Next Article in Journal
The Zebrafish (Danio rerio) Is a Relevant Model for Studying Sex-Specific Effects of 17β-Estradiol in the Adult Heart
Next Article in Special Issue
Meniscus-Derived Matrix Bioscaffolds: Effects of Concentration and Cross-Linking on Meniscus Cellular Responses and Tissue Repair
Previous Article in Journal
Expression Profile of the Chromosome 14 MicroRNA Cluster (C14MC) Ortholog in Equine Maternal Circulation throughout Pregnancy and Its Potential Implications
Previous Article in Special Issue
Current and Future Concepts for the Treatment of Impaired Fracture Healing
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Usefulness of Mesenchymal Cell Lines for Bone and Cartilage Regeneration Research

by
M. Piñeiro-Ramil
1,2,3,*,
C. Sanjurjo-Rodríguez
1,2,3,4,
R. Castro-Viñuelas
1,2,3,
S. Rodríguez-Fernández
1,2,3,
I.M. Fuentes-Boquete
1,2,3,4,
F.J. Blanco
4,5 and
S.M. Díaz-Prado
1,2,3,4,*
1
Grupo de Investigación en Terapia Celular e Medicina Rexenerativa, Departamento de Fisioterapia, Medicina e Ciencias Biomédicas, Facultade de Ciencias da Saúde, Universidade da Coruña (UDC), Campus de A Coruña, 15006 A Coruña, Spain
2
Grupo de Investigación en Terapia Celular e Medicina Rexenerativa, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario A Coruña (CHUAC), Servizo Galego de Saúde (SERGAS), Universidade da Coruña (UDC), 15006 A Coruña, Spain
3
Grupo de Investigación en Terapia Celular e Medicina Rexenerativa, Centro de Investigacións Científicas Avanzadas (CICA), Agrupación Estratéxica entre o CICA e o Instituto de Investigación Biomédica de A Coruña (INIBIC), Universidade da Coruña (UDC), 15071 A Coruña, Spain
4
Centro de Investigación Biomédica en Red (CIBER) de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain
5
Grupo de Investigación en Reumatología (GIR), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario A Coruña (CHUAC), Servizo Galego de Saúde (SERGAS), 15006 A Coruña, Spain
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2019, 20(24), 6286; https://doi.org/10.3390/ijms20246286
Submission received: 17 October 2019 / Revised: 10 December 2019 / Accepted: 11 December 2019 / Published: 13 December 2019

Abstract

:
The unavailability of sufficient numbers of human primary cells is a major roadblock for in vitro repair of bone and/or cartilage, and for performing disease modelling experiments. Immortalized mesenchymal stromal cells (iMSCs) may be employed as a research tool for avoiding these problems. The purpose of this review was to revise the available literature on the characteristics of the iMSC lines, paying special attention to the maintenance of the phenotype of the primary cells from which they were derived, and whether they are effectively useful for in vitro disease modeling and cell therapy purposes. This review was performed by searching on Web of Science, Scopus, and PubMed databases from 1 January 2015 to 30 September 2019. The keywords used were ALL = (mesenchymal AND (“cell line” OR immortal*) AND (cartilage OR chondrogenesis OR bone OR osteogenesis) AND human). Only original research studies in which a human iMSC line was employed for osteogenesis or chondrogenesis experiments were included. After describing the success of the immortalization protocol, we focused on the iMSCs maintenance of the parental phenotype and multipotency. According to the literature revised, it seems that the maintenance of these characteristics is not guaranteed by immortalization, and that careful selection and validation of clones with particular characteristics is necessary for taking advantage of the full potential of iMSC to be employed in bone and cartilage-related research.

1. Introduction

Therapeutic options capable of restoring the physiological properties of bone and cartilage are still lacking [1,2]. Due to the increase in life expectancy of the population, the incidence of musculoskeletal disorders, such as fractures, osteoporosis, maxillofacial pathologies, and rheumatic diseases such as osteoarthritis, is rising [3,4]. In this context, tissue engineering has emerged as a potential alternative treatment that could provide biological tissue substitutes for replacing the damaged ones, using scaffolds and cells. However, although many efforts have been made, very few tissue engineering techniques have been translated into clinical practice, and the ideal scaffold for engineering bone and cartilage substitutes has not yet been developed [1,4,5,6].
Tissue engineering techniques, combining scaffolds and cells, must undergo in vitro testing before translation into clinic or preclinical models. Human mesenchymal stromal cells (MSCs) are often employed in bone and cartilage tissue engineering approaches because of their proliferation and multidifferentiation abilities [7,8,9]. However, much more research is still needed to optimize isolation, expansion, differentiation, and preconditioning of MSCs before implantation [10] to maximize cell retention and viability and, in the case of bone engineering, to improve vascular network formation [1]. Furthermore, there are still some concerns about biosafety and efficacy of MSCs for clinical applications [11] as well as several other associated risk factors, such as the MSC differentiation status [12]. The unavailability of sufficient numbers of human primary cells is likely to delay the advance of research in these fields. This lack of primary MSCs is not only due to them being scarce (mainly healthy ones), but also the limited lifespan of the cells after isolation and in vitro culture. It has been described that human MSCs can achieve a maximum of 30–40 population doublings (PDs) in vitro before they lose their proliferation potential [13,14,15]. In addition, heterogeneity increases between MSCs which have been derived from the same donor at different passages, and the expanded MSCs progressively lose their differentiation potential [7,8,13]. There is also variability among donors [16], apart from the single MSC-derived clones isolated from the same donor [17,18,19]. For these reasons, human cell lines, and specifically iMSC lines, are only being used for research purposes.
Nowadays, a high number of MSC lines that display specific characteristics and differentiation capabilities have been generated and are valuable tools as part of models of disease and tissue repairing strategies. Different MSC lines have been employed for testing [17,20,21,22,23,24,25] or producing [26,27] engineered scaffolds for skeletal applications, and for both investigating the MSC differentiation process [28,29,30,31,32,33,34] and finding new ways to improve it [35,36,37,38,39]. Additionally, these cell lines have also been used for analyzing functional makers [19,40] or even for exploring their roles in different diseases, such as osteoarthritis [41,42].
The aim of this review was to analyze the characteristics of the MSC lines that are being used currently. Also, we aimed to investigate whether the MSC lines keep the phenotype of the primary cells from which they were derived, and if they could indeed be good models of tissue regeneration and disease.

2. Methodology

This review was carried out by employing Web of Science, Scopus, and PubMed databases from 1st January 2015 to 30th September 2019. In order to identify the human immortal MSC (iMSC) lines that are being used currently in the fields of bone and cartilage research, the keywords used were ALL = (mesenchymal AND (“cell line” OR immortal*) AND (cartilage OR chondrogenesis OR bone OR osteogenesis) AND human). Only original research studies were included in the analysis. References of the selected articles were included when relevant, and duplicates were excluded. After screening the title/abstract or full text, articles in which no human iMSC line was employed for osteogenesis or chondrogenesis experiments were excluded. The PRISMA flow diagram [43] is shown in Figure 1. This way, we identified 38 human iMSC lines derived from MSCs of single or “pooled” donors and whose osteogenic and/or chondrogenic potential had been tested.
For each iMSC line identified, we collected the following data: immortalization genes and method employed; tissue of origin and donor characteristics; if the iMSC line was clonal; whether it was tumorigenic and how its tumorigenicity had been assayed; and if it had been validated by short tandem repeat (STR) genotyping, as seen in Table 1. In addition, we investigated how its multidifferentiation (osteogenic, chondrogenic, and adipogenic) potential had been assessed and what results were obtained, as seen in Table 2. Afterwards, we described the immortalization strategies and their mechanism of action, as well as the outcomes and the characteristics of the iMSC lines revised, focusing on their osteogenic and chondrogenic capacities and potential usefulness for bone and cartilage regeneration research.

3. Immortal Mesenchymal Stromal Cell (iMSC) Lines

3.1. Immortalizing Human Adult MSCs

Immortalization is the process by which cells acquire an unlimited proliferation potential by bypassing senescence [106]. There are two types of senescence that cells must evade for their immortalization: replicative senescence, caused by telomere shortening and resulting chromosomal instability, and nonreplicative senescence, promoted by cellular stress, DNA damage, or oncogenic signals.
The initial in vitro growth arrest of human primary MSCs is presumed to be due to nonreplicative senescence, which is regulated by p53 and Rb-related pathways. In response to stress, the tumor suppressor p53 is phosphorylated and liberated from its binding to E3 ubiquitin ligase Mdm2, hence activating the senescence pathways. During quiescence, unphosphorylated Rb proteins control cell proliferation by binding and inhibiting E2F transcription factors, thus blocking cell cycle progression. During cell growth, signaling pathways that phosphorylate Rb proteins are activated, promoting its disassociation from E2F and allowing for the expression of E2F-dependent genes necessary for cell division [107,108]. Inhibition of p53 and inactivation of Rb by viral oncogenes have been shown to extend the life span of several cell types in culture, but telomeres maintenance is also needed for preventing replicative senescence [106]. Since human primary MSCs undergo progressive telomere shortening during serial passaging, human telomerase reverse transcriptase (hTERT) expression is needed to avoid telomere shortening [70]; otherwise, telomeres will shorten with every cell division until a critical threshold at which cells enter senescence.
Both simian virus 40 large T antigen (SV40LT) and human papillomavirus (HPV) E6/E7 gene transduction promote cell cycle progression by interfering with p53 and Rb-mediated pathways. SV40LT binds to these two proteins, thus releasing the activity of E2F transcription factors and avoiding growth arrest [107]. HPV E6/E7 proteins work in a similar manner, with p53 being the principal target of E6 and Rb being degraded via the ubiquitin proteasome pathway by the action of E7 [109]. SV40LT transduction has been employed for immortalizing MSCs derived from bone marrow from young and old donors [45,47], umbilical cord [49], cranial periosteum [51], coronal sutures [50], dental follicle [52], peripheral blood [48], and osteoarthritic cartilage [41]. SV40LT expression increases the lifespan of MSCs and usually raises its proliferation rate as well, but it has also been observed that this increased lifespan is not unlimited. Lee et al. (2015) reported that after more than 80 passages, SV40LT-transduced MSCs decreased their growth rate and entered senescence, indicating that this antigen is not enough for complete immortalization of MSCs [44].
HPV E6/E7 genes have also been used for immortalizing MSCs derived from bone marrow [54,58], and, in a similar way, E6/E7-transduced MSCs have been reported to enter a period of growth arrest after 70 PDs, suggesting a limited effect of E6/E7 in prolonging lifespan [58]. The same occurs with the p16 antagonist Bmi1, which has been reported to extend the lifespan of ligament-derived MSCs [62], but also to be insufficient for immortalization, with Bmi1-transduced adipose tissue-derived MSCs entering senescence after 55–60 PDs [110]. Since neither SV40LT nor E6/E7 proteins can promote the telomere maintenance needed to achieve an unlimited lifespan, SV40LT and E6/E7-transduced MSCs reported as immortal MSCs must have acquired a mechanism to prevent telomere shortening, or they will eventually undergo replicative senescence. Nevertheless, short telomeres are a source of chromosomal instability, and, if p53 activity is inhibited by SV40LT or E6/E7 proteins, alterations resulting from this instability will increase the mutability of the genome and might eventually give rise to telomerase re-expression [106].
It has been stated that hTERT transduction allows senescence evasion while maintaining in vitro and in vivo osteogenic ability of MSCs [63,91]. Transduction of hTERT alone has been employed to generate iMSC lines, but, since hTERT has no effect over non-replicative senescence, it has also been reported to fail to immortalize MSCs derived from bone marrow [13,18,58,74,104] and adipose tissue [85]. Skårn et al. (2014) described that only one out of nine hTERT-transduced bone marrow-derived MSC clones was able to proliferate over 40 PDs, and even this clone showed a slow proliferation rate similar to that of primary MSCs [71]. Other authors have confirmed that hTERT-transduced MSCs displayed a lifespan similar to that of primary MSCs (about 30–40 PDs) [18,81]. Okamoto et al. (2002) observed that p16 expression was upregulated in hTERT-transduced MSCs during passaging, finally leading to senescence despite maintenance of telomeres length. Conversely, MSCs transduced with both hTERT and E6/E7 were able to proliferate during more than 80 PDs [18], overcoming senescence, in the same way that MSCs transduced with both hTERT and SV40LT acquired an unlimited lifespan [13,85,110].
However, in addition to giving rise to MSC lines derived from bone marrow from young and/or healthy donors [15,63,71], which may well be less prone to suffer nonreplicative senescence, hTERT transduction has also been shown to immortalize placenta-derived MSCs of fetal and maternal origin [91], umbilical cord-derived MSCs [86], periodontal ligament-derived MSCs [89], adipose tissue-derived MSCs [84], and, importantly, osteoarthritic cartilage-derived MSCs [94]. Therefore, whether hTERT is sufficient to immortalize MSCs remains controversial. Immortalization requirements could be dependent on cell characteristics, cell culture conditions, and any factors that influence proneness to nonreplicative senescence, and seems not to be related to tissue of origin.
Transduction of hTERT has also been employed in combination with E6/E7 [18,55,58,85,101,104] or SV40LT [13,85,110]. This combination of genes leaded to an unlimited proliferation potential of the cells, which could not be obtained with the transduction of one gene only [58,85]. In the study of Balducci et al. (2014), the combination of hTERT and SV40LT was more efficient than hTERT and E6/E7 in improving growth rate of adipose-tissue-derived MSCs, but both combinations were efficient in overcoming senescence [85]. In short, hTERT alone, SV40LT alone, or E6/E7 alone could be enough to achieve immortalization of MSCs or not, but the combination of p53/Rb repression together with a mechanism of telomeres maintenance has always proven to be successful.

3.2. Multidifferentiation Potential of iMSCs

3.2.1. Osteogenic Potential

Out of the 35 iMSC lines included in this review whose osteogenic potential had been tested, only one line showed no mineralization ability. This hASCs-T cell line was derived from adipose tissue and immortalized with a combination of hTERT and SV40LT [85]. All the resting iMSCs were capable of osteogenically differentiating upon induction, as shown by the standard histochemical staining—alizarin red staining (ARS), Von Kossa staining (VKS), and alkaline phosphatase staining (APS)—and gene expression analysis of bone-related genes RUNX2, osteocalcin, alkaline phosphatase (ALP), BMP-2, bone sialoprotein (BSP), and COL1A1, as seen in Table 2.
In four out of the thirty iMSC lines which displayed osteogenic potential, this differentiation ability was increased in comparison with primary MSCs, in terms of mineralization [14,85] and osteogenesic-related gene expression [44,51]. This increment of the osteogenic potential neither seems to be related to tissue’s origin, since it has been detected in iMSCs from bone marrow [14,44], cranial periosteum [51], and adipose tissue origin [85], nor seems to be related to immortalization protocol, as two of these cell lines were transduced with SV40LT [44,51], one was transduced with hTERT alone [14] and the other one with hTERT and E6/E7 [85].
On the other hand, there were four iMSC lines that showed reduced osteogenic potential in comparison with primary MSCs, all of them transduced with hTERT alone: BMA13H, hASCs-T, GB/hTERT MSCs, and Pelt cells. Two of these cell lines, BMA13H and hASCs-T, were incompletely immortalized [74,85], and so in these cases the reduction of osteogenic potential could be a result of the progressive loss of differentiation potential that occurs in primary MSCs as well. Also, nonreplicative senescence effects cannot be discarded in the reduction of osteogenic potential observed in GB/hTERT MSCs (derived from umbilical cord) and Pelt cells (derived from the periodontal ligament) [86,90].
Of note, all the reviewed MSC cell lines immortalized with hTERT and E6/E7 were described to maintain or enhance their osteogenic potential. Interestingly, the hTERT and E6/E7-transduced iMSC line 3A6 showed a higher osteogenic potential than their E6/E7-only-transduced counterpart, the KP cells [55], suggesting that a complete immortalization could be beneficial for the bone-forming capacity of MSCs. Nonetheless, hTERT expression in MSCs has also been shown to upregulate osteogenesis-related genes such as RUNX2, osterix, and osteocalcin [91], and SV40LT-transduced iMSCs have shown higher levels of RUNX2 than primary MSCs without any osteogenic stimuli as well [51]. It is commonly accepted that osteogenesis is the default differentiation pathway for MSCs [15,111] and the most commonly retained differentiation lineage at later passages. Thus, we hypothesize that this high expression of bone-related transcription factors might be due to an osteogenic commitment of later passaged MSCs instead of being due to an effect of the transduction of the immortalization genes. These signs of “spontaneous differentiation” have also been observed in the KP cells, but have been lost by complete immortalization of these cells with hTERT in addition to E6/E7 genes [55].
A reduction of osteogenic potential was also observed in some MSC cell lines immortalized with hTERT and SV40LT, such as hASCs-TS [85]. Song et al. (2017) also pointed out that reversibly immortalized iSuPs (derived from coronal sutures) presented increased osteogenic potential when the immortalization gene SV40LT was removed [50]. Shu et al. (2018) have proposed that MSCs with higher proliferative activity, such as SV40LT-transduced MSCs, may need a longer time to differentiate towards the osteogenic lineage [49]. Tátrai et al. (2012) reported that adipose tissue-derived MSCs immortalized with a combination of SV40LT and hTERT showed a higher growth rate, as a well as a reduced osteogenic and adipogenic potency [110]. The literature shows that the osteogenic differentiation potential is the most commonly retained path after immortalization, and that MSCs are able to differentiate towards this lineage if they are completely immortalized and adequate times and strategies of osteogenic induction are used.

3.2.2. Chondrogenic Potential

Only 23 iMSC lines out of the 38 included in this review have been submitted for analysis of their chondrogenic potential. Two iMSC lines did not show any chondrogenic potential when assessed in two-dimensional culture: UE6E7T-2, derived from bone marrow and transduced with E6/E7 and hTERT [99]; and iSuPS, derived from coronal sutures and transduced with SV40LT [50]. Three other iMSC lines (SCP-1, BMA13H, and 3 Hits hMPC), all derived from bone marrow-MSCs, showed reduced chondrogenic potential in comparison to primary cells; all these iMSC lines were chondrogenically induced in three-dimensional culture and contained hTERT as immortalization gene, and two of them were incompletely immortalized [14,74,101].
In many cases, the chondrogenic potential of iMSCs has been scarcely analyzed by one single histochemical staining, either alcian blue staining (ABS) or toluidine blue staining (TBS) [14,54,55,71,91,101]. Although chondrogenic transcription factor Sox9 and type II collagen upregulation have been detected in chondrogenic-induced iMSCs, these cells showed the same proneness to hypertrophy as primary MSCs, with type X collagen expression [15,35] and low-quality cartilage production [22] with scarce exceptions [41,94], even when three-dimensional culture was performed [15,22,35]. This low-quality cartilage generation is relatively common among MSCs. It has been proposed that these cells are intrinsically committed to bone formation through the endochondral ossification pathway, and that they follow this differentiation program after being exposed to chondrogenic stimuli [111]. However, the lack of reproducibility among chondrogenic protocols is also a feasible explanation [112], and it is important to note that good results have been obtained when performing chondrogenesis onto suitable scaffolds [95]. Finger et al. (2003) found that immortalized cell lines obtained from chondrocytes were highly proliferative and showed less expression of genes involved in matrix synthesis and turnover than expected [113], in the same way that higher proliferation rates are related to reduced mineralization upon osteogenic induction [49,110].
Despite this, it has been shown that low chondrogenic iMSCs can stimulate chondrocyte differentiation when cocultured [71], possibly through the same trophic effects as primary MSCs. It is important to take into account that MSCs exists as heterogeneous populations, and that the iMSCs differentiation properties could be derived from primary cells or be related to the clonal selection, as has been proposed by Bourgine et al. (2014) [15]. Therefore, the differentiation potential of iMSCs should be assessed in comparison with their untransduced counterparts—the same clone or the primary cells derived from the same donor—in order to detect variations as a consequence of immortalization. Selection of MSC subsets or development of methods to stimulate MSCs to induce and/or to modulate specific attributes of the cells could give rise to more chondrogenic iMSC lines [9].

3.2.3. Adipogenic Potential

The adipogenic potential of 30 out of 38 iMSC lines was assessed, mainly by oil red O staining (OROS), but also by gene expression analysis of adipogenesis-related genes, such as transcription factor PPARγ, with different results, as seen in Table 1. Several hTERT-transduced iMSC lines derived from the bone marrow [19,101], placenta [91], and adipose tissue [85] showed a reduction in adipogenic potential in comparison with primary MSCs. In addition, the bone marrow-derived KM101 iMSC line was unable to differentiate towards the adipogenic lineage [45], similar to incompletely immortalized hASCs-T [85]. Moreover, adipogenic potential of the 3A6 iMSC line was reduced in comparison with the KP cells from which they were derived, unlike osteogenic potential [55]. Conversely, all the reviewed MSC cell lines immortalized with SV40LT maintained their adipogenic potential. The adipose-tissue-derived ASC/TERT1 cell line showed an increase in adipogenic potential after immortalization with hTERT [22]. Once more, iSUPS differentiation potential towards the adipogenic lineage was increased when SV40LT was removed [50].

3.3. Surface Markers Expression of iMSCs

In 2006, the International Society for Cell Therapy proposed a panel of cell surface markers to identify human MSC, including CD73, CD90, and CD105 [114]. However, none of these markers are specific for MSCs, and their expression does not imply a multidifferentiation ability [115], since the same expression pattern can be found in other cell types, such as fibroblasts [116]. Although the expression of these surface markers is usually investigated in primary MSCs before immortalization, none of the articles performed sorting selection; instead, the whole isolated population or uncharacterized clones were employed for transduction. Nevertheless, the level of expression of these makers may change due to passaging and culture conditions [81,115], and their expression in primary cells does not guarantee that they will be expressed in immortalized cells, even if they are previously sorted.
For example, Abarrategi et al. (2018) noticed a lowering of CD73 and CD105 expression in iMSCs in comparison with primary MSCs [102], and Alexander et al. (2015) observed that cranial periosteum-derived TAg cells were less CD105-positive, but more CD146-positive than primary cells [51]. Adipose-tissue-derived hASCs-TS and hASCs-TE showed the same decrease in CD105 expression and an increase in CD146 expression [85]. On the contrary, hTERT-transduced BMA13H maintained CD105, but displayed reduced CD90 expression [81]. It is not possible to know if these changes were caused by transduction of immortalization genes, subculturing, or both.
In addition, it is not clear whether the expression of a certain surface marker of this traditional panel confers an advantage to differentiate towards a specific lineage; for example, it has been shown that there are no differences in chondrogenic potential of MSCs caused by CD105 expression [117]. Therefore, the value of these surface markers for iMSCs characterization or selection may be limited. However, the expression of several surface markers outside this panel may have functional characteristics. For example, James et al. (2015) noted that nondifferentiating iMSC clones were uniquely CD137-positive [19], and Jayasuriya et al. (2018) pointed that SV40LT-transduced OA-MSCs expressed high levels of CD54, which is lowly expressed by bone marrow-derived MSCs but constitutively expressed by articular chondrocytes [41].

3.4. Clonality, Selection and Validation

It is known that polyclonal expansion favors selection of faster growing cells, while clone characterization and selection may enable the maintenance of a subpopulation of cells with more desirable characteristics [81]. Variations in differentiation potential exists among clones derived from one single donor [19], and even among subclones derived from single MSCs [84]. Therefore, careful selection of clones could favor certain applications.
Half of the iMSC lines that are reviewed here were clonal, while the other half were nonclonal. However, some clones were not selected by their differentiation abilities or surface markers expression profile, but were randomly picked [91] or chosen because of their proliferation capacity [71]. On the contrary, Bourgine et al. (2014) decided to select the clone with the most prominent osteogenic differentiation capacity, thus obtaining an iMSC line likely to be suitable for bone regeneration approaches, but still with weak chondrogenic potential. Further studies are needed to know if the use of a similar approach would give rise to an iMSC line with better chondrogenic potential [15].
Of note, only eight of the iMSC lines reviewed here—four of them derived from the same donor—have been submitted to short tandem repeat (STR) analysis to confirm whether they originated from one particular donor. This validation is important, especially if these iMSC lines are employed for basic research about MSCs biology; for instance, comparing cell lines originating from young and elderly donors or investigating the characteristics or behavior of these cells in skeletal diseases. Using this approach, Jayasuriya et al. (2018) generated and analyzed clonal iMSC lines from knee articular cartilage of osteoarthritic patients, identifying the existence of two MSC populations in human osteoarthritic cartilage; one preferentially undergoing chondrogenesis and the other exhibiting higher osteogenic potential. In this case, the generated cell lines were properly submitted to STR genotyping [41].

3.5. Tumorigenicity

MSCs have been described as resistant to malignant transformation, requiring the combination of several events to achieve an oncogenic phenotype. Primary MSCs have been widely used in clinical trials, but immortalized MSCs transduced with proto-oncogenes can eventually become tumorigenic, making them useless for clinical approaches, but not for research purposes. The tumorigenicity of 18 out of 38 iMSC lines included in this review was investigated by either soft agar colony formation assay or in vivo tumorigenicity test in immunodeficient mice (IDM). There were only two cases in which these cells showed signs of tumorigenicity; c-Fos-transduced 3 Hits hMPCs [102] and high passaged UE6E7T-3 [96], as seen in Table 1. In the case of 3 Hits hMPCs, it is not surprising that the transduction of the proto-oncogene c-Fos led to oncogenic transformation of iMSCs already transduced with hTERT and E6/E7 genes. Importantly, transformed iMSCs lost their phenotype and experienced changes in their differentiation potential, with c-Fos-transformed iMSCs showing reduced adipogenic and osteogenic potential and a conserved ability to specifically differentiate towards the chondrogenic lineage, as well as forming chondrogenic tumors in IDM. However, 3 Hits hMPCs did not display tumorigenic features despite accumulating oncogenic mutations in hTERT and E6/E7 genes [102], thus confirming that iMSCs need further signals to initiate carcinogenesis [106].
However, oncogenic mutations may arise during passaging of iMSCs [118]. In this regard, culture conditions are important, since hTERT-transduced iMSCs seeded at low densities during long periods of time have been reported to be tumorigenic [70]. Low density seeding provides an advantage for clones with oncogenic mutations that display higher growth rates; the lower the density seeding, the faster the accumulation of these oncogenic clones in the population. hTERT and E6/E7-transduced UE6E7T-3 at high passages (252 PDs) were capable of forming colonies in soft agar and sarcomas in IDM, while lower passaged cells (less than 200 PDs) did not shown any sign of malignant transformation [96]. This may indicate that although iMSCs have an unlimited lifespan, their ability to maintain their phenotype may be restricted to the first PDs, and that their characteristics should be submitted to periodical testing.
It is important to note that hTERT expression also has a role in the achievement of cellular capacities related to tumorigenesis, such as angiogenesis and immune system evasion. In the same way, the inactivation of p53 and Rb by SV40LT and E6/E7 proteins is related to the acquisition of cancer-related features. Moreover, if hTERT is re-expressed after SV40LT or E6/E7 transduction, its recovery could favor the fixation of aberrant karyotypes that lead to malignant phenotypes [106]. Unsurprisingly, the introduction of immortalization genes in MSCs alters the expression levels of genes associated with stem cell functions [91], which highlights the need for detailed characterization of iMSC lines.

3.6. In Vivo Bone Formation Capacity

One of the fundamental characteristics of MSCs is their ability to form ectopic “ossicles” which mimic the architecture of bone marrow [119]. The ectopic bone formation ability of hTERT-transduced MSCs has been investigated in IDM. Simonsen et al. (2002) found that hMSCs-hTERT generated bone-enclosing bone marrow cells and adipocytes when implanted subcutaneously with hydroxyapatite/tricalcum phosphate powder [63]. hMSCs-hTERT-derived clones formed ectopic bone marrow stroma-supporting hematopoiesis and adipocytes after in vivo transplantation as well [70]. However, after extensive subculturing, one of these clones was found to produce tumors, composed mostly of mesoderm-type cells [70]. Bourgine et al. (2014) also assessed the bone formation ability of MSOD together with ceramic granules in a fibrinogen/thrombin gel. They found that MSOD secreted a dense collagen matrix and formed osteoid tissue [15].

4. Conclusions

A number of iMSC lines have been generated in an attempt to overcome the limitations associated with primary MSCs. These cell lines have had many in vitro applications, including testing of engineered scaffolds for bone and cartilage repair, decellularized extracellular matrix production, investigation of the MSCs differentiation process at the molecular level, optimization of the current differentiation protocols, and analysis of their behavior in the pathological joints. However, the application of these cells has been for research purposes only as they present a risk of tumorigenicity.
Several approaches have been employed to confer an unlimited proliferation potential to MSCs, mainly involving viral genes and hTERT transduction, with different degrees of success. It is still unclear which set of genetic alterations are necessary and sufficient for MSC immortalization, but it probably involves abrogation of replicative and nonreplicative senescence.
Alterations of the multidifferentiation potential of MSCs after immortalization have been described, with the osteogenic potential being the best conserved in fully immortalized MSC lines. However, there are also iMSC lines capable of differentiating towards the chondrogenic lineage when cultured in 3D environment. In addition, some studies suggest that other characteristics of iMSC lines, such as secretion of trophic factors, are maintained in MSCs despite immortalization.
The ideal iMSC line should retain the phenotypic and functional characteristics of MSCs, as well as a normal karyotype. The usefulness of these cell lines in bone and cartilage regenerative medicine research would be increased if clones were carefully selected and validated. Before employing iMSCs for basic or applied bone and cartilage research, their characteristics should be fully understood. Moreover, the maintenance of these characteristics should be assessed periodically throughout passaging, as immortalization does not guarantee it, and both polyclonal expansion and low-density seeding should be avoided to prevent malignant transformation. If these requirements are fulfilled, iMSC lines could be useful and convenient tools for basic research, testing of tissue engineering approaches, and production of biotechnological products, among other applications.

Funding

This study was carried out thanks to the funding from Rede Galega de Terapia Celular and Grupos con Potencial de Crecemento, Xunta de Galicia (R2016/036, R2014/050, CN2012/142 and GPC2014/048); Deputación da Coruña (BINV-CS/2016); Fundación Española de Reumatología (Proyectos 2014); Universidade da Coruña (UDC) and Centro de Investigación Biomédica en Red-Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN).

Acknowledgments

María Piñeiro-Ramil, Rocío Castro-Viñuelas and Silvia Rodríguez-Fernández were granted by a predoctoral fellowship from Xunta de Galicia and European Union (European Social Fund) and Clara Sanjurjo-Rodríguez is beneficiary of a postdoctoral fellowship from Xunta de Galicia.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

MSCMesenchymal stromal cells
PDsPopulation doublings
iMSCImmortal mesenchymal stromal cells
hTERTHuman telomerase reverse transcriptase
SV40LTSimian virus 40 large T antigen
HPVHuman papillomavirus
ARSAlizarin Red Staining
VKSVon Kossa Staining
APSAlkaline Phosphatase Staining
ABSAlcian Blue Staining
TBSToluidine Blue Staining
OROS
PSR
Oil Red O Staining
Picro-Sirius Red
STRShort Tandem Repeat
IDMImmunodeficient Mice

References

  1. Grayson, W.L.; Bunnell, B.A.; Martin, E.; Frazier, T.; Hung, B.P.; Gimble, J.M. Stromal cells and stem cells in clinical bone regeneration. Nat. Rev. Endocrinol. 2015, 11, 140–150. [Google Scholar] [CrossRef] [PubMed]
  2. Tamaddon, M.; Wang, L.; Liu, Z.; Liu, C. Osteochondral tissue repair in osteoarthritic joints: Clinical challenges and opportunities in tissue engineering. Bio Des. Manuf. 2018, 1, 101–114. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Roseti, L.; Parisi, V.; Petretta, M.; Cavallo, C.; Desando, G.; Bartolotti, I.; Grigolo, B. Scaffolds for Bone Tissue Engineering: State of the art and new perspectives. Mater. Sci. Eng. C 2017, 78, 1246–1262. [Google Scholar] [CrossRef] [PubMed]
  4. Piñeiro-Ramil, M.; Castro-Viñuelas, R.; Sanjurjo-Rodríguez, C.; Hermida-Gómez, T.; Fuentes-Boquete, I.; Toro-Santos, F.J.; de Blanco-García, F.J.; Díaz-Prado, S.M. Cell Therapy and Tissue Engineering for Cartilage Repair. In Cartilage Repair and Regeneration; InTech: London, UK, 2018. [Google Scholar]
  5. Deng, C.; Zhu, H.; Li, J.; Feng, C.; Yao, Q.; Wang, L.; Chang, J.; Wu, C. Bioactive Scaffolds for Regeneration of Cartilage and Subchondral Bone Interface. Theranostics 2018, 8, 1940–1955. [Google Scholar] [CrossRef] [PubMed]
  6. Ghassemi, T.; Shahroodi, A.; Ebrahimzadeh, M.H.; Mousavian, A.; Movaffagh, J.; Moradi, A. Current Concepts in Scaffolding for Bone Tissue Engineering. Arch. Bone Jt. Surg. 2018, 6, 90–99. [Google Scholar] [PubMed]
  7. Samsonraj, R.M.; Raghunath, M.; Nurcombe, V.; Hui, J.H.; van Wijnen, A.J.; Cool, S.M. Concise Review: Multifaceted Characterization of Human Mesenchymal Stem Cells for Use in Regenerative Medicine. Stem Cells Transl. Med. 2017, 6, 2173–2185. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Szychlinska, M.A.; Stoddart, M.J.; D’Amora, U.; Ambrosio, L.; Alini, M.; Musumeci, G. Mesenchymal Stem Cell-Based Cartilage Regeneration Approach and Cell Senescence: Can We Manipulate Cell Aging and Function? Tissue Eng. Part B Rev. 2017, 23, 529–539. [Google Scholar] [CrossRef]
  9. Kouroupis, D.; Sanjurjo-Rodriguez, C.; Jones, E.; Correa, D. MSC functionalization for enhanced therapeutic applications. Tissue Eng. Part B Rev. 2018. [Google Scholar] [CrossRef]
  10. Richardson, S.M.; Kalamegam, G.; Pushparaj, P.N.; Matta, C.; Memic, A.; Khademhosseini, A.; Mobasheri, R.; Poletti, F.L.; Hoyland, J.A.; Mobasheri, A. Mesenchymal stem cells in regenerative medicine: Focus on articular cartilage and intervertebral disc regeneration. Methods 2016, 99, 69–80. [Google Scholar] [CrossRef]
  11. Yong, K.W.; Choi, J.R.; Dolbashid, A.S.; Wan Safwani, W.K.Z. Biosafety and bioefficacy assessment of human mesenchymal stem cells: What do we know so far? Regen Med. 2018, 13, 219–232. [Google Scholar] [CrossRef]
  12. Herberts, C.A.; Kwa, M.S.; Hermsen, H.P. Risk factors in the development of stem cell therapy. J. Transl. Med. 2011, 9, 29. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Koch, C.M.; Reck, K.; Shao, K.; Lin, Q.; Joussen, S.; Ziegler, P.; Walenda, G.; Drescher, W.; Opalka, B.; May, T.; et al. Pluripotent stem cells escape from senescence-associated DNA methylation changes. Genome Res. 2013, 23, 248–259. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Böcker, W.; Yin, Z.; Drosse, I.; Haasters, F.; Rossmann, O.; Wierer, M.; Popov, C.; Locher, M.; Mutschler, W.; Docheva, D.; et al. Introducing a single-cell-derived human mesenchymal stem cell line expressing hTERT after lentiviral gene transfer. J. Cell. Mol. Med. 2008, 12, 1347–1359. [Google Scholar] [CrossRef] [Green Version]
  15. Bourgine, P.; Le Magnen, C.; Pigeot, S.; Geurts, J.; Scherberich, A.; Martin, I. Combination of immortalization and inducible death strategies to generate a human mesenchymal stromal cell line with controlled survival. Stem Cell Res. 2014, 12, 584–598. [Google Scholar] [CrossRef] [Green Version]
  16. Baker, N.; Boyette, L.B.; Tuan, R.S. Characterization of bone marrow-derived mesenchymal stem cells in aging. Bone 2015, 70, 37–47. [Google Scholar] [CrossRef]
  17. Stölzel, K.; Schulze-Tanzil, G.; Olze, H.; Schwarz, S.; Feldmann, E.M.; Rotter, N. Immortalised human mesenchymal stem cells undergo chondrogenic differentiation in alginate and PGA/PLLA scaffolds. Cell Tissue Bank. 2015, 16, 159–170. [Google Scholar] [CrossRef]
  18. Okamoto, T.; Aoyama, T.; Nakayama, T.; Nakamata, T.; Hosaka, T.; Nishijo, K.; Nakamura, T.; Kiyono, T.; Toguchida, J. Clonal heterogeneity in differentiation potential of immortalized human mesenchymal stem cells. Biochem. Biophys. Res. Commun. 2002, 295, 354–361. [Google Scholar] [CrossRef] [Green Version]
  19. James, S.; Fox, J.; Afsari, F.; Lee, J.; Clough, S.; Knight, C.; Ashmore, J.; Ashton, P.; Preham, O.; Hoogduijn, M.; et al. Multiparameter Analysis of Human Bone Marrow Stromal Cells Identifies Distinct Immunomodulatory and Differentiation-Competent Subtypes. Stem Cell Rep. 2015, 4, 1004–1015. [Google Scholar] [CrossRef] [Green Version]
  20. Ahlfeld, T.; Cidonio, G.; Kilian, D.; Duin, S.; Akkineni, A.R.; Dawson, J.I.; Yang, S.; Lode, A.; Oreffo, R.O.C.; Gelinsky, M. Development of a clay based bioink for 3D cell printing for skeletal application. Biofabrication 2017, 9. [Google Scholar] [CrossRef]
  21. Akmammedov, R.; Huysal, M.; Isik, S.; Senel, M. Preparation and characterization of novel chitosan/zeolite scaffolds for bone tissue engineering applications. Int. J. Polym. Mater. Polym. Biomater. 2018, 67, 110–118. [Google Scholar] [CrossRef]
  22. Nürnberger, S.; Schneider, C.; van Osch, G.V.M.; Keibl, C.; Rieder, B.; Monforte, X.; Teuschl, A.H.; Mühleder, S.; Holnthoner, W.; Schädl, B.; et al. Repopulation of an auricular cartilage scaffold, AuriScaff, perforated with an enzyme combination. Acta Biomater. 2019, 86, 207–222. [Google Scholar] [CrossRef] [PubMed]
  23. Hossain, K.M.Z.; Patel, U.; Kennedy, A.R.; Macri-Pellizzeri, L.; Sottile, V.; Grant, D.M.; Scammell, B.E.; Ahmed, I. Porous calcium phosphate glass microspheres for orthobiologic applications. Acta Biomater. 2018, 72, 396–406. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Prasopthum, A.; Shakesheff, K.M.; Yang, J. Direct three-dimensional printing of polymeric scaffolds with nanofibrous topography. Biofabrication 2018, 10. [Google Scholar] [CrossRef] [PubMed]
  25. Tendulkar, G.; Sreekumar, V.; Rupp, F.; Teotia, A.K.; Athanasopulu, K.; Kemkemer, R.; Buck, A.; Buck, A.; Kaps, H.-P.; Geis-Gerstorfer, J.; et al. Characterisation of porous knitted titanium for replacement of intervertebral disc nucleus pulposus. Sci. Rep. 2017, 7. [Google Scholar] [CrossRef] [Green Version]
  26. Bourgine, P.E.; Gaudiello, E.; Pippenger, B.; Jaquiery, C.; Klein, T.; Pigeot, S.; Todorov, A.; Feliciano, S.; Banfi, A.; Martin, I. Engineered Extracellular Matrices as Biomaterials of Tunable Composition and Function. Adv. Funct. Mater. 2017, 27. [Google Scholar] [CrossRef]
  27. da Conceicao Ribeiro, R.; Pal, D.; Ferreira, A.M.; Gentile, P.; Benning, M.; Dalgarno, K. Reactive jet impingement bioprinting of high cell density gels for bone microtissue fabrication. Biofabrication 2018, 11. [Google Scholar] [CrossRef]
  28. Aida, Y.; Kurihara, H.; Kato, K. Wnt3a promotes differentiation of human bone marrow-derived mesenchymal stem cells into cementoblast-like cells. Vitr. Cell. Dev. Biol. Anim. 2018, 54, 468–476. [Google Scholar] [CrossRef]
  29. Cesarz, Z.; Funnell, J.L.; Guan, J.; Tamama, K. Soft Elasticity-Associated Signaling and Bone Morphogenic Protein 2 Are Key Regulators of Mesenchymal Stem Cell Spheroidal Aggregates. Stem Cells Dev. 2016, 25, 622–635. [Google Scholar] [CrossRef]
  30. Elsafadi, M.; Manikandan, M.; Alajez, N.M.; Hamam, R.; Dawud, R.A.; Aldahmash, A.; Iqbal, Z.; Alfayez, M.; Kassem, M.; Mahmood, A. MicroRNA-4739 regulates osteogenic and adipocytic differentiation of immortalized human bone marrow stromal cells via targeting LRP3. Stem Cell Res. 2017, 20, 94–104. [Google Scholar] [CrossRef] [Green Version]
  31. Torii, D.; Tsutsui, T.W.; Watanabe, N.; Konishi, K. Bone morphogenetic protein 7 induces cementogenic differentiation of human periodontal ligament-derived mesenchymal stem cells. Odontology 2016, 104, 1–9. [Google Scholar] [CrossRef]
  32. Wilson, K.M.; Jagger, A.M.; Walker, M.; Seinkmane, E.; Fox, J.M.; Kröger, R.; Genever, P.; Ungar, D. Glycans modify mesenchymal stem cell differentiation to impact on the function of resulting osteoblasts. J. Cell Sci. 2018, 131. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Ali, D.; Hamam, R.; Alfayez, M.; Kassem, M.; Aldahmash, A.; Alajez, N.M. Epigenetic Library Screen Identifies Abexinostat as Novel Regulator of Adipocytic and Osteoblastic Differentiation of Human Skeletal (Mesenchymal) Stem Cells. Stem Cells Transl. Med. 2016, 5, 1036–1047. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Hossan, T.; Nagarajan, S.; Baumgart, S.J.; Xie, W.; Magallanes, R.T.; Hernandez, C.; Chiaroni, P.-M.; Indenbirken, D.; Spitzner, M.; Thomas-Chollier, M.; et al. Histone Chaperone SSRP1 is Essential for Wnt Signaling Pathway Activity During Osteoblast Differentiation. Stem Cells 2016, 34, 1369–1376. [Google Scholar] [CrossRef] [Green Version]
  35. Armbruster, N.; Krieg, J.; Weißenberger, M.; Scheller, C.; Steinert, A.F. Rescued Chondrogenesis of Mesenchymal Stem Cells under Interleukin 1 Challenge by Foamyviral Interleukin 1 Receptor Antagonist Gene Transfer. Front. Pharmacol. 2017, 8, 255. [Google Scholar] [CrossRef]
  36. Duan, P.; Toumpaniari, R.; Partridge, S.; Birch, M.A.; Genever, P.G.; Bull, S.J.; Dalgarno, K.W.; McCaskie, A.W.; Chen, J. How cell culture conditions affect the microstructure and nanomechanical properties of extracellular matrix formed by immortalized human mesenchymal stem cells: An experimental and modelling study. Mater. Sci. Eng. C 2018, 89, 149–159. [Google Scholar] [CrossRef] [Green Version]
  37. Hu, X.; Xie, P.; Li, W.; Li, Z.; Shan, H. Direct induction of hepatocyte-like cells from immortalized human bone marrow mesenchymal stem cells by overexpression of HNF4α. Biochem. Biophys. Res. Commun. 2016, 478, 791–797. [Google Scholar] [CrossRef]
  38. Tendulkar, G.; Ehnert, S.; Sreekumar, V.; Chen, T.; Kaps, H.-P.; Golombek, S.; Wendel, H.-P.; Nüssler, A.; Avci-Adali, M. Exogenous Delivery of Link N mRNA into Chondrocytes and MSCs—The Potential Role in Increasing Anabolic Response. Int. J. Mol. Sci. 2019, 20, 1716. [Google Scholar] [CrossRef] [Green Version]
  39. Lin, W.-H.; Yu, J.; Chen, G.; Tsai, W.-B. Fabrication of multi-biofunctional gelatin-based electrospun fibrous scaffolds for enhancement of osteogenesis of mesenchymal stem cells. Colloids Surf. B Biointerfaces 2016, 138, 26–31. [Google Scholar] [CrossRef]
  40. Harkness, L.; Zaher, W.; Ditzel, N.; Isa, A.; Kassem, M. CD146/MCAM defines functionality of human bone marrow stromal stem cell populations. Stem Cell Res. Ther. 2016, 7, 4. [Google Scholar] [CrossRef] [Green Version]
  41. Jayasuriya, C.T.; Hu, N.; Li, J.; Lemme, N.; Terek, R.; Ehrlich, M.G.; Chen, Q. Molecular characterization of mesenchymal stem cells in human osteoarthritis cartilage reveals contribution to the OA phenotype. Sci. Rep. 2018, 8, 7044. [Google Scholar] [CrossRef]
  42. Hu, N.; Gao, Y.; Jayasuriya, C.T.; Liu, W.; Du, H.; Ding, J.; Feng, M.; Chen, Q. Chondrogenic induction of human osteoarthritic cartilage-derived mesenchymal stem cells activates mineralization and hypertrophic and osteogenic gene expression through a mechanomiR. Arthr. Res. Ther. 2019, 21, 167. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Moher, D.; Liberati, A.; Tetzlaff, J.; Altman, D.G. Preferred Reporting Items for Systematic Reviews and Meta-Analyses: The PRISMA Statement. PLoS Med. 2009, 6, e1000097. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Lee, K.S.; Shim, J.S.; Paik, M.J.; Joo, W.H.; Kim, S.H.; Lee, G.; Kim, D.W. Characterization of a growth-elevated cell line of human bone marrow-derived mesenchymal stem cells by SV40 T-antigen. Biotechnol. Bioprocess. Eng. 2015, 20, 498–505. [Google Scholar] [CrossRef]
  45. Harigaya, K.; Handa, H. Generation of functional clonal cell lines from human bone marrow stroma. Proc. Natl. Acad. Sci. USA 1985, 82, 3477–3480. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Liang, X.; Glowacki, J.; Hahne, J.; Xie, L.; LeBoff, M.S.; Zhou, S. Dehydroepiandrosterone Stimulation of Osteoblastogenesis in Human MSCs Requires IGF-I Signaling. J. Cell. Biochem. 2016, 117, 1769–1774. [Google Scholar] [CrossRef]
  47. Thalmeier, K.; Meissner, P.; Reisbach, G.; Falk, M.; Brechtel, A.; Dörmer, P. Establishment of two permanent human bone marrow stromal cell lines with long-term post irradiation feeder capacity. Blood 1994, 83, 1799–1807. [Google Scholar] [CrossRef] [Green Version]
  48. Conrad, C.; Gottgens, B.; Kinston, S.; Ellwart, J.; Huss, R. GATA transcription in a small rhodamine 123(low)CD34(+) subpopulation of a peripheral blood-derived CD34(-)CD105(+) mesenchymal cell line. Exp. Hematol. 2002, 30, 887–895. [Google Scholar] [CrossRef]
  49. Shu, Y.; Yang, C.; Ji, X.; Zhang, L.; Bi, Y.; Yang, K.; Gong, M.; Liu, X.; Guo, Q.; Su, Y.; et al. Reversibly immortalized human umbilical cord-derived mesenchymal stem cells (UC-MSCs) are responsive to BMP9-induced osteogenic and adipogenic differentiation. J. Cell. Biochem. 2018, 119, 8872–8886. [Google Scholar] [CrossRef]
  50. Song, D.; Zhang, F.; Reid, R.R.; Ye, J.; Wei, Q.; Liao, J.; Zou, Y.; Fan, J.; Ma, C.; Hu, X.; et al. BMP9 induces osteogenesis and adipogenesis in the immortalized human cranial suture progenitors from the patent sutures of craniosynostosis patients. J. Cell. Mol. Med. 2017, 21, 2782–2795. [Google Scholar] [CrossRef] [Green Version]
  51. Alexander, D.; Biller, R.; Rieger, M.; Ardjomandi, N.; Reinert, S. Phenotypic Characterization of a Human Immortalized Cranial Periosteal Cell Line. Cell. Physiol. Biochem. 2015, 35, 2244–2254. [Google Scholar] [CrossRef]
  52. Wu, Y.; Feng, G.; Song, J.; Zhang, Y.; Yu, Y.; Huang, L.; Zheng, L.; Deng, F. TrAmplification of Human Dental Follicle Cells by piggyBac Transposon—Mediated Reversible Immortalization System. PLoS ONE 2015, 10, e0130937. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Dou, L.; Wu, Y.; Yan, Q.; Wang, J.; Zhang, Y.; Ji, P. Secretome profiles of immortalized dental follicle cells using iTRAQ-based proteomic analysis. Sci. Rep. 2017, 7, 7300. [Google Scholar] [CrossRef] [PubMed]
  54. Hung, S.-C.; Yang, D.-M.; Chang, C.-F.; Lin, R.-J.; Wang, J.-S.; Low-Tone Ho, L.; Yang, W.K. Immortalization without neoplastic transformation of human mesenchymal stem cells by transduction with HPV16E6/E7 genes. Int. J. Cancer 2004, 110, 313–319. [Google Scholar] [CrossRef] [PubMed]
  55. Tsai, C.-C.; Chen, C.-L.; Liu, H.-C.; Lee, Y.-T.; Wang, H.-W.; Hou, L.-T.; Hung, S.-C. Overexpression of hTERT increases stem-like properties and decreases spontaneous differentiation in human mesenchymal stem cell lines. J. Biomed. Sci. 2010, 17, 64. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Fernández-Moreno, M.; Hermida-Gómez, T.; Gallardo, M.E.; Dalmao-Fernández, A.; Rego-Pérez, I.; Garesse, R.; Blanco, F.J. Generating Rho-0 Cells Using Mesenchymal Stem Cell Lines. PLoS ONE 2016, 11, e0164199. [Google Scholar] [CrossRef]
  57. Chen, Y.-C.; Wu, K.-C.; Huang, B.-M.; So, E.C.; Wang, Y.-K. Midazolam inhibits chondrogenesis via peripheral benzodiazepine receptor in human mesenchymal stem cells. J. Cell. Mol. Med. 2018, 22, 2896–2907. [Google Scholar] [CrossRef]
  58. Mori, T.; Kiyono, T.; Imabayashi, H.; Takeda, Y.; Tsuchiya, K.; Miyoshi, S.; Makino, H.; Matsumoto, K.; Saito, H.; Ogawa, S.; et al. Combination of hTERT and bmi-1, E6, or E7 Induces Prolongation of the Life Span of Bone Marrow Stromal Cells from an Elderly Donor without Affecting Their Neurogenic Potential. Mol. Cell. Biol. 2005, 25, 5183–5195. [Google Scholar] [CrossRef] [Green Version]
  59. Kusuyama, J.; Kamisono, A.; ChangHwan, S.; Amir, M.S.; Bandow, K.; Eiraku, N.; Ohnishi, T.; Matsuguchi, T. Spleen tyrosine kinase influences the early stages of multilineage differentiation of bone marrow stromal cell lines by regulating phospholipase C gamma activities. J. Cell. Physiol. 2018, 233, 2549–2559. [Google Scholar] [CrossRef]
  60. Roecklein, B.A.; Torok-Storb, B. Functionally distinct human marrow stromal cell lines immortalized by transduction with the human papilloma virus E6/E7 genes. Blood 1995, 85, 997–1005. [Google Scholar] [CrossRef] [Green Version]
  61. Liu, B.; Wu, S.; Han, L.; Zhang, C. β-catenin signaling induces the osteoblastogenic differentiation of human pre-osteoblastic and bone marrow stromal cells mainly through the upregulation of osterix expression. Int. J. Mol. Med. 2015, 36, 1572–1582. [Google Scholar] [CrossRef] [Green Version]
  62. Wei, K.; Xie, Y.; Chen, T.; Fu, B.; Cui, S.; Wang, Y.; Cai, G.; Chen, X. ERK1/2 signaling mediated naringin-induced osteogenic differentiation of immortalized human periodontal ligament stem cells. Biochem. Biophys. Res. Commun. 2017, 489, 319–325. [Google Scholar] [CrossRef] [PubMed]
  63. Simonsen, J.L.; Rosada, C.; Serakinci, N.; Justesen, J.; Stenderup, K.; Rattan, S.I.S.; Jensen, T.G.; Kassem, M. Telomerase expression extends the proliferative life-span and maintains the osteogenic potential of human bone marrow stromal cells. Nat. Biotechnol. 2002, 20, 592–596. [Google Scholar] [CrossRef] [PubMed]
  64. Leber, J.; Barekzai, J.; Blumenstock, M.; Pospisil, B.; Salzig, D.; Czermak, P. Microcarrier choice and bead-to-bead transfer for human mesenchymal stem cells in serum-containing and chemically defined media. Process. Biochem. 2017, 59, 255–265. [Google Scholar] [CrossRef]
  65. Ziouti, F.; Ebert, R.; Rummler, M.; Krug, M.; Müller-Deubert, S.; Lüdemann, M.; Jakob, F.; Willie, B.M.; Jundt, F. NOTCH Signaling Is Activated through Mechanical Strain in Human Bone Marrow-Derived Mesenchymal Stromal Cells. Stem Cells Int. 2019, 2019, 1–13. [Google Scholar] [CrossRef]
  66. Müller-Deubert, S.; Seefried, L.; Krug, M.; Jakob, F.; Ebert, R. Epidermal growth factor as a mechanosensitizer in human bone marrow stromal cells. Stem Cell Res. 2017, 24, 69–76. [Google Scholar] [CrossRef]
  67. Salzig, D.; Leber, J.; Merkewitz, K.; Lange, M.C.; Köster, N.; Czermak, P. Attachment, Growth, and Detachment of Human Mesenchymal Stem Cells in a Chemically Defined Medium. Stem Cells Int. 2016, 2016. [Google Scholar] [CrossRef] [Green Version]
  68. Larsen, K.H.; Frederiksen, C.M.; Burns, J.S.; Abdallah, B.M.; Kassem, M. Identifying A Molecular Phenotype for Bone Marrow Stromal Cells With In Vivo Bone Forming Capacity. J. Bone Miner. Res. 2009, 25. [Google Scholar] [CrossRef]
  69. Elsafadi, M.; Manikandan, M.; Atteya, M.; Abu Dawud, R.; Almalki, S.; Ali Kaimkhani, Z.; Aldahmash, A.; Alajez, N.M.; Alfayez, M.; Kassem, M.; et al. SERPINB2 is a novel TGFβ-responsive lineage fate determinant of human bone marrow stromal cells. Sci. Rep. 2017, 7. [Google Scholar] [CrossRef] [Green Version]
  70. Abdallah, B.M.; Haack-Sørensen, M.; Burns, J.S.; Elsnab, B.; Jakob, F.; Hokland, P.; Kassem, M. Maintenance of differentiation potential of human bone marrow mesenchymal stem cells immortalized by human telomerase reverse transcriptase gene despite of extensive proliferation. Biochem. Biophys. Res. Commun. 2005, 326, 527–538. [Google Scholar] [CrossRef]
  71. Skårn, M.; Noordhuis, P.; Wang, M.-Y.; Veuger, M.; Kresse, S.H.; Egeland, E.V.; Micci, F.; Namløs, H.M.; Håkelien, A.-M.; Olafsrud, S.M.; et al. Generation and Characterization of an Immortalized Human Mesenchymal Stromal Cell Line. Stem Cells Dev. 2014, 23, 2377–2389. [Google Scholar] [CrossRef] [Green Version]
  72. Wu, L.; Leijten, J.C.H.; Georgi, N.; Post, J.N.; van Blitterswijk, C.A.; Karperien, M. Trophic effects of mesenchymal stem cells increase chondrocyte proliferation and matrix formation. Tissue Eng. Part A 2011, 17, 1425–1436. [Google Scholar] [CrossRef] [PubMed]
  73. Fayyad, A.; Khan, A.; Abdallah, S.; Alomran, S.; Bajou, K.; Khattak, M. Rosiglitazone Enhances Browning Adipocytes in Association with MAPK and PI3-K Pathways During the Differentiation of Telomerase-Transformed Mesenchymal Stromal Cells into Adipocytes. Int. J. Mol. Sci. 2019, 20, 1618. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Dale, T.P.; de Castro, A.; Kuiper, N.J.; Parkinson, E.K.; Forsyth, N.R. Immortalisation with hTERT Impacts on Sulphated Glycosaminoglycan Secretion and Immunophenotype in a Variable and Cell Specific Manner. PLoS ONE 2015, 10, e0133745. [Google Scholar] [CrossRef] [PubMed]
  75. Dale, T.P.; Forsyth, N.R. Ectopic Telomerase Expression Fails to Maintain Chondrogenic Capacity in Three-Dimensional Cultures of Clinically Relevant Cell Types. Biores. Open Access 2018, 7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Blaschke, M.; Koepp, R.; Lenz, C.; Kruppa, J.; Jung, K.; Siggelkow, H. Crohn’s disease patient serum changes protein expression in a human mesenchymal stem cell model in a linear relationship to patients’ disease stage and to bone mineral density. J. Clin. Transl. Endocrinol. 2018, 13, 26–38. [Google Scholar] [CrossRef] [PubMed]
  77. Schmid, J.; Schwarz, S.; Meier-Staude, R.; Sudhop, S.; Clausen-Schaumann, H.; Schieker, M.; Huber, R. A Perfusion Bioreactor System for Cell Seeding and Oxygen-Controlled Cultivation of Three-Dimensional Cell Cultures. Tissue Eng. Part C. Methods 2018, 24, 585–595. [Google Scholar] [CrossRef]
  78. Sreekumar, V.; Aspera-Werz, R.; Ehnert, S.; Strobel, J.; Tendulkar, G.; Heid, D.; Schreiner, A.; Arnscheidt, C.; Nussler, A.K. Resveratrol protects primary cilia integrity of human mesenchymal stem cells from cigarette smoke to improve osteogenic differentiation in vitro. Arch. Toxicol. 2018, 92, 1525–1538. [Google Scholar] [CrossRef]
  79. Westphal, I.; Jedelhauser, C.; Liebsch, G.; Wilhelmi, A.; Aszodi, A.; Schieker, M. Oxygen mapping: Probing a novel seeding strategy for bone tissue engineering. Biotechnol. Bioeng. 2017, 114, 894–902. [Google Scholar] [CrossRef]
  80. Aspera-Werz, R.H.; Chen, T.; Ehnert, S.; Zhu, S.; Fröhlich, T.; Nussler, A.K. Cigarette Smoke Induces the Risk of Metabolic Bone Diseases: Transforming Growth Factor Beta Signaling Impairment via Dysfunctional Primary Cilia Affects Migration, Proliferation, and Differentiation of Human Mesenchymal Stem Cells. Int. J. Mol. Sci. 2019, 20, 2915. [Google Scholar] [CrossRef] [Green Version]
  81. Duan, P.; Chen, J. Nanomechanical and microstructure analysis of extracellular matrix layer of immortalized cell line Y201 from human mesenchymal stem cells. Surf. Coat. Technol. 2015, 284, 417–421. [Google Scholar] [CrossRef]
  82. Galarza Torre, A.; Shaw, J.E.; Wood, A.; Gilbert, H.T.J.; Dobre, O.; Genever, P.; Brennan, K.; Richardson, S.M.; Swift, J. An immortalised mesenchymal stem cell line maintains mechano-responsive behaviour and can be used as a reporter of substrate stiffness. Sci. Rep. 2018, 8, 8981. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Vanderburgh, J.; Sterling, J.A.; Guelcher, S.A. 3D Printing of Tissue Engineered Constructs for In Vitro Modeling of Disease Progression and Drug Screening. Ann. Biomed. Eng. 2017, 45, 164–179. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Wolbank, S.; Stadler, G.; Peterbauer, A.; Gillich, A.; Karbiener, M.; Streubel, B.; Wieser, M.; Katinger, H.; van Griensven, M.; Redl, H.; et al. Telomerase Immortalized Human Amnion- and Adipose-Derived Mesenchymal Stem Cells: Maintenance of Differentiation and Immunomodulatory Characteristics. Tissue Eng. Part A 2009, 15, 1843–1854. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Balducci, L.; Blasi, A.; Saldarelli, M.; Soleti, A.; Pessina, A.; Bonomi, A.; Coccè, V.; Dossena, M.; Tosetti, V.; Ceserani, V.; et al. Immortalization of human adipose-derived stromal cells: Production of cell lines with high growth rate, mesenchymal marker expression and capability to secrete high levels of angiogenic factors. Stem Cell Res. Ther. 2014, 5, 63. [Google Scholar] [CrossRef] [Green Version]
  86. Siska, E.K.; Weisman, I.; Romano, J.; Ivics, Z.; Izsvák, Z.; Barkai, U.; Petrakis, S.; Koliakos, G. Generation of an immortalized mesenchymal stem cell line producing a secreted biosensor protein for glucose monitoring. PLoS ONE 2017, 12, e0185498. [Google Scholar] [CrossRef] [Green Version]
  87. Akazawa, Y.; Hasegawa, T.; Yoshimura, Y.; Chosa, N.; Asakawa, T.; Ueda, K.; Sugimoto, A.; Kitamura, T.; Nakagawa, H.; Ishisaki, A.; et al. Recruitment of mesenchymal stem cells by stromal cell-derived factor 1α in pulp cells from deciduous teeth. Int. J. Mol. Med. 2015, 36, 442–448. [Google Scholar] [CrossRef]
  88. Sugimoto, A.; Miyazaki, A.; Kawarabayashi, K.; Shono, M.; Akazawa, Y.; Hasegawa, T.; Ueda-Yamaguchi, K.; Kitamura, T.; Yoshizaki, K.; Fukumoto, S.; et al. Piezo type mechanosensitive ion channel component 1 functions as a regulator of the cell fate determination of mesenchymal stem cells. Sci. Rep. 2017, 7. [Google Scholar] [CrossRef] [Green Version]
  89. Tsutsui, T.; Kumakura, S.-I.; Yamamoto, A.; Kanai, H.; Tamura, Y.; Kato, T.; Anpo, M.; Tahara, H.; Barrett, J.C. Association of p16INK4a and pRb inactivation with immortalization of human cells. Carcinogenesis 2002, 23, 2111–2117. [Google Scholar] [CrossRef] [Green Version]
  90. Torii, D.; Konishi, K.; Watanabe, N.; Goto, S.; Tsutsui, T. Cementogenic potential of multipotential mesenchymal stem cells purified from the human periodontal ligament. Odontology 2015, 103, 27–35. [Google Scholar] [CrossRef]
  91. Qin, S.Q.; Kusuma, G.D.; Al-Sowayan, B.; Pace, R.A.; Isenmann, S.; Pertile, M.D.; Gronthos, S.; Abumaree, M.H.; Brennecke, S.P.; Kalionis, B. Establishment and characterization of fetal and maternal mesenchymal stem/stromal cell lines from the human term placenta. Placenta 2016, 39, 134–146. [Google Scholar] [CrossRef]
  92. Kusuma, G.D.; Brennecke, S.P.; O’Connor, A.J.; Kalionis, B.; Heath, D.E. Decellularized extracellular matrices produced from immortal cell lines derived from different parts of the placenta support primary mesenchymal stem cell expansion. PLoS ONE 2017, 12, e0171488. [Google Scholar] [CrossRef] [PubMed]
  93. Kusuma, G.D.; Yang, M.C.; Brennecke, S.P.; O’Connor, A.J.; Kalionis, B.; Heath, D.E. Transferable Matrixes Produced from Decellularized Extracellular Matrix Promote Proliferation and Osteogenic Differentiation of Mesenchymal Stem Cells and Facilitate Scale-Up. ACS Biomater. Sci. Eng. 2018, 8, 744–752. [Google Scholar] [CrossRef]
  94. Matta, C.; Fodor, J.; Miosge, N.; Takács, R.; Juhász, T.; Rybaltovszki, H.; Tóth, A.; Csernoch, L.; Zákány, R. Purinergic signalling is required for calcium oscillations in migratory chondrogenic progenitor cells. Pflüg. Arch. Eur. J. Physiol. 2015, 467, 429–442. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Koelling, S.; Kruegel, J.; Irmer, M.; Path, J.R.; Sadowski, B.; Miro, X.; Miosge, N. Migratory Chondrogenic Progenitor Cells from Repair Tissue during the Later Stages of Human Osteoarthritis. Cell Stem Cell 2009, 4, 324–335. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Takeuchi, M.; Higashino, A.; Takeuchi, K.; Hori, Y.; Koshiba-Takeuchi, K.; Makino, H.; Monobe, Y.; Kishida, M.; Adachi, J.; Takeuchi, J.; et al. Transcriptional Dynamics of Immortalized Human Mesenchymal Stem Cells during Transformation. PLoS ONE 2015, 10, e0126562. [Google Scholar]
  97. Takeuchi, M.; Takeuchi, K.; Kohara, A.; Satoh, M.; Shioda, S.; Ozawa, Y.; Ohtani, A.; Morita, K.; Hirano, T.; Terai, M.; et al. Chromosomal instability in human mesenchymal stem cells immortalized with human papilloma virus E6, E7, and hTERT genes. Vitr. Cell. Dev. Biol. Anim. 2007, 43, 129–138. [Google Scholar] [CrossRef]
  98. Kajiyama, S.; Ujiie, Y.; Nishikawa, S.; Inoue, K.; Shirakawa, S.; Hanada, N.; Liddell, R.; Davies, J.E.; Gomi, K. Bone formation by human umbilical cord perivascular cells. J. Biomed. Mater. Res. Part A 2015, 103, 2807–2814. [Google Scholar] [CrossRef]
  99. Okita, N.; Honda, Y.; Kishimoto, N.; Liao, W.; Azumi, E.; Hashimoto, Y.; Matsumoto, N. Supplementation of strontium to a chondrogenic medium promotes chondrogenic differentiation of human dedifferentiated fat cells. Tissue Eng. Part A 2015, 21, 1695–1704. [Google Scholar] [CrossRef]
  100. Toda, H.; Yamamoto, M.; Uyama, H.; Tabata, Y. Fabrication of hydrogels with elasticity changed by alkaline phosphatase for stem cell culture. Acta Biomater. 2016, 29, 215–227. [Google Scholar] [CrossRef]
  101. Funes, J.M.; Quintero, M.; Henderson, S.; Martinez, D.; Qureshi, U.; Westwood, C.; Clements, M.O.; Bourboulia, D.; Pedley, R.B.; Moncada, S.; et al. Transformation of human mesenchymal stem cells increases their dependency on oxidative phosphorylation for energy production. Proc. Natl. Acad. Sci. USA 2007, 104, 6223–6228. [Google Scholar] [CrossRef] [Green Version]
  102. Abarrategi, A.; Gambera, S.; Alfranca, A.; Rodriguez-Milla, M.A.; Perez-Tavarez, R.; Rouault-Pierre, K.; Waclawiczek, A.; Chakravarty, P.; Mulero, F.; Trigueros, C.; et al. c-Fos induces chondrogenic tumor formation in immortalized human mesenchymal progenitor cells. Sci. Rep. 2018, 8. [Google Scholar] [CrossRef] [PubMed]
  103. Gambera, S.; Abarrategi, A.; Rodríguez-Milla, M.A.; Mulero, F.; Menéndez, S.T.; Rodriguez, R.; Navarro, S.; García-Castro, J. Role of Activator Protein-1 Complex on the Phenotype of Human Osteosarcomas Generated from Mesenchymal Stem Cells. Stem Cells 2018, 36, 1487–1500. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Takeda, Y.; Mori, T.; Imabayashi, H.; Kiyono, T.; Gojo, S.; Miyoshi, S.; Hida, N.; Ita, M.; Segawa, K.; Ogawa, S.; et al. Can the life span of human marrow stromal cells be prolonged by bmi-1, E6, E7, and/or telomerase without affecting cardiomyogenic differentiation? J. Gene Med. 2004, 6, 833–845. [Google Scholar] [CrossRef] [PubMed]
  105. Nemoto, A.; Chosa, N.; Kyakumoto, S.; Yokota, S.; Kamo, M.; Noda, M.; Ishisaki, A. Water-soluble factors eluated from surface pre-reacted glass-ionomer filler promote osteoblastic differentiation of human mesenchymal stem cells. Mol. Med. Rep. 2017, 17, 3448–3454. [Google Scholar] [CrossRef] [PubMed]
  106. Carnero, A.; Blanco-Aparicio, C.; Kondoh, H.; Lleonart, M.E.; Martinez-Leal, J.F.; Mondello, C.; Scovassi, A.I.; Bisson, W.H.; Amedei, A.; Roy, R.; et al. Disruptive chemicals, senescence and immortality. Carcinogenesis 2015, 36, S19–S37. [Google Scholar] [CrossRef] [PubMed]
  107. An, P.; Sáenz Robles, M.T.; Pipas, J.M. Large T Antigens of Polyomaviruses: Amazing Molecular Machines. Annu. Rev. Microbiol. 2012, 66, 213–236. [Google Scholar] [CrossRef]
  108. Ozono, E.; Yamaoka, S.; Ohtani, K. To Grow, Stop or Die?—Novel Tumor-Suppressive Mechanism Regulated by the Transcription Factor E2F. In Future Aspects of Tumor Suppressor Gene; InTech: London, UK, 2013. [Google Scholar]
  109. Tomaić, V. Functional Roles of E6 and E7 Oncoproteins in HPV-Induced Malignancies at Diverse Anatomical Sites. Cancers 2016, 8, 95. [Google Scholar] [CrossRef] [Green Version]
  110. Tátrai, P.; Szepesi, Á.; Matula, Z.; Szigeti, A.; Buchan, G.; Mádi, A.; Uher, F.; Német, K. Combined introduction of Bmi-1 and hTERT immortalizes human adipose tissue-derived stromal cells with low risk of transformation. Biochem. Biophys. Res. Commun. 2012, 422, 28–35. [Google Scholar] [CrossRef]
  111. Somoza, R.A.; Welter, J.F.; Correa, D.; Caplan, A.I. Chondrogenic differentiation of mesenchymal stem cells: Challenges and unfulfilled expectations. Tissue Eng. Part B Rev. 2014, 20, 596–608. [Google Scholar] [CrossRef] [Green Version]
  112. Castro-Viñuelas, R.; Sanjurjo-Rodríguez, C.; Piñeiro-Ramil, M.; Hermida-Gómez, T.; Fuentes-Boquete, I.; de Toro-Santos, F.; Blanco-García, F.; Díaz-Prado, S. Induced pluripotent stem cells for cartilage repair: Current status and future perspectives. Eur. Cells Mater. 2018, 36, 96–109. [Google Scholar] [CrossRef]
  113. Finger, F.; Schörle, C.; Zien, A.; Gebhard, P.; Goldring, M.B.; Aigner, T. Molecular phenotyping of human chondrocyte cell lines T/C-28a2, T/C-28a4, and C-28/I2. Arthr. Rheumatol. 2003, 48, 3395–3403. [Google Scholar] [CrossRef] [PubMed]
  114. Dominici, M.; Le Blanc, K.; Mueller, I.; Slaper-Cortenbach, I.; Marini, F.; Krause, D.S.; Deans, R.J.; Keating, A.; Prockop, D.J.; Horwitz, E.M. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 2006, 8, 315–317. [Google Scholar] [CrossRef] [PubMed]
  115. Uder, C.; Brückner, S.; Winkler, S.; Tautenhahn, H.-M.; Christ, B. Mammalian MSC from selected species: Features and applications. Cytom. Part A 2018, 93, 32–49. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. 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] [PubMed]
  117. Cleary, M.A.; Narcisi, R.; Focke, K.; van der Linden, R.; Brama, P.A.J.; van Osch, G.J.V.M. Expression of CD105 on expanded mesenchymal stem cells does not predict their chondrogenic potential. Osteoarthr. Cartil. 2016, 24, 868–872. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Burns, J.S.; Harkness, L.; Aldahmash, A.; Gautier, L.; Kassem, M. Chromosome copy number variation in telomerized human bone marrow stromal cells; insights for monitoring safe ex-vivo expansion of adult stem cells. Stem Cell Res. 2017, 25, 6–17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Bianco, P.; Robey, P.G. Skeletal stem cells. Development 2015, 142, 1023–1027. [Google Scholar] [CrossRef] [Green Version]
Figure 1. PRISMA flow diagram showing the search process carried out to select articles to be analyzed for this review.
Figure 1. PRISMA flow diagram showing the search process carried out to select articles to be analyzed for this review.
Ijms 20 06286 g001
Table 1. Basic characteristics of the reviewed immortal MSC (iMSC) lines.
Table 1. Basic characteristics of the reviewed immortal MSC (iMSC) lines.
MSC LineImmortalization GenesImmortalization MethodTissueDonor CharacteristicsSTR GenotypingClonalityTumorigenicityReferences
hMSC-TSV40LTTransfectionBone marrowUnknownNoUnclear 1No (tested by soft agar)[44]
KM101SV40LTTransfectionBone marrow48-year-old maleNoYesNot tested[45,46]
L87/4SV40LTTransfectionBone marrow70-year-old maleNoYesNot tested/shown[17,47]
V54/2SV40LTTransfectionPeripheral bloodHealthy donorNoYesNot tested/shown[17,48]
iUC-MSCsSV40LTRetroviral transductionUmbilical cordUnknownNoNoNo (tested in IDM)[49]
iSuPsSV40LTRetroviral transductionCoronal sutures15 to 17-month-old malesNoNoNo (tested in IDM)[50]
TAg cellsSV40LTLentiviral transductionCranial periosteumHealthy (fracture patient)NoNoNot tested[51]
iDFCsSV40LTRetroviral transductionDental follicleThree young adults (18–20 years old)NoYesNot tested[52,53]
OA-MSCsSV40LTRetroviral transductionArticular cartilage (knee)Osteoarthritic 61-year-old male and 69-year-old femaleYesYes 2Not tested/shown[41,42]
KPE6/E7Retroviral transductionBone marrow61-year-old femaleNoNoNo (tested in IDM)[54,55,56,57]
UE6E7-16 3E6/E7Retroviral transductionBone marrow91-year-old femaleYesYesNot shown[58,59]
HS-27E6/E7Retroviral transductionBone marrowAdult donorNoYesNot tested[60,61]
PDLSC-Bmi1 4Bmi1Retroviral transductionPeriodontal ligament15 to 20-year-old donorsNoNoNot tested[62]
hMSC-hTERThTERTRetroviral transductionBone marrowHealthy 33-year-old maleNoNo 5No (tested in IDM)[30,33,34,63,64,65,66,67,68,69]
TERT4 (hMSC-hTERT derived)hTERTRetroviral transductionBone marrowHealthy 33-year-old maleNoNoNo (tested in IDM) 6[35,40,70]
iMSC#3hTERTRetroviral transductionBone marrowHealthy maleNoYesNo (tested in IDM)[71,72,73]
BMA13H 7hTERTRetroviral transductionBone marrowUnknownNoNoNot tested[74,75]
SCP-1hTERTLentiviral transductionBone marrowUnknownNoYesNo (tested in IMD and by soft agar assay)[14,20,24,25,38,76,77,78,79,80]
Y201hTERTLentiviral transductionBone marrowUnknownNoYesNo (tested in IDM)[19,36,81,82,83]
Y101hTERTLentiviral transductionBone marrowUnknown [19,32]
MSODhTERTLentiviral transductionBone marrowHealthy 55-year-old femaleYesYesNo (tested in IDM)[15,26]
ASC/TERT1hTERTRetroviral transductionAdipose tissueUnknownYesNoNo (soft agar assay)[22,84]
hASCs-T 7hTERTLentiviral transductionAdipose tissueTwo males and two females (21 to 59 years old)NoNoNo (soft agar assay)[85]
GB/hTERT MSCshTERTTransfectionUmbilical cordUnknownNoNoNo (soft agar assay)[86]
SDP11hTERTTransfectionDental pulp6 to 8-year-old donorsNoYesNot tested[87,88]
Pelt cellshTERTRetroviral transductionPeriodontal ligamentAdult donorNoNoNot tested/shown[31,89,90]
CMSC29hTERTRetroviral transductionPlacenta (Chorionic Villi)UnknownNoYesNo (tested by soft agar assay)[91,92,93]
DMSC23hTERTRetroviral transductionPlacenta (Decidua Basalis)UnknownNoYesNo (tested by soft agar assay)[91,92,93]
CPC531hTERTLentiviral transductionArticular cartilage (knee)65 to 75-year-old patientsNoUnclear 1Not tested/shown[94,95]
hASCs-TS
(same parental cells as hASCs-T)
hTERT and SV40LTLentiviral transductionAdipose tissueTwo males and two females (21 to 59 years old)NoNoNo (soft agar assay)[85]
3A6 (KP-derived)hTERT and E6/E7Transfection (hTERT)Bone marrow61-year-old femaleNoYesNot tested[39,55,56]
hASCs-TE
(same parental cells as hASCs-T)
hTERT and E6/E7 Lentiviral transductionAdipose tissueTwo males and two females (21 to 59 years old)NoNoNo (soft agar assay)[85]
UE6E7T-3 (same parental cells as UE6E7-16)hTERT and E6/E7Retroviral transductionBone marrow91-year-old femaleYesYesTested in soft agar at “low” (PDs ≤ 200) and high (PDs = 252) passages, with only high passage UE6E7T-3 being capable of forming colonies; high passage UE6E7T-3 formed sarcomas in IDM[28,96,97]
UE6E7T-11 (same parental cells as UE6E7-16)hTERT and E6/E7Retroviral transductionBone marrow91-year-old femaleYesYesNot shown[58,98]
UE6E7T-2 (same parental cells as UE6E7-16)hTERT and E6/E7Retroviral transductionBone marrow91-year-old femaleYesYesNot shown[99]
imhMSCshTERT and E6/E7Retroviral transductionBone marrowUnknownNoUnclear 1No (tested in IDM)[18,23,29,100]
3 Hits hMPChTERT and E6/E7Retroviral transductionBone marrowHealthy 34-year-old maleYesNoNo (tested in IDM; only c-Fos-transduced cells were tumorigenic)[101,102,103]
UE7T-13 (same parental cells as UE6E7-16)hTERT and E7Retroviral transductionBone marrow91-year-old femaleYesYesNot shown[21,37,88,104,105]
1 Clones have been generated but it is unclear if they have been used afterwards. 2 Several clones have been generated and analyzed. 3 Not completely immortalized because E6/E7 was sufficient to extend lifespan but not to bypass senescence. 4 Probably incompletely immortalized. 5 [30,68,69] employed hMSC-hTERT-derived clones. 6 TERT20 at higher passages formed tumors composed of mesoderm type cells in IDM. 7 Not completely immortalized because hTERT was not sufficient to bypass senescence.
Table 2. Differentiation potential of reviewed iMSC lines.
Table 2. Differentiation potential of reviewed iMSC lines.
MSC LineOsteogenic PotentialChondrogenic PotentialAdipogenic Potential
hMSC-TPositive for VKS and osteocalcin upregulation (increased compared with primary MSCs) [44]Not testedNot tested
KM101Positive for ALP activity [46]Not testedTested and no adipogenic differentiation potential was found (also not shown) [45]
L87/4Not tested/shownPositive for ABS and ColII immunostaining in 3D alginate and PGA/PLLA scaffolds [17]Not tested/shown
V54/2Not tested/shownPositive for ABS and ColII immunostaining in 3D alginate and PGA/PLLA scaffolds [17]Not tested/shown
iUC-MSCsPositive for Runx2 and Osteocalcin upregulation [49]Positive for Sox9 upregulation [49]Positive for PPARγ upregulation [49]
iSuPsPositive for ARS (increased if SV40LT is removed) and osteogenesis-related genes upregulation [50]No chondrogenic differentiation potential was found (also not shown) [50]Positive for OROS (increased if SV40LT is removed) [50]
TAg cellsPositive for hydroxyapatite formation (showing earlier and stronger mineralization than parental cells) and upregulation of osteogenesis-related genes (increased compared with primary cells) [51]Not testedNot tested
iDFCsPositive for ARS, APS, and osteogenesis-related genes upregulation; osteogenic potential similar to primary cells [52]Positive for ABS and SOX9 upregulation in 2D culture [52]Positive for OROS and adipogenesis-related genes upregulation (PPARγ and LPL) [52]
OA-MSCsPositive for ARS and ALP upregulation [41,42]Positive for SOS [41], ABS [42] and upregulation of Sox9, Col2A1, ACAN and COL10A1 [41,42] in pellet [41] and 2D culture [42]Positive for OROS (weak staining) and LPL upregulation [41]
KPPositive for APS, ARS, and VKS [54]Proved by ABS [54,57] and ColII immunostaining [57] in pellet culturePositive for OROS [54]
UE6E7-16Positive for osteocalcin production [59]Not tested/shownPositive for PPARγ production [59]
HS-27Positive for ALP activity, calcium deposition and osterix upregulation [61]Not tested/shownPositive for OROS in presence of steroids [60]
PDLSC-Bmi1Positive for ARS, ALP activity, and osteogenesis-related genes upregulation [62]Not testedPositive for OROS [62]
hMSC-hTERTPositive for ARS [64], ALP activity [33], upregulation of osteogenesis-related genes [33,63,68], and in vivo bone formation [63,68]Positive for ABS [64] and ColII immunostaining [63,67] in 2D culturePositive for OROS and upregulation of adipogenesis-related genes [33]
TERT4 (hMSC-hTERT derived)Positive for ARS [40,70], ALP activity [40] and upregulation of osteogenesis-related genes [40] Positive for ABS [35,70], GAG assay [35], and upregulation of ColII [35,70] but also ColX [35], in pellet culture; reduced compared with primary MSCsPositive for OROS [40,70] and upregulation of adipogenesis-related genes [40]
iMSC#3Positive for ARS, APS, and Runx2 upregulation [71]Positive for ABS, TBS, and GAG assay in pellet culture [72]; low chondrogenic potential but stimulation of chondrocyte differentiationPositive for OROS, adipogenesis-related genes upregulation [71,73], and NRS [73]
BMA13HPositive for ARS (reduced compared with primary cells) [74]Positive for ABS and GAG assay in 2D culture [74]; also positive for TBS, PSR and aggrecan and ColII immunostaining in 3D culture [75]; chondrogenic potential reduced compared with primary cells [74]Positive for OROS (reduced compared with primary cells) [74]
SCP-1Positive for VKS (increased compared with MSCs) [14], ARS [78], ALP activity [78], and upregulation of osteogenesis-related genes [76]Positive for TBS in pellet culture [14]; ColII and GAG production in 3D printed scaffolds [24]Positive for OROS [14]
Y201Positive for ARS [19], ALP activity [19,82,83], and Runx2 upregulation [19,82]Positive for ABS, GAG assay, and Sox9 upregulation in pellet culture [19]Positive for OROS (reduced compared to primary MSCs) and upregulation of adipogenesis-related genes [19,82]
Y101 (derived from the same donor than Y201)Proved by ARS [19,32], VKS [32], ALP activity, and osteogenesis-related genes’ upregulation [19,32]; osteogenic potential similar to Y201 [19]Positive for ABS, GAG assay and Sox9 upregulation in pellet culture; chondrogenic potential similar to Y201 [19]Positive for OROS and upregulation of adipogenesis-related genes; adipogenic potential reduced compared to Y201 [19]
MSODPositive for ARS [15], upregulation of osteogenesis-related genes [15,26], and in vivo bone formation [15]Weak positivity for ABS and upregulation of ColX but not ColII nor Sox9, similarly to primary parental cells; tested in pellet culture [15]Positive for OROS and PPARγ upregulation [15]
ASC/TERT1Positive for VKS and ALP activity [84]Positive for ABS, trichrome staining and ColII immunostaining in 3D scaffolds; reduced cartilage quality in comparison with chondrocytes [22]Positive for OROS and PPARγ upregulation; adipogenic potential increased compared with primary cells [84]
hASCs-TPositive for APS; reduced osteogenic potential in comparison with primary cells [85]Not testedTested by OROS, but almost no lipid droplets detected [85]
GB/hTERT MSCsPositive for ARS; reduced compared with primary cells [86]Not testedPositive for OROS [86]
SDP11Positive for BMP-2 and ALP upregulation [88]Not testedPositive for OROS but not shown [88]
Pelt cellsPositive for ARS (slightly reduced compared with primary cells) [90] and cementogenesis-related gene expression [31]Not testedNot tested
CMSC29Positive for ARS [91]Positive for ABS in pellet culture [91,92]Very weak positivity for OROS [91]
DMSC23Positive for ARS (increased compared with CMSC29) [91,93]Positive for ABS in pellet culture [91,92]Very weak positivity for OROS [91]
CPC531Positive for APS and upregulation of osteogenesis-related genes [95]Spontaneous chondrogenesis in 3D alginate culture, proved by upregulation of ColII and Sox9 and downregulation of Runx2 and ColI [95]Positive for OROS and upregulation of adipogenesis-related genes [95]
hASCs-TS (same parental cells as hASCs-T)Tested by APS, but no mineralization detected [85]Not testedPositive for OROS; reduced adipogenic potential in comparison with primary cells [85]
3A6 (KP-derived)Positive for ARS and VKS (increased compared with KP) [55], and also ALP activity [39]Positive for ABS [55] and ColII upregulation [56] in pellet culturePositive for OROS (reduced compared with KP) [55]
hASCs-TE (same parental cells as hASCs-T)Positive for APS; increased in comparison with primary cells [85]Not testedPositive for OROS; slightly reduced in comparison with primary cells [85]
UE6E7T-3 (same parental cells as UE6E7-16)Positive for ALP activity [97], ARS and upregulation of osteogenesis-related genes [28]Not tested/shownPositive for OROS [97]
UE6E7T-11 (same parental cells as UE6E7-16)Positive for APS and bone sialoprotein (BSP) upregulation [98]Not tested/shownNot tested/shown
UE6E7T-2 (same parental cells as UE6E7-16)Not tested/shownTested by ABS in 2D culture; negative under employed conditions [99]Not tested/shown
imhMSCsPositive for VKS and upregulation of osteogenesis-related genes [18]Weak positivity for ABS and with weak upregulation of chondrogenesis-related genes (similarly to primary parental cells); tested in pellet culture [18]Positive for OROS and PPARγ upregulation [18]
3 Hits hMPCPositive for ARS [102,103], APS [101,102] and Runx2 upregulation [102]Positive for ABS [102,103] and TBS [101] in pellet culture, but reduced compared with primary MSCsPositive for OROS [101,102,103], but reduced compared with primary MSCs
UE7T-13 (same parental cells as UE6E7-16)Positive for ARS [37,88,105] and ALP activity [88]Not tested/shownPositive for OROS [37,88]

Share and Cite

MDPI and ACS Style

Piñeiro-Ramil, M.; Sanjurjo-Rodríguez, C.; Castro-Viñuelas, R.; Rodríguez-Fernández, S.; Fuentes-Boquete, I.M.; Blanco, F.J.; Díaz-Prado, S.M. Usefulness of Mesenchymal Cell Lines for Bone and Cartilage Regeneration Research. Int. J. Mol. Sci. 2019, 20, 6286. https://doi.org/10.3390/ijms20246286

AMA Style

Piñeiro-Ramil M, Sanjurjo-Rodríguez C, Castro-Viñuelas R, Rodríguez-Fernández S, Fuentes-Boquete IM, Blanco FJ, Díaz-Prado SM. Usefulness of Mesenchymal Cell Lines for Bone and Cartilage Regeneration Research. International Journal of Molecular Sciences. 2019; 20(24):6286. https://doi.org/10.3390/ijms20246286

Chicago/Turabian Style

Piñeiro-Ramil, M., C. Sanjurjo-Rodríguez, R. Castro-Viñuelas, S. Rodríguez-Fernández, I.M. Fuentes-Boquete, F.J. Blanco, and S.M. Díaz-Prado. 2019. "Usefulness of Mesenchymal Cell Lines for Bone and Cartilage Regeneration Research" International Journal of Molecular Sciences 20, no. 24: 6286. https://doi.org/10.3390/ijms20246286

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