Skip to main content

Adipose-derived mesenchymal stem cell therapy for connective tissue diseases and complications

Abstract

Mesenchymal stem cells (MSCs) may be effective in treating connective tissue disease and associated organ damage, leveraging their anti-inflammatory and immunoregulatory effects. Moreover, MSCs may possess the ability to produce antiapoptotic, proliferative, growth, angiogenic, and antifibrotic factors. Among MSCs, adipose-derived MSCs (ASCs) stand out for their relative ease of harvesting and abundance. Additionally, studies have indicated that compared with bone marrow-derived MSCs, ASCs have superior immunomodulatory, proangiogenic, antiapoptotic, and antioxidative properties. However, relatively few reviews have focused on the efficacy of ASC therapy in treating connective tissue disease (CTD) and interstitial lung disease (ILD). Therefore, this review aims to evaluate evidence from preclinical studies that investigate the effectiveness of MSC therapy, specifically ASC therapy, in managing CTD and ILD. Moreover, we explore the outcomes of documented clinical trials. We also introduce an innovative approach involving the utilization of pharmacologically primed ASCs in the CTD model to address the current challenges associated with ASC therapy.

Background

Recent advances have transformed the treatment landscape of connective tissue disease (CTD), resulting in the availability of various treatment options. For example, the treatment of rheumatoid arthritis (RA) now focuses on achieving remission by combining biological disease-modifying antirheumatic drugs (DMARDs) and targeted synthetic DMARDs with conventional synthetic DMARDs, including methotrexate [1]. For systemic lupus erythematosus (SLE), conventional immunosuppressants, including cyclophosphamide (CYC), azathioprine, and cyclosporine, alongside novel immunosuppressants such as tacrolimus and mycophenolate mofetil (MMF), have demonstrated efficacy in treating lupus nephritis and organ damage associated with SLE [2]. Belimumab—a monoclonal antibody targeting soluble B lymphocyte stimulator—is also effective in suppressing disease activity in SLE and serves as a maintenance therapy for this disease [3]. Advancements in novel immunosuppressants and biological products have improved disease control and CTD prognosis. However, challenges such as the risks of side effects and opportunistic infection owing to immunosuppression persist. Furthermore, effective treatments for many types of CTD, such as systemic sclerosis (SSc), remain few.

CTD is associated with various types of organ damage, with interstitial lung disease (ILD) being particularly important due to its significant impact on patient prognosis. Progressive ILD associated with dermatomyositis/polymyositis, progressive ILD linked to SSc, acute exacerbation of ILD correlated with RA, and comparable conditions present therapeutic challenges and are associated with a poor prognosis [4]. Treatment for these progressive CTD-ILDs typically requires a combination of corticosteroids and immunosuppressive agents, including calcineurin inhibitors, CYC, and MMF, among others [4, 5]. However, numerous cases continue to progress despite the advances, leading to respiratory failure and, ultimately, mortality. Additionally, the prolonged and high-dose utilization of these immunosuppressants raises concerns regarding infections and potential side effects. Recently, pirfenidone and nintedanib have been employed to inhibit fibrotic progression in ILD; however, their efficacy remains limited [5, 6]. Therefore, developing more effective and safer treatments for refractory CTD and complicating ILD, considering their efficacy and safety, is crucial.

Mesenchymal stem cells (MSCs) are being explored extensively in the field of regenerative medicine, given their ability to differentiate into various mesenchymal cells, including osteoblasts, adipocytes, myocytes, and chondrocytes [7, 8]. Moreover, MSCs exhibit antiapoptotic, anti-inflammatory, and antifibrotic effects, along with their ability to modulate the immune response and modify the microenvironment at the engraftment site [7, 9,10,11]. Thus, the current focus involves investigating the efficacy of MSC therapy for inflammatory and autoimmune diseases. Additionally, MSCs can serve as allografts owing to their low expression levels of human leukocyte antigen (HLA) class I and II [12, 13], and they are well-tolerated when administered intravenously [9]. While research on MSCs has primarily focused on those harvested from the bone marrow, recent studies indicate that these cells can also be harvested from various other tissues, including the cord blood, placenta, and adipose tissue. Adipose tissue specifically contains substantial numbers of MSCs, with subcutaneous adipose tissue being easily accessible. Consequently, adipose-derived mesenchymal stem cells (ASCs) are currently gaining much attention.

Therefore, this review aims to examine evidence from preclinical studies that investigated the efficacy of MSC therapy, particularly focusing on ASC therapy, in managing CTD and ILD. Additionally, we explored the findings of documented clinical trials. Furthermore, this review aims to introduce an innovative approach involving the use of pharmacologically primed ASCs in the CTD model. The study findings could offer a potential strategy to prevent current challenges associated with ASC therapy.

Immunomodulatory effects of MSCs

MSCs modulate the activities of various immune cells, including lymphocytes, monocytes, and neutrocytes. Furthermore, MSCs inhibit cyclin D2 expression, leading to cell cycle arrest at the G0/G1 phase [14]. They also inhibit the proliferation of CD4+ and CD8+ T cells, including memory and naïve T cells [15]. The effect on T cells is mediated by specific factors, namely transforming growth factor-β1 (TGF-β1), prostaglandin E2 (PGE2), and indoleamine 2,3 dioxygenase, which are generated by MSCs [16]. Other factors have been observed to mediate the effect of MSCs on T cells. These include IL-6, hepatocyte growth factor (HGF), heme oxygenase 1, HLA-G5, interleukin 1 receptor antagonist, and soluble TNF-receptor 1 [17,18,19,20]. MSCs induce anergy by inhibiting proinflammatory cytokines, including IFN-γ, TNF-α, and IL-17, while simultaneously elevating the expression of IL-10 and IL-4 [16, 21, 22]. In addition, inducible nitric oxide synthase, which triggers differentiation into Th2-type T cells and regulatory T cells (Tregs), exhibits cytotoxic effects on T cells and natural killer (NK) cells. Additionally, MSCs suppress the function of Th17-type T cells through cell contact via the programmed death-1/programmed death ligand-1 pathway [23]. Soluble factors produced by MSCs, including PGE2 and TGF-β1, influence the proliferative capacity and/or cytotoxicity of NK cells through direct and indirect mechanisms [24]. The TNF-α-stimulated gene 6 protein (TSG-6), secreted by MSCs, exerts anti-inflammatory effects in macrophages by attenuating TLR2/NF-κB signaling [25].

MSCs inhibit the differentiation of T cell-dependent B cells into plasma cells by suppressing the activity of CD4+ T cells. Furthermore, MSCs directly inhibit the proliferation, differentiation into plasma cells, and chemotaxis [26], consequently exerting an indirect suppression on T cell activation [27]. In addition, MSCs inhibit the differentiation, maturation, and activity of dendritic cells while inducing differentiation into M2 macrophages [28,29,30]. PGE2 and IL-6—produced by MSCs—induce the secretion of IL-10 by M2 macrophages, consequently inhibiting the neutrophil oxidative burst [31].

The trophic effect of MSCs is predominantly attributed to antiapoptotic, proliferative, growth, and angiogenic factors. Besides their antiapoptotic and angiogenic effects, MSCs prevent fibrosis by promoting the secretion of HFG and matrix metalloproteinases (MMPs) while concurrently downregulating collagen synthesis [32]. Exosomes released by MSCs encompass proteins, messenger RNA (mRNA), and micro RNA, crucial factors that significantly contribute to the trophic effect of MSCs [32].

The anti-inflammatory and immunoregulatory effects of MSCs and their capacity to produce antiapoptotic, proliferative, growth, angiogenic, and antifibrotic factors such as MMPs and HGF render MSC therapy an appealing treatment option for CTD and its associated organ involvement (Fig 1).

Fig. 1
figure 1

Schematic overview of anti-inflammatory, immunoregulatory effects, and other beneficial factors for preventing disease progression of connective tissue diseases and complications. MSCs exert a multifaceted suppressive effect on various immune cells implicated in CTDs. By inhibiting the proliferation and effector functions of CD4+ and CD8+ T cells, including memory and naïve subsets, MSCs wield their influence through specific factors such as TGF-β1, PGE2, HO-1, iNOS, sHLA-G5, and IDO. This induction of T cell anergy, suppression of proinflammatory cytokines, and elevation of anti-inflammatory cytokines collectively contribute to immune regulation. Furthermore, MSCs modulate the function of regulatory T cells, fostering their differentiation alongside Th2-type T cells, thus further dampening immune responses. Not only do MSCs hinder the differentiation, maturation, and function of dendritic cells, but they also steer the polarization of M2 macrophages, inducing an anti-inflammatory milieu. Through the secretion of soluble factors such as IDO1, PGE2, sHLA-G5, HO-1, and TSG-6, MSCs exert influence on the activity of NK cells and macrophages, enhancing their anti-inflammatory properties. PGE2 and IL-6 from MSCs prompt M2 macrophages to release IL-10, which, in turn, suppresses neutrophil oxidative burst. MSCs further impede CD4+ T cell activity, thwarting T cell-driven B cell differentiation into plasma cells while directly curtailing plasma cell functions. Moreover, through the secretion of HGF and matrix MMPs, along with the downregulation of collagen synthesis, MSCs impede fibrosis. Their trophic effects are significantly augmented by the encapsulation of various bioactive molecules within exosomes released by MSCs. In summary, the collective anti-inflammatory, immunoregulatory, and trophic actions of MSCs position them as highly promising therapeutic candidates for CTDs and their associated complications. MSCs mesenchymal stem cells, CTDs connective tissue diseases, MMPs matrix metalloproteinases, NK natural killer, IDO indoleamine 2,3 dioxygenase, PGE2 prostaglandin E2, TSG-6 TNF-α-stimulated gene 6 protein, sHLA-G5 soluble human leukocyte antigen G5, HO-1 heme oxygenase 1, TGF-β1 transforming growth factor-β1, IL-6 interleukin-6, iNOS inducible nitric oxide synthase, HGF hepatocyte growth factor, ILD interstitial lung disease. Solid line indicates promotion, dotted line indicates suppression

Advantages of ASCs compared to other types of MSCs, including bone marrow-derived MSCs and umbilical cord blood-derived MSCs

MSCs exhibit remarkable abilities for self-replication and self-renewal. As adult stem cells are distributed throughout the body, MSCs can be harvested from various sources, including the bone marrow, cord blood, adipose tissue, and placenta. Thus, the utilization of MSCs is associated with fewer ethical concerns compared to that of embryonic stem cells. In addition, compared to induced pluripotent stem cells, MSCs are considered safer to use since there is no requirement for gene transfers.

ASCs are fibroblast-like cells resembling MSCs harvested from other tissues, with differences in cell surface characteristics and gene expression [33]. However, the physiological significance of these differences remains unclear. Bone marrow-derived MSCs (BM-MSCs) constitute only 0.001–0.01% of all nucleated cells [34]. Additionally, studies suggest that the proliferation rate of harvested MSCs tends to decelerate with aging [35]. Therefore, harvesting a sufficient quantity of MSCs for therapy from the bone marrow poses a challenge. Conversely, the number of ASCs that can be harvested from 1 g of adipose tissue (5×103 cells/g) is 500 times greater than that of BM-MSCs for the equivalent amount of the bone marrow [34]. Additionally, ASCs exhibit a faster proliferation rate than BM-MSCs. Therefore, harvesting the required number of ASCs is relatively easy. Regarding immunomodulatory effects, ASCs surpass other types of MSCs, including BM-MSCs, in suppressing the proliferation of T cells, inhibiting the activation and immunoglobulin production of B cells, and suppressing monocyte differentiation into mature dendritic cells [36,37,38,39,40]. Furthermore, ASCs exhibit greater potency than other MSCs based on their proangiogenic, antiapoptotic, and antioxidative effects [41,42,43]. Collectively, these findings indicate that ASCs may be more effective in suppressing immune response compared to other types of MSCs, including BM-MSCs.

While both ASCs and umbilical cord blood-derived MSCs (UC-MSCs) exhibit significant immunomodulatory properties, current literature delineates several distinct advantages of ASCs over UC-MSCs. Firstly, ASCs are sourced from the adipose tissue, which is not only more abundant but also more readily accessible than the umbilical cord blood, where availability may be constrained [44,45,46]. Secondly, ASCs display enhanced multipotency and differentiation potential. Empirical evidence suggests that ASCs possess a superior ability to differentiate into adipocytes, osteoblasts, and neuronal cells, thus offering broader therapeutic applications than UC-MSCs [46, 47]. Thirdly, ASCs demonstrate more potent immunomodulatory effects than UC-MSCs. Comparative studies indicate that ASCs may have a more pronounced cytokine secretion profile, significantly impacting immune responses [45, 46]. Fourthly, analyses of proliferation and anti-apoptotic capacities revealed no significant differences between ASCs and UC-MSCs, indicating that both cell types exhibit comparable growth and survival capabilities [46]. In summary, the advantages of ASCs over UC-MSCs, particularly their more accessible source, enhanced multipotency, differentiation potential, and stronger immunomodulatory effects, while maintaining comparable proliferative and anti-apoptotic abilities, position them as preferable candidates for applications in tissue engineering and regenerative medicine.

Table 1 shows a comparison of characteristics among ASCs, BM-MSCs, and UC-MSCs.

Table 1 Comparison of the characteristics between ASCs, BM-MSCs, and UC-MSCs

MSC/AdSC therapy for preclinical models of connective tissue and interstitial lung diseases

Rheumatoid arthritis model

Studies using the collagen type II-induced arthritis model reported conflicting results. While some studies showed that the systemic administration of MSCs improved arthritis-associated inflammation [48,49,50,51], others reported its ineffectiveness or observed worsening of symptoms [52, 53]. The efficacy of MSC therapy depends on factors, including the route, frequency, and timing of administration. Based on the findings of these previous studies, MSC transplantation should precede collagen induction, with short intervals between each administration. Studies reporting improvements after MSC therapy in the collagen type II-induced arthritis model found a reduced incidence of arthritis, lower disease activity score (DAS), an improved balance between pro- and anti-inflammatory cytokines, and reduced pathological scores reflecting the degree of joint destruction. Regarding ASCs, the systemic administration of human ASCs in DBA/1 mice induced with collagen-induced arthritis notably alleviated arthritis severity. This therapeutic effect originated from the suppression of two pivotal disease components: the Th1-driven autoimmune response and the associated inflammatory reaction [50]. Ueyama et al. reported reduced intra-articular inflammation and significant cartilage regeneration in SKG mice through the local injection of single-cell mouse ADSCs and three-dimensionally cultured ADSC spheroids [51].

Systemic lupus erythematosus model

The therapeutic effectiveness of MSCs in the SLE mouse model shows variability, influenced by factors such as the mouse strain and origin or condition of the MSCs. Upon transplantation of BM-MSCs into MRL/Ipr mice, a decrease in serum levels of anti-ds-DNA antibodies, antinuclear antibodies, and immunoglobulins was observed. Concurrently, kidney function improved, complement 3 levels decreased, and glomerular IgG deposition lessened [54, 55]. Conversely, the systemic administration of BM-MSCs into NZB/W F1 mice was reported ineffective and, in some cases, exacerbating the disease [56, 57]. Gu et al. utilized MRL/Ipr and NZB/W F1 mouse models, demonstrating that BM-MSCs derived from healthy young and diseased mice ameliorated SLE-like disease while reducing T and B lymphocyte levels in the spleen. However, BM-MSCs derived from old NZB/W F1 mice did not reduce spleen weight, IgG deposition, kidney disease, or inflammation in the renal interstitium [58]. In contrast to BM-MSCs, Gu et al. utilized umbilical cord blood-derived MSCs (UC-MSCs) in MRL/lpr mice. They demonstrated a dose-dependent improvement in lupus nephritis-associated [59]. Chang et al., using NZB/W F1 mice, demonstrated that the transplantation of human UC-MSCs significantly delayed the onset of proteinuria, reduced serum anti-ds-DNA antibody levels, mitigated renal dysfunction, and prolonged mouse survival [60]. Zhang et al. also reported marked improvement in nephritis in MRL/lpr mice following weekly systemic administration of ASCs at a dose of 1×106 cells for eight consecutive weeks. They observed a substantial decrease in Th17 cells within the spleen and a significant increase in Treg cells [61]. Choi et al. also reported that long-term repeated administration of human ASCs improved SLE symptoms in NZB/W F1 mice [62]. Mice-administered ASCs exhibited a higher survival rate than those in the control group, demonstrating improvements in pathological and serological abnormalities, enhanced immune functions, and reduced proteinuria incidence. This administration also resulted in a notable decrease in serum anti-ds-DNA antibody and urea nitrogen levels, alongside increased serum granulocyte–macrophage colony-stimulating factor, IL-4, and IL-10. Furthermore, a significant rise in Treg cell proportion was observed in the spleen of ASC-administered mice.

Systemic sclerosis model

In a hypochlorous acid (HOCl)-induced SSc mouse model, characterized by induced fibrosis in the skin and lung through HOCl, a systemic administration of 2.5×105 BM-MSCs was conducted thrice [63]. The result showed that BM-MSC administration reduced the deposition of all collagen in the skin and lung tissues, and it downregulated the expression of αSMA and TGF-β1 mRNAs. Additionally, the level of anti-Scl-70 antibodies in the serum decreased, alongside a reduction in macrophage infiltration and T cells in the skin. The treatment also led to an improvement in tissue remodeling. Moreover, BM-MSCs did not accumulate in the skin, and they were cleared from the lungs within several days. Similar results were observed in allotransplantation, xenotransplantation, and syngeneic transplantation. The authors also compared the treatment effects of human BM-MSCs and ASCs in HOCl-SSc mice, revealing that ASCs were more effective in suppressing dermal thickening. Furthermore, ASCs significantly downregulated mRNA expression for inflammatory cytokines and factors associated with tissue remodeling in the skin and lung tissues compared to BM-MSCs [64]. The subcutaneous administration of autologous ASCs improved dermal fibrosis in the bleomycin-induced skin fibrosis mouse model [65]. Furthermore, intravenous administration of allogeneic ASCs attenuated skin fibrosis in the bleomycin-induced scleroderma and Scl-cGVHD mouse models [66, 67].

Interstitial lung disease model

An ILD model utilizing BLM-induced lung fibrosis has been employed to evaluate the efficacy of MSCs. A review consolidating 36 preclinical trials of MSC therapy for BLM-induced pulmonary fibrosis models has been published [68]. In these studies, the therapy consistently demonstrated a reduction in various aspects of BLM-induced pulmonary damage, including tissue inflammation, inflammatory cell infiltration, inflammatory cytokine expression, extracellular matrix production, and collagen deposition. This collectively demonstrated an improvement in lung fibrosis scores. Several studies have highlighted the effectiveness of intraperitoneally or intravenously administered ASCs and condition medium from ASCs in inhibiting pulmonary fibrosis in BLM-induced ILD mice [69,70,71,72].

Specific animal models for ILD associated with CTD have not yet been established. The ILD induced by BLM administration through the airway triggers localized inflammatory and fibrotic changes around the peribronchiolar region. However, these alterations do not precisely mimic the pathological manifestations of ILD associated with CTD. Conversely, continuous subcutaneous BLM administration via osmotic minipump results in inflammatory and fibrotic changes on the pleural side. The lesion distribution originating from these changes is comparable to that observed in ILD associated with CTD [73]. Consequently, we investigated the therapeutic effect of intravenous ASC transplantation on a BLM-induced ILD mice model exhibiting diffuse lesions on the pleural side. The results showed that ASCs inhibited inflammation and fibrosis in the lungs in a manner dependent on the number of administered cells [74].

The abovementioned MSCs/ASCs therapies for preclinical models of CTDs and ILD are presented in Table 2.

Table 2 MSCs/ASCs therapy for preclinical models of CTDs and ILD

Pharmacologically primed AdSC therapy in CTD model

Current challenges in AdSC therapy

Intravenous administration of ASCs presents several associated challenges. A preclinical study found that mice developed pulmonary embolisms following the intravenous administration of a large number of mASCs [75]. Moreover, a clinical study revealed that three patients who were intravenously administered ASCs developed pulmonary embolism [76]. Although minimizing the number of intravenously administered ASCs is preferable, the treatment effect may also be diminished with a low cell count. Elderly individual-derived ASCs typically exhibit poorer proliferative and chemotactic activities than those derived from young individuals [77]. Furthermore, MSCs derived from patients with SLE exhibit characteristics associated with the initial phase of aging and demonstrate reduced function [78], while ASCs derived from patients with scleroderma demonstrate reduced proliferative, metabolic, and chemotactic activities compared to those from healthy individuals [79]. Therefore, treating the potential induction of pulmonary embolism by intravenous ASC administration and improving the functionality of ASCs in elderly patients and those with underlying diseases necessitate the development of novel strategies.

Previous reports on pharmacologically primed AdSC therapy in the CTD model

To address the challenges associated with ASC treatment, recent studies have explored the therapeutic application of pharmacologically enhanced ASCs in CTD animal models. Kim et al. observed a significant alleviation of symptoms related to graft-versus-host disease in NOD-SCID mice with the administration of MSCs, including ASCs with IFN-γ. This finding implies that the therapeutic effect may be attributed to the induction of IDO expression in MSCs through the IFN-γ-JAK-STAT1 pathway [80]. Zolfaghari et al. demonstrated that TLR3 ligand-primed ASCs using polyinosinic:polycytidylic acid (poly I:C) reduced splenocyte proliferation in vitro. Furthermore, in animal models of adjuvant-induced arthritis, these cells significantly improved clinical and histopathological severity, notably reducing TNF-α and IL-6 levels in serum [81]. Jang et al. reported that metformin enhanced the immunoregulatory effect of ASCs by upregulating STAT1 expression through the AMPK/mTOR pathway in vitro. Administering metformin-treated ASCs markedly improved disease activity, including inflammatory phenotype, glomerulonephritis, proteinuria, and anti-dsDNA IgG antibody production in MRL/lpr mice. Additionally, metformin-treated ASCs inhibited CD4-CD8- T cell proliferation and modulated the Th17/Treg cell ratio [82].

Heparin-primed ASC therapy in the CTD model

Heparin—an inhibitor of antithrombin III and factor Xa used in preventing and treating thrombosis [83]—interacts with various proteins, demonstrating multifaceted efficacy [84]. Heparin stabilizes HGF dimers, thereby promoting the dimerization and activation of the c-Met receptor [85]. It augments fibroblast growth factor and bone morphogenetic protein 4 gene expression, consequently enhancing proliferation and pluripotency in BM-MSCs and embryonic stem cells, respectively [86,87,88]. Furthermore, heparin stimulates HGF biosynthesis in diverse cell types, including lung fibroblasts, promyelocytic leukemia cells, and umbilical vein endothelial cells [89, 90]. Therefore, we hypothesized that heparin-activated ASCs might exhibit a synergistic beneficial effect on SSc or ILD by promoting anti-inflammatory and antifibrotic responses. We investigated the effect of heparin on ASC functions. We compared the therapeutic effects of heparin-enhanced ASCs (hepASCs) to that of ASCs alone in mouse models of SSc and ILD [91, 92]. Figure 2 shows the procedures for isolating and culturing mASCs, the preparation and treatment of SSc and ILD model mice, and the therapeutic effects of SSc skins and ILD lungs. The results showed that heparin significantly increased ASC numbers and enhanced their migratory, anti-inflammatory, and antifibrotic effects in vitro. Additionally, hepASCs demonstrated increased accumulation in the skin or lung tissues compared to ASCs alone. Moreover, in mice with bleomycin-induced SSc or ILD, intravenously administered hepASCs significantly reduced skin thickness and hydroxyproline content compared to the SSc pathological model group. It also decreased collagen deposition and hydroxyproline levels in the lungs compared to those in the ILD pathological model group.

Fig. 2
figure 2

Enhanced techniques for isolation and co-cultivating heparin-enhanced murine adipose-derived stem cells, with details on preparation, treatment protocols, and therapeutic effects for systemic sclerosis and interstitial lung disease model mice. Isolation and culture of ASCs: ASCs were isolated from the inguinal adipose tissue of 8-week-old female Balb/c mice (for SSc) and 8-week-old female C57BL/6J mice (for ILD), following euthanasia by cervical dislocation under isoflurane anesthesia. The adipose tissue was washed in PBS, minced, and digested with type I collagenase (1.0 mg/mL in 1% BSA/HBSS (+)) at 37 °C for 30 min. After filtration and centrifugation, the cell pellet was resuspended in erythrocyte lysis buffer, followed by medium addition and centrifugation. The resulting mesenchymal ASCs were cultured at a density of 1×106 cells per 90 mm cell-culture dish and utilized at the third passage. Preparation and treatment of SSc and ILD model mice: HepASCs were cultured in DMEM/F-12 with 10% FBS, 1% Pen-Strep, and LMWH. For SSc studies, female BALB/c mice (8 weeks old) received daily subcutaneous injections of 100 μg/100 μL BLM for 21 days. These mice were divided into three groups: BLM alone, BLM with ASCs, and BLM with hepASCs. For ILD studies, female C57BL/6J mice (13 weeks old) were divided into four groups: untreated (normal), BLM alone, BLM with ASCs, and BLM with hepASCs. BLM (3 mg in 100 μL saline) was administered subcutaneously over 7 days. For both studies, cells (1.0×105 for lung, 2.5×104 for skin in 100 μL PBS) or PBS was intravenously injected. Mice were euthanized on day 21 for skin analysis and day 28 for lung analysis, with respective organs harvested for evaluation. Therapeutic effects of SSc skins and ILD lungs: intravenously administered hepASCs significantly reduced skin thickness and hydroxyproline content in mice with bleomycin-induced SSc compared to that of the SSc pathological model group. Additionally, hepASCs decreased collagen deposition and hydroxyproline levels in the lungs compared to those in the ILD pathological model group. ASCs adipose-derived mesenchymal stem/stromal cells, LMWH low-molecular-weight heparin, SSc systemic sclerosis, ILD interstitial lung disease, hepASCs ASCs enhanced by LMWH, BLM bleomycin. Arrows indicate collagen deposition area; scale bars in skins and lungs are 300 μm and 100 μm, respectively. Data were analyzed using nonparametric one-way ANOVA, followed by a multiple comparison test. *P < 0.05, **P < 0.01, and ***P < 0.001, a significant difference between the linked groups

Potential of MSCs transplant rejection and activation of the immune system

Several reports show that transplanting human MSCs into mice can trigger transplant rejection despite MSCs’ immunosuppressive properties. Various studies indicate that human MSCs provoke an immune response in mice, contrary to prior beliefs about their immune-privileged status [93,94,95]. This response is most pronounced in xenogeneic transplantation (human MSCs into mice), followed by allogeneic and syngeneic transplantation [94, 95]. Notably, xenotransplantation results in significant infiltration of leukocytes at the injection site, indicating innate immune system activation [93]. While inevitable for preclinical evaluation, xenotransplantation poses challenges for clinical translation of human MSC therapy. Studies suggest using immunosuppressants, such as dexamethasone and tacrolimus, to manage immune responses [93, 96]. Effective immunosuppression protocols are crucial for successful xenotransplantation experiments. In essence, despite MSCs’ immunosuppressive properties, transplant rejection can occur when transplanting them into mice. Mitigating immune responses through careful immunosuppressant use is thus vital for successful xenotransplantation studies.

Clinical applications of MSCs/ASCs for CTDs and ILD

Table 3 shows the published major clinical trials of MSCs/ASCs therapy for RA, SLE, SSc, and ILD patients.

Table 3 Published major clinical trials of MSCs/ASCs therapy for RA, SLE, SSc, and ILD patients

Rheumatoid arthritis

In an uncontrolled phase I/II study, 136 patients with RA resistant to conventional drug treatments were treated with intravenous administration of 4×107 UC-MSCs. They were followed up for 8 months [97]. The result showed significant improvements in the DAS of RA, health assessment questionnaire, and American College of Rheumatology (ACR) response and an increased frequency of Tregs in the peripheral blood. Patients whose disease activity worsened following the treatment received an additional administration of UC-MSCs after 3 months. At the end of the study, 58% of the patients had achieved ACR20, and none of them experienced severe adverse events. Additionally, reports indicate the potential inhibitory effects of single intravenous administrations of BM-MSCs and UC-MSCs on the disease activity of RA in a limited number of cases [98,99,100,101,102]. In 2017, Álvaro-Gracia et al. conducted a multicenter, non-randomized, single-blinded (double-blinded for efficacy assessment), placebo-controlled phase Ib/IIa study to examine the safety of intravenously administered allogenic ASCs in 53 patients with RA with active disease refractory to at least two administrations of biological DMARDs [103]. These researchers administered 1×106, 2×106, or 4×106 ASCs. The results showed that a higher proportion of patients achieved ACR20 and ACR50 and improved DAS in a dose-dependent manner than those in the placebo group. This study primarily aimed to determine the safety of ASCs, and the findings revealed that the treatment was well-tolerated with no serious side effects, except for one patient who developed lacunar infarction.

Systemic lupus erythematosus

Studies conducted to date have examined the use of MSCs for treatment-resistant SLE, involving a few to over 80 patients. MSCs employed in these studies were allogeneic BM- or UC-MSCs, administered intravenously. These studies collectively found that MSC therapy was well-tolerated and effective in improving kidney function and disease activity, reducing proteinuria, and reducing anti-ds-DNA antibody levels. MSC transplantation resulted in an elevation in Tregs in the peripheral blood and restored the balance between Th1 and Th2 cytokines [104,105,106,107,108,109,110]. One of the representative trials conducted by Wang et al. was a multicenter study in which 40 patients with refractory SLE were administered 1×106/kg UC-MSCs on days 0 and 7 [106]. Following a 6-month follow-up, 32.5% of the patients achieved complete remission, while 27.5% attained partial remission. However, 17.5% of the patients experienced recurrence. The dosage of immunosuppressants required was significantly reduced in most patients. Throughout the follow-up period, three patients developed herpes simplex virus infection, and one patient contracted tuberculosis. Furthermore, three patients died during the follow-up. The causes of death included acute cardiac arrest 7 days after MSC administration, severe pulmonary hypertension that manifested 8 months after the treatment, and pulmonary infection. Nonetheless, none of these causes were likely associated with MSC therapy. A reported case described a 9-year-old girl with SLE showed improved disease activity after AdSC administration via one nasal injection, one lymph node injection, and two intravenous injections [111].

Systemic sclerosis

Based on previous studies on MSC therapy for SSc patients, a systematic review and meta-analysis were conducted to assess the efficacy and safety of MSCs in treating SSc [112,113,114,115,116,117,118,119,120]. The study encompassed nine clinical trials involving 133 adult patients with SSc up to February 1, 2021. These trials included one case of intravenous BM-MSC administration, two cases of intravenous UC-MSC administration, three cases of subcutaneous stromal vascular fraction administration, and three cases of subcutaneous AdSC administration. MSC therapy significantly reduced the modified Rodnan skin score, digital ulcer count, oral handicap scales, and visual analog scales in patients with SSc. Although a few patients presented with injection site swelling, diarrhea, and joint pain, these issues resolved on their own, and no severe adverse events were observed. Overall, the utilization of MSCs was deemed safe.

Interstitial lung disease

To date, no clinical studies have specifically examined MSC therapy for ILD associated with CTD. However, studies on MSC therapy for idiopathic pulmonary fibrosis (IPF) exist. Glassberg et al. conducted the first trial assessing BM-MSCs in patients with mild to moderate IPF [121]. They infused BM-MSCs from two donors into nine patients. They observed no severe treatment-related side effects with doses of up to 2×108 cells over 60 weeks. However, two deaths unrelated to the study occurred, and 78% of patients reported non-serious adverse events such as bronchitis and colds. Nevertheless, declines in lung function persisted below the recognized threshold for disease progression, with a 3.0% and 5.4% decrease in predicted FVC and DLCO 5.4% after 60 weeks, respectively. Chambers et al. conducted an open-label, single-center, non-randomized, uncontrolled, phase Ib dose-escalation study involving eight patients with moderately severe but not advanced IPF (FVC ≥50% of predicted normal; DLCO ≥35% of predicted normal) [122]. The patients were administered two intravenous doses of HLA-unmatched placenta-derived MSCs (1×106/kg body weight, n = 4; 2×106/kg/kg body weight, n = 4). During the 6-month follow-up period, one episode of lingular left lobe consolidation, considered possibly treatment-related, occurred 5 days after the infusion of the lowest cell dose. No other severe side effects attributed to the administration of MSCs were reported. Tzouvelekis et al. conducted an open-label, single-group, non-comparative, phase-Ib clinical trial comprising 14 patients with mild or moderate IPF (FVC >50% of predicted normal; DLCO >35% of predicted normal) [123]. The patients were treated with endobronchial administration of autologous adipose tissue-derived stromal cell-stromal vascular fraction (0.5×106/kg body weight/dose, administered thrice at monthly intervals). No other severe side effects directly attributed to the administration of these cells were reported.

Table 4 presents the current clinical trials identified through the National Library of Medicine website of the National Institutes of Health for RA, SLE, SSc, and ILD (visit: www.clinicaltrials.gov). The number of clinical trials assessing the effectiveness and safety of BM-MSCs and UC-MSCs for CTD and ILD is on the rise. However, studies on AdSCs in this context remain limited, posing a future challenge.

Table 4 Current clinical trials for connective tissue disease (CTD) and interstitial lung disease (ILD) from the NIH National Library of Medicine database

Conclusion

In conclusion, recent advances in the treatment of CTD have yielded a myriad of therapeutic options, particularly in the realm of RA and SLE. Although in this regard, conventional immunosuppressants and biological or targeted synthetic DMARDs have shown certain efficacy, challenges persist with respect to issues such as side effects and the lack of effective treatments for various CTDs, including SSc. CTD often manifests with organ damage, and ILD has emerged as a particular concern, given its significant impact on patient prognosis. Despite the current application of treatment modalities involving corticosteroids, immunosuppressive agents, and anti-fibrotic agents, CTD is still associated with a high incidence of respiratory failure and mortality.

In this context, MSCs have gain considerable attention with respect to their regenerative potential. Specifically, ASCs are gaining prominence on account of their accessibility and immunomodulatory effects. In this review, we have provided an overview of evidence obtained from preclinical studies that have evaluated the efficacy of MSC therapy in managing CTD and ILD, with a particular emphasis on AdSC therapy. The immunomodulatory effects of MSCs are highlighted, emphasizing their capacity to modulate a range of different immune cell types and cytokines, thereby identifying these stem cells as a promising avenue for CTD treatment. Notably, compared with other MSC types, such as BM-MSCs, ASCs have been established to have a number advantages in terms of accessibility, proliferation rates, and immunomodulatory potency.

Furthermore, our review of preclinical models, including those for RA, SLE, SSc, and ILD, highlight the potential of MSC/ASC therapy in ameliorating disease severity and improving patient outcomes. In addition, we also assess the challenges associated with the application ASC therapy, including intravenous administration-related complications. In response to these challenges, recent studies have begun the evaluate the efficacy of pharmacologically primed ASC therapy, introducing innovative approaches designed to enhance therapeutic effects. Among these, investigations into heparin-primed ASCs have yielded promising results in mitigating CTD-related symptoms.

We conclude the review by providing insights into the clinical applications of MSCs/ASCs for CTDs, including rheumatoid arthritis, systemic lupus erythematosus, systemic sclerosis, and interstitial lung disease. Clinical trials conducted to date have verified the safety and potential efficacy of MSCs, particularly ASCs, in treating refractory cases, paving the way for further research and development. In summary, the multifaceted potential of MSCs, particularly ASCs, in managing CTD and ILD represents a promising avenue for future therapeutic interventions. Moreover, the development of pharmacologically enhanced ASCs will provide innovative strategies for overcoming current challenges, thereby broadening the scope for advancing the field and improving patient outcomes.

Availability of data and materials

Not applicable.

Abbreviations

MSCs:

Mesenchymal stem cells

ASCs:

Adipose-derived MSCs

CTD:

Connective tissue disease

ILD:

Interstitial lung disease

RA:

Rheumatoid arthritis

DMARDs:

Disease-modifying anti-rheumatic drugs

SLE:

Systemic lupus erythematosus

CYC:

Cyclophosphamide

MMF:

Mycophenolate mofetil

SSc:

Systemic sclerosis

HLA:

Human leukocyte antigen

TGF-β1:

Transforming growth factor-β1

PGE2:

Prostaglandin E2

HGF:

Hepatocyte growth factor

NK:

Natural killer

TSG-6:

TNF-α-stimulated gene 6

MMPs:

Matrix metalloproteinases

mRNA:

Messenger RNA

BM-MSCs:

Bone marrow-derived MSCs

UC-MSCs:

Umbilical cord blood-derived MSCs

HOCl:

Hypochlorous acid

hepASCs:

Heparin-enhanced ASCs

ACR:

American College of Rheumatology

IPF:

Idiopathic pulmonary fibrosis

References

  1. Smolen JS, Landewé R, Bijlsma J, Burmester G, Chatzidionysiou K, Dougados M, et al. EULAR recommendations for the management of rheumatoid arthritis with synthetic and biological disease-modifying antirheumatic drugs: 2016 update. Ann Rheum Dis. 2017;76:960–77.

    Article  PubMed  Google Scholar 

  2. Thong B, Olsen NJ. Systemic lupus erythematosus diagnosis and management. Rheumatology (Oxford). 2017;56:i3–13.

    CAS  PubMed  Google Scholar 

  3. Strand V, Levy RA, Cervera R, Petri MA, Birch H, Freimuth WW, et al. Improvements in health-related quality of life with belimumab, a B-lymphocyte stimulator-specific inhibitor, in patients with autoantibody-positive systemic lupus erythematosus from the randomised controlled BLISS trials. Ann Rheum Dis. 2014;73:838–44.

    Article  CAS  PubMed  Google Scholar 

  4. Jeganathan N, Sathananthan M. Connective tissue disease-related interstitial lung disease: prevalence, patterns, predictors, prognosis, and treatment. Lung. 2020;198:735–59.

    Article  PubMed  Google Scholar 

  5. Vacchi C, Sebastiani M, Cassone G, Cerri S, Casa GD, Salvarani C, et al. Therapeutic options for the treatment of interstitial lung disease related to connective tissue diseases A narrative review. J Clin Med. 2020;9:407.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Yang M, Wu Y, Liu X, Zhao C, Li T, Li T, et al. Efficacy and safety of antifibrotic agents in the treatment of CTD-ILD and RA-ILD: a systematic review and meta-analysis. Respir Med. 2023;216:107329.

    Article  PubMed  Google Scholar 

  7. Murphy MB, Moncivais K, Caplan AI. Mesenchymal stem cells: environmentally responsive therapeutics for regenerative medicine. Exp Mol Med. 2013;45:e54.

  8. Caruso M, Evangelista M, Parolini O. Human term placental cells: phenotype, properties and new avenues in regenerative medicine. Int J Mol Cell Med. 2012;1:64–74.

  9. Lalu MM, McIntyre L, Pugliese C, Fergusson D, Winston BW, Marshall JC, et al. Safety of cell therapy with mesenchymal stromal cells (SafeCell): a systematic review and meta-analysis of clinical trials. PLoS One. 2012;7:e47559.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Qin L, Liu N, Bao CL, Yang DZ, Ma GX, Yi WH, et al. Mesenchymal stem cells in fibrotic diseases—the two sides of the same coin. Acta Pharmacol Sin. 2023;44:268–87.

  11. Molnar V, Pavelić E, Vrdoljak K, Čemerin M, Klarić E, Matišić V, et al. Mesenchymal stem cell mechanisms of action and clinical effects in osteoarthritis: A narrative review. Genes (Basel). 2022;13:949.

  12. Murphy MB, Moncivais K, Caplan AI. Mesenchymal stem cells: environmentally responsive therapeutics for regenerative medicine. Exp Mol Med. 2013;45:e54.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Weiss DJ. Stem cells, cell therapies, and bioengineering in lung biology and diseases. Comprehensive review of the recent literature 2010–2012. Ann Am Thorac Soc. 2013;10:S45–97.

    Article  PubMed  PubMed Central  Google Scholar 

  14. Glennie S, Soeiro I, Dyson PJ, Lam EW, Dazzi F. Bone marrow mesenchymal stem cells induce division arrest anergy of activated T cells. Blood. 2005;105:2821–7.

    Article  CAS  PubMed  Google Scholar 

  15. Di Nicola M, Carlo-Stella C, Magni M, Milanesi M, Longoni PD, Matteucci P, et al. Human bone marrow stromal cells suppress T-lymphocyte proliferation induced by cellular or nonspecific mitogenic stimuli. Human bone marrow stromal cells suppress T-lymphocyte proliferation induced by cellular or nonspecific mitogenic stimuli. Blood. 2002;99:3838–43.

    Article  PubMed  Google Scholar 

  16. Meisel R, Zibert A, Laryea M, Göbel U, Däubener W, Dilloo D. Human bone marrow stromal cells inhibit allogeneic T-cell responses by indoleamine 2,3-dioxygenase-mediated tryptophan degradation. Blood. 2004;103:4619–21.

    Article  CAS  PubMed  Google Scholar 

  17. Bouffi C, Bony C, Courties G, Jorgensen C, Noël D. IL-6-dependent PGE2 secretion by mesenchymal stem cells inhibits local inflammation in experimental arthritis. PLoS One. 2010;5:e14247.

    Article  PubMed  PubMed Central  Google Scholar 

  18. Nauta AJ, Fibbe WE. Immunomodulatory properties of mesenchymal stromal cells. Blood. 2007;110:3499–506.

    Article  CAS  PubMed  Google Scholar 

  19. Luz-Crawford P, Djouad F, Toupet K, Bony C, Franquesa M, Hoogduijn MJ, et al. Mesenchymal stem cell-derived interleukin 1 receptor antagonist promotes macrophage polarization and inhibits B cell differentiation. Stem Cells. 2016;34:483–92.

    Article  CAS  PubMed  Google Scholar 

  20. Ke F, Zhang L, Liu Z, Yan S, Xu Z, Bai J, et al. Soluble tumor necrosis factor receptor 1 released by skin-derived mesenchymal stem cells is critical for inhibiting Th17 cell differentiation. Stem Cells Transl Med. 2016;5:301–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Noël D, Djouad F, Bouffi C, Mrugala D, Jorgensen C. Multipotent mesenchymal stromal cells and immune tolerance. Leuk Lymphoma. 2007;48:1283–9.

    Article  PubMed  Google Scholar 

  22. Djouad F, Bouffi C, Ghannam S, Noël D, Jorgensen C. Mesenchymal stem cells: innovative therapeutic tools for rheumatic diseases. Nat Rev Rheumatol. 2009;5:392–9.

    Article  CAS  PubMed  Google Scholar 

  23. Luz-Crawford P, Noël D, Fernandez X, Khoury M, Figueroa F, Carrión F, et al. Mesenchymal stem cells repress Th17 molecular program through the PD-1 pathway. PLoS One. 2012;7:e45272.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Sotiropoulou PA, Perez SA, Gritzapis AD, Baxevanis CN, Papamichail M. Interactions between human mesenchymal stem cells and natural killer cells. Stem Cells. 2006;24:74–85.

    Article  PubMed  Google Scholar 

  25. Choi H, Lee RH, Bazhanov N, Oh JY, Prockop DJ. Anti-inflammatory protein TSG-6 secreted by activated MSCs attenuates zymosan-induced mouse peritonitis by decreasing TLR2/NF-κB signaling in resident macrophages. Blood. 2011;118:330–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Corcione A, Benvenuto F, Ferretti E, Giunti D, Cappiello V, Cazzanti F, et al. Human mesenchymal stem cells modulate B-cell functions. Blood. 2006;107:367–72.

    Article  CAS  PubMed  Google Scholar 

  27. Rosado MM, Bernardo ME, Scarsella M, Conforti A, Giorda E, Biagini S, et al. Inhibition of B-cell proliferation and antibody production by mesenchymal stromal cells is mediated by T cells. Stem Cells Dev. 2015;24:93–103.

    Article  CAS  PubMed  Google Scholar 

  28. Jiang XX, Zhang Y, Liu B, Zhang SX, Wu Y, Yu XD, et al. Human mesenchymal stem cells inhibit differentiation and function of monocyte-derived dendritic cells. Blood. 2005;105:4120–6.

    Article  CAS  PubMed  Google Scholar 

  29. Nauta AJ, Kruisselbrink AB, Lurvink E, Willemze R, Fibbe WE. Mesenchymal stem cells inhibit generation and function of both CD34+-derived and monocyte-derived dendritic cells. J Immunol. 2006;177:2080–7.

    Article  CAS  PubMed  Google Scholar 

  30. Ramasamy R, Fazekasova H, Lam EW, Soeiro I, Lombardi G, Dazzi F. Mesenchymal stem cells inhibit dendritic cell differentiation and function by preventing entry into the cell cycle. Transplantation. 2007;83:71–6.

    Article  PubMed  Google Scholar 

  31. Németh K, Leelahavanichkul A, Yuen PS, Mayer B, Parmelee A, Doi K, et al. Bone marrow stromal cells attenuate sepsis via prostaglandin E(2)-dependent reprogramming of host macrophages to increase their interleukin-10 production. Nat Med. 2009;15:42–9.

    Article  PubMed  Google Scholar 

  32. Maumus M, Jorgensen C, Noël D. Mesenchymal stem cells in regenerative medicine applied to rheumatic diseases: role of secretome and exosomes. Biochimie. 2013;95:2229–34.

    Article  CAS  PubMed  Google Scholar 

  33. Zannettino AC, Paton S, Arthur A, Khor F, Itescu S, Gimble JM, et al. Multipotential human adipose-derived stromal stem cells exhibit a perivascular phenotype in vitro and in vivo. J Cell Physiol. 2008;214:413–21.

    Article  CAS  PubMed  Google Scholar 

  34. Ohgushi H, Caplan AI. Stem cell technology and bioceramics: from cell to gene engineering. J Biomed Mater Res. 1999;48:913–27.

    Article  CAS  PubMed  Google Scholar 

  35. Mareschi K, Ferrero I, Rustichelli D, Aschero S, Gammaitoni L, Aglietta M, et al. Expansion of mesenchymal stem cells isolated from pediatric and adult donor bone marrow. J Cell Biochem. 2006;97:744–54.

    Article  CAS  PubMed  Google Scholar 

  36. Noël D, Caton D, Roche S, Bony C, Lehmann S, Casteilla L, et al. Cell specific differences between human adipose-derived and mesenchymal-stromal cells despite similar differentiation potentials. Exp Cell Res. 2008;314:1575–84.

    Article  PubMed  Google Scholar 

  37. Puissant B, Barreau C, Bourin P, Clavel C, Corre J, Bousquet C, et al. Immunomodulatory effect of human adipose tissue-derived adult stem cells: comparison with bone marrow mesenchymal stem cells. Br J Haematol. 2005;129:118–29.

    Article  PubMed  Google Scholar 

  38. Strioga M, Viswanathan S, Darinskas A, Slaby O, Michalek J. Same or not the same? Comparison of adipose tissue-derived versus bone marrow-derived mesenchymal stem and stromal cells. Stem Cells Dev. 2012;21:2724–52.

    Article  CAS  PubMed  Google Scholar 

  39. Bochev I, Elmadjian G, Kyurkchiev D, Tzvetanov L, Altankova I, Tivchev P, et al. Mesenchymal stem cells from human bone marrow or adipose tissue differently modulate mitogen-stimulated B-cell immunoglobulin production in vitro. Cell Biol Int. 2008;32:384–93.

    Article  CAS  PubMed  Google Scholar 

  40. Ivanova-Todorova E, Bochev I, Mourdjeva M, Dimitrov R, Bukarev D, Kyurkchiev S, et al. Adipose tissue-derived mesenchymal stem cells are more potent suppressors of dendritic cells differentiation compared to bone marrow-derived mesenchymal stem cells. Immunol Lett. 2009;126:37–42.

    Article  CAS  PubMed  Google Scholar 

  41. Rehman J, Traktuev D, Li J, Merfeld-Clauss S, Temm-Grove CJ, Bovenkerk JE, et al. Secretion of angiogenic and antiapoptotic factors by human adipose stromal cells. Circulation. 2004;109:1292–8.

    Article  PubMed  Google Scholar 

  42. Kim WS, Park BS, Kim HK, Park JS, Kim KJ, Choi JS, et al. Evidence supporting antioxidant action of adipose-derived stem cells: protection of human dermal fibroblasts from oxidative stress. J Dermatol Sci. 2008;49:133–42.

    Article  CAS  PubMed  Google Scholar 

  43. Kim WS, Park BS, Sung JH. The wound-healing and antioxidant effects of adipose-derived stem cells. Expert Opin Biol Ther. 2009;9:879–87.

    Article  CAS  PubMed  Google Scholar 

  44. Nagamura-Inoue T, He H. Umbilical cord-derived mesenchymal stem cells: Their advantages and potential clinical utility. World J Stem Cells. 2014;6:195–202.

    Article  PubMed  PubMed Central  Google Scholar 

  45. Tan K, Zheng K, Li D, Lu H, Wang S, Sun X. Impact of adipose tissue or umbilical cord derived mesenchymal stem cells on the immunogenicity of human cord blood derived endothelial progenitor cells. PLoS One. 2017;12:e0178624.

    Article  PubMed  PubMed Central  Google Scholar 

  46. Hu L, Hu J, Zhao J, Liu J, Ouyang W, Yang C, et al. Side-by-side comparison of the biological characteristics of human umbilical cord and adipose tissue-derived mesenchymal stem cells. Biomed Res Int. 2013;2013:438243.

    Article  PubMed  PubMed Central  Google Scholar 

  47. Rashnonejad A, Ercan G, Gunduz C, Akdemir A, Tiftikcioglu YO. Comparative analysis of human UCB and adipose tissue derived mesenchymal stem cells for their differentiation potential into brown and white adipocytes. Mol Biol Rep. 2018;45:233–44.

    Article  CAS  PubMed  Google Scholar 

  48. Augello A, Tasso R, Negrini SM, Cancedda R, Pennesi G. Cell therapy using allogeneic bone marrow mesenchymal stem cells prevents tissue damage in collagen-induced arthritis. Arthritis Rheum. 2007;56:1175–86.

    Article  CAS  PubMed  Google Scholar 

  49. Liu Y, Mu R, Wang S, Long L, Liu X, Li R, et al. Therapeutic potential of human umbilical cord mesenchymal stem cells in the treatment of rheumatoid arthritis. Arthritis Res Ther. 2010;12:R210.

    Article  PubMed  PubMed Central  Google Scholar 

  50. González MA, Gonzalez-Rey E, Rico L, Büscher D, Delgado M. Treatment of experimental arthritis by inducing immune tolerance with human adipose-derived mesenchymal stem cells. Arthritis Rheum. 2009;60:1006–19.

    Article  PubMed  Google Scholar 

  51. Ueyama H, Okano T, Orita K, Mamoto K, Ii M, Sobajima S, et al. Local transplantation of adipose-derived stem cells has a significant therapeutic effect in a mouse model of rheumatoid arthritis. Sci Rep. 2020;10:3076.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Chen B, Hu J, Liao L, Sun Z, Han Q, Song Z, et al. Flk-1+ mesenchymal stem cells aggravate collagen-induced arthritis by up-regulating interleukin-6. Clin Exp Immunol. 2010;159:292–302.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Djouad F, Fritz V, Apparailly F, Louis-Plence P, Bony C, Sany J, et al. Reversal of the immunosuppressive properties of mesenchymal stem cells by tumor necrosis factor alpha in collagen-induced arthritis. Arthritis Rheum. 2005;52:1595–603.

    Article  CAS  PubMed  Google Scholar 

  54. Sun L, Akiyama K, Zhang H, Yamaza T, Hou Y, Zhao S, et al. 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 

  55. Zhou K, Zhang H, Jin O, Feng X, Yao G, Hou Y, et al. Transplantation of human bone marrow mesenchymal stem cell ameliorates the autoimmune pathogenesis in MRL/lpr mice. Cell Mol Immunol. 2008;5:417–24.

    Article  PubMed  PubMed Central  Google Scholar 

  56. Schena F, Gambini C, Gregorio A, Mosconi M, Reverberi D, Gattorno M, et al. 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 

  57. Youd M, Blickarz C, Woodworth L, Touzjian T, Edling A, Tedstone J, et al. Allogeneic mesenchymal stem cells do not protect NZBxNZW F1 mice from developing lupus disease. Clin Exp Immunol. 2010;161:176–86.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Gu F, Molano I, Ruiz P, Sun L, Gilkeson GS. Differential effect of allogeneic versus syngeneic mesenchymal stem cell transplantation in MRL/lpr and (NZB/NZW) F1 mice. Clin Immunol. 2012;145:142–52.

    Article  CAS  PubMed  Google Scholar 

  59. Gu Z, Akiyama K, Ma X, Zhang H, Feng X, Yao G, et al. Transplantation of umbilical cord mesenchymal stem cells alleviates lupus nephritis in MRL/lpr mice. Lupus. 2010;19:1502–14.

    Article  CAS  PubMed  Google Scholar 

  60. Chang JW, Hung SP, Wu HH, Wu WM, Yang AH, Tsai HL, et al. Therapeutic effects of umbilical cord blood-derived mesenchymal stem cell transplantation in experimental lupus nephritis. Cell Transplant. 2011;20:245–57.

    Article  PubMed  Google Scholar 

  61. Zhang W, Feng YL, Pang CY, Lu FA, Wang YF. Transplantation of adipose tissue-derived stem cells ameliorates autoimmune pathogenesis in MRL/lpr mice: modulation of the balance between Th17 and Treg. Z Rheumatol. 2019;78:82–8.

    Article  CAS  PubMed  Google Scholar 

  62. Choi EW, Shin IS, Park SY, Park JH, Kim JS, Yoon EJ, et al. Reversal of serologic, immunologic, and histologic dysfunction in mice with systemic lupus erythematosus by long-term serial adipose tissue-derived mesenchymal stem cell transplantation. Arthritis Rheum. 2012;64:243–53.

    Article  CAS  PubMed  Google Scholar 

  63. Maria AT, Toupet K, Bony C, Pirot N, Vozenin MC, Petit B, et al. Antifibrotic, antioxidant, and immunomodulatory effects of mesenchymal stem cells in HOCl-induced systemic sclerosis. Arthritis Rheumatol. 2016;68:1013–25.

    Article  CAS  PubMed  Google Scholar 

  64. Maria AT, Toupet K, Maumus M, Fonteneau G, Le Quellec A, Jorgensen C, et al. Human adipose mesenchymal stem cells as potent anti-fibrosis therapy for systemic sclerosis. J Autoimmun. 2016;70:31–9.

    Article  CAS  PubMed  Google Scholar 

  65. Chen W, Xia ZK, Zhang MH, Ding GC, Zhang XY, Wang ZX, et al. Adipose tissue-derived stem cells ameliorates dermal fibrosis in a mouse model of scleroderma. Asian Pac J Trop Med. 2017;10:52–6.

    Article  CAS  PubMed  Google Scholar 

  66. Rubio GA, Elliot SJ, Wikramanayake TC, Xia X, Pereira-Simon S, Thaller SR, et al. 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 

  67. Okamura A, Matsushita T, Komuro A, Kobayashi T, Maeda S, Hamaguchi Y, et al. Adipose-derived stromal/stem cells successfully attenuate the fibrosis of scleroderma mouse models. Int J Rheum Dis. 2020;23:216–25.

    Article  CAS  PubMed  Google Scholar 

  68. Zhao Y, Yan Z, Liu Y, Zhang Y, Shi J, Li J, et al. Effectivity of mesenchymal stem cells for bleomycin-induced pulmonary fibrosis: a systematic review and implication for clinical application. Stem Cell Res Ther. 2021;12:470.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Lee SH, Lee EJ, Lee SY, Kim JH, Shim JJ, Shin C, et al. The effect of adipose stem cell therapy on pulmonary fibrosis induced by repetitive intratracheal bleomycin in mice. Exp Lung Res. 2014;40:117–25.

    Article  CAS  PubMed  Google Scholar 

  70. Tashiro J, Elliot SJ, Gerth DJ, Xia X, Pereira-Simon S, Choi R, et al. Therapeutic benefits of young, but not old, adipose-derived mesenchymal stem cells in a chronic mouse model of bleomycin-induced pulmonary fibrosis. Transl Res. 2015;166:554–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Rathinasabapathy A, Bruce E, Espejo A, Horowitz A, Sudhan DR, Nair A, et al. Therapeutic potential of adipose stem cell-derived conditioned medium against pulmonary hypertension and lung fibrosis. Br J Pharmacol. 2016;173:2859–79.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Zhao X, Wu J, Yuan R, Li Y, Yang Q, Wu B, et al. Adipose-derived mesenchymal stem cell therapy for reverse bleomycin-induced experimental pulmonary fibrosis. Sci Rep. 2023;13:13183.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Harrison JH Jr, Lazo JS. High dose continuous infusion of bleomycin in mice: a new model for drug-induced pulmonary fibrosis. J Pharmacol Exp Ther. 1987;243:1185–94.

    CAS  PubMed  Google Scholar 

  74. Kotani T, Masutani R, Suzuka T, Oda K, Makino S, Ii M. Anti-inflammatory and anti-fibrotic effects of intravenous adipose-derived stem cell transplantation in a mouse model of bleomycin-induced interstitial pneumonia. Sci Rep. 2017;7:14608.

    Article  PubMed  PubMed Central  Google Scholar 

  75. Tatsumi K, Ohashi K, Matsubara Y, Kohori A, Ohno T, Kakidachi H, et al. Tissue factor triggers procoagulation in transplanted mesenchymal stem cells leading to thromboembolism. Biochem Biophys Res Commun. 2013;431:203–9.

    Article  CAS  PubMed  Google Scholar 

  76. Jung JW, Kwon M, Choi JC, Shin JW, Park IW, Choi BW, et al. Familial occurrence of pulmonary embolism after intravenous, adipose tissue-derived stem cell therapy. Yonsei Med J. 2013;54:1293–6.

    Article  PubMed  PubMed Central  Google Scholar 

  77. Liu M, Lei H, Dong P, Fu X, Yang Z, Yang Y, et al. Adipose-derived mesenchymal stem cells from the elderly exhibit decreased migration and differentiation abilities with senescent properties. Cell Transplant. 2017;26:1505–19.

    Article  PubMed  PubMed Central  Google Scholar 

  78. Sun LY, Zhang HY, Feng XB, Hou YY, Lu LW, Fan LM. Abnormality of bone marrow-derived mesenchymal stem cells in patients with systemic lupus erythematosus. Lupus. 2007;16:121–8.

    Article  CAS  PubMed  Google Scholar 

  79. Griffin M, Ryan CM, Pathan O, Abraham D, Denton CP, Butler PE. Characteristics of human adipose derived stem cells in scleroderma in comparison to sex and age matched normal controls: implications for regenerative medicine. Stem Cell Res Ther. 2017;8:23.

    Article  PubMed  PubMed Central  Google Scholar 

  80. Kim DS, Jang IK, Lee MW, Ko YJ, Lee DH, Lee JW, et al. Enhanced immunosuppressive properties of human mesenchymal stem cells primed by interferon-γ. EBioMedicine. 2018;28:261–73.

    Article  PubMed  PubMed Central  Google Scholar 

  81. Zolfaghari S, Milan PB, Dehpour AR, Fomeshi MR, Eskandari F, Ebrahimi L, et al. The effect of poly I: C or LPS priming on the therapeutic efficacy of mesenchymal stem cells in an adjuvant-induced arthritis rat model. Pharmacol Rep. 2022;74:654–68.

    Article  CAS  PubMed  Google Scholar 

  82. Jang SG, Lee J, Hong SM, Kwok SK, Cho ML, Park SH. Metformin enhances the immunomodulatory potential of adipose-derived mesenchymal stem cells through STAT1 in an animal model of lupus. Rheumatology (Oxford). 2020;59:1426–38.

    Article  CAS  PubMed  Google Scholar 

  83. Casu B, Naggi A, Torri G. Re-visiting the structure of heparin. Carbohydr Res. 2015;403:60–8.

    Article  CAS  PubMed  Google Scholar 

  84. Lane DA, Adams L. Non-anticoagulant uses of heparin. N Engl J Med. 1993;329:129–30.

    Article  CAS  PubMed  Google Scholar 

  85. Zioncheck TF, Richardson L, Liu J, Chang L, King KL, Bennett GL, et al. Sulfated oligosaccharides promote hepatocyte growth factor association and govern its mitogenic activity. J Biol Chem. 1995;270:16871–8.

    Article  CAS  PubMed  Google Scholar 

  86. Sasaki N, Okishio K, Ui-Tei K, Saigo K, Kinoshita-Toyoda A, Toyoda H, et al. Heparan sulfate regulates self-renewal and pluripotency of embryonic stem cells. J Biol Chem. 2008;283:3594–606.

    Article  CAS  PubMed  Google Scholar 

  87. Furue MK, Na J, Jackson JP, Okamoto T, Jones M, Baker D, et al. Heparin promotes the growth of human embryonic stem cells in a defined serum-free medium. Proc Natl Acad Sci U S A. 2008;105:13409–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Ling L, Dombrowski C, Foong KM, Haupt LM, Stein GS, Nurcombe V, et al. Synergism between Wnt3a and heparin enhances osteogenesis via a phosphoinositide 3-kinase/Akt/RUNX2 pathway. J Biol Chem. 2010;285:26233–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Matsumoto K, Tajima H, Okazaki H, Nakamura T. Heparin as an inducer of hepatocyte growth factor. J Biochem. 1993;114:820–6.

    Article  CAS  PubMed  Google Scholar 

  90. Saito T, Kotani T, Suzuki K. Antifibrotic therapy by sustained release of low molecular weight heparin from poly(lactic-co-glycolic acid) microparticles on bleomycin-induced pulmonary fibrosis in mice. Sci Rep. 2020;10:19019.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Suzuka T, Kotani T, Saito T, Matsuda S, Sato T, Takeuchi T. Therapeutic effects of adipose-derived mesenchymal stem/stromal cells with enhanced migration ability and hepatocyte growth factor secretion by low-molecular-weight heparin treatment in bleomycin-induced mouse models of systemic sclerosis. Arthritis Res Ther. 2022;24:228.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Saito T, Kotani T, Suzuka T, Matsuda S, Takeuchi T, Sato T. Adipose-derived stem/stromal cells with heparin-enhanced anti-inflammatory and antifibrotic effects mitigate induced pulmonary fibrosis in mice. Biochem Biophys Res Commun. 2022;629:135–41.

    Article  CAS  PubMed  Google Scholar 

  93. Hwang JW, Myeong SH, Lee NH, Kim H, Son HJ, Chang JW, et al. Immunosuppressant drugs mitigate immune responses generated by human mesenchymal stem cells transplanted into the mouse parenchyma. Cell Transplant. 2021;30:9636897211019024.

    Article  PubMed  Google Scholar 

  94. Norte-Muñoz M, Gallego-Ortega A, Lucas-Ruiz F, González-Riquelme MJ, Changa-Espinoza YI, Galindo-Romero C, et al. Immune recognition of syngeneic, allogeneic and xenogeneic stromal cell transplants in healthy retinas. Stem Cell Res Ther. 2022;13:430.

    Article  PubMed  PubMed Central  Google Scholar 

  95. Hwang JW, Lee NK, Yang JH, Son HJ, Bang SI, Chang JW, et al. A comparison of immune responses exerted following syngeneic, allogeneic, and xenogeneic transplantation of mesenchymal stem cells into the mouse brain. Int J Mol Sci. 2020;21:3052.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Diehl R, Ferrara F, Müller C, Dreyer AY, McLeod DD, Fricke S, et al. Immunosuppression for in vivo research: state-of-the-art protocols and experimental approaches. Cell Mol Immunol. 2017;14:146–79.

    Article  CAS  PubMed  Google Scholar 

  97. Wang L, Wang L, Cong X, Liu G, Zhou J, Bai B, et al. Human umbilical cord mesenchymal stem cell therapy for patients with active rheumatoid arthritis: safety and efficacy. Stem Cells Dev. 2013;22:3192–202.

    Article  CAS  PubMed  Google Scholar 

  98. Park EH, Lim HS, Lee S, Roh K, Seo KW, Kang KS, et al. 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 

  99. Ghoryani M, Shariati-Sarabi Z, Tavakkol-Afshari J, Mohammadi M. The sufficient immunoregulatory effect of autologous bone marrow-derived mesenchymal stem cell transplantation on regulatory T cells in patients with refractory rheumatoid arthritis. J Immunol Res. 2020;2020:3562753.

    Article  PubMed  PubMed Central  Google Scholar 

  100. Shadmanfar S, Labibzadeh N, Emadedin M, Jaroughi N, Azimian V, Mardpour S, et al. 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 

  101. Wang L, Huang S, Li S, Li M, Shi J, Bai W, 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;13:4331–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Ghoryani M, Shariati-Sarabi Z, Tavakkol-Afshari J, Ghasemi A, Poursamimi J, Mohammadi M. Amelioration of clinical symptoms of patients with refractory rheumatoid arthritis following treatment with autologous bone marrow-derived mesenchymal stem cells: a successful clinical trial in Iran. Biomed Pharmacother. 2019;109:1834–40.

    Article  CAS  PubMed  Google Scholar 

  103. Álvaro-Gracia JM, Jover JA, García-Vicuña R, Carreño L, Alonso A, Marsal S, et al. 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 

  104. Liang J, Zhang H, Hua B, Wang H, Lu L, Shi S, et al. 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 

  105. Sun L, Wang D, Liang J, Zhang H, Feng X, Wang H, et al. Umbilical cord mesenchymal stem cell transplantation in severe and refractory systemic lupus erythematosus. Arthritis Rheum. 2010;62:2467–75.

    Article  CAS  PubMed  Google Scholar 

  106. Wang D, Li J, Zhang Y, Zhang M, Chen J, Li X, et al. Umbilical cord mesenchymal stem cell transplantation in active and refractory systemic lupus erythematosus: a multicenter clinical study. Arthritis Res Ther. 2014;16:R79.

    Article  PubMed  PubMed Central  Google Scholar 

  107. Deng D, Zhang P, Guo Y, Lim TO. A randomised double-blind, placebo-controlled trial of allogeneic umbilical cord-derived mesenchymal stem cell for lupus nephritis. Ann Rheum Dis. 2017;76:1436–9.

    Article  CAS  PubMed  Google Scholar 

  108. Yuan X, Qin X, Wang D, Zhang Z, Tang X, Gao X, et al. Mesenchymal stem cell therapy induces FLT3L and CD1c+ dendritic cells in systemic lupus erythematosus patients. Nat Commun. 2019;10:2498.

    Article  PubMed  PubMed Central  Google Scholar 

  109. Chen C, Liang J, Yao G, Chen H, Shi B, Zhang Z, et al. Mesenchymal stem cells upregulate Treg cells via sHLA-G in SLE patients. Int Immunopharmacol. 2017;44:234–41.

    Article  CAS  PubMed  Google Scholar 

  110. Wang Y, Ma D, Wu Z, Yang B, Li R, Zhao X, et al. Clinical application of mesenchymal stem cells in rheumatic diseases. Stem Cell Res Ther. 2021;12:567.

    Article  PubMed  PubMed Central  Google Scholar 

  111. Wong SC, Medrano LC, Hoftman AD, Jones OY, McCurdy DK. Uncharted waters: mesenchymal stem cell treatment for pediatric refractory rheumatic diseases; a single center case series. Pediatr Rheumatol Online J. 2021;19:87.

    Article  PubMed  PubMed Central  Google Scholar 

  112. Almadori A, Griffin M, Ryan CM, Hunt DF, Hansen E, Kumar R, et al. Stem cell enriched lipotransfer reverses the effects of fibrosis in systemic sclerosis. PLoS One. 2019;14:e0218068.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Blezien O, D’Andrea F, Nicoletti GF, Ferraro GA. Effects of fat grafting containing stem cells in microstomia and microcheilia derived from systemic sclerosis. Aesthetic Plast Surg. 2017;41:839–44.

    Article  PubMed  Google Scholar 

  114. Virzì F, Bianca P, Giammona A, Apuzzo T, Di Franco S, Mangiapane LR, et al. Combined platelet-rich plasma and lipofilling treatment provides great improvement in facial skin-induced lesion regeneration for scleroderma patients. Stem Cell Res Ther. 2017;8:236.

    Article  PubMed  PubMed Central  Google Scholar 

  115. Granel B, Daumas A, Jouve E, Harlé JR, Nguyen PS, Chabannon C, et al. Safety, tolerability and potential efficacy of injection of autologous adipose-derived stromal vascular fraction in the fingers of patients with systemic sclerosis: an open-label phase I trial. Ann Rheum Dis. 2015;74:2175–82.

    Article  CAS  PubMed  Google Scholar 

  116. Keyszer G, Christopeit M, Fick S, Schendel M, Taute BM, Behre G, et al. Treatment of severe progressive systemic sclerosis with transplantation of mesenchymal stromal cells from allogeneic related donors: report of five cases. Arthritis Rheum. 2011;63:2540–2.

    Article  PubMed  Google Scholar 

  117. Park Y, Lee YJ, Koh JH, Lee J, Min HK, Kim MY, et al. Clinical efficacy and safety of injection of stromal vascular fraction derived from autologous adipose tissues in systemic sclerosis patients with hand disability: a proof-of-concept trial. J Clin Med. 2020;9:3023.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Scuderi N, Ceccarelli S, Onesti MG, Fioramonti P, Guidi C, Romano F, et al. Human adipose-derived stromal cells for cell-based therapies in the treatment of systemic sclerosis. Cell Transplant. 2013;22:779–95.

    Article  PubMed  Google Scholar 

  119. Zhang H, Liang J, Tang X, Wang D, Feng X, Wang F, et al. Sustained benefit from combined plasmapheresis and allogeneic mesenchymal stem cells transplantation therapy in systemic sclerosis. Arthritis Res Ther. 2017;19:165.

    Article  PubMed  PubMed Central  Google Scholar 

  120. Cui J, Jin L, Ding M, He J, Yang L, Cui S, et al. Efficacy and safety of mesenchymal stem cells in the treatment of systemic sclerosis: a systematic review and meta-analysis. Stem Cell Res Ther. 2022;13:118.

    Article  PubMed  PubMed Central  Google Scholar 

  121. Antoniou KM, Karagiannis K, Tsitoura E, Bibaki E, Lasithiotaki I, Proklou A, et al. Clinical applications of mesenchymal stem cells in chronic lung diseases. Biomed Rep. 2018;8:314–8.

    CAS  PubMed  PubMed Central  Google Scholar 

  122. Chambers DC, Enever D, Lawrence S, Sturm MJ, Herrmann R, Yerkovich S, et al. Mesenchymal stromal cell therapy for chronic lung allograft dysfunction: results of a first-in-man study. Stem Cells Transl Med. 2017;6:1152–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Tzouvelekis A, Paspaliaris V, Koliakos G, Ntolios P, Bouros E, Oikonomou A, Zissimopoulos A, Boussios N, Dardzinski B, Gritzalis D, Antoniadis A, Froudarakis M, et al. A prospective, non-randomized, no placebo-controlled, phase Ib clinical trial to study the safety of the adipose derived stromal cells-stromal vascular fraction in idiopathic pulmonary fibrosis. J Transl Med. 2013;11:171.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank and extend our appreciation to all staff members of the Osaka Medical and Pharmaceutical University, Division of Research Equipment and Devices, for their assistance and support. We also thank Naomi Takahashi and Kaori Suzuki for their technical assistance. We would like to thank Editage (www.editage.jp) for English language editing.

Funding

Our work shown in this review was supported by grants as follows: Grant-in-Aid for Young Scientists (B; 16K19470, to T. K.); Grant-in-Aid for Young Scientists (21K16312, to T. S.); and Grant-in-Aid for Young Scientists (B; 17K16216, to Y. W.).

Author information

Authors and Affiliations

Authors

Contributions

TK wrote the manuscript. TS prepared Table 4 and Fig. 2. All authors contributed to the data collection and the preparation of our research papers cited in this review. TK and TS revised the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Takuya Kotani.

Ethics declarations

Ethics approval and consent to participate

The Institutional Animal Care and Use Committee of Osaka Medical and Pharmaceutical University approved all research protocols (approval ID: AM23-010), including surgical procedures and animal care. All experiments were performed in accordance with relevant guidelines and regulations.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

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

Rights and permissions

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

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kotani, T., Saito, T., Suzuka, T. et al. Adipose-derived mesenchymal stem cell therapy for connective tissue diseases and complications. Inflamm Regener 44, 35 (2024). https://doi.org/10.1186/s41232-024-00348-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s41232-024-00348-z

Keywords