Back to Journals » Journal of Inflammation Research » Volume 17

Emerging Role and Mechanism of Mesenchymal Stem Cells-Derived Extracellular Vesicles in Rheumatic Disease

Authors Wang Z , Yang C, Yan S, Sun J, Zhang J, Qu Z , Sun W , Zang J, Xu D

Received 23 July 2024

Accepted for publication 20 September 2024

Published 30 September 2024 Volume 2024:17 Pages 6827—6846

DOI https://doi.org/10.2147/JIR.S488201

Checked for plagiarism Yes

Review by Single anonymous peer review

Peer reviewer comments 2

Editor who approved publication: Professor Ning Quan



Zhangxue Wang,1,2,* Chunjuan Yang,3,* Shushan Yan,4,* Jiamei Sun,3,* Jin Zhang,1,2 Zhuojian Qu,5 Wenchang Sun,3 Jie Zang,3 Donghua Xu1– 3

1School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, 261000, People’s Republic of China; 2Department of Rheumatology and Immunology, Weifang People’s Hospital, Shandong Second Medical University, Weifang, Shandong, 261000, People’s Republic of China; 3Central Laboratory, Weifang People’s Hospital, Shandong Second Medical University, Weifang, Shandong, 261000, People’s Republic of China; 4Department of Gastrointestinal and Anal Diseases Surgery, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, 261000, People’s Republic of China; 5School of Basic Medicine Sciences, Shandong Second Medical University, Weifang, Shandong, 261000, People’s Republic of China

*These authors contributed equally to this work

Correspondence: Donghua Xu; Jie Zang, Email [email protected]; [email protected]

Abstract: Mesenchymal stem cells (MSCs) are pluripotent stem cells derived from mesoderm. Through cell-to-cell contact or paracrine effects, they carry out biological tasks like immunomodulatory, anti-inflammatory, regeneration, and repair. Extracellular vesicles (EVs) are the primary mechanism for the paracrine regulation of MSCs. They deliver proteins, nucleic acids, lipids, and other active compounds to various tissues and organs, thus facilitating intercellular communication. Rheumatic diseases may be treated using MSCs and MSC-derived EVs (MSC-EVs) due to their immunomodulatory capabilities, according to mounting data. Since MSC-EVs have low immunogenicity, high stability, and similar biological effects as to MSCs themselves, they are advantageous over cell therapy for potential therapeutic applications in rheumatoid arthritis, systemic erythematosus lupus, systemic sclerosis, Sjogren’s syndrome, and other rheumatoid diseases. This review integrates recent advances in the characteristics, functions, and potential molecular mechanisms of MSC-EVs in rheumatic diseases and provides a new understanding of the pathogenesis of rheumatic diseases and MSC-EV-based treatment strategies.

Keywords: mesenchymal stem cells, extracellular vesicles, exosome, immunomodulation, rheumatic disease

Introduction

Rheumatic diseases are refractory conditions that affect multiple systems and organs and are primarily brought on by autoimmune responses.1 Abnormal activation of immune cells due to acute or chronic infection results in abundant immune reaction, large amounts of autoantibodies, deposition of immune complexes, and inflammatory response, which thus causes damages to specific tissues and organs, leading to arthralgia, skin lesions, myalgia, dry mouth, dry eyes, hair loss, and other clinical symptoms. The most prevalent conditions are rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), systemic sclerosis (SSc), Sjogren’s syndrome (SS), and osteoarthritis (OA). Currently, hormones, antirheumatic drugs, and biological agents are the principal medications to treat rheumatic disorders.2 However, not all patients respond well to those drug therapies, and some patients still can not achieve disease remission after standardized treatments. Furthermore, long-term use of antirheumatic drugs often leads to serious side effects in several patients with rheumatic disorders. Therefore, one problem that needs to be resolved is identifying novel therapeutic targets for rheumatic disease.

Mesenchymal Stem Cells (MSCs)

Stem cells have proven beneficial in recent times for autoimmune illnesses like SLE, RA, and SSc.3–5 MSCs widely exist in the bone marrow, umbilical cords, peripheral blood, fatty tissue, and other tissues. They are adult stem cells with multidirectional differentiation potentials. MSCs serve as a promising stem cell-based therapeutic choice and possess properties of regeneration, repair, anti-inflammation, and immunomodulation. However, MSC treatment, as a live cell therapy, inevitably has safety concerns of tumorigenicity and transplant rejection. Accumulated evidence has revealed that MSC-derived extracellular vesicles (MSC-EVs) also perform strong biological functions similar to MSCs by transferring active molecules to the corresponding organs and tissues, such as proteins, nucleic acids, and lipids. More importantly, MSC-EVs exert biological effects with excellent biocompatibility and stability in rheumatic disorders, such as exosomes, which are the most common particles with diameters less than 200 nm in size. In our previously published review, the role of MSCs and MSC-EVs in SLE has been summarized.6 In the current paper, an updated review of MSCs and MSC-EVs in regulating innate and adaptive immunity rheumatoid arthritis has been performed, primarily including SLE, SS, RA, and SSc. This review will be useful to explore new therapeutic strategies for rheumatic diseases based on MSC-EVs.

Biological Characterization of MSC and MSC-EVs

MSCs can differentiate into various cells, including osteoblasts,7 fat cells,8 and chondrocytes.9 MSCs display low immunogenicity, multidirectional differentiation, self-renewal, and immunomodulatory characteristics. Thus, MSCs are star cells for regeneration medicine suggested by several studies on preclinical research and Phase I/II clinical trials.10–12 MSCs express surface markers such as CD105, CD73 and CD90, while lacking expression of CD45, CD34, CD11b, CD19, and HLA-DR.13 Multiple studies have confirmed that MSCs can influence monocytes/macrophages, T cells, and B cells by producing bioactive factors in autoimmune and inflammatory diseases.14–16

In recent years, many studies have reported that many bioactive factors are encapsulated and delivered by MSC-EVs, which subsequently participate in the intercellular communications between MSCs and the recipient cells, including Immune and histological cells (Figure 1). EVs are heterogeneous particles with lipid bilayers that can be secreted by all cell types and mediate intercellular communication. EVs can transport lipids, proteins, and nucleic acids to target cells.17 EVs are categorized into exosomes, microvesicles, and apoptotic vesicles by size. These vesicles play a crucial role in intracellular signaling transduction in MSCs. Apoptotic vesicles are the largest vesicles with a diameter of up to 5000 nm, forming during the late stage of apoptosis through direct budding of the membrane.18 Microvesicles, with the diameter ranging from 100 to 1000 nm, are generated by the budding and shedding of the cell membrane following fusion with the cell membrane.19 Exosomes are known as the smallest particles, ranging from 30 and 150 nm in size, which are formed when the cell membrane is endocytosed (Figure 1).20 Exosomes are one of the most prevalent EVs among them. They have a diameter of 30–150nm, specifically expressing phenotypic markers like CD9, CD63, and CD81. Exosomes confer biological effects by delivering functional molecules to specific sites. Via EVs, T cells, B cells, and MSCs can carry out biological functions by secreting paracrine substances (Figure 2). MSC-EVs are secreted from inside the cell to the outside and like MSCs, have functions of immune regulation, tissue regeneration, and wound healing. EVs derived from MSCs contain a substantial quantity of miRNAs, which play significant roles in various physiological and pathological processes (Figure 2).

Figure 1 The formation of EVs derived from MSCs. Inside MSCs, the membrane invaginations form early endosomes, which further mature into multivesicular bodies (MVB). MVBs fuse with the cell membrane and release their internal intracavicular vesicles outside the cell to form exosomes. As an important medium of intercellular communication, exosomes carry and deliver a variety of bioactive molecules, such as proteins, DNA, mRNA, etc., to regulate the function of recipient cells by direct contaction or transporting these bioactive molecules.

Figure 2 Role of MSC-derived EVs in regulating autoimmunity and inflammation. MSC-EVs suppress activation of B and NK cells, and reverse M1 phenotype into M2 phenotype. Further, MSC-EVs suppress DCs, leading to increased anti-inflammatory (TGF-β and IL-10) and decreased pro-inflammatory (IL-6) factors. MSC-EVs up-regulate Treg and Th2 but down-regulate Th1 and Th17 In addition to immunological cells, MSC-EV improves rheumatic disorders by acting on salivary gland cells, fibroblasts, and synovial cells and exerting anti-inflammatory effects.

MSC-EVs exhibit biological activities similar to those of MSCs, with much lower immunogenicity and higher stability. Although most research has demonstrated that MSC has anti-cancer effects, Gloria Bonuccelli et al have found that MSC makes osteosarcoma cells more aggressive.21 Furthermore, two patients receiving MSC therapy for renal illness experienced thrombosis.22 Pulmonary embolism after treatment with MSC in both in vivo and clinical trials.23,24 For patients, these MSC side effects might be quite harmful. In terms of safety, MSC-EV is not affected by these and can be used as a good treatment.25 Patients with grade III–IV CKD treated with cell-free cord-blood mesenchymal stem cells derived extracellular vesicles (CF-CB-MSCs-EVs) have shown improvements in their overall renal function, and there are no known concerns related to MSC therapy, such as tumorigenicity or pulmonary embolism (Table 1).26 In a Phase 2a clinical trial study (NCT04276987) conducted in Wuhan, China, seven patients with severe COVID-19 pneumonia who received nebulized inhalation of human adipose-derived MSCs-Exosomes (haMSC-Exos) showed varying degrees of regression of lung lesions and significant relief of symptoms (Table 1).27 In a different clinical study, a patient treated with MSC-Exo for Graft-versus-Host disease demonstrated a reduction in inflammatory factors in the peripheral blood and an improvement in clinical symptoms of GvHD (Table 1).28 Although the patient died of pneumonia after 7 months of exosomes, the improvement in these symptoms still provides evidence for effective treatment of GvHD.28 Thus, MSC-EVs are promising cell-free nanoparticles for future biotherapeutics.

Table 1 Application of MSC-Derived Extracellular Vesicles in the Clinic

Regulation of Innate Immunity by MSC-EVs

Regulation of Macrophages by MSC-EVs

Macrophages are critical cells involved in innate immunity, which are responsible for removing necrotic debris and pathogens from the damaged tissues. Macrophages are highly plastic and can be divided into M1 proinflammatory cells and M2 anti-inflammatory cells. MSC-EVs can help convert M1 to M2, according to numerous research (Table 2 and Figure 2). After receiving MSC-Exos, mice’s colitis is lessened because their M2-like substance is expressed, while their M1-like substance is less.30 Exosomes derived from human umbilical cord MSCs (hUMSC-Exos) have been suggested to alleviate steroid-resistant asthma by inhibiting M1 but promoting M2 polarization via suppressing tumor necrosis factor receptor-related factor 1.31 Also, MSC-EV reduced salpingitis by shaping macrophage from M1 to M2.32 Furthermore, some bioactive molecules encapsulated in MSC-EVs can also regulate macrophage functions and phenotypes, such as cytokines, miRNAs, and peptides. MiR-216a-5p derived from hypoxia-preconditioned MSC-Exo has been found to repair spinal cord injury by regulating microglia M1/M2 polarization.33 By preventing M1, MSC-EVs transfer of non-coding RNA tSNA-21109 has also been shown to dramatically reduce SLE.34 Moreover, adipose tissue-derived MSC-EV (AD-MSC-EV) is demonstrated to improve healing, reduce matrix degradation, and switch synovial macrophages towards M2-type in knee OA via encapsulating microRNA.35

Table 2 Application of MSC-Derived EVs in Animal Research

Li’s team discovers that subconjunctival injection of MSC-EV induces the transformation of macrophages into M2, which in turn alleviates autoimmune dacryoadenitis.36 It is also very interesting to see how MSC-EVs control SLE macrophages, particularly in lupus nephritis. Our previous work has confirmed that hUC-MSC-Exo improved lupus nephritis by reshaping macrophage polarization to M2 in MRL/lpr mice.37 Tang et al have reported that MSC-EV reduced the expression of CD14 and the pro-inflammatory cytokine IL-1, suppressed M1, and improved OA.38 It is evident that MSC-EV inhibits the inflammatory response to illness.

According to more research, MSC-EV controls macrophages’ capacity for repair in trauma-related illnesses. MSC-EVs carry miR-139-3p, which inhibits the Stat1 pathway and improves myocardial infarction by encouraging M2 macrophage.39 Besides, BMSC-Exo-derived indoleamine 2,3-dioxygenase (IDO) participated in the tryptophan metabolism and promoted renal repair in acute renal injury by upregulating M2.40 HUMSC-MVs improved renal fibrosis in ischemia partial nephrectomy rats by promoting M2 polarization via hepatocyte growth factor (HGF).41 Additionally, MSC-Exos from the infrapatellar fat pad are demonstrated to promote tendon-bone repair by regulating M1/M2 polarization.42 As a noninvasive strategy, inhalation of MSC-EVs has been documented to ameliorate acute lung injury by exerting anti-inflammatory and immunomodulatory effects.43 The study by Li Qet al has implicated that platelet membrane-engineered EVs functioned to target immunomodulation of cardiac repair via delivering bioactive miRNAs into the cytoplasm and switching M1 polarization towards M2, suggesting the engineered EVs-based membrane fusion way to macrophages.44

Preconditioning MSCs with specific stimulation or gene editing technology may guide the future applications of MSC-EVs. Preconditioning MSCs with hypoxia and induction high expression of HIF-1α resulted in MSC-EVs with highly immunosuppressive and anti-inflammatory effects in experimental Crohn’s disease.45 Similarly, deferoxamine is documented to enhance the functions of MSC-EVs in effectively reprograming macrophage into M2 by activating the HIF-1α signaling pathway.111 EVs from TNF-α-preconditioned MSCs can promote bone repair by decreasing M1 and boosting M2.46 Engineered cytokine-primed MSC-EVs loaded hexyl 5-aminolevulinate hydrochloride (HAL) can regulate the PD-L1/PD-1 signaling pathway to suppress the activation of CD4+T cells and induce the transition of macrophages from M1 to M2, thereby alleviating T1D.47 Besides, the antibacterial and self-healing hydrogels loaded with BMSC-Exos are found to promote the healing of diabetic wounds by stimulating angiogenesis and transforming M2.48 Moreover, melatonin-stimulated MSC-derived Exos enhance diabetic wound healing by inducing M2 polarization through the PTEN/AKT pathway.49 Most interestingly, MSC-Exo is documented to improve peripheral neuropathy in a diabetic mice model by inducing M2 polarization, indicating that MSC-EVs also have significant advantages in regulating M1/M2 bias in treating diabetes-associated nerve injury.50 However, MSC-EVs can accelerate tumor development by promoting M2 polarization in a hypoxic environment.51 Accordingly, MSCs may be a double-edged sword. Taken together, MSC-EVs have significant impacts on regulating macrophage differentiation, polarization, activation, and functions in various inflammatory diseases.

In a brain injury mice model, MSC-EVs are documented to reduce the neuroinflammatory response, promote neural cell proliferation, and enhance oligodendrocyte maturation as well as the expressions of M2 markers of YM-1 and TGF-β.52 Besides, MSC-Exos treatment is found to promote and inhibit neuroinflammation in the brain and spinal cord by inducing M2 polarization via the TLR2/IRAK1/NF-κβ signaling pathway.53 Small EVs derived from four-dimensional-cultured MSCs (MSC-sEV) induce the transformation of M1 to M2 and inhibit inflammation in spinal cord injury rats by activating the IGFBP2/EGFR signal.54 The cysteinyl leukotrienes (CysLTs) are a family of potent inflammatory mediators. MSC-Exos improve acute brain injury and inflammation by inhibiting the M1 polarization of the microglial cells via inactivating the CysLT2R-ERK1/2 signaling pathway.55 MSC-Exos overexpressing microRNA-223-3p is reported to alleviate cerebral ischemia injury by inhibiting neuroinflammation and the M1 polarization of microglial cells.56 As mentioned above, MSC-EVs exert significant effects on neuropathy, and spinal cord injury by regulating the M1/M2 balance of macrophage. Overall, by regulating macrophages, EVs, or exosomes from MSCs regulate inflammation, damage, and immunological disorders (Table 2 and Figure 2).

Regulation of Dendritic Cells (DCs) by MSC-EVs

DCs are also known as antigen-presenting cells.112 DCs in many illnesses are regulated by MSC-EVs (Table 2 and Figure 2). TGF-β and IL-6 are critical in inflammatory and immune diseases.113,114 According to a study by Shahir, M.’s group, AD-MSC-Exo promotes the generation of tolerogenic DC, IL-10, and TGF-β generation but reduces IL-6 in mice.57 Another study has also documented that mature DC treated with MSC-EV resulted in a large rise in TGF-β and a decrease in IL-6, which suggests that MSC-EV is critical in limiting DC maturation.115 Furthermore, MSC-EVs are demonstrated to alleviate allergic rhinitis by enhancing the generation of IL-10 and Treg cell but reducing Th2 response.116 In addition, BMSC-derived microvesicles (BMSC-MVs) can enhance the longevity of transplanted kidneys by increasing the expression of micro-146a in DCs.58 As is well known, the complexity of the dendritic structure of DCs reflects their maturation ability. It has been implicated that the administration of MSC-EV led to significantly decreased intricacy of DC dendrites in mice corneas, slower DC maturation, and reduced dry eye symptoms in mice.59 Consequently, MSCs and MSC-EVs have great potential for modulating DC maturation and DC-mediated immunological effects.

Regulation of Natural Killer (NK) Cells by MSC-EVs

Bone marrow lymphocytes give rise to natural killer (NK) cells, which are engaged in immunological and inflammatory responses.117 MSC-EVs-derived miRNA-155 and miRNA-146 have been shown to primarily block the G0 and G1 phases of the NK cell cycle.118 Besides, MSCs-Exos derived from the fetal liver have been well documented to prevent NK cell proliferation through the TGF-β/Smad signaling pathway.119 There was an increase in NK cells in the wounded kidneys and splenic organs following renal ischemia-reperfusion, according to another study.60 However, reduced severity of renal ischemia-reperfusion injury was found due to a significantly decreased proportion of NK cells and reduced expressions of TLR-2 and CX3CL1 in kidneys after the intravenous infusion of MSC-EV.60 It has been found that the biological effects of bone marrow-derived MSCs (BMSC) were superior to adipose-derived MSCs (ADMSC) in rat kidney transplantation.120 However, the study has reported no improvement in renal function from the EVs of either source, whereas the ADMSC-EV promoted T cells and NK cells infiltration into the kidneys, accelerating the progression of end-stage renal disease.120 Hence, MSC-EVs may control the biological activity of NK cells, thereby compromising their immunomodulatory role. Nonetheless, the source of MSC-EVs should be seriously considered before application. Table 2 and Figure 2 illustrate how MSC-EV controls NK cells.

Modulation of Adaptive Immune Cells by MSC-EVs

Regulation of T Cells by MSC-EVs

T cells are involved in adaptive immunity.121 According to many studies, MSC-EV can regulate T cells. MSC-EVs are reported to inhibit CD4+T cell expansion and Th1 response.122 There are also studies that show MSC-EVs dramatically lower T cell activation and improve Treg cell activity.123 In several disorders, MSC-EV can regulate T-cells. For example, F. Jenner et al found that Small extracellular vesicles (sEVs) derived from hUC-MSC markedly reduced T-cell proliferation in the context of rotator cuff repair.61 MSC-Exo-delivering miR-223 could hinder donor T cell migration in mice of aGVH.62 Regulatory cells (Tregs) are necessary to keep the peripheral immunological environment steady. Foxp3 is a widely recognized transcription factor responsible for Treg differentiation. Yeganeh, A. and his team have revealed that MSC-EVs increased CD4+CD25+FOXP3+ T cells.124 Pro-inflammatory cell subpopulation Th17 causes harm to tissues. RORγt is a pivotal transcription factor for pro-inflammatory Th17 cells. MSC-EVs can inhibit Th17 cells by destabilizing RORγt.63 As a result, MSC-EVs are essential for controlling T cell differentiation, proliferation, and function.

Multiple research results have revealed that MSC-EVs can be used to treat other autoimmune diseases. Small EVs from MSCs are found to alleviate autoimmune uveitis by suppressing Th1 and Th17 cells but promoting Treg activation.64 HUC-MSC-EV can reduce T-lymphocytes, while decreasing Th17 cells and increasing Treg cells, thereby ameliorating collagen-induced arthritis (CIA).65 Several studies have also shown that MSC-Exo regulated the Th17/Tregs balance and alleviated colitis.66–68 sEVs from programmed death receptor (PD-L1)-modified MSCs are demonstrated to foster Tregs differentiation and extend allograft survival.125 MSC-Exo has also been shown to decrease Th1- and Th17-associated cytokines while enhancing Tregs in multiple sclerosis.69 Additionally, MSC-EVs attenuate experimental autoimmune encephalomyelitis by stimulating Treg cells.70 As previously mentioned, our research team has determined that MSC-Exo improved SLE via Treg augmentation.37 Other studies have shown that MSC-EVs suppress the Th17 cell response to modify SLE.126 Accordingly, MSC-EVs may improve the condition by balancing Th17/Treg and Th1/Th2 in autoimmune diseases.

MSC-EVs maintain immune tolerance and immune microenvironment homeostasis by regulating T-cell bioactivity. However, not all tissue-derived MSCs and their EVs inherently possess robust anti-inflammatory properties. MSC-EVs extracted from the mesenteric tissue of Crohn’s disease patients had diminished capacity to inhibit T cells, unlike MSCs and their EVs sourced from subcutaneous adipose tissue (SubQ), which effectively suppressed T-cell, IFN-γ, and IL-17a.71 Moreover, miR-503 loaded by MSC-EV is found to facilitate glioma immune escape.72 Therefore, the varying disease conditions, diverse tissue sources, and other factors may influence or reshape the action of MSC-EVs. In summary, MSC-EVs can control T cell activation, proliferation, and polarization (Table 2 and Figure 2).

Regulation of B Cells by MSC-EVs

B cells develop into a vast quantity of plasma cells, which subsequently generate autoantibodies.127 MSC-EVs have a regulatory effect on B cells (Table 2 and Figure 2). There is evidence that MSC-EVs inhibit Tfh’s interaction with reproductive center B cells in vivo.73 Additionally, BMSC-derived Exos can inhibit B cells by regulating the expression of mRNA genes related to B cell maturation and differentiation.128 MSC-EVs exert immunosuppressive effects by suppressing B cell.118 Recent research has revealed that MSC-EV inhibited B cells by regulating the PI3K-AKT signaling pathway.129 In addition, BMSC-EV can prevent B cells apoptosis in chronic lymphocytic leukemia.130 However, the study by Carreras-Planella, Let al has discovered that MSCs could confer an immunomodulatory effect on B cells independent of MSC-EV.131 Future research is required to figure out the precise functions and mechanisms of the various MSC-EV subtypes in controlling B cells.

Regulatory Role and Mechanism of MSC-EVs in Rheumatic Diseases

MSC-EVs and RA

The main feature of RA is the presence of multiple joints.132 UC-MSCs have shown good efficacy and tolerance in patients with RA.133 When RA patients received intravenous UC-MSC injections, their symptoms and other indications improved, according to a domestic clinical trial.133 Liming Wang et al conducted a three-year prospective phase I/II study using UC-MSC cells in combination with DMARDs, and most patients showed significant improvement in joint symptoms with fewer side effects. Two of these patients, including one elderly male and young female, recovered from joint deformities after 3 years of UC-MSC use, suggesting that UC-MSC cells combined with DMARDs may be a safe and effective long-term treatment for RA patients.134 Several studies have pointed to the involvement of MSC-EV in RA (Table 2 and Figure 3). Fibroblast-like synovial cells (FLSs) are involved in joint lesions. MSC-EVs influence RA by interacting with FLSs and promoting joint repair by delivering bioactive molecules to the damaged joints. For instance, in a CIA mouse model, miR-205-5p delivered by BMSC-Exos dramatically reduced arthritis by regulating double minute 2 (MDM2) in FLSs.74 According to Su. et al, lncRNA HAND2-AS1 loaded via MSC-Exos inhibited RA-FLS.135 Besides, miR-34a delivered by BM-MSC-EVs could inhibit RA-FLS and enhance inflammation by targeting cyclin I through the ATM/ATR/p53 signaling pathway.75 Meng, H. Y. et al found that miRNA-124a-overexpressed MSC-derived EVs prevented RA-FLS by affecting the cell cycle G0/G1 conversion.136 Other bioactive molecules such as circFBXW7, microRNA-21, and microRNA-320a loaded by MSC-Exo were also demonstrated to inhibit FLS, suggesting critical roles of non-coding RNAs delivered by MSC-Exo in RA.76–78 High levels of AFT2 expression encouraged RA-FLS invasion.137 Exosomes loaded with miR-451a that are generated from hUC-MSC bind to ATF2 to prevent FLS.79 Joints can be destroyed by activated synovial fibroblasts. Activation of synovial fibrogenesis and other joint deterioration are inhibited by gingival mesenchymal stem cells (GMSC) and their exosomes have been demonstrated by the development of a hybrid human/mouse model of synovitis.80 Taken together, MSC-EVs significantly affect the development and progression of arthritis and joint damage in RA by delivering bioactive mediators involved in RA-FLS proliferation, cell cycle regulation, and inflammation.

Figure 3 Regulatory effects and mechanisms of MSC-EVs in common rheumatic diseases. MSC-EV improves RA by inhibiting inflammation in FLS through some miRNAs. In addition to miRNA delivery, MSC-EV also acts on salivary gland bodies through several pathways to improve dry mouth, dry throat, or other symptoms in SS mice models. In addition, MSC-EV improves skin sclerosis in SSc or alleviate pulmonary fibrosis by delivering miRNAs. Similarly, in SLE, MSC-EV exerts immunomodulatory and anti-inflammatory effects by interacting with immune cells through complicated mechanisms.

Matrix metalloproteinases (MMPs) are crucial enzymes involved in RA that promote synovium invasion and cartilage damage by degrading the extracellular matrix. MSC-Exos-miR-150-5p alleviated RA by reducing the expression of targeted genes of MMP14 and VEGF.81 Interleukin-27 (IL-27) is overexpressed in RA. According to L. Ma et al, MSC-derived exosomes induced by IL-27 exacerbate RA mainly via upregulating MMP3 expression through the miR-206/L3MBTL4 axis.138 This provides us with a new target for the treatment of R, which can be performed by inhibiting IL-27. It has been well documented that inflammation promotes angiogenesis, which thus aggravates RA. Zhang et al revealed that circEDIL3 delivered by MSC-Exos from human synovium alleviates RA by inhibiting VEGF production via the miR-485-3p/PIAS3/STAT3 signaling pathway.82 Many investigations conducted in the last few years have discovered that iron death inducers can exacerbate synovial angiogenesis and cause RA-FLS, while SMSCs-sEV that overexpresses MiR-433-3p increases VEGF expression and lessens the severity of arthritis.83 AMSC-derived EV suppressed inflammation by delivering IL-1ra and inhibiting IL-1 and TNF-α secretions in an RA mouse model.84 Exosomes from iMSCs reduce IL-17, IL-10, and IL-1β while increasing TGF-β1, polarizing Th2 and M2, and improving RA.85 Small extracellular vesicles derived from MSCs decreased inflammatory cytokine IL-6 and increased M2, which reduced joint inflammation in CIA mice. However, the researchers did not find any statistically significant differences in the therapeutic effects of MSC-sEVs at low and high dosages.86 It is evident that managing inflammation is essential to managing RA.

In RA, MSC-EVs can regulate immune disorders. Gingival MSC-derived exosome (GMSC-Exo) regulated the Th17/Treg balance to alleviate RA.87 Additionally, mouse adipose MSC-Exo-derived miR-146a/miR-155 up-regulated Treg cells and relieved joint inflammation.88 According to Huang et al, miR-223 encapsulated in MSC-Exos could alleviate arthritis by suppressing macrophage-induced inflammation via targeting NLRP3.89 T cells are a major factor in RA pathogenesis. MSC-EV has shown a more potent inhibitory effect on RA pathogenesis as compared to MSC. This was attributed to the MSCs’ IFN-β-induced suppression of CD4 T cells.139 To suppress the IRF1/STAT1 axis and reduce inflammation in RA animals, BMSC-EV distributes miR-378a-5p.90 Accordingly, MSC-EVs and the encapsulated bioactive molecules can control RA (Table 2 and Figure 3).

MSC-EVs and SLE

Chronic inflammation, autoimmune diseases, and numerous organ damage are the final results of SLE, an autoimmune illness marked by excessive immune cell proliferation, activation, and abundance of autoantibodies.140 According to recent clinical research, UC-MSCs are safe and effective for lupus patients via significantly increasing GARP-TGFβ complexes and reducing CD27IgD double-negative B cells.141 We should continue to monitor the impact of MSC-secreted vesicles on the therapy of early SLE as, as per F. Guo et al, hUC-MSC transplantation reduces B-cell proliferation in the early peripheral blood of SLE mice, which is useful for the treatment of early SLE.142 Umbilical cord blood MSCs (UC-BSC)-derived exosomes exerted immunomodulatory and anti-inflammatory effects in SLE by regulating Th17/Treg balance through the miR-19b/KLF13 axis.143 According to our earlier research, hUC-MSC-Exos can reduce SLE by modifying M2 polarization and increasing Treg cells.37 Another work has also reported that MSC-Exos-derived miR-16 and miR-21 alleviated SLE by regulating macrophage polarization.91 Dou et al have demonstrated that tsRNA-21109 encapsulated by MSC-Exo alleviated SLE by inhibiting M1 macrophage polarization.34 Chen et al have also pointed out that microRNA-146a-5p delivered by hUC-MSC-EVs significantly improved SLE-related diffuse alveolar hemorrhage by promoting anti-inflammatory M2 polarization via targeting the NOTCH1 signal.92 Similarly, hUC-MSCs and hUC-MSC-EVs could exert immunoregulatory effects by upregulating Th17 and increasing TGF-β1 in SLE.126 UCMSC-Exos can suppress miR-155 in B cells and raise SHIP-1 levels, which encourages B cell death and reduces SLE.144 Apoptotic vesicles formed from MSCs, such as EVs, block TCR signaling to prevent T cell activation and IL-2 release. Additionally, apoVs have been shown to not harm mice’s organs, which helps to alleviate SLE.93 Accordingly, MSC-EVs work similarly to MSCs and provide SLE patients with a different kind of biological therapy (Table 2 and Figure 3).

MSC-EVs and SSc

SSc manifests as vascular lesions and fibrosis of the skin or organs.145 MSC-EVs can serve as miRNA carriers and exert immunomodulatory in SSc (Table 2 and Figure 3), such as MSC-EVs-derived miRNA clusters, miR-196b-5p, and miR-29a-3p.94–97 BMSC-EVs-delivering miRNA clusters are documented to suppress SSc by regulating the WNT signal and TGF-β signal.95 TGF-β1 is part of the process of tissue fibrosis. MSC-Exo has been demonstrated to improve SSc by downregulating the TGF-β/Smad signaling pathway.96 Remarkably, EVs produced from AD-MSC outperformed parental cells in terms of enhancing myofibroblasts.146 Additionally, IFNγ-primed MSCs-EVs confer immunoregulatory function and improve lung fibrosis by upregulating iNOS, IL1ra, IL6, and PGE2.98 Regulation of M1/M2 macrophage balance is another effect of MSC-Exos in treating SSc.99 Interstitial lung disease (ILD) is a manifestation of severe lung involvement by SSc. Patients with SSc were found to benefit from placental MSC-EV, according to a recent clinical case report. After undergoing traditional medical treatment, the patient still needed oxygen therapy; however, after utilizing MSC-EV, the patient’s symptoms, including dyspnea, dramatically improved, necessitating no longer oxygen therapy. Additionally, a lung CT scan revealed a considerable reduction in fibrosis (Table 1).29 In systemic sclerosis (SSc), interleukin (IL)-33 functions as a pro-inflammatory cytokine and stimulates fibrosis. By delivering miR-214 and inhibiting the IL-33/ST2 axis, BMSC-Exos reduce cutaneous fibrosis.100 Altogether, MSC-EVs as a whole exhibit a therapeutic impact on SSc.

MSC-EVs and SS

SS is characteristic of loss of exocrine glands structure and function, which results in symptoms such as dry mouth, dry eyes, and swallowing difficulty.147 The progression of SS is associated with Th17/Treg.148 In pSS, UCMSC-Exos primarily inhibit CD4 T cell growth by blocking the G0/G1 phase and limiting the cells’ ability to enter the S phase, which in turn balances Th17/Treg.149 Salivary gland inflammation was successfully reduced by EV derived from early passaged iMSCs, which also reduced Th17 and boosted M2.101 MSC and MSC-Exos derived from the labial gland (LG) can ameliorate murine SS by suppressing Th17 but up-regulating Treg.102 Moreover, it has also been found that LG-MSC-Exos-derived miR-125b could alleviate SS by inhibiting plasma cell and targeting PRDM1.103 According to reports, EV isolated from human induced pluripotent stem cells (IPSCs) can activate APC, suppress Tfh and Th17 cells, and relieve SS.104 Under pathological microenvironment, IL-6 is essential for enhancing the suppressive ability of myeloid-derived suppressor cells (MDSC). Olfactory MSC-derived exosomes (OE-MSC-Exo) are demonstrated to increase MDSC proliferation and attenuate SS via activating the IL-6/Jak2/Stat3 pathway.105 OE-MSC-Exo-derived PD-L1 is documented to alleviate SS by inhibiting Tfh response.106 Additionally, the contents of MSC-EVs may vary according to the parent MSCs. In an SS mouse model, Lee et al have found that iPSC-MSC-EVs at an earlier stage exhibited higher immunomodulatory potencies than iPSC-MSC-EVs at a later stage.107 Salivary secretion was stimulated by exosecreted by human exfoliated deciduous teeth (SHEDs), which suppressed p-ERK1/2 activation and glandular cell death.108 According to another study, ZO-1 expression regulated by the Akt/GSK-3β/Slug pathway is the primary mechanism by which Exo generated from human deciduous teeth can enhance SS and increase paracellular permeability of glandular epithelial cells.109 A transmembrane estrogen receptor is called GPER. Exosomes produced from endodontic stem cells (DPSCs) stimulate the cAMP/PKA/CREB pathway via GPER to enhance salivary gland epithelial cell activity.110 The regulatory mechanisms of MSC-EVs in SS are summarized in Table 2 and Figure 3, which lays a theoretical foundation of MSC-EVs for future clinical applications in SS.

Conclusions

The impacts of MSC-EVs with various origins on controlling both innate and adaptive immune responses are methodically covered in this review. In particular, we summarize the underlying mechanisms of MSC-EVs in rheumatic diseases, primarily including RA, SLE, and SS. MSC-EVs have exhibited great potentials in inhibiting inflammation and excessive immune responses, thereby defending against organ and tissue damage. Compared to MSCs, MSC-derived EVs are less immunogenic, safe, and simple to utilize and store. Nevertheless, we need to focus on some issues including quality standards of MSC-EVs, intra-batch and inter-batch repeatability, and characterization and standards of MSC-EVs for a clinical-grade reagent. Besides, further research on the pharmacokinetics, long-term safety, targeted homing mechanism, and active and nonactive components of MSC-EVs needs further exploration.

Data Sharing Statement

The data supporting this review are from previously reported studies and datasets, which have been cited. The processed data are available from Donghua Xu upon request.

Author Contributions

All authors made a significant contribution to the work reported, whether that is in the conception, study design, execution, acquisition of data, analysis and interpretation, or in all these areas; took part in drafting, revising or critically reviewing the article; gave final approval of the version to be published; have agreed on the journal to which the article has been submitted; and agree to be accountable for all aspects of the work.

Funding

This work was supported by the National Natural Science Foundation, China (82171790, 82201925, 82402112), Natural Science Foundation, Shandong Province (ZR2022QH203), and Medical and Health Science and Technology Development Plan, Shandong Province, China (202102070121).

Disclosure

All authors declare no conflicts of interest.

References

1. Weisman MH. Guideline development and implementation in rheumatic disease. Rheum Dis Clin North Am. 2022;48(3):xiii–xiv. doi:10.1016/j.rdc.2022.06.013

2. Higgins GC. Complications of treatments for pediatric rheumatic diseases. Pediatr Clin North Am. 2018;65(4):827–854. doi:10.1016/j.pcl.2018.04.008

3. Jiang Z, Shao M, Dai X, Pan Z, Liu D. Identification of diagnostic biomarkers in systemic lupus erythematosus based on bioinformatics analysis and machine learning. Front Genet. 2022;13:865559. doi:10.3389/fgene.2022.865559

4. Rahimi Khorashad M, Ghoryani M, Gowhari Shabgah A, Shariati-Sarabi Z, Tavakkol Afshari J, Mohammadi M. The effects of mesenchymal stem cells on the gene expression of tgf-beta and ifn-gamma in patients with rheumatoid arthritis. Iran J Allergy Asthma Immunol. 2023;22(2):183–189. doi:10.18502/ijaai.v22i2.12679

5. Ganesan N, Chang YD, Hung SC, et al. Mesenchymal stem cells suppressed skin and lung inflammation and fibrosis in topoisomerase I-induced systemic sclerosis associated with lung disease mouse model. Cell Tissue Res. 2023;391(2):323–337. doi:10.1007/s00441-022-03716-8

6. Yang C, Sun J, Tian Y, et al. Immunomodulatory effect of mscs and mscs-derived extracellular vesicles in systemic lupus erythematosus. Front Immunol. 2021;12:714832. doi:10.3389/fimmu.2021.714832

7. Jinteng L, Peitao X, Wenhui Y, et al. BMAL1-TTK-H2Bub1 loop deficiency contributes to impaired BM-MSC-mediated bone formation in senile osteoporosis. Mol Ther Nucleic Acids. 2023;31:568–585. doi:10.1016/j.omtn.2023.02.014

8. Gimble JM, Katz AJ, Bunnell BA. Adipose-derived stem cells for regenerative medicine. Circ Res. 2007;100(9):1249–1260. doi:10.1161/01.RES.0000265074.83288.09

9. Fortier LA, Nixon AJ, Williams J, Cable CS. Isolation and chondrocytic differentiation of equine bone marrow-derived mesenchymal stem cells. Am J Vet Res. 1998;59(9):1182–1187. doi:10.2460/ajvr.1998.59.09.1182

10. Lian XF, Lu DH, Liu HL, et al. Safety evaluation of human umbilical cord-mesenchymal stem cells in type 2 diabetes mellitus treatment: a phase 2 clinical trial. World J Clin Cases. 2023;11(21):5083–5096. doi:10.12998/wjcc.v11.i21.5083

11. Lightner AL, Reese JS, Ream J, et al. A phase IB/IIA study of ex vivo expanded allogeneic bone marrow-derived mesenchymal stem cells for the treatment of rectovaginal fistulizing Crohn’s disease. Surgery. 2024;175(2):242–249. doi:10.1016/j.surg.2023.07.020

12. McGuirk JP, Metheny L 3rd, Pineiro L, et al. Placental expanded mesenchymal-like cells (PLX-R18) for poor graft function after hematopoietic cell transplantation: a Phase I study. Bone Marrow Transplant. 2023;58(11):1189–1196. doi:10.1038/s41409-023-02068-3

13. LR T, Sanchez-Abarca LI, Muntion S, et al. MSC surface markers (CD44, CD73, and CD90) can identify human MSC-derived extracellular vesicles by conventional flow cytometry. Cell Commun Signal. 2016;14(1):2. doi:10.1186/s12964-015-0124-8

14. Zhang X, He J, Zhao K, et al. Mesenchymal stromal cells ameliorate chronic GVHD by boosting thymic regeneration in a CCR9-dependent manner in mice. Blood Adv. 2023;7(18):5359–5373. doi:10.1182/bloodadvances.2022009646

15. Pan C, Hu T, Liu P, et al. BM-MSCs display altered gene expression profiles in B-cell acute lymphoblastic leukemia niches and exert pro-proliferative effects via overexpression of IFI6. J Transl Med. 2023;21(1):593. doi:10.1186/s12967-023-04464-1

16. Li T, Su X, Lu P, et al. Bone marrow mesenchymal stem cell-derived dermcidin-containing migrasomes enhance LC3-associated phagocytosis of pulmonary macrophages and protect against post-stroke pneumonia. Adv Sci. 2023;10(22):e2206432. doi:10.1002/advs.202206432

17. Keshtkar S, Azarpira N, Ghahremani MH. Mesenchymal stem cell-derived extracellular vesicles: novel frontiers in regenerative medicine. Stem Cell Res Ther. 2018;9(1):63. doi:10.1186/s13287-018-0791-7

18. S ELA, Mager I, Breakefield XO, Wood MJ. Extracellular vesicles: biology and emerging therapeutic opportunities. Nat Rev Drug Discov. 2013;12(5):347–357. doi:10.1038/nrd3978

19. Klimczak A. Mesenchymal stem/progenitor cells and their derivates in tissue regeneration. Int J Mol Sci. 2022;23(12):6652. doi:10.3390/ijms23126652

20. Rao D, Huang D, Sang C, Zhong T, Zhang Z, Tang Z. Advances in mesenchymal stem cell-derived exosomes as drug delivery vehicles. Front Bioeng Biotechnol. 2021;9:797359. doi:10.3389/fbioe.2021.797359

21. Bonuccelli G, Avnet S, Grisendi G, et al. Role of mesenchymal stem cells in osteosarcoma and metabolic reprogramming of tumor cells. Oncotarget. 2014;5(17):7575–7588. doi:10.18632/oncotarget.2243

22. Wu Z, Zhang S, Zhou L, et al. Thromboembolism induced by umbilical cord mesenchymal stem cell infusion: a report of two cases and literature review. Transplant Proc. 2017;49(7):1656–1658. doi:10.1016/j.transproceed.2017.03.078

23. Furlani D, Ugurlucan M, Ong L, et al. Is the intravascular administration of mesenchymal stem cells safe? Mesenchymal stem cells and intravital microscopy. Microvasc Res. 2009;77(3):370–376. doi:10.1016/j.mvr.2009.02.001

24. Jung JW, Kwon M, Choi JC, et al. Familial occurrence of pulmonary embolism after intravenous, adipose tissue-derived stem cell therapy. Yonsei Med J. 2013;54(5):1293–1296. doi:10.3349/ymj.2013.54.5.1293

25. Gowen A, Shahjin F, Chand S, Odegaard KE, Yelamanchili SV. Mesenchymal stem cell-derived extracellular vesicles: challenges in clinical applications. Front Cell Dev Biol. 2020;8:149. doi:10.3389/fcell.2020.00149

26. Nassar W, El-Ansary M, Sabry D, et al. Umbilical cord mesenchymal stem cells derived extracellular vesicles can safely ameliorate the progression of chronic kidney diseases. Biomater Res. 2016;20(1):21. doi:10.1186/s40824-016-0068-0

27. Zhu YG, Shi MM, Monsel A, et al. Nebulized exosomes derived from allogenic adipose tissue mesenchymal stromal cells in patients with severe COVID-19: a pilot study. Stem Cell Res Ther. 2022;13(1):220. doi:10.1186/s13287-022-02900-5

28. Kordelas L, Rebmann V, Ludwig AK, et al. MSC-derived exosomes: a novel tool to treat therapy-refractory graft-versus-host disease. Leukemia. 2014;28(4):970–973. doi:10.1038/leu.2014.41

29. Assar S, Mohammadzadeh D, Norooznezhad AH, et al. Improvement in the clinical manifestations of interstitial lung disease following treatment with placental mesenchymal stromal cell extracellular vesicles in a patient with systemic sclerosis: a case report. Respir Med Case Rep. 2023;46:101923. doi:10.1016/j.rmcr.2023.101923

30. Liu H, Liang Z, Wang F, et al. Exosomes from mesenchymal stromal cells reduce murine colonic inflammation via a macrophage-dependent mechanism. JCI Insight. 2019;4(24). doi:10.1172/jci.insight.131273

31. Dong B, Wang C, Zhang J, et al. Exosomes from human umbilical cord mesenchymal stem cells attenuate the inflammation of severe steroid-resistant asthma by reshaping macrophage polarization. Stem Cell Res Ther. 2021;12(1):204. doi:10.1186/s13287-021-02244-6

32. Zhang C, Liao W, Li W, et al. Human umbilical cord mesenchymal stem cells derived extracellular vesicles alleviate salpingitis by promoting M1-to-M2 transformation. Front Physiol. 2023;14:1131701. doi:10.3389/fphys.2023.1131701

33. Liu W, Rong Y, Wang J, et al. Exosome-shuttled miR-216a-5p from hypoxic preconditioned mesenchymal stem cells repair traumatic spinal cord injury by shifting microglial M1/M2 polarization. J Neuroinflammation. 2020;17(1):47. doi:10.1186/s12974-020-1726-7

34. Dou R, Zhang X, Xu X, Wang P, Yan B. Mesenchymal stem cell exosomal tsRNA-21109 alleviate systemic lupus erythematosus by inhibiting macrophage M1 polarization. Mol Immunol. 2021;139:106–114. doi:10.1016/j.molimm.2021.08.015

35. Ragni E, Perucca Orfei C, De Luca P, Colombini A, Vigano M, de Girolamo L. Secreted factors and EV-miRNAs orchestrate the healing capacity of adipose mesenchymal stem cells for the treatment of knee osteoarthritis. Int J Mol Sci. 2020;21(5):1582. doi:10.3390/ijms21051582

36. Li N, Gao Z, Zhao L, et al. MSC-derived small extracellular vesicles attenuate autoimmune dacryoadenitis by promoting M2 macrophage polarization and inducing tregs via miR-100-5p. Front Immunol. 2022;13:888949. doi:10.3389/fimmu.2022.888949

37. Sun W, Yan S, Yang C, et al. Mesenchymal stem cells-derived exosomes ameliorate lupus by inducing m2 macrophage polarization and regulatory T cell expansion in MRL/lpr mice. Immunol Invest. 2022;51(6):1785–1803. doi:10.1080/08820139.2022.2055478

38. Tang S, Chen P, Zhang H, et al. Comparison of curative effect of human umbilical cord-derived mesenchymal stem cells and their small extracellular vesicles in treating osteoarthritis. Int J Nanomed. 2021;16:8185–8202. doi:10.2147/IJN.S336062

39. Ning Y, Huang P, Chen G, et al. Atorvastatin-pretreated mesenchymal stem cell-derived extracellular vesicles promote cardiac repair after myocardial infarction via shifting macrophage polarization by targeting microRNA-139-3p/Stat1 pathway. BMC Med. 2023;21(1):96. doi:10.1186/s12916-023-02778-x

40. Xie X, Yang X, Wu J, et al. Exosome from indoleamine 2,3-dioxygenase-overexpressing bone marrow mesenchymal stem cells accelerates repair process of ischemia/reperfusion-induced acute kidney injury by regulating macrophages polarization. Stem Cell Res Ther. 2022;13(1):367. doi:10.1186/s13287-022-03075-9

41. Du T, Ju G, Zhou J, et al. Microvesicles derived from human umbilical cord mesenchyme promote M2 macrophage polarization and ameliorate renal fibrosis following partial nephrectomy via hepatocyte growth factor. Hum Cell. 2021;34(4):1103–1113. doi:10.1007/s13577-021-00525-z

42. Xu J, Ye Z, Han K, et al. Infrapatellar fat pad mesenchymal stromal cell-derived exosomes accelerate tendon-bone healing and intra-articular graft remodeling after anterior cruciate ligament reconstruction. Am J Sports Med. 2022;50(3):662–673. doi:10.1177/03635465211072227

43. Zhao R, Wang L, Wang T, Xian P, Wang H, Long Q. Inhalation of MSC-EVs is a noninvasive strategy for ameliorating acute lung injury. J Control Release. 2022;345:214–230. doi:10.1016/j.jconrel.2022.03.025

44. Li Q, Huang Z, Wang Q, et al. Targeted immunomodulation therapy for cardiac repair by platelet membrane engineering extracellular vesicles via hitching peripheral monocytes. Biomaterials. 2022;284:121529. doi:10.1016/j.biomaterials.2022.121529

45. Gomez-Ferrer M, Amaro-Prellezo E, Dorronsoro A, et al. HIF-overexpression and pro-inflammatory priming in human mesenchymal stromal cells improves the healing properties of extracellular vesicles in experimental crohn’s disease. Int J Mol Sci. 2021;22(20):11269. doi:10.3390/ijms222011269

46. Kang M, Huang CC, Gajendrareddy P, et al. Extracellular vesicles from TNFalpha preconditioned MSCs: effects on immunomodulation and bone regeneration. Front Immunol. 2022;13:878194. doi:10.3389/fimmu.2022.878194

47. Wang L, Qi C, Cao H, et al. Engineered cytokine-primed extracellular vesicles with high PD-L1 expression ameliorate type 1 diabetes. Small. 2023;19(38):e2301019. doi:10.1002/smll.202301019

48. Geng X, Qi Y, Liu X, Shi Y, Li H, Zhao L. A multifunctional antibacterial and self-healing hydrogel laden with bone marrow mesenchymal stem cell-derived exosomes for accelerating diabetic wound healing. Biomater Adv. 2022;133:112613. doi:10.1016/j.msec.2021.112613

49. Liu W, Yu M, Xie D, et al. Melatonin-stimulated MSC-derived exosomes improve diabetic wound healing through regulating macrophage M1 and M2 polarization by targeting the PTEN/AKT pathway. Stem Cell Res Ther. 2020;11(1):259. doi:10.1186/s13287-020-01756-x

50. Fan B, Li C, Szalad A, et al. Mesenchymal stromal cell-derived exosomes ameliorate peripheral neuropathy in a mouse model of diabetes. Diabetologia. 2020;63(2):431–443. doi:10.1007/s00125-019-05043-0

51. Ren W, Hou J, Yang C, et al. Extracellular vesicles secreted by hypoxia pre-challenged mesenchymal stem cells promote non-small cell lung cancer cell growth and mobility as well as macrophage M2 polarization via miR-21-5p delivery. J Exp Clin Cancer Res. 2019;38(1):62. doi:10.1186/s13046-019-1027-0

52. Kaminski N, Koster C, Mouloud Y, et al. Mesenchymal stromal cell-derived extracellular vesicles reduce neuroinflammation, promote neural cell proliferation and improve oligodendrocyte maturation in neonatal hypoxic-ischemic brain injury. Front Cell Neurosci. 2020;14:601176. doi:10.3389/fncel.2020.601176

53. Zhang J, Buller BA, Zhang ZG, et al. Exosomes derived from bone marrow mesenchymal stromal cells promote remyelination and reduce neuroinflammation in the demyelinating central nervous system. Exp Neurol. 2022;347:113895. doi:10.1016/j.expneurol.2021.113895

54. Wang J, Wei Q, Yang Y, et al. Small extracellular vesicles derived from four dimensional-culture of mesenchymal stem cells induce alternatively activated macrophages by upregulating IGFBP2/EGFR to attenuate inflammation in the spinal cord injury of rats. Front Bioeng Biotechnol. 2023;11:1146981. doi:10.3389/fbioe.2023.1146981

55. Zhao Y, Gan Y, Xu G, Yin G, Liu D. MSCs-derived exosomes attenuate acute brain injury and inhibit microglial inflammation by reversing CysLT2R-ERK1/2 mediated microglia M1 polarization. Neurochem Res. 2020;45(5):1180–1190. doi:10.1007/s11064-020-02998-0

56. Zhao Y, Gan Y, Xu G, Hua K, Liu D. Exosomes from MSCs overexpressing microRNA-223-3p attenuate cerebral ischemia through inhibiting microglial M1 polarization mediated inflammation. Life Sci. 2020;260:118403. doi:10.1016/j.lfs.2020.118403

57. Shahir M, Mahmoud Hashemi S, Asadirad A, et al. Effect of mesenchymal stem cell-derived exosomes on the induction of mouse tolerogenic dendritic cells. J Cell Physiol. 2020;235(10):7043–7055. doi:10.1002/jcp.29601

58. Wu XQ, Yan TZ, Wang ZW, Wu X, Cao GH, Zhang C. BM-MSCs-derived microvesicles promote allogeneic kidney graft survival through enhancing micro-146a expression of dendritic cells. Immunol Lett. 2017;191:55–62. doi:10.1016/j.imlet.2017.09.010

59. Guo R, Liang Q, He Y, et al. Mesenchymal stromal cells-derived extracellular vesicles regulate dendritic cell functions in dry eye disease. Cells. 2022;12(1):33. doi:10.3390/cells12010033

60. Zou X, Gu D, Zhang G, et al. NK cell regulatory property is involved in the protective role of MSC-derived extracellular vesicles in renal ischemic reperfusion injury. Hum Gene Ther. 2016;27(11):926–935. doi:10.1089/hum.2016.057

61. Jenner F, Wagner A, Gerner I, et al. Evaluation of the potential of umbilical cord mesenchymal stromal cell-derived small extracellular vesicles to improve rotator cuff healing: a pilot ovine study. Am J Sports Med. 2023;51(2):331–342. doi:10.1177/03635465221145958

62. Liu W, Zhou N, Liu Y, et al. Mesenchymal stem cell exosome-derived miR-223 alleviates acute graft-versus-host disease via reducing the migration of donor T cells. Stem Cell Res Ther. 2021;12(1):153. doi:10.1186/s13287-021-02159-2

63. Jung S, Lee S, Kim HJ, et al. Mesenchymal stem cell-derived extracellular vesicles subvert Th17 cells by destabilizing RORgammat through posttranslational modification. Exp Mol Med. 2023;55(3):665–679. doi:10.1038/s12276-023-00949-7

64. Li Y, Ren X, Zhang Z, et al. Effect of small extracellular vesicles derived from IL-10-overexpressing mesenchymal stem cells on experimental autoimmune uveitis. Stem Cell Res Ther. 2022;13(1):100. doi:10.1186/s13287-022-02780-9

65. Xu K, Ma D, Zhang G, et al. Human umbilical cord mesenchymal stem cell-derived small extracellular vesicles ameliorate collagen-induced arthritis via immunomodulatory T lymphocytes. Mol Immunol. 2021;135:36–44. doi:10.1016/j.molimm.2021.04.001

66. Heidari N, Abbasi-Kenarsari H, Namaki S, et al. Adipose-derived mesenchymal stem cell-secreted exosome alleviates dextran sulfate sodium-induced acute colitis by Treg cell induction and inflammatory cytokine reduction. J Cell Physiol. 2021;236(8):5906–5920. doi:10.1002/jcp.30275

67. Heidari N, Abbasi-Kenarsari H, Namaki S, et al. Regulation of the Th17/Treg balance by human umbilical cord mesenchymal stem cell-derived exosomes protects against acute experimental colitis. Exp Cell Res. 2022;419(1):113296. doi:10.1016/j.yexcr.2022.113296

68. Yang R, Huang H, Cui S, Zhou Y, Zhang T, Zhou Y. IFN-gamma promoted exosomes from mesenchymal stem cells to attenuate colitis via miR-125a and miR-125b. Cell Death Dis. 2020;11(7):603. doi:10.1038/s41419-020-02788-0

69. Riazifar M, Mohammadi MR, Pone EJ, et al. Stem cell-derived exosomes as nanotherapeutics for autoimmune and neurodegenerative disorders. ACS Nano. 2019;13(6):6670–6688. doi:10.1021/acsnano.9b01004

70. Ahmadvand Koohsari S, Absalan A, Azadi D. Human umbilical cord mesenchymal stem cell-derived extracellular vesicles attenuate experimental autoimmune encephalomyelitis via regulating pro and anti-inflammatory cytokines. Sci Rep. 2021;11(1):11658. doi:10.1038/s41598-021-91291-3

71. Dadgar N, Altemus J, Li Y, Lightner AL. Effect of Crohn’s disease mesenteric mesenchymal stem cells and their extracellular vesicles on T-cell immunosuppressive capacity. J Cell Mol Med. 2022;26(19):4924–4939. doi:10.1111/jcmm.17483

72. Wang XS, Yu XJ, Wei K, et al. Mesenchymal stem cells shuttling miR-503 via extracellular vesicles enhance glioma immune escape. Oncoimmunology. 2022;11(1):1965317. doi:10.1080/2162402X.2021.1965317

73. Guo L, Lai P, Wang Y, et al. Extracellular vesicles derived from mesenchymal stem cells prevent skin fibrosis in the cGVHD mouse model by suppressing the activation of macrophages and B cells immune response. Int Immunopharmacol. 2020;84:106541. doi:10.1016/j.intimp.2020.106541

74. Ma W, Tang F, Xiao L, et al. miR-205-5p in exosomes divided from chondrogenic mesenchymal stem cells alleviated rheumatoid arthritis via regulating MDM2 in fibroblast-like synoviocytes. J Musculoskelet Neuronal Interact. 2022;22(1):132–141.

75. Wu H, Zhou X, Wang X, et al. miR-34a in extracellular vesicles from bone marrow mesenchymal stem cells reduces rheumatoid arthritis inflammation via the cyclin I/ATM/ATR/p53 axis. J Cell Mol Med. 2021;25(4):1896–1910. doi:10.1111/jcmm.15857

76. Chang L, Kan L. Mesenchymal stem cell-originated exosomal circular RNA circFBXW7 attenuates cell proliferation, migration and inflammation of fibroblast-like synoviocytes by targeting miR-216a-3p/HDAC4 in rheumatoid arthritis. J Inflamm Res. 2021;14:6157–6171. doi:10.2147/JIR.S336099

77. Li GQ, Fang YX, Liu Y, et al. MicroRNA-21 from bone marrow mesenchymal stem cell-derived extracellular vesicles targets TET1 to suppress KLF4 and alleviate rheumatoid arthritis. Ther Adv Chronic Dis. 2021;12:20406223211007369. doi:10.1177/20406223211007369

78. Meng Q, Qiu B. Exosomal microrna-320a derived from mesenchymal stem cells regulates rheumatoid arthritis fibroblast-like synoviocyte activation by suppressing CXCL9 expression. Front Physiol. 2020;11:441. doi:10.3389/fphys.2020.00441

79. Mi L, Gao J, Li N, et al. Human umbilical cord mesenchymal stem cell-derived exosomes loaded miR-451a targets ATF2 to improve rheumatoid arthritis. Int Immunopharmacol. 2024;127:111365. doi:10.1016/j.intimp.2023.111365

80. Bruckner S, Capria VM, Zeno B, et al. The therapeutic effects of gingival mesenchymal stem cells and their exosomes in a chimeric model of rheumatoid arthritis. Arthritis Res Ther. 2023;25(1):211. doi:10.1186/s13075-023-03185-6

81. 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. 2018;201(8):2472–2482. doi:10.4049/jimmunol.1800304

82. Zhang J, Zhang Y, Ma Y, Luo L, Chu M, Zhang Z. Therapeutic potential of exosomal circrna derived from synovial mesenchymal cells via targeting circEDIL3/miR-485-3p/PIAS3/STAT3/VEGF functional module in rheumatoid arthritis. Int J Nanomed. 2021;16:7977–7994. doi:10.2147/IJN.S333465

83. Lin Z, Li W, Wang Y, et al. SMSCs-derived sEV overexpressing miR-433-3p inhibits angiogenesis induced by sEV released from synoviocytes under triggering of ferroptosis. Int Immunopharmacol. 2023;116:109875. doi:10.1016/j.intimp.2023.109875

84. Tsujimaru K, Takanashi M, Sudo K, et al. Extracellular microvesicles that originated adipose tissue derived mesenchymal stem cells have the potential ability to improve rheumatoid arthritis on mice. Regen Ther. 2020;15:305–311. doi:10.1016/j.reth.2020.08.004

85. Choi EW, Lim IR, Park JH, Song J, Choi B, Kim S. Exosomes derived from mesenchymal stem cells primed with disease-condition-serum improved therapeutic efficacy in a mouse rheumatoid arthritis model via enhanced TGF-beta1 production. Stem Cell Res Ther. 2023;14(1):283. doi:10.1186/s13287-023-03523-0

86. Zhang B, Lai RC, Sim WK, Lim SK. Therapeutic efficacy of mesenchymal stem/stromal cell small extracellular vesicles in alleviating arthritic progression by restoring macrophage balance. Biomolecules. 2023;13(10):1501. doi:10.3390/biom13101501

87. Tian X, Wei W, Cao Y, et al. Gingival mesenchymal stem cell-derived exosomes are immunosuppressive in preventing collagen-induced arthritis. J Cell Mol Med. 2022;26(3):693–708. doi:10.1111/jcmm.17086

88. 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(4):297–312. doi:10.2174/1566523220666200916120708

89. Huang Y, Lu D, Ma W, et al. miR-223 in exosomes from bone marrow mesenchymal stem cells ameliorates rheumatoid arthritis via downregulation of NLRP3 expression in macrophages. Mol Immunol. 2022;143:68–76. doi:10.1016/j.molimm.2022.01.002

90. Zhang Y, Jiao Z, Wang S. Bone marrow mesenchymal stem cells release miR-378a-5p-carried extracellular vesicles to alleviate rheumatoid arthritis. J Innate Immun. 2023;15(1):893–910. doi:10.1159/000534830

91. Zhang M, Johnson-Stephenson TK, Wang W, et al. Mesenchymal stem cell-derived exosome-educated macrophages alleviate systemic lupus erythematosus by promoting efferocytosis and recruitment of IL-17(+) regulatory T cell. Stem Cell Res Ther. 2022;13(1):484. doi:10.1186/s13287-022-03174-7

92. Chen X, Su C, Wei Q, Sun H, Xie J, Nong G. Exosomes derived from human umbilical cord mesenchymal stem cells alleviate diffuse alveolar hemorrhage associated with systemic lupus erythematosus in mice by promoting m2 macrophage polarization via the microRNA-146a-5p/NOTCH1 axis. Immunol Invest. 2022;51(7):1975–1993. doi:10.1080/08820139.2022.2090261

93. Wang R, Hao M, Kou X, et al. Apoptotic vesicles ameliorate lupus and arthritis via phosphatidylserine-mediated modulation of T cell receptor signaling. Bioact Mater. 2023;25:472–484. doi:10.1016/j.bioactmat.2022.07.026

94. Baral H, Uchiyama A, Yokoyama Y, et al. Antifibrotic effects and mechanisms of mesenchymal stem cell-derived exosomes in a systemic sclerosis mouse model: possible contribution of miR-196b-5p. J Dermatol Sci. 2021;104(1):39–47. doi:10.1016/j.jdermsci.2021.08.006

95. Jin J, Ou Q, Wang Z, et al. BMSC-derived extracellular vesicles intervened the pathogenic changes of scleroderma in mice through miRNAs. Stem Cell Res Ther. 2021;12(1):327. doi:10.1186/s13287-021-02400-y

96. Li M, Zhang HP, Wang XY, Chen ZG, Lin XF, Zhu W. Mesenchymal stem cell-derived exosomes ameliorate dermal fibrosis in a murine model of bleomycin-induced scleroderma. Stem Cells Dev. 2021;30(19):981–990. doi:10.1089/scd.2021.0112

97. Rozier P, Maumus M, Maria ATJ, et al. Mesenchymal stromal cells-derived extracellular vesicles alleviate systemic sclerosis via miR-29a-3p. J Autoimmun. 2021;121:102660. doi:10.1016/j.jaut.2021.102660

98. Rozier P, Maumus M, Maria ATJ, et al. Lung fibrosis is improved by extracellular vesicles from IFNgamma-primed mesenchymal stromal cells in murine systemic sclerosis. Cells. 2021;10(10):2727. doi:10.3390/cells10102727

99. Yu Y, Shen L, Xie X, Zhao J, Jiang M. The therapeutic effects of exosomes derived from human umbilical cord mesenchymal stem cells on scleroderma. Tissue Eng Regen Med. 2022;19(1):141–150. doi:10.1007/s13770-021-00405-5

100. Xie L, Long X, Mo M, et al. Bone marrow mesenchymal stem cell-derived exosomes alleviate skin fibrosis in systemic sclerosis by inhibiting the IL-33/ST2 axis via the delivery of microRNA-214. Mol Immunol. 2023;157:146–157. doi:10.1016/j.molimm.2023.03.017

101. Zhao Q, Bae EH, Zhang Y, et al. Inhibitory effects of extracellular vesicles from iPS-cell-derived mesenchymal stem cells on the onset of sialadenitis in sjogren’s syndrome are mediated by immunomodulatory splenocytes and improved by inhibiting miR-125b. Int J Mol Sci. 2023;24(6). doi:10.3390/ijms24065258

102. Li B, Xing Y, Gan Y, He J, Hua H. Labial gland-derived mesenchymal stem cells and their exosomes ameliorate murine Sjogren’s syndrome by modulating the balance of Treg and Th17 cells. Stem Cell Res Ther. 2021;12(1):478. doi:10.1186/s13287-021-02541-0

103. Xing Y, Li B, He J, Hua H. Labial gland mesenchymal stem cell derived exosomes-mediated mirna-125b attenuates experimental Sjogren’s syndrome by targeting PRDM1 and suppressing plasma cells. Front Immunol. 2022;13:871096. doi:10.3389/fimmu.2022.871096

104. Hai B, Shigemoto-Kuroda T, Zhao Q, Lee RH, Liu F. Inhibitory effects of ipsc-mscs and their extracellular vesicles on the onset of sialadenitis in a mouse model of Sjogren’s syndrome. Stem Cells Int. 2018;2018:2092315. doi:10.1155/2018/2092315

105. Rui K, Hong Y, Zhu Q, et al. Olfactory ecto-mesenchymal stem cell-derived exosomes ameliorate murine Sjogren’s syndrome by modulating the function of myeloid-derived suppressor cells. Cell Mol Immunol. 2021;18(2):440–451. doi:10.1038/s41423-020-00587-3

106. Rui K, Shen Z, Peng N, et al. Olfactory ecto-mesenchymal stem cell-derived exosomes ameliorate murine Sjogren’s syndrome via suppressing tfh cell response. Rheumatol Immunol Res. 2022;3(4):198–207. doi:10.2478/rir-2022-0035

107. Kim H, Zhao Q, Barreda H, et al. Identification of molecules responsible for therapeutic effects of extracellular vesicles produced from iPSC-derived mscs on sjo gren’s syndrome. Aging Dis. 2021;12(6):1409–1422. doi:10.14336/AD.2021.0621

108. Chu WX, Ding C, Du ZH, et al. SHED-exos promote saliva secretion by suppressing p-ERK1/2-mediated apoptosis in glandular cells. Oral Dis. 2023;30(5):3066–3080. doi:10.1111/odi.14776

109. Du Z, Wei P, Jiang N, Wu L, Ding C, Yu G. SHED-derived exosomes ameliorate hyposalivation caused by Sjogren’s syndrome via Akt/GSK-3beta/Slug-mediated ZO-1 expression. Chin Med J. 2023;136(21):2596–2608. doi:10.1097/CM9.0000000000002610

110. Hu S, Chen B, Zhou J, et al. Dental pulp stem cell-derived exosomes revitalize salivary gland epithelial cell function in NOD mice via the GPER-mediated cAMP/PKA/CREB signaling pathway. J Transl Med. 2023;21(1):361. doi:10.1186/s12967-023-04198-0

111. Park SM, An JH, Lee JH, et al. Extracellular vesicles derived from DFO-preconditioned canine AT-MSCs reprogram macrophages into M2 phase. PLoS One. 2021;16(7):e0254657. doi:10.1371/journal.pone.0254657

112. Liu Z, Wang H, Li Z, et al. Dendritic cell type 3 arises from Ly6C(+) monocyte-dendritic cell progenitors. Immunity. 2023;56(8):1761–1777e6. doi:10.1016/j.immuni.2023.07.001

113. Liu L, Wang Y, Yu S, et al. Transforming growth factor beta promotes inflammation and tumorigenesis in Smad4-deficient intestinal epithelium in a YAP-dependent manner. Adv Sci. 2023;10(23):e2300708. doi:10.1002/advs.202300708

114. Ding Q, Fang H, Jin P, et al. Pretreating mesenchymal stem cells with IL-6 regulates the inflammatory response of DSS-induced ulcerative colitis in rats. Transpl Immunol. 2023;76:101765. doi:10.1016/j.trim.2022.101765

115. Reis M, Mavin E, Nicholson L, Green K, Dickinson AM, Wang XN. Mesenchymal stromal cell-derived extracellular vesicles attenuate dendritic cell maturation and function. Front Immunol. 2018;9:2538. doi:10.3389/fimmu.2018.02538

116. Peng YQ, Wu ZC, Xu ZB, et al. Mesenchymal stromal cells-derived small extracellular vesicles modulate DC function to suppress Th2 responses via IL-10 in patients with allergic rhinitis. Eur J Immunol. 2022;52(7):1129–1140. doi:10.1002/eji.202149497

117. Wang L, Mao L, Xiao W, Chen P. Natural killer cells immunosenescence and the impact of lifestyle management. Biochem Biophys Res Commun. 2023;689:149216. doi:10.1016/j.bbrc.2023.149216

118. Di Trapani M, Bassi G, Midolo M, et al. Differential and transferable modulatory effects of mesenchymal stromal cell-derived extracellular vesicles on T, B and NK cell functions. Sci Rep. 2016;6(1):24120. doi:10.1038/srep24120

119. Fan Y, Herr F, Vernochet A, Mennesson B, Oberlin E, Durrbach A. Human fetal liver mesenchymal stem cell-derived exosomes impair natural killer cell function. Stem Cells Dev. 2019;28(1):44–55. doi:10.1089/scd.2018.0015

120. Ramirez-Bajo MJ, Rovira J, Lazo-Rodriguez M, et al. Impact of mesenchymal stromal cells and their extracellular vesicles in a rat model of kidney rejection. Front Cell Dev Biol. 2020;8:10. doi:10.3389/fcell.2020.00010

121. Sun L, Su Y, Jiao A, Wang X, Zhang B. T cells in health and disease. Signal Transduct Target Ther. 2023;8(1):235. doi:10.1038/s41392-023-01471-y

122. Franco da Cunha F, Andrade-Oliveira V, Candido de Almeida D, et al. Extracellular vesicles isolated from mesenchymal stromal cells modulate CD4(+) T lymphocytes toward a regulatory profile. Cells. 2020;9(4):1059. doi:10.3390/cells9041059

123. Bolandi Z, Mokhberian N, Eftekhary M, et al. Adipose derived mesenchymal stem cell exosomes loaded with miR-10a promote the differentiation of Th17 and treg from naive CD4(+) T cell. Life Sci. 2020;259:118218. doi:10.1016/j.lfs.2020.118218

124. Yeganeh A, Fathollahi A, Hashemi SM, Yeganeh F. In vitro treatment of murine splenocytes with extracellular vesicles derived from mesenchymal stem cells altered the mRNA levels of the master regulator genes of T helper cell subsets. Mol Biol Rep. 2023;50(4):3309–3316. doi:10.1007/s11033-023-08247-1

125. Ou Q, Dou X, Tang J, Wu P, Pan D. Small extracellular vesicles derived from PD-L1-modified mesenchymal stem cell promote Tregs differentiation and prolong allograft survival. Cell Tissue Res. 2022;389(3):465–481. doi:10.1007/s00441-022-03650-9

126. Xie M, Li C, She Z, et al. Human umbilical cord mesenchymal stem cells derived extracellular vesicles regulate acquired immune response of lupus mouse in vitro. Sci Rep. 2022;12(1):13101. doi:10.1038/s41598-022-17331-8

127. Evans LS, Lewis KE, DeMonte D, et al. Povetacicept, an enhanced dual april/BAFF antagonist that modulates B lymphocytes and pathogenic autoantibodies for the treatment of lupus and other B cell-related autoimmune diseases. Arthritis Rheumatol. 2023;75(7):1187–1202. doi:10.1002/art.42462

128. Khare D, Or R, Resnick I, Barkatz C, Almogi-Hazan O, Avni B. Mesenchymal stromal cell-derived exosomes affect mRNA expression and function of B-lymphocytes. Front Immunol. 2018;9:3053. doi:10.3389/fimmu.2018.03053

129. Adamo A, Brandi J, Caligola S, et al. Extracellular vesicles mediate mesenchymal stromal cell-dependent regulation of B cell PI3K-AKT signaling pathway and actin cytoskeleton. Front Immunol. 2019;10:446. doi:10.3389/fimmu.2019.00446

130. Crompot E, Van Damme M, Pieters K, et al. Extracellular vesicles of bone marrow stromal cells rescue chronic lymphocytic leukemia B cells from apoptosis, enhance their migration and induce gene expression modifications. Haematologica. 2017;102(9):1594–1604. doi:10.3324/haematol.2016.163337

131. Carreras-Planella L, Monguio-Tortajada M, Borras FE, Franquesa M. Immunomodulatory Effect of MSC on B Cells Is Independent of Secreted Extracellular Vesicles. Front Immunol. 2019;10:1288. doi:10.3389/fimmu.2019.01288

132. Leon L, Madrid-Garcia A, Lopez-Viejo P, et al. Difficult-to-treat rheumatoid arthritis (D2T RA): clinical issues at early stages of disease. RMD Open. 2023;9(1):e002842. doi:10.1136/rmdopen-2022-002842

133. Lv X, Wang L, Zou X, Huang S. Umbilical cord mesenchymal stem cell therapy for regenerative treatment of rheumatoid arthritis: opportunities and challenges. Drug Des Devel Ther. 2021;15:3927–3936. doi:10.2147/DDDT.S323107

134. Wang L, Huang S, Li S, et al. Efficacy and safety of umbilical cord mesenchymal stem cell therapy for rheumatoid arthritis patients: a prospective phase I/II study. Drug Des Devel Ther. 2019;13:4331–4340. doi:10.2147/DDDT.S225613

135. Su Y, Liu Y, Ma C, Guan C, Ma X, Meng S. Mesenchymal stem cell-originated exosomal lncRNA HAND2-AS1 impairs rheumatoid arthritis fibroblast-like synoviocyte activation through miR-143-3p/TNFAIP3/NF-kappaB pathway. J Orthop Surg Res. 2021;16(1):116. doi:10.1186/s13018-021-02248-1

136. Meng HY, Chen LQ, Chen LH. The inhibition by human MSCs-derived miRNA-124a overexpression exosomes in the proliferation and migration of rheumatoid arthritis-related fibroblast-like synoviocyte cell. BMC Musculoskelet Disord. 2020;21(1):150. doi:10.1186/s12891-020-3159-y

137. Zhang X, Zhang D, Wang Q, et al. Sprouty2 inhibits migration and invasion of fibroblast-like synoviocytes in rheumatoid arthritis by down-regulating ATF2 expression and phosphorylation. Inflammation. 2021;44(1):91–103. doi:10.1007/s10753-020-01311-z

138. Ma L, Liu Y, Xu F, et al. IL-27-induced, MSC-derived exosomes promote MMP3 expression through the miR-206/L3MBTL4 axis in synovial fibroblasts. Altern Ther Health Med. 2023;29(8):680–688.

139. Tsiapalis D, Floudas A, Tertel T, et al. Therapeutic effects of mesenchymal/stromal stem cells and their derived extracellular vesicles in rheumatoid arthritis. Stem Cells Transl Med. 2023;12(12):849–862. doi:10.1093/stcltm/szad065

140. Barber MRW, Falasinnu T, Ramsey-Goldman R, Clarke AE. The global epidemiology of SLE: narrowing the knowledge gaps. Rheumatology. 2023;62(Suppl 1):i4–i9. doi:10.1093/rheumatology/keac610

141. Kamen DL, Wallace C, Li Z, et al. Safety, immunological effects and clinical response in a phase I trial of umbilical cord mesenchymal stromal cells in patients with treatment refractory SLE. Lupus Sci Med. 2022;9(1):e000704. doi:10.1136/lupus-2022-000704

142. Guo F, Pan Q, Chen T, et al. hUC-MSC transplantation therapy effects on lupus-prone MRL/lpr mice at early disease stages. Stem Cell Res Ther. 2023;14(1):211. doi:10.1186/s13287-023-03432-2

143. Tu J, Zheng N, Mao C, Liu S, Zhang H, Sun L. UC-BSCs exosomes regulate Th17/Treg balance in patients with systemic lupus erythematosus via miR-19b/KLF13. Cells. 2022;11(24):4123. doi:10.3390/cells11244123

144. Zhao Y, Song W, Yuan Z, et al. Exosome derived from human umbilical cord mesenchymal cell exerts immunomodulatory effects on B cells from SLE patients. J Immunol Res. 2023;2023:3177584. doi:10.1155/2023/3177584

145. Truchetet ME, Brembilla NC, Chizzolini C. Current concepts on the pathogenesis of systemic sclerosis. Clin Rev Allergy Immunol. 2023;64(3):262–283. doi:10.1007/s12016-021-08889-8

146. Rozier P, Maumus M, Bony C, et al. Extracellular vesicles are more potent than adipose mesenchymal stromal cells to exert an anti-fibrotic effect in an in vitro model of systemic sclerosis. Int J Mol Sci. 2021;22(13):6837. doi:10.3390/ijms22136837

147. Thorlacius GE, Bjork A, Wahren-Herlenius M. Genetics and epigenetics of primary Sjogren syndrome: implications for future therapies. Nat Rev Rheumatol. 2023;19(5):288–306. doi:10.1038/s41584-023-00932-6

148. Qi J, Zhou X, Bai Z, et al. FcgammaRIIIA activation-mediated up-regulation of glycolysis alters MDSCs modulation in CD4(+) T cell subsets of Sjogren syndrome. Cell Death Dis. 2023;14(2):86. doi:10.1038/s41419-023-05631-4

149. Ma D, Wu Z, Zhao X, et al. Immunomodulatory effects of umbilical mesenchymal stem cell-derived exosomes on CD4(+) T cells in patients with primary Sjogren’s syndrome. Inflammopharmacology. 2023;31(4):1823–1838. doi:10.1007/s10787-023-01189-x

Creative Commons License © 2024 The Author(s). This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, 3.0) License. By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.