Next Article in Journal
A Holistic Approach to Hard-to-Treat Cancers: The Future of Immunotherapy for Glioblastoma, Triple Negative Breast Cancer, and Advanced Prostate Cancer
Next Article in Special Issue
Cellular and Molecular Processes in Wound Healing
Previous Article in Journal
Induction of Periodontitis Using Bacterial Strains Isolated from the Human Oral Microbiome in an Experimental Rat Model
Previous Article in Special Issue
Laser in the Treatment of Atonic Wounds
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Advancements and Insights in Exosome-Based Therapies for Wound Healing: A Comprehensive Systematic Review (2018–June 2023)

1
Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, No. 228, 4050-313 Porto, Portugal
2
Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal
3
Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 1300-477 Lisboa, Portugal
4
Instituto Universitário de Ciências da Saúde (CESPU), Avenida Central de Gandra 1317, 4585-116 Paredes, Portugal
5
Centre for Rapid and Sustainable Product Development, Polytechnic of Leiria, 2430-028 Marinha Grande, Portugal
6
Department of Clinical and Biological Sciences, Cavalieri Ottolenghi Neuroscience Institute, University of Turin, Ospedale San Luigi, 10043 Turin, Italy
*
Author to whom correspondence should be addressed.
Biomedicines 2023, 11(8), 2099; https://doi.org/10.3390/biomedicines11082099
Submission received: 25 June 2023 / Revised: 14 July 2023 / Accepted: 22 July 2023 / Published: 25 July 2023
(This article belongs to the Special Issue Basic and Clinical Research in Wound Healing)

Abstract

:
Exosomes have shown promising potential as a therapeutic approach for wound healing. Nevertheless, the translation from experimental studies to commercially available treatments is still lacking. To assess the current state of research in this field, a systematic review was performed involving studies conducted and published over the past five years. A PubMed search was performed for English-language, full-text available papers published from 2018 to June 2023, focusing on exosomes derived from mammalian sources and their application in wound healing, particularly those involving in vivo assays. Out of 531 results, 148 papers were selected for analysis. The findings revealed that exosome-based treatments improve wound healing by increasing angiogenesis, reepithelization, collagen deposition, and decreasing scar formation. Furthermore, there was significant variability in terms of cell sources and types, biomaterials, and administration routes under investigation, indicating the need for further research in this field. Additionally, a comparative examination encompassing diverse cellular origins, types, administration pathways, or biomaterials is imperative. Furthermore, the predominance of rodent-based animal models raises concerns, as there have been limited advancements towards more complex in vivo models and scale-up assays. These constraints underscore the substantial efforts that remain necessary before attaining commercially viable and extensively applicable therapeutic approaches using exosomes.

Graphical Abstract

1. Introduction

The skin serves as the body’s external protection against harmful agents, regulating the internal temperature and integrity while maintaining homeostasis. Under normal conditions, the skin can regenerate itself through a complex process that comprises four distinct phases: hemostasis, inflammation, proliferation, and remodeling. However, when this process fails or is disrupted, it can culminate in impaired tissue regeneration or prolonged wound healing, leading to the formation of chronic wounds [1,2,3].
Chronic wounds are characterized by a prolonged inflammation (lasting from 4 to 12 weeks), often associated with infections, microbial biofilms, and impaired response from epithelial cells [4]. These wounds are multifactorial and frequently occur in individuals with several diseases, including diabetes, infections, and arterial/venous insufficiency [5].
The prevalence of chronic non-healing wounds is increasing due to factors such as population aging and aging-associated diseases, concomitant diseases, tumors, and congenital defects, negatively impacting the quality of life of millions of people worldwide. Therefore, the socioeconomical and health care burden is also increasing [1,2,3].
Current therapies such as debridement, antibiotherapy, and dressings remain insufficient, as they are not efficient, and there is still a need for new treatments. In the last few years, regenerative medicine has gained popularity, and extensive research has been conducted on mesenchymal stem cells (MSCs) and their derivates in several fields, including wound healing and skin regeneration.
MSCs are defined by the International Society of Cellular Therapy as similar to fibroblasts, adherent to plastic, and with the ability to differentiate into three different cell lines in vitro (chondrocytes, osteoblasts, and adipocytes). These cells should express the surface markers CD73, CD90, and CD105 while not expressing hematopoietic markers (CD14, CD45, CD34, CD19/HLA-DR, and CD11b/CD79). These undifferentiated cells have the potential to repair different tissues as they undergo differentiation. Furthermore, MSCs can be obtained from different sources and species. In recent years, there has been a notable rise in the use of MSCs for wound-healing purposes, with numerous studies showcasing promising outcomes utilizing cells obtained from diverse sources [1,2].
The use of MSC-derived products, such as secretome and exosomes, when compared to MSCs offers some advantages, including a reduced risk of tumorigenesis and minimal immune rejection [4], which is steadily rising. Exosomes are nanovesicles secreted from the endosomal system (30–150 nm) and represent one of the three major subpopulations of extracellular vesicles [6]. The other subpopulations include apoptotic bodies (>100 nm) and microvesicles (100–1000 nm). Exosomes are produced by several different cell types from different origins [7].
Previous research has highlighted the crucial role of exosomes in facilitating cell-to-cell communication, namely by sharing their cargo as miRNAs and proteins. Several studies have indicated that exosomes obtained from stem cells have the potential to assist with and promote tissue repair. This is attributed to their unique advantages, including exceptional stability, minimal risk of immune rejection, targeted delivery to specific tissues, straightforward control of dosage, and definable concentration [8].
Several studies have shown that exosomes derived from MSCs have similar therapeutic properties, angiogenic ability, and immune modulation as the cells from which they originate [3,9,10,11].
The quality of wound healing relies on the fibroblast’s migration and proliferation as well as collagen synthesis and deposition. Exosomes, with their abundant content of RNAs and proteins relevant to fibroblast functions, are thought to facilitate these processes. This optimization of fibroblast activities ultimately contributes to the accelerated wound-healing mechanism [8].
Multiple studies have demonstrated the therapeutic potential of exosomes in various stages of wound healing. In the inflammation phase, exosomes have been shown to modulate immune cells and resident tissue cells, leading to a reduction in uncontrolled inflammatory responses. During the proliferation phase, exosomes play a role in wound closure by activating endothelial cells and fibroblasts. This activation promotes a proangiogenic environment and initiates the deposition of extracellular matrix. In the remodeling phase, exosomes influence the balance between matrix metalloproteinases and tissue inhibitors of matrix metalloproteinases, favoring optimal wound-healing outcomes. Exosome therapy also enhances wound healing by stabilizing and stimulating a wide range of mediators involved in each phase [10].
The aim of this systematic review was to access the exosome-based therapies wound-healing effects and methodological heterogeneity in studies and furnish the scientific community with a comprehensive overview of the advancements made in this field over the past five years. Additionally, the review encompasses an analysis of the animal models used to evaluate the translational potential of the data to human medicine.

2. Data and Methods

This systematic review was performed according to PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-Analyses) guidelines.
The main goal of this study was to assess the methodology heterogeneity and provide insights into the progress of exosome research in wound healing over the past five years.
The research involved using the PubMed database, covering the period from 2018 to June 2023. The search query used the following keywords: “wound healing” (Title/Abstract) AND “exosomes” (Title/Abstract) NOT “review” (Title/Abstract), which initially retrieved 531 results.
All 531 publications underwent title, abstract, and full-text article examination.
The eligibility criteria included: (1) English language, (2) full access to the publication, (3) use of exosomes from mammalian sources, (4) exosomes applications in wound healing, and (5) use of animal models (in vivo studies).
Exclusion criteria were applied to filter out the studies that did not meet the research goals, such as: (1) in vitro studies, (2) review articles, (3) non-English-language publication, (4) studies unrelated to wound healing, (5) no full access to the publication, and (6) studies not involving exosomes.
To ensure rigorous study selection, all authors participated in the process and conducted double checks. Any discrepancies or disagreements were resolved through discussion and consensus. Duplicates were searched through Endnote software, and data analysis was performed using an Excel form specifically designed by the authors.
Among the initial 531 studies, 383 were excluded based on the exclusion criteria mentioned above, resulting in 148 papers. These remaining papers underwent a thorough double check by all authors.
The extracted information from the selected studies included PMID (PubMed Identification), paper title, publication date, corresponding author’s country, cell species, cell type, biomaterial usage, administration route, animal models, and exclusion criteria (when applicable).
GraphPad Prism version 8.0.1 was used to elaborate the graphical representations of the collected data.
Bias assessment was evaluated for each study regarding the adherence to MSCs minimal criteria, and animal models were examined to determine external validity.
The selection process is summarized in Figure 1.
Through this systematic review, the study aimed to provide valuable insights and contribute to the understanding of exosome-derived treatments in wound healing, with potential implications for potential translation into human medicine.

3. Results

3.1. Retrieved Data

The following table summarizes the retrieved data from the selected 148 papers (Table 1).

3.2. Scientific Data Production and Publication Distribution between 2018 and June 2023

All 148 papers selected were comprehensively analyzed to assess the temporal distribution of their publication across the five-year timeframe (Figure 2). In 2018, a total of seven papers were published (4.7%). The subsequent year, 2019, witnessed a notable increase in publications, with 16 papers, accounting for 10.8% of the selection. The publication rate continued to rise in 2020, reaching 30 papers (20.3%). In 2021, 29 papers were published (19.6%). The most significant publication rate occurred in 2022, with 42 papers published (28.4%). Up until June 2023, 24 additional papers had already been published, indicating that the year will probably surpass previous records (16.2%). These data demonstrate that there has been a continuous and stable increase in scientific investment in this research area, with the number of works carried out and published results increasing continuously.
The geographical distribution of scientific publications in the field of exosome application in wound healing was examined, focusing on the corresponding author’s country over a 5-year timeframe (Figure 3). China emerged as the country with the highest scientific publication rate in this field, with 122 publications (82.4%). The United States of America (USA) followed with eight publications (5.4%), South Korea with five publications (3.4%) and Portugal with three publications (2.0%). Other countries have lower publication rates: Japan recorded two publications (1.4%), as did Iran (1.4%) and India (1.4%). Similarly, Egypt and Taiwan each had one publication (0.7%). Additionally, a collaboration between China and Finland resulted in two publications (1.4%).

3.3. Cell Source and Type

Cell source was analyzed as seen in Figure 4. Among the papers analyzed, human tissues were the preferred cell source for exosome extraction (73.6%). Then, rodent-derived cells corresponded to 23%, with 15.5% from mice and 7.4% from rat. The other sources, although less frequent, consisted of dog (0.7%) and macaque (0.7%). Furthermore, one study compared mice- and rat-derived exosomes (0.7%), while two independent studies compared both mice- and human-derived exosomes (1.4%).
According to the data presented in Table 2, the most commonly used to extract exosomes in humans is the adipose tissue (ADSC) with 26.6%, followed by the UCMSCs (umbilical cord mesenchymal stem cells) with 22.9%, and the BMSCs (bone-marrow-derived mesenchymal stem cells) with 10.1%. Other tissues still present considerable percentages of use, such as placenta (2.8%), peripheral blood (3.7%), epidermal (3.7%), DP (dental-pulp-derived mesenchymal stem cells) (2.8%), and umbilical vein (7.3%). Regarding mice, adipose tissue emerges as the preferred source (30.4%), followed by bone marrow (26.1%). In rats, both adipose tissue and bone marrow (36.4%) are favored as primary sources for exosome extraction. These results are in agreement with the general scientific literature related to the use of cell-based therapies, where adipose tissue, bone marrow, and umbilical cord are the most explored tissues, and the respective MSCs are the most studied and characterized cells both for their direct use and use of their secretion products.

3.4. Biomaterials and Administration Route

As seen in Figure 5, the analysis of biomaterials used in all 148 papers revealed that the majority of studies chose to use exosomes without any biomaterial (73.6%). However, when a biomaterial was selected, hydrogels were the most commonly used (18.2%). Other biomaterials were also employed in several studies, such as scaffolds (2.7%), patches (1.4%), sponges (2.0%), nanoparticles (0.7%), and dressings (1.4%).
In the analysis of these 148 papers, the administration route of exosomes was analyzed and compared to the selected biomaterials, as illustrated in Figure 6. The preferred method, regardless of the presence of biomaterials, was via subcutaneous (SC) injection at the wound margins (66.2%). Within this route, subcutaneous injection without a biomaterial (57.8%) was the most common approach, while the association with a biomaterial was only 8.8%.
Topical administration was used 25.7%, with 16.9% involving the use of a biomaterial and 8.8% without one. Other administration routes included intradermal (ID) injection (2.7%), intraperitoneal (IP) injection (1.4%), and endovenous (EV) injection (2.0%). There were also two papers that compared topical and subcutaneous injections (1.4%), while one study compared topical and intradermal injections (0.7%).

3.5. Animal Models

Among the studies included in the analysis, rodents were used in the majority, accounting for 96.6% of the total (143 studies), with mice comprising 66.9% (99 studies) and rats 29.7% (44 studies). Also, two studies used both mice and rats (1.4%), while one study used a non-human primate model (0.7%), and the last used a canine model (0.7%). Figure 7 provides a visual representation of the distribution of in vivo models used in the selected studies.

4. Discussion

4.1. Scientific Data Production and Publication Distribution between 2018 and June 2023

The publication rate regarding the use of exosomes for wound healing has shown a significant increase in the last five years, with 2022 marking the highest publication rate to date. Based on the count of 24 publications as of June, 2023 is expected to surpass previous records.
This notable increase in scientific publications reflects the recent emergence and promising outcomes of exosome-based therapies in wound healing. It is expected that even more valuable data will be published in the next years.
In addition, China stands out as the leading country regarding publication rate, which demonstrates the high importance this topic in this country. However, there remains a gap in research development and publication in this field in other countries, particularly in Europe and America. It is essential to encourage research and publication in these regions in order to promote advancements in the field of exosome-based therapy worldwide.

4.2. Cell Source and Type

Among the preferred tissue source for exosome production, human-derived cells accounted for 73.6% of the studies, followed by rodent-derived cells at 23%, with 15.5% from mice and 7.4% from rat.
Within the human-derived cells, the most commonly used tissue is the adipose tissue (26.6%), followed by the umbilical cord (22.9%) and the bone marrow (10.1%). In mice, adipose tissue emerges as the preferred source (30.4%), followed by bone marrow (26.1%). In rats, both adipose tissue and bone marrow (36.4%) are favored as primary sources for exosome extraction.
Given that the ultimate objective of most studies is the development of exosome-based therapies for human medicine, the retrieval of exosomes from human tissues appears to be a logical approach. However, the significant heterogeneity within the tissues poses challenges in comparing results, as researchers have not reached an understanding of the most efficacious treatment option.
The preferred use of ADSCs (adipose-tissue-derived mesenchymal stem cells) is probably due to its low ethical issues, easy extraction, and cost-effectiveness. ADSCs have shown potential in wound healing by increasing vascularization, fibroblasts migration, and differentiation and upregulating macrophages chemotaxis [2,160,161].
BMSCs have also demonstrated great potential in wound healing, increasing angiogenesis, and reducing wound contraction [161]. Additionally, BM-MSCs have garnered significant attention as the most extensively investigated subset of MSCs and have been recognized for their relatively low immunogenicity [2].
UCMSCs have also shown their wound-healing potential, as they can differentiate into epidermal tissue and are easier to harvest than BM-MSCs [1,2].
The variation observed in exosomes derived from different species and tissues still needs further research and understanding. The goal should be to identify the most effective sources and optimize and standardize the isolation processes to ensure consistent and reliable outcomes. By doing so, the scientific community can more accurately make comparisons between studies and advance towards the development of efficacious therapeutic approaches.
In addition, it is valuable to explore alternative sources of exosomes beyond those currently described, as it may uncover potential benefits and characteristics, broadening the range of therapeutic options.
Overall, while human-derived exosomes remain the preferred choice due to their ultimate clinical relevance, efforts should be made to refine the current methodologies and promote collaborations between research groups to better understand the most effective exosome-based treatments.

4.3. Biomaterials and Administration Route

In most studies, exosomes were administered via SC injection in the wound margins, either without a biomaterial (57.8%) or in combination with a biomaterial (8.8%). This delivery method offers ease, speed, and localized treatment administration. Considering that most skin wounds are created on the animal’s dorsum, incorporating biomaterials can be difficult, requiring prior development and testing. SC injection of MSCs has also demonstrated great results regarding wound closure, angiogenesis, and re-epithelization. Alternatively, topical administration is also used (25.7%), as it is less invasive and less painful than the injection methods [162].
However, when a biomaterial was selected, it was predominantly a hydrogel (18.2%). Other biomaterials were also applied in several studies, including scaffolds (2.7%), patches (1.4%), sponges (2.0%), nanoparticles (0.7%), and dressings (1.4%).
The combination of biomaterials aims to improve the therapeutic functionality of exosomes by stabilizing them and prolonging their release at the wound site, thereby preventing rapid entry into blood circulation and systemic dilution. Hydrogels, specifically, offer several advantages in wound healing, such as antibacterial activity, facilitation of tissue adhesion, protection against UV radiation, hemostatic capacity, promotion of spontaneous regeneration, and easy injectability. They can also provide a 3D environment and mimic the extracellular matrix while maintaining proper moister levels at the wound site. Therefore, the use of exosomes associated with hydrogels has shown to improve wound healing, enhancing re-epithelization and vascularization [4,163].
Hydrogels based on chitosan or methylcellulose are considered great options for diabetic wound treatment and have been used in some of the selected studies. These polymers have good biodegradability and biocompatibility and are nontoxic. Geng et al. developed a loaded carboxyethyl chitosan hydrogel loaded with bone-marrow-derived exosomes to improve chronic diabetic wound healing. It increased angiogenesis and neovascularization, reduced local inflammation, and improved wound healing in diabetic rats [27].
Pluronic F-12 hydrogels have also been used in selected studies, as they are injectable, biocompatible, and thermosensitive. Zhou et al. used a pluronic F-12 hydrogel combined with adipose-tissue-derived exosomes to improve re-epithelization, angiogenesis, collagen synthesis, and wound healing and cellular proliferation in mice [17]. Yang et al. also used a similar hydrogel combined with human umbilical-cord-derived exosomes with increased wound-closure rate and granulation tissue in rats [13].
Gelatin methacryloyl (GelMA) hydrogels were also chosen in different studies due to their mechanical properties and ability to retain exosomes for a prolonged time. Zhao et al. used human umbilical vein endothelial cells (HUVECs)-derived exosomes in association with a GelMA hydrogel and demonstrated an improvement of angiogenesis and collagen maturity in rats [56]. Hu et al. used a similar hydrogel with ADSCs-derived exosomes and showed an improvement of wound healing with increased blood vessel regeneration, proliferation, and migration in mice [43].
Several studies have combined hydrogels and MSCs with promising results in skin regeneration. The use of BMSCs seeded into hydrogels improved angiogenesis and accelerated wound healing in mice [164]. Another study demonstrated reduced scar formation and improved angiogenesis, collagen, granulation, and re-epithelization in rabbits through ADSCs combined with a hydrogel [165].
These findings emphasize the importance of carefully selecting the biomaterials and administration route to optimize the therapeutic effects of exosomes in wound healing.

4.4. Animal Models

The results showed that rodents are the main animal models in studies involving exosome-based therapies in wound healing (96.6%). These findings are consistent with Al-Masawa et al.’s previous findings up until March 2021 [3].
The use of small-animal models has several advantages, such as researchers’ familiarity, easy handling, affordability, and availability. However, there are also limitations associated with these models, including skin thickness, fast hair growth cycles, follicular pattern, and wound size [166]. Rodents exhibit a thin epidermis and loose skin adherence along with dense hair that has been suggested to potentially enhance the wound-healing rate. In addition, these animals lack apocrine and eccrine glands but possess a subcutaneous panniculus carnosus muscle that enhances rapid wound contraction. Moreover, they also have stronger immune systems and have endogenous sources of vitamin C, which plays a significant role in wound healing [167,168].
Although rodent models are frequently used in the initial stages of new therapy approaches, it is necessary to scale up to more complex animal models to better reflect the similarities between such models and the human species. The main goal of most researchers is to develop new treatments options for non-healing chronic wounds and make them commercially available to the human population. Therefore, the consistent use of rodents in 96.6% of studies over the last five years limits the broader application of these data.
Although the use of exosome-based therapies has been showing promising results over last few years, the inclusion of larger animal models, such as ovine, swine, dog, and non-human primates, is crucial. In addition, it is important to fulfill the 3Rs principle (replace, reduce, and refine) regarding animal use, implying that research data should evolve until commercialization becomes possible [169,170].
However, these more complex models present challenges, as they are more expensive, more difficult to handle, and require large set-ups. Pigs, for instance, are regarded as standard models for wound-healing research due to the resemblance of their skin to that of humans. They also present physiological and anatomical similarity to the human species. Nonetheless, to date, no studies have been conducted on this particular species. Non-human primates, although sharing greater similarity with humans, are rarely used mainly due to ethical concerns [166,167].
The porcine model has been used in wound-healing research with promising results. In particular, the administration of BMSC and ADSC intradermally into partial-thickness wounds enhanced local epithelization and improved wound appearance when compared to the control [171,172]. This suggests that the use of these cells can accelerate the wound-healing process.
Martinello et al. used the ovine model in wound healing and achieved great results. The local injection of peripheral-blood MSCs into the wound margins revealed improved re-epithelization, proliferation, neovascularization, and contraction, with a higher wound-closer rate [173].
In dogs, the use of MSCs to treat chronic wounds has also demonstrated great potential. UCMSCs used in association with a PVA hydrogel showed significant progress in wound regeneration and decreased local ulceration [174]. Other study using ADSc also improved re-epithelization, reduced local inflammation, and promoted epidermal and dermal regeneration in both acute and chronic wounds [175]. In the selected study using a dog model, Bahr et al. used BM-MSCs-derived exosomes in association with a carboxymethylcellulose hydrogel. The results were promising, as the treatment enhanced wound healing with no scaring, with organized collagen deposition and increased dermal fibroblasts [158].
Lu et al. used autologous and allogeneic iPSCs-derived exosomes to improve wound healing in macaques. It demonstrated an increased angiogenesis, collagen deposition, epithelial coverage, and wound-closure rate [56].
While acknowledging the differences among animal models, current approaches in wound healing remain highly relevant. However, it is crucial to increase the use of diverse and more complex animal models to bridge the gap between current findings and their practical application in the human species [176].
All the selected papers consistently reported positive outcomes in vivo, highlighting the ability of exosomes to promote wound healing.
Exosome treatment was found to enhance wound-closure rates, stimulate local angiogenesis and reepithelization, and facilitate collagen deposition [12,14,17,18]. Furthermore, exosomes promoted a reduction in scar formation and decreased local inflammation in multiple studies [18,23,158]. Additionally, exosome treatments resulted in increased granulation tissue formation and enhanced the proliferation and migration of dermal fibroblasts [65,155,157].
The findings consistently indicate that exosomes possess therapeutic properties and contribute to the healing of skin wounds. Importantly, these beneficial effects remain consistent across various experimental animal models, methods of administration, exosome concentrations, number of administrations, and sources of exosomes.
Some meta-analyses have been published with positive outcomes that corroborate these findings. Qiao et al. and Masawa et al. demonstrated that exosome-based therapies improve angiogenesis, reepithelization, and collagen deposition while decreasing local inflammation. Therefore, the results indicate that the treatments accelerate wound healing [3,177].

5. Conclusions

Addressing the need for effective therapeutic options to promote skin regeneration remains a significant goal, as it poses an ongoing challenge to public health. This challenge is expected to intensify with the increasing population suffering from chronic diseases and the general aging of the population associated with an increase in average life expectancy. As a potential biological therapeutic approach, exosome-based therapies emerge as a promising strategy for wound healing.
This comprehensive systematic review highlights the great potential of exosomes as therapeutic options for non-healing chronic wounds. In summary, exosome treatment has shown consistent positive outcomes, including enhanced wound-closure rates, stimulation of local angiogenesis and reepithelization, and increased collagen deposition. Moreover, exosomes have demonstrated the ability to reduce scar formation, alleviate local inflammation, promote increased granulation tissue formation, and enhance the proliferation and migration of dermal fibroblasts. These findings underscore the therapeutic efficacy of exosomes in promoting wound healing. The field has also witnessed significant advancements in the last 5 years by combining exosomes with innovative engineering strategies. Exosome-based therapies have emerged as promising tools for wound healing, with advantages such as abundant sources; ease of preparation, storage, and transportation; as well as minimal immunogenicity.
Despite the potential of exosomes in wound treatment, challenges persist. The methodology associated with the use of exosomes-based therapies in wound healing remains highly heterogeneous. The mechanisms underlying exosome biogenesis, cellular interactions, and communication remain largely unknown. Comprehensive research is necessary to deepen our understanding of exosome biology for safe and effective applications. Standardized preparation methods and technical issues pose obstacles that impact the pharmacokinetic and pharmacodynamic performance of exosomes. Precise cargo loading, large-scale production, targeted delivery, reduced contamination, and cytotoxicity all require further exploration. Nonetheless, standardization and optimization of exosomes isolation and purification methods, scalability for clinical applications, and enhancing therapeutic potential through tissue engineering strategies are important challenges to address.
Further research endeavors are imperative to facilitate the commercial availability and clinical application of these treatments.
The considerable variability in cell sources, types, biomaterials, and administration routes under investigation shows the urgent need of further research in this field. Moreover, the lack of comparative studies exploring different cell sources/types, administration routes, or even biomaterials is a critical gap that must be addressed. Furthermore, the predominant use of rodent-based animal models raises concerns, as limited progress has been made in advancing toward more complex in vivo models that closely resemble human physiology.
This study also has certain limitations, primarily due to the potential bias associated with study design and methodology of the included studies. To address these limitations, it is crucial that future research incorporates measures to mitigate bias, such as randomization, blinding, and standardized protocols. Due to the absence of a universally agreed-upon set of techniques for isolating, characterizing, and applying exosomes, we were unable to strictly enforce inclusion and exclusion criteria in our study. Additionally, the scope of our study did not allow for a comprehensive comparison between various wound-healing models. It is worth noting that a bias may have been introduced during data collection and reporting due to the prevalent practice of publishing only successful or efficacious studies.
To achieve a commercially viable and widely accessible range of therapeutic options, several key objectives must be pursued in the future. Standardizing methodologies is paramount to ensure more reliable and comparable results. In addition, the inclusion of more complex animal models that closely mimic the human species will enable the effective translation of research outcomes. These collective efforts will drive the field closer to its ultimate goal of achieving large-scale production and widespread availability of exosome-based therapeutic option for wound healing.
To overcome the limitations associated with solely relying on rodent models, we suggest expanding our investigation to include a diverse range of models, including non-rodent and non-animal alternatives. Furthermore, it is crucial to extend the follow-up period to thoroughly evaluate the impact of exosomes on the maturation and scarring of healed skin as well as the immunological response. Additionally, future research endeavors should encompass various skin lesions, such as burns, ulcers, and incisional ischemic lesions, and utilize more representative models of diabetic type 2 and non-healing wounds.
In conclusion, our research findings support the hypothesis that exosomes have great potential as therapeutic options for wound-healing applications. However, it is crucial to establish a consensus regarding the definition and standardization of variables related to exosome isolation, quantification, administration, and reporting. Multidisciplinary research is crucial to address the scientific questions and technical challenges associated with exosomes and to bridge the gap between experimental studies and commercialization. Continued advancements in the field will pave the way for the translation of exosome-based therapies into clinically applicable approaches. With ongoing advancements in the field of exosome research and continued efforts in addressing these challenges, exosome-based therapies hold great promise for future clinical applications. Achieving unanimous agreement on these variables will undoubtedly facilitate the advancement of the exosome field and pave the way for a promising future, allowing its translation into clinical practice.

Author Contributions

Conceptualization, P.S., B.L., A.C.S., R.A., A.M., A.C., N.A., S.G. and A.C.M.; methodology, P.S., B.L., A.C.S., R.A., A.M., A.C., N.A., S.G. and A.C.M.; software, P.S., B.L., A.C.S., R.A., A.M., A.C., N.A., S.G. and A.C.M.; validation, P.S., B.L., A.C.S., R.A., A.M., A.C., N.A., S.G. and A.C.M.; formal analysis, P.S., B.L., A.C.S., R.A., A.M., A.C., N.A., S.G. and A.C.M.; investigation, P.S., B.L., A.C.S., R.A., A.M., A.C., N.A., S.G. and A.C.M.; resources, P.S., B.L., A.C.S., R.A., A.M., A.C., N.A., S.G. and A.C.M.; data curation, P.S., B.L., A.C.S., R.A., A.M., A.C., N.A., S.G. and A.C.M.; writing—original draft preparation, P.S., B.L., A.C.S., R.A., A.M., A.C., N.A., S.G. and A.C.M.; writing—review and editing, P.S., B.L., A.C.S., R.A., A.M., A.C., N.A., S.G. and A.C.M.; visualization, P.S., B.L., A.C.S., R.A., A.M., A.C., N.A., S.G. and A.C.M.; supervision, R.A., N.A. and A.C.M.; project administration, N.A. and A.C.M.; funding acquisition, N.A. and A.C.M. All authors have read and agreed to the published version of the manuscript.

Funding

Patrícia Sousa acknowledges University of Porto (UP) and Centro de Estudos de Ciêcia Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA) for the funding and availability of all resources needed for this work. Ana Catarina Sousa (SFRH/BD/146689/2019) and Bruna Lopes (2021.05265.BD) acknowledge Fundaçao para a Ciência e Tecnologia (FCT) for financial support. Rui Damásio Alvites acknowledges the Animal Science Studies Centre (CECA), Agroenvironment, Technologies and Sciences Institute (ICETA), Porto University (UP), and FCT for the funding and availability of all technical, structural, and human resources necessary for the development of this work. The work was supported through the project UIDB/00211/2020 funded by FCT/MCTES national funds. This research was funded by Projects PEst-OE/AGR/UI0211/2011 from FCT and COMPETE 2020, from ANI–Projetos ID&T Empresas em Copromoçao, and by the project “H2Cure—Desenvolvimento de formulações de géis e pensos de Mel, Goma Gelana e Ácido Hialurónico para tratamento de feridas”(POCI-01-0247-FEDER-047032).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Further data on the reported results are available from the corresponding author on request.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

ADSCAdipose-tissue-derived mesenchymal stem cells
BMSCBone-marrow-derived mesenchymal stem cells
DPsDental-pulp-derived mesenchymal stem cells
EVEndovenous
IDIntradermal
IPIntraperitoneal
iPSCsInduced pluripotent stem cells
MSCsMesenchymal stem cells
PMIDPubMed identification
PRISMAPreferred Reporting Items for Systematic Reviews and Meta-Analyses
SCSubcutaneous
UCMSCUmbilical cord mesenchymal stem cells
UVECUmbilical vein endothelial cells

References

  1. Carolina, M.; Bruna, L.; Patrícia, S.; Mariana, B.; Ana Catarina, S.; Ana Lúcia, L.; Rui, A.; Ana Colette, M. Application of Cell-Based Therapies in Veterinary Dermatology. In Wound Healing—Recent Advances and Future Opportunities; Ana Colette, M., Rui Damásio, A., Müzeyyen, G., Eds.; IntechOpen: Rijeka, Croatia, 2023; Chapter 1. [Google Scholar]
  2. Lopes, B.; Sousa, P.; Alvites, R.; Branquinho, M.; Sousa, A.; Mendonça, C.; Atayde, L.M.; Maurício, A.C. The Application of Mesenchymal Stem Cells on Wound Repair and Regeneration. Appl. Sci. 2021, 11, 3000. [Google Scholar] [CrossRef]
  3. Al-Masawa, M.E.; Alshawsh, M.A.; Ng, C.Y.; Ng, A.M.H.; Foo, J.B.; Vijakumaran, U.; Subramaniam, R.; Ghani, N.A.A.; Witwer, K.W.; Law, J.X. Efficacy and safety of small extracellular vesicle interventions in wound healing and skin regeneration: A systematic review and meta-analysis of animal studies. Theranostics 2022, 12, 6455–6508. [Google Scholar] [CrossRef] [PubMed]
  4. Zhao, H.; Li, Z.; Wang, Y.; Zhou, K.; Li, H.; Bi, S.; Wang, Y.; Wu, W.; Huang, Y.; Peng, B.; et al. Bioengineered MSC-derived exosomes in skin wound repair and regeneration. Front. Cell Dev. Biol. 2023, 11, 1029671. [Google Scholar] [CrossRef]
  5. Wu, J.; Chen, L.H.; Sun, S.Y.; Li, Y.; Ran, X.W. Mesenchymal stem cell-derived exosomes: The dawn of diabetic wound healing. World J. Diabetes 2022, 13, 1066–1095. [Google Scholar] [CrossRef] [PubMed]
  6. Bailey, A.J.M.; Li, H.; Kirkham, A.M.; Tieu, A.; Maganti, H.B.; Shorr, R.; Fergusson, D.A.; Lalu, M.M.; Elomazzen, H.; Allan, D.S. MSC-Derived Extracellular Vesicles to Heal Diabetic Wounds: A Systematic Review and Meta-Analysis of Preclinical Animal Studies. Stem Cell Rev. Rep. 2022, 18, 968–979. [Google Scholar] [CrossRef]
  7. Nikfarjam, S.; Rezaie, J.; Zolbanin, N.M.; Jafari, R. Mesenchymal stem cell derived-exosomes: A modern approach in translational medicine. J. Transl. Med. 2020, 18, 449. [Google Scholar] [CrossRef] [PubMed]
  8. Hu, L.; Wang, J.; Zhou, X.; Xiong, Z.; Zhao, J.; Yu, R.; Huang, F.; Zhang, H.; Chen, L. Exosomes derived from human adipose mensenchymal stem cells accelerates cutaneous wound healing via optimizing the characteristics of fibroblasts. Sci. Rep. 2016, 6, 32993. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. An, Y.; Lin, S.; Tan, X.; Zhu, S.; Nie, F.; Zhen, Y.; Gu, L.; Zhang, C.; Wang, B.; Wei, W.; et al. Exosomes from adipose-derived stem cells and application to skin wound healing. Cell Prolif. 2021, 54, e12993. [Google Scholar] [CrossRef]
  10. Prasai, A.; Jay, J.W.; Jupiter, D.; Wolf, S.E.; El Ayadi, A. Role of Exosomes in Dermal Wound Healing: A Systematic Review. J. Investig. Dermatol. 2022, 142, 662–678.e668. [Google Scholar] [CrossRef]
  11. Hu, J.C.; Zheng, C.X.; Sui, B.D.; Liu, W.J.; Jin, Y. Mesenchymal stem cell-derived exosomes: A novel and potential remedy for cutaneous wound healing and regeneration. World J. Stem Cells 2022, 14, 318–329. [Google Scholar] [CrossRef]
  12. Wang, C.; Wang, M.; Xu, T.; Zhang, X.; Lin, C.; Gao, W.; Xu, H.; Lei, B.; Mao, C. Engineering Bioactive Self-Healing Antibacterial Exosomes Hydrogel for Promoting Chronic Diabetic Wound Healing and Complete Skin Regeneration. Theranostics 2019, 9, 65–76. [Google Scholar] [CrossRef] [PubMed]
  13. Yang, J.; Chen, Z.; Pan, D.; Li, H.; Shen, J. Umbilical Cord-Derived Mesenchymal Stem Cell-Derived Exosomes Combined Pluronic F127 Hydrogel Promote Chronic Diabetic Wound Healing and Complete Skin Regeneration. Int. J. Nanomed. 2020, 15, 5911–5926. [Google Scholar] [CrossRef]
  14. Hu, Y.; Rao, S.S.; Wang, Z.X.; Cao, J.; Tan, Y.J.; Luo, J.; Li, H.M.; Zhang, W.S.; Chen, C.Y.; Xie, H. Exosomes from human umbilical cord blood accelerate cutaneous wound healing through miR-21-3p-mediated promotion of angiogenesis and fibroblast function. Theranostics 2018, 8, 169–184. [Google Scholar] [CrossRef] [PubMed]
  15. Liu, W.; Yu, M.; Xie, D.; Wang, L.; Ye, C.; Zhu, Q.; Liu, F.; Yang, L. Melatonin-stimulated MSC-derived exosomes improve diabetic wound healing through regulating macrophage M1 and M2 polarization by targeting the PTEN/AKT pathway. Stem Cell Res. Ther. 2020, 11, 259. [Google Scholar] [CrossRef]
  16. Yuan, M.; Liu, K.; Jiang, T.; Li, S.; Chen, J.; Wu, Z.; Li, W.; Tan, R.; Wei, W.; Yang, X.; et al. GelMA/PEGDA microneedles patch loaded with HUVECs-derived exosomes and Tazarotene promote diabetic wound healing. J. Nanobiotechnol. 2022, 20, 147. [Google Scholar] [CrossRef] [PubMed]
  17. Zhou, Y.; Zhang, X.L.; Lu, S.T.; Zhang, N.Y.; Zhang, H.J.; Zhang, J.; Zhang, J. Human adipose-derived mesenchymal stem cells-derived exosomes encapsulated in pluronic F127 hydrogel promote wound healing and regeneration. Stem Cell Res. Ther. 2022, 13, 407. [Google Scholar] [CrossRef]
  18. Wu, D.; Kang, L.; Tian, J.; Wu, Y.; Liu, J.; Li, Z.; Wu, X.; Huang, Y.; Gao, B.; Wang, H.; et al. Exosomes Derived from Bone Mesenchymal Stem Cells with the Stimulation of Fe3O4 Nanoparticles and Static Magnetic Field Enhance Wound Healing Through Upregulated miR-21-5p. Int. J. Nanomed. 2020, 15, 7979–7993. [Google Scholar] [CrossRef]
  19. Hu, Y.; Tao, R.; Chen, L.; Xiong, Y.; Xue, H.; Hu, L.; Yan, C.; Xie, X.; Lin, Z.; Panayi, A.C.; et al. Exosomes derived from pioglitazone-pretreated MSCs accelerate diabetic wound healing through enhancing angiogenesis. J. Nanobiotechnol. 2021, 19, 150. [Google Scholar] [CrossRef]
  20. Wang, J.; Wu, H.; Peng, Y.; Zhao, Y.; Qin, Y.; Zhang, Y.; Xiao, Z. Hypoxia adipose stem cell-derived exosomes promote high-quality healing of diabetic wound involves activation of PI3K/Akt pathways. J. Nanobiotechnol. 2021, 19, 202. [Google Scholar] [CrossRef]
  21. Li, X.; Xie, X.; Lian, W.; Shi, R.; Han, S.; Zhang, H.; Lu, L.; Li, M. Exosomes from adipose-derived stem cells overexpressing Nrf2 accelerate cutaneous wound healing by promoting vascularization in a diabetic foot ulcer rat model. Exp. Mol. Med. 2018, 50, 1–14. [Google Scholar] [CrossRef] [Green Version]
  22. Xiong, Y.; Chen, L.; Liu, P.; Yu, T.; Lin, C.; Yan, C.; Hu, Y.; Zhou, W.; Sun, Y.; Panayi, A.C.; et al. All-in-One: Multifunctional Hydrogel Accelerates Oxidative Diabetic Wound Healing through Timed-Release of Exosome and Fibroblast Growth Factor. Small 2022, 18, e2104229. [Google Scholar] [CrossRef] [PubMed]
  23. Wang, P.; Theocharidis, G.; Vlachos, I.S.; Kounas, K.; Lobao, A.; Shu, B.; Wu, B.; Xie, J.; Hu, Z.; Qi, S.; et al. Exosomes Derived from Epidermal Stem Cells Improve Diabetic Wound Healing. J. Investig. Dermatol. 2022, 142, 2508–2517.e2513. [Google Scholar] [CrossRef] [PubMed]
  24. Zhou, X.; Brown, B.A.; Siegel, A.P.; El Masry, M.S.; Zeng, X.; Song, W.; Das, A.; Khandelwal, P.; Clark, A.; Singh, K.; et al. Exosome-Mediated Crosstalk between Keratinocytes and Macrophages in Cutaneous Wound Healing. ACS Nano 2020, 14, 12732–12748. [Google Scholar] [CrossRef]
  25. Shi, R.; Jin, Y.; Zhao, S.; Yuan, H.; Shi, J.; Zhao, H. Hypoxic ADSC-derived exosomes enhance wound healing in diabetic mice via delivery of circ-Snhg11 and induction of M2-like macrophage polarization. Biomed. Pharmacother. 2022, 153, 113463. [Google Scholar] [CrossRef] [PubMed]
  26. Yu, M.; Liu, W.; Li, J.; Lu, J.; Lu, H.; Jia, W.; Liu, F. Exosomes derived from atorvastatin-pretreated MSC accelerate diabetic wound repair by enhancing angiogenesis via AKT/eNOS pathway. Stem Cell Res. Ther. 2020, 11, 350. [Google Scholar] [CrossRef]
  27. Geng, X.; Qi, Y.; Liu, X.; Shi, Y.; Li, H.; Zhao, L. A multifunctional antibacterial and self-healing hydrogel laden with bone marrow mesenchymal stem cell-derived exosomes for accelerating diabetic wound healing. Biomater. Adv. 2022, 133, 112613. [Google Scholar] [CrossRef]
  28. Xiong, Y.; Chen, L.; Yan, C.; Zhou, W.; Endo, Y.; Liu, J.; Hu, L.; Hu, Y.; Mi, B.; Liu, G. Circulating Exosomal miR-20b-5p Inhibition Restores Wnt9b Signaling and Reverses Diabetes-Associated Impaired Wound Healing. Small 2020, 16, e1904044. [Google Scholar] [CrossRef] [Green Version]
  29. Shiekh, P.A.; Singh, A.; Kumar, A. Exosome laden oxygen releasing antioxidant and antibacterial cryogel wound dressing OxOBand alleviate diabetic and infectious wound healing. Biomaterials 2020, 249, 120020. [Google Scholar] [CrossRef]
  30. Xie, Y.; Yu, L.; Cheng, Z.; Peng, Y.; Cao, Z.; Chen, B.; Duan, Y.; Wang, Y. SHED-derived exosomes promote LPS-induced wound healing with less itching by stimulating macrophage autophagy. J. Nanobiotechnol. 2022, 20, 239. [Google Scholar] [CrossRef]
  31. Wang, M.; Wang, C.; Chen, M.; Xi, Y.; Cheng, W.; Mao, C.; Xu, T.; Zhang, X.; Lin, C.; Gao, W.; et al. Efficient Angiogenesis-Based Diabetic Wound Healing/Skin Reconstruction through Bioactive Antibacterial Adhesive Ultraviolet Shielding Nanodressing with Exosome Release. ACS Nano 2019, 13, 10279–10293. [Google Scholar] [CrossRef]
  32. Zhang, Y.; Pan, Y.; Liu, Y.; Li, X.; Tang, L.; Duan, M.; Li, J.; Zhang, G. Exosomes derived from human umbilical cord blood mesenchymal stem cells stimulate regenerative wound healing via transforming growth factor-β receptor inhibition. Stem Cell Res. Ther. 2021, 12, 434. [Google Scholar] [CrossRef]
  33. He, X.; Dong, Z.; Cao, Y.; Wang, H.; Liu, S.; Liao, L.; Jin, Y.; Yuan, L.; Li, B. MSC-Derived Exosome Promotes M2 Polarization and Enhances Cutaneous Wound Healing. Stem Cells Int. 2019, 2019, 7132708. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Kwak, G.; Cheng, J.; Kim, H.; Song, S.; Lee, S.J.; Yang, Y.; Jeong, J.H.; Lee, J.E.; Messersmith, P.B.; Kim, S.H. Sustained Exosome-Guided Macrophage Polarization Using Hydrolytically Degradable PEG Hydrogels for Cutaneous Wound Healing: Identification of Key Proteins and MiRNAs, and Sustained Release Formulation. Small 2022, 18, e2200060. [Google Scholar] [CrossRef] [PubMed]
  35. Shi, R.; Jin, Y.; Hu, W.; Lian, W.; Cao, C.; Han, S.; Zhao, S.; Yuan, H.; Yang, X.; Shi, J.; et al. Exosomes derived from mmu_circ_0000250-modified adipose-derived mesenchymal stem cells promote wound healing in diabetic mice by inducing miR-128-3p/SIRT1-mediated autophagy. Am. J. Physiol. Cell Physiol. 2020, 318, C848–C856. [Google Scholar] [CrossRef] [PubMed]
  36. Qiu, X.; Liu, J.; Zheng, C.; Su, Y.; Bao, L.; Zhu, B.; Liu, S.; Wang, L.; Wang, X.; Wang, Y.; et al. Exosomes released from educated mesenchymal stem cells accelerate cutaneous wound healing via promoting angiogenesis. Cell Prolif. 2020, 53, e12830. [Google Scholar] [CrossRef]
  37. Qian, L.; Pi, L.; Fang, B.R.; Meng, X.X. Adipose mesenchymal stem cell-derived exosomes accelerate skin wound healing via the lncRNA H19/miR-19b/SOX9 axis. Lab. Investig. 2021, 101, 1254–1266. [Google Scholar] [CrossRef] [PubMed]
  38. Wang, Y.; Cao, Z.; Wei, Q.; Ma, K.; Hu, W.; Huang, Q.; Su, J.; Li, H.; Zhang, C.; Fu, X. VH298-loaded extracellular vesicles released from gelatin methacryloyl hydrogel facilitate diabetic wound healing by HIF-1α-mediated enhancement of angiogenesis. Acta Biomater. 2022, 147, 342–355. [Google Scholar] [CrossRef]
  39. Heo, J.S. Selenium-Stimulated Exosomes Enhance Wound Healing by Modulating Inflammation and Angiogenesis. Int. J. Mol. Sci. 2022, 23, 11543. [Google Scholar] [CrossRef]
  40. Ren, S.; Chen, J.; Guo, J.; Liu, Y.; Xiong, H.; Jing, B.; Yang, X.; Li, G.; Kang, Y.; Wang, C.; et al. Exosomes from Adipose Stem Cells Promote Diabetic Wound Healing through the eHSP90/LRP1/AKT Axis. Cells 2022, 11, 3229. [Google Scholar] [CrossRef]
  41. Kim, H.; Wang, S.Y.; Kwak, G.; Yang, Y.; Kwon, I.C.; Kim, S.H. Exosome-Guided Phenotypic Switch of M1 to M2 Macrophages for Cutaneous Wound Healing. Adv. Sci. 2019, 6, 1900513. [Google Scholar] [CrossRef] [Green Version]
  42. Li, B.; Luan, S.; Chen, J.; Zhou, Y.; Wang, T.; Li, Z.; Fu, Y.; Zhai, A.; Bi, C. The MSC-Derived Exosomal lncRNA H19 Promotes Wound Healing in Diabetic Foot Ulcers by Upregulating PTEN via MicroRNA-152-3p. Mol. Ther. Nucleic Acids 2020, 19, 814–826. [Google Scholar] [CrossRef] [PubMed]
  43. Hu, N.; Cai, Z.; Jiang, X.; Wang, C.; Tang, T.; Xu, T.; Chen, H.; Li, X.; Du, X.; Cui, W. Hypoxia-pretreated ADSC-derived exosome-embedded hydrogels promote angiogenesis and accelerate diabetic wound healing. Acta Biomater. 2023, 157, 175–186. [Google Scholar] [CrossRef] [PubMed]
  44. Xiao, S.; Xiao, C.; Miao, Y.; Wang, J.; Chen, R.; Fan, Z.; Hu, Z. Human acellular amniotic membrane incorporating exosomes from adipose-derived mesenchymal stem cells promotes diabetic wound healing. Stem Cell Res. Ther. 2021, 12, 255. [Google Scholar] [CrossRef] [PubMed]
  45. Chen, L.; Qin, L.; Chen, C.; Hu, Q.; Wang, J.; Shen, J. Serum exosomes accelerate diabetic wound healing by promoting angiogenesis and ECM formation. Cell Biol. Int. 2021, 45, 1976–1985. [Google Scholar] [CrossRef]
  46. Xia, W.; Li, M.; Jiang, X.; Huang, X.; Gu, S.; Ye, J.; Zhu, L.; Hou, M.; Zan, T. Young fibroblast-derived exosomal microRNA-125b transfers beneficial effects on aged cutaneous wound healing. J. Nanobiotechnol. 2022, 20, 144. [Google Scholar] [CrossRef]
  47. Zhang, W.; Bai, X.; Zhao, B.; Li, Y.; Zhang, Y.; Li, Z.; Wang, X.; Luo, L.; Han, F.; Zhang, J.; et al. Cell-free therapy based on adipose tissue stem cell-derived exosomes promotes wound healing via the PI3K/Akt signaling pathway. Exp. Cell Res. 2018, 370, 333–342. [Google Scholar] [CrossRef] [PubMed]
  48. Ren, S.; Chen, J.; Duscher, D.; Liu, Y.; Guo, G.; Kang, Y.; Xiong, H.; Zhan, P.; Wang, Y.; Wang, C.; et al. Microvesicles from human adipose stem cells promote wound healing by optimizing cellular functions via AKT and ERK signaling pathways. Stem Cell Res. Ther. 2019, 10, 47. [Google Scholar] [CrossRef]
  49. Li, Q.; Hu, W.; Huang, Q.; Yang, J.; Li, B.; Ma, K.; Wei, Q.; Wang, Y.; Su, J.; Sun, M.; et al. MiR146a-loaded engineered exosomes released from silk fibroin patch promote diabetic wound healing by targeting IRAK1. Signal Transduct. Target. Ther. 2023, 8, 62. [Google Scholar] [CrossRef]
  50. Chen, B.; Sun, Y.; Zhang, J.; Zhu, Q.; Yang, Y.; Niu, X.; Deng, Z.; Li, Q.; Wang, Y. Human embryonic stem cell-derived exosomes promote pressure ulcer healing in aged mice by rejuvenating senescent endothelial cells. Stem Cell Res. Ther. 2019, 10, 142. [Google Scholar] [CrossRef] [Green Version]
  51. Zhang, Z.; Mi, T.; Jin, L.; Li, M.; Zhanghuang, C.; Wang, J.; Tan, X.; Lu, H.; Shen, L.; Long, C.; et al. Comprehensive proteomic analysis of exosome mimetic vesicles and exosomes derived from human umbilical cord mesenchymal stem cells. Stem Cell Res. Ther. 2022, 13, 312. [Google Scholar] [CrossRef]
  52. Liang, Z.H.; Pan, N.F.; Lin, S.S.; Qiu, Z.Y.; Liang, P.; Wang, J.; Zhang, Z.; Pan, Y.C. Exosomes from mmu_circ_0001052-modified adipose-derived stem cells promote angiogenesis of DFU via miR-106a-5p and FGF4/p38MAPK pathway. Stem Cell Res. Ther. 2022, 13, 336. [Google Scholar] [CrossRef] [PubMed]
  53. Li, Y.; Yu, Y.; Xie, Z.; Ye, X.; Liu, X.; Xu, B.; Mao, J. Serum-derived exosomes accelerate scald wound healing in mice by optimizing cellular functions and promoting Akt phosphorylation. Biotechnol. Lett. 2021, 43, 1675–1684. [Google Scholar] [CrossRef] [PubMed]
  54. Chen, J.; Zhou, R.; Liang, Y.; Fu, X.; Wang, D.; Wang, C. Blockade of lncRNA-ASLNCS5088-enriched exosome generation in M2 macrophages by GW4869 dampens the effect of M2 macrophages on orchestrating fibroblast activation. FASEB J. 2019, 33, 12200–12212. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Zhu, Z.; Zhang, X.; Hao, H.; Xu, H.; Shu, J.; Hou, Q.; Wang, M. Exosomes Derived From Umbilical Cord Mesenchymal Stem Cells Treat Cutaneous Nerve Damage and Promote Wound Healing. Front. Cell Neurosci. 2022, 16, 913009. [Google Scholar] [CrossRef] [PubMed]
  56. Lu, M.; Peng, L.; Ming, X.; Wang, X.; Cui, A.; Li, Y.; Wang, X.; Meng, D.; Sun, N.; Xiang, M.; et al. Enhanced wound healing promotion by immune response-free monkey autologous iPSCs and exosomes vs. their allogeneic counterparts. EBioMedicine 2019, 42, 443–457. [Google Scholar] [CrossRef] [Green Version]
  57. Zhu, J.; Quan, H. Adipose-derived stem cells-derived exosomes facilitate cutaneous wound healing by delivering XIST and restoring discoidin domain receptor 2. Cytokine 2022, 158, 155981. [Google Scholar] [CrossRef]
  58. Yan, C.; Xv, Y.; Lin, Z.; Endo, Y.; Xue, H.; Hu, Y.; Hu, L.; Chen, L.; Cao, F.; Zhou, W.; et al. Human Umbilical Cord Mesenchymal Stem Cell-Derived Exosomes Accelerate Diabetic Wound Healing via Ameliorating Oxidative Stress and Promoting Angiogenesis. Front. Bioeng. Biotechnol. 2022, 10, 829868. [Google Scholar] [CrossRef]
  59. Zhang, Y.; Han, F.; Gu, L.; Ji, P.; Yang, X.; Liu, M.; Tao, K.; Hu, D. Adipose mesenchymal stem cell exosomes promote wound healing through accelerated keratinocyte migration and proliferation by activating the AKT/HIF-1α axis. J. Mol. Histol. 2020, 51, 375–383. [Google Scholar] [CrossRef]
  60. Teng, L.; Maqsood, M.; Zhu, M.; Zhou, Y.; Kang, M.; Zhou, J.; Chen, J. Exosomes Derived from Human Umbilical Cord Mesenchymal Stem Cells Accelerate Diabetic Wound Healing via Promoting M2 Macrophage Polarization, Angiogenesis, and Collagen Deposition. Int. J. Mol. Sci. 2022, 23, 10421. [Google Scholar] [CrossRef]
  61. Zhao, G.; Liu, F.; Liu, Z.; Zuo, K.; Wang, B.; Zhang, Y.; Han, X.; Lian, A.; Wang, Y.; Liu, M.; et al. MSC-derived exosomes attenuate cell death through suppressing AIF nucleus translocation and enhance cutaneous wound healing. Stem Cell Res. Ther. 2020, 11, 174. [Google Scholar] [CrossRef]
  62. Zhao, D.; Yu, Z.; Li, Y.; Wang, Y.; Li, Q.; Han, D. GelMA combined with sustained release of HUVECs derived exosomes for promoting cutaneous wound healing and facilitating skin regeneration. J. Mol. Histol. 2020, 51, 251–263. [Google Scholar] [CrossRef] [PubMed]
  63. Li, J.; Gao, H.; Xiong, Y.; Wang, L.; Zhang, H.; He, F.; Zhao, J.; Liu, S.; Gao, L.; Guo, Y.; et al. Enhancing Cutaneous Wound Healing Based on Human Induced Neural Stem Cell-derived Exosomes. Int. J. Nanomed. 2022, 17, 5991–6006. [Google Scholar] [CrossRef] [PubMed]
  64. Ge, L.; Wang, K.; Lin, H.; Tao, E.; Xia, W.; Wang, F.; Mao, C.; Feng, Y. Engineered exosomes derived from miR-132-overexpresssing adipose stem cells promoted diabetic wound healing and skin reconstruction. Front. Bioeng. Biotechnol. 2023, 11, 1129538. [Google Scholar] [CrossRef] [PubMed]
  65. Wei, P.; Zhong, C.; Yang, X.; Shu, F.; Xiao, S.; Gong, T.; Luo, P.; Li, L.; Chen, Z.; Zheng, Y.; et al. Exosomes derived from human amniotic epithelial cells accelerate diabetic wound healing via PI3K-AKT-mTOR-mediated promotion in angiogenesis and fibroblast function. Burn. Trauma 2020, 8, tkaa020. [Google Scholar] [CrossRef] [PubMed]
  66. Lyu, L.; Cai, Y.; Zhang, G.; Jing, Z.; Liang, J.; Zhang, R.; Dang, X.; Zhang, C. Exosomes derived from M2 macrophages induce angiogenesis to promote wound healing. Front. Mol. Biosci. 2022, 9, 1008802. [Google Scholar] [CrossRef] [PubMed]
  67. Zhou, Y.; Zhao, B.; Zhang, X.L.; Lu, Y.J.; Lu, S.T.; Cheng, J.; Fu, Y.; Lin, L.; Zhang, N.Y.; Li, P.X.; et al. Combined topical and systemic administration with human adipose-derived mesenchymal stem cells (hADSC) and hADSC-derived exosomes markedly promoted cutaneous wound healing and regeneration. Stem Cell Res. Ther. 2021, 12, 257. [Google Scholar] [CrossRef] [PubMed]
  68. Han, C.; Liu, F.; Zhang, Y.; Chen, W.; Luo, W.; Ding, F.; Lu, L.; Wu, C.; Li, Y. Human Umbilical Cord Mesenchymal Stem Cell Derived Exosomes Delivered Using Silk Fibroin and Sericin Composite Hydrogel Promote Wound Healing. Front. Cardiovasc. Med. 2021, 8, 713021. [Google Scholar] [CrossRef] [PubMed]
  69. Li, M.; Wang, T.; Tian, H.; Wei, G.; Zhao, L.; Shi, Y. Macrophage-derived exosomes accelerate wound healing through their anti-inflammation effects in a diabetic rat model. Artif. Cells Nanomed. Biotechnol. 2019, 47, 3793–3803. [Google Scholar] [CrossRef] [Green Version]
  70. Zhang, X.; Gan, J.; Fan, L.; Luo, Z.; Zhao, Y. Bioinspired Adaptable Indwelling Microneedles for Treatment of Diabetic Ulcers. Adv. Mater. 2023, 35, e2210903. [Google Scholar] [CrossRef]
  71. Zhang, Y.; Zhang, P.; Gao, X.; Chang, L.; Chen, Z.; Mei, X. Preparation of exosomes encapsulated nanohydrogel for accelerating wound healing of diabetic rats by promoting angiogenesis. Mater. Sci. Eng. C Mater. Biol. Appl. 2021, 120, 111671. [Google Scholar] [CrossRef]
  72. Shen, Y.F.; Huang, J.H.; Wang, K.Y.; Zheng, J.; Cai, L.; Gao, H.; Li, X.L.; Li, J.F. PTH Derivative promotes wound healing via synergistic multicellular stimulating and exosomal activities. Cell Commun. Signal 2020, 18, 40. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Wang, L.; Cai, Y.; Zhang, Q.; Zhang, Y. Pharmaceutical Activation of Nrf2 Accelerates Diabetic Wound Healing by Exosomes from Bone Marrow Mesenchymal Stem Cells. Int. J. Stem Cells 2022, 15, 164–172. [Google Scholar] [CrossRef] [PubMed]
  74. Li, C.; An, Y.; Sun, Y.; Yang, F.; Xu, Q.; Wang, Z. Adipose Mesenchymal Stem Cell-Derived Exosomes Promote Wound Healing Through the WNT/β-catenin Signaling Pathway in Dermal Fibroblasts. Stem Cell Rev. Rep. 2022, 18, 2059–2073. [Google Scholar] [CrossRef] [PubMed]
  75. Liu, Y.; Zhang, M.; Liao, Y.; Chen, H.; Su, D.; Tao, Y.; Li, J.; Luo, K.; Wu, L.; Zhang, X.; et al. Human umbilical cord mesenchymal stem cell-derived exosomes promote murine skin wound healing by neutrophil and macrophage modulations revealed by single-cell RNA sequencing. Front. Immunol. 2023, 14, 1142088. [Google Scholar] [CrossRef] [PubMed]
  76. Liu, Y.; Liu, Y.; Zhao, Y.; Wu, M.; Mao, S.; Cong, P.; Zou, R.; Hou, M.; Jin, H.; Bao, Y. Application of adipose mesenchymal stem cell-derived exosomes-loaded β-chitin nanofiber hydrogel for wound healing. Folia Histochem. Cytobiol. 2022, 60, 167–178. [Google Scholar] [CrossRef]
  77. Zhang, Y.; Bai, X.; Shen, K.; Luo, L.; Zhao, M.; Xu, C.; Jia, Y.; Xiao, D.; Li, Y.; Gao, X.; et al. Exosomes Derived from Adipose Mesenchymal Stem Cells Promote Diabetic Chronic Wound Healing through SIRT3/SOD2. Cells 2022, 11, 2568. [Google Scholar] [CrossRef]
  78. Sjöqvist, S.; Ishikawa, T.; Shimura, D.; Kasai, Y.; Imafuku, A.; Bou-Ghannam, S.; Iwata, T.; Kanai, N. Exosomes derived from clinical-grade oral mucosal epithelial cell sheets promote wound healing. J. Extracell. Vesicles 2019, 8, 1565264. [Google Scholar] [CrossRef]
  79. Zhou, Z.; Zheng, J.; Lin, D.; Xu, R.; Chen, Y.; Hu, X. Exosomes derived from dental pulp stem cells accelerate cutaneous wound healing by enhancing angiogenesis via the Cdc42/p38 MAPK pathway. Int. J. Mol. Med. 2022, 50, 143. [Google Scholar] [CrossRef]
  80. Han, X.; Wu, P.; Li, L.; Sahal, H.M.; Ji, C.; Zhang, J.; Wang, Y.; Wang, Q.; Qian, H.; Shi, H.; et al. Exosomes derived from autologous dermal fibroblasts promote diabetic cutaneous wound healing through the Akt/β-catenin pathway. Cell Cycle 2021, 20, 616–629. [Google Scholar] [CrossRef]
  81. Camões, S.P.; Bulut, O.; Yazar, V.; Gaspar, M.M.; Simões, S.; Ferreira, R.; Vitorino, R.; Santos, J.M.; Gursel, I.; Miranda, J.P. 3D-MSCs A151 ODN-loaded exosomes are immunomodulatory and reveal a proteomic cargo that sustains wound resolution. J. Adv. Res. 2022, 41, 113–128. [Google Scholar] [CrossRef]
  82. Zhang, Y.; Shi, L.; Li, X.; Liu, Y.; Zhang, G.; Wang, Y. Placental stem cells-derived exosomes stimulate cutaneous wound regeneration via engrailed-1 inhibition. Front. Bioeng. Biotechnol. 2022, 10, 1044773. [Google Scholar] [CrossRef] [PubMed]
  83. Zhu, J.; Liu, Z.; Wang, L.; Jin, Q.; Zhao, Y.; Du, A.; Ding, N.; Wang, Y.; Jiang, H.; Zhu, L. Exosome Mimetics-Loaded Hydrogel Accelerates Wound Repair by Transferring Functional Mitochondrial Proteins. Front. Bioeng. Biotechnol. 2022, 10, 866505. [Google Scholar] [CrossRef] [PubMed]
  84. Chen Md, G.; Wu Md, Y.; Zou Md, L.; Zeng Md, Y. Effect of MicroRNA-146a Modified Adipose-Derived Stem Cell Exosomes on Rat Back Wound Healing. Int. J. Low. Extrem. Wounds 2021, 1–9. [Google Scholar] [CrossRef]
  85. Yu, Q.; Liu, L.; Zhang, X.; Chang, H.; Ma, S.; Xie, Z.; Tang, S.; Ju, X.; Zhu, H.; Shen, B.; et al. MiR-221-3p targets HIPK2 to promote diabetic wound healing. Microvasc. Res. 2022, 140, 104306. [Google Scholar] [CrossRef]
  86. Wang, X.; Jiao, Y.; Pan, Y.; Zhang, L.; Gong, H.; Qi, Y.; Wang, M.; Gong, H.; Shao, M.; Wang, X.; et al. Fetal Dermal Mesenchymal Stem Cell-Derived Exosomes Accelerate Cutaneous Wound Healing by Activating Notch Signaling. Stem Cells Int. 2019, 2019, 2402916. [Google Scholar] [CrossRef] [PubMed]
  87. Xu, J.; Bai, S.; Cao, Y.; Liu, L.; Fang, Y.; Du, J.; Luo, L.; Chen, M.; Shen, B.; Zhang, Q. miRNA-221-3p in Endothelial Progenitor Cell-Derived Exosomes Accelerates Skin Wound Healing in Diabetic Mice. Diabetes Metab. Syndr. Obes. 2020, 13, 1259–1270. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Ahmadpour, F.; Rasouli, H.R.; Talebi, S.; Golchin, D.; Esmailinejad, M.R.; Razie, A. Effects of exosomes derived from fibroblast cells on skin wound healing in Wistar rats. Burns 2023. [Google Scholar] [CrossRef]
  89. Hsu, H.H.; Wang, A.Y.L.; Loh, C.Y.Y.; Pai, A.A.; Kao, H.K. Therapeutic Potential of Exosomes Derived from Diabetic Adipose Stem Cells in Cutaneous Wound Healing of db/db Mice. Pharmaceutics 2022, 14, 1206. [Google Scholar] [CrossRef]
  90. Parvanian, S.; Yan, F.; Su, D.; Coelho-Rato, L.S.; Venu, A.P.; Yang, P.; Zou, X.; Jiu, Y.; Chen, H.; Eriksson, J.E.; et al. Exosomal vimentin from adipocyte progenitors accelerates wound healing. Cytoskeleton 2020, 77, 399–413. [Google Scholar] [CrossRef]
  91. Born, L.J.; Chang, K.H.; Shoureshi, P.; Lay, F.; Bengali, S.; Hsu, A.T.W.; Abadchi, S.N.; Harmon, J.W.; Jay, S.M. HOTAIR-Loaded Mesenchymal Stem/Stromal Cell Extracellular Vesicles Enhance Angiogenesis and Wound Healing. Adv. Healthc. Mater. 2022, 11, e2002070. [Google Scholar] [CrossRef]
  92. Jiang, L.; Zhang, Y.; Liu, T.; Wang, X.; Wang, H.; Song, H.; Wang, W. Exosomes derived from TSG-6 modified mesenchymal stromal cells attenuate scar formation during wound healing. Biochimie 2020, 177, 40–49. [Google Scholar] [CrossRef] [PubMed]
  93. Liu, J.; Yan, Z.; Yang, F.; Huang, Y.; Yu, Y.; Zhou, L.; Sun, Z.; Cui, D.; Yan, Y. Exosomes Derived from Human Umbilical Cord Mesenchymal Stem Cells Accelerate Cutaneous Wound Healing by Enhancing Angiogenesis through Delivering Angiopoietin-2. Stem Cell Rev. Rep. 2021, 17, 305–317. [Google Scholar] [CrossRef] [PubMed]
  94. Sun, Y.; Ju, Y.; Fang, B. Exosomes from human adipose-derived mesenchymal stromal/stem cells accelerate angiogenesis in wound healing: Implication of the EGR-1/lncRNA-SENCR/DKC1/VEGF-A axis. Hum. Cell 2022, 35, 1375–1390. [Google Scholar] [CrossRef] [PubMed]
  95. Shiekh, P.A.; Singh, A.; Kumar, A. Data supporting exosome laden oxygen releasing antioxidant and antibacterial cryogel wound dressing OxOBand alleviate diabetic and infectious wound healing. Data Brief. 2020, 31, 105671. [Google Scholar] [CrossRef]
  96. Wang, C.; Liang, C.; Wang, R.; Yao, X.; Guo, P.; Yuan, W.; Liu, Y.; Song, Y.; Li, Z.; Xie, X. The fabrication of a highly efficient self-healing hydrogel from natural biopolymers loaded with exosomes for the synergistic promotion of severe wound healing. Biomater. Sci. 2019, 8, 313–324. [Google Scholar] [CrossRef]
  97. Jiang, T.; Wang, Z.; Sun, J. Human bone marrow mesenchymal stem cell-derived exosomes stimulate cutaneous wound healing mediates through TGF-β/Smad signaling pathway. Stem Cell Res. Ther. 2020, 11, 198. [Google Scholar] [CrossRef]
  98. Bakadia, B.M.; Qaed Ahmed, A.A.; Lamboni, L.; Shi, Z.; Mutu Mukole, B.; Zheng, R.; Pierre Mbang, M.; Zhang, B.; Gauthier, M.; Yang, G. Engineering homologous platelet-rich plasma, platelet-rich plasma-derived exosomes, and mesenchymal stem cell-derived exosomes-based dual-crosslinked hydrogels as bioactive diabetic wound dressings. Bioact. Mater. 2023, 28, 74–94. [Google Scholar] [CrossRef]
  99. Parvanian, S.; Zha, H.; Su, D.; Xi, L.; Jiu, Y.; Chen, H.; Eriksson, J.E.; Cheng, F. Exosomal Vimentin from Adipocyte Progenitors Protects Fibroblasts against Osmotic Stress and Inhibits Apoptosis to Enhance Wound Healing. Int. J. Mol. Sci. 2021, 22, 4678. [Google Scholar] [CrossRef]
  100. Zhang, X.F.; Wang, T.; Wang, Z.X.; Huang, K.P.; Zhang, Y.W.; Wang, G.L.; Zhang, H.J.; Chen, Z.H.; Wang, C.Y.; Zhang, J.X.; et al. Hypoxic ucMSC-secreted exosomal miR-125b promotes endothelial cell survival and migration during wound healing by targeting TP53INP1. Mol. Ther. Nucleic Acids 2021, 26, 347–359. [Google Scholar] [CrossRef]
  101. Duan, M.; Zhang, Y.; Zhang, H.; Meng, Y.; Qian, M.; Zhang, G. Epidermal stem cell-derived exosomes promote skin regeneration by downregulating transforming growth factor-β1 in wound healing. Stem Cell Res. Ther. 2020, 11, 452. [Google Scholar] [CrossRef]
  102. Lv, Q.; Deng, J.; Chen, Y.; Wang, Y.; Liu, B.; Liu, J. Engineered Human Adipose Stem-Cell-Derived Exosomes Loaded with miR-21-5p to Promote Diabetic Cutaneous Wound Healing. Mol. Pharm. 2020, 17, 1723–1733. [Google Scholar] [CrossRef] [PubMed]
  103. Xu, N.; Wang, L.; Guan, J.; Tang, C.; He, N.; Zhang, W.; Fu, S. Wound healing effects of a Curcuma zedoaria polysaccharide with platelet-rich plasma exosomes assembled on chitosan/silk hydrogel sponge in a diabetic rat model. Int. J. Biol. Macromol. 2018, 117, 102–107. [Google Scholar] [CrossRef] [PubMed]
  104. Xu, M.; Su, X.; Xiao, X.; Yu, H.; Li, X.; Keating, A.; Wang, S.; Zhao, R.C. Hydrogen Peroxide-Induced Senescence Reduces the Wound Healing-Promoting Effects of Mesenchymal Stem Cell-Derived Exosomes Partially via miR-146a. Aging Dis. 2021, 12, 102–115. [Google Scholar] [CrossRef] [PubMed]
  105. Dalirfardouei, R.; Jamialahmadi, K.; Jafarian, A.H.; Mahdipour, E. Promising effects of exosomes isolated from menstrual blood-derived mesenchymal stem cell on wound-healing process in diabetic mouse model. J. Tissue Eng. Regen. Med. 2019, 13, 555–568. [Google Scholar] [CrossRef] [PubMed]
  106. Mi, B.; Chen, L.; Xiong, Y.; Yan, C.; Xue, H.; Panayi, A.C.; Liu, J.; Hu, L.; Hu, Y.; Cao, F.; et al. Saliva exosomes-derived UBE2O mRNA promotes angiogenesis in cutaneous wounds by targeting SMAD6. J. Nanobiotechnol. 2020, 18, 68. [Google Scholar] [CrossRef] [PubMed]
  107. Zhang, Y.; Li, M.; Wang, Y.; Han, F.; Shen, K.; Luo, L.; Li, Y.; Jia, Y.; Zhang, J.; Cai, W.; et al. Exosome/metformin-loaded self-healing conductive hydrogel rescues microvascular dysfunction and promotes chronic diabetic wound healing by inhibiting mitochondrial fission. Bioact. Mater. 2023, 26, 323–336. [Google Scholar] [CrossRef]
  108. Chen, K.; Yu, T.; Wang, X. Inhibition of Circulating Exosomal miRNA-20b-5p Accelerates Diabetic Wound Repair. Int. J. Nanomed. 2021, 16, 371–381. [Google Scholar] [CrossRef]
  109. Xu, Y.; Lin, Z.; He, L.; Qu, Y.; Ouyang, L.; Han, Y.; Xu, C.; Duan, D. Platelet-Rich Plasma-Derived Exosomal USP15 Promotes Cutaneous Wound Healing via Deubiquitinating EIF4A1. Oxid. Med. Cell Longev. 2021, 2021, 9674809. [Google Scholar] [CrossRef]
  110. Kobayashi, H.; Ebisawa, K.; Kambe, M.; Kasai, T.; Suga, H.; Nakamura, K.; Narita, Y.; Ogata, A.; Kamei, Y. Effects of exosomes derived from the induced pluripotent stem cells on skin wound healing. Nagoya J. Med. Sci. 2018, 80, 141–153. [Google Scholar] [CrossRef]
  111. Abdelsaid, K.; Sudhahar, V.; Harris, R.A.; Das, A.; Youn, S.W.; Liu, Y.; McMenamin, M.; Hou, Y.; Fulton, D.; Hamrick, M.W.; et al. Exercise improves angiogenic function of circulating exosomes in type 2 diabetes: Role of exosomal SOD3. FASEB J. 2022, 36, e22177. [Google Scholar] [CrossRef]
  112. Li, P.; Hong, G.; Zhan, W.; Deng, M.; Tu, C.; Wei, J.; Lin, H. Endothelial progenitor cell derived exosomes mediated miR-182-5p delivery accelerate diabetic wound healing via down-regulating PPARG. Int. J. Med. Sci. 2023, 20, 468–481. [Google Scholar] [CrossRef] [PubMed]
  113. Gao, S.; Chen, T.; Hao, Y.; Zhang, F.; Tang, X.; Wang, D.; Wei, Z.; Qi, J. Exosomal miR-135a derived from human amnion mesenchymal stem cells promotes cutaneous wound healing in rats and fibroblast migration by directly inhibiting LATS2 expression. Stem Cell Res. Ther. 2020, 11, 56. [Google Scholar] [CrossRef] [PubMed]
  114. Fu, W.; Liang, D.; Wu, X.; Chen, H.; Hong, X.; Wang, J.; Zhu, T.; Zeng, T.; Lin, W.; Chen, S.; et al. Long noncoding RNA LINC01435 impedes diabetic wound healing by facilitating YY1-mediated HDAC8 expression. iScience 2022, 25, 104006. [Google Scholar] [CrossRef] [PubMed]
  115. Liu, Y.; Liu, Y.; Wu, M.; Zou, R.; Mao, S.; Cong, P.; Hou, M.; Jin, H.; Zhao, Y.; Bao, Y. Adipose-derived mesenchymal stem cell-loaded β-chitin nanofiber hydrogel promote wound healing in rats. J. Mater. Sci. Mater. Med. 2022, 33, 12. [Google Scholar] [CrossRef] [PubMed]
  116. Jin, M.H.; Yu, N.N.; Jin, Y.H.; Mao, Y.Y.; Feng, L.; Liu, Y.; Wang, A.G.; Sun, H.N.; Kwon, T.; Han, Y.H. Peroxiredoxin II with dermal mesenchymal stem cells accelerates wound healing. Aging 2021, 13, 13926–13940. [Google Scholar] [CrossRef]
  117. Park, D.J.; Duggan, E.; Ho, K.; Dorschner, R.A.; Dobke, M.; Nolan, J.P.; Eliceiri, B.P. Serpin-loaded extracellular vesicles promote tissue repair in a mouse model of impaired wound healing. J. Nanobiotechnol. 2022, 20, 474. [Google Scholar] [CrossRef]
  118. Li, Q.; Guo, L.; Wang, J.; Tao, S.; Jin, P. Exosomes derived from Nr-CWS pretreated MSCs facilitate diabetic wound healing by promoting angiogenesis via the circIARS1/miR-4782-5p/VEGFA axis. Chin. J. Nat. Med. 2023, 21, 172–184. [Google Scholar] [CrossRef]
  119. Ding, J.; Wang, X.; Chen, B.; Zhang, J.; Xu, J. Exosomes Derived from Human Bone Marrow Mesenchymal Stem Cells Stimulated by Deferoxamine Accelerate Cutaneous Wound Healing by Promoting Angiogenesis. Biomed. Res. Int. 2019, 2019, 9742765. [Google Scholar] [CrossRef] [Green Version]
  120. Zhang, Y.; Yan, J.; Liu, Y.; Chen, Z.; Li, X.; Tang, L.; Li, J.; Duan, M.; Zhang, G. Human Amniotic Fluid Stem Cell-Derived Exosomes as a Novel Cell-Free Therapy for Cutaneous Regeneration. Front. Cell Dev. Biol. 2021, 9, 685873. [Google Scholar] [CrossRef]
  121. Pi, L.; Yang, L.; Fang, B.R.; Meng, X.X.; Qian, L. Exosomal microRNA-125a-3p from human adipose-derived mesenchymal stem cells promotes angiogenesis of wound healing through inhibiting PTEN. Mol. Cell Biochem. 2022, 477, 115–127. [Google Scholar] [CrossRef]
  122. Zhao, B.; Li, X.; Shi, X.; Shi, X.; Zhang, W.; Wu, G.; Wang, X.; Su, L.; Hu, D. Exosomal MicroRNAs Derived from Human Amniotic Epithelial Cells Accelerate Wound Healing by Promoting the Proliferation and Migration of Fibroblasts. Stem Cells Int. 2018, 2018, 5420463. [Google Scholar] [CrossRef] [PubMed]
  123. Qiu, J.; Shu, C.; Li, X.; Ye, C.; Zhang, W.C. Exosomes from linc00511-overexpressing ADSCs accelerates angiogenesis in diabetic foot ulcers healing by suppressing PAQR3-induced Twist1 degradation. Diabetes Res. Clin. Pract. 2021, 180, 109032. [Google Scholar] [CrossRef] [PubMed]
  124. Xiong, Y.; Chen, L.; Yu, T.; Yan, C.; Zhou, W.; Cao, F.; You, X.; Zhang, Y.; Sun, Y.; Liu, J.; et al. Inhibition of circulating exosomal microRNA-15a-3p accelerates diabetic wound repair. Aging 2020, 12, 8968–8986. [Google Scholar] [CrossRef]
  125. Shi, M.; Gao, Y.; Lee, L.; Song, T.; Zhou, J.; Yan, L.; Li, Y. Adaptive Gelatin Microspheres Enhanced Stem Cell Delivery and Integration With Diabetic Wounds to Activate Skin Tissue Regeneration. Front. Bioeng. Biotechnol. 2022, 10, 813805. [Google Scholar] [CrossRef] [PubMed]
  126. Li, X.; Wang, Y.; Shi, L.; Li, B.; Li, J.; Wei, Z.; Lv, H.; Wu, L.; Zhang, H.; Yang, B.; et al. Magnetic targeting enhances the cutaneous wound healing effects of human mesenchymal stem cell-derived iron oxide exosomes. J. Nanobiotechnol. 2020, 18, 113. [Google Scholar] [CrossRef]
  127. Kim, J.; Kim, E.H.; Lee, H.; Sung, J.H.; Bang, O.Y. Clinical-Scale Mesenchymal Stem Cell-Derived Extracellular Vesicle Therapy for Wound Healing. Int. J. Mol. Sci. 2023, 24, 4273. [Google Scholar] [CrossRef]
  128. Liu, Z.; Yang, S.; Li, X.; Wang, S.; Zhang, T.; Huo, N.; Duan, R.; Shi, Q.; Zhang, J.; Xu, J. Local transplantation of GMSC-derived exosomes to promote vascularized diabetic wound healing by regulating the Wnt/β-catenin pathways. Nanoscale Adv. 2023, 5, 916–926. [Google Scholar] [CrossRef] [PubMed]
  129. Li, Q.; Gong, S.; Yao, W.; Yang, Z.; Wang, R.; Yu, Z.; Wei, M. Exosome loaded genipin crosslinked hydrogel facilitates full thickness cutaneous wound healing in rat animal model. Drug Deliv. 2021, 28, 884–893. [Google Scholar] [CrossRef]
  130. Han, Z.F.; Cao, J.H.; Liu, Z.Y.; Yang, Z.; Qi, R.X.; Xu, H.L. Exosomal lncRNA KLF3-AS1 derived from bone marrow mesenchymal stem cells stimulates angiogenesis to promote diabetic cutaneous wound healing. Diabetes Res. Clin. Pract. 2022, 183, 109126. [Google Scholar] [CrossRef]
  131. Kuang, L.; Zhang, C.; Li, B.; Deng, H.; Chen, R.; Li, G. Human Keratinocyte-Derived Exosomal MALAT1 Promotes Diabetic Wound Healing by Upregulating MFGE8 via microRNA-1914-3p. Int. J. Nanomed. 2023, 18, 949–970. [Google Scholar] [CrossRef]
  132. Liu, Y.; Zhuang, X.; Yu, S.; Yang, N.; Zeng, J.; Liu, X.; Chen, X. Exosomes derived from stem cells from apical papilla promote craniofacial soft tissue regeneration by enhancing Cdc42-mediated vascularization. Stem Cell Res. Ther. 2021, 12, 76. [Google Scholar] [CrossRef] [PubMed]
  133. Qian, Z.; Bai, Y.; Zhou, J.; Li, L.; Na, J.; Fan, Y.; Guo, X.; Liu, H. A moisturizing chitosan-silk fibroin dressing with silver nanoparticles-adsorbed exosomes for repairing infected wounds. J. Mater. Chem. B 2020, 8, 7197–7212. [Google Scholar] [CrossRef] [PubMed]
  134. Nie, W.; Huang, X.; Zhao, L.; Wang, T.; Zhang, D.; Xu, T.; Du, L.; Li, Y.; Zhang, W.; Xiao, F.; et al. Exosomal miR-17-92 derived from human mesenchymal stem cells promotes wound healing by enhancing angiogenesis and inhibiting endothelial cell ferroptosis. Tissue Cell 2023, 83, 102124. [Google Scholar] [CrossRef] [PubMed]
  135. Huang, J.; Yu, M.; Yin, W.; Liang, B.; Li, A.; Li, J.; Li, X.; Zhao, S.; Liu, F. Development of a novel RNAi therapy: Engineered miR-31 exosomes promoted the healing of diabetic wounds. Bioact. Mater. 2021, 6, 2841–2853. [Google Scholar] [CrossRef]
  136. Liu, M.; Liu, Z.; Chen, Y.; Peng, S.; Yang, J.; Chen, C.; Wang, J.; Shang, R.; Tang, Y.; Huang, Y.; et al. Dendritic epidermal T cells secreting exosomes promote the proliferation of epidermal stem cells to enhance wound re-epithelialization. Stem Cell Res. Ther. 2022, 13, 121. [Google Scholar] [CrossRef]
  137. Qian, J.; Park, D.J.; Perrott, S.; Patel, P.; Eliceiri, B.P. Genetic Background and Kinetics Define Wound Bed Extracellular Vesicles in a Mouse Model of Cutaneous Injury. Int. J. Mol. Sci. 2021, 22, 3551. [Google Scholar] [CrossRef]
  138. Yang, H.; Xu, H.; Wang, Z.; Li, X.; Wang, P.; Cao, X.; Xu, Z.; Lv, D.; Rong, Y.; Chen, M.; et al. Analysis of miR-203a-3p/SOCS3-mediated induction of M2 macrophage polarization to promote diabetic wound healing based on epidermal stem cell-derived exosomes. Diabetes Res. Clin. Pract. 2023, 197, 110573. [Google Scholar] [CrossRef]
  139. Wang, S.; Shi, M.; Zhou, J.; Wang, W.; Zhang, Y.; Li, Y. Circulating Exosomal miR-181b-5p Promoted Cell Senescence and Inhibited Angiogenesis to Impair Diabetic Foot Ulcer via the Nuclear Factor Erythroid 2-Related Factor 2/Heme Oxygenase-1 Pathway. Front. Cardiovasc. Med. 2022, 9, 844047. [Google Scholar] [CrossRef]
  140. Henriques-Antunes, H.; Cardoso, R.M.S.; Zonari, A.; Correia, J.; Leal, E.C.; Jiménez-Balsa, A.; Lino, M.M.; Barradas, A.; Kostic, I.; Gomes, C.; et al. The Kinetics of Small Extracellular Vesicle Delivery Impacts Skin Tissue Regeneration. ACS Nano 2019, 13, 8694–8707. [Google Scholar] [CrossRef]
  141. Zeng, J.; Sun, Z.; Zeng, F.; Gu, C.; Chen, X. M2 macrophage-derived exosome-encapsulated microneedles with mild photothermal therapy for accelerated diabetic wound healing. Mater. Today Bio 2023, 20, 100649. [Google Scholar] [CrossRef]
  142. Cardoso, R.M.S.; Rodrigues, S.C.; Gomes, C.F.; Duarte, F.V.; Romao, M.; Leal, E.C.; Freire, P.C.; Neves, R.; Simões-Correia, J. Development of an optimized and scalable method for isolation of umbilical cord blood-derived small extracellular vesicles for future clinical use. Stem Cells Transl. Med. 2021, 10, 910–921. [Google Scholar] [CrossRef]
  143. Zhao, X.; Fu, L.; Zou, H.; He, Y.; Pan, Y.; Ye, L.; Huang, Y.; Fan, W.; Zhang, J.; Ma, Y.; et al. Optogenetic engineered umbilical cord MSC-derived exosomes for remodeling of the immune microenvironment in diabetic wounds and the promotion of tissue repair. J. Nanobiotechnol. 2023, 21, 176. [Google Scholar] [CrossRef]
  144. Zhang, L.; Ouyang, P.; He, G.; Wang, X.; Song, D.; Yang, Y.; He, X. Exosomes from microRNA-126 overexpressing mesenchymal stem cells promote angiogenesis by targeting the PIK3R2-mediated PI3K/Akt signalling pathway. J. Cell Mol. Med. 2021, 25, 2148–2162. [Google Scholar] [CrossRef]
  145. Kou, X.; Xu, X.; Chen, C.; Sanmillan, M.L.; Cai, T.; Zhou, Y.; Giraudo, C.; Le, A.; Shi, S. The Fas/Fap-1/Cav-1 complex regulates IL-1RA secretion in mesenchymal stem cells to accelerate wound healing. Sci. Transl. Med. 2018, 10, eaai8524. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Xia, W.; Liu, Y.; Jiang, X.; Li, M.; Zheng, S.; Zhang, Z.; Huang, X.; Luo, S.; Khoong, Y.; Hou, M.; et al. Lean adipose tissue macrophage derived exosome confers immunoregulation to improve wound healing in diabetes. J. Nanobiotechnol. 2023, 21, 128. [Google Scholar] [CrossRef] [PubMed]
  147. Chen, T.; Song, P.; He, M.; Rui, S.; Duan, X.; Ma, Y.; Armstrong, D.G.; Deng, W. Sphingosine-1-phosphate derived from PRP-Exos promotes angiogenesis in diabetic wound healing via the S1PR1/AKT/FN1 signalling pathway. Burn. Trauma 2023, 11, tkad003. [Google Scholar] [CrossRef] [PubMed]
  148. Yang, H.; Zhang, Y.; Du, Z.; Wu, T.; Yang, C. Hair follicle mesenchymal stem cell exosomal lncRNA H19 inhibited NLRP3 pyroptosis to promote diabetic mouse skin wound healing. Aging 2023, 15, 791–809. [Google Scholar] [CrossRef] [PubMed]
  149. Cao, G.; Chen, B.; Zhang, X.; Chen, H. Human Adipose-Derived Mesenchymal Stem Cells-Derived Exosomal microRNA-19b Promotes the Healing of Skin Wounds Through Modulation of the CCL1/TGF-β Signaling Axis. Clin. Cosmet. Investig. Dermatol. 2020, 13, 957–971. [Google Scholar] [CrossRef]
  150. Wu, M.; Tu, J.; Huang, J.; Wen, H.; Zeng, Y.; Lu, Y. Exosomal IRF1-loaded rat adipose-derived stem cell sheet contributes to wound healing in the diabetic foot ulcers. Mol. Med. 2023, 29, 60. [Google Scholar] [CrossRef]
  151. Zhang, Y.; Fang, M.; Xie, W.; Zhang, Y.A.; Jiang, C.; Li, N.; Li, L.; Tian, J.; Zhou, C. Sprayable alginate hydrogel dressings with oxygen production and exosome loading for the treatment of diabetic wounds. Int. J. Biol. Macromol. 2023, 242, 125081. [Google Scholar] [CrossRef]
  152. Xu, Y.; Ouyang, L.; He, L.; Qu, Y.; Han, Y.; Duan, D. Inhibition of exosomal miR-24-3p in diabetes restores angiogenesis and facilitates wound repair via targeting PIK3R3. J. Cell Mol. Med. 2020, 24, 13789–13803. [Google Scholar] [CrossRef]
  153. Deng, D.; Li, X.; Zhang, J.J.; Yin, Y.; Tian, Y.; Gan, D.; Wu, R.; Wang, J.; Tian, B.M.; Chen, F.M.; et al. Biotin-Avidin System-Based Delivery Enhances the Therapeutic Performance of MSC-Derived Exosomes. ACS Nano 2023, 17, 8530–8550. [Google Scholar] [CrossRef]
  154. Bae, Y.U.; Son, Y.; Kim, C.H.; Kim, K.S.; Hyun, S.H.; Woo, H.G.; Jee, B.A.; Choi, J.H.; Sung, H.K.; Choi, H.C.; et al. Embryonic Stem Cell-Derived mmu-miR-291a-3p Inhibits Cellular Senescence in Human Dermal Fibroblasts Through the TGF-β Receptor 2 Pathway. J. Gerontol. A Biol. Sci. Med. Sci. 2019, 74, 1359–1367. [Google Scholar] [CrossRef] [PubMed]
  155. Yang, C.; Luo, L.; Bai, X.; Shen, K.; Liu, K.; Wang, J.; Hu, D. Highly-expressed micoRNA-21 in adipose derived stem cell exosomes can enhance the migration and proliferation of the HaCaT cells by increasing the MMP-9 expression through the PI3K/AKT pathway. Arch. Biochem. Biophys. 2020, 681, 108259. [Google Scholar] [CrossRef] [PubMed]
  156. Sanders, M.C.; Balaji, S.; Martin, W.B.; Siegmund, N.; Poland, L.; Sanders Hanna, M.; Wei, D.; Kaliada, H.; Littlejohn, S.; Ganey, T. Protecting human amnion and chorion matrices (HACM) during processing: Performance enhancement in a Diabetic Mouse Model and Human Co-culture System. Wound Repair Regen. 2023, 31, 475–488. [Google Scholar] [CrossRef] [PubMed]
  157. Li, B.; Qian, L.; Pi, L.; Meng, X. A therapeutic role of exosomal lncRNA H19 from adipose mesenchymal stem cells in cutaneous wound healing by triggering macrophage M2 polarization. Cytokine 2023, 165, 156175. [Google Scholar] [CrossRef] [PubMed]
  158. Bahr, M.M.; Amer, M.S.; Abo-El-Sooud, K.; Abdallah, A.N.; Shehab, G.G.; El-Tookhy, O.S. Proficiency of Carboxymethylcellulose as a Cryoprotectant. Clinical and Histological Evaluation of Cryopreserved Heterogenous Mesenchymal Stem Cell-Exosomal Hydrogel on Critical Size Skin Wounds in Dogs. Int. J. Hematol. Oncol. Stem Cell Res. 2021, 15, 178–191. [Google Scholar] [CrossRef]
  159. Li, Y.; Zhang, J.; Shi, J.; Liu, K.; Wang, X.; Jia, Y.; He, T.; Shen, K.; Wang, Y.; Liu, J.; et al. Exosomes derived from human adipose mesenchymal stem cells attenuate hypertrophic scar fibrosis by miR-192-5p/IL-17RA/Smad axis. Stem Cell Res. Ther. 2021, 12, 221. [Google Scholar] [CrossRef]
  160. Huayllani, M.T.; Sarabia-Estrada, R.; Restrepo, D.J.; Boczar, D.; Sisti, A.; Nguyen, J.H.; Rinker, B.D.; Moran, S.L.; Quiñones-Hinojosa, A.; Forte, A.J. Adipose-derived stem cells in wound healing of full-thickness skin defects: A review of the literature. J. Plast. Surg. Hand Surg. 2020, 54, 263–279. [Google Scholar] [CrossRef]
  161. Pang, C.; Ibrahim, A.; Bulstrode, N.W.; Ferretti, P. An overview of the therapeutic potential of regenerative medicine in cutaneous wound healing. Int. Wound J. 2017, 14, 450–459. [Google Scholar] [CrossRef]
  162. Shojaei, F.; Rahmati, S.; Banitalebi Dehkordi, M. A review on different methods to increase the efficiency of mesenchymal stem cell-based wound therapy. Wound Repair Regen. 2019, 27, 661–671. [Google Scholar] [CrossRef] [PubMed]
  163. Golchin, A.; Shams, F.; Basiri, A.; Ranjbarvan, P.; Kiani, S.; Sarkhosh-Inanlou, R.; Ardeshirylajimi, A.; Gholizadeh-Ghaleh Aziz, S.; Sadigh, S.; Rasmi, Y. Combination Therapy of Stem Cell-derived Exosomes and Biomaterials in the Wound Healing. Stem Cell Rev. Rep. 2022, 18, 1892–1911. [Google Scholar] [CrossRef] [PubMed]
  164. Rustad, K.C.; Wong, V.W.; Sorkin, M.; Glotzbach, J.P.; Major, M.R.; Rajadas, J.; Longaker, M.T.; Gurtner, G.C. Enhancement of mesenchymal stem cell angiogenic capacity and stemness by a biomimetic hydrogel scaffold. Biomaterials 2012, 33, 80–90. [Google Scholar] [CrossRef] [Green Version]
  165. Nilforoushzadeh, M.A.; Khodadadi Yazdi, M.; Baradaran Ghavami, S.; Farokhimanesh, S.; Mohammadi Amirabad, L.; Zarrintaj, P.; Saeb, M.R.; Hamblin, M.R.; Zare, M.; Mozafari, M. Mesenchymal Stem Cell Spheroids Embedded in an Injectable Thermosensitive Hydrogel: An In Situ Drug Formation Platform for Accelerated Wound Healing. ACS Biomater. Sci. Eng. 2020, 6, 5096–5109. [Google Scholar] [CrossRef]
  166. Grada, A.; Mervis, J.; Falanga, V. Research Techniques Made Simple: Animal Models of Wound Healing. J. Investig. Dermatol. 2018, 138, 2095–2105.e1. [Google Scholar] [CrossRef] [Green Version]
  167. Sami, D.G.; Heiba, H.H.; Abdellatif, A. Wound healing models: A systematic review of animal and non-animal models. Wound Med. 2019, 24, 8–17. [Google Scholar] [CrossRef]
  168. Saeed, S.; Martins-Green, M. Animal models for the study of acute cutaneous wound healing. Wound Repair Regen. 2023, 31, 6–16. [Google Scholar] [CrossRef]
  169. Peric, M.; Dumic-Cule, I.; Grcevic, D.; Matijasic, M.; Verbanac, D.; Paul, R.; Grgurevic, L.; Trkulja, V.; Bagi, C.M.; Vukicevic, S. The rational use of animal models in the evaluation of novel bone regenerative therapies. Bone 2015, 70, 73–86. [Google Scholar] [CrossRef] [Green Version]
  170. De Vries, R.B.M.; Wever, K.E.; Avey, M.T.; Stephens, M.L.; Sena, E.S.; Leenaars, M. The Usefulness of Systematic Reviews of Animal Experiments for the Design of Preclinical and Clinical Studies. ILAR J. 2014, 55, 427–437. [Google Scholar] [CrossRef] [Green Version]
  171. Hanson, S.E.; Kleinbeck, K.R.; Cantu, D.; Kim, J.; Bentz, M.L.; Faucher, L.D.; Kao, W.J.; Hematti, P. Local delivery of allogeneic bone marrow and adipose tissue-derived mesenchymal stromal cells for cutaneous wound healing in a porcine model. J. Tissue Eng. Regen. Med. 2016, 10, E90–E100. [Google Scholar] [CrossRef] [Green Version]
  172. Ochiai, H.; Kishi, K.; Kubota, Y.; Oka, A.; Hirata, E.; Yabuki, H.; Iso, Y.; Suzuki, H.; Umezawa, A. Transplanted mesenchymal stem cells are effective for skin regeneration in acute cutaneous wounds of pigs. Regen. Ther. 2017, 7, 8–16. [Google Scholar] [CrossRef] [PubMed]
  173. Martinello, T.; Gomiero, C.; Perazzi, A.; Iacopetti, I.; Gemignani, F.; DeBenedictis, G.M.; Ferro, S.; Zuin, M.; Martines, E.; Brun, P.; et al. Allogeneic mesenchymal stem cells improve the wound healing process of sheep skin. BMC Vet. Res. 2018, 14, 202. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Ribeiro, J.; Pereira, T.; Amorim, I.; Caseiro, A.R.; Lopes, M.A.; Lima, J.; Gartner, A.; Santos, J.D.; Bártolo, P.J.; Rodrigues, J.M.; et al. Cell therapy with human MSCs isolated from the umbilical cord Wharton jelly associated to a PVA membrane in the treatment of chronic skin wounds. Int. J. Med. Sci. 2014, 11, 979–987. [Google Scholar] [CrossRef] [Green Version]
  175. Enciso, N.; Avedillo, L.; Fermín, M.L.; Fragío, C.; Tejero, C. Cutaneous wound healing: Canine allogeneic ASC therapy. Stem Cell Res. Ther. 2020, 11, 261. [Google Scholar] [CrossRef] [PubMed]
  176. Cibelli, J.; Emborg, M.E.; Prockop, D.J.; Roberts, M.; Schatten, G.; Rao, M.; Harding, J.; Mirochnitchenko, O. Strategies for Improving Animal Models for Regenerative Medicine. Cell Stem Cell 2013, 12, 271–274. [Google Scholar] [CrossRef] [Green Version]
  177. Qiao, Z.; Wang, X.; Zhao, H.; Deng, Y.; Zeng, W.; Yang, K.; Chen, H.; Yan, Q.; Li, C.; Wu, J.; et al. The effectiveness of cell-derived exosome therapy for diabetic wound: A systematic review and meta-analysis. Ageing Res. Rev. 2023, 85, 101858. [Google Scholar] [CrossRef]
Figure 1. PRISMA Flow Diagram that summarizes the selection process.
Figure 1. PRISMA Flow Diagram that summarizes the selection process.
Biomedicines 11 02099 g001
Figure 2. Graphical representation of publication distribution per year, from 2018 to June 2023.
Figure 2. Graphical representation of publication distribution per year, from 2018 to June 2023.
Biomedicines 11 02099 g002
Figure 3. Graphical representation of publication distribution per corresponding authors country (2018–June 2023).
Figure 3. Graphical representation of publication distribution per corresponding authors country (2018–June 2023).
Biomedicines 11 02099 g003
Figure 4. Graphical representation of cell tissue source distribution in the scientific literature between 2018 and June 2023.
Figure 4. Graphical representation of cell tissue source distribution in the scientific literature between 2018 and June 2023.
Biomedicines 11 02099 g004
Figure 5. Graphical representation of the biomaterials used in publications related to wound healing between 2018 and June 2023.
Figure 5. Graphical representation of the biomaterials used in publications related to wound healing between 2018 and June 2023.
Biomedicines 11 02099 g005
Figure 6. Graphical representation of the administration route of exosomes used in wound healing between 2018 and June 2023. EV, endovenous; ID, intradermal; IP, intraperitoneal; SC, subcutaneous.
Figure 6. Graphical representation of the administration route of exosomes used in wound healing between 2018 and June 2023. EV, endovenous; ID, intradermal; IP, intraperitoneal; SC, subcutaneous.
Biomedicines 11 02099 g006
Figure 7. Graphical representation of the animal models used in wound healing between 2018 and June 2023.
Figure 7. Graphical representation of the animal models used in wound healing between 2018 and June 2023.
Biomedicines 11 02099 g007
Table 1. Summary of the retrieved data from the 148 papers. x—No biomaterial.
Table 1. Summary of the retrieved data from the 148 papers. x—No biomaterial.
RefYearCountryCell SourceCell TypeBiomaterialAdministration RouteAnimal
Models
[12]2019ChinaHumanADSCHydrogelSC InjectionMice
[13]2020ChinaHumanUCMSCHydrogelSC InjectionRat
[14]2018ChinaHumanUCMSCxSC InjectionMice
[15]2020ChinaHumanBMSCxSC InjectionRat
[16]2022ChinaHumanUVECPatchPatchMice
[17]2022ChinaHumanADSCHydrogelTopicalMice
[18]2020ChinaHumanBMSCxSC InjectionRat
[19]2021ChinaRatBMSCxSC InjectionRat
[20]2021ChinaHumanADSCxSC InjectionMice
[21]2018ChinaHumanADSCxSC InjectionRat
[22]2022ChinaMiceBMSCHydrogelSC InjectionMice
[23]2022USAHumanEpidermalxSC InjectionMice
[24]2020USAMiceKeratinocytexSC InjectionMice
[25]2022ChinaHumanADSCxSC InjectionMice
[26]2020ChinaHumanBMSCxSC InjectionRat
[27]2022ChinaRatBMSCHydrogelTopicalRat
[28]2020ChinaHumanPeripheral BloodxSC InjectionMice
[29]2020IndiaRatADSCScaffoldScaffoldRat
[30]2022ChinaHumanDPsxSC InjectionMice
[31]2019ChinaHumanADSCScaffoldScaffoldMice
[32]2021ChinaHumanUCMSCxEV InjectionRat
[33]2019ChinaHumanBMSCxSC InjectionMice
[34]2022Republic of KoreaMiceBMSCHydrogelSC InjectionMice
[35]2020ChinaHumanADSCxSC InjectionRat
[36]2020ChinaMiceBMSCxID InjectionMice
[37]2021ChinaHumanADSCxSC InjectionMice
[38]2022ChinaHumanEpidermalHydrogelSC InjectionMice
[39]2022Republic of KoreaHumanADSCxSC InjectionMice
[40]2022ChinaHumanADSCxSC InjectionMice
[41]2019Republic of KoreaMiceBMSCxSC InjectionMice
[42]2020ChinaHumanBMSCxSC InjectionMice
[43]2023ChinaMiceADSCHydrogelSC InjectionMice
[44]2021ChinaHumanADSCScaffoldScaffoldMice
[45]2021ChinaMiceSerumxSC InjectionMice
[46]2022ChinaMiceFibroblastxID InjectionMice
[47]2018ChinaHumanADSCxSC and ID InjectionMice
[48]2019ChinaHumanADSCxSC InjectionMice
[49]2023ChinaHumanPlacentaPatchPatchMice
[50]2019ChinaHumanEmbryonicxTopicalMice
[51]2022ChinaHumanUCMSCxSC InjectionMice
[52]2022ChinaMiceADSCxSC InjectionMice
[53]2021ChinaRat and MiceSerumxSC InjectionMice
[54]2019ChinaHumanMacrophagexSC InjectionMice
[55]2022ChinaHumanUCMSCxTopicalMice
[56]2019ChinaMacaqueiPSCsxTopicalMacaque
[57]2022ChinaMiceADSCxSC InjectionMice
[58]2022ChinaHumanUCMSCxSC InjectionMice
[59]2020ChinaHumanADSCxSC InjectionMice
[60]2022ChinaHumanUCMSCxSC InjectionRat
[61]2020ChinaHumanUCMSCxSC InjectionMice
[62]2020ChinaHumanUVECHydrogelTopicalRat
[63]2022ChinaHumaniPSCsHydrogelTopicalMice
[64]2023ChinaMiceADSCxSC InjectionMice
[65]2020ChinaHumanAmniotic MembranexSC InjectionMice
[66]2022ChinaHumanUVECxSC InjectionMice
[67]2021ChinaHumanADSCxInjection and TopicalMice
[68]2021ChinaHumanUCMSCHydrogelTopicalMice
[69]2019ChinaHumanUVECxSC InjectionRat
[70]2023ChinaHumanUVECHydrogelMicroneedleRat
[71]2021ChinaHumanUCMSCHydrogelSC InjectionRat
[72]2020ChinaHumanUVECHydrogelSC InjectionRat
[73]2022ChinaRatBMSCxSC InjectionRat
[74]2022ChinaHumanADSCxSC InjectionRat
[75]2023ChinaHumanUCMSCxSC InjectionMice
[76]2022ChinaMiceADSCHydrogelTopicalRat
[77]2022ChinaHumanADSCxSC InjectionMice
[78]2019JapanHumanEpithelialxTopicalRat
[79]2022ChinaHumanDPsxSC InjectionMice
[80]2021ChinaRatDermalxSC InjectionRat
[81]2022PortugalHumanUCMSCxSC InjectionRat
[82]2022ChinaRatPlacenta and ADSCxSC InjectionRat
[83]2022ChinaHumanUCMSCHydrogelSC InjectionMice
[84]2021ChinaHumanADSCxSC InjectionRat
[85]2022ChinaHumanUVECxSC InjectionMice
[86]2019ChinaHumanFetal dermalxSC InjectionMice
[87]2020ChinaMiceBMSCxTopicalMice
[88]2023IranHumanFetal dermalxTopicalRat
[89]2022TaiwanMiceADSC and dermalxTopicalMice
[90]2020China and FinlandHumanADSCxIP InjectionMice
[91]2022USAHumanBMSCxSC InjectionMice and Rat
[92]2020ChinaHumanBMSCxSC InjectionMice
[93]2021ChinaHumanUCMSCxSC InjectionRat
[94]2022ChinaHumanADSCxSC InjectionMice
[95]2020IndiaRatADSCScaffoldScaffoldRat
[96]2019ChinaHumanPlacentaHydrogelSC InjectionMice
[97]2020ChinaHumanBMSCxSC InjectionRat
[98]2023ChinaRatBMSC and plasmaHydrogelTopicalRat
[99]2021China and FinlandHumanADSCxIP InjectionMice
[100]2021ChinaHumanUCMSCxSC InjectionMice
[101]2020ChinaHumanEpidermalxSC InjectionRat
[102]2020ChinaHumanADSCHydrogelTopicalRat
[103]2018ChinaHumanPlasmaSpongeSpongeRat
[104]2021ChinaHumanUCMSC and ADSCxSC InjectionMice
[105]2019IranHumanMenstrual BloodxID InjectionMice
[106]2020ChinaHumanSalivaxSC InjectionMice
[107]2023ChinaHumanADSCHydrogelSC InjectionMice
[108]2021ChinaHumanPeripheral BloodxSC InjectionMice
[109]2021ChinaMicePlasmaxSC InjectionMice
[110]2018JapanHumaniPSCsxSC InjectionMice
[111]2022USAMice and HumanPlasmaxTopicalMice
[112]2023ChinaHumanUCMSCxTopicalMice
[113]2020ChinaHumanAmnionxSC InjectionRat
[114]2022ChinaHumanKeratinocytexSC InjectionMice
[115]2022ChinaMiceADSCHydrogelSC InjectionRat
[116]2021ChinaMiceDermalxID InjectionMice
[117]2022USAMiceSkinSpongeSpongeMice
[118]2023ChinaHumanUCMSCxSC InjectionMice
[119]2019ChinaHumanBMSCxSC InjectionRat
[120]2021ChinaHumanAmniotic FluidxSC InjectionRat
[121]2022ChinaHumanADSCxSC InjectionMice
[122]2018ChinaHumanAmniotic FluidxSC InjectionMice
[123]2021ChinaHumanADSCxSC InjectionRat
[124]2020ChinaHumanPeripheral BloodxInjectionMice
[125]2022ChinaRatADSCHydrogelTopicalRat
[126]2020ChinaHumanUCMSCNanoparticlesEV InjectionRat
[127]2023Republic of KoreaHumanUCMSCxInjection and TopicalMice and Rat
[128]2022ChinaHumanGingivalxSC InjectionMice
[129]2021ChinaHumanUCMSCHydrogelTopicalRat
[130]2022ChinaHumanBMSCxEV InjectionMice
[131]2023ChinaHumanKeratinocytexSC InjectionMice
[132]2021ChinaHumanDPsxTopicalMice
[133]2020ChinaHumanUCMSCDressingTopicalMice
[134]2023ChinaHumanUCMSCxSC InjectionMice
[135]2021ChinaHumanEmbryonicxSC InjectionRat
[136]2022ChinaMiceDendritic epidermal T cellsxSC InjectionMice
[137]2021USAMiceSkinSpongeTopicalMice
[138]2023ChinaHumanEpidermalxSC InjectionMice
[139]2022ChinaHumanUVECxSC InjectionMice
[140]2019PortugalHumanUCMSCHydrogelTopicalMice
[141]2023ChinaMiceMacrophagexTopicalRat
[142]2021PortugalHumanUCMSCHydrogelTopicalMice
[143]2023ChinaHumanUCMSCxSC InjectionMice
[144]2021ChinaHumanBMSCxSC InjectionMice
[145]2018USAMice and HumanBMSCs, Skin and GingivaxSC InjectionMice
[146]2023ChinaMiceADSCxSC InjectionMice
[147]2023ChinaHumanPlasmaxSC InjectionMice
[148]2023ChinaHumanHair folliclexSC InjectionMice
[149]2020ChinaHumanADSCxSC InjectionMice
[150]2023ChinaRatADSCxSC InjectionRat
[151]2023ChinaMiceBMSCDressingTopicalRat
[152]2020ChinaHumanPeripheral BloodxSC InjectionMice
[153]2023ChinaRatBMSCHydrogelSC InjectionMice and Rat
[154]2019Republic of KoreaHumanFibroblastxTopicalMice
[155]2020ChinaHumanADSCxSC InjectionMice
[156]2023USAHumanPlacentaxTopicalMice
[157]2023ChinaHumanADSCxSC InjectionMice
[158]2021EgyptDogBMSCHydrogelTopicalDog
[159]2021ChinaHumanADSCxSC InjectionMice
Table 2. Summary of the most used cell types of the most common species. X—Not applicable.
Table 2. Summary of the most used cell types of the most common species. X—Not applicable.
Human (109 Studies)Mice (23 Studies)Rat (11 Studies)
BMSC11 (10.1%)6 (26.1%)4 (36.4%)
ADSC29 (26.6%)7 (30.4%)4 (36.4%)
UCMSC25 (22.9%)XX
UVECS8 (7.3%)XX
DP3 (2.8%)XX
Epidermal4 (3.7%)XX
Peripherical blood4 (3.7%)XX
Placenta3 (2.8%)XX
Others22 (20.2%)10 (43.5%)3 (27.3%)
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Sousa, P.; Lopes, B.; Sousa, A.C.; Moreira, A.; Coelho, A.; Alvites, R.; Alves, N.; Geuna, S.; Maurício, A.C. Advancements and Insights in Exosome-Based Therapies for Wound Healing: A Comprehensive Systematic Review (2018–June 2023). Biomedicines 2023, 11, 2099. https://doi.org/10.3390/biomedicines11082099

AMA Style

Sousa P, Lopes B, Sousa AC, Moreira A, Coelho A, Alvites R, Alves N, Geuna S, Maurício AC. Advancements and Insights in Exosome-Based Therapies for Wound Healing: A Comprehensive Systematic Review (2018–June 2023). Biomedicines. 2023; 11(8):2099. https://doi.org/10.3390/biomedicines11082099

Chicago/Turabian Style

Sousa, Patrícia, Bruna Lopes, Ana Catarina Sousa, Alícia Moreira, André Coelho, Rui Alvites, Nuno Alves, Stefano Geuna, and Ana Colette Maurício. 2023. "Advancements and Insights in Exosome-Based Therapies for Wound Healing: A Comprehensive Systematic Review (2018–June 2023)" Biomedicines 11, no. 8: 2099. https://doi.org/10.3390/biomedicines11082099

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop