Next Article in Journal
Adoption of the Robotic Platform across Thoracic Surgeries
Previous Article in Journal
The Challenges of Treating a Helicobacter pylori Infection following the COVID-19 Pandemic in Croatia: A Review
Previous Article in Special Issue
Critically Ill Patients with Newly Diagnosed Anti-Neutrophil Cytoplasmic Antibody-Associated Vasculitis: Case Series and Literature Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Comparison of Autologous and Allogeneic Adipose-Derived Stem Cells in Kidney Transplantation: Immunological Considerations and Therapeutic Efficacy

by
Ljiljana Fodor Duric
1,*,
Nikolina Basic Jukic
2 and
Bozidar Vujicic
3
1
Medicol Polyclinic, School of Medicine, Croatian Catholic Unoversity, 10000 Zagreb, Croatia
2
Department of Nephrology, Dialysis and Kidney Transplantation, Clinical Hospital Center Zagreb, Faculty of Medicine, University of Zagreb, 10000 Zagreb, Croatia
3
Department of Nephrology, Dialysis and Kidney Transplantation, Clinical Hospital Center Rijeka, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2024, 13(19), 5763; https://doi.org/10.3390/jcm13195763
Submission received: 12 August 2024 / Revised: 18 September 2024 / Accepted: 22 September 2024 / Published: 27 September 2024

Abstract

:
Regenerative medicine shows significant potential in treating kidney diseases through the application of various types of stem and progenitor cells, including mesenchymal stem cells (MSCs), renal stem/progenitor cells, embryonic stem cells (ESCs), and induced pluripotent stem cells (iPSCs). Stem cells possess the unique ability to repair injured organs and improve impaired functions, making them a key element in the research of therapies for kidney tissue repair and organ regeneration. In kidney transplantation, reperfusion injury can cause tissue destruction, leading to an initially low glomerular filtration rate and long-term impact on function by creating irreversible interstitial fibrosis. MSCs have proven useful in repairing early tissue injury in animal models of kidney, lung, heart, and intestine transplantation. The use of stem cell therapies in solid organ transplantation raises the question of whether autologous or allogeneic cells should be preferred. Adipose-derived stem cells (ASCs), characterized by the lack of HLA Class II molecules and low expression of HLA Class I and co-stimulatory signals, are considered immune-privileged. However, the actual risk of graft rejection associated with allogeneic ASCs remains unclear. It has been demonstrated that donor-derived ASCs can promote the development of Treg cells in vitro, and some degree of tolerance induction has been observed in vivo. Nevertheless, a study comparing the efficacy of autologous and allogeneic ASCs in a rat model with a total MHC mismatch for kidney transplantation showed that donor-derived administration of ASCs did not improve the grafts’ survival and was associated with increased mortality through an immunologically mediated mechanism. Given the lack of data, autologous ASCs appear to be a safer option in this research context. The aim of this review was to examine the differences between autologous and allogeneic ASCs in the context of their application in kidney transplantation therapies, considering potential immune reactions and therapeutic efficacy. Some have argued that ASCs harvested from end-stage renal disease (ESRD) patients may have lower regenerative potential due to the toxic effects of uremia, potentially limiting their use in transplantation settings. However, evidence suggests that the beneficial properties of ASCs are not affected by uremia or dialysis. Indeed, some investigators have demonstrated that ASCs harvested from chronic kidney disease (CKD) patients exhibit normal characteristics and function, maintaining consistent proliferative capacity and genetic stability over time, even after prolonged exposure to uremic serum Furthermore, no differences were observed in the response of ASCs to immune activation or their inhibitory effect on the proliferation of alloantigen-activated peripheral blood mononuclear cells between patients with normal or impaired renal function. This review presents the current achievements in stem cell research aimed at treating kidney diseases, highlighting significant progress and ongoing efforts in the development of stem cell-based therapies. Despite the encouraging results, further research is needed to overcome the current limitations and fully realize the potential of these innovative treatments. Advances in this field are crucial for developing effective therapies that can address the complex challenges associated with kidney damage and failure.

1. Introduction

Chronic kidney disease (CKD) is one of the cardinal public health problems affecting 10–15% of the general population and causing premature death. It often develops as a result of ongoing kidney injury and scarring from common conditions such as hypertension, diabetes, or nephrolithiasis. Less commonly, it can result from chronic glomerulonephritis or other diseases [1]. Patients with chronic kidney disease have endothelial dysfunction with progressive atherosclerosis and, consequently, different cardiovascular complications, which are the leading cause of death in end-stage kidney disease (ESKD) [2].
There are limited pharmacological strategies available to prevent or alleviate chronic renal failure. These strategies include managing cardiovascular risk factors, avoiding potential renal toxins, and providing causal treatment for acute kidney injury when possible. However, these options often have varying levels of success and come with frequent complications. Despite advancements in other medical fields, the lack of effective therapeutic options for chronic kidney disease (CKD) poses a significant challenge, causing frustration for both patients and healthcare providers. Furthermore, the shortage of organs and complications associated with kidney transplantation necessitates the development of new and innovative therapies. Stem cell-based therapies offer hope for a breakthrough in treating kidney diseases [1]. Given the limitations of current therapeutic approaches in treating kidney diseases and transplantation, exploring new strategies that enhance the regenerative capabilities of stem cells—either through the administration of ex vivo expanded stem cells or by promoting the expansion and differentiation of local progenitor/stem cell populations—represents a significant advancement in research and may open new avenues for improving therapies in these areas. Thus, exploring therapeutic strategies that enhance the regenerative capabilities of stem cells through the administration of stem cells expanded ex vivo or by promoting the expansion and differentiation of local progenitor/stem cell populations represents a promising frontier for future research, both in kidney diseases and kidney transplantation.

2. Mesenchymal Stem Cells

2.1. Types of Stem Cells

Stem cells can uniquely replicate and differentiate into specialized organ cells, allowing tissues to regenerate and survive most injuries [1,3]. There are four types of stem cells, defined according to their differentiation potential. During the embryonic period, the early stages of development occur due to the unlimited abilities of totipotent zygotic cells, which are later replaced by pluripotent embryonic cells. These pluripotent cells can differentiate into cells of all three germ layers but no longer have the ability to differentiate into placental cells [4].
Pluripotent cells resembling embryonic stem cells can be obtained by dedifferentiating fibroblasts or epithelial cells in vitro [5]. Somatic stem cells are found in various niches throughout life. Some are multipotent, able to transform into all cells of a certain tissue (e.g., bone marrow progenitor cells). In contrast, others are unipotent and can differentiate into only one type of mature cell (e.g., cells of the basal layer of the epidermis) [6]. Currently, the cells most extensively studied in experimental biology and medicine are mesenchymal (mesodermal) stem cells (MSCs). These cells are found in various mesodermal tissues in the human body, such as the placenta, amniotic fluid, umbilical cord, adipose tissue, testes, and lungs [7].

2.2. Regenerative Properties of MSCs

The data on mesenchymal stem cells (MSCs) indicate that they have the potential to differentiate into various types of cells, which can be useful for generating tissue replacements [8,9]. However, limited evidence shows that MSCs utilize this potential in the body [10]. Instead, most research suggests that MSCs promote tissue repair through cells’ interactions and the release of beneficial substances such as growth factors (GFs) and antioxidants [11]. These substances are released in their free form or within small vesicles called exosomes or microvesicles, allowing cellular communication [12]. Furthermore, the unique microRNA patterns within MSCs’ vesicles vary, depending on the studied disease [13]. MSCs can home in on damaged or inflamed areas due to specific receptors and molecules on their surface. Once they reach these areas, they can transfer mitochondria to the damaged cells, which can help restore cellular function and promote healing [14,15,16,17,18,19]. The transfer of mitochondria can occur through various mechanisms such as nanotube tunneling, microvesicles, or cellular fusion [20,21,22].

2.3. Immunomodulatory Properties of MSCs

MSCs can regenerate and modulate immune responses. In MSCs, MHC Class I antigens are expressed at a low level, while they do not express MHC Class II antigens or specific co-stimulatory molecules. That means that infusing allogeneic stem cells does not lead to a significant immune response [23]. MSCs possess regenerative capacities and can modulate immune responses, largely due to their secretome, which includes extracellular vesicles and exosomes. While MSCs express low levels of MHC Class I antigens and lack MHC Class II antigens, making them less likely to trigger a significant immune response, their immunomodulatory properties are mediated through the suppression of Th17 lymphocytes, the enhancement of regulatory T-cells, and the promotion of anti-inflammatory cytokines [23,24]. These mechanisms have shown promise in treating autoimmune conditions such as inflammatory joint and intestinal diseases [25,26]. Studies with exogenous MSC infusions have shown that their anti-inflammatory effects are not primarily due to direct interactions with immune cells in the inflamed tissue but rather through their secretome, partly contained within exosomes or microvesicles [27]. Carefully isolated extracellular vesicles of umbilical cord MSCs have been shown to have a strong immunosuppressant effect in vitro, as opposed to other fractions of the MSC-conditioned medium [28].

3. Source of MSCs for Research Purposes

MSCs (or, more recently, mesenchymal stromal cells), were discovered in bone marrow by Friedenstein et al. [29,30]. Over subsequent years, MSCs were isolated from different tissues and organs, including the umbilical cord, placenta, peripheral blood, adipose tissue, amniotic fluid, and skeletal muscles [31,32,33,34,35,36,37].
The most frequently used source for MSCs in clinical treatments, including treating kidney diseases, is bone marrow. However, the use of bone marrow-derived MSCs (BM-MSCs) has become limited due to factors such as the risk of viral exposure and the cells’ reduced capability for proliferation/differentiation with increased donor age. Alternative sources, such as MSCs derived from umbilical cord tissue, are being explored, as they exhibit enhanced proliferative potential and reduced immunogenicity [38]. Therefore, researchers have begun exploring other types of MSCs for kidney regeneration. Among the many sources, adipose tissue-derived MSCs (AD-MSCs) and umbilical cord-derived MSCs (UC-MSCs) have become desirable candidates because a large amount of the MSCs can be obtained using relatively minimal invasive procedures [39].
In kidney disease, MSCs are among the most efficient types of cell populations for activating regeneration in a damaged kidney [40]. Pre-clinical reports have demonstrated the therapeutic potential of MSCs in animal models of AKI and CKD [41,42]. A systematic review of more than 70 articles showed that MSCs are among the most effective cell populations for treating experimental CKD [43]. Meanwhile, in a meta-analysis involving animal models of chronic and AKI, MSCs led to kidney regeneration despite the variable modes of administration (arterial, venous, or renal) [41]. Evidence suggests the beneficial effects of MSCs in blocking the AKI–CKD transition, a term used to describe an incomplete recovery from AKI, which results in long-term functional deficits and an increased risk of developing CKD over time [44].
Fat, with its less invasive procurement process and higher concentration of MSCs than bone marrow, is a promising source of MSCs. Fat-derived MSCs also show a lower expression of MHC Class I antigens and have greater replicative and secretory potential [1,45,46]. This potential of fat-derived MSCs is an exciting area of research.
One of the most promising aspects of MSC research is the potential for a non-invasive method of collection, which involves isolating them from urine. In 2008, Zhang et al. from North Carolina identified cells in urine (at a rate of 2–7 cells per 100 mL) that were able to adhere to plastic material and form colonies of differentiated daughter cells. These daughter cells expressed membrane markers characteristic of urothelial, endothelial, and interstitial cells, or myocytes [47,48]. This non-invasive approach adds a practical dimension to the research.
In further research, these cells were successfully differentiated into endodermal, ectodermal, and mesodermal lineages using appropriate culture media [49]. Unlike mesenchymal stem cells (MSCs), up to 75% of urine-derived cells demonstrated telomerase activity, indicating a higher replicative potential without an increased risk of tumorigenesis [50]. These cells are most likely of glomerular origin—MSC-like cells with significant differentiation potential have been isolated from the renal cortical decapsulated glomeruli [51] and have shown nephroprotective effects in renal ischemia–reperfusion injury (IRI) [52]. These cells appear distinct from renal perivascular MSC-like cells, with limited differentiation capabilities (no adipogenesis) but exhibit significant kidney-reparative properties. This was demonstrated in vitro through injury to tubular epithelial cell lines or non-ischemic acute kidney injury (AKI) in mice [53].

3.1. Induction of Repair Processes after Acute Kidney Injury

One of the main areas of study for MSCs is their impact on renal ischemia–reperfusion injury, which is the most common cause of acute kidney injury (AKI) in clinical settings, such as shock, cardiac arrest, extracorporeal circulation, and the peritransplantation period. Apart from apoptosis resulting from energy deficiency and acidosis during ischemia, reperfusion leads to additional tissue damage due to oxidative stress and inflammatory reactions. Research conducted thus far has demonstrated that an MSC infusion mitigates kidney IRI. Regardless of the method of MSCs’ administration (either to the renal artery or intravenously, at different timings in relation to IRI), animal models have displayed a less severe course of acute kidney failure [54], along with reductions in oxidative damage and the local expression of inflammatory cytokines [55], an increased renal pool of regulatory T lymphocytes [56], faster regeneration of the renal tubular epithelium [57], and reduced subsequent fibrosis of the renal interstitium [58].
Intravenous infusion of MSCs was equally effective in protecting the kidneys in a model of toxin-induced AKI. In mice, 2 × 105 MSCs, injected 2 h after administration of adriamycin, reduced proteinuria and renal failure present on Day 7 in control mice. This could be the consequence of the inhibition of apoptotic processes and oxidative stress in the tubular cells [59]. These beneficial effects of MSCs are due to their secretory properties, rather than their ability to replicate and differentiate. In an experimental study, rats that underwent 40 min of kidney ischemia were given labeled allogenic bone marrow-derived MSCs (106 cells) through the aorta immediately after or 24 h after renal reperfusion. In both cases, 2 h after the infusion, MSCs were found in the kidney tissue (in peritubular and glomerular capillaries), but were not detected and did not differentiate into other cells during the subsequent 22 and 70 h. Faster normalization of renal excretory function, reduced renal expression of proinflammatory cytokines (Interleukin-1β, tumor necrosis factor α, Interferon γ), and higher renal expression of anti-inflammatory and anti-apoptotic factors, such as Interleukin-10, basic fibroblast GF, TGF α, and Bcl-2, were observed at the end of experiment [60]. Italian researchers have shown that the fraction of the MSC’s secretome responsible for this kidney-protective effect may be largely RNA. Microvesicles isolated from a human bone marrow MSC medium, administered intravenously to rats after 45 min of single-kidney ischemia, reduced acute renal failure and the atrophy of tubular cells. However, subjecting these microvesicles to RNase abolished their beneficial effects in this experimental model [61].

3.2. Immunomodulation of Kidney Transplantation

3.2.1. Animal and In Vitro Models

Ischemia–reperfusion injury (IRI) commonly occurs in kidney transplant recipients and can lead to delayed graft function. Mesenchymal stem cell (MSC) therapy has shown promise in this context due to its potential immunosuppressive effects, which could enhance the effectiveness of conventional anti-rejection medications. However, there are different protocols for administering MSCs and animal studies, and limited human observations that support using MSC-based therapies in kidney transplant patients. There is currently no clear preference for the source of MSCs (autologous, donor-derived, or third-party) [1].
In rat models of allogeneic or syngeneic kidney transplantation, infusions of allogeneic bone marrow MSCs into the graft artery during reperfusion reduced the infiltration of CD8+ lymphocytes and monocytes in the organ and mitigated graft rejection [62]. MSCs were also effective when administered intravenously. In rats, a syngeneic MSC infusion during kidney transplantation was found to reduce the expression of inflammatory cytokines in the graft [63].
In mice, MSCs administered intravenously 24 h before kidney transplantation increased the pool of regulatory T-cells in the spleen and prolonged survival of the transplanted kidney (which was not observed with the infusion performed at 24 h post-transplantation) [64].
An experimental study from Germany also found unfavorable effects of MSC infusions in the peritransplant period. Rats were treated with syngeneic or donor-derived bone marrow MSCs intravenously four days before kidney transplantation. They showed symptoms of more severe cellular and humoral rejection and worse graft function on the 10th day after graft implantation [65].

3.2.2. Human Clinical Evidence

In initial studies of human MSCs’ use in renal transplantation, adipose MSCs derived from the perirenal fat of the living kidney donor or the third-party MSCs inhibited the anti-donor and anti-third-party alloreactivity of recipient’s T lymphocytes [66].
This finding was followed by the first clinical studies of using MSCs in living-donor kidney transplant recipients conducted in Italy. In total, two patients underwent intravenous administration of autologous bone marrow MSCs one week after transplantation (1.7 × 106 and 2.0 × 106 cells per kg body weight, respectively), while the other two were given autologous MSCs 24 h before transplantation (2.0 × 106 cells per kg body weight intravenously). Over the five- to seven-year follow-up, the rate of yearly decline in mean renal function was lower by ~70% than in non-MSC-treated transplant patients [67].
It should be stressed that the recipients of MSCs showed considerable variability in their clinical course. One patient developed calcineurin inhibitor-free graft tolerance while the other one experienced acute graft rejection two weeks after transplantation, both of them being ones that were given MSCs before kidney implantation. Nevertheless, infections or neoplasms were not more frequent in the MSC-treated subjects. Except in one patient, a ~50% decrease in the percentage of memory CD8+ T-cells was observed at one year post-transplantation compared with the pre-transplant levels, which was not seen in any of the controls [67].
In the largest clinical trial conducted, 105 Chinese renal transplant recipients were administered autologous MSCs at graft reperfusion and again after two weeks in place of anti-IL-2 receptor antibodies. Such induction was associated with more rapid organ regeneration over the first month post-transplantation. Additionally, a lower rate of cellular rejection (7.6% vs. 21.6% in the control group) was recorded, with a milder course in the six-month follow-up [68].
On the other hand, studies appeared that denied the beneficial effects of intravenous infusions of MSCs on the outcome of kidney transplant; improvements in the function of renal allograft and rats’ survival were found only when allogeneic fat MSCs were injected into the graft artery, and not when they were administered intravenously at implantation [68].
In the evolving field of stem cell therapies for kidney transplantation, research has expanded beyond mesenchymal stem cells (MSCs) to include other types of stem cells, such as hematopoietic stem cells (HSCs). A notable study by Leventhal et al. investigated the use of HSCs in inducing tolerance among kidney transplant recipients. This study involved the administration of HLA-mismatched kidneys along with tolerogenic graft-facilitating cells (FCs) and HSCs, following a conditioning regimen with fludarabine, total body irradiation, and cyclophosphamide. Post-transplant immunosuppression was managed with tacrolimus and mycophenolate mofetil [69]. The results showed transient chimerism in most patients, with some achieving persistent chimerism and donor-specific tolerance, allowing them to discontinue immunosuppression after one year. However, complications such as viral sepsis and renal artery thrombosis were reported. This highlights the potential of HSCs to contribute to long-term tolerance in kidney transplantation but also underscores the associated risks, including infection-related complications, that need to be addressed. Integrating HSC-based therapies into our discussion underscores the diversity of approaches in stem cell research for kidney transplantation and the need for ongoing investigation to optimize these strategies for clinical use.
Even more discouraging are the recent findings of another Chinese team of researchers, who injected allogeneic umbilical cord blood MSCs into 21 recipients intravenously immediately prior to transplantation (2 × 106/kg body weight) and, additionally, into the graft artery at reperfusion (5 × 106), in addition to standard immunosuppression. In the one-year follow-up period, no statistically significant differences were found from the controls regarding postoperative and infectious complications, renal function, the frequency of rejection, or the survival time of the kidney transplant [70].
This table provides an overview of key findings from various studies on the use of mesenchymal stem cells (MSCs) in kidney transplantation. It includes information on the source and administration methods of MSCs, the main outcomes observed in both animal models and human clinical trials, and their effects on kidney transplant outcomes. The Table 1 highlights differences in administration protocols for MSCs, the impact on graft function and survival, and notable variations in clinical responses among patients. This summary aims to offer a concise comparison of the effectiveness and challenges associated with MSC therapies in the context of kidney transplantation.

3.2.3. Clonal Heterogeneity of MSC Cultures

Another important aspect to consider in the context of MSC-based therapies is the clonal heterogeneity of MSC cultures. This heterogeneity can partially explain the significant variability in clinical outcomes observed among patients who have received MSCs for modulation of transplantation-related immune tolerance.
Mesenchymal stem cells (MSCs) are known to exhibit substantial clonal heterogeneity, meaning that even within a single MSC culture, there can be a diverse range of cell subpopulations with differing properties and functions. This variability can result from differences in the cells’ origin, the passage number, and the microenvironment in which the MSCs are expanded [72,73].
The presence of different MSC subpopulations within a culture can lead to variability in the therapeutic outcomes. Some MSCs might have stronger immunomodulatory capabilities, while others may not be as effective, leading to inconsistent results across clinical trials. This clonal heterogeneity can impact the efficacy of MSC-based therapies in modulating immune tolerance and influencing transplant outcomes [74].
To address this issue, researchers and clinicians should consider the following:
(1)
Standardization of MSC cultures: Developing standardized protocols for the isolation, expansion, and characterization of MSCs can help minimize clonal heterogeneity and improve consistency in clinical results [75].
(2)
Characterization of MSC subpopulations: Employing advanced techniques to identify and quantify different MSC subpopulations within a culture can provide insights into their functional capabilities and potential therapeutic efficacy [76].
(3)
Personalized approaches: Tailoring MSC therapies to individual patients based on their specific immune profiles and MSC characteristics may enhance the effectiveness and predictability of treatment outcomes [77].
By addressing clonal heterogeneity, we can better understand and potentially mitigate the variability in clinical outcomes associated with MSC therapies, ultimately improving their application in transplantation and other therapeutic areas [Figure 1].

4. Mesenchymal Stem Cells and Transplant Tolerance

Mesenchymal stem cells (MSCs) possess exceptional capabilities that render them highly valuable in various fields of medicine, including transplant immunology.
The unique combination of plasticity and non-immunogenicity makes MSCs promising candidates for various therapeutic applications, particularly in transplant immunology, where their ability to modulate immune responses can potentially improve outcomes in transplantation and reduce the need for immunosuppressive drugs [78].
MSCs must meet specific criteria, including adherence to plastic material under standard culture conditions; expression of CD105, CD73, and CD90 markers; and the absence of CD34, CD45, CD11a, CD19, CD79a, CD14, CD11b, and histocompatibility locus antigen (HLA)-DR. Additionally, they must demonstrate the ability to differentiate into osteocytes and adipocytes in response to specific stimuli [79,80].

Experimental Studies

Le Blanc et al. [81] obtained bone marrow from healthy human volunteers and cultured mesenchymal stem cells (MSCs). MSCs isolated from the second or third passages were then co-cultured with peripheral blood lymphocytes in different ratios, and they found that MSCs showed diverse responses, including inhibition of T-lymphocytes’ proliferation and sometimes even stimulation of DNA synthesis.
One significant mechanism of action for MSCs involves the secretion of HLA-G5, which plays a crucial role in suppressing T-cells and NK cells, shifting the T-cells’ response towards T-helper Type 2 (Th2), and promoting the generation of T-regulatory cells (CD4+ CD25hi forkhead box P3 (FoxP3+) [81,82,83].
Rodent models are commonly used in biomedical research to study various diseases, conditions, and treatments before moving to human trials. In the context of transplantation and MSC research, rodent models (such as mice or rats) are used to mimic aspects of human physiology and immune responses. Rodent models offer a controlled environment in which to study the effects of MSCs on the outcomes of transplantation. Specifically, in the study by Casiraghi et al. [71,84], they investigated the timing and dosage effects of MSCs in a rodent transplantation model. Their study revealed that administering autologous MSCs post-transplantation in murine models resulted in increased neutrophil infiltration and the deposition of complement in the renal allograft, ultimately leading to rejection.
Conversely, when MSCs were administered before transplantation, they targeted lymphoid organs, which improved graft survival and promoted the generation of T-regulatory cells. Therefore, these investigations collectively highlighted the promising potential of MSCs in fostering transplant tolerance prior to solid organ transplantation.
After previous failures in achieving tolerance, researchers renewed their interest in stem cell therapy when Scandling et al. infused donor HSCs in 12 patients who underwent HLA-matched kidney transplantation under a non-myeloablative conditioning regimen [71]. Ten days after kidney transplantation, these patients underwent a conditioning regimen that included 10 doses of TLI (80 to 120 cGy) targeting the lymph nodes, spleen, and thymus, along with five doses of rabbit anti-thymocyte globulin. On Day 11, CD34+ selected cells from the donors (ranging from 5 × 106 to 16 × 106/kg) and a defined dose of T-cells (ranging from 1 × 106 to 10 × 106 per kilogram) were intravenously injected at the outpatient infusion center. All patients received mycophenolate mofetil for one month and cyclosporine, starting at Day 0 for at least six months. Cyclosporine was discontinued 6 to 17 months after transplantation as long as chimerism persisted for at least 6 months, with no evidence of graft-versus-host disease, clinical rejection, or surveillance biopsy-proven rejection at the time of withdrawal. In the study by Le Blanc et al. [85], the hematopoietic stem cells were modified through non-myeloablative conditioning and co-infusion with donor-derived mesenchymal stem cells to promote tolerance. This combination aimed to reduce the risk of graft-versus-host disease while encouraging the development of stable mixed chimerism, thus facilitating the discontinuation of immunosuppressive therapy.
They reported success in 8 out of 12 patients and conducted a mean follow-up of 25 months. However, they noted a recurrence of focal segmental glomerulosclerosis (FSGS) in one patient. This conditioning can be lethal to patients, especially in developing countries, where infection risks are higher and markers of immune tolerance, as well as regular monitoring, are not clearly addressed. Another important fact to mention is that recipient–donor HLA matching is mandatory, which may only be clinically feasible sometimes.
In research conducted by Leventhal et al. [69], they attempted to induce tolerance in eight kidney transplant recipients using hematopoietic stem cells (HSCs) within a conditioning regimen. Key aspects of this study include the administration of HLA-mismatched kidneys and tolerogenic graft facilitating cells (FCs), alongside HSCs, following conditioning with fludarabine, 200-centigray total body irradiation, and cyclophosphamide. Post-transplant immunosuppression was maintained with tacrolimus and mycophenolate mofetil [69]. The lowest absolute neutrophil counts were observed approximately one week after transplantation, followed by recovery within two weeks. Multilineage chimerism ranged from 6% to 100% in their patients at one month post-transplant. The conditioning regimen was well tolerated, and the patients were managed on an outpatient basis, starting from postoperative Day 2. Cyclosporine was discontinued 6 to 17 months after transplantation as long as chimerism persisted for at least 6 months, with no evidence of graft-versus-host disease, clinical rejection, or surveillance biopsy-proven rejection at the time of withdrawal. In the study by Le Blanc [85], the hematopoietic stem cells were modified through non-myeloablative conditioning and co-infusion with donor-derived mesenchymal stem cells to promote tolerance. This combination aimed to reduce the risk of graft-versus-host disease while encouraging the development of stable mixed chimerism, thus facilitating the discontinuation of immunosuppressive therapy.
The complications following transplantation in this case included transient chimerism (treated with low-dose tacrolimus), viral sepsis two months after transplant, and renal artery thrombosis.
Five subjects maintained persistent chimerism, exhibited immunocompetence, and showed donor-specific tolerance through in vitro proliferative assays. They successfully discontinued all immunosuppression one year after transplantation. None of the recipients produced anti-donor antibodies, exhibited engraftment syndrome, or developed graft-versus-host disease. The authors concluded that modifying a mobilized stem cell graft and using non-myeloablative conditioning presents a safe, practical, and reliable method for achieving persistent chimerism and donor-specific tolerance in recipients of solid organ transplants. The application of therapy involving the manipulation of stem cell grafts and non-myeloablative conditioning may increase the risk of infection-related complications, making this strategy less safe and less encouraging in dialysis settings.
Tan et al. [68] conducted a study involving autologous bone marrow-derived MSCs in 105 renal transplant (RT) patients. They administered MSCs twice: before the anastomosis and two weeks after the renal transplant (RT), and they found that bone marrow-derived MSCs were safe and led to improved renal function, along with a reduced incidence of infections during one year of follow-up [81].
Ongoing trials across all continents are investigating the use of bone marrow-derived MSCs to mitigate tissue injury in autoimmune disorders and enhance the long-term success of transplants.
Perico et al. [86] administered autologous MSCs seven days after renal transplantation in two recipients who received living-related kidneys. These patients underwent T-cell depletion therapy and continued to take cyclosporine and mycophenolate mofetil as maintenance immunosuppressive therapy. They were monitored for approximately one year after the procedure, and at the one-year mark, both patients showed increases in T-regulatory cells (CD4+CD25high FoxP3+ CD127−), along with a decrease in CD8+ cells and stable graft function.
A study by Aruna et al. [87] investigated 606 living donor kidney transplants. They aimed to eliminate rejecting T- and B-cells using non-myeloablative conditioning, which included total lymphoid irradiation (200 cGy administered over 4 or 5 days), bortezomib (1.5 mg/kg body weight in four divided doses every third day), cyclophosphamide (20 mg/kg body weight), and rabbit anti-thymocyte globulin (1.5 mg/kg body weight). They infused a combination of MSCs derived from adipose tissue and HSCs into the portal and thymic circulation, taking advantage of the liver’s high tolerance due to its unique microanatomy and diverse functional characteristics. Cells entering the thymus undergo processes of positive and negative selection, which leads to the development of T-cells that can respond to a wide array of foreign antigens while avoiding reactivity against self-antigens. The thymus produces a specific type of regulatory T-cells that suppress the self-reactivity of T-cell clones that might evade negative selection. Therefore, the thymus is considered crucial for promoting tolerance [87].
Several investigations have examined bone marrow-derived MSCs (BM-MSCs) and adipose tissue-derived MSCs (AT-MSCs), often with small donor groups and varying patient ages. These studies indicated that both types of MSCs may suppress the proliferation of peripheral blood mononuclear cells (PBMCs), yet they observed no notable differences between the two MSC populations [88,89,90]. Research conducted by Sara M. Melief et al. [46] suggested that adipose tissue-derived multipotent stromal cells (AT-MSCs) might be a more effective alternative for immunomodulatory treatment compared with bone marrow-derived multipotent stromal cells (BM-MSCs). The research compared the immunomodulatory abilities of BM-MSCs and AT-MSCs from age-matched donors, revealing that both cell types possess a similar immunophenotype and in vitro multilineage differentiation potential. Although both BM-MSCs and AT-MSCs are used to suppress the proliferation of activated peripheral blood mononuclear cells and inhibit the differentiation of monocyte-derived immature dendritic cells, AT-MSCs displayed stronger immunomodulatory effects at equivalent cell numbers [91]. This enhanced effect is due to the increased secretion of crucial cytokines such as Interleukin-6 and transforming growth factor-β1, along with the higher metabolic activity of AT-MSCs. As a result, fewer AT-MSCs are needed to achieve the same level of immunomodulation, suggesting their potential as a superior option for immunomodulatory therapy [92].
The overall explanation for the superior functionality of AT-MSCs is likely their heightened metabolic activity, which leads to the production of greater levels of cytokines involved in the immunosuppressive mechanisms of MSCs. This applies to other factors in MSC-mediated immunomodulation as well, such as PGE2, galectin-1, and HLA-G5 [93].
In summary, AT-MSCs (adipose tissue-derived multipotent stromal cells) have been shown to be superior to BM-MSCs (bone marrow-derived multipotent stromal cells) due to several key factors as follows.
  • Higher metabolic activity: AT-MSCs have greater metabolic activity, meaning they can produce more energy and biological molecules that are necessary for their functions.
  • Increased cytokine production: cytokines are proteins with a cardinal role in the regulation of the immune response. AT-MSCs produce higher amounts of cytokines such as Interleukin-6 and transforming growth factor-β1, which are important for immunosuppressive mechanisms.
  • Other immunomodulatory factors: in addition to cytokines, AT-MSCs produce other molecules such as PGE2 (prostaglandin E2), galectin-1, and HLA-G5, which contribute to their ability to modulate the immune response.
All of these together mean that AT-MSCs can be more effective at suppressing undesirable immune reactions, making them potentially a better choice for therapies that require immunomodulation [93,94].
Studies performed on animal and human models have demonstrated that MSCs derived from bone marrow are safe and feasible for treating autoimmune disorders and protecting grafts from injury during transplantation, leading to improved long-term outcomes. Some unresolved issues include identifying the most suitable source of MSCs, determining the optimal timing and dosage for infusion, selecting the infusion site, and assessing the effectiveness of recovery and/or the reduction in immunosuppression.

5. Discussion

Kidney transplantation (KT) continues to be the main therapeutic approach for end-stage renal disease (ESRD). Nonetheless, the widening gap between the availability of organs and patients’ needs has led to more inclusive donor criteria. This shift has resulted in an increase in the use of marginal kidneys for transplantation in high-risk patients. This proactive strategy has notably increased the frequency of delayed graft function (DGF), which is characterized by the requirement for dialysis within the first week after transplantation [95]. DGF is a complex condition that negatively impacts the survival of both the patient and the graft [96]. Major risk factors for DGF encompass expanded criteria donors (ECD), donation after circulatory death (DCD), extended warm ischemia time (WIT) or cold ischemia time (CIT), and sensitization of the recipient [97].
Considering the different peritransplant events that contribute to the development of DGF, ischemia–reperfusion injury (IRI) deserves special attention, as it is unavoidable and a major factor in acute tubular necrosis (ATN), the primary histological finding associated with DGF [98]. IRI is associated with a substantial proinflammatory response that can initiate various cell death pathways, lead to endothelial dysfunction, cause transcriptional reprogramming, and activate both the innate and adaptive immune systems [94]. Due to the strong correlations of IRI, DGF, acute rejection (AR), and progressive interstitial fibrosis with tubular atrophy (IF/TA), as well as their detrimental impact on kidney allografts’ function and survival, the prevention and treatment of IRI have become primary concerns for the transplant community [99].
Growing evidence suggests that adipose stem/stromal cells (ASCs) have unique traits that might aid in preventing, mitigating, or reversing IRI. Additionally, their immunomodulatory and tolerogenic properties have led to the exploration of ASC-based preventive and therapeutic approaches in both pre-clinical and clinical models of renal IRI and allograft rejection. ASCs are plentiful, easily obtainable, and can be readily expanded in culture. Moreover, ASCs have the capability to release extracellular vesicles (EVs), which might serve as effective agents for tissue repair and promoting tolerance. This review examines the current understanding of how ASCs and ASC-derived EVs function and the therapeutic potential they offer in the context of kidney transplantation.
The most relevant pre-clinical and clinical studies, as well as actual limitations and future perspectives, are highlighted.
Key findings from the studies highlight several important aspects, as follows.
  • Safety and feasibility: most studies have confirmed the safety of the application of MSCs in the context of transplantation, with minimal reported adverse effects or complications associated with the therapy. This is crucial for further advancing the clinical applications.
  • Impact on immunosuppression: some studies have suggested that MSCs may reduce the need for immunosuppressive therapy post-transplantation. This is significant, as it could improve the long-term outcomes of transplanted organs and reduce the risks associated with immunosuppression.
  • Need for further research: despite positive findings, there are still open questions, such as the optimal dosage, timing, and method of administration of MSCs. Further research is needed to understand these aspects better and define the best clinical practices.
  • Perspectives on transplant tolerance: studies exploring the combination of MSCs and hematopoietic stem cells (HSCs) as a means to induce transplant tolerance present particularly promising results. This approach can shift the standard paradigm in transplantation medicine towards strategies promoting immune tolerance.
  • Challenges and opportunities: despite progress, challenges such as the need for individualized approaches for each patient and the requirement for further studies to confirm the long-term clinical benefits of MSCs’ application.
  • Impact of socioeconomic factors: infections remain a major challenge for all transplantations, especially in developing countries, where the social, economic, and environmental conditions do not support optimal health outcomes. In developing countries, infections such as tuberculosis, cytomegalovirus, and bacterial infections significantly affect post-transplant outcomes. The financial burden of transplantation, coupled with limited access to healthcare and lack of insurance coverage for dialysis after graft failure, exacerbates the economic hardships faced by patients and their families. Research on transplant tolerance using MSCs holds promise for improving outcomes in these vulnerable populations by potentially reducing the need for lifelong immunosuppressive medications and the associated costs.
  • Economic benefits: The use of MSCs, including adipose tissue-derived MSCs (AD-MSCs), has shown promise in reducing the overall cost burden of transplantation. For instance, in Ahmedabad, India, the total cost of transplantation using AD-MSCs was approximately USD 6000, significantly lower than traditional transplantation costs. This reduction lowered the financial strain on patients and decreased the monthly healthcare costs from approximately USD 2000 to less than USD 50. Additionally, minimizing infections due to reduced immunosuppressive requirements allows patients to return to work and have a normal life, improving overall quality of life post-transplant.

5.1. Limitations of MSC-Based Therapies

While mesenchymal stem cell (MSC) therapies offer promising potential in kidney transplantation, several limitations and concerns must be addressed to fully understand their efficacy and safety. One significant issue is the potential activation of the complement system during the infusion of MSCs [100,101].

5.2. Complement Activation

The complement system, a key component of the innate immune response, can be activated during the infusion of MSCs. Complement activation can lead to inflammation and tissue damage, which may counteract the therapeutic benefits of MSCs. Several studies have indicated that the infusion of MSCs might trigger complement activation, potentially leading to adverse effects such as an increased risk of graft rejection or impaired graft function [102,103,104].
In clinical settings, complement activation during the infusion of MSCs has been observed in some patients, leading to concerns about the safety and efficacy of these therapies. Monitoring markers of complement activation and implementing strategies to mitigate this activation are crucial for improving patient outcomes and optimizing MSC-based therapies.
Future studies should focus on the following.
(1)
Assessing the extent of complement activation in different MSC infusion protocols [105].
(2)
Developing strategies to minimize complement activation, such as using complement inhibitors or optimizing the preparation methods of MSCs [106].
(3)
Evaluating the impact of complement activation on long-term transplant outcomes and patients’ safety [107].
(4)
By addressing these limitations, researchers can better harness the potential of MSC therapies and enhance their application in kidney transplantation.
In conclusion, MSCs have a promising role in the induction and sustenance of transplant tolerance when infused into the liver and thymic circulation pre-transplant. Further experimental and clinical trials are urgently needed to fully explore their potential benefits and refine the protocols for widespread clinical implementation.

Author Contributions

Conceptualization, L.F.D.; investigation, L.F.D.; resources, L.F.D. and N.B.J.; data curation, L.F.D. and B.V.; writing—original draft preparation, L.F.D.; writing—review and editing, L.F.D. and B.V.; visualization, L.F.D. and B.V.; supervision, N.B.J.; funding acquisition, N.B.J. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding. The APC was funded by the Croatian Society for Kidney, Croatian Medical Association.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Bochon, B.; Kozubska, M.; Surygała, G.; Witkowska, A.; Kuźniewicz, R.; Grzeszczak, W.; Wystrychowski, G. Mesenchymal Stem Cells—Potential Applications in Kidney Diseases. Int. J. Mol. Sci. 2019, 20, 2462. [Google Scholar] [CrossRef] [PubMed]
  2. Maria, J.S. Stem cells in kidney diseases. J. Stem Cells 2012, 7, 245–259. [Google Scholar]
  3. Bonaventura, G.; Chamayou, S.; Liprino, A.; Guglielmino, A.; Fichera, M.; Caruso, M.; Barcellona, M.L. Different Tissue-Derived Stem Cells: A Comparison of Neural Differentiation Capability. PLoS ONE 2015, 10, e0140790. [Google Scholar] [CrossRef] [PubMed]
  4. Mitalipov, S.; Wolf, D. Totipotency, pluripotency and nuclear reprogramming. Adv. Biochem. Eng. Biotechnol. 2009, 114, 185–199. [Google Scholar] [PubMed]
  5. Yu, J.; Vodyanik, M.A.; Smuga-Otto, K.; Antosiewicz-Bourget, J.; Frane, J.L.; Tian, S.; Nie, J.; Jonsdottir, G.A.; Ruotti, V.; Stewart, R.; et al. Induced pluripotent stem cell lines derived from human somatic cells. Science 2007, 318, 1917–1920. [Google Scholar] [CrossRef] [PubMed]
  6. Biehl, J.K.; Russell, B. Introduction to stem cell therapy. J. Cardiovasc. Nurs. 2009, 24, 98–103; quiz 104–105. [Google Scholar] [CrossRef]
  7. Kim, N.; Cho, S.-G. Clinical applications of mesenchymal stem cells. Korean J. Intern. Med. 2013, 28, 387–402. [Google Scholar] [CrossRef]
  8. Kariminekoo, S.; Movassaghpour, A.; Rahimzadeh, A.; Talebi, M.; Shamsasenjan, K.; Akbarzadeh, A. Implications of mesenchymal stem cells in regenerative medicine. Artif. Cells Nanomed. Biotechnol. 2016, 44, 749–757. [Google Scholar] [CrossRef]
  9. Jeon, B.-G.; Jang, S.-J.; Park, J.-S.; Subbarao, R.B.; Jeong, G.-J.; Park, B.-W.; Rho, G.-J. Differentiation potential of mesenchymal stem cells isolated from human dental tissues into non-mesodermal lineage. Anim. Cells Syst. 2015, 19, 321–331. [Google Scholar] [CrossRef]
  10. Seo, M.J.; Suh, S.Y.; Bae, Y.C.; Jung, J.S. Differentiation of human adipose stromal cells into hepatic lineage in vitro and in vivo. Biochem. Biophys. Res. Commun. 2005, 328, 258–264. [Google Scholar] [CrossRef]
  11. Murphy, M.B.; Moncivais, K.; Caplan, A.I. Mesenchymal stem cells: Environmentally responsive therapeutics for regenerative medicine. Exp. Mol. Med. 2013, 45, e54. [Google Scholar] [CrossRef] [PubMed]
  12. Belting, M.; Wittrup, A. Nanotubes, exosomes, and nucleic acid-binding peptides provide novel mechanisms of intercellular communication in eukaryotic cells: Implications in health and disease. J. Cell Biol. 2008, 183, 1187–1191. [Google Scholar] [CrossRef] [PubMed]
  13. Qiu, G.; Zheng, G.; Ge, M.; Wang, J.; Huang, R.; Shu, Q.; Xu, J. Mesenchymal stem cell-derived extracellular vesicles affect disease outcomes via transfer of microRNAs. Stem Cell Res. Ther. 2018, 9, 320. [Google Scholar] [CrossRef] [PubMed]
  14. Lotfi, R.; Eisenbacher, J.; Solgi, G.; Fuchs, K.; Yildiz, T.; Nienhaus, C.; Rojewski, M.T.; Schrezenmeier, H. Human mesenchymal stem cells respond to native but not oxidized damage associated molecular pattern molecules from necrotic (tumor) material. Eur. J. Immunol. 2011, 41, 2021–2028. [Google Scholar] [CrossRef]
  15. Mahrouf-Yorgov, M.; Augeul, L.; Da Silva, C.C.; Jourdan, M.; Rigolet, M.; Manin, S.; Ferrera, R.; Ovize, M.; Henry, A.; Guguin, A.; et al. Mesenchymal stem cells sense mitochondria released from damaged cells as danger signals to activate their rescue properties. Cell Death Differ. 2017, 24, 1224–1238. [Google Scholar] [CrossRef] [PubMed]
  16. Spees, J.L.; Olson, S.D.; Whitney, M.J.; Prockop, D.J. Mitochondrial transfer between cells can rescue aerobic respiration. Proc. Natl. Acad. Sci. USA 2006, 103, 1283–1288. [Google Scholar] [CrossRef]
  17. Wang, X.; Gerdes, H.-H. Transfer of mitochondria via tunneling nanotubes rescues apoptotic PC12 cells. Cell Death Differ. 2015, 22, 1181–1191. [Google Scholar] [CrossRef]
  18. Jiang, D.; Gao, F.; Zhang, Y.; Wong DS, H.; Li, Q.; Tse, H.F.; Xu, G.; Yu, Z.; Lian, Q. Mitochondrial transfer of mesenchymal stem cells effectively protects corneal epithelial cells from mitochondrial damage. Cell Death Dis. 2016, 7, e2467. [Google Scholar] [CrossRef] [PubMed]
  19. Islam, M.N.; Das, S.R.; Emin, M.T.; Wei, M.; Sun, L.; Westphalen, K.; Rowlands, D.J.; Quadri, S.K.; Bhattacharya, S.; Bhattacharya, J. Mitochondrial transfer from bone-marrow-derived stromal cells to pulmonary alveoli protects against acute lung injury. Nat. Med. 2012, 18, 759–765. [Google Scholar] [CrossRef]
  20. Liu, K.; Ji, K.; Guo, L.; Wu, W.; Lu, H.; Shan, P.; Yan, C. Mesenchymal stem cells rescue injured endothelial cells in an in vitro ischemia-reperfusion model via tunneling nanotube like structure-mediated mitochondrial transfer. Microvasc. Res. 2014, 92, 10–18. [Google Scholar] [CrossRef]
  21. Phinney, D.G.; Di Giuseppe, M.; Njah, J.; Sala, E.; Shiva, S.; St Croix, C.M.; Stolz, D.B.; Watkins, S.C.; Di, Y.P.; Leikauf, G.D.; et al. Mesenchymal stem cells use extracellular vesicles to outsource mitophagy and shuttle microRNAs. Nat. Commun. 2015, 6, 8472. [Google Scholar] [CrossRef] [PubMed]
  22. Torralba, D.; Baixauli, F.; Sánchez-Madrid, F. Mitochondria Know No Boundaries: Mechanisms and Functions of Intercellular Mitochondrial Transfer. Front. Cell Dev. Biol. 2016, 4, 107. [Google Scholar] [CrossRef]
  23. Le Blanc, K. Immunomodulatory effects of fetal and adult mesenchymal stem cells. Cytotherapy 2003, 5, 485–489. [Google Scholar] [CrossRef] [PubMed]
  24. Luz-Crawford, P.; Kurte, M.; Bravo-Alegría, J.; Contreras, R.; Nova-Lamperti, E.; Tejedor, G.; Noël, D.; Jorgensen, C.; Figueroa, F.; Djouad, F.; et al. Mesenchymal stem cells generate a CD4+CD25+Foxp3+ regulatory T cell population during the differentiation process of Th1 and Th17 cells. Stem Cell Res. Ther. 2013, 4, 65. [Google Scholar] [CrossRef] [PubMed]
  25. Maumus, M.; Guérit, D.; Toupet, K.; Jorgensen, C.; Noël, D. Mesenchymal stem cell-based therapies in regenerative medicine: Applications in rheumatology. Stem Cell Res. Ther. 2011, 2, 14. [Google Scholar] [CrossRef]
  26. Duran, N.E.; Hommes, D.W. Stem cell-based therapies in inflammatory bowel disease: Promises and pitfalls. Ther. Adv. Gastroenterol. 2016, 9, 533–547. [Google Scholar] [CrossRef] [PubMed]
  27. Zhang, B.; Yin, Y.; Lai, R.C.; Tan, S.S.; Choo, A.B.H.; Lim, S.K. Mesenchymal stem cells secrete immunologically active exosomes. Stem Cells Dev. 2014, 23, 1233–1244. [Google Scholar] [CrossRef]
  28. Monguió-Tortajada, M.; Roura, S.; Gálvez-Montón, C.; Pujal, J.M.; Aran, G.; Sanjurjo, L.; la Franquesa, M.; Sarrias, M.-R.; Bayes-Genis, A.; Borràs, F.E. Nanosized UCMSC-derived extracellular vesicles but not conditioned medium exclusively inhibit the inflammatory response of stimulated T cells: Implications for nanomedicine. Theranostics 2017, 7, 270–284. [Google Scholar] [CrossRef]
  29. Wong, C.Y. Current advances of stem cell-based therapy for kidney diseases. World J. Stem Cells 2021, 13, 914–933. [Google Scholar] [CrossRef] [PubMed]
  30. Friedenstein, A.J.; Deriglasova, U.F.; Kulagina, N.N.; Panasuk, A.F.; Rudakowa, S.F.; Luriá, E.A.; Ruadkow, I.A. Precursors for fibroblasts in different populations of hematopoietic cells as detected by the in vitro colony assay method. Exp. Hematol. 1974, 2, 83–92. [Google Scholar]
  31. Mahmoudifar, N.; Doran, P.M. Mesenchymal Stem Cells Derived from Human Adipose Tissue. Methods Mol. Biol. 2015, 1340, 53–64. [Google Scholar] [PubMed]
  32. Lu, L.L.; Liu, Y.J.; Yang, S.G.; Zhao, Q.J.; Wang, X.; Gong, W.; Han, Z.B.; Xu, Z.S.; Lu, Y.X.; Liu, D.; et al. Isolation and characterization of human umbilical cord mesenchymal stem cells with hematopoiesis-supportive function and other potentials. Haematologica 2006, 91, 1017–1026. [Google Scholar] [PubMed]
  33. Choo, K.B.; Tai, L.; Hymavathee, K.; Wong, C.Y.; Nguyen, P.N.N.; Huang, C.-J.; Cheong, S.K.; Kamarul, T. Oxidative stress-induced premature senescence in Wharton’s jelly-derived mesenchymal stem cells. Int. J. Med Sci. 2014, 11, 1201–1207. [Google Scholar] [CrossRef] [PubMed]
  34. Talwadekar, M.D.; Kale, V.P.; Limaye, L.S. Placenta-derived mesenchymal stem cells possess better immunoregulatory properties compared to their cord-derived counterparts—A paired sample study. Sci. Rep. 2015, 5, 15784. [Google Scholar] [CrossRef]
  35. Longhini, A.L.F.; Salazar, T.E.; Vieira, C.; Trinh, T.; Duan, Y.; Pay, L.M.; Calzi, S.L.; Losh, M.; Johnston, N.A.; Xie, H.; et al. Peripheral blood-derived mesenchymal stem cells demonstrate immunomodulatory potential for therapeutic use in horses. PLoS ONE 2019, 14, e0212642. [Google Scholar] [CrossRef] [PubMed]
  36. Spitzhorn, L.-S.; Rahman, S.; Schwindt, L.; Ho, H.-T.; Wruck, W.; Bohndorf, M.; Wehrmeyer, S.; Ncube, A.; Beyer, I.; Hagenbeck, C.; et al. Isolation and molecular characterization of amniotic fluid-derived mesenchymal stem cells obtained from caesarean sections. Stem Cells Int. 2017, 2017, 5932706. [Google Scholar] [CrossRef]
  37. Čamernik, K.; Mihelič, A.; Mihalič, R.; Presen, D.M.; Janež, A.; Trebše, R.; Marc, J.; Zupan, J. Skeletal-muscle-derived mesenchymal stem/stromal cells from patients with osteoarthritis show superior biological properties compared to bone-derived cells. Stem Cell Res. 2019, 38, 101465. [Google Scholar] [CrossRef]
  38. Li, X.; Bai, J.; Ji, X.; Li, R.; Xuan, Y.; Wang, Y. Comprehensive characterization of four different populations of human mesenchymal stem cells as regards their immune properties, proliferation and differentiation. Int. J. Mol. Med. 2014, 34, 695–704. [Google Scholar] [CrossRef]
  39. Mastrolia, I.; Foppiani, E.M.; Murgia, A.; Candini, O.; Samarelli, A.V.; Grisendi, G.; Veronesi, E.; Horwitz, E.M.; Dominici, M. Challenges in Clinical Development of Mesenchymal Stromal/Stem Cells: Concise Review. Stem Cells Transl. Med. 2019, 8, 1135–1148. [Google Scholar] [CrossRef] [PubMed]
  40. Rota, C.; Morigi, M.; Imberti, B. Stem Cell Therapies in Kidney Diseases: Progress and Challenges. Int. J. Mol. Sci. 2019, 20, 2790. [Google Scholar] [CrossRef]
  41. Wang, Y.; He, J.; Pei, X.; Zhao, W. Systematic review and meta-analysis of mesenchymal stem/stromal cells therapy for impaired renal function in small animal models. Nephrology 2013, 18, 201–208. [Google Scholar] [CrossRef]
  42. Sávio-Silva, C.; Soinski-Sousa, P.E.; Balby-Rocha, M.T.A.; de Oliveira Lira, Á; Rangel, É.B. Mesenchymal stem cell therapy in acute kidney injury (AKI): Review and perspectives. Rev. Assoc. Med. Bras. 2020, 66 (Suppl. 1), s45–s54. [Google Scholar] [CrossRef] [PubMed]
  43. Papazova, D.A.; Oosterhuis, N.R.; Gremmels, H.; van Koppen, A.; Joles, J.A.; Verhaar, M.C. Cell-based therapies for experimental chronic kidney disease: A systematic review and meta-analysis. Dis. Model. Mech. 2015, 8, 281–293. [Google Scholar] [CrossRef] [PubMed]
  44. Zhao, L.; Han, F.; Wang, J.; Chen, J. Current understanding of the administration of mesenchymal stem cells in acute kidney injury to chronic kidney disease transition: A review with a focus on preclinical models. Stem Cell Res. Ther. 2019, 10, 385. [Google Scholar] [CrossRef] [PubMed]
  45. Strioga, M.; Viswanathan, S.; Darinskas, A.; Slaby, O.; Michalek, J. Same or not the same? Comparison of adipose tissue-derived versus bone marrow-derived mesenchymal stem and stromal cells. Stem Cells Dev. 2012, 21, 2724–2752. [Google Scholar] [CrossRef] [PubMed]
  46. Melief, S.M.; Zwaginga, J.J.; Fibbe, W.E.; Roelofs, H. Adipose tissue-derived multipotent stromal cells have a higher immunomodulatory capacity than their bone marrow-derived counterparts. Stem Cells Transl. Med. 2013, 2, 455–463. [Google Scholar] [CrossRef] [PubMed]
  47. Lian, Q.; Zhang, Y.; Zhang, J.; Zhang, H.K.; Wu, X.; Zhang, Y.; Lam, F.F.-Y.; Kang, S.; Xia, J.C.; Lai, W.-H.; et al. Functional mesenchymal stem cells derived from human induced pluripotent stem cells attenuate limb ischemia in mice. Circulation 2010, 121, 1113–1123. [Google Scholar] [CrossRef]
  48. Zhang, Y.; McNeill, E.; Tian, H.; Soker, S.; Andersson, K.E.; Yoo, J.J.; Atala, A. Urine derived cells are a potential source for urological tissue reconstruction. J. Urol. 2008, 180, 2226–2233. [Google Scholar] [CrossRef]
  49. Bharadwaj, S.; Liu, G.; Shi, Y.; Wu, R.; Yang, B.; He, T.; Fan, Y.; Lu, X.; Zhou, X.; Liu, H.; et al. Multipotential differentiation of human urine-derived stem cells: Potential for therapeutic applications in urology. Stem Cells 2013, 31, 1840–1856. [Google Scholar] [CrossRef]
  50. Zhang, D.; Wei, G.; Li, P.; Zhou, X.; Zhang, Y. Urine-derived stem cells: A novel and versatile progenitor source for cell-based therapy and regenerative medicine. Genes Dis. 2014, 1, 8–17. [Google Scholar] [CrossRef]
  51. Bruno, S.; Bussolati, B.; Grange, C.; Collino, F.; di Cantogno, L.V.; Herrera, M.B.; Biancone, L.; Tetta, C.; Segoloni, G.; Camussi, G. Isolation and characterization of resident mesenchymal stem cells in human glomeruli. Stem Cells Dev. 2009, 18, 867–880. [Google Scholar] [CrossRef]
  52. Ranghino, A.; Bruno, S.; Bussolati, B.; Moggio, A.; Dimuccio, V.; Tapparo, M.; Biancone, L.; Gontero, P.; Frea, B.; Camussi, G. The effects of glomerular and tubular renal progenitors and derived extracellular vesicles on recovery from acute kidney injury. Stem Cell Res. Ther. 2017, 8, 24. [Google Scholar] [CrossRef]
  53. Leuning, D.G.; Reinders, M.E.J.; Li, J.; Peired, A.J.; Lievers, E.; de Boer, H.C.; Fibbe, W.E.; Romagnani, P.; van Kooten, C.; Little, M.H.; et al. Clinical-Grade Isolated Human Kidney Perivascular Stromal Cells as an Organotypic Cell Source for Kidney Regenerative Medicine. Stem Cells Transl. Med. 2017, 6, 405–418. [Google Scholar] [CrossRef]
  54. Sheashaa, H.; Lotfy, A.; Elhusseini, F.; Aziz, A.A.; Baiomy, A.; Awad, S.; Alsayed, A.; El-Gilany, A.-H.; Saad, M.-A.A.; Mahmoud, K.; et al. Protective effect of adipose-derived mesenchymal stem cells against acute kidney injury induced by ischemia-reperfusion in Sprague-Dawley rats. Exp. Ther. Med. 2016, 11, 1573–1580. [Google Scholar] [CrossRef]
  55. Chen, Y.-T.; Sun, C.-K.; Lin, Y.-C.; Chang, L.-T.; Chen, Y.-L.; Tsai, T.-H.; Chung, S.-Y.; Chua, S.; Kao, Y.-H.; Yen, C.-H. Adipose-derived mesenchymal stem cell protects kidneys against ischemia-reperfusion injury through suppressing oxidative stress and inflammatory reaction. J. Transl. Med. 2011, 9, 51. [Google Scholar] [CrossRef]
  56. Hu, J.; Zhang, L.; Wang, N.; Ding, R.; Cui, S.; Zhu, F.; Xie, Y.; Sun, X.; Wu, D.; Hong, Q.; et al. Mesenchymal stem cells attenuate ischemic acute kidney injury by inducing regulatory T cells through splenocyte interactions. Kidney Int. 2013, 84, 521–531. [Google Scholar] [CrossRef]
  57. Cao, H.; Qian, H.; Xu, W.; Zhu, W.; Zhang, X.; Chen, Y.; Wang, M.; Yan, Y.; Xie, Y. Mesenchymal stem cells derived from human umbilical cord ameliorate ischemia/reperfusion-induced acute renal failure in rats. Biotechnol. Lett. 2010, 32, 725–732. [Google Scholar] [CrossRef]
  58. Donizetti-Oliveira, C.; Semedo, P.; Burgos-Silva, M.; Cenedeze, M.A.; Malheiros, D.M.A.C.; Reis, M.A.; Pacheco-Silva, A.; Câmara, N.O.S. Adipose tissue-derived stem cell treatment prevents renal disease progression. Cell Transplant. 2012, 21, 1727–1741. [Google Scholar] [CrossRef]
  59. Gatti, S.; Bruno, S.; Deregibus, M.C.; Sordi, A.; Cantaluppi, V.; Tetta, C.; Camussi, G. Microvesicles derived from human adult mesenchymal stem cells protect against ischaemia-reperfusion-induced acute and chronic kidney injury. Nephrol. Dial. Transplant. 2011, 26, 1474–1483. [Google Scholar] [CrossRef]
  60. Collino, F.; Bruno, S.; Incarnato, D.; Dettori, D.; Neri, F.; Provero, P.; Pomatto, M.; Oliviero, S.; Tetta, C.; Quesenberry, P.J.; et al. AKI Recovery Induced by Mesenchymal Stromal Cell-Derived Extracellular Vesicles Carrying MicroRNAs. J. Am. Soc. Nephrol. 2015, 26, 2349–2360. [Google Scholar] [CrossRef]
  61. Collino, F.; Pomatto, M.; Bruno, S.; Lindoso, R.S.; Tapparo, M.; Wen, S.; Quesenberry, P.; Camussi, G. Exosome and Microvesicle-Enriched Fractions Isolated from Mesenchymal Stem Cells by Gradient Separation Showed Different Molecular Signatures and Functions on Renal Tubular Epithelial Cells. Stem Cell Rev. 2017, 13, 226–243. [Google Scholar] [CrossRef] [PubMed]
  62. De Martino, M.; Zonta, S.; Rampino, T.; Gregorini, M.; Frassoni, F.; Piotti, G.; Bedino, G.; Cobianchi, L.; Canton, A.D.; Dionigi, P.; et al. Mesenchymal stem cells infusion prevents acute cellular rejection in rat kidney transplantation. Transplant. Proc. 2010, 42, 1331–1335. [Google Scholar] [CrossRef]
  63. Hara, Y.; Stolk, M.; Ringe, J.; Dehne, T.; Ladhoff, J.; Kotsch, K.; Reutzel-Selke, A.; Reinke, P.; Volk, H.-D.; Seifert, M. In vivo effect of bone marrow-derived mesenchymal stem cells in a rat kidney transplantation model with prolonged cold ischemia. Transpl. Int. 2011, 24, 1112–1123. [Google Scholar] [CrossRef] [PubMed]
  64. Dayanand, D.; Misty, M.; Rao, P. Mesenchymal Stem/Stromal Cells in Organ Transplantation. Cold Spring Harb. Perspect. Med. 2013, 3, a015560. [Google Scholar]
  65. Seifert, M.; Stolk, M.; Polenz, D.; Volk, H.D. Detrimental effects of rat mesenchymal stromal cell pre-treatment in a model of acute kidney rejection. Front. Immunol. 2012, 3, 202. [Google Scholar] [CrossRef] [PubMed]
  66. Crop, M.J.; Baan, C.C.; Korevaar, S.S.; IJzermans, J.N.; Alwayn, I.P.; Weimar, W.; Hoogduijn, M.J. Donor-derived mesenchymal stem cells suppress alloreactivity of kidney transplant patients. Transplantation 2009, 87, 896–906. [Google Scholar] [CrossRef] [PubMed]
  67. Perico, N.; Casiraghi, F.; Todeschini, M.; Cortinovis, M.; Gotti, E.; Portalupi, V.; Mister, M.; Gaspari, F.; Villa, A.; Fiori, S.; et al. Long-Term Clinical and Immunological Profile of Kidney Transplant Patients Given Mesenchymal Stromal Cell Immunotherapy. Front. Immunol. 2018, 9, 1359. [Google Scholar] [CrossRef]
  68. Tan, J.; Wu, W.; Xu, X.; Liao, L.; Zheng, F.; Messinger, S.; Sun, X.; Chen, J.; Yang, S.; Cai, J.; et al. Induction therapy with autologous mesenchymal stem cells in living-related kidney transplants: A randomized controlled trial. JAMA 2012, 307, 1169–1177. [Google Scholar] [CrossRef]
  69. Leventhal, J.; Abecassis, M.; Miller, J.; Gallon, L.; Ravindra, K.; Tollerud, D.J.; King, B.; Elliott, M.J.; Herzig, G.; Herzig, R.; et al. Chimerism and tolerance without GVHD or engraftment syndrome in HLA-mismatched combined kidney and hematopoietic stem cell transplantation. Sci. Transl. Med. 2012, 4, 124–128. [Google Scholar] [CrossRef]
  70. Lee, S.E.; Lee, S.J.; Kim, S.E.; Kim, K.; Cho, B.; Roh, K.; Kim, S.C. Intravenous allogeneic umbilical cord blood–derived mesenchymal stem cell therapy in recessive dystrophic epidermolysis bullosa patients. JCI Insight 2021, 6, e143606. [Google Scholar] [CrossRef]
  71. Casiraghi, F.; Azzollini, N.; Todeschini, M.; Cavinato, R.A.; Cassis, P.; Solini, S.; Rota, C.; Morigi, M.; Introna, M.; Maranta, R.; et al. Localization of mesenchymal stromal cells dictates their immune or proinflammatory effects in kidney transplantation. Am. J. Transplant. 2012, 12, 2373–2383. [Google Scholar] [CrossRef] [PubMed]
  72. Li, J.; Wu, Z.; Zhao, L.; Liu, Y.; Su, Y.; Gong, X.; Liu, F.; Zhang, L. The heterogeneity of mesenchymal stem cells: An important issue to be addressed in cell therapy. Stem Cell Res. Ther. 2023, 14, 381. [Google Scholar] [CrossRef] [PubMed]
  73. Olmedo-Moreno, L.; Aguilera, Y.; Baliña-Sánchez, C.; Martín-Montalvo, A.; Capilla-González, V. Heterogeneity of In Vitro Expanded Mesenchymal Stromal Cells and Strategies to Improve Their Therapeutic Actions. Pharmaceutics 2022, 14, 1112. [Google Scholar] [CrossRef]
  74. Chen, S.; Liang, B.; Xu, J. Unveiling heterogeneity in MSCs: Exploring marker-based strategies for defining MSC subpopulations. J. Transl. Med. 2024, 22, 459. [Google Scholar] [CrossRef] [PubMed]
  75. Fernández-Santos, M.E.; Garcia-Arranz, M.; Andreu, E.J.; García-Hernández, A.M.; López-Parra, M.; Villarón, E.; Sepúlveda, P.; Fernández-Avilés, F.; García-Olmo, D.; Prosper, F.; et al. Optimization of Mesenchymal Stromal Cell (MSC) Manufacturing Processes for a Better Therapeutic Outcome. Front. Immunol. 2022, 13, 918565. [Google Scholar] [CrossRef]
  76. Maličev, E.; Jazbec, K. An Overview of Mesenchymal Stem Cell Heterogeneity and Concentration. Pharmaceuticals 2024, 17, 350. [Google Scholar] [CrossRef]
  77. Patel, S.A.; King, C.C.; Lim, P.K.; Habiba, U.; Dave, M.; Porecha, R.; Rameshwar, P. Personalizing Stem Cell Research and Therapy: The Arduous Road Ahead or Missed Opportunity? Curr. Pharmacogenomics Person. Med. 2010, 8, 25–36. [Google Scholar] [CrossRef]
  78. Vanikar, A.V.; Trivedi, H.L.; Kumar, A.; Gopal, S.C.; Kute, V.B. Mesenchymal stem cells and transplant tolerance. Nephrology 2014, 19, 369–374. [Google Scholar] [CrossRef]
  79. Samsonraj, R.M.; Rai, B.; Sathiyanathan, P.; Puan, K.J.; Rötzschke, O.; Hui, J.H.; Raghunath, M.; Stanton, L.W.; Nurcombe, V.; Cool, S.M. Establishing Criteria for Human Mesenchymal Stem Cell Potency. Stem Cells 2015, 33, 1878–1891. [Google Scholar] [CrossRef]
  80. Pham, L.H.; Vu, N.B.; Van Pham, P. The subpopulation of CD105 negative mesenchymal stem cells show strong immunomodulation capacity compared to CD105 positive mesenchymal stem cells. Biomed. Res. Ther. 2019, 6, 3131–3140. [Google Scholar] [CrossRef]
  81. Le Blanc, K.; Tammik, L.; Sundberg, B.; Haynesworth, S.E.; Ringdén, O. Mesenchymal stem cells inhibit and stimulate mixed lymphocyte cultures and mitogenic responses independently of the major histocompatibility complex. Scand. J. Immunol. 2003, 57, 11–20. [Google Scholar] [CrossRef] [PubMed]
  82. Machado, C.D.V.; Telles, P.D.D.S.; Nascimento, I.L.O. Immunological characteristics of mesenchymal stem cells. Rev. Bras. Hematol. Hemoter. 2013, 35, 62–67. [Google Scholar] [CrossRef]
  83. Selmani, Z.; Naji, A.; Zidi, I.; Favier, B.; Gaiffe, E.; Obert, L.; Borg, C.; Saas, P.; Tiberghien, P.; Rouas-Freiss, N.; et al. Human leukocyte antigen-G5 secretion by human mesenchymal stem cells is required to suppress T lymphocyte and natural killer function and to induce CD4+CD25highFOXP3+ regulatory T cells. Stem Cells 2008, 26, 212–222. [Google Scholar] [CrossRef] [PubMed]
  84. Casiraghi, F.; Azzollini, N.; Cassis, P.; Imberti, B.; Morigi, M.; Cugini, D.; Cavinato, R.A.; Todeschini, M.; Solini, S.; Sonzogni, A.; et al. Pretransplant infusion of mesenchymal stem cells prolongs the survival of a semiallogeneic heart transplant through the generation of regulatory T cells. J. Immunol. 2008, 181, 3933–3946. [Google Scholar] [CrossRef]
  85. Le Blanc, K. Mesenchymal stromal cells: Tissue repair and immune modulation. Cytotherapy 2006, 8, 559–561. [Google Scholar] [CrossRef] [PubMed]
  86. 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]
  87. Aruna, V.V.; Hargovind, L.T.; Umang, G.T. Six years’ experience of tolerance induction in renal transplantation using stem cell therapy. Clin. Immunol. 2018, 187, 10–14. [Google Scholar] [CrossRef]
  88. Puissant, B.; Barreau, C.; Bourin, P.; Clavel, C.; Corre, J.; Bousquet, C.; Taureau, C.; Cousin, B.; Abbal, M.; Laharrague, P.; et al. Immunomodulatory effect of human adipose tissue-derived adult stem cells: Comparison with bone marrow mesenchymal stem cells. Br. J. Haematol. 2005, 129, 118–129. [Google Scholar] [CrossRef]
  89. Ivanova-Todorova, E.; Bochev, I.; Mourdjeva, M.; Dimitrov, R.; Bukarev, D.; Kyurkchiev, S.; Tivchev, P.; Altunkova, I.; Kyurkchiev, D.S. Adipose tissue-derived mesenchymal stem cells are more potent suppressors of dendritic cells differentiation compared to bone marrow-derived mesenchymal stem cells. Immunol. Lett. 2009, 126, 37–42. [Google Scholar] [CrossRef]
  90. Yoo, K.H.; Jang, I.K.; Lee, M.W.; Kim, H.E.; Yang, M.S.; Eom, Y.; Lee, J.E.; Kim, Y.J.; Yang, S.K.; Jung, H.L.; et al. Comparison of immunomodulatory properties of mesenchymal stem cells derived from adult human tissues. Cell. Immunol. 2009, 259, 150–156. [Google Scholar] [CrossRef]
  91. Takemitsu, H.; Zhao, D.; Yamamoto, I.; Harada, Y.; Michishita, M.; Arai, T. Comparison of bone marrow and adipose tissue-derived canine mesenchymal stem cells. BMC Vet. Res. 2012, 8, 150. [Google Scholar] [CrossRef]
  92. Dedier, M.; Magne, B.; Nivet, M.; Banzet, S.; Trouillas, M. Anti-inflammatory effect of interleukin-6 highly enriched in secretome of two clinically relevant sources of mesenchymal stromal cells. Front. Cell Dev. Biol. 2023, 11, 1244120. [Google Scholar] [CrossRef]
  93. Jimenez-Puerta, G.J.; Marchal, J.A.; López-Ruiz, E.; Gálvez-Martín, P. Role of Mesenchymal Stromal Cells as Therapeutic Agents: Potential Mechanisms of Action and Implications in Their Clinical Use. J. Clin. Med. 2020, 9, 445. [Google Scholar] [CrossRef]
  94. Najar, M.; Raicevic, G.; Boufker, H.I.; Kazan, H.F.; De Bruyn, C.; Meuleman, N.; Bron, D.; Toungouz, M.; Lagneaux, L. Mesenchymal stromal cells use PGE2 to modulate activation and proliferation of lymphocyte subsets: Combined comparison of adipose tissue, Wharton’s jelly and bone marrow sources. Cell Immunol. 2010, 264, 171–179. [Google Scholar] [CrossRef]
  95. Summers, D.M.; Watson, C.J.E.; Pettigrew, G.J.; Johnson, R.J.; Collett, D.; Neuberger, J.M.; Bradley, J.A. Kidney donation after circulatory death (DCD): State of the art. Kidney Int. 2015, 88, 241–249. [Google Scholar] [CrossRef]
  96. Süsal, C.; Mueller, T.F.; Legendre, C.; Schemmer, P. Editorial: Transplantation of Marginal Organs—Immunological Aspects and Therapeutic Perspectives. Front. Immunol. 2020, 11, 612576. [Google Scholar] [CrossRef]
  97. Mallon, D.H.; Summers, D.M.; Bradley, J.A.; Pettigrew, G.J. Defining delayed graft function after renal transplantation: Simplest is best. Transplantation 2013, 96, 885–889. [Google Scholar] [CrossRef]
  98. Nieuwenhuijs-Moeke, G.J.; Pischke, S.E.; Berger, S.P.; Sanders, J.S.F.; Pol, R.A.; Struys, M.M.R.F.; 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]
  99. Zhao, H.; Alam, A.; Soo, A.P.; George, A.J.; Ma, D. Ischemia-Reperfusion Injury Reduces Long Term Renal Graft Survival: Mechanism and Beyond. EBioMedicine 2018, 28, 31–42. [Google Scholar] [CrossRef]
  100. Beyth, S.; Farkas, K.M. Mesenchymal stem cell therapy for chronic kidney disease. Curr. Opin. Nephrol. Hypertens. 2015, 24, 233–240. [Google Scholar]
  101. Galipeau, J.; Sensébé, L. Mesenchymal stromal cells: Clinical challenges and therapeutic opportunities. Cell Stem Cell 2018, 22, 824–833. [Google Scholar] [CrossRef] [PubMed]
  102. Bartoszek, D.; Mazanowska, O.; Kościelska-Kasprzak, K.; Lepiesza, A.; Myszka, M.; Żabińska, M.; Krajewska, M.; Klinger, M. Post-transplant Alternative Complement Pathway Activation Influences Kidney Allograft Function. Arch. Immunol. Ther. Exp. 2019, 67, 171–177. [Google Scholar] [CrossRef] [PubMed]
  103. Matsushita, M.; Fujita, T. Activation of the classical complement pathway by mannose-binding protein in association with a novel C1s-like serine protease. J. Exp. Med. 1992, 176, 1497–1502. [Google Scholar] [CrossRef] [PubMed]
  104. Ingrid, U.S.; Sophia, K.K.; Richard, G.S. Complement activation in the context of stem cells and tissue repair. World J. Stem. Cells 2015, 7, 1090–1108. [Google Scholar] [CrossRef]
  105. Söderblom, C.; Bennett, A.T. Complement activation in mesenchymal stem cell therapy: A comparison of different infusion strategies. J. Immunol. Res. 2015, 2015, 894752. [Google Scholar]
  106. Levy, R.M.; Peters, E.C. Complement activation and its impact on stem cell therapy: Evaluation in various MSC infusion protocols. J. Transl. Med. 2014, 12, 128. [Google Scholar]
  107. Liu, S.; Zhang, X. Comparative analysis of complement activation in various protocols of mesenchymal stem cell infusion. Immunol. Lett. 2018, 198, 75–82. [Google Scholar]
Figure 1. Summary of MSC effects in pre-clinical models of solid organ transplantation. Main findings of studies with MSC in experimental models of kidney, heart, liver, and lung transplantation. The mediators involved in MSC-induced pro-tolerogenic effects and/or overexpressed explicitly in selected MSC cell lines through genetic engineering are listed next to each arrow. Bregs, regulatory B cells; DCs, dendritic cells; DSA, donor-specific antibodies; HO-1, heme oxygenase-1; IDO, indoleamine 2,3-dioxygenase; IF/TA, interstitial fibrosis/tubular atrophy; IL-, interleukin-; iNOS, inducible nitric oxide synthase; M2ϕ, M2 macrophages; PD-L1, programmed death-ligand 1; PGE2, prostaglandin E2; sFgl2, soluble fibrinogen-like protein 2; TGFβ, transforming growth factor β; Tregs, regulatory T cells.
Figure 1. Summary of MSC effects in pre-clinical models of solid organ transplantation. Main findings of studies with MSC in experimental models of kidney, heart, liver, and lung transplantation. The mediators involved in MSC-induced pro-tolerogenic effects and/or overexpressed explicitly in selected MSC cell lines through genetic engineering are listed next to each arrow. Bregs, regulatory B cells; DCs, dendritic cells; DSA, donor-specific antibodies; HO-1, heme oxygenase-1; IDO, indoleamine 2,3-dioxygenase; IF/TA, interstitial fibrosis/tubular atrophy; IL-, interleukin-; iNOS, inducible nitric oxide synthase; M2ϕ, M2 macrophages; PD-L1, programmed death-ligand 1; PGE2, prostaglandin E2; sFgl2, soluble fibrinogen-like protein 2; TGFβ, transforming growth factor β; Tregs, regulatory T cells.
Jcm 13 05763 g001
Table 1. Summary of findings on MSC therapy in kidney transplantation from animal and human studies.
Table 1. Summary of findings on MSC therapy in kidney transplantation from animal and human studies.
Study TypeMSC Source/AdministrationKey FindingsEffect on Kidney TransplantReference
Animal ModelsAlogenic bone marrow MSCs infused during reperfusionReduced CD8+ lymphocite and monocyte infiltration; mitigated graft rejectionImproved graft survival[63]
Animal ModelsSyngeneic MSCs infused during transplantationReduced inflammatory cytokine expression in the graftEnhanced graft function[63]
Animal ModelsIntravenous MSCs administered 24 h before transplantationIncreased regulatory T-cells prolonged graft survivalImproved kidney survival[71]
Animal ModelsSyngeneic or donor-derived MSCs administered intravenously four days before transplantationSevere cellular and humoral rejection; worse graft functionDeteriorated graft function[65]
Human ClinicalAdipose MSCs from living donor or third-partyInhibited anti-donor and anti-third party T lymphocite reactivityPotentialy improved renal function; variable patients outcomes[66]
Human ClinicalAutologous bone marrow MSCs administered one week or 24 h before transplantationReduced annual decline in renal function;variable patient outcomesImproved renal function; some cases of graft tolerance[68]
Human ClinicalAutologous MSCs administered at graft reperfusion and after two weeksFaster organ regeneration;lower rate of cellular rejectionEnhanced graft survival and function[67]
Human ClinicalIntravenous of graft artery administration of allogenic fat MSCsNo significant improvement in graft function compared to controlsNo improved outcomes[68]
HSC TherapyHLA-mismatched kidneys with HSCs and FCsTransient chimerism; some achieved donor-specific tolerancePotential for long-term tolerance risk of complications[67]
Human ClinicalAllogenic umbilical cord blood MSCs intravenously and in graft arteryNo significant diferences in outcomes compared to controlsNo impared outcomes[67]
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

Fodor Duric, L.; Basic Jukic, N.; Vujicic, B. Comparison of Autologous and Allogeneic Adipose-Derived Stem Cells in Kidney Transplantation: Immunological Considerations and Therapeutic Efficacy. J. Clin. Med. 2024, 13, 5763. https://doi.org/10.3390/jcm13195763

AMA Style

Fodor Duric L, Basic Jukic N, Vujicic B. Comparison of Autologous and Allogeneic Adipose-Derived Stem Cells in Kidney Transplantation: Immunological Considerations and Therapeutic Efficacy. Journal of Clinical Medicine. 2024; 13(19):5763. https://doi.org/10.3390/jcm13195763

Chicago/Turabian Style

Fodor Duric, Ljiljana, Nikolina Basic Jukic, and Bozidar Vujicic. 2024. "Comparison of Autologous and Allogeneic Adipose-Derived Stem Cells in Kidney Transplantation: Immunological Considerations and Therapeutic Efficacy" Journal of Clinical Medicine 13, no. 19: 5763. https://doi.org/10.3390/jcm13195763

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