Skip to main content
Stem Cells and Development logoLink to Stem Cells and Development
. 2013 Jan 22;22(9):1455–1476. doi: 10.1089/scd.2012.0637

Reviewing and Updating the Major Molecular Markers for Stem Cells

Raquel Calloni 1, Elvira Alicia Aparicio Cordero 2, João Antonio Pêgas Henriques 1, Diego Bonatto 1,
PMCID: PMC3629778  PMID: 23336433

Abstract

Stem cells (SC) are able to self-renew and to differentiate into many types of committed cells, making SCs interesting for cellular therapy. However, the pool of SCs in vivo and in vitro consists of a mix of cells at several stages of differentiation, making it difficult to obtain a homogeneous population of SCs for research. Therefore, it is important to isolate and characterize unambiguous molecular markers that can be applied to SCs. Here, we review classical and new candidate molecular markers that have been established to show a molecular profile for human embryonic stem cells (hESCs), mesenchymal stem cells (MSCs), and hematopoietic stem cells (HSCs). The commonly cited markers for embryonic ESCs are Nanog, Oct-4, Sox-2, Rex-1, Dnmt3b, Lin-28, Tdgf1, FoxD3, Tert, Utf-1, Gal, Cx43, Gdf3, Gtcm1, Terf1, Terf2, Lefty A, and Lefty B. MSCs are primarily identified by the expression of CD13, CD29, CD44, CD49e, CD54, CD71, CD73, CD90, CD105, CD106, CD166, and HLA-ABC and lack CD14, CD31, CD34, CD45, CD62E, CD62L, CD62P, and HLA-DR expression. HSCs are mainly isolated based on the expression of CD34, but the combination of this marker with CD133 and CD90, together with a lack of CD38 and other lineage markers, provides the most homogeneous pool of SCs. Here, we present new and alternative markers for SCs, along with microRNA profiles, for these cells.

Introduction

Stem cells (SC) are defined as a class of undifferentiated cells capable of self-renewal, perpetuating their population and giving rise to many types of committed or more specialized cells through differentiation [1]. SCs can be found during all stages of development from the embryo to the adult organism, and they consist of cells with varying differentiation potential.

Cells taken from the zygote to as far as the blastocyst stage are considered totipotent because they have the potential to generate a whole organism [2]. By the blastocyst stage, the cells become more specialized and are considered pluripotent. Embryonic stem cells (ESCs) are obtained at this stage and can generate tissues from the 3 germ layers [3], but they are not able to originate a whole individual [2]. Fetal and adult tissues also have several sources of SCs. These cells, however, have a limited differentiation potential that is less than that of ESCs. Mesenchymal stem cells (MSCs) and hematopoietic stem cells (HSCs) are found among this type of SCs.

The discovery of SCs has brought new possibilities to the scientific and clinical area, as they have the potential to be applied in cell replacement therapy, gene therapy, drug discovery, disease modeling, and developmental biology [46]. Nevertheless, the pool of SCs obtained from in vivo and in vitro conditions is not homogeneous; rather, the cells are in several stages of differentiation. Therefore, identifying unambiguous markers is essential for isolating the most primitive cells and for clearly identifying the different stages of undifferentiated and committed cells.

In this context, the aim of this review is to construct a molecular profile, including classical and new candidate molecular markers, of the 3 most studied human SCs: ESCs, MSCs, and HSCs.

Molecular Markers for ESC Characterization

ESCs are commonly isolated from the inner cell mass (ICM) during the blastocyst stage and possess the capacity to self-renew and to originate all cell types of an organism [7]. Since the first cultures of ESCs were established [8,9], considerable effort has been made to characterize a unique ESC-associated molecular signature. In 2007, the International Stem Cell Forum created the so-called “International Stem Cells Initiative” to establish an ESC molecular identity [10]. A total of 59 human ESC (hESC) lines were analyzed for cell-surface antigens and gene expression as potential markers for ESCs [10]. In the same year, a consensus ESC gene list and a consensus differentiation gene list were proposed by Assou and coworkers [11] based on 38 publications regarding ESC transcriptomes. They also created an online database [http://amazonia.montp.inserm.fr] where the transcriptome dataset is available.

The set of molecular markers commonly applied to identify ESCs consists of cell-surface proteins and genes specifically expressed in ESCs (Table 1). The characteristic cell-surface markers of ESCs were first detected in human embryonic carcinoma [1214]. Among them are stage-specific embryonic antigen-3 (SSEA-3) and 4 (SSEA-4) and the tumor rejection antigens (TRA-1-60 and TRA-1-81) [9,15]. These surface markers are observed in the ICM, but they are absent in the 2–8 cell and morula stages [16]. When ESCs are induced to differentiate, these antigens are downregulated, and SSEA-1 is upregulated [16,17]. Moreover, GCTM2, GCTM343, alkaline phosphatase, CD90, CD24, and CD9 are other surface molecules identified in hESCs [9,10,15,16, 18,19].

Table 1.

The Most Common Molecular Markers Used for Embryonic Stem Cells, Mesenchymal Stem Cells, and Hematopoietic Stem Cells Characterization

SC Molecular markers
ESCs
 Positive markers SSEA-3, SSEA-4, TRA-1-60, TRA-1-81, alkaline phosphatase, Nanog, Oct-4, and Sox-2.
 Negative markers SSEA-1.
MSCs
 Positive markers CD13, CD29, CD44, CD49e, CD54, CD71, CD73, CD90, CD105, CD106, CD166, and HLA-ABC.
 Negative markers CD14, CD31, CD34, CD45, CD62E, CD62L, CD62P, and HLA-DR.
HSCs
 Positive markers CD34, CD90, and CD133.
 Negative markers CD38 and lineage markersa.
a

A detailed list of negative lineage markers can be found on Table 8.

SC, stem cell; ESCs, embryonic stem cells; MSCs, mesenchymal stem cells; HSCs, hematopoietic stem cells; SSEA, stage-specific embryonic antigen; TRA, tumor rejection antigens.

In addition to surface molecules, there are some genes whose expression is characteristic of ESCs. Classically, the 3 transcription factors Nanog, Oct-4, and Sox-2 are used as indicators of the uncommitted status of an ESC [15,20]. Alternatively, other molecules (Table 2) are cited in the scientific literature as putative markers of ESCs, and all of them have their expression downregulated when these cells are induced to differentiate [9,15,18,19,2126]. Below, we discuss the genes most commonly used to confirm ESC identity. It should be noted that some of the genes listed in Table 2 are not discussed because there are none or very few studies about their roles in ESCs.

Table 2.

The Most Cited Candidate Embryonic Stem Cell Maker Genes in Literature

Gene abbreviation Gene name Biochemical functiona
Cx43 Connexin 43 Component of connexons.
DNMT3B DNA (cytosine-5) methyltransferase 3β Required for genome-wide de novo methylation and is essential for the establishment of DNA methylation patterns during development.
FOXD3 Forkhead box D3 Required for maintenance of pluripotent cells in the pre-implantation and peri-implantation stages of embryogenesis.
GAL Galanin Contracts smooth muscle of the gastrointestinal and genitourinary tract, regulates growth hormone release, modulates insulin release, and may be involved in the control of adrenal secretion.
GDF3 Growth differentiation factor 3 Control differentiation of ESCs in mice and humans. This molecule plays a role in mesoderm and definitive endoderm formation during the pre-gastrulation stages of development.
PODXL Podocalyxin-like Pro-adhesive molecule, enhancing the adherence of cells to immobilized ligands, increasing the rate of migration and cell-cell contacts in an integrin-dependent manner.
LEFTYA Left-right determination factor A Required for left-right (L-R) asymmetry determination of organ systems in mammals.
LEFTYB Left-right determination factor A Required for left-right axis determination as a regulator of LEFTY2 and NODAL.
LIN28 Cell lineage protein 28 Acts as a “translational enhancer,” driving specific mRNAs to polysomes and thus increasing the efficiency of protein synthesis. Its association with the translational machinery and target mRNAs results in an increased number of initiation events per molecule of mRNA and, indirectly, in stabilizing the mRNAs.
NANOG Nanog Transcription regulator involved in inner cell mass and ESCs proliferation and self-renewal. Imposes pluripotency on ESCs and prevents their differentiation towards extraembryonic endoderm and trophectoderm lineages.
OCT4 Octamer binding protein 4 Forms a trimeric complex with SOX2 on DNA and controls the expression of a number of genes involved in embryonic development
REX1 Zinc finger protein 42 Involved in self-renewal property of ESCs.
SOX2 SRY-related HMG box 2 Transcription factor that forms a trimeric complex with OCT4 on DNA and controls the expression of a number of genes involved in embryonic development.
TDGF1 Teratocarcinoma-derived growth factor 1 Play a role in the determination of the epiblastic cells that subsequently give rise to the mesoderm.
TERF1 Telomeric repeat binding factor 1 Component of the shelterin complex (telosome) that is involved in the regulation of telomere length and protection.
TERF2 Telomeric repeat binding factor 1 Component of the shelterin complex (telosome) that is involved in the regulation of telomere length and protection.
TERT Telomerase Catalytic component of the teleromerase holoenzyme complex whose main activity is the elongation of telomeres by acting as a reverse transcriptase that adds simple sequence repeats to chromosome ends by copying a template sequence within the RNA component of the enzyme.
UTF-1 Undifferentiated embryonic cell transcription factor-1 Acts as a transcriptional coactivator of ATF2, a transcriptional activator, probably constitutive, which binds to the cAMP-responsive element.
a

Extracted from GeneCards (www.genecards.org).

Classical Molecular Markers for ESC

Nanog

Named after the mythological Celtic land of the ever-young Tir nan Og, Nanog was first described in 2002 by 2 groups independently [27,28]. This transcription factor is a homeodomain protein whose expression is observed in the morula and ICM but is absent from unfertilized oocytes, 2- to 16-cell embryos, early morula, and trophectoderm [27,29]. Nanog is downregulated when organogenesis is initiated at the time of embryo implantation [27]. The silencing of the Nanog gene leads to the differentiation of ESCs into trophoectoderm and extraembryonic endodermal lineages, along with a downregulation of Oct-4 [29]. In murine ESCs (mESCs), the overexpression of Nanog can maintain these cells in an undifferentiated state even without LIF, likely by the inhibition of Gata4 and Gata6 [28]. The expression level of Nanog seems to be regulated by the inhibitor of differentiation 1 (Id1) protein [30], which acts as a negative regulator of helix-loop-helix DNA-binding proteins [31]. ESCs in which Id1 is knocked down display Nanog expression levels that are 35% lower than wild-type ESCs and exhibit a loss of the capacity to self-renew [31].

Oct-4

Oct-4, also known as Oct-3, Oct-3/4, POU5f1, OTF3, or NF-A3 [32], is another transcription factor that has roles in controlling the pluripotency of ESCs. It is expressed in unfertilized oocytes [7,32] and after fertilization as far as the 10-cell stage the observed transcripts are mainly of maternal origin and were expressed before zygote formation [32]. After the 10-cell stage, Oct-4 expression stabilizes, indicating the beginning of the embryonic production of Oct-4. During the blastocyst stage, Oct-4 can be observed in both the ICM and trophoectoderm, with Oct-4 levels higher in the former [32]. However, Oct-4 is highly expressed in the ICM of the early blastocyst but is absent from the trophoectoderm in mice [33]. The levels of Oct-4 determine the fate of ESCs because its downregulation leads to ESC differentiation into trophoectoderm [33,34], and an upregulation of less than 2-fold leads to ESC differentiation into extraembryonic endoderm and mesoderm [33].

An important point that Oct-4 alone is not sufficient to maintain an undifferentiated phenotype. The withdrawal of LIF from mouse ESCs leads to their differentiation despite the expression of Oct-4 [33].

Sox-2

Sox-2 is included in the SOX B1 group of transcription factors and has a single high-mobility group DNA-binding domain [35]. Together with Oct-4 and Nanog, Sox-2 plays a role in the maintenance of ESC pluripotency [36]. Its expression is first observed during the morula stage, followed by the ICM, epiblast, and cells from the extraembryonic ectoderm [36]. In addition, Sox-2 was also detected in the precursor cells of the developing central nervous system and in both male and female germ cells [37]. An up to 2-fold increase in the expression of Sox-2 leads to the differentiation of ESCs to ectoderm (mainly neuroectoderm), endoderm, and trophoectoderm lineages, likely due to an observed downregulation of genes (Oct-4, Nanog, FGF-4, UTF-1, Lefty-1) controlled by Sox-2 [36].

Sox-2-disrupted mice embryos fail to survive after implantation. Cultured cells from the entire blastocyst or ICM of these embryos differentiate into trophoblast cells and extraembryonic endoderm [37], thus suggesting that Sox-2 has a role in the maintenance of the undifferentiated status of epiblast cells.

The transcription factors detailed above act together in the maintenance of the SC status of ESCs. The knockdown of Sox-2 downregulates Oct-4 and Nanog, indicating that these transcription factors act together to maintain SC status [38]. Additionally, Sox-2 is unable to activate its target genes alone and must have a partner protein, which in ESCs is Oct-4 [35]. These 2 transcription factors bind to DNA as a heterodimer [33], and their targets include their own coding genes and Nanog [3941]. Nanog also regulates the expression of Sox-2 and Oct-4 [37,38] in a feed-forward manner.

Rex-1

Rex-1, also named zinc finger protein 42 (Zfp-42), is a transcription factor that was first identified in mice teratocarcinoma [12]. It has been studied primarily in mESCs, but its expression has also already been reported in hESCs [16,25]. In mice, Rex-1 is downregulated when the cells from the ICM differentiate into embryonic ectoderm, but it remains expressed in trophoblast cells [42]. In ESCs, Rex-1 appears to inhibit their differentiation, which is evidenced by the increased susceptibility of Rex-1−/− cells to differentiate after exposure to retinoic acid [43]. The expression of this transcription factor is regulated by Sox-2, Nanog, and Oct-4 [44]. An Oct-4-binding site has already been reported in the promoter of Rex-1, and its expression seems to be regulated by the levels of this transcription factor [45].

Dnmt3b

Dnmt3b is a de novo methyltransferase detected in oocytes, 2- to 4-cell embryos, and in the blastocyst stage in humans [46]. In mice, it is expressed in the ICM, epiblast, and embryonic ectoderm in a pattern similar to that observed for Oct-4 [46]. It presents 4 splicing variants, but only the Dnmt3b1 isoform is observed at these stages. This variant is observed in ESCs and, upon differentiation, its expression shifts to the Dnmt3b3 variant [47]. In mESCs, Dnmt3b interacts physically with Dnmt3a and stimulates its reciprocal activities [48]. Dnmt3a−/−/3b−/− mESCs show a progressive decrease in the levels of methylation together with an increasing inability to differentiate [49]. The impairment in the methylation levels affects the promoters of Oct-4 and Nanog; consequently, abnormal expression of these transcription factors during differentiation is observed [48]. In contrast, Dnmt3b does not seem to have a role in ESC self-renewal [50].

Foxd3

Foxd3, initially termed Genesis, belongs to the HNF-3/Forkhead transcriptional regulatory family [51]. In mice, Foxd3 is first detected during the blastocyst stage. It is not observed in either oocytes or during the first cleavage stages [52]. After ESCs differentiate, Foxd3 can still be detected in neural crest cells [53]. A lack of Foxd3 in mESCs increases the number of apoptotic cells [54]. Foxd3−/− mice embryos die during the gastrulation stage because of a loss of epiblast cells and the expansion of extraembryonic tissues [52]. However, the expression of the genes necessary for ESC maintenance (Oct-4, Sox-2, and Nanog) is not altered in Foxd3−/− embryos [52,54]. Thus, Foxd3 seems to be important for the survival and self-renewal of ESCs and to repress their differentiation [54]. There are studies reporting the interaction of Oct-4 and Nanog with Foxd3, although mRNA levels are not altered in cells lacking Foxd3 [54]. Oct-4 was shown to act as a co-repressor of Foxd3 [55], and it is regulated by Foxd3 together with Nanog, which counterbalance the inhibitory effect of Oct-4 on its own promoter [56].

Tdgf1

Tdgf1, also termed Cripto, is a member of the EGFP/TGFα growth factor family [57]. It is first detected at the blastocyst stage; during later stages of embryonic development, it is observed in the developing heart in mice [58]. It appears to have a role in blocking neural differentiation and in allowing ESCs to differentiate into cardiac cells [59]. Cripto−/− embryos die before birth and exhibit aberrant development and a lack of cardiogenic differentiation. Tdgf1 suppression in ESCs leads to their differentiation into neuronal lineages [60]. However, the proliferation levels of Crypto−/− ESCs are not different from wild-type ESCs [61].

Lin-28

Lin-28 is an RNA-binding protein that is highly expressed in hESCs [62] and is important for their growth and survival [63]. ESCs lacking Lin-28 show a decrease in their proliferation and a higher number of apoptotic cells when compared to wild-type ESCs [63,64]. High levels of Lin-28 expression at low cell densities slow the cell cycle and leads to differentiation toward an extraembryonic endoderm lineage [62]. Regarding its roles in ESCs, Lin-28 is involved in enhancing mRNA translation and the inhibition of some microRNA (miRNAs). Lin-28 acts on the let-7 miRNA family to block the processing of pri-let-7a and 7g in vitro. When Lin-28 is knocked down, the levels of mature let-7 family members are increased and are accompanied by decreasing in Oct-4 and Nanog expression. [65]. Lin-28 also regulates Oct-4 at the translational level, as its knockdown leads to a reduction in Oct-4 protein levels but not of its mRNA [63,64,66]. Oct-4 is also observed in Lin-28-associated polysomes, indicating that Lin-28 might be involved in the active translation of this transcription factor [66]. Other targets for translational activation are Cdk4 and cyclins A and B [64].

UTF-1

UTF-1 is a transcription factor that is stably associated with chromatin and acts as a transcriptional repressor [67,68]. During embryonic development in mice, UTF-1 cannot be observed in the morula but is upregulated at the blastocyst stage, specifically in the ICM. Recently, it has been observed in the primitive ectoderm and extraembryonic ectoderm [69]. ESCs with reduced levels of UTF-1 were delayed in differentiation and experienced perturbed EB formation [67,68], but their self-renewal was not affected, which resulted in increased expression levels of several genes. The explanation for this phenotype is that UTF-1 promotes chromatin condensation of its target genes, preventing their aberrant expression [68]. Moreover, it has been suggested that UTF-1 might maintain an ESC chromatin state that is susceptible to differentiation stimuli [67].

UTF-1 is bound by Oct-4 and Sox-2 in regulatory regions located at 3′ position of its gene, as demonstrated by in vitro assays [70,71]. There is an overlap between genes regulated by UTF-1 and those that are targets of Nanog, Sox2, Dax1, Nac1, Oct-4, Klf4, Zfp-281, Rex1, and c-Myc [69].

Novel Cell Markers for ESCs

Applying ESCs to cellular therapy is not feasible for many reasons. First, ESCs display a high potential for generating tumors in vivo. Moreover, the isolation of a pool of ESCs requires the destruction of human embryos, which raises ethical concerns about their use in cell therapy.

For clinical applications, the determination of markers that identify undifferentiated ESCs from a pool of cells ready for transplantation is desirable because this would allow tumor induction to be avoided [72]. Additionally, it is important to achieve a homogeneous pool of ESCs for basic and applied studies in vitro, allowing for the better characterization of cellular and molecular properties of those cells [72].

In addition to the classical ESC markers discussed above, surface proteins and highly expressed genes have been proposed as new ESC markers. Regarding surface proteins, ESCs have been reported to be positive for CD24, CD30, CD49f, CD50, CD90, CD133, CD200, and CD326. From this set of markers, CD133 and CD326 have been proposed as ESC markers due to their downregulation after the induction of neuronal differentiation [73,74]. On the other hand, CD24 may not be proposed as a marker due to its detection in differentiated cells [75]. CD30, although expressed in ESCs and downregulated under differentiation, was reported to be present on ESCs plasma membrane as a consequence of the culture media, specifically in serum-free condition [76]. Together with those proteins, a new surface marker was recently added to the list of the SSEA-associated membrane molecules of ESCs. Named SSEA-5, its expression is detected in the ICM of human blastocist and it was reported to be 5-fold lower when ESCs were induced to differentiate. Moreover, SSEA-5 was suggested as a marker for removing remaining undifferentiated ESCs in conditions of differentiation induction [74]. Additionally, the combined analysis of SSEA-5 together with CD9, CD50, CD90, and CD200 was more effective in detecting potential teratoma cells within differentiating ESCs [74].

In this sense, it is important to note that additional surface markers can be discovered from plasma membrane proteomics studies (for more details on ESCs plasma membrane proteomics, see the Ref. [77]).

Within ESCs, other highly expressed genes and putative new markers include line-type transposase domain containing 1 protein (L1TD1), Forkhead box O1 (FOXO1), and E1B-AP5. L1TD1 is highly expressed in ESCs and is absent from most adult tissues. In silico analysis revealed that it is restricted to the blastocyst stage, where its expression is downregulated during differentiation in a pattern similar to that observed for Oct-4, Nanog, and Sox-2. In addition, L1TD1 is a downstream target for Nanog protein [78].

FOXO1 is also expressed at higher levels in ESCs, is downregulated during ESC differentiation, and has been implicated in the regulation of ESC pluripotency. Knockdown of FOXO1 does not alter the self-renewal of ESCs but is accompanied by the downregulation of Oct-4, Nanog, and Sox-2, leading to the spontaneous differentiation of ESCs into mesoderm and endoderm lineages. Moreover, FOXO1 appears to act by activating Sox-2 and Oct-4 expression [79].

Adenovirus early region 1B-associated protein 5 (E1B-AP5) is a nuclear RNA-binding protein observed in the nucleus and cytoplasm, where its phosphorylated form is presented at the ESC surface and can be used as a specific marker for ESCs. Cells positive for this marker are also positive for the classical ESC markers (ie, SSEA-3, SSEA-4, TRA-1-60 and TRA-1-81, Nanog, Oct-4, and Sox-2). Moreover, this surface protein was not detected after cell differentiation [72].

Transcriptome studies are also a potential source of new ESC markers [18,19,2126]. These reports have revealed many genes that are enriched in ESCs and also downregulated during cell differentiation. All of these genes can be considered putative markers of an undifferentiated state, but only a few have had their roles in ESCs investigated. Table 3 summarizes the results from transcriptome studies comparing the expression profiles of ESCs and differentiated cells.

Table 3.

Genes Expressed at Higher Levels in Embryonic Stem Cells When Compared to Differentiated Cells Based in Transcriptomic Data

ESC lineage Differentiated cells ESC-associated transcripts differentially expressed Reference
HES3, HES4 Cells from fetal brain, fetal liver, adult brain, placenta, adult testis, adult kidney, adult lung, adult heart CCNB1, CFL1, CGI-48, CHK2, CKS1B, CLDN6, Cx43, DNMT3B, ERH, FAM60A, FAS, FLJ10713, FZD7, GDF3, GTCM1, HMGA1, LIN-28, NCL, Numatrin, PCNA, PNF1, POU5F1, REX-1, SFRP1, SOX-2, TDGF-1, TERF-1, TMSB15A. [25]
BG02 Embryoid body ARL8, BRIX, C15orf15, C20orf1, C20orf129, C20orf168, CCNB1, CCNC, CCT8, CDC2, CER1, CRABP1, CRABP2, CYP26A1, DDX21, DNMT3B, EIF4A1, ELOVL6, EPRS, FABP5, GAL, GDF3, GJA1, GSH1, GTCM-1, HDAC2, HMGB2, HMGIY, HNRPA1, HNRPAB, HSSG1, IDH1, IFITM1, IMP-2, IMPDH2, Jade-1, KIAA1573, KIF4A, KPNA2, Laminin receptor, LAPTM4B, LDHB, LEFTB, Lin-28, LRRN1, MAD2L2, MGST1, MTHFD1, MTHFD2, Nanog, NASP, NBR2, NME2, NPM1, NS, Numatrin, POU5F1, PPAT, PSMA2, PSMA3, PTTG1, RAMP, RPL24, RPL4, RPL6, RPL7, SEMA6A, SET, SFRP2, SLC16A1, SMS, SNRPF, SOX2, SSB, STK12, TD-60, TDGF1, TK1. [26]
GE01/GE07/GE09 (pooled RNA) Embryoid body C20orf129, CCNB1, CCNC, CRABP1, CYP26A1, ELOVL6, FABP5, FLJ12581/Nanog, HDAC2, HSPA4, JADE-1, KIAA1573, KPNA2, LEFTB, MGC27165, GST1, NASP, NS, PSMA3, PTTG1, RAMP, RPL17, SEMA6A, SFRP2, SLC16A1, TDGF1, TNNT1, ZNF257. [80]
H1 Embryoid bodies AK3, DUSP6, E2IG5, FLJ10713, FRAT2, GAL, LEFT B, MYO10, PLP1, POU5F1, PROML1, PSIP2, SPS, STRIN, VRK1, VSNL1. [81]
CH3, CH4 Embryoid bodies A2ML1, AASS, ADCY2, ADD2, AK5, ARTN, C14orf115, C1orf182, C9orf61, CABYR, CACNA1G, CAMKV, CDCA7L, CHST4, CKMT1, CNTN1, CRABP1, CTGF, CXCL6, DCAMKL1, DDX25, DEPDC2, DNMT3B, DPPA2, DPPA4, FBXL16, FGF2, FLJ12505, FLJ12684, FLJ30707, GABRA5, GABRB3, GAP43, GPC4, GPR19, GPR23, GRPR, HESX1, INA, INDO, INHBE, ITGB1BP3, LEFTY1, LOC168474, LOC283174, MDN1, NALP4, NANOG, NAP1L2, NEF3, NEFL, NELL2, NMNAT2, NMU, NPTX2, OLFM1, OSBPL6, PCSK9, POU5F1, PTHB1, PTPRB, PTPRZ1, RAB39B, RARRES2, RASL11B, RDH12, RET, RNF182,SAMHD, SCG3, SCGB3A2, SEPHS1, SLC10A4, SLC7A3, SOX2, SYT1, TAC1, TAF4B, TDGF1,TERF1, TIMP4 TNFRSF8, USP44, WIF1, ZIC3. [82]
H1, H7, H9 Embryoid bodies, immature hepatocytes and putative neural cells FLJ35207, FOXH1, FOXO1A, GABRB3, GAP43, GRPR, PHC1, PODXL1, POU5F1, PRDM14, PTPRZ1, SALL1, SALL2, SZF1, THY1, ZIC2, ZIC3, ZNF206. [21]
BG01, BG02, BG03 Embryoid bodies CKMT1, DIAPH2, DNMT3B, EBAF, GABRB3, GDF3, GYLTL1B, IFITM1, LCK, LIN28, MIBP, NTS, PMAIP1, POU5F1, TDGF1, UTF1, ZNF206, ZPF42. [83]

FOXO1, forkhead box O1.

miRNAs associated with ESCs

Studies of mESCs have revealed that null Dicer cells display a pronounced loss in proliferative capacity [84]. The lack of DGCR8, another important member of the miRNA generation pathway [85], appears to cause alterations in mESCs; these cells exhibited extended population doubling, increased numbers of cells in the G1 phase, and differentiation impairment [86]. These results indicate that miRNAs possibly have important roles in the biology of ESCs, and several have been identified as being exclusive to those cells, such as clusters miR-302-367, miR-520, and miR-371/372/373.

The miR-302-367 cluster is located on chromosome 4 and is composed of 9 miRNAs disposed in a polycistronic manner: miR-302a, miR-302a*, miR-302b, miR-302b*, miR-302c, miR-302c*, miR-302d, miR-367, and miR-367* [87]. This cluster is highly expressed in ESCs [88,89], more than 20-fold when compared to adult cells or hMSCs, and its expression is rapidly downregulated when cells are induced to differentiate. It has also been detected in induced pluripotent cells [90]. In addition, putative binding sites for Oct3/4, Nanog, Rex1, and Sox-2 have been identified within its promoter sequence, proving its function in ESCs [88]. Indeed, Card et al. [91] showed that Oct-4 and Sox-2 bind to the promoter of this cluster.

Another typical ESC miRNA cluster is miR-520. It is located on chromosome 19 and is composed of 21 miRNAs: 515-5p, miR-517a, miR-517b, miR-517c, miR-518b, miR-518c, miR-519b, miR-519c, miR-519e, miR-520a, miR-520b, miR-520c, miR-520d, miR-520e, miR-520f, miR-520g, miR-520h, miR-521, miR-524*, miR-525-3p, and miR-526b* [89]. The roles of these 2 clusters in ESCs include (i) cell growth arrest, (ii) negative regulation of cellular metabolic processes, (iii) negative regulation of transcription, and (iv) small GTPase-mediated signal transduction [89].

The miRNAs miR-371-372-373 also comprise a cluster located on chromosome 19. This cluster is highly expressed in ESCs [88], and as observed for other miRNA clusters, its expression is rapidly downregulated when the cells are induced to differentiate [92].

Moreover, miRNAs initially observed in MSCs were also identified in ESCs, such as miR-9, miR-28, miR-29, miR-42, miR-63, and miR-89 [93]. These miRNAs are downregulated during the transition from ESCs to endothelial cells. Interestingly, miR-9 and miR-28 act by inhibiting CDH5 and endoglin translation, respectively [94]. In addition to the miRNAs identified in ESCs, low levels of piRNA, small noncoding RNAs that interact with Piwi proteins, were also observed in these cells [94]. The occurrence of 3′ modifications and RNA editing in miRNAs observed in ESCs have also been reported [94].

The roles of miRNAs in ESCs are still not fully understood. Research in mice has revealed that miRNAs typical of mESCs have roles in cell proliferation, being implicated in promoting G1 to S phase transition through the suppression of the G1/S inhibitors [95]. Similar results were observed in hESCs; the miR-302 cluster was shown to be involved in the progression from G1 to S phase. Cell cycle regulators, cyclin D1, and possibly Cdk4 are targets of the miRNAs from this cluster [91]. Recently, NR2F2 (nuclear receptor subfamily 2, group F, member 2) was reported as a target for miR-302 [96]. Oct-4 induces the miR-302 expression and it is inhibited by NR2F2 [96].

Although little is known about the function of the miRNAs that are enriched in ESCs, some of them have already been reported as good reprogramming factors for the induced pluripotent stem cells (iPSCs) generation. Among the miRNAs successfully applied on reprogramming of fully differentiated cells into iPSCs, there are the cluster miR-302-367 [97], the miR-302b alone [96,98], the combination of miR-302, miR-369, and miR-200c [99], the miR-302b associated with miR-372, and the miR-372 alone [98]. These miRNAs are generally associated with factors as Oct-4, Sox-2, Klf4, Nanog, and c-Myc [9698].

Published miRNA profiles of hESCs are summarized on Table 4. The data displayed correspond to the highly expressed miRNAs as listed by the authors of each article.

Table 4.

microRNAs Highly Expressed in Embryonic Stem Cells, When Compared to Embryoid Bodies or Differentiated Cells

Lineage Highly expressed miRNA profile Reference
hES-T3 miR-20b, miR-26b, miR-200c, miR-302a*, miR-302b*, miR-302c*, miR-302d, miR-367, miR-371, miR-372, miR-373. [100]
H9, I6,BG01v miR-96, miR-127, miR-141, miR-200b, miR-200c, miR-299-3p, miR-302a, miR-302a*, miR-302b, miR-302b*, miR-302c, miR-302d, miR-324-3p, miR-367, miR-369-3p, miR-372, miR-515-5p, miR-517a, miR-517b, miR-517c, miR-518b, miR-518c, miR-519b, miR-519c, miR-519e, miR-520a, miR-520b, miR-520c, miR-520d, miR-520e, miR-520f, miR-520g, miR-520h, miR-521, miR-524*, miR-525, miR-525*, miR-526b*, miR-550-2, miR-612. [89]
WA09, WA01, HSF6, HUES7, HUES13 miR-302 cluster, miR-371/372/373 cluster, primate-specific placenta associated cluster (54 miRNAs), miR-17, miR-106a. [101]
SNU-hES3 miR-154*, miR-200c, miR-302a*, miR-302a, miR-302b*, miR-302b, miR-302c, miR-302c*, miR-302d, miR-371, miR-372, miR-373, miR-373*, miR-368. [97]
Cyt25, Cyt203, HES2, HES 3, HES4 miR-21, miR-200c, miR-222, miR-296, miR-302a, miR-302c, miR-367, miR-371, miR-372, miR-373, miR-320d, miR-494. [102]
H1 miR-18b, miR-20b, miR-92b, miR-154, miR-184, miR-187, miR-302a, miR-302b, miR-302c, miR-302d, miR-324-3p, miR-363*, miR-512-3p, miR-518b, miR-518c, miR-519d, miR-520g, miR-524*, miR-1323, miR-1901, miR-1908, miR-1910, miR-1911. [103]
H9 Let-7a, Let-7b, Let-7c, Let-7d, Let-7g, Let-7i, miR-7, miR-92, miR-106b, miR-155, miR-181d, miR-184, miR-185, miR-187, miR-211, miR-222, miR-296, miR-302a, miR-302d, miR-331, miR-424, miR-484, miR-486, miR-503, miR-519c, miR-520, miR-518c, miR-519a, miR-574, miR-594, miR-744, miR-760, miR-766, miR-766*, miR-874, miR-877, miR-941, miR-1298, miR-1308, miR-1246, miR-1254, miR-1261, miR-1266, miR-1268, miR-1272, miR-1275, miR-1301, miR-1306, miR-1307, miR-1308. [98]

Asterisk indicates miRNA strands less commonly found associated with Argonauta complex.

miRNA, microRNA.

Interestingly, a comparison between ESCs and iPSCs showed that both cells have similar groups of upregulated miRNAs. Among them there are the clusters of miR-302-367 and miR-17-92 (composed by miR-17, miR-18a, miR-19a, miR-20a, miR-19b-1, and miR-92). However, members of the miR-371-372-373 and miR-520 clusters were downregulated in iPSCs [90]. For more details on iPSCs miRNA profile, see Ref. [90].

Molecular Markers for MSC Characterization

MSCs are multipotent, adherent SCs capable of differentiating into osteoblasts, adipocytes, and chondroblasts [104] and also to nonmesenchymal lineages such as pancreatic islands [105], hepatocytes [106,107], and neuron-like cells [108]. MSCs were first isolated from bone marrow by A.J. Friedenstein [109]; nevertheless, many researchers have been able to isolate them from other regions of the human body, including adipose tissue [110,111], heart [111], trabecular bone [112], vessels [113], peripheral blood [114], skin [115], deciduous teeth [116], and many others. MSCs are also found in fetal tissues and fluids, such as amniotic fluid, umbilical cord blood [117], amnion, and placenta [118].

When MSCs are isolated from different tissues and organs, a heterogeneous pool of cells with several differentiation potentials can be obtained [119]. In addition, MSCs are not present in high quantities in tissues. For example, in the bone marrow, MSCs comprise 0.01% to 0.001% of the total cell number [96]. Therefore, the definition of markers that make it possible to isolate the most primitive MSCs and to identify those subpopulations with different potentials to generate mature cells is necessary to improve the cellular and molecular characterization of MSCs. Thus, the focus of this section will be on information about classical markers for MSCs, recently reported or alternative markers, and the miRNA profile of MSCs.

In 2006, The International Society for Cellular Therapy published the minimal criteria to identify a human SC as an MSC [120]. Among these are the expression of the surface proteins CD73, CD90, and CD105 together with the lack of expression of CD45, CD34, CD14 or CD11b, CD79a or CD19, and HLA-DR [120]. However, many other markers have been identified and used by researchers (Table 5).

Table 5.

Surface Markers for Different Mesenchymal Stem Cells Reported in Scientific Literature

Source Positive markers Negative markers References
Adult origin
Bone marrow CD29, CD44, CD71, CD73, CD90, CD105, CD106, STRO-1. CD13, CD14, CD34, CD45, CD133, CD144. [106,117,121]
Deciduous teeth CD13, CD73, CD105, CD146, STRO-1. CD34, CD43, CD45. [116,122]
Permanent teeth STRO-1. No negative markers reported. [122]
Periodontal ligament CD9, CD13, CD29, CD44, CD49a, CD49b, CD49c, CD49d, CD49e, CD51/61, CD71, CD73, CD90, CD105, CD106, CD119, CD120a, CD146, CD166, STRO-1, HLA-ABC. CD2, CD3, CD8a, CD11a, CD14, CD16, CD18, CD19, CD20, CD24, CD25, CD31, CD33, CD34, CD38, CD41a, CD45, CD50, CD54, CD56, CD62E, CD62L, CD62p, CD66b, CD104, CD117, CD121a, CD123, CD124, CD126, CD133, CD235a, CD318, HLA-DR, STRO-1. [124126]
Apical papilla CD146, STRO-1. No negative markers reported. [127]
Skin CD13, CD29, CD44, CD49d, CD71, CD73, CD105, CD166. CD10, CD11b, CD14, CD31, CD34, CD45, CD133, CD106, HLA-DR. [128130]
Endometrium CD29, CD44, CD73, CD90, CD105, CD146. CD31, CD34, CD45, STRO-1. [131]
Adipose tissue CD9, CD10, CD13, CD29, CD34*, CD44, CD49d, CD49e*, CD54, CD55, CD59, CD71, CD73, CD90, CD105, CD166.STRO-1, HLA-ABC. CD11a, CD11b, CD11c, CD14, CD16, CD18, CD31, CD34, CD45, CD50, CD56, CD61, CD62e, CD104, CD106, CD133, CD144, HLA-DR. [110,117,132]
Periferal blood CD44, CD105, STRO-1. CD3, CD14, CD20, CD45, CD68, CD106, HLA-DR. [114]
Menstrual blood CD9, CD29, CD41a, CD44, CD59, CD73, CD90, CD105. CD14, CD31, CD34, CD38, CD45, CD133, STRO-1. [133]
Saphenous vein CD13, CD29, CD44, CD54, CD90, HLA-I CD14, CD31, CD34, CD45, CD133. [134]
Limbal stroma–eye CD29, CD54, CD71, CD90, CD105, CD106, CD166. CD11a, CD11c, CD14, CD31, CD34, CD45, CD138. [135]
Lung CD13, CD73, CD90, CD105, CD166, HLA-I, STRO-1. CD14, CD34, CD45, HLA-DR, HLA-DQ. [136,137]
Fetal origin
Amniotic fluid CD13, CD29, CD44, CD49e, CD73, CD90, CD105, CD166, HLA-ABC. CD1a,CD10,CD11a,CD11b,CD14,CD31,CD34,CD45, CD49d,CD50,CD54,CD62E,CD62P,CD117,CD133, HLA-DR,HLA-DP,HLA-DQ. [138,139]
Bone marrow CD29, CD44, CD49e, CD54, CD58, CD73, CD90, C105, CD106, CD123, CD166, HLA-ABC. CD14,CD25,CD31,CD34,CD40,CD45,CD49d,CD50, CD62E,CD62L,CD62P,CD68,CD80,CD86,CD106, CD120a,CD120b,CD127,HLA-DR. [140,141]
Peripheral blood CD29, CD44, CD73, C105, CD106. CD14,CD31,CD34,CD45,CD68,HLA-DR [140]
Liver CD29, CD44, CD49e, CD54, CD58, CD73, CD90, CD105, CD106, CD123, CD166, HLA-ABC. CD14,CD25,CD31,CD34,CD40,CD45,CD49d,CD50, CD62L,CD62P,CD68,CD80,CD86,CD106,CD120a, CD120b,CD127,HLA-DR. [140,141]
Lung CD44, CD49e, CD54, CD58, CD73, CD90, CD105, CD123, CD166, HLA-ABC. CD14,CD25,CD31,CD34,CD40,CD45,CD49d, CD50,CD62E,CD62L,CD62P,CD80,CD86,CD106, CD120a,CD120b,CD127,HLA-DR. [141]
Pâncreas CD13, CD29, CD44. CD34,CD45,HLA-DR. [142]
Placenta CD29, CD44, CD49e, CD54, CD73, CD90, CD105, CD166, HLA-ABC. CD31,CD34,CD45,CD49d,CD123,CD133,HLA-DR. [118,143]
Spleen CD44, CD49e, CD54, CD58, CD73, CD90, CD105, CD123, CD166, HLA-ABC. CD14,CD25,CD31,CD34,CD40,CD45,CD49d, CD50,CD62E,CD62L,CD62P,CD80,CD86,CD106, CD120a,CD120b,CD127,HLA-DR. [141]
Umbilical cord vein CD29, CD73, CD90, CD105, CD166. CD14,CD31,CD34,CD45. [105]
Umbilical cord blood CD29, CD44, CD49b, CD49d, CD51, CD58, CD71, CD73, CD90, CD105, CD106, HLA-ABC. CD3,CD7,CD11a,CD11b,CD14,CD19,CD33,CD34, CD45,CD62L,CD62P,CD117,CD133,CD135,CD31, CD144, LA-DR. [118,144,145]
Wharton's jelly CD10, CD13, CD29, CD44, CD49e, CD51, CD68,CD73, CD80, CD90, CD105, CD117, CD166, HLA-ABC. CD14,CD31,CD33,CD34,CD45,CD56,CD86,CD163, HLA-DR. [146]
*

Less than 28% of positive cells for this marker. The authors considered this percentage as positive.

Some of the markers listed above appear to be dependent on the original tissue where the MSCs were isolated, but many are common among all MSCs. Based on the scientific literature, we suggest a list of common positive and negative surface markers found in MSCs (Table 1).

Together with these surface markers, several articles have reported the expression of some ESC-associated markers in MSCs from different sources (Table 6). The expression levels of some of these markers are downregulated when MSCs are induced to differentiate followed by an increase in SSEA-1 [122,124]. These changes in MSC marker expression recapitulate what is observed during ESC differentiation.

Table 6.

Major Embryonic Stem Cells Markers Observed in Mesenchymal Stem Cells

ESC marker detected Source of cells Reference
Oct-4, Nanog, Sox2, SSEA-4 Bone marrow, adipose tissue, dermis, and heart [115]
OCT-4, Nanog, SSEA-3, SSEA-4, TRA-1-60, low level of cells were TRA-1-80 Pulp of deciduous teeth [122]
OCT-4, Nanog, SSEA-4 Amniotic fluid [139,147]
OCT-4, Nanog, SSEA-3, SSEA-4, TRA-1-60, TRA-1-81 Fetal bone marrow, liver, and blood [148]
SSEA-4 Dermis [149]
SSEA-3, SSEA-4, TRA-1-60, TRA-1-81, OCT-4, Nanog, Sox-2, REX-1 Periodontal ligament [124,150]
Nanog Bone marrow, adipose, and cardiac tissue [111]
Oct-4 Menstrual blood [133]
Oct-4, Nanog, SSEA-4 Bone marrow and placenta [151]

The real function of the ESC-associated markers in MSCs is not completely understood, and their presence has been considered as a primitive phenotype and an indication of the stem potential of the cells [141]. On the other hand, the expression of Nanog in MSCs could be due to a transition from in vivo to in vitro conditions, from the quiescent to the proliferative state [111]. In fact, Nanog seems to have roles in the maintenance and differentiation of MSCs in vitro. Studies with murine MSCs reported that the expression of this transcription factor is downregulated during differentiation. In addition, Nanog overexpression or knockdown leads to an increase or a reduction in cell proliferation, respectively [152]. In vitro, the knockdown of NANOG also resulted in the elevation of osteocalcin expression, a marker of osteogenic differentiation. In vivo, during the healing of an induced bone injury, Nanog expression was detected early in the process, preceding the expression of osteogenic differentiation markers. The timing of Nanog expression can be explained by the necessity of MSC population expansion, whose cells will be recruited for the healing process [152]. When the same healing experiment was repeated and Nanog expression was blocked, osteogenic differentiation was impaired, and adipogenic cells were observed [152]. In fact, Nanog seems to be related to favoring MSC differentiation to an osteogenic rather than an adipogenic fate. A decrease in Nanog expression is observed during adipogenic differentiation [153], and when Nanog is overexpressed in MSCs induced to adipogenic differentiation, there is a decrease in the expression of adipogenic markers and weaker Oil red staining [154].

Novel and Alternative MSC-Associated Markers

Although great progress has been made regarding the definition of MSC markers, we are still far from defining a specific molecular signature for these cell types. Here, we discuss novel and alternative markers reported for MSCs.

A study of plasma membrane surface proteins of MSCs derived from bone marrow detected 113 transcripts, including 20 CDs, expressed by MSCs but not by hematopoietic cells. From this group, 8 markers (CD49b, CD73, CD90, CD105, CD130, CD146, CD200, and integrin αV/β5) allowed for the isolation of MSCs from bone marrow mononuclear cells. CD200 has been proposed as a molecular marker to isolate bone marrow MSCs because cells isolated using this marker display a high enrichment in colony-forming units-fibroblasts when compared to the total mononuclear fraction before sorting and were able to differentiate into osteogenic, adipogenic, and chondrogenic lineages [155].

Battula et al. [151] have proposed FZD9 (Frizzled-9 or CD349) as a marker for primitive MSCs. The cells isolated from placenta that display FZD9 exhibited high clonogenic potential, which was enhanced when FZD9 was combined with CD10 and CD26 [156]. Additionally, this fraction expressed high levels of Oct-4, Nanog, and SSEA-4 [147]. However, Tran et al. [157] proposed that FZD9 is a good marker for the isolation of MSCs specifically for arterio/angiogenic therapy but not for discriminating between MSCs and non-MSCs. The expression of this marker was also observed in MSCs isolated from the periodontal ligament and was downregulated after the second passage [157].

Kaltz et al. [158] reported NOTCH-3 as a marker for the enrichment of MSCs capable of both osteogenic and adipogenic differentiation from bone marrow. In addition, the same research group demonstrated that ITGA11 and MSCA-1 could be used as markers for bone marrow MSC-derived cells that are mainly unipotent: osteogenic or adipogenic, respectively.

Using an antibody against the nerve growth factor receptor (NGFR or CD271), Quirici et al. [159] were able to isolate highly proliferative MSCs that were prone to osteogenic and adipogenic differentiation. Moreover, the authors observed that these cells were able to support the growth of hematopoietic progenitors [159]. The CD271bright subpopulation had been reported to contain a fraction of highly clonogenic bone marrow MSCs [160]. The MSCA-1+CD56+ fraction of this subpopulation demonstrated a high clonogenic potential and osteogenic, chondrogenic, and pancreatic differentiation, but it was unable to generate adipose cells [161]. These cells also present the capacity to inhibit T-cell proliferation and the differentiation of monocytes to dendritic cells [161]. Bühring et al. [160] observed that combining CD271 with CD140b, W8B, HEK-3D6, FZD-9, and CD56 makes it possible to isolate highly clonogenic MSCs.

GD2, a disialoganglioside, is another proposed MSC marker. It is detected in CD45 and CD73+, CD90+ MSCs isolated from bone marrow, but it is not observed in other cells from the same compartment, such as leukocytes, myeloid cells, T-lymphocytes, B-lymphocytes, or hematopoietic progenitors [162]. MSCs isolated from adipose tissue also present GD2 at the same levels as detected in MSCs from the bone marrow [162]. MSCs isolated from the umbilical cord also present GD2, and it can be detected as far as the 10th passage [163]. Cells isolated based on this marker were able to differentiate into adipocytes, osteoblasts, chondrocytes, and neuronal cells [162164]. However, it has been reported that the GD2 and GD2+ fractions have the same MSC marker profile and a similar potential to differentiate. Moreover, GD2 cells show higher proliferation rates than GD2+ cells [164]. Nevertheless, the inhibition of GD2 synthesis leads to a block of neuronal differentiation [164].

SSEA-4, a classical ESC marker, has been demonstrated to be useful for isolating MSCs with potential to differentiate into adipocytes, chondroblasts, osteoblasts, pancreatic, and neuronal cells, together with the capacity for forming organized bone tissue in vivo [124,151,165].

miRNAs associated with MSCs

Little is known about the miRNAs involved in the regulation of MSCs. The majority of the reports about miRNAs expressed in MSCs have focused on molecules with roles in the osteogenic [166168], adipogenic [169171], and chondrogenic [172,174,175] differentiation pathways. There are few reports exploring miRNA profiles and their functions associated with the maintenance of the stem state of MSCs (Table 7).

Table 7.

microRNA Profile Reported for Mesenchymal Stem Cells from Different Sources

MSC source miRNAs Reference
Bone marrow miR-15b, miR-16, miR-21, miR-22, miR-23a, miR-23b, miR-24, miR-26a, miR-27a, miR-27b, miR-145, miR-29a, miR-30d, miR-99a, miR-100, miR-103, miR-107, miR-125a, miR-130a, miR-143, miR-181a, miR-191, miR-193a, miR-193b, miR-199a, miR-199a*, miR-210, miR-214, miR-221, miR-222, miR-320. [166]
Bone marrow miR-18b, miR-21, miR-122a, miR-132, miR-140, miR-143, miR-145, miR-181a*, miR-181a-2, miR-181c, miR-335*, miR-337, miR-340, miR-409-5p, miR-431, miR-491, miR-519b, miR-520f, miR-520e, miR-520g, miR-652. [173]
Adipose tissue miR-16, miR-19b, miR-20a, miR-21, miR-24, miR-26a, miR-26b, miR-29a, miR-30c, miR-31, miR-92, miR-93, miR-99a, miR-119a, miR-125a, miR-125b, miR-127, miR-140, miR-146a, miR-146b, miR-152, miR-106b, miR-181d, miR-186, miR-190, miR-19, miR-214, miR-221, miR-270, miR-320, miR-339, miR-342, miR-365, miR-376a, Let-7a, Let-7b, Let-7g, Let-7i. [176]
Bone marrow miR-16, miR-21, miR-22, miR-23a, miR-23b, miR-24, miR-26a, miR-27a, miR-27b, miR-29a, miR-30a-5p, miR-31, miR-100, miR-125a, miR-125b, miR-143, miR-145, miR-152, miR-191, miR-199a, miR-199a-AS, miR-214, miR-221, miR-222, miR-320, Let-7a, Let-7c, Let-7d, Let-7e, Let-7f, Let-7i, Lrt-7b. [177]

Asterisk indicates miRNA strands less commonly found associated with Argonauta complex.

Molecular Markers for the Characterization of HSCs

HSCs are adult SCs found in the bone marrow [178], umbilical cord blood [179], fetal liver [180], and peripheral blood after mobilization [181,182]. A true HSC must be able to self-renew and give rise to all of the mature cells that comprise the hematopoietic system [183]. One approach to check the stemness of a candidate HSC is to test its capacity for performing a long-term reconstitution of hematopoiesis in recipient animals subjected to myeloablative treatment [183].

The identification of molecular markers that can characterize true primitive HSCs will make their isolation from the heterogeneous pool of cells where they are located easier. Below, we list the classical molecular markers reported by the scientific literature for the enrichment of highly primitive HSCs (Table 1), together with new candidate markers and the miRNA profiles of these cells.

Classical and Alternative HSC-Associated Markers

CD34+

CD34+ is a member of the sialomucin family of surface molecules [184]. It is the classical marker for HSCs, although its functions are not completely understood. It has been speculated that CD34 plays roles in cell adhesion and/or HSC differentiation [184]. Murine cells constitutively expressing this surface protein failed to completely differentiate [185]. Nevertheless, Nielsen and McNagny [186] argue that this blockage of differentiation may be due to inhibitory effects of CD34 on cell adhesion.

The bone marrow fraction positive for this molecule is known to be capable of hematopoietic reconstitution in recipients that underwent myeloablative therapy [187,188], indicating the presence of primitive HSCs. Interestingly, this marker is also expressed in vascular endothelial cells [189].

Nevertheless, there is no consensus regarding whether the most primitive HSCs are located in the CD34+ fraction. In 1996, Osawa et al. [190] observed that murine cells negative for this marker were able to engraft and reconstitute hematopoiesis in irradiated animals more efficiently than CD34+ cells. Further studies using human HSCs confirmed that cells lacking CD34 are able to engraft and differentiate into multilineage hematopoietic cells in vivo [191193]. These cells can be isolated from the fetal liver, fetal blood, umbilical cord blood, peripheral blood, and bone marrow [191]. Analysis of CD34 cells isolated from the bone marrow and umbilical cord blood revealed that they lack the HLA-DR and CD90 antigens and are rarer than CD34+ cells [191].

Some authors have raised the possibility of CD34 cells being a more primitive precursor than CD34+ [191] based in the fact that CD34+ cells were identified in myeloablated receptors that received CD34 cells [190193]. Dao et al. [194] proposed that cells can interconvert between CD34+ and CD34 based on their observation that CD34+ grafts give rise to CD34 cells, and when these cells were transplanted to secondary recipients, they were able generate CD34+ cells. However, the issue about what kind of cell (CD34+ or CD34) is the most primitive HSC is a long way from being solved. In contrast to the previously mentioned observations, Gao et al. [195] utilized 3 different sources of HSCs and 4 purification methods and observed no engraftment in mice by the CD34 fraction. The same results were observed by Bhatia et al. [196], who reported that no engraftment was achieved even when 2.6×106 CD34 cells were injected into recipient mice [196].

Nevertheless, some interesting findings can possibly help to explain these contradictory results. Nakamura et al. [197] observed that, when cultured with murine stromal cells as feeders, CD34Lin cells turn from nonproliferative to proliferative, generate CD34+ cells, and exhibit colony-forming activity. Additionally, these cells were only able to engraft into nonobese diabetes/severe combined immunodeficiency (NOD/SCID) mice when cultured under these conditions. Interestingly, the range of engraftment was positively related to the level of CD34+ cells. In the reports showing CD34 engraftment in irradiated recipients, the cells or the mice were exposed to human interleukin-3 (IL-3) and granulocyte/macrophage colony-stimulating factor [191193]. Moreover, Gallacher et al. [198] isolated different subfractions of CD34CD38Lin and observed that only CD133+ cells were capable of engrafting in NOD/SCID mice. This fraction represents only 0.2% of the CD34CD38 cells. It is possible that the CD34 fraction contains primitive HSCs, and the negative results for hematopoietic multilineage differentiation in vitro and lack of engraftment in vivo may be due to the low levels of the true primitive CD34 cells and/or lack of pre-stimulation.

Although a consensus has not been established, HSCs are still being commonly obtained based on their expression of CD34. However, the enrichment of HSCs based on this marker results in a heterogeneous pool of cells [199] in which only a small number of cells can be considered SCs [200]. Therefore, other markers must be applied to subdivide this fraction and identify the most primitive HSCs.

CD133

CD133, also known as AC133, is an HSC surface marker that is restricted to the CD34+ fraction. The percentage of CD133+ cells is generally lower than that of CD34+ cells, comprising 20%–60% of the CD34+ cells isolated from the adult bone marrow, umbilical cord blood, peripheral blood, fetal liver, and bone marrow [201]. The CD133+ fraction of the C34+ HSC pool exhibits a high capacity for expansion, is enriched for megakaryocytic progenitor cells, and is able to differentiate into a larger number of erythroid cells [202]. These cells can home to and engraft in the bone marrow of myeloablated mice and sheep, primary and secondary recipients in the last case [201,203].

However, CD133 is not a marker that is exclusive to HSCs. In mice ESCs, CD133 is upregulated in committed and early progenitor cells and has been proposed to be a molecular marker of this stage of differentiation [204]; CD133 is also found in fetal and adult hepatic SCs [205] and in neural SCs [206]. Moreover, CD133 has been proposed to be a common marker for 2 cell fractions, CD34+ and CD34, which may be enriched for the most primitive SCs [199]. The engraftment of the latter fraction in NOD/SCID mice is only observed when they are enriched for CD133.

Thy-1

Thy-1, also known as CD90 or CDw90, is observed in cells lacking or with low levels of expression of Lin, c-kit, CD38, CD45RA, CD71, and HLA-DR [207, 208].

Studies using umbilical cord blood [209], peripheral blood [210], and fetal [207] and adult bone marrow-derived cells have shown that the CD34+Thy-1+ subset is capable of generating long-term cultures and can give raise to multilineage differentiated cells in vitro. This type of cells is also able to engraft in radiation-ablated recipients and generates myeloerythroid and lymphocyte B lineages more effectively than the Thy-1 fraction [207]. However, it has been suggested that both Thy1+ and Thy1 subsets have self-renewal potential and are capable of engrafting in recipient mice and that the unique difference between them is the HSC frequency. The levels of chimerism of these 2 subset fractions are similar under nonlimiting cell transplantation conditions, but a limiting dilution analysis revealed that HSCs are 5 times more abundant in the Thy-1+ fraction [211].

There are also reports demonstrating the generation of Thy-1+ cells from Thy-1 cells [211,212]. However, Majeti et al. [213] were able to establish a cell hierarchy, where Thy-1+CD45RA cells give rise to Thy-1CD45RA cells that are, in turn, upstream of Thy-1CD45+ cells.

Although it is known that the Thy-1+ fraction includes primitive HSCs, the function of this surface protein has not yet been elucidated. A reduction in the number of hematopoietic colonies was observed when an anti-Thy-1 antibody was added to cultures of Thy-1+ cells, suggesting that it may be involved in the development of HSCs [209]. Thy-1 has also been proposed as a cell cycle status indicator based on the observation that only the Thy-1+ fraction enters the S/G2/M-phases when cells are stimulated by cytokines [212].

Kinase insert domain receptor

Kinase insert domain receptor (KDR), also known as vascular endothelial growth factor receptor 2 (VEGFR2) or fetal liver kinase-1 (Flk1) in the mouse, is a less-known HSC marker. In vitro and in vivo studies have revealed that primitive HSCs are enriched in the CD34+KDR+ fraction, whereas the KDR subset is composed mainly of lineage-committed hematopoietic progenitor cells. KDR+ cells are very rare in the CD34+ fraction, comprising 0.1 to 0.5% of the cells [214]. It has also been proposed that the CD34+KDR+ fraction comprises 5% to 6% of hemangioblasts, cells with bi-lineage differentiation potential, that are capable of generating hematopoietic and endothelial cells [215].

Cub domain protein 1

Cub domain protein 1 (CDCP1) is co-expressed with CD34 and CD133 in HSCs and is absent from mature cells. The HSC fraction isolated based on this marker and subsequently injected into the NOD/SCID mouse is able to engraft and generate various mature hematopoietic lineages [216].

HSC-Associated Negative Markers

CD38

This surface molecule is considered a negative marker for HSCs. It is expressed in differentiated erythroid, myeloid, B-lymphoid precursors, and T-lymphoid lineages [199]. The CD34+CD38 fraction consists of a highly primitive set of cells that are able to generate progeny in long-term cell culture [199,217] and can engraft NOD/SCID mice to produce differentiated hematopoietic cells [196]. These cells are quiescent, and the increase in CD38+ cells correlates with the beginning of cycling and differentiation [199].

Even when the CD34 cells are enriched, the fraction that demonstrates a greater engraftment and proliferation capacity is CD38 [191].

HSC-Associated Lineage Markers

Uncommitted HSCs lack characteristic markers that distinguish them from differentiated lineages [196] (Table 8). When HSCs are purified, it is usually by negatively selecting for these markers.

Table 8.

Differentiated Hematopoietic Lineage Markers Absent from the Surface of Hematopoietic Stem Cellsa

Cell type Marker
Lymphocytes T CD2, CD3, CD4, CD5, CD7, CD8.
Lymphocytes B CD10, CD19, CD20, CD24.
Myeloid CD14, CD15, CD16, CD33, CD41.
Erythroid CD71, glycophorin A.
Natural killer CD56.
Granulocyte CD66b.

New HSC-Associated Markers

Many molecular markers have been established for HSCs. However, HSCs exist within a highly heterogeneous pool of cells, which makes it difficult to isolate the most primitive SCs. Therefore, the discovery of new HSC markers will help to obtain a more homogeneous pool of HSCs. Below, we discuss some of the potential new markers for these cells.

CD49f, also known as integrin α6, yielded positive results when applied together with CD34 and Thy-1 to sort HSCs. The CD34+ Thy1+/Thy1CD49f+ fractions showed high levels of chimerism in receptor mice [211].

Complement component 1 q subcomponent receptor 1 (C1qRp), or CD93 and a human homolog of the murine protein AA4, has been proposed to be a marker for the simultaneous isolation of primitive HSCs found in both CD34+ and CD34 fractions. In vitro and in vivo assays have demonstrated that sorting cells using C1qRp leads to the isolation of primitive hematopoietic progenitors. This molecule has also been proposed to be a positive sorter for the HSCs found in the CD34 fraction, as the isolation of these cells is difficult due to the lack of a characteristic positive marker [219].

The isolation of primitive HSCs based on the activity of aldehyde dehydrogenase (ALDH) has also been reported. ALDH consists of a group of enzymes that are involved in the oxidation of aldehydes to carboxylic acids [220]. Cells positive for ALDH are negative for lineage markers but enriched for CD34+ cells and for cells with short- and long-term activities [221,222]. This is a simple and inexpensive method for isolating HSCs; the fluorescent substrate for ALDH can also be combined with other markers [223].

Some additional HSC markers have been established in mice and are interesting for future studies to confirm their status as HSC markers in humans. Among them are ecotropic viral integration site 1 (Evi1), endothelial cell-selective adhesion molecule 1 (Esam1) and Flk2. Evi1 is a transcription factor exclusively expressed in HSCs and is downregulated during their differentiation [214]. Evi1 expression identifies HSCs with long-term repopulating activity, which are considered the most primitive HSCs [223]. Evi1-positive cells exhibit significant self-renewal and differentiation potential in vitro [223]. In studies in vivo, Evi1+ cells showed the potential to engraft in irradiated recipients and to give rise to differentiated lineages, as evidenced by the presence of myeloid, B, and T cells in the peripheral blood of recipients [223]. The same was observed in secondary transplantations, demonstrating the in vivo long-term multilineage repopulating potential of Evi1-positive HSCs, suggesting that Evi1 is needed to maintain long-term HSC activity [223]. Evi1 overexpression blocks the differentiation of HSCs and induces their expansion. Therefore, Evi1 appears to regulate the transition from HSC to a more committed progenitor. In other words, it controls the balance between self-renewal and differentiation [223].

Esam1 is a transmembrane protein that is highly expressed in both human and murine HSCs and is downregulated when these cells become committed to differentiate [224]. Mature hematopoietic cells, with the exception of megakaryocyte progenitors, do not express this marker. Higher levels of chimerism were observed when mouse cells were sorted using an Esam1 antibody [224]. It has been proposed that Esam1 may be a more effective marker and could be a substitute for other markers in the isolation of HSCs [224].

Flk2 has been proposed as a negative marker for HSCs with long-term repopulating capacity. Murine cells isolated from bone marrow and fetal liver that are negative for this marker show a greater capacity to repopulate irradiated recipients. Flk2 is present in short-term HSCs and is upregulated as the cells become more mature [225].

miRNAs Associated with HSCs

It is known that miRNAs also have a role in SC mechanisms [226]. However, little research has focused on the miRNAs related to the most primitive HSCs. Most of the work has focused on miRNAs related to more committed cells or to diseases associated with the hematopoietic system [227]. Below, we summarize the latest reports on miRNAs in HSCs from the adult bone marrow, umbilical cord blood and mobilized peripheral blood (Table 9).

Table 9.

microRNAs Expression Observed in Different Hematopoietic Stem Cells Fractions from Different Tissues

HSC origin Cell fraction miRNAs Reference
Adult bone marrowa CD34+CD133 miR-15a, miR-15b, miR-16, miR-17, miR-19a, miR-19b, miR-20a, miR-20b, miR-21, miR-23a, miR-25, miR-26a, miR-26b, miR-30b, miR-92a, miR-92b, miR-101, miR-106a, miR-126-3p, miR-142sp, miR-142-3p, miR-142-5p, miR-144, miR-181a, miR-191, miR-221, miR-222, miR-223, miR-451, miR-663, miR-638, Let-7a, Let-7c, Let 7f, Let-7g. [228,229]
  CD133+ miR-15a, miR-16, miR-17, miR-19a, miR-19b, miR-20a, miR-20b, miR-21, miR-26a, miR-26b, miR-29c, miR-30b, miR-92a, miR-92b, miR-101, miR-126-3p, miR-142-3p, miR-142-5p, miR-144, miR-221, miR-222, miR-223, miR-451, Let-7a, Let-7f. [229]
  CD34+ miR-9-3, miR-16a, miR-16b, miR-17, miR-20, miR-23a, miR-23b, miR-24-1, miR-24-2, miR-25, miR-26a, miR-26b, miR-27a, miR-29a, miR-29c, miR-30a, miR-30b, miR-30d, miR-32, miR-33, miR-92, miR-93, miR-95, miR-96, miR-100-1/2, miR-102, miR-103, miR-103-2, miR-106, miR-107, miR-122a, miR-123, miR-128a, miR-128b, miR-130a, miR-135-2, miR-146, miR-155, miR-181a, miR-181b, miR-181c, miR-182, miR-183, miR-190, miR-191, miR-193, miR-192-2/3, miR-194, miR-197, miR-198, miR-199a, miR-202, miR-203, miR-204, miR-205, miR-206, miR-212, miR-213, miR-221, miR-222, miR-223, Let-7b, Let-7c, Let-7d. [230]
Umbilical cord blood CD34+ miR-10a, miR-10b, miR-15a, miR-16, miR-17-5p, miR-18a, miR-19a, miR-19b, miR-20a, miR-20b, miR-21, miR-27a, miR-27b, miR-93, miR-99a, miR-99b, miR-100, miR-101, miR-106a, miR-106b, miR-125a, miR-125b, miR-126, miR-129, miR-130a, miR-142-3p, miR-142-5p, miR-144, miR-155, miR-181a, miR-181c, miR-181d, miR-196b, miR-222, miR-451, miR-519d, miR-551b, miR-520h, Let-7e. [231233]
  CD34+CD38 miR-127, miR-365, miR-452, miR-520h, miR-526b*.Predicted miRNAS: miR-100, miR-105, miR-149, miR-209. [231]
Peripheral blood CD34+CD133+b miR-10a, miR-17-5p, miR-18a, miR-19a, miR-19b, miR-20a, miR-20b, miR-34a, miR-93, miR-106a, miR-126, miR-130a, miR-146a, miR-221, miR-363, miR-599. [233]
  CD34+ miR-10a, miR-15a, miR-16a, miR-16b3, miR-17, miR-19a, miR-19b, miR-20, miR-20a, miR-23a, miR-23b, miR-24, miR-24-1, miR-25, miR-26a, miR-26b, miR-27a, miR-30b, miR-30c, miR-30d, miR-30e, miR-92, miR-93, miR-103, miR-103-2, miR-106, miR-107, miR-123, miR-126, miR-130a, miR-140, miR-142, miR-143, miR-146, miR-155, miR-181a, miR-191, miR-193, miR-196b, miR-197, miR-213, miR-221, miR-222, miR-223, miR-320, miR-363, miR-424/322, Let-7a, Let-7b, Let-7c, Let-7d, Let-7f, Let-7g. [230,234]
a

The miRNAs cited here are the listed by the article's authors as the highly expressed in the cells.

b

Common miRNAs found in 2 pools of HSCs, either CD34+ or CD133+.

Asterisk indicates miRNA strands less commonly found associated with Argonauta complex.

The miRNAs differentially expressed in the CD133+ fraction of the cells isolated from bone marrow (miR-146a, miR-146b-sp, miR-99a, miR-10a, miR-125b, miR-551b, miR-125a-sp) are involved in the inhibition of differentiation, apoptosis, and cytoskeletal remodeling. miRNAs expressed in the CD34+CD133 subset (miR-142-3p and miR-425) were observed to have negative effects on cell proliferation [229].

Merkerova et al. [232] detected differences in the miRNAs expressed by HSCs obtained from bone marrow and umbilical cord blood. Bone marrow-derived CD34+ cells differentially express let-7b, miR-1, miR-34a, miR-195, miR-203, miR-214, miR-545, and miR-548d, and umbilical cord blood cells express the set of miRNAs that is clustered on chromosome 19q13: miR-517c, miR-518a, miR-519d, and miR-520 h.

The miRNA miR-155 seems to block myeloid and erythroid differentiation in hHSCs [230]. Moreover, it is thought that miR-17, 24, 146, 155, 128, and 181 may also maintain hematopoietic cells at an early stem-progenitor stage by blocking their differentiation [230].

Conclusions

ESCs are generally identified by a set of surface markers and the expression of 3 transcription factors (Nanog, Oct-4, and Sox-2). However, transcriptome assays have revealed a set of genes that are highly expressed in ESCs and are not found in their differentiated counterparts. The commonly cited markers are Rex-1, Dnmt3b, Lin-28, Tdgf1, FoxD3, Tert, Utf-1, Gal, Cx43, Gdf3, Gtcm1, Terf1, Terf2, Lefty A, and Lefty B. Nevertheless, these putative markers are far less studied than Nanog, Oct-4, and Sox-2. Some lack studies in hESCs, and others have not even been studied in ESCs.

MSCs are usually identified by the expression of CD73, CD90, and CD105 along with the absence of CD34. Although there is variation among the MSCs isolated from many different regions, it is possible to establish a common set of markers for these cells in which the cells are positive for CD13, CD29, CD44, CD49e, CD54, CD71, CD73, CD90, CD105, CD106, CD166, and HLA-ABC expression and negative for CD14, CD31, CD34, CD45, CD62E, CD62 L, CD62P, and HLA-DR expression. Moreover, many articles have reported the expression of ESC markers; SSEA-4 has been proposed as a new marker for isolating MSCs. Nevertheless, there is no consensus about the expression of these molecules, and little is known about their roles in MSCs.

HSCs are primarily isolated based on the expression of CD34, but the pool of cells obtained is composed of cells with many degrees of differentiation. Therefore, the combination of this marker with other surface molecules, such as CD133 and CD90, along with the lack of CD38 and lineage markers provides the most homogeneous pool of SCs. In addition, the CD34 fraction has also been reported as containing true HSCs, but there is no consensus regarding this finding.

Regarding miRNAs, profiles are being established for each type of SC. However, their functions in the status of SCs have not been completely elucidated. Although there has been considerable progress in the study of SC markers, it is still far from being fully understood.

Acknowledgments

This work was supported by research grants from the Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq; grant no. 474117/2010-3), the Programa Institutos Nacionais de Ciência e Tecnologia (INCT de Processos Redox em Biomedicina-REDOXOMA; grant no. 573530/2008-4), Fundação de Amparo a Pesquisa do Rio Grande do Sul FAPERGS (PRONEM grant no. 11/2072-2), and CAPES (Cordenação de Aperfeiçoamento de Pessoal do Ensino Superior).

Author Disclosure Statement

No competing financial interests exist.

References

  • 1.Reya T. Morrison SJ. Clarke MF. Weissman IL. Stem cells, cancer, and cancer stem cells. Nature. 2001;414:105–111. doi: 10.1038/35102167. [DOI] [PubMed] [Google Scholar]
  • 2.Verfaillie CM. Pera MF. Lansdorp PM. Stem cells: hype and reality. Hematology. 2002;2002:369–391. doi: 10.1182/asheducation-2002.1.369. [DOI] [PubMed] [Google Scholar]
  • 3.Bongso A. Richards M. History and perspective of stem cell research. Best Pract Res Clin Obstet Gynaecol. 2004;18:827–842. doi: 10.1016/j.bpobgyn.2004.09.002. [DOI] [PubMed] [Google Scholar]
  • 4.Rathjen PDA. Lake J. Whyatt LM. Bettess MD. Rathjen J. Properties and uses of embryonic stem cells: prospects for application to human biology and gene therapy. Reprod Fertil Dev. 1998;10:31–47. doi: 10.1071/r98041. [DOI] [PubMed] [Google Scholar]
  • 5.Lerou PH. Daley GQ. Therapeutic potential of embryonic stem cells. Blood Rev. 2005;19:321–331. doi: 10.1016/j.blre.2005.01.005. [DOI] [PubMed] [Google Scholar]
  • 6.Wobus M. Boheler KR. Embryonic stem cells: prospects for developmental biology and cell therapy. Physiol Rev. 2005;85:635–678. doi: 10.1152/physrev.00054.2003. [DOI] [PubMed] [Google Scholar]
  • 7.Pan GJ. Chang ZY. Scher HR. Pei D. Stem cell pluripotency and transcription factor Oct4. Cell Res. 2002;12:321–329. doi: 10.1038/sj.cr.7290134. [DOI] [PubMed] [Google Scholar]
  • 8.Evans MJ. Kaufman MH. Establishment in culture of pluripotential cells from mouse embryos. Nature. 1981;292:154–156. doi: 10.1038/292154a0. [DOI] [PubMed] [Google Scholar]
  • 9.Thompson JA. Itskovitz-Eldor J. Shapiro SS. Waknitz MA. Swiergiel JJ. Jones JM. Marshall VS. Embryonic stem cell lines derived from human blastocysts. Science. 1998;282:1145–1147. doi: 10.1126/science.282.5391.1145. [DOI] [PubMed] [Google Scholar]
  • 10.Adewumi O. Aflatoonia B. Ahrlund-Richte L, et al. Characterization of human embryonic stem cell lines by the International Stem Cell Initiative. Nat Biotechnol. 2007;25:803–816. doi: 10.1038/nbt1318. [DOI] [PubMed] [Google Scholar]
  • 11.Assou S. Carrour T. Tondeur S. Ström S. Gabelle A. Marty S. Nadal L. Pantesco V. Réme T, et al. A meta-analysis of human embryonic stem cells transcriptome integrated into a web-based expression atlas. Stem Cells. 2007;25:961–973. doi: 10.1634/stemcells.2006-0352. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Hosler BA. Larosa GJ. Grippo JF. Gudas LJ. Expression of REX-I, a gene containing zinc finger motifs, is rapidly reduced by retinoic acid in F9 teratocarcinoma cells. Mol Cell Biol. 1989;9:5623–5629. doi: 10.1128/mcb.9.12.5623. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Kannagi R. Cochran NA. Ishigami F. Hakimori S. Andrews PW. Knowles BB. Solterl D. Stage-specific embryonic antigens (SSEA-3 and -4) are epitopes of a unique globo-series ganglioside isolated from human teratocarcinoma cells. EMBO J. 1983;2:2355–2361. doi: 10.1002/j.1460-2075.1983.tb01746.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Badcock G. Pigott C. Goepel J. Andrews PW. The human embryonal carcinoma marker antigen TRA-1-60 is a sialylated keratan sulfate proteoglycan. Cancer Res. 1999;59:4715–4719. [PubMed] [Google Scholar]
  • 15.Reubinoff BE. Pera MF. Fong CY. Trounson A. Bongso A. Embryonic stem cell lines from human blastocysts: somatic differentiation in vitro. Nat Biotechnol. 2000;18:399–404. doi: 10.1038/74447. [DOI] [PubMed] [Google Scholar]
  • 16.Henderson JK. Draper JS. Baillie HS. Fishel S. Thomson JA. Moore H. Andrews PH. Preimplantation human embryos and embryonic stem cells show comparable expression of stage-specific embryonic antigens. Stem Cells. 2002;20:329–333. doi: 10.1634/stemcells.20-4-329. [DOI] [PubMed] [Google Scholar]
  • 17.Draper JS. Pigott C. Thomson JA. Andrews PW. Surface antigens of human embryonic stem cells: changes upon differentiation in culture. J Anat. 2002;200:249–258. doi: 10.1046/j.1469-7580.2002.00030.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Abeyta MJ. Clark AT. Rodriguez RT. Bodnar MS. Pera RAR. Firpo MT. Unique gene expression signatures of independently derived human embryonic stem cell lines. Hum Mol Genet. 2004;13:601–608. doi: 10.1093/hmg/ddh068. [DOI] [PubMed] [Google Scholar]
  • 19.Skottman H. Mikkola M. Lundin K. Olsson C. Strömberg A. Tuuri T. Otonkoski T. Hovatta O. Lahesmaa R. Gene expression signatures of seven individual human embryonic stem cell lines. Stem Cells. 2005;23:1343–1356. doi: 10.1634/stemcells.2004-0341. [DOI] [PubMed] [Google Scholar]
  • 20.Clark AT. Rodriguez RT. Bodnar MS. Abeyta MJ. Cedars MI. Turek PJ. Firpo MT. Pera RRA. Human STELLAR, NANOG, and GDF3 genes are expressed in pluripotent cells and map to chromosome 12p13, a hotspot for teratocarcinoma. Stem Cells. 2004;22:169–179. doi: 10.1634/stemcells.22-2-169. [DOI] [PubMed] [Google Scholar]
  • 21.Brandenberger R. Khrebtukova I. Thies RS. Miura T. Jingli C. Puri R. Vasicek T. Lebkowski J. Rao M. MPSS profiling of human embryonic stem cells. BMC Dev Biol. 2004;4:10. doi: 10.1186/1471-213X-4-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Ginis I. Luo Y. Miura T. Thies S. Brandenberger R. Gerecht-Nir S. Amit M. Hoke A. Carpenter MK. Itskovitz-Eldor J. Rao MS. Differences between human and mouse embryonic stem cells. Dev Biol. 2004;269:360–380. doi: 10.1016/j.ydbio.2003.12.034. [DOI] [PubMed] [Google Scholar]
  • 23.Miura TK. Luo Y. Khrebtukova I. Brandenberger R. Zhou D. Thies RS. Vasicek T. Young H. Lebkowski J. Carpenter MK. Rao MS. Monitoring early differentiation events in human embryonicstem cells by massively parallel signature sequencing and expressed sequence tag scan. Stem Cells Dev. 2004;13:694–715. doi: 10.1089/scd.2004.13.694. [DOI] [PubMed] [Google Scholar]
  • 24.Rao RR. Stice SL. Gene expression profiling of embryonic stem cells leads to greater understanding of pluripotency and early developmental events. Biol Reprod. 2004;71:1772–1778. doi: 10.1095/biolreprod.104.030395. [DOI] [PubMed] [Google Scholar]
  • 25.Richards M. Tan S. Tan J. Chan W. Bongso A. The transcriptome profile of human embryonic stem cells as defined by SAGE. Stem Cells. 2004;22:51–64. doi: 10.1634/stemcells.22-1-51. [DOI] [PubMed] [Google Scholar]
  • 26.Bhattacharya B. Cai J. Lu Y. Miura T. Mejido J. Brimble SN. Zeng X. Schulz TC. Rao MS. Puri RK. Comparison of the gene expression profile of undifferentiated human embryonic stem cell lines and differentiating embryoid bodies. BMC Dev Biol. 2005;5:22. doi: 10.1186/1471-213X-5-22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Chambers I. Colby D. Robertson M. Functional expression cloning of Nanog, a pluripotency sustaining factor in embryonic stem cells. Cell. 2003;113:643–655. doi: 10.1016/s0092-8674(03)00392-1. [DOI] [PubMed] [Google Scholar]
  • 28.Mitsui K. Tokuzawa Y. Itoh H. Segawa K. Murakami M. Takahashi K. Maruyama M. Maeda M. Yamanaka S. The homeoprotein Nanog is required for maintenance of pluripotency in mouse epiblast and ES Cells. Cell. 2003;113:631–642. doi: 10.1016/s0092-8674(03)00393-3. [DOI] [PubMed] [Google Scholar]
  • 29.Hyslop L. Stojkovic M. Armstrong L. Walter T. Stojkovic P. Przyborski S. Herbert M. Murdoch A. Strachan T. Lako M. Downregulation of NANOG induces differentiation of human embryonic stem cells to extraembryonic lineages. Stem Cells. 2005;23:1035–1043. doi: 10.1634/stemcells.2005-0080. [DOI] [PubMed] [Google Scholar]
  • 30.Romero-Lanman EE. Pavlovic S. Amlani B. Chin Y. Benezra R. Id1 maintains embryonic stem cell self-renewal by up-regulation of Nanog and repression of brachyury expression. Stem Cells Dev. 2012;21:384–393. doi: 10.1089/scd.2011.0428. [DOI] [PubMed] [Google Scholar]
  • 31.Benezra R. Davis RL. Lokshon D. Turner DL. Weintraub H. The Protein Id: a negative regulator of helix-loop-helix DNA binding proteins. Cell. 1990;61:49–59. doi: 10.1016/0092-8674(90)90214-y. [DOI] [PubMed] [Google Scholar]
  • 32.Cauffman G. Van de Velde H. Liebaers I. Van Steirteghem A. Oct-4 mRNA and protein expression during human preimplantation development. Molecular Hum Reprod. 2005;11:173–181. doi: 10.1093/molehr/gah155. [DOI] [PubMed] [Google Scholar]
  • 33.Niwa H. Miyazaki J. Smith AG. Quantitative expression of Oct-3/4 defines differentiation, dedifferentiation or self-renewal of ES cells. Nat Genet. 2000;24:372–376. doi: 10.1038/74199. [DOI] [PubMed] [Google Scholar]
  • 34.Martin MM. Walsh JR. Gokhale PJ. Draper JS. Bahrami AR. Morton I. Moore HD. Andrews PW. Specific knockdown of Oct4 and β2-microglobulin expression by RNA interference in human embryonic stem cells and embryonic carcinoma cells. Stem Cells. 2004;22:659–668. doi: 10.1634/stemcells.22-5-659. [DOI] [PubMed] [Google Scholar]
  • 35.Kamachi Y. Uchikawa M. Kondoh H. Pairing SOX off with partners in the regulation of embryonic. Trends Genet. 2000;16:182–187. doi: 10.1016/s0168-9525(99)01955-1. [DOI] [PubMed] [Google Scholar]
  • 36.Kopp JL. Ormsbee BD. Desler M. Rizzino A. Small increases in the level of Sox 2 trigger the differentiation of mouse embryonic stem cells. Stem Cells. 2008;26:903–911. doi: 10.1634/stemcells.2007-0951. [DOI] [PubMed] [Google Scholar]
  • 37.Avilion AA. Nicolis SK. Pevny LH. Perez L. Vivian N. Lovell-Badge R. Multipotent cell lineages in early mouse development depend on SOX2 function. Genes Dev. 2003;17:126–140. doi: 10.1101/gad.224503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Fong H. Hohenstein KA. Donovan PJ. Regulation of self-renewal and pluripotency by Sox2 in human embryonic stem cells. Stem Cells. 2008;26:1931–1938. doi: 10.1634/stemcells.2007-1002. [DOI] [PubMed] [Google Scholar]
  • 39.Chew J. Loh Y. Zhang W. Chen X. Tam W. Yeap L. Li P. Ang Y. Lim B. Robson P. Ng H. Reciprocal transcriptional regulation of Pou5f1 and Sox2 via the Oct4/Sox2 complex in embryonic stem cells. Mol Cell Biol. 2005;25:6031–6046. doi: 10.1128/MCB.25.14.6031-6046.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Rodda DJ. Chew J. Lim L. Loh Y. Wang B. Ng H. Robson P. Transcriptional regulation of Nanog by OCT4 and SOX2. J Biol Chem. 2005;280:24731–24737. doi: 10.1074/jbc.M502573200. [DOI] [PubMed] [Google Scholar]
  • 41.Loh Y. Wu Q. Chew J. Vega VB. Zhang W. Chen X. Bourque G. George J. Leong B, et al. The Oct4 and Nanog transcription network regulates pluripotency in mouse embryonic stem cells. Nat Genet. 2006;38:431–440. doi: 10.1038/ng1760. [DOI] [PubMed] [Google Scholar]
  • 42.Mongan NP. Martin KM. Gudas LJ. The putative human stem cell marker, Rex-1 (Zfp42): structural classification and expression in normal human epithelial and carcinoma cell cultures. Mol Carcinog. 2006;45:887–900. doi: 10.1002/mc.20186. [DOI] [PubMed] [Google Scholar]
  • 43.Scotland KB. Chen S. Sylvester R. Gudas LJ. Analysis of Rex1 (Zfp42) function in embryonic stem cell differentiation. Dev Dyn. 2009;238:1863–1877. doi: 10.1002/dvdy.22037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Shi W. Wang H. Pan G. Geng Y. Guo Y. Pei D. Regulation of the pluripotency marker Rex-1 by Nanog and Sox2. J Biol Chem. 2006;281:23319–23325. doi: 10.1074/jbc.M601811200. [DOI] [PubMed] [Google Scholar]
  • 45.Ben-Shushan E. Thompson JR. Gudas LJ. Bergman Y. Rex-1, a gene encoding a transcription factor expressed in the early embryo, is regulated via Oct-3/4 and Oct 6 binding to an octamer site and an novel protein, Rox-1, binding to an adjacent site. Mol Cell Biol. 1998;18:1866–1878. doi: 10.1128/mcb.18.4.1866. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Watanabe D. Suetakeb I. Tadac T. Tajimab S. Stage and cell-specific expression of Dnmt3a and Dnmt3b during embryogenesis. Mech Dev. 2002;118:187–190. doi: 10.1016/s0925-4773(02)00242-3. [DOI] [PubMed] [Google Scholar]
  • 47.Huntriss J. Hinkins M. Oliver B. Harris SE. Beazley JC. Rutherford AJ. Gosden RG. Lanzendorf SE. Picton HM. Expression of mRNAs for DNA methyltransferases and methyl-CpG-binding proteins in the human female germ line, pre-implantation embryos, and embryonic stem cells. Mol Reprod Dev. 2004;67:323–336. doi: 10.1002/mrd.20030. [DOI] [PubMed] [Google Scholar]
  • 48.Li J. Pu M. Hirasawa R. Li B. Huang Y. Zeng R. Jing N. Chen T. Li E. Sasaki H. Xu G. Synergistic function of DNA methyltransferases Dnmt3a and Dnmt3b in the methylation of Oct4 and Nanog. Mol Cell Biol. 2007;27:8748–8759. doi: 10.1128/MCB.01380-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Jackson M. Krassowska A. Gilbert N. Chevassut T. Forrester L. Ansell J. Ramsahoye B. Severe global DNA hypomethylation blocks differentiation and induces histone hyperacetylation in embryonic stem cells. Mol Cell Biol. 2004;24:8862–8871. doi: 10.1128/MCB.24.20.8862-8871.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Tsumura A. Hayakawa T. Kumaki Y. Takebayashi S. Sakaue M. Matsuoka C. Shimotohno K. Ishikawa F. Li E, et al. Maintenance of self-renewal ability of mouse embryonic stem cells in the absence of DNA methyltransferases Dnmt1, Dnmt3a and Dnmt3b. Genes Cells. 2006;11:805–814. doi: 10.1111/j.1365-2443.2006.00984.x. [DOI] [PubMed] [Google Scholar]
  • 51.Sutton J. Costa R. Klugi M. Fieldi L. Xu D. Largaespada DA. Fletcher CF. Jenkins NA. Copeland NG. Klemsz M. Hromas R. Genesis, a winged helix transcriptional repressor with expression restricted to embryonic stem cells. J Biol Chem. 1996;271:23126–23133. doi: 10.1074/jbc.271.38.23126. [DOI] [PubMed] [Google Scholar]
  • 52.Hanna LA. Foreman RK. Tarasenko IA. Kessler DS. Labosky PA. Requirement for Foxd3 in maintaining pluripotent cells of the early mouse embryo. Genes Dev. 2002;16:2650–2661. doi: 10.1101/gad.1020502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Hromas R. Ye H. Spinella M. Dmitrovsky E. Xu D. Costa RH. Genesis, a winged helix transcriptional repressor, has embryonic expression limited to the neural crest, and stimulates proliferation in vitro in a neural development model. Cell Tissue Res. 1999;297:371–382. doi: 10.1007/s004410051365. [DOI] [PubMed] [Google Scholar]
  • 54.Liu Y. Labosky PA. Regulation of embryonic stem cell self-renewal and pluripotency by Foxd3. Stem Cells. 2008;26:2475–2484. doi: 10.1634/stemcells.2008-0269. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Guo Y. Costa R. Ramsey H. Starnes T. Vance G. Robertson K. Kelley M. Reinbold R. Scholer H. Hromas R. The embryonic stem cell transcription factors Oct-4 and FoxD3 interact to regulate endodermal specific promoter expression. Proc Natl Acad Sci U S A. 2002;99:3663–3667. doi: 10.1073/pnas.062041099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Pan G. Li J. Zhou Y. Zheng H. Pei D. A negative feedback loop of transcription factors that controls stem cell pluripotency and self-renewal. FASEB J. 2006;20:1730–1732. doi: 10.1096/fj.05-5543fje. [DOI] [PubMed] [Google Scholar]
  • 57.Brandt R. Normanno N. Gullick WJ. Lin J. Harkins R. Schneider D. Jones B. Ciardiello F. Persico MG, et al. Identification and biological characterization of an epidermal growth factor-related protein: Cripto-l. J Biol Chem. 1994;269:17320–17328. [PubMed] [Google Scholar]
  • 58.Johnson SE. Rothstein JL. Knowless BB. Expression of epidermal growth factor family gene members in early mouse development. Dev Dyn. 1994;201:216–226. doi: 10.1002/aja.1002010305. [DOI] [PubMed] [Google Scholar]
  • 59.Parisi S. D'Andrea D. Lago CT. Adamson ED. Persico MG. Minchiotti G. Nodal-dependent Cripto signaling promotes cardiomyogenesis and redirects the neural fate of embryonic stem cells. J Cell Biol. 2003;163:303–314. doi: 10.1083/jcb.200303010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Parish CL. Parisi S. Persico MG. Arenas E. Minchiottib G. Cripto as a target for improving embryonic stem cell-based therapy in Parkinson's disease. Stem Cells. 2005;23:471–476. doi: 10.1634/stemcells.2004-0294. [DOI] [PubMed] [Google Scholar]
  • 61.Xu C. Liguori G. Persico MG. Adamson ED. Abrogation of the Cripto gene in mouse leads to failure of postgastrulation morphogenesis and lack of differentiation of cardiomyocytes. Development. 1999;126:483–494. doi: 10.1242/dev.126.3.483. [DOI] [PubMed] [Google Scholar]
  • 62.Darr H. Benvenisty N. Genetic analysis of the role of the reprogramming gene LIN-28 in human embryonic stem cells. Stem Cells. 2009;27:352–362. doi: 10.1634/stemcells.2008-0720. [DOI] [PubMed] [Google Scholar]
  • 63.Peng L. Chen S. Lei X. Yang L. Lin H. Carmichael GG. Huang Y. Genome-wide studies reveal that Lin28 enhances the translation of genes important for growth and survival of human embryonic stem cells. Stem Cells. 2011;29:496–504. doi: 10.1002/stem.591. [DOI] [PubMed] [Google Scholar]
  • 64.Xu B. Zhang K. Huang Y. Lin28 modulates cell growth and associates with a subset of cell cycle regulator mRNAs in mouse embryonic stem cells. RNA. 2009;15:357–361. doi: 10.1261/rna.1368009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Viswanathan SR. Daley GQ. Gregory RI. Selective blockade of microRNA processing by Lin28. Science. 2008;320:97–100. doi: 10.1126/science.1154040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Qiu C. Ma Y. Wang J. Peng S. Huang Y. Lin28-mediated post-transcriptional regulation of Oct4 expression in human embryonic stem cells. Nucleic Acids Res. 2010;38:1240–1248. doi: 10.1093/nar/gkp1071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.van den Boom V. Kooistra SM. Boesjes M. Geverts B. Houtsmuller AB. Monzen K. Komuro I. Essers J. Drenth-Diephuis LJ. Eggen BJL. UTF1 is a chromatin associated protein involved in ES cell differentiation. J Cell Biol. 2007;178:913–924. doi: 10.1083/jcb.200702058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Kooistra SM. Thummer RP. Eggen BJL. Characterization of human UTF1, a chromatin-associated protein with repressor activity expressed in pluripotent cells. Stem Cell Res. 2009;2:211–218. doi: 10.1016/j.scr.2009.02.001. [DOI] [PubMed] [Google Scholar]
  • 69.Okuda A. Fukushima A. Nishimoto M. Orimo A. Yamagishi T. Nabeshima Y. Kuro-o M. Nabeshima Y. Boon K, et al. UTF1, a novel transcriptional coactivator expressed in pluripotent embryonic stem cells and extra-embryonic cells. EMBO J. 1998;17:2019–2032. doi: 10.1093/emboj/17.7.2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Nishimoto M. Fukushima A. Okuda A. Muramats M. The gene for the embryonic stem cell coactivator UTF1 carries a regulatory element which selectively interacts with a complex composed of Oct-3/4 and Sox-2. Mol Cell Biol. 1999;19:5453–5465. doi: 10.1128/mcb.19.8.5453. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Nishimoto M. Miyagi S. Yamagishi T. Sakaguchi T. Niwa H. Muramatsu M. Okuda A. Oct-3/4 maintains the proliferative embryonic stem cell state via specific binding to a variant octamer sequence in the regulatory region of the UTF1 locus. Mol Cell Biol. 2005;25:5084–5094. doi: 10.1128/MCB.25.12.5084-5094.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Choi HS. Kim W. Kim H. Kim J. Ko J. Lee S. Jang YJ. Kim SJ. Lee M, et al. Identification and characterization of adenovirus early region 1B-associated protein 5 as a surface marker on undifferentiated human embryonic stem cells. Stem Cells Dev. 2011;20:609–620. doi: 10.1089/scd.2010.0265. [DOI] [PubMed] [Google Scholar]
  • 73.Sundberg M. Janson L. Ketolainen J. Pihlajamäki H. Suuronen R. Skottman H. Inzunza J. Hovatta O. Narkilahti S. CD marker expression profiles of human embryonic stem cells and their neural derivatives, determined using flow-cytometric analysis, reveal a novel CD marker for exclusion of pluripotent stem cells. Stem Cell Res. 2009;2:113–124. doi: 10.1016/j.scr.2008.08.001. [DOI] [PubMed] [Google Scholar]
  • 74.Tang C. Lee AS. Volkmer JP. Sahoo D. Nag D. Mosley AR. Inlay MA. Ardehali R. Chavez SL, et al. An antibody against SSEA-5 glycan on human pluripotent stem cells enables removal of teratoma-forming cells. Nat Biotechnol. 2011;29:829–835. doi: 10.1038/nbt.1947. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Naujok O. Lenzen S. A critical re-evaluation of CD24-positivity of human embryonic stem cells differentiated into pancreatic progenitors. Stem Cell Rev. 2012;8:779–791. doi: 10.1007/s12015-012-9362-y. [DOI] [PubMed] [Google Scholar]
  • 76.Mateizel I. Spits C. Verloes A. Mertzanidou A. Liebaers I. Sermon K. Characterization of CD30 expression in human embryonic stem cell lines cultured in serum-free media and passaged mechanically. Hum Reprod. 2009;24:2477–2489. doi: 10.1093/humrep/dep234. [DOI] [PubMed] [Google Scholar]
  • 77.Dormeyer W. van Hoof D. Braam SR. Heck AJR. Mummery CL. Krijgsveld J. Plasma membrane proteomics of human embryonic stem cells and human embryonal carcinoma cells. J Proteome Res. 2008;7:2936–2951. doi: 10.1021/pr800056j. [DOI] [PubMed] [Google Scholar]
  • 78.Wong RC. Ibrahim A. Fong H. Thompson N. Lock LF. Donovan PJ. L1TD1 is a marker for undifferentiated human embryonic stem cells. PLoS One. 2011;6:e19355. doi: 10.1371/journal.pone.0019355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Zhang X. Yalcin S. Lee D. Yeh TJ. Lee S. Su J. Mungamuri SK. Rimmelé P. Kennedy M, et al. FOXO1 is an essential regulator of pluripotency in human embryonic stem cells. Nat Cell Biol. 2011;13:1092–1099. doi: 10.1038/ncb2293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Bhattacharya B. Miura T. Brandenberger R. Mejido J. Luo Y. Yang AX. Joshi BH. Ginis I. Thies RA, et al. Gene expression in human embryonic stem cell lines: unique molecular signature. Blood. 2004;103:2956–2964. doi: 10.1182/blood-2003-09-3314. [DOI] [PubMed] [Google Scholar]
  • 81.Sato N. Sanjuan IM. Heke M. Uchida M. Naef F. Brivanlou AH. Molecular signature of human embryonic stem cells and its comparison with the mouse. Dev Biol. 2003;260:404–413. doi: 10.1016/s0012-1606(03)00256-2. [DOI] [PubMed] [Google Scholar]
  • 82.Moon SH. Kim SW. Kim JS. Park SJ. Do JT. Lee DR. Chung HM. Gene expression profiles in CHA3 and CHA4 human embryonic stem cells and embryoid bodies. Mol Cells. 2011;31:315–326. doi: 10.1007/s10059-011-0039-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Cai J. Chen J. Liu Y. Miura T. Luo Y. Loring JF. Freed WJ. Rao MS. Zeng X. Assessing self-renewal and differentiation in human embryonic stem cell lines. Stem Cells. 2006;24:516–530. doi: 10.1634/stemcells.2005-0143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Murchison EP. Partridge JF. Tam OH. Cheloufi S. Hannon GJ. Characterization of Dicer-deficient murine embryonic stem cells. Proc Natl Acad Sci U S A. 2005;102:12135–12140. doi: 10.1073/pnas.0505479102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Denli AM. Tops BBJ. Plasterk RHA. Ketting RF. Hannon GJ. Processing of primary microRNAs by the microprocessor complex. Nature. 2004;432:231–235. doi: 10.1038/nature03049. [DOI] [PubMed] [Google Scholar]
  • 86.Wang Y. Medvid R. Melton C. Jaenisch R. Blelloch R. DGCR8 is essential for microRNA biogenesis and silencing of embryonic stem cell self-renewal. Nat Genet. 2007;39:380–385. doi: 10.1038/ng1969. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Barroso-del Jesus AB. Lucena-Aguilar G. Menendez P. The miR-302-367 cluster as a potential stemness regulator in ESCs. Cell Cycle. 2009;8:394–398. doi: 10.4161/cc.8.3.7554. [DOI] [PubMed] [Google Scholar]
  • 88.Barroso-delJesus AB. Romero-López C. Lucena-Aguilar G. Melen GJ. Sanchez L. Ligero G. Berzal-Herranz A. Menendez P. Promoter functional characterization of its core cluster: human gene structure and embryonic stem cell-specific miR302-367. Mol Cell Biol. 2008;28:6609–6619. doi: 10.1128/MCB.00398-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Ren J. Jin P. Wang E. Marincola FM. Stroncek DF. MicroRNA and gene expression patterns in the differentiation of human embryonic stem cells. J Transl Med. 2009;7:20. doi: 10.1186/1479-5876-7-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Wilson KD. Venkatasubrahmanyam S. Jia F. Sun N. Butte AJ. Wu JC. MicroRNA profiling of human-induced pluripotent stem cells. Stem Cells Dev. 2009;18:749–758. doi: 10.1089/scd.2008.0247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Card DAG. Hebbar PB. Li L. Trotter KW. Komatsu Y. Mishina Y. Archer TK. Oct4/Sox2-regulated miR-302 targets cyclin D1 in human embryonic stem cells. Mol Cell Biol. 2008;20:6426–6438. doi: 10.1128/MCB.00359-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Hu S. Wilson KD. Ghosh Z. Han L. Wang Y. Lan F. Ransohoff KJ. Wu JC. MicroRNA-302 increases reprogramming efficiency via repression of NR2F2. Stem Cells. 2012 doi: 10.1002/stem.1278. [Epub ahead of print] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Liao B. Bao X. Liu L. Feng S. Zovoilis A. Liu W. Xue Y. Cai J. Guo X, et al. MicroRNA cluster 302-367 enhances somatic cell reprogramming by accelerating a mesenchymal-to-epithelial transition. J Biol Chem. 2011;286:17359–17364. doi: 10.1074/jbc.C111.235960. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Subramanyam D. Lamouille S. Judson RL. Liu JY. Bucay N. Derynck R. Blelloch R. Multiple targets of miR-302 and miR-372 promote reprogramming of human fibroblasts to induced pluripotent stem cells. Nat Biotechnol. 2011;29:443–449. doi: 10.1038/nbt.1862. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Miyoshi N. Ishii H. Nagano H. Haraguchi N. Dewi DL. Kano Y. Nishikawa S. Tanemura M. Mimori K, et al. Reprogramming of mouse and human cells to pluripotency using mature microRNAs. Cell Stem Cell. 2011;8:633–638. doi: 10.1016/j.stem.2011.05.001. [DOI] [PubMed] [Google Scholar]
  • 96.Suh M. Lee Y. Kim JY. Kim S. Moon S. Lee JY. Cha K. Chung HM. Yoon HS, et al. Human embryonic stem cells express a unique set of microRNAs. Dev Biol. 2004;270:488–498. doi: 10.1016/j.ydbio.2004.02.019. [DOI] [PubMed] [Google Scholar]
  • 97.Yoo JK. Kim J. Choi S. Noh HM. Kwon YD. Yoo H. Yi HS. Chung HM. Kim JK. Discovery and characterization of novel miRNAs during endothelial differentiation of human embryonic stem cells. Stem Cells Dev. 2011;20:2049–2057. doi: 10.1089/scd.2011.0500. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Morin RD. O'Connor MD. Griffith M. Kuchenbauer F. Delaney A. Prabhu A. Zhao Y. McDonald H. Zeng T, et al. Application of massively parallel sequencing to microRNA profiling and discovery in human embryonic stem cells. Genome Res. 2008;18:610–621. doi: 10.1101/gr.7179508. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Wang Y. Baskerville S. Shenoy A. Babiarz JE. Baehner L. Blelloch R. Embryonic stem cell-specific microRNAs regulate the G1-S transition and promote rapid proliferation. Nat Genet. 2008;40:1478–1483. doi: 10.1038/ng.250. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Li SSL. Yu SL. Kao LP. Tsai ZY. Singh S. Chen BZ. Ho BC. Liu YH. Yang PC. Target identification of microRNAs expressed highly in human embryonic stem cells. J Cell Biochem. 2009;106:1020–1030. doi: 10.1002/jcb.22084. [DOI] [PubMed] [Google Scholar]
  • 101.Laurent LC. Chen J. Ulitsky I. Mueller FJ. Lu C. Shamir R. Fan JB. Loring JF. Comprehensive microRNA profiling reveals a unique human embryonic stem cell signature dominated by a single seed sequence. Stem Cells. 2008;26:1506–1516. doi: 10.1634/stemcells.2007-1081. [DOI] [PubMed] [Google Scholar]
  • 102.Lakshmipathy U. Love B. Goff LA. Jörnsten R. Graichen R. Hart RP. Chesnut JD. MicroRNA expression pattern of undifferentiated and differentiated human embryonic stem cells. Stem Cells Dev. 2007;16:1003–1016. doi: 10.1089/scd.2007.0026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Bar M. Wyman SK. Fritz BR. QI J. Garg KS. Parkin RK. Kroh EM. Bendoraite A. Mitchell PS, et al. MicroRNA discovery and profiling in human embryonic stem cells by deep sequencing of small RNA libraries. Stem Cells. 2005;26:2496–2505. doi: 10.1634/stemcells.2008-0356. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Pittenger MF. Mackay AM. Beck SC. Jaiswal RK. Douglas R. Mosca JD. Moorman MA. Simonetti DW. Craig S. Marshak DR. Multilineage potential of adult human mesenchymal stem cells. Science. 1999;284:143–147. doi: 10.1126/science.284.5411.143. [DOI] [PubMed] [Google Scholar]
  • 105.Santos TM. Percegona LS. González P. Calil A. Perini CC. Faucz FR. Câmara NOS. Aita CAM. Expression of pancreatic endocrine markers by mesenchymal stem cells from human umbilical cord vein. Transplant Proc. 2010;42:563–565. doi: 10.1016/j.transproceed.2010.01.032. [DOI] [PubMed] [Google Scholar]
  • 106.Lee KD. Kuo TKC. Whang-Peng J. Chung YF. Lin CT. Chow SH. Chen JR. Chen YP. Lee OKS. In vitro hepatic differentiation of human mesenchymal stem cells. Hepatology. 2004;40:1275–1284. doi: 10.1002/hep.20469. [DOI] [PubMed] [Google Scholar]
  • 107.Pournasr B. Mohamadnejad M. Bagheri M. Aghdami N. Shahsavani M. Malekzadeh R. Baharvand H. In vitro differentiation of human bone marrow mesenchymal stem cells into hepatocyte-like cells. Arch Iran Med. 2011;14:244–249. [PubMed] [Google Scholar]
  • 108.Dezawa M. Kanno H. Hoshino M. Cho H. Matsumoto N. Itokazu Y. Tajima N. Yamada H. Sawada H, et al. Specific induction of neuronal cells from bone marrow stromal cells and application for autologous transplantation. J Clin Invest. 2004;113:1701–1710. doi: 10.1172/JCI20935. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Afannsyev BV. Elstner EE. Zander AR. A. J. Friedenstein, founder of the mesenchymal stem cell concept. Cell Ther Transplant. 2009;1:35–37. [Google Scholar]
  • 110.Zuk PA. Zhu M. Ashjian P. De Ugarte DA. Huang JI. Mizuno H. Alfonso ZC. Fraser JK. Benhaim P. Hedrick MH. Human adipose tissue is a source of multipotent stem cells. Mol Biol Cell. 2002;13:4279–4295. doi: 10.1091/mbc.E02-02-0105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Pierantozzi E. Gava B. Manini I. Roviello F. Marotta G. Chiavarelli M. Sorrentino V. Pluripotency regulators in human mesenchymal stem cells: expression of NANOG but not of OCT-4 and SOX-2. Stem Cells Dev. 2011;20:915–923. doi: 10.1089/scd.2010.0353. [DOI] [PubMed] [Google Scholar]
  • 112.Nöth U. Osyczka AM. Tuli R. Hickok NJ. Danielson KG. Tuan RS. Multilineage mesenchymal differentiation potential of human trabecular bone-derived cells. J Orthop Res. 2002;20:1060–1069. doi: 10.1016/S0736-0266(02)00018-9. [DOI] [PubMed] [Google Scholar]
  • 113.Pasquinelli G. Pacilli A. Alviano F. Foroni L. Ricci F. Valente S. Orrico C. Lanzoni G. Buzzi M, et al. Multidistrict human mesenchymal vascular cells: pluripotency and stemness characteristics. Cytotherapy. 2010;12:275–287. doi: 10.3109/14653241003596679. [DOI] [PubMed] [Google Scholar]
  • 114.Zvaifler NJ. Marinova-Matafchieva L. Adams G. Edwards CJ. Moss J. Burger JA. Maini RN. Mesenchymal precursor cells in the blood of normal individuals. Arthritis Res. 2000;2:477–488. doi: 10.1186/ar130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Riekstina U. Cakstina I. Parfejevs V. Hoogduijn M. Jankoyskis G. Muiznieks I. Muceniece R. Ancans J. Embryonic stem cell marker expression pattern in human mesenchymal stem cells derived from bone marrow, adipose tissue, heart and dermis. Stem Cell Rev. 2009;5:378–386. doi: 10.1007/s12015-009-9094-9. [DOI] [PubMed] [Google Scholar]
  • 116.Miura M. Gronthos S. Zhao M. Lu B. Fisher LW. Robey PG. Shi S. SHED: stem cells from human exfoliated deciduous teeth. Proc Natl Acad Sci U S A. 2003;100:5807–5812. doi: 10.1073/pnas.0937635100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Kern S. Eichler H. Stoeve J. Klünter H. Bieback K. Comparative analysis of mesenchymal stem cells from bone marrow, umbilical cord blood, or adipose tissue. Stem Cells. 2006;24:1294–1301. doi: 10.1634/stemcells.2005-0342. [DOI] [PubMed] [Google Scholar]
  • 118.in‘t Anker PS. Scherjon SA. Kleijburg-van der Keur C. de Groot-Swings GM. Claas FH. Fibbe WE. Kanhaia HH. Isolation of mesenchymal stem cells of fetal or maternal origin from human placenta. Stem Cells. 2004;22:1338–1345. doi: 10.1634/stemcells.2004-0058. [DOI] [PubMed] [Google Scholar]
  • 119.Bianco P. Riminucci M. Gronthos S. Robey PG. Bone marrow stromal stem cells: nature, biology, and potential applications. Stem Cells. 2001;19:180–192. doi: 10.1634/stemcells.19-3-180. [DOI] [PubMed] [Google Scholar]
  • 120.Dominici M. Le Blanc K. Mueller I. Slaper-Cortenbach I. Marini F. Krause D. Deans R. Keating A. Prockop DJ. Horwitz E. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy. 2006;8:315–317. doi: 10.1080/14653240600855905. [DOI] [PubMed] [Google Scholar]
  • 121.Gronthos S. Zannettino ACW. Hay SJ. Shi S. Graves SE. Kortesidis A. Simmons PJ. Molecular and cellular characterization of highly purified stromal stem cells derived from human bone marrow. J Cell Sci. 2003;116:1827–1835. doi: 10.1242/jcs.00369. [DOI] [PubMed] [Google Scholar]
  • 122.Kerkis I. Kerkis A. Dozortsev D. Stukart-Parsons GC. Massironi SMG. Pereira LV. Caplan AI. Cerruti HF. Isolation and characterization of a population of immature dental pulp stem cells expressing OCT-4 and other embryonic stem cell markers. Cells Tissues Organs. 2006;184:105–116. doi: 10.1159/000099617. [DOI] [PubMed] [Google Scholar]
  • 123.Kadar K. Kiraly M. Porcsalmy B. Molnar B. Racz GZ. Blazsek J. Kallok K. Szabo EL. Gera I. Gerber G. Varga G. Differentiation potential of stem cells from human dental origin—promise for tissue engineering. J Physiol Pharmacol. 2009;60:167–175. [PubMed] [Google Scholar]
  • 124.Kawanabe N. Murata S. Murakami K. Ishihara Y. Hayano S. Kurosaka H. Kamioka H. Takano-Yamamoto T. Yamashiro T. Isolation of multipotent stem cells in human periodontal ligament using stage-specific embryonic antigen-4. Differentiation. 2010;79:74–78. doi: 10.1016/j.diff.2009.10.005. [DOI] [PubMed] [Google Scholar]
  • 125.Seo BM. Miura M. Gronthos S. Bartold PM. Batouli S. Brahim J. Young M. Robey PG. Wang CY. Shi S. Investigation of multipotent postnatal stem cells from human periodontal ligament. Lancet. 2004;364:149–55. doi: 10.1016/S0140-6736(04)16627-0. [DOI] [PubMed] [Google Scholar]
  • 126.Iwata T. Yamato M. Zhang Z. Mukobata S. Washio K. Ando T. Feijen J. Okano T. Ishikawa I. Validation of human periodontal ligament-derived cells as a reliable source for cytotherapeutic use. J Clin Periodontol. 2010;37:1088–1099. doi: 10.1111/j.1600-051X.2010.01597.x. [DOI] [PubMed] [Google Scholar]
  • 127.Sonoyama W. Liu Y. Yamaza T. Tuan RS. Wang S. Shi S. Huang GTJ. Characterization of apical papilla and its residing stem cells from human immature permanent teeth—a pilot study. J Endod. 2008;34:166–171. doi: 10.1016/j.joen.2007.11.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Chen FG. Zhang WJ. Bi D. Liu W. Wei X. Chen FF. Zhu L. Cui L. Cao Y. Clonal analysis of nestin-vimentin+ multipotent fibroblasts isolated from human dermis. J Cell Sci. 2007;120:2875–2883. doi: 10.1242/jcs.03478. [DOI] [PubMed] [Google Scholar]
  • 129.Lorenz K. Sicker M. Schmelzer E. Rupf T. Salvetter T. Schulz-Siegmund M. Bader A. Multilineage differentiation potential of human dermal skin-derived fibroblasts. Exp Dermatol. 2008;17:925–932. doi: 10.1111/j.1600-0625.2008.00724.x. [DOI] [PubMed] [Google Scholar]
  • 130.Orciani M. Mariggiò MA. Morabito C. Di Benedetto G. Di Primio R. Functional characterization of calcium-signaling pathways of human skin-derived mesenchymal stem cells. Skin Pharmacol Physiol. 2010;23:124–132. doi: 10.1159/000270383. [DOI] [PubMed] [Google Scholar]
  • 131.Gargett CE. Schwab KE. Zillwood RM. Nguyen HPT. Wu D. Isolation and culture of epithelial progenitors and mesenchymal stem cells from human endometrium. Biol Reprod. 2009;80:1136–1145. doi: 10.1095/biolreprod.108.075226. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Gronthos S. Franklin DM. Leddy HA. Robey PG. Storms RW. Gimbles JM. Surface protein characterization of human adipose tissue-derived stromal cells. J Cell Physiol. 2001;189:54–63. doi: 10.1002/jcp.1138. [DOI] [PubMed] [Google Scholar]
  • 133.Meng X. Ichim TE. Zhong J. Rogers A. Yin Z. Jackson J. Wang H. Ge W. Bogin V, et al. Endometrial regenerative cells: a novel stem cell population. J Transl Med. 2007;5:57. doi: 10.1186/1479-5876-5-57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Covas DT. Piccinato CE. Orellana MD. Siufi JLC. Silva WA., Jr. Proto-Siqueira R. Rizzatti EG. Neder L. Silva AR. Rocha V. Zago MA. Mesenchymal stem cells can be obtained from the human saphena vein. Exp Cell Res. 2005;309:340–344. doi: 10.1016/j.yexcr.2005.06.005. [DOI] [PubMed] [Google Scholar]
  • 135.Polisetty N. Fatima A. Madhira SL. Sangwan VS. Vemuganti GK. Mesenchymal cells from limbal stroma of human eye. Mol Vis. 2008;14:431–442. [PMC free article] [PubMed] [Google Scholar]
  • 136.Lama VN. Smith L. Badri L. Flint A. Andrei A. Murray S. Wang Z. Liao H. Toews GB, et al. Evidence for tissue-resident mesenchymal stem cells in human adult lung from studies of transplanted allografts. J Clin Invest. 2007;117:989–996. doi: 10.1172/JCI29713. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Hennrick KT. Keeton AG. Nanua S. Kijek TG. Goldsmith AM. Sajjan US. Bentley JK. Lama VN. Moore BB, et al. Lung cells from neonates show a mesenchymal stem cell phenotype. Am J Respir Crit Care Med. 2007;175:1158–1164. doi: 10.1164/rccm.200607-941OC. [DOI] [PubMed] [Google Scholar]
  • 138.Tsai MS. Lee JL. Chang YJ. Hwang SM. Isolation of human multipotent mesenchymal stem cells from second-trimester amniotic fluid using a novel two-stage culture protocol. Hum Reprod. 2004;19:1450–1456. doi: 10.1093/humrep/deh279. [DOI] [PubMed] [Google Scholar]
  • 139.Roubelakis MG. Pappa KI. Bitsika V. Zagura D. Vlahou A. Papadaki HA. Antsaklis A. Anagnou NP. Molecular and proteomic characterization of human mesenchymal stem cells derived from amniotic fluid: comparison to bone marrow mesenchymal stem cells. Stem Cells Dev. 2007;16:931–951. doi: 10.1089/scd.2007.0036. [DOI] [PubMed] [Google Scholar]
  • 140.Campagnoli C. Roberts IAG. Kumar S. Bennett PR. Bellantuono I. Fisk NM. Identification of mesenchymal stem/progenitor cells in human first-trimester fetal blood, liver, and bone marrow. Blood. 2001;98:2396–2402. doi: 10.1182/blood.v98.8.2396. [DOI] [PubMed] [Google Scholar]
  • 141.in't Anker PS. Noort WA. Scherjon SA. Kleijburg-van der Keur C. Kruisselbrink AB. van Bezooijen RL. Beekhuizen W. Willemze R. Kanhai HH. Fibbe WE. Mesenchymal stem cells in human second-trimester bone marrow, liver, lung, and spleen exhibit a similar immunophenotype but a heterogeneous multilineage differentiation potential. Haematologica. 2003;88:845–852. [PubMed] [Google Scholar]
  • 142.Hu Y. Liao L. Wang Q. Ma L. Ma G. Jiang X. Zhao RC. Isolation and identification of mesenchymal stem cells from human fetal pancreas. J Lab Clin Med. 2003;141:342–349. doi: 10.1016/S0022-2143(03)00022-2. [DOI] [PubMed] [Google Scholar]
  • 143.Fukuchi Y. Nakajima H. Sugiyama D. Hirose I. Kitamura T. Tsuji K. Human placenta-derived cells have mesenchymal stem/progenitor cell potential. Stem Cells. 2004;22:649–658. doi: 10.1634/stemcells.22-5-649. [DOI] [PubMed] [Google Scholar]
  • 144.Lee OK. Kuo TK. Chen WM. Lee KD. Hsieh SL. Chen TH. Isolation of multipotent mesenchymal stem cells from umbilical cord blood. Blood. 2004;103:1669–1675. doi: 10.1182/blood-2003-05-1670. [DOI] [PubMed] [Google Scholar]
  • 145.Hou L. Cao L. Wang D. Wei G. Bai C. Zhang Y. Pei X. Induction of umbilical cord blood mesenchymal stem cells into neuron-like cells in vitro. Int J Hematol. 2003;78:256–261. doi: 10.1007/BF02983804. [DOI] [PubMed] [Google Scholar]
  • 146.Anzalone R. Lo Iacono M. Corrao S. Magno F. Loria T. Cappello F. Zummo G. Farina F. La Rocca G. New emerging potentials for human Wharton's Jelly mesenchymal stem cells: immunological features and hepatocyte-like differentiative capacity. Stem Cells Dev. 2010;19:423–438. doi: 10.1089/scd.2009.0299. [DOI] [PubMed] [Google Scholar]
  • 147.Coppi P. Bartsch G., Jr. Siddiqui MM. Xu T. Santos CC. Perin L. Mostoslavsky G. Serre AC. Snyder EY, et al. Isolation of amniotic stem cell lines with potential for therapy. Nat Biotechnol. 2007;25:100–106. doi: 10.1038/nbt1274. [DOI] [PubMed] [Google Scholar]
  • 148.Guillot PV. Gotherstrom C. Chan J. Kurata H. Fisk NM. Human first-trimester fetal MSC express pluripotency markers and grow faster and have longer telomeres than adult MSC. Stem Cells. 2007;25:646–654. doi: 10.1634/stemcells.2006-0208. [DOI] [PubMed] [Google Scholar]
  • 149.Bartsch G., Jr. Yoo JJ. Coppi P. Siddiqui MM. Schuch G. Pohl HG. Fuhr J. Perin L. Soker S. Atala A. Propagation, expansion, and multilineage differentiation of human somatic stem cells from dermal progenitors. Stem Cells Dev. 2005;14:337–348. doi: 10.1089/scd.2005.14.337. [DOI] [PubMed] [Google Scholar]
  • 150.Trubiani O. Zalzal SF. Paganelli R. Marchisio M. Giancola R. Pizzicannella J. Bühring H. Piattelli M. Caputi S. Nanci A. Expression profile of the embryonic markers Nanog, OCT-4, SSEA-1, SSEA-4, and frizzled-9 receptor in human periodontal ligament mesenchymal stem cells. J Cell Physiol. 2010;225:123–131. doi: 10.1002/jcp.22203. [DOI] [PubMed] [Google Scholar]
  • 151.Battula VL. Bareiss PM. Treml S. Conrad S. Albert I. Hojak S. Abele H. Schewe B. Just L. Skutella T. Bühring H. 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;75:279–291. doi: 10.1111/j.1432-0436.2006.00139.x. [DOI] [PubMed] [Google Scholar]
  • 152.Bais MV. Shabin ZM. Young M. Einhorn TA. Kotton DN. Gerstnefeld LC. Role of Nanog in the maintenance of marrow stromal stem cells during postnatal bone regeneration. Biochem Biophys Res Commun. 2012;417:211–216. doi: 10.1016/j.bbrc.2011.11.087. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Katsara O. Mahaira LG. Iliopoulou EG. Moustaki A. Antsaklis A. Loutradis D. Stefanidis K. Baxevanis CN. Papamichail M. Perez SA. Effects of donor age, gender, and in vitro cellular aging on the phenotypic, functional, and molecular characteristics of mouse bone marrow-derived mesenchymal stem cells. Stem Cells Dev. 2011;20:1549–1561. doi: 10.1089/scd.2010.0280. [DOI] [PubMed] [Google Scholar]
  • 154.Liu TM. Wu YN. Guo XM. Hui JHP. Lee EH. Lim B. Effects of ectopic Nanog and Oct4 overexpression on mesenchymal stem cells. Stem Cells Dev. 2009;18:1013–1021. doi: 10.1089/scd.2008.0335. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Dolorme D. Ringe J. Gallay N. Le Vern Y. Kerboeuf D. Jorgensen C. Rosset P. Sensebé L. Layrolle P. Häupl T. Charbord P. Specific plasma membrane protein phenotype of culture-aplified and native human bone marrow mesenchymal stem cells. Blood. 2008;111:2631–2635. doi: 10.1182/blood-2007-07-099622. [DOI] [PubMed] [Google Scholar]
  • 156.Battula VL. Treml S. Abele H. Bühring HRG. Prospective isolation and characterization of mesenchymal stem cells from human placenta using a frizzled-9-specific monoclonal antibody. Differentiation. 2008;76:326–336. doi: 10.1111/j.1432-0436.2007.00225.x. [DOI] [PubMed] [Google Scholar]
  • 157.Tran TC. Kimura K. Nagano M. Yamashita T. Ohneda K. Sugimori H. Sato F. Sakakibara Y. Hamada H, et al. Identification of human placenta-derived mesenchymal stem cells involved in re-endothelialization. J Cell Physiol. 2010;226:224–235. doi: 10.1002/jcp.22329. [DOI] [PubMed] [Google Scholar]
  • 158.Kaltz N. Ringe J. Holzwarth C. Charbord P. Niemeyer M. Jacobs VR. Peschel C. Haupl T. Oostendorp RAJ. Novel markers of mesenchymal stem cells defined by genome-wide gene expression analysis of stromal cells from different sources. Exp Cell Res. 2010;316:2609–2617. doi: 10.1016/j.yexcr.2010.06.002. [DOI] [PubMed] [Google Scholar]
  • 159.Quirici N. Soligo D. Bossolasco P. Servida F. Lumini C. Deliliers GL. Isolation of bone marrow mesenchymal stem cells by anti-nerve growth factor receptor antibodies. Exp Hematol. 2002;30:783–791. doi: 10.1016/s0301-472x(02)00812-3. [DOI] [PubMed] [Google Scholar]
  • 160.Bühring HJ. Battula L. Treml S. Schewe B. Kanz L. Vogel W. Novel markers for the prospective isolation of human MSC. Ann N Y Acad Sci. 2007;1106:262–271. doi: 10.1196/annals.1392.000. [DOI] [PubMed] [Google Scholar]
  • 161.Battula VL. Treml S. Bareiss PM. Gieseke F. Roelofs H. de Zwart P. Müller I. Schewe B. Skutella T, et al. Isolation of functionally distinct mesenchymal stem cells subsets using antibodies against CD56, CD271, and mesenchymal stem cell antigen-1. Haematologica. 2009;94:173–184. doi: 10.3324/haematol.13740. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Martinez C. Hofmann TJ. Marino R. Dominici M. Horwitz EM. Human bone marrow mesenchymal stromal cells express the neural ganglioside GD2: a novel surface marker for the identification of MSCs. Blood. 2007;109:4245–4248. doi: 10.1182/blood-2006-08-039347. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Xu J. Liao WB. Gu DS. Liang L. Liu M. Du WT. Liu PX. Zhang L. Lu SH, et al. Neural ganglioside GD2 identifies a subpopulation of mesenchymal stem cells in umbilical cord. Cell Physiol Biochem. 2009;23:415–424. doi: 10.1159/000218188. [DOI] [PubMed] [Google Scholar]
  • 164.Jin Jin H. Nam HY. Bae YK. Kim SY. Im IR. Oh W. Yang YS. Choi SJ. Kim SW. GD2 expression is closely associated with neuronal differentiation of human umbilical cord blood-derived mesenchymal stem cells. Cell Mol Life Sci. 2010;67:1845–1858. doi: 10.1007/s00018-010-0292-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Gang EJ. Bosnakovski D. Figueiredo CA. Visser JW. Perlingeiro RCR. SSEA-4 identifies mesenchymal stem cells from bone marrow. Blood. 2007;109:1743–1751. doi: 10.1182/blood-2005-11-010504. [DOI] [PubMed] [Google Scholar]
  • 166.Gao J. Yang T. Han J. Yan K. Qiu X. Zhou Y. Fan Q. Ma B. MicroRNA expression during osteogenic differentiation of human multipotent mesenchymal stromal cells from bone marrow. J Cell Biochem. 2011;112:1844–1856. doi: 10.1002/jcb.23106. [DOI] [PubMed] [Google Scholar]
  • 167.Huang S. Wang S. Bian C. Yang Z. Zhou H. Zeng Y. Li H. Han Q. Zhao RC. Upregulation of miR-22 promotes osteogenic differentiation and inhibits adipogenic differentiation of human adipose tissue-derived mesenchymal stem cells by repressing HDAC6 protein expression. Stem Cells Dev. 2012;21:2531–2540. doi: 10.1089/scd.2012.0014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Kim YJ. Bae SW. Yu SS. Bae YC. Jun JS. miR-196a regulates proliferation and osteogenic differentiation in mesenchymal stem cells derived from human adipose tissue. J Bone Miner Res. 2009;24:816–825. doi: 10.1359/jbmr.081230. [DOI] [PubMed] [Google Scholar]
  • 169.Bork S. Horn P. Castoldi M. Hellwig I. Ho AS. Wagner W. Adipogenic differentiation of human mesenchymal stromal cells is downregulated by microRNA-369-5p and up-deregulated by microRNA-371. J Cell Physiol. 2010;226:2226–2223. doi: 10.1002/jcp.22557. [DOI] [PubMed] [Google Scholar]
  • 170.Skårn M. Namløs HM. Noordhuis P. Wang MY. Meza-Zepeda LA. Myklebost O. Adipocyte differentiation of human bone marrow-derived stromal cells is modulated by microRNA-155, microRNA-221, and microRNA-222. Stem Cells Dev. 2012;21:873–883. doi: 10.1089/scd.2010.0503. [DOI] [PubMed] [Google Scholar]
  • 171.Yang Z. Bian C. Zhou H. Huang S. W S. Liao L. Zhao RC. MicroRNA hsa-miR-138 inhibits adipogenic differentiation of human adipose tissue-derived mesenchymal stem cells through adenovirus EID-1. Stem Cells Dev. 2011;20:259–267. doi: 10.1089/scd.2010.0072. [DOI] [PubMed] [Google Scholar]
  • 172.Kim D. Song J. Jin EJ. MicroRNA-221 regulates chondrogenic differentiation through promoting proteosomal degradation of slug by targeting Mdm2. J Biol Chem. 2010;285:26900–26907. doi: 10.1074/jbc.M110.115105. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 173.Bae S. Ahn JH. Park CW. Son HK. Kim K. Lim N. Jeon C. Kim H. Gene and microRNA expression signatures of human mesenchymal stromal cells in comparison to fibroblasts. Cell Tissue Res. 2009;335:565–573. doi: 10.1007/s00441-008-0729-y. [DOI] [PubMed] [Google Scholar]
  • 174.Yang B. Guo H. Zhang Y. Chen L. Ying D. Dong S. MicroRNA-145 regulates chondrogenic differentiation of mesenchymal stem cells by targeting Sox9. PLoS One. 2011;6:e21679. doi: 10.1371/journal.pone.0021679. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Xu J. Kang Y. Liao W. Yu L. MiR-194 regulates chondrogenic differentiation of human adipose-derived stem cells by targeting Sox5. PLoS One. 2012;7:e31861. doi: 10.1371/journal.pone.0031861. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Kao L. Yu S. Singh S. Wang K. Kao A. Li SS. Comparative profiling of mRNA and microRNA expression in human mesenchymal stem cells derived from adult adipose and lipoma tissues. Open Stem Cell J. 2009;1:1–9. [Google Scholar]
  • 177.Greco SJ. Rameshwar P. MicroRNAs regulate synthesis of the neurotransmitter substance P in human mesenchymal stem cell-derived neuronal cells. Proc Natl Acad Sci U S A. 2007;104:15484–15489. doi: 10.1073/pnas.0703037104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Sutherland HJ. Eaves CJ. Eaves AC. Dragowska W. Lansdorp PM. Characterization and partial purification of human marrow cells capable of initiating long-term hematopoiesis in vitro. Blood. 1989;74:1563–1570. [PubMed] [Google Scholar]
  • 179.Nayar B. Raju GMK. Deka D. Hematopoietic stem/progenitor cell harvesting from umbilical cord Blood. Int J Gynaecol Obstet. 2002;79:31–32. doi: 10.1016/s0020-7292(02)00187-x. [DOI] [PubMed] [Google Scholar]
  • 180.Craig W. Kay R. Cutler RL. Lansdorp PM. Expression of Thy-1 on human hematopoietic progenitor cells. J Exp Med. 1993;177:1331–1342. doi: 10.1084/jem.177.5.1331. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.McCredie KB. Hersh EM. Freireich EJ. Cells capable of colony formation in the peripheral blood of man. Science. 1971;271:293–294. doi: 10.1126/science.171.3968.293. [DOI] [PubMed] [Google Scholar]
  • 182.Jansen J. Hanks S. Thompson JM. Dugan MJ. Akard LP. Transplantation of hematopoietic stem cells from the peripheral blood. J Cell Mol Med. 2005;9:37–50. doi: 10.1111/j.1582-4934.2005.tb00335.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Ogawa M. Differentiation and proliferation of hematopoietic stem cells. Blood. 1993;81:2844–2853. [PubMed] [Google Scholar]
  • 184.Krause DS. Fackler MJ. Civin CI. May WS. CD34: structure, biology, and clinical utility. Blood. 1996;87:1–13. [PubMed] [Google Scholar]
  • 185.Fackler MJ. Krause DS. Smith OM. Civin CI. May MS. Full-length but not truncated CD34 inhibits hematopoietic cell differentiation of M1 cells. Blood. 1995;85:3040–3047. [PubMed] [Google Scholar]
  • 186.Nielsen JS. Mcnagny KM. CD34 is a key regulator of hematopoietic stem cell trafficking to bone marrow and mast cell progenitor trafficking in the periphery. Microcirculation. 2009;16:487–496. doi: 10.1080/10739680902941737. [DOI] [PubMed] [Google Scholar]
  • 187.Berenson RJ. Andrews RG. Bensinger WL. Kalamasz D. Knitter G. Buckner CD. Bernstein ID. Antigen CD34+ marrow cells engraft lethally irradiated baboon. J Clin Invest. 1988;81:951–955. doi: 10.1172/JCI113409. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Berenson RJ. Bensinger WI. Hill RS. Andrews RG. Garcia-Lopez J. Kalamasz DF. Still BJ. Spitzer G. Buckner CD. Bernstein ID. Thomas ED. Engraftment after infusion of CD34+ marrow cells in patients with breast cancer or neuroblastoma. Blood. 1991;177:1717–1722. [PubMed] [Google Scholar]
  • 189.Fina L. Molgaard HV. Robertson D. Bradley NJ. Monaghan P. Delia D. Sutherland DR. Baker MA. Greaves MF. Expression of the CD34 gene in vascular endothelial cells. Blood. 1990;75:2417–2426. [PubMed] [Google Scholar]
  • 190.Osawa M. Hanada K. Hamada H. Nakauchi H. Long-term lymphohematopoietic reconstitution by a single CD34-low/egative hematopoietic stem cell. Science. 1996;273:242–245. doi: 10.1126/science.273.5272.242. [DOI] [PubMed] [Google Scholar]
  • 191.Bhatia M. Bonnet D. Murdoch B. Gan OI. Dick JE. A newly discovered class of human hematopoietic cells with SCID repopulating activity. Nat Med. 1998;4:1038–1045. doi: 10.1038/2023. [DOI] [PubMed] [Google Scholar]
  • 192.Zanjani ED. Almeida-Porada G. Livingston AG. Porada CD. Ogawa M. Engraftment and multilineage expression of human bone marrow CD34- cells in vivo. Ann N Y Acad Sci. 1999;872:220–232. doi: 10.1111/j.1749-6632.1999.tb08467.x. [DOI] [PubMed] [Google Scholar]
  • 193.Zanjani ED. Almeida-Porada G. Livingston AG. Zeng H. Ogawa M. Reversible expression of CD34 by adult human bone marrow long-term engrafting hematopoietic stem cells. Exp Hematol. 2003;31:406–412. doi: 10.1016/s0301-472x(03)00051-1. [DOI] [PubMed] [Google Scholar]
  • 194.Dao MA. Arevalo J. Nolta JA. Reversibility of CD34 expression on human hematopoietic stem cells that retain the capacity for secondary reconstitution. Blood. 2003;101:212–218. doi: 10.1182/blood-2002-01-0025. [DOI] [PubMed] [Google Scholar]
  • 195.Gao Z. Fackler MJ. Leung W. Lumkul R. Ramirez M. Theobald N. Malech HL. Civin CI. Human CD34+ cell preparations contain over 100-fold greater NOD/SCID mouse engrafting capacity than do CD34- cell preparations. Exp Hematol. 2001;29:910–921. doi: 10.1016/s0301-472x(01)00654-3. [DOI] [PubMed] [Google Scholar]
  • 196.Bhatia M. Wang JCY. Kapp U. Bonnet D. Dick JE. Purification of primitive human hematopoietic cells capable of repopulating immune-deficient mice. Proc Natl Acad Sci U S A. 1997;94:5320–5325. doi: 10.1073/pnas.94.10.5320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Nakamura Y. Ando K. Chargui J. Kawada H. Sato T. Tsuji T. Hotta T. Kato S. Ex vivo generation of CD34+ cells from CD34- hematopoietic cells. Blood. 1999;94:4053–4059. [PubMed] [Google Scholar]
  • 198.Gallacher L. Murdoch B. Wu DM. Karanu FN. Keeney M. Bhatia M. Isolation and characterization of human CD34-Lin- and CD34+Lin- hematopoietic stem cells using cell surface markers AC133 and CD7. Blood. 2000;95:2813–2820. [PubMed] [Google Scholar]
  • 199.Terstappen LWMM. Huang S. Safford M. Lansdorp PM. Loken MR. Sequential generations of hematopoietic colonies derived from single nonlineage-committed CD34+CD38- progenitor cells. Blood. 1991;77:1218–1227. [PubMed] [Google Scholar]
  • 200.Ivanovic Z. Hematopoietic stem cells in research and clinical applications: the “CD34 issue.”. World J Stem Cells. 2010;2:18–23. doi: 10.4252/wjsc.v2.i2.18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Yin AH. Miraglia S. Zanjani ED. Almeida-Porada G. Ogawa M. Leary AG. Olweus J. Kearney J. Buck DW. AC133, a novel marker for human hematopoietic stem and progenitor cells. Blood. 1997;90:5002–5012. [PubMed] [Google Scholar]
  • 202.Goussetis E. Theodosaki M. Paterakis G. Peristeri J. Petropoulos D. Kitra V. Papassarandis C. Graphakos S. A functional hierarchy among the CD34 hematopoietic cells based on in vitro proliferative and differentiative potential of AC133+, CD34bright and AC133dim/-CD34+ human cord blood cells. J Hematother Stem Cell Res. 2000;9:827–840. doi: 10.1089/152581600750062255. [DOI] [PubMed] [Google Scholar]
  • 203.Wynter EA. Buck D. Hart C. Heywood R. Coutinho LH. Clayton A. Rafferty JA. Burt D. Guenechea G, et al. CD34+ AC133+ cells isolated from cord blood are highly enriched in long-term culture-initiating cells, NOD/SCIDRepopulating cells and dentritic cell progenitors. Stem Cells. 1998;16:387–396. doi: 10.1002/stem.160387. [DOI] [PubMed] [Google Scholar]
  • 204.Kania G. Corbeil D. Fuchs J. Tarasov KV. Blyszczuk P. Huttner WB. Boheler KR. Wobus AM. Somatic stem cell marker prominin-1/CD133 is expressed in embryonic stem cell-derived progenitors. Stem Cells. 2005;23:791–804. doi: 10.1634/stemcells.2004-0232. [DOI] [PubMed] [Google Scholar]
  • 205.Schmelzer E. Zhang L. Bruce A. Wauthier E. Ludlow J. Yao H. Moss N. Melhem A. McClelland R, et al. Human hepatic stem cells from fetal and postnatal donors. J Exp Med. 2007;204:1973–1987. doi: 10.1084/jem.20061603. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Lee A. Kessler JD. Read T. Kaiser C. Corbeil D. Huttner WB. Johnson JE. Wechsler-Reya RJ. Isolation of neural stem cells from the postnatal cerebellum. Nat Neurosci. 2005;8:723–729. doi: 10.1038/nn1473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Baum CM. Weissman IL. Tsukamoto AN. Buckle AM. Peault B. Isolation of a candidate human hematopoietic stem-cell population. Proc Natl Acad Sci U S A. 1992;89:2804–2808. doi: 10.1073/pnas.89.7.2804. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Haas R. Möhle R. Pftjrsich M. Fruehauf S. Witt B. Goldschmidt H. Hunstein W. Blood-Derived autografts collected during granulocyte colony-stimulating factor-enhanced recovery are enriched with early Thy-l+ hematopoietic progenitor cells. Blood. 1995;5:1936–1943. [PubMed] [Google Scholar]
  • 209.Mayani H. Lansdorp PM. Thy-l expression is linked to functional properties of primitive hematopoietic progenitor cells from human umbilical cord blood. Blood. 1994;83:2410–2417. [PubMed] [Google Scholar]
  • 210.Humeau L. Bardin F. Maroc C. Alario T. Galindo R. Mannoni P. Chabannon C. Phenotypic, molecular, and functional characterization of human peripheral blood CD34+/THY-1+ cells. Blood. 1996;87:949–955. [PubMed] [Google Scholar]
  • 211.Notta F. Doulatov S. Laurenti S. Poeppl A. Jurisica I. Dick JE. Isolation of single human hematopoietic stem cells capable of long-term multilineage engraftment. Science. 2011;333:218–221. doi: 10.1126/science.1201219. [DOI] [PubMed] [Google Scholar]
  • 212.Takeda H. Yamamoto M. Morita N. Tanizawa T. Relationship between Thy-1 expression and cell-cycle distribution in human bone marrow hematopoietic progenitors. Am J Hematol. 2005;79:187–193. doi: 10.1002/ajh.20362. [DOI] [PubMed] [Google Scholar]
  • 213.Majeti R. Park CY. Weissman IL. Identification of a hierarchy of multipotent hematopoietic progenitors in human cord blood. Cell Stem Cell. 2007;1:635–645. doi: 10.1016/j.stem.2007.10.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214.Ziegler BL. Valtieri M. Porada GA. De Maria R. Müller R. Masella B. Gabbianelli M. Casella I. Pelosi E, et al. KDR receptor: a key marker defining hematopoietic stem cells. Science. 1999;285:1553–1558. doi: 10.1126/science.285.5433.1553. [DOI] [PubMed] [Google Scholar]
  • 215.Pelosi E. Valtieri M. Coppola S. Botta R. Gabbianelli M. Lulli V. Marziali G. Masella B. Müller R, et al. Identification of the hemangioblast in postnatal life. Blood. 2002;100:3203–3208. doi: 10.1182/blood-2002-05-1511. [DOI] [PubMed] [Google Scholar]
  • 216.Conze T. Lammers R. Kuci S. Scherl-Mostageer M. Schweifer N. Kanz L. Buhring HJ. CDCP1 is a novel marker for hematopoietic stem cells. Ann N Y Acad Sci. 2003;996:222–226. doi: 10.1111/j.1749-6632.2003.tb03249.x. [DOI] [PubMed] [Google Scholar]
  • 217.Hao Q. Shah AJ. Thiemann FT. Smogorzewska EM. Crooks GM. A functional comparison of CD34+CD38- cells in cord blood and bone marrow. Blood. 1995;86:3745–3753. [PubMed] [Google Scholar]
  • 218.Koutnaa I. Klabusayb M. Kohutovab V. Krontorada P. Svobodaa Z. Kozubeka M. Mayerb J. Evaluation of CD34+ and Lin2- selected cells from peripheral blood stem cell grafts of patients with lymphoma during differentiation in culture ex vivo using a cDNA microarray technique. Exp Hematol. 2006;34:832–840. doi: 10.1016/j.exphem.2006.04.006. [DOI] [PubMed] [Google Scholar]
  • 219.Danet JH. Luongo JL. Butler G. Lu MM. Tenner AJ. Simon MC. Bonnet DA. C1qRp defines a new human stem cell population with hematopoietic and hepatic potential. Proc Natl Acad Sci U S A. 2002;99:10441–10445. doi: 10.1073/pnas.162104799. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220.Moreb JS. Aldehyde dehydrogenase as a marker for stem cells. Stem Cell Res Curr Stem Cell Res Ther. 2008;3:237–246. doi: 10.2174/157488808786734006. [DOI] [PubMed] [Google Scholar]
  • 221.Storms RW. Trujillo AP. Springer JB. Shah L. Colvin OM. Ludeman SM. Smith C. Isolation of primitive human hematopoietic progenitors on the basis of aldehyde dehydrogenase activity. Proc Natl Acad Sci U S A. 1999;96:9118–9123. doi: 10.1073/pnas.96.16.9118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222.Christ O. Lucke K. Imren S. Leung K. Hamilton M. Eaves A. Smith C. Eaves C. Improved purification of hematopoietic stem cells based on their elevated aldehyde dehydrogenase activity. Haematologica. 2007;92:1165–1172. doi: 10.3324/haematol.11366. [DOI] [PubMed] [Google Scholar]
  • 223.Kataoka K. Sato T. Yoshimi A. Goyama S. Tsuruta T. Kobayashi H. Shimabe M. Arai S. Nakagawa M, et al. Evi1 is essential for hematopoietic stem cell self-renewal, and its expression marks hematopoietic cells with long-term multilineage repopulating activity. J Exp Med. 2011;208:2403–2416. doi: 10.1084/jem.20110447. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224.Ooi AGL. Karsunky H. Majeti R. Butz S. Vestweber D. Ishida T. Quertermous T. WeissmanN IL. Forseberg EC. The adhesion molecule Esam1 is a novel hematopoietic stem cell marker. Stem Cells. 2009;27:653–661. doi: 10.1634/stemcells.2008-0824. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 225.Christensen JL. Weissman IL. Flk-2 is a marker in hematopoietic stem cell differentiation: a simple method to isolate long-term stem cells. Proc Natl Acad Sci USA. 2001;98:14541–14546. doi: 10.1073/pnas.261562798. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226.Liu S. Fu R. Yu H. Li K. Tsai C. Shyu andS Lin W. MicroRNAs regulation modulated self-renewal and lineage differentiation of stem cells. Cell Transplant. 2009;18:1039–1045. doi: 10.3727/096368909X471224. [DOI] [PubMed] [Google Scholar]
  • 227.Vasilatou D. Papageorgiou S. Pappa V. Papageorgiou E. Dervenoulas J. The role of microRNAs in normal and malignant hematopoiesis. Eur J Haematol. 2009;84:1–16. doi: 10.1111/j.1600-0609.2009.01348.x. [DOI] [PubMed] [Google Scholar]
  • 228.Bissels U. Wild S. Tomiuk S. Holste A. Hafner M. Tuschl T. Bosio A. Absolute quantification of microRNAs by using a universal reference. RNA. 2009;15:2375–2384. doi: 10.1261/rna.1754109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229.Bissels U. Wild S. Tomiuk S. Hafner M. Scheel H. Mihailovic A. Choi Y. Tuschl T. Bosio A. Combined characterization of microRNA and mRNA profiles delineates early differentiation pathways of CD133+ and CD34+ hematopoietic stem and progenitor cells. Stem Cells. 2011;29:847–857. doi: 10.1002/stem.627. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230.Georgantas RW. Hildreth R. Morisot S. Alder J. Liu C. Heimfeld S. Calin GA. Croce CM. Civin CI. CD34+ hematopoietic stem-progenitor cell microRNA expression and function: a circuit diagram of differentiation control. Proc Natl Acad Sci U S A. 2007;104:2750–2755. doi: 10.1073/pnas.0610983104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 231.Liao R. Sun J. Zhang L. Lou G. Chen M. Zhou D. Chen Z. Zhang S. MicroRNAs play a role in the development of human hematopoietic stem cells. J Cell Biochem. 2008;104:805–817. doi: 10.1002/jcb.21668. [DOI] [PubMed] [Google Scholar]
  • 232.Merkerova M. Vasikova A. Belickova M. Bruchova H. MicroRNA expression profiles in umbilical cord blood cell lineages. Stem Cells Dev. 2010;19:17–25. doi: 10.1089/scd.2009.0071. [DOI] [PubMed] [Google Scholar]
  • 233.Jin P. Wang E. Ren J. Childs R. Shin JW. Khuu H. Marincola FM. Stroncek DF. Differentiation of two types of mobilized peripheral blood stem cells by microRNA and cDNA expression analysis. J Transl Med. 2008;6:39. doi: 10.1186/1479-5876-6-39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234.Landgraf P. Rusu M. Sheridan R. Sewer A. Iovino N. Aravin A. Pfeffer S. Rice A. Kamphorst AO, et al. A mammalian microRNA expression atlas based on small RNA library sequencing. Cell. 2007;129:1401–1414. doi: 10.1016/j.cell.2007.04.040. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Stem Cells and Development are provided here courtesy of Mary Ann Liebert, Inc.

RESOURCES