Abstract
Exosome therapy holds great promise as a novel approach to improve acute skin wound healing. This review provides a comprehensive overview of the current understanding of exosome biology and its potential applications in acute skin wound healing and beyond. Exosomes, small extracellular vesicles secreted by various stem cells, have emerged as potent mediators of intercellular communication and tissue repair. One advantage of exosome therapy is its ability to avoid potential risks associated with stem cell therapy, such as immune rejection or stem cells differentiating into unwanted cell types. However, further research is necessary to optimize exosome therapy, not only in the areas of exosome isolation, characterization, and engineering, but also in determining the optimal dose, timing, administration, and frequency of exosome therapy. Thus, optimization of exosome therapy is critical for the development of more effective and safer exosome-based therapies for acute skin wound healing and other diseases induced by cancer, ischemia, or inflammation. This review provides valuable insights into the potential of exosome therapy and highlights the need for further research to optimize exosome therapy for clinical use.
Keywords: exosomes, stem cells, therapeutic impact, skin, wound healing
Introduction
Acute skin wounds generally result from surgery, traumatic wounds, superficial burns, and abrasions, and can lead to complications such as wound infection, delayed healing or non-healing, and scar formation. These complications not only require significant medical resources but can also affect the mental health of some patients [1,2]. The severity of acute skin wounds is rising rapidly, making it a global public health concern. Therefore, the therapy and care of acute skin wounds is an urgent health issue.
Skin is the largest organ in the human body and it is exposed to various types of damage, such as burns, cuts, abrasions, and infections. There are several types of therapies that can help repair or replace damaged skin, including cell therapy and tissue-engineered skin. Cell therapy involves using stem cells or other types of cells to promote skin regeneration. Stem cells have the ability to differentiate into different types of cells, such as skin cells, endothelial cells, and stromal cells [3-6]. These adult stem cells can be obtained from various sources, such as bone marrow or adipose tissue. Cell therapy can also involve the use of growth factors or cytokines to stimulate cell growth and differentiation. Recent studies have demonstrated the effect of growth factors and cytokines, including platelet-derived growth factors, epidermal growth factor, fibroblast growth factors, vascular endothelial growth factor and transforming growth factor-β (TGF-β), and keratinocyte growth factors on therapeutic strategies in wound healing, which can significantly increase the level of neovascularization and the number of capillaries to promote angiogenesis and reduce scar formation at the wound site [7,8]. Tissue-engineered skin, on the other hand, involves creating a skin substitute using a combination of cells and biomaterials. The process typically involves seeding skin cells onto a scaffold made of biodegradable materials, such as collagen or hyaluronic acid. The scaffold provides a framework for the cells to grow and organize into tissue that can be transplanted onto the patient’s skin. Both cell therapy and tissue-engineered skin have shown promise in treating various skin conditions and injuries [9,10]. Our preliminary research also indicates that stem cells derived from mesenchymal tissues and kidney present in the urine can promote skin wound healing [11-13].
While stem cells have great potential for skin regeneration, they also have limitations and risks. One of the potential risks associated with stem cell therapy is the formation of tumors during the differentiation process, especially embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) [14-17]. What’s more, Min et al. discovered that dysplastic stem cells can drive the neoplastic transformation of precancerous gastric mucosa [18]. In addition, stem cell therapy may be limited by immune rejection [19-21], which occurs when the patient’s immune system attacks the transplanted cells as foreign [22]. A recent study explored the immunogenicity of iPSCs derivatives in allogeneic recipients. While iPSCs themselves are considered less immunogenic, the study reveals that their derivatives may still trigger an immune response, particularly if they are not properly modulated for immune evasion [19]. This can lead to the failure of the treatment and potential harm to the patient [23]. Furthermore, stem cell therapy can be time-consuming and expensive, as it requires the growth and differentiation of large numbers of cells in culture. This can limit the practicality and affordability of the therapy, making it less accessible to patients [24].
The extracellular matrix (ECM) plays a critical role in the repair of skin tissue. It is a complex network of proteins and other molecules that provide structural support and help regulate cellular behavior. Our previous studies have shown that skin ECM can better maintain skin-derived cells, and promote cell proliferation and differentiation, but their phenotype and function may change during ECM culture [25]. In skin tissue repair, xenogenous skin decellularized matrix serves as a scaffold for the migration and proliferation of skin cells, such as keratinocytes and fibroblasts. Skin ECM also contains various growth factors and cytokines that help regulate cell behavior and promote tissue regeneration [26].
The use of stem cell-derived exosomes as a novel therapy for skin wound healing is becoming increasingly important [27]. Exosomes are tiny vesicles ranging in size from 30 to 150 nm, which are released by cells and contain various proteins, lipids, and nucleic acids that can affect the behavior of other cells [28,29]. In various tissue-repairing processes, implanted exosomes regulate necrotic apoptosis, reduce renal tubular epithelial cell damage and enhance repair, enhance the function of the pancreas and provide immune isolation for pancreatic transplantation, and promote endogenous sperm recovery in non-obstructive azoospermic mice [30-32]. Recent studies have shown that exosome therapy has the potential to promote skin regeneration and reduce inflammation in various types of skin injury [33,34].
Stem cell-derived exosome therapy is suitable for several types of skin injuries, such as wounds, burns, scars, dermatitis, and aging-related skin changes. For example, exosome therapy promotes wound healing by stimulating the growth of new blood vessels, promoting cell proliferation and differentiation, and reducing inflammation [33]. Exosome therapy can also reduce the inflammatory response and improves tissue repair and regeneration in burn injuries [33]. For scars, exosome therapy helps reduce scar formation by promoting the remodeling of collagen fibers and reducing inflammation [35]. Moreover, exosome therapy reduces inflammation and promote skin regeneration in various types of dermatitis, such as atopic dermatitis and allergic contact dermatitis [36,37]. Finally, exosome therapy helps to reduce the appearance of wrinkles and fine lines, improve skin texture and tone, and promote the production of collagen and elastin in aging-related skin changes [38].
Although exosomes hold great potential in the treatment of skin injuries, their application is currently limited due to changes in isolation and purification, loading efficiency, low yield, and biological distribution in vivo. In addition, it is important to note that exosome therapy is still in the early stages of development for skin injury. Therefore, further research is needed to optimize exosome therapy, and its safety and efficacy for skin injury therapy needs to be thoroughly evaluate.
Exosome biology and potential applications in skin acute wound healing
Biomarkers of exosomes
Exosomes are nano-sized vesicles (with a diameter of 30–100 nm) with a bilayer lipid membrane structure. They are originated from the intima, stored in polyvesicles, and released outside the cell through plasma membrane fusion [39]. Exosomes carry a variety of bioactive contents such as nucleic acid and protein. They not only mediate signal transduction and information exchange between cells but are also involved in the pathophysiological processes of various diseases in the human body [28,39]. Exosomes contain four transmembrane proteins (CD9, CD63, CD81, CD82), membrane transport and fusion proteins (GTPases, annexins, flotillin), major histocompatibility complex (MHC Class I/Class II), and heat shock proteins (HSP70, HSP90) [40]. Surface proteins and internal biological substances (such as protein, mRNA, miRNA, and DNA) determine the specificity of exosomes [28,41].
Exosome delivery
Exosomes are small extracellular vesicles that are secreted by cells and are involved in intercellular communication. Exosomes can deliver various types of biomolecules, including proteins, lipids, and nucleic acids, to target cells [42]. The process of exosome delivery can be divided into three main steps [43,44].
(1) Biogenesis and packaging: Exosomes are formed by the release of intraluminal vesicles after the fusion of multivesicular endosomes (MVEs) with the plasma membrane. In the early stage, extracellular matrix or membrane proteins are encapsulated by the plasma membrane to form early endosomes, which form intraluminal vesicles and are encapsulated by MVEs over time [29,39,45,46]. During this process, specific proteins, lipids, and nucleic acids are selectively packaged into the exosomes; (2) Release and uptake: Once exosomes are formed, they are released into the extracellular space and can be taken up by neighboring or distant cells. Exosomes can be internalized by target cells through various mechanisms, including endocytosis, direct fusion, and ligand-receptor interaction with the plasma membrane; and (3) Targeting and function: Once exosomes are internalized, the biomolecules they contain can exert various effects on target cells. For example, exosomes can deliver miRNAs that regulate gene expression, proteins that modulate cell signaling pathways, or lipids that affect membrane properties [47].
Therefore, exosomes can serve as a delivery system for therapeutic agents such as drugs or nucleic acids [48], which can target specific cells and promote healing. Studies have suggested that exosomes derived from various cell types such as adipose-derived stem cells, mesenchymal stem cells can promote skin regeneration and repair [49]. These studies demonstrate that exosome therapy can promote wound healing, reduce scar formation, and reduce inflammation in acute skin injury. In sum, exosomes are well tolerated and stable, and have been considered as ideal nanocarriers that can penetrate deep tissues and evade immune cell attacks to deliver drugs to the injured site [44].
Mechanisms of exosome-mediated intercellular communication
Exosomes have the biological function of mediating intercellular communication. They can directly activate cell surface receptors through proteins and bioactive lipid ligands, and fuse with target cell membranes and target cells or act as signal bodies, transfer effector molecules and multifunctional signaling complexes to maintain normal cell physiological functions, i.e., immune surveillance, blood coagulation, stem cell maintenance, tissue repair, and communication functions [40]. When the normal biological function of exosomes suffers unbalanced, it will induce pathological changes, including tumor occurrence and metastasis, autoimmune diseases, neurodegenerative diseases, and HIV infection [29,40].
The involvement of exosomes in skin acute wound healing
Acute skin injury, such as a cut or burn, triggers a series of pathophysiological changes that are part of the body’s natural wound healing response. These changes can be divided into four overlapping phases: hemostasis, inflammation, proliferation, and remodeling [50-52]. Here is a brief overview of the pathophysiological changes that occur during each phase (Fig. 1).
Fig. 1.
Pathophysiological changes at different stages of skin wound healing. (A) At the hemostasis stage, platelets are activated and release various cytokines and growth factors, recruit immune cells and induce vasoconstriction. (B) In the stage of inflammatory response, macrophages and neutrophils gather at the site of injury, macrophages release inflammatory factors, and the M1 phenotype transitions to M2 phenotype to promote wound healing. (C) Endothelial cell vascularization and fibronectin migration promote collagen formation during the proliferation stage. (D) The main process during the remodeling stage involves the formation of type I and II collagen, as well as the reorganization of collagen and ECM. ECM, extracellular matrix.
(1) Hemostasis phase: This is the initial phase of wound healing that occurs immediately after the injury. The body’s first response is to stop bleeding and promote clotting. Platelets aggregate at the site of injury and release various cytokines, growth factors, and chemokines that promote vasoconstriction and recruit immune cells to the site of injury [51]. (2) Inflammatory phase: This phase begins within hours of injury and lasts for up to 5–7 days. During this phase, neutrophils, monocytes, and macrophages migrate to the wound site and remove debris, foreign particles, and pathogens [53,54]. These immune cells release various pro-inflammatory cytokines and chemokines that activate the immune response and recruit more immune cells to the site of injury [51]. (3) Proliferation phase: This phase begins around 3–5 days and lasts up to 3 weeks [54]. During this phase, fibroblasts migrate to the wound site and begin to synthesize collagen and other extracellular matrix components that provide structural support for the healing tissue [54]. New blood vessels form to supply nutrients and oxygen to the healing tissue [55]. Epithelial cells at the wound edges proliferate and migrate across the wound bed to cover the wound. (4) Remodeling phase: This phase can last for up to several months and involves the reorganization of the extracellular matrix and the remodeling of collagen fibers [56]. Collagen synthesis and degradation are balanced to strengthen and align the collagen fibers in the healing tissue. The wound site becomes less vascularized and less cellular as the healing progresses. Those pathophysiological changes are essential for the normal wound healing response.
Studies have shown that exosome signal transduction plays an important role in different stages of wound healing. Exosomes by themselves contain various bioactive molecules such as growth factors, cytokines, and nucleic acids, cannot only promote wound healing by inducing angiogenesis, cell proliferation and differentiation, and vascular remodeling by mediating signal communication between cells, but also play an effective therapeutic role as vehicles for the delivery of drug payload(s) [27,28]. Therefore, exosome therapy has shown promise for treating acute skin injury by promoting tissue regeneration and reducing inflammation. Here are some of the ways in which exosome therapy can potentially treat acute skin injury (Fig. 2).
Fig. 2.
The mechanism of exosomes therapy for acute skin wound. (A) Exosomes of different cellular origins, such as fibroblast, macrophage, stem cell and platelet etc. (B) The role of exosomes in wound healing of the skin includes alleviating inflammatory reactions, promoting proliferation and migration of fibroblasts, promoting angiogenesis of endothelial cells, and accelerating remodeling of extracellular matrix and collagen. (C) Exosomes accelerate skin wound healing and reduce scar formation.
Wound healing is a complex biological process involving multiple cell types and signaling molecules, including cytokines, to orchestrate tissue repair and regeneration. Exosomes, small extracellular vesicles secreted by many cell types, are increasingly recognized for their role in intercellular communication and tissue repair. These exosomes can carry bioactive molecules, including cytokines, growth factors, and microRNAs, modulating the wound healing process. The cascade of cytokine expression in wound healing is a dynamic and coordinated process involving multiple steps. Here is a simplified overview of how cytokines in exosomes are thought to participate in normal wound healing.
Exosomes in hemostasis phase
Platelets can release a variety of cell growth factors, cytokines, and extracellular matrix to promote vasoconstriction and play an important role in hemostasis. Blood contains a large number of exosomes, and platelet-derived exosomes are the most abundant in circulation. When platelet-derived exosomes in circulation are stimulated by inflammation and stress, their secretion increases [57]. Studies have shown that the surface coagulants of exosomes derived from platelets in circulation are 50–100 times more than activated platelets, and there are binding sites of coagulant promoters [58]. Therefore, platelet-derived exosomes can accelerate wound hemostasis.
In addition, platelet-derived exosomes are rich in platelet-derived growth factors (PDGF), fibroblast growth factor (FGF), transforming growth factor-β (TGF-β), and vascular endothelial growth factor (VEGF) [59]. Therefore, platelet-derived exosomes can enhance tissue regeneration through paracrine mode [60], and platelet-rich plasma exosomes can promote the healing of diabetic chronic skin wounds by upregulating collagen synthesis, inducing the proliferation and migration of endothelial cells and fibroblasts, and angiogenesis [61,62]. These studies highlight the role of platelet-derived exosomes in skin and vascular regeneration.
In conclusion, platelet-derived exosomes have been used in the field of skin wound healing. In addition, platelet-derived exosomes do not have nuclei, which can avoid the occurrence of teratogenesis. Currently, the downstream industrial manufacturing process, pathogen safety measures, quality control and release standards of platelet-derived exosomes are under development [57], which are worthy of further research.
Exosomes in inflammation phase
The wound healing process begins with an inflammatory response to remove debris and bacteria from the wound site. During this phase, immune cells and other cell types secrete pro-inflammatory cytokines, such as tumor necrosis factor-alpha (TNF-α), interleukin-1 (IL-1), and interleukin-6 (IL-6). Exosomes released by various cells can also contain these cytokines and deliver them to target cells, amplifying the inflammatory response.
In addition, exosomes can regulate immune responses by influencing gene expression and signaling pathways in recipient cells [29,48], and mediate the transformation of M1-proinflammatory phenotype to M2-anti-inflammatory phenotype to accelerate wound healing [63-65]. Cheng et al. has found that exosome PD-L1 can inhibit cytokine production of CD8+ T cells and suppressed CD8+ T cell numbers in spleen and peripheral lymph nodes to promote tissue repair [66]. Macrophage-derived exosomes can induce the reprogramming of macrophages from Ml to M2, which not only enhances the anti-inflammatory ability, but also accelerates the migration of fibroblasts and the tubular forming ability of vascular endothelial cells, thus promoting wound healing [63,67].
Exosomes in proliferation phase
During this phase, cell proliferation and tissue remodeling occur. Exosomes released from various cell types can carry cytokines such as transforming growth factor-beta (TGF-β), fibroblast growth factor (FGF), and vascular endothelial growth factor (VEGF). These cytokines play an essential role in promoting cell proliferation, angiogenesis (formation of new blood vessels), and collagen synthesis. Mesenchymal stem cells derived exosomes (MSC-Exos) cultured from neonatal serum demonstrated superior therapeutic efficacy in skin wound healing by targeting endothelial cells through the regulation of the AKT/eNOS signaling pathway to enhance angiogenesis [68]. Additional studies have shown that human MSC-Exos promote wound healing by stimulating angiogenesis [69,70]. Therefore, exosomes from different cell origins have a positive effect on skin healing.
Exosomes also can promote the proliferation and migration of various cell types involved in wound healing, including keratinocytes, fibroblasts, and endothelial cells [65,71]. This can accelerate the formation of new tissue and promote re-epithelialization of the wound. Studies have shown that exosomes derived from human adipose stem cells promote the proliferation and migration of human dermal fibroblasts in a dose-dependent manner and by inducing the expression of genes involved in dermal cell proliferation [49]. MSC-Exo promotes the growth and migration of diabetic wound fibroblasts and the formation of blood vessels to promote wound healing through AKT, ERK1/2, STAT3 and other signaling pathways [72], and it can enhance the proliferation and migration of keratinocytes and fibroblasts by regulating their biological characteristics, thus promoting the healing of skin wounds [69].
Exosomes in remodeling phase
The final phase of wound healing involves tissue remodeling. Exosomes can deliver cytokines that modulate matrix metalloproteinases (MMPs) to regulate the degradation and remodeling of the extracellular matrix.
Collagen synthesis plays an important role in wound remodeling. Exosomes can stimulate the synthesis of collagen, a major component of the extracellular matrix [71,73,74], which provides structural support for the healing tissue. Human adipose MSC-Exo enhance collagen synthesis by downregulating MMP3 while promoting skin barrier function recovery [73]. Exosomes released from human pluripotent stem cell derived MSCs promote skin wound healing through type I and III collagen synthesis and angiogenesis [74]. In addition, fibroblast-derived exosomes can increase collagen synthesis, migration, and vascular endothelial formation in injured skin to promote wound healing [75].
Exosomes act as mediators of intercellular communication by transferring their cargo of cytokines and other bioactive molecules to target cells. This process allows cells involved in wound healing to communicate with each other, promoting a coordinated and regulated healing response. This evidence highlights the role of exosomes in various stages of wound healing. It is important to note that the exact composition and functions of exosomes in wound healing are still an active area of research, and our understanding of their role in different stages of tissue repair is still evolving. In addition, different wound types and individual patient factors may influence the specific cytokines carried by exosomes and their overall impact on wound healing.
Comparison of stem cell therapy and exosome therapy
In general, both exosome therapy and stem cell therapy have the potential for treating skin injury, and the choice of therapy may depend on the severity of the injury, the therapeutic goal, and the patient’s individual circumstances. Exosome therapy for acute skin injury has several potential advantages and disadvantages compared to stem cell therapy (Table 1), as follows.
Table 1.
Comparison of stem cell therapy and exosome therapy
Stem cell therapy | Exosome therapy | |
---|---|---|
Advantages | Extensively studied | No ethical concerns for ESCs/iPSCs |
Cell differentiation capacity | Less invasive | |
Self-renewal | Fewer safety concerns | |
Longer-lasting effect | No need for genetic manipulation or expansion | |
Storage and transportation of exosome more easily | ||
Disadvantages | Time-consuming, expensive | Shorter half-life |
Safety concerns | Require multiple doses or sustained release formulations | |
Ethical concerns for iPSCs/ESCs | Failure to replicate the regenerative potential of stem cells in severe tissue damage | |
Risk of immune rejection |
Abbreviations: ESCs, embryonic stem cells; iPSCs, induced pluripotent stem cells.
Advantages of exosome therapy
Exosomes have regenerative potential and can promote tissue regeneration and healing in the wound bed, but less invasive compared to stem cell therapy that requests wound cleaning procedures. Injection of exosomes into the wound bed makes it an attractive option for patients who may not be suitable for more invasive treatments.
Importantly, exosome therapy avoids the ethical concerns associated with the use of embryonic stem cells or induced pluripotent stem cells. In addition, there is a reduced risk of immune rejection compared to other treatments as exosomes are derived from the patient’s own cells or from a compatible donor [76]. Furthermore, exosome therapy is generally considered to be safe, with minimal side effects reported in clinical trials to date.
Exosome can be isolated and purified from various sources, including mesenchymal stem cells, without the need for genetic manipulation or expansion. Exosome can be stored and transported more easily than living cells, allowing for more flexible treatment options [76].
Limitations of exosome therapy
Exosome therapy is a relatively new field, and further research is needed to determine the optimal dose, delivery method, and therapeutic potential of exosomes for different types of chronic skin injury. Exosomes may have a shorter half-life than stem cells and may require multiple doses or sustained release formulations to achieve optimal therapeutic effects [77]. Exosomes may not be able to replicate the regenerative potential of stem cells, especially in cases of severe or extensive tissue damage. In addition, currently, the availability of exosomes for clinical use is limited, which may restrict the widespread use of exosome therapy [78]. Furthermore, there is currently no standardized protocol for producing, isolating, and characterizing exosomes, which can lead to variability in the quality and effectiveness of exosome therapy [42,78]. Finally, exosome therapy can be expensive, which may limit access for some patients.
Advantages of stem cell therapy
Stem cell therapy has been extensively studied and has demonstrated a high potential for tissue regeneration and repair in preclinical and clinical studies. Stem cell can differentiate into multiple cell types and have the ability to self-renew [79], making them a versatile tool for tissue engineering and regenerative medicine [80]. Stem cell therapy may have a longer-lasting effect compared to exosome therapy.
Disadvantages of stem cell therapy
Stem cell therapy requires the isolation, culture, and expansion of living cells, which can be time-consuming, expensive, and may have safety concerns [81,82]. Stem cell therapy may have ethical concerns associated with the use of embryonic stem cells or induced pluripotent stem cells. In addition, stem cell therapy may have a risk of immune rejection, requiring immunosuppressive therapy in some cases [81,82].
Optimization of exosome therapy for skin acute wound healing
Exosome therapy has shown potential for promoting skin regeneration and reducing inflammation in various models of skin injury. We proposed the strategies to optimize adult stem cell therapy to enhance greatly for tissue regeneration [80]. Similarly, exosomes are a promising therapeutic tool, exosome optimization can better promote tissue regeneration and accelerate wound healing, but optimizing their yield and quality is crucial to the success in the improvement of their therapeutic efficacy and makes exosomes a more viable option for clinical use (Table 2).
Table 2.
Optimization of the yield and quality of exosomes as well as exosome therapy
Optimizing their yield | Optimizing their quality | Optimizing their therapy |
---|---|---|
Cell source | Purity | Timing |
Culture conditions | Cargo loading | Dose |
Isolation methods | Targeting | Frequency |
Identification | Characterization | Interval |
Storage conditions | Stability | Route of administration |
First, optimizing the yield of exosomes for therapy requires careful consideration of the cell source, culture conditions, isolation methods, characterization, and storage conditions (Fig. 3). By optimizing these factors, researchers can improve the yield and quality of exosomes and enhance their therapeutic potential.
Fig. 3.
Optimization of exosomes yield and therapy for skin acute wound healing, including optimization of exosome sources, culture conditions, storage conditions, isolation, genetic engineering and optimal does, timing, frequency, administration routine of exosomes.
Optimizing exosomes yield
Selection of exosome sources
The yield and quality of exosomes can vary depending on the cell type used for their isolation. Exosomes can be derived from blood, urine and a variety of cell types [28], including stem cells [83], fibroblasts [84], immune cells [85], etc. Different cell types produce exosomes with different cargoes, so it is essential to select a cell type that can secrete exosomes with the desired therapeutic payload. Stem cells derived exosomes (SC-Exos) include adipose-derived exosomes (AD-Exos), human umbilical mesenchymal stem cells derived exosomes (hUMSC-Exos), bone mesenchymal stem cells derived exosomes (BMSC-Exos), etc. Hence, in addition to containing abundant biomolecules of donor cells, such as RNA, DNA, nucleic acid, lipids, metabolites, and cytosolic, SCExos also contain plentiful growth factors, such as TGF-β, fibroblast growth factor, vascular endothelial growth factor, and neurotrophic factor, which can deliver a large number of functional biological molecules to the recipient cells to do their job. For example, human AD-Exos accelerate the healing of full-layer skin wounds in mice by reducing macrophage infiltration and pro-inflammatory cytokine secretion and increasing the formation and proliferation of new blood vessels [86,87]. Human amniotic fluid derived exosomes can accelerate the wound healing in the full skin wound rats, improve the regeneration of hair follicles, nerves, and blood vessels, and the natural distribution of collagen, and increase the proliferation of skin cells, as well as inhibit the TGF-β signaling pathway by targeting TGF-β receptors to reduce the formation of fibrosis scar [88]. Similarly, epidermal stem cells derived exosome specific microRNA can inhibit the activity of TGF-β1 and its downstream genes to promote wound healing and reduce scar formation [89]. Xu et al. summarized the therapeutic effects of SC-Exos in wound healing, indicating that SC-Exos are small size, high efficiency, low immune repulsion, and have more biological functions, which have significant advantages in promoting wound healing [90]. Recent research suggested that skin cells can release extracellular vehicles (EVs) to regulate intercellular skin communication [91]. Wang et al. described the therapeutic role of exosomes derived from specific skin cells in skin wound healing, and found that exosomes derived from epidermal keratinocytes, epidermal stem cells, and dermal fibroblasts play a unique role in promoting skin wound repair and can reflect cross-talk between cells in the wound healing environment, but the potential risks and more still need to be clarified [91]. Inflammation plays an important role in the process of wound healing. Macrophages are the main immune cells in the process of wound healing and play a key regulatory role in the inflammatory and proliferative stages of skin wound repair. Studies have shown that M2-macrophage-derived exosomes promote angiogenesis in vitro by targeting PTEN via miR-21 to activate the AKT/mTOS signaling pathway, and significantly promote angiogenesis, reduce scar formation, and accelerate wound healing in a mouse full-layer skin injury model [92].
In sum, exosomes from different cell sources have been proven to promote and accelerate wound healing during acute skin wound healing, but most of the current research is still focused on exosomes from stem cells, and some of the studies have entered clinical trials. Although skin- or immune-cells-derived exosomes may promote acute skin wound healing, the underlying mechanisms and clinical applications require further investigation. Therefore, SC-Exos show greater advantages and clinical applications in the treatment of acute skin wound healing.
Optimizing cell culture conditions
The culture conditions of the cells used for exosome production can also influence the yield and quality of exosomes. Optimizing cell culture conditions, such as pH, oxygen concentration, and nutrient availability, can enhance exosome production.
Cell starvation
It has been shown that depletion of the culture conditions required for cell growth can promote exosome biogenesis. Oxygen, glucose, and serum deprivation are common starvation methods that can increase exosome biogenesis. Serum deprivation of MSC-Exos increased 22 times in the delivery efficiency of exosomes compared with control [93]. In addition, Jeremy et al. suggested that serum has an inhibitory effect on exosomes production, and serum-free Opti-MEM is the optimal cell culture media for exosomes production [94]. Furthermore, hypoxia also effect exosome biogenesis. For instance, hypoxic can stimulate the biogenesis of exosomes derived from adipose and stem cells, etc. Gonzalez-King et al. overexpressed hypoxia-inducing factor-1a (HIF-1a) in MSC by transduction, and overexpressed HIF-1a stimulated stem cells to produce more exosomes [95]. Stem cells are more bioactive and can secrete more exosomes when cultured in hypoxia environment [96]. Overall, serum-free Opti-MEM promotes exosome biogenesis due to the combination of physical, molecular, and transcriptional effects. In the future, optimizing its production can be developed to exert more therapeutic effects of exosomes.
pH and others
Exosomes are also affected by pH. Researches have suggested that the loss of exosome concentration at pH 4 and pH 10 was more than at pH 7; moreover, exosomes stored at pH 4 degrade faster than at pH 10 [97]. Moreover, studies have observed that MSC-EV production depends on cell seeding density, per cell between MSCs seeded at 1E2 or 1E4 cells/cm2 for P2, P3, P4, and P5 MSCs measuring ~126-fold, ~152-fold, ~201-fold, and ~126-fold, respectively; and increased EV collection frequency can augment EV production. When the inoculation density was fixed, increasing the collection frequency could increase the total exosomes by 2.0–2.4 fold [98].
Optimizing the isolation of exosomes
At present, the traditional separation and purification methods of exosomes have some shortcomings. There are several methods available for exosome isolation, including ultracentrifugation, size-exclusion chromatography, and polymer-based precipitation, etc., and we showed the pros and cons of different separation methods (Table 3).
Table 3.
Exosome isolation techniques and their comparison
Isolation methods | Advantages | Limitations | Sample matrix |
---|---|---|---|
UC | Golden standard | Time consuming | Cell culture medium Serum Urine |
Easily required | High requirements for equipment | Serum | |
Simplicity of operator | Decreased in biological activity | Urine | |
Limited mass production | |||
Differential centrifugation | Rapid | Heterogeneity | Cell culture medium |
Mass production | Easy to drain | Serum | |
Urine | |||
UF | Rapid | Protein contamination | Cell culture medium |
Simplicity of operation | Exosomes are damaged | Serum | |
Urine | |||
Cerebrospinal fluid | |||
Immuno-isolation | Rapid | Additional separation and purification are required | Cell culture medium |
High purity and specificity | Not suitable for mass generation | Serum | |
Polymer precipitation | Mass production | Decreased purity | Cell culture medium |
Simplicity of operator | Protein contamination | ||
Expensive kit | |||
SEC | High purity | Co-separation of proteins with similar diameters | Cell culture medium |
Commercial kits available | Not satisfy the downstream application | Serum | |
High productivity | Urine | ||
Cerebrospinal fluid | |||
Saliva, etc. | |||
AF4 | High purity | High requirements for equipment and personnel | Cell culture medium |
High efficiency | Limited mass production | Serum | |
Identify subset | Urine | ||
Ion exchange | Simplicity of operator | Unknown | Cell culture medium |
High purityy | |||
Microfluidic technology | Rapid | Not suitable for mass generation | Cell culture medium |
Save samples and reagents | Methods need to be further standardized | Serum | |
High purity and efficiency |
Abbreviations: UC, ultracentrifugation; UF, ultrafiltration; SEC, size-exclusion chromatography; AF4, asymmetric flow field-flow fractionation.
Traditional methods are density-based capture using ultra or differential centrifugation [99], immunoaffinity based capture using specific membrane proteins (e.g., CD9, CD63, and CD31) [100], and size based capture using volume exclusion chromatography, such as ultracentrifugation (UC), size-exclusion chromatography (SEC), ultrafiltration (UF), precipitation, etc. Although exosomes separation technology is constantly updated, there are some shortcomings in the separation and purification process of exosomes, such as low purity, contamination, coexistence of immunoglobulins, vesicle destruction and heterogeneity, which affect the biological function of exosomes.
Therefore, some studies have optimized SEC, using longer columns (56 mm to 222 mm), reduced protein and immunoglobulin contamination by 90% without affecting EV particle size distribution and number, and improved EV purity [101]. In addition, 2D SEC (including 2 columns) can be used for further isolation of exosome subpopulations, which has been achieved in the isolation of urine exosomes [102]. However, the separation of EV subpopulations by SEC separation techniques above is limited, which hinders characterization of their molecular composition and biogenesis. Recent studies describing new isolation strategies for exosomes show great progress in harvesting high quality EVs with increasingly convenient operating steps and high-level devices. For example, the methods of microfluidics, asymmetric flow field-flow fractionation (AF4), ion charge and combined multi-step methods are becoming increasingly popular [103-105].
Flow field-flow fractionation
Flow field-flow fractionation (FFF) is a size-based exosome separation technique, of which asymmetric flow field-flow fractionation (AF4) is most common in FFFS. David et al. [106] successfully isolated and identified EV subsets with asymmetric flow field-flow fractionation (AF4), thus identifying their heterogeneity, and compared other isolate methods, AF4 can obtain high purity, efficiency and integrity, but the technology has high requirements for equipment and personnel and fail to dispose large volume sample [103].
Microfluidics
Microfluidics, a separation technology that can integrate separation and detection functions into a single chip, is widely used for the separation and detection of micro-nanoparticles, biomarkers, cells, and proteins. The separation of exosomes by microfluidic chip technology is mainly related to the size, density, and surface antigen of exosomes. Current microfluid-based exosome separation techniques include capture, filtration, magnetic separation, and acoustic fluid separation [104]. (1) Capture: The capture is mainly to modify the antibodies on the inner wall of the microchannel, and anti-CD63 and CD81 antibodies are often used for exosome capture [107]. Other studies have shown that it can be optimized, such as filling microchannels with proteins, novel GO/polydopamine, etc., which can improve the separation of exosomes [108]. (2) Filtration: Microfluid-based filtration is similar to traditional ultrafiltration in that the microfluid-based filtration chip is driven by direct current to separate exosomes from plasma. Microfluid-based filtration can achieve high throughput, but its purity is limited. (3) Magnetic: Magnetic separation begins with the capture of exosomes by magnetic nanoparticles modified with antibodies, and then magnetically separates the resulting exosomes [109], mainly used for cell separation, can achieve high specificity, high throughput, and so on [104]. In addition, compared with UC, Magnetic bead-mediated selective adsorption strategy can obtain more than 2 times the yield and relatively stable purity, could meet the requirements of various EV-associated downstream applications [110]. (4) Acoustic fluid: Acoustic fluid separation is the separation of exosomes of different sizes by acoustic force in the environment of sound field, which is often used in cells culture [104].
Ion exchange
Ion exchange is a charge-based separation technique in exosome separation mainly through the interaction between a negatively charged EV membrane component and a positively charged functional group or cation anion exchanger. The team of Hiroshi Shiku used an anion-exchange method to obtain high-performance exosomes, and showed that it is suitable for the large-scale separation of bioactive exosomes and micro vesicle (MV)-like EVs as a cargo for dangerous nucleic acids at high-purity [111]. The all-in-one nanowire-integrated well plate assay system can capture and analyze EVs based on the surface charge of vesicle membranes, especially in EV-mediated tumor-microenvironment crosstalk [112,113]. But ion exchange is challenging for exosome separation in blood or plasma, which contains other charged materials.
In addition, the combined exosome isolation technology is recommended because it clearly outperformed single exosome isolation method to obtain higher purity and quantity, such as UC combination with microfiltration, SEC combined low-speed centrifugal method, combination of iodixanol density gradient ultracentrifugation and bind-elute chromatography [114-116].
Overall, the method of isolation and purification can affect the purity and quality of the exosomes, and it is important to use standardized and validated methods to ensure consistent and high-quality exosomes. Each method has its own advantages and limitations, and choosing the appropriate isolation method can optimize the yield and purity of exosomes.
Selection of exosome identification methods
Proper identification of the exosomes is crucial to ensure their quality and efficacy. Techniques such as nanoparticle tracking analysis (NTA), electron microscopy (EM), flow cytometry (FCM), and western blotting (WB) can be used to characterize the size, morphology, and concentration of exosomes. EM can identify the morphology of exosomes; ANT can track the distribution of exosomes; FCM and WB can identify molecular phenotyping with high sensitivity and specificity [117]. However, each identification method has certain disadvantages, such as: transmission electron microscopy destroys the morphology of exosomes in the identification process; NTA shows poor sensitivity, low efficiency; and WB takes a long time [118]. Therefore, combination of the four methods can better characterize the characteristics of exosomes and improve the quality.
Engineered exosomes for wound healing
Exosomes are promising therapeutics for skin injury due to their ability to modulate cellular processes such as inflammation, proliferation, and differentiation. However, their efficacy may be limited by factors such as low bioavailability, poor targeting, and short half-life in vivo. To address these limitations, researchers are exploring the use of engineered exosomes to improve therapeutic outcomes for skin injury, and we summarize some methods for optimizing exosome quality, as follows (Fig. 4, Table 4).
Fig. 4.
Engineered exosomes for skin wound healing, including surface and genetic engineering, precondition and loaded.
Table 4.
Engineered exosomes better improve skin wound healing
Methods | Mechanism | Outcomes | References |
---|---|---|---|
Surface engineering | Modified with the surface antibodies |
Modulate inflammatory response and promote skin tissue repair in immune cells, keratinocytes or fibroblasts | [44,119,120] |
Genetic engineering | Modify donor cells cargo | Overexpressed or loaded with growth factors or miRNA can promote cell proliferation and differentiation, angiogenesis, and extracellular matrix formation | [121-128] |
Hydrogels combined with exosomes | Exosomes are transported to the site of injury by hydrogels | Loaded Exos on hydrogel facilitate cell proliferation and accelerate collagen synthesis to promote wound healing | [129,130] |
Preconditioning | Preconditioning donor cells | ATV-pretreated MSC-Exos upregulating miR-221-3p and AKT/eNOS pathway to accelerate diabetic wound repair | [131-137] |
Fe3O4 nanoparticles preconditioning BMSc-Exos upregulating miR-21-5 to promote wound healing | |||
HGF-ADSC-Exos promote wound healing by increasing neovascularization |
Abbreviations: Exos, Exosomes; ATV, atorvastatin; MSC-Exos, mesenchymal stem cells derived exosomes; BMSC-Exos, bone mesenchymal stem cells derived exosomes; HGF-ASC-Exos, exosomes from hepatocyte growth factor-modified human adipose mesenchymal stem cells.
Surface engineering
One approach to engineering exosomes for skin injury is to modify their surface with targeting ligands or antibodies that can enhance their uptake by specific cell types in the skin. For example, exosomes can be modified with antibodies that target specific markers on immune cells, such as macrophages, to modulate the inflammatory response and promote tissue repair [119]. Similarly, exosomes can be modified with peptides or proteins that target specific cell types in the skin, such as keratinocytes or fibroblasts, to promote wound healing and tissue regeneration [44,120].
Genetic engineering
Engineered exosomes for skin wound are transfected into donor cells with specific proteins, growth factors, or small molecules to modify their cargo to enhance their therapeutic efficacy. Studies have shown that exosome-based genetic modification has higher sensitivity and specificity for the treatment of certain diseases and can improve the efficiency of drug delivery. For example, exosomes can be overexpressed or loaded with growth factors such as TGF-β or PDGF, which can promote cell proliferation and differentiation, angiogenesis, and extracellular matrix formation [121,122]. Exosomes can also be loaded with small molecules such as microRNAs or other non-coding RNAs that can modulate gene expression and promote tissue regeneration [123-126]. For instance, Chen et al. have found that AD-Exos can highly express miR-21 in process of mouse full layer wound healing, miR-21 plasmid was used to overexpress miR-21 by transfection of HaCaT cells (representative of keratinocytes in vitro), and miR-21 can promote the migration and proliferation of the keratinocytes via PI3K/AKT signal pathway to improve wound healing [127]. However, the modification of exosome genes is mainly used in cancer research, and more investment is needed to research skin wound healing in the future.
Hydrogels combined with exosomes
Researchers are exploring the use of exosome-based hydrogels or scaffolds for skin injury [138]. These hydrogels or scaffolds can be engineered to release exosomes in a controlled manner over time, providing a sustained therapeutic effect [67]. Furthermore, these hydrogels or scaffolds can be designed to mimic the structure and function of the extracellular matrix in the skin, providing a favorable environment for tissue regeneration. In addition, recent studies have shown that a YARA peptide based equipping technique can enhance the cargo carrying potential of exosomes. YARA peptide covalently conjugated with mammalian miR-21-5p to form the conjugate YARA-miR-21-5p and incubated with mesenchymal stem cells-derived exosomes, which significantly enhanced the proliferation, migration of fibroblasts in human and mouse wound healing [129]. Zhu et al. has suggested that loaded human umbilical mesenchymal stem cell (UMSC) derived exosomes on hydrogel can facilitate cells proliferation and accelerate collagen synthesis to promote wound healing through upregulating oxidative phosphorylation-related proteins in mitochondria [130]. Therefore, loading exosomes on hydrogels may be a potential treatment strategy for wound healing.
Preconditioning
Donor cells preconditioned using drugs, factor, and hypoxia, etc. can accelerate skin wound healing. For example, exosomes derived from atorvastatin-pretreated mesenchymal stem cell accelerate diabetic wound repair by enhancing biological function of endothelial cells and angiogenesis via upregulating miR-221-3p and AKT/eNOS pathway [131]. BMSC-Exos preconditioned with Fe3O4 nanoparticles and a static magnetic field can improve angiogenesis and fibroblast function by upregulating miR-21-5 to promote wound healing [132]. In addition, growth factor-stimulated exosomes also can facilitate skin wound healing. For example, exosomes from hepatocyte growth factor-modified human adipose mesenchymal stem cells can reduce exudate in early stage, and significantly promote wound healing by increasing neovascularization in wound tissue, and the mechanism may be related to activate PI3K/Akt signaling pathway [137].
Electrical stimulation
Electrical stimulation (ES) is a non-invasive operation that can stimulate cell differentiation, migration, proliferation, and so on. ES as a physical stimulator can regulate cellular behavior for applying to regenerative medicine and tissue engineering [139]. Studies have found that ES can increase EV secretion and cargo loading [140,141]. However, different cells receive different electric fields, which can change the size, concentration, and cargo of exosomes. In addition, the application of electric field can also impair the normal physiological function of cells. Therefore, the intensity of the electric field needs to be controlled according to different cell types. In the future, the role of electrical stimulation in promoting skin wound healing needs to be further explored.
Ultraviolet B
Ultraviolet B (UVB) can stimulate the generation of exosomes by increasing HaCaT exosome secretion in dose- and time-dependent manners, and promote the expression of miRNA, especially miR-126, in skin cells-derived exosomes in diabetic mice [142]. However, UVB does not always promote the treatment of skin diseases. It was found that UVB could stimulate the secretion of exosome miR-769-5p in human skin fibroblasts, but miR-769-5p could aggravate the oxidative damage of human skin fibroblasts and accelerate cell apoptosis, while inhibition of miR-769-5p can reverse those effects [143]. Therefore, more experiments are needed to further confirm the effect of UVB stimulation on exosomes.
In addition, exosomes secreted from hypoxic-preconditioned mesenchymal stem cells show great therapeutic potential in the treatment of diseases [144]. Hypoxic microenvironment plays a negative role in wound healing, and prolonged hypoxia can also lead to delayed wound healing. Hypoxic adipose-derived exosomes can regulate the function of cell metabolism, growth and transformation factors by upregulating and downregulating the expression of miRNA related to skin healing, and promote wound healing and inhibit inflammation through PI3K/AKT signaling pathway [133]; and compared with MSC-Exo, hypoxic MSc-Exo has greater potential to promote angiogenesis, cell proliferation and differentiation [134]. Besides, BMSC-Exos facilitates human umbilical vein endothelial cells proliferation, migration and angiogenesis by increasing the expression of high mobility box 1 protein to stimulate JNK signaling pathway [135]. Exosomes derived from hypoxic umbilical cord mesenchymal stem cells could induce the expression of miR-125b, which targeted inhibition of the expression of tumor protein p53 inducible nuclear protein 1 for increasing the proliferation and migration of endothelial cells, and inhibiting cell apoptosis to promote wound healing [136]. Therefore, preconditioning stem cells-derived exosomes may open a new strategy for skin wound treatment.
Overall, engineering exosomes for skin injury has the potential to improve therapeutic outcomes by enhancing their bioavailability, targeting, and therapeutic effects. However, further research is needed to optimize these strategies and evaluate their safety and efficacy in preclinical and clinical settings.
Storage of exosomes
Exosomes can be sensitive to changes in their environment, such as temperature and pH levels. Therefore, it is important to optimize the storage conditions to ensure their stability and efficacy over time. Currently, the storage of exosomes mainly contains freezing, freeze-drying, and spray-drying [145,146]. Studies have shown that more quantity of exosomes were stored at 4 °C for 24 h than at −80 °C, but the best long-storage method for exosomes is frozen storage at −80 °C [97]; and minimizing the storage time can avoid structural changes and bioactivity degradation of exosomes [145]. In addition, a certain concentration of antifreeze can be added in the freezing process to extend the shelf life of exosomes. The study found that storing exosomes in disaccharide antifreeze solution trehalose is the best antifreezing method [147,148].
In short, by optimizing the factors affecting the quantity and quality, therapeutic efficacy of exosomes can be improved as a more viable option for clinical use.
Optimizing exosome therapy for skin wound healing
Although exosomes show great advantages in the treatment of skin wounds, the dosage, mode of administration, safety and efficacy of exosomes should be considered (Fig. 3). The optimal dose and route of delivery of exosomes vary depending on the severity and location of the skin injury. Exosomes can be delivered topically, injected directly into the injured area, or administered systemically. The timing of exosome therapy may also be important, as early treatment can be more effective in promoting skin regeneration.
Doses of exosome therapy
The optimal dose of exosome therapy for wound healing is still an area of ongoing research, and there is no consensus on the ideal dose. The dose of exosome therapy may depend on various factors such as the type and severity of the wound, the characteristics of the exosomes used, and the mode of administration.
A systematic review has shown that various doses of exosomes have been used to treat skin injury, ranging from 1 μg to 200 μg per treatment, depending on the study design and animal model used [27]. The higher doses of exosomes may result in improved therapeutic outcomes, while lower doses may be sufficient for achieving therapeutic effects [149]. For example, MSC-Exos eye drops were formulated at 0.5 × 1010, 2.5 × 1010, or 12.5 × 1010 particles/mL in 5 μL per drop. When administrated twice a day for 7 days, the 2.5 × 1010 particles/mL formulation was the most optimal and effective dose for the mouses with dry eyes [150]. On the contrary, human corneal mesenchymal stem cell-derived exosomes with 1.0 × 108 exosome/mL treated a monolayer of confluent human corneal endothelial cells (HCECs) were scratched for 24 h can accelerate the re-epithelialization of HCECs, while 105 and 107 exosomes/mL has a slight dose dependent effect, but this was not statistically significant [151]. In clinical studies, the optimal dosage of exosomes for skin injury has not yet been established, as these studies are still in the early stages of development. However, some clinical trials have used doses ranging from 1011 to 1013 exosomes in antitumor [152]. In a clinical trial of human umbilical cord mesenchymal stem cell-derived exosomes (UCSC-Exos) for 28 patients with graft-versus-host disease (GVHD)-associated dry eye, UCSC-Exos at a concentration of 10 μg/50 μL and administrated 4 times a day per eye for 2 weeks can significantly remission dry eye symptoms and improve quality of life [150].
In the end, the optimal dose of exosome therapy for wound healing may depend on the specific wound and the treatment protocol. Further research is needed to determine the best dose and dosing schedule of exosome therapy for different types of wounds.
Timing of exosome therapy
Exosome therapy for wound healing is an emerging field of research, and there is still much to learn about the optimal timing of exosome therapy. However, based on current research, the timing of exosome therapy may depend on the type and severity of the wound. Similarly, to stem cell therapy, exosome therapy may be most effective when administered during the early stages of wound healing, when inflammation and angiogenesis are occurring. At this stage, exosomes may help to promote the recruitment of immune cells and the formation of new blood vessels, which are important steps in the wound healing process.
However, it is also possible that exosome therapy could be beneficial during the later stages of wound healing, when tissue remodeling and maturation are occurring. Exosomes may help to promote the production of extracellular matrix components and the organization of new tissue, which are critical for the proper healing of wounds. Thus, the optimal timing of exosome therapy for wound healing depends on the specific characteristics of the wound and the underlying tissue damage. Further research is needed to determine the best timing and dosing of exosome therapy for different types of wounds.
The timing of exosome therapy may depend on various factors such as the type and severity of the wound, the characteristics of the exosomes used, and the mode of administration. In general, the timing of exosome therapy may be influenced by the phases of wound healing, which include inflammation, proliferation, and remodeling. During the inflammation phase (early phase), which typically lasts up to 3–7 days after injury, exosomes may help to modulate the immune response and promote angiogenesis, which is the formation of new blood vessels. In the proliferation phase, which occurs after the inflammation phase and lasts for several weeks, exosomes may promote cell growth and tissue regeneration. In the remodeling phase, which can last for several months, exosomes may help to promote tissue remodeling and the organization of new tissue.
However, it is important to note that the optimal timing of exosome therapy for wound healing may vary depending on the specific wound and the treatment protocol. It is likely that further research will be needed to determine the best timing and dosing of exosome therapy for different types of wounds.
Administration of exosome therapy
The optimal administration of exosome therapy for wound healing may depend on various factors such as the type and location of the wound, the characteristics of the exosomes used, and the desired therapeutic effect. However, several administration routes have been studied for exosome therapy for wound healing (Table 5): (1) Local injection: Exosomes can be injected directly into the wound site or the surrounding tissue. This mode of administration can deliver exosomes directly to the site of injury, promoting local tissue repair and regeneration [153]. (2) Topical application: Exosomes can be applied topically to the surface of the wound or incorporated into a wound dressing. This mode of administration can promote healing of the wound surface and prevent infection [154]. (3) Systemic injection: Exosomes can be injected systemically, such as intravenously or intramuscularly. This mode of administration can deliver exosomes to the entire body, promoting general tissue repair and regeneration [87,155]. (4) Inhalation: Exosomes can be delivered to the lungs through inhalation. This mode of administration has been studied for pulmonary diseases but may also have potential for wound healing of respiratory tissues [156]. (5) Oral administration: Exosomes can be administered orally, such as in the form of a capsule or tablet [157]. Due to the poor oral bioavailability, further studies are needed to characterize the efficacy of this delivery method. However, recent studies have shown that milk exosomes can survive the highly acidic conditions in the stomach and degradation conditions in the gut. In addition, they can cross biological barriers to reach the target tissue. The ability of milk exosomes to cross the gastrointestinal barrier makes them a promising drug delivery vehicle for oral delivery [158].
Table 5.
Comparison of exosome administration
Administration | Diseases | Outcomes | References |
---|---|---|---|
Local injection (dose: 1 g/L) | Diabetic wound | Enhanced the biological functions of HUVECs in vitro; enhanced angiogenesis to accelerated diabetic wound healing in vivo. | [153] |
Topical application | Diabetic wound | Less inflammation, collagen deposition, and neovascularization to drive wound healing | [154] |
Systemic injection (dose: 200 μg) | Acute skin wound | Stimulate cell migration, proliferation and collagen synthesis to accelerate wound healing | [87,155] |
Inhalation (dose: 10 × 109 particles/kg, 30 min/day, successive 10 days) | Idiopathic pulmonary fibrosis | Reestablishing normal alveolar structure and decreasing both collagen accumulation and myofibroblast proliferation | [156] |
Oral administration (dose: 3 mg/day, successive 7 days) | Ulcerative colitis | Ameliorate mice colitis and accelerate colitis resolution via regulating the expression of the pro-inflammatory cytokines | [157] |
In summary, the above exosome delivery methods can better promote disease healing. However, the current study shows that inhalation is mainly used for lung diseases, and due to the low bioavailability of oral methods, more studies are needed to further confirm the effectiveness of promoting skin wound healing. Local application, local administration and systemic intravenous administration of exosomes all play a positive role in promoting skin wound healing, but studies have shown that the therapeutic effect of intravenous injection and local application is superior to that of local injection [87,155]. This may be related to the loss of exosomes during local injection, and in addition, local injection will inevitably interfere with the wound, thus interfering with wound healing. Intravenous exosomes may recruit injured areas via the membrane surface of receptors or adhesion molecules, targeting fibroblasts and promoting wound healing.
The optimal administration of exosome therapy for wound healing may depend on the specific characteristics of the wound and the exosomes used. However, intravenous injection and topical application may be particularly effective for promoting wound healing, as they allow direct delivery of exosomes to the site of injury. Further research is needed to determine the most effective administration route for exosome therapy for wound healing.
Frequencies of exosome therapy
The optimal frequency of exosome therapy for wound healing may depend on various factors such as the type and severity of the wound, the characteristics of the exosomes used, and the mode of administration. Frequencies of exosome therapy for wound healing, including: (1) Single administration: Exosome therapy can be given as a single dose, typically at the time of injury or after the initial wound healing phase. This mode of administration may be effective in promoting wound healing and tissue regeneration. (2) Repeated administration: Exosome therapy can be given multiple times, either at regular intervals or as needed depending on the wound healing progress. This mode of administration may be more effective in promoting sustained tissue repair and regeneration. (3) Continuous administration: Exosome therapy can be given continuously over an extended period of time, such as through a slow-release implant or infusion. This mode of administration may be effective in promoting long-term tissue repair and regeneration. Zhang et al. compared the administration frequency of ASC-Exos in skin wounds and found that increasing the administration frequency (3 times a day) could promote skin wound healing more than once a day and twice a day [87].
The optimal frequency of exosome therapy for wound healing may depend on the specific characteristics of the wound and the exosomes used. However, repeated administration may be particularly effective for promoting sustained tissue repair and regeneration, as it allows for ongoing delivery of exosomes to the site of injury. Further research is needed to determine the most effective frequency and dosing schedule of exosome therapy for wound healing.
Interval between repeated exosome therapies
The optimal interval between two exosome therapies for wound healing may depend on various factors such as the type and severity of the wound, the characteristics of the exosomes used, and the mode of administration. However, several general guidelines have been suggested based on existing studies.
For repeated administration of exosome therapy, the optimal interval can be vary depending on the desired therapeutic effect and the duration of exosome activity. If exosomes have a short half-life, more frequent administration is necessary to maintain their therapeutic effect. Exosome therapy has been administered at intervals ranging from daily to weekly [149]. Injections of ASC-Exos every three times one days for two weeks were effective in promoting full-skin wound healing [87]. Another study on diabetic mice with impaired wound healing found that topical application of ASC-exosomes every two days for two weeks was effective in promoting wound healing. However, the optimal interval between two exosome therapies for wound healing may vary depending on the specific characteristics of the wound and the exosomes used. Further research is needed to determine the most effective dosing schedule for exosome therapy for different types of wounds.
In summary, optimizing exosomes for therapy involves a comprehensive approach that includes isolation and purification, characterization, loading of therapeutic cargo, targeting, quality control, and scale-up. By optimizing these parameters, exosomes can be developed as effective and safe therapeutic agents for a wide range of diseases.
Safety and efficacy
The safety and efficacy of exosome therapy should be carefully evaluated in preclinical and clinical studies. Although local administration is safer and more bioavailable than systemic administration, which can reduce the influence on other cells of the body and avoid the influence of phagocytes and cyclic clearance in vivo, drug concentration is easily interfered by external factors. Moreover, whether local or systemic administration, different concentrations of administration will produce different biological effects [159,160]. Adverse effects such as immune reactions or unintended effects on other tissues or organs should be monitored, and the efficacy of exosome therapy should be assessed using objective measures such as wound healing time and scar formation. To optimize exosome therapy for skin injury, it is important to carefully consider the source of exosomes, isolation and purification methods, dosing and delivery, and safety and efficacy in preclinical and clinical studies. Further research is needed to fully evaluate the potential of exosome therapy for promoting skin regeneration and reducing inflammation in various types of skin injuries.
Exosome therapy used in other diseases
Exosome therapy has been widely studied in the management of other diseases, such as cancer, ischemia-induced diseases, and inflammatory-related diseases (Table 6).
Table 6.
Exosome therapy used in other diseases
Indication | Diseases | Exosome source | Effective molecules | Outcomes | References |
---|---|---|---|---|---|
Cancer | Glioma | Neutrophils | – | Loaded with doxorubicin, inhibit glioma progression via BBB transfer to the TME | [162] |
Colorectal cancer | 293T cells | miR-21↓, PTEN and hMSH2↑ | Decrease tumor proliferation and increase apoptosis to inhibit tumor growth | [164] | |
Gastric cancer | HEK293T cells culture media and plasma | miR-214↓ | Induce cell apoptosis, reduce proliferation and migration, and increase drug sensitivity | [165] | |
BC | Breast tumor cells | miRNA-16-5p↓, CD73↑ | BC-derived exosome SNHG16/miR-16-5p/SMAD5-regulatory axis potentiates TGF-β1/SMAD5 pathway activation, to induce CD73 expression in Vδ1 T cells | [168] | |
BC | CAF | miR-92, PD-L1↑ | Promotes apoptosis and impaired proliferation of T cells | [169] | |
Ischemia-induced injury or diseases | MI | hUSC | Cx43, Ki67, CD31, α-SMA, Vwf, TGF-β1, MMP-9, etc.↑ | Promote cell proliferation and angiogenesis, enhancing ejection fraction | [171] |
IS | IPAS | circSHOC2, miR-7670-3p, SIRT1↑ | Regulate autophagy and reduce neuronal apoptosis | [172] | |
IS | Lactobacillus plantarum | miR-101a-3p↑ | Inhibit neuron apoptosis to protect against ischemic brain injury through the micro RNA-101a-3p/c-Fos/TGF-β axis | [173] | |
Kidney I/R | HUVEC | NF-κb activity↓ | Decreased NF-κB activity, expression of pro-inflammatory cytokines and adhesion molecules | [174] | |
Inflammatory-related diseases | Atherosclerosis | HEK293T cells | miR-155 ↑ | Exosome-based delivery of the engineered IL-10 could alleviate the atherosclerosis in ApoE mice | [180] |
AA | M2 macrophage | Inflammatory cytokines ↓ | Ameliorated AA with a marked reduction of lung inflammation | [181] | |
TBI | Astrocyte | miR-873a-5p↑ | Inhibited LPS-induced microglial M1 phenotype transformation and inflammation through decreased phosphorylation of ERK and NF-κB p65 | [182] | |
Murine colitis | M2 macrophage | miR-590-3p, LATS1↑ | Reduces inflammatory signals and promotes epithelial regeneration | [183] | |
NASH | HLSCs | α-SMA, Col1a, Tgf-β1, TNF, IL-1β↓ | Display anti-fibrotic and anti-inflammatory effects to improve liver function | [186] | |
EAE | HUCB | IL-2, MMP-9, HSP-72↑ | Inhibited T cell proliferation and EAE development by regulating IL-2 signaling | [189] |
Abbreviations: BBB, blood-brain barrier; TME, tumour microenvironment; PTEN, phosphatase and tensin homolog; hMSH2, human DNA MutS homolog 2; BC, breast cancer; CAF, cancer-associated fibroblasts; hUSC, human umbilical cord mesenchymal stem cells; MI, myocardial infarction; IS, ischemic stroke; IPAS, ischemic-preconditioned astrocyte; I/R, ischemia-reperfusion; HUVEC, human umbilical vein endothelial cells; CIRI, cerebral ischemia-reperfusion injury; AA, allergic asthma; TBI, traumatic brain injury; NASH, non-alcoholic steatohepatitis; HLSCs, human liver stem cells; EAE, experimental autoimmune encephalomyelitis; HUCB, human umbilical cord blood.
Exosome therapy used in the treatment of cancer
Exosome therapy is an emerging approach in cancer treatment in skin or other tissues, which involves the use of exosomes or small extracellular vesicles released by cells [161]. These exosomes can carry various molecules, such as proteins, nucleic acids, and lipids, which can influence cellular behavior and signaling [43].
In cancer treatment, exosome therapy holds promise for several reasons. First, exosomes can be isolated from various cell sources, including immune cells, stem cells, and tumor cells, allowing for the potential delivery of therapeutic cargo to cancer cells [43]. Second, exosomes naturally have the ability to cross biological barriers, including the blood–brain barrier, which can enhance their accessibility to tumors located in challenging anatomical sites [162]. Third, exosomes can serve as mediators of intercellular communication, facilitating the transfer of information between cells and modulating tumor growth, immune response, and drug resistance [163].
Exosome-based therapies in cancer treatment can be categorized into two main strategies: therapeutic cargo delivery and immunomodulation. In therapeutic cargo delivery, exosomes can be loaded with anticancer agents, such as chemotherapeutic drugs or small interfering RNAs (siRNAs), and delivered to cancer cells to exert their effects. Studies have shown that loading 5-fluorouracil and miRNA-21 inhibitors into exosomes through electroporation can effectively promote cellular uptake, reduce miRNA-21 expression, decrease tumor proliferation, increase apoptosis, and reduce expression of PTEN and hMSH2 to inhibit tumor growth [164]. In addition, exosome delivering anti-miRNA-214 to gastric cancer cells can induce cell apoptosis, reduce proliferation and migration, and increase drug sensitivity [165]. Exosome can handle the payload of anti-tumor drugs, and can also be used as a carrier of genetic tools to increase the effectiveness of anti-tumor treatment. Exosome loaded siRNAs may solve the drawbacks of RNA enzyme degradation, tumor barrier, and off-target effects [166]. This approach can potentially enhance drug delivery specificity and reduce off-target toxicity to improve the effectiveness of treatment.
On the other hand, exosomes can also be utilized to modulate the immune response against cancer. They can be engineered to carry immune-stimulating molecules or tumor antigens to activate the immune system and promote antitumor immune responses [167]. Additionally, exosomes derived from immune cells can be modified to enhance their immunosuppressive properties, which may help counteract the immunosuppressive tumor microenvironment and promote better immune surveillance against cancer cells. Exosome transmits SNHG16 to induce the TGF-β/SMAD16 axis, reducing miRNA-16-5p and mediating CD73 overexpression on Treg cells, leading to immune suppression in breast cancer [168]. Study has shown that miRNA-92 mediated interaction between LATS2 and YAP1 in breast cancer cells is facilitated by the exosome contained in cancer-associated fibroblasts. YAP1 is translocated to the cell nucleus and binds to the PD-L1 promoter to enhance its expression, leading to T cell apoptosis and decreased proliferation [169].
Exosome therapy used in ischemia-induced injury or diseases
Ischemia refers to a condition where there is inadequate blood supply to the skin, other tissues or organs, resulting in reduced oxygen and nutrient availability. This can lead to skin ischemia or tissue damage and various diseases, such as ischemic stroke, myocardial infarction, renal failure, liver dysfunction, severe acute respiratory syndrome and peripheral artery disease. Exosome therapy has shown promise in the context of ischemia-induced injury or diseases [170].
In preclinical studies, administration of MSC-derived exosomes has demonstrated beneficial effects in ischemia-induced injury models. For example, Zou et al. has found that hydrogel USC-Exos can improve cardiac function and promote blood vessel formation in myocardial infarction models by promoting cell proliferation and angiogenesis [171]. Chen et al. discovered that the expression of circSHOC2, an exosome derived from astrocytes, was significantly upregulated, which can promote neuroprotection and enhance functional recovery via the miR-7670-3p/SIRT1 axis to regulate autophagy and reduce neuronal apoptosis [172]. In addition to mammalian-derived exosomes showing a positive therapeutic effect on ischemic diseases, Lactobacillus plantarum-derived exosome can inhibit neuron apoptosis to protect against ischemic brain injury through the microRNA-101a-3p/c-Fos/TGF-β axis [173]. Furthermore, exosomes have emerged as a promising delivery tool for treating ischemic-related diseases. For example, exosome can delivery super-repressor IκBα via decreasing NF-κB activity and reducing apoptosis to ameliorates kidney ischemia-reperfusion injury [174].
Exosome therapy used in chronic inflammatory-related diseases
Chronic inflammatory conditions are characterized by persistent inflammation in the affected tissues or organs, leading to tissue damage and dysfunction. Examples of such skin disease include eczema, seborrheic dermatitis, and psoriasis. Other diseases include rheumatoid arthritis, inflammatory bowel disease, and asthma. Some studies have reported that exosome therapy holds promise for the treatment of chronic inflammatory-related diseases in skin or other tissues, including inflammatory bowel disease, neuroinflammation, osteoarthritis, atherosclerosis, allergic asthma, etc. [36,175-181].
Exosomes derived from immune cells, such as dendritic cells and macrophages, can influence the immune response by delivering specific molecules that either promote or suppress inflammation [182,183], while exosome also can act as transport vehicles to deliver cargo to the inflammation site for better anti-inflammatory effects [184,185]. Bruno et al. found that EVs derived from human liver stem cells can reduce pro-inflammatory genes such as TNF, IL-1β, and IFN-γ in the mouse model of non-alcoholic steatohepatitis (NASH), thereby alleviating the inflammatory response of NASH [186]. Other research has shown that EVs released by inflammasome activated macrophages contain interferon beta and induce interferon signatures to inhibit NLRP3 inflammasome activation to prevent systemic inflammatory responses [187].
Furthermore, the unique properties of exosomes are involved in immune regulation of inflammatory or autoimmune diseases and are considered as potential alternatives to cell therapy [188]. For example, EVs derived from human umbilical cord blood cells can suppress T cell proliferation by reducing the production of IL-2 and express MMP-9 and HSP-72 to achieve significant T cell immune suppression, improving experimental autoimmune encephalomyelitis (EAE) [189]. EVs derived from oligodendrocytes protect EAE mice from demyelination and axonal damage in a myelin self-antigen dependent manner, and reduce infiltration of CD45 and CD4 in the central nervous system [190].
The therapeutic cargo within exosomes, such as microRNAs, anti-inflammatory proteins, and growth factors, can modulate immune cell activity, reduce pro-inflammatory cytokines, and promote tissue repair and regeneration [191]. These exosomes can also influence the communication between immune cells and damaged tissues, helping to restore tissue homeostasis and alleviate chronic inflammation.
Taken together, the use of extracellular vesicles as a delivery tool and their direct action on cancer, ischemic-related diseases, and chronic inflammatory-related diseases, etc., make them a promising therapeutic approach for treating a range of conditions. To fully understand the potential of these tiny vesicles, and early results are highly encouraging. While the field of exosome therapy is still in its early stages, these findings highlight the potential of utilizing exosomes as a therapeutic approach. Further research and clinical studies are necessary to optimize exosome isolation, characterization, and delivery methods to harness their full therapeutic potential in treating diseases.
Future work and conclusions
Exosome therapy for acute skin injury is an area of active research, and there are several promising directions that this research is taking. Here are some future directions of exosome therapy for acute skin injury.
Potential for clinical translation of exosome therapy
Exosomes have emerged as promising candidates for clinical translation due to their unique properties and potential therapeutic applications. The potential for clinical translation of exosomes is listed below.
(1). Natural intercellular communication:
Playing a critical role in natural intercellular communication, exosomes act as shuttles to transfer bioactive molecules from donor cells to recipient cells. This unique feature allows exosomes to influence cellular behavior, promote tissue repair, and regulate immune responses. Harnessing this natural communication system offers great potential for targeted therapies.
(2). Minimal immunogenicity:
Exosomes have been shown to have minimal immunogenicity, making them attractive for therapeutic use. They have a reduced likelihood of eliciting immune responses when administered to patients, even across allogeneic or xenogeneic systems. This characteristic increases their compatibility for clinical translation and reduces the risk of adverse reactions.
(3). Specific targeting and payload delivery:
Exosomes can be engineered to carry specific cargo, such as therapeutic proteins, small RNAs, or drugs. By modifying the parent cells, researchers can load exosomes with the desired cargo and direct them to specific target tissues or cells. This targeted delivery system minimizes off-target effects and enhances the therapeutic potential of various treatments.
(4). Combined with biomaterials:
Combined exosomes with hydrogels or 3D scaffolds, exosomes can achieve the effect of continuous and even distribution on the wound, or exosomes can be used as drug carriers to deliver drugs or genes to the wound site, regulate the activity of related growth factors and the formation of blood vessels, thus shortening the time of wound healing and improving wound healing.
(5). Potential for regenerative medicine:
Exosomes derived from stem cells, mesenchymal stromal cells, and other cell types have demonstrated promising regenerative properties. They can promote tissue repair, reduce inflammation, and stimulate cell proliferation and differentiation. These characteristics make exosomes potential candidates for treating various degenerative diseases, such as cardiovascular diseases, neurodegenerative disorders, and tissue injuries.
(6). Drug delivery applications:
Exosomes’ ability to cross biological barriers, such as the blood–brain barrier, and their favorable biodistribution properties make them excellent candidates for drug delivery applications. They can protect their cargo from degradation, extend circulation time, and enhance drug bioavailability, leading to improved therapeutic outcomes.
(7). Non-invasive biomarkers:
Exosomes carry unique biomolecules that reflect physiological status of their parent cells. As such, exosomes provide non-invasive and readily accessible source of biomarkers for diagnosing diseases and monitoring treatment response. This potential for liquid biopsy and disease monitoring holds great promise for personalized medicine.
(8). Clinical trials:
As the safety and efficacy of exosome therapy for acute skin injury is further established, more clinical trials will be conducted to determine the optimal dosage, frequency, and delivery methods for exosome therapy.
While the potential for clinical translation of exosomes is significant, several challenges must be addressed. These include standardizing isolation and purification methods, optimizing loading and targeting strategies, ensuring scalability and reproducibility, and understanding potential long-term effects.
In summary, exosomes represent a promising avenue for clinical translation in several therapeutic areas, including regenerative medicine, targeted drug delivery, and disease diagnostics. Their ability to mediate intercellular communication, minimal immunogenicity, and potential for specific targeting make them attractive candidates for future therapeutic interventions. However, further research, preclinical studies, and clinical trials are needed to fully understand their safety, efficacy, and long-term implications before they can be widely adopted in the clinical setting. With continuing advancements and innovations in the field, exosomes hold great potential to revolutionize modern medicine and improve patient outcomes.
Challenges and opportunities
Although a large number of techniques have been applied to the separation and purification of exosomes, different techniques have some shortcomings, and some of them are not fully mature. Therefore, how to obtain a large number of exosomes with high purity, specificity, and low cost is the current difficulty. At present, with the continuous update of exosome separation technology, great progress has been made in the separation and purification of exosomes, especially researchers are focusing on how to obtain high quality and low cost exosomes through simple procedures. Secondly, exosomes were obtained by combining different separation methods to meet the accuracy of downstream experimental research results. Finally, we need to evaluate the composition of exosomes, although Dennis et al. have identified that annex A1 as a specific marker for microvesicles shed directly from the plasma membrane and found that small extracellular vesicles are not vectors for active DNA release. Therefore, future studies are needed to evaluate exosome composition and better understand exosome heterogeneity [192]. In addition, the activity of exosomes decreases over time. Therefore, it is necessary to develop methods to maintain exosome activity in the future to increase the therapeutic effect of exosomes. We expect that the separation and purification techniques of exosomes will be developed more rapidly and achieve great success in the future, so as to solve the key problems that have long hindered the study of exosomes.
In addition, increasing studies have shown that exosomes can promote skin wound regeneration. But in animal studies, exosomes show fatal shortcomings such as low bioavailability, poor targeting, and short half-life in vivo. Therefore, how to optimize exosomes is a hot and difficult topic in current research, and more studies are needed to confirm the therapeutic effect of different methods to optimize exosomes on skin wounds. What’s more, the effects of exosome administration modes, dose, and time on different types of skin wounds also need to be further studied.
The process of exosome delivery is a complex and dynamic process, involving multiple steps and mechanisms. The specific factors that govern exosome biogenesis, release, and targeting still need research, and a better understanding of these processes could have important implications for the development of new therapies for wound healing. Therefore, optimization of exosomes in isolation and treatment is crucial, and more studies are needed in the future to confirm the therapeutic effect of exosomes in skin wound healing.
The delayed or non-healed skin wound is a common clinical complication. Therefore, effective and rapid promotion of skin wound healing is an urgent clinical problem. Basic medical research provides a powerful theoretical basis for clinical treatment, and clinical research provides human tissue samples and data sources for basic research. Only by combining clinical and basic practice can it be better applied in clinical practice. For example, storing the samples obtained in clinical practice into the biological sample database for basic research; and researchers work with clinicians to better integrate the clinical and basic.
Conclusions
Exosomes are considered as a promising regenerative therapy for skin tissue repair and beyond as they carry a variety of bioactive substances from the parental cell, which can mediate intercellular communication. Exosomes accelerate skin wound healing by promoting vascular regeneration, collagen synthesis, cell proliferation and migration. In this review, we mainly describe how to optimize the regeneration potential of exosomes in skin wounds. Continued research in this area is expected to lead to new breakthroughs in the treatment of acute skin injury and prevent complications.
Acknowledgements
This work was supported by the National Natural Science Foundation of China (Nos. 82060245 and 82260254); Federal funds from the National Institute of Allergy and Infectious Diseases, National Institutes of Health, under Contract Nos. R21 AI152832 and R03 AI165170 (PI: Yuanyuan Zhang).
Footnotes
Conflicts of interest Tianjing Sun, Mo Li, Qi Liu, Anyong Yu, Kun Chen, Jianxing Ma, Sean Murphy, Patrick Michael McNutt, and Yuanyuan Zhang declare that they have no conflict of interest.
This manuscript is a review article and does not involve a research protocol requiring approval by the relevant institutional review board or ethics committee.
References
- 1.Sen CK. Human wounds and its burden: an updated compendium of estimates. Adv Wound Care (New Rochelle) 2019; 8(2): 39–48 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Sen CK. Human wound and its burden: updated 2020 compendium of estimates. Adv Wound Care (New Rochelle) 2021; 10(5): 281–292 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Bacakova L, Zarubova J, Travnickova M, Musilkova J, Pajorova J, Slepicka P, Kasalkova NS, Svorcik V, Kolska Z, Motarjemi H, Molitor M. Stem cells: their source, potency and use in regenerative therapies with focus on adipose-derived stem cells — a review. Biotechnol Adv 2018; 36(4): 1111–1126 [DOI] [PubMed] [Google Scholar]
- 4.Xue M, Dervish S, Jackson CJ. Isolation of human skin epidermal stem cells based on the expression of endothelial protein C receptor. In: Turksen K. Skin Stem Cells. Methods in Molecular Biology, vol 1879. New York: Humana Press, 2018: 165–174 [DOI] [PubMed] [Google Scholar]
- 5.Mokry J, Pisal R. Development and maintenance of epidermal stem cells in skin adnexa. Int J Mol Sci 2020; 21(24): 9736. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Brockmann I, Ehrenpfordt J, Sturmheit T, Brandenburger M, Kruse C, Zille M, Rose D, Boltze J. Skin-derived stem cells for wound treatment using cultured epidermal autografts: clinical applications and challenges. Stem Cells Int 2018; 2018: 4623615. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Veith AP, Henderson K, Spencer A, Sligar AD, Baker AB. Therapeutic strategies for enhancing angiogenesis in wound healing. Adv Drug Deliv Rev 2019; 146: 97–125 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Xiaojie W, Banda J, Qi H, Chang AK, Bwalya C, Chao L, Li X. Scarless wound healing: current insights from the perspectives of TGF-β, KGF-1, and KGF-2. Cytokine Growth Factor Rev 2022; 66: 26–37 [DOI] [PubMed] [Google Scholar]
- 9.Liu J, Qiu X, Lv Y, Zheng C, Dong Y, Dou G, Zhu B, Liu A, Wang W, Zhou J, Liu S, Liu S, Gao B, Jin Y. Apoptotic bodies derived from mesenchymal stem cells promote cutaneous wound healing via regulating the functions of macrophages. Stem Cell Res Ther 2020; 11(1): 507. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Zhou X, Ning K, Ling B, Chen X, Cheng H, Lu B, Gao Z, Xu J. Multiple injections of autologous adipose-derived stem cells accelerate the burn wound healing process and promote blood vessel regeneration in a rat model. Stem Cells Dev 2019; 28(21): 1463–1472 [DOI] [PubMed] [Google Scholar]
- 11.Li M, Qiu L, Hu W, Deng X, Xu H, Cao Y, Xiao Z, Peng L, Johnson S, Alexey L, Kingston PA, Li Q, Zhang Y. Genetically-modified bone mesenchymal stem cells with TGF-β(3) improve wound healing and reduce scar tissue formation in a rabbit model. Exp Cell Res 2018; 367(1): 24–29 [DOI] [PubMed] [Google Scholar]
- 12.Li Q, Qi G, Lutter D, Beard W, Souza CRS, Highland MA, Wu W, Li P, Zhang Y, Atala A, Sun X. Injectable peptide hydrogel encapsulation of mesenchymal stem cells improved viability, stemness, anti-inflammatory effects, and early stage wound healing. Biomolecules 2022; 12(9): 1317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Yin X, Li Q, McNutt PM, Zhang Y. Urine-derived stem cells for epithelial tissues reconstruction and wound healing. Pharmaceutics 2022; 14(8): 1669. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Jeong JO, Han JW, Kim JM, Cho HJ, Park C, Lee N, Kim DW, Yoon YS. Malignant tumor formation after transplantation of short-term cultured bone marrow mesenchymal stem cells in experimental myocardial infarction and diabetic neuropathy. Circ Res 2011; 108(11): 1340–1347 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Trounson A, McDonald C. Stem cell therapies in clinical trials: progress and challenges. Cell Stem Cell 2015; 17(1): 11–22 [DOI] [PubMed] [Google Scholar]
- 16.Lee AS, Tang C, Rao MS, Weissman IL, Wu JC. Tumorigenicity as a clinical hurdle for pluripotent stem cell therapies. Nat Med 2013; 19(8): 998–1004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Ben-David U, Benvenisty N. The tumorigenicity of human embryonic and induced pluripotent stem cells. Nat Rev Cancer 2011; 11(4): 268–277 [DOI] [PubMed] [Google Scholar]
- 18.Min J, Zhang C, Bliton RJ, Caldwell B, Caplan L, Presentation KS, Park DJ, Kong SH, Lee HS, Washington MK, Kim WH, Lau KS, Magness ST, Lee HJ, Yang HK, Goldenring JR, Choi E. Dysplastic stem cell plasticity functions as a driving force for neoplastic transformation of precancerous gastric mucosa. Gastroenterology 2022; 163(4): 875–890 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Deuse T, Hu X, Gravina A, Wang D, Tediashvili G, De C, Thayer WO, Wahl A, Garcia JV, Reichenspurner H, Davis MM, Lanier LL, Schrepfer S. Hypoimmunogenic derivatives of induced pluripotent stem cells evade immune rejection in fully immunocompetent allogeneic recipients. Nat Biotechnol 2019; 37(3): 252–258 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Pearl JI, Lee AS, Leveson-Gower DB, Sun N, Ghosh Z, Lan F, Ransohoff J, Negrin RS, Davis MM, Wu JC. Short-term immunosuppression promotes engraftment of embryonic and induced pluripotent stem cells. Cell Stem Cell 2011; 8(3): 309–317 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Zhao T, Zhang ZN, Rong Z, Xu Y. Immunogenicity of induced pluripotent stem cells. Nature 2011; 474(7350): 212–215 [DOI] [PubMed] [Google Scholar]
- 22.Gopalarethinam J, Nair AP, Iyer M, Vellingiri B, Subramaniam MD. Advantages of mesenchymal stem cell over the other stem cells. Acta Histochem 2023; 125(4): 152041. [DOI] [PubMed] [Google Scholar]
- 23.Ford E, Pearlman J, Ruan T, Manion J, Waller M, Neely GG, Caron L. Human pluripotent stem cells-based therapies for neurodegenerative diseases: current status and challenges. Cells 2020; 9(11): 2517. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Shpichka A, Butnaru D, Bezrukov EA, Sukhanov RB, Atala A, Burdukovskii V, Zhang Y, Timashev P. Skin tissue regeneration for burn injury. Stem Cell Res Ther 2019; 10(1): 94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Zhang Y, He Y, Bharadwaj S, Hammam N, Carnagey K, Myers R, Atala A, Van Dyke M. Tissue-specific extracellular matrix coatings for the promotion of cell proliferation and maintenance of cell phenotype. Biomaterials 2009; 30(23-24): 4021–4028 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Orlando G, Wood KJ, De Coppi P, Baptista PM, Binder KW, Bitar KN, Breuer C, Burnett L, Christ G, Farney A, Figliuzzi M, Holmes JH, Koch K, Macchiarini P, Mirmalek Sani SH, Opara E, Remuzzi A, Rogers J, Saul JM, Seliktar D, Shapira-Schweitzer K, Smith T, Solomon D, Van Dyke M, Yoo JJ, Zhang Y, Atala A, Stratta RJ, Soker S. Regenerative medicine as applied to general surgery. Ann Surg 2012; 255(5): 867–880 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Prasai A, Jay JW, Jupiter D, Wolf SE, El Ayadi A. Role of exosomes in dermal wound healing: a systematic review. J Invest Dermatol 2022; 142(3): 662–678.e8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Kourembanas S. Exosomes: vehicles of intercellular signaling, biomarkers, and vectors of cell therapy. Annu Rev Physiol 2015; 77(1): 13–27 [DOI] [PubMed] [Google Scholar]
- 29.Kalluri R, LeBleu VS. The biology, function, and biomedical applications of exosomes. Science 2020; 367(6478): eaau6977. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Yu Y, Chen M, Guo Q, Shen L, Liu X, Pan J, Zhang Y, Xu T, Zhang D, Wei G. Human umbilical cord mesenchymal stem cell exosome-derived miR-874-3p targeting RIPK1/PGAM5 attenuates kidney tubular epithelial cell damage. Cell Mol Biol Lett 2023; 28(1): 12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Canning P, Alwan A, Khalil F, Zhang Y, Opara EC. Perspectives and challenges on the potential use of exosomes in bioartificial pancreas engineering. Ann Biomed Eng 2022; 50(10): 1177–1186 [DOI] [PubMed] [Google Scholar]
- 32.Deng C, Xie Y, Zhang C, Ouyang B, Chen H, Lv L, Yao J, Liang X, Zhang Y, Sun X, Deng C, Liu G. Urine-derived stem cells facilitate endogenous spermatogenesis restoration of busulfan-induced nonobstructive azoospermic mice by paracrine exosomes. Stem Cells Dev 2019; 28(19): 1322–1333 [DOI] [PubMed] [Google Scholar]
- 33.Hu P, Yang Q, Wang Q, Shi C, Wang D, Armato U, Prà ID, Chiarini A. Mesenchymal stromal cells-exosomes: a promising cell-free therapeutic tool for wound healing and cutaneous regeneration. Burns Trauma 2019; 7: 38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Abdelsaid K, Sudhahar V, Harris RA, Das A, Youn SW, Liu Y, McMenamin M, Hou Y, Fulton D, Hamrick MW, Tang Y, Fukai T, Ushio-Fukai M. Exercise improves angiogenic function of circulating exosomes in type 2 diabetes: role of exosomal SOD3. FASEB J 2022; 36(3): e22177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Zhao W, Zhang R, Zang C, Zhang L, Zhao R, Li Q, Yang Z, Feng Z, Zhang W, Cui R. Exosome derived from mesenchymal stem cells alleviates pathological scars by inhibiting the proliferation, migration and protein expression of fibroblasts via delivering miR-138-5p to target SIRT1. Int J Nanomedicine 2022; 17: 4023–4038 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Park KY, Han HS, Park JW, Kwon HH, Park GH, Seo SJ. Exosomes derived from human adipose tissue-derived mesenchymal stem cells for the treatment of dupilumab-related facial redness in patients with atopic dermatitis: a report of two cases. J Cosmet Dermatol 2022; 21(2): 844–849 [DOI] [PubMed] [Google Scholar]
- 37.Yang GH, Lee YB, Kang D, Choi E, Nam Y, Lee KH, You HJ, Kang HJ, An SH, Jeon H. Overcome the barriers of the skin: exosome therapy. Biomater Res 2021; 25(1): 22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Ha DH, Kim HK, Lee J, Kwon HH, Park GH, Yang SH, Jung JY, Choi H, Lee JH, Sung S, Yi YW, Cho BS. Mesenchymal stem/stromal cell-derived exosomes for immunomodulatory therapeutics and skin regeneration. Cells 2020; 9(5): 1157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Théry C, Zitvogel L, Amigorena S. Exosomes: composition, biogenesis and function. Nat Rev Immunol 2002; 2(8): 569–579 [DOI] [PubMed] [Google Scholar]
- 40.EL Andaloussi S, Mäger I, Breakefield XO, Wood MJ. Extracellular vesicles: biology and emerging therapeutic opportunities. Nat Rev Drug Discov 2013; 12(5): 347–357 [DOI] [PubMed] [Google Scholar]
- 41.Karnati HK, Garcia JH, Tweedie D, Becker RE, Kapogiannis D, Greig NH. Neuronal enriched extracellular vesicle proteins as biomarkers for traumatic brain injury. J Neurotrauma 2019; 36(7): 975–987 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Hade MD, Suire CN, Mossell J, Suo Z. Extracellular vesicles: emerging frontiers in wound healing. Med Res Rev 2022; 42(6): 2102–2125 [DOI] [PubMed] [Google Scholar]
- 43.Han QF, Li WJ, Hu KS, Gao J, Zhai WL, Yang JH, Zhang SJ. Exosome biogenesis: machinery, regulation, and therapeutic implications in cancer. Mol Cancer 2022; 21(1): 207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Farooqi AA, Desai NN, Qureshi MZ, Librelotto DRN, Gasparri ML, Bishayee A, Nabavi SM, Curti V, Daglia M. Exosome biogenesis, bioactivities and functions as new delivery systems of natural compounds. Biotechnol Adv 2018; 36(1): 328–334 [DOI] [PubMed] [Google Scholar]
- 45.Pan BT, Teng K, Wu C, Adam M, Johnstone RM. Electron microscopic evidence for externalization of the transferrin receptor in vesicular form in sheep reticulocytes. J Cell Biol 1985; 101(3): 942–948 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Pan BT, Johnstone RM. Fate of the transferrin receptor during maturation of sheep reticulocytes in vitro: selective externalization of the receptor. Cell 1983; 33(3): 967–978 [DOI] [PubMed] [Google Scholar]
- 47.Yan C, Chen J, Wang C, Yuan M, Kang Y, Wu Z, Li W, Zhang G, Machens HG, Rinkevich Y, Chen Z, Yang X, Xu X. Milk exosomes-mediated miR-31-5p delivery accelerates diabetic wound healing through promoting angiogenesis. Drug Deliv 2022; 29(1): 214–228 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Gurunathan S, Kang MH, Kim JH. A comprehensive review on factors influences biogenesis, functions, therapeutic and clinical implications of exosomes. Int J Nanomedicine 2021; 16: 1281–1312 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Choi EW, Seo MK, Woo EY, Kim SH, Park EJ, Kim S. Exosomes from human adipose-derived stem cells promote proliferation and migration of skin fibroblasts. Exp Dermatol 2018; 27(10): 1170–1172 [DOI] [PubMed] [Google Scholar]
- 50.Narauskaitė D, Vydmantaitė G, Rusteikaitė J, Sampath R, Rudaitytė A, Stašytė G, Aparicio Calvente MI, Jekabsone A. Extracellular vesicles in skin wound healing. Pharmaceuticals (Basel) 2021; 14(8): 811. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Rodrigues M, Kosaric N, Bonham CA, Gurtner GC. Wound healing: a cellular perspective. Physiol Rev 2019; 99(1): 665–706 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Čoma M, Manning JC, Kaltner H, Gál P. The sweet side of wound healing: galectins as promising therapeutic targets in hemostasis, inflammation, proliferation, and maturation/remodeling. Expert Opin Ther Targets 2023; 27(1): 41–53 [DOI] [PubMed] [Google Scholar]
- 53.Hassanshahi A, Moradzad M, Ghalamkari S, Fadaei M, Cowin AJ, Hassanshahi M. Macrophage-mediated inflammation in skin wound healing. Cells 2022; 11(19): 2953. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Reinke JM, Sorg H. Wound repair and regeneration. Eur Surg Res 2012; 49(1): 35–43 [DOI] [PubMed] [Google Scholar]
- 55.Moretti L, Stalfort J, Barker TH, Abebayehu D. The interplay of fibroblasts, the extracellular matrix, and inflammation in scar formation. J Biol Chem 2022; 298(2): 101530. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Talbott HE, Mascharak S, Griffin M, Wan DC, Longaker MT. Wound healing, fibroblast heterogeneity, and fibrosis. Cell Stem Cell 2022; 29(8): 1161–1180 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Johnson J, Wu YW, Blyth C, Lichtfuss G, Goubran H, Burnouf T. Prospective therapeutic applications of platelet extracellular vesicles. Trends Biotechnol 2021; 39(6): 598–612 [DOI] [PubMed] [Google Scholar]
- 58.Sinauridze EI, Kireev DA, Popenko NY, Pichugin AV, Panteleev MA, Krymskaya OV, Ataullakhanov FI. Platelet microparticle membranes have 50- to 100-fold higher specific procoagulant activity than activated platelets. Thromb Haemost 2007; 97(3): 425–434 [PubMed] [Google Scholar]
- 59.Antich-Rosselló M, Forteza-Genestra MA, Monjo M, Ramis JM. Platelet-derived extracellular vesicles for regenerative medicine. Int J Mol Sci 2021; 22(16): 8580. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Graça AL, Domingues RMA, Gomez-Florit M, Gomes ME. Platelet-derived extracellular vesicles promote tenogenic differentiation of stem cells on bioengineered living fibers. Int J Mol Sci 2023; 24(4): 3516. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.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 [DOI] [PubMed] [Google Scholar]
- 62.Guo SC, Tao SC, Yin WJ, Qi X, Yuan T, Zhang CQ. Exosomes derived from platelet-rich plasma promote the re-epithelization of chronic cutaneous wounds via activation of YAP in a diabetic rat model. Theranostics 2017; 7(1): 81–96 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Kim H, Wang SY, Kwak G, Yang Y, Kwon IC, Kim SH. Exosome-guided phenotypic switch of M1 to M2 macrophages for cutaneous wound healing. Adv Sci (Weinh) 2019; 6(20): 1900513. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Hu JC, Zheng CX, Sui BD, Liu WJ, Jin Y. Mesenchymal stem cell-derived exosomes: a novel and potential remedy for cutaneous wound healing and regeneration. World J Stem Cells 2022; 14(5): 318–329 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Su D, Tsai HI, Xu Z, Yan F, Wu Y, Xiao Y, Liu X, Wu Y, Parvanian S, Zhu W, Eriksson JE, Wang D, Zhu H, Chen H, Cheng F. Exosomal PD-L1 functions as an immunosuppressant to promote wound healing. J Extracell Vesicles 2020; 9(1): 1709262. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Kwak G, Cheng J, Kim H, Song S, Lee SJ, Yang Y, Jeong JH, Lee JE, Messersmith PB, Kim SH. 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(15): 2200060. [DOI] [PubMed] [Google Scholar]
- 68.Qiu X, Liu J, Zheng C, Su Y, Bao L, Zhu B, Liu S, Wang L, Wang X, Wang Y, Zhao W, Zhou J, Deng Z, Liu S, Jin Y. Exosomes released from educated mesenchymal stem cells accelerate cutaneous wound healing via promoting angiogenesis. Cell Prolif 2020; 53(8): e12830. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Tutuianu R, Rosca AM, Iacomi DM, Simionescu M, Titorencu I. Human mesenchymal stromal cell-derived exosomes promote in vitro wound healing by modulating the biological properties of skin keratinocytes and fibroblasts and stimulating angiogenesis. Int J Mol Sci 2021; 22(12): 6239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Marofi F, Alexandrovna KI, Margiana R, Bahramali M, Suksatan W, Abdelbasset WK, Chupradit S, Nasimi M, Maashi MS. MSCs and their exosomes: a rapidly evolving approach in the context of cutaneous wounds therapy. Stem Cell Res Ther 2021; 12(1): 597. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Than UTT, Guanzon D, Leavesley D, Parker T. Association of extracellular membrane vesicles with cutaneous wound healing. Int J Mol Sci 2017; 18(5): 956. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Shabbir A, Cox A, Rodriguez-Menocal L, Salgado M, Van Badiavas E. Mesenchymal stem cell exosomes induce proliferation and migration of normal and chronic wound fibroblasts, and enhance angiogenesis in vitro. Stem Cells Dev 2015; 24(14): 1635–1647 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Zhao B, Zhang X, Zhang Y, Lu Y, Zhang W, Lu S, Fu Y, Zhou Y, Zhang J, Zhang J. Human exosomes accelerate cutaneous wound healing by promoting collagen synthesis in a diabetic mouse model. Stem Cells Dev 2021; 30(18): 922–933 [DOI] [PubMed] [Google Scholar]
- 74.Zhang J, Guan J, Niu X, Hu G, Guo S, Li Q, Xie Z, Zhang C, Wang Y. Exosomes released from human induced pluripotent stem cells-derived MSCs facilitate cutaneous wound healing by promoting collagen synthesis and angiogenesis. J Transl Med 2015; 13(1): 49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Oh EJ, Gangadaran P, Rajendran RL, Kim HM, Oh JM, Choi KY, Chung HY, Ahn BC. Extracellular vesicles derived from fibroblasts promote wound healing by optimizing fibroblast and endothelial cellular functions. Stem Cells 2021; 39(3): 266–279 [DOI] [PubMed] [Google Scholar]
- 76.Bian D, Wu Y, Song G, Azizi R, Zamani A. The application of mesenchymal stromal cells (MSCs) and their derivative exosome in skin wound healing: a comprehensive review. Stem Cell Res Ther 2022; 13(1): 24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Huang J, Xiong J, Yang L, Zhang J, Sun S, Liang Y. Cell-free exosome-laden scaffolds for tissue repair. Nanoscale 2021; 13(19): 8740–8750 [DOI] [PubMed] [Google Scholar]
- 78.Tenchov R, Sasso JM, Wang X, Liaw WS, Chen CA, Zhou QA. Exosomes—nature’s lipid nanoparticles, a rising star in drug delivery and diagnostics. ACS Nano 2022; 16(11): 17802–17846 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca JD, Moorman MA, Simonetti DW, Craig S, Marshak DR. Multilineage potential of adult human mesenchymal stem cells. Science 1999; 284(5411): 143–147 [DOI] [PubMed] [Google Scholar]
- 80.Liu S, Zhou J, Zhang X, Liu Y, Chen J, Hu B, Song J, Zhang Y. Strategies to optimize adult stem cell therapy for tissue regeneration. Int J Mol Sci 2016; 17(6): 982. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Mousaei Ghasroldasht M, Seok J, Park HS, Liakath Ali FB, Al-Hendy A. Stem cell therapy: from idea to clinical practice. Int J Mol Sci 2022; 23(5): 2850. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Nourian Dehkordi A, Mirahmadi Babaheydari F, Chehelgerdi M, Raeisi Dehkordi S. Skin tissue engineering: wound healing based on stem-cell-based therapeutic strategies. Stem Cell Res Ther 2019; 10(1): 111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Yousefi Dehbidi M, Goodarzi N, Azhdari MH, Doroudian M. Mesenchymal stem cells and their derived exosomes to combat Covid-19. Rev Med Virol 2022; 32(2): e2281. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Ren XS, Tong Y, Qiu Y, Ye C, Wu N, Xiong XQ, Wang JJ, Han Y, Zhou YB, Zhang F, Sun HJ, Gao XY, Chen Q, Li YH, Kang YM, Zhu GQ. miR155-5p in adventitial fibroblasts-derived extracellular vesicles inhibits vascular smooth muscle cell proliferation via suppressing angiotensin-converting enzyme expression. J Extracell Vesicles 2020; 9(1): 1698795. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Mahmoudi F, Hanachi P, Montaseri A. Extracellular vesicles of immune cells; immunomodulatory impacts and therapeutic potentials. Clin Immunol 2023; 248: 109237. [DOI] [PubMed] [Google Scholar]
- 86.Shen K, Wang XJ, Liu KT, Li SH, Li J, Zhang JX, Wang HT, Hu DH. Effects of exosomes from human adipose-derived mesenchymal stem cells on inflammatory response of mouse RAW264.7 cells and wound healing of full-thickness skin defects in mice Chin J Burns (Zhonghua Shao Shang Za Zhi) 2022; 38(3): 215–226 (in Chinese) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Zhou Y, Zhao B, Zhang XL, Lu YJ, Lu ST, Cheng J, Fu Y, Lin L, Zhang NY, Li PX, Zhang J, Zhang J. 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(1): 257. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.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(1): 452. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Lv H, Liu H, Sun T, Wang H, Zhang X, Xu W. Exosome derived from stem cell: a promising therapeutics for wound healing. Front Pharmacol 2022; 13: 957771. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Wang M, Wu P, Huang J, Liu W, Qian H, Sun Y, Shi H. Skin cell-derived extracellular vesicles: a promising therapeutic strategy for cutaneous injury. Burns Trauma 2022; 10: tkac037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Haraszti RA, Miller R, Dubuke ML, Rockwell HE, Coles AH, Sapp E, Didiot MC, Echeverria D, Stoppato M, Sere YY, Leszyk J, Alterman JF, Godinho B, Hassler MR, McDaniel J, Narain NR, Wollacott R, Wang Y, Shaffer SA, Kiebish MA, DiFiglia M, Aronin N, Khvorova A. Serum deprivation of mesenchymal stem cells improves exosome activity and alters lipid and protein composition. iScience 2019; 16: 230–241 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Bost JP, Saher O, Hagey D, Mamand DR, Liang X, Zheng W, Corso G, Gustafsson O, Görgens A, Smith CE, Zain R, El Andaloussi S, Gupta D. Growth media conditions influence the secretion route and release levels of engineered extracellular vesicles. Adv Healthc Mater 2022; 11(5): 2101658. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Gonzalez-King H, García NA, Ontoria-Oviedo I, Ciria M, Montero JA, Sepúlveda P. Hypoxia Inducible factor-1α potentiates jagged 1-mediated angiogenesis by mesenchymal stem cell-derived exosomes. Stem Cells 2017; 35(7): 1747–1759 [DOI] [PubMed] [Google Scholar]
- 96.Ng CY, Kee LT, Al-Masawa ME, Lee QH, Subramaniam T, Kok D, Ng MH, Law JX. Scalable production of extracellular vesicles and its therapeutic values: a review. Int J Mol Sci 2022; 23(14): 7986. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Cheng Y, Zeng Q, Han Q, Xia W. Effect of pH, temperature and freezing-thawing on quantity changes and cellular uptake of exosomes. Protein Cell 2019; 10(4): 295–299 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Patel DB, Gray KM, Santharam Y, Lamichhane TN, Stroka KM, Jay SM. Impact of cell culture parameters on production and vascularization bioactivity of mesenchymal stem cell-derived extracellular vesicles. Bioeng Transl Med 2017; 2(2): 170–179 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Linares R, Tan S, Gounou C, Arraud N, Brisson AR. High-speed centrifugation induces aggregation of extracellular vesicles. J Extracell Vesicles 2015; 4(1): 29509. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Ludwig AK, De Miroschedji K, Doeppner TR, Börger V, Ruesing J, Rebmann V, Durst S, Jansen S, Bremer M, Behrmann E, Singer BB, Jastrow H, Kuhlmann JD, El Magraoui F, Meyer HE, Hermann DM, Opalka B, Raunser S, Epple M, Horn PA, Giebel B. Precipitation with polyethylene glycol followed by washing and pelleting by ultracentrifugation enriches extracellular vesicles from tissue culture supernatants in small and large scales. J Extracell Vesicles 2018; 7(1): 1528109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Arntz OJ, Pieters BCH, van Lent P, Koenders MI, van der Kraan PM, van de Loo FAJ. An optimized method for plasma extracellular vesicles isolation to exclude the copresence of biological drugs and plasma proteins which impairs their biological characterization. PLoS One 2020; 15(7): e0236508. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Zheng H, Guan S, Wang X, Zhao J, Gao M, Zhang X. Deconstruction of heterogeneity of size-dependent exosome subpopulations from human urine by profiling N-glycoproteomics and phosphoproteomics simultaneously. Anal Chem 2020; 92(13): 9239–9246 [DOI] [PubMed] [Google Scholar]
- 103.Liangsupree T, Multia E, Riekkola ML. Modern isolation and separation techniques for extracellular vesicles. J Chromatogr A 2021; 1636: 461773. [DOI] [PubMed] [Google Scholar]
- 104.Lin S, Yu Z, Chen D, Wang Z, Miao J, Li Q, Zhang D, Song J, Cui D. Progress in microfluidics-based exosome separation and detection technologies for diagnostic applications. Small 2020; 16(9): 1903916. [DOI] [PubMed] [Google Scholar]
- 105.Sitar S, Kejžar A, Pahovnik D, Kogej K, Tušek-Žnidarič M, Lenassi M, Žagar E. Size characterization and quantification of exosomes by asymmetrical-flow field-flow fractionation. Anal Chem 2015; 87(18): 9225–9233 [DOI] [PubMed] [Google Scholar]
- 106.Zhang H, Freitas D, Kim HS, Fabijanic K, Li Z, Chen H, Mark MT, Molina H, Martin AB, Bojmar L, Fang J, Rampersaud S, Hoshino A, Matei I, Kenific CM, Nakajima M, Mutvei AP, Sansone P, Buehring W, Wang H, Jimenez JP, Cohen-Gould L, Paknejad N, Brendel M, Manova-Todorova K, Magalhães A, Ferreira JA, Osório H, Silva AM, Massey A, Cubillos-Ruiz JR, Galletti G, Giannakakou P, Cuervo AM, Blenis J, Schwartz R, Brady MS, Peinado H, Bromberg J, Matsui H, Reis CA, Lyden D. Identification of distinct nanoparticles and subsets of extracellular vesicles by asymmetric flow field-flow fractionation. Nat Cell Biol 2018; 20(3): 332–343 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Chen C, Skog J, Hsu CH, Lessard RT, Balaj L, Wurdinger T, Carter BS, Breakefield XO, Toner M, Irimia D. Microfluidic isolation and transcriptome analysis of serum microvesicles. Lab Chip 2010; 10(4): 505–511 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Zhang P, He M, Zeng Y. Ultrasensitive microfluidic analysis of circulating exosomes using a nanostructured graphene oxide/polydopamine coating. Lab Chip 2016; 16(16): 3033–3042 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Bohmer N, Demarmels N, Tsolaki E, Gerken L, Keevend K, Bertazzo S, Lattuada M, Herrmann IK. Removal of cells from body fluids by magnetic separation in batch and continuous mode: influence of bead size, concentration, and contact time. ACS Appl Mater Interfaces 2017; 9(35): 29571–29579 [DOI] [PubMed] [Google Scholar]
- 110.Fang X, Chen C, Liu B, Ma Z, Hu F, Hi H, Gu H, Xu H. A magnetic bead-mediated selective adsorption strategy for extracellular vesicle separation and purification. Acta Biomater 2021; 124: 336–347 [DOI] [PubMed] [Google Scholar]
- 111.Seo N, Nakamura J, Kaneda T, Tateno H, Shimoda A, Ichiki T, Furukawa K, Hirabayashi J, Akiyoshi K, Shiku H. Distinguishing functional exosomes and other extracellular vesicles as a nucleic acid cargo by the anion-exchange method. J Extracell Vesicles 2022; 11(3): e12205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Chattrairat K, Yasui T, Suzuki S, Natsume A, Nagashima K, Iida M, Zhang M, Shimada T, Kato A, Aoki K, Ohka F, Yamazaki S, Yanagida T, Baba Y. All-in-one nanowire assay system for capture and analysis of extracellular vesicles from an ex vivo brain tumor model. ACS Nano 2023; 17(3): 2235–2244 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Yasui T, Paisrisarn P, Yanagida T, Konakade Y, Nakamura Y, Nagashima K, Musa M, Thiodorus IA, Takahashi H, Naganawa T, Shimada T, Kaji N, Ochiya T, Kawai T, Baba Y. Molecular profiling of extracellular vesicles via charge-based capture using oxide nanowire microfluidics. Biosens Bioelectron 2021; 194: 113589. [DOI] [PubMed] [Google Scholar]
- 114.Stam J, Bartel S, Bischoff R, Wolters JC. Isolation of extracellular vesicles with combined enrichment methods. J Chromatogr B Analyt Technol Biomed Life Sci 2021; 1169: 122604. [DOI] [PubMed] [Google Scholar]
- 115.Onódi Z, Pelyhe C, Terézia Nagy C, Brenner GB, Almási F, Kittel Á, Manček-Keber M, Ferdinandy P, Buzás EI, Giricz Z. Isolation of high-purity extracellular vesicles by the combination of iodixanol density gradient ultracentrifugation and bind-elute chromatography from blood plasma. Front Physiol 2018; 9: 1479. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Hettich BF, Ben-Yehuda Greenwald M, Werner S, Leroux JC. Exosomes for wound healing: purification optimization and identification of bioactive components. Adv Sci (Weinh) 2020; 7(23): 2002596. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Jin C, Wu P, Li L, Xu W, Qian H. Exosomes: emerging therapy delivery tools and biomarkers for kidney diseases. Stem Cells Int 2021; 2021: 7844455. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Singh K, Nalabotala R, Koo KM, Bose S, Nayak R, Shiddiky MJA. Separation of distinct exosome subpopulations: isolation and characterization approaches and their associated challenges. Analyst (Lond) 2021; 146(12): 3731–3749 [DOI] [PubMed] [Google Scholar]
- 119.Wei Z, Chen Z, Zhao Y, Fan F, Xiong W, Song S, Yin Y, Hu J, Yang L, Yang F, Xu B, Ge J. Mononuclear phagocyte system blockade using extracellular vesicles modified with CD47 on membrane surface for myocardial infarction reperfusion injury treatment. Biomaterials 2021; 275: 121000. [DOI] [PubMed] [Google Scholar]
- 120.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(5): 1723–1733 [DOI] [PubMed] [Google Scholar]
- 121.Shi A, Li J, Qiu X, Sabbah M, Boroumand S, Huang TC, Zhao C, Terzic A, Behfar A, Moran SL. TGF-β loaded exosome enhances ischemic wound healing in vitro and in vivo. Theranostics 2021; 11(13): 6616–6631 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Togliatto G, Dentelli P, Rosso A, Lombardo G, Gili M, Gallo S, Gai C, Solini A, Camussi G, Brizzi MF. PDGF-BB carried by endothelial cell-derived extracellular vesicles reduces vascular smooth muscle cell apoptosis in diabetes. Diabetes 2018; 67(4): 704–716 [DOI] [PubMed] [Google Scholar]
- 123.Lou R, Chen J, Zhou F, Wang C, Leung CH, Lin F. Exosome-cargoed microRNAs: Potential therapeutic molecules for diabetic wound healing. Drug Discov Today 2022; 27(10): 103323. [DOI] [PubMed] [Google Scholar]
- 124.Shen J, Zhao X, Zhong Y, Yang P, Gao P, Wu X, Wang X, An W. Exosomal ncRNAs: the pivotal players in diabetic wound healing. Front Immunol 2022; 13: 1005307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Xu YX, Pu SD, Li X, Yu ZW, Zhang YT, Tong XW, Shan YY, Gao XY. Exosomal ncRNAs: novel therapeutic target and biomarker for diabetic complications. Pharmacol Res 2022; 178: 106135. [DOI] [PubMed] [Google Scholar]
- 126.Matsuzaka Y, Yashiro R. Advances in purification, modification, and application of extracellular vesicles for novel clinical treatments. Membranes (Basel) 2022; 12(12): 1244. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.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. [DOI] [PubMed] [Google Scholar]
- 128.Li M, Qiu L, Hu W, Deng X, Xu H, Cao Y, Xiao Z, Peng L, Johnson S, Alexey L, Kingston PA, Li Q, Zhang Y. Genetically-modified bone mesenchymal stem cells with TGF-beta3 improve wound healing and reduce scar tissue formation in a rabbit model. Exp Cell Res 2018; 367(1): 24–29 [DOI] [PubMed] [Google Scholar]
- 129.Hade MD, Suire CN, Suo Z. An effective peptide-based platform for efficient exosomal loading and cellular delivery of a microRNA. ACS Appl Mater Interfaces 2023; 15(3): 3851–3866 [DOI] [PubMed] [Google Scholar]
- 130.Zhu J, Fiu 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.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(1): 350. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Wu D, Kang L, Tian J, Wu Y, Liu J, Li Z, Wu X, Huang Y, Gao B, Wang H, Wu Z, Qiu G. 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 Nanomedicine 2020; 15: 7979–7993 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.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 Nanobiotechnology 2021; 19(1): 202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Liu W, Li L, Rong Y, Qian D, Chen J, Zhou Z, Luo Y, Jiang D, Cheng L, Zhao S, Kong F, Wang J, Zhou Z, Xu T, Gong F, Huang Y, Gu C, Zhao X, Bai J, Wang F, Zhao W, Zhang L, Li X, Yin G, Fan J, Cai W. Hypoxic mesenchymal stem cell-derived exosomes promote bone fracture healing by the transfer of miR-126. Acta Biomater 2020; 103: 196–212 [DOI] [PubMed] [Google Scholar]
- 135.Gao W, He R, Ren J, Zhang W, Wang K, Zhu L, Liang T. Exosomal HMGB1 derived from hypoxia-conditioned bone marrow mesenchymal stem cells increases angiogenesis via the JNK/HIF-1α pathway. FEBS Open Bio 2021; 11(5): 1364–1373 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Zhang XF, Wang T, Wang ZX, Huang KP, Zhang YW, Wang GL, Zhang HJ, Chen ZH, Wang CY, Zhang JX, Wang H. 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 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Cao T, Xiao D, Ji P, Zhang Z, Cai WX, Han C, Li W, Tao K. Effects of exosomes from hepatocyte growth factor-modified human adipose mesenchymal stem cells on full-thickness skin defect in diabetic mice. Chin J Burns (Zhonghua Shao Shang Za Zhi) 2022; 38(11): 1004–1013 (in Chinese) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Amengual-Tugores AM, Ráez-Meseguer C, Forteza-Genestra MA, Monjo M, Ramis JM. Extracellular vesicle-based hydrogels for wound healing applications. Int J Mol Sci 2023; 24(4): 4104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Chen C, Bai X, Ding Y, Lee IS. Electrical stimulation as a novel tool for regulating cell behavior in tissue engineering. Biomater Res 2019; 23(1): 25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Hu M, Hong L, Liu C, Hong S, He S, Zhou M, Huang G, Chen Q. Electrical stimulation enhances neuronal cell activity mediated by Schwann cell derived exosomes. Sci Rep 2019; 9(1): 4206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Klein JD, Wang XH. Electrically stimulated acupuncture increases renal blood flow through exosome-carried miR-181. Am J Physiol Renal Physiol 2018; 315(6): F1542–F1549 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Wang J, Pothana K, Chen S, Sawant H, Travers JB, Bihl J, Chen Y. Ultraviolet B irradiation alters the level and miR contents of exosomes released by keratinocytes in diabetic condition. Photochem Photobiol 2022; 98(5): 1122–1130 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Ni N, Ma W, Tao Y, Liu J, Hua H, Cheng J, Wang J, Zhou B, Luo D. Exosomal miR-769-5p exacerbates ultraviolet-induced bystander effect by targeting TGFBR1. Front Physiol 2020; 11: 603081. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Dong J, Wu B, Tian W. Exosomes derived from hypoxia-preconditioned mesenchymal stem cells (hypoMSCs-Exo): advantages in disease treatment. Cell Tissue Res 2023; 392(3): 621–629 [DOI] [PubMed] [Google Scholar]
- 145.Zhang Y, Bi J, Huang J, Tang Y, Du S, Li P. Exosome: a review of its classification, isolation techniques, storage, diagnostic and targeted therapy applications. Int J Nanomedicine 2020; 15: 6917–6934 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Maroto R, Zhao Y, Jamaluddin M, Popov VL, Wang H, Kalubowilage M, Zhang Y, Luisi J, Sun H, Culbertson CT, Bossmann SH, Motamedi M, Brasier AR. Effects of storage temperature on airway exosome integrity for diagnostic and functional analyses. J Extracell Vesicles 2017; 6(1): 1359478. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Bosch S, de Beaurepaire L, Allard M, Mosser M, Heichette C, Chrétien D, Jegou D, Bach JM. Trehalose prevents aggregation of exosomes and cryodamage. Sci Rep 2016; 6(1): 36162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Charoenviriyakul C, Takahashi Y, Nishikawa M, Takakura Y. Preservation of exosomes at room temperature using lyophilization. Int J Pharm 2018; 553(1-2): 1–7 [DOI] [PubMed] [Google Scholar]
- 149.Gupta D, Zickler AM, El Andaloussi S. Dosing extracellular vesicles. Adv Drug Deliv Rev 2021; 178: 113961. [DOI] [PubMed] [Google Scholar]
- 150.Zhou T, He C, Lai P, Yang Z, Liu Y, Xu H, Lin X, Ni B, Ju R, Yi W, Liang L, Pei D, Egwuagu CE, Liu X. miR-204-containing exosomes ameliorate GVHD-associated dry eye disease. Sci Adv 2022; 8(2): eabj9617. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Samaeekia R, Rabiee B, Putra I, Shen X, Park YJ, Hematti P, Eslani M, Djalilian AR. Effect of human corneal mesenchymal stromal cell-derived exosomes on corneal epithelial wound healing. Invest Ophthalmol Vis Sci 2018; 59(12): 5194–5200 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Chen YS, Lin EY, Chiou TW, Harn HJ. Exosomes in clinical trial and their production in compliance with good manufacturing practice. Ci Ji Yi Xue Za Zhi 2020; 32(2): 113–120 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Hu Y, Tao R, Chen L, Xiong Y, Xue H, Hu L, Yan C, Xie X, Lin Z, Panayi AC, Mi B, Liu G. Exosomes derived from pioglitazone-pretreated MSCs accelerate diabetic wound healing through enhancing angiogenesis. J Nanobiotechnology 2021; 19(1): 150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Li Q, Hu W, Huang Q, Yang J, Li B, Ma K, Wei Q, Wang Y, Su J, Sun M, Cui S, Yang R, Li H, Fu X, Zhang C. miR146a-loaded engineered exosomes released from silk fibroin patch promote diabetic wound healing by targeting IRAK1. Signal Transduct Target Ther 2023; 8(1): 62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.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(1): 32993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Dinh PC, Paudel D, Brochu H, Popowski KD, Gracieux MC, Cores J, Huang K, Hensley MT, Harrell E, Vandergriff AC, George AK, Barrio RT, Hu S, Allen TA, Blackburn K, Caranasos TG, Peng X, Schnabel LV, Adler KB, Lobo LJ, Goshe MB, Cheng K. Inhalation of lung spheroid cell secretome and exosomes promotes lung repair in pulmonary fibrosis. Nat Commun 2020; 11(1): 1064. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Liu C, Yan X, Zhang Y, Yang M, Ma Y, Zhang Y, Xu Q, Tu K, Zhang M. Oral administration of turmeric-derived exosome-like nanovesicles with anti-inflammatory and pro-resolving bioactions for murine colitis therapy. J Nanobiotechnology 2022; 20(1): 206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Zhong J, Xia B, Shan S, Zheng A, Zhang S, Chen J, Liang XJ. High-quality milk exosomes as oral drug delivery system. Biomaterials 2021; 277: 121126. [DOI] [PubMed] [Google Scholar]
- 159.Al-Masawa ME, Alshawsh MA, Ng CY, Ng AMH, Foo JB, Vijakumaran U, Subramaniam R, Ghani NAA, Witwer KW, Law JX. 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(15): 6455–6508 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Jia Y, Yu L, Ma T, Xu W, Qian H, Sun Y, Shi H. Small extracellular vesicles isolation and separation: current techniques, pending questions and clinical applications. Theranostics 2022; 12(15): 6548–6575 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Li MY, Liu LZ, Dong M. Progress on pivotal role and application of exosome in lung cancer carcinogenesis, diagnosis, therapy and prognosis. Mol Cancer 2021; 20(1): 22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Wang J, Tang W, Yang M, Yin Y, Li H, Hu F, Tang L, Ma X, Zhang Y, Wang Y. Inflammatory tumor microenvironment responsive neutrophil exosomes-based drug delivery system for targeted glioma therapy. Biomaterials 2021; 273: 120784. [DOI] [PubMed] [Google Scholar]
- 163.Paskeh MDA, Entezari M, Mirzaei S, Zabolian A, Saleki H, Naghdi MJ, Sabet S, Khoshbakht MA, Hashemi M, Hushmandi K, Sethi G, Zarrabi A, Kumar AP, Tan SC, Papadakis M, Alexiou A, Islam MA, Mostafavi E, Ashrafizadeh M. Emerging role of exosomes in cancer progression and tumor microenvironment remodeling. J Hematol Oncol 2022; 15(1): 83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Liang G, Zhu Y, Ali DJ, Tian T, Xu H, Si K, Sun B, Chen B, Xiao Z. Engineered exosomes for targeted co-delivery of miR-21 inhibitor and chemotherapeutics to reverse drug resistance in colon cancer. J Nanobiotechnology 2020; 18(1): 10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Wang X, Zhang H, Bai M, Ning T, Ge S, Deng T, Liu R, Zhang L, Ying G, Ba Y. Exosomes serve as nanoparticles to deliver anti-miR-214 to reverse chemoresistance to cisplatin in gastric cancer. Mol Ther 2018; 26(3): 774–783 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Ashrafizadeh M, Delfi M, Hashemi F, Zabolian A, Saleki H, Bagherian M, Azami N, Farahani MV, Sharifzadeh SO, Hamzehlou S, Hushmandi K, Makvandi P, Zarrabi A, Hamblin MR, Varma RS. Biomedical application of chitosan-based nanoscale delivery systems: potential usefulness in siRNA delivery for cancer therapy. Carbohydr Polym 2021; 260: 117809. [DOI] [PubMed] [Google Scholar]
- 167.Xu Z, Zeng S, Gong Z, Yan Y. Exosome-based immunotherapy: a promising approach for cancer treatment. Mol Cancer 2020; 19(1): 160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Ni C, Fang QQ, Chen WZ, Jiang JX, Jiang Z, Ye J, Zhang T, Yang L, Meng FB, Xia WJ, Zhong M, Huang J. Breast cancer-derived exosomes transmit lncRNA SNHG16 to induce CD73+γδ1 Treg cells. Signal Transduct Target Ther 2020; 5(1): 41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Dou D, Ren X, Han M, Xu X, Ge X, Gu Y, Wang X. Cancer-associated fibroblasts-derived exosomes suppress immune cell function in breast cancer via the miR-92/PD-L1 pathway. Front Immunol 2020; 11: 2026. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 170.Xin W, Qin Y, Lei P, Zhang J, Yang X, Wang Z. From cerebral ischemia towards myocardial, renal, and hepatic ischemia: exosomal miRNAs as a general concept of intercellular communication in ischemia-reperfusion injury. Mol Ther Nucleic Acids 2022; 29: 900–922 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Zou Y, Li L, Li Y, Chen S, Xie X, Jin X, Wang X, Ma C, Fan G, Wang W. Restoring cardiac functions after myocardial infarction-ischemia/reperfusion via an exosome anchoring conductive hydrogel. ACS Appl Mater Interfaces 2021; 13(48): 56892–56908 [DOI] [PubMed] [Google Scholar]
- 172.Chen W, Wang H, Zhu Z, Feng J, Chen L. Exosome-shuttled circSHOC2 from IPASs regulates neuronal autophagy and ameliorates ischemic brain injury via the miR-7670-3p/SIRT1 axis. Mol Ther Nucleic Acids 2020; 22: 657–672 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Yang Z, Gao Z, Yang Z, Zhang Y, Chen H, Yang X, Fang X, Zhu Y, Zhang J, Ouyang F, Li J, Cai G, Li Y, Lin X, Ni R, Xia C, Wang R, Shi X, Chu L. Lactobacillus plantarum-derived extracellular vesicles protect against ischemic brain injury via the microRNA-101a-3p/c-Fos/TGF-β axis. Pharmacol Res 2022; 182: 106332. [DOI] [PubMed] [Google Scholar]
- 174.Kim S, Lee SA, Yoon H, Kim MY, Yoo JK, Ahn SH, Park CH, Park J, Nam BY, Park JT, Han SH, Kang SW, Kim NH, Kim HS, Han D, Yook JI, Choi C, Yoo TH. Exosome-based delivery of super-repressor IκBα ameliorates kidney ischemia-reperfusion injury. Kidney Int 2021; 100(3): 570–584 [DOI] [PubMed] [Google Scholar]
- 175.Wang C, Xu M, Fan Q, Li C, Zhou X. Therapeutic potential of exosome-based personalized delivery platform in chronic inflammatory diseases. Asian J Pharm Sci 2023; 18(1): 100772. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Ocansey DKW, Zhang L, Wang Y, Yan Y, Qian H, Zhang X, Xu W, Mao F. Exosome-mediated effects and applications in inflammatory bowel disease. Biol Rev Camb Philos Soc 2020; 95(5): 1287–1307 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Noonin C, Thongboonkerd V. Exosome-inflammasome crosstalk and their roles in inflammatory responses. Theranostics 2021; 11(9): 4436–4451 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Zeng X, Zhang YD, Ma RY, Chen YJ, Xiang XM, Hou DY, Li XH, Huang H, Li T, Duan CY. Activated Drp1 regulates p62-mediated autophagic flux and aggravates inflammation in cerebral ischemia-reperfusion via the ROS-RIP1/RIP3-exosome axis. Mil Med Res 2022; 9(1): 25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Wu Y, Li J, Zeng Y, Pu W, Mu X, Sun K, Peng Y, Shen B. Exosomes rewire the cartilage microenvironment in osteoarthritis: from intercellular communication to therapeutic strategies. Int J Oral Sci 2022; 14(1): 40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Bu T, Li Z, Hou Y, Sun W, Zhang R, Zhao L, Wei M, Yang G, Yuan L. Exosome-mediated delivery of inflammation-responsive Il-10 mRNA for controlled atherosclerosis treatment. Theranostics 2021; 11(20): 9988–10000 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Pei W, Li X, Bi R, Zhang X, Zhong M, Yang H, Zhang Y, Lv K. Exosome membrane-modified M2 macrophages targeted nanomedicine: treatment for allergic asthma. J Control Release 2021; 338: 253–267 [DOI] [PubMed] [Google Scholar]
- 182.Long X, Yao X, Jiang Q, Yang Y, He X, Tian W, Zhao K, Zhang H. Astrocyte-derived exosomes enriched with miR-873a-5p inhibit neuroinflammation via microglia phenotype modulation after traumatic brain injury. J Neuroinflammation 2020; 17(1): 89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Deng F, Yan J, Lu J, Luo M, Xia P, Liu S, Wang X, Zhi F, Liu D. M2 macrophage-derived exosomal miR-590-3p attenuates DSS-induced mucosal damage and promotes epithelial repair via the LATS1/YAP/ β-catenin signalling axis. J Crohn’s Colitis 2021; 15(4): 665–677 [DOI] [PubMed] [Google Scholar]
- 184.Dad HA, Gu TW, Zhu AQ, Huang LQ, Peng LH. Plant exosome-like nanovesicles: emerging therapeutics and drug delivery nanoplatforms. Mol Ther 2021; 29(1): 13–31 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Kang Y, Xu C, Meng L, Dong X, Qi M, Jiang D. Exosome-functionalized magnesium-organic framework-based scaffolds with osteogenic, angiogenic and anti-inflammatory properties for accelerated bone regeneration. Bioact Mater 2022; 18: 26–41 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Bruno S, Pasquino C, Herrera Sanchez MB, Tapparo M, Figliolini F, Grange C, Chiabotto G, Cedrino M, Deregibus MC, Tetta C, Camussi G. HLSC-derived extracellular vesicles attenuate liver fibrosis and inflammation in a murine model of non-alcoholic steatohepatitis. Mol Ther 2020; 28(2): 479–489 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Budden CF, Gearing LJ, Kaiser R, Standke L, Hertzog PJ, Latz E. Inflammasome-induced extracellular vesicles harbour distinct RNA signatures and alter bystander macrophage responses. J Extracell Vesicles 2021; 10(10): e12127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Haghighitalab A, Dominici M, Matin MM, Shekari F, Ebrahimi Warkiani M, Lim R, Ahmadiankia N, Mirahmadi M, Bahrami AR, Bidkhori HR. Extracellular vesicles and their cells of origin: open issues in autoimmune diseases. Front Immunol 2023; 14: 1090416. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Kim S, Maeng JY, Hyun SJ, Sohn HJ, Kim SY, Hong CH, Kim TG. Extracellular vesicles from human umbilical cord blood plasma modulate interleukin-2 signaling of T cells to ameliorate experimental autoimmune encephalomyelitis. Theranostics 2020; 10(11): 5011–5028 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Casella G, Rasouli J, Boehm A, Zhang W, Xiao D, Ishikawa LLW, Thome R, Li X, Hwang D, Porazzi P, Molugu S, Tang HY, Zhang GX, Ciric B, Rostami A. Oligodendrocyte-derived extracellular vesicles as antigen-specific therapy for autoimmune neuroinflammation in mice. Sci Transl Med 2020; 12(568): eaba0599. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Tang TT, Wang B, Lv LL, Liu BC. Extracellular vesicle-based nanotherapeutics: emerging frontiers in anti-inflammatory therapy. Theranostics 2020; 10(18): 8111–8129 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Jeppesen DK, Fenix AM, Franklin JL, Higginbotham JN, Zhang Q, Zimmerman LJ, Liebler DC, Ping J, Liu Q, Evans R, Fissell WH, Patton JG, Rome LH, Burnette DT, Coffey RJ. Reassessment of exosome composition. Cell 2019; 177(2): 428–445.e18 [DOI] [PMC free article] [PubMed] [Google Scholar]