Next Article in Journal
Bioavailability Enhancement and Formulation Technologies of Oral Mucosal Dosage Forms: A Review
Previous Article in Journal
Transdermal Delivery of Botulinum Neurotoxin A: A Novel Formulation with Therapeutic Potential
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Exosome Source Matters: A Comprehensive Review from the Perspective of Diverse Cellular Origins

Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 33 Badachu Road, Shijingshan, Beijing 100144, China
*
Author to whom correspondence should be addressed.
Pharmaceutics 2025, 17(2), 147; https://doi.org/10.3390/pharmaceutics17020147
Submission received: 29 November 2024 / Revised: 2 January 2025 / Accepted: 14 January 2025 / Published: 22 January 2025
(This article belongs to the Special Issue Stem Cell Therapy and Nanodelivery Systems for Tissue Engineering)

Abstract

:
Exosomes have emerged as promising therapeutic agents in regenerative medicine. This review introduces a novel cell type-oriented perspective to systematically analyze exosomal properties in regenerative therapies. To our knowledge, this review is the first to comprehensively compare exosomes based on cellular source type, offering unprecedented insights into selecting optimal exosome producers for targeted regenerative applications. Factors beyond cellular origin influencing exosomal therapeutic efficacy, such as donor sites and collection methods, are also explored here. By synthesizing key advances, we propose promising research directions in the end. We aim to accelerate the development of more effective exosome-based regenerative therapies and highlight underexplored directions in this rapidly evolving field.

1. Introduction

Exosomes, a subset of extracellular vesicles with a general size range smaller than 200 nm in diameter, secreted by almost all cell types [1,2], serve as intercellular communicators, and play critical roles in multiple physiological and pathological processes [3]. Specifically, exosomes can promote tissue repair and regeneration, which is mainly attributed to their ability to carry a variety of bioactive molecules and deliver them to target cells, low immunogenicity, and high biosafety [4,5]. These characteristics enable exosomes to demonstrate significant application potential in tissue engineering, wound healing, neurodegenerative disease treatment, and cardiovascular disease intervention [6,7].
The isolation and characterization of exosomes are crucial steps in exosomal research. To improve the reproducibility and quality of research in this field, MISEV (Minimal Information for Studies of Extracellular Vesicles) guidelines play a critical role in ensuring that scientists adopt standardized isolation methods [2].
Before clinical application, it is essential to select the cellular sources of exosomes based on the specific intention, while optimizing the potential influencing factors to maximize therapeutic effects. Growing numbers of exosome-related research are emerging, but there is no review to systematically compare exosomes from various cell types when considering applications in regenerative medicine. This study will provide a comprehensive and novel perspective by reviewing relevant studies in Pubmed, Web of Science, and Embase databases, focusing on a cell type-oriented approach. We aim to provide a reference for choosing appropriate cellular sources of exosomes for specific usage, bridging the gap between regenerative medicine and nanomedicine.

2. Production, Isolation, Modification, and Storage of Exosomes

2.1. Production

There are two primary sources of exosome production: biological fluid and cell culture medium. Extensively studied biological fluids include serum, platelet-rich plasma, urine, cerebrospinal fluid, breast milk, and saliva. The production yield varies depending on parent cell types and the conditions under which the cells are cultured, or the biological fluid is preconditioned. For example, MSCs produce substantial exosomes [8], while dendritic cells produce a more limited number [9]. Preconditioning conditions, including oxygen levels and growth factors, significantly influence production [10]. Stimulating factors related to the yield can be divided into biochemical, mechanical, and exosome structural engineering [11]. Optimizing these factors is crucial for high yields of production.

2.2. Isolation and Purification

Exosomes’ overlap in size, density, content, and biological markers has made obtaining high exosomal purity challenging. Most existing technologies can not completely distinguish exosomes from lipoproteins with similar biophysical or biochemical properties and extracellular vesicles from non-endosomal pathways, leading to a low purity of desired exosomes [12].
Commonly used techniques include ultracentrifugation, size-exclusion chromatography (SEC), polymer precipitation, immunoaffinity capture, and microfluidics-derived techniques [13]. Ultracentrifugation is widely used due to its high recovery rate, but the co-isolation of other vesicles and the time-consuming process limit its applications [12]. SEC utilizes the size difference to isolate exosomes. Their procedures could largely preserve the integrity and bioactivity of exosomes but could not separate exosomes from other similar-size vesicles [14]. Every single technique above has advantages and disadvantages. Therefore, scientists have been trying to combine multiple complementary methods and found that the combination performs better in reducing contamination, improving separation purity, and maintaining the natural characteristics of exosomes [15].

2.3. Modification

Three main strategies for modifying exosomes are surface engineering, cargo loading, and genetic modification (Figure 1), with the goal of enhancing the therapeutic potential, targeting specificity, and stability.
Surface engineering involves modifying the exosomal membrane to improve targeting capabilities, circulation time, and uptake by specific cell types. Many scientists have summarized various surface engineering strategies, which can be categorized into modification at the cellular level and modification after exosomal isolation, or chemical and biological strategies [16,17,18]. For instance, Yu F et al. have engineered adipose stem cell-derived exosomes (ADSC-Exos) with specific peptide linkers to conjugate therapeutic molecules and have obtained ideal anti-inflammatory and tissue regenerative effects [19].
Cargo loading refers to incorporating therapeutic effector molecules, such as drugs, RNA, or proteins, into exosomes for specific purposes. These methods have also been comprehensively summarized by various studies [20,21]. It can be categorized into active loading and passive loading. Active loading normally means employing techniques such as electroporation, sonication, or extrusion to enhance cargo incorporation. Passive loading often refers to incubating exosomes with the desired cargo, allowing for diffusion-based incorporation, which includes co-culture, transfection, and incubation.
Genetic modification entails manipulating donor cells to express particular proteins or RNAs that are subsequently incorporated into exosomes. It can be divided into modification of parent cells and surface display via genetic engineering [22]. By transfecting donor cells with genes encoding desired therapeutic agents, these cells can produce exosomes enriched with specific proteins or RNAs. For instance, engineering cells to express therapeutic mRNA or siRNA enables the secretion of exosomes loaded with these genetic materials, facilitating targeted gene therapy applications [23].

2.4. Storage

To ensure the repeatability of studies on exosomal structure, content, and function, while keeping exosomes easy to transport and handle, scientists have developed three main storage techniques: cryopreservation, freeze-drying, and spray-drying.
For cryopreservation, exosomes are commonly stored at a temperature of 4 °C or −196 °C [12], but they might be negatively affected by repeated freezing and thawing [24]. Freeze-drying removes water from the exosome samples under low pressure [25], which stabilizes the exosomes and makes them easier to reconstitute. When lyophilized, exosomes can be stored at room temperature for extended periods. However, the type of cryoprotectants is crucial to prevent aggregation and ensure the exosomes’ functional integrity upon reconstitution [26]. Spray-drying converts the liquid exosome suspension into a dry powder by rapidly drying it with hot gas, offering easy transportation [12]. But high temperatures potentially denature sensitive proteins on the exosome surface, making it less suitable for exosomes with heat-sensitive components [27].

3. Factors Influencing the Efficacy of Exosome-Based Applications in Tissue Regeneration

Several other factors critically affect the therapeutic outcomes of exosome-based interventions, including donor cell condition, dosage, administration route, and delivery vehicles used (Figure 2).

3.1. Donor Cell Condition

Age, health status, donor sites, collection methods, and preconditioning methods of donor cells are non-negligible factors that affect the therapeutic outcomes.
Aging in donor cells generally leads to a decline in the quality of exosomes, which usually manifests as reduced anti-inflammatory and regenerative capabilities. For instance, exosomes derived from older BMSCs exhibit diminished effects in osteogenic and lipogenic abilities compared to those derived from younger BMSCs [28]. In addition, cells with different health statuses may produce exosomes with significant differences in morphology, function, or molecules. Cells derived from diseased individuals produce exosomes with altered molecular profiles, leading to impaired functionality or adverse effects. Oncoproteins, including EGFR, GRB2, and SRC, have been reported to be enriched in the non-small cell lung cancer (NSCLC) exosomes, which promote the cancer’s development [29]. Different sites and collection methods may also lead to exosomes with varying traits. Visconte C et al. reported that abdominal adipose tissue obtained by surgical excision and lipoaspiration might produce exosomes with distinct characteristics [30]. They also demonstrated that exosomes released by ADSCs from different anatomical locations contained a different abundance of miRNAs. Preconditioning strategies, such as hypoxia or cytokine stimulation, can change the regenerative potential of donor cells and their exosomes. Hypoxia could upregulate pro-angiogenic factors in MSCs, improving exosomal abilities to promote neovascularization and tissue repair in mouse models of femoral fracture [31]. Preconditioning methods with rapamycin and growth factor deprivation have also been proven to increase the release of exosomes [32].

3.2. Dosage

Higher exosomal doses generally lead to a more pronounced regenerative effect; however, there is a threshold beyond which additional exosomes may not provide further benefits or may even cause adverse effects (Table 1). In traumatic brain injury (TBI) models, researchers found that compared with 50 μg or 200 μg groups, 100 μg exosomes per rat showed more significant efficacy in improving sensory-motor and cognitive function, reducing hippocampal neuron cell loss and promoting neurogenesis [33]. The therapeutic dose of exosomes commonly ranges from 10 to 100 μg of protein in mouse models [34], but evaluation of the effective dose in specific conditions is lacking.

3.3. Administration Route

The route of exosomal administration substantially affects exosomal capabilities [41]. The most frequently applied routes include systemic (intravenous), local (e.g., intra-muscular, intra-articular, or intra-dermal), and targeted delivery methods.
Systemic administration is usually applied to deliver exosomes throughout the body, targeting multiple sites simultaneously. Although it is beneficial in treating conditions where widespread distribution is needed, rapid clearance by the liver, spleen, and kidneys decreases the therapeutic efficacy, and non-specific uptake by off-target tissues can reduce the concentration of exosomes at the desired site [42]. In contrast, the biggest advantage of local administration is the high exosomal levels at the target site and the low risk of systemic side effects [43]. This route is commonly used for conditions where localized action is desired [44]. However, repeated administration procedures might be required to maintain adequate levels at the site. Targeted delivery methods, such as ligand-modified exosomes or magnetic targeting, enable precise delivery to diseased tissues [45]. Maximizing efficacy while minimizing off-target effects, they have become attractive research hotspots. Superparamagnetic iron oxide nanoparticles (SPIONs) possess excellent magnetic properties and can be concentrated using an external magnetic field [46]. Combining the magnetic targeting properties of SPIONs with the homing abilities of exosomes, scientists have achieved a “dual targeting” effect and excellent therapeutic efficacy in NSCLC treatment [47].

3.4. Delivery Vehicles

To enhance the effectiveness and address the drawbacks of existing therapies, growing numbers of studies have been exploring new materials as delivery vehicles.
Hydrogels are effective delivery systems due to their sustained release, biocompatibility, and biodegradability [48]. They can be engineered to respond to environmental stimuli, such as pH or temperature, to achieve controlled exosome release [49]. Nevertheless, controlling the hydrogels’ degradation rate to align with the tissue regeneration process is a crucial challenge. Functionalized with ligands, nanoparticles (NPs) become practical tools to protect exosomes from rapid clearance [50]. Nonetheless, targeted ligands may trigger an immune response, and certain NP materials’ biocompatibility and potential toxicity remain significant hurdles [51]. Various scaffold materials are used as exosomal delivery platforms to provide controlled release. Collagen-based scaffolds are frequently chosen due to collagen’s role as a major component of the extracellular matrix (ECM) and its natural involvement in tissue repair processes. Promoting cell adhesion and proliferation notwithstanding, their mechanical properties may be inadequate in load-bearing tissues [52]. In these scenarios, the lack of long-term structural integrity in collagen can hinder the sustained bioactive function of exosomes at the injury site.

4. Exosomes from Different Cellular Sources

The regenerative potential of exosomes is intricately tied to their cellular origin, with each source imparting unique properties and functionalities to the secreted vesicles. This cellular specificity not only underlies the diverse applications but also highlights the need for a thorough understanding of how different sources of exosomes contribute to tissue restoration. Currently, several cellular sources have been broadly studied (Figure 3), such as MSCs, immune cells, neural stem cells (NSCs), fibroblasts, umbilical vein endothelial cells (UVECs), and Schwann cells (SCs).

4.1. MSC-Derived Exosome

MSCs are a diverse group of multipotent stromal cells capable of differentiating into various cell types, which is also evident in their exosomes (MSC-Exos) [53].
Extensively studied MSC types as sources of exosomes include bone marrow MSC (BMSC), ADSC, umbilical cord MSC (UCMSC), placental MSC (PMSC), and DPSC.

4.1.1. BMSC-Derived Exosome

BMSC-derived exosomes (BMSC-Exos) have attracted research attention since the early years, correlated studies are relatively more extensive than other types of exosomes [54].
For wound healing, BMSC-Exos outperform exosomes derived from ADSC and UCMSC in enhancing the proliferation of dermal fibroblast (DF) [55]. Pomatto M et al. reported that, unlike ADSC-derived extracellular vesicle proteins, proteins uniquely enriched in BMSC-derived extracellular vesicles (BMSC-EVs) are associated with cell adhesion, glycolysis, and fructose–galactose metabolism, suggesting that BMSC-Exos mainly promote cell proliferation [56]. However, their results indicated that BMSC-Exos might be inferior to ADSC-derived exosomes (ADSC-Exos) in achieving optimal effects, possibly due to insufficient angiogenic abilities. This conclusion contrasts with the later findings of Soni et al., who demonstrated that BMSC-Exos have superior angiogenesis-promoting capabilities [57].
BMSC-Exos exhibit a high propensity to promote bone tissue regeneration due to their enrichment in osteogenic factors like microRNA-148a and long non-coding RNA-H19 [58,59]. Additionally, studies have shown that BMSC-Exos possess enhanced immunomodulatory effects, creating a favorable microenvironment for healing [57]. In addition, their beneficial effects in other fields such as cardiovascular diseases and neural regeneration were also reported [60,61].
However, obtaining BMSCs from primary sources involves an invasive and painful procedure, which poses a notable disadvantage when contrasted with more readily accessible sources of MSCs [62,63]. This invasiveness can limit the scalability of BMSC-Exo production and may reduce donor willingness. BMSC-Exos are also highly susceptible to age-related changes in their parent cells. As donor age increases, their regenerative capacity and therapeutic efficacy may decline more significantly than exosomes from other MSC sources [64].
Apart from the primary sources mentioned above, it is essential to develop sustainable and scalable alternatives such as cell lines. Commonly used BMSC lines are immortalized cell lines like hTERT-BMSC (human telomerase reverse transcriptase-BMSC) [65], which exhibit extended proliferative capacity. Though more scalable, immortalized BMSC lines may differ phenotypically and functionally from their primary counterparts, raising concerns about their translational relevance [66].

4.1.2. ADSC-Derived Exosome

ADSC exhibits shorter population doubling time and stronger antiapoptotic potential than other types of MSC [67]. Studies have also indicated that ADSCs yield more exosomes with simpler isolation techniques and lower invasiveness [68]. Furthermore, the increasing prevalence of overweight and obesity due to improved modern living standards has made acquiring adipose stem cells more feasible.
A distinctive feature of ADSC is higher lipid content. The elevated lipid composition contributes to their enhanced paracrine signaling capacity, mediated by exosomes. They promote tissue repair through increased cellular communication, improved membrane fusion, and better interaction with target cells [69]. Enriched with anti-inflammatory cytokines, growth factors, and miRNAs, ADSC-Exos can modulate the local immune environment and promote angiogenesis and adipogenesis [70]. Their anti-fibrotic effects were also observed, which is beneficial for scar reduction [71].
Emerging evidence suggests that the source of adipose tissue used to derive ADSCs significantly impacts the properties of the resulting exosomes. Studies examining ADSCs from various fat depots, such as subcutaneous, visceral, perivascular, and infrapatellar fat, have revealed depot-specific variations in exosomal composition and therapeutic efficacy. For instance, the expression levels of key chondrogenic and osteogenic genes in cells isolated from infrapatellar fat pad (IPFP) were proved to be higher than those from subcutaneous adipose tissue, indicating IPFP might be a better source for ADSC-Exos for cartilage and bone regeneration [72]. For subcutaneous adipose tissue, there are also variations between different anatomical locations [73]. These depot-specific differences further highlight the versatility for customization of ADSC-Exos. Despite the abundance of sources, ADSC-Exos from elder individuals may be less effective than those from younger individuals in terms of therapeutic outcomes, as ADSCs derived from different age ranges might have significant differences in function [74].
In addition, ADSC-Exos were also revealed to possess therapeutic effects on inflammation-related diseases (e.g., Crohn’s disease, arthritis, etc.), myocardial ischemia, hair loss, and delayed photoaging [75,76,77].
Primary cultures of ADSCs were indicated to be inferior to their commercially obtained counterparts in cell growth [78]. Regenerative capabilities of immortalized adipose-derived mesenchymal stem cell lines have been revealed [79], but their application in exosomal research needs more investigation.

4.1.3. Human UCMSC-Derived Exosome

In recent years, there has been a surge in research exploring the therapeutic potential of human UCMSC-derived exosomes (hUCMSC-Exos).
Firstly, the collection methods of hUCMSCs are much simpler than those for most other MSC sources. Derived from umbilical cord blood, hUCMSCs can be obtained through a painless procedure simply by extracting blood from the umbilical cord. In contrast, harvesting MSCs from alternative sources such as bone marrow or adipose tissue typically involves invasive techniques.
Existing studies have exhibited superior regenerative properties of hUCMSC-Exos. A study analyzing exosomes derived from BMSCs, ADSCs, and hUCMSCs demonstrated the greatest capacity of hUCMSC-Exos to promote keratinocyte migration [55]. Scientists have conducted a systematic analysis of protein components in these three types of exosomes, indicating that hUCMSC-Exos are more prominent in tissue repair [80].
Due to the notable immunomodulation ability, hUCMSC-Exo is regarded as an effective strategy for treating inflammatory or autoimmune diseases. Applications of hUCMSC-Exos in inflammatory bowel disease have been a research hotspot in recent years [81,82]. Periocular injections of hUCMSC-Exos have shown substantial inhibition of autoimmune uveitis progression in rats [83]. Moreover, complete subacute spinal cord injury (SCI) patients after intrathecal injection of hUCMSC-Exos have observed functional improvements [84]. These exosomes can also obviously mitigate graft-versus-host disease injury by alleviating the oxidative–reductive metabolic dysfunction [85].
However, several issues must be considered. Maternal factors such as maternal obesity and diabetes have been widely studied. Maternal obesity influences the metabolism and bioenergetic profile of hUSMSCs [86], while gestational diabetes mellitus (DM) might contribute to premature aging and mitochondrial dysfunction [87]. Additionally, immortalized hUCMSCs have been reported to exert therapeutic influence in other fields [88], but the use of them in exosomal research remains blank.

4.1.4. Human PMSC-Derived Exosome

The human placenta serves as a crucial source of MSC-Exos. The availability of this tissue is well established, as the placenta is typically discarded as medical waste after birth and can be readily obtained with minimal ethical concerns [89]. PMSCs have attracted extensive attention due to their unique biological characteristics. However, there is still no widely recognized “standard” commercial immortalized PMSC cell line. Therefore, most of the existing studies on PMSCs use cells that are directly isolated from placental tissue.
The placenta tissue can be divided into four layers: amniotic membrane (AM), chorionic membrane (CM), chorionic villi (CV), and deciduae (DC) [90]. Each layer yields distinct MSCs: AMMSCs, CMMSCs, CVMSCs, and DCMSCs, respectively. Current research focusing on human AMMSC-derived exosomes (AMMSC-Exos) is relatively abundant, whereas studies on others remain limited.

Human AMMSC-Derived Exosome

AMMSCs and their secretome have consistently exhibited immune-regulatory capabilities, such as suppressing T cell proliferation induced by alloantigens and shifting the macrophage 1 (M1)/macrophage 2 (M2) ratio of synovial macrophages in osteoarthritis [91]. AMMSC-Exos can also reduce the number of Kupffer cells and the levels of inflammatory cytokines in nonalcoholic steatohepatitis rat models [92]. Although a few studies have directly examined AMMSC-Exos alongside exosomes from other cell types, comparisons between their parent cells offer valuable insights into the potential of AMMSC-Exos. Topoluk N et al. demonstrated that AMMSCs possess stronger chondroprotective abilities than ADSCs and can effectively mitigate cartilage damage induced by macrophages, an effect not observed with human ADSCs [93].
A significant advantage of AMMSC-Exos is their small particle size, with a mean diameter of 72 nm, notably smaller than the mean sizes of ADSC-Exos (220 nm) and UCMSC-Exos (120 nm), facilitating their passage through physiological barriers [94].
For nervous system regeneration, AMMSC-Exos were found to effectively enhance neuron survival by regulating cell apoptosis and improving the neurobehavioral function of cerebellar palsy rat models [95]. In acute traumatic spinal cord injury (TSCI) rats, human AMMSC-Exos were shown to significantly reduce the lesion volume [96].
In wound healing, scientists have found that combining acellular amniotic membrane scaffolds with ADSC-Exos significantly enhances the therapeutic effects on diabetic wound healing compared to the single application of ADSC-Exos [97]. Subsequently, Noh CH et al. demonstrated that exosome-rich conditioned medium from AMMSCs improved whole-skin-excision rats’ wound healing, with rapid wound closure and reduced scar tissue formation [94].

Other Human PMSC-Derived Exosome

Research correlated with exosomes from CMMSCs (CMMSC-Exos), CVMSCs (CVMSC-Exos), or DCMSCs (DCMSC-Exos) is limited, but the existing studies could provide a reference for future exploration.
CMMSC-Exos could be absorbed by multiple cells (including MSCs derived from synovial fibroblasts, osteoblasts, and periosteum) isolated from tissues associated with osteoarthritis, suggesting their potential in the treatment of osteoarthritis [98]. CVMSC-Exos have shown anti-cancer effects on ovarian cancer cells and promote trophoblast migration and proliferation [99]. Salomon C et al. isolated CVMSC from placental villi and further obtained CVMSC-Exos from acellular CVMSC, finding that CVMSC-Exos contribute to placental adaptation to low oxygen [100]. Zhang CP’s group provided evidence for the regenerative effects of DCMSC-Exos, they revealed that DCMSC-Exos can promote the proliferation, migration, and differentiation of high-glucose-induced senescent fibroblasts [101].

4.1.5. DPSC-Derived Exosome

DPSCs can be obtained during routine dental procedures [102] and exhibit robustness under cryopreservation [103]. Exosomes derived from DPSCs (DPSC-Exos) possess higher drug-loading efficiency than many other MSC-derived exosomes [104]. Notably, unlike the common effect of aging on the regenerative ability of exosomes, senescent DPSC-derived exosomes can improve the antioxidant ability, proliferation, migration, and survival rate of young DPSCs [105].
DPSC-Exos demonstrate exceptional efficacy in oral diseases, surpassing other exosomes. When isolated from odontogenic differentiation culture conditions, DPSC-Exos can increase the expression of genes necessary for odontogenic differentiation in vitro and promote dental pulp regeneration in vivo [106,107]. They were also proven to have stronger immunoregulation activity, anti-necrosis, and anti-apoptosis capabilities than BMSC-Exos [108,109]. Shen Z et al. claimed that combining DPSC-Exos with chitosan hydrogel effectively inhibited periodontal inflammation and reduced epithelial damage and alveolar bone loss [110], consistent with Zheng J et al. [111].
In nervous system regeneration, DPSC-Exos were found to reduce neuronal apoptosis by transferring miR-877–3p, improving brain edema and infarct volume in cerebral ischemia–reperfusion injury (I/R) rat models [112]. Liang X et al. reported that DPSC-Exos attenuated the neuroinflammation and microglial pyroptosis in subarachnoid hemorrhage through miR-197-3p/FOXO3 axis [113]. In addition, they can enhance sciatic nerve regeneration by increasing the secretion of neurotrophic factors, proliferation, and migration of SCs [114].
DPSC-Exo is also widely studied in other areas such as bone-related realms, flap transplantation, and wound healing [115,116]. Compared with BMSC-Exos and ADSC-Exos, isolating sufficient quantities of DPSC-Exos remains a significant challenge, which needs more innovative technologies [117].
Similarly, commercialized or immortalized DPSC lines have been researched in some fields [118,119]. But the tumorigenic potential and long-term influence are not fully explored and there is a long way before applying these cell lines to an exosome-related field.

4.2. Immune Cell-Derived Exosome

The overall body environment is nonnegligible for tissue regeneration. Immune modulation is a key entry point for treating inflammation and autoimmune diseases. While MSC-Exos exhibit certain immunomodulatory properties, immune cells serve as a potential source of exosomes with more specific effects [120].
In contrast to MSC-Exos, the potential effects of immune cell-derived exosomes still harbor many unknowns. There has been relatively more literature on exosomes derived from macrophages and T cells.

4.2.1. Macrophage-Derived Exosome

Macrophages exist in various activation states (e.g., M1 pro-inflammatory and M2 anti-inflammatory), with different contents and functions of exosomes. M1 macrophages and their exosomes are known for pro-inflammatory effects, which might adversely affect tissue regeneration [121]. Therefore, related research on M1 macrophage-derived exosomes is limited. Meanwhile, M2 macrophages are widely acknowledged to enhance inflammation regression and tissue repair. Exosomes derived from them (M2-Exos) replicate multiple functions of M2 macrophages [122], exhibiting promising immunomodulation properties. In this section, we will review the applications of M2-Exos in regenerative medicine.
M2-Exos possess a strong capability to guide the phenotypic switch of M1 to M2 macrophages, providing a promising strategy for the therapy of diseases associated with imbalances of pro-inflammatory and anti-inflammatory responses. Due to this, they were revealed to facilitate diabetic fracture healing [122]. CCL24 and MFG-E8, abundant in M2-Exos, are key regulators of this switch, promoting wound repair by strengthening angiogenesis and epithelialization [123]. In addition, M2-Exos can enhance tendon-to-bone healing in aged rats by alleviating cellular senescence and improving the chondrogenic potential of BMSCs [124]. Other properties, such as promoting functional recovery after SCI, have also been reported [125].
In addition to primary macrophage cultures mentioned above, macrophage cell lines, such as the commonly used THP-1, have also been employed to generate exosomes for various research [126]. They are robust and easy to culture, offering a more consistent and reproducible source of exosomes compared to primary macrophages, which are often subject to donor variability and difficult to maintain over extended periods. For instance, RAW 264.7-derived M2-Exos have been reported to facilitate osteogenesis and promote bone regeneration after specific modification [127,128].
Notably, some miRNAs contained in M2-Exos may have unintended effects on tumor biology. For example, some M2-Exos content can increase tumor metastasis and progression [126,129], requiring a more comprehensive investigation.

4.2.2. T Cell-Derived Exosome

T cells, a critical component of the immune system, encompass various subtypes. Regulatory T cell (Treg)-derived exosomes (Treg-Exos) possess potent immunomodulatory properties and have a strong research foundation in regenerative medicine [130].
Treg-Exos exhibit remarkable potential in modulating immune responses during tissue repair processes. Their ability to suppress effector T cells and promote tolerance makes them particularly valuable in scenarios where fine-tuning the immune system is crucial for optimal healing [120].
The application of Treg-Exos in transplantation rejection has been researched widely. In the humanized mouse skin transplantation model, human Treg-Exos were observed to inhibit homologous immunity-mediated skin tissue damage by reducing immune cell infiltration and prolonging skin allograft survival [131]. Beyond this, Treg-Exos can also prolong the survival time of rat kidney transplantation models [132] and rat orthotopic liver transplantation models [133].
Moreover, Treg-Exos were found to exert beneficial effects in the field of neural injuries, wound healing, and cardiovascular diseases. Yang C et al. demonstrated that Treg-Exos could prevent the apoptosis and inflammation of BV-2 microglia induced by oxygen–glucose deprivation/reperfusion [134]. Treg-Exos could also promote the phenotypic switch of macrophages, facilitating angiogenesis and tissue remodeling for successful diabetic wound healing [135]. For acute myocardial infarction, Treg-Exos were observed to improve cardiac function by promoting macrophage M2 polarization [136].
Several challenges must be addressed. For example, Tregs can be categorized into two types with distinct properties: those directly derived from the thymus and peripheral-derived Tregs (pTregs) [134]. Similar variations in Treg cell source types and isolation methods of Tregs and exosomes may lead to heterogeneity of the effects of Treg-Exos. Additionally, stable Treg cell lines that can be used for exosomal research are still lacking, and relevant studies are limited.

4.3. NSC-Derived Exosome

NSC-Exos have also become a promising focus. One of the most famous properties is their neuroregenerative effect. Apparent decreases in infarct size and brain atrophy in murine thromboembolic stroke models were observed after applying NSC-Exos [137], effects not typically observed with MSC-Exos [138]. They can also mediate autophagy to inhibit neuroinflammation and promote functional recovery in SCI model rats at an early stage [139]. Furthermore, their function to promote neural recovery was reported to surpass those of MSC-Exos [137].
Another unique advantage is their potential to improve the integrity of the blood–brain barrier (BBB). Liu Y et al. established a BBB model using 5 × FAD primary cerebral endothelial cells and NSC-Exos, which reversed BBB defects caused by Alzheimer’s disease [140].
Beyond neuroregenerative effects, NSC-Exos were found to support wound repair and might play roles in controlling systemic aging speed. These exosomes contain abundant neurotrophic factors such as neuron-derived neurotrophic factor (NDNF) and immunoregulation proteins [141]. Zhang Y et al. transplanted healthy hypothalamic NSCs into the aging brain, finding that the exosomal miRNAs from these cells can decelerate aging [142].
A few research studies in other fields have used commercial or immortalized NSC lines, but primary NSCs remain the most widely studied source of exosomes currently. Nevertheless, establishing a stable amplification system without heterologous components and defining a method for single-exosome NSC analysis are still needed to improve NSC-Exos application [143].

4.4. Exosome Derived from Other Cellular Origins

4.4.1. Fibroblast-Derived Exosome

Fibroblasts are indispensable for the body’s response to injuries, the synthesis and remodeling of the local ECM, and the formation of new tissue—processes in which exosomes produced by them (FC-Exos) play a crucial role [144]. For FC-Exos, exosomes can be obtained from primary fibroblasts and commercial or immortalized cell lines.
Compared with MSCs, primary fibroblasts can be isolated from the skin through less invasive procedures and are more closely associated with wound healing [145]. Scientists have identified several mechanisms underlying FC-Exos’ beneficial effects on wound healing, such as accelerating local collagen deposition and maturation [146]. In neural regeneration, primary fibroblasts derived from the sciatic nerve of rats were observed to enhance axonal regeneration and contribute to Schwann cell-mediated myelination [147,148,149].
Commercial or immortalized cell lines of fibroblasts have been extensively studied. They were found to possess antioxidant activity and could prevent ultraviolet B-induced senescence, indicating excellent potential as an anti-photoaging strategy [150]. Hu S et al. compared the anti-skin-aging capabilities of exosomes derived from three-dimensional spheroids of human DFs (3D HDF-XOs) with those of MSC-Exos, finding that 3D HDF-XOs are more effective in reducing skin aging [151]. Jang YN et al. demonstrated that human DF-neonatal-derived exosomes might exert anti-inflammation activity and improve the recovery of damaged skin barrier in atopic dermatitis [152].

4.4.2. Human Umbilical Vein Endothelial Cell-Derived Exosome

Exosomes derived from human umbilical vein endothelial cells (HUVEC-Exos) have been applied in many regenerative areas. At present, HUVECs in the majority of existing studies were purchased from suppliers, which facilitates their exosomal research.
One of the most extensively studied applications is cutaneous repair. Scientists reported that HUVEC-Exos could facilitate diabetic wound healing when pretreated under hypoxic conditions or cooperated with bioengineered scaffolds [153]. Interestingly, when pretreated with advanced glycation end products to mimic conditions in DM, HUVEC-Exos were found to delay wound healing by modulating fibroblast autophagy [154]. In common skin wounds, apoptotic HUVEC-Exos could promote skin repair by increasing angiogenesis [155]. However, contrasting findings by Qi L et al. suggested that the exosomal miR-106b, which diminished the adhesion and viability of fibroblasts and keratinocytes, might harm wound healing [156].
For pro-angiogenic function, Guo L et al. demonstrated that pretreating HUVEC-Exos with an appropriate centration of H2O2 can enhance the angiogenic ability of endothelial progenitor cells, leading to increased skin flap survival [157]. Maiullari F et al. explored a 3D bioprinting strategy and revealed that HUVEC-Exos could be potentially employed as bioadditives for the formulation of bioinks, supporting the formation of a new functional vasculature when being loaded onto the bioprinted 3D structures [158]. Regarding neuroprotective effects, HUVEC-Exos have been proven to attenuate inflammation and apoptosis of neural cells [159] and protect nerve cells against I/R injuries [160]. In bone regeneration, HUVEC-Exos can drive osteogenic differentiation and boost the migratory potential of BMSCs [161].
There are still many unknowns. For example, some characteristics of HUVEC-Exos remain controversial as mentioned previously (e.g., the effects on wound healing). For preconditioning methods, contrary to the common belief that longer treatment times yield stronger effects, a 3-h hypoxic treatment had a similar or even weaker impact on HUVEC exosomes compared to a 15 min treatment [162].

4.4.3. Schwann Cell-Derived Exosome

Due to the close relationship with the nervous system, exosomes derived from SCs (SC-Exos) have garnered significant attention in neural treatment.
In existing exosome-related studies, primary SCs could be isolated from sciatic nerves or derived from skin precursors (SKPs). For peripheral nerve regeneration, Lopez-Verrilli MA et al. first reported in 2013 that SC-Exos significantly increased axonal regeneration in vitro and enhanced the regenerative capacity of the post-injury sciatic nerve in vivo [163]. Subsequent research showed that this effect might be primarily produced by exosomes derived from repair Schwann cells (rSCs) rather than differentiated Schwann cells (dSCs) [164]. Moreover, exosomes derived from SKP-derived SCs could enhance the survival and repair of sensory neurons after oxygen–glucose deprivation (OGD) exposure [165], facilitate axon regeneration [166] and alleviate denervation-induced muscle atrophy [167].
Although SCs are an important component of PNS, their exosomes could also affect the regeneration after SCI. Pan D et al. identified that SC-Exos could induce axonal protection after SCI by enhancing autophagy and reducing apoptosis [168]. Consistent with this, Xu B et al.’s study showed that SC-Exos could activate mitophagy mediated by the AMPK pathway and improve mitochondrial dysfunction and necroptosis after SCI [169]. In bone tissue engineering, hydrogel-encapsulated SC-Exos optimized the microenvironment for bone regeneration by improving innervation, immunoregulation, angiogenesis, and osteogenesis [170].
Li Z et al. used rat cell lines of SCs (RSC96) and observed promoting dental pulp regeneration effects of their exosomes [171]. In addition, their multiple beneficial regenerative effects were also revealed in the in vitro model of cyclic mechanical strain (CMS)-induced dorsal root ganglion (DRG) cell injury [172].
It is undeniable that some properties of SC-Exos limit their application. For example, because of their terminal status, the culturing of primary SCs is complex and costly [173]. Moreover, SC-Exos’ regenerative capabilities might decrease after remaining denervated without axonal contact for a prolonged period, which needs more exploration and improvement [174].

5. Future Insights

Table 2 consolidates all the examples of exosome applications provided in the above text. There are many other cellular sources of exosomes showing regenerative effects, such as cardiosphere-derived cells (CDC) and hepatocytes. For instance, CDC-derived exosomes (CDC-Exos) could improve cardiac function after acute myocardial infarction [175]. Extracellular vesicles produced by hepatocytes were proven to promote liver regeneration [176].
In general, exosomes offer new possibilities for tissue regeneration across a wide range of medical conditions (Figure 4). Firstly, their acellular nature mitigates many safety concerns associated with cell transplantation [177]. Secondly, exosomes can cross biological barriers due to their nano-size, more efficiently than larger particles or cells to reach target tissues [178]. Thirdly, exosomes exhibit lower immunogenicity than whole cells, reducing the risk of immune rejection and potentially allowing for allogeneic applications [179]. These characteristics significantly broaden the donor pool and increase the scalability of exosome-based therapies. So far, researchers have compared the properties of different cellular exosomes in specific conditions. Table 3 shows studies comparing different types of exosomes, aiming to provide a reference for choosing the cellular origin of exosomes.
Several promising but underexplored strategies in exosome research warrant further investigation (Figure 5).
Firstly, technological innovations in exosome isolation, characterization, and large-scale production methodologies represent a critical area for advancement. Current techniques often yield heterogeneous exosome populations, potentially confounding research findings and limiting therapeutic efficacy. Developing novel approaches for high-purity and high-yield exosome isolation is crucial for research and clinical applications. Additionally, establishing standardized protocols for exosome characterization, encompassing physical and molecular attributes, is essential for ensuring reproducibility across studies and translating research findings into clinical practice.
While promising, current exosome modification methodologies are often limited in their precision and scalability. Future investigations should focus on developing techniques for fine-tuning exosome surface proteins and internal cargo with unprecedented specificity. This could involve the integration of synthetic biology principles, such as using engineered cell lines or artificial exosome-mimetic nanovesicles, to produce exosomes with tailored therapeutic properties. Such advancements may lead to the creation of “designer exosomes” capable of addressing specific regenerative challenges with enhanced efficacy and reduced off-target effects.
While the immunomodulatory properties of certain exosome populations have been documented, our understanding of the complex interplay between exosomes and various immune cell subsets remains limited. Delineating the mechanisms by which exosomes modulate immune responses in different tissue microenvironments could inform the development of exosome-based therapies that promote tissue regeneration and create a favorable immune milieu conducive to healing. Moreover, investigating the potential of exosomes in mitigating autoimmune responses or promoting tolerance in the context of tissue transplantation might open new avenues in regenerative medicine and beyond.
As we contemplate the future of exosome research, it is imperative to consider the long-term effects and potential risks. While short-term safety profiles have generally been favorable, comprehensive longitudinal studies are needed to assess the long-term impact of exosome treatments on multiple aspects such as tissue homeostasis, cellular senescence, and potential oncogenic risks. Therefore, employing advanced in vivo imaging techniques and molecular profiling methods to track the fate and effects of administered exosomes over extended periods is also essential.
Integrating exosome biology with emerging fields such as 3D bioprinting presents an exciting frontier. Incorporating exosomes into bioengineered scaffolds or using exosome-laden bioinks in 3D printing applications to enhance tissue regeneration and vascularization deserves further exploration. This approach could develop more complex, functional tissue constructs that better recapitulate native tissue architecture and function.
Another underexplored area is the exosome’s potential role in epigenetic reprogramming for tissue regeneration. Exosomes carry a variety of epigenetic modifiers, such as non-coding RNAs, which can alter the epigenetic landscape of recipient cells. Harnessing this epigenetic modulation capacity for regenerative purposes is a promising research direction. Profiling the epigenetic cargo of exosomes from different cell types, developing methods to selectively enrich exosomes with specific epigenetic modifiers, and investigating the long-term stability of exosome-induced epigenetic changes in regenerating tissues are crucial aspects to explore. Investigating the potential of exosomes in reprogramming somatic cells into induced pluripotent stem cells or directly into specific cell types needed for tissue repair, without genetic manipulation, could open new avenues for precise and efficient tissue regeneration.
To the best of our knowledge, the present study is the first review to discuss the applications of exosomes in regenerative medicine from the perspective of cellular sources, aiming to provide a reference for researchers to choose the cell type of exosome sources. The future research directions in exosome-based regenerative medicine are diverse and interconnected, necessitating a holistic approach that spans basic science, translational research, and clinical development. Addressing these multifaceted challenges could unlock the transformative potential of exosomes in regenerative medicine, ushering in a new era of personalized and cell-free therapies.

Author Contributions

Conceptualization, Y.C.; formal analysis, F.N., W.Q. and Z.W.; writing—original draft preparation, Y.C.; writing—review and editing, Y.C. and F.N.; visualization, Y.C. All authors have read and agreed to the published version of the manuscript.

Funding

This study was funded by CAMS Innovation Fund for Medical Sciences (2021-I2M-1-068).

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Kourembanas, S. Exosomes: Vehicles of Intercellular Signaling, Biomarkers, and Vectors of Cell Therapy. Annu. Rev. Physiol. 2015, 77, 13–27. [Google Scholar] [CrossRef] [PubMed]
  2. Welsh, J.A.; Goberdhan, D.C.I.; O’Driscoll, L.; Buzas, E.I.; Blenkiron, C.; Bussolati, B.; Cai, H.; Di Vizio, D.; Driedonks, T.A.P.; Erdbrügger, U.; et al. Minimal information for studies of extracellular vesicles (MISEV2023): From basic to advanced approaches. J. Extracell. Vesicles 2024, 13, e12404. [Google Scholar] [CrossRef] [PubMed]
  3. Kalluri, R.; LeBleu, V.S. The biology, function, and biomedical applications of exosomes. Science 2020, 367, eaau6977. [Google Scholar] [CrossRef] [PubMed]
  4. O’brien, K.; Breyne, K.; Ughetto, S.; Laurent, L.C.; Breakefield, X.O. RNA delivery by extracellular vesicles in mammalian cells and its applications. Nat. Rev. Mol. Cell Biol. 2020, 21, 585–606. [Google Scholar] [CrossRef]
  5. Zhang, Y.; Liu, Y.; Liu, H.; Tang, W.H. Exosomes: Biogenesis, biologic function and clinical potential. Cell Biosci. 2019, 9, 19. [Google Scholar] [CrossRef]
  6. Han, C.; Yang, J.; Sun, J.; Qin, G. Extracellular vesicles in cardiovascular disease: Biological functions and therapeutic implications. Pharmacol. Ther. 2022, 233, 108025. [Google Scholar] [CrossRef]
  7. Zhang, S.; Chu, W.C.; Lai, R.C.; Lim, S.K.; Hui, J.H.P.; Toh, W.S. Exosomes derived from human embryonic mesenchymal stem cells promote osteochondral regeneration. Osteoarthr. Cartil. 2016, 24, 2135–2140. [Google Scholar] [CrossRef]
  8. Chen, T.S.; Arslan, F.; Yin, Y.; Tan, S.S.; Lai, R.C.; Choo, A.B.H.; Padmanabhan, J.; Lee, C.N.; De Kleijn, D.P.V.; Lim, S.K. Enabling a robust scalable manufacturing process for therapeutic exosomes through oncogenic immortalization of human ESC-derived MSCs. J. Transl. Med. 2011, 9, 47. [Google Scholar] [CrossRef]
  9. Luo, S.; Chen, J.; Xu, F.; Chen, H.; Li, Y.; Li, W. Dendritic Cell-Derived Exosomes in Cancer Immunotherapy. Pharmaceutics 2023, 15, 2070. [Google Scholar] [CrossRef]
  10. Cai, H.; Huang, L.-Y.; Hong, R.; Song, J.-X.; Guo, X.-J.; Zhou, W.; Hu, Z.-L.; Wang, W.; Wang, Y.-L.; Shen, J.-G.; et al. Momordica charantia Exosome-Like Nanoparticles Exert Neuroprotective Effects Against Ischemic Brain Injury via Inhibiting Matrix Metalloproteinase 9 and Activating the AKT/GSK3β Signaling Pathway. Front. Pharmacol. 2022, 13, 908830. [Google Scholar] [CrossRef]
  11. Bei, H.P.; Hung, P.M.; Yeung, H.L.; Wang, S.; Zhao, X. Bone-a-Petite: Engineering Exosomes towards Bone, Osteochondral, and Cartilage Repair. Small 2021, 17, 2101741. [Google Scholar] [CrossRef]
  12. 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. Nanomed. 2020, 15, 6917–6934. [Google Scholar] [CrossRef]
  13. Chen, J.; Li, P.; Zhang, T.; Xu, Z.; Huang, X.; Wang, R.; Du, L. Review on Strategies and Technologies for Exosome Isolation and Purification. Front. Bioeng. Biotechnol. 2021, 9, 811971. [Google Scholar] [CrossRef] [PubMed]
  14. Sidhom, K.; Obi, P.O.; Saleem, A. A Review of Exosomal Isolation Methods: Is Size Exclusion Chromatography the Best Option? Int. J. Mol. Sci. 2020, 21, 6466. [Google Scholar] [CrossRef]
  15. La Shu, S.; Allen, C.L.; Benjamin-Davalos, S.; Koroleva, M.; MacFarland, D.; Minderman, H.; Ernstoff, M.S. A Rapid Exosome Isolation Using Ultrafiltration and Size Exclusion Chromatography (REIUS) Method for Exosome Isolation from Melanoma Cell Lines. Methods Mol. Biol. 2021, 2265, 289–304. [Google Scholar] [CrossRef]
  16. Antimisiaris, S.G.; Mourtas, S.; Marazioti, A. Exosomes and Exosome-Inspired Vesicles for Targeted Drug Delivery. Pharmaceutics 2018, 10, 218. [Google Scholar] [CrossRef] [PubMed]
  17. Richardson, J.J.; Ejima, H. Surface Engineering of Extracellular Vesicles through Chemical and Biological Strategies. Chem. Mater. 2019, 31, 2191–2201. [Google Scholar] [CrossRef]
  18. Johnson, V.; Vasu, S.; Kumar, U.S.; Kumar, M. Surface-Engineered Extracellular Vesicles in Cancer Immunotherapy. Cancers 2023, 15, 2838. [Google Scholar] [CrossRef]
  19. Yu, F.; Zhao, X.; Wang, Q.; Fang, P.-H.; Liu, L.; Du, X.; Li, W.; He, D.; Zhang, T.; Bai, Y.; et al. Engineered Mesenchymal Stromal Cell Exosomes-Loaded Microneedles Improve Corneal Healing after Chemical Injury. ACS Nano 2024, 18, 20065–20082. [Google Scholar] [CrossRef]
  20. Zeng, H.; Guo, S.; Ren, X.; Wu, Z.; Liu, S.; Yao, X. Current Strategies for Exosome Cargo Loading and Targeting Delivery. Cells 2023, 12, 1416. [Google Scholar] [CrossRef]
  21. Han, Y.; Jones, T.W.; Dutta, S.; Zhu, Y.; Wang, X.; Narayanan, S.P.; Fagan, S.C.; Zhang, D. Overview and Update on Methods for Cargo Loading into Extracellular Vesicles. Processes 2021, 9, 356. [Google Scholar] [CrossRef]
  22. Bahadorani, M.; Nasiri, M.; Dellinger, K.; Aravamudhan, S.; Zadegan, R. Engineering Exosomes for Therapeutic Applications: Decoding Biogenesis, Content Modification, and Cargo Loading Strategies. Int. J. Nanomed. 2024, 19, 7137–7164. [Google Scholar] [CrossRef] [PubMed]
  23. Wang, J.-H.; Forterre, A.V.; Zhao, J.; Frimannsson, D.O.; Delcayre, A.; Antes, T.J.; Efron, B.; Jeffrey, S.S.; Pegram, M.D.; Matin, A.C. Anti-HER2 scFv-Directed Extracellular Vesicle-Mediated mRNA-Based Gene Delivery Inhibits Growth of HER2-Positive Human Breast Tumor Xenografts by Prodrug Activation. Mol. Cancer Ther. 2018, 17, 1133–1142. [Google Scholar] [CrossRef] [PubMed]
  24. Wu, Y.; Deng, W.; Ii, D.J.K. Exosomes: Improved methods to characterize their morphology, RNA content, and surface protein biomarkers. Analyst 2015, 140, 6631–6642. [Google Scholar] [CrossRef] [PubMed]
  25. Nireesha, G.; Divya, L.; Sowmya, C.; Venkateshan, N.N.B.M.; Lavakumar, V. Lyophilization/freeze drying—An review. Int. J. Nov. Trends Pharm. Sci. 2013, 3, 87–98. [Google Scholar]
  26. Bahr, M.M.; Amer, M.S.; Abo-El-Sooud, K.; Abdallah, A.N.; El-Tookhy, O.S. Preservation techniques of stem cells extracellular vesicles: A gate for manufacturing of clinical grade therapeutic extracellular vesicles and long-term clinical trials. Int. J. Vet. Sci. Med. 2020, 8, 1–8. [Google Scholar] [CrossRef]
  27. Ruprecht, N.A.; Kohlus, R. Thermal damages in spray drying: Particle size-dependent protein denaturation using phycocyanin as model substrate. Dry. Technol. 2023, 41, 2357–2370. [Google Scholar] [CrossRef]
  28. Zhang, J.; Cai, Z.; Feng, F.; Peng, Y.; Cui, Y.; Xu, Y. Age-different BMSCs-derived exosomes accelerate tendon-bone interface healing in rotator cuff tears model. Gene 2024, 895, 148002. [Google Scholar] [CrossRef]
  29. Clark, D.J.; Fondrie, W.E.; Yang, A.; Mao, L. Triple SILAC quantitative proteomic analysis reveals differential abundance of cell signaling proteins between normal and lung cancer-derived exosomes. J. Proteom. 2016, 133, 161–169. [Google Scholar] [CrossRef]
  30. Visconte, C.; Taiana, M.M.; Colombini, A.; De Luca, P.; Ragni, E.; de Girolamo, L. Donor Sites and Harvesting Techniques Affect miRNA Cargos of Extracellular Vesicles Released by Human Adipose-Derived Mesenchymal Stromal Cells. Int. J. Mol. Sci. 2024, 25, 6450. [Google Scholar] [CrossRef]
  31. Liu, W.; Li, L.; Rong, Y.; Qian, D.; Chen, J.; Zhou, Z.; Luo, Y.; Jiang, D.; Cheng, L.; Zhao, S.; et al. Hypoxic mesenchymal stem cell-derived exosomes promote bone fracture healing by the transfer of miR-126. Acta Biomater. 2020, 103, 196–212. [Google Scholar] [CrossRef]
  32. Zou, W.; Lai, M.; Zhang, Y.; Zheng, L.; Xing, Z.; Li, T.; Zou, Z.; Song, Q.; Zhao, X.; Xia, L.; et al. Exosome Release Is Regulated by mTORC1. Adv. Sci. 2019, 6, 1801313. [Google Scholar] [CrossRef] [PubMed]
  33. Zhang, Y.; Zhang, Y.; Chopp, M.; Zhang, Z.G.; Mahmood, A.; Xiong, Y. Mesenchymal Stem Cell-Derived Exosomes Improve Functional Recovery in Rats After Traumatic Brain Injury: A Dose-Response and Therapeutic Window Study. Neurorehabilit. Neural Repair 2020, 34, 616–626. [Google Scholar] [CrossRef] [PubMed]
  34. Willis, G.R.; Kourembanas, S.; Mitsialis, S.A. Toward Exosome-Based Therapeutics: Isolation, Heterogeneity, and Fit-for-Purpose Potency. Front. Cardiovasc. Med. 2017, 4, 63. [Google Scholar] [CrossRef]
  35. Zhao, J.; Ding, Y.; He, R.; Huang, K.; Liu, L.; Jiang, C.; Liu, Z.; Wang, Y.; Yan, X.; Cao, F.; et al. Dose-effect relationship and molecular mechanism by which BMSC-derived exosomes promote peripheral nerve regeneration after crush injury. Stem Cell Res. Ther. 2020, 11, 360. [Google Scholar] [CrossRef] [PubMed]
  36. Kemaloğlu, C.A.; Dursun, E.N.; Yay, A.H.; Gökdemir, N.S.M.; Mat, Ö.C.; Gönen, Z.B.D. The Optimal Effective Dose of Adipose-Derived Stem Cell Exosomes in Wound Healing. Ann. Plast. Surg. 2024, 93, 253–260. [Google Scholar] [CrossRef]
  37. Xie, X.; Song, Q.; Dai, C.; Cui, S.; Tang, R.; Li, S.; Chang, J.; Li, P.; Wang, J.; Li, J.; et al. Clinical safety and efficacy of allogenic human adipose mesenchymal stromal cells-derived exosomes in patients with mild to moderate Alzheimer’s disease: A phase I/II clinical trial. Gen. Psychiatry 2023, 36, e101143. [Google Scholar] [CrossRef]
  38. Lu, Y.; Huangfu, S.; Ma, C.; Ding, Y.; Zhang, Y.; Zhou, C.; Liao, L.; Li, M.; You, J.; Chen, Y.; et al. Exosomes derived from umbilical cord mesenchymal stem cells promote healing of complex perianal fistulas in rats. Stem Cell Res. Ther. 2024, 15, 414. [Google Scholar] [CrossRef]
  39. Xiao, Y.; Zou, D.; Liu, J.; Dai, F.; Zhao, A.; Yang, P. Dose-responsive effects of endothelial cell-sourced exosomes on vascular cell proliferation and phenotype transition. Biochim. Biophys. Acta (BBA) Gen. Subj. 2024, 1869, 130745. [Google Scholar] [CrossRef]
  40. Kang, Y.X.; Han, S.F.; Chai, W.; Zhang, Y.; Zhang, J.; Yu, H.Q.; Qian, P.P.; Wang, Z.; Liu, L.Y. Comparative study of the effect of different doses of human umbilical cord mesenchymal stem cells exosomes on intestinal barrier injury in severely burned rats. Zhonghua Yi Xue Za Zhi 2024, 104, 4153–4161. [Google Scholar] [CrossRef]
  41. Zhang, B.; Lai, R.C.; Sim, W.K.; Tan, T.T.; Lim, S.K. An Assessment of Administration Route on MSC-sEV Therapeutic Efficacy. Biomolecules 2024, 14, 622. [Google Scholar] [CrossRef]
  42. Gurung, S.; Perocheau, D.; Touramanidou, L.; Baruteau, J. The exosome journey: From biogenesis to uptake and intracellular signalling. Cell Commun. Signal. 2021, 19, 47. [Google Scholar] [CrossRef]
  43. Yang, Y.; Li, J.; Xia, Z.; Tang, B.; Li, Y. Mesenchymal stem cells-derived exosomes alleviate temporomandibular joint disc degeneration in temporomandibular joint disorder. Biochem. Biophys. Res. Commun. 2024, 726, 150278. [Google Scholar] [CrossRef] [PubMed]
  44. Yin, Z.; Qin, C.; Pan, S.; Shi, C.; Wu, G.; Feng, Y.; Zhang, J.; Yu, Z.; Liang, B.; Gui, J. Injectable hyperbranched PEG crosslinked hyaluronan hydrogel microparticles containing mir-99a-3p modified subcutaneous ADSCs-derived exosomes was beneficial for long-term treatment of osteoarthritis. Mater. Today Bio 2023, 23, 100813. [Google Scholar] [CrossRef] [PubMed]
  45. Liang, Y.; Duan, L.; Lu, J.; Xia, J. Engineering exosomes for targeted drug delivery. Theranostics 2021, 11, 3183–3195. [Google Scholar] [CrossRef] [PubMed]
  46. Kheyrolahzadeh, K.; Tohidkia, M.R.; Tarighatnia, A.; Shahabi, P.; Nader, N.D.; Aghanejad, A. Theranostic chimeric antigen receptor (CAR)-T cells: Insight into recent trends and challenges in solid tumors. Life Sci. 2023, 328, 121917. [Google Scholar] [CrossRef]
  47. Wang, J.; Zhu, X.; Jiang, H.; Ji, M.; Wu, Y.; Chen, J. Cancer cell-derived exosome based dual-targeted drug delivery system for non-small cell lung cancer therapy. Colloids Surf. B Biointerfaces 2024, 244, 114141. [Google Scholar] [CrossRef]
  48. Khayambashi, P.; Iyer, J.; Pillai, S.; Upadhyay, A.; Zhang, Y.; Tran, S.D. Hydrogel Encapsulation of Mesenchymal Stem Cells and Their Derived Exosomes for Tissue Engineering. Int. J. Mol. Sci. 2021, 22, 684. [Google Scholar] [CrossRef]
  49. Wang, S.; Xing, X.; Peng, W.; Huang, C.; Du, Y.; Yang, H.; Zhou, J. Fabrication of an exosome-loaded thermosensitive chitin-based hydrogel for dental pulp regeneration. J. Mater. Chem. B 2023, 11, 1580–1590. [Google Scholar] [CrossRef]
  50. Tanziela, T.; Shaikh, S.; Jiang, H.; Lu, Z.; Wang, X. Efficient encapsulation of biocompatible nanoparticles in exosomes for cancer theranostics. Nano Today 2020, 35, 100964. [Google Scholar] [CrossRef]
  51. Hua, S.; de Matos, M.B.C.; Metselaar, J.M.; Storm, G. Current Trends and Challenges in the Clinical Translation of Nanoparticulate Nanomedicines: Pathways for Translational Development and Commercialization. Front. Pharmacol. 2018, 9, 790. [Google Scholar] [CrossRef] [PubMed]
  52. Dong, C.; Lv, Y. Application of Collagen Scaffold in Tissue Engineering: Recent Advances and New Perspectives. Polymers 2016, 8, 42. [Google Scholar] [CrossRef] [PubMed]
  53. Phinney, D.G.; Di Giuseppe, M.; Njah, J.; Sala, E.; Shiva, S.; St Croix, C.M.; Stolz, D.B.; Watkins, S.C.; Di, Y.P.; Leikauf, G.D.; et al. Mesenchymal stem cells use extracellular vesicles to outsource mitophagy and shuttle microRNAs. Nat. Commun. 2015, 6, 8472. [Google Scholar] [CrossRef] [PubMed]
  54. Valadi, H.; Ekström, K.; Bossios, A.; Sjöstrand, M.; Lee, J.J.; Lötvall, J.O. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat. Cell Biol. 2007, 9, 654–659. [Google Scholar] [CrossRef] [PubMed]
  55. Hoang, D.H.; Nguyen, T.D.; Nguyen, H.-P.; Nguyen, X.-H.; Dang, V.D.; Dam, P.T.M.; Bui, H.T.H.; Trinh, M.Q.; Vu, D.M.; Hoang, N.T.M.; et al. Differential Wound Healing Capacity of Mesenchymal Stem Cell-Derived Exosomes Originated From Bone Marrow, Adipose Tissue and Umbilical Cord Under Serum- and Xeno-Free Condition. Front. Mol. Biosci. 2020, 7, 119. [Google Scholar] [CrossRef]
  56. Pomatto, M.; Gai, C.; Negro, F.; Cedrino, M.; Grange, C.; Ceccotti, E.; Togliatto, G.; Collino, F.; Tapparo, M.; Figliolini, F.; et al. Differential Therapeutic Effect of Extracellular Vesicles Derived by Bone Marrow and Adipose Mesenchymal Stem Cells on Wound Healing of Diabetic Ulcers and Correlation to Their Cargoes. Int. J. Mol. Sci. 2021, 22, 3851. [Google Scholar] [CrossRef]
  57. Soni, N.; Gupta, S.; Rawat, S.; Krishnakumar, V.; Mohanty, S.; Banerjee, A. MicroRNA-Enriched Exosomes from Different Sources of Mesenchymal Stem Cells Can Differentially Modulate Functions of Immune Cells and Neurogenesis. Biomedicines 2021, 10, 69. [Google Scholar] [CrossRef]
  58. Huang, S.; Li, Y.; Wu, P.; Xiao, Y.; Duan, N.; Quan, J.; Du, W. microRNA-148a-3p in extracellular vesicles derived from bone marrow mesenchymal stem cells suppresses SMURF1 to prevent osteonecrosis of femoral head. J. Cell. Mol. Med. 2020, 24, 11512–11523. [Google Scholar] [CrossRef]
  59. Behera, J.; Kumar, A.; Voor, M.J.; Tyagi, N. Exosomal lncRNA-H19 promotes osteogenesis and angiogenesis through mediating Angpt1/Tie2-NO signaling in CBS-heterozygous mice. Theranostics 2021, 11, 7715–7734. [Google Scholar] [CrossRef]
  60. Bian, S.; Zhang, L.; Duan, L.; Wang, X.; Min, Y.; Yu, H. Extracellular vesicles derived from human bone marrow mesenchymal stem cells promote angiogenesis in a rat myocardial infarction model. J. Mol. Med. 2014, 92, 387–397. [Google Scholar] [CrossRef]
  61. Liu, X.; Wu, C.; Zhang, Y.; Chen, S.; Ding, J.; Chen, Z.; Wu, K.; Wu, X.; Zhou, T.; Zeng, M.; et al. Hyaluronan-based hydrogel integrating exosomes for traumatic brain injury repair by promoting angiogenesis and neurogenesis. Carbohydr. Polym. 2023, 306, 120578. [Google Scholar] [CrossRef] [PubMed]
  62. Berebichez-Fridman, R.; Gómez-García, R.; Granados-Montiel, J.; Berebichez-Fastlicht, E.; Olivos-Meza, A.; Granados, J.; Velasquillo, C.; Ibarra, C. The Holy Grail of Orthopedic Surgery: Mesenchymal Stem Cells—Their Current Uses and Potential Applications. Stem Cells Int. 2017, 2017, 2638305. [Google Scholar] [CrossRef]
  63. Berebichez-Fridman, R.; Montero-Olvera, P.R. Sources and Clinical Applications of Mesenchymal Stem Cells: State-of-the-art review. Sultan Qaboos Univ. Med. J. 2018, 18, e264–e277. [Google Scholar] [CrossRef] [PubMed]
  64. Beane, O.S.; Fonseca, V.C.; Cooper, L.L.; Koren, G.; Darling, E.M. Impact of aging on the regenerative properties of bone marrow-, muscle-, and adipose-derived mesenchymal stem/stromal cells. PLoS ONE 2014, 9, e115963. [Google Scholar] [CrossRef] [PubMed]
  65. Gopi, I.K.; Rattan, S.I.S. Biphasic Dose–Response and Hormetic Effects of Stress Hormone Hydrocortisone on Telomerase-Immortalized Human Bone Marrow Stem Cells In Vitro. Dose Response 2019, 17, 1559325819889819. [Google Scholar] [CrossRef]
  66. Voloshin, N.; Tyurin-Kuzmin, P.; Karagyaur, M.; Akopyan, Z.; Kulebyakin, K. Practical Use of Immortalized Cells in Medicine: Current Advances and Future Perspectives. Int. J. Mol. Sci. 2023, 24, 12716. [Google Scholar] [CrossRef]
  67. Peng, L.; Jia, Z.; Yin, X.; Zhang, X.; Liu, Y.; Chen, P.; Ma, K.; Zhou, C. Comparative analysis of mesenchymal stem cells from bone marrow, cartilage, and adipose tissue. Stem Cells Dev. 2008, 17, 761–774. [Google Scholar] [CrossRef]
  68. Ding, D.C.; Shyu, W.C.; Lin, S.Z. Mesenchymal stem cells. Cell Transplant. 2011, 20, 5–14. [Google Scholar] [CrossRef]
  69. Wang, Y.; Li, Q.; Zhou, S.; Tan, P. Contents of exosomes derived from adipose tissue and their regulation on inflammation, tumors, and diabetes. Front. Endocrinol. 2024, 15, 1374715. [Google Scholar] [CrossRef]
  70. Li, C.; Wei, S.; Xu, Q.; Sun, Y.; Ning, X.; Wang, Z. Application of ADSCs and their Exosomes in Scar Prevention. Stem Cell Rev. Rep. 2022, 18, 952–967. [Google Scholar] [CrossRef]
  71. Qin, X.; He, J.; Wang, X.; Wang, J.; Yang, R.; Chen, X. The functions and clinical application potential of exosomes derived from mesenchymal stem cells on wound repair: A review of recent research advances. Front. Immunol. 2023, 14, 1256687. [Google Scholar] [CrossRef] [PubMed]
  72. Tangchitphisut, P.; Srikaew, N.; Numhom, S.; Tangprasittipap, A.; Woratanarat, P.; Wongsak, S.; Kijkunasathian, C.; Hongeng, S.; Murray, I.R.; Tawonsawatruk, T. Infrapatellar Fat Pad: An Alternative Source of Adipose-Derived Mesenchymal Stem Cells. Arthritis 2016, 2016, 4019873. [Google Scholar] [CrossRef] [PubMed]
  73. Schipper, B.M.; Marra, K.G.; Zhang, W.B.; Donnenberg, A.D.; Rubin, J.P. Regional anatomic and age effects on cell function of human adipose-derived stem cells. Ann. Plast. Surg. 2008, 60, 538–544. [Google Scholar] [CrossRef] [PubMed]
  74. Castaño, C.; Kalko, S.; Novials, A.; Párrizas, M. Obesity-associated exosomal miRNAs modulate glucose and lipid metabolism in mice. Proc. Natl. Acad. Sci. USA 2018, 115, 12158–12163. [Google Scholar] [CrossRef]
  75. Woo, C.H.; Kim, H.K.; Yang, S.; Park, J.H.; Jo, D.; Cho, Y.W.; Jung, G.Y.; Jung, Y.J.; Lee, K.S.; Yun, Y.E.; et al. Small extracellular vesicles from human adipose-derived stem cells attenuate cartilage degeneration. J. Extracell. Vesicles 2020, 9, 1735249. [Google Scholar] [CrossRef]
  76. Cui, X.; He, Z.; Liang, Z.; Chen, Z.; Wang, H.; Zhang, J. Exosomes From Adipose-derived Mesenchymal Stem Cells Protect the Myocardium Against Ischemia/Reperfusion Injury Through Wnt/β-Catenin Signaling Pathway. J. Cardiovasc. Pharmacol. 2017, 70, 225–231. [Google Scholar] [CrossRef]
  77. Liang, Y.; Tang, X.; Zhang, X.; Cao, C.; Yu, M.; Wan, M. Adipose Mesenchymal Stromal Cell-Derived Exosomes Carrying MiR-122-5p Antagonize the Inhibitory Effect of Dihydrotestosterone on Hair Follicles by Targeting the TGF-β1/SMAD3 Signaling Pathway. Int. J. Mol. Sci. 2023, 24, 5703. [Google Scholar] [CrossRef]
  78. Pendleton, C.; Li, Q.; Chesler, D.A.; Yuan, K.; Guerrero-Cazares, H.; Quinones-Hinojosa, A. Mesenchymal stem cells derived from adipose tissue vs bone marrow: In vitro comparison of their tropism towards gliomas. PLoS ONE 2013, 8, e58198. [Google Scholar] [CrossRef]
  79. Iacomi, D.-M.; Rosca, A.-M.; Tutuianu, R.; Neagu, T.P.; Pruna, V.; Simionescu, M.; Titorencu, I. Generation of an Immortalized Human Adipose-Derived Mesenchymal Stromal Cell Line Suitable for Wound Healing Therapy. Int. J. Mol. Sci. 2022, 23, 8925. [Google Scholar] [CrossRef]
  80. Wang, Z.-G.; He, Z.-Y.; Liang, S.; Yang, Q.; Cheng, P.; Chen, A.-M. Comprehensive proteomic analysis of exosomes derived from human bone marrow, adipose tissue, and umbilical cord mesenchymal stem cells. Stem Cell Res. Ther. 2020, 11, 511. [Google Scholar] [CrossRef]
  81. Xu, Y.; Tang, X.; Fang, A.; Yan, J.; Ocansey, D.K.W.; Zhang, X.; Mao, F. HucMSC-Ex carrying miR-203a-3p.2 ameliorates colitis through the suppression of caspase11/4-induced macrophage pyroptosis. Int. Immunopharmacol. 2022, 110, 108925. [Google Scholar] [CrossRef]
  82. Zhang, L.; Yuan, J.; Ocansey, D.K.W.; Lu, B.; Wan, A.; Chen, X.; Zhang, X.; Qiu, W.; Mao, F. Exosomes derived from human umbilical cord mesenchymal stem cells regulate lymphangiogenesis via the miR-302d-3p/VEGFR3/AKT axis to ameliorate inflammatory bowel disease. Int. Immunopharmacol. 2022, 110, 109066. [Google Scholar] [CrossRef] [PubMed]
  83. Bai, L.; Shao, H.; Wang, H.; Zhang, Z.; Su, C.; Dong, L.; Yu, B.; Chen, X.; Li, X.; Zhang, X. Effects of Mesenchymal Stem Cell-Derived Exosomes on Experimental Autoimmune Uveitis. Sci. Rep. 2017, 7, 4323. [Google Scholar] [CrossRef] [PubMed]
  84. Akhlaghpasand, M.; Tavanaei, R.; Hosseinpoor, M.; Yazdani, K.O.; Soleimani, A.; Zoshk, M.Y.; Soleimani, M.; Chamanara, M.; Ghorbani, M.; Deylami, M.; et al. Safety and potential effects of intrathecal injection of allogeneic human umbilical cord mesenchymal stem cell-derived exosomes in complete subacute spinal cord injury: A first-in-human, single-arm, open-label, phase I clinical trial. Stem Cell Res. Ther. 2024, 15, 264. [Google Scholar] [CrossRef] [PubMed]
  85. Li, W.; Si, Y.; Wang, Y.; Chen, J.; Huo, X.; Xu, P.; Jiang, B.; Li, Z.; Shang, K.; Luo, Q.; et al. hUCMSC-derived exosomes protect against GVHD-induced endoplasmic reticulum stress in CD4+ T cells by targeting the miR-16-5p/ATF6/CHOP axis. Int. Immunopharmacol. 2024, 135, 112315. [Google Scholar] [CrossRef] [PubMed]
  86. Iaffaldano, L.; Nardelli, C.; D’Alessio, F.; D’Argenio, V.; Nunziato, M.; Mauriello, L.; Procaccini, C.; Maruotti, G.M.; Martinelli, P.; Matarese, G.; et al. Altered Bioenergetic Profile in Umbilical Cord and Amniotic Mesenchymal Stem Cells from Newborns of Obese Women. Stem Cells Dev. 2018, 27, 199–206. [Google Scholar] [CrossRef]
  87. Kim, J.; Piao, Y.; Pak, Y.K.; Chung, D.; Han, Y.M.; Hong, J.S.; Jun, E.J.; Shim, J.-Y.; Choi, J.; Kim, C.J. Umbilical cord mesenchymal stromal cells affected by gestational diabetes mellitus display premature aging and mitochondrial dysfunction. Stem Cells Dev. 2015, 24, 575–586. [Google Scholar] [CrossRef]
  88. Chen, Q.; Jin, M.; Wang, S.; Wang, K.; Chen, L.; Zhu, X.; Zhang, Y.; Wang, Y.; Li, Y.; Li, S.; et al. Establishing an hTERT-driven immortalized umbilical cord-derived mesenchymal stem cell line and its therapeutic application in mice with liver failure. J. Tissue Eng. 2023, 14, 20417314231200328. [Google Scholar] [CrossRef]
  89. Zhou, H.-L.; Zhang, X.-J.; Zhang, M.-Y.; Yan, Z.-J.; Xu, Z.-M.; Xu, R.-X. Transplantation of Human Amniotic Mesenchymal Stem Cells Promotes Functional Recovery in a Rat Model of Traumatic Spinal Cord Injury. Neurochem. Res. 2016, 41, 2708–2718. [Google Scholar] [CrossRef]
  90. Parolini, O.; Alviano, F.; Bagnara, G.P.; Bilic, G.; Bühring, H.-J.; Evangelista, M.; Hennerbichler, S.; Liu, B.; Magatti, M.; Mao, N.; et al. Concise review: Isolation and characterization of cells from human term placenta: Outcome of the first international Workshop on Placenta Derived Stem Cells. Stem Cells 2008, 26, 300–311. [Google Scholar] [CrossRef]
  91. Banas, R.A.; Trumpower, C.; Bentlejewski, C.; Marshall, V.; Sing, G.; Zeevi, A. Immunogenicity and immunomodulatory effects of amnion-derived multipotent progenitor cells. Hum. Immunol. 2008, 69, 321–328. [Google Scholar] [CrossRef] [PubMed]
  92. Ohara, M.; Ohnishi, S.; Hosono, H.; Yamamoto, K.; Yuyama, K.; Nakamura, H.; Fu, Q.; Maehara, O.; Suda, G.; Sakamoto, N. Extracellular Vesicles from Amnion-Derived Mesenchymal Stem Cells Ameliorate Hepatic Inflammation and Fibrosis in Rats. Stem Cells Int. 2018, 2018, 3212643. [Google Scholar] [CrossRef] [PubMed]
  93. Topoluk, N.; Steckbeck, K.; Siatkowski, S.; Burnikel, B.; Tokish, J.; Mercuri, J. Amniotic mesenchymal stem cells mitigate osteoarthritis progression in a synovial macrophage-mediated in vitro explant coculture model. J. Tissue Eng. Regen. Med. 2018, 12, 1097–1110. [Google Scholar] [CrossRef] [PubMed]
  94. Noh, C.H.; Park, S.; Seong, H.-R.; Lee, A.-Y.; Tsolmon, K.-E.; Geum, D.; Hong, S.-C.; Kim, T.M.; Choi, E.-K.; Kim, Y.-B. An Exosome-Rich Conditioned Medium from Human Amniotic Membrane Stem Cells Facilitates Wound Healing via Increased Reepithelization, Collagen Synthesis, and Angiogenesis. Cells 2023, 12, 2698. [Google Scholar] [CrossRef] [PubMed]
  95. Zhou, Y.; He, L.-N.; Wang, L.-N.; Chen, K.-Y.; Qian, S.-D.; Li, X.-H.; Zang, J.; Wang, D.-M.; Yu, X.-F.; Gao, J. Human amniotic mesenchymal stromal cell-derived exosomes promote neuronal function by inhibiting excessive apoptosis in a hypoxia/ischemia-induced cerebral palsy model: A preclinical study. Biomed. Pharmacother. 2024, 173, 116321. [Google Scholar] [CrossRef]
  96. Zhou, H.; Wang, J.; Zhao, P.; Le, D.; Cai, S.; Mao, G. Intravenous administration exosomes derived from human amniotic mesenchymal stem cells improves neurological recovery after acute traumatic spinal cord injury in rats. Iran. J. Basic Med. Sci. 2024, 27, 1284–1292. [Google Scholar] [CrossRef]
  97. Xiao, S.; Xiao, C.; Miao, Y.; Wang, J.; Chen, R.; Fan, Z.; Hu, Z. Human acellular amniotic membrane incorporating exosomes from adipose-derived mesenchymal stem cells promotes diabetic wound healing. Stem Cell Res. Ther. 2021, 12, 255. [Google Scholar] [CrossRef]
  98. Janockova, J.; Matejova, J.; Moravek, M.; Homolova, L.; Slovinska, L.; Nagyova, A.; Rak, D.; Sedlak, M.; Harvanova, D.; Spakova, T.; et al. Small Extracellular Vesicles Derived from Human Chorionic MSCs as Modern Perspective towards Cell-Free Therapy. Int. J. Mol. Sci. 2021, 22, 13581. [Google Scholar] [CrossRef]
  99. Li, Y.; Wang, C.; Xi, H.; Li, W.; Liu, Y.; Feng, S.; Chu, Y.; Wang, Y. Chorionic villus-derived mesenchymal stem cells induce E3 ligase TRIM72 expression and regulate cell behaviors through ubiquitination of p53 in trophoblasts. FASEB J. 2021, 35, e22005. [Google Scholar] [CrossRef]
  100. Salomon, C.; Ryan, J.; Sobrevia, L.; Kobayashi, M.; Ashman, K.; Mitchell, M.; Rice, G.E. Exosomal signaling during hypoxia mediates microvascular endothelial cell migration and vasculogenesis. PLoS ONE 2013, 8, e68451. [Google Scholar] [CrossRef]
  101. Bian, X.; Li, B.; Yang, J.; Ma, K.; Sun, M.; Zhang, C.; Fu, X. Regenerative and protective effects of dMSC-sEVs on high-glucose-induced senescent fibroblasts by suppressing RAGE pathway and activating Smad pathway. Stem Cell Res. Ther. 2020, 11, 166. [Google Scholar] [CrossRef] [PubMed]
  102. Egusa, H.; Sonoyama, W.; Nishimura, M.; Atsuta, I.; Akiyama, K. Stem cells in dentistry—Part I: Stem cell sources. J. Prosthodont. Res. 2012, 56, 151–165. [Google Scholar] [CrossRef] [PubMed]
  103. Zhang, W.; Walboomers, X.F.; Shi, S.; Fan, M.; Jansen, J.A. Multilineage differentiation potential of stem cells derived from human dental pulp after cryopreservation. Tissue Eng. 2006, 12, 2813–2823. [Google Scholar] [CrossRef] [PubMed]
  104. Li, Y.; Chen, Y.; Liu, B.; Nie, Q.; Li, L.; Duan, X.; Wu, L.; Chen, G. Deciphering the Heterogeneity Landscape of Mesenchymal Stem/Stromal Cell-Derived Extracellular Vesicles for Precise Selection in Translational Medicine. Adv. Healthc. Mater. 2023, 12, e2202453. [Google Scholar] [CrossRef] [PubMed]
  105. Mas-Bargues, C.; Sanz-Ros, J.; Romero-García, N.; Huete-Acevedo, J.; Dromant, M.; Borrás, C. Small extracellular vesicles from senescent stem cells trigger adaptive mechanisms in young stem cells by increasing antioxidant enzyme expression. Redox Biol. 2023, 62, 102668. [Google Scholar] [CrossRef]
  106. Huang, C.-C.; Narayanan, R.; Alapati, S.; Ravindran, S. Exosomes as biomimetic tools for stem cell differentiation: Applications in dental pulp tissue regeneration. Biomaterials 2016, 111, 103–115. [Google Scholar] [CrossRef]
  107. Swanson, W.B.; Gong, T.; Zhang, Z.; Eberle, M.; Niemann, D.; Dong, R.; Rambhia, K.J.; Ma, P.X. Controlled release of odontogenic exosomes from a biodegradable vehicle mediates dentinogenesis as a novel biomimetic pulp capping therapy. J. Control Release 2020, 324, 679–694. [Google Scholar] [CrossRef]
  108. Ji, L.; Bao, L.; Gu, Z.; Zhou, Q.; Liang, Y.; Zheng, Y.; Xu, Y.; Zhang, X.; Feng, X. Comparison of immunomodulatory properties of exosomes derived from bone marrow mesenchymal stem cells and dental pulp stem cells. Immunol. Res. 2019, 67, 432–442. [Google Scholar] [CrossRef]
  109. Venugopal, C.; Shobha, K.; Rai, K.S.; Pinnelli, V.B.; Kutty, B.M.; Dhanushkodi, A. Neuroprotection by Human Dental Pulp Mesenchymal Stem Cells: From Billions to Nano. Curr. Gene Ther. 2018, 18, 307–323. [Google Scholar] [CrossRef]
  110. Shen, Z.; Kuang, S.; Zhang, Y.; Yang, M.; Qin, W.; Shi, X.; Lin, Z. Chitosan hydrogel incorporated with dental pulp stem cell-derived exosomes alleviates periodontitis in mice via a macrophage-dependent mechanism. Bioact. Mater. 2020, 5, 1113–1126. [Google Scholar] [CrossRef]
  111. Zheng, J.; Kong, Y.; Hu, X.; Li, Z.; Li, Y.; Zhong, Y.; Wei, X.; Ling, J. MicroRNA-enriched small extracellular vesicles possess odonto-immunomodulatory properties for modulating the immune response of macrophages and promoting odontogenesis. Stem Cell Res. Ther. 2020, 11, 517. [Google Scholar] [CrossRef] [PubMed]
  112. Miao, Y.; Liang, X.; Chen, J.; Liu, H.; He, Z.; Qin, Y.; Liu, A.; Zhang, R. Transfer of miR-877-3p via extracellular vesicles derived from dental pulp stem cells attenuates neuronal apoptosis and facilitates early neurological functional recovery after cerebral ischemia–reperfusion injury through the Bclaf1/P53 signaling pathway. Pharmacol. Res. 2024, 206, 107266. [Google Scholar] [CrossRef] [PubMed]
  113. Liang, X.; Miao, Y.; Tong, X.; Chen, J.; Liu, H.; He, Z.; Liu, A.; Hu, Z. Dental pulp mesenchymal stem cell-derived exosomes inhibit neuroinflammation and microglial pyroptosis in subarachnoid hemorrhage via the miRNA-197-3p/FOXO3 axis. J. Nanobiotechnol. 2024, 22, 426. [Google Scholar] [CrossRef] [PubMed]
  114. Chai, Y.; Liu, Y.; Liu, Z.; Wei, W.; Dong, Y.; Yang, C.; Chen, M. Dental Pulp Stem Cell-Derived Exosomes Promote Sciatic Nerve Regeneration via Optimizing Schwann Cell Function. Cell. Reprogram. 2024, 26, 67–78. [Google Scholar] [CrossRef] [PubMed]
  115. Lee, A.; Choi, J.; Shi, S.; He, P.; Zhang, Q.; Le, A. DPSC-Derived Extracellular Vesicles Promote Rat Jawbone Regeneration. J. Dent. Res. 2023, 102, 313–321. [Google Scholar] [CrossRef]
  116. Zhou, Z.; Zheng, J.; Lin, D.; Xu, R.; Chen, Y.; Hu, X. Exosomes derived from dental pulp stem cells accelerate cutaneous wound healing by enhancing angiogenesis via the Cdc42/p38 MAPK pathway. Int. J. Mol. Med. 2022, 50, 143. [Google Scholar] [CrossRef]
  117. Stanko, P.; Altanerova, U.; Jakubechova, J.; Repiska, V.; Altaner, C. Dental Mesenchymal Stem/Stromal Cells and Their Exosomes. Stem Cells Int. 2018, 2018, 8973613. [Google Scholar] [CrossRef]
  118. Ikbale, E.-A.; Goorha, S.; Reiter, L.T.; Miranda-Carboni, G.A. Effects of hTERT immortalization on osteogenic and adipogenic differentiation of dental pulp stem cells. Data Brief 2016, 6, 696–699. [Google Scholar] [CrossRef]
  119. Victor, A.K.; Reiter, L.T. Dental pulp stem cells for the study of neurogenetic disorders. Hum. Mol. Genet. 2017, 26, R166–R171. [Google Scholar] [CrossRef]
  120. Heydari, Z.; Peshkova, M.; Gonen, Z.B.; Coretchi, I.; Eken, A.; Yay, A.H.; Dogan, M.E.; Gokce, N.; Akalin, H.; Kosheleva, N.; et al. EVs vs. EVs: MSCs and Tregs as a source of invisible possibilities. J. Mol. Med. 2023, 101, 51–63. [Google Scholar] [CrossRef]
  121. Ying, W.; Riopel, M.; Bandyopadhyay, G.; Dong, Y.; Birmingham, A.; Seo, J.B.; Ofrecio, J.M.; Wollam, J.; Hernandez-Carretero, A.; Fu, W.; et al. Adipose Tissue Macrophage-Derived Exosomal miRNAs Can Modulate In Vivo and In Vitro Insulin Sensitivity. Cell 2017, 171, 372–384.e12. [Google Scholar] [CrossRef]
  122. Wang, Y.; Lin, Q.; Zhang, H.; Wang, S.; Cui, J.; Hu, Y.; Liu, J.; Li, M.; Zhang, K.; Zhou, F.; et al. M2 macrophage-derived exosomes promote diabetic fracture healing by acting as an immunomodulator. Bioact. Mater. 2023, 28, 273–283. [Google Scholar] [CrossRef] [PubMed]
  123. Kim, H.; Wang, S.Y.; Kwak, G.; Yang, Y.; Kwon, I.C.; Kim, S.H. Exosome-Guided Phenotypic Switch of M1 to M2 Macrophages for Cutaneous Wound Healing. Adv. Sci. 2019, 6, 1900513. [Google Scholar] [CrossRef] [PubMed]
  124. Cai, Z.; Shu, L.; Wang, C.; Xie, X.; Liu, X. M2 macrophage-derived exosomes promote tendon-to-bone healing by alleviating cellular senescence in aged rats. Arthrosc. J. Arthrosc. Relat. Surg. 2024; in press. [Google Scholar] [CrossRef] [PubMed]
  125. Huang, J.-H.; He, H.; Chen, Y.-N.; Liu, Z.; Romani, M.D.; Xu, Z.-Y.; Xu, Y.; Lin, F.-Y. Exosomes derived from M2 Macrophages Improve Angiogenesis and Functional Recovery after Spinal Cord Injury through HIF-1α/VEGF Axis. Brain Sci. 2022, 12, 1322. [Google Scholar] [CrossRef] [PubMed]
  126. Yin, J.; Pei, Z.; Wu, C.; Liu, J.; Huang, J.; Xia, R.; Xiang, D. M2 Macrophage-Derived Exosomal circ_0088494 Inhibits Ferroptosis via Promoting H3K4me1 Modification of STEAP3 in Cutaneous Squamous Cell Carcinoma. Mol. Carcinog. 2024. [Google Scholar] [CrossRef] [PubMed]
  127. Zhou, Y.-K.; Han, C.-S.; Zhu, Z.-L.; Chen, P.; Wang, Y.-M.; Lin, S.; Chen, L.-J.; Zhuang, Z.-M.; Zhou, Y.-H.; Yang, R.-L. M2 exosomes modified by hydrogen sulfide promoted bone regeneration by moesin mediated endocytosis. Bioact. Mater. 2024, 31, 192–205. [Google Scholar] [CrossRef] [PubMed]
  128. Hou, C.; Zhang, Y.; Lv, Z.; Luan, Y.; Li, J.; Meng, C.; Liu, K.; Luo, X.; Chen, L.; Liu, F. Macrophage exosomes modified by miR-365-2-5p promoted osteoblast osteogenic differentiation by targeting OLFML1. Regen. Biomater. 2024, 11, rbae018. [Google Scholar] [CrossRef]
  129. Ma, Y.-S.; Wu, T.-M.; Ling, C.-C.; Yu, F.; Zhang, J.; Cao, P.-S.; Gu, L.-P.; Wang, H.-M.; Xu, H.; Li, L.; et al. M2 macrophage-derived exosomal microRNA-155-5p promotes the immune escape of colon cancer by downregulating ZC3H12B. Mol. Ther. Oncolytics 2021, 20, 484–498. [Google Scholar] [CrossRef]
  130. Bahmani, L.; Ullah, M. Different Sourced Extracellular Vesicles and Their Potential Applications in Clinical Treatments. Cells 2022, 11, 1989. [Google Scholar] [CrossRef]
  131. Tung, S.L.; Fanelli, G.; Matthews, R.I.; Bazoer, J.; Letizia, M.; Vizcay-Barrena, G.; Faruqu, F.N.; Philippeos, C.; Hannen, R.; Al-Jamal, K.T.; et al. Regulatory T Cell Extracellular Vesicles Modify T-Effector Cell Cytokine Production and Protect Against Human Skin Allograft Damage. Front. Cell Dev. Biol. 2020, 8, 317. [Google Scholar] [CrossRef] [PubMed]
  132. Aiello, S.; Rocchetta, F.; Longaretti, L.; Faravelli, S.; Todeschini, M.; Cassis, L.; Pezzuto, F.; Tomasoni, S.; Azzollini, N.; Mister, M.; et al. Extracellular vesicles derived from T regulatory cells suppress T cell proliferation and prolong allograft survival. Sci. Rep. 2017, 7, 11518. [Google Scholar] [CrossRef] [PubMed]
  133. Chen, L.; Huang, H.; Zhang, W.; Ding, F.; Fan, Z.; Zeng, Z. Exosomes Derived From T Regulatory Cells Suppress CD8+ Cytotoxic T Lymphocyte Proliferation and Prolong Liver Allograft Survival. Med. Sci. Monit. 2019, 25, 4877–4884. [Google Scholar] [CrossRef] [PubMed]
  134. Yang, C.; Yuan, F.; Shao, W.; Yao, L.; Jin, S.; Han, F. Protective role of exosomes derived from regulatory T cells against inflammation and apoptosis of BV-2 microglia under oxygen-glucose deprivation/reperfusion challenge. Genet. Mol. Biol. 2022, 45, e20220119. [Google Scholar] [CrossRef]
  135. Yang, F.; Cai, D.; Kong, R.; Bi, Y.; Zhang, Y.; Lei, Y.; Peng, Y.; Li, X.; Xiao, Y.; Zhou, Z.; et al. Exosomes derived from cord blood Treg cells promote diabetic wound healing by targeting monocytes. Biochem. Pharmacol. 2024, 226, 116413. [Google Scholar] [CrossRef]
  136. Hu, H.; Wu, J.; Cao, C.; Ma, L. Exosomes derived from regulatory T cells ameliorate acute myocardial infarction by promoting macrophage M2 polarization. IUBMB Life 2020, 72, 2409–2419. [Google Scholar] [CrossRef]
  137. Webb, R.L.; Kaiser, E.E.; Scoville, S.L.; Thompson, T.A.; Fatima, S.; Pandya, C.; Sriram, K.; Swetenburg, R.L.; Vaibhav, K.; Arbab, A.S.; et al. Human Neural Stem Cell Extracellular Vesicles Improve Tissue and Functional Recovery in the Murine Thromboembolic Stroke Model. Transl. Stroke Res. 2018, 9, 530–539. [Google Scholar] [CrossRef]
  138. Xin, H.; Li, Y.; Cui, Y.; Yang, J.J.; Zhang, Z.G.; Chopp, M. Systemic administration of exosomes released from mesenchymal stromal cells promote functional recovery and neurovascular plasticity after stroke in rats. J. Cereb. Blood Flow Metab. 2013, 33, 1711–1715. [Google Scholar] [CrossRef]
  139. Rong, Y.; Liu, W.; Wang, J.; Fan, J.; Luo, Y.; Li, L.; Kong, F.; Chen, J.; Tang, P.; Cai, W. Neural stem cell-derived small extracellular vesicles attenuate apoptosis and neuroinflammation after traumatic spinal cord injury by activating autophagy. Cell Death Dis. 2019, 10, 340. [Google Scholar] [CrossRef]
  140. Liu, Y.; Huber, C.C.; Wang, H. Disrupted blood-brain barrier in 5xFAD mouse model of Alzheimer’s disease can be mimicked and repaired in vitro with neural stem cell-derived exosomes. Biochem. Biophys. Res. Commun. 2020, 525, 192–196. [Google Scholar] [CrossRef]
  141. Li, J.; Gao, H.; Xiong, Y.; Wang, L.; Zhang, H.; He, F.; Zhao, J.; Liu, S.; Gao, L.; Guo, Y.; et al. Enhancing Cutaneous Wound Healing Based on Human Induced Neural Stem Cell-derived Exosomes. Int. J. Nanomed. 2022, 17, 5991–6006. [Google Scholar] [CrossRef] [PubMed]
  142. Zhang, Y.; Kim, M.S.; Jia, B.; Yan, J.; Zuniga-Hertz, J.P.; Han, C.; Cai, D. Hypothalamic stem cells control ageing speed partly through exosomal miRNAs. Nature 2017, 548, 52–57. [Google Scholar] [CrossRef] [PubMed]
  143. Zhong, L.; Wang, J.; Wang, P.; Liu, X.; Liu, P.; Cheng, X.; Cao, L.; Wu, H.; Chen, J.; Zhou, L. Neural stem cell-derived exosomes and regeneration: Cell-free therapeutic strategies for traumatic brain injury. Stem Cell Res. Ther. 2023, 14, 198. [Google Scholar] [CrossRef] [PubMed]
  144. Clément, V.; Roy, V.; Paré, B.; Goulet, C.R.; Deschênes, L.T.; Berthod, F.; Bolduc, S.; Gros-Louis, F. Tridimensional cell culture of dermal fibroblasts promotes exosome-mediated secretion of extracellular matrix proteins. Sci. Rep. 2022, 12, 19786. [Google Scholar] [CrossRef] [PubMed]
  145. Li, B.; Wang, J.H.-C. Fibroblasts and myofibroblasts in wound healing: Force generation and measurement. J. Tissue Viability 2011, 20, 108–120. [Google Scholar] [CrossRef]
  146. Gangadaran, P.; Oh, E.J.; Rajendran, R.L.; Kim, H.M.; Oh, J.M.; Kwak, S.; Hong, C.M.; Choi, K.Y.; Chung, H.Y.; Ahn, B.-C. Identification of Angiogenic Cargoes in Human Fibroblasts-Derived Extracellular Vesicles and Induction of Wound Healing. Pharmaceuticals 2022, 15, 702. [Google Scholar] [CrossRef]
  147. Zhou, X.; Lv, Y.; Xie, H.; Li, Y.; Liu, C.; Zheng, M.; Wu, R.; Zhou, S.; Gu, X.; Li, J.; et al. RNA sequencing of exosomes secreted by fibroblast and Schwann cells elucidates mechanisms underlying peripheral nerve regeneration. Neural Regen. Res. 2024, 19, 1812–1821. [Google Scholar] [CrossRef]
  148. Zhao, Y.; Liang, Y.; Xu, Z.; Liu, J.; Liu, X.; Ma, J.; Sun, C.; Yang, Y. Exosomal miR-673-5p from fibroblasts promotes Schwann cell-mediated peripheral neuron myelination by targeting the TSC2/mTORC1/SREBP2 axis. J. Biol. Chem. 2022, 298, 101718. [Google Scholar] [CrossRef]
  149. Zhao, Y.; Liu, J.; Liu, S.; Yang, P.; Liang, Y.; Ma, J.; Mao, S.; Sun, C.; Yang, Y. Fibroblast exosomal TFAP2C induced by chitosan oligosaccharides promotes peripheral axon regeneration via the miR-132-5p/CAMKK1 axis. Bioact. Mater. 2023, 26, 249–263. [Google Scholar] [CrossRef]
  150. Park, A.Y.; Lee, J.; Jang, Y.; Kim, Y.-J.; Kim, S.-Y.; Kim, B.J.; Yoo, K.H. Exosomes derived from human dermal fibroblasts protect against UVB-induced skin photoaging. Int. J. Mol. Med. 2023, 52, 120. [Google Scholar] [CrossRef]
  151. Hu, S.; Li, Z.; Cores, J.; Huang, K.; Su, T.; Dinh, P.-U.; Cheng, K. Needle-Free Injection of Exosomes Derived from Human Dermal Fibroblast Spheroids Ameliorates Skin Photoaging. ACS Nano 2019, 13, 11273–11282. [Google Scholar] [CrossRef] [PubMed]
  152. Na Jang, Y.; Lee, J.M.; Park, A.Y.; Kim, Y.J.; Kim, S.Y.; Seok, J.; Yoo, K.H.; Kim, B.J. Exosomes derived from human dermal fibroblasts (HDFn-Ex) alleviate DNCB-induced atopic dermatitis (AD) via PPARα. Exp. Dermatol. 2024, 33, e14970. [Google Scholar] [CrossRef] [PubMed]
  153. Xiang, K.; Chen, J.; Guo, J.; Li, G.; Kang, Y.; Wang, C.; Jiang, T.; Zhang, M.; Jiang, G.; Yuan, M.; et al. Multifunctional ADM hydrogel containing endothelial cell-exosomes for diabetic wound healing. Mater. Today Bio 2023, 23, 100863. [Google Scholar] [CrossRef] [PubMed]
  154. Zeng, T.; Wang, X.; Wang, W.; Feng, Q.; Lao, G.; Liang, Y.; Wang, C.; Zhou, J.; Chen, Y.; Liu, J.; et al. Endothelial cell-derived small extracellular vesicles suppress cutaneous wound healing through regulating fibroblasts autophagy. Clin. Sci. 2019, 133, CS20190008. [Google Scholar] [CrossRef] [PubMed]
  155. Liu, J.; Dong, J.; Pei, X. Apoptotic Extracellular Vesicles Derived from Human Umbilical Vein Endothelial Cells Promote Skin Repair by Enhancing Angiogenesis: From Death to Regeneration. Int. J. Nanomed. 2024, 19, 415–428. [Google Scholar] [CrossRef]
  156. Qi, L.; Lu, Y.; Wang, Z.; Zhang, G. microRNA-106b derived from endothelial cell–secreted extracellular vesicles prevents skin wound healing by inhibiting JMJD3 and RIPK3. J. Cell. Mol. Med. 2021, 25, 4551–4561. [Google Scholar] [CrossRef]
  157. Guo, L.; Chen, Y.; Feng, X.; Sun, D.; Sun, J.; Mou, S.; Zhao, K.; An, R. Oxidative stress-induced endothelial cells-derived exosomes accelerate skin flap survival through Lnc NEAT1-mediated promotion of endothelial progenitor cell function. Stem Cell Res. Ther. 2022, 13, 325. [Google Scholar] [CrossRef]
  158. Maiullari, F.; Chirivì, M.; Costantini, M.; Ferretti, A.M.; Recchia, S.; Maiullari, S.; Milan, M.; Presutti, D.; Pace, V.; Raspa, M.; et al. In vivo organized neovascularization induced by 3D bioprinted endothelial-derived extracellular vesicles. Biofabrication 2021, 13, 035014. [Google Scholar] [CrossRef]
  159. Yu, Y.; Zhou, H.; Xiong, Y.; Liu, J. Exosomal miR-199a-5p derived from endothelial cells attenuates apoptosis and inflammation in neural cells by inhibiting endoplasmic reticulum stress. Brain Res. 2020, 1726, 146515. [Google Scholar] [CrossRef]
  160. Xiao, B.; Chai, Y.; Lv, S.; Ye, M.; Wu, M.; Xie, L.; Fan, Y.; Zhu, X.; Gao, Z. Endothelial cell-derived exosomes protect SH-SY5Y nerve cells against ischemia/reperfusion injury. Int. J. Mol. Med. 2017, 40, 1201–1209. [Google Scholar] [CrossRef]
  161. Chen, Y.; Wu, Y.; Guo, L.; Yuan, S.; Sun, J.; Zhao, K.; Wang, J.; An, R. Exosomal Lnc NEAT1 from endothelial cells promote bone regeneration by regulating macrophage polarization via DDX3X/NLRP3 axis. J. Nanobiotechnol. 2023, 21, 98. [Google Scholar] [CrossRef] [PubMed]
  162. Li, L.; Mu, J.; Zhang, Y.; Zhang, C.; Ma, T.; Chen, L.; Huang, T.; Wu, J.; Cao, J.; Feng, S.; et al. Stimulation by Exosomes from Hypoxia Preconditioned Human Umbilical Vein Endothelial Cells Facilitates Mesenchymal Stem Cells Angiogenic Function for Spinal Cord Repair. ACS Nano 2022, 16, 10811–10823. [Google Scholar] [CrossRef] [PubMed]
  163. Lopez-Verrilli, M.A.; Picou, F.; Court, F.A. Schwann cell-derived exosomes enhance axonal regeneration in the peripheral nervous system. Glia 2013, 61, 1795–1806. [Google Scholar] [CrossRef]
  164. López-Leal, R.; Díaz-Viraqué, F.; Catalán, R.J.; Saquel, C.; Enright, A.; Iraola, G.; Court, F.A. Schwann cell reprogramming into repair cells increases miRNA-21 expression in exosomes promoting axonal growth. J. Cell Sci. 2020, 133, jcs239004. [Google Scholar] [CrossRef] [PubMed]
  165. Wu, X.; Wang, L.; Cong, M.; Shen, M.; He, Q.; Ding, F.; Shi, H. Extracellular vesicles from skin precursor-derived Schwann cells promote axonal outgrowth and regeneration of motoneurons via Akt/mTOR/p70S6K pathway. Ann. Transl. Med. 2020, 8, 1640. [Google Scholar] [CrossRef] [PubMed]
  166. Cong, M.; Li, J.; Wang, L.; Liu, C.; Zheng, M.; Zhou, Q.; Du, M.; Ye, X.; Feng, M.; Ye, Y.; et al. MircoRNA-25-3p in skin precursor cell-induced Schwann cell-derived extracellular vesicles promotes axon regeneration by targeting Tgif1. Exp. Neurol. 2024, 376, 114750. [Google Scholar] [CrossRef]
  167. Wang, W.; Shen, D.; Zhang, L.; Ji, Y.; Xu, L.; Chen, Z.; Shen, Y.; Gong, L.; Zhang, Q.; Shen, M.; et al. SKP-SC-EVs Mitigate Denervated Muscle Atrophy by Inhibiting Oxidative Stress and Inflammation and Improving Microcirculation. Antioxidants 2021, 11, 66. [Google Scholar] [CrossRef]
  168. Pan, D.; Zhu, S.; Zhang, W.; Wei, Z.; Yang, F.; Guo, Z.; Ning, G.; Feng, S. Autophagy induced by Schwann cell-derived exosomes promotes recovery after spinal cord injury in rats. Biotechnol. Lett. 2022, 44, 129–142. [Google Scholar] [CrossRef]
  169. Xu, B.; Zhou, Z.; Fang, J.; Wang, J.; Tao, K.; Liu, J.; Liu, S. Exosomes derived from schwann cells alleviate mitochondrial dysfunction and necroptosis after spinal cord injury via AMPK signaling pathway-mediated mitophagy. Free Radic. Biol. Med. 2023, 208, 319–333. [Google Scholar] [CrossRef]
  170. Hao, Z.; Ren, L.; Zhang, Z.; Yang, Z.; Wu, S.; Liu, G.; Cheng, B.; Wu, J.; Xia, J. A multifunctional neuromodulation platform utilizing Schwann cell-derived exosomes orchestrates bone microenvironment via immunomodulation, angiogenesis and osteogenesis. Bioact. Mater. 2023, 23, 206–222. [Google Scholar] [CrossRef]
  171. Li, Z.; Liang, Y.; Pan, K.; Li, H.; Yu, M.; Guo, W.; Chen, G.; Tian, W. Schwann cells secrete extracellular vesicles to promote and maintain the proliferation and multipotency of hDPCs. Cell Prolif. 2017, 50, e12353. [Google Scholar] [CrossRef] [PubMed]
  172. Zhou, M.; Hu, M.; He, S.; Li, B.; Liu, C.; Min, J.; Hong, L. Effects of RSC96 Schwann Cell-Derived Exosomes on Proliferation, Senescence, and Apoptosis of Dorsal Root Ganglion Cells In Vitro. Med. Sci. Monit. 2018, 24, 7841–7849. [Google Scholar] [CrossRef] [PubMed]
  173. Jiang, H.; Qu, W.; Han, F.; Liu, D.; Zhang, W. Establishment of immortalized Schwann cells derived from rat embryo dorsal root ganglia. Int. J. Mol. Med. 2012, 30, 480–486. [Google Scholar] [CrossRef] [PubMed]
  174. Jessen, K.R.; Mirsky, R. The Success and Failure of the Schwann Cell Response to Nerve Injury. Front. Cell. Neurosci. 2019, 13, 33. [Google Scholar] [CrossRef] [PubMed]
  175. Mentkowski, K.I.; Tarvirdizadeh, T.; Manzanero, C.A.; Eagler, L.A.; Lang, J.K. Surface engineering enhances the therapeutic potential of systemically delivered extracellular vesicles following acute myocardial infarction. FASEB J. 2024, 38, e70070. [Google Scholar] [CrossRef]
  176. Ying, S.-Q.; Cao, Y.; Zhou, Z.-K.; Luo, X.-Y.; Zhang, X.-H.; Shi, K.; Qiu, J.-Y.; Xing, S.-J.; Li, Y.-Y.; Zhang, K.; et al. Hepatocyte-derived tissue extracellular vesicles safeguard liver regeneration and support regenerative therapy. J. Nanobiotechnol. 2024, 22, 521. [Google Scholar] [CrossRef]
  177. Larson, A.; Natera-Rodriguez, D.E.; Crane, A.; Larocca, D.; Low, W.C.; Grande, A.W.; Lee, J. Emerging Roles of Exosomes in Stroke Therapy. Int. J. Mol. Sci. 2024, 25, 6507. [Google Scholar] [CrossRef]
  178. Rehman, F.U.; Liu, Y.; Zheng, M.; Shi, B. Exosomes based strategies for brain drug delivery. Biomaterials 2023, 293, 121949. [Google Scholar] [CrossRef]
  179. Einabadi, M.; Ai, J.; Kargar, M.; Kafilzadeh, F.; Nooshabadi, V.T.; Jamali, H. Mesenchymal cell-derived exosomes as novel useful candidates for drug delivery. Arch. Neurosci. 2020, 7, e98722. [Google Scholar] [CrossRef]
  180. Wang, H.; Zhang, X.; Zhang, W.; Sun, H. The effects of exosomes originating from different cell sources on the differentiation of bone marrow mesenchymal stem cells into Schwann cells. J. Nanobiotechnol. 2024, 22, 220. [Google Scholar] [CrossRef]
  181. Tan, X.; Xiao, H.; Yan, A.; Li, M.; Wang, L. Effect of Exosomes From Bone Marrow–Derived Mesenchymal Stromal Cells and Adipose-Derived Stromal Cells on Bone-Tendon Healing in a Murine Rotator Cuff Injury Model. Orthop. J. Sports Med. 2024, 12, 23259671231210304. [Google Scholar] [CrossRef] [PubMed]
  182. Zheng, Y.; Lu, H.; Mu, Q.; Yi, P.; Lin, L.; Li, P.; Yu, D.; Zhao, W. Effects of sEV derived from SHED and DPSC on the proliferation, migration and osteogenesis of PDLSC. Regen. Ther. 2023, 24, 489–498. [Google Scholar] [CrossRef] [PubMed]
  183. Deng, H.; Zhu, L.; Zhang, Y.; Zheng, L.; Hu, S.; Zhou, W.; Zhang, T.; Xu, W.; Chen, Y.; Zhou, H.; et al. Differential Lung Protective Capacity of Exosomes Derived from Human Adipose Tissue, Bone Marrow, and Umbilical Cord Mesenchymal Stem Cells in Sepsis-Induced Acute Lung Injury. Oxid. Med. Cell. Longev. 2022, 2022, 7837837. [Google Scholar] [CrossRef] [PubMed]
  184. Walravens, A.-S.; Smolgovsky, S.; Li, L.; Kelly, L.; Antes, T.; Peck, K.; Quon, T.; Ibrahim, A.; Marbán, E.; Berman, B.; et al. Mechanistic and therapeutic distinctions between cardiosphere-derived cell and mesenchymal stem cell extracellular vesicle non-coding RNA. Sci. Rep. 2021, 11, 8666. [Google Scholar] [CrossRef] [PubMed]
  185. Niada, S.; Giannasi, C.; Magagnotti, C.; Andolfo, A.; Brini, A.T. Proteomic analysis of extracellular vesicles and conditioned medium from human adipose-derived stem/stromal cells and dermal fibroblasts. J. Proteom. 2021, 232, 104069. [Google Scholar] [CrossRef]
  186. Wang, A.; Liu, J.; Zhuang, X.; Yu, S.; Zhu, S.; Liu, Y.; Chen, X. Identification and Comparison of piRNA Expression Profiles of Exosomes Derived from Human Stem Cells from the Apical Papilla and Bone Marrow Mesenchymal Stem Cells. Stem Cells Dev. 2020, 29, 511–520. [Google Scholar] [CrossRef]
  187. Merckx, G.; Hosseinkhani, B.; Kuypers, S.; Deville, S.; Irobi, J.; Nelissen, I.; Michiels, L.; Lambrichts, I.; Bronckaers, A. Angiogenic Effects of Human Dental Pulp and Bone Marrow-Derived Mesenchymal Stromal Cells and their Extracellular Vesicles. Cells 2020, 9, 312. [Google Scholar] [CrossRef]
  188. Villatoro, A.; Alcoholado, C.; Martín-Astorga, M.; Fernández, V.; Cifuentes, M.; Becerra, J. Comparative analysis and characterization of soluble factors and exosomes from cultured adipose tissue and bone marrow mesenchymal stem cells in canine species. Vet. Immunol. Immunopathol. 2019, 208, 6–15. [Google Scholar] [CrossRef]
Figure 1. Three main strategies to modify exosomes (created with BioRender.com).
Figure 1. Three main strategies to modify exosomes (created with BioRender.com).
Pharmaceutics 17 00147 g001
Figure 2. Different factors that may influence the exosome-based therapeutic effects (created with BioRender.com).
Figure 2. Different factors that may influence the exosome-based therapeutic effects (created with BioRender.com).
Pharmaceutics 17 00147 g002
Figure 3. Cellular sources of exosomes that are widely researched in regenerative medicine (created with BioRender.com).
Figure 3. Cellular sources of exosomes that are widely researched in regenerative medicine (created with BioRender.com).
Pharmaceutics 17 00147 g003
Figure 4. Characteristics of exosomes and their potential applications (created with BioRender.com) (miRNA: microRNA, ROS: reactive oxygen species, M1: macrophage 1, M2: macrophage 2).
Figure 4. Characteristics of exosomes and their potential applications (created with BioRender.com) (miRNA: microRNA, ROS: reactive oxygen species, M1: macrophage 1, M2: macrophage 2).
Pharmaceutics 17 00147 g004
Figure 5. Underexplored exosomal directions in regenerative medicine (created with BioRender.com).
Figure 5. Underexplored exosomal directions in regenerative medicine (created with BioRender.com).
Pharmaceutics 17 00147 g005
Table 1. Examples of different exosomal dosages and exosomal administration routes.
Table 1. Examples of different exosomal dosages and exosomal administration routes.
Exosomal
Species Origin
Cellular SourceApplication/EffectThe Most Effective Dosage/ConcentrationAdministration RouteRefs.
RatsBMSCsciatic nerve crush injury0.9 × 1010 particles/mLin vitro administration[35]
HumanADSCwound healing200 μg/mLlocal administration[36]
HumanAlzheimer’s disease4 × 108 particles in saline (1 mL) two times per weekintranasally administration[37]
HumanUCMSCcomplex perianal fistulas10 ug/100 uL per ratlocal administration[38]
HumanHUVECvascular repair0.35–1.75 μg/mLin vitro administration[39]
repair of intestinal structure and function in rats with severe burn injuries200 μg/0.5 mL per ratintraperitoneal injection[40]
Table 2. The examples of exosome applications mentioned above.
Table 2. The examples of exosome applications mentioned above.
Exosome SourcesApplication AreasSpecific EffectsRefs.
BMSCBone and cartilage regeneration; wound healing; osteonecrosis of the Femeral Head; immunomodulation; myocardial infarction; nervous system injury(+)Osteogenesis; angiogenesis; keratinocyte and DF proliferation; astrocyte differentiation; BMSC proliferation and differentiation; NSC differentiation[55,56,57,58,59,60,61]
(−)Inflammation; T cell proliferation; glial scar
ADSCWound healing; scar prevention; osteoarthritis; immunomodulation; myocardial ischemia; hair loss; skin photoaging(+)Angiogenesis; adipogenesis; keratinocyte and DF proliferation; collagen synthesis; growth of hair follicles; dermal papilla cell proliferation[55,57,70,71,72,73,74,75,76,77]
(−)Inflammation; fibrotic; cartilage degeneration; skin photoaging; T cell proliferation
Human UCMSCWound healing; cutaneous regeneration; colitis; autoimmune uveoretinitis; GVHD(+)Keratinocyte and DF proliferation; intestinal lymphatic drainage improvement[55,57,80,81,82,83,85]
(−)Inflammation; T cell proliferation; macrophage pyroptosis; lymphangiogenesis; endoplasmic reticulum stress in CD4+ T cells
Human AMMSCLiver fibrosis; nonalcoholic steatohepatitis; wound healing; hypoxia/ischemia-induced cerebral palsy; acute TSCI(+)Reepithelization; collagen synthesis; angiogenesis; axonal regeneration[92,93,94,95,96,97]
(−)Inflammation; activation of Kupffer cell and hepatic stellate cell; excessive apoptosis; astrogliosis; blood-spinal cord barrier leakage; spinal cord edema
Human CMMSCOsteoarthritisCould be uptaken by different types of cells isolated from tissues associated with osteoarthritis[98]
Human CVMSCHypoxic placenta(+)Trophoblast migration and proliferation; placental vascular adaptation to low oxygen tension[99,100]
Human DCMSCWound healing(+)Fibroblast proliferation, migration, and differentiation; fibroblast senescent state improvement; collagen deposition[101]
(−)Oxidative stress
DPSCWound healing; dental pulp-like tissue regeneration; molar defect; periodontitis; cerebral I/R injury; subarachnoid hemorrhage; sciatic nerve injury; mandibular bone defect(+)Odontogenic differentiation of DPSC; dentinogenesis; neuroprotection; SC proliferation; migration and secreting neurotrophic factors; osteogenesis[106,107,108,109,110,111,112,113,114,115,116,117]
(−)Imbalance of Th17/Treg; inflammation; alveolar bone loss; neuronal apoptosis; microglial pyroptosis; brain edema
M2Fracture healing; wound healing; chronic rotator cuff tear; contusion SCI; calvarial bone defect(+)Macrophage polarization (M1 to M2); osteogenesis; angiogenesis; reepithelialization; collagen deposition; neurogenesis; tube formation, migration and proliferation of brain endothelial cell[122,123,124,125,127,128,130]
(−)Inflammation; cellular senescence of BMSC
TregSkin xenograft transplantation; kidney allotransplantation; orthotopic liver transplantation; oxygen-glucose deprivation/reperfusion; wound healing; acute myocardial infarction(+)Naive T cell converting into Treg; migration of human DF and HUVEC; macrophage polarization (M1 to M2)[120,131,132,133,134,135,136]
(−)Inflammation; effector T cell proliferation; BV-2 microglia apoptosis; myocardial cell apoptosis
NSCThromboembolic stroke; SCI; Alzheimer’s disease; wound healing(+)Neurogenesis; angiogenesis; neurite remodeling; autophagy activation of spinal cord neuron; human DF migration; tube formation of HUVEC[137,138,139,140,141,142,143]
(−)Brain atrophy; neuronal apoptosis; lipopolysaccharide-induced nitric oxide production by macrophage; activation of microglia; inflammation; BBB leakage; aging
FibroblastWound healing; nerve defect; skin photoaging; atopic dermatitis(+)ECM formation; angiogenesis; collagen deposition and maturity; axon regeneration and functional recovery; Schwann cell-mediated peripheral neuron myelination; [144,146,147,148,149,150,151,152]
(−)Scar formation; oxidative stress; inflammation; collagen degradation
HUVECWound healing; hypoxia/reoxygenation; flap transplantation; transient cerebral I/R; cranial defect; SCI(+)HUVEC and human cutaneous keratinocyte migration; angiogenesis; skin proliferation; reepithelialization; granulation tissue formation; vascularization; endothelial progenitor cell proliferation, tube formation, and invasion; nerve cell migration and invasion; osteogenesis; macrophage polarization (M1 to M2); osteogenic differentiation and migration of BMSC[153,155,156,157,158,159,160,161,162]
(−)Inflammation; scar proliferation; endoplasmic reticulum stress; neural cell apoptosis
SCNerve crush; nerve axotomy; OGD-injured motoneuron; optic nerve crush; denervated muscle atrophy: SCI; cranial defect; CMS-induced DRG injury;(+)Axonal regeneration; motoneuron repair; angiogenesis; injured neuron autophogy; mitophagy; BMSC osteogenesis; proliferation and multipotency of human dental pulp cell; proliferation of injured DRG cell[147,163,164,165,166,167,168,169,170,171,172]
(−)Oxidative stress; inflammation; mitochondrial damage; senescence of injured DRG cell
((+): promoting effects, (−): suppressing effects).
Table 3. Studies comparing different types of exosomes and their findings.
Table 3. Studies comparing different types of exosomes and their findings.
Species OriginExosomesIn Vitro Model and FindingsIn Vivo Model and FindingsRefs.
Ability/EffectComparing FindingsAbility/EffectComparing Findings
RatsSC-Exos vs. FC-Exos vs. NSC-ExosThe ability to induce BMSCs into SCs SC-Exos  ≥  NSC-Exos >  FC-Exos > ControlNANA[180]
MiceBMSC-Exos vs. ADSC-ExosPromotion of proliferation, migration, osteogenic differentiation, and chondrogenic differentiation ability of BMSCsBMSC-Exos ≈ ADSC-ExosThe ability to accelerate bone-tendon injury healing in murine rotator cuff injury modelBMSC-Exos ≈ ADSC-Exos[181]
HumanSmall extracellular vesicles derived from stem cell from human exfoliated deciduous teeth (SHED-sEVs) vs. Small extracellular vesicles derived from DPSC (DPSC-sEVs)Promotion of the proliferation, migration, and osteogenesis of periodontal ligament stem cells (PDLSCs)SHED-sEVs > DPSC-sEVsNANA[182]
HumanBMSC-EVs vs. ADSC-EVs vs. UCMSC-EVs vs. Extracellular vesicles derived from dermal stem cell (DSC-EVs) vs. DPSC-EVsProductivityUCMSC-EVs > othersBiodistribution in full-sickness skin defect mouse modelsEnrichment in the spleen, lungs, kidneys, and lymphonodus: UCMSC-EVs > others, in bone marrows: BMSC-EVs > others.[104]
Cell affinityImmune cells and recipient cells in tissue regeneration: UCMSC-EVs > others, neuroblastoma cells: DPSC-EVs > others.Wound-healing potential in full-sickness skin defect mouse modelsBMSC-EVs ≈ CMSC-EVs ≈ DSC-EVs > ADSC-EVs ≈ DPSC-EVs
Drug loading/delivery capacityUCMSC-EVs/DPSC-EVs > others
HumanBMSC-Exos vs. ADSC-Exos vs. UCMSC-ExosSuppressing glycolysis and pro-inflammatory cytokine release in LPS-treated macrophagesADSC-Exos > othersAlleviating sepsis-induced ALI and systemic inflammation and improving survival of ALI miceADSC-Exos > others[183]
HumanCDC-EVs vs. BMSC-EVs vs. ADSC-EVsUpregulating the Arg1/Nos2 ratio of peritoneal Mϕ of thioglycolate-stimulated miceCDC-EVs > MSC-EVsReduction in scar size and increase in infarct wall thickness in a mouse model of MICDC-EVs > MSC-EVs[184]
HumanBMSC-Exos vs. ADSC-Exos vs. WJMSC-ExosEnhancing neuronal differentiation, postponing neutrophil apoptosis, and PBMC proliferation.BMSC-Exos > othersNANA[57]
Promoting angiogenesisWJMSC-Exos > others
HumanBMSC-EVs vs. ADSC-EVsPromoting endothelial cell proliferation
The effects on the most relevant cell types involved in skin wound healing
BMSC-EVs > ADSC-EVsAccelerating wound closure in a mouse model of diabetic ulcersADSC-EVs > BMSC-EVs[56]
Promoting fibroblast, keratinocyte, and endothelial cell viabilityBMSC-EVs > ADSC-EVs
Promoting migration of endothelial cellsADSC-EVs > BMSC-EVs
Inducing vessel formationADSC-EVs > BMSC-EVs
HumanADSC-EVs vs. Extracellular vesicles derived from DF (DF-EVs)Top 3 enriched biological pathwaysADSC-EVs: Positive regulation of macrophage cytokine production, type B pancreatic cell proliferation, and positive regulation of I-κB kinase/NF-κB signalingNANA[185]
DF-EVs: non-canonical Wnt signaling pathway, Wnt signaling pathway, planar cell polarity pathway, and receptor-mediated endocytosis
HumanBMSC-Exos vs. ADSC-Exos vs. UCMSC-ExosPotential applications of exosomes in different fields via proteomicsBMSC-Exos: superior regeneration abilityNANA[80]
ADSC-Exos: immune regulation
UCMSC-Exos: tissue damage repair
HumanBMSC-Exos vs. Exosomes derived from stem cells from the apical papilla (SCAP-Exos)Different expression profiles of PIWI-interacting RNAs (piRNAs) and the related functionsThe higher expression of piRNAs in BMSC-Exos: the regulation of apoptosis and osteogenic differentiationNANA[186]
The higher expression of piRNAs in SCAP-Exos: metabolism, cell proliferation and differentiation, and other signaling pathways closely related to the development of teeth and the formation of bone tissue
HumanBMSC-EVs vs. DPSC-EVsChemotactic capacity on endothelial cellsBMSC-EVs > ADSC-EVs, this difference was neutralized when the results were normalized for the higher particle secretion of BMSCs.NANA[187]
HumanBMSC-Exos vs. ADSC-Exos vs. UCMSC-ExosInducing primary DF proliferationBMSC-Exos > othersNANA[55]
Stimulating keratinocyte migrationUCMSC-Exos > others
Canine speciesBMSC-EVs vs. ADSC-EVsProductivityBMSC-Exos > ADSC-ExosNANA[188]
Differential function of exosomal proteinsBMSC-Exos: cell differentiation, cell organization and biogenesis, cellular component and movement, metabolic process, regulation of biological process, response to stimulus and transport
HumanBMSC-Exos vs. ADSC-ExosSuppressing the differentiation of CD4 + T cells into Th17 and the secretions of pro-inflammatory factors IL-17 and TNF-αDPSC-Exos > BMSC-ExosNANA[108]
Promoting the polarization of CD4 + T cells into Treg and increasing the release of anti-inflammatory factors IL-10 and TGF-βDPSC-Exos > BMSC-Exos
Inducing apoptosis of CD4 + T cellsDPSC-Exos > BMSC-Exos
HumanNSC-EVs vs. MSC-EVsNANAImproving cellular, tissue, and functional outcomes in middle-aged mouse TE stroke modelsNSC-EVs > MSC-EVs[137]
HumanBMSC-Exos vs. DPSC-ExosNeuroprotective propertiesBMSC-Exos ≈ DPSC-ExosNANA[109]
(NA: not applicable).
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Chen, Y.; Qi, W.; Wang, Z.; Niu, F. Exosome Source Matters: A Comprehensive Review from the Perspective of Diverse Cellular Origins. Pharmaceutics 2025, 17, 147. https://doi.org/10.3390/pharmaceutics17020147

AMA Style

Chen Y, Qi W, Wang Z, Niu F. Exosome Source Matters: A Comprehensive Review from the Perspective of Diverse Cellular Origins. Pharmaceutics. 2025; 17(2):147. https://doi.org/10.3390/pharmaceutics17020147

Chicago/Turabian Style

Chen, Yiru, Weikun Qi, Zhenghao Wang, and Feng Niu. 2025. "Exosome Source Matters: A Comprehensive Review from the Perspective of Diverse Cellular Origins" Pharmaceutics 17, no. 2: 147. https://doi.org/10.3390/pharmaceutics17020147

APA Style

Chen, Y., Qi, W., Wang, Z., & Niu, F. (2025). Exosome Source Matters: A Comprehensive Review from the Perspective of Diverse Cellular Origins. Pharmaceutics, 17(2), 147. https://doi.org/10.3390/pharmaceutics17020147

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

Article Metrics

Back to TopTop