Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Connective-Tissue Growth Factor (CTGF/CCN2) Induces Astrogenesis and Fibronectin Expression of Embryonic Neural Cells In Vitro

  • Fabio A. Mendes ,

    Contributed equally to this work with: Fabio A. Mendes, Juliana M. Coelho Aguiar

    Affiliation Instituto de Ciências Biomédicas, Programa de Biologia Celular e do Desenvolvimento, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil

  • Juliana M. Coelho Aguiar ,

    Contributed equally to this work with: Fabio A. Mendes, Juliana M. Coelho Aguiar

    Affiliation Instituto de Ciências Biomédicas, Programa de Biologia Celular e do Desenvolvimento, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil

  • Suzana A. Kahn,

    Affiliations Instituto de Ciências Biomédicas, Programa de Biologia Celular e do Desenvolvimento, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil, Inserm, UMR894, Team Glial Plasticity, University Paris Descartes, Paris, France

  • Alice H. Reis,

    Affiliation Instituto de Ciências Biomédicas, Programa de Biologia Celular e do Desenvolvimento, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil

  • Luiz Gustavo Dubois,

    Affiliation Instituto Estadual do Cérebro Paulo Niemeyer (IEC), Rio de Janeiro, RJ, Brazil

  • Luciana Ferreira Romão,

    Affiliation Universidade Federal do Rio de Janeiro, pólo Xerém, Rio de Janeiro, RJ, Brazil

  • Lais S. S. Ferreira,

    Affiliation Instituto de Ciências Biomédicas, Programa de Biologia Celular e do Desenvolvimento, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil

  • Hervé Chneiweiss,

    Affiliation Inserm, UMR894, Team Glial Plasticity, University Paris Descartes, Paris, France

  • Vivaldo Moura Neto,

    Affiliation Instituto Estadual do Cérebro Paulo Niemeyer (IEC), Rio de Janeiro, RJ, Brazil

  • José G. Abreu

    garciajr@icb.ufrj.br

    Affiliation Instituto de Ciências Biomédicas, Programa de Biologia Celular e do Desenvolvimento, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil

Abstract

Connective-tissue growth factor (CTGF) is a modular secreted protein implicated in multiple cellular events such as chondrogenesis, skeletogenesis, angiogenesis and wound healing. CTGF contains four different structural modules. This modular organization is characteristic of members of the CCN family. The acronym was derived from the first three members discovered, cysteine-rich 61 (CYR61), CTGF and nephroblastoma overexpressed (NOV). CTGF is implicated as a mediator of important cell processes such as adhesion, migration, proliferation and differentiation. Extensive data have shown that CTGF interacts particularly with the TGFβ, WNT and MAPK signaling pathways. The capacity of CTGF to interact with different growth factors lends it an important role during early and late development, especially in the anterior region of the embryo. ctgf knockout mice have several cranio-facial defects, and the skeletal system is also greatly affected due to an impairment of the vascular-system development during chondrogenesis. This study, for the first time, indicated that CTGF is a potent inductor of gliogenesis during development. Our results showed that in vitro addition of recombinant CTGF protein to an embryonic mouse neural precursor cell culture increased the number of GFAP- and GFAP/Nestin-positive cells. Surprisingly, CTGF also increased the number of Sox2-positive cells. Moreover, this induction seemed not to involve cell proliferation. In addition, exogenous CTGF activated p44/42 but not p38 or JNK MAPK signaling, and increased the expression and deposition of the fibronectin extracellular matrix protein. Finally, CTGF was also able to induce GFAP as well as Nestin expression in a human malignant glioma stem cell line, suggesting a possible role in the differentiation process of gliomas. These results implicate ctgf as a key gene for astrogenesis during development, and suggest that its mechanism may involve activation of p44/42 MAPK signaling. Additionally, CTGF-induced differentiation of glioblastoma stem cells into a less-tumorigenic state could increase the chances of successful intervention, since differentiated cells are more vulnerable to cancer treatments.

Introduction

The connective-tissue growth factor (CTGF/CCN2) was first described in 1991 by Bradham and coworkers, as a novel growth factor secreted by human umbilical-vein endothelial cells (HUVECs), with high chemotactic and mitogenic activity [12]. Since then, an increasing amount of data has revealed several functions of CTGF, which seem to be related to its modular architecture [310].

The amino-acid sequence of CTGF is characterized by the presence of an N-terminal secretory signal and four distinct domains: a domain that shows homology to the IGF-binding protein, a von Willebrand factor type C repeat (VWC) or chordin-like cysteine-rich (CR) domain, a thrombospondin type 1 repeat (TSP-1) and a cysteine knot in the carboxy-terminal domain. This modular architecture is also found in NOV, CYR61, WISP1 and WISP3, which together with CTGF form the so-called CCN family of secreted factors [11].

The characteristic modular architecture of CTGF is responsible for the modulation of growth-factor signaling by binding and sequestering different ligands. Abreu et al. (2002) demonstrated that CTGF directly binds BMP4 and TGFβ-1 through its CR domain. CTGF can antagonize BMP4 signaling by blocking its binding to receptors, and has an opposite effect on TGFβ-1, enhancing its binding to receptors. CTGF cooperates with TGFβ-1 in most TGFβ responses, such as induction of the expression of extracellular matrix components, fibroblast proliferation, wound repair and fibrotic disorders [1216]. A SMAD binding element was found within the ctgf promoter, indicating that ctgf is a direct target of TGFβs [14]. Indeed, fibroblasts produce high levels of CTGF mRNA and protein after activation with TGFβ-1 [3].

Mercurio et al. (2004) demonstrated that CTGF also modulates both canonical and non-canonical WNT signaling. CTGF interacts with the WNT pathway through a domain distinct from the TGFβ/BMP interacting domain. In this case, the CT domain of CTGF is important in the interaction with all four EGF repeats of the WNT co-receptor LRP6, and can also bind weakly to the WNT receptor Frizzled. CTGF competes with WNT for binding to LRP6 and Frizzled, blocking the access of WNT to its receptor. Moreover, CTGF was able to inhibit convergent extension movements of Xenopus laevis animal caps, also indicating its ability to control the non-canonical WNT pathway; however, the mechanism is not completely understood since the non-canonical pathways comprise several β-catenin-independent cascades [7]. In addition, CTGF may also be a downstream target of WNT/β-catenin signaling. Luo et al. (2004) showed that CTGF is upregulated at the early stages of BMP-9 and WNT-3a stimulation of osteoblast differentiation, and that CTGF expression is β-catenin-dependent [17].

CTGF expression can also be controlled by mitogen-activated protein (MAP) kinase pathways [1820]. The three major MAP kinase pathways, extracellular signal-regulated kinases (ERK, also known as p42/44 MAPK), p38 and jun N-terminal kinase (JNK) serve as mediators for a variety of signaling molecules and growth factors [2123]. ERK is classically activated via the sequential activation of Ras G proteins, Raf kinases and MEK1 and 2. MEKs, in turn, phosphorylate and activate ERK1 and 2. The minimal consensus phosphorylation motif in MAPK substrates is [Ser/Thr]-Pro. Activated MAPKs can phosphorylate a large number of intracellular substrates, leading to diverse cellular outcomes. ERK activates CTGF expression in different cell types including fibroblasts, mesangial cells and osteoblast cells [1820, 24].

During the development of the nervous system, all cell types in the cortex are generated following a distinct temporal program. Neurons are differentiated with a peak at embryonic day 14 (E14); the astrocytes appear second, with a peak at postnatal day 2 (P2); and then the oligodendrocytes with a peak at P14. Since both neurons and astrocytes are derived from the same progenitor, a neurogenic to gliogenic switch has to occur during the temporal differentiation program. BMPs and Notch are both important signals involved in astrogenesis. BMP-2, together with gliogenic cytokines, causes the formation of a SMAD/STAT complex that activates transcription of gliogenic genes. Notch signaling can also promote astrogenesis in the presence of an active JAK-STAT signal. The ERK pathway also has an important role during the neural-cell differentiation program. During early stages, ERK pathway directly represses gp130 and the JAK-STAT pathway, promoting neurogenesis. However, some data have pointed to a pro-gliogenic role for the ERK pathway. Rajan and McKay (1998) showed that CNTF activates the JAK-STAT and MAPK pathways in multipotent stem cells of the central nervous system, to instruct them to an astrocytic fate. Moreover, Lee et al. (2004) showed that the cytokine erythropoietin is able to promote astrogenesis, activating the ERK pathway, and Nakanishi et al. (2007) showed that IL-6 and LIF also activate the ERK pathway, together with JAK-STAT during astrogenesis [25].

Considering that several signaling pathways modulated by CTGF are important in nervous-system development, this study was undertaken to determine whether CTGF can be important for the differentiation program of central nervous system cells. In order to achieve this, we added recombinant CTGF protein [5] to a culture of mouse embryonic E14 hemisphere cells. Our results showed that the exogenous CTGF protein increased the number of glial fibrillary acidic protein (GFAP) and GFAP/Nestin-positive cells without changing the proliferation index. Surprisingly, CTGF was also able to increase the number of Sox2-positive cells. Moreover, the exogenous CTGF protein was able to induce phosphorylation of ERK proteins and also increased the expression and deposition of the extracellular matrix protein fibronectin. Finally, using human malignant glioma stem cells (OB1 cell line), we showed that CTGF treatment induced GFAP, CD133, Nestin and inhibited Sox2 expression, suggesting a possible role for CTGF not only during normal brain development but also in the glioma differentiation program.

Methods

Primary embryonic neural cell culture

Mice experiments were carried out according to the guidelines granted by the Animal Care and Use Ethic Committee (Comissão de Ética no Uso de Animais—CEUA) of the Federal University of Rio de Janeiro and were approved by this committee under the permission number DHE-ICB 015. The experiments were also approved by the Brazilian Ministry of Health Ethics Committee (CONEP No. 2340). The mice were euthanized in CO2 chamber followed by cervical dislocation. The primary cultures were established as previously described [2628]. Briefly, 14-day-old mouse embryo cerebral hemispheres were dissected in phosphate-buffered saline (PBS), stripped of meninges, minced and mechanically dissociated. Aggregates were removed by decantation and cells were obtained by centrifugation at 180 x g for 4 min. The pellet was resuspended in DMEM-F12 (Invitrogen) enriched with 2 nM glutamine and 14 mM sodium bicarbonate with 10% FBS (Invitrogen) and plated at a density of 5 x 106 cells in 100-mm cell culture dishes or of 2 x 105 cells in 24-well plates, with glass coverslips previously treated with poly-L-ornithine (1.5 μg/mL, PM 41000, Sigma). These cultures were kept in 37°C at 5%/95% CO2/H2O atmosphere for 5 days. The medium was changed 24 and 72 h after plating and on the last day, 30 min before processing for immunocytochemistry or total protein extraction. In all the experiments, 5nM CTGF was re-added in each medium change. The recombinant protein CTGF-Flag was obtained as described [5]. Xenopus laevis full-length Flag-CTGF from S2 cells was affinity purified with anti-Flag M2 affinity gel column and eluted with Flag peptide according to manufacturer’s directions (Sigma, St Louis, MO).

Human glioblastoma stem cell line

The cell line of non-adherent human glioblastoma stem cells (OB1) were established as described in Patru et al., 2010. OB1 were grown in 75 cm2 tissue culture flasks (2500–5000 cells/ cm2) in DMEM-F12, supplemented with B27, N2 and G5 (Invitrogen, France). All cultures were maintained at 37°C in humidified atmosphere with 5% CO2.

Immunocytochemistry

The cells were fixed with 4% paraformaldehyde for 15 min at room temperature. Fixed cultures were washed with PBS, permeabilized with 0.2% Triton X-100 in PBS for 3 min, and blocked for 1 h with 5% bovine serum albumin (BSA). Next, the cells were incubated with anti-BrdU (Sigma, 1:500), polyclonal anti-fibronectin (Sigma, 1:400) and anti-laminin (Sigma, 1:50), anti-Sox2 (Millipore, 1:100), anti-CD133 (Millipore, 1:20) antibodies or double-stained with polyclonal anti-GFAP (DAKP, 1:500) and monoclonal anti-Nestin (BD Biosciences 1:50) antibodies overnight at 4°C. Incubation with specific secondary Alexa Fluor 546 goat anti-mouse IgG (Molecular Probes 1:800), Alexa Fluor 488 goat anti-rabbit (Molecular Probes 1:400) or Alexa Fluor 546 goat anti-rat IgG (Molecular Probes 1:500) proceeded for 1 h at room temperature. Then, the cells were washed with PBS, stained with DAPI (Sigma), washed again and mounted. For negative controls, cells received similar treatment but the primary antibodies were omitted. The slides were observed in a Nikon TE 2000 inverted microscope. Images were captured using a CoolSNAP-Pro (Media Cybernetics) digital camera.

BrdU incorporation assay

5’-Bromo-2’-deoxyuridine (BrdU, Sigma) was administered in the culture, with or without CTGF, for de last 2 h of treatment, at a final concentration of 10 μM. After fixation with 4% paraformaldehyde and permeabilization with 0.4% Triton X-100, cell were treated with 2 N HCl, then neutralized with 0.1 M Na2B4O7. After PBS washes, the rat anti-BrdU antibody (1:500) was incubated. The Cy3-labeled goat anti-rat antibody was used as a secondary antibody. A negative control for the BrdU assay was performed by omitting the primary antibody during immunostaining.

[3H]-Thymidine incorporation assay

The cultured cells were seeded at 5x104 cells per well in DMEM-F12 with 10% FBS in 24-well plates. In the last 6 hours of CTGF treatment, a pulse of [3H]-thymidine was added. After the five days of CTGF treatment, the medium was carefully removed and 300 μL of ice-cold 10% thrichloroacetic acid was added. Cells were harvested and [3H]-thymidine incorporation was measured on a scintillation counter.

Cell viability assay

To measure the viability of the cells in culture we used the premixed WST-1 reagent (Clontech). This colorimetric assay is based on the cleavage of the tetrazolium salt WST-1 to a formazan-class dye by mitochondrial succinate-tetrazolium reductase in viable cells. 5x104 cells were plated in a 96-well plate previously treated with poly-L ornithine (1,5μg/mL, PM 41000, Sigma) and cultured in presence of recombinant CTGF protein for 24, 48, 72, 96 e 120 hours. After each 24 hours, the reagent was added according to manufactures protocol and the absorbance was read at 450nm.

Immunoblotting

The cultures were lysed in 50 μl of RIPA buffer (0.05 M Tris-HCl pH 7.4, 0.15 M NaCl, 1% NP-40, 2 mM EDTA, 1 mg/mL pepstatin, 1 mM PMSF, 1 mM NaF, 1 mM Na3VO4) 30 min after the last medium change. Protein concentration was measured by the Bradford method. Before loading, samples were mixed with sample buffer (200 mM DTT 4% SDS, 125 mM Tris pH 6.8, 20% glycerol and 0.02% bromophenol blue). Samples were heated for 5 min at 95°C and the proteins were separated in SDS-PAGE. Proteins were transferred to PVDF membranes (Amersham Biosciences) in transfer buffer (25 mM Tris, 192 mM glycine, 0.1% SDS and 20% methanol). Membranes were blocked in 5% BSA for phosphorylated proteins or in nonfat dry milk for all others, in TBSt (20 mM Tris pH 7.6, 137 mM NaCl and 0.1% Tween 20). Primary polyclonal antibodies anti-total p44/p42 MAPK (Cell Signaling 1:1000), anti-phosphorylated p44/p42 MAPK (Cell Signaling 1:1000), anti-phosphorylated p38 (Cell Signaling 1:1000), anti-total SAPK/JNK (Cell Signaling 1:1000), anti-phosphorylated SAPK/JNK (Cell Signaling 1:1000), anti-fibronectin (Sigma 1:1000), anti-laminin (Sigma 1:400), anti-tubulin (Sigma 1:1000), monoclonal anti-tubulin (Sigma 1:2000), monoclonal anti-β-tubulin (Covance, Eurogentec, Belgium), polyclonal anti-GFAP (Dakocytomation, France), polyclonal anti-OLIG-2 (R&D systems, France) and monoclonal anti-Sox2 (Neuromics, Acris, Germany) in blocking solution were incubated with the membranes overnight at 4°C, followed by incubation with Peroxidase-conjugated anti-rabbit IgG and peroxidase-conjugated anti-mouse IgG secondary antibodies (Amersham Biosciences 1:2000). The immunoblot reaction was developed with Super Signal West Pico chemiluminescent substrate (Pierce, USA) and exposed in Kodak X-OMAT film.

Densitometries and statistical analysis

Densitometric analyses of the immunoblots were performed using ImageQuant software. The ratio between phosphor-specific bands and their respective loading controls was calculated and the results were normalized to the respective control groups. Nonparametric statistical tests were performed (Student’s t test) with GraphPad PRISM software (GraphPad Software, Inc., San Diego, CA). The results were considered to be significant when P < 0.05.

Results

Recombinant CTGF protein increased the number of GFAP and Sox2-positive cells in vitro

In order to test the ability of CTGF to affect the differentiation program of neural progenitors during development, we dissociated cells from 14-day-old mouse embryo cerebral hemispheres and grew them in DMEM-F12 (Invitrogen) with 10% fetal bovine serum (FBS, Invitrogen) in the presence of 5 nM recombinant CTGF protein [5]. Cells were maintained in culture for 5 days and the medium was replaced 24 and 72 h after plating and 30 min before fixation. 5nM recombinant CTGF protein was added after each medium change. Control cultures were maintained in DMEM-F12 supplemented with 10% FBS. Then, cells were processed for double immunocytochemistry against GFAP and Nestin or against Sox2.

After 5 days of culture, it was possible to identify cells that stained only for GFAP, some that stained only for Nestin, others that double-stained for GFAP/Nestin, and a large number of unstained cells (Fig 1). Interestingly, almost twice as many Nestin-/GFAP+ cells were observed when the cells were grown in the presence of CTGF (Fig 1A and 1B and Nestin-/GFAP+ bars in Fig 1G, P = 0.0215), and six times as many cells double-stained for Nestin and GFAP (Fig 1A–1F and Nestin+/GFAP+ bars in Fig 1G, P = 0.0103). However, no changes were observed in the number of cells that stained only for Nestin (Fig 1C–1F and Nestin+/GFAP- bars in Fig 1G). In addition, we observed a 43% decrease in the percentage of cells that did not stain for GFAP or Nestin (Fig 1E and 1F and Nestin-/GFAP- bars in Fig 1G, P = 0.0277). Finally, the percentages of beta-III tubulin-positive cells were the same in both CTGF-treated and untreated cultures (data not shown). In order to check the effect of CTGF treatment on the number of neural progenitors we also stained CTGF-treated and untreated cells for Sox2. Surprisingly, CTGF was able to increase 20% the number of Sox2-positive cells (Fig 2).

thumbnail
Fig 1. Exogenous CTGF protein increases the number of GFAP-positive cells of a neural progenitor culture.

Double immunostaining showing GFAP (A and B green) and Nestin (C and D red) expression of untreated and CTGF-treated astrocytes. E and F show merged pictures of GFAP and Nestin immunostaining together with nuclei-DAPI staining. Scale bars 50 μm. G shows the percentage of cells that were unstained or stained with GFAP and/or Nestin. CTGF was able to increase the number of GFAP-positive cells and, more dramatically, the number of Nestin/GFAP-positive cells.

https://doi.org/10.1371/journal.pone.0133689.g001

thumbnail
Fig 2. Exogenous CTGF protein increases the number of Sox2-positive cells of a neural progenitor culture.

Immunostaining showing Sox2 (A and D) expression of untreated and CTGF-treated cells. C and F show merged pictures of Sox2 immunostaining together with nuclei-DAPI staining. Scale bars 10 μm. G shows the percentage of cells that were positive for Sox2.

https://doi.org/10.1371/journal.pone.0133689.g002

Recombinant CTGF did not affect the proliferation rate of neural cells in vitro

Since it has been shown that CTGF induces proliferation in different cell types, we asked if addition of recombinant CTGF in neural progenitor-cell cultures derived from 14-day-old embryo hemisphere-cell cultures cultivated for 5 days could affect the cell growth rate. As described above, 5x104 cells were plated in a 96-well plate and cultivated in the presence of 5 nM recombinant CTGF protein in DMEM-F12 with 10% FBS for 5 days. The medium containing CTGF was changed 24 and 72 h after plating and 30 min before the cells and controls were maintained in DMEM-F12 with 10% FBS. To test for changes in the proliferation index, we performed a WST-1 cell proliferation assay with these cells during the 5 days in culture. WST-1 reagent was added to the CTGF-treated cultures every 24 h during the 5 days of culture. The absorbance at 450 nm showed no differences in the proliferation index between untreated and CTGF-treated cells (S1 Fig). We also performed other proliferation assays on the 5th day in culture, including incorporation of 5-bromo-2-deoxyuridine (BrdU) (S2 Fig) and incorporation of [3H] thymidine (S2 Fig). Both assays indicated no changes in the proliferation rate.

Recombinant CTGF increased expression and deposition of fibronectin

CTGF is also known to induce expression of the extracellular matrix protein fibronectin. In order to investigate if CTGF is able to increase the expression and deposition of fibronectin and laminin, two major extracellular matrix proteins essential for neural-cell development, we immunostained progenitor cell cultures derived from 14-day-old embryo hemispheres and cultivated for 5 days in the presence of 5 nM recombinant CTGF protein. The medium was replaced every 24 and 72 h after plating and 30 min before fixing and processing for immunocytochemistry against fibronectin and laminin. We also performed immunoblot assays (as described in Methods) using total protein extracts of these neural-cell cultures after 5 days of the same treatments. Tubulin was used as a loading control. The CTGF increased the deposition of fibronectin in the extracellular matrix (Fig 3A and 3B) and increased the expression almost six fold (Fig 3E, P = 0.0171). Differently from the untreated condition, fibronectin showed a fibrillary organization after CTGF treatment (Fig 3A and 3B). However, CTGF seemed not to modify the expression or deposition of laminin (Fig 3C, 3D and 3E). Treated or untreated cells had the same organization of laminin in their extracellular matrix.

thumbnail
Fig 3. Recombinant CTGF increases expression and deposition of fibronectin.

Immunostaining and immunoblotting for fibronectin and laminin. (A-D) Untreated or CTGF-treated progenitor neural cells immunostained for fibronectin (A, B) and laminin (C, D). (E) Immunoblotting for fibronectin and laminin of untreated (lane 1) or CTGF-treated neural progenitor cells (lane 2). CTGF incubation of neural progenitor cells for 120 h increased the expression of fibronectin threefold (compare untreated versus CTGF bars of graph in E). Scale bar 50 μm.

https://doi.org/10.1371/journal.pone.0133689.g003

CTGF increased phosphorylation of p44/42 MAPK (ERK1/2) but not p38 MAPK or p54/46 MAPK (SAPK/JNK)

Because p44/42 MAPK (ERK1/2) signaling is one of the principal pathways activated by CTGF and given that it has been shown to be involved in p44/42 MAPK (ERK1/2) activation and GFAP expression, we decided to test whether CTGF can modulate MAPK signaling effectors, phospho44/42 (ERK1/2), p38 and p54/46 (SAPK/JNK). Cell extracts from neural progenitor-cell cultures derived from 14-day-old mouse embryo hemispheres were separated on SDS-PAGE gels, and the proteins were transferred to PVDF membranes and probed with anti-phospho p44/42 MAPK, anti-phospho p38 MAPK and anti-phospho p54/46 SAPK/JNK antibodies. Tubulin was used as a loading control. As observed by densitometry, we demonstrated that addition of 5 nM recombinant CTGF protein in these cultures for 5 days dramatically increased (almost 7-fold) the phosphorylation of p44/42 MAPK (ERK1/2) level (Fig 4, compare lanes 1 and 2). This difference was highly significant by the Mann-Whitney test (P = 0.0181). In contrast, the same treatment did not produce any significant change in p38 MAPK or p54/46 SAPK/JNK phosphorylation (Fig 5, compare lanes 1 and 2).

thumbnail
Fig 4. CTGF increases activation of p44/42 MAPK (ERK1/2).

Immunoblot for phosphorylated p44/42 MAPK (ERK 1 and 2) of untreated or CTGF-treated neural progenitor cells. CTGF increased the expression of phosphorylated p44/42 MAPK almost sevenfold (compare lanes 1 and 2 and the graph of the densitometry). As a loading control we used total p44/42 MAPK and tubulin.

https://doi.org/10.1371/journal.pone.0133689.g004

thumbnail
Fig 5. CTGF does not increase p38 MAPK or p54/46 SAPK/JNK MAPK pathways.

Immunoblot for phosphorylated p38 MAPK and phosphorylated p46/p54 MAPK of untreated or CTGF-treated neural progenitor cells. CTGF did not change the levels of expression of these proteins (compare lanes 1 and 2). As a loading control we used total p46/p54 MAPK and tubulin.

https://doi.org/10.1371/journal.pone.0133689.g005

Recombinant CTGF increased GFAP and reduced Sox2 expression in human cancer stem cells

To investigate whether CTGF also plays an important role in neural cell differentiation in a human model, we treated OB1 cells [29], a malignant glioma stem cell line, with recombinant CTGF protein. Both 1 nM and 5 nM CTGF induced an astrocytic differentiation of these cells, as GFAP expression increased in a dose-dependent manner (Fig 6), 40% with 1 nM CTGF and 80% with 5 nM CTGF. Sox2 expression decreased in a dose-dependent manner (Fig 6), 30% with 1 nM CTGF and 60% with 5 nM CTGF. As expected, no significant difference was observed in β-tubulin III expression levels. These differences were highly significant by the Mann-Whitney test, P < 0.001 and P = 0.005 respectively. We also performed immunocytochemistry against the glial marker GFAP and the markers of neural progenitors, CD133 and Nestin. 5nM CTGF was able to increase approximately 2.0 fold the intensity of GFAP, 2.7 fold the intensity of CD133 and 1.6 fold the intensity of Nestin immunostaining (Fig 7).

thumbnail
Fig 6. Exogenous CTGF protein increases GFAP and reduces Sox2 expression in human cancer stem cells.

Immunoblotting showing GFAP, β-tubulin III and Sox2 expression in untreated and 1 nM and 5 nM CTGF-treated cells. CTGF incubation of human glioma stem cells for 120 h increased GFAP and reduced Sox2 expression in a dose-dependent manner. Alpha-tubulin was used as a loading control. **P < 0.001 and *P < 0.005.

https://doi.org/10.1371/journal.pone.0133689.g006

thumbnail
Fig 7. Exogenous CTGF protein increases the number of GFAP-, CD133- and Nestin-positive cells of human cancer stem cells.

Immunostaining showing GFAP (A and D), CD133 (H and K) and Nestin (O and R) expression of untreated and CTGF-treated astrocytes. C and F, J and M and Q and T show merged pictures of GFAP, CD133 and Nestin immunostaining together with nuclei-DAPI staining respectively. Scale bars 25 μm. G, N and U show the fluorescence intensity of cells that were stained with GFAP, CD133 and Nestin respectively.

https://doi.org/10.1371/journal.pone.0133689.g007

Discussion

In this study, we associated CTGF with astrocyte differentiation for the first time. Recombinant CTGF protein added during the first steps of the embryonic neural-cell differentiation process in vitro was able to increase the number of GFAP-positive cells and to activate p44/42 MAPK (ERK1/2). CTGF did not activate other MAPK pathways such as p38 or JNK. CTGF was also able to increase fibronectin expression and deposition in the extracellular matrix of these newly differentiated astrocytes. Together, these findings suggest that CTGF induces astrocytic differentiation and modulates the MAPK signaling pathway in the first steps of neural cells differentiation.

The CTGF protein contains different modules that capacitate CTGF to interact with a large number of cytokines, growth factors and different extracellular matrix proteins, modulating different signaling pathways [5, 7, 10, 30]. CTGF was first implicated in the promotion of cell adhesion and mitogenesis in both fibroblasts and endothelial cells and in the stimulation of cell migration in fibroblasts [1, 12, 31]. CTGF has also been extensively implicated in the differentiation of chondrocytes and osteoblasts [3237] as well as in the differentiation of diverse cell types such as myofibroblasts, adipocytes, endothelial cells and mammary epithelial cells [4, 31, 3840]. Very little is known about the roles of CTGF in neural development or neural-cell differentiation. CTGF is expressed in early stages of neural-tube development in both Xenopus laevis and the mouse. CTGF mRNA was detected in the floor plate and notochord of the Xenopus neural-plate stage [5] and its mRNA and protein staining was observed in the mouse neural tube as early as 10.5 days of gestation [34, 41]. Moreover, Freemantle et al. (2002)[42] showed by microarray that ctgf is one of 57 genes that are upregulated during retinoic acid-induced differentiation of the human embryonal carcinoma (EC) cell line, NT2/D1, toward a neuronal lineage. More recently, Obayashi et al. (2009) [43] also showed by microarray that ctgf is one of 45 genes upregulated during serum-induced differentiation of human neural progenitor cells into astrocytes. Our results showed that recombinant purified CTGF, added to the first steps of neural-cell differentiation in vitro, increased the number of cells that were immunostained for the astrocytic marker GFAP or cells double-marked for Nestin and GFAP, indicating an increase in the number of cells committed to the astrocytic phenotype. Although the maturity of these GFAP-positive cells was not revealed by these results, they suggest that these cells are in a transient stage, entering the astrocytic differentiation program. According to the literature, GFAP expression in primary cultures of radial glia starts after day 5, which correlates with the transition period of astrocyte maturation in rodents in vivo [44]. This result supports our in vitro system and indicates that CTGF’s role in the developing brain is related to the astrocyte differentiation program.

To induce cell differentiation, CTGF modulates important signaling pathways. Since most CTGF partners such as TGFβ-1, BMP, EGF and WNT are implicated in nervous-system development [4547], we could predict an important role for CTGF in the differentiation process of neural cells during development. The main interaction of CTGF is with the TGFβ-1 signaling pathway. TGFβ-1 induces expression and secretion of CTGF, and CTGF binds directly to the TGFβ-1 ligand and increases its binding to the receptors, enhancing TGFβ-1/SMAD2 signaling as a positive feedback loop [5]. This partnership has been implicated in many diseases, including most types of fibrosis [4851]. TGFβ-1/SMAD2 signaling also has a role during astrocyte fate commitment of embryonic neural-cell cultures [5253]. TGFβ-1 protein activated SMAD2 translocation to the nucleus of early-differentiated astrocytes and increased the overall number of GFAP-positive cells in these cultures while it decreased the expression of progenitor markers, suggesting that TGFβ-1 drives progenitor cells toward an astrocyte phenotype [52]. It is still not known if CTGF can increase TGFβ-1 signaling in these astrocyte precursors or in any of the developing neural cells. However, a few studies suggest that this cooperation between CTGF and TGFβ-1 can occur during development [5455]. In a previous study, our group showed that CTGF and components of the TGFβ-1 signaling pathway are expressed in the same structures during tooth development. Yet, CTGF seems not to cooperate with TGFβ-1 signaling in this situation since ctgf knockout mice did not show any reduction in the levels of phosphorylated SMAD2, suggesting that they might function independently [56].

CTGF also interacts with the MAPK signaling pathway. CTGF expression induced by TGFβ requires both phosphorylation of SMAD and p44/42 MAPK/ERK activation in rat mesangial cell line and human skin fibroblast primary cultures [18, 57]. The ERK pathway is required for CTGF to potentiate myofibroblast differentiation induced by TGFβ [58]. During chondrocyte differentiation, CTGF can block canonical signaling of BMP, a member of the TGFβ superfamily. However, it can also modulate non-canonical signaling pathways of BMP. CTGF inhibits BMP-induced ERK1/2 phosphorylation, blocking chondrocyte proliferation, and activates p38 MAPK signaling inducing chondrocyte differentiation [59, 37]. A vast amount of data implicates MAPK signaling pathways in astrocyte differentiation in vitro [53, 6062]. Our results showed that CTGF increased more than six fold the phosphorylation of p44/p42 MAPK but not p38 MAPK or P54/p46 SAPK/JNK in astrocyte progenitor cells, suggesting that at least part of the CTGF function in astrocyte differentiation may involve activation of the specific MAPK signaling cascade. Another well-known function of CTGF is to coordinate extracellular matrix production and degradation during development and in pathological conditions [12, 63]. Nagai and coworkers [64] showed that CTGF activates both p44/42 and p38 MAPK in the human retinal pigment epithelial cell line, leading to the production of laminin and fibronectin. The induction of fibronectin expression by CTGF seemed to require p44/42 MAPK activation, while the induction of laminin seemed to require both p44/42 and p38MAPK activation [64]. Our results showed that CTGF is able to induce the expression and deposition of fibronectin but not of laminin in these newly differentiated astrocytes. This result is partly in agreement with the results of Nagai et al. (2009), in that CTGF is able to activate only p44/42 MAPK and not p38, which correlates with the induction of fibronectin but not laminin.

Laminin and fibronectin are the two main extracellular matrix proteins of the developing brain. They are important, for example, in cell adhesion, proliferation and axon migration during the development of the nervous system. The finding that CTGF induces the expression of fibronectin by astrocytes during development suggests a possible role for CTGF in cellular interactions and/or axon migration during the development of the nervous system. Our study provided the first evidence that CTGF induces astrocyte differentiation, activating the MAPK signaling pathway and promoting fibronectin expression and deposition.

Incubation of glioblastoma stem cells with CTGF protein increased the expression of GFAP and a decrease of Sox2 proteins, indicating an increase in the differentiation rate of these cells (Fig 6). Targeting glioblastoma stem cells may involve indirect and direct strategies. Indirect targeting strategies involve the perivascular [65], hypoxic [66] and immune [67] niches. Direct approaches involve overcoming the resistance of glioblastoma stem cells to standard treatment [68], blocking their functions [69], and inducing differentiation into a less-tumorigenic state that is more vulnerable to cancer treatments [70]. The precise mechanisms by which CTGF induces glioblastoma stem-cell differentiation, however, need to be investigated further. These data, together with previously published information, point to a more-complex mechanism that could involve multiple pathways. Therefore, it is likely that CTGF can interact with different pathways and promote a link between them, during the first steps of astrocyte and also during glioblastoma stem-cell differentiation.

Supporting Information

S1 Fig. Recombinant CTGF did not affect the proliferation rate of neural cells in vitro.

(A) Graph showing the result of the WST-1 cell proliferation assay of untreated or CTGF-treated neural progenitor cells every 24 h after plating.

https://doi.org/10.1371/journal.pone.0133689.s001

(TIF)

S2 Fig. Recombinant CTGF did not affect the number of BrdU-positive cells in vitro.

(A and B) Immunocytochemistry showing BrdU-positive cells of untreated or CTGF-treated neural progenitor cells after 120 h in culture. (C) Graph showing the percentage of BrdU-positive cells of untreated or CTGF-treated neural progenitor cells after 120 h in culture. (D) [3H] Timidin incorporation of untreated or CTGF-treated neural progenitor cells after 120 h in culture.

https://doi.org/10.1371/journal.pone.0133689.s002

(TIF)

Acknowledgments

We thank Adiel Batista, Grasiella Ventura, Fernando Almeida and Fabio Zuim for animal care and technical support.

Author Contributions

Conceived and designed the experiments: FAM HC JGA VMN. Performed the experiments: FAM JMCA SAK AR LFR LGD LSSF. Analyzed the data: FAM JMCA SAK LGD LFR HC JGA LSSF. Contributed reagents/materials/analysis tools: HC JGA VMN. Wrote the paper: FAM JGA.

References

  1. 1. Bradham DM, Igarashi A, Potter RL, Grotendorst GR (1991) Connective tissue growth factor: a cysteine-rich mitogen secreted by human vascular endothelial cells is related to the SRC-induced immediate early gene product CEF-10. J Cell Biol 114:1285–1294. pmid:1654338
  2. 2. Aguiar DP, Pontes B, Mendes FA, Andrade LR, Viana NB, Abreu JG (2011) CTGF/CCN2 has a chemoattractive function but a weak adhesive property to embryonic carcinoma cells. Biochem Biophys Res Commun 413:582–587. pmid:21925146
  3. 3. Igarashi A, Okochi H, Bradham DM, Grotendorst GR (1993) Regulation of connective tissue growth factor gene expression in human skin fibroblasts and during wound repair. Mol Biol Cell 6:637–645.
  4. 4. Babic AM, Chen CC, Lau LF (1999) Fisp12/mouse connective tissue growth factor mediates endothelial cell adhesion and migration through integrin alphavbeta3, promotes endothelial cell survival, and induces angiogenesis in vivo. Mol Cell Biol 19:2958–2966. pmid:10082563
  5. 5. Abreu JG, Keptura NI, Reversade B, De Robertis EM (2002) Connective-tissue growth factor (CTGF) modulates cell signaling by BMP and TGF-beta. Nat Cell Biol 8:599–604.
  6. 6. Kang Y, Siegel PM, Shu W, Drobnjak M, Kakonen SM, Cordon-Cardo C, et al. (2003) A multigenic program mediating breast cancer metastasis to bone. Cancer Cell 6:537–549.
  7. 7. Mercurio S, Latinkic B, Itasaki N, Krumlauf R, Smith JC (2004) Connective-tissue growth factor modulates WNT signaling and interacts with the WNT receptor complex. Development 131:2137–2147. pmid:15105373
  8. 8. Crean JK, Furlong F, Finlay D, Mitchell D, Murphy M Conway B, et al. (2004) Connective tissue growth factor [CTGF]/CCN2 stimulates mesangial cell migration through integrated dissolution of focal adhesion complexes and activation of cell polarization. FASEB J 13:1541–1543
  9. 9. Grotendorst GR, Duncan MR (2005) Individual domains of connective tissue growth factor regulate fibroblast proliferation and myofibroblast differentiation. FASEB J 7:729–738.
  10. 10. Hoshijima M, Hattori T, Inoue M, Araki D, Hanagata H, Miyauchi A, Takigawa M (2006) CT domain of CCN2/CTGF directly interacts with Fibronectin and enhances cell adhesion of chondrocytes through integrin alpha5beta1. FEBS Lett. 580:1376–1382. pmid:16457822
  11. 11. Bork P (1993) The modular architecture of a new family of growth regulators related to connective tissue growth factor. FEBS Lett 327:125–130. pmid:7687569
  12. 12. Franzier K, Williams S, Kothapalli D, Klapper H, Grotendorst GR (1996) Stimulation of fibroblast cell growth, matrix production, and granulation tissue formation by connective tissue growth factor. J Invest Dermatol 107:404–411. pmid:8751978
  13. 13. Kothapalli D1, Frazier KS, Welply A, Segarini PR, Grotendorst GR. (1997) Transforming growth factor beta induces anchorage-independent growth of NRK fibroblasts via a connective tissue growth factor-dependent signaling pathway. Cell Growth Differ 8(1):61–8. pmid:8993835
  14. 14. Grotendorst GR, Okochi H, Hayashi N (1996) A novel transforming growth factor beta response element controls the expression of connective tissue growth factor gene. Cell Growth Differ 7:469–480. pmid:9052988
  15. 15. Weston BS, Wahab NA, Mason RM (2003) CTGF mediates TGF-beta-induced Fibronectin matrix deposition by upregulating active alpha5beta1 integrin in human mesangial cells. J Am Soc Nephrol 14:601–610. pmid:12595495
  16. 16. Arnott JA, Nuglozeh E, Rico MC, Arango-Hisijara I, Odgren PR, Safadi FF et al. (2007) Connective tissue growth factor (CTGF/CCN2) is a downstream mediator for TGF-beta 1-induced extracellular matrix production in osteoblasts. J Cell Physiol 210:843–852. pmid:17133352
  17. 17. Luo Q, Kang Q, Si W, Jiang W, Park JK, Peng Y, et al. (2004) Connective tissue growth factor (CTGF) is regulated by WNT and bone morphogenetic proteins signaling in osteoblast differentiation of mesenchymal stem cells. J Biol Chem 279:55958–55968. pmid:15496414
  18. 18. Chen Y, Blom IE, Sa S, Goldschmeding R, Abraham DJ, Leask A (2002) CTGF expression in mesangial cells: involvement of SMADs, MAP kinase and PKC. Kidney Int 62:1149–1159. pmid:12234285
  19. 19. Phanish MK, Wahab NA, Hendry BM, Dockrell ME (2005) TGF-beta 1-induced connective tissue growth factor (CCN2) expression in human renal proximal tubule ephitilial cells requires Ras/MEK/ERK and Smad signaling. Nephron Exp Nephrol 100:156–165.
  20. 20. Yuan LQ, Lu Y, Luo XH, Xie H, Wu XP, Liao EY (2007) Taurine promotes connective tissue growth factor (CTGF) expression in osteoblasts through the ERK signal pathway. Amino Acids 32:425–430. pmid:16937320
  21. 21. Gotoh Y, Nishida E, Yamashita T, Hoshi M, Kawakami M, Sakai H (1990) Microtubule-associated-protein (MAP) kinase activated by nerve growth factor and epidermal growth factor in PC12 cells. Identity with the mitogen-activated MAP kinase of fibroblastic cells. Eur J Biochem 193:661–669. pmid:2174361
  22. 22. Creuzet C, Loeb J, Barbin G (1995) Fibroblast growth factor stimulate protein tyrosine phosphorylation and mitogen-activated protein kinase activity in primary cultures of hippocampal neurons. J Neurochem 64:1541–1547. pmid:7891081
  23. 23. Northrop J, Woods A, Seger R, Suzuki A, Ueno N, Krebs E, et al. (1995) BMP-4 regulates the dorsal-ventral differences in FGF/MAPKK-mediated mesoderm induction in Xenopus. Dev Biol 172:242–252. pmid:7589804
  24. 24. Liu B, Yu J, Taylor L, Zhou X, Polgar P (2006) Microarray and phosphokinase screenings leading to studies on ERK and JNK regulation of connective tissue growth factor expression by angiotensin II 1a and bradykinin B2 receptors in Rat1 fibroblasts. J Cell Biochem 97:1104–1120. pmid:16294326
  25. 25. Nakanishi M, Niidome T, Matsuda S, Akaike A, Kihara T, Sugimoto H (2007) Microglia-derived interleukin-6 and leukaemia inhibitory factor promote astrocytic differentiation of neural stem/progenitor cells. Eur. Journal of Neuroscience 25:649–658.
  26. 26. Garcia-Abreu J, Mendes FA, Onofre GR, De Freitas MS, Silva LC, Moura Neto V, et al. (2000) Contribution of heparan sulfate to the non-permissive role of the midline glia to the growth of midbrain neurites. Glia 29: 260–272. pmid:10642752
  27. 27. Onofre GR, Werneck CC, Mendes FA, Garcia-Abreu J, Moura Neto V, Cavalcante LA, et al. (2001) Astroglial cells derived from lateral and medial midbrain sectors differ in their synthesis and secretion of sulfated glycosaminoglycans. Braz J Med Biol Res 34:251–8. pmid:11175502
  28. 28. Mendes FA, Onofre GR, Silva LC, Cavalcante LA, Garcia-Abreu J (2003) Concentration-dependent actions of glial chondroitin sulfate on the neuritic growth of midbrain neurons. Brain Res Dev Brain Res 142: 111–119. pmid:12711362
  29. 29. Silvestre DC, Pineda JR, Hoffschir F, Studler JM, Mouthon MA, Pflumio F, et al. (2011) Alternative lengthening of telomeres in human glioma stem cells. Stem Cells. 29:440–451. pmid:21425407
  30. 30. Ball DK, Rachfal AW, Kemper SA, Brigstock DR (2003) The heparin-binding 10 kDa fragment of connective tissue growth factor (CTGF) containing module 4 alone stimulates cell adhesion. J Endocrinol 176:R1–7 pmid:12553878
  31. 31. Shimo T, Nakanishi T, Nishida T, Asano M, Kanyama M, Kuboki T, et al. (1999) Connective tissue growth factor induces the proliferation, migration, and tube formation of vascular endothelial cells in vitro, and angiogenesis in vivo. J. Biochem 126:137–45. pmid:10393331
  32. 32. Nakanishi T, Nishida T, Shimo T, Kobayashi K, Kubo T, Tamatani T, et al. (2000) Effects of CTGF/Hcs24, a product of a hypertrophic chondrocyte-specific gene, on the proliferation and differentiation of chondrocytes in culture. Endocrinology 141:264–73. pmid:10614647
  33. 33. Nishida T, Nakanishi T, Asano M, Shimo T, Takigawa M (2000) Effects of CTGF/Hcs24, a hypertrophic chondrocyte-specific gene product, on the proliferation and differentiation of osteoblastic cells in vitro. J Cell Physiol 184:197–206. pmid:10867644
  34. 34. Ivkovic s, Yoon BS, Popoff SN, Safadi FF, Libuda DE, Stephenson RC, et al. (2003) Connective tissue growth factor coordinates chondrogenesis and angiogenesis during skeletal development. Development 130: 2779–2791. pmid:12736220
  35. 35. Safadi FF, Xu J, Smock SL, Kanaan RA, Selim AH, Odgren PR, Marks SC Jr, et al. (2003) Expression of connective tissue growth factor in bone: its role in osteoblast proliferation and differentiation in vitro and bone formation in vivo. J Cell Physiol 196:51–62. pmid:12767040
  36. 36. Kawaki H, Kubota S, Suzuki A, Yamada T, Matsumura T, Mandai T, et al. (2008) Functional requirement of CCN2 for intramembranous bone formation in embryonic mice. Biochem Biophys Res Commun 366:450–6. pmid:18067859
  37. 37. Maeda A, Nishida T, Aoyama E, Kubota S, Lyons KM, Kuboki T, et al. (2009) CCN family 2/connective tissue growth factor modulates BMP signalling as a signal conductor, which action regulates the proliferation and differentiation of chondrocytes. J Biochem 145:207–16. pmid:19038999
  38. 38. Tan JT, McLennan SV, Song WW, Lo LW, Bonner JG, Williams PF, et al. (2008) Connective tissue growth factor inhibits adipocyte differentiation. Am J Physiol Cell Physiol 295:C740–51. pmid:18596209
  39. 39. Wang W, Morrison B, Galbaugh T, Jose CC, Kenney N, Cutler ML (2008) Glucocorticoid induced expression of connective tissue growth factor contributes to lactogenic differentiation of mouse mammary epithelial cells. J Cell Physiol 214:38–46. pmid:17541935
  40. 40. Wu S, Platteau A, Chen S, McNamara G, Whitsett J, Bancalari E (2010) Conditional overexpression of connective tissue growth factor disrupts postnatal lung development. Am J Respir Cell Mol Biol 42:552–63. pmid:19541844
  41. 41. Surveyor GA, Brigstock DR (1999) Immunohistochemical localization of connective tissue growth factor (CTGF) in the mouse embryo between days 7.5 and 14.5 of gestation. Growth Factors 17:115–124. pmid:10595311
  42. 42. Freemantle SJ, Kerley JS, Olsen SL, Gross RH, Spinella MJ (2002) Developmentally-related candidate retinoic acid target genes regulated early during neuronal differentiation of human embryonal carcinoma. Oncogene 21:2880–9. pmid:11973648
  43. 43. Obayashi S, Tabunoki H, Kim SU, Satoh J (2009) Gene expression profiling of human neural progenitor cells following the serum-induced astrocyte differentiation. Cell. Mol. Neurobio. 29:423–438.
  44. 44. Sancho-Tello M, Vallés S, Montoliu C, Renau-Piqueras J, Guerri C (1995) Developmental pattern of GFAP and vimentin gene expression in rat brain and in radial glial cultures. Glia 15:157–66. pmid:8567067
  45. 45. Lie DC, Colamarino SA, Song HJ, Désiré L, Mira H, Consiglio A, et al. (2005) WNT signalling regulates adult hippocampal neurogenesis. Nature 437: 1370–1375. pmid:16251967
  46. 46. Gomes FC, Sousa Vde O, Romão L (2005) Emerging roles for TGF-beta1 in nervous system development. Int J Dev Neurosci 23: 413–424. pmid:15936920
  47. 47. D'Alessandro JS, Yetz-Aldape J, Wang EA (1994) Bone morphogenetic proteins induce differentiation in astrocyte lineage cells. Growth Factors 11:53–69. pmid:7530466
  48. 48. Rachfal AW, Brigstock DR (2003) Connective tissue growth factor (CTGF/CCN2) in hepatic fibrosis. Hepatol Res 26:1–9. pmid:12787797
  49. 49. Cooker LA, Peterson D, Rambow J, Riser ML, Riser RE, Najmabadi F, et al. (2007) TNF-alpha, but not IFN-gamma, regulates CCN2 (CTGF), collagen type I, and proliferation in mesangial cells: possible roles in the progression of renal fibrosis. Am J Physiol Renal Physiol 293:F157–165. pmid:17376761
  50. 50. Brigstock DR (2010) Connective tissue growth factor (CCN2, CTGF) and organ fibrosis: lessons from transgenic animals. J Cell Commun Signal 4:1–4. pmid:19798591
  51. 51. Kodama T, Takehara T, Hikita H, Shimizu S, Shigekawa M, Tsunematsu H, et al. (2011) Increases in p53 expression induce CTGF synthesis by mouse and human hepatocytes and result in liver fibrosis in mice. J Clin Invest 121:3343–56. pmid:21747166
  52. 52. Stipursky J and Gomes FCA (2007) TGF-β1/SMAD signaling induces astrocyte fate commitment in vitro: Implications for radial glia development. Glia 55:1023–1033. pmid:17549683
  53. 53. Romão LF, Sousa V de O, Neto VM, Gomes FC (2008) Glutamate activates GFAP gene promoter from cultured astrocytes through TGF-beta1 pathways. J. Neurochem 106:746–56. pmid:18419760
  54. 54. Oka K, Oka S, Sasaki T, Ito Y, Bringas P Jr, Nonaka K, et al. (2007) The role of TGF-beta signaling in regulating chondrogenesis and osteogenesis during mandibular development. Dev Biol 303:391–404. pmid:17204263
  55. 55. Nagashima T, Kim J, Li Q, Lydon JP, Demayo FJ, Lyons KM, et al. (2011) Connective tissue growth factor is required for normal follicle development and ovulation. Mol Endocrinol 25:1740–59. pmid:21868453
  56. 56. Pacheco MS, Reis AH, Aguiar DP, Lyons KM, Abreu JG (2008) Dynamic analysis of the expression of the TGFbeta/SMAD2 pathway and CCN2/CTGF during early steps of tooth development. Cells Tissues Organs 187:199–210. pmid:18089935
  57. 57. Stratton R, Rajkumar V, Ponticos M, Nichols B, Shiwen X, Black CM, Abraham DJ, et al. (2002) Prostacyclin derivatives prevent the fibrotic response to TGFβ2 by inhibiting the Ras/MEK/ERK pathway. FASEB J. 16:1949–51. pmid:12368229
  58. 58. Yang M, Huang H, Li J, Li D and Wang H. (2004) Tyrosine phosphorylation of the LDL receptor-related protein (LRP) and activation of the ERK pathway are required for connective tissue growth factor to potentiate myofibroblast differentiation. FASEB J. 18:1920–1940. pmid:15469966
  59. 59. Yosimichi G, Nakanishi T, Nishida T, Hattori T, Takano-Yamamoto T, Takigawa M. (2001) CTGF/Hcs24 induces chondrocyte differentiation through a p38 mitogen-activated protein kinase (p38MAPK), and proliferation through a p44/42 MAPK/extracellular-signal regulated kinase (ERK) Eur. J. Biochem. 268:6058–6065.
  60. 60. Rajan P. and McKay R.D., (1998) Multiple routes to astrocytic differentiation in the CNS. J Neurosci 18:3620–3629. pmid:9570793
  61. 61. Lee SM, Nga NTH, Park MH, Kim KS, Cho KJ, Moon DC, et al. (2004) EPO receptor-mediated ERK kinase and NF-kB activation in erythropoietin-promoted differentiation of astrocytes. Biochemical and Biophysical Research Communications 320:1087–1095. pmid:15249201
  62. 62. Osinde M, Mullershausen F, Dev KK (2007) Phosphorylated FTY720 stimulates ERK phosphorylation in astrocytes via S1P receptors. Neuropharmacology 52:1210–8. pmid:17379261
  63. 63. Mori T, Kawara S, Shinozaki M, Hayashi N, Kakinuma T, Igarashi A, et al. (1999) Role and interaction of connective tissue growth factor with transforming growth factor-beta in persistent fibrosis: A mouse fibrosis model. J Cell Physiol 181:153–159. pmid:10457363
  64. 64. Nagai N, Klimava A, Lee WH, Izumi-Nagai K, Handa JT (2009) CTGF is increased in basal deposits and regulates matrix production through the ERK (p42/44 mapk) MAPK and the p38 MAPK signaling pathways. Invest Ophthalmol Vis Sci 50:1903–1910. pmid:19011018
  65. 65. Folkins C, Man S, Xu P, Shaked Y, Hicklin DJ, Kerbel RS (2007) Anticancer therapies combining antiangiogenic and tumor cell cytotoxic effects reduce the tumor stem-like cell fraction in glioma xenograft tumors. Cancer Res 67:3560–4. pmid:17440065
  66. 66. Li Z, Bao S, Wu Q, Wang H, Eyler C, Sathornsumetee S, et al. (2009) Hypoxia-inducible factors regulate tumorigenic capacity of glioma stem cells. Cancer Cell 15:501–13. pmid:19477429
  67. 67. Patru C, Romao L, Varlet P, Coulombel L, Raponi E, Cadusseau J, et al. (2010) CD133, CD15/SSEA-1, CD34 or side populations do not resume tumor-initiating properties of long-term cultured cancer stem cells from human malignant glio-neuronal tumors. BMC Cancer 10:66. pmid:20181261
  68. 68. Wang J, Wakeman TP, Lathia JD, Hjelmeland AB, Wang XF, White RR, Rich JN, et al. (2010) Notch promotes radioresistance of glioma stem cells. Stem Cells 28:17–28. pmid:19921751
  69. 69. Suvà ML, Riggi N, Janiszewska M, Radovanovic I, Provero P, Stehle JC, Baumer K, et al. (2009) EZH2 is essential for glioblastoma cancer stem cell maintenance. Cancer Res 69:9211–8. pmid:19934320
  70. 70. Piccirillo SG, Reynolds BA, Zanetti N, Lamorte G, Binda E, Broggi G, et al. (2006) Bone morphogenetic proteins inhibit the tumorigenic potential of human brain tumour-initiating cells. Nature 444:761–5. pmid:17151667