Next Article in Journal
Bone Defect Treatment in Regenerative Medicine: Exploring Natural and Synthetic Bone Substitutes
Previous Article in Journal
Can Humanized Immune System Mouse and Rat Models Accelerate the Development of Cytomegalovirus-Based Vaccines Against Infectious Diseases and Cancers?
Previous Article in Special Issue
Ingenol-3-Angelate Enhances the B Cell Inhibitory Potential of Mesenchymal Stem Cells, Leading to Marked Alleviation of Lupus Symptoms in MRL.faslpr Mice
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Therapeutic Use and Potential of MSCs: Advances in Regenerative Medicine

by
Alin Constantin Pînzariu
1,†,
Roxana Moscalu
2,*,
Radu Petru Soroceanu
3,*,
Minela Aida Maranduca
1,†,
Ilie Cristian Drochioi
4,
Vlad Ionut Vlasceanu
3,
Sergiu Timofeiov
3,
Daniel Vasile Timofte
3,
Bogdan Huzum
5,
Mihaela Moscalu
6,
Dragomir Nicolae Serban
1 and
Ionela Lacramioara Serban
1
1
Department of Morpho-Functional Sciences II, Discipline of Physiology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
2
Division of Cell Matrix Biology & Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PL, UK
3
Department of Surgery I, Discipline of Surgical Semiology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
4
Department of Oral and Maxillo Facial Surgery, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
5
Department of Orthopaedic and Traumatology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
6
Department of Preventive Medicine and Interdisciplinarity, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2025, 26(7), 3084; https://doi.org/10.3390/ijms26073084
Submission received: 10 March 2025 / Revised: 23 March 2025 / Accepted: 25 March 2025 / Published: 27 March 2025
(This article belongs to the Special Issue Latest Research on Mesenchymal Stem Cells)

Abstract

:
Mesenchymal stem cells (MSCs) have emerged as a relevant strategy in regenerative medicine due to their multipotent differentiation capacity, immunomodulatory properties, and therapeutic applications in various medical fields. This review explores the therapeutic use of MSCs, focusing on their role in treating autoimmune disorders and neoplastic diseases and in tissue regeneration. We discuss the mechanisms underlying MSC-mediated tissue repair, including their paracrine activity, migration to injury sites, and interaction with the immune system. Advances in cellular therapies such as genome engineering and MSC-derived exosome treatments further enhance their applicability. Key methodologies analyzed include genomic studies, next-generation sequencing (NGS), and bioinformatics approaches to optimize MSC-based interventions. Additionally, we reviewed preclinical and clinical evidence demonstrating the therapeutic potential of MSCs in conditions such as graft-versus-host disease, osteoarthritis, liver cirrhosis, and neurodegenerative disorders. While promising, challenges remain regarding standardization, long-term safety, and potential tumorigenic risks associated with MSC therapy. Future research should focus on refining MSC-based treatments to enhance efficacy and minimize risks. This review underscores the need for large-scale clinical trials to validate MSC-based interventions and fully harness their therapeutic potential.

1. Introduction

Mesenchymal stem cells (MSCs) have garnered increasing interest due to their remarkable applicability in a vast range of clinical challenges, from their key role in supporting tissue regeneration and wound healing to more immunomodulatory effects [1]. MSCs are multipotent stem cells able to self-renew and easily proliferate and can be widely derived from bone marrow, adipose tissue, placentas, umbilical cords, skin, connective muscle tissue, the liver, and the spleen, amongst other sources [2]. The inherent ability of MSCs to undergo differentiation into a wide array of cell lineages including osteoblasts and chondrocytes, adipocytes, and neuronal or muscle cells [3,4,5], and their accessible in vitro expansion and low ethical impact [2], have attracted considerable attention for their effective utilization in the realm of therapeutic strategies for various human diseases (Figure 1).
The literature has described their role in tissue regeneration through their unique ability to migrate to the site of injury, their paracrine potential, their ability to release growth factors and cytokines [6], and their low immunogenic effects, allowing for easier allogeneic transplants that do not require immunosuppressive therapies [2]. Upon transplantation, they acquire immunosuppressive properties, adapt to the structure of the transplanted tissue, and subsequently advance into progenitor cells within the microenvironment, functioning both as stem cells and their progeny [7]. They possess natural abilities to detect changes in their environment, such as the appearance of inflammation. Under such conditions, they can induce the release of bioactive agents and form progenitor cells in response to these changes [8]. Additionally, it has been demonstrated that MSCs migrate to sites of inflammation far from their injection site [9,10].
As research into regenerative medicine and cellular therapies continues to progress, the use of MSCs is becoming increasingly recognized for its valuable therapeutic properties [9]. In most cases, these properties have a cumulative effect on the successful achievement of the desired biological effects of MSCs. However, their exact roles in mediating therapeutic actions are not yet fully understood [11].
In the clinical field, the therapeutic effects of MSCs are currently under investigation in clinical trials involving patients with diverse medical conditions. These conditions include orthopedic injuries, acute graft rejection following bone marrow transplantation, cardiovascular diseases, autoimmune disorders, and liver diseases. Moreover, the genetic engineering of MSCs to enhance the expression of antitumor genes has opened up promising perspectives for their potential clinical application as an antineoplastic therapy [11].
The first clinical study using MSCs developed ex vivo was conducted in 1995 by Lazarus et al. [12]. During this Phase I feasibility study, 15 patients with hematological malignancies successfully received autologous MSC injections without any toxic adverse effects [11,12]. Subsequent to this initial study, many clinical investigations have been undertaken to assess the viability and effectiveness of MSC therapy across diverse pathological conditions. These studies primarily fall within Phase I, which evaluates safety; Phase II, focused on establishing proof of concept and efficacy in human subjects; or a combination of both (Phase I/II). Only a limited proportion of these clinical trials have advanced to Phase III, where newer treatments are compared to standard therapies or the most established methods of therapy or involve a combination of Phase II/III studies [11].
In general, MSCs exhibit a favorable safety profile, with most investigations indicating a lack of medium-term adverse effects, as evidenced by the majority of investigations indicating the absence of significant medium-term adverse reactions. Nevertheless, a few studies have reported the occurrence of mild and transient peri-injection effects, which are generally short-lived and not of substantial concern [8]. Furthermore, many completed clinical studies have demonstrated the effectiveness of MSC infusions for various other diseases, such as acute myocardial ischemia, stroke, liver cirrhosis, amyotrophic lateral sclerosis, and osteoarticular diseases [11].
In the present review, we will discuss the diverse successful utilizations of MSCs to address a wide variety of common important global clinical challenges, including solid organ transplant rejection and graft-versus-host disease; bone and cartilage disease, including osteoarthritis and osteogenesis imperfect; liver diseases; autoimmune conditions, with emphasis on systemic lupus erythematosus, multiple sclerosis, and rheumatoid arthritis; and neoplastic diseases. We will also investigate the immunomodulatory mechanisms of MSCs that improve graft survival and reduce inflammation, promoting tissue regeneration in conditions such as osteoarthritis or cirrhosis, and examine developing targeted therapies for cancer that use the ability of MSCs to deliver anticancer agents. This review highlights the versatility of MSC therapies and the potential for further development of therapeutic strategies that can benefit from their vast availability, ease of culturing ex vivo, and safe integration, whether in vivo or in vitro.

Highlights from the Specialized Literature Regarding the Therapeutic Use and Potential of MSCs

The literature search was conducted in the PubMed database (accessed on 1 February 2025), focusing on studies published within the last 10 years. Our search queried “mesenchymal stem cells [AND] regenerative medicine [OR] tissue regeneration [OR]; immunomodulation [AND] stem cell therapy” and was limited only to prospective and retrospective studies and metaanalyses, omitting abstracts, documents, and reviews. In this way, our search resulted in 48 references within our scope of interest, covering the period from 2015 to 2025, of which 20 articles were identified in the last five years. Additionally, the bibliographic references of the identified studies were examined, and specific observations relevant to the objectives of our study were integrated. Significant viewpoints consistent with our research are included in the discussion section, leading to the referencing of articles prior to 2015 when deemed relevant.

2. Therapeutic Use of MSCs in Acute Graft Rejection

The successful use of MSC therapy for a severe case of acute graft rejection refractory to steroid treatment in a nine-year-old leukemia patient following allogeneic hematopoietic stem cell transplantation (HSCT) has sparked increased interest in the research world [13,14]. This study led the way for further clinical trials and preclinical studies that thoroughly investigated the potential of MSCs not only in graft-versus-host disease (GVHD)-related HSCT but also for the treatment and prevention of solid organ transplant rejection [14]. Linked with significant morbidity and mortality rates, the current standard-of-care treatment for acute transplant rejection is immunosuppression using corticosteroids [15]. However, this therapy has demonstrated effectiveness only in a small subset of patients, leaving room for further improvement in treatment outcomes.
A follow-up study by Fang et al. [16] tested adipose tissue-derived MSCs in six patients with severe grade III-IV acute GVHD that did not respond to steroids. They found that five out of six—around 83%—had their GVHD resolve, with the intestine, liver, and skin completely healing in those cases. Monitoring these patients later showed they lived longer, anywhere from a few months to three years for some, though survival varied greatly from one person to the next [10,16,17,18,19].
The mechanism of action of MSC therapy in GVHD or solid organ transplantation rejection has been widely linked with their ability to inhibit the activity of effector T cells, which drive rejection by causing excessive transplanted tissue damage [20], and instead modulate the increase in anti-inflammatory regulatory T cells (Treg), which suppress the immune system, thus improving graft survival. This was also demonstrated in vivo in a murine semiallogeneic heart transplant model, which highlighted the immunosuppressive potential of MSC therapy by inducing donor-specific CD4+CD25+Foxp3+ Treg cells, key factors in preventing autoimmune disease and maintaining immune tolerance [21]. These effects are largely due to the paracrine activity of MSCs, secreting factors such as transforming growth factor beta (TGF-β), nitric oxide (NO)—which modulates to prevent T-cell division by interfering with their signals, prostaglandin E2 (PGE2)—which facilitates attenuation by cutting pro-inflammatory cytokines and directing macrophages to a subdued M2 type, or indoleamine 2,3-dioxygenase (IDO) as a response to the GVHD-related elevation of pro-inflammatory cytokines including interferon gamma (IFN-γ), tumor necrosis factor alpha (TNF-α), or interleukin 1β (IL-1β) [14]. The anti-inflammatory effects of MSCs have also been identified through their role in supporting the change in polarization of macrophages, from the pro-inflammatory M1 phenotype to a pro-healing M2 phenotype [22], further supporting a dampening of the inflammatory response and promoting tissue repair and graft survival.
MSC therapy has also shown promising results in improving graft survival and reducing the rate of transplant rejection in a number of other tissue transplants, including liver, kidney, cornea, and skin [14,23,24,25,26], and multiple clinical trials are underway to further address, test, and demonstrate the successful immunomodulatory effects of MSCs [14].

3. Therapeutic Use of MSCs in Bone and Cartilage Diseases

The multipotent capacity of MSCs and, specifically, their ease of differentiation into osteoblasts (bone-forming cells), tenocytes (tendon cells), and chondrocytes (cartilage cells) has garnered significant attention for their potential application in orthopedic conditions. MSCs have been used in the development of regenerative therapy strategies aimed at treating orthopedic injuries and promoting tissue repair, as well as regeneration in the musculoskeletal system [27,28,29].
MSCs have demonstrated a beneficial role in the treatment of bone system diseases, particularly conditions like osteogenesis imperfecta and hypophosphatasia. Osteogenesis imperfecta (OI) is a genetic disorder characterized by increased skeletal fragility and alterations in connective tissue due to the deficient production of type I collagen by osteoblasts [11]. Also, a genetic disorder, hypophosphatasia, is the result of a loss of function mutation in the alkaline phosphatase gene that encodes the activity of tissue-nonspecific alkaline phosphatase (TNSALP) [30]. These diseases are characterized by abnormalities in bone formation and mineralization, leading to brittle bones and skeletal deformities. MSCs differentiate into osteoblasts under the influence of signaling molecules such as bone morphogenetic proteins (BMPs) and Runx2, a transcription factor that initiates the whole bone-making process. Once they differentiate, they produce type I collagen to reinforce the structural deficiencies in OI’s bone setup and enhance alkaline phosphatase levels to address the impaired mineralization in hypophosphatasia [28,29,30]. Apart from the anti-inflammatory and immunomodulatory advantages brought about by MSC therapy, these cells have also been shown to contribute to bone regeneration by being able to migrate to the site of a bone injury due to specialized chemokine receptor activity [31], promote osteoblast differentiation, and localize new blood vessel formation, which is essential for bone repair [31,32,33].
A series of clinical studies focused on pediatric patients with OI who underwent allogeneic hematopoietic stem cell transplants has shown promising outcomes. The transplanted bone marrow cells successfully integrated into the recipient’s body and gave rise to functional osteoblasts, contributing to the enhancement of bone structure and overall function [34,35,36,37,38]. These findings offer encouraging prospects for the potential therapeutic use of hematopoietic stem cell transplantation in managing OI and promoting bone health in affected pediatric patients.
MSCs have also shown great therapeutic potential in the treatment of osteoarthritis (OA). OA has a high incidence worldwide, with a high socioeconomic and psychological impact due to persistent pain and disability that erode patients’ quality of life (QoL) [39,40]. For OA, MSCs undergo chondrogenic differentiation into chondrocytes when transforming growth factor-beta (TGF-β) and Sox9, a transcription factor serving as a pivotal regulator of chondrogenic differentiation and cartilage formation, which directs the production of cartilage matrix components—such as type II collagen and aggrecan—to repair cartilage degradation. They also release anti-inflammatory factors to mitigate the joint’s inflammatory disruption, helping repair it over time [41]. However, there is still a paucity of effective therapeutic strategies beyond the surgical replacement of the affected joint. Advances in MSCs’ regenerative therapies have brought about the successful integration of these cells for OA management. A meta-analysis by Tan et al. [41], comprising nine studies and 440 osteoarthritic knees, concluded that even in the absence of adjuvant therapies, intra-articular MSC injections significantly improved function and joint pain in affected individuals [41,42]. The results were also reflected throughout patient questionnaires, such as the Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC) analyzing pain, stiffness, and knee function, and the Knee Injury and Osteoarthritis Outcome Score (KOOS) questionnaire looking into the impact of knee injuries on patients’ QoL [40,41].
These are just a few examples of the impact that MSC therapies can have in orthopedics, providing effective solutions to ongoing clinical challenges that are likely to increase in the modern population.

4. Therapeutic Use of MSCs in Liver Diseases

While MSCs have been closely studied in the aforementioned clinical contexts, the research to date shows that their use in treating hepatic cirrhosis has remained restricted to a narrowly defined patient group.
In a Phase I clinical trial conducted by Mohamadnejad et al. [42], four patients with decompensated hepatic cirrhosis were administered autologous MSCs intravenously. Throughout the study, no adverse effects were observed in the patients, and notably, their QoL exhibited a considerable improvement by the end of the study’s follow-up phase [43]. A deeper look into the results suggests that the boost in quality of life (QoL) stemmed from better liver performance, particularly marked by increased albumin levels, rather than MSCs turning into hepatocytes or endothelial cells—something this study did not convincingly show [43].
In a separate Phase I-II clinical trial, eight patients diagnosed with end-stage liver disease (comprising four with hepatitis B, one with hepatitis C, one with alcoholic hepatopathy, and two with cryptogenic etiology) were administered injectable autologous MSCs. The research demonstrated that all participants responded well to the therapy, with notable enhancements in liver function being particularly evident. These findings support the notion that MSCs may offer a feasible, safe, and efficient treatment option for individuals with end-stage liver diseases [44]. Through paracrine effects, MSCs seem to mitigate chronic inflammation—a key contributor to these disorders—by secreting anti-inflammatory factors like IL-10 and TGF-β, which reduce excessive immune responses, including overactive macrophages [44].
A systematic review and meta-analysis by Lu et al. [45] identified 11 randomized control clinical trials addressing the use of MSC strategies in liver cirrhosis between 2011 and 2023. These studies showed that MSC infusions were safe, with no presented adverse side effects, and had made a significant positive improvement on albumin levels from two weeks up to six months from infusion compared with the controls. Moreover, an increased three-month survival rate was reported based on the patients’ MELD scores at one, two, and six months post-treatment [45].
However, in the context of a progressive disease with debilitating factors that can significantly impact patients’ QoL, there is a paucity of studies that needs to be addressed before MSCs can routinely be used in liver disease. This mainly revolves around the effects of different sources of MSCs, optimization of dosages and routes of administration, as well as frequency of treatment [45,46].

5. Therapeutic Use of MSCs in Autoimmune Diseases

The literature data highlight attempts to use MSCs in the therapy of autoimmune conditions such as refractory cases of systemic lupus erythematosus (SLE), multiple sclerosis (MS), polymyositis, vasculitis (including Behcet’s disease), rheumatoid arthritis, Crohn’s disease, polychondritis, and encephalomyelitis [47,48,49,50].
Recent in vitro studies have revealed that MSCs possess immunomodulatory characteristics and can exert immunosuppressive effects on various immune cell types. These effects include the inhibition of lymphocyte proliferation, memory, and activated T cells, B cells, natural killer (NK) cells, and dendritic cells. It is important to differentiate this concept from that of autologous hematopoietic stem cell transplantation, as MSC transplantation does not necessitate immunosuppression of patients beforehand. Furthermore, the therapeutic impact of MSCs is considered to occur locally within the inflamed organ, facilitated by the engraftment of MSCs in that particular area. Consequently, the effects of MSC therapy are relatively short-lived, limited to the immediate vicinity of grafting [51,52].
SLE is a systemic chronic autoimmune and inflammatory disorder that can lead to multiorgan injury, including the brain, lungs, and kidneys, as well as the hematopoietic system [53]. Some researchers revealed that the transplantation of bone marrow-derived mesenchymal stem cells improved multi-organ function in genetically modified mice and individuals diagnosed with SLE. The genetically modified mice (MRL/lpr) were found to have a deficiency in the osteoblastic niche, which seems to be partially responsible for the pathogenesis of systemic lupus erythematosus [54,55,56]. Moreover, the allogeneic transplantation of bone marrow-derived MSCs has the capability to rebuild the bone marrow osteoblastic niche and shows enhanced effectiveness in reducing multi-organ dysfunction when compared to cyclophosphamide, the conventional standard therapy. Patients with SLE who received allogenic bone marrow-derived mesenchymal stem cell transplants experienced stable disease remission for a period ranging from 12 to 18 months. Additionally, improvements in symptoms like joint pain and fatigue, alongside decreasing serological markers and better renal function, were seen [57,58]. Other experimental research has demonstrated favorable effects of MSC use in the treatment of multiple sclerosis, but the mechanisms underlying these actions are not fully elucidated, suggesting possible involvement in immunomodulation, neuroprotection, and neuro-regeneration as shown in (Table 1) [59,60,61,62].
The use of MSC strategies for patients with MS, an autoimmune neurodegenerative disease causing localized inflammation, demyelination, and axon degeneration within the central nervous system [63], has also been investigated. A systematic review and meta-analysis by Vaheb et al. [64], updated in January 2024 and covering 30 studies, examined the safety and efficacy of this cell therapy in patients with various types of MS, including relapsing-remitting, primary, and secondary progressive disease. Interestingly, the intrathecal (IT) approach demonstrated significantly higher improvements in patients’ expanded disability status scale (EDSS) when compared with the control, rather than the intravenous (IV) approach of administering the MSCs. This could be due to the limited ability of IV-administered MSCs to cross the blood–brain barrier, which hinders their therapeutic effects [64,65]. MRI imaging has also highlighted that 71.8% of patients had activity-free MRI scans in short-term follow-up and 100% clearance with no Gadolinium-enhancement lesions (GELs) on long-term follow-up scans [64]. This confirms MSCs’ role in plaque remyelination, as repeated exposure through IT MSC infiltrations for a prolonged duration has been shown to promote the secretion of trophic factors, including leukemia inhibitory factor (LIF), insulin-like growth factors (IGF), glial cell-line-derived neutrophic factor (GDNF), and hepatocyte growth factor (HGF), which can potentiate the neural repair in the context of plaque demyelination [64,66,67,68] and also dampen the localized inflammation, thus inhibiting further development of GELs [64,69,70].
Table 1. MSC therapy research.
Table 1. MSC therapy research.
StudyStudy TypeTherapyTargeted
Condition
Mechanism
of Action
Main Findings
Islam et al., 2023 [62]Meta-
analysis
MSC therapy for multiple sclerosisMultiple
Sclerosis
Neuroprotection, reducing relapses and disabilityMeta-analysis confirms MSCs’ potential in reducing relapses and disability in MS patients.
Vaheb et al., 2024 [63]Meta-
analysis
MSCs’ neurological efficacy in MSMultiple
Sclerosis
Enhancing neurological function, reducing progressionUpdated review highlights the safety and efficacy of MSCs in progressive MS therapy.
Terstappen et al., 2021 [64]ReviewTherapeutics across the blood–brain barrierBlood–Brain
Barrier
Improving MSC delivery for CNS repairDiscusses methods to improve MSC delivery to CNS via blood–brain barrier crossing.
Harris et al., 2012 [65]Preclinical studyAutologous MSC-derived neural progenitorsMultiple
Sclerosis
Using MSC-derived neural progenitors for CNS regenerationDemonstrates feasibility of MSC-derived neural progenitors for CNS applications.
Huang and Dreyfus, 2016 [67]ReviewGrowth factors for demyelinating diseasesDemyelinating DiseasesUsing growth factors to support remyelinationGrowth factors like IGF and FGF can support MSC-based therapies in demyelinating diseases.
Petrou et al., 2020 [68]Clinical trialMSC transplantation for progressive MSProgressive
Multiple
Sclerosis
Autologous MSC transplantation for neurological repairClinical benefits observed in MS patients post MSC transplantation, improving neurological function.
Harris et al., 2020 [69]Clinical trialMSC-derived neural progenitors for MSProgressive
Multiple
Sclerosis
Long-term benefits of MSC-derived neural progenitorsPhase I study indicates long-term benefits of MSC-derived progenitors in progressive MS.
Zhang et al., 2019 [71]Preclinical studyDifferent MSCs for rheumatoid arthritisRheumatoid
Arthritis
Comparison of MSC types for immune regulationComparison of different MSC sources shows varying efficacy in RA models.
Hwang et al., 2021 [72]ReviewClinical applications of MSCs in RA and OARheumatoid
Arthritis and Osteoarthritis
Reduction of inflammation and joint damageRecent trials confirm the effectiveness of MSCs in reducing inflammation in RA and OA.
Wang et al., 2013 [11]Clinical trial (Phase II)Umbilical cord MSCs for rheumatoid arthritisRheumatoid
Arthritis
Umbilical cord MSCs modulating immune responseHuman umbilical cord MSCs show good safety and efficacy profile for RA patients.
Park et al., 2018 [73]Clinical trial (Phase I)Intravenous MSCs for RA (Phase 1 trial)Rheumatoid
Arthritis
MSC infusion reducing symptoms in RA patientsPhase 1 clinical trial supports intravenous MSC administration as a viable RA treatment.
Ghoryani et al., 2020 [74]Clinical trialImmunoregulatory effects of MSCs in RARheumatoid
Arthritis
Enhancing regulatory T cells for immune suppressionMSC therapy modulates immune response, increasing regulatory T cells in refractory RA.
Treatment strategies involving MSCs have also been developed for rheumatoid arthritis (RA), an autoimmune condition affecting patients’ joints, muscles, and tendons, as well as connective and fibrous tissue, leading to severe and debilitating joint deformities, dysfunction, and pain [71]. Although a much more limited number of studies have been presented in the specialized literature, the safety and therapeutic effects of MSC administration have been highlighted in in vivo mice models [72]. Clinical trials have also been developed. Wang et al. [14] also confirmed the safe administration of MSCs in patients with RA and disease-modifying anti-rheumatic drugs (DMARDS) and showed improvements in disease activity based on a disease activity score of 28 (DAS28), joint swelling and pain, and overall QoL [14,72]. Another clinical trial by Park et al. [73] also noted a dose-dependent decrease in DAS28 and reported no severe adverse effects following MSC therapy; apart from a mild elevation of serum uric acid levels, no other changes were noted in the hematological or chemical investigations of patients [71,72]. Ghoryani et al. [74] went further, highlighting the immunomodulatory effects of MSC therapy in refractory RA, identifying a post-infiltration elevation of IL-10, TGF-β, and head box P3 (FOXP3), a protein regulating the development and function of Treg cells [72,74].
It is clear that there is still space for the development of MSC-targeted strategies for auto-immune disorders. Previous studies have provided the basis of ensuring safety and efficacy in human applications of MSCs; however, considering the complexity of these disorders and their systemic effects, there is a high need for large, comprehensive studies that can inform the decision-making and guide the adjustment of MSC therapies for the affected population.

6. Therapeutic Use of MSCs in Neoplastic Diseases

Although preclinical models extensively explored the utilization of genetically modified MSCs for treating diverse cancer types such as breast, lung, liver, pancreatic, ovarian, prostate, sarcoma, and glioma, clinical trials implementing MSCs in neoplastic disease therapy have yet to be documented [75]. While no significant adverse events have been observed in their application, the safety of MSC administration remains a current area of concern.
The potential of MSCs to undergo malignant transformation is a significant concern that poses limitations on ensuring their safety in therapeutic applications [76]. The tumor microenvironment plays a crucial role in influencing tumor-cell behavior, creating an atmosphere that can either promote or suppress tumor development. MSCs have demonstrated the ability to modulate the fate of tumor cells, attracting attention to their potential utility in this context. MSCs are capable of integrating into the tumor microenvironment, a region abundant in inflammatory mediators, cytokines, and growth factors. This setting may prompt MSCs to secrete agents that amplify angiogenesis, cellular proliferation, and tumor invasion, thereby potentially aggravating tumor expansion and metastatic spread. For instance, the liberation of vascular endothelial growth factor (VEGF), a key angiogenic promoter, or other proliferative cues could enhance vascular support and drive tumor growth within this context [77].
MSCs have been acknowledged for their dual effects, encompassing both tumorigenic and antitumoral activity. On the one hand, clinical studies have shown promising potential for MSCs in the treatment of human cancer. Notably, MSCs possess intrinsic migratory abilities, which hold promise for targeted drug delivery and therapies aimed at tumor and metastatic cells. Moreover, different manipulation techniques have been explored to achieve the desired expression of anti-angiogenic, anti-proliferative, and pro-apoptotic properties tailored to specific tumor types [78]. For instance, liberating vascular endothelial growth factor (VEGF), a key angiogenic promoter, or other proliferative cues could enhance vascular support and drive tumor growth within this context [75,79].
Tumor angiogenesis, a critical feature of tumor advancement and metastasis, has garnered significant attention as a therapeutic target in cancer treatment since the introduction of the first angiogenesis inhibitor, bevacizumab, for metastatic colorectal cancer therapy. MSCs demonstrate pathotropic migratory attributes, rendering them promising con-tenders for the targeted delivery of antineoplastic agents. Given the advancements in specific anticancer drugs and considering the incorporation and targeted delivery capabilities of MSCs, a novel area of research has emerged aiming to develop efficacious cancer therapies by manipulating these cells [80].
The scientific literature provides evidence that unmodified MSCs exhibit antitumor effects in vitro and in various mouse tumor models. These effects are attributed, in part, to the release of factors by MSCs that possess antitumor functions, leading to reduced proliferation of melanoma, glioma, lung cancer, hepatoma, and breast adenocarcinoma cells [1]. It has been highlighted that bone marrow-derived MSCs injected intravenously into a mouse Kaposi’s sarcoma model colonize tumor sites and strongly inhibit tumor development [81].
Furthermore, experimental observations have demonstrated that MSCs display anti-angiogenic properties in both in vitro investigations and a murine model of melanoma. Moreover, when MSCs are directly injected into murine subcutaneous melanoma, it induces apoptosis and leads to a noteworthy reduction in tumor growth. In separate investigations, MSCs derived from umbilical cord blood have been utilized as untainted cellular entities in therapeutic approaches for treating multiform glioblastoma [7].
Stem cells derived from umbilical cord blood, exhibiting elevated levels of CD44 and CD133, undergo apoptosis upon co-culture with multiform glioblastoma cells. Additionally, the application of umbilical cord blood-derived stem cells in the treatment of glioma cells exerts an inhibitory effect on angiogenesis mediated by focal adhesion kinase (FAK), resulting in the upregulation of homologous phosphatase and tension in glioma cells, as well as the downregulation of Akt and PI3K signaling molecules. Consequently, this inhibits migration and characteristic cellular lesion healing in glioma [7]. Furthermore, favorable outcomes have been achieved by modifying MSCs to overexpress specific target genes and express or secrete molecules such as interleukins, on the one hand, and additionally, interferons, pro-apoptotic proteins, and anti-angiogenic agents on the other hand. This selective modulation exerts beneficial effects, making targeted therapy a viable option for various types of cancer [7].
Only a few clinical trials have been reported on cancer therapy using MSCs. The initial clinical trial (Phase I/II), referred to as TREAT-ME1, demonstrated promising outcomes by employing genetically changed autologous mesenchymal stromal cells as a delivery vector for herpes simplex virus thymidine kinase (HSV-TK) to treat advanced gastrointestinal tumors [81,82].
Two additional clinical trials concentrated on MSC usage in ovarian cancer. The first study (Phase I/II) aimed to determine adverse effects and establish the appropriate dose of MSC transfected with oncolytic measles virus encoding NIS (MV-NIS) while evaluating its effectiveness in patients with ovarian cancer [81]. Another Phase I clinical trial employed human MSCs transfected with IFN-β to evaluate its safety and ascertain a well-tolerated dosage for treating ovarian cancer patients.
In an alternative Phase I clinical study, the safety and potential benefits of allogeneic bone marrow-derived MSCs in patients with prostate cancer were investigated. The investigations showed that MSCs were not efficiently targeted to tumor sites to exert therapeutic effects [82]. These studies were conducted with the aim of assessing the potential of MSCs in treating different types of cancer; however, the results appear to be insufficient and require additional preclinical studies and clinical trials.

7. Conclusions

In conclusion, MSCs hold great potential for regenerative medicine and therapeutic applications, making them effective in treating a wide range of medical conditions, including acute graft rejection, bone and cartilage diseases, liver diseases, autoimmune diseases, and select neoplastic diseases. Their ability to differentiate into various cell types, secrete bioactive molecules, and exhibit immunomodulatory properties makes them effective in treating a wide range of medical conditions, thereby facilitating tissue repair and improving damaged tissue functions. However, the research also underscores the importance of further investigations to establish comprehensive safety and efficacy profiles and to refine treatment methodologies. If, for some areas such as cardiovascular diseases, orthopedic diseases, or transplant rejection and graft-versus-host disease, the literature is more extensive and offers vast alternatives of treatment strategies for both the pediatric and adult populations, other areas, such as autoimmune disorders, neoplastic or solid organ diseases (i.e., liver, kidney, lung, etc.), still present a paucity of information regarding the efficacy and best administration strategies for achieving the desired results. As cellular therapies continue to advance in the field of regenerative medicine, MSCs remain a leading contender, offering hope for improved treatments and better patient outcomes in some of the biggest clinical challenges worldwide.

Author Contributions

Conceptualization, A.C.P., R.M., R.P.S., M.M., D.N.S. and I.L.S.; methodology, A.C.P., R.M., M.A.M., I.C.D., D.V.T., B.H. and D.N.S.; software, M.M. and S.T.; validation, A.C.P., R.M., M.M., D.N.S. and I.L.S.; formal analysis, M.A.M., I.C.D., V.I.V., S.T., D.V.T. and B.H.; resources, A.C.P., R.M., V.I.V., S.T. and B.H.; writing—original draft preparation, R.M., M.M., R.P.S. and I.C.D.; writing—review and editing, R.M., M.M., R.P.S., M.A.M., V.I.V., S.T., D.V.T., B.H. and D.N.S.; visualization, R.M. and R.P.S.; supervision A.C.P., M.M. and I.L.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Merimi, M.; El-Majzoub, R.; Lagneaux, L.; Moussa Agha, D.; Bouhtit, F.; Meuleman, N.; Fahmi, H.; Lewalle, P.; Fayyad-Kazan, M.; Najar, M. The Therapeutic Potential of Mesenchymal Stromal Cells for Regenerative Medicine: Current Knowledge and Future Understandings. Front. Cell Dev. Biol. 2021, 9, 661532. [Google Scholar] [CrossRef]
  2. Hassanzadeh, A.; Rahman, H.S.; Markov, A.; Endjun, J.J.; Zekiy, A.O.; Chartrand, M.S.; Beheshtkhoo, N.; Kouhbanani, M.A.J.; Marofi, F.; Nikoo, M.; et al. Mesenchymal Stem/Stromal Cell-Derived Exosomes in Regenerative Medicine and Cancer; Overview of Development, Challenges, and Opportunities. Stem Cell Res. Ther. 2021, 12, 297. [Google Scholar] [CrossRef]
  3. Haliga, R.; Zugun-Eloae, F.; Oboroceanu, T.; Pînzariu, A.; Mocanu, V. Vitamin D and tissular expression of vitamin D receptor in obesity. Rev. Med. Chir. Soc. Med. Nat. Iasi 2016, 120, 404–408. [Google Scholar] [PubMed]
  4. Hristov, I.; Mocanu, V.; Zugun-Eloae, F.; Labusca, L.; Cretu-Silivestru, I.; Oboroceanu, T.; Tiron, C.; Tiron, A.; Burlacu, A.; Pinzariu, A.C.; et al. Association of Intracellular Lipid Accumulation in Subcutaneous Adipocyte Precursors and Plasma Adipokines in Bariatric Surgery Candidates. Lipids Health Dis. 2019, 18, 141. [Google Scholar] [CrossRef] [PubMed]
  5. Pinzariu, A.C.; Oboroceanu, T.; Eloae, F.Z.; Hristov, I.; Costan, V.V.; Labusca, L.; Cianga, P.; Verestiuc, L.; Hanganu, B.; Crauciuc, D.V.; et al. Vitamin D as a Regulator of Adipocyte Differentiation Effects in Vivo and in Vitro. Rev. Chim. 2018, 69, 731–734. [Google Scholar] [CrossRef]
  6. Maldonado, V.V.; Patel, N.H.; Smith, E.E.; Barnes, C.L.; Gustafson, M.P.; Rao, R.R.; Samsonraj, R.M. Clinical Utility of Mesenchymal Stem/Stromal Cells in Regenerative Medicine and Cellular Therapy. J. Biol. Eng. 2023, 17, 44. [Google Scholar] [CrossRef]
  7. Javan, M.R.; Khosrojerdi, A.; Moazzeni, S.M. New Insights Into Implementation of Mesenchymal Stem Cells in Cancer Therapy: Prospects for Anti-Angiogenesis Treatment. Front. Oncol. 2019, 9, 840. [Google Scholar] [CrossRef]
  8. Otto, W.R.; Wright, N.A. Mesenchymal Stem Cells: From Experiment to Clinic. Fibrogenesis Tissue Repair. 2011, 4, 20. [Google Scholar] [CrossRef]
  9. Zhang, Q.; Zhou, S.N.; Fu, J.M.; Chen, L.J.; Fang, Y.X.; Xu, Z.Y. Interferon-γ Priming Enhances the Therapeutic Effects of Menstrual Blood-Derived Stromal Cells in a Mouse Liver Ischemia-Reperfusion Model. World J. Stem Cells 2023, 15, 876–896. [Google Scholar] [CrossRef]
  10. Penack, O.; Marchetti, M.; Ruutu, T.; Aljurf, M.; Bacigalupo, A.; Bonifazi, F.; Ciceri, F.; Cornelissen, J.; Malladi, R.; Duarte, R.F.; et al. Prophylaxis and Management of Graft versus Host Disease after Stem-Cell Transplantation for Haematological Malignancies: Updated Consensus Recommendations of the European Society for Blood and Marrow Transplantation. Lancet Haematol. 2020, 7, e157–e167. [Google Scholar] [CrossRef]
  11. Wang, L.; Wang, L.; Cong, X.; Liu, G.; Zhou, J.; Bai, B.; Li, Y.; Bai, W.; Li, M.; Ji, H.; et al. Human umbilical cord mesenchymal stem cell therapy for patients with active rheumatoid arthritis: Safety and efficacy. Stem Cells Dev. 2013, 22, 3192–3202. [Google Scholar] [CrossRef] [PubMed]
  12. Lazarus, H.M.; Haynesworth, S.E.; Gerson, S.L.; Rosenthal, N.S.; Caplan, A.I. Ex Vivo Expansion and Subsequent Infusion of Human Bone Marrow-Derived Stromal Progenitor Cells (Mesenchymal Progenitor Cells): Implications for Therapeutic Use. Bone Marrow Transplant. 1995, 16, 557–564. [Google Scholar] [PubMed]
  13. Le Blanc, K.; Rasmusson, I.; Sundberg, B.; Götherström, C.; Hassan, M.; Uzunel, M.; Ringdén, O. Treatment of Severe Acute Graft-versus-Host Disease with Third Party Haploidentical Mesenchymal Stem Cells. Lancet 2004, 363, 1439–1441. [Google Scholar] [CrossRef]
  14. Wang, L.-T.; Liu, K.-J.; Sytwu, H.-K.; Yen, M.-L.; Yen, B.L. Advances in Mesenchymal Stem Cell Therapy for Immune and Inflammatory Diseases: Use of Cell-Free Products and Human Pluripotent Stem Cell-Derived Mesenchymal Stem Cells. Stem Cells Transl. Med. 2021, 10, 1288–1303. [Google Scholar] [CrossRef]
  15. Justiz Vaillant, A.A.; Misra, S.; Fitzgerald, B.M. Acute Transplantation Rejection. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2025. [Google Scholar]
  16. Fang, B.; Song, Y.; Liao, L.; Zhang, Y.; Zhao, R.C. Favorable Response to Human Adipose Tissue-Derived Mesenchymal Stem Cells in Steroid-Refractory Acute Graft-versus-Host Disease. Transplant. Proc. 2007, 39, 3358–3362. [Google Scholar] [CrossRef]
  17. Bonig, H.; Kuçi, Z.; Kuçi, S.; Bakhtiar, S.; Basu, O.; Bug, G.; Dennis, M.; Greil, J.; Barta, A.; Kállay, K.M.; et al. Children and Adults with Refractory Acute Graft-versus-Host Disease Respond to Treatment with the Mesenchymal Stromal Cell Preparation “MSC-FFM”-Outcome Report of 92 Patients. Cells 2019, 8, 1577. [Google Scholar] [CrossRef]
  18. Weng, J.Y.; Du, X.; Geng, S.X.; Peng, Y.W.; Wang, Z.; Lu, Z.S.; Wu, S.J.; Luo, C.W.; Guo, R.; Ling, W.; et al. Mesenchymal Stem Cell as Salvage Treatment for Refractory Chronic GVHD. Bone Marrow Transplant. 2010, 45, 1732–1740. [Google Scholar] [CrossRef]
  19. Branisteanu, D.E.; Georgescu, S.; Serban, I.L.; Pinzariu, A.C.; Boda, D.; Maranduca, M.A.; Glod, M.; Branisteanu, C.I.; Bilibau, R.; Dimitriu, A.; et al. Management of Psoriasis in Children (Review). Exp. Ther. Med. 2021, 22, 1429. [Google Scholar] [CrossRef] [PubMed]
  20. Noyan, F.; Zimmermann, K.; Hardtke-Wolenski, M.; Knoefel, A.; Schulde, E.; Geffers, R.; Hust, M.; Huehn, J.; Galla, M.; Morgan, M.; et al. Prevention of Allograft Rejection by Use of Regulatory T Cells With an MHC-Specific Chimeric Antigen Receptor. Am. J. Transplant. 2017, 17, 917–930. [Google Scholar] [CrossRef]
  21. 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]
  22. Arabpour, M.; Saghazadeh, A.; Rezaei, N. Anti-inflammatory and M2 macrophage polarization-promoting effect of mesenchymal stem cell-derived exosomes. Int. Immunopharmacol. 2021, 97, 107823. [Google Scholar] [CrossRef] [PubMed]
  23. Niu, J.; Yue, W.; Song, Y.; Zhang, Y.; Qi, X.; Wang, Z.; Liu, B.; Shen, H.; Hu, X. Prevention of Acute Liver Allograft Rejection by IL-10-Engineered Mesenchymal Stem Cells. Clin. Exp. Immunol. 2014, 176, 473–484. [Google Scholar] [CrossRef]
  24. Cao, Z.; Zhang, G.; Wang, F.; Liu, H.; Liu, L.; Han, Y.; Zhang, J.; Yuan, J. Protective Effects of Mesenchymal Stem Cells with CXCR4 Up-Regulation in a Rat Renal Transplantation Model. PLoS ONE 2013, 8, e82949. [Google Scholar] [CrossRef]
  25. Jia, S.; Liu, Y.; Yuan, J. Evidence in Guidelines for Treatment of Coronary Artery Disease. Adv. Exp. Med. Biol. 2020, 1177, 37–73. [Google Scholar] [CrossRef]
  26. Zhang, Y.; Zhao, D.; Tian, C.; Li, F.; Li, X.; Zhang, L.; Yang, H. Stro-1-Positive Human Mesenchymal Stem Cells Prolong Skin Graft Survival in Mice. Transplant. Proc. 2013, 45, 726–729. [Google Scholar] [CrossRef]
  27. Franceschini, M.; Boffa, A.; Andriolo, L.; Di Martino, A.; Zaffagnini, S.; Filardo, G. The 50 Most-Cited Clinical Articles in Cartilage Surgery Research: A Bibliometric Analysis. Knee Surg. Sports Traumatol. Arthrosc. 2022, 30, 1901–1914. [Google Scholar] [CrossRef] [PubMed]
  28. Kuroda, R.; Ishida, K.; Matsumoto, T.; Akisue, T.; Fujioka, H.; Mizuno, K.; Ohgushi, H.; Wakitani, S.; Kurosaka, M. Treatment of a Full-Thickness Articular Cartilage Defect in the Femoral Condyle of an Athlete with Autologous Bone-Marrow Stromal Cells. Osteoarthr. Cartil. 2007, 15, 226–231. [Google Scholar] [CrossRef]
  29. Pînzariu, A.; Sindilar, A.; Haliga, R.; Chelaru, L.; Mocanu, V. Nutritional Factors in Transdifferentiation of Scheletal Muscles to Adipocytes. Rev. Med. Chir. Soc. Med. Nat. Iasi 2014, 118, 699–705. [Google Scholar]
  30. Villa-Suárez, J.M.; García-Fontana, C.; Andújar-Vera, F.; González-Salvatierra, S.; de Haro-Muñoz, T.; Contreras-Bolívar, V.; García-Fontana, B.; Muñoz-Torres, M. Hypophosphatasia: A Unique Disorder of Bone Mineralization. Int. J. Mol. Sci. 2021, 22, 4303. [Google Scholar] [CrossRef]
  31. Amin, H.D.; Brady, M.A.; St-Pierre, J.-P.; Stevens, M.M.; Overby, D.R.; Ethier, C.R. Stimulation of Chondrogenic Differentiation of Adult Human Bone Marrow-Derived Stromal Cells by a Moderate-Strength Static Magnetic Field. Tissue Eng. Part A 2014, 20, 1612–1620. [Google Scholar] [CrossRef]
  32. Formiga, F.R.; Pelacho, B.; Garbayo, E.; Abizanda, G.; Gavira, J.J.; Simon-Yarza, T.; Mazo, M.; Tamayo, E.; Jauquicoa, C.; Ortiz-de-Solorzano, C.; et al. Sustained Release of VEGF through PLGA Microparticles Improves Vasculogenesis and Tissue Remodeling in an Acute Myocardial Ischemia-Reperfusion Model. J. Control. Release 2010, 147, 30–37. [Google Scholar] [CrossRef] [PubMed]
  33. Malekpour, K.; Hazrati, A.; Zahar, M.; Markov, A.; Zekiy, A.O.; Navashenaq, J.G.; Roshangar, L.; Ahmadi, M. The Potential Use of Mesenchymal Stem Cells and Their Derived Exosomes for Orthopedic Diseases Treatment. Stem Cell Rev. Rep. 2022, 18, 933–951. [Google Scholar] [CrossRef] [PubMed]
  34. Undale, A.H.; Westendorf, J.J.; Yaszemski, M.J.; Khosla, S. Mesenchymal Stem Cells for Bone Repair and Metabolic Bone Diseases. Mayo Clin. Proc. 2009, 84, 893–902. [Google Scholar] [CrossRef]
  35. Wenkert, D.; McAlister, W.H.; Coburn, S.P.; Zerega, J.A.; Ryan, L.M.; Ericson, K.L.; Hersh, J.H.; Mumm, S.; Whyte, M.P. Hypophosphatasia: Nonlethal Disease despite Skeletal Presentation in Utero (17 New Cases and Literature Review). J. Bone Miner. Res. 2011, 26, 2389–2398. [Google Scholar] [CrossRef] [PubMed]
  36. Cabanillas Stanchi, K.M.; Böhringer, J.; Strölin, M.; Groeschel, S.; Lenglinger, K.; Treuner, C.; Kehrer, C.; Laugwitz, L.; Bevot, A.; Kaiser, N.; et al. Hematopoietic Stem Cell Transplantation with Mesenchymal Stromal Cells in Children with Metachromatic Leukodystrophy. Stem Cells Dev. 2022, 31, 163–175. [Google Scholar] [CrossRef] [PubMed]
  37. Kon, E.; Filardo, G.; Roffi, A.; Di Martino, A.; Hamdan, M.; De Pasqual, L.; Merli, M.L.; Marcacci, M. Bone Regeneration with Mesenchymal Stem Cells. Clin. Cases Miner. Bone Metab. 2012, 9, 24–27. [Google Scholar] [PubMed]
  38. Kouraki, A.; Bast, T.; Ferguson, E.; Valdes, A.M. The Association of Socio-Economic and Psychological Factors with Limitations in Day-to-Day Activity over 7 Years in Newly Diagnosed Osteoarthritis Patients. Sci. Rep. 2022, 12, 943. [Google Scholar] [CrossRef]
  39. Mezey, G.A.; Paulik, E.; Máté, Z. Effect of Osteoarthritis and Its Surgical Treatment on Patients’ Quality of Life: A Longitudinal Study. BMC Musculoskelet. Disord. 2023, 24, 537. [Google Scholar] [CrossRef]
  40. Molnar, V.; Pavelić, E.; Vrdoljak, K.; Čemerin, M.; Klarić, E.; Matišić, V.; Bjelica, R.; Brlek, P.; Kovačić, I.; Tremolada, C.; et al. Mesenchymal Stem Cell Mechanisms of Action and Clinical Effects in Osteoarthritis: A Narrative Review. Genes 2022, 13, 949. [Google Scholar] [CrossRef]
  41. Tan, S.H.S.; Kwan, Y.T.; Neo, W.J.; Chong, J.Y.; Kuek, T.Y.J.; See, J.Z.F.; Wong, K.L.; Toh, W.S.; Hui, J.H.P. Intra-Articular Injections of Mesenchymal Stem Cells Without Adjuvant Therapies for Knee Osteoarthritis: A Systematic Review and Meta-Analysis. Am. J. Sports Med. 2021, 49, 3113–3124. [Google Scholar] [CrossRef]
  42. Mohamadnejad, M.; Alimoghaddam, K.; Mohyeddin-Bonab, M.; Bagheri, M.; Bashtar, M.; Ghanaati, H.; Baharvand, H.; Ghavamzadeh, A.; Malekzadeh, R. Phase 1 Trial of Autologous Bone Marrow Mesenchymal Stem Cell Transplantation in Patients with Decompensated Liver Cirrhosis. Arch. Iran. Med. 2007, 10, 459–466. [Google Scholar]
  43. Kharaziha, P.; Hellström, P.M.; Noorinayer, B.; Farzaneh, F.; Aghajani, K.; Jafari, F.; Telkabadi, M.; Atashi, A.; Honardoost, M.; Zali, M.R.; et al. Improvement of Liver Function in Liver Cirrhosis Patients after Autologous Mesenchymal Stem Cell Injection: A Phase I-II Clinical Trial. Eur. J. Gastroenterol. Hepatol. 2009, 21, 1199–1205. [Google Scholar] [CrossRef] [PubMed]
  44. Lu, W.; Qu, J.; Yan, L.; Tang, X.; Wang, X.; Ye, A.; Zou, Z.; Li, L.; Ye, J.; Zhou, L. Efficacy and Safety of Mesenchymal Stem Cell Therapy in Liver Cirrhosis: A Systematic Review and Meta-Analysis. Stem Cell Res. Ther. 2023, 14, 301. [Google Scholar] [CrossRef] [PubMed]
  45. Liu, Y.; Dong, Y.; Wu, X.; Xu, X.; Niu, J. The Assessment of Mesenchymal Stem Cells Therapy in Acute on Chronic Liver Failure and Chronic Liver Disease: A Systematic Review and Meta-Analysis of Randomized Controlled Clinical Trials. Stem Cell Res. Ther. 2022, 13, 204. [Google Scholar] [CrossRef]
  46. Choi, E.W. Adult Stem Cell Therapy for Autoimmune Disease. Int. J. Stem Cells 2009, 2, 122–128. [Google Scholar] [CrossRef]
  47. Hernanz, N.; Sierra, M.; Volpato, N.; Núñez-Gómez, L.; Mesonero, F.; Herrera-Puente, P.; García-Gutiérrez, V.; Albillos, A.; López-San Román, A. Autologous Haematopoietic Stem Cell Transplantation in Refractory Crohn’s Disease: Experience in Our Centre. Gastroenterol. Hepatol. 2019, 42, 16–22. [Google Scholar] [CrossRef] [PubMed]
  48. Burt, R.K.; Traynor, A.; Statkute, L.; Barr, W.G.; Rosa, R.; Schroeder, J.; Verda, L.; Krosnjar, N.; Quigley, K.; Yaung, K.; et al. Nonmyeloablative Hematopoietic Stem Cell Transplantation for Systemic Lupus Erythematosus. JAMA 2006, 295, 527–535. [Google Scholar] [CrossRef]
  49. Puyade, M.; Patel, A.; Lim, Y.J.; Blank, N.; Badoglio, M.; Gualandi, F.; Ma, D.D.; Maximova, N.; Greco, R.; Alexander, T.; et al. Autologous Hematopoietic Stem Cell Transplantation for Behçet’s Disease: A Retrospective Survey of Patients Treated in Europe, on Behalf of the Autoimmune Diseases Working Party of the European Society for Blood and Marrow Transplantation. Front. Immunol. 2021, 12, 638709. [Google Scholar] [CrossRef]
  50. Madec, A.M.; Mallone, R.; Afonso, G.; Abou Mrad, E.; Mesnier, A.; Eljaafari, A.; Thivolet, C. Mesenchymal Stem Cells Protect NOD Mice from Diabetes by Inducing Regulatory T Cells. Diabetologia 2009, 52, 1391–1399. [Google Scholar] [CrossRef]
  51. Lu, Z.; Hu, X.; Zhu, C.; Wang, D.; Zheng, X.; Liu, Q. Overexpression of CNTF in Mesenchymal Stem Cells Reduces Demyelination and Induces Clinical Recovery in Experimental Autoimmune Encephalomyelitis Mice. J. Neuroimmunol. 2009, 206, 58–69. [Google Scholar] [CrossRef]
  52. Yuan, X.; Sun, L. Stem Cell Therapy in Lupus. Rheumatol. Immunol. Res. 2022, 3, 61–68. [Google Scholar] [CrossRef]
  53. Fava, A.; Petri, M. Systemic Lupus Erythematosus: Diagnosis and Clinical Management. J. Autoimmun. 2019, 96, 1–13. [Google Scholar] [CrossRef] [PubMed]
  54. Cheng, R.-J.; Xiong, A.-J.; Li, Y.-H.; Pan, S.-Y.; Zhang, Q.-P.; Zhao, Y.; Liu, Y.; Marion, T.N. Mesenchymal Stem Cells: Allogeneic MSC May Be Immunosuppressive but Autologous MSC Are Dysfunctional in Lupus Patients. Front. Cell. Dev. Biol. 2019, 7, 285. [Google Scholar] [CrossRef]
  55. Sun, L.; Akiyama, K.; Zhang, H.; Yamaza, T.; Hou, Y.; Zhao, S.; Xu, T.; Le, A.; Shi, S. Mesenchymal Stem Cell Transplantation Reverses Multiorgan Dysfunction in Systemic Lupus Erythematosus Mice and Humans. Stem Cells 2009, 27, 1421–1432. [Google Scholar] [CrossRef]
  56. Smith-Berdan, S.; Gille, D.; Weissman, I.L.; Christensen, J.L. Reversal of Autoimmune Disease in Lupus-Prone New Zealand Black/New Zealand White Mice by Nonmyeloablative Transplantation of Purified Allogeneic Hematopoietic Stem Cells. Blood 2007, 110, 1370–1378. [Google Scholar] [CrossRef]
  57. Zhou, K.; Zhang, H.; Jin, O.; Feng, X.; Yao, G.; Hou, Y.; Sun, L. Transplantation of Human Bone Marrow Mesenchymal Stem Cell Ameliorates the Autoimmune Pathogenesis in MRL/Lpr Mice. Cell. Mol. Immunol. 2008, 5, 417–424. [Google Scholar] [CrossRef]
  58. Turovsky, E.A.; Golovicheva, V.V.; Varlamova, E.G.; Danilina, T.I.; Goryunov, K.V.; Shevtsova, 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]
  59. Yamout, B.; Hourani, R.; Salti, H.; Barada, W.; El-Hajj, T.; Al-Kutoubi, A.; Herlopian, A.; Baz, E.K.; Mahfouz, R.; Khalil-Hamdan, R.; et al. Bone Marrow Mesenchymal Stem Cell Transplantation in Patients with Multiple Sclerosis: A Pilot Study. J. Neuroimmunol. 2010, 227, 185–189. [Google Scholar] [CrossRef] [PubMed]
  60. Mallam, E.; Kemp, K.; Wilkins, A.; Rice, C.; Scolding, N. Characterization of in Vitro Expanded Bone Marrow-Derived Mesenchymal Stem Cells from Patients with Multiple Sclerosis. Mult. Scler. 2010, 16, 909–918. [Google Scholar] [CrossRef]
  61. Jetta, D.; Shireen, T.; Hua, S.Z. Epithelial Cells Sense Local Stiffness via Piezo1 Mediated Cytoskeletal Reorganization. Front. Cell. Dev. Biol. 2023, 11, 1198109. [Google Scholar] [CrossRef]
  62. Islam, M.A.; Alam, S.S.; Kundu, S.; Ahmed, S.; Sultana, S.; Patar, A.; Hossan, T. Mesenchymal Stem Cell Therapy in Multiple Sclerosis: A Systematic Review and Meta-Analysis. J. Clin. Med. 2023, 12, 6311. [Google Scholar] [CrossRef] [PubMed]
  63. Vaheb, S.; Afshin, S.; Ghoshouni, H.; Ghaffary, E.M.; Farzan, M.; Shaygannejad, V.; Thapa, S.; Zabeti, A.; Mirmosayyeb, O. Neurological Efficacy and Safety of Mesenchymal Stem Cells (MSCs) Therapy in People with Multiple Sclerosis (pwMS): An Updated Systematic Review and Meta-Analysis. Mult. Scler. Relat. Disord. 2024, 87, 105681. [Google Scholar] [CrossRef]
  64. Terstappen, G.C.; Meyer, A.H.; Bell, R.D.; Zhang, W. Strategies for Delivering Therapeutics across the Blood-Brain Barrier. Nat. Rev. Drug Discov. 2021, 20, 362–383. [Google Scholar] [CrossRef]
  65. Harris, V.K.; Faroqui, R.; Vyshkina, T.; Sadiq, S.A. Characterization of Autologous Mesenchymal Stem Cell-Derived Neural Progenitors as a Feasible Source of Stem Cells for Central Nervous System Applications in Multiple Sclerosis. Stem Cells Transl. Med. 2012, 1, 536–547. [Google Scholar] [CrossRef]
  66. Bai, Y.-B.; Zhao, F.; Wu, Z.-H.; Shi, G.-N.; Jiang, N. Left Ventricular Thrombosis Caused Cerebral Embolism during Venoarterial Extracorporeal Membrane Oxygenation Support: A Case Report. World J. Clin. Cases 2024, 12, 973–979. [Google Scholar] [CrossRef] [PubMed]
  67. Huang, Y.; Dreyfus, C.F. The Role of Growth Factors as a Therapeutic Approach to Demyelinating Disease. Exp. Neurol. 2016, 283, 531–540. [Google Scholar] [CrossRef] [PubMed]
  68. Petrou, P.; Kassis, I.; Levin, N.; Paul, F.; Backner, Y.; Benoliel, T.; Oertel, F.C.; Scheel, M.; Hallimi, M.; Yaghmour, N.; et al. Beneficial Effects of Autologous Mesenchymal Stem Cell Transplantation in Active Progressive Multiple Sclerosis. Brain 2020, 143, 3574–3588. [Google Scholar] [CrossRef]
  69. Harris, V.K.; Stark, J.W.; Yang, S.; Zanker, S.; Tuddenham, J.; Sadiq, S.A. Mesenchymal Stem Cell-Derived Neural Progenitors in Progressive MS: Two-Year Follow-up of a Phase I Study. Neurol. Neuroimmunol. Neuroinflamm. 2020, 8, e928. [Google Scholar] [CrossRef]
  70. Sarsenova, M.; Issabekova, A.; Abisheva, S.; Rutskaya-Moroshan, K.; Ogay, V.; Saparov, A. Mesenchymal Stem Cell-Based Therapy for Rheumatoid Arthritis. Int. J. Mol. Sci. 2021, 22, 11592. [Google Scholar] [CrossRef] [PubMed]
  71. Zhang, Q.; Li, Q.; Zhu, J.; Guo, H.; Zhai, Q.; Li, B.; Jin, Y.; He, X.; Jin, F. Comparison of Therapeutic Effects of Different Mesenchymal Stem Cells on Rheumatoid Arthritis in Mice. PeerJ 2019, 7, e7023. [Google Scholar] [CrossRef]
  72. Hwang, J.J.; Rim, Y.A.; Nam, Y.; Ju, J.H. Recent Developments in Clinical Applications of Mesenchymal Stem Cells in the Treatment of Rheumatoid Arthritis and Osteoarthritis. Front. Immunol. 2021, 12, 631291. [Google Scholar] [CrossRef]
  73. Park, E.H.; Lim, H.-S.; Lee, S.; Roh, K.; Seo, K.-W.; Kang, K.-S.; Shin, K. Intravenous Infusion of Umbilical Cord Blood-Derived Mesenchymal Stem Cells in Rheumatoid Arthritis: A Phase Ia Clinical Trial. Stem Cells Transl. Med. 2018, 7, 636–642. [Google Scholar] [CrossRef]
  74. Ghoryani, M.; Shariati-Sarabi, Z.; Tavakkol-Afshari, J.; Mohammadi, M. The Sufficient Immunoregulatory Effect of Autologous Bone Marrow-Derived Mesenchymal Stem Cell Transplantation on Regulatory T Cells in Patients with Refractory Rheumatoid Arthritis. J. Immunol. Res. 2020, 2020, 3562753. [Google Scholar] [CrossRef]
  75. Sage, E.K.; Thakrar, R.M.; Janes, S.M. Genetically Modified Mesenchymal Stromal Cells in Cancer Therapy. Cytotherapy 2016, 18, 1435–1445. [Google Scholar] [CrossRef] [PubMed]
  76. Lalu, M.M.; McIntyre, L.; Pugliese, C.; Fergusson, D.; Winston, B.W.; Marshall, J.C. Safety of Cell Therapy with Mesenchymal Stromal Cells (SafeCell): A Systematic Review and Meta-Analysis of Clinical Trials. PLoS ONE 2012, 7, e47559. [Google Scholar] [CrossRef]
  77. Tolar, J.; Nauta, A.J.; Osborn, M.J.; Panoskaltsis Mortari, A.; McElmurry, R.T.; Bell, S.; Xia, L.; Zhou, N.; Riddle, M.; Schroeder, T.M.; et al. Sarcoma Derived from Cultured Mesenchymal Stem Cells. Stem Cells 2007, 25, 371–379. [Google Scholar] [CrossRef]
  78. Ning, H.; Yang, F.; Jiang, M.; Hu, L.; Feng, K.; Zhang, J.; Yu, Z.; Li, B.; Xu, C.; Li, Y.; et al. The Correlation between Cotransplantation of Mesenchymal Stem Cells and Higher Recurrence Rate in Hematologic Malignancy Patients: Outcome of a Pilot Clinical Study. Leukemia 2008, 22, 593–599. [Google Scholar] [CrossRef]
  79. Sage, E.K.; Kolluri, K.K.; McNulty, K.; Lourenco, S.D.S.; Kalber, T.L.; Ordidge, K.L.; Davies, D.; Gary Lee, Y.C.; Giangreco, A.; Janes, S.M. Systemic but Not Topical TRAIL-Expressing Mesenchymal Stem Cells Reduce Tumour Growth in Malignant Mesothelioma. Thorax 2014, 69, 638–647. [Google Scholar] [CrossRef]
  80. Wang, X.; Chen, W.; Yuan, Y. KSHV Enhances Mesenchymal Stem Cell Homing and Promotes KS-like Pathogenesis. Virology 2020, 549, 5–12. [Google Scholar] [CrossRef]
  81. Niess, H.; von Einem, J.C.; Thomas, M.N.; Michl, M.; Angele, M.K.; Huss, R.; Günther, C.; Nelson, P.J.; Bruns, C.J.; Heinemann, V. Treatment of Advanced Gastrointestinal Tumors with Genetically Modified Autologous Mesenchymal Stromal Cells (TREAT-ME1): Study Protocol of a Phase I/II Clinical Trial. BMC Cancer 2015, 15, 237. [Google Scholar] [CrossRef]
  82. Schweizer, M.T.; Wang, H.; Bivalacqua, T.J.; Partin, A.W.; Lim, S.J.; Chapman, C.; Abdallah, R.; Levy, O.; Bhowmick, N.A.; Karp, J.M.; et al. A Phase I Study to Assess the Safety and Cancer-Homing Ability of Allogeneic Bone Marrow-Derived Mesenchymal Stem Cells in Men with Localized Prostate Cancer. Stem Cells Transl. Med. 2019, 8, 441–449. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Applications of MSCs with multiple differentiation potential for repair of various tissues.
Figure 1. Applications of MSCs with multiple differentiation potential for repair of various tissues.
Ijms 26 03084 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

Pînzariu, A.C.; Moscalu, R.; Soroceanu, R.P.; Maranduca, M.A.; Drochioi, I.C.; Vlasceanu, V.I.; Timofeiov, S.; Timofte, D.V.; Huzum, B.; Moscalu, M.; et al. The Therapeutic Use and Potential of MSCs: Advances in Regenerative Medicine. Int. J. Mol. Sci. 2025, 26, 3084. https://doi.org/10.3390/ijms26073084

AMA Style

Pînzariu AC, Moscalu R, Soroceanu RP, Maranduca MA, Drochioi IC, Vlasceanu VI, Timofeiov S, Timofte DV, Huzum B, Moscalu M, et al. The Therapeutic Use and Potential of MSCs: Advances in Regenerative Medicine. International Journal of Molecular Sciences. 2025; 26(7):3084. https://doi.org/10.3390/ijms26073084

Chicago/Turabian Style

Pînzariu, Alin Constantin, Roxana Moscalu, Radu Petru Soroceanu, Minela Aida Maranduca, Ilie Cristian Drochioi, Vlad Ionut Vlasceanu, Sergiu Timofeiov, Daniel Vasile Timofte, Bogdan Huzum, Mihaela Moscalu, and et al. 2025. "The Therapeutic Use and Potential of MSCs: Advances in Regenerative Medicine" International Journal of Molecular Sciences 26, no. 7: 3084. https://doi.org/10.3390/ijms26073084

APA Style

Pînzariu, A. C., Moscalu, R., Soroceanu, R. P., Maranduca, M. A., Drochioi, I. C., Vlasceanu, V. I., Timofeiov, S., Timofte, D. V., Huzum, B., Moscalu, M., Serban, D. N., & Serban, I. L. (2025). The Therapeutic Use and Potential of MSCs: Advances in Regenerative Medicine. International Journal of Molecular Sciences, 26(7), 3084. https://doi.org/10.3390/ijms26073084

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