Journal Search Engine
Search Advanced Search Adode Reader(link)
Download PDF Export Citaion korean bibliography PMC previewer
ISSN : 1226-7155(Print)
ISSN : 2287-6618(Online)
International Journal of Oral Biology Vol.41 No.2 pp.69-74
DOI : https://doi.org/10.11620/IJOB.2016.41.2.069

Stemness and Proliferation of Murine Skin-Derived Precursor Cells under Hypoxic Environment

Hyewon Kim, Sangkyu Park, Sangho Roh*
Cellular Reprogramming and Embryo Biotechnology Lab, BK21 and Dental Research Institute, Seoul National University School of Dentistry, Seoul, Korea
Correspondence to: Sangho Roh, Seoul National University School of Dentistry, Gwanak-ro, Gwanak-gu, Seoul, Korea 82-2-880-2333, 82-2-882-0228sangho@snu.ac.kr
May 18, 2016 June 4, 2016 June 5, 2016

Abstract

Skin-derived precursors (SKPs) have potential to differentiate to various cell types including osteoblasts, adipocytes and neurons. SKPs are a candidate for cell-based therapy since they are easily accessible and have multipotency. Most mammalian cells are exposed to a low oxygen environment with 1 to 5% O2 concentration in vivo, while 21% O2 concentration is common in in vitro culture. The difference between in vitro and in vivo O2 concentration may affect to the behavior of cultured cells.

In this report, we investigated the effect of hypoxic condition on stemness and proliferation of SKPs. The results indicated that SKPs exposed to hypoxic condition for 5 days showed no change in proliferation. In terms of mRNA expression, hypoxia maintained expression of stemness markers; whereas, oncogenes, such as Klf4 and c-Myc, were downregulated, and the expression of Nestin, related to cancer migration, was also downregulated. Thus, SKPs cultured in hypoxia may reduce the risk of cancer in SKP cell-based therapy.


초록


    National Research Foundation
    NRF-2006-2004042 Seoul National University
    No. 2015048003
    © The Korean Academy of Oral Biology. All rights reserved.

    This is an Open-Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/3.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

    Introduction

    Skin derived precursor cells (SKPs) are multipotent stem cells that have self-renewal potential and differentiate into various cell types such as mesenchymal lineages, neurons and glial cells. SKPs can be isolated from dorsal skin and whisker follicle and are cultured in serum-free medium containing epidermal growth factor and fibroblast growth factor [1, 2]. There are many researches related to cell therapy and these researches often use neural stem cells and mesenchymal stem cells as the cell sources [3-6]. Some researches suggest that SKPs can also be used as autologous cell sources for cell therapy because they have some advantages [7, 8]. First, SKPs can be obtained from skin. It is relatively easy to be collected by noninvasive procedure. Second, SKPs can be propagated in in vitro and have an ability to differentiate into several cell types [7, 9]. There are some cases that in vivo transplanted SKPs are functional, for example, SKP-derived schwann cells form myelin and repair in transected sciatic nerve model [10].

    The control of self-renewal and cell-fate commitment is an important issue for therapeutic use of cells [4, 5]. In general, most mammalian cells are routinely cultured in vitro at 21% O2 concentration (normoxia) in laboratory, whereas the cells are exposed to much lower O2 concentration environment (1-5%; hypoxia) in mammalian bodies. Oxygen tension affects the cellular metabolism and hypoxia has an influence on proliferation and differentiation of stem cells [11-13]. However, the effect of hypoxic culture condition on SKPs has not been investigated yet. In the present study, we focused on how hypoxic condition affect to the proliferation and stemness of SKPs.

    Materials and Methods

    Animals

    Pregnant Imprinting Control Region (ICR) mice were purchased from Taconic Biosciences Inc. (Eumseong, Korea). E17.5 fetuses of ICR mice were used in this study. All procedures were approved by the Experimental Animal Committee of Seoul National University.

    Primary cell culture and subculture

    Embryonic murine SKPs were cultured as previously described [1, 14]. Briefly, dorsal back skin from mouse embryos (E17.5) was dissected and cut into 1 mm2 pieces. Skin was digested with 0.05% trypsin-EDTA for 60 min at 37℃ and filtered through a 40 μm cell strainer (BD Biosciences, MA, USA). The filtered cells were plated in a 10 cm tissue culture dish in a 37℃, 5% CO2 incubator. Cells were grown in DMEM/F12 (3:1; Life Technologies, CA, USA), containing 1% penicillin/streptomycin (Life Technologies), 2% B27 supplement (Life Technologies), 20 ng/ml epidermal growth factor (EGF; PeproTech, NJ, USA), and 40 ng/ml basic fibroblast growth factor (bFGF; PeproTech). Cells formed spheres and the spheres were passaged every 7 days. Spheres were collected and digested in accutase (Life Technologies) for 5min and then plated at a density of 3x105 cells per ml.

    Hypoxic environments

    Hypoxia culture conditions were maintained by incubating SKPs in an N2/CO2 multi gas incubator (SMA-30D, Astec, Japan). Cells were exposed to hypoxic conditions (2% O2 and 5% CO2) at 37℃ for 5 days.

    Cell proliferation

    The proliferation was assessed by the EZ-Cytox (Daeil Lab Service, Seoul, Korea), based on the water soluble tetrazolium salt-1 (WST) method. Briefly, cultured SKPs in normoxia and hypoxia were seeded in a 96 well plate and the WST reagent solution (10 μl) was added to each well and incubated for 2 h at normoxic and hypoxic condition, respectively. The absorbance was measured at 450 nm using a microplate reader (EMax Plus, Molecular devices, CA, USA).

    RT-PCR and Quantitative reverse transcriptionpolymerase chain reaction (qRT- PCR)

    RNA was extracted using the PureLinkTM RNA Mini Kit (Life Technologies) and the synthesis of cDNA was performed using LabopassTM M-MuLV Reverse Transcriptase (Cosmo Genetech, Seoul, Korea) according to the manufacturer’s instructions. Primers are shown in Table 1. PCR was performed using T100TM Thermal cycler (Biorad, CA, USA) with the following program: 95℃ for 5 min followed by 34 cycles of 95℃ for 30 s, 62℃ for 15 s, 72℃ for 30 s and 72℃ for 5 min. PCR product were confirmed by electrophoresis on a 1% agarose gel with SYBR Safe DNA gel stain (Life Technology) and visualized by InGenius System (Syngene, Cambridge, UK). Quantitative PCR was performed using SYBR Premix Ex TaqTM Ⅱ (Takara, Shiga, Japan) and 7500 Real-time PCR System (Applied Biosystems, CA, USA). The program was set as below: 95℃ for 30 s followed by 40 cycles of 95℃ for 5 s, 60℃ for 34 s, 95℃ for 15 s, 60℃ for 1 min and 95℃ for 15 s. Comparative CT method ddCt was used to measure the level of expression. Glyceraldehyde 3-phosphate dehydrogenase (GAPDH) was used as a housekeeping gene for normalization.

    Data analysis

    Data were analyzed using a student’s t-test. All data were presented as means ± standard deviation (SD). P < 0.05 was defined statistical significance.

    Result

    Morphologic differences of spheres in the normoxia and hypoxia

    SKPs formed floating spheres in non-adherent conditions. Single cells from SKP were plated in culture dishes and then after 2-3 days, primary spheres were observed and the spheres grew in size until 5-7 days. There were no significant morphological differences between normoxic spheres and hypoxic spheres, except for their size. In hypoxia group, size of SKP spheres was smaller than the spheres in normoxia group (Fig 1A). The spheres were counted after 5 days of seeding. The number of spheres tended to increase in hypoxia condition, although the change was not statistically significant (Fig 1B).

    Proliferation of SKPs in hypoxia

    SKPs were cultured in the SKP medium containing EGF and bFGF. There was no effect on the proliferation of the cells between two experimental groups (Fig 2).

    Gene expression changes in hypoxia

    RT-PCR was performed to evaluate the change of gene expression levels in two experimental groups. As shown in Fig 3A, Hif1a expression was significantly higher in hypoxic condition. Expression level of Oct4, Sox2, Nanog, and Rex1 genes were not significantly different between normoxia and hypoxia groups. Interestingly, Klf4, c-Myc, and Nestin genes showed approximately 40% lower levels of mRNA expression in hypoxic condition when compared with in normoxic condition (Fig 3B).

    Discussion

    SKPs derived from dermis have ability to form floating colonies, which are called spheres in suspension cultures [1, 2, 14]. SKPs maintain their multipotency and have capacity to differentiate into multiple lineages [15, 16]. The newly isolated SKPs contain heterogenous populations that are mixed with SKPs, adipose cells, muscle cells and blood cells [1]. In this study, passage 2 SKPs were used to obtain more homogenous population of the cells.

    Oxygen is important to life. It is required to produce metabolic energy and maintain cellular processes [17]. Oxygen concentration in standard cell culture is 21% (normoxia). However, most tissues are exposed to low O2, 1-5% (hypoxia) in mammalian body and hypoxic condition is considered as stem cell niche [18]. In previous studies, hypoxic condition has influences on the proliferation and differentiation of stem cells including neural stem cells [19], mesenchymal stem cells [20], and induced pluripotent stem cells [21]. In the present study, the size of SKP spheres in hypoxia were smaller than normoxia group ones. Hypoxia had no effect on the proliferation of the cells although a few more spheres were observed. These results indicate that O2 concentration is not critical for the formation of spheres and proliferation in SKPs when SKPs were cultured in hypoxia for 5 days. The proliferation of cells is not affected in some studies [22, 23]. Since the lifespan of multipotential stromal cells (MSCs) is prolonged by increasing senescence marker, P16INK4A, under normoxic condition, the proliferation of MSCs has no changes between hypoxia and normoxia [23]. Increased level of senescence marker in normoxic condition also might be the reason for our results.

    Maintenance of oxygen homeostasis is essential for cell survival in hypoxic condition [24]. Hypoxia-inducible factor (Hif) is a heterodimeric transcription factor composed of alpha (a) subunit and beta (b) subunit [25]. Hif b subunit is constitutive expressed, whereas Hif a subunits (Hif1a, Hif2a and Hif3a) are expressed in an O2 dependent manner [24, 26]. The role of Hif3a is not yet clearly understood while Hif2a is insensitive to O2 changes [27]. Hif1a is sensitive to hypoxia [27], and is also confirmed in this report. Hypoxic condition suppressed the expression of Klf4, c-Myc, and Nestin. Klf4 and c-Myc are well known as transcription factors that have an oncogenic function [28, 29]. These oncogenes are required for maintenance of cancer stem cells and tumor growth was also delayed and reduced when these oncogenes were downregulated [30, 31]. Nestin, an intermediate filament protein and marker of several progenitor cells, is expressed in some cancer cells such as lung [32], breast [33] prostate [34, 35] and pancreatic [34, 35] cancer cells. Since the expression of Nestin is related to malignancy and metastasis of cancers [36], downregulation of this gene inhibits the migration and growth of tumors [37]. Although hypoxia is also known as a critical factor of cancer, the effect of hypoxia in cancer has not been clearly determined yet. The tumor growth and metastasis are increased in acute hypoxia, from 30 min to 72 h [38]. On the other hand, tumor progression was repressed when tumor cells were exposed to the chronic hypoxia, from 72 h to 2 weeks. Interestingly, this effect was maintained for 3 weeks even after the hypoxic condition was removed [39]. Although the precise effect of chronic hypoxia in stem cells and precursors has not been understood yet, acute hypoxic condition promote the proliferation and increase the expression of stemness genes in various stem and cancer cells [38, 40]. In the present study, the SKPs exposed to hypoxic condition for 5 days, which can be considered as chronic hypoxia, show repression of oncogenes similar to tumor cells under hypoxic condition whereas the genes related with stemness maintenance was not affected under same condition.

    In conclusion, hypoxic culture condition may reduce the risk of cancer, when SKPs are used for cell transplantation therapy.

    Acknowledgements

    This study was supported by a grant from the National Research Foundation of Korea (NRF-2006-2004042, and No. 2015048003 through the Oromaxillofacial Dysfunction Research Center for the Elderly at Seoul National University)

    Figure

    IJOB-41-69_F1.gif
    Effect of hypoxic condition for sphere formation of Skin-derived precursor cells (SKPs).

    (A) Sphere morphology of skin-derived precursor cells in normoxic and hypoxic condition. (B) The average number of spheres in normoxia group and hypoxia group. Data is expressed as mean ± standard deviation (n=4).

    IJOB-41-69_F2.gif
    Proliferation of SKPs.

    Proliferation was assessed by water soluble tetrazolium salt-1 assay. Data of proliferation is expressed as mean ± standard deviation (n=3).

    IJOB-41-69_F3.gif
    Expression patterns of mRNA in skin-derived precursor cells.

    (A) Expression of patterns of Hif1a, Oct4, Sox2, Nanog, Rex1, Klf4, c-Myc and Nestin. (B) Relative mRNA levels of the Klf4, c-Myc and Nestin genes. GAPDH was used for normalization. Data are expressed as mean ± standard deviation (n=3). * indicates statistical significance between two groups. *P < 0.05.

    Table

    Reference

    1. Biernaskie JA , McKenzie IA , Toma JG , Miller FD (2006) Isolation of skin-derived precursors (SKPs) and differentiation and enrichment of their Schwann cell progeny , Nat Protoc, Vol.1 ; pp.2803-2812
    2. Toma JG , Akhavan M , Fernandes KJ , Barnabe-Heider F , Sadikot A , Kaplan DR , Miller FD (2001) Isolation of multipotent adult stem cells from the dermis of mammalian skin , Nat Cell Biol, Vol.3 ; pp.778-784
    3. Kim JA , Ha S , Shin KY , Kim S , Lee KJ , Chong YH , Chang KA , Suh YH (2015) Neural stem cell transplantation at critical period improves learning and memory through restoring synaptic impairment in Alzheimer's disease mouse model , Cell Death Dis, Vol.6 ; pp.e1789
    4. Kim SU , Lee HJ , Kim YB (2013) Neural stem cell-based treatment for neurodegenerative diseases , Neuropathology, Vol.33 ; pp. 491-504
    5. Xu S , Liu H , Xie Y , Sang L , Liu J , Chen B (2015) Effect of mesenchymal stromal cells for articular cartilage degeneration treatment: a meta-analysis , Cytotherapy, Vol.17 ; pp.1342-1352
    6. Ho MS , Mei SH , Stewart DJ (2015) The Immunomodulatory and Therapeutic Effects of Mesenchymal Stromal Cells for Acute Lung Injury and Sepsis , J Cell Physiol, Vol.230 ; pp.2606-2617
    7. Tunici P , Bulte JW , Bruzzone MG , Poliani PL , Cajola L , Grisoli M , Douglas T , Finocchiaro G (2006) Brain engraftment and therapeutic potential of stem/progenitor cells derived from mouse skin , J Gene Med, Vol.8 ; pp.506-513
    8. Kwok CK , Tam PK , Ngan ES (2013) Potential use of skin-derived precursors (SKPs) in establishing a cell-based treatment model for Hirschsprung's disease , J Pediatr Surg, Vol.48 ; pp.619-628
    9. Mao D , Yao X , Feng G , Yang X , Mao L , Wang X , Ke T , Che Y , Kong D (2015) Skin-derived precursor cells promote angiogenesis and stimulate proliferation of endogenous neural stem cells after cerebral infarction , Biomed Res Int, Vol.2015 ; pp.945846
    10. Liu Y , Chen J , Liu W , Lu X , Liu Z , Zhao X , Li G , Chen Z (2016) A Modified Approach to Inducing Bone Marrow Stromal Cells to Differentiate into Cells with Mature Schwann Cell Phenotypes , Stem Cells Dev, Vol.25 ; pp.347-359
    11. Santilli G , Lamorte G , Carlessi L , Ferrari D , Rota Nodari L , Binda E , Delia D , Vescovi AL , De Filippis L (2010) Mild hypoxia enhances proliferation and multipotency of human neural stem cells , PLoS One, Vol.5 ; pp.e8575
    12. Braunschweig L , Meyer AK , Wagenfuhr L , Storch A (2015) Oxygen regulates proliferation of neural stem cells through Wnt/ beta-catenin signalling , Mol Cell Neurosci, Vol.67 ; pp.84-92
    13. Chen HC , Sytwu HK , Chang JL , Wang HW , Chen HK , Kang BH , Liu DW , Chen CH , Chao TT , Wang CH (2011) Hypoxia enhances the stemness markers of cochlear stem/progenitor cells and expands sphere formation through activation of hypoxiainducible factor-1 alpha , Hear Res, Vol.275 ; pp.43-52
    14. Fernandes KJ , McKenzie IA , Mill P , Smith KM , Akhavan M , Barnabe-Heider F , Biernaskie J , Junek A , Kobayashi NR , Toma JG , Kaplan DR , Labosky PA , Rafuse V , Hui CC , Miller FD (2004) A dermal niche for multipotent adult skin-derived precursor cells , Nat Cell Biol, Vol.6 ; pp.1082-1093
    15. Fernandes KJ , Kobayashi NR , Gallagher CJ , Barnabe-Heider F , Aumont A , Kaplan DR , Miller FD (2006) Analysis of the neurogenic potential of multipotent skin-derived precursors , Exp Neurol, Vol.201 ; pp.32-48
    16. Lorenz K , Sicker M , Schmelzer E , Rupf T , Salvetter J , Schulz-Siegmund M , Bader A (2008) Multilineage differentiation potential of human dermal skin-derived fibroblasts , Exp Dermatol, Vol.17 ; pp.925-932
    17. Csete M (2005) Oxygen in the cultivation of stem cells , Ann N Y Acad Sci, Vol.1049 ; pp.1-8
    18. Mohyeldin A , Garzon-Muvdi T , Quinones-Hinojosa A (2010) Oxygen in stem cell biology: a critical component of the stem cell niche , Cell Stem Cell, Vol.7 ; pp.150-161
    19. Gustafsson MV , Zheng X , Pereira T , Gradin K , Jin S , Lundkvist J , Ruas JL , Poellinger L , Lendahl U , Bondesson M (2005) Hypoxia requires notch signaling to maintain the undifferentiated cell state , Dev Cell, Vol.9 ; pp.617-628
    20. Beegle J , Lakatos K , Kalomoiris S , Stewart H , Isseroff RR , Nolta JA , Fierro FA (2015) Hypoxic preconditioning of mesenchymal stromal cells induces metabolic changes, enhances survival, and promotes cell retention in vivo , Stem Cells, Vol.33 ; pp.1818-1828
    21. Yoshida Y , Takahashi K , Okita K , Ichisaka T , Yamanaka S (2009) Hypoxia enhances the generation of induced pluripotent stem cells , Cell Stem Cell, Vol.5 ; pp.237-241
    22. Holzwarth C , Vaegler M , Gieseke F , Pfister SM , Handgretinger R , Kerst G , Muller I (2010) Low physiologic oxygen tensions reduce proliferation and differentiation of human multipotent mesenchymal stromal cells , BMC Cell Biol, Vol.11 ; pp.11
    23. Tamama K , Kawasaki H , Kerpedjieva SS , Guan J , Ganju RK , Sen CK (2011) Differential roles of hypoxia inducible factor subunits in multipotential stromal cells under hypoxic condition , J Cell Biochem, Vol.112 ; pp.804-817
    24. Bruick RK (2003) Oxygen sensing in the hypoxic response pathway regulation of the hypoxia-inducible transcription factor , Genes Dev, Vol.17 ; pp.2614-2623
    25. Ivan M , Kondo K , Yang H , Kim W , Valiando J , Ohh M , Salic A , Asara JM , Lane WS , Kaelin WG Jr (2001) HIFalpha targeted for VHL-mediated destruction by proline hydroxylation implications for O2 sensing , Science, Vol.292 ; pp.464-468
    26. Jaakkola P , Mole DR , Tian YM , Wilson MI , Gielbert J , Gaskell SJ , von Kriegsheim A , Hebestreit HF , Mukherji M , Schofield CJ , Maxwell PH , Pugh CW , Ratcliffe PJ (2001) Targeting of HIF-alpha to the von Hippel-Lindau ubiquitylation complex by O2-regulated prolyl hydroxylation , Science, Vol.292 ; pp. 468-472
    27. Wenger RH , Stiehl DP , Camenisch G (2005) Integration of oxygen signaling at the consensus HRE , Sci STKE, Vol.2005 ; pp.re12
    28. Shi YAi W (2013)
    29. Miller DM , Thomas SD , Islam A , Muench D , Sedoris K (2012) c-Myc and cancer metabolism , Clin Cancer Res, Vol.18 ; pp.5546-5553
    30. Yu F , Shi Y , Wang J , Li J , Fan D , Ai W (2013) Deficiency of Kruppel-like factor KLF4 in mammary tumor cells inhibits tumor growth and pulmonary metastasis and is accompanied by compromised recruitment of myeloid-derived suppressor cells , Int J Cancer, Vol.133 ; pp.2872-2883
    31. Gordan JD , Bertout JA , Hu CJ , Diehl JA , Simon MC (2007) HIF-2alpha promotes hypoxic cell proliferation by enhancing c-myc transcriptional activity , Cancer Cell, Vol.11 ; pp.335-347
    32. Chen Z , Wang J , Cai L , Zhong B , Luo H , Hao Y , Yu W , Wang B , Su C , Lei Y , Bella AE , Xiang AP , Wang T (2014) Role of the stem cell-associated intermediate filament nestin in malignant proliferation of non-small cell lung cancer , PLoS One, Vol.9 ; pp.e85584
    33. Zhao Z , Lu P , Zhang H , Xu H , Gao N , Li M , Liu C (2014) Nestin positively regulates the Wnt/beta-catenin pathway and the proliferation, survival and invasiveness of breast cancer stem cells , Breast Cancer Res, Vol.16 ; pp.408
    34. Hyder CL , Lazaro G , Pylvanainen JW , Roberts MW , Qvarnstrom SM , Eriksson JE (2014) Nestin regulates prostate cancer cell invasion by influencing the localisation and functions of FAK and integrins , J Cell Sci, Vol.127 ; pp.2161-2173
    35. Matsuda Y (2013) The Roles and Molecular Mechanisms of Nestin Expression in Cancer with a Focus on Pancreatic Cancer , Journal of Carcinogenesis & Mutagenesis, Vol.01
    36. Ehrmann J , Kolar Z , Mokry J (2005) Nestin as a diagnostic and prognostic marker immunohistochemical analysis of its expression in different tumours , J Clin Pathol, Vol.58 ; pp. 222-223
    37. Narita K , Matsuda Y , Seike M , Naito Z , Gemma A , Ishiwata T (2014) Nestin regulates proliferation, migration, invasion and stemness of lung adenocarcinoma , Int J Oncol, Vol.44 ; pp. 1118-1130
    38. Cairns RAHill RP (2004) Acute hypoxia enhances spontaneous lymph node metastasis in an orthotopic murine model of human cervical carcinoma , Cancer Res, Vol.64 ; pp.2054-2061
    39. Yu LHales CA (2011) Long-term exposure to hypoxia inhibits tumor progression of lung cancer in rats and mice , BMC Cancer, Vol.11 ; pp.331
    40. Yu X , Lu C , Liu H , Rao S , Cai J , Liu S , Kriegel AJ , Greene AS , Liang M , Ding X (2013) Hypoxic preconditioning with cobalt of bone marrow mesenchymal stem cells improves cell migration and enhances therapy for treatment of ischemic acute kidney injury , PLoS One, Vol.8 ; pp.e62703