1. Introduction
Skin wound healing is a complex biological process that involves keratinocytes, fibroblasts, and endothelial cells. Wounds can be caused by pathological conditions, such as surgery, injury, diabetes, or vascular disease [
1]. These injuries are categorized as acute or chronic wounds depending on their underlying cause and consequences. Acute wounds, such as abrasions, bites, and lacerations, tend to heal within two weeks. Disturbances during the wound healing phase can cause delays in healing, leading to chronic wounds, which can persist for longer than three months and may never fully heal. Under certain conditions, burns and bites may develop into chronic infections that last a lifetime, which not only seriously affects the quality of life and physical and mental health of patients, but also increases the treatment cost and burdens the economic situation of patients [
2,
3]. Patients with diabetes mellitus, vascular disease, aging, and hemoglobinopathies are especially prone to abnormal wound healing, which, if not properly addressed, can lead to wound recurrence and even amputation or death in severe cases [
4,
5].
Wounds can be divided into hemostatic, inflammatory, proliferative, and maturation stages from wound formation to tissue healing [
6,
7,
8]. The body’s first response to a wound is to constrict injured blood vessels and activate platelets to form a fibrin clot, which blocks blood flow and serves as a scaffold for incoming inflammatory cells [
9]. The immune system and immune cells are then activated to combat self and foreign antigens. The onset of angiogenesis marks the end of the inflammatory phase, as endothelial cells proliferate, migrate, and branch into new blood vessels. Following the emergence of the new blood vessels, the existing fibroblasts proliferate and differentiate into myofibroblasts, which pull the edges of the wound together. This causes the microenvironment of the wound to shift from an inflammatory state to a growth state, with re-epithelialization occurring simultaneously [
10,
11,
12,
13].
Mesenchymal stem cells (MSCs) are pluripotent mesenchymal cells, and the paracrine effects of MSCs play a role in promoting angiogenesis, proliferation, and the migration of epithelial cells and fibroblasts during the wound healing response [
14,
15,
16,
17,
18]. Stem cell exosomes can be used as nanoscale membrane-bound vesicular particles with sizes ranging from 30–150 nm [
19]. They have the advantages of high stability, low immunogenicity, biocompatibility, and a long circulation time, making them natural drug carriers [
20,
21]. Exosomes can also remodel the extracellular matrix to deliver signals and molecules to other cells, and their use avoids risks such as the tumorigenicity of stem cells and host rejection. Exosomes can migrate to the site of skin injury to perform their functions, improving skin regeneration and reducing skin scarring through inhibiting inflammation and increasing the growth and differentiation capacity of fibroblasts, epidermal cells, and endothelial cells. Exosomes repair wounds by controlling several steps in the wound healing and regeneration process [
14,
22]. MSC exosomes contain extracellular matrix, nucleic acids, and multiple growth factors, which can play a therapeutic role [
23]. In addition, MSC exosomes can also wrap small-molecule drugs with special functions to become drug-loaded exosomes, which have the dual advantages of both exosomes and drugs and play a synergistic effect.
Valproic acid (VPA) is a histone deacetylase (HDAC) inhibitor with potential protective and reparative effects against acute central nervous system (CNS) injury. Because of its good tolerability and safety, VPA has become the drug of choice for the prevention and treatment of epilepsy [
24]. VPA not only mediates nuclear factor kappa-B (NF-κB) to attenuate inflammatory responses, but also down-regulates the production of reactive oxygen species (ROS) [
25,
26]. VPA can attenuate the expression and activation of matrix metalloprotein-9 (MMP-9) through inhibiting histone deacetylases, hinder the production of inflammatory mediators such as TNF-α and IL-6, and influence the cell cycle, cell differentiation, and apoptosis. VPA can promote skin wound healing through enhancing the activity of keratinocytes in mouse models. Cell activity promotes skin wound healing, and VPA also inhibits the inflammatory activation of macrophages and promotes the macrophage phagocytosis of apoptotic cells, suggesting that VPA can accelerate wound closure by acting on macrophages [
27]. Therefore, VPA is considered a safe small-molecule drug with the potential to promote wound repair. In this study, human umbilical cord MSC exosomes loaded with VPA were prepared to explore the therapeutic effect of drug-loaded exosomes in wound healing and their potential mechanism of action, providing new ideas for therapeutic approaches to wound repair.
3. Discussion
The treatment of wound healing has long been a key research area in medicine. Cell therapy through the introduction of biologically active stem cells is not only expected to promote wound healing but also may improve therapeutic efficacy, especially for difficult-to-heal or chronic wounds. The use of stem-cell-derived exosomes in wound healing therapy has received increasing attention and has become a research hotspot in recent years [
35]. In the inflammatory stage of the wound, exosomes can promote macrophage activation and tissue repair. Exosomes have high pro-angiogenic and cell growth factor secretion activity, which can promote cell proliferation and migration, inhibit inflammation, and enhance vascular neogenesis in the process of wound healing. Exosomes have achieved remarkable efficacy in the treatment of tumors and immune diseases and have shown great potential for application in clinical trials, representing an ideal solution for cell-free therapy in the field of regenerative medicine [
36,
37]. In addition, some therapeutic drugs still have limitations in their application. For example, large-molecule drugs are not easily absorbed in the human body, are prone to cytotoxicity, and cannot promote the healing of trauma sites, limiting their clinical application. Exosomes derived from MSCs have certain advantages, such as natural targeting, low immunogenicity, and their ability to be easily frozen and transported. As a result, MSC-derived exosomes are considered to exhibit potential as drug delivery carriers. Exosome-based drug delivery systems can overcome biological barriers and thus provide excellent therapeutic effects. Exosomes can be used as carriers for many types of drugs, including nucleic acids, proteins, and even small-molecule drugs. Using exosomes as drug carriers can increase the solubility, stability, and bioavailability of drugs and improve drug delivery efficiency. This means that MSC exosomes can not only be used in treatment but can also act as drug carriers, thereby improving the drug treatment effect. Drug-loaded exosomes have great therapeutic potential in tissue repair and regeneration. VPA, as a histone deacetylase inhibitor, exhibits anti-inflammatory, antioxidant, and anti-tumor activities, especially for the nerves, with potential protective and restorative effects. VPA can up-regulate the expression of neurotrophic factors, which means that VPA can promote not only angiogenesis but also nerve regeneration, thus further accelerating wound closure [
38]. In the near future, VPA can be utilized in combination with other pro-nerve repair drugs, enabling VPA to play a greater role.
Although the experimental results demonstrate that VPA-EXO has good prospects in wound repair, more clinical practice is needed to verify its effect and safety in actual treatment, which will be favorable for future clinical applications. In addition, in terms of the drug-loading method, indirect drug-loading is commonly used but it also suffers from insufficient characterization and low loading efficiency, while the direct drug-loading method is relatively simple and requires further development. In terms of isolation and purification, the classical ultracentrifugation method requires costly equipment, and the other methods need to be improved in terms of isolation effectiveness and integrity. In the future, it is necessary to further investigate the molecular mechanism of drug-loaded exosomes, as well as factors including the source, isolation methods, culture conditions, drug-loaded methods, and drug delivery schemes. In addition, research in this field should move towards the realization of standardized large-scale production as soon as possible. We believe that drug-loaded exosomes will not only be effective in the field of wound healing and dermatological diseases, but will also enable further breakthroughs in other systemic diseases in the future.
4. Materials and Methods
4.1. MSC-EXO Preparation and Extraction
MSC cells with a fusion rate of 80–90% were washed with phosphate-buffered saline (PBS) two to three times, added to a serum-free medium for stem cells, and cultured for another 24 h. Twenty-four hours later, the cell supernatant was collected and centrifuged at 3000× g for 10 min, after which the supernatant was taken and exosome concentration solution (ECS reagent) was added according to the selected ratio, mixed well, and then allowed to stand for 14 h at 4 °C. After another round of centrifugation (10,000× g for 1 h), the supernatant was discarded and PBS was added for resuspension, followed by another round of centrifugation (12,000× g for 2 min). The supernatant was then transferred to the upper chamber of the exosome purification filter (EPF column). Centrifugation (3000× g for 10 min) was performed a final time and the liquid at the bottom of the EPF column was collected and stored at −80 °C for subsequent use.
4.2. MSC-EXO Identification
4.2.1. Transmission Electron Microscopy (TEM)
Each exosome sample (10 μL) was dropped onto a copper grid so that the hydrophilic side was in contact with the liquid, and the excess liquid was absorbed with filter paper. The sample was washed with ultrapure water and then stained with drops of uranyl acetate (UA) staining solution for 30 s. The sample was allowed to dry naturally. The samples were placed under a transmission electron microscope and the morphology and size of exosomes were observed.
4.2.2. Nanoparticle Tracking Analysis (NTA)
The particle size and concentration of exosomes were detected using a nanoparticle tracking analyzer (NanoSight NS500, Zetaview, Dusseldorf, Germany). Calibration was performed before use and the samples were washed with PBS three times. PBS-diluted samples were added to the nanopore to measure and analyze the particle size and concentration of exosomes.
4.2.3. Western Blot Assay
The lysis solution was added to the sample proportionally and left on ice for 3–5 min. The sample was centrifuged at 12,000× g for 5 min, the supernatant was collected, the sample buffer was added according to the specified proportion, and the sample was heated for 10 min. The gelator was placed into the electrophoresis tank, the electrophoresis solution was added, the sample was dotted in, and electrophoresis was initiated. When the bromophenol blue was about 1 cm from the bottom, electrophoresis was terminated and the membrane was transferred. During the membrane transfer process, the membrane transfer equipment was cooled down. Tris buffered saline with tween (TBST) was added to wash the membrane, skimmed milk was added, and the membrane was closed for 30 min at 25 °C. After washing, the prepared primary antibodies TSG101 (1:1000) and CD81 (1:1000) were added and incubation proceeded at 4 °C overnight. After washing the membrane with TBST, the secondary antibody was added, incubation was performed at room temperature for 2 h, and the membrane was washed again. Well-mixed ECL solution (1:1) was added and allowed to react fully. After 1–2 min, strip images were obtained using a chemiluminescence image analysis system.
4.3. Preparation and Characterization of Drug-Loaded Exosomes
The exosomes and VPA mixture were loaded with the drug using an ultrasonic cell crusher. After sonication, the mixture was incubated at 37 °C for 60 min, and the free drug was isolated via centrifugation at 5000× g for 10 min using an ultrafiltration tube. The drug-loading efficiency of exosomes was determined using liquid chromatography with mobile phase A as acetonitrile: 0.1 mol/L sodium acetate (4.2 g of anhydrous sodium acetate was taken and 30 mL of acetonitrile was dissolved in 970 mL of water) solution (3:97), with the pH adjusted to 6.5 using acetic acid. Mobile phase B consisted of acetonitrile: water (4:1). The flow rate was 1.0 mL/min and the detection wavelength was 210 nm. TEM observations, the NTA assay, and the Western blot assay were also performed on the drug-loaded exosomes as described above.
4.4. Cell Culture for In Vitro Models
Human umbilical cord mesenchymal stem cells (HUC-MSCs) were purchased from SAIYE (Guangzhou, China) Biotechnology Co., Ltd. and cultured to the third generation using serum-free medium for stem cells. Human skin fibroblasts (HSFs) were purchased from Peking Union Medical College Hospital (Beijing, China) and cultured to the ninth generation using Dulbecco’s modified eagle medium (DMEM) high-sugar medium supplemented with fetal bovine serum and penicillin–streptomycin. Human umbilical vein endothelial cells (HUVECs) were purchased from Henan Provincial Industrial Microbial Strain Engineering and Technology Research Center (Nanyang, China) and cultured to the third generation using endothelial cell culture medium for experiments. The cell lines were cultured in a cell culture incubator (37 °C, 5% CO2).
4.5. Cytotoxicity Assay
HSF cells and HUVECs in the logarithmic growth phase were collected and the cell density was adjusted to 8 × 104 cells/mL. The cells were inoculated into 16-well and 96-well plates and incubated at 37 °C and 5% CO2 for 24 h. The 16-well plates were placed into the real-time cell analysis (RTCA, Agilent, Santa Clara, CA, USA) system for real-time monitoring, and the 96-well plates were subjected to the Cell Counting Kit-8 (CCK-8) assay. Intervention treatments using different concentrations of VPA were conducted for 24 h, while control wells were set up. After replacing the original culture medium with culture medium containing 10 μL of CCK-8 solution, the cells were incubated for 2 h and the optical density value at 450 nm was determined using an enzyme-labeling instrument.
4.6. VPA-EXO to Promote Skin Wound Healing In Vitro
4.6.1. Uptake of Drug-Loaded Exosomes
The PKH26-labeled VPA-EXO was utilized in co-culture with HSFs or HUVECs for 24 h. The cells were washed with pre-cooled PBS, fixed with 4% paraformaldehyde for 10 min, washed three times with PBS, blocked using an anti-fluorescent bursting agent, and placed under a fluorescence microscope for observations.
4.6.2. Migration Ability of HSFs and HUVECs
HSF cells and HUVECs in the logarithmic growth phase were collected, the cell density was adjusted to 5 × 105 cells/mL, the two-well inserts from the scratch assay were placed into six-well plates, and 70 μL of cell suspension was added to the inserts. The cells were cultured in an incubator at 37 °C and 5% CO2 for 24 h. One milliliter each of different drugs was added for treatment. Culture was performed in an incubator and pictures were collected at the same position at 0, 4, 8, 12, and 24 h of culture. The width and area of the scratched area were measured using Image J software, and the cell migration rate at each period was calculated as (scratched area at 0 h − scratched area at x h)/scratched area at 0 h.
4.6.3. Transwell Assay of the Invasive Ability of HSFs and HUVECs
A cell invasion assay was performed in 24-well plates in a Transwell filtration chamber. First, 600 μL of serum-free medium containing VPA-EXO, EXO, and VPA was added to the lower chamber and an equal amount of serum-free medium containing PBS was added to the control group. Finally, 200 μL of cell suspension was added to the upper chamber, which was incubated for 20–24 h at 37 °C with 5% CO2, fixed in 4% tissue cell fixative for 20 min, and stained using 0.1% crystal violet for 5 min.
4.6.4. Tube-Forming Ability of HUVECs
The Ibidi angiogenesis slides were removed and 10 μL Matrigel was added to each well. After standing in the incubator for 30 min and waiting for the matrix gel to solidify, 50 μL of cell suspension with a concentration of 2 × 105 cells/mL was added to the upper well of the angiogenesis slides and images were collected at regular intervals in accordance with the growth rate of the cells. Angiogenesis was observed using a microscope. Then, 50 μL of Calcein AM solution at a concentration of 1 μM was added and the cells were incubated at 37 °C for 30 min away from light, washed with PBS, and observed using a fluorescence microscope for immunofluorescence imaging. The tubule lengths of each experimental group were statistically analyzed using Image J software to examine the effects of different drugs on the experimental results.
4.6.5. Examination of Cytokines in the Supernatants of HSFs and HUVECs
The levels of interleukin 1β (IL-1β), interleukin 8 (IL-8), tumor necrosis factor (TNF-α), matrix metalloproteinase 9 (MMP-9), and prostaglandin E2 (PG-E2) in the supernatant of HSF cells and vascular endothelial growth factor (VEGF) in the supernatant of HUVECs were detected using enzyme-linked immunosorbent assay (ELISA). The expression of relevant cytokine mRNA in HUVECs was detected using quantitative reverse transcription polymerase chain reaction (qRT-PCR). RNA was extracted from HUVECs and 15 μL of nuclease-free water was added to solubilize the RNA. Incubation was performed at 55 °C for 5 min and each sample was diluted to a concentration of 100–500 ng/μL. The total samples collected from the experimental group and the blank group were quantified using a microspectrophotometer. cDNA was quantified using a microspectrophotometer, the reaction system was configured, and the corresponding primers were designed according to the target genes, after which the corresponding PCR program was initiated. The expression of VEGF and PEDF in HUVECs was detected via Western blot, and the experimental procedure was the same as that described above.
4.6.6. In Vivo Study of VPA-EXO to Promote Skin Wound Healing
Forty-four male C57BL/6 mice aged 6–8 weeks were used to construct a whole-layer skin injury model and randomly divided into VPA-EXO, EXO, VPA, and PBS groups. The mice were acclimatized for 7 days. After fasting for 12 h before the experiment and anesthesia, the backs of the mice were shaved. The depilated area was about 2.5 cm × 2.5 cm, and one circular wound measuring 1 cm in diameter was constructed on the backs of all the mice to ensure adequate exfoliation from the skin, dermis, and muscularis propria. The date of model establishment was recorded as day 0. Each mouse was injected subcutaneously with 50 μL of VPA-EXO (EXO/VPA/PBS) into the four midpoints of the wound margin. Wounds were observed on postoperative days 0, 3, 7, 10, and 14 to assess wound healing. On the 14th postoperative day, the trauma tissues were sampled, and the skin tissues at the wound were excised, flash-frozen in liquid nitrogen, and stored at −80 °C. The frozen skin tissue was rapidly ground until no visible particles were present. The total RNA was extracted from the tissues and the expression levels of the related factors Col-І, IL-1β, TGF-β1, HIF-1α, Tie2, and Ang-1 in the skin tissues were detected using qRT-PCR following the method described above. The skin tissues of the wounds of mice on days 7 and 14 were taken and embedded, and then they were subjected to Hematoxylin and Eosin(H&E) and Masson staining. Wound skin tissues collected on day 14 were subjected to immunohistochemistry to detect the expression levels of CD31 and VEGF. The histologic changes of the wounds were observed under the microscope.
4.7. Statistical Analysis
One-way analysis of variance (ANOVA) was applied to the results obtained using GraphPad Prism 9 to analyze the differences between the groups, with p < 0.05 considered statistically significant, * indicating p < 0.05, ** indicating p < 0.01, and *** indicating p < 0.001.
5. Conclusions
Umbilical cord MSC exosomes can promote skin wound healing through inhibiting inflammation and promoting collagen production and angiogenesis. To study the therapeutic effect and mechanism of action of exosomes combined with VPA in wound healing, this study successfully loaded VPA into exosomes derived from HUC-MSCs. The VPA-EXO prepared using ultrasound technology exhibited the morphological characteristics of exosomes, with particle size mainly distributed from 30–150 nm and a membrane structure. In addition, the relative expression levels of CD9 and TSG101 proteins, which are characteristic surface markers of exosomes, were positive. When the mass ratio of VPA to exosome was 5:1, the drug-loading effect was the best and the encapsulation rate could reach 11.54%.
In vitro investigations showed that fibroblasts and vascular endothelial cells were able to uptake PKH26-labeled VPA-EXO, and in vitro cellular experiments (including cell proliferation and migration assays and tubule formation) demonstrated that VPA-EXO effectively promoted the proliferation, migration, and angiogenic effects of HSFs and HUVECs. ELISA and qRT-PCR were performed on a number of cytokines, the expression of inflammatory regulatory factors, and angiogenesis-related factors, demonstrating that VPA-EXO could significantly inhibit the expression of MMP-9, IL-1β, IL-8, TNF-α, and PG-E2 and promote the expression of VEGFs. In addition, VPA-EXO significantly inhibited the expression of inflammation-related cytokines IL-1β and TNF-α mRNA and induced the up-regulated expression of Ang-1, Tie2, VEGFR-2, VEGFA, and HIF-1α mRNA in HUVECs. VPA-EXO promoted the expression of VEGF protein and inhibited the expression of PEDF protein. These findings suggest that VPA-EXO regulates wound healing through inhibiting inflammation and enhancing angiogenesis, thus promoting wound healing.
In a mouse skin trauma model, VPA-EXO significantly reduced the inflammatory manifestations of skin wounds, increased the wound healing rate, reduced the inflammatory factors in skin tissues, significantly increased the collagen content, accelerated angiogenesis and the re-epithelialization of wounds, thereby shortening the time of wound repair, and accelerated the recovery of the skin structure. These results were consistent with the in vitro experiments and demonstrated that VPA-EXO treatment accelerated skin healing in mice. VPA-EXO shows potential for utilization in wound healing and vascular regeneration, and the mechanism of action of drug-loaded exosomes in promoting wound healing and vascular regeneration can be further investigated to expand their therapeutic role in wound healing.