Next Article in Journal
Toxoplasma gondii in Wild Boars (Sus scrofa) in Germany: Serological Screening from Thuringia
Previous Article in Journal
Differences in Milk Fatty Acids Profile of Two Breeds of Water Buffaloes Explained by Their Gastrointestinal Microbiota
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Therapeutic Application of Extracellular Vesicles Derived from Mesenchymal Stem Cells in Domestic Animals

1
Department of Veterinary Medical Sciences, University of Bologna, Via Tolara di Sora 50, Ozzano dell’Emilia, 40064 Bologna, Italy
2
Health Science and Technologies Interdepartmental Center for Industrial Research (HST-ICIR), University of Bologna, 40100 Bologna, Italy
*
Author to whom correspondence should be addressed.
Animals 2024, 14(15), 2147; https://doi.org/10.3390/ani14152147
Submission received: 9 June 2024 / Revised: 12 July 2024 / Accepted: 20 July 2024 / Published: 24 July 2024
(This article belongs to the Section Veterinary Clinical Studies)

Abstract

:

Simple Summary

Extracellular vesicles (EVs) secreted by mesenchymal stem cells (MSCs) have recently been described in human and veterinary medicine and great interest is directed toward them for their therapeutic potential. EVs are vesicles produced and secreted by Mesenchymal Stem Cells (MSCs) that possess the same anti-inflammatory and regenerative properties as the cells themselves but seem safer for clinical applications because of lower immunogenicity and lower size. MSCs that produce EVs can be derived from adult tissues such as bone marrow or adipose tissue or from fetal adnexa such as amniotic membrane and Wharton’s jelly. This manuscript gives a summary of the current description of composition, characteristics, sources, and function of EVs, focusing especially on their therapeutic applications in veterinary medicine to date. Knowledge of these features and the steps taken to date will help researchers and veterinary practitioners discover new approaches to regenerative medicine that are ever closer to being achievable.

Abstract

Recently, the therapeutic potential of extracellular vesicles (EVs) derived from mesenchymal stem cells (MSCs) has been extensively studied in both human and veterinary medicine. EVs are nano-sized particles containing biological components commonly found in other biological materials. For that reason, EV isolation and characterization are critical to draw precise conclusions during their investigation. Research on EVs within veterinary medicine is still considered in its early phases, yet numerous papers were published in recent years. The conventional adult tissues for deriving MSCs include adipose tissue and bone marrow. Nonetheless, alternative sources such as synovial fluid, endometrium, gingiva, and milk have also been intermittently used. Fetal adnexa are amniotic membrane/fluid, umbilical cord and Wharton’s jelly. Cells derived from fetal adnexa exhibit an intermediate state between embryonic and adult cells, demonstrating higher proliferative and differentiative potential and longer telomeres compared to cells from adult tissues. Summarized here are the principal and recent preclinical and clinical studies performed in domestic animals such as horse, cattle, dog and cat. To minimize the use of antibiotics and address the serious issue of antibiotic resistance as a public health concern, they will undoubtedly also be utilized in the future to treat infections in domestic animals. A number of concerns, including large-scale production with standardization of EV separation and characterization techniques, must be resolved for clinical application.

1. Introduction

The study and application of mesenchymal stem cells (MSCs) has exponentially increased over the past decade since their versatility, safety and great potential exhibit a strong attraction for researchers [1]. In particular, MSCs are studied extensively for their application in regenerative medicine and tissue engineering [2] and they can be isolated from adult or fetal tissues [3,4]. In fact, there are a great number of in vitro and clinical studies on humans, and laboratory studies on small and large animals. The key to MSCs therapeutic potential is their ability to migrate to sites of tissue injury or inflammation, perceive hypoxia and tissue damage [5], stimulate endogenous repair of injured tissues [6], and modulate immune responses [7,8,9,10]. MSCs abilities are due to the fact they release many factors (composing the secretoma) into the environment (in vivo) or into the culture medium (in vitro) forming, in the latter case, the conditioned medium (CM). The secretoma is composed of small soluble factors (chemokines, cytokines, and hormones) and extracellular vesicles (EVs) that provide a vehicle for the transfer of lipids, proteins, and nuclear acids from one cell to another [11,12].
EVs are secreted from many cell types and differ with respect to their origin within the cell, size, and contents [13,14,15]. Since they are not only secreted by MSCs, the biological significance of EVs has for many years been largely overlooked, with them regarded like apoptotic bodies, as merely cellular fragments or debris [13,14,15]. By the most recent definitions [16,17] there are two main classes of EVs: exosomes, derived from budding of endosomal membranes and ranging in size from 40 to 100 nm, and microvesicles (MVs), which originate from outward budding and fission of the plasma membrane and which range from 50 nm to 1 μm in size. The diversity of proteins, lipids, and nucleic acids contained in EVs depends on their cell of origin and may be influenced by physiological stress or other conditions [14,18,19,20]. EVs have an important role in intercellular communication and are able to modify the activity of target cells through interactions with surface receptors and the transfer of proteins, mRNAs, and miRNAs [14]. EVs are responsible for the paracrine action of MSCs, exerting an effective mediating role that directly activates target cells, transferring information to damaged cells or stimulating adjacent cells to secrete other factors [16,21]. In particular, first, EVs may stimulate target cells directly by surface-expressed ligands, acting as signaling complexes [22]. Second, EVs may transfer surface receptors from one cell to another, deliver proteins, mRNA, bioactive lipids and even whole organelles (e.g., mitochondria) into target cells [22]. As an editorial noted, this fascinating EVs-mediated cell–cell communication system developed very early in evolution and served as a model for further development of intercellular interaction mechanisms involving soluble bioactive mediators and precision ligand–receptor interactions [22]. The earliest descriptions of EVs were performed in the early 1980s and EVs were initially thought to represent a mechanism for the elimination of proteins and other undesirable molecules [13,23]. Since the exact route that MSC-EVs follow from donor cells to recipient cells is still unknown, previous research has shown several important features [24]. EVs’ surface molecules have the ability to both direct them to recipient cells and prevent the inside components from deteriorating [24,25,26]. EVs can enter cells by a variety of routes once they are connected to a target cell, such as receptor–ligand contact, internalization through phagocytosis and/or endocytosis, or direct fusion with the plasma membrane [25,26]. Nevertheless, EVs have the capacity to interact with recipient cells by delivering a particular cargo directly into the cytoplasm [24,25,26].
The great attraction for the therapeutic use of EVs is derived from the reduced risks associated with MSCs grafting, from possible immune reactions against MSCs, and especially from the opportunity to load or modify the content of bioactive factors to address specific therapeutic needs [15]. Indeed, MSC-EVs have the same therapeutic potential as MSCs, which includes stimulating angiogenesis, encouraging cell migration and proliferation, inhibiting apoptosis, and reducing inflammation [24]. Furthermore, the quick development of EV-based treatment has been spurred by recent advances in nanotechnology. Because MSC-EVs are naturally biocompatible, biodegradable, and non-immunogenic, they are a promising class of nanomaterials for drug delivery applications [27].
These properties make MSC-EVs a viable method for EV-based therapies and they have been discovered to be relevant in a lot of fields. In human medicine, the main areas of application of EVs in preclinical studies conducted in laboratory animals have been cardiovascular disease [28,29,30], kidney [31,32,33], liver [34,35], lung injury and diseases [36,37], wound healing [38,39,40], tumor growth [41,42,43] ophthalmology [44,45], immune-related diseases [46,47,48,49,50], neurological diseases [51,52,53] and musculoskeletal system [54,55,56].
The present review will provide an overview of clinical–therapeutic applications of MSCs-EVs in veterinary medicine. First of all, the characterization of EVs derived from adult tissues and fetal adnexa is presented. Preclinical research carried out in sheep, pigs, and rodents will be mentioned in addition to the therapeutic applicability in vitro and in vivo for the main domestic animals. Furthermore, the future perspectives of the clinical application will be discussed to highlighting the importance of the challenges that need to be addressed and solved to make routine MSCs-EV therapy possible in domestica animals.
To conduct the present study, relevant information was sourced from databases such as Medline and Science Direct, Google Scholar, Pubmed, Scopus and CAB Abstracts. Multiple search queries were employed to find all related articles. Full-text reports in the form of reviews or research articles written in English language were included, while conference or congress contributions were excluded.

2. Characterization and Sources

EVs are nano-sized particles containing biological components commonly found in other biological materials [57]. For that reason, EV isolation and characterization are critical to draw precise conclusions during their investigation. In this context, the International Society for Extracellular Vesicles (ISEV) identified the minimal experimental requirements for the definition of EVs and their functions (MISEV 2014) [57]. Initially, they proposed this thorough documentation of the origin of materials and methods used for isolation, emphasizing their influence on the ability to replicate findings. Secondly, they advocated for a comprehensive assessment of the protein composition, including quantifying proteins anticipated to be concentrated in extracellular EVs. Moreover, they insisted on employing two complementary methods for characterizing individual particles. Lastly, the functional analysis should encompass appropriate control samples.
The initial suggestions underwent revision in 2018 [17]. MISEV2018 is presently regarded as the fundamental set of information to be disclosed when presenting findings involving EVs. The guidelines were enhanced by urging the provision of more detailed data concerning sample collection, as well as cell-culture specifics for EV gathering and storage. The reporting of isolation and concentration methods was emphasized, and no specific purification protocol was recommended. The catalog of methods for EV characterization was extensively updated to include recent advancements in single-particle techniques. Quantification of fundamental components like lipids, proteins, and RNA, along with their ratios (e.g., particle-to-protein ratio), was highlighted as crucial for assessing EV enrichment [17]. It was suggested that dose–response studies should be performed using EV preparations normalized to particle count or another approach of quantifying the biological cargo of the EVs, and to use multiple control samples, including distinct fractions of the enrichment protocol, in light of the significantly expanded list of specific protein markers [17]. Although EV research in veterinary medicine is still in its early phases, several studies have been published recently. It is strongly advised that specialized EV scientists and emerging EV-focused veterinary researchers work closely together to minimize biases and technical issues, provide optimal outcomes, and maximize the potential of group EV research initiatives [58].

2.1. Adult Tissues

The conventional adult tissues for deriving MSCs include adipose tissue and bone marrow. Nonetheless, alternative sources such as synovial fluid [59], endometrium [60], gingiva [61], and milk [62] have also been intermittently used. MSC-derived EVs have undergone purification and investigation across various domestic animal species including pig [63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90], horse [59,60,91,92,93,94,95,96,97,98,99,100,101,102,103,104], dog [61,105,106,107,108,109,110,111,112,113,114,115,116,117], cat [118,119,120,121,122], cow [62], and sheep [123]. While not all studies have comprehensively characterized EVs [58], most have involved isolation, purification, and characterization procedures focusing on size, morphology, and protein composition. Some studies have delved into nucleic acid content, whereas lipid quantification has been performed in only two studies focusing on Lyosecretome [98,112]. Tetraspanins (CD9, CD63, CD81) are commonly employed as identifying markers for EVs, although other biomarkers such as CD29, CD73, CD40, TSG101, β-catenin, and β1-integrins have also been utilized. The collective characterization of MSCs-EVs sourced from adult tissues in domestic animals is illustrated in Figure 1.

2.2. Fetal Adnexa

Cells derived from fetal adnexa exhibit an intermediate state between embryonic and adult cells [124,125,126,127], demonstrating higher proliferative and differentiative potential and longer telomeres compared to cells from adult tissues [128,129,130]. These characteristics are related to the early embryological origin of MSCs derived from fetal adnexa. Following blastocyst implantation, the inner cell mass undergoes morphological changes, leading to the formation of the bilaminar embryonic disc, which consists of the epiblast and the hypoblast. Some cells from the hypoblast migrate along the outer edges of the extraembryonic reticulum to form a connective tissue known as the extraembryonic mesoderm, which surrounds the yolk sac and amniotic cavity, later forming the amniotic mesoderm and chorionic mesoderm [131]. These cells, derived from extraembryonic mesoderm, are fundamental for maintaining feto–maternal tolerance during pregnancy, having important immunomodulatory characteristics and low immunogenicity [131,132]. Despite these important attributes of MSCs from fetal adnexa, MSCs-EVs have been purified and investigated only in equine and canine species [3,133,134,135,136,137,138,139,140,141,142].
In equine species, only Iacono et al. [3] noticed the presence of complex extracellular vesicles measuring 500 nM–1 μM, observed in MSCs from Wharton’s jelly (WJ) using TEM. In contrast, the research group of Lange-Consiglio reported in various studies [133,134,135] that amniotic membrane (AM)-MSCs produce EVs ranging in size from 100 nm to 1000 nm, with a predominance of vesicles between 100 and 200 nm, which they considered as shedding vesicles. In 2018, Lange-Consiglio et al. [135] performed miRNA sequencing of EVs derived from AM-MSCs for the first time, finding that EVs contain a lower percentage of miRNAs than AM-MSCs and that several miRNAs are enriched hundreds or thousands of times in EVs, while others remain at the same level as in AM-MSCs. Moreover, the authors reported that many of the miRNAs enriched in EVs regulate the inflammatory response, such as the overexpression of miR-146, which decreases the expression of the inflammatory cytokine IL-6 in lipopolysaccharide (LPS)-stimulated macrophage cells, and MiR-223, which negatively regulates the expression of many inflammatory genes in macrophage cells. The same research group supported these observations using AM-MSCs-EVs, in an in vitro model, to counteract the stress induced by LPS in endometrial, lung, and tendon cells [133,134,136]. In all cases, the authors observed the incorporation of EVs within cells and the downregulation of tumor necrosis factor-alpha (TNF-α), interleukin 6 (IL-6), IL-1β, matrix metalloproteinase-1 (MMP-1), and MMP-13 genes. Most recently, the same research group evaluated the surface glycosylation pattern of AM-MSCs-EVs released in conditioned medium, using a microarray procedure [137]. The signal intensity detected by microarray scanner indicated a high simultaneous presence of Galβ1,3GalNAc, α2,3 sialic acid, and high-mannose N-linked glycans, which the authors suggest may constitute markers of AM-MSCs-EVs in equine species.
Canine MSCs are of interest for both veterinary and comparative models of disease; however, in this species, as well as in humans, there are difficulties in acquiring adult tissues and there are ethical implications. For these reasons, in recent years, researchers’ attention has been directed towards identifying innovative sources of MSCs and EVs from easily accessible materials. In 2019, Crain et al. [138] reported that canine WJ-MSCs produce EVs of 125 nm in diameter. In this preliminary study on the mechanism of immune EVs modulation in canines, the authors observed that EVs inhibited CD4 T cell proliferation in a dose-dependent manner, hypothesizing a mechanism regulated by a TGF-βRI antagonist, neutralizing antibodies to TGF-β, or the A2A adenosine receptor blockade. Most recently, Wright et al. [139] characterized EVs derived from umbilical cord (UC) cells in canines, following MISEV guidelines. Canine UC-MSCS-EVs were found to be within the size range of exosomes (50–150 nm) with a median protein concentration of 3 g/mL. Isolated EVs were positively stained for CD9, CD63, CD81, ALIX protein, and CD142 (TF: Tissue Factor), similar to canine UC-MSCs. These findings indicate that EVs potentially share TF expression and potential pro-coagulant activity with MSCs of origin, which must be considered when using them as therapeutic agents. Regarding EVs derived from canine AM-MSCs, Karam et al. compared their morphological aspects at different culture passages, finding that, while the EV size did not differ between culture passages, their number decreased from passage 0 to passage 2 of in vitro culture [140]. Based on these observations, the authors suggest that cell–cell communication is greater in the early phase, making this the optimal phase for clinical EV application. These results were confirmed by Scassiotti et al. [141]. In line with the findings reported in human amniotic fluid and membranes, Pastore et al. [142] isolated EVs of different sizes enriched for Alix (ALG-2 interacting protein X), CD81 (Tetraspanin-28), and TSG101 (Tumor susceptibility gene 101) after sequential centrifugations. Since CD59 seems to be involved in regulating the immunomodulation of feto–maternal interaction during pregnancy, the authors investigated the expression of this cluster by AM-MSCs-EVs in canines. All isolated EVs fractions expressed CD59, indicating that EVs derived from the amnion carry a complement inhibitor and play a crucial role in promoting immune tolerance to embryo–fetal antigens and reducing the risk of abortion in dogs.

3. Clinical Application of EVs

The therapeutic potential of MSCs-EVs has been increasingly studied over the past decade.

3.1. Preclinical Studies

In pigs and sheep there has been less research than that on rodents, but it is all quite recent. Application areas in sheep include the musculoskeletal system, neurological system, and sepsis/pneumonia, while in pigs the respiratory and circulatory systems and renal/liver injury are currently being utilized. The main clinical studies conducted on these species are summarized in Table 1.
Only recently, some studies have characterized MSCs-EVs in the equine and canine species [101,141]. More recently, domestic animals, such as horse, dog, and cat have also been used in preclinical and clinical studies. Table 2 summarizes the main preclinical studies carried out in domestic animals. Regarding preclinical studies, EVs derived from adult and fetal tissues have been applied to cultures of tenocytes, chondrocytes, fibroblasts, and endometrial cells, during in vitro embryo production, and to alveolar macrophages, microglial cells and mesenchymal stem cells.

3.2. Clinical Studies/Applications

Table 3 summarizes the main clinical studies involving the in vivo application of EV-MSCs in veterinary medicine.

3.2.1. Orthopedic Field

Two studies have been conducted in the orthopedic field: one study involved one horse with ligament injury [96], and three dogs with spontaneous osteoarthritis [98]. In the ligament injury, the injection of 25 μg/mL EVs increased angiogenesis and elasticity in the ligament injury and also promoted lesion filling without adverse reaction [96]. The study conducted in the dog used a product called Lyosecretome (freeze-dried secretome), which has previously created by the same research group using MSCs obtained from human adipose tissue [150]. In order to concentrate and purify the MSC-derived secretome, this procedure included an ultrafiltration phase. After that, the secretome was freeze-dried to provide a powdered dosage form with improved long-term stability [98]. Dogs suffering osteoarthritis received 20 mg, or 2 × 106 cell equivalents, resuspended in hyaluronic acid for application [98]. Allogeneic Lyosecretome injected intra-articularly is safe and does not cause a clinically relevant systemic or local adverse response [98].

3.2.2. Reproductive Field

In the reproductive field, two studies have been conducted on mares with chronic endometritis [103,149]. A case report was conducted on an 11-year-old Friesian mare with a history of failed pregnancies despite numerous insemination attempts [150]. Two treatments with 20 billion EVs diluted in 50 mL of NaCl 0.9% were performed, followed by an uterine biopsy. The success of the intrauterine administration of EVs is demonstrated by an improvement in the classification of endometritis and in a successful artificial insemination with implantation of an embryo, as detected at day 14, and with a pregnancy that is still ongoing [149]. Day-8 equine embryos are thought to release EVs that transfer early pregnancy factors including HSP10 and miRNA, hence modulating the function of the oviductal epithelium [151]. The endometrial epithelium also secretes EVs, which target cellular pathways important for embryo implantation [152]. Pro-inflammatory cytokines, growth factors, and chemokines operate on fibroblasts and other cells in the physiopathological mechanisms of equine endometriosis, as well as in other inflamed tissues, influencing extra-cellular matrix deposition and tissue fibrosis [153].
In the second study of chronic endometritis, 14 mares were included and were divided into control and endometritis groups [103]. During the first and second ovulation, EVs were injected twice, separated by 21 days; 400 μg/mL of EVs was added to the sterile carboxymethylcellulose (CMC) at a concentration of 22 mg/mL. Each mL of the gel contained 200 μg EVs. The CMC gel with MSCs-EVs is preserved by lyophilization, and lyophilized EV solution was administered intrauterinely. Doppler and hormonal analysis were performed in addition to uterine biopsy [103]. After the second EV treatment, the histological evaluation revealed the regression of fibrous tissue and restoration of healthy endometrial glands with normal epithelium. In all treated mares, on the ninth day after insemination, an embryonic vesicle, and thus a pregnancy, was identified. The activity of EVs and the miRNAs contained in them triggered tissue regeneration, resulting in a restoration to the original histological features and thus, normal endometrial function. This probably restored the conditions needed for the appropriate implantation and development of maternal–embryonic paracrine communication [103].
The third study concerns the persistent post-breeding-induced endometritis, which is considered a major cause of subfertility in mares. Authors identified an optimal concentration of 400 × 106 EVs with 10 × 106 spermatozoa/mL: at this concentration, sperm mobility parameters were not negatively affected [149]. Semen alone or semen enhanced with EVs was used for insemination of sixteen susceptible mares. The supplementation resulted in a decrease in intrauterine fluid accumulation and polymorphonuclear neutrophil infiltration, along with a noteworthy decrease in intrauterine TNF-α and IL-6 and an increase in anti-inflammatory IL-10 in mares in the EV group, indicating effective regulation of the post-insemination inflammatory response [149].
Comparing these studies is challenging because of the limited sample size, and the substantial difference between the two considered diseases with a distinct etiology, as well as the different sources of EVs used (AM and BM). Despite this, the results are encouraging, although the different authors also measured and evaluated different molecules in addition to the common result represented by the positive diagnosis of pregnancy. A large clinical trial should be conducted on the problem-mares with persistent post-breeding-induced endometritis and chronic endometritis by standardizing the amount of EVs and the protocol of administration. Results should be supported by a histologic examination, in addition to a positive pregnancy diagnosis.
In cattle, the embryo culture was supplemented with or without 100 × 106 EVs/mL in Holstein Friesian cows, and this seemed to partially modify the expression of certain miRNAs involved in successful embryo implantation and prevent the detrimental effects of in vitro culture [146].

3.2.3. Skin-Wound Field

Regarding skin wounds, El-Tookhy et al. [105] used a dog full-thickness skin defect model. The wounds were induced using a dermal punch and the wounds were 2.5 cm apart. The results showed that MSCs-EVs significantly accelerated and increased cutaneous wound healing, collagen synthesis, and vascularization at wound sites, and showed faster wound closure. Additionally, it was determined that the application of EVs sped up the maturity of freshly created capillaries at wound sites, in addition to encouraging the formation of new ones [105] The amount of EV injected in this study equals the amount produced by a 2 × 106 MSCs/1 mL/wound [105].
One of the more complex biological processes that may be observed is wound healing, which requires coordinated action between cells, growth factors, and extracellular matrix proteins. Conducting well-designed studies is crucial to compare the literature and obtain practical insights that might aid clinicians, especially considering the substantial amount of research that has been carried out on this topic in both human and veterinary medicine. Dogs provide ideal research models, and studies on wound healing have used them as translational models for both human and veterinary applications [154].
Induced-wound sizes used in this study were performed considering the critical size defect for dogs [105]. Future trials should consider different types of wounds, such as those that have arisen spontaneously and those that are more difficult to heal such as pressure sores, as performed in other studies in which MSCs have been applied [155,156,157].

3.2.4. Urinary Tract

Artificial urinary occlusion was used to create the cat models of PR-AKI, which were subsequently treated with MSCs-EVs [122]. The infusion dose was equivalent to the amount of EV secreted by 10 million allogenic feline AT-MSCs in 24 h. Treatment with EVs was found to be effective in restoring plasma phosphorus, urea nitrogen, and creatinine. A routine blood examination revealed that the PR-AKI cats treated with EVs had faster return of their leukocytes to the normal physiological range than the control group. The plasma metabolome profile of PR-AKI cats treated with EVs was shown to be strikingly comparable to that of normal cats using ultra-high performance liquid chromatography analysis. Additionally, the examination of plasma demonstrated a strong correlation between the dynamic process of PR-AKI in cats and six metabolites found in plasma: carnitine, melibiose, d-glucosamine, cytidine, dihydroorotic acid, and stachyose [122]. The study demonstrated the efficacy of treatment with MSC-EVs and also discovered new PR-AKI indicators in addition to six metabolites, which may be potential targets for MSC-EVs treatment [122].
In the dog, Liu et al. 2023 [116] included 20 dogs as a model of renal ischemia-reperfusion injury, and the renal cortex of the left kidney was injected with EVs (180 μg/kg) in the experimental group. The EV treatment group showed reduced mitochondrial damage and a decrease in mitochondrial number as compared to the renal ischemia-reperfusion injury model group [116]. Renal ischemia-reperfusion injury resulted in severe histological abnormalities and significant increases in markers for renal function, inflammation and apoptosis, which were reduced by the infusion of MSCs-EVs [116].

3.2.5. Mammary Gland

Regarding the application of the mammary gland in cattle, 48 animals were enrolled, of whom 32 had acute mastitis and 16 had chronic mastitis [147]. After milking, the treated cows received CM through intramammary application with 3 mL of CM alone or, in the control group, they received the antibiotic alone, intramammarily, chosen from an antibiogram test, for three consecutive days. The standard experimental volume of CM was set at 3 mL since, after lyophilization, CM was concentrated 4-fold, and a uniform treatment protocol was needed to compare this to the antibiotic treatment [147]. In vitro results showed that the addition of CM inhibited CFU bacteria and decreased bacterial growth; in cell culture infected with S. aureus, cells died in 12 h, while with the addition of CM 60–89% of cells remained viable. Regarding the in vivo application, there was no difference in the improvement of clinically affected quarters treated with CM compared to antibiotic treatment, but the rate of relapse was different. There was no statistically significant difference between the antibiotic group compared to the CM groups but, in the antibiotic treatment, the mean value of somatic cell count decreased, compared to the CM treatments. The study showed that treating mastitis with CM would reduce the need for antibiotics, minimizing antibiotic resistance and preventing the need for costly and inefficient treatments. The mammary gland appears to be trying to repair itself, and, in this situation, using CM, which is high in growth factors, could facilitate the regenerative process [147]. Furthermore, animals with chronic mastitis are frequently culled from the herd rather than receiving treatment. In the future, this research could help restore milk production even in cows who would otherwise be forced to leave the production cycle, in addition to significantly reducing the use of antibiotics. The successful recovery of glandular tissue and the prevention of antibiotic residues in milk could be two additional financial benefits of using MSCs-CM [147].

3.3. Future Perspectives and Challenges in Clinical Applications

Compared with MSC therapy, the use of MSCs-EVs offers more advantages. In fact, MSCs-EVs are highly stable, suitable for long-term storage, and can induce intercellular communication by directly transferring functional proteins and miRNAs [158]. Furthermore, allogeneic administration of EVs does not result in an immune response, and their ability to avoid the possible carcinogenicity of MSCs is a major benefit [158].
Treatment of infectious disorders is another clinical area where potential EV therapy is growing, according to recent studies. Infectious diseases that were thought to be under control are becoming more severe and new ones are emerging [159]. The effectiveness of antibiotic treatment of these diseases is threatened by antibiotic resistance. In fact, worldwide, one of the most significant problems endangering the health of humans and animals is antibiotic resistance (AMR) [160]. Alternative therapeutic approaches to treat infections are becoming increasingly essential, as a result of the failure of regularly used treatment approaches and an increase in the number of outbreaks of serious infectious diseases [159]. Nowadays, the majority of research on infectious diseases has been conducted on rodents, including studies on sepsis, lung infections, wound infections, urinary tract infections and intestinal infections. Regenerative tissue, direct antibacterial effects, and immunomodulation are the therapeutic mechanisms [159]. Given the importance of reducing antibiotic therapy in order to decrease antibiotic resistance for public health, it is certain that numerous studies on domestic animals will also be carried out in the coming years.
Despite the encouraging results of preclinical and clinical studies of the application of MSCs-EVs in domestic animals, there are several challenges to overcome in order to achieve routine clinical application, such as large-scale production and precise isolation methods, taking into account and reducing factors that affect EV quality and quantity, finding rapid and accurate characterization of EVs, the precise content of the EV cargo, and the safety profile.

3.4. Large-Scale Production, Isolation Methods and Factors That Affect EV Quality/Quantity

Although MSCs are relatively easy to expand using conventional tissue flasks and bioreactors, their growth in culture is finite and their biological properties may become altered with repeated passage. There is an urgent need for development of methods for reliable expansion of MSCs to mass-produce EVs for clinical use. In fact, current methods of expansion of MSCs are labor-intensive and involve several procedures. In order to facilitate large-scale MSC-EV production, new batches of MSCs will have to be periodically derived, with significant impact on the costs [161]. To overcome this limitation different methods could be used, such as immortalization by natural selection or by genetic modification or clonal isolation [162,163]. A recent study in human medicine showed that hUC-MSC culture in scalable three-dimensional cultures resulted in a twenty-fold greater yield of EV than two-dimensional cultures [164]. In human medicine, several research groups have demonstrated that EVs isolated from MSCs culture by ultrafiltration followed by size-exclusion chromatography results in a higher yield while preserving EVs’ biophysical and functional properties [165,166,167,168].
The production of MSCs-EVs can be influenced by a number of factors, including cellular confluence, early vs. later cell passages, oxygen concentration, cytokines, and medium serum content [169]. A recent standard protocol for Good Manufacturing Practices (GMPs) offers a solution to produce MSCs and MSC-EVs on a large scale [170]. A similar procedure could be carried out with MSCs-EVs derived from horse and dog in veterinary medicine.

3.5. Safety Profile

Establishing a safety profile is another fundamental condition for clinical application. The greatest apprehension about the in vivo therapeutic use of cells is the differentiation of the transplanted MSCs and the potential of MSCs to suppress anti-tumor immune responses and to act as a progenitor for blood vessels, which potentially promote tumor growth and metastasis [171].

4. Conclusions

In the present review, we have summarized what EVs are, and that they can be readily isolated from MSCs derived from adult and fetal tissues. MSCs-EVs are known to have therapeutic benefits in different animal disease models and have theoretical advantages over intact MSCs as a therapeutic product, and in the future may be preferred over whole cells in the regenerative medicine field. For clinical application, several issues need to be addressed, such as large-scale production with standardization of EV isolation and characterization protocols.

Author Contributions

Conceptualization, E.I. and B.M.; methodology, A.L.; validation, A.L., E.I. and B.M.; investigation, A.L., E.I. and B.M.; data curation, A.L.; writing—original draft preparation, A.L., E.I. and B.M.; writing—review and editing, A.L., E.I. and B.M.; visualization, A.L., E.I. and B.M.; supervision, A.L., E.I. and B.M.; project administration, E.I. and B.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Lalu, M.M.; McIntyre, L.; Pugliese, C.; Fergusson, D.; Winston, B.W.; Marshall, J.C.; Granton, J.; Stewart, D.J.; Canadian Critical Care Trials Group. Safety of Cell Therapy with Mesenchymal Stromal Cells (SafeCell): A Systematic Review and Meta-Analysis of Clinical Trials. PLoS ONE 2012, 7, e47559. [Google Scholar] [CrossRef] [PubMed]
  2. Brooke, G.; Cook, M.; Blair, C.; Han, R.; Heazlewood, C.; Jones, B.; Kambouris, M.; Kollar, K.; McTaggart, S.; Pelekanos, R.; et al. Therapeutic Applications of Mesenchymal Stromal Cells. Semin. Cell Dev. Biol. 2007, 18, 846–858. [Google Scholar] [CrossRef]
  3. Iacono, E.; Pascucci, L.; Rossi, B.; Bazzucchi, C.; Lanci, A.; Ceccoli, M.; Merlo, B. Ultrastructural characteristics and immune profile of equine MSCs from fetal adnexa. Reproduction 2017, 154, 509–519. [Google Scholar] [CrossRef]
  4. Merlo, B.; Teti, G.; Lanci, A.; Burk, J.; Mazzotti, E.; Falconi, M.; Iacono, E. Comparison between Adult and Foetal Adnexa Derived Equine Post-Natal Mesenchymal Stem Cells. BMC Vet. Res. 2019, 15, 277. [Google Scholar] [CrossRef] [PubMed]
  5. Hofmann, N.A.; Ortner, A.; Jacamo, R.O.; Reinisch, A.; Schallmoser, K.; Rohban, R.; Etchart, N.; Fruehwirth, M.; Beham-Schmid, C.; Andreeff, M.; et al. Oxygen Sensing Mesenchymal Progenitors Promote Neo-Vasculogenesis in a Humanized Mouse Model In Vivo. PLoS ONE 2012, 7, e44468. [Google Scholar] [CrossRef]
  6. Bell, G.I.; Meschino, M.T.; Hughes-Large, J.M.; Broughton, H.C.; Xenocostas, A.; Hess, D.A. Combinatorial Human Progenitor Cell Transplantation Optimizes Islet Regeneration Through Secretion of Paracrine Factors. Stem Cells Dev. 2012, 21, 1863–1876. [Google Scholar] [CrossRef]
  7. Abumaree, M.H.; Abomaray, F.M.; Alshabibi, M.A.; AlAskar, A.S.; Kalionis, B. Immunomodulatory Properties of Human Placental Mesenchymal Stem/Stromal Cells. Placenta 2017, 59, 87–95. [Google Scholar] [CrossRef]
  8. Carrade, D.D.; Borjesson, D.L. Immunomodulation by Mesenchymal Stem Cells in Veterinary Species. Comp. Med. 2013, 63, 207–217. [Google Scholar] [PubMed]
  9. Le Blanc, K.; Davies, L.C. Mesenchymal Stromal Cells and the Innate Immune Response. Immunol. Lett. 2015, 168, 140–146. [Google Scholar] [CrossRef]
  10. Zhao, S.; Wehner, R.; Bornhäuser, M.; Wassmuth, R.; Bachmann, M.; Schmitz, M. Immunomodulatory Properties of Mesenchymal Stromal Cells and Their Therapeutic Consequences for Immune-Mediated Disorders. Stem Cells Dev. 2010, 19, 607–614. [Google Scholar] [CrossRef]
  11. Raposo, G.; Stoorvogel, W. Extracellular Vesicles: Exosomes, Microvesicles, and Friends. J. Cell Biol. 2013, 200, 373–383. [Google Scholar] [CrossRef] [PubMed]
  12. Yáñez-Mó, M.; Siljander, P.R.M.; Andreu, Z.; Bedina Zavec, A.; Borràs, F.E.; Buzas, E.I.; Buzas, K.; Casal, E.; Cappello, F.; Carvalho, J.; et al. Biological Properties of Extracellular Vesicles and Their Physiological Functions. J. Extracell. Vesicles 2015, 4, 27066. [Google Scholar] [CrossRef] [PubMed]
  13. George, J.N.; Thoi, L.L.; McManus, L.M.; Reimann, T.A. Isolation of Human Platelet Membrane Microparticles from Plasma and Serum. Blood 1982, 60, 834–840. [Google Scholar] [CrossRef]
  14. Ratajczak, J.; Wysoczynski, M.; Hayek, F.; Janowska-Wieczorek, A.; Ratajczak, M.Z. Membrane-Derived Microvesicles: Important and Underappreciated Mediators of Cell-to-Cell Communication. Leukemia 2006, 20, 1487–1495. [Google Scholar] [CrossRef] [PubMed]
  15. Akyurekli, C.; Le, Y.; Richardson, R.B.; Fergusson, D.; Tay, J.; Allan, D.S. A Systematic Review of Preclinical Studies on the Therapeutic Potential of Mesenchymal Stromal Cell-Derived Microvesicles. Stem Cell Rev. 2015, 11, 150–160. [Google Scholar] [CrossRef]
  16. Rani, S.; Ryan, A.E.; Griffin, M.D.; Ritter, T. Mesenchymal Stem Cell-Derived Extracellular Vesicles: Toward Cell-Free Therapeutic Applications. Mol. Ther. 2015, 23, 812–823. [Google Scholar] [CrossRef] [PubMed]
  17. Théry, C.; Witwer, K.W.; Aikawa, E.; Alcaraz, M.J.; Anderson, J.D.; Andriantsitohaina, R.; Antoniou, A.; Arab, T.; Archer, F.; Atkin-Smith, G.K.; et al. Minimal Information for Studies of Extracellular Vesicles 2018 (MISEV2018): A Position Statement of the International Society for Extracellular Vesicles and Update of the MISEV2014 Guidelines. J. Extracell. Vesicles 2018, 7, 1535750. [Google Scholar] [CrossRef] [PubMed]
  18. Lee, J.K.; Park, S.R.; Jung, B.K.; Jeon, Y.K.; Lee, Y.S.; Kim, M.K.; Kim, Y.G.; Jang, J.Y.; Kim, C.W. Exosomes Derived from Mesenchymal Stem Cells Suppress Angio-genesis by Down-Regulating VEGF Expression in Breast Cancer Cells. PLoS ONE 2013, 8, e84256. [Google Scholar] [CrossRef] [PubMed]
  19. Roccaro, A.M.; Sacco, A.; Maiso, P.; Azab, A.K.; Tai, Y.T.; Reagan, M.; Azab, F.; Flores, L.M.; Campigotto, F.; Weller, E.; et al. BM mesenchymal stromal cell–derived exosomes facilitate multiple myeloma progression. J. Clin. Investig. 2012, 123, 1542–1555. [Google Scholar] [CrossRef] [PubMed]
  20. Im, H.; Shao, H.; Park, Y.I.; Peterson, V.M.; Castro, C.M.; Weissleder, R.; Lee, H. Label-Free Detection and Molecular Profiling of Exosomes with a Nano-Plasmonic Sensor. Nat. Biotechnol. 2014, 32, 490–495. [Google Scholar] [CrossRef]
  21. Caplan, A.I.; Dennis, J.E. Mesenchymal Stem Cells as Trophic Mediators. J. Cell Biochem. 2006, 98, 1076–1084. [Google Scholar] [CrossRef] [PubMed]
  22. Ratajczak, M.Z. The Emerging Role of Microvesicles in Cellular Therapies for Organ/Tissue Regeneration. Nephrol. Dial. Transpl. 2011, 26, 1453–1456. [Google Scholar] [CrossRef] [PubMed]
  23. Pan, B.T.; Johnstone, R.M. Fate of the Transferrin Receptor during Maturation of Sheep Reticulocytes in Vitro: Selective Externalization of the Receptor. Cell 1983, 33, 967–978. [Google Scholar] [CrossRef] [PubMed]
  24. Wang, Y.; Wen, J.; Lu, T.; Han, W.; Jiao, K.; Li, H. Mesenchymal Stem Cell-Derived Extracellular Vesicles in Bone-Related Diseases: Intercellular Communication Messengers and Therapeutic Engineering Protagonists. Int. J. Nanomed. 2024, 19, 3233–3257. [Google Scholar] [CrossRef]
  25. Mathieu, M.; Martin-Jaular, L.; Lavieu, G.; Théry, C. Specificities of secretion and uptake of exosomes and other extracellular vesicles for cell-to- cell communication. Nat. Cell Biol. 2019, 21, 9–17. [Google Scholar] [CrossRef] [PubMed]
  26. van Niel, G.; Carter, D.R.F.; Clayton, A.; Lambert, D.W.; Raposo, G.; Vader, P. Challenges and directions in studying cell-cell communication by extracellular vesicles. Nat. Rev. Mol. Cell Biol. 2022, 23, 369–382. [Google Scholar] [CrossRef]
  27. Varderidou-Minasian, S.; Lorenowicz, M.J. Mesenchymal stromal/stem cell-derived extracellular vesicles in tissue repair: Challenges and opportunities. Theranostics 2020, 10, 5979–5997. [Google Scholar] [CrossRef]
  28. Sluijter, J.P.; Verhage, V.; Deddens, J.C.; van den Akker, F.; Doevendans, P.A. Microvesicles and exosomes for intracardiac communication. Cardiovasc. Res. 2014, 102, 302–311. [Google Scholar] [CrossRef] [PubMed]
  29. 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]
  30. Khoei, S.G.; Dermani, F.K.; Malih, S.; Fayazi, N.; Sheykhhasan, M. The use of mesenchymal stem cells and their derived extracellular vesicles in cardiovascular disease treatment. Curr. Stem Cell Res. Ther. 2020, 15, 623–638. [Google Scholar] [CrossRef]
  31. Bruno, S.; Grange, C.; Collino, F.; Deregibus, M.C.; Cantaluppi, V.; Biancone, L.; Tetta, C.; Camussi, G. Microvesicles derived from mesenchymal stem cells enhance survival in a lethal model of acute kidney injury. PLoS ONE 2012, 7, e33115. [Google Scholar] [CrossRef] [PubMed]
  32. Kilpinen, L.; Impola, U.; Sankkila, L.; Ritamo, I.; Aatonen, M.; Kilpinen, S.; Tuimala, J.; Valmu, L.; Levijoki, J.; Finckenberg, P. Extracellular membrane vesicles from umbilical cord blood-derived MSC protect against ischemic acute kidney injury, a feature that is lost after inflammatory conditioning. J. Extracell. Vesicles 2013, 2, 21927. [Google Scholar] [CrossRef] [PubMed]
  33. Sun, X.; Meng, H.; Wan, W.; Xie, M.; Wen, C. Application potential of stem/progenitor cell-derived extracellular vesicles in renal diseases. Curr. Stem Cell Res. Ther. 2019, 10, 8. [Google Scholar] [CrossRef] [PubMed]
  34. Mardpour, S.; Hassani, S.N.; Mardpour, S.; Sayahpour, F.; Vosough, M.; Ai, J.; Aghdami, N.; Hamidieh, A.A.; Baharvand, H. Extra-cellular vesicles derived from human embryonic stem cell-MSCs ameliorate cirrhosis in thioacetamide-induced chronic liver injury. J. Cell Physiol. 2018, 233, 9330–9344. [Google Scholar] [CrossRef] [PubMed]
  35. Wu, R.; Fan, X.; Wang, Y.; Shen, M.; Zheng, Y.; Zhao, S.; Yang, L. Mesenchymal Stem Cell-Derived Extracellular Vesicles in Liver immunity and therapy. Front. Immunol. 2022, 13, 833878. [Google Scholar] [CrossRef] [PubMed]
  36. Du, Y.M.; Zhuansun, Y.X.; Chen, R.; Lin, L.; Lin, Y.; Li, J.G. Mesenchymal stem cell exosomes promote immunosuppression of regulatory T cells in asthma. Exp. Cell Res. 2018, 363, 114–120. [Google Scholar] [CrossRef] [PubMed]
  37. Nataliya, B.; Mikhail, A.; Vladimir, P.; Olga, G.; Maksim, V.; Ivan, Z.; Novoseletskaya, E.; Sagaradze, G.; Danilova, N.; Malkov, P.; et al. Mesenchymal stromal cells facilitate resolution of pulmonary fibrosis by miR-29c and miR-129 intercellular transfer. Exp. Mol. Med. 2023, 55, 1399–1412. [Google Scholar] [CrossRef] [PubMed]
  38. Casado-Díaz, A.; Quesada-Gómez, J.M.; Dorado, G. Extracellular vesicles derived from mesenchymal stem cells (MSC) in regenerative medicine: Applications in skin wound healing. Front. Bioeng. Biotechnol. 2020, 8, 146. [Google Scholar] [CrossRef] [PubMed]
  39. Ha, D.H.; Kim, H.K.; Lee, J.; Kwon, H.H.; Park, G.H.; Yang, S.H.; Jung, J.Y.; Choi, H.; Lee, J.H.; Sung, S.; et al. Mesenchymal Stem/Stromal Cell-Derived Exosomes for Immunomodulatory Therapeutics and Skin Regeneration. Cells 2020, 9, 1157. [Google Scholar] [CrossRef]
  40. Kim, J.; Kim, E.H.; Lee, H.; Sung, J.H.; Bang, O.Y. Clinical-Scale Mesenchymal Stem Cell-Derived Extracellular Vesicle Therapy for Wound Healing. Int. J. Mol. Sci. 2023, 24, 4273. [Google Scholar] [CrossRef]
  41. Dong, L.; Pu, Y.; Zhang, L.; Qi, Q.; Xu, L.; Li, W.; Wei, C.; Wang, X.; Zhou, S.; Zhu, J.; et al. Human umbilical cord mesenchymal stem cell-derived extracellular vesicles promote lung adenocarcinoma growth by transferring miR-410. Cell Death Dis. 2018, 9, 218. [Google Scholar] [CrossRef] [PubMed]
  42. Zhao, Q.; Hai, B.; Kelly, J.; Wu, S.; Liu, F. Extracellular vesicle mimics made from iPS cell-derived mesenchymal stem cells improve the treatment of metastatic prostate cancer. Stem Cell Res. Ther. 2021, 12, 29. [Google Scholar] [CrossRef]
  43. Weng, Z.; Zhang, B.; Wu, C.; Yu, F.; Han, B.; Li, B.; Li, L. Therapeutic roles of mesenchymal stem cell-derived extracellular vesi-cles in cancer. J. Hematol. Oncol. 2021, 14, 136. [Google Scholar] [CrossRef] [PubMed]
  44. Mathew, B.; Torres, L.A.; Gamboa Acha, L.; Tran, S.; Liu, A.; Patel, R.; Chennakesavalu, M.; Aneesh, A.; Huang, C.C.; Feinstein, D.L.; et al. Uptake and distribution of administered bone marrow mesenchymal stem cell extracellular vesicles in retina. Cells 2021, 10, 730. [Google Scholar] [CrossRef] [PubMed]
  45. Reddy, S.K.; Ballal, A.R.; Shailaja, S.; Seetharam, R.N.; Raghu, C.H.; Sankhe, R.; Pai, K.; Tender, T.; Mathew, M.; Aroor, A.; et al. Small extracellular vesicle-loaded bevacizumab reduces the frequency of intravitreal injection required for diabetic retinopathy. Theranostics 2023, 13, 2241–2255. [Google Scholar] [CrossRef] [PubMed]
  46. Wang, M.; Yuan, Q.; Xie, L. Mesenchymal Stem Cell-Based Immunomodulation: Properties and Clinical Application. Stem Cells Int. 2018, 2018, 3057624. [Google Scholar] [CrossRef] [PubMed]
  47. Chen, Y.; Yu, Q.; Hu, Y.; Shi, Y. Current Research and Use of Mesenchymal Stem Cells in the Therapy of Autoimmune Diseases. Curr. Stem Cell Res. Ther. 2019, 14, 579–582. [Google Scholar] [CrossRef] [PubMed]
  48. Gómez-Ferrer, M.; Amaro-Prellezo, E.; Dorronsoro, A.; Sánchez-Sánchez, R.; Vicente, Á.; Cosín-Roger, J.; Barrachina, M.D.; Baquero, M.C.; Valencia, J.; Sepúlveda, P. HIF-Overexpression and Pro-Inflammatory Priming in Human Mesenchymal Stro-mal Cells Improves the Healing Properties of Extracellular Vesicles in Experimental Crohn’s Disease. Int. J. Mol. Sci. 2021, 22, 11269. [Google Scholar] [CrossRef]
  49. Madel, R.J.; Börger, V.; Dittrich, R.; Bremer, M.; Tertel, T.; Phuong, N.N.T.; Baba, H.A.; Kordelas, L.; Staubach, S.; Stein, F.; et al. Independent human mesenchymal stromal cell-derived extracellular vesicle preparations differentially attenuate symptoms in an advanced murine graft-versus-host disease model. Cytotherapy 2023, 25, 821–836. [Google Scholar] [CrossRef]
  50. Hackel, A.; Vollmer, S.; Bruderek, K.; Lang, S.; Brandau, S. Immunological priming of mesenchymal stromal/stem cells and their extracellular vesicles augments their therapeutic benefits in experimental graft-versus-host disease via engagement of PD-1 ligands. Front. Immunol. 2023, 14, 1078551. [Google Scholar] [CrossRef]
  51. van Velthoven, C.T.; Braccioli, L.; Willemen, H.L.; Kavelaars, A.; Heijnen, C.J. Therapeutic potential of genetically modified mesenchymal stem cells after neonatal hypoxic-ischemic brain damage. Mol. Ther. 2014, 22, 645–654. [Google Scholar] [CrossRef] [PubMed]
  52. Mao, Q.; Nguyen, P.D.; Shanti, R.M.; Shi, S.; Shakoori, P.; Zhang, Q.; Le, A.D. Gingiva-derived mesenchymal stem cell-extracellular vesicles activate schwann cell repair phenotype and promote nerve regeneration. Tissue Eng. Part A 2019, 25, 887–900. [Google Scholar] [CrossRef] [PubMed]
  53. Guy, R.; Herman, S.; Benyamini, H.; Ben-Zur, T.; Kobo, H.; Pasmanik-Chor, M.; Yaacobi, D.; Barel, E.; Yagil, C.; Yagil, Y.; et al. Mesenchymal Stem Cell-Derived Extracellular Vesicles as Proposed Therapy in a Rat Model of Cerebral Small Vessel Disease. Int. J. Mol. Sci. 2022, 23, 11211. [Google Scholar] [CrossRef] [PubMed]
  54. Yang, Y.; Yuan, L.; Cao, H.; Guo, J.; Zhou, X.; Zeng, Z. Application and Molecular Mechanisms of Extracellular Vesicles De-rived from Mesenchymal Stem Cells in Osteoporosis. Curr. Issues Mol. Biol. 2022, 44, 6346–6367. [Google Scholar] [CrossRef] [PubMed]
  55. Yin, B.; Ni, J.; Witherel, C.E.; Yang, M.; Burdick, J.A.; Wen, C.; Wong, S.H.D. Harnessing Tissue-derived Extracellular Vesicles for Osteoarthritis theranostics. Theranostics 2022, 12, 207–231. [Google Scholar] [CrossRef]
  56. Zou, J.; Yang, W.; Cui, W.; Li, C.; Ma, C.; Ji, X.; Hong, J.; Qu, Z.; Chen, J.; Liu, A.; et al. Therapeutic potential and mechanisms of mesenchymal stem cell-derived exosomes as bioactive materials in tendon-bone healing. J. Nanobiotechnol. 2023, 21, 14. [Google Scholar] [CrossRef] [PubMed]
  57. Lötvall, J.; Hill, A.F.; Hochberg, F.; Buzás, E.I.; Di Vizio, D.; Gardiner, C.; Gho, Y.S.; Kurochkin, I.V.; Mathivanan, S.; Quesenberry, P.; et al. Minimal experimental requirements for definition of extracellular vesicles and their functions (MISEV2014): A position statement from the International Society for Extracellular Vesicles. J. Extracell. Vesicles 2014, 3, 26913. [Google Scholar] [CrossRef] [PubMed]
  58. Moccia, V.; Sammarco, A.; Cavicchioli, L.; Castagnaro, M.; Bongiovanni, L.; Zappulli, V. Extracellular Vesicles in Veterinary Medicine. Animals 2022, 12, 2716. [Google Scholar] [CrossRef] [PubMed]
  59. Arévalo-Turrubiarte, M.; Baratta, M.; Ponti, G.; Chiaradia, E.; Martignani, E. Extracellular vesicles from equine mesenchymal stem cells decrease inflammation markers in chondrocytes in vitro. Equine Vet. J. 2022, 54, 1133–1143. [Google Scholar] [CrossRef]
  60. Navarrete, F.; Wong, Y.S.; Cabezas, J.; Riadi, G.; Manríquez, J.; Rojas, D.; Furlanetto Mancanares, A.C.; Rodriguez-Alvarez, L.; Saravia, F.; Castro, F.O. Distinctive cellular transcriptomic signature and MicroRNA cargo of extracellular vesicles of horse adipose and endometrial mesenchymal stem cells from the same donors. Cell Reprogramming 2020, 22, 311–327. [Google Scholar] [CrossRef]
  61. Ji, Y.; Jiang, W.; Zeng, F.; Zou, D.; Li, S.; Zhang, X.; Zhu, Q.; Liang, Q.; Li, M.; Li, D. Characterization of Canine Gingival-Derived Mesenchymal Stem Cells and Their Exosomes. J. Vet. Dent. 2023. [Google Scholar] [CrossRef] [PubMed]
  62. Pipino, C.; Mandatori, D.; Buccella, F.; Lanuti, P.; Preziuso, A.; Castellani, F.; Grotta, L.; Di Tomo, P.; Marchetti, S.; Di Pietro, N.; et al. Identification and characterization of a stem cell-like population in bovine milk: A potential new source for regenerative medicine in veterinary. Stem Cells Dev. 2018, 27, 1587–1597. [Google Scholar] [CrossRef] [PubMed]
  63. Eirin, A.; Riester, S.M.; Zhu, X.Y.; Tang, H.; Evans, J.M.; O’Brien, D.; van Wijnen, A.J.; Lerman, L.O. MicroRNA and mRNA cargo of extracellular vesicles from porcine adipose tissue-derived mesenchymal stem cells. Gene 2014, 551, 55–64. [Google Scholar] [CrossRef]
  64. Chen, K.H.; Chen, C.H.; Wallace, C.G.; Yuen, C.M.; Kao, G.S.; Chen, Y.L.; Shao, P.L.; Chen, Y.L.; Chai, H.T.; Lin, K.C.; et al. Intravenous administration of xenogenic adipose-derived mesenchymal stem cells (ADMSC) and ADMSC-derived exosomes markedly reduced brain infarct volume and preserved neurological function in rat after acute ischemic stroke. Oncotarget 2016, 7, 74537. [Google Scholar] [CrossRef] [PubMed]
  65. Eirin, A.; Zhu, X.Y.; Puranik, A.S.; Woollard, J.R.; Tang, H.; Dasari, S.; Lerman, A.; van Wijnen, A.J.; Lerman, L.O. Comparative proteomic analysis of extracellular vesicles isolated from porcine adipose tissue-derived mesenchymal stem/stromal cells. Sci. Rep. 2016, 6, 36120. [Google Scholar] [CrossRef]
  66. Eirin, A.; Zhu, X.Y.; Puranik, A.S.; Woollard, J.R.; Tang, H.; Dasari, S.; Lermna, A.; van Wijnen, A.J.; Lerman, L.O. Comparative Integrated transcriptomic and proteomic analysis of the molecular cargo of extracellular vesicles derived from porcine adipose tissue-derived mesenchymal stem cells. PLoS ONE 2017, 12, e0174303. [Google Scholar] [CrossRef]
  67. Eirin, A.; Zhu, X.Y.; Puranik, A.S.; Tang, H.; McGurren, K.A.; van Wijnen, A.J.; Lerman, A.; Lerman, L.O. Mesenchymal stem cell–derived extracellular vesicles attenuate kidney inflammation. Kidney Int. 2017, 92, 114–124. [Google Scholar] [CrossRef]
  68. Eirin, A.; Zhu, X.Y.; Jonnada, S.; Lerman, A.; van Wijnen, A.J.; Lerman, L.O. Mesenchymal stem cell-derived extracellular vesicles improve the renal microvasculature in metabolic renovascular disease in swine. Cell Transplant. 2018, 27, 1080–1095. [Google Scholar] [CrossRef]
  69. Meng, Y.; Eirin, A.; Zhu, X.Y.; O’Brien, D.R.; Lerman, A.; Van Wijnen, A.J.; Lerman, L.O. The metabolic syndrome modifies the mRNA expression profile of extracellular vesicles derived from porcine mesenchymal stem cells. Diabetol. Metab. Syndr. 2018, 10, 58. [Google Scholar] [CrossRef]
  70. Khatri, M.; Richardson, L.A.; Meulia, T. Mesenchymal stem cell-derived extracellular vesicles attenuate influenza virus-induced acute lung injury in a pig model. Stem Cell Res. Ther. 2018, 9, 17. [Google Scholar] [CrossRef]
  71. Meng, Y.; Eirin, A.; Zhu, X.Y.; Tang, H.; Chanana, P.; Lerman, A.; Van Wijnen, A.J.; Lerman, L.O. The metabolic syndrome alters the miRNA signature of porcine adipose tissue-derived mesenchymal stem cells. Cytom. Part A 2018, 93, 93–103. [Google Scholar] [CrossRef] [PubMed]
  72. Eirin, A.; Zhu, X.Y.; Woollard, J.R.; Tang, H.; Dasari, S.; Lerman, A.; Lerman, L.O. Metabolic syndrome interferes with packaging of proteins within porcine mesenchymal stem cell-derived extracellular vesicles. Stem Cells Transl. Med. 2019, 8, 430–440. [Google Scholar] [CrossRef] [PubMed]
  73. Conley, S.M.; Shook, J.E.; Zhu, X.Y.; Eirin, A.; Jordan, K.L.; Woollard, J.R.; Isik, B.; Hickson, L.J.; Puranik, A.S.; Lerman, L.O. Metabolic syndrome induces release of smaller extracellular vesicles from porcine mesenchymal stem cells. Cell Transplant. 2019, 28, 1271–1278. [Google Scholar] [CrossRef] [PubMed]
  74. Thankam, F.G.; Chandra, I.; Diaz, C.; Dilisio, M.F.; Fleegel, J.; Gross, R.M.; Agrawal, D.K. Matrix regeneration proteins in the hypoxia-triggered exosomes of shoulder tenocytes and adipose-derived mesenchymal stem cells. Mol. Cell Biochem. 2020, 465, 75–87. [Google Scholar] [CrossRef]
  75. Zhang, L.; Zhu, X.Y.; Zhao, Y.; Eirin, A.; Liu, L.; Ferguson, C.M.; Tang, H.; Lerman, A.; Lerman, L.O. Selective intrarenal delivery of mesenchymal stem cell-derived extracellular vesicles attenuates myocardial injury in experimental metabolic renovascular disease. Basic Res. Cardiol. 2020, 115, 16. [Google Scholar] [CrossRef]
  76. Pawar, A.S.; Eirin, A.; Tang, H.; Zhu, X.Y.; Lerman, A.; Lerman, L.O. Upregulated tumor necrosis factor-α transcriptome and proteome in adipose tissue-derived mesenchymal stem cells from pigs with metabolic syndrome. Cytokine 2020, 130, 155080. [Google Scholar] [CrossRef] [PubMed]
  77. Zhao, Y.; Zhu, X.; Zhang, L.; Ferguson, C.M.; Song, T.; Jiang, K.; Conley, S.M.; Krier, J.D.; Tang, H.; Saadiq, I.; et al. Mesenchymal stem/stromal cells and their extracellular vesicle progeny decrease injury in poststenotic swine kidney through different mechanisms. Stem Cells Dev. 2020, 29, 1190–1200. [Google Scholar] [CrossRef]
  78. Song, T.; Eirin, A.; Zhu, X.; Zhao, Y.; Krier, J.D.; Tang, H.; Jordan, K.L.; Woollard, J.R.; Taner, T.; Lerman, A.; et al. Mesenchymal stem cell–derived extracellular vesicles induce regulatory t cells to ameliorate chronic kidney injury. Hypertension 2020, 75, 1223–1232. [Google Scholar] [CrossRef]
  79. Li, Y.; Meng, Y.; Zhu, X.; Saadiq, I.M.; Jordan, K.L.; Eirin, A.; Lerman, L.O. Metabolic syndrome increases senescence-associated micro-RNAs in extracellular vesicles derived from swine and human mesenchymal stem/stromal cells. Cell Commun. Signal. 2020, 18, 124. [Google Scholar] [CrossRef]
  80. Farahani, R.A.; Zhu, X.Y.; Tang, H.; Jordan, K.L.; Lerman, A.; Lerman, L.O.; Eirin, A. Metabolic syndrome alters the cargo of mitochondria-related microRNAs in swine mesenchymal stem cell-derived extracellular vesicles, impairing their capacity to repair the stenotic kidney. Stem Cells Int. 2020, 2020, 8845635. [Google Scholar] [CrossRef]
  81. Monguió-Tortajada, M.; Prat-Vidal, C.; Moron-Font, M.; Clos-Sansalvador, M.; Calle, A.; Gastelurrutia, P.; Cserkoova, A.; Morancho, A.; Ramírez, M.A.; Rosell, A.; et al. Local administration of porcine immunomodulatory, chemotactic and angiogenic extracellular vesicles using engineered cardiac scaffolds for myocardial infarction. Bioact. Mater. 2021, 6, 3314–3327. [Google Scholar] [CrossRef] [PubMed]
  82. Ferguson, C.M.; Farahani, R.A.; Zhu, X.Y.; Tang, H.; Jordan, K.L.; Saadiq, I.M.; Lerman, A.; Lerman, L.O.; Eirin, A. Mesenchymal stem/stromal cell-derived extracellular vesicles elicit better preservation of the intra-renal microvasculature than renal revascularization in pigs with renovascular disease. Cells 2021, 10, 763. [Google Scholar] [CrossRef] [PubMed]
  83. Jiang, Y.; Hong, S.; Zhu, X.; Zhang, L.; Tang, H.; Jordan, K.L.; Saadiq, I.M.; Huang, W.; Lerman, A.; Eirin, A.; et al. IL-10 partly mediates the ability of MSC-derived extracellular vesicles to attenuate myocardial damage in experimental metabolic renovascular hypertension. Front. Immunol. 2022, 13, 940093. [Google Scholar] [CrossRef] [PubMed]
  84. Monguió-Tortajada, M.; Prat-Vidal, C.; Martínez-Falguera, D.; Teis, A.; Soler-Botija, C.; Courageux, Y.; Munizaga-Larroudé, M.; Moron-Font, M.; Bayes-Genis, A.; Borràs, F.E.; et al. Acellular cardiac scaffolds enriched with MSC-derived extracellular vesicles limit ventricular remodelling and exert local and systemic immunomodulation in a myocardial infarction porcine model. Theranostics 2022, 12, 4656. [Google Scholar] [CrossRef] [PubMed]
  85. Jiang, W.; Cui, Y.; Li, X.; Xiong, Y.; Yue, G.; Yang, Y. Isolation and Identification of Porcine Bone Marrow Mesenchymal Stem Cells and their Derived Extracellular Vesicles. JoVE 2022, 182, e63785. [Google Scholar] [CrossRef] [PubMed]
  86. Hong, S.; Zhu, X.Y.; Jiang, Y.; Zhang, L.; Tang, H.; Jordan, K.L.; Saadiq, I.M.; Huang, W.; Lerman, A.; Eirin, A.; et al. Autologous Extracellular Vesicles attenuate cardiac injury in experimental atherosclerotic renovascular disease more effectively than their parent Mesenchymal Stem/Stromal Cells. Stem Cell Rev. Rep. 2023, 19, 700–712. [Google Scholar] [CrossRef] [PubMed]
  87. Wang, Y.; Piao, C.; Liu, T.; Lu, X.; Ma, Y.; Zhang, J.; Liu, G.; Wang, H. Effects of the exosomes of adipose-derived mesenchymal stem cells on apoptosis and pyroptosis of injured liver in miniature pigs. Biomed. Pharmacother. 2023, 169, 115873. [Google Scholar] [CrossRef] [PubMed]
  88. Wang, Y.; Liu, T.; Jiao, G.; Lv, Y.; Piao, C.; Lu, X.; Ma, H.; Wang, H. Exosomes from adipose-derived mesenchymal stem cells can attenuate liver injury caused by minimally invasive hemihepatectomy combined with ischemia-reperfusion in minipigs by modulating the endoplasmic reticulum stress response. Life Sci. 2023, 321, 121618. [Google Scholar] [CrossRef] [PubMed]
  89. Shulman, I.; Ageeva, T.; Kostennikov, A.; Ogurcov, S.; Tazetdinova, L.; Kabdesh, I.; Rogozhin, A.; Ganiev, I.; Rizvanov, I.; Mukhamedshina, Y. Intrathecal Injection of Autologous Mesenchymal Stem-Cell-Derived Extracellular Vesicles in Spinal Cord Injury: A Feasibility Study in Pigs. Int. J. Mol. Sci. 2023, 24, 8240. [Google Scholar] [CrossRef]
  90. Aggarwal, R.; Shao, A.; Potel, K.N.; So, S.W.; Swingen, C.M.; Wright, C.A.; Hocum Stone, L.L.; McFalls, E.O.; Butterick, T.A.; Kelly, R.F. Stem cell-derived exosome patch with coronary artery bypass graft restores cardiac function in chronically ischemic porcine myocardium. J. Thorac. Cardiovasc. Surg. 2023, 166, e512–e530. [Google Scholar] [CrossRef]
  91. Pascucci, L.; Alessandri, G.; Dall’Aglio, C.; Mercati, F.; Coliolo, P.; Bazzucchi, C.; Dante, S.; Petrini, S.; Curina, G.; Ceccarelli, P. Membrane vesicles mediate pro-angiogenic activity of equine adipose-derived mesenchymal stromal cells. Vet. J. 2014, 202, 361–366. [Google Scholar] [CrossRef] [PubMed]
  92. Pascucci, L.; Dall’Aglio, C.; Bazzucchi, C.; Mercati, F.; Mancini, M.G.; Pessina, A.; Alessandri, G.; Giammarioli, M.; Dante, S.; Brunati, G.; et al. Horse adipose-derived mesenchymal stromal cells constitutively produce membrane vesicles: A morphological study. Histol. Histopathol. 2015, 30, 549–557. [Google Scholar] [PubMed]
  93. Marycz, K.; Michalak, I.; Kocherova, I.; Marędziak, M.; Weiss, C. The Cladophora glomerata enriched by biosorption process in Cr (III) improves viability, and reduces oxidative stress and apoptosis in equine metabolic syndrome derived adipose mesenchymal stromal stem cells (ASCs) and their extracellular vesicles (MV’s). Mar. Drugs 2017, 15, 385. [Google Scholar] [CrossRef] [PubMed]
  94. Capomaccio, S.; Cappelli, K.; Bazzucchi, C.; Coletti, M.; Gialletti, R.; Moriconi, F.; Passamonti, F.; Pepe, M.; Petrini, S.; Mecocci, S.; et al. Equine adipose-derived mesenchymal stromal cells release extracellular vesicles enclosing different subsets of small RNAs. Stem Cells Int. 2019, 2019, 4957806. [Google Scholar] [CrossRef] [PubMed]
  95. Klymiuk, M.C.; Balz, N.; Elashry, M.I.; Heimann, M.; Wenisch, S.; Arnhold, S. Exosomes isolation and identification from equine mesenchymal stem cells. BMC Vet. Res. 2019, 15, 42. [Google Scholar] [CrossRef] [PubMed]
  96. Kornicka-Garbowska, K.; Pędziwiatr, R.; Woźniak, P.; Kucharczyk, K.; Marycz, K. Microvesicles isolated from 5-azacytidine-and-resveratrol-treated mesenchymal stem cells for the treatment of suspensory ligament injury in horse—A case report. Stem Cell Res. Ther. 2019, 10, 394. [Google Scholar] [CrossRef] [PubMed]
  97. Hotham, W.E.; Thompson, C.; Szu-Ting, L.; Henson, F.M.D. The anti-inflammatory effects of equine bone marrow stem cell-derived extracellular vesicles on autologous chondrocytes. Vet. Rec. 2021, 8, e22. [Google Scholar] [CrossRef] [PubMed]
  98. Mocchi, M.; Grolli, S.; Dotti, S.; Di Silvestre, D.; Villa, R.; Berni, P.; Conti, V.; Passignani, G.; Brambilla, F.; Del Bue, M.; et al. Equine mesenchymal stem/stromal cells freeze-dried secretome (Lyosecretome) for the treatment of musculoskeletal diseases: Production process validation and batch release test for clinical use. Pharmaceuticals 2021, 14, 553. [Google Scholar] [CrossRef]
  99. Contentin, R.; Jammes, M.; Bourdon, B.; Cassé, F.; Bianchi, A.; Audigié, F.; Branly, T.; Velot, E.; Galéra, P. Bone marrow MSC secretome increases equine articular chondrocyte collagen accumulation and their migratory capacities. Int. J. Mol. Sci. 2022, 23, 5795. [Google Scholar] [CrossRef]
  100. Soukup, R.; Gerner, I.; Gültekin, S.; Baik, H.; Oesterreicher, J.; Grillari, J.; Jenner, F. Characterisation of extracellular vesicles from equine mesenchymal stem cells. Int. J. Mol. Sci. 2022, 23, 5858. [Google Scholar] [CrossRef]
  101. Clarke, E.J.; Johnson, E.; Caamaño Gutierrez, E.; Andersen, C.; Berg, L.C.; Jenkins, R.E.; Lindegaard, C.; Uvebrant, K.; Lundgren-Åkerlund, E.; Turlo, A.; et al. Temporal extracellular vesicle protein changes following intraarticular treatment with integrin α10β1-selected mesenchymal stem cells in equine osteoarthritis. Front. Vet. Sci. 2022, 9, 1057667. [Google Scholar] [CrossRef] [PubMed]
  102. Soukup, R.; Gerner, I.; Mohr, T.; Gueltekin, S.; Grillari, J.; Jenner, F. Mesenchymal Stem Cell Conditioned Medium Modulates Inflammation in Tenocytes: Complete Conditioned Medium Has Superior Therapeutic Efficacy than Its Extracellular Vesicle Fraction. Int. J. Mol. Sci. 2023, 24, 10857. [Google Scholar] [CrossRef] [PubMed]
  103. Abdelnaby, E.A.; Abdallah, A.N.; Anwar, I.M.; El-Tookhy, O.S.; Shamaa, A.A. The therapeutic effect of stem cell-derived exosomes in the treatment of chronic endometritis as assessed by histopathological, Doppler and hormonal expression in Arabian mares. Equine Vet. Educ. 2023, 36, 347–356. [Google Scholar] [CrossRef]
  104. Cassé, F.; Velot, E.; Bianchi, A.; Audigié, F.; Contentin, R.; Galéra, P. Pro-Inflammatory Cytokine Priming and Purification Method Modulate the Impact of Exosomes Derived from Equine Bone Marrow Mesenchymal Stromal Cells on Equine Articular Chondrocytes. Int. J. Mol. Sci. 2023, 24, 14169. [Google Scholar] [CrossRef] [PubMed]
  105. El-Tookhy, O.S.; Shamaa, A.A.; Shehab, G.G.; Abdallah, A.N.; Azzam, O.M. Histological evaluation of experimentally induced critical size defect skin wounds using exosomal solution of mesenchymal stem cells derived microvesicles. Int. J. Stem Cells 2017, 10, 144–153. [Google Scholar] [CrossRef]
  106. Qamar, A.Y.; Fang, X.; Kim, M.J.; Cho, J. Improved post-thaw quality of canine semen after treatment with exosomes from conditioned medium of adipose-derived mesenchymal stem cells. Animals 2019, 9, 865. [Google Scholar] [CrossRef]
  107. Villatoro, A.J.; Alcoholado, C.; Martín-Astorga, M.C.; 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]
  108. An, J.H.; Li, Q.; Bhang, D.H.; Song, W.J.; Youn, H.Y. TNF-α and INF-γ primed canine stem cell-derived extracellular vesicles alleviate experimental murine colitis. Sci. Rep. 2020, 10, 2115. [Google Scholar] [CrossRef]
  109. An, J.H.; Li, Q.; Ryu, M.O.; Nam, A.R.; Bhang, D.H.; Jung, Y.C.; Song, W.J.; Youn, H.Y. TSG-6 in extracellular vesicles from canine mesenchymal stem/stromal is a major factor in relieving DSS-induced colitis. PLoS ONE 2020, 15, e0220756. [Google Scholar] [CrossRef]
  110. Park, S.M.; An, J.H.; Lee, J.H.; Kim, K.B.; Chae, H.K.; Oh, Y.I.; Song, W.J.; Youn, H.Y. Extracellular vesicles derived from DFO-preconditioned canine AT-MSCs reprogram macrophages into M2 phase. PLoS ONE 2021, 16, e0254657. [Google Scholar] [CrossRef]
  111. Teshima, T.; Yuchi, Y.; Suzuki, R.; Matsumoto, H.; Koyama, H. Immunomodulatory effects of canine adipose tissue mesenchymal stem cell-derived extracellular vesicles on stimulated CD4+ T cells isolated from peripheral blood mononuclear cells. J. Immunol. Res. 2021, 2021, 2993043. [Google Scholar] [CrossRef]
  112. Mocchi, M.; Bari, E.; Dotti, S.; Villa, R.; Berni, P.; Conti, V.; Del Bue, M.; Squassino, G.P.; Segale, L.; Ramoni, R.; et al. Canine mesenchymal cell lyosecretome production and safety evaluation after allogenic intraarticular injection in osteoarthritic dogs. Animals 2021, 11, 3271. [Google Scholar] [CrossRef]
  113. Kuwahara, Y.; Yoshizaki, K.; Nishida, H.; Kamishina, H.; Maeda, S.; Takano, K.; Fujita, N.; Nishimura, R.; Jo, J.I.; Tabata, Y.; et al. Extracellular vesicles derived from canine mesenchymal stromal cells in serum free culture medium have anti-inflammatory effect on microglial cells. Front. Vet. Sci. 2021, 8, 633426. [Google Scholar] [CrossRef]
  114. Kim, S.Y.; Yoon, T.H.; Na, J.; Yi, S.J.; Jin, Y.; Kim, M.; Oh, T.H.; Chung, T.W. Mesenchymal stem cells and extracellular vesicles derived from canine adipose tissue ameliorates inflammation, skin barrier function and pruritus by reducing JAK/STAT signaling in atopic dermatitis. Int. J. Mol. Sci. 2022, 23, 4868. [Google Scholar] [CrossRef]
  115. Cho, B.S.; Kim, S.B.; Kim, S.; Rhee, B.; Yoon, J.; Lee, J.W. Canine Mesenchymal-Stem-Cell-Derived Extracellular Vesicles Attenuate Atopic Dermatitis. Animals 2023, 13, 2215. [Google Scholar] [CrossRef]
  116. Liu, H.; Huang, L.; Chen, F.; Zhong, Z.; Ma, X.; Zhou, Z.; Cao, S.; Shen, L.; Peng, G. Adipose-derived mesenchymal stem cells secrete extracellular vesicles: A potential cell-free therapy for canine renal ischaemia-reperfusion injury. Vet. Med. Sci. 2023, 9, 1134–1142. [Google Scholar] [CrossRef]
  117. Yoshizaki, K.; Nishida, H.; Tabata, Y.; Jo, J.I.; Nakase, I.; Akiyoshi, H. Controlled release of canine MSC-derived extracellular vesicles by cationized gelatin hydrogels. Regen. Ther. 2023, 22, 1–6. [Google Scholar] [CrossRef]
  118. Sung, S.E.; Seo, M.S.; Kang, K.K.; Choi, J.H.; Lee, S.; Sung, M.; Kim, K.; Lee, G.W.; Lim, J.H.; Yang, S.Y.; et al. Mesenchymal stem cell exosomes derived from feline adipose tissue enhance the effects of anti-inflammation compared to fibroblasts-derived exosomes. Vet. Sci. 2021, 8, 182. [Google Scholar] [CrossRef]
  119. Villatoro, A.J.; Martín-Astorga, M.D.C.; Alcoholado, C.; Cárdenas, C.; Fariñas, F.; Becerra, J.; Visser, R. Altered proteomic profile of adipose tissue-derived mesenchymal stem cell exosomes from cats with severe chronic gingivostomatitis. Animals 2021, 11, 2466. [Google Scholar] [CrossRef]
  120. Villatoro, A.J.; Martín-Astorga, M.D.C.; Alcoholado, C.; Sánchez-Martín, M.D.M.; Becerra, J. Proteomic analysis of the secretome and exosomes of feline adipose-derived mesenchymal stem cells. Animals 2021, 11, 295. [Google Scholar] [CrossRef]
  121. Li, D.; Luo, H.; Ruan, H.; Chen, Z.; Chen, S.; Wang, B.; Xie, Y. Isolation and identification of exosomes from feline plasma, urine and adipose-derived mesenchymal stem cells. BMC Vet. Res. 2021, 17, 272. [Google Scholar] [CrossRef]
  122. Li, W.; Wang, W.; He, X.; Liao, Z.; Aierken, A.; Hua, J.; Wang, Y.; Lu, D.; Zhang, S. Rapid recovery of male cats with postrenal acute kidney injury by treating with allogeneic adipose mesenchymal stem cell-derived extracellular vesicles. Stem Cell Res. Ther. 2022, 13, 379. [Google Scholar] [CrossRef]
  123. von Stade, D.; Meyers, M.; Johnson, J.; Schlegel, T.T.; Romeo, A.; Regan, D.; McGilvray, K. Exosome Cell Origin Affects In Vitro Markers of Tendon Repair in Ovine Macrophages and Tenocytes. Tissue Eng. Part A 2023, 29, 282–291. [Google Scholar] [CrossRef]
  124. De Coppiz, P.; Bartsch, G.J.; Siddiqui, M.M.; Xu, T.; Santos, C.C.; Perin, L.; Mostoslavsky, G.; Serre, A.C.; Snyder, E.Y.; Yoo, J.J.; et al. Isolation of amniotic stem cell lines with potential for therapy. Nat. Biotechnol. 2007, 25, 100–106. [Google Scholar] [CrossRef]
  125. Delo, D.M.; De Coppi, P.; Bartsch, G.J.; Atala, A. Amniotic fluid e placental stem cells. Methods Enzymol. 2006, 419, 426–438. [Google Scholar]
  126. Gucciardo, L.; Lories, R.; Ochsenbein-Kolble, N.; Done, E.; Zwijsen, A.; Depresta, J. Fetal mesenchymal stem cells: Isolation, properties e potential use in perinatology e regenerative medicine. BJOG 2009, 116, 166–172. [Google Scholar] [CrossRef]
  127. In’t Anker, P.S.; Scherjon, S.A.; Kleijburg-van der Keur, C.; De Groot Swings, G.M.; Claas, F.H.; Fibbe, W.E.; Kanhai, H.H.H. Isolation of mesenchymal stem cells of fetal or maternal origin from human placenta. Stem Cells 2004, 22, 1338–1345. [Google Scholar] [CrossRef]
  128. Kern, S.; Eichler, H.; Stoeve, J.; Kluter, H.; Bieback, K. Comparative analysis of mesenchymal stem cells from bone marrow, umbilical cord blood, or adipose tissue. Stem Cells 2006, 24, 1294–1301. [Google Scholar] [CrossRef]
  129. Kögler, G.; Sensken, S.; Airey, J.A.; Trapp, T.; Mschen, M.; Feldhahn, N.; Liedtke, S.; Sorg, R.V.; Fischer, J.; Rosenbaum, C.; et al. New human somatic stem cell from placental cord blood with intrinsic pluripotent differentiation potential. J. Exp. Med. 2004, 200, 123–135. [Google Scholar] [CrossRef]
  130. Lange-Consiglio, A.; Rossi, D.; Tassan, S.; Perego, R.; Cremonesi, F.; Parolini, O. Conditioned medium from horse amniotic membrane-derived multipotent progenitor cells: Immunomodulatory activity in vitro and first clinical application in tendon and ligament injuries in vivo. Stem Cells Dev. 2013, 22, 3015–3024. [Google Scholar] [CrossRef] [PubMed]
  131. Moore, K.L.; Persaud, T.V.N. The developing human: Clinically oriented embryology. 1998, Sanders.
  132. Evangelista, M.; Soncini, M.; Parolini, O. Placenta-derived stem cells: New hope for cell therapy? Cytotechnology 2008, 58, 33–42. [Google Scholar] [CrossRef] [PubMed]
  133. Zucca, E.; Corsini, E.; Galbiati, V.; Lange-Consiglio, A.; Ferrucci, F. Evaluation of amniotic mesenchymal cell derivatives on cytokine production in equine alveolar macrophages: An in vitro approach to lung inflammation. Stem Cell Res. Ther. 2016, 7, 137. [Google Scholar] [CrossRef] [PubMed]
  134. Perrini, C.; Strillacci, M.G.; Bagnato, A.; Esposti, P.; Marini, M.G.; Corradetti, B.; Bizzaro, D.; Idda, A.; Ledda, S.; Capra, E.; et al. Microvesicles secreted from equine amniotic-derived cells and their potential role in reducing inflammation in endometrial cells in an in-vitro model. Stem Cell Res. Ther. 2016, 7, 169. [Google Scholar] [CrossRef]
  135. Lange-Consiglio, A.; Lazzari, B.; Perrini, C.; Pizzi, F.; Stella, A.; Cremonesi, F.; Capra, E. MicroRNAs of equine amniotic mesenchymal cell-derived microvesicles and their involvement in anti-inflammatory processes. Cell Transplant. 2018, 27, 45–54. [Google Scholar] [CrossRef] [PubMed]
  136. Lange-Consiglio, A.; Perrini, C.; Tasquier, R.; Deregibus, M.C.; Camussi, G.; Pascucci, L.; Marini, M.G.; Corradetti, B.; Bizzaro, D.; De Vita, B.; et al. Equine amniotic microvesicles and their anti-inflammatory potential in a tenocyte model in vitro. Stem Cells Dev. 2016, 25, 610–621. [Google Scholar] [CrossRef] [PubMed]
  137. Desantis, S.; Accogli, G.; Albrizio, M.; Rossi, R.; Cremonesi, F.; Lange Consiglio, A. Glycan profiling analysis of Equine amniotic progenitor mesenchymal cells and their derived extracellular microvesicles. Stem Cells Dev. 2019, 28, 812–821. [Google Scholar] [CrossRef] [PubMed]
  138. Crain, S.K.; Robinson, S.R.; Thane, K.E.; Davis, A.M.; Meola, D.M.; Barton, B.A.; Yang, V.K.; Hoffman, A.M. Extracellular vesicles from Wharton’s Jelly mesenchymal stem cells suppress CD4 expressing T Cells through Transforming Growth Factor Beta and Adenosine Signaling in a canine model. Stem Cells Dev. 2019, 28, 212–226. [Google Scholar] [CrossRef] [PubMed]
  139. Wright, A.; Snyder, O.L.; He, H.; Christenson, L.K.; Fleming, S.; Weiss, M.L. Procoagulant Activity of Umbilical Cord-Derived Mesenchymal Stromal Cells’ Extracellular Vesicles (MSC-EVs). Int. J. Mol. Sci. 2023, 24, 9216. [Google Scholar] [CrossRef] [PubMed]
  140. Karam, R.G.; Motta, L.C.B.; de Almeida, M.F.; Bridi, A.; da Silveira, J.C.; Ambrósio, C.E. Secretion pattern of canine amniotic stem cells derived extracellular vesicles. Anim. Reprod. 2022, 19, e20220063. [Google Scholar] [CrossRef]
  141. Scassiotti, R.F.; de Paula Coutinho, M.; Pinto Santos, S.I.; Ferreira Pinto, P.A.; Ferreira de Almeida, M.; Karam, R.G.; da Silva Rosa, P.M.; Dos Santos Martins, D.; Coelho da Silveira, J.; Ambrósio, C.E. Adipose and amnion-derived mesenchymal stem cells: Extracellular vesicles characterization and implication for reproductive biotechnology. Theriogenology 2023, 198, 264–272. [Google Scholar] [CrossRef]
  142. Pastore, S.; Troisi, A.; Romani, R.; Bellezza, I.; Gargaro, M.; De Michele, A.; Orlandi, R.; Guerrera, G.; Bazzano, M.; Polisca, A. Isolation of extracellular vesicles from bitch’s amnion-derived cells culture and their CD59 expression: Preliminary results. Theriogenology 2023, 198, 164–171. [Google Scholar] [CrossRef] [PubMed]
  143. Ophelders, D.R.; Wolfs, T.G.; Jellema, R.K.; Zwanenburg, A.; Andriessen, P.; Delhaas, T.; Ludwig, A.K.; Radtke, S.; Peters, V.; Janssen, L.; et al. Mesenchymal stromal cell-derived extracellular vesicles protect the fetal brain after hypoxia-ischemia. Stem Cells Transl. Med. 2016, 5, 754–763. [Google Scholar] [CrossRef] [PubMed]
  144. Jenner, F.; Wagner, A.; Gerner, I.; Ludewig, E.; Trujanovic, R.; Rohde, E.; von Rechenberg, B.; Gimona, M.; Traweger, A. Evaluation of the potential of umbilical cord mesenchymal stromal cell–derived small extracellular vesicles to improve rotator cuff healing: A pilot ovine study. Am. J. Sports Med. 2023, 51, 331–342. [Google Scholar] [CrossRef] [PubMed]
  145. Homma, K.; Bazhanov, N.; Hashimoto, K.; Shimizu, M.; Heathman, T.; Hao, Q.; Nawgiri, R.; Muthukumarana, V.; Lee, J.W.; Prough, D.S.; et al. Mesenchymal stem cell-derived exosomes for treatment of sepsis. Front. Immunol. 2023, 14, 1136964. [Google Scholar] [CrossRef] [PubMed]
  146. Lange-Consiglio, A.; Lazzari, B.; Pizzi, F.; Idda, A.; Cremonesi, F.; Capra, E. Amniotic microvesicles impact hatching and pregnancy percentages of in vitro bovine embryos and blastocyst microRNA expression versus in vivo controls. Sci. Rep. 2020, 10, 501. [Google Scholar] [CrossRef] [PubMed]
  147. Lange-Consiglio, A.; Gusmara, C.; Manfredi, E.; Idda, A.; Soggiu, A.; Greco, V.; Bonizzi, L.; Cremonesi, F.; Zecconi, A. Antimicrobial effects of conditioned medium from amniotic progenitor cells in vitro and in vivo: Toward tissue regenerative therapies for bovine mastitis. Front. Vet. Sci. 2019, 6, 443. [Google Scholar] [CrossRef] [PubMed]
  148. Lange-Consiglio, A.; Funghi, F.; Cantile, C.; Idda, A.; Cremonesi, F.; Riccaboni, P. Case report: Use of amniotic microvesicles for regenerative medicine treatment of a mare with chronic endometritis. Front. Vet. Sci. 2020, 7, 347. [Google Scholar] [CrossRef] [PubMed]
  149. Lange-Consiglio, A.; Gaspari, G.; Funghi, F.; Capra, E.; Cretich, M.; Frigerio, R.; Bosi, G.; Cremonesi, F. Amniotic Mesenchymal-Derived Extracellular Vesicles and Their Role in the Prevention of Persistent Post-Breeding Induced Endometritis. Int. J. Mol. Sci. 2023, 24, 5166. [Google Scholar] [CrossRef] [PubMed]
  150. Mocchi, M.; Bari, E.; Marrubini, G.; Bonda, A.F.; Perteghella, S.; Tartara, F.; Cofano, F.; di Perna, G.; Giovannelli, L.; Mandracchia, D.; et al. Freeze-Dried Mesenchymal Stem Cell-Secretome pharmaceuticalization: Optimization of formulation and manufacturing process robustness. Pharmaceutics 2021, 13, 1129. [Google Scholar] [CrossRef]
  151. Bemis, L.T.; McCue, P.M.; Hatzel, J.N.; Bemis, J.; Ferris, R.A. Evidence for production of early pregnancy factor (Hsp10), micro RNAs and exosomes by day 8 equine embryos. 8th ISEET Abstracts. J. Equine Vet. Sci. 2012, 32, 398. [Google Scholar] [CrossRef]
  152. Ng, Y.H.; Rome, S.; Jalabert, A.; Forterre, A.; Singh, H.; Hincks, C.L.; Salamonsen, L.A. Endometrial exosomes/microvesicles in the uterine microenvironment: A new paradigm for embryo-endometrial cross talk at implantation. PLoS ONE 2013, 8, e58502. [Google Scholar] [CrossRef]
  153. Rebordao, M.R.; Amaral, A.; Lukasik, K.; Szostek-Mioduchowska, A.; Pinto-Bravo, P.; Galvao, A.; Skarzynski, D.J.; Ferreira-Dias, G. Impairment of the antifi-brotic prostaglandin E2 pathway may influence neutrophil extracellular trapsinduced fibrosis in the mare endometrium. Domest. Anim. Endocrinol. 2019, 67, 1–10. [Google Scholar] [CrossRef]
  154. Volk, S.W.; Bohling, M.W. Comparative wound healing--are the small animal veterinarian’s clinical patients an improved translational model for human wound healing research? Wound Repair Regen. 2013, 21, 372–381. [Google Scholar] [CrossRef] [PubMed]
  155. Iacono, E.; Lanci, A.; Merlo, B.; Ricci, F.; Pirrone, A.; Antonelli, C.; Mariella, J.; Castagnetti, C. Effects of amniotic fluid mesenchymal stem cells in carboxymethyl cellulosegel on healing of spontaneous pressure sores: Clinical outcome in seven hospitalized neonatal foals. Turk. J. Biol. 2016, 40, 484–492. [Google Scholar] [CrossRef]
  156. Lanci, A.; Merlo, B.; Mariella, J.; Castagnetti, C.; Iacono, E. Heterologous Wharton’s jelly derived mesenchymal stem cells application on a large chronic skin wound in a 6-month-old filly. Front. Vet. Sci. 2019, 6, 9. [Google Scholar] [CrossRef]
  157. Enciso, N.; Avedillo, L.; Fermín, M.L.; Fragío, C.; Tejero, C. Cutaneous wound healing: Canine allogeneic ASC therapy. Stem Cell Res. Ther. 2020, 11, 261. [Google Scholar] [CrossRef]
  158. Cheng, Y.; Cao, X.; Qin, L. Mesenchymal Stem Cell-Derived Extracellular Vesicles: A Novel Cell-Free Therapy for Sepsis. Front. Immunol. 2020, 11, 647. [Google Scholar] [CrossRef] [PubMed]
  159. Manzoor, T.; Saleem, A.; Farooq, N.; Dar, L.A.; Nazir, J.; Saleem, S.; Ismail, S.; Gugjoo, M.B.; Shiekh, P.A.; Ahmad, S.M. Extracellular vesicles derived from mesenchymal stem cells—A novel therapeutic tool in infectious diseases. Inflamm. Regen. 2023, 43, 17. [Google Scholar] [CrossRef]
  160. Palma, E.; Tilocca, B.; Roncada, P. Antimicrobial resistance in veterinary medicine: An overview. Int. J. Mol. Sci. 2020, 21, 1914. [Google Scholar] [CrossRef] [PubMed]
  161. Indira, V.; Corteling, R.; Stevanato, L.; Hicks, C.; Sinden, J. The development of stem cell-derived exosomes as a cell-free regenerative medicine. J. Circ. Biomark. 2014, 3, 2. [Google Scholar]
  162. Pollock, K.; Stroemer, P.; Patel, S.; Stevanato, L.; Hope, A.; Miljan, E.; Dong, Z.; Hodges, H.; Price, J.; Sinden, J.D. A conditionally immortal clonal stem cell line from human cortical neuroepithelium for the treatment of ischemic stroke. Exp. Neurol. 2006, 199, 143–155. [Google Scholar] [CrossRef]
  163. Yeo, R.W.; Lai, R.C.; Zhang, B.; Tan, S.S.; Yin, Y.; Teh, B.J.; Lim, S.K. Mesenchymal stem cell: An efficient mass producer of exosomes for drug delivery. Adv. Drug Deliv. Rev. 2013, 65, 336–341. [Google Scholar] [CrossRef] [PubMed]
  164. Haraszti, R.A.; Miller, R.; Stoppato, M.; Sere, Y.Y.; Coles, A.; Didiot, M.C.; Wollacott, R.; Sapp, E.; Dubuke, M.L.; Li, X.; et al. Exosomes produced from 3D cultures of MSCs by tangential flow filtration show higher yield and improved activity. Mol. Ther. 2018, 26, 2838–2847. [Google Scholar] [CrossRef]
  165. Nordin, J.Z.; Lee, Y.; Vader, P.; Mager, I.; Johansson, H.J.; Heusermann, W.; Wiklander, O.P.B.; Hällbrink, M.; Seow, Y.; Bultema, J.J.; et al. Ultrafiltration with size-exclusion liquid chromatography for high yield isolation of extracellular vesicles preserving intact biophysical and functional properties. Nanomedicine 2015, 11, 879–883. [Google Scholar] [CrossRef]
  166. Benedikter, B.J.; Bouwman, F.G.; Vajen, T.; Heinzmann, A.C.A.; Grauls, G.; Mariman, E.C.; Wouters, E.F.M.; Savelkoul, P.H.; Lopez-Iglesias, C.; Koenen, R.R.; et al. Ultrafiltration combined with size exclusion chromatography efficiently isolates extracellular vesicles from cell culture media for compositional and functional studies. Sci. Rep. 2017, 7, 15297. [Google Scholar] [CrossRef]
  167. Mol, E.A.; Goumans, M.J.; Doevendans, P.A.; Sluijter, J.P.G.; Vader, P. Higher functionality of extracellular vesicles isolated using sizeexclusion chromatography compared to ultracentrifugation. Nanomedicine 2017, 13, 2061–2065. [Google Scholar] [CrossRef] [PubMed]
  168. Monguio-Tortajada, M.; Galvez-Monton, C.; Bayes-Genis, A.; Roura, S.; Borras, F.E. Extracellular vesicle isolation methods: Rising impact of size-exclusion chromatography. Cell Mol. Life Sci. 2019, 76, 2369–2382. [Google Scholar] [CrossRef] [PubMed]
  169. Lener, T.; Gimona, M.; Aigner, L.; Börger, V.; Buzas, E.; Camussi, G.; Chaput, N.; Chatterjee, D.; Court, F.A.; Del Portillo, H.A.; et al. Applying extracellular vesicles based therapeutics in clinical trials—An ISEV position paper. J. Extracell. Vesicles 2015, 4, 30087. [Google Scholar] [CrossRef]
  170. Pachler, K.; Lener, T.; Streif, D.; Dunai, Z.A.; Desgeorges, A.; Feichtner, M.; Öller, M.; Schallmoser, K.; Rohde, E.; Gimona, M. A good manufacturing prac-tice-grade standard protocol for exclusively human mesenchymal stromal cell-derived extracellular vesicles. Cytotherapy 2017, 19, 458–472. [Google Scholar] [CrossRef]
  171. Burrello, J.; Monticone, S.; Gai, C.; Gomez, Y.; Kholia, S.; Camussi, G. Stem cell-derived extracellular vesicles and immune-modulation. Front. Cell Dev. Biol. 2016, 4, 83. [Google Scholar] [CrossRef]
Figure 1. Source and overall characterization of MSCs-EVs in domestic animals.
Figure 1. Source and overall characterization of MSCs-EVs in domestic animals.
Animals 14 02147 g001
Table 1. Application of MSCs-derived EVs in pig and sheep. p = porcine; h = human; BM = bone marrow; AT = adipose tissue; UC = umbilical cord.
Table 1. Application of MSCs-derived EVs in pig and sheep. p = porcine; h = human; BM = bone marrow; AT = adipose tissue; UC = umbilical cord.
AnimalSource of EVsDistrictApplicationMode of AdministrationReference
PigpBM-MSCsRespiratoryInfluenza virusIntratracheal[70]
pAT-MSCsCardiovascularMyocardial infarctionIntraoperative insertion of EVs combined with biocompatible cardiac scaffolds[81]
pBM-MSCsCardiovascularMyocardial infarctionIntraoperative insertion of EV collagen patch[90]
pAT-MSCsUrinaryModel of metabolic syndrome and renal artery stenosis in cardiovascular complicationsIntrarenal injection[67,68,75,77,78,79,80,82,83]
pAT-MSCsVertebral columnInducted spinal cord injuryIntrathecal injection[89]
pAT-MSCsHepaticInducted liver injury (hemi-hepatectomy and hepatic ischemia-reperfusion injury)Intravenous[87,88]
SheephBM-MSCsNeurologicalHypoxic ischemic encephalopathy (HIE)Intravenous[143]
hUC-MSCsMusculoskeletalLigament injuryApplication onto a type 1 collagen sponge[144]
hBM-MSCsSystemicPneumonia/sepsisIntravenous[145]
Table 2. Preclinical studies in domestic animals with the use of EV/CM in culture. AM = amniotic membrane; BM = bone marrow; AT = adipose tissue; Syn Fluid = synovial fluid; WJ = Wharton’s jelly; e = equine; b = bovine; c = canine; f = feline.
Table 2. Preclinical studies in domestic animals with the use of EV/CM in culture. AM = amniotic membrane; BM = bone marrow; AT = adipose tissue; Syn Fluid = synovial fluid; WJ = Wharton’s jelly; e = equine; b = bovine; c = canine; f = feline.
AnimalSource of EVsApplication CultureEffectsReference
HorseeAM-MSCsEndometrial cellsreduced the apoptosis rate, increased cell proliferation values, downregulated pro-inflammatory gene expression, and decreased the secretion of pro-inflammatory cytokines[134]
eAM-MSCsTenocytesinduced a down-regulation of MMP1, MMP9, MMP13 and TNFα expression[130]
eBM-MSCs eAT-MSCs ad Syn FluidChondrocytesreduced inflammation[59]
eBM-MSCsChondrocytesincreased the articular chondrocyte collagen protein amounts, mRNA levels of Prg4, and enhanced the proliferation and migratory capacities of chondrocytes[99]
eBM-MSCs
(autologous)
Chondrocytesanti-inflammatory effects on gene expression following chondrocyte exposure to tumor necrosis factor α and Interleukin 1β[97]
eBM-MSCsChondrocytesinduced a greater increase in equine articular chondrocyte-neosynthesized hyaline-like matrix by modulating collagen levels, increasing PCNA, and decreasing Htra1 synthesis[104]
eAM-MSCsAlveolar macrophagesModulatory-effect release of TGF-alfa and β and possibly IL-6[133]
BovinebAM-MSCsBlastocystsaddition of EVs during in vitro embryo production seemed to influence the developmental capacity and implantation potential of the embryos and regulate the expression of specific miRNAs that regulated blastocyst development[146]
bAM-MSCs (CM)Mammary epithelial cellscould attenuate bacterial growth, as evaluated by the number of CFUs. After 24 h of culture with S. aureus, 89.67% of mammary epithelial cells treated were still alive, whereas all cells cultured and not treated were dead[147]
DogcWJ-MSCsFibroblastssuppressed the proliferation of cell T CD4+ using TGF-β and adenosin[138]
cBM-MSCsMurine microglia cellsdecreased inflammation (decrease IL-1β)[113]
cAT-MSCs (Lyosecretoma)Tenocytes, chondrocytes and AT-MSCsinduced proliferation of cells in dose-dependent manner and showed anti-elastase activity[98]
cAM-MSCsCoculture with AM-MSCs and AT-MSCs15–20% increased expansion rate[141]
cAT-MSCsSemen during cryopreservationinitiated damaged-sperm repair (higher motility, live sperm percentage, membrane and acrosome integrity; higher expression of genes related to the repair of plasma membrane and chromatin material) and decreased reactive oxygen species production[106]
CatfAT-MSCs and fibroblastsHuman THP-1 MacrophagesMSCs-EVs had lower levels of pro-inflammatory cytokines (IL-1β, TNF-α) and higher level of IL-10. MSCs-EVs played a crucial role in immune defense compared with EVs–fibroblasts[118]
Table 3. Application of EVs/CM (culture medium)-derived MSCs in veterinary medicine. e = equine; b = bovine; c = canine; f = feline; AT = adipose tissue; AM = amniotic membrane; BM = bone marrow; UC = umbilical cord.
Table 3. Application of EVs/CM (culture medium)-derived MSCs in veterinary medicine. e = equine; b = bovine; c = canine; f = feline; AT = adipose tissue; AM = amniotic membrane; BM = bone marrow; UC = umbilical cord.
AnimalSource of EVsDistrictApplicationMode of AdministrationReference
HorseeAT-MSCsMusculoskeletalLigament injuryUltrasound-guided
injection at the injury site
[96]
eAT-MSCsMusculoskeletalOsteoarthritisIntra-articular injection[101]
eAM-MSCsReproductiveChronic endometritisIntrauterine[148]
eBM-MSCsReproductiveChronic endometritisIntrauterine[103]
eAM-MSCs (CM)ReproductivePersistent post-breeding-induced endometritisIntrauterine[149]
CattlebAM-MSCsReproductiveBlastocyst developmentIn vitro during embryo production[146]
bAM-MSCs (CM)Mammary glandAcute and chronic mastitisIntramammarily[147]
DogcBM-MSCsSkinInducted skin woundSubcutaneous injection[105]
cAT-MSCs
(Lyosectetoma)
MusculoskeletalOsteoarthritisIntra-articular injection[98]
cAT-MSCsUrinaryRenal ischemia-reperfusion injuryRenal cortex injection[116]
CatfAT-MSCsUrinaryPost-renal acute kidney injury (PR-AKI)Intravenous[122]
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

Lanci, A.; Iacono, E.; Merlo, B. Therapeutic Application of Extracellular Vesicles Derived from Mesenchymal Stem Cells in Domestic Animals. Animals 2024, 14, 2147. https://doi.org/10.3390/ani14152147

AMA Style

Lanci A, Iacono E, Merlo B. Therapeutic Application of Extracellular Vesicles Derived from Mesenchymal Stem Cells in Domestic Animals. Animals. 2024; 14(15):2147. https://doi.org/10.3390/ani14152147

Chicago/Turabian Style

Lanci, Aliai, Eleonora Iacono, and Barbara Merlo. 2024. "Therapeutic Application of Extracellular Vesicles Derived from Mesenchymal Stem Cells in Domestic Animals" Animals 14, no. 15: 2147. https://doi.org/10.3390/ani14152147

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

Article Metrics

Article metric data becomes available approximately 24 hours after publication online.
Back to TopTop