document

13

Click here to load reader

Transcript of document

Page 1: document

progress

nature genetics • volume 29 • october 2001 117

TGF-β signaling in tumor suppression andcancer progression

Rik Derynck1, Rosemary J. Akhurst2 & Allan Balmain2,3

Epithelial and hematopoietic cells have a high turnover and their progenitor cells divide continuously,making them prime targets for genetic and epigenetic changes that lead to cell transformation andtumorigenesis. The consequent changes in cell behavior and responsiveness result not only from genet-ic alterations such as activation of oncogenes or inactivation of tumor suppressor genes, but also fromaltered production of, or responsiveness to, stimulatory or inhibitory growth and differentiation fac-tors. Among these, transforming growth factor β (TGF-β) and its signaling effectors act as key deter-minants of carcinoma cell behavior. The autocrine and paracrine effects of TGF-β on tumor cells andthe tumor micro-environment exert both positive and negative influences on cancer development.Accordingly, the TGF-β signaling pathway has been considered as both a tumor suppressor pathwayand a promoter of tumor progression and invasion. Here we evaluate the role of TGF-β in tumor devel-opment and attempt to reconcile the positive and negative effects of TGF-β in carcinogenesis.

1Departments of Growth and Development, and Anatomy, Programs in Cell Biology and Developmental Biology, 2Mount Zion Cancer Research Institute,3Department of Biochemistry and Biophysics, University of California at San Francisco, San Francisco, California, USA. Correspondence should be addressedto R.D. (e-mail: [email protected]).

In mammals, there are three different TGF-βs, β1, β2 and β3,which are encoded by different genes and which all functionthrough the same receptor signaling systems1,2. Of these, TGF-β1is most frequently upregulated in tumor cells3,4 and is the focusof most studies on the role of TGF-β in tumorigenesis.

The TGF-β protein is released as an inactive ‘latent’ complex,comprising a TGF-β dimer in a non-covalent complex with twoprosegments, to which one of several ‘latent TGF-β–binding pro-teins’ is often linked5–7. This latent complex represents an impor-tant safeguard against ‘inadvertent’ activation, and may stabilizeand target latent TGF-β to the extracellular matrix, where it issequestered7,8. The matrix thus acts as a reservoir from which TGF-β can readily be recruited without the need for new synthesis.

The secretion of TGF-β as a latent complex necessitates theexistence of a regulated activation process, which is mostprobably mediated through the activities of proteases thatpreferentially degrade the TGF-β prosegments and therebyrelease the highly stable, active TGF-β dimer. Because plasminactivates latent TGF-β9,10 and plasminogen is converted intoplasmin at sites of cell migration and invasion11, we assumethat endothelial and tumor cells are exposed to active TGF-β.

Latent TGF-β can also be activated by the metalloproteasesMMP-9 and MMP-2 (ref. 12), which are frequently expressedby malignant cells, especially at sites of tumor cell inva-sion13,14. Other activation mechanisms might not depend onproteases. For example, the extracellular matrix proteinthrombospondin15,16 and the αvβ6 integrin, which isexpressed at the surface of epithelial cells in response toinflammation17, may mediate TGF-β activation through aconformational change in the TGF-β complex. Thus, differentmechanisms may regulate TGF-β activation in different physi-ological contexts, and tumor cells are well equipped to activateTGF-β locally.

Mechanisms of TGF-β signalingThe TGF-β receptors. As the mechanisms of TGF-β receptor sig-naling have been extensively reviewed recently18–23, we will sum-marize only the salient points (Fig. 1) before focusing on the roleof TGF-β in cancer cells. TGF-β signals through a heteromericcell-surface complex of two types of transmembrane serine/thre-onine kinases, named ‘type I’ and ‘type II’ receptors. Althoughthere is only one type II TGF-β receptor (TβRII), there are threetype I receptors (ALK1/TSR-1, ALK2/Tsk7L, ALK5/TβRI). Mostgene expression responses are presumably mediated by the TβRIreceptor. TβRII and TβRI show a widespread pattern of expres-sion and are expressed by tumor cells. After TGF-β binds to thereceptor complex, the TβRII kinase phosphorylates TβRI in the‘GS sequence’, which is located upstream from the kinasedomain. This phosphorylation activates the TβRI kinase thatmediates TβRI autophosphorylation and phosphorylation ofdownstream target proteins.

Several intracellular proteins have been shown to interact withTGF-β receptor complexes on the cell surface. Among these,FKBP12 interacts constitutively with the juxtamembrane domainof type I receptors and regulates the conformation of this domainand the signaling sensitivity24–27. Three proteins containing WDrepeats associate with and are phosphorylated by the receptorcomplex after ligand-induced activation and regulate TGF-βreceptor signaling. Whereas TRIP-1 interacts with TβRII (refs28,29), the Bα subunit of the protein phosphatase 2A interactswith type I receptors30, and STRAP interacts with both TβRIIand TβRI to decrease TGF-β signaling31,32. The regulatory rolesof these proteins are well documented, but there is no convincingevidence to show that these WD-repeat proteins act as effectorsof TGF-β responses, with the exception of the Bα subunit of pro-tein phosphatase 2A, which may be involved in growth arrestthrough inactivation of the S6 kinase pathway33.

©20

01 N

atu

re P

ub

lish

ing

Gro

up

h

ttp

://g

enet

ics.

nat

ure

.co

m© 2001 Nature Publishing Group http://genetics.nature.com

Page 2: document

progress

118 nature genetics • volume 29 • october 2001

Signaling through Smads. The only well-characterized signal-ing effector pathway that is initiated by activated TGF-β receptorsis provided by the Smads, a small family of structurally relatedproteins (Fig. 1) 19–23,34,35. Smad2 and Smad3 are activated viacarboxy-terminal phosphorylation by type I TGF-β receptorkinases and form heterotrimeric complexes with Smad4. Thesecomplexes then translocate into the nucleus and act as TGF-β-induced transcriptional activators of target genes. By contrast,Smad6 and Smad7 interfere with the activation of the effectorSmads and act as ‘inhibitory’ Smads. Both Smad6 and Smad7 caninteract with type I receptors, thus competitively preventing the‘receptor-activated’ Smads from being phosphorylated; Smad6also interferes with the formation of the heteromeric Smad com-plex. TGF-β signaling induces expression of Smad7, which pro-vides a TGF-β–induced negative feedback loop.

The TGF-β protein activates or represses the transcription ofdefined target genes, and many of these responses are immediateresponses to TGF-β receptor activation. So far, Smads have beenshown primarily to activate transcription19–23,34,35, but they arealso involved in TGF-β–mediated repression of transcrip-tion36–38. Smads also interact with many DNA-binding tran-scription factors; this interaction, together with theDNA-binding activity of Smads, specifies the promoter bindingand transcriptional activation21,34,35. For example, Smad3 caninteract with bZIP transcription factors such as c-Jun (refs.39–41), ATF-2 (refs. 42,43) or CREB (ref. 44); basic

helix–loop–helix transcription factors such as TFE3 (refs. 45,46);runt domain transcription factors such as AML-1, AML-2 andCBFA1 (refs. 37,44,47,48); nuclear receptors such as the vitaminD3 (ref. 49), glucocorticoid (ref. 50) and androgen receptors(refs. 38,51), and STAT-3 (ref. 52).

The receptor-activated Smads are linked to the general tran-scriptional machinery through a direct physical interactionwith the transcriptional coactivator CBP/p300, which is essen-tial for the transcriptional activation function of Smads53–55.The high-affinity binding of the Smad transcription complex toa defined promoter sequence is mediated primarily by the inter-acting transcription factor, although Smad3 and Smad4, butnot Smad2, have an intrinsic DNA-binding capability aswell39,56–59. A Smad-binding DNA sequence thus provides afavorable sequence context for Smad binding close to the high-affinity binding sequence for the Smad-interacting transcrip-tion factor34,56,58. This DNA context–dependent binding of aSmad to both the interacting transcription factor and the pro-moter DNA might explain why TGF-β activates only a subpop-ulation of the promoters that bind the transcription factor withwhich a Smad can interact.

Crosstalk with Smad signaling. The TGF-β–dependentrecruitment of Smad complexes to the transcription machin-ery also allows the recruitment of additional coactivators orcorepressors, which regulate the amplitude of transcriptionalactivation. Besides the interaction of the Smad complex with

Fig. 1 TGF-β-induced signaling through Smads, and several non-Smad signaling mechanisms. After ligand-induced activation of the receptor, Smad2 and/or Smad3interact transiently with the TβRI receptor (RI), and this interaction is stabilized by the FYVE protein SARA. Smad2 and Smad3 are phosphorylated on their C terminalsby TβRI, and then dissociate from the receptor to form a heterotrimeric complex comprising two receptor-activated Smads and Smad4. This complex then translocatesinto the nucleus, where it interacts at the promoter with transcription factors with sequence-specific DNA binding to regulate gene expression. The heteromeric Smadcomplex also interacts with the CBP/p300 transcriptional coactivator, which connects the Smad complex with the general transcription factors (GTF). Smad7 inhibitsactivation of Smad2 and/or Smad3 by the receptors, and Smad7 expression is induced on stimulation of one of several signaling pathways—for example, in response toEGF, interferon-γ (IFN-γ) or tumor necrosis factor-α (TNF-α). Several other signaling pathways also regulate both signaling by Smads and Smad-mediated gene expres-sion, as exemplified here by the activation of JNK and p38 MAP kinase signaling in response to various stress signals, and β-catenin signaling in response to Wnt pro-teins. TGF-β also induces activation of Ras, RhoB and RhoA, as well as of TAK1 and protein phosphatase 2A, which leads to the activation of several MAP kinasepathways and the downregulation of S6 kinase activity. The mechanisms of activation of these non-Smad signaling events and how they connect to the het-eromeric TGF-β receptor complex remain to be characterized.

RII RI

TGF-β

PP

Smad2/3

Smad4

P

p38

c-Jun

β-catenin

JNK

ATF2

stress signals Wnt

STAT1

IFN-γ

NF-kB

TNF-αEGF

MAPK JAK1

SMAD7

target genes

PP

GTF

S6Kp38 MAPKErk /MAPK

PP2ATAK1RhoARhoRas

JNK p160ROCK

©20

01 N

atu

re P

ub

lish

ing

Gro

up

h

ttp

://g

enet

ics.

nat

ure

.co

m© 2001 Nature Publishing Group http://genetics.nature.com

Page 3: document

Fig. 2 TGF-β acts on both the tumor cell and itsenvironment. Both tumor and stromal cellssecrete elevated levels of TGF-β that are self-perpetuating via a cycle of enhanced proteaseactivation of latent TGF-β, auto-induction ofTGF-β1 gene activity and recruitment of TGF-β–secreting cells such as macrophages andneutrophils. Most TGF-β activities, apart fromthe growth inhibition of early tumor cells,promote tumor progression. The TGF-β signal-ing pathway is thus a target for therapeuticintervention in invasive tumors. TGF-β activitycan be targeted by using endogenous pro-teins such as soluble betaglycan or decorin, orby using artificial agents such as antisenseoligonucleotides, antibodies or small-drugmolecules selective for specific components ofthe signaling pathway. ECM, extracellularmatrix; Fas-L, Fas ligand.

progress

nature genetics • volume 29 • october 2001 119

CBP/p300, Smad4 can also engageanother coactivator, named MSG1,into the transcription complex toenhance the Smad response60,61. Bycontrast, recruitment of a corepres-sor decreases the Smad and TGF-βresponsiveness. The corepressorsEvi-1 (ref. 62) and c-Ski (ref. 63,64),as well as SnoN (ref. 65), TGIF (ref.66), SNIP1 (ref. 67) and SIP1 (ref.68), can interact with Smad3 and/orSmad2 and inhibit TGF-β responses.Thus, the expression of Smad-inter-acting corepressors by tumor cellsmight block some TGF-β responses,including its inhibitory effect ongrowth, and contribute to tumorprogression.

The transcriptional activation by Smads, through their physicalinteractions and functional cooperativity with transcription factors,allows crosstalk with other signaling pathways19,21,69,70. For example,the activation of mitogen-activated protein (MAP) kinase pathways,which is commonly observed in tumor cells, and of Jun kinase or p38MAP kinase is likely to regulate TGF-β-induced transcription frompromoters with TGF-β-responsive AP-1 (c-Jun/c-Fos)– orCREB/ATF-binding sites and Smad-binding sites39,42,43,71–74.

Most notably, the expression of extracellular matrix proteins andproteases in response to TGF-β often requires an intact AP-1–bind-ing promoter sequence75,76, suggesting that there is transcriptionalcooperation of Smads with the AP-1 complex and a dependence ofthese TGF-β responses on Ras/MAP kinase and/or phosphatidyli-nositol-3-OH kinase (PI-3-K) signaling39,72,77. Ras/MAP kinasesignaling also induces expression of TGF-β1 (refs. 73,78,79), whichcan be enhanced further by TGF-β signaling73,80 and thus mayexplain the often-observed increase in expression of TGF-β1 bytumor cells. Because Ras signaling also induces the expression ofurokinase and the consequent generation of plasmin81, and acti-vates expression of the cell-surface protease MMP-9 (refs.12,82–84), Ras/MAP kinase signaling might enhance the cell-autonomous effects of TGF-β1 and the effects of TGF-β activationon the tumor micro-environment.

Ras signaling has also been proposed to inhibit TGF-β signal-ing. Specifically, extracellular signal–regulated kinase (Erk) MAPkinase has been shown to phosphorylate Smad2 and Smad3, andthis phosphorylation inhibits nuclear translocation of these TGF-β-activated Smads85. This mechanism might explain why in somecells with hyperactive Ras signaling the response to TGF-β isinhibited85,86. But other studies have not reported impairednuclear translocation of Smads in Ras-transformed cells or in cells

with activated MAP kinase signaling74,87, and impaired Smad sig-naling in Ras-transformed cells cannot be easily reconciled withthe cooperativity between Ras/MAP kinase signaling and TGF-βsignaling in tumor cell differentiation and behavior87,88. Finally,Jun N-terminal kinase (JNK)–mediated phosphorylation ofSmad3 enhances its activation and nuclear translocation74.Clearly, the crosstalk between these two pathways may depend onthe physiological cell context, a general theme in TGF-β signaling.

Several additional mechanisms of crosstalk with the Smadpathway may be relevant for the role of TGF-β signaling in can-cer cells. For example, signaling by Wnt differentiation factors,which direct tissue specification and growth control, can inter-sect with TGF-β/Smad signaling because Smad3 can interactdirectly with β-catenin and LEF/TCF—transcription regulatorsof Wnt signaling89,90. Virally encoded nuclear proteins may alsoregulate Smad-mediated gene expression through interactionswith the Smads at responsive promoters. For example, thehuman T cell leukemia virus type I (HTLV-1)–encoded Tax pro-tein is a potent repressor of Smad-mediated transcription91,whereas the hepatitis B virus–encoded nuclear pX proteinenhances Smad-mediated transcription in response to TGF-β92.Perhaps conceptually similar, menin, a putative tumor suppres-sor associated with multiple endocrine neoplasia type 1(MEN1), interacts with Smad3 in the nucleus, and its inactiva-tion suppresses Smad3-mediated transcription and TGF-β-induced responses93. The activation of some signaling pathways,including epidermal growth factor (EGF) receptor activation94,interferon-γ signaling through STATs95 and tumor-necrosis fac-tor-α (TNF-α)–induced activation of NF-κB96, induces theexpression of Smad7, which in turn inhibits TGF-β signalingthrough Smads (Fig. 1).

stromal cell

proteasesthrombospondin

integrins

latent TGF-β

active TGF-β

decorinbetaglycan

effects on tumor environmenteffects on tumor cell1. growth inhibition (early)

2. invasion (late)

1. stroma proteases ECM production

3. immune cells Fas-L activity NK cells T cells B cells

2. endothelial cells angiogenisis

tumor cell

©20

01 N

atu

re P

ub

lish

ing

Gro

up

h

ttp

://g

enet

ics.

nat

ure

.co

m© 2001 Nature Publishing Group http://genetics.nature.com

Page 4: document

progress

120 nature genetics • volume 29 • october 2001

Crosstalk with Smad signaling may also result from the abilityof TGF-β to activate signaling pathways independently of Smads(Fig. 1). TGF-β can activate Erk MAP kinase97–99, p38 MAPkinase42,43 and JNK71,74, although the extent and kinetics of acti-vation differ among different cell lines and types. The MAPkinase kinase kinase TAK1, which is rapidly activated by TGF-β100 but is also involved in other signaling pathways101–104, mayinitiate these signaling cascades. The activation of p38 MAPkinase and JNK can enhance Smad signaling through eitherSmad phosphorylation74 or the phosphorylation of c-Jun andATF-2 (refs. 42,43,71), which are transcription factors that coop-erate with Smad3, resulting in functional crosstalk with Smad-mediated transcription at defined promoters.

In addition, TGF-β can activate or stabilize the small GTPasesRhoA105 and RhoB106; these may in turn have roles in severalresponses to TGF-β, for example through a requirement of RhoBfor activation of JNK74. Finally, TGF-β induces an interaction ofprotein phosphatase 2A with S6 kinase, which regulates proteintranslation and growth control, decreasing its activity33.Although the mechanisms of activation by TGF-β and the rolesof these non-Smad signaling cascades remain to be better charac-terized, these observations indicate that inactivation of the Smadpathways may not leave the cell unresponsive to TGF-β.

TGF-β–mediated growth arrest and apoptosisMost pertinent to our understanding of the role of TGF-β in car-cinoma development is the fact that TGF-β is a potent inducer ofgrowth inhibition in several cell types, including epithelialcells107–109. One key event that leads to TGF-β–induced growtharrest is the induction of expression of the CDK inhibitorsp15INK4B (refs. 110,111) and/or p21CIP1 (ref. 112), depending onthe cell type. The inhibitor p21CIP1 interacts with complexes ofCDK2 and cyclin A or cyclin E and thereby inhibits CDK2 activ-ity, preventing progression of the cell cycle22,111. By contrast,p15INK4B interacts with and inactivates CDK4 and CDK6, orassociates with cyclin D complexes of CDK4 or CDK6. The latterinteraction not only inactivates the catalytic activity of these

CDKs but also displaces p21CIP1 or the related p27KIP1 fromthese complexes, allowing them to bind to and inactivate theCDK2 complexes with cyclin A and E22,111,113.

Induction of p15INK4B or p21CIP1 expression in response toTGF-β is brought about by Smad-mediated transcriptional acti-vation114,115. In contrast to many TGF-β responses that are medi-ated by Smad3 and Smad4, a heteromeric complex of Smad2,Smad3 and Smad4 induces transcription by interacting with Sp1at the p15INK4B (ref. 114) or the p21CIP1 (ref. 115) promoter.Consequently, the Smad complex recruits the coactivatorCBP/p300 into the complex and strongly potentiates the tran-scriptional activity of Sp1, which activates transcription of thep15INK4B or p21CIP1 genes114,115.

Additional mechanisms also contribute to TGF-β–mediatedgrowth arrest, again depending on the cell type22. For example,TGF-β inhibits the expression of c-Myc (ref. 107), CDK4 (refs.116,117) and CDC25A, a tyrosine phosphatase of CDK4 andCDK6 (refs. 118,119). High levels of c-Myc render the cells resis-tant to the growth inhibitory activity of TGF-β (refs. 120,121),and downregulation of c-Myc is required for the induction ofp15INK4B (ref. 121) and p21CIP1 (ref. 122) expression. The inter-action of c-Myc in a complex at the p15INK4B promoter correlateswith transcriptional repression123,124; TGF-β–induced downreg-ulation of c-Myc thus allows derepression124 and TGF-β–inducedtranscription through Smads114. Decreased expression of c-Mycmay also be involved in the downregulation of CDC25A expres-sion in response to TGF-β (refs. 125,126).

The TGF-β protein has also been shown to induce apoptosis inseveral cell types127–133. The initial events that follow receptor acti-vation and result in cell death are not understood, although theDaxx adaptor protein has been proposed to mediate TGF-β–induced apoptosis through its ability to interact with the TβRIIreceptor134. Increased levels of Smad3 or Smad4 can induce apop-tosis135–137, and dominant-negative interference with Smad3 func-tion protects against apoptosis138. In addition, Smad7 can act as aneffector of TGF-β–induced cell death131,139, but can also protectagainst cell death133,140, depending on the context.

rapid growth

normalcell

benignlesion

migration/invasion

BOB CRIMI

Ras Smad

carcinoma

carcinoma

metastasis

metastasis

TGF-β

loss of TGF-βsignaling, e.g. TGFBR2

mutation

perturbation of TGF-βsignaling

further geneticchanges

Fig. 3 Alternative roles for TGF-β sig-naling in cancer progression. Alter-ations in the TGF-β signaling pathwaycontribute to increased tumor pro-gression, invasion and metastasis bytwo different routes. First, normalepithelial cells are inhibited in growthby TGF-β. Early genetic loss of signal-ing components, such as TβRII, leadsto increased synthesis of DNA, morerapid tumor growth and clonalexpansion, which results in anincreased probability of accumulatingfurther mutations and cytogeneticchanges. This ultimately drives tumorprogression. Second, and more fre-quently, the TGF-β signaling pathwayremains intact but is perturbed byepigenetic mechanisms, such as acti-vation of the Ras pathway, whichleads to a diminished growthinhibitory response but accentuationof activities that enhance tumor pro-gression (see Fig. 2). Synergismbetween an elevated TGF-β signalingpathway and an enhanced Ras path-way leads to a direct increase intumor cell plasticity, invasion andmetastasis.

©20

01 N

atu

re P

ub

lish

ing

Gro

up

h

ttp

://g

enet

ics.

nat

ure

.co

m© 2001 Nature Publishing Group http://genetics.nature.com

Page 5: document

progress

nature genetics • volume 29 • october 2001 121

These findings implicate Smad signaling in TGF-β–mediatedapoptosis, although the pro-apoptotic gene targets of Smad signal-ing remain to be identified. Downregulation of Bcl-XL (refs.141,142) and activation of caspase 3 and 8 (refs. 141,143,144) havealso been implicated in this process, and a mitochondrial septin,ARTS, has a role in the sensitivity of cells to TGF-β–mediated apop-tosis132. The activation of tyrosine kinase–receptor signaling145,through stimulation of the PI-3-K/Akt146–148 or the Raf87 signalingpathway, protects against TGF-β-induced apoptosis and is likely tocontribute to carcinogenesis.

TGF-β signaling as a tumor suppressor pathwayAlterations in TGF-βreceptor signaling. The role of TGF-β signal-ing as a tumor suppressor pathway is best illustrated by the presenceof inactivating mutations in genes encoding TGF-β receptors andSmads in human carcinomas, and by studies of tumor developmentin mouse models. Somatic mutations in TGFBR2, the gene thatencodes the TβRII receptor, occur most frequently in tumors frompatients with hereditary non-polyposis colorectal cancer(HNPCC). A repeat stretch of adenines in the TβRII codingsequence is prone to mutation in these patients, owing to germlinedefects in their capacity for DNA mismatch repair. Resultingnucleotide additions or deletions give rise to a truncated TβRII,which is incapable of signaling149–151.

Mutations in TGFBR2 are frequently found in colon can-cers149–151, gastric cancers150,152–154 and gliomas155 withmicrosatellite instability, but are less common in microsatellite-instability tumors from the endometrium, pancreas, liver andbreast150,156–159. Missense and inactivating mutations in thekinase domain of TβRII have also been reported to occur in coloncancers, which do not display microsatellite instability160, and invarious other carcinomas22,161. Overall, inactivating TGFBR2mutations may be present in 20–25% of all colon cancers. Albeitless common, inactivating mutations in TβRI, encoded byTGFBR1 have also been observed, most notably in ovarian can-cers, metastatic breast cancers, pancreatic carcinomas and T-celllymphomas162–166. Mutations in TGFBR1 have not, however,been found to be accompanied by TGFBR2 mutations166. Alto-gether, these mutations suggest that TβRII and TβRI might func-tion as tumor suppressors in the development of carcinomas.

Various observations suggest that TGF-β receptor expression isoften downregulated or that TGF-β receptor availability at the cellsurface is impaired167–170 in tumor cells, and that these defectsallow cells to escape the growth inhibitory activities of TGF-β.TGF-β receptor expression may also be reduced in tumor cellsthrough altered levels or activities of transcription factors, such asthe Ets transcription factors that are required for expression ofTβRII (ref. 170). Transcriptional silencing may also result fromhypermethylation of CpG islands in the TβRI or TβRII gene pro-moters, or from mutations in the TβRII promoter that interferewith transcription factor binding170. Whereas decreased TβRIIfunction confers resistance against the growth inhibitory effect ofTGF-β, other TGF-β responses may not be similarly affectedbecause they require different thresholds of signaling171–176.

The tumor suppressor role of TβRII has been demonstrated byexpressing wildtype TβRII in colon or breast carcinoma cells,which lack a functional TGFBR2 allele177,178, and by overexpress-ing it in thyroid carcinomas179. Expression of TβRII conferredgrowth inhibition and suppressed anchorage independence, andstrongly reduced tumor formation in nude mice when comparedwith the parental cells. Overexpression of TGF-β1 or TβRII in theskin of transgenic mice also provided evidence for a tumor sup-pressor activity180,181, and expression of dominant-negativeforms of TβRII in the skin or mammary gland182,183 increasedtumor formation. In addition, mice with one inactivated allele of

the gene encoding TGF-β1 (TGFB1) show increased propensityfor developing carcinoma, either spontaneously184 or after car-cinogen exposure185. In the latter case, haploinsufficiency ofTGFB1 increased tumor susceptibility, as the tumors retainedone wildtype allele185. Consistent with these results, decreasedexpression of TβRII correlates with high tumor grade in humancancers170 and experimental systems186–189.

Although these observations suggest that loss of TGF-βresponsiveness provides a distinct advantage for developingtumors, most tumors do not have inactivated TGF-β receptors;individuals with HNPCC, who frequently have TGFBR2 muta-tions in their tumors149, have a better prognosis than individualswith sporadic colon cancer190. Abrogation of TGF-β signaling,while leading to a loss of growth inhibition mediated by TGF-βand early tumor onset, paradoxically protects against tumor pro-gression. TGF-β receptors can thus act as tumor suppressors atearly stages of carcinoma development, but at later stages TGF-βresponsiveness may provide distinct advantages for cancer pro-gression (see below and Fig. 3).

Smads as tumor suppressors. Mutations of the Smad2- andSmad4-encoding gene sequences, but not those of Smad3 or theinhibitory Smad6 or Smad7, have been detected in several carci-nomas22,191, but are uncommon overall. These observations sug-gest that some Smads act as tumor suppressors. Inactivation ofthe genes encoding Smad2 (MADH2) or Smad4 (MADH4)occurs by loss of entire chromosome segments, small deletions,frameshift, nonsense or missense mutations22,191. MADH4mutations occur primarily in pancreatic carcinomas, in whichthe MADH4 gene was first identified as DPC4 (deleted in pancre-atic carcinomas)192, and in colon carcinomas, and less frequentlyin other types of carcinomas22,191.

Whereas biallelic inactivation of MADH4 often occurs inpancreatic and colon carcinomas, haploinsufficiency of theMADH4 locus may also contribute to progression of can-cer193,194. The occurrence of MADH4 mutations in thegermline of a subset of juvenile polyposis families195 furthersupports the notion that Smad4 acts as a tumor suppressor. Incontrast to MADH4, inactivating mutations of MADH2 are rareand occur primarily in colorectal and lung carcinomas196–199.Finally, enhanced Smad7 levels, as observed in pancreatic carci-nomas200 (possibly as a result of TGF-β signaling) may alsodecrease Smad responsiveness.

Tumor-associated mutations in the Smad4 and Smad2 proteinsoccur most frequently in the MH2 domain, which mediates het-eromeric complex formation and transcriptional activation22,191.C-terminal deletions or mutations often inactivate the Smad, butalso provide dominant-negative interference with wildtype Smadfunction201–203. Other mutations confer decreased stabilitythrough increased ubiquitin-mediated degradation204. Mutationsin the MH1 domain of Smad4 interfere with its DNA-bindingability205,206. Most if not all mutations impair Smad function;however, some mutations might alter TGF-β signaling to theadvantage of the tumor. Indeed, the Smad2 mutations D450E andP445H, which are found in colorectal carcinomas, may enhancethe invasive behavior of the tumor cells207, as do some mutationsin the type I TGF-β receptor162.

Studies that use mice carrying an inactivated allele of MADH4support a role for this gene in tumor suppression. Whereashomozygous inactivation of MADH4 leads to early embryoniclethality, heterozygous mice are viable but develop intestinalpolyps that can progress to carcinomas193,208,209. When com-bined with an inactivated allele of the adenomatous polyposiscoli (Apc) gene, simultaneous loss of the wildtype alleles at bothloci results in the development of multiple polyps and in progres-sion to heterogeneous invasive adenocarcinomas208.

©20

01 N

atu

re P

ub

lish

ing

Gro

up

h

ttp

://g

enet

ics.

nat

ure

.co

m© 2001 Nature Publishing Group http://genetics.nature.com

Page 6: document

progress

122 nature genetics • volume 29 • october 2001

Mice with homozygous inactivation of the gene encodingSmad3 (MADH3) have also been shown to develop colon carci-nomas210, although this phenotype was not seen in two similarstudies211,212. This discrepancy could be due to differences ingenetic background or in environmental factors associatedwith animal husbandry. For example, the absence of Smad3expression might impair TGF-β–mediated immunosuppres-sion and contribute to immune or inflammatory responsesthat predispose to cancer formation. Although a role ofMADH3 as tumor suppressor gene is conceivable, no inactivat-ing MADH3 mutations have been observed in human tumors.The occurrence of MADH2, but not MADH3, mutations canbe rationalized by the crucial role of Smad2 in the TGF-β–induced expression of p21CIP1 or p15INK4B CDKinhibitors114,115. Inactivation of Smad2 might therefore inacti-vate the growth arrest response without affecting the Smad3-mediated gene expression that provides an advantage fortumor development, that is, the induction of some extracellu-lar matrix proteins by TGF-β.

The loss of Smad4 function may not abolish TGF-β respon-siveness, even though Smad4 is generally perceived as essentialfor TGF-β responses. Indeed, mouse fibroblasts derived fromMadh4–/– embryos213, as well as some Smad4-deficient tumorcell lines214,215, retain at least some TGF-β responses. This is con-sistent with the observation that TGF-β–induced synthesis offibronectin can occur in the absence of Smad4 (ref. 71). Thus, thehigh frequency of MADH4 deletions in tumors might representa selective perturbation of TGF-β signaling, rather than its com-plete abrogation.

In addition, loss of Smad4 expression may enhance Ras signal-ing and progression to undifferentiated carcinomas216, furtheremphasizing the crosstalk between TGF-β and Ras signaling.Moreover, different TGF-β responses may have differential sensi-tivity to Smad signaling. Accordingly, overexpression of Smad7in a carcinoma cell line suppressed TGF-β–induced growtharrest, without affecting TGF-β–induced expression of plas-minogen activator inhibitor 1, and enhanced anchorage-inde-pendent growth and tumorigenicity200. Finally, inactivatingMADH4 mutations have been found in conjunction with muta-tions in TGFBR2 or TGFBR1 (ref. 160), which strongly suggeststhat Smad4 has tumor suppressive activities that are unrelated toTGF-β signaling.

Other mechanisms of impaired TGF-β responsiveness.Upregulated expression of proto-oncogenes or expression oftheir oncogenic counterparts may also provide a mechanism bywhich tumor cells downregulate TGF-β responsiveness andescape its tumor suppressor effects. For example, elevated expres-sion of the proto-oncogene c-Ski in melanoma217 has been corre-lated with decreased TGF-β responsiveness, presumably owing torepression of Smad-mediated transcription63,64.

Expression of c-Myc may also be involved in the growthinhibitory response to TGF-β. The downregulation of c-Mycexpression by TGF-β, as observed in epithelial cells, is lost invarious cancer cell lines, concomitant with the loss of thegrowth inhibitory response to TGF-β36. The EWS-FLI1 onco-gene, which incorporates a partial coding sequence for the Etsfamily transcription factor Fli-1 and was first identified inEwing sarcoma, represses expression of TβRII and mayaccount for decreased TGF-β responsiveness218. In addition,the Tax protein, which is encoded by HTLV-1 and is thought tocontribute to leukemogenesis in adult T-cell leukemia, is apotent repressor of Smad-mediated transcription91. Clearly,tumor cells have developed several strategies to escape thegrowth inhibitory response of TGF-β, and most carcinomacells are resistant to growth inhibition by TGF-β.

Finally, the mannose-6-phosphate receptor may also have arole in the tumor suppressor activity of TGF-β. This receptorbinds latent TGF-β and is required for activation of TGF-β byplasminogen conversion into plasmin219. Loss of heterozygosityat the receptor locus and inactivating receptor mutations occurwith high incidence in hepatocellular and squamous lung carci-nomas220,221, and are early events during carcinogenesis222. Asthe levels of mannose-6-phosphate receptor and loss of heterozy-gosity of this locus correlate with TGF-β1 activation, it is con-ceivable that part of its tumor suppressor role is linked toactivation of TGF-β1 (ref. 223).

Cell-autonomous stimulation of tumor developmentIn spite of the tumor suppressor activity of TGF-β1, tumor cellsoften show increased production of this growth factor3,4, and con-siderable evidence documents its tumor-promoting role throughits effects on tumor cell invasion and changes in the tumor micro-environment. Tumor metastasis depends on various factors,including the ability of tumor cells to migrate and invade thestroma, and to migrate in and out of blood and lymphatic vessels.The epithelial to mesenchymal differentiation of tumor cells hasan important role in this invasive phenotype224. Accordingly,fibroblastoid or ‘spindle-cell’ tumors of epithelial origin havebeen characterized as highly malignant and invasive88,225–229.

This type of transdifferentiation and invasion of epithelial cellsinto the underlying mesenchyme is not restricted to cancer pro-gression; it also occurs during developmental processes such asplacentation224. During cardiac230,231, palatal232,233 and hair-fol-licle development234, TGF-βs are involved in epithelial to mes-enchymal transdifferentiation, and related family members servethe same function in mesoderm induction235. This morphologi-cal transition is characterized by extensive changes in the expres-sion of cell-adhesion molecules and by a switch from acytoskeleton of mainly cytokeratin intermediate filaments to onecomprising predominantly vimentin88,228,229,236. The ability ofepithelial or carcinoma cells to undergo epithelial to mesenchy-mal transition in culture correlates with cell changes that facili-tate invasion and metastasis in vivo88,176,229.

The TGF-β protein can induce an epithelial to mesenchymaltransition in cultures of normal and transformed breast epithelialcells88,229,236–238, squamous carcinoma cells176,239, ovarianadenosarcoma cells240 and melanoma cells241. The phenotypicchanges have been best characterized in NMuMG cells, in whichTGF-β induces changes in cell shape, downregulates the expres-sion of E-cadherin, ZO-1, vinculin and keratin, and induces theexpression of vimentin and N-cadherin, which has been shownto increase cell motility and scattering105,236,238.

The mechanism of the TGF-β-induced epithelial to mesenchy-mal transition is complex, and Ras/PI-3-K signaling and TGF-β-induced activation of Smad and RhoA signaling seem to havedistinct roles in the process105,216,238,242. Whereas TGF-β inducesthis phenotypic transition in some cell lines, growth factors thatact through tyrosine kinase receptors and activation of Ras sig-naling induce similar changes in other cell systems224,243. In somecases, both signaling cascades need to be enhanced to achieveepithelial to mesenchymal transition88. These findings suggestthat these two pathways synergize in the transition of squamousto spindle carcinoma formation.

This synergy explains why, for example, the invasive growthof Ras- or Raf-transformed epithelial cells in vitro depends onintact autocrine TGF-β signaling87,229. Accordingly, spindle-cellcarcinoma tissue has an increased mutant Hras gene dosagewhen compared with squamous cells originating from the sameskin tumors227. Together, these observations suggest that acti-vation of TGF-β signaling and an involvement of Ras/MAP

©20

01 N

atu

re P

ub

lish

ing

Gro

up

h

ttp

://g

enet

ics.

nat

ure

.co

m© 2001 Nature Publishing Group http://genetics.nature.com

Page 7: document

progress

nature genetics • volume 29 • october 2001 123

kinase signaling may be essential for carcinoma cell invasionand metastasis, whether through epigenetic alterations ofautocrine growth factor signaling or genetic alterations. Thisinterpretation is also consistent with the cooperation of bothpathways in mesoderm induction235, and in epithelial to mes-enchymal differentiation and invasion during hair-follicle for-mation234 and cardiogenesis244.

The cell-autonomous role of TGF-β in the spindle-cell pheno-type and invasive behavior of carcinomas has been well docu-mented for skin carcinomas in mice176,180. Transgenic expressionof activated TGF-β1 in mouse epidermis in vivo increased themalignant conversion to carcinomas and the incidence of spin-dle-cell carcinomas, even though the hyperplastic response to thetumor promoter 12-O-tetradecanoylphorbol 13-acetate and thenumber of papillomas after treatment with 7,12-dimethylbenz-[a]-anthracene were reduced180. In another study, expression of adominant-negative TβRII prevented squamous carcinoma cellsfrom undergoing an epithelial to mesenchymal transition inresponse to TGF-β in vivo176. Similar observations on the role ofTGF-β signaling have been made using a Ras-transformed mam-mary epithelial cell line and a fibroblastoid colon carcinoma cellline229. Expression of a dominant-negative TβRII preventedTGF-β-induced epithelial to mesenchymal changes and revertedthe colon carcinoma cells to an epithelial phenotype. In addition,blocking TβRII function suppressed the in vitro invasion and thehighly metastatic phenotype of this colon carcinoma cell line229.Finally, restoration of TβRII signaling in HNPCC cells with anendogenous TβRII mutation rendered the cells invasive in con-trast to their normally non-invasive phenotype229.

The increased incidence of aggressive skin neoplasia associatedwith the use of cyclosporin (for example, in kidney transplantpatients) may also result from a cell-autonomous effect mediatedby TGF-β. Cyclosporin induces invasive behavior in adenocarci-noma cells in vitro and enhances tumor growth in immunodefi-cient SCID-beige mice, whereas anti-TGF-β antibodies preventthe cyclosporin-induced increase in metastasis245. TGF-β hasalso been shown to be an autocrine effector of breast tumormetastasis246. By using a breast cancer cell line that metastasizesto bone in nude mice, it was shown that dominant-negativeinterference with TβRII function decreases tumor formation andmetastasis, and the resulting destruction of bone246. In thismodel, TGF-β1 induced the expression and secretion of parathy-roid hormone-releasing peptide (PTHrP), resulting in therecruitment of bone-resorbing osteoclasts. In complementaryexperiments, tumor cells expressing a partially activated TβRIaccelerated bone destruction and reduced survival246. This find-ing is likely to be relevant for the clinical correlation that womenwith PTHrP-positive breast cancer are more likely to developmetastatic disease than those with PTHrP-negative tumors246.

In summary, in vitro and in vivo studies link autocrine TGF-βresponsiveness to epithelial to mesenchymal transdifferentiation,the acquisition of an invasive phenotype, and metastatic behav-ior. The TGF-β-dependent, reversible nature of this phenotypemay explain why many metastatic tumors show a well-differenti-ated epithelial phenotype, presumably depending on the localtissue micro-environment and levels of active TGF-β.

Effects of TGF-β on the tumor micro-environmentThe increased expression and activation of TGF-β1 by tumor cellsprofoundly affects the micro-environment. Increased proteaseexpression and plasmin generation by tumor cells11 enhancesactivation of TGF-β and degradation of the extracellular matrix,with a consequent release of stored TGF-β. But a concomitantincrease in production of TGF-β1 stimulates synthesis of extracel-lular matrix proteins and chemoattraction of fibroblasts.

Together, all these changes result in a micro-environment thatpromotes tumor growth and invasion and angiogenesis247.

Tumor angiogenesis is crucial for tumor growth and invasion,as blood vessels deliver nutrients and oxygen to the tumor cellsand allow them to intravasate the blood system, which leads tometastasis. TGF-β1 acts as a potent inducer of angiogenesis inseveral assays248–252, and mouse models defective in TGF-β sig-naling components indicate the importance of TGF-β1 in nor-mal vascular development. Targeted inactivation of the geneencoding TGF-β1 or TβRII results in embryonic lethality owingto defective vasculogenesis and angiogenesis253,254, whereasangiogenesis-defective phenotypes are also seen in mice with nullmutations of the genes encoding TβRI (ref. 255), ALK-1, a TGF-β type I receptor that is expressed in endothelial cells and signalsthrough Smad1 and Smad5 (ref. 256) endoglin, an endotheliallyrestricted TGF-β type III receptor (refs. 257,258) and Smad5(refs. 259,260).

Several models indicate the role of tumor cell–secreted TGF-β1in tumor angiogenesis. Increased expression of TGF-β1 in trans-fected prostate carcinoma or Chinese hamster ovary cellsenhanced tumor angiogenesis in immunodeficient mice261,262,and local administration of neutralizing antibodies to TGF-β1strongly reduced tumor angiogenesis261. Intraperitoneal injectionof TGF-β antibodies reduced angiogenesis and tumorigenicity ofa renal carcinoma cell line that lacks TβRII, and is therefore notresponsive to TGF-β, in natural killer (NK)-, T- and B-cell–defi-cient mice263. This cell line also lacks the von Hippel–Lindau(VHL) tumor suppressor gene, and reintroduction of this genedecreased TGF-β1 synthesis, suggesting that VHL regulates TGF-β1 levels and has tumor suppressor effects through an anti-angio-genic mechanism involving TGF-β1 (ref. 263).

Finally, re-expression of Smad4 in Smad4-deficient pancreascancer cells suppresses tumor development primarily by repressingtumor angiogenesis264. In human breast tumors, high levels ofTGF-β1 messenger RNA are associated with increased microvesseldensity, and both parameters correlate with poor patient progno-sis265. Diagnostic studies on other carcinoma types suggest thathigh tumor burden and circulating plasma levels of TGF-β1 areassociated with enhanced tumor angiogenesis and poor patientprognosis266–269. In one study, TGF-β1 levels were also correlatedwith expression levels of the angiogenic growth factor vascularendothelial growth factor (VEGF)270.

The mechanisms of angiogenesis stimulation by TGF-β1 pre-sumably combine direct and indirect effects. TGF-β inducesexpression of VEGF, which directly acts on endothelial cells tostimulate cell proliferation and migration271. TGF-β also inducescapillary formation of endothelial cells, which are cultured oncollagen matrix249,252. Indirect stimulation of angiogenesis byTGF-β1 may occur through the potent chemoattractant activityof TGF-β for monocytes, which release angiogeniccytokines248,250,272,273.

TGF-β1–induced changes in the microenvironment providefavorable conditions for endothelial cell migration and capillaryformation. Moreover, TGF-β–induced expression of the metallo-proteases MMP-2 and MMP-9, and downregulation of the proteaseinhibitor TIMP in tumor and endothelial cells274–279, are expectedto enhance the migratory and invasive properties of endothelialcells required for angiogenesis280,281. Thus, both the direct effects ofTGF-β and its indirect effects on the tumor micro-environmentstimulate tumor angiogenesis.

Secretion and activation of TGF-β1 by tumor cells alsostimulate tumor development and progression through escapefrom immunosurveillance. TGF-β inhibits the proliferationand functional differentiation of T lymphocytes, lymphokine-activated killer cells, NK cells, neutrophils, macrophages and

©20

01 N

atu

re P

ub

lish

ing

Gro

up

h

ttp

://g

enet

ics.

nat

ure

.co

m© 2001 Nature Publishing Group http://genetics.nature.com

Page 8: document

progress

124 nature genetics • volume 29 • october 2001

B cells273,282,283. Several observations show that TGF-β expres-sion by tumor cells promotes tumorigenicity by locallyrepressing immune functions. For example, the increasedexpression of TGF-β1 by tumor cells enhanced their tumori-genicity and prevented activation of cytotoxic T-lymphocytefunction when compared with parental cells284. This repres-sion was presumably elaborated by the ability of TGF-β toinhibit the expression and function of interleukin 2 (IL-2) andIL-2 receptors285,286. Accordingly, the inhibition of the cyto-toxic T-lymphocyte response by EMT6 mammary tumor cells,which produce high levels of TGF-β1, was overcome byectopic expression of IL-2 in these cells287. Thus, TGF-β–mediated suppression of the cytotoxic T-cell response pro-motes tumor development288.

TGF-β1 expression by tumor cells may also inhibit otherimmune functions. Increased expression of TGF-β1 by tumorcells decreased NK cell activity289 and promoted tumor forma-tion in nude mice that lack T cells289,290. In addition, anti–TGF-βantibodies suppressed tumor formation and metastasis of abreast carcinoma cell line in nude mice, while enhancing NK cellfunction291,292. This suppression was not seen in nude beigemice, which lack both T and NK cells, which implicates TGF-β1–induced suppression of NK cells in cancer progression292,293.

TGF-β–mediated suppression of neutrophil function may alsobe involved in tumor progression. Indeed, Fas ligand–expressingcarcinoma cells underwent neutrophil-mediated rejection, butthis rejection did not occur at a site with high levels of TGF-β orwhen TGF-β1 was injected at the tumor site294. Finally, strongdownregulation of the expression of major histocompatibilitycomplex (MHC) class II antigens by TGF-β295–297 may make thetumor cell surface less immunogenic. This regulation contributesto the local immunosuppression and the escape of TGF-β1-expressing tumor cells from immune surveillance298,299.

Potential avenues toward a cancer therapyConsidering the complex roles of TGF-β signaling in tumor sup-pression and progression, which are context- and stage-depen-dent, any strategy to exploit this knowledge for therapeuticpurposes is fraught with difficulties. For example, stimulatingTGF-β signaling might suppress tumor growth, but might alsopromote tumor invasion and metastasis, particularly of cells thatare no longer sensitive to the growth inhibitory effect of TGF-β.To suppress cancer progression by exogenous administration ofTGF-β, induction of TGF-β synthesis or enhanced TGF-β signal-ing would require that the tumors are sensitive to the growthinhibitory effects of TGF-β; unfortunately, however, resistance tothe growth inhibitory effect of TGF-β is often acquired in earlytumor development, often before diagnosis.

It has been proposed, on the one hand, that the beneficialeffects of tamoxifen in preventing or treating breast cancer maybe due, in part, to its ability to induce expression of TGF-β300.On the other hand, tumor resistance to tamoxifen correlateswith increased expression of TGF-β1, which is possibly areflection of insensitivity to the antiproliferative effects ofTGF-β301,302. These elevated levels of TGF-β1 and the conse-quent suppression of NK cell activity may be responsible forthe failure of tamoxifen therapy. Accordingly, tamoxifen resis-tance of a breast tumor cell line has been reversed through theuse of anti-TGF-β antibodies293,303.

As indicated by this example, inhibiting TGF-β1 expression oractivation may represent a valid approach to treatment of carci-noma. Inhibiting TGF-β production should affect the cell-autonomous response and the effects of TGF-β on itsmicro-environment, whereas interfering with the TGF-β respon-siveness of the tumor cells would primarily inhibit its cell-

autonomous effects. But any approach that inhibits TGF-β sig-naling in tumor cells might induce the regression of advancedcancers while stimulating the growth of otherwise quiescent pre-malignant lesions.

The most sensible approach may therefore be to activate TGF-β in a preventive setting, but to inhibit signaling in advancedmetastatic cancer. Inhibiting the production and/or signaling ofTGF-β through antisense RNA, anti–TGF-β antibodies or solu-ble TGF-β receptors might inhibit cancer progression. For exam-ple, antisense RNA inhibition of TGF-β2 synthesis by breastcancer cells enhanced the cytotoxicity of NK cells293, which isconsistent with the ability of anti–TGF-β therapy to decreasesuppression of the NK cell response289,292. In other studies, anti-sense RNA inhibition of TGF-β1 or TGF-β2 synthesis in breastcancer, mesothelioma cells or glioma cells restored the tumorimmunogenicity and the cytotoxic T-lymphocyte response, andinhibited tumor development304–307. As discussed above, anti-bodies against TGF-β have also been shown to inhibit tumor for-mation through an increase in NK cell activity292 or a decrease inmicrovessel density, without directly affecting cell prolifera-tion261,263 or the innate tumorigenicity of cancer cells245.

In addition, administration of an anti-endoglin immunotoxintargeted the endothelial cells and selectively inhibited tumorangiogenesis and carcinoma development308. Sequestration ofTGF-β through the use of soluble receptor ectodomains may pro-vide another approach to inhibit cancer progression. Indeed,expression of a soluble TβRII (ref. 309) or the soluble proteogly-cans betaglycan310 or decorin311,312, which bind TGF-β, inhibitedtumor formation and/or metastasis and even induced tumorregression, depending on the tumor model. These approaches tofacilitating immune rejection might be enhanced further bycombinatorial therapy with IL-2. For example, the combinationof anti-TGF-β antibodies with IL-2–reduced tumor formation ina highly metastatic melanoma cell line, which was unaffected byeither treatment alone313.

In future studies, it would be desirable to retain the TGF-β–mediated stimulation of apoptosis in tumor cells, but toinhibit the cell-autonomous (invasion and metastatic spread)and non–cell-autonomous (angiogenesis and immunosuppres-sion) activities of this multifaceted molecule. In view of theimportant role of the Ras pathway in these effects, combinatorialapproaches involving inhibitors of the Ras pathway coupled withinhibitors of TGF-β might lead to a marked synergistic inhibitionof tumor growth. Future insight into the TGF-β signaling mecha-nisms that lead to epithelial to mesenchymal transition and inva-sion, or to apoptosis, might lead to the design of more targetedand more effective therapeutic interventions.

AcknowledgmentsIt is impossible to mention every important contribution to ourunderstanding of the mechanisms of TGF-β signaling and its role in cancer,and we apologize to those investigators whose contributions could not beacknowledged. Relevant research in the laboratories of the authors is fundedby NIH grants to R.D.; NIH, March of Dimes and American HeartAssociation grants to R.J.A.; and an NIH grant to A.B. We thank J. Qing forthe design and drawing of Fig. 1, and A. Roberts, B. Lechleider and C.Arteaga for their comments.

Received 8 August; accepted 29 August 2001.

1. Roberts, A.B. & Sporn, M.B. in Peptide Growth Factors and their Receptors —Handbook of Experimental Pharmacology (eds Sporn, M.B. & Roberts, A.B.)(Springer, Heidelberg, 1990). 419–472.

2. Massagué, J. TGF-β signal transduction. Annu. Rev. Biochem. 67, 753–791 (1998).Review

3. Derynck, R. et al. Synthesis of messenger RNAs for transforming growth factors αand β and the epidermal growth factor receptor by human tumors. Cancer Res.47, 707–712 (1987).

©20

01 N

atu

re P

ub

lish

ing

Gro

up

h

ttp

://g

enet

ics.

nat

ure

.co

m© 2001 Nature Publishing Group http://genetics.nature.com

Page 9: document

progress

nature genetics • volume 29 • october 2001 125

4. Dickson, R.B. et al. Activation of growth factor secretion in tumorigenic states ofbreast cancer induced by 17β-estradiol or v-Ha-ras oncogene. Proc. Natl Acad. Sci.USA 84, 837–841 (1987).

5. Miyazono, K., Ichijo, H. & Heldin, C.H. Transforming growth factor-β: latentforms, binding proteins and receptors. Growth Factors 8, 11–22 (1993). Review

6. Munger, J. et al. Latent transforming growth factor-β: structural features andmechanisms of activation. Kidney Int. 51, 1376–1382 (1997). Review

7. Oklu, R. & Hesketh, R. The latent transforming growth factor β binding protein(LTBP) family. Biochem. J. 352, 601–610 (2000). Review

8. Taipale, J., Saharinen, J. & Keski-Oja, J. Extracellular matrix-associatedtransforming growth factor-β: role in cancer cell growth and invasion. Adv.Cancer Res. 75, 87–134 (1998). Review

9. Sato, Y. & Rifkin, D.B. Inhibition of endothelial cell movement by pericytes andsmooth muscle cells: activation of a latent transforming growth factor-β1-likemolecule by plasmin during co-culture. J. Cell Biol. 109, 309–315 (1989).

10. Lyons, R.M., Gentry, L.E., Purchio, A.F. & Moses, H.L. Mechanism of activation oflatent recombinant transforming growth factor β1 by plasmin. J. Cell Biol. 110,1361–1367 (1990).

11. Andreasen, P.A., Kjoller, L., Christensen, L. & Duffy, M.J. The urokinase-typeplasminogen activator system in cancer metastasis: a review. Int. J. Cancer 72,1–22 (1997). Review

12. Yu, Q. & Stamenkovic, I. Cell surface-localized matrix metalloproteinase-9proteolytically activates TGF-β and promotes tumor invasion and angiogenesis.Genes Dev. 14, 163–176 (2000).

13. Stamenkovic, I. Matrix metalloproteinases in tumor invasion and metastasis.Semin. Cancer Biol. 10, 415–433 (2000). Review

14. Stetler-Stevenson, W.G. & Yu, A.E. Proteases in invasion: matrixmetalloproteinases. Semin. Cancer Biol. 11, 143–153 (2001). Review

15. Schultz-Cherry, S. & Murphy-Ullrich, J.E. Thrombospondin causes activation oflatent transforming growth factor-β secreted by endothelial cells by a novelmechanism. J. Cell Biol. 122, 923–932 (1993).

16. Crawford, S.E. et al. Thrombospondin-1 is a major activator of TGF-β1 in vivo. Cell93, 1159–1170 (1998).

17. Munger, J.S. et al. The integrin αvβ6 binds and activates latent TGF β1: amechanism for regulating pulmonary inflammation and fibrosis. Cell 96, 319–328(1999).

18. Derynck, R. & Feng, X. H. TGF-β receptor signaling. Biochim Biophys Acta. 1333,F105–F150 (1997). Review

19. Piek, E., Heldin, C.H. & ten Dijke, P. Specificity, diversity, and regulation in TGF-βsuperfamily signaling. FASEB J. 13, 2105–2124 (1999). Review

20. Itoh, S., Itoh, F., Goumans, M.J. & ten Dijke, P. Signaling of transforming growthfactor-β family members through Smad proteins. Eur. J. Biochem. 267, 6954–6967(2000). Review

21. Massagué, J. How cells read TGF-β signals. Nature Rev. Mol. Cell. Biol. 1, 169–178(2000). Review

22. Massagué, J., Blain, S.W. & Lo, R.S. TGF-β signaling in growth control, cancer, andheritable disorders. Cell 103, 295–309 (2000). Review

23. Miyazono, K., ten Dijke, P. & Heldin, C.H. TGF-β signaling by Smad proteins. Adv.Immunol. 75, 115–157 (2000). Review

24. Wang, T. et al. The immunophilin FKBP12 functions as a common inhibitor of theTGF-β family type I receptors. Cell 86, 435–444 (1996).

25. Chen, Y.G., Liu, F. & Massagué, J. Mechanism of TGF-β receptor inhibition byFKBP12. EMBO J. 16, 3866–3876 (1997).

26. Yao, D., Doré, J.J. Jr & Leof, E.B. FKBP12 is a negative regulator of transforminggrowth factor-β receptor internalization. J. Biol. Chem. 275, 13149–13154 (2000).

27. Aghdasi, B. et al. FKBP12, the 12-kDa FK506-binding protein, is a physiologicregulator of the cell cycle. Proc. Natl Acad. Sci. USA 98, 2425–2430 (2001).

28. Chen, R.H., Miettinen, P.J., Maruoka, E.M., Choy, L. & Derynck, R. A WD-domainprotein that is associated with and phosphorylated by the type II TGF-β receptor.Nature 377, 548–552 (1995).

29. Choy, L. & Derynck, R. The type II transforming growth factor (TGF)-β receptor-interacting protein TRIP-1 acts as a modulator of the TGF-β response. J. Biol.Chem. 273, 31455–31462 (1998).

30. Griswold-Prenner, I., Kamibayashi, C., Maruoka, E.M., Mumby, M.C. & Derynck, R.Physical and functional interactions between type I transforming growth factor βreceptors and Bα, a WD-40 repeat subunit of phosphatase 2A. Mol. Cell. Biol. 18,6595–6604 (1998).

31. Datta, P.K., Chytil, A., Gorska, A.E. & Moses, H.L. Identification of STRAP, a novelWD domain protein in transforming growth factor-β signaling. J. Biol. Chem. 273,34671–34674 (1998).

32. Datta, P.K. & Moses, H.L. STRAP and Smad7 synergize in the inhibition oftransforming growth factor β signaling. Mol. Cell. Biol. 20, 3157–3167 (2000).

33. Petritsch, C., Beug, H., Balmain, A. & Oft, M. TGF-β inhibits p70 S6 kinase viaprotein phosphatase 2A to induce G1 arrest. Genes Dev. 14, 3093–3101 (2000).

34. Derynck, R., Zhang, Y. & Feng, X.-H. Smads: transcriptional activators of TGF-βresponses. Cell 95, 737–740 (1998). Review

35. Massagué, J. & Wotton, D. Transcriptional control by the TGF-β/Smad signalingsystem. EMBO J. 19, 1745–1754 (2000). Review

36. Chen, C.R., Kang, Y. & Massagué, J. Defective repression of c-myc in breast cancercells: a loss at the core of the transforming growth factor β growth arrestprogram. Proc. Natl Acad. Sci. USA 98, 992–999 (2001).

37. Alliston, T., Choy, L., Ducy, P., Karsenty, G. & Derynck, R. TGF-β-induced repressionof CBFA1 by Smad3 decreases cbfa1 and osteocalcin expression and inhibitsosteoblast differentiation. EMBO J. 20, 2254–2272 (2001).

38. Hayes, S. A. et al. Smad3 represses androgen receptor-mediated transcription.Cancer Res. 61, 2112–2118 (2001).

39. Zhang, Y., Feng, X.H. & Derynck, R. Smad3 and Smad4 cooperate with c-Jun/c-Fosto mediate TGF-β-induced transcription. Nature 394, 909–913 (1998).

40. Liberati, N. T. et al. Smads bind directly to the Jun family of AP-1 transcriptionfactors. Proc. Natl Acad. Sci. USA 96, 4844–4849 (1999).

41. Qing, J., Zhang, Y. & Derynck, R. Structural and functional characterization of thetransforming growth factor-β-induced Smad3/c-Jun transcriptional cooperativity.J. Biol. Chem. 275, 38802–38812 (2000).

42. Sano, Y. et al. ATF-2 is a common nuclear target of Smad and TAK1 pathways intransforming growth factor-β signaling. J. Biol. Chem. 274, 8949–8957 (1999).

43. Hanafusa, H. et al. Involvement of the p38 mitogen-activated protein kinasepathway in transforming growth factor-β-induced gene expression. J. Biol. Chem.274, 27161–27167 (1999).

44. Zhang, Y. & Derynck, R. Transcriptional regulation of the transforming growthfactor-β-inducible mouse germ line Igα constant region gene by functionalcooperation of Smad, CREB, and AML family members. J. Biol. Chem. 275,16979–16985 (2000).

45. Hua, X., Liu, X., Ansari, D.O. & Lodish, H.F. Synergistic cooperation of TFE3 andSmad proteins in TGF-β-induced transcription of the plasminogen activatorinhibitor-1 gene. Genes Dev. 12, 3084–3095 (1998).

46. Hua, X., Miller, Z.A., Wu, G., Shi, Y. & Lodish, H.F. Specificity in transforminggrowth factor β-induced transcription of the plasminogen activator inhibitor-1gene: interactions of promoter DNA, transcription factor µE3, and Smad proteins.Proc. Natl Acad. Sci. USA 96, 13130–13135 (1999).

47. Hanai, J. et al. Interaction and functional cooperation of PEBP2/CBF with Smads.Synergistic induction of the immunoglobulin germline Cα promoter. J. Biol.Chem. 274, 31577–31582 (1999).

48. Pardali, E. et al. Smad and AML proteins synergistically confer transforminggrowth factor β1 responsiveness to human germ-line Igα genes. J. Biol. Chem.275, 3552–3560 (2000).

49. Yanagisawa, J. et al. Convergence of transforming growth factor-β and vitamin Dsignaling pathways on SMAD transcriptional coactivators. Science 283, 1317–1321(1999).

50. Song, C.Z., Tian, X. & Gelehrter, T.D. Glucocorticoid receptor inhibits transforminggrowth factor-β signaling by directly targeting the transcriptional activationfunction of Smad3. Proc. Natl Acad. Sci. USA 96, 11776–11781 (1999).

51. Kang, H.Y. et al. From transforming growth factor-β signaling to androgenaction: identification of Smad3 as an androgen receptor coregulator in prostatecancer cells. Proc. Natl Acad. Sci. USA 98, 3018–3023 (2001).

52. Nakashima, K. et al. Synergistic signaling in fetal brain by STAT3–Smad1 complexbridged by p300. Science 284, 479–482 (1999).

53. Janknecht, R., Wells, N.J. & Hunter, T. TGF-β-stimulated cooperation of Smadproteins with the coactivators CBP/p300. Genes Dev. 12, 2114–2119 (1998).

54. Feng, X.H., Zhang, Y., Wu, R.Y. & Derynck, R. The tumor suppressor Smad4/DPC4and transcriptional adaptor CBP/p300 are coactivators for Smad3 in TGF-β-induced transcriptional activation. Genes Dev. 12, 2153–2163 (1998).

55. Shen, X. et al. TGF-β-induced phosphorylation of Smad3 regulates its interactionwith coactivator p300/CREB-binding protein. Mol. Biol. Cell 9, 3309–3319 (1998).

56. Yingling, J.M. et al. Tumor suppressor Smad4 is a transforming growth factor β-inducible DNA binding protein. Mol. Cell. Biol. 17, 7019–7028 (1997).

57. Zawel, L. et al. Human Smad3 and Smad4 are sequence-specific transcriptionactivators. Mol. Cell 1, 611–617 (1998).

58. Dennler, S. et al. Direct binding of Smad3 and Smad4 to critical TGF β-inducibleelements in the promoter of human plasminogen activator inhibitor-type 1 gene.EMBO J. 17, 3091–3100 (1998).

59. Shi, Y. et al. Crystal structure of a Smad MH1 domain bound to DNA: insights onDNA binding in TGF-β signaling. Cell 94, 585–594 (1998).

60. Shioda, T. et al. Transcriptional activating activity of Smad4: roles of SMADhetero-oligomerization and enhancement by an associating transactivator. Proc.Natl Acad. Sci. USA 95, 9785–9790 (1998).

61. Yahata, T. et al. The MSG1 non-DNA-binding transactivator binds to the p300/CBPcoactivators, enhancing their functional link to the Smad transcription factors. J.Biol. Chem. 275, 8825–8834 (2000).

62. Kurokawa, M. et al. The oncoprotein Evi-1 represses TGF-β signalling by inhibitingSmad3. Nature 394, 92–96 (1998).

63. Luo, K. et al. The Ski oncoprotein interacts with the Smad proteins to repress TGFβsignaling. Genes Dev. 13, 2196–2206 (1999).

64. Akiyoshi, S. et al. c-Ski acts as a transcriptional co-repressor in transforminggrowth factor-β signaling through interaction with Smads. J. Biol. Chem. 274,35269–35277 (1999).

65. Stroschein, S.L., Wang, W., Zhou, S., Zhou, Q. & Luo, K. Negative feedbackregulation of TGF-β signaling by the SnoN oncoprotein. Science 286, 771–774(1999).

66. Wotton, D., Lo, R.S., Lee, S. & Massagué, J. A Smad transcriptional corepressor.Cell 97, 29–39 (1999).

67. Kim, R.H. et al. A novel Smad nuclear interacting protein, SNIP1, suppresses p300-dependent TGF-β signal transduction. Genes Dev. 14, 1605–1616 (2000).

68. Verschueren, K. et al. SIP1, a novel zinc finger/homeodomain repressor, interactswith Smad proteins and binds to 5′-CACCT sequences in candidate target genes. J.Biol. Chem. 274, 20489–20498 (1999).

69. Zhang, Y. & Derynck, R. Regulation of Smad signalling by protein associations andsignalling crosstalk. Trends Cell Biol. 9, 274–279 (1999). Review

70. ten Dijke, P., Miyazono, K. & Heldin, C.H. Signaling inputs converge on nucleareffectors in TGF-β signaling. Trends Biochem. Sci. 25, 64–70 (2000). Review

71. Hocevar, B.A., Brown, T.L. & Howe, P.H. TGF-β induces fibronectin synthesisthrough a c-Jun N-terminal kinase-dependent, Smad4-independent pathway.EMBO J. 18, 1345–1356 (1999).

72. Wong, C. et al. Smad3-Smad4 and AP-1 complexes synergize in transcriptionalactivation of the c-Jun promoter by transforming growth factor β. Mol. Cell. Biol.19, 1821–1830 (1999).

73. Yue, J. & Mulder, K. M. Requirement of Ras/MAPK pathway activation bytransforming growth factor β for transforming growth factor β1 production in aSmad-dependent pathway. J. Biol. Chem. 275, 30765–30773 (2000).

74. Engel, M.E., McDonnell, M.A., Law, B.K. & Moses, H.L. Interdependent SMAD andJNK signaling in transforming growth factor-β-mediated transcription. J. Biol.Chem. 274, 37413–37420 (1999).

75. Chang, E. & Goldberg, H. Requirements for transforming growth factor-βregulation of the pro-α 2(I) collagen and plasminogen activator inhibitor-1promoters. J. Biol. Chem. 270, 4473–4477 (1995).

76. Chung, K.Y., Agarwal, A., Uitto, J. & Mauviel, A. An AP-1 binding sequence isessential for regulation of the human α2(I) collagen (COL1A2) promoter activityby transforming growth factor-β. J. Biol. Chem. 271, 3272–3278 (1996).

77. Peron, P. et al. Potentiation of Smad transactivation by Jun proteins during acombined treatment with epidermal growth factor and transforming growthfactor-β in rat hepatocytes. Role of phosphatidylinositol 3-kinase-induced AP-1activation. J. Biol. Chem. 276, 10524–10531 (2001).

78. Owen, R.D. & Ostrowski, M.C. Transcriptional activation of a conserved sequenceelement by Ras requires a nuclear factor distinct from c-Fos or c-Jun. Proc. NatlAcad. Sci. USA 87, 3866–3870 (1990).

79. Geiser, A.G, Kim, S.J., Roberts, A.B. & Sporn, M.B. Characterization of the mousetransforming growth factor-β1 promoter and activation by the Ha-ras oncogene.

©20

01 N

atu

re P

ub

lish

ing

Gro

up

h

ttp

://g

enet

ics.

nat

ure

.co

m© 2001 Nature Publishing Group http://genetics.nature.com

Page 10: document

progress

126 nature genetics • volume 29 • october 2001

Mol. Cell. Biol. 11, 84–92 (1991).80. Van Obberghen-Schilling, E., Roche, N.S., Flanders, K.C., Sporn, M.B. & Roberts,

A.B. Transforming growth factor β1 positively regulates its own expression innormal and transformed cells. J. Biol. Chem. 263, 7741–7746 (1988).

81. Glick, A.B., Sporn, M.B. & Yuspa, S.H. Altered regulation of TGF-β1 and TGF-α inprimary keratinocytes and papillomas expressing v-Ha-ras. Mol. Carcinog. 4,210–219 (1991).

82. Gum, R. et al. Stimulation of 92-kDa gelatinase B promoter activity by ras ismitogen-activated protein kinase kinase 1-independent and requires multipletranscription factor binding sites including closely spaced PEA3/ets and AP-1sequences. J. Biol. Chem. 271, 10672–10680 (1996).

83. Himelstein, B.P., Lee, E.J., Sato, H., Seiki, M. & Muschel, R.J. Transcriptionalactivation of the matrix metalloproteinase-9 gene in an H-ras and v-myctransformed rat embryo cell line. Oncogene 14, 1995–1998 (1997).

84. Reddy, K.B., Krueger, J.S., Kondapaka, S.B. & Diglio, C.A. Mitogen-activatedprotein kinase (MAPK) regulates the expression of progelatinase B (MMP-9) inbreast epithelial cells. Int. J. Cancer 82, 268–273 (1999).

85. Kretzschmar, M., Doody, J., Timokhina, I. & Massagué, J. A mechanism ofrepression of TGFβ/Smad signaling by oncogenic Ras. Genes Dev. 13, 804–816(1999).

86. Calonge, M.J. & Massagué, J. Smad4/DPC4 silencing and hyperactive Ras jointlydisrupt transforming growth factor-β antiproliferative responses in colon cancercells. J. Biol. Chem. 274, 33637–33643 (1999).

87. Lehmann, K. et al. Raf induces TGFβ production while blocking its apoptotic butnot invasive responses: a mechanism leading to increased malignancy in epithelialcells. Genes Dev. 14, 2610–2622 (2000).

88. Oft, M. et al. TGF-β1 and Ha-Ras collaborate in modulating the phenotypicplasticity and invasiveness of epithelial tumor cells. Genes Dev. 10, 2462–2477(1996).

89. Labbé, E., Letamendia, A. & Attisano, L. Association of Smads with lymphoidenhancer binding factor 1/T cell-specific factor mediates cooperative signaling bythe transforming growth factor-β and Wnt pathways. Proc. Natl Acad. Sci. USA97, 8358–8363 (2000).

90. Nishita, M. et al. Interaction between Wnt and TGF-β signalling pathways duringformation of Spemann’s organizer. Nature 403, 781–785 (2000).

91. Mori, N. et al. Human T-cell leukemia virus type I oncoprotein Tax represses Smad-dependent transforming growth factor β signaling through interaction withCREB-binding protein/p300. Blood 97, 2137–2144 (2001).

92. Lee, D.K. et al. The hepatitis B virus encoded oncoprotein pX amplifies TGF-βfamily signaling through direct interaction with Smad4: potential mechanism ofhepatitis B virus-induced liver fibrosis. Genes Dev. 15, 455–466 (2001).

93. Kaji, H., Canaff, L., Lebrun, J.J., Goltzman, D. & Hendy, G.N. Inactivation of menin,a Smad3-interacting protein, blocks transforming growth factor type β signaling.Proc. Natl Acad. Sci. USA 98, 3837–3842 (2001).

94. Afrakhte, M. Induction of inhibitory Smad6 and Smad7 mRNA by TGF-β familymembers. Biochem. Biophys. Res. Commun. 249, 505–511 (1998).

95. Ulloa, L., Doody, J. & Massagué, J. Inhibition of transforming growth factor-β/SMAD signalling by the interferon-γ/STAT pathway. Nature 397, 710–713 (1999).

96. Bitzer, M. et al. A mechanism of suppression of TGF-β/SMAD signaling by NF-κB/RelA. Genes Dev. 14, 187–197 (2000).

97. Mulder, K.M. & Morris, S.L. Activation of p21Ras by transforming growth factor βin epithelial cells. J. Biol. Chem. 267, 5029–5031 (1992).

98. Hartsough, M.T. & Mulder, K.M. Transforming growth factor β activation ofp44MAPK in proliferating cultures of epithelial cells. J. Biol. Chem. 270,7117–7124 (1995).

99. Mulder, K.M. Role of Ras and MAPKs in TGFβ signaling. Cytokine Growth FactorRev. 11, 23–35 (2000). Review

100.Yamaguchi, K. et al. Identification of a member of the MAPKKK family as apotential mediator of TGF-β signal transduction. Science 270, 2008–2011 (1995).

101.Shirakabe, K. et al. TAK1 mediates the ceramide signaling to stress-activatedprotein kinase/c-Jun N-terminal kinase. J. Biol. Chem. 272, 8141–8144 (1997).

102.Shibuya, H. et al. Role of TAK1 and TAB1 in BMP signaling in early Xenopusdevelopment. EMBO J. 17, 1019–1028 (1998).

103.Ninomiya-Tsuji, J. et al. The kinase TAK1 can activate the NIK-IκB as well as theMAP kinase cascade in the IL-1 signalling pathway. Nature 398, 252–256 (1999).

104.Takaesu, G. et al. TAB2, a novel adaptor protein, mediates activation of TAK1MAPKKK by linking TAK1 to TRAF6 in the IL-1 signal transduction pathway. Mol.Cell 5, 649–658 (2000).

105.Bhowmick, N.A. et al. Transforming growth factor-β1 mediates epithelial tomesenchymal transdifferentiation through a RhoA-dependent mechanism. Mol.Biol. Cell 12, 27–36 (2001).

106.Engel, M.E., Datta, P.K. & Moses, H.L. RhoB is stabilized by transforming growthfactor β and antagonizes transcriptional activation. J. Biol. Chem. 273, 9921–9926(1998).

107.Coffey, R.J. Jr et al. Selective inhibition of growth-related gene expression inmurine keratinocytes by transforming growth factor β. Mol. Cell. Biol. 8,3088–3093 (1988).

108.Laiho, M., DeCaprio, J.A., Ludlow, J.W., Livingston, D.M. & Massagué, J. Growthinhibition by TGF-β linked to suppression of retinoblastoma proteinphosphorylation. Cell 62, 175–185 (1990).

109.Moses, H.L., Yang, E.Y. & Pietenpol, J.A. TGF-β stimulation and inhibition of cellproliferation: new mechanistic insights. Cell 63, 245–247 (1990).

110.Hannon, G.J. & Beach, D. p15INK4B is a potential effector of TGF-β-induced cellcycle arrest. Nature 371, 257–261 (1994).

111.Reynisdóttir, I., Polyak, K., Iavarone, A. & Massagué, J. Kip/CIP and INK4 CDKinhibitors cooperate to induce cell cycle arrest in response to TGF-β. Genes Dev. 9,1831–1845 (1995).

112.Datto, M.B. et al. Transforming growth factor β induces the cyclin-dependentkinase inhibitor p21 through a p53-independent mechanism. Proc. Natl Acad. Sci.USA 92, 5545–5549 (1995).

113.Sandhu, C. et al. Transforming growth factor β stabilizes p15INK4B protein,increases p15INK4B–CDK4 complexes, and inhibits cyclin D1–CDK4 association inhuman mammary epithelial cells. Mol. Cell. Biol. 17, 2458–2467 (1997).

114.Feng, X.H., Lin, X. & Derynck, R. Smad2, Smad3 and Smad4 cooperate with Sp1 toinduce p15INK4B transcription in response to TGF-β. EMBO J. 19, 5178–5193 (2000).

115.Pardali, K. et al. Role of Smad proteins and transcription factor Sp1 in p21Waf1/CIP1

regulation by transforming growth factor-β. J. Biol. Chem. 275, 29244–29256 (2000).116.Ewen, M.E., Sluss, H.K., Whitehouse, L.L. & Livingston, D.M. TGF-β inhibition of

Cdk4 synthesis is linked to cell cycle arrest. Cell 74, 1009–1020 (1993).117.Ewen, M.E., Oliver, C.J., Sluss, H.K., Miller, S.J. & Peeper, D.S. p53-dependent

repression of CDK4 translation in TGF-β-induced G1 cell-cycle arrest. Genes Dev. 9,204–217 (1995).

118.Iavarone, A. & Massagué, J. Repression of the CDK activator Cdc25A and cell-cyclearrest by cytokine TGF-β in cells lacking the CDK inhibitor p15. Nature 387,417–422 (1997).

119.Iavarone, A. & Massagué, J. E2F and histone deacetylase mediate transforminggrowth factor β repression of cdc25A during keratinocyte cell cycle arrest. Mol.Cell. Biol. 19, 916–922 (1999).

120.Alexandrow, M.G., Kawabata, M., Aakre, M. & Moses, H.L. Overexpression of thec-Myc oncoprotein blocks the growth-inhibitory response but is required for themitogenic effects of transforming growth factor β1. Proc. Natl Acad. Sci. USA 92,3239–3243 (1995).

121.Warner, B.J., Blain, S.W., Seoane, J. & Massagué, J. Myc downregulation bytransforming growth factor β required for activation of the p15INK4B G1 arrestpathway. Mol. Cell. Biol. 19, 5913–5922 (1999).

122.Claassen, G.F. & Hann, S.R. A role for transcriptional repression of p21CIP1 by c-Mycin overcoming transforming growth factor β -induced cell-cycle arrest. Proc. NatlAcad. Sci. USA 97, 9498–9503 (2000).

123.Staller, P. et al. Repression of p15INK4b expression by Myc through association withMiz-1. Nature Cell Biol. 3, 392–399 (2001).

124.Seoane, J. et al. TGFβ influences Myc, Miz-1 and Smad to control the CDKinhibitor p15INK4b. Nature Cell Biol. 3, 400–408 (2001).

125.Galaktionov, K., Chen, X. & Beach, D. Cdc25 cell-cycle phosphatase as a target ofc-myc. Nature 382, 511–517 (1996).

126.Santoni-Rugiu, E., Falck, J., Mailand, N., Bartek, J. & Lukas, J. Involvement of Mycactivity in a G1/S-promoting mechanism parallel to the pRb/E2F pathway. Mol.Cell. Biol. 20, 3497–3509 (2000).

127.Rotello, R.J., Lieberman, R.C., Purchio, A.F. & Gerschenson, L.E. Coordinatedregulation of apoptosis and cell proliferation by transforming growth factor β1 incultured uterine epithelial cells. Proc. Natl Acad. Sci. USA 88, 3412–3415 (1991).

128.Oberhammer, F.A. et al. Induction of apoptosis in cultured hepatocytes and inregressing liver by transforming growth factor β1. Proc. Natl Acad. Sci. USA 89,5408–5412 (1992).

129.Selvakumaran, M. et al. The novel primary response gene MyD118 and the proto-oncogenes myb, myc, and bcl-2 modulate transforming growth factor β1-inducedapoptosis of myeloid leukemia cells. Mol. Cell. Biol. 14, 2352–2360 (1994).

130.Chaouchi, N. et al. Characterization of transforming growth factor-β1 inducedapoptosis in normal human B cells and lymphoma B cell lines. Oncogene 11,1615–1622 (1995).

131.Landstrom, M. et al. Smad7 mediates apoptosis induced by transforming growthfactor β in prostatic carcinoma cells. Curr. Biol. 10, 535–538 (2000).

132.Larisch, S. et al. A novel mitochondrial septin-like protein, ARTS, mediatesapoptosis dependent on its P-loop motif. Nature Cell Biol. 2, 915–921 (2000).

133.Patil, S. et al. Smad7 is induced by CD40 and protects WEHI 231 B-lymphocytesfrom transforming growth factor-β-induced growth inhibition and apoptosis. J.Biol. Chem. 275, 38363–38370 (2000).

134.Perlman, R., Schiemann, W.P., Brooks, M.W., Lodish, H.F. & Weinberg, R.A. TGF-β-induced apoptosis is mediated by the adapter protein Daxx that facilitates JNKactivation. Nature Cell Biol. 3, 708–714 (2001).

135.Atfi, A., Buisine, M., Mazars, A. & Gespach, C. Induction of apoptosis by DPC4, atranscriptional factor regulated by transforming growth factor-β through stress-activated protein kinase/c-Jun N-terminal kinase (SAPK/JNK) signaling pathway. J.Biol. Chem. 272, 24731–24734 (1997).

136.Yanagisawa, K. et al. Induction of apoptosis by Smad3 and down-regulation ofSmad3 expression in response to TGF-β in human normal lung epithelial cells.Oncogene 17, 1743–1747 (1998).

137.Dai, J.L., Bansal, R.K. & Kern, S.E. G1 cell cycle arrest and apoptosis induction bynuclear Smad4/Dpc4: phenotypes reversed by a tumorigenic mutation. Proc. NatlAcad. Sci. USA 96, 1427–1432 (1999).

138.Yamamura, Y., Hua, X., Bergelson, S. & Lodish, H.F. Critical role of Smads and AP-1complex in transforming growth factor-β-dependent apoptosis. J. Biol. Chem.275, 36295–36302 (2000).

139.Lallemand, F. et al. Smad7 inhibits the survival nuclear factor κB and potentiatesapoptosis in epithelial cells. Oncogene 20, 879–884 (2001).

140.Ishisaki, A. et al. Smad7 is an activin-inducible inhibitor of activin-induced growtharrest and apoptosis in mouse B cells. J. Biol. Chem. 273, 24293–24296 (1998).

141.Saltzman, A. et al. Transforming growth factor-β-mediated apoptosis in theRamos B-lymphoma cell line is accompanied by caspase activation and Bcl-XLdownregulation. Exp. Cell. Res. 242, 244–254 (1998).

142.Chipuk, J.E., Bhat, M., Hsing, A.Y., Ma, J. & Danielpour, D. Bcl-xL blocks TGF-β1-induced apoptosis by inhibiting cytochrome C release and not by directlyantagonizing Apaf-1-dependent caspase activation in prostate epithelial cells. J.Biol. Chem. 276, 26614–26621 (2001).

143.Shima, Y. et al. Activation of caspase-8 in transforming growth factor-β-inducedapoptosis of human hepatoma cells. Hepatology 30, 1215–1222 (1999).

144.Chen, R.H. & Chang, T.Y. Involvement of caspase family proteases in transforminggrowth factor-β-induced apoptosis. Cell Growth Differ. 8, 821–827 (1997).

145.Tanaka, S. & Wands, J.R. Insulin receptor substrate 1 overexpression in humanhepatocellular carcinoma cells prevents transforming growth factor β1-inducedapoptosis. Cancer Res. 56, 3391–3394 (1996).

146.Chen, R.H., Su, Y.H., Chuang, R.L. & Chang, T.Y. Suppression of transforminggrowth factor-β-induced apoptosis through a phosphatidylinositol 3-kinase/Akt-dependent pathway. Oncogene 17, 1959–1968 (1998).

147.Chen, R.H., Chang, M.C., Su, Y.H., Tsai, Y.T. & Kuo, M.L. Interleukin-6 inhibitstransforming growth factor-β-induced apoptosis through thephosphatidylinositol 3-kinase/Akt and signal transducers and activators oftranscription 3 pathways. J. Biol. Chem. 274, 23013–23019 (1999).

148.Shih, W.L., Kuo, M.L, Chuang, S.E., Cheng, A.L. & Doong, S.L. Hepatitis B virus Xprotein inhibits transforming growth factor-β-induced apoptosis through theactivation of phosphatidylinositol 3-kinase pathway. J. Biol Chem. 275,25858–25864 (2000).

149.Markowitz, S. et al. Inactivation of the type II TGF-β receptor in colon cancer cellswith microsatellite instability. Science 268, 1336–1338 (1995).

150.Myeroff, L.L. et al. A transforming growth factor β receptor type II gene mutationcommon in colon and gastric but rare in endometrial cancers with microsatelliteinstability. Cancer Res. 55, 5545–5547 (1995).

©20

01 N

atu

re P

ub

lish

ing

Gro

up

h

ttp

://g

enet

ics.

nat

ure

.co

m© 2001 Nature Publishing Group http://genetics.nature.com

Page 11: document

progress

nature genetics • volume 29 • october 2001 127

151.Lu, S.L., Zhang, W.C., Akiyama, Y., Nomizu, T. & Yuasa, Y. Genomic structure of thetransforming growth factor β type II receptor gene and its mutations inhereditary nonpolyposis colorectal cancers. Cancer Res. 56, 4595–4598 (1996).

152.Park, K. et al. Genetic changes in the transforming growth factor β (TGF-β) type IIreceptor gene in human gastric cancer cells: correlation with sensitivity to growthinhibition by TGF-β. Proc. Natl Acad. Sci. USA 91, 8772–8776 (1994).

153.Chung, Y.J. et al. Microsatellite instability-associated mutations associatepreferentially with the intestinal type of primary gastric carcinomas in a high-riskpopulation. Cancer Res. 56, 4662–4665 (1996).

154.Ohue, M. et al. Mutations of the transforming growth factor β type II receptorgene and microsatellite instability in gastric cancer. Int. J. Cancer 68, 203–206(1996).

155.Izumoto, S. et al. Microsatellite instability and mutated type II transforminggrowth factor-β receptor gene in gliomas. Cancer Lett. 112, 251–256 (1997).

156.Vincent, F. et al. Mutation analysis of the transforming growth factor β type IIreceptor in sporadic human cancers of the pancreas, liver, and breast. Biochem.Biophys. Res. Commun. 223, 561–564 (1996).

157.Kawate, S. et al. Mutation analysis of transforming growth factor β type IIreceptor, Smad2, and Smad4 in hepatocellular carcinoma. Int. J. Oncol. 14,127–131 (1999).

158.Furuta, K. et al. Gene mutation of transforming growth factor β1 type II receptorin hepatocellular carcinoma. Int. J. Cancer 81, 851–853 (1999).

159.Tomita, S. et al. Analyses of microsatellite instability and the transforming growthfactor-β receptor type II gene mutation in sporadic breast cancer and theircorrelation with clinicopathological features. Breast Cancer Res. Treat. 53, 33–39(1999).

160.Grady, W.M. et al. Mutational inactivation of transforming growth factor βreceptor type II in microsatellite stable colon cancers. Cancer Res. 59, 320–324(1999).

161.Reiss, M. TGF-β and cancer. Microbes Infect. 1, 1327–1347 (1999). Review162.Chen, T., Carter, D., Garrigue-Antar, L. & Reiss, M. Transforming growth factor β

type I receptor kinase mutant associated with metastatic breast cancer. CancerRes. 58, 4805–4810 (1998).

163.Chen, T. et al. Transforming growth factor-β receptor type I gene is frequentlymutated in ovarian carcinomas. Cancer Res. 61, 4679–4682 (2001).

164.Goggins, M. et al. Genetic alterations of the transforming growth factor βreceptor genes in pancreatic and biliary adenocarcinomas. Cancer Res. 58,5329–5332 (1998).

165.Schiemann, W.P., Pfeifer, W.M., Levi, E., Kadin, M.E. & Lodish, H.F. A deletion inthe gene for transforming growth factor β type I receptor abolishes growthregulation by transforming growth factor β in a cutaneous T-cell lymphoma.Blood 94, 2854–2861 (1999).

166.Wang, D. et al. Analysis of specific gene mutations in the transforming growthfactor-β signal transduction pathway in human ovarian cancer. Cancer Res. 60,4507–4512 (2000).

167.Knaus, P.I. et al. A dominant inhibitory mutant of the type II transforming growthfactor β receptor in the malignant progression of a cutaneous T-cell lymphoma.Mol. Cell. Biol. 16, 3480–3489 (1996).

168.Guo, Y., Jacobs, S.C. & Kyprianou, N. Down-regulation of protein and mRNAexpression for transforming growth factor-β (TGF-β1) type I and type II receptorsin human prostate cancer. Int. J. Cancer 71, 573–579 (1997).

169.Amoroso, S.R., Huang, N., Roberts, A.B., Potter, M. & Letterio, J.J. Consistent lossof functional transforming growth factor β receptor expression in murineplasmacytomas. Proc. Natl Acad. Sci. USA 95, 189–194 (1998).

170.Kim, S.J., Im, Y.H., Markowitz, S.D. & Bang, Y.J. Molecular mechanisms ofinactivation of TGF-β receptors during carcinogenesis. Cytokine Growth FactorRev. 11, 159–168 (2000). Review

171.Geiser, A.G., Burmester, J.K., Webbink, R., Roberts, A.B. & Sporn, M.B. Inhibitionof growth by transforming growth factor-β following fusion of twononresponsive human carcinoma cell lines. Implication of the type II receptor ingrowth inhibitory responses. J. Biol. Chem. 267, 2588–2593 (1992).

172.Chen, R.H., Ebner, R. & Derynck, R. Inactivation of the type II receptor reveals tworeceptor pathways for the diverse TGF-β activities. Science 260, 1335–1338 (1993).

173.Fafeur, V., O’Hara, B. & Böhlen, P. A glycosylation-deficient endothelial cellmutant with modified responses to transforming growth factor-β and othergrowth inhibitory cytokines: evidence for multiple growth inhibitory signaltransduction pathways. Mol. Biol. Cell 4, 135–144 (1993).

174.Feng, X.H., Filvaroff, E.H. & Derynck, R. Transforming growth factor-β (TGF-β)-induced down-regulation of cyclin A expression requires a functional TGF-βreceptor complex. Characterization of chimeric and truncated type I and type IIreceptors. J. Biol. Chem. 270, 24237–24245 (1995).

175.Wieser, R., Attisano, L., Wrana, J.L. & Massagué, J. Signaling activity oftransforming growth factor β type II receptors lacking specific domains in thecytoplasmic region. Mol. Cell. Biol. 13, 7239–7247 (1993).

176.Portella, G. et al. Transforming growth factor β is essential for spindle cellconversion of mouse skin carcinoma in vivo: implications for tumor invasion. CellGrowth Differ. 9, 393–404 (1998).

177.Sun, L. et al. Expression of transforming growth factor β type II receptor leads toreduced malignancy in human breast cancer MCF-7 cells. J. Biol. Chem. 269,26449–26455 (1998).

178.Wang, J. et al. Demonstration that mutation of the type II transforming growthfactor β receptor inactivates its tumor suppressor activity in replication error-positive colon carcinoma cells. J. Biol. Chem. 270, 22044–22049 (1995).

179.Turco, A. et al. Overexpression of transforming growth factor β-type II receptorreduces tumorigenicity and metastastic potential of K-ras-transformed thyroidcells. Int. J. Cancer 80, 85–91 (1999).

180.Cui, W. et al. TGFβ1 inhibits the formation of benign skin tumours but enhancesprogression to invasive spindle cell carcinomas in transgenic mice. Cell 86,531–542 (1996).

181.Wang, X.J., Liefer, K.M., Tsai, S., O’Malley, B.W. & Roop, D.R. Development ofgene-switch transgenic mice that inducibly express transforming growth factorβ1 in the epidermis. Proc. Natl Acad. Sci. USA 96, 8483–8488 (1999).

182.Böttinger, E.P., Jakubczak, J.L., Haines, D.C., Bagnall, K. & Wakefield, L.M.Transgenic mice overexpressing a dominant-negative mutant type II transforminggrowth factor β receptor show enhanced tumorigenesis in the mammary glandand lung in response to the carcinogen 7,12-dimethylbenz-[a]-anthracene.Cancer Res. 57, 5564–5570 (1997).

183.Go, C., Li, P. & Wang, X.J. Blocking transforming growth factor β signaling in

transgenic epidermis accelerates chemical carcinogenesis: a mechanismassociated with increased angiogenesis. Cancer Res. 59, 2861–2868 (1999).

184.Engle, S.J. et al. Transforming growth factor β1 suppresses nonmetastatic coloncancer at an early stage of tumorigenesis. Cancer Res. 59, 3379–3386 (1999).

185.Tang, B. et al. Transforming growth factor-β1 is a new form of tumor suppressorwith true haploid insufficiency. Nature Med. 4, 802–807 (1998).

186.Tang, B. et al. Loss of responsiveness to transforming growth factor β inducesmalignant transformation of nontumorigenic rat prostate epithelial cells. CancerRes. 59, 4834–4842 (1999).

187.Venkatasubbarao, K. et al. Differential expression of transforming growth factorβ receptors in human pancreatic adenocarcinoma. Anticancer Res. 20, 43–51(2000).

188.Kim, T.K. et al. Alteration of cell growth and morphology by overexpression oftransforming growth factor β type II receptor in human lung adenocarcinomacells. Lung Cancer 31, 181–191 (2001).

189.Lee, B.I. et al. MS-275, a histone deacetylase inhibitor, selectively inducestransforming growth factor β type II receptor expression in human breast cancercells. Cancer Res. 61, 931–934 (2001).

190.Bubb, V.J. et al. Microsatellite instability and the role of hMSH2 in sporadiccolorectal cancer. Oncogene 12, 2641–2649 (1996).

191.Hata, A., Shi, Y. & Massagué, J. TGF-β signaling and cancer: structural andfunctional consequences of mutations in Smads. Mol. Med. Today 4, 257–262(1998). Review

192.Hahn, S.A. et al. DPC4, a candidate tumor suppressor gene at humanchromosome 18q21.1. Science 271, 350–353 (1996).

193.Xu, X. et al. Haploid loss of the tumor suppressor Smad4/Dpc4 initiates gastricpolyposis and cancer in mice. Oncogene 19, 1868–1874 (2000).

194.Luttges, J. et al. Allelic loss is often the first hit in the biallelic inactivation of thep53 and DPC4 genes during pancreatic carcinogenesis. Am. J. Pathol. 158,1677–1683 (2001).

195.Howe, J.R. et al. Mutations in the SMAD4/DPC4 gene in juvenile polyposis. Science280, 1086–1088 (1998).

196.Eppert, K. et al. MADR2 maps to 18q21 and encodes a TGFβ-regulated MAD-related protein that is functionally mutated in colorectal carcinoma. Cell 86,543–552 (1996).

197.Takagi, Y. et al. Somatic alterations of the SMAD-2 gene in human colorectalcancers. Br. J. Cancer 78, 1152–1155 (1998).

198.Yakicier, M.C. et al. Smad2 and Smad4 gene mutations in hepatocellularcarcinoma. Oncogene 18, 4879–4883 (1999).

199.Ohtaki, N. et al. Somatic alterations of the DPC4 and Madr2 genes in colorectalcancers and relationship to metastasis. Int. J. Oncol. 18, 265–270 (2001).

200.Kleeff, J. et al. The TGF-β signaling inhibitor Smad7 enhances tumorigenicity inpancreatic cancer. Oncogene 18, 5363–5372 (1999).

201.Zhang, Y. Musci, T. & Derynck, R. The tumor suppressor Smad4/DPC4 as a centralmediator of Smad function. Curr. Biol. 7, 270–276 (1997).

202.Goto, D. et al. A single missense mutant of Smad3 inhibits activation of bothSmad2 and Smad3, and has a dominant negative effect on TGF-β signals. FEBSLett. 430, 201–204 (1998).

203.Prunier, C., Ferrand, N., Frottier, B., Pessah, M. & Atfi, A. Mechanism formutational inactivation of the tumor suppressor Smad2. Mol. Cell. Biol. 21,3302–3313 (2001).

204.Xu, J. & Attisano, L. Mutations in the tumor suppressors Smad2 and Smad4inactivate transforming growth factor β signaling by targeting Smads to theubiquitin-proteasome pathway. Proc. Natl Acad. Sci. USA 97, 4820–4825 (2000).

205.Dai, J.L., Turnacioglu, K.K., Schutte, M., Sugar, A.Y. & Kern, S.E. Dpc4transcriptional activation and dysfunction in cancer cells. Cancer Res. 58,4592–4597 (1998).

206.Morén, A., Itoh, S., Moustakas, A., Dijke, P. & Heldin, C.H. Functionalconsequences of tumorigenic missense mutations in the amino-terminal domainof Smad4. Oncogene 19, 4396–4404 (2000).

207.Prunier, C. et al. Evidence that Smad2 is a tumor suppressor implicated in thecontrol of cellular invasion. J. Biol. Chem. 274, 22919–22922 (1999).

208.Takaku, K. et al. Intestinal tumorigenesis in compound mutant mice of both Dpc4(Smad4) and Apc genes. Cell 92, 645–656 (1998).

209.Takaku, K. et al. Gastric and duodenal polyps in Smad4 (Dpc4) knockout mice.Cancer Res. 9, 6113–6117 (1999).

210.Zhu, Y., Richardson, J.A., Parada, L.F. & Graff, J.M. Smad3 mutant mice developmetastatic colorectal cancer. Cell 94, 703–714 (1998).

211.Datto, M.B. et al. Targeted disruption of Smad3 reveals an essential role intransforming growth factor β-mediated signal transduction. Mol. Cell. Biol. 19,2495–2504 (1999).

212.Yang, X. et al. Targeted disruption of SMAD3 results in impaired mucosalimmunity and diminished T cell responsiveness to TGF-β. EMBO J. 18, 1280–1291(1999).

213.Sirard, C. et al. Targeted disruption in murine cells reveals variable requirementfor Smad4 in transforming growth factor β-related signaling. J. Biol. Chem. 275,2063–2070 (2000).

214.Dai, J.L. et al. Transforming growth factor-β responsiveness in DPC4/SMAD4-nullcancer cells. Mol. Carcinog. 26, 37–43 (1999).

215.Fink, S.P. et al. Transforming growth factor-β-induced growth inhibition in aSmad4 mutant colon adenoma cell line. Cancer Res. 61, 256–260 (2001).

216.Iglesias, M., Frontelo, P., Gamallo, C. & Quintanilla, M. Blockade of Smad4 intransformed keratinocytes containing a Ras oncogene leads to hyperactivation ofthe Ras-dependent Erk signalling pathway associated with progression toundifferentiated carcinomas. Oncogene 19, 4134–4145 (2000).

217.Fumagalli, S., Doneda, L., Nomura, N. & Larizza, L. Expression of the c-ski proto-oncogene in human melanoma cell lines. Melanoma Res. 3, 23–27 (1993).

218.Hahm, K.B. et al. Repression of the gene encoding the TGF-β type II receptor is amajor target of the EWS-FLI1 oncoprotein. Nature Genet. 23, 222–227 (1999).

219.Dennis, P.A. & Rifkin, D.B. Cellular activation of latent transforming growth factorβ requires binding to the cation-independent mannose 6-phosphate/insulin-likegrowth factor type II receptor. Proc. Natl Acad. Sci. USA 88, 580–584 (1991).

220.Kong, F.M., Anscher, M.S., Washington, M.K., Killian, J.K. & Jirtle, R.L. M6P/IGF2Ris mutated in squamous cell carcinoma of the lung. Oncogene 19, 1572–1578(2000).

221.De Souza, A.T., Hankins, G.R., Washington, M.K., Orton, T.C. & Jirtle, R.L.M6P/IGF2R gene is mutated in human hepatocellular carcinomas with loss ofheterozygosity. Nature Genet. 11, 447–449 (1995).

©20

01 N

atu

re P

ub

lish

ing

Gro

up

h

ttp

://g

enet

ics.

nat

ure

.co

m© 2001 Nature Publishing Group http://genetics.nature.com

Page 12: document

progress

128 nature genetics • volume 29 • october 2001

222.Yamada, T., De Souza, A.T., Finkelstein, S. & Jirtle, R.L. Loss of the gene encodingmannose 6-phosphate/insulin-like growth factor II receptor is an early event inliver carcinogenesis. Proc. Natl Acad. Sci. USA 94, 10351–10355 (1997).

223.Jirtle, R.L., Hankins, G.R., Reisenbichler, H. & Boyer, I.J. Regulation of mannose 6-phosphate/insulin-like growth factor-II receptors and transforming growth factorβ during liver tumor promotion with phenobarbital. Carcinogenesis 15,1473–1478 (1994).

224.Thiery, J.P. & Chopin, D. Epithelial cell plasticity in development and tumorprogression. Cancer Metastasis Rev. 18, 31–42 (1999). Review

225.Battifora, H. Spindle cell carcinoma: ultrastructural evidence of squamous originand collagen production by the tumor cells. Cancer 37, 2275–2282 (1976).

226.Evans, H.L. and Baer, S.C. Epithelioid sarcoma: a clinicopathologic and prognosticstudy of 26 cases. Semin. Diagn. Pathol. 10, 286–291 (1993).

227.Buchmann, A., Ruggeri, B., Klein-Szanto, A.J.P. & Balmain, A. Progression ofsquamous carcinoma cells to spindle carcinoma of the mouse skin is associatedwith an imbalance of H-ras alleles on chromosome 7. Cancer Res. 51, 4097–4101(1991).

228.Stoler, A., Stenback, F. & Balmain, A. The conversion of mouse skin squamous cellcarcinomas to spindle cell carcinomas is a recessive event. J. Cell Biol. 122,1103–1117 (1993).

229.Oft, M., Heider, K.H. & Beug, H. TGFβ signaling is necessary for carcinoma cellinvasiveness and metastasis. Curr. Biol. 8, 1243–1252 (1998).

230.Potts, J.D. & Runyan, R.B. Epithelial-mesenchymal cell transformation in theembryonic heart can be mediated, in part, by transforming growth factor β. Dev.Biol. 134, 392–401 (1989).

231.Brown, C.B., Boyer, A.S., Runyan, R.B. & Barnett, J.V. Requirement of type III TGF-βreceptor for endocardial cell transformation in the heart. Science 283, 2080–2082(1999).

232.Proetzel, G. et al. Transforming growth factor-β3 is required for secondary palatefusion. Nature Genet. 11, 409–414 (1995).

233.Kaartinen, V. et al. Abnormal lung development and cleft palate in mice lackingTGF-β3 indicates defects of epithelial–mesenchymal interaction. Nature Genet.11, 415–421 (1995).

234.Weinberg, W.C., Brown, P.D., Stetler-Stevenson, W.G. & Yuspa, S.H. Growthfactors specifically alter hair follicle cell proliferation and collagenolytic activityalone or in combination. Differentiation 45, 168–178 (1990).

235.Kimelman, D. & Griffin, K.J. Vertebrate mesendoderm induction and patterning.Curr. Opin. Genet. Dev. 10, 350–356 (2000). Review

236.Miettinen, P.J., Ebner, R., Lopez, A.R. & Derynck, R. TGF-β inducedtransdifferentiation of mammary epithelial cells to mesenchymal cells:involvement of type I receptors. J. Cell Biol. 127, 2021–2036 (1994).

237.Hosobuchi, M. & Stampfer, M. R. Effects of transforming growth factor β ongrowth of human mammary epithelial cells in culture. In vitro Cell Dev. Biol. 25,705–713 (1989).

238.Piek, E., Moustakas, A., Kurisaki, A., Heldin, C.H. & ten Dijke, P. TGF-β type Ireceptor/ALK-5 and Smad proteins mediate epithelial to mesenchymaltransdifferentiation in NMuMG breast epithelial cells. J. Cell Sci. 112, 4557–4568(1999).

239.Caulin, C., Scholl, S.G., Frontelo, P., Gamallo, C. & Quintanilla, M. Chronicexposure of cultured transfected mouse epidermal cells to TGFβ1 induces anepithelial-mesenchymal transdifferentiation and a spindle tumoral phenotype.Cell Growth Diff. 6, 1027–1035 (1995).

240.Kitagawa, K. et al. Epithelial–mesenchymal transformation of a newly establishedcell line from ovarian adenosarcoma by transforming growth factor-β1. Int. J.Cancer 66, 91–97 (1996).

241.Janji, B., Melchior, C., Gouon, V., Vallar, L. & Kieffer, N. Autocrine TGF-β-regulatedexpression of adhesion receptors and integrin-linked kinase in HT-144 melanomacells correlates with their metastatic phenotype. Int. J. Cancer 83, 255–262 (1999).

242.Santibanez, J.F., Frontelo, P., Iglesias, M., Martinez, J. & Quintanilla, M. Urokinaseexpression and binding activity associated with the transforming growth factorβ1-induced migratory and invasive phenotype of mouse epidermal keratinocytes.J. Cell. Biochem. 74, 61–73 (1999).

243.Jeffers, M., Rong, S. & Vande Woude, G.F. Enhanced tumorigenicity and invasion-metastasis by hepatocyte growth factor/scatter factor-met signalling in humancells concomitant with induction of the urokinase proteolysis network. Mol. CellBiol. 16, 1115–1125 (1996).

244.Lakkis, M.M. & Epstein, J.A. Neurofibromin modulation of ras activity is requiredfor normal endocardial-mesenchymal transformation in the developing heart.Development 125, 4359–4367 (1998).

245.Hojo, M. et al. Cyclosporine induces cancer progression by a cell-autonomousmechanism. Nature 397, 530–534 (1999).

246.Yin, J.J. et al. TGF-β signaling blockade inhibits PTHrP secretion by breast cancercells and bone metastases development. J. Clin. Invest. 103, 197–206 (1999).

247.Hanahan, D. & Weinberg, R.A. The hallmarks of cancer. Cell 100, 57–70 (2000).248.Roberts, A.B. et al. Transforming growth factor type β: rapid induction of fibrosis

and angiogenesis in vivo and stimulation of collagen formation in vitro. Proc. NatlAcad. Sci. USA 83, 4167–4171 (1986).

249.Madri, J.A., Pratt, B.M. & Tucker, A.M. Phenotypic modulation of endothelial cellsby transforming growth factor-β depends upon the composition andorganization of the extracellular matrix. J. Cell Biol. 106, 1375–1384 (1988).

250.Yang, E.Y. & Moses, H.L. Transforming growth factor β1-induced changes in cellmigration, proliferation, and angiogenesis in the chicken chorioallantoicmembrane. J. Cell Biol. 111, 731–741 (1990).

251.Gajdusek, C.M., Luo, Z. & Mayberg, M.R. Basic fibroblast growth factor andtransforming growth factor β1: synergistic mediators of angiogenesis in vitro. J.Cell. Physiol. 157, 133–144 (1993).

252.Choi, M.E. & Ballermann, B.J. Inhibition of capillary morphogenesis andassociated apoptosis by dominant negative mutant transforming growth factor-βreceptors. J. Biol. Chem. 270, 21144–21150 (1995).

253.Dickson, R.B. et al. Activation of growth factor secretion in tumorigenic states ofbreast cancer induced by 17β-estradiol or v-Ha-ras oncogene. Proc. Natl Acad. Sci.USA 84, 837–841 (1987).

254.Oshima, M., Oshima, H. & Taketo, M.M. TGF-β receptor type II deficiency results indefects of yolk sac hematopoiesis and vasculogenesis. Dev. Biol. 179, 297–302(1996).

255.Larsson, J. et al. Abnormal angiogenesis but intact hematopoietic potential inTGF-β type I receptor-deficient mice. EMBO J. 20, 1663–1673 (2001).

256.Oh, S.P. et al. Activin receptor-like kinase 1 modulates transforming growth

factor-β1 signaling in the regulation of angiogenesis. Proc. Natl Acad. Sci. USA 97,2626–2631 (2000).

257.Li, D.Y. Defective angiogenesis in mice lacking endoglin. Science 284, 1534–1537(1999).

258.Arthur, H.M. et al. Endoglin, an ancillary TGFβ receptor, is required forextraembryonic angiogenesis and plays a key role in heart development. Dev.Biol. 217, 42–53 (1999).

259.Chang, H. et al. Smad5 knockout mice die at mid-gestation due to multipleembryonic and extraembryonic defects. Development 126, 1631–1642 (1999).

260.Yang, X. et al. Angiogenesis defects and mesenchymal apoptosis in mice lackingSmad5. Development 126, 1571–1580 (1999).

261.Ueki, N. et al. Excessive production of transforming growth-factor β1 can play animportant role in the development of tumorigenesis by its action forangiogenesis: validity of neutralizing antibodies to block tumor growth. Biochim.Biophys. Acta 1137, 189–196 (1992).

262.Stearns, M.E., Garcia, F.U., Fudge, K., Rhim, J. & Wang, M. Role of interleukin 10and transforming growth factor β1 in the angiogenesis and metastasis of humanprostate primary tumor lines from orthotopic implants in severe combinedimmunodeficiency mice. Clin. Cancer Res. 5, 711–720 (1999).

263.Ananth, S. et al. Transforming growth factor β1 is a target for the von Hippel-Lindau tumor suppressor and a critical growth factor for clear cell renalcarcinoma. Cancer Res. 59, 2210–2216 (1999).

264.Schwarte-Waldhoff, I. et al. Smad4/DPC4-mediated tumor suppression throughsuppression of angiogenesis. Proc. Natl Acad. Sci. USA 97, 9624–9629 (2000).

265.de Jong, J.S., van Diest, P.J., van der Valk, P. & Baak, J.P. Expression of growthfactors, growth-inhibiting factors, and their receptors in invasive breast cancer. II.Correlations with proliferation and angiogenesis. J. Pathol. 184, 53–57 (1998).

266.Ito, N. et al. Positive correlation of plasma transforming growth factor-β1 levelswith tumor vascularity in hepatocellular carcinoma. Cancer Lett. 89, 45–48 (1995).

267.Ivanovic, V., Melman, A., Davis-Joseph, B., Valcic, M. & Geliebter, J. Elevatedplasma levels of TGF-β1 in patients with invasive prostate cancer. Nature Med. 1,282–284 (1995).

268.Junker, U. et al. Transforming growth factor β 1 is significantly elevated in plasmaof patients suffering from renal cell carcinoma. Cytokine 8, 794–798 (1996).

269.Wikstrom, P., Stattin, P., Franck-Lissbrant, I., Damber, J.E. & Bergh, A.Transforming growth factor β1 is associated with angiogenesis, metastasis, andpoor clinical outcome in prostate cancer. Prostate 37, 19–29 (1998).

270.Saito, H. et al. The expression of transforming growth factor-β1 is significantlycorrelated with the expression of vascular endothelial growth factor and poorprognosis of patients with advanced gastric carcinoma. Cancer 86, 1455–1462(1999).

271.Pertovaara, L. et al. Vascular endothelial growth factor is induced in response totransforming growth factor-β in fibroblastic and epithelial cells. J. Biol. Chem.269, 6271–6274 (1994).

272.Sunderkotter, C., Goebeler, M., Schulze-Osthoff, K., Bhardwaj, R. & Sorg, C.Macrophage-derived angiogenesis factors. Pharmacol Ther. 51, 195–216 (1991).Review

273.Ashcroft, G.S. Bidirectional regulation of macrophage function by TGF-β.Microbes Infect. 1, 1275–1282 (1999). Review

274.Edwards, D.R. et al. Transforming growth factor β modulates the expression ofcollagenase and metalloproteinase inhibitor. EMBO J. 6, 1899–1904 (1987).

275.Kordula, T. et al. Synthesis of tissue inhibitor of metalloproteinase-1 (TIMP-1) inhuman hepatoma cells (HepG2). Up-regulation by interleukin-6 and transforminggrowth factor β1. FEBS Lett. 313, 143–147 (1992).

276.Shimizu, S. et al. Involvement of transforming growth factor β1 in autocrineenhancement of gelatinase B secretion by murine metastatic colon carcinomacells. Cancer Res. 56, 3366–3370 (1996).

277.Sehgal, I. & Thompson, T.C. Novel regulation of type IV collagenase (matrixmetalloproteinase-9 and -2) activities bytransforming growth factor-β1 in humanprostate cancer cell lines. Mol. Biol. Cell 10, 407–416 (1999).

278.Duivenvoorden, W.C., Hirte, H.W. & Singh, G. Transforming growth factor β1 actsas an inducer of matrix metalloproteinase expression and activity in human bone-metastasizing cancer cells. Clin. Exp. Metastasis 17, 27–34 (1999).

279.Hagedorn, H.G., Bachmeier, B.E. & Nerlich, A.G. Synthesis and degradation ofbasement membranes and extracellular matrix and their regulation by TGF-β ininvasive carcinomas. Int. J. Oncol. 18, 669–681 (2001). Review

280.Vu, T.H. et al. MMP-9/gelatinase B is a key regulator of growth plate angiogenesisand apoptosis of hypertrophic chondrocytes. Cell 93, 411–422 (1998).

281.Hiraoka, N., Allen, E., Apel, I.J., Gyetko, M.R. & Weiss, S.J. Matrixmetalloproteinases regulate neovascularization by acting as pericellularfibrinolysins. Cell 95, 365–377 (1998).

282.Letterio, J.J. & Roberts, A.B. Regulation of immune responses by TGF-β. Annu.Rev. Immunol. 16, 137–161 (1998). Review

283.Fortunel, N.O., Hatzfeld, A. & Hatzfeld, J.A. Transforming growth factor-β:pleiotropic role in the regulation of hematopoiesis. Blood 96, 2022–2036 (2000).Review

284.Torre-Amione, G. et al. A highly immunogenic tumor transfected with a murinetransforming growth factor type β1 cDNA escapes immune surveillance. Proc.Natl Acad. Sci. USA 87, 1486–1490 (1990).

285.Kehrl, J.H. et al. Production of transforming growth factor β by human Tlymphocytes and its potential role in the regulation of T cell growth. J. Exp. Med.163, 1037–1050 (1986).

286.Kehrl, J.H. et al. Transforming growth factor β is an importantimmunomodulatory protein for human B lymphocytes. J. Immunol. 137,3855–3860 (1986).

287.McAdam, A.J. et al. Transfection of transforming growth factor-β producingtumor EMT6 with interleukin-2 elicits tumor rejection and tumor reactivecytotoxic T-lymphocytes. J. Immunother. Emphasis Tumor Immunol. 15, 155–164(1994).

288.Inge, T.H., Hoover, S.K., Susskind, B.M., Barrett, S.K. & Bear, H.D. Inhibition oftumor-specific cytotoxic T-lymphocyte responses by transforming growth factorβ1. Cancer Res. 52, 1386–1392 (1992).

289.Horwitz, D.A., Gray, J.D., Ohtsuka, K., Hirokawa, M. & Takahashi, T. Theimmunoregulatory effects of NK cells: the role of TGF-β and implications forautoimmunity. Immunol Today. 18, 538–542 (1997). Review

290.Wallick, S.C., Figari, I.S., Morris, R.E., Levinson, A.D. & Palladino, M.A.Immunoregulatory role of transforming growth factor β (TGF-β) in developmentof killer cells: comparison of active and latent TGF-β1. J. Exp. Med. 172,

©20

01 N

atu

re P

ub

lish

ing

Gro

up

h

ttp

://g

enet

ics.

nat

ure

.co

m© 2001 Nature Publishing Group http://genetics.nature.com

Page 13: document

progress

nature genetics • volume 29 • october 2001 129

1777–17784 (1990).291.Arteaga, C.L., Carty-Dugger, T., Moses, H.L., Hurd, S.D. & Pietenpol, J.A.

Transforming growth factor β1 can induce estrogen-independent tumorigenicity ofhuman breast cancer cells in athymic mice. Cell Growth Differ. 4, 193–201 (1993).

292.Arteaga, C.L. et al. Anti-transforming growth factor (TGF)-β antibodies inhibitbreast cancer cell tumorigenicity and increase mouse spleen natural killer cellactivity. Implications for a possible role of tumor cell/host TGF-β interactions inhuman breast cancer progression. J. Clin. Invest. 92, 2569–2576 (1993).

293.Arteaga, C.L., Koli, K.M., Dugger, T.C. & Clarke, R. Reversal of tamoxifenresistance of human breast carcinomas in vivo by neutralizing antibodies totransforming growth factor-β. J. Natl Cancer Inst. 91, 46–53 (1999).

294.Chen, J.J., Sun, Y. & Nabel, G.J. Regulation of the proinflamatory effects of Fasligand (CD95L). Science 282, 1714–1717 (1998).

295.Czarniecki, C.W., Chiu, H.H., Wong, G.H., McCabe, S.M. & Palladino, M.A.Transforming growth factor-β1 modulates the expression of class IIhistocompatibility antigens on human cells. J. Immunol. 140, 4217–4223 (1988).

296.Geiser, A.G. et al. Transforming growth factor β1 (TGF-β1) controls expression ofmajor histocompatibility genes in the postnatal mouse: aberranthistocompatibility antigen expression in the pathogenesis of the TGF-β1 nullmouse phenotype. Proc. Natl Acad. Sci. USA 90, 9944–9948 (1993).

297.Letterio, J.J. et al. Autoimmunity associated with TGF-β-deficiency in mice isdependent on MHC class II antigen expression. J. Clin. Invest. 98, 2109–2119 (1996).

298.Ma, D. & Niederkorn, J.Y. Transforming growth factor-β down-regulates majorhistocompatibility complex class I antigen expression and increases thesusceptibility of uveal melanoma cells to natural killer cell-mediated cytolysis.Immunology 86, 263–269 (1995).

299.Botti, C., Seregni, E., Ferrari, L., Martinetti, A. & Bombardieri, E.Immunosuppressive factors: role in cancer development and progression. Int. J.Biol. Markers 13, 51–69 (1998). Review

300.Knabbe, C. et al. Evidence that transforming growth factor-β is a hormonallyregulated negative growth factor in human breast cancer cells. Cell 48, 417–428(1987).

301.Thompson, A.M., Kerr, D.J. & Steel, C.M. Transforming growth factor β1 isimplicated in the failure of tamoxifen therapy in human breast cancer. Br. J.Cancer 63, 609–614 (1991).

302.Butta, A. et al. Induction of transforming growth factor β1 in human breastcancer in vivo following tamoxifen treatment. Cancer Res. 52, 4261–4264 (1992).

303.Koli, K.M. et al. Blockade of transforming growth factor-β signaling does notabrogate antiestrogen-induced growth inhibition of human breast carcinomacells. J. Biol. Chem. 272, 8296–8302 (1997).

304.Fakhrai, H. et al. Eradication of established intracranial rat gliomas bytransforming growth factor β antisense gene therapy. Proc. Natl Acad. Sci. USA93, 2909–2914 (1996).

305.Marzo, A.L., Fitzpatrick, D.R., Robinson, B.W. & Scott, B. Antisenseoligonucleotides specific for transforming growth factor β2 inhibit the growth ofmalignant mesothelioma both in vitro and in vivo. Cancer Res. 57, 3200–3207(1997).

306.Park, J.A. et al. Expression of an antisense transforming growth factor-β1transgene reduces tumorigenicity of EMT6 mammary tumor cells. Cancer GeneTher. 4, 42–50 (1997).

307.Yamanaka, R. et al. Suppression of TGF-β1 in human gliomas by retroviral genetransfection enhances susceptibility to LAK cells. J. Neurooncol. 43, 27–34 (1999).

308.Seon, B.K., Matsuno, F., Haruta, Y., Kondo, M. & Barcos, M. Long-lasting completeinhibition of human solid tumors in SCID mice by targeting endothelial cells oftumor vasculature with antihuman endoglin immunotoxin. Clin. Cancer Res. 3,1031–1044 (1997).

309.Won, J. et al. Tumorigenicity of mouse thymoma is suppressed by soluble type IItransforming growth factor β receptor therapy. Cancer Res. 59, 1273–1277 (1999).

310.Bandyopadhyay, A. et al. A soluble transforming growth factor β type III receptorsuppresses tumorigenicity and metastasis of human breast cancer MDA-MB-231cells. Cancer Res. 59, 5041–5046 (1999).

311.Yamaguchi, Y., Mann, D.M. & Ruoslahti, E. Negative regulation of transforminggrowth factor-β by the proteoglycan decorin. Nature 346, 281–284 (1990).

312.Stander, M. et al. Decorin gene transfer-mediated suppression of TGF-β synthesisabrogates experimental malignant glioma growth in vivo. Gene Ther. 5,1187–1194 (1998).

313.Wojtowicz-Praga, S. et al. Modulation of B16 melanoma growth and metastasisby anti-transforming growth factor β antibody and interleukin-2. J. Immunother.Emphasis Tumor Immunol. 19, 169–175 (1996).

©20

01 N

atu

re P

ub

lish

ing

Gro

up

h

ttp

://g

enet

ics.

nat

ure

.co

m© 2001 Nature Publishing Group http://genetics.nature.com