Skip to main content

The potential use of mesenchymal stem cells-derived exosomes as microRNAs delivery systems in different diseases

Abstract

MicroRNAs (miRNAs) are a group of small non-coding RNAs that regulate gene expression by targeting mRNA. Moreover, it has been shown that miRNAs expression are changed in various diseases, such as cancers, autoimmune disease, infectious diseases, and neurodegenerative Diseases. The suppression of miRNA function can be easily attained by utilizing of anti-miRNAs. In contrast, an enhancement in miRNA function can be achieved through the utilization of modified miRNA mimetics. The discovery of appropriate miRNA carriers in the body has become an interesting subject for investigators. Exosomes (EXOs) therapeutic efficiency and safety for transferring different cellular biological components to the recipient cell have attracted significant attention for their capability as miRNA carriers. Mesenchymal stem cells (MSCs) are recognized to generate a wide range of EXOs (MSC-EXOs), showing that MSCs may be effective for EXO generation in a clinically appropriate measure as compared to other cell origins. MSC-EXOs have been widely investigated because of their immune attributes, tumor-homing attributes, and flexible characteristics. In this article, we summarized the features of miRNAs and MSC-EXOs, including production, purification, and miRNA loading methods of MSC-EXOs, and the modification of MSC-EXOs for targeted miRNA delivery in various diseases.

Graphical abstract

Video abstract

Introduction

MicroRNA (miRNAs) are short single-stranded non-coding RNA (ncRNA) that control and affect the expression of various genes [1]. About 30–60 percent of all mammalian proteins can be targeted via miRNAs, which are involved in different cellular and developmental procedures. MiRNAs control cell proliferation, differentiation, renewal, and apoptosis [2]. In addition, with the quick progress of next-generation sequencing (NGS), investigators have understood that miRNA interposition can alter associated physiological roles, causing inflammation of cell penetration, cancer, infectious diseases, neurological and immunological disorders, and other diseases [3,4,5]. Currently, some miRNA medications are undergoing clinical phases, and excellent advance has been made in the investigation and progress of miRNA medication patents and miRNA treatment. However, there are some obstacles to the clinical usage of miRNAs, including inconstancy in vivo and problems related to crossing biological barriers [6].

Exosomes (EXOs) are endosomal source membrane extracellular vehicles (EVs) with a dimension of about 30 to 150 nm secreted via types of cells. EXOs contained different compounds, including nucleic acid (such as miRNAs), proteins, enzymes, and lipids [7,8,9]. EXOs are natural biological materials with the intrinsic roles and protein machinery that safely and effectively deliver their load biomaterials among origin and target cells over long distances. EXOs play a crucial function in intercellular communication. EXOs are being progressively used as curative carriers. There is robust literature that EXOs therapy emerges as harmless with low immunogenicity [10,11,12]. Furthermore, EXOs are amenable to in vivo and in vitro loading of curative agents and membrane modifications to improve tissue-particular homing [13]. Several EXOs are obtained from various cells, such as mesenchymal stromal cells (MSCs) [14, 15]. MiRNA-loaded EXOs have been employed in multiple diseases. MiRNAs encapsulated in EXOs are usually attained via transfecting adipose tissue-obtained stem cells (SCs) and MSCs with the desired miRNA [16].

The properties of MSCs include the facility of production and isolation, low immunogenicity, harmless, lack of side effects, and efficient therapeutic method. Robust clinical assessments suggest that MSC-based therapies lack side effects, practicable and are efficient [17,18,19]. In addition, several investigations have obtained and identified EXOs from different MSC sources, such as bone marrow (BM), adipose tissues (AD), and umbilical cord (UC), approving their robust anti-inflammatory, anti-fibrotic and angiogenesis-regeneration capabilities [20]. MSC-EXOs have numerous exclusive features, including small dimensions, low immunogenicity, long-term circulating, sustained release, tumor-homing, tissue-particular homing, excellent permeation, and excellent biocompatibility. In novel investigations, researchers use MSC-EXOs as a vehicle to transport RNA, protein, and molecular medications to particular sections of the body to attain targeted therapy [21,22,23]. Up to now, about 150 miRNAs and more than 900 proteins have been recognized in loads of MSC-EXOs, leading to the modification of a diversity of actions in target cells through diverse pathways [24, 25]. However, difficulties, including carrier isolation and purification, maintenance and transport, medication loading method, and targeting, still exist [21,22,23].

In this review, we will summarize recent advances regarding the application and miRNAs loading techniques of MSC-EXOs, MSC-EXOs production method, and routes of administration of MSC-EXOs. Moreover, the transfer of different miRNAs through MSC-EXOs to treat diverse diseases will be highlighted.

MiRNAs biogenesis and characteristics

MiRNAs are having the size of 18 ~ 25 nucleotides (nt) in length that post-transcriptionally up or down-regulate genes by connecting to the 3′-untranslated regions (3′-UTRs) of mRNAs [26, 27]. Briefly, the biogenesis of miRNAs begins with their transcription via RNA polymerase II (RNAP II), leading to an early transcript recognized as pri-miRNA [28, 29]. Afterward transcription, the ordinary hairpin-loop secondary construction existing in pri-miRNAs is detected and cleaved via the microprocessor complex (formed via DGCR8 and Drosha). The produced precursor miRNAs (pre-miRNAs) are transported to the cytoplasm and subsequently processed via the Dicer nuclease to produce a double-stranded RNA. The mature miRNA sequence is elected via Ago2 and involved in the RNA-induced silencing complex (RISC) to use its regulatory function [4, 30, 31]. Each miRNA can bind to several mRNAs. The role of miRNAs is to suppress the protein synthesis of protein-coding genes by suppressing the translation of targeted mRNA or through mRNA degradation. Furthermore, miRNAs can activate the translation of targeted mRNAs, switching between translation suppression and triggering in coordination with the cell cycle. In addition, miRNAs have been shown to play a crucial function in many biological activities, such as metabolism, immunity, cell development, apoptosis, differentiation, and signal transduction [32,33,34]. The deregulation of miRNAs in disease situations can be used as possible curatives via either miRNA substitution treatment utilizing miRNA mimics or suppression of miRNA role via antagomiRs [35, 36] (Fig. 1).

Fig. 1
figure 1

The biogenesis of miRNAs and their pathways to the target cell. a-d miRNA biogenesis starts in the nucleus. Pri-miRNA transcripts become pre-miRNA through the Drosha. Pre-miRNA is carried out of the nucleus through Exportin-5 into the cytoplasm. e-j MiRNA maturation is the elective loading of the functional strand of the small RNA duplex onto the RISC. miRISC induces the destruction of mRNA and translational inhibition via the interplay with the supplementary sequences in the 3′-UTR of target mRNA. k) MiRNAs are released in the extracellular environment or circulate through AGO2 protein, microvesicles, EXOs, high-density lipoprotein (HDL), and low-density lipoprotein (LDL) particles

MiRNA delivery system

Notwithstanding the excellent capability of miRNA in treating various diseases, it has several limitations, which must be resolved. Firstly, because of the negative charge of miRNAs, they are hard to penetrate the cell. Moreover, miRNAs are unstable in vitro due to, destruction by nucleases, and immunotoxicity. Hence, the advance of harmless and effective miRNA delivery methods is highly important to the most effective use of miRNA as therapeutic agents [37]. Chemical changes have been a principal method for antagomiRs to inhibit their destruction via nucleases in cells and in vivo. Viral and non-viral carriers have been produced to transfer miRNA mimics or antagomiRs for the targeted treatment of various diseases. Viral carriers contain viruses that are altered to be replication-deficient, however, they can be utilized to carry nucleic acid for expression [38]. Moreover, different types of nanoparticles (NPs) propose unique chances for cell-particular controlled transfer of miRNAs to treat diseases. MiRNA-encapsulated in NPs has been offered, by NPs potential to protect the packed factor from the extracellular environment, thereby decreasing destruction, and increasing circulation time and selective accumulation [39, 40]. Notwithstanding the significant progress and achievements in their formulation, preparation, and efficiency of synthetic miRNA delivery systems, there is a developing acknowledgment that nature has particulates with some of the highly favorable properties of miRNA delivery systems, including immunologically inert, an inherent tropism that causes exceedingly selective and effective entrance into special host cells, stability in the different condition, and diverse therapeutic cargos and that such particulates should be used as miRNAs transfer carriers. Red blood cells (RBCs), bacteria, lymphocytes, extracellular vesicles (EVs), and EXOs are examples of natural miRNA delivery systems [41,42,43,44] (Fig. 2).

Fig. 2
figure 2

Various miRNAs delivery systems, including organic and inorganic nanoparticles, viral vectors, and extracellular vesicles. Inorganic NPs, including lipid-based nanoparticles, polymeric carriers/dendrimer-based, cell-isolated membrane vesicles, and 3D scaffold-based carriers

MSCs-EXOs biogenesis and characteristics

MSCs are recognized as the most desirable origin for curative EXOs. This can be ascribed to the harmlessness of MSCs, the possibility of the generating MSC‐EXOs as an off‐the‐shelf production, and lack of tumorigenic possibility of MSC-EXOs. Therefore, the use of MSC‐EXOs substitute MSCs in a diversity of diseases, such as cancer, autoimmune diseases, tissue injury, neurodegenerative diseases, infectious diseases, ocular diseases, and dermal diseases [45, 46]. Moreover, MSCs are the only recognized cells able to generate EXOs at a large scale. Large scale generation can be attained via obtaining and, culturing MSCs in vitro [47].

They have been classified based on EXOs dimensions, compounds, and formation mechanism in the last years. There are recognized subtypes of EVs comprising microvesicles (MVs), apoptotic bodies (ABs), and EXOs. In addition to EXOs, other membrane vesicles generated via cells contain plasma membrane-budded MVs and ABs [48]. In further investigations, Wnt and mTOR pathways, which lead to overexpression of β-catenin, are presented as main controllers essential both for MSC-EXO discharge and as well as supporting the self-renewal of MSCs [49, 50].

The mechanism of EXO generation during EXO biogenesis includes the beginning; the plasma membrane is internalized to form an endocytic vesicle called an early endosome, followed by early endosome to late endosome transformation. The budding of late endosomal membranes results in forming intraluminal vesicles (ILVs) within large MVBs [51]. Comprehensively, EXOs biogenesis is controlled via two distinct molecular pathways, comprising endosomal sorting complex required for transport (ESCRT) machinery- affiliate and ESCRT-autonomous [52, 53]. EXOs discharge relates to transfer and plasma membrane fusion of the secretory MVBs subsequent internal budding of ILVs, which needs multiple critical agents, such as molecular switches (small GTPase), cytoskeleton (microtubule and microfilament), molecular motors (dynein and kinesin) and the membrane fusion proteins (SNARE complex). Rab GTPase is the most crucial agent, with more than 70 subtypes placed on the surface of membranes, where they can control vesicle traffic, such as budding, motility, and fusion. In addition, Rab35 localizes to the surface of oligodendroglia cells in a GTP-related method, regulating the docking of endocytic vesicles with the membrane. MVBs and plasma membranes can amalgamate through intermediation via Rab and the corresponding effector on the MVB membrane [54]. EXOs are taken up via the recipient cells by direct connecting to the plasma membrane, binding to the host-receptor, and by endocytosis. EXOs comprise proteins, DNA, RNA (such as miRNA, lncRNA, mRNA, tRNA, and circRNA), and cholesterol (Fig. 3 and Fig. 4) [55, 56].

Fig. 3
figure 3

The biogenesis of MSC-EXOs and their pathways to the target cell. a EXOs biogenesis starts within the endosomal system. Within the endosomal system, internalized loads are encapsulated into early endosomes, b which then mature into late endosomes or MVB. Different compounds are also transported from the trans-Golgi network and presumably from the cytosol. c In addition, MVBs can be transported to lysosomes for decay or d move along microtubules to amalgamate with the cell membrane and discharge EXOs into the extracellular environment. MVB amalgamation with the membrane is a fine-tuned process, which needs various vital agents, including Rab GTPases and SNARE complexes. e) EXOs intercede their effects on the target cells via three essential methods: 1- recipient cell receptor interceded signal amplification, 2- direct connecting to the plasma membrane and fusion, and 3- endocytosis

Fig. 4
figure 4

a 3D schematic structure and contents of MSCs-EXOs. b EXOs usually contain dynamic loads, including different proteins, enzymes, lipids, RNA (such as miRNAs), and DNA. Moreover, MSC-EXOs carry and deliver different hydrophobic and hydrophilic drugs

MSC-EXOs preparation by different techniques

To ensure the biological function of EXOs, a standardized engineering procedure, including a process by good manufacturing practice (GMP), of EXOs is essential. As EXOs are released via cells, a generation method could be created by a large-scale cell cultivation technique. EXOs in clinical experiments need to comply with GMP. Three crucial challenges are prevalent in GMP for EXOs, including upstream of cell culture, downstream of the purification method, and EXOs quality control [57, 58].

The minimum prerequisites for utilizing EXOs as medication delivery systems are an excellent efficiency cell origin of EXOs and a reproducible, scalable purification procedure to form a highly determined population of EXOs. Though most cell kinds generate EXOs, the quantity of EXOs generated via each cell kind is highly changeable. EXOs generated via cultured cells are ordinarily obtained from the medium conditioned for 1–7 days via the cells utilizing either medium without serum or medium with the serum that had been discharged of MVs [13]. Investigators have applied scaffolds, spherical culture, a mercantile hollow-fiber bioreactor method, stirred suspension bioreactors, perfusion-based bioreactors, and microcarrier-based 3D culture techniques to attain more significant EXOs generation [59, 60].

MSC-EXOs produced by 3D cultures

MSCs cultivated in two-dimensional (2D) traditional tissue culture polystyrene flasks can lead to low efficiency, restricting their clinical use. 3D culture might be an effective method for the enhanced biological purposes [61, 62]. The advantages of using the three-dimensional cultivation method include the culture, a multitude of cells the high efficiency of EXOs. Compared with 2D culture, the EXO generation of 3D culture is 19.4 times higher. In addition, 3D-EXO cultures are more concentrated in the harvested supernatants (15.5-fold) than 2D-EXOs, which results in a greater EXOs collection yield [63]. Scaffold-free and scaffold-based culture methods produced of natural or synthetic substances are the two broad groups of 3D culture methods. There are various scaffold-based 3D culture techniques, including hydrogels and solid scaffolds. For example, researchers showed that contrasted with 2D culture, human BM-MSCs cultured in the 3D collagen scaffolds produced further EXOs with enhanced repair role in rats afterward intracranial injury. In another investigation, researchers showed that scalable microcarrier-in 3D cultures could twofold the accumulation of MSCs and efficiency more EXOs than 2D cultures [62, 64, 65].

MSC-EXOs produced by bioreactor production methods

The conventional 2D culture technique to generate EXOs from adherent cells does not allow for a sustained generation of large amounts of biological production and thus prevents their usage for clinical trials. On the other hand, bioreactor methods remove this limitation while preparing the needful milieu to preserve excellent cell viability and homeostasis. EXO manufacture according to a bioreactor method offers numerous advantages where scalability, decreased manual manipulation, and simple monitoring and regulating of culture parameters can be attained. Moreover, the usage of a bioreactor can enhance the translational competency of bio therapeutics as this milieu is an improved presentation of the cell–cell interplay found in vivo as compared to the flask-based technique [64, 66]. The commercial hollow-fiber bioreactor (HFB) method has been used efficiently for the mass production of EXOs. The HFB from Fiber Cell Systems allows for seeding large quantities of adherent cells based on its HFB technology which subsequently enhances the cell seeding surface area (an average-sized cartridge provides 4000 cm2 of surface area) [64, 67, 68]. Novel investigations have used more straightforward methods (such as flat-plate bioreactors), permitting a more accurate approach for comprehend and restraining flow-derived shear stress. However, still absence the adjustability essential for dynamic construction and experimental investigation requirements [69]. Moreover, stirred suspension bioreactors (SSBs) have been efficiently utilized to scale up MSCs manufacture. These techniques are partly simple to perform, propose scalability benefits, supply an excellent capacity for cell development, can help adherent cell development with the addition of microcarriers or accumulations, and allow a homogenous culture microenvironment because of continuous stirring [70] (Fig. 5).

Fig. 5
figure 5

MSCs have been isolated from different sources (A and B). Exosomes are produced by various methods from diverse types of MSCs (C). The standard diagram of the HFB-based 3D culture method. The technique was prepared of a pulsatile perfusion pump, an oxygenator, a cartridge comprising thousands of hollow fibers, a bottle of culture media, and the linking tube

Separation and purification of MSC-EXOs

MSC-EXOs as a medication delivery system in vivo, a harmless, effective, and trustworthy purification technique is essential. Presently, there is numerous generally utilized techniques for EXO filtration [13]. Various recognized methods, including differential ultracentrifugation, density gradients, sedimentation, ultra-filtration, and size exclusion chromatography, have been used for EXO isolation [71]. The most popular utilized separation technique for EXOs is differential centrifugation, where an enhancing centrifugal force from 200 × g to 100,000 × g is used to primarily deplete the media of bigger units and cell remnants before finally precipitating EXOs at 100,000 × g [13, 72]. Newly, some commercial kits have been offered to separate EXOs for several targets [73]. These kits are more significant to ultracentrifugation because of being less time-consuming, less method sensitive, and higher compatibility with confined content of samples [71, 74, 75].

Newly, microfluidic methods have been used in EXOs isolation. The attributes of the microfluidic-based EXOs isolation methods, such as high surface-area-to-volume proportion, reduce use of samples, quick analysis, laminar flow, and simple implementation method, make them appropriate for EXOs isolation with an excellent recovery level and purity for clinical uses [76]. To efficiently exploit the dimensional difference among EXOs, other types of EVs, and cellular debris, researchers manufactured a poriferous silicon nanowire-on-micropillar “nano-trap” produced from ciliated micropillars. This manufactured microfluidic machine more favorably traps EXOs with a size of 40–100 nm, while purifying proteins, larger EVs, and cellular debris. Furthermore, trapped EXOs can be recovered by solving the porous silicon nanowires in phosphate-buffered saline buffer [77].

Among the several separation techniques, tangential flow filtration (TFF) has been offered as the good technique for the large-scale production of EXOs. The TFF methods accessible for GMP are now in application and offer confirmed procedures and GMP documents. This method was primarily presented in 2010 for the separation of EXOs based on size and was progressively used for EXOs purification or concentration in different investigational settings. More significantly, novel investigation displayed the superior efficiency and activity of EXOs separated via TFF compared with those separated through ultracentrifugation. The great-purity separation of EXOs is attainable with extra diafiltration by TFF with suitable pore dimensions and parameters, such as transmembrane pressure, flow rate, and diafiltration factor [78] (Fig. 6).

Fig. 6
figure 6

Traditional and new techniques of EXO purification. EXOs are classified into three main kinds associated with their location of source, density, expression of markers, and dimensions. Traditional techniques of EXO separation contain differential ultracentrifugation and size-exclusion chromatography. a Size-exclusion chromatography utilizes biofluids as a mobile state versus a porous static state to differentially elute molecules with a reverse speed relative to their dimension. b Differential ultracentrifugation depends on the isolation of EXO subpopulations through progressively higher speed rates. Polyethylene glycol (PEG)-based sedimentation uses a solution to ease a polymer-encapsulated vesicle collection in large amounts. c Immunoaffinity absorption utilizes antibodies targeted versus exosomal surface proteins to separate particular vesicle populations. The microfluidics (MF) method utilizes chips with particular antibody-interceded connections to capture EXOs effectively. d Ultrafiltration is associated with a filter of a particular pore size that forms a vesicle-rich filtrate particular to the favorable dimensions [164, 165]

MiRNAs loading methods in MSC-EXOs

EXOs could be packed with different therapeutic agents either in vivo during their biogenesis or in vitro in isolated EXOs. However, the progress and improvement of effective use of EXO loading methods are presently restricted via our little knowledge of EXO biology, construction, and biogenesis and an absence of EXO-associated investigation and progress tools [79, 80]. To load miRNAs in EXOs, two techniques have been used. A cell culture overexpressing the miRNA of interest is produced, leading to enhanced miRNA expression and EXO discharge with the entrapped miRNA. Another method is separating EXOs and loading them with miRNAs [16]. Other methods of extracellular or in vitro MSC-EXOs drug loading includes: medication co-incubation, electroporation, acoustic processing, lipofection, sonication, squeeze technique, extrusion, saponin-helped loading, and freeze–thaw cycles. However, these techniques also have drawbacks, including the accumulation of EXOs, exosomal membrane damage, toxicity to target cells, and extreme separation and purification stages [81,82,83,84,85].

Another method for MSC-EXOs loading is to combine medications into EXOs during their biogenesis. This method is mainly related to the payload that cannot be encapsulated onto isolated EXOs, including cytosolic and transmembrane proteins or extraordinary molecular weight RNA, including mRNA [13, 86, 87]. A relationship matrix analysis investigation demonstrated a weak correlation between the RNA amount of MSC-EXOs and the principal MSCs, showing that miRNAs were electively packed into the EXOs. It has been proposed that RNA-binding proteins (RBPs), including hnRNPA2B1 and hnRNPA1 per se, connect to miRNAs and control their elective loading into EXOs. The endosomal sorting complex required for transport (ESCRT) has a significant function in loading protein-miRNA compounds into EXOs. Moreover, ESCRT-autonomous pathways, including ceramide–interceded mechanisms, have been demonstrated to be implicated in load sorting into EXOs. Additional study is required to discover the particulars of the elective miRNA loading method [88]. Moreover, RBPs and cell priming (such as hypoxia and inflammatory cytokines) methods can also be used to regulate the miRNA load of EXOs. Methods including sonication, electroporation, and CaCl2-heat shock can pack the miRNAs per se into the EXOs [89].

MSCs-EXOs routes of administration

Choosing the best appropriate MSC-EXOs injection method, containing the dose injected, and considering the constancy, biological distribution, and toxicity of delivered EXOs is one of the main to attain effective cell-free treatment [90]. Diverse methods of administration utilized in MSC-EXOs have been studied, and it was defined that the route of administration had a significant effect on the preparation design (Fig. 7).

Fig. 7
figure 7

Diverse ways of administration utilized in MSC-EXOs delivery, including a the parenteral administration comprises; the three most common ways are intramuscular (i.m.), intravenous (i.v.), and subcutaneous (S.C.) administration; b Oral administration is a more straightforward and non-invasive method, although the transported EXOs must remain stable pass via the gastrointestinal tract [104]; c Intranasal injection resulted in improved brain aggregation of EXOs at the damaged brain location, compared to i.v. administration; d Intraperitoneal injection of EXOs is another injection method, with widespread EXO distribution capability [97]; e Regarding dermal injection routes, administration into the dermis, recognized as intradermal administration, and topical use are other easy choices to remedy dermal diseases; f Intra-articular administration next to the injured tissue is utilized for rheumatoid arthritis, leading to more effectiveness tissue repair because of the direct use of the drugs to the injured region; g MSCs-EXOs permeate through ocular surface and enhance the bioavailability of therapeutic agents

Parenteral route

The parenteral administration comprises; the three most common ways intramuscular (IM.), intravenous (IV), and subcutaneous (S.C.) administration. It allows the direct administration of the drugs in systemic blood circulation. It is usually utilized when the drug is removed from the system by initial crossing metabolism or drug destruction in the stomach and gastrointestinal tract [91,92,93]. However, this medication route of injection has unique problems, including the aggregation of EXOs in non-targeted organs, usually the liver, spleen, and lungs, and rapid elimination from the organism [94]. For example, in bleeding into the brain tissue of a rat, DiI-labeled MSC-EXOs attained brain, liver, lung, and spleen afterward, IV administration. Biological distribution of systemically injected EXOs is an active procedure: a fast stage of delivery in the liver, spleen, and lungs within about 30 min upon injection is followed by a removal stage by hepatic and renal processing, eliminating EXOs in 1 to 6 h afterward injection [95]. In another study, Yang Zhou et al. showed that the topical drug of smearing human adipose-MSC-EXOs (hAD-MSC-EXOs) is more useful in improving skin injury regeneration than the S.C. administration of hAD-MSC-EXOs. For systemic injection, local injection composed treatment with the IV administration of hAD-MSC-EXOs presented an extra advantage over either therapy alone to improve cutaneous repair [96].

Intraperitoneal delivery

Intraperitoneal injection of EXOs is another injection method with widespread EXO distribution capability. Moreover, researchers showed that intraperitoneal administration of AD-MSC-EXOs leads to an immunomodulatory influence on autoimmune type 1 diabetes, enhancing regulatory T-cell population and their production unchanged in lymphocyte proliferation [97].

Intra-articular injection

Intra-articular administration next to the injured tissue is utilized for rheumatoid arthritis, leading to more effective tissue repair because of the direct use of the drugs to the injured region. For example, researchers showed that 12 intra-articular administrations per week of hMSC-EXOs were efficient in regenerating osteochondral lesions of the talus in rats [98]. Another investigation showed that the amalgamation of MSC-EXOs and hyaluronic acid injected at a clinically admissible frequency of 3 intra-articular administrations can improve stable and functional cartilage restoration in a rabbit after traumatic cartilage defect model when compared with HA alone [99].

Intranasal route

Intranasal injection resulted in improved the brain aggregation of EXOs at the damaged brain location, compared to IV administration. This injection route is an appropriate choice when the purpose is to pass the blood–brain barrier, preventing EXO disintegration in other organs and supporting aggregation in the brain, leading to improved neuroprotective efficacy [100, 101].

Transdermal route

Regarding dermal injection routes, administration into the dermis, recognized as intradermal administration, and topical use are other easy choices to remedy dermal diseases. The topical use of EXOs released via MSC-EXOs on the skin is a novel and interesting subject in the pharmaceutical. Newly, researchers showed that the topical use of human umbilical cord blood MSC-EXOs (hUC-MSC-EXOs) on ex vivo human skin, led to an enhanced expression of skin extracellular matrix (ECM) genes and therefore helped to rejuvenate the skin. In addition, the penetration and effectiveness of EXOs can be additionally enhanced by augmenting skin penetrance [102, 103].

Oral route

Oral administration is a more straightforward and non-invasive method, although the transported EXOs must remain stable and pass via the gastrointestinal tract [104]. The oral route, similar to other routes, has been used to inject chemotherapeutic medications, including curcumin, leading to 3–fivefold greater amounts of curcumin in different organs compared to without encapsulated drug delivery [105]. Newly, an immune tolerance mechanism interceded via free light chain-covered, antigen-particular, miR-150-EXOs that affect the antigen-offering cells proved more efficient afterward oral injection [106].

Ocular route

Ocular injection methods, comprising subconjunctival, intravitreal, and intraocular injection, despite being invasive and disturbing injection routes, have been used to treat diabetic retinopathy (damaging the retina) complications with progressive consequences. MSC-EXOs were as effective as transplanted MSCs in restricting the size of eye damage and inflammation. Immediately afterward, intravitreal administration of MSC-EXOs, because of nano-size, distributed quickly all over the retina and considerably decreased retinal injury and inflammation. MSC-EXOs effectively delivered trophic and immunomodulatory agents to the inner retina and effectively improved survival and neuritogenesis of damaged retinal ganglion cells [107,108,109].

Different types of MSC-EXOs as miRNAs delivery systems in different diseases

MSC has been effectively isolated from various origins such as bone marrow (BM), adipose tissue (AD), Wharton’s Jelly (WJ), umbilical cord (UC), placenta, the dental pulp (DP), or amniotic fluid (AF) among others [110]. BM-MSCs have the benefits of less infection amount of pathogenic microorganisms, constant biological efficiency, low immunogenicity, and a large number of feasible passages [74]. BM-MSC-EXOs could speed up the production and migration of endothelial cells and osteoblast cells, improving angiogenesis, and osteogenesis to improve fracture repair [111]. In another study, researchers showed that hBM-MSC-EXOs therapy considerably decreased liver fibrosis in rats [112]. UC-MSCs can be obtained via a non-invasive strategy and easy cultured, thus offering their advantage over other types of MSCs for therapeutic objectives. Because of their unique characteristics, such as self-renewal, multipotency, and availability simultaneous with their immunosuppressive capability and lesser ethical worries, UC-MSCs treatment is defined as hopeful curative choices in cell-based treatments [113]. HUC-MSC-EXOs, acquired by extensively expanding hUC-MSCs in vitro, is convenient to extract, store, and transport, lower in immunogenicity, and better in biocompatibility. Over the past decades, it has been found that hUC-MSC-EXOs are mainly involved in improving tissue regeneration via delivering proteins, lipids, RNAs, and DNAs, which increase the progress of “cell-free therapy” [114]. The human body is abundant in AD tissue and, therefore, contrasted with UC-MSCs and BM-MSCs. AD-MSCs are plentiful with widespread origins and have extraordinary separation efficiency. However, AD-MSCs have challenging needs for storing situations and have low immunogenicity, which has limited their clinical use. The membrane construction of EXOs is relatively constant, and AD-MSC-EXOs can be stored for long times; hence, AD-MSC-EXOs has produced enhanced attention from investigators [74, 115]. AD-MSC-EXOs used preservative influences versus radiation-stimulated brain damage via reducing oxidative stress injury and decreasing inflammation and microglial penetration [116].WJ-MSCs, a mucosal-linked tissue of the UC, may be improved alternatives to BM-MSCs or AD-MSCs since WJ-MSCs are younger and preserved from injuries caused by senescent, environmental toxins and diseases [117].

MSC-EXOs can be an effective transfer mechanism for miRNAs. It's been investigated widely as a manner to release miRNAs in a controlled and targeted method to treat different cancers, neurodegeneration, autoimmune disorders, infectious diseases, and other diseases.

MSC-EXOs as miRNA delivery system in cancer

Numerous investigations have displayed that MSC-EXOs play a significant function in tumor development, angiogenesis, malignancy, and medication resistance. However, inconsistent consequences have shown that MSC-EXOs could as well as inhibit tumors by particular mechanisms, including controlling immune reactions and intercellular signaling [118]. Unchanged MSC-EXOs can suppress tumors, while altered MSC-EXOs have participated in the inhibition of cancer development and expansion by the transfer of numerous therapeutics agents [119, 120]. In most cancers, the disorder in the regulation of miRNAs not only happens as a result of malignancy development, but is per se implicated in tumor suppression and progress because of their functions as oncomiRs or tumor suppressor miRNAs. MiRNA repair is commonly attained via increasing the expression level of tumor suppressors miRNAs utilizing synthetic miRNA mimics and viral carriers or even decreased expression level of oncomiRs utilizing antagomiRs [119] (Fig. 8).

Fig. 8
figure 8

Function of MSC-EXOs as miRNA delivery method in different types of cancer. a/b Following discharge, MSC-EXOs are absorbed through cancer cells, c and the miRNAs encapsulated in MSC-EXOs regulate various processes, including involvement in cancer and tumor microenvironment immune responses, possibly tumor development, invasion, metastasis, angiogenesis, and chemotherapy resistance

BM-MSC-EXOs miR-205–5p exerts inhibitory efficacy on developing hepatocellular carcinoma (HCC) via controlling CDKL3. This miRNA expression was decreased while CDKL3 was increased in HCC. BM-MSCs-EXOs inhibited the cellular growth of HCC in vitro and in vivo. Loss of CDKL3 impaired the malignant processes of HCC cells and could even disturb the pro-tumor efficacy of downregulated BM MSC-EXOs miR-205–5p [121]. Recently, researchers used AD-MSC-EXOs to carry miR-381 mimic to MDA-MB-231 cells to clarify their effectiveness on triple-negative breast cancer cells. This miRNA encapsulated within AD-MSC-EXOs considerably decreases the expression of epithelial-to-mesenchymal transition (EMT) associated genes and proteins. Remarkably, this method suppressed the proliferation, migration, and malignancy capability of MDA-MB-231 and improved their apoptosis in vitro. As a result, this type of EXOs could be applied as an effective drug delivery system for miRNAs [122]. In another study, Jiang et al. showed that Exo-miR-7-5p isolated from BM-MSCs stimulated the organization of acute myeloid leukemia (AML) cells susceptible to apoptosis and a low viability amount, with OSBPL11 expression suppressed via the PI3K/AKT/mTOR signaling pathway. This method showed tumor-homing efficacy in vitro and in vivo and stopped AML progression [123]. Kurniawati et al. used BM-MSCs-EXOs to deliver miR-let-7c for suppression of the development of Castrate-resistant prostate cancer (CRPC). This investigation showed the tumor-preventative role of miR-let-7c in inhibiting cell proliferation and migration of CRPC-like cells [124]. In another study, researchers transfected BM-MSCs with a miR-146b expression plasmid, and isolated EXOs obtained via the MSCs. Intra-tumor administration of EXOs received from miR-146-expressing MSCs remarkably decreased glioma xenograft development in an animal model of early brain malignant growth [125] (Table 1).

Table 1 MSC-EXOs as miRNAs delivery system in cancer

MSC-EXOs as miRNAs delivery system in infectious diseases

The application of MSC-EXOs in the therapy of infectious diseases is a promising host-directed therapy. MSC-EXOs have an excellent capability for the treatment of inflammatory and infectious diseases. Moreover, MSC-EXOs have analogous capabilities to their parent cells, which have an extraordinary ability to regulate immune reactions because of their curative biomolecules [126, 127]. Furthermore, protein profiling of MSC-EVs shows that exosomal proteins are associated with the biological procedures, including the innate immune system, anti-bacterial, host-virus interplay, cellular oxidant detoxification, and complementarity and clotting cascades [128]. Infectious diseases, such as viral infections are related to changed amounts of host miRNAs. The efficacy of host miRNAs on viral infection can be used per se and indirectly. The direct effectiveness of miRNAs on virus control happens via directly binding various zones of the RNA virus genome. The indirect effectiveness comprises regulating a cellular transcript encoding a host agent required for one or more stages in viral replication. However, the virus may inhibit the production of miRNAs in an antiviral reaction [4, 129, 130]. In an investigation, researchers showed that UC-MSC-EXOs suppress Hepatitis C virus (HCV) infection in vitro, particularly viral reproduction, with less cell toxicity. The results showed that miRNAs from UC-MSC-EXOs had their exclusive expression profiles, and these functional miRNAs, mainly demonstrated via let-7f, miR-145, miR-199a, and miR-221 discharged from UC-MSC-EXO, widely involved in the inhibition of HCV RNA reproduction. Furthermore, UC-MSC-EXO treatment presented synergistic results when incorporated with U.S. FDA accepted interferon-α or telaprevir (VX-950), improving their anti-HCV capability and therefore enhancing the clinical importance of these regenerative ingredients for future usage as optimal adjuvants of anti-HCV treatment [131].

Moreover, UC-MSC-EXOs were isolated and applied to the in vitro acute lung injury (ALI) model. UC-MSC-EXOs restored the impaired alveolar fluid clearance of alveolar epithelial cells induced by influenza A (H5N1) infection. Still, this change was less statistically significant than the restoration by UC-MSCs. However, EXOs prevented changes in the alveolar protein permeability similarly to that of UC-MSCs [132].

In viral pneumonia, apart from the suppression of hypercytokinemia, inhibition of viral reproduction and attack on viruses are based pathways of MSC-EXOs treatment. MSC-EXOs miRNAs might bind to the viral genome to inhibit viral RNA transcription or protein translation vital for viral reproduction [133]. Sepsis is an acute deadly factor in COVID-19, and treatment via MSC-EXOs has improved the quantity of recovery in the sepsis animal model. At the same time, MSC-EXOs also suppressed the release of pro-inflammatory factors, such as TNF-α, IFN-γ, IL-6, IL-17, and IL-1β. They augmented the discharge of anti-inflammatory factors, containing IL-4, IL-10, and TGF-β. Furthermore, MSC-EXOs reduced the number of chemokines in the serum when injected. In another investigation, researchers utilized one dose, IV injection (15 mL) of allogeneic BM-MSC-EXOs (ExoFlo) to cure severe COVID-19. Consequently, 71 percent of the patients recovered, 13% remained extremely badly ill although stable, and 16% died for causes not related to this treatment method. Furthermore, laboratory quantities showed important improvement in absolute neutrophil amount and lymphopenia, with average CD3 + , CD4 + , and CD8 + cell amounts increasing [91]. In another investigation, Li and colleagues discovered the controlling pathway of miR-133 released from BM-MSC-EXO on myocardial fibrosis and EMT in viral myocarditis (VMC) animal model by regulating mastermind-like 1 (MAML1). EXOs were isolated and attained by ultracentrifugation, which was recognized via transmission electron microscope and western blot analysis. BM-MSC-EXO enhanced miR-133a expression in VMC rats and successfully enhanced the VMC rat cardiac action and myocardial fibrosis, enhanced cardiomyocyte viability, and suppressed the EMT procedure. Enhanced miR-133a in EXOs reinforced the developments. Inhibited miR-133a efficiently reversed the efficacy of BM-MSC-EXOs on VMC rats [134].

MSC-EXOs as miRNAs delivery system in autoimmune and neurodegenerative diseases

In a study, investigators studied whether MSC-EXOs can carry miR-223-3p to remedy autoimmune hepatitis in an animal model. Researchers showed that MSC-EXOs was effectively combined with miR-223-3p and transported miR-223-3p into macrophages. Moreover, therapies of either naked EXOs or EXOs-miR-223-3p effectively decreased inflammatory reactions in the autoimmune chronic active hepatitis and IL-1, IL-6, and TNF-α discharge in both the liver and macrophages. The pathway may be associated with controlling miR-223-3p rate and STAT3 expression level in the liver and macrophages [135]. In another investigation, researchers showed that therapy of diabetic peripheral neuropathy in diabetic mice with MSC-EXOs-miR-146a for two weeks considerably augmented and reduced the nerve conduction speed and thermic and mechanical stimuli threshold, in order, while it took four weeks of EXO-naive therapy to attain this recovery. Contrasted with EXO-naïve, MSC-EXOs-146a considerably inhibited the peripheral blood inflammatory monocytes and the triggering of endothelial cells by suppressing the TLR-4/NF-κB signaling pathway [136]. BM-MSC-EXOs loaded with atorvastatin could show superior pro-angiogenic capability in diabetic wound healing. Besides, BM-MSC-EXOs-atorvastatin improved the proliferation, migration, tube organization, and VEGF rate of endothelial cells in vitro. MiR-221-3p was upregulated via atorvastatin-EXO induction, and miR-221-3p suppressor inhibited the pro-angiogenesis efficacy of atorvastatin-EXOs [137]. Furthermore, hBM-MSC-EXOs overexpressing miR-26a-5p postponed the harm of synovial fibroblasts in vitro and reduced osteoarthritis harm in vivo. Overall, this method used as an inhibitor for the damage of synovial fibroblasts by PTGS2 in osteoarthritis, which is of importance for the therapy of osteoarthritis in rats [138].

In another study, in vitro studies demonstrated that EXOs miR-146a released from BM-MSCs was delivered into astrocytic glial cells, and an enhanced rate of miR-146a and a reduced rate of NF-κB were detected in astrocytic glial cells. This investigation shows that exosomal delivery of miR-146a is implicated in the repair of cognitive disorder in an animal model of Alzheimer’s disease [139]. Qiang Li et al. used AD-MSCs-EXOs to deliver miR-188-3p for therapy-inhibited autophagy and pyroptosis while enhancing proliferation by binding to CDK5 and NLRP3 in mice and MN9D cells. It was shown that miR-188-3p could be a novel curative purpose for treating Parkinson's disease [140].

Damage to Retinal Ganglion Cells (RGC) and their axons is the primary reason for blindness. Researchers showed that MSC-EXOs were efficient in preserving RGC. And also, this method improved RGC survival and axon efficiency in the animal ocular nerve damage model while partially inhibiting RGC axon damage and dysfunction. To additionally study, the pathway of RGC preservation via MSC-EXOs, transfected MSCs with siRNA to inhibit the Argonaute-2 gene (the main miRNA effector) and separated the EXOs produced. It was found that the EXOs effectively carried their “payload” to the internal retina and that the efficacy was miRNA-related, with the curative efficacy of MSC-EXOs being decreased when Argonaute-2 was knocked out [141] (Table 2).

Table 2 MSC-EXOs as miRNAs delivery system in Autoimmune and Neurodegenerative Diseases

MSC-EXOs as miRNAs delivery system in other diseases

MSC-EXOs control vascular smooth muscle cell (VSMC) roles to suppress neointimal hyperplasia (NIH). Injection of hUC-MSC-EXOs inhibited NIH afterward artery ligation. HUC-MSC-EXOs reduced the intima and media region and intima/media proportion, enhanced the contractile phenotype protein SM22a in the media coating, and decreased Serpine1 expression in the carotid artery. MiR-148a-3p was improved in hUC-MSC-EXOs and inhibited Serpine1 via direct binding to its 3′-UTR area. Furthermore, hUC-MSC-EXOs miR-148a-3p inhibited VSMC phenotypic switching and migration via targeting Serpine1 [142]. The separated UC-MSC-EXOs had a characteristic cup-formed morphology, expressed the particular exosomal markers Alix, CD63, and TSG101, and were about 50–150 nm in size. TGFβ1 at 10 ng/ml considerably improved endometrial fibrosis, which was reversed via 20 µg/ml UC-MSC-EXOs. Exosomal miR-145-5p enhanced TGFβ1-stimulated endometrial fibrosis. UC-MSC-EXOs might reverse endometrial stromal cell fibrosis by controlling miR-145-5p/ZEB2 axis, demonstrating a possible new approach to augment endometrial regeneration [143]. In another investigation, MSCs were delivered with anti-let-7i-5p afterward, EXOs were separated and purified to carry anti-let-7i-5p oligonucleotides to suppress the rate of let-7i-5p in kidney tubular epithelial cells (NRK-52E). Furthermore, mice injected with MSC-EXOs anti-let-7i-5p exhibited decreased renal fibrosis and enhanced kidney function when challenged with a unilateral ureteral obstacle (UUO) [144]. In another study, 24 h afterward surgical treatment, BM-MSCs-EXOs-miR-146a-5p injection enhanced neurological function decreased apoptotic and neurodegenerative diseases and suppressed inflammatory reaction [145] (Table 3).

Table 3 MSCs-EXOs as miRNAs delivery system in different diseases

MSC-EXOs as miRNAs delivery system in the clinical stage

Up to now, an investigation on www.clinicaltrials.gov, which found 26 outcomes related to the keywords "Mesenchymal Stem Cells Exosomes". Moreover, clinical evaluations of MSC-EXOs are presently underway for insulin-dependent diabetes, cerebral infarction, COVID-19, Alzheimer's disease, and degenerative arthritis. However, of those studies, only one comprises what can be regarded as MSCs-EXOs as a medication transfer method: iExosomes in treating participants with metastatic pancreas cancer with KrasG12D mutation [146,147,148]. MSC-EXOs, as one of the most possible in vivo drug delivery systems, require to be additional studied and advanced via investigators [149]. For example, available pre-clinical information shows the efficient transfer of EXOs encapsulated with siRNA targeting KRASG12D resulting in tumor control in several mouse models of Pancreatic ductal adenocarcinoma (PDAC). Extensive generation of KRASG12D-siRNA loaded EXOs from MSCs will be implemented at the MD Anderson Cancer Center utilizing predefined GMP-compliant protocols. This is a single arm, single institute, phase I test assessing therapy with KRASG12D-siRNA loaded EXOs. This phase I trial investigates the most significant amount and adverse events of MSC-EXOs with KrasG12D siRNA (iExosomes) in curing patients with pancreatic malignant with KrasG12D mutation that has metastasized. iExosomes may work improved at curing patients [150]. Furthermore, a recently recorded trial (NCT04276987) aims to study the side effects and effectiveness of aerosol inhalation of allogenic AD-MSC-EXOs in patients with SARS-CoV-2 infection. The results demonstrate that a successive five days inhalation injection of clinical level hAD-MSC-EXOs up to a total quantity of 2.0 × 109 nanovesicles were possible and well tolerated in seven SARS-CoV-2 patients, with no proof of pre-identified side effects, instant clinical inconsistency, or dosage-related toxicity at each of the amounts evaluated. This harmlessness profile is followed by CT scan recovery within seven days [151].

Currently, several investigations on novel or improved techniques for injecting curative miRNAs to the body, preserved transfer in the blood, targeted delivery of miRNAs to host cells, effective absorption via host cells, and improved gene targeting in the cell. MiRNAs as therapeutic agents have seldom been implicated in these clinical phases; however, numerous investigations have been done on miRNAs in MSC-EXOs in animal models. The clinical usage of miRNAs in the therapy of EXOs isolated from MSCs is a very significant investigation direction in the future [152, 153]. For example, in a study (NCT03562715), peripheral blood EXOs miRNA136, miRNA494, and miRNA495 genes expression in contrast to UC-MSC-EXOs in patients with pregnancy-associated hypertension and toxemia were detected. According to the recognized and recorded information, MSC-EXOs are going to be excellent biological vehicles for the treatment of different diseases. Moreover, it is encouraged to explore deeper into the possibility of MSC-EXOs between COVID-19 treatment and provide efficient therapy with maximum safety [154] (Table 4).

Table 4 Different MSC-EXOs in a clinical trial

MSC-EXOs as a drug delivery system advantages and disadvantages

The use of MSC-EXOs is safe regarding adverse effects of MSC therapy, which may include consequences such as potential tumorigenesis by cell transplantation and obstruction in the distal vasculature by intravascular injection. The advantages are that MSC-EXOs can be mass-produced and sterilized via filtration and have a long shelf-life, but these properties do not extend to MSCs themselves [155].

One restriction to utilizing EXOs to their completest possible is their confined release from cells, which constitutes the main blockage to effective, mass-scale EXOs generation. This is particularly true for MSCs, which, while capable to self-renew, have restricted development ability. MSCs tolerate aging afterward only a few passages, and EXOs isolated from aged MSCs have damaged regenerative capability compared to young MSCs [156,157,158]. Consequently, improving EXO generation is vital for both allogeneic and autologous EXOs-based treatments since 1- Time limitation, because EXOs-based therapies require to be injected as rapidly as possible, especially for cancer vaccination or to decrease the immunogenic efficacy arising from mismatched allogeneic cells; 2- high amounts of EXOs are needed for EXO-based treatments in a concise time, and while upscaling of cell cultures is a selection, this method is restricted via the growth kinetics and the number of cells separated from patients; and 3- developed EXOs synthesis would allow the generation of off-the-shelf curative [158, 159]. From interior to exterior, from physical situations to biomolecular parameters, investigators have offered techniques to enhance the generation of MSC-EXOs from different aspects. However, the methods suggested by the investigators will more or less influence the biological function of MSC-EXOs. And some of the effects could be uncontrollable and unverified. In the future, investigators may require to incorporate numerous techniques to form standardized and stable quality processes to offer strategies for large-scale generation of MSC-EXOs [149]. Furthermore, by comparing standard miRNA delivery systems, EXOs have the benefits, such as 1. EXOs are natural vehicles of cell generation and have excellent biocompatibility; 2. the outside of EXOs contains several types of proteins to inhibit the phagocytosis of macrophages and present the capability for a long-term circulation; 3. EXOs can avoid diverse biological obstacles and penetrate cells in different methods, including endocytosis; 4. EXOs can be packed with miRNA medications in an endogenous approach. Thus, EXOs can fill more miRNA as a therapeutic agent via exploring the endogenous loading method; 5. EXOs have homing potentials, which can be targeted via active and passive techniques [6] (Fig. 9).

Fig. 9
figure 9

A road map for the large-scale production of various targeted miRNAs based on MSC-EXOs as a delivery system in different diseases. a Based on the type of diseases and microRNAs, we choose the appropriate and practical type of MSC (AD, BM, UC, WJ, and DP). b To load miRNAs in EXOs, two techniques have been used (cell modification for miRNA mimics and Direct loading of miRNAs). c After producing MSC-EXOs by hollow-Fiber Bioreactor, we need to purify them from other substances by tangential flow filtration (TFF) methods. d It is essential to establish proper storage methods and suitable transportation techniques for the mass production of MSC-EXOs containing miRNA

Newly, it has been demonstrated that MSC-EXOs play an essential role in MSC-mediated paracrine effects through the delivery of miRNAs. Extracellular miRNAs exist with significant consistency in several body fluids and cell culture media in the vesicle-related form [160]. In addition, targeted miRNA delivery to tumors has been researched to target particular subcellular compartments, and receptor-interceded endocytosis is the most encouraging method. As hopeful cancer treatment methods, miRNAs are hard to pass through cell membranes due to their negative charge and lipophobic property. In addition, they are simply destruction next, entering the body. As high-quality carriers, MSC-EXOs can address this concern. In a related studies, the MSC-EXOs expressing miRNAs have been highlighted as crucial vehicles for gene or drug therapy [161]. The limitations of using this method include the identification of MSC-EXOs absence of uniform international standards, the low efficiency and expensive; traditional purification techniques need a long term to extract EXO, and available kits are costly. In addition, because the quality of EXOs is highly affected by temperature and time, the storage of EXOs is challenging [162]. In addition, refining the culture situation of MSCs will remarkably affect not only the generation efficiency but also the effectiveness of MSC-EXOs in consideration of the proteomic and genomic complications of EXOs. The culture situation alone may not be sufficient to address the further restrictions on the efficacy of MSC-EXOs. EXOs isolated from different cell kinds may have preferential targeting towards some cell kinds based on their membrane combination, therefore, imparting a differential effect on body systems [163].

Conclusion

The use of EXOs as hopeful miRNAs delivery systems is remarkably associated with a trustworthy cell origin. Of the cell kinds recognized to release EXOs, the human MSC shows the most encouraging cell origin. MSC is not only a simply available cell kind that could be isolated from approximately all human tissues, and it is highly proliferative. One of the most attractive properties is the relative safety of MSCs. MSC-EXOs are therapeutically effective in animal models and display immunosuppressive action. MSC-EXOs have significant attributes that are examples of functional MSCs. Researchers showed the clinical effectiveness of MSC-EXOs for the treatment of different diseases. Currently, there are several studies are offering that MSC-EXO can be used for cancer therapy, gene therapy, medication delivery, regenerative medicine, and some other biomedical usages. Due to of some restrictions with the usage of MSCs themselves, such as controversial utilization in the presence of tumors, MSC-EXOs could be considered to be a cell-free alternative to intact MSCs. Moreover, the production of cell-based therapeutics is a challenging method. In contrast, MSC-EXOs can be simply stored at − 20 °C for six months without losing biological function. Therefore, EXO-based, cell-free therapies in regenerative medicine can be simpler to produce and prima facie safer. By EXOs, MSCs deliver their curative agents, particularly miRNAs, to target cells, and therein change gene expression and thereby improve therapeutic reaction. In addition, the results of the new miRNA study showed that some MSC-EXOs and MSCs had similar miRNA expression profiles, which is one of the causes why MSC-EXOs can replace MSCs for therapy. Present disease models are frequently mice or in vitro cell trials applied for cancer and other disease studies, which are relatively easy and absent information for comparative investigation with other disease models. Commonly, MSC-EXOs, as one of the most potential in vivo drug delivery systems, require to be additional studied and advanced via investigators.

Availability of data and materials

Not applicable.

References

  1. Sufianov A, et al., MicroRNAs as prognostic markers and therapeutic targets in gliomas. Non-coding RNA Research, 2022.

  2. Abdolahi S, et al., A review of molecular interplay between neurotrophins and miRNAs in neuropsychological disorders. Mol Neurobiol, 2022; 1–21.

  3. Zhou W, et al. New challenges for microRNAs in acute pancreatitis: progress and treatment. J Transl Med. 2022;20(1):1–14.

    Article  Google Scholar 

  4. Hu J, et al. The potential use of microRNAs as a therapeutic strategy for SARS-CoV-2 infection. Adv Virol. 2021;166(10):2649–72.

    CAS  Google Scholar 

  5. Dias TR, et al. MicroRNAs as potential tools for predicting cancer patients’ susceptibility to SARS-CoV-2 infection and vaccination response. Cells. 2022;11(15):2279.

    Article  CAS  Google Scholar 

  6. Fang Z, et al. Exosome based miRNA delivery strategy for disease treatment. Chin Chem Lett. 2022;33(4):1693–704.

    Article  CAS  Google Scholar 

  7. Bian D, et al. The application of mesenchymal stromal cells (MSCs) and their derivative exosome in skin wound healing: a comprehensive review. Stem Cell Res Ther. 2022;13(1):1–17.

    Article  Google Scholar 

  8. Cha S, et al. Clock-modified mesenchymal stromal cells therapy rescues molecular circadian oscillation and age-related bone loss via miR142-3p/Bmal1/YAP signaling axis. Cell Death Discov. 2022;8(1):1–13.

    Article  Google Scholar 

  9. Ma C, et al. Amelioration of ligamentum flavum hypertrophy using umbilical cord mesenchymal stromal cell-derived extracellular vesicles. Bioactive Mater. 2023;19:139–54.

    Article  CAS  Google Scholar 

  10. Zhang J, et al. Exosomes derived from bone marrow mesenchymal stromal cells promote remyelination and reduce neuroinflammation in the demyelinating central nervous system. Exp Neurol. 2022;347: 113895.

    Article  CAS  Google Scholar 

  11. Mukherjee A, et al, Current advances in the use of exosomes, liposomes, and bioengineered hybrid nanovesicles in cancer detection and therapy. Acta Pharmacologica Sinica, 2022: 1–18.

  12. Yu D, et al. Exosomes as a new frontier of cancer liquid biopsy. Mol Cancer. 2022;21(1):1–33.

    Article  CAS  Google Scholar 

  13. Lai RC, et al. Exosomes for drug delivery—a novel application for the mesenchymal stem cell. Biotechnol Adv. 2013;31(5):543–51.

    Article  CAS  Google Scholar 

  14. Yang, Z.-L., et al., The role of exosomes and exosomal noncoding RNAs from different cell sources in spinal cord injury. Front Cell Neurosci, 2022. 16.

  15. Xiong H, et al. Recent progress in detection and profiling of cancer cell-derived exosomes. Small. 2021;17(35):2007971.

    Article  CAS  Google Scholar 

  16. Dasgupta I, Chatterjee A. Recent advances in miRNA delivery systems. Methods Protocols. 2021;4(1):10.

    Article  CAS  Google Scholar 

  17. Jasim SA, et al. Shining the light on clinical application of mesenchymal stem cell therapy in autoimmune diseases. Stem Cell Res Ther. 2022;13(1):1–15.

    Article  Google Scholar 

  18. Sarmah D, et al, Sirtuin-1-mediated NF-κB pathway modulation to mitigate inflammasome signaling and cellular apoptosis is one of the neuroprotective effects of intra-arterial mesenchymal stem cell therapy following ischemic stroke. Stem Cell Rev Rep, 2022, 1–18

  19. Freitag J, et al, Real-world evidence of mesenchymal stem cell therapy in knee osteoarthritis: a large prospective two-year case series. Regen Med, 2022.

  20. Kouroupis D, Kaplan LD, Best TM. Human infrapatellar fat pad mesenchymal stem cells show immunomodulatory exosomal signatures. Sci Rep. 2022;12(1):1–15.

    Article  Google Scholar 

  21. Martinez-Arroyo O, et al. Mesenchymal stem cell-derived extracellular vesicles as non-coding RNA therapeutic vehicles in autoimmune diseases. Pharmaceutics. 2022;14(4):733.

    Article  CAS  Google Scholar 

  22. Wang S, et al. Targeted therapy for inflammatory diseases with mesenchymal stem cells and their derived exosomes: from basic to clinics. Int J Nanomed. 2022;17:1757.

    Article  Google Scholar 

  23. Yang Z, Li Y, Wang Z. Recent advances in the application of mesenchymal stem cell-derived exosomes for cardiovascular and neurodegenerative disease therapies. Pharmaceutics. 2022;14(3):618.

    Article  CAS  Google Scholar 

  24. Cao Q, et al, Mesenchymal stem cell-derived exosomes: toward cell-free therapeutic strategies in chronic kidney disease. Front Med, 2022. 9.

  25. Allegretta C, et al. Mesenchymal stem cell-derived extracellular vesicles and their therapeutic use in central nervous system demyelinating disorders. Int J Mol Sci. 2022;23(7):3829.

    Article  CAS  Google Scholar 

  26. Fu Y, Chen J, Huang Z. Recent progress in microRNA-based delivery systems for the treatment of human disease. ExRNA. 2019;1(1):1–14.

    Article  Google Scholar 

  27. Norouzi M, et al. Recent advances on nanomaterials-based fluorimetric approaches for microRNAs detection. Mater Sci Eng, C. 2019;104: 110007.

    Article  CAS  Google Scholar 

  28. Romero-Cordoba SL, et al. miRNA biogenesis: biological impact in the development of cancer. Cancer Biol Ther. 2014;15(11):1444–55.

    Article  CAS  Google Scholar 

  29. Vishnoi A, Rani S, MiRNA biogenesis and regulation of diseases: an overview. MicroRNA Profiling, 2017: 1–10

  30. Li M, Yu B. Recent advances in the regulation of plant miRNA biogenesis. RNA Biol. 2021;18(12):2087–96.

    Article  CAS  Google Scholar 

  31. Leitão AL, Enguita FJ. A structural view of miRNA biogenesis and function. Non-coding RNA. 2022;8(1):10.

    Article  Google Scholar 

  32. Heydarzadeh S, et al. Overview of host miRNA properties and their association with epigenetics, long non-coding RNAs, and Xeno-infectious factors. Cell Biosci. 2021;11(1):1–17.

    Article  Google Scholar 

  33. Lee Y, Im E. Regulation of miRNAs by natural antioxidants in cardiovascular diseases: Focus on SIRT1 and eNOS. Antioxidants. 2021;10(3):377.

    Article  CAS  Google Scholar 

  34. Saghazadeh A, Rezaei N. MicroRNA machinery in Parkinson’s disease: a platform for neurodegenerative diseases. Expert Rev Neurother. 2022;22(6):427–53.

    Article  CAS  Google Scholar 

  35. Mehta M, et al. Targeting respiratory diseases using miRNA inhibitor based nanotherapeutics: Current status and future perspectives. Nanomed Nanotechnol Biol Med. 2021;31:102303.

    Article  CAS  Google Scholar 

  36. Rupaimoole R, Slack FJ. MicroRNA therapeutics: towards a new era for the management of cancer and other diseases. Nat Rev Drug Discovery. 2017;16(3):203–22.

    Article  CAS  Google Scholar 

  37. Yan C, et al. Milk exosomes-mediated miR-31-5p delivery accelerates diabetic wound healing through promoting angiogenesis. Drug Deliv. 2022;29(1):214–28.

    Article  CAS  Google Scholar 

  38. Ban E, Kwon T-H, Kim A. Delivery of therapeutic miRNA using polymer-based formulation. Drug Deliv Transl Res. 2019;9(6):1043–56.

    Article  CAS  Google Scholar 

  39. Lee SWL, et al. MicroRNA delivery through nanoparticles. J Control Release. 2019;313:80–95.

    Article  CAS  Google Scholar 

  40. Ahmadzada T, Reid G, McKenzie DR. Fundamentals of siRNA and miRNA therapeutics and a review of targeted nanoparticle delivery systems in breast cancer. Biophys Rev. 2018;10(1):69–86.

    Article  CAS  Google Scholar 

  41. Yeo RWY, et al. Mesenchymal stem cell: an efficient mass producer of exosomes for drug delivery. Adv Drug Deliv Rev. 2013;65(3):336–41.

    Article  CAS  Google Scholar 

  42. Elsharkasy OM, et al. Extracellular vesicles as drug delivery systems: Why and how? Adv Drug Deliv Rev. 2020;159:332–43.

    Article  CAS  Google Scholar 

  43. Chen W-X, et al. MicroRNAs delivered by extracellular vesicles: an emerging resistance mechanism for breast cancer. Tumor Biol. 2014;35(4):2883–92.

    Article  CAS  Google Scholar 

  44. Esmaeili A, Hosseini S, BaghabanEslaminejad M. Engineered-extracellular vesicles as an optimistic tool for microRNA delivery for osteoarthritis treatment. Cell Mol Life Sci. 2021;78(1):79–91.

    Article  CAS  Google Scholar 

  45. Alzhrani GN, et al. Exosomes: isolation, characterization, and biomedical applications. Cell Biol Int. 2021;45(9):1807–31.

    Article  CAS  Google Scholar 

  46. Ahmadi M, Rezaie J. Ageing and mesenchymal stem cells derived exosomes: molecular insight and challenges. Cell Biochem Funct. 2021;39(1):60–6.

    Article  CAS  Google Scholar 

  47. Gutierrez-Millan C, et al. Advances in exosomes-based drug delivery systems. Macromol Biosci. 2021;21(1):2000269.

    Article  CAS  Google Scholar 

  48. Zhang Y, et al. Exosomes: biogenesis, biologic function and clinical potential. Cell Biosci. 2019;9(1):1–18.

    Article  Google Scholar 

  49. Kim J-A, et al. Regulation of mesenchymal stromal cells through fine tuning of canonical Wnt signaling. Stem cell research. 2015;14(3):356–68.

    Article  CAS  Google Scholar 

  50. Lai, R.C., R.W.Y. Yeo, and S.K. Lim. Mesenchymal stem cell exosomes. In: Seminars in cell & developmental biology. 2015. Elsevier, New York

  51. Magdy Beshbishy A, et al. Biogenesis, biologic function and clinical potential of exosomes in different diseases. Appl Sci. 2020;10(13):4428.

    Article  Google Scholar 

  52. Jeppesen DK, et al. Reassessment of exosome composition. Cell. 2019;177(2):428–45.

    Article  CAS  Google Scholar 

  53. Gurunathan S, et al. Review of the isolation, characterization, biological function, and multifarious therapeutic approaches of exosomes. Cells. 2019;8(4):307.

    Article  CAS  Google Scholar 

  54. Yue B, et al. Exosome biogenesis, secretion and function of exosomal miRNAs in skeletal muscle myogenesis. Cell Prolif. 2020;53(7): e12857.

    Article  CAS  Google Scholar 

  55. Hassanzadeh A, et al. Mesenchymal stem/stromal cell-derived exosomes in regenerative medicine and cancer; overview of development, challenges, and opportunities. Stem Cell Res Ther. 2021;12(1):1–22.

    Article  Google Scholar 

  56. Liu W-Z, et al. Mesenchymal stem cell-derived exosomes: therapeutic opportunities and challenges for spinal cord injury. Stem Cell Res Ther. 2021;12(1):1–15.

    Article  Google Scholar 

  57. Chen Y-S, et al. Exosomes in clinical trial and their production in compliance with good manufacturing practice. Tzu-Chi Med J. 2020;32(2):113.

    Article  CAS  Google Scholar 

  58. Antinori A, et al. Epidemiological, clinical and virological characteristics of four cases of monkeypox support transmission through sexual contact, Italy, May 2022. Eurosurveillance. 2022;27(22):2200421.

    Article  CAS  Google Scholar 

  59. Patel DB, et al. Towards rationally designed biomanufacturing of therapeutic extracellular vesicles: impact of the bioproduction microenvironment. Biotechnol Adv. 2018;36(8):2051–9.

    Article  CAS  Google Scholar 

  60. Cheng J, et al. Engineering of MSC-derived exosomes: a promising cell-free therapy for osteoarthritis. Membranes. 2022;12(8):739.

    Article  CAS  Google Scholar 

  61. Yan L, Wu X. Exosomes produced from 3D cultures of umbilical cord mesenchymal stem cells in a hollow-fiber bioreactor show improved osteochondral regeneration activity. Cell Biol Toxicol. 2020;36(2):165–78.

    Article  CAS  Google Scholar 

  62. Haraszti RA, et al. Exosomes produced from 3D cultures of MSCs by tangential flow filtration show higher yield and improved activity. Mol Ther. 2018;26(12):2838–47.

    Article  CAS  Google Scholar 

  63. Zhang Y, et al. Exosomes derived from 3D-cultured MSCs improve therapeutic effects in periodontitis and experimental colitis and restore the Th17 cell/Treg balance in inflamed periodontium. Int J Oral Sci. 2021;13(1):1–15.

    Article  Google Scholar 

  64. Cao J, et al. Three-dimensional culture of MSCs produces exosomes with improved yield and enhanced therapeutic efficacy for cisplatin-induced acute kidney injury. Stem Cell Res Ther. 2020;11(1):1–13.

    Article  Google Scholar 

  65. Zhang Y, et al. Systemic administration of cell-free exosomes generated by human bone marrow derived mesenchymal stem cells cultured under 2D and 3D conditions improves functional recovery in rats after traumatic brain injury. Neurochem Int. 2017;111:69–81.

    Article  CAS  Google Scholar 

  66. Gobin J, et al. Hollow-fiber bioreactor production of extracellular vesicles from human bone marrow mesenchymal stromal cells yields nanovesicles that mirrors the immuno-modulatory antigenic signature of the producer cell. Stem Cell Res Ther. 2021;12(1):1–20.

    Article  Google Scholar 

  67. Park JE, et al. Hypoxia-induced tumor exosomes promote M2-like macrophage polarization of infiltrating myeloid cells and microRNA-mediated metabolic shift. Oncogene. 2019;38(26):5158–73.

    Article  CAS  Google Scholar 

  68. Ebrahim AS, et al. Application of a flow-based hollow-Fiber co-culture system to study cellular influences under hyperglycemic conditions. Sci Rep. 2019;9(1):1–8.

    Article  CAS  Google Scholar 

  69. Kronstadt SM, et al, Mesenchymal stem cell culture within perfusion bioreactors incorporating 3D-printed scaffolds enables improved extracellular vesicle yield with preserved bioactivity. bioRxiv, 2022.

  70. Phelps J, et al. Production of mesenchymal progenitor cell-derived extracellular vesicles in suspension bioreactors for use in articular cartilage repair. Stem Cells Transl Med. 2022;11(1):73–87.

    Article  Google Scholar 

  71. Vakhshiteh F, Atyabi F, Ostad SN. Mesenchymal stem cell exosomes: a two-edged sword in cancer therapy. Int J Nanomed. 2019;14:2847.

    Article  CAS  Google Scholar 

  72. Zitvogel L, et al. Eradication of established murine tumors using a novel cell-free vaccine: dendritic cell derived exosomes. Nat Med. 1998;4(5):594–600.

    Article  CAS  Google Scholar 

  73. Chopra N, et al. Biophysical characterization and drug delivery potential of exosomes from human Wharton’s jelly-derived mesenchymal stem cells. ACS Omega. 2019;4(8):13143–52.

    Article  CAS  Google Scholar 

  74. Tang Y, Zhou Y, Li H-J. Advances in mesenchymal stem cell exosomes: a review. Stem Cell Res Ther. 2021;12(1):1–12.

    Article  Google Scholar 

  75. Lai RC, et al. Mesenchymal stem cell exosomes: the future MSC-based therapy? In: Mesenchymal stem cell therapy. Springer; 2013. p. 39–61.

    Chapter  Google Scholar 

  76. Shirejini SZ, Inci F, The Yin and Yang of exosome isolation methods: Conventional practice, microfluidics, and commercial kits. Biotechnol Adv, 2021: 107814.

  77. Willis GR, Kourembanas S, Mitsialis SA. Toward exosome-based therapeutics: isolation, heterogeneity, and fit-for-purpose potency. Front Cardiovasc Med. 2017;4:63.

    Article  Google Scholar 

  78. Lee JH, et al. Reproducible large-scale isolation of exosomes from adipose tissue-derived mesenchymal stem/stromal cells and their application in acute kidney injury. Int J Mol Sci. 2020;21(13):4774.

    Article  CAS  Google Scholar 

  79. Takayama Y, et al. Anticancer drug-loaded mesenchymal stem cells for targeted cancer therapy. J Control Release. 2021;329:1090–101.

    Article  CAS  Google Scholar 

  80. Zhang S, et al. Application of mesenchymal stem cell exosomes and their drug-loading systems in acute liver failure. J Cell Mol Med. 2020;24(13):7082–93.

    Article  Google Scholar 

  81. Baek G, et al. Mesenchymal stem cell-derived extracellular vesicles as therapeutics and as a drug delivery platform. Stem Cells Transl Med. 2019;8(9):880–6.

    Article  Google Scholar 

  82. Antimisiaris SG, Mourtas S, Marazioti A. Exosomes and exosome-inspired vesicles for targeted drug delivery. Pharmaceutics. 2018;10(4):218.

    Article  CAS  Google Scholar 

  83. Alvarez-Erviti L, et al. Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes. Nat Biotechnol. 2011;29(4):341–5.

    Article  CAS  Google Scholar 

  84. Kim MS, et al. Development of exosome-encapsulated paclitaxel to overcome MDR in cancer cells. Nanomed Nanotechnol Biol Med. 2016;12(3):655–64.

    Article  CAS  Google Scholar 

  85. Rao D, et al, Advances in mesenchymal stem cell-derived exosomes as drug delivery vehicles. Front Bioeng Biotechnol, 2021: 9.

  86. Ma Z-J, et al. Mesenchymal stem cell-derived exosomes: Toward cell-free therapeutic strategies in regenerative medicine. World J Stem Cells. 2020;12(8):814.

    Article  Google Scholar 

  87. Zhao Y, et al. MSCs-derived exosomes attenuate acute brain injury and inhibit microglial inflammation by reversing CysLT2R-ERK1/2 mediated microglia M1 polarization. Neurochem Res. 2020;45(5):1180–90.

    Article  CAS  Google Scholar 

  88. Nasirishargh A, et al. Exosomal microRNAs from mesenchymal stem/stromal cells: biology and applications in neuroprotection. World J Stem Cells. 2021;13(7):776.

    Article  Google Scholar 

  89. Foo JB, et al. Mesenchymal stem cell-derived exosomes and micrornas in cartilage regeneration: biogenesis, efficacy, mirna enrichment and delivery. Pharmaceuticals. 2021;14(11):1093.

    Article  CAS  Google Scholar 

  90. Kurtz A. Mesenchymal stem cell delivery routes and fate. Int J Stem Cells. 2008;1(1):1.

    Article  Google Scholar 

  91. Yasamineh S, et al, A state-of-the-art review on the recent advances of niosomes as a targeted drug delivery system. Int J Pharmaceut, 2022: 121878

  92. Lou G, et al. Mesenchymal stem cell-derived exosomes as a new therapeutic strategy for liver diseases. Exp Mol Med. 2017;49(6):e346–e346.

    Article  Google Scholar 

  93. Charles CJ, et al, Systemic mesenchymal stem cell-derived exosomes reduce myocardial infarct size: characterization with MRI in a porcine model. Front Cardiovasc Med, 2020; 258

  94. Al-Khawaga S, Abdelalim EM. Potential application of mesenchymal stem cells and their exosomes in lung injury: an emerging therapeutic option for COVID-19 patients. Stem Cell Res Ther. 2020;11(1):1–33.

    Article  Google Scholar 

  95. Yin K, Wang S, Zhao RC. Exosomes from mesenchymal stem/stromal cells: a new therapeutic paradigm. Biomarker Res. 2019;7(1):1–8.

    Article  CAS  Google Scholar 

  96. Zhou Y, et al. Combined topical and systemic administration with human adipose-derived mesenchymal stem cells (hADSC) and hADSC-derived exosomes markedly promoted cutaneous wound healing and regeneration. Stem Cell Res Ther. 2021;12(1):1–12.

    Article  Google Scholar 

  97. Nojehdehi S, et al. Immunomodulatory effects of mesenchymal stem cell–derived exosomes on experimental type-1 autoimmune diabetes. J Cell Biochem. 2018;119(11):9433–43.

    Article  CAS  Google Scholar 

  98. Zhang S, et al. Exosomes derived from human embryonic mesenchymal stem cells promote osteochondral regeneration. Osteoarthritis Cartilage. 2016;24(12):2135–40.

    Article  CAS  Google Scholar 

  99. Wong KL, et al. Intra-articular injections of mesenchymal stem cell exosomes and hyaluronic acid improve structural and mechanical properties of repaired cartilage in a rabbit model. Arthroscopy J Arthrosc Relat Surg. 2020;36(8):2215–28.

    Article  Google Scholar 

  100. Betzer O, et al. In vivo neuroimaging of exosomes using gold nanoparticles. ACS Nano. 2017;11(11):10883–93.

    Article  CAS  Google Scholar 

  101. Lai CP, et al. Dynamic biodistribution of extracellular vesicles in vivo using a multimodal imaging reporter. ACS Nano. 2014;8(1):483–94.

    Article  CAS  Google Scholar 

  102. Zhang K, et al. Topical application of exosomes derived from human umbilical cord mesenchymal stem cells in combination with sponge spicules for treatment of photoaging. Int J Nanomed. 2020;15:2859.

    Article  CAS  Google Scholar 

  103. Golchin A, et al. The clinical trials of mesenchymal stem cell therapy in skin diseases: an update and concise review. Curr Stem Cell Res Ther. 2019;14(1):22–33.

    Article  CAS  Google Scholar 

  104. Ortega A, et al. Exosomes as drug delivery systems: endogenous nanovehicles for treatment of systemic lupus erythematosus. Pharmaceutics. 2020;13(1):3.

    Article  Google Scholar 

  105. Aqil F, et al. Exosomes for the enhanced tissue bioavailability and efficacy of curcumin. AAPS J. 2017;19(6):1691–702.

    Article  CAS  Google Scholar 

  106. Nazimek K, et al. Orally administered exosomes suppress mouse delayed-type hypersensitivity by delivering miRNA-150 to antigen-primed macrophage APC targeted by exosome-surface anti-peptide antibody light chains. Int J Mol Sci. 2020;21(15):5540.

    Article  CAS  Google Scholar 

  107. Harrell CR, et al. Therapeutic potential of mesenchymal stem cell-derived exosomes in the treatment of eye diseases. Cell Biol Transl Med. 2018;2:47–57.

    Google Scholar 

  108. Nuzzi R, Caselgrandi P, Vercelli A, Effect of mesenchymal stem cell-derived exosomes on retinal injury: a review of current findings. Stem Cells Int, 2020. 2020.

  109. Cui Y, et al. Protective effects of intravitreal administration of mesenchymal stem cell-derived exosomes in an experimental model of optic nerve injury. Exp Cell Res. 2021;407(1): 112792.

    Article  CAS  Google Scholar 

  110. Álvarez-Viejo M. Mesenchymal stem cells from different sources and their derived exosomes: a pre-clinical perspective. World J Stem Cells. 2020;12(2):100.

    Article  Google Scholar 

  111. Zhang L, et al. Exosomes from bone marrow mesenchymal stem cells enhance fracture healing through the promotion of osteogenesis and angiogenesis in a rat model of nonunion. Stem Cell Res Ther. 2020;11(1):1–15.

    Google Scholar 

  112. Jiang T, Wang Z, Sun J. Human bone marrow mesenchymal stem cell-derived exosomes stimulate cutaneous wound healing mediates through TGF-β/Smad signaling pathway. Stem Cell Res Ther. 2020;11(1):1–10.

    Article  Google Scholar 

  113. Ahani-Nahayati M, et al. Umbilical cord mesenchymal stem/stromal cells potential to treat organ disorders; an emerging strategy. Curr Stem Cell Res Ther. 2022;17(2):126–46.

    Article  CAS  Google Scholar 

  114. Yin S, et al. Human umbilical cord mesenchymal stem cells and exosomes: bioactive ways of tissue injury repair. Am J Transl Res. 2019;11(3):1230.

    CAS  Google Scholar 

  115. Navajas R, Corrales FJ, Paradela A. Serum exosome isolation by size-exclusion chromatography for the discovery and validation of preeclampsia-associated biomarkers. In: Proteomics for Biomarker Discovery. Springer; 2019. p. 39–50.

    Chapter  Google Scholar 

  116. Liu M, et al, Exosomes derived from adipose-derived mesenchymal stem cells ameliorate radiation-induced brain injury by activating the sirt1 pathway. Front Cell Dev Biol, 2021: 2083

  117. Taghavi-Farahabadi M, et al. Wharton’s Jelly Mesenchymal Stem cells exosomes and conditioned media increased Neutrophil lifespan and phagocytosis capacity. Immunol Invest. 2021;50(8):1042–57.

    Article  CAS  Google Scholar 

  118. Zhang F, et al. Mesenchymal stem cell-derived exosome: A tumor regulator and carrier for targeted tumor therapy. Cancer Lett. 2022;526:29–40.

    Article  CAS  Google Scholar 

  119. Sohrabi B, et al, Mesenchymal stem cell (msc)-derived exosomes as novel vehicles for delivery of mirnas in cancer therapy. Cancer Gene Therapy, 2022: 1–12.

  120. Yassine S, Alaaeddine N. Mesenchymal stem cell exosomes and cancer: controversies and prospects. Adv Biol. 2022;6(2):2101050.

    Article  Google Scholar 

  121. Sun Q, et al. Bone marrow mesenchymal stem cells-secreted exosomal microRNA-205–5p exerts inhibitory effect on the progression of liver cancer through regulating CDKL3. Pathol Res Pract. 2021;225: 153549.

    Article  CAS  Google Scholar 

  122. Shojaei S, et al. Delivery of miR-381-3p mimic by mesenchymal stem cell-derived exosomes inhibits triple negative breast cancer aggressiveness; an in vitro study. Stem Cell Rev Rep. 2021;17(3):1027–38.

    Article  CAS  Google Scholar 

  123. Jiang D, et al. Bone mesenchymal stem cell-derived exosomal microRNA-7-5p inhibits progression of acute myeloid leukemia by targeting OSBPL11. J Nanobiotechnol. 2022;20(1):1–19.

    Article  Google Scholar 

  124. Kurniawati I, et al. Targeting castration-resistant prostate cancer using mesenchymal stem cell exosomes for therapeutic MicroRNA-let-7c delivery. Front Biosci Landmark. 2022;27(9):256.

    Article  Google Scholar 

  125. Katakowski M, et al. Exosomes from marrow stromal cells expressing miR-146b inhibit glioma growth. Cancer Lett. 2013;335(1):201–4.

    Article  CAS  Google Scholar 

  126. Zohrabi M, et al, Potential of mesenchymal stem cell-derived exosomes as a novel treatment for female infertility caused by bacterial infections. Front Microbiol, 2022: 4115.

  127. Doyle LM, Wang MZ. Overview of extracellular vesicles, their origin, composition, purpose, and methods for exosome isolation and analysis. Cells. 2019;8(7):727.

    Article  CAS  Google Scholar 

  128. Pierce LM, Kurata WE. Priming with toll-like receptor 3 agonist poly (I: C) enhances content of innate immune defense proteins but not MicroRNAs in human mesenchymal stem cell-derived extracellular vesicles. Front Cell Dev Biol. 2021;9: 676356.

    Article  Google Scholar 

  129. Chen L, Zhou Y, Li H. LncRNA, miRNA and lncRNA-miRNA interaction in viral infection. Virus Res. 2018;257:25–32.

    Article  CAS  Google Scholar 

  130. Girardi E, López P, Pfeffer S. On the importance of host microRNAs during viral infection. Front Genet. 2018;9:439.

    Article  CAS  Google Scholar 

  131. Qian X, et al. Exosomal microRNAs derived from umbilical mesenchymal stem cells inhibit hepatitis C virus infection. Stem Cells Transl Med. 2016;5(9):1190–203.

    Article  CAS  Google Scholar 

  132. Loy H, et al. Therapeutic implications of human umbilical cord mesenchymal stromal cells in attenuating influenza A (H5N1) virus–associated acute lung injury. J Infect Dis. 2019;219(2):186–96.

    Article  CAS  Google Scholar 

  133. Wu X, et al, Mesenchymal stem cell-derived exosome therapy of microbial diseases: from bench to bed. Front Microbiol, 2021. 12.

  134. Lin N, et al. Antiviral nanoparticles for sanitizing surfaces: a roadmap to self-sterilizing against COVID-19. Nano Today. 2021;40: 101267.

    Article  CAS  Google Scholar 

  135. Lu F-B, et al. Attenuation of experimental autoimmune hepatitis in mice with bone mesenchymal stem cell-derived exosomes carrying microRNA-223-3p. Mol Cells. 2019;42(12):906.

    CAS  Google Scholar 

  136. Fan B, et al. Treatment of diabetic peripheral neuropathy with engineered mesenchymal stromal cell-derived exosomes enriched with microRNA-146a provide amplified therapeutic efficacy. Exp Neurol. 2021;341: 113694.

    Article  CAS  Google Scholar 

  137. Yu M, et al. Exosomes derived from atorvastatin-pretreated MSC accelerate diabetic wound repair by enhancing angiogenesis via AKT/eNOS pathway. Stem Cell Res Ther. 2020;11(1):1–17.

    Article  CAS  Google Scholar 

  138. Jin Z, Ren J, Qi S. Human bone mesenchymal stem cells-derived exosomes overexpressing microRNA-26a-5p alleviate osteoarthritis via down-regulation of PTGS2. Int Immunopharmacol. 2020;78: 105946.

    Article  CAS  Google Scholar 

  139. Nakano M, et al. Bone marrow-derived mesenchymal stem cells improve cognitive impairment in an Alzheimer’s disease model by increasing the expression of microRNA-146a in hippocampus. Sci Rep. 2020;10(1):1–15.

    Article  Google Scholar 

  140. Li Q, et al. Exosomes derived from miR-188-3p-modified adipose-derived mesenchymal stem cells protect Parkinson’s disease. Mol Therapy-Nucl Acids. 2021;23:1334–44.

    Article  CAS  Google Scholar 

  141. Mead B, Tomarev S. Bone marrow-derived mesenchymal stem cells-derived exosomes promote survival of retinal ganglion cells through miRNA-dependent mechanisms. Stem Cells Transl Med. 2017;6(4):1273–85.

    Article  CAS  Google Scholar 

  142. Zhang X, et al. Human umbilical cord mesenchymal stem cell-derived exosomal microRNA-148a-3p inhibits neointimal hyperplasia by targeting Serpine1. Arch Biochem Biophys. 2022;719: 109155.

    Article  CAS  Google Scholar 

  143. Rizk JG, et al. Prevention and treatment of Monkeypox. Drugs. 2022;82(9):957–63.

    Article  CAS  Google Scholar 

  144. Jin J, et al. Mesenchymal stem cells attenuate renal fibrosis via exosomes-mediated delivery of microRNA Let-7i-5p antagomir. Int J Nanomed. 2021;16:3565.

    Article  Google Scholar 

  145. Duan S, et al. Exosomes derived from MicroRNA-146a-5p-enriched bone marrow mesenchymal stem cells alleviate intracerebral hemorrhage by inhibiting neuronal apoptosis and microglial M1 polarization. Drug Des Dev Ther. 2020;14:3143.

    Article  CAS  Google Scholar 

  146. Massa M, et al. Clinical applications of mesenchymal stem/stromal cell derived extracellular vesicles: therapeutic potential of an acellular product. Diagnostics. 2020;10(12):999.

    Article  CAS  Google Scholar 

  147. Huda MN, et al. Potential use of exosomes as diagnostic biomarkers and in targeted drug delivery: progress in clinical and preclinical applications. ACS Biomater Sci Eng. 2021;7(6):2106–49.

    Article  CAS  Google Scholar 

  148. Harrell CR, et al. Mesenchymal stem cell-derived exosomes and other extracellular vesicles as new remedies in the therapy of inflammatory diseases. Cells. 2019;8(12):1605.

    Article  CAS  Google Scholar 

  149. Sun Y, et al. Mesenchymal stem cells-derived exosomes for drug delivery. Stem Cell Res Ther. 2021;12(1):1–15.

    Article  Google Scholar 

  150. Surana R, et al, Phase I study of mesenchymal stem cell (MSC)-derived exosomes with KRASG12D siRNA in patients with metastatic pancreatic cancer harboring a KRASG12D mutation. 2022, American Society of Clinical Oncology.

  151. Zhu Y-G, et al. Nebulized exosomes derived from allogenic adipose tissue mesenchymal stromal cells in patients with severe COVID-19: a pilot study. Stem Cell Res Ther. 2022;13(1):1–10.

    Article  Google Scholar 

  152. Xu H-K, et al. Multifunctional role of microRNAs in mesenchymal stem cell-derived exosomes in treatment of diseases. World J Stem Cells. 2020;12(11):1276.

    Article  Google Scholar 

  153. Diener C, Keller A, Meese E, Emerging concepts of miRNA therapeutics: From cells to clinic. Trends Genet, 2022.

  154. Janockova J, et al. New therapeutic approaches of mesenchymal stem cells-derived exosomes. J Biomed Sci. 2021;28(1):1–26.

    Article  Google Scholar 

  155. Ha DH, et al. Toxicological evaluation of exosomes derived from human adipose tissue-derived mesenchymal stem/stromal cells. Regul Toxicol Pharmacol. 2020;115: 104686.

    Article  CAS  Google Scholar 

  156. Chen TS, et al. Enabling a robust scalable manufacturing process for therapeutic exosomes through oncogenic immortalization of human ESC-derived MSCs. J Transl Med. 2011;9(1):1–10.

    Article  Google Scholar 

  157. Severino V, et al. Insulin-like growth factor binding proteins 4 and 7 released by senescent cells promote premature senescence in mesenchymal stem cells. Cell Death Dis. 2013;4(11):e911–e911.

    Article  CAS  Google Scholar 

  158. Wang J, et al. Boosting the biogenesis and secretion of mesenchymal stem cell-derived exosomes. Cells. 2020;9(3):660.

    Article  CAS  Google Scholar 

  159. Patel DB, et al. Enhanced extracellular vesicle production and ethanol-mediated vascularization bioactivity via a 3D-printed scaffold-perfusion bioreactor system. Acta Biomater. 2019;95:236–44.

    Article  CAS  Google Scholar 

  160. Gong M, et al. Mesenchymal stem cells release exosomes that transfer miRNAs to endothelial cells and promote angiogenesis. Oncotarget. 2017;8(28):45200.

    Article  Google Scholar 

  161. Sun Z, et al, Roles of mesenchymal stem cell-derived exosomes in cancer development and targeted therapy. Stem Cells Int, 2021. 2021.

  162. Jafari D, et al. Designer exosomes: a new platform for biotechnology therapeutics. BioDrugs. 2020;34(5):567–86.

    Article  Google Scholar 

  163. Phan J, et al. Engineering mesenchymal stem cells to improve their exosome efficacy and yield for cell-free therapy. J Extracell Vesicles. 2018;7(1):1522236.

    Article  CAS  Google Scholar 

  164. Sidhom K, Obi PO, Saleem A. A review of exosomal isolation methods: is size exclusion chromatography the best option? Int J Mol Sci. 2020;21(18):6466.

    Article  CAS  Google Scholar 

  165. Contreras-Naranjo JC, Wu H-J, Ugaz VM. Microfluidics for exosome isolation and analysis: enabling liquid biopsy for personalized medicine. Lab Chip. 2017;17(21):3558–77.

    Article  CAS  Google Scholar 

  166. Liu T, et al, Evaluating adipose‐derived stem cell exosomes as miRNA drug delivery systems for the treatment of bladder cancer. Cancer Med 2022.

  167. Xu Y, et al. Human umbilical cord mesenchymal stem cells-derived exosomal microRNA-451a represses epithelial–mesenchymal transition of hepatocellular carcinoma cells by inhibiting ADAM10. RNA Biol. 2021;18(10):1408–23.

    Article  CAS  Google Scholar 

  168. Huang H, et al. Human umbilical cord-mesenchymal stem cells-derived exosomes carrying microRNA-15a-5p possess therapeutic effects on Wilms tumor via regulating septin 2. Bioengineered. 2022;13(3):6136–48.

    Article  CAS  Google Scholar 

  169. Naseri Z, et al. Exosome-mediated delivery of functionally active miRNA-142-3p inhibitor reduces tumorigenicity of breast cancer in vitro and in vivo. Int J Nanomed. 2018;13:7727.

    Article  CAS  Google Scholar 

  170. Vakhshiteh F, et al. Exosomes derived from miR-34a-overexpressing mesenchymal stem cells inhibit in vitro tumor growth: A new approach for drug delivery. Life Sci. 2021;266: 118871.

    Article  CAS  Google Scholar 

  171. Munoz JL, et al. Delivery of functional anti-miR-9 by mesenchymal stem cell–derived exosomes to glioblastoma multiforme cells conferred chemosensitivity. Mol Therapy Nucl Acids. 2013;2: e126.

    Article  Google Scholar 

  172. Lv Q, et al. Engineered human adipose stem-cell-derived exosomes loaded with miR-21-5p to promote diabetic cutaneous wound healing. Mol Pharm. 2020;17(5):1723–33.

    Article  CAS  Google Scholar 

  173. Wei H, et al. Mesenchymal stem cell-derived exosomal miR-223 regulates neuronal cell apoptosis. Cell Death Dis. 2020;11(4):1–11.

    Article  Google Scholar 

  174. Tao S-C, et al. Exosomes derived from miR-140-5p-overexpressing human synovial mesenchymal stem cells enhance cartilage tissue regeneration and prevent osteoarthritis of the knee in a rat model. Theranostics. 2017;7(1):180.

    Article  CAS  Google Scholar 

  175. Li Q, et al. Bone marrow mesenchymal stem cell-derived exosomal MicroRNA-133a restrains myocardial fibrosis and epithelial–mesenchymal transition in viral myocarditis rats through suppressing MAML1. Nanoscale Res Lett. 2021;16(1):1–17.

    Article  Google Scholar 

  176. Facility S-FH, Samara regional medical center dinasty. Evaluation of safety and efficiency of method of exosome inhalation in SARS-CoV-2 associated pneumonia; (COVID-19EXO); ClinicalTrials. gov Identifier: NCT04491240.

  177. Wang G, Gao X, Hospital R, The safety and the efficacy evaluation of allogenic adipose MSC-Exos in patients with Alzheimer's disease. Identifier NCT04388982, 2020.

  178. Zhang X, et al. Effects of mesenchymal stem cells and their exosomes on the healing of large and refractory macular holes. Graefes Arch Clin Exp Ophthalmol. 2018;256(11):2041–52.

    Article  CAS  Google Scholar 

  179. Özmert E, Arslan U. Management of retinitis pigmentosa by Wharton’s jelly-derived mesenchymal stem cells: prospective analysis of 1-year results. Stem Cell Res Ther. 2020;11(1):1–17.

    Google Scholar 

  180. Sun Y, et al. Human mesenchymal stem cell derived exosomes alleviate type 2 diabetes mellitus by reversing peripheral insulin resistance and relieving β-cell destruction. ACS Nano. 2018;12(8):7613–28.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

The authors would like to thank the Department of Medical Biotechnology and Department of Bacteriology and Department of Virology, Tabriz University of Medical Sciences for supporting this project.

Funding

There is no Funding.

Author information

Authors and Affiliations

Authors

Contributions

S.Y., O.GH.: Writing–original draft. S.V., E.O., H.B., Y.E., E.M.: conceptualization, supervision, writing – review & editing. All authors participated in the manuscript in the critical review process of the manuscript and approved the final version.

Corresponding authors

Correspondence to Saman Yasamineh or Omid Gholizadeh.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Oveili, E., Vafaei, S., Bazavar, H. et al. The potential use of mesenchymal stem cells-derived exosomes as microRNAs delivery systems in different diseases. Cell Commun Signal 21, 20 (2023). https://doi.org/10.1186/s12964-022-01017-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12964-022-01017-9

Keywords