Next Article in Journal
Estrogens in Adipose Tissue Physiology and Obesity-Related Dysfunction
Next Article in Special Issue
Mesenchymal Stem Cells Reduce the Extracellular Mitochondrial DNA-Mediated TLR9 Activation in Neonatal Hyperoxia-Induced Lung Injury
Previous Article in Journal
Prognostic Value of Liver Kinase B1 (LKB1) in Gastric Cancer-Associated Tumor Microenvironment Immunity
Previous Article in Special Issue
Current Perspectives on Adult Mesenchymal Stromal Cell-Derived Extracellular Vesicles: Biological Features and Clinical Indications
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Role of Mesenchymal Stem/Stromal Cells in Modulating Ischemia/Reperfusion Injury: Current State of the Art and Future Perspectives

Research Department, IRCSS ISMETT (Istituto Mediterraneo per I Trapianti e Terapie ad Alta Specializzazione), 90127 Palermo, Italy
*
Authors to whom correspondence should be addressed.
Biomedicines 2023, 11(3), 689; https://doi.org/10.3390/biomedicines11030689
Submission received: 7 February 2023 / Revised: 15 February 2023 / Accepted: 21 February 2023 / Published: 23 February 2023

Abstract

:
Ischemia/reperfusion injury (IRI) is a multistep damage that occurs in several tissues when a blood flow interruption is inevitable, such as during organ surgery or transplantation. It is responsible for cell death and tissue dysfunction, thus leading, in the case of transplantation, to organ rejection. IRI takes place during reperfusion, i.e., when blood flow is restored, by activating inflammation and reactive oxygen species (ROS) production, causing mitochondrial damage and apoptosis of parenchymal cells. Unfortunately, none of the therapies currently in use are definitive, prompting the need for new therapeutic approaches. Scientific evidence has proven that mesenchymal stem/stromal cells (MSCs) can reduce inflammation and ROS, prompting this cellular therapy to also be investigated for treatment of IRI. Moreover, it has been shown that MSC therapeutic effects were mediated in part by their secretome, which appears to be involved in immune regulation and tissue repair. For these reasons, mediated MSC paracrine function might be key for injury amelioration upon IRI damage. In this review, we highlight the scientific literature on the potential beneficial use of MSCs and their products for improving IRI outcomes in different tissues/organs, focusing in particular on the paracrine effects mediated by MSCs, and on the molecular mechanisms behind these effects.

1. Introduction

Ischemia/reperfusion injury (IRI) is defined as organ damage due to cellular dysfunction. It usually occurs after organ surgery, and in particular during organ transplantation [1,2,3,4]. IRI is considered a multistep damage: for example, in transplantation, the first step is induced by organ resection and the inevitable ischemia, due to the interruption of blood flow and cold preservation of the organ for its storage before the transplant. During the ischemic phase, which varies according to the quality of the donor organ, disruption of blood flow and organ preservation at cold temperatures lead to mitochondrial dysfunction, intracellular adenosine triphosphate (ATP) depletion, and a switch to anaerobic metabolism [3]. Restoration of the blood flow during reperfusion exacerbates the damage, causing destruction of the function and viability of the organ. In particular, re-establishment of blood supplies causes an extensive production of pro-inflammatory cytokines and reactive oxygen species (ROS), which eventually leads to neutrophil infiltration and apoptosis of parenchymal cells [5,6]. IRI occurs in a wide range of organs, including lung, pancreas, kidney, gut, heart, brain, and liver, and can also induce systemic damage, potentially leading to multisystem organ failure [7]. Despite the variety of tissues involved, the biological processes activated by IRI are relatively common, and include inflammation, ROS production, and apoptosis. Currently, different therapeutic strategies have been implemented to attenuate IRI, though none have been considered definitive [8,9,10]. For example, ischemic pre-conditioning, which consists of brief and repetitive episodes of IRI before the induction of sustained organ ischemia, has been found to be effective for a number of operative settings where ischemia can be tightly controlled, such as transplantation, coronary bypass grafting, and elective major vascular procedures [11,12]. Ischemic post-conditioning is instead defined as the rapid and sequential intermittent interruption of blood supplies in the early stages of organ reperfusion. This therapeutic approach is usually applied when the ischemic damage cannot be predicted [13,14]. Finally, together with what is cited above, pharmacological treatments, despite their encouraging results in animal models of IRI, have not provided the expected results in a large number of clinical trials [8,15,16,17,18]. Consequently, it seems clear that new potential therapeutic approaches need to be identified and tested in order to reduce IRI side effects in various clinical settings.

2. Mesenchymal Stem/Stromal Cell (MSC)-Based Therapy as a New Strategy for Treating IRI

Scientific evidence has revealed that both cellular and acellular therapies based on the use of mesenchymal stem/stromal cells (MSCs), represent promising approaches to mitigate IRI-related pathological processes [19,20,21,22]. MSCs have been found in different tissues, including umbilical cord (UC-MSCs) [23], bone marrow (BM-MSCs) [24], adipose tissue (Ad-MSCs) [25], and placenta (AM-MSCs) [26], where they participate in the maintenance of stem cell niches and tissue homeostasis [27,28]. Several studies have demonstrated that MSCs possess therapeutic properties arising from their ability to secrete a plethora of functional factors involved in both immune regulation and tissue repair [29,30,31,32,33]. In particular, MSCs can secrete growth factors, cytokines, chemokines, and extracellular vesicles (EVs), such as exosomes (EXOs), which confer on MSCs paracrine therapeutic capabilities that can lead to, e.g., immune modulation, tissue injury amelioration, and reduction in fibrosis [32,34,35,36,37,38]. Oxidative stress is strongly correlated with cellular injury, and involved in the onset of several pathologies, including IRI [39,40]. Growing evidence supports the hypothesis that MSCs exert antioxidant properties in several pathological processes, which may explain MSCs’ cytoprotective properties in IRI experimental models [41,42]. These cells have been extensively tested in different areas of therapeutic application, including organ transplantation [43,44], where the beneficial strategies are aimed at reducing IRI and acute inflammatory responses (Table 1). Indeed, in many experimental models, including organ transplant studies, it has been found that infusion of MSCs promotes regeneration and prolonged recipient survival by reducing IRI and acute inflammation (Figure 1) [45,46,47,48,49,50,51].
In the future, the routine use of allogeneic MSCs may well present some hurdles to be overcome. In fact, MSCs can be found in the parenchyma of different tissues after intravascular administration, leading to the risk of tumor formation and/or the possibility of immune rejection for allogenic cells. Consequently, in order to obtain the therapeutic effects of MSCs without using the cells, MSC-derived products such as conditioned medium (CM) and EXOs have been investigated. For example, various in vivo studies of organ injury have highlighted the efficacy of CM derived from MSC cultures [52,53], supporting the emerging consensus that MSCs secrete bioactive factors that can mediate beneficial therapeutic effects through a paracrine mechanism. Indeed, individual components of the MSC secretome have been involved in a number of essential cellular processes, including angiogenesis, immunomodulation, wound healing, and tissue repair [32,54]. MSC-derived CM also contains functional factors that have shown therapeutic efficacy against ischemia reperfusion injury. In several IRI experimental models, it has been demonstrated that the whole secretome or EXOs alone were able to alleviate IRI side effects by attenuating inflammation and apoptosis, and improving tissue regeneration (Table 1).
Table 1. Summary of in vitro and in vivo studies reporting the use of MSCs and/or their products in preventing ischemia/reperfusion injury (IRI).
Table 1. Summary of in vitro and in vivo studies reporting the use of MSCs and/or their products in preventing ischemia/reperfusion injury (IRI).
Use of Cells or Their Products Study ModelEffects Due to MSC TreatmentReferences
AMSC-derived CMIn vitro model of human lung IRIAttenuation of IRI effects by improving
the efficacy of in vitro EVLP
[20]
AMSC-derived CMIn vitro model of hepatic IRIInhibition of activation of inflammatory macrophages and apoptosis in hepatocytes[22]
UC-MSCsRat lung IRIReduction in oxidative stress damage and inflammation[41]
BM-MSC-derived EVsMouse intestinal IRIMitigation of intestinal pathological injury, reduction in intestinal cell apoptosis and oxidative stress[42]
UC-MSCsSwine lung IRIAttenuation of IRI by improving the efficacy of EVLP[49]
BM-MSCsMouse lung IRIProtection against cold IRI in lung transplants[55]
AdMSCsRat lung IRIAttenuation of inflammation and oxidative stress[56]
BM-MSCsRat lung IRIReduction in both pulmonary edema and pro-inflammatory factors, and increase in anti-inflammatory factors[57]
AdMSCsRat lung IRIAttenuation of lung damage after IRI[58]
BM-MSCsRat lung IRIAttenuation of lung pathologic injury[59]
BM-MSCsHuman lung IRI and EVLPDecreased cold ischemic injury[60]
BM-MSCsEVLP in human lungs rejected for transplantationImprovement in alveolar fluid clearance and
reduction in both acute IRI and fibrotic responses
[61]
MSC-derived EVsRat lung IRI and EVLPImproved tissue integrity and metabolism[62]
UC-MSCs and UC-MSC-derived EVsMouse lung IRIAttenuation of lung dysfunction and injury by improving the efficacy of EVLP[63]
BM-MSC-derived CMRat lung IRIProtection against lung IRI[64]
BM-MSCsMouse model of kidney IRIInduction of M1 to M2 transition
and tissue regeneration
[65]
BM-MSCsHuman renal allograft model with exsanguinous metabolic supportReduction in ischemic damage and
inflammatory cytokines
[66]
BM-MSCsHuman renal allograft model with normothermic machine perfusionReduction in ischemia reperfusion injury[67]
BM-MSCsRat intestinal IRIReduction in inflammatory response and intestinal ischemic damage [68]
BM-MSCsRat intestinal IRIReduction in intestinal ischemic damage[69]
BM-MSCsSwine myocardial infarction modelAttenuation of contractile dysfunction and pathologic thinning[70]
AMSCsSwine myocardial infarction modelReduction in histological and functional impairment of myocardium[71]
BM-MSC-derived EVsRat myocardial infarction modelImprovement of blood flow recovery, reduction in infarct size and preservation of cardiac systolic and diastolic performance[72]
MSC-derived CMRat cardiac allograft model with hypothermic perfusionImprovement of cardiac function and reduction in pro-inflammatory cytokines[73]
BM-MSC-derived CM and EVsHypothermic perfusion of mouse donor heartAttenuation of ischemia-induced myocardial
damage in donor heart and
improvement of heart function after
transplantation
[74]
BM-MSC-derived CMRat cardiac allograft modelImprovement of cardiac post-operatory functions and reduction in pro-inflammatory cytokines[75]
BM-MSCsMouse model of embolic middle cerebral artery occlusionImprovement in functional recovery[76]
BM-MSCsRat model of embolic middle cerebral artery occlusionImprovement of function, induction of angiogenesis, and reduction in apoptosis[77]
BM-MSCsRat model of hepatic IRIProtection from the progression of the damage and reduction in neutrophil infiltration[78]
AdMSCsRat model of hepatic IRI and hepatectomyInhibition of hepatic apoptosis and
improvement of tissue regeneration
[79]
AdMSCsRat model of hepatic IRIInhibition of inflammasome activation[80]
AdMSC-derived CMSwine model of hepatic IRI and hepatectomyAttenuation of hepatic IRI and
hepatectomy-induced liver damage
[81]
MSC-derived EVsMouse model of hepatic IRIAttenuation of liver damage and improvement in liver regeneration after IRI[82]
BM-MSC-derived EVsMouse model of hepatic IRIAttenuation of liver damage and
inflammatory responses
[83]
UC-MSC-derived EVsMouse model of hepatic IRIReduction in liver IRI by reducing
apoptosis and inflammation
[84]
UC-MSCRat model of hepatic IRIAttenuation of injury by inhibiting inflammation, neutrophil infiltration, and apoptosis[85]
EVLP: ex vivo lung perfusion; EVs: extracellular vesicles; CM: conditioned medium; MSCs: mesenchymal stem cells; BM-MSCs: bone marrow-derived MSCs; AMSCs: amnion-derived MSCs; UC-MSCs: umbilical cord-derived MSCs; AdMSCs: adipose-derived MSCs; IRI: ischemia-reperfusion injury.
Therefore, the therapeutic potential of both MSCs and MSC-derived products, are of particular interest as a strategy for modulating injury due to ischemia/reperfusion in several IRI-related diseases. In this review, we summarize the major studies investigating the clinical efficacy of MSC-based therapy in different IRI experimental models.

2.1. MSCs and Ischemia/Reperfusion Injury in the Lung

Ischemia/reperfusion injury is the leading cause of postoperative dysfunction after lung transplantation (LTx) [86,87]. This pathological condition is characterized by ROS production (production of toxic molecules), increased inflammation, alveolar damage, and lung edema, with consequent injury to the lung parenchyma, which can result in both early primary graft dysfunction (PGD) and/or chronic lung allograft dysfunction (CLAD) [19,41,88,89,90,91]. Lungs are particularly susceptible to IRI which, together with infections, can contribute to lung rejection and post-transplant mortality [90,92]. Therefore, a reduction in IRI adverse effects is a crucial step to improving LTx.
Over the last decade, a number of reports have shown that MSCs and/or their products (CM and EVs) are able to stimulate tissue regeneration, inhibit immunological responses, and block ROS production [32,35,93,94,95,96,97,98]. They have been shown capable of decreasing inflammation and IRI in both in vitro and in vivo models, and those effects were mediated, at least in part, by the paracrine activity of MSCs (Figure 1) [99]. MSC therapeutic action has also been evaluated both in human lung diseases [19,100,101,102,103] and in different lung experimental models [19,55,97,104,105]. In those cases, MSCs and/or their products were capable of potentiating anti-microbial action, and mitigating both lung injury and inflammation, as found with IRI of LTx. Lin et al., in a rat model of lung injury, demonstrated that both xenogeneic and allogeneic MSCs were able to protect the lung against IRI by downregulating inflammation and oxidative stress [56]. Using the same model, Lu et al. showed that intravenous injection of BM-MSCs reduced both pulmonary edema and pro-inflammatory factors, while increasing anti-inflammatory factors [57]. In a rodent model, Sun et al. found that the autologous transplantation of Ad-MSCs was able to reduce lung IRI [58]. Moreover, Guillamat-Prats, Chen and colleagues demonstrated that the engraftment of allogenic MSCs reduced acute lung injury [59,106], while Chambers et al. showed that MSCs, infused via the peripheral vein twice weekly for 2 weeks, also decreased CLAD in human lung-transplant recipients [107].
Recently, it has been postulated that the use of normothermic ex vivo lung perfusion (EVLP) may be helpful in mitigating ischemic injury to the lung [108,109]. During EVLP, the organ is placed in a device, providing ventilation and perfusion for 4–6 h, and allowing the evaluation and recovery of compromised donor lungs [110,111]. Interestingly, for the duration of EVLP, while the estimation of donor lung function can be made, different treatments can also be used to further reduce IRI [15,18]. In this regard, MSC-based treatments (MSCs or MSC-derived products) have been tested to improve EVLP. The promising results obtained from those studies have led MSCs to be considered good candidates for integration with EVLP, aiming at improvement in LTx. Several reports have suggested that MSC-based treatment administered during EVLP is associated with a decrease in inflammation and ischemic injury of human donor lungs [49,60,112]. McAuley et al. showed that when MSCs were added to the EVLP perfusate, they can exercise the ability to restore alveolar fluid clearance, and reduce both acute IRI and fibrotic responses in human lungs rejected for transplantation [61]. Moreover, Pacienza et al., in a rat preservation model of lung injury, found that MSC treatment during EVLP was able to reduce oxidative damage and IRI [41]. Promising results have also been found by using CM or EVs obtained from MSC cultures. It has been shown that MSC-derived EVs were able to reduce inflammation, and enhance tissue regeneration in acute lung injury models [62,63,113]. In a mouse model of lung IRI, treatment with EVs derived from UC-MSCs improved the efficacy of EVLP, and attenuated lung dysfunction by decreasing pro-inflammatory cytokines, neutrophil infiltration, and edema [63]. Moreover, EVs derived from BM-MSCs, when administered during EVLP, were able to increase the alveolar fluid clearance in human donor lungs rejected for transplantation [114]. Similarly to EVs, MSC-derived CM has shown beneficial effects on different experimental models of lung diseases [20,64,115,116,117]. For example, BM-MSC-derived CM, when administered during EVLP, was able to increase alveolar fluid clearance in human lung injured by the E. coli endotoxin [117]. The use of MSC-derived products, rather than the direct use of cells, can avoid all risks associated with live-cell transplants and, therefore, represents an emerging and promising approach to treating IRI in the field of LTx.
The use of MSCs and/or their products is a promising approach to reducing IRI in several lung pathological conditions; therefore, the integration of MSC-based treatment with EVLP in the context of LTx seems to be the best way to improve LTx outcomes.

2.2. MSCs and Ischemia/Reperfusion Injury in the Pancreas

The transplantation of the whole pancreas or pancreatic islets is considered an effective treatment for restoring glycemia in specific patients with unstable type 1 diabetes mellitus (T1D). However, in the case of the whole pancreas, similarly to other solid organs, IRI pathological processes often lead to complications which occur after transplantation, resulting in poor transplant success [118]. On the other hand, the clinical outcome also needs to be improved for islet transplantation because a variable degree and length of insulin independency have been observed [119]. A number of aspects can affect clinical outcomes for those transplants and, among these, the quality of donor organ is often considered a key factor [120,121,122]. Pancreas or pancreatic islet transplantation techniques are characterized by different phases, including organ procurement, preservation, and islet isolation, during which IRI processes occur that affect organ/islet function and survival. In this case, IRI not only impacts on transplant outcomes, but also reduces the number of pancreas/islets suitable for transplantation.
To address those issues, a more effective preservation before transplantation represents a promising strategy for improving the quality of the pancreas/islets, and to achieving complete and long-term insulin independence [123,124,125,126]. For the pancreas, the most common organ preservation solution used is the University of Wisconsin (UW) solution, which, however, does not prevent the deleterious effects of IRI [127,128,129]. Recently, in a T1D mouse model, Nishime et al. demonstrated that UW solution, supplemented with the antioxidant and cytoprotective agent AP39, mitigates cold IRI and promotes higher islet yields before and after purification. In this study, graft pancreatitis was prevented, and the outcome of islet transplantation was improved [130].
Across the last decade, it has been shown that the treatment with MSCs was able to induce islet regeneration, increase islet function, and inhibit immune system reactions in various animal models of T1D [131]. Kasahara et al. found that AdMSCs-CM administered through a preservation solution was able to restore rat islets to the condition they were in before transport, culture, and transplantation [132]. Similar results were found by Teratani et al., in which the MSC-derived CM added to the UW solution was able to preserve porcine islets during both transportation and cultures [133].
In the early phase after islet transplantation, the lack of vasculature and the hypoxic environment contribute to islet acute injury, mediated by ischemia [134]. In addition, after transplantation, graft failure continues to occur because of immunological reactions [135]. MSCs have the ability to both stimulate angiogenesis and attenuate inflammation [32], and are thus considered a useful therapeutic tool for alleviating IRI and improving islet transplantation outcomes. MSCs can be utilized in the early phase of the transplant to suppress immune-mediated rejection. Moreover, MSCs can provide a favorable environment for improving islet engraftment and stimulating their regeneration [136]. In a mouse model of allogenic islet transplantation, it has been shown that MSCs were able to prevent islet allograft rejection, leading to long-term normoglycemia. Ding et al. demonstrated that those effects were due to the MSCs’ suppressive activity on the immune system, which reduced hypersensitivity responses to allogeneic antigens, and allowed the survival of allogeneic islet grafts [137]. In a rat model of allogenic islet transplantation, Longoni et al. observed the ability of both syngeneic and allogeneic MSCs to prevent acute rejection and prolong graft function. In this study, the efficacy of MSCs was related to a reduction in inflammation, and this effect was independent of the administration route [138]. Li et al., in a diabetic mouse model, investigated the mechanisms by which the co-transplantation of MSCs and allogenic islets alleviated allograft rejection. They showed that MSCs exerted immunosuppressive effects through inhibition of both T lymphocytes and the activation of dendritic cells, resulting in the survival of transplanted pancreatic islets [139]. Promising results have also been obtained in non-human primates. Berman and colleagues showed that the intra-portal or intra-venous co-transplantation of MSCs and pancreatic islets significantly enhanced islet engraftment and function, and further infusions of MSCs were also able, in some cases, to avoid rejection and maintain islet function [140].
Therefore, thanks to their beneficial properties (anti-oxidant, proangiogenic, and immunomodulatory properties), MSC-based therapies can prevent IRI pathological processes during the transplantation of either the whole pancreas or pancreatic islets. Moreover, addition of MSCs or their secreted products to preservation solution during organ preservation or isolation of islets could conceivably offer a novel pharmacological approach to improving the outcome of pancreas/islet transplantation.

2.3. MSCs and Ischemia/Reperfusion Injury in the Kidney

Currently, renal transplantation is the principal treatment option for patients with end-stage kidney disease (ESKD), and IRI is an inevitable event during renal transplantation, with a significant impact on the function of transplanted kidneys [141]. IRI is the main cause of acute kidney injury (AKI) in surgery, and is characterized by endothelial cell activation, and leukocyte recruitment and infiltration, as well as renal cell necrosis and apoptosis [142]. In one study, it was found that treatment with MSC-derived EVs can ameliorate kidney fibrosis in a cisplatinum-induced AKI mouse model by reducing the levels of ROS and of pro-apoptotic molecules, such as 8-hydroxy-2-deoxyguanosine (8-OHdG), malonaldehyde (MDA), Bax, and caspase-3 [143]. The regenerative and anti-inflammatory properties of MSCs have been explored in a large number of animal injury models, explicating how these cells promote endothelial repair [144,145,146,147,148]. In several phase I clinical trials for kidney disease and transplantation, it has been found that the effects of MSCs on endothelial cells might lead to an improved kidney transplantation outcome [149,150,151,152,153]. Among the factors involved in the AKI pathogenesis, inflammatory response runs through the entire process of IRI-derived AKI, in which innate immunity, principally through macrophages, has a pivotal role in both injury and repair processes [154,155]. Macrophages can differentiate in two different phenotypes: M1, which produces pro-inflammatory molecules, and M2, which can inhibit inflammation and promote the repair of injured tissue [156]. Hence, the M1/M2 ratio can regulate the progress of AKI toward chronic kidney disease (CKD) or, conversely, kidney repair [157]. In the early stage of IRI, M1 macrophages are the prevalent inflammatory cells in kidney tissue, while M2 macrophages appear in later stages. Thus, to alleviate kidney injury, it is necessary to eliminate pro-inflammatory M1 macrophages before IRI [158]. The transition of macrophages from M1 to M2 phenotype induces the production of growth factors, such as platelet-derived growth factor (PDGF), transforming growth factor beta 1 (TGF-β1), vascular endothelial growth factor A (VEGF-A), and insulin-like growth factor I (IGF-1), which promote the regeneration and repair of renal tubular epithelial cells [157,159,160]. In a recent study using a mouse model of kidney IRI, it was demonstrated that EXOs from indoleamine 2,3-dioxygenase-(IDO)-overexpressing BM-MSCs accelerates tissue regeneration upon IRI by inducing the M1/M2 transition [65]. To date, most of the research and clinical trials have been focused mainly on MSC therapy after kidney transplantation, but not prior to it [152]. The systemic administration of MSCs to transplant recipients is safe; however, it has been shown that the cells never reach the kidney, mostly because intravenously infused MSCs are largely trapped in the lungs [161,162,163]. By contrast, the administration of MSCs to donor kidneys in an ex vivo isolated organ-perfusion system will deliver cells directly to the injured organ. With this purpose in mind, normothermic machine perfusion (NMP) of donor organs has been recognized as an opportunity to maintain organ viability and allow therapeutic interventions prior to transplantation [164,165,166]. Two different studies of MSC renal infusions during NMP on discarded human kidneys reported beneficial effects of MSCs. In one case, the potentiation of renal regeneration by the increased synthesis of adenosine triphosphate (ATP), reduction in inflammatory response, increased synthesis of growth factor, and the normalization of the cytoskeleton and mitosis was found [66]. In the other case, a significant reduction in IRI through the improvement of clinically relevant parameters, such as urine output and micro-vascular perfusion, and injury biomarkers, such as the downregulation of interleukin (IL)-1β, upregulation of IL-10 and IDO, as well as decreased neutrophil recruitment were reported [67]. In conclusion, the use of MSCs or their derivatives represents a promising approach to treating kidney IRI and/or improving kidney transplantation outcomes.

2.4. MSCs and Ischemia/Reperfusion Injury in the Gut

Intestinal IRI is a pathological process characterized by local vasoconstriction, thrombosis, mitochondrial damage, inflammatory response, cellular damage, and cell death [167], resulting in an impaired intestinal mucosal barrier function [168], associated with severe clinical conditions such as extracorporeal circulation, mesenteric artery thrombosis, strangulated ileus, trauma, abdominal aortic aneurysm surgery, and intestinal transplantation [169,170].
The visceral inflammation caused by intestinal IRI can alter the epithelial barrier morphology and function, allowing bacterial translocation [171], and their products as pathogen-associated molecular patterns (PAMPs), from the lumen into the lamina propria [172], thus driving inflammation [173]. This process eventually results in endotoxemia, the release of multiple pro-inflammatory cytokines [174], systemic inflammatory response syndrome (SIRS) [50], and even multi-organ failure and death [175,176,177,178]. Therefore, in patients with critical illness, the development of effective therapies and discovery of novel agents capable of ameliorating intestinal IRI are crucial for reducing mortality.
The potential of MSC therapy has been widely discussed for the treatment of end-stage organ ischemia. Even in intestinal IRI, these cells boost functional recovery and limit inflammation, while the exact mechanisms have not yet been defined (Figure 1). It has been postulated that MSCs may play a protective role against intestinal IRI via exogenous migration into the damaged intestinal tissue, and differentiation into intestinal epithelial cells (IECs), to enhance the integrity of the gut barrier or through the exogenous release of paracrine and/or endocrine cytokines with anti-inflammatory, anti-apoptotic, and pro-angiogenetic characteristics [50], principally mediated by EXOs [179]. As for the first mechanism of action, there are few studies demonstrating the cellular differentiation of MSCs within the damaged bowel as the primary mechanism by which MSCs attenuate intestinal IRI [180,181]. Most evidence supports the hypothesis that paracrine mechanisms drive the therapeutic efficacy of MSCs within damaged tissue [182,183]. One of the most promising molecular targets downregulated by MSC therapy is NF-kB, known to activate the expression of several genes expressed during intestinal IRI, and involved in the inflammatory response process [184], including TNF-α [185], IL-1β [186], IL-6, and ICAM [187]. These molecules drive complementary activation and subsequent production of inflammatory mediators, including IL-8, IL-17, and IL-18 [188]. The role of MSC immunomodulation in intestinal IRI acts on one side by decreasing production of inflammatory mediators, such as TNF-α [68], IL-1β [189], and IFN-γ [190], and by increasing anti-inflammatory cytokine production, via monocytes’ stimulation, such as IL-10 [191]. As an alternative mechanism of action, it has been demonstrated that BM-MSCs are able to attenuate intestinal IRI by reducing tight junction disruption and ZO-1 downregulation, thereby restoring the intestinal mucosal barrier, likely by regulating the levels of TNF-α [69,192].
Several studies have found that MSCs upregulate expression of numerous growth factors, such as VEGF, FGF2, and TGF-β by a p38 MAPK-dependent mechanism, resulting in enhanced tissue restoration [193,194]. Among the multiple roles played by MSCs, there is increasing evidence that the efficacy of MSCs resides in the EXOs released, carriers of a discrete set of proteins, and coding and non-coding RNA. In vivo experiments have shown that MSC-derived exosomal miR-34a/c-5p, and miR-29b-3p improved the intestinal barrier, thus alleviating intestinal IRI via the Snail/claudin pathway [195]. Recently, it was reported that BM-MSC EXOs can alleviate intestinal IRI through the PTEN/Akt/Nrf2 pathway by targeting miR-144-3p [42].

2.5. MSCs and Ischemia/Reperfusion Injury in the Heart

Ischemic heart diseases are the leading cause of mortality worldwide, with more than 2 million deaths in 2019, 16% of the world’s total (https://www.who.int/, accessed on 11 December 2022). Transplantation is a vital option for patients with challenging heart failure, despite the limitations imposed by organ availability. The shortage in organ supply obviously depends on the number of potential and actual donors, but also in large part on organ preservation. In fact, organ deterioration after donor explant is a key issue in this life-saving procedure. Perfusion has been adopted for prolonging organ storage, providing oxygen and nutrients, and maintaining the conditions for organ preservation. Though perfusion techniques have improved since the first heart transplant, in 1967 [196], organ transport for location and matching constitutes a very demanding problem in the prevention of damage caused by ischemia and hypoxia. Temperature lowering can reduce the cell death process with the release of autolytic enzymes, without altering cell metabolism; however, prolonged ischemia and hypothermia can induce oxidative processes [197], swelling [198], and acidosis [199]. In order to reduce these effects, several protective formulations have been developed [200]. Currently, another pioneering strategy has been considered together with cold and chemical preparations, and based on the protective action of MSCs on various organs and, in particular, heart injuries [201]. Though not less than two decades ago MSCs were viewed as playing a directly plastic role in heart remodeling after injury [70,202], further experiments have demonstrated that MSCs act mostly via paracrine effects (Figure 1). The first experiments revealed MSCs’ effects in in vivo animal models after MI, identifying cardiomyocyte transdifferentiation, and the integration into cardiac tissues [71,203]. However, further experiments have highlighted a limited amount of transdifferentiated MSC-derived cardiomyocytes [203,204]. This opened an important debate on the integration of MSCs into cardiac tissues after injection, and subsequent cardiomyocyte transdifferentiation [205]. Further evidence has demonstrated that the therapeutic effects of MSCs on the heart are based mostly on factors released, since their action is exerted in a paracrine mode by the secretome [206]. For example, it has been shown that MSC-derived EVs promote angiogenesis, an effect that can be ascribed principally to the miRNA content in EVs [207]. MiRNAs have been involved at various levels in cardiac repair or cardiomyocyte proliferation/aging [208,209] by modulating pathways implicated in the inhibition of fibrosis after MI in repair processes. MSC-derived EVs can promote cardiac protection, given that this treatment shows a positive effect in rat models on the MI heart by reducing scar-size formation, and stimulating angiogenesis [72]. Proteomic analysis of MSC EVs has revealed the presence of growth factor receptors such as PDGFR, cytokines, signaling, and adhesion molecules [210], which account for the induction of angiogenesis. The effect of MSCs in improving hypothermic perfusion in ex vivo organ preservation was recently tested in allograft in vivo models [73,74,75]. The authors here found that the MSC secretome considerably improved post-operatory heart functions in rat allograft experiments [75]. Moreover, they showed that hypoxic MSC-CM have cardioprotective effects by significantly reducing apoptotic indexes (e.g., TUNNEL, DNA breaks). At the same time, pro-inflammatory cytokines have been reduced by MSC-CM treatment, with a stronger lowering of IL-6 and TNF-α in the hypoxic MSC-CM compared to normoxic MSC-CM. Similarly, another group evaluated MSC-CM in heart transplantation after extended organ storage in 15-month-old rats, which is approximately comparable to a 40-year-old human [73]. Myocardial evaluation of 120 genes involved in apoptosis, oxidative stress, and inflammatory response showed a significant modification in gene expression signature by MSC-CM treatment. This was associated with an improvement in the cardiac functions, such as a reduced re-beating time after transplant, and an enhancement of the LV systolic and diastolic functions, with an increase in the pressure rate [73]. Likewise, comparable results have been obtained in mouse models, where the MSC secretome determined cardiac protection against IRI after prolonged organ preservation [74]. MSC-CM substantially reduced the presence of pro-inflammatory cytokines (TNF-α, IL-1β, and IL-6) in allotransplanted hearts. Moreover, the authors found the presence of miR-199a-3p in MSC EVs, which showed a cardioprotective effect in IRI [211]. miR-199a-3p expression in donor hearts is strongly reduced after prolonged cold preservation. Thus, MSC EXOs containing miR-199a-3p can restore myocardial levels in experimental allograft models. The opportunity to modulate the release of miRNAs by engineering EXOs is an additional option, together with a MSC-based therapeutic approach [212]. The MSC secretome may turn out to be an efficient cell-free therapeutic option for reducing organ damage, and mitigating the problems experienced by long and slow-moving waiting lists for this life-saving approach.

2.6. MSCs and Ischemia/Reperfusion Injury in the Brain

Cerebral IRI is a common event in ischemic stroke, induced by the impaired blood supply to the brain for a short period of time, followed by its restoration [213]. The impaired blood supply is usually caused by a blood pressure perturbation, and induced by the presence of a thrombus or embolus in vessels. Depending on the site, size, and duration of cerebral ischemia, it can cause permanent and irreversible brain tissue damage, including, in the worst-case scenario, neuronal cell death and cerebral infarctions [214]. In recent years, the literature has extensively described the main mechanisms involved in IRI. Among the most studied are oxidative stress, leukocyte infiltration, mitochondrial mechanisms, activation and aggregation of platelets, and the prompting of protein complement. Notably, a substantial number of studies supports the hypothesis that inflammation is a key factor in initiating the process at the base of the pathogenesis of cerebral IRI, which culminates in severe neurological dysfunction, such as blood–brain barrier (BBB) disruption, and edema or hemorrhagic transformation [213,215]. The disease is characterized by a robust activation and release of cytokines, chemokines, adhesion molecules, and proteolytic enzymes, which exacerbate tissue damage [216]. The current novel therapeutic approaches aim to modulate the neuroinflammatory response in order to reduce the inflammatory mediators involved in tissue damage [214,216]. Recent studies have demonstrated the strong immunomodulatory capacity of MSCs in the field of tissue regeneration. It has been found that they are able to inhibit tissue inflammation by directly or indirectly targeting the immune response, and promote tissue repair by the stimulation of endogenous cell function and revascularization of the damaged tissue [32,217]. MSCs can also inhibit the pro-inflammatory T-cell response, leading to a reduction in IFNγ, thus promoting an anti-inflammatory environment, which leads, in turn, to an increase in IL-10 production that induces the protective activity of Tregs [202,218,219]. The current data in the literature indicate that the administration of MSCs intracranially (intrastriatal or intracerebroventricular) or intravascularly (intra-arterial or intravenous) improves the restoration of tissue damage after ischemia both in mouse and rat models. The authors confirm that during cerebral stroke, the transplanted MSCs migrate to the damaged brain tissue site, inhibiting apoptosis and stimulating the expression of a series of growth factors, including brain-derived neurotrophic factor (BDNF), nerve growth factor (NGF), basic fibroblast growth factor (bFGF), IGF, HGF, VEGF, and angiogenic and stem cell factors, which synergistically prompt the recovery of the neuronal functions [76,220,221]. The recovery function carried out by MSCs also occurs through the induction of angiogenesis, the reduction in apoptosis, the restoration of synapses and dendrites, and the promotion of axonal regeneration and differentiation of autologous neuronal stem cells [77]. Recently, in vitro studies on astrocytes and neurons undergoing ischemic damage nicely demonstrated that MSC-EVs strongly protect the cells from intracellular Ca2+ accumulation, a biological effect of ischemia, and cell death, thus promoting tissue regeneration. In particular, the authors showed that the MSC-EVs acted by suppressing PI3K-Akt pathway. Furthermore, proteomic analysis on MSC-EVs revealed that the protective effect might be related to the EV-protein content, including factors such as HGF, CXCL1, VEGF-A, and MIF, all associated with brain regeneration [222]. Despite the fact that at present the MSC-evoked neurorestorative effects on IRI are not clear, recent clinical reports carried out on humans have highlighted a series of beneficial effects obtained in the treatment of ischemic stroke. Qiao et al. demonstrated the safety and feasibility of the co-transplantation of neural stem/progenitor cells (NSPCs) and UC-MSCs in patients with ischemic stroke, observing an improvement in neurological function and daily life activities [223]. Consonant with this study, Jing et al., in a small pilot study, demonstrated the safety and efficacy of allogeneic stem cells in treating strokes in the middle cerebral artery territory [224]. More recently, a series of clinical trials (phase 1/2) have confirmed that autologous MSCs, if delivered intravenously, are able to reduce post-stroke IRI damage, inducing a reduction in infarct size and an improvement in the functional outcome of the patients [225,226].

2.7. MSCs and Ischemia/Reperfusion Injury in the Liver

IRI in the liver occurs frequently during surgery for intrahepatic lesions, and is considered an inevitable injury during organ transplantation [227,228,229]. This is due mainly to the increasing number of potential recipients and the subsequent necessity of extending the criteria of liver eligibility for transplantation [230]. Thus, clinical needs require the identification of therapies that can attenuate the damage upon organ resection. Stem-cell therapy is currently considered an important approach, and has garnered attention in bench to bedside for the treatment of different diseases [32,231]. In particular, MSCs, because of their immunomodulatory effects and their tissue regeneration potential, have been used in liver surgery, including IRI [78,232,233]. Over the last decade, several studies from different research groups have used MSCs or their cellular products to detect protection from IRI, as well as to identify the molecular mechanisms underlining this biological effect (Figure 1). In vivo infusion of MSCs in mouse and rat models of liver IRI have shown an important protection against the injury. Pan et al., in 2012, found that BM-MSC infusion in IRI-injured rat livers protected the animals from the progression of the damage by reducing serum biomarkers of injury (AST and ALT), and by inhibiting neutrophil infiltration [234]. Furthermore, other studies have arrived at similar results in rat models, in which administration of Ad-MSCs inhibited hepatic apoptosis and increased tissue regeneration [79,235]. Reduced neutrophil infiltration upon BM-MSC infusion was confirmed a few years later by Li et al., who found that BM-MSCs stimulate MAPK phosphorylation and CXCR2 downregulation, both at transcriptional and protein levels. Taken together, these findings were associated with a significant reduction in the number of CD11+/CD18+ cells [80]. Along with the latter, MSC infusion in vivo inhibits pro-inflammatory phenotype during early organ reperfusion by modulating the transcription and the protein release of several pro-inflammatory cytokines, including TNF-α, IL-1β, and IL-6. More importantly, two different groups assessed the important role of MSCs (from bone marrow or adipose tissue) in regulating NLRP3 inflammasome activation, leading to reduced IL-1β and IL-18 protein release [236,237]. Interestingly, by using different mouse models, Li et al. discovered that BM-MSCs reduced NLRP3 activation via the Hippo/Wnt signaling axis [237]. MSCs either from bone marrow or adipose tissue have been employed in liver IRI attenuation because of their capacity to activate autophagy and mitophagy [81,238]. Wang et al. recently found that a BM-MSC infusion activates autophagy, as shown by increased levels of LC3b, a typical marker of terminal autophagy [239]. Moreover, they demonstrated that activation of BM-MSC-mediated autophagy depends on heme-oxygenase-1 (HO-1) upregulation. Furthermore, a recent paper by Zheng’s group showed that UC-MSCs activate mitophagy, a mitochondrial-related type of autophagy, thus promoting mitochondrial stability and cell survival [240]. Along the same line, the authors further confirmed mitochondrial protection by increased superoxide dismutase 1 (SOD1) activation and mitochondrial ROS (mtROS) inhibition after in vivo injection of UC-MSCs [240]. Endoplasmic reticulum stress (ERS), responsible for protein misfolding and subsequent cellular apoptosis, has recently been shown to be involved in liver IRI progression. Interestingly, it was recently found that Ad-MSCs attenuate ERS by downregulating ERS-related genes (ATF6 and XBP1), and by inhibiting hepatic apoptosis [241]. Viewed together, these findings clearly demonstrate that in vivo infusion of MSCs ameliorates liver IRI by tackling different biological processes that are usually responsible for the progression of the damage. However, in the last few years, mounting evidence suggests that the biological function exerted by MSCs might depend on their biological products, including the whole secretome or, more specifically, the EVs. As important cell–cell communicators, they have been indicated as a new therapeutic approach that could eventually replace infusion of MSCs, thus avoiding the risk of tumorigenicity, pulmonary embolism, and alloimmune response [82,242]. Independent of the source, MSC secretome and EVs have been shown to significantly reduce IRI damage by reducing ALT and AST blood levels, and by accelerating tissue regeneration upon injury [83,243]. In particular, Anger et al. demonstrated that treatment with UC-MSC-derived EVs was able to induce a drastic reduction in pro-inflammatory proteins, including HMGB1 and IL-1β, and a strong reduction in the expression of ICAM-1, a well-known adhesion molecule expressed in endothelial sinusoidal cells, and involved in neutrophil recruitment and infiltration. These data are in line with the results from Yao’s group [244]. In this study, the authors further characterized EV content, thus proposing Mn-SOD as a target protein involved in reduced IRI-mediated oxidative stress [244]. Moreover, UC-MSC EVs have been shown to inhibit CD4+ T cell activation in liver IRI. In particular, through unbiased proteomic approaches, the authors demonstrated that UC-MSC EVs contain CCT2, a Ca2+ modulator protein, which in turn downregulates NFAT1, thus reducing CD154 and the subsequent pro-inflammatory role of CD4+ T cells [245]. MSC EVs are enriched in miRNA, which are responsible for post-transcriptional changes in the target cells. Interestingly, only a few studies have identified potential miRNAs within MSC EVs that can attenuate liver IRI damage. For example, mir-1246 contained in UC-MSC EVs seems to alleviate liver IRI by reducing apoptosis and inflammation of parenchymal cells, and by modulating T helper/Treg balance [84,85]. Here, the authors showed that miR-1246 activates Wnt signaling in the target cells during IRI by inhibiting GSK3β, thus sustaining proliferation and inhibiting inflammation and apoptosis [85]. The same group in back-to-back studies demonstrated that miR-1246 modulates the T helper/Treg ratio by inhibiting the IL6ST-gp130-STAT3 signaling axis in an in vitro model of hepatic IRI [84]. Across recent years, mounting evidence suggests that MSC pre-conditioning might be useful to specify secretome content [32]. However, very few studies have focused on this type of approach in the context of liver IRI. In 2018, Sun et al. demonstrated in vivo that the 3D culture of UC-MSCs generates spheroids that, when implanted in an IRI-damaged liver in rats, attenuates the injury by inhibiting inflammation, neutrophil infiltration, and parenchymal apoptosis [246]. Finally, our group recently showed that 3D and IFN-γ priming of human amnion-derived MSCs (AMSCs) are able to modulate IRI damage in an in vitro model of IRI carried out on primary macrophages and hepatocytes. We found that several bioactive factors, including HGF, IL-10, IL-1RA, and BDNF, selectively inhibit inflammation of primary macrophages and apoptosis of IRI hepatocytes in vitro [22].

3. Discussion

The main biological processes activated during IRI are shared among the different types of organs where the damage occurs. As described above, inflammation, ROS production, and apoptosis of the parenchymal cells are responsible for the start and progress of the injury, leading eventually to organ rejection. Thus, it does not seem unusual that proteins and factors released during the injury are common in all the damaged tissues. For these reasons, using MSCs for the treatment of multi-organ IRI is currently considered a valid approach to reducing the injury (Figure 1) [21]. The scientific literature is replete with interesting studies that report the resolution of IRI after MSC treatment [50,56,67,75,80,226,247]. Because of their properties, MSCs can modulate the damage at different levels. For example, they strongly reduce inflammation, thus inhibiting the release of several pro-inflammatory cytokines, which in turn regulate neutrophil infiltration in the damaged tissue [192,248,249]. In addition, MSCs have been shown to block ROS production, thus improving mitochondrial damage [192]. Altogether, the inhibition of such biological mechanisms eventually leads to the reduction in parenchymal cell death by preventing organ rejection. Currently, there are seven clinical trials registered on clinicaltrials.gov, aimed at investigating MSC therapeutic efficacy for the treatment of IRI for both heart and kidney. Moreover, MSCs have demonstrated significant therapeutic effects in numerous preclinical IRI models (Table 1) and in the context of human solid-organ transplantation. In particular, in different preclinical human studies in which organs from deceased donors were not suitable for transplantation, it has been shown that MSCs were able to reduce IRI side effects in both lung and kidney in the setting of mechanical organ perfusion [60,61,66,67]. However, despite the safety of MSC treatment having been widely demonstrated for different diseases in numerous patients [250], several studies have also reported adverse events and side effects associated with MSC therapy [251]. For instance, the long-term cell culture of MSCs can lead to genetic abnormalities with consequent tumorigenic effects. Moreover, different clinical trials reported fibrosis and thromboembolism as the most common adverse events of MSCs therapy [251]. All these problems may be avoided by exploiting the paracrine properties of MSCs. Interestingly, in the recent past it has been demonstrated that the MSC secretome accounts for most of the positive effects of MSC therapy for IRI. The MSC secretome, containing biological factors that include EVs enriched in proteins and miRNAs, has been shown to be strongly involved in the modulation of the IRI phenotype for several reasons. First, it contains immunomodulatory factors, as well as pro-angiogenic and tissue repair properties, which by themselves seem to account for the majority of the therapeutic effects [20,22,252,253,254]. Second, because of the MSC secretome requirement for resolution of IRI, its use has recently been proposed instead of MSC transplantation. This has prompted reflection, as several studies have demonstrated that MSCs, when intravenously transplanted, die before tissue homing, becoming trapped in the lungs, or because they do not survive the highly hypoxic environments generated by ischemic tissues [162,255,256,257]. MSC paracrine effects seem to be more efficient, thus suggesting that MSC secretome-based therapy might be beneficial for IRI treatment. In the last few years, mounting evidence has suggested that MSC priming may be the future of MSC-based therapies. Many recent studies, mostly in vitro, have shown that MSC pre-conditioning might specify the secretome content, thus allowing the selection of the type of factors within the CM according to the disease to be tackled. For instance, it has been shown that IFNγ or IL-17 MSC priming generates a secretome enriched in anti-inflammatory factors, setting the stage for its use in inflammatory diseases, including IRI [22,258]. In addition, if the resolution of the injury requires tissue regeneration or angiogenesis, 3D culture priming, and hypoxia pre-conditioning stimulate the generation of MSC secretomes enriched in molecules, growth factors, and EVs required for tissue repair or formation of new vessels [259,260,261]. Very few clinical studies have described strategies to attenuate IRI damage, though the in vitro results obtained in the recent past have paved the way for further development of this novel approach in preclinical studies.

4. Conclusions

IRI damage currently represents an important impairment in organ surgery, given the variety of biological processes involved, and the multiple cell types targeted during the injury. Inflammation, ROS production, and apoptosis are the main consequences of the damage, thus therapeutic interventions are required and new strategies are under investigation. MSC-based cell therapy seems to be one of the most promising, considering the immuno-modulating and tissue regeneration properties provided by this cell type. However, several issues need to be addressed, as MSCs’ injection still has adverse effects in the short term, while no data are yet available in the long term. New alternatives are in place, which implies MSC-cell-free therapies; however, the studies are still in their infancy and further research needs to be performed before validation in pre-clinical human subjects.

Author Contributions

Conceptualization, G.Z. and V.M.; methodology, G.Z., V.M. and P.G.C.; investigation, M.B., A.G., R.B. and G.I.; writing—original draft preparation, G.Z., V.M., M.B., A.G., R.B. and G.I; writing—review and editing, G.Z., V.M. and P.G.C.; visualization, V.M.; supervision, P.G.C.; funding acquisition, P.G.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Ministero della Salute a valere sui fondi Ricerca Corrente. Italian Ministry of Health, Ricerca Corrente 2022 (Grant Number 23670065).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Dar, W.A.; Sulliv an, E.; Bynon, J.S.; Eltzschig, H.; Ju, C. Ischaemia reperfusion injury in liver transplantation: Cellular and molecular mechanisms. Liver Int. Off. J. Int. Assoc. Study Liver 2019, 39, 788–801. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Ito, T.; Naini, B.V.; Markovic, D.; Aziz, A.; Younan, S.; Lu, M.; Hirao, H.; Kadono, K.; Kojima, H.; DiNorcia, J., 3rd; et al. Ischemia-reperfusion injury and its relationship with early allograft dysfunction in liver transplant patients. Am. J. Transplant. Off. J. Am. Soc. Transplant. Am. Soc. Transpl. Surg. 2021, 21, 614–625. [Google Scholar] [CrossRef] [PubMed]
  3. Kalogeris, T.; Baines, C.P.; Krenz, M.; Korthuis, R.J. Cell biology of ischemia/reperfusion injury. Int. Rev. Cell Mol. Biol. 2012, 298, 229–317. [Google Scholar] [PubMed] [Green Version]
  4. Nieuwenhuijs-Moeke, G.J.; Pischke, S.E.; Berger, S.P.; Sanders, J.S.F.; Pol, R.A.; Struys, M.; Ploeg, R.J.; Leuvenink, H.G.D. Ischemia and Reperfusion Injury in Kidney Transplantation: Relevant Mechanisms in Injury and Repair. J. Clin. Med. 2020, 9, 253. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Kasimsetty, S.G.; Hawkes, A.; Barekatain, K.; Soo, E.; Welch, A.K.; McKay, D.B. TLR2 and NODs1 and 2 cooperate in inflammatory responses associated with renal ischemia reperfusion injury. Transpl. Immunol. 2020, 58, 101260. [Google Scholar] [CrossRef] [PubMed]
  6. Jaeschke, H. Molecular mechanisms of hepatic ischemia-reperfusion injury and preconditioning. Am. J. Physiol. Gastrointest. Liver Physiol. 2003, 284, G15–G26. [Google Scholar] [CrossRef]
  7. Pierro, A.; Eaton, S. Intestinal ischemia reperfusion injury and multisystem organ failure. Semin. Pediatr. Surg. 2004, 13, 11–17. [Google Scholar] [CrossRef]
  8. Abu-Amara, M.; Gurusamy, K.S.; Hori, S.; Glantzounis, G.; Fuller, B.; Davidson, B.R. Pharmacological interventions versus no pharmacological intervention for ischaemia reperfusion injury in liver resection surgery performed under vascular control. Cochrane Database Syst. Rev. 2009, 4, CD007472. [Google Scholar] [CrossRef]
  9. Kharbanda, R.K.; Nielsen, T.T.; Redington, A.N. Translation of remote ischaemic preconditioning into clinical practice. Lancet 2009, 374, 1557–1565. [Google Scholar] [CrossRef]
  10. Mockford, K.A.; Girn, H.R.; Homer-Vanniasinkam, S. Postconditioning: Current controversies and clinical implications. Eur. J. Vasc. Endovasc. Surg. Off. J. Eur. Soc. Vasc. Surg. 2009, 37, 437–442. [Google Scholar] [CrossRef] [Green Version]
  11. Ali, Z.A.; Callaghan, C.J.; Lim, E.; Ali, A.A.; Nouraei, S.A.; Akthar, A.M.; Boyle, J.R.; Varty, K.; Kharbanda, R.K.; Dutka, D.P.; et al. Remote ischemic preconditioning reduces myocardial and renal injury after elective abdominal aortic aneurysm repair: A randomized controlled trial. Circulation 2007, 116 (Suppl. S11), I98–I105. [Google Scholar] [CrossRef] [Green Version]
  12. Botker, H.E.; Kharbanda, R.; Schmidt, M.R.; Bottcher, M.; Kaltoft, A.K.; Terkelsen, C.J.; Munk, K.; Andersen, N.H.; Hansen, T.M.; Trautner, S.; et al. Remote ischaemic conditioning before hospital admission, as a complement to angioplasty, and effect on myocardial salvage in patients with acute myocardial infarction: A randomised trial. Lancet 2010, 375, 727–734. [Google Scholar] [CrossRef] [PubMed]
  13. Kaur, S.; Jaggi, A.S.; Singh, N. Molecular aspects of ischaemic postconditioning. Fundam. Clin. Pharmacol. 2009, 23, 521–536. [Google Scholar] [CrossRef] [PubMed]
  14. Zhao, Z.Q. Postconditioning in reperfusion injury: A status report. Cardiovasc. Drugs Ther. 2010, 24, 265–279. [Google Scholar] [CrossRef]
  15. Huerter, M.E.; Sharma, A.K.; Zhao, Y.; Charles, E.J.; Kron, I.L.; Laubach, V.E. Attenuation of Pulmonary Ischemia-Reperfusion Injury by Adenosine A2B Receptor Antagonism. Ann. Thorac. Surg. 2016, 102, 385–393. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Kertai, M.D.; Boersma, E.; Westerhout, C.M.; van Domburg, R.; Klein, J.; Bax, J.J.; van Urk, H.; Poldermans, D. Association between long-term statin use and mortality after successful abdominal aortic aneurysm surgery. Am. J. Med. 2004, 116, 96–103. [Google Scholar] [CrossRef] [PubMed]
  17. Laws, P.E.; Spark, J.I.; Cowled, P.A.; Fitridge, R.A. The role of statins in vascular disease. Eur. J. Vasc. Endovasc. Surg. Off. J. Eur. Soc. Vasc. Surg. 2004, 27, 6–16. [Google Scholar] [CrossRef] [PubMed]
  18. Martens, A.; Boada, M.; Vanaudenaerde, B.M.; Verleden, S.E.; Vos, R.; Verleden, G.M.; Verbeken, E.K.; Van Raemdonck, D.; Schols, D.; Claes, S.; et al. Steroids can reduce warm ischemic reperfusion injury in a porcine donation after circulatory death model with ex vivo lung perfusion evaluation. Transpl. Int. Off. J. Eur. Soc. Organ Transplant. 2016, 29, 1237–1246. [Google Scholar]
  19. Miceli, V.; Bertani, A. Mesenchymal Stromal/Stem Cells and Their Products as a Therapeutic Tool to Advance Lung Transplantation. Cells 2022, 11, 826. [Google Scholar] [CrossRef]
  20. Miceli, V.; Bertani, A.; Chinnici, C.M.; Bulati, M.; Pampalone, M.; Amico, G.; Carcione, C.; Schmelzer, E.; Gerlach, J.C.; Conaldi, P.G. Conditioned Medium from Human Amnion-Derived Mesenchymal Stromal/Stem Cells Attenuating the Effects of Cold Ischemia-Reperfusion Injury in an In Vitro Model Using Human Alveolar Epithelial Cells. Int. J. Mol. Sci. 2021, 22, 510. [Google Scholar] [CrossRef]
  21. Rowart, P.; Erpicum, P.; Detry, O.; Weekers, L.; Gregoire, C.; Lechanteur, C.; Briquet, A.; Beguin, Y.; Krzesinski, J.M.; Jouret, F. Mesenchymal Stromal Cell Therapy in Ischemia/Reperfusion Injury. J. Immunol. Res. 2015, 2015, 602597. [Google Scholar] [CrossRef] [Green Version]
  22. Zito, G.; Miceli, V.; Carcione, C.; Busa, R.; Bulati, M.; Gallo, A.; Iannolo, G.; Pagano, D.; Conaldi, P.G. Human Amnion-Derived Mesenchymal Stromal/Stem Cells Pre-Conditioning Inhibits Inflammation and Apoptosis of Immune and Parenchymal Cells in an In Vitro Model of Liver Ischemia/Reperfusion. Cells 2022, 11, 709. [Google Scholar] [CrossRef]
  23. Miranda, J.P.; Camoes, S.P.; Gaspar, M.M.; Rodrigues, J.S.; Carvalheiro, M.; Barcia, R.N.; Cruz, P.; Cruz, H.; Simoes, S.; Santos, J.M. The Secretome Derived From 3D-Cultured Umbilical Cord Tissue MSCs Counteracts Manifestations Typifying Rheumatoid Arthritis. Front. Immunol. 2019, 10, 18. [Google Scholar] [CrossRef]
  24. Colter, D.C.; Sekiya, I.; Prockop, D.J. Identification of a subpopulation of rapidly self-renewing and multipotential adult stem cells in colonies of human marrow stromal cells. Proc. Natl. Acad. Sci. USA 2001, 98, 7841–7845. [Google Scholar] [CrossRef] [Green Version]
  25. Zuk, P.A.; Zhu, M.; Ashjian, P.; De Ugarte, D.A.; Huang, J.I.; Mizuno, H.; Alfonso, Z.C.; Fraser, J.K.; Benhaim, P.; Hedrick, M.H. Human adipose tissue is a source of multipotent stem cells. Mol. Biol. Cell 2002, 13, 4279–4295. [Google Scholar] [CrossRef]
  26. Magatti, M.; De Munari, S.; Vertua, E.; Gibelli, L.; Wengler, G.S.; Parolini, O. Human amnion mesenchyme harbors cells with allogeneic T-cell suppression and stimulation capabilities. Stem Cells 2008, 26, 182–192. [Google Scholar] [CrossRef]
  27. Keating, A. Mesenchymal stromal cells: New directions. Cell Stem Cell 2012, 10, 709–716. [Google Scholar] [CrossRef] [Green Version]
  28. Mendez-Ferrer, S.; Michurina, T.V.; Ferraro, F.; Mazloom, A.R.; Macarthur, B.D.; Lira, S.A.; Scadden, D.T.; Ma’ayan, A.; Enikolopov, G.N.; Frenette, P.S. Mesenchymal and haematopoietic stem cells form a unique bone marrow niche. Nature 2010, 466, 829–834. [Google Scholar] [CrossRef] [Green Version]
  29. Baldari, S.; Di Rocco, G.; Piccoli, M.; Pozzobon, M.; Muraca, M.; Toietta, G. Challenges and Strategies for Improving the Regenerative Effects of Mesenchymal Stromal Cell-Based Therapies. Int. J. Mol. Sci. 2017, 18, 2087. [Google Scholar] [CrossRef] [Green Version]
  30. Chang, T.H.; Wu, C.S.; Chiou, S.H.; Chang, C.H.; Liao, H.J. Adipose-Derived Stem Cell Exosomes as a Novel Anti-Inflammatory Agent and the Current Therapeutic Targets for Rheumatoid Arthritis. Biomedicines 2022, 10, 1725. [Google Scholar] [CrossRef]
  31. Kolios, G.; Paspaliaris, V. Mesenchyme Stem Cell-Derived Conditioned Medium as a Potential Therapeutic Tool in Idiopathic Pulmonary Fibrosis. Biomedicines 2022, 10, 2298. [Google Scholar] [CrossRef]
  32. Miceli, V.; Bulati, M.; Iannolo, G.; Zito, G.; Gallo, A.; Conaldi, P.G. Therapeutic Properties of Mesenchymal Stromal/Stem Cells: The Need of Cell Priming for Cell-Free Therapies in Regenerative Medicine. Int. J. Mol. Sci. 2021, 22, 763. [Google Scholar] [CrossRef]
  33. Wu, Y.; Hoogduijn, M.J.; Baan, C.C.; Korevaar, S.S.; de Kuiper, R.; Yan, L.; Wang, L.; van Besouw, N.M. Adipose Tissue-Derived Mesenchymal Stem Cells Have a Heterogenic Cytokine Secretion Profile. Stem Cells Int. 2017, 2017, 4960831. [Google Scholar] [CrossRef] [Green Version]
  34. Alberti, G.; Russo, E.; Corrao, S.; Anzalone, R.; Kruzliak, P.; Miceli, V.; Conaldi, P.G.; Di Gaudio, F.; La Rocca, G. Current Perspectives on Adult Mesenchymal Stromal Cell-Derived Extracellular Vesicles: Biological Features and Clinical Indications. Biomedicines 2022, 10, 2822. [Google Scholar] [CrossRef]
  35. Cittadini, E.; Brucculeri, A.M.; Quartararo, F.; Vaglica, R.; Miceli, V.; Conaldi, P.G. Stem cell therapy in the treatment of organic and dysfunctional endometrial pathology. Minerva Obstet. Gynecol. 2022, 74, 504–515. [Google Scholar] [CrossRef]
  36. Garnier, D.; Ratcliffe, E.; Briand, J.; Cartron, P.F.; Oliver, L.; Vallette, F.M. The Activation of Mesenchymal Stem Cells by Glioblastoma Microvesicles Alters Their Exosomal Secretion of miR-100-5p, miR-9-5p and let-7d-5p. Biomedicines 2022, 10, 112. [Google Scholar] [CrossRef]
  37. Soni, N.; Gupta, S.; Rawat, S.; Krishnakumar, V.; Mohanty, S.; Banerjee, A. MicroRNA-Enriched Exosomes from Different Sources of Mesenchymal Stem Cells Can Differentially Modulate Functions of Immune Cells and Neurogenesis. Biomedicines 2021, 10, 69. [Google Scholar] [CrossRef]
  38. Vu, H.T.; Han, M.R.; Lee, J.H.; Kim, J.S.; Shin, J.S.; Yoon, J.Y.; Park, J.H.; Dashnyam, K.; Knowles, J.C.; Lee, H.H.; et al. Investigating the Effects of Conditioned Media from Stem Cells of Human Exfoliated Deciduous Teeth on Dental Pulp Stem Cells. Biomedicines 2022, 10, 906. [Google Scholar] [CrossRef]
  39. Li, H.; Xia, Z.; Chen, Y.; Qi, D.; Zheng, H. Mechanism and Therapies of Oxidative Stress-Mediated Cell Death in Ischemia Reperfusion Injury. Oxidative Med. Cell. Longev. 2018, 2018, 2910643. [Google Scholar] [CrossRef]
  40. Zhang, H.; Yan, Q.; Wang, X.; Chen, X.; Chen, Y.; Du, J.; Chen, L. The Role of Mitochondria in Liver Ischemia-Reperfusion Injury: From Aspects of Mitochondrial Oxidative Stress, Mitochondrial Fission, Mitochondrial Membrane Permeable Transport Pore Formation, Mitophagy, and Mitochondria-Related Protective Measures. Oxidative Med. Cell. Longev. 2021, 2021, 6670579. [Google Scholar] [CrossRef]
  41. Pacienza, N.; Santa-Cruz, D.; Malvicini, R.; Robledo, O.; Lemus-Larralde, G.; Bertolotti, A.; Marcos, M.; Yannarelli, G. Mesenchymal Stem Cell Therapy Facilitates Donor Lung Preservation by Reducing Oxidative Damage during Ischemia. Stem Cells Int. 2019, 2019, 8089215. [Google Scholar] [CrossRef] [Green Version]
  42. Zhang, G.; Wan, Z.; Liu, Z.; Liu, D.; Zhao, Z.; Leng, Y. Exosomes Derived from BMSCs Ameliorate Intestinal Ischemia-Reperfusion Injury by Regulating miR-144-3p-Mediated Oxidative Stress. Dig. Dis. Sci. 2022, 67, 5090–5106. [Google Scholar] [CrossRef]
  43. Esrefoglu, M. Role of stem cells in repair of liver injury: Experimental and clinical benefit of transferred stem cells on liver failure. World J. Gastroenterol. 2013, 19, 6757–6773. [Google Scholar] [CrossRef]
  44. Meier, R.P.; Muller, Y.D.; Morel, P.; Gonelle-Gispert, C.; Buhler, L.H. Transplantation of mesenchymal stem cells for the treatment of liver diseases, is there enough evidence? Stem Cell Res. 2013, 11, 1348–1364. [Google Scholar] [CrossRef]
  45. Hu, H.; Zou, C. Mesenchymal Stem Cells in Renal Ischemia-Reperfusion Injury: Biological and Therapeutic Perspectives. Curr. Stem Cell Res. Ther. 2017, 12, 183–187. [Google Scholar] [CrossRef]
  46. Lai, T.S.; Wang, Z.H.; Cai, S.X. Mesenchymal stem cell attenuates neutrophil-predominant inflammation and acute lung injury in an in vivo rat model of ventilator-induced lung injury. Chin. Med. J. 2015, 128, 361–367. [Google Scholar] [CrossRef]
  47. Lee, J.W.; Fang, X.; Krasnodembskaya, A.; Howard, J.P.; Matthay, M.A. Concise review: Mesenchymal stem cells for acute lung injury: Role of paracrine soluble factors. Stem Cells 2011, 29, 913–919. [Google Scholar] [CrossRef] [Green Version]
  48. Li, J.; Peng, Q.; Yang, R.; Li, K.; Zhu, P.; Zhu, Y.; Zhou, P.; Szabo, G.; Zheng, S. Application of Mesenchymal Stem Cells During Machine Perfusion: An Emerging Novel Strategy for Organ Preservation. Front. Immunol. 2021, 12, 713920. [Google Scholar] [CrossRef]
  49. Mordant, P.; Nakajima, D.; Kalaf, R.; Iskender, I.; Maahs, L.; Behrens, P.; Coutinho, R.; Iyer, R.K.; Davies, J.E.; Cypel, M.; et al. Mesenchymal stem cell treatment is associated with decreased perfusate concentration of interleukin-8 during ex vivo perfusion of donor lungs after 18-hour preservation. J. Heart Lung Transplant. Off. Publ. Int. Soc. Heart Transplant. 2016, 35, 1245–1254. [Google Scholar] [CrossRef]
  50. Shi, Y.; Zhang, X.; Wan, Z.; Liu, X.; Chen, F.; Zhang, J.; Leng, Y. Mesenchymal stem cells against intestinal ischemia-reperfusion injury: A systematic review and meta-analysis of preclinical studies. Stem Cell Res. Ther. 2022, 13, 216. [Google Scholar] [CrossRef]
  51. Wang, Y.; Chen, X.; Cao, W.; Shi, Y. Plasticity of mesenchymal stem cells in immunomodulation: Pathological and therapeutic implications. Nat. Immunol. 2014, 15, 1009–1016. [Google Scholar] [CrossRef]
  52. Fouraschen, S.M.; Pan, Q.; de Ruiter, P.E.; Farid, W.R.; Kazemier, G.; Kwekkeboom, J.; Ijzermans, J.N.; Metselaar, H.J.; Tilanus, H.W.; de Jonge, J.; et al. Secreted factors of human liver-derived mesenchymal stem cells promote liver regeneration early after partial hepatectomy. Stem Cells Dev. 2012, 21, 2410–2419. [Google Scholar] [CrossRef]
  53. Ionescu, L.; Byrne, R.N.; van Haaften, T.; Vadivel, A.; Alphonse, R.S.; Rey-Parra, G.J.; Weissmann, G.; Hall, A.; Eaton, F.; Thebaud, B. Stem cell conditioned medium improves acute lung injury in mice: In vivo evidence for stem cell paracrine action. Am. J. Physiol. Lung Cell. Mol. Physiol. 2012, 303, L967–L977. [Google Scholar] [CrossRef] [Green Version]
  54. Lavoie, J.R.; Rosu-Myles, M. Uncovering the secretes of mesenchymal stem cells. Biochimie 2013, 95, 2212–2221. [Google Scholar] [CrossRef]
  55. Tian, W.; Liu, Y.; Zhang, B.; Dai, X.; Li, G.; Li, X.; Zhang, Z.; Du, C.; Wang, H. Infusion of mesenchymal stem cells protects lung transplants from cold ischemia-reperfusion injury in mice. Lung 2015, 193, 85–95. [Google Scholar] [CrossRef]
  56. Lin, K.C.; Yeh, J.N.; Chen, Y.L.; Chiang, J.Y.; Sung, P.H.; Lee, F.Y.; Guo, J.; Yip, H.K. Xenogeneic and Allogeneic Mesenchymal Stem Cells Effectively Protect the Lung Against Ischemia-reperfusion Injury Through Downregulating the Inflammatory, Oxidative Stress, and Autophagic Signaling Pathways in Rat. Cell Transplant. 2020, 29, 963689720954140. [Google Scholar] [CrossRef]
  57. Lu, W.; Si, Y.I.; Ding, J.; Chen, X.; Zhang, X.; Dong, Z.; Fu, W. Mesenchymal stem cells attenuate acute ischemia-reperfusion injury in a rat model. Exp. Ther. Med. 2015, 10, 2131–2137. [Google Scholar] [CrossRef] [Green Version]
  58. Sun, C.K.; Yen, C.H.; Lin, Y.C.; Tsai, T.H.; Chang, L.T.; Kao, Y.H.; Chua, S.; Fu, M.; Ko, S.F.; Leu, S.; et al. Autologous transplantation of adipose-derived mesenchymal stem cells markedly reduced acute ischemia-reperfusion lung injury in a rodent model. J. Transl. Med. 2011, 9, 118. [Google Scholar] [CrossRef] [Green Version]
  59. Chen, S.; Chen, L.; Wu, X.; Lin, J.; Fang, J.; Chen, X.; Wei, S.; Xu, J.; Gao, Q.; Kang, M. Ischemia postconditioning and mesenchymal stem cells engraftment synergistically attenuate ischemia reperfusion-induced lung injury in rats. J. Surg. Res. 2012, 178, 81–91. [Google Scholar] [CrossRef]
  60. La Francesca, S.; Ting, A.E.; Sakamoto, J.; Rhudy, J.; Bonenfant, N.R.; Borg, Z.D.; Cruz, F.F.; Goodwin, M.; Lehman, N.A.; Taggart, J.M.; et al. Multipotent adult progenitor cells decrease cold ischemic injury in ex vivo perfused human lungs: An initial pilot and feasibility study. Transplant. Res. 2014, 3, 19. [Google Scholar] [CrossRef] [Green Version]
  61. McAuley, D.F.; Curley, G.F.; Hamid, U.I.; Laffey, J.G.; Abbott, J.; McKenna, D.H.; Fang, X.; Matthay, M.A.; Lee, J.W. Clinical grade allogeneic human mesenchymal stem cells restore alveolar fluid clearance in human lungs rejected for transplantation. Am. J. Physiol. Lung Cell. Mol. Physiol. 2014, 306, L809–L815. [Google Scholar] [CrossRef] [Green Version]
  62. Lonati, C.; Bassani, G.A.; Brambilla, D.; Leonardi, P.; Carlin, A.; Maggioni, M.; Zanella, A.; Dondossola, D.; Fonsato, V.; Grange, C.; et al. Mesenchymal stem cell-derived extracellular vesicles improve the molecular phenotype of isolated rat lungs during ischemia/reperfusion injury. J. Heart Lung Transplant. Off. Publ. Int. Soc. Heart Transplant. 2019, 38, 1306–1316. [Google Scholar] [CrossRef]
  63. Stone, M.L.; Zhao, Y.; Robert Smith, J.; Weiss, M.L.; Kron, I.L.; Laubach, V.E.; Sharma, A.K. Mesenchymal stromal cell-derived extracellular vesicles attenuate lung ischemia-reperfusion injury and enhance reconditioning of donor lungs after circulatory death. Respir. Res. 2017, 18, 212. [Google Scholar] [CrossRef]
  64. Hwang, B.; Liles, W.C.; Waworuntu, R.; Mulligan, M.S. Pretreatment with bone marrow-derived mesenchymal stromal cell-conditioned media confers pulmonary ischemic tolerance. J. Thorac. Cardiovasc. Surg. 2016, 151, 841–849. [Google Scholar] [CrossRef] [Green Version]
  65. Xie, X.; Yang, X.; Wu, J.; Tang, S.; Yang, L.; Fei, X.; Wang, M. Exosome from indoleamine 2,3-dioxygenase-overexpressing bone marrow mesenchymal stem cells accelerates repair process of ischemia/reperfusion-induced acute kidney injury by regulating macrophages polarization. Stem Cell Res. Ther. 2022, 13, 367. [Google Scholar] [CrossRef]
  66. Brasile, L.; Henry, N.; Orlando, G.; Stubenitsky, B. Potentiating Renal Regeneration Using Mesenchymal Stem Cells. Transplantation 2019, 103, 307–313. [Google Scholar] [CrossRef]
  67. Thompson, E.R.; Bates, L.; Ibrahim, I.K.; Sewpaul, A.; Stenberg, B.; McNeill, A.; Figueiredo, R.; Girdlestone, T.; Wilkins, G.C.; Wang, L.; et al. Novel delivery of cellular therapy to reduce ischemia reperfusion injury in kidney transplantation. Am. J. Transplant. Off. J. Am. Soc. Transplant. Am. Soc. Transpl. Surg. 2021, 21, 1402–1414. [Google Scholar] [CrossRef]
  68. Jiang, H.; Qu, L.; Dou, R.; Lu, L.; Bian, S.; Zhu, W. Potential role of mesenchymal stem cells in alleviating intestinal ischemia/reperfusion impairment. PLoS ONE 2013, 8, e74468. [Google Scholar] [CrossRef]
  69. Jiang, H.; Qu, L.; Li, Y.; Gu, L.; Shi, Y.; Zhang, J.; Zhu, W.; Li, J. Bone marrow mesenchymal stem cells reduce intestinal ischemia/reperfusion injuries in rats. J. Surg. Res. 2011, 168, 127–134. [Google Scholar] [CrossRef]
  70. Shake, J.G.; Gruber, P.J.; Baumgartner, W.A.; Senechal, G.; Meyers, J.; Redmond, J.M.; Pittenger, M.F.; Martin, B.J. Mesenchymal stem cell implantation in a swine myocardial infarct model: Engraftment and functional effects. Ann. Thorac. Surg. 2002, 73, 1919–1925. [Google Scholar] [CrossRef]
  71. Kimura, M.; Toyoda, M.; Gojo, S.; Itakura, Y.; Kami, D.; Miyoshi, S.; Kyo, S.; Ono, M.; Umezawa, A. Allogeneic amniotic membrane-derived mesenchymal stromal cell transplantation in a porcine model of chronic myocardial ischemia. J. Stem Cells Regen. Med. 2012, 8, 171–180. [Google Scholar]
  72. Bian, S.; Zhang, L.; Duan, L.; Wang, X.; Min, Y.; Yu, H. Extracellular vesicles derived from human bone marrow mesenchymal stem cells promote angiogenesis in a rat myocardial infarction model. J. Mol. Med. 2014, 92, 387–397. [Google Scholar] [CrossRef] [PubMed]
  73. Korkmaz-Icoz, S.; Li, S.; Huttner, R.; Ruppert, M.; Radovits, T.; Loganathan, S.; Sayour, A.A.; Brlecic, P.; Lasitschka, F.; Karck, M.; et al. Hypothermic perfusion of donor heart with a preservation solution supplemented by mesenchymal stem cells. J. Heart Lung Transplant. Off. Publ. Int. Soc. Heart Transplant. 2019, 38, 315–326. [Google Scholar] [CrossRef] [PubMed]
  74. Wang, M.; Yan, L.; Li, Q.; Yang, Y.; Turrentine, M.; March, K.; Wang, I.W. Mesenchymal stem cell secretions improve donor heart function following ex vivo cold storage. J. Thorac. Cardiovasc. Surg. 2022, 163, e277–e292. [Google Scholar] [CrossRef]
  75. Zhou, P.; Liu, H.; Liu, X.; Ling, X.; Xiao, Z.; Zhu, P.; Zhu, Y.; Lu, J.; Zheng, S. Donor heart preservation with hypoxic-conditioned medium-derived from bone marrow mesenchymal stem cells improves cardiac function in a heart transplantation model. Stem Cell Res. Ther. 2021, 12, 56. [Google Scholar] [CrossRef] [PubMed]
  76. Li, Y.; Chopp, M.; Chen, J.; Wang, L.; Gautam, S.C.; Xu, Y.X.; Zhang, Z. Intrastriatal transplantation of bone marrow nonhematopoietic cells improves functional recovery after stroke in adult mice. J. Cereb. Blood Flow Metab. Off. J. Int. Soc. Cereb. Blood Flow Metab. 2000, 20, 1311–1319. [Google Scholar] [CrossRef] [Green Version]
  77. Zhang, J.; Li, Y.; Chen, J.; Yang, M.; Katakowski, M.; Lu, M.; Chopp, M. Expression of insulin-like growth factor 1 and receptor in ischemic rats treated with human marrow stromal cells. Brain Res. 2004, 1030, 19–27. [Google Scholar] [CrossRef]
  78. Saidi, R.F.; Rajeshkumar, B.; Shariftabrizi, A.; Bogdanov, A.A.; Zheng, S.; Dresser, K.; Walter, O. Human adipose-derived mesenchymal stem cells attenuate liver ischemia-reperfusion injury and promote liver regeneration. Surgery 2014, 156, 1225–1231. [Google Scholar] [CrossRef]
  79. Kanazawa, H.; Fujimoto, Y.; Teratani, T.; Iwasaki, J.; Kasahara, N.; Negishi, K.; Tsuruyama, T.; Uemoto, S.; Kobayashi, E. Bone marrow-derived mesenchymal stem cells ameliorate hepatic ischemia reperfusion injury in a rat model. PLoS ONE 2011, 6, e19195. [Google Scholar] [CrossRef] [Green Version]
  80. Li, S.; Zheng, X.; Li, H.; Zheng, J.; Chen, X.; Liu, W.; Tai, Y.; Zhang, Y.; Wang, G.; Yang, Y. Mesenchymal Stem Cells Ameliorate Hepatic Ischemia/Reperfusion Injury via Inhibition of Neutrophil Recruitment. J. Immunol. Res. 2018, 2018, 7283703. [Google Scholar] [CrossRef] [Green Version]
  81. Li, J.; Zhou, J.; Zhang, D.; Song, Y.; She, J.; Bai, C. Bone marrow-derived mesenchymal stem cells enhance autophagy via PI3K/AKT signalling to reduce the severity of ischaemia/reperfusion-induced lung injury. J. Cell. Mol. Med. 2015, 19, 2341–2351. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Du, Z.; Wei, C.; Yan, J.; Han, B.; Zhang, M.; Peng, C.; Liu, Y. Mesenchymal stem cells overexpressing C-X-C chemokine receptor type 4 improve early liver regeneration of small-for-size liver grafts. Liver Transplant. Off. Publ. Am. Assoc. Study Liver Dis. Int. Liver Transplant. Soc. 2013, 19, 215–225. [Google Scholar] [CrossRef] [PubMed]
  83. Anger, F.; Camara, M.; Ellinger, E.; Germer, C.T.; Schlegel, N.; Otto, C.; Klein, I. Human Mesenchymal Stromal Cell-Derived Extracellular Vesicles Improve Liver Regeneration After Ischemia Reperfusion Injury in Mice. Stem Cells Dev. 2019, 28, 1451–1462. [Google Scholar] [CrossRef] [PubMed]
  84. Xie, K.; Liu, L.; Chen, J.; Liu, F. Exosomal miR-1246 derived from human umbilical cord blood mesenchymal stem cells attenuates hepatic ischemia reperfusion injury by modulating T helper 17/regulatory T balance. IUBMB Life 2019, 71, 2020–2030. [Google Scholar] [CrossRef] [PubMed]
  85. Xie, K.; Liu, L.; Chen, J.; Liu, F. Exosomes derived from human umbilical cord blood mesenchymal stem cells improve hepatic ischemia reperfusion injury via delivering miR-1246. Cell Cycle 2019, 18, 3491–3501. [Google Scholar] [CrossRef] [PubMed]
  86. Chen-Yoshikawa, T.F. Ischemia-Reperfusion Injury in Lung Transplantation. Cells 2021, 10, 1333. [Google Scholar] [CrossRef]
  87. Jin, Z.; Suen, K.C.; Wang, Z.; Ma, D. Review 2: Primary graft dysfunction after lung transplant-pathophysiology, clinical considerations and therapeutic targets. J. Anesth. 2020, 34, 729–740. [Google Scholar] [CrossRef]
  88. De Perrot, M.; Liu, M.; Waddell, T.K.; Keshavjee, S. Ischemia-reperfusion-induced lung injury. Am. J. Respir. Crit. Care Med. 2003, 167, 490–511. [Google Scholar] [CrossRef]
  89. Kawashima, M.; Juvet, S.C. The role of innate immunity in the long-term outcome of lung transplantation. Ann. Transl. Med. 2020, 8, 412. [Google Scholar] [CrossRef]
  90. Laubach, V.E.; Sharma, A.K. Mechanisms of lung ischemia-reperfusion injury. Curr. Opin. Organ Transplant. 2016, 21, 246–252. [Google Scholar] [CrossRef] [Green Version]
  91. Van der Mark, S.C.; Hoek, R.A.S.; Hellemons, M.E. Developments in lung transplantation over the past decade. Eur. Respir. Rev. Off. J. Eur. Respir. Soc. 2020, 29, 190132. [Google Scholar] [CrossRef]
  92. Gagliotti, C.; Morsillo, F.; Moro, M.L.; Masiero, L.; Procaccio, F.; Vespasiano, F.; Pantosti, A.; Monaco, M.; Errico, G.; Ricci, A.; et al. Infections in liver and lung transplant recipients: A national prospective cohort. Eur. J. Clin. Microbiol. Infect. Dis. Off. Publ. Eur. Soc. Clin. Microbiol. 2018, 37, 399–407. [Google Scholar] [CrossRef] [PubMed]
  93. Gallo, A.; Cuscino, N.; Contino, F.; Bulati, M.; Pampalone, M.; Amico, G.; Zito, G.; Carcione, C.; Centi, C.; Bertani, A.; et al. Changes in the Transcriptome Profiles of Human Amnion-Derived Mesenchymal Stromal/Stem Cells Induced by Three-Dimensional Culture: A Potential Priming Strategy to Improve Their Properties. Int. J. Mol. Sci. 2022, 23, 863. [Google Scholar] [CrossRef]
  94. Lo Nigro, A.; Gallo, A.; Bulati, M.; Vitale, G.; Paini, D.S.; Pampalone, M.; Galvagno, D.; Conaldi, P.G.; Miceli, V. Amnion-Derived Mesenchymal Stromal/Stem Cell Paracrine Signals Potentiate Human Liver Organoid Differentiation: Translational Implications for Liver Regeneration. Front. Med. 2021, 8, 746298. [Google Scholar] [CrossRef]
  95. Miceli, V.; Chinnici, C.M.; Bulati, M.; Pampalone, M.; Amico, G.; Schmelzer, E.; Gerlach, J.C.; Conaldi, P.G. Comparative study of the production of soluble factors in human placenta-derived mesenchymal stromal/stem cells grown in adherent conditions or as aggregates in a catheter-like device. Biochem. Biophys. Res. Commun. 2020, 522, 171–176. [Google Scholar] [CrossRef]
  96. Miceli, V.; Pampalone, M.; Vella, S.; Carreca, A.P.; Amico, G.; Conaldi, P.G. Comparison of Immunosuppressive and Angiogenic Properties of Human Amnion-Derived Mesenchymal Stem Cells between 2D and 3D Culture Systems. Stem Cells Int. 2019, 2019, 7486279. [Google Scholar] [CrossRef]
  97. Schmelzer, E.; Miceli, V.; Chinnici, C.M.; Bertani, A.; Gerlach, J.C. Effects of Mesenchymal Stem Cell Coculture on Human Lung Small Airway Epithelial Cells. BioMed Res. Int. 2020, 2020, 9847579. [Google Scholar] [CrossRef] [PubMed]
  98. Stavely, R.; Nurgali, K. The emerging antioxidant paradigm of mesenchymal stem cell therapy. Stem Cells Transl. Med. 2020, 9, 985–1006. [Google Scholar] [CrossRef]
  99. Oliva, J. Therapeutic Properties of Mesenchymal Stem Cell on Organ Ischemia-Reperfusion Injury. Int. J. Mol. Sci. 2019, 20, 5511. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  100. Pelizzo, G.; Silvestro, S.; Avanzini, M.A.; Zuccotti, G.; Mazzon, E.; Calcaterra, V. Mesenchymal Stromal Cells for the Treatment of Interstitial Lung Disease in Children: A Look from Pediatric and Pediatric Surgeon Viewpoints. Cells 2021, 10, 3270. [Google Scholar] [CrossRef]
  101. Walter, J.; Ware, L.B.; Matthay, M.A. Mesenchymal stem cells: Mechanisms of potential therapeutic benefit in ARDS and sepsis. Lancet. Respir. Med. 2014, 2, 1016–1026. [Google Scholar] [PubMed] [Green Version]
  102. Wick, K.D.; Leligdowicz, A.; Zhuo, H.; Ware, L.B.; Matthay, M.A. Mesenchymal stromal cells reduce evidence of lung injury in patients with ARDS. JCI Insight 2021, 6, e148983. [Google Scholar] [PubMed]
  103. Wilson, J.G.; Liu, K.D.; Zhuo, H.; Caballero, L.; McMillan, M.; Fang, X.; Cosgrove, K.; Vojnik, R.; Calfee, C.S.; Lee, J.W.; et al. Mesenchymal stem (stromal) cells for treatment of ARDS: A phase 1 clinical trial. Lancet. Respir. Med. 2015, 3, 24–32. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Abreu, S.C.; Weiss, D.J.; Rocco, P.R. Extracellular vesicles derived from mesenchymal stromal cells: A therapeutic option in respiratory diseases? Stem Cell Res. Ther. 2016, 7, 53. [Google Scholar] [CrossRef] [Green Version]
  105. Morrison, T.J.; Jackson, M.V.; Cunningham, E.K.; Kissenpfennig, A.; McAuley, D.F.; O’Kane, C.M.; Krasnodembskaya, A.D. Mesenchymal Stromal Cells Modulate Macrophages in Clinically Relevant Lung Injury Models by Extracellular Vesicle Mitochondrial Transfer. Am. J. Respir. Crit. Care Med. 2017, 196, 1275–1286. [Google Scholar] [CrossRef] [Green Version]
  106. Guillamat-Prats, R.; Camprubi-Rimblas, M.; Puig, F.; Herrero, R.; Tantinya, N.; Serrano-Mollar, A.; Artigas, A. Alveolar Type II Cells or Mesenchymal Stem Cells: Comparison of Two Different Cell Therapies for the Treatment of Acute Lung Injury in Rats. Cells 2020, 9, 1816. [Google Scholar] [CrossRef]
  107. Chambers, D.C.; Enever, D.; Lawrence, S.; Sturm, M.J.; Herrmann, R.; Yerkovich, S.; Musk, M.; Hopkins, P.M. Mesenchymal Stromal Cell Therapy for Chronic Lung Allograft Dysfunction: Results of a First-in-Man Study. Stem Cells Transl. Med. 2017, 6, 1152–1157. [Google Scholar] [CrossRef]
  108. Charles, E.J.; Huerter, M.E.; Wagner, C.E.; Sharma, A.K.; Zhao, Y.; Stoler, M.H.; Mehaffey, J.H.; Isbell, J.M.; Lau, C.L.; Tribble, C.G.; et al. Donation After Circulatory Death Lungs Transplantable Up to Six Hours After Ex Vivo Lung Perfusion. Ann. Thorac. Surg. 2016, 102, 1845–1853. [Google Scholar] [CrossRef] [Green Version]
  109. Cypel, M.; Yeung, J.C.; Machuca, T.; Chen, M.; Singer, L.G.; Yasufuku, K.; de Perrot, M.; Pierre, A.; Waddell, T.K.; Keshavjee, S. Experience with the first 50 ex vivo lung perfusions in clinical transplantation. J. Thorac. Cardiovasc. Surg. 2012, 144, 1200–1206. [Google Scholar] [CrossRef] [Green Version]
  110. Cypel, M.; Yeung, J.C.; Liu, M.; Anraku, M.; Chen, F.; Karolak, W.; Sato, M.; Laratta, J.; Azad, S.; Madonik, M.; et al. Normothermic ex vivo lung perfusion in clinical lung transplantation. New Engl. J. Med. 2011, 364, 1431–1440. [Google Scholar] [CrossRef] [Green Version]
  111. Machuca, T.N.; Cypel, M. Ex vivo lung perfusion. J. Thorac. Dis. 2014, 6, 1054–1062. [Google Scholar] [PubMed]
  112. Lee, J.W.; Fang, X.; Gupta, N.; Serikov, V.; Matthay, M.A. Allogeneic human mesenchymal stem cells for treatment of E. coli endotoxin-induced acute lung injury in the ex vivo perfused human lung. Proc. Natl. Acad. Sci. USA 2009, 106, 16357–16362. [Google Scholar] [CrossRef] [Green Version]
  113. Varkouhi, A.K.; Jerkic, M.; Ormesher, L.; Gagnon, S.; Goyal, S.; Rabani, R.; Masterson, C.; Spring, C.; Chen, P.Z.; Gu, F.X.; et al. Extracellular Vesicles from Interferon-gamma-primed Human Umbilical Cord Mesenchymal Stromal Cells Reduce Escherichia coli-induced Acute Lung Injury in Rats. Anesthesiology 2019, 130, 778–790. [Google Scholar] [CrossRef]
  114. Gennai, S.; Monsel, A.; Hao, Q.; Park, J.; Matthay, M.A.; Lee, J.W. Microvesicles Derived From Human Mesenchymal Stem Cells Restore Alveolar Fluid Clearance in Human Lungs Rejected for Transplantation. Am. J. Transplant. Off. J. Am. Soc. Transplant. Am. Soc. Transpl. Surg. 2015, 15, 2404–2412. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Hayes, M.; Curley, G.F.; Masterson, C.; Devaney, J.; O’Toole, D.; Laffey, J.G. Mesenchymal stromal cells are more effective than the MSC secretome in diminishing injury and enhancing recovery following ventilator-induced lung injury. Intensive Care Med. Exp. 2015, 3, 29. [Google Scholar] [CrossRef] [Green Version]
  116. Lu, H.; Poirier, C.; Cook, T.; Traktuev, D.O.; Merfeld-Clauss, S.; Lease, B.; Petrache, I.; March, K.L.; Bogatcheva, N.V. Conditioned media from adipose stromal cells limit lipopolysaccharide-induced lung injury, endothelial hyperpermeability and apoptosis. J. Transl. Med. 2015, 13, 67. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Shologu, N.; Scully, M.; Laffey, J.G.; O’Toole, D. Human Mesenchymal Stem Cell Secretome from Bone Marrow or Adipose-Derived Tissue Sources for Treatment of Hypoxia-Induced Pulmonary Epithelial Injury. Int. J. Mol. Sci. 2018, 19, 2996. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Prudhomme, T.; Kervella, D.; Le Bas-Bernardet, S.; Cantarovich, D.; Karam, G.; Blancho, G.; Branchereau, J. Ex situ Perfusion of Pancreas for Whole-Organ Transplantation: Is it Safe and Feasible? A Systematic Review. J. Diabetes Sci. Technol. 2020, 14, 120–134. [Google Scholar] [CrossRef] [PubMed]
  119. Barton, F.B.; Rickels, M.R.; Alejandro, R.; Hering, B.J.; Wease, S.; Naziruddin, B.; Oberholzer, J.; Odorico, J.S.; Garfinkel, M.R.; Levy, M.; et al. Improvement in outcomes of clinical islet transplantation: 1999–2010. Diabetes Care 2012, 35, 1436–1445. [Google Scholar] [CrossRef] [Green Version]
  120. Hilling, D.E.; Bouwman, E.; Terpstra, O.T.; Marang-van de Mheen, P.J. Effects of donor-, pancreas-, and isolation-related variables on human islet isolation outcome: A systematic review. Cell Transplant. 2014, 23, 921–928. [Google Scholar] [CrossRef] [Green Version]
  121. Kaddis, J.S.; Danobeitia, J.S.; Niland, J.C.; Stiller, T.; Fernandez, L.A. Multicenter analysis of novel and established variables associated with successful human islet isolation outcomes. Am. J. Transplant. Off. J. Am. Soc. Transplant. Am. Soc. Transpl. Surg. 2010, 10, 646–656. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Wang, Y.; Danielson, K.K.; Ropski, A.; Harvat, T.; Barbaro, B.; Paushter, D.; Qi, M.; Oberholzer, J. Systematic analysis of donor and isolation factor’s impact on human islet yield and size distribution. Cell Transplant. 2013, 22, 2323–2333. [Google Scholar] [CrossRef] [Green Version]
  123. Brandhorst, H.; Iken, M.; Scott, W.E., 3rd; Papas, K.K.; Theisinger, B.; Johnson, P.R.; Korsgren, O.; Brandhorst, D. Quality of isolated pig islets is improved using perfluorohexyloctane for pancreas storage in a split lobe model. Cell Transplant. 2013, 22, 1477–1483. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Fridell, J.A.; Mangus, R.S.; Powelson, J.A. Organ preservation solutions for whole organ pancreas transplantation. Curr. Opin. Organ Transplant. 2011, 16, 116–122. [Google Scholar] [CrossRef] [PubMed]
  125. Iwanaga, Y.; Sutherland, D.E.; Harmon, J.V.; Papas, K.K. Pancreas preservation for pancreas and islet transplantation. Curr. Opin. Organ Transplant. 2008, 13, 445–451. [Google Scholar] [CrossRef]
  126. Salehi, P.; Hansen, M.A.; Avila, J.G.; Barbaro, B.; Gangemi, A.; Romagnoli, T.; Wang, Y.; Qi, M.; Murdock, P.; Benedetti, E.; et al. Human islet isolation outcomes from pancreata preserved with Histidine-Tryptophan Ketoglutarate versus University of Wisconsin solution. Transplantation 2006, 82, 983–985. [Google Scholar] [CrossRef] [PubMed]
  127. Hubert, T.; Gmyr, V.; Arnalsteen, L.; Jany, T.; Triponez, F.; Caiazzo, R.; Vandewalle, B.; Vantyghem, M.C.; Kerr-Conte, J.; Pattou, F. Influence of preservation solution on human islet isolation outcome. Transplantation 2007, 83, 270–276. [Google Scholar] [CrossRef]
  128. Paushter, D.H.; Qi, M.; Danielson, K.K.; Harvat, T.A.; Kinzer, K.; Barbaro, B.; Patel, S.; Hassan, S.Z.; Oberholzer, J.; Wang, Y. Histidine-tryptophan-ketoglutarate and University of Wisconsin solution demonstrate equal effectiveness in the preservation of human pancreata intended for islet isolation: A large-scale, single-center experience. Cell Transplant. 2013, 22, 1113–1121. [Google Scholar] [CrossRef] [Green Version]
  129. Pileggi, A.; Ribeiro, M.M.; Hogan, A.R.; Molano, R.D.; Cobianchi, L.; Ichii, H.; Embury, J.; Inverardi, L.; Fornoni, A.; Ricordi, C.; et al. Impact of pancreatic cold preservation on rat islet recovery and function. Transplantation 2009, 87, 1442–1450. [Google Scholar] [CrossRef]
  130. Nishime, K.; Miyagi-Shiohira, C.; Kuwae, K.; Tamaki, Y.; Yonaha, T.; Sakai-Yonaha, M.; Saitoh, I.; Watanabe, M.; Noguchi, H. Preservation of pancreas in the University of Wisconsin solution supplemented with AP39 reduces reactive oxygen species production and improves islet graft function. Am. J. Transplant. Off. J. Am. Soc. Transplant. Am. Soc. Transpl. Surg. 2021, 21, 2698–2708. [Google Scholar] [CrossRef]
  131. Donzelli, E.; Scuteri, A. Mesenchymal Stem Cells: A Trump Card for the Treatment of Diabetes? Biomedicines 2020, 8, 112. [Google Scholar] [CrossRef] [PubMed]
  132. Kasahara, N.; Teratani, T.; Doi, J.; Iijima, Y.; Maeda, M.; Uemoto, S.; Fujimoto, Y.; Sata, N.; Yasuda, Y.; Kobayashi, E. Use of Mesenchymal Stem Cell-Conditioned Medium to Activate Islets in Preservation Solution. Cell Med. 2013, 5, 75–81. [Google Scholar] [CrossRef] [PubMed]
  133. Teratani, T.; Kasahara, N.; Fujimoto, Y.; Sakuma, Y.; Miki, A.; Goto, M.; Sata, N.; Kitayama, J. Mesenchymal Stem Cells Secretions Enhanced ATP Generation on Isolated Islets during Transplantation. Islets 2022, 14, 69–81. [Google Scholar] [CrossRef] [PubMed]
  134. Miao, G.; Ostrowski, R.P.; Mace, J.; Hough, J.; Hopper, A.; Peverini, R.; Chinnock, R.; Zhang, J.; Hathout, E. Dynamic production of hypoxia-inducible factor-1alpha in early transplanted islets. Am. J. Transplant. Off. J. Am. Soc. Transplant. Am. Soc. Transpl. Surg. 2006, 6, 2636–2643. [Google Scholar] [CrossRef]
  135. Figliuzzi, M.; Bonandrini, B.; Silvani, S.; Remuzzi, A. Mesenchymal stem cells help pancreatic islet transplantation to control type 1 diabetes. World J. Stem Cells 2014, 6, 163–172. [Google Scholar] [CrossRef]
  136. Hematti, P.; Kim, J.; Stein, A.P.; Kaufman, D. Potential role of mesenchymal stromal cells in pancreatic islet transplantation. Transplant. Rev. 2013, 27, 21–29. [Google Scholar] [CrossRef]
  137. Ding, Y.; Xu, D.; Feng, G.; Bushell, A.; Muschel, R.J.; Wood, K.J. Mesenchymal stem cells prevent the rejection of fully allogenic islet grafts by the immunosuppressive activity of matrix metalloproteinase-2 and -9. Diabetes 2009, 58, 1797–1806. [Google Scholar] [CrossRef] [Green Version]
  138. Longoni, B.; Szilagyi, E.; Quaranta, P.; Paoli, G.T.; Tripodi, S.; Urbani, S.; Mazzanti, B.; Rossi, B.; Fanci, R.; Demontis, G.C.; et al. Mesenchymal stem cells prevent acute rejection and prolong graft function in pancreatic islet transplantation. Diabetes Technol. Ther. 2010, 12, 435–446. [Google Scholar] [CrossRef]
  139. Li, F.R.; Wang, X.G.; Deng, C.Y.; Qi, H.; Ren, L.L.; Zhou, H.X. Immune modulation of co-transplantation mesenchymal stem cells with islet on T and dendritic cells. Clin. Exp. Immunol. 2010, 161, 357–363. [Google Scholar] [CrossRef]
  140. Berman, D.M.; Willman, M.A.; Han, D.; Kleiner, G.; Kenyon, N.M.; Cabrera, O.; Karl, J.A.; Wiseman, R.W.; O’Connor, D.H.; Bartholomew, A.M.; et al. Mesenchymal stem cells enhance allogeneic islet engraftment in nonhuman primates. Diabetes 2010, 59, 2558–2568. [Google Scholar] [CrossRef] [Green Version]
  141. Chadban, S.J.; Ahn, C.; Axelrod, D.A.; Foster, B.J.; Kasiske, B.L.; Kher, V.; Kumar, D.; Oberbauer, R.; Pascual, J.; Pilmore, H.L.; et al. KDIGO Clinical Practice Guideline on the Evaluation and Management of Candidates for Kidney Transplantation. Transplantation 2020, 104 (Suppl. S1), S11–S103. [Google Scholar] [CrossRef]
  142. Han, S.J.; Lee, H.T. Mechanisms and therapeutic targets of ischemic acute kidney injury. Kidney Res. Clin. Pract. 2019, 38, 427–440. [Google Scholar] [CrossRef] [Green Version]
  143. Zhou, Y.; Xu, H.; Xu, W.; Wang, B.; Wu, H.; Tao, Y.; Zhang, B.; Wang, M.; Mao, F.; Yan, Y.; et al. Exosomes released by human umbilical cord mesenchymal stem cells protect against cisplatin-induced renal oxidative stress and apoptosis in vivo and in vitro. Stem Cell Res. Ther. 2013, 4, 34. [Google Scholar] [CrossRef] [Green Version]
  144. Baulier, E.; Favreau, F.; Le Corf, A.; Jayle, C.; Schneider, F.; Goujon, J.M.; Feraud, O.; Bennaceur-Griscelli, A.; Hauet, T.; Turhan, A.G. Amniotic fluid-derived mesenchymal stem cells prevent fibrosis and preserve renal function in a preclinical porcine model of kidney transplantation. Stem Cells Transl. Med. 2014, 3, 809–820. [Google Scholar] [CrossRef] [PubMed]
  145. Ebrahimi, B.; Eirin, A.; Li, Z.; Zhu, X.Y.; Zhang, X.; Lerman, A.; Textor, S.C.; Lerman, L.O. Mesenchymal stem cells improve medullary inflammation and fibrosis after revascularization of swine atherosclerotic renal artery stenosis. PLoS ONE 2013, 8, e67474. [Google Scholar] [CrossRef] [Green Version]
  146. Eirin, A.; Zhu, X.Y.; Krier, J.D.; Tang, H.; Jordan, K.L.; Grande, J.P.; Lerman, A.; Textor, S.C.; Lerman, L.O. Adipose tissue-derived mesenchymal stem cells improve revascularization outcomes to restore renal function in swine atherosclerotic renal artery stenosis. Stem Cells 2012, 30, 1030–1041. [Google Scholar] [CrossRef] [Green Version]
  147. Morigi, M.; Introna, M.; Imberti, B.; Corna, D.; Abbate, M.; Rota, C.; Rottoli, D.; Benigni, A.; Perico, N.; Zoja, C.; et al. Human bone marrow mesenchymal stem cells accelerate recovery of acute renal injury and prolong survival in mice. Stem Cells 2008, 26, 2075–2082. [Google Scholar] [CrossRef] [PubMed]
  148. Semedo, P.; Palasio, C.G.; Oliveira, C.D.; Feitoza, C.Q.; Goncalves, G.M.; Cenedeze, M.A.; Wang, P.M.; Teixeira, V.P.; Reis, M.A.; Pacheco-Silva, A.; et al. Early modulation of inflammation by mesenchymal stem cell after acute kidney injury. Int. Immunopharmacol. 2009, 9, 677–682. [Google Scholar] [CrossRef]
  149. Mudrabettu, C.; Kumar, V.; Rakha, A.; Yadav, A.K.; Ramachandran, R.; Kanwar, D.B.; Nada, R.; Minz, M.; Sakhuja, V.; Marwaha, N.; et al. Safety and efficacy of autologous mesenchymal stromal cells transplantation in patients undergoing living donor kidney transplantation: A pilot study. Nephrology 2015, 20, 25–33. [Google Scholar] [CrossRef] [PubMed]
  150. Perico, N.; Casiraghi, F.; Gotti, E.; Introna, M.; Todeschini, M.; Cavinato, R.A.; Capelli, C.; Rambaldi, A.; Cassis, P.; Rizzo, P.; et al. Mesenchymal stromal cells and kidney transplantation: Pretransplant infusion protects from graft dysfunction while fostering immunoregulation. Transpl. Int. Off. J. Eur. Soc. Organ Transplant. 2013, 26, 867–878. [Google Scholar] [CrossRef]
  151. Perico, N.; Casiraghi, F.; Introna, M.; Gotti, E.; Todeschini, M.; Cavinato, R.A.; Capelli, C.; Rambaldi, A.; Cassis, P.; Rizzo, P.; et al. Autologous mesenchymal stromal cells and kidney transplantation: A pilot study of safety and clinical feasibility. Clin. J. Am. Soc. Nephrol. 2011, 6, 412–422. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Perico, N.; Casiraghi, F.; Remuzzi, G. Clinical Translation of Mesenchymal Stromal Cell Therapies in Nephrology. J. Am. Soc. Nephrol. 2018, 29, 362–375. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Reinders, M.E.; de Fijter, J.W.; Roelofs, H.; Bajema, I.M.; de Vries, D.K.; Schaapherder, A.F.; Claas, F.H.; van Miert, P.P.; Roelen, D.L.; van Kooten, C.; et al. Autologous bone marrow-derived mesenchymal stromal cells for the treatment of allograft rejection after renal transplantation: Results of a phase I study. Stem Cells Transl. Med. 2013, 2, 107–111. [Google Scholar] [CrossRef] [PubMed]
  154. Kinsey, G.R. Macrophage dynamics in AKI to CKD progression. J. Am. Soc. Nephrol. 2014, 25, 209–211. [Google Scholar] [CrossRef] [Green Version]
  155. Su, X.; Liu, B.; Wang, S.; Wang, Y.; Zhang, Z.; Zhou, H.; Li, F. NLRP3 inflammasome: A potential therapeutic target to minimize renal ischemia/reperfusion injury during transplantation. Transpl. Immunol. 2022, 75, 101718. [Google Scholar] [CrossRef]
  156. Murray, P.J. Macrophage Polarization. Annu. Rev. Physiol. 2017, 79, 541–566. [Google Scholar] [CrossRef]
  157. Meng, X.M.; Tang, P.M.; Li, J.; Lan, H.Y. Macrophage Phenotype in Kidney Injury and Repair. Kidney Dis. 2015, 1, 138–146. [Google Scholar] [CrossRef]
  158. Lee, S.; Huen, S.; Nishio, H.; Nishio, S.; Lee, H.K.; Choi, B.S.; Ruhrberg, C.; Cantley, L.G. Distinct macrophage phenotypes contribute to kidney injury and repair. J. Am. Soc. Nephrol. 2011, 22, 317–326. [Google Scholar] [CrossRef] [Green Version]
  159. Lech, M.; Grobmayr, R.; Ryu, M.; Lorenz, G.; Hartter, I.; Mulay, S.R.; Susanti, H.E.; Kobayashi, K.S.; Flavell, R.A.; Anders, H.J. Macrophage phenotype controls long-term AKI outcomes--kidney regeneration versus atrophy. J. Am. Soc. Nephrol. 2014, 25, 292–304. [Google Scholar] [CrossRef] [Green Version]
  160. Tammaro, A.; Kers, J.; Scantlebery, A.M.L.; Florquin, S. Metabolic Flexibility and Innate Immunity in Renal Ischemia Reperfusion Injury: The Fine Balance Between Adaptive Repair and Tissue Degeneration. Front. Immunol. 2020, 11, 1346. [Google Scholar] [CrossRef]
  161. De Witte, S.F.H.; Luk, F.; Sierra Parraga, J.M.; Gargesha, M.; Merino, A.; Korevaar, S.S.; Shankar, A.S.; O’Flynn, L.; Elliman, S.J.; Roy, D.; et al. Immunomodulation By Therapeutic Mesenchymal Stromal Cells (MSC) Is Triggered Through Phagocytosis of MSC By Monocytic Cells. Stem Cells 2018, 36, 602–615. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Eggenhofer, E.; Benseler, V.; Kroemer, A.; Popp, F.C.; Geissler, E.K.; Schlitt, H.J.; Baan, C.C.; Dahlke, M.H.; Hoogduijn, M.J. Mesenchymal stem cells are short-lived and do not migrate beyond the lungs after intravenous infusion. Front. Immunol. 2012, 3, 297. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Thompson, M.; Mei, S.H.J.; Wolfe, D.; Champagne, J.; Fergusson, D.; Stewart, D.J.; Sullivan, K.J.; Doxtator, E.; Lalu, M.; English, S.W.; et al. Cell therapy with intravascular administration of mesenchymal stromal cells continues to appear safe: An updated systematic review and meta-analysis. EClinicalMedicine 2020, 19, 100249. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Hamar, M.; Urbanellis, P.; Kaths, M.J.; Kollmann, D.; Linares, I.; Ganesh, S.; Wiebe, A.; Cen, J.Y.; Yip, P.; John, R.; et al. Normothermic Ex Vivo Kidney Perfusion Reduces Warm Ischemic Injury of Porcine Kidney Grafts Retrieved After Circulatory Death. Transplantation 2018, 102, 1262–1270. [Google Scholar] [CrossRef] [PubMed]
  165. Hosgood, S.A.; Saeb-Parsy, K.; Hamed, M.O.; Nicholson, M.L. Successful Transplantation of Human Kidneys Deemed Untransplantable but Resuscitated by Ex Vivo Normothermic Machine Perfusion. Am. J. Transplant. Off. J. Am. Soc. Transplant. Am. Soc. Transpl. Surg. 2016, 16, 3282–3285. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Weissenbacher, A.; Lo Faro, L.; Boubriak, O.; Soares, M.F.; Roberts, I.S.; Hunter, J.P.; Voyce, D.; Mikov, N.; Cook, A.; Ploeg, R.J.; et al. Twenty-four-hour normothermic perfusion of discarded human kidneys with urine recirculation. Am. J. Transplant. Off. J. Am. Soc. Transplant. Am. Soc. Transpl. Surg. 2019, 19, 178–192. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  167. Hayase, N.; Doi, K.; Hiruma, T.; Matsuura, R.; Hamasaki, Y.; Noiri, E.; Nangaku, M.; Morimura, N. Recombinant Thrombomodulin on Neutrophil Extracellular Traps in Murine Intestinal Ischemia-Reperfusion. Anesthesiology 2019, 131, 866–882. [Google Scholar] [CrossRef]
  168. Gutierrez-Sanchez, G.; Garcia-Alonso, I.; Gutierrez Saenz de Santa Maria, J.; Alonso-Varona, A.; Herrero de la Parte, B. Antioxidant-Based Therapy Reduces Early-Stage Intestinal Ischemia-Reperfusion Injury in Rats. Antioxidants 2021, 10, 853. [Google Scholar] [CrossRef]
  169. Ma, Y.; Zabell, T.; Creasy, A.; Yang, X.; Chatterjee, V.; Villalba, N.; Kistler, E.B.; Wu, M.H.; Yuan, S.Y. Gut Ischemia Reperfusion Injury Induces Lung Inflammation via Mesenteric Lymph-Mediated Neutrophil Activation. Front. Immunol. 2020, 11, 586685. [Google Scholar] [CrossRef]
  170. Wu, M.; Rowe, J.M.; Fleming, S.D. Complement Initiation Varies by Sex in Intestinal Ischemia Reperfusion Injury. Front. Immunol. 2021, 12, 649882. [Google Scholar] [CrossRef]
  171. Morris, N.L.; Cannon, A.R.; Li, X.; Choudhry, M.A. Protective effects of PX478 on gut barrier in a mouse model of ethanol and burn injury. J. Leukoc. Biol. 2021, 109, 1121–1130. [Google Scholar] [CrossRef] [PubMed]
  172. Swank, G.M.; Deitch, E.A. Role of the gut in multiple organ failure: Bacterial translocation and permeability changes. World J. Surg. 1996, 20, 411–417. [Google Scholar] [CrossRef] [PubMed]
  173. Doster, D.L.; Jensen, A.R.; Khaneki, S.; Markel, T.A. Mesenchymal stromal cell therapy for the treatment of intestinal ischemia: Defining the optimal cell isolate for maximum therapeutic benefit. Cytotherapy 2016, 18, 1457–1470. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Souza, D.G.; Amaral, F.A.; Fagundes, C.T.; Coelho, F.M.; Arantes, R.M.; Sousa, L.P.; Matzuk, M.M.; Garlanda, C.; Mantovani, A.; Dias, A.A.; et al. The long pentraxin PTX3 is crucial for tissue inflammation after intestinal ischemia and reperfusion in mice. Am. J. Pathol. 2009, 174, 1309–1318. [Google Scholar] [CrossRef] [Green Version]
  175. Clair, D.G.; Beach, J.M. Mesenteric Ischemia. New Engl. J. Med. 2016, 374, 959–968. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Clark, J.A.; Coopersmith, C.M. Intestinal crosstalk: A new paradigm for understanding the gut as the “motor” of critical illness. Shock 2007, 28, 384–393. [Google Scholar] [CrossRef] [Green Version]
  177. Nadatani, Y.; Watanabe, T.; Shimada, S.; Otani, K.; Tanigawa, T.; Fujiwara, Y. Microbiome and intestinal ischemia/reperfusion injury. J. Clin. Biochem. Nutr. 2018, 63, 26–32. [Google Scholar] [CrossRef] [Green Version]
  178. Oldenburg, W.A.; Lau, L.L.; Rodenberg, T.J.; Edmonds, H.J.; Burger, C.D. Acute mesenteric ischemia: A clinical review. Arch. Intern. Med. 2004, 164, 1054–1062. [Google Scholar] [CrossRef]
  179. 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]
  180. Brittan, M.; Hunt, T.; Jeffery, R.; Poulsom, R.; Forbes, S.J.; Hodivala-Dilke, K.; Goldman, J.; Alison, M.R.; Wright, N.A. Bone marrow derivation of pericryptal myofibroblasts in the mouse and human small intestine and colon. Gut 2002, 50, 752–757. [Google Scholar] [CrossRef]
  181. Mifflin, R.C.; Pinchuk, I.V.; Saada, J.I.; Powell, D.W. Intestinal myofibroblasts: Targets for stem cell therapy. Am. J. Physiol. Gastrointest. Liver Physiol. 2011, 300, G684–G696. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  182. Mishra, P.J.; Mishra, P.J.; Banerjee, D. Cell-free derivatives from mesenchymal stem cells are effective in wound therapy. World J. Stem Cells 2012, 4, 35–43. [Google Scholar] [CrossRef] [PubMed]
  183. Weil, B.R.; Markel, T.A.; Herrmann, J.L.; Abarbanell, A.M.; Meldrum, D.R. Mesenchymal stem cells enhance the viability and proliferation of human fetal intestinal epithelial cells following hypoxic injury via paracrine mechanisms. Surgery 2009, 146, 190–197. [Google Scholar] [CrossRef] [PubMed]
  184. Nichols, T.C. NF-kappaB and reperfusion injury. Drug News Perspect. 2004, 17, 99–104. [Google Scholar] [CrossRef] [PubMed]
  185. Grootjans, J.; Lenaerts, K.; Derikx, J.P.; Matthijsen, R.A.; de Bruine, A.P.; van Bijnen, A.A.; van Dam, R.M.; Dejong, C.H.; Buurman, W.A. Human intestinal ischemia-reperfusion-induced inflammation characterized: Experiences from a new translational model. Am. J. Pathol. 2010, 176, 2283–2291. [Google Scholar] [CrossRef] [PubMed]
  186. O’Neill, L.A. The interleukin-1 receptor/Toll-like receptor superfamily: Signal transduction during inflammation and host defense. Sci. STKE Signal Transduct. Knowl. Environ. 2000, 2000, re1. [Google Scholar]
  187. Alexandropoulos, D.; Bazigos, G.V.; Doulamis, I.P.; Tzani, A.; Konstantopoulos, P.; Tragotsalou, N.; Kondi-Pafiti, A.; Kotsis, T.; Arkadopoulos, N.; Smyrniotis, V.; et al. Protective effects of N-acetylcystein and atorvastatin against renal and hepatic injury in a rat model of intestinal ischemia-reperfusion. Biomed. Pharmacother. = Biomed. Pharmacother. 2017, 89, 673–680. [Google Scholar] [CrossRef]
  188. Edgerton, C.; Crispin, J.C.; Moratz, C.M.; Bettelli, E.; Oukka, M.; Simovic, M.; Zacharia, A.; Egan, R.; Chen, J.; Dalle Lucca, J.J.; et al. IL-17 producing CD4+ T cells mediate accelerated ischemia/reperfusion-induced injury in autoimmunity-prone mice. Clin. Immunol. 2009, 130, 313–321. [Google Scholar] [CrossRef] [Green Version]
  189. 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]
  190. Maggini, J.; Mirkin, G.; Bognanni, I.; Holmberg, J.; Piazzon, I.M.; Nepomnaschy, I.; Costa, H.; Canones, C.; Raiden, S.; Vermeulen, M.; et al. Mouse bone marrow-derived mesenchymal stromal cells turn activated macrophages into a regulatory-like profile. PLoS ONE 2010, 5, e9252. [Google Scholar] [CrossRef]
  191. Nemeth, K.; Leelahavanichkul, A.; Yuen, P.S.; Mayer, B.; Parmelee, A.; Doi, K.; Robey, P.G.; Leelahavanichkul, K.; Koller, B.H.; Brown, J.M.; et al. Bone marrow stromal cells attenuate sepsis via prostaglandin E(2)-dependent reprogramming of host macrophages to increase their interleukin-10 production. Nat. Med. 2009, 15, 42–49. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  192. Barzegar, M.; Kaur, G.; Gavins, F.N.E.; Wang, Y.; Boyer, C.J.; Alexander, J.S. Potential therapeutic roles of stem cells in ischemia-reperfusion injury. Stem Cell Res. 2019, 37, 101421. [Google Scholar] [CrossRef] [PubMed]
  193. Kinnaird, T.; Stabile, E.; Burnett, M.S.; Lee, C.W.; Barr, S.; Fuchs, S.; Epstein, S.E. Marrow-derived stromal cells express genes encoding a broad spectrum of arteriogenic cytokines and promote in vitro and in vivo arteriogenesis through paracrine mechanisms. Circ. Res. 2004, 94, 678–685. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. Wang, M.; Crisostomo, P.R.; Herring, C.; Meldrum, K.K.; Meldrum, D.R. Human progenitor cells from bone marrow or adipose tissue produce VEGF, HGF, and IGF-I in response to TNF by a p38 MAPK-dependent mechanism. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2006, 291, R880–R884. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  195. Li, Y.Y.; Xu, Q.W.; Xu, P.Y.; Li, W.M. MSC-derived exosomal miR-34a/c-5p and miR-29b-3p improve intestinal barrier function by targeting the Snail/Claudins signaling pathway. Life Sci. 2020, 257, 118017. [Google Scholar] [CrossRef] [PubMed]
  196. Barnard, C.N. The operation. A human cardiac transplant: An interim report of a successful operation performed at Groote Schuur Hospital, Cape Town. South Afr. Med. J. = Suid-Afrik. Tydskr. Vir Geneeskd. 1967, 41, 1271–1274. [Google Scholar]
  197. Salahudeen, A.K.; Huang, H.; Patel, P.; Jenkins, J.K. Mechanism and prevention of cold storage-induced human renal tubular cell injury. Transplantation 2000, 70, 1424–1431. [Google Scholar] [CrossRef] [Green Version]
  198. Enerson, D.M.; Merola, J. Cellular swelling: 3. Effects of hypothermia and addition of low molecular weight dextran on oxygen consumption and electrolyte composition of isolated tissues. Ann. Surg. 1967, 165, 244–249. [Google Scholar] [CrossRef]
  199. Bonventre, J.V.; Cheung, J.Y. Effects of metabolic acidosis on viability of cells exposed to anoxia. Am. J. Physiol. 1985, 249, C149–C159. [Google Scholar] [CrossRef]
  200. Hess, N.R.; Ziegler, L.A.; Kaczorowski, D.J. Heart Donation and Preservation: Historical Perspectives, Current Technologies, and Future Directions. J. Clin. Med. 2022, 11, 5762. [Google Scholar] [CrossRef]
  201. Zhang, L.L.; Xiong, Y.Y.; Yang, Y.J. The Vital Roles of Mesenchymal Stem Cells and the Derived Extracellular Vesicles in Promoting Angiogenesis After Acute Myocardial Infarction. Stem Cells Dev. 2021, 30, 561–577. [Google Scholar] [CrossRef] [PubMed]
  202. Pittenger, M.F.; Mackay, A.M.; Beck, S.C.; Jaiswal, R.K.; Douglas, R.; Mosca, J.D.; Moorman, M.A.; Simonetti, D.W.; Craig, S.; Marshak, D.R. Multilineage potential of adult human mesenchymal stem cells. Science 1999, 284, 143–147. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  203. Zhao, P.; Ise, H.; Hongo, M.; Ota, M.; Konishi, I.; Nikaido, T. Human amniotic mesenchymal cells have some characteristics of cardiomyocytes. Transplantation 2005, 79, 528–535. [Google Scholar] [CrossRef] [PubMed]
  204. Alvarez-Dolado, M.; Pardal, R.; Garcia-Verdugo, J.M.; Fike, J.R.; Lee, H.O.; Pfeffer, K.; Lois, C.; Morrison, S.J.; Alvarez-Buylla, A. Fusion of bone-marrow-derived cells with Purkinje neurons, cardiomyocytes and hepatocytes. Nature 2003, 425, 968–973. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  205. Rota, M.; Kajstura, J.; Hosoda, T.; Bearzi, C.; Vitale, S.; Esposito, G.; Iaffaldano, G.; Padin-Iruegas, M.E.; Gonzalez, A.; Rizzi, R.; et al. Bone marrow cells adopt the cardiomyogenic fate in vivo. Proc. Natl. Acad. Sci. USA 2007, 104, 17783–17788. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  206. Nikfarjam, S.; Rezaie, J.; Zolbanin, N.M.; Jafari, R. Mesenchymal stem cell derived-exosomes: A modern approach in translational medicine. J. Transl. Med. 2020, 18, 449. [Google Scholar] [CrossRef]
  207. Chinnici, C.M.; Iannolo, G.; Cittadini, E.; Carreca, A.P.; Nascari, D.; Timoneri, F.; Bella, M.D.; Cuscino, N.; Amico, G.; Carcione, C.; et al. Extracellular Vesicle-Derived microRNAs of Human Wharton’s Jelly Mesenchymal Stromal Cells May Activate Endogenous VEGF-A to Promote Angiogenesis. Int. J. Mol. Sci. 2021, 22, 2045. [Google Scholar] [CrossRef]
  208. Iannolo, G.; Sciuto, M.R.; Cuscino, N.; Carcione, C.; Coronnello, C.; Chinnici, C.M.; Raffa, G.M.; Pilato, M.; Conaldi, P.G. miRNA expression analysis in the human heart: Undifferentiated progenitors vs. bioptic tissues-Implications for proliferation and ageing. J. Cell. Mol. Med. 2021, 25, 8687–8700. [Google Scholar] [CrossRef]
  209. Iannolo, G.; Sciuto, M.R.; Raffa, G.M.; Pilato, M.; Conaldi, P.G. MiR34 inhibition induces human heart progenitor proliferation. Cell Death Dis. 2018, 9, 368. [Google Scholar] [CrossRef] [Green Version]
  210. Kim, H.S.; Choi, D.Y.; Yun, S.J.; Choi, S.M.; Kang, J.W.; Jung, J.W.; Hwang, D.; Kim, K.P.; Kim, D.W. Proteomic analysis of microvesicles derived from human mesenchymal stem cells. J. Proteome Res. 2012, 11, 839–849. [Google Scholar] [CrossRef]
  211. Park, K.M.; Teoh, J.P.; Wang, Y.; Broskova, Z.; Bayoumi, A.S.; Tang, Y.; Su, H.; Weintraub, N.L.; Kim, I.M. Carvedilol-responsive microRNAs, miR-199a-3p and -214 protect cardiomyocytes from simulated ischemia-reperfusion injury. Am. J. Physiol. Heart Circ. Physiol. 2016, 311, H371–H383. [Google Scholar] [CrossRef] [Green Version]
  212. Badami, E.; Busa, R.; Douradinha, B.; Russelli, G.; Miceli, V.; Gallo, A.; Zito, G.; Conaldi, P.G.; Iannolo, G. Hepatocellular carcinoma, hepatitis C virus infection and miRNA involvement: Perspectives for new therapeutic approaches. World J. Gastroenterol. 2022, 28, 2417–2428. [Google Scholar] [CrossRef]
  213. Lin, L.; Wang, X.; Yu, Z. Ischemia-reperfusion Injury in the Brain: Mechanisms and Potential Therapeutic Strategies. Biochem. Pharmacol. Open Access 2016, 5, 213. [Google Scholar]
  214. Shin, T.H.; Lee, D.Y.; Basith, S.; Manavalan, B.; Paik, M.J.; Rybinnik, I.; Mouradian, M.M.; Ahn, J.H.; Lee, G. Metabolome Changes in Cerebral Ischemia. Cells 2020, 9, 1630. [Google Scholar] [CrossRef]
  215. Miceli, V.; Russelli, G.; Iannolo, G.; Gallo, A.; Lo Re, V.; Agnese, V.; Sparacia, G.; Conaldi, P.G.; Bulati, M. Role of non-coding RNAs in age-related vascular cognitive impairment: An overview on diagnostic/prognostic value in Vascular Dementia and Vascular Parkinsonism. Mech. Ageing Dev. 2020, 191, 111332. [Google Scholar] [CrossRef] [PubMed]
  216. Wang, Y.; Xiao, G.; He, S.; Liu, X.; Zhu, L.; Yang, X.; Zhang, Y.; Orgah, J.; Feng, Y.; Wang, X.; et al. Protection against acute cerebral ischemia/reperfusion injury by QiShenYiQi via neuroinflammatory network mobilization. Biomed. Pharmacother. = Biomed. Pharmacother. 2020, 125, 109945. [Google Scholar] [CrossRef] [PubMed]
  217. Virani, S.S.; Alonso, A.; Benjamin, E.J.; Bittencourt, M.S.; Callaway, C.W.; Carson, A.P.; Chamberlain, A.M.; Chang, A.R.; Cheng, S.; Delling, F.N.; et al. Heart Disease and Stroke Statistics-2020 Update: A Report From the American Heart Association. Circulation 2020, 141, e139–e596. [Google Scholar] [PubMed]
  218. Liu, N.; Chen, R.; Du, H.; Wang, J.; Zhang, Y.; Wen, J. Expression of IL-10 and TNF-alpha in rats with cerebral infarction after transplantation with mesenchymal stem cells. Cell. Mol. Immunol. 2009, 6, 207–213. [Google Scholar] [CrossRef] [Green Version]
  219. Uccelli, A.; Moretta, L.; Pistoia, V. Mesenchymal stem cells in health and disease. Nat. Rev. Immunol. 2008, 8, 726–736. [Google Scholar] [CrossRef]
  220. Aihara, N.; Mizukawa, K.; Koide, K.; Mabe, H.; Nishino, H. Striatal grafts in infarct striatopallidum increase GABA release, reorganize GABAA receptor and improve water-maze learning in the rat. Brain Res. Bull. 1994, 33, 483–488. [Google Scholar] [CrossRef]
  221. Nishino, H.; Koide, K.; Aihara, N.; Kumazaki, M.; Sakurai, T.; Nagai, H. Striatal grafts in the ischemic striatum improve pallidal GABA release and passive avoidance. Brain Res. Bull. 1993, 32, 517–520. [Google Scholar] [CrossRef] [PubMed]
  222. Turkovsky, E.A.; Golovicheva, V.V.; Varlamova, E.G.; Danilina, T.I.; Goryunov, K.V.; Shevstova, Y.A.; Pevzner, I.B.; Zorova, L.D.; Babenko, V.A.; Evtushenko, E.A.; et al. Mesenchymal stromal cell-derived extracellular vesicles afford neuroprotection by modulating PI3K/AKT pathway and calcium oscillations. Int. J. Biol. Sci. 2022, 18, 5345–5368. [Google Scholar] [CrossRef] [PubMed]
  223. Qiao, L.Y.; Huang, F.J.; Zhao, M.; Xie, J.H.; Shi, J.; Wang, J.; Lin, X.Z.; Zuo, H.; Wang, Y.L.; Geng, T.C. A two-year follow-up study of cotransplantation with neural stem/progenitor cells and mesenchymal stromal cells in ischemic stroke patients. Cell Transplant. 2014, 23 (Suppl. S1), S65–S72. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  224. Jiang, Y.; Zhu, W.; Zhu, J.; Wu, L.; Xu, G.; Liu, X. Feasibility of delivering mesenchymal stem cells via catheter to the proximal end of the lesion artery in patients with stroke in the territory of the middle cerebral artery. Cell Transplant. 2013, 22, 2291–2298. [Google Scholar] [CrossRef] [PubMed]
  225. Honmou, O.; Houkin, K.; Matsunaga, T.; Niitsu, Y.; Ishiai, S.; Onodera, R.; Waxman, S.G.; Kocsis, J.D. Intravenous administration of auto serum-expanded autologous mesenchymal stem cells in stroke. Brain A J. Neurol. 2011, 134, 1790–1807. [Google Scholar] [CrossRef] [Green Version]
  226. Kawabori, M.; Shichinohe, H.; Kuroda, S.; Houkin, K. Clinical Trials of Stem Cell Therapy for Cerebral Ischemic Stroke. Int. J. Mol. Sci. 2020, 21, 7380. [Google Scholar] [CrossRef]
  227. Casillas-Ramirez, A.; Mosbah, I.B.; Ramalho, F.; Rosello-Catafau, J.; Peralta, C. Past and future approaches to ischemia-reperfusion lesion associated with liver transplantation. Life Sci. 2006, 79, 1881–1894. [Google Scholar] [CrossRef]
  228. Motino, O.; Frances, D.E.; Casanova, N.; Fuertes-Agudo, M.; Cucarella, C.; Flores, J.M.; Vallejo-Cremades, M.T.; Olmedilla, L.; Perez Pena, J.; Banares, R.; et al. Protective Role of Hepatocyte Cyclooxygenase-2 Expression Against Liver Ischemia-Reperfusion Injury in Mice. Hepatology 2019, 70, 650–665. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  229. Zhai, Y.; Petrowsky, H.; Hong, J.C.; Busuttil, R.W.; Kupiec-Weglinski, J.W. Ischaemia-reperfusion injury in liver transplantation--from bench to bedside. Nat. Rev. Gastroenterol. Hepatol. 2013, 10, 79–89. [Google Scholar] [CrossRef]
  230. Alqahtani, S.A. Update in liver transplantation. Discov. Med. 2012, 14, 133–141. [Google Scholar] [CrossRef]
  231. Sagaradze, G.; Basalova, N.; Kirpatovsky, V.; Ohobotov, D.; Nimiritsky, P.; Grigorieva, O.; Popov, V.; Kamalov, A.; Tkachuk, V.; Efimenko, A. A magic kick for regeneration: Role of mesenchymal stromal cell secretome in spermatogonial stem cell niche recovery. Stem Cell Res. Ther. 2019, 10, 342. [Google Scholar] [CrossRef] [Green Version]
  232. Chinnici, C.M.; Russelli, G.; Bulati, M.; Miceli, V.; Gallo, A.; Busa, R.; Tinnirello, R.; Conaldi, P.G.; Iannolo, G. Mesenchymal stromal cell secretome in liver failure: Perspectives on COVID-19 infection treatment. World J. Gastroenterol. 2021, 27, 1905–1919. [Google Scholar] [CrossRef] [PubMed]
  233. Lee, S.C.; Jeong, H.J.; Lee, S.K.; Kim, S.J. Hypoxic Conditioned Medium From Human Adipose-Derived Stem Cells Promotes Mouse Liver Regeneration Through JAK/STAT3 Signaling. Stem Cells Transl. Med. 2016, 5, 816–825. [Google Scholar] [CrossRef] [PubMed]
  234. Pan, G.Z.; Yang, Y.; Zhang, J.; Liu, W.; Wang, G.Y.; Zhang, Y.C.; Yang, Q.; Zhai, F.X.; Tai, Y.; Liu, J.R.; et al. Bone marrow mesenchymal stem cells ameliorate hepatic ischemia/reperfusion injuries via inactivation of the MEK/ERK signaling pathway in rats. J. Surg. Res. 2012, 178, 935–948. [Google Scholar] [CrossRef]
  235. Seki, T.; Yokoyama, Y.; Nagasaki, H.; Kokuryo, T.; Nagino, M. Adipose tissue-derived mesenchymal stem cell transplantation promotes hepatic regeneration after hepatic ischemia-reperfusion and subsequent hepatectomy in rats. J. Surg. Res. 2012, 178, 63–70. [Google Scholar] [CrossRef]
  236. Chen, K.; Obara, H.; Matsubara, Y.; Fukuda, K.; Yagi, H.; Ono-Uruga, Y.; Matsubara, K.; Kitagawa, Y. Adipose-Derived Mesenchymal Stromal/Stem Cell Line Prevents Hepatic Ischemia/Reperfusion Injury in Rats by Inhibiting Inflammasome Activation. Cell Transplant. 2022, 31, 9636897221089629. [Google Scholar] [CrossRef]
  237. Li, C.; Jin, Y.; Wei, S.; Sun, Y.; Jiang, L.; Zhu, Q.; Farmer, D.G.; Busuttil, R.W.; Kupiec-Weglinski, J.W.; Ke, B. Hippo Signaling Controls NLR Family Pyrin Domain Containing 3 Activation and Governs Immunoregulation of Mesenchymal Stem Cells in Mouse Liver Injury. Hepatology 2019, 70, 1714–1731. [Google Scholar] [CrossRef] [PubMed]
  238. Ma, Y.; Jiao, Z.; Liu, X.; Zhang, Q.; Piao, C.; Xu, J.; Wang, H. Protective effect of adipose-derived stromal cell-secretome attenuate autophagy induced by liver ischemia-reperfusion and partial hepatectomy. Stem Cell Res. Ther. 2022, 13, 427. [Google Scholar] [CrossRef]
  239. Wang, X.; Wang, S.; Zhou, Y.; Obulkasim, H.; Zhang, Z.H.; Dai, B.; Zhu, W.; Shi, X.L. BM-MSCs protect against liver ischemia/reperfusion injury via HO-1 mediated autophagy. Mol. Med. Rep. 2018, 18, 2253–2262. [Google Scholar] [CrossRef]
  240. Zheng, J.; Chen, L.; Lu, T.; Zhang, Y.; Sui, X.; Li, Y.; Huang, X.; He, L.; Cai, J.; Zhou, C.; et al. MSCs ameliorate hepatocellular apoptosis mediated by PINK1-dependent mitophagy in liver ischemia/reperfusion injury through AMPKalpha activation. Cell Death Dis. 2020, 11, 256. [Google Scholar] [CrossRef] [Green Version]
  241. Jiao, Z.; Liu, X.; Ma, Y.; Ge, Y.; Zhang, Q.; Liu, B.; Wang, H. Adipose-Derived Stem Cells Protect Ischemia-Reperfusion and Partial Hepatectomy by Attenuating Endoplasmic Reticulum Stress. Front. Cell Dev. Biol. 2020, 8, 177. [Google Scholar] [CrossRef]
  242. Baglio, S.R.; Pegtel, D.M.; Baldini, N. Mesenchymal stem cell secreted vesicles provide novel opportunities in (stem) cell-free therapy. Front. Physiol. 2012, 3, 359. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  243. Haga, H.; Yan, I.K.; Borrelli, D.A.; Matsuda, A.; Parasramka, M.; Shukla, N.; Lee, D.D.; Patel, T. Extracellular vesicles from bone marrow-derived mesenchymal stem cells protect against murine hepatic ischemia/reperfusion injury. Liver Transplant. Off. Publ. Am. Assoc. Study Liver Dis. Int. Liver Transplant. Soc. 2017, 23, 791–803. [Google Scholar] [CrossRef] [Green Version]
  244. Yao, J.; Zheng, J.; Cai, J.; Zeng, K.; Zhou, C.; Zhang, J.; Li, S.; Li, H.; Chen, L.; He, L.; et al. Extracellular vesicles derived from human umbilical cord mesenchymal stem cells alleviate rat hepatic ischemia-reperfusion injury by suppressing oxidative stress and neutrophil inflammatory response. FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol. 2019, 33, 1695–1710. [Google Scholar] [CrossRef] [Green Version]
  245. Zheng, J.; Lu, T.; Zhou, C.; Cai, J.; Zhang, X.; Liang, J.; Sui, X.; Chen, X.; Chen, L.; Sun, Y.; et al. Extracellular Vesicles Derived from Human Umbilical Cord Mesenchymal Stem Cells Protect Liver Ischemia/Reperfusion Injury by Reducing CD154 Expression on CD4+ T Cells via CCT2. Adv. Sci. 2020, 7, 1903746. [Google Scholar] [CrossRef] [PubMed]
  246. Sun, Y.; Wang, Y.; Zhou, L.; Zou, Y.; Huang, G.; Gao, G.; Ting, S.; Lei, X.; Ding, X. Spheroid-cultured human umbilical cord-derived mesenchymal stem cells attenuate hepatic ischemia-reperfusion injury in rats. Sci. Rep. 2018, 8, 2518. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  247. Rackham, C.L.; Amisten, S.; Persaud, S.J.; King, A.J.F.; Jones, P.M. Mesenchymal stromal cell secretory factors induce sustained improvements in islet function pre- and post-transplantation. Cytotherapy 2018, 20, 1427–1436. [Google Scholar] [CrossRef] [Green Version]
  248. 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]
  249. Saat, T.C.; van den Engel, S.; Bijman-Lachger, W.; Korevaar, S.S.; Hoogduijn, M.J.; JN, I.J.; de Bruin, R.W. Fate and Effect of Intravenously Infused Mesenchymal Stem Cells in a Mouse Model of Hepatic Ischemia Reperfusion Injury and Resection. Stem Cells Int. 2016, 2016, 5761487. [Google Scholar] [CrossRef]
  250. 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]
  251. Baranovskii, D.S.; Klabukov, I.D.; Arguchinskaya, N.V.; Yakimova, A.O.; Kisel, A.A.; Yatsenko, E.M.; Ivanov, S.A.; Shegay, P.V.; Kaprin, A.D. Adverse events, side effects and complications in mesenchymal stromal cell-based therapies. Stem Cell Investig. 2022, 9, 7. [Google Scholar] [CrossRef]
  252. Cunningham, C.J.; Redondo-Castro, E.; Allan, S.M. The therapeutic potential of the mesenchymal stem cell secretome in ischaemic stroke. J. Cereb. Blood Flow Metab. Off. J. Int. Soc. Cereb. Blood Flow Metab. 2018, 38, 1276–1292. [Google Scholar] [CrossRef] [Green Version]
  253. Jiao, Z.; Ma, Y.; Zhang, Q.; Wang, Y.; Liu, T.; Liu, X.; Piao, C.; Liu, B.; Wang, H. The adipose-derived mesenchymal stem cell secretome promotes hepatic regeneration in miniature pigs after liver ischaemia-reperfusion combined with partial resection. Stem Cell Res. Ther. 2021, 12, 218. [Google Scholar] [CrossRef] [PubMed]
  254. Lai, R.C.; Arslan, F.; Lee, M.M.; Sze, N.S.; Choo, A.; Chen, T.S.; Salto-Tellez, M.; Timmers, L.; Lee, C.N.; El Oakley, R.M.; et al. Exosome secreted by MSC reduces myocardial ischemia/reperfusion injury. Stem Cell Res. 2010, 4, 214–222. [Google Scholar] [CrossRef] [Green Version]
  255. Antebi, B.; Rodriguez, L.A., 2nd; Walker, K.P., 3rd; Asher, A.M.; Kamucheka, R.M.; Alvarado, L.; Mohammadipoor, A.; Cancio, L.C. Short-term physiological hypoxia potentiates the therapeutic function of mesenchymal stem cells. Stem Cell Res. Ther. 2018, 9, 265. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  256. Chang, W.; Lee, C.Y.; Park, J.H.; Park, M.S.; Maeng, L.S.; Yoon, C.S.; Lee, M.Y.; Hwang, K.C.; Chung, Y.A. Survival of hypoxic human mesenchymal stem cells is enhanced by a positive feedback loop involving miR-210 and hypoxia-inducible factor 1. J. Vet. Sci. 2013, 14, 69–76. [Google Scholar] [CrossRef] [Green Version]
  257. Fischer, U.M.; Harting, M.T.; Jimenez, F.; Monzon-Posadas, W.O.; Xue, H.; Savitz, S.I.; Laine, G.A.; Cox, C.S., Jr. Pulmonary passage is a major obstacle for intravenous stem cell delivery: The pulmonary first-pass effect. Stem Cells Dev. 2009, 18, 683–692. [Google Scholar] [CrossRef] [PubMed]
  258. Ma, T.; Wang, X.; Jiao, Y.; Wang, H.; Qi, Y.; Gong, H.; Zhang, L.; Jiang, D. Interleukin 17 (IL-17)-Induced Mesenchymal Stem Cells Prolong the Survival of Allogeneic Skin Grafts. Ann. Transplant. 2018, 23, 615–621. [Google Scholar] [CrossRef] [PubMed]
  259. Bhang, S.H.; Lee, S.; Shin, J.Y.; Lee, T.J.; Kim, B.S. Transplantation of cord blood mesenchymal stem cells as spheroids enhances vascularization. Tissue Eng. Part A 2012, 18, 2138–2147. [Google Scholar] [CrossRef]
  260. Han, Y.S.; Lee, J.H.; Yoon, Y.M.; Yun, C.W.; Noh, H.; Lee, S.H. Hypoxia-induced expression of cellular prion protein improves the therapeutic potential of mesenchymal stem cells. Cell Death Dis. 2016, 7, e2395. [Google Scholar] [CrossRef]
  261. Lee, J.H.; Han, Y.S.; Lee, S.H. Long-Duration Three-Dimensional Spheroid Culture Promotes Angiogenic Activities of Adipose-Derived Mesenchymal Stem Cells. Biomol. Ther. 2016, 24, 260–267. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Schematic representation of MSC effects on IRI, with the main biological processes involved in the damage that are attenuated with MSC therapy.
Figure 1. Schematic representation of MSC effects on IRI, with the main biological processes involved in the damage that are attenuated with MSC therapy.
Biomedicines 11 00689 g001
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Miceli, V.; Bulati, M.; Gallo, A.; Iannolo, G.; Busà, R.; Conaldi, P.G.; Zito, G. Role of Mesenchymal Stem/Stromal Cells in Modulating Ischemia/Reperfusion Injury: Current State of the Art and Future Perspectives. Biomedicines 2023, 11, 689. https://doi.org/10.3390/biomedicines11030689

AMA Style

Miceli V, Bulati M, Gallo A, Iannolo G, Busà R, Conaldi PG, Zito G. Role of Mesenchymal Stem/Stromal Cells in Modulating Ischemia/Reperfusion Injury: Current State of the Art and Future Perspectives. Biomedicines. 2023; 11(3):689. https://doi.org/10.3390/biomedicines11030689

Chicago/Turabian Style

Miceli, Vitale, Matteo Bulati, Alessia Gallo, Gioacchin Iannolo, Rosalia Busà, Pier Giulio Conaldi, and Giovanni Zito. 2023. "Role of Mesenchymal Stem/Stromal Cells in Modulating Ischemia/Reperfusion Injury: Current State of the Art and Future Perspectives" Biomedicines 11, no. 3: 689. https://doi.org/10.3390/biomedicines11030689

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