Skip to main content

RETRACTED ARTICLE: Mesenchymal stem/stromal cells as a valuable source for the treatment of immune-mediated disorders

This article was retracted on 22 April 2024

This article has been updated

Abstract

Over recent years, mesenchymal stem/stromal cells (MSCs) and their potential biomedical applications have received much attention from the global scientific community in an increasing manner. Firstly, MSCs were successfully isolated from human bone marrow (BM), but in the next steps, they were also extracted from other sources, mostly from the umbilical cord (UC) and adipose tissue (AT). The International Society for Cellular Therapy (ISCT) has suggested minimum criteria to identify and characterize MSCs as follows: plastic adherence, surface expression of CD73, D90, CD105 in the lack of expression of CD14, CD34, CD45, and human leucocyte antigen-DR (HLA-DR), and also the capability to differentiate to multiple cell types including adipocyte, chondrocyte, or osteoblast in vitro depends on culture conditions. However, these distinct properties, including self-renewability, multipotency, and easy accessibility are just one side of the coin; another side is their huge secretome which is comprised of hundreds of mediators, cytokines, and signaling molecules and can effectively modulate the inflammatory responses and control the infiltration process that finally leads to a regulated tissue repair/healing or regeneration process. MSC-mediated immunomodulation is a direct result of a harmonic synergy of MSC-released signaling molecules (i.e., mediators, cytokines, and chemokines), the reaction of immune cells and other target cells to those molecules, and also feedback in the MSC-molecule-target cell axis. These features make MSCs a respectable and eligible therapeutic candidate to be evaluated in immune-mediated disorders, such as graft versus host diseases (GVHD), multiple sclerosis (MS), Crohn’s disease (CD), and osteoarthritis (OA), and even in immune-dysregulating infectious diseases such as the novel coronavirus disease 2019 (COVID-19). This paper discussed the therapeutic applications of MSC secretome and its biomedical aspects related to immune-mediated conditions. Sources for MSC extraction, their migration and homing properties, therapeutic molecules released by MSCs, and the pathways and molecular mechanisms possibly involved in the exceptional immunoregulatory competence of MSCs were discussed. Besides, the novel discoveries and recent findings on immunomodulatory plasticity of MSCs, clinical applications, and the methods required for their use as an effective therapeutic option in patients with immune-mediated/immune-dysregulating diseases were highlighted.

Introduction

As known, mesenchymal stem/stromal cells (MSCs) are the plastic adherent spindle-shaped cells isolated from bone marrow (BM), adipose tissue (AT), umbilical cord (UC), and other tissue sources showing multipotent differentiation characteristic in vitro [1]. For the first time, MSCs were isolated from murine BM by Friendenstein et al. and were termed as hematopoiesis-supporting cells in BM [2]. They showed that these cells were separate from the hematopoietic cells because of dissimilarities in the capability to adhere to the tissue culture vessels and the fibroblast-like morphology of their progeny in culture [2, 3]. Friendenstein et al. offered a prominent advance by demonstrating that the expansion of BM cell suspensions at clonal density led to the creation of separate colonies that originated from single cells (the colony-forming unit-fibroblasts, CFU-Fs) [4]. Though exclusive characteristics quoted for MSCs vary among specialists due to the lack of a generally accepted surface marker phenotype, all suggested that MSC’s populations show plastic adherent property along with the expression of CD73, CD90, and CD105 in the absence of hematopoietic markers, most importantly, CD45, CD34, CD14, CD19, and CD3 [5]. Moreover, MSCs can give rise to three mesodermal lineages osteoblasts, adipocytes, and chondrocytes in vitro [6]. The minimal criteria provided by the International Society for Cellular Therapy (ISCT) could be applied to all types of MSCs, while some discrepancy has been reported. During the last decades, MSC’s particular possessions, such as self-renewal, multipotency, accessibility, less ethical concerns, and immunomodulatory attributes, have emphasized their importance in stem cell-based therapies and regenerative medicine [7]. They can expand ex vivo in culture upon procurement and differentiate into osteogenic, chondrogenic, adipogenic, and myogenic cells and other lineages for repair and recovery of target tissues [8]. Interestingly, given the unique immunomodulatory competence of MSC, which are predominantly exerted by a synergy of cell contact-dependent processes and soluble factors, they attracted increasing attention in enabling tissue regeneration and homeostasis in immunological disorders, such as graft versus host diseases (GVHD), multiple sclerosis (MS), inflammatory lung and musculoskeletal disorders, and Crohn’s disease (CD) [9, 10]. A variety of studies on animal models of immune-mediated disorders have evidenced that MSCs are capable of survival and interfere with the growth, activation, and function of immune cells following transplantation. For example, MSCs inhibited the proliferation and infiltration of immune cells into the skin through reduction of CCR4 and CCR8 expression on CD4-positive T cells and CCR1 on CD11b-positive monocyte/macrophages cells [11] concomitant with a decrease in expression of chemokines such as CCL1, CCL3, CCL8, CCL17, and CCL22 in skin resulted in alleviated cutaneous sclerodermatous GVHD in rodent models [12]. Furthermore, the MSC secretome includes cytokines, chemokines, microRNAs (miRNAs), growth factors, and proteins which can signify a reasonable alternative to their application [13]. Now, there exists robust evidence supporting the hypothesis that proximity of MSCs from adjacent tissues is not required as their soluble trophic factors are conveyed to the target tissues, allowing their repair and hemostasis [14]. Thereby, the use of MSC secretome encompassing exosomes and microvesicles (MV), generally known as extracellular vesicles (EVs), can be considered a rational and practical therapeutic strategy to treat immunological disorders. Compared to their parent MSCs, EVs expose a higher safety profile and can be safely kept without losing their functional activities [15]. The exosomes are significantly complicated in cell communication and immunomodulatory functions [16]. They are nano-sized (30–100 nm) lipid-bilayer membrane vesicles produced by inward budding of the intracellular endosomal membrane upon the formation of multivesicular bodies (MVBs) and are identified in different body fluids [17,18,19]. Also, MVs size usually ranges from 100 nm to 1 μm secreted through direct plasma membrane budding [20].

In this review, a brief overview of MSC sources, migration process, and unique immunomodulatory attribute’s mechanisms was provided while focusing on the current findings on immunoregulatory plasticity of MSCs which contribute to the regulation of immune response to elicit the desired therapeutic outcomes in patients suffering from immune-mediated/immune-dysregulating diseases.

Sources of mesenchymal stem/stromal cells (MSCs)

Mesenchymal stem/stromal cells (MSCs) can be isolated from multiple human tissues, implying the significance of the selection of more appropriated sources concerning their logistical, practical, in vitro characteristics, target tissue, and therapeutic goal [21, 22]. Today, the major and most well-known sources of MSCs are BM, AT, and UC; however, they can be isolated from dental pulps (DP), endometrium, peripheral blood (PB), skin, placenta (PL), synovial fluid (SF), muscle, Wharton’s jelly (WJ), etc. [4]. MSCs can supposedly be isolated from any human tissue, while there exist concrete restrictions based on the availability of source tissues and invasiveness of the isolation procedures and also different donor’s features. It is of paramount importance to select a fitting cell source, evaluate the difficulty of samples procurement process, and consider the possible untoward effects of collecting cells from donors [23, 24]. For instance, obtaining MSCs from BM can result in pain, bleeding, or infection, thereby making it more challenging than isolation from PB or surgical remnants (e.g., AT, DP, and UC) [25]. There are some differences in terms of marker expression, proliferation and differentiation potential, clonality, and paracrine activities among cells from various sources. In this regard, UC-MSCs displayed a more significant rate of cell proliferation and clonality in association with lower expression of p53, p21, and p16 compared to the cells isolated from BM and AT. Furthermore, UC-MSCs showed more prominent inhibitory effects on serum levels of the IL-1α, IL-6, and IL-8 in lipopolysaccharides (LPS)-treated rats compared to AT-MSCs and UC-MSCs [26]. On the other hand, MSCs derived from human placenta (PL-MSCs) demonstrated exclusive proteome profiles and revealed higher therapeutic efficacy than the cells isolated from BM and AT in the hindlimb ischemia in animal models [27]. Other studies have revealed a higher frequency of non-functional cells in BM-MSCs than in WJ-MSCs and stem cells derived from human exfoliated deciduous teeth (SHED) [28]. Additionally, molecular investigations presented the augmented expression of INF-γ, PDGFA, VEGF, IL10, and stromal-derived factor (SDF) in SHED compared to WJ-MSC and BM-MSC, indicating that SHED are possibly more effective than BM-MSC and WJ-MSC in modulating the immune response and fibrosis process [28]. MSCs isolated from AT, BM, and WJ presented similar cell surface antigen expression levels and showed comparable differentiation competence, BM-MSCs and WJ-MSC were superior over AT-MSCs concerning proliferation and clonality potential [29]. Regarding differential capacity, Bernardo et al. found that BM-MSCs have a more prominent chondrogenic differentiation potential than cells isolated from PL and fetal tissues [30], as displayed through the presence of representative morphological properties of cartilage, the concentration of toluidine blue stain, and the expression of collagen type II, IX, and X upon culture under chondrogenic conditions [30]. Furthermore, AT-MSCs and UC-MSCs displayed greater osteogenic potential compared to the chorionic membrane (CM)- and decidua (DC)-MSCs [31], and fibronectin could dramatically improve the osteogenic potential of MSCs mainly mediated by the promotion of phosphorylation and activation of Akt and ERK signaling axis [31].

In sum, though MSCs isolated from various tissues display a variety of common appearances, their biological functions, and some markers are dissimilar depending on the their origins. MSCs derived from diverse origins are phenotypically heterogeneous and demonstrate varied differentiation possibilities and release of bioactive factors related to tissue origin. The selection of MSCs with particular biological possessions provides the opportunity to use targeted therapies, in which the source of MSCs and the duration of culture act as influential marker [32].

Immunomodulatory properties of MSCs

As mentioned earlier, mesenchymal stem/stromal cells have the competence to modify immunological reactions through several mechanisms such as T cell suppression accompanied by induction of macrophages shift from M1 to M2 [33]. Therefore, they have been considered as an emerging therapeutic approach to treat immune-mediated disorders, such as GVHD, MS, and CD [34]. Furthermore, the therapeutic efficacy of MSC administration has been evidenced in acute lung injuries (ALI) and musculoskeletal diseases. In this regard, MSCs can migrate to injured sites after systemic injection and subsequently elicit a therapeutic effect through several mechanisms, particularly immunomodulation, and angiogenesis [35, 36]. While the corresponding mechanism involved in MSC immunomodulation has not yet been fully found, it seems that cell-to-cell contact along with trophic factors plays the central role in this process. MSCs can modify cytokine release’s profile of dendritic cells (DCs), naive and effector T cells, and natural killer (NK) cells to induce a superior anti-inflammatory or tolerant phenotype. They commonly affect mature DC type 1 (DC1) to diminish the secretion of tumor necrosis factor-α (TNF-α), modify DC2 to promote IL-10 secretion, adjust Th1 cells to decrease IFN-γ release, and finally provoke TH2 cells to upsurge IL-4 secretion [37]. Moreover, they trigger a rise in the frequency of regulatory T cells (Tregs) and a decrease in IFN-γ produced by NK cells [38]. A wide spectrum of soluble ingredients, in particular, transforms growth factor-β1 (TGF-β1), prostaglandin E2 (PGE2), hepatocyte growth factor (HGF), indoleamine-pyrrole 2, 3-dioxygenase (IDO), nitric oxide (NO), and IL-10 [4, 39,40,41] and has been supported that contribute to the immunomodulation axis. The PGE2 is a lipid intermediate proposed as a central factor stimulating T cell suppression by MSCs. It is generated from arachidonic acid through the functions of either the constitutive cyclooxygenase-1 (COX-1) or the inducible COX-2 enzymes, commonly expressed by human MSCs [42]. In addition, IDO as another soluble factor released by MSCs enables breakdown of tryptophan, which is required for T lymphocyte effector functions, and thereby resulted in immunosuppression in injured sites after MSC transplantation. MSCs do not constitutively express IDO, but they can be stimulated to express IDO upon inducement by IFN-γ but not TNF-α [43]. Sundrud et al. have suggested that IDO may hinder T cell proliferation and effector T cell activation and also induce NK cell apoptosis [44]. Regarding other reports, programmed cell death 1 ligand 1 (PD-L1) and FasL molecules may contribute to the immunoregulation stimulated by human MSCs (e.g., PL-MSCs) [45]. Observations have evidenced promoted levels of PD receptor expression on the surface of human T-effector cells following co-culture with MSCs in vitro, indicating the potential role of PD-1/B7-H1 axis in the mediation of the inhibitory effect of MSCs on effector T cells [46]. Furthermore, AT-MSC stimulated suppressive effects on T cells by promoting the expression of immunomodulatory cytokines, encompassing TGF-β, and IL-10, in association with IFN-γ inhibition and expression of T-bet transcription factors [47]. It has already been found that TGF-β and IL-10 contribute to the suppressive activities of Tregs and are critical for supporting immune homeostasis [48]. The performance of TGF-β as an immune regulator of T cell function is demonstrated by similarities between TGF-β1 knockout and T cell-specific TGF-β receptor II knockout rodents. Rodents in both models suffered from severe multiorgan autoimmunity, leading to premature death [34, 49]. Concerning genomic and proteomics analysis displaying high-level HGF expression and secretion from MSCs, other studies have clarified its potent role in MSC-induced immunomodulation. Investigations have revealed that HGF-treated monocytes remained undifferentiated and could alter Th cell profile from Th1 toward Th2 [50]. Also, in vivo studies have indicated that MSCs alleviated early ALI via paracrine HGF which induced mature DC differentiation into regulatory DCs in rodent models. Also, some studies have delivered proof of the concept that enhancing endogenous HGF secretion may induce partial rescue in patients suffering from inflammatory lung diseases [51].

Briefly, transplanted MSCs can migrate to the inflammation site and stimulate potent immunomodulatory and anti-inflammatory effects through cell-cell contact between MSCs and lymphocytes or generation of soluble factors, signifying that MSC application in many conditions is full of potentials for future clinical treatment [52, 53].

MSC homing and migration

One of the central advantages of MSC-based therapies is their ability to favorably home deteriorated tissue or organ. Homing encompasses both non-systemic and systemic homing [54]. In non-systemic homing, MSCs are grafted locally at the target tissue and are previously directed to the damage area by a chemokine gradient. However, in systemic homing, MSCs are injected or endogenously recruited into the bloodstream and experience a sequential process to exit circulation and migrate to the damaged area. The process of systemic homing is commonly split into five steps: (1) tethering and rolling (2) activation, (3) arrest, (4) transmigration, and (5) migration. In this section, MSC homing and migration both in vitro and in vivo is discussed and the crucial role of important chemokines and other factors in this perspective is elucidated [55].

In vitro MSC migration

In vitro, MSCs migrate in response to multiple chemotactic factors such as platelet-derived growth factor-AB (PDGF-AB), insulin-like growth factor-1 (IGF-1), chemokines RANTES, macrophage-derived chemokine (MDC), and stromal-derived factor-1 (SDF-1). MSC expresses these factor-related receptors, including the receptor tyrosine kinases for PDGF and IGF, CCR2, CCR3, and CCR4 for RANTES, MDC receptors for MDC, and CXCR4 receptor for SDF-1 [56]. Chemokines are more active on TNF-α-primed cells, signifying the high association between MSC recruitment, their succeeding homing to damaged tissue, and systemic and local inflammatory circumstances [56]. Bhakta et al. suggested that MSCs can be proficiently transduced to overexpress CXCR4, which consequently allows swift migration of transduced MSCs toward SDF-1 [57]. On the other hand, in vitro analysis showed that platelet-rich concentrates improved the migration potential of MSCs because of the persistent release of TGF-β1, IGF, VEGF, and PDGF-AB [57]. Also, preconditioning of MSCs with all-trans retinoic acid (ATRA) improved survival signaling axis activation, trophic factor release, and proangiogenic molecules, including COX-2, HIF-1, CXCR4, CCR2, VEGF, Ang-2, and Ang-4, which in turn, led to the upheld migration competence of MSCs [58]. Although MSCs are extensively used in clinical trials upon ex vivo expansion due to their low frequency, it is not clear how expansion and GMP manufacturing procedures may affect MSC homing capacity following transplantation. Additionally, it seems that the duration of cell culture, medium ingredients, and cell expansion levels may strongly affect MSC’s morphology, differentiation, viability, and migratory attributes [59]. Furthermore, studies revealed that freshly procured MSCs possess higher homing capability compared to expanded MSC and that diverse MSC subtypes, such as classical MSC and multipotent adult progenitor cells, display non-similar migration potential during in vitro migration assays [60]. This theory that altered MSC provisions can stimulate discrepancy based on their homing receptor expression leading to a different therapeutic outcome highlights the importance of optimizing of MSC expansion procedures before transplantation.

Endogenous MSC migration and homing

MSCs are localized in the BM from where they are recruited to other sites by processes possibly comparable to those applied by HSCs. Nevertheless, MSC may be located and circulated in PB, making it difficult to specifically identify migrating MSCs. MSC recognition in the PB is debated, whereas some studies confirmed that cord blood and mobilized PB may contain a significant number of cells [61]. Besides, Alm et al. identified MSCs in PB in patients suffering from hip bone fractures [62]; however, it could be asked whether MSC exists in PB of those patients by active migration or involvement of mechanical disturbance of bone tissue. Observations in murine have revealed that hypoxia induces MSC recruitment in PB [63] and also evidenced a promoted number of fluorescent MSC in murine PB following liver injury stimulation [64], indicating that systemic signals induce MSC secretion from BM. Besides, it was detected that MSCs may be released by adipose tissue in response to inflammation and that they are collected in lymph nodes and blood vessels by SDF-1/CXCR4-dependent axis (Fig. 1a) [65]. Recently, a study in murine models signified that CCR9, CXCR4, and c-MET play pivotal role in directing endogenous MSC migration toward the injured liver. The migrated murine BM-MSCs elicited diverse functions, particularly hepatic fate specification, and obstruction of hepatic stellate cell functions which led to suppression of collagen accumulations and liver fibrosis progression [64]. Given that a comprehensive array of human tissues have their own MSC, other findings have showed that local MSC from tissue or blood vessels can migrate only a short distance to reach the injured organ and thus cut the bloodstream route short (Fig. 1b) [60].

Fig. 1
figure 1

Two mechanisms suggested for recruiting endogenous MSC after tissue injury. a Special mediators (e.g., cytokines and growth factors) secreted by the injured tissue can stimulate recruitment of MSC from BM to injured sites through circulation. b Otherwise, MSC can be recruited from within tissues to the injured sites by migration within the stroma or through micro-capillaries. Mesenchymal stem/stromal cell (MSC); bone marrow (BM); stem cell factor (SCF); stromal cell-derived factor-1 (SDF-1); granulocyte colony-stimulating factor (G-CSF)

Migration and homing of transplanted MSCs

MSCs are progressively utilized as an intravenously used cellular therapeutic. The homing potential and engraftment to the injured site determine the potent efficacy of MSC-based cell therapy. There are some missing understandings for the biodistribution of MSCs, their cellular or molecular target structures, and responsible mechanisms by which MSCs are recruited to the target site [66]. MSC migration and engraftment process is affected by both chemical (e.g., chemokines, cytokines, and growth factors) and mechanical factors (e.g., hemodynamic forces) [67]. In vivo researches proved that the SDF-1/CXCR4 axis acts as an influential factor in the modification of motility of MSC-transplanted through intravenous (IV) routes, and also revealed that improvement in CXCR4 expression may be a possible approach to develop engraftment of MSC in BM and improve the recovery of hematopoiesis in NOD/SCID mice [68]. Besides, the promotion of myocardial SDF-1 expression after induction of myocardial infarction (MI) could promote the engraftment of transplanted MSCs in the injured heart and thus restore cardiac performance by upholding neovascularization in animal models [69]. In other MI animal models, studies showed that labeling of MSCs with superparamagnetic iron oxide (SPIO) nanoparticles makes tracking of administrated cells possible [70]. Examining the potential role of chemotactic SDF-1/CXCR4 signaling axis in the recruitment of engrafted BM-MSCs to the damaged cochlea following a noise-induced hearing loss (NIHL) confirmed the presence of the labeled transplanted cells in cochlear tissue of the murine models. Meanwhile, elevated levels of SDF-1 found in cochlear tissue confirmed that the SDF-1/CXCR4 signaling pathway plays a central role in BM-MSC migration into the injured sites after administration [71]. In this regard, other reports demonstrated that Fe3O4@polydopamine nanoparticles (Fe3O4@PDA NPs) improved the migration ability of MSCs by increasing CXCR4 expression levels [72]. A study on a murine model of burn injury showed that IV transplantation of labeled MSCs with Fe3O4@PDA NPs caused more reduction in inflammation of transplanted control mice. Additionally, the labeled MSC group displayed heightened cytokines and decreased production of proinflammatory factors [72]. In general, an extensive variety of mechanical and chemical factors have been elucidated that may affect MSC migration; however, most of these findings are developed by single-factor analysis at the cellular level in vitro, emphasizing the accomplishment of more comprehensive and multifaceted in vivo studies.

Though MSC homing after transplantation has been evidenced, this process failed to be prominently effective since only a small number of cells reach the target tissue and remain there after systemic injection. This has been attributed to the low expression rate of homing molecules concomitant with attenuation of expression of such molecules throughout expansion along with the heterogeneity of MSCs in cultures and MSC cultivation methods. A better comprehension of MSC’s biology, migration, and the homing mechanisms allow preparing MSCs with ideal homing competencies [73]. Moreover, despite the endogenous recruitment of MSCs, most adult tissues fail to heal after injury, which proves that these mechanisms are inadequate [74].

Application of MSC therapy in immune-mediated disorders

Mesenchymal stem/stromal cells (MSCs) exhibit anti-inflammatory and regenerative properties in addition to the multipotency capability. Following extensive preclinical in vitro and in vivo studies, autologous and allogeneic MSCs have been used in clinical trials in a variety of immune-mediated disorders, encompassing GVHD, SLE, OA, RA, MS, COVID-19, ALI/ARDS, etc. (Table 1). Current findings suggest that MSCs may not only replace the injured tissues but also deliver a pool of growth factors and regenerative molecules. Interestingly, MSCs can modify their gene expression profile in the damaged microenvironment and modulate the expression profiles of adjacent cells. For example, Cho et al. revealed that under the co-culture of MSCs and normal liver cells, expression levels of the CXCR6, CCR3, IL-2, IL-11, CD34, CD74, pro-collagen, FMS-like tyrosine kinase (FLT-3), neuregulin 4, Wnt2, and catenins were promoted. Conversely, under the co-culture of MSCs and the CCl4-injured liver cells, expression levels of CXCL2, cytoglobin, erythropoietin (EPO), v-Erb, retinoic acid receptor beta (RAR-ß), and Vav2 were boosted [75]. These findings represent the significance of identifying the differential molecular mechanisms that adjust the potentials of MSCs in the regeneration of damaged tissue.

Table 1 A brief overview of clinical trials in the context of the MSC-based therapy for immune-mediated disorders registered in ClinicalTrails.gov (January 2021)

MSCs in graft versus host disease (GVHD)

Graft versus host disease (GVHD) is a severe complication detected after approximately 40–60% of allogeneic HSC transplants but infrequently upon solid organ transplants. Acute GVHD is a multifaceted inflammatory disease in which various factors such as conditioning, recruitment of donor immune cells, and the release of proinflammatory cytokines are proposed to be contributed. MSC therapy is now a promising alternative for the treatment of acute GVHD (Fig. 2) [76]. Studies have shown that subconjunctival transplantation of human MSCs in ocular GVHD models reduced the number of CD3-positive cells in the injured site. In addition to the decreased tear osmolarity in transplanted eyes, MSC transplantation resulted in diminished Pax6 in experimental corneal models. These findings demonstrated that MSC therapy can modify corneal inflammation and squamous metaplasia in ocular GVHD, signifying the therapeutic potential of local MSC administration in this condition [77]. Tang et al. observed that the use of genetically engineered MSCs to overexpress intercellular adhesion molecule-1 (MSCs-ICAM-1) inhibited DC maturation and T cell immune response according to the mixed lymphocyte response (MLR) and lymphoblast transformation test (LTT) in vitro [78]. On the other hand, MSCs-ICAM-1 administration robustly extended the overall survival rate of the animal models of GVHD. The injected MSCs-ICAM-1 were recruited to secondary lymphoid organs (SLOs) in vivo, hindered the maturation of DCs and CD4+ T cell differentiation to Th1 cells, and also improved the frequency of Treg cells [78]. Although they failed to describe the rationality of ICAM-1 application, studies in a murine autoimmune thyroiditis model have indicated that ICAM-1 could affect the immunomodulatory potential of MSCs by targeting their migration in vivo [79]. Other observations exhibited that CXCR4 overexpressing MSCs (MSC-CXCR4) retained their immunomodulatory potential and exposed promoted migration competency in vitro [80]. In a murine GVHD model, intravenous infusion of MSC-CXCR4 ameliorated survival rate and alleviated clinical and pathological GVHD scores. Serological analyses evidenced a reduction in IL-2, IL-6, IFN-γ, and TNF-α and conversely an increase in IL-4 and IL-10 plasma levels in transplanted mice [80]. Likewise, a study on murine sclerodermatous GVHD showed that MSC therapy relieved the clinical and pathological gravity of cutaneous sclerodermatous GVHD [12]. Moreover, a reduction in skin collagen production in association with inhibition of TGF-β expression and function was supported in experimental transplanted models. Observations verified that MSCs not only migrated to the skin but also suppressed the recruitment of immune functional cells into the skin through inhibition of CCR4 and CCR8 expression on CD4+ T cells, which play a critical role in GVHD onset and progression [12]. Similarly, other studies revealed that MSC-derived EVs (MSC-EVs) recapped the therapeutic effects of MSCs on GVHD. For instance, IV injection of human MSC-EVs enabled extended survival of rodents with GVHD and recovered the pathologic injuries in various GVHD-affected organs, possibly mediated by inhibition of CD4+ and CD8+ T cell function and infiltration, and also promotion of Treg cell population. Microarray analysis exposed promoted levels of miR-125a-3p in the MSC-EVs [76]. As upregulated levels of the miR-125a family can suppress macrophage and effector T cell function [81], it seems that miR-125a-3p may be responsible for the alleviated clinical symptoms of GVHD in vivo. A phase II clinical trial carried out between October 2001 and January 2007 on 55 participants with steroid-resistant acute GVHD developed after HSC transplantation revealed that systemic BM-MSC injection could partially rescue the clinical presentation of transplant patients. Regarding observations, no participant experienced untoward effects during or immediately after MSC infusions and 9 participants presented a significant recovery [82]. These findings implied that injection of MSCs expanded in vitro, regardless of the donor, can be an operative and effective therapeutic modality for patients with steroid-resistant, acute GVHD. Besides, a double-blind randomized controlled trial showed that UC-MSC transplantation remarkably reduced the onset of chronic GVHD following HLA-haploidentical stem cell transplantation in the transplanted groups (27.4%) compared to control groups (49.0%) during 24-month follow-up. More importantly, UC-MSC therapy promoted memory B lymphocytes and the percentage of Tregs in association with increased Th1 to Th2 ratio; however, it stimulated a reduction in the number of NK cells [83].

Fig. 2
figure 2

MSC-based therapy for treating GVHD. Owing to their exclusive immunomodulatory properties, MSC injection can restore clinical symptoms in GVHD in vivo. Mesenchymal stem/stromal cell (MSC); graft versus host diseases (GVHD); transforming growth factor-beta (TGF-β); hepatocyte growth factor (HGF); indoleamine 2,3-dioxygenase (IDO); cyclooxygenase-2 (COX-2); prostaglandin E2 (PGE2); programmed death receptor (PD); programmed death-ligand 1 (PD-L1); tumor necrosis factor-alpha (TNF-α), TNFα-stimulated gene-6 (TSG6); interferon-gamma (IFN-γ); immunoglobulin (Ig); T helper cell (Th); T regulatory cell (T reg); M1 and M2 macrophage (M1 and M2); natural killer cell (NKC); dendritic cell (DC); cytotoxic T lymphocyte (CTL)

MSCs in systemic lupus erythematosus

The systemic lupus erythematosus (SLE) is a polymorphic, multisystemic autoimmune disease leading to extensive inflammation, which in turn, induces tissue’s deterioration in joints, skin, brain, lungs, kidneys, and blood vessels. It is characterized by a comprehensive disturbance of self-tolerance by autoreactive T and B cell activation leading to the generation of pathogenic autoantibodies and tissue deterioration [84]. Concerning underlying pathological mechanisms, rapidly evolving clinical trials suggest that MSC-based therapy may be an optimal treatment strategy for severe and refractory SLE [85,86,87]. Interestingly, reports exhibited that BM-MSCs procured from SLE patients show high levels of abnormalities, most importantly, cytoskeleton-related dysfunctions and intensified cellular senescence due to the upregulated expression of p53 and p16 accompanied by promoted apoptosis in comparison with normal MSCs [88]. In addition to the compromised differentiation and recruitment potential, expression profiles of genes related to immunological events in SLE-MSCs, including IDO, IL-6, IL-7, and TGF-β, are generally discrete from those in normal cells [89]. Consistently, biological activities of MSCs from SLE patients or lupus animal models are rigorously impaired, fail to modify multiple immune cell functions, and may support the autoimmunity onset through increased reactive oxygen species (ROS) levels as well as DNA damage [90]. Observations have demonstrated that murine BM-MSC transplantation into the SLE murine model had no significant effect on serum levels of anti-double-stranded DNA (anti-dsDNA) or proteinuria, while a restoration in glomerular immune complexes, lymphocytic infiltration, and glomerular proliferation was evidenced, representing the therapeutic potential of MSCs in the rescue of glomerular damage in SLE animal models [91]. Other in vivo studies revealed that dental pulp MSCs (DP-MSCs) and periodontal ligament MSCs (PDL-MSCs) had an immunoregulatory potential in SLE B6/LPR murine models [92]. Findings verified that both DP-MSCs and PDL-MSCs proficiently diminished proteinuria, anti-nuclear antibodies (ANA), and glomerular IgG/IgM in transplanted mice [92]. Also, the frequency of Th1 and plasma cells in the spleen dwindled in transplanted groups in the absence of any moderation in Th2, Th17, Tfh, and Treg cell percentages and IL-6, IL-10, IL-17, and MCP-1 serum levels, suggesting that DP-derived stem cells can restore renal glomerular defects and perivascular inflammation and may be recruited as alternative sources for SLE treatment [92]. In addition to the experimental animal models, clinical trials have provided proof of the notion that MSC therapy can exert beneficial therapeutic effects in patients with SLE by alleviating the disease progression and development in serologic scores and renal function. In this regard, a clinical trial conducted between March 2007 and November 2008 on 15 patients with active SLE evidenced the safety and significant efficacy of allogeneic MSC transplant, as presented by a reduction in SLE disease activity index (SLEDAI), a validated instrument for lupus disease activity in the preceding 10 days, and a significant reduction in serum levels of ANA, concomitant with an improvement in kidney function and percentage of peripheral blood Tregs [87]. These findings imply that MSC transplantation can elicit beneficial effects in patients with SLE, refractory to conventional treatment approaches. Conversely, another clinical trial on 2 females with SLE revealed that autologous BM-MSC transplantation had no significant effect on Tregs percentage in peripheral blood of grafted patients. However, disease activity indexes were modified and no unwanted events were reported during a 14-week follow-up [93]. These observations signify the importance of conducting more trials before MSC application in clinical settings to clarify the underlying mechanism contributed to observed desired effects of MSCs in patients with SLE. Moreover, several trials have verifies the UC-MSC potential for SLE therapy. Accordingly, a study on 30 patients with refractory SLE indicated that UC-MSCs promoted Tregs and inhibited Th17 cell frequencies and activation, which were mediated by adjustment of TGF-β and PGE2 expression in lupus patients [94]. Correspondingly, another trial on 40 participants with refractory SLE revealed that UC-MSC administration was well-tolerated and had no severe transplantation-induced side effects. In addition to a reduction in SLEDAI scores, UC-MSC transplantation diminished proteinuria and attenuated serum creatinine, urea nitrogen, and ANA levels [94], describing UC as a promising source to isolate MSCs and use them in SLE treatment.

MSCs in multiple sclerosis

Multiple sclerosis (MS) is a chronic autoimmune disease of the central nervous system (CNS) characterized by damage to the CNS, stimulating physical or cognitive deficits, as well as neurological dysfunctions [95]. To identify an appropriate treatment to alleviate the neurological signs and remyelination, autologous and allogeneic MSC transplantation was introduced as an operational and effective therapeutic approach. Various preclinical and clinical trainings have established that MSC transplantation can ameliorate the CNS restoration and improve functional neurological signs. For instance, human amniotic mesenchymal stem/stromal cells (hAMSCs) improved the expression of neurotrophic factors that participated in promoting the survival, progression, and function of neurons in vitro. More importantly, it has been found that co-culture of neural progenitor cells (NPCs) with hAMSCs supports their differentiation into functional neurons [96]. Moreover, hAMSCs suppressed MMP dysfunctions and accordingly sustained endothelial cell survival, angiogenesis, and maintenance of vascular networks [96]. Regarding the observations showing that the use of specific and broad-spectrum inhibitors for MMPs can diminish neuroinflammation and brain lesion in neurodegenerative diseases (e.g., MS) [97], it seems that the inhibitory effect of the MSCs on MMPs plays a pivotal role in improving motor deficits in MS patients upon transplantation. On the other hand, in vivo investigation in a canine MS model verified the efficacy of MSC infusion leading to a better quality of life in grafted dogs, offering hopefulness for comparable encouraging outcomes in patients with MS [98]. Further, a similar report in experimental allergic encephalomyelitis (EAE) mice, a common MS experimental models, suggested that human BM-MSC infusion improved functional recovery in transplanted models. Findings revealed that infused human BM-MSCs collected in the CNS condensed the lesion volume and finally augmented the frequency of oligodendrocyte (ODC) lineage cells in the lesion zone [99]. Furthermore, assessment of particular percentages of effector T cell subtypes in PB and their related cytokine serum levels confirmed a decrease in Th1 cells and IL-17 generating Th17 inflammatory cells and their related cytokines and conversely demonstrated an improvement in IL-4 generating Th2 cells and anti-inflammatory cytokines in transplanted models [99]. Due to the generally accepted protective role of Th2 cells in MS patients and the importance of the Th1/Th2 ratio in determining disease progression or alleviation, MSC therapy can be proposed as a rational therapeutic strategy in these patients. Moreover, a study in murine MS models supported the positive role of intravenous MSC-EV injection, such as restored motor deficits, attenuated brain atrophy, improved cell proliferation in the subventricular zone (SVZ), and reduced immune cells infiltration. A strong decline in serum levels of Th1- and Th17-produced cytokine approved MSC-EV-induced immunomodulation in transplanted murine models [100]; however, more comprehensive studies are required to introduce EV delivery as a potential therapeutic approach for the neurodegenerative phase of MS. Other investigations respecting the therapeutic efficacy of EV in experimental MS models evidenced that BM-MSC-EVs could improve neural behavioral scores, suppress immune cell infiltration into the CNS, and alleviate the demyelination process compared to control animals [84]. In addition, injection of EVs promoted IL-10 and TGF-β levels, though reduced serum levels of TNF-α and IL-12 [84]. These findings suggest that the polarization of microglia is another potential mechanism used by MSCs and their secretome to alleviate MS-related deficits. Furthermore, studies in EAE mice confirmed the potential of PDL-MSC secretome in hindering activation of NALP3 inflammasome and supporting maintenance from EAE [101]. Regardless of a decrease in cleaved caspase 1, IL-1β, and IL-18 levels, transplantation of MSC secretome downregulated proinflammatory toll-like receptor (TLR)-4 and NF-κB in transplanted EAE models. Analyses verified high levels of anti-inflammatory IL-10, TGF-β, and SDF-1α in the human PDL-MSC secretome [101]. Based on promising results of the MSC-based therapies in MS, several clinical trials have been conducted to address the safety and efficacy of MSCs in humans. Accordingly, the safety and feasibility of UC-MSC therapy has been supported by a study on 20 patients with MS [102]. Observations approved the absence of any severe adverse events during a 12-month follow-up following multiple MSC injections, while symptoms of rescue were significant 1 month after injection. Moreover, improvements were observed in the Kurtzke Expanded Disability Status Scale (EDSS), bladder, bowel, and sexual dysfunctions, average scores for non-dominant hand, distance walked over time, and general views on positive health alterations and developed quality of life [102]. Though these observations approved the safety and feasibility of IV injection in patients with MS, its potential therapeutic benefits should be further investigated. Additionally, assessment of autologous AT-MSC injection in 34 patients with MS showed the safety of stem cell transplantation in enrolled participants, but evaluation of the treatment outcomes displayed a non-significant rate of efficacy [103]. Moreover, a phase 2 clinical trial (registered at ClinicalTrials.gov, NCT00395200) suggested that autologous MSC systemic injection not only was safe and feasible but also had positive therapeutic outcomes in participants with secondary progressive MS most likely mediated by induction of neuroprotection concerning the structural, functional, and physiological recovery [104]. Overall, it appears that inhibition of Th1 and Th17 activation and infiltration, promotion of Tregs, and TH2 function along with induction of neuroprotection may contribute to optimal effects elicited by MSC transplantation in patients with MS.

MSCs in Crohn’s disease

Crohn’s disease (CD) is an inflammatory bowel disease (IBD) that typically affects the terminal ileum (outer ends of the intestines) but can also target the whole gastrointestinal tract, from mouth to anus [105]. The CD is associated with full-thickness inflammation in the gastrointestinal tract leading to pain, discomfort, unusual bowel activities, and digestive problems. It is generally characterized by severe Th1 cell-induced inflammation of the colon partially resulting from a disrupted immune tolerance to mucosal antigens [106]. The anti-inflammatory properties of MSCs propose their potential for improving the damaging symptoms accompanying CD [107]. In vivo studies provide evidence suggesting that intralesional administration of human embryonic stem cell-derived MSCs (hESC-MSCs) could decrease serum levels of IL-2 and IL-6, two main inflammatory cytokines associated with CD, in canine models [108]. In this regard, other studies showed that IV infusion of human AT-MSCs had the potential to hinder body weight loss, diarrhea, and inflammation and raise the survival rate of experimental CD models. Findings revealed that the observed positive therapeutic effects were mediated by mitigation of Th1-driven autoimmune and inflammatory reactions along with improved Tregs population and activation [109], introducing AT-MSC as a regulator of immune tolerance and assuring cell-based therapy candidates for CD. Moreover, compartmental analysis evaluating the therapeutic potential of intraperitoneal AT-MSC and BM-MSC transplantation in a trinitrobenzene sulfonic acid (TNBS)-induced murine CD model revealed that both of them could improve the clinical and histopathologic severity of intestinal inflammation, leading to the augmented survival of murine CD model [110]. Additionally, transplanted cells efficiently improved IL-10 expression and decreased the secretion of proinflammatory cytokines TNF- α, IL-12, and proangiogenic factor VEGF [110]. Likewise, other examinations indicated that AT-MSC administration attenuated the disease activity index (DAI) and improved the severity of colitis in a rodent CD model. Significantly, regulation of intestinal epithelial cell (IEC) proliferation, Wnt signaling pathway, and T cell immunity were suggested as the underlying mechanism of the AT-MSC-prompted therapeutic effect in the rodent CD model [111]. The crucial role of the Wnt axis has already been confirmed in murine IBD, where Roger et al. showed that injection of a Wnt agonist to STAT6 (−/−) mice induced the Wnt signaling in the damaged mucosa and accelerated wound-healing in the TNBS-induced CD model [112]. Based on the results of animal studies, several clinical trials were designed and accomplished to confirm the safety, feasibility, and efficacy of MSC therapy in CD. A survey conducted between 2007 and 2014 on 10 patients with CD showed that locally injected MSCs were safe and feasible and restored refractory patients, and regained responsiveness to the therapeutic agents formerly shown ineffective [113]. Another study displayed the allogeneic expanded AT-MSC (Cx601) administration is effective strategy for treating CD. The trial, which was carried out at 49 hospitals in seven European countries and Israel from 2012 to 2015 on 212 participants, proposed that a single intralesional injection of Cx601 resulted in the rescue of pathological symptoms in the transplanted group compared to the placebo group. However, 17% and 29% of participants in the transplanted and placebo group showed treatment-associated adverse events, most frequently anal abscess [114]. Moreover, investigating the potential of IV injection of allogeneic MSCs in 16 participants with luminal CD during a phase 2 clinical trial signified a remarkable decrease in Crohn’s disease activity index (CDAI) scores, which are commonly applied in clinical trials to evaluate CD activity, only 6 weeks post-transplantation. Concerning observations, 12 participants had a clinical response, 8 participants had clinical remission, and 7 of them experienced an endoscopic improvement in the absence of any severe treatment-related adverse events [115]. Overall, analyses imply that MSC administration, particularly, the cells isolated from adipose tissue, can improve the quality of life of treated CD patients after local or systemic injection mediated by suppression of acute mucosal inflammation through downregulating the secretion of a broad spectrum of mediators contributing in the local and systemic inflammatory reactions.

MSCs in acute lung injury/acute respiratory distress syndrome

Acute respiratory distress syndrome (ARDS) and its milder form acute lung injury (ALI) are characterized by acute respiratory failure after multiple invasions to the pulmonary parenchyma or vasculature [116]. It has been verified that macrophages play important role in the inflammatory response in ALI/ARDS. Remarkably, they play a dual proinflammation and anti-inflammation role according to the microenvironment in various pathological phases. In the acute phase of ALI/ARDS, local alveolar macrophages, characteristically showing the M2 phenotype, shift into the M1 phenotype and eventually trigger the secretion of proinflammatory mediators [117]. In the last years, because of their multipotency and unique aptitude to release multiple paracrine factors, ranging from growth factors, factors fluctuating endothelial and epithelial permeability, and anti-inflammatory cytokines, MSCs have been introduced as a therapeutic option which can alleviate major complications underlying lung disease (e.g., ALI/ARDS), such as disrupted alveolar fluid clearance, modified pulmonary endothelial permeability, and dysregulated immune responses (Table 2) [144, 145]. Studies have exhibited that inhibition of the Hippo signaling pathway improves MSC proliferation, motility, and differentiation in vitro, supporting the theory that MSCs with downregulated Hippo signaling pathway can rescue lipopolysaccharide (LPS)-induced ARDS in vivo [146]. As known, the Hippo signaling pathway is conserved and modifies a variety of cellular processes, surrounding cell survival, proliferation, and differentiation. In mammals, the activation of the Hippo pathway leads to the inactivation of Yes-associated protein (YAP) by large tumor suppressor 1/2 (LATS1/2)-mediated direct phosphorylation. Contrariwise, dephosphorylation of YAP results in its transport into the nucleus and its succeeding interaction with TEA/ATTS domain (TEAD), forkhead box protein O1 (FOXO1), and other transcription factors, and therefore can exert cell proliferation, organ growth, and stem cell self-renewal [147]. Other studies on murine LPS models demonstrated that transplantation of murine BM-MSCs with downregulated Hippo pathways led to the intensified retention of murine MSC in ARDS lung tissue and their differentiation into alveolar epithelial type II (AE2) cell as a supporter of the alveolus [120]. Moreover, injected cells supported a decline in lung wet weight to body weight ratio, the diminished total protein and albumin concentrations in bronchoalveolar lavage fluid (BALF) accompanied by downregulation of proinflammatory cytokines, and upregulation of anti-inflammatory mediators [120]. Concerning the elevated release of proinflammatory cytokines and also reactive oxygen species (ROS), which in turn, induces the activation of neutrophil-derived proteases and the formation of neutrophil extracellular traps (NETs) during ALI/ARDS, some investigations addressed the effect of MSCs on NET formation in LPS-induced murine models. Accordingly, transplanted MSCs were capable of survival and modifying pulmonary inflammation, reducing ROS generation, and suppressing NET formation in the experimental transplanted model [122]. Moreover, a preclinical study evaluated the therapeutic efficacy of systemic infusion of BM-MSCs, AT-MSCs, and lung tissue MSCs (L-MSCs) in Wistar rats ARDS models. Regardless of their source, transplanted cells ameliorated lung function and decreased alveolar collapse, tissue cellularity, collagen, and elastic fiber content in lung tissue. Correspondingly, BM- and AT-derived MSCs attenuated the expression rate of several immune mediators, such as TNF-α, IL-1β, CXCL1, TGF-β, and VEGF, and reduced the number of damaged and dead cells in lung and kidney. Besides, they could improve the expression of keratinocyte growth factor (KGF) in lung tissue [121]. Moreover, various studies have suggested that MSC-EVs can elicit promising effects in ALI/ARDS patients. Accordingly, an assessment of the anti-influenza potential of swine MSC-EVs in vitro as well as in lung epithelial cells, and its anti-viral and immunoregulatory properties in vivo in a swine influenza virus model revealed that MSC-EVs could suppress the hemagglutination functions of avian, swine, and human influenza viruses. On the other hand, MSC-EVs obstructed the replication of influenza virus and virus-stimulated apoptosis in epithelial cells of the lung, and also intratracheal administration MSC-EVs could decrease virus shedding in the nasal swabs, attenuate proliferation of influenza virus in the lungs, and diminish virus-simulated generation of proinflammatory mediators in the lungs of transplanted pigs [119]. Similarly, systemic injection of MSC-exosomal miR-30b-3p exerted protective effects against ALI in murine models [123]. The negative relation between miR-30b-3p and TNF-α, NF-κB, IL-6, and IL-8 levels in the lung tissue and BALF in murine ALI models, as shown by Zhou et al. [148], signifies that the induced protective effects of MSC-exosomal miR-30b-3p are possibly achieved by downregulation of NF-κB and proinflammatory cytokines in experimental models. These findings are in consistent with other observations, representing the central role of miRNAs in determining the outcomes of therapeutic approaches in lung inflammatory diseases [149,150,151]. Interestingly, some studies have demonstrated that BM-MSCs could transfer mitochondria to pulmonary alveoli and support protection from acute lung injury. In this regard, Islam et al. verified the mitochondrial transfer in intact lungs in a rodent model treated with LPS. They noticed that human or murine BM-MSCs injected in murine airways could transfer mitochondria and repair mitochondrial bioenergetics in the lungs [150]. Other reports have also proposed that mitochondrial dysfunction is detected in case of prolonged inflammation, and MSCs can transfer mitochondria to alleviate inflammation, which reveals their rescue capabilities via stimulating anti-inflammatory responses [152].

Table 2 Mesenchymal stem/stromal cell (MSC)-based therapy for common immune-mediated lung disorders (animal studies)

On the other hand, a phase 1 clinical trial carried out between July 2013 and January 2014 on 9 participants with severe ARDS verified the safety and feasibility of a single-dose systemic injection of allogeneic BM-MSCs in transplanted patients. One patient died 1 month after transplantation and one experienced multiple embolic infarcts of the spleen, kidneys, and brain. None of these intense untoward events were supposed to be treatment-related [153].

MSCs in coronavirus disease 2019

The coronavirus disease 2019 (COVID-19) is a contagious respiratory and vascular disorder caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) [154]. While the first human case was identified in Wuhan, China, in December 2019, recent evidence suggests that the virus may have been moderately disseminated months earlier in Italy [155]. Angiotensin-converting enzyme 2 (ACE 2) proteins, which are significantly expressed on various human cells, such as alveolar type II cells (AT2), oral, esophageal, ileal epithelial cells, myocardial cells, proximal tubule cells of the kidneys, and urothelial cells of the bladder, are suggested to contribute to the SARS-CoV2 internalization [156, 157]. The COVID-19 contagion is appeared by forceful inflammatory reactions with the secretion of a massive quantity of proinflammatory cytokines, triggering cytokine storm events [158]. ICU patients with COVID-19 have exposed higher plasma levels of the inflammatory mediators, including IL-2, IL-6, and TNF-α, granulocyte colony-stimulating factor (GCSF), CCL2, macrophage inflammatory protein 1-α (MIP-1α), and interferon-gamma inducible protein 10 kDa (IP-10) [159]. Correspondingly, it is supposed that MSCs can modulate the cytokine storm elicited by coronavirus infection due to their unique properties in modifying the immune response and regulating immune cell infiltration and motility (Fig. 3) [160]. In this context, the first clinical trial was designed and carried out in Beijing Hospital, China, from January 23 to February 16, 2020, to evaluate whether MSC therapy can ameliorate the outcomes of 7 participants with COVID-19 pneumonia. About the observations, MSCs remarkably restored the outcomes of all transplanted participants without severe unwanted events only 2 days post-transplantation. A promotion in PB lymphocyte counts, as well as a reduction in the C-reactive protein (CRP) levels, concomitant with a reduction in cytokine-secreting immune cells, CXCR3 + CD4+ T cells, CXCR3 + CD8+ T cells, and CXCR3+ NK cells were found upon administration. The serological analysis also verified reduced serum levels of TNF-α simultaneously increased levels of IL-10 after transplantation [161]. These observations offered first evidence suggesting that systemic injection of MSCs is safe and effective for treating COVID-19 patients. Further, the study of possible effects of IV human UC-MSC infusion in COVID-19 patients indicated that human UC-MSC transplantation shortened time to clinical improvement in the transplanted group compared to the control group. Meanwhile, clinical symptoms of weakness, fatigue, and respiratory distress perceptibly alleviated after human UC-MSC therapy [162]. Another clinical trial in a patient with severe COVID-19 infection showed that systemic infusion of human UC-MSC alleviated the inflammation signs, as approved by assessment of laboratory indexes and computed tomography (CT) images, leading to the discharge of the patient from ICU [163]. Likewise, transplantation of human Wharton’s jelly MSCs (hWJCs) improved pulmonary function and symptoms of participants suffering from COVID-19 pneumonia 48 h post-transplantation. The immunological analysis revealed enhanced frequencies of lymphocyte subsets and diminished levels of IL-6, TNF-α, and post-transplant CRP [164]. Moreover, the safety and efficacy of allogeneic BM-MSC-derived exosomes (ExoFlo™) was evidenced for treating severe COVID-19 during a trial conducted on 24 participants within 2 weeks follow-up. In addition to verifying the safety and feasibility of the method, 71% of participants recovered, 13% remained stable, and 16% expired for causes not associated with cell transplantation, highlighting the ExoFlo potential to be considered as a capable therapeutic modality for severe COVID-19 [165].

Fig. 3
figure 3

MSC-based therapy for treating COVID-19. MSC transplantation can stimulate COVID-19 rescue by affecting immune cells proliferation, differentiation, and activation, through releasing special mediators and triggering cytokine storm alleviation. Mesenchymal stem/stromal cell (MSC); coronavirus disease 2019 (COVID-19)

Taken together, despite encouraging results about the therapeutic potential of MSC therapy, there is no widespread evidence on its efficacy in defeating COVID-19 disorder. Though 42 clinical studies have been registered in ClinicalTrails.gov (January 2021) (Fig. 4), they are almost in phases I and II, and the therapeutic effects of MSC therapy on COVID-19 development are not yet clarified. The opportunity to use various administration routes such as inhalation and improvement of MSC immunoregulatory potential by MSC pre-treatment with hypoxia or ischemia includes many attractions for large-scale studies [166].

Fig. 4
figure 4

Clinical trials in MSC therapy for COVID-19 registered in ClinicalTrials.gov (January 2021). This graph presents the distribution of MSC-based clinical trials for COVID-19 treatment based on cell source (a), study phase (b), number of participants (c), and study locations (d). Mesenchymal stem/stromal cell (MSC); coronavirus disease 2019 (COVID-19); bone marrow (BM); adipose tissue (AT); umbilical cord (UC); Wharton’s jelly (WJ)

MSCs in osteoarthritis

Osteoarthritis (OA) is a common chronic joint condition caused by degeneration of articular cartilage and also other joint changes, including bone hyperplasia. Given the MSC’s potential to differentiate into chondrocytes and exert immunomodulation in the target tissue, their administration has turned into the most comprehensively discovered cell-based therapy approach for osteoarthritis (Fig. 5) (Table 3) [204]. MSC is found in synovial fluid (SF) and can simply be procured by arthrocentesis or arthroscopy. In vitro, chondrogenic stimulation of SF-MSCs in collagen sponges showed the respectable potential of chondrogenic gene stimulation and ECM formation. An in vivo study on murine OA models revealed that intra-articular injection of xenogenic SF-MSCs fails to elicit chondroprotection in transplanted models [172]. However, UC-MSC injection into a rabbit model of temporomandibular joint (TMJ)-OA induced by monosodium iodoacetate led to the regenerative outcome and anti-inflammatory influences as well as high-level neuroprotection. The observed therapeutic effects were dependent on promoted expression of growth factors, ECM markers, anti-inflammatory cytokines, and conversely the lessened expression of proinflammatory cytokines (e.g., TNF-α, IL-1β, and IL-6) [174]. Findings which support the UC-MSC potential to provoke both chondrogenesis and chondroprotection imply that they can be an effective source for OA therapy. Moreover, evaluation of intra-articular MSC infusion in murine OA models resulted in suppressed expression of A disintegrin and metalloproteinase with thrombospondin motifs 5 (ADAMTS5) in joint cartilage in transplanted models [176]. Due to the verified destructive role of ADAMTS5 in OA progression [205], scholars seek to discover novel strategies to suppress their activation in joint cartilages. Consequently, the inhibitory effect of MSCs on ADMATS5 activation evidenced the rationality of MSC-based therapies for treating cartilage disorders. Conversely, a noticeable increase in the expression of TNF-α-stimulated gene/protein 6 (TSG-6), an anti-inflammatory and cartilage protective factor, in transplanted OA models suggested that this method can stimulate neuroprotection in damaged cartilages [176]. In addition, intra-articular transplantation of BM-MSC secretome alleviated pain and cartilage damage, but not subchondral bone modifications and synovial inflammation in a murine collagenase-induced OA model [169]. It appears that using the regenerative potential of MSC secretome, it is conceivable to improve the optimization, affordability, and clinical translatability of this approach. Concerning other studies in this context, exosomes derived from miR-140-5p-overexpressing human synovial MSC (SMSC-140-Exos) can effectively treat OA. It has been supposed that SMSC-140-Exos promoted the proliferation and recruitment of articular chondrocytes (ACs) without any negative effects on ECM releases [167]. In detail, Wnt5a and Wnt5b were highly expressed in SMSC-140-Exos, which in turn led to YAP activation, as a mediator of cell proliferation. Then, YAP obstructed the expression of SRY-box transcription factor 9 (SOX9) and suppressed ECM formation, which improved the proliferation and recruitment of ACs [167]. Correspondingly, Xu et al. found that co-culture of MSCs with ACs can reshape and induce their proliferation by releasing soluble factors in vitro [206]. As ACs generate and retain substantial quantities of active and inactive BMPs [207] and are recognized to improve ECM production and trigger chondrogenesis and osteogenesis, their improved proliferation and activation by MSCs or other treatments can develop OA rescue. Similarly, there is some evidence confirming the potential of exosomes derived from miR-26a-5p overexpressing BM-MSC (BM-MSC-26a-Exos) to trigger positive therapeutic effects in a rodent OA model by targeting prostaglandin-endoperoxide synthase 2 (PTGS2) [208] frequently detected in damaged cartilages. In this respect, other observations revealed that exosomes from human embryonic stem cell-derived MSCs (ESC-MSC-Exos) had a profitable effect on OA via augmenting collagen type II (CII) production and inhibition of ADAMTS5, providing a balance between generation and degradation of chondrocyte ECM which elicited OA restoration in vivo [209]. Also, a clinical trial conducted on 18 participants with OA evidenced the safety and efficacy of human amniotic MSCs (hAMSCs) transplantation (5 × 107 cells each time). Observations demonstrated that intra-articular administration of hAMSCs reduced pain and restored knee joint function and cartilage, describing them as potential candidates for knee OA therapy [210]. Moreover, single intra-articular injection of autologous AT-MSCs in 12 patients with knee OA supported a noticeable amelioration of Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC) score, which is commonly used to assess pain, stiffness, and function in patients with OA, during a 6-month follow-up in the absence of any rigorous adverse effects [211]. Similarly, other in vivo studies demonstrated that intra-articular injection of autologous adipose AT-MSCs (1 × 108 cells each time), in addition to improving WOMAC score, could diminish cartilage defects and induce a rescue in the cartilage volume in the medial femoral and tibial condyles of transplant patients, possibly mediated by hyaline-like articular cartilage restoration [212].

Fig. 5
figure 5

Therapeutic potential of MSCs and their secretome for treating OA. Rendering literature, MSCs can be isolated from several sources ranging from bone marrow to endometrium and be injected into OA patients via the intra-articular route to induce encouraging outcomes. Mesenchymal stem/stromal cell (MSC); osteoarthritis (OA); matrix metalloproteinase (MMPs); fibroblast-like synoviocytes (FLS)

Table 3 Mesenchymal stem/stromal cells (MSC)-based therapy for common immune-mediated musculoskeletal disorders (animal studies)

MSCs in rheumatoid arthritis

Rheumatoid arthritis (RA) is a chronic autoimmune inflammatory disorder resulting from peripheral tolerance’s impairment stimulating the immune cell’s unregulated infiltration into the synovial membrane [213]. Also, the unbalanced immune reactions in proinflammatory and anti-inflammatory cells, most significantly, between memory Th17 and memory regulatory T cells (Tregs) seems to play a pivotal role in RA onset and progression [214]. Now, MSC therapy has become a promising therapeutic plan for RA recovery given their immunoregulatory belongings (Table 3) [215, 216]. Meanwhile, MSCs can alter the function of memory lymphocytes such as Th17, follicular helper T (Tfh) cells, and gamma delta (γδ) T cells while supporting Treg cell production and facilitating alleviation of RA clinical symptoms [170]. A variety of in vivo studies have suggested that human BM-MSCs can improve arthritis in animal models, such as collagen-induced arthritis (CIA). A recent report has signified that MSCs alleviated the severity of arthritis by reducing proinflammatory cytokine levels in association with attenuating the ratio of Th17 to Tregs cells in IL-1 receptor antagonist-deficient mice (IL-1RaKO) [170]. As Tfh cells are promoted and associated with autoantibodies in patients with OA, other investigations focused on its role in the RA progression. Accordingly, Liu et al. found that UC-MSC transplantation in CIA mice inhibited the development of arthritis by suppressing Tfh proliferation and also activation in vivo likely achieved by IDO releases [189]. Rising proofs suggest that MSCs induce antioxidant characteristics in a variety of animal disorders, which enable their cytoprotective and anti-inflammatory capabilities. However, evolving approaches to improve their therapeutic effect are of paramount importance. In this context, a study revealed that combined use of human MSCs with hesperidin, a natural compound with antioxidant activity, could ameliorate oxidative stress and intensify MSC immunosuppressive function through targeting IL-9 expression and serum levels in adjuvant-induced arthritis (AIA) of a murine OA model [171]. The significance of the IL-9 in RA depends on its potential to sustain the survival of neutrophils, increase MMP expression and activation, and assist Th17 cell differentiation supported by induction of transcription factor RORγt and STAT3 phosphorylation [217]. On the other hand, MSC-derived miR-150-5p exosomes (Exo-150) could suppress the migration of fibroblast-like synoviocytes (FLS), which play a crucial role in RA pathogenesis, and diminish tube formation in human umbilical vein endothelial cells (HUVECs) through targeting matrix metalloproteinase 14 (MMP14) and vascular endothelial growth factor (VEGF) in vitro [175]. In a murine CIA model, Exo-150 infusion improved clinical arthritic scores likely by suppressing synoviocyte hyperplasia, delivering the first proof of therapeutic efficacy of exosome therapy for RA [175]. Similarly, MSC-derived miR-192-5p exosomes (Exo-192) could delay the onset of the inflammatory response through targeting Ras-related C3 botulinum toxin substrate 2 (RAC2) in experimental models [218]. Rendering the findings by Dey et al. that interaction between RAC2 and inducible nitric oxide synthase (iNOS) may provoke NO upregulation and consequently initiate chronic inflammation in the RA synovium, application of therapeutic strategies focusing on RAC2 inhibition can exert beneficial effects in RA patients [219]. Another preclinical study suggested that MSC-derived exosomes with overexpressed miR-146a, a well-known miRNA involved in regulation of immune response, improved FoxP3, TGF-β, and IL-10 gene expression in murine CIA models, proposing their potential for treating RA through enhancing Treg cell populations and anti-inflammatory cytokine levels [220]. According to the promising results based on MSC therapy for RA in animal models, several clinical trials have been accomplished to report the safety and efficacy of these cell transplantation in human models. For instance, a phase I, uncontrolled, open-label trial on 9 participants showed that infusion of 1 × 108 UC-MSCs decreased levels of IL-1β, IL-6, IL-8, and TNF-α without any serious adverse events post-transplantation [221]. Besides, a phase Ib/IIa clinical trial revealed that systemic injection of expanded Cx611 allogeneic adipose-derived stem cells was safe and well-tolerated in 43 patients with refractory RA [222]. Likewise, intra-articular knee injection of autologous BM-MSCs in 15 RA participants improved WOMAC score and supported its potential efficacy in transplant patients during a 12-month follow-up [223].

In sum, these findings justify the necessity for large-scale studies over a prolonged evaluation period before utilizing MSCs in the clinical setting to restore RA.

Conclusion and prospect

As mentioned, given their unique attributes, such as differentiation into a wide spectrum of adult cell lineages, immunomodulatory competence along with lower ethical concerns and secretion of angiogenic factors, mesenchymal stem/stromal cells (MSCs) have attracted growing attention worldwide to restore immune-mediated disorders (e.g., GVHD, MS, COVID-19, and OA). The underlying mechanism contributing to MSC immunomodulation has not entirely been elucidated, while it seems that cell-cell contact in association with trophic factors ranging from cytokine to growth factors play pivotal roles in this process. In addition to animal studies, various clinical trials have also evidenced the safety, feasibility, and efficacy of administration of MSCs and their secretome in immunological disorders. Nonetheless, their promising effect on human clinical outcomes has not yet been reliably realized. Moreover, the oncogenic potential of uncontrolled MSC differentiation needs to be further investigated, as some studies have shown that human AT-MSC experience spontaneous transformation following prolonged expansion by consecutive c-Myc upregulation and p16 downregulation [224]. In this regard, another report revealed that in vitro expansion of human BM-MSCs produced a subpopulation of cells with improved telomerase functions, chromosomal aneuploidy, and translocations, capable of developing tumors in multiple organs in NOD/SCID mice [224]. Moreover, large-scale studies are required to extend knowledge about recruiting MSCs to improve their migration and homing following transplantation. Additionally, identifying MSC secretome, as a cell-free alternative that exerts inherently advantageous therapeutic effects, delivers a new paradigm for their application in regenerative medicine. Exosomes uphold the therapeutic merits of their origin cells in the absence of revealing concerns such as possible tumorigenesis and unwanted mutation in MSC [225]. Moreover, the therapeutic potential of MSC exosomes may be developed through genetically modified MSC exosomes to express special ligands that direct them toward a target tissue and transfer genes and other molecules directly to the target area as a gene delivery system.

Taken together, it is supposed that enrichment of the MSC culture, choosing appropriate induction factors, and finding novel strategies to promote MSCs homing post-transplantation accompanied by optimization of MSC delivery dose and route in various diseases can elicit optimal therapeutic outcomes in patients with immune-mediated/immune-dysregulating diseases.

Availability of data and materials

Not applicable.

Change history

References

  1. Ding D-C, Shyu W-C, Lin S-Z. Mesenchymal stem cells. Cell Transplant. 2011;20:5–14.

    Article  PubMed  Google Scholar 

  2. Friedenstein A, Piatetzky-Shapiro I, Petrakova K. Osteogenesis in transplants of bone marrow cells. Development. 1966;16:381–90.

    Article  CAS  Google Scholar 

  3. Stefańska K, Bryl R, Moncrieff L, Pinto N, Shibli JA, Dyszkiewicz-Konwińska M. Mesenchymal stem cells–a historical overview. Med J Cell Biol. 2020;8:83–7.

    Article  Google Scholar 

  4. Tavakoli S, Ghaderi Jafarbeigloo HR, Shariati A, Jahangiryan A, Jadidi F, Jadidi Kouhbanani MA, Hassanzadeh A, Zamani M, Javidi K, Naimi A. Mesenchymal stromal cells; a new horizon in regenerative medicine. J Cell Physiol. 2020;235:9185–210.

    Article  CAS  PubMed  Google Scholar 

  5. Baghaei K, Hashemi SM, Tokhanbigli S, Asadi Rad A, Assadzadeh-Aghdaei H, Sharifian A, Zali MR. Isolation, differentiation, and characterization of mesenchymal stem cells from human bone marrow. Gastroenterol Hepatol Bed Bench. 2017;10:208–13.

    PubMed  PubMed Central  Google Scholar 

  6. Hu MS, Borrelli MR, Lorenz HP, Longaker MT, Wan DC. Mesenchymal stromal cells and cutaneous wound healing: a comprehensive review of the background, role, and therapeutic potential. Stem Cells Int. 2018;2018:6901983.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Samadi P, Saki S, Khoshinani HM, Sheykhhasan M. Therapeutic applications of mesenchymal stem cells: a comprehensive review. Curr Stem Cell Res Ther. 2020;

  8. Gnecchi M, Danieli P, Malpasso G, Ciuffreda MC. Paracrine mechanisms of mesenchymal stem cells in tissue repair. In: Mesenchymal Stem Cells: Springer; 2016. p. 123–46.

    Chapter  Google Scholar 

  9. Volarevic V, Gazdic M, Markovic BS, Jovicic N, Djonov V, Arsenijevic N. Mesenchymal stem cell-derived factors: immuno-modulatory effects and therapeutic potential. Biofactors (Oxford, England). 2017;43:633–44.

    Article  CAS  PubMed  Google Scholar 

  10. Marofi F, Hassanzadeh A, Solali S, Vahedi G, Mousavi Ardehaie R, Salarinasab S, Aliparasti MR, Ghaebi M, Farshdousti Hagh M. Epigenetic mechanisms are behind the regulation of the key genes associated with the osteoblastic differentiation of the mesenchymal stem cells: the role of zoledronic acid on tuning the epigenetic changes. J Cell Physiol. 2019;234:15108–22.

    Article  CAS  PubMed  Google Scholar 

  11. Zhao S, Wehner R, Bornhäuser M, Wassmuth R, Bachmann M, Schmitz M. Immunomodulatory properties of mesenchymal stromal cells and their therapeutic consequences for immune-mediated disorders. Stem Cells Dev. 2010;19:607–14.

    Article  CAS  PubMed  Google Scholar 

  12. Lim JY, Ryu DB, Lee SE, Park G, Min CK. Mesenchymal stem cells (MSCs) attenuate cutaneous sclerodermatous graft-versus-host disease (Scl-GVHD) through inhibition of immune cell infiltration in a mouse model. J Invest Dermatol. 2017;137:1895–904.

    Article  CAS  PubMed  Google Scholar 

  13. Gnecchi M, Ciuffreda MC, Mura M. Mesenchymal stromal cell secretome for tissue repair. Cell Eng Regen. 2020:641–66.

  14. Toh WS, Zhang B, Lai RC, Lim SK. Immune regulatory targets of mesenchymal stromal cell exosomes/small extracellular vesicles in tissue regeneration. Cytotherapy. 2018;20:1419–26.

    Article  CAS  PubMed  Google Scholar 

  15. Marolt Presen D, Traweger A, Gimona M, Redl H. Mesenchymal stromal/stem cell-based bone regeneration therapies: from cell transplantation and tissue engineering to therapeutic secretomes and extracellular vesicles. Front Bioeng Biotechnol. 2019;7:352.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Zhou Y, Yamamoto Y, Xiao Z, Ochiya T. The immunomodulatory functions of mesenchymal stromal/stem cells mediated via paracrine activity. J Clin Med. 2019;8:1025.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Lässer C, Alikhani VS, Ekström K, Eldh M, Paredes PT, Bossios A, Sjöstrand M, Gabrielsson S, Lötvall J, Valadi H. Human saliva, plasma and breast milk exosomes contain RNA: uptake by macrophages. J Transl Med. 2011;9:1–8.

    Article  Google Scholar 

  18. Nawaz M, Camussi G, Valadi H, Nazarenko I, Ekström K, Wang X, Principe S, Shah N, Ashraf NM, Fatima F. The emerging role of extracellular vesicles as biomarkers for urogenital cancers. Nat Rev Urol. 2014;11:688.

    Article  PubMed  Google Scholar 

  19. Simpson RJ, Lim JW, Moritz RL, Mathivanan S. Exosomes: proteomic insights and diagnostic potential. Expert Rev Proteomics. 2009;6:267–83.

    Article  CAS  PubMed  Google Scholar 

  20. Raposo G, Stoorvogel W. Extracellular vesicles: exosomes, microvesicles, and friends. J Cell Biol. 2013;200:373–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Marofi F, Vahedi G, Hasanzadeh A, Salarinasab S, Arzhanga P, Khademi B, Farshdousti Hagh M. Mesenchymal stem cells as the game-changing tools in the treatment of various organs disorders: mirage or reality? J Cell Physiol. 2019;234:1268–88.

    Article  CAS  PubMed  Google Scholar 

  22. Ahani-Nahayati M, Solali S, Shams Asenjan K, Movassaghpour Akbari AA, Talebi M, Zadi Heydarabad M, Baharaghdam S, Farshdousti Hagh M. Promoter methylation status of survival-related genes in MOLT- 4 cells co-cultured with bone marrow mesenchymal stem cells under hypoxic conditions. Cell J. 2018;20:188–94.

    PubMed  PubMed Central  Google Scholar 

  23. Mushahary D, Spittler A, Kasper C, Weber V, Charwat V. Isolation, cultivation, and characterization of human mesenchymal stem cells. Cytom Part A. 2018;93:19–31.

    Article  CAS  Google Scholar 

  24. Shariati A, Nemati R, Sadeghipour Y, Yaghoubi Y, Baghbani R, Javidi K, Zamani M, Hassanzadeh A. Mesenchymal stromal cells (MSCs) for neurodegenerative disease: a promising frontier. Eur J Cell Biol. 2020;99:151097.

    Article  CAS  PubMed  Google Scholar 

  25. Fan X-L, Zhang Y, Li X, Fu Q-L. Mechanisms underlying the protective effects of mesenchymal stem cell-based therapy. Cell Mol Life Sci. 2020:77;1–24.

  26. Jin HJ, Bae YK, Kim M, Kwon SJ, Jeon HB, Choi SJ, Kim SW, Yang YS, Oh W, Chang JW. Comparative analysis of human mesenchymal stem cells from bone marrow, adipose tissue, and umbilical cord blood as sources of cell therapy. Int J Mol Sci. 2013;14:17986–8001.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Jeon YJ, Kim J, Cho JH, Chung HM, Chae JI. Comparative analysis of human mesenchymal stem cells derived from bone marrow, placenta, and adipose tissue as sources of cell therapy. J Cell Biochem. 2016;117:1112–25.

    Article  CAS  PubMed  Google Scholar 

  28. Konala VBR, Bhonde R, Pal R. Secretome studies of mesenchymal stromal cells (MSCs) isolated from three tissue sources reveal subtle differences in potency. In Vitro Cell Dev Biol Anim. 2020:56;1–12.

  29. Wang Q, Yang Q, Wang Z, Tong H, Ma L, Zhang Y, Shan F, Meng Y, Yuan Z. Comparative analysis of human mesenchymal stem cells from fetal-bone marrow, adipose tissue, and Wharton’s jelly as sources of cell immunomodulatory therapy. Hum Vaccin Immunother. 2016;12:85–96.

    Article  PubMed  Google Scholar 

  30. Bernardo M, Emons J, Karperien M, Nauta A, Willemze R, Roelofs H, Romeo S, Marchini A, Rappold G, Vukicevic S. Human mesenchymal stem cells derived from bone marrow display a better chondrogenic differentiation compared with other sources. Connect Tissue Res. 2007;48:132–40.

    Article  CAS  PubMed  Google Scholar 

  31. Shen C, Yang C, Xu S, Zhao H. Comparison of osteogenic differentiation capacity in mesenchymal stem cells derived from human amniotic membrane (AM), umbilical cord (UC), chorionic membrane (CM), and decidua (DC). Cell Biosci. 2019;9:17.

    Article  PubMed  PubMed Central  Google Scholar 

  32. Han Y, Li X, Zhang Y, Han Y, Chang F, Ding J. Mesenchymal stem cells for regenerative medicine. Cells. 2019;8:886.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Abdi R, Fiorina P, Adra CN, Atkinson M, Sayegh MH. Immunomodulation by mesenchymal stem cells: a potential therapeutic strategy for type 1 diabetes. Diabetes. 2008;57:1759–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Marie JC, Liggitt D, Rudensky AY. Cellular mechanisms of fatal early-onset autoimmunity in mice with the T cell-specific targeting of transforming growth factor-β receptor. Immunity. 2006;25:441–54.

    Article  CAS  PubMed  Google Scholar 

  35. Swartzlander MD, Blakney AK, Amer LD, Hankenson KD, Kyriakides TR, Bryant SJ. Immunomodulation by mesenchymal stem cells combats the foreign body response to cell-laden synthetic hydrogels. Biomaterials. 2015;41:79–88.

    Article  CAS  PubMed  Google Scholar 

  36. Rigotti G, Charles-de-Sá L, Gontijo-de-Amorim NF, Takiya CM, Amable PR, Borojevic R, Benati D, Bernardi P, Sbarbati A. Expanded stem cells, stromal-vascular fraction, and platelet-rich plasma enriched fat: comparing results of different facial rejuvenation approaches in a clinical trial. Aesthet Surg J. 2016;36:261–70.

    Article  PubMed  PubMed Central  Google Scholar 

  37. Li F, Guo X, Chen S-Y. Function and therapeutic potential of mesenchymal stem cells in atherosclerosis. Front Cardiovasc Med. 2017;4:32.

    Article  PubMed  PubMed Central  Google Scholar 

  38. Luz-Crawford P, Kurte M, Bravo-Alegría J, Contreras R, Nova-Lamperti E, Tejedor G, Noël D, Jorgensen C, Figueroa F, Djouad F, Carrión F. Mesenchymal stem cells generate a CD4+CD25+Foxp3+ regulatory T cell population during the differentiation process of Th1 and Th17 cells. Stem Cell Res Ther. 2013;4:65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Salah RB, Snoussi M, Louati N, Donia C, Frikha F, Hela M, Zouhir B. The lymphoproliferative auto-immune syndrome: a rare cause of peripheral cytopenia. Electron J Gen Med. 2018:5;15.

  40. Umit EG, Baysal M, Bas V, Goze H, Asoglu V, Kirkizlar O, Demir AM. Value of extracellular high mobility group box 1 (HMGB1) in the clinical context of immune thrombocytopenia. J Clin Exp Invest. 2019;10:em00724.

    Article  Google Scholar 

  41. Meisel R, Brockers S, Heseler K, Degistirici Ö, Bülle H, Woite C, Stuhlsatz S, Schwippert W, Jäger M, Sorg R. Human but not murine multipotent mesenchymal stromal cells exhibit broad-spectrum antimicrobial effector function mediated by indoleamine 2, 3-dioxygenase. Leukemia. 2011;25:648–54.

    Article  CAS  PubMed  Google Scholar 

  42. Bai M, Zhang L, Fu B, Bai J, Zhang Y, Cai G, Bai X, Feng Z, Sun S, Chen X. IL-17A improves the efficacy of mesenchymal stem cells in ischemic-reperfusion renal injury by increasing Treg percentages by the COX-2/PGE2 pathway. Kidney Int. 2018;93:814–25.

    Article  CAS  PubMed  Google Scholar 

  43. Wang G, Cao K, Liu K, Xue Y, Roberts AI, Li F, Han Y, Rabson AB, Wang Y, Shi Y. Kynurenic acid, an IDO metabolite, controls TSG-6-mediated immunosuppression of human mesenchymal stem cells. Cell Death Differ. 2018;25:1209–23.

    Article  CAS  PubMed  Google Scholar 

  44. Sundrud MS, Koralov SB, Feuerer M, Calado DP, Kozhaya AE, Rhule-Smith A, Lefebvre RE, Unutmaz D, Mazitschek R, Waldner H. Halofuginone inhibits TH17 cell differentiation by activating the amino acid starvation response. Science (New York, N.Y.). 2009;324:1334–8.

    Article  CAS  PubMed  Google Scholar 

  45. Gu Y-z, Xue Q, Chen Y-j, Yu G-H, Shen Y, Wang M-y, Shi Q, Zhang X-G. Different roles of PD-L1 and FasL in immunomodulation mediated by human placenta-derived mesenchymal stem cells. Hum Immunol. 2013;74:267–76.

    Article  CAS  PubMed  Google Scholar 

  46. Yan Z, Zhuansun Y, Liu G, Chen R, Li J, Ran P. Mesenchymal stem cells suppress T cells by inducing apoptosis and through PD-1/B7-H1 interactions. Immunol Lett. 2014;162:248–55.

    Article  CAS  PubMed  Google Scholar 

  47. Mohammadzadeh A, Pourfathollah AA, Shahrokhi S, Hashemi SM, Moradi SLA, Soleimani M. Immunomodulatory effects of adipose-derived mesenchymal stem cells on the gene expression of major transcription factors of T cell subsets. Int Immunopharmacol. 2014;20:316–21.

    Article  CAS  PubMed  Google Scholar 

  48. Vignali DA, Collison LW, Workman CJ. How regulatory T cells work. Nat Rev Immunol. 2008;8:523–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Li MO, Sanjabi S, Flavell RA. Transforming growth factor-β controls development, homeostasis, and tolerance of T cells by regulatory T cell-dependent and-independent mechanisms. Immunity. 2006;25:455–71.

    Article  CAS  PubMed  Google Scholar 

  50. Chen PM, Liu KJ, Hsu PJ, Wei CF, Bai CH, Ho LJ, Sytwu HK, Yen BL. Induction of immunomodulatory monocytes by human mesenchymal stem cell-derived hepatocyte growth factor through ERK1/2. J Leukoc Biol. 2014;96:295–303.

    Article  PubMed  Google Scholar 

  51. Barnes PJ. Hepatocyte growth factor deficiency in COPD: a mechanism of emphysema and small airway fibrosis? Chest. 2014;146:1135–6.

    Article  PubMed  Google Scholar 

  52. Weiss ARR, Dahlke MH. Immunomodulation by mesenchymal stem cells (MSCs): mechanisms of action of living, apoptotic, and dead MSCs. Front Immunol. 2019;10:1191.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Kim N, Cho S-G. New strategies for overcoming limitations of mesenchymal stem cell-based immune modulation. Int J Stem Cells. 2015;8:54–68.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Naji A, Eitoku M, Favier B, Deschaseaux F, Rouas-Freiss N, Suganuma N. Biological functions of mesenchymal stem cells and clinical implications. Cell Mol Life Sci. 2019;76:3323–48.

    Article  CAS  PubMed  Google Scholar 

  55. Ullah M, Liu DD, Thakor AS. Mesenchymal stromal cell homing: mechanisms and strategies for improvement. iScience. 2019;15:421–38.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Ponte AL, Marais E, Gallay N, Langonné A, Delorme B, Hérault O, Charbord P, Domenech J. The in vitro migration capacity of human bone marrow mesenchymal stem cells: comparison of chemokine and growth factor chemotactic activities. Stem Cells. 2007;25:1737–45.

    Article  CAS  PubMed  Google Scholar 

  57. Bhakta S, Hong P, Koc O. The surface adhesion molecule CXCR4 stimulates mesenchymal stem cell migration to stromal cell-derived factor-1 in vitro but does not decrease apoptosis under serum deprivation. Cardiovasc Revasc Med. 2006;7:19–24.

    Article  PubMed  Google Scholar 

  58. Pourjafar M, Saidijam M, Mansouri K, Ghasemibasir H, Karimi Dermani F, Najafi R. All-trans retinoic acid preconditioning enhances proliferation, angiogenesis and migration of mesenchymal stem cell in vitro and enhances wound repair in vivo. Cell Prolif. 2017:50;50.

  59. Hassan M, Yazid MD, Yunus MHM, Chowdhury SR, Lokanathan Y, Idrus RBH, Ng AMH, Law JX. Large-scale expansion of human mesenchymal stem cells. Stem Cells Int. 2020;2020:9529465.

    Article  PubMed  PubMed Central  Google Scholar 

  60. Eggenhofer E, Luk F, Dahlke MH, Hoogduijn MJ. The life and fate of mesenchymal stem cells. Front Immunol. 2014;5:148.

    Article  PubMed  PubMed Central  Google Scholar 

  61. Tondreau T, Meuleman N, Delforge A, Dejeneffe M, Leroy R, Massy M, Mortier C, Bron D, Lagneaux L. Mesenchymal stem cells derived from CD133-positive cells in mobilized peripheral blood and cord blood: proliferation, Oct4 expression, and plasticity. Stem Cells. 2005;23:1105–12.

    Article  CAS  PubMed  Google Scholar 

  62. Alm JJ, Koivu HM, Heino TJ, Hentunen TA, Laitinen S, Aro HT. Circulating plastic adherent mesenchymal stem cells in aged hip fracture patients. J Orthop Res. 2010;28:1634–42.

    Article  CAS  PubMed  Google Scholar 

  63. Rochefort GY, Delorme B, Lopez A, Hérault O, Bonnet P, Charbord P, Eder V, Domenech J. Multipotential mesenchymal stem cells are mobilized into peripheral blood by hypoxia. Stem Cells. 2006;24:2202–8.

    Article  CAS  PubMed  Google Scholar 

  64. Chen Y, Xiang LX, Shao JZ, Pan RL, Wang YX, Dong XJ, Zhang GR. Recruitment of endogenous bone marrow mesenchymal stem cells towards injured liver. J Cell Mol Med. 2010;14:1494–508.

    Article  PubMed  Google Scholar 

  65. Gil-Ortega M, Garidou L, Barreau C, Maumus M, Breasson L, Tavernier G, García-Prieto CF, Bouloumié A, Casteilla L, Sengenès C. Native adipose stromal cells egress from adipose tissue in vivo: evidence during lymph node activation. Stem Cells. 2013;31:1309–20.

    Article  CAS  PubMed  Google Scholar 

  66. Ghorbanzade S, Naghib SM, Sadr A, Fateminia FS, Ghaffarinejad A, Majidzadeh-A K, Sanati-Nezhad A. Multifunctional magnetic nanoparticles-labeled mesenchymal stem cells for hyperthermia and bioimaging applications; 2019.

    Book  Google Scholar 

  67. Fu X, Liu G, Halim A, Ju Y, Luo Q, Song G. Mesenchymal stem cell migration and tissue repair. Cells. 2019;8:784.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Shi M, Li J, Liao L, Chen B, Li B, Chen L, Jia H, Zhao RC. Regulation of CXCR4 expression in human mesenchymal stem cells by cytokine treatment: role in homing efficiency in NOD/SCID mice. Haematologica. 2007;92:897–904.

    Article  PubMed  Google Scholar 

  69. Zhuang Y, Chen X, Xu M, Zhang LY, Xiang F. Chemokine stromal cell-derived factor 1/CXCL12 increases homing of mesenchymal stem cells to injured myocardium and neovascularization following myocardial infarction. Chin Med J. 2009;122:183–7.

    Article  PubMed  Google Scholar 

  70. Amsalem Y, Mardor Y, Feinberg MS, Landa N, Miller L, Daniels D, Ocherashvilli A, Holbova R, Yosef O, Barbash IM. Iron-oxide labeling and outcome of transplanted mesenchymal stem cells in the infarcted myocardium. Circulation. 2007;116:I-38–45.

    Article  CAS  Google Scholar 

  71. Peyvandi AA, Roozbahany NA, Peyvandi H, Abbaszadeh H-A, Majdinasab N, Faridan M, Niknazar S. Critical role of SDF-1/CXCR4 signaling pathway in stem cell homing in the deafened rat cochlea after acoustic trauma. Neural Regen Res. 2018;13:154–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Li X, Wei Z, Li B, Li J, Lv H, Wu L, Zhang H, Yang B, Zhu M, Jiang J. In vivo migration of Fe 3 O 4@ polydopamine nanoparticle-labeled mesenchymal stem cells to burn injury sites and their therapeutic effects in a rat model. Biomater Sci. 2019;7:2861–72.

    Article  CAS  PubMed  Google Scholar 

  73. De Becker A, Van Riet I. Homing and migration of mesenchymal stromal cells: how to improve the efficacy of cell therapy? World J Stem Cells. 2016;8:73.

    Article  PubMed  PubMed Central  Google Scholar 

  74. Rennert RC, Sorkin M, Garg RK, Gurtner GC. Stem cell recruitment after injury: lessons for regenerative medicine. Regen Med. 2012;7:833–50.

    Article  CAS  PubMed  Google Scholar 

  75. Cho KA, Ju SY, Ryu KH, Woo SY. Gene expression profile of mesenchymal stromal cells after co-culturing with injured liver tissue. Mol Med Rep. 2009;2:51–61.

    CAS  PubMed  Google Scholar 

  76. Fujii S, Miura Y, Fujishiro A, Shindo T, Shimazu Y, Hirai H, Tahara H, Takaori-Kondo A, Ichinohe T, Maekawa T. Graft-versus-host disease amelioration by human bone marrow mesenchymal stromal/stem cell-derived extracellular vesicles is associated with peripheral preservation of naive T cell populations. Stem Cells. 2018;36:434–45.

    Article  CAS  PubMed  Google Scholar 

  77. Martínez-Carrasco R, Sánchez-Abarca LI, Nieto-Gómez C, Martín García E, Sánchez-Guijo F, Argüeso P, Aijón J, Hernández-Galilea E, Velasco A. Subconjunctival injection of mesenchymal stromal cells protects the cornea in an experimental model of GVHD. Ocul Surf. 2019;17:285–94.

    Article  PubMed  Google Scholar 

  78. Tang B, Li X, Liu Y, Chen X, Li X, Chu Y, Zhu H, Liu W, Xu F, Zhou F, Zhang Y. The therapeutic effect of ICAM-1-overexpressing mesenchymal stem cells on acute graft-versus-host disease. Cell Physiol Biochem. 2018;46:2624–35.

    Article  CAS  PubMed  Google Scholar 

  79. Ma S, Chen X, Wang L, Wei Y, Ni Y, Chu Y, Liu Y, Zhu H, Zheng R, Zhang Y. Repairing effects of ICAM-1-expressing mesenchymal stem cells in mice with autoimmune thyroiditis. Exp Ther Med. 2017;13:1295–302.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Chen W, Li M, Li Z, Yan Z, Cheng H, Pan B, Cao J, Chen C, Zeng L, Xu K. CXCR4-transduced mesenchymal stem cells protect mice against graft-versus-host disease. Immunol Lett. 2012;143:161–9.

    Article  CAS  PubMed  Google Scholar 

  81. Lee H-M, Kim TS, Jo E-K. MiR-146 and miR-125 in the regulation of innate immunity and inflammation. BMB Rep. 2016;49:311–8.

    Article  PubMed  PubMed Central  Google Scholar 

  82. Le Blanc K, Frassoni F, Ball L, Locatelli F, Roelofs H, Lewis I, Lanino E, Sundberg B, Bernardo ME, Remberger M, Dini G, Egeler RM, Bacigalupo A, Fibbe W, Ringdén O. Mesenchymal stem cells for treatment of steroid-resistant, severe, acute graft-versus-host disease: a phase II study. Lancet (London, England). 2008;371:1579–86.

    Article  PubMed  Google Scholar 

  83. Gao L, Zhang Y, Hu B, Liu J, Kong P, Lou S, Su Y, Yang T, Li H, Liu Y, Zhang C, Gao L, Zhu L, Wen Q, Wang P, Chen X, Zhong J, Zhang X. Phase II multicenter, randomized, double-blind controlled study of efficacy and safety of umbilical cord-derived mesenchymal stromal cells in the prophylaxis of chronic graft-versus-host disease after HLA-haploidentical stem-cell transplantation. J Clin Oncol. 2016;34:2843–50.

    Article  CAS  PubMed  Google Scholar 

  84. Pan L, Lu M-P, Wang J-H, Xu M, Yang S-R. Immunological pathogenesis and treatment of systemic lupus erythematosus. World J Pediatr. 2020;16:19–30.

    Article  PubMed  Google Scholar 

  85. Sun L, Akiyama K, Zhang H, Yamaza T, Hou Y, Zhao S, Xu T, Le A, Shi S. Mesenchymal stem cell transplantation reverses multiorgan dysfunction in systemic lupus erythematosus mice and humans. Stem Cells. 2009;27:1421–32.

    Article  CAS  PubMed  Google Scholar 

  86. Sharma J, Hampton JM, Valiente GR, Wada T, Steigelman H, Young MC, Spurbeck RR, Blazek AD, Bösh S, Jarjour WN. Therapeutic development of mesenchymal stem cells or their extracellular vesicles to inhibit autoimmune-mediated inflammatory processes in systemic lupus erythematosus. Front Immunol. 2017;8:526.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Liang J, Zhang H, Hua B, Wang H, Lu L, Shi S, Hou Y, Zeng X, Gilkeson GS, Sun L. Allogenic mesenchymal stem cells transplantation in refractory systemic lupus erythematosus: a pilot clinical study. Ann Rheum Dis. 2010;69:1423–9.

    Article  PubMed  Google Scholar 

  88. Shi D, Li X, Chen H, Che N, Zhou S, Lu Z, Shi S, Sun L. High level of reactive oxygen species impaired mesenchymal stem cell migration via overpolymerization of F-actin cytoskeleton in systemic lupus erythematosus. Pathol Biol. 2014;62:382–90.

    Article  CAS  PubMed  Google Scholar 

  89. Wang D, Feng X, Lu L, Konkel JE, Zhang H, Chen Z, Li X, Gao X, Lu L, Shi S. A CD8 T cell/indoleamine 2, 3-dioxygenase axis is required for mesenchymal stem cell suppression of human systemic lupus erythematosus. Arthritis Rheumatol. 2014;66:2234–45.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Zhu Y, Feng X. Genetic contribution to mesenchymal stem cell dysfunction in systemic lupus erythematosus. Stem Cell Res Ther. 2018;9:1–6.

    Article  Google Scholar 

  91. Schena F, Gambini C, Gregorio A, Mosconi M, Reverberi D, Gattorno M, Casazza S, Uccelli A, Moretta L, Martini A. Interferon-γ–dependent inhibition of B cell activation by bone marrow–derived mesenchymal stem cells in a murine model of systemic lupus erythematosus. Arthritis Rheum. 2010;62:2776–86.

    Article  CAS  PubMed  Google Scholar 

  92. Tang X, Li W, Wen X, Zhang Z, Chen W, Yao G, Chen H, Wang D, Shi S, Sun L. Transplantation of dental tissue-derived mesenchymal stem cells ameliorates nephritis in lupus mice. Ann Transl Med. 2019;7:132.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Carrion F, Nova E, Ruiz C, Diaz F, Inostroza C, Rojo D, Mönckeberg G, Figueroa F. Autologous mesenchymal stem cell treatment increased T regulatory cells with no effect on disease activity in two systemic lupus erythematosus patients. Lupus. 2010;19:317–22.

    Article  CAS  PubMed  Google Scholar 

  94. Wang D, Huang S, Yuan X, Liang J, Xu R, Yao G, Feng X, Sun L. The regulation of the Treg/Th17 balance by mesenchymal stem cells in human systemic lupus erythematosus. Cell Mol Immunol. 2017;14:423–31.

    Article  CAS  PubMed  Google Scholar 

  95. Dulamea A. Mesenchymal stem cells in multiple sclerosis - translation to clinical trials. J Med Life. 2015;8:24–7.

    CAS  PubMed  PubMed Central  Google Scholar 

  96. Abbasi-Kangevari M, Ghamari SH, Safaeinejad F, Bahrami S, Niknejad H. Potential therapeutic features of human amniotic mesenchymal stem cells in multiple sclerosis: immunomodulation, inflammation suppression, angiogenesis promotion, oxidative stress inhibition, neurogenesis induction, MMPs regulation, and remyelination stimulation. Front Immunol. 2019;10:238.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Muri L, Leppert D, Grandgirard D, Leib SL. MMPs and ADAMs in neurological infectious diseases and multiple sclerosis. Cell Mol Life Sci. 2019;76:3097–116.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Pinheiro LL, de Lima AR, Martins DM, de Oliveira EHC, Souza MPC, de Carvalho Miranda CMF, Baleeiro Beltrão-Braga PC, Russo FB, Pignatari GC, da Silva Filho E, Branco É. Mesenchymal stem cells in dogs with demyelinating leukoencephalitis as an experimental model of multiple sclerosis. Heliyon. 2019;5:e01857.

    Article  PubMed  PubMed Central  Google Scholar 

  99. Bai L, Lennon DP, Eaton V, Maier K, Caplan AI, Miller SD, Miller RH. Human bone marrow-derived mesenchymal stem cells induce Th2-polarized immune response and promote endogenous repair in animal models of multiple sclerosis. Glia. 2009;57:1192–203.

    Article  PubMed  PubMed Central  Google Scholar 

  100. Laso-García F, Ramos-Cejudo J, Carrillo-Salinas FJ, Otero-Ortega L, Feliú A, Gómez-de Frutos M, Mecha M, Díez-Tejedor E, Guaza C, Gutiérrez-Fernández M. Therapeutic potential of extracellular vesicles derived from human mesenchymal stem cells in a model of progressive multiple sclerosis. PLoS One. 2018;13:e0202590.

    Article  PubMed  PubMed Central  Google Scholar 

  101. Soundara Rajan T, Giacoppo S, Diomede F, Bramanti P, Trubiani O, Mazzon E. Human periodontal ligament stem cells secretome from multiple sclerosis patients suppresses NALP3 inflammasome activation in experimental autoimmune encephalomyelitis. Int J Immunopathol Pharmacol. 2017;30:238–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Riordan NH, Morales I, Fernández G, Allen N, Fearnot NE, Leckrone ME, Markovich DJ, Mansfield D, Avila D, Patel AN, Kesari S, Paz Rodriguez J. Clinical feasibility of umbilical cord tissue-derived mesenchymal stem cells in the treatment of multiple sclerosis. J Transl Med. 2018;16:57.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Fernández O, Izquierdo G, Fernández V, Leyva L, Reyes V, Guerrero M, León A, Arnaiz C, Navarro G, Páramo MD, Cuesta A, Soria B, Hmadcha A, Pozo D, Fernandez-Montesinos R, Leal M, Ochotorena I, Gálvez P, Geniz MA, Barón FJ, Mata R, Medina C, Caparrós-Escudero C, Cardesa A, Cuende N. Adipose-derived mesenchymal stem cells (AdMSC) for the treatment of secondary-progressive multiple sclerosis: A triple blinded, placebo controlled, randomized phase I/II safety and feasibility study. PLoS One. 2018;13:e0195891.

    Article  PubMed  PubMed Central  Google Scholar 

  104. Connick P, Kolappan M, Crawley C, Webber DJ, Patani R, Michell AW, Du M-Q, Luan S-L, Altmann DR, Thompson AJ. Autologous mesenchymal stem cells for the treatment of secondary progressive multiple sclerosis: an open-label phase 2a proof-of-concept study. Lancet Neurol. 2012;11:150–6.

    Article  PubMed  PubMed Central  Google Scholar 

  105. Gohil K, Carramusa B. Ulcerative colitis and Crohn’s disease. P T. 2014;39:576–7.

    PubMed  PubMed Central  Google Scholar 

  106. Faleiro R, Liu J, Karunarathne D, Edmundson A, Winterford C, Nguyen TH, Simms LA, Radford-Smith G, Wykes M. Crohn’s disease is facilitated by a disturbance of programmed death-1 ligand 2 on blood dendritic cells. Clin Transl Immunol. 2019;8:e01071.

    Article  Google Scholar 

  107. Mao F, Tu Q, Wang L, Chu F, Li X, Li HS, Xu W. Mesenchymal stem cells and their therapeutic applications in inflammatory bowel disease. Oncotarget. 2017;8:38008–21.

    Article  PubMed  PubMed Central  Google Scholar 

  108. Mannon PJ. Remestemcel-L: human mesenchymal stem cells as an emerging therapy for Crohn's disease. Expert Opin Biol Ther. 2011;11:1249–56.

    Article  PubMed  Google Scholar 

  109. González MA, Gonzalez–Rey E, Rico L, Büscher D, Delgado M. Adipose-derived mesenchymal stem cells alleviate experimental colitis by inhibiting inflammatory and autoimmune responses. Gastroenterology. 2009;136:978–89.

    Article  PubMed  Google Scholar 

  110. Xie M, Qin H, Luo Q, He X, He X, Lan P, Lian L. Comparison of adipose-derived and bone marrow mesenchymal stromal cells in a murine model of Crohn’s disease. Dig Dis Sci. 2017;62:115–23.

    Article  CAS  PubMed  Google Scholar 

  111. Gao J-G, Yu M-S, Zhang M-M, Gu X-W, Ren Y, Zhou X-X, Chen D, Yan T-L, Li Y-M, Jin X. Adipose-derived mesenchymal stem cells alleviate TNBS-induced colitis in rats by influencing intestinal epithelial cell regeneration, Wnt signaling, and T cell immunity. World J Gastroenterol. 2020;26:3750.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Cosín-Roger J, Ortiz-Masiá D, Calatayud S, Hernández C, Esplugues JV, Barrachina MD. The activation of Wnt signaling by a STAT6-dependent macrophage phenotype promotes mucosal repair in murine IBD. Mucosal Immunol. 2016;9:986–98.

    Article  PubMed  Google Scholar 

  113. Ciccocioppo R, Gallia A, Sgarella A, Kruzliak P, Gobbi PG, Corazza GR. Long-term follow-up of Crohn disease fistulas after local injections of bone marrow-derived Mesenchymal stem cells. Mayo Clin Proc. 2015;90:747–55.

    Article  PubMed  Google Scholar 

  114. Panés J, García-Olmo D, Van Assche G, Colombel JF, Reinisch W, Baumgart DC, Dignass A, Nachury M, Ferrante M, Kazemi-Shirazi L. Expanded allogeneic adipose-derived mesenchymal stem cells (Cx601) for complex perianal fistulas in Crohn's disease: a phase 3 randomised, double-blind controlled trial. Lancet. 2016;388:1281–90.

    Article  PubMed  Google Scholar 

  115. Forbes GM, Sturm MJ, Leong RW, Sparrow MP, Segarajasingam D, Cummins AG, Phillips M, Herrmann RP. A phase 2 study of allogeneic mesenchymal stromal cells for luminal Crohn's disease refractory to biologic therapy. Clin Gastroenterol Hepatol. 2014;12:64–71.

    Article  PubMed  Google Scholar 

  116. Vlaar AP, Toy P, Fung M, Looney MR, Juffermans NP, Bux J, Bolton-Maggs P, Peters AL, Silliman CC, Kor DJ. A consensus redefinition of transfusion-related acute lung injury. Transfusion. 2019;59:2465–76.

    Article  PubMed  PubMed Central  Google Scholar 

  117. Cleary SJ, Kwaan N, Tian JJ, Calabrese DR, Mallavia B, Magnen M, Greenland JR, Urisman A, Singer JP, Hays SR, Kukreja J, Hay AM, Howie HL, Toy P, Lowell CA, Morrell CN, Zimring JC, Looney MR. Complement activation on endothelium initiates antibody-mediated acute lung injury. J Clin Invest. 2020;130:5909–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Li X, Michaeloudes C, Zhang Y, Wiegman CH, Adcock IM, Lian Q, Mak JCW, Bhavsar PK, Chung KF. Mesenchymal stem cells alleviate oxidative stress-induced mitochondrial dysfunction in the airways. J Allergy Clin Immunol. 2018;141:1634–1645.e1635.

    Article  CAS  PubMed  Google Scholar 

  119. Khatri M, Richardson LA, Meulia T. Mesenchymal stem cell-derived extracellular vesicles attenuate influenza virus-induced acute lung injury in a pig model. Stem Cell Res Ther. 2018;9:17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Li L, Dong L, Zhang J, Gao F, Hui J, Yan J. Mesenchymal stem cells with downregulated hippo signaling attenuate lung injury in mice with lipopolysaccharide-induced acute respiratory distress syndrome. Int J Mol Med. 2019;43:1241–52.

    CAS  PubMed  Google Scholar 

  121. Silva JD, Lopes-Pacheco M, Paz AHR, Cruz FF, Melo EB, de Oliveira MV, Xisto DG, Capelozzi VL, Morales MM, Pelosi P, Cirne-Lima E, Rocco PRM. Mesenchymal stem cells from bone marrow, adipose tissue, and lung tissue differentially mitigate lung and distal organ damage in experimental acute respiratory distress syndrome. Crit Care Med. 2018;46:e132–40.

    Article  PubMed  Google Scholar 

  122. Pedrazza L, Cunha AA, Luft C, Nunes NK, Schimitz F, Gassen RB, Breda RV, Donadio MV, de Souza Wyse AT, Pitrez PMC, Rosa JL, de Oliveira JR. Mesenchymal stem cells improves survival in LPS-induced acute lung injury acting through inhibition of NETs formation. J Cell Physiol. 2017;232:3552–64.

    Article  CAS  PubMed  Google Scholar 

  123. Yi X, Wei X, Lv H, An Y, Li L, Lu P, Yang Y, Zhang Q, Yi H, Chen G. Exosomes derived from microRNA-30b-3p-overexpressing mesenchymal stem cells protect against lipopolysaccharide-induced acute lung injury by inhibiting SAA3. Exp Cell Res. 2019;383:111454.

    Article  CAS  PubMed  Google Scholar 

  124. Guan XJ, Song L, Han FF, Cui ZL, Chen X, Guo XJ, Xu WG. Mesenchymal stem cells protect cigarette smoke-damaged lung and pulmonary function partly via VEGF-VEGF receptors. J Cell Biochem. 2013;114:323–35.

    Article  CAS  PubMed  Google Scholar 

  125. Zhen G, Xue Z, Zhao J, Gu N, Tang Z, Xu Y, Zhang Z. Mesenchymal stem cell transplantation increases expression of vascular endothelial growth factor in papain-induced emphysematous lungs and inhibits apoptosis of lung cells. Cytotherapy. 2010;12:605–14.

    Article  CAS  PubMed  Google Scholar 

  126. Chen S, Cui G, Peng C, Lavin MF, Sun X, Zhang E, Yang Y, Guan Y, Du Z, Shao H. Transplantation of adipose-derived mesenchymal stem cells attenuates pulmonary fibrosis of silicosis via anti-inflammatory and anti-apoptosis effects in rats. Stem Cell Res Ther. 2018;9:110.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Willis GR, Fernandez-Gonzalez A, Anastas J, Vitali SH, Liu X, Ericsson M, Kwong A, Mitsialis SA, Kourembanas S. Mesenchymal stromal cell exosomes ameliorate experimental bronchopulmonary dysplasia and restore lung function through macrophage immunomodulation. Am J Respir Crit Care Med. 2018;197:104–16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Rubio GA, Elliot SJ, Wikramanayake TC, Xia X, Pereira-Simon S, Thaller SR, Glinos GD, Jozic I, Hirt P, Pastar I, Tomic-Canic M, Glassberg MK. Mesenchymal stromal cells prevent bleomycin-induced lung and skin fibrosis in aged mice and restore wound healing. J Cell Physiol. 2018;233:5503–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Lan YW, Choo KB, Chen CM, Hung TH, Chen YB, Hsieh CH, Kuo HP, Chong KY. Hypoxia-preconditioned mesenchymal stem cells attenuate bleomycin-induced pulmonary fibrosis. Stem Cell Res Ther. 2015;6:97.

    Article  PubMed  PubMed Central  Google Scholar 

  130. Xiang B, Chen L, Wang X, Zhao Y, Wang Y, Xiang C. Transplantation of menstrual blood-derived mesenchymal stem cells promotes the repair of LPS-induced acute lung injury. Int J Mol Sci. 2017:18;18.

  131. Zhong H, Fan XL, Fang SB, Lin YD, Wen W, Fu QL. Human pluripotent stem cell-derived mesenchymal stem cells prevent chronic allergic airway inflammation via TGF-β1-Smad2/Smad3 signaling pathway in mice. Mol Immunol. 2019;109:51–7.

    Article  CAS  PubMed  Google Scholar 

  132. Lee FY, Chen KH, Wallace CG, Sung PH, Sheu JJ, Chung SY, Chen YL, Lu HI, Ko SF, Sun CK, Chiang HJ, Chang HW, Lee MS, Yip HK. Xenogeneic human umbilical cord-derived mesenchymal stem cells reduce mortality in rats with acute respiratory distress syndrome complicated by sepsis. Oncotarget. 2017;8:45626–42.

    Article  PubMed  PubMed Central  Google Scholar 

  133. Chen J, Si L, Zhou L, Deng Y. Role of bone marrow mesenchymal stem cells in the development of PQ-induced pulmonary fibrosis. Mol Med Rep. 2019;19:3283–90.

    CAS  PubMed  Google Scholar 

  134. Moroncini G, Paolini C, Orlando F, Capelli C, Grieco A, Tonnini C, Agarbati S, Mondini E, Saccomanno S, Goteri G, Svegliati Baroni S, Provinciali M, Introna M, Del Papa N, Gabrielli A. Mesenchymal stromal cells from human umbilical cord prevent the development of lung fibrosis in immunocompetent mice. PLoS One. 2018;13:e0196048.

    Article  PubMed  PubMed Central  Google Scholar 

  135. Han XP, Zhang FQ, Tan XS, Liu L, Ma WX, Ou-Yang HF, Wu CG. EPO modified MSCs can inhibit asthmatic airway remodeling in an animal model. J Cell Biochem. 2018;119:1008–16.

    Article  CAS  PubMed  Google Scholar 

  136. Liu F, Lin Q, Liu Z. A study on the role of apoptotic human umbilical cord mesenchymal stem cells in bleomycin-induced acute lung injury in rat models. Eur Rev Med Pharmacol Sci. 2016;20:969–82.

    PubMed  Google Scholar 

  137. Abreu SC, Xisto DG, de Oliveira TB, Blanco NG, de Castro LL, Kitoko JZ, Olsen PC, Lopes-Pacheco M, Morales MM, Weiss DJ, Rocco PRM. Serum from asthmatic mice potentiates the therapeutic effects of mesenchymal stromal cells in experimental allergic asthma. Stem Cells Transl Med. 2019;8:301–12.

    Article  CAS  PubMed  Google Scholar 

  138. Jin Z, Wang Q, Bi H, Zhou K, He L, He X, Wu Q, Pan X. Effects and possible mechanisms of mesenchymal stem cell transplantation on emphysema in rats. Zhonghua Yi Xue Za Zhi. 2015;95:1731–5.

    CAS  PubMed  Google Scholar 

  139. Chang Q, Tian X, Huo R, Liu D, Chen J, Wang X, Tian X. Therapeutic effect of human umbilical cord mesenchymal stem cells on airway remodeling in an asthma model of rat and its possible mechanism. Zhonghua Yi Xue Za Zhi. 2018;98:2258–63.

    CAS  PubMed  Google Scholar 

  140. Soliman MG, Mansour HA, Hassan WA, El-Sayed RA, Hassaan NA. Mesenchymal stem cells therapeutic potential alleviate lipopolysaccharide-induced acute lung injury in rat model. J Biochem Mol Toxicol. 2018;32:e22217.

    Article  PubMed  Google Scholar 

  141. Curley GF, Hayes M, Ansari B, Shaw G, Ryan A, Barry F, O'Brien T, O'Toole D, Laffey JG. Mesenchymal stem cells enhance recovery and repair following ventilator-induced lung injury in the rat. Thorax. 2012;67:496–501.

    Article  PubMed  Google Scholar 

  142. Moodley Y, Sturm M, Shaw K, Shimbori C, Tan DB, Kolb M, Graham R. Human mesenchymal stem cells attenuate early damage in a ventilated pig model of acute lung injury. Stem Cell Res. 2016;17:25–31.

    Article  CAS  PubMed  Google Scholar 

  143. Asmussen S, Ito H, Traber DL, Lee JW, Cox RA, Hawkins HK, McAuley DF, McKenna DH, Traber LD, Zhuo H, Wilson J, Herndon DN, Prough DS, Liu KD, Matthay MA, Enkhbaatar P. Human mesenchymal stem cells reduce the severity of acute lung injury in a sheep model of bacterial pneumonia. Thorax. 2014;69:819–25.

    Article  PubMed  Google Scholar 

  144. Zhu H, Xiong Y, Xia Y, Zhang R, Tian D, Wang T, Dai J, Wang L, Yao H, Jiang H. Therapeutic effects of human umbilical cord-derived mesenchymal stem cells in acute lung injury mice. Sci Rep. 2017;7:1–11.

    Google Scholar 

  145. Du Y-m, Zhuansun Y-x, Chen R, Lin L, Lin Y, Li J-g. Mesenchymal stem cell exosomes promote immunosuppression of regulatory T cells in asthma. Exp Cell Res. 2018;363:114–20.

    Article  CAS  PubMed  Google Scholar 

  146. Li L, Dong L, Hui J, Gao F, Wang Q, Yang L, Zhang J, Yan J. Under-expression of LATS1 promotes the differentiation, proliferation and migration of mesenchymal stem cells by inhibition the Hippo signaling pathway in vitro. Zhonghua Wei Zhong Bing Ji Jiu Yi Xue. 2017;29:731–7.

    PubMed  Google Scholar 

  147. Chen X, Xu C-X, Liang H, Xi Z, Pan J, Yang Y, Sun Q, Yang G, Sun Y, Bian L. Bone marrow mesenchymal stem cells transplantation alleviates brain injury after intracerebral hemorrhage in mice through the Hippo signaling pathway. Aging. 2020;12:6306–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Zhou T, Chen YL. The functional mechanisms of miR-30b-5p in acute lung injury in children. Med Sci Monit. 2019;25:40–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Fen L, Rong J, Zhenguo Z, Ning Z, Liang X, Cheng N, Kejian Q. The expression changes in microRNA-132 in the lipopolysaccharide-induced inflammation of rat alveolar macrophages. Zhonghua Wei Zhong Bing Ji Jiu Yi Xue. 2014;26:80–3.

    PubMed  Google Scholar 

  150. Islam MN, Das SR, Emin MT, Wei M, Sun L, Westphalen K, Rowlands DJ, Quadri SK, Bhattacharya S, Bhattacharya J. Mitochondrial transfer from bone-marrow-derived stromal cells to pulmonary alveoli protects against acute lung injury. Nat Med. 2012;18:759–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Zeng Z, Gong H, Li Y, Nie Z, Jie K, Zhan Y, Nie C, Liu F, Ding C, Shao Q. Effect of Shenfu injection on expression of lipopolysaccharide--induced microRNA-146a in alveolar macrophages. Zhongguo Wei Zhong Bing Ji Jiu Yi Xue. 2012;24:166–9.

    PubMed  Google Scholar 

  152. Paliwal S, Chaudhuri R, Agrawal A, Mohanty S. Regenerative abilities of mesenchymal stem cells through mitochondrial transfer. J Biomed Sci. 2018;25:31.

    Article  PubMed  PubMed Central  Google Scholar 

  153. Wilson JG, Liu KD, Zhuo H, Caballero L, McMillan M, Fang X, Cosgrove K, Vojnik R, Calfee CS, Lee JW, Rogers AJ, Levitt J, Wiener-Kronish J, Bajwa EK, Leavitt A, McKenna D, Thompson BT, Matthay MA. Mesenchymal stem (stromal) cells for treatment of ARDS: a phase 1 clinical trial. Lancet Respir Med. 2015;3:24–32.

    Article  PubMed  Google Scholar 

  154. Velavan TP, Meyer CG. The COVID-19 epidemic. Tropical Med Int Health. 2020;25:278.

    Article  CAS  Google Scholar 

  155. Apolone G, Montomoli E, Manenti A, Boeri M, Sabia F, Hyseni I, Mazzini L, Martinuzzi D, Cantone L, Milanese G. Unexpected detection of SARS-CoV-2 antibodies in the prepandemic period in Italy. Tumori J. 2020:0300891620974755.

  156. Ni W, Yang X, Yang D, Bao J, Li R, Xiao Y, Hou C, Wang H, Liu J, Yang D. Role of angiotensin-converting enzyme 2 (ACE2) in COVID-19. Crit Care. 2020;24:1–10.

    Article  Google Scholar 

  157. Sharquie IK. BCG is a good immunotherapeutic agent for viral and autoimmune diseases: is it a new weapon against coronavirus (COVID-19)? Electron J Gen Med. 2020:17;17.

  158. Ragab D, Salah Eldin H, Taeimah M, Khattab R, Salem R. The COVID-19 cytokine storm; what we know so far. Front Immunol. 2020;11:1446.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. Guo YR, Cao QD, Hong ZS, Tan YY, Chen SD, Jin HJ, Tan KS, Wang DY, Yan Y. The origin, transmission and clinical therapies on coronavirus disease 2019 (COVID-19) outbreak - an update on the status. Mil Med Res. 2020;7:11.

    CAS  PubMed  PubMed Central  Google Scholar 

  160. Kavianpour M, Saleh M, Verdi J. The role of mesenchymal stromal cells in immune modulation of COVID-19: focus on cytokine storm. Stem Cell Res Ther. 2020;11:404.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Leng Z, Zhu R, Hou W, Feng Y, Yang Y, Han Q, Shan G, Meng F, Du D, Wang S, Fan J, Wang W, Deng L, Shi H, Li H, Hu Z, Zhang F, Gao J, Liu H, Li X, Zhao Y, Yin K, He X, Gao Z, Wang Y, Yang B, Jin R, Stambler I, Lim LW, Su H, Moskalev A, Cano A, Chakrabarti S, Min KJ, Ellison-Hughes G, Caruso C, Jin K, Zhao RC. Transplantation of ACE2(−) mesenchymal stem cells improves the outcome of patients with COVID-19 pneumonia. Aging Dis. 2020;11:216–28.

    Article  PubMed  PubMed Central  Google Scholar 

  162. Shu L, Niu C, Li R, Huang T, Wang Y, Huang M, Ji N, Zheng Y, Chen X, Shi L, Wu M, Deng K, Wei J, Wang X, Cao Y, Yan J, Feng G. Treatment of severe COVID-19 with human umbilical cord mesenchymal stem cells. Stem Cell Res Ther. 2020;11:361.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. Liang B, Chen J, Li T, Wu H, Yang W, Li Y, Li J, Yu C, Nie F, Ma Z, Yang M, Xiao M, Nie P, Gao Y, Qian C, Hu M. Clinical remission of a critically ill COVID-19 patient treated by human umbilical cord mesenchymal stem cells: A case report. Medicine (Baltimore). 2020;99:e21429.

    Article  CAS  PubMed  Google Scholar 

  164. Zhang Y, Ding J, Ren S, Wang W, Yang Y, Li S, Meng M, Wu T, Liu D, Tian S, Tian H, Chen S, Zhou C. Intravenous infusion of human umbilical cord Wharton’s jelly-derived mesenchymal stem cells as a potential treatment for patients with COVID-19 pneumonia. Stem Cell Res Ther. 2020;11:207.

    Article  PubMed  PubMed Central  Google Scholar 

  165. Sengupta V, Sengupta S, Lazo A, Woods P, Nolan A, Bremer N. Exosomes derived from bone marrow mesenchymal stem cells as treatment for severe COVID-19. Stem Cells Dev. 2020;29:747–54.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. Sadeghi S, Soudi S, Shafiee A, Hashemi SM. Mesenchymal stem cell therapies for COVID-19: current status and mechanism of action. Life Sci. 2020;262:118493.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  167. Tao S-C, Yuan T, Zhang Y-L, Yin W-J, Guo S-C, Zhang C-Q. 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;7:180.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  168. Fernandez-Pernas P, Rodríguez-Lesende I, de la Fuente A, Mateos J, Fuentes I, De Toro J, Blanco FJ, Arufe MC. CD105+-mesenchymal stem cells migrate into osteoarthritis joint: An animal model. PLoS One. 2017;12:e0188072.

    Article  PubMed  PubMed Central  Google Scholar 

  169. Khatab S, van Osch GJ, Kops N, Bastiaansen-Jenniskens YM, Bos PK, Verhaar JA, Bernsen MR, van Buul GM. Mesenchymal stem cell secretome reduces pain and prevents cartilage damage in a murine osteoarthritis model. Eur Cells Mater. 2018;36:218–30.

    Article  CAS  Google Scholar 

  170. Lee K, Park N, Jung H, Rim YA, Nam Y, Lee J, Park SH, Ju JH. Mesenchymal stem cells ameliorate experimental arthritis via expression of interleukin-1 receptor antagonist. PLoS One. 2018;13:e0193086.

    Article  PubMed  PubMed Central  Google Scholar 

  171. Abd-Elhalem SS, Haggag NZ, El-Shinnawy NA. Bone marrow mesenchymal stem cells suppress IL-9 in adjuvant-induced arthritis. Autoimmunity. 2018;51:25–34.

    Article  CAS  PubMed  Google Scholar 

  172. Neybecker P, Henrionnet C, Pape E, Mainard D, Galois L, Loeuille D, Gillet P, Pinzano A. In vitro and in vivo potentialities for cartilage repair from human advanced knee osteoarthritis synovial fluid-derived mesenchymal stem cells. Stem Cell Res Ther. 2018;9:329.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  173. Yun S, Ku SK, Kwon YS. Adipose-derived mesenchymal stem cells and platelet-rich plasma synergistically ameliorate the surgical-induced osteoarthritis in Beagle dogs. J Orthop Surg Res. 2016;11:9.

    Article  PubMed  PubMed Central  Google Scholar 

  174. Kim H, Yang G, Park J, Choi J, Kang E, Lee BK. Therapeutic effect of mesenchymal stem cells derived from human umbilical cord in rabbit temporomandibular joint model of osteoarthritis. Sci Rep. 2019;9:13854.

    Article  PubMed  PubMed Central  Google Scholar 

  175. Chen Z, Wang H, Xia Y, Yan F, Lu Y. Therapeutic potential of mesenchymal cell-derived miRNA-150-5p-expressing exosomes in rheumatoid arthritis mediated by the modulation of MMP14 and VEGF. J Immunol (Baltimore, Md. : 1950). 2018;201:2472–82.

    Article  CAS  Google Scholar 

  176. Ichiseki T, Shimazaki M, Ueda Y, Ueda S, Tsuchiya M, Souma D, Kaneuji A, Kawahara N. Intraarticularly-injected mesenchymal stem cells stimulate anti-inflammatory molecules and inhibit pain related protein and chondrolytic enzymes in a monoiodoacetate-induced rat arthritis model. Int J Mol Sci. 2018:19;19.

  177. Chen W, Sun Y, Gu X, Hao Y, Liu X, Lin J, Chen J, Chen S. Conditioned medium of mesenchymal stem cells delays osteoarthritis progression in a rat model by protecting subchondral bone, maintaining matrix homeostasis, and enhancing autophagy. J Tissue Eng Regen Med. 2019;13:1618–28.

    Article  CAS  PubMed  Google Scholar 

  178. Haikal SM, Abdeltawab NF, Rashed LA, Abd El-Galil TI, Elmalt HA, Amin MA. Combination therapy of mesenchymal stromal cells and interleukin-4 attenuates rheumatoid arthritis in a collagen-induced murine model. Cells. 2019;8:823.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  179. Liu Y, Lin L, Zou R, Wen C, Wang Z, Lin F. MSC-derived exosomes promote proliferation and inhibit apoptosis of chondrocytes via lncRNA-KLF3-AS1/miR-206/GIT1 axis in osteoarthritis. Cell Cycle. 2018;17:2411–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  180. Li F, Li X, Liu G, Gao C, Li X. Bone marrow mesenchymal stem cells decrease the expression of RANKL in collagen-induced arthritis rats via reducing the levels of IL-22. J Immunol Res. 2019;2019:8459281.

    Article  PubMed  PubMed Central  Google Scholar 

  181. Ma D, Xu K, Zhang G, Liu Y, Gao J, Tian M, Wei C, Li J, Zhang L. Immunomodulatory effect of human umbilical cord mesenchymal stem cells on T lymphocytes in rheumatoid arthritis. Int Immunopharmacol. 2019;74:105687.

    Article  CAS  PubMed  Google Scholar 

  182. Toghraie FS, Chenari N, Gholipour MA, Faghih Z, Torabinejad S, Dehghani S, Ghaderi A. Treatment of osteoarthritis with infrapatellar fat pad derived mesenchymal stem cells in rabbit. Knee. 2011;18:71–5.

    Article  CAS  PubMed  Google Scholar 

  183. Wu CC, Liu FL, Sytwu HK, Tsai CY, Chang DM. CD146+ mesenchymal stem cells display greater therapeutic potential than CD146- cells for treating collagen-induced arthritis in mice. Stem Cell Res Ther. 2016;7:23.

    Article  PubMed  PubMed Central  Google Scholar 

  184. Wu J, Kuang L, Chen C, Yang J, Zeng WN, Li T, Chen H, Huang S, Fu Z, Li J, Liu R, Ni Z, Chen L, Yang L. miR-100-5p-abundant exosomes derived from infrapatellar fat pad MSCs protect articular cartilage and ameliorate gait abnormalities via inhibition of mTOR in osteoarthritis. Biomaterials. 2019;206:87–100.

    Article  CAS  PubMed  Google Scholar 

  185. Frisbie DD, Kisiday JD, Kawcak CE, Werpy NM, McIlwraith CW. Evaluation of adipose-derived stromal vascular fraction or bone marrow-derived mesenchymal stem cells for treatment of osteoarthritis. J Orthop Res. 2009;27:1675–80.

    Article  PubMed  Google Scholar 

  186. Yan X, Cen Y, Wang Q. Mesenchymal stem cells alleviate experimental rheumatoid arthritis through microRNA-regulated IκB expression. Sci Rep. 2016;6:28915.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  187. Cui YP, Cao YP, Liu H, Yang X, Meng ZC, Wang R. Bone marrow mesenchymal stem cells in Sprague-Dawley rat model of osteoarthritis. Beijing Da Xue Xue Bao Yi Xue Ban. 2015;47:211–8.

    CAS  PubMed  Google Scholar 

  188. Xia Q, Zhu S, Wu Y, Wang J, Cai Y, Chen P, Li J, Heng BC, Ouyang HW, Lu P. Intra-articular transplantation of atsttrin-transduced mesenchymal stem cells ameliorate osteoarthritis development. Stem Cells Transl Med. 2015;4:523–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  189. Liu R, Li X, Zhang Z, Zhou M, Sun Y, Su D, Feng X, Gao X, Shi S, Chen W, Sun L. Allogeneic mesenchymal stem cells inhibited T follicular helper cell generation in rheumatoid arthritis. Sci Rep. 2015;5:12777.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  190. Nazemian V, Manaheji H, Sharifi AM, Zaringhalam J. Long term treatment by mesenchymal stem cells conditioned medium modulates cellular, molecular and behavioral aspects of adjuvant-induced arthritis. Cell Mol Biol (Noisy-le-Grand, France). 2018;64:19–26.

    Article  Google Scholar 

  191. Stancker TG, Vieira SS, Serra AJ, do Nascimento Lima R, Feliciano RDS, Silva JA Jr, Santos SAD, Vieira MADS, Simões MCB, Leal-Junior EC, de Tarso Camillo de Carvalho P. Can photobiomodulation associated with implantation of mesenchymal adipose-derived stem cells attenuate the expression of MMPs and decrease degradation of type II collagen in an experimental model of osteoarthritis? Lasers Med Sci. 2018;33:1073–84.

    Article  PubMed  Google Scholar 

  192. Grigolo B, Lisignoli G, Desando G, Cavallo C, Marconi E, Tschon M, Giavaresi G, Fini M, Giardino R, Facchini A. Osteoarthritis treated with mesenchymal stem cells on hyaluronan-based scaffold in rabbit, tissue engineering. Part C Methods. 2009;15:647–58.

    Article  CAS  Google Scholar 

  193. Wang XD, Wan XC, Liu AF, Li R, Wei Q. Effects of umbilical cord mesenchymal stem cells loaded with graphene oxide granular lubrication on cytokine levels in animal models of knee osteoarthritis. Int Orthop. 2021;45:381–90.

    Article  PubMed  Google Scholar 

  194. Tseng WJ, Huang S-W, Fang C-H, Hsu L-T, Chen C-Y, Shen H-H, Chang J, Sun J-S, Lin F-H. Treatment of osteoarthritis with collagen-based scaffold: a porcine animal model with xenograft mesenchymal stem cells. Histol Histopathol. 2018;33:1271–86.

    CAS  PubMed  Google Scholar 

  195. McGonagle D, Baboolal TG, Jones E. Native joint-resident mesenchymal stem cells for cartilage repair in osteoarthritis. Nat Rev Rheumatol. 2017;13:719–30.

    Article  CAS  PubMed  Google Scholar 

  196. Wang Y, Yu D, Liu Z, Zhou F, Dai J, Wu B, Zhou J, Heng BC, Zou XH, Ouyang H. Exosomes from embryonic mesenchymal stem cells alleviate osteoarthritis through balancing synthesis and degradation of cartilage extracellular matrix. Stem Cell Res Ther. 2017;8:1–13.

    Article  Google Scholar 

  197. Gonzalo-Gil E, Pérez-Lorenzo MJ, Galindo M, Díaz de la Guardia R, López-Millán B, Bueno C, Menéndez P, Pablos JL, Criado G. Human embryonic stem cell-derived mesenchymal stromal cells ameliorate collagen-induced arthritis by inducing host-derived indoleamine 2,3 dioxygenase. Arthritis Res Ther. 2016;18:77.

    Article  PubMed  PubMed Central  Google Scholar 

  198. Martel-Pelletier J, Pelletier JP, Fahmi H. Cyclooxygenase-2 and prostaglandins in articular tissues. Semin Arthritis Rheum. 2003;33:155–67.

    Article  CAS  PubMed  Google Scholar 

  199. van Buul GM, Siebelt M, Leijs MJ, Bos PK, Waarsing JH, Kops N, Weinans H, Verhaar JA, Bernsen MR, van Osch GJ. Mesenchymal stem cells reduce pain but not degenerative changes in a mono-iodoacetate rat model of osteoarthritis. J Orthop Res. 2014;32:1167–74.

    Article  PubMed  Google Scholar 

  200. Gu Y, Shi S. Transplantation of gingiva-derived mesenchymal stem cells ameliorates collagen-induced arthritis. Arthritis Res Ther. 2016;18:262.

    Article  PubMed  PubMed Central  Google Scholar 

  201. Garimella MG, Kour S, Piprode V, Mittal M, Kumar A, Rani L, Pote ST, Mishra GC, Chattopadhyay N, Wani MR. Adipose-derived mesenchymal stem cells prevent systemic bone loss in collagen-induced arthritis. J Immunol (Baltimore, Md. : 1950). 2015;195:5136–48.

    Article  CAS  Google Scholar 

  202. Shin TH, Kim HS, Kang TW, Lee BC, Lee HY, Kim YJ, Shin JH, Seo Y, Won Choi S, Lee S, Shin K, Seo KW, Kang KS. Human umbilical cord blood-stem cells direct macrophage polarization and block inflammasome activation to alleviate rheumatoid arthritis. Cell Death Dis. 2016;7:e2524.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  203. Song F, Tang J, Geng R, Hu H, Zhu C, Cui W, Fan W. Comparison of the efficacy of bone marrow mononuclear cells and bone mesenchymal stem cells in the treatment of osteoarthritis in a sheep model. Int J Clin Exp Pathol. 2014;7:1415.

    PubMed  PubMed Central  Google Scholar 

  204. Koh Y-G, Jo S-B, Kwon O-R, Suh D-S, Lee S-W, Park S-H, Choi Y-J. Mesenchymal stem cell injections improve symptoms of knee osteoarthritis. Arthrosc J Arthrosc Relat Surg. 2013;29:748–55.

    Article  Google Scholar 

  205. Apte SS. Anti-ADAMTS5 monoclonal antibodies: implications for aggrecanase inhibition in osteoarthritis. Biochem J. 2016;473:e1–4.

    Article  CAS  PubMed  Google Scholar 

  206. Xu L, Wu Y, Xiong Z, Zhou Y, Ye Z, Tan WS. Mesenchymal stem cells reshape and provoke proliferation of articular chondrocytes by paracrine secretion. Sci Rep. 2016;6:32705.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  207. Sandell LJ, Aigner T. Articular cartilage and changes in arthritis. An introduction: cell biology of osteoarthritis. Arthritis Res. 2001;3:107–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  208. 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  PubMed  Google Scholar 

  209. Wang Y, Yu D, Liu Z, Zhou F, Dai J, Wu B, Zhou J, Heng BC, Zou XH, Ouyang H, Liu H. Exosomes from embryonic mesenchymal stem cells alleviate osteoarthritis through balancing synthesis and degradation of cartilage extracellular matrix. Stem Cell Res Ther. 2017;8:189.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  210. Song Y, Du H, Dai C, Zhang L, Li S, Hunter DJ, Lu L, Bao C. Human adipose-derived mesenchymal stem cells for osteoarthritis: a pilot study with long-term follow-up and repeated injections. Regen Med. 2018;13:295–307.

    Article  CAS  PubMed  Google Scholar 

  211. Lee WS, Kim HJ, Kim KI, Kim GB, Jin W. Intra-articular injection of autologous adipose tissue-derived mesenchymal stem cells for the treatment of knee osteoarthritis: a phase IIb, randomized, placebo-controlled clinical trial. Stem Cells Transl Med. 2019;8:504–11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  212. Jo CH, Lee YG, Shin WH, Kim H, Chai JW, Jeong EC, Kim JE, Shim H, Shin JS, Shin IS. Intra-articular injection of mesenchymal stem cells for the treatment of osteoarthritis of the knee: a proof-of-concept clinical trial. Stem Cells. 2014;32:1254–66.

    Article  CAS  PubMed  Google Scholar 

  213. Tang CH. Research of pathogenesis and novel therapeutics in arthritis. Int J Mol Sci. 2019;20

  214. Tang C-H. Research of Pathogenesis and Novel Therapeutics in Arthritis 2.0. In: Multidisciplinary Digital Publishing Institute; 2020.

    Google Scholar 

  215. Ross CL, Ang DC, Almeida-Porada G. Targeting mesenchymal stromal cells/pericytes (MSCs) with pulsed electromagnetic field (PEMF) has the potential to treat rheumatoid arthritis. Front Immunol. 2019;10:266.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  216. Miranda JP, Camões SP, Gaspar MM, Rodrigues JS, Carvalheiro M, Bárcia RN, Cruz P, Cruz H, Simões S, Santos JM. The secretome derived from 3D-cultured umbilical cord tissue MSCs counteracts manifestations typifying rheumatoid arthritis. Front Immunol. 2019;10:18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  217. Chowdhury K, Kumar U, Das S, Chaudhuri J, Kumar P, Kanjilal M, Ghosh P, Sircar G, Basyal RK, Kanga U, Bandyopadhaya S, Mitra DK. Synovial IL-9 facilitates neutrophil survival, function and differentiation of Th17 cells in rheumatoid arthritis. Arthritis Res Ther. 2018;20:18.

    Article  PubMed  PubMed Central  Google Scholar 

  218. Zheng J, Zhu L, Iok In I, Chen Y, Jia N, Zhu W. Bone marrow-derived mesenchymal stem cells-secreted exosomal microRNA-192-5p delays inflammatory response in rheumatoid arthritis. Int Immunopharmacol. 2020;78:105985.

    Article  CAS  PubMed  Google Scholar 

  219. Dey P, Panga V, Raghunathan S. A cytokine signalling network for the regulation of inducible nitric oxide synthase expression in rheumatoid arthritis. PLoS One. 2016;11:e0161306.

    Article  PubMed  PubMed Central  Google Scholar 

  220. Tavasolian F, Hosseini AZ, Soudi S, Naderi M. miRNA-146a improves immunomodulatory effects of MSC-derived exosomes in rheumatoid arthritis. Curr Gene Ther. 2020;20:297–312.

    Article  CAS  PubMed  Google Scholar 

  221. Park EH, Lim HS, Lee S, Roh K, Seo KW, Kang KS, Shin K. Intravenous infusion of umbilical cord blood-derived mesenchymal stem cells in rheumatoid arthritis: a phase Ia clinical trial. Stem Cells Transl Med. 2018;7:636–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  222. Álvaro-Gracia JM, Jover JA, García-Vicuña R, Carreño L, Alonso A, Marsal S, Blanco F, Martínez-Taboada VM, Taylor P, Martín-Martín C, DelaRosa O, Tagarro I, Díaz-González F. Intravenous administration of expanded allogeneic adipose-derived mesenchymal stem cells in refractory rheumatoid arthritis (Cx611): results of a multicentre, dose escalation, randomised, single-blind, placebo-controlled phase Ib/IIa clinical trial. Ann Rheum Dis. 2017;76:196–202.

    Article  PubMed  Google Scholar 

  223. Shadmanfar S, Labibzadeh N, Emadedin M, Jaroughi N, Azimian V, Mardpour S, Kakroodi FA, Bolurieh T, Hosseini SE, Chehrazi M, Niknejadi M, Baharvand H, Gharibdoost F, Aghdami N. Intra-articular knee implantation of autologous bone marrow-derived mesenchymal stromal cells in rheumatoid arthritis patients with knee involvement: results of a randomized, triple-blind, placebo-controlled phase 1/2 clinical trial. Cytotherapy. 2018;20:499–506.

    Article  PubMed  Google Scholar 

  224. Rubio D, Garcia-Castro J, Martín MC, de la Fuente R, Cigudosa JC, Lloyd AC, Bernad A. Spontaneous human adult stem cell transformation. Cancer Res. 2005;65:3035–9.

    Article  CAS  PubMed  Google Scholar 

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

Download references

Acknowledgements

Not applicable.

Funding

No funders.

Author information

Authors and Affiliations

Authors

Contributions

All authors contributed to the conception and the main idea of the work. M. J, S.SH, A. M, L. T, S. A, A.O.Z, A. H, and F. M drafted the main text, figures, and tables. A. H supervised the work and provided the comments and additional scientific information. M.S.C, Y. P, and M. J also reviewed and revised the text. All authors read and approved the final version of the work to be published.

Corresponding author

Correspondence to Ali Hassanzadeh.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

There is no conflict of interests.

Additional information

Publisher’s Note

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

This article has been retracted. Please see the retraction notice for more detail: https://doi.org/10.1186/s13287-024-03733-0

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.

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Markov, A., Thangavelu, L., Aravindhan, S. et al. RETRACTED ARTICLE: Mesenchymal stem/stromal cells as a valuable source for the treatment of immune-mediated disorders. Stem Cell Res Ther 12, 192 (2021). https://doi.org/10.1186/s13287-021-02265-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13287-021-02265-1

Keywords