Next Article in Journal
Alpha Mangostin and Cisplatin as Modulators of Exosomal Interaction of Ovarian Cancer Cell with Fibroblasts
Next Article in Special Issue
Presume Why Probiotics May Not Provide Protection in Inflammatory Bowel Disease through an Azoxymethane and Dextran Sodium Sulfate Murine Model
Previous Article in Journal
Externalization of Mitochondrial PDCE2 on Irradiated Endothelium as a Target for Radiation-Guided Drug Delivery and Precision Thrombosis of Pathological Vasculature
Previous Article in Special Issue
A Preclinical Validation of Iron Oxide Nanoparticles for Treatment of Perianal Fistulizing Crohn’s Disease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Mesenchymal (Stem) Stromal Cells Based as New Therapeutic Alternative in Inflammatory Bowel Disease: Basic Mechanisms, Experimental and Clinical Evidence, and Challenges

by
Noemi Eiro
1,*,
Maria Fraile
1,
Alberto González-Jubete
1,
Luis O. González
2 and
Francisco J. Vizoso
1,3,*
1
Research Unit, Fundación Hospital de Jove, Av. de Eduardo Castro, 161, 33290 Gijón, Spain
2
Department of Anatomical Pathology, Fundación Hospital de Jove, Av. de Eduardo Castro, 161, 33290 Gijón, Spain
3
Department of Surgery, Fundación Hospital de Jove, Av. de Eduardo Castro, 161, 33290 Gijón, Spain
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(16), 8905; https://doi.org/10.3390/ijms23168905
Submission received: 6 July 2022 / Revised: 26 July 2022 / Accepted: 26 July 2022 / Published: 10 August 2022
(This article belongs to the Special Issue Pathophysiology and Treatment of Inflammatory Bowel Disease)

Abstract

:
Inflammatory bowel diseases (IBD) are an example of chronic diseases affecting 40% of the population, which involved tissue damage and an inflammatory process not satisfactorily controlled with current therapies. Data suggest that mesenchymal stem cells (MSC) may be a therapeutic option for these processes, and especially for IBD, due to their multifactorial approaches such as anti-inflammatory, anti-oxidative stress, anti-apoptotic, anti-fibrotic, regenerative, angiogenic, anti-tumor, or anti-microbial. However, MSC therapy is associated with important limitations as safety issues, handling difficulties for therapeutic purposes, and high economic cost. MSC-derived secretome products (conditioned medium or extracellular vesicles) are therefore a therapeutic option in IBD as they exhibit similar effects to their parent cells and avoid the issues of cell therapy. In this review, we proposed further studies to choose the ideal tissue source of MSC to treat IBD, the implementation of new standardized production strategies, quality controls and the integration of other technologies, such as hydrogels, which may improve the therapeutic effects of derived-MSC secretome products in IBD.

1. Introduction

Inflammatory bowel disease (IBD) encompasses principally two disorders defined by a chronic inflammation of the gastrointestinal tract: ulcerative colitis (UC) and Crohn’s disease (CD), affecting 1.4 million Americans and 2.4 million Europeans [1]. The incidence and prevalence of IBD, which can lead to life-threatening problems, have increased over the last decades, and is thus a growing public health concern [2,3,4].
IBD has a multifactorial etiology implicating environmental factors, microbiota imbalance, genetic predisposition and mucosa immune defects [5,6]. In the intestine, response to the pathogens produces a controlled inflammation that continues until the pathogen is eradicated; this response is excessive in the IBD [7]. In this scenario, a dysregulate immune response to commensal antigens in genetically susceptible people led to an inflammatory environment characterized by high expression of pro-inflammatory molecular factors such as TLRs, TNF-α, IL-1β and INF-γ, which globally produce both high proliferation and low apoptosis rates of T cells, as well as their differentiation to pro-inflammatory T cells subsets such as dendritic cells, T helper type 1 and M1 macrophages, whereas regulatory T cells (Treg) are down-regulated (Figure 1). The immune imbalance between Treg and Th17 contribute to the progression of colitis [8], since both subset of cellular immune response are interconnected. Th17 express IL-17, a pro-inflammatory interleukin that contributes to tissue inflammation and damage. IL-17 plays a key role in the recruitment and activation of granulocytes and induces the release of pro-inflammatory mediators (IL-1β, TNF-α, IL-6, MMPs, among others) from macrophages leading to exacerbated inflammation [9]. A failure in the regulatory capacity of Treg worsens inflammation. However, Treg inhibit the inflammation through the suppression of effector T cells proliferation and the production of IL-10 and TGF-β [10].
Especially relevant is also the increased polarization of macrophages to the activated pro-inflammatory M1 phenotype which produce high pro-inflammatory cytokines, reactive nitrogen and oxygen species [11]. Taken together, led to an inflammatory cytokine cascade (mainly high levels of TNF-α IL-1β, IL-6, MMPs and nitric oxide) which exacerbate the inflammation [9] by increasing migration of leukocytes and provoking oedema, granuloma formation, ulceration and mucosa degradation, an attack of commensal microbiota resulting in a severe tissue damage and a fibrotic reaction.
Currently, the treatment for IBD starts with 5-ASA agents and antibiotics, depending on the response patients are treated with corticosteroids, immunomodulators and/or biologic agents, but when all this fails, patients require surgery [12]. Such multifactorial factors implicated are obviously difficult to target by using a conventional therapy strategy, which are not completely effective. In addition, all these current therapies are generally noted to have several side effects related to the gastrointestinal tract and liver and severe immune suppressive side effects that impact on lifestyle and predispose individuals to opportunistic infection [13].
For all this, development of more effective treatment for IBD patients, which preserves immune homeostasis and restores the gut barrier, remains to be a challenge [14,15,16,17].

2. Stem Cell-Based Therapies in IBD

Cell therapy is defined as the introduction of cells into an organism or tissue in order to treat a disease. In this context, stem cell therapies have significant potential to lead therapeutic development [18]. Stem cells, defined as undifferentiated cells with the capability of differentiate into specific cells of tissues or organs, are characterized as embryonic stem cells, induced pluripotent stem cells (iPSC) or somatic (adult) stem cells, according to their sources. Embryonic stem cells and iPSC are pluripotent cells with the highest multi-differentiation potential; nonetheless, due to ethical concerns and the oncogenic potential, respectively, their clinical use is limited. Somatic stem cells, which are found in many organs and tissues from adults, do not raise special ethical concerns and do not require to be genetically reprogrammed and make them the most used cells for “stem cell-based therapy. Indeed, autologous (from the patient himself) or allogeneic (isolated from a donor) somatic stem cells, such as haematopoietic stem cells (HSC) or mesenchymal (stem) stromal cells (MSC), are used in most clinical trials for IBD.

2.1. Haematopoietic Stem Cells

Haematopoietic stem cells transplantation (HSCT) involves the collection and the reinfusion of HSC (from the patient or from a donor) to restore the lymphohematopoietic system [19]. The treatment of non-oncohaematological diseases with HSCT is based on evidences from animal studies and from some cases of remission observed in patients [20].
In the last decade, HSCT has been explore as a new therapy for severe autoimmune diseases, refractory to conventional therapy, including systemic lupus erythematosus and rheumatoid arthritis [21,22]; but also, in severe forms of gastrointestinal pathologies, such as refractory CD. Autologous HSCT to CD patients, with concomitant neoplasms, showed a high efficacy including one case with a remission up to 7 years [23,24,25], but there is the doubt if the maintenance of remission is due to the transplantation (for concomitant neoplasms) in an already quiescent intestinal disease. Later studies report significant remissions or significant symptomatic improvement with these therapies [26,27]. In addition, several aspects were criticized for their designs and unconventional endpoint [28], supporting the need for further controlled clinical trials. In addition, it has been hypothesized that through the plastic properties of stem cells, the self-reactive immune system is eliminated and replaced directly at the source (bone marrow), but the mechanisms of action of auto-HSCT in CD are not well known and understood. Data support that an immunomodulatory “resetting” effect occurs, leading to the restoration of a dynamic balance between self-reactivity and immunological tolerance, probably due to immunologically “naive” stem cells. In fact, after HSCT it was observed a significant decrease of TNF-α and IL-10 secretion and positive TLR-4 cells and an increase of T-regulatory (T-reg) [29].
Allogeneic HSCT seems to reset the immune response and impact on the modification of genetic predisposition to the disease, since HSC with polymorphisms causing the disease are replaced by HSC from a healthy donor [30]. Early reports of allo-HSCT, usually retrospective studies carried out on CD patients with concomitant haematological malignancies, showed the achievement and maintenance of long-term clinical remission (up to 15 years) (Revised by [31]). However, studies focused on IBD patients do not show such a favorable benefit/risk ratio (Revised by [31]). As well, compared with auto-HSCT, allo-HSCT is associated with a higher morbidity and mortality which limits its implementation as a treatment of IBD. In allo-HSCT, despite donor/recipient HLA-matching, both are immunologically different; therefore, after HSCT immune cells from the donor can generate a response against the recipient, called Graft Versus Host Disease (GVHD). This situation requires an adequate immunosuppressive prophylaxis to avoid transplant rejection, which is associated with significant morbidity and mortality [32,33,34]. Nucleotide-binding oligomerization domain 2 (NOD2) mutations or deletion are independent risk factors for CD and GVHD. Host NOD2 expression in the hematopoietic compartment protects against GVHD, as it has been evidenced in bone marrow transplantation in a mouse model [35] and regulates epithelial paracellular permeability [36]. Nevertheless, it has been described that NOD2 contributes to intestinal crypt survival in vivo [37], but when stimulated by muramyl dipeptide, NOD2 in the non-hematopoietic compartment normalized the permeability [36]. So, NOD2 expression in hematopoietic or non-hematopoietic compartment could exhibit a protective role GVHD [38] and regulate intestinal barrier function.
On the other hand, none studies regarding HSCT as a treatment for UC have been published, only some reports when UC patients undergoing HSCT for other indications [31]. Maybe it is due to the identification of UC, but not CD, as an independent risk factor for morbidity and mortality [34].
MSC therapy is considered an interesting therapeutic option for IBD since MSC have low immunogenicity, allowing MSC to escape recognition by the immune system due to low expression of HLA class I and no expression of HLA class II and co-stimulatory molecules (CD40, CD40L, CD80 and CD86) [39,40,41,42].

2.2. Mesenchymal (Stem) Stromal Cells

About 50 years ago, MSC were described as a “colony forming unit-fibroblast” from rodent’s bone marrow [43]. MSC can be found in several locations such as bone marrow (BM), adipose tissue (AT), umbilical cord (UC), Wharton’s jelly (WJ), amniotic fluid, placenta, uterus, dermis, skeletal muscle, periosteum, periodontal ligament dental pulp or peripheral blood, among others [44]. In 2006, the “International Society for Cellular Therapy” defined MSC as follows: “(i) MSC must display plastic-adherent capacities; (ii) a simultaneous expression of stromal markers (CD29, CD44, CD73, CD90 and CD105), but the absence of hematopoietic (CD45 and CD14) or endothelial (CD31 and CD34) markers and HLA-DR surface molecules and (iii) an in vitro differentiation potential into osteoblasts, adipocytes and chondroblasts” [45]. MSC are attractive for cell therapies due to their ability to spontaneously migrate to the injured region when the body is damaged, which is named homing effect.
Till the date, over 1000 phase I, II and III clinical trials have been carried out in several pathologies, such as autoimmune and inflammatory diseases [46]. MSC can interact with immune cells and secrete factors, resulting in an immunomodulation. Thus, a change in concept of the therapeutic utility of MSC occurred, from the primarily regenerative effect to the anti-inflammatory and, more recently, to other relevant ones such as angiogenic, anti-oxidative stress, anti-tumoral or anti-microbial [47].
MSC participate and control tissue renewal through their function as sentinels and regulators in tissue homeostasis. Through their ability to regulate oxidative stress, cell proliferation, angiogenesis or apoptosis, MSC influence the tissue microenvironment using paracrine signals, secreting soluble bioactive molecules and/or extracellular vesicles (EVs). The depletion or poor function of MSC has been implicated in several degenerative or autoimmune diseases (such as rheumatoid arthritis [48], systemic lupus erythematosus [49], diabetes mellitus [50] or psoriasis [51]) and in processes associated with aging [52,53]. Consequently, the use of allogenic MSC to restore physiological function has motivated several investigations around the world [18].

2.2.1. MSC and Intestinal Homeostasis

In the intestinal tract, epithelial cells and intestinal mesenchymal cells (iMC) create a physical barrier dividing the microbiome from the underlying tissues and immune system [54]. iMC, a heterogeneous cell population consisting of MSC, myofibroblasts, fibroblasts and pericytes, are present in the subepithelial stroma where they supply a large part of the intestinal structure. More importantly, iMC regulate homeostasis and maintenance of the epithelium barrier [54,55] and foster immunologic tolerance against the commensal bacteria and dietary antigens by the expression of immune-modulating molecules, including programmed death-ligand-1 (PD-L1) [56], chemokines and retinoic acid [57]. As well, recent studies have suggested that iMC play an important role in inhibiting and promoting intestinal inflammation and IBD damage, which can partly, by TGF-β production, contribute to the homeostasis of intestinal epithelium associated with wound healing and mucosal integrity. Furthermore, TGF-β regulates the adhesion molecules and tight junction molecules’ expression and thus adjusts the barrier function of the intestinal epithelium [58]. In this context, iMC isolated from human resected intestine, exhibiting a fibroblast-like morphology and lacking haemopoietic, epithelial and endothelial markers, have showed, in a recent ground-breaking study, to have an immunomodulatory effect and their contribution to epithelial integrity preservation in vitro and in vivo [59]. Remarkably, preconditioning of iMC with TNF and IFN-γ improved their immunosuppressive ability, through indoleamine 2,3-dioxygenase (IDO-1) pathway indicating its role in iMC activity improvement. Moreover, iMC conditioned media enhanced wound healing in vitro. Besides, data show that human iMC isolated from un-inflamed intestine possess tissue-regenerative and immunomodulatory capabilities that could potentially be promoted/restored in order to reduce IBD disease severity. Thus, these data suggest that an impoverishment or dysfunction of iMC and more precisely, MSC may contribute to the pathophysiology of IBD.

2.2.2. MSC and Gut Microbiota

The gastrointestinal tract harbors a complex and dynamic population of microorganisms, known as the gut microbiota, which influence the host during homeostasis and disease, since the microbiota plays a key role in maintaining immune and metabolic homeostasis. The alteration of the gut microbiota, called dysbiosis, is associated with several inflammatory diseases such as IBD [60] and the identification of the functional and integrative map of the microbiota has been proposed to be used as a diagnostic, prognostic and therapeutic tools [61].
Considering that dysbiosis coupled with immune dysregulation could lead to IBD, MSC therapy and microbiota focused therapy like fecal microbiota transplantation (FMT) gain much interest and emerge as a novel approach. Common therapeutic points of both FMT and MSC-therapy in IBD is based on their main functional effects: immunoregulation, tissue damage repair and remodeling, and gut microbiota restoration [62].
It has been evidenced that the crosstalk between MSC and gut microbiota improves the function of each one. Indeed, MSC can promote microbiota normalization in AOM/DSS mice [63], while the microbiota can enhance the immunomodulation capacity of BM-MSC in a DSS mouse model [64]. Hence the need to explore further this approach and its possible mechanisms trough NOD2 [65,66] or EVs [67].

3. Therapeutic Potential of MSC in IBD

According to the above hypothesis, many experimental animal studies indicate that the transplantation of MSC has therapeutic potential for IBD. In addition, MSC therapy have also emerged as therapeutic option for refractory IBD patients.
The importance of MSC capabilities for their efficacy on experimental colitis and IBD continues to be studied (Table 1). Several studies shown that MSC ameliorate experimental colitis endpoints, such as body weight changes, bleeding, stool consistency, mortality rate, colon length, colonic inflammation or inflammatory cytokines expression [68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88]. MSC from different sources were used in those studies, such as adipose tissue-derived MSC (AT-MSC) [77,78,81,85,89,90], bone marrow-derived MSC (BM-MSC) [68,69,70,74,82,91], umbilical cord blood-derived MSC (UCB-MSC) [71,72,79,92], amnion derived MSC, endometrial derived MSC (E-MSC) [86], tonsil derived MSC, or MSC derived from induced pluripotent stem cells (iPSC) equivalent to AT-MSC [80]. Moreover, MSC were applied by different methods, such as intravenous injection [68,69,70,74,80,82,86,87,93], intraperitoneally administration [69,72,73,75,76,77,79,83,84,85,89,91], mesenteric injection [78] or local injection [69,81,87,88,90,91]. Furthermore, intravenous administration of canine AT-MSC to dogs with IBD induce an improvement of the disease severity and allow to progressively reduce until to suppress steroid dosage [94,95].
A recent meta-analysis, which assessed 46 studies, 28 of which were animal studies (n = 567), showed that the disease activity index decreased significantly in MSC treatment groups compared to the control group [96].
Table 1. Experimental in vivo studies based on MSC-therapy in IBD.
Table 1. Experimental in vivo studies based on MSC-therapy in IBD.
First
Author/Year
ExperimentalCell SurgeAdministration RouteDose of ProductTimeMore Relevant Results
Barnhoorn (2020) [97]DSS-induced colitisBM-MSCIP2 × 106 cells11 daysMSC after in vivo aggregation show a favorable RNA expression profile for the treatment of colitis. MSC spheroids showed high expression of Ki-67 and low levels of apoptotic marker cleaved caspase-3. Locally applied MSC and MSC spheroids are both able to ameliorate DSS-induced colitis and show similar clinical effects, including improvement in the macro and microscopic IBD score.
Barnhoorn (2020) [97]DSS-induced colitisBM-MSCEndoscopic2 × 106 cells4–6 daysEndoscopic injection can be a feasible and effective novel application route for MSC therapy in patients with luminal IBD.
Chao (2016) [71]TNBS-induced colitisUC-MSCIP10 × 106 cells14 daysThe mortality in UC-MSC-treated TNBS mice was 20% (55% in colitis model); the treatment reduced the inflammation of the transmural area, depletion of epithelial cells and focal loss of crypts. IL-20 and TGF-Beta were significantly higher in UC-MSC-treated mice (p = 0.04 and 0.02 respectively).
Cheng (2017) [82]DSS-induced colitisBM-MSC/IL-25-BM-MSCIV5 × 106 cells8 daysIL-25-MSC treatment significantly attenuate the colon shortening (12 ± 0.62 cm); IL-25 could enhance immunomodulatory ability of MSC via inhibiting Th17 immune response and promoting the regulation of Tregs cells. However, the study failed to confirm that IL-25 affected the migratory and regenerative capacities of MSC in vivo.
de Aguiar (2018) [84]DSS-induced colitisAT-MSCIP10 × 106 cells7 daysADMSC-treated mice did not present severe reduction in colon length, and presented a reduced tissue damage score index (3); less detachment of mucosa and submucosa layers, low villous blunting and partial preservation of crypt and epithelial integrity. The level of ZO-1 expression in the colon was re-established in ADMSC-treated mice. Significant reduction of IFN-gamma and TNF-Alpha, and reduction of IL-6 and MCP-1 protein levels. ADMSC treatment reduced DCs and macrophages presence in the colon.
de Aguiar (2018) [84]DSS-induced colitisAT-MSCIP2 × 106 cells7 daysDMSC ameliorated the severity of DSS-induced colitis, reducing colitis pathological score and preventing colon shortening.
de la Portilla (2013) [98]TNBS-induced colitisAT-MSCLocal60 × 106 cells24 weeksFirst study which shows the homing migration of MSC to areas of experimentally-induced colitis following rectal installation.
de la Portilla (2018) [90]TNBS-induced colitisAT-MSCLocal2 × 106 cells10 daysThere were no differences in component rectal wall thicknesses with a higher Hunter score in the treated group compared with the controls.
Fu (2017) [78]TNBS-induced colitisAT-MSCMesenteric injection2 × 106 cells6 daysDcreased the weight loss and DAI score, MPO activity; moreover relieved colitis, decreased colonic shortening, inflammatory cell infiltration and mucosal ulceration. Reduced levels of ROR(lamda)t and IL-17A; inhibited STAT3 phosphorylation, but increased STAT5 phosphorylation.
González-Rey (2009) [77]DSS-induced chronic colitisAT-MSCIP10 × 106 cells27 daysAT-MSC treatment protects against DSS-induced acute colitis as well as chronic severe colitis (p 0.01 and p 0.001 respectively); reduces colonic inflammatory responses in DSS-induced chronic colitis (p 0.001).
Gregoire (2018) [99]Fistulising Crohn’s diseaseAT-MSCSingle intrafistular injection3 × 106−30 × 106 cells8 weeks6/8 fistulas healed, 2/8 improved.
Heidari (2021) [100]DSS-induced colitisAT-MSC/MSC-CMIP10 × 106 cells34 daysThere was no significant difference in the survival rate among the study groups; however, there was a significant increase in terms of the colon length (p 0.005). In the treated mice the level of mucosal damage was significantly lower (p 0.005), and the structure of the crypts also showed improvement of tissue healing.
Heidari (2021) [100]DSS-induced colitisAT-MSCIP2 × 106 cells34 dayThe regulatory effects of AT-MSC and their CM in inflammatory conditions because of colitis
In Kap (2010) [93]DSS-induced colitisMSCIV1 × 106 cells7 daysAnti-addressin Ab coating on MSC increased cell delivery to inflamed colon and increased the efficacy of MSC treatment of IBD.
Jianxia Hu (2016) [101]Luminal Crohn’s diseaseUCB-MSCIV0.5 × 106 cells3 month30/36 patients showed good response and diffuse and deep ulcer formation and severe inflammatory mucosa were improved markedly.
Lee (2016) [70]DSS-induced colitisBM-MSCIV10 × 106 cells33 daysIL-10 production was upregulated by about 10-fold in BM-MSC-treated mice and showed a preventive effect on weight loss.
Lee (2016) [70]DSS-induced colitisBM-MSCIV30 × 106 cells33 daysInfusion of BM-MSC at the onset of disease exerted preventive and rapid recovery effects.
Lee (2018) [79]DSS-induced colitisUCB-MSCIP2 × 106 cells12 daysThe survival rate was further increased by co-treatment compared to UCB-MSC or MIS416 single treatments; colon lengths were significantly increased in co-treatment; colonic inflammation was more effectively resolved by co-treatment with MSI416 and UCB-MSC, and only co-treatment markedly decreased fibrosis and enhanced tissue regeneration. Exposure to MIS416 increases the number of immune cells via activation of CD14+ macrophages.
Legaki (2016) [83] DSS-induced colitisAmniotic fluid-MSCIP1.5 × 106 cells, 200 μL/dose7 daysCM treatment significantly decreased the extension and severity of the inflammation in comparison to the DSS-treated mice; the relative expression levels of IL-10 mRNA were significantly increased, similarly TNF-a and IL-1B levels were decreased at mRNA level. Additionally, TGFb1 was significantly higher (p 0.0001).
Mao (2017) [102]DSS-induced colitisUCB-MSCIV400 μg
exosomes/1.3 × 106 cells
11 daysExosomes from MSC have profound effects on alleviating DSS-induced IBD and may exert their impact through the modulation of IL-7 expression in macrophages.
Martín (2018) [103]TNBS-induced colitisAT-MSCLocal10 × 106 cells11 daysSubmucosal injection of human ASCs ameliorates the course of TNBS colitis in immunocompetent rats.
Martin Arranz (2018) [81]TNBS-induced colitisAT-MSCEndoscopic10 × 106 cells11 daysThe endoscopic score improved in the ASC group by 47.1% ± 5.3% vs. 21.8% ± 6.6% in the vehicle group.
Miyamoto (2017) [87]TNBS-induced colitisAT-MSCIV and Local1 × 106 cells IV and 400 μL Local7 dayshAMSC transplantation significantly decreased the number of neutrophils, attenuated acute inflammation, suppressed the expression levels of inflammatory mediators in the colons; in the TNBS-CM gel group ulcers were shallow and bleeding was not detected, therefore improved endoscopic score. In the gel group mRNA expression levels of TNF-Alpha, CXCL1, CCL2 and IL-6 were increased.
Molendijk (2015) [104]Fistulising Crohn’s diseaseBM-MSCSingle intrafistular injection10, 30, 90 × 106 cells6, 12, 24 weeksAt week twelve, 3 of 9 individual fistulas had healed in group 1 (33.3%), 6 of 7 had healed in group 2 (85.7%), 2 of 7 had healed in group 3 (28.6%), and 3 of 9 had healed in the placebo group (33.3%).
Pak (2018) [88]DSS-induced colitisBM-MSC/AT-MSCEndoscopic8 × 105 cells/1.1 × 106 cells1–3 daysThe success rate was 37.60% for AT-MSC group and 35.20% for BM-MSC group.
Panés (2016) [105]Fistulising Crohn’s diseaseAT-MSCSingle intrafistular injection120 × 106 cells24 weeksRemission in the ITT (53 of 107 [50%] vs. 36 of 105 [34%]; difference 15.2%, 97.5% CI 0.2–30.3; p = 0·024) C × 601 vs. placebo.
Panés (2018) [106]Fistulising Crohn’s diseaseAT-MSCSingle intrafistular injection120 × 106 cells52 weeksC × 601 achieved combined remission (56.3%) vs. controls (38.6%) (a difference of 17.7 percentage points; 95% CI 4.2–31.2; p = 0.010).
Park (2018) [107]DSS-induced chronic colitisAT-MSCIP10 × 106 cells20 daysIn DSS-induced chronic colitis model, hASCs decreased the frequency of macrophage transition, specially M1 macrophages. The results suggest that PGE2, produced by co-culture of ASCs and THP-1, reduces M1 population.
Park (2018) [107]DSS-induced colitisAT-MSCIP2 × 106 cells20 dayASCs can suppress the inflammatory response by controlling the macrophage population, and ASCs may be therapeutically useful for the treatment of IBD.
Pouya (2018) [75]DSS-induced colitisMSC-CMIP500 μL, ×310 daysAfter infusion, colon inflammation was reduced and histopathological analysis showed a decrease in mucosal degeneration.
Song (2017) [72]DSS-induced acute/chronic colitisMSC-Ex/UC-MSCIP10 × 106 cells; 150 μg/mouse36 daysMSC-Ex ameliorates the clinical parameters in DSS-induced colitis; the treated group showed significantly less MPO activity. The level of IL-17 was significantly decreased, whereas those of IL-10 and TGF-Beta1 were increased. MSC-Ex is superior to UC-MSC in chronic IBD models, without differences in the colon length.
Song (2018) [85]DSS-induced colitiscanine AT-MSCIP2 × 106 + TSG-6 siRNA10 daysAT-MSC-secreted TSG-6 reduced inflammatory response and apoptosis in the colon; intraperitoneally infused cAT-MSC did not migrate to the inflamed colon; increased M2 macrophages in the inflamed colon.
Soontararak (2018) [80]DSS-induced colitisiMSC/AT-MSCIV3 × 106 cells19 daysThe clinical illness scores were significantly reduced (iMSC-treated p = 0.003, adMSC-treated p = 0.001); colonic tissues from mice treated with either iMSC or adMSC exhibited an overall reduction in transmural inflammation, with significantly less infiltration of inlammatory cells in the lamina propria, diminished mucosal ulceration and decreased mucosal collapse and granulation tissue formation.
Tanaka (2008) [68]DSS-induced colitisBM-MSCIV5 × 106 cells7 daysIn the rectum of treated rats the mRNA expression of TNF-alpha and IL-1Beta was markedly decreased to (43.7 ± 25.5% p 0.05 and 14.5 ± 12% p 0.01 respectively), as well as COX-2 16.5 ± 15.2% (p 0.01).
Tanaka (2008) [68]DSS-induced colitisMSCIV5 × 106 cells7 daysExogenous MSC accumulated in inflamed tissues and ameliorated DSS-induced colitis via a local anti-inflammatory action.
Wang (2016) [108]DSS-induced colitisBM-MSCIP0.5 × 106 cells10 daysIntraperitoneal injection is the best delivery way for MSC: showed better mucosa recovery and higher cell engraftment at inflamed colon.
Wu (2018) [109]DSS-induced colitisUCB-MSCIV400 μg UC-MSC11 daysExosomes from hucMSC have profound effects on alleviating DSS-induced IBD and may exert their function by regulating the ubiquitin modification level.
Xu (2018) [86]DSS-induced colitisEndometrial regenerative cells (ERC)IV3 × 10610 daysERC treatment significantly reduced the levels of TNF-Alpha, IL-1Beta and IL-6; ERCs downregulated the expanded Th1 and Th17 cells in colitis, and elevated the proportion of Tregs in lymphocytes; ERCs inhibited B-cell activation, differentiation and IgG production in colitis. ERC treatment enhanced the concentration of IL-10 in the colon and spleen, as well as CD1dhiCD5 + B cells in the spleen, peritoneal cavity and MLN.
Yu (2017) [73]DSS-induced colitisTonsil-MSCIP20 × 106/40 × 106 cells30 daysCo-culture with T-MSC clearly inhibited the PMA-stimulated proliferation of splenocytes by 60%; TMSC [×4] treated mice’s survival rate was improved to that of the normal. TMSC [×2] injection also significantly improved the survival rate to 89% of the control. TMSC [×4] treatment inhibits DSS-induced colon shortening; TMSC injection does no inhibit histopathological alterations in the distal colon in the chronic colitis mouse model, although it ameliorates IL-1Beta and IL-6 mRNA production in chronic colitis mice.
IP: Intraperitoneal; IV: intravenous.

3.1. Potential Therapeutic Mechanisms of MSC in IBD

3.1.1. Anti-Inflammatory Effects

MSC modulate immune adaptive cells from a high amount of T-cell effector to a regulatory T (Treg)-rich microenvironment, through paracrine factors like transforming growth factor beta (TGF-β) [110], hepatocyte growth factor (HGF) [111], prostaglandin E2 (PGE2) [112], nitric oxide (NO) [113], and indoleamine 2,3-dioxygenase (IDO) [114]. IL-10 is another MSC-generated immunoregulatory cytokine which contributes to bowel homeostasis, its deficiency aggravates DSS-mediated colitis whereas its supplementation could become an alternative treatment for IBD [115]. As a result of these soluble factors, MSC exert an anti-inflammatory effect on different types of immune cells. In this sense, it has been reported that MSC downregulate Th1 and Th17 responses and upregulate Th2 and Treg-mediated responses, which contribute to the improvement of colonic inflammation [116,117]. Also, MSC express NOD2, and its binding to muramyl dipeptide (MDP) ligand, enhances the secretion of anti-inflammatory factors such as PEG2 and IL-10, and stimulates the production of Tregs. In this regard, the anti-inflammatory effect of UC-MSC was enhanced by the activation of NOD2 through the activation of COX-2 signalling, which reduced disease severity in a mouse model of colitis [118]. Furthermore, co-administration of MIS416 (a microparticle that activates NOD2 and TLR9 signalling) and UC-MSCs improved therapeutic efficacy of MSC [79]. Additionally, there are other specific mechanisms associated with anti-inflammatory effects through EVs derived from MSC, as discussed below.

3.1.2. Regenerative Effects

MSC secrete several biological molecules such as cytokines, growth factors and lipids, which impact on tissue renewal [107]. Thus, paracrine signalling of MSC was proposed as the primary mechanism for their regenerative effect on parenchymal and mesenchymal cells [119], acting on processes implicated in tissue regeneration, like migration, immunomodulation and re-epithelization [47]. A correct tissue regeneration requires new blood vessels formation for the administration of oxygen, nutrients, and growth factor; in this sense, it was suggested that the induction of angiogenesis is another one of the principal mechanisms of action of MSC on tissue regeneration. Through the secretion of molecular factors, such as VEGF, PDGF, ANG-1 y 2, EGF, FGF, TGF-β1, TGF-α, MCP-1, CXCL5, and MMPs [44,120], MSC increase the proliferation and migration of endothelial cells. It has been described that MSC through growth factors, cytokines, and EVs can promote the survival and regeneration of colonic epithelial cells [121] and limit colonic tissue damage such as atrophy, inflammation, and dysplasia [122].

3.1.3. Antifibrotic Effects

A pro-fibrotic state, induced by oxidative stress, inflammation, and aging, is related to numerous progressive and terminal illnesses. This architecture entails the over-deposition of extracellular matrix (ECM) proteins, such as fibronectin and collagens I and III, resulting in altered biological functions and reduced tissues regeneration capacity [47,123]. In vivo studies disclose an anti-fibrotic effect of MSC like against skeletal muscular fibrosis, primarily through MMP-1 [124], or kidney or liver fibrosis via VEGF and HGF secretion, and further eliminates TGF-β1-induced fibrotic changes [125,126], with TGF-β1/Smad route which is a major pathogenic mechanism in tissue fibrosis [127,128]. MSC secretome and EVs have also an antifibrotic effect by containing growth factors and cytokines, such as HGF, TGF-β3, TNF-α, and IL-10 [129,130,131], and through the inhibition of dermal fibroblast–myofibroblast transition by blocking the TGF-β1/Smad2/3 signalling pathway, respectively [129,132,133].
Recently, in a phase I-II pilot trial it was assessed safety and efficacy of local MSC injection to treat CD strictures reporting that it was well tolerated and may offer a benefit. This is an interesting discovery since intestinal fibrosis is a common complication in IBD, especially in CD, causing bowel constriction and obstruction.

3.1.4. Anti-Apoptotic Effects

The study of anti-apoptotic mechanisms of MSC in various organ injury models is of great interest [47]. AT-MSC induce the upregulation of the cell proliferation marker Ki67 and the antiapoptotic markers BCL2 and SURVIVIN, and the downregulation of apoptosis indicators like TUNEL, annexin V, CASPASE3, and CASPASE9 [134]. Also, exosomes from BM-MSC containing miRNA-146a-5p reduce neuron apoptosis and inflammation through the downregulation of IRAK1 and NFAT5 expression [135]. For IBD, it has been proposed the use of MSC expressing IL-37b, cells transduced with an adenovirus vector expressing IL-37b, since they can inhibit the inflammatory process and cellular apoptosis [108]. Also, it has been evidenced that EVs from BM-MSC suppress the apoptosis through the reduction of the cleavage of caspase-3, caspase-8 and caspase-9 in an animal model [136].

3.1.5. Anti-Oxidative Stress Effects

Oxidative stress is a common and crucial pathophysiological mechanism of several illnesses [137]. Oxidative stress is due to the imbalance of pro-oxidants, or free radicals, and antioxidants, inducing a structural modification of lipids, proteins, and DNA causing pathology or damage to a cell or tissue [138]. Reactive oxygen species (ROS), such as superoxide anion (O2 (−)), hydroxyl radical ((·)OH), and hydrogen peroxide (H2O2) are the most studied free radicals [139,140]. The dynamic balance of ROS production and metabolism is crucial for tissue homeostasis, when the disruption of this balance lead to oxidative stress and tissue damage [141]. Although the relationship between the immune system and oxidative stress is not elucidated, leukocytes and pro-inflammatory mediators increase free radicals composition and disrupt the redox environment inducing a positive feedback loop [142]. Neutrophils are contributors to oxidative stress in inflammation, since they have an extensive activity of myeloperoxidase (MPO), which play a significant role in oxidative stress by catalyzing H2O2 to hypochlorite [143,144].
Several studies reported the resistance of MSC to oxidative injury, related to the constitutive expression of antioxidant enzymes (SOD1, SOD2, catalase (CAT), glutathione peroxidase (Gpx), high levels of the antioxidant glutathione (GSH)) [145], and other proteins such as HSP70 and SIRT, which may also play a role in the resistance to oxidative stress [146]. Antioxidant effects of MSC have been observed in vitro and in vivo using multiple disease models (aging, gastrointestinal inflammation, ischemic injuries) [47] identifying various mechanisms such as retrieval of free radicals, promotion of native antioxidant defenses, immunomodulation via ROS suppression and endowing functional mitochondria to damaged cells [137]. Also, MSC can decrease ROS and MPO activity in monocytes and macrophages, reducing their pro-inflammatory phenotype [147,148]. In a rat model of colitis, EVs from BM-MSC reduce oxidative perturbations increasing antioxidant enzymes (SOD and GSH) and decreasing in the activity of malondialdehyde (MDA) and MPO [136]. All these data indicate that to prevent oxidative injury, MSC induce an immunomodulation depending on their antioxidant properties.

3.1.6. Antimicrobial Effects

MSC resistance to viral infection is mainly due to the expression of interferon (IFN) and IFN-stimulated genes (ISGs) [149] which hamper infection before viruses can cross the cell membrane, as demonstrated to be so for influenza A virus and SARS coronavirus [150]. MSC can act directly by releasing antimicrobial peptides [151,152] leading to cell destruction, interfering with membrane integrity, inhibiting DNA, RNA or protein synthesis; and engaging with specific intracellular targets [153,154,155,156]. LL-37 is one of this kind of peptide showing a broad spectrum of antibacterial endeavors against both Gram-negative and Gram-positive bacteria [157,158,159] and antifungal [160] and antiviral activities [161]. Also, LL-37 is related to the modulation of Toll-like receptors (TLRs), which is a precursor of the immunomodulatory activity of MSC [162]. Also, MSC can act by releasing soluble proteins with a known antimicrobial effect such as interleukin-10 (IL-10), prostaglandin E2 (PGE2), tumor necrosis and factor-alpha (TNF-α) [163], IDO [164], and interleukin-17 [165]. In addition, the secretoma of MSC (conditioned medium) has an anti-microbial activity, including against E. coli and S. aureus [152,166], E. epidemidis [166], Vibrio cholerae [129,167], P. aeruginosa [168], Mycobacterium tuberculosis [169], Acinetobacter baumannii [170], and several Candida species [171,], including effects against biofilm build-up [172,173].

3.1.7. Anti-Tumor Effects

Cell communication between tumor cells and stromal environment is key to cancer pathophysiology. Pro- or anti-tumorigenic effect of MSC depends on the tissue origin of MSC and the kind of tumor [174,175] and MSC from uterus and pregnancy-related tissues seems to have a wider antitumor effect, which make them good candidates for MSC-based therapy for cancer [176].
MSC produce high level of cytokines, such as IFN-α, DKK-1/3, IL12, TRAIL, TNFSF14 (also known as LIGHT), FLT-3 ligand, CXCL10 and LAP, known to inhibit tumor growth in vivo in breast cancer cells [177,178,179,180]. Also, the antitumoral effect of MSC may be, in part, due to the presence in the secretome of tissue inhibitors of matrix metalloproteinase (TIMP-1 and TIMP-2) [177,181], associated with the migration suppression and invasion of cancer cells.
Other mechanism used by MSC to take effect on cancer cells is through EVs, since cancer cells internalize EVs more efficiently than normal cells [182,183]. It has been reported that EVs from human UC-MSC inhibit the development of bladder carcinoma, reducing the phosphorylation of Akt protein kinase and increasing cleaved caspase-3 [184], and EVs from AT-MSC suppress ovarian cancer cells proliferation through their exosomal miRNA content [185].

3.1.8. IBD and Colon Cancer

It was noted that anti-inflammatory drugs can effectively reduce the risk of developing colitis associated with colorectal cancer (CAC) [186]. However, long-term use of these therapies is restricted due to their potentially fatal adverse reactions [187]. Therefore, there is a pressing need to explore novel anti-inflammatory therapeutic approaches to cancer.
Several studies showed that MSC inhibited CRC initiation in an azoxymethane (AOM)/dextran sulfate sodium (DSS) mouse model [188,189,190], which closely mimics the mechanisms of human CAC [191]. Furthermore, several mechanisms for this effect have been identified, such as through IL-6-STAT3 signalling [188] or regulating the differentiation of Treg cells by Smad2 [190].
Recently, MSC have been shown to migrate to the intestine following intraperitoneal injection and prevent the appearance of colitis-associated colorectal cancer. This inhibition effect was characterized by a decrease in weight loss, a longer colon length, and a reduction in the number of tumors. Moreover, MSC also reduced tumor cell proliferation and caused tumor cell apoptosis [63]. This study also found that MSC did not change the abundance, but rather the diversity and composition of the intestinal microbiome. The analysis in genus levels revealed that an injection of MSC increased the abundance of potentially beneficial bacteria and reduced the amount of potentially harmful bacteria in the gut microbiome of mice. It is worth noting that the wealth of Parabacteroides, Staphylococcus, Acetatifactor, Intestinimonas, and Candidatus Saccharimonas was increased after administering MSC. Parabacteroides in faecal matter was displayed to bind inversely to colon tumor numbers and has anti-inflammatory and anticancer properties [192]. Staphylococcus is a commensal strain allegedly apoptosis trigger [193] and may protect from neoplasia [194]. Other research also demonstrated that MSC attenuate gut microbiota dysbiosis [80]. These are interesting discoveries as intestinal microbes are implicated in the intestinal defense function and immune system maturation, whereas gut microbiome dysbiosis plays a role in the pathogenesis of IBD and CRC [195]. Consequently, all of these data suggest that MSC may be a promising strategy for colitis-associated colorectal cancer.

3.2. Clinical Trials

3.2.1. Clinical Trials on MSC Transplantation in IBD

Over 1000 clinical trials based on MSC therapy are registered on the NIH Clinical Trial Database (https://ClinicalTrials.gov/), of them 491 trials (47.1%) for immune disorders and of these last, 34 trials conducted on IBD patients [196].
Trials on IBD used autologous or allogeneic (most of them) MSC transplantation by local injection to treat fistulizing CD or by systemic intravenous infusions to treat UC or luminal Crohn’s disease. Allogeneic MSC are more convenient since they can be expanded in mass to obtain fully characterized and enough cells dose prior to administration.

3.2.2. MSC in Fistulizing and Perianal CD

Fistula relapse occurs in approximately 30% of CD patients and is associated with abscess formation, decreased organ function and reduced quality of life [197]. Although the pathogenesis of complex perianal fistula is unknown, it is assumed that it is due to an immune-mediated disturb in the context of CD. The perianal fistula treatment aims to block fistula from draining, to avoid complications such as the fecal incontinence, and reach a durable closure. Currently, the treatment includes conventional drugs (antibiotics and immunomodulators), biological therapy based on anti-TNF antibodies (e.g., infliximab, adalimumab, and certolizumab) and surgical treatments [198], but they fail to maintain a durable closure of the fistula.
In 2003, the first study on MSC-therapy for perianal fistula on CD patients was published reporting a successful healing after local administration [199]. Since then, more phase I/II studies were carried with AT-MSC and BM-MSC, so autologous [200,201,202,203,204] as allogeneic [97,98,104,205], transplanted after ex vivo expansion. In these studies, peri-fistula injections of MSC promoted healing in many cases of complex perianal fistulas in CD patients, refractory to conventional or biological therapy.
In 2016, Panés et al. completed the ADMIRE trial, a phase III randomized, double-blind, placebo-controlled study using the above mentioned Cx601 product (now called darvadstrocel) for allogeneic AT-MSC transplantation in CD patients with complex perianal fistulas [105]. This randomized clinical trial involved 212 CD patients, conducted in 49 hospitals. A higher number of patients treated with Cx601 vs. placebo achieved remission at 24 weeks. The benefit over placebo was sustained 52 weeks after local injection [106]. Nevertheless, one randomized clinical trial reported no differences between treated and control patients after one year of follow-up [206].
Recently, a meta-analysis on CD perianal fistulas has reported that MSC therapy increased healing rate compared to control groups (OR 0.379, 95% CI 0.152–0.947), existing significant differences between autologous and allogenic MSC (69.4% and 50.7% (p = 0.020), respectively), but CD perianal fistulas treated with MSC achieve an overall healing rate of 64.1% [207]. In addition, one more recent meta-analysis evidenced the effective and safety application of MSC combined with fibrin glue for complex perianal fistula (CD or non-CD) evidencing an angiogenic effect of fibrin glue which together with MSC differentiation capacity show a synergistic effect on fistula’s healing [208]. Nevertheless, studies evaluated in this meta-analysis seems to be heterogeneous regarding fistula closure definition and MSC administration and doses. There is a controversial regarding these results which will be referred below in the section of “limitations of MSC therapy in IBD”.

3.2.3. MSC Transplantation in Luminal Crohn’s Disease

Until today, only BM-MSC and MSC from neonatal tissues have been employed as systemic therapy to treat luminal CD. Two phase I trials have evaluated the intravenous administration of autologous BM-MSC in luminal CD patients, but the results are disappointing, since the therapy failed to achieve clinical remission [209,210]. Similarly, phase I-II trials on intravenous administration of allogeneic BM-MSC in moderate-severe refractory CD only showed a clinical remission rate of 32% [99,211,212,213]. Regarding the use of MSC from neonatal tissues such as placenta, umbilical cord, or amnion in clinical trial, the results are preliminary or pending to be published [214,215,216].
A recent meta-analysis, which includes 18 human trials on CD patients (n = 360), reported that MSC therapy induces maintained remission for 3 to 24 months and decreases activity index, endoscopic index of severity and simplified endoscopy score. But also concluded the need to carry out high-quality randomized controlled clinical trials and basic research [96].

3.2.4. MSC Transplantation in Ulcerative Colitis

Although several clinical trials were conducted with MSC therapy in more than 200 UC patients, few of them are fully published and the clinical remission rate is doubtful due to imprecise definitions [101,217,218,219]. Recently, a meta-analysis reviewed 7 trials (n = 216 MSC treated patients) of whom 2 studies (n = 33 patients) use submucosal MSC administration by colonoscopy, 3 studies (n = 136 patients) opted to intravenous infusion and for the remaining 2 (n = 47) the administration route is not available [121]. Four of these studies do not include a control group and only one was a randomized controlled trial. Although the healing rate, the main outcome of trials, was not clearly defined in 4 single-arm study the overall healing rate was 0.787 and in the other 3 studies the healing rate was 0.791 and 0.853, respectively for the 2 group of treatment (MSC vs. 5-ASA and MSC + 5-ASA vs. placebo + 5-ASA).

4. Limitations of MSC Therapy in IBD

The limitations associated with MSC clinical use are based on the difficulty to provide a consistent supply complying with the required cell stability, the high cost related to the isolation and handling process and adverse events such as the infusional toxicity [220] or cellular rejection [221,222]. Despite of some positive outcomes [101], until now disappointing results have been found with regards to the systemic administration of MSC to patients with IBD [223]. Safety of systemic application of MSC, still need to be explored since several clinical trials reported aggravation of UC or CD [224]. Nevertheless, the optimal source of MSC, route of administration and dosage have to be resolved yet. [223,224]. In addition, the basic limitation of this therapy is the great number of cells required for transplantation [225,226,227,228,229].
Instead, regarding CD treatment, there are still many unsolved questions that are concerning: (i) differences in the dose of MSC injected and the number of injections; (ii) the most appropriate technique for the MSC transplant is still controversial in the literature (e.g., direct injection, injection with fibrin glue, and delivery on a fistula plug); (iii) the time point of fistula healing; (iv) criteria for defining fistula healing; (v) the best tissue to obtain the MSC; (vi) unclear treatment mechanisms; and (vii) eligible patients were all at least 18 years of age. In the future, an optimized and standardized treatment scheme should be adopted to enable patients to achieve long-term healing [208].

Regulatory Aspects

Over 1000 clinical in more than 40 countries have been conducted with MSC or their derivatives but only nine MSC-based products have been approved [230]. Of them, two are treatment for IBD patients such as allogenic AT-MSC to treat complex perianal fistulas in adult CD patients, approved by the European Medicines Agency of the European Union, and autologous AT-MSC for CD patients approved by the Korean Ministry of Food and Drug Safety.
Despite the clinical efficacy demonstrated in various clinical trials, few MSC products have been approved as a treatment, indicating on one side, significant differences in regulations between countries [231] and on the other side, obstacles to establish a transition from preclinical platforms to platforms clinics [232].
There is an important lack of agreement on relevant characterization criteria for clinical application since the current criteria were established in 2006 and did not consider some properties as immunomodulation which was demonstrated subsequently. Moreover, carry out a double-blind randomized clinical trial, considered the gold standard, is much more challenging with MSC-based products than with a classical drug, specially to dose, timing and delivery route testing [233]. Some changes are taking place to define clinical effectiveness for future clinical trial and drug approval [230].

5. New Perspectives for MSC Therapy in IBD

5.1. Importance of the Type of MSC

On the other hand, there are recognized limitations of MSC-based therapies related to the transplantation of proliferating living cells including immunological incompatibility, tumorigenicity, transmissible infections, and the initiation of the process of MSC senescence [18,234,235]. MSC administration can also induce thromboembolism [236], since they express procoagulant activity (PCA) that can initiate the coagulation process, when in contact with blood [237]. However, despite the mentioned limitations, MSC administration was safe [238,239].
The functional alterations evidenced in MSC from patients affected by systemic diseases such as diabetes mellitus, obesity, systemic lupus erythematosus, and rheumatoid arthritis, make them non-ideal donors. Also, the aging of MSC is a limiting factor as older cells lose stem cells competence and may enter in senescence. MSC aging can induce a senescence-associated phenotype and produce proinflammatory cytokines, which inhibits the regenerative process [240]. In this sense, the culture of MSCs induces a progressive loss of self-renewal and multipotency capacity [241,242], allowing no more than 30 to 40 population doublings, which constitutes a limitation for research and cell therapy [46,243,244,245]. This last limitation can be solved with the immortalization process [246] through telomere maintenance and suppression of p53- and Rb-mediated pathways, which allows the preservation of MSC phenotype and multipotency and increase the proliferation rate. In addition, both MSC morphology and functionality were similar after transduction of immortalization genes [245,247].
MSC functionality differs depending on their tissue origin, as it was reported between AT-MSC and BM-MSC regarding the proliferation and differentiation capacity, or paracrine mechanisms, like the secretion of pro-angiogenic factors [176]. With regard to anti-inflammatory effects, heterogeneity of MSC from different sources often secrete varying levels of soluble factors and thus exert diverse suppressive effects [229]. As it was described above, different tissue sources of MSC were employed depending on the clinical application. In this sense, AT-MSC were predominantly used and exhibited successful healing in fistulizing and perianal CD whereas BM-MSC seem induce clinical remission in UC patients. Some sources of MSC may be attractive for specific indications, such as MSC from reproductive tissues by their anti-tumor activity [180,248], or human uterine cervical stem cells (hUCESC) for their immunomodulatory [180] and antifungal activity [171], or MSC from dental pulp by neurological disorders [249].
The genetic manipulation of MSC provide a possibility to improve some of their capabilities; as it has been demonstrated with the incorporation of anti-inflammatory genes to MSC (such as IL-10; IDO; or Foxp3) or the induced overexpression of factors leading to cell survival; angiogenic; neuroprotective; or anti-cancer activities [176]. Nevertheless; biological attributes of the cell and gene therapy products; such as MSC integrating viral vectors; usually replication-deficient such as adenovirus; adeno- retrovirus or lentivirus; are associated with toxicity and carcinogenicity/tumorigenicity potential [250] which limit clinical application. It has been reported that CRISPR-Cas system may improve MSC effect [251,252]
For all these reasons, with a vision of future studies for IBD therapies, it is important to consider the origin of the MSC.

5.2. Cell-Free Therapy

The therapeutic effect of MSC is mainly produced via paracrine signalling, soluble factors (growth factors and cytokines) and EVs. Therefore, MSC secretome may be considered a new therapeutic product [253] which can avoid some concerns derived from the use of stem cells themselves and offers several advantages: (i) easier to handle in the clinical practice; (ii) easier evaluation of safety, dose, and potency; (iii) storage without toxic cryopreservative agents; and (iv) its use is cheaper since it could be mass produced, stored and been available for a ready-to-use application [44,254].
The cell-free therapy modalities used in experimental models of IBD, include cell extracts, conditioned medium (CM) and extracellular vesicles (EVs).

5.2.1. Cell Extracts

In DSS colitis mouse model, the intraperitoneally administration of UC-MSC cell extracts is more effective than UC-MSC to decrease activity index and histological scores, and to increase the body weight [72].

5.2.2. Conditioned Medium

The secretome is composed by factors/molecules (soluble proteins, lipids or nucleic acids) and vesicles secreted to the extracellular space by the cells, which is obtained culturing MSC, this is why it is also named conditioned medium (CM). MSC-CM can significantly improve criteria of pathophysiology in different animal models and is usually as effective as MSC transplantation (Revised by [44]). MSC-CM administered intraperitoneally has demonstrated to improve the symptoms of experimental colitis and decrease TNF-α and MMP2 expression in mice [83].

5.2.3. Extracellular Vesicles

EVs are membrane-bound phospholipid particles secreted by cells, containing RNA, proteins, chemokines, cytokines, integrins, and signal transduction factors, among others [47,255].
EVs can be classified as: (i) exosomes (30–120 nm); (ii) microparticles (150–1000 nm); and (iii) apoptotic bodies (500–2000 nm). EVs is a relevant intercellular communication pathway, acting through different mechanisms such as bind directly with a surface receptors to activate signalling pathway, or internalization trough membrane fusion or endocytosis process to release its content into the cytosol of the recipient cell [255].
EVs from MSC secretome are producing a great interest as a promising alternative to exploit MSC properties. Indeed, EVs show important advantages such as a less immunogenicity than MSC [256], the possibility of a high scale production and easier storage than MSC and a longer half-life in the bloodstream [257] and tropism towards inflamed tissues and tumors [258,259]. EVs have a similar immunoregulatory potential than MSC, but due to their small size, EVs are able to pass-through capillaries, enter the peripheral circulation and even cross the blood-brain barrier.
There are data indicating that EVs modulate macrophage polarization from M1-type to M2-type in the colonic tissue from DSS colitis mice through the enhancement of enhanced Arg-1 and CD206 expression in colon macrophages [260,261,262,263].
It has been also reported that MSC-secreted exosomes, by intraperitoneal injection, alleviates experimental-induced colitis (both DSS or 2,4,6-trinitrobenzenesulfonic acid (TNBS)-induced IBD mouse models) and reduced mortality in mice [100,102,136,264,265,266]. In these studies, EVs used were derived from BM, UC or AT tissues, and their administration improved the symptoms of UC by reducing weight loss, disease activity index, and colon mucosa damage and severity while increasing colon length. These therapeutic effects may be due, in part, to the anti-inflammatory and regenerative mechanisms induced by MSC-derived EVs.
On the other hand, it was revealed that the levels of VEGF-A, IFN-γ, TNF-α, IL-12, CCL-24, and CCL-17 and IL-17 were decreased, whereas the level of TGF-β, IL-4, and IL-10 were increased in lymph node and spleen of mice treated with exosomes. In addition, the percentages of CD4+ CD25+ Foxp3+ Treg cells were grown in these organ places [100]. Also, EVs-MSC induce the conversion of T helper type 1(Th1) into Th2 and decrease Th17 [266,267,268]. Also, EVs induce macrophage polarization to an M2 phenotype and promote UC repair through the JAK1/STAT1/STAT6 signalling pathway.
There are also data showing a protective effect of MSC-exosomes on the intestinal mucosal. After DSS or TNBS administration, the number of goblet cells in the colonic mucosa is significantly reduced, and it was shown that MSC-Exo injection significantly rescues the goblet cell population [266]. In addition, it was reported that intraperitoneal injection of MSC-Exo increased the expression of ZO-1, occludin, and claudin-1 in colon tissue and alleviates the disruption of the intestinal barrier.
Interestingly, it has also been reported that EVs-MSC restored the intestinal barrier function by improving mitochondrial dynamic balance [269]. This is also a relevant finding considering that mitochondria have a key role in energy metabolism, but are also involved in various pathophysiological processes, including cell apoptosis, and proliferation [270].

6. New Technologies for MSC Cultures and Mass Production of Secretome Derived Products

6.1. MSC In Vitro Production

MSC are widespread in many human organs and tissues, but in small numbers, so in vitro expansion is required for several weeks to achieve enough cells for cell-based therapies. Then, a large-scale expansion process is crucial to obtain enough cells for a therapy in a time and cost-effective manner. A large-scale production can be carry out using multi-layered flask, spinner flask, roller bottle, or bioreactor, which are used for expansion of MSC [271].
The use of bioreactors for MSC expansion allows to develop an industrial and commercial manufacturing and to obtain a monitoring of the expansion process, limiting batch-to-batch variability and fluctuations of cell culture conditions (pH, oxygen concentration, or nutrient gradients) caused by manual medium exchange.
Specific technological parameters must be considered to establish an expansion process in bioreactor such as the type of microcarriers, hydrodynamic parameters and agitation. Microcarriers are small beads (100 to 300 μm) made of diverse materials (polystyrene, dextran, cellulose, gelatine, glass, or decellularized tissue…) and used to increase the surface area available for cell attachment, providing a high surface-to-volume ratio for high-density cell culture with a cost reduction [272,273]. Recently, it was reported that, compared with 2D-cultured tonsil-derived MSC (TMSC), 3D-cultured TMSC showed a higher anti-inflammatory cytokine expression and induce a significant lower disease activity index score, body weight, colon length shortening and histological scoring index [274].
Compared with classical 2D cell culture systems [275], high scale-production is more advantageous, but there is the need to understand cell culture conditions which impact on MSC therapeutic effect [276].

6.2. Ex Vivo MSC Modifications toward More Specific Therapeutic Applications

Oxygen pressure, pH or cell pre-conditioning with inflammatory factors are ex vivo modifications that impact on MSC effect.
MSC are aerobic cells that need an adequate supply of oxygen; however, tissue oxygen saturation (5–7% O2) is at least 3 times lower than in standard cell culture flasks (21% O2) which can increase ROS production [277,]. Cell culture in hypoxic conditions can enhance MSC proliferation and decrease apoptosis [278], which suggest that it would be preferable to impose hypoxia during the expansion phase, to mimic the natural niche, instead of preconditioning MSC [279]. In addition, hypoxia can enhance pluripotent markers [280,281], cytokines and growth factors expression [282], improving MSC angiogenic capacity [283], anticancer effects [284] and their ability to migrate to the site of injury markers [285]. In this sense, it was reported that hypoxia-preconditioned MSC could reduce colon inflammation to a large extent compared with normoxia-preconditioned MSC in a DSS mouse model [286].
Usually, cell expansion is performed at 37 °C and neutral pH (7.2–7.4) it has been described [287] that pH can influence MSC metabolism which impact on their secretome and therapeutic effect. Studies about temperature and pH impact on MSC effect must be deeply evaluated.
MSC preconditioning with inflammatory cytokines, such as IFN-γ [288] and TNF-α [289] can improve therapeutic effect. Preconditioned MSC with IFN-γ and/or TNF-α, increase the expression of anti-inflammatory factors and therefore, improve the immunosuppressive function of MSC [290]. Also, preconditioning rat BM-MSC with licochalcone A (a flavoid compound extracted from legume liquorice) enhances MSC therapeutic activity in animal models of colitis, by increasing the number of MSC migrating to the inflammation site probably promoting CXCR4 expression [286].
It is also relevant the possibility to manipulate the EVs production by MSC in order to improve their anti-inflammatory properties. Thus, EVs from cultured medium of BM-MSC transfected with recombinant lentiviruses overexpress an anti-inflammatory miRNA (miR-146a), a well-known and which acts as a negative feedback regulator of the innate immune response. It was showed that these obtained EV significantly inhibited TNF receptor-associated factor 6 (TRAF6) and IL-1 receptor-associated kinase 1 (IRAK1) expression in TNBS-induced colitis of rats [291].
It has been also reported that EVs from thapsigargin (TSG-EV)-treated Warton’s jelly-derived MSC intraperitoneally injected ameliorated experimental colitis decreasing (or at least maintain similarly) pro-inflammatory factors expression (IFNγ, TNFα, and IL-1β) and inducing anti-inflammatory factors secretion such as TGFβ, COX-2 and IDO, up to 15 fold for this latter [292], which can induce the development of regulatory T cells and M2-type macrophages [293]. Indeed, TSG-EVs increase T cell proliferation and amount of Treg and M2-type macrophage and induce Th1 and Th17 differentiation. Thus, TSG-EV reduce the inflammatory response, maintain intestinal barrier integrity, and therefore, substantially alleviated colitis symptoms [292].
In addition, it was reported that UC-MSC cultured in a xeno-free medium showed an enhanced therapeutic effect in a mouse model for UC, compared with UC-MSC cultured in a conventional medium [294].
Another strategy is to engineer MSC to overexpressed specific factors to improve their therapeutic effect in IBD. In this sense, recently Zhou et al. showed that Nrf-2-modified hair follicle MSC improved DSS-induced UC in rats [11]. Nrf-2 is a transcription factor which regulates the expression of gene related to detoxification and anti-inflammatory response, among others [295].
The capacity of MSC and their secretome can be modulated through molecular or chemical stimuli, which suggest the interest to investigate these aspects to adapt the potentiality of MSC or their derivates to different therapeutic applications, such as IBD.

6.3. Standardization and Functional Tests Research for Specific Applications

Standardized criteria for the donor selection, transport, cell culture and storage of MSC during the manufacturing process are not established. Currently, allo-MSC from one or various donors are used as a universal drug for multiple patients; however, there is donor-to-donor, tissue source and cell culture strategies (with or without preconditioning) differences that can impact of clinical effectiveness [296,297,298,299]. Indeed, MSC expansion strategies impact deeply on their molecular phenotype compared to donor age [103,300,301,302], since the high-scale production of allo-MSC can minimize donor and bioprocessing variability, standardized practices should be established between laboratories/companies for MSC manufacturing and derived products, like the secretome. On the other hand, although MSC clinical effectiveness is in part due to the secretion of trophic factors, there is no established and standardized potency assays for the release of MSC- based therapy [303]. Thus, a functional characterization of allo-MSC or secretome is required for each therapeutic indication. In this sense, Caco-2 cells in vitro may be a biological platform to explore the functional capacity of the optimal MSC and their secretome products.

6.4. Route of Administration

The route of administration of MSC used in IBD were intravenous, intra-peritoneal or local. Intravenous administration was the most used in experimental studies in clinical trials. Despite the homing properties of MSC, a previous study has shown that less than 1% of MSC injected intravenously homing at the damaged intestinal tissue [304]. Another alternative route of administration in IBD is intraperitoneally, proven to be satisfactory in view of MSC migration capability towards the inflamed intestine and therapeutic effectiveness [305]. In addition, such as was referred more above, experimental studies demonstrated that EVs administrated by this way produced therapeutic effect in IBD [266,292]. Another possibility is the local administration which demonstrate positive beneficials in clinical trials for perianal fistulas in CD. In this sense, the follow-up study through week 104 of the clinical trial known as “Adipose derived mesenchymal stem cells for induction of remission in perianal fistulizing Crohn’s disease,” or ADMIRE-CD reported 7 treatment-emergent serious adverse events and clinical remission in 56% patients of the darvadstrocel group and 40% in the control group [306]. Regarding intraluminal lesions, the local administration through mucosa, that exhibit a high permeability but a low risk of overdose, seems to be a good option for drug delivery but the retention time is limited, and this is the main disadvantage and may be the cause of a less therapeutic effect [307,308]. Nevertheless, there is the possibility to combine the administration of MSC or their secretome-derived products with new technologies, such as mucoadhesive hydrogels. In this line, a recent study assessed the effect of rectal administration of a thermosensitive hydrogel loaded with CM from human uterine cervical mesenchymal stem cells (hUCESC-CM) in DSS-induced colitis mouse model [122]. Treatment hydrogel loaded with hUCESC-CM (H-hUCESC-CM) reduced body weight loss, gene expression of pro-inflammatory factors (TNF-α, IFN-γ, and IL-6) and colonic tissue damage such as atrophy, inflammation, and dysplasia. It has been reported that hydrogel alone can reduce pro-inflammatory cytokines expression, such as TNF-α and IFN-γ; probably due to polymers properties [309,310].
This last study shows the importance of the tissue source of MSC, the use of their secretome and the administration route using new technologies such as hydrogels. Human uterine cervical stem cells (hUCESC) are a characterized new population of MSC [180] isolated from the transformation zone of the human uterine cervix of healthy women [248] and whose secretome shows a potent regenerative, anti-inflammatory, anti-tumor and anti-microbial capacities [166,180,311]. On the other hand, hydrogels are employed due to their biocompatibility, similarity to the extracellular matrix, permeability for oxygen, small molecules and nutrients [312,313], converting them into optimal carriers of MSC secretome to restore tissue damages in IBD. Hydrogels can spread over colonic mucosa and release bioactive molecules from the MSC secretome, allowing a faster regeneration.
In this previous work [314], the hydrogel was developed using Pluronic® and one hydroxypropylcellulose (MK4M). Pluronics® are amphiphilic, thermosensitive, and bioadhesive and are mainly used to induce gelation (from liquid to solid state) [314,315]. MK4M is a viscous cellulose derivative used to generate mucoadhesion or even regulating the release of molecules. Therefore, the combination of both compounds improves hydrogel adhesion and protection to the colonic mucosa [316].

7. Conclusions

The complexity of IBD pathophysiology makes difficult to find a therapeutic strategy that effectively and globally addresses the different mechanisms implicated and clinical manifestations.
There is an evolution of the concept of the therapeutic interest of MSC: from the use of autologous to allogeneic MSC, from the first conception of their regenerative therapeutic effect to anti-inflammatory, and more recently to others such as anti-tumor and anti-inflammatory, anti-microbial or anti-oxidative stress. Likewise, there is an evolution from cell-based therapies to cell-free therapies based on their secretome-derived products. The development of all these evolutions could lead to the birth of a new therapeutic strategy for IBD.
However, for this, new challenges must be faced, such as conveniently integrating new approaches and technologies, such as the appropriate choice of the ideal source of MSC to counteract the different multifactorial pathophysiological mechanisms involved in IBD. In addition, for the implementation of the new products in the clinical practice, we must develop an adequate mass production and standardization process and the integration of new technological developments such as hydrogels that, based on nanotechnology, can optimize the therapeutic topic application. All this will allow us to design new therapeutic horizons for IBD based on biological therapy against its diverse and complex therapeutic targets.

Author Contributions

N.E., M.F., A.G.-J. and L.O.G. analyzed the bibliography and prepared the table and figure. F.J.V. and N.E. designed the project and wrote the manuscript. All authors reviewed the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by grants to N.E. and F.J.V from Instituto de Salud Carlos III (PI20/01122) and co-funded by European Union (ERDF/ESF, “Investing in your future”).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Cosnes, J.; Gower-Rousseau, C.; Seksik, P.; Cortot, A. Epidemiology and natural history of inflammatory bowel diseases. Gastroenterology 2011, 140, 1785–1794. [Google Scholar] [CrossRef] [PubMed]
  2. Burisch, J.; Pedersen, N.; Čuković-Čavka, S.; Brinar, M.; Kaimakliotis, I.; Duricova, D.; Shonová, O.; Vind, I.; Avnstrøm, S.; Thorsgaard, N.; et al. East-West gradient in the incidence of inflammatory bowel disease in Europe: The ECCO-EpiCom inception cohort. Gut 2014, 63, 588–597. [Google Scholar] [CrossRef] [PubMed]
  3. Gómez-Gómez, G.J.; Masedo, Á.; Yela, C.; Martínez-Montiel Mdel, P.; Casís, B. Current stage in inflammatory bowel disease: What is next? World J. Gastroenterol. 2015, 21, 11282–11303. [Google Scholar] [CrossRef] [PubMed]
  4. Lam, S.; Zuo, T.; Ho, M.; Chan, F.K.L.; Chan, P.K.S.; Ng, S.C. Review article: Fungal alterations in inflammatory bowel diseases. Aliment. Pharmacol. Ther. 2019, 50, 1159–1171. [Google Scholar] [CrossRef]
  5. Abdel Salam, A.G.; Ata, H.M.; Salman, T.M.; Rashed, L.A.; Sabry, D.; Schaalan, M.F. Potential therapeutic utility of mesenchymal stem cells in inflammatory bowel disease in mice. Int. Immunopharmacol. 2014, 22, 515–521. [Google Scholar] [CrossRef]
  6. Zhu, J.F.; Xu, Y.; Zhao, J.; Li, X.; Meng, X.; Wang, T.Q.; Zou, B.Y.; Zhao, P.Y.; Liu, Q.; Lu, C.L.; et al. IL-33 Protects Mice against DSS-Induced Chronic Colitis by Increasing Both Regulatory B Cell and Regulatory T Cell Responses as Well as Decreasing Th17 Cell Response. J. Immunol. Res. 2018, 2018, 1827901. [Google Scholar] [CrossRef]
  7. Abraham, C.; Cho, J.H. Inflammatory bowel disease. N. Engl. J. Med. 2009, 361, 2066–2078. [Google Scholar] [CrossRef]
  8. Gong, Y.; Lin, Y.; Zhao, N.; He, X.; Lu, A.; Wei, W.; Jiang, M. The Th17/Treg Immune Imbalance in Ulcerative Colitis Disease in a Chinese Han Population. Mediat. Inflamm 2016, 2016, 7089137. [Google Scholar] [CrossRef] [Green Version]
  9. Gálvez, J. Role of Th17 Cells in the Pathogenesis of Human IBD. ISRN Inflamm 2014, 2014, 928461. [Google Scholar] [CrossRef] [Green Version]
  10. Zhang, B.; Yeo, R.W.Y.; Lai, R.C.; Sim, E.W.K.; Chin, K.C.; Lim, S.K. Mesenchymal stromal cell exosome-enhanced regulatory T-cell production through an antigen-presenting cell-mediated pathway. Cytotherapy 2018, 20, 687–696. [Google Scholar] [CrossRef]
  11. Isidro, R.A.; Appleyard, C.B. Colonic macrophage polarization in homeostasis, inflammation, and cancer. Am. J. Physiol. Liver Physiol. 2016, 311, G59–G73. [Google Scholar] [CrossRef]
  12. Thomas, A.; Lodhia, N. Advanced therapy for inflammatory bowel disease: A guide for the primary care physician. J. Am. Board Fam. Med. 2014, 27, 411–420. [Google Scholar] [CrossRef] [Green Version]
  13. Jneid, H.; Anderson, J.L.; Wright, R.S.; Adams, C.D.; Bridges, C.R.; Casey, D.E., Jr.; Ettinger, S.M.; Fesmire, F.M.; Ganiats, T.G.; Lincoff, A.M.; et al. 2012 ACCF/AHA focused update of the guideline for the management of patients with unstable angina/non-ST-elevation myocardial infarction (updating the 2007 guideline and replacing the 2011 focused update): A report of the American College of Cardiology Foundation/American Heart Association Task Force on Practice Guidelines. J. Am. Coll. Cardiol. 2012, 60, 645–681. [Google Scholar] [CrossRef] [Green Version]
  14. Kappelman, M.D.; Rifas-Shiman, S.L.; Porter, C.Q.; Ollendorf, D.A.; Sandler, R.S.; Galanko, J.A.; Finkelstein, J.A. Direct health care costs of Crohn’s disease and ulcerative colitis in US children and adults. Gastroenterology 2008, 135, 1907–1913. [Google Scholar] [CrossRef] [Green Version]
  15. Mozaffari, S.; Nikfar, S.; Abdolghaffari, A.H.; Abdollahi, M. New biologic therapeutics for ulcerative colitis and Crohn’s disease. Expert Opin. Biol. Ther. 2014, 14, 583–600. [Google Scholar] [CrossRef]
  16. Bernstein, C.N. Treatment of IBD: Where we are and where we are going. Am. J. Gastroenterol. 2015, 110, 114–126. [Google Scholar] [CrossRef]
  17. Abdu-Allah, H.; El-Shorbagi, A.-N.; Abdel-Moty, S.; El-awady, R.; Abdel-Alim, A.-A. 5-Aminosalyclic Acid (5-ASA): A Unique Anti-Inflammatory Salicylate. Med. Chem. 2016, 6, 306–315. [Google Scholar] [CrossRef]
  18. Vizoso, F.J.; Eiro, N.; Costa, L.; Esparza, P.; Landin, M.; Diaz-Rodriguez, P.; Schneider, J.; Perez-Fernandez, R. Mesenchymal Stem Cells in Homeostasis and Systemic Diseases: Hypothesis, Evidences, and Therapeutic Opportunities. Int. J. Mol. Sci. 2019, 20, 3738. [Google Scholar] [CrossRef] [Green Version]
  19. Copelan, E.A. Hematopoietic stem-cell transplantation. N. Engl. J. Med. 2006, 354, 1813–1826. [Google Scholar] [CrossRef]
  20. Marmont, A.M. New horizons in the treatment of autoimmune diseases: Immunoablation and stem cell transplantation. Annu Rev. Med. 2000, 51, 115–134. [Google Scholar] [CrossRef]
  21. Snowden, J.A.; Badoglio, M.; Labopin, M.; Giebel, S.; McGrath, E.; Marjanovic, Z.; Burman, J.; Moore, J.; Rovira, M.; Wulffraat, N.M.; et al. Evolution, trends, outcomes, and economics of hematopoietic stem cell transplantation in severe autoimmune diseases. Blood Adv. 2017, 1, 2742–2755. [Google Scholar] [CrossRef] [Green Version]
  22. Balassa, K.; Danby, R.; Rocha, V. Haematopoietic stem cell transplants: Principles and indications. Br. J. Hosp. Med. 2019, 80, 33–39. [Google Scholar] [CrossRef]
  23. Kashyap, A.; Forman, S.J. Autologous bone marrow transplantation for non-Hodgkin’s lymphoma resulting in long-term remission of coincidental Crohn’s disease. Br. J. Haematol. 1998, 103, 651–652. [Google Scholar] [CrossRef]
  24. Musso, M.; Porretto, F.; Crescimanno, A.; Bondì, F.; Polizzi, V.; Scalone, R. Crohn’s disease complicated by relapsed extranodal Hodgkin’s lymphoma: Prolonged complete remission after unmanipulated PBPC autotransplant. Bone Marrow Transplant. 2000, 26, 921–923. [Google Scholar] [CrossRef] [Green Version]
  25. Söderholm, J.D.; Malm, C.; Juliusson, G.; Sjödahl, R. Long-term endoscopic remission of crohn disease after autologous stem cell transplantation for acute myeloid leukaemia. Scand. J. Gastroenterol. 2002, 37, 613–616. [Google Scholar] [CrossRef]
  26. Hawkey, C.J.; Allez, M.; Clark, M.M.; Labopin, M.; Lindsay, J.O.; Ricart, E.; Rogler, G.; Rovira, M.; Satsangi, J.; Danese, S.; et al. Autologous Hematopoetic Stem Cell Transplantation for Refractory Crohn Disease: A Randomized Clinical Trial. Jama 2015, 314, 2524–2534. [Google Scholar] [CrossRef]
  27. López-García, A.; Rovira, M.; Jauregui-Amezaga, A.; Marín, P.; Barastegui, R.; Salas, A.; Ribas, V.; Feu, F.; Elizalde, J.I.; Fernández-Avilés, F.; et al. Autologous Haematopoietic Stem Cell Transplantation for Refractory Crohn’s Disease: Efficacy in a Single-Centre Cohort. J. Crohn’s Colitis 2017, 11, 1161–1168. [Google Scholar] [CrossRef] [Green Version]
  28. Burt, R.K.; Kaiser, R.L., Jr.; Ruiz, M.A. Stem-cell transplantation for Crohn’s disease: Same authors, different conclusions? Lancet Gastroenterol. Hepatol. 2017, 2, 386–387. [Google Scholar] [CrossRef]
  29. Clerici, M.; Cassinotti, A.; Onida, F.; Trabattoni, D.; Annaloro, C.; Della Volpe, A.; Rainone, V.; Lissoni, F.; Duca, P.; Sampietro, G.; et al. Immunomodulatory effects of unselected haematopoietic stem cells autotransplantation in refractory Crohn’s disease. Dig. Liver Dis. 2011, 43, 946–952. [Google Scholar] [CrossRef] [PubMed]
  30. Marmont, A.M. Immunoablation followed or not by hematopoietic stem cells as an intense therapy for severe autoimmune diseases. New perspectives, new problems. Haematologica 2001, 86, 337–345. [Google Scholar] [PubMed]
  31. Cassinotti, A.; Passamonti, F.; Segato, S. CELL THERAPY IN INFLAMMATORY BOWEL DISEASE. Pharmacol. Res. 2021, 163, 105247. [Google Scholar] [CrossRef]
  32. Gooley, T.A.; Chien, J.W.; Pergam, S.A.; Hingorani, S.; Sorror, M.L.; Boeckh, M.; Martin, P.J.; Sandmaier, B.M.; Marr, K.A.; Appelbaum, F.R.; et al. Reduced mortality after allogeneic hematopoietic-cell transplantation. N. Engl. J. Med. 2010, 363, 2091–2101. [Google Scholar] [CrossRef] [Green Version]
  33. Hahn, T.; McCarthy, P.L., Jr.; Hassebroek, A.; Bredeson, C.; Gajewski, J.L.; Hale, G.A.; Isola, L.M.; Lazarus, H.M.; Lee, S.J.; Lemaistre, C.F.; et al. Significant improvement in survival after allogeneic hematopoietic cell transplantation during a period of significantly increased use, older recipient age, and use of unrelated donors. J. Clin. Oncol. 2013, 31, 2437–2449. [Google Scholar] [CrossRef] [Green Version]
  34. Mehta, K.; Jaiswal, P.; Briggs, F.; Faubion, W.A.; Tabibian, J.H.; Cominelli, F.; Dave, M. In-patient outcomes of Hematopoietic Stem Cell Transplantation in Patients with Immune Mediated Inflammatory Diseases: A Nationwide Study. Sci. Rep. 2018, 8, 6825. [Google Scholar] [CrossRef] [Green Version]
  35. Penack, O.; Smith, O.M.; Cunningham-Bussel, A.; Liu, X.; Rao, U.; Yim, N.; Na, I.K.; Holland, A.M.; Ghosh, A.; Lu, S.X.; et al. NOD2 regulates hematopoietic cell function during graft-versus-host disease. J. Exp. Med. 2009, 206, 2101–2110. [Google Scholar] [CrossRef]
  36. Al Nabhani, Z.; Montcuquet, N.; Roy, M.; Dussaillant, M.; Hugot, J.P.; Barreau, F. Complementary Roles of Nod2 in Hematopoietic and Nonhematopoietic Cells in Preventing Gut Barrier Dysfunction Dependent on MLCK Activity. Inflamm. Bowel Dis. 2017, 23, 1109–1119. [Google Scholar] [CrossRef] [Green Version]
  37. Lee, C.; Choi, C.; Kang, H.S.; Shin, S.W.; Kim, S.Y.; Park, H.C.; Hong, S.N. NOD2 Supports Crypt Survival and Epithelial Regeneration after Radiation-Induced Injury. Int. J. Mol. Sci. 2019, 20, 4297. [Google Scholar] [CrossRef] [Green Version]
  38. Jansen, S.A.; Nieuwenhuis, E.E.S.; Hanash, A.M.; Lindemans, C.A. Challenges and opportunities targeting mechanisms of epithelial injury and recovery in acute intestinal graft-versus-host disease. Mucosal Immunol. 2022, 15, 605–619. [Google Scholar] [CrossRef]
  39. Tse, W.T.; Pendleton, J.D.; Beyer, W.M.; Egalka, M.C.; Guinan, E.C. Suppression of allogeneic T-cell proliferation by human marrow stromal cells: Implications in transplantation. Transplantation 2003, 75, 389–397. [Google Scholar] [CrossRef]
  40. Jiang, X.X.; Zhang, Y.; Liu, B.; Zhang, S.X.; Wu, Y.; Yu, X.D.; Mao, N. Human mesenchymal stem cells inhibit differentiation and function of monocyte-derived dendritic cells. Blood 2005, 105, 4120–4126. [Google Scholar] [CrossRef] [Green Version]
  41. Lotfinegad, P.; Shamsasenjan, K.; Movassaghpour, A.; Majidi, J.; Baradaran, B. Immunomodulatory nature and site specific affinity of mesenchymal stem cells: A hope in cell therapy. Adv. Pharm. Bull. 2014, 4, 5–13. [Google Scholar] [CrossRef] [PubMed]
  42. Ankrum, J.A.; Ong, J.F.; Karp, J.M. Mesenchymal stem cells: Immune evasive, not immune privileged. Nat. Biotechnol 2014, 32, 252–260. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Friedenstein, A.J.; Chailakhjan, R.K.; Lalykina, K.S. The development of fibroblast colonies in monolayer cultures of guinea-pig bone marrow and spleen cells. Cell Prolif. 1970, 3, 393–403. [Google Scholar] [CrossRef]
  44. Vizoso, F.J.; Eiro, N.; Cid, S.; Schneider, J.; Perez-Fernandez, R. Mesenchymal Stem Cell Secretome: Toward Cell-Free Therapeutic Strategies in Regenerative Medicine. Int. J. Mol. Sci. 2017, 18, 1852. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Dominici, M.; Le Blanc, K.; Mueller, I.; Slaper-Cortenbach, I.; Marini, F.; Krause, D.; Deans, R.; Keating, A.; Prockop, D.; Horwitz, E. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 2006, 8, 315–317. [Google Scholar] [CrossRef] [PubMed]
  46. Samsonraj, R.M.; Raghunath, M.; Nurcombe, V.; Hui, J.H.; van Wijnen, A.J.; Cool, S.M. Concise Review: Multifaceted Characterization of Human Mesenchymal Stem Cells for Use in Regenerative Medicine. Stem Cells Transl. Med. 2017, 6, 2173–2185. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Fernández-Francos, S.; Eiro, N.; Costa, L.A.; Escudero-Cernuda, S.; Fernández-Sánchez, M.L.; Vizoso, F.J. Mesenchymal Stem Cells as a Cornerstone in a Galaxy of Intercellular Signals: Basis for a New Era of Medicine. Int. J. Mol. Sci. 2021, 22, 3576. [Google Scholar] [CrossRef]
  48. De Bari, C. Are mesenchymal stem cells in rheumatoid arthritis the good or bad guys? Arthritis Res. Ther. 2015, 17, 113. [Google Scholar] [CrossRef] [Green Version]
  49. Cheng, R.J.; Xiong, A.J.; Li, Y.H.; Pan, S.Y.; Zhang, Q.P.; Zhao, Y.; Liu, Y.; Marion, T.N. Mesenchymal Stem Cells: Allogeneic MSC May Be Immunosuppressive but Autologous MSC Are Dysfunctional in Lupus Patients. Front. Cell Dev. Biol. 2019, 7, 285. [Google Scholar] [CrossRef]
  50. Rennert, R.C.; Sorkin, M.; Januszyk, M.; Duscher, D.; Kosaraju, R.; Chung, M.T.; Lennon, J.; Radiya-Dixit, A.; Raghvendra, S.; Maan, Z.N.; et al. Diabetes impairs the angiogenic potential of adipose-derived stem cells by selectively depleting cellular subpopulations. Stem Cell Res. Ther. 2014, 5, 79. [Google Scholar] [CrossRef] [Green Version]
  51. Hou, R.; Liu, R.; Niu, X.; Chang, W.; Yan, X.; Wang, C.; Li, J.; An, P.; Li, X.; Yin, G.; et al. Biological characteristics and gene expression pattern of bone marrow mesenchymal stem cells in patients with psoriasis. Exp. Dermatol. 2014, 23, 521–523. [Google Scholar] [CrossRef] [Green Version]
  52. Cárdenes, N.; Álvarez, D.; Sellarés, J.; Peng, Y.; Corey, C.; Wecht, S.; Nouraie, S.M.; Shanker, S.; Sembrat, J.; Bueno, M.; et al. Senescence of bone marrow-derived mesenchymal stem cells from patients with idiopathic pulmonary fibrosis. Stem Cell Res. Ther. 2018, 9, 257. [Google Scholar] [CrossRef]
  53. Zhou, X.; Hong, Y.; Zhang, H.; Li, X. Mesenchymal Stem Cell Senescence and Rejuvenation: Current Status and Challenges. Front. Cell Dev. Biol. 2020, 8, 364. [Google Scholar] [CrossRef]
  54. Degirmenci, B.; Valenta, T.; Dimitrieva, S.; Hausmann, G.; Basler, K. GLI1-expressing mesenchymal cells form the essential Wnt-secreting niche for colon stem cells. Nature 2018, 558, 449–453. [Google Scholar] [CrossRef]
  55. Nowarski, R.; Jackson, R.; Flavell, R.A. The Stromal Intervention: Regulation of Immunity and Inflammation at the Epithelial-Mesenchymal Barrier. Cell 2017, 168, 362–375. [Google Scholar] [CrossRef] [Green Version]
  56. Beswick, E.J.; Johnson, J.R.; Saada, J.I.; Humen, M.; House, J.; Dann, S.; Qiu, S.; Brasier, A.R.; Powell, D.W.; Reyes, V.E.; et al. TLR4 activation enhances the PD-L1-mediated tolerogenic capacity of colonic CD90+ stromal cells. J. Immunol. 2014, 193, 2218–2229. [Google Scholar] [CrossRef] [Green Version]
  57. Thomson, C.A.; Nibbs, R.J.; McCoy, K.D.; Mowat, A.M. Immunological roles of intestinal mesenchymal cells. Immunology 2020, 160, 313–324. [Google Scholar] [CrossRef] [Green Version]
  58. Biancheri, P.; Di Sabatino, A.; Corazza, G.R.; MacDonald, T.T. Proteases and the gut barrier. Cell Tissue Res. 2013, 351, 269–280. [Google Scholar] [CrossRef]
  59. Hidalgo-Garcia, L.; Molina-Tijeras, J.A.; Huertas-Peña, F.; Ruiz-Malagón, A.J.; Diez-Echave, P.; Vezza, T.; Rodríguez-Sojo, M.J.; Morón, R.; Becerra-Massare, P.; Rodríguez-Nogales, A.; et al. Intestinal mesenchymal cells regulate immune responses and promote epithelial regeneration in vitro and in dextran sulfate sodium-induced experimental colitis in mice. Acta Physiol. 2021, 233, e13699. [Google Scholar] [CrossRef]
  60. Thursby, E.; Juge, N. Introduction to the human gut microbiota. Biochem. J. 2017, 474, 1823–1836. [Google Scholar] [CrossRef]
  61. Lacroix, V.; Cassard, A.; Mas, E.; Barreau, F. Multi-Omics Analysis of Gut Microbiota in Inflammatory Bowel Diseases: What Benefits for Diagnostic, Prognostic and Therapeutic Tools? Int. J. Mol. Sci. 2021, 22, 1255. [Google Scholar] [CrossRef] [PubMed]
  62. Ocansey, D.K.W.; Wang, L.; Wang, J.; Yan, Y.; Qian, H.; Zhang, X.; Xu, W.; Mao, F. Mesenchymal stem cell-gut microbiota interaction in the repair of inflammatory bowel disease: An enhanced therapeutic effect. Clin. Transl. Med. 2019, 8, 31. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. He, R.; Han, C.; Li, Y.; Qian, W.; Hou, X. Cancer-Preventive Role of Bone Marrow-Derived Mesenchymal Stem Cells on Colitis-Associated Colorectal Cancer: Roles of Gut Microbiota Involved. Front. Cell Dev. Biol. 2021, 9, 642948. [Google Scholar] [CrossRef] [PubMed]
  64. Xiao, E.; He, L.; Wu, Q.; Li, J.; He, Y.; Zhao, L.; Chen, S.; An, J.; Liu, Y.; Chen, C.; et al. Microbiota regulates bone marrow mesenchymal stem cell lineage differentiation and immunomodulation. Stem Cell Res. Ther. 2017, 8, 213. [Google Scholar] [CrossRef]
  65. Lauro, M.L.; Burch, J.M.; Grimes, C.L. The effect of NOD2 on the microbiota in Crohn’s disease. Curr. Opin. Biotechnol. 2016, 40, 97–102. [Google Scholar] [CrossRef] [Green Version]
  66. Sidiq, T.; Yoshihama, S.; Downs, I.; Kobayashi, K.S. Nod2: A Critical Regulator of Ileal Microbiota and Crohn’s Disease. Front. Immunol. 2016, 7, 367. [Google Scholar] [CrossRef]
  67. Gu, L.; Ren, F.; Fang, X.; Yuan, L.; Liu, G.; Wang, S. Exosomal MicroRNA-181a Derived From Mesenchymal Stem Cells Improves Gut Microbiota Composition, Barrier Function, and Inflammatory Status in an Experimental Colitis Model. Front. Med. 2021, 8, 660614. [Google Scholar] [CrossRef]
  68. Tanaka, F.; Tominaga, K.; Ochi, M.; Tanigawa, T.; Watanabe, T.; Fujiwara, Y.; Ohta, K.; Oshitani, N.; Higuchi, K.; Arakawa, T. Exogenous administration of mesenchymal stem cells ameliorates dextran sulfate sodium-induced colitis via anti-inflammatory action in damaged tissue in rats. Life Sci. 2008, 83, 771–779. [Google Scholar] [CrossRef]
  69. Wang, M.; Liang, C.; Hu, H.; Zhou, L.; Xu, B.; Wang, X.; Han, Y.; Nie, Y.; Jia, S.; Liang, J.; et al. Intraperitoneal injection (IP), Intravenous injection (IV) or anal injection (AI)? Best way for mesenchymal stem cells transplantation for colitis. Sci. Rep. 2016, 6, 30696. [Google Scholar] [CrossRef]
  70. Lee, H.J.; Oh, S.H.; Jang, H.W.; Kwon, J.H.; Lee, K.J.; Kim, C.H.; Park, S.J.; Hong, S.P.; Cheon, J.H.; Kim, T.I.; et al. Long-Term Effects of Bone Marrow-Derived Mesenchymal Stem Cells in Dextran Sulfate Sodium-Induced Murine Chronic Colitis. Gut Liver 2016, 10, 412–419. [Google Scholar] [CrossRef] [Green Version]
  71. Chao, K.; Zhang, S.; Qiu, Y.; Chen, X.; Zhang, X.; Cai, C.; Peng, Y.; Mao, R.; Pevsner-Fischer, M.; Ben-Horin, S.; et al. Human umbilical cord-derived mesenchymal stem cells protect against experimental colitis via CD5(+) B regulatory cells. Stem Cell Res. Ther. 2016, 7, 109. [Google Scholar] [CrossRef] [Green Version]
  72. Song, J.Y.; Kang, H.J.; Hong, J.S.; Kim, C.J.; Shim, J.Y.; Lee, C.W.; Choi, J. Umbilical cord-derived mesenchymal stem cell extracts reduce colitis in mice by re-polarizing intestinal macrophages. Sci. Rep. 2017, 7, 9412. [Google Scholar] [CrossRef] [Green Version]
  73. Yu, Y.; Song, E.M.; Lee, K.E.; Joo, Y.H.; Kim, S.E.; Moon, C.M.; Kim, H.Y.; Jung, S.A.; Jo, I. Therapeutic potential of tonsil-derived mesenchymal stem cells in dextran sulfate sodium-induced experimental murine colitis. PLoS ONE 2017, 12, e0183141. [Google Scholar] [CrossRef] [Green Version]
  74. Yu, Y.; Zhao, T.; Yang, D. Cotransfer of regulatory T cells improve the therapeutic effectiveness of mesenchymal stem cells in treating a colitis mouse model. Exp. Anim. 2017, 66, 167–176. [Google Scholar] [CrossRef]
  75. Pouya, S.; Heidari, M.; Baghaei, K.; Asadzadeh Aghdaei, H.; Moradi, A.; Namaki, S.; Zali, M.R.; Hashemi, S.M. Study the effects of mesenchymal stem cell conditioned medium injection in mouse model of acute colitis. Int. Immunopharmacol. 2018, 54, 86–94. [Google Scholar] [CrossRef]
  76. Heidari, M.; Pouya, S.; Baghaei, K.; Aghdaei, H.A.; Namaki, S.; Zali, M.R.; Hashemi, S.M. The immunomodulatory effects of adipose-derived mesenchymal stem cells and mesenchymal stem cells-conditioned medium in chronic colitis. J. Cell. Physiol. 2018, 233, 8754–8766. [Google Scholar] [CrossRef]
  77. Gonzalez-Rey, E.; Anderson, P.; González, M.A.; Rico, L.; Büscher, D.; Delgado, M. Human adult stem cells derived from adipose tissue protect against experimental colitis and sepsis. Gut 2009, 58, 929–939. [Google Scholar] [CrossRef]
  78. Fu, Z.W.; Zhang, Z.Y.; Ge, H.Y. Mesenteric injection of adipose-derived mesenchymal stem cells relieves experimentally-induced colitis in rats by regulating Th17/Treg cell balance. Am. J. Transl. Res. 2018, 10, 54–66. [Google Scholar]
  79. Lee, B.C.; Shin, N.; Lee, J.Y.; Kang, I.; Kim, J.J.; Lee, S.E.; Choi, S.W.; Webster, G.A.; Kang, K.S. MIS416 Enhances Therapeutic Functions of Human Umbilical Cord Blood-Derived Mesenchymal Stem Cells Against Experimental Colitis by Modulating Systemic Immune Milieu. Front. Immunol. 2018, 9, 1078. [Google Scholar] [CrossRef] [Green Version]
  80. Soontararak, S.; Chow, L.; Johnson, V.; Coy, J.; Wheat, W.; Regan, D.; Dow, S. Mesenchymal Stem Cells (MSC) Derived from Induced Pluripotent Stem Cells (iPSC) Equivalent to Adipose-Derived MSC in Promoting Intestinal Healing and Microbiome Normalization in Mouse Inflammatory Bowel Disease Model. Stem Cells Transl. Med. 2018, 7, 456–467. [Google Scholar] [CrossRef] [Green Version]
  81. Martín Arranz, E.; Martín Arranz, M.D.; Robredo, T.; Mancheño-Corvo, P.; Menta, R.; Alves, F.J.; Suárez de Parga, J.M.; Mora Sanz, P.; de la Rosa, O.; Büscher, D.; et al. Endoscopic submucosal injection of adipose-derived mesenchymal stem cells ameliorates TNBS-induced colitis in rats and prevents stenosis. Stem Cell Res. Ther. 2018, 9, 95. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Cheng, W.; Su, J.; Hu, Y.; Huang, Q.; Shi, H.; Wang, L.; Ren, J. Interleukin-25 primed mesenchymal stem cells achieve better therapeutic effects on dextran sulfate sodium-induced colitis via inhibiting Th17 immune response and inducing T regulatory cell phenotype. Am. J. Transl. Res. 2017, 9, 4149–4160. [Google Scholar] [PubMed]
  83. Legaki, E.; Roubelakis, M.G.; Theodoropoulos, G.E.; Lazaris, A.; Kollia, A.; Karamanolis, G.; Marinos, E.; Gazouli, M. Therapeutic Potential of Secreted Molecules Derived from Human Amniotic Fluid Mesenchymal Stem/Stroma Cells in a Mice Model of Colitis. Stem Cell Rev. Rep. 2016, 12, 604–612. [Google Scholar] [CrossRef] [PubMed]
  84. de Aguiar, C.F.; Castoldi, A.; Andrade-Oliveira, V.; Ignacio, A.; da Cunha, F.F.; Felizardo, R.J.F.; Bassi, Ê.J.; Câmara, N.O.S.; de Almeida, D.C. Mesenchymal stromal cells modulate gut inflammation in experimental colitis. Inflammopharmacology 2018, 26, 251–260. [Google Scholar] [CrossRef]
  85. Song, W.-J.; Li, Q.; Ryu, M.-O.; Ahn, J.-O.; Bhang, D.H.; Jung, Y.C.; Youn, H.-Y. TSG-6 released from intraperitoneally injected canine adipose tissue-derived mesenchymal stem cells ameliorate inflammatory bowel disease by inducing M2 macrophage switch in mice. Stem Cell Res. Ther. 2018, 9, 91. [Google Scholar] [CrossRef]
  86. Xu, X.; Wang, Y.; Zhang, B.; Lan, X.; Lu, S.; Sun, P.; Li, X.; Shi, G.; Zhao, Y.; Han, H.; et al. Treatment of experimental colitis by endometrial regenerative cells through regulation of B lymphocytes in mice. Stem Cell Res. Ther. 2018, 9, 146. [Google Scholar] [CrossRef]
  87. Miyamoto, S.; Ohnishi, S.; Onishi, R.; Tsuchiya, I.; Hosono, H.; Katsurada, T.; Yamahara, K.; Takeda, H.; Sakamoto, N. Therapeutic effects of human amnion-derived mesenchymal stem cell transplantation and conditioned medium enema in rats with trinitrobenzene sulfonic acid-induced colitis. Am. J. Transl. Res. 2017, 9, 940–952. [Google Scholar]
  88. Pak, S.; Hwang, S.W.; Shim, I.; Bae, S.; Ryu, Y.; Kim, H.-B.; Do, E.-J.; Son, H.-N.; Choi, E.-j.; Park, S.-h.; et al. Endoscopic Transplantation of Mesenchymal Stem Cell Sheets in Experimental Colitis in Rats. Sci. Rep. 2018, 8, 11314. [Google Scholar] [CrossRef]
  89. Park, H.J.; Kim, J.; Saima, F.T.; Rhee, K.J.; Hwang, S.; Kim, M.Y.; Baik, S.K.; Eom, Y.W.; Kim, H.S. Adipose-derived stem cells ameliorate colitis by suppression of inflammasome formation and regulation of M1-macrophage population through prostaglandin E2. Biochem. Biophys. Res. Commun. 2018, 498, 988–995. [Google Scholar] [CrossRef]
  90. de la Portilla, F.; Yuste, Y.; Pereira, S.; Olano, C.; Maestre, M.V.; Padillo, F.J. Local Mesenchymal Stem Cell Therapy in Experimentally Induced Colitis in the Rat. Int. J. Stem Cells 2018, 11, 39–47. [Google Scholar] [CrossRef] [Green Version]
  91. Barnhoorn, M.; de Jonge-Muller, E.; Molendijk, I.; van Gulijk, M.; Lebbink, O.; Janson, S.; Schoonderwoerd, M.; van der Helm, D.; van der Meulen-de Jong, A.; Hawinkels, L.; et al. Endoscopic Administration of Mesenchymal Stromal Cells Reduces Inflammation in Experimental Colitis. Inflamm. Bowel Dis. 2018, 24, 1755–1767. [Google Scholar] [CrossRef]
  92. Pan, X.H.; Li, Q.Q.; Zhu, X.Q.; Li, Z.A.; Cai, X.M.; Pang, R.Q.; Ruan, G.P. Mechanism and therapeutic effect of umbilical cord mesenchymal stem cells in inflammatory bowel disease. Sci. Rep. 2019, 9, 17646. [Google Scholar] [CrossRef]
  93. Ko, I.K.; Kim, B.G.; Awadallah, A.; Mikulan, J.; Lin, P.; Letterio, J.J.; Dennis, J.E. Targeting improves MSC treatment of inflammatory bowel disease. Mol. Ther. 2010, 18, 1365–1372. [Google Scholar] [CrossRef]
  94. Pérez-Merino, E.M.; Usón-Casaús, J.M.; Zaragoza-Bayle, C.; Duque-Carrasco, J.; Mariñas-Pardo, L.; Hermida-Prieto, M.; Barrera-Chacón, R.; Gualtieri, M. Safety and efficacy of allogeneic adipose tissue-derived mesenchymal stem cells for treatment of dogs with inflammatory bowel disease: Clinical and laboratory outcomes. Vet. J. 2015, 206, 385–390. [Google Scholar] [CrossRef]
  95. Cristobal, J.I.; Duque, F.J.; Uson-Casaus, J.M.; Ruiz, P.; Nieto, E.L.; Perez-Merino, E.M. Effects of Allogeneic Mesenchymal Stem Cell Transplantation in Dogs with Inflammatory Bowel Disease Treated with and without Corticosteroids. Animals 2021, 11, 2061. [Google Scholar] [CrossRef]
  96. Wang, R.; Yao, Q.; Chen, W.; Gao, F.; Li, P.; Wu, J.; Yu, J.; Cao, H. Stem cell therapy for Crohn’s disease: Systematic review and meta-analysis of preclinical and clinical studies. Stem Cell Res. Ther. 2021, 12, 463. [Google Scholar] [CrossRef]
  97. Barnhoorn, M.C.; Wasser, M.; Roelofs, H.; Maljaars, P.W.J.; Molendijk, I.; Bonsing, B.A.; Oosten, L.E.M.; Dijkstra, G.; van der Woude, C.J.; Roelen, D.L.; et al. Long-term Evaluation of Allogeneic Bone Marrow-derived Mesenchymal Stromal Cell Therapy for Crohn’s Disease Perianal Fistulas. J. Crohn’s Colitis 2020, 14, 64–70. [Google Scholar] [CrossRef]
  98. de la Portilla, F.; Alba, F.; García-Olmo, D.; Herrerías, J.M.; González, F.X.; Galindo, A. Expanded allogeneic adipose-derived stem cells (eASCs) for the treatment of complex perianal fistula in Crohn’s disease: Results from a multicenter phase I/IIa clinical trial. Int. J. Color. Dis. 2013, 28, 313–323. [Google Scholar] [CrossRef]
  99. Gregoire, C.; Briquet, A.; Pirenne, C.; Lechanteur, C.; Louis, E.; Beguin, Y. Allogeneic mesenchymal stromal cells for refractory luminal Crohn’s disease: A phase I-II study. Dig. Liver. Dis. 2018, 50, 1251–1255. [Google Scholar] [CrossRef]
  100. Heidari, N.; Abbasi-Kenarsari, H.; Namaki, S.; Baghaei, K.; Zali, M.R.; Ghaffari Khaligh, S.; Hashemi, S.M. 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, 5906–5920. [Google Scholar] [CrossRef]
  101. Hu, J.; Zhao, G.; Zhang, L.; Qiao, C.; Di, A.; Gao, H.; Xu, H. Safety and therapeutic effect of mesenchymal stem cell infusion on moderate to severe ulcerative colitis. Exp. Ther. Med. 2016, 12, 2983–2989. [Google Scholar] [CrossRef] [Green Version]
  102. Mao, F.; Wu, Y.; Tang, X.; Kang, J.; Zhang, B.; Yan, Y.; Qian, H.; Zhang, X.; Xu, W. Exosomes Derived from Human Umbilical Cord Mesenchymal Stem Cells Relieve Inflammatory Bowel Disease in Mice. Biomed Res. Int. 2017, 2017, 5356760. [Google Scholar] [CrossRef]
  103. Martin, C.; Olmos, É.; Collignon, M.-L.; De Isla, N.; Blanchard, F.; Chevalot, I.; Marc, A.; Guedon, E. Revisiting MSC expansion from critical quality attributes to critical culture process parameters. Process Biochem. 2017, 59, 231–243. [Google Scholar] [CrossRef]
  104. Molendijk, I.; Bonsing, B.A.; Roelofs, H.; Peeters, K.C.; Wasser, M.N.; Dijkstra, G.; van der Woude, C.J.; Duijvestein, M.; Veenendaal, R.A.; Zwaginga, J.J.; et al. Allogeneic Bone Marrow-Derived Mesenchymal Stromal Cells Promote Healing of Refractory Perianal Fistulas in Patients With Crohn’s Disease. Gastroenterology 2015, 149, 918–927.e916. [Google Scholar] [CrossRef] [Green Version]
  105. Panés, J.; García-Olmo, D.; Van Assche, G.; Colombel, J.F.; Reinisch, W.; Baumgart, D.C.; Dignass, A.; Nachury, M.; Ferrante, M.; Kazemi-Shirazi, L.; et al. Expanded allogeneic adipose-derived mesenchymal stem cells (Cx601) for complex perianal fistulas in Crohn’s disease: A phase 3 randomised, double-blind controlled trial. Lancet 2016, 388, 1281–1290. [Google Scholar] [CrossRef]
  106. Panés, J.; García-Olmo, D.; Van Assche, G.; Colombel, J.F.; Reinisch, W.; Baumgart, D.C.; Dignass, A.; Nachury, M.; Ferrante, M.; Kazemi-Shirazi, L.; et al. Long-term Efficacy and Safety of Stem Cell Therapy (Cx601) for Complex Perianal Fistulas in Patients With Crohn’s Disease. Gastroenterology 2018, 154, 1334–1342.e4. [Google Scholar] [CrossRef] [Green Version]
  107. Park, S.R.; Kim, J.W.; Jun, H.S.; Roh, J.Y.; Lee, H.Y.; Hong, I.S. Stem Cell Secretome and Its Effect on Cellular Mechanisms Relevant to Wound Healing. Mol. Ther. 2018, 26, 606–617. [Google Scholar] [CrossRef] [Green Version]
  108. Wang, W.Q.; Dong, K.; Zhou, L.; Jiao, G.H.; Zhu, C.Z.; Li, W.W.; Yu, G.; Wu, W.T.; Chen, S.; Sun, Z.N.; et al. IL-37b gene transfer enhances the therapeutic efficacy of mesenchumal stromal cells in DSS-induced colitis mice. Acta Pharmacol. Sin. 2015, 36, 1377–1387. [Google Scholar] [CrossRef]
  109. Wu, Y.; Qiu, W.; Xu, X.; Kang, J.; Wang, J.; Wen, Y.; Tang, X.; Yan, Y.; Qian, H.; Zhang, X.; et al. Exosomes derived from human umbilical cord mesenchymal stem cells alleviate inflammatory bowel disease in mice through ubiquitination. Am. J. Transl. Res. 2018, 10, 2026–2036. [Google Scholar]
  110. Li, M.O.; Wan, Y.Y.; Sanjabi, S.; Robertson, A.K.; Flavell, R.A. Transforming growth factor-beta regulation of immune responses. Annu. Rev. Immunol. 2006, 24, 99–146. [Google Scholar] [CrossRef]
  111. Di Nicola, M.; Carlo-Stella, C.; Magni, M.; Milanesi, M.; Longoni, P.D.; Matteucci, P.; Grisanti, S.; Gianni, A.M. Human bone marrow stromal cells suppress T-lymphocyte proliferation induced by cellular or nonspecific mitogenic stimuli. Blood 2002, 99, 3838–3843. [Google Scholar] [CrossRef] [PubMed]
  112. Aggarwal, S.; Pittenger, M.F. Human mesenchymal stem cells modulate allogeneic immune cell responses. Blood 2005, 105, 1815–1822. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Sato, K.; Ozaki, K.; Oh, I.; Meguro, A.; Hatanaka, K.; Nagai, T.; Muroi, K.; Ozawa, K. Nitric oxide plays a critical role in suppression of T-cell proliferation by mesenchymal stem cells. Blood 2007, 109, 228–234. [Google Scholar] [CrossRef] [PubMed]
  114. 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, 4619–4621. [Google Scholar] [CrossRef] [Green Version]
  115. Marlow, G.J.; van Gent, D.; Ferguson, L.R. Why interleukin-10 supplementation does not work in Crohn’s disease patients. World J. Gastroenterol. 2013, 19, 3931–3941. [Google Scholar] [CrossRef]
  116. Chen, Q.Q.; Yan, L.; Wang, C.Z.; Wang, W.H.; Shi, H.; Su, B.B.; Zeng, Q.H.; Du, H.T.; Wan, J. Mesenchymal stem cells alleviate TNBS-induced colitis by modulating inflammatory and autoimmune responses. World J. Gastroenterol. 2013, 19, 4702–4717. [Google Scholar] [CrossRef]
  117. Alves, V.B.F.; de Sousa, B.C.; Fonseca, M.T.C.; Ogata, H.; Caliári-Oliveira, C.; Yaochite, J.N.U.; Rodrigues Júnior, V.; Chica, J.E.L.; da Silva, J.S.; Malmegrim, K.C.R.; et al. A single administration of human adipose tissue-derived mesenchymal stromal cells (MSC) induces durable and sustained long-term regulation of inflammatory response in experimental colitis. Clin. Exp. Immunol. 2019, 196, 139–154. [Google Scholar] [CrossRef] [Green Version]
  118. Kim, H.S.; Shin, T.H.; Lee, B.C.; Yu, K.R.; Seo, Y.; Lee, S.; Seo, M.S.; Hong, I.S.; Choi, S.W.; Seo, K.W.; et al. Human umbilical cord blood mesenchymal stem cells reduce colitis in mice by activating NOD2 signaling to COX2. Gastroenterology 2013, 145, 1392–1403.e8. [Google Scholar] [CrossRef]
  119. Bluguermann, C.; Wu, L.; Petrigliano, F.; McAllister, D.; Miriuka, S.; Evseenko, D.A. Novel aspects of parenchymal-mesenchymal interactions: From cell types to molecules and beyond. Cell Biochem. Funct. 2013, 31, 271–280. [Google Scholar] [CrossRef]
  120. Ahangar, P.; Mills, S.J.; Cowin, A.J. Mesenchymal Stem Cell Secretome as an Emerging Cell-Free Alternative for Improving Wound Repair. Int. J. Mol. Sci. 2020, 21, 7038. [Google Scholar] [CrossRef]
  121. Shi, X.; Chen, Q.; Wang, F. Mesenchymal stem cells for the treatment of ulcerative colitis: A systematic review and meta-analysis of experimental and clinical studies. Stem Cell Res. Ther. 2019, 10, 266. [Google Scholar] [CrossRef] [Green Version]
  122. Sendon-Lago, J.; Rio, L.G.; Eiro, N.; Diaz-Rodriguez, P.; Avila, L.; Gonzalez, L.O.; Vizoso, F.J.; Perez-Fernandez, R.; Landin, M. Tailored Hydrogels as Delivery Platforms for Conditioned Medium from Mesenchymal Stem Cells in a Model of Acute Colitis in Mice. Pharmaceutics 2021, 13, 1127. [Google Scholar] [CrossRef]
  123. Pardali, E.; Sanchez-Duffhues, G.; Gomez-Puerto, M.C.; Ten Dijke, P. TGF-β-Induced Endothelial-Mesenchymal Transition in Fibrotic Diseases. Int. J. Mol. Sci. 2017, 18, 2157. [Google Scholar] [CrossRef] [Green Version]
  124. Choi, A.; Park, S.E.; Jeong, J.B.; Choi, S.J.; Oh, S.Y.; Ryu, G.H.; Lee, J.; Jeon, H.B.; Chang, J.W. Anti-Fibrotic Effect of Human Wharton’s Jelly-Derived Mesenchymal Stem Cells on Skeletal Muscle Cells, Mediated by Secretion of MMP-1. Int. J. Mol. Sci. 2020, 21, 6269. [Google Scholar] [CrossRef]
  125. Ishiuchi, N.; Nakashima, A.; Doi, S.; Yoshida, K.; Maeda, S.; Kanai, R.; Yamada, Y.; Ike, T.; Doi, T.; Kato, Y.; et al. Hypoxia-preconditioned mesenchymal stem cells prevent renal fibrosis and inflammation in ischemia-reperfusion rats. Stem Cell Res. Ther. 2020, 11, 130. [Google Scholar] [CrossRef] [Green Version]
  126. Yao, X.; Wang, J.; Zhu, J.; Rong, X. The anti-fibrotic effect of human fetal skin-derived stem cell secretome on the liver fibrosis. Stem Cell Res. Ther. 2020, 11, 379. [Google Scholar] [CrossRef]
  127. Hu, H.H.; Chen, D.Q.; Wang, Y.N.; Feng, Y.L.; Cao, G.; Vaziri, N.D.; Zhao, Y.Y. New insights into TGF-β/Smad signaling in tissue fibrosis. Chem. Biol. Interact. 2018, 292, 76–83. [Google Scholar] [CrossRef] [Green Version]
  128. Walton, K.L.; Johnson, K.E.; Harrison, C.A. Targeting TGF-β Mediated SMAD Signaling for the Prevention of Fibrosis. Front. Pharmacol. 2017, 8, 461. [Google Scholar] [CrossRef] [Green Version]
  129. Driscoll, J.; Patel, T. The mesenchymal stem cell secretome as an acellular regenerative therapy for liver disease. J. Gastroenterol. 2019, 54, 763–773. [Google Scholar] [CrossRef] [Green Version]
  130. Alfaifi, M.; Eom, Y.W.; Newsome, P.N.; Baik, S.K. Mesenchymal stromal cell therapy for liver diseases. J. Hepatol. 2018, 68, 1272–1285. [Google Scholar] [CrossRef] [Green Version]
  131. Jang, Y.J.; An, S.Y.; Kim, J.H. Identification of MFGE8 in mesenchymal stem cell secretome as an anti-fibrotic factor in liver fibrosis. BMB Rep. 2017, 50, 58–59. [Google Scholar] [CrossRef] [Green Version]
  132. Rong, X.; Liu, J.; Yao, X.; Jiang, T.; Wang, Y.; Xie, F. Human bone marrow mesenchymal stem cells-derived exosomes alleviate liver fibrosis through the Wnt/β-catenin pathway. Stem Cell Res. Ther. 2019, 10, 98. [Google Scholar] [CrossRef] [Green Version]
  133. Hu, J.; Chen, Y.; Huang, Y.; Su, Y. Human umbilical cord mesenchymal stem cell-derived exosomes suppress dermal fibroblasts-myofibroblats transition via inhibiting the TGF-β1/Smad 2/3 signaling pathway. Exp. Mol. Pathol. 2020, 115, 104468. [Google Scholar] [CrossRef]
  134. Qian, C.; Meng, Q.; Lu, J.; Zhang, L.; Li, H.; Huang, B. Human amnion mesenchymal stem cells restore spermatogenesis in mice with busulfan-induced testis toxicity by inhibiting apoptosis and oxidative stress. Stem Cell Res. Ther. 2020, 11, 290. [Google Scholar] [CrossRef]
  135. Duan, S.; Wang, F.; Cao, J.; Wang, C. Exosomes Derived from MicroRNA-146a-5p-Enriched Bone Marrow Mesenchymal Stem Cells Alleviate Intracerebral Hemorrhage by Inhibiting Neuronal Apoptosis and Microglial M1 Polarization. Drug Des. Dev. Ther. 2020, 14, 3143–3158. [Google Scholar] [CrossRef]
  136. Yang, J.; Liu, X.X.; Fan, H.; Tang, Q.; Shou, Z.X.; Zuo, D.M.; Zou, Z.; Xu, M.; Chen, Q.Y.; Peng, Y.; et al. Extracellular Vesicles Derived from Bone Marrow Mesenchymal Stem Cells Protect against Experimental Colitis via Attenuating Colon Inflammation, Oxidative Stress and Apoptosis. PLoS ONE 2015, 10, e0140551. [Google Scholar] [CrossRef] [Green Version]
  137. Stavely, R.; Nurgali, K. The emerging antioxidant paradigm of mesenchymal stem cell therapy. Stem Cells Transl. Med. 2020, 9, 985–1006. [Google Scholar] [CrossRef]
  138. Sies, H. Oxidative stress: A concept in Redox Biology and medicine. Redox Biol. 2015, 4, 180–183. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  139. Birben, E.; Sahiner, U.M.; Sackesen, C.; Erzurum, S.; Kalayci, O. Oxidative stress and antioxidant defense. World Allergy Organ. J. 2012, 5, 9–19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  140. Schieber, M.; Chandel, N.S. ROS function in redox signaling and oxidative stress. Curr. Biol. 2014, 24, R453–R462. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  141. Tian, T.; Wang, Z.; Zhang, J. Pathomechanisms of Oxidative Stress in Inflammatory Bowel Disease and Potential Antioxidant Therapies. Oxidative Med. Cell. Longev. 2017, 2017, 4535194. [Google Scholar] [CrossRef]
  142. Kim, Y.W.; West, X.Z.; Byzova, T.V. Inflammation and oxidative stress in angiogenesis and vascular disease. J. Mol. Med. 2013, 91, 323–328. [Google Scholar] [CrossRef] [Green Version]
  143. Zhang, R.; Brennan, M.L.; Shen, Z.; MacPherson, J.C.; Schmitt, D.; Molenda, C.E.; Hazen, S.L. Myeloperoxidase functions as a major enzymatic catalyst for initiation of lipid peroxidation at sites of inflammation. J. Biol. Chem. 2002, 277, 46116–46122. [Google Scholar] [CrossRef] [Green Version]
  144. Pattison, D.I.; Davies, M.J. Reactions of myeloperoxidase-derived oxidants with biological substrates: Gaining chemical insight into human inflammatory diseases. Curr. Med. Chem. 2006, 13, 3271–3290. [Google Scholar] [CrossRef]
  145. Valle-Prieto, A.; Conget, P.A. Human mesenchymal stem cells efficiently manage oxidative stress. Stem Cells Dev. 2010, 19, 1885–1893. [Google Scholar] [CrossRef]
  146. Gorbunov, N.V.; Garrison, B.R.; McDaniel, D.P.; Zhai, M.; Liao, P.J.; Nurmemet, D.; Kiang, J.G. Adaptive redox response of mesenchymal stromal cells to stimulation with lipopolysaccharide inflammagen: Mechanisms of remodeling of tissue barriers in sepsis. Oxidative Med. Cell. Longev. 2013, 2013, 186795. [Google Scholar] [CrossRef]
  147. Guillén, M.I.; Platas, J.; Pérez Del Caz, M.D.; Mirabet, V.; Alcaraz, M.J. Paracrine Anti-inflammatory Effects of Adipose Tissue-Derived Mesenchymal Stem Cells in Human Monocytes. Front. Physiol. 2018, 9, 661. [Google Scholar] [CrossRef]
  148. Oh, J.Y.; Ko, J.H.; Lee, H.J.; Yu, J.M.; Choi, H.; Kim, M.K.; Wee, W.R.; Prockop, D.J. Mesenchymal stem/stromal cells inhibit the NLRP3 inflammasome by decreasing mitochondrial reactive oxygen species. Stem Cells 2014, 32, 1553–1563. [Google Scholar] [CrossRef]
  149. Wu, X.; Dao Thi, V.L.; Huang, Y.; Billerbeck, E.; Saha, D.; Hoffmann, H.H.; Wang, Y.; Silva, L.A.V.; Sarbanes, S.; Sun, T.; et al. Intrinsic Immunity Shapes Viral Resistance of Stem Cells. Cell 2018, 172, 423–438.e425. [Google Scholar] [CrossRef]
  150. Bailey, C.C.; Zhong, G.; Huang, I.C.; Farzan, M. IFITM-Family Proteins: The Cell’s First Line of Antiviral Defense. Annu. Rev. Virol. 2014, 1, 261–283. [Google Scholar] [CrossRef] [Green Version]
  151. Zhang, L.J.; Gallo, R.L. Antimicrobial peptides. Curr. Biol. 2016, 26, R14–R19. [Google Scholar] [CrossRef]
  152. Yagi, H.; Chen, A.F.; Hirsch, D.; Rothenberg, A.C.; Tan, J.; Alexander, P.G.; Tuan, R.S. Antimicrobial activity of mesenchymal stem cells against Staphylococcus aureus. Stem Cell Res. Ther. 2020, 11, 293. [Google Scholar] [CrossRef]
  153. Brogden, K.A. Antimicrobial peptides: Pore formers or metabolic inhibitors in bacteria? Nat. Rev. Microbiol. 2005, 3, 238–250. [Google Scholar] [CrossRef] [PubMed]
  154. Alcayaga-Miranda, F.; Cuenca, J.; Khoury, M. Antimicrobial Activity of Mesenchymal Stem Cells: Current Status and New Perspectives of Antimicrobial Peptide-Based Therapies. Front. Immunol. 2017, 8, 339. [Google Scholar] [CrossRef] [Green Version]
  155. Marrazzo, P.; Crupi, A.N.; Alviano, F.; Teodori, L.; Bonsi, L. Exploring the roles of MSCs in infections: Focus on bacterial diseases. J. Mol. Med. 2019, 97, 437–450. [Google Scholar] [CrossRef] [PubMed]
  156. Agerberth, B.; Charo, J.; Werr, J.; Olsson, B.; Idali, F.; Lindbom, L.; Kiessling, R.; Jörnvall, H.; Wigzell, H.; Gudmundsson, G.H. The human antimicrobial and chemotactic peptides LL-37 and alpha-defensins are expressed by specific lymphocyte and monocyte populations. Blood 2000, 96, 3086–3093. [Google Scholar] [CrossRef] [PubMed]
  157. Wang, G.; Narayana, J.L.; Mishra, B.; Zhang, Y.; Wang, F.; Wang, C.; Zarena, D.; Lushnikova, T.; Wang, X. Design of Antimicrobial Peptides: Progress Made with Human Cathelicidin LL-37. Adv. Exp. Med. Biol. 2019, 1117, 215–240. [Google Scholar] [CrossRef]
  158. Xhindoli, D.; Pacor, S.; Benincasa, M.; Scocchi, M.; Gennaro, R.; Tossi, A. The human cathelicidin LL-37--A pore-forming antibacterial peptide and host-cell modulator. Biochim. Biophys. Acta 2016, 1858, 546–566. [Google Scholar] [CrossRef]
  159. De Smet, K.; Contreras, R. Human antimicrobial peptides: Defensins, cathelicidins and histatins. Biotechnol. Lett. 2005, 27, 1337–1347. [Google Scholar] [CrossRef]
  160. López-García, B.; Lee, P.H.; Yamasaki, K.; Gallo, R.L. Anti-fungal activity of cathelicidins and their potential role in Candida albicans skin infection. J. Investig. Dermatol. 2005, 125, 108–115. [Google Scholar] [CrossRef]
  161. Bergman, P.; Walter-Jallow, L.; Broliden, K.; Agerberth, B.; Söderlund, J. The antimicrobial peptide LL-37 inhibits HIV-1 replication. Curr. HIV Res. 2007, 5, 410–415. [Google Scholar] [CrossRef]
  162. Rivas-Santiago, B.; Hernandez-Pando, R.; Carranza, C.; Juarez, E.; Contreras, J.L.; Aguilar-Leon, D.; Torres, M.; Sada, E. Expression of cathelicidin LL-37 during Mycobacterium tuberculosis infection in human alveolar macrophages, monocytes, neutrophils, and epithelial cells. Infect. Immun. 2008, 76, 935–941. [Google Scholar] [CrossRef] [Green Version]
  163. Alcayaga-Miranda, F.; Cuenca, J.; Martin, A.; Contreras, L.; Figueroa, F.E.; Khoury, M. Combination therapy of menstrual derived mesenchymal stem cells and antibiotics ameliorates survival in sepsis. Stem Cell Res. Ther. 2015, 6, 199. [Google Scholar] [CrossRef] [Green Version]
  164. Meisel, R.; Brockers, S.; Heseler, K.; Degistirici, O.; Bülle, H.; Woite, C.; Stuhlsatz, S.; Schwippert, W.; Jäger, M.; Sorg, R.; et al. Human but not murine multipotent mesenchymal stromal cells exhibit broad-spectrum antimicrobial effector function mediated by indoleamine 2,3-dioxygenase. Leukemia 2011, 25, 648–654. [Google Scholar] [CrossRef]
  165. Yang, R.; Liu, Y.; Kelk, P.; Qu, C.; Akiyama, K.; Chen, C.; Atsuta, I.; Chen, W.; Zhou, Y.; Shi, S. A subset of IL-17(+) mesenchymal stem cells possesses anti-Candida albicans effect. Cell Res. 2013, 23, 107–121. [Google Scholar] [CrossRef] [Green Version]
  166. Bermudez, M.A.; Sendon-Lago, J.; Eiro, N.; Treviño, M.; Gonzalez, F.; Yebra-Pimentel, E.; Giraldez, M.J.; Macia, M.; Lamelas, M.L.; Saa, J.; et al. Corneal epithelial wound healing and bactericidal effect of conditioned medium from human uterine cervical stem cells. Investig. Ophthalmol. Vis. Sci. 2015, 56, 983–992. [Google Scholar] [CrossRef]
  167. Saberpour, M.; Bakhshi, B.; Najar-Peerayeh, S. Evaluation of the Antimicrobial and Antibiofilm Effect of Chitosan Nanoparticles as Carrier for Supernatant of Mesenchymal Stem Cells on Multidrug-Resistant Vibrio cholerae. Infect. Drug Resist. 2020, 13, 2251–2260. [Google Scholar] [CrossRef]
  168. Krasnodembskaya, A.; Samarani, G.; Song, Y.; Zhuo, H.; Su, X.; Lee, J.W.; Gupta, N.; Petrini, M.; Matthay, M.A. Human mesenchymal stem cells reduce mortality and bacteremia in gram-negative sepsis in mice in part by enhancing the phagocytic activity of blood monocytes. Am. J. Physiol. Lung Cell. Mol. Physiol. 2012, 302, L1003–L1013. [Google Scholar] [CrossRef] [Green Version]
  169. Skrahin, A.; Jenkins, H.E.; Hurevich, H.; Solodovnikova, V.; Isaikina, Y.; Klimuk, D.; Rohava, Z.; Skrahina, A. Effectiveness of a novel cellular therapy to treat multidrug-resistant tuberculosis. J. Clin. Tuberc. Other Mycobact. Dis. 2016, 4, 21–27. [Google Scholar] [CrossRef] [Green Version]
  170. Harman, R.M.; Yang, S.; He, M.K.; Van de Walle, G.R. Antimicrobial peptides secreted by equine mesenchymal stromal cells inhibit the growth of bacteria commonly found in skin wounds. Stem Cell Res. Ther. 2017, 8, 157. [Google Scholar] [CrossRef]
  171. Schneider, J.; Mateo, E.; Marcos-Arias, C.; Eiró, N.; Vizoso, F.; Pérez-Fernández, R.; Eraso, E.; Quindós, G. Antifungal Activity of the Human Uterine Cervical Stem Cells Conditioned Medium (hUCESC-CM) Against Candida albicans and Other Medically Relevant Species of Candida. Front. Microbiol. 2018, 9, 2818, Correction in Front. Microbiol. 2019, 10, 1297. [Google Scholar] [CrossRef] [PubMed]
  172. Chow, L.; Johnson, V.; Impastato, R.; Coy, J.; Strumpf, A.; Dow, S. Antibacterial activity of human mesenchymal stem cells mediated directly by constitutively secreted factors and indirectly by activation of innate immune effector cells. Stem Cells Transl. Med. 2020, 9, 235–249. [Google Scholar] [CrossRef] [Green Version]
  173. Marx, C.; Gardner, S.; Harman, R.M.; Van de Walle, G.R. The mesenchymal stromal cell secretome impairs methicillin-resistant Staphylococcus aureus biofilms via cysteine protease activity in the equine model. Stem Cells Transl. Med. 2020, 9, 746–757. [Google Scholar] [CrossRef] [Green Version]
  174. Rhee, K.J.; Lee, J.I.; Eom, Y.W. Mesenchymal Stem Cell-Mediated Effects of Tumor Support or Suppression. Int. J. Mol. Sci. 2015, 16, 30015–30033. [Google Scholar] [CrossRef] [Green Version]
  175. Lin, W.; Huang, L.; Li, Y.; Fang, B.; Li, G.; Chen, L.; Xu, L. Mesenchymal Stem Cells and Cancer: Clinical Challenges and Opportunities. Biomed Res. Int. 2019, 2019, 2820853. [Google Scholar] [CrossRef] [Green Version]
  176. Costa, L.A.; Eiro, N.; Fraile, M.; Gonzalez, L.O.; Saá, J.; Garcia-Portabella, P.; Vega, B.; Schneider, J.; Vizoso, F.J. Functional heterogeneity of mesenchymal stem cells from natural niches to culture conditions: Implications for further clinical uses. Cell. Mol. Life Sci. 2021, 78, 447–467. [Google Scholar] [CrossRef]
  177. Lozito, T.P.; Tuan, R.S. Mesenchymal stem cells inhibit both endogenous and exogenous MMPs via secreted TIMPs. J. Cell. Physiol 2011, 226, 385–396. [Google Scholar] [CrossRef]
  178. Leng, L.; Wang, Y.; He, N.; Wang, D.; Zhao, Q.; Feng, G.; Su, W.; Xu, Y.; Han, Z.; Kong, D.; et al. Molecular imaging for assessment of mesenchymal stem cells mediated breast cancer therapy. Biomaterials 2014, 35, 5162–5170. [Google Scholar] [CrossRef] [Green Version]
  179. Ren, C.; Kumar, S.; Chanda, D.; Chen, J.; Mountz, J.D.; Ponnazhagan, S. Therapeutic potential of mesenchymal stem cells producing interferon-alpha in a mouse melanoma lung metastasis model. Stem Cells 2008, 26, 2332–2338. [Google Scholar] [CrossRef] [Green Version]
  180. Eiró, N.; Sendon-Lago, J.; Seoane, S.; Bermúdez, M.A.; Lamelas, M.L.; Garcia-Caballero, T.; Schneider, J.; Perez-Fernandez, R.; Vizoso, F.J. Potential therapeutic effect of the secretome from human uterine cervical stem cells against both cancer and stromal cells compared with adipose tissue stem cells. Oncotarget 2014, 5, 10692–10708. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  181. Ueda, G.; Shimizu, C.; Saito, J.; Tanaka, Y.; Inoue, M.; Tanizawa, O. An immunohistochemical study of colon-ovarian tumor antigen and colon-specific antigen in gynecologic tumors. Gynecol. Oncol. 1989, 35, 90–92. [Google Scholar] [CrossRef]
  182. Greco, K.A.; Franzen, C.A.; Foreman, K.E.; Flanigan, R.C.; Kuo, P.C.; Gupta, G.N. PLK-1 Silencing in Bladder Cancer by siRNA Delivered with Exosomes. Urology 2016, 91, 241.e1–241.e7. [Google Scholar] [CrossRef]
  183. Smyth, T.J.; Redzic, J.S.; Graner, M.W.; Anchordoquy, T.J. Examination of the specificity of tumor cell derived exosomes with tumor cells in vitro. Biochim. Biophys. Acta 2014, 1838, 2954–2965. [Google Scholar] [CrossRef] [Green Version]
  184. Wu, S.; Ju, G.Q.; Du, T.; Zhu, Y.J.; Liu, G.H. Microvesicles derived from human umbilical cord Wharton’s jelly mesenchymal stem cells attenuate bladder tumor cell growth in vitro and in vivo. PLoS ONE 2013, 8, e61366. [Google Scholar] [CrossRef]
  185. Reza, A.; Choi, Y.J.; Yasuda, H.; Kim, J.H. Human adipose mesenchymal stem cell-derived exosomal-miRNAs are critical factors for inducing anti-proliferation signalling to A2780 and SKOV-3 ovarian cancer cells. Sci. Rep. 2016, 6, 38498. [Google Scholar] [CrossRef]
  186. Vendramini-Costa, D.B.; Carvalho, J.E. Molecular link mechanisms between inflammation and cancer. Curr. Pharm. Des. 2012, 18, 3831–3852. [Google Scholar] [CrossRef]
  187. Wang, D.; DuBois, R.N. The role of anti-inflammatory drugs in colorectal cancer. Annu. Rev. Med. 2013, 64, 131–144. [Google Scholar] [CrossRef]
  188. Chen, Z.; He, X.; He, X.; Chen, X.; Lin, X.; Zou, Y.; Wu, X.; Lan, P. Bone marrow mesenchymal stem cells ameliorate colitis-associated tumorigenesis in mice. Biochem. Biophys. Res. Commun. 2014, 450, 1402–1408. [Google Scholar] [CrossRef]
  189. Nasuno, M.; Arimura, Y.; Nagaishi, K.; Isshiki, H.; Onodera, K.; Nakagaki, S.; Watanabe, S.; Idogawa, M.; Yamashita, K.; Naishiro, Y.; et al. Mesenchymal stem cells cancel azoxymethane-induced tumor initiation. Stem Cells 2014, 32, 913–925. [Google Scholar] [CrossRef]
  190. Tang, R.J.; Shen, S.N.; Zhao, X.Y.; Nie, Y.Z.; Xu, Y.J.; Ren, J.; Lv, M.M.; Hou, Y.Y.; Wang, T.T. Mesenchymal stem cells-regulated Treg cells suppress colitis-associated colorectal cancer. Stem Cell Res. Ther. 2015, 6, 71. [Google Scholar] [CrossRef] [Green Version]
  191. De Robertis, M.; Massi, E.; Poeta, M.L.; Carotti, S.; Morini, S.; Cecchetelli, L.; Signori, E.; Fazio, V.M. The AOM/DSS murine model for the study of colon carcinogenesis: From pathways to diagnosis and therapy studies. J. Carcinog. 2011, 10, 9. [Google Scholar] [CrossRef]
  192. Koh, G.Y.; Kane, A.; Lee, K.; Xu, Q.; Wu, X.; Roper, J.; Mason, J.B.; Crott, J.W. Parabacteroides distasonis attenuates toll-like receptor 4 signaling and Akt activation and blocks colon tumor formation in high-fat diet-fed azoxymethane-treated mice. Int. J. Cancer 2018, 143, 1797–1805. [Google Scholar] [CrossRef] [Green Version]
  193. Zhang, X.; Hu, X.; Rao, X. Apoptosis induced by Staphylococcus aureus toxins. Microbiol. Res. 2017, 205, 19–24. [Google Scholar] [CrossRef]
  194. Nakatsuji, T.; Chen, T.H.; Butcher, A.M.; Trzoss, L.L.; Nam, S.J.; Shirakawa, K.T.; Zhou, W.; Oh, J.; Otto, M.; Fenical, W.; et al. A commensal strain of Staphylococcus epidermidis protects against skin neoplasia. Sci. Adv. 2018, 4, eaao4502. [Google Scholar] [CrossRef] [Green Version]
  195. Jostins, L.; Ripke, S.; Weersma, R.K.; Duerr, R.H.; McGovern, D.P.; Hui, K.Y.; Lee, J.C.; Schumm, L.P.; Sharma, Y.; Anderson, C.A.; et al. Host-microbe interactions have shaped the genetic architecture of inflammatory bowel disease. Nature 2012, 491, 119–124. [Google Scholar] [CrossRef] [Green Version]
  196. Wang, L.T.; Liu, K.J.; Sytwu, H.K.; Yen, M.L.; Yen, B.L. Advances in mesenchymal stem cell therapy for immune and inflammatory diseases: Use of cell-free products and human pluripotent stem cell-derived mesenchymal stem cells. Stem Cells Transl. Med. 2021, 10, 1288–1303. [Google Scholar] [CrossRef]
  197. Mahadev, S.; Young, J.M.; Selby, W.; Solomon, M.J. Quality of life in perianal Crohn’s disease: What do patients consider important? Dis. Colon Rectum 2011, 54, 579–585. [Google Scholar] [CrossRef]
  198. Panés, J.; Rimola, J. Perianal fistulizing Crohn’s disease: Pathogenesis, diagnosis and therapy. Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 652–664. [Google Scholar] [CrossRef]
  199. García-Olmo, D.; García-Arranz, M.; García, L.G.; Cuellar, E.S.; Blanco, I.F.; Prianes, L.A.; Montes, J.A.; Pinto, F.L.; Marcos, D.H.; García-Sancho, L. Autologous stem cell transplantation for treatment of rectovaginal fistula in perianal Crohn’s disease: A new cell-based therapy. Int. J. Color. Dis. 2003, 18, 451–454. [Google Scholar] [CrossRef]
  200. García-Olmo, D.; García-Arranz, M.; Herreros, D.; Pascual, I.; Peiro, C.; Rodríguez-Montes, J.A. A phase I clinical trial of the treatment of Crohn’s fistula by adipose mesenchymal stem cell transplantation. Dis. Colon Rectum 2005, 48, 1416–1423. [Google Scholar] [CrossRef] [PubMed]
  201. Garcia-Olmo, D.; Herreros, D.; Pascual, I.; Pascual, J.A.; Del-Valle, E.; Zorrilla, J.; De-La-Quintana, P.; Garcia-Arranz, M.; Pascual, M. Expanded adipose-derived stem cells for the treatment of complex perianal fistula: A phase II clinical trial. Dis. Colon Rectum 2009, 52, 79–86. [Google Scholar] [CrossRef] [PubMed]
  202. Guadalajara, H.; Herreros, D.; De-La-Quintana, P.; Trebol, J.; Garcia-Arranz, M.; Garcia-Olmo, D. Long-term follow-up of patients undergoing adipose-derived adult stem cell administration to treat complex perianal fistulas. Int. J. Color. Dis. 2012, 27, 595–600. [Google Scholar] [CrossRef] [PubMed]
  203. Ciccocioppo, R.; Bernardo, M.E.; Sgarella, A.; Maccario, R.; Avanzini, M.A.; Ubezio, C.; Minelli, A.; Alvisi, C.; Vanoli, A.; Calliada, F.; et al. Autologous bone marrow-derived mesenchymal stromal cells in the treatment of fistulising Crohn’s disease. Gut 2011, 60, 788–798. [Google Scholar] [CrossRef]
  204. Ciccocioppo, R.; Gallia, A.; Sgarella, A.; Kruzliak, P.; Gobbi, P.G.; Corazza, G.R. Long-Term Follow-Up of Crohn Disease Fistulas After Local Injections of Bone Marrow-Derived Mesenchymal Stem Cells. Mayo Clin. Proc. 2015, 90, 747–755. [Google Scholar] [CrossRef]
  205. Park, K.J.; Ryoo, S.B.; Kim, J.S.; Kim, T.I.; Baik, S.H.; Kim, H.J.; Lee, K.Y.; Kim, M.; Kim, W.H. Allogeneic adipose-derived stem cells for the treatment of perianal fistula in Crohn’s disease: A pilot clinical trial. Color. Dis. 2016, 18, 468–476. [Google Scholar] [CrossRef]
  206. Garcia-Arranz, M.; Garcia-Olmo, D.; Herreros, M.D.; Gracia-Solana, J.; Guadalajara, H.; de la Portilla, F.; Baixauli, J.; Garcia-Garcia, J.; Ramirez, J.M.; Sanchez-Guijo, F.; et al. Autologous adipose-derived stem cells for the treatment of complex cryptoglandular perianal fistula: A randomized clinical trial with long-term follow-up. Stem Cells Transl. Med. 2020, 9, 295–301. [Google Scholar] [CrossRef] [Green Version]
  207. Choi, S.; Jeon, B.G.; Chae, G.; Lee, S.J. The clinical efficacy of stem cell therapy for complex perianal fistulas: A meta-analysis. Tech. Coloproctol. 2019, 23, 411–427. [Google Scholar] [CrossRef]
  208. Cheng, F.; Huang, Z.; Li, Z. Efficacy and Safety of Mesenchymal Stem Cells in Treatment of Complex Perianal Fistulas: A Meta-Analysis. Stem Cells Int. 2020, 2020, 8816737. [Google Scholar] [CrossRef]
  209. Duijvestein, M.; Vos, A.C.; Roelofs, H.; Wildenberg, M.E.; Wendrich, B.B.; Verspaget, H.W.; Kooy-Winkelaar, E.M.; Koning, F.; Zwaginga, J.J.; Fidder, H.H.; et al. Autologous bone marrow-derived mesenchymal stromal cell treatment for refractory luminal Crohn’s disease: Results of a phase I study. Gut 2010, 59, 1662–1669. [Google Scholar] [CrossRef]
  210. Dhere, T.; Copland, I.; Garcia, M.; Chiang, K.Y.; Chinnadurai, R.; Prasad, M.; Galipeau, J.; Kugathasan, S. The safety of autologous and metabolically fit bone marrow mesenchymal stromal cells in medically refractory Crohn’s disease—A phase 1 trial with three doses. Aliment. Pharmacol. Ther. 2016, 44, 471–481. [Google Scholar] [CrossRef] [Green Version]
  211. Liang, J.; Zhang, H.; Wang, D.; Feng, X.; Wang, H.; Hua, B.; Liu, B.; Sun, L. Allogeneic mesenchymal stem cell transplantation in seven patients with refractory inflammatory bowel disease. Gut 2012, 61, 468–469. [Google Scholar] [CrossRef]
  212. Forbes, G.M.; Sturm, M.J.; Leong, R.W.; Sparrow, M.P.; Segarajasingam, D.; Cummins, A.G.; Phillips, M.; Herrmann, R.P. A phase 2 study of allogeneic mesenchymal stromal cells for luminal Crohn’s disease refractory to biologic therapy. Clin. Gastroenterol. Hepatol. 2014, 12, 64–71. [Google Scholar] [CrossRef]
  213. Onken, J.; Gallup, D.; Hanson, J.; Pandak, M.; Custer, L. Successful outpatient treatment of refractory Crohn’s disease using adult mesenchymal stem cells. In Proceedings of the American College of Gastroenterology Conference, Las Vegas, NV, USA, 20–25 October 2006. [Google Scholar]
  214. Mayer, L.; Pandak, W.M.; Melmed, G.Y.; Hanauer, S.B.; Johnson, K.; Payne, D.; Faleck, H.; Hariri, R.J.; Fischkoff, S.A. Safety and tolerability of human placenta-derived cells (PDA001) in treatment-resistant crohn’s disease: A phase 1 study. Inflamm. Bowel Dis. 2013, 19, 754–760. [Google Scholar] [CrossRef] [Green Version]
  215. Zhang, J.; Lv, S.; Liu, X.; Song, B.; Shi, L. Umbilical Cord Mesenchymal Stem Cell Treatment for Crohn’s Disease: A Randomized Controlled Clinical Trial. Gut Liver 2018, 12, 73–78. [Google Scholar] [CrossRef]
  216. Otagiri, S.; Ohnishi, S.; Miura, A.; Hayashi, H.; Kumagai, I.; Ito, Y.M.; Katsurada, T.; Nakamura, S.; Okamoto, R.; Yamahara, K.; et al. Evaluation of amnion-derived mesenchymal stem cells for treatment-resistant moderate Crohn’s disease: Study protocol for a phase I/II, dual-centre, open-label, uncontrolled, dose-response trial. BMJ Open Gastroenterol. 2018, 5, e000206. [Google Scholar] [CrossRef]
  217. Lazebnik, L.B.; Kniazev, O.V.; Konopliannikov, A.G.; Parfenov, A.I.; Ruchkina, I.N.; Mikhaĭlova, Z.F.; Tsaregorodtseva, T.M.; Khomeriki, S.G.; Rogozina, V.A.; Gudkova, R.B.; et al. Allogeneic mesenchymal stromal cells in patients with ulcerative colitis: Two years of observation. Eksp. Klin. Gastroenterol. 2010, 3–15. [Google Scholar]
  218. Fadeeva, N.; Knyazev, O.; Kagramanova, A.; Babayan, A.; Lishchinskaya, A.; Ruchkina, I.; Konoplyannikov, A.; Parfenov, A. P413 Relative frequency of relapses in patients with ulcerative colitis and Crohn’s disease treated with mesenchymal stromal cells: Five-years of follow-up. J. Crohn’s Colitis 2018, 12, S313. [Google Scholar] [CrossRef]
  219. Lazebnik, L.; Knyazev, O.; Sagynbaeva, V.; Parfenov, A. Laboratory prediction of the effectiveness of transplantation of allogeneic mesenchymal stromal cells of bone marrow in patients with ulcerative colitis.: P-131. Inflamm. Bowel Dis. 2011, 17, S51. [Google Scholar] [CrossRef]
  220. Urbanelli, L.; Buratta, S.; Sagini, K.; Ferrara, G.; Lanni, M.; Emiliani, C. Exosome-based strategies for Diagnosis and Therapy. Recent Pat. CNS Drug Discov. 2015, 10, 10–27. [Google Scholar] [CrossRef]
  221. Eliopoulos, N.; Stagg, J.; Lejeune, L.; Pommey, S.; Galipeau, J. Allogeneic marrow stromal cells are immune rejected by MHC class I- and class II-mismatched recipient mice. Blood 2005, 106, 4057–4065. [Google Scholar] [CrossRef]
  222. Poncelet, A.J.; Nizet, Y.; Vercruysse, J.; Hiel, A.L.; Saliez, A.; Gianello, P. Inhibition of humoral response to allogeneic porcine mesenchymal stem cell with 12 days of tacrolimus. Transplantation 2008, 86, 1586–1595. [Google Scholar] [CrossRef]
  223. Markovic, B.S.; Kanjevac, T.; Harrell, C.R.; Gazdic, M.; Fellabaum, C.; Arsenijevic, N.; Volarevic, V. Molecular and Cellular Mechanisms Involved in Mesenchymal Stem Cell-Based Therapy of Inflammatory Bowel Diseases. Stem Cell Rev. 2018, 14, 153–165. [Google Scholar] [CrossRef]
  224. Gregoire, C.; Lechanteur, C.; Briquet, A.; Baudoux, E.; Baron, F.; Louis, E.; Beguin, Y. Review article: Mesenchymal stromal cell therapy for inflammatory bowel diseases. Aliment. Pharmacol. Ther. 2017, 45, 205–221. [Google Scholar] [CrossRef] [Green Version]
  225. Touboul, T.; Hannan, N.R.; Corbineau, S.; Martinez, A.; Martinet, C.; Branchereau, S.; Mainot, S.; Strick-Marchand, H.; Pedersen, R.; Di Santo, J.; et al. Generation of functional hepatocytes from human embryonic stem cells under chemically defined conditions that recapitulate liver development. Hepatology 2010, 51, 1754–1765. [Google Scholar] [CrossRef]
  226. Martinez-Montiel Mdel, P.; Gomez-Gomez, G.J.; Flores, A.I. Therapy with stem cells in inflammatory bowel disease. World J. Gastroenterol. 2014, 20, 1211–1227. [Google Scholar] [CrossRef]
  227. Kim, N.; Cho, S.G. New strategies for overcoming limitations of mesenchymal stem cell-based immune modulation. Int. J. Stem Cells 2015, 8, 54–68. [Google Scholar] [CrossRef] [Green Version]
  228. Lee, J.M.; Jung, J.; Lee, H.J.; Jeong, S.J.; Cho, K.J.; Hwang, S.G.; Kim, G.J. Comparison of immunomodulatory effects of placenta mesenchymal stem cells with bone marrow and adipose mesenchymal stem cells. Int. Immunopharmacol. 2012, 13, 219–224. [Google Scholar] [CrossRef]
  229. Ankrum, J.; Karp, J.M. Mesenchymal stem cell therapy: Two steps forward, one step back. Trends Mol. Med. 2010, 16, 203–209. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  230. López-Beas, J.; Guadix, J.A.; Clares, B.; Soriano-Ruiz, J.L.; Zugaza, J.L.; Gálvez-Martín, P. An overview of international regulatory frameworks for mesenchymal stromal cell-based medicinal products: From laboratory to patient. Med. Res. Rev. 2020, 40, 1315–1334. [Google Scholar] [CrossRef]
  231. FDA Rejects Mesoblast Flagship Treatment. 2020. Available online: https://www.pharmamanufacturing.com/industrynews/2020/fda-rejects-mesoblast-flagship-treatment/ (accessed on 2 July 2022).
  232. Lipsitz, Y.Y.; Timmins, N.E.; Zandstra, P.W. Quality cell therapy manufacturing by design. Nat. Biotechnol. 2016, 34, 393–400. [Google Scholar] [CrossRef] [PubMed]
  233. Temple, S.; Studer, L. Lessons Learned from Pioneering Neural Stem Cell Studies. Stem Cell Rep. 2017, 8, 191–193. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  234. Nakanishi, C.; Nagaya, N.; Ohnishi, S.; Yamahara, K.; Takabatake, S.; Konno, T.; Hayashi, K.; Kawashiri, M.A.; Tsubokawa, T.; Yamagishi, M. Gene and protein expression analysis of mesenchymal stem cells derived from rat adipose tissue and bone marrow. Circ. J. 2011, 75, 2260–2268. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  235. Al-Nbaheen, M.; Vishnubalaji, R.; Ali, D.; Bouslimi, A.; Al-Jassir, F.; Megges, M.; Prigione, A.; Adjaye, J.; Kassem, M.; Aldahmash, A. Human stromal (mesenchymal) stem cells from bone marrow, adipose tissue and skin exhibit differences in molecular phenotype and differentiation potential. Stem Cell Rev. Rep. 2013, 9, 32–43. [Google Scholar] [CrossRef] [Green Version]
  236. Wu, Z.; Zhang, S.; Zhou, L.; Cai, J.; Tan, J.; Gao, X.; Zeng, Z.; Li, D. Thromboembolism Induced by Umbilical Cord Mesenchymal Stem Cell Infusion: A Report of Two Cases and Literature Review. Transplant. Proc. 2017, 49, 1656–1658. [Google Scholar] [CrossRef]
  237. Coppin, L.; Sokal, E.; Stephenne, X. Thrombogenic Risk Induced by Intravascular Mesenchymal Stem Cell Therapy: Current Status and Future Perspectives. Cells 2019, 8, 1160. [Google Scholar] [CrossRef] [Green Version]
  238. Wang, Y.; Yi, H.; Song, Y. The safety of MSC therapy over the past 15 years: A meta-analysis. Stem Cell Res. Ther. 2021, 12, 545. [Google Scholar] [CrossRef]
  239. Musial-Wysocka, A.; Kot, M.; Majka, M. The Pros and Cons of Mesenchymal Stem Cell-Based Therapies. Cell Transplant. 2019, 28, 801–812. [Google Scholar] [CrossRef] [Green Version]
  240. Barekzai, J.; Petry, F.; Zitzmann, J.; Czermak, P.; Salzig, D. Bioprocess Development for Human Mesenchymal Stem Cell Therapy Products. New Adv. Ferment. Processes 2020, 1–25. [Google Scholar] [CrossRef] [Green Version]
  241. Szychlinska, M.A.; Stoddart, M.J.; D’Amora, U.; Ambrosio, L.; Alini, M.; Musumeci, G. Mesenchymal Stem Cell-Based Cartilage Regeneration Approach and Cell Senescence: Can We Manipulate Cell Aging and Function? Tissue Eng. Part B Rev. 2017, 23, 529–539. [Google Scholar] [CrossRef]
  242. Knuth, C.A.; Kiernan, C.H.; Palomares Cabeza, V.; Lehmann, J.; Witte-Bouma, J.; Ten Berge, D.; Brama, P.A.; Wolvius, E.B.; Strabbing, E.M.; Koudstaal, M.J.; et al. Isolating Pediatric Mesenchymal Stem Cells with Enhanced Expansion and Differentiation Capabilities. Tissue Eng. Part C Methods 2018, 24, 313–321. [Google Scholar] [CrossRef]
  243. Yang, Y.K.; Ogando, C.R.; Wang See, C.; Chang, T.Y.; Barabino, G.A. Changes in phenotype and differentiation potential of human mesenchymal stem cells aging in vitro. Stem Cell Res. Ther. 2018, 9, 131. [Google Scholar] [CrossRef] [Green Version]
  244. Bourgine, P.; Le Magnen, C.; Pigeot, S.; Geurts, J.; Scherberich, A.; Martin, I. Combination of immortalization and inducible death strategies to generate a human mesenchymal stromal cell line with controlled survival. Stem Cell Res. 2014, 12, 584–598. [Google Scholar] [CrossRef] [Green Version]
  245. Skårn, M.; Noordhuis, P.; Wang, M.Y.; Veuger, M.; Kresse, S.H.; Egeland, E.V.; Micci, F.; Namløs, H.M.; Håkelien, A.M.; Olafsrud, S.M.; et al. Generation and characterization of an immortalized human mesenchymal stromal cell line. Stem Cells Dev. 2014, 23, 2377–2389. [Google Scholar] [CrossRef] [Green Version]
  246. Balducci, L.; Blasi, A.; Saldarelli, M.; Soleti, A.; Pessina, A.; Bonomi, A.; Coccè, V.; Dossena, M.; Tosetti, V.; Ceserani, V.; et al. Immortalization of human adipose-derived stromal cells: Production of cell lines with high growth rate, mesenchymal marker expression and capability to secrete high levels of angiogenic factors. Stem Cell Res. Ther. 2014, 5, 63. [Google Scholar] [CrossRef] [Green Version]
  247. Piñeiro-Ramil, M.; Castro-Viñuelas, R.; Sanjurjo-Rodríguez, C.; Rodríguez-Fernández, S.; Hermida-Gómez, T.; Blanco-García, F.J.; Fuentes-Boquete, I.; Díaz-Prado, S. Immortalizing Mesenchymal Stromal Cells from Aged Donors While Keeping Their Essential Features. Stem Cells Int. 2020, 2020, 5726947. [Google Scholar] [CrossRef]
  248. Schneider, J.; Eiró, N.; Pérez-Fernández, R.; Martínez-Ordóñez, A.; Vizoso, F. Human Uterine Cervical Stromal Stem Cells (hUCESCs): Why and How they Exert their Antitumor Activity. Cancer Genom. Proteom. 2016, 13, 331–337. [Google Scholar]
  249. Apel, C.; Forlenza, O.V.; de Paula, V.J.; Talib, L.L.; Denecke, B.; Eduardo, C.P.; Gattaz, W.F. The neuroprotective effect of dental pulp cells in models of Alzheimer’s and Parkinson’s disease. J. Neural Transm. 2009, 116, 71–78. [Google Scholar] [CrossRef]
  250. Boulaiz, H.; Marchal, J.A.; Prados, J.; Melguizo, C.; Aránega, A. Non-viral and viral vectors for gene therapy. Cell. Mol. Biol. 2005, 51, 3–22. [Google Scholar]
  251. Filho, D.M.; de Carvalho Ribeiro, P.; Oliveira, L.F.; Dos Santos, A.; Parreira, R.C.; Pinto, M.C.X.; Resende, R.R. Enhancing the Therapeutic Potential of Mesenchymal Stem Cells with the CRISPR-Cas System. Stem Cell Rev. Rep. 2019, 15, 463–473. [Google Scholar] [CrossRef]
  252. Meng, X.; Zheng, M.; Yu, M.; Bai, W.; Zuo, L.; Bu, X.; Liu, Y.; Xia, L.; Hu, J.; Liu, L.; et al. Transplantation of CRISPRa system engineered IL10-overexpressing bone marrow-derived mesenchymal stem cells for the treatment of myocardial infarction in diabetic mice. J. Biol. Eng. 2019, 13, 49. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  253. Teixeira, F.G.; Salgado, A.J. Mesenchymal stem cells secretome: Current trends and future challenges. Neural Regen. Res. 2020, 15, 75–77. [Google Scholar] [CrossRef] [PubMed]
  254. Osugi, M.; Katagiri, W.; Yoshimi, R.; Inukai, T.; Hibi, H.; Ueda, M. Conditioned media from mesenchymal stem cells enhanced bone regeneration in rat calvarial bone defects. Tissue Eng. Part A 2012, 18, 1479–1489. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  255. van Niel, G.; D’Angelo, G.; Raposo, G. Shedding light on the cell biology of extracellular vesicles. Nat. Rev. Mol. Cell Biol. 2018, 19, 213–228. [Google Scholar] [CrossRef]
  256. Lou, G.; Chen, Z.; Zheng, M.; Liu, Y. Mesenchymal stem cell-derived exosomes as a new therapeutic strategy for liver diseases. Exp. Mol. Med. 2017, 49, e346. [Google Scholar] [CrossRef]
  257. Lasda, E.; Parker, R. Circular RNAs Co-Precipitate with Extracellular Vesicles: A Possible Mechanism for circRNA Clearance. PLoS ONE 2016, 11, e0148407. [Google Scholar] [CrossRef] [Green Version]
  258. Sun, B.; Shi, L.; Shi, Q.; Jiang, Y.; Su, Z.; Yang, X.; Zhang, Y. Circular RNAs are abundantly expressed and upregulated during repair of the damaged endometrium by Wharton’s jelly-derived mesenchymal stem cells. Stem Cell Res. Ther. 2018, 9, 314. [Google Scholar] [CrossRef]
  259. Bach, D.H.; Lee, S.K.; Sood, A.K. Circular RNAs in Cancer. Mol. Ther. Nucleic Acids 2019, 16, 118–129. [Google Scholar] [CrossRef] [Green Version]
  260. Ogino, H.; Nakamura, K.; Ihara, E.; Akiho, H.; Takayanagi, R. CD4+CD25+ regulatory T cells suppress Th17-responses in an experimental colitis model. Dig. Dis. Sci. 2011, 56, 376–386. [Google Scholar] [CrossRef]
  261. Lee, S.H.; Kwon, J.E.; Cho, M.L. Immunological pathogenesis of inflammatory bowel disease. Intest. Res. 2018, 16, 26–42. [Google Scholar] [CrossRef] [Green Version]
  262. Proto, J.D.; Doran, A.C.; Gusarova, G.; Yurdagul, A., Jr.; Sozen, E.; Subramanian, M.; Islam, M.N.; Rymond, C.C.; Du, J.; Hook, J.; et al. Regulatory T Cells Promote Macrophage Efferocytosis during Inflammation Resolution. Immunity 2018, 49, 666–677.e666. [Google Scholar] [CrossRef] [Green Version]
  263. Liu, G.; Ma, H.; Qiu, L.; Li, L.; Cao, Y.; Ma, J.; Zhao, Y. Phenotypic and functional switch of macrophages induced by regulatory CD4+CD25+ T cells in mice. Immunol. Cell Biol. 2011, 89, 130–142. [Google Scholar] [CrossRef]
  264. Cao, L.; Xu, H.; Wang, G.; Liu, M.; Tian, D.; Yuan, Z. Extracellular vesicles derived from bone marrow mesenchymal stem cells attenuate dextran sodium sulfate-induced ulcerative colitis by promoting M2 macrophage polarization. Int. Immunopharmacol. 2019, 72, 264–274. [Google Scholar] [CrossRef]
  265. Liu, H.; Liang, Z.; Wang, F.; Zhou, C.; Zheng, X.; Hu, T.; He, X.; Wu, X.; Lan, P. Exosomes from mesenchymal stromal cells reduce murine colonic inflammation via a macrophage-dependent mechanism. JCI Insight 2019, 4, e131273. [Google Scholar] [CrossRef] [Green Version]
  266. Yang, S.; Liang, X.; Song, J.; Li, C.; Liu, A.; Luo, Y.; Ma, H.; Tan, Y.; Zhang, X. A novel therapeutic approach for inflammatory bowel disease by exosomes derived from human umbilical cord mesenchymal stem cells to repair intestinal barrier via TSG-6. Stem Cell Res. Ther. 2021, 12, 315. [Google Scholar] [CrossRef]
  267. Chen, W.; Huang, Y.; Han, J.; Yu, L.; Li, Y.; Lu, Z.; Li, H.; Liu, Z.; Shi, C.; Duan, F.; et al. Immunomodulatory effects of mesenchymal stromal cells-derived exosome. Immunol. Res. 2016, 64, 831–840. [Google Scholar] [CrossRef]
  268. Harrell, C.R.; Jovicic, N.; Djonov, V.; Arsenijevic, N.; Volarevic, V. Mesenchymal Stem Cell-Derived Exosomes and Other Extracellular Vesicles as New Remedies in the Therapy of Inflammatory Diseases. Cells 2019, 8, 1605. [Google Scholar] [CrossRef] [Green Version]
  269. Zheng, D.; Zhou, H.; Wang, H.; Zhu, Y.; Wu, Y.; Li, Q.; Li, T.; Liu, L. Mesenchymal stem cell-derived microvesicles improve intestinal barrier function by restoring mitochondrial dynamic balance in sepsis rats. Stem Cell Res. Ther. 2021, 12, 299. [Google Scholar] [CrossRef]
  270. Giacomello, M.; Pyakurel, A.; Glytsou, C.; Scorrano, L. The cell biology of mitochondrial membrane dynamics. Nat. Rev. Mol. Cell Biol. 2020, 21, 204–224. [Google Scholar] [CrossRef]
  271. Hassan, M.; Yazid, M.D.; Yunus, M.H.M.; Chowdhury, S.R.; Lokanathan, Y.; Idrus, R.B.H.; Ng, A.M.H.; Law, J.X. Large-Scale Expansion of Human Mesenchymal Stem Cells. Stem Cells Int. 2020, 2020, 9529465. [Google Scholar] [CrossRef]
  272. Chen, A.K.; Reuveny, S.; Oh, S.K. Application of human mesenchymal and pluripotent stem cell microcarrier cultures in cellular therapy: Achievements and future direction. Biotechnol. Adv. 2013, 31, 1032–1046. [Google Scholar] [CrossRef] [Green Version]
  273. Yu, C.; Kornmuller, A.; Brown, C.; Hoare, T.; Flynn, L.E. Decellularized adipose tissue microcarriers as a dynamic culture platform for human adipose-derived stem/stromal cell expansion. Biomaterials 2017, 120, 66–80. [Google Scholar] [CrossRef]
  274. Song, E.M.; Joo, Y.H.; Choe, A.R.; Park, Y.; Tae, C.H.; Hong, J.T.; Moon, C.M.; Kim, S.E.; Jung, H.K.; Shim, K.N.; et al. Three-dimensional culture method enhances the therapeutic efficacies of tonsil-derived mesenchymal stem cells in murine chronic colitis model. Sci. Rep. 2021, 11, 19589. [Google Scholar] [CrossRef]
  275. Barckhausen, C.; Rice, B.; Baila, S.; Sensebé, L.; Schrezenmeier, H.; Nold, P.; Hackstein, H.; Rojewski, M.T. GMP-Compliant Expansion of Clinical-Grade Human Mesenchymal Stromal/Stem Cells Using a Closed Hollow Fiber Bioreactor. Methods Mol. Biol. 2016, 1416, 389–412. [Google Scholar] [CrossRef]
  276. Castilla-Casadiego, D.A.; Reyes-Ramos, A.M.; Domenech, M.; Almodovar, J. Effects of Physical, Chemical, and Biological Stimulus on h-MSC Expansion and Their Functional Characteristics. Ann. Biomed. Eng. 2020, 48, 519–535. [Google Scholar] [CrossRef]
  277. Noronha, N.C.; Mizukami, A.; Caliári-Oliveira, C.; Cominal, J.G.; Rocha, J.L.M.; Covas, D.T.; Swiech, K.; Malmegrim, K.C.R. Priming approaches to improve the efficacy of mesenchymal stromal cell-based therapies. Stem Cell Res. Ther. 2019, 10, 131, Correction in Stem Cell Res. Ther. 2019, 10, 132. [Google Scholar] [CrossRef] [Green Version]
  278. Hoch, A.I.; Leach, J.K. Concise review: Optimizing expansion of bone marrow mesenchymal stem/stromal cells for clinical applications. Stem Cells Transl. Med. 2014, 3, 643–652. [Google Scholar] [CrossRef]
  279. Henn, A.; Darou, S.; Yerden, R. Full-time physioxic culture conditions promote MSC proliferation more than hypoxic preconditioning. Cytotherapy 2019, 21, S73–S74. [Google Scholar] [CrossRef]
  280. Basciano, L.; Nemos, C.; Foliguet, B.; de Isla, N.; de Carvalho, M.; Tran, N.; Dalloul, A. Long term culture of mesenchymal stem cells in hypoxia promotes a genetic program maintaining their undifferentiated and multipotent status. BMC Cell Biol. 2011, 12, 12. [Google Scholar] [CrossRef] [Green Version]
  281. Zhu, C.; Yu, J.; Pan, Q.; Yang, J.; Hao, G.; Wang, Y.; Li, L.; Cao, H. Hypoxia-inducible factor-2 alpha promotes the proliferation of human placenta-derived mesenchymal stem cells through the MAPK/ERK signaling pathway. Sci. Rep. 2016, 6, 35489. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  282. Bader, A.M.; Klose, K.; Bieback, K.; Korinth, D.; Schneider, M.; Seifert, M.; Choi, Y.H.; Kurtz, A.; Falk, V.; Stamm, C. Hypoxic Preconditioning Increases Survival and Pro-Angiogenic Capacity of Human Cord Blood Mesenchymal Stromal Cells In Vitro. PLoS ONE 2015, 10, e0138477. [Google Scholar] [CrossRef] [PubMed]
  283. Lee, E.Y.; Xia, Y.; Kim, W.S.; Kim, M.H.; Kim, T.H.; Kim, K.J.; Park, B.S.; Sung, J.H. Hypoxia-enhanced wound-healing function of adipose-derived stem cells: Increase in stem cell proliferation and up-regulation of VEGF and bFGF. Wound Repair Regen. 2009, 17, 540–547. [Google Scholar] [CrossRef] [PubMed]
  284. Han, K.H.; Kim, A.K.; Jeong, G.J.; Jeon, H.R.; Bhang, S.H.; Kim, D.I. Enhanced Anti-Cancer Effects of Conditioned Medium from Hypoxic Human Umbilical Cord-Derived Mesenchymal Stem Cells. Int. J. Stem Cells 2019, 12, 291–303. [Google Scholar] [CrossRef] [PubMed]
  285. Vertelov, G.; Kharazi, L.; Muralidhar, M.G.; Sanati, G.; Tankovich, T.; Kharazi, A. High targeted migration of human mesenchymal stem cells grown in hypoxia is associated with enhanced activation of RhoA. Stem Cell Res. Ther. 2013, 4, 5. [Google Scholar] [CrossRef] [Green Version]
  286. Ying, J.; You, Q.; Wang, Z.; Hu, Z. Hypoxic preconditioning promotes the immunosuppressive effects of mesenchymal stem cells in mice with colitis. Res. Vet. Sci. 2021, 144, 157–163. [Google Scholar] [CrossRef]
  287. Monfoulet, L.E.; Becquart, P.; Marchat, D.; Vandamme, K.; Bourguignon, M.; Pacard, E.; Viateau, V.; Petite, H.; Logeart-Avramoglou, D. The pH in the microenvironment of human mesenchymal stem cells is a critical factor for optimal osteogenesis in tissue-engineered constructs. Tissue Eng. Part A 2014, 20, 1827–1840. [Google Scholar] [CrossRef] [Green Version]
  288. Krampera, M.; Cosmi, L.; Angeli, R.; Pasini, A.; Liotta, F.; Andreini, A.; Santarlasci, V.; Mazzinghi, B.; Pizzolo, G.; Vinante, F.; et al. Role for interferon-gamma in the immunomodulatory activity of human bone marrow mesenchymal stem cells. Stem Cells 2006, 24, 386–398. [Google Scholar] [CrossRef]
  289. Ren, G.; Zhang, L.; Zhao, X.; Xu, G.; Zhang, Y.; Roberts, A.I.; Zhao, R.C.; Shi, Y. Mesenchymal stem cell-mediated immunosuppression occurs via concerted action of chemokines and nitric oxide. Cell Stem Cell 2008, 2, 141–150. [Google Scholar] [CrossRef] [Green Version]
  290. Campaña-Seoane, M.; Peleteiro, A.; Laguna, R.; Otero-Espinar, F.J. Bioadhesive emulsions for control release of progesterone resistant to vaginal fluids clearance. Int. J. Pharm. 2014, 477, 495–505. [Google Scholar] [CrossRef]
  291. Wu, H.; Fan, H.; Shou, Z.; Xu, M.; Chen, Q.; Ai, C.; Dong, Y.; Liu, Y.; Nan, Z.; Wang, Y.; et al. Extracellular vesicles containing miR-146a attenuate experimental colitis by targeting TRAF6 and IRAK1. Int. Immunopharmacol. 2019, 68, 204–212. [Google Scholar] [CrossRef]
  292. Joo, H.; Oh, M.K.; Kang, J.Y.; Park, H.S.; Chae, D.H.; Kim, J.; Lee, J.H.; Yoo, H.M.; Choi, U.; Kim, D.K.; et al. Extracellular Vesicles from Thapsigargin-Treated Mesenchymal Stem Cells Ameliorated Experimental Colitis via Enhanced Immunomodulatory Properties. Biomedicines 2021, 9, 209. [Google Scholar] [CrossRef]
  293. Kang, J.Y.; Oh, M.K.; Joo, H.; Park, H.S.; Chae, D.H.; Kim, J.; Lee, H.R.; Oh, I.H.; Yu, K.R. Xeno-Free Condition Enhances Therapeutic Functions of Human Wharton’s Jelly-Derived Mesenchymal Stem Cells against Experimental Colitis by Upregulated Indoleamine 2,3-Dioxygenase Activity. J. Clin. Med. 2020, 9, 2913. [Google Scholar] [CrossRef]
  294. Wu, X.; Wu, D.; Mu, Y.; Zhao, Y.; Ma, Z. Serum-Free Medium Enhances the Therapeutic Effects of Umbilical Cord Mesenchymal Stromal Cells on a Murine Model for Acute Colitis. Front. Bioeng. Biotechnol. 2020, 8, 586. [Google Scholar] [CrossRef]
  295. Chen, C.; Han, X.; Wang, G.; Liu, D.; Bao, L.; Jiao, C.; Luan, J.; Hou, Y.; Xu, Y.; Wang, H.; et al. Nrf2 deficiency aggravates the kidney injury induced by subacute cadmium exposure in mice. Arch. Toxicol. 2021, 95, 883–893. [Google Scholar] [CrossRef]
  296. Billing, A.M.; Ben Hamidane, H.; Dib, S.S.; Cotton, R.J.; Bhagwat, A.M.; Kumar, P.; Hayat, S.; Yousri, N.A.; Goswami, N.; Suhre, K.; et al. Comprehensive transcriptomic and proteomic characterization of human mesenchymal stem cells reveals source specific cellular markers. Sci. Rep. 2016, 6, 21507. [Google Scholar] [CrossRef] [Green Version]
  297. Menard, C.; Pacelli, L.; Bassi, G.; Dulong, J.; Bifari, F.; Bezier, I.; Zanoncello, J.; Ricciardi, M.; Latour, M.; Bourin, P.; et al. Clinical-grade mesenchymal stromal cells produced under various good manufacturing practice processes differ in their immunomodulatory properties: Standardization of immune quality controls. Stem Cells Dev. 2013, 22, 1789–1801. [Google Scholar] [CrossRef]
  298. Isobe, Y.; Koyama, N.; Nakao, K.; Osawa, K.; Ikeno, M.; Yamanaka, S.; Okubo, Y.; Fujimura, K.; Bessho, K. Comparison of human mesenchymal stem cells derived from bone marrow, synovial fluid, adult dental pulp, and exfoliated deciduous tooth pulp. Int. J. Oral Maxillofac. Surg. 2016, 45, 124–131. [Google Scholar] [CrossRef]
  299. Schäfer, R.; Spohn, G.; Baer, P.C. Mesenchymal Stem/Stromal Cells in Regenerative Medicine: Can Preconditioning Strategies Improve Therapeutic Efficacy? Transfus. Med. Hemother. 2016, 43, 256–267. [Google Scholar] [CrossRef] [Green Version]
  300. Teixeira, F.G.; Panchalingam, K.M.; Assunção-Silva, R.; Serra, S.C.; Mendes-Pinheiro, B.; Patrício, P.; Jung, S.; Anjo, S.I.; Manadas, B.; Pinto, L.; et al. Modulation of the Mesenchymal Stem Cell Secretome Using Computer-Controlled Bioreactors: Impact on Neuronal Cell Proliferation, Survival and Differentiation. Sci. Rep. 2016, 6, 27791. [Google Scholar] [CrossRef]
  301. Cunha, B.; Aguiar, T.; Carvalho, S.B.; Silva, M.M.; Gomes, R.A.; Carrondo, M.J.T.; Gomes-Alves, P.; Peixoto, C.; Serra, M.; Alves, P.M. Bioprocess integration for human mesenchymal stem cells: From up to downstream processing scale-up to cell proteome characterization. J. Biotechnol. 2017, 248, 87–98. [Google Scholar] [CrossRef]
  302. Andrzejewska, A.; Catar, R.; Schoon, J.; Qazi, T.H.; Sass, F.A.; Jacobi, D.; Blankenstein, A.; Reinke, S.; Krüger, D.; Streitz, M.; et al. Multi-Parameter Analysis of Biobanked Human Bone Marrow Stromal Cells Shows Little Influence for Donor Age and Mild Comorbidities on Phenotypic and Functional Properties. Front. Immunol. 2019, 10, 2474. [Google Scholar] [CrossRef]
  303. Bieback, K.; Kuçi, S.; Schäfer, R. Production and quality testing of multipotent mesenchymal stromal cell therapeutics for clinical use. Transfusion 2019, 59, 2164–2173. [Google Scholar] [CrossRef]
  304. Sala, E.; Genua, M.; Petti, L.; Anselmo, A.; Arena, V.; Cibella, J.; Zanotti, L.; D’Alessio, S.; Scaldaferri, F.; Luca, G.; et al. Mesenchymal Stem Cells Reduce Colitis in Mice via Release of TSG6, Independently of Their Localization to the Intestine. Gastroenterology 2015, 149, 163–176.e120. [Google Scholar] [CrossRef]
  305. Castelo-Branco, M.T.; Soares, I.D.; Lopes, D.V.; Buongusto, F.; Martinusso, C.A.; do Rosario, A., Jr.; Souza, S.A.; Gutfilen, B.; Fonseca, L.M.; Elia, C.; et al. Intraperitoneal but not intravenous cryopreserved mesenchymal stromal cells home to the inflamed colon and ameliorate experimental colitis. PLoS ONE 2012, 7, e33360. [Google Scholar] [CrossRef] [Green Version]
  306. Garcia-Olmo, D.; Gilaberte, I.; Binek, M.; AJL, D.H.; Lindner, D.; Selvaggi, F.; Spinelli, A.; Panes, J. Follow-up Study to Evaluate the Long-term Safety and Efficacy of Darvadstrocel (Mesenchymal Stem Cell Treatment) in Patients With Perianal Fistulizing Crohn’s Disease: ADMIRE-CD Phase 3 Randomized Controlled Trial. Dis. Colon Rectum 2022, 65, 713–720. [Google Scholar] [CrossRef]
  307. Laroui, H.; Ingersoll, S.A.; Liu, H.C.; Baker, M.T.; Ayyadurai, S.; Charania, M.A.; Laroui, F.; Yan, Y.; Sitaraman, S.V.; Merlin, D. Dextran sodium sulfate (DSS) induces colitis in mice by forming nano-lipocomplexes with medium-chain-length fatty acids in the colon. PLoS ONE 2012, 7, e32084. [Google Scholar] [CrossRef]
  308. Koffi, A.A.; Agnely, F.; Ponchel, G.; Grossiord, J.L. Modulation of the rheological and mucoadhesive properties of thermosensitive poloxamer-based hydrogels intended for the rectal administration of quinine. Eur. J. Pharm. Sci. 2006, 27, 328–335. [Google Scholar] [CrossRef]
  309. Soliman, G.M.; Fetih, G.; Abbas, A.M. Thermosensitive bioadhesive gels for the vaginal delivery of sildenafil citrate: In vitro characterization and clinical evaluation in women using clomiphene citrate for induction of ovulation. Drug Dev. Ind. Pharm. 2017, 43, 399–408. [Google Scholar] [CrossRef]
  310. Liu, Y.; Lu, W.L.; Wang, J.C.; Zhang, X.; Zhang, H.; Wang, X.Q.; Zhou, T.Y.; Zhang, Q. Controlled delivery of recombinant hirudin based on thermo-sensitive Pluronic F127 hydrogel for subcutaneous administration: In vitro and in vivo characterization. J. Control. Release 2007, 117, 387–395. [Google Scholar] [CrossRef]
  311. Sendon-Lago, J.; Seoane, S.; Martinez-Ordoñez, A.; Eiro, N.; Saa, J.; Vizoso, F.J.; Gonzalez, F.; Perez-Fernandez, R.; Bermudez, M.A. Corneal regeneration by conditioned medium of human uterine cervical stem cells is mediated by TIMP-1 and TIMP-2. Exp. Eye Res. 2019, 180, 110–121. [Google Scholar] [CrossRef] [PubMed]
  312. Sosnik, A.; Seremeta, K.P. Polymeric Hydrogels as Technology Platform for Drug Delivery Applications. Gels 2017, 3, 25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  313. Chatterjee, S.; Hui, P.C.; Kan, C.W. Thermoresponsive Hydrogels and Their Biomedical Applications: Special Insight into Their Applications in Textile Based Transdermal Therapy. Polymers 2018, 10, 480. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  314. Garcia-Del Rio, L.; Diaz-Rodriguez, P.; Landin, M. New tools to design smart thermosensitive hydrogels for protein rectal delivery in IBD. Mater. Sci. Eng. C 2020, 106, 110252. [Google Scholar] [CrossRef]
  315. Din, F.U.; Mustapha, O.; Kim, D.W.; Rashid, R.; Park, J.H.; Choi, J.Y.; Ku, S.K.; Yong, C.S.; Kim, J.O.; Choi, H.G. Novel dual-reverse thermosensitive solid lipid nanoparticle-loaded hydrogel for rectal administration of flurbiprofen with improved bioavailability and reduced initial burst effect. Eur. J. Pharm. Biopharm. 2015, 94, 64–72. [Google Scholar] [CrossRef]
  316. Santos Akkari, A.C.; Ramos Campos, E.V.; Keppler, A.F.; Fraceto, L.F.; de Paula, E.; Tófoli, G.R.; de Araujo, D.R. Budesonide-hydroxypropyl-β-cyclodextrin inclusion complex in binary poloxamer 407/403 system for ulcerative colitis treatment: A physico-chemical study from micelles to hydrogels. Colloids Surf. B Biointerfaces 2016, 138, 138–147. [Google Scholar] [CrossRef] [Green Version]
Figure 1. IBD pathophysiology and MSC properties.
Figure 1. IBD pathophysiology and MSC properties.
Ijms 23 08905 g001
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Eiro, N.; Fraile, M.; González-Jubete, A.; González, L.O.; Vizoso, F.J. Mesenchymal (Stem) Stromal Cells Based as New Therapeutic Alternative in Inflammatory Bowel Disease: Basic Mechanisms, Experimental and Clinical Evidence, and Challenges. Int. J. Mol. Sci. 2022, 23, 8905. https://doi.org/10.3390/ijms23168905

AMA Style

Eiro N, Fraile M, González-Jubete A, González LO, Vizoso FJ. Mesenchymal (Stem) Stromal Cells Based as New Therapeutic Alternative in Inflammatory Bowel Disease: Basic Mechanisms, Experimental and Clinical Evidence, and Challenges. International Journal of Molecular Sciences. 2022; 23(16):8905. https://doi.org/10.3390/ijms23168905

Chicago/Turabian Style

Eiro, Noemi, Maria Fraile, Alberto González-Jubete, Luis O. González, and Francisco J. Vizoso. 2022. "Mesenchymal (Stem) Stromal Cells Based as New Therapeutic Alternative in Inflammatory Bowel Disease: Basic Mechanisms, Experimental and Clinical Evidence, and Challenges" International Journal of Molecular Sciences 23, no. 16: 8905. https://doi.org/10.3390/ijms23168905

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

Article Metrics

Back to TopTop