Open access peer-reviewed chapter

Clinical Relevance of Mesenchymal Stromal Cells from Various Sources: Insights into Transcriptome Analysis for Identifying Inherent Potential

Written By

Dana M. Alhattab, Salwa Alshehri and Fatima Jamali

Submitted: 01 December 2023 Reviewed: 04 December 2023 Published: 21 December 2023

DOI: 10.5772/intechopen.1004004

From the Edited Volume

Recent Update on Mesenchymal Stem Cells

Khalid Ahmed Al-Anazi

Chapter metrics overview

41 Chapter Downloads

View Full Metrics

Abstract

This book chapter provides an in-depth overview of the clinical relevance of mesenchymal stem cells (MSCs) derived from various sources, highlighting the importance of whole transcriptome analysis in revealing their inherent potential. The chapter delves into different sources of MSCs, such as bone marrow, adipose tissue, umbilical cord, and placenta, and compares their respective properties and capabilities. Additionally, it explores the latest advancements in whole transcriptome analysis, including RNA sequencing and microarray analysis, and their applications in MSC research. The aim is to provide a comprehensive understanding of how high-end technologies, such as whole transcriptome analysis, can aid in identifying the inherent potential of cells for therapeutic applications. It will also discuss how such gene expression approaches helped identify the inherent potential of specific MSC sources, tailoring their use towards specific clinical applications, including immune tolerance and modulation, osteogenesis, and chondrogenesis. Additionally, it highlights the importance of extracellular vesicles derived from MSCs. This knowledge will be beneficial for researchers and clinicians working towards developing MSC-based treatments for regenerative medicine and cellular therapy.

Keywords

  • mesenchymal stromal cells
  • transcriptome analysis
  • RNA-seq
  • inherent potential
  • immune modulation
  • osteogenesis
  • chondrogenesis
  • exosomes

1. Introduction

Mesenchymal stromal cells (MSCs), also referred to as mesenchymal stem cells, are multipotent stromal cells with self-renewal capacity and multilineage differentiation potential [1]. There has been an increasing interest in MSCs in recent years due to their unique properties, including long-term proliferation, multilineage differentiation potential, and immunomodulatory capabilities [1]. These properties make MSCs a promising candidate for use in regenerative medicine and cellular therapy.

The discovery of MSCs dates back to 1976, when Friedenstein et al. identified a group of non-hematopoietic, plastic-adherent, fibroblast-like cells that could be extracted from the bone marrow (BM). Since then, MSCs have been identified in various tissues, including adipose tissue (AT), umbilical cord (UC), and amniotic fluid [2, 3]. It is, however, essential to note that MSCs are not homogenous in their properties and capabilities. The source from which they are derived significantly determines specific cellular characteristics. MSCs from different tissues exhibit distinctive properties that may influence their potential clinical applications [4, 5]. Several studies have been conducted that compare MSCs from various sources [6, 7, 8]. For instance, Hsieh et al. performed a side-by-side functional comparison between UC-MSCs and BM-MSCs and found that BM-MSCs can be easily differentiated into osteocytes compared to UC-MSCs, which showed delayed and insufficient differentiation into osteocytes [8]. Another study by Karahuseyinoglu et al. identified that UC-MSCs possess higher chondrogenic differentiation capacity compared to BM-MSCs [9]. Therefore, it is critical to understand the inherent potential of MSCs from various sources before they can be used for clinical applications.

Whole transcriptome analysis is a powerful tool that enables researchers to gain a deep understanding of the gene expression patterns and regulatory networks that govern the properties and functions of MSCs. This cutting-edge approach allows for a comprehensive exploration of the molecular mechanisms that underlie the unique characteristics of MSCs. To this end, various studies have analyzed the global gene expression profile for different types of MSCs [10, 11, 12]. Some of these studies focused on evaluating the expression levels of pluripotency genes and embryonic stem cell (ESC) markers [13, 14]. Other studies investigated the expression of genes that have functional significance, such as genes involved in bone and cardiovascular development [13, 15]. Overall, whole transcriptome analysis is a valuable tool for gaining insights into the complex mechanisms that govern the behavior of MSCs.

This chapter will review the clinical relevance of MSCs derived from various sources, highlighting the importance of whole transcriptome analysis in unraveling their inherent potential. It will discuss the different sources of MSCs, including BM, AT, UC, and placenta (PL), and compare their properties and capabilities. Additionally, it will explore the latest developments in whole transcriptome analysis, including RNA sequencing and microarray analysis, and their applications in MSC research. The aim is to provide a comprehensive insight into high-end technologies, such as whole transcriptome analysis, and how they can aid in identifying the inherent potential of cells for therapeutic applications. This knowledge will be useful for researchers and clinicians working towards developing MSC-based treatments for regenerative medicine and cellular therapy. Furthermore, new strategies to enhance the therapeutic potential of MSCs are discussed.

Advertisement

2. Mesenchymal stem cell sources

While bone marrow-derived MSCs (BM-MSCs) have been extensively studied for the past four decades, the last decade has witnessed a shift towards exploring alternative sources of MSCs. This shift is driven by the search for cell sources that are easily accessible, noninvasive, and available in abundant quantity [16]. As a result, MSCs have been successfully isolated from diverse tissues, such as AT and perinatal tissues like the UC and PL [2, 3, 17]. Each source of MSCs has its advantages and limitations, and ongoing research aims to further understand their unique properties and refine their applications in clinical settings. Additionally, studies are exploring other sources, such as synovial fluid, dental pulp, and more, expanding the potential repertoire of MSCs for therapeutic purposes. Besides the MSCs derived from the BM, other tissue sources of MSCs provide technical advantages. For instance, harvesting BM-MSCs involves an invasive procedure, and their expansion is imperative due to the restricted number of stem cells [18]. Conversely, adipose tissue-derived MSCs (AT-MSCs) are more accessible and produce a greater number of cells, simplifying the expansion process [19]. Such technical distinctions highlight the importance of tailoring protocols for isolating and expanding MSCs based on their specific tissue source.

2.1 Bone marrow-derived MSCs (BM-MSCs)

Traditionally considered the gold standard, BM-MSCs have been the focus of extensive research and clinical trials [20]. Isolation from the bone marrow involves a relatively invasive procedure, typically bone marrow aspiration from the iliac crest [21]. BM-MSCs exhibit robust self-renewal capacity and the ability to differentiate into various cell lineages, including osteoblasts, adipocytes, and chondrocytes [22]. Surface markers commonly associated with BM-MSCs include CD90, CD105, CD73, and CD44, while they lack the expression of hematopoietic markers such as CD34 and CD45 [23]. Research on the application of BM-MSCs has shown promising results in treating various diseases [24]. Nonetheless, discrepancies in results among studies can be ascribed to factors like donor variability, cell preparation procedures, and a deficiency in standardization before transplantation [25]. It is important to note that the expansion and cultivation process can vary based on specific protocols, and researchers continuously work on optimizing these procedures for the best outcomes in terms of cell yield, quality, and therapeutic efficacy.

2.2 Adipose tissue-derived MSCs (AT-MSCs)

AT has emerged as a rich and easily accessible source of MSCs. AT-MSCs can be isolated from fat tissue through a minimally invasive procedure, usually involving liposuction or fat removal during other surgical procedures. AT-MSCs share similarities with BM-MSCs in terms of their differentiation potential [26, 27]. However, they may offer advantages in terms of abundance and ease of isolation, making them an attractive alternative to regenerative therapies. As BM-MSCs, AT-MSCs express typical stem cell surface markers and possess the potential to differentiate into various lineages [23, 28]. Due to its accessibility and abundance, AT yields the isolation of stem cells at a quantity 500 times greater than those obtained from bone marrow [29]. AT-MSCs have been studied for their ability to enhance wound healing processes. The cells contribute to tissue regeneration by promoting the proliferation and migration of various cells involved in wound repair [30, 31, 32]. Other studies have investigated the potential of AT-MSCs in neural tissue repair. AT-MSCs may contribute to neuroprotection and neurodegeneration after injury [33]. They have also demonstrated a pro-angiogenic effect, promoting the formation of new blood vessels. Studies suggest that the paracrine factors released by AT-MSCs contribute to angiogenesis, which is crucial for tissue repair and regeneration [34]. Research and preclinical studies have explored the use of AT-MSCs in specific autoimmune diseases such as rheumatoid arthritis, multiple sclerosis, and systemic lupus erythematosus. Clinical trials have been conducted or are ongoing to assess the safety and efficacy of AT-MSCs in treating these conditions [35]. However, additional research is crucial to establish the optimal dosage, administration route, and safety parameters for clinical applications [36].

2.3 Umbilical cord-derived MSCs (UC-MSCs)

MSCs derived from the UC present a unique perinatal source with distinct properties. Isolation from the Wharton’s jelly of the UC is considered noninvasive and ethically uncontentious. UC-MSCs exhibit multilineage differentiation potential, like BM-MSCs. Moreover, they possess immunomodulatory properties that make them particularly interesting for therapeutic applications, especially in the context of immune-related disorders [24, 37]. In 1991, McElreavey et al. [38] made a groundbreaking discovery by isolating fibroblast-like cells from the Wharton’s Jelly (WJ) of the human UC. These cells are readily available after childbirth, providing a noninvasive and ethical source of MSCs. Obtaining a significant quantity of UC-MSCs through several passages and extensive ex vivo expansion is a simple process [39]. UC-MSCs are considered to be in a more primitive state compared to MSCs from other tissues, and they possess a high proliferative capacity [40]. In addition, UC-MSCs can be cryopreserved and stored for future use, allowing the creation of cell banks for potential therapeutic applications [41]. UC-MSCs display comparable surface phenotypes, adherence to plastic surfaces, and multipotency characteristics as MSCs obtained from other MSC tissue sources. UC-MSCs present versatile applications in both autologous and allogeneic contexts. Autologous use involves employing UC-MSCs for the same individuals they are sourced from. On the other hand, allogeneic use refers to the application of UC-MSCs across different individuals. Allogeneic UC-MSCs can be expanded and cryopreserved in cell banks, ready for use by patients in need. However, it is crucial to verify the health of the baby, who is the donor of UC-MSCs, through genomic or chromosomal tests, as their normal growth without health problems cannot be guaranteed in advance. Understanding the advantages and disadvantages is essential for each specific clinical application in both autologous and allogeneic scenarios [42].

2.4 Placenta-derived MSCs (PL-MSCs)

The PL plays a crucial role in fetal development by providing nutrition and supporting immunological tolerance. In recent times, stem cells derived from the PL, known as PL-MSCs, have gained attention as an alternative perinatal source of MSCs in regenerative medicine. PL-MSCs, originating from the fetus, exhibit the capacity for self-renewal and multipotency, possessing immunomodulatory properties, making them valuable for therapeutic applications [43]. Additionally, the PL harbors a substantial number of MSCs, and its utilization for research is not encumbered by the ethical concerns associated with human embryonic stem cells [44, 45].

2.5 Other sources of MSCs

2.5.1 Dental pulp-derived MSCs (DP-MSCs)

The dental pulp is the soft tissue at the core of a tooth, a highly vascularized connective tissue. It is enveloped by mineralized hard tissue and contains diverse cell types, including odontoblasts and undifferentiated progenitor cells [46]. Within this population of undifferentiated progenitors, MSCs are found to exhibit a high proliferation rate and a high degree of multipotency [47, 48, 49]. Adherent colonies of spindle-shaped cells, known as Colony-Forming Units Fibroblast (CFU-F), are formed by human dental pulp-derived stem cells. The analysis of CFU-F reveals that human dental pulp harbors a more abundant population of MSCs compared to human bone marrow. Specifically, the CFU-F capacity of DPSCs is five times greater than that of BM-MSCs [50, 51]. DP-MSCs have shown great promise in dental applications. They held the potential for dental pulp regeneration [52] and dentin repair [53], and showed regenerative capacity for nerve repair [54].

2.5.2 Synovial fluid-derived MSCs (SF-MSCs)

The existence and attributes of MSCs in synovium specimens were initially documented by De Bari et al. [55], and subsequent studies have extensively explored this topic [56, 57]. MSCs derived from synovium specimens (SF-MSCs) exhibit superior proliferative capacity and chondrogenic potential compared to MSCs from other sources. Various studies have demonstrated the multilineage differentiation potential of SF-MSCs in humans [58, 59]. Recent findings indicate that the chondrogenic differentiation capacity of equine SF-derived MSCs is comparable to that of bone marrow MSCs [60, 61], confirming the stemness of these cells according to the criteria set by the International Society for Cellular Therapy [23].

Advertisement

3. Transcriptome analysis and identification of inherent potentials

MSCs are being actively used in clinical trials as potential cellular therapies for various clinical conditions, with over 1000 trials registered on ClinicalTrials.gov (http://www.clinicaltrials.gov). MSCs isolated from the bone marrow are the first to be used in clinical trials and are the most commonly studied MSC source in preclinical and clinical studies [62]. MSCs from other sources are also being used to treat different clinical conditions, including MSCs from AT, UC, and others. However, variations in the clinical outcomes of MSC treatments exist, and to date, there is no consensus on the most suitable MSC source to treat a specific disease [63]. The inconsistent clinical outcomes of MSC treatments can be attributed to the heterogeneous potency and functional variations of the MSCs and the lack of efficient assays for assessing their potency [7, 62].

MSCs are still identified by their expression of a specific subset of surface marker proteins, trilineage differentiation potential, and immunomodulatory potential [23, 64]. Although these criteria have served the research community for a long time, they do not represent the heterogeneity of the MSC population. The scientific community should benefit from advanced technologies, such as whole transcriptome analysis and single-cell RNA-seq technology, to explore MSC identity and function regarding their tissue origin and functional status.

These approaches could provide a comprehensive view of gene expression patterns in MSCs, enabling researchers to gain a deeper understanding of the inherent capabilities of MSCs, including their differentiation potential, immunomodulatory effects, and regenerative properties. In a prior study, we conducted a gene expression analysis of four types of MSCs, including MSCs derived from the BM, AT, UC, and PL [16]. Our analysis revealed a unique set of genes that are either up- or down-regulated in one type of MSC compared to all other stem cell types. Moreover, we identified signature genes exclusively expressed in one type of MSC [16]. These findings clearly indicate intrinsic differences in the potential of MSCs from different sources.

Such information can be leveraged to develop more effective and targeted therapies for various conditions, such as osteoarthritis, cardiovascular disease, and neurological disorders. Moreover, whole transcriptome analysis can identify the specific genes and pathways involved in the biological processes of MSCs. Such an approach can refine the operational definition of MSCs and further our understanding of their native physiological function. Furthermore, this knowledge can inform the development of optimized culture conditions and clinical manufacturing protocols, leading to more consistent and effective MSC-based patient therapies. In the following sections, we will discuss how such gene expression approaches helped identify the inherent potential of specific MSC sources, tailoring their use towards specific clinical applications, including immune tolerance and modulation, osteogenesis, and chondrogenesis.

3.1 Immune tolerance and modulation

The modulation of the immune system and its tolerance is a complex process involving gene expression related to various biological functions. One of the critical properties of MSCs is their immune tolerance and survival mechanisms, which are essential for the success of stem cell transplantations in treating diseases [65]. MSCs typically express major histocompatibility (MHC) Class I antigens on their surface and not Class II; however, Class II antigens are upregulated by inflammatory agents [66]. Studies have shown that both autologous and allogenic MSCs prevent lymphocyte proliferation without causing apoptosis of T cells [67, 68]. MSCs release factors such as transforming growth factor β (TGF-β), interleukin 10 (IL-10), interleukin 6 (IL-6), and nitric oxide, which are known to affect immune cells [69, 70]. MSCs also affect the maturation of immune cells, increasing regulatory T cells (Treg), anti-inflammatory T helper 2 (TH2), and dendritic (DC2) cells [66, 71, 72]. Furthermore, they were found to induce M1 macrophages to adopt the anti-inflammatory M2 form and reduce IgG production from B cells [73]. Although the immunoregulatory potential of MSCs has been well established, the mechanisms underlying their actions are not yet fully understood [65]. Moreover, variations in the immunomodulatory capabilities among different types of MSCs are identified [6, 74]. These findings are manifested not only in in vitro studies but also in the outcomes of various clinical trials. For example, AT-MSCs were found to possess greater immunosuppressive capabilities than BM-MSCs in vitro and in vivo [75, 76].

Whole-gene expression analysis of MSCs from various sources can help identify the most suitable cells for clinical use by uncovering differences in their immunomodulatory potential. We previously conducted a comprehensive transcriptome analysis of MSCs from four sources to gauge their inherent potential [16]. We studied around 1400 immune modulation-related genes to understand if there were any differences in the MSCs’ ability to modulate immune responses. Our findings indicate that toll-like receptor 4 (TLR-4) expression was highest in BM-MSCs and lowest in UC- and PL-MSCs. While TLR-4 is traditionally known for activating innate immune cells against pathogens, its activation in MSCs has also been discovered to facilitate interactions with the surrounding environment [77] and induce Treg cell activation, which counteracts the inflammatory aspect of several diseases [78]. We have identified a significant immune modulatory trend in BM-MSCs, which was emphasized with increased expression of interleukin 7 (IL-7), an inhibitor of T-cell proliferation [79]. We also found an increase in CD200 expression, an immune player, in both BM- and UC-MSCs compared to other MSC types. In addition to its immune tolerance roles, CD200 inhibits the maturation of myeloid progenitors into inflammatory cells and suppresses the secretion of proinflammatory TNF-α in stimulated macrophages [80, 81]. On the other hand, CD274, a known immune-modulatory protein that plays a negative role in immune modulation, was found to be uniquely upregulated in UC- and PL-MSCs. Additionally, we found several proinflammatory pathways to be activated in other MSC types compared to BM-MSCs, such as interleukin 8 (IL-8) signaling, diabetes mellitus signaling, and TWEAK signaling. Our transcriptome analysis suggests that BM-MSCs may have a higher immune modulation potential than other MSC types.

Several studies have been conducted to evaluate the similarity and variability of gene expression in MSC samples [11, 82, 83]. In a study by Sun C et al., the transcriptomic variation of MSCs from different tissues, including PL, UC, and dental pulp, was investigated in relation to immunomodulatory function [10]. They found that among the genes that exhibited highly variable expression were genes involved in the differentiation process of MSCs and genes that regulated immunomodulation, such as CD274, C-C motif chemokine ligand 2 (CCL2), C-C motif chemokine ligand 5 (CCL5), IL6, colony-stimulating factor 3 (CSF3), and hepatocyte growth factor (HGF). Conversely, genes involved in other biological processes, such as metabolic processes, gene expression, RNA processes, and RNA binding, showed minimal gene expression changes. The study also examined the transcriptomic changes of INFγ-preconditioned MSCs to identify the molecular mechanisms behind the varying immunomodulatory potencies of different MSC samples. They found that different groups of MSCs use similar regulation networks in response to inflammatory stimulations, but gene expression variations within those networks result in differences in immunosuppressive capability. These gene expressions were found to vary greatly between MSC samples. Accordingly, the study identified a panel of these responsive genes that can be used to assess the immunosuppressive potency of MSCs.

Generally, whole transcriptome analysis studies enable investigation of the cause of immunomodulatory functional variation in different MSC types at the molecular level. Consequently, it can aid in establishing minimum clinical release criteria in our pursuit to identify the best MSC source with immunomodulatory potency.

3.2 Osteogenesis and chondrogenesis

In accordance with the guidelines set forth by the International Society for Cellular Therapy (ISCT), human MSCs are required to possess the ability to differentiate into three distinct lineages in vitro, namely osteoblasts, adipocytes, and chondroblasts [23]. While all MSCs from different tissue sources must be able to demonstrate osteogenic and chondrogenic differentiation capabilities, variations in their differentiation potential have been extensively studied and reported [8, 84, 85, 86]. These inherent differences have significant implications, particularly when deciding on the source of MSCs to be used in clinical trials for related medical conditions such as cartilage regeneration in patients with knee osteoarthritis. The selection of appropriate MSCs is crucial to ensuring successful outcomes in such clinical trials and the development of effective therapies.

Our previous study, which analyzed the global gene expression profile of MSCs from different sources, revealed that BM-MSCs and AT-MSCs exhibit a higher expression of genes related to osteogenesis. We identified forkhead box C1 (FOXC1) and distal-less homeobox 5 (DLX5) among the upregulated genes in BM-MSCs. FOXC1 plays a crucial role in regulating initial osteoblast differentiation by directly regulating the expression of Msx2, a key regulator of early osteogenic events [87]. Similarly, DLX5 is a transcription factor that plays a role in later stages of osteogenic differentiation and is regulated by bone morphogenic protein 2 (BMP2) signaling. On the other hand, AT-MSCs showed upregulation in the bone morphogenetic protein receptor type 1B (BMPR1B) gene, which binds to BMP ligands and transduces BMP signaling [88]. These results suggest that both BM- and AT-MSCs have a higher capacity to differentiate into osteoblasts than PL-MSCs and UC-MSCs. Our findings are consistent with other studies that analyzed gene expression profiles of MSCs from various sources, which also demonstrated that BM-MSCs have better osteogenic potential than MSCs from other sources [8, 89]. It is worth noting that several studies have reported comparable osteogenic differentiation capabilities of MSCs derived from different sources, such as BM, UC, or AT [90, 91]. However, in this discussion, we are primarily concerned with the inherent osteogenic potential that undoubtedly reflects the osteogenic differentiation capacity of MSCs, although it does not exclude the differentiation capacity of MSCs obtained from other sources.

A recent study conducted by Zhang et al. utilized advanced single-cell RNA sequencing to investigate the similarity and heterogeneity of BM-MSCs and MSCs derived from Wharton’s Jelly [92]. The study revealed a unique gene expression profile for each MSC cell source and identified heterogeneity among MSCs from the same tissue source, with distinct subpopulations of cells being identified. These subpopulations were categorized according to their gene expression patterns. Notably, the study identified a multipotent progenitor subpopulation that had an expression signature enriched for trilineage differentiation, including osteogenic and chondrogenic differentiation. However, the study did not draw any conclusions about which cell source possesses a higher osteogenic or chondrogenic differentiation capability. This study provides valuable insights into the inherent cellular composition of MSCs from different sources. The unique gene expression profiles identified for each cell source highlight the potential differences in their therapeutic applications. Furthermore, the identification of a multipotent progenitor subpopulation with an expression signature enriched for trilineage differentiation is of particular interest for regenerative medicine applications.

The ability of MSCs to differentiate into chondrocytes has made cartilage repair a successful regenerative application of these cells. Among the several bone and cartilage disorders investigated in MSC-based clinical studies, knee osteoarthritis is one of the most extensively studied, with various clinical trials employing MSCs from different sources [93, 94, 95]. Most of these trials have utilized an autologous source of MSCs, either from the BM or AT, and have produced encouraging results, with one study progressing to the phase III stage [96]. Other sources, such as WJ, PL, and amniotic membrane/fluid, have also been explored [96]. Evaluating the inherent chondrogenic and survival potential of the MSC type before its use in clinical settings could minimize the risk of suboptimal cell sourcing, leading to improved patient outcomes.

We have conducted a whole transcriptome analysis of four tissue-specific MSCs and evaluated the expression level of 43 genes related to chondrogenesis [16]. Our results revealed that BM- and AT-MSCs exhibited the highest number of upregulated genes. In BM-MSCs, chondroitin Sulfate N Acetylgalactosaminyltransferase 1 (CSGalNAcT-1), chitinase-3-like 1 protein (CHI3L1), and mothers against decapentaplegic homolog 1 (SMAD1) were specifically upregulated. CSGalNAcT-1 plays a crucial role in initiating cartilage chondroitin sulfate biosynthesis [97], while CHI3L1 is associated with proliferation and differentiation in osteogenic and chondrogenic cell lineages during fetal development and MSC differentiation [98]. SMAD1 is an essential regulator of Smad-dependent signal transduction pathways, which is vital for initiating chondrogenic differentiation [99]. In contrast, transforming growth factor beta-induced (TGFBI), transforming growth factor beta receptor 2 (TGFBR2), and short stature homeobox 2 (SHOX2) were found to be downregulated in all MSCs compared to AT-MSCs. Research has shown that transforming growth factor beta (TGFB) proteins are the most potent activators of chondrogenesis in human MSCs [100]. Inactivation of TGFBR2 in neural crest cells resulted in the aberrant formation of Meckel’s cartilage and altered the development of the mandible [101]. SHOX2 has been shown to control chondrocyte maturation by regulating the expression of Runt-related transcription factor 1 (RUNX) genes through bone morphogenic protein 4 (BMP4) [102]. Therefore, our results propose that BM and AT-MSCs have higher chondrogenic differentiation potential than PL and UC-MSCs. Such findings provide a deeper understanding of the molecular mechanisms involved in chondrogenic differentiation and could potentially inform the development of new regenerative therapies for cartilage repair.

3.3 Extracellular vesicles of MSCs and exosome potential

Besides direct cell-to-cell contact, the therapeutic potential of MSCs is attributed mainly to their paracrine actions [103, 104]. These actions are primarily mediated by the secretion of extracellular vesicles (EVs), particularly exosomes. Exosomes are small EVs with an average size of ~100 nm that are released by the cells into the extracellular environment [105]. They are enriched with bioactive molecules, such as nucleic acids, proteins, and metabolites, with various important biological functions [106]. Exosomes play a crucial role in intercellular communication and can transfer important biomolecules to target cells, thereby modulating various physiological and pathological processes [107, 108]. Studies have demonstrated that EVs of MSCs can exert biological effects comparable to MSCs themselves [109, 110]. As such, the paracrine effects of MSCs through EV secretion have emerged as a promising avenue for developing novel cell-free therapies for multiple diseases.

The unique advantage of EV as an alternative therapy for MSCs stems from the fact that they are cell-free therapies and accordingly require less authority approval due to their superior safety profile and lower immunogenicity [111, 112]. In addition, the viability, longevity, and entrapment of MSCs in the lung microvasculature after implantation hinder their use [113, 114, 115], whereas EVs of MSCs do not follow these limits [103]. Although the therapeutic potential of MSCs-EV has been extensively studied in vivo, its applications in a clinical setting have been poorly investigated [111, 116]. Only a few clinical studies have demonstrated the effectiveness of their use [117, 118, 119].

Given the high potential of exosomes in modulating cellular behaviors and their crucial role in the paracrine action of MSCs, it is imperative to gain a deeper understanding of their function to develop effective exosome-based therapeutics. It has been observed that exosomes derived from different MSC types have varying effects, and different culture conditions also result in exosomes with greater therapeutic effects [103, 120]. Transcriptome analysis of exosome cargo can aid in elucidating key factors that govern their biological activities. Accordingly, such data could lead to the development of more effective therapies. For instance, Yao et al. investigated the effect of exosomes from BM-MSCs on pancreatic cancer cell characteristics [104]. They performed transcriptome sequencing on the BM-MSCs-derived exosomes at different stages and identified, for the first time, the role of circular RNA (circRNA) found in the BM-MSCs-derived exosomes in pancreatic cancer. The circRNA (circ_0030167) was found to be enriched and identified as the main effector molecule of the BM-MSCs-derived exosome. From there, they identified that circ-0030167 inhibited the malignant progression and stemness of pancreatic cancer via the miR-338-5p/wif1/wnt8/β-catenin axis.

Similarly, Liu W et al. employed a high-throughput sequencing approach to study miRNA expression profiling in exosomes derived from human UC-MSCs and murine compact bone MSCs (cm-MSCs) to explore their effects on acute graft versus host disease (aGVHD) [121]. Their findings revealed a high expression level of miRNA-223 and identified this miRNA’s mechanistic action in attenuating aGVHD. In another study by Terunuma A et al., the transcriptomes of EV from DP-MSCS and AT-MSCs were analyzed and compared to the transcriptomes of MSCs for the same tissue types [121]. EVs obtained from DP-MSCs exhibit transcriptomic signatures associated with neurogenesis and neural retinal development, while those obtained from AT-MCSs exhibit signatures related to mitochondrial activity and skeletal system development. Notably, the transcriptomes of EV-derived MSCs closely resemble those of their parent MSCs, and genes associated with neurogenesis were found to be highly expressed in both DP-MSCs and their EVs. Conversely, AT-MSCs and their EVs were found to exhibit high expression of genes associated with angiogenesis, hair growth, and dermal matrices. These findings suggest that EVs derived from DP-MSCs may hold promise as a therapeutic target for neurodegenerative disorders and retinal diseases, while EVs-derived AT-MSCs may be useful in rejuvenating the musculoskeletal system and skin.

The transcriptome studies of EVs derived from MSCs have demonstrated the significance of advanced technologies. Specifically, the analysis of the exosome cargo transcriptome can offer valuable insights into the biological activities of exosomes and facilitate the development of effective exosome-based therapeutics. Further investigation into MSC-EVs from diverse tissue types may broaden the range of potential therapeutic targets and enhance our comprehension of the mechanisms underlying the therapeutic effects of MSC-EVs.

Advertisement

4. Strategies to improve the therapeutic outcome of MSCs

While the current clinical successes of MSC therapies are encouraging, reports of failure and minimum efficacy persist. These limitations may not necessarily stem from the potential of MSCs but rather from their suboptimal use. As evidenced by single-cell RNA-seq studies, MSCs are a heterogeneous group of populations with varying cellular characteristics and potential. Additional heterogeneity in the MSC population could be introduced during the manufacturing process (isolation and expansion) [122, 123]. Standardizing the therapeutic potency of MSCs is therefore crucial to ensuring reliable and reproducible results in clinical trials. This can be achieved by establishing consistent protocols for the selection, isolation, and expansion of MSCs, as well as developing protocols for assessing cell quality and potency prior to clinical application. Furthermore, the automation of the expansion process and large-scale production will result in consistent product quality, thereby eliminating variabilities related to the production process. Namely, isolating a homogenous population of cells with the desired potency characteristics and following standardized protocols for expansion can produce high-quality MSCs for therapeutic use.

Another strategy to enhance the therapeutic potential of MSCs is through priming or preconditioning with specific agents before clinical administration. Such a strategy entails priming (treating) MSCs with small molecules, growth factors, and other biological or chemical agents to boost their reparative and regenerative properties and immunomodulatory capabilities. Preconditioning can be viewed as a way of educating MSCs before clinical administration. The main aim is to circumvent problems of MSC proliferation and in vivo survival and to boost the cells’ therapeutic potential. Depending on the intended clinical use of the MSCs, different agents may be used. For instance, the role of growth factors in promoting the survival of transplanted MSCs has been investigated. In cardiac therapy, preconditioning MSCs with TGF-α for 24 hours before transplantation into ischemic sites in a rat model of myocardial infarction increased cell survival [124]. Additionally, priming MSCs with IFNγ was found to enhance their immunomodulatory potential [92]. Alternatively, MCSs can be genetically engineered to induce the expression of genes with desired therapeutic outcomes, including growth factors, cytokines, enzymes, and microRNA, without interfering with cell differentiation and self-regeneration capabilities [125]. Various strategies can be employed for genetic modification to enhance the therapeutic benefits of MSCs while retaining their inherent properties [125].

Cells in vivo reside within three-dimensional (3D) niches, where cell-matrix and cell-cell interactions play a significant role in maintaining cellular characteristics. The current culture protocols for MSCs rely on two-dimensional (2D) expansion systems, which do not accurately recapitulate the native environment of the cells. Consequently, these 2D cultures induce cellular changes that might impact certain therapeutic characteristics of the cells. Differences in the transcriptome profile of MSCs in 2D and 3D cultures have been reported [126, 127, 128]. These differences encompass the expression levels of growth factors and cytokines, which play an essential role in the paracrine mechanism of MSCs’ protection [127, 129]. Therefore, the employment of 3D culture, which closely mimics the native conditions, could be considered an alternative strategy to maintain the therapeutic potential of MSCs. Alternatively, 3D cultures of MSCs could be viewed as a route of cell administration, particularly if they support MSCs’ proliferation and differentiation [130, 131].

Advertisement

5. Conclusions

The clinical relevance of MSCs from various sources cannot be overstated, as they have demonstrated great promise in treating a wide range of diseases and injuries. MSCs are a heterogeneous population of cells with varying inherent potential. Analyzing the whole transcriptome of MSCs offers a robust tool for identifying gene expression patterns and regulatory networks. Through this approach, researchers can gain insights into the molecular mechanisms that govern the properties and functions of MSCs. Identifying key gene expression signatures can help predict the therapeutic potential of MSCs and optimize their use in clinical settings. This high-end technology could help identify the optimal source of MSCs with the desired inherent potential for specific clinical applications. Furthermore, data obtained from analyses could aid in enhancing the efficacy of clinical applications of MSCs. Additionally, new strategies, such as potency testing, priming of MSCs, and the development of 3D biomimicry conditions, should be deployed to enhance the therapeutic outcomes of MSCs.

Advertisement

Acknowledgments

The authors would like to express their gratitude to Professor Abdalla Awidi for his mentorship and guidance.

Advertisement

Conflict of interest

The authors declare no conflict of interest.

References

  1. 1. Wei X, Yang X, Han Z-P, Qu F-F, Shao L, Shi Y-F. Mesenchymal stem cells: A new trend for cell therapy. Acta Pharmacologica Sinica. 2013;34(6):747-754
  2. 2. Francis MP, Sachs PC, Elmore LW, Holt SE. Isolating adipose-derived mesenchymal stem cells from lipoaspirate blood and saline fraction. Organogenesis. 2010;6(1):11-14
  3. 3. De Coppi P, Bartsch G Jr, Siddiqui MM, Xu T, Santos CC, Perin L, et al. Isolation of amniotic stem cell lines with potential for therapy. Nature Biotechnology. 2007;25(1):100-106
  4. 4. Prockop DJ, Oh JY. Medical therapies with adult stem/progenitor cells (MSCs): A backward journey from dramatic results in vivo to the cellular and molecular explanations. Journal of Cellular Biochemistry. 2012;113(5):1460-1469
  5. 5. Aslam N, Abusharieh E, Abuarqoub D, Hattab D, Jafar H, Alshaer W, et al. An in vitro comparison of anti-tumoral potential of Wharton’s jelly and bone marrow mesenchymal stem cells exhibited by cell cycle arrest in glioma cells (U87MG). Pathology and Oncology Research. 2021;27:584710
  6. 6. Wegmeyer H, Bröske A-M, Leddin M, Kuentzer K, Nisslbeck AK, Hupfeld J, et al. Mesenchymal stromal cell characteristics vary depending on their origin. Stem Cells and Development. 2013;22(19):2606-2618
  7. 7. Phinney DG. Functional heterogeneity of mesenchymal stem cells: Implications for cell therapy. Journal of Cellular Biochemistry. 2012;113(9):2806-2812
  8. 8. Hsieh J-Y, Fu Y-S, Chang S-J, Tsuang Y-H, Wang H-W. Functional module analysis reveals differential osteogenic and stemness potentials in human mesenchymal stem cells from bone marrow and Wharton's jelly of umbilical cord. Stem Cells and Development. 2010;19(12):1895-1910
  9. 9. Karahuseyinoglu S, Kocaefe C, Balci D, Erdemli E, Can A. Functional structure of adipocytes differentiated from human umbilical cord stroma-derived stem cells. Stem Cells. 2008;26(3):682-691
  10. 10. Sun C, Zhang K, Yue J, Meng S, Zhang X. Deconstructing transcriptional variations and their effects on immunomodulatory function among human mesenchymal stromal cells. Stem Cell Research & Therapy. 2021;12(1):1-16
  11. 11. Cho K-A, Park M, Kim Y-H, Woo S-Y, Ryu K-H. RNA sequencing reveals a transcriptomic portrait of human mesenchymal stem cells from bone marrow, adipose tissue, and palatine tonsils. Scientific Reports. 2017;7(1):17114
  12. 12. Tsai MS, Hwang SM, Chen KD, Lee YS, Hsu LW, Chang YJ, et al. Functional network analysis of the transcriptomes of mesenchymal stem cells derived from amniotic fluid, amniotic membrane, cord blood, and bone marrow. Stem Cells. 2007;25(10):2511-2523
  13. 13. Gao LR, Zhang NK, Ding QA, Chen HY, Hu X, Jiang S, et al. Common expression of stemness molecular markers and early cardiac transcription factors in human Wharton's jelly-derived mesenchymal stem cells and embryonic stem cells. Cell Transplantation. 2013;22(10):1883-1900
  14. 14. Nekanti U, Rao VB, Bahirvani AG, Jan M, Totey S, Ta M. Long-term expansion and pluripotent marker array analysis of Wharton’s jelly-derived mesenchymal stem cells. Stem Cells and Development. 2010;19(1):117-130
  15. 15. Yu S, Long J, Yu J, Du J, Ma P, Ma Y, et al. Analysis of differentiation potentials and gene expression profiles of mesenchymal stem cells derived from periodontal ligament and Wharton’s jelly of the umbilical cord. Cells, Tissues, Organs. 2013;197(3):209-223
  16. 16. Alhattab D, Jamali F, Ali D, Hammad H, Adwan S, Rahmeh R, et al. An insight into the whole transcriptome profile of four tissue-specific human mesenchymal stem cells. Regenerative Medicine. 2019;14(9):841-865
  17. 17. Lee OK, Kuo TK, Chen W-M, Lee K-D, Hsieh S-L, Chen T-H. Isolation of multipotent mesenchymal stem cells from umbilical cord blood. Blood. 2004;103(5):1669-1675
  18. 18. Lotfy A, El-Sherbiny YM, Cuthbert R, Jones E, Badawy A. Comparative study of biological characteristics of mesenchymal stem cells isolated from mouse bone marrow and peripheral blood. Biomedical Reports. 2019;11(4):165-170
  19. 19. Bunnell BA, Flaat M, Gagliardi C, Patel B, Ripoll C. Adipose-derived stem cells: Isolation, expansion and differentiation. Methods. 2008;45(2):115-120
  20. 20. Roberto BF, Pablo MO. Sources and clinical applications of mesenchymal stem cells: State-of-the-art review. Sultan Qaboos University Medical Journal. Aug 2018;18(3):e264-e277
  21. 21. Berebichez-Fridman R, Gómez-García R, Granados-Montiel J, Berebichez-Fastlicht E, Olivos-Meza A, Granados J, et al. The holy grail of orthopedic surgery: Mesenchymal stem cells—Their current uses and potential applications. Stem Cells International. 2017;2017:2638305
  22. 22. Williams AR, Hare JM. Mesenchymal stem cells: Biology, pathophysiology, translational findings, and therapeutic implications for cardiac disease. Circulation Research. 2011;109(8):923-940
  23. 23. Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini F, Krause D, et al. Minimal criteria for defining multipotent mesenchymal stromal cells. The international society for cellular therapy position statement. Cytotherapy. 2006;8(4):315-317
  24. 24. Aslam N, Abusharieh E, Abuarqoub D, Ali D, Al-Hattab D, Wehaibi S, et al. Anti-oncogenic activities exhibited by paracrine factors of MSCs can be mediated by modulation of KITLG and DKK1 genes in glioma SCs in vitro. Molecular Therapy-Oncolytics. 2021;20:147-165
  25. 25. Kramer J, Dazzi F, Dominici M, Schlenke P, Wagner W. Clinical perspectives of mesenchymal stem cells. Hindawi. 2012;2012:684827
  26. 26. Yoshimura K, Suga H, Eto H. Adipose-derived stem/progenitor cells: Roles in adipose tissue remodeling and potential use for soft tissue augmentation. Regenerative Medicine. 2009;4:265-273
  27. 27. Zuk PA, Zhu M, Ashjian P, De Ugarte DA, Huang JI, Mizuno H, et al. Human adipose tissue is a source of multipotent stem cells. Molecular Biology of the Cell. 2002;13(12):4279-4295
  28. 28. Sober SA, Darmani H, Alhattab D, Awidi A. Flow cytometric characterization of cell surface markers to differentiate between fibroblasts and mesenchymal stem cells of different origin. Archives of Medical Science: AMS. 2023;19(5):1487
  29. 29. Fan X-L, Zhang Y, Li X, Fu Q-L. Mechanisms underlying the protective effects of mesenchymal stem cell-based therapy. Cellular and Molecular Life Sciences. 2020;77:2771-2794
  30. 30. Hassan WU, Greiser U, Wang W. Role of adipose-derived stem cells in wound healing. Wound Repair and Regeneration. 2014;22(3):313-325
  31. 31. Shingyochi Y, Orbay H, Mizuno H. Adipose-derived stem cells for wound repair and regeneration. Expert Opinion on Biological Therapy. 2015;15:1285-1292
  32. 32. Si Z, Wang X, Sun C, Kang Y, Xu J, Wang X, et al. Adipose-derived stem cells: Sources, potency, and implications for regenerative therapies. Biomedicine & Pharmacotherapy. 2019;114:108765
  33. 33. Ribeiro TB, Duarte AS, Longhini ALF, Pradella F, Farias AS, Luzo AC, et al. Neuroprotection and immunomodulation by xenografted human mesenchymal stem cells following spinal cord ventral root avulsion. Scientific Reports. 2015;5(1):16167
  34. 34. Krawczenko A, Klimczak A. Adipose tissue-derived mesenchymal stem/stromal cells and their contribution to angiogenic processes in tissue regeneration. International Journal of Molecular Sciences. 2022;23(5):2425
  35. 35. Sabol RA, Bowles AC, Côté A, Wise R, Pashos N, Bunnell BA. Therapeutic potential of adipose stem cells. Cell Biology and Translational Medicine. 2018;13:15-25 Stem cells in development and disease
  36. 36. Kim Y-J, Jeong J-H. Clinical application of adipose stem cells in plastic surgery. Journal of Korean Medical Science. 2014;29(4):462-467
  37. 37. Minguell JJ. Umbilical cord-derived mesenchymal stem cells. In: Regenerative Medicine Using Pregnancy-Specific Biological Substances. London: Springer; 2010. pp. 249-253
  38. 38. McElreavey KD, Irvine AI, Ennis KT, McLean WI. Isolation, culture and characterisation of fibroblast-like cells derived from the Wharton's jelly portion of human umbilical cord. Biochemical Society Transactions (Portland Press Ltd.). 1991;19(1):29S
  39. 39. Sarugaser R, Lickorish D, Baksh D, Hosseini MM, Davies JE. Human umbilical cord perivascular (HUCPV) cells: A source of mesenchymal progenitors. Stem Cells. 2005;23(2):220-229
  40. 40. Troyer DL, Weiss ML. Concise review: Wharton's jelly-derived cells are a primitive stromal cell population. Stem Cells. 2008;26(3):591-599
  41. 41. Horie S, Gonzalez H, Brady J, Devaney J, Scully M, O’Toole D, et al. Fresh and cryopreserved human umbilical-cord-derived mesenchymal stromal cells attenuate injury and enhance resolution and repair following ventilation-induced lung injury. International Journal of Molecular Sciences. 2021;22(23):12842
  42. 42. Nagamura-Inoue T, He H. Umbilical cord-derived mesenchymal stem cells: Their advantages and potential clinical utility. World Journal of Stem Cells. 2014;6(2):195
  43. 43. Li CD, Zhang WY, Li HL, Jiang XX, Zhang Y, Tang PH, et al. Mesenchymal stem cells derived from human placenta suppress allogeneic umbilical cord blood lymphocyte proliferation. Cell Research. 2005;15(7):539-547
  44. 44. Li C, Zhang W, Jiang X, Mao N. Human-placenta-derived mesenchymal stem cells inhibit proliferation and function of allogeneic immune cells. Cell and Tissue Research. 2007;330(3):437-446
  45. 45. Atala A. Recent developments in tissue engineering and regenerative medicine. Current Opinion in Pediatrics. 2006;18(2):167-171
  46. 46. Orban B. Oral Histology and Embryology. 2nd ed. St. Louis, MO: CV Mosby; 1949. p. 75
  47. 47. Kawashima N. Characterisation of dental pulp stem cells: A new horizon for tissue regeneration? Archives of Oral Biology. 2012;57(11):1439-1458
  48. 48. Rodas-Junco BA, Villicana C. Dental pulp stem cells: Current advances in isolation, expansion and preservation. Tissue Engineering and Regenerative Medicine. 2017;14(4):333-347
  49. 49. Yang X, Li L, Xiao L, Zhang D. Recycle the dental fairy’s package: Overview of dental pulp stem cells. Stem Cell Research & Therapy. 2018;9:1-8
  50. 50. Sonoyama W, Liu Y, Fang D, Yamaza T, Seo B-M, Zhang C, et al. Mesenchymal stem cell-mediated functional tooth regeneration in swine. PLoS One. 2006;1(1):e79
  51. 51. Makino Y, Yamaza H, Akiyama K, Ma L, Hoshino Y, Nonaka K, et al. Immune therapeutic potential of stem cells from human supernumerary teeth. Journal of Dental Research. 2013;92(7):609-615
  52. 52. Itoh Y, Sasaki J, Hashimoto M, Katata C, Hayashi M, Imazato S. Pulp regeneration by 3-dimensional dental pulp stem cell constructs. Journal of Dental Research. 2018;97(10):1137-1143
  53. 53. Li K, O’Dwyer R, Yang F, Cymerman J, Li J, Feldman JD, et al. Enhancement of acellular biomineralization, dental pulp stem cell migration, and differentiation by hybrid fibrin gelatin scaffolds. Dental Materials. 2023;39(3):305-319
  54. 54. Saez DM, Sasaki RT, Martins DO, Chacur M, Kerkis I, da Silva MCP. Rat facial nerve regeneration with human immature dental pulp stem cells. Cell Transplantation. 2019;28(12):1573-1584
  55. 55. De Bari C, Dell'Accio F, Tylzanowski P, Luyten FP. Multipotent mesenchymal stem cells from adult human synovial membrane. Arthritis and Rheumatism. 2001;44(8):1928-1942
  56. 56. Imanishi Y, Miyagawa S, Kitagawa-Sakakida S, Taketani S, Sekiya N, Sawa Y. Impact of synovial membrane-derived stem cell transplantation in a rat model of myocardial infarction. Journal of Artificial Organs. 2009;12:187-193
  57. 57. Chen K, Man C, Zhang B, Hu J, Zhu S-S. Effect of in vitro chondrogenic differentiation of autologous mesenchymal stem cells on cartilage and subchondral cancellous bone repair in osteoarthritis of temporomandibular joint. International Journal of Oral and Maxillofacial Surgery. 2013;42(2):240-248
  58. 58. Koyama N, Okubo Y, Nakao K, Osawa K, Fujimura K, Bessho K. Pluripotency of mesenchymal cells derived from synovial fluid in patients with temporomandibular joint disorder. Life Sciences. 2011;89(19-20):741-747
  59. 59. Matsukura Y, Muneta T, Tsuji K, Koga H, Sekiya I. Mesenchymal stem cells in synovial fluid increase after meniscus injury. Clinical Orthopaedics and Related Research. 2014;472:1357-1364
  60. 60. Lee W-J, Hah Y-S, Ock S-A, Lee J-H, Jeon R-H, Park J-S, et al. Cell source-dependent in vivo immunosuppressive properties of mesenchymal stem cells derived from the bone marrow and synovial fluid of minipigs. Experimental Cell Research. 2015;333(2):273-288
  61. 61. Ando W, Kutcher JJ, Krawetz R, Sen A, Nakamura N, Frank CB, et al. Clonal analysis of synovial fluid stem cells to characterize and identify stable mesenchymal stromal cell/mesenchymal progenitor cell phenotypes in a porcine model: A cell source with enhanced commitment to the chondrogenic lineage. Cytotherapy. 2014;16(6):776-788
  62. 62. Levy O, Kuai R, Siren EM, Bhere D, Milton Y, Nissar N, et al. Shattering barriers toward clinically meaningful MSC therapies. Science. Advances. 2020;6(30):eaba6884
  63. 63. Krampera M, Le Blanc K. Mesenchymal stromal cells: Putative microenvironmental modulators become cell therapy. Cell Stem Cell. 2021;28(10):1708-1725
  64. 64. Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca JD, et al. Multilineage potential of adult human mesenchymal stem cells. Science. 1999;284(5411):143-147
  65. 65. Shi Y, Wang Y, Li Q , Liu K, Hou J, Shao C, et al. Immunoregulatory mechanisms of mesenchymal stem and stromal cells in inflammatory diseases. Nature Reviews Nephrology. 2018;14(8):493-507
  66. 66. Pittenger MF, Discher DE, Péault BM, Phinney DG, Hare JM, Caplan AI. Mesenchymal stem cell perspective: Cell biology to clinical progress. NPJ Regenerative Medicine. 2019;4(1):22
  67. 67. Klyushnenkova E, Mosca JD, Zernetkina V, Majumdar MK, Beggs KJ, Simonetti DW, et al. T cell responses to allogeneic human mesenchymal stem cells: Immunogenicity, tolerance, and suppression. Journal of Biomedical Science. 2005;12:47-57
  68. 68. Di Nicola M, Carlo-Stella C, Magni M, Milanesi M, Longoni PD, Matteucci P, et al. Human bone marrow stromal cells suppress T-lymphocyte proliferation induced by cellular or nonspecific mitogenic stimuli. Blood, The Journal of the American Society of Hematology. 2002;99(10):3838-3843
  69. 69. Aggarwal S, Pittenger MF. Human mesenchymal stem cells modulate allogeneic immune cell responses. Blood. 2005;105(4):1815-1822
  70. 70. Nasef A, Chapel A, Mazurier C, Bouchet S, Lopez M, Mathieu N, et al. Identification of IL-10 and TGF-β transcripts involved in the inhibition of T-lymphocyte proliferation during cell contact with human mesenchymal stem cells. Gene Expression The Journal of Liver Research. 2006;13(4-5):217-226
  71. 71. Ma S, Xie N, Li W, Yuan B, Shi Y, Wang Y. Immunobiology of mesenchymal stem cells. Cell Death & Differentiation. 2014;21(2):216-225
  72. 72. Akiyama K, Chen C, Wang D, Xu X, Qu C, Yamaza T, et al. Mesenchymal-stem-cell-induced immunoregulation involves FAS-ligand-/FAS-mediated T cell apoptosis. Cell Stem Cell. 2012;10(5):544-555
  73. 73. Le Blanc K, Mougiakakos D. Multipotent mesenchymal stromal cells and the innate immune system. Nature Reviews Immunology. 2012;12(5):383-396
  74. 74. Neofytou E, Deuse T, Beygui RE, Schrepfer S. Mesenchymal stromal cell therapy: Different sources exhibit different immunobiological properties. Transplantation. 2015;99(6):1113-1118
  75. 75. Melief SM, Zwaginga JJ, Fibbe WE, Roelofs H. Adipose tissue-derived multipotent stromal cells have a higher immunomodulatory capacity than their bone marrow-derived counterparts. Stem Cells Translational Medicine. 2013;2(6):455-463
  76. 76. Ivanova-Todorova E, Bochev I, Mourdjeva M, Dimitrov R, Bukarev D, Kyurkchiev S, et al. Adipose tissue-derived mesenchymal stem cells are more potent suppressors of dendritic cells differentiation compared to bone marrow-derived mesenchymal stem cells. Immunology Letters. 2009;126(1-2):37-42
  77. 77. Ghosh AK, Sinha D, Mukherjee S, Biswas R, Biswas T. LPS stimulates and Hsp70 down-regulates TLR4 to orchestrate differential cytokine response of culture-differentiated innate memory CD8+ T cells. Cytokine. 2015;73(1):44-52
  78. 78. Rashedi I, Gómez-Aristizábal A, Wang X-H, Viswanathan S, Keating A. TLR3 or TLR4 activation enhances mesenchymal stromal cell-mediated Treg induction via notch signaling. Stem Cells. 2017;35(1):265-275
  79. 79. Nemoto Y, Kanai T, Takahara M, Oshima S, Nakamura T, Okamoto R, et al. Bone marrow-mesenchymal stem cells are a major source of interleukin-7 and sustain colitis by forming the niche for colitogenic CD4 memory T cells. Gut. 2013;62(8):1142-1152
  80. 80. Amouzegar A, Mittal SK, Sahu A, Sahu SK, Chauhan SK. Mesenchymal stem cells modulate differentiation of myeloid progenitor cells during inflammation. Stem Cells. 2017;35(6):1532-1541
  81. 81. Pietilä M, Lehtonen S, Tuovinen E, Lähteenmäki K, Laitinen S, Leskelä H-V, et al. CD200 positive human mesenchymal stem cells suppress TNF-alpha secretion from CD200 receptor positive macrophage-like cells. PLoS One. 2012;7(2):e31671
  82. 82. Roson-Burgo B, Sanchez-Guijo F, Del Cañizo C, De Las RJ. Transcriptomic portrait of human mesenchymal stromal/stem cells isolated from bone marrow and placenta. BMC Genomics. 2014;15(1):1-18
  83. 83. Jansen BJ, Gilissen C, Roelofs H, Schaap-Oziemlak A, Veltman JA, Raymakers RA, et al. Functional differences between mesenchymal stem cell populations are reflected by their transcriptome. Stem Cells and Development. 2010;19(4):481-490
  84. 84. Bellavia M, Altomare R, Cacciabaudo F, Santoro A, Allegra A, Concetta Gioviale M, et al. Towards an ideal source of mesenchymal stem cell isolation for possible therapeutic application in regenerative medicine. Biomedical Papers. 2014;158(3):356-360
  85. 85. Secunda R, Vennila R, Mohanashankar A, Rajasundari M, Jeswanth S, Surendran R. Isolation, expansion and characterisation of mesenchymal stem cells from human bone marrow, adipose tissue, umbilical cord blood and matrix: A comparative study. Cytotechnology. 2015;67(5):793-807
  86. 86. Fong C-Y, Subramanian A, Gauthaman K, Venugopal J, Biswas A, Ramakrishna S, et al. Human umbilical cord Wharton’s jelly stem cells undergo enhanced chondrogenic differentiation when grown on nanofibrous scaffolds and in a sequential two-stage culture medium environment. Stem Cell Reviews and Reports. 2012;8:195-209
  87. 87. Mirzayans F, Lavy R, Penner-Chea J, Berry FB. Initiation of early osteoblast differentiation events through the direct transcriptional regulation of Msx2 by FOXC1. PLoS One. 2012;7(11):e49095
  88. 88. Wu M, Chen G, Li Y-P. TGF-β and BMP signaling in osteoblast, skeletal development, and bone formation, homeostasis and disease. Bone Research. 2016;4(1):1-21
  89. 89. Mohamed-Ahmed S, Fristad I, Lie SA, Suliman S, Mustafa K, Vindenes H, et al. Adipose-derived and bone marrow mesenchymal stem cells: A donor-matched comparison. Stem Cell Research & Therapy. 2018;9:1-15
  90. 90. Rebelatto CK, Aguiar A, Moretao M, Senegaglia A, Hansen P, Barchiki F, et al. Dissimilar differentiation of mesenchymal stem cells from bone marrow, umbilical cord blood, and adipose tissue. Experimental Biology and Medicine. 2008;233(7):901-913
  91. 91. Jo CH, Yoon PW, Kim H, Kang KS, Yoon KS. Comparative evaluation of in vivo osteogenic differentiation of fetal and adult mesenchymal stem cell in rat critical-sized femoral defect model. Cell and Tissue Research. 2013;353(1):41-52
  92. 92. Zhang C, Han X, Liu J, Chen L, Lei Y, Chen K, et al. Single-cell transcriptomic analysis reveals the cellular heterogeneity of mesenchymal stem cells. Genomics, Proteomics & Bioinformatics. 2022;20(1):70-86
  93. 93. Lee W-S, Kim HJ, Kim K-I, Kim GB, Jin W. Intra-articular injection of autologous adipose tissue-derived mesenchymal stem cells for the treatment of knee osteoarthritis: A phase IIb, randomized, placebo-controlled clinical trial. Stem Cells Translational Medicine. 2019;8(6):504-511
  94. 94. Al-Najar M, Khalil H, Al-Ajlouni J, Al-Antary E, Hamdan M, Rahmeh R, et al. Intra-articular injection of expanded autologous bone marrow mesenchymal cells in moderate and severe knee osteoarthritis is safe: A phase I/II study. Journal of Orthopaedic Surgery and Research. 2017;12:1-6
  95. 95. Samara O, Jafar H, Hamdan M, Al-Ta’mari A, Rahmeh R, Hourani B, et al. Ultrasound-guided intra-articular injection of expanded umbilical cord mesenchymal stem cells in knee osteoarthritis: A safety/efficacy study with MRI data. Regenerative Medicine. 2022;17(5):299-312
  96. 96. Negoro T, Takagaki Y, Okura H, Matsuyama A. Trends in clinical trials for articular cartilage repair by cell therapy. NPJ Regenerative Medicine. 2018;3(1):17
  97. 97. Sakai K, Kimata K, Sato T, Gotoh M, Narimatsu H, Shinomiya K, et al. Chondroitin sulfate N-acetylgalactosaminyltransferase-1 plays a critical role in chondroitin sulfate synthesis in cartilage. Journal of Biological Chemistry. 2007;282(6):4152-4161
  98. 98. Hoover DJ, Zhu V, Chen R, Briley K, Rameshwar P, Cohen S, et al. Expression of the chitinase family glycoprotein YKL-40 in undifferentiated, differentiated and trans-differentiated mesenchymal stem cells. PLoS One. 2013;8(5):e62491
  99. 99. Cleary MA, van Osch GJM, Brama PA, Hellingman CA, Narcisi R. FGF, TGFβ and Wnt crosstalk: Embryonic to in vitro cartilage development from mesenchymal stem cells. Journal of Tissue Engineering and Regenerative Medicine. 2015;9(4):332-342
  100. 100. Green JD, Tollemar V, Dougherty M, Yan Z, Yin L, Ye J, et al. Multifaceted signaling regulators of chondrogenesis: Implications in cartilage regeneration and tissue engineering. Genes & Diseases. 2015;2(4):307-327
  101. 101. Oka K, Oka S, Sasaki T, Ito Y, Bringas P Jr, Nonaka K, et al. The role of TGF-β signaling in regulating chondrogenesis and osteogenesis during mandibular development. Developmental Biology. 2007;303(1):391-404
  102. 102. Yu L, Liu H, Yan M, Yang J, Long F, Muneoka K, et al. Shox2 is required for chondrocyte proliferation and maturation in proximal limb skeleton. Developmental Biology. 2007;306(2):549-559
  103. 103. Han Y, Yang J, Fang J, Zhou Y, Candi E, Wang J, et al. The secretion profile of mesenchymal stem cells and potential applications in treating human diseases. Signal Transduction and Targeted Therapy. 2022;7(1):92
  104. 104. Yao X, Mao Y, Wu D, Zhu Y, Lu J, Huang Y, et al. Exosomal circ_0030167 derived from BM-MSCs inhibits the invasion, migration, proliferation and stemness of pancreatic cancer cells by sponging miR-338-5p and targeting the Wif1/Wnt8/β-catenin axis. Cancer Letters. 2021;512:38-50
  105. 105. Kalluri R, LeBleu VS. The biology, function, and biomedical applications of exosomes. Science. 2020;367(6478):eaau6977
  106. 106. McAndrews KM, Kalluri R. Mechanisms associated with biogenesis of exosomes in cancer. Molecular Cancer. 2019;18(1):52
  107. 107. Emanueli C, Shearn AI, Angelini GD, Sahoo S. Exosomes and exosomal miRNAs in cardiovascular protection and repair. Vascular Pharmacology. 2015;71:24-30
  108. 108. Liao FL, Tan L, Liu H, Wang JJ, Ma XT, Zhao B, et al. Hematopoietic stem cell-derived exosomes promote hematopoietic differentiation of mouse embryonic stem cells in vitro via inhibiting the miR126/Notch1 pathway. Acta Pharmacologica Sinica. 2018;39(4):552-560
  109. 109. Nawaz M, Fatima F, Vallabhaneni KC, Penfornis P, Valadi H, Ekström K, et al. Extracellular vesicles: Evolving factors in stem cell biology. Stem Cells International. 2016;2016:1073140
  110. 110. Shao L, Zhang Y, Lan B, Wang J, Zhang Z, Zhang L, et al. MiRNA-sequence indicates that mesenchymal stem cells and exosomes have similar mechanism to enhance cardiac repair. BioMed Research International. 2017;2017:4150705
  111. 111. Gowen A, Shahjin F, Chand S, Odegaard KE, Yelamanchili SV. Mesenchymal stem cell-derived extracellular vesicles: Challenges in clinical applications. Frontiers in Cell and Developmental Biology. 2020;8:149
  112. 112. Yeo RWY, Lai RC, Zhang B, Tan SS, Yin Y, Teh BJ, et al. Mesenchymal stem cell: An efficient mass producer of exosomes for drug delivery. Advanced Drug Delivery Reviews. 2013;65(3):336-341
  113. 113. Wang S, Guo L, Ge J, Yu L, Cai T, Tian R, et al. Excess integrins cause lung entrapment of mesenchymal stem cells. Stem Cells. 2015;33(11):3315-3326
  114. 114. Badillo AT, Beggs KJ, Javazon EH, Tebbets JC, Flake AW. Murine bone marrow stromal progenitor cells elicit an in vivo cellular and humoral alloimmune response. Biology of Blood and Marrow Transplantation. 2007;13(4):412-422
  115. 115. Tran C, Damaser MS. Stem cells as drug delivery methods: Application of stem cell secretome for regeneration. Advanced Drug Delivery Reviews. 2015;82:1-11
  116. 116. Moghadasi S, Elveny M, Rahman HS, Suksatan W, Jalil AT, Abdelbasset WK, et al. A paradigm shift in cell-free approach: The emerging role of MSCs-derived exosomes in regenerative medicine. Journal of Translational Medicine. 2021;19(1):1-21
  117. 117. Nassar W, El-Ansary M, Sabry D, Mostafa MA, Fayad T, Kotb E, et al. Umbilical cord mesenchymal stem cells derived extracellular vesicles can safely ameliorate the progression of chronic kidney diseases. Biomaterials Research. 2016;20(1):1-11
  118. 118. Sun Y, Shi H, Yin S, Ji C, Zhang X, Zhang B, et al. Human mesenchymal stem cell derived exosomes alleviate type 2 diabetes mellitus by reversing peripheral insulin resistance and relieving β-cell destruction. ACS Nano. 2018;12(8):7613-7628
  119. 119. Kordelas L, Rebmann V, Ludwig A, Radtke S, Ruesing J, Doeppner T, et al. MSC-derived exosomes: A novel tool to treat therapy-refractory graft-versus-host disease. Leukemia. 2014;28(4):970-973
  120. 120. Liu W, Li L, Rong Y, Qian D, Chen J, Zhou Z, et al. Hypoxic mesenchymal stem cell-derived exosomes promote bone fracture healing by the transfer of miR-126. Acta Biomaterialia. 2020;103:196-212
  121. 121. Liu W, Zhou N, Liu Y, Zhang W, Li X, Wang Y, et al. Mesenchymal stem cell exosome-derived miR-223 alleviates acute graft-versus-host disease via reducing the migration of donor T cells. Stem Cell Research & Therapy. 2021;12:1-16
  122. 122. Juneja SC, Viswanathan S, Ganguly M, Veillette C. A simplified method for the aspiration of bone marrow from patients undergoing hip and knee joint replacement for isolating mesenchymal stem cells and in vitro chondrogenesis. Bone Marrow Research. 2016;2016:3152065
  123. 123. De Wolf C, Van De Bovenkamp M, Hoefnagel M. Regulatory perspective on in vitro potency assays for human mesenchymal stromal cells used in immunotherapy. Cytotherapy. 2017;19(7):784-797
  124. 124. Herrmann JL, Wang Y, Abarbanell AM, Weil BR, Tan J, Meldrum DR. Preconditioning mesenchymal stem cells with transforming growth factor-alpha improves mesenchymal stem cell-mediated cardioprotection. Shock. 2010;33(1):24-30
  125. 125. Damasceno PKF, De Santana TA, Santos GC, Orge ID, Silva DN, Albuquerque JF, et al. Genetic engineering as a strategy to improve the therapeutic efficacy of mesenchymal stem/stromal cells in regenerative medicine. Frontiers in Cell and Developmental Biology. 2020;8:737
  126. 126. Kim B-C, Kwack KH, Chun J, Lee J-H. Comparative transcriptome analysis of human adipose-derived stem cells undergoing osteogenesis in 2D and 3D culture conditions. International Journal of Molecular Sciences. 2021;22(15):7939
  127. 127. Alhattab DM, Isaioglou I, Alshehri S, Khan ZN, Susapto HH, Li Y, et al. Fabrication of a three-dimensional bone marrow niche-like acute myeloid Leukemia disease model by an automated and controlled process using a robotic multicellular bioprinting system. Biomaterials Research. 2023;27(1):111
  128. 128. Gallo A, Cuscino N, Contino F, Bulati M, Pampalone M, Amico G, et al. Changes in the transcriptome profiles of human amnion-derived mesenchymal stromal/stem cells induced by three-dimensional culture: A potential priming strategy to improve their properties. International Journal of Molecular Sciences. 2022;23(2):863
  129. 129. Chang C, Yan J, Yao Z, Zhang C, Li X, Mao HQ. Effects of mesenchymal stem cell-derived paracrine signals and their delivery strategies. Advanced Healthcare Materials. 2021;10(7):2001689
  130. 130. Alhattab DM, Khan Z, Alshehri S, Susapto HH, Hauser CA. 3D bioprinting of ultrashort self-assembling peptides to engineer scaffolds with different matrix stiffness for chondrogenesis. International Journal of Bioprinting. 2023;9(4):719
  131. 131. Alshehri S, Susapto HH, Hauser CA. Scaffolds from self-assembling tetrapeptides support 3D spreading, osteogenic differentiation, and angiogenesis of mesenchymal stem cells. Biomacromolecules. 2021;22(5):2094-2106

Written By

Dana M. Alhattab, Salwa Alshehri and Fatima Jamali

Submitted: 01 December 2023 Reviewed: 04 December 2023 Published: 21 December 2023