Skip to main content

Mesenchymal stromal/stem cells from perinatal sources: biological facts, molecular biomarkers, and therapeutic promises

Abstract

The use of mesenchymal stem cells (MSCs) from perinatal tissue sources has gained attention due to their availability and lack of significant ethical or moral concerns. These cells have a higher proliferative capability than adult MSCs and less immunogenic or tumorigenesis risk than fetal and embryonic stem cells. Additionally, they do not require invasive isolation methods like fetal and adult MSCs. We reviewed the main biological and therapeutic aspects of perinatal MSCs in a three-part article. In the first part, we revised the main biological features and characteristics of MSCs and the advantages of perinatal MSCs over other types of SCs. In the second part, we provided a detailed molecular background for the main biomarkers that can be used to identify MSCs. In the final part, we appraised the therapeutic application of perinatal MSCs in four major degenerative disorders: degenerative disc disease, retinal degenerative diseases, ischemic heart disease, and neurodegenerative diseases. In conclusion, there is no single specific molecular marker to identify MSCs. We recommend using at least two positive markers of stemness (CD29, CD73, CD90, or CD105) and two negative markers (CD34, CD45, or CD14) to exclude the hematopoietic origin. Moreover, utilizing perinatal MSCs for managing degenerative diseases presents a promising therapeutic approach. This review emphasizes the significance of employing more specialized progenitor cells that originated from the perinatal MSCs. The review provides scientific evidence from the literature that applying these progenitor cells in therapeutic procedures provides a greater regenerative capacity than the original primitive MSCs. Finally, this review provides a valuable reference for researchers exploring perinatal MSCs and their therapeutic applications.

Biological facts

Mesenchymal stem cells (MSCs) are multipotent regenerative cells that can differentiate into several tissue cells [1, 2]. They have been designated the name “multipotent mesenchymal stromal cells” by the International Society for Cellular Therapy (ISCT) [2]. However, the term “MSCs” remains the most widely used name in literature. According to the Mesenchymal and Tissue Stem Cell Committee of the ISCT, human MSCs must exhibit the following standards to be considered for scientific investigations and/or pre-clinical trials:

  • They must be plastic-adherent.

  • They must express the following cell surface antigen biomarkers: CD73 (ecto 5’ nucleotidase), CD90 (Thy−1), and CD105 (endoglin).

  • They must lack the expression of hematopoietic antigens. These include the following: the hematopoietic progenitor cells marker CD34; the pan-leukocyte marker CD45; the monocyte and macrophage markers CD14 and CD11b; the B-cell markers CD19 and CD79α.

  • They must display trilineage differentiation in vitro. Thus, the cells can differentiate into osteoblasts, chondroblasts, and adipocytes in culture.

  • In addition, the following fifth criterion of MSCs has been emphasized in the literature:

  • They must be able to show a fibroblastoid morphology [1, 3, 4].

Despite sharing the aforementioned standards, human MSCs can still be classified into three types based on their source tissue. These include (i) adult MSCs, isolated from adult tissues, especially bone marrow (BM) and adipose tissue; (ii) fetal MSCs, isolated from collected samples of amniotic fluid or fetal tissues after terminated pregnancies; and (iii) perinatal MSCs, which are isolated from the placentome tissues (i.e., umbilical cord and placenta membranes). Perinatal MSCs can be harvested from five distinct regions of the placentome (Fig. 1), including the umbilical cord (UC) lining membrane, Wharton’s jelly (WJ), The junction between the UC and placenta, the fetal chorion, and the maternal part of the placenta (i.e., Decidua basalis) [1].

Fig. 1
figure 1

Illustration of the five different regions of the placentome to isolate perinatal MSCs. These are cord lining (CL), Wharton’s jelly (WJ), cordplacenta junction (CPJ), the chorion (Ch), and the maternal placenta (P). Created in https://BioRender.com

The MSCs from perinatal sources have attracted the attention of many researchers due to their abundance in easily accessible tissue sources without significant ethical and safety concerns. For example, perinatal MSCs do not carry major ethical concerns like embryonic stem cells (ESCs). The primary ethical conflict associated with ESCs consists of using human embryos to obtain these cells [5, 6]. Therefore, the ethical debate surrounding the ESCs revolves around whether it is justifiable to develop new cell-based therapies at the cost of harming human embryos. In contrast, MSCs derived from perinatal sources do not raise significant ethical concerns. They only require institutional ethical approval along with the consent of the mother or guardian to use the discarded placentome tissue. Additionally, perinatal MSCs do not need invasive harvesting procedures, unlike MSCs from adult sources such as bone marrow and adipose tissue. The invasive procedures required to harvest adult MSCs can cause tissue damage, discomfort or pain to the donor, and place him at risk. Therefore, harvesting adult MSCs requires more ethical and safety considerations than the perinatal MSCs.

Perinatal MSCs have numerous advantages over MSCs from other sources (Fetal and adult), making them the center of attention for both experimental research and therapeutic applications. These benefits include:

  • Higher proliferation capability than adult MSCs [7].

  • Lower immunogenicity than adult MSCs with the ability to ameliorate immune response [7, 8].

  • Less likely to have mutations than adult MSCs since they are immature cells, which reduces the risk of tumorigenesis [4, 7, 9].

  • They do not require invasive isolation methods like fetal and adult MSCs [1, 9].

  • They have no ethical issues like fetal and adult MSCs since placentome tissues are considered medical waste that is discarded after birth [1, 9, 10].

  • large-scale availability due to the abundance of placentome tissues that yield a high number of cells [4, 7].

Molecular biomarkers

Numerous surface biomarkers have been used to identify MSCs. However, there is currently no single marker that can be used specifically to identify MSCs. Herewith, we discuss the most recommended and most frequently adopted biomarkers in the literature for MSC identification. These include the three recommended markers by the ISCT committee (CD73, CD90, & CD105), the three markers of stemness, known as the pluripotency triad (Sox2, Oct4, & Nanog), and two widely used integrin biomarkers (CD29 & CD49f). A summary of the structural and functional characteristics of these markers is provided in Table 1.

Table 1 Summary of the most commonly used biomarkers to identify MSCs from perinatal tissues
Fig. 2
figure 2

Role of CD73 in the hydrolysis of extracellular ATP into Adenosine. Created in https://BioRender.com

Cluster of differentiation 73 (CD73, NT5E)

This is a membrane-bound enzyme (ectoenzyme) that is known as ecto-5’-nucleotidase (NT5E) and is encoded by the NT5E gene [11, 12]. It degrades extracellular AMP into adenosine [11]. The successive enzymatic activities of ectonucleotidases CD39 and CD73 form the main pathway for hydrolysis of extracellular ATP into adenosine (Fig. 2). CD39 hydrolyzes ATP into AMP, and CD73 further converts AMP into adenosine [13]. Therefore, CD73 is considered an immune modulator since it helps convert the pro-inflammatory ATP molecule into the immune suppressive nucleoside adenosine [11, 13].

As noted in the previous section, CD73 has been recognized by the ISCT as one of the three surface markers required to define the MSCs [1, 2]. However, it has been revealed that CD73 is unequally expressed among different MSC populations, with MSCs from perinatal sources (especially from the UC) having higher expression levels of CD73 than MSCs from adult sources (including BM and adipose tissue) [14]. This heterogenous pattern of CD73 expression may explain the inconsistency in regenerative potentials among different MSCs. For example, MSCs expressing high levels of CD73 promote significantly better cardiac repair in infarcted murine hearts than MSCs with low levels of CD73 [14].

Cluster of differentiation 90 (CD90, Thy-1)

This protein is anchored to the outer surface of the plasma membrane by a glycosylphosphatidylinositol (GPI) segment [15, 16]. It is also known as thymocyte differentiation antigen 1 (Thy-1) because it was initially discovered in murine thymocytes [17]. It is associated with many biological functions, including cell death (apoptosis), cell adhesion, cell proliferation and migration, and as a pluripotency marker for stem cells [16, 18].

CD90 is expressed in different types of stem cells, including cancer stem cells (CSCs) [19, 20], hematopoietic stem cells (HSCs) [21, 22], and MSCs [1, 2, 9]. Therefore, when using CD90 to identify MSCs, it is necessary to use additional markers to distinguish them from other stem cell populations. For example, CD34, a common marker for HSCs, is often used with CD90 to differentiate between MSCs and HSCs. HSCs are CD34+/CD90+, while MSCs are CD34-/CD90+ [18, 22].

Cluster of differentiation 105 (CD105, Endoglin)

This is a transmembrane glycoprotein commonly known as endoglin because it is predominantly expressed in the endothelial cells of blood vessels. Despite being initially identified in a lymphoblastic leukemia cell line, it has been exclusively investigated in vascular endothelial cells [23, 24]. Endoglin (CD105) is composed of three distinct domains. These include (i) an extracellular tripeptide domain, which is heavily glycosylated; (ii) a hydrophobic transmembrane domain; and (iii) an intracellular domain, which is rich in serine/threonine phosphorylation sites [24]. The intracellular domain is involved in modulating response only, and it does not commence a signaling cascade [25].

Endoglin functions as an auxiliary receptor or coreceptor since it is a component of the receptor complex for the transforming growth factor beta (TGF-β) superfamily ligands. This receptor complex is made of three types of receptors. These are TGF-β receptor type 1 (TGF-βR1), TGF-β receptor type 2 (TGF-βR2), and CD105 (endoglin). Endoglin is also known by some investigators as TGF-β receptor type 3 (TGF-βR3). CD105 is first associated with receptor TGR-βR2. This association allows the binding of TGF-β 1 or 3 ligands (Fig. 3). The binding of these ligands to the association will then activate TGF-βR2 to phosphorylate TGF-βR1. Phosphorylation of TGF-βR1, also known as activin receptor-like kinase (ALK), will initiate intracellular downstream signaling cascades. There are two downstream signaling pathways that are activated by two different subtypes of TGF-βR1. These include ALK1 or ALK5 kinase (Fig. 3) [24,25,26].

Fig. 3
figure 3

Mechanism of action of CD105 through TGF-β receptor complex. CD105 is first associated with receptor TGR-βR2. This association allows the binding of TGF-β 1 or 3 ligands to the complex. The binding of these ligands to the receptor complex will then activate TGF-βR2 to phosphorylate TGF-βR1. Phosphorylation of TGF-βR1, also known as activin receptor-like kinase (ALK), will initiate two intracellular downstream signaling pathways that are activated by two different subtypes of TGF-βR1 (ALK1 or ALK5). Created in https://BioRender.com

Endoglin is expressed in various cell types, including vascular endothelial cells, MSCs, HSCs, tumor-associated fibroblasts, and epithelial cancer cells [23, 24, 26]. Additionally, its expression levels are significantly elevated in the highly proliferating than in low proliferating and quiescent cells. Therefore, it is a predominant marker of both tumorigenesis and stemness. For example, MSCs expressing CD105 have a higher proliferation rate and better colony formation than MSCs not expressing CD105 [27]. In summary, similar to CD90, CD105 is a marker of proliferation and cannot be used as a single marker to identify MSCs. Additional markers are required to confirm the stemness of the cells (i.e., CD73 + and CD90+) and to exclude the hematopoietic origin (i.e., CD45− and CD34−).

Sex determining region Y – Box 2 (Sox2)

This nuclear transcriptional factor is essential for the maintenance and stemness of different types of stem cells [28]. It is one of three critical transcription factors that are referred to as the pluripotency triad of stem cells. These are Sox2, Oct4, and Nanog [29, 30]. Sox2 is a core transcription factor in embryonic stem cells (ESCs) and MSCs from perinatal and adult sources [28,29,30,31]. It is expressed at a higher level in MSCs from perinatal sources than MSCs in adult sources (BM and Adipose tissue). However, this level of expression is not as high as the expression level in ESCs [28].

Sox2 interacts with Oct4 to form a binary complex that activates pluripotent genes and represses differentiation genes [32]. It has been reported that Sox2 is essential for maintaining the self-renewal capability of MSCs, and without Sox2, the cells may lose their stemness [30]. In fact, the expression level of Sox2 gradually decreases as the MSC passaging number increases. Additionally, the knockdown of Sox2 inhibits proliferation and significantly reduces the colony-forming ability of MSCs [30]. On the other hand, induced expression of Sox2 in aged bone marrow-derived MSCs (BM-MSCs), which display flattened morphology and decreased proliferation, restored their normal fibroblastoid morphology and proliferation rate [33]. It is anticipated that higher expression of Sox2 is associated with greater stemness, increased proliferative potential, and better cell migration and adhesion in perinatal MSCs [31].

Octamer-binding transcription factor 4 (Oct4, POU5F1)

This is an important transcription factor found in both ESCs and MSCs. It is involved in sustaining the self-renewal ability of undifferentiated stem cells [9, 34]. It is the first and most used marker for identifying ESCs [34]. However, it is also expressed as a marker in perinatal MSCs isolated from WJ and the amniotic membrane of the placenta [9].

Nanog

Nanog is a homeobox transcription factor that is a member of the pluripotency triad (Sox2, Oct4, & Nanog) [35]. It is a 305 amino acids (AAs) long protein with a homeobox domain in the middle that spans 60 AAs from AA 95 to AA 154 [35]. In addition, it contains an N-terminal region that constitutes 94 AAs and is rich in serine residues (17 serine residues in its N-terminal). The C-terminal region contains a tryptophan repeating domain where every fifth residue is a tryptophan (residues 200, 205, & 210). The tryptophan repeating domain is essential for Nanog’s interaction with other pluripotency factors and for mediating Nanog’s dimerization [36, 37]. It is important to note that in addition to the original NANOG gene on chromosome 12, there are 11 NANOG pseudogenes marked as P1 to P11 [35, 38]. The first pseudogene (NANOGP1) results from a tandem duplication of the original NANOG gene and has a high homology to the original gene. The remaining pseudogenes (NANOGP2-NANOGP11) are located on different chromosomes and show 85–90% homology to the original NANOG gene [35, 38].

Nanog’s name is derived from the Celtic Irish mythology “Tir nan Og,” which means the land of ever young, where people never age [39, 40]. This explains the main role of Nanog in stem cells, which includes maintaining the stemness and self-renewal of these cells and retaining them in an undifferentiated state [35]. Nanog is expressed in different types of stem cells, including ESCs [39, 40], CSCs [35, 38], and both perinatal [41,42,43] and adult [44, 45] MSCs.

Nanog is a key factor in maintaining the self-renewal of perinatal MSCs by preserving stemness, delaying senescence, and increasing the expression of both Sox2 and Oct4 [28]. It is important to clarify that Nanog, along with Sox2 and Oct 4, are forming a pluripotency triad that is essential for maintaining self-renewal and pluripotency of ESCs. In MSCs, this triad is also speculated to play similar roles in maintaining MSCs self-renewal and retaining them in an undifferentiated state [46]. This is evident by the fact that overexpression of these factors promotes the proliferation rate and enhances the colony formation of MSCs [47]. In contrast, the knockdown of these factors significantly reduces the growth rate and multipotency of MSCs [48, 49]. Furthermore, it has been established that Nanog functions by activating the repressors and suppressing the activators of differentiation [35]. Therefore, Nanog inhibits the differentiation of various types of stem cells.

One of Nanog’s suggested mechanisms of action is that it collaborates with Oct4 to suppress the expression of genes that promote differentiation and senescence by enhancing DNA methylation. It has been revealed that both Nanog and Oct4 can upregulate the expression of a significant DNA methyltransferase gene called Dnmt1 by binding directly to its promoter region [46]. In turn, Dnmt1 will promote DNA methylation and inhibit the transcription of p16, p21, and other senescence genes, ultimately retaining cell proliferation and inhibiting differentiation (Fig. 4).

Fig. 4
figure 4

Illustrating diagram for a suggested mechanism of action of Nanog in maintaining self-renewal of MSCs

Cluster of differentiation 29 (CD29, ITGβ1)

This is a transmembrane adhesion molecule that is also known as integrin beta 1 (ITGβ1) [50, 51]. This molecule has four isoforms, and it can be associated with different integrin alpha subunits to form integrin complexes that bind extracellular adhesion proteins, including collagen, laminin, and fibronectin [52, 53]. CD29 is widely expressed in different types of cell lineages and in both progenitor and differentiating cells [52].

Previous studies showed that this glycoprotein biomarker is crucial for the survival, proliferation, and migration of MSCs. Interrupting interactions between ITGβ1 and extracellular matrix proteins can result in MSC death [54]. On the other hand, overexpression of ITGβ1 increases MSC survival and improves their regenerative efficacy [55]. Ode et al. 2011 reported that decreased CD29 expression results in reduced migratory capacity of MSCs [51]. Furthermore, Ip et al. (2007) found that CD29 blockade reduces the migration and engraftment accumulation of MSCs into infarcted sites of ischemic murine hearts [56].

Several previous studies used CD29 to identify MSCs from perinatal tissues [4, 57,58,59,60]. Brown et al. (2019) showed that more than 99% of MSCs isolated from the five different regions of the perinatal tissues (Fig. 1) express CD29 [1]. Additionally, researchers used CD29 as a marker to investigate the impact of maternal age on the frequency and distribution of perinatal MSCs within different areas of the placenta [59] and the UC [58]. In conclusion, to define MSCs using CD29, it is necessary to use at least two additional markers. First, a positive marker is needed to confirm the stemness of the cells, such as CD73 or CD90. Second, a negative marker is required to exclude the hematopoietic origin of the cell, such as CD34 or CD45.

Integrin alpha 6 (ITGA6, CD49f)

This is another transmembrane protein that functions as a cell adhesion receptor [61, 62]. This cell surface biomarker is expressed in about 35 different populations of stem cells, indicating its integral role in stem cell biology, which maintains the self-renewal mechanism of stem cells [62]. It has been identified in both adult and perinatal MSCs. Nieto-Nicolau et al. (2020) reported that the expression of ITGA6 can indicate the progenitor potential of BM-MSCs since BM-MSCs that express higher levels of ITGA6 have higher clonogenicity, migration, and differentiation potentials [63]. Comparable to these findings, Al-Obaide et al. (2022) revealed that higher expression of ITGA6 is correlated with higher clonogenicity and shorter doubling time in perinatal MSCs [61]. Therefore, the expression of ITGA6 is necessary for the proliferation and self-renewal of MSCs. Perinatal MSCs isolated from the WJ in the UC had higher expression levels of ITGA6 than MSCs isolated from the placenta [61].

The suggested mechanism of action for ITGA6 is summarized in Fig. 5 below. It has been reported that Oct4 and Sox2 transcription factors play positive roles in the expression of ITGA6. Both Oct4 and Sox2 bind to the promoter region of the ITGA6 gene [62, 64]. On the other hand, the silencing of Oct4 and Sox2 leads to the downregulation of ITGA6 activity [64]. It is speculated that ITGA6 sustains the stemness of human MSCs by the activation of the PI3K/AKT pathway and downregulation of p53, a cell cycle regulator protein [64]. In pluripotent stem cells, ITGA6 levels diminish during differentiation, and integrin β1 is activated, leading to phosphorylation and activation of Focal adhesion kinase (FAK), which induces the cell differentiation and reduces the expression of the pluripotency factors Oct4, Sox2, and Nanog (Fig. 5) [65].

Fig. 5
figure 5

Illustrating diagram for the mechanism of action of Integrin Alpha 6 (ITGA6, CD49f) in maintaining self-renewal of MSCs

Therapeutic promises

Degenerative disc disease (DDD)

Medical research has been increasingly focused on applying stem cell-based therapy in degenerative disc diseases (DDDs). Perinatal MSCs, particularly those isolated from WJ in the UC, have shown considerable promise in cartilage regeneration and intervertebral disc (IVD) repair [66]. The first goal in using MSC therapy for DDD is to overcome the pathological catabolic microenvironment inside the disc by inhibiting the abnormal production of matrix-degrading enzymes and inflammatory cytokines, like interleukin-1β (IL-1β) and TNF-α, by the resident nucleus pulposus (NP) cells [67,68,69]. The second goal is to promote a healthy and more anabolic environment by promoting the implanted MSCs to produce growth factors (i.e., TGF-β, GDF5, & GDF6), anti-inflammatory agents (i.e., IL-1 receptor antagonist), and anti-catabolic factors known as tissue inhibitors of metalloproteinases (TIMPs) [66]. Previous studies revealed that MSCs are capable of improving the disc microenvironment mainly through paracrine activity by producing exosomes (extracellular vesicles) that contain proteins, microRNAs, and lipids [66, 70, 71]. These exosomes can release their contents directly into the extracellular matrix (ECM) or deliver them into a nearby NP cell after being ingested by endocytosis [71].

The MSC-secreted exosomes can produce a more favorable environment inside the IVD by several mechanisms. MSC-exosomes can release microRNAs (i.e., miR-21 & miR-31-5p) that are capable of inhibiting caspase-3 activity, thereby reducing NP cell apoptosis [71, 72]. Additionally, MSC-exosomes contain TIMPs natural proteins that bind directly to the metalloproteinases (ECM-degrading enzymes) and prevent their catabolic activity, thereby maintaining the ECM [73]. Moreover, MSC-exosomes comprise several anti-inflammatory proteins and miRNAs that suppress the inflammatory cytokines IL-1β and TNF-α, thereby reducing the inflammatory response within the IVD [66, 71]. For example, MSC-exosomes containing miR-532-5p were found to suppress TNF-α by silencing the pro-apoptotic gene RASSF5 [74]. Also, MSC-exosomes contain factors that enhance the expression of cartilage-specific genes aggrecan, collagen II, and SOX9, thereby improving cartilage regeneration [75]. Finally, MSC-exosomes contain several anabolic growth factors like TGF-β1, HGF, BMP2, and GDF5 that contribute to the regeneration of the IVD [66, 70, 76].

Nevertheless, several challenges still hinder the clinical application of MSCs in DDDs. These are mainly technical difficulties related to pre-conditioning and preparation of implanted stem cells and overcoming the harsh pathological environment of degenerated IVD. Research has shown that implanted MSCs cannot survive in the acidic, hypoxic, and nutrient-deprived avascular environment inside the IVD [77].

In 2018, Beeravolu et al. investigated the potential of using human umbilical cord-derived MSCs (UC-MSCs) in remedying IVD degeneration. They compared the therapeutic outcomes between UC-MSCs and chondrogenic progenitor cells (ChPCs) derived from these cord cells in vitro. They showed a far greater capacity for the in vitro differentiated ChPCs to regenerate the IVD than their original UC-MSCs [3]. The UC-MSCs were differentiated into ChPCs by culturing them in a chondrogenic medium rich in TGF-β1, insulin, dexamethasone, and ascorbic acid. The ChPCs were then identified by expressing the chondrogenic markers Sox9, aggrecan, Collage type II, and the NP-specific cell marker FOXF1 [3]. Additionally, researchers at the Oakland University-William Beaumont Institute for Stem Cell and Regenerative Medicine (OU-WB ISCRM) were able to optimize in vitro differentiation of human UC-MSCs into nucleus pulposus-like cells (NPCs) by culturing them in a special differentiation medium that contained TGF-β1, BMP7, GDF5, insulin, dexamethasone, and ascorbic acid [78]. The NPCs were identified by their expression for the chondrogenic markers Sox9, Aggrecan, and Collagen type II. More importantly, the differentiated cells were able to express NP-specific markers, including FOXF1, PAX6, CA12, and KRT19. When these NPCs were injected into degenerated rabbit IVDs, they provided much better regenerative capacity than their original UC-MSCs [78].

However, many studies revealed that MSC therapy alone in DDD is not enough. They emphasized the imperative need to use biocompatible materials with mechanical strength as scaffolds to restore mechanical stability and support spine loading [79,80,81,82,83]. Additionally, these scaffolds can prevent cell leakage from the injection site, therefore improving cell growth and differentiation. Most of the scaffolds investigated were injectable hydrogels to avoid invasive surgical interventions. Investigators at OU-WB ISCRM used self-assembling hydrogel scaffolds composed of polyethylene glycol biocompatible materials to examine the efficacy of combined MSC therapy and tissue engineering in managing DDD. They discovered that using self-assembling scaffolds can promote retention of the implanted cells within the IVD, which improves cellularity and ECM accumulation of the regenerated tissue [83].

Nevertheless, the hydrogel scaffolds do not provide sufficient mechanical support to completely restore the degenerated disc height. Therefore, the researchers at OU-WB ISCRM suggested a novel multi-pronged therapeutic approach for treating DDD [84]. This approach can be summarized in three main ideas: (1) Using a special distractive medical device to gradually restore the disc space to its natural height (Fig. 6). (2) Injection of self-assembling hydrogel scaffolds into the NP of the disc to prevent cell leakage. (3) The use of more chondrogenic lineage descendant progenitor cells like NPCs, which proved to be more effective in the regeneration process than the primitive UC-MSCs.

Fig. 6
figure 6

Distractive medical device to restore intervertebral disc space

Retinal degenerative diseases (RDDs)

Retinal degenerative diseases (RDDs) refer to a group of pathologies that involve progressive degeneration of the cells in the retina, leading to low vision that subsequently worsens to induce loss of visual fields (partial blindness) and eventually end up in complete blindness [85,86,87]. These pathologies include (1) age-related macular degeneration, (2) retinitis pigmentosa, (3) pediatric Stargardt macular dystrophy (Stargardt disease), (4) diabetic retinopathy, and (5) glaucoma [86, 88]. The visual damage induced by RDDs is irreversible since the lost retinal cells are not replaced [86]. Hence, using MSC-based therapy provides a therapeutic promise to replace degenerated cells.

Several studies investigated the role of perinatal MSCs in managing RDDs [85, 89,90,91]. It is anticipated that MSCs can act through two main mechanisms to halt and overcome the damage induced by RDDs. The first mechanism is to support and protect the existing retinal cells by producing growth and anti-apoptotic factors. This mechanism is achieved in two ways: (i) secretion of several growth and neurotrophic factors that improve the persistence and growth of retinal cells, such as hepatocyte growth factor (HGF), nerve growth factor (NGF), pigment epithelium growth factor (PEGF), epidermal growth factor (EGF), Brain-derived neurotrophic factor (BDNF), and ciliary neurotrophic factor (CNTF). (ii) secretion of anti-inflammatory and anti-apoptotic agents that protect and support the survival of the retinal cells, such as TGF-β, interleukin-6 (IL-6), indoleamine 2,3-deoxygenase (IDO), TNF-α stimulated gene 6 protein (TSG-6), and programmed death-ligand 1 (PDL-1). The second mechanism is to rejuvenate the pool of degenerated cells. This mechanism is achieved by direct differentiation into new retinal cells that replace the degenerated ones. It is suggested that signals from the damaged retina stimulate this differentiation [88, 92]. Additional mechanisms are also speculated to contribute to the treatment of RDDs by MSCs, such as the ability of MSCs to release extracellular vesicles that comprise functional molecules like anti-inflammatory proteins, growth factors, and microRNAs [93,94,95]. These vesicles were found to be endocytosed by the existing retinal cells in a receptor-mediated, dose-dependent, and saturable manner [96]. Other mechanisms may include mitochondrial transfer and/or fusion with pre-existing retinal cells [88].

Similar to the previous principle in DDD, it is anticipated that using more specialized derivative progenitor cells would be more effective for retinal regeneration than using the original primitive MSCs. Brown et al. (2022) differentiated perinatal MSCs obtained from the human UC into retinal progenitor cells (RPCs) in vitro by culturing them for two weeks in a special neurobasal medium that contains EGF, retinoic acid, taurine, glutamine, and B27 neuronal supplement [85]. Then, they compared the therapeutic effects between the original primitive MSCs and their differentiated RPCs by intravitreally injecting these cells into a retinitis pigmentosa mouse model. They found that the primitive MSCs were restricted to the epithelial layer of the retina. In contrast, the RPCs could migrate and integrate into different neuronal layers of the retina. The RPCs significantly increased the retinal thickness and improved the visual function in the retinitis pigmentosa mouse model compared to the primitive MSCs. Additionally, it has been indicated that RPCs are the best candidate for stem cell-based therapy of RDDs [97].

In summary, RDDs are the primary sources of visual impairment and blindness worldwide. A promising therapeutic approach is available by using MSC-based therapy. The experimental findings on animal models are promising; however, these findings still need to be validated by clinical trials. Many phase I and II trials were performed and reviewed in the literature [86, 87, 97,98,99]. Most of these trials focused mainly on the safety and efficacy of the application of MSCs in RDDs. However, several questions still need to be investigated and verified in future clinical trials. These include questions about the best source of MSCs to be used (embryonic vs. perinatal vs. adult), the in vitro differentiation state of the cells (primitive vs. RPCs), the route of administration (intravenous vs. intravitreal), and the optimized dose required without inducing potential hazards.

Ischemic heart disease (IHD)

Ischemic heart disease, also known as coronary artery disease, is the leading cause of death worldwide [100,101,102]. It involves a reduction of blood supply to the cardiac muscle due to a narrowing in a coronary artery [102, 103]. The reduced blood supply can lead to damage of part of the cardiac muscle and death of cardiomyocytes, known as myocardial infarction (MI). The MI will eventually result in heart failure [102].

It has been demonstrated that intravenous infusion of perinatal MSCs can enhance heart function and improve quality of life in patients with stable heart failure [104]. The perinatal MSCs can be induced to differentiate into cardiomyocytes in vitro by either treating them with special chemical agents and supplements [105] or by coculturing them with pre-existing cardiomyocytes [106,107,108]. The most common and widely used chemical to induce differentiation of MSCs into cardiomyocytes is 5-azacytidine (5-AZA). This chemical agent is an analog of the pyrimidine nucleoside cytidine [109]. It can disrupt RNA metabolism and inhibit DNA methylation; therefore, it is used as an anticancer drug for treating different kinds of leukemia [109,110,111]. It has been anticipated that 5-AZA induces cardiomyogenic differentiation of MSCs by activating the MEK-ERK and Notch signaling pathways [105, 112]. The suggested mechanism of action of 5-AZA to induce differentiation of MSCs into cardiomyocytes is illustrated in Fig. 7.

Fig. 7
figure 7

The suggested mechanism of action of 5-azacytidine (5-AZA) in inducing MSC differentiation into Cardiomyocytes. 5-AZA inhibits MSC proliferation by suppressing the expression of Nanog and Sox2. It is anticipated that 5-AZA is acting through Notch and MAPK pathways to stimulate the expression of cardiac-specific genes and eventually induce differentiation into cardiomyocytes. Abbreviations: DLL4, delta-like 4 ligand; GATA4, GATA-binding protein 4; Nkx2.5, NK-2 homeobox 5 protein; cTnT, cardiac troponin T; ANP, A-type natriuretic peptide

Previous studies recommended adding culture supplements, such as fetal bovine serum and platelet lysate, to support MSC growth, expansion, and differentiation [113,114,115,116]. Platelet lysate (PL) is a media supplement that is rich in several growth and differentiation factors; therefore, it can be used as a differentiation inducer in vitro [117]. Among these factors are the platelet-derived growth factor (PDGF), vascular endothelial growth factor (VEGF), insulin-like growth factor 1 (IGF-1), TGF-β, and EGF [115, 117, 118]. It has been revealed that human PL (hPL) is an effective cardiomyogenic supplement. The combined treatment of perinatal MSCs with hPL and 5-AZA resulted in greater cell viability and significantly higher expression of cardiomyogenic markers than treatment with 5-AZA alone [117]. The PL is prepared by several freezing and thawing cycles of platelet concentrate or platelet-rich plasma samples, resulting in mechanical disruption and lysis of the platelets [115, 117]. To induce cardiomyogenic differentiation, the MSCs are incubated in a cardiomyogenic-induced medium containing 10 µM 5-AZA for 24 h only. After that, the cells are incubated in a basal growth medium containing 20% hPL for three weeks. The hPL medium is changed every three days [105, 117, 119].

The cardiomyogenic markers that indicate the differentiation of MSCs into cardiomyocytes are divided into two categories based on the differentiation stage [100]. Markers of early differentiation, which indicate the initiation of transformation of MSCs into cardiomyocytes, and markers of late differentiation, which indicate the maturation of cardiomyocytes. The essential markers of cardiomyogenic initiation are the GATA-binding protein 4 (GATA4) and the homeobox protein Nkx2.5 [100, 119, 120]. GATA4 belongs to the zinc finger family of transcription factors that bind to the specific DNA sequence “GATA” [121, 122]. It is well established that GATA4 is an essential factor in inducing differentiation of functionally beating cardiomyocytes in both Xenopus and mammalian systems [123]. Nkx2.5 is a homeodomain transcription factor essential in cardiomyogenesis and cardiac specification [120, 124, 125]. Moreover, mutations in Nkx2.5 are a common cause of congenital heart disease [120].

The main markers of late differentiation or cardiomyocyte maturation are cardiac troponin T (cTnT) and connexin-43 (Cx43) [100, 117]. The cTnT is one of three troponin subunits (C, I, and T) that form the troponin protein complex. This complex is a component of the cardiac thin myofilaments and plays a crucial role in cardiac muscle contraction [126]. Mutations in the cTnT gene can lead to hypertrophic cardiomyopathy [127]. Cx43 is a prominent transmembrane protein in cardiomyocytes that is also known as gap junction alpha-1 (GJA1) protein because it is encoded by the GJA1 gene [128, 129]. It forms conductive channels between cardiomyocytes that allow the intercellular exchange of ions and metabolites [130, 131]. The cTnT is found in the cytoplasm, while Cx43 is localized to the surface membrane of cardiomyocytes [117]. Additional cardiac markers include desmin and A-type natriuretic peptide (Fig. 7).

The identification of a resident population of cardiac multipotent stem cells that are pre-committed to the cardiac lineage has reformed the stem cell-based therapeutic approaches in IHD [132, 133]. These are known as cardiac progenitor cells (CPCs), and they can differentiate into all types of cardiac cells, including cardiomyocytes, endothelial cells, and vascular smooth muscle cells [134]. Under normal conditions, CPCs are quiescent in their cardiac niche. Upon cardiac damage or stimulation, they become active, proliferate, and differentiate to repair the damaged tissue [133, 135]. They are characterized and subclassified by their expression of different surface markers. The most common markers are the tyrosine kinase receptor c-kit (CD117), the stem cell antigen 1 (Sca-1), and the insulin gene enhancer protein ISL-1 [132, 133, 135]. It is anticipated that CPCs have a higher cardiac regenerative potential than MSCs. Experimental research showed that CPCs can reduce infarct size, improve cardiac function, and promote the release of growth and angiogenic factors in the hearts of different animal models [136,137,138]. However, the extent of cardiac damage usually exceeds the capacity of endogenous CPCs to repair due to their very limited numbers [135]. Therefore, several studies have investigated the ex-vivo production of CPCs from different cell sources [132]. Currently, we are focusing on the in vitro differentiation of perinatal MSCs into CPCs and the therapeutic potential of these descendant cardiac lineage cells in IHD.

Neurodegenerative diseases (NDDs)

Neurodegenerative diseases (NDDs) are a group of debilitating disorders characterized by the progressive loss of structure and function in the nervous system. While these conditions are commonly associated with aging, they can occur at any stage of life due to genetic, environmental, or idiopathic factors. Some of the most well-known NDDs include Parkinson’s disease, Multiple Sclerosis, Alzheimer’s disease, Huntington’s disease, and amyotrophic lateral sclerosis. There is increasing interest in MSCs due to their regenerative, immunomodulatory, and neuroprotective properties, especially since curative options are limited.

Parkinson’s disease (PD) is a progressive neurodegenerative disorder characterized by a loss of motor function and the presence of tremors. The pathology of the disease involves the accumulation of misfolded α-synuclein aggregates within dopaminergic (DA) neurons, which form Lewy bodies. This buildup disrupts synaptic transmission and affects dopamine regulation [139], eventually resulting in damage and death of DA neurons in the substantia nigra pars compacta and noradrenergic neurons in the locus coeruleus of the brain [140]. The DA neurons produce and release dopamine, a neurotransmitter that relays signals controlling motor functions. Additionally, noradrenergic neurons are part of the autonomic nervous system, which, when activated by noradrenaline, is responsible for alertness [141, 142]. Since these brain areas control motor function, patients with later-stage PD exhibit symptoms such as muscle tremors, rigidity, slow movements, bradykinesia, and other motor dysfunctions [140]. PD is the second most common neurodegenerative disease and is projected to affect nearly 1.2 million people in the next 20 years; it significantly impacts the quality of life of individuals affected and causes a socioeconomic burden that accounts for almost 14.4 billion dollars annually in the United States [143]. The onset of PD symptoms commonly occurs after the age of 60 and affects about 1% of that age group (> 60 years) [144]. Although there are treatments for this disease, they are not very effective and cannot reverse or stop the progression of the disease.

Currently, there is no cure for PD. However, treatment options, such as pharmaceuticals and surgical intervention, are available to help PD patients manage their symptoms. These treatments include medications that either increase the dopamine levels in the brain or mimic dopamine’s effects on the brain, such as dopamine precursors or dopamine agonists, in addition to surgical interventions such as deep brain stimulation [145]. Although these therapies can relieve some of the symptoms of PD, they cannot reverse or stop the damaging effects of the disease. The limited effectiveness of these drugs in halting disease progression and the anticipated increase in disease prevalence in future generations exhibit an immense need to develop a more effective treatment to stop and reverse the pathology of PD. We hypothesize that MSCs and DA neuron progenitors (DAPs) can halt neurodegeneration and provide neuro-regenerative effects.

We examined the effect of cell therapy on the 6-OHDA toxin-induced PD rat model. The toxin induces neural cell death via oxidative stress. Latchoumycandane et al. found that some of these pathways are via free radical generation, mitochondrial dysfunction, cytochrome c release, activation of caspase-9 and caspase-3, proteolytic activation PKCδ, and DNA fragmentation [146]. In earlier studies, perinatal MSCs have been shown to decrease oxidative stress by reducing levels of malondialdehyde and enhancing superoxide dismutase, glutathione, and the antioxidant enzyme glutathione peroxidase [147]. This may be why the MSC-treated animals displayed decreased necrosing DA neurons and increased tyrosine hydroxylase-positive (TH+) cells. Tyrosine hydroxylase (TH) is an important enzyme needed to produce dopamine. It acts by converting the amino acid tyrosine into L-DOPA, a precursor of dopamine. Our studies showed that DAPs are more effective than MSCs in mitigating disease symptoms in PD rats (unpublished results). This might be attributed to the high levels of neurotrophic factors GDNF and BDNF expressed by DAPs, which are known to promote neuroprotection in DA neurons. Additionally, the ability of DAPs to integrate into the damaged substantia nigra pars compacta and differentiate into DA neurons may contribute to their more promising results compared to MSCs.

On the other hand, multiple sclerosis (MS) is a chronic autoimmune disorder that affects the central nervous system (CNS) and results in demyelination, neuroinflammation, and axonal degeneration. MS presents a unique challenge due to its inflammatory and demyelinating characteristics, especially in young adults [148, 149]. The illness features either relapsing-remitting phases or progressive forms, significantly affecting motor, sensory, and cognitive functions [150, 151]. It is believed to be caused by immune-mediated attacks on myelin, the protective covering of nerve fibers. This condition arises from an unusual interaction between genetic factors and environmental triggers. Clinically, MS is characterized by symptoms such as fatigue, muscle weakness, spasticity, visual disturbances, and cognitive impairments, which can lead to varying levels of disability [152,153,154,155]. Although there have been advancements in disease-modifying therapies aimed at reducing inflammation and slowing disease progression, these treatments primarily focus on the immune response. They do not address the underlying neurodegeneration or promote repair mechanisms [156, 157]. This has generated interest in stem cell-based therapies, particularly MSCs because they can modulate the immune response, promote remyelination, and repair neuronal damage [158,159,160].

The MSCs derived from perinatal sources have significant potential for treating MS, due to their unique biological properties, as previously mentioned. One of the primary mechanisms MSCs exert their therapeutic effects on MS is through immunomodulation [161, 162]. MS is driven by autoreactive T cells, particularly Th1 and Th17 subtypes, which attack the myelin sheath surrounding neurons. MSCs can suppress these pathogenic cells by secreting anti-inflammatory cytokines, including interleukin-10 (IL-10), TGF-β, and prostaglandin E2 (PGE2) [161,162,163,164]. Additionally, MSCs enhance the differentiation of regulatory T cells and reduce the activation of dendritic cells and macrophages, effectively shifting the immune response towards a more tolerogenic state [163,164,165].

In addition to immunomodulation, MSCs also contribute significantly to the remyelination process in MS. These cells can differentiate into oligodendrocyte progenitor-like cells, which further mature into oligodendrocytes, the myelin-producing cells in the CNS [166]. Furthermore, MSCs secrete a range of growth factors, such as BDNF, NGF, and GDNF [167, 168]. These factors enhance the survival and functionality of existing oligodendrocytes and support neuronal health by contributing to the repair of damaged neural tissue [169].

MSCs play a crucial role in remyelination and in offering neuroprotective benefits. They achieve this by secreting anti-apoptotic factors and releasing exosomes that contain microRNAs, proteins, and lipids, all supporting neuronal survival and aiding in axonal repair. By reducing oxidative stress and inflammation in the CNS, MSCs help create an environment favorable for effective tissue repair [170,171,172,173,174,175,176,177]. Additionally, the components of the extracellular matrix derived from MSCs, including laminin and fibronectin, support the structural integrity of neural networks and enhance their neuroprotective role [178].

Another critical mechanism MSCs aid in MS treatment is angiogenesis and restoring the blood-brain barrier (BBB). MSCs secrete VEGF and HGF, which enhance vascular repair and reduce BBB permeability [179,180,181]. This is especially crucial in multiple sclerosis, where BBB dysfunction permits immune cells to enter the central nervous system, worsening the disease [161]. Several preclinical studies provide substantial evidence supporting the efficacy of primitive MSCs in animal models of MS, such as experimental autoimmune encephalomyelitis [182,183,184,185]. These studies have demonstrated reductions in clinical scores, improved motor function, and enhanced remyelination. Similarly, clinical trials in MS patients have reported promising outcomes, including significant improvements in disability scores and reductions in relapse rates. However, the heterogeneity in trial designs, cell sources, doses, and administration routes present challenges in drawing definitive conclusions about MSC therapy’s overall effectiveness [186,187,188].

Intravenous (IV) delivery is the most commonly used method for MSC transplantation in MS due to its safety and practicality. However, intrathecal (IT) administration, which involves directly injecting the cells into the cerebrospinal fluid, may offer enhanced therapeutic effects by improving MSC availability within the CNS [187]. Optimal dosing regimens are still actively being investigated, with new evidence suggesting that repeated administrations may be needed to maintain therapeutic benefits over time [189,190,191].

While there is significant potential for MSCs in treating MS, several challenges persist. The long-term safety and effectiveness of MSC therapies, especially for chronic neurodegenerative diseases, are still not completely understood [186, 192]. Additionally, variability in MSC isolation, expansion, and delivery protocols hinders the standardization required for broad clinical adoption [161, 193]. We believe more research is needed to elucidate the precise mechanisms underlying MSC-mediated neuroregeneration and immunomodulation. Additionally, novel strategies to enhance MSC functionality, such as preconditioning with hypoxia or bioactive molecules, could further improve their efficacy [162, 186].

Conclusion

This review provides the fundamental understanding of perinatal MSCs that is essential for future experimental research and therapeutic applications of these cells. It revises the main biological characteristics that make perinatal MSCs superior to stem cells from other sources. The main advantages of perinatal MSCs over MSCs from other sources can be summarized by their high proliferative capability, low immunogenicity and tumorigenesis, large-scale availability, and easy accessibility without major ethical concerns. These advantages render the perinatal MSCs a more favorable position, making them the center of attention for both medical researchers and clinicians. However, no single molecular marker can definitely identify MSCs, and a combination of several biological markers is a must. We recommend using at least two positive markers of stemness (CD29, CD73, CD90, and/or CD105) and two negative markers to exclude the hematopoietic origin of the cells (specifically CD34 and CD45).

In the realm of therapeutic applications, perinatal MSCs could be a promising solution for degenerative disorders such as degenerative disc disease, retinal degenerative diseases, ischemic heart disease, and neurodegenerative diseases. Scientific research indicates that more specialized progenitor cells derived from perinatal MSCs in vitro can provide a more potent regenerative response than the original undifferentiated MSCs. As we advance, it is crucial to continue this research trajectory, progressing from laboratory experiments to practical applications, in order to fully exploit the regenerative abilities of perinatal MSCs. This can revolutionize the field of regenerative medicine.

Data availability

Not applicable.

Abbreviations

5-AZA:

5-azacytidine

AA:

Amino acid

BBB:

Blood-brain barrier

BDNF:

Brain-derived neurotrophic factor

BM:

Bone marrow

BMP:

Bone morphogenetic protein

CD:

Cluster of differentiation

CNS:

Central nervous system

CNTF:

Ciliary neurotrophic factor

CPCs:

Cardiac progenitor cells

ChPCs:

Chondrogenic progenitor cells

CSCs:

Cancer stem cells

DA:

Dopaminergic

DDD:

Degenerative disc disease

ECM:

Extracellular matrix

EGF:

Epidermal growth factor

ESCs:

Embryonic stem cells

GDF:

Growth and differentiation factor

GDNF:

Glial cell-derived neurotrophic factor

HGF:

Hepatocyte growth factor

HSCs:

Hematopoietic stem cells

IGF-1:

Insulin-like growth factor 1

IHD:

Ischemic heart disease

IL:

Interleukin

ISCT:

International Society for Cellular Therapy

IVD:

Intervertebral disc

MI:

Myocardial infarction

MS:

Multiple sclerosis

MSCs:

Mesenchymal stem cells

NP:

Nucleus pulposus

NPCs:

Nucleus pulposus-like cells

PD:

Parkinson’s disease

PDGF:

Platelet-derived growth factor

PEGF:

Pigment epithelium growth factor

PL:

Platelet lysate

RDDs:

Retinal degenerative diseases

RPCs:

Retinal progenitor cells

Sca-1:

Stem cell antigen 1

TGF-β:

Transforming growth factor beta

TIMPs:

Tissue inhibitors of metalloproteinases

UC:

Umbilical cord

UC-MSCs:

Umbilical cord-derived mesenchymal stem cells

VEGF:

Vascular endothelial growth factor

WJ:

Wharton’s jelly

References

  1. Brown C, McKee C, Bakshi S, Walker K, Hakman E, Halassy S, et al. Mesenchymal stem cells: cell therapy and regeneration potential. J Tissue Eng Regen Med. 2019;13:1738–55.

    Article  CAS  PubMed  Google Scholar 

  2. 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;8:315–7.

    Article  CAS  PubMed  Google Scholar 

  3. Beeravolu N, Brougham J, Khan I, McKee C, Perez-Cruet M, Chaudhry GR. Human umbilical cord derivatives regenerate intervertebral disc. J Tissue Eng Regen Med. 2018;12:e579–91.

    Article  CAS  PubMed  Google Scholar 

  4. Beeravolu N, McKee C, Alamri A, Mikhael S, Brown C, Perez-Cruet M et al. Isolation and characterization of mesenchymal stromal cells from human umbilical cord and fetal placenta. J Vis Exp JoVE. 2017;55224.

  5. Espinoza N, Peterson M. How to depolarise the ethical debate over human embryonic stem cell research (and other ethical debates too!). J Med Ethics. 2012;38:496–500.

    Article  PubMed  Google Scholar 

  6. Volarevic V, Markovic BS, Gazdic M, Volarevic A, Jovicic N, Arsenijevic N, et al. Ethical and safety issues of stem Cell-Based therapy. Int J Med Sci. 2018;15:36–45.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Ma J, Wu J, Han L, Jiang X, Yan L, Hao J, et al. Comparative analysis of mesenchymal stem cells derived from amniotic membrane, umbilical cord, and chorionic plate under serum-free condition. Stem Cell Res Ther. 2019;10:19.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Deuse T, Stubbendorff M, Tang-Quan K, Phillips N, Kay MA, Eiermann T, et al. Immunogenicity and Immunomodulatory properties of umbilical cord lining mesenchymal stem cells. Cell Transpl. 2011;20:655–67.

    Article  Google Scholar 

  9. Teoh PL, Mohd Akhir H, Abdul Ajak W, Hiew VV. Human mesenchymal stromal cells derived from perinatal tissues: sources, characteristics and isolation methods. Malays J Med Sci MJMS. 2023;30:55–68.

    Article  PubMed  Google Scholar 

  10. Hass R, Kasper C, Böhm S, Jacobs R. Different populations and sources of human mesenchymal stem cells (MSC): A comparison of adult and neonatal tissue-derived MSC. Cell Commun Signal CCS. 2011;9:12.

    Article  CAS  PubMed  Google Scholar 

  11. Allard B, Cousineau I, Spring K, Stagg J. Chapter Fifteen - Measurement of CD73 enzymatic activity using luminescence-based and colorimetric assays. In: Galluzzi L, Rudqvist N-P, editors. Methods Enzymol [Internet]. Academic Press; 2019 [cited 2023 May 23]. pp. 269–89. Available from: https://www.sciencedirect.com/science/article/pii/S0076687919304094

  12. Misumi Y, Ogata S, Ohkubo K, Hirose S, Ikehara Y. Primary structure of human placental 5′-nucleotidase and identification of the glycolipid anchor in the mature form. Eur J Biochem. 1990;191:563–9.

    Article  CAS  PubMed  Google Scholar 

  13. Allard B, Longhi MS, Robson SC, Stagg J. The ectonucleotidases CD39 and CD73: novel checkpoint inhibitor targets. Immunol Rev. 2017;276:121–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Tan K, Zhu H, Zhang J, Ouyang W, Tang J, Zhang Y, et al. CD73 expression on mesenchymal stem cells dictates the reparative properties via its Anti-Inflammatory activity. Stem Cells Int. 2019;2019:8717694.

    Article  PubMed  PubMed Central  Google Scholar 

  15. Rege TA, Hagood JS. Thy-1 as a regulator of cell-cell and cell-matrix interactions in axon regeneration, apoptosis, adhesion, migration, cancer, and fibrosis. FASEB J Off Publ Fed Am Soc Exp Biol. 2006;20:1045–54.

    CAS  Google Scholar 

  16. Yang J, Zhan X-Z, Malola J, Li Z-Y, Pawar JS, Zhang H-T, et al. The multiple roles of Thy-1 in cell differentiation and regeneration. Differ Res Biol Divers. 2020;113:38–48.

    CAS  Google Scholar 

  17. Reif AE, Allen JMV, THE AKR THYMIC ANTIGEN, AND ITS DISTRIBUTION IN LEUKEMIAS AND NERVOUS TISSUES. J Exp Med. 1964;120:413–33.

  18. Sauzay C, Voutetakis K, Chatziioannou A, Chevet E, Avril T. CD90/Thy-1, a Cancer-Associated cell surface signaling molecule. Front Cell Dev Biol. 2019;7:66.

    Article  PubMed  PubMed Central  Google Scholar 

  19. Connor EV, Saygin C, Braley C, Wiechert AC, Karunanithi S, Crean-Tate K, et al. Thy-1 predicts poor prognosis and is associated with self-renewal in ovarian cancer. J Ovarian Res. 2019;12:112.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Shaikh MV, Kala M, Nivsarkar M. CD90 a potential cancer stem cell marker and a therapeutic target. Cancer Biomark Sect Dis Markers. 2016;16:301–7.

    Article  CAS  Google Scholar 

  21. Craig W, Kay R, Cutler R, Lansdrop P. Expression of Thy-1 on human hematopoietic progenitor cells. J Exp Med. 1993;177:1331–42.

    Article  CAS  PubMed  Google Scholar 

  22. Majeti R, Park CY, Weissman IL. Identification of a hierarchy of multipotent hematopoietic progenitors in human cord blood. Cell Stem Cell. 2007;1:635–45.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Lin C-S, Xin Z-C, Dai J, Lue TF. Commonly used mesenchymal stem cell markers and tracking labels: limitations and challenges. Histol Histopathol. 2013;28:1109–16.

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Schoonderwoerd MJA, Goumans M-JTH, Hawinkels LJAC. Endoglin: beyond the endothelium. Biomolecules. 2020;10:289.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Gougos A, Letarte M. Primary structure of endoglin, an RGD-containing glycoprotein of human endothelial cells. J Biol Chem. 1990;265:8361–4.

    Article  CAS  PubMed  Google Scholar 

  26. Nassiri F, Cusimano MD, Scheithauer BW, Rotondo F, Fazio A, Yousef GM, et al. Endoglin (CD105): a review of its role in angiogenesis and tumor diagnosis, progression and therapy. Anticancer Res. 2011;31:2283–90.

    CAS  PubMed  Google Scholar 

  27. Lv X-J, Zhou G-D, Liu Y, Liu X, Chen J-N, Luo X-S, et al. In vitro proliferation and differentiation of adipose-derived stem cells isolated using anti-CD105 magnetic beads. Int J Mol Med. 2012;30:826–34.

    Article  CAS  PubMed  Google Scholar 

  28. Park SB, Seo KW, So AY, Seo MS, Yu KR, Kang SK, et al. SOX2 has a crucial role in the lineage determination and proliferation of mesenchymal stem cells through Dickkopf-1 and c-MYC. Cell Death Differ. 2012;19:534–45.

    Article  CAS  PubMed  Google Scholar 

  29. Świstowska M, Gil-Kulik P, Krzyżanowski A, Bielecki T, Czop M, Kwaśniewska A, et al. Potential effect of SOX2 on the cell cycle of Wharton’s jelly stem cells (WJSCs). Oxid Med Cell Longev. 2019;2019:5084689.

    Article  PubMed  PubMed Central  Google Scholar 

  30. Yoon DS, Kim YH, Jung HS, Paik S, Lee JW. Importance of Sox2 in maintenance of cell proliferation and multipotency of mesenchymal stem cells in low-density culture. Cell Prolif. 2011;44:428–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Gil-Kulik P, Świstowska M, Krzyżanowski A, Petniak A, Kwaśniewska A, Płachno BJ, et al. Evaluation of the impact of Pregnancy-Associated factors on the quality of Wharton’s Jelly-Derived stem cells using SOX2 gene expression as a marker. Int J Mol Sci. 2022;23:7630.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Zhang S, Cui W. Sox2, a key factor in the regulation of pluripotency and neural differentiation. World J Stem Cells. 2014;6:305–11.

    Article  PubMed  PubMed Central  Google Scholar 

  33. Go MJ, Takenaka C, Ohgushi H. Forced expression of Sox2 or Nanog in human bone marrow derived mesenchymal stem cells maintains their expansion and differentiation capabilities. Exp Cell Res. 2008;314:1147–54.

    Article  CAS  PubMed  Google Scholar 

  34. Das BC, Tyagi A. Chapter 23 - Stem cells: A trek from laboratory to clinic to industry. In: Verma AS, Singh A, editors. Anim Biotechnol. San Diego: Academic; 2014. pp. 425–50.

    Chapter  Google Scholar 

  35. Gawlik-Rzemieniewska N, Bednarek I. The role of NANOG transcriptional factor in the development of malignant phenotype of cancer cells. Cancer Biol Ther. 2016;17:1–10.

    Article  CAS  PubMed  Google Scholar 

  36. Mullin NP, Yates A, Rowe AJ, Nijmeijer B, Colby D, Barlow PN, et al. The pluripotency rheostat Nanog functions as a dimer. Biochem J. 2008;411:227–31.

    Article  CAS  PubMed  Google Scholar 

  37. Wang J, Levasseur DN, Orkin SH. Requirement of Nanog dimerization for stem cell self-renewal and pluripotency. Proc Natl Acad Sci U S A. 2008;105:6326–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Zhang W, Sui Y, Ni J, Yang T. Insights into the Nanog gene: A propeller for stemness in primitive stem cells. Int J Biol Sci. 2016;12:1372–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Chambers I, Colby D, Robertson M, Nichols J, Lee S, Tweedie S, et al. Functional expression cloning of Nanog, a pluripotency sustaining factor in embryonic stem cells. Cell. 2003;113:643–55.

    Article  CAS  PubMed  Google Scholar 

  40. Mitsui K, Tokuzawa Y, Itoh H, Segawa K, Murakami M, Takahashi K, et al. The Homeoprotein Nanog is required for maintenance of pluripotency in mouse epiblast and ES cells. Cell. 2003;113:631–42.

    Article  CAS  PubMed  Google Scholar 

  41. Balzano F, Bellu E, Basoli V, Dei Giudici S, Santaniello S, Cruciani S, et al. Lessons from human umbilical cord: gender differences in stem cells from Wharton’s jelly. Eur J Obstet Gynecol Reprod Biol. 2019;234:143–8.

    Article  CAS  PubMed  Google Scholar 

  42. Pirjali T, Azarpira N, Ayatollahi M, Aghdaie MH, Geramizadeh B, Talai T. Isolation and characterization of human mesenchymal stem cells derived from human umbilical cord Wharton’s jelly and amniotic membrane. Int J Organ Transpl Med. 2013;4:111–6.

    CAS  Google Scholar 

  43. Song Y, Lim J-Y, Lim T, Im K-I, Kim N, Nam Y-S, et al. Human mesenchymal stem cells derived from umbilical cord and bone marrow exert Immunomodulatory effects in different mechanisms. World J Stem Cells. 2020;12:1032–49.

    Article  PubMed  PubMed Central  Google Scholar 

  44. Han S, Zhao Y, Xiao Z, Han J, Chen B, Chen L, et al. The three-dimensional collagen scaffold improves the stemness of rat bone marrow mesenchymal stem cells. J Genet Genomics Yi Chuan Xue Bao. 2012;39:633–41.

    Article  CAS  PubMed  Google Scholar 

  45. Riekstina U, Cakstina I, Parfejevs V, Hoogduijn M, Jankovskis G, Muiznieks I, et al. Embryonic stem cell marker expression pattern in human mesenchymal stem cells derived from bone marrow, adipose tissue, heart and dermis. Stem Cell Rev Rep. 2009;5:378–86.

    Article  CAS  PubMed  Google Scholar 

  46. Tsai C-C, Su P-F, Huang Y-F, Yew T-L, Hung S-C. Oct4 and Nanog directly regulate Dnmt1 to maintain self-renewal and undifferentiated state in mesenchymal stem cells. Mol Cell. 2012;47:169–82.

    Article  CAS  PubMed  Google Scholar 

  47. Liu TM, Wu YN, Guo XM, Hui JHP, Lee EH, Lim B. Effects of ectopic Nanog and Oct4 overexpression on mesenchymal stem cells. Stem Cells Dev. 2009;18:1013–22.

    Article  CAS  PubMed  Google Scholar 

  48. Greco SJ, Liu K, Rameshwar P. Functional similarities among genes regulated by OCT4 in human mesenchymal and embryonic stem cells. Stem Cells Dayt Ohio. 2007;25:3143–54.

    Article  CAS  Google Scholar 

  49. Seo K-W, Lee S-R, Bhandari DR, Roh K-H, Park S-B, So A-Y, et al. OCT4A contributes to the stemness and multi-potency of human umbilical cord blood-derived multipotent stem cells (hUCB-MSCs). Biochem Biophys Res Commun. 2009;384:120–5.

    Article  CAS  PubMed  Google Scholar 

  50. Davies OG, Cooper PR, Shelton RM, Smith AJ, Scheven BA. Isolation of adipose and bone marrow mesenchymal stem cells using CD29 and CD90 modifies their capacity for osteogenic and adipogenic differentiation. J Tissue Eng. 2015;6:2041731415592356.

    Article  PubMed  PubMed Central  Google Scholar 

  51. Ode A, Kopf J, Kurtz A, Schmidt-Bleek K, Schrade P, Kolar P, et al. CD73 and CD29 concurrently mediate the mechanically induced decrease of migratory capacity of mesenchymal stromal cells. Eur Cell Mater. 2011;22:26–42.

    Article  CAS  PubMed  Google Scholar 

  52. Togarrati PP, Dinglasan N, Desai S, Ryan WR, Muench MO. CD29 is highly expressed on epithelial, myoepithelial, and mesenchymal stromal cells of human salivary glands. Oral Dis. 2018;24:561–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Zwolanek D, Flicker M, Kirstätter E, Zaucke F, van Osch GJVM, Erben RG. β1 integrins mediate attachment of mesenchymal stem cells to cartilage lesions. BioResearch Open Access. 2015;4:39–53.

    Article  PubMed  PubMed Central  Google Scholar 

  54. Popov C, Radic T, Haasters F, Prall WC, Aszodi A, Gullberg D, et al. Integrins Α2β1 and Α11β1 regulate the survival of mesenchymal stem cells on collagen I. Cell Death Dis. 2011;2:e186.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Li L, Guan Q, Dai S, Wei W, Zhang Y. Integrin Β1 increases stem cell survival and cardiac function after myocardial infarction. Front Pharmacol. 2017;8:135.

    PubMed  PubMed Central  Google Scholar 

  56. Ip JE, Wu Y, Huang J, Zhang L, Pratt RE, Dzau VJ. Mesenchymal stem cells use integrin beta1 not CXC chemokine receptor 4 for myocardial migration and engraftment. Mol Biol Cell. 2007;18:2873–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Wang H-S, Hung S-C, Peng S-T, Huang C-C, Wei H-M, Guo Y-J, et al. Mesenchymal stem cells in the Wharton’s jelly of the human umbilical cord. Stem Cells Dayt Ohio. 2004;22:1330–7.

    Article  Google Scholar 

  58. Alrefaei GI, Ayuob NN, Ali SS, Al-Karim S. Effects of maternal age on the expression of mesenchymal stem cell markers in the components of human umbilical cord. Folia Histochem Cytobiol. 2015;53:259–71.

    Article  CAS  PubMed  Google Scholar 

  59. Alrefaei GI, Al-Karim S, Ayuob NN, Ali SS. Does the maternal age affect the mesenchymal stem cell markers and gene expression in the human placenta? What is the evidence? Tissue Cell. 2015;47:406–19.

    Article  CAS  PubMed  Google Scholar 

  60. Yang Y, Hu M, Zhang Y, Li H, Miao Z. CD29 of human umbilical cord mesenchymal stem cells is required for expansion of CD34(+) cells. Cell Prolif. 2014;47:596–603.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Al-Obaide M, Ishmakej A, Brown C, Mazzella M, Agosta P, Perez-Cruet M, et al. The potential role of integrin alpha 6 in human mesenchymal stem cells. Front Genet. 2022;13:968228.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Krebsbach PH, Villa-Diaz LG. The role of integrin Α6 (CD49f) in stem cells: more than a conserved biomarker. Stem Cells Dev. 2017;26:1090–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Nieto-Nicolau N, de la Torre RM, Fariñas O, Savio A, Vilarrodona A, Casaroli-Marano RP. Extrinsic modulation of integrin Α6 and progenitor cell behavior in mesenchymal stem cells. Stem Cell Res. 2020;47:101899.

    Article  CAS  PubMed  Google Scholar 

  64. Yu K-R, Yang S-R, Jung J-W, Kim H, Ko K, Han DW, et al. CD49f enhances multipotency and maintains stemness through the direct regulation of OCT4 and SOX2. Stem Cells Dayt Ohio. 2012;30:876–87.

    Article  CAS  Google Scholar 

  65. Villa-Diaz LG, Kim JK, Laperle A, Palecek SP, Krebsbach PH. Inhibition of focal adhesion kinase signaling by integrin Α6β1 supports human pluripotent stem cell Self-Renewal. Stem Cells Dayt Ohio. 2016;34:1753–64.

    Article  CAS  Google Scholar 

  66. Richardson SM, Kalamegam G, Pushparaj PN, Matta C, Memic A, Khademhosseini A, et al. Mesenchymal stem cells in regenerative medicine: focus on articular cartilage and intervertebral disc regeneration. Methods San Diego Calif. 2016;99:69–80.

    Article  CAS  PubMed  Google Scholar 

  67. Bachmeier BE, Nerlich AG, Weiler C, Paesold G, Jochum M, Boos N. Analysis of tissue distribution of TNF-α, TNF-α-Receptors, and the activating TNF-α–Converting enzyme suggests activation of the TNF-α system in the aging intervertebral disc. Ann N Y Acad Sci. 2007;1096:44–54.

    Article  CAS  PubMed  Google Scholar 

  68. Hoyland JA, Le Maitre C, Freemont AJ. Investigation of the role of IL-1 and TNF in matrix degradation in the intervertebral disc. Rheumatology. 2008;47:809–14.

    Article  CAS  PubMed  Google Scholar 

  69. Le Maitre CL, Freemont AJ, Hoyland JA. The role of interleukin-1 in the pathogenesis of human intervertebral disc degeneration. Arthritis Res Ther. 2005;7:R732.

    Article  PubMed  PubMed Central  Google Scholar 

  70. Huang H, Liu X, Wang J, Suo M, Zhang J, Sun T, et al. Umbilical cord mesenchymal stem cells for regenerative treatment of intervertebral disc degeneration. Front Cell Dev Biol. 2023;11:1215698.

    Article  PubMed  PubMed Central  Google Scholar 

  71. Liang W, Han B, Hai Y, Sun D, Yin P. Mechanism of action of mesenchymal stem Cell-Derived exosomes in the intervertebral disc degeneration treatment and bone repair and regeneration. Front Cell Dev Biol. 2021;9:833840.

    Article  PubMed  Google Scholar 

  72. Xie L, Chen Z, Liu M, Huang W, Zou F, Ma X, et al. MSC-Derived exosomes protect vertebral endplate chondrocytes against apoptosis and calcification via the miR-31-5p/ATF6 Axis. Mol Ther Nucleic Acids. 2020;22:601–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Lozito TP, Tuan RS. Mesenchymal stem cells inhibit both endogenous and exogenous MMPs via secreted timps. J Cell Physiol. 2011;226:385–96.

    Article  CAS  PubMed  Google Scholar 

  74. Zhu G, Yang X, Peng C, Yu L, Hao Y. Exosomal miR-532-5p from bone marrow mesenchymal stem cells reduce intervertebral disc degeneration by targeting RASSF5. Exp Cell Res. 2020;393:112109.

    Article  CAS  PubMed  Google Scholar 

  75. Li Q, Yu H, Sun M, Yang P, Hu X, Ao Y, et al. The tissue origin effect of extracellular vesicles on cartilage and bone regeneration. Acta Biomater. 2021;125:253–66.

    Article  CAS  PubMed  Google Scholar 

  76. He C, Yang C, Zeng Q, Liu Z, Wang F, Chen Q, et al. Umbilical cord-derived mesenchymal stem cells cultured in the MCL medium for aplastic anemia therapy. Stem Cell Res Ther. 2023;14:224.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Sakai D, Andersson GBJ. Stem cell therapy for intervertebral disc regeneration: Obstacles and solutions. Nat Rev Rheumatol. 2015;11:243–56.

    Article  PubMed  Google Scholar 

  78. Perez-Cruet M, Beeravolu N, McKee C, Brougham J, Khan I, Bakshi S, et al. Potential of human nucleus Pulposus-Like cells derived from umbilical cord to treat degenerative disc disease. Neurosurgery. 2019;84:272–83.

    Article  PubMed  Google Scholar 

  79. Ahn J, Park E-M, Kim BJ, Kim J-S, Choi B, Lee S-H, et al. Transplantation of human Wharton’s jelly-derived mesenchymal stem cells highly expressing TGFβ receptors in a rabbit model of disc degeneration. Stem Cell Res Ther. 2015;6:190.

    Article  PubMed  PubMed Central  Google Scholar 

  80. Bertram H, Kroeber M, Wang H, Unglaub F, Guehring T, Carstens C, et al. Matrix-assisted cell transfer for intervertebral disc cell therapy. Biochem Biophys Res Commun. 2005;331:1185–92.

    Article  CAS  PubMed  Google Scholar 

  81. Growney Kalaf EA, Pendyala M, Bledsoe JG, Sell SA. Characterization and restoration of degenerated IVD function with an injectable, in situ gelling alginate hydrogel: an in vitro and ex vivo study. J Mech Behav Biomed Mater. 2017;72:229–40.

    Article  CAS  PubMed  Google Scholar 

  82. Huang Y-C, Hu Y, Li Z, Luk KDK. Biomaterials for intervertebral disc regeneration: current status and looming challenges. J Tissue Eng Regen Med. 2018;12:2188–202.

    Article  CAS  PubMed  Google Scholar 

  83. McKee C, Beeravolu N, Brown C, Perez-Cruet M, Chaudhry GR. Mesenchymal stem cells transplanted with self-assembling scaffolds differentiated to regenerate nucleus pulposus in an ex vivo model of degenerative disc disease. Appl Mater Today. 2020;18:100474.

    Article  Google Scholar 

  84. McKee C, Perez-Cruet M, Chaudhry R. A combinational approach using cell therapy, tissue engineering, and distracting device for the treatment of degenerative disc disease. HSOA J Stem Cell Res Dev Ther. 2021;7:064.

    Google Scholar 

  85. Brown C, Agosta P, McKee C, Walker K, Mazzella M, Alamri A, et al. Human primitive mesenchymal stem cell-derived retinal progenitor cells improved neuroprotection, neurogenesis, and vision in rd12 mouse model of retinitis pigmentosa. Stem Cell Res Ther. 2022;13:148.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Jin Z-B, Gao M-L, Deng W-L, Wu K-C, Sugita S, Mandai M, et al. Stemming retinal regeneration with pluripotent stem cells. Prog Retin Eye Res. 2019;69:38–56.

    Article  CAS  PubMed  Google Scholar 

  87. Labrador-Velandia S, Alonso-Alonso ML, Alvarez-Sanchez S, González-Zamora J, Carretero-Barrio I, Pastor JC, et al. Mesenchymal stem cell therapy in retinal and optic nerve diseases: an update of clinical trials. World J Stem Cells. 2016;8:376–83.

    Article  PubMed  PubMed Central  Google Scholar 

  88. Holan V, Palacka K, Hermankova B. Mesenchymal stem Cell-Based therapy for retinal degenerative diseases: experimental models and clinical trials. Cells. 2021;10:588.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Koh AE-H, Alsaeedi HA, Rashid MBA, Lam C, Harun MHN, Ng MH, et al. Transplanted Erythropoietin-Expressing mesenchymal stem cells promote Pro-survival gene expression and protect photoreceptors from sodium Iodate-Induced cytotoxicity in a retinal degeneration model. Front Cell Dev Biol. 2021;9:652017.

    Article  PubMed  PubMed Central  Google Scholar 

  90. Mohamed EM, Abdelrahman SA, Hussein S, Shalaby SM, Mosaad H, Awad AMB. Effect of human umbilical cord blood mesenchymal stem cells administered by intravenous or intravitreal routes on cryo-induced retinal injury. IUBMB Life. 2017;69:188–201.

    Article  CAS  PubMed  Google Scholar 

  91. Zhao T, Liang Q, Meng X, Duan P, Wang F, Li S, et al. Intravenous infusion of umbilical cord mesenchymal stem cells maintains and partially improves visual function in patients with advanced retinitis pigmentosa. Stem Cells Dev. 2020;29:1029–37.

    Article  CAS  PubMed  Google Scholar 

  92. Huang C, Zhang J, Ao M, Li Y, Zhang C, Xu Y, et al. Combination of retinal pigment epithelium cell-conditioned medium and photoreceptor outer segments stimulate mesenchymal stem cell differentiation toward a functional retinal pigment epithelium cell phenotype. J Cell Biochem. 2012;113:590–8.

    Article  CAS  PubMed  Google Scholar 

  93. Adak S, Magdalene D, Deshmukh S, Das D, Jaganathan BG. A review on mesenchymal stem cells for treatment of retinal diseases. Stem Cell Rev Rep. 2021;17:1154–73.

    Article  PubMed  PubMed Central  Google Scholar 

  94. Safwat A, Sabry D, Ragiae A, Amer E, Mahmoud RH, Shamardan RM. Adipose mesenchymal stem cells-derived exosomes attenuate retina degeneration of streptozotocin-induced diabetes in rabbits. J Circ Biomark. 2018;7:1849454418807827.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Yu B, Shao H, Su C, Jiang Y, Chen X, Bai L, et al. Exosomes derived from MSCs ameliorate retinal laser injury partially by Inhibition of MCP-1. Sci Rep. 2016;6:34562.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Mathew B, Ravindran S, Liu X, Torres L, Chennakesavalu M, Huang C-C, et al. Mesenchymal stem cell-derived extracellular vesicles and retinal ischemia-reperfusion. Biomaterials. 2019;197:146–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Wang Y, Tang Z, Gu P. Stem/progenitor cell-based transplantation for retinal degeneration: a review of clinical trials. Cell Death Dis. 2020;11:793.

    Article  PubMed  PubMed Central  Google Scholar 

  98. Bhattacharya S, Gangaraju R, Chaum E. Recent advances in retinal stem cell therapy. Curr Mol Biol Rep. 2017;3:172–82.

    Article  PubMed  PubMed Central  Google Scholar 

  99. Cotrim CC, Toscano L, Messias A, Jorge R, Siqueira RC. Intravitreal use of bone marrow mononuclear fraction containing CD34 + stem cells in patients with atrophic age-related macular degeneration. Clin Ophthalmol Auckl NZ. 2017;11:931–8.

    Article  Google Scholar 

  100. Chen Y, Shen H, Ding Y, Yu Y, Shao L, Shen Z. The application of umbilical cord-derived MSCs in cardiovascular diseases. J Cell Mol Med. 2021;25:8103–14.

    Article  PubMed  PubMed Central  Google Scholar 

  101. Golpanian S, Wolf A, Hatzistergos KE, Hare JM. Rebuilding the damaged heart: mesenchymal stem cells, Cell-Based therapy, and engineered heart tissue. Physiol Rev. 2016;96:1127–68.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Malik A, Chhabra L. Congestive heart failure. StatPearls. Treasure Island (FL): StatPearls Publishing; 2025.

    Google Scholar 

  103. Severino P, D’Amato A, Pucci M, Infusino F, Adamo F, Birtolo LI, et al. Ischemic heart disease pathophysiology paradigms overview: from plaque activation to microvascular dysfunction. Int J Mol Sci. 2020;21:8118.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Bartolucci J, Verdugo FJ, González PL, Larrea RE, Abarzua E, Goset C, et al. Safety and efficacy of the intravenous infusion of umbilical cord mesenchymal stem cells in patients with heart failure: A phase 1/2 randomized controlled trial (RIMECARD trial [Randomized Clinical trial of intravenous infusion umbilical cord mesenchymal stem cells on Cardiopathy]). Circ Res. 2017;121:1192–204.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Ruan Z-B, Zhu L, Yin Y-G, Chen G-C. The mechanism underlying the differentiation of human umbilical cord-derived mesenchymal stem cells into myocardial cells induced by 5-azacytidine. Indian J Med Sci. 2010;64:402–7.

    Article  PubMed  Google Scholar 

  106. López Y, Lutjemeier B, Seshareddy K, Trevino EM, Hageman KS, Musch TI, et al. Wharton’s jelly or bone marrow mesenchymal stromal cells improve cardiac function following myocardial infarction for more than 32 weeks in a rat model: a preliminary report. Curr Stem Cell Res Ther. 2013;8:46–59.

    Article  PubMed  Google Scholar 

  107. Szaraz P, Gratch YS, Iqbal F, Librach CL. In vitro differentiation of human mesenchymal stem cells into functional Cardiomyocyte-like cells. J Vis Exp JoVE. 2017;55757.

  108. Yannarelli G, Dayan V, Pacienza N, Lee C-J, Medin J, Keating A. Human umbilical cord perivascular cells exhibit enhanced cardiomyocyte reprogramming and cardiac function after experimental acute myocardial infarction. Cell Transpl. 2013;22:1651–66.

    Article  Google Scholar 

  109. Müller AMS, Florek M. 5-Azacytidine/5-Azacitidine. Recent Results Cancer Res Fortschr Krebsforsch Progres Dans Rech Sur Cancer. 2014;201:299–324.

    Article  Google Scholar 

  110. Bohl SR, Bullinger L, Rücker FG. Epigenetic therapy: Azacytidine and decitabine in acute myeloid leukemia. Expert Rev Hematol. 2018;11:361–71.

    Article  CAS  PubMed  Google Scholar 

  111. Katagiri T, Ushiki T, Masuko M, Tanaka T, Miyakoshi S, Fuse K, et al. Successful 5-azacytidine treatment of myeloid sarcoma and leukemia cutis associated with myelodysplastic syndrome: A case report and literature review. Med (Baltim). 2017;96:e7975.

    Article  Google Scholar 

  112. Qian Q, Qian H, Zhang X, Zhu W, Yan Y, Ye S, et al. 5-Azacytidine induces cardiac differentiation of human umbilical cord-derived mesenchymal stem cells by activating extracellular regulated kinase. Stem Cells Dev. 2012;21:67–75.

    Article  CAS  PubMed  Google Scholar 

  113. Burnouf T, Strunk D, Koh MBC, Schallmoser K. Human platelet lysate: replacing fetal bovine serum as a gold standard for human cell propagation? Biomaterials. 2016;76:371–87.

    Article  CAS  PubMed  Google Scholar 

  114. Doucet C, Ernou I, Zhang Y, Llense J-R, Begot L, Holy X, et al. Platelet lysates promote mesenchymal stem cell expansion: a safety substitute for animal serum in cell-based therapy applications. J Cell Physiol. 2005;205:228–36.

    Article  CAS  PubMed  Google Scholar 

  115. Naskou MC, Sumner SM, Chocallo A, Kemelmakher H, Thoresen M, Copland I, et al. Platelet lysate as a novel serum-free media supplement for the culture of equine bone marrow-derived mesenchymal stem cells. Stem Cell Res Ther. 2018;9:75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Schallmoser K, Bartmann C, Rohde E, Reinisch A, Kashofer K, Stadelmeyer E, et al. Human platelet lysate can replace fetal bovine serum for clinical-scale expansion of functional mesenchymal stromal cells. Transfus (Paris). 2007;47:1436–46.

    Article  CAS  Google Scholar 

  117. Markmee R, Aungsuchawan S, Tancharoen W, Narakornsak S, Pothacharoen P. Differentiation of cardiomyocyte-like cells from human amniotic fluid mesenchymal stem cells by combined induction with human platelet lysate and 5-azacytidine. Heliyon. 2020;6:e04844.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Lippross S, Loibl M, Hoppe S, Meury T, Benneker L, Alini M, et al. Platelet released growth factors boost expansion of bone marrow derived CD34(+) and CD133(+) endothelial progenitor cells for autologous grafting. Platelets. 2011;22:422–32.

    Article  CAS  PubMed  Google Scholar 

  119. Almalki SG, Agrawal DK. Key transcription factors in the differentiation of mesenchymal stem cells. Differ Res Biol Divers. 2016;92:41–51.

    CAS  Google Scholar 

  120. de Sena-Tomás C, Aleman AG, Ford C, Varshney A, Yao D, Harrington JK, et al. Activation of Nkx2.5 transcriptional program is required for adult myocardial repair. Nat Commun. 2022;13:2970.

    Article  PubMed  PubMed Central  Google Scholar 

  121. Gao J, Chen Y-H, Peterson LC. GATA family transcriptional factors: emerging suspects in hematologic disorders. Exp Hematol Oncol. 2015;4:28.

    Article  PubMed  PubMed Central  Google Scholar 

  122. Shimizu S, Sunagawa Y, Hajika N, Yorimitsu N, Katanasaka Y, Funamoto M, et al. Multimerization of the GATA4 transcription factor regulates transcriptional activity and cardiomyocyte hypertrophic response. Int J Biol Sci. 2022;18:1079–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Afouda BA, Lynch AT, de Paiva Alves E, Hoppler S. Genome-wide transcriptomics analysis identifies sox7 and sox18 as specifically regulated by gata4 in cardiomyogenesis. Dev Biol. 2018;434:108–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Prall OWJ, Menon MK, Solloway MJ, Watanabe Y, Zaffran S, Bajolle F, et al. An Nkx2-5/Bmp2/Smad1 negative feedback loop controls heart progenitor specification and proliferation. Cell. 2007;128:947–59.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Targoff KL, Schell T, Yelon D. Nkx genes regulate heart tube extension and exert differential effects on ventricular and atrial cell number. Dev Biol. 2008;322:314–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Katrukha IA. Human cardiac troponin complex. Structure and functions. Biochem Biokhimiia. 2013;78:1447–65.

    Article  CAS  Google Scholar 

  127. Moolman JC, Corfield VA, Posen B, Ngumbela K, Seidman C, Brink PA, et al. Sudden death due to troponin T mutations. J Am Coll Cardiol. 1997;29:549–55.

    Article  CAS  PubMed  Google Scholar 

  128. Paznekas WA, Karczeski B, Vermeer S, Lowry RB, Delatycki M, Laurence F, et al. GJA1 mutations, variants, and connexin 43 dysfunction as it relates to the oculodentodigital dysplasia phenotype. Hum Mutat. 2009;30:724–33.

    Article  CAS  PubMed  Google Scholar 

  129. Ren D, Zheng P, Zou S, Gong Y, Wang Y, Duan J, et al. GJA1-20K enhances mitochondria transfer from astrocytes to neurons via Cx43-TnTs after traumatic brain injury. Cell Mol Neurobiol. 2022;42:1887–95.

    Article  CAS  PubMed  Google Scholar 

  130. Bonacquisti EE, Nguyen J. Connexin 43 (Cx43) in cancer: implications for therapeutic approaches via gap junctions. Cancer Lett. 2019;442:439–44.

    Article  CAS  PubMed  Google Scholar 

  131. Boengler K, Schulz R. Connexin 43 and mitochondria in cardiovascular health and disease. Adv Exp Med Biol. 2017;982:227–46.

    Article  CAS  PubMed  Google Scholar 

  132. Barreto S, Hamel L, Schiatti T, Yang Y, George V. Cardiac progenitor cells from stem cells: learning from genetics and biomaterials. Cells. 2019;8:1536.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Shouman S, Zaher A, Abdelhameed A, Elshaboury S, Sakr S, Fouda BE, et al. Cardiac progenitor cells. Adv Exp Med Biol. 2021;1312:51–73.

    Article  CAS  PubMed  Google Scholar 

  134. Lyngbaek S, Schneider M, Hansen JL, Sheikh SP. Cardiac regeneration by resident stem and progenitor cells in the adult heart. Basic Res Cardiol. 2007;102:101–14.

    Article  PubMed  Google Scholar 

  135. Bollini S, Smart N, Riley PR. Resident cardiac progenitor cells: at the heart of regeneration. J Mol Cell Cardiol. 2011;50:296–303.

    Article  CAS  PubMed  Google Scholar 

  136. Dawn B, Stein AB, Urbanek K, Rota M, Whang B, Rastaldo R, et al. Cardiac stem cells delivered intravascularly traverse the vessel barrier, regenerate infarcted myocardium, and improve cardiac function. Proc Natl Acad Sci U S A. 2005;102:3766–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Linke A, Müller P, Nurzynska D, Casarsa C, Torella D, Nascimbene A, et al. Stem cells in the dog heart are self-renewing, clonogenic, and multipotent and regenerate infarcted myocardium, improving cardiac function. Proc Natl Acad Sci U S A. 2005;102:8966–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Maring JA, Lodder K, Mol E, Verhage V, Wiesmeijer KC, Dingenouts CKE, et al. Cardiac progenitor cell-Derived extracellular vesicles reduce infarct size and associate with increased cardiovascular cell proliferation. J Cardiovasc Transl Res. 2019;12:5–17.

    Article  PubMed  Google Scholar 

  139. Sivandzade F, Prasad S, Bhalerao A, Cucullo L. NRF2 and NF-қB interplay in cerebrovascular and neurodegenerative disorders: molecular mechanisms and possible therapeutic approaches. Redox Biol. 2019;21:101059.

    Article  CAS  PubMed  Google Scholar 

  140. Dickson DW. Parkinson’s disease and parkinsonism: neuropathology. Cold Spring Harb Perspect Med. 2012;2:a009258.

    Article  PubMed  PubMed Central  Google Scholar 

  141. Samuels ER, Szabadi E. Functional neuroanatomy of the noradrenergic locus coeruleus: its roles in the regulation of arousal and autonomic function part I: principles of functional organisation. Curr Neuropharmacol. 2008;6:235–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Ernsberger U, Rohrer H. Sympathetic Tales: subdivisons of the autonomic nervous system and the impact of developmental studies. Neural Develop. 2018;13:20.

    Article  Google Scholar 

  143. Kowal SL, Dall TM, Chakrabarti R, Storm MV, Jain A. The current and projected economic burden of Parkinson’s disease in the united States. Mov Disord Off J Mov Disord Soc. 2013;28:311–8.

    Article  Google Scholar 

  144. Tysnes O-B, Storstein A. Epidemiology of Parkinson’s disease. J Neural Transm Vienna Austria. 1996. 2017;124:901–5.

  145. Ellis JM, Fell MJ. Current approaches to the treatment of Parkinson’s disease. Bioorg Med Chem Lett. 2017;27:4247–55.

    Article  CAS  PubMed  Google Scholar 

  146. Latchoumycandane C, Anantharam V, Jin H, Kanthasamy A, Kanthasamy A. Dopaminergic neurotoxicant 6-OHDA induces oxidative damage through proteolytic activation of PKCδ in cell culture and animal models of Parkinson’s disease. Toxicol Appl Pharmacol. 2011;256:314–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Chi H, Guan Y, Li F, Chen Z. The effect of human umbilical cord mesenchymal stromal cells in protection of dopaminergic neurons from apoptosis by reducing oxidative stress in the early stage of a 6-OHDA-Induced Parkinson’s disease model. Cell Transpl. 2019;28:S87–99.

    Article  Google Scholar 

  148. Rekatsina M, Paladini A, Piroli A, Zis P, Pergolizzi JV, Varrassi G. Pathophysiology and therapeutic perspectives of oxidative stress and neurodegenerative diseases: A narrative review. Adv Ther. 2020;37:113–39.

    Article  PubMed  Google Scholar 

  149. Davis SE, Cirincione AB, Jimenez-Torres AC, Zhu J. The impact of neurotransmitters on the neurobiology of neurodegenerative diseases. Int J Mol Sci. 2023;24:15340.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Musella A, Gentile A, Rizzo FR, De Vito F, Fresegna D, Bullitta S, et al. Interplay between age and neuroinflammation in multiple sclerosis: effects on motor and cognitive functions. Front Aging Neurosci. 2018;10:238.

    Article  PubMed  PubMed Central  Google Scholar 

  151. Gómez-Melero S, Caballero-Villarraso J, Escribano BM, Galvao-Carmona A, Túnez I, Agüera-Morales E. Impact of cognitive impairment on quality of life in multiple sclerosis Patients-A comprehensive review. J Clin Med. 2024;13:3321.

    Article  PubMed  PubMed Central  Google Scholar 

  152. Filippi M, Bar-Or A, Piehl F, Preziosa P, Solari A, Vukusic S, et al. Multiple sclerosis. Nat Rev Dis Primer. 2018;4:43.

    Article  Google Scholar 

  153. Waubant E, Lucas R, Mowry E, Graves J, Olsson T, Alfredsson L, et al. Environmental and genetic risk factors for MS: an integrated review. Ann Clin Transl Neurol. 2019;6:1905–22.

    Article  PubMed  PubMed Central  Google Scholar 

  154. Zielińska-Nowak E, Włodarczyk L, Kostka J, Miller E. New strategies for rehabilitation and Pharmacological treatment of fatigue syndrome in multiple sclerosis. J Clin Med. 2020;9:3592.

    Article  PubMed  PubMed Central  Google Scholar 

  155. Cavallo S. Immune-mediated genesis of multiple sclerosis. J Transl Autoimmun. 2020;3:100039.

    Article  PubMed  Google Scholar 

  156. Cummings J. Disease modification and neuroprotection in neurodegenerative disorders. Transl Neurodegener. 2017;6:25.

    Article  PubMed  PubMed Central  Google Scholar 

  157. Müller T, Mueller BK, Riederer P. Perspective: treatment for disease modification in chronic neurodegeneration. Cells. 2021;10:873.

    Article  PubMed  PubMed Central  Google Scholar 

  158. De Gioia R, Biella F, Citterio G, Rizzo F, Abati E, Nizzardo M, et al. Neural stem cell transplantation for neurodegenerative diseases. Int J Mol Sci. 2020;21:3103.

    Article  PubMed  PubMed Central  Google Scholar 

  159. Boshuizen MCS, Steinberg GK. Stem Cell-Based Immunomodulation after stroke: effects on brain repair processes. Stroke. 2018;49:1563–70.

    Article  PubMed  PubMed Central  Google Scholar 

  160. Palanisamy CP, Pei J, Alugoju P, Anthikapalli NVA, Jayaraman S, Veeraraghavan VP, et al. New strategies of neurodegenerative disease treatment with extracellular vesicles (EVs) derived from mesenchymal stem cells (MSCs). Theranostics. 2023;13:4138–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Dadfar S, Yazdanpanah E, Pazoki A, Nemati MH, Eslami M, Haghmorad D, et al. The role of mesenchymal stem cells in modulating adaptive immune responses in multiple sclerosis. Cells. 2024;13:1556.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Rizvi SFA, Wasim B, Usman S, Borges KJJ, Sahibdad I, Salim A, et al. Zinc and hypoxic preconditioning: a strategy to enhance the functionality and therapeutic potential of bone marrow-derived mesenchymal stem cells. Mol Cell Biochem. 2022;477:2735–49.

    Article  CAS  PubMed  Google Scholar 

  163. Baharlooi H, Salehi Z, Minbashi Moeini M, Rezaei N, Azimi M. Immunomodulatory potential of human mesenchymal stem cells and their exosomes on multiple sclerosis. Adv Pharm Bull. 2022;12:389–97.

    CAS  PubMed  Google Scholar 

  164. Abbasi-Kangevari M, Ghamari S-H, Safaeinejad F, Bahrami S, Niknejad H. Potential therapeutic features of human amniotic mesenchymal stem cells in multiple sclerosis: Immunomodulation, inflammation suppression, angiogenesis promotion, oxidative stress Inhibition, neurogenesis induction, MMPs regulation, and remyelination stimulation. Front Immunol. 2019;10:238.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  165. 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;20:263.

    Article  PubMed  PubMed Central  Google Scholar 

  166. Rivera FJ, Aigner L. Adult mesenchymal stem cell therapy for Myelin repair in multiple sclerosis. Biol Res. 2012;45:257–68.

    Article  PubMed  Google Scholar 

  167. Crigler L, Robey RC, Asawachaicharn A, Gaupp D, Phinney DG. Human mesenchymal stem cell subpopulations express a variety of neuro-regulatory molecules and promote neuronal cell survival and neuritogenesis. Exp Neurol. 2006;198:54–64.

    Article  CAS  PubMed  Google Scholar 

  168. De la Bierlein M, Sharma AD, Mallapragada SK, Sakaguchi DS. Transdifferentiation of brain-derived neurotrophic factor (BDNF)-secreting mesenchymal stem cells significantly enhance BDNF secretion and Schwann cell marker proteins. J Biosci Bioeng. 2017;124:572–82.

    Article  Google Scholar 

  169. Kurozumi K, Nakamura K, Tamiya T, Kawano Y, Ishii K, Kobune M, et al. Mesenchymal stem cells that produce neurotrophic factors reduce ischemic damage in the rat middle cerebral artery occlusion model. Mol Ther. 2005;11:96–104.

    Article  CAS  PubMed  Google Scholar 

  170. Ahn SY, Sung DK, Kim YE, Sung S, Chang YS, Park WS. Brain-derived neurotropic factor mediates neuroprotection of mesenchymal stem cell‐derived extracellular vesicles against severe intraventricular hemorrhage in newborn rats. Stem Cells Transl Med. 2020;10:374–84.

    Article  PubMed  PubMed Central  Google Scholar 

  171. Han M, Cao Y, Guo X, Chu X, Li T, Xue H, et al. Mesenchymal stem cell-derived extracellular vesicles promote microglial M2 polarization after subarachnoid hemorrhage in rats and involve the AMPK/NF-κB signaling pathway. Biomed Pharmacother Biomedecine Pharmacother. 2021;133:111048.

    Article  CAS  Google Scholar 

  172. Han M, Cao Y, Xue H, Chu X, Li T, Xin D, et al. Neuroprotective effect of mesenchymal stromal Cell-Derived extracellular vesicles against cerebral Ischemia-Reperfusion-Induced neural functional injury: A pivotal role for AMPK and JAK2/STAT3/NF-κB signaling pathway modulation. Drug Des Devel Ther. 2020;14:2865–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  173. Losurdo M, Pedrazzoli M, D’Agostino C, Elia CA, Massenzio F, Lonati E, et al. Intranasal delivery of mesenchymal stem cell-derived extracellular vesicles exerts Immunomodulatory and neuroprotective effects in a 3xTg model of Alzheimer’s disease. Stem Cells Transl Med. 2020;9:1068–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  174. Turovsky EA, Golovicheva VV, Varlamova EG, Danilina TI, Goryunov KV, Shevtsova YA, et al. Mesenchymal stromal cell-derived extracellular vesicles afford neuroprotection by modulating PI3K/AKT pathway and calcium oscillations. Int J Biol Sci. 2022;18:5345–68.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  175. Wang C, Börger V, Sardari M, Murke F, Skuljec J, Pul R, et al. Mesenchymal stromal Cell-Derived small extracellular vesicles induce ischemic neuroprotection by modulating leukocytes and specifically neutrophils. Stroke. 2020;51:1825–34.

    Article  CAS  PubMed  Google Scholar 

  176. Xin D, Li T, Chu X, Ke H, Yu Z, Cao L, et al. Mesenchymal stromal cell-derived extracellular vesicles modulate microglia/macrophage polarization and protect the brain against hypoxia-ischemic injury in neonatal mice by targeting delivery of miR-21a-5p. Acta Biomater. 2020;113:597–613.

    Article  CAS  PubMed  Google Scholar 

  177. Zhou X, Chu X, Yuan H, Qiu J, Zhao C, Xin D, et al. Mesenchymal stem cell derived EVs mediate neuroprotection after spinal cord injury in rats via the microRNA-21-5p/FasL gene axis. Biomed Pharmacother Biomedecine Pharmacother. 2019;115:108818.

    Article  CAS  Google Scholar 

  178. Jarmalavičiūtė A, Pivoriūnas A. Neuroprotective properties of extracellular vesicles derived from mesenchymal stem cells. Neural Regen Res. 2016;11:904–5.

    Article  PubMed  PubMed Central  Google Scholar 

  179. Bronckaers A, Hilkens P, Martens W, Gervois P, Ratajczak J, Struys T, et al. Mesenchymal stem/stromal cells as a Pharmacological and therapeutic approach to accelerate angiogenesis. Pharmacol Ther. 2014;143:181–96.

    Article  CAS  PubMed  Google Scholar 

  180. Ortiz GG, Pacheco-Moisés FP, Macías-Islas MÁ, Flores-Alvarado LJ, Mireles-Ramírez MA, González-Renovato ED, et al. Role of the blood-brain barrier in multiple sclerosis. Arch Med Res. 2014;45:687–97.

    Article  CAS  PubMed  Google Scholar 

  181. Tashima T. Mesenchymal stem cell (MSC)-Based drug delivery into the brain across the Blood-Brain barrier. Pharmaceutics. 2024;16:289.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  182. Clark K, Zhang S, Barthe S, Kumar P, Pivetti C, Kreutzberg N, et al. Placental mesenchymal stem Cell-Derived extracellular vesicles promote Myelin regeneration in an animal model of multiple sclerosis. Cells. 2019;8:1497.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  183. Ghareghani M, Arneaud A, Rivest S. The evolution of mesenchymal stem cell-derived neural progenitor therapy for multiple sclerosis: from concept to clinic. Front Cell Neurosci. 2024;18:1428652.

    Article  PubMed  PubMed Central  Google Scholar 

  184. Gramlich OW, Brown AJ, Godwin CR, Chimenti MS, Boland LK, Ankrum JA, et al. Systemic mesenchymal stem cell treatment mitigates structural and functional retinal ganglion cell degeneration in a mouse model of multiple sclerosis. Transl Vis Sci Technol. 2020;9:16.

    Article  PubMed  PubMed Central  Google Scholar 

  185. Liao W, Pham V, Liu L, Riazifar M, Pone EJ, Zhang SX, et al. Mesenchymal stem cells engineered to express selectin ligands and IL-10 exert enhanced therapeutic efficacy in murine experimental autoimmune encephalomyelitis. Biomaterials. 2016;77:87–97.

    Article  CAS  PubMed  Google Scholar 

  186. Islam MA, Alam SS, Kundu S, Ahmed S, Sultana S, Patar A, et al. Mesenchymal stem cell therapy in multiple sclerosis: A systematic review and Meta-Analysis. J Clin Med. 2023;12:6311.

    Article  PubMed  PubMed Central  Google Scholar 

  187. Petrou P, Kassis I, Levin N, Paul F, Backner Y, Benoliel T, et al. Beneficial effects of autologous mesenchymal stem cell transplantation in active progressive multiple sclerosis. Brain J Neurol. 2020;143:3574–88.

    Article  Google Scholar 

  188. Uccelli A, Laroni A, Ali R, Battaglia MA, Blinkenberg M, Brundin L, et al. Safety, tolerability, and activity of mesenchymal stem cells versus placebo in multiple sclerosis (MESEMS): a phase 2, randomised, double-blind crossover trial. Lancet Neurol. 2021;20:917–29.

    Article  CAS  PubMed  Google Scholar 

  189. Lu Z, Zhu L, Liu Z, Wu J, Xu Y, Zhang C-J. IV/IT hUC-MSCs infusion in RRMS and NMO: A 10-Year Follow-Up study. Front Neurol. 2020;11:967.

    Article  PubMed  PubMed Central  Google Scholar 

  190. Karussis D, Karageorgiou C, Vaknin-Dembinsky A, Gowda-Kurkalli B, Gomori JM, Kassis I, et al. Safety and immunological effects of mesenchymal stem cell transplantation in patients with multiple sclerosis and amyotrophic lateral sclerosis. Arch Neurol. 2010;67:1187–94.

    Article  PubMed  PubMed Central  Google Scholar 

  191. Petrou P, Kassis I, Ginzberg A, Halimi M, Yaghmour N, Abramsky O, et al. Long-Term clinical and immunological effects of repeated mesenchymal stem cell injections in patients with progressive forms of multiple sclerosis. Front Neurol. 2021;12:639315.

    Article  PubMed  PubMed Central  Google Scholar 

  192. Kvistad CE, Kråkenes T, Gjerde C, Mustafa K, Rekand T, Bø L. Safety and clinical efficacy of mesenchymal stem cell treatment in traumatic spinal cord injury, multiple sclerosis and ischemic Stroke - A systematic review and Meta-Analysis. Front Neurol. 2022;13:891514.

    Article  PubMed  PubMed Central  Google Scholar 

  193. Gugliandolo A, Bramanti P, Mazzon E. Mesenchymal stem cells in multiple sclerosis: recent evidence from Pre-Clinical to clinical studies. Int J Mol Sci. 2020;21:8662.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This work was supported by grants to M. Z. Allouh from the United Arab Emirates University (CMHS grant # G00004697, fund # 12M176; and UPAR grant # G00004977, fund # 12M219) and by NIH grant # R16 NS129748-01 to G. Rasul Chaudhry at the OU-WB ISCRM, Oakland University, Rochester, USA.

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization: M.Z.A., and G.R.C. Writing – Original Draft: All authors. Writing – Review & Editing: M.Z.A., S.F.A.R., A.A., Y.J. and G.R.C. Funding Acquisition: M.Z.A., and G.R.C. Figures: M.Z.A.

Corresponding authors

Correspondence to Mohammed Z. Allouh or G. Rasul Chaudhry.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Artificial intelligence

The authors declare that they have not used artificial intelligence in this review.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Allouh, M.Z., Rizvi, S.F.A., Alamri, A. et al. Mesenchymal stromal/stem cells from perinatal sources: biological facts, molecular biomarkers, and therapeutic promises. Stem Cell Res Ther 16, 127 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13287-025-04254-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13287-025-04254-0

Keywords