Cell-cell adhesion through N-cadherin enhances VCAM-1 expression via PDGFRβ in a ligand-independent manner in mesenchymal stem cells

  • Authors:
    • Emiko Aomatsu
    • Naoyuki Chosa
    • Soko Nishihira
    • Yoshiki Sugiyama
    • Hiroyuki Miura
    • Akira Ishisaki
  • View Affiliations

  • Published online on: December 27, 2013     https://doi.org/10.3892/ijmm.2013.1607
  • Pages: 565-572
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Cell-cell adhesions induce various intracellular signals through hierarchical and synergistic molecular interactions. Recently, we demonstrated that a high cell density induces the expression of vascular cell adhesion molecule-1 (VCAM-1) through the nuclear factor-κB (NF-κB) pathway in human bone marrow-derived mesenchymal stem cells (MSCs). However, the specific molecules that activated the NF-κB pathway were not determined. In the present study, in experiments with receptor tyrosine kinase inhibitors, VCAM-1 expression was completely suppressed by platelet-derived growth factor (PDGF) receptor (PDGFR) inhibitors. In addition, VCAM-1 expression was significantly suppressed by knockdown with PDGFRβ siRNA, but not with PDGFRα siRNA. However, VCAM-1 expression did not increase following treatment with PDGF. The overexpression of N-cadherin, a structural molecule in adherence junctions in MSCs, promoted VCAM-1 expression and induced the marked phosphorylation of the intracellular signaling factor, Src. In addition, VCAM-1 expression and Src phosphorylation were reduced by the overexpression of a dominant negative mutant of N-cadherin. These results suggest that cell-cell adhesion, through N-cadherin, enhances the expression of VCAM-1 via PDGFRβ and the activation of Src in a ligand-independent manner in MSCs.

Introduction

Mesenchymal stem cells (MSCs) are non-hematopoietic stromal cells, which retain the ability to self-renew and differentiate into mesenchymal cells, such as osteoblasts, adipocytes, chondrocytes and skeletal muscle cells (1). Various surface markers, including CD49a, CD73, CD105, vascular cell adhesion molecule-1 (VCAM-1)/CD106, CD140b, CD146, CD271, mesenchymal stromal cell antigen-1 (MSCA-1) and STRO-1, have been used alone or in combination to identify and isolate human MSCs (hMSCs) (28). STRO-1 is a popular MSC marker and is often used in combination with VCAM-1 for MSC isolation. A number of studies have analyzed the changes in surface marker expression caused by prolonged cultivation, and have reported the attenuated expression of several markers, including VCAM-1 (9,10). Cell adhesion molecules, such as VCAM-1 mediate the interaction of MSCs with endothelial cells (ECs), which is essential for MSC homing. Therefore, the reduction in VCAM-1 expression during expansion in culture may be related to the decreased homing ability of senescent MSCs. Mabuchi et al recently demonstrated that clones of hMSCs retaining high rates of colony-forming unit fibroblasts (CFU-Fs) expressing the surface markers, CD271/low affinity nerve growth factor receptor (LNGFR), thymocyte antigen-1 (Thy-1) and VCAM-1, exhibited robust multilineage differentiation and self-renewal ability (11). An in-depth investigation of MSC markers has made it possible to identify and purify MSCs; for instance, anti-CD49a antibody is useful for identifying hMSCs (12,13). Notably, rapidly expanding clones of hMSCs express high levels of CD49a and VCAM-1 and are highly migratory (11). These results suggest that VCAM-1 can be used as a marker for enriching migratory, multipotent and proliferative cells from culture-expanded MSCs.

Cadherins are members of a family of transmembrane proteins involved in mediating homophilic adhesion in a Ca2+-dependent manner. These proteins are major components of adherence junctions (AJs) in cells. E-cadherin is the main cadherin in the AJs of epithelial cells, whereas other cadherins, including N-cadherin, P-cadherin, R-cadherin and VE-cadherin form AJs in other cell types. Although fibroblasts express a number of different cadherins, including P-cadherin, R-cadherin, OB-cadherin and fat-like cadherins (14,15), N-cadherin is the predominant cadherin expressed by these cells (14,16). N-cadherin-mediated AJs are of great importance in connective tissue physiology and are critical for the regulation of cell attachment and migration (17), wound healing (18), metastatic potential (19) and embryonic development (20,21), as well as the differentiation and formation of numerous specialized tissues, including fibrous connective tissues (2226). N-cadherin is considered to be the key factor in directing cell-cell interactions during mesenchymal condensation, a process mediated by surface contact that results in the aggregation of progenitor cells (2730). Studies have indicated that the expression of deletion mutant forms of N-cadherin, which lack either the extracellular homotypic interaction domains or the intracellular β-catenin binding site, results in decreased cellular condensation and impaired chondrogenesis (31,32). These findings suggest that both extracellular homotypic interaction and intracellular interaction with the catenin complex are essential for proper N-cadherin signaling (33).

In a recent study of ours, we found that the expression level of VCAM-1/CD106 was markedly upregulated in human bone marrow-derived MSCs, UE7T-13 cells, under conditions of high cell density (34). The high cell density-induced expression of VCAM-1 was markedly suppressed by nuclear factor-κB (NF-κB) signaling-related protein kinase inhibitors, such as the IκB kinase-2 (IKK-2) inhibitor VI, a phosphoinositide 3-kinase (PI3K) inhibitor, an Src inhibitor and a protein kinase C (PKC) inhibitor. Therefore, the high cell density-induced VCAM-1 expression was regulated by the NF-κB pathway in human bone marrow-derived MSCs. However, the identities of the inducing factor(s) that activate the NF-κB pathway in MSC cell-cell adhesion are not yet clear. Herein, we demonstrate that cell-cell adhesion by N-cadherin activates the NF-κB pathway via platelet-derived growth factor (PDGF) receptor (PDGFR)-β in a ligand-independent manner.

Materials and methods

Reagents

The SCADS inhibitor kit, including various protein kinase inhibitors was generously supplied by the Screening Committee of Anticancer Drugs supported by a Grant-in-Aid for Scientific Research on Priority Area ‘Cancer’ from the Ministry of Education, Culture, Sports, Science and Technology, Japan.

Cell culture

Human bone marrow-derived MSCs, UE7T-13 cells, the life span of which was prolonged by infection with a retrovirus encoding human papillomavirus E7 and human telomerase reverse transcriptase (hTERT) (35,36), were purchased from the Health Science Research Resources Bank (JCRB no. 1154, Japan Health Sciences Foundation, Japan). UE7T-13 cells were cultured in Dulbecco’s modified Eagle’s medium (DMEM; Sigma-Aldrich, St. Louis, MO, USA) supplemented with 10% fetal bovine serum (FBS; HyClone, Logan, UT, USA) at 37ºC in a humidified incubator with an atmosphere of 5% CO2 (3.0×103 cells/cm2, ‘low’ cell density; 1.0×105 cells/cm2, ‘high’ cell density).

RNA isolation and quantitative RT-PCR (qRT-PCR)

Total RNA from low, medium and high cell density cultured UE7T-13 cells was isolated using Isogen reagent (Nippon Gene, Tokyo, Japan) according to the manufacturer’s instructions. First-strand cDNA was synthesized from total RNA by using the PrimeScript RT reagent kit (Takara, Kyoto, Japan). qRT-PCR was performed on a Thermal Cycler Dice Real-time System (Takara) using SYBR Premix Ex Taq II (Takara) with specific oligonucleotide primers (presented in Table I). The mRNA expression levels for VCAM-1, PDGFRα and PDGFRβ were normalized to those obtained for glyceraldehyde adenosine-phosphate dehydrogenase (GAPDH), and the relative expression levels were shown as fold-increase or decrease relative to the control.

Table I

Sequences of the qRT-PCR primers used in this study.

Table I

Sequences of the qRT-PCR primers used in this study.

GenePrimer sequencesLocation (bp)Product size (bp)
VCAM-1Foward: 5′-CGAAAGGCCCAGTTGAAGGA-3′2086–2226141
Reverse: 5′-GAGCACGAGAAGCTCAGGAGAAA-3′
PDGFRαForward: 5′-GTGCGAAGACTGAGCCAGATTG-3′5708–5828121
Reverse: 5′-CGATAAACAGAATGCTTGAGCTGTG-3′
PDGFRβForward: 5′-TGCCTTGCCAGCACTAACATTC-3′4769–4908140
Reverse: 5′-CCAGAGTGTGATGTGTGATCTGGA-3′
GAPDHForward: 5′-GCACCGTCAAGGCTGAGAAC-3′248–389142
Reverse: 5′-ATGGTGGTGAAGACGCCAGT-3′

[i] VCAM-1, vascular cell adhesion molecule-1; PDGFRα, platelet-derived growth factor receptor α; PDGFRβ, platelet-derived growth factor receptor β; GAPDH, glyceraldehyde adenosine-phosphate dehydrogenase.

Western blot analysis

The UE7T-13 cells were washed twice with ice-cold PBS and then lysed in RIPA 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 of the samples was measured using the BCA reagent (Pierce Biotechnology, Rockford, IL, USA). Samples containing equal amounts of protein were separated by 12.5% SDS-polyacrylamide gel electrophoresis (SDS-PAGE) and transferred onto a polyvinylidene difluoride (PVDF) membrane (Millipore, Billerica, MA, USA). After being blocked with 5% non-fat dry milk in T-TBS (50 mM Tris-HCl, pH 7.2, 150 mM NaCl and 0.1% Tween-20), the membrane was incubated with primary anti-Src (Cell Signaling, Technologies, Beverly, MA, USA) and anti-phospho-Src (p-Src, Cell Signaling) antibodies, using an anti-β-actin (clone C4, Santa Cruz Biotechnology Inc., Santa Cruz, CA, USA) antibody as a loading control for normalization. The blots were subsequently incubated with horseradish peroxidase-conjugated secondary antibody and developed using chemiluminescence with the Amersham ECL western blot analysis system (GE Healthcare Biosciences, Pittsburgh, PA, USA). The detected blots were photographed using the photo image detection system CL-Cube L (Tohoku Electronic Industrial Co., Ltd., Sendai, Japan) and densitometrically measured using ImageJ software (version 1.44). Data are expressed as the ratio of phosphorylated to total molecular bands.

siRNA transfection for PDGFRs

Three sets of Stealth siRNA oligonucleotide duplexes targeting PDGFRα and PDGFRβ were designed using the online BLOCK-iT RNAi Designer software (Invitrogen, Carlsbad, CA, USA). Sequences of the siRNA oligonucleotide duplexes are listed in Table II. UE7T-13 cells were seeded in 24-well culture plates without antibiotic selection at a density of 2.0×104 cells/well, 24 h before siRNA transfection. Subsequently, transcriptional knockdown was performed by transfection of the cells with siRNA oligonucleotide duplexes at a final concentration of 20 nM in DMEM using Lipofectamine RNAiMAX (Invitrogen) for 24 h according to the manufacturer’s instructions. The effects of RNAi knockdown of target genes were assayed by qRT-PCR. Stealth siRNA Negative Control medium GC Duplex (Invitrogen) was also included as a control for sequence independent RNAi knockdown.

Table II

Oligonucleotide sequences of the Stealth siRNA duplex.

Table II

Oligonucleotide sequences of the Stealth siRNA duplex.

GenesiRNA no.RNA duplex sequence
PDGFRα−387Sense: 5′-AAUGAAAGCUGGCAGAGGAUUAGGC-3′
Antisense: 5′-GCCUAAUCCUCUGCCAGCUUUCAUU-3′
−751Sense: 5′-UAUAAUGGCAGAAUCAUCAUCCUCC-3′
Antisense: 5′-GGAGGAUGAUGAUUCUGCCAUUAUA-3′
−843Sense: 5′-UUAAAGCCCUGUCUGCUGUCGUAGG-3′
Antisense: 5′-CCUACGACAGCAGACAGGGCUUUAA-3′
PDGFRβ−803Sense: 5′-CAAAGAUGUAGAGCCGUUUCCGCUC-3′
Antisense: 5′-CCUACGACAGCAGACAGGGCUUUAA-3′
−1061Sense: 5′-CAUAGUAGGCAUCAGAAUCCACCUC-3′
Antisense: 5′-GAGGUGGAUUCUGAUGCCUACUAUG-3′
−1506Sense: 5′-UUGUCUUUGAACCACAGGACAGUGG-3′
Antisense: 5′-CCACUGUCCUGUGGUUCAAAGACAA-3′

[i] PDGFRα, platelet-derived growth factor receptor α; PDGFRβ, platelet-derived growth factor receptor β.

Overexpression of N-cadherin

cDNA encoding full-length N-cadherin (Met1-Asp906) or an N-cadherin deletion mutant lacking the C-terminal intercellular domain (Met1-Trp745) was amplified by PCR. The cDNA was then subcloned into the pcDNA/V5/GW/D-TOPO vector (Invitrogen) using the pcDNA Gateway Directional TOPO Expression kit (Invitrogen) according to the manufacturer’s instructions. The overexpression vectors, containing full-length N-cadherin (pCDH2-full) or the deletion mutant (pCDH2-Δ), were transfected into the UE7T-13 cells using Lipofectamine LTX (Invitrogen) for 48 h according to the manufacturer’s instructions. The effects of N-cadherin overexpression were assayed by flow cytometric analysis. The transfected UE7T-13 cells were stripped with cell dissociation buffer (Invitrogen) and washed with PBS containing 0.5% FBS and 2 mM EDTA. The cells (1.0×105) were then incubated with an anti-N-cadherin antibody (clone GC-4; Abcam, Cambridge, MA, USA) for 1 h at room temperature. The cells were then incubated with phycoerythrin-conjugated secondary antibody for 1 h. Image acquisition was performed using the EPICS XL ADC System (Beckman Coulter, Brea, CA, USA).

Cell proliferation assay

Cell proliferation was analyzed using a colorimetric assay for the quantification of the cleavage of the tetrazolium salt WST-1 (Roche Applied Science, Basel, Switzerland) by mitochondrial dehydrogenases in viable cells. The dye formed can be quantified using a spectrophotometer and directly correlates with the number of metabolically active cells in the culture. The cells were grown in 96-well plates (Nunclon®; Sigma-Aldrich) for 1, 3 and 5 days treated with or without 10 ng/ml of recombinant human PDGF-BB (Acris Antibodies, San Diego, CA, USA). After each incubation period, the cells were incubated for a further 1 h at 37ºC with 100 μl medium containing 10 μl WST-1 reagent. The samples were shaken for 1 min, and absorbance was measured at 450 nm using a MPR-A4i microplate reader (Tosoh Corp., Tokyo, Japan).

Statistical analysis

All experiments were repeated at least 3 times. Representative images or data are shown. Data are presented as the means ± standard deviation (SD). Differences between averages and percentages between the control and test samples were statistically analyzed using paired two-tailed Student’s t-tests. Values of p<0.05 were considered to indicate statistically significant differences.

Results

Effect of receptor tyrosine kinase (RTK) inhibitors on high cell density-induced VCAM-1 expression

In a recent study, we demonstrated that high cell density induces VCAM-1 expression through the NF-κB pathway in UE7T-13 cells (34) and that the expression of VCAM-1 in high cell density culture was significantly inhibited by treatment with IKK-2 inhibitor VI. In the present study, we investigated the effects of RTK inhibitors on VCAM-1 expression. As illustrated in Fig. 1, the high cell density-induced VCAM-1 expression decreased in a dose-dependent manner upon the addition of PDGFR inhibitors (SU11652 and PDGFR tyrosine kinase inhibitor IV) to the cell culture. However, the addition of the following RTK inhibitors did not decrease VCAM-1 expression: vascular endothelial growth factor receptor (VEGFR) inhibitors (SU1498 and VEGFR kinase inhibitor I), an insulin-like growth factor-1 receptor (IGF-1R) inhibitor (AG1024), an epidermal growth factor receptor (EGFR) inhibitor (AG1478), fibroblast growth factor receptor (FGFR) inhibitors (SU4984 and SU5402), and a colony stimulating factor-1 receptor (CSF1R) tyrosine kinase inhibitor (Fig. 1A). Similarly, the addition of a receptor type serine/threonine kinase inhibitor, such as a transforming growth factor-β receptor (TGFβR) inhibitor (SB431542) did not decrease VCAM-1 expression (Fig. 1A).

Role of PDGFRβ on high cell density-induced VCAM-1 expression

In order to determine the role of PDGFRs in the high cell density-induced intracellular signal transduction culminating into VCAM-1 expression in UE7T-13 cells, we investigated the effects of knocking down PDGFRα and PDGFRβ expression on VCAM-1 expression. As shown in Fig. 2, VCAM-1 mRNA expression in high cell density culture was reduced effectively by transfection with siRNA targeting PDGFRβ, but not siRNA targeting PDGFRα. In order to investigate whether PDGFRβ mediates the intracellular signal for VCAM-1 expression in a ligand-dependent manner, PDGF-BB was added to the UE7T-13 cell culture at high and low cell densities. As shown in Fig. 3A, PDGF-BB did not affect the level of VCAM-1 mRNA expression in the MSCs at low or high cell densities. By contrast, exogenously added recombinant human PDGF-BB markedly accelerated the proliferation of UE7T-13 cells, suggesting that functional PDGFR is present at the cell surface in UE7T-13 cells (Fig. 3B).

Role of N-cadherin in high cell density-induced VCAM-1 expression

A previous study revealed that a mechanostress-induced intracellular signal is mediated by an RTK in ECs, even if the receptor was not stimulated with any ligand: vascular endothelial (VE)-cadherin forms a mechanosensory complex with VEGFR2 in ECs in a ligand-independent manner (37). Therefore, in this study, we investigated the role of N-cadherin as a major structural molecule in AJs in MSCs on high cell density-induced VCAM-1 expression. UE7T-13 cells were transfected with vectors expressing either full-length N-cadherin or a truncated version lacking the intracellular domain. The expression of N-cadherin, which localizes on the cell surface, was confirmed by flow cytometry (Fig. 4A), and then the cells were seeded at a high density. Under conditions of high density, the overexpression of full-length N-cadherin markedly increased the level of VCAM-1 mRNA expression. By contrast, the overexpression of the truncated N-cadherin markedly reduced the high cell density-induced VCAM-1 expression (Fig. 4B). These results indicate that, in MSCs, the high cell density induction of VCAM-1 expression is mediated by N-cadherin.

Signaling pathway for high cell density-induced VCAM-1 expression

In ECs, even if VEGFR2 is not stimulated with VEGF, the VE-cadherin-VEGFR2 complex responds to a subset of endothelial shear stresses, resulting in the activation of NF-κB-mediated Src signaling that upregulates VCAM-1 expression (37). Therefore, in this study, we investigated the phosphorylation status of Src by western blot analysis. As shown in Fig. 5, the phosphorylation levels of Src in UE7T-13 cells were markedly upregulated by the overexpression of full-length N-cadherin (pCDH2-full). On the other hand, the overexpression of the truncated mutant (pCDH2-Δ) had no effect compared with the control vector-transfected cells (pCTRL). This result strongly suggests that the phosphorylation of Src through the intracellular domain of N-cadherin plays an important role in the regulation of VCAM-1 expression by cell-cell adhesion.

Discussion

VCAM-1 is an important marker for enriching migratory, multipotent and proliferative cells from culture-expanded MSCs (11). In addition, it has been shown that VCAM-1 is upregulated in bone marrow-derived MSCs cultured to overconfluency (38). However, a comprehensive analysis of the types of adhesion molecules that play important roles in cell-cell contact between MSCs has not been performed. In a recent study, we demonstrated that VCAM-1 expression was markedly upregulated in human bone marrow-derived MSCs at high cell density through the activation of the NF-κB pathway (34).

Notably, a previous study revealed that a mechanostress-induced intracellular signal is mediated by an RTK in ECs in a ligand-independent manner: VE-cadherin forms a mechanosensory complex with VEGFR2 in ECs that responds to a subset of endothelial shear stresses, resulting in the activation of NF-κB-mediated Src signaling, upregulating VCAM-1 expression, even when VEGFR2 is not stimulated with VEGF (37). Therefore, we hypothesized that an RTK may similarly mediate the cell-cell contact-induced signal responsible for upregulating VCAM-1 expression in MSCs. When we evaluated the effects of various RTK inhibitors on high cell density-induced VCAM-1 expression, we found that VCAM-1 expression was decreased by PDGFR inhibitors (SU11652 and PDGFR tyrosine kinase inhibitor IV) in a dose-dependent manner (Fig. 1). However, other RTK inhibitors, e.g., inhibitors of VEGFR, IGF1R, EGFR and FGFR, as well as a receptor type serine/threonine kinase TGFβR inhibitor, did not decrease VCAM-1 expression (Fig. 1A). In general, PDGFR is activated by PDGF as its ligand. PDGF has four isoforms (A–D) that form homo- or heterodimers, such as PDGF-AA, PDGF-AB and PDGF-BB (39). Of these, PDGF-BB exhibits the strongest activity (39) and has been approved by the FDA for the treatment of patients with bone defects in the oral and maxillofacial regions (4043). PDGF-BB is mainly produced by platelets and has been implicated in tissue repair (fracture repair) (39). PDGFR has two isoforms (α and β) that also form homo- or heterodimers, such as PDGFRα/α, α/β and β/β (39). PDGFRα is reportedly expressed in MSCs and osteoblast progenitor cells, and PDGFRα-positive cells exhibit a high osteoblastic differentiation capacity (44). In MSCs, a recent study provided evidence that PDGF-BB promotes PDGFα-positive cell migration into artificial bones without inhibiting osteoblastogenesis (45). In this study, high cell density-induced VCAM-1 expression was significantly suppressed by the knockdown of PDGFRβ (Fig. 2). Of note, the PDGF-BB-induced PDGFR activation did not affect the level of VCAM-1 mRNA expression in MSCs at both low and high cell densities (Fig. 3A), although PDGF-BB clearly upregulated MSC proliferation (Fig. 3B). These results strongly suggest that the high cell density-induced intracellular signal to upregulate VCAM-1 expression in MSCs was mediated by PDGFRβ in a PDGF-BB-independent manner.

In MSCs, VE-cadherin does not exhibit dominant expression, although N-cadherin does (46). Therefore, it may be possible that the formation of a mechanosensory interaction occurs between N-cadherin and PDGFRβ in high cell density cultures and that this interaction may then activate the NF-κB-mediated signaling pathway, thus upregulating VCAM-1 expression. In support of this hypothesis, in the present study, the overexpression of full-length N-cadherin in UE7T-13 cells markedly increased high cell density-induced VCAM-1 expression, whereas the overexpression of truncated N-cadherin lacking its intracellular domain suppressed the high cell density-induced VCAM-1 expression (Fig. 4). As, in ECs, the VE-cadherin-VEGFR2 complex responds to a subset of endothelial shear stresses by activating NF-κB-mediated Src signaling (37). Therefore, we examined the phosphorylation status of Src by western blot analysis and found that the phosphorylation levels of Src in UE7T-13 cells were enhanced at high cell density (Fig. 5A). Src phosphorylation levels in the UE7T-13 cells were also upregulated by the overexpression of full-length N-cadherin (Fig. 5B). By contrast, the overexpression of the truncated mutant did not affect Src phosphorylation. In addition, in a previous study, we demonstrated that high cell density-induced VCAM-1 expression was decreased by the Src inhibitor, PP2 analogue (34). These results strongly suggest that the phosphorylation of Src through the intracellular domain of N-cadherin plays an important role in the upregulation of VCAM-1 by cell-cell adhesion.

In conclusion, the data presented in our study demonstrate that cell-cell adhesion induced by high cell density through N-cadherin enhances the expression of VCAM-1 via PDGFRβ in a ligand-independent manner in human bone marrow-derived MSCs. These findings may eventually lead to the development of new, MSC-based clinical therapies in regenerative medicine.

Acknowledgements

This study was supported in part by JSPS KAKENHI Grant nos. 25463053 to N.C., 23592896 to A.I.; the Open Research Project from the Ministry of Education, Culture, Sports, Science and Technology of Japan, 2008–2012; and Grant-in-Aid for Strategic Medical Science Research Centre from the Ministry of Education, Culture, Sports, Science and Technology of Japan, 2010–2014.

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 

Aslan H, Zilberman Y, Kandel L, et al: Osteogenic differentiation of noncultured immunoisolated bone marrow-derived CD105+ cells. Stem Cells. 7:1728–1737. 2006. View Article : Google Scholar : PubMed/NCBI

3 

Battula VL, Treml S, Bareiss PM, et al: Isolation of functionally distinct mesenchymal stem cell subsets using antibodies against CD56, CD271, and mesenchymal stem cell antigen-1. Haematologica. 94:173–184. 2009. View Article : Google Scholar : PubMed/NCBI

4 

Boiret N, Rapatel C, Veyrat-Masson R, et al: Characterization of nonexpanded mesenchymal progenitor cells from normal adult human bone marrow. Exp Hematol. 33:219–225. 2005. View Article : Google Scholar : PubMed/NCBI

5 

Bühring HJ, Battula VL, Treml S, et al: Novel markers for the prospective isolation of human MSC. Ann NY Acad Sci. 1106:262–271. 2007.PubMed/NCBI

6 

Gronthos S, Zannettino AC, Hay SJ, et al: Molecular and cellular characterisation of highly purified stromal stem cells derived from human bone marrow. J Cell Sci. 116:1827–1835. 2003. View Article : Google Scholar : PubMed/NCBI

7 

Quirici N, Soligo D, Bossolasco P, et al: Isolation of bone marrow mesenchymal stem cells by anti-nerve growth factor receptor antibodies. Exp Hematol. 30:783–791. 2002. View Article : Google Scholar : PubMed/NCBI

8 

Sacchetti B, Funari A, Michienzi S, et al: Self-renewing osteoprogenitors in bone marrow sinusoids can organize a hematopoietic microenvironment. Cell. 131:324–336. 2007. View Article : Google Scholar : PubMed/NCBI

9 

Honczarenko M, Le Y, Swierkowski M, et al: Human bone marrow stromal cells express a distinct set of biologically functional chemokine receptors. Stem Cells. 24:1030–1041. 2006. View Article : Google Scholar : PubMed/NCBI

10 

Wagner W, Horn P, Castoldi M, et al: Replicative senescence of mesenchymal stem cells: a continuous and organized process. PLoS One. 3:e22132008. View Article : Google Scholar : PubMed/NCBI

11 

Mabuchi Y, Morikawa S, Harada S, et al: LNGFR(+)THY-1(+)VCAM-1(hi+) cells reveal functionally distinct subpopulations in mesenchymal stem cells. Stem Cell Reports. 1:152–165. 2013.

12 

Deschaseaux F, Gindraux F, Saadi R, et al: Direct selection of human bone marrow mesenchymal stem cells using an anti-CD49a antibody reveals their CD45med, low phenotype. Br J Haematol. 122:506–517. 2003. View Article : Google Scholar : PubMed/NCBI

13 

Jones EA, English A, Kinsey SE, et al: Optimization of a flow cytometry-based protocol for detection and phenotypic characterization of multipotent mesenchymal stromal cells from human bone marrow. Cytometry B Clin Cytom. 70:391–399. 2006. View Article : Google Scholar : PubMed/NCBI

14 

Matsuyoshi N and Imamura S: Multiple cadherins are expressed in human fibroblasts. Biochem Biophys Res Commun. 235:355–358. 1997. View Article : Google Scholar : PubMed/NCBI

15 

Simonneau L, Kitagawa M, Suzuki S and Thiery JP: Cadherin 11 expression marks the mesenchymal phenotype: towards new functions for cadherins? Cell Adhes Commun. 3:115–130. 1995. View Article : Google Scholar : PubMed/NCBI

16 

Hatta K and Takeichi M: Expression of N-cadherin adhesion molecules associated with early morphogenetic events in chick development. Nature. 320:447–449. 1986. View Article : Google Scholar : PubMed/NCBI

17 

Akitaya T and Bronner-Fraser M: Expression of cell adhesion molecules during initiation and cessation of neural crest cell migration. Dev Dyn. 194:12–20. 1992. View Article : Google Scholar : PubMed/NCBI

18 

De Wever O, Westbroek W, Verloes A, et al: Critical role of N-cadherin in myofibroblast invasion and migration in vitro stimulated by colon-cancer-cell-derived TGF-beta or wounding. J Cell Sci. 117:4691–4703. 2004.

19 

Kashima T, Nakamura K, Kawaguchi J, et al: Overexpression of cadherins suppresses pulmonary metastasis of osteosarcoma in vivo. Int J Cancer. 104:147–154. 2003. View Article : Google Scholar : PubMed/NCBI

20 

Radice GL, Rayburn H, Matsunami H, et al: Developmental defects in mouse embryos lacking N-cadherin. Dev Biol. 181:64–78. 1997. View Article : Google Scholar : PubMed/NCBI

21 

García-Castro MI, Vielmetter E and Bronner-Fraser M: N-Cadherin, a cell adhesion molecule involved in establishment of embryonic left-right asymmetry. Science. 288:1047–1051. 2000.PubMed/NCBI

22 

Hinz B, Pittet P, Smith-Clerc J, et al: Myofibroblast development is characterized by specific cell-cell adherens junctions. Mol Biol Cell. 15:4310–4320. 2004. View Article : Google Scholar : PubMed/NCBI

23 

Tomasek JJ, Gabbiani G, Hinz B, et al: Myofibroblasts and mechano-regulation of connective tissue remodelling. Nat Rev Mol Cell Biol. 3:349–363. 2002. View Article : Google Scholar : PubMed/NCBI

24 

Marthiens V, Gavard J, Lambert M and Mège RM: Cadherin-based cell adhesion in neuromuscular development. Biol Cell. 94:315–326. 2002. View Article : Google Scholar : PubMed/NCBI

25 

Krauss RS, Cole F, Gaio U, et al: Close encounters: regulation of vertebrate skeletal myogenesis by cell-cell contact. J Cell Sci. 118:2355–2362. 2005. View Article : Google Scholar : PubMed/NCBI

26 

Marie PJ: Role of N-cadherin in bone formation. J Cell Physiol. 190:297–305. 2002. View Article : Google Scholar : PubMed/NCBI

27 

Ahrens PB, Solursh M and Reiter RS: Stage-related capacity for limb chondrogenesis in cell culture. Dev Biol. 60:69–82. 1977. View Article : Google Scholar : PubMed/NCBI

28 

San Antonio JD and Tuan RS: Chondrogenesis of limb bud mesenchyme in vitro: stimulation by cations. Dev Biol. 115:313–324. 1986.PubMed/NCBI

29 

Oberlender SA and Tuan RS: Spatiotemporal profile of N-cadherin expression in the developing limb mesenchyme. Cell Adhes Commun. 2:521–537. 1994. View Article : Google Scholar : PubMed/NCBI

30 

Tavella S, Raffo P, Tacchetti C, et al: N-CAM and N-cadherin expression during in vitro chondrogenesis. Exp Cell Res. 215:354–362. 1994. View Article : Google Scholar : PubMed/NCBI

31 

DeLise AM and Tuan RS: Alterations in the spatiotemporal expression pattern and function of N-cadherin inhibit cellular condensation and chondrogenesis of limb mesenchymal cells in vitro. J Cell Biochem. 87:342–359. 2002. View Article : Google Scholar : PubMed/NCBI

32 

Delise AM and Tuan RS: Analysis of N-cadherin function in limb mesenchymal chondrogenesis in vitro. Dev Dyn. 225:195–204. 2002. View Article : Google Scholar : PubMed/NCBI

33 

Tuan RS: Cellular signaling in developmental chondrogenesis: N-cadherin, Wnts, and BMP-2. J Bone Joint Surg Am. 85:137–141. 2003.PubMed/NCBI

34 

Nishihira S, Okubo N, Takahashi N, et al: High-cell density-induced VCAM-1 expression inhibits the migratory ability of mesenchymal stem cells. Cell Biol Int. 35:475–481. 2011. View Article : Google Scholar : PubMed/NCBI

35 

Mori T, Kiyono T, Imabayashi H, et al: Combination of hTERT and bmi-1, E6, or E7 induces prolongation of the life span of bone marrow stromal cells from an elderly donor without affecting their neurogenic potential. Mol Cell Biol. 25:5183–5195. 2005. View Article : Google Scholar : PubMed/NCBI

36 

Shimomura T, Yoshida Y, Sakabe T, et al: Hepatic differentiation of human bone marrow-derived UE7T-13 cells: Effects of cytokines and CCN family gene expression. Hepatol Res. 37:1068–1079. 2007. View Article : Google Scholar : PubMed/NCBI

37 

Liu Y, Sweet DT, Irani-Tehrani M, et al: Shc coordinates signals from intercellular junctions and integrins to regulate flow-induced inflammation. J Cell Biol. 182:185–196. 2008. View Article : Google Scholar : PubMed/NCBI

38 

Lee RH, Seo MJ, Pulin AA, et al: The CD34-like protein PODXL and alpha6-integrin (CD49f) identify early progenitor MSCs with increased clonogenicity and migration to infarcted heart in mice. Blood. 113:816–826. 2009. View Article : Google Scholar : PubMed/NCBI

39 

Hollinger JO, Hart CE, Hirsch SN, et al: Recombinant human platelet-derived growth factor: biology and clinical applications. J Bone Joint Surg Am. 90:48–54. 2008. View Article : Google Scholar

40 

Jayakumar A, Rajababu P, Rohini S, et al: Multi-centre, randomized clinical trial on the efficacy and safety of recombinant human platelet-derived growth factor with β-tricalcium phosphate in human intra-osseous periodontal defects. J Clin Periodontol. 38:163–172. 2011.PubMed/NCBI

41 

Ridgway HK, Mellonig JT and Cochran DL: Human histologic and clinical evaluation of recombinant human platelet-derived growth factor and beta-tricalcium phosphate for the treatment of periodontal intraosseous defects. Int J Periodontics Restorative Dent. 28:171–179. 2008.

42 

Nevins M, Giannobile WV, McGuire MK, et al: Platelet-derived growth factor stimulates bone fill and rate of attachment level gain: results of a large multicenter randomized controlled trial. J Periodontol. 76:2205–2215. 2005. View Article : Google Scholar

43 

McGuire MK, Kao RT, Nevins M and Lynch SE: rhPDGF-BB promotes healing of periodontal defects: 24-month clinical and radiographic observations. Int J Periodontics Restorative Dent. 26:223–231. 2006.PubMed/NCBI

44 

Morikawa S, Mabuchi Y, Kubota Y, et al: Prospective identification, isolation, and systemic transplantation of multipotent mesenchymal stem cells in murine bone marrow. J Exp Med. 206:2483–2496. 2009. View Article : Google Scholar : PubMed/NCBI

45 

Yoshida S, Iwasaki R, Kawana H, et al: PDGFBB promotes PDGFRα-positive cell migration into artificial bone in vivo. Biochem Biophys Res Commun. 421:785–789. 2012.

46 

Wuchter P, Boda-Heggemann J, Straub BK, et al: Processus and recessus adhaerentes: giant adherens cell junction systems connect and attract human mesenchymal stem cells. Cell Tissue Res. 328:499–514. 2007. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

2014-March
Volume 33 Issue 3

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Aomatsu E, Chosa N, Nishihira S, Sugiyama Y, Miura H and Ishisaki A: Cell-cell adhesion through N-cadherin enhances VCAM-1 expression via PDGFRβ in a ligand-independent manner in mesenchymal stem cells. Int J Mol Med 33: 565-572, 2014
APA
Aomatsu, E., Chosa, N., Nishihira, S., Sugiyama, Y., Miura, H., & Ishisaki, A. (2014). Cell-cell adhesion through N-cadherin enhances VCAM-1 expression via PDGFRβ in a ligand-independent manner in mesenchymal stem cells. International Journal of Molecular Medicine, 33, 565-572. https://doi.org/10.3892/ijmm.2013.1607
MLA
Aomatsu, E., Chosa, N., Nishihira, S., Sugiyama, Y., Miura, H., Ishisaki, A."Cell-cell adhesion through N-cadherin enhances VCAM-1 expression via PDGFRβ in a ligand-independent manner in mesenchymal stem cells". International Journal of Molecular Medicine 33.3 (2014): 565-572.
Chicago
Aomatsu, E., Chosa, N., Nishihira, S., Sugiyama, Y., Miura, H., Ishisaki, A."Cell-cell adhesion through N-cadherin enhances VCAM-1 expression via PDGFRβ in a ligand-independent manner in mesenchymal stem cells". International Journal of Molecular Medicine 33, no. 3 (2014): 565-572. https://doi.org/10.3892/ijmm.2013.1607