Open access peer-reviewed chapter

A Closer Look at Mesenchymal Stem Cells (MSCs), Their Potential and Function as Game-Changers of Modern Medicine

Written By

Mohammad Ali Khodadoust, Amirreza Boroumand, Alireza Sedaghat, Hamidreza Reihani, Najmeh Kaffash Farkhad and Jalil Tavakol Afshari

Submitted: 21 September 2023 Reviewed: 01 October 2023 Published: 07 December 2023

DOI: 10.5772/intechopen.1003599

From the Edited Volume

Recent Update on Mesenchymal Stem Cells

Khalid Ahmed Al-Anazi

Chapter metrics overview

47 Chapter Downloads

View Full Metrics

Abstract

Mesenchymal stem cells (MSCs) have garnered significant attention in modern medicine as a potentially effective therapeutic intervention, owing to their distinctive characteristics, including the ability to self-renew, exhibit multipotency, elicit immunomodulatory effects, and promote tissue repair. MSCs are being studied extensively for their potential use in a wide range of clinical applications, including regenerative medicine, immunotherapy, and tissue engineering. In this chapter, we provide a comprehensive overview of the biology, potential, and function of MSCs, highlighting their role in modulating the immune system, promoting tissue repair, and restoring homeostasis in various disease conditions. We also discuss the challenges and limitations associated with MSC-based therapies, including issues related to their isolation, expansion, and delivery. Further research is needed to fully understand the mechanisms underlying MSCs’ therapeutic effects and to optimize their clinical application. Nevertheless, MSCs hold great promise as game-changers in modern medicine, and their potential to revolutionize the field of regenerative medicine and immunotherapy cannot be overlooked.

Keywords

  • mesenchymal stem cells
  • regenerative medicine
  • tissue repair
  • multipotency
  • immunotherapy

1. Introduction

In the 1960s, scientists led by Friedenstein initially identified a population of fibroblast-like cells in bone marrow tissue that had the unique ability to renew themselves. Ever since this landmark discovery, these cells, now known as mesenchymal stem/stromal cells, have been the target of extensive research efforts due to their intriguing characteristics [1]. MSCs represent a subset of adult stem cells distinguished by their potential to develop into different cell lineages, their self-propagating capacity, and immunoregulatory functions [2]. Their proven capacity to mature into cell types like osteocytes, chondrocytes, and adipocytes makes them highly valuable for promoting tissue repair and regeneration. Further research has shown that MSCs secrete a variety of biomolecules like growth factors, cytokines, and extracellular vehicles (EVs) involved in healing processes [3].

MSCs show great potential in the regenerative medicine field due to their unique traits and therapeutic capabilities. These cells, found in tissues like bone marrow (BM), fat, umbilical cord (UC), and dental pulp, hold significant promise for addressing many diseases and injuries [2]. Scholars have extensively investigated MSC isolation and expansion methods to benefit from their therapeutic properties. Researchers have created various isolation techniques like density centrifugation, adherence-based selection, and fluorescence-activated sorting [4]. These approaches help capture a population enriched with MSCs, which can then be grown in culture. Standardizing isolation protocols and characterization standards is crucial to make results across labs and clinical applications consistent and comparable. One remarkable MSC characteristic is immunomodulation. MSCs can alter immune responses by regulating various immune cell activities. They suppress the proliferation and functions of T cells, B cells, and natural killer (NK) cells, while promoting regulatory T cell (Treg) expansion. This immunosuppressive ability benefits autoimmune disorders, graft versus host disease (GVHD), and organ transplant treatment by curbing excessive immunity and fostering tolerance [5].

The therapeutic applications of MSCs and their derivatives like exosomes span a wide range of medical disciplines [6]. In orthopedics, MSCs have shown promise in regenerating bone and cartilage tissues, offering potential alternatives to traditional approaches such as joint replacements [7]. In cardiovascular medicine, MSCs have been investigated for their ability to improve cardiac function and promote blood vessel formation, holding the potential for treating heart failure and ischemic conditions [8]. Additionally, MSC-based therapies have shown encouraging results in neurological disorders, autoimmune diseases, and tissue injuries, demonstrating their versatility and potential to revolutionize patient care [9]. Due to these key characteristics, MSCs hold tremendous promise for regenerative medicine applications.

Advertisement

2. Biology and characteristics of MSCs

The International Society for Cellular Therapy (ISCT) defines human MSCs as fibroblast-like, plastic-adherent cells that express surface markers CD73, CD90 and CD105. At the same time, they lack expression of CD45, CD34, CD14 or CD11b, CD79a or CD19, and HLA-DR. According to ISCT guidelines, the ability to differentiate into osteoblasts, adipocytes, and chondroblasts when cultured in vitro is another key characteristic used to identify MSCs [10]. However, self-renewal – the process by which a single stem cell divides to produce at least one new identical stem cell – is a fundamental property of all stem cells. The ability to proliferate and generate clonal progeny through successive cell divisions while maintaining the potential to differentiate is crucial for stem cell characterization and relies on self-renewal capability [11]. Importantly, it should be noted that the expression of surface markers on MSCs may fluctuate depending on the specific tissue they are derived from, in vitro expansion protocols, and the culture conditions used. Careful phenotypic analysis is therefore required to accurately characterize MSC populations [2]. For example, the surface marker CD105, also known as endoglin and a component of the TGF-β receptor complex, has been shown to be expressed on mesenchymal stem cells derived from various tissues but with differing intensities. Research findings indicate that over 50% of liver-derived MSCs are positive for CD105, whereas less than 10% of MSCs from the amniotic membrane (AM-MSCs) and UC (UC-MSCs) express this antigen. Additionally, liver-derived MSCs were reported to have a low-level expression (5–10%) of CD45, a lineage marker that the majority of MSCs typically lack [12]. In a separate study comparing mesenchymal stem cells derived from BM (BM-MSCs) to those obtained from menstrual blood (referred to as MenSCs), distinct expression profiles were observed between the two populations. The research found that although both BM-MSCs and MenSCs lacked surface markers associated with pluripotency, namely SSEA-3, SSEA-4, and TRA-1-60, MenSCs demonstrated high levels of expression of HLA-ABC and CD49α [13]. So, proper identification of MSCs necessitates controlling for variables that can potentially impact marker expression. In terms of origin, MSCs have been identified in multiple tissues throughout the body, with sources comprising BM, adipose tissue, UC, and dental pulp, among others. Of these, MSCs derived from BM have undergone significant research and are viewed as the archetypal MSC population. However, it is important to recognize that MSCs from diverse tissues can demonstrate differences in their attributes and responses. Studies have highlighted variations in the properties of MSCs depending on their exact anatomical source. For example, a comparative study investigated the properties of MSCs isolated from BM, adipose tissue (AD-MSCs), placenta (P-MSCs), and cord blood (CB-MSCs). While all MSC types exhibited similarities in proliferation capacity, clonogenic potential (ability to form colonies), and surface marker expression, BM-MSCs and AD-MSCs demonstrated stronger osteogenic differentiation ability as determined by von Kossa staining. BM-MSCs and AD-MSCs also expressed higher, more consistent levels of the DLX-5 gene known to regulate osteogenesis [14]. Additionally, the research reported that CB-MSCs and P-MSCs lacked osteogenic potential and exhibited poor adipogenic differentiation as well [14]. Similar findings were reported in another study that compared the in vivo bone-forming capacity of MSCs derived from different tissues. Although BM-MSCs successfully generated bone tissue under the experimental conditions, AD-MSCs, UC-MSCs, and skin-derived MSCs did not exhibit the same ability to produce bone [15]. The results mirrored previous research indicating that while MSCs share a multilineage potential profile, their precise anatomical source can influence the extent of osteogenic differentiation, with BM-MSCs demonstrating superior skeletal lineage commitment ability compared to MSCs from adipose tissue, UC, and skin in this experimental setup [15].

Additionally, MSCs appear as spindle-shaped cells when observed under a microscope. They possess elongated and slender cytoplasmic extensions, enabling them to interact with their microenvironment and neighboring cells. This morphology allows MSCs to migrate to sites of injury or inflammation efficiently and participate in tissue repair processes [16].

Advertisement

3. Functional properties of MSCs

MSCs have an array of traits supporting their usefulness in regenerative medicine. Their immunomodulatory influences, capacity for tissue restoration and regeneration, the potential to evolve into numerous cell types, and role in regulating the microenvironment collectively underlie their therapeutic power.

3.1 Immunomodulatory effects of MSCs

The immunomodulatory function of MSCs involves complex interactions between cell-cell contact mechanisms and soluble factor secretion to regulate immune homeostasis. Both modes of action work synergistically to influence immune cell behavior. Their impact on immune signaling pathways is another key feature of MSCs in immunomodulatory function [17], which is explained with more detail below:

3.1.1 Cell-to-cell contact mechanisms

Direct contact between MSCs and immune cells allows interaction through cell surface molecules. MSC expression of indoleamine 2,3-dioxygenase (IDO) depletes the amino acid tryptophan locally, which inhibits T cell proliferation and activation while promoting Treg cell development [18]. Upregulation of programmed death ligand 1(PD-L1) engages with programmed cell death protein 1 (PD-1) on T cells, hindering their activation and inducing apoptosis. Fas ligand (FasL) expression carried out apoptosis of activated T cells through the Fas receptor, diminishing T cell responses [19]. Additionally, cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) augmentation enables outcompeting co-stimulatory molecules on antigen-presenting cells (APCS) for binding, interrupting signals necessary for T cell activation [20]. Collectively, these membrane-bound pathways facilitate MSC modulation of multiple localized immune cell subsets.

3.1.2 Soluble factor-mediated mechanisms

MSCs demonstrate potent immunomodulation by secreting a diverse array of soluble factors like cytokines, chemokines, growth factors and EVs. These factors act autonomously and on neighboring cells to shape immune responses. Transforming growth factor-beta (TGF-β) released from MSCs strongly suppresses immunity, affecting T cell activity, regulatory T cell development, and natural killer/dendritic cell function. Prostaglandin E2 (PGE2) similarly governs T cell behavior and Treg amplification while transitioning the cytokine profile to anti-inflammation [21]. Interleukin-10 (IL-10) blocks proinflammatory cytokine generation and bolsters Treg cell’s function. Hepatocyte growth factor (HGF) impacts T cell proliferation/dendritic cells and enables repair [22]. EVs such as exosomes transport molecules like TGF-β, PGE2, and IL-10 between MSCs and immune cells to regulate recipient responses. Collectively, these secreted components downscale excessive immune activation through coordinated suppression of proinflammatory signaling and augmentation of regulatory networks [23].

3.1.3 Modulation of immune signaling pathways

MSCs wield robust immunoregulation by targeting major networks governing immune initiation and inflammation. Precisely adjusting these pathways optimizes immune balance:

  • Nuclear factor-kappa β (NF-κβ) signaling coordinates proinflammatory reactions. MSCs curb and dampens inflammatory triggers, shielding tissues from collateral harm [24].

  • The Janus kinase-signal transducer/activator of transcription (JAK-STAT) cascade strengthens cytokine and growth factor signaling, impacting immune effectors. MSCs suppress JAK-STAT transmission, soothing inflammatory mediator synthesis, and immune cell behavior globally [25, 26].

  • Toll-like receptor (TLR) engagement alerts cells to microbe detection, kicking off innate activation. MSCs deter TLR relay, easing downstream proinflammatory cytokine induction. This directly confronts inflammatory triggers at multiple receptors [27].

  • Notch intercellular communication decides cell identity crucial for immune cell development. MSCs interact with Notch receptors on immune targets, steering differentiation toward regulatory phenotypes through guided differentiation [28].

  • Additional influenced pathways include Wnt/β-catenin signaling hindered by MSCs. This pathway promotes survival but also proinflammatory cytokine synthesis when awry. Via multiple membrane and exosome proteins, MSCs achieve these functional suppressions [29].

Yet, studies showed MSC groups vary quantitatively in immune regulation strength and evasion depending on the source. For example, a study contrasting bone marrow and fat tissue MSCs found fat MSCs exerted stronger, less immunogenic effects. When exposed to low interferon-gamma (IFN-γ), fat MSCs made higher IDO versus bone marrow MSCs. Activated fat MSCs also exhibited lower HLA-DR and higher CD55 [30]. These enabled more efficient immune bypasses. Following activation, fat MSCs better halted T cell proliferation than bone marrow MSCs. In contrast, bone marrow MSCs suppressed natural killer cell proliferation more [30]. Another study assessed BM, fat, and Wharton’s jelly MSC (WJ-MSC) immunomodulation – a co-culture combined each MSC type with immune cells, where fat MSCs most potently hindered proliferation. Additionally, WJ-MSCs uniquely failed to curb activated B cell and NK cell multiplication [1].

3.2 Tissue repair and regenerative potential of MSCs

Tissue damage and degenerative diseases are major health challenges worldwide, necessitating the development of innovative therapeutic strategies. MSCs, found in various tissues, hold great promise for tissue repair and regeneration [31]. In this section, we will discuss the role of MSCs in tissue repair in the context of different diseases.

3.2.1 Musculoskeletal disorders

Research shows MSCs demonstrate excellent potential for treating musculoskeletal disorders like osteoarthritis, tendon injuries, and bone breaks. In osteoarthritis, MSCs can mature into cartilage cells and promote repair. Additionally, their immunomodulation helps reduce inflammation and pain from joint degeneration [32]. For bone defects, MSCs aid regeneration by maturing into bone-forming cells and boosting the recruitment of other formative cells. They also play a pivotal role in remodeling the extracellular matrix (ECM), which is vital for repairing such conditions. MSCs make and secrete ECM constituents such as collagens, proteoglycans, and glycosaminoglycans. By synthesizing and depositing these ECM components, MSCs fortify repaired musculoskeletal tissue structure and functionality [7].

3.2.2 Cardiovascular disorders

Research shows MSC-based therapies hold promise for heart conditions like heart attack and clogged arteries. MSCs encourage new blood vessel growth by releasing an array of pro-angiogenic factors including vascular endothelial growth factor (VEGF), fibroblast growth factor (FGF), insulin-like growth factor (IGF), and hepatocyte growth factor [8]. These stimulate endothelial cell proliferation, migration, and tube formation, vital steps in blood vessel formation that aid angiogenesis in cardiovascular disease. MSCs may also decrease scar tissue and improve heart function by secreting anti-fibrotic factors. They secrete substances inhibiting extracellular matrix component formation, especially collagen, contributing to scars. These include HGF and Matrix metalloproteinases (MMPs) that can degrade excess collagen plus TGF-β blockers [33]. Additionally, MSCs can govern fibroblast and highly contractile myofibroblast behavior, which contributes to scar tightening. Released factors inhibit fibroblast activation, proliferation, and transitioning to myofibroblasts. This activity regulates scar formation. MSCs can also mature into endothelial and smooth muscle cells, assisting cardiac repair [34].

3.2.3 Neurological disorders

Neurological issues like stroke, spinal cord injury, and neurodegenerative diseases have limited treatment due to the nervous system’s low regenerative capacity. MSCs can develop into neural cells such as neurons, astrocytes, and oligodendrocytes. Administered to the central nervous system (CNS), MSCs integrate damaged areas, replacing lost or nonfunctional cells to promote repair and regrowth. They secrete various neurotrophic factors including brain-derived neurotrophic factor (BDNF), nerve growth factor (NGF), and glial cell line-derived neurotrophic factor (GDNF) supporting existing neuron survival and growth [35]. These stimulate brain angiogenesis and new neural connections. MSC-secreted neurotrophins enhance neuronal survival and functional recovery. MSCs also possess immunomodulation, curbing inflammatory responses in the central nervous system by inhibiting microglia and T cell activation/proliferation and cytokine production. Reducing neuroinflammation favors neural recovery while decreasing secondary neuron damage. Moreover, MSCs activate resident brain stem and progenitor cells, aiding their maturation into functional neurons or other neural cells to boost endogenous repair mechanisms, contributing to neuronal recovery and tissue restoration [36].

By governing glial cell behavior and transitioning them to a more protective phenotype, MSCs assist inflammatory regulation, lessen glial scar formation, and build a supportive environment for neuronal survival and repair. These properties make them attractive candidates for treating neurological disorders and spinal cord injuries [37].

3.2.4 Autoimmune disorders

MSCs have a powerful ability to modulate immune responses, making them beneficial for treating conditions caused by abnormal immune activation like GVHD, COVID-19, and inflammatory bowel disease (IBD). MSCs can dampen disproportionate immune reactions, balance proinflammatory and anti-inflammatory cytokine levels, and spur tissue healing by adjusting the local microenvironment [38]. MSCs have the adaptability to evolve into various cell types, including mesodermal derivatives such as fat, cartilage, and bone cells, plus non-mesodermal varieties including nerve and liver cells. In autoimmune diseases where tissue is harmed, MSCs can mature into specific cell types needed for repair and regrowth [39]. For example, in an autoimmune arthritis, MSCs can become cartilage cells to further mend cartilage. In autoimmune liver conditions, MSCs can evolve into liver cells to facilitate organ regrowth [40].

Table 1 shows a summary of the therapeutic application of MSCs in the mentioned diseases in some clinical trials.

Disease’s categoryDiseaseTrial phaseSample sizeMSC sourceMSC doseMain outcomeRef
OsteoarthritisPhase I/II15BM-MSCs40.9 × 106 ± 0.4×106Significant improvement in parameters including bodily pain, physical role, and physical functioning[41]
Musculoskeletal disordersBone defectsphase IIa18BM-MSCs15 ± 4.5 × 106Significant clinical improvement in patients.[42]
Lateral epicondylosisPilot study6AD-MSCs106 or 107 cellsSignificant decrease in patients’ VAS & increase in elbow performance scores[43]
Severe ischemic heart failurePhase II60BM-MSCsNMSignificant improvement in myocardial function[34]
Cardiovascular diseasesMyocardial infarctionPhase II116WJ-MSC6 × 106 cellsSignificant improvement in LVEF, and the myocardial viability (PET) and perfusion within the infarcted territory (SPECT) was observed.[44]
AtaxiaNM24UC-MSCs1 × 106 cell/kg BWImprovement in patient’s movement and quality of life with delay in desease progression.[45]
Neurological disordersALSPhase I/II15BM-MSCs1 × 106 cell/kg BWImprovement in patient’s status, ALS-FRS & FVC[36]
MSPhase I/II160BM-MSCs1–2 × 106 cell/kg BWMSC transplantation was safe and well tolerable by patients and showed improvement in EDSS score.[46]
MSAPhase I/II24AD-MSCsTwo doses (1 × 107 and 1 × 108 cell/kg BW)Significant decrease in the rate of disease progression (UMSARS)[47]
Refractory luminal Crohn’s diseasePhase I10BM-MSCs1–2 × 106 cell/kg BWSignificant decrease in patient’s CDAI[48]
Autoimmune diseasesCOVID-19Phase I20UC-MSCs1 × 106Cell/kg BWImprovement in patient’s oxygenation and significant decrease in CRP and inflammatory cytokines[49, 50]
GVHDNM11BM-MSCsNMSignificant decrease in TH17 cells, but increase in Treg cells and a shift toward TH2-cell responses[51]

Table 1.

Therapeutic application of MSCs in various disorders.

Abbreviations: MSC, Mesenchymal stem cells; BM-MSCs, Bone Marrow derived Mesenchymal stem cells; AD-MSCs, Adipose-derived Mesenchymal stem cells; WJ-MSC, Wharton’s jelly-derived mesenchymal stem cells; VAS, patients’ visual analog scale; UC-MSCs, Umbilical cord-derived Mesenchymal stem cells; LVEF, Left Ventricular Ejection Fraction; ALS, Amyotrophic Lateral Sclerosis; MSA, Multiple System Atrophy; UMSARS, Unified Multiple System Atrophy Rating Scale; MS, Multiple sclerosis; EDSS, Expanded Disability Status Scale; GVHD, Graft-versus-host disease; CRP, C-reactive protein; CDAI, Crohn’s disease activity index; TH17, T helper 17; Treg, T regulatory cell; NM, Not Mentioned.

Advertisement

4. MSCs and exosomes

MSCs have been extensively studied in clinical trials due to their diverse roles including tissue repair, anti-inflammation, immunosuppression, and neuroprotection. Initially, it was believed that MSCs traveled to injury sites, differentiated, and replaced damaged cells. However, later research showed MSC engraftment and differentiation at injuries is brief and limited [52, 53]. Current research suggests MSCs mainly act through secreting trophic factors. Some propose exosomes mediate MSC cell communication as they are abundantly secreted by MSCs [54].

Exosomes are tiny lipid bilayer vesicles formed through endocytosis and budding from late endosomes, released when multivesicular bodies fuse with the plasma membrane. They release from various cells including plasma, breast milk, serum, and cerebral fluid (CSF) under inflammation, disease, or immune imbalance [6, 55]. Exosomes have potential to treat infections like COVID-19, cancers, and neurodegeneration supported by evidence. Originally discovered in the 1980s studying sheep blood cell maturation, exosomes garnered attention as key communicators [56]. Though believed to dispose of proteins, exosomes stimulate immunity in vivo and in vitro, secreting from nearly all body cells found in fluids affecting lung, kidney, and liver functions [57, 58].

Exosomes uniquely carry a diverse cargo of lipids, proteins, DNAs, and RNAs specific to their cellular origins, regulating immunity and communication, unlike other extracellular vesicles. Exosomes are characterized by membrane proteins, such as Alix, TSG101, Rab5, Rab27a, Rab27b, and multiple tetraspanins (CD37, CD63, CD81, and CD82), distinguishing them from apoptotic bodies [59]. Exosomal mRNA or miRNA transported from donor cells may alter recipient cell fates. For example, brain cancer or regulatory immune cell exosomes impacted recipient cells [59, 60]. Figure 1 schematically illustrates MSC exosome structures, contents, and target cells.

Figure 1.

Biogenesis, release, and uptake of exosomes (the figure is created using Adobe illustrator 2019).

MSC exosomes are considered MSCs’ paracrine effectors with comparable functions to MSCs. For instance, both alleviated neuroprotection and endotoxin-induced lung injury [61]. MSC exosomes contained miR-16 downregulating VEGF similarly to MSCs [62]. MSC exosomes express MSC antigens and adhesion molecules such as CD29, CD44, and CD73. Moreover, like MSCs, MSC exosomes contain genetic information for signaling pathways [63, 64].

Advertisement

5. Challenges and limitations in MSC therapy

There are currently some obstacles tied to using stem cells for medical functions because the way transplanted cells act is not fully decoded. Still, comprehending stem cell traits, behavior, and mechanisms more profoundly could facilitate customized treatments for many sicknesses. Before meshing stem cells with recipient tissues for lifelong gains, certain crucial points demand prudent consideration. More knowledge about stem cell properties may eventually alleviate the challenges of applying them therapeutically, leading to improved prognosis and quality of life [65].

5.1 Heterogeneity and inherent differential potency of MSCs

MSCs used for medical purposes can originate from either the patient (autologous) or a donor (allogeneic). These cells inhabit complex biological systems in the body and have diverse subpopulations. Ignoring this heterogeneity is a primary reason cell-based therapies have inconsistent results. Stem cells intrinsically proliferate indefinitely while maintaining an undifferentiated state when environmental conditions permit. Thus, appropriate signaling from adjacent cell types and local microenvironments is required for proper functioning. Variations in how cells signal to each other or respond to their surroundings can alter functional pathways. A thorough understanding of diversity across cell populations and their characteristics is essential for developing well-designed clinical trials. As such, fully considering a stem cell’s innate properties when selecting candidates for specific therapies is crucial [66, 67].

5.2 Heterogeneity of disease progression

Carefully reviewing disease worsening over time is pivotal for cell therapy success and demands serious attention. Such review informs customized remedies for enduring impacts. A thorough understanding of illness details, especially for degenerative conditions, allows individual care. Induced pluripotent stem cells (iPSCs) offer game-changing ways to model human sicknesses, particularly genetic ones, by developing iPSCs from uncommon and prevalent disease patients. This provides invaluable disease modeling and medication development, letting progression study and disorder remedies. This process enhances comprehension of underlying molecular drivers as most maladies contain numerous intertwined subsets rather than single illnesses. Initially, stem cell use may concentrate on an affliction, then an individual, potentially ushering a more targeted therapy approach. However, early efforts involve trial and error that markedly improves over time [65, 68].

5.3 Homing and targeted MSC delivery

Stem cells prove highly effective in cell therapy due to sensing their surroundings via cytokine receptors, permitting migration toward damaged tissues or tumors by chemokine gradient trails. This innate navigation aids targeted delivery for enhanced treatment. Beyond natural locating traits, induced direction strategies developed encourage further precise transportation. Methods alter cells through membrane receptor swaps, lipid particles conveying customized cells, viral vectors transmitting genes, or antibody/peptide-hooked particles delivering targets. Still, just a small fraction embeds at wished spots, and engraftment relies on administration technique. Therefore, transportation optimization can emerge from route suitability consideration, physiological forces concentrating cells, preconditioning, and transgenes actuating homing expertly. Additional efforts are required to enhance the understanding and utilization of stem cell manipulation and targeted tissue environments in order to effectively leverage these techniques, either independently or in combination, to provide optimal healthcare outcomes [65, 69, 70].

5.4 Complexity of mechanism of action of cell therapy

When exploring new treatments, a key challenge lies in understanding intricate biochemical and physiological events during application. Animal models often study such processes pre-human use. Cell therapies proved workable clinically, relying on safe, reproducible advantages. Basically, therapy forestalls or reverses sickness worsening. Both approaches overlap and differ at the technique level for cell-based remedies, necessitating customizing to meet illness-specific demands. Identifying diverse biotherapeutics such as anti-death, immune modulation, anti-scarring, pro-vessel growth, cell-cell signaling chemotaxis, local stem/progenitor growth, and diversification aspect discharge is daunting. Defining biochemical pathways and intermolecular machinery involved in cell therapy is critical to grasping stem cell behavior/roles better, a compelling cell therapy platform necessity to heighten predictive abilities. Further exploration embracing cell intricacy centers on understanding underlying behavior/roles [65, 71, 72].

Advertisement

6. Future directions

MSC therapies hold tremendous promise for regenerative medicine, but their complete potential can be realized by implementing various techniques and approaches to reduce adverse effects and improve treatment outcomes. One strategy is the genetic engineering of MSCs, which permits precise modifications to their genetic makeup in order to enhance their therapeutic properties. By introducing specific genes into MSCs, it is possible to improve their survival, ability to differentiate into desired cell types, promote tissue repair, modulate the immune response, or secrete therapeutic factors. This genetic engineering can be achieved through methods such as viral vectors, non-viral vectors, or genome editing tools like CRISPR-Cas9, ensuring targeted and efficient modification of MSCs [73, 74].

Furthermore, the utilization of MSC-derived products, such as exosomes and EVs, has emerged as a promising approach to augment the therapeutic effects of MSC therapies. These vesicles contain a diverse array of bioactive molecules, including proteins, nucleic acids, and signaling molecules that can exert potent therapeutic effects by facilitating intercellular communication, promoting tissue regeneration, and reducing inflammation [75]. To boost the efficacy of MSC-derived products, various techniques can be employed, including optimization of isolation procedures, standardization of production protocols, and characterization of cargo contents. Additionally, engineering MSCs to enhance the secretion of specific exosomes or EVs with desired therapeutic cargo can further enhance their therapeutic potential.

Besides, to mitigate adverse effects associated with MSC therapies, careful consideration should be given to factors such as cell dose, route of administration, and patient selection. Optimizing the dose of MSCs administered ensures that the therapeutic effects are maximized while avoiding potential complications. Choosing the appropriate route of administration, whether it be systemic, local, or targeted delivery, can enhance the homing and engraftment of MSCs to the desired tissue, improving treatment efficacy. Moreover, thorough patient evaluation and selection based on disease characteristics, immune compatibility, and pre-existing conditions can help minimize adverse reactions and maximize the safety and effectiveness of MSC therapies. Continued research and advancements in these areas will undoubtedly contribute to unlocking the full therapeutic potential of MSC-based therapies and pave the way for transformative regenerative medicine.

Advertisement

7. Conclusion

In summary, MSCs possess unique properties making them highly beneficial for therapeutic applications. Their ability to self-renew, differentiate into various cell types, and modulate immunological responses provide MSCs with regenerative capabilities across multiple organ systems. Extensive research over the past decades has revealed the therapeutic potential of MSCs and their exosomes for treating a wide range of medical conditions. As the introduction described, MSCs have shown promise in orthopedic applications like bone and cartilage repair. They have also been studied for their cardiovascular benefits in improving heart function and promoting blood vessel growth. Perhaps most importantly, MSC therapies have demonstrated encouraging results in central nervous system disorders as well as autoimmune and inflammatory conditions.

Looking ahead, further research is still needed to optimize isolation and culture methods to standardize MSC production. Additional pre-clinical and clinical studies are also required to fully characterize MSC engraftment, bio distribution, and mechanisms of action in vivo. Despite these remaining challenges, MSCs represent a promising tool for regenerative medicine. Their multipotency and immunomodulatory properties provide a versatile cell-based therapeutic approach with potential applications across many areas of human disease. With continued advances in basic and translational research, MSC therapies may transform patient care and management of conditions currently lacking effective medical solutions.

Advertisement

Acknowledgments

We thank all those who have directly and indirectly helped in writing this chapter.

Advertisement

Conflict of interest

The authors declare no conflict of interest.

References

  1. 1. Yoo KH, Jang IK, Lee MW, Kim HE, Yang MS, Eom Y, et al. Comparison of immunomodulatory properties of mesenchymal stem cells derived from adult human tissues. Cellular Immunology. 2009;259(2):150-156
  2. 2. Farkhad NK, Mahmoudi A, Mahdipour E. How similar are human mesenchymal stem cells derived from different origins? A review of comparative studies. Current Stem Cell Research & Therapy. 2021;16(8):980-993
  3. 3. 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
  4. 4. Harichandan A, Bühring HJ. Prospective isolation of human MSC. Best Practice & Research Clinical Haematology. 2011;24(1):25-36
  5. 5. Song N, Scholtemeijer M, Shah K. Mesenchymal stem cell immunomodulation: Mechanisms and therapeutic potential. Trends in Pharmacological Sciences. 2020;41(9):653-664
  6. 6. Farkhad NK, Mahmoudi A, Mahdipour E. Regenerative therapy by using mesenchymal stem cells-derived exosomes in COVID-19 treatment. The potential role and underlying mechanisms. Regenerative Therapy. 2022;20:61-71
  7. 7. Lin H, Sohn J, Shen H, Langhans MT, Tuan RS. Bone marrow mesenchymal stem cells: Aging and tissue engineering applications to enhance bone healing. Biomaterials. 2019;203:96-110
  8. 8. Oliveira MS, Saldanha-Araujo F, Goes AM, Costa FF, de Carvalho JL. Stem cells in cardiovascular diseases: Turning bad days into good ones. Drug Discovery Today. 2017;22(11):1730-1739
  9. 9. Qu M, Xing F, Xing N. Mesenchymal stem cells for the treatment of cognitive impairment caused by neurological diseases. Biotechnology Letters. 2022;44(8):903-916
  10. 10. 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
  11. 11. Viswanathan S, Shi Y, Galipeau J, Krampera M, Leblanc K, Martin I, et al. Mesenchymal stem versus stromal cells: International Society for Cell & gene therapy (ISCT®) mesenchymal stromal cell committee position statement on nomenclature. Cytotherapy. 2019;21(10):1019-1024
  12. 12. Schmelzer E, McKeel DT, Gerlach JC. Characterization of human mesenchymal stem cells from different tissues and their membrane encasement for prospective transplantation therapies. BioMed Research International. 2019;2019:6376271
  13. 13. Alcayaga-Miranda F, Cuenca J, Luz-Crawford P, Aguila-Díaz C, Fernandez A, Figueroa FE, et al. Characterization of menstrual stem cells: Angiogenic effect, migration and hematopoietic stem cell support in comparison with bone marrow mesenchymal stem cells. Stem Cell Research & Therapy. 2015;6(1):32
  14. 14. Heo JS, Choi Y, Kim HS, Kim HO. Comparison of molecular profiles of human mesenchymal stem cells derived from bone marrow, umbilical cord blood, placenta and adipose tissue. International Journal of Molecular Medicine. 2016;37(1):115-125
  15. 15. Reinisch A, Etchart N, Thomas D, Hofmann NA, Fruehwirth M, Sinha S, et al. Epigenetic and in vivo comparison of diverse MSC sources reveals an endochondral signature for human hematopoietic niche formation. Blood, The Journal of the American Society of Hematology. 2015;125(2):249-260
  16. 16. Bobis S, Jarocha D, Majka M. Mesenchymal stem cells: Characteristics and clinical applications. Folia Histochemica et Cytobiologica. 2006;44(4):215-230
  17. 17. Herrero C, Pérez-Simón J. Immunomodulatory effect of mesenchymal stem cells. Brazilian Journal of Medical and Biological Research. 2010;43:425-430
  18. 18. Farkhad NK, Reihani H, Moghadam AA, Moghadam AB, Tavakol-Afshari J. Are mesenchymal stem cells able to manage cytokine storm in COVID-19 patients? A review of recent studies. Regenerative Therapy. 2021;18:152-160
  19. 19. Atsuta I, Liu S, Miura Y, Akiyama K, Chen C, An Y, et al. Mesenchymal stem cells inhibit multiple myeloma cells via the Fas/Fas ligand pathway. Stem Cell Research & Therapy. 2013;4:1-14
  20. 20. Gaber T, Schönbeck K, Hoff H, Tran CL, Strehl C, Lang A, et al. CTLA-4 mediates inhibitory function of mesenchymal stem/stromal cells. International Journal of Molecular Sciences. 2018;19(8):2312
  21. 21. Bassi ÊJ, de Almeida DC, Moraes-Vieira PMM, Câmara NOS. Exploring the role of soluble factors associated with immune regulatory properties of mesenchymal stem cells. Stem Cell Reviews and Reports. 2012;8:329-342
  22. 22. Lee RH, Oh JY, Choi H, Bazhanov N. Therapeutic factors secreted by mesenchymal stromal cells and tissue repair. Journal of Cellular Biochemistry. 2011;112(11):3073-3078
  23. 23. Huang Y, Wu Q, Tam PKH. Immunomodulatory mechanisms of mesenchymal stem cells and their potential clinical applications. International Journal of Molecular Sciences. 2022;23(17):10023
  24. 24. Castro-Manrreza ME, Montesinos JJ. Immunoregulation by mesenchymal stem cells: Biological aspects and clinical applications. Journal of Immunology Research. 2015;2015:1-21
  25. 25. Wang L, Deng Z, Sun Y, Zhao Y, Li Y, Yang M, et al. The study on the regulation of Th cells by mesenchymal stem cells through the JAK-STAT signaling pathway to protect naturally aged sepsis model rats. Frontiers in Immunology. 2022;13:820685
  26. 26. Pokrywczynska M, Rasmus M, Jundzill A, Balcerczyk D, Adamowicz J, Warda K, et al. Mesenchymal stromal cells modulate the molecular pattern of healing process in tissue-engineered urinary bladder: The microarray data. Stem Cell Research & Therapy. 2019;10(1):1-17
  27. 27. Romieu-Mourez R, François M, Boivin M-N, Bouchentouf M, Spaner DE, Galipeau J. Cytokine modulation of TLR expression and activation in mesenchymal stromal cells leads to a proinflammatory phenotype. The Journal of Immunology. 2009;182(12):7963-7973
  28. 28. Fu D, Yao Q, Qi X. The role of Notch signaling pathway in bone immune regulation of osteogenic differentiation of macrophages and mesenchymal stem cells. 2023:1-20
  29. 29. Elbaz EM, Helmy HS, El-Sahar AE, Saad MA, Sayed RH. Lercanidipine boosts the efficacy of mesenchymal stem cell therapy in 3-NP-induced Huntington’s disease model rats via modulation of the calcium/calcineurin/NFATc4 and Wnt/β-catenin signalling pathways. Neurochemistry International. 2019;131:104548
  30. 30. Ménard C, Dulong J, Roulois D, Hébraud B, Verdière L, Pangault C, et al. Integrated transcriptomic, phenotypic, and functional study reveals tissue-specific immune properties of mesenchymal stromal cells. Stem Cells. 2020;38(1):146-159
  31. 31. Patel DM, Shah J, Srivastava AS. Therapeutic potential of mesenchymal stem cells in regenerative medicine. Stem Cells International. 2013;2013:1-16
  32. 32. Xiang X-N, Zhu S-Y, He H-C, Yu X, Xu Y, He C-Q. Mesenchymal stromal cell-based therapy for cartilage regeneration in knee osteoarthritis. Stem Cell Research & Therapy. 2022;13(1):1-20
  33. 33. Kasper G, Glaeser JD, Geissler S, Ode A, Tuischer J, Matziolis G, et al. Matrix metalloprotease activity is an essential link between mechanical stimulus and mesenchymal stem cell behavior. Stem Cells. 2007;25(8):1985-1994
  34. 34. Mathiasen AB, Qayyum AA, Jørgensen E, Helqvist S, Fischer-Nielsen A, Kofoed KF, et al. Bone marrow-derived mesenchymal stromal cell treatment in patients with severe ischaemic heart failure: A randomized placebo-controlled trial (MSC-HF trial). European Heart Journal. 2015;36(27):1744-1753
  35. 35. Najafi S, Najafi P, Farkhad NK, Torshizi GH, Darban RA, Boroumand AR, et al. Mesenchymal stem cell therapy in amyotrophic lateral sclerosis (ALS) patients: A comprehensive review of disease information and future perspectives. Iranian Journal of Basic Medical Sciences. 2023;26(8):872
  36. 36. Tavakol-Afshari J, Boroumand AR, Farkhad NK, Moghadam AA, Sahab-Negah S, Gorji A. Safety and efficacy of bone marrow derived-mesenchymal stem cells transplantation in patients with amyotrophic lateral sclerosis. Regenerative Therapy. 2021;18:268-274
  37. 37. Li M, Chen H, Zhu M. Mesenchymal stem cells for regenerative medicine in central nervous system. Frontiers in Neuroscience. 2022;16:1068114
  38. 38. Jasim SA, Yumashev AV, Abdelbasset WK, Margiana R, Markov A, Suksatan W, et al. Shining the light on clinical application of mesenchymal stem cell therapy in autoimmune diseases. Stem Cell Research & Therapy. 2022;13(1):1-15
  39. 39. Wang Y, Fang J, Liu B, Shao C, Shi Y. Reciprocal regulation of mesenchymal stem cells and immune responses. Cell Stem Cell. 2022;29(11):1515-1530
  40. 40. Liu J, Gao J, Liang Z, Gao C, Niu Q, Wu F, et al. Mesenchymal stem cells and their microenvironment. Stem Cell Research & Therapy. 2022;13(1):429
  41. 41. Soler R, Orozco L, Munar A, Huguet M, López R, Vives J, et al. Final results of a phase I–II trial using ex vivo expanded autologous mesenchymal stromal cells for the treatment of osteoarthritis of the knee confirming safety and suggesting cartilage regeneration. The Knee. 2016;23(4):647-654
  42. 42. Šponer P, Filip S, Kučera T, Brtková J, Urban K, Palička V, et al. Utilizing autologous multipotent mesenchymal stromal cells and-Tricalcium phosphate scaffold in human bone defects: A prospective, controlled feasibility trial. BioMed Research International. 2016;2016:1-13
  43. 43. Lee SY, Kim W, Lim C, Chung SG. Treatment of lateral epicondylosis by using allogeneic adipose-derived mesenchymal stem cells: A pilot study. Stem Cells. 2015;33(10):2995-3005
  44. 44. Gao LR, Chen Y, Zhang NK, Yang XL, Liu HL, Wang ZG, et al. Intracoronary infusion of Wharton’s jelly-derived mesenchymal stem cells in acute myocardial infarction: Double-blind, randomized controlled trial. BMC Medicine. 2015;13:1-15
  45. 45. Dongmei H, Jing L, Mei X, Ling Z, Hongmin Y, Zhidong W, et al. Clinical analysis of the treatment of spinocerebellar ataxia and multiple system atrophy-cerebellar type with umbilical cord mesenchymal stromal cells. Cytotherapy. 2011;13(8):913-917
  46. 46. Uccelli A, Laroni A, Brundin L, Clanet M, Fernandez O, Nabavi SM, et al. MEsenchymal StEm cells for multiple sclerosis (MESEMS): A randomized, double blind, cross-over phase I/II clinical trial with autologous mesenchymal stem cells for the therapy of multiple sclerosis. Trials. 2019;20(1):1-13
  47. 47. Singer W, Dietz AB, Zeller AD, Gehrking TL, Schmelzer JD, Schmeichel AM, et al. Intrathecal administration of autologous mesenchymal stem cells in multiple system atrophy. Neurology. 2019;93(1):e77-e87
  48. 48. Duijvestein M, Vos ACW, Roelofs H, Wildenberg ME, Wendrich BB, Verspaget HW, et al. Autologous bone marrow-derived mesenchymal stromal cell treatment for refractory luminal Crohn’s disease: Results of a phase I study. Gut. 2010;59(12):1662-1669
  49. 49. Kaffash Farkhad N, Sedaghat A, Reihani H, Adhami Moghadam A, Bagheri Moghadam A, Khadem Ghaebi N, et al. Mesenchymal stromal cell therapy for COVID-19-induced ARDS patients: A successful phase 1, control-placebo group, clinical trial. Stem Cell Research & Therapy. 2022;13(1):1-13
  50. 50. Farkhad NK, Sedaghat A, Reihani H, Moghadam AA, Moghadam AB, Ghaebi NK, et al. Specific clinical and immunological changes following mesenchymal stem cell transplantation in COVID-19–induced acute respiratory distress syndrome patients: A phase-I clinical trial. Iranian Journal of Allergy, Asthma and Immunology. 2022;21:1-17
  51. 51. Jitschin R, Mougiakakos D, Von Bahr L, Völkl S, Moll G, Ringden O, et al. Alterations in the cellular immune compartment of patients treated with third-party mesenchymal stromal cells following allogeneic hematopoietic stem cell transplantation. Stem Cells. 2013;31(8):1715-1725
  52. 52. Nikfarjam S, Rezaie J, Zolbanin NM, Jafari R. Mesenchymal stem cell derived-exosomes: A modern approach in translational medicine. Journal of Translational Medicine. 2020;18(1):1-21
  53. 53. Moghadam MS, Azimian H, Afshari JT, Toossi MTB, Farkhad NK, Aghaee-Bakhtiari SH. Chromosomal instability in various generations of human mesenchymal stem cells following the therapeutic radiation doses. 2023;2023:1-22
  54. 54. Elahi FM, Farwell DG, Nolta JA, Anderson JD. Preclinical translation of exosomes derived from mesenchymal stem/stromal cells. Stem Cells. 2020;38(1):15-21
  55. 55. Pegtel DM, Gould SJ. Exosomes. Annual Review of Biochemistry. 2019;88:487-514
  56. 56. Vlassov AV, Magdaleno S, Setterquist R, Conrad R. Exosomes: Current knowledge of their composition, biological functions, and diagnostic and therapeutic potentials. Biochimica et Biophysica Acta (BBA)-General Subjects. 2012;1820(7):940-948
  57. 57. Gao K, Jin J, Huang C, Li J, Luo H, Li L, et al. Exosomes derived from septic mouse serum modulate immune responses via exosome-associated cytokines. Frontiers in Immunology. 2019;10:1560
  58. 58. Zhang X, Sheng Y, Li B, Wang Q, Liu X, Han J. Ovarian cancer derived PKR1 positive exosomes promote angiogenesis by promoting migration and tube formation in vitro. Cell Biochemistry and Function. 2021;39(2):308-316
  59. 59. Li Q, Wang Z, Xing H, Wang Y, Guo Y. Exosomes derived from miR-188-3p-modified adipose-derived mesenchymal stem cells protect Parkinson’s disease. Molecular Therapy-Nucleic Acids. 2021;23:1334-1344
  60. 60. Okoye IS, Coomes SM, Pelly VS, Czieso S, Papayannopoulos V, Tolmachova T, et al. MicroRNA-containing T-regulatory-cell-derived exosomes suppress pathogenic T helper 1 cells. Immunity. 2014;41(1):89-103
  61. 61. Chen J, Li C, Liang Z, Li C, Li Y, Zhao Z, et al. Human mesenchymal stromal cells small extracellular vesicles attenuate sepsis-induced acute lung injury in a mouse model: The role of oxidative stress and the mitogen-activated protein kinase/nuclear factor kappa B pathway. Cytotherapy. 2021;23(10):918-930
  62. 62. Lee J-K, Park S-R, Jung B-K, Jeon Y-K, Lee Y-S, Kim M-K, et al. Exosomes derived from mesenchymal stem cells suppress angiogenesis by down-regulating VEGF expression in breast cancer cells. PLoS One. 2013;8(12):e84256
  63. 63. Yu B, Zhang X, Li X. Exosomes derived from mesenchymal stem cells. International Journal of Molecular Sciences. 2014;15(3):4142-4157
  64. 64. Eirin A, Riester SM, Zhu X-Y, Tang H, Evans JM, O'Brien D, et al. MicroRNA and mRNA cargo of extracellular vesicles from porcine adipose tissue-derived mesenchymal stem cells. Gene. 2014;551(1):55-64
  65. 65. Srijaya TC, Ramasamy TS, Kasim NHA. Advancing stem cell therapy from bench to bedside: Lessons from drug therapies. Journal of Translational Medicine. 2014;12:1-17
  66. 66. Hope K, Bhatia M. Clonal interrogation of stem cells. Nature Methods. 2011;8(Suppl 4):S36-S40
  67. 67. Cahan P, Daley GQ. Origins and implications of pluripotent stem cell variability and heterogeneity. Nature Reviews Molecular Cell Biology. 2013;14(6):357-368
  68. 68. Ebben JD, Zorniak M, Clark PA, Kuo JS. Introduction to induced pluripotent stem cells: Advancing the potential for personalized medicine. World Neurosurgery. 2011;76(3–4):270-275
  69. 69. Komarova S, Roth J, Alvarez R, Curiel DT, Pereboeva L. Targeting of mesenchymal stem cells to ovarian tumors via an artificial receptor. Journal of Ovarian Research. 2010;3(1):1-14
  70. 70. Ansboro S, Greiser U, Barry F, Murphy M. Strategies for improved targeting of therapeutic cells: Implications for tissue repair. European Cells & Materials. 2012;23:310-318
  71. 71. Laflamme MA, Zbinden S, Epstein SE, Murry CE. Cell-based therapy for myocardial ischemia and infarction: Pathophysiological mechanisms. Annual Review of Pathology and Mechanical Diseases. 2007;2:307-339
  72. 72. Drukker M, Tang C, Ardehali R, Rinkevich Y, Seita J, Lee AS, et al. Isolation of primitive endoderm, mesoderm, vascular endothelial and trophoblast progenitors from human pluripotent stem cells. Nature Biotechnology. 2012;30(6):531-542
  73. 73. Nowakowski A, Walczak P, Lukomska B, Janowski M. Genetic engineering of mesenchymal stem cells to induce their migration and survival. Stem Cells International. 2016;2016:1-10
  74. 74. 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
  75. 75. Bian D, Wu Y, Song G, Azizi R, Zamani A. The application of mesenchymal stromal cells (MSCs) and their derivative exosome in skin wound healing: A comprehensive review. Stem Cell Research & Therapy. 2022;13(1):24

Written By

Mohammad Ali Khodadoust, Amirreza Boroumand, Alireza Sedaghat, Hamidreza Reihani, Najmeh Kaffash Farkhad and Jalil Tavakol Afshari

Submitted: 21 September 2023 Reviewed: 01 October 2023 Published: 07 December 2023