+ All Categories
Home > Documents > Hematopoiesis Controlled by Distinct TIF1γ and Smad4 Branches of the TGFβ Pathway

Hematopoiesis Controlled by Distinct TIF1γ and Smad4 Branches of the TGFβ Pathway

Date post: 21-Nov-2023
Category:
Upload: nyu
View: 0 times
Download: 0 times
Share this document with a friend
13
Hematopoiesis Controlled by Distinct TIF1 g and Smad4 Branches of the TGF b Pathway Wei He, 1 David C. Dorn, 2 Hediye Erdjument-Bromage, 3 Paul Tempst, 3 Malcolm A.S. Moore, 2 and Joan Massague ´ 1, * 1 Cancer Biology and Genetics Program and Howard Hughes Medical Institute 2 Cell Biology Program 3 Molecular Biology Program Memorial Sloan-Kettering Cancer Center, New York, NY 10021, USA *Contact: [email protected] DOI 10.1016/j.cell.2006.03.045 SUMMARY Tissue homeostasis in mammals relies on pow- erful cytostatic and differentiation signals deliv- ered by the cytokine TGFb and relayed within the cell via the activation of Smad transcription factors. Formation of transcription regulatory complexes by the association of Smad4 with re- ceptor-phosphorylated Smads 2 and 3 is a cen- tral event in the canonical TGFb pathway. Here we provide evidence for a branching of this pathway. The ubiquitious nuclear protein Tran- scriptional Intermediary Factor 1g (TIF1g) selec- tively binds receptor-phosphorylated Smad2/3 in competition with Smad4. Rapid and robust binding of TIF1g to Smad2/3 occurs in hemato- poietic, mesenchymal, and epithelial cell types in response to TGFb. In human hematopoietic stem/progenitor cells, where TGFb inhibits pro- liferation and stimulates erythroid differentia- tion, TIF1g mediates the differentiation response while Smad4 mediates the antiproliferative response with Smad2/3 participating in both responses. Thus, Smad2/3-TIF1g and Smad2/ 3-Smad4 function as complementary effector arms in the control of hematopoietic cell fate by the TGFb/Smad pathway. INTRODUCTION The transforming growth factor-b (TGFb) family of cyto- kines play central roles in many aspects of metazoan em- bryonic development and tissue homeostasis (Massague ´ et al., 2000; Shen and Schier, 2000; Sporn and Roberts, 1990). TGFb family members bind two types of membrane serine/threonine kinases, the type I and type II receptors, forming a heteromeric receptor complex. The type II receptor then phosphorylates and activates the type I re- ceptor, which in turn phosphorylates Smad transcription factors (Shi and Massague ´ , 2003). Of the eight Smad family members in human, five function as receptor substrates (RSmads). Smad2 and Smad3 do so as substrates of TGFb, nodal and activin receptors, and Smads 1, 5, and 8 as substrates of the receptors for bone morphogenetic proteins, myostatin and anti-muellerian hormone. Recep- tor-mediated phosphorylation triggers nuclear accumula- tion of RSmads and their binding to Smad4. Smad4, itself not a receptor substrate, is an essential partner of RSmads in transcriptional regulation of many genes. RSmad-Smad4 complexes bind a diverse group of DNA binding factors to achieve target gene selection and recruit transcriptional co- activators or corepressors for gene regulation (Feng and Derynck, 2005; Massague ´ et al., 2005). This general mech- anism underlies a large number of TGFb gene responses controlling cell proliferation, organization, and fate. Smad proteins consist of a conserved N-terminal DNA binding domain (or MH1 domain) containing a DNA binding site, a variable linker region, and a C-terminal MH2 domain. At different steps during signal transduction, the MH2 do- main binds cytoplasmic anchor proteins, TGFb receptors, partner Smads, nucleoporins, DNA binding cofactors, or transcriptional coregulators (Massague ´ et al., 2005). Receptor-mediated phosphorylation occurs at two serine residues in the extreme C-terminal sequence Ser-Val-Ser (Ser-Met-Ser in Smad2) of RSmads. This creates an acidic tail that binds to a basic pocket in the Smad4 MH2 domain, forming the RSmad-Smad4 complex (Wu et al., 2001). A decade-long focus on the central role of Smad4 as a shared partner of all RSmads and a mediator of many TGFb effects has sidestepped the possibility of alternative RSmads partners. However, evidence from Smad4-defi- cient organisms and tumor cell lines indicates that Smad4 is not required for all the actions of the TGFb family (Sirard et al., 1998; Subramanian et al., 2004; Wisotzkey et al., 1998). TGFb may also signal via MAPK and PI3K kinases, PP2A phosphatases, and Rho family members (Derynck and Zhang, 2003; Massague ´, 2000). Yet, Cell 125, 929–941, June 2, 2006 ª2006 Elsevier Inc. 929
Transcript

Hematopoiesis Controlled by DistinctTIF1g and Smad4 Branchesof the TGFb PathwayWei He,1 David C. Dorn,2 Hediye Erdjument-Bromage,3 Paul Tempst,3 Malcolm A.S. Moore,2

and Joan Massague1,*1Cancer Biology and Genetics Program and Howard Hughes Medical Institute2Cell Biology Program3Molecular Biology Program

Memorial Sloan-Kettering Cancer Center, New York, NY 10021, USA

*Contact: [email protected] 10.1016/j.cell.2006.03.045

SUMMARY

Tissue homeostasis in mammals relies on pow-erful cytostatic and differentiation signals deliv-ered by the cytokine TGFb and relayed withinthe cell via the activation of Smad transcriptionfactors. Formation of transcription regulatorycomplexes by the association of Smad4 with re-ceptor-phosphorylated Smads 2 and 3 is a cen-tral event in the canonical TGFb pathway. Herewe provide evidence for a branching of thispathway. The ubiquitious nuclear protein Tran-scriptional Intermediary Factor 1g (TIF1g) selec-tively binds receptor-phosphorylated Smad2/3in competition with Smad4. Rapid and robustbinding of TIF1g to Smad2/3 occurs in hemato-poietic, mesenchymal, and epithelial cell typesin response to TGFb. In human hematopoieticstem/progenitor cells, where TGFb inhibits pro-liferation and stimulates erythroid differentia-tion, TIF1g mediates the differentiation responsewhile Smad4 mediates the antiproliferativeresponse with Smad2/3 participating in bothresponses. Thus, Smad2/3-TIF1g and Smad2/3-Smad4 function as complementary effectorarms in the control of hematopoietic cell fateby the TGFb/Smad pathway.

INTRODUCTION

The transforming growth factor-b (TGFb) family of cyto-

kines play central roles in many aspects of metazoan em-

bryonic development and tissue homeostasis (Massague

et al., 2000; Shen and Schier, 2000; Sporn and Roberts,

1990). TGFb family members bind two types of membrane

serine/threonine kinases, the type I and type II receptors,

forming a heteromeric receptor complex. The type II

receptor then phosphorylates and activates the type I re-

ceptor, which in turn phosphorylates Smad transcription

factors (Shi and Massague, 2003). Of the eight Smad family

members in human, five function as receptor substrates

(RSmads). Smad2 and Smad3 do so as substrates of

TGFb, nodal and activin receptors, and Smads 1, 5, and 8

as substrates of the receptors for bone morphogenetic

proteins, myostatin and anti-muellerian hormone. Recep-

tor-mediated phosphorylation triggers nuclear accumula-

tion of RSmads and their binding to Smad4. Smad4, itself

not a receptor substrate, is an essential partner of RSmads

in transcriptional regulationof manygenes. RSmad-Smad4

complexes bind a diverse group of DNA binding factors to

achieve targetgeneselectionand recruit transcriptionalco-

activators or corepressors for gene regulation (Feng and

Derynck, 2005; Massague et al., 2005). This general mech-

anism underlies a large number of TGFb gene responses

controlling cell proliferation, organization, and fate.

Smad proteins consist of a conserved N-terminal DNA

binding domain (or MH1 domain) containing a DNA binding

site, a variable linker region, and a C-terminal MH2 domain.

At different steps during signal transduction, the MH2 do-

main binds cytoplasmic anchor proteins, TGFb receptors,

partner Smads, nucleoporins, DNA binding cofactors,

or transcriptional coregulators (Massague et al., 2005).

Receptor-mediated phosphorylation occurs at two serine

residues in the extreme C-terminal sequence Ser-Val-Ser

(Ser-Met-Ser in Smad2) of RSmads. This creates an acidic

tail that binds to a basic pocket in the Smad4 MH2 domain,

forming the RSmad-Smad4 complex (Wu et al., 2001).

A decade-long focus on the central role of Smad4 as

a shared partner of all RSmads and a mediator of many

TGFb effects has sidestepped the possibility of alternative

RSmads partners. However, evidence from Smad4-defi-

cient organisms and tumor cell lines indicates that

Smad4 is not required for all the actions of the TGFb family

(Sirard et al., 1998; Subramanian et al., 2004; Wisotzkey

et al., 1998). TGFb may also signal via MAPK and PI3K

kinases, PP2A phosphatases, and Rho family members

(Derynck and Zhang, 2003; Massague, 2000). Yet,

Cell 125, 929–941, June 2, 2006 ª2006 Elsevier Inc. 929

Figure 1. Phosphorylation-Dependent Interaction of Smad2/3 and TIF1g

(A) Scheme of GST-tagged linker (L) and MH2 region of Smad2 and its phosphorylation-site mutants. Two C-terminal Ser (S) residues were mutated to

Asp residues (marked in red) to mimic receptor-mediated phosphorylation.

(B and C) HeLa cell extracts were subjected to affinity purification with the indicated baits. Eluted proteins were resolved on SDS-PAGE and stained

with Coomassie blue. Bands were excised and identified by mass spectrometry analysis.

(D and E) COS-1 cells were transfected with vectors encoding TIF1g, Smad2, Smad3, or Smad4 in the presence of constitutively active [TbRI(AAD)] or

kinase-dead [TbRI(K-R)] TGFb type I receptor constructs, as indicated. Lysates were immunoprecipitated (IP) and subsequently immunoblotted (IB)

with antibodies indicated on the left. Protein expression was monitored by immunoblot analysis of total cell extracts (Input).

signaling via RSmads independently of Smad4 remains

possible, as suggested by the more severe phenotype of

mad (Smad1) mutants compared to medea (Smad4)

mutants in Drosophila (Wisotzkey et al., 1998), by differ-

ences in the phenotype of Smad2 null and Smad4 null

mice (Sirard et al., 1998), and by the prominent presence

of phospho-Smad2/3 in Smad4-defective pancreatic

cancer cells (Subramanian et al., 2004).

Here we report the identification of TIF1g (also known as

TRIM33, RFG7, PTC7, and Ectodermin) as a protein that

selectively binds receptor-activated Smads 2 and 3. Four

TIF1 family members (a to d) are known in mammals, and

orthologs are present in organisms through Drosophila

(Beckstead et al., 2001; Friedman et al., 1996; Khetchou-

mian et al., 2004; Le Douarin et al., 1996; Moosmann

et al., 1996). TIF1a binds nuclear hormone receptors and

functions as a coactivator of retinoic acid receptor (Le

Douarin et al., 1996). TIF1b is an intrinsic component of

the histone deacetylase N-CoR1/HDAC3 complex (Under-

hill et al., 2000) and functions as a corepressor for Kruppel-

associated box (KRAB) zinc-finger transcription factors

(Friedman et al., 1996; Moosmann et al., 1996; Schultz

et al., 2001) and a cofactor for HP1 proteins in heterochro-

matin-mediated gene silencing (Le Douarin et al., 1996).

TIF1g is not known to have any of these functions. Cued

by the observation that the TIF1g homolog in the zebrafish

is essential for blood formation (Ransom et al., 2004), we

investigated the role of TIF1g in the TGFb response of

human hematopoietic progenitor cells (Ohta et al., 1987).

930 Cell 125, 929–941, June 2, 2006 ª2006 Elsevier Inc.

We found that in these cells, Smad4 mediates growth inhi-

bition whereas TIF1g mediates erythroid differentiation in

response to TGFb, thus establishing separate effector

functions for these two branches of the Smad pathway.

RESULTS

Identification of TIF1g as a Smad2/3 Binding Protein

We developed an affinity-purification strategy to isolate

proteins that selectively bind to activated RSmads. Mu-

tant forms of Smad1, Smad2, and Smad3 were created

in which the C-terminal Ser residues at the�1 and�3 po-

sitions were mutated to Asp (S2D mutants). X-ray crystal

structure analysis and biochemical studies have shown

that these mutations mimic the receptor-phosphorylated

pSer-Xxx-pSer Smad C-terminal motif in driving RSmad-

Smad4 complex formation (Chacko et al., 2004; Qin

et al., 2001; Wu et al., 2001). Recombinant glutathione-

S-transferase (GST) fusion proteins containing the linker

and MH2 (L+MH2) region of either wild-type RSmads or

S2D mutants (Figure 1A) were immobilized and used in

large-scale affinity purification of human HeLa cell ex-

tracts. A reproducible pattern of eluted proteins was

detected by Coomassie staining of eluates from GST-

Smad2(L+MH2) (Figure 1B). A similar pattern was ob-

tained with GST-Smad3(L+MH2) as the bait (data not

shown), as may be expected from the similarity (91% se-

quence identity) between the L+MH2 regions of Smad2

and Smad3. Very few protein bands could be visualized

when using either wild-type or S2D mutant forms of GST-

Smad1(L+MH2) as the bait (data not shown).

As identified by mass spectrometry, the proteins that

bound equally well to wild-type and S2D Smad2/3 baits in-

cluded the transcriptional coactivator CBP, which is known

to bind RSmads (Massague et al., 2005); BRG1, BAF155,

BAF170, and OSA2, which are subunits of SWI/SNF chro-

matin remodeling complexes (Narlikar et al., 2002; Roberts

and Orkin, 2004) previously unknown to interact with

Smads; and the transcriptional coactivator NCoA3 (Spie-

gelman and Heinrich, 2004). A 140 kDa protein identified

as TIF1g drew our attention because it bound several-fold

more abundantly to Smad2(L+MH2)(S2D) than to the

wild-type bait (Figure 1B). TIF1g was also present in eluates

from the Smad3(L+MH2)(S2D) column (data not shown). Of

the four known mammalian TIF1 family members, only

TIF1g was isolated under these conditions. The molecular

size of the L+MH2 baits is close to that of Smad4 and would

obscure Smad4 on SDS-PAGE of the eluates. Smad4 was

identified by mass spectrometry analysis in the eluate from

a bait of smaller size, Smad2 MH2(S2D) (Figure 1C). No

other components were identified that bound preferentially

to the wild-type or (S2D) baits.

TIF1g Selectively Binds Activated Smad2/3

To determine whether Smads 2 and 3 bind to TIF1g in vivo,

and whether this interaction is stimulated by TGFb recep-

tor signaling, we transfected COS-1 cells with vectors en-

coding Flag-tagged TIF1g, HA-tagged Smad2 or Smad3,

and a constitutively active mutant form of type I TGFb re-

ceptor, TbRI(AAD) (Chen et al., 1998), or a kinase-inactive

mutant, TbRI(KR) (Wrana et al., 1994) (Figure 1D). TIF1g

complexes with both Smad2 (Figure 1D, left panel) and

Smad3 (Figure 1D, right panel) could be coimmunopreci-

pitated from these cells, and this interaction was stimu-

lated by the activated receptor. In similar experiments us-

ing HA-tagged Smad4, only a trace amount of the

overexpressed protein could be coimmunoprecipitated

with TIF1g, and this was not increased by the activated re-

ceptor (data not shown).

To directly compare the affinity of TIF1g for different

Smad proteins, we cotransfected cells with vectors en-

coding TIF1g, activated receptor, Smad2, and either

Smad3 or Smad4 (Figure 1E). Equivalent amounts of

Smad2 and Smad3 could be coimmunoprecipitated with

TIF1g (Figure 1E, left panel). In contrast, in cells express-

ing similar levels of exogenous Smad2 and Smad4,

a very small amount of Smad4 compared to Smad2 was

coimmunoprecipitated with TIF1g (Figure 1E, right panel).

These results indicated that TIF1g has high affinity for ac-

tivated Smads 2 and 3.

TIF1g and Smad2/3 Interaction Domains

TIF1a and TIF1b tested under the same conditions as

TIF1g failed to bind Smad2 (Figure 2A). TIF1 family mem-

bers share sequence similarity within the RBCC (Ring fin-

ger–B boxes–Coiled Coil) and PHD/bromodomain regions

(PB) but are quite divergent in the middle region (M) linking

these domains (Figure 2A). Indeed, domain deletion ex-

periments mapped the Smad binding region to the middle

region of TIF1g (Figure 2B). Recombinant portions of

these proteins were made in bacteria as fusion products,

purified, and tested for binding in vitro. A MBP (maltose

binding protein) fusion protein containing the linker and

MH2 domain of Smad2 [MBP-Smad2(L+MH2)] was able

to bind a GST fusion with the middle region and PHD/bro-

modomain of TIF1g but not a GST fusion with only the

PHD/bromodomain region (Figure 2C). The version

MBP-Smad2(L+MH2)(S2D), containing C-terminal acidic

mutations, showed a higher affinity for TIF1g in vitro

(Figure 2C). When overexpressed in cells, the middle re-

gion of TIF1g inhibited the Smad2/3-Smad4 interaction

(Figure 2D), suggesting competition (see below). These

results argue that the activated MH2 domain in Smads 2

and 3 specifically and selectively binds to the middle re-

gion of TIF1g through direct interactions (Figure 2E).

Coexistence of Smad2/3-TIF1g and Smad2/3-Smad4

Complexes

We raised anti-TIF1g polyclonal antibodies that recognize

the endogenous protein by immunoblotting, immunopre-

cipitation and cell immunofluorescence. TIF1g mRNA is

expressed in many tissues (Venturini et al., 1999). Accord-

ingly, we detected TIF1g in all cell types tested, including

primary human hematopoietic progenitors, mouse em-

bryo fibroblasts, and mouse T cells, as well as cell lines

derived from normal tissues and tumors (Figure 3 and

data not shown). Immunofluorescence staining using af-

finity-purified anti-TIF1g antibodies revealed that TIF1g

is localized in the nucleus, outside of nucleoli, in HaCaT

human keratinocytes and all other cell types tested

(Figure 3A, and data not shown). A punctate pattern coex-

isted with a more diffuse nuclear distribution, all of which

was specific as shown by loss of the signal upon depletion

of TIF1g by means of a short-hairpin RNA (shRNA) (Fig-

ure 3A). A punctate TIF1g nuclear pattern has also been

noted by others (Ransom et al., 2004).

Importantly, TGFb addition induced the formation of an

endogenous Smad2/3-TIF1g along with a Smad2/3-

Smad4 complex (Figure 3B). Formation of both complexes

was entirely dependent on TGFb addition. The relative

abundance of these two complexes was proportional to

the relative abundance of TIF1g and Smad4. Thus, primary

mouse embryo fibroblasts, human M091 acute myeloge-

nous leukemia (Figure 3B), and mouse T lymphocytes

(data not shown) revealed abundant Smad2/3-TIF1g and

Smad2/3-Smad4 complexes in response to TGFb, whereas

the Smad2/3-TIF1g complex was less abundant in HaCaT

keratinocytes and other cell lines that express high levels

of Smad4 relative to TIF1g (Figure 3C, and data not shown).

The Smad2/3-TIF1g complex was more abundant in

the Smad4-defective tumor cell lines MDA-MB-468

(Figure 3C), SW480 (Figure 3E), and BxPC3 (Figure S1A).

Formation of a TGFb-dependent Smad2/3-TIF1g com-

plex was also demonstrated by anti-Smad2/3 immuno-

blotting of anti-TIF1g immunoprecipitates (Figure S1A).

Cell 125, 929–941, June 2, 2006 ª2006 Elsevier Inc. 931

Figure 2. Interaction of TIF1g Middle Region with Smad2 MH2 Domain

(A) Top: representation of human TIF1a, TIF1b, and TIF1g and percent amino acid sequence identity of each domain. Bottom: COS-1 cells were trans-

fected with vectors encoding TIF1 family members, Smad2, and receptor constructs, as indicated. Lysate immunoprecipitates (IP) were immunoblot-

ted (IB) with antibodies as indicated. Protein expression was monitored by immunoblotting of total cell extracts (Input).

(B) COS-1 cells were transfected with vectors encoding TIF1g, TIF1g fragments, Smad2, and receptor construct as indicated. Lysates were immu-

noprecipitated (IP) and subsequently immunoblotted (IB) with antibodies as indicated. TIF1g protein domains are as shown in (E).

(C) Purified maltose binding protein (MBP) Smad fusion products were prebound to amylose beads and mixed with purified GST-tagged TIF1g protein

domains. MBP-tagged proteins bound to the beads were detected by Coomassie blue staining. GST-tagged proteins were detected by Western im-

munoblotting using anti-GST antibodies.

(D) HEK293 Cells were transfected with vectors encoding TIF1g or TIF1g fragments and receptor constructs, as indicated. Endogenous Smad2 was

immunoprecipitated, and the precipitates were immunoblotted as indicated.

(E) Scheme of Smad2- and TIF1g-interacting domains. The location of truncating mutations in zebrafish TIF1g (Ransom et al., 2004) is indicated by

arrowheads.

In order to compare the relative contribution of endoge-

nous Smad2 and Smad3 to the formation of TIF1g com-

plexes, we decreased the level of endogenous Smads 2

or 3 using specific shRNAs. The results show that both

Smad2 and Smad3 contribute to the formation of com-

plexes with TIF1g (Figure S1B).

TIF1g and Smad4 Competitively Share Smad2/3

Quantitation by immunoblotting using recombinant pro-

teins as standards indicated a content of 0.1 pmol of

TIF1g, 1.4 pmol of Smad4, and 0.6 pmol of Smad2 per

106 HaCaT cells (data not shown). The higher abundance

of Smad4 over TIF1g may explain, at least partly, the prev-

alence of the Smad2/3-Smad4 complex over Smad2/3-

TIF1g complex. Forced overexpression of Smad4 inhibited

932 Cell 125, 929–941, June 2, 2006 ª2006 Elsevier Inc.

the formation of the Smad2/3-TIF1g complex to near com-

pletion (Figure 3D). Conversely, TIF1g overexpression in-

hibited the Smad2/3-Smad4 interaction (Figures 3E, left

panel, and 2D). HaCaT cells formed more Smad2/3-

TIF1g complex when Smad4 was depleted using a specific

shRNA (Figure 3E, central panel). Furthermore, formation

of an endogenous Smad2/3-TIF1g complex in the

Smad4-defective cell line SW480 was inhibited by restora-

tion of Smad4 expression, whereas a truncated Smad4

construct [Smad4(1–363)] that does not bind Smad2/3

(Hata et al., 1997) lacked this effect (Figure 3E, right panel).

These results suggest that Smad4 and TIF1g competitively

share the pool of TGFb-activated Smad2/3.

Xenopus TIF1g, which is called ectodermin, was re-

cently suggested to act as a general inhibitor of TGFb

Figure 3. TGFb-Dependent Formation of Competing Smad2/3-TIF1g and Smad2/3-Smad4 Complexes

(A) HaCaT cells were stably infected with retroviruses encoding a TIF1g short hairpin RNA, sh(TIF1g). Parental and TIF1g-depleted HaCaT cells were

subjected to immunofluorescence analysis with anti-TIF1g antibodies. Nuclei were visualized by DNA staining with DAPI. TIF1g knockdown was eval-

uated by immunoblot analysis, with immunoblot of Smad2 as loading control (bottom panel).

(B and C) The indicated cell types were treated with (+) or without (�) 200 pM TGFb for 2 hr. Lysate immunoprecipitates were immunoblotted as

shown.

(D) HEK293 cells were transfected with vectors encoding full-length TIF1g, Smads, and TGFb receptor constructs, as indicated. Lysate immunopre-

cipitates were immunoblotted as indicated.

(E) HaCaT cells stably expressing exogenous TIF1g (left panel) or depleted of endogenous Smad4 using a Smad4 shRNA (central panel), and SW480

cells stably expressing a full-length Smad4 or a truncated Smad4(1–363) (right panel), were stimulated with 200 pM TGFb for 2 hr. Lysate immuno-

precipitates were immunoblotted as indicated.

(F) HaCaT cells stably infected by retroviruses encoding either empty vector or TIF1g were incubated with the indicated TGFb concentrations and

periods. RNA was subjected to Northern blot analysis and each band was quantified by phosphorimager. The relative levels of PAI-1 and CDKN1A

mRNAs, normalized against GAPDH, are plotted.

(G) MDA-MB-231 cells were stably infected with sh(TIF1g) retrovirus or empty vector. The level of TIF1g knockdown was evaluated by immunoblot

analysis of knockdown lysates and serial dilution of control lysates.

(H) Control (black symbols) and TIF1g-depleted MDA-MB-231 cells (green symbols) were treated with 10 pM TGFb for the indicated periods. RNA was

subjected to Northern blot analysis with the indicated probes, and the data are presented as in panel (F).

and BMP signaling by binding to Smad4 and causing

Smad4 ubiquitination and degradation (Dupont et al.,

2005). However, we observed that TIF1g has a strong

binding preference for Smad2/3 over Smad4 (Figures 1E

and 3D); TIF1g overexpression did not cause a decrease

in the level of endogenous Smad4 (Figures 2D and 3E,

left panel); TIF1g overexpression did not cause Smad4

ubiquitination under conditions in which Smad2 was

polyubiquinated by the Smad ubiquitin ligase Smurf2

(Figure S1C). Depletion of endogenous TIF1g by shRNA

did not affect the rate of Smad2 phosphorylation or de-

phosphorylation after TGFb stimulation, or the level of en-

dogenous Smad4 (Figure S1D).

Mild Inhibition of Smad4-Dependent Gene

Responses by TIF1g

We tested the ability of TIF1g to affect various TGFb gene

responses that depend on Smad4. These genes include

Cell 125, 929–941, June 2, 2006 ª2006 Elsevier Inc. 933

Figure 4. Expression of TIF1g and Activated Smad2/3 in Mouse and Human Hematopoietic Tissues

(A) Immunohistochemical analysis of mouse embryo E12.5 sections with anti-TIF1g and control antibodies. Sections were counterstained by H&E.

(B) Mouse E8.5 sections were stained with anti-TIF1g, monoclonal rabbit anti-phopho-Smad2 (S465/S467) or polyclonal rabbit anti-phopho-Smad3

(S423/S425) without counterstaining.

(C) Human cord blood CD34+ stem/progenitor cells were subjected to immunostaining with anti-TIF1g or Giemsa staining.

plasminogen activator inhibitor-1 (PAI-1), JUNB, and

CDKN1A encoding the CDK inhibitor p21Cip1/WAF. Ec-

topic overexpression of TIF1g in HaCaT cells partially in-

hibited the induction of PAI-1 and CDKN1A by TGFb (Fig-

ures 3F and S2A). In complementary experiments, MDA-

MB-231 cells subjected to depletion of endogenous

TIF1g by shRNA (Figure 3G) showed a mild enhancement

in the induction of CDKN1A and JUNB by TGFb (Figures

3H and S2B). These results argue that TIF1g is neither

a mediator nor a potent inhibitor of Smad4-dependent

TGFb gene responses.

Role of TIF1g in Human Erythropoiesis

The closest homolog of TIF1g in the zebrafish is encoded

by moonshine (mon), an essential gene in blood formation

(Ransom et al., 2004). The most carboxy-terminal muta-

tion reported in mon spares the RBCC domain but trun-

cates the middle region, which corresponds to the

Smad-interacting domain of TIF1g (refer to Figure 2E).

Based on these clues, we investigated the possible role

of TIF1g in mammalian hematopoietic cells and their re-

sponse to TGFb.

Immunohistochemical analysis of mouse embryo sec-

tions revealed a widespread pattern of TIF1g expression

with nuclear staining of most tissues throughout gestation

(Figure 4A for E12.5; data not shown for other stages).

TIF1g immunostaining was particularly pronounced in

934 Cell 125, 929–941, June 2, 2006 ª2006 Elsevier Inc.

round hematopoietic cells in yolk sac blood islands in

E8.5 embryos (Figure 4B, top), which are sites of primitive

hematopoiesis. Interestingly, these hematopoietic cells

were also strongly stained by anti-phospho-Smad2 anti-

bodies (Figure 4B, middle), suggesting an important role

of TGFb in hematopoiesis. Only weak staining of hemato-

poietic cells was observed in blood islands from anti-

phospho-smad3 antibodies (Figure 4B, bottom), which

may reflect a low total Smad3 expression level. Strong nu-

clear staining was also observed in CD34+ hematopoietic

stem/progenitor cells isolated from human umbilical cord

blood (Figure 4C).

To investigate the role of TIF1g in hematopoietic devel-

opment, human umbilical cord blood was utilized as

a source of CD34+ hematopoietic stem/progenitor cells

(Cohen and Nagler, 2004; Shizuru et al., 2005). These cells

were infected with retroviruses encoding TIF1g shRNA,

followed by selection of the transduced cell population.

This achieved a 70% drop in endogenous TIF1g mRNA

level, as determined using quantitative RT-PCR (data not

shown). Cells were then assayed for proliferation and dif-

ferentiation in several well-established assay systems

(Figure 5A).

When placed in methylcellulose semi-solid medium with

stem cell factor (SCF), interleukin-3 (IL-3), and erythropoi-

etin (EPO) as a differentiation factor, CD34+ hematopoietic

progenitors form burst colonies that progressively

Figure 5. TIF1g Mediates Erythroid Differentiation of Cord Blood Hematopoietic Stem/Progenitor Cells

(A) Assays used to test the effect of TIF1g and Smad proteins on hematopoiesis.

(B) CD34+ cells infected with shRNA vectors targeting SMAD2, SMAD3, SMAD4, or TIF1g were plated for colony-forming cell (CFC) assays in semi-

solid media, in triplicate. After 2 weeks, colonies were scored (top panel) and cells from each plate were subjected to secondary CFC assays (bottom

panel). Data are presented as mean ± SD (n = 3).

(C) Representative pictures from cytospin preparations of cells from primary CFC assays described in (B).

(D) CD34+ cells infected with the indicated shRNAs retroviruses were placed in liquid culture proliferation conditions, in the absence and presence of

80 pM TGFb. Cell numbers were counted 3 days later. Data are presented as mean ± SD (n = 3).

differentiate into erythrocytes in two weeks. In this colony-

forming assay, the knockdown of TIF1g did not have a ma-

jor effect on the number of burst-forming units-erythoid

(BFU-E) (Figure 5B, top) or size distribution of the colonies

(Table S1). However, the BFU-E colonies formed by

TIF1g-depleted cells were less differentiated, as deter-

mined by cytospin of plucked colonies examined for dif-

ferentiation (Figure 5C). Compared to 2 week control cul-

tures, which already had a large proportion of cells in the

final stages of erythroid differentiation, the TIF1g-depleted

cultures showed a shift toward earlier stages of differenti-

ation (Figure 5C). In secondary passages of these colonies

(refer to Figure 5A), the control cultures formed very few

erythroid colonies, indicating that the majority of cells in

these cultures had already entered the late differentiation

stages (Figure 5B, bottom). TIF1g depletion increased

secondary erythroid colony formation, including large

multifocal BFU-E, 7-fold compared to control transduced

cells (Figure 5B, bottom). Depletion of Smad2 or Smad3

resulted in small increases in secondary colony formation,

and depletion of Smad4 had no effect. Collectively,

these results argue that TIF1g promotes hematopoietic

stem/progenitor cell differentiation along the erythroid line-

age at the expense of expansion of the early BFU-E stages.

Distinct Roles of TIF1g and Smad4 in TGFb Action

Culture in liquid media containing SCF, thrombopoietin

(TPO), and Flt3 ligand (FL) has been shown to stimulate

the proliferation of CD34+ hematopoietic stem/progenitor

cells. TGFb addition profoundly inhibited cell proliferation

under these conditions (Figure 5D). TIF1g knockdown did

not protect against this effect whereas Smad4 knockdown

blunted the growth-inhibitory action of TGFb (Figure 5D).

Smad2 and Smad3 knockdown also significantly blunted

the growth-inhibitory action of TGFb (Figure 5D). These

results suggest that Smad2 and Smad3 together with

Smad4, but not TIF1g, mediate the growth-inhibitory re-

sponse to TGFb in hematopoietic stem/progenitor cells.

To analyze effects on erythroid differentiation, we

placed control or knockdown CD34+ hematopoietic

stem/progenitor cells in liquid media containing EPO and

SCF. Three days after sorting, most of the cells were still

in the early stages of differentiation (i.e., proerythroblast

and basophilic erythroblast stages) (Figures 6A and 6C).

Cell 125, 929–941, June 2, 2006 ª2006 Elsevier Inc. 935

Figure 6. Distinct Roles of TIF1g and Smads in TGFb-Induced Growth Inhibition and Erythroid Differentiation(A and C) CD34+ cells infected with different shRNA retroviruses against TIF1g (A) or against the indicated components (C) were sorted and placed in

liquid culture differentiation conditions, in the absence or presence of TGFb (40 pM). Cell numbers were counted at day 3. The differentiation state of

the cells was determined by analysis of cytospin preparations. Data are presented as mean ± SD (n = 3).

(B and D) Number of erythrocytes in the ‘‘+TGFb’’ condition from experiments described in (A) and (C).

(E) Representative pictures of cytospin preparations from experiments described in (C).

Knockdown of TIF1g or Smads 2, 3, or 4 had little effect on

the differentiation state of these cells at this early stage, al-

though a trend toward differentiation was apparent in

Smad4 knockdown cells and an opposite trend in the

other knockdown cells (Figure 6C). Addition of TGFb at

the start of the 3 day incubation markedly accelerated

the differentiation of hematopoietic stem/progenitor cells

into erythrocytes and concurrently inhibited cell prolifera-

tion (Figures 6A–6E). Note that erythrocytes forming after

3 days in these assays would have originated from pro-

genitors (CFU-E) committing to differentiation on the first

day in culture. Therefore, effects on cell differentiation

would correspond to changes in the rate of differentiation

of committed progenitors, whereas effects on cell prolifer-

ation would correspond to changes in cells that remain in

the progenitor state for most of the 3 day incubation.

Smad4 knockdown blunted the growth-inhibitory effect

of TGFb but did not diminish the TGFb-dependent accu-

mulation of erythrocytes (Figures 6C and 6D). In contrast,

TIF1g knockdown with two different shRNAs strongly in-

hibited the TGFb-dependent accumulation of erythro-

cytes but did not diminish the growth-inhibitory effect of

TGFb (Figures 6A and 6B). Knockdown of Smad2 or

Smad3 decreased both the TGFb-dependent accumulation

of erythrocytes and the growth-inhibitory effect (Figures

6C and 6D). Addition of a TGFb type I receptor kinase

936 Cell 125, 929–941, June 2, 2006 ª2006 Elsevier Inc.

inhibitor SB431542 (Laping et al., 2002) as a blocker of au-

tocrine TGFb caused a trend toward less differentiation

and blocked the effect of exogenous TGFb (data not

shown).

Smad2/3-TIF1g Interaction in TGFb-Dependent

Erythroid Differentiation

To test the role of the Smad-TIF1g interaction in erythroid

differentiation we used constructs encoding the Smad2/3-

interacting middle region of TIF1g. Trimming of the N-ter-

minal or C-terminal portions of the middle region abol-

ished its ability to bind Smad2 in transfected cells

(Figure 7A). Therefore, the entire middle region is required

for this interaction. CD34+ hematopoietic stem/progenitor

cells were transfected with a vector encoding the TIF1g

middle region. The cultures were maintained in the pres-

ence of TGFb for 4 days in order to allow the accumulation

of erythrocytes. Overexpression of the TIF1g middle re-

gion, which served as a potential dominant-negative mu-

tant, indeed inhibited the effect of TGFb on erythroid

differentiation (Figure 7B). A construct encoding the mid-

dle region with a small C-terminal deletion (M-del3

construct), which fails to bind Smad2 (Figure 7A), did not

inhibit TGFb-induced differentiation (Figure 7B). This sug-

gests that the TIF1g middle region also recognizes Smads

in CD34+ cells.

Figure 7. Requirement of Smad2/3-TIF1g Complexes in TGFb-Dependent Erythroid Differentiation

(A) COS-1 cells were transfected with vectors encoding the middle region of TIF1g or fragments of this region, Smad2, and receptor construct, as

indicated. Lysate immunoprecipitates were immunoblotted as indicated.

(B and C) CD34+ cells transfected with the indicated plasmids were sorted and placed in liquid culture differentiation conditions, in the presence of

TGFb (40 pM). Cultures were maintained in the presence of TGFb for 4 days in order to allow the accumulation of erythrocytes. The differentiation state

of the cells was determined by analysis of cytospin preparations. Data are presented as mean ± SD (n = 3). In (B), TIF1g-Middle represents the Smad2/

3 binding middle region of TIF1g. TIF1g-M-del3 represents a C-terminal truncation mutant of this region (refer to A). In (C), rTIF1g represents

a sh(TIF1g)-resistant TIF1g mutant that has two silent mutations on the sh(TIF1g) targeting site (star). rTIF1g/a and rTIF1g/b constructs were

made by replacing the Smad2/3 binding middle region of TIF1g with the middle regions of TIF1a or b, which do not bind Smad2/3. The vectors

also encoded green or yellow fluorescent proteins (G and Y, respectively) in order to facilitate the isolation of doubly transfected cells.

(D) TIF1g as a branch in the TGFb-activated Smad pathway. The canonical TGFb signaling pathway involves ligand-induced receptor phosphorylation

of Smads 2 and 3, which allows the accumulation of phospho-Smad2/3 in the nucleus. Smad4 specifically recognizes receptor-phosphorylated

Smad2/3, forming transcriptional complexes that mediate antiproliferative responses in hematopoietic stem/progenitor cells. In the newly identified

branch of this pathway, TIF1g specifically recognizes receptor-phosphorylated Smad2/3 and mediates erythroid differentiation of hematopoietic

stem/progenitor cells. Additional effects of TIF1g may be mediated by TGFb- and Smad-independent inputs.

To further test the requirement of the Smad2/3-TIF1g in-

teraction in TGFb-dependent erythroid differentiation, we

checked the ability of Smad binding defective and wild-

type TIF1g constructs to rescue TGFb-dependent ery-

throid differentiation in TIF1g-depleted CD34+ hematopoi-

etic stem/progenitor cells (Figure 7C). We created

a sh(TIF1g)-resistant version of TIF1g (rTIF1g) by introduc-

ing silent mutations into the cDNA region that is targeted

by the sh(TIF1g). We then used this construct to create chi-

meras in which the Smad binding middle region of TIF1g

was replaced with the nonbinding middle region of TIF1a

or TIF1b (rTIF1g/a and rTIF1g/b constructs; Figure 7C).

The expression level of these chimeras in transfected

HEK293 cells was equal to or higher than that of the wild-

type TIF1g (data not shown). These constructs were engi-

neered into an IRES green fluorescent protein vector and

the sh(TIF1g) into a yellow fluorescent protein vector, in

order to allow sorting of doubly transfected cells. In 4 day

differentiation assays in the presence of TGFb, sh(TIF1g)-

transfected CD34+ cells showed a diminished erythroid

maturation compared to control transfectants (Figure 7C).

Cotransfection of rTIF1g rescued the ability of TGFb to

stimulate erythroid differentiation in sh(TIF1g)-transfected

CD34+ cells, providing further evidence of the specificity

Cell 125, 929–941, June 2, 2006 ª2006 Elsevier Inc. 937

of this sh(TIF1g) vector. More importantly, cotransfection

of the rTIF1g/a or rTIF1g/b constructs did not reverse the

effect of sh(TIF1g) on TGFb-induced differentiation

(Figure 7C). We conclude that the Smad2/3-TIF1g-inter-

acting region is important for TIF1g to mediate the effect

of TGFb on erythroid differentiation. Although the only

known function of this region is to bind Smad2/3, we can-

not exclude the possibility that another protein might bind

to this same region and mediate the TGFb effect.

DISCUSSION

Novel Smad Interactions

C-terminal phosphorylation of RSmad proteins by TGFb

receptor kinases is a critical event in signal transduction

because it creates a docking site for Smad4 allowing the

assembly of signaling complexes (Shi and Massague,

2003). Smad4 was the only factor known to occupy this

key position in the pathway. Searching for new partners

of activated Smads, we have identified TIF1g as an alter-

native effector of TGFb-activated Smad2/3. TIF1g is a nu-

clear protein expressed in most tissues during mouse em-

bryo development and in the adult. TIF1g competitively

shares Smad2/3 with Smad4, mediating signaling func-

tions that are distinct from those of the classical Smad2/

3-Smad4 complex (Figure 7E). We show a TGFb-depen-

dent interaction between endogenous Smad2/3 and

TIF1g alongside the Smad2/3-Smad4 interaction in differ-

ent mammalian cell types. Thus, TIF1g could play a role in

TGFb signaling in different contexts.

TIF1g and Smad4 were the only proteins from HeLa cell

extracts that preferentially bound to activated Smad2/3 in

protein affinity-chromatography experiments. Smad2/3

binding maps to the region of TIF1g linking the RBCC

domain and the PHD/bromodomain. This region is poorly

conserved in TIF1a and TIF1b, which do not bind Smad2/

3. Our protein purification approach also yielded a class of

proteins that bind equally well to the basal and activated

forms of Smads 2 and 3. These proteins include the tran-

scriptional coactivators CBP and NCoA3 (Spiegelman

and Heinrich, 2004) and several subunits of the SWI/SNF

ATP-dependent nucleosome remodeling complex (Narli-

kar et al., 2002; Roberts and Orkin, 2004). The important

role of CBP as a transactivation cofactor of RSmads is

well established (Feng and Derynck, 2005; Massague

et al., 2005). The previously unknown ability of Smad2/3

to interact with SWI/SNF components and NCoA3 sug-

gests a larger role of Smad proteins in chromatin remodel-

ing than previously appreciated.

A TGFb-Dependent Smad2/3-TIF1g Complex

Formation of an endogenous Smad2/3-TIF1g complex is

strictly dependent on TGFb stimulation. In vitro, however,

TIF1g can bind to the basal-state Smad2 and Smad3

MH2 domains, albeit less avidly than it binds to these pro-

teins in the activated state. Thus, TIF1g may recognize the

C-terminal motif pSer-Met/Val-pSer (or its mimic Asp-Met/

Val-Asp) in the context of other contacts with the MH2

938 Cell 125, 929–941, June 2, 2006 ª2006 Elsevier Inc.

domain. These properties are also characteristic of the

Smad2/3-Smad4 interaction (Shi and Massague, 2003).

Consistent with these similarities, we find that TIF1g and

Smad4 competitively share the pool of activated Smad2/

3 in TGFb-stimulated cells. The relative abundance of

these two complexes depends on the relative abundance

of TIF1g and Smad4 in a cell. Overexpression of TIF1g or

Smad4 inhibits binding of Smad2/3 to the other, and deple-

tion of one augments the binding of Smad2/3 to the other.

Smad4-deficient tumor cells form a high level of Smad2/3-

TIF1g complex, whereas cells with a high endogenous

Smad4:TIF1g ratio show a high ratio of Smad2/3-Smad4

over Smad2/3-TIF1g complexes in response to TGFb.

This ability to compete for Smad2/3 notwithstanding,

our evidence argues against TIF1g acting primarily as

a negative regulator of Smad4. Even under conditions in

which TIF1g overexpression markedly decreased the level

of endogenous Smad2/3-Smad4, TIF1g caused only

a small decrease in Smad4-dependent gene responses.

We have not been able to obtain evidence that TIF1g tar-

gets Smad4 for ubiquitination and degradation, as was re-

cently proposed by others for the human and Xenopus pro-

teins (Dupont et al., 2005). We observed no decrease in the

levels of endogenous or cotransfected Smad4 when TIF1g

was overexpressed by transfection, and no increase in

Smad4 upon RNAi-mediated TIF1g depletion. We de-

tected an interaction between TIF1g and Smad4 under

conditions of protein overexpression, but the level of this

complex was very low compared to the level of Smad2/

3-TIF1g complex on TGFb stimulation. The functional

consequences of genetically depleting TIF1g or Smad4

in hematopoietic progenitor cells are also inconsistent

with an anti-Smad4 role of TIF1g. It is possible that the pre-

viously reported inhibitory effect of TIF1g on TGFb signal-

ing in Xenopus embryos (Dupont et al., 2005) involves an

anti-Smad4 mechanism that is unique to that system.

Distinct Effector Roles of TIF1g and Smad4

in TGFb-Dependent Erythropoiesis

Based on sequence similarity, chromosomal location, and

exon boundaries, human TIF1g is closely related to zebra-

fish moonshine (mon) (Ransom et al., 2004), which was

identified as a gene required for blood formation. Muta-

tions in mon disrupt both embryonic and adult hematopoi-

esis, as mutant hemangioblasts fail to express hematopoi-

etic transcription factors, and undergo apoptosis. All

reported mon mutations truncate the middle region (Ran-

som et al., 2004), which we show to be a Smad2/3 binding

region that cannot be trimmed without loss of this func-

tion. As TGFb controls the proliferation and differentiation

of hematopoietic progenitor cells (Ruscetti et al., 2005;

Ohta et al., 1987), and Smads are implicated in erythropoi-

esis (Davidson and Zon, 2000; Epperly et al., 2005; Lars-

son and Karlsson, 2005; Schmerer and Evans, 2003), we

investigated these intriguing links.

In human umbilical cord blood CD34+ cells, which are

a significant source for allogeneic stem cell transplantation

(Cohen and Nagler, 2004), Smad2/3 and TIF1g stimulate

erythroid differentiation in response to TGFb whereas

Smad2/3 and Smad4 mediate the antiproliferative re-

sponse. RNAi-mediated deletion of TIF1g interfered with

the differentiative response of these cells to TGFb but

had no effect on the antiproliferative response. In contrast,

Smad4 depletion prevented the antiproliferative response

to TGFb without preventing the differentiative response.

Depletion of Smad2 and Smad3 inhibited both the differen-

tiative response and the antiproliferative response. Our re-

sults also suggest the existence of TGFb- and Smad-inde-

pendent inputs in the control of hematopoietic stem/

progenitor cell development by TIF1g (Figure 7D).

Smad2/3-TIF1g complexes, like Smad2/3-Smad4,

could influence cell behavior through transcriptional regu-

lation. Other TIF1 proteins serve as scaffolds for the as-

sembly of nuclear protein complexes via the RBCC protein

interaction domain (Peng et al., 2002). Comparative tran-

scriptomic analyses are underway to identify TIF1g-de-

pendent TGFb gene responses that may mediate erythroid

differentiation. However, whether TIF1g is a general medi-

ator of cell differentiation by the TGFb family is unknown. It

should also be noted that the hematopoietic collapse of

TIF1g-deficient zebrafish occurs without gross defects in

earlier developmental events that are known to require

extensive TGFb signaling (Ransom et al., 2004).

TIF1g is on human chromosome 1p13, which is an ac-

tive breakpoint for chromosomal abnormalities in cancer

(Johansson et al., 1994; Ng et al., 1999; Sawyer et al.,

2002). Translocations resulting in a fusion of TIF1g and

RET, which encodes a receptor tyrosine kinase, occur in

childhood papillary thyroid carcinomas (Klugbauer and

Rabes, 1999). Thus, alterations in TIF1g-dependent differ-

entiative functions might contribute to carcinogenesis. The

present findings may also have implications for pancreatic

cancers and other tumors that suffer loss of Smad4 while

retaining TGFb receptors, Smad2/3, and TIF1g (Subrama-

nian et al., 2004). Smad4-defective cancer cells are profi-

cient at forming Smad2/3-TIF1g complexes, which could

participate in the pro-tumorigenic responsiveness of

tumor cells to TGFb.

In sum, our results suggest that TIF1g associates with

Smad2/3 to stimulate erythroid differentiation of hemato-

poietic stem/progenitor cells whereas Smad4 associates

with Smad2/3 to inhibit the proliferation of these cells in re-

sponse to TGFb. The widespread expression of TIF1g and

the ability of different cell types to form Smad2/3-TIF1g

complexes suggest that this complex may play other

important roles in TGFb action.

EXPERIMENTAL PROCEDURES

Affinity Purification of HeLa Extracts and Protein Identification

For affinity purification, one liter of HeLa-S3 cells (about 560 million)

from National Cell Culture Center was lysed by sonication in 2 ml of

HKG lysis buffer (10 mM HEPES pH 7.9, 100 mM KCl, 10% Glycerol,

5 mM MgCl2, 10 mM NaF, 20 mM b-glycerolphosphate, 1 mM

Na3VO4, 0.2% NP-40) supplemented with protease inhibitors cocktail

(Roche), then precleared with 100 mg GST and centrifuged at 50,000 g

for 30 min. The supernatants were incubated with glutathione-

Sepharose prebound with 50 mg proteins for 4 hr at 4ºC on a rotating

platform. The bound proteins were eluted by the elution buffer (50

mM HEPES pH 7.4, 100 mM NaCl, 2 mM DTT, 0.15% Sarcosyl) at

room temperature for 10 min. The eluted proteins were subjected to

SDS-PAGE and Coomassie blue staining. The bands were excised

from the gel and proteins identified by peptide mass fingerprinting us-

ing matrix-assisted laser desorption/ionization-time-of-flight (MALDI-

TOF) mass spectrometry (MS).

Anti-TIF1g Antibodies

Purified N-terminal His6-tag TIF1g-M (456–887) and TIF1g-PB (888–

1127) proteins of human TIF1g were used to immunize rabbits for

antisera (Bethyl Labs), which were further affinity purified using col-

umns packed with antigen-conjugated Sepharose following standard

protocols.

Retroviral Transduction of CD34+ Cells

Human cord blood CD34+ cells were prestimulated for 48 hr in serum-

free Quality Biological serum-free–60 (QBSF) medium (Quality Biolog-

ical) supplemented with SCF (100 ng/ml) (Peprotech), FL (100 ng/ml)

(Imclone), and TPO (100 ng/ml) (Peprotech). High-titer retroviral super-

natants are applied immediately to the CD34+ cells on retronectin-

coated 6-well plates (Takara). Two consecutive transduction rounds

were performed 8 hr apart in polybrene (4 mg/ml). Transduction rates

of >30% were routinely obtained with CD34+ cells under these condi-

tions. Transduced cells were allowed to recover for 48 hr with fresh

medium and selected by FACS sorting.

Transfection of CD34+ Cells by Electroporation

One or two million CD34+ cells were resuspended in buffer provided in

human CD34 cell nucleofector kit (Amaxa), mixed with the desired

amount of plasmid DNA, and electroporated with U-08 program of

nucleofector (Amaxa) as suggested by the manufacturer.

Colony-Forming Cell Assays

Colony-forming cell (CFC) assays were performed in triplicate in 35

mm plates using 1.2% methylcellulose (Dow Chemical), 30% FBS,

57.2 mM b-mercaptoethanol, 2 mM glutamine, 0.5 mM hemin (Sigma),

20 ng/ml IL-3 (Peprotech), 20 ng/ml SCF, and 6 U/ml EPO (Ortho Bio-

tech). Colonies were scored 14 days after plating. Five colonies were

plucked and pooled to cytospin and stain for Wright-Giemsa to deter-

mine the differentiation status of the cells, or all the colonies were re-

suspended and replated for secondary CFC assays.

Liquid Culture Assays for TGFb-Induced Growth Inhibition

and Erythroid Differentiation

Cytokine-driven liquid expansion cultures were performed with FACS-

sorted GFP-positive CD34+ cord blood cells (1.5 3 104 cells/ml) in

QBSF serum-free medium, and 100 ng/ml SCF, FL, and TPO in the

absence and presence of 80 pM TGFb. After 3 days in liquid expansion

culture, cells were counted by trypan blue exclusion staining.

Cytokine-driven liquid erythroid differentiation cultures were per-

formed as described below based on whether transduction of retrovi-

ruses or transfection by electroporation were used to introduce desired

genes into CD34+ cells. For transduction of retroviruses, CD34+ cells

were stimulated for 48 hr as described above and a cytokine cocktail

of SCF (100 ng/ml) and EPO (6 U/ml) was added during transduction

for 48 hr. After a total of 4 days of stimulation and transduction, cells

were sorted and assayed for 3 days in SCF (100 ng/ml) and EPO (6 U/

ml). For transfection by electroporation, CD34+ cells were stimulated

for 72 hr and a cytokine cocktail of IL-3 (100 ng/ml), SCF (100 ng/ml),

and EPO (6 U/ml) was added during transfection for 48 hr. After a total

of 5 days of stimulation and transfection, cells were sorted and assayed

for 4 days in IL-3 (100 ng/ml), SCF (100 ng/ml), and EPO (6 U/ml).The

erythroid differentiation status was determined by assessing cytospin

preparations of the cultures stained with Wright-Giemsa and counting

cells in different fields of view for a total of 500 to 600 cells.

Cell 125, 929–941, June 2, 2006 ª2006 Elsevier Inc. 939

Subsequently, absolute cell numbers were determined through count-

ing by trypan blue dye exclusion staining.

Supplemental Data

Supplemental Data include Experimental Procedures, two figures, and

one table and can be found with this article online at http://www.cell.

com/cgi/content/full/125/5/929/DC1/.

ACKNOWLEDGMENTS

We are indebted to P. Rubinstein and L. Dobrila (New York Blood Cen-

ter) for cord blood; W. Mark, F.J. Rauscher III, and R. Agami for re-

agents; E. Montalvo for technical assistance; and Y. Kang, H.V. Le,

and current members of the Massague lab for insightful discussions.

We acknowledge the assistance of the MSKCC Genomics, Molecular

Cytology, and Flow Cytometry Core Facilities. This work was sup-

ported by NIH grants (R01 HL61401 and P01 CA59350), the Leukemia

and Lymphoma Society Specialized Centers for Research grants, and

the Gar Reichman Fund of the Cancer Research Institute to M.A.S.M.,

an NIH grant (R01 CA34610) to J.M., and a Cancer Center Support

Grant (P30 CA08748). J.M. is an Investigator of the Howard Hughes

Medical Institute.

Received: October 27, 2005

Revised: February 26, 2006

Accepted: March 27, 2006

Published: June 1, 2006

REFERENCES

Beckstead, R., Ortiz, J.A., Sanchez, C., Prokopenko, S.N., Chambon,

P., Losson, R., and Bellen, H.J. (2001). Bonus, a Drosophila homolog of

TIF1 proteins, interacts with nuclear receptors and can inhibit be-

taFTZ-F1-dependent transcription. Mol. Cell 7, 753–765.

Chacko, B.M., Qin, B.Y., Tiwari, A., Shi, G., Lam, S., Hayward, L.J., De

Caestecker, M., and Lin, K. (2004). Structural basis of heteromeric

smad protein assembly in TGF-beta signaling. Mol. Cell 15, 813–823.

Chen, Y.G., Hata, A., Lo, R.S., Wotton, D., Shi, Y., Pavletich, N., and

Massague, J. (1998). Determinants of specificity in TGF-beta signal

transduction. Genes Dev. 12, 2144–2152.

Cohen, Y., and Nagler, A. (2004). Umbilical cord blood transplantation–

how, when and for whom? Blood Rev. 18, 167–179.

Davidson, A.J., and Zon, L.I. (2000). Turning mesoderm into blood: the

formation of hematopoietic stem cells during embryogenesis. Curr.

Top. Dev. Biol. 50, 45–60.

Derynck, R., and Zhang, Y.E. (2003). Smad-dependent and Smad-in-

dependent pathways in TGF-beta family signalling. Nature 425, 577–

584.

Dupont, S., Zacchigna, L., Cordenonsi, M., Soligo, S., Adorno, M.,

Rugge, M., and Piccolo, S. (2005). Germ-layer specification and con-

trol of cell growth by Ectodermin, a Smad4 ubiquitin ligase. Cell 121,

87–99.

Epperly, M.W., Cao, S., Goff, J., Shields, D., Zhou, S., Glowacki, J.,

and Greenberger, J.S. (2005). Increased longevity of hematopoiesis

in continuous bone marrow cultures and adipocytogenesis in marrow

stromal cells derived from Smad3(�/�) mice. Exp. Hematol. 33, 353–

362.

Feng, X.H., and Derynck, R. (2005). Specificity and selectivity in TGF-

beta signaling through Smads. Annu. Rev. Cell Dev. Biol. 21, 659–666.

Friedman, J.R., Fredericks, W.J., Jensen, D.E., Speicher, D.W.,

Huang, X.P., Neilson, E.G., and Rauscher, F.J., 3rd. (1996). KAP-1,

a novel corepressor for the highly conserved KRAB repression domain.

Genes Dev. 10, 2067–2078.

940 Cell 125, 929–941, June 2, 2006 ª2006 Elsevier Inc.

Hata, A., Lo, R.S., Wotton, D., Lagna, G., and Massague, J. (1997). Mu-

tations increasing autoinhibition inactivate tumour suppressors

Smad2 and Smad4. Nature 388, 82–87.

Johansson, M., Dietrich, C., Mandahl, N., Hambraeus, G., Johansson,

L., Clausen, P.P., Mitelman, F., and Heim, S. (1994). Karyotypic charac-

terization of bronchial large cell carcinomas. Int. J. Cancer 57, 463–467.

Khetchoumian, K., Teletin, M., Mark, M., Lerouge, T., Cervino, M.,

Oulad-Abdelghani, M., Chambon, P., and Losson, R. (2004). TIF1delta,

a novel HP1-interacting member of the transcriptional intermediary

factor 1 (TIF1) family expressed by elongating spermatids. J. Biol.

Chem. 279, 48329–48341.

Klugbauer, S., and Rabes, H.M. (1999). The transcription coactivator

HTIF1 and a related protein are fused to the RET receptor tyrosine

kinase in childhood papillary thyroid carcinomas. Oncogene 18,

4388–4393.

Laping, N.J., Grygielko, E., Mathur, A., Butter, S., Bomberger, J.,

Tweed, C., Martin, W., Fornwald, J., Lehr, R., Harling, J., et al.

(2002). Inhibition of transforming growth factor (TGF)-beta1-induced

extracellular matrix with a novel inhibitor of the TGF-beta type I recep-

tor kinase activity: SB-431542. Mol. Pharmacol. 62, 58–64.

Larsson, J., and Karlsson, S. (2005). The role of Smad signaling in

hematopoiesis. Oncogene 24, 5676–5692.

Le Douarin, B., Nielsen, A.L., Garnier, J.M., Ichinose, H., Jeanmougin,

F., Losson, R., and Chambon, P. (1996). A possible involvement of TIF1

alpha and TIF1 beta in the epigenetic control of transcription by nuclear

receptors. EMBO J. 15, 6701–6715.

Massague, J. (2000). How cells read TGF-beta signals. Nat. Rev. Mol.

Cell Biol. 1, 169–178.

Massague, J., Blain, S.W., and Lo, R.S. (2000). TGFbeta signaling in

growth control, cancer, and heritable disorders. Cell 103, 295–309.

Massague, J., Seoane, J., and Wotton, D. (2005). Smad transcription

factors. Genes Dev. 19, 2783–2810.

Moosmann, P., Georgiev, O., Le Douarin, B., Bourquin, J.P., and

Schaffner, W. (1996). Transcriptional repression by RING finger protein

TIF1 beta that interacts with the KRAB repressor domain of KOX1.

Nucleic Acids Res. 24, 4859–4867.

Narlikar, G.J., Fan, H.Y., and Kingston, R.E. (2002). Cooperation be-

tween complexes that regulate chromatin structure and transcription.

Cell 108, 475–487.

Ng, K.C., Tan, A.M., Chong, Y.Y., Lau, L.C., and Lou, J. (1999). Con-

genital acute megakaryoblastic leukemia (M7) with chromosomal

t(1;22)(p13;q13) translocation in a set of identical twins. J. Pediatr.

Hematol. Oncol. 21, 428–430.

Ohta, M., Greenberger, J.S., Anklesaria, P., Bassols, A., and Mas-

sague, J. (1987). Two forms of transforming growth factor-beta distin-

guished by multipotential haematopoietic progenitor cells. Nature 329,

539–541.

Peng, H., Feldman, I., and Rauscher, F.J., 3rd. (2002). Hetero-oligo-

merization among the TIF family of RBCC/TRIM domain-containing

nuclear cofactors: a potential mechanism for regulating the switch

between coactivation and corepression. J. Mol. Biol. 320, 629–644.

Qin, B.Y., Chacko, B.M., Lam, S.S., de Caestecker, M.P., Correia, J.J.,

and Lin, K. (2001). Structural basis of Smad1 activation by receptor

kinase phosphorylation. Mol. Cell 8, 1303–1312.

Ransom, D.G., Bahary, N., Niss, K., Traver, D., Burns, C., Trede, N.S.,

Paffett-Lugassy, N., Saganic, W.J., Lim, C.A., Hersey, C., et al. (2004).

The zebrafish moonshine gene encodes transcriptional intermediary

factor 1gamma, an essential regulator of hematopoiesis. PLoS Biol.

2, E237. 10.1371/journal.pbio.0020237.

Roberts, C.W., and Orkin, S.H. (2004). The SWI/SNF complex–chro-

matin and cancer. Nat. Rev. Cancer 4, 133–142.

Ruscetti, F.W., Akel, S., and Bartelmez, S.H. (2005). Autocrine trans-

forming growth factor-beta regulation of hematopoiesis: many out-

comes that depend on the context. Oncogene 24, 5751–5763.

Sawyer, J.R., Thomas, E.L., Lukacs, J.L., Swanson, C.M., Ding, Y.,

Parham, D.M., Thomas, J.R., and Nicholas, R.W. (2002). Recurring

breakpoints of 1p13 approximately p22 in osteochondroma. Cancer

Genet. Cytogenet. 138, 102–106.

Schmerer, M., and Evans, T. (2003). Primitive erythropoiesis is regu-

lated by Smad-dependent signaling in postgastrulation mesoderm.

Blood 102, 3196–3205.

Schultz, D.C., Friedman, J.R., and Rauscher, F.J., 3rd. (2001). Targeting

histone deacetylase complexes via KRAB-zinc finger proteins: the PHD

and bromodomainsof KAP-1 forma cooperativeunit that recruits a novel

isoform of the Mi-2alpha subunit of NuRD. Genes Dev. 15, 428–443.

Shen, M.M., and Schier, A.F. (2000). The EGF-CFC gene family in

vertebrate development. Trends Genet. 16, 303–309.

Shi, Y., and Massague, J. (2003). Mechanisms of TGF-beta signaling

from cell membrane to the nucleus. Cell 113, 685–700.

Shizuru, J.A., Negrin, R.S., and Weissman, I.L. (2005). Hematopoietic

stem and progenitor cells: clinical and preclinical regeneration of the

hematolymphoid system. Annu. Rev. Med. 56, 509–538.

Sirard, C., de la Pompa, J.L., Elia, A., Itie, A., Mirtsos, C., Cheung, A.,

Hahn, S., Wakeham, A., Schwartz, L., Kern, S.E., et al. (1998). The tu-

mor suppressor gene Smad4/Dpc4 is required for gastrulation and

later for anterior development of the mouse embryo. Genes Dev. 12,

107–119.

Spiegelman, B.M., and Heinrich, R. (2004). Biological control through

regulated transcriptional coactivators. Cell 119, 157–167.

Sporn, M.B., and Roberts, A.B. (1990). TGF-beta: problems and pros-

pects. Cell Regul. 1, 875–882.

Subramanian, G., Schwarz, R.E., Higgins, L., McEnroe, G., Chakra-

varty, S., Dugar, S., and Reiss, M. (2004). Targeting endogenous trans-

forming growth factor beta receptor signaling in SMAD4-deficient

human pancreatic carcinoma cells inhibits their invasive phenotype1.

Cancer Res. 64, 5200–5211.

Underhill, C., Qutob, M.S., Yee, S.P., and Torchia, J. (2000). A novel

nuclear receptor corepressor complex, N-CoR, contains components

of the mammalian SWI/SNF complex and the corepressor KAP-1.

J. Biol. Chem. 275, 40463–40470.

Venturini, L., You, J., Stadler, M., Galien, R., Lallemand, V., Koken,

M.H., Mattei, M.G., Ganser, A., Chambon, P., Losson, R., and deThe,

H. (1999). TIF1gamma, a novel member of the transcriptional interme-

diary factor 1 family. Oncogene 18, 1209–1217.

Wisotzkey, R.G., Mehra, A., Sutherland, D.J., Dobens, L.L., Liu, X.,

Dohrmann, C., Attisano, L., and Raftery, L.A. (1998). Medea is a Dro-

sophila Smad4 homolog that is differentially required to potentiate

DPP responses. Development 125, 1433–1445.

Wrana, J.L., Attisano, L., Wieser, R., Ventura, F., and Massague, J.

(1994). Mechanism of activation of the TGF-beta receptor. Nature

370, 341–347.

Wu, J.W., Hu, M., Chai, J., Seoane, J., Huse, M., Li, C., Rigotti, D.J.,

Kyin, S., Muir, T.W., Fairman, R., et al. (2001). Crystal structure of

a phosphorylated Smad2. Recognition of phosphoserine by the MH2

domain and insights on Smad function in TGF-beta signaling. Mol.

Cell 8, 1277–1289.

Cell 125, 929–941, June 2, 2006 ª2006 Elsevier Inc. 941


Recommended