Expand this Topic clickable element to expand a topic
Skip to content
Optica Publishing Group

Label-free assessment of replicative senescence in mesenchymal stem cells by Raman microspectroscopy

Open Access Open Access

Abstract

Here, Raman microspectroscopy was employed to assess replicative senescence of mesenchymal stem cells (MSC). A regular spectral change related to the cell senescence was found in the ratio of two peaks at 1157 cm−1 and 1174 cm−1, which are assigned to C-C, C-N stretching vibrations in proteins and C-H bending vibrations in tyrosine and phenylalanine, respectively. With the cell aging, the ratio I1157 / I1174 exhibited a monotonic decline and showed small standard deviations, so that it can statistically distinguish between cells having slight changes in terms of aging. We propose that I1157 / I1174 can act as a characteristic spectral signature for label-free assessment of MSC senescence.

© 2015 Optical Society of America

1. Introduction

Mesenchymal stem cells (MSCs), which are present in various tissues such as bone marrow, adipose, amniotic membrane, etc., have been proposed to be a promising candidate for regenerative medicine [1] because of their multi-differentiation potential, hematopoietic-supporting activity, immunomodulatory potency and self-renewal. Umbilical cord is an important source of MSCs [2–5 ]. Compared with those derived from bone marrow, MSCs from human umbilical cord (hUC-MSCs) can effectively avoid the reduction in cell number, proliferation and differentiation capacity with age or age-related disease. Therefore, hUC-MSCs are considered to be an ideal source for cell-based therapy.

Generally, cell subculture in vitro is essential to provide a sufficient number of MSCs for a successful clinical application [6]. However, this process is inescapably accompanied by replicative senescence, which has been found to inhibit cell proliferation [6], impair cell differentiation [7], and further abrogate cells’ therapeutic potential [8]. Thus, to assess the cell senescence is of great necessity for their therapeutic use.

Empirically, one can roughly estimate cell senescence according to the times of cell passage. However, due to individual differences in cells, there is no strict correspondence between the times of cell passage and the level of cell aging, and thus easy to cause a large error. In addition, cell aging is accompanied with morphological changes such as enlarged cell size and flattened morphology [9–11 ]. Nevertheless, morphological methods are susceptible to interference of individual judgment, and also difficult to quantitatively determine the cell senescence. Furthermore, molecular changes associated with cell senescence have been widely studied, and several meaningful biomolecular markers were revealed. For instance, β-galactosidase has been widely accepted as an effective indicator for cell aging detection [9–12 ]. The expression of the proteins p16, p21, p53, etc. were also found closely related to MSC senescence [12–14 ]. Therefore, corresponding to the molecular markers, biochemical methods including SA-β-gal Staining, Real Time PCR, Western Blotting, etc. can offer relatively accurate results and make quantitative analysis. However, most of these methods need labeling or complicated operations, which may not be conducive to detection of a large number of cell samples. Hence, development of a label-free optical method for cell senescence assessment is necessary for MSCs’ quality control and their applications.

In this regard, Raman spectroscopy may be a promising candidate to tackle the task. This technology is based on inelastic light scattering, in which the frequency of the scattered light is changed as compared to that of the incident light, and their frequency differences are dependent on the molecular vibrations. Therefore, Raman spectroscopy is able to illustrate the intrinsic molecular structures [15] and make quantitative analysis of molecular changes in biological samples [16]. Owing to the excellent features of label-free and “molecular fingerprint”, this technology has been increasingly employed in stem cell studies, such as discrimination of undifferentiated and differentiated embryonic stem cells (ESCs) [17–19 ], distinguishing between MSCs and fibroblasts [20], identification of abnormal stem cells [21], in situ monitoring of the MSCs’ osteogenic differentiation [22,23 ] and the cardiac differentiation of ESCs [24], determination of the cell cycle phase in ESCs [25], as well as rapid cell sorting of cardiomyocytes derived from ESCs [26]. In our previous works, the Raman spectral markers specific to viability of hUC-MSCs and their correlations with reactive oxygen species (ROS) were also revealed [27,28 ]. These encouraging findings showed a tremendous application potential of Raman spectroscopy in living cell analysis, and laid the foundation for its further use in stem cell researches.

In this study, we focused on the Raman spectral variations of hUC-MSCs during their aging process, aimed at looking for the specific Raman spectral features that can be linked to the cell replicative senescence. The findings may be helpful to provide new evidences in terms of molecular structures for the cell aging studies, and also contribute to development of a label-free method for assessment of MSC senescence.

2. Materials and methods

2.1 Isolation and culture of hUC-MSCs

We isolated hUC-MSCs from human umbilical cord tissues by the method previously described [4]. hUC-MSCs were cultured in monolayer at 37 °C and 5% CO2 in DMEM with 10% FCS. Cells were harvested after reaching 90% confluence using trypsin and passaged (subplated) at a 1:3 ratio until passage (P) 30. We used cells in passage 5, 10, 20, 30 respectively for experiments to obtain samples at different aging levels.

2.2 Raman microspectroscopy

A Raman microspectrometer (LRS-5, GANGDONG, China) was used to record the Raman scattering from the center of single hUC-MSCs by means of the microscopic imaging. Briefly, excitation light at 532 nm was provided by a Nd:YAG laser with the output power of 8 mW. A 50 × objective was used to focus the excitation light and to collect the Raman signal. For each spectral scan, Raman spectrum was recorded in the range of 600–1800 cm−1, and the signal was integrated for 50 s. For each cell sample, 6 ~7 cells were selected randomly for Raman spectroscopy measurements.

2.3 Spectral processing and statistical analysis

The background signal was subtracted from each spectrum, and a multipoint baseline was corrected in the whole spectral region. Spectra were then normalized to the peak intensity at 1449 cm−1 that was widely accepted as a normalization standard of Raman spectra from cells [18,29 ]. The statistical analyses were performed by the Student’s t-test.

3. Results

In order to find the spectral differences related to MSC senescence, we first compared the Raman spectra of cells in passage 5 (P5) with those in P30.

As is shown in Fig. 1 , obvious spectral differences can be found in the regions of 905 - 1016, 1087 - 1190, and 1510 - 1530 cm−1. These spectral regions include Raman peaks at 934, 980, 1003, 1097, 1127, 1157, 1174, and 1521 cm−1, which are mainly corresponding to vibrations in proteins, lipids and DNA [18,27,29,30 ]. The assignments of the Raman peaks are listed in Table 1 .

 figure: Fig. 1

Fig. 1 Average Raman spectra from senescent hUC-MSCs in P30 (a) and from young cells in P5 (b) and their difference spectrum (c = a-b). Spectra are shifted vertically for clarity. Gray bars highlight the spectral differences.

Download Full Size | PDF

Tables Icon

Table 1. Raman peak assignment

The spectral differences showed above were observed from average spectra between young and aging cell-groups. However, individual differences among cells in the same group were not taken into account. If individual differences are too large, the corresponding differences from average spectra will be meaningless. Thus, each Raman peak listed above needs to be further validated by statistical analysis. Here, Student’s t-test is employed and its result is described by the p value. As a rule of thumb, p<0.05 represents the difference is of statistical significance.

The peak intensity comparisons between young (P5) and aging (P30) hUC-MSCs are illustrated in Fig. 2 . It can be seen that the average intensities of Raman peaks at 1003, 1127, 1174 cm−1 did not show obvious differences between the two cell-groups and thus cannot be used as spectral markers. As for the peaks at 934, 980, 1097 and 1521 cm−1, although their average intensities exhibited visible differences, these differences were not statistically significant (p>0.05) because of the relative large standard deviations. In contrast, the Raman peak at 1157 cm−1, corresponding to C-C, C-N stretching vibrations in proteins, showed significant difference in peak intensity as indicated by p<0.05. In comparison of the case of young cells, the peak intensity of 1157 cm−1 reduced by about 60% in the spectra of aging cells, indicating this peak is sensitive to replicative senescence of hUC-MSCs.

 figure: Fig. 2

Fig. 2 Comparison of relative intensities of Raman peaks between young (P5) and aging (P30) hUC-MSCs. * means p<0.05 obtained by Student’s t-test.

Download Full Size | PDF

However, the results above are not enough to demonstrate the Raman peak at 1157 cm−1 can be used as a spectral marker for the cell aging assessment. It is noted that a spectral marker should not only show a significant difference between young and aging cells, but also exhibit a monotonic change with cell senescence. Thus, we further analyzed Raman spectra from hUC-MSCs in P5, P10, P20 and P30, corresponding to increasingly serious aging states. As is shown in Fig. 3 , average spectra of the cells in P5, P10, P20, P30 are visually compared and the Raman peak at 1157 cm−1 is highlighted by a gray bar.

 figure: Fig. 3

Fig. 3 Average Raman spectra from hUC-MSCs in P5 (a), P10 (b), P20 (c) and P30 (d). Spectra are shifted vertically for clarity. The gray bar highlights the spectral changes with cell senescence.

Download Full Size | PDF

Figure 4(A) shows the change of peak intensity at 1157 cm−1 with the cell senescence. It can be seen that the average peak intensity underwent a monotonic decline as the cell passage increased. However, this decline showed significant nonlinearity i.e. sharp decline could be observed from P5 to P10, while only a slight decrease occurred from P10 to P30. This is not conducive to the characterization of the cell aging. More disadvantageously, for each cell group, the peak intensity exhibited so large standard deviation that the p values obtained from Student’s t-test between cells in P5 and P10, P10 and P20, P20 and P30 were all larger than 0.05. Statistically, it means the peak intensity at 1157 cm−1 does not have the ability to detect slight changes in the cell aging state.

 figure: Fig. 4

Fig. 4 (A) Change of I1157 with cell senescence. (B) Change of I1157 / I1174 with cell senescence. Values p1, p2, p3 were obtained by the Student’s t-test analysis between cells in P5 and P10, P10 and P20, P20 and P30 respectively.

Download Full Size | PDF

Since no single Raman peak can be an effective indicator for the cell aging assessment, multi-peak complex should be considered. In this respect, we try to find a composite indicator by calculation of two or more peak intensities. As is shown in Fig. 3, a monotonic change in the ratio of peak intensities at 1157 cm−1 and 1174 cm−1 can be clearly observed and highlighted by the gray bar. Further, the change of ratio I1157 / I1174 with the cell aging and the results of Student’s t-test are shown in Fig. 4(B). Compared with the case of I1157 (Fig. 4(A)), the ratio I1157 / I1174 showed a more linear decline with the cell passage increase, especially from P10 to P30. More importantly, the standard deviations of I1157 / I1174 in the cell groups P5 and P10 were much smaller, indicating the ratio is more stable, and less affected by individual differences of the cells. Statistically, the p values by Student’s t-test of I1157 / I1174 between cells in P5 and P10, P10 and P20, P20 and P30 were all less than 0.05, suggesting I1157 / I1174 is potential to detect slight changes in the cell aging state. Besides, It is necessary to mention that this result can be verified by other cell lines of hUC-MSCs derived from different donors. In those cases, it can also be observed that I1157 / I1174 of young cells was significantly higher than that of aging cells (data not shown). Based on these findings, we suggest that I1157 / I1174 can be an effective spectral marker for assessment of MSC senescence.

4. Discussion

Similar to any normal somatic cell, MSCs undergo replicative senescence and have a limited lifespan in vitro [6,9 ]. It has been reported that replicative senescence of MSCs is a continuous process, during which numerous changes in phenotype, differentiation potential, global gene expression patterns, and miRNA profiles occur [31]. These far-reaching alterations will inevitably affect the cell therapy and other MSC applications. Thus, cell senescence is an important factor that needs to be considered and evaluated in MSC preparation, especially through a label-free manner. In this work, we found a Raman spectral feature (I1157 / I1174) that showed high sensitivity and effectiveness for MSC aging assessment, indicating Raman spectroscopy can be an ideal choice. Moreover, owing to the “molecular fingerprint” feature, Raman spectroscopy also showed a great potential in monitoring MSC viability [27] and differentiation [23]. These encouraging findings make it hopeful to further develop a label-free method for monitoring multiple physiological parameters of MSCs.

In addition, the spectral alterations may provide new evidences for understanding of replicative senescence in MSCs. The results showed that significant spectral difference between young and aging cells can be found at 1157 cm−1 (Fig. 2), which is assigned to C-C, C-N stretching vibration in peptide bonds and sensitive to the conformational changes of proteins [30]. With the cell aging, the peak intensity of 1157 cm−1 exhibited a significant decline (Fig. 4), while no alterations in peak position and full width at half maximum (FWHM) were observed, indicating a damage of the peptide bonds. The changes in protein structures were also indicated by the Raman peak at 934 cm−1 that is assigned to C-C backbone stretching vibrations in proteins. As the cell passage increased, an obvious expansion in the peak FWHM of 934 cm−1 can be observed (Fig. 3), indicating a change in the protein backbones. These results suggest that protein cleavage may occur in the cell aging process.

Besides, the change of peak at 1174 cm−1 should also be discussed. Although there is no statistically significant difference in this peak between young and aging cells can be found (Fig. 2), it is noted that the standard deviation of the peak intensity ratio I1157 / I1174 was much smaller than that of single peak at 1157 cm−1, and the ratio showed a much better performance in distinguishing cells at different aging states (Fig. 4). In other words, the ratio I1157 / I1174 largely reduced the fluctuations of I1157 caused by individual differences and showed a greater ability for assessing the cell senescence. It indicates that for a single cell, the rise of I1174 occures concomitantly with the decrease of I1157 during the cell aging process. The Raman peak at 1174 cm−1 is corresponding to C-H bending vibrations in tyrosine and phenylalanine [18], which are cellular endogenous fluorescent substances. Thus, the rise of I1174 with the cell senescence was further confirmed by autofluorescence measurement. As is shown in Fig. 5 , the fluorescence of aromatic amino acids (emission peak at 336nm, excitation wavelength at 280nm) in aging cells (P20) was higher than that of young cells (P5), and their statistically significant difference was observed (p<0.05).

 figure: Fig. 5

Fig. 5 (A) Average autofluorescence spectra from aging hUC-MSCs (P20) and young cells (P5). (B) Comparison of the fluorescent peak height at 336nm. Excitation wavelength at 280nm. p value was obtained by Student’s t-test.

Download Full Size | PDF

Taken together, we infer that the aging of hUC-MSCs is accompanied by cleavage of proteins and polypeptides, and thus free amino acid residues will increase in the senescent cells. In this case, more amino acid residues are exposed to irradiation, causing an enhancement of their Raman scattering and fluorescence intensity. These molecular changes may be closely related to oxidation reactions and oxidation end products, which have been proven to play an important role in MSC senescence [32].

Finally, it is necessary to mention that the sample size is relatively small, due to the limited cell lines and the long-term cell passage. The small sample size may introduce somewhat influences on the results caused by systematic drifts and baseline corrections, etc., which are almost unavoidable in such weak signal measurements and potentially lead to minor deviations in the results. Future work based on an expanded sample size is needed to further explore quantitative correspondence between the Raman spectral feature and MSC senescence.

5. Conclusions

In this study, we demonstrated that the C-C, C-N stretching vibrations in proteins at 1157 cm−1 and C-H bending vibrations in tyrosine and phenylalanine at 1174 cm−1 can be associated with the replicative senescence of hUC-MSCs, and their intensity ratio I1157 / I1174 can act as a characteristic spectral signature for label-free assessment of the cell senescence. The decrease of I1157 / I1174 with the cell aging could be related to the cleavage of proteins and increase of free amino acid residues in the senescent cells.

Acknowledgments

We would like to thank Tianjin Gangdong Sci. & Tech. Development Co., Ltd. for assistance in Raman spectral measurements. This work was supported by Tianjin Municipal Science and Technology Commission (14JCQNJC01800 and 08ZCDFGX09400), National Natural Science Fundation of China (61201106, 31470951, 81172837 and 61307094) and Tianjin Special Scientific Grant (14JCPJC00529).

References and links

1. H. Motaln, C. Schichor, and T. T. Lah, “Human mesenchymal stem cells and their use in cell-based therapies,” Cancer 116(11), 2519–2530 (2010). [CrossRef]   [PubMed]  

2. K. E. Mitchell, M. L. Weiss, B. M. Mitchell, P. Martin, D. Davis, L. Morales, B. Helwig, M. Beerenstrauch, K. Abou-Easa, T. Hildreth, D. Troyer, and S. Medicetty, “Matrix cells from Wharton’s jelly form neurons and glia,” Stem Cells 21(1), 50–60 (2003). [CrossRef]   [PubMed]  

3. H. S. Wang, S. C. Hung, S. T. Peng, C. C. Huang, H. M. Wei, Y. J. Guo, Y. S. Fu, M. C. Lai, and C. C. Chen, “Mesenchymal stem cells in the Wharton’s jelly of the human umbilical cord,” Stem Cells 22(7), 1330–1337 (2004). [CrossRef]   [PubMed]  

4. W. Gong, Z. Han, H. Zhao, Y. Wang, J. Wang, J. Zhong, B. Wang, S. Wang, Y. Wang, L. Sun, and Z. Han, “Banking human umbilical cord-derived mesenchymal stromal cells for clinical use,” Cell Transplant. 21(1), 207–216 (2012). [CrossRef]   [PubMed]  

5. D. Baksh, R. Yao, and R. S. Tuan, “Comparison of proliferative and multilineage differentiation potential of human mesenchymal stem cells derived from umbilical cord and bone marrow,” Stem Cells 25(6), 1384–1392 (2007). [CrossRef]   [PubMed]  

6. H. B. Noh, H. J. Ahn, W. J. Lee, K. B. Kwack, and Y. D. Kwon, “The molecular signature of in vitro senescence in human mesenchymal stem cells,” Genes Genomics 32(1), 87–93 (2010). [CrossRef]  

7. M. C. Mitterberger, S. Lechner, M. Mattesich, and W. Zwerschke, “Adipogenic differentiation is impaired in replicative senescent human subcutaneous adipose-derived stromal/progenitor cells,” J. Gerontol. A Biol. Sci. Med. Sci. 69(1), 13–24 (2014). [CrossRef]   [PubMed]  

8. J. C. Sepúlveda, M. Tomé, M. E. Fernández, M. Delgado, J. Campisi, A. Bernad, and M. A. González, “Cell senescence abrogates the therapeutic potential of human mesenchymal stem cells in the lethal endotoxemia model,” Stem Cells 32(7), 1865–1877 (2014). [CrossRef]   [PubMed]  

9. H. Cheng, L. Qiu, J. Ma, H. Zhang, M. Cheng, W. Li, X. Zhao, and K. Liu, “Replicative senescence of human bone marrow and umbilical cord derived mesenchymal stem cells and their differentiation to adipocytes and osteoblasts,” Mol. Biol. Rep. 38(8), 5161–5168 (2011). [CrossRef]   [PubMed]  

10. H. Hao, G. Chen, J. Liu, D. Ti, Y. Zhao, S. Xu, X. Fu, and W. Han, “Culturing on Wharton’s jelly extract delays mesenchymal stem cell senescence through p53 and p16INK4a/pRb pathways,” PLoS One 8(3), e58314 (2013). [CrossRef]   [PubMed]  

11. J. Ren, D. F. Stroncek, Y. Zhao, P. Jin, L. Castiello, S. Civini, H. Wang, J. Feng, K. Tran, S. A. Kuznetsov, P. G. Robey, and M. Sabatino, “Intra-subject variability in human bone marrow stromal cell (BMSC) replicative senescence: molecular changes associated with BMSC senescence,” Stem Cell Res. (Amst.) 11(3), 1060–1073 (2013). [CrossRef]   [PubMed]  

12. J. C. Estrada, Y. Torres, A. Benguría, A. Dopazo, E. Roche, L. Carrera-Quintanar, R. A. Pérez, J. A. Enríquez, R. Torres, J. C. Ramírez, E. Samper, and A. Bernad, “Human mesenchymal stem cell-replicative senescence and oxidative stress are closely linked to aneuploidy,” Cell Death Dis. 4(6), e691 (2013). [CrossRef]   [PubMed]  

13. J. S. Park, H. Y. Kim, H. W. Kim, G. N. Chae, H. T. Oh, J. Y. Park, H. Shim, M. Seo, E. Y. Shin, E. G. Kim, S. C. Park, and S. J. Kwak, “Increased caveolin-1, a cause for the declined adipogenic potential of senescent human mesenchymal stem cells,” Mech. Ageing Dev. 126(5), 551–559 (2005). [CrossRef]   [PubMed]  

14. M. Terai, T. Uyama, T. Sugiki, X. K. Li, A. Umezawa, and T. Kiyono, “Immortalization of human fetal cells: the life span of umbilical cord blood-derived cells can be prolonged without manipulating p16INK4a/RB braking pathway,” Mol. Biol. Cell 16(3), 1491–1499 (2005). [CrossRef]   [PubMed]  

15. A. R. Boyd, G. A. Burke, and B. J. Meenan, “Monitoring cellular behaviour using Raman spectroscopy for tissue engineering and regenerative medicine applications,” J. Mater. Sci. Mater. Med. 21(8), 2317–2324 (2010). [CrossRef]   [PubMed]  

16. S. O. Konorov, H. G. Schulze, C. G. Atkins, J. M. Piret, S. A. Aparicio, R. F. B. Turner, and M. W. Blades, “Absolute quantification of intracellular glycogen content in human embryonic stem cells with Raman microspectroscopy,” Anal. Chem. 83(16), 6254–6258 (2011). [CrossRef]   [PubMed]  

17. I. Notingher, I. Bisson, J. M. Polak, and L. L. Hench, “In situ spectroscopic study of nucleic acids in differentiating embryonic stem cells,” Vib. Spectrosc. 35(1-2), 199–203 (2004). [CrossRef]  

18. J. W. Chan, D. K. Lieu, T. Huser, and R. A. Li, “Label-free separation of human embryonic stem cells and their cardiac derivatives using Raman spectroscopy,” Anal. Chem. 81(4), 1324–1331 (2009). [CrossRef]   [PubMed]  

19. H. G. Schulze, S. O. Konorov, N. J. Caron, J. M. Piret, M. W. Blades, and R. F. B. Turner, “Assessing differentiation status of human embryonic stem cells noninvasively using Raman microspectroscopy,” Anal. Chem. 82(12), 5020–5027 (2010). [CrossRef]   [PubMed]  

20. M. Pudlas, D. A. C. Berrio, M. Votteler, S. Koch, S. Thude, H. Walles, and K. Schenke-Layland, “Non-contact discrimination of human bone marrow-derived mesenchymal stem cells and fibroblasts using Raman spectroscopy,” Med. Laser Appl. 26(3), 119–125 (2011). [CrossRef]  

21. L. Harkness, S. M. Novikov, J. Beermann, S. I. Bozhevolnyi, and M. Kassem, “Identification of abnormal stem cells using Raman spectroscopy,” Stem Cells Dev. 21(12), 2152–2159 (2012). [CrossRef]   [PubMed]  

22. H. K. Chiang, F. Y. Peng, S. C. Hung, and Y. C. Feng, “In situ Raman spectroscopic monitoring of hydroxyapatite as human mesenchymal stem cells differentiate into osteoblasts,” J. Raman Spectrosc. 40(5), 546–549 (2009). [CrossRef]  

23. L. L. McManus, G. A. Burke, M. M. McCafferty, P. O’Hare, M. Modreanu, A. R. Boyd, and B. J. Meenan, “Raman spectroscopic monitoring of the osteogenic differentiation of human mesenchymal stem cells,” Analyst (Lond.) 136(12), 2471–2481 (2011). [CrossRef]   [PubMed]  

24. F. C. Pascut, S. Kalra, V. George, N. Welch, C. Denning, and I. Notingher, “Non-invasive label-free monitoring the cardiac differentiation of human embryonic stem cells in-vitro by Raman spectroscopy,” Biochim. Biophys. Acta 1830(6), 3517–3524 (2013). [CrossRef]   [PubMed]  

25. S. O. Konorov, H. G. Schulze, J. M. Piret, M. W. Blades, and R. F. Turner, “Label-free determination of the cell cycle phase in human embryonic stem cells by Raman microspectroscopy,” Anal. Chem. 85(19), 8996–9002 (2013). [CrossRef]   [PubMed]  

26. F. C. Pascut, H. T. Goh, V. George, C. Denning, and I. Notingher, “Toward label-free Raman-activated cell sorting of cardiomyocytes derived from human embryonic stem cells,” J. Biomed. Opt. 16(4), 045002 (2011). [CrossRef]   [PubMed]  

27. H. Bai, P. Chen, H. Fang, L. Lin, G. Q. Tang, G. G. Mu, W. Gong, Z. P. Liu, H. Wu, H. Zhao, and Z. C. Han, “Detecting viability transitions of umbilical cord mesenchymal stem cells by Raman micro-spectroscopy,” Laser Phys. Lett. 8(1), 78–84 (2011). [CrossRef]  

28. H. Bai, P. Chen, G. Q. Tang, L. Lin, H. Fang, G. G. Mu, W. Gong, Z. P. Liu, Z. B. Han, H. Zhao, and Z. C. Han, “Relations between reactive oxygen species and Raman spectral variations of human umbilical cord mesenchymal stem cells with different viability,” Laser Phys. 21(6), 1122–1129 (2011). [CrossRef]  

29. I. Notingher, S. Verrier, S. Haque, J. M. Polak, and L. L. Hench, “Spectroscopic study of human lung epithelial cells (A549) in culture: living cells versus dead cells,” Biopolymers 72(4), 230–240 (2003). [CrossRef]   [PubMed]  

30. Y. M. Xu, Raman spectroscopy in application of structure biology (Chemical Industry Press, 2005).

31. W. Wagner, P. Horn, M. Castoldi, A. Diehlmann, S. Bork, R. Saffrich, V. Benes, J. Blake, S. Pfister, V. Eckstein, and A. D. Ho, “Replicative senescence of mesenchymal stem cells: a continuous and organized process,” PLoS One 3(5), e2213 (2008). [CrossRef]   [PubMed]  

32. S. Sethe, A. Scutt, and A. Stolzing, “Aging of mesenchymal stem cells,” Ageing Res. Rev. 5(1), 91–116 (2006). [CrossRef]   [PubMed]  

Cited By

Optica participates in Crossref's Cited-By Linking service. Citing articles from Optica Publishing Group journals and other participating publishers are listed here.

Alert me when this article is cited.


Figures (5)

Fig. 1
Fig. 1 Average Raman spectra from senescent hUC-MSCs in P30 (a) and from young cells in P5 (b) and their difference spectrum (c = a-b). Spectra are shifted vertically for clarity. Gray bars highlight the spectral differences.
Fig. 2
Fig. 2 Comparison of relative intensities of Raman peaks between young (P5) and aging (P30) hUC-MSCs. * means p<0.05 obtained by Student’s t-test.
Fig. 3
Fig. 3 Average Raman spectra from hUC-MSCs in P5 (a), P10 (b), P20 (c) and P30 (d). Spectra are shifted vertically for clarity. The gray bar highlights the spectral changes with cell senescence.
Fig. 4
Fig. 4 (A) Change of I1157 with cell senescence. (B) Change of I1157 / I1174 with cell senescence. Values p1, p2, p3 were obtained by the Student’s t-test analysis between cells in P5 and P10, P10 and P20, P20 and P30 respectively.
Fig. 5
Fig. 5 (A) Average autofluorescence spectra from aging hUC-MSCs (P20) and young cells (P5). (B) Comparison of the fluorescent peak height at 336nm. Excitation wavelength at 280nm. p value was obtained by Student’s t-test.

Tables (1)

Tables Icon

Table 1 Raman peak assignment

Select as filters


Select Topics Cancel
© Copyright 2024 | Optica Publishing Group. All rights reserved, including rights for text and data mining and training of artificial technologies or similar technologies.