The potential use of stem cells (SCs) for tissue engineering, regenerative medicine, disease modeling, toxicological studies, drug delivery, and as in vitro model for the study of basic developmental processes implies large-scale in vitro culture. Here, after a brief description of the main techniques used for karyotype analysis, we will give a detailed overview of the chromosome abnormalities described in pluripotent (embryonic and induced pluripotent SCs) and somatic SCs, and the possible causes of their origin during culture.

Stem cells (SCs) are undifferentiated cells capable of self-renewal and of differentiation into specific terminally-differentiated cell types. Based on their source of derivation, they can be classified into embryonic stem cells (ESCs) or somatic stem cells (SSCs).

The first mouse ESC (mESC) lines were derived in 2 independent laboratories at the beginning of the 1980s from preimplantation embryos at the blastocyst stage [Evans and Kaufman, 1981; Martin, 1981]. About 15 years later, the first primate non-human ESC lines (rhesus monkey, Callithrix jacchus) were generated [Thomson et al., 1995, 1996], followed by the first human (hESC) [Thomson et al., 1998], rabbit [Fang et al., 2006; Wang et al., 2007], canine [Hayes et al., 2008], and rat [Buehr et al., 2008; Li et al., 2008] lines. Once established in vitro, ESCs are able to differentiate into almost all mature fetal and adult cell types, and thus they are defined as pluripotent cells. When transferred into a blastocyst, they participate in the formation of both somatic and germ lines of the growing individual [Rossant, 2001; Smith, 2001; Cockburn and Rossant, 2010]. In 2006, Takahashi and Yamanaka demonstrated the feasibility to obtain pluripotent ESC-like cells by reprogramming somatic terminally-differentiated mouse or human fibroblasts [Takahashi and Yamanaka, 2006; Takahashi et al., 2007]. These authors showed that the induced ectopic expression of 4 genes (Oct4, Sox2, Klf4, and cMyc) was sufficient to reprogram fibroblasts into the so-called induced pluripotent stem cells (iPSCs), which show similar developmental potential and pluripotency as ESCs [Takahashi and Yamanaka, 2006; Okita et al., 2007; Wernig et al., 2007].

The first SSCs were identified long before ESCs or iPSCs. In the early 1960s, hematopoietic SSCs were purified from mouse bone marrow [Till and McCullough, 1961]. Since then, SSCs have been identified in many different organs (i.e. skeletal muscle, heart, liver, fat, umbilical cord blood, or placenta). SSCs exert a crucial role in the maintenance of tissue homeostasis and participate in the repairing processes within their specific tissue. In vivo, SSCs can differentiate either in one, few or multiple cell lineages and thus are classified as unipotent (e.g. spermatogonia, oogonia), oligopotent (e.g. neural stem cells, NSCs) or multipotent (e.g. hematopoietic SCs). In vitro, SSCs display greater plasticity, showing higher differentiation potential than in vivo [Jiang et al., 2002; Franco Lambert et al., 2009]. Among SSCs, human mesenchymal stromal, hematopoietic and neural SCs already find an application in the clinic [Bajada et al., 2008; Thomsen et al., 2014; Porada et al., 2015].

Both ESCs and SSCs have boosted existing and, sometimes, given rise to novel and interdisciplinary research fields such as tissue engineering [Katari et al., 2015], regenerative medicine [Grompe, 2012], disease modeling [Merkle and Eggan, 2013], toxicological studies, and drug delivery [Seiler and Spielmann, 2011; Liu et al., 2013]. Furthermore, ESCs are an in vitro model for the study of basic developmental processes [Neri et al., 2011; Rebuzzini et al., 2013].

If, on one side, this interdisciplinarity allowed a quantum leap in our basic knowledge of the biology of SCs, it has also brought out a number of criticalities that limit their clinical use. For example, the potential risk of forming tumors, the migration far away from the site of infusion and colonization of other tissues, the dedifferentiation of SC-derived differentiated cells, the establishment of an incorrect epigenetic and genetic status, and an abnormal chromosome complement.

The reflection paper on SC-based medicinal products - EMA/CAT 14 January 2011 - defines, among other requirements, that ‘stem cell preparations undergoing extensive in vitro manipulation such as prolonged cell culture, as well as those derived from hESCs or iPSCs are evaluated for both their tumorigenicity and chromosomal stability before their initial clinical use'.

During the last decade, several papers evidenced the difficulty of SCs to maintain a correct chromosome complement during prolonged expansion [for reviews see Rebuzzini et al., 2011; Oliveira et al., 2014]. After a brief description of the main techniques used for the karyotype analysis, this review will give a detailed overview of the chromosome abnormalities described in both pluripotent SCs (ESCs and iPSCs) and SSCs, and the possible causes of their origin during prolonged culture.

Several techniques are currently available to investigate the integrity of the chromosome complement of a cell line. Each method has advantages and disadvantages in terms of sensitivity, resolution and costs [Catalina et al., 2007].

Conventional banding techniques (G-, Q- or DAPI-banding) allow a snapshot of the entire chromosome complement and the ordinary gross karyotype control of a cell line. These techniques, providing 300-400 stained bands, facilitate the detection of incorrect chromosome numbers (aneuploidies), mosaicism and large structural chromosome abnormalities, such as translocations, deletions or insertions, with a resolution of 5-10 Mb.

Spectral karyotyping (SKY) and multicolor FISH represent an advancement of the conventional cytogenetic banding analysis [Liyanage et al., 1996; Schröck et al., 1996]. These techniques, using chromosome-specific fluorescently labeled probes, allow the detection of submicroscopic deletions, insertions or amplifications of DNA, with a resolution of ∼1-2 Mb.

To detect smaller genetic imbalances, the array-based comparative genomic hybridization (array-CGH) [Sanlaville et al., 2005] and the single nucleotide polymorphism (SNP) array [Peiffer et al., 2006] are useful techniques. They allow the identification of tiny aberrations, comprising homo- or hemizygous deletions, copy-neutral loss of heterozygosity, duplications, and amplifications. Their resolution ranges from 1 Mb to <100 kb. Both CGH and SNP-array are unable to evaluate the frequency of a specific abnormality within a cell population.

Human Embryonic Stem Cells

Variations in the chromosome complement of hESC lines, detected by conventional banding techniques or by CGH assays, comprise both numerical and structural aberrations. Chromosomes 1, 12, 17, 20, and X are most frequently involved (∼50% of the aberrations found involved these chromosomes). Duplication of 1q11q32, whole or partial gain of chromosomes 12 and 17, duplication of the 20q11.21 region or aneuploidy of chromosome X have been reported by many independent laboratories (table 1) [Brimble et al., 2004; Draper et al., 2004; Inzunza et al., 2004; Mitalipova et al., 2005; Imreh et al., 2006; Baker et al., 2007; Lefort et al., 2008, 2009; Spits et al., 2008; International Stem Cell Initiative, 2011]. The recurrence of these specific types of mutations, linked to the presence of additional copies of some specific genes, may confer carrier cells with a selective and/or proliferative advantage, higher culture adaptation and resistance to apoptosis [Clark et al., 2004; Rosler et al., 2004; Caldas and Brenton, 2005; Maitra et al., 2005; Baker et al., 2007; Lefort et al., 2008; Navarro et al., 2008; Spits et al., 2008; Blum and Benvenisty, 2009; Werbowetski-Ogilvie et al., 2009; International Stem Cell Initiative, 2011; Oliveira et al., 2014].

Table 1

Frequency of pluripotent SC lines with recurrent numerical and structural aberrations

Frequency of pluripotent SC lines with recurrent numerical and structural aberrations
Frequency of pluripotent SC lines with recurrent numerical and structural aberrations

Mouse Embryonic Stem Cells

A general feature of mESCs is the alteration of the chromosome number. Extensive cytogenetic studies on 540 [Sugawara et al., 2006] and 97 [Kim et al., 2013] mESC lines described high heterogeneity in numerical aberrations, detected by conventional banding techniques, SKY or CGH assays, with recurrent trisomies of chromosomes 8 and 11. Similar results were obtained in our and other laboratories [Liu et al., 1997; Rebuzzini et al., 2008a, b, 2012; Gaztelumendi and Nogués, 2014; Luft et al., 2014]. Also, several random subchromosomal variations were detected by CGH in mESC samples, among which the most recurrent were deletions of chromosomes 10qB and 14qC-14qE and the duplications of specific regions on chromosomes 1, 2 and 12 (table 1) [Ben-David and Benvenisty, 2012; Liang and Zhang, 2013]. However, the analysis until passage 22-34 of the chromosome complement of 4 mESC lines derived from B6C3F1 blastocysts showed a very heterogeneous spectrum of abnormalities that varied during the culture period, suggesting for these specific cell lines a lack of selective pressure in favor of specific chromosome complements [Rebuzzini et al., 2008a, b].

Parthenogenetic Embryonic Stem Cells

Parthenogenetic ESCs (pESCs), derived from non-human primate embryos [Cibelli et al., 2002; Vrana et al., 2003], display a normal diploid karyotype set of 42 chromosomes, even after 80-120 passages, analyzed using G-banding [Wei et al., 2011]. On the contrary, human pESCs [Kim et al., 2007; Revazova et al., 2007; Brevini et al., 2009, 2012) appear to be more variable. Some cell lines are stable, showing a normal karyotype after long-term culture (>50-100 passages) after standard G-banding analysis [Lin et al., 2007; Mai et al., 2007]; others lose 1 copy of the X chromosome with a frequency that increases during passages [Liu et al., 2011]. Chromosome deletions and translocations have also been reported in the hPES-2 cell line, when cultivated for 120 passages [Mai et al., 2007]. Few cytogenetic data are available on mouse pESC lines, reporting a stable karyotype during culture (up to passage 120), analyzed by standard G-banding [Shao et al., 2007; Ju et al., 2008; Shan et al., 2012].

Induced Pluripotent Stem Cells

iPSC lines seem to maintain a normal karyotype during early (∼5) culture passages [Takahashi et al., 2007]; whereas, during long-term culture (∼60 passages), large-scale screenings have evidenced the presence of several chromosomal (detected by standard karyotyping and FISH) and subchromosomal abnormalities such as translocations, duplications or deletions and point mutations (detected by array-CGH and SNP-arrays) [Takahashi et al., 2007; Mayshar et al., 2010; Gore et al., 2011; Martins-Taylor et al., 2011; Martins-Taylor and Xu, 2012]. Full trisomy of chromosomes 8 and 12, amplification of portions of chromosome 1, 17 or 20 (1q31.1, 17q21.1 and 20q11.21, respectively) and deletion of chromosome 8 and 17 (8q24.3 and 17q21.1, respectively) are the most frequent aberrations observed (table 1) [Aasen et al., 2008; Mayshar et al., 2010; Boulting et al., 2011; Martins-Taylor and Xu, 2012; Peterson and Loring, 2014]. Up to date, it is unclear whether the chromosomal and subchromosomal variations detected in iPSC lines originate early in individual cells and are propagated during the prolonged culture or are caused by the reprogramming process itself. The presence of recurrent aberrations may facilitate the adaptation process during reprogramming [Mayshar et al., 2010].

Mesenchymal Stromal Stem Cells

Most studies on chromosome variability have been performed on human mesenchymal stromal cells (MSCs) derived from bone marrow. Independent laboratories reported contrasting results on the accumulation of chromosomal aberrations during in vitro culture, a debate that still remains open [Ben-David et al., 2011, 2012; Sensebé et al., 2012]. Some authors showed that human bone marrow-derived MSCs remain chromosomally stable throughout long-term culture [Pittenger and Martin, 2004; Soukup et al., 2006; Bernardo et al., 2007; Zhang et al., 2007; Sensebé et al., 2012], whereas others claimed the occurrence of spontaneous numerical and structural chromosome aberrations within 10 passages of in vitro culture (table 2). To this regard, monosomy of chromosomes 13 and X (in 0.3-1.3% of cells analyzed) [Ben-David et al., 2011; Nikitina et al., 2011], trisomies of chromosome 5 and, less frequently, of chromosomes 8 and 20 (in 15-20% of cell analyzed) [Grigorian et al., 2010; Tarte et al., 2010] were reported after conventional G-banding.

Table 2

SSC lines in which numerical and structural aberrations were described

SSC lines in which numerical and structural aberrations were described
SSC lines in which numerical and structural aberrations were described

Similarly, discordant results have been obtained for MSCs derived from other tissues. Human adipose-derived MSC lines cultivated for more than 6 months seem to maintain chromosome stability [Meza-Zepeda et al., 2008; Ra et al., 2011; Yang et al., 2011]. Minor deletions and pericentromeric instabilities were detected by high-resolution microarray-based CGH analyses in few cells, but they were spontaneously eliminated from the population [Meza-Zepeda et al., 2008]. More recently, trisomy of chromosomes 8 or 17 and tetrasomy of chromosomes 11 or 17, acquired within few passages of in vitro culture, were described in human adipose-derived MSCs (at passage 2, 8-12% of cells were aneuploid, at passage 20, 18-28%) [Estrada et al., 2013]. Human dental pulp MSCs showed a normal karyotype until the Hayflick limit (passage 65) [Gronthos et al., 2000; Suchánek et al., 2007; Tamaki et al., 2013]. Beyond this limit, some chromosome abnormalities have been described [2n = 44,XX,t(13;14),-22 in 3% of cells analyzed; Suchánek et al., 2007]. More recently, polyploid aberrant metaphases (loss of chromosomes 2, 3, 5, 6, 7, 8, 9, and 16 in ∼70% of the cells) were found in 4 human dental pulp-derived MSC lines, in association with the presence of ring chromosomes (table 2) [Duailibi et al., 2012]. Finally, data on chorionic villi-, human umbilical cord- and amniotic fluid-derived MSCs showed normal chromosome complements after 6-10 passages [Poloni et al., 2011; Wang et al., 2013; Ruan et al., 2014] or after 150 days of cryopreservation [Angelo et al., 2012].

High chromosomal instability is characteristic of murine bone marrow-derived MSCs [Fan et al., 2011]. Several papers showed the accumulation of both clonal numerical and structural chromosome abnormalities in different cell lines since the very early passages from their isolation (table 2). Double-minute chromosomes, clonal gain of chromosome 2, chromosomal imbalances, non-clonal gains or losses of several chromosomes were extensively described [Miura et al., 2006; Josse et al., 2010; Fan et al., 2011].

Hematopoietic Stem Cells

Expanded umbilical cord blood (UCB)-derived CD34+ SCs showed karyotype stability even after 20 and 43 days of culture [Tian et al., 2005]. A recent comprehensive analysis of 55 CD34+ hematopoietic SC samples derived from bone marrow, peripherical blood or UCB of healthy donors revealed that these cells preserved their correct chromosome complement [Ben-David et al., 2011]. However, a paper published in 2011 reported that already after 7-14 days of in vitro culture ∼10-15% of UCB-derived CD34+ SCs displayed various chromosomal abnormalities [i.e. 46,XX,t(2;8); 45,XX,-8; 45,XX,-7,6q+; tetraploidy of chromosome 13, loss of an X chromosome]. After 28 days of culture, loss of entire chromosomes was described (i.e. 45,XX,-19 in 10% of cells analyzed) (table 2) [Ge et al., 2011].

Endothelial Progenitor Cells

To our knowledge, few papers reported the karyotype analysis of endothelial progenitor cells (EPCs). Although samples of cord blood-derived EPCs showed a normal karyotype throughout the ex vivo expansion, some cord blood EPC samples showed a high incidence of chromosomal alterations during culture, such as tetraploidy (92,XXYY; 33% of cells at passage 2), several types of aneuploidy (+77,XX, 68% of cells at passages 2 and 4; +55,XX, 100% of cells from passage 2 to 8) including gain of chromosomes 11 and 14 (74% of cells at passage 10) (table 2) [Corselli et al., 2008]. On the contrary, peripheral blood-derived EPCs showed a normal karyotype during in vitro culture (40-60 cell population doublings) [Untergasser et al., 2006].

Neural Stem Cells

Neural progenitor stem cells (NSCs) show a limited capacity of mitotic expansion in vitro; for this reason, few papers tackled the analysis of their chromosome complement. NSCs display high variability in the chromosome complement when cultured. For example, neither numerical nor structural chromosome abnormalities were detected in both rat and human NSC samples, cultivated up to 70 passages and analyzed with G-banding [Bai et al., 2004; Jiang et al., 2008; Sareen et al., 2009]; on the contrary, using the same technique, other samples (human NSCs derived from the striatum fetal) displayed different numerical (from 60-70 chromosomes per metaphase) and structural chromosome aberrations [Wu et al., 2011]. Also, the analysis of more than 70 other human NSC samples identified trisomy of chromosomes 7, 10, 18, or 19 [Sareen et al., 2009; Ben-David et al., 2011] and monosomy of chromosome 18 [Ben-David et al., 2011] in long-term cultures (table 2).

The occurrence of chromosome aberrations in mouse NSCs after extensive passaging was also reported. NSCs derived from fetal mouse brain (analyzed within 39 passages) showed no clonal structural aberrations but clonal monosomy 17 in 6.1% of cells (at passage 21). NSCs derived from the subventricular zone displayed a higher frequency of clonal aneuploidies than structural aberrations. Trisomy of chromosome 1 was detected in 29% of subventricular zone-derived NSCs at passage 10 and in 62% at passage 40. Also, trisomies of chromosomes 18 and 19 and duplication of chromosome 19q were reported with a lower frequency (table 2) [Diaferia et al., 2011].

The effect of in vitro environmental conditions on genomic stability of cells received much attention during the last years. However, the variability among culture protocols applied in different laboratories for derivation and cultivation of SCs complicates the identification of the source of such variations.

The techniques used for cell detachment and disaggregation seem to be a major factor affecting the maintenance of genome integrity during long culture. Mechanical (i.e. pipetting in and out and flushing the medium until the colonies are detached and disaggregated) or manual (i.e. colonies are cut and removed using a blade) methods are more gentle passaging techniques for subculturing than the use of enzymes (i.e. trypsin or collagenase), being less aggressive and able to better preserve genome integrity [Buzzard et al., 2004; Mitalipova et al., 2005; Lefort et al., 2008]. A modified enzymatic dissociation solution (0.25% trypsin, 0.1% collagenase IV, 20% KSR, and 1 mM CaCl2 in PBS), in combination with manual dissection for bulk passaging, has been proposed for hESC dissociation, demonstrating the maintenance of a normal chromosome complement even after more than 100 passages [Suemori et al., 2006].

Several studies focused their attention on oxygen concentration during culture although with contrasting results. In some studies, physiological concentrations of O2 (ranging from 1 to 7%) seemed to significantly reduce the incidence of aneuploidies in hMSCs and promote the maintenance of genetic integrity [Holzwarth et al., 2010; Li et al., 2011; Tsai et al., 2011; Estrada et al., 2012]; whereas, in others, 1-5% O2 concentrations were found to increase the risk of aneuploidies, chromosome and microsatellite instability in mouse NSCs, human bone marrow MSCs and human adipose SCs, even at early passages [Oliveira et al., 2012; Ueyama et al., 2012]. Also, high rates of aneuploidy, gaps and breaks were reported in hESC lines cultured with air concentration of O2 (21%), in comparison with lower concentrations [Forsyth et al., 2006; Lim et al., 2011].

A fundamental component of the SC medium is the serum. However, its animal (calf or bovine) or artificial (knockout serum replacement of defined composition, only used for ESC culture) origin does not seem to play a role in the maintenance or in the alteration of the genome stability [Inzunza at al., 2005; Ludwig at al., 2006]. Similarly, the choice of a cell feeder layer (mouse embryonic or immortalized fibroblasts) or of supportive matrixes (i.e. gelatin, fibronectin or matrigel™) during the derivation and maintenance of ESC lines does not seem to influence either the onset or the restraint of aberrations in the ESC genome [Cowan et al., 2004; Draper et al., 2004; Guo et al., 2005; Maitra et al., 2005; Mitalipova et al., 2005; Imreh et al., 2006; Sugawara et al., 2006; Rebuzzini et al., 2008a, b].

It should not be forgotten that various ESC lines have been derived from different strains and at different time points of preimplantation embryonic development, when gene expression and DNA methylation vary significantly. Hence, a differing tolerance to genetic instability might be also a characteristic defined at the time of ESC derivation. Unfortunately, up to date, there are no studies that have undertaken these comparisons.

As evidenced in this review, SCs accumulate during long-term culture both numerical and structural aberrations. This genomic instability does not affect their stemness and, upon challenging, SCs achieve a differentiated phenotype [Rebuzzini et al., 2012, 2013; Gaztelumendi and Nogués, 2014; Luft et al., 2014]. Strikingly, there is a lack of studies that analytically investigate if and which type of aneuploidies is permissive for cell differentiation, an aspect that is central to the use of SCs in the clinic. Given the enormous investments in terms of human and economic resources undertaken for the use of SCs in regenerative medicine and the recent approval of their use as medicinal products, we foresee a timely discussion among scientists, clinicians and regulatory agencies to produce appropriate guidelines for SC derivation and culture.

1.
Aasen T, Raya A, Barrero MJ, Garreta E, Consiglio A, et al: Efficient and rapid generation of induced pluripotent stem cells from human keratinocytes. Nat Biotechnol 26:1276-1284 (2008).
[PubMed]
2.
Angelo PC, Ferreira AC, Fonseca VD, Frade SP, Ferreira CS, et al: Cryopreservation does not alter karyotype, multipotency, or NANOG/SOX2 gene expression of amniotic fluid mesenchymal stem cells. Genet Mol Res 11:1002-1012 (2012).
[PubMed]
3.
Bai Y, Hu Q, Li X, Wang Y, Lin C, et al: Telomerase immortalization of human neural progenitor cells. Neuroreport 15:245-249 (2004).
[PubMed]
4.
Bajada S, Mazakova I, Richardson JB, Ashammakhi N: Updates on stem cells and their applications in regenerative medicine. J Tissue Eng Regen Med 2:169-183 (2008).
[PubMed]
5.
Baker DE, Harrison NJ, Maltby E, Smith K, Moore HD, et al: Adaptation to culture of human embryonic stem cells and oncogenesis in vivo. Nat Biotechnol 25:207-215 (2007).
[PubMed]
6.
Ben-David U, Benvenisty N: High prevalence of evolutionarily conserved and species-specific genomic aberrations in mouse pluripotent stem cells. Stem Cells 30:612-622 (2012).
[PubMed]
7.
Ben-David U, Mayshar Y, Benvenisty N: Large-scale analysis reveals acquisition of lineage-specific chromosomal aberrations in human adult stem cells. Cell Stem Cell 9:97-102 (2011).
[PubMed]
8.
Ben-David U, Kopper O, Benvenisty N: Expanding the boundaries of embryonic stem cells. Cell Stem Cell 10:666-677 (2012).
[PubMed]
9.
Bernardo ME, Zaffaroni N, Novara F, Cometa AM, Avanzini MA, et al: Human bone marrow derived mesenchymal stem cells do not undergo transformation after long-term in vitro culture and do not exhibit telomere maintenance mechanisms. Cancer Res 67:9142-9149 (2007).
[PubMed]
10.
Blum B, Benvenisty N: The tumorigenicity of diploid and aneuploid human pluripotent stem cells. Cell Cycle 8:3822-3830 (2009).
[PubMed]
11.
Boulting GL, Kiskinis E, Croft GF, Amoroso MW, Oakley DH, et al: A functionally characterized test set of human induced pluripotent stem cells. Nat Biotechnol 29:279-286 (2011).
[PubMed]
12.
Brevini TA, Pennarossa G, Antonini S, Paffoni A, Tettamanti G, et al: Cell lines derived from human parthenogenetic embryos can display aberrant centriole distribution and altered expression levels of mitotic spindle check-point transcripts. Stem Cell Rev 5:340-352 (2009).
[PubMed]
13.
Brevini TA, Pennarossa G, Maffei S, Tettamanti G, Vanelli A, et al: Centrosome amplification and chromosomal instability in human and animal parthenogenetic cell lines. Stem Cell Rev 8:1076-1087 (2012).
[PubMed]
14.
Brimble SN, Zeng X, Weiler DA, Luo Y, Liu Y, et al: Karyotypic stability, genotyping, differentiation, feeder-free maintenance, and gene expression sampling in three human embryonic stem cell lines derived prior to August 9, 2001. Stem Cells Dev 13:585-597 (2004).
[PubMed]
15.
Buehr M, Meek S, Blair K, Yang J, Ure J, et al: Capture of authentic embryonic stem cells from rat blastocysts. Cell 135:1287-1298 (2008).
[PubMed]
16.
Buzzard JJ, Gough NM, Crook JM, Colman A: Karyotype of human ES cells during extended culture. Nat Biotechnol 22:381-382 (2004).
[PubMed]
17.
Caldas C, Brenton JD: Sizing up miRNAs as cancer genes. Nat Med 11:712-714 (2005).
[PubMed]
18.
Catalina P, Cobo F, Cortés JL, Nieto AI, Cabrera C, et al: Conventional and molecular cytogenetic diagnostic methods in stem cell research: a concise review. Cell Biol Int 31:861-869 (2007).
[PubMed]
19.
Cibelli JB, Grant KA, Chapman KB, Cunniff K, Worst T, et al: Parthenogenetic stem cells in nonhuman primates. Science 295:819 (2002).
[PubMed]
20.
Clark AT, Rodriguez RT, Bodnar MS, Abeyta MJ, Cedars MI, et al: Human STELLAR, NANOG, and GDF3 genes are expressed in pluripotent cells and map to chromosome 12p13, a hotspot for teratocarcinoma. Stem Cells 22:169-179 (2004).
[PubMed]
21.
Cockburn K, Rossant J: Making the blastocyst: lessons from the mouse. J Clin Invest 120:995-1003 (2010).
[PubMed]
22.
Corselli M, Parodi A, Mogni M, Sessarego N, Kunkl A, et al: Clinical scale ex vivo expansion of cord blood-derived outgrowth endothelial progenitor cells is associated with high incidence of karyotype aberrations. Exp Hematol 36:340-349 (2008).
[PubMed]
23.
Cowan CA, Klimanskaya I, McMahon J, Atienza J, Witmyer J, et al: Derivation of embryonic stem-cell lines from human blastocysts. N Engl J Med 350:1353-1356 (2004).
[PubMed]
24.
Diaferia GR, Conti L, Redaelli S, Cattaneo M, Mutti C, et al: Systematic chromosomal analysis of cultured mouse neural stem cell lines. Stem Cells Dev 20:1411-1423 (2011).
[PubMed]
25.
Draper JS, Smith K, Gokhale P, Moore HD, Maltby E, et al: Recurrent gain of chromosomes 17q and 12 in cultured human embryonic stem cells. Nat Biotechnol 22:53-54 (2004).
[PubMed]
26.
Duailibi MT, Kulikowski LD, Duailibi SE, Lipay MV, Melaragno MI, et al: Cytogenetic instability of dental pulp stem cell lines. J Mol Histol 43:89-94 (2012).
[PubMed]
27.
Estrada JC, Albo C, Benguría A, Dopazo A, López-Romero P, et al: Culture of human mesenchymal stem cells at low oxygen tension improves growth and genetic stability by activating glycolysis. Cell Death Differ 19:743-755 (2012).
[PubMed]
28.
Estrada JC, Torres Y, Benguría A, Dopazo A, Roche E, et al: Human mesenchymal stem cell-replicative senescence and oxidative stress are closely linked to aneuploidy. Cell Death Dis 4:e691 (2013).
[PubMed]
29.
Evans MJ, Kaufman MH: Establishment in culture of pluripotential cells from mouse embryos. Nature 292:154-156 (1981).
[PubMed]
30.
Fan G, Wen L, Li M, Li C, Luo B, et al: Isolation of mouse mesenchymal stem cells with normal ploidy from bone marrows by reducing oxidative stress in combination with extracellular matrix. BMC Cell Biol 12:30 (2011).
[PubMed]
31.
Fang ZF, Gai H, Huang YZ, Li SG, Chen XJ, et al: Rabbit embryonic stem cell lines derived from fertilized, parthenogenetic or somatic cell nuclear transfer embryos. Exp Cell Res 312:3669-3682 (2006).
[PubMed]
32.
Forsyth NR, Musio A, Vezzoni P, Simpson AH, Noble BS, McWhir J: Physiologic oxygen enhances human embryonic stem cell clonal recovery and reduces chromosomal abnormalities. Cloning Stem Cells 8:16-23 (2006).
[PubMed]
33.
Franco Lambert AP, Fraga Zandonai A, Bonatto D, Cantarelli Machado D, Pêgas Henriques JA: Differentiation of human adipose-derived adult stem cells into neuronal tissue: does it work? Differentiation 77:221-228 (2009).
[PubMed]
34.
Gaztelumendi N, Nogués C: Chromosome instability in mouse embryonic stem cells. Sci Rep 4:5324 (2014).
[PubMed]
35.
Ge J, Cai H, Tan WS: Chromosomal stability during ex vivo expansion of UCB CD34(+) cells. Cell Prolif 44:550-557 (2011).
[PubMed]
36.
Gore A, Li Z, Fung HL, Young JE, Agarwal S, et al: Somatic coding mutations in human induced pluripotent stem cells. Nature 471:63-67 (2011).
[PubMed]
37.
Grigorian AS, Kruglyakov PV, Taminkina UA, Efimova OA, Pendina AA, et al: Alterations of cytological and karyological profile of human mesenchymal stem cells during in vitro culturing. Bull Exp Biol Med 150:125-130 (2010).
[PubMed]
38.
Grompe M: Tissue stem cells: new tools and functional diversity. Cell Stem Cell 10:685-689 (2012).
[PubMed]
39.
Gronthos S, Mankani M, Brahim J, Robey PG, Shi S: Postnatal human dental pulp stem cells (DPSCs) in vitro and in vivo. Proc Natl Acad Sci USA 97:13625-13630 (2000).
[PubMed]
40.
Guo J, Jauch A, Heidi HG, Schoell B, Erz D, et al: Multicolor karyotype analyses of mouse embryonic stem cells. In Vitro Cell Dev Biol Anim 41:278-283 (2005).
[PubMed]
41.
Hayes B, Fagerlie SR, Ramakrishnan A, Baran S, Harkey M, et al: Derivation, characterization, and in vitro differentiation of canine embryonic stem cells. Stem Cells 26:465-473 (2008).
[PubMed]
42.
Holzwarth C, Vaegler M, Gieseke F, Pfister SM, Handgretinger R, et al: Low physiologic oxygen tensions reduce proliferation and differentiation of human multipotent mesenchymal stromal cells. BMC Cell Biol 11:11 (2010).
[PubMed]
43.
Imreh MP, Gertow K, Cedervall J, Unger C, Holmberg K, et al: In vitro culture conditions favoring selection of chromosomal abnormalities in human ES cells. J Cell Biochem 99:508-516 (2006).
[PubMed]
44.
International Stem Cell Initiative: Screening ethnically diverse human embryonic stem cells identifies a chromosome 20 minimal amplicon conferring growth advantage. Nat Biotechnol 29:1132-1144 (2011).
[PubMed]
45.
Inzunza J, Sahlén S, Holmberg K, Strömberg AM, Teerijoki H, et al: Comparative genomic hybridization and karyotyping of human embryonic stem cells reveals the occurrence of an isodicentric X chromosome after long-term cultivation. Mol Hum Reprod 10:461-466 (2004).
[PubMed]
46.
Inzunza J, Gertow K, Strömberg MA, Matilainen E, Blennow E, et al: Derivation of human embryonic stem cell lines in serum replacement medium using postnatal human fibroblasts as feeder cells. Stem Cells 23:544-549 (2005).
[PubMed]
47.
Jiang X, Xu R, Yang Z, Jin P, Xu Q, et al: Experimental study on trace marking and oncogenicity of neural stem cells derived from bone marrow. Cell Mol Neurobiol 28:689-711 (2008).
[PubMed]
48.
Jiang Y, Jahagirdar BN, Reinhardt RL, Schwartz RE, Keene CD, et al: Pluripotency of mesenchymal stem cells derived from adult marrow. Nature 418:41-49 (2002).
[PubMed]
49.
Josse C, Schoemans R, Niessen NA, Delgaudine M, Hellin AC, et al: Systematic chromosomal aberrations found in murine bone marrow-derived mesenchymal stem cells. Stem Cells Dev 19:1167-1173 (2010).
[PubMed]
50.
Ju JY, Park CY, Gupta MK, Uhm SJ, Paik EC, et al: Establishment of stem cell lines from nuclear transferred and parthenogenetically activated mouse oocytes for therapeutic cloning. Fertil Steril 89:1314-1323 (2008).
[PubMed]
51.
Katari R, Peloso A, Orlando G: Tissue engineering and regenerative medicine: semantic considerations for an evolving paradigm. Front Bioeng Biotechnol 2:57 (2015).
[PubMed]
52.
Kim K, Ng K, Rugg-Gunn PJ, Shieh JH, Kirak O, et al: Recombination signatures distinguish embryonic stem cells derived by parthenogenesis and somatic cell nuclear transfer. Cell Stem Cell 1:346-352 (2007).
[PubMed]
53.
Kim YM, Lee JY, Xia L, Mulvihill JJ, Li S: Trisomy 8: a common finding in mouse embryonic stem (ES) cell lines. Mol Cytogenet 6:3 (2013).
[PubMed]
54.
Lefort N, Feyeux M, Bas C, Féraud O, Bennaceur-Griscelli A, et al: Human embryonic stem cells reveal recurrent genomic instability at 20q11.21. Nat Biotechnol 26:1364-1366 (2008).
[PubMed]
55.
Lefort N, Perrier AL, Laâbi Y, Varela C, Peschanski M: Human embryonic stem cells and genomic instability. Regen Med 4:899-909 (2009).
[PubMed]
56.
Li P, Tong C, Mehrian-Shai R, Jia L, Wu N, et al: Germline competent embryonic stem cells derived from rat blastocysts. Cell 135:1299-1310 (2008).
[PubMed]
57.
Li R, Chen B, Wang G, Yu B, Ren G, Ni G: Effects of mechanical strain on oxygen free radical system in bone marrow mesenchymal stem cells from children. Injury 42:753-757 (2011).
[PubMed]
58.
Liang G, Zhang Y: Genetic and epigenetic variations in iPSCs: potential causes and implications for application. Cell Stem Cell 13:149-159 (2013).
[PubMed]
59.
Lim HJ, Han J, Woo DH, Kim SE, Kim SK, et al: Biochemical and morphological effects of hypoxic environment on human embryonic stem cells in long-term culture and differentiating embryoid bodies. Mol Cells 31:123-132 (2011).
[PubMed]
60.
Lin G, OuYang Q, Zhou X, Gu Y, Yuan D, et al: A highly homozygous and parthenogenetic human embryonic stem cell line derived from a one-pronuclear oocyte following in vitro fertilization procedure. Cell Res 17:999-1007 (2007).
[PubMed]
61.
Liu LN, Wang G, Hendricks K, Lee K, Bohnlein E, et al: Comparison of drug and cell-based delivery: engineered adult mesenchymal stem cells expressing soluble tumor necrosis factor receptor II prevent arthritis in mouse and rat animal models. Stem Cells Transl Med 2:362-375 (2013).
[PubMed]
62.
Liu W, Yin Y, Jiang Y, Kou C, Luo Y, et al: Genetic and epigenetic X-chromosome variations in a parthenogenetic human embryonic stem cell line. J Assist Reprod Genet 28:303-313 (2011).
[PubMed]
63.
Liu X, Wu H, Loring J, Hormuzdi S, Disteche CM, et al: Trisomy eight in ES cells is a common potential problem in gene targeting and interferes with germ line transmission. Dev Dyn 209:85-91 (1997).
[PubMed]
64.
Liyanage M, Coleman A, du Manoir S, Veldman T, McCormack S, et al: Multicolour spectral karyotyping of mouse chromosomes. Nat Genet 14:312-315 (1996).
[PubMed]
65.
Ludwig TE, Levenstein ME, Jones JM, Berggren WT, Mitchen ER, et al: Derivation of human embryonic stem cells in defined conditions. Nat Biotechnol 24:185-187 (2006).
[PubMed]
66.
Luft S, Pignalosa D, Nasonova E, Arrizabalaga O, Helm A, et al: Fate of D3 mouse embryonic stem cells exposed to X-rays or carbon ions. Mutat Res Genet Toxicol Environ Mutagen 760:56-63 (2014).
[PubMed]
67.
Mai Q, Yu Y, Li T, Wang L, Chen MJ, et al: Derivation of human embryonic stem cell lines from parthenogenetic blastocysts. Cell Res 17:1008-1019 (2007).
[PubMed]
68.
Maitra A, Arking DE, Shivapurkar N, Ikeda M, Stastny V, et al: Genomic alterations in cultured human embryonic stem cells. Nat Genet 37:1099-1103 (2005).
[PubMed]
69.
Martin GR: Isolation of a pluripotent cell line from early mouse embryos cultured in medium conditioned by teratocarcinoma stem cells. Proc Natl Acad Sci USA 78:7634-7638 (1981).
[PubMed]
70.
Martins-Taylor K, Xu RH: Concise review: genomic stability of human induced pluripotent stem cells. Stem Cells 30:22-27 (2012).
[PubMed]
71.
Martins-Taylor K, Nisler BS, Taapken SM, Compton T, Crandall L, et al: Recurrent copy number variations in human induced pluripotent stem cells. Nat Biotechnol 29:488-491 (2011).
[PubMed]
72.
Mayshar Y, Ben-David U, Lavon N, Biancotti JC, Yakir B, et al: Identification and classification of chromosomal aberrations in human induced pluripotent stem cells. Cell Stem Cell 7:521-531 (2010).
[PubMed]
73.
Merkle FT, Eggan K: Modeling human disease with pluripotent stem cells: from genome association to function. Cell Stem Cell 12:656-668 (2013).
[PubMed]
74.
Meza-Zepeda LA, Noer A, Dahl JA, Micci F, Myklebost O, Collas P: High-resolution analysis of genetic stability of human adipose tissue stem cells cultured to senescence. J Cell Mol Med 12:553-563 (2008).
[PubMed]
75.
Mitalipova MM, Rao RR, Hoyer DM, Johnson JA, Meisner LF, et al: Preserving the genetic integrity of human embryonic stem cells. Nat Biotechnol 23:19-20 (2005).
[PubMed]
76.
Miura M, Miura Y, Padilla-Nash HM, Molinolo AA, Fu B, et al: Accumulated chromosomal instability in murine bone marrow mesenchymal stem cells leads to malignant transformation. Stem Cells 24:1095-1103 (2006).
[PubMed]
77.
Navarro P, Chambers I, Karwacki-Neisius V, Chureau C, Morey C, et al: Molecular coupling of Xist regulation and pluripotency. Science 321:1693-1695 (2008).
[PubMed]
78.
Neri T, Merico V, Fiordaliso F, Salio M, Rebuzzini P, et al: The differentiation of cardiomyocytes from mouse embryonic stem cells is altered by dioxin. Toxicol Lett 202:226-236 (2011).
[PubMed]
79.
Nikitina VA, Osipova EY, Katosova LD, Rumyantsev SA, Skorobogatova EV, et al: Study of genetic stability of human bone marrow multipotent mesenchymal stromal cells. Bull Exp Biol Med 150:627-631 (2011).
[PubMed]
80.
Okita K, Ichisaka T, Yamanaka S: Generation of germline-competent induced pluripotent stem cells. Nature 448:313-317 (2007).
[PubMed]
81.
Oliveira PH, Boura JS, Abecasis MM, Gimble JM, da Silva CL, Cabral JM: Impact of hypoxia and long-term cultivation on the genomic stability and mitochondrial performance of ex vivo expanded human stem/stromal cells. Stem Cell Res 9:225-236 (2012).
[PubMed]
82.
Oliveira PH, da Silva CL, Cabral JM: Concise review: Genomic instability in human stem cells: current status and future challenges. Stem Cells 32:2824-2832 (2014).
[PubMed]
83.
Peiffer DA, Le JM, Steemers FJ, Chang W, Jenniges T, et al: High-resolution genomic profiling of chromosomal aberrations using Infinium whole-genome genotyping. Genome Res 16:1136-1148 (2006).
[PubMed]
84.
Peterson SE, Loring JF: Genomic instability in pluripotent stem cells: implications for clinical applications. J Biol Chem 289:4578-4584 (2014).
[PubMed]
85.
Pittenger MF, Martin BJ: Mesenchymal stem cells and their potential as cardiac therapeutics. Circ Res 95:9-20 (2004).
[PubMed]
86.
Poloni A, Maurizi G, Babini L, Serrani F, Berardinelli E, et al: Human mesenchymal stem cells from chorionic villi and amniotic fluid are not susceptible to transformation after extensive in vitro expansion. Cell Transplant 20:643-654 (2011).
[PubMed]
87.
Porada CD, Atala AJ, Almeida-Porada G: The hematopoietic system in the context of regenerative medicine. Methods S1046-2023:30057-30058 (2015).
[PubMed]
88.
Ra JC, Shin IS, Kim SH, Kang SK, Kang BC, et al: Safety of intravenous infusion of human adipose tissue-derived mesenchymal stem cells in animals and humans. Stem Cells Dev 20:1297-1308 (2011).
[PubMed]
89.
Rebuzzini P, Neri T, Mazzini G, Zuccotti M, Redi CA, Garagna S: Karyotype analysis of the euploid cell population of a mouse embryonic stem cell line revealed a high incidence of chromosome abnormalities that varied during culture. Cytogenet Genome Res 121:18-24 (2008a).
[PubMed]
90.
Rebuzzini P, Neri T, Zuccotti M, Redi CA, Garagna S: Chromosome number variation in three mouse embryonic stem cell lines during culture. Cytotechnology 58:17-23 (2008b).
[PubMed]
91.
Rebuzzini P, Zuccotti M, Redi CA, Garagna S: Genome stability in embryonic stem cells, in Atwood CS (ed): Embryonic Stem Cells - Recent Advantages in Pluripotent Stem Cell-Based Regenerative Medicine, pp 399-410 (InTech, Rijeka 2011).
92.
Rebuzzini P, Pignalosa D, Mazzini G, Di Liberto R, Coppola A, et al: Mouse embryonic stem cells that survive γ-rays exposure maintain pluripotent differentiation potential and genome stability. J Cell Physiol 227:1242-1249 (2012).
[PubMed]
93.
Rebuzzini P, Fassina L, Mulas F, Bellazzi R, Redi CA, et al: Mouse embryonic stem cells irradiated with γ-rays differentiate into cardiomyocytes but with altered contractile properties. Mutat Res 756:37-45 (2013).
[PubMed]
94.
Revazova ES, Turovets NA, Kochetkova OD, Kindarova LB, Kuzmichev LN, et al: Patient-specific stem cell lines derived from human parthenogenetic blastocysts. Cloning Stem Cells 9:432-449 (2007).
[PubMed]
95.
Rosler ES, Fisk GJ, Ares X, Irving J, Miura T, et al: Long-term culture of human embryonic stem cells in feeder-free conditions. Dev Dyn 229:259-274 (2004).
[PubMed]
96.
Rossant J: Stem cells from the mammalian blastocyst. Stem Cells 19:477-482 (2001).
[PubMed]
97.
Ruan ZB, Zhu L, Yin YG, Chen GC: Karyotype stability of human umbilical cord-derived mesenchymal stem cells during in vitro culture. Exp Ther Med 8:1508-1512 (2014).
[PubMed]
98.
Sanlaville D, Lapierre JM, Turleau C, Coquin A, Borck G, et al: Molecular karyotyping in human constitutional cytogenetics. Eur J Med Genet 48:214-231 (2005).
[PubMed]
99.
Sareen D, McMillan E, Ebert AD, Shelley BC, Johnson JA, et al: Chromosome 7 and 19 trisomy in cultured human neural progenitor cells. PLoS One 4:e7630 (2009).
[PubMed]
100.
Schröck E, Blume C, Meffert MC, du Manoir S, Bersch W, et al: Recurrent gain of chromosome arm 7q in low-grade astrocytic tumors studied by comparative genomic hybridization. Genes Chromosomes Cancer 15:199-205 (1996).
[PubMed]
101.
Seiler AE, Spielmann H: The validated embryonic stem cell test to predict embryotoxicity in vitro. Nat Protoc 6:961-978 (2011).
[PubMed]
102.
Sensebé L, Tarte K, Galipeau J, Krampera M, Martin I, et al: Limited acquisition of chromosomal aberrations in human adult mesenchymal stromal cells. Cell Stem Cell 10:9-10 (2012).
[PubMed]
103.
Shan ZY, Wu YS, Shen XH, Li X, Xue Y, et al: Aggregation of pre-implantation embryos improves establishment of parthenogenetic stem cells and expression of imprinted genes. Dev Growth Differ 54:481-488 (2012).
[PubMed]
104.
Shao H, Wei Z, Wang L, Wen L, Duan B, et al: Generation and characterization of mouse parthenogenetic embryonic stem cells containing genomes from non-growing and fully grown oocytes. Cell Biol Int 31:1336-1344 (2007).
[PubMed]
105.
Smith AG: Embryo-derived stem cells: of mice and men. Annu Rev Cell Dev Biol 17:435-462 (2001).
[PubMed]
106.
Soukup T, Mokrý J, Karbanová J, Pytlík R, Suchomel P, Kucerová L: Mesenchymal stem cells isolated from the human bone marrow: cultivation, phenotypic analysis and changes in proliferation kinetics. Acta Medica (Hradec Kralove) 49:27-33 (2006).
[PubMed]
107.
Spits C, Mateizel I, Geens M, Mertzanidou A, Staessen C, et al: Recurrent chromosomal abnormalities in human embryonic stem cells. Nat Biotechnol 26:1361-1363 (2008).
[PubMed]
108.
Suchánek J, Soukup T, Ivancaková R, Karbanová J, Hubková V, et al: Human dental pulp stem cells-isolation and long term cultivation. Acta Medica (Hradec Kralove) 50:195-201 (2007).
[PubMed]
109.
Suemori H, Yasuchika K, Hasegawa K, Fujioka T, Tsuneyoshi N, Nakatsuji N: Efficient establishment of human embryonic stem cell lines and long-term maintenance with stable karyotype by enzymatic bulk passage. Biochem Biophys Res Commun 345:926-932 (2006).
[PubMed]
110.
Sugawara A, Goto K, Sotomaru Y, Sofuni T, Ito T: Current status of chromosomal abnormalities in mouse embryonic stem cell lines used in Japan. Comp Med 56:31-34 (2006).
[PubMed]
111.
Takahashi K, Yamanaka S: Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 126:663-676 (2006).
[PubMed]
112.
Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, et al: Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 131:861-872 (2007).
[PubMed]
113.
Tamaki Y, Nakahara T, Ishikawa H, Sato S: In vitro analysis of mesenchymal stem cells derived from human teeth and bone marrow. Odontology 101:121-132 (2013).
[PubMed]
114.
Tarte K, Gaillard J, Lataillade JJ, Fouillard L, Becker M, et al: Clinical-grade production of human mesenchymal stromal cells: occurrence of aneuploidy without transformation. Blood 115:1549-1553 (2010).
[PubMed]
115.
Thomsen GM, Gowing G, Svendsen S, Svendsen CN: The past, present and future of stem cell clinical trials for ALS. Exp Neurol 262:127-137 (2014).
[PubMed]
116.
Thomson JA, Kalishman J, Golos TG, Durning M, Harris CP, et al: Isolation of a primate embryonic stem cell line. Proc Natl Acad Sci USA 92:7844-7848 (1995).
[PubMed]
117.
Thomson JA, Kalishman J, Golos TG, Durning M, Harris CP, Hearn JP: Pluripotent cell lines derived from common marmoset (Callithrix jacchus) blastocysts. Biol Reprod 55:254-259 (1996).
[PubMed]
118.
Thomson JA, Itskovitz-Eldor J, Shapiro SS, Waknitz MA, Swiergiel JJ, et al: Embryonic stem cell lines derived from human blastocysts. Science 282:1145-1147 (1998).
[PubMed]
119.
Tian H, Huang S, Gong F, Tian L, Chen Z: Karyotyping, immunophenotyping, and apoptosis analyses on human hematopoietic precursor cells derived from umbilical cord blood following long-term ex vivo expansion. Cancer Genet Cytogenet 157:33-36 (2005).
[PubMed]
120.
Till JE, McCullough EA: A direct measurement of the radiation sensitivity of normal mouse bone marrow cells. Radiat Res 14:213-222 (1961).
[PubMed]
121.
Tsai CC, Chen YJ, Yew TL, Chen LL, Wang JY, et al: Hypoxia inhibits senescence and maintains mesenchymal stem cell properties through down-regulation of E2A-p21 by HIF-TWIST. Blood 117:459-469 (2011).
[PubMed]
122.
Ueyama H, Horibe T, Hinotsu S, Tanaka T, Inoue T, et al: Chromosomal variability of human mesenchymal stem cells cultured under hypoxic conditions. J Cell Mol Med 16:72-82 (2012).
[PubMed]
123.
Untergasser G, Koeck R, Wolf D, Rumpold H, Ott H, et al: CD34+/CD133- circulating endothelial precursor cells (CEP): characterization, senescence and in vivo application. Exp Gerontol 41:600-608 (2006).
[PubMed]
124.
Vrana KE, Hipp JD, Goss AM, McCool BA, Riddle DR, et al: Nonhuman primate parthenogenetic stem cells. Proc Natl Acad Sci USA 100 Suppl 1:11911-11916 (2003).
[PubMed]
125.
Wang S, Tang X, Niu Y, Chen H, Li B, et al: Generation and characterization of rabbit embryonic stem cells. Stem Cells 25:481-489 (2007).
[PubMed]
126.
Wang Y, Zhang Z, Chi Y, Zhang Q, Xu F, et al: Long-term cultured mesenchymal stem cells frequently develop genomic mutations but do not undergo malignant transformation. Cell Death Dis 4:e950 (2013).
[PubMed]
127.
Wei Q, Sun Z, He X, Tan T, Lu B, et al: Derivation of rhesus monkey parthenogenetic embryonic stem cells and its microRNA signature. PLoS One 6:e25052 (2011).
[PubMed]
128.
Werbowetski-Ogilvie TE, Bossé M, Stewart M, Schnerch A, Ramos-Mejia V, et al: Characterization of human embryonic stem cells with features of neoplastic progression. Nat Biotechnol 27:91-97 (2009).
[PubMed]
129.
Wernig M, Meissner A, Foreman R, Brambrink T, Ku M, et al: In vitro reprogramming of fibroblasts into a pluripotent ES-cell-like state. Nature 448:318-324 (2007).
[PubMed]
130.
Wu W, He Q, Li X, Zhang X, Lu A, et al: Long-term cultured human neural stem cells undergo spontaneous transformation to tumor-initiating cells. Int J Biol Sci 7:892-901 (2011).
[PubMed]
131.
Yang XF, He X, He J, Zhang LH, Su XJ, et al: High efficient isolation and systematic identification of human adipose-derived mesenchymal stem cells. J Biomed Sci 18:59 (2011).
[PubMed]
132.
Zhang ZX, Guan LX, Zhang K, Wang S, Cao PC, et al: Cytogenetic analysis of human bone marrow-derived mesenchymal stem cells passaged in vitro. Cell Biol Int 31:645-648 (2007).
[PubMed]