Open Access

Establishment of mesenchymal stem cell lines derived from the bone marrow of green fluorescent protein‑transgenic mice exhibiting a diversity in intracellular transforming growth factor‑β and bone morphogenetic protein signaling

  • Authors:
    • Shunsuke Sawada
    • Naoyuki Chosa
    • Naoki Takizawa
    • Jun Yokota
    • Yasuyuki Igarashi
    • Koichi Tomoda
    • Hisatomo Kondo
    • Takashi Yaegashi
    • Akira Ishisaki
  • View Affiliations

  • Published online on: January 18, 2016     https://doi.org/10.3892/mmr.2016.4794
  • Pages: 2023-2031
  • Copyright: © Sawada et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Cytokines and their intercellular signals regulate the multipotency of mesenchymal stem cells (MSCs). The present study established the MSC lines SG‑2, ‑3, and ‑5 from the bone marrow of green fluorescent protein (GFP)‑transgenic mice. These cell lines clearly expressed mouse MSC markers Sca‑1 and CD44, and SG‑2 and ‑5 cells retained the potential for osteogenic and adipogenic differentiation in the absence of members of the transforming growth factor (TGF)‑β superfamily. By contrast, SG‑3 cells only retained adipogenic differentiation potential. Analysis of cytokine and cytokine receptor expression in these SG cell lines showed that bone morphogenetic protein (BMP) receptor 1B was most highly expressed in the SG‑3 cells, which underwent osteogenesis in response to BMP, while TGF‑β receptor II was most highly expressed in SG‑3 and ‑5 cells. However, it was unexpectedly noted that phosphorylation of Smad 2, a major transcription factor, was induced by TGF‑β1 in SG‑2 cells but not in SG‑3 or ‑5 cells. Furthermore, TGF‑β1 clearly induced the expression of Smad‑interacting transcription factor CCAAT/enhancer binding protein‑β in SG‑2 but not in SG‑3 or ‑5 cells. These results demonstrated the establishment of TGF‑β‑responsive SG‑2 MSCs, BMP‑responsive SG‑3 MSCs and TGF‑β/BMP‑unresponsive SG‑5 MSCs, each of which was able to be traced by GFP fluorescence after transplantation into in vivo experimental models. In conclusion, the present study suggested that these cell lines may be used to explore how the TGF‑β superfamily affects the proliferation and differentiation status of MSCs in vivo.

Introduction

Mesenchymal stem cells (MSCs), which were first derived from bone marrow, have self-renewal properties and are able to differentiate into a variety of mesenchymal tissue types (13). In stem cell therapy, human bone marrow-derived MSCs (BM-MSCs) are expanded in vitro and subsequently autoimplanted, which eliminates the risk of immune rejection. BM-MSCs are able to differentiate into osteoblasts, chondrocytes and adipocytes (4), and are a major source of bone regeneration and remodeling during homeostasis (58). In addition, immunophenotype evaluation demonstrated that mouse BM-MSCs express Sca-1 and CD44, but not CD11b or CD45 (9).

The transforming growth factor (TGF)-β superfamily includes the TGF-β/activin/Nodal family and the bone morphogenetic protein (BMP)/growth and differentiation factor (GDF)/Mullerian inhibiting substance (MIS) family (10). On the cell surface, binding of ligands to receptors triggers the formation of a tetrameric complex of type I and II receptors. Type II receptor kinase activates type I receptor kinase, which transduces the signal through phosphorylation of receptor-activated Smads (R-Smads) (1114). Smad proteins are the central mediators of TGF-β superfamily signaling. R-Smads, including Smad 1, Smad 5 and Smad 8, are primarily activated by BMP-specific type I receptors, whereas Smad 2 and Smad 3 are activated by the TGF-β-specific type I receptors. Activated R-Smads form complexes with the common mediator Smads (Co-Smads; e.g., Smad 4), which translocate into the nucleus, where they and their partner proteins regulate the transcription of specific target genes. Abnormal intensity of Smad-mediated TGF-β/BMP signals is associated with various human diseases, including bone and immune disorders, fibrosis, and cancer progression or metastasis (15). Of note, TGF-β superfamily-induced intracellular signals affect osteogenesis and adipogenesis of MSCs; for instance, BMP has been observed to potentiate osteogenic and adipogenic differentiation of undifferentiated mesenchymal cells (16). By contrast, TGF-β potentiates osteogenic differentiation of BM-MSCs (17,18), although none of these results have been confirmed in vivo.

Recent studies have focused on controlling TGF-β/BMP signals for the discovery of pharmacotherapeutics; however, the detection of therapeutic molecular targets in these pathways has not been successful, probably because most trials are performed in vitro, not in vivo (19,20). Therefore, it is important to establish appropriate in vivo experimental models to evaluate the role of TGF-β/BMP signaling in disease development or healing. The present study aimed to establish MSC cell lines derived from bone marrow of green fluorescent protein (GFP)-transgenic mice; the cells and their diverse, intracellular BMP and TGF-β signals can be tracked after transplantation into in vivo experimental models. These cell lines are available for in vivo molecular studies that aim to determine how the TGF-β superfamily affects MSC proliferation and differentiation in diseases including fibrosis and cancer progression or metastasis (21,22), and in tissue repair processes, including tissue reconstruction and anti-inflammatory responses (23).

Materials and methods

Bone marrow-derived cells from GFP-transgenic mice

All experimental procedures were performed in accordance with the guidelines established by the Animal Studies Committee at Iwate Medical University (Iwate, Japan). A total of four GFP-transgenic mice (24) were obtained from the Center for in vivo Science, Iwate Medical University (Iwate, Japan). The mice were sacrificed by excessive inhalation of CO2. Cells were flushed from the tibia of three-week-old GFP-transgenic mice with phosphate-buffered saline (PBS) containing 0.5% fetal bovine serum (FBS; PAA Laboratories, GE Healthcare, Piscataway, NJ, USA) and 2 mM EDTA, and then seeded into plastic cell culture dishes (Nunc; Thermo Fisher Scientific, Waltham, MA, USA) with Dulbecco's modified Eagle's medium (DMEM; Sigma-Aldrich, St. Louis, MO, USA) containing 10% FBS. The cells were cultured for 1 week under hypoxic conditions (5% O2, 5% CO2 and 90% N2). Cells were re-plated upon reaching 80% confluence.

Co-transfection of hTERT and SV40 large T antigen (SV40LT) genes

The expanded cells were transfected with pBABE-neo-hTERT and pBABE-pur-SV40LT plasmids encoding neomycin and puromycin resistance (provided by Addgene, Cambridge, MA, USA) with Lipofectamine LTX (Invitrogen Life Technologies, Carlsbad, CA, USA) according to manufacturer instructions. Cells were then incubated in DMEM containing 10% FBS, 150 µg/ml G418 (Gibco-BRL, Thermo Fisher Scientific) or 1 µg/ml puromycin (Gibco-BRL) under hypoxic conditions for 12–15 days. The surviving cells were trypsinized and allowed to grow in 90-mm culture dishes.

Single-cell cloning

Single-cell clones were obtained using the limited dilution method. After hTERT and SV40LT transfection and selection with G418 and puromycin, the surviving cells were seeded on a 96-well plate (Nunc) at 0.5% cells per well, and then cultured under hypoxic conditions. After 10 days, the cells were sub-cultured in 24-well plates (Nunc). This was repeated until confluence was reached at 20 days after single cell cloning. Population doubling (PD) was defined as the number of doublings required for a single cell to reach confluence in a 60-mm culture dish (Nunc) under hypoxic conditions. PD was estimated for clones SG-2, -3, -5 and -6.

Telomeric repeat amplification protocol

Telomerase activity in bone marrow-derived cell lines was assayed by the stretch PCR method using the Quantitative Telomerase Detection kit (Allied Biotech, Vallejo, CA, USA) according to manufacturer's instructions. The PCR mixture contained QTD premixed buffer and SYBR green one dye (cat. no. MT3010; Allied Biotech, Inc., St. Benicia, CA, USA). Quantification of telomerase activity was performed under the following amplification conditions using a Thermal Cycler Dice Real Time system (Takara Bio, Otsu, Japan) according to manufacturer's instructions: 25°C for 20 min, initial activation at 95°C for 10 min, denaturation at 90°C for 30 s, annealing at 60°C for 30 s, and a final extension of 40 cycles at 72°C for 30 s. The PCR products were separated by 10–20% polyacrylamide gel electrophoresis and stained with ethidium bromide.

Detection of SV40LT by immunocytochemistry

Bone marrow-derived cell lines were seeded onto eight-well culture slides (BD Biosciences, Franklin Lakes, NJ, USA). After 24 h, the cells were fixed with 4% paraformaldehyde and washed five times with PBS. For detection of SV40LT, cells were incubated with mouse monoclonal anti-SV40LT (1:100; cat. no. ab16879; Abcam, Cambridge, UK) antibody for 1 h at room temperature. The cells were then incubated with Alexa Fluor 594 goat polyclonal anti-mouse secondary antibodies (1:500; cat. no. A11005; Thermo Fisher Scientific) and DAPI (1:500; cat. no. D9542; Sigma-Aldrich) for 30 min at room temperature. Fluorescence was examined by using a fluorescence microscope (Olympus ix70; Olympus Corporation, Tokyo, Japan).

Detection of MSC markers by flow cytometry

A total of 1.0×105 bone marrow-derived cell lines (SG-2, -3, -5 and -6) were suspended in PBS containing 0.5 FBS and 2 mM EDTA and incubated with phycoerythrin-conjugated monoclonal anti-mouse Sca-1 (1:10; cat. no. 130-093-224), monoclonal anti-mouse CD44 (1:10; cat. no. 130-096-838), monoclonal anti-mouse CD11b (1:10; cat. no. 130-091-240) or monoclonal anti-mouse CD45 (1:10; cat. no. 130-091-610) antibody for 1 h at 4°C in the dark. All antibodies were purchased from Miltenyi Biotec (Bergisch Gladbach, Germany). Acquisition was performed using an EPICS XL ADC system (Beckman Coulter, Brea, CA, USA).

Adipogenic and osteogenic differentiation

In vitro differentiation was performed according to a previous study by our group (25). To induce osteogenic differentiation, confluent cells were incubated in osteogenic differentiation medium (ODM) under hypoxic conditions for two weeks. Bone matrix mineralization was evaluated by Alizarin red S (Sigma-Aldrich) staining. To induce adipogenic differentiation, cells were cultured to near confluence and cultured in adipogenic differentiation medium (ADM) under hypoxic conditions for two weeks. At the end of the differentiation period, lipid droplets were stained with Oil Red O (Sigma-Aldrich).

Expression profiling of cytokines and cytokine receptors

Gene expression profiling was performed using a PrimerArray of mouse cytokine-cytokine receptor interaction (Takara Bio) in combination with a Thermal Cycler Dice Real Time System (Takara Bio) according to manufacturer's instructions. This PrimerArray is a set of real-time reverse transcription-polymerase chain reaction (RT-PCR) primers used for the analysis of RNA expression. The array contains a mixture of 96 primer pairs for 88 target genes and eight housekeeping genes. Quantification of gene expression was performed using a PrimerArray Analysis Tool version 2.0 (Takara Bio).

Western blot analysis

SG-2, -3 and -5 cells were serum-starved overnight and stimulated with 50 ng/ml BMP-2 (R&D Systems, Minneapolis, MN, USA) or 50 ng/ml TGF-β1 (R&D Systems). The cells were washed twice with ice-cold PBS and then lysed in radioimmunoprecipitation assay buffer (50 mM Tris-HCl, pH 7.2, 150 mM NaCl, 1% NP-40, 0.5% sodium deoxycholate, and 0.1% SDS) containing protease and phosphatase inhibitor cocktails (Sigma-Aldrich). The protein content was quantified using the bicinchoninic acid method (Pierce; Thermo Fisher Scientific). Samples containing equal amounts of protein were separated by 12.5% SDS-PAGE and transferred onto a polyvinylidene difluoride membrane (Merck Millipore, Darmstadt, Germany). After blocking with 5% nonfat dry milk in 50 mM Tris-HCl, pH 7.2, containing 150 mM NaCl and 0.1% Tween-20 for 2 h at room temperature, the membrane was incubated with primary rabbit monoclonal anti-phospho-Smad 5 (1:1,000; cat. no. ab92698; Abcam), anti-Smad 1/5/8 (1:1,000, cat. no. 12656; Cell Signaling Technology, Danvers, MA, USA), rabbit monoclonal anti-phospho-Smad 2 (1:1,000; cat. no. 04-953; Merck Millipore), or anti-Smad 2/3 (1:1,000; cat. no. 610843; BD Biosciences) antibody overnight at 4°C, with mouse monoclonal anti-β-actin (1:1,000; cat. no. sc-47778; Santa Cruz Biotechnology, Dallas, TX, USA) antibody as a loading control. The blots were incubated with alkaline phosphatase-conjugated secondary antibody and developed using the 5-bromo-4-chloro-3′-indolyphosphate/nitro-blue tetrazolium membrane phosphatase substrate system (cat. no. 50-81-00; Kirkegaard & Perry Laboratories, Gaithersburg, MD, USA).

RNA isolation and RT-quantitative (q)PCR

SG-2, -3 and -5 cells were stimulated with TGF-β1 (1–50 ng/ml; R&D Systems) for 24 h. Total RNA was isolated using ISOGEN reagent (Nippongene, Tokyo, Japan) according to the manufacturer's instructions. First-strand cDNA was synthesized from total RNA with the PrimeScript RT Reagent kit (Takara Bio). RT-qPCR was performed on a Thermal Cycler Dice Real Time System (Takara Bio) with SYBR Premix Ex Taq II (Takara Bio). Expression of CCAAT/enhancer binding protein-β (C/EBPβ) was normalized to β-actin and relative expression levels were calculated as a fold-increase or -decrease relative to the control. Transcripts were detected with primers (designed using a Perfect Real Time Support system; Takara Bio) for C/EBPβ (sense, 5′-GACAAGCTGAGCGACGAGTA-3′; and anti-sense, 5′-AGCTGCTCCACCTTCTTCTG-3′) and β-actin (sense, 5′-CATCCGTAAAGACCTCTATGCCAAC-3′ and anti-sense, 5′-ATGGAGCCACCGATCCACA-3′). For each PCR run, cDNA derived from 50 ng total RNA was used. Following initial denaturation at 95°C for 30 s, a two-step cycle procedure was used (denaturation at 95°C for 5 s and annealing and extension at 60°C for 30 s) for 40 cycles. The relative mRNA expression levels in each sample were calculated using the 2−ΔΔCT method (26).

Statistical analysis

All experiments were repeated at least three times. Representative images or data are shown. Values are expressed as the mean ± standard deviation. Differences between control and test samples were analyzed using paired two-tailed Student's t-tests. P<0.05 was considered to indicate a statistically significant difference between values.

Results

Establishment of cell lines from the bone marrow of GFP-transgenic mice

Bone marrow cells were flushed from the tibia of GFP mice and cultured under hypoxic conditions. Adherent cells were transformed with hTERT and SV40LT vectors, yielding four single cell-derived cell lines SG-2, -3, -5 and -6. As shown in Fig. 1, the cell lines exhibited fibroblastic morphology (Fig. 1, upper panel) and GFP fluorescence (Fig. 1, lower panel). All cell lines exhibited nuclear SV40LT expression (Fig. 2A). At PD 20, a stretch PCR assay indicated telomerase activity in all cell lines, but not in the negative control (Fig. 2B). Thus, the bone marrow-derived cell lines exhibited telomerase activity and SV40LT expression. All cell lines grew at a similar rate of ~1 PD every two days (Fig. 2C). The cells divided at least 60 times and were passaged >30 times, thus demonstrating successful immortalization.

Bone marrow-derived SG cell lines have MSC-like features

To determine their MSC character, the SG-2, -3, 5, and -6 cell lines were analyzed for the expression of mouse MSC markers and differentiation potential. Sca-1 is the most reliable MSC marker in mice and was strongly expressed in SG-2, -3 and -5 cells, but only weakly expressed in SG-6 cells (Fig. 3). CD44 was detected at similar levels in all cell lines, although the hematopoietic stem cell markers CD11b and CD45 were not detected. The expression patterns of MSC markers suggested that the SG-2, -3 and -5 cell lines were MSCs. Next, the present study evaluated the osteogenic and adipogenic differentiation potentials of these cell lines. Bone matrix mineralization indicated by Alizarin red staining was highest in SG-5 cells (Fig. 4A). Although miner-alization was observed in SG-2 cells, it was markedly lower than that in SG-5 and was not detected in SG-3 and SG-6 cells. Thus, SG-2 and -5 cells retained their osteogenic differentiation potential, although at different levels of efficiency. Lipid droplet formation indicated by Oil Red O staining was more intense in SG-2 cells than in SG-3 and -5 cells (Fig. 4B). Thus, SG-2, -3, and -5 retained their adipogenic differentiation potential to various degrees.

Intercellular signaling by TGF-β and BMP in SG cells

In order to identify the expression profiles of cytokines and cytokine receptors in SG cells, the present study performed PrimerArray analyses and compared the results derived from SG-2, -3 and -5 cells. Differentially expressed genes are shown in Table I. BMP receptor 1B (Bmpr1b) was most highly expressed in SG-3 cells, while TGF-β receptor II (Tgfbr2) was most highly expressed in SG-3 and -5 cells, indicating differential sensitivities to BMP-2 and TGF-β. Smad 5, which is a major signaling factor activated by BMP, was most significantly phosphorylated in BMP 2-stimulated SG-3 cells (Fig. 5A). In addition, osteogenic differentiation in SG-3 cells was induced by stimulation with BMP-2 in a dose-dependent manner (Fig. 5B), suggesting these cells retained the capacity to differentiate into adipogenic (Fig. 4B) and osteogenic lineages. However, phosphorylation of Smad 2, which is activated by TGF-β, was highest in the MSC-like SG-2 cells in response to TGF-β1 (Fig. 6A). Of note, mRNA expression of C/EBPβ, which is an immune- and inflammatory response-associated as well as Smad-interacting transcription factor, was induced in SG-2 cells by TGF-β1 in a dose-dependent manner (Fig. 6B). Thus, the SG-3 cells were BMP-responsive and the SG-2 cells were TGF-β-responsive, while SG-5 cells were BMP/TGF-β-unresponsive MSCs.

Table I

Genes for which expression changed by >2-fold in SG-2 vs. SG-3 and SG-5.

Table I

Genes for which expression changed by >2-fold in SG-2 vs. SG-3 and SG-5.

Gene symbolGene nameFold change with SG-2
vs. SG-3vs. SG-5
Bmpr1bBone morphogenetic protein receptor, type 1B45.89−2.362
EgfrEpidermal growth factor receptor4.2286.453
Ifngr2Interferon γ receptor 21.181−3.534
Il17raInterleukin 17 receptor A2.2661.790
Il18r1Interleukin 18 receptor 13.8111.007
PdgfrbPlatelet derived growth factor receptor, beta polypeptide2.5141.625
Cx3cl1Chemokine (C-X3-C motif) ligand 15.3152.346
Tgfbr1Transforming growth factor, beta receptor I2.0561.778
Tgfbr2Transforming growth factor, beta receptor II23202702
HgfHepatocyte growth factor14.835.098
KdrKinase insert domain protein receptor8.5742.888
LeprLeptin receptor2.1441.495
PdgfbPlatelet-derived growth factor, B polypeptide4.8234.993
PdgfraPlatelet-derived growth factor receptor, alpha polypeptide2.3291.444
PrlrProlactin receptor−2.071−1.014
Ccl9Chemokine (C-C motif) ligand 9−2.1741.050
Tnfrsf1bTumor necrosis factor receptor superfamily, member 1b2.1291.879
Cd40CD40 antigen−1.319−27.48
Il2rgInterleukin 2 receptor, γ chain7.1602.789
LifrLeukemia inhibitory factor receptor1.6024.112
KitlKit ligand2.0001.283
CntfrCiliary neurotrophic factor receptor5.9385.242
Cxcl14Chemokine (C-X-C motif) ligand 147.8355.205
Il17rbInterleukin 17 receptor B−2.2503.074
Ccl8Chemokine (C-C motif) ligand 81.9192.346

Discussion

In the present study, the bone marrow of GFP mice was used to establish three MSC lines immortalized by transfection with SV40LT and hTERT. SV40LT-transformed cells are not tumorigenic (2730); therefore, SV40LT is commonly used to immortalize primary mammalian cells. Another commonly used gene for immortalization is TERT, which maintains telomere length to enable cells to indefinitely proliferate. TERT expression is high in stem cells, while it is reduced upon differentiation. Restoration of TERT activity in normal somatic cells can lead to their immortalization and may be associated with the acquisition of characteristics associated with cellular transformation (31). It has been indicated that ectopic expression of the mouse TERT catalytic subunit does not affect embryonic stem cell proliferation or differentiation in vitro, but protects them from cell death during differentiation (32). Therefore, the cell lines generated in the present study may be used to study stem cell proliferation and differentiation.

The expression of MSC markers Sca-1+ and CD44+ and the absence of hematopoietic stem cell markers CD11b and CD45 were confirmed in SG-2, -3 and -5 cells, which exhibited osteogenic and adipogenic differentiation potential. These results strongly suggested that the MSC-like potential of the cells was preserved. The present study focused on cytokines and cytokine receptors that are expressed specifically in each cell line to clarify the differentiation mechanism of MSCs: Bmpr1b was most highly expressed in SG-3 but not in SG-2 or -5 cells, whereas Tgfbr2 was most highly expressed in SG-3 and -5 but not in SG-2 cells. BMP-2 induced phosphorylation of Smad 5 in SG-3 but not in SG-2 and -5 cells. Furthermore, BMP-2 induced osteogenic differentiation of SG-3 cells but did not affect osteogenic differentiation in SG-2 and -5 cells (data not shown). By contrast, TGF-β unexpectedly but clearly induced Smad 2 phosphorylation in SG-2 cells, in which expression of TGF-β receptors I and II was lower than in SG-3 and -5 cells, suggesting that Smad 2 itself or signal transduction molecules upstream of Smad 2 may have been inactivated in SG-3 and -5 cells. Thus, the present study established TGF-β-responsive SG-2 cells, BMP-responsive SG-3 cells and TGF-β/BMP-unresponsive SG-5 cells that can be traced by GFP fluorescence after transplantation into in vivo experimental models.

Of note, TGF-β stimulation of SG-2 induced expression of C/EBPβ, a Smad-interacting transcription factor (33). C/EBPβ is a member of the C/EBP family of transcription factors (C/EBPα, C/EBPβ, C/EBPγ, C/EBPδ, C/EBPε and C/EBPξ) (3436). C/EBPβ was first described in 1990 as a basic leucine zipper-structured factor that binds to the interleukin (IL)-1-responsive element in the IL-6 promoter (37). C/EBPβ is highly expressed in myelomonocytic cells and macrophages (3841). Extracellular signals, including differentiation- or proliferation-inducing agents, hormones, cytokines and inflammatory substances, as well as bacterial and other microbial products can activate or inhibit C/EBPβ via distinct signal transduction pathways. The expression and/or activation of C/EBPβ is regulated by transcriptional mechanisms, mammalian target of rapamycin (mTOR)-mediated alternative translation, post-translational modifications and protein-protein interactions (35,42,43). Upon its activation, C/EBPβ induces or represses a variety of genes, including cytokines, chemokines and their receptors, other pro-inflammatory genes and pro-proliferative or differentiation-associated markers, as well as metabolic enzymes (34). C/EBPβ thereby affects associated cellular functions, including proliferation (40,42), differentiation (39,41,44), metabolic regulation (45,46) and orchestration of the immune response (4749). Furthermore, C/EBPβ is implicated in the pathogenesis of various common diseases, including cancer, hyper-/hypo-inflammation and bacterial/viral infections (34,50). Expression and activation of C/EBPβ induces the production of monocyte chemotactic protein-1 (MCP-1) (5154), a member of the C-C motif chemokine ligand-2, which induces leukocyte migration to inflamed tissues and organs (55,56). In addition, MCP-1 is secreted by primary breast tumors and stimulates migration of MSCs to tumor lesions (57). MCP-1, stromal cell-derived factor-1, macrophage inflammatory protein-1α and monocyte chemotactic protein-3 are the most widely reported MSC homing factors (5861). Stem cell therapy relies on the appropriate homing and engraftment capacity of stem cells (62). Therefore, SG-2 can be used for in vivo studies of TGF-β-dependent anti-inflammation and stem cell homing.

Abnormal intensity of Smad-mediated TGF-β/BMP signals in MSCs is associated with various human diseases, including bone and immune disorders, fibrosis, and cancer progression or metastasis. However, the detection of therapeutic molecular targets for these diseases in the TGF-β/BMP signaling pathways has not been successful as most studies have been performed in vitro. The present study established MSC lines, including TGF-β-responsive SG-2, BMP-responsive SG-3, and TGF-β/BMP-unresponsive SG-5 cells, which can be traced by GFP fluorescence after transplantation into in vivo experimental models. These cell lines can be used to explore how TGF-β/BMP-induced Smad-mediated signals affect proliferation and differentiation of MSCs in vivo, providing insight into various human diseases, including bone and immune disorders, fibrosis and cancer progression or metastasis.

Acknowledgments

The present study was supported in part by KAKENHI grants-in-aid from the Japan Society for the Promotion of Science (grant nos. 25893221 to S.S., 25463053 to N.C., 26893248 to J.Y., 26462932 to H.K. and 26670852 to A.I.), a grant-in-aid for Strategic Medical Science Research Centre from the Ministry of Education, Culture, Sports, Science and Technology of Japan (2010–2014), a grant from the Keiryokai Research Foundation (no. 120 to S.S., 2013) and the Fund of Academic Society for Research in Otolaryngology (Kansai Medical University, Hirakata, Japan).

References

1 

Prockop DJ: Marrow stromal cells as stem cells for nonhematopoietic tissues. Science. 276:71–74. 1997. View Article : Google Scholar : PubMed/NCBI

2 

Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca JD, Moorman MA, Simonetti DW, Craig S and Marshak DR: Multilineage potential of adult human mesenchymal stem cells. Science. 284:143–147. 1999. View Article : Google Scholar : PubMed/NCBI

3 

Docheva D, Popov C, Mutschler W and Schieker M: Human mesenchymal stem cells in contact with their environment: Surface characteristics and the integrin system. J Cell Mol Med. 11:21–38. 2007. View Article : Google Scholar : PubMed/NCBI

4 

Baksh D, Song L and Tuan RS: Adult mesenchymal stem cells: Characterization, differentiation and application in cell and gene therapy. J Cell Mol Med. 8:301–316. 2004. View Article : Google Scholar : PubMed/NCBI

5 

Kassem M, Abdallah BM and Saeed H: Osteoblastic cells: Differentiation and transdifferentiation. Arch Biochem Biophys. 473:183–187. 2008. View Article : Google Scholar : PubMed/NCBI

6 

Jones E and Yang X: Mesenchymal stem cells and bone regeneration: Current status. Injury. 42:562–568. 2011. View Article : Google Scholar : PubMed/NCBI

7 

Proff P and Römer P: The molecular mechanism behind bone remodelling: A review. Clin Oral Investig. 13:355–362. 2009. View Article : Google Scholar : PubMed/NCBI

8 

Lazar-Karsten P, Dorn I, Meyer G, Lindner U, Driller B and Schlenke P: The influence of extracellular matrix proteins and mesenchymal stem cells on erythropoietic cell maturation. Vox Sang. 101:65–76. 2011. View Article : Google Scholar

9 

Yew TL, Chang MC, Hsu YT, Weng WH, Tsai CC, Chiu FY, Hung SC and He FY: Efficient expansion of mesenchymal stem cells from mouse bone marrow under hypoxic conditions. J Tissue Eng Regen Med. 7:984–993. 2013. View Article : Google Scholar

10 

Weiss A and Attisano L: The TGF beta superfamily signaling pathway. Wiley Interdiscip Rev Dev Biol. 2:47–63. 2013. View Article : Google Scholar : PubMed/NCBI

11 

Miyazawa K, Shinozaki M, Hara T, Furuya T and Miyazono K: Two major Smad pathways in TGF-beta superfamily signalling. Genes Cells. 7:1191–1204. 2002. View Article : Google Scholar : PubMed/NCBI

12 

Heldin CH, Landström M and Moustakas A: Mechanism of TGF-beta signaling to growth arrest, apoptosis and epithelial-mesenchymal transition. Curr Opin Cell Biol. 21:166–176. 2009. View Article : Google Scholar : PubMed/NCBI

13 

Meulmeester E and Ten Dijke P: The dynamic roles of TGF-β in cancer. J Pathol. 223:205–218. 2011. View Article : Google Scholar

14 

Imamura T, Oshima Y and Hikita A: Regulation of TGF-β family signaling by ubiquitination and de ubiquitination. J Biochem. 154:481–489. 2013. View Article : Google Scholar : PubMed/NCBI

15 

Herhaus L and Sapkota GP: The emerging roles of deubiquitylating enzymes (DUBs) in the TGFβ and BMP pathways. Cell Signal. 26:2186–2192. 2014. View Article : Google Scholar : PubMed/NCBI

16 

Lee SY, Lee JH, Kim JY, Bae YC, Suh KT and Jung JS: BMP2 increases adipogenic differentiation in the presence of dexamethasone, which is inhibited by the treatment of TNF-α in human adipose tissue-derived stromal cells. Cell Physiol Biochem. 34:1339–1350. 2014. View Article : Google Scholar

17 

Pountos I, Georgouli T, Henshaw K, Bird H, Jones E and Giannoudis PV: The effect of bone morphogenetic protein-2, bone morphogenetic protein-7, parathyroid hormone and platelet-derived growth factor on the proliferation and osteogenic differentiation of mesenchymal stem cells derived from osteoporotic bone. J Orthop Trauma. 24:552–556. 2010. View Article : Google Scholar : PubMed/NCBI

18 

Yokota J, Chosa N, Sawada S, Okubo N, Takahashi N, Hasegawa T, Kondo H and Ishisaki A: PDGF-induced PI3K-mediated signal enhances TGF-β-induced osteogenic differentiation of human mesenchymal stem cells in the TGF-β-activated MEK-dependent manner. Int J Mol Med. 33:534–542. 2014.PubMed/NCBI

19 

Ray P and Chapman SC: Cytoskeletal reorganaization drives mesenchymal condensation and regulates downstream molecular signaling. PLoS One. 10:e01347022015. View Article : Google Scholar

20 

Sengle G, Carlberg V, Tufa SF, Charbonneau NL, Smaldone S, Carlson EJ, Ramirez F, Keene DR and Sakai LY: Abnormal activation of BMP signaling causes myopathy in Fbn2 null mice. PLoS Genet. 11:e10053402015. View Article : Google Scholar : PubMed/NCBI

21 

Kramman R and Humphreys BD: Kidney pericytes: Roles in regeneration and fibrosis. Semin Nephrol. 34:374–383. 2014. View Article : Google Scholar

22 

Touboul C, Vidal F, Pasquier J, Lis R and Rafii A: Role of mesenchymal cells in the natural history of ovarian cancer: A review. J Transl Med. 12:2712014. View Article : Google Scholar : PubMed/NCBI

23 

Wise AF and Ricardo SD: Mesenchymal stem cells in kidney inflammation and repair. Nephrology (Carlton). 17:1–10. 2012. View Article : Google Scholar

24 

Okabe M, Ikawa M, Kominami K, Nakanishi T and Nishimune Y: 'Green mice' as a source of ubiquitous green cells. FEBS Lett. 407:313–319. 1997. View Article : Google Scholar : PubMed/NCBI

25 

Aomatsu E, Takahashi N, Sawada S, Okubo N, Hasegawa T, Taira M, Miura H, Ishisaki A and Chosa N: Novel SCRG1/BST1 axis regulates self-renewal, migration and osteogenic differentiation potential in mesenchymal stem cells. Sci Rep. 4:36522014. View Article : Google Scholar

26 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T)) Method. Methods. 25:402–408. 2001. View Article : Google Scholar

27 

Gee CJ and Harris H: Tumorigenicity of cells transformed by Simian virus 40 and of hybrids between such cells and normal diploid cells. J Cell Sci. 36:223–240. 1979.PubMed/NCBI

28 

Howell N: Suppression of transformation and tumorigenicity in interspecies hybrids of human SV40-transformed and mouse 3T3 cell lines. Cytogenet Cell Genet. 34:215–229. 1982. View Article : Google Scholar : PubMed/NCBI

29 

Kahn P, Topp WC and Shin S: Tumorigenicity of SV40-transformed human and monkey cells in immunodeficient mice. Virology. 126:348–360. 1983. View Article : Google Scholar : PubMed/NCBI

30 

Nitta M, Katabuchi H, Ohtake H, Tashiro H, Yamaizumi M and Okamura H: Characterization and tumorigenicity of human ovarian surface epithelial cells immortalized by SV40 large T antigen. Gynecol Oncol. 81:10–17. 2001. View Article : Google Scholar : PubMed/NCBI

31 

Kang MK and Park NH: Extension of cell life span using exogenous telomerase. Methods Mol Biol. 371:151–165. 2007. View Article : Google Scholar : PubMed/NCBI

32 

Lee MK, Hande MP and Sabapathy K: Ectopic mTERT expression in mouse embryonic stem cells does not affect differentiation but confers resistance to differentiation- and stress-induced p53-dependent apoptosis. J Cell Sci. 118:819–829. 2005. View Article : Google Scholar : PubMed/NCBI

33 

Abraham S, Sweet T, Khalili K, Sawaya BE and Amini S: Evidence for activation of the TGF-beta1 promoter by C/EBP beta and its modulation by Smads. J Interferon Cytokine Res. 29:1–7. 2009. View Article : Google Scholar

34 

Ramji DP and Foka P: CCAAT/enhancer-binding proteins: Structure, function and regulation. Biochem J. 365:561–575. 2002. View Article : Google Scholar : PubMed/NCBI

35 

Zahnow CA: CCAAT/enhancer-binding protein beta: Its role in breast cancer and associations with receptor tyrosine kinases. Expert Rev Mol Med. 11:e122009. View Article : Google Scholar : PubMed/NCBI

36 

Lekstrom-Himes J and Xanthopoulos KG: Biological role of the CCAAT/enhancer-binding protein family of transcription factors. J Biol Chem. 273:28545–28548. 1998. View Article : Google Scholar : PubMed/NCBI

37 

Akira S, Isshiki H, Sugita T, Tanabe O, Kinoshita S, Nishio Y, Nakajima T, Hirano T and Kishimoto T: A nuclear factor for IL-6 expression (NF-IL6) is a member of a C/EBP family. EMBO J. 9:1897–1906. 1990.PubMed/NCBI

38 

Williams SC, Cantwell CA and Johnson PF: A family of C/EBP-related proteins capable of forming covalently linked leucine zipper dimers in vitro. Genes Dev. 5:1553–1567. 1991. View Article : Google Scholar : PubMed/NCBI

39 

Katz S, Kowenz-Leutz E, Müller C, Meese K, Ness SA and Leutz A: The NF-M transcription factor is related to C/EBP beta and plays a role in signal transduction, differentiation and leukemogenesis of avian myelomonocytic cells. EMBO J. 12:1321–1332. 1993.PubMed/NCBI

40 

Haas SC, Huber R, Gutsch R, Kandemir JD, Cappello C, Krauter J, Duyster J, Ganser A and Brand K: ITD- and FL-induced FLT3 signal transduction leads to increased C/EBP beta-LIP expression and LIP/LAP ratio by different signalling modules. Br J Haematol. 148:777–790. 2010. View Article : Google Scholar

41 

Gutsch R, Kandemir JD, Pietsch D, Cappello C, Meyer J, Simanowski K, Huber R and Brand K: CCAAT/enhancer-binding protein beta inhibits proliferation in monocytic cells by affecting the retinoblastoma protein/E2F/cyclin E pathway but is not directly required for macrophage morphology. J Biol Chem. 286:22716–22729. 2011. View Article : Google Scholar : PubMed/NCBI

42 

Nerlov C: The C/EBP family of transcription factors: A paradigm for interaction between gene expression and proliferation control. Trends Cell Biol. 17:318–324. 2007. View Article : Google Scholar : PubMed/NCBI

43 

Tsukada J, Yoshida Y, Kominato Y and Auron PE: The CCAAT/enhancer (C/EBP) family of basic-leucine zipper (bZIP) transcription factors is a multifaceted highly-regulated system for gene regulation. Cytokine. 54:6–19. 2011. View Article : Google Scholar : PubMed/NCBI

44 

Pham TH, Langmann S, Schwarzfischer L, El Chartouni C, Lichtinger M, Klug M, Krause SW and Rehli M: CCAAT enhancer-binding protein beta regulates constitutive gene expression during late stages of monocyte to macrophage differentiation. J Biol Chem. 282:21924–21933. 2007. View Article : Google Scholar : PubMed/NCBI

45 

Liu S, Croniger C, Arizmendi C, Harada-Shiba M, Ren J, Poli V, Hanson RW and Friedman JE: Hypoglycemia and impaired hepatic glucose production in mice with a deletion of the C/EBP beta gene. J Clin Invest. 103:207–213. 1999. View Article : Google Scholar : PubMed/NCBI

46 

Croniger CM, Millward C, Yang J, Kawai Y, Arinze IJ, Liu S, Harada-Shiba M, Chakravarty K, Friedman JE, Poli V and Hanson RW: Mice with a deletion in the gene for CCAAT/enhancer-binding protein beta have an attenuated response to cAMP and impaired carbohydrate metabolism. J Biol Chem. 276:629–638. 2001. View Article : Google Scholar

47 

Poli V: The role of C/EBP isoforms in the control of inflammatory and native immunity functions. J Biol Chem. 273:29279–29282. 1998. View Article : Google Scholar : PubMed/NCBI

48 

Zwergal A, Quirling M, Saugel B, Huth KC, Sydlik C, Poli V, Neumeier D, Ziegler-Heitbrock HW and Brand K: C/EBP beta blocks p65 phosphorylation and thereby NF-kappa B-mediated transcription in TNF-tolerant cells. J Immunol. 177:665–672. 2006. View Article : Google Scholar : PubMed/NCBI

49 

Cappello C, Zwergal A, Kanclerski S, Haas SC, Kandemir JD, Huber R, Page S and Brand K: C/EBP beta enhances NF-kappaB-associated signalling by reducing the level of IkappaB-alpha. Cell Signal. 21:1918–1924. 2009. View Article : Google Scholar : PubMed/NCBI

50 

Liu Y, Nonne macher MR and Wigdahl B: CCAAT/enhancer-binding proteins and the pathogenesis of retrovirus infection. Future Microbiol. 4:299–321. 2009. View Article : Google Scholar : PubMed/NCBI

51 

Spooner CJ, Guo X, Johnson PF and Schwartz RC: Differential roles of C/EBP beta regulatory domains in specifying MCP-1 and IL-6 transcription. Mol Immunol. 44:1384–1392. 2007. View Article : Google Scholar

52 

Abraham M, Shapiro S, Karni A, Weiner HL and Miller A: Gelatinases (MMP-2 and MMP-9) are preferentially expressed by Th1 vs. Th2 cells. J Neuroimmunol. 163:157–164. 2005. View Article : Google Scholar : PubMed/NCBI

53 

Guo J, Zhang H, Xiao J, Wu J, Ye Y, Li Z, Zou Y and Li X: Monocyte chemotactic protein-1 promotes the myocardial homing of mesenchymal stem cells in dilated cardiomyopathy. Int J Mol Sci. 14:8164–8178. 2013. View Article : Google Scholar : PubMed/NCBI

54 

Belema-Bedada F, Uchida S, Martire A, Kostin S and Braun T: Efficient homing of multipotent adult mesenchymal stem cells depends on FROUNT-mediated clustering of CCR2. Cell Stem Cell. 2:566–575. 2008. View Article : Google Scholar : PubMed/NCBI

55 

Gerard C and Rollins BJ: Chemokines and disease. Nat Immunol. 2:108–115. 2001. View Article : Google Scholar : PubMed/NCBI

56 

Rot A and von Andrian UH: Chemokines in innate and adaptive host defense: Basic chemokinese grammar for immune cells. Annu Rev Immunol. 22:891–928. 2004. View Article : Google Scholar : PubMed/NCBI

57 

Dwyer RM, Potter-Beirne SM, Harrington KA, Lowery AJ, Hennessy E, Murphy JM, Barry FP, O'Brien T and Kerin MJ: Monocyte chemotactic protein-1 secreted by primary breast tumors stimulates migration of mesenchymal stem cells. Clin Cancer Res. 13:5020–5027. 2007. View Article : Google Scholar : PubMed/NCBI

58 

Schenk S, Mal N, Finan A, Zhang M, Kiedrowski M, Popovic Z, McCarthy PM and Penn MS: Monocyte chemotactic protein-3 is a myocardial mesenchymal stem cell homing factor. Stem Cells. 25:245–251. 2007. View Article : Google Scholar

59 

Zhou YL, Zhang HF, Li XL, Di RM, Yao WM, Li DF, Feng JL, Huang J, Cao KJ and Fu M: Increased stromal-cell-derived factor 1 enhances the homing of bone marrow derived mesenchymal stem cells in dilated cardiomyopathy in rats. Chin Med J (Engl). 123:3282–3287. 2010.

60 

Zhuang Y, Chen X, Xu M, Zhang LY and Xiang F: Chemokine stromal cell-derived factor 1/CXCL12 increases homing of mesenchymal stem cells to injured myocardium and neovascularization following myocardial infarction. Chin Med J (Engl). 122:183–187. 2009.

61 

Abbott JD, Huang Y, Liu D, Hickey R, Krause DS and Giordano FJ: Stromal cell-derived factor-1 alpha plays a critical role in stem cell recruitment to the heart after myocardial infarction but is not sufficient to induce homing in the absence of injury. Circulation. 110:3300–3305. 2004. View Article : Google Scholar : PubMed/NCBI

62 

Mozid AM, Arnous S, Sammut EC and Mathur A: Stem cell therapy for heart diseases. Br Med Bull. 98:143–159. 2011. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2016
Volume 13 Issue 3

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Sawada S, Chosa N, Takizawa N, Yokota J, Igarashi Y, Tomoda K, Kondo H, Yaegashi T and Ishisaki A: Establishment of mesenchymal stem cell lines derived from the bone marrow of green fluorescent protein‑transgenic mice exhibiting a diversity in intracellular transforming growth factor‑β and bone morphogenetic protein signaling. Mol Med Rep 13: 2023-2031, 2016
APA
Sawada, S., Chosa, N., Takizawa, N., Yokota, J., Igarashi, Y., Tomoda, K. ... Ishisaki, A. (2016). Establishment of mesenchymal stem cell lines derived from the bone marrow of green fluorescent protein‑transgenic mice exhibiting a diversity in intracellular transforming growth factor‑β and bone morphogenetic protein signaling. Molecular Medicine Reports, 13, 2023-2031. https://doi.org/10.3892/mmr.2016.4794
MLA
Sawada, S., Chosa, N., Takizawa, N., Yokota, J., Igarashi, Y., Tomoda, K., Kondo, H., Yaegashi, T., Ishisaki, A."Establishment of mesenchymal stem cell lines derived from the bone marrow of green fluorescent protein‑transgenic mice exhibiting a diversity in intracellular transforming growth factor‑β and bone morphogenetic protein signaling". Molecular Medicine Reports 13.3 (2016): 2023-2031.
Chicago
Sawada, S., Chosa, N., Takizawa, N., Yokota, J., Igarashi, Y., Tomoda, K., Kondo, H., Yaegashi, T., Ishisaki, A."Establishment of mesenchymal stem cell lines derived from the bone marrow of green fluorescent protein‑transgenic mice exhibiting a diversity in intracellular transforming growth factor‑β and bone morphogenetic protein signaling". Molecular Medicine Reports 13, no. 3 (2016): 2023-2031. https://doi.org/10.3892/mmr.2016.4794