brief communications

Connective-tissue growth factor (CTGF) modulates cell signalling by BMP and TGF-β José G. Abreu*, Nan I. Ketpura, Bruno Reversade and E. M. De Robertis† Howard Hughes Medical Institute and Department of Biological Chemistry, University of California, Los Angeles, CA 90095-1662, USA *Present address: Departamento de Anatomia, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, 21949-590, Rio de Janeiro, Brazil †e-mail: [email protected]

Published online: 22 July 2002; DOI:10.1038/ncb826

Connective-tissue growth factor (CTGF) is a secreted protein implicated in multiple cellular events including angiogenesis, skeletogenesis and wound healing1. It is a member of the CCN family of secreted proteins, named after CTGF, cysteine-rich 61 (CYR61), and nephroblastoma overexpressed (NOV) proteins. The molecular mechanism by which CTGF or other CCN proteins regulate cell signalling is not known. CTGF contains a cysteine-rich domain (CR) similar to those found in chordin and other secreted proteins2, which in some cases have been reported to function as bone morphogenetic protein (BMP) and TGF-β binding domains3–6. Here we show that CTGF directly binds BMP4 and TGF-β1 through its CR domain. CTGF can antagonize BMP4 activity by preventing its binding to BMP receptors and has the opposite effect, enhancement of receptor binding, on TGF-β1. These results show that CTGF inhibits BMP and activates TGF-β signals by direct binding in the extracellular space.

B

y sequence comparison to Chordin we noticed that CTGF contains a CR module previously designated as von Willebrand type c domain7 (vwc). CTGF contains four distinct structural modules: an amino-terminal insulin-like growthfactor-binding domain (IGFB), followed by the CR/vwc domain, a thrombospondin type 1 repeat (TSP-1), and a carboxy-terminal

cystine knot (CT) domain8 (Fig. 1a). The same modular architecture is shared by the other CCN family members. A comparison of the CR domain of CTGF with the CR domains of von Willebrand factor (vWF), thrombospondin (TSP), procollagens I and II and chordin showed conservation of ten regularly spaced cysteines and a few additional amino-acid residues characteristic of CR domains2 (Fig. 1b). Here we present functional studies on Xenopus laevis CTGF, a gene that is expressed in the floor plate, notochord, somites and other tissues known to be involved in early embryonic patterning (see Fig. S1 in the Supplementary Information). Synthetic CTGF mRNA injected into a single ventral blastomere at the four-cell stage was able to induce secondary axes9,10 in Xenopus embryos (66%, n = 115, Fig. 2b), which is consistent with inhibition of BMP signalling. The CTGF-induced axes were partial and never formed head structures with eyes, but included somites marked by the muscle-specific marker antibody 12-101 (data not shown). A construct encoding a secreted form of only the CR domain of CTGF (CTGF-CR, Fig. S2 in the Supplementary Information) was sufficient to induce ectopic axes, although at a lower frequency (30%, n = 78, Fig. 2c). Conversely, mRNA encoding CTGF lacking the CR domain (CTGF-∆CR, Fig. S2) failed to induce ectopic axes (0%, n = 50, Fig. 2d). Reverse transcriptionpolymerase chain reaction (RT-PCR) analysis of ectodermal explants showed that microinjected CTGF mRNA strongly induced the anterior marker Otx-2 and the cement-gland marker XAG-1

a CTGF SP

IGFB

CR

TSP-1

CT

b CTGF vWF TSP Procoll 2 Procoll 1 Chordin

CIF-GGTVYRSGESFQSS----CKYQCTCLDGA-VGCMPL---CSMDVRLPSPDCPFPRRVKLPGKCCEEWVC CIGEDGVQHQFLEAWVPDHQP-CQI-CTCLSGRKVNCTTQP--CPTAKAPTCGLCEVARLRQNADQCCPEYEC CLH-NGVLHKNRDEWTVD---SCTE-CTCQNSATI-CRKVS--CPL------MPCTNA--TIPDGECCP--RC CLQ-NGQRYKDKDVWKPS---SCRI-CVCDTGNVL-CDDII--CED------PDCLNP--EIPFGECCP--IC CVQ-NGLRYHDRDVWKPE---PCRI-CVCDNGKVL-CDDVI--CDET-----KNCPGA--EVPEGECCP--VC CSF-EGQLRAHGSRWAPDYDRKCSV-CSCQKRTVI-CDPIV--CPP------LNCSQP--VHLPDQCCP--VC

*

* **

Figure 1 CTGF encodes a CR-containing protein. a, The connective tissue growth factor (CTGF) protein contains four structural modules following the signal peptide (SP): insulin-like growth-factor binding (IGFB) domain, chordin-like cysteinerich (CR) domain, thrombospondin type 1 repeat (TSP-1), and a C-terminal cystineknot (CT). b, Alignment of the CR domains of CTGF, von Willebrand Factor, throm-

*

*

*

**

*

bospondin, procollagen 1 and 2 and Chordin (GenBank accession numbers XP037056, NM000552, P35448, NP001835, P02452 and AAC42222, respectively). Black boxes represent identical amino acids and gray boxes similar amino acids; dashes indicate gaps introduced to optimize the alignment. Asterisks mark the ten identical cysteine residues characteristic of CR domains.

NATURE CELL BIOLOGY VOL 4 AUGUST 2002 http://cellbio.nature.com

©2003 Nature Publishing Group

599

d

CTGF-CR

CTGF-∆CR

f

g

Uninj.

CTGF

ntr co

AC

sta

c

WE

e

GF

CTGF

AC

Uninj.

CT

25

b

ge

a

ol

brief communications

Otx-2 XAG-1 NCAM Msx-1

i

h

Cytokerati α-Actin

Uninj.

XAG-1

j

k

XAG-1

l

Uninj.

Sox-2 CTGF

Sox-2 CTGF-CR Sox-2

Uninj.

Msx-1 CTGF

Msx-1 CTGF-CR Msx-1

m

n

EF1-α

o

Figure 2 CTGF mRNA injections induce anti-BMP phenotypes in Xenopus embryos. a–d, Dorsal views of tail-bud stage embryos. a, Uninjected; b, CTGF mRNA (150 pg); c, CTGF-CR mRNA (800 pg) and d, CTGF-∆CR mRNA (500 pg) injected ventrally at 4-cell stage. e, RT-PCR analysis of whole embryo (WE), animal caps uninjected (AC control) or injected with 50 pg of CTGF mRNA per blastomere at the 4-cell stage (AC CTGF). Animal cap explants were dissected at stage 9 and cultured until sibling embryos reached stage 26. EF1-α was used as RNA loading control and α-Actin to monitor mesoderm in the ectodermal explants. Three-day tadpoles, f, uninjected, g, injected with 50 pg of CTGF mRNA into the animal region of each blastomere at 4-cell stage. Cement-gland marker XAG-1, h, uninjected, i, injected with 150 pg of CTGF mRNA per blastomere at 4-cell stage. j–o, in situ hybridization at neural plate stage for Sox-2 (j–l) and Msx-1 (m–o); j, m, Uninjected embryos; k, n, injected with 50 pg of CTGF; i, o, 150 pg of CTGF-CR mRNA injected per blastomere at 4-cell stage.

(Fig. 2e). In addition, CTGF caused a weaker induction of the panneural marker N-CAM and downregulation of the epidermal markers Msx-1 and Cytokeratin (Fig. 2e). Microinjection of CTGF mRNA into the animal pole resulted in embryos with enlarged heads, shortened trunk and tail structures (Fig. 2f, g), and expanded expression of the anterior cement-gland marker XAG-1 (Fig. 2h, i). In CTGF-injected embryos analysed at the neurula stage, the expression domain of the neural-plate marker Sox-2 was expanded (Fig. j, k) and, reciprocally, the expression domain of Msx-1, which demarcates the border between the neural plate and epidermis, was displaced laterally (Fig. 2m, n). Embryos injected with CTGF-CR also developed with expanded neural plates (Fig. 2l, o). We conclude from these in vivo results that CTGF can induce anti-BMP phenotypes in microinjected Xenopus embryos through its CR domain. To test whether CTGF and BMP interacted biochemically, Flagepitope-tagged constructs of full-length CTGF, CTGF-CR, CTGF∆CR and the C terminus of CTGF (CTGF-CT) were generated and the proteins produced in cultured cells (Fig. S2). To produce CTGF in sufficient amounts for biochemical experiments we generated a Drosophila melanogaster S2 stable cell line secreting Flag-tagged 600

Xenopus CTGF. Under these conditions the protein was full length and could be affinity-purified via its Flag tag (Fig. S2). Immunoprecipitation assays showed that full-length CTGF, like the positive control chordin, could bind BMP4 in solution (Fig. 3a, lanes 2, 3). Using a monoclonal antibody that is entirely specific for BMP4 (ref. 11), we showed in western blots that the binding of BMP4 to CTGF could be competed by an excess of BMP2 or TGFβ1 but not by IGF-1 (Fig. 3b, lanes 2–5). The interaction between CTGF and BMP4 was direct, because in chemical crosslinking experiments, full-length CTGF or the CR domain of CTGF (CTGFCR) could be crosslinked to BMP4, forming complexes of the expected molecular weights (Fig. 3c, lanes 2, 5). Conversely, CTGF constructs lacking the CR domain (CTGF-∆CR and CTGF-CT) were unable to form complexes with BMP4 (Fig. 3c, lanes 3, 4). The observation that binding of BMP4 to CTGF could be partially competed by TGF-β1 (Fig. 3b, lane 4) suggested that CTGF might also bind TGF-β1. Chemical crosslinking experiments showed that TGF-β1 could also bind directly to CTGF or to CTGF-CR (Fig. 3d, e). The binding affinity of CTGF for BMP4 and TGF-β1 was determined by surface plasmon resonance (SPR) analysis, a method used previously to evaluate real-time interactions between BMP and follistatin12. Kinetic measurements using different concentrations of CTGF yielded dissociation constants (KD) of 5 nM for BMP4 and of 30 nM for TGF-β1 (Fig. 3f). We conclude from these biochemical studies that CTGF can directly bind BMP4 and TGF-β1 through its CR domain, and has a higher affinity for BMP4 than for TGF-β1. Purified CTGF inhibited BMP signalling in 10T1/2 cells13,14 (Fig. 4a, b). Increasing amounts of CTGF inhibited the alkaline phosphatase (AP) activity induced by 2.5 nM BMP4 in a dosedependent manner (Fig. 4b) with 50% inhibition (IC50) at approximately 12 nM. The phosphorylation of Smad1 induced by 0.5 nM BMP4 was inhibited by the addition of 20 nM CTGF (Fig. 4c, lanes 2 and 3) to human 293T cell cultures15. Interestingly, an excess of BMP4 (10 nM) was able to quench the antagonistic effect of CTGF on BMP4 action (Fig. 4c, lanes 4 and 5). To address the molecular mechanism by which CTGF inhibited BMP4 signalling we next examined the effect of CTGF on the binding of BMP4 to its cognate receptor16. Increasing amounts of CTGF antagonized BMP4 binding to a recombinant type Ia BMP-receptor-Fc fusion protein, as shown by western blot analysis with anti-BMP4 antibody (Fig. 4d, lanes 2–5). We conclude that binding of BMP4 to CTGF inhibits BMP signalling by preventing the binding of BMP4 to its cognate receptor. This molecular mechanism of action is analogous to that of the BMP antagonist chordin13. CTGF did not inhibit binding of TGF-β1 to a TGFβ-receptor IIFc (TβRII-Fc) recombinant protein. On the contrary, preincubation with CTGF increased the crosslinking of [125I]TGF-β1 to recombinant TβRII-Fc in solution (Fig. 5a). CTGF also enhanced the binding of [125I]TGF-β1 to endogenous receptors on the surface of cultured cells. Using the method of Massague17, it was found that crosslinking of TGF-β to all three TGF-β interacting proteins — betaglycan, TβRII and TβRI — increased in the presence of CTGF (Fig. 5b). Because CTGF binds TGF-β1 with relatively low affinity (with a KD value of only 30 nM, Fig. 3f), it is conceivable that CTGF may function as a chaperone to modify the conformation or solubility of TGF-β1 and facilitate presentation to its cognate receptors, which have affinities in the picomolar range17. Signalling by TGF-β1 was also increased by CTGF. At low TGF-β1 concentrations, CTGF potentiated the phosphorylation of Smad2 induced by TGF-β1 (Fig. 5c, lanes 2 and 3) in fetal mink lung (Mv1Lu) cell cultures. At higher concentrations of TGF-β1, CTGF did not increase Smad2 phosphorylation, indicating that CTGF serves to potentiate the effects of limiting amounts of TGF-β1 and has no detectable effects of its own on these cells (Fig. 5c, lanes 4–6). In addition, a TGF-β reporter gene18 was induced 6–10-fold by as little as 3 nM CTGF (Fig 5d). TGF-β1 was required, as CTGF alone did not induce reporter-gene expression, even at 45 nM (Fig. 5d). NATURE CELL BIOLOGY VOL 4 AUGUST 2002 http://cellbio.nature.com

©2003 Nature Publishing Group

brief communications b

BMP4 Flag (CTGF)

+ – – +

Myc (Chordin) 37 -

- 100

1

3

d

2

e CTGF (20 nM) –

4



TGF-β1

50 C

- IgG - CTGF

37 -

B:C B:CR B

25 -

5

1

2

3

4

5

400

TGF-β1

Mr(K)

C:T

75 -

f CTGF-CR (25 nM)

Mr(K) 100 -

3

75 -

CR2:T

37 -

CR2

25 CR

25 -

Relative resonance units

2

Mr(K) - BMP4

15 -

- 37

1

BMP4 (2 nM) R

+ – + –

-C

+ + – –

GF

+ – – –

CT

– – – –

R

+

T

+

-C

+

-∆C

+

GF

TGF-β1 IGF-1 Mr(K)

+

GF

Mr(K) - 25

Blot: anti-

c BMP4 CTGF BMP2

CT

+ + –

GF

+ – +

CT

BMP4 + CTGF – Chordin –

CT

a

300 Kd = 5 × 10-9 M (BMP4) 200 100

Kd = 30 × 10-9 M (TGF-β1)

0 50

130

210

290

370

450

Time (s)

β1 through the CR domain. a, WesternFigure 3 CTGF binds BMP4 and TGF-β blot analysis of BMP4 (5 nM) bound to CTGF (10 nM, lane 3) or chordin (5 nM, lane 2) after immunoprecipitation (IP) as described previously 4. To determine IP effectiveness, the same blots were subsequently probed with anti-Flag antibody for CTGF and anti-myc antibody for chordin. b, Competition of BMP4 (5 nM) bound to CTGF (10 nM) by IP in the absence (lane 2) or presence of 20-fold molar excess of BMP2 (lane 3), TGF-β1 (lane 4) or IGF-1 (lane 5). CTGF IP efficiency was monitored with anti-Flag antibody. c, Chemical crosslinking of CTGF and CTGF mutant constructs to BMP4. Membrane was probed with anti-BMP4 antibody. Bands corresponding to BMP4 dimer (B), BMP4–CTGF complex (B–C) and BMP4:CTGF-CR complex (B–CR)

are indicated. d, e, Western blot probed with anti-Flag antibody after chemical crosslinking of CTGF and CTGF-CR to TGF-β1. Bands corresponding to CTGF monomer (C), CTGF–TGF-β1 (C–T) complex, and to CTGF-CR monomer (CR), to the dimer of CTGF-CR (CR2) and to the complex of a dimer of CTGF-CR with TGF-β1 (CR2–T) are indicated. The TGF-β1 was added at 25 nM and 50 nM. f, Sensorgrams of SPR analyses showing the binding of purified CTGF to BMP4 or to TGF-β1. CTGF protein was run (black arrowhead) over BMP4 or TGF-β1 sensor chips. After CTGF flow ended (white arrowhead), dissociation was monitored by a decrease in the resonance units. Kinetic experiments were performed using CTGF concentrations ranging between 6.5 nM and 210 nM of full-length CTGF.

The synergy between CTGF and TGF-β1 was accompanied by a striking and unexpected morphological change in the cultures. Spherical aggregates of cells were observed in Mv1Lu cells treated with TGF-β1 and purified CTGF, but not when treated with either factor on its own (Fig. 5e and Fig. S3 in the Supplementary Information). These effects of CTGF were not caused by a blockade of endogenous BMP signals, because addition of 10 nM chordin or BMPRIa-Fc (which are sufficient to inhibit BMP4 signalling in cell culture, data not shown) together with TGF-β1 did not induce spherical structures in Mv1Lu cells (Fig. 5e). In embryonal carcinoma P19 cells similar structures were induced by CTGF alone (Fig. 5f), presumably because these cells produce endogenous TGF-β (see the legend to Fig. S3). The cell aggregates were positive for platelet-endothelial cell-adhesion molecule (PECAM-1) and for vWF (Fig. 5f and Fig. S3). Although these molecules are markers for endothelial cells, it has recently been reported that PECAM-1 is not entirely specific for blood vessels19 and therefore the determination of whether the spherical structures reported here correspond to blood-vessel-like structures must await further experimentation. We conclude that CTGF protein enhances TGF-β1 signalling by several criteria, including receptor and cell-surface binding, Smad2 phosphorylation, activation of gene expression, and differentiation of cultured cell lines. Although multiple physiological roles have been proposed for CTGF, its signalling pathway remains unknown. CTGF can bind

to low-density lipoprotein receptor-related protein and integrins20,21, but the consequences of these interactions on downstream signaltransduction pathways are not known. The results reported here indicate that CTGF signals in part through the binding of BMP4/2 and TGF-β1 in the extracellular space. We find that CTGF inhibits BMP but activates TGF-β1 signalling. TGF-β is a well-known inducer of extracellular matrix (ECM) components such as collagen and fibronectin22,23. The CTGF gene may participate in this event, as its promoter contains a TGF-β response element24,25. In support of this view, collagen deposition and anchorage-independent growth induced by TGF-β in fibroblasts can be inhibited by adding neutralizing antibodies against CTGF23,26, indicating a synergistic relationship between CTGF and TGF-β. The function of the other domains in CTGF remains to be investigated: the IGFB domain has been shown to bind IGF although this interaction is of low affinity (ref. 27 and our unpublished observations), TSP-1 domains have been implicated in extracellular matrix and vascular endothelial growth factor (VEGF) binding28,29, and the cystine-knot module may contribute a dimerization motif or additional signalling capabilities30. In conclusion, the results presented here suggest a molecular mechanism for CTGF function by which this protein can activate TGF-β signalling and inhibit BMP activity by directly binding to these growth factors in the extracellular space through its Chordin-like domain.

NATURE CELL BIOLOGY VOL 4 AUGUST 2002 http://cellbio.nature.com

©2003 Nature Publishing Group

601

brief communications c

b

20 10 0

CTGF 45 nM 0

0.1 1

2.5 0.1

1

2.5

CTGF (20 nM)



BMP4 (nM)



30



+



0.5 0.5 10

+

+

10



Mr(K) 20

-75 P Smad1

-50

10 0

2.5 nM BMP4

Actin

-37 1

2

3

4

5

6

No

BM

BMP4 (nM)

P4 No CT CT GF GF 3.5 n CT M GF 35 nM CT GF BM 70 PR nM laFc 5n M

30

10T1/2 AP activity × 104

10T1/2 AP activity × 104

a

d

BMPRla-Fc 1nM BMP4 1nM Input 50%

1

CTGF

2

3

4

5

6

Figure 4 CTGF antagonizes BMP4 signalling by inhibiting receptor binding. a, Dose-dependent induction of alkaline phosphatase (AP) by BMP4 in 10T1/2 cells is inhibited by CTGF. b, Adding increasing amounts of CTGF inhibits AP induced by 2.5 nM of BMP4. We estimate that CTGF can block BMP4 activity with an IC50 of about 12 nM. Error bars represent standard deviations of triplicate samples. Experiments were independently performed three times. c, Inhibition of Smad1 phosphorylation in 293T cells by CTGF at low, but not high, BMP4 concentrations.

d, CTGF inhibits binding of BMP4 to its cognate receptor. BMP4 (1 nM) was preincubated 1 h at room temperature with 10, 25 and 50 nM (black triangle) of affinity-purified CTGF (lanes 3–5), and BMPRIa-Fc was added at 1 nM for 2 h. Lane 1 corresponds to the loading of 50% of the total BMP4 used in each reaction. Lane 6 shows negative control of BMP4 binding to protein A beads in the absence of the BMPRIa-Fc.

Methods

buffer (128 mM NaCl, 5 nM KCl, 5 mM MgSO4, 1.2 mM CaCl2, 50 mM HEPES, 2 mg ml−1 BSA, pH 7.5) and incubated for 4 h at 4 °C on an oscillating platform at 120 rpm. After extensive washes with ice-cold binding buffer, DSS was added in binding buffer without BSA for 15 min at 4 °C. Cells were extracted in detachment buffer (0.25 M sucrose, 10 mM Tris, 1 mM EDTA, pH 7.4, 0.3 mM PMSF), sedimented by centrifugation and the pellets solubilized with solubilization buffer (125 mM NaCl, 10 mM Tris, 1 mM EDTA, pH 7.0, 1% Triton X-100). Cell debris were removed by centrifugation, and samples were subjected to SDS–PAGE.

DNA constructs and embryo manipulations The complete open reading frame of Xenopus CTGF was amplified by PCR from cDNA of stage 25 embryos using the following primers: forward 5′-AAA CTC GAG GAA CTA ATC TGC TGC TGT-3′ and reverse 5′-AAA TCT AGA TGT GTT TCT CTG CCT CTA-3′, based on the sequence from GenBank accession number U43523 entered by M. L. King and Z. Ying in 1997. The PCR product was cloned in the expression vector pCS2+. Epitope-tag subclones were generated by amplifying CTGF lacking the signal peptide and inserting the fragment in-frame into pCS2+ containing a chordin signal peptide until Ala41, followed by the Flag-tag sequence (pchd-Flag). The primers used for Flag-tagged fulllength CTGF or deletion mutants were as follows: CTGF 5′-AAA CTC GAG CAG GAG TGT AAT GGG GAA T-3′ and 5′-AAA TCT AGA TGT GTT TCT CTG CCT CTA-3′; CTGF-CR 5′-AAA CTC GAG GCC CCT TGT TGT GTT CGG-3′ and 5′-AAA TCT AGA TAG CGC GGA TTA GGG ATG GA-3′; CTGF-CT, 5′-AAA CTC GAG GGT CCT GAT CCA TCC CTA-3′ and T7 primer. CTGF-∆CR was generated by deletion of sequences between Ala95 and Ala191 of CTGF by PCR mutagenesis. Detailed protocols for embryo fertilizations, in situ hybridization and RT-PCR can be found at http://www.hhmi.ucla.edu/derobertis/index.html.

Protein purification, immunoprecipitation and crosslinking Flag-tagged secreted proteins were obtained by transient tranfection of human 293T cells or from Drosophila S2 cells stably expressing CTGF (Fig. S2). Full-length Flag-CTGF from S2 cells was affinitypurified with anti-Flag M2 affinity gel column and eluted with Flag peptide according to manufacturer’s directions (Sigma, St Louis, MO). Culture medium obtained from Drosophila S2 cells transfected with empty vector was purified in the same way and used as negative control in all experiments. Protein concentrations were determined using bovine serum albumen (BSA) standards stained with Coommassie blue in SDS–polyacrylamide gel electrophoresis (SDS–PAGE). Immunoprecipitation analyses and chemical crosslinking with disuccinimidyl suberate (DSS) were performed as described previously4,13.

TGF-β-receptor binding [125I]-TGF-β1 (specific activity 3000 Ci/mmol, NEN) was crosslinked to recombinant TGF-β-receptor II-Fc (TβRII-Fc) (R&D systems, Minneapolis, MN). Different concentrations of [125I]-TGF-β1 were pre-incubated for 1 h in absence or presence of 10 nM of purified CTGF. 1 nM of TβRII-Fc was added to the reactions for 30 min and the samples were crosslinked with DSS for 30 min4. Binding of [125I]-TGF-β1 to cell surface receptors of NRK9F cultured cells was performed as described previously17. Briefly, 100 pM [125I]-TGF-β1 was pre-incubated with or without 6 nM of purified CTGF for 1 h. Proteins were added to semi-confluent normal rat kidney (NRK) cells pre-equilibrated in binding

602

Surface plasmon resonance (SPR) analysis Real-time binding experiments were performed on the Biacore 2000 apparatus (Biacore AB, Uppsala, Sweden) as described previously12. Carrier-free recombinant human BMP4 and TGF-β1 from human platelets (R&D Systems) were immobilized on the sensor-chip surface (CM5, certified grade, Biacore AB). HBS-EP buffer (10 mM HEPES pH 7.4, 150 mM NaCl, 3 mM EDTA and 0.005% v/v surfactant P20) was used as running buffer.

Cell culture assays and immunocytochemistry Alkaline phosphatase (AP) induction assay13 was performed in the multipotent mesodermal cell line 10T1/2 (ATCC) using the fluorescent substrate 4-methyl umbelliferyl-phosphate (Sigma). AP values were normalized using the amount of protein in each tissue-culture well (Bio-Rad Dc protein kit). Mink lung epithelial cells (Mv1Lu – CCL 64, ATCC) were cultured in alpha-minimal essential media (MEM; Gibco, Grand Island, NY) supplemented with 10% foetal bovine serum. Cells plated in 96-well plates were transiently co-transfected with 0.1 µg of p3TP–lux (ref. 18) and 0.015 µg of pBSKSβ-gal expression plasmid using FuGENE (Roche, Indianapolis, IN) as a transfection reagent. Before growth factors were added, cells were washed with phosphate buffer saline and pre-incubated for 2 h with alpha-MEM containing 2% NuSerum (BD Biosciences, San Jose, CA). Growth factors diluted in alpha-MEM containing 2% NuSerum were pre-incubated at room temperature for 1 h before they were added to cells. After 18 h cells were lysed in Reporter Lysis Buffer (Promega, Madison, WI), and enzyme activity measured using the Luciferase Assay System and the β-Galactosidase Enzyme Assay System (Promega). Enzyme activities were measured in a microplate luminometer and enzyme-linked immunosorbent assay (ELISA) plate reader, respectively. Smad phosphorylation was detected on western blots using 1:6000 dilution of anti-phosphoSmad1 (specific for Smad 1, 5 and 8, gift of C. H. Heldin), 1:500 dilution of anti-phosphoSmad2 (Upstate Biotechnology, Waltham, MA), and 1:400 actin antibodies (Sigma) for protein loading controls. Purified growth factors were premixed for 1 h in serum-free medium (DMEM, F12, Iscove’s, 1:1:1) at room temperature, and added to 80% confluent 293T or Mv1Lu cells for 2 h. Immunocytochemistry was performed with an endothelial-specific anti-CD31 (PECAM-1) monoclonal antibody22 (MEC 13.3, Pharmingen, San Diego, CA) diluted 1:400, and an anti-vWF polyclonal antibody (Dako Co., Burlingame, CA) diluted 1:200. Staining was visualized after incubation with secondary

NATURE CELL BIOLOGY VOL 4 AUGUST 2002 http://cellbio.nature.com

©2003 Nature Publishing Group

brief communications a

b TβRII-Fc (1 nM)

125l-TGF-β1

125l-TGF-β1

– –

CTGF (6 nM) TGF-β1 (5 nM)

CTGF (10 nM) + – + – + – + –

+ –

+ +

+ –

2

3

4

βG

-TβRII-Fc–TGF-β1 Complex

RII RI -[TGF-β1]2 -TGF-β1 1

1 2 3 4 5 6 7 8

c CTGF (6 nM)





+



+

3TP-luciferase activity

d +

TGF-β1 (pM) – 10 10 50 50 1 Mr(K) 75 Smad2 P

50 -

Actin

37 -

5500 4500 3500 2500 1500 500 0 TGF-β1 GF

GF 45

nM

CT

GF 35

nM

CT

CT

CT M

F-β

CT

No

TG No

e

GF

6

nM

5

3n

4

15

3

1

2

GF

1

f

Condition Control TGF-β1 CTGF TGF-β1 + CTGF TGF-β1 + CTGF + TβRII-Fc TGF-β1 + Chordin TGF-β1 + BMPR1a-Fc Chordin BMPRqa-Fc

No. of Mv1Lu cell aggregates per cm2 0 0 0 53 13 0 0 0 0

PECAM-1

vWF

CTGF-treated

P19 cells

DAPI

β1 receptor binding and signalling. Figure 5 CTGF potentiates TGF-β a, Crosslinking of [125I]-TGF-β1 to 1 nM TGF-β receptor Fc fusion protein (TβRII-Fc) in the presence (+) or absence (−) of 10 nM purified CTGF. The TβRII-Fc–TGF-β1 complex is seen in the upper part of the gel. Different concentrations of [125I]-TGF-β1, ranging from 10 pM to 0.5 nM, were used. b, Labelling of cell-surface proteins by [125I]-TGF-β1 in NRK cells after chemical crosslinking with DSS. 125I-TGF-β1 (100 pM) was pre-incubated 1 h with (+) or without (−) 6 nM CTGF before adding for receptor binding17. Specific binding was shown by competition with 50-fold unlabelled TGF-β1 (lane 3). Lane 4 shows negative control crosslinking of CTGF, and [I125]-TGF-β1 in the absence of cells. The three major cell surface TGF-β receptors17 are indicated: betaglycan (βG, 250 kDa), TGF-β-receptor II (TβRII, 80 kDa) and TGF-β-receptor I (TβRI, 68 kDa). c, Two hours after addition of proteins, Smad2 phosphorylation in Mv1Lu cells was stimulated by 6 nM CTGF in the presence of 10 pM TGF-β1. This effect was not seen when TGF-β1 was increased fivefold. Lane 6 shows that CTGF

has no effect on its own. Note that Smad2 phosphorylation was induced at lower concentrations of TGF-β1 than those required in reporter-gene or cell-differentiation assays (see below), perhaps reflecting the shorter treatment times involved. d, Luciferase activity 36 h after transfection of Mv1Lu cells with the TGF-β inducible reporter p3TP–lux. Constant TGF-β1 and different concentrations of CTGF were used. Note that CTGF protein stimulates TGF-β1 signalling even when as little as 3 nM is used. e, Number of spherical cell aggregates per square centimeter observed in Mv1Lu cells after 12 h of treatment with different growth factors as described above. The concentrations of CTGF and TGF-β1 were 3 nM and 200 pM, respectively. Note that the BMP inhibitors Chordin and BMPRIa-Fc could not replace CTGF, and that soluble TGFRII-Fc inhibited spherical aggregate formation. f, Immunofluorescence showing the formation of spherical cell aggregates in P19 embryonal carcinoma cells treated with 5 nM CTGF. P19 cell aggregates were positive for PECAM-1 and vWF.

antibodies conjugated to fluorescein isothiocyanate (FITC) or Cy3 (Jackson Immunoresearch Laboratories) using fluorescent microscopy. Coverslips were mounted in Vectashield mounting media containing DAPI (Vector, Carpinteria, CA). Negative controls were performed by omitting the primary antibody during the immunostaining reaction.

and A. Cuellar for technical assistance, and C. Coffinier, E. Delot, J. I. Kim, J. Larraín, K. M. Lyons, M. Oelgeschläger, E. Pera, O. Wessely for discussions and comments on the manuscript. J.G.A. was a Latin American PEW fellow. This work was supported by the National Institutes of Health (R37 HD21502-16). E.M.D.R. is an Investigator of the Howard Hughes Medical Institute. Correspondence and requests for material should be addressed to E.M.D.R. Supplementary information is available on Nature Cell Biology’s website (http://cellbio.nature.com).

ACKNOWLEDGEMENTS We thank K. M. Lyons and S. Ivkovic for unpublished information, K. Masuhara and C.H. Heldin for antibodies, J. Massague for reporter plasmid, Naoto Ueno for the protocol to immobilize BMP4 to Biacore chips, M. L. King and Z. Ying for entering the CTGF full-length sequence in GenBank, S.-Y. Li

COMPETING FINANCIAL INTERESTS The authors declare that they have no competing financial interests.

NATURE CELL BIOLOGY VOL 4 AUGUST 2002 http://cellbio.nature.com

©2003 Nature Publishing Group

603

brief communications RECEIVED 28 MARCH 2002; REVISED 10 MAY 2002; ACCEPTED 24 MAY 2002; PUBLISHED 22 JULY 2002

1. Moussad, E. E. & Brigstock, D. R. Mol. Genet. Metab. 71, 276–292 (2000). 2. Abreu, J., Coffinier, C., Larraín, J., Oelgeschläger, M. & De Robertis, E. M. Gene 287, 39–47 (2002). 3. Zhu, Y., Oganesian, A., Keene, D.R. & Sandell, L. J. J. Cell Biol. 144, 1069–1080 (1999). 4. Larraín, J. et al. Development 127, 821–830 (2000). 5. Nakayama, N. et al. Dev. Biol. 232, 372–387 (2001). 6. Sakuta, H. et al. Science 293, 111–115 (2001). 7. Hunt, L.T. & Barker, W.C. Biochem. Biophys. Res. Commun. 144, 876–882 (1987). 8. Bork, P. FEBS Lett. 327, 125–130 (1993). 9. Smith, W.C. & Harland, R.M. Cell 70, 829–840 (1992). 10. Sasai, Y. et al. Cell 79, 779–790 (1994). 11. Masuhara, K. et al. Bone 16, 91–96. (1995). 12. Iemura, S. et al. Proc. Natl. Acad. Sci. USA 95, 9337–9342 (1998). 13. Piccolo, S., Sasai, Y., Lu, B. & De Robertis, E. M. Cell 86, 589–598 (1996). 14. Katagiri, T. et al. Biochem. Biophys. Res. Commun. 172, 295–299 (1990). 15 Persson, V. et al. FEBS Lett. 434, 83–87 (1998).

604

16. Larraín, J. et al. Development 128, 4439–44347 (2001). 17 Massagué, J. Methods Enzymol. 46, 174–195 (1987). 18. Wrana, J. L. et al. Cell 71, 1003–1014 (1992). 19. Robson, P., Stein, P., Zhou, B., Schultz, R. M. & Baldoin, H. S. Dev. Biol. 234, 317–329 (2001). 20. Segarini, P. R. et al. J. Biol. Chem. 276, 40659–40667 (2001). 21. Kireeva, M. L. et al. Exp. Cell Res. 233, 63–77 (1997). 22. Roberts, A. B. et al. Proc. Natl. Acad. Sci. USA 83, 4167–4171 (1986). 23. Frazier, K., Williams, S., Kothapalli, D., Klapper, H. & Grotendorst, G. R. J. Invest. Dermatol. 107, 404–411 (1996). 24. Grotendorst, G. R., Okochi, H. & Hayashi, N. Cell Growth Differ. 7, 469–480 (1996). 25. Holmes, A. et al. J. Biol. Chem. 276, 10594–10601 (2001). 26. Kothapalli, D., Frazier, K. S., Welply, A., Segarini, P. R. & Grotendorst, G. R. Cell Growth Differ. 8, 61–68 (1997). 27. Kim, H. S. et al. Proc. Natl Acad. Sci. USA 94, 12981–12986 (1997). 28. Adams, J. C. & Tucker, R. P. Dev. Dyn. 218, 280–299 (2000). 29. Inoki, I. et al. FASEB J. 16, 219–221 (2002). 30. Pearce, J. J., Penny, G. & Rossant, J. Dev. Biol. 209, 98–110 (1999).

NATURE CELL BIOLOGY VOL 4 AUGUST 2002 http://cellbio.nature.com

©2003 Nature Publishing Group

supplementary infomation

Figure S1 Expression pattern of CTGF in Xenopus embryogenesis detected by in situ hybridization. a, lateral view of four-cell stage embryo; b, dorsal view of early neural plate stage; c, close up of section at neural plate stage, note staining in floor plate (Fp) and notochord (No); d, e, dorsal and lateral view at the late neurula stage showing expression in somites; f, cross-section of a late neurula; g, i, lateral view of tailbud stage embryos; h, section through early tailbud stage. The sequence of Xenopus CTGF had been presented in the Genbank database (accession # U43523), but its expression during development had been not described. Our in situ hybridization analysis revealed the presence of maternal CTGF transcripts in the animal region of four-cell stage embryos and very little expression during gastrulation (panel a and data not shown). At early neurula stage zygotic transcripts were detected in the dorsal midline, mainly in the floor plate, and at lower levels in the underlying notochord (b-c). CTGF was also expressed throughout the developing somites at the late neurula stage and later became restricted to the

dorsal aspect of the somite (f-i). At tailbud stage additional expression domains were seen in the developing heart, nasal placode and branchial arches (g, i). Thus, CTGF is expressed at various sites during early Xenopus development, including dorsal tissues known to be involved in embryonic patterning. An antisense morpholino oligonucleotides directed against the immediate 5’ leader region of CTGF mRNA (sequence: GTACAGCAGCAGATTAGTTCTCTTC) was microinjected into 2-cell Xenopus embryos. No specific morphological phenotypes were observed at 8, 10 or 80 ng of oligonucleotides per embryo (n=34, 33 and 19, respectively). The CTGF gene has been inactivated in the mouse by S. Ivkovic and K. M. Lyons (UCLA) and although the mice die perinatally, the overall body patterning is not affected by the mutation (personal communication). Thus, even if the morpholino caused phenotypes similar to those seen in mouse, we would not have expected to observe them by examination of Xenopus embryos at the tailbud tadpole stage.

1

©2003 Nature Publishing Group

supplementary infomation

Figure S2 CTGF constructs and expression of CTGF proteins in human 293T cells, Xenopus embryos and Drosophila S2 cells. a, Schematic representation of full-length Flag epitope-tagged CTGF containing its four structural modules: the IGF binding domain (IGFB), the CR/vwc domain, the Thrombospondin-1 domain and the C-terminal cystine knot (CT). Mutants lacking the CR domain (CTGF-∆CR and CTGF-CT), or consisting of the CR domain only (CTGF-CR) are shown. After cleavage of the signal peptide (dashed line), proteins retain twenty amino acids of the N-terminal part of Chordin (N-chd) followed by a Flag epitope sequence. The Chordin epitope tag allowed us to perform immunoprecipitation assays using an anti-N-chd 1 polyclonal antibody . b, Western blot using anti-Flag antibody of the proteins represented in the previous panel. Proteins were produced by transient transfection in 293T cells. Note that full-length CTGF and CTGF-∆CR are partially cleaved and N-terminal fragments of 20 kDa and 18 kDa are generated, respectively. In other experiments the cleavage products were more prevalent than the unprocessed proteins. c, Expression of CTGF and CTGF-∆CR in Xenopus embryonic cells. After injection of 100 pg CTGF or CTGF-∆CR mRNA into each blastomere of the four-cell stage Xenopus, animal halves were dissected at stage 9 and cells cultured overnight in 2 calcium-magnesium-free media . Supernatant was collected, centrifuged and western blot using anti-Flag antibody performed. Note that the proteins produced in Xenopus were not cleaved. d, e, Large scale production and purification of fulllength CTGF. Expression of Flag-CTGF in Drosophila S2 cells was performed as 3 described . Briefly, Flag-CTGF was sub-cloned into pUAST vector. To establish a cell

line 5 βg of pUAST-CTGF were co-transfected with 1 βg of pCoHygro (Invitrogen, for hygromycin resistance) and 5 βg of pGal/pRmHa3, which contains the yeast Gal-4 protein under the control of the methalothioneine promoter. FuGENE (Roche) was used as transfection reagent and cells were selected with 250 βg/ml of Hygromycin for 60 days. For protein secretion, 500 µM of CuSO4 was added to induce Gal4, which in turn induced CTGF expression via its UAS promoter. Note that the flag-CTGF produced by S2 cells is full-length. e, Flag-CTGF in S2 cell secreted media was purified by anti-Flag affinity gel (Sigma) column and eluted with Flag peptide (Sigma). Fractions were analyzed by SDS-PAGE stained with Coomassie blue. The gel was loaded with aliquots of the input in the column (I), of the flow through (FT), of the first and third washes (W1, W3) and of the three elutions (E1, E2 and E3). This method yielded approximately 100 βg of pure CTGF per 30 ml of tissue culture medium. We found that the use of full-length affinity-purified CTGF was essential for the crosslinking, binding analysis and cell culture results shown here. The use of Drosophila S2 cells was crucial as human 293T cells secrete large amounts of proteolytically processed CTGF. It is not known whether this proteolytic cleavage, which occurs close to the center of the protein, has a biological role on CTGF function in vivo. 1. Larraín, J. et al. Development 127, 821–830 (2000). 2. Sive, H.L., Grainger, R.M. & Harland, R.M. Early development of Xenopus laevis: A laborator manual, (Cold Spring Harbor Laboratory Press, New York, 2000). 3. Garrity, P.A. et al. Neuron 22, 707-717 (1999).

2

©2003 Nature Publishing Group

supplementary infomation

β1 and CTGF: Induction of striking morphoFigure S3 Synergy between TGF-β logical changes in Mv1Lu cells. The combination of TGF-β1 (0.2 nM) and CTGF (3 nM) induced spherical fluid-filled aggregates of Mv1Lu cells after 12 hours of culture (panel a). Previous studies have implicated TGF-β1 and CTGF in angiogenesis14 . Immunocytochemistry was performed with the cell surface antigen5-6 CD31, also known as platelet-endothelial cell adhesion molecule or PECAM-1. As shown in panel a, it was found that, indeed, the spherical aggregates were PECAM-1 positive. Spherical aggregates of PECAM-1-positive cells were not observed in Mv1Lu cells treated with TGF-β1 or CTGF alone. Interestingly, in the pluripotent mouse embryonal carcinoma P19 cell line, addition of CTGF induced PECAM-1 positive cell aggregates without requiring the addition of TGF-β1 (Fig. 5f). The mouse P19 cell structures were positive for PECAM-1 and for von Willebrand Factor, another endothelial marker (Fig. 5f). Panel b shows that increased amounts of vWF protein were detected in Mv1Lu cells treated with CTGF and TGF-β1 by Western blot (lane 6); polyclonal anti-vWF (Dako Co) was used at 1:200, and purified vWF and mouse placental proteins were used as positive controls. Spherical structures are induced by CTGF alone in P19 cells (see Fig. 5f of main text) presumably because they express endogenous TGF-β1 signals, since P19 cells have higher basal luciferase levels than Mv1Lu cells when transfected with a TGF-β-inducible reporter (data not shown). Mv1Lu cells have been very useful in TGF-β research because they have an intact TGF-β signal transduction pathway7 yet express low levels of endogenous TGF-β signals, which strongly inhibit their proliferation8. Further experiments will be required

to determine whether Mv1Lu cells, which were established from fetal mink lung tissue, are of endothelial origin. If the aggregates induced in Mv1Lu cells treated with CTGF and TGF-β1 eventually are proven to form blood vessel-like spherical structures, they will provide a valuable system for the molecular dissection of the signals involved in angiogenesis. The CTGF gene has been inactivated by homologous recombination in the mouse and defects in angiogenesis are apparent in regions where CTGF and TGF-β are normally co-expressed, identifying CTGF as an essential mediator of angiogenesis in vivo (S. Ivkovic and K. M. Lyons personal communication). In gain-of-function experiments, CTGF ectopically expressed in chicken chorioallantoic membranes or in the rat cornea has been shown to promote angio3,4 genesis and neovascularization . Even if it is eventually demonstrated that these remarkable structures are not of vascular nature, the formation of these cell aggregates may still provide an interesting in vitro system for the study of mesenchymal to epithelial transformation under the control of extracellular signals. 1. 2. 3. 4. 5. 6. 7. 8.

Roberts, A.B. et al. Proc. Natl. Acad. Sci. U S A 83, 4167-4171 (1986). Dickson, M.C. et al. Development 121, 1845-1854. (1995). Shimo, T. et al. J. Biochem. (Tokyo) 126, 137-145 (1999). Babic, A.M., Chen, C.C. & Lau, L.F. Mol. Cell. Biol. 19, 2958-2966 (1999). Baldwin, H.S. et al. Development 120, 2539-2553 (1994). Vecchi, A. et al. Eur. J. Cell Biol. 63, 247-254 (1994). Cheifetz, S., Like, B. & Massagué, J. J. Biol. Chem. 261, 9972-9978 (1986). Tucker, R.F., Shipley, G.D., Moses, H.L. & Holley, R.W. Science 226, 705-707 (1984).

3

©2003 Nature Publishing Group

Connective-tissue growth factor (CTGF) modulates cell ... - Nature

Jul 22, 2002 - ber of the CCN family of secreted proteins, named after. CTGF, cysteine-rich 61 (CYR61), and nephroblastoma overexpressed (NOV) proteins.

862KB Sizes 0 Downloads 169 Views

Recommend Documents

Epidermal growth factor and serum activate distinct pathways ... - Nature
Jun 12, 2006 - National University of Singapore, 8 Medical Drive, MD7, Singapore. 117 597 .... monoclonal antibody and incubated with 1 μg of GSK-3 fusion.

Nitric Oxide Modulates Tumor Cell Death Induced by ...
using a cell death detection enzyme-linked immunosorbent assay kit. (Roche, Carnaxide, Portugal), according ... instructions (ECF Western Blotting kit, Amersham Biosciences), using primary antibodies for caspase-3 ...... Wang, X. and P. J. Robinson (

Growth Factor Upregulation during Obliterative
Raleigh, NC) were obtained from pathogen-free colonies and were housed and used in ... sion (using ImageQuant software), and a secondary endpoint, graft epi- thelialization .... and/or myofibroblast recruitment and activation as these events.

The effect of vascular endothelial growth factor and brain‐derived ...
from the site of injury [6]. ... adhered to charged slides (Superfrost Plus, ... *all comparisons P < 0.05, except 3 vs 4; †number of nerve fibres positive per ...

Increased nerve growth factor after rat plantar incision ...
After tissue damage a number of substances are released within the local tissue environment ... to the sensory nerve cell body (Richardson and Riopelle,. 1984).

On the growth factor for Hadamard matrices
Determinants. Preliminaries. Solution. The proposed idea. Pivots from the beginning. Pivots from the end. Numerical experiments. Pivot patterns. Summary-. References. Backward error analysis for GE −→ growth factor g(n,A) = maxi,j,k |a. (k) ij. |

Expression of Vascular Endothelial Growth Factor ...
Sep 20, 2004 - 2004;110;3699-3707; originally published online Sep 20, 2004; ... Administration of ... to Dr Suchitra Sumitran-Holgersson, Department of Transplantation Surgery B56, Huddinge University Hospital, S-141 86 Stockholm,.

Fibroblast Growth Factor 23 and Mortality among ...
ratio per unit increase in log-transformed cFGF-23 values, 1.8; 95% CI, 1.4 to 2.4) or in quartiles, with ... erated by Fresenius Medical Care North America. (Waltham, MA) in 2004 ..... osteomalacia express genes important in bone and mineral ...

Growth Facts, Factor Shares and Intellectual Property ...
in R&D (SEV-2015-0563) for financial support. .... separation is essential for our study of the effects of IPP capitalization on growth and the big ... counting (2) has focused on the change in the accounting treatment of IPP in the business sector.

Human CTGF 100-60 SDS.pdf
Retrying... Download. Connect more apps... Try one of the apps below to open or edit this item. Human CTGF 100-60 SDS.pdf. Human CTGF 100-60 SDS.pdf.

High-throughput gene silencing using cell arrays - Nature
Cell array as a functional genomics tool ... statistical analysis of the data acquired by microscope or ..... Brummelkamp TR, Bernards R and Agami R. (2002).

Design of a muscle cell-specific expression vector utilising ... - Nature
p21, p27 and p579,10 or constitutively active forms of the retinoblastoma gene ... VSMC -actin have been defined in the chicken,24,25 rat,26,27 mouse28–30.

The growth of the cell death field
... Tor Vergata, via. Tor Vergata 135,00133 Rome, Italy tel t39 6 20427299; fax t39 6 20427290; ...... ideas. SCI-Map is a ISI PC program running under. Microsoft .... 'Apopt(free ending)' and/or '(free) Cell (free ending) Death', unless otherwise ..

Cell Growth Inhibition and Gene Expression Induced by ...
Accessible online at: ... Department of Medicine and Clinical Oncology, Graduate School of Medicine, Chiba ...... 14 McBain JA, Eastman A, Nobel CS, Mueller.

Ontogenic growth of the haemopoietic stem cell ... - Semantic Scholar
rate, which has been shown to follow an allometric exponent of 3/4 .... Let us now deduce the best possible scaling for B(t). To ..... Organization Tech. Rep. Ser.

Musical Meter Modulates the Allocation of Attention across Time
ERPs elicited by tones presented at times of high and low metric strength in ... waveforms less than 250 msec after auditory and visual on- sets (Hillyard, 1985 ...

Greatest Common Factor/Greatest Common Factor
Greatest Common Factor/Greatest Common Factor - (20 Multiple Choices Questions) Quiz Assignment.pdf. Greatest Common Factor/Greatest Common Factor ...

Activation of methionine synthase by insulin-like growth factor-1 and ...
Jan 27, 2004 - cold unlabeled Hank's solution followed by 500 μl ... 1ml of ice-cold lysis buffer. ...... maintained by the Centers for Disease Control, found.

Activation of methionine synthase by insulin-like growth factor-1 and ...
Jan 27, 2004 - IGF-1 has been shown to promote recovery from ..... 1737–1741. 66 Stubbs G, Litt M, Lis E, Jackson R, Voth W, Lindberg A, Litt R. Adenosine ...