Next Article in Journal
Lumbar Spinal Stenosis Treatment: Is Surgery Better than Non-Surgical Treatments in Afro-Descendant Populations?
Next Article in Special Issue
A Family with Myh7 Mutation and Different Forms of Cardiomyopathies
Previous Article in Journal
Secondary Terpenes in Cannabis sativa L.: Synthesis and Synergy
Previous Article in Special Issue
Vascular Calcification: In Vitro Models under the Magnifying Glass
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Brief Report

Cardiac Mesenchymal Stem Cell-like Cells Derived from a Young Patient with Bicuspid Aortic Valve Disease Have a Prematurely Aged Phenotype

1
Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, William Henry Duncan Building, 6 West Derby Street, Liverpool L7 8TX, UK
2
Newcastle University Bioscience Institute, Newcastle University, International Centre for Life, Central Parkway, Newcastle upon Tyne NE1 3BZ, UK
3
Department of Cardiothoracic Surgery, South Tees Hospitals NHS Foundation Trust, Middlesbrough TS4 3BW, UK
4
Graduate Institute of Biomedical Materials and Tissue Engineering, Taipei Medical University, Taipei 110, Taiwan
*
Author to whom correspondence should be addressed.
Biomedicines 2022, 10(12), 3143; https://doi.org/10.3390/biomedicines10123143
Submission received: 4 November 2022 / Revised: 25 November 2022 / Accepted: 28 November 2022 / Published: 6 December 2022

Abstract

:
There is significant interest in the role of stem cells in cardiac regeneration, and yet little is known about how cardiac disease progression affects native cardiac stem cells in the human heart. In this brief report, cardiac mesenchymal stem cell-like cells (CMSCLC) from the right atria of a 21-year-old female patient with a bicuspid aortic valve and aortic stenosis (referred to as biscuspid aortic valve disease BAVD-CMSCLC), were compared with those of a 78-year-old female patient undergoing coronary artery bypass surgery (referred to as coronary artery disease CAD-CMSCLC). Cells were analyzed for expression of MSC markers, ability to form CFU-Fs, metabolic activity, cell cycle kinetics, expression of NANOG and p16, and telomere length. The cardiac-derived cells expressed MSC markers and were able to form CFU-Fs, with higher rate of formation in CAD-CMSCLCs. BAVD-CMSCLCs did not display normal MSC morphology, had a much lower cell doubling rate, and were less metabolically active than CAD-CMSCLCs. Cell cycle analysis revealed a population of BAVD-CMSCLC in G2/M phase, whereas the bulk of CAD-CMSCLC were in the G0/G1 phase. BAVD-CMSCLC had lower expression of NANOG and shorter telomere lengths, but higher expression of p16 compared with the CAD-CMSCLC. In conclusion, BAVD-CMSCLC have a prematurely aged phenotype compared with CAD-CMSCLC, despite originating from a younger patient.

1. Introduction

A number of resident cardiac stem cell populations have been identified, several of which are present in the human heart [1,2,3,4,5]. However, clinical trials utilizing bone marrow or cardiac-derived stem cells have delivered only modestly successful results, with only a few patients showing meaningful improvements in cardiac function [6,7].
It is now accepted that most therapeutic benefits of MSC transplantation for cardiac repair are driven by paracrine secretions of numerous growth factors, cytokines and extracellular vesicles that aid in promoting cardiomyocyte survival, angiogenesis, and steering remodeling towards a less fibrotic phenotype [8]. Despite this, their mechanisms of action within the heart remain controversial and there has also been consideration of their possible role in tumor formation [9,10]. Further challenges are presented in the production of clinically relevant cell therapy products, including the influence of in vitro ageing during culture expansion [11], and limited retention and survival of donor cells at the target site, which requires combination of cells with supporting biocompatible materials [12,13,14,15,16].
It is well-known that not all donor cells are created equal [17]. For example, a previous study has shown that myocardial infarction adversely influences the therapeutic potential of bone marrow-derived cells from the same donor [18]. Furthermore, the secretome varies dynamically according to phenotype of the originating cells. Extracellular vesicles derived from post-MI mouse hearts were shown to aggravate inflammation and worsened heart function in other animals [19]. Thus, the source of donor therapeutic cells might also impact on the therapeutic outcome. By their very nature, patients requiring cardiac surgical intervention are rarely healthy or absent of cardiovascular disease. Therefore, if cardiac-derived cells are to be used for therapy, better understanding of the effects of cardiac disease and/or aging on those cells is needed.
Coronary artery disease (CAD) is a common disease caused by the buildup of atheroma in the coronary arteries that causes narrowing of the arteries and reduced blood flow to the heart [20]. This atherosclerotic process is, however, not restricted to the coronary arteries or even to the heart and can affect a number of different organs in the body and can therefore be considered a systemic disease [21]. The loss and/or dysfunction of MSCs has been associated with many systemic diseases (reviewed in Vizoso et al., 2019 [22]), while circulating endothelial progenitor cells (CPCs) expressing osteocalcin have been shown to be present in higher numbers in patients with coronary atherosclerosis than those without [23]. These studies support a role for progenitors/stem cells as either contributing to, or as being targets of, disease progression in some systemic diseases, including CAD. Contrary to this, bicuspid aortic valve defects are a form of congenital heart defect, often associated with several other cardiac complications, the symptoms of which frequently become apparent with increasing age [24,25].
One cell fate known to impede stem cell function is cellular senescence, controlled by the p16-pRb and p53-p21 pathways and defined as a cell cycle arrest, alterations in gene and protein expression and the production of the senescence-associated secretory phenotype (SASP) [26], a cocktail of pro-inflammatory cytokines, chemokines, matrix proteases and growth factors, which in the heart can impact tissue function, attenuate regeneration, induce fibrosis, extracellular matrix degeneration and drive inflammation [27]. Cardiac regenerative potential declines with age [28] and populations of human cardiac progenitor cells (CPCs) accumulate the senescence phenotype with age, express p16 and are unable to replicate, differentiate, regenerate or restore cardiac function following transplantation into the infarcted heart [29]. Our recent studies demonstrate that in the heart, cardiomyocyte, fibroblast and endothelial senescence can be induced not only by ageing but also as a result of cellular stress and disease [30,31]. The effect of disease on progenitor cells independent of ageing has not yet been investigated.
We have previously reported on the identification of human cardiac mesenchymal stem cell-like cells (CMSCLC), which have stem cell-like characteristics and an immunophenotype typical of MSC. These cells were capable of low levels of adipogenic differentiation but failed to differentiate into osteoblasts or chondrocytes. However, these CMSCLC did, under cardiac differentiation conditions, have the phenotype of both mature and immature cardiac cells, expressing troponin C and Nkx2.5, respectively [5]. CMSCLC express many of the bioactive molecules that make up the cardio-beneficial paracrine secretome, including, interleukin-10 (IL10), fibroblast growth factor-2 (FGF2), vascular endothelial growth factor (VEGF), transforming growth factor (TGF) and hepatocyte growth factor (HGF) [5], and their potential for therapeutic application demonstrated in vivo by improved cell retention, survival, extracellular vesicle production, and promotion of functional cardiac repair when encapsulated and delivered to a murine model of myocardial infarction ischemic injury [32].
Clinical presentation of a 21-year-old female patient with relatively rare bicuspid aortic valve disease (BAVD) provided an opportunity to present a case-based study that contributes to advancing the understanding of the impact of both age and different cardiomyopathies on the health of CMSCLC, and thus the potential implications for application of cardiac-derived stem cell-based therapies in clinic. We have isolated and compared CMSCLC from the atrial appendage of both the BAVD patient and from a 78-year-old female patient with coronary artery disease (CAD). We provide evidence that CMSCLC from the 21-year-old BAVD patient have a prematurely aged phenotype when compared with CMSCLC from the 78-year-old CAD patient. This suggests that premature aging of resident cardiac stem cells may contribute to additional cardiac complications observed in some patients with BAVD.

2. Materials and Methods

2.1. Cardiac MSC Isolation and Derivation

All cardiac tissue samples used in this study were collected from consenting patients undergoing cardiac surgery under REC number UKCRN ID: 20120092. Right atrial appendage CMSCLC were derived and cultured as described previously [5]. Assessment of colony-forming unit-fibroblast (CFU-F) formation was performed as described previously [33].

2.2. Immunophenotyping of Cardiac MSC Populations

Immunophenotyping of CMSCLC for expression of cell surface antigens, including CD44, CD73, CD90, CD105, CD106, CD146, CD166, CD19 and CD45 was performed and analyzed as described previously with all primary antibodies purchased from R&D Biosystems, Abingdon, UK [5].

2.3. Measurement of Metabolic Activity

The metabolic activity of CMSCLC as an indicator of cell health was measured by intracellular reduction of resazurin (IUPAC: 7-hydroxy-10-oxidophenoxazin-10-ium-3-one) to resorufin (absorbance = A570) using alamarBlue® metabolic assay (Thermo Fisher Scientific, Paisley, UK) as previously described [33,34,35]. For normalization of cell numbers DNA was quantified using Quant-iT™ PicoGreen® dsDNA Kit (Thermo Fisher Scientific) as previously described [33].

2.4. Measurement of Telomere Length

Measurement of telomere length was performed at passage 5 of culture using the TeloTAGGG Telomere Length Assay (Roche, St Albans, UK) according to manufacturer’s instructions. Genomic (g)DNA (5 μg) was purified using the QIAamp DNA mini kit (Qiagen, Manchester, UK) and digested in parallel with 1.5 μg positive control DNA with 20 U/μL each of restriction endonucleases Hinf I and Rsa I. Telomere fragments were resolved by 0.8% (wt/vol) agarose gel electrophoresis, blotted by capillary action onto nylon membrane and hybridized with a digoxigenin (DIG)-labelled telomere-specific hybridization probe. Telomere fragments were visualized by chemiluminescence following incubation with 75 mU/mL of anti-DIG antibody conjugated with alkaline phosphatase and CDP-Star chemiluminescence substrate.

2.5. Immunocytochemistry

Cells were cultured on chamberslides and fixed in 4% (wt/vol) paraformaldehyde for 5 min and labelled with primary antibody specific to Nanog (dilution 1:200, ab62734, Abcam, Cambridge, UK) or p16 (dilution 1:50, J0411, Santa Cruz, Heidelberg, UK). An Alexa-488 conjugated secondary antibody (dilution 1:1000, Molecular Probes; Thermo Fisher Scientific) was used to detect primary labelling. Nuclei were labelled with 0.2 mg/mL 4′,6-diamidino-2-phenylindole (DAPI, Sigma, Poole, UK). Analysis was performed using an Axioimager M1 fluorescence microscope (Carl Zeiss) running OpenLab software (Improvision). The specificity of all secondary antibodies and the absence of autofluorescence were tested by omitting primary control. To allow comparison of protein expression level in the different patient samples the same exposure time was used for each sample.

2.6. Cell Cycle Analysis

Cell cycle analysis was carried out using the CyStain DNA 2 step kit (Partec, Wymbush, UK) CMSCs at 90% confluence were resuspended in 100 μL extraction buffer and incubated at room temperature for 15 min. Cells were labelled with the addition of 500 μL staining solution (containing DAPI) and incubated overnight at 4 °C. Flow cytometry was performed on FACS Canto (BD) using 405 450/50 filters.

3. Results

3.1. Colony-Forming Unit Fibroblast Potential, Cell Derivation Rate and Morphological Analysis of CMSCCLC

Comparative analysis of CMSCLC in vitro derivation was made between BAVD and CAD tissues. The ability of CMSCLC to form colony-forming unit fibroblasts (CFU-Fs) was examined with BAVD-CMSCLC producing a total of 5 CFU-Fs, fewer than the CAD-CMSCLC, which formed 14 CFU-Fs (Figure 1A). The number of cells that could be derived from these CFU-Fs was determined at passage 1. BAVD-CMSCLC CFU-Fs gave rise to 3.34 × 105 cells and CAD-CMSCLC CFU-Fs gave rise to 5.5 × 106 cells (Figure 1B). To allow time for cells to adapt from an in vivo to an in vitro environment, cell doublings were assessed at passages 3–5 and showed that BAVD-CMSCLC had a much lower cell doubling rate than CAD-CMSCLC (Figure 1C). The morphology of BAVD- and CAD-CMSCLC was examined at passage 5. BAVD-CMSCLC appeared ‘prematurely aged’, being flatter and more irregular in shape with large cytoplasmic volume in comparison to CAD-CMSCLC, which had a more typical fibroblastic MSC morphology, having a small cell body with long cellular protrusions (Figure 1D,E).

3.2. CMSCLC Immunophenotyping

The immunophenotype of CMSCLC derived from BAVD and CAD tissues was determined by profiling a panel of cell surface antigens established as being positively or negatively expressed by MSC. CMSCLC were immunolabelled using antibodies to CD44, CD73, CD90, CD105, CD106, CD146, CD166, CD45 and CD19 and used in combination with flow cytometry [36]. Analysis of the data generated revealed that both cell populations expressed markers normally expressed by MSCs and CMSCLCs, and were negative for expression of CD45 and CD19 (Figure 2).

3.3. Expression of NANOG and p16 in CMSCLC

Evaluation of the CMSCLC stem cell phenotype was performed by immunolabelling BAVD- and CAD-CMSCLC with anti-human NANOG antibody at passage 5. Nuclear-specific expression of NANOG was observed in both BAVD-CMSCLC and CAD-CMSCLC, however expression of NANOG appeared to be downregulated in BAVD-CMSCLC, being expressed in 3.8% of the cell population compared to 22.9% of CAD-CMSCLC (Figure 3A,B).
In response to the ‘prematurely aged’ cellular morphology observed in BAV-CMSCLC, cells were also immunolabelled for p16, a key regulator of cellular senescence, which is upregulated in cells causal to systemic ageing [37], including senescent MSC [38] and senescent cardiac cells that contribute to myocardial remodeling [31,39], and which in a recent systemic review was identified as a robust marker for the detection of senescence in human tissue samples [40]. BAVD-CMSCLC were shown to have higher expression of p16 (48.6% of culture) compared to CAD-CMSCLC (14.7% of culture) (Figure 3C,D).

3.4. Evaluation of Cellular Ageing My Measurement of Telomere Length, Metabolic Activity and Cell Cycle Kinetics

Further comparative investigation of cellular ageing in BAV-CMSCLC and CAD-CMSCLC was performed at passage 5. Analysis of telomere length demonstrated that BAVD-CMSCLC had shorter telomeres (5.3 kbp) than CAD-CMSCLC (8.3 kbp) (Figure 4A).
Metabolic activity was also measured using an alamarBlue® assay normalized to total DNA and was lower in BAVD-CMSCLC compared to CAD-CMSCLC (Figure 4B). CMSCCL were also stained and analyzed for cell cycle progression, which showed a population of BAVD-CMSCLC in G2/M phase, whereas the bulk of CAD-CMSCLC were in the G0/G1 phase of the cell cycle (Figure 4C,D).

4. Discussion

Bicuspid aortic valve defects are a common form of congenital heart defect, often associated with a number of other cardiac complications, the symptoms of which frequently become apparent with increasing age [24,25]. The causes of BAVD, however, remain unclear. We report for the first time on differences between CMSCLC isolated from of a young female patient with BAVD disease and an elderly female patient with CAD disease. We observed that both the CAD-CMSCLC and BAVD-CMSCLC expressed cell surface makers normally expressed by MSCs and lack expression of hematopoietic lineage makers. Moreover, the CMSCLCs adhere to plastic under standard MSC culture conditions [41,42]. However, morphologically, while the CAD-CMSCLC displayed an MSC-like morphology [41], the BAVD-CMSCLC did not; they displayed characteristics associated with aged MSCs [43].
One well-recognized stem cell characteristic is the ability to form colonies in culture; we observed that CAD-CMSCLC formed more CFU-Fs than the BAVD-CMSCLC. They also formed fewer CFU-Fs than for other patient-derived CMSCLC reported in our previous study [5]. Moreover, the number of cells that could be derived from the CAD-CFU-Fs was higher than those that could be derived from the BAVD-CFU-Fs. BAVD-CMSCLC also had a slower doubling rate and a reduced capacity for cell division compared with CAD-CMSCLC, both of which are characteristics of aging [43]. The BAVD-CMSCLC also had a shorter telomere length than the CAD-CMSCLC. The replicative potential of hematopoietic stem cells is related to telomere length and as such this provides an indication of stem cell function and there is increasing evidence that telomerase activity and telomere length are important for the function of bone marrow derived MSCs. Mouse MSCs that lack telomerase activity show an inability to differentiate into adipocytes or chondrocytes [44,45] while human MSCs, forced to overexpress telomerase, have increased proliferative potential [45]. It has been suggested that in human MSCs telomerase activity is required to bring about regenerative capacity and differentiation potential [46]. The shorter telomeres in the BAVD-CMSCLC may therefore be an indicator of the inferior quality of these cells compared with the CAD-CMSCLC and suggest that like bone marrow MSCs telomerase activity and telomere length is required to maintain the CMSCLC proliferative potential.
NANOG is associated with biological processes important for stem cell function including proliferation and differentiation potential [47,48,49]. In aging murine bone marrow MSCs, the over-expression of NANOG reversed ageing-associated loss of proliferation and myogenic potential [50]. We observed NANOG expression in both CMSCLC populations; however, there was decreased expression in terms of cell numbers in BAVD-CMSCLC. Furthermore, p16 an inhibitor of cell cycle progression most commonly associated with cell senescence including, tissue resident MSC and cardiomyocytes, was upregulated in BAVD-CMSCLC compared to CAD-CMSCLC [30,51,52]. While p16 is a robust marker of human senescence in vivo [40], the senescent phenotype can be heterogeneric and dependent on cell type and stimuli as such a deeper analysis of senescence-associated gene expression would provide insight into the signature of senescent CMSCLCs [53].
Cell cycle analysis revealed difference in cell cycle kinetics between the CAD and BAVD CMSCLC, with a subpopulation of BAVD cells being in the G2/M phase of the cell cycle which can be indicative of a stressed or senescent cell phenotype [50]. Recent studies have demonstrated that epigenetic biomarkers of ageing are prognostic of disease across multiple tissues including the cardiovascular system. [54,55,56]. While it is beyond the scope of the current study, a future investigation of epigenetic changes within CMSCLC populations could shed light on if epigenetic changes are associated with CMSCLC age and disease-related dysfunction.
Finally, we investigated the health of both populations of cells. Quantification of metabolic activity by alamarBlue® assay is an established indicator of cell health whereby measurement of the reducing power of the intracellular environment includes contribution from mitochondrial and cytoplasmic reductases, and is therefore a function of both aerobic glycolysis and oxidative phosphorylation, respectively [34]. Our observation that the BAVD-CMSCLC were less metabolically active than the CAD-CMSCLC suggests that these cells have an impaired cellular metabolism, consistent with the established phenotype of senescent cells [34,57,58]. Metabolic balance is known to couple bioenergetic state with broader physiological pathways that regulate MSCs phenotype and function, including the metabolic switch of aerobic glycolysis to oxidative phosphorylation that drives differentiation [59]. Whilst aerobic glycolysis might be expected to be the predominant metabolic pathway in rapidly proliferating CMSCLC, the involvement of oxidative phosphorylation in the production of metabolite intermediates that form the precursors of biosynthetic pathways, as well as the metabolism of glutamine to glutathione for the regulation of REDOX signaling should also be considered [60,61,62]. Dysregulation of these integrated metabolic pathways is linked to elevated production of reactive oxygen species (ROS) and cellular oxidative stress that drives the senescent phenotype, and we propose this mechanism as one that warrants further interrogation in determining the health status of patient-derived CMSCLC [60,61].
The premature senescence evidenced in the BAVD-CMSCLCs is intriguing when considering that BAVD has no anatomical or embryological link with the right atrium (the aortic valve originates from neural crest and mutations tend not to affect right heart), whereas CAD associated with significant atherosclerosis is a systemic illness as opposed to a very organ-specific abnormality [63,64]. We suggest that in atherosclerotic cardiovascular disease, vascular remodeling, whilst being initiated by dysfunctional endothelial cells, is contributed to by stem cells originating from several sources. However, these cells have been shown to differentiate primarily to adipocytes, chondrocytes and osteocytes [65]. In our previously published study, CMSCLC from RAA of patients with CAD also had a very poor ability to undergo this tri-lineage differentiation [5], suggesting that CMSCLC from RAA are distinct from the stem cells contributing to cardiovascular disease. Resident cardiac stem cells known as CASCs, have also been isolated from atrial appendages based on aldehyde dehydrogenase activity and have been shown to be present in both human and pig heart. In the pig examination of different regions of the heart for the presence of CASCs revealed the highest numbers to be present in RAA and it is therefore a good source of stem cells that are relevant to cardiac-specific therapeutic application [66]. Interestingly, it has been proposed that individuals with BAV are predisposed to senescence [67]. Individuals with BAV display an increased prevalence of thoracic aortic aneurysm (TAA) and have a significantly increased risk of aortic dissection compared to those with a typical tricuspid valve (TAV), while senescence is associated with TAA in both TAV and BAV patients, only aortas of individuals with BAV contained senescence cells in the absence of TAA [68]. Observations that suggest BAV aortas have an increased predisposition to senescence even in the absence of symptomatic disease. Although the cause of this predisposition is unknown, however the genetics contributing to the disease may play a role, therefore it is possible that other cardiac resident cells including CMSCLCs share this susceptibility to senescence.
The ultimate goal for stem cell research is cellular therapy; therefore, it is imperative that we fully understand how donor disease and genetics effects stem cell biology. Whilst our study is limited by access to cardiac tissue of one donor with relatively rare BAVD, our data suggests that genetic valvular disease can promote a senescence phenotype, and raises important questions regarding the appropriateness of using stem cells from diseased individuals, which may be required for autologous transplantation. Studies investigating age-related senescence have demonstrated that c-kit expressing cardiac progenitor cells have impaired stem cell function and express a proinflammatory SASP that promotes senescence in healthy CPC populations [29]. As such, cellular therapies that use cell populations, which include senescent cells may not only be less effective but may in fact be detrimental to both the transplanted population as a whole and the organ into which they are transplanted. Indeed, in animal models, transplantation of small numbers of senescent cells induces age-related disease, increases frailty, and increases mortality [6,7,8]. It is possible that in the future some of these challenges could be overcome isolated senescent CPCs can be “rejuvenated” via the treatment with senolytics which induce senescent but not proliferative cells to apoptosis [29].

5. Conclusions

In conclusion, our data suggest that the CMSCLC derived from the 21-year-old patient with BAVD disease were less metabolically active, less proliferative, had begun to show less ‘stemness’ characteristics and were prematurely aged compared with those of the 78-year-old patient with CAD. These findings have implications for the use of autologous cardiac derived stem cells for use as a therapeutic tool.

Author Contributions

Conceptualization: R.A.O. and A.M.; Methodology: R.A.O., G.R., P.C. and A.M.; Validation: R.A.O., G.R., P.C., W.A.O., D.J.L. and A.M.; Formal Analysis: R.A.O., G.R., P.C., W.A.O., D.J.L. and A.M.; Investigation: R.A.O., G.R., P.C., W.A.O., D.J.L. and A.M.; Resources: R.A.O., W.A.O. and A.M.; Data Curation: R.A.O. and A.M.; Writing—Original Draft Preparation: R.A.O. and A.M.; Writing—Review and Editing: R.A.O., G.R., P.C., W.A.O., D.J.L. and A.M.; Project Administration: A.M.; Funding Acquisition: R.A.O. and A.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded the Wellcome Trust Institutional Strategic Support Fund awarded to the University of Liverpool (grant number 097826/z/11/z). The APC was funded by the University of Liverpool.

Institutional Review Board Statement

The study was conducted in accordance with the Declaration of Helsinki, and approved by Research Ethics Committee UKCRN ID: 20120092.

Informed Consent Statement

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

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Mishra, R.; Vijayan, K.; Colletti, E.J.; Harrington, D.A.; Matthiesen, T.S.; Simpson, D.; Goh, S.K.; Walker, B.L.; Al-meida-Porada, G.; Wang, D.; et al. Characterization and functionality of car-diac progenitor cells in congenital heart patients. Circulation 2011, 123, 364–373. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Alfakir, M.; Dawe, N.; Eyre, R.; Tyson-Capper, A.; Britton, K.; Robson, S.C.; Meeson, A.P. The temporal and spatial expression pattern of ABCG2 in the embryonic/fetal human heart. Int. J. Cardiol. 2012, 156, 133–138. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Sandstedt, J.; Jonsson, M.; Kajic, K.; Sandstedt, M.; Lindahl, A.; Dellgren, G.; Jeppsson, A.; Asp, J. Left atrium of the human adult heart contains a population of side population cells. Basic Res. Cardiol. 2012, 107, 255. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Smith, R.R.; Barile, L.; Cho, H.C.; Leppo, M.K.; Hare, J.; Messina, E.; Giacomello, A.; Abraham, M.R.; Marbán, E. Regenerative Potential of Cardiosphere-Derived Cells Expanded from Percutaneous Endomyocardial Biopsy Specimens. Circulation 2007, 115, 896–908. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Oldershaw, R.; Owens, W.A.; Sutherland, R.; Linney, M.; Liddle, R.; Magana, L.; Lash, G.E.; Gill, J.H.; Richardson, G.; Mee-son, A. Human Cardiac-Mesenchymal Stem Cell-Like Cells, a Novel Cell Population with Therapeutic Potential. Stem Cells Dev. 2019, 28, 593–607. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Schächinger, V.; Erbs, S.; Elsässer, A.; Haberbosch, W.; Hambrecht, R.; Hölschermann, H.; Yu, J.; Corti, R.; Mathey, D.G.; Hamm, C.W.; et al. REPAIR-AMI Investigators. Intracoronary bone marrow-derived progenitor cells in acute myocardial infarction. N. Engl. J. Med. 2006, 355, 1210–1221. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Yousef, M.; Schannwell, C.M.; Köstering, M.; Zeus, T.; Brehm, M.; Strauer, B.E. The BALANCE Study: Clinical benefit and long-term outcome after intracoronary autologous bone marrow cell transplantation in patients with acute myocardial infarction. J. Am. Coll. Cardiol. 2009, 53, 2262–2269. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Blume, G.G.; Machado-Junior, P.A.B.; Simeoni, R.B.; Bertinato, G.P.; Tonial, M.S.; Nagashima, S.; Pinho, R.A.; de Noronha, L.; Olandoski, M.; de Carvalho, K.A.T.; et al. Bone-Marrow Stem Cells and Acellular Human Amniotic Membrane in a Rat Model of Heart Failure. Life 2021, 11, 958. [Google Scholar] [CrossRef] [PubMed]
  9. Klopp, A.H.; Gupta, A.; Andreeff, M.; Marini, F. Concise Review: Dissecting a discrepancy in the literature: Do mesenchymal stem cells support or suppress tumor growth? Stem Cells 2011, 29, 11–19. [Google Scholar] [CrossRef] [Green Version]
  10. Chien, K.R.; Frisén, J.; Fritsche-Danielson, R.; Melton, D.A.; Murry, C.E.; Weissman, I.L. Regenerating the field of cardiovascular cell therapy. Nat. Biotechnol. 2019, 37, 232–237. [Google Scholar] [CrossRef] [PubMed]
  11. Yang, Y.H.K.; Ogando, C.R.; Wang See, C.; Tsui-Yun, C.; Barabino, G.A. Changes in phenotype and differentiation potential of human mesenchymal stem cells aging in vitro. Stem Cell Res. Ther. 2018, 9, 131. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Gao, L.; Kupfer, M.E.; Jung, J.P.; Yang, L.; Zhang, P.; Da Sie, Y.; Tran, Q.; Ajeti, V.; Freeman, B.T.; Fast, V.G.; et al. Myocardial Tissue Engineering with cells derived from human-induced pluripotent stem cells and a native-like, high-resolution, 3-dimensionally printed scaffold. Circ. Res. 2017, 120, 1318–1325. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Lee, A.S.; Inayathullah, M.; Lijkwan, M.A.; Zhao, X.; Sun, W.; Park, S.; Hong, W.X.; Parekh, M.B.; Malkovskiy, A.V.; Lau, E.; et al. Prolonged survival of transplanted stem cells after ischaemic injury via the slow release of pro-survival peptides from a collagen matrix. Nat. Biomed. Eng. 2018, 2, 104–113. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Su, T.; Huang, K.; Daniele, M.A.; Hensley, M.T.; Young, A.T.; Tang, J.; Allen, T.A.; Vandergriff, A.C.; Erb, P.D.; Ligler, F.S.; et al. Cardiac Stem Cell Patch Integrated with Microengineered Blood Vessels Promotes Cardiomyocyte Proliferation and Neovascularization after Acute Myocardial Infarction. ACS Appl. Mater. Interfaces 2018, 10, 33088–33096. [Google Scholar] [CrossRef]
  15. Zhang, J.; Zhu, W.; Radisic, M.; Vunjak-Novakovic, G. Can We Engineer a Human Cardiac Patch for Therapy? Circ. Res. 2018, 123, 244–265. [Google Scholar] [CrossRef] [PubMed]
  16. Christman, K.L. Biomaterials for Tissue Repair. Science 2019, 363, 340–341. [Google Scholar] [CrossRef] [PubMed]
  17. Johnston, P.V.; Duckers, H.J.; Amish, N.; Raval, A.N.; Cook, T.D.; Pepine, C.J. Not all stem cells are created equal, the case for prospective assessment of stem cell potency in the CardiAMP Heart Failure Trial. Circ. Res. 2018, 123, 944–946. [Google Scholar] [CrossRef]
  18. Wang, X.; Takagawa, J.; Lam, V.C.; Haddad, D.J.; Tobler, D.L.; Mok, P.Y.; Zhang, Y.; Clifford, B.T.; Pinnamaneni, K.; Saini, S.A.; et al. Donor myocardial infarction impairs the therapeutic potential of bone marrow cells by an interleukin-1-mediated inflammatory response. Sci. Transl. Med. 2011, 3, 100ra90. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Ge, X.; Meng, Q.; Wei, L.; Liu, J.; Li, M.; Liang, X.; Lin, F.; Zhang, Y.; Li, Y.; Liu, Z.; et al. Myocardial ischemia-reperfusion induced cardiac extracellular vesicles harbour proinflammatory features and aggravate heart injury. J. Ex-Tracell Vesicles 2021, 10, e12072. [Google Scholar] [CrossRef] [PubMed]
  20. Malakar, A.K.; Choudhury, D.; Halder, B.; Paul, P.; Uddin, A.; Chakraborty, S. A review on coronary artery disease, its risk factors, and therapeutics. J. Cell Physiol. 2019, 234, 16812–16823. [Google Scholar] [CrossRef] [PubMed]
  21. Montecucco, F.; Mach, F. Atherosclerosis is an inflammatory disease. Semin. Immunopathol. 2009, 31, 1–3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Vizoso, F.J.; Eiro, N.; Costa, L.; Esparza, P.; Landin, M.; Diaz-Rodriguez, P.; Schneider, J.; Perez-Fernandez, R. Mesenchymal Stem Cells in Homeostasis and Systemic Diseases: Hypothesis, Evidences, and Therapeutic Opportuni-ties. Int. J. Mol. Sci. 2019, 20, 3738. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Gössl, M.; Mödder, U.I.; Gulati, R.; Rihal, C.S.; Prasad, A.; Loeffler, D.; Lerman, L.O.; Khosla, S.; Lerman, A. Coronary endothelial dysfunction in humans is associated with coronary retention of osteogenic endothelial progenitor cells. Eur. Heart J. 2010, 31, 2909–2914. [Google Scholar] [CrossRef]
  24. Ward, G. Clinical significance of the bicuspid aortic valve. Heart 2000, 83, 81–85. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Siu, S.C.; Silversides, C.K. Bicuspid Aortic Valve Disease. JACC 2010, 55, 2789–2800. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Coppé, J.-P.; Desprez, P.-Y.; Krtolica, A.; Campisi, J. The senescence-associated secretory phenotype: The dark side of tumor suppression. Annu. Rev. Pathol 2010, 5, 99–118. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Dookun, E.; Passos, J.F.; Arthur, H.M.; Richardson, R.D. Therapeutic Potential of Senolytics in Cardiovascular Disease. Cardiovasc. Drugs Ther. 2022, 36, 187–196. [Google Scholar] [CrossRef] [PubMed]
  28. Richardson, G.D.; Laval, S.; Owens, W.A. Cardiomyocyte Regeneration in the mdx Mouse Model of Nonischemic Cardiomyopathy. Stem Cells Dev. 2015, 24, 1672–1679. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Lewis-McDougall, F.C.; Ruchaya, P.J.; Domenjo-Vila, E.; Teoh, T.S.; Prata, L.; Cottle, B.J.; Clark, J.E.; Punjabi, P.P.; Awad, W.; Torella, D.; et al. Aged-senescent cells contribute to impaired heart regeneration. Aging Cell 2019, 18, e12931. [Google Scholar] [CrossRef] [Green Version]
  30. Anderson, R.; Lagnado, A.; Maggiorani, D.; Walaszczyk, A.; Dookun, E.; Chapman, J.; Birch, J.; Salmonowicz, H.; Ogrodnik, M.; Jurk, D.; et al. Length-independent telomere damage drives post-mitotic cardiomyocyte senescence. EMBO J. 2019, 38, e100492. [Google Scholar] [CrossRef] [PubMed]
  31. Dookun, E.; Walaszczyk, A.; Redgrave, R.; Palmowski, P.; Tual-Chalot, S.; Suwana, A.; Chapman, J.; Jirkovsky, E.; Sosa, L.D.; Gill, E.; et al. Clearance of senescent cells during cardiac ischemia–reperfusion injury improves recovery. Aging Cell 2020, 19, e13249. [Google Scholar] [CrossRef] [PubMed]
  32. Czosseck, A.; Chen, M.M.; Nguyen, H.; Meeson, A.; Hsu, C.-C.; Chen, C.-C.; George, T.A.; Ruan, S.-C.; Cheng, Y.-Y.; Lin, P.-J.; et al. Porous scaffold for mesenchymal cell encapsulation and exosome-based therapy of is-chemic diseases. J. Control. Release 2022, 352, 879–892. [Google Scholar] [CrossRef] [PubMed]
  33. Knuth, C.A.; Clark, M.E.; Meeson, A.P.; Khan, S.K.; Dowen, D.J.; Deehan, D.J.; Oldershaw, R.A. Low oxygen tension is critical for the culture of human mesenchymal stem cells with strong osteogenic potential from haemarthrosis fluid. Stem Cell Rev. Rep. 2013, 9, 599–608. [Google Scholar] [CrossRef] [PubMed]
  34. Rampersad, S.N. Multiple applications of alamar blue as an indicator of metabolic function and cellular health in cell viability bioassays. Sensors 2012, 12, 12347–12360. [Google Scholar] [CrossRef] [PubMed]
  35. Präbst, K.; Engelhardt, H.; Ringgeler, S.; Hübner, H. Basic colorimetric proliferation assays: MTT, WST, and resazurin. Methods Mol. Biol. 2017, 1601, 1–17. [Google Scholar]
  36. 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]
  37. Baker, D.; Childs, B.; Durik, M.; Wilers, M.E.; Sieben, C.J.; Zhong, J.; Saltness, R.A.; Jeganathan, K.B.; Casaclang Verzosa, G.; Pezeshki, A.; et al. Naturally occurring p16Ink4a-positive cells shorten healthy lifespan. Nature 2016, 530, 184–189. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Chou, L.-Y.; Ho, C.-T.; Hung, S.-C. Paracrine Senescence of Mesenchymal Stromal Cells Involves Inflammatory Cytokines and the NF-κB Pathway. Cells 2022, 11, 3324. [Google Scholar] [CrossRef]
  39. Walaszczyk, A.; Dookun, E.; Redgrave, R.; Tual-Chalot, S.; Victorelli, S.; Spyridopoulos, I.; Owens, A.; Arthur, H.M.; Passos, J.F.; Richardson, G.D. Pharmacological clearance of senescent cells improves survival and recovery in aged mice following acute myocardial infarction. Aging Cell 2019, 18, e12945. [Google Scholar] [CrossRef] [PubMed]
  40. Tuttle, C.S.L.; Luesken, S.W.M.; Waaijer, M.E.C.; Maier, A.B. Senescence in tissue samples of humans with age-related diseases: A systematic review. Ageing Res. Rev. 2021, 68, 101334. [Google Scholar] [CrossRef] [PubMed]
  41. Mauney, J.R.; Kaplan, D.L.; Volloch, V. Matrix-mediated retention of osteogenic differentiation potential by human adult bone marrow stromal cells during ex vivo expansion. Biomaterials 2004, 25, 3233–3243. [Google Scholar] [CrossRef] [PubMed]
  42. Dominici, M.; Le Blanc, K.; Mueller, I.; Slaper-Cortenbach, I.; Marini, F.; Krause, D.; Deans, R.; Keating, A.; Prockop, D.; Horwitz, E. 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]
  43. Sethe, S.; Scutt, A.; Stolzing, A. Aging of Mesenchymal Stem Cells. Aging Res. Rev. 2006, 5, 91–116. [Google Scholar] [CrossRef] [PubMed]
  44. Liu, L.; DiGirolamo, C.M.; Navarro, P.A.A.S.; Blasco, M.A.; Keefe, D.L. Telomerase deficiency impairs differentiation of mesenchymal stem cells. Exp. Cell Res. 2004, 294, 1–8. [Google Scholar] [CrossRef]
  45. Simonsen, J.L.; Rosada, C.; Serakinci, N.; Justesen, J.; Stenderup, K.; Rattan, S.I.; Jensen, T.G.; Kassem, M. Telomerase expression extends the proliferative lifespan and maintains the osteogenic potential of human bone marrow stromal cells. Nat. Biotechnol. 2002, 20, 592–596. [Google Scholar] [CrossRef]
  46. Hiyama, E.; Hiyama, K. Telomere and telomerase in stem cells. Br. J. Cancer 2007, 96, 1020–1024. [Google Scholar] [CrossRef] [Green Version]
  47. Go, M.J.; Takenaka, C.; Ohgushi, H. Forced expression of Sox2 or Nanog in human bone marrow derived mesenchymal stem cells maintains their expansion and differentiation capabilities. Exp. Cell Res. 2008, 314, 1147–1154. [Google Scholar] [CrossRef] [PubMed]
  48. Han, J.; Mistriotis, P.; Lei, P.; Wang, D.; Liu, S.; Andreadis, S.T. Nanog reverses the effects of organismal aging on mesenchymal stem cell proliferation and myogenic differentiation potential. Stem Cells 2012, 12, 2746–2759. [Google Scholar] [CrossRef] [Green Version]
  49. Alvarez, A.; Hossain, M.; Dantuma, E.; Merchant, S.; Sugaya, K. Nanog overexpression allows human mesenchymal stem cells to differentiate into neural cells. Neurosci. Med. 2010, 1, 1–13. [Google Scholar] [CrossRef] [Green Version]
  50. Wada, T.; Joza, N.; Cheng, H.Y.; Sasaki, T.; Kozieradzki, I.; Bachmaier, K.; Katada, T.; Schreiber, M.; Wagner, E.F.; Nishina, H.; et al. MKK7 couples stress signalling to G2/M cell-cycle progression and cellular senescence. Nat. Cell Biol. 2004, 6, 215–226. [Google Scholar] [CrossRef] [PubMed]
  51. Shvedova, M.; Samdavid Thanapaul, R.J.R.; Thompson, E.L.; Niedernhofer, L.J.; Roh, D.S. Cellular Senescence in Aging, Tissue Repair, and Regeneration. Plast Reconstr. Surg. 2022, 150, 4S–11S. [Google Scholar] [CrossRef] [PubMed]
  52. Silva, A.D.; Piccinato, C.A.; Sardinha, L.R.; Aloia, T.P.A.; Goldberg, A.C. Comparison of senescence progression in mesenchymal cells from human umbilical cord walls measured by immunofluorescence and flow cytometry of p16 and p21. Einstein 2020, 18, eAO5236. [Google Scholar] [CrossRef] [PubMed]
  53. Casella, G.; Munk, R.; Kim, K.M.; Piao, Y.; Supriyo, S.; Abdelmohsen, K.; Gorospe, M. Transcriptome signature of cellular senescence. Nucleic Acids Res. 2019, 47, 11476. [Google Scholar] [CrossRef] [PubMed]
  54. Lu, A.T.; Quach, A.; Wilson, J.G.; Reiner, A.P.; Aviv, A.; Raj, K.; Hou, L.; Baccarelli, A.A.; Li, Y.; Stewart, J.D.; et al. DNA methylation GrimAge strongly predicts lifespan and healthspan. Aging 2019, 11, 303–327. [Google Scholar] [CrossRef] [PubMed]
  55. Levine, M.E.; Lu, A.T.; Quach, A.; Chen, B.H.; Assimes, T.L.; Bandinelli, S.; Hou, L.; Baccarelli, A.A.; Stewart, J.D.; Li, Y.; et al. An epigenetic biomarker of aging for lifespan and healthspan. Aging 2018, 10, 573–591. [Google Scholar] [CrossRef] [Green Version]
  56. Lind, L.; Ingelsson, E.; Sundström, J.; Siegbahn, A.; Lampa, E. Methylation-based estimated biological age and cardiovascular disease. Eur. J. Clin. Investig. 2018, 48, e12872. [Google Scholar] [CrossRef]
  57. Niemann, J.; Johne, C.; Schroder, S.; Koch, F.; Ibrahim, S.M.; Schultz, J.; Tiedge, M.; Baltrusch, S. An mtDNA mutation accelerates liver aging by interfering with the ROS response and mitochondrial life cycle. Free Radic. Biol. Med. 2017, 102, 174–187. [Google Scholar]
  58. Di Micco, R.; Krizhanovsky, V.; Baker, D.; d’Adda di Fagagna, F. Cellular senescence in ageing: From mechanisms to therapeutic opportunities. Nat. Rev. Mol. Cell Biol. 2021, 22, 75–95. [Google Scholar] [CrossRef]
  59. Monsour, M.; Gorsky, A.; Nguyen, H.; Castelli, V.; Lee, J.Y.; Borlongan, C.V. Enhancing oxidative phosphorylation over glycolysis for energy production in cultured mesenchymal stem cells. Neuroreport 2022, 33, 635–640. [Google Scholar] [CrossRef] [PubMed]
  60. Subramani, B.; Subbannagounder, S.; Ramanathanpullai, C.; Palanivel, S.; Ramasamy, R. Impaired redox environment modulates cardiogenic and ion-channel gene expression in cardiac-resident and non-resident mesen-chymal stem cells. Exp. Biol. Med. 2017, 242, 645–656. [Google Scholar] [CrossRef] [Green Version]
  61. Yuan, X.; Liu, Y.; Bijonowski, B.M.; Tsai, A.C.; Fu, Q.; Logan, T.M.; Ma, T.; Li, Y. NAD+/NADH redox alterations reconfigure metabolism and rejuvenate senescent human mesenchymal stem cells in vitro. Commun. Biol. 2020, 3, 774. [Google Scholar] [CrossRef] [PubMed]
  62. Coope, A.; Ghanameh, Z.; Kingston, O.; Sheridan, C.M.; Barrett-Jolley, R.; Phelan, M.M.; Oldershaw, R.A. 1H NMR Metabolite Monitoring during the Differentiation of Human Induced Pluripotent Stem Cells Provides New Insights into the Molecular Events That Regulate Embryonic Chondrogenesis. Int. J. Mol. Sci. 2022, 23, 9266. [Google Scholar] [CrossRef] [PubMed]
  63. Zhu, Y.; Xian, X.; Wang, Z.; Bi, Y.; Chen, Q.; Han, X.; Tang, D.; Chen, R. Research Progress on the Relationship between Atherosclerosis and Inflammation. Biomolecules 2018, 8, 80. [Google Scholar] [CrossRef] [Green Version]
  64. Soto-Navarrete, M.T.; López-Unzu, M.Á.; Durán, A.C.; Fernández, B. Embryonic development of bicuspid aortic valves. Prog. Cardiovasc. Dis. 2020, 63, 407–418. [Google Scholar] [CrossRef] [PubMed]
  65. Wang, D.; Li, L.K.; Dai, T.; Wang, A.; Li, S. Adult Stem Cells in Vascular Remodeling. Theranostics 2018, 8, 815–829. [Google Scholar] [CrossRef] [Green Version]
  66. Willems, L.; Daniëls, A.; Fanton, Y.; Linsen, L.; Evens, L.; Bito, V.; Declercq, J.; Rummens, J.L.; Hensen, K.; Hendrikx, M. Differentiation of Human Cardiac Atrial Appendage Stem Cells into Adult Cardiomyocytes: A Role for the Wnt Pathway? Int. J. Mol. Sci. 2020, 21, 3931. [Google Scholar] [CrossRef] [PubMed]
  67. Owens, W.A.; Anna Walaszczyk, A.; Ioakim Spyridopoulos, I.; Dookun, E.; Richardson, G.D. Senescence and senolytics in cardiovascular disease: Promise and potential pitfalls. Mech. Ageing Dev. 2021, 198, 111540. [Google Scholar] [CrossRef]
  68. Balint, B.; Yin, H.; Nong, Z.; Arpino, J.-M.; O’Neil, C.; Rogers, S.R.; Randhawa, V.K.; Fox, S.A.; Chevalier, J.; Lee, J.J.; et al. Seno-destructive smooth muscle cells in the ascending aorta of patients with bicuspid aortic valve disease. eBioMedicine 2019, 43, 34–66. [Google Scholar] [CrossRef]
Figure 1. Derivation and characterization of CMSCLC from BAVD and CAD tissue. (A) Derivation of CMSCLC from BAVD and CAD cardiac tissue was quantified by counting the formation of established CFU-Fs. (B) Total number of CMSCLC counted from established CFU-Fs. (C) Quantification of cell doubling per day for BAVD-CMSCLC and CAD-CMSCLC over passages 3–5 of in vitro culture. (D,E) Brightfield images of BAVDMSCs and CADMSCs. Scale bar = 50 μm.
Figure 1. Derivation and characterization of CMSCLC from BAVD and CAD tissue. (A) Derivation of CMSCLC from BAVD and CAD cardiac tissue was quantified by counting the formation of established CFU-Fs. (B) Total number of CMSCLC counted from established CFU-Fs. (C) Quantification of cell doubling per day for BAVD-CMSCLC and CAD-CMSCLC over passages 3–5 of in vitro culture. (D,E) Brightfield images of BAVDMSCs and CADMSCs. Scale bar = 50 μm.
Biomedicines 10 03143 g001
Figure 2. Immunophenotyping of MSC-associated cell surface antigens. Results for CMSCLCs immunolabelled with antibodies to human CD cell surface antigens. BAVD-CMSCLC express a number of markers of MSCs (AG) but are negative for CD19 (H) and CD45 (I). CAD-CMSCLC also express the expected MSC markers (JP) but are also negative for CD19 (Q) and CD45 (R).
Figure 2. Immunophenotyping of MSC-associated cell surface antigens. Results for CMSCLCs immunolabelled with antibodies to human CD cell surface antigens. BAVD-CMSCLC express a number of markers of MSCs (AG) but are negative for CD19 (H) and CD45 (I). CAD-CMSCLC also express the expected MSC markers (JP) but are also negative for CD19 (Q) and CD45 (R).
Biomedicines 10 03143 g002
Figure 3. Immunolabelling of CMSCLC for NANOG and p16. (A,B) ICC analysis for NANOG expression in BAVD-CMSCLC and CAD-CMSCLC. NANOG (Green), all nuclei labelled with Dapi (Blue). (C,D) ICC analysis for p16 expression in BAVD-CMSCLC and CAD-CMSCLC. p16 (Green), all nuclei labelled with DAPI (Blue). Scale bar = 50μm.
Figure 3. Immunolabelling of CMSCLC for NANOG and p16. (A,B) ICC analysis for NANOG expression in BAVD-CMSCLC and CAD-CMSCLC. NANOG (Green), all nuclei labelled with Dapi (Blue). (C,D) ICC analysis for p16 expression in BAVD-CMSCLC and CAD-CMSCLC. p16 (Green), all nuclei labelled with DAPI (Blue). Scale bar = 50μm.
Biomedicines 10 03143 g003
Figure 4. Evaluation of CMSCLC senescent phenotype (A) Measurement of CMSCLC telomere length. (A) TeloTAGGG Telomere Length Assay was used to measure telomere length of BAVD-CMSCLC and CAD-CMSCLC. Resolved telomere fragments from Hinf I/Rsa I-digested control gDNA, CAD-CMSCLC gDNA and BAV-CMSCLC gDNA were hybridized with DIG-labelled telomere-specific probe, incubated with anti-DIG antibody conjugated to alkaline phosphatase and visualized by chemiluminescence. Average telomere fragment lengths were calibrated against a DIG-labelled molecular weight marker (MWM). (B) Measurement of metabolic activity. Measurement of metabolic activity as an indicator of cell health was performed on CMSCLC using an alamarBlue® assay, the fluorescence of the resulting culture media was normalized against total DNA content. * = p < 0.05 of n = 3 independent technical replicates, Mann–Whitney test. (C,D) Cell cycle analysis. Results of flow cytometry analysis for cell cycle of BAVD-CMSCLC, note the presence of a subpopulation of cells in the G2/M phase that is absent in the CAD-MSCs.
Figure 4. Evaluation of CMSCLC senescent phenotype (A) Measurement of CMSCLC telomere length. (A) TeloTAGGG Telomere Length Assay was used to measure telomere length of BAVD-CMSCLC and CAD-CMSCLC. Resolved telomere fragments from Hinf I/Rsa I-digested control gDNA, CAD-CMSCLC gDNA and BAV-CMSCLC gDNA were hybridized with DIG-labelled telomere-specific probe, incubated with anti-DIG antibody conjugated to alkaline phosphatase and visualized by chemiluminescence. Average telomere fragment lengths were calibrated against a DIG-labelled molecular weight marker (MWM). (B) Measurement of metabolic activity. Measurement of metabolic activity as an indicator of cell health was performed on CMSCLC using an alamarBlue® assay, the fluorescence of the resulting culture media was normalized against total DNA content. * = p < 0.05 of n = 3 independent technical replicates, Mann–Whitney test. (C,D) Cell cycle analysis. Results of flow cytometry analysis for cell cycle of BAVD-CMSCLC, note the presence of a subpopulation of cells in the G2/M phase that is absent in the CAD-MSCs.
Biomedicines 10 03143 g004
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Oldershaw, R.A.; Richardson, G.; Carling, P.; Owens, W.A.; Lundy, D.J.; Meeson, A. Cardiac Mesenchymal Stem Cell-like Cells Derived from a Young Patient with Bicuspid Aortic Valve Disease Have a Prematurely Aged Phenotype. Biomedicines 2022, 10, 3143. https://doi.org/10.3390/biomedicines10123143

AMA Style

Oldershaw RA, Richardson G, Carling P, Owens WA, Lundy DJ, Meeson A. Cardiac Mesenchymal Stem Cell-like Cells Derived from a Young Patient with Bicuspid Aortic Valve Disease Have a Prematurely Aged Phenotype. Biomedicines. 2022; 10(12):3143. https://doi.org/10.3390/biomedicines10123143

Chicago/Turabian Style

Oldershaw, Rachel A., Gavin Richardson, Phillippa Carling, W. Andrew Owens, David J. Lundy, and Annette Meeson. 2022. "Cardiac Mesenchymal Stem Cell-like Cells Derived from a Young Patient with Bicuspid Aortic Valve Disease Have a Prematurely Aged Phenotype" Biomedicines 10, no. 12: 3143. https://doi.org/10.3390/biomedicines10123143

APA Style

Oldershaw, R. A., Richardson, G., Carling, P., Owens, W. A., Lundy, D. J., & Meeson, A. (2022). Cardiac Mesenchymal Stem Cell-like Cells Derived from a Young Patient with Bicuspid Aortic Valve Disease Have a Prematurely Aged Phenotype. Biomedicines, 10(12), 3143. https://doi.org/10.3390/biomedicines10123143

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