Skip to main content

Unveiling heterogeneity in MSCs: exploring marker-based strategies for defining MSC subpopulations

Abstract

Mesenchymal stem/stromal cells (MSCs) represent a heterogeneous cell population distributed throughout various tissues, demonstrating remarkable adaptability to microenvironmental cues and holding immense promise for disease treatment. However, the inherent diversity within MSCs often leads to variability in therapeutic outcomes, posing challenges for clinical applications. To address this heterogeneity, purification of MSC subpopulations through marker-based isolation has emerged as a promising approach to ensure consistent therapeutic efficacy. In this review, we discussed the reported markers of MSCs, encompassing those developed through candidate marker strategies and high-throughput approaches, with the aim of explore viable strategies for addressing the heterogeneity of MSCs and illuminate prospective research directions in this field.

Introduction

MSCs (mesenchymal stem/stromal cells) are heterogenous cell populations, residing in various tissues (such as bone marrow, umbilical cord, teeth, adipose, and so on). Differing from other types of adult stem cells or terminal differentiated cells, the main function of MSCs is sensing and responding to micro-environmental disturbances. Due to their innate characteristics and functions, they have multiple ways to respond to micro-environmental changes, such as extracellular matrix modification, recruiting other cells (the immune cells, for example), secreting small factors with various functions (immune modulation and regeneration, for example) [1]. It is well-known that many diseases are resulting from micro-environment dysfunctions. Therefore, the MSCs have been intensively and extensively applied in treating different kinds of diseases. Both pre-clinical and clinical investigations have shown that the MSCs hold great promise in developing one new therapeutic approach for treating many kinds of diseases [1,2,3,4,5].

Soon after the first demonstration of MSCs, its therapeutic applications have been investigated for decades. Unfortunately, in contrast to the rapid growth of clinical trials, few of them eventually have been developed as applicable therapeutic products. In addition to other factors inducing the therapeutic inconsistency of MSCs, cell heterogeneity is one tough challenge in their way to achieve the expected clinical outcomes [1, 2, 4, 6, 7].

The heterogeneity of MSCs is reflected in different levels, such as the molecular levels (transcriptomics, proteomics, secretomics, and epigenomics), and the function levels (tri-lineage differentiation potentials, immunomodulatory capabilities, and regenerative activities) [2, 7, 8]. The heterogeneity of MSCs could be induced by various factors including the donor conditions (age, gender, health condition, genetic background, and so on), tissue origin, and the strategies to isolate and expand the MSCs (digestion enzyme, matrix protein, cell culture medium, passage number, and so on) [2, 6, 7, 9,10,11,12] (Fig. 1). The causes of MSC heterogeneity have been extensively described in the preceding reviews, and we will not delve into them further. Pluripotent stem cell derived MSC, which can avoid the heterogeneity induced by the aforementioned factors, is also an important category of MSCs [13, 14]. However, we will focus exclusively on MSCs derived from somatic cells under natural conditions in this review.

Fig. 1
figure 1

Illustration of factors inducing MSC heterogeneity and potential solutions. The MSC heterogeneity results from various factors, including donor conditions (age, gender, health conditions), tissue origin (bone, fat, placenta/umbilical cord, teeth), and the methods employed for isolating (plastic adherence, MACS, FACS) and expanding MSCs (2D, 3D bioreactor, 3D matrix). To address MSC heterogeneity and enhance their therapeutic stability, three primary strategies are currently employed. These strategies encompass standardizing the MSC production procedures and purifying MSC subpopulations by markers. MSC mesenchymal stem/stromal cell, MACS magnetic-activated cell sorting, FACS fluorescence-activated cell sorting, 2D 2 dimensional, 3D 3 dimensional

Among different strategies to reduce the heterogeneity and improve the therapeutic consistency of MSCs, purifying the homogenous MSC subpopulations is suggested to yield more consistent clinical outcomes [6]. MSC subpopulations refer to distinct groups or subsets within the broader MSC population that are identified based on specific characteristics or markers. These characteristics can include surface protein expressions, functional properties, gene expression profiles, or responsiveness to different environmental cues. According to the minimal criteria for defining MSCs, stated by the International Society for Cellular Therapy in 2006 [15], 55 MSC markers have been identified so far from different tissues and species (Fig. 2, Table 1).

Fig. 2
figure 2

Timeline of MSC marker identification

Table 1 Basic information for MSC markers

Function enrichment by GO (Gene Ontology) analysis indicates that these MSC markers mainly regulate the process of leukocyte migration, wound healing, cell chemotaxis, and so on (Fig. 3A). Although some markers are involved in multiple functions, some of them are also cross-interacted in a network way (Fig. 3B). KEGG (Kyoto Encyclopedia of Genes and Genomes) also indicates that these MSC markers are mainly involved in the signal pathways in PI3K-AKT, adhesion, and so on (Fig. 4A). Similar to the GO analysis, some markers regulated multiple pathways (Fig. 4B) and they are cross-interacted (Fig. 4C). Most of these MSC markers are localized on the cell membrane, which is suitable for cell purification with FACS (fluorescence-activated cell sorting) and MACS (magnetic-activated cell sorting), while some of them are also intracellularly or extracellularly localized (Table 2). Normally, MSCs enriched with specific makers have functional advantages (Table 2). However, in some cases, these enriched MSCs also have some disadvantages (Table 2).

Fig. 3
figure 3

GO analysis of MSC markers. The bioinformatic analysis of GO enrichment of MSC markers was performed with Dotplotting (A), Cnetplotting (B). GO, gene ontology. MSC mesenchymal stem/stromal cell

Fig. 4
figure 4

KEGG analysis of MSC markers. The bioinformatic analysis of KEGG enrichment of MSC markers was performed with Dotplotting (A), Cnetplotting (B), and Emapplotting (C). Bioinformatic analysis was conducted with package ‘enrichplot’ in R. KEGG, kyoto encyclopedia of genes and genomes. MSC mesenchymal stem/stromal cell

Table 2 Pros and cons of MSC markers

There are various strategies available for biomarker discovery, and among them, two classic approaches stand out: the candidate biomarker strategy and the high-throughput screening strategy. The candidate biomarker strategy is based on existing biological knowledge, where one or more molecules or features possibly related to a specific disease or biological process are selected as candidate biomarkers. These candidates are then experimentally validated for their expression levels or variations under different conditions. This strategy relies heavily on a profound comprehensive understanding of medical domains and relevant biological processes [16]. In contrast, the high-throughput screening strategy employs techniques like genomics, transcriptomics, proteomics, lipidomics, and metabolomics to simultaneously analyze a large number of molecules and features. Through these techniques, it becomes feasible to detect thousands of molecules, facilitating the comparison of differences between heterogenous cell populations. Notably, this approach allows for the identification of biomarkers associated with specific functions without being reliant on prior knowledge [16,17,18,19,20]. It's worth noting that these strategies can be combined to enhance the comprehensive development of potential biomarkers. This integrated approach harnesses the biological knowledge of the candidate biomarker strategy while utilizing the technical capabilities of the high-throughput strategy to discover biomarkers associated with distinct functional subgroups in a more comprehensive and precise manner [16, 21,22,23,24]. In the realm of MSCs, with the use of prior knowledge and high-throughput technologies such as single-cell RNA sequencing (scRNA-seq), specific markers related to different functional subsets of mesenchymal stem cells can be more comprehensively and accurately mined [17,18,19,20,21,22,23,24].

Therefore, in the current review, we would discuss the MSC markers that have been identified so far. Furthermore, based on the identification approaches, these markers have been categorized into two groups: the 1st generation of MSC markers, which has been identified by the candidate biomarker strategy; and the 2nd generation of MSC makers, which has been identified by high-throughput screening approaches (Table 2).

Techniques of identifying mesenchymal stem cell subpopulations

In most studies reviewed in this paper, Flow cytometry and fluorescence-activated cell sorting (FACS) is predominantly utilized for sorting MSC subpopulations. FACS are the primary methods for identifying MSC subpopulations, celebrated for their precision and versatility in scientific research. These technologies use fluorescently labeled antibodies to target specific surface markers, allowing researchers to conduct multiparameter analyses [25, 26]. This facilitates simultaneous assessment of various markers and functional properties within MSC populations, aiding in the identification and isolation of distinct subpopulations based on differential expression of markers. Such detailed analysis provides crucial insights into MSC heterogeneity.

Another vital technique, immunomagnetic cell sorting, utilizes magnetic beads tagged with antibodies targeting specific surface markers for selective isolation of MSC subpopulations [27, 28]. This method ensures high specificity and efficiency, essential for distinguishing and harvesting functionally diverse MSC subsets.

Additionally, functional assays are integral for understanding the biological characteristics of MSC subpopulations. Immunomodulatory assays, for instance, involve co-culture setups with immune cells to evaluate MSCs' effects on immune cell proliferation, activation, and cytokine production [29,30,31,32,33]. These studies highlight the potential therapeutic uses of distinct MSC subsets in treating immune-related conditions. Differentiation assays, including those for osteogenic, adipogenic, and chondrogenic pathways, further elucidate the multilineage potential of MSC subpopulations, critical for identifying suitable cell sources for tissue engineering and regenerative medicine.

Gene expression profiling, through techniques such as RNA sequencing, provides deep insights into the transcriptomic landscapes that define specific functional states or lineage commitments within MSC populations [17,18,19, 23, 34]. These analyses help pinpoint molecular signatures characteristic of unique MSC subsets, enhancing our understanding of their heterogeneity.

Together, these techniques not only facilitate a comprehensive analysis of MSC heterogeneity but also specialize in pinpointing distinct MSC subpopulations. By employing these advanced methodologies, researchers can effectively characterize the diverse functional capacities and biological properties inherent to each subpopulation, significantly enhancing the precision of mesenchymal stem cell-based therapeutic strategies and the development of personalized regenerative medicine.

1st generation of MSC markers

Immune suppression related markers

Although the MSCs have been widely investigated in the animal models of different diseases, the only approved clinical product of MSCs is for GVHD (Graft Versus Host Disease) treatment in clinics [35, 36], because of their immune suppression capabilities. The immune modulation activity is one of those important contributors to the therapeutic effects of MSCs [1].

Extracellular secreted modulators

It has been demonstrated that TNFAIP6 (Tumor Necrosis Factor Alpha-Induced Protein 6) is a potential cell marker for mouse MSCs, irrespective of tissue origin and laboratory origin, with higher immune suppression activities and improved therapeutic effects [12]. However, the membrane expression level of TNFAIP6 is significantly lower than its cytoplasm level [12]. Indeed, TNFAIP6, also known as TSG6, is a small secreted protein with extracellular matrix remodeling and immunomodulation functions [37]. On the other hand, the importance of these secreted modulators, such as the TNFAIP6 having been characterized as one efficacy predictor of MSCs in treating inflammation in vivo [38], makes it necessary to develop novel strategies to purifying these MSC subpopulations for improving their therapeutic effects.

Extracellular ATP clearance

Dying or stressed cells could release ATP (Adenosine 5'-triphosphate) to the extracellular spaces and induce the pro-inflammatory cascade [39, 40]. The immune regulatory cells, such as Treg and MSCs, could express genes, such as ENTPD1 (Ecto-Nucleoside Triphosphate Diphosphohydrolase 1, also known as CD39) and NT5E (Ecto-5′-AMP-nucleotidase, also known as CD73), responsible for clearing these extracellular ATP [39, 41]. CD39 could hydrolyze the extracellular ATP, into ADP and then AMP; while CD73 converts AMP into adenosine [39, 40]. The extracellular adenosine has strong immune suppression activities via binding to the corresponding P1 receptors (including A1R, A2AR, A2BR, and A3R), and activating the downstream pathways (such as PKA, NF-κB, CREB, AKT, PI3K, ERK, JNK, and p38) [42]. Furthermore, the extracellular adenosine also regulates other cell functions, such as cell proliferation, adhesion, migration, invasion, tight junction formation, and vascular remodeling [39, 40, 42, 43].

The expression levels of CD39, CD73, and adenosine receptors could be induced by tissue damage, remodeling, and also the conditions of hypoxia and inflammation [41,42,43]. It has been demonstrated that MSCs express both CD73 and CD39 and could convert ATP into adenosine, resulting in suppressing T cell proliferation [44,45,46,47], and the activation of B cells [48]. The expression levels of CD73 modulate the proliferation and differentiation capabilities of MSCs [49, 50]. Its expression level decreases during the differentiation process [51].

The purified CD73+ MSCs have higher levels of colony-forming capabilities [52], even higher than the ENG+ and THY1+ MSCs [51]. In addition, CD73+ MSCs have much higher tri-differentiation abilities (adipocytes, osteoblasts, and chondrocytes) and higher immune suppression activities [52, 53]. Through EGFP reporter analysis in mice, CD73 could identify the MSCs in different organs in vivo [54, 55]. Furthermore, CD73+ MSCs are much more smaller with spindle and rod-like shapes, while CD73 MSCs are more polygonal larger cells [33]. CD73+ MSCs secrete higher levels of regeneration cytokines, such as VEGF, SDF-1α, and HGF than CD73 MSCs, and show improved therapeutic effects on the rat model of myocardial infarction [33]. Furthermore, CD73+CD39+ MSCs have great potential in bone regeneration, including better efficiency in chondrogenic and osteogenic differentiation [56], preventing osteoclastogenesis [57], and promoting bone formation via the Wnt/β-catenin pathway[58].

Other immune regulators

CD200 is an immune suppressor and promotes peripheral immune tolerance [59, 60]. Its immune suppression function works through binding to its receptor CD200R, which then activates multiple pathways, such as MAPK-ERK, p38 MAPK, and JNK, via Dok and p120-RasGAP [61], resulting in upregulating the downstream effectors including IDO (indoleamine-2,3-dioxygenase), TGF-β, and IL-10 [59]. A higher expression level of CD200 in MSCs correlates with enhanced immune suppression activities in vitro and in vivo [62]. CD200 expressed on MSCs recognizes and binds to its receptor CD200R, which is expressed on myeloid progenitors, resulting in myeloid differentiation inhibition and immune suppression [63]. CD200+ MSCs have much higher levels of colony-forming activity [51]. However, it has been demonstrated that the expression of CD200 is undetectable in MSCs derived from umbilical cord blood [64], or very low in MSCs derived from adipose [65]. In contrast, MSCs derived from the umbilical cord express higher levels of CD200 [65]. Interestingly, the pro-inflammatory cytokine IFN-γ upregulates the expression of CD200 in MSCs derived from bone marrow but not adipose or umbilical cord [65].

BST2 (bone marrow stromal cell antigen 2), also known as CD317, is a type of transmembrane glycoprotein involved in virus reproduction suppression and immune regulation [66]. Using the hTERT immortalized human bone marrow MSC colonies, it has been demonstrated that the MSCs from the CD317+ colony have increased cell areas and up-regulated mRNA levels of immunosuppressive genes than the CD317 MSCs in vitro [67]. Furthermore, CD317+ bone marrow-derived MSCs have better regeneration capabilities than the CD317 MSCs [68]. However, fresh CD317 MSCs isolated from human bone marrow have better immune suppression activities but not CD317+ MSCs [68]. However, our unpublished data show that CD317+ MSCs isolated from the human umbilical cord and expanded with chemically defined media have better immune suppression capabilities (unpublished data).

CD274, also known as PD-L1 (programmed death ligand 1), is a type I transmembrane protein and is widely expressed on multiple types of cells, such as lymphocytes [69]. Its expression can be induced by pro-inflammatory cytokines, such as interferon-γ (IFN-γ), TNF-α, and IL-17 [69, 70]. And it has strong immune suppression activities through binding to its receptor PD-1 [71]. It has been demonstrated that PD-L1 is expressed in MSCs [70, 72, 73]. PD-L1+ MSCs have enhanced immune suppression activities and improved therapeutic effects on the collagen-induced mouse model of arthritis [74].

MX1, for ‘myxovirus resistance’, is the gene responsible for virus immunity and an important component of interferon pathway [75]. It has been demonstrated that Mx1+ MSCs are clonogenic at the single-cell level and have tri-differentiation abilities [32]. Although its antivirus mechanism remains unsolved, the Mx1+ MSCs might also have immune regulatory functions.

Cell adhesion related markers

In addition to the important role of MSCs in modulating immune responses [1], another critical function is regulating cell adhesion, including both the cell adhesion and migration of MSCs, as well as the recruitment and adhesion of other types of cells, such as lymphocytes.

Mediating cell migration

CD44 is an important adhesion molecule involved in recruiting immune cells or stem cells into the inflammatory or injured tissues, via interacting with hyaluronic acid (HA), which is expressed in the injured/inflammatory sites [76, 77].Their interactions induce conformational changes of CD44, recruit adaptor proteins, and lead to cytoskeletal rearrangement, resulting in the activation of various signaling pathways that involve cell growth, adhesion, and migration [76, 77]. In addition, CD44 also functions as a co-receptor to regulate the activities of other receptors, such as VEGFR, EGFR, FGFR and PDGFR [78]. CD44 is widely expressed in multiple types of cells, including MSCs, and it also contributes to MSC recruitment [79, 80]. Its expression level is further induced by PDGF [79]. The migration and adhesion of MSCs depend on CD44-HA (hyaluronic acid) interaction [79, 80]. Therefore, CD44 is a potentially important cell surface marker for MSC purification [81]. However, later investigations indicate that freshly isolated mouse/human MSCs derived from bone marrow express very low levels of CD44 [82, 83]. MSCs show enrichment in the CD44 fractions, as evidenced by their marker expression, colony-forming capacity, and in vitro differentiation abilities [82, 83]. Interestingly, CD44 is gradually up-regulated during cell expansion, even for the CD44 fractions of MSCs [82, 83]. Thus, the CD44 expression levels after in vitro expansion, may not reflect their original cell identity [82]. The CD44+ MSCs have enhanced colony-forming capacity and differentiation abilities [84].

MCAM (melanoma cell adhesion molecule), also known as CD146, is involved in cell-ECM (extracellular matrix) interactions [85, 86]. Upregulation of CD146 could switch cell–cell adhesion to cell-ECM adhesion by interacting with its ligands in the ECM, preparing cells for migration and invasion by secreting related cytokines and proteins [85, 86]. CD146 is expressed in many cell types, especially in those cells constituting blood vessels, such as endothelial cells [86] and MSCs [87,88,89,90,91]. And it has been proposed that CD146 is an MSC marker of multipotency [90,91,92,93]. CD146+ MSCs have a much stronger chemotactic attraction [94,95,96,97], and enhanced immune suppression activities in vitro and in vivo [27, 97,98,99]. Higher levels of CD146 expression correlate with a faster proliferation rate, enhanced multilineage differentiation potentials, stronger stemness characteristics, and less senescent phenotypes [98,99,100]. However, Tormin et al. have demonstrated that the colony-forming cells are exclusively enriched in the CD271+ population of MSCs in human bone marrow, regardless of the expression level of CD146 [101]. Within the CD271+ MSCs, both CD146+ and CD146 share similar genotypes and phenotypes [101]. Furthermore, other studies have also demonstrated that CD146+ and CD146 share similar levels of MSC marker expression, colony-forming, proliferation and differentiation capabilities [94, 96, 102, 103]. And the CD146 MSCs even proliferate significantly faster than the CD146+ population [103]. Higher expression of CD146 also indicates more prone to differentiate into vascular smooth muscle cells [103]. In MSCs derived from human dental cysts, CD146Low MSCs have higher levels of cell proliferation, colony-formation, and osteogenesis [102].

SDC2 (Syndecan-2), also known as CD362, is a type of transmembrane heparan sulfate proteoglycan, involved in modulating cell adhesion, proliferation, migration, and apoptosis through its interactions with the extracellular matrix and various proteins, such as proteases and cytokines. These interactions induce downstream pathway activations through intracellular protein partners [104]. CD362 is mainly expressed in MSCs [104]. CD362+ MSCs have enhanced colony forming, immune suppression and regeneration activities [105,106,107]. Furthermore, both Phase 1 and Phase 2 clinical studies show that CD362+ MSCs are safe, feasible, and effective in treating COVID-19 infections [108].

Mediating lymphocyte adhesion

VCAM1 (vascular cell adhesion molecule 1), also known as CD106, mediates cell–cell adhesion and plays an important role in mediating the rolling, adhesion, and migration of circulating lymphocytes on the endothelium under inflammatory conditions [109,110,111]. The CD106 is induced by pro-inflammatory cytokines in MSCs [112], and is involved in the immune suppression function of MSCs [113]. CD106+ MSCs derived from placenta and umbilical cord have stronger abilities to suppress immune responses [112, 114] and better pro-angiogenic activity, with enhanced promoting endothelial cell proliferation and migration [28, 115]. Furthermore, CD106+ MSCs have enhanced homing capacity [28, 112].

ITGA1 (integrin subunit alpha 1) is identified in the very late stage of activated T cells. ITGA1 is the major component of the ECM by binding to collagens (mainly collagen I and IV) and laminin, supporting the migration and activation of leukocytes, such as T cells, NK cells, NKT cells, and monocytes, especially the long-term activated or resident T cells [116, 117]. The ITGA1 has been proposed as an MSC marker for human bone marrow [118,119,120]. The ITGA1+ MSCs have stronger colony-forming activity [118, 119].

CD9, also known as MRP1 (motility related protein-1), is widely expressed in many cell types, including MSCs and lymphocytes, and is involved in regulating cell migration and invasion through integrin receptors [121, 122]. It has been demonstrated that CD9 is involved in the recognition and binding between MSCs and lymphocytes [123]. CD9+ human MSCs have higher NOS (nitric oxide synthase) expression, proliferation rate, colony formation ratio, and stronger cell adhesion capability, resulting in better engraftment and improved therapeutic effects in the mouse model of hindlimb ischemia [124, 125].

Other adhesion molecules

THY1, also known as CD90, is a small membrane protein located in the lipid raft [126]. Although CD90 does not have an intracellular domain, it is involved in cell adhesion, migration, proliferation, and apoptosis through modulating the cell–cell and cell–matrix interactions via binding to its ligands, such as integrins, syndecan, CD90 and CD97 [126, 127]. CD90 has been identified as an important marker for MSCs from different species and tissues [128,129,130,131,132], and could be a potential marker for predicting the immune suppressive function of MSCs [133, 134]. Later studies also indicate that CD90+ MSCs have a faster proliferation rate and better differentiation capabilities [135,136,137]. However, CD90 is also expressed in the fibroblasts, which might induce fibrosis [138].

Other adhesion genes also have been demonstrated as potential MSC makers, such as the SUSD2 [139,140,141], ALCAM [142, 143], NCAM1 [144,145,146,147,148,149], CD51 (also known as ITGAV) [150, 151], and ITGA6 (also known as CD49f) [152] (Tables 1, 2).

Regeneration related markers

Ephrin receptors

The Ephrin receptors (EphA and EphB), which can be recognized by ephrin ligands, play an important role in modulating multiple cellular functions, such as the self-renewal and differentiation of stem cells [153,154,155,156,157,158]. Proteomics studies indicate that EphA2 is expressed in the MSCs from human bone marrow and umbilical cord, and regulates the functions of MSCs [159, 160]. Follow-up studies showed that EphA2 could be a cell surface marker to distinguish MSCs from fibroblasts [161]. Furthermore, EphA7+ MSCs proliferate faster and have higher levels of colony formation and differentiation capabilities [162]. And EphB2+ MSCs have improved intestinal homing abilities and promoted the intestinal stem cell regeneration [31]. It has been demonstrated that Eph/ephrin pathway is also involved in the cell migration of MSCs [163,164,165,166], and is essential for suppressing the proliferation of activated T cells by MSCs [167].

PDGFR

PDGFR (platelet-derived growth factor receptor), including PDGFRA and PDGFRB (also known as CD140α and CD140β, respectively), plays an important role in embryonic development and organogenesis, particularly in regulating the proliferation, migration, and differentiation of MSCs in various organs [168,169,170,171]. Although both CD140α and CD140β have been identified as MSC markers [91, 168, 169], their investigation also indicates that CD140α is the negative selection marker for human MSCs derived from bone marrow, which differs from mouse MSCs [172].

Wnt pathway

FZD9, also known as CD349, is a receptor for Wnt ligands and activates β-catenin signaling pathway, which is involved in embryonic development and stem cell renewal [173, 174]. It has been demonstrated that CD349 is expressed in MSCs from both bone marrow and placenta, and proposed as a feasible marker for MSC isolation [175, 176]. Although both CD349+ and CD349 MSCs show similar levels of MSC marker expression and differentiation abilities, the CD349 MSCs have better neovascularization abilities than the CD349+ MSCs [177].

ROR2 is a tyrosine kinase-like orphan receptor, which can be activated by Wnt5a and regulate the tissue polarity and cell movement through downstream WNT/PCP (planar cell polarity) signaling pathway [178, 179]. It has been demonstrated that ROR2+ MSCs derived from human bone marrow have enhanced chondrogenic differentiation efficiency [30].

Others

ALDH (aldehyde dehydrogenase) belongs to the metabolic enzyme family, which is involved in regulating glycolysis/gluconeogenesis and the detoxification of aldehydes via oxidation [180, 181]. It plays an important role in cell survival, proliferation, differentiation, and has been characterized as a classical stem cell marker [180, 181]. In human adipose tissues, the ALDHHigh MSCs represent a more primitive subpopulation than the ALDHLow MSCs, from the perspectives of cell proliferation and tri-differentiation capabilities [182, 183].

STRO-1 can bind to an uncharacterized cell surface antigen, and identify around 10% of mononuclear cells in the human bone marrow [184]. Purified STRO-1+ cells from human bone marrow have higher levels of colony-forming activity, proliferative rate, multilineage differentiation capabilities, and immune suppression activities by expressing higher levels of immune inhibitory factors (IL-8, LIF, IDO, HLA-G, VCAM1, TGF-β, and IL-10) [185], suggesting that STRO-1 is a potential MSC marker [184, 186]. Later study showed that STRO-1+ MSCs have better homing activities than STRO-1 MSCs in the bone marrow, spleen, muscle, liver and kidney, while STRO-1 MSCs are more prone to be trapped in the lung [187].

STRO-3, which recognizes TNSALP (tissue nonspecific alkaline phosphatase, a cell-surface glycoprotein), also identifies a MSC subpopulation with higher proliferation and differentiation potencies [188, 189]. The STRO-3+ MSCs have been identified in various species and tissues and applied in treating various disease models [189,190,191,192,193,194,195,196].

STRO-4 is a monoclonal antibody recognizing the cell surface expressed chaperone protein, Hsp90β. STRO-4+ MSCs have higher colony-forming activities, proliferation rates, and multiple differentiation abilities [29].

TLX1, also known as Hoxa11, belongs to Hox gene family which is essential for patterning during embryonic development. It has been demonstrated that the Hoxa11-lineage marked (Hoxa11-eGFP) could identify the multi-potent MSCs in the mouse bone marrow [197]. Hoxa11+ MSCs have better colony forming potentials and tri-lineage differentiation abilities [198, 199].

Transcription factor GLI1, the effector of the Hh signaling pathway, which regulates tissue development and homeostasis, has been used to mark MSCs in vivo [24, 200,201,202]. These Gli1+ MSCs are responsible for tissue regeneration after injury [200, 203,204,205,206]. However, the Gli1+ MSCs have the tendency to differentiate into osteochondrogenic lineages [201, 204]. Furthermore, the Gli1+ MSCs also contribute to tissue fibrosis [205, 207, 208].

ISLR, also known as Meflin, is a glycoprotein (cell membrane located or secreted) with anti-fibrosis functions through interacting with BMP7 (bone morphogenetic protein 7) and inhibiting TGF-β pathway and myofibroblast differentiation [209]. It has been demonstrated that Meflin is one MSC marker, and its expression positively correlates with its differentiation efficiency [210, 211].

Sca-1 (stem cell antigen-1) has been characterized as a common marker in multiple types of stem cells, such as hematopoietic stem cells and MSCs [212, 213]. It has been demonstrated that mouse MSCs derived from bone marrow and ear express high levels of Sca-1 [214,215,216]. The expression of Sca-1 is fundamental for maintaining the stem cell state of MSCs [22, 213, 215, 217]. Furthermore, they have higher proliferation rates and better immune suppression abilities [22, 217]. Other common stem cell markers, such as SSEA-4 (stage-specific embryonic antigen-4), KIT, and ABCG2, have also been identified as MSC markers [218,219,220,221,222,223,224,225,226,227].

Neuron related markers

Interestingly, the MSCs express some neural development related genes and some of them have been identified as MSC markers, such as CSPG4 (chondroitin sulfate proteoglycan 4) [228,229,230], GD2 (Disialoganglioside) [231], CD271 [232,233,234,235,236,237,238], and Nestin [239,240,241,242,243]. Whether the expression of neuron related genes indicates the dedifferentiated state of MSCs or potential interactions between MSCs and neurons remains unclear and needs further investigation.

Other markers

Since the first demonstration of MSCs, the ENG (Endoglin), also known as CD105, has been identified as a classical MSC marker [15, 128]. Using CD105 to purify MSCs is feasible and efficient in human bone marrow and adipose [244,245,246,247,248]. Furthermore, CD105+ MSCs have increased osteogenic and chondrogenic differentiation efficiency, and reduced adipogenic differentiation efficiency [248, 249]. However, controversial results also show that a low expression level of CD105 is correlated with increased osteogenic and chondrogenic differentiation [250]. Indeed, as a coreceptor of the TGF-β superfamily, CD105 is involved in regulating osteogenic differentiation [251,252,253].

The SDF1-CXCR4 is the major pathway responsible for cell recruitment and retention [254, 255]. CXCR4 is expressed in human MSCs and contributes to the MSCs homing process [256,257,258,259,260]. For example, in the mouse model of osteogenesis imperfecta, the human MSCs migrate into the bone marrow through the SDF1-CXCR4 pathway and reduce the fracture rate [261]. Furthermore, in the rat model of ischemic brain injuries, rat MSCs migrate into the injured sites of the brain and show therapeutic effects via the SDF1-CXCR4 pathway [262, 263]. Although the expression level of CXCR4 is high in MSCs, few the on the cell surface [256, 261]. However, Honczarenko et al. have demonstrated that the surface expression of CXCR4 is up to around 43% [264], indicating that some factors might induce the cell surface expression of CXCR4, such as culture conditions, stimuli, and passage numbers [265]. Indeed, the expression level of CXCR4 decreases during passaging [264, 266] and aging [267]. The cell membrane localization of CXCR4 is induced by cytokine stimulation (such as SDF-1) [256, 261, 268] or 3D culture conditions [269].

Some other MSC markers have also been demonstrated, such as LepR (Leptin receptor) [270,271,272,273], CD34 [274,275,276], and TNFRSF10D [277]. However, their functions in MSCs remain unclear. Purifying MSC subpopulation with one single maker has many disadvantages (Table 2). Therefore, the combination of multiple markers is a promising strategy to improve the efficiency and efficacy of MSC subpopulation purification. It has been demonstrated that the PODXLhi/ITGA6hi MSCs have better activities of colony formation, differentiation, proliferation, homing activities, regeneration activity, and anti-apoptotic activities [152, 278,279,280,281,282]. The PDGFR+Sca-1+ MSCs could differentiate into both mesenchymal and endothelial at single-cell level with enhanced self-renewal and multipotency abilities [129], and the CD146+PDGFRβ+ MSCs have higher levels of colony-forming activities [91]. Combining PDGFRα and other markers, such as Ly6a, Sca-1, and CD51, would further enrich the MSC subpopulation with enhanced colony-forming and differentiation activities [129, 243, 283, 284]. On the other hand, identifying novel MSC markers with novel high-throughput technologies is also critical for both MSC subpopulation purification and understanding the heterogeneity of MSCs.

2nd generation of MSC maker identification-high-throughput approach

The emergence and development of high-throughput technologies (genomics, transcriptomics, proteomics, lipidomics, metabolomics, and so on) have revolutionized various fields of life sciences [285,286,287]. These high-throughput technologies have not only expedited the pace of research but also transformed our understanding of life itself by providing a comprehensive and intricate view of biological systems. Since the introduction of Illumina's Solexa sequencing technology in 2005, a new era has been heralded by paving the way for high-throughput technologies [288]. This pioneering approach, built upon parallel sequencing principles, enables the simultaneous analysis of millions of DNA fragments, dramatically boosting sequencing efficiency. Notably, this breakthrough laid the foundation for subsequent advancements, with other platforms such as 454 Life Sciences, Ion Torrent, and PacBio also contributing to the progress of high-throughput technologies [285].

The impact of high-throughput technology extends far beyond genomics, reverberating profoundly across various domains of omics research. In the realm of transcriptomics, we can now unravel intricate gene regulatory networks by simultaneously analyzing the expression of thousands of genes. Technologies like RNA-Seq have empowered scientists to assess gene expression patterns across different conditions, tissues, or developmental stages, shedding light on cellular processes and signaling pathways [286]. High-throughput mass spectrometry techniques in proteomics offer a swift and comprehensive understanding of protein–protein interactions, modifications, and functions within cells. These methods allow researchers to identify and quantify proteins in complex samples, revealing insights into cellular processes, biomarker discovery, and disease mechanisms [287]. In lipidomics, mass spectrometry-based methods have enabled the comprehensive analysis of lipid molecules in biological samples, uncovering lipid profiles associated with health and disease [289]. Similarly, metabolomics, utilizing high-throughput mass spectrometry and nuclear magnetic resonance (NMR) techniques, offers insights into the global metabolite composition of cells or organisms, contributing to our understanding of metabolic pathways and disease biomarkers [290]. Epigenomics, focusing on epigenetic modifications like DNA methylation and histone modifications, benefits from high-throughput techniques such as DNA methylation arrays and next-generation sequencing. These tools provide a genome-wide view of epigenetic modifications, aiding in deciphering their roles in gene regulation, development, and disease [291].

Among these high-throughput technologies, single-cell RNA sequencing technology (scRNA-seq) is a significant innovation in the field of MSCs that has sparked widespread interest in recent years [17,18,19,20,21,22,23,24]. By deciphering the gene expression of each individual cell within a cell population, this technique reveals the astonishing complexity of cellular diversity and heterogeneity, bringing about a revolutionary breakthrough in cellular biology research [292, 293]. Distinct from traditional bulk RNA sequencing methods, scRNA-seq can precisely analyze cell function and types, regardless of sample heterogeneity [293,294,295].

Several novel MSC markers have been discovered since the application of scRNA-seq technology in the MSC field, such as the LRRC75A+ MSCs with enhanced VEGF production [23]; the CMKLR1+ MSCs with improved immune suppression capabilities [19]; the F3+ and S100A9+ MSCs with better regenerative activities [17, 18, 34].

Among these novel MSC markers identified by scRNA-seq, the CMKLR1+ subpopulation with enhanced immune suppression capabilities [19] has been investigated in detail. The CMKLR1 (Chemokine-like receptor 1), also known as CCRL2 (chemokine C–C motif receptor-like 2), is the transmembrane receptor for chemoattractant chemerin, involved in recruiting and migrating of lymphocytes and immune suppression via its ligand resolvin E1, an important anti-inflammatory mediator [296]. Furthermore, it has been demonstrated that the CMKLR1+ MSCs have better osteogenic differentiation potential and weaker adipogenic differentiation potentials than the CMKLR1 MSCs [19]. Indeed, the CMKLR1 pathway regulates the differentiation balance between the osteoblastogenic and adipogenic MSCs [297]. However, their data also indicate that inhibiting the CMKLR1 pathway promotes the osteoblastogenic differentiation of MSCs and suppresses the adipogenic differentiation of the mouse MSCs [297]. Whether the controversial data resulting from species differences needs further investigation [298,299,300].

The applications of scRNA-seq not only promote the identification of novel MSC markers, but also uncover new potential functions of MSCs. The MSC marker F3 [17, 18], also known as CD142 or thromboplastin, is a transmembrane glycoprotein and a receptor for coagulation factors, which is involved in platelet activation and coagulation development after tissue injury [301, 302]. The discovery of F3 in MSCs might indicate that MSCs play an important role in blood clot formation at the site of injury.

The extracellular matrix modification function of MSCs is well-known [303, 304]. However, the extracellular matrix microenvironment is a highly complex and dynamic biological component and is critical for the functions of MSCs, including the immune modulation function and stem cell characteristics [3, 305, 306]. Identifying new extracellular matrix-related MSC markers, such as Serpinf1 [22] and HMMR [20], would enhance our understanding of MSC biology in greater depth and breadth..

In addition, scRNA-seq is a powerful strategy for investigating the heterogeneity of MSCs. Purifying a homogenous MSC subpopulation is proposed to have improved therapeutic advantages [17, 19, 23, 307]. However, it has been demonstrated that the Gli1+ MSCs are still heterogenous, as revealed by scRNA-seq [24]. Furthermore, scRNA-seq can also uncover the diversity of functions and interactions among different MSC subpopulations. Two major MSC subpopulations (Lgr5+ and Lgr6+) residing in the mouse lung have completely different functions, uncovered by scRNA-seq analysis [21]. Lgr6+ MSCs support the airway differentiation, while the Lgr5+ MSCs promote alveolar differentiation [21]. In the human umbilical cord, four different MSC subpopulations (proliferative, niche-supporting, metabolism-related, and biofunctional MSCs) have been revealed by scRNA-sequencing [17].

High-throughput techniques for purifying MSC subpopulations

Throughout the developmental trajectory of scRNA-seq, various innovative platforms have emerged, each catering to different research needs based on their unique principles and features. The 10 × Genomics Chromium system is one widely used platform. It employs droplet technology to combine individual cells with specific molecular barcode particles, enabling high-throughput cell capture and transcriptome sequencing [308]. Similarly, Drop-seq utilizes droplet technology to encapsulate cells and molecular barcode beads in droplets, providing a cost-effective option for large-scale cell sequencing [308]. For studies requiring more accurate and comprehensive gene expression information, SMART-seq2 is an ideal choice. Its principle involves introducing specific sample labels after reverse transcription of RNA, allowing individual processing and sequencing of each cell's RNA for deeper insights [309]. For large-scale sample processing, CEL-seq2 proves to be a powerful selection, utilizing cell-specific molecular barcode primers to provide unique identification for each cell [310]. Additionally, the C1 platform, also known as Fluidigm C1, is an advanced single-cell analysis technology platform. It combines microfluidics technology and real-time fluorescence PCR technology, enabling high-throughput capture, processing, and analysis of individual cells. The workflow of the C1 platform includes key steps such as cell capture, lysis, reverse transcription, and amplification, resulting in high-quality single-cell transcriptome data. The C1 platform can be applied to various types of cell analysis, offering crucial support for cellular biology research [311].

Apart from the aforementioned platforms, other unique single-cell sequencing platforms continue to advance the field of cell analysis. For instance, inDrop, a platform similar to Drop-seq, utilizes microfluidic chips for cell capture, boasting high-throughput performance. Its distinctive design involves encapsulating cells and molecular barcode beads together in droplets, enhancing efficiency and accuracy in cell capture and analysis [308]. Moreover, sci-ATAC-seq is another notable platform that not only focuses on single-cell gene expression but also integrates transcriptome and chromatin accessibility information, providing researchers with more comprehensive data [312]. SPLiT-seq, a high-throughput single-cell sequencing technology, simultaneously captures the transcriptomes of thousands of cells. Through specialized fragmentation tags, cellular lysates are split into multiple fragments, each containing a cell-specific molecular barcode. This tag design enables the concurrent amplification of RNA fragments from multiple cells in a single reaction, achieving high-throughput cell capture and sequencing [313].

The exploration of the majority of these markers (S100A9, F3, LRRC75A, SERPINF1, CMKLR1, GL1) in the context of scRNA-seq applications has primarily relied on the 10 × Genomics Chromium system [17,18,19, 22,23,24], while the CD168 identification was conducted with the C1 platform [20] and the interaction between Lgr5+ and Lgr6+ MSCs were carried out by using the SMART-seq2 technology [21]. The reliability of these three platforms for developing novel MSC markers has been successfully validated. However, other single-cell sequencing platforms have yet to be applied in the MSC marker field so far. Their respective unique advantages, however, suggest they still hold immense potential for the development of new and effective MSC markers.

Beyond accelerating the pace of analysis, these technologies facilitate the simultaneous analysis of expansive datasets, laying bare the intricate network of molecular mechanisms and relationships that drive biological systems [285]. With the wide application of high-throughput technologies in biomedicine, we also had a deeper understanding of the complexity of biological systems and sought to go beyond the limitations of single omics. The rise of high-throughput technologies not only accelerated data generation but also paved the way for the emergence of multi-omics. This approach, fueled by the copious data generated, marries different omics layers (genomics, transcriptomics, proteomics, and metabolomics) into a comprehensive narrative of biological intricacies. By merging diverse omics datasets, multi-omics integration offers a more comprehensive biological context, enhancing accuracy and facilitating meaningful interpretation of findings [314, 315]. Notably, multi-omics technologies have already been applicated in MSCs [316,317,318,319]. Gao et al. demonstrated the utility of multi-omics analysis in understanding the immunosuppressive efficacy of MSCs, shedding light on cellular senescence and PD-L1 expression through single-cell transcriptome and proteomic data analysis [319]. Their findings underscore the potential of multi-omics approaches in discovering new effective MSC markers. This indicates that multi-omics is a feasible strategy to find new effective MSC subpopulations.

Enhanced therapeutic efficacy of marker-sorted MSC subpopulations

Above-mentioned MSC subpopulations exhibit enhanced therapeutic efficacy in various disease models, offering tailored treatment approaches for regenerative medicine and immunotherapy. CD73+ MSCs, characterized by heightened regeneration cytokine secretion and colony-forming capabilities, have shown promising results, particularly in myocardial infarction models [33, 52]. Similarly, CD200+, CD317+, and PD-L1+ MSC subpopulations demonstrate superior colony-forming activity and immune modulation, with CD317+ MSCs exhibiting notable immune suppression capabilities [51, 68, 74]. Moreover, functional diversity is evident among MSC subpopulations. For instance, CD146+ MSCs exhibit strong chemotactic attraction and immune suppression, while CD362+ MSCs display enhanced colony formation and immune suppression, validated in COVID-19 clinical trials [27, 94,95,96,97,98,99, 105,106,107,108]. Additionally, CD106+ MSCs from placenta and umbilical cord tissues demonstrate potent immunomodulation and pro-angiogenic activities [28, 112, 114, 115]. Furthermore, ITGA1+ MSCs and STRO-4+ MSCs exhibit robust colony-forming and proliferation rates [29, 118, 119].

In the clinical translation of marker-based sorting, personalized therapeutic interventions are becoming increasingly feasible. However, further clinical studies are needed to validate the efficacy and safety of these approaches. Overall, leveraging the unique properties of marker-sorted MSC subpopulations holds great promise for advancing regenerative medicine and immunotherapy, offering tailored treatments for diverse medical conditions.

Conclusions and perspectives

Although the therapeutic applications of the MSCs have great promises, challenges still need to be overcome [320, 321]. And the heterogeneity of MSCs constitutes one of those important barriers before their clinical application [6, 7]. Through bioinformatic analysis of the RNA-seq data from different labs and tissues, it is shown that the isolation and expansion procedures induce more heterogeneity than the tissue origin [12]. It should be noted that purifying and expanding the MSCs in vitro is a kind of stress similar to tissue damage in vivo, which might affect the molecular pathways and functions of MSCs [322]. Indeed, the expanded MSCs in vitro are very different from their counterpart in vivo [323]. The MSC expansion strategy would select the cell population which could adapt to these stimuli and stresses [322], indicating the necessity of standardizing the MSC processing procedures and developing a full chemical defined medium [6, 10,11,12]. Therefore, selecting the suitable MSC subpopulations with specific markers based on their functions and applications is necessary and mandatory [6, 7].

So far, the quest for identifying markers of MSCs has been incessant. The emergence of advanced high-throughput multi-omics techniques offers a promising avenue for discovering novel markers. In this review, numerous MSC subpopulations identified through marker-based sorting have demonstrated significant therapeutic efficacy in animal models. These subpopulations mainly exhibit enhanced therapeutic effects through their potent immunosuppressive capabilities, which have been validated across various animal models of inflammation [38, 68, 74, 112, 114]. Additionally, some subpopulations possess superior homing and regenerative properties, contributing to tissue repair in the rat model of myocardial infarction and the mouse model of hindlimb ischemia [31, 33, 124, 125]. These findings offer promising directions for future therapeutic applications of MSCs. Furthermore, ongoing advances in understanding and manipulating the properties of MSC subpopulations hold great promise for the development of more targeted and effective therapies in regenerative medicine and immune modulation.

However, upon evaluating the majority of currently developed markers, a trend becomes apparent: many subpopulations that are sorted tend to revert to an unsorted state after multiple generations of in vitro proliferation. For instance, during the isolation of MSCs using markers like MCAM, CD9, CXCR4, and STRO-1, their expression diminishes upon subsequent in vitro expansion and cultivation [100, 124, 264, 266, 324]. This situation might indicate that the sole reliance on biomarkers cannot purify consistent and stable subpopulations of MSCs, and eventually achieve successful applications in clinical medicine.

Under diverse physiological or pathological conditions, MSCs exhibit various forms of plasticity, including alterations in morphology, surface markers, secretion, differentiation, proliferation, migration, and apoptotic potential [325]. This plasticity is intimately linked to the microenvironment surrounding MSCs, where physical, chemical, and biological factors impact MSCs’ functions through distinct mechanisms [325]. These mechanisms might involve critical processes like signaling pathway modulation and cellular reprogramming, ultimately influencing MSCs’ capabilities [325]. Illustrating the immunomodulatory role of MSCs exemplifies this phenomenon. During the acute phase or relapse of inflammation, effector T cells secrete pro-inflammatory cytokines, including IFN-γ, TNF, IL-1, and IL-7. These pro-inflammatory cytokines stimulate MSCs to produce substantial amounts of IDO (indoleamine 2,3-dioxygenase) and chemokines. Chemokines serve to attract activated T cells toward MSCs. The elevated concentration of IDO metabolites stemming from this process directly inhibits T cells, resulting in an overall attenuation of the immune response and promotion of tissue repair [1, 326]. On the contrary, in chronic inflammation or during remission, the concentration of anti-inflammatory cytokines, such as TGF-β, increases while pro-inflammatory cytokines decline. Consequently, the production of IDO by MSCs drops below the immunosuppressive threshold. Despite the continued expression of chemokines albeit at lower levels, recruited T cells are not restricted, thus exacerbating the inflammatory immune response [1, 326].

As previously highlighted, MSCs are characterized by their exceptional plasticity. The exclusive focus on purifying MSC subpopulations could potentially impose certain limitations. A more intricate strategy revolves around carefully shaping the extracellular environment of MSCs through deliberate in vitro cultivation, a process terming ‘MSC education’. The objective of this educational initiative is to tap into the inherent variability present within the cell population, steering it towards a consistent manifestation of the intended functions. Across a spectrum of models, diverse categories of educated MSCs have unveiled a range of distinctive functionalities [327,328,329]. For instance, when BM-MSCs are exposed to WNT5a secreted by gastric cancer cells, a noticeable upregulation of α-SMA expression and an amplified capacity for driving tumorigenesis have been observed [327]. Furthermore, the exosomes released by MSCs primed with neonatal serum have proven capable of expediting the healing of cutaneous wounds by actively stimulating angiogenesis [328]. Notably, MSCs that have undergone a process of education through exposure to chemotherapy have emerged as critical mediators in facilitating communication between MSCs and tumor-initiating cells within specific tumor contexts. This communication is achieved through the selective secretion of cytokines and/or chemokines [329]. In the realm of immune regulation, differently educated MSCs can even exhibit contrasting functionalities. Waterman et al. found that MSCs educated by T-cell signaling (referred to as MSC-I) are primarily geared toward producing pro-inflammatory factors, while MSCs educated by TLR3 signaling (referred to as MSC-II) predominantly express immune-suppressive factors [330]. Similarly, MSCs educated by immune factors such as IFN-γ and TNF-α also demonstrate enhanced immune-suppressive capabilities [1, 331].

To sum up, the development of MSC markers, bolstered by high-throughput techniques, holds substantial potential. Looking at the broader field of MSC research, in addressing the challenge of inconsistent therapeutic efficacy due to MSC heterogeneity, MSC education also presents a viable avenue alongside MSC markers.

Data availability

Not applicable.

References

  1. Jiang W, Xu J. Immune modulation by mesenchymal stem cells. Cell Prolif. 2020;53(1): e12712.

    Article  PubMed  Google Scholar 

  2. Levy O, et al. Shattering barriers toward clinically meaningful MSC therapies. Sci Adv. 2020;6(30):eaba6884.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Soliman H, et al. Multipotent stromal cells: one name, multiple identities. Cell Stem Cell. 2021;28(10):1690–707.

    Article  CAS  PubMed  Google Scholar 

  4. Krampera M, Le Blanc K. Mesenchymal stromal cells: putative microenvironmental modulators become cell therapy. Cell Stem Cell. 2021;28(10):1708–25.

    Article  CAS  PubMed  Google Scholar 

  5. Hoang DM, et al. Stem cell-based therapy for human diseases. Signal Transduct Target Ther. 2022;7(1):272.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Dunn CM, et al. Strategies to address mesenchymal stem/stromal cell heterogeneity in immunomodulatory profiles to improve cell-based therapies. Acta Biomater. 2021;133:114–25.

    Article  CAS  PubMed  Google Scholar 

  7. Costa LA, et al. Functional heterogeneity of mesenchymal stem cells from natural niches to culture conditions: implications for further clinical uses. Cell Mol Life Sci. 2021;78(2):447–67.

    Article  CAS  PubMed  Google Scholar 

  8. Zhang Q, Xu Y, Xu J. Editorial: targeting heterogeneity of mesenchymal stem cells. Front Cell Dev Biol. 2022;10: 894008.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Xu J, et al. Additive therapeutic effects of mesenchymal stem cells and IL-37 for systemic lupus erythematosus. J Am Soc Nephrol. 2020;31(1):54–65.

    Article  CAS  PubMed  Google Scholar 

  10. Xu J, et al. Improved therapeutic consistency and efficacy of mesenchymal stem cells expanded with chemically defined medium for systemic lupus erythematosus. Cell Mol Immunol. 2020;17(10):1104–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Xu J, et al. Chemical-defined medium supporting the expansion of human mesenchymal stem cells. Stem Cell Res Ther. 2020;11(1):125.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Li L, et al. TNFAIP6 defines the MSC subpopulation with enhanced immune suppression activities. Stem Cell Res Ther. 2022;13(1):479.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Luzzani CD, Miriuka SG. Pluripotent stem cells as a robust source of mesenchymal stem cells. Stem Cell Rev Rep. 2017;13(1):68–78.

    Article  CAS  PubMed  Google Scholar 

  14. Dias IX, et al. Potential and limitations of induced pluripotent stem cells-derived mesenchymal stem cells in musculoskeletal disorders treatment. Biomolecules. 2023;13(9):1342.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Dominici M, et al. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy. 2006;8(4):315–7.

    Article  CAS  PubMed  Google Scholar 

  16. Antoranz A, et al. Mechanism-based biomarker discovery. Drug Discov Today. 2017;22(8):1209–15.

    Article  PubMed  Google Scholar 

  17. Chen P, et al. Single-cell and spatial transcriptomics decodes Wharton’s jelly-derived mesenchymal stem cells heterogeneity and a subpopulation with wound repair signatures. Adv Sci (Weinh). 2023;10(4): e2204786.

    Article  PubMed  Google Scholar 

  18. Sun C, et al. Single-cell RNA-seq highlights heterogeneity in human primary Wharton’s jelly mesenchymal stem/stromal cells cultured in vitro. Stem Cell Res Ther. 2020;11(1):149.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Xie Z, et al. Single-cell RNA sequencing analysis of human bone-marrow-derived mesenchymal stem cells and functional subpopulation identification. Exp Mol Med. 2022;54(4):483–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Huang Y, et al. Single cell transcriptomic analysis of human mesenchymal stem cells reveals limited heterogeneity. Cell Death Dis. 2019;10(5):368.

    Article  PubMed  PubMed Central  Google Scholar 

  21. Lee JH, et al. Anatomically and functionally distinct lung mesenchymal populations marked by Lgr5 and Lgr6. Cell. 2017;170(6):1149-1163e12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Mizikova I, et al. Single-cell RNA sequencing-based characterization of resident lung mesenchymal stromal cells in bronchopulmonary dysplasia. Stem Cells. 2022;40(5):479–92.

    Article  PubMed  PubMed Central  Google Scholar 

  23. Miura T, et al. Single-cell RNA-Seq reveals LRRC75A-expressing cell population involved in VEGF secretion of multipotent mesenchymal stromal/stem cells under ischemia. Stem Cells Transl Med. 2023;12(6):379–90.

    Article  PubMed  PubMed Central  Google Scholar 

  24. Chen S, et al. Runx2+ niche cells maintain incisor mesenchymal tissue homeostasis through IGF signaling. Cell Rep. 2020;32(6): 108007.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Adan A, et al. Flow cytometry: basic principles and applications. Crit Rev Biotechnol. 2017;37(2):163–76.

    Article  CAS  PubMed  Google Scholar 

  26. McKinnon KM. Flow cytometry: an overview. Curr Protoc Immunol. 2018;120:5.1.1-5.1.11.

    Article  PubMed  Google Scholar 

  27. Wu CC, et al. CD146+ mesenchymal stem cells display greater therapeutic potential than CD146− cells for treating collagen-induced arthritis in mice. Stem Cell Res Ther. 2016;7:23.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Jia Y, et al. An optimized method for obtaining clinical-grade specific cell subpopulations from human umbilical cord-derived mesenchymal stem cells. Cell Prolif. 2022;55:e13300.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Gronthos S, et al. Heat shock protein-90 beta is expressed at the surface of multipotential mesenchymal precursor cells: generation of a novel monoclonal antibody, STRO-4, with specificity for mesenchymal precursor cells from human and ovine tissues. Stem Cells Dev. 2009;18(9):1253–62.

    Article  CAS  PubMed  Google Scholar 

  30. Dickinson SC, et al. The Wnt5a receptor, receptor tyrosine kinase-like orphan receptor 2, is a predictive cell surface marker of human mesenchymal stem cells with an enhanced capacity for chondrogenic differentiation. Stem Cells. 2017;35(11):2280–91.

    Article  CAS  PubMed  Google Scholar 

  31. Colletti E, et al. EphB2 isolates a human marrow stromal cell subpopulation with enhanced ability to contribute to the resident intestinal cellular pool. FASEB J. 2013;27(6):2111–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Park D, et al. Endogenous bone marrow MSCs are dynamic, fate-restricted participants in bone maintenance and regeneration. Cell Stem Cell. 2012;10(3):259–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Li Q, et al. CD73(+) mesenchymal stem cells ameliorate myocardial infarction by promoting angiogenesis. Front Cell Dev Biol. 2021;9: 637239.

    Article  PubMed  PubMed Central  Google Scholar 

  34. Chen Y, et al. S100A8 and S100A9 in cancer. Biochim Biophys Acta Rev Cancer. 2023;1878(3): 188891.

    Article  CAS  PubMed  Google Scholar 

  35. Kadri N, et al. Current perspectives on mesenchymal stromal cell therapy for graft versus host disease. Cell Mol Immunol. 2023;20(6):613–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Zhao K, et al. Mesenchymal stromal cells plus basiliximab, calcineurin inhibitor as treatment of steroid-resistant acute graft-versus-host disease: a multicenter, randomized, phase 3, open-label trial. J Hematol Oncol. 2022;15(1):22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Day AJ, Milner CM. TSG-6: A multifunctional protein with anti-inflammatory and tissue-protective properties. Matrix Biol. 2019;78–79:60–83.

    Article  PubMed  Google Scholar 

  38. Lee RH, et al. TSG-6 as a biomarker to predict efficacy of human mesenchymal stem/progenitor cells (hMSCs) in modulating sterile inflammation in vivo. Proc Natl Acad Sci USA. 2014;111(47):16766–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Moesta AK, Li XY, Smyth MJ. Targeting CD39 in cancer. Nat Rev Immunol. 2020;20(12):739–55.

    Article  CAS  PubMed  Google Scholar 

  40. Antonioli L, et al. CD39 and CD73 in immunity and inflammation. Trends Mol Med. 2013;19(6):355–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Liu Y, et al. Review immune response of targeting CD39 in cancer. Biomark Res. 2023;11(1):63.

    Article  PubMed  PubMed Central  Google Scholar 

  42. Galgaro BC, et al. The adenosinergic pathway in mesenchymal stem cell fate and functions. Med Res Rev. 2021;41(4):2316–49.

    Article  PubMed  Google Scholar 

  43. Alcedo KP, Bowser JL, Snider NT. The elegant complexity of mammalian ecto-5’-nucleotidase (CD73). Trends Cell Biol. 2021;31(10):829–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Haynesworth SE, Baber MA, Caplan AI. Cell surface antigens on human marrow-derived mesenchymal cells are detected by monoclonal antibodies. Bone. 1992;13(1):69–80.

    Article  CAS  PubMed  Google Scholar 

  45. Barry F, et al. The SH-3 and SH-4 antibodies recognize distinct epitopes on CD73 from human mesenchymal stem cells. Biochem Biophys Res Commun. 2001;289(2):519–24.

    Article  CAS  PubMed  Google Scholar 

  46. Sattler C, et al. Inhibition of T-cell proliferation by murine multipotent mesenchymal stromal cells is mediated by CD39 expression and adenosine generation. Cell Transplant. 2011;20(8):1221–30.

    Article  PubMed  Google Scholar 

  47. Huang F, et al. Human gingiva-derived mesenchymal stem cells inhibit xeno-graft-versus-host disease via CD39-CD73-adenosine and IDO signals. Front Immunol. 2017;8:68.

    Article  PubMed  PubMed Central  Google Scholar 

  48. Dang J, et al. Human gingiva-derived mesenchymal stem cells are therapeutic in lupus nephritis through targeting of CD39(-)CD73 signaling pathway. J Autoimmun. 2020;113: 102491.

    Article  CAS  PubMed  Google Scholar 

  49. Mistry D, Chambers MG, Mason RM. The role of adenosine in chondrocyte death in murine osteoarthritis and in a murine chondrocyte cell line. Osteoarthrit Cartil. 2006;14(5):486–95.

    Article  CAS  Google Scholar 

  50. Evans BA, et al. Human osteoblast precursors produce extracellular adenosine, which modulates their secretion of IL-6 and osteoprotegerin. J Bone Miner Res. 2006;21(2):228–36.

    Article  CAS  PubMed  Google Scholar 

  51. Delorme B, et al. Specific plasma membrane protein phenotype of culture-amplified and native human bone marrow mesenchymal stem cells. Blood. 2008;111(5):2631–5.

    Article  CAS  PubMed  Google Scholar 

  52. Suto EG, et al. Prospectively isolated mesenchymal stem/stromal cells are enriched in the CD73(+) population and exhibit efficacy after transplantation. Sci Rep. 2017;7(1):4838.

    Article  PubMed  PubMed Central  Google Scholar 

  53. Canepa DD, et al. Identification of ALP+/CD73+ defining markers for enhanced osteogenic potential in human adipose-derived mesenchymal stromal cells by mass cytometry. Stem Cell Res Ther. 2021;12(1):7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Breitbach M, et al. In vivo labeling by CD73 marks multipotent stromal cells and highlights endothelial heterogeneity in the bone marrow niche. Cell Stem Cell. 2018;22(2):262-276.e7.

    Article  CAS  PubMed  Google Scholar 

  55. Severe N, et al. Stress-induced changes in bone marrow stromal cell populations revealed through single-cell protein expression mapping. Cell Stem Cell. 2019;25(4):570-583 e7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Gullo F, De Bari C. Prospective purification of a subpopulation of human synovial mesenchymal stem cells with enhanced chondro-osteogenic potency. Rheumatology (Oxford). 2013;52(10):1758–68.

    Article  CAS  PubMed  Google Scholar 

  57. Luo Y, et al. Human gingival tissue-derived MSC suppress osteoclastogenesis and bone erosion via CD39-adenosine signal pathway in autoimmune arthritis. EBioMedicine. 2019;43:620–31.

    Article  PubMed  PubMed Central  Google Scholar 

  58. Wu W, et al. CD39 produced from human GMSCs regulates the balance of osteoclasts and osteoblasts through the Wnt/beta-catenin pathway in osteoporosis. Mol Ther. 2020;28(6):1518–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Kotwica-Mojzych K, Jodłowska-Jędrych B, Mojzych M. CD200:CD200R interactions and their importance in immunoregulation. Int J Mol Sci. 2021;22(4):1602.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Snelgrove RJ, et al. A critical function for CD200 in lung immune homeostasis and the severity of influenza infection. Nat Immunol. 2008;9(9):1074–83.

    Article  CAS  PubMed  Google Scholar 

  61. van der Vlist M, et al. Signaling by the inhibitory receptor CD200R is rewired by type I interferon. Sci Signal. 2021;14(704):eabb324.

    Google Scholar 

  62. Zhu Y, et al. Placental mesenchymal stem cells of fetal and maternal origins demonstrate different therapeutic potentials. Stem Cell Res Ther. 2014;5(2):48.

    Article  PubMed  PubMed Central  Google Scholar 

  63. Amouzegar A, et al. Mesenchymal stem cells modulate differentiation of myeloid progenitor cells during inflammation. Stem Cells. 2017;35(6):1532–41.

    Article  CAS  PubMed  Google Scholar 

  64. Pietila M, et al. CD200 positive human mesenchymal stem cells suppress TNF-alpha secretion from CD200 receptor positive macrophage-like cells. PLoS ONE. 2012;7(2): e31671.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Najar M, et al. Characterization and functionality of the CD200-CD200R system during mesenchymal stromal cell interactions with T-lymphocytes. Immunol Lett. 2012;146(1–2):50–6.

    Article  CAS  PubMed  Google Scholar 

  66. Zhao Y, et al. Multi-functional BST2/tetherin against HIV-1, other viruses and LINE-1. Front Cell Infect Microbiol. 2022;12: 979091.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. James S, et al. Multiparameter analysis of human bone marrow stromal cells identifies distinct immunomodulatory and differentiation-competent subtypes. Stem Cell Reports. 2015;4(6):1004–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Kay AG, et al. CD317-positive immune stromal cells in human “mesenchymal stem cell” populations. Front Immunol. 2022;13: 903796.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Xie F, et al. The role of exosomal PD-L1 in tumor progression and immunotherapy. Mol Cancer. 2019;18(1):146.

    Article  PubMed  PubMed Central  Google Scholar 

  70. Wang S, et al. Interleukin-17 promotes nitric oxide-dependent expression of PD-L1 in mesenchymal stem cells. Cell Biosci. 2020;10:73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Dong H, et al. B7–H1, a third member of the B7 family, co-stimulates T-cell proliferation and interleukin-10 secretion. Nat Med. 1999;5(12):1365–9.

    Article  CAS  PubMed  Google Scholar 

  72. Sheng H, et al. A critical role of IFNgamma in priming MSC-mediated suppression of T cell proliferation through up-regulation of B7–H1. Cell Res. 2008;18(8):846–57.

    Article  CAS  PubMed  Google Scholar 

  73. Augello A, et al. Bone marrow mesenchymal progenitor cells inhibit lymphocyte proliferation by activation of the programmed death 1 pathway. Eur J Immunol. 2005;35(5):1482–90.

    Article  CAS  PubMed  Google Scholar 

  74. Wu W, et al. B7–H1 promotes the functional effect of human gingiva-derived mesenchymal stem cells on collagen-induced arthritis murine model. Mol Ther. 2020;28(11):2417–29.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Haller O, et al. The discovery of the antiviral resistance gene Mx: a story of great ideas, great failures, and some success. Annu Rev Virol. 2018;5(1):33–51.

    Article  CAS  PubMed  Google Scholar 

  76. Chen C, et al. The biology and role of CD44 in cancer progression: therapeutic implications. J Hematol Oncol. 2018;11(1):64.

    Article  PubMed  PubMed Central  Google Scholar 

  77. Weng X, et al. The membrane receptor CD44: novel insights into metabolism. Trends Endocrinol Metab. 2022;33(5):318–32.

    Article  CAS  PubMed  Google Scholar 

  78. Orian-Rousseau V, Sleeman J. CD44 is a multidomain signaling platform that integrates extracellular matrix cues with growth factor and cytokine signals. Adv Cancer Res. 2014;123:231–54.

    Article  PubMed  Google Scholar 

  79. Zhu H, et al. The role of the hyaluronan receptor CD44 in mesenchymal stem cell migration in the extracellular matrix. Stem Cells. 2006;24(4):928–35.

    Article  CAS  PubMed  Google Scholar 

  80. Herrera MB, et al. Exogenous mesenchymal stem cells localize to the kidney by means of CD44 following acute tubular injury. Kidney Int. 2007;72(4):430–41.

    Article  CAS  PubMed  Google Scholar 

  81. Chamberlain G, et al. Concise review: mesenchymal stem cells: their phenotype, differentiation capacity, immunological features, and potential for homing. Stem Cells. 2007;25(11):2739–49.

    Article  CAS  PubMed  Google Scholar 

  82. Qian H, Le Blanc K, Sigvardsson M. Primary mesenchymal stem and progenitor cells from bone marrow lack expression of CD44 protein. J Biol Chem. 2012;287(31):25795–807.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Hall SR, et al. Identification and isolation of small CD44-negative mesenchymal stem/progenitor cells from human bone marrow using elutriation and polychromatic flow cytometry. Stem Cells Transl Med. 2013;2(8):567–78.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Srinivasan A, et al. Substrate stiffness modulates the multipotency of human neural crest derived ectomesenchymal stem cells via CD44 mediated PDGFR signaling. Biomaterials. 2018;167:153–67.

    Article  CAS  PubMed  Google Scholar 

  85. Wang Z, et al. CD146, from a melanoma cell adhesion molecule to a signaling receptor. Signal Transduct Target Ther. 2020;5(1):148.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Leroyer AS, et al. CD146 (Cluster of Differentiation 146). Arterioscler Thromb Vasc Biol. 2019;39(6):1026–33.

    Article  CAS  PubMed  Google Scholar 

  87. Filshie RJ, et al. MUC18, a member of the immunoglobulin superfamily, is expressed on bone marrow fibroblasts and a subset of hematological malignancies. Leukemia. 1998;12(3):414–21.

    Article  CAS  PubMed  Google Scholar 

  88. Shih IM. The role of CD146 (Mel-CAM) in biology and pathology. J Pathol. 1999;189(1):4–11.

    Article  CAS  PubMed  Google Scholar 

  89. Shi S, Gronthos S. Perivascular niche of postnatal mesenchymal stem cells in human bone marrow and dental pulp. J Bone Miner Res. 2003;18(4):696–704.

    Article  PubMed  Google Scholar 

  90. Baksh D, Yao R, Tuan RS. Comparison of proliferative and multilineage differentiation potential of human mesenchymal stem cells derived from umbilical cord and bone marrow. Stem Cells. 2007;25(6):1384–92.

    Article  CAS  PubMed  Google Scholar 

  91. Schwab KE, Gargett CE. Co-expression of two perivascular cell markers isolates mesenchymal stem-like cells from human endometrium. Hum Reprod. 2007;22(11):2903–11.

    Article  CAS  PubMed  Google Scholar 

  92. Russell KC, et al. In vitro high-capacity assay to quantify the clonal heterogeneity in trilineage potential of mesenchymal stem cells reveals a complex hierarchy of lineage commitment. Stem Cells. 2010;28(4):788–98.

    Article  CAS  PubMed  Google Scholar 

  93. Sacchetti B, et al. Self-renewing osteoprogenitors in bone marrow sinusoids can organize a hematopoietic microenvironment. Cell. 2007;131(2):324–36.

    Article  CAS  PubMed  Google Scholar 

  94. Harkness L, et al. CD146/MCAM defines functionality of human bone marrow stromal stem cell populations. Stem Cell Res Ther. 2016;7:4.

    Article  PubMed  PubMed Central  Google Scholar 

  95. Wangler S, et al. CD146/MCAM distinguishes stem cell subpopulations with distinct migration and regenerative potential in degenerative intervertebral discs. Osteoarthrit Cartil. 2019;27(7):1094–105.

    Article  CAS  Google Scholar 

  96. Al Bahrawy M. Comparison of the migration potential through microperforated membranes of CD146+ GMSC population versus heterogeneous GMSC population. Stem Cells Int. 2021;2021:5583421.

    Article  PubMed  PubMed Central  Google Scholar 

  97. Bowles AC, et al. Signature quality attributes of CD146(+) mesenchymal stem/stromal cells correlate with high therapeutic and secretory potency. Stem Cells. 2020;38(8):1034–49.

    Article  CAS  PubMed  Google Scholar 

  98. Ma L, et al. CD146 controls the quality of clinical grade mesenchymal stem cells from human dental pulp. Stem Cell Res Ther. 2021;12(1):488.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Zhang L, et al. Comparison of CD146 +/− mesenchymal stem cells in improving premature ovarian failure. Stem Cell Res Ther. 2022;13(1):267.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Jin HJ, et al. Downregulation of melanoma cell adhesion molecule (MCAM/CD146) accelerates cellular senescence in human umbilical cord blood-derived mesenchymal stem cells. Stem Cells Transl Med. 2016;5(4):427–39.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Tormin A, et al. CD146 expression on primary nonhematopoietic bone marrow stem cells is correlated with in situ localization. Blood. 2011;117(19):5067–77.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Paduano F, et al. CD146 expression influences periapical cyst mesenchymal stem cell properties. Stem Cell Rev Rep. 2016;12(5):592–603.

    Article  CAS  PubMed  Google Scholar 

  103. Espagnolle N, et al. CD146 expression on mesenchymal stem cells is associated with their vascular smooth muscle commitment. J Cell Mol Med. 2014;18(1):104–14.

    Article  CAS  PubMed  Google Scholar 

  104. Tsoyi K, et al. Lung adenocarcinoma syndecan-2 potentiates cell invasiveness. Am J Respir Cell Mol Biol. 2019;60(6):659–66.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Masterson C, et al. Syndecan-2-positive, bone marrow-derived human mesenchymal stromal cells attenuate bacterial-induced acute lung injury and enhance resolution of ventilator-induced lung injury in rats. Anesthesiology. 2018;129(3):502–16.

    Article  CAS  PubMed  Google Scholar 

  106. Horie S, et al. Umbilical cord-derived CD362(+) mesenchymal stromal cells for E. coli pneumonia: impact of dose regimen, passage, cryopreservation, and antibiotic therapy. Stem Cell Res Ther. 2020;11(1):116.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Gonzalez H, et al. Umbilical cord-derived CD362(+) mesenchymal stromal cells attenuate polymicrobial sepsis induced by caecal ligation and puncture. Int J Mol Sci. 2020;21(21):8270.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Gorman E, et al. Repair of acute respiratory distress syndrome by stromal cell administration (REALIST) trial: A phase 1 trial. EClinicalMedicine. 2021;41: 101167.

    Article  PubMed  PubMed Central  Google Scholar 

  109. VanHeyst KA, et al. Ectopic tumor VCAM-1 expression in cancer metastasis and therapy resistance. Cells. 2022;11(23):3922.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Troncoso MF, et al. VCAM-1 as a predictor biomarker in cardiovascular disease. Biochim Biophys Acta Mol Basis Dis. 2021;1867(9): 166170.

    Article  CAS  PubMed  Google Scholar 

  111. Kokovay E, et al. VCAM1 is essential to maintain the structure of the SVZ niche and acts as an environmental sensor to regulate SVZ lineage progression. Cell Stem Cell. 2012;11(2):220–30.

    Article  CAS  PubMed  Google Scholar 

  112. Wei Y, et al. High-efficient generation of VCAM-1(+) mesenchymal stem cells with multidimensional superiorities in signatures and efficacy on aplastic anaemia mice. Cell Prolif. 2020;53(8): e12862.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Ren G, et al. Inflammatory cytokine-induced intercellular adhesion molecule-1 and vascular cell adhesion molecule-1 in mesenchymal stem cells are critical for immunosuppression. J Immunol. 2010;184(5):2321–8.

    Article  CAS  PubMed  Google Scholar 

  114. Yang ZX, et al. CD106 identifies a subpopulation of mesenchymal stem cells with unique immunomodulatory properties. PLoS ONE. 2013;8(3): e59354.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Du W, et al. VCAM-1+ placenta chorionic villi-derived mesenchymal stem cells display potent pro-angiogenic activity. Stem Cell Res Ther. 2016;7:49.

    Article  PubMed  PubMed Central  Google Scholar 

  116. Sun H, et al. Accumulation of tumor-infiltrating CD49a(+) NK cells correlates with poor prognosis for human hepatocellular carcinoma. Cancer Immunol Res. 2019;7(9):1535–46.

    Article  CAS  PubMed  Google Scholar 

  117. Bromley SK, et al. CD49a regulates cutaneous resident memory CD8(+) T cell persistence and response. Cell Rep. 2020;32(9): 108085.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Deschaseaux F, Charbord P. Human marrow stromal precursors are alpha 1 integrin subunit-positive. J Cell Physiol. 2000;184(3):319–25.

    Article  CAS  PubMed  Google Scholar 

  119. Deschaseaux F, et al. Direct selection of human bone marrow mesenchymal stem cells using an anti-CD49a antibody reveals their CD45med, low phenotype. Br J Haematol. 2003;122(3):506–17.

    Article  PubMed  Google Scholar 

  120. Stewart K, et al. STRO-1, HOP-26 (CD63), CD49a and SB-10 (CD166) as markers of primitive human marrow stromal cells and their more differentiated progeny: a comparative investigation in vitro. Cell Tissue Res. 2003;313(3):281–90.

    Article  CAS  PubMed  Google Scholar 

  121. Brosseau C, et al. CD9 tetraspanin: a new pathway for the regulation of inflammation? Front Immunol. 2018;9:2316.

    Article  PubMed  PubMed Central  Google Scholar 

  122. Reyes R, et al. Tetraspanin CD9: a key regulator of cell adhesion in the immune system. Front Immunol. 2018;9:863.

    Article  PubMed  PubMed Central  Google Scholar 

  123. Oritani K, et al. Stromal cell CD9 and the differentiation of hematopoietic stem/progenitor cells. Leuk Lymphoma. 2000;38(1–2):147–52.

    Article  CAS  PubMed  Google Scholar 

  124. Kim YJ, et al. Role of CD9 in proliferation and proangiogenic action of human adipose-derived mesenchymal stem cells. Pflugers Arch. 2007;455(2):283–96.

    Article  CAS  PubMed  Google Scholar 

  125. Gronthos S, et al. Surface protein characterization of human adipose tissue-derived stromal cells. J Cell Physiol. 2001;189(1):54–63.

    Article  CAS  PubMed  Google Scholar 

  126. Sauzay C, et al. CD90/Thy-1, a cancer-associated cell surface signaling molecule. Front Cell Dev Biol. 2019;7:66.

    Article  PubMed  PubMed Central  Google Scholar 

  127. Morris RJ. Thy-1, a pathfinder protein for the post-genomic era. Front Cell Dev Biol. 2018;6:173.

    Article  PubMed  PubMed Central  Google Scholar 

  128. Pittenger MF, et al. Multilineage potential of adult human mesenchymal stem cells. Science. 1999;284(5411):143–7.

    Article  CAS  PubMed  Google Scholar 

  129. Morikawa S, et al. Prospective identification, isolation, and systemic transplantation of multipotent mesenchymal stem cells in murine bone marrow. J Exp Med. 2009;206(11):2483–96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Kaukua N, et al. Glial origin of mesenchymal stem cells in a tooth model system. Nature. 2014;513(7519):551–4.

    Article  CAS  PubMed  Google Scholar 

  131. Michelis KC, et al. CD90 identifies adventitial mesenchymal progenitor cells in adult human medium- and large-sized arteries. Stem Cell Reports. 2018;11(1):242–57.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Picke AK, et al. Thy-1 (CD90) promotes bone formation and protects against obesity. Sci Transl Med. 2018;10(453):eaa06806.

    Article  Google Scholar 

  133. Campioni D, et al. A decreased positivity for CD90 on human mesenchymal stromal cells (MSCs) is associated with a loss of immunosuppressive activity by MSCs. Cytometry B Clin Cytom. 2009;76(3):225–30.

    Article  PubMed  Google Scholar 

  134. Martini H, et al. Aging induces cardiac mesenchymal stromal cell senescence and promotes endothelial cell fate of the CD90 + subset. Aging Cell. 2019;18(5): e13015.

    Article  PubMed  PubMed Central  Google Scholar 

  135. Kim YK, et al. Osteogenic potential of mouse periosteum-derived cells sorted for CD90 in vitro and in vivo. Stem Cells Transl Med. 2016;5(2):227–34.

    Article  CAS  PubMed  Google Scholar 

  136. Moraes DA, et al. A reduction in CD90 (THY-1) expression results in increased differentiation of mesenchymal stromal cells. Stem Cell Res Ther. 2016;7(1):97.

    Article  PubMed  PubMed Central  Google Scholar 

  137. An Z, et al. A quiescent cell population replenishes mesenchymal stem cells to drive accelerated growth in mouse incisors. Nat Commun. 2018;9(1):378.

    Article  PubMed  PubMed Central  Google Scholar 

  138. Schmidt M, et al. Controlling the balance of fibroblast proliferation and differentiation: impact of Thy-1. J Invest Dermatol. 2015;135(7):1893–902.

    Article  CAS  PubMed  Google Scholar 

  139. Masuda H, et al. A novel marker of human endometrial mesenchymal stem-like cells. Cell Transplant. 2012;21(10):2201–14.

    Article  PubMed  Google Scholar 

  140. Sivasubramaniyan K, et al. Prospective isolation of mesenchymal stem cells from human bone marrow using novel antibodies directed against Sushi domain containing 2. Stem Cells Dev. 2013;22(13):1944–54.

    Article  CAS  PubMed  Google Scholar 

  141. Khanmohammadi M, et al. Identification and characterisation of maternal perivascular SUSD2(+) placental mesenchymal stem/stromal cells. Cell Tissue Res. 2021;385(3):803–15.

    Article  CAS  PubMed  Google Scholar 

  142. Arai F, et al. Mesenchymal stem cells in perichondrium express activated leukocyte cell adhesion molecule and participate in bone marrow formation. J Exp Med. 2002;195(12):1549–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Bruder SP, et al. Mesenchymal stem cell surface antigen SB-10 corresponds to activated leukocyte cell adhesion molecule and is involved in osteogenic differentiation. J Bone Miner Res. 1998;13(4):655–63.

    Article  CAS  PubMed  Google Scholar 

  144. Van Acker HH, et al. CD56 in the immune system: more than a marker for cytotoxicity? Front Immunol. 2017;8:892.

    Article  PubMed  PubMed Central  Google Scholar 

  145. Wang X, et al. Neural cell adhesion molecule contributes to hemopoiesis-supporting capacity of stromal cell lines. Stem Cells. 2005;23(9):1389–99.

    Article  CAS  PubMed  Google Scholar 

  146. Wang X, et al. Mouse mesenchymal stem cells can support human hematopoiesis both in vitro and in vivo: the crucial role of neural cell adhesion molecule. Haematologica. 2010;95(6):884–91.

    Article  CAS  PubMed  Google Scholar 

  147. Skog MS, et al. Expression of neural cell adhesion molecule and polysialic acid in human bone marrow-derived mesenchymal stromal cells. Stem Cell Res Ther. 2016;7(1):113.

    Article  PubMed  PubMed Central  Google Scholar 

  148. Battula VL, et al. Isolation of functionally distinct mesenchymal stem cell subsets using antibodies against CD56, CD271, and mesenchymal stem cell antigen-1. Haematologica. 2009;94(2):173–84.

    Article  CAS  PubMed  Google Scholar 

  149. Studle C, et al. Challenges toward the identification of predictive markers for human mesenchymal stromal cells chondrogenic potential. Stem Cells Transl Med. 2019;8(2):194–204.

    Article  PubMed  PubMed Central  Google Scholar 

  150. Xie DM, et al. CD51 distinguishes a subpopulation of bone marrow mesenchymal stem cells with distinct migratory potential: a novel cell-based strategy to treat acute myocardial infarction in mice. Stem Cell Res Ther. 2019;10(1):331.

    Article  PubMed  PubMed Central  Google Scholar 

  151. Xie DM, et al. Cardiac derived CD51-positive mesenchymal stem cells enhance the cardiac repair through SCF-mediated angiogenesis in mice with myocardial infarction. Front Cell Dev Biol. 2021;9: 642533.

    Article  PubMed  PubMed Central  Google Scholar 

  152. Lee RH, et al. The CD34-like protein PODXL and alpha6-integrin (CD49f) identify early progenitor MSCs with increased clonogenicity and migration to infarcted heart in mice. Blood. 2009;113(4):816–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Xiao T, et al. Targeting EphA2 in cancer. J Hematol Oncol. 2020;13(1):114.

    Article  PubMed  PubMed Central  Google Scholar 

  154. Gucciardo E, Sugiyama N, Lehti K. Eph- and ephrin-dependent mechanisms in tumor and stem cell dynamics. Cell Mol Life Sci. 2014;71(19):3685–710.

    Article  CAS  PubMed  Google Scholar 

  155. Liu W, et al. The roles of EphB2 in cancer. Front Cell Dev Biol. 2022;10: 788587.

    Article  PubMed  PubMed Central  Google Scholar 

  156. Arthur A, et al. Eph/ephrinB mediate dental pulp stem cell mobilization and function. J Dent Res. 2009;88(9):829–34.

    Article  CAS  PubMed  Google Scholar 

  157. Himanen JP, Saha N, Nikolov DB. Cell-cell signaling via Eph receptors and ephrins. Curr Opin Cell Biol. 2007;19(5):534–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Pasquale EB. Eph receptor signalling casts a wide net on cell behaviour. Nat Rev Mol Cell Biol. 2005;6(6):462–75.

    Article  CAS  PubMed  Google Scholar 

  159. Holley RJ, et al. Comparative quantification of the surfaceome of human multipotent mesenchymal progenitor cells. Stem Cell Reports. 2015;4(3):473–88.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  160. Alfaro D, Zapata AG. Eph/Ephrin-mediated stimulation of human bone marrow mesenchymal stromal cells correlates with changes in cell adherence and increased cell death. Stem Cell Res Ther. 2018;9(1):172.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Al Madhoun A, et al. Comparative proteomic analysis identifies EphA2 as a specific cell surface marker for Wharton’s jelly-derived mesenchymal stem cells. Int J Mol Sci. 2020;21(17):6437.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Yoshida Y, et al. Capillary-resident EphA7(+) pericytes are multipotent cells with anti-ischemic effects through capillary formation. Stem Cells Transl Med. 2020;9(1):120–30.

    Article  CAS  PubMed  Google Scholar 

  163. Arthur A, et al. EphB/ephrin-B interactions mediate human MSC attachment, migration and osteochondral differentiation. Bone. 2011;48(3):533–42.

    Article  CAS  PubMed  Google Scholar 

  164. Compagni A, et al. Control of skeletal patterning by ephrinB1-EphB interactions. Dev Cell. 2003;5(2):217–30.

    Article  CAS  PubMed  Google Scholar 

  165. Matsuo K, Otaki N. Bone cell interactions through Eph/ephrin: bone modeling, remodeling and associated diseases. Cell Adh Migr. 2012;6(2):148–56.

    Article  PubMed  PubMed Central  Google Scholar 

  166. Mundy GR, Elefteriou F. Boning up on ephrin signaling. Cell. 2006;126(3):441–3.

    Article  CAS  PubMed  Google Scholar 

  167. Nguyen TM, et al. EphB and Ephrin-B interactions mediate human mesenchymal stem cell suppression of activated T-cells. Stem Cells Dev. 2013;22(20):2751–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  168. Murayama E, et al. Alcam-a and Pdgfr-α are essential for the development of sclerotome-derived stromal cells that support hematopoiesis. Nat Commun. 2023;14(1):1171.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  169. Miwa H, Era T. Tracing the destiny of mesenchymal stem cells from embryo to adult bone marrow and white adipose tissue via Pdgfralpha expression. Development. 2018;145(2): dev155879.

    Article  PubMed  Google Scholar 

  170. Chan RW, Schwab KE, Gargett CE. Clonogenicity of human endometrial epithelial and stromal cells. Biol Reprod. 2004;70(6):1738–50.

    Article  CAS  PubMed  Google Scholar 

  171. Schwab KE, Chan RW, Gargett CE. Putative stem cell activity of human endometrial epithelial and stromal cells during the menstrual cycle. Fertil Steril. 2005;84(Suppl 2):1124–30.

    Article  CAS  PubMed  Google Scholar 

  172. Li H, et al. Low/negative expression of PDGFR-alpha identifies the candidate primary mesenchymal stromal cells in adult human bone marrow. Stem Cell Reports. 2014;3(6):965–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  173. Zhang Z, et al. Upregulation of frizzled 9 in astrocytomas. Neuropathol Appl Neurobiol. 2006;32(6):615–24.

    Article  CAS  PubMed  Google Scholar 

  174. Logan CY, Nusse R. The Wnt signaling pathway in development and disease. Annu Rev Cell Dev Biol. 2004;20:781–810.

    Article  CAS  PubMed  Google Scholar 

  175. Battula VL, et al. Human placenta and bone marrow derived MSC cultured in serum-free, b-FGF-containing medium express cell surface frizzled-9 and SSEA-4 and give rise to multilineage differentiation. Differentiation. 2007;75(4):279–91.

    Article  CAS  PubMed  Google Scholar 

  176. Battula VL, et al. Prospective isolation and characterization of mesenchymal stem cells from human placenta using a frizzled-9-specific monoclonal antibody. Differentiation. 2008;76(4):326–36.

    Article  CAS  PubMed  Google Scholar 

  177. Tran TC, et al. Identification of human placenta-derived mesenchymal stem cells involved in re-endothelialization. J Cell Physiol. 2011;226(1):224–35.

    Article  CAS  PubMed  Google Scholar 

  178. DeChiara TM, et al. Ror2, encoding a receptor-like tyrosine kinase, is required for cartilage and growth plate development. Nat Genet. 2000;24(3):271–4.

    Article  CAS  PubMed  Google Scholar 

  179. Sato A, et al. Wnt5a regulates distinct signalling pathways by binding to Frizzled2. EMBO J. 2010;29(1):41–54.

    Article  CAS  PubMed  Google Scholar 

  180. Vasilogiannakopoulou T, Piperi C, Papavassiliou AG. Impact of aldehyde dehydrogenase activity on gliomas. Trends Pharmacol Sci. 2018;39(7):605–9.

    Article  CAS  PubMed  Google Scholar 

  181. Ginestier C, et al. ALDH1 is a marker of normal and malignant human mammary stem cells and a predictor of poor clinical outcome. Cell Stem Cell. 2007;1(5):555–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  182. Hardy WR, et al. Transcriptional networks in single perivascular cells sorted from human adipose tissue reveal a hierarchy of mesenchymal stem cells. Stem Cells. 2017;35(5):1273–89.

    Article  CAS  PubMed  Google Scholar 

  183. Gomez-Salazar MA, et al. Aldehyde dehydrogenase, a marker of normal and malignant stem cells, typifies mesenchymal progenitors in perivascular niches. Stem Cells Transl Med. 2023;12(7):474–84.

    Article  PubMed  PubMed Central  Google Scholar 

  184. Simmons PJ, Torok-Storb B. Identification of stromal cell precursors in human bone marrow by a novel monoclonal antibody, STRO-1. Blood. 1991;78(1):55–62.

    Article  CAS  PubMed  Google Scholar 

  185. Nasef A, et al. Selected Stro-1-enriched bone marrow stromal cells display a major suppressive effect on lymphocyte proliferation. Int J Lab Hematol. 2009;31(1):9–19.

    Article  CAS  PubMed  Google Scholar 

  186. Psaltis PJ, et al. Enrichment for STRO-1 expression enhances the cardiovascular paracrine activity of human bone marrow-derived mesenchymal cell populations. J Cell Physiol. 2010;223(2):530–40.

    Article  CAS  PubMed  Google Scholar 

  187. Bensidhoum M, et al. Homing of in vitro expanded Stro-1− or Stro-1+ human mesenchymal stem cells into the NOD/SCID mouse and their role in supporting human CD34 cell engraftment. Blood. 2004;103(9):3313–9.

    Article  CAS  PubMed  Google Scholar 

  188. Gronthos S, et al. A novel monoclonal antibody (STRO-3) identifies an isoform of tissue nonspecific alkaline phosphatase expressed by multipotent bone marrow stromal stem cells. Stem Cells Dev. 2007;16(6):953–63.

    Article  CAS  PubMed  Google Scholar 

  189. See F, et al. Therapeutic effects of human STRO-3-selected mesenchymal precursor cells and their soluble factors in experimental myocardial ischemia. J Cell Mol Med. 2011;15(10):2117–29.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  190. Hamamoto H, et al. Allogeneic mesenchymal precursor cell therapy to limit remodeling after myocardial infarction: the effect of cell dosage. Ann Thorac Surg. 2009;87(3):794–801.

    Article  PubMed  PubMed Central  Google Scholar 

  191. Psaltis PJ, et al. Reparative effects of allogeneic mesenchymal precursor cells delivered transendocardially in experimental nonischemic cardiomyopathy. JACC Cardiovasc Interv. 2010;3(9):974–83.

    Article  PubMed  Google Scholar 

  192. Ghosh P, et al. Immunoselected STRO-3+ mesenchymal precursor cells and restoration of the extracellular matrix of degenerate intervertebral discs. J Neurosurg Spine. 2012;16(5):479–88.

    Article  PubMed  Google Scholar 

  193. Houtgraaf JH, et al. Intracoronary infusion of allogeneic mesenchymal precursor cells directly after experimental acute myocardial infarction reduces infarct size, abrogates adverse remodeling, and improves cardiac function. Circ Res. 2013;113(2):153–66.

    Article  CAS  PubMed  Google Scholar 

  194. Oehme D, et al. Reconstitution of degenerated ovine lumbar discs by STRO-3-positive allogeneic mesenchymal precursor cells combined with pentosan polysulfate. J Neurosurg Spine. 2016;24(5):715–26.

    Article  PubMed  Google Scholar 

  195. Abdalmula A, et al. Immunoselected STRO-3(+) mesenchymal precursor cells reduce inflammation and improve clinical outcomes in a large animal model of monoarthritis. Stem Cell Res Ther. 2017;8(1):22.

    Article  PubMed  PubMed Central  Google Scholar 

  196. Amirdelfan K, et al. Allogeneic mesenchymal precursor cells treatment for chronic low back pain associated with degenerative disc disease: a prospective randomized, placebo-controlled 36-month study of safety and efficacy. Spine J. 2021;21(2):212–30.

    Article  PubMed  Google Scholar 

  197. Kfoury Y, Scadden DT. Mesenchymal cell contributions to the stem cell niche. Cell Stem Cell. 2015;16(3):239–53.

    Article  CAS  PubMed  Google Scholar 

  198. Rux DR, et al. Regionally restricted Hox function in adult bone marrow multipotent mesenchymal stem/stromal cells. Dev Cell. 2016;39(6):653–66.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  199. Pineault KM, et al. Hox11 expressing regional skeletal stem cells are progenitors for osteoblasts, chondrocytes and adipocytes throughout life. Nat Commun. 2019;10(1):3168.

    Article  PubMed  PubMed Central  Google Scholar 

  200. Zhao H, et al. Secretion of shh by a neurovascular bundle niche supports mesenchymal stem cell homeostasis in the adult mouse incisor. Cell Stem Cell. 2014;14(2):160–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  201. Zhao H, et al. The suture provides a niche for mesenchymal stem cells of craniofacial bones. Nat Cell Biol. 2015;17(4):386–96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  202. Men Y, et al. Gli1+ periodontium stem cells are regulated by osteocytes and occlusal force. Dev Cell. 2020;54(5):639-654e6.

    Article  CAS  PubMed  Google Scholar 

  203. Kramann R, et al. Adventitial MSC-like cells are progenitors of vascular smooth muscle cells and drive vascular calcification in chronic kidney disease. Cell Stem Cell. 2016;19(5):628–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  204. Shi Y, et al. Gli1 identifies osteogenic progenitors for bone formation and fracture repair. Nat Commun. 2017;8(1):2043.

    Article  PubMed  PubMed Central  Google Scholar 

  205. Cassandras M, et al. Gli1(+) mesenchymal stromal cells form a pathological niche to promote airway progenitor metaplasia in the fibrotic lung. Nat Cell Biol. 2020;22(11):1295–306.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  206. Magallanes J, et al. A new mouse model of post-traumatic joint injury allows to identify the contribution of Gli1+ mesenchymal progenitors in arthrofibrosis and acquired heterotopic endochondral ossification. Front Cell Dev Biol. 2022;10: 954028.

    Article  PubMed  PubMed Central  Google Scholar 

  207. Kramann R, et al. Perivascular Gli1+ progenitors are key contributors to injury-induced organ fibrosis. Cell Stem Cell. 2015;16(1):51–66.

    Article  CAS  PubMed  Google Scholar 

  208. Schneider RK, et al. Gli1(+) mesenchymal stromal cells are a key driver of bone marrow fibrosis and an important cellular therapeutic target. Cell Stem Cell. 2017;20(6):785-800e8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  209. Hara A, et al. Roles of the mesenchymal stromal/stem cell marker meflin in cardiac tissue repair and the development of diastolic dysfunction. Circ Res. 2019;125(4):414–30.

    Article  CAS  PubMed  Google Scholar 

  210. Maeda K, et al. Identification of Meflin as a potential marker for mesenchymal stromal cells. Sci Rep. 2016;6:22288.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  211. Takahashi M, et al. Roles of the mesenchymal stromal/stem cell marker Meflin/Islr in cancer fibrosis. Front Cell Dev Biol. 2021;9: 749924.

    Article  PubMed  PubMed Central  Google Scholar 

  212. Holmes C, Stanford WL. Concise review: stem cell antigen-1: expression, function, and enigma. Stem Cells. 2007;25(6):1339–47.

    Article  CAS  PubMed  Google Scholar 

  213. Bonyadi M, et al. Mesenchymal progenitor self-renewal deficiency leads to age-dependent osteoporosis in Sca-1/Ly-6A null mice. Proc Natl Acad Sci USA. 2003;100(10):5840–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  214. Meirellesda LS, Nardi NB. Murine marrow-derived mesenchymal stem cell: isolation, in vitro expansion, and characterization. Br J Haematol. 2003;123(4):702–11.

    Article  Google Scholar 

  215. Staszkiewicz J, et al. IFATS collection: stem cell antigen-1-positive ear mesenchymal stem cells display enhanced adipogenic potential. Stem Cells. 2008;26(10):2666–73.

    Article  CAS  PubMed  Google Scholar 

  216. Hamidouche Z, et al. Bistable epigenetic states explain age-dependent decline in mesenchymal stem cell heterogeneity. Stem Cells. 2017;35(3):694–704.

    Article  CAS  PubMed  Google Scholar 

  217. Rangasamy T, et al. Host defense against Klebsiella pneumoniae pneumonia is augmented by lung-derived mesenchymal stem cells. J Immunol. 2021;207(4):1112–27.

    Article  CAS  PubMed  Google Scholar 

  218. Gang EJ, et al. SSEA-4 identifies mesenchymal stem cells from bone marrow. Blood. 2007;109(4):1743–51.

    Article  CAS  PubMed  Google Scholar 

  219. Rosu-Myles M, et al. The globoseries glycosphingolipid SSEA-4 is a marker of bone marrow-derived clonal multipotent stromal cells in vitro and in vivo. Stem Cells Dev. 2013;22(9):1387–97.

    Article  CAS  PubMed  Google Scholar 

  220. Block TJ, et al. Restoring the quantity and quality of elderly human mesenchymal stem cells for autologous cell-based therapies. Stem Cell Res Ther. 2017;8(1):239.

    Article  PubMed  PubMed Central  Google Scholar 

  221. Kawanabe N, et al. Isolation of multipotent stem cells in human periodontal ligament using stage-specific embryonic antigen-4. Differentiation. 2010;79(2):74–83.

    Article  CAS  PubMed  Google Scholar 

  222. Fukushima H, et al. SSEA-4 is a marker of human deciduous periodontal ligament stem cells. J Dent Res. 2012;91(10):955–60.

    Article  CAS  PubMed  Google Scholar 

  223. Kawanabe N, et al. Stage-specific embryonic antigen-4 identifies human dental pulp stem cells. Exp Cell Res. 2012;318(5):453–63.

    Article  CAS  PubMed  Google Scholar 

  224. Mihaila SM, et al. The osteogenic differentiation of SSEA-4 sub-population of human adipose derived stem cells using silicate nanoplatelets. Biomaterials. 2014;35(33):9087–99.

    Article  CAS  PubMed  Google Scholar 

  225. Varma MJ, et al. Phenotypical and functional characterization of freshly isolated adipose tissue-derived stem cells. Stem Cells Dev. 2007;16(1):91–104.

    Article  PubMed  Google Scholar 

  226. Blazquez-Martinez A, et al. c-Kit identifies a subpopulation of mesenchymal stem cells in adipose tissue with higher telomerase expression and differentiation potential. Differentiation. 2014;87(3–4):147–60.

    Article  CAS  PubMed  Google Scholar 

  227. Jun D, et al. The pathology of bleomycin-induced fibrosis is associated with loss of resident lung mesenchymal stem cells that regulate effector T-cell proliferation. Stem Cells. 2011;29(4):725–35.

    Article  PubMed  Google Scholar 

  228. Ilieva KM, et al. Chondroitin sulfate proteoglycan 4 and its potential as an antibody immunotherapy target across different tumor types. Front Immunol. 2017;8:1911.

    Article  PubMed  Google Scholar 

  229. Kozanoglu I, et al. Human bone marrow mesenchymal cells express NG2: possible increase in discriminative ability of flow cytometry during mesenchymal stromal cell identification. Cytotherapy. 2009;11(5):527–33.

    Article  CAS  PubMed  Google Scholar 

  230. Kunisaki Y, et al. Arteriolar niches maintain haematopoietic stem cell quiescence. Nature. 2013;502(7473):637–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  231. Martinez C, et al. Human bone marrow mesenchymal stromal cells express the neural ganglioside GD2: a novel surface marker for the identification of MSCs. Blood. 2007;109(10):4245–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  232. Cattoretti G, et al. Bone marrow stroma in humans: anti-nerve growth factor receptor antibodies selectively stain reticular cells in vivo and in vitro. Blood. 1993;81(7):1726–38.

    Article  CAS  PubMed  Google Scholar 

  233. Alvarez R, et al. Single CD271 marker isolates mesenchymal stem cells from human dental pulp. Int J Oral Sci. 2015;7(4):205–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  234. Buhring HJ, et al. Novel markers for the prospective isolation of human MSC. Ann N Y Acad Sci. 2007;1106:262–71.

    Article  PubMed  Google Scholar 

  235. Cuthbert R, et al. Single-platform quality control assay to quantify multipotential stromal cells in bone marrow aspirates prior to bulk manufacture or direct therapeutic use. Cytotherapy. 2012;14(4):431–40.

    Article  CAS  PubMed  Google Scholar 

  236. Kuci S, et al. CD271 antigen defines a subset of multipotent stromal cells with immunosuppressive and lymphohematopoietic engraftment-promoting properties. Haematologica. 2010;95(4):651–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  237. Vaculik C, et al. Human dermis harbors distinct mesenchymal stromal cell subsets. J Invest Dermatol. 2012;132(3 Pt 1):563–74.

    Article  CAS  PubMed  Google Scholar 

  238. Smith RJP, et al. The angiogenic potential of CD271+ human adipose tissue-derived mesenchymal stem cells. Stem Cell Res Ther. 2021;12(1):160.

    Article  PubMed  PubMed Central  Google Scholar 

  239. Mendez-Ferrer S, et al. Mesenchymal and haematopoietic stem cells form a unique bone marrow niche. Nature. 2010;466(7308):829–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  240. Jiang MH, et al. Nestin(+) kidney resident mesenchymal stem cells for the treatment of acute kidney ischemia injury. Biomaterials. 2015;50:56–66.

    Article  CAS  PubMed  Google Scholar 

  241. Liao Y, et al. Cardiac Nestin(+) mesenchymal stromal cells enhance healing of ischemic heart through periostin-mediated M2 macrophage polarization. Mol Ther. 2020;28(3):855–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  242. Lu D, et al. Bone-derived Nestin-positive mesenchymal stem cells improve cardiac function via recruiting cardiac endothelial cells after myocardial infarction. Stem Cell Res Ther. 2019;10(1):127.

    Article  PubMed  PubMed Central  Google Scholar 

  243. Pinho S, et al. PDGFRalpha and CD51 mark human nestin+ sphere-forming mesenchymal stem cells capable of hematopoietic progenitor cell expansion. J Exp Med. 2013;210(7):1351–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  244. Robledo MM, et al. Characterization of TGF-beta 1-binding proteins in human bone marrow stromal cells. Br J Haematol. 1996;93(3):507–14.

    Article  CAS  PubMed  Google Scholar 

  245. Majumdar MK, et al. Isolation, characterization, and chondrogenic potential of human bone marrow-derived multipotential stromal cells. J Cell Physiol. 2000;185(1):98–106.

    Article  CAS  PubMed  Google Scholar 

  246. Aslan H, et al. Osteogenic differentiation of noncultured immunoisolated bone marrow-derived CD105+ cells. Stem Cells. 2006;24(7):1728–37.

    Article  PubMed  Google Scholar 

  247. Roura S, et al. Effect of aging on the pluripotential capacity of human CD105+ mesenchymal stem cells. Eur J Heart Fail. 2006;8(6):555–63.

    Article  CAS  PubMed  Google Scholar 

  248. Jiang T, et al. Potent in vitro chondrogenesis of CD105 enriched human adipose-derived stem cells. Biomaterials. 2010;31(13):3564–71.

    Article  CAS  PubMed  Google Scholar 

  249. Chang CB, et al. Chondrogenic potentials of human synovium-derived cells sorted by specific surface markers. Osteoarthrit Cartil. 2013;21(1):190–9.

    Article  CAS  Google Scholar 

  250. Levi B, et al. CD105 protein depletion enhances human adipose-derived stromal cell osteogenesis through reduction of transforming growth factor β1 (TGF-β1) signaling. J Biol Chem. 2011;286(45):39497–509.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  251. Maeda S, et al. Endogenous TGF-beta signaling suppresses maturation of osteoblastic mesenchymal cells. EMBO J. 2004;23(3):552–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  252. Ollauri-Ibáñez C, Ayuso-Íñigo B, Pericacho M. Hot and cold tumors: is endoglin (CD105) a potential target for vessel normalization? Cancers (Basel). 2021;13(7):1552.

    Article  PubMed  Google Scholar 

  253. Jeng KS, et al. The role of endoglin in hepatocellular carcinoma. Int J Mol Sci. 2021;22(6):3208.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  254. Daniel SK, Seo YD, Pillarisetty VG. The CXCL12-CXCR4/CXCR7 axis as a mechanism of immune resistance in gastrointestinal malignancies. Semin Cancer Biol. 2020;65:176–88.

    Article  CAS  PubMed  Google Scholar 

  255. Xu J, et al. Therapeutic effects of CXCR4(+) subpopulation of transgene-free induced cardiosphere-derived cells on experimental myocardial infarction. Cell Prolif. 2021;54(6): e13041.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  256. Wynn RF, et al. A small proportion of mesenchymal stem cells strongly expresses functionally active CXCR4 receptor capable of promoting migration to bone marrow. Blood. 2004;104(9):2643–5.

    Article  CAS  PubMed  Google Scholar 

  257. Shi M, et al. Regulation of CXCR4 expression in human mesenchymal stem cells by cytokine treatment: role in homing efficiency in NOD/SCID mice. Haematologica. 2007;92(7):897–904.

    Article  PubMed  Google Scholar 

  258. Lapidot T, Kollet O. The essential roles of the chemokine SDF-1 and its receptor CXCR4 in human stem cell homing and repopulation of transplanted immune-deficient NOD/SCID and NOD/SCID/B2m(null) mice. Leukemia. 2002;16(10):1992–2003.

    Article  CAS  PubMed  Google Scholar 

  259. Broxmeyer HE, et al. Rapid mobilization of murine and human hematopoietic stem and progenitor cells with AMD3100, a CXCR4 antagonist. J Exp Med. 2005;201(8):1307–18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  260. Kyriakou C, et al. Factors that influence short-term homing of human bone marrow-derived mesenchymal stem cells in a xenogeneic animal model. Haematologica. 2008;93(10):1457–65.

    Article  CAS  PubMed  Google Scholar 

  261. Jones GN, et al. Upregulating CXCR4 in human fetal mesenchymal stem cells enhances engraftment and bone mechanics in a mouse model of osteogenesis imperfecta. Stem Cells Transl Med. 2012;1(1):70–8.

    Article  CAS  PubMed  Google Scholar 

  262. Chen J, et al. Therapeutic benefit of intravenous administration of bone marrow stromal cells after cerebral ischemia in rats. Stroke. 2001;32(4):1005–11.

    Article  CAS  PubMed  Google Scholar 

  263. Ji JF, et al. Interactions of chemokines and chemokine receptors mediate the migration of mesenchymal stem cells to the impaired site in the brain after hypoglossal nerve injury. Stem Cells. 2004;22(3):415–27.

    Article  CAS  PubMed  Google Scholar 

  264. Honczarenko M, et al. Human bone marrow stromal cells express a distinct set of biologically functional chemokine receptors. Stem Cells. 2006;24(4):1030–41.

    Article  CAS  PubMed  Google Scholar 

  265. Deng C, et al. Up-regulation of CXCR4 in rat umbilical mesenchymal stem cells induced by serum from rat with acute liver failure promotes stem cells migration to injured liver tissue. Mol Cell Biochem. 2014;396(1–2):107–16.

    Article  CAS  PubMed  Google Scholar 

  266. Son BR, et al. Migration of bone marrow and cord blood mesenchymal stem cells in vitro is regulated by stromal-derived factor-1-CXCR4 and hepatocyte growth factor-c-met axes and involves matrix metalloproteinases. Stem Cells. 2006;24(5):1254–64.

    Article  CAS  PubMed  Google Scholar 

  267. Singh P, et al. Aging-related reduced expression of CXCR4 on bone marrow mesenchymal stromal cells contributes to hematopoietic stem and progenitor cell defects. Stem Cell Rev Rep. 2020;16(4):684–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  268. Wysoczynski M, et al. Incorporation of CXCR4 into membrane lipid rafts primes homing-related responses of hematopoietic stem/progenitor cells to an SDF-1 gradient. Blood. 2005;105(1):40–8.

    Article  CAS  PubMed  Google Scholar 

  269. Potapova IA, et al. Culturing of human mesenchymal stem cells as three-dimensional aggregates induces functional expression of CXCR4 that regulates adhesion to endothelial cells. J Biol Chem. 2008;283(19):13100–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  270. Zhang KW, et al. LEP and LEPR are possibly a double-edged sword for wound healing. J Cell Physiol. 2023;238(2):355–65.

    Article  CAS  PubMed  Google Scholar 

  271. Voigtmann F, et al. Identification of a novel leptin receptor (LEPR) variant and proof of functional relevance directing treatment decisions in patients with morbid obesity. Metabolism. 2021;116: 154438.

    Article  CAS  PubMed  Google Scholar 

  272. Zhou BO, et al. Leptin-receptor-expressing mesenchymal stromal cells represent the main source of bone formed by adult bone marrow. Cell Stem Cell. 2014;15(2):154–68.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  273. Yue R, et al. Leptin receptor promotes adipogenesis and reduces osteogenesis by regulating mesenchymal stromal cells in adult bone marrow. Cell Stem Cell. 2016;18(6):782–96.

    Article  CAS  PubMed  Google Scholar 

  274. Simmons PJ, Torok-Storb B. CD34 expression by stromal precursors in normal human adult bone marrow. Blood. 1991;78(11):2848–53.

    Article  CAS  PubMed  Google Scholar 

  275. Kaiser S, et al. BM cells giving rise to MSC in culture have a heterogeneous CD34 and CD45 phenotype. Cytotherapy. 2007;9(5):439–50.

    Article  CAS  PubMed  Google Scholar 

  276. Lee PH, et al. Antifibrotic activity of human placental amnion membrane-derived CD34+ mesenchymal stem/progenitor cell transplantation in mice with thioacetamide-induced liver injury. Stem Cells Transl Med. 2016;5(11):1473–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  277. Madsen SD, et al. Decoy TRAIL receptor CD264: a cell surface marker of cellular aging for human bone marrow-derived mesenchymal stem cells. Stem Cell Res Ther. 2017;8(1):201.

    Article  PubMed  PubMed Central  Google Scholar 

  278. Zha K, et al. Evaluation of CD49f as a novel surface marker to identify functional adipose-derived mesenchymal stem cell subset. Cell Prolif. 2021;54(5): e13017.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  279. Yu KR, et al. CD49f enhances multipotency and maintains stemness through the direct regulation of OCT4 and SOX2. Stem Cells. 2012;30(5):876–87.

    Article  CAS  PubMed  Google Scholar 

  280. Nystedt J, et al. Cell surface structures influence lung clearance rate of systemically infused mesenchymal stromal cells. Stem Cells. 2013;31(2):317–26.

    Article  CAS  PubMed  Google Scholar 

  281. Yang Z, et al. CD49f acts as an inflammation sensor to regulate differentiation, adhesion, and migration of human mesenchymal stem cells. Stem Cells. 2015;33(9):2798–810.

    Article  CAS  PubMed  Google Scholar 

  282. Yang Z, et al. CD49f(high) defines a distinct skin mesenchymal stem cell population capable of hair follicle epithelial cell maintenance. J Invest Dermatol. 2020;140(3):544–5559.

    Article  CAS  PubMed  Google Scholar 

  283. Kayaba A, et al. Bone marrow PDGFRα+Sca-1+-enriched mesenchymal stem cells support survival of and antibody production by plasma cells in vitro through IL-6. Int Immunol. 2018;30(6):241–53.

    Article  CAS  PubMed  Google Scholar 

  284. Houlihan DD, et al. Isolation of mouse mesenchymal stem cells on the basis of expression of Sca-1 and PDGFR-alpha. Nat Protoc. 2012;7(12):2103–11.

    Article  CAS  PubMed  Google Scholar 

  285. Metzker ML. Sequencing technologies—the next generation. Nat Rev Genet. 2010;11(1):31–46.

    Article  CAS  PubMed  Google Scholar 

  286. Shen X, et al. Recent advances in high-throughput single-cell transcriptomics and spatial transcriptomics. Lab Chip. 2022;22(24):4774–91.

    Article  CAS  PubMed  Google Scholar 

  287. Zhang Z, et al. High-throughput proteomics. Annu Rev Anal Chem (Palo Alto Calif). 2014;7:427–54.

    Article  CAS  PubMed  Google Scholar 

  288. Bentley DR, et al. Accurate whole human genome sequencing using reversible terminator chemistry. Nature. 2008;456(7218):53–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  289. Baek J, et al. Lipidomic approaches to dissect dysregulated lipid metabolism in kidney disease. Nat Rev Nephrol. 2022;18(1):38–55.

    Article  CAS  PubMed  Google Scholar 

  290. Wishart DS. Metabolomics for investigating physiological and pathophysiological processes. Physiol Rev. 2019;99(4):1819–75.

    Article  CAS  PubMed  Google Scholar 

  291. Wang KC, Chang HY. Epigenomics: technologies and applications. Circ Res. 2018;122(9):1191–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  292. Jovic D, et al. Single-cell RNA sequencing technologies and applications: a brief overview. Clin Transl Med. 2022;12(3): e694.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  293. Papalexi E, Satija R. Single-cell RNA sequencing to explore immune cell heterogeneity. Nat Rev Immunol. 2018;18(1):35–45.

    Article  CAS  PubMed  Google Scholar 

  294. Kolodziejczyk AA, et al. The technology and biology of single-cell RNA sequencing. Mol Cell. 2015;58(4):610–20.

    Article  CAS  PubMed  Google Scholar 

  295. Shapiro E, Biezuner T, Linnarsson S. Single-cell sequencing-based technologies will revolutionize whole-organism science. Nat Rev Genet. 2013;14(9):618–30.

    Article  CAS  PubMed  Google Scholar 

  296. Yoshimura T, Oppenheim JJ. Chemokine-like receptor 1 (CMKLR1) and chemokine (C-C motif) receptor-like 2 (CCRL2); two multifunctional receptors with unusual properties. Exp Cell Res. 2011;317(5):674–84.

    Article  CAS  PubMed  Google Scholar 

  297. Muruganandan S, et al. Chemokine-like receptor 1 is a novel wnt target gene that regulates mesenchymal stem cell differentiation. Stem Cells. 2017;35(3):711–24.

    Article  CAS  PubMed  Google Scholar 

  298. Muruganandan S, et al. Chemerin neutralization blocks hematopoietic stem cell osteoclastogenesis. Stem Cells. 2013;31(10):2172–82.

    Article  CAS  PubMed  Google Scholar 

  299. Muruganandan S, et al. Chemerin, a novel peroxisome proliferator-activated receptor gamma (PPARgamma) target gene that promotes mesenchymal stem cell adipogenesis. J Biol Chem. 2011;286(27):23982–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  300. Muruganandan S, Roman AA, Sinal CJ. Role of chemerin/CMKLR1 signaling in adipogenesis and osteoblastogenesis of bone marrow stem cells. J Bone Miner Res. 2010;25(2):222–34.

    Article  CAS  PubMed  Google Scholar 

  301. Subramaniam S, Kothari H, Bosmann M. Tissue factor in COVID-19-associated coagulopathy. Thromb Res. 2022;220:35–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  302. Unruh D, Horbinski C. Beyond thrombosis: the impact of tissue factor signaling in cancer. J Hematol Oncol. 2020;13(1):93.

    Article  PubMed  PubMed Central  Google Scholar 

  303. Deng M, et al. Modification of PLGA Scaffold by MSC-derived extracellular matrix combats macrophage inflammation to initiate bone regeneration via TGF-β-induced protein. Adv Healthc Mater. 2021;10(12): e2100872.

    Article  PubMed  Google Scholar 

  304. Yang Y, et al. Mesenchymal stem cell-derived extracellular matrix enhances chondrogenic phenotype of and cartilage formation by encapsulated chondrocytes in vitro and in vivo. Acta Biomater. 2018;69:71–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  305. Ding JY, et al. Mesenchymal stem cell-derived extracellular vesicles in skin wound healing: roles, opportunities and challenges. Mil Med Res. 2023;10(1):36.

    CAS  PubMed  PubMed Central  Google Scholar 

  306. Pinho SS, et al. Immune regulatory networks coordinated by glycans and glycan-binding proteins in autoimmunity and infection. Cell Mol Immunol. 2023;20:1101.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  307. Yin JQ, Zhu J, Ankrum JA. Manufacturing of primed mesenchymal stromal cells for therapy. Nat Biomed Eng. 2019;3(2):90–104.

    Article  CAS  PubMed  Google Scholar 

  308. Zhang X, et al. Comparative analysis of droplet-based ultra-high-throughput single-cell RNA-Seq systems. Mol Cell. 2019;73(1):130-142.e5.

    Article  PubMed  Google Scholar 

  309. Picelli S, et al. Smart-seq2 for sensitive full-length transcriptome profiling in single cells. Nat Methods. 2013;10(11):1096–8.

    Article  CAS  PubMed  Google Scholar 

  310. Hashimshony T, et al. CEL-Seq2: sensitive highly-multiplexed single-cell RNA-Seq. Genome Biol. 2016;17:77.

    Article  PubMed  PubMed Central  Google Scholar 

  311. See P, et al. A single-cell sequencing guide for immunologists. Front Immunol. 2018;9:2425.

    Article  PubMed  PubMed Central  Google Scholar 

  312. Cusanovich DA, et al. A single-cell atlas of in vivo mammalian chromatin accessibility. Cell. 2018;174(5):1309-1324.e18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  313. Rosenberg AB, et al. Single-cell profiling of the developing mouse brain and spinal cord with split-pool barcoding. Science. 2018;360(6385):176–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  314. Hasin Y, Seldin M, Lusis A. Multi-omics approaches to disease. Genome Biol. 2017;18(1):83.

    Article  PubMed  PubMed Central  Google Scholar 

  315. Baysoy A, et al. The technological landscape and applications of single-cell multi-omics. Nat Rev Mol Cell Biol. 2023;25:695–713.

    Article  Google Scholar 

  316. Lu GM, et al. Multiomics global landscape of stemness-related gene clusters in adipose-derived mesenchymal stem cells. Stem Cell Res Ther. 2020;11(1):310.

    Article  PubMed  PubMed Central  Google Scholar 

  317. Zou X, et al. Multi-omics analysis of an in vitro photoaging model and protective effect of umbilical cord mesenchymal stem cell-conditioned medium. Stem Cell Res Ther. 2022;13(1):435.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  318. Rampler E, et al. The power of LC-MS based multiomics: exploring adipogenic differentiation of human mesenchymal stem/stromal cells. Molecules. 2019;24(19):3615.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  319. Gao Y, et al. Multi-omics analysis of human mesenchymal stem cells shows cell aging that alters immunomodulatory activity through the downregulation of PD-L1. Nat Commun. 2023;14(1):4373.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  320. Zhou T, et al. Challenges and advances in clinical applications of mesenchymal stromal cells. J Hematol Oncol. 2021;14(1):24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  321. Giacomini C, et al. The critical role of apoptosis in mesenchymal stromal cell therapeutics and implications in homeostasis and normal tissue repair. Cell Mol Immunol. 2023;20(6):570–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  322. Kulebyakina M, Makarevich P. Hox-positive adult mesenchymal stromal cells: beyond positional identity. Front Cell Dev Biol. 2020;8:624.

    Article  PubMed  PubMed Central  Google Scholar 

  323. Sacchetti B, et al. No identical “mesenchymal stem cells” at different times and sites: human committed progenitors of distinct origin and differentiation potential are incorporated as adventitial cells in microvessels. Stem Cell Reports. 2016;6(6):897–913.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  324. RangaRao S, Subbarayan R. Passage-dependent expression of STRO-1 in human gingival mesenchymal stem cells. J Cell Biochem. 2019;120(3):2810–5.

    Article  CAS  Google Scholar 

  325. Tan L, et al. Characteristics and regulation of mesenchymal stem cell plasticity by the microenvironment-specific factors involved in the regulation of MSC plasticity. Genes Dis. 2022;9(2):296–309.

    Article  CAS  PubMed  Google Scholar 

  326. Wang Y, et al. Plasticity of mesenchymal stem cells in immunomodulation: pathological and therapeutic implications. Nat Immunol. 2014;15(11):1009–16.

    Article  CAS  PubMed  Google Scholar 

  327. Wang M, et al. Lymph node metastasis-derived gastric cancer cells educate bone marrow-derived mesenchymal stem cells via YAP signaling activation by exosomal Wnt5a. Oncogene. 2021;40(12):2296–308.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  328. Qiu X, et al. Exosomes released from educated mesenchymal stem cells accelerate cutaneous wound healing via promoting angiogenesis. Cell Prolif. 2020;53(8): e12830.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  329. Timaner M, et al. Therapy-educated mesenchymal stem cells enrich for tumor-initiating cells. Cancer Res. 2018;78(5):1253–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  330. Waterman RS, et al. A new mesenchymal stem cell (MSC) paradigm: polarization into a pro-inflammatory MSC1 or an Immunosuppressive MSC2 phenotype. PLoS ONE. 2010;5(4): e10088.

    Article  PubMed  PubMed Central  Google Scholar 

  331. Ryan JM, et al. Interferon-gamma does not break, but promotes the immunosuppressive capacity of adult human mesenchymal stem cells. Clin Exp Immunol. 2007;149(2):353–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Funding

Guangdong Basic and Applied Basic Research Foundation (2021A1515012484, 2023A1515010181); The fundamental research project of the Shenzhen Science and Technology Innovation Commission (20231120113324002).

Author information

Authors and Affiliations

Authors

Contributions

Si Chen and Bowei Liang wrote the draft; Jianyong Xu revised the manuscript.

Corresponding author

Correspondence to Jianyong Xu.

Ethics declarations

Competing interests

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

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 4.0 International License, which permits use, sharing, adaptation, 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 changes were made. 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/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chen, S., Liang, B. & Xu, J. Unveiling heterogeneity in MSCs: exploring marker-based strategies for defining MSC subpopulations. J Transl Med 22, 459 (2024). https://doi.org/10.1186/s12967-024-05294-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12967-024-05294-5

Keywords