations involving human subjects in this paper.

    Data sharing statement

    The data used to support the findings of this study are available from the corresponding author upon request.

    Open access

    This work is licensed under the Attribution-NonCommercial-NoDerivatives 4.0 Unported License. To view a copy of this license, visit http://creativecommons.org/licenses/by-nc-nd/4.0/

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. Wong S, Ning A, Lee C, Feeley BT. Return to sport after muscle injury. Curr. Rev. Musculoskelet. Med. 8(2), 168–175 (2015).
    • 2. Zissler A, Steinbacher P, Zimmermann R et al. Extracorporeal shock wave therapy accelerates regeneration after acute skeletal muscle injury. Am. J. Sports Med. 45(3), 676–684 (2017).
    • 3. Smith C, Kruger MJ, Smith RM, Myburgh KH. The inflammatory response to skeletal muscle injury: illuminating complexities. Sports Med. 38(11), 947–969 (2008).
    • 4. Gharaibeh B, Chun-Lansinger Y, Hagen T et al. Biological approaches to improve skeletal muscle healing after injury and disease. Birth Defects Res. C Embryo Today 96(1), 82–94 (2012).
    • 5. Qazi TH, Duda GN, Ort MJ, Perka C, Geissler S, Winkler T. Cell therapy to improve regeneration of skeletal muscle injuries. J. Cachexia Sarcopenia Muscle 10(3), 501–516 (2019).
    • 6. Mann CJ, Perdiguero E, Kharraz Y et al. Aberrant repair and fibrosis development in skeletal muscle. Skelet. Muscle 1(1), 21 (2011).
    • 7. Buckingham M, Montarras D. Skeletal muscle stem cells. Curr. Opin. Genet. Dev. 18(4), 330–336 (2008).
    • 8. Zammit PS, Relaix F, Nagata Y et al. Pax7 and myogenic progression in skeletal muscle satellite cells. J. Cell Sci. 119(9), 1824–1832 (2006).
    • 9. Relaix F, Zammit PS. Satellite cells are essential for skeletal muscle regeneration: the cell on the edge returns centre stage. Development 139(16), 2845–2856 (2012).
    • 10. Shi H, Xie H, Zhao Y et al. Myoprotective effects of bFGF on skeletal muscle injury in pressure-related deep tissue injury in rats. Burns Trauma 4, s41038-016-0051-y (2016).
    • 11. Zhu Y, Dean AE, Horikoshi N, Heer C, Spitz DR, Gius D. Emerging evidence for targeting mitochondrial metabolic dysfunction in cancer therapy. J. Clin. Invest. 128(9), 3682–3691 (2018).
    • 12. Bhattacharya D, Scimè A. Mitochondrial function in muscle stem cell fates. Front. Cell Dev. Biol. 8, 480 (2020). • Details how mitochondrial content, function, dynamics and adaptations affect the fate of satellite cells.
    • 13. Bhatti JS, Bhatti GK, Reddy PH. Mitochondrial dysfunction and oxidative stress in metabolic disorders – a step towards mitochondria based therapeutic strategies. Biochim. Biophys. Acta Mol. Basis Dis. 1863(5), 1066–1077 (2017).
    • 14. Abrigo J, Simon F, Cabrera D, Vilos C, Cabello-Verrugio C. Mitochondrial dysfunction in skeletal muscle pathologies. Curr. Protein and Peptide Sci. 20(6), 536–546 (2019).
    • 15. Kowaltowski AJ, de Souza-Pinto NC, Castilho RF, Vercesi AE. Mitochondria and reactive oxygen species. Free Radic. Biol. Med. 47(4), 333–343 (2009).
    • 16. Le Moal E, Pialoux V, Juban G et al. Redox control of skeletal muscle regeneration. Antioxid. Redox Signal. 27(5), 276–310 (2017).
    • 17. García-Prat L, Martínez-Vicente M, Muñoz-Cánoves P. Methods for mitochondria and mitophagy flux analyses in stem cells of resting and regenerating skeletal muscle. In: Skeletal Muscle Regeneration in the Mouse. Kyba M (Ed.). Springer New York, NY, USA, 223–240 (2016).
    • 18. Pessemesse L, Tintignac L, Blanchet E et al. Regulation of mitochondrial activity controls the duration of skeletal muscle regeneration in response to injury. Sci. Rep. 9(1), 12249 (2019). • Illustrates that mitochondrial activity plays an important role in acute skeletal muscle injury.
    • 19. Dell'Orso S, Juan AH, Ko K-D et al. Single-cell analysis of adult skeletal muscle stem cells in homeostatic and regenerative conditions. Development 146(12), dev.174177 (2019).
    • 20. Myburgh KH, Kruger MJ, Smith C. Accelerated skeletal muscle recovery after in vivo polyphenol administration. J. Nutr. Biochem. 23(9), 1072–1079 (2012).
    • 21. Rayman MP. The importance of selenium to human health. Lancet 356(9225), 233–241 (2000).
    • 22. Zhang J, Wang X, Xu T. Elemental selenium at nano size (nano-Se) as a potential chemopreventive agent with reduced risk of selenium toxicity: comparison with Se-methylselenocysteine in mice. Toxicol. Sci. 101(1), 22–31 (2008).
    • 23. Sun H-J. Arsenic and selenium toxicity and their interactive effects in humans. Environ. Int. 69, 148–158(2014).
    • 24. Li C, Wang Q, Gu X et al. Porous Se@SiO2 nanocomposite promotes migration and osteogenic differentiation of rat bone marrow mesenchymal stem cell to accelerate bone fracture healing in a rat model. Int. J. Nanomed. 14, 3845–3860 (2019). •• Reports on the potential of porous Se@SiO2 nanoparticles in the locomotor system.
    • 25. Groitl B, Jakob U. Thiol-based redox switches. Biochim. Biophys. Acta Proteins Proteom. 1844(8), 1335–1343 (2014).
    • 26. Wang M, Wang K, Deng G et al. Mitochondria-modulating porous Se@SiO2 nanoparticles provide resistance to oxidative injury in airway epithelial cells: implications for acute lung injury. Int. J. Nanomed. 15, 2287–2302 (2020). •• Reports the mitochondria-targeted bioactivity modulation of porous Se@SiO2 nanoparticles.
    • 27. Liu X, Deng G, Wang Y et al. A novel and facile synthesis of porous SiO2-coated ultrasmall Se particles as a drug delivery nanoplatform for efficient synergistic treatment of cancer cells. Nanoscale 8(16), 8536–8541 (2016).
    • 28. Musarò A, Carosio S. Isolation and culture of satellite cells from mouse skeletal muscle. In: Adult Stem Cells. Di Nardo PDhingra SSingla DK (Eds). Springer New York, NY, USA, 155–167 (2017).
    • 29. Deng G, Dai C, Chen J et al. Porous Se@SiO2 nanocomposites protect the femoral head from methylprednisolone-induced osteonecrosis. Int. J. Nanomed. 13, 1809–1818 (2018).
    • 30. Garry GA, Antony ML, Garry DJ. Cardiotoxin induced injury and skeletal muscle regeneration. In: Skeletal Muscle Regeneration in the Mouse. Kyba M (Ed.). Springer New York, NY, USA, 61–71 (2016).
    • 31. Gutiérrez J, Escalante T, Hernández R, Gastaldello S, Saravia-Otten P, Rucavado A. Why is skeletal muscle regeneration impaired after myonecrosis induced by viperid snake venoms? Toxins 10(5), 182 (2018).
    • 32. Jensen EC. Quantitative analysis of histological staining and fluorescence using ImageJ: histological staining/fluorescence using ImageJ. Anat. Rec. 296(3), 378–381 (2013).
    • 33. Menconi M, Gonnella P, Petkova V, Lecker S, Hasselgren P-O. Dexamethasone and corticosterone induce similar, but not identical, muscle wasting responses in cultured L6 and C2C12 myotubes. J. Cell. Biochem. 105(2), 353–364 (2008).
    • 34. López-Otín C, Blasco MA, Partridge L, Serrano M, Kroemer G. The hallmarks of aging. Cell 153(6), 1194–1217 (2013).
    • 35. Lepper C, Partridge TA, Fan C-M. An absolute requirement for Pax7-positive satellite cells in acute injury-induced skeletal muscle regeneration. Development 138(17), 3639–3646 (2011).
    • 36. Khurana A, Tekula S, Saifi MA, Venkatesh P, Godugu C. Therapeutic applications of selenium nanoparticles. Biomed. Pharmacother. 111, 802–812 (2019).
    • 37. Yang B-Y, Deng G-Y, Zhao R-Z et al. Porous Se@SiO2 nanosphere-coated catheter accelerates prostatic urethra wound healing by modulating macrophage polarization through reactive oxygen species-NF-κB pathway inhibition. Acta Biomaterialia 88, 392–405 (2019).
    • 38. Gholami M, Zendedel A, Khanipour Khayat Z, Ghanad K, Nazari A, Pirhadi A. Selenium effect on ischemia-reperfusion injury of gastrocnemius muscle in adult rats. Biol. Trace Elem. Res. 164(2), 205–211 (2015). •• Reveals the important role of Se antioxidant damage.
    • 39. Ostadalova I, Vobecky M, Chvojkova Z et al. Selenium protects the immature rat heart against ischemia/reperfusion injury. Mol. Cell. Biochem. 300(1–2), 259–267 (2007).
    • 40. Zheng Z, Deng G, Qi C et al. Porous Se@SiO2 nanospheres attenuate ischemia/reperfusion (I/R)-induced acute kidney injury (AKI) and inflammation by antioxidative stress. Int. J. Nanomed. 14, 215–229 (2018).
    • 41. Li X, Wang Q, Deng G et al. Porous Se@SiO2 nanospheres attenuate cisplatin-induced acute kidney injury via activation of Sirt1. Toxicol. Appl. Pharmacol. 380, 114704 (2019).
    • 42. Goncalves RLS, Quinlan CL, Perevoshchikova IV, Hey-Mogensen M, Brand MD. Sites of superoxide and hydrogen peroxide production by muscle mitochondria assessed ex vivo under conditions mimicking rest and exercise. J. Biol. Chem. 290(1), 209–227 (2015).
    • 43. Dickinson BC, Chang CJ. Chemistry and biology of reactive oxygen species in signaling or stress responses. Nat. Chem. Biol. 7(8), 504–511 (2011).
    • 44. Ardite E, Barbera JA, Roca J, Fernández-Checa JC. Glutathione depletion impairs myogenic differentiation of murine skeletal muscle C2C12 cells through sustained NF-κB activation. Am. J. Pathol. 165(3), 719–728 (2004).
    • 45. Kozakowska M, Pietraszek-Gremplewicz K, Jozkowicz A, Dulak J. The role of oxidative stress in skeletal muscle injury and regeneration: focus on antioxidant enzymes. J. Muscle Res. Cell Motil. 36(6), 377–393 (2015).
    • 46. Singh K, Dilworth FJ. Differential modulation of cell cycle progression distinguishes members of the myogenic regulatory factor family of transcription factors. FEBS J. 280(17), 3991–4003 (2013).
    • 47. Sestili P, Barbieri E, Martinelli C et al. Creatine supplementation prevents the inhibition of myogenic differentiation in oxidatively injured C2C12 murine myoblasts. Mol. Nutr. Food Res. 53(9), 1187–1204 (2009).
    • 48. Kujoth GC, Hiona A, Pugh TD et al. Mitochondrial DNA mutations, oxidative stress, and apoptosis in mammalian aging. Science 309(5733), 481–484 (2005).
    • 49. Serrano AL, Muñoz-Cánoves P. Regulation and dysregulation of fibrosis in skeletal muscle. Exp. Cell Res. 316(18), 3050–3058 (2010).
    • 50. Bersini S, Gilardi M, Mora M et al. Tackling muscle fibrosis: from molecular mechanisms to next generation engineered models to predict drug delivery. Adv. Drug Deliv. Rev. 129, 64–77 (2018).
    • 51. Brack AS, Conboy MJ, Roy S et al. Increased Wnt signaling during aging alters muscle stem cell fate and increases fibrosis. Science 317(5839), 807–810 (2007).
    • 52. Li EW, McKee-Muir OC, Gilbert PM. Cellular biomechanics in skeletal muscle regeneration. Curr. Top Dev. Biol. 126, 125–176 (2018).