Next Article in Journal
Artificial Intelligence Its Uses and Application in Pediatric Dentistry: A Review
Previous Article in Journal
Morpho-Functional Effect of a New Collagen-Based Medical Device on Human Gingival Fibroblasts: An In Vitro Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

A Simplified and Effective Approach for the Isolation of Small Pluripotent Stem Cells Derived from Human Peripheral Blood

by
Eirini Filidou
1,†,
Leonidas Kandilogiannakis
1,†,
Gesthimani Tarapatzi
1,
Michail Spathakis
1,
Colin Su
2,
Alin Rai
3,4,
David W. Greening
3,4,5,6,
Konstantinos Arvanitidis
1,
Vasilis Paspaliaris
2,* and
George Kolios
1
1
Laboratory of Pharmacology, Faculty of Medicine, Democritus University of Thrace, Dragana, 68100 Alexandroupolis, Greece
2
Tithon Biotech Inc., San Diego, CA 92127, USA
3
Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
4
Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Melbourne, VIC 3086, Australia
5
Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC 3052, Australia
6
Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
*
Author to whom correspondence should be addressed.
These authors have equal contributions.
Biomedicines 2023, 11(3), 787; https://doi.org/10.3390/biomedicines11030787
Submission received: 25 January 2023 / Revised: 24 February 2023 / Accepted: 3 March 2023 / Published: 5 March 2023
(This article belongs to the Section Molecular and Translational Medicine)

Abstract

:
Pluripotent stem cells are key players in regenerative medicine. Embryonic pluripotent stem cells, despite their significant advantages, are associated with limitations such as their inadequate availability and the ethical dilemmas in their isolation and clinical use. The discovery of very small embryonic-like (VSEL) stem cells addressed the aforementioned limitations, but their isolation technique remains a challenge due to their small cell size and their efficiency in isolation. Here, we report a simplified and effective approach for the isolation of small pluripotent stem cells derived from human peripheral blood. Our approach results in a high yield of small blood stem cell (SBSC) population, which expresses pluripotent embryonic markers (e.g., Nanog, SSEA-3) and the Yamanaka factors. Further, a fraction of SBSCs also co-express hematopoietic markers (e.g., CD45 and CD90) and/or mesenchymal markers (e.g., CD29, CD105 and PTH1R), suggesting a mixed stem cell population. Finally, quantitative proteomic profiling reveals that SBSCs contain various stem cell markers (CD9, ITGA6, MAPK1, MTHFD1, STAT3, HSPB1, HSPA4), and Transcription reg complex factors (e.g., STAT5B, PDLIM1, ANXA2, ATF6, CAMK1). In conclusion, we present a novel, simplified and effective isolating process that yields an abundant population of small-sized cells with characteristics of pluripotency from human peripheral blood.

1. Introduction

Regenerative medicine is an ever-growing scientific field that employs stem cells for the generation of differentiated cells and/or tissues with possible applications in clinical practice [1,2,3]. Stem cells are characterized by three main traits, their self-renewal, their differentiation capacity and their regenerative potential, and can be categorized based on either their differentiation potential or the tissue origin [4,5,6]. Depending on their differentiation potential, stem cells can be categorized as totipotent, pluripotent, multipotent, oligopotent or unipotent [7,8,9]. Regarding tissue origin, the two major categories are embryonic and somatic stem cells. Embryonic stem cells are pluripotent, having significant potential for self-renewal and cell differentiation, whereas somatic stem cells are more confined, and their characteristics vary depending on the tissue origin [10,11,12]. Thus, for many years, there has been a great interest in embryonic stem cells, as they can be excellent candidates for applications in regenerative medicine, such as cell-based therapy [13,14,15]. Nonetheless, there are ethical and experimental limitations to using these cells, including scalable, efficient isolation [16,17].
In adults, stem cells can be found in various tissues, such as the bone marrow, peripheral blood, adipose tissue, intestine and many others, but, in most cases, they are characterized as multipotent, giving rise to limited types of different cells [18,19]. Hematopoietic, mesenchymal and epithelial stem cells are the three major multipotent stem cells found in adult tissues [11,12,20]. Although most multipotent stem cells can be easily isolated and cultured, their use in regenerative medicine is limited due to their restricted differentiation capacity [16,20]. Therefore, regenerative medicine requires stem cells that are pluripotent, can be easily obtained in large numbers and do not pose ethical constraints.
The discovery of induced pluripotent stem cells (iPSCs) was considered a milestone in regenerative medicine because, for the first time, researchers had the opportunity to utilize a natural source of adult pluripotent stem cells [21,22]. iPSCs were generated in vitro by introducing four transcription factors, OCT4, SOX2, MYC and KLF4, the “Yamanaka factors”, into differentiated adult cells, thus reversing the cell fate and resulting in pluripotent undifferentiated stem cells [21,23,24]. One of the main advantages of iPSCs was the possibility of having an unlimited supply of autologous cells, with several clinical applications, either as undifferentiated stem cells or differentiated into other adult cell types and, most importantly, without the fear of immune rejection [25,26]. Nonetheless, iPSCs also presented several challenges, with the most significant ones being their high cost and manufacturing time, and thus, in most cases, iPSCs were clinically impractical to use [27,28]. One possible solution was the generation and banking of donor iPSC cell lines, which can give rise to allogeneic tissue-matching cell products. Although this approach resulted in a significant reduction in the manufacturing time, it is still a relatively high-cost procedure, which also demands the use of immunosuppressive agents when clinically applied [29,30,31]. Therefore, the ultimate solution to the aforementioned challenges would be the discovery and identification of a naturally occurring and autologous pluripotent stem cell population, which could be found in abundance and have low cost and processing time [32,33].
Fifteen years ago, the group of Ratajczak identified in adults a population of very small cells bearing markers of pluripotency, such as stage-specific antigen (SSEA), nuclear Oct-4A, Nanog, and Rex1, and thus, named these cells “very small embryonic-like” (VSEL) stem cells [34,35]. VSELs have a size of 5–7 μm in humans, are thought to give rise to various cell types (including hematopoietic, mesenchymal and endothelial stem cells) and are believed to act as a backup regeneration for adult tissues [36,37]. Nonetheless, isolation of VSELs from peripheral blood has since remained a challenge due to their small cell size and the limited number of recoverable cells [38,39]. Most of the isolation techniques use density gradients or reagents for erythrocyte lysis [40,41,42] that not only results in a limited number of isolated cells but also makes their isolation complex and time-consuming. Therefore, to take advantage of the regenerative and translational potential of VSELs, we require a streamlined, efficient approach to gain this pluripotent blood-derived stem cell population.
Taking into consideration all the aforementioned complications in isolating VSELs, this study employed a simplified approach using processing steps of centrifugation and filtration of fresh whole blood, without the use of density gradients and immuno-cell separation methods, in order to isolate and biochemically characterize small blood stem cells (SBSCs) from peripheral blood.

2. Materials and Methods

2.1. Healthy Volunteers

Peripheral blood was collected from healthy volunteers, who were informed and agreed to participate in this study by giving their written consent. All principles outlined in the Declaration of Helsinki for all human experimental investigations were followed. Healthy volunteers were of all sexes and their ages varied from 20 to 30 years old.

2.2. Isolation of SBSCs

Isolation of SBSCs was performed with the following steps. Briefly, 30 mL peripheral blood was collected in tubes containing sodium citrate and the tubes were then centrifuged at 600× g for 10 min to separate red blood cells from the plasma layer. The plasma layer was then carefully transferred into a new centrifuge tube and centrifuged again at 1200× g for another 10 min. Supernatant plasma was then discarded, and the pellet containing the SBSCs was resuspended in 1 ml of sterile saline. Finally, the suspension was filtered through a 5 µm syringe-driven filter and SBSCs were collected for further studies.

2.3. SBSCs Counting

SBSCs were isolated, as described above, from healthy young volunteers, and appropriate dilutions with sterile saline were made. SBSCs were then stained with 0.4% trypan blue solution (Gibco™—Thermo Fisher Scientific, Waltham, MA, USA) in order to exclude any possible dead cells and debris, and a 10 µL of the sample was loaded on each loading groove of the Neubauer chamber. Using a 40× magnification, the five RBC squares of the counting chambers were identified, and the live population of SBSCs was counted. The final number of live SBSCs was calculated using the formula:
SBSCs   per   μ L = counted   SBSCs counted   surface   ×   chamber   depth   ×   dilution   factor
Finally, the calculated number of isolated SBSCs was presented as the number of SBCSs per ml of plasma from peripheral blood.

2.4. Kyoto Probe 1 Staining

SBSCs were stained using Kyoto Probe 1 (KP-1; Goryo Chemical, Sapporo Japan) according to the manufacturer’s instructions. Briefly, SBSCs were centrifuged at 1200× g for 10 min, and the pellet was then reconstituted in phosphate-buffered saline (PBS; Sigma-Aldrich, St. Louis, MO, USA). Next, 10 μg of KP-1 were dissolved in Dimethyl Sulfoxide (DMSO; Sigma-Aldrich, St. Louis, MO, USA) to prepare a 5 mM stock solution and then diluted again in PBS to prepare a 2 μM cell stain solution. The stain solution was then added to the cells, which were incubated for 3 h, and then the culture slides were studied using a fluorescent microscope (Leica DM2000, Leica Microsystems GmbH, Wetzlar, Germany).

2.5. Immunofluorescence

SBSCs were characterized using the immunofluorescence method, as previously described [43,44]. Cells were centrifuged at 1200× g for 10 min, and the pellet was then reconstituted and fixed in ice-cold 4% paraformaldehyde (PFA; Sigma-Aldrich, St. Louis, MO, USA) for 20 min at 4 °C. After fixation, cells were washed with PBS (Sigma-Aldrich, St. Louis, MO, USA), and then permeabilization was achieved using 0.05% Triton X-100 (Sigma-Aldrich, St. Louis, MO, USA) in 1X PBS for 10 min. To eliminate nonspecific staining, samples were incubated for 60 min with 5% bovine serum albumin (BSA; Sigma-Aldrich, Darmstadt, Germany). Next, antibodies against isotype controls (Supplementary Figure S1) or targets of interest (Table 1) (Supplementary Figure S2) were added at a 1:50 dilution and incubated for 1 h at room temperature. Finally, DAPI (Sigma-Aldrich, Darmstadt, Germany) was added to stain the nuclei, and the samples were observed under a fluorescent microscope (Leica DM2000, Leica Microsystems GmbH, Wetzlar, Germany).

2.6. Hematoxylin and Eosin (H&E) Staining

Once isolated, SBSCs were fixed using ice-cold 4% PFA (Sigma-Aldrich, St. Louis, MO, USA) for 20 min at 4 °C. After fixation, cells were washed with PBS (Sigma-Aldrich, St. Louis, MO, USA), and then Hematoxylin (Sigma-Aldrich, St. Louis, MO, USA) was added and incubated for 3 min. Cells were washed with deionized water and then with tap water, and then destain was accomplished using acid ethanol. Cells were washed again with deionized water and then with tap water, and then Eosin (Sigma-Aldrich, St. Louis, MO, USA) was added and incubated for 30 s. Lastly, cells were incubated in ethanol, and the culture slides were studied using a light microscope (Leica DM2000, Leica Microsystems GmbH, Wetzlar, Germany).

2.7. Culture of Human Embryonic Stem Cell Line H1

The human embryonic stem cell line H1 (Wicell International Stem Cell Bank, Wicell Research Institute, Madison, WI, USA) was cultured as previously described [45] and served as a positive control in antibody verification of embryonic stem cell markers. H1 stem cells were cultured in 6-well plates coated with Matrigel (BD Biosciences, San Jose, CA, USA), fed daily with mTeSR1 (Stem Cell Technologies, Vancouver, BC, Canada) and observed under a microscope for any spontaneous differentiation. When confluence reached around 90%, H1 stem cells were passaged, seeded in Matrigel-coated chamber slides (ibidi GmbH, Martinsried, Planegg, Germany) and later stained for the detection of embryonic stem cell markers using immunofluorescence.

2.8. Isolation of White Blood Cells (WBCs)

Peripheral blood was collected in tubes containing sodium citrate, and equal parts of whole blood and sterile saline were mixed and then carefully applied over Biocoll separating solution (Sigma-Aldrich, St. Louis, MO, USA). Samples were then centrifuged at 1200× g for 20 min, and an enriched layer of WBCs was later formed and retrieved using a Pasteur’s pipette. WBCs were then washed twice using sterile saline and used for further studies.

2.9. Sample Preparation for Proteomic Analysis

SBSCs were first cultured for 3 days in matrigel-coated (Corning, New York, NY, USA) plates and in DMEM (Gibco Thermo Fisher Scientific, Waltham, MA, USA) in order to discard any inhibitory components found in the blood. In addition, plasma cells, which were obtained after blood centrifugation at 600× g for 10 min and removal of red and white cells, served as controls in the proteomic analysis. Plasma cells and SBSCs (n = 4) were then lysed in 1% (v/v) sodium dodecyl sulfate (SDS; Sigma-Aldrich, St. Louis, MO, USA) and 50 mM HEPES pH 8.0 (Sigma-Aldrich, St. Louis, MO, USA). Sample homogenization was performed using tip probe sonication (Misonix—S-4000 Ultrasonic Liquid Processor). Lysates were quantified with microBCA (Thermo Fisher Scientific, Waltham, MA, USA) and then normalized samples (10 μg protein) were prepared for proteomic analysis as described [46,47] using the single-pot solid-phase-enhanced sample preparation (SP3) method [48]. The samples were reduced (10 mM dithiothreitol, DTT; Sigma-Aldrich, St. Louis, MO, USA) for 45 min at 25 °C and alkylated (20 mM iodoacetamide; Sigma-Aldrich, St. Louis, MO, USA) for 30 min at 25 °C in the dark before the Sera-Mag-based workflow as described before [46,47].

2.10. Proteomic Liquid Chromatography-Tandem Mass Spectrometry

Tryptic peptides for one-shot analysis were analyzed on a Dionex UltiMate NCS-3500RSLC nanoUHPLC coupled to a Q-Exactive HF-X hybrid quadrupole-Orbitrap mass spectrometer equipped with nanospray ion source in data-dependent acquisition analysis and positive mode as previously described [46,47,49]. Peptides were loaded (Acclaim PepMap100 C18 3 μm beads with 100 Å pore-size, Thermo Fisher Scientific, Waltham, MA, USA) and separated (1.9 µm particle size C18, 0.075 × 250 mm, Nikkyo Technos Co. Ltd., Tokyo, Japan) with a gradient of 2–28% acetonitrile (Sigma-Aldrich, St. Louis, MO, USA) containing 0.1% formic acid (Sigma-Aldrich, St. Louis, MO, USA), and 28–80% over 56 min at 300 nL min−1 at 55 °C (butterfly portfolio heater, Phoenix S&T, Chester, PA, USA). All MS acquisition parameters were as described before [46,47]. Data were acquired using Xcalibur (v4.0, Thermo Fisher Scientific, Waltham, MA, USA). RAW and parameter/processed proteomics data were deposited to the ProteomeXchange Consortium using the PRIDE partner repository and are available in ProteomeXchange with the identifier (PXD039464).

2.11. Database Search and Analysis

RAW MS data were processed using MaxQuant [50] (v1.6.14.0) with its built-in search engine Andromeda [47]. Tandem mass spectra were searched as a single batch against the Homo Sapien database (UniProt, UP000005640, Jun 2022; 79,684) supplemented with common contaminants. Specific search parameters included: carbamidomethylated cysteine (fixed modification), oxidation of methionine and N-terminal protein acetylation (variable modifications), trypsin/P (C-terminal to arginine/lysine) with ≤2 missed cleavage sites, FDR at PSM/protein at 1%, initial precursor mass deviation (7 ppm), fragment mass deviation (20 ppm), up to two missed cleavages, with all other parameters at default. Match between runs (MBRs) (time window 1 min) and label-free protein quantitation (LFQ) were enabled. False discovery rate (FDR) at the PSM, protein and site levels were each 1%. Peptides were identified with an initial precursor mass deviation of up to 7 ppm and a fragment mass deviation of 20 ppm. A maximum of two missed cleavages were allowed. The 0.01 Protein groups were analyzed using Perseus (v1.6.7), with contaminants and reverse peptides removed. Comparative analysis based on stem cell marker proteins [51] and abundance treemap using proteomap [52] was performed. Perseus was used to analyze the proteomic data and generate plots [53]. G:Profiler, Reactome and STRING were used for Gene Ontology/functional enrichment analysis.

3. Results

3.1. Isolation of a Rich and Pluripotent Population of SBSCs Using a Simplified Approach

SBSCs were isolated from peripheral blood using simple processing steps of centrifugation and filtration that are depicted in Figure 1.
From 30 mL whole blood, our approach resulted in a high concentration of very small cells (about 5 μm), and after staining with trypan blue, over 90% of them were viable (n = 3) (Figure 2A). The mean of the counted live cell population was 2,965,333 cells/mL (SEM: 276,746) of whole blood, and Figure 2B shows a representative field of isolated small stem cells stained with H&E. Black arrows indicate several small stem cells, suggesting that our method can indeed result in high yield isolation.
Furthermore, we investigated the pluripotency of these isolated cells using staining with KP-1. KP-1 staining detects pluripotent cells, as they fluoresce in the green spectrum due to their lack of ABC transporters, a set of proteins that are normally expressed in differentiated cells, eliminating this dye [54]. We observed positive KP-1 staining of the isolated peripheral blood small cells, suggesting that this population contains pluripotent stem cells (Figure 3A).
To further confirm the pluripotency of our small cell population, we next stained them with antibodies against the pluripotent markers Nanog, CXCR4, SSEA-3 and SSEA-4 and the Yamanaka pluripotent factors Oct4, SOX-2, cMyc and KLF4, known to be expressed in embryonic stem cells [55,56,57,58]. The expression of CXCR4, a marker found not only in embryonic stem cells but also in adult differentiated cells, such as intestinal epithelial cells [59], was also examined. Before proceeding with staining the SBSCs, we tested our antibodies on the H1 embryonic stem cell line and found that they were stained positive for all the pluripotent markers (Supplementary Figure S3).
We observed that some of the isolated cells expressed all the studied pluripotent markers (Figure 4), and although we did not perform any quantitative analysis, the expression of SSEA-3, SSEA-4, Nanog and Oct4 were semi-quantitatively determined to be around 10–20%, 10–20%, 5–10% and 20–30%, respectively, based on the acquired immunofluorescence images. This finding suggests that our novel methodology approach results in isolating a rich population of SBSCs in which some are of pluripotent origin.

3.2. SBSCs Express Markers of Mesenchymal Origin

We investigated the mesenchymal characteristics of SBSCs by staining against the known mesenchymal markers, CD29, parathyroid hormone 1 receptor (PTH1R), CD105 and CD106 [60,61,62]. We observed that a portion of SBSCs expressed these markers (Figure 5), and approximately 80%, 90%, 10% and 10% were found positive for CD29, PTH1R, CD105 and CD106, respectively, suggesting that a number of these stem cells may have been differentiated towards mesenchyme lineages.

3.3. SBSCs Express Certain Hematopoietic Markers

We further examined the identity of SBSCs and, in particular, we investigated the possible expression of factors of hematopoietic origin, such as CD90, CD133, CD45 and CD34 [63]. Upon testing, SBSCs were found positive to CD90, CD133 and CD45, but not to CD34 (Figure 6), suggesting that many of the isolated SBSCs were committed towards the hematopoietic lineages.
To exclude any false staining from CD45, we also investigated its expression on WBCs and found that both WBCs and SBSCs were stained positive (Supplementary Figure S4).
We further performed a simultaneous double-stain to SBSCs with antibodies against CD45 and the embryonic stem cell markers, Nanog and SOX-2, and the marker CXCR4, in order to investigate whether both pluripotent and already differentiated stem cells co-exist in this isolated stem cell population. Indeed, we observed a mixed population of SBSCs, as most of them expressed both CD45 and the embryonic stem cell markers Nanog, SOX-2 or CXCR4 (Figure 7, white arrows), while a small portion of them expressed CD45 and expressed weakly or not at all the other markers (Figure 7, red arrows). These results indicate that SBSCs are indeed a mixed population of stem cells that may bear both embryonic and hematopoietic markers.

3.4. SBSC Proteomics

To further investigate the expression profile of SBSCs, we performed proteomics analysis of SBSCs and plasma cells (as a control). SBSCs were cultured in DMEM and a pure cell population, free of debris or blood components, was harvested and analyzed (Figure 8A). Proteomic analysis revealed that SBSCs comprised 809 proteins based on stringent inclusion criteria (quantified LFQ, two or more peptides) (Figure 8B, Supplementary Table S1). Various stem cell marker proteins were identified, including CD9, HSPA4, HSPB1, ITGA6, MAPK1, MTHFD1 and STAT3, that were previously characterized either as stem cell markers or as being significant factors for stem cell maintenance [51]. In addition, we report various transcriptional regulator complex proteins in SBSCs, including STAT3, STAT5B, PDLIM1, ANXA2, ATF6 and CAMK1 (Supplementary Table S1). There was strong enrichment for cytoskeletal and adhesion-related components (tight junctions, focal adhesion), and various signaling pathways including Rap1, MAPK and PI3K-Akt in the SBSC proteome (Figure 8C). To gain further insight into the SBSC proteome, functional enrichment analyses were performed, highlighting enrichment in the SBSC proteome indicating source (blood cell, coagulation, platelet), localization (cell-substrate, actin cytoskeleton, membrane), function (adhesion, binding) and pathways associated with metabolism, adhesion, ECM-receptor interaction, platelet activation and membrane trafficking (Figure 8D–H, Supplementary Table S2).
We further compared the SBSC proteome with isolated plasma cells. We show that the SBSC proteome is distinct in the composition of factors associated with transcriptional regulator complex, and differential proteome analysis (based on LFQ intensity) revealed enrichment in various stem cell markers including CD9, MAPK1 and STAT3 (Figure 9A, Supplementary Table S3). Importantly, various cell membrane surface markers were identified enriched in the SBSC proteome (Figure 9B), including various annexins, integrins and Leukocyte surface antigens. Further analysis of proteins uniquely identified in SBSCs revealed their association with transcription factors (ATF6), the extracellular matrix (COL4A1, COL4A2) and membrane/signaling (HSPG2) (Figure 9C). In addition, functional enrichment analyses of the most abundant proteins in SBSCs (LFQ intensity rank, vs Plasma cell proteome, average LFQ) were performed to highlight enrichment in blood microparticle, integrin signaling, ECM/cell-substrate adhesion and antioxidant activity (Figure 9D).

4. Discussion

In this study, we report a simplified and efficient approach for the isolation of small stem cells (the SBSCs) from human peripheral blood using straightforward steps of centrifugation and filtration of blood, without the use of density gradients and immuno-cell separation methods as previously reported [64,65]. Our approach results in the isolation of a rich population of SBSCs, with various characteristics, as some of them expressed all the widely studied pluripotent stem cell markers (KP-1, Nanog, Oct4, SOX-2, cMyc, KLF4, CXCR4, SSEA-3 and SSEA-4), other certain hematopoietic markers (CD90, CD133 and CD45) and portions of them being mesenchymal markers (CD29, PTH1R, CD105 and CD106). Therefore, we show that the isolated SBSCs consist of a mixed pluripotent population of stem cells that includes a fraction with mesenchymal and hematopoietic characteristics, aligning with previous studies supporting the heterogeneous character of small stem cells in the cord blood [66,67,68].
More specifically, regarding the embryonic stem cell markers, the KP-1, even though not precisely sensitive for pluripotent stem cell detection, has been reported to be a particularly useful tool for screening undifferentiated pluripotent stem cells [54,69,70]. KP-1 has also been used as the selective part for the creation of synthetic hybrid molecules that can target, attach to, and remove unwanted pluripotent stem cells from cell mixtures, to help avoid tumorigenesis [71]. Our approach resulted in the isolation of a pluripotent subpopulation of SBCSs, positively stained with this specific pluripotent marker, KP-1. The other embryonic stem cell markers that were investigated in this study are widely used in stem cell characterization of pluripotency [55,56,57]. In particular, SSEA-3 was proven to be the selective marker to distinguish the Multilineage-differentiating stress-enduring (MUSE) cells, a pluripotent subpopulation that lies among mesenchymal stem cells and fibroblasts [72,73]. SSEA-4 was one of the embryonic markers [74,75] that characterize primate embryonic stem cells and was used in the characterization and establishment of the well-known embryonic stem cell lines derived from human blastocysts (H1, H9) [76]. Nanog has been described as the new “master gene” of embryonic stem cell pluripotency [77,78,79] and was shown to work together with other key pluripotent factors, such as Oct4 [80,81] and SOX-2 [82,83], to control a set of target genes that have important functions in embryonic stem cell pluripotency [84,85,86,87]. CXCR4, the stromal cell-derived factor 1 (SDF-1) chemokine receptor, is expressed in leukocytes, epithelial cells [59,88] and also in embryonic stem cells [89,90]. These findings strongly suggest the pluripotent character of the isolated SBSCs, as they expressed all the aforementioned markers.
In addition, Oct4, KLF4, SOX-2 and c-Myc have been established as the “Yamanaka factors”, as Yamanaka et al. proved that they are crucial key players in the process of creating induced pluripotent stem cells (iPSCs) [21,23,24,91]. According to the suggestions laid out by the International Stem Cell Banking Initiative, there are specific criteria that should be met before banking an iPSC line [92]. Common characterization methods for pluripotency assessment in establishing iPSC lines include the detection of Nanog and Oct4, as well as the TRA-1-60 and TRA-1-81 cell surface antigens, that are expressed along with SSEA-3, SSEA-4 in human embryonic stem cells and embryonal carcinoma cells [24,93]. A portion of our SBSCs was found to be positive for these Yamanaka markers, and the iPSC pluripotency assessment markers indicated that a subpopulation of them has a strong pluripotent identity.
Apart from the aforementioned embryonic stem cell markers, our proteomic analysis revealed that SBCSs express various proteins (CD9, HSPA4, HSPB1, ITGA6, MAPK1, MTHFD1 and STAT3), which have been previously reported either as stem cell markers or as being significant players in stem cell maintenance [51]. CD9 has been found to be expressed in human embryonic stem cells [94,95] and in mesenchymal stem cells, where its expression was correlated with their ability to proliferate and expand [96], suggesting that its expression in SBSCs may be of equal importance for their proliferation and expansion. HSPA4, a heat-shock protein found to be also expressed in embryonic stem cells [97,98], has been shown to have a critical role in spermatogenesis, as its lack of expression led to reduced numbers of spermatozoa that bore apoptotic characteristics [99], suggesting again that the expression of this marker in SBSCs may be significant for their survival. The expression intensity of HSPB1, another heat-shock protein, has been correlated with the differentiation status of stem cells; HSPB1 is overexpressed in undifferentiating stem cells and downregulated in stem cells already committed towards a differentiation pathway [100,101,102]. ITGA6, an integrin α-6 or otherwise known as CD49f, has been shown to be expressed in various stem cells, having a significant role in self-renewal and proliferation [103,104,105]. MAPK1, otherwise known as Erk2, along with Erk1, are two kinases with significant roles in cell proliferation [106,107,108], and their expression has been shown to be crucial for the survival and self-renewal of hematopoietic stem cells [109,110,111]. Regarding embryonic stem cells, various studies have shown that inhibition of the Erk1/2 signaling pathway is essential for their self-renewal [112,113,114]. MTHFD1 is an enzyme involved in folate metabolism [115,116,117], and although it was reported as a potential stem cell marker by Maguire et al. and Bhattacharya et al. [51,118], there is no other knowledge in regard to its role in stem cell pluripotency, while its polymorphisms have been associated with therapeutic response to methotrexate in childhood acute lymphoblastic leukemia [119,120,121]. STAT3 is well known as a transcription factor for its role in the regulation of the self-renewal of embryonic stem cells [122,123,124], suggesting that its expression in SBSCs may indeed promote their self-renewal.
Identified factors in SBSCs further included proteins associated with cell differentiation. These included LIMS1, HSP90AB1, EIF5A, LYN, S100A4, PA2G4, FYN, CAMK1 and PTBP1, and most of them have an already known role in stem cell differentiation, as seen in Table 2.
Our proteome data also supports the identification (and enrichment to control plasma-derived cells) of various factors highly enriched in SBSCs associated with cell adhesion, matrix localization, antioxidant function and cytoskeleton localization, suggesting that SBSCs might have the potential to migrate through the bloodstream and attach to specific tissues and differentiate towards various cell types. In addition, various signaling pathways including Rap1, MAPK and PI3K-Akt were identified as enriched in SBSCs (Figure 8), as well as integrin signaling factors/pathways highly abundant in SBSCs in comparison to isolated blood cells (Figure 9). These signaling pathways have been previously reported to be also highly enriched in mesenchymal stem cells [168], and more specifically, the MAPK signaling pathway has been found to promote neuronal stem cell differentiation [169]. Rap1 has been shown to regulate the fate of calvarial mesenchymal cells, as it promotes chondrogenesis, while its depletion results in osteogenesis [170]. The PI3K-Akt signaling pathway has been reported to be involved in the differentiation process of mesenchymal cells toward the osteogenic lineage [171].
Regarding the widely studied markers for mesenchymal stem cells, CD29 and CD105 [172,173,174], portions of SBSCs were found to be positive for both, indicating that they may be prone toward mesenchymal lineage differentiation. On the other hand, CD106, which was also found to be expressed in a small portion of SBSCs, is a marker primarily expressed in endothelial cells [175] and secondarily in some cases of mesenchymal stem cells [62,173,176], suggesting that a subpopulation of SBSCs may be committed either towards endothelial cells or specific types of mesenchymal stem cells.
Furthermore, a subpopulation of SBSCs was also found to stain for membrane-bound PTH1R. It was previously proposed that VSELs may have an important role in bone metabolism and hence, it is plausible to suggest that they may express parathyroid hormone (PTH) receptors [177]. PTH receptors have been shown to be expressed in mesenchymal stem cells [178,179,180] and are thought to be important in driving the anabolic effect on the bone that has been observed with intermittent PTH and parathyroid hormone-related protein (PTHrP) agonists [177,181,182]. The fact that these cells seem to be of more primitive origin than mesenchymal stem cells, due to their size, large quantity, and pluripotent nature, suggests that SBSCs may be of paramount importance in bone metabolism.
Finally, the hematopoietic marker CD90, a surface marker of different types of cells, including T cells and mesenchymal stem cells [183,184,185], was expressed in SBSCs, suggesting again that these cells may have a mixed phenotype. Regarding CD133, although it was initially described as a stem cell marker of hematopoietic origin [186,187,188], it was later used for the identification of VSELs [67,189,190]. Our results agree with the aforementioned report on VSEL characterization, as SBSCs were also found to be CD133 positive. In contrast to previous studies on VSEL characterization using CD45 and CD34 markers [34,189], in our study, SBSCs were found to be CD45 positive, but CD34 negative, suggesting that our method may result in the isolation of a different stem cell population. Similar findings to our results come from the research study by Virant-Klun et al. on small ovarian stem cells found on the ovarian surface epithelium of postmenopausal women with no naturally present oocytes and follicles. These small ovarian stem cells expressed the pluripotent markers SSEA-4, Oct-4, Nanog and Sox-2, and, in addition, they were detected to be also positive for c-kit, a receptor implicated in hematopoiesis and gametogenesis, suggesting that although pluripotent, these cells were also committed to specific lineages [191]. Finally, our results agree with a recent study that identified a novel population of stem cells (termed MUSE cells) isolated from peripheral blood that simultaneously express SSEA-3 and CD45 [192]. Although these MUSE cells were 10 µm, this leads further to the notion that indeed there is a primitive subpopulation of small stem cells that bears both pluripotent, mesenchymal and hematopoietic stem cell markers. On the other hand, all these findings on the different stem cell populations with different expressing markers, such as hematopoietic or mesenchymal ones, might suggest that there are circulating stem cell subpopulations at different stages of differentiation depending on the organism’s needs. For instance, Parte et al. found that there are two distinctive stem cell populations in the ovarian surface epithelium that differ in size, embryonic marker site expression and differentiation potential [193].
Apart from the various expressed markers, SBSCs appeared to differ from hematopoietic stem cells (HSCs) in terms of other characteristics as well. One of these characteristics was their population size found in peripheral blood; while most HSCs are increased in the bone marrow and limited in the peripheral blood [194,195,196], the SBSCs population seemed to be high in number in the peripheral blood. Another characteristic in regards to their size, SBSCs seem to be smaller than 5 μm, while the size of HSCs usually varies from 6 to 10 μm [197,198,199]. Finally, regarding their nucleus-to-cytoplasm ratio, SBSCs appear to have very little cytoplasm with nuclear Oct-4 staining, as shown in our study, while HSCs have larger cytoplasm and cytoplasmic Oct-4 staining [200].
Finally, although our results indicate that the isolated SBSCs express various pluripotent markers, suggesting that they are a mixed stem cell population, further characterization is required. Flow cytometry analysis and cell sorting would be an interesting approach, as these methods would not only further characterize this cell population, but could also compare the SBSCs’ expression profile with that of other stem and non-stem cells and provide information about the subpopulations’ actual cell number size. Apart from their protein content, another significant approach would be to analyze and compare their transcriptomic profile with other differentiated and non-differentiated cell populations, and this could be achieved using next-generation sequencing methods.

5. Conclusions

In this study, we presented an easy process to isolate small peripheral blood pluripotent stem cells with a strong pluripotent stem cell identity, while also sharing certain mesenchymal and hematopoietic characteristics. Although this population was morphologically similar to the VSELs, they were characterized as SBSCs due to the expression of different markers, as well as having different sizes and isolation methods. Proteomic analyses provided not only a composition analysis of SBSCs (stem cell markers CD9, MAPK1, STAT3, cell surface markers and signaling molecules) but also showed how specific factors were enriched in comparison to control blood cells. Currently, almost every VSEL isolation method requires the use of density gradients and erythrocyte lysis reagents that ultimately result in few isolated stem cells, while our approach uses only two centrifugation steps and no extra reagents and results in a rich stem cell population. VSELs may be a subgroup of SBSCs as SBSCs were found in higher quantity and mostly CD45 positive; however, further studies on the role and nature of SBSCs are crucial. The findings of the present study do not explain the functional role of SBSCs; whether they are a stand-alone cell population or a population of early tissue progenitor cells in blood that move from blood to tissue, and vice versa, is the subject of future work. Additional work is also necessary to determine their self-renewal and differentiating capacity into ectodermal, endodermal and mesodermal tissues and to examine their possible role in regenerative medicine applications. Furthermore, in terms of future diagnostic and therapeutic use, which is the main focus in our laboratory, it is plausible to suggest that these cells have the potential to be a natural source of adult pluripotent stem cells that could fulfill the need for an unlimited supply of autologous pluripotent stem cells that could be manufactured as end-products for personalized diagnostics and immune-rejection free autologous therapeutics.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/biomedicines11030787/s1, Figure S1: Staining with isotype controls; Figure S2: SBSCs stained against the various targets of interest; Figure S3: H1 embryonic stem cell line stained for embryonic stem cell markers; Figure S4: SBSCs and WBC stained for CD45; Table S1: SBSC proteome; Table S2: SBSC functional enrichment analysis; Table S3: SBSC and control plasma cell proteome comparison.

Author Contributions

Conceptualization, E.F., L.K., V.P. and G.K.; methodology, E.F., L.K., G.T., M.S., C.S., A.R., D.W.G. and K.A.; formal analysis and investigation, E.F. and L.K.; writing—original draft preparation, E.F. and L.K.; writing—review and editing, V.P. and G.K.; supervision, V.P. and G.K. All authors reviewed the manuscript. E.F. and L.K. contributed equally to this work. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

This study was conducted in accordance with the Declaration of Helsinki, and approved by the Research Ethics Committee of University Hospital of Alexandroupolis (Protocol number Θ9/Δ.Σ37/21-12-2018).

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

The data used to support the findings of this study are available from the corresponding author upon request.

Conflicts of Interest

V.P. holds a patent on the SBSC isolation process described in this study.

References

  1. Suman, S.; Domingues, A.; Ratajczak, J.; Ratajczak, M.Z. Potential Clinical Applications of Stem Cells in Regenerative Medicine. Adv. Exp. Med. Biol. 2019, 1201, 1–22. [Google Scholar] [CrossRef]
  2. Jarrige, M.; Frank, E.; Herardot, E.; Martineau, S.; Darle, A.; Benabides, M.; Domingues, S.; Chose, O.; Habeler, W.; Lorant, J.; et al. The Future of Regenerative Medicine: Cell Therapy Using Pluripotent Stem Cells and Acellular Therapies Based on Extracellular Vesicles. Cells 2021, 10, 240. [Google Scholar] [CrossRef] [PubMed]
  3. Bajada, S.; Mazakova, I.; Richardson, J.B.; Ashammakhi, N. Updates on stem cells and their applications in regenerative medicine. J. Tissue Eng. Regen. Med. 2008, 2, 169–183. [Google Scholar] [CrossRef] [PubMed]
  4. Kolios, G.; Moodley, Y. Introduction to stem cells and regenerative medicine. Respiration 2013, 85, 3–10. [Google Scholar] [CrossRef]
  5. Ilic, D.; Polak, J.M. Stem cells in regenerative medicine: Introduction. Br. Med. Bull. 2011, 98, 117–126. [Google Scholar] [CrossRef] [Green Version]
  6. Hipp, J.; Atala, A. Sources of Stem Cells for Regenerative Medicine. Stem Cell Rev. 2008, 4, 3–11. [Google Scholar] [CrossRef]
  7. Zakrzewski, W.; Dobrzyński, M.; Szymonowicz, M.; Rybak, Z. Stem cells: Past, present, and future. Stem Cell Res. Ther. 2019, 10, 68. [Google Scholar] [CrossRef]
  8. Triffitt, J.T. Stem cells and the philosopher’s stone. J. Cell. Biochem. Suppl. 2002, 38, 13–19. [Google Scholar] [CrossRef]
  9. Bongso, A.; Richards, M. History and perspective of stem cell research. Best Pract. Res. Clin. Obstet. Gynaecol. 2004, 18, 827–842. [Google Scholar] [CrossRef]
  10. Ntege, E.H.; Sunami, H.; Shimizu, Y. Advances in regenerative therapy: A review of the literature and future directions. Regen. Ther. 2020, 14, 136–153. [Google Scholar] [CrossRef]
  11. Graf, T.; Stadtfeld, M. Heterogeneity of Embryonic and Adult Stem Cells. Cell Stem Cell 2008, 3, 480–483. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Passier, R.; Mummery, C. Origin and use of embryonic and adult stem cells in differentiation and tissue repair. Cardiovasc. Res. 2003, 58, 324–335. [Google Scholar] [CrossRef] [PubMed]
  13. Aly, R.M. Current state of stem cell-based therapies: An overview. Stem Cell Investig. 2020, 7, 8. [Google Scholar] [CrossRef]
  14. Doss, M.X.; Koehler, C.I.; Gissel, C.; Hescheler, J.; Sachinidis, A. Embryonic stem cells: A promising tool for cell replacement therapy. J. Cell. Mol. Med. 2004, 8, 465–473. [Google Scholar] [CrossRef] [PubMed]
  15. Menendez, P.; Bueno, C.; Wang, L. Human embryonic stem cells: A journey beyond cell replacement therapies. Cytotherapy 2006, 8, 530–541. [Google Scholar] [CrossRef]
  16. De Luca, M.; Aiuti, A.; Cossu, G.; Parmar, M.; Pellegrini, G.; Robey, P.G. Advances in stem cell research and therapeutic development. Nat. Cell Biol. 2019, 21, 801–811. [Google Scholar] [CrossRef]
  17. Volarevic, V.; Markovic, B.S.; Gazdic, M.; Volarevic, A.; Jovicic, N.; Arsenijevic, N.; Armstrong, L.; Djonov, V.; Lako, M.; Stojkovic, M. Ethical and Safety Issues of Stem Cell-Based Therapy. Int. J. Med. Sci. 2018, 15, 36–45. [Google Scholar] [CrossRef] [Green Version]
  18. Brunet, A.; Goodell, M.A.; Rando, T.A. Ageing and rejuvenation of tissue stem cells and their niches. Nat. Rev. Mol. Cell Biol. 2023, 24, 45–62. [Google Scholar] [CrossRef]
  19. Sobhani, A.; Khanlarkhani, N.; Baazm, M.; Mohammadzadeh, F.; Najafi, A.; Mehdinejadiani, S.; Aval, F.S. Multipotent stem cell and current application. Acta Med. Iran. 2017, 55, 6–23. [Google Scholar]
  20. Mirzaei, H.; Sahebkar, A.; Sichani, L.S.; Moridikia, A.; Nazari, S.; Sadri Nahand, J.; Salehi, H.; Stenvang, J.; Masoudifar, A.; Mirzaei, H.R.; et al. Therapeutic application of multipotent stem cells. J. Cell. Physiol. 2018, 233, 2815–2823. [Google Scholar] [CrossRef]
  21. Takahashi, K.; Yamanaka, S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 2006, 126, 663–676. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Takahashi, K.; Yamanaka, S. Induced pluripotent stem cells in medicine and biology. Development 2013, 140, 2457–2461. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Mostoslavsky, G. Concise Review: The Magic Act of Generating Induced Pluripotent Stem Cells: Many Rabbits in the Hat. Stem Cells 2011, 30, 28–32. [Google Scholar] [CrossRef] [PubMed]
  24. Maherali, N.; Hochedlinger, K. Guidelines and Techniques for the Generation of Induced Pluripotent Stem Cells. Cell Stem Cell 2008, 3, 595–605. [Google Scholar] [CrossRef] [Green Version]
  25. Singh, V.K.; Kalsan, M.; Kumar, N.; Saini, A.; Chandra, R. Induced pluripotent stem cells: Applications in regenerative medicine, disease modeling, and drug discovery. Front. Cell Dev. Biol. 2015, 3, 2. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Omole, A.E.; Fakoya, A.O.J. Ten years of progress and promise of induced pluripotent stem cells: Historical origins, characteristics, mechanisms, limitations, and potential applications. PeerJ 2018, 6, e4370. [Google Scholar] [CrossRef] [Green Version]
  27. Neofytou, E.; O’Brien, C.G.; Couture, L.A.; Wu, J.C. Hurdles to clinical translation of human induced pluripotent stem cells. J. Clin. Investig. 2015, 125, 2551–2557. [Google Scholar] [CrossRef] [Green Version]
  28. Madrid, M.; Sumen, C.; Aivio, S.; Saklayen, N. Autologous Induced Pluripotent Stem Cell–Based Cell Therapies: Promise, Progress, and Challenges. Curr. Protoc. 2021, 1, e88. [Google Scholar] [CrossRef]
  29. Shi, Y.; Inoue, H.; Wu, J.C.; Yamanaka, S. Induced pluripotent stem cell technology: A decade of progress. Nat. Rev. Drug Discov. 2017, 16, 115–130. [Google Scholar] [CrossRef]
  30. Taylor, C.J.; Bolton, E.M.; Bradley, J.A. Immunological considerations for embryonic and induced pluripotent stem cell banking. Philos. Trans. R. Soc. B: Biol. Sci. 2011, 366, 2312–2322. [Google Scholar] [CrossRef] [Green Version]
  31. Azuma, K.; Yamanaka, S. Recent policies that support clinical application of induced pluripotent stem cell-based regenerative therapies. Regen. Ther. 2016, 4, 36–47. [Google Scholar] [CrossRef] [Green Version]
  32. Sullivan, S.; Fairchild, P.J.; Marsh, S.G.E.; Müller, C.R.; Turner, M.L.; Song, J.; Turner, D. Haplobanking induced pluripotent stem cells for clinical use. Stem Cell Res. 2020, 49, 102035. [Google Scholar] [CrossRef]
  33. Li, C.; Chen, S.; Zhou, Y.; Zhao, Y.; Liu, P.; Cai, J. Application of induced pluripotent stem cell transplants: Autologous or allogeneic? Life Sci. 2018, 212, 145–149. [Google Scholar] [CrossRef] [PubMed]
  34. Kucia, M.; Reca, R.; Campbell, F.R.; Zuba-Surma, E.; Majka, M.; Ratajczak, J.; Ratajczak, M.Z. A population of very small embryonic-like (VSEL) CXCR4(+)SSEA-1(+)Oct-4+ stem cells identified in adult bone marrow. Leukemia 2006, 20, 857–869. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Shin, D.M.; Liu, R.; Wu, W.; Waigel, S.J.; Zacharias, W.; Ratajczak, M.Z.; Kucia, M. Global gene expression analysis of very small embryonic-like stem cells reveals that the Ezh2-dependent bivalent domain mechanism contributes to their pluripotent state. Stem Cells Dev. 2012, 21, 1639–1652. [Google Scholar] [CrossRef] [Green Version]
  36. Ratajczak, M.Z.; Ratajczak, J.; Suszynska, M.; Miller, D.M.; Kucia, M.; Shin, D.M. A Novel View of the Adult Stem Cell Compartment From the Perspective of a Quiescent Population of Very Small Embryonic-Like Stem Cells. Circ. Res. 2017, 120, 166–178. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Ratajczak, M.Z.; Ratajczak, J.; Kucia, M. Very small embryonic-like stem cells (VSELs) an update and future directions. Circ. Res. 2019, 124, 208–210. [Google Scholar] [CrossRef] [PubMed]
  38. Ratajczak, M.Z.; Zuba-Surma, E.; Wojakowski, W.; Suszynska, M.; Mierzejewska, K.; Liu, R.; Ratajczak, J.; Shin, D.M.; Kucia, M. Very small embryonic-like stem cells (VSELs) represent a real challenge in stem cell biology: Recent pros and cons in the midst of a lively debate. Leukemia 2014, 28, 473–484. [Google Scholar] [CrossRef] [Green Version]
  39. Weissman, I.L. Stem cells: Units of development, units of regeneration, and units in evolution. Cell 2000, 100, 157–168. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Kuruca, S.E.; Çelik, D.D.; Özerkan, D.; Erdemir, G. Characterization and Isolation of Very Small Embryonic-like (VSEL) Stem Cells Obtained from Various Human Hematopoietic Cell Sources. Stem Cell Rev. Rep. 2019, 15, 730–742. [Google Scholar] [CrossRef]
  41. Gounari, E.; Daniilidis, A.; Tsagias, N.; Michopoulou, A.; Kouzi, K.; Koliakos, G. Isolation of a novel embryonic stem cell cord blood-derived population with in vitro hematopoietic capacity in the presence of Wharton’s jelly-derived mesenchymal stromal cells. Cytotherapy 2019, 21, 246–259. [Google Scholar] [CrossRef] [PubMed]
  42. Monti, M.; Imberti, B.; Bianchi, N.; Pezzotta, A.; Morigi, M.; Del Fante, C.; Redi, C.A.; Perotti, C. A Novel Method for Isolation of Pluripotent Stem Cells from Human Umbilical Cord Blood. Stem Cells Dev. 2017, 26, 1258–1269. [Google Scholar] [CrossRef]
  43. Bamias, G.; Filidou, E.; Goukos, D.; Valatas, V.; Arvanitidis, K.; Panagopoulou, M.; Kouklakis, G.; Daikos, G.L.; Ladas, S.D.; Kolios, G. Crohn’s disease-associated mucosal factors regulate the expression of TNF-like cytokine 1A and its receptors in primary subepithelial intestinal myofibroblasts and intestinal epithelial cells. Transl. Res. J. Lab. Clin. Med. 2017, 180, 118–130.e112. [Google Scholar] [CrossRef] [PubMed]
  44. Filidou, E.; Valatas, V.; Drygiannakis, I.; Arvanitidis, K.; Vradelis, S.; Kouklakis, G.; Kolios, G.; Bamias, G. Cytokine receptor profiling in human colonic subepithelial myofibroblasts: A differential effect of Th polarization associated cytokines in intestinal fibrosis. Inflamm. Bowel Dis. 2018, 24, 2224–2241. [Google Scholar] [CrossRef]
  45. Kandilogiannakis, L.; Filidou, E.; Drygiannakis, I.; Tarapatzi, G.; Didaskalou, S.; Koffa, M.; Arvanitidis, K.; Bamias, G.; Valatas, V.; Paspaliaris, V.; et al. Development of a Human Intestinal Organoid Model for In Vitro Studies on Gut Inflammation and Fibrosis. Stem Cells Int. 2021, 2021, 9929461. [Google Scholar] [CrossRef]
  46. Claridge, B.; Rai, A.; Fang, H.; Matsumoto, A.; Luo, J.; McMullen, J.R.; Greening, D.W. Proteome characterisation of extracellular vesicles isolated from heart. Proteomics 2021, 21, e2100026. [Google Scholar] [CrossRef]
  47. Lozano, J.; Rai, A.; Lees, J.G.; Fang, H.; Claridge, B.; Lim, S.Y.; Greening, D.W. Scalable Generation of Nanovesicles from Human-Induced Pluripotent Stem Cells for Cardiac Repair. Int. J. Mol. Sci. 2022, 23, 14334. [Google Scholar] [CrossRef]
  48. Hughes, C.S.; Moggridge, S.; Muller, T.; Sorensen, P.H.; Morin, G.B.; Krijgsveld, J. Single-pot, solid-phase-enhanced sample preparation for proteomics experiments. Nat. Protoc. 2019, 14, 68–85. [Google Scholar] [CrossRef]
  49. Kompa, A.R.; Greening, D.W.; Kong, A.M.; McMillan, P.J.; Fang, H.; Saxena, R.; Wong, R.C.B.; Lees, J.G.; Sivakumaran, P.; Newcomb, A.E.; et al. Sustained subcutaneous delivery of secretome of human cardiac stem cells promotes cardiac repair following myocardial infarction. Cardiovasc. Res. 2021, 117, 918–929. [Google Scholar] [CrossRef] [PubMed]
  50. Cox, J.; Mann, M. MaxQuant enables high peptide identification rates, individualized p.p.b.-range mass accuracies and proteome-wide protein quantification. Nat. Biotechnol. 2008, 26, 1367–1372. [Google Scholar] [CrossRef] [PubMed]
  51. Maguire, C.T.; Demarest, B.L.; Hill, J.T.; Palmer, J.D.; Brothman, A.R.; Yost, H.J.; Condic, M.L. Genome-wide analysis reveals the unique stem cell identity of human amniocytes. PLoS ONE 2013, 8, e53372. [Google Scholar] [CrossRef] [Green Version]
  52. Liebermeister, W.; Noor, E.; Flamholz, A.; Davidi, D.; Bernhardt, J.; Milo, R. Visual account of protein investment in cellular functions. Proc. Natl. Acad. Sci. USA 2014, 111, 8488–8493. [Google Scholar] [CrossRef] [Green Version]
  53. Tyanova, S.; Cox, J. Perseus: A Bioinformatics Platform for Integrative Analysis of Proteomics Data in Cancer Research. Methods Mol. Biol. 2018, 1711, 133–148. [Google Scholar] [CrossRef] [Green Version]
  54. Hirata, N.; Nakagawa, M.; Fujibayashi, Y.; Yamauchi, K.; Murata, A.; Minami, I.; Tomioka, M.; Kondo, T.; Kuo, T.F.; Endo, H.; et al. A chemical probe that labels human pluripotent stem cells. Cell Rep. 2014, 6, 1165–1174. [Google Scholar] [CrossRef] [Green Version]
  55. Miller, R.J.; Banisadr, G.; Bhattacharyya, B.J. CXCR4 signaling in the regulation of stem cell migration and development. J. Neuroimmunol. 2008, 198, 31–38. [Google Scholar] [CrossRef] [Green Version]
  56. Galkowski, D.; Ratajczak, M.Z.; Kocki, J.; Darzynkiewicz, Z. Of Cytometry, Stem Cells and Fountain of Youth. Stem Cell Rev. Rep. 2017, 13, 465–481. [Google Scholar] [CrossRef] [PubMed]
  57. Wright, A.J.; Andrews, P.W. Surface marker antigens in the characterization of human embryonic stem cells. Stem Cell Res. 2009, 3, 3–11. [Google Scholar] [CrossRef] [Green Version]
  58. Takahashi, K.; Tanabe, K.; Ohnuki, M.; Narita, M.; Ichisaka, T.; Tomoda, K.; Yamanaka, S. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 2007, 131, 861–872. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Jordan, N.J.; Kolios, G.; Abbot, S.E.; Sinai, M.A.; Thompson, D.A.; Petraki, K.; Westwick, J. Expression of functional CXCR4 chemokine receptors on human colonic epithelial cells. J. Clin. Investig. 1999, 104, 1061–1069. [Google Scholar] [CrossRef] [Green Version]
  60. Brown, C.; McKee, C.; Bakshi, S.; Walker, K.; Hakman, E.; Halassy, S.; Svinarich, D.; Dodds, R.; Govind, C.K.; Chaudhry, G.R. Mesenchymal stem cells: Cell therapy and regeneration potential. J. Tissue Eng. Regen. Med. 2019, 13, 1738–1755. [Google Scholar] [CrossRef] [PubMed]
  61. Zhang, Y.; Kumagai, K.; Saito, T. Effect of parathyroid hormone on early chondrogenic differentiation from mesenchymal stem cells. J. Orthop. Surg. Res. 2014, 9, 68. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Mo, M.; Wang, S.; Zhou, Y.; Li, H.; Wu, Y. Mesenchymal stem cell subpopulations: Phenotype, property and therapeutic potential. Cell. Mol. Life Sci. 2016, 73, 3311–3321. [Google Scholar] [CrossRef]
  63. Tarnok, A.; Ulrich, H.; Bocsi, J. Phenotypes of stem cells from diverse origin. Cytom. A 2010, 77, 6–10. [Google Scholar] [CrossRef]
  64. Chang, Y.J.; Tien, K.E.; Wen, C.H.; Hsieh, T.B.; Hwang, S.M. Recovery of CD45(-)/Lin(-)/SSEA-4(+) very small embryonic-like stem cells by cord blood bank standard operating procedures. Cytotherapy 2014, 16, 560–565. [Google Scholar] [CrossRef] [PubMed]
  65. Halasa, M.; Baskiewicz-Masiuk, M.; Dabkowska, E.; Machalinski, B. An efficient two-step method to purify very small embryonic-like (VSEL) stem cells from umbilical cord blood (UCB). Folia Histochem. Cytobiol. 2008, 46, 239–243. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Alvarez-Gonzalez, C.; Duggleby, R.; Vagaska, B.; Querol, S.; Gomez, S.G.; Ferretti, P.; Madrigal, A. Cord blood Lin(-)CD45(-) embryonic-like stem cells are a heterogeneous population that lack self-renewal capacity. PLoS ONE 2013, 8, e67968. [Google Scholar] [CrossRef] [Green Version]
  67. Shaikh, A.; Nagvenkar, P.; Pethe, P.; Hinduja, I.; Bhartiya, D. Molecular and phenotypic characterization of CD133 and SSEA4 enriched very small embryonic-like stem cells in human cord blood. Leukemia 2015, 29, 1909–1917. [Google Scholar] [CrossRef] [PubMed]
  68. Bhartiya, D.; Jha, N.; Tripathi, A.; Tripathi, A. Very small embryonic-like stem cells have the potential to win the three-front war on tissue damage, cancer, and aging. Front. Cell Dev. Biol. 2022, 10, 1061022. [Google Scholar] [CrossRef]
  69. Kuo, T.F.; Mao, D.; Hirata, N.; Khambu, B.; Kimura, Y.; Kawase, E.; Shimogawa, H.; Ojika, M.; Nakatsuji, N.; Ueda, K.; et al. Selective elimination of human pluripotent stem cells by a marine natural product derivative. J. Am. Chem. Soc. 2014, 136, 9798–9801. [Google Scholar] [CrossRef] [PubMed]
  70. Miyagi-Shiohira, C.; Saitoh, I.; Watanabe, M.; Noguchi, H. Kyoto probe-1 reveals phenotypic differences between mouse ES cells and iTS-P cells. Sci. Rep. 2020, 10, 18084. [Google Scholar] [CrossRef]
  71. Mao, D.; Ando, S.; Sato, S.I.; Qin, Y.; Hirata, N.; Katsuda, Y.; Kawase, E.; Kuo, T.F.; Minami, I.; Shiba, Y.; et al. A Synthetic Hybrid Molecule for the Selective Removal of Human Pluripotent Stem Cells from Cell Mixtures. Angew. Chem. Int. Ed. Engl. 2017, 56, 1765–1770. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Wakao, S.; Kitada, M.; Kuroda, Y.; Shigemoto, T.; Matsuse, D.; Akashi, H.; Tanimura, Y.; Tsuchiyama, K.; Kikuchi, T.; Goda, M.; et al. Multilineage-differentiating stress-enduring (Muse) cells are a primary source of induced pluripotent stem cells in human fibroblasts. Proc. Natl. Acad. Sci. USA 2011, 108, 9875–9880. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Kuroda, Y.; Kitada, M.; Wakao, S.; Nishikawa, K.; Tanimura, Y.; Makinoshima, H.; Goda, M.; Akashi, H.; Inutsuka, A.; Niwa, A.; et al. Unique multipotent cells in adult human mesenchymal cell populations. Proc. Natl. Acad. Sci. USA 2010, 107, 8639–8643. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Zhao, W.; Ji, X.; Zhang, F.; Li, L.; Ma, L. Embryonic Stem Cell Markers. Molecules 2012, 17, 6196–6236. [Google Scholar] [CrossRef]
  75. Breimer, M.E.; Säljö, K.; Barone, A.; Teneberg, S. Glycosphingolipids of human embryonic stem cells. Glycoconj. J. 2017, 34, 713–723. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Thomson, J.A.; Itskovitz-Eldor, J.; Shapiro, S.S.; Waknitz, M.A.; Swiergiel, J.J.; Marshall, V.S.; Jones, J.M. Embryonic stem cell lines derived from human blastocysts. Science 1998, 282, 1145–1147. [Google Scholar] [CrossRef] [Green Version]
  77. Pan, G.; Thomson, J.A. Nanog and transcriptional networks in embryonic stem cell pluripotency. Cell Res. 2007, 17, 42–49. [Google Scholar] [CrossRef]
  78. Cavaleri, F.; Schöler, H.R. Nanog: A New Recruit to the Embryonic Stem Cell Orchestra. Cell 2003, 113, 551–552. [Google Scholar] [CrossRef] [Green Version]
  79. Blinka, S.; Rao, S. Nanog Expression in Embryonic Stem Cells—An Ideal Model System to Dissect Enhancer Function. Bioessays 2017, 39, 86. [Google Scholar] [CrossRef] [Green Version]
  80. Lee, J.; Go, Y.; Kang, I.; Han, Y.-M.; Kim, J. Oct-4 controls cell-cycle progression of embryonic stem cells. Biochem. J. 2010, 426, 171–181. [Google Scholar] [CrossRef] [Green Version]
  81. Pesce, M.; Anastassiadis, K.; Schöler, H.R. Oct-4: Lessons of Totipotency from Embryonic Stem Cells. Cells Tissues Organs 1999, 165, 144–152. [Google Scholar] [CrossRef]
  82. Zhang, S.; Cui, W. Sox2, a key factor in the regulation of pluripotency and neural differentiation. World J. Stem Cells 2014, 6, 305–311. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Schaefer, T.; Steiner, R.; Lengerke, C. SOX2 and p53 Expression Control Converges in PI3K/AKT Signaling with Versatile Implications for Stemness and Cancer. Int. J. Mol. Sci. 2020, 21, 4902. [Google Scholar] [CrossRef] [PubMed]
  84. Boyer, L.A.; Lee, T.I.; Cole, M.F.; Johnstone, S.E.; Levine, S.S.; Zucker, J.P.; Guenther, M.G.; Kumar, R.M.; Murray, H.L.; Jenner, R.G. Core transcriptional regulatory circuitry in human embryonic stem cells. Cell 2005, 122, 947–956. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Wang, J.; Rao, S.; Chu, J.; Shen, X.; Levasseur, D.N.; Theunissen, T.W.; Orkin, S.H. A protein interaction network for pluripotency of embryonic stem cells. Nature 2006, 444, 364–368. [Google Scholar] [CrossRef]
  86. Chen, X.; Xu, H.; Yuan, P.; Fang, F.; Huss, M.; Vega, V.B.; Wong, E.; Orlov, Y.L.; Zhang, W.; Jiang, J. Integration of external signaling pathways with the core transcriptional network in embryonic stem cells. Cell 2008, 133, 1106–1117. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Rao, S.; Zhen, S.; Roumiantsev, S.; McDonald, L.T.; Yuan, G.-C.; Orkin, S.H. Differential roles of Sall4 isoforms in embryonic stem cell pluripotency. Mol. Cell. Biol. 2010, 30, 5364–5380. [Google Scholar] [CrossRef] [Green Version]
  88. Ward, S.G.; Bacon, K.; Westwick, J. Chemokines and T lymphocytes: More than an attraction. Immunity 1998, 9, 1–11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. McGrath, K.E.; Koniski, A.D.; Maltby, K.M.; McGann, J.K.; Palis, J. Embryonic expression and function of the chemokine SDF-1 and its receptor, CXCR4. Dev. Biol. 1999, 213, 442–456. [Google Scholar] [CrossRef] [Green Version]
  90. Cencioni, C.; Capogrossi, M.C.; Napolitano, M. The SDF-1/CXCR4 axis in stem cell preconditioning. Cardiovasc. Res. 2012, 94, 400–407. [Google Scholar] [CrossRef] [Green Version]
  91. Karagiannis, P.; Takahashi, K.; Saito, M.; Yoshida, Y.; Okita, K.; Watanabe, A.; Inoue, H.; Yamashita, J.K.; Todani, M.; Nakagawa, M.; et al. Induced Pluripotent Stem Cells and Their Use in Human Models of Disease and Development. Physiol. Rev. 2019, 99, 79–114. [Google Scholar] [CrossRef] [PubMed]
  92. Crook, J.M.; Hei, D.; Stacey, G. The International Stem Cell Banking Initiative (ISCBI): Raising standards to bank on. Vitr. Cell. Dev. Biol. Anim. 2010, 46, 169–172. [Google Scholar] [CrossRef] [PubMed]
  93. Huang, C.Y.; Liu, C.L.; Ting, C.Y.; Chiu, Y.T.; Cheng, Y.C.; Nicholson, M.W.; Hsieh, P.C.H. Human iPSC banking: Barriers and opportunities. J. Biomed. Sci. 2019, 26, 87. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Kolle, G.; Ho, M.; Zhou, Q.; Chy, H.S.; Krishnan, K.; Cloonan, N.; Bertoncello, I.; Laslett, A.L.; Grimmond, S.M. Identification of human embryonic stem cell surface markers by combined membrane-polysome translation state array analysis and immunotranscriptional profiling. Stem Cells 2009, 27, 2446–2456. [Google Scholar] [CrossRef] [PubMed]
  95. Oka, M.; Tagoku, K.; Russell, T.L.; Nakano, Y.; Hamazaki, T.; Meyer, E.M.; Yokota, T.; Terada, N. CD9 is associated with leukemia inhibitory factor-mediated maintenance of embryonic stem cells. Mol. Biol. Cell 2002, 13, 1274–1281. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Kim, Y.J.; Yu, J.M.; Joo, H.J.; Kim, H.K.; Cho, H.H.; Bae, Y.C.; Jung, J.S. Role of CD9 in proliferation and proangiogenic action of human adipose-derived mesenchymal stem cells. Pflug. Arch. 2007, 455, 283–296. [Google Scholar] [CrossRef]
  97. Prinsloo, E.; Setati, M.M.; Longshaw, V.M.; Blatch, G.L. Chaperoning stem cells: A role for heat shock proteins in the modulation of stem cell self-renewal and differentiation? BioEssays 2009, 31, 370–377. [Google Scholar] [CrossRef] [PubMed]
  98. Fan, G.-C. Role of heat shock proteins in stem cell behavior. Prog. Mol. Biol. Transl. Sci. 2012, 111, 305–322. [Google Scholar] [PubMed] [Green Version]
  99. Held, T.; Barakat, A.Z.; Mohamed, B.A.; Paprotta, I.; Meinhardt, A.; Engel, W.; Adham, I.M. Heat-shock protein HSPA4 is required for progression of spermatogenesis. Reproduction 2011, 142, 133–144. [Google Scholar] [CrossRef] [Green Version]
  100. Osman, A.M.; van Dartel, D.A.; Zwart, E.; Blokland, M.; Pennings, J.L.; Piersma, A.H. Proteome profiling of mouse embryonic stem cells to define markers for cell differentiation and embryotoxicity. Reprod. Toxicol. 2010, 30, 322–332. [Google Scholar] [CrossRef]
  101. Battersby, A.; Jones, R.D.; Lilley, K.S.; McFarlane, R.J.; Braig, H.R.; Allen, N.D.; Wakeman, J.A. Comparative proteomic analysis reveals differential expression of Hsp25 following the directed differentiation of mouse embryonic stem cells. Biochim. Biophys. Acta (BBA)-Mol. Cell Res. 2007, 1773, 147–156. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Winger, Q.A.; Guttormsen, J.; Gavin, H.; Bhushan, F. Heat shock protein 1 and the mitogen-activated protein kinase 14 pathway are important for mouse trophoblast stem cell differentiation. Biol. Reprod. 2007, 76, 884–891. [Google Scholar] [CrossRef] [PubMed]
  103. Krebsbach, P.H.; Villa-Diaz, L.G. The Role of Integrin α6 (CD49f) in Stem Cells: More than a Conserved Biomarker. Stem Cells Dev. 2017, 26, 1090–1099. [Google Scholar] [CrossRef]
  104. Al-Obaide, M.; Ishmakej, A.; Brown, C.; Mazzella, M.; Agosta, P.; Perez-Cruet, M.; Chaudhry, G.R. The potential role of integrin alpha 6 in human mesenchymal stem cells. Front. Genet. 2022, 13, 968228. [Google Scholar] [CrossRef] [PubMed]
  105. Hakki, S.S.; Kayis, S.A.; Hakki, E.E.; Bozkurt, S.B.; Duruksu, G.; Unal, Z.S.; Turaç, G.; Karaoz, E. Comparison of mesenchymal stem cells isolated from pulp and periodontal ligament. J. Periodontol. 2015, 86, 283–291. [Google Scholar] [CrossRef]
  106. Meloche, S.; Pouysségur, J. The ERK1/2 mitogen-activated protein kinase pathway as a master regulator of the G1- to S-phase transition. Oncogene 2007, 26, 3227–3239. [Google Scholar] [CrossRef] [Green Version]
  107. Buscà, R.; Pouysségur, J.; Lenormand, P. ERK1 and ERK2 Map Kinases: Specific Roles or Functional Redundancy? Front. Cell Dev. Biol. 2016, 4, 53. [Google Scholar] [CrossRef] [Green Version]
  108. Zou, J.; Lei, T.; Guo, P.; Yu, J.; Xu, Q.; Luo, Y.; Ke, R.; Huang, D. Mechanisms shaping the role of ERK1/2 in cellular senescence (Review). Mol. Med. Rep. 2019, 19, 759–770. [Google Scholar] [CrossRef] [Green Version]
  109. Chan, G.; Gu, S.; Neel, B.G. Erk1 and Erk2 are required for maintenance of hematopoietic stem cells and adult hematopoiesis. Blood 2013, 121, 3594–3598. [Google Scholar] [CrossRef] [Green Version]
  110. Zhang, C.; Lv, J.; He, Q.; Wang, S.; Gao, Y.; Meng, A.; Yang, X.; Liu, F. Inhibition of endothelial ERK signalling by Smad1/5 is essential for haematopoietic stem cell emergence. Nat. Commun. 2014, 5, 3431. [Google Scholar] [CrossRef] [Green Version]
  111. Saulnier, N.; Guihard, S.; Holy, X.; Decembre, E.; Jurdic, P.; Clay, D.; Feuillet, V.; Pages, G.; Pouysségur, J.; Porteu, F. ERK1 regulates the hematopoietic stem cell niches. PLoS ONE 2012, 7, e30788. [Google Scholar] [CrossRef]
  112. Kim, M.O.; Kim, S.H.; Cho, Y.Y.; Nadas, J.; Jeong, C.H.; Yao, K.; Kim, D.J.; Yu, D.H.; Keum, Y.S.; Lee, K.Y.; et al. ERK1 and ERK2 regulate embryonic stem cell self-renewal through phosphorylation of Klf4. Nat. Struct. Mol. Biol. 2012, 19, 283–290. [Google Scholar] [CrossRef] [PubMed]
  113. Hacohen Lev-Ran, A.; Seger, R. Retention of ERK in the cytoplasm mediates the pluripotency of embryonic stem cells. Stem Cell Rep. 2023, 18, 305–318. [Google Scholar] [CrossRef]
  114. Ma, X.; Chen, H.; Chen, L. A dual role of Erk signaling in embryonic stem cells. Exp. Hematol. 2016, 44, 151–156. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Nilsson, R.; Jain, M.; Madhusudhan, N.; Sheppard, N.G.; Strittmatter, L.; Kampf, C.; Huang, J.; Asplund, A.; Mootha, V.K. Metabolic enzyme expression highlights a key role for MTHFD2 and the mitochondrial folate pathway in cancer. Nat. Commun. 2014, 5, 3128. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Sdelci, S.; Rendeiro, A.F.; Rathert, P.; You, W.; Lin, J.-M.G.; Ringler, A.; Hofstätter, G.; Moll, H.P.; Gürtl, B.; Farlik, M.; et al. MTHFD1 interaction with BRD4 links folate metabolism to transcriptional regulation. Nat. Genet. 2019, 51, 990–998. [Google Scholar] [CrossRef]
  117. Field, M.S.; Kamynina, E.; Agunloye, O.C.; Liebenthal, R.P.; Lamarre, S.G.; Brosnan, M.E.; Brosnan, J.T.; Stover, P.J. Nuclear enrichment of folate cofactors and methylenetetrahydrofolate dehydrogenase 1 (MTHFD1) protect de novo thymidylate biosynthesis during folate deficiency. J. Biol. Chem. 2014, 289, 29642–29650. [Google Scholar] [CrossRef] [Green Version]
  118. Bhattacharya, B.; Miura, T.; Brandenberger, R.; Mejido, J.; Luo, Y.; Yang, A.X.; Joshi, B.H.; Ginis, I.; Thies, R.S.; Amit, M.; et al. Gene expression in human embryonic stem cell lines: Unique molecular signature. Blood 2004, 103, 2956–2964. [Google Scholar] [CrossRef] [Green Version]
  119. Krajinovic, M.; Lemieux-Blanchard, E.; Chiasson, S.; Primeau, M.; Costea, I.; Moghrabi, A. Role of polymorphisms in MTHFR and MTHFD1 genes in the outcome of childhood acute lymphoblastic leukemia. Pharm. J. 2004, 4, 66–72. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  120. Erčulj, N.; Kotnik, B.F.; Debeljak, M.; Jazbec, J.; Dolžan, V. Influence of folate pathway polymorphisms on high-dose methotrexate-related toxicity and survival in childhood acute lymphoblastic leukemia. Leuk. Lymphoma 2012, 53, 1096–1104. [Google Scholar] [CrossRef]
  121. de Jonge, R.; Hooijberg, J.H.; van Zelst, B.D.; Jansen, G.; van Zantwijk, C.H.; Kaspers, G.J.L.; Peters, F.G.; Ravindranath, Y.; Pieters, R.; Lindemans, J. Effect of polymorphisms in folate-related genes on in vitro methotrexate sensitivity in pediatric acute lymphoblastic leukemia. Blood 2005, 106, 717–720. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Chen, G.; Yin, S.; Zeng, H.; Li, H.; Wan, X. Regulation of Embryonic Stem Cell Self-Renewal. Life 2022, 12, 1151. [Google Scholar] [CrossRef] [PubMed]
  123. Burdon, T.; Smith, A.; Savatier, P. Signalling, cell cycle and pluripotency in embryonic stem cells. Trends Cell Biol. 2002, 12, 432–438. [Google Scholar] [CrossRef] [Green Version]
  124. Kraunsoe, S.; Azami, T.; Pei, Y.; Martello, G.; Jones, K.; Boroviak, T.; Nichols, J. Requirement for STAT3 and its target, TFCP2L1, in self-renewal of naïve pluripotent stem cells in vivo and in vitro. Biol. Open 2023, 12, bio059650. [Google Scholar] [CrossRef] [PubMed]
  125. Legate, K.R.; Montañez, E.; Kudlacek, O.; Fässler, R. ILK, PINCH and parvin: The tIPP of integrin signalling. Nat. Rev. Mol. Cell Biol. 2006, 7, 20–31. [Google Scholar] [CrossRef]
  126. Braun, A.; Bordoy, R.; Stanchi, F.; Moser, M.; ünter Kostka, G.; Ehler, E.; Brandau, O.; Fässler, R. PINCH2 is a new five LIM domain protein, homologous to PINCHand localized to focal adhesions☆. Exp. Cell Res. 2003, 284, 237–248. [Google Scholar] [CrossRef]
  127. Xu, H.; Cao, H.; Xiao, G. Signaling via PINCH: Functions, binding partners and implications in human diseases. Gene 2016, 594, 10–15. [Google Scholar] [CrossRef] [Green Version]
  128. Su, J.; Guo, L.; Wu, C. A mechanoresponsive PINCH-1-Notch2 interaction regulates smooth muscle differentiation of human placental mesenchymal stem cells. Stem Cells 2021, 39, 650–668. [Google Scholar] [CrossRef] [PubMed]
  129. Lei, Y.; Fu, X.; Li, P.; Lin, S.; Yan, Q.; Lai, Y.; Liu, X.; Wang, Y.; Bai, X.; Liu, C.; et al. LIM domain proteins Pinch1/2 regulate chondrogenesis and bone mass in mice. Bone Res. 2020, 8, 37. [Google Scholar] [CrossRef]
  130. Liao, H.; Wang, F.; Lu, K.; Ma, X.; Yan, J.; Luo, L.; Sun, Y.; Liang, X. Requirement for PINCH in skeletal myoblast differentiation. Cell Tissue Res. 2023, 391, 205–215. [Google Scholar] [CrossRef] [PubMed]
  131. Radli, M.; Veprintsev, D.B.; Rüdiger, S.G.D. Production and purification of human Hsp90β in Escherichia coli. PLoS ONE 2017, 12, e0180047. [Google Scholar] [CrossRef] [Green Version]
  132. Taipale, M.; Jarosz, D.F.; Lindquist, S. HSP90 at the hub of protein homeostasis: Emerging mechanistic insights. Nat. Rev. Mol. Cell Biol. 2010, 11, 515–528. [Google Scholar] [CrossRef] [PubMed]
  133. Buchner, J.; Li, J. Structure, function and regulation of the hsp90 machinery. Biomed. J. 2013, 36, 106. [Google Scholar] [CrossRef] [PubMed]
  134. Jing, R.; Duncan, C.B.; Duncan, S.A. A small-molecule screen reveals that HSP90β promotes the conversion of induced pluripotent stem cell-derived endoderm to a hepatic fate and regulates HNF4A turnover. Development 2017, 144, 1764–1774. [Google Scholar] [CrossRef] [Green Version]
  135. Nenasheva, V.V.; Kovaleva, G.V.; Khaidarova, N.V.; Novosadova, E.V.; Manuilova, E.S.; Antonov, S.A.; Tarantul, V.Z. Trim14 overexpression causes the same transcriptional changes in mouse embryonic stem cells and human HEK293 cells. Vitr. Cell. Dev. Biol. Anim. 2014, 50, 121–128. [Google Scholar] [CrossRef] [PubMed]
  136. Schnier, J.; Schwelberger, H.G.; Smit-McBride, Z.; Kang, H.A.; Hershey, J.W. Translation initiation factor 5A and its hypusine modification are essential for cell viability in the yeast Saccharomyces cerevisiae. Mol. Cell. Biol. 1991, 11, 3105–3114. [Google Scholar] [CrossRef]
  137. Jasiulionis, M.G.; Luchessi, A.D.; Moreira, A.G.; Souza, P.P.; Suenaga, A.P.; Correa, M.; Costa, C.A.; Curi, R.; Costa-Neto, C.M. Inhibition of eukaryotic translation initiation factor 5A (eIF5A) hypusination impairs melanoma growth. Cell Biochem. Funct. 2007, 25, 109–114. [Google Scholar] [CrossRef]
  138. Park, M.H.; Lee, Y.B.; Joe, Y.A. Hypusine is essential for eukaryotic cell proliferation. Biol. Signals 1997, 6, 115–123. [Google Scholar] [CrossRef]
  139. Dihazi, H.; Dihazi, G.H.; Jahn, O.; Meyer, S.; Nolte, J.; Asif, A.R.; Mueller, G.A.; Engel, W. Multipotent adult germline stem cells and embryonic stem cells functional proteomics revealed an important role of eukaryotic initiation factor 5A (Eif5a) in stem cell differentiation. J. Proteome Res. 2011, 10, 1962–1973. [Google Scholar] [CrossRef]
  140. Luchessi, A.D.; Cambiaghi, T.D.; Hirabara, S.M.; Lambertucci, R.H.; Silveira, L.R.; Baptista, I.L.; Moriscot, A.S.; Costa-Neto, C.M.; Curi, R. Involvement of eukaryotic translation initiation factor 5A (eIF5A) in skeletal muscle stem cell differentiation. J. Cell. Physiol. 2009, 218, 480–489. [Google Scholar] [CrossRef] [PubMed]
  141. Ingley, E. Functions of the Lyn tyrosine kinase in health and disease. Cell Commun. Signal. 2012, 10, 21. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Boggon, T.J.; Eck, M.J. Structure and regulation of Src family kinases. Oncogene 2004, 23, 7918–7927. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Resh, M.D. Fatty acylation of proteins: New insights into membrane targeting of myristoylated and palmitoylated proteins. Biochim. Biophys. Acta 1999, 1451, 1–16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Zhang, X.; Simerly, C.; Hartnett, C.; Schatten, G.; Smithgall, T.E. Src-family tyrosine kinase activities are essential for differentiation of human embryonic stem cells. Stem Cell Res. 2014, 13, 379–389. [Google Scholar] [CrossRef] [Green Version]
  145. Meyn, M.A., 3rd; Schreiner, S.J.; Dumitrescu, T.P.; Nau, G.J.; Smithgall, T.E. SRC family kinase activity is required for murine embryonic stem cell growth and differentiation. Mol. Pharm. 2005, 68, 1320–1330. [Google Scholar] [CrossRef] [Green Version]
  146. Alvandi, Z.; Opas, M. c-Src kinase inhibits osteogenic differentiation via enhancing STAT1 stability. PLoS ONE 2020, 15, e0241646. [Google Scholar] [CrossRef]
  147. Chow, K.H.; Park, H.J.; George, J.; Yamamoto, K.; Gallup, A.D.; Graber, J.H.; Chen, Y.; Jiang, W.; Steindler, D.A.; Neilson, E.G.; et al. S100A4 Is a Biomarker and Regulator of Glioma Stem Cells That Is Critical for Mesenchymal Transition in Glioblastoma. Cancer Res. 2017, 77, 5360–5373. [Google Scholar] [CrossRef] [Green Version]
  148. Ishikawa, M.; Osaki, M.; Yamagishi, M.; Onuma, K.; Ito, H.; Okada, F.; Endo, H. Correlation of two distinct metastasis-associated proteins, MTA1 and S100A4, in angiogenesis for promoting tumor growth. Oncogene 2019, 38, 4715–4728. [Google Scholar] [CrossRef]
  149. Li, Y.; Wang, J.; Song, K.; Liu, S.; Zhang, H.; Wang, F.; Ni, C.; Zhai, W.; Liang, J.; Qin, Z.; et al. S100A4 promotes hepatocellular carcinogenesis by intensifying fibrosis-associated cancer cell stemness. Oncoimmunology 2020, 9, 1725355. [Google Scholar] [CrossRef] [Green Version]
  150. Mishra, S.K.; Siddique, H.R.; Saleem, M. S100A4 calcium-binding protein is key player in tumor progression and metastasis: Preclinical and clinical evidence. Cancer Metastasis Rev. 2012, 31, 163–172. [Google Scholar] [CrossRef]
  151. Tochimoto, M.; Oguri, Y.; Hashimura, M.; Konno, R.; Matsumoto, T.; Yokoi, A.; Kodera, Y.; Saegusa, M. S100A4/non-muscle myosin II signaling regulates epithelial-mesenchymal transition and stemness in uterine carcinosarcoma. Lab. Invest. 2020, 100, 682–695. [Google Scholar] [CrossRef] [PubMed]
  152. Stary, M.; Schneider, M.; Sheikh, S.P.; Weitzer, G. Parietal endoderm secreted S100A4 promotes early cardiomyogenesis in embryoid bodies. Biochem. Biophys. Res. Commun. 2006, 343, 555–563. [Google Scholar] [CrossRef]
  153. Schneider, M.; Hansen, J.L.; Sheikh, S.P. S100A4: A common mediator of epithelial–mesenchymal transition, fibrosis and regeneration in diseases? J. Mol. Med. 2008, 86, 507–522. [Google Scholar] [CrossRef] [PubMed]
  154. Figeac, N.; Serralbo, O.; Marcelle, C.; Zammit, P.S. ErbB3 binding protein-1 (Ebp1) controls proliferation and myogenic differentiation of muscle stem cells. Dev. Biol. 2014, 386, 135–151. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Neilson, K.M.; Abbruzzesse, G.; Kenyon, K.; Bartolo, V.; Krohn, P.; Alfandari, D.; Moody, S.A. Pa2G4 is a novel Six1 co-factor that is required for neural crest and otic development. Dev. Biol. 2017, 421, 171–182. [Google Scholar] [CrossRef]
  156. Yoo, J.; Wang, X.; Rishi, A.; Lessor, T.; Xia, X.; Gustafson, T.; Hamburger, A. Interaction of the PA2G4 (EBP1) protein with ErbB-3 and regulation of this binding by heregulin. Br. J. Cancer 2000, 82, 683–690. [Google Scholar] [CrossRef] [Green Version]
  157. Somanath, P.; Bush, K.M.; Knoepfler, P.S. ERBB3-Binding Protein 1 (EBP1) Is a Novel Developmental Pluripotency-Associated-4 (DPPA4) Cofactor in Human Pluripotent Cells. Stem Cells 2018, 36, 671–682. [Google Scholar] [CrossRef] [Green Version]
  158. Song, Y.; Cao, P.; Gu, Z.; Xiao, J.; Lian, M.; Huang, D.; Xing, J.; Zhang, Y.; Feng, X.; Wang, C. The Role of Neuropilin-1-FYN Interaction in Odontoblast Differentiation of Dental Pulp Stem Cells. Cell Reprogram 2018, 20, 117–126. [Google Scholar] [CrossRef] [PubMed]
  159. Thompson, W.R.; Guilluy, C.; Xie, Z.; Sen, B.; Brobst, K.E.; Yen, S.S.; Uzer, G.; Styner, M.; Case, N.; Burridge, K.; et al. Mechanically activated fyn utilizes mTORC2 to regulate RhoA and adipogenesis in mesenchymal stem cells. Stem Cells 2013, 31, 2528–2537. [Google Scholar] [CrossRef] [Green Version]
  160. Brzozowski, J.S.; Skelding, K.A. The Multi-Functional Calcium/Calmodulin Stimulated Protein Kinase (CaMK) Family: Emerging Targets for Anti-Cancer Therapeutic Intervention. Pharmaceuticals 2019, 12, 8. [Google Scholar] [CrossRef] [Green Version]
  161. Takemoto-Kimura, S.; Suzuki, K.; Horigane, S.I.; Kamijo, S.; Inoue, M.; Sakamoto, M.; Fujii, H.; Bito, H. Calmodulin kinases: Essential regulators in health and disease. J. Neurochem. 2017, 141, 808–818. [Google Scholar] [CrossRef] [Green Version]
  162. Skelding, K.A.; Rostas, J.A.P. Regulation of Multifunctional Calcium/Calmodulin Stimulated Protein Kinases by Molecular Targeting. Adv. Exp. Med. Biol. 2020, 1131, 649–679. [Google Scholar] [CrossRef]
  163. Boutz, P.L.; Stoilov, P.; Li, Q.; Lin, C.-H.; Chawla, G.; Ostrow, K.; Shiue, L.; Ares, M.; Black, D.L. A post-transcriptional regulatory switch in polypyrimidine tract-binding proteins reprograms alternative splicing in developing neurons. Genes Dev. 2007, 21, 1636–1652. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Coutinho-Mansfield, G.C.; Xue, Y.; Zhang, Y.; Fu, X.D. PTB/nPTB switch: A post-transcriptional mechanism for programming neuronal differentiation. Genes Dev. 2007, 21, 1573–1577. [Google Scholar] [CrossRef] [Green Version]
  165. Dai, S.; Wang, C.; Zhang, C.; Feng, L.; Zhang, W.; Zhou, X.; He, Y.; Xia, X.; Chen, B.; Song, W. PTB: Not just a polypyrimidine tract-binding protein. J. Cell. Physiol. 2022, 237, 2357–2373. [Google Scholar] [CrossRef] [PubMed]
  166. Rehn, M.; Wenzel, A.; Frank, A.K.; Schuster, M.B.; Pundhir, S.; Jørgensen, N.; Vitting-Seerup, K.; Ge, Y.; Jendholm, J.; Michaut, M.; et al. PTBP1 promotes hematopoietic stem cell maintenance and red blood cell development by ensuring sufficient availability of ribosomal constituents. Cell Rep. 2022, 39, 110793. [Google Scholar] [CrossRef]
  167. Iannone, C.; Kainov, Y.; Zhuravskaya, A.; Hamid, F.; Nojima, T.; Makeyev, E.V. PTBP1-activated co-transcriptional splicing controls epigenetic status of pluripotent stem cells. Mol. Cell 2023, 83, 203–218.e209. [Google Scholar] [CrossRef]
  168. Wang, D.H.; Chen, J.S.; Hou, R.; Li, Y.; An, J.H.; He, P.; Cai, Z.G.; Liang, X.H.; Liu, Y.L. Comparison of transcriptome profiles of mesenchymal stem cells derived from umbilical cord and bone marrow of giant panda (Ailuropoda melanoleuca). Gene 2022, 845, 146854. [Google Scholar] [CrossRef] [PubMed]
  169. Jiang, J.; Hai, J.; Liu, W.; Luo, Y.; Chen, K.; Xin, Y.; Pan, J.; Hu, Y.; Gao, Q.; Xiao, F. Gallic Acid Induces Neural Stem Cell Differentiation into Neurons and Proliferation through the MAPK/ERK Pathway. J. Agric. Food Chem. 2021, 69, 12456–12464. [Google Scholar] [CrossRef]
  170. Maruyama, T.; Jiang, M.; Abbott, A.; Yu, H.I.; Huang, Q.; Chrzanowska-Wodnicka, M.; Chen, E.I.; Hsu, W. Rap1b Is an Effector of Axin2 Regulating Crosstalk of Signaling Pathways During Skeletal Development. J. Bone Min. Res. 2017, 32, 1816–1828. [Google Scholar] [CrossRef] [Green Version]
  171. Gao, S.; Chen, B.; Zhu, Z.; Du, C.; Zou, J.; Yang, Y.; Huang, W.; Liao, J. PI3K-Akt signaling regulates BMP2-induced osteogenic differentiation of mesenchymal stem cells (MSCs): A transcriptomic landscape analysis. Stem Cell Res. 2023, 66, 103010. [Google Scholar] [CrossRef] [PubMed]
  172. Halfon, S.; Abramov, N.; Grinblat, B.; Ginis, I. Markers distinguishing mesenchymal stem cells from fibroblasts are downregulated with passaging. Stem Cells Dev. 2011, 20, 53–66. [Google Scholar] [CrossRef]
  173. Ise, H.; Matsunaga, K.; Shinohara, M.; Sakai, Y. Improved Isolation of Mesenchymal Stem Cells Based on Interactions between N-Acetylglucosamine-Bearing Polymers and Cell-Surface Vimentin. Stem Cells Int. 2019, 2019, 4341286. [Google Scholar] [CrossRef]
  174. Lee, T.C.; Lee, T.H.; Huang, Y.H.; Chang, N.K.; Lin, Y.J.; Chien, P.W.; Yang, W.H.; Lin, M.H. Comparison of surface markers between human and rabbit mesenchymal stem cells. PLoS ONE 2014, 9, e111390. [Google Scholar] [CrossRef] [Green Version]
  175. Malekmohammad, K.; Sewell, R.D.E.; Rafieian-Kopaei, M. Antioxidants and Atherosclerosis: Mechanistic Aspects. Biomolecules 2019, 9, 301. [Google Scholar] [CrossRef] [Green Version]
  176. Yang, J.; Song, T.; Wu, P.; Chen, Y.; Fan, X.; Chen, H.; Zhang, J.; Huang, C. Differentiation potential of human mesenchymal stem cells derived from adipose tissue and bone marrow to sinus node-like cells. Mol. Med. Rep. 2012, 5, 108–113. [Google Scholar] [CrossRef] [PubMed]
  177. Paspaliaris, V.; Kolios, G. Stem cells in Osteoporosis: From Biology to New Therapeutic Approaches. Stem Cells Int. 2019, 2019, 1730978. [Google Scholar] [CrossRef] [Green Version]
  178. Ahlström, M.; Pekkinen, M.; Lamberg-Allardt, C. Dexamethasone downregulates the expression of parathyroid hormone-related protein (PTHrP) in mesenchymal stem cells. Steroids 2009, 74, 277–282. [Google Scholar] [CrossRef]
  179. Lyu, P.; Li, B.; Li, P.; Bi, R.; Cui, C.; Zhao, Z.; Zhou, X.; Fan, Y. Parathyroid Hormone 1 Receptor Signaling in Dental Mesenchymal Stem Cells: Basic and Clinical Implications. Front. Cell Dev. Biol. 2021, 9, 654715. [Google Scholar] [CrossRef] [PubMed]
  180. Fan, Y.; Hanai, J.I.; Le, P.T.; Bi, R.; Maridas, D.; DeMambro, V.; Figueroa, C.A.; Kir, S.; Zhou, X.; Mannstadt, M.; et al. Parathyroid Hormone Directs Bone Marrow Mesenchymal Cell Fate. Cell Metab. 2017, 25, 661–672. [Google Scholar] [CrossRef] [Green Version]
  181. Reeve, J.; Meunier, P.J.; Parsons, J.A.; Bernat, M.; Bijvoet, O.; Courpron, P.; Edouard, C.; Klenerman, L.; Neer, R.M.; Renier, J.C. Anabolic effect of human parathyroid hormone fragment on trabecular bone in involutional osteoporosis: A multicentre trial. Br. Med. J. 1980, 280, 1340–1344. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  182. Chen, T.; Wang, Y.; Hao, Z.; Hu, Y.; Li, J. Parathyroid hormone and its related peptides in bone metabolism. Biochem. Pharmacol. 2021, 192, 114669. [Google Scholar] [CrossRef] [PubMed]
  183. Sauzay, C.; Voutetakis, K.; Chatziioannou, A.; Chevet, E.; Avril, T. CD90/Thy-1, a Cancer-Associated Cell Surface Signaling Molecule. Front. Cell Dev. Biol. 2019, 7, 66. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Leyton, L.; Díaz, J.; Martínez, S.; Palacios, E.; Pérez, L.A.; Pérez, R.D. Thy-1/CD90 a Bidirectional and Lateral Signaling Scaffold. Front. Cell Dev. Biol. 2019, 7, 132. [Google Scholar] [CrossRef]
  185. Rege, T.A.; Hagood, J.S. Thy-1 as a regulator of cell-cell and cell-matrix interactions in axon regeneration, apoptosis, adhesion, migration, cancer, and fibrosis. FASEB J. 2006, 20, 1045–1054. [Google Scholar] [CrossRef] [Green Version]
  186. Horn, P.A.; Tesch, H.; Staib, P.; Kube, D.; Diehl, V.; Voliotis, D. Expression of AC133, a novel hematopoietic precursor antigen, on acute myeloid leukemia cells. Blood 1999, 93, 1435–1437. [Google Scholar] [CrossRef] [PubMed]
  187. Majka, M.; Ratajczak, J.; Machalinski, B.; Carter, A.; Pizzini, D.; Wasik, M.A.; Gewirtz, A.M.; Ratajczak, M.Z. Expression, regulation and function of AC133, a putative cell surface marker of primitive human haematopoietic cells. Folia Histochem. Cytobiol. 2000, 38, 53–63. [Google Scholar] [PubMed]
  188. Patel, P.; Abbasian, J.; Mahmud, D.; Mahmud, N.; Horsthemke, P.M.; Chunduri, S.; Rondelli, D. Allostimulatory activity of CD133+ hematopoietic cells. Bone Marrow Transpl. 2013, 48, 742–744. [Google Scholar] [CrossRef] [PubMed]
  189. Kucia, M.; Halasa, M.; Wysoczynski, M.; Baskiewicz-Masiuk, M.; Moldenhawer, S.; Zuba-Surma, E.; Czajka, R.; Wojakowski, W.; Machalinski, B.; Ratajczak, M.Z. Morphological and molecular characterization of novel population of CXCR4+ SSEA-4+ Oct-4+ very small embryonic-like cells purified from human cord blood: Preliminary report. Leukemia 2007, 21, 297–303. [Google Scholar] [CrossRef] [Green Version]
  190. Ratajczak, M.Z.; Mierzejewska, K.; Ratajczak, J.; Kucia, M. CD133 Expression Strongly Correlates with the Phenotype of Very Small Embryonic-/Epiblast-Like Stem Cells. Adv. Exp. Med. Biol. 2013, 777, 125–141. [Google Scholar] [CrossRef] [Green Version]
  191. Virant-Klun, I.; Zech, N.; Rozman, P.; Vogler, A.; Cvjeticanin, B.; Klemenc, P.; Malicev, E.; Meden-Vrtovec, H. Putative stem cells with an embryonic character isolated from the ovarian surface epithelium of women with no naturally present follicles and oocytes. Differentiation 2008, 76, 843–856. [Google Scholar] [CrossRef] [PubMed]
  192. Sato, T.; Wakao, S.; Kushida, Y.; Tatsumi, K.; Kitada, M.; Abe, T.; Niizuma, K.; Tominaga, T.; Kushimoto, S.; Dezawa, M. A Novel Type of Stem Cells Double-Positive for SSEA-3 and CD45 in Human Peripheral Blood. Cell Transpl. 2020, 29, 963689720923574. [Google Scholar] [CrossRef]
  193. Parte, S.; Bhartiya, D.; Telang, J.; Daithankar, V.; Salvi, V.; Zaveri, K.; Hinduja, I. Detection, characterization, and spontaneous differentiation in vitro of very small embryonic-like putative stem cells in adult mammalian ovary. Stem Cells Dev. 2011, 20, 1451–1464. [Google Scholar] [CrossRef] [PubMed]
  194. Dzierzak, E.; de Pater, E. Regulation of Blood Stem Cell Development. Curr. Top. Dev. Biol. 2016, 118, 1–20. [Google Scholar] [CrossRef]
  195. Dzierzak, E.; Speck, N.A. Of lineage and legacy: The development of mammalian hematopoietic stem cells. Nat. Immunol. 2008, 9, 129–136. [Google Scholar] [CrossRef] [Green Version]
  196. Orkin, S.H.; Zon, L.I. Hematopoiesis: An evolving paradigm for stem cell biology. Cell 2008, 132, 631–644. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  197. Boltze, J.; Arnold, A.; Walczak, P.; Jolkkonen, J.; Cui, L.; Wagner, D.C. The Dark Side of the Force—Constraints and Complications of Cell Therapies for Stroke. Front. Neurol. 2015, 6, 155. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  198. Lengefeld, J.; Amon, A. 3022—CELL SIZE IS A DETERMINANT OF STEM CELL POTENTIAL DURING AGING. Exp. Hematol. 2021, 100, S54. [Google Scholar] [CrossRef]
  199. Gao, D.Y.; Chang, Q.; Liu, C.; Farris, K.; Harvey, K.; McGann, L.E.; English, D.; Jansen, J.; Critser, J.K. Fundamental cryobiology of human hematopoietic progenitor cells. I: Osmotic characteristics and volume distribution. Cryobiology 1998, 36, 40–48. [Google Scholar] [CrossRef] [PubMed]
  200. Bhartiya, D.; Shaikh, A.; Nagvenkar, P.; Kasiviswanathan, S.; Pethe, P.; Pawani, H.; Mohanty, S.; Rao, S.G.; Zaveri, K.; Hinduja, I. Very small embryonic-like stem cells with maximum regenerative potential get discarded during cord blood banking and bone marrow processing for autologous stem cell therapy. Stem Cells Dev. 2012, 21, 1–6. [Google Scholar] [CrossRef]
Figure 1. Methodology for the isolation of SBSCs.
Figure 1. Methodology for the isolation of SBSCs.
Biomedicines 11 00787 g001
Figure 2. Representative brightfield microscopy image of Trypan Blue-stained and H&E-stained SBSCs. (A) Black arrows indicate live cells, while red arrows show dead cells. (B) Black arrows indicate H&E-stained SBSCs.
Figure 2. Representative brightfield microscopy image of Trypan Blue-stained and H&E-stained SBSCs. (A) Black arrows indicate live cells, while red arrows show dead cells. (B) Black arrows indicate H&E-stained SBSCs.
Biomedicines 11 00787 g002
Figure 3. Immunofluorescence and light microscopy images of SBSCs. (A) SBSCs stained with KP-1. (B) SBSCs observed using a bright field light microscope. Representative 400× snapshots are shown.
Figure 3. Immunofluorescence and light microscopy images of SBSCs. (A) SBSCs stained with KP-1. (B) SBSCs observed using a bright field light microscope. Representative 400× snapshots are shown.
Biomedicines 11 00787 g003
Figure 4. SBSCs stained for embryonic stem cell markers. Representative 400× immunofluorescence snapshots are shown. Isotype control staining was examined for each tested antibody, and the result was negative (Supplementary Figure S1). Blue and red/green colors correspond to DAPI and embryonic stem cell marker antibody staining, respectively.
Figure 4. SBSCs stained for embryonic stem cell markers. Representative 400× immunofluorescence snapshots are shown. Isotype control staining was examined for each tested antibody, and the result was negative (Supplementary Figure S1). Blue and red/green colors correspond to DAPI and embryonic stem cell marker antibody staining, respectively.
Biomedicines 11 00787 g004
Figure 5. SBSCs stained for mesenchymal stem cell markers. Representative 400× immunofluorescence snapshots are shown. Isotype control staining was examined for each tested antibody, and the result was negative (Supplementary Figure S1). Blue and green colors correspond to DAPI and mesenchymal stem cell marker antibody staining, respectively.
Figure 5. SBSCs stained for mesenchymal stem cell markers. Representative 400× immunofluorescence snapshots are shown. Isotype control staining was examined for each tested antibody, and the result was negative (Supplementary Figure S1). Blue and green colors correspond to DAPI and mesenchymal stem cell marker antibody staining, respectively.
Biomedicines 11 00787 g005
Figure 6. SBSCs stained for hematopoietic stem cell markers. Representative 400× immunofluorescence snapshots are shown. Isotype control staining was examined for each tested antibody, and the result was negative (Supplementary Figure S1). Blue and red/green colors correspond to DAPI and hematopoietic stem cell marker antibody staining, respectively.
Figure 6. SBSCs stained for hematopoietic stem cell markers. Representative 400× immunofluorescence snapshots are shown. Isotype control staining was examined for each tested antibody, and the result was negative (Supplementary Figure S1). Blue and red/green colors correspond to DAPI and hematopoietic stem cell marker antibody staining, respectively.
Biomedicines 11 00787 g006
Figure 7. SBSCs double stained for CD45 and embryonic stem cell markers. Representative 400× immunofluorescence snapshots are shown. White arrows indicate simultaneous co-staining of both CD45 and embryonic stem cell markers. Red arrows indicate strong CD45 staining and weak or not at all staining of embryonic stem cell markers. Isotype control staining was examined for each tested antibody, and the result was negative (Supplementary Figure S1). Blue and green/red colors correspond to DAPI and CD45/embryonic stem cell marker antibody staining, respectively.
Figure 7. SBSCs double stained for CD45 and embryonic stem cell markers. Representative 400× immunofluorescence snapshots are shown. White arrows indicate simultaneous co-staining of both CD45 and embryonic stem cell markers. Red arrows indicate strong CD45 staining and weak or not at all staining of embryonic stem cell markers. Isotype control staining was examined for each tested antibody, and the result was negative (Supplementary Figure S1). Blue and green/red colors correspond to DAPI and CD45/embryonic stem cell marker antibody staining, respectively.
Biomedicines 11 00787 g007
Figure 8. Proteomic analysis of SBSCs. (A) Experimental workflow of isolation and proteome analysis of SBSCs. (B) Proteome composition of SBSCs, including various stem cell marker proteins, and factors associated with transcriptional regulator complex. (C) SBSC proteome abundance treemap, highlighting various adhesion, cytoskeletal, signaling and metabolic factors abundant in SBSCs. The cell size of the SBSC treemap indicates the protein abundance of 809 proteins. (DH) Profiler enrichment of the SBSC proteome, Gene Ontology (D), Component (E), Process (F), Function (G), KEGG (H), Reactome functional enrichments (FDR < 0.05).
Figure 8. Proteomic analysis of SBSCs. (A) Experimental workflow of isolation and proteome analysis of SBSCs. (B) Proteome composition of SBSCs, including various stem cell marker proteins, and factors associated with transcriptional regulator complex. (C) SBSC proteome abundance treemap, highlighting various adhesion, cytoskeletal, signaling and metabolic factors abundant in SBSCs. The cell size of the SBSC treemap indicates the protein abundance of 809 proteins. (DH) Profiler enrichment of the SBSC proteome, Gene Ontology (D), Component (E), Process (F), Function (G), KEGG (H), Reactome functional enrichments (FDR < 0.05).
Biomedicines 11 00787 g008
Figure 9. Analysis and comparison of the SBSC and plasma cell proteome. (A) Comparison of SBSC proteome with isolated plasma cells (control plasma cells). Proteins uniquely identified in SBSCs (13) and co-identified (796) are shown. Proteins differentially expressed based on LFQ intensity are shown—upregulated expression (↑, LFQ) and downregulated expression (↓) of SBSCs vs plasma cells (average LFQ). Specific proteins identified in each group are indicated, including various stem cell marker proteins, and factors associated with transcriptional regulator complex. (B) Enriched cell surface marker proteins in SBSCs (based on UniProt). (C) Proteins uniquely identified in the SBSC (vs control plasma cells) proteome. (D) Profiler enrichment of top 50 proteins identified in the SBSC vs control plasma cell proteome (LFQ intensity) (FDR < 0.05).
Figure 9. Analysis and comparison of the SBSC and plasma cell proteome. (A) Comparison of SBSC proteome with isolated plasma cells (control plasma cells). Proteins uniquely identified in SBSCs (13) and co-identified (796) are shown. Proteins differentially expressed based on LFQ intensity are shown—upregulated expression (↑, LFQ) and downregulated expression (↓) of SBSCs vs plasma cells (average LFQ). Specific proteins identified in each group are indicated, including various stem cell marker proteins, and factors associated with transcriptional regulator complex. (B) Enriched cell surface marker proteins in SBSCs (based on UniProt). (C) Proteins uniquely identified in the SBSC (vs control plasma cells) proteome. (D) Profiler enrichment of top 50 proteins identified in the SBSC vs control plasma cell proteome (LFQ intensity) (FDR < 0.05).
Biomedicines 11 00787 g009
Table 1. List of antibodies used for the characterization of SBSCs.
Table 1. List of antibodies used for the characterization of SBSCs.
PluripotencyAntigen
EmbryonicNanog 1, Oct4 1, CXCR4 1, SOX2 1, KLF4 1, cMyc 1, SSEA-3 1 and SSEA-4 2
MesenchymalCD29 3, PTH1R 1, CD105 1 and CD106 3
HematopoieticCD45 3, CD34 1, CD90 1 and CD133 1
1 Novus Biologicals, Littleton, CO, USA, 2 Merck Millipore, Burlington, MA, USA, 3 R&D Systems, Minneapolis, MN, USA.
Table 2. Identified proteins that are associated with stem cell differentiation processes.
Table 2. Identified proteins that are associated with stem cell differentiation processes.
ProteinFunctional RoleRole in Stem Cell Differentiation
LIMS1Adaptor protein that binds to integrin-linked kinase and has a vital role in the processes of cell adhesion [125,126,127]Promotes chondrogenesis, differentiation of placental mesenchymal cells to smooth muscle cells and plays a crucial part in skeletal myogenic differentiation [128,129,130]
HSP90AB1Molecular chaperone involved in protein degradation [131,132,133]Plays a role in the endoderm differentiation towards hepatic cells [134] and in mesodermal differentiation [135]
EIF5AAssociated with protein translation and cell proliferation and viability [136,137,138]Supports embryonic stem cell differentiation and promotes skeletal muscle stem cell differentiation [139,140]
LYNBelongs to the Src-family of tyrosine kinases and it is a nonreceptor cytoplasmic protein. It has a pivotal role in various cellular processes such as migration, cell growth, apoptosis, adhesion, differentiation, metabolism and immune response [141,142,143]It has significant role in the differentiation process of embryonic stem cells [144,145,146]
S100A4Belongs to the S100 family of calcium-binding proteins and is involved in various processes, such as angiogenesis, cell growth, motility, differentiation, apoptosis and invasion [147,148,149,150,151]Promotes the differentiation of endoderm towards cardiomyocytes [152] and has a central role in the epithelial-to-mesenchymal transition [153]
PA2G4It is a protein regulator of the ErbB3 signaling pathway, and it plays a role in cell growth, apoptosis and differentiation [154,155,156]It plays a significant role in the differentiation of muscle stem cells and pluripotency of stem cells [154,157]
FYNBelongs to the Src-family of tyrosine kinases and it is involved in various processes, such as cell growth, survival, adhesion, motility and tumor metastasis [142,143,158]It is implicated in the differentiation process of mesenchymal stem cells [159] and has a significant role in the self-renewal of murine embryonic stem cells [145]
CAMK1It is a protein kinase, primarily expressed in nerve cells, with various functions [160,161,162]Unknown
PTBP1Belongs to the heterogeneous nuclear ribonucleoprotein family and has a pivotal role in neuronal development [163,164,165]It plays a significant role in the self-renewal of hematopoietic stem cells [166] and in the neuronal development [167]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Filidou, E.; Kandilogiannakis, L.; Tarapatzi, G.; Spathakis, M.; Su, C.; Rai, A.; Greening, D.W.; Arvanitidis, K.; Paspaliaris, V.; Kolios, G. A Simplified and Effective Approach for the Isolation of Small Pluripotent Stem Cells Derived from Human Peripheral Blood. Biomedicines 2023, 11, 787. https://doi.org/10.3390/biomedicines11030787

AMA Style

Filidou E, Kandilogiannakis L, Tarapatzi G, Spathakis M, Su C, Rai A, Greening DW, Arvanitidis K, Paspaliaris V, Kolios G. A Simplified and Effective Approach for the Isolation of Small Pluripotent Stem Cells Derived from Human Peripheral Blood. Biomedicines. 2023; 11(3):787. https://doi.org/10.3390/biomedicines11030787

Chicago/Turabian Style

Filidou, Eirini, Leonidas Kandilogiannakis, Gesthimani Tarapatzi, Michail Spathakis, Colin Su, Alin Rai, David W. Greening, Konstantinos Arvanitidis, Vasilis Paspaliaris, and George Kolios. 2023. "A Simplified and Effective Approach for the Isolation of Small Pluripotent Stem Cells Derived from Human Peripheral Blood" Biomedicines 11, no. 3: 787. https://doi.org/10.3390/biomedicines11030787

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop