<<

Downloaded from http://cshperspectives.cshlp.org/ on September 24, 2021 - Published by Cold Spring Harbor Laboratory Press

TGF-b and the TGF-b Family: Context-Dependent Roles in Cell and Tissue Physiology

Masato Morikawa,1 Rik Derynck,2 and Kohei Miyazono3

1Ludwig Cancer Research, Science for Life Laboratory, Uppsala University, Biomedical Center, SE-751 24 Uppsala, Sweden 2Department of Cell and Tissue Biology, University of California at San Francisco, San Francisco, California 94143 3Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan Correspondence: [email protected]

The transforming growth factor-b (TGF-b) is the prototype of the TGF-b family of growth and differentiation factors, which is encoded by 33 in mammals and comprises homo- and heterodimers. This review introduces the reader to the TGF-b family with its complexity of names and biological activities. It also introduces TGF-b as the best-studied factor among the TGF-b family , with its diversity of roles in the control of cell proliferation and differentiation, wound healing and immune system, and its key roles in pathology, for exam- ple, skeletal diseases, fibrosis, and cancer.

lthough initially thought to stimulate cell TGF-b has been well documented in most cell Aproliferation, just like many growth factors, types, and has been best characterized in epithe- it became rapidly accepted that transforming lial cells. The bifunctional and context-depen- growth factor b (TGF-b) is a bifunctional reg- dent nature of TGF-b activities was further con- ulator that either inhibits or stimulates cell pro- firmed in a large variety of cell systems and liferation. TGF-b was originally isolated as a biological responses. For example, TGF-b can that, together with epidermal growth inhibit EGF-dependent proliferation of cells factor (EGF), induces cellular transformation in monolayer culture, whereas TGF-b and and anchorage-independent growth of selected EGF synergistically enhance anchorage-inde- fibroblast cell lines (Roberts et al. 1981), yet did pendent growth of the same cells in soft agar not require the presence of EGF to induce phe- medium (Roberts et al. 1985). Now, it is widely notypic transformation of other fibroblast cell accepted that TGF-b regulates a variety of lines (Shipley et al. 1984). In contrast, TGF-b key events in normal development and physiol- was also identified as a growth inhibitor se- ogy, and perturbation of TGF-b signaling creted from confluent BSC-1 cells, epithelial has been implicated in the pathogenesis of dis- cells of African green monkey kidney (Tucker eases such as connective tissue disorders, fibro- et al. 1984). The growth inhibitory activity of sis, and cancer.

Editors: Rik Derynck and Kohei Miyazono Additional Perspectives on The Biology of the TGF-b Family available at www.cshperspectives.org Copyright # 2016 Cold Spring Harbor Laboratory Press; all rights reserved; doi: 10.1101/cshperspect.a021873 Cite this article as Cold Spring Harb Perspect Biol 2016;8:a021873

1 Downloaded from http://cshperspectives.cshlp.org/ on September 24, 2021 - Published by Cold Spring Harbor Laboratory Press

M. Morikawa et al.

The identification of TGF-b family mem- 1995; Sekelsky et al. 1995). In C. elegans, daf-1 bers and their signaling components has en- and daf-4 turned out to also encode serine/thre- abled the characterization of the complex biol- onine transmembrane kinase receptors for ogy of the TGF-b family members. Molecular TGF-b family members. Screening for mutants cloning of TGF-b family members and their with similar phenotypes with daf-4 revealed signaling mediators started in 1985 with the re- three genes, sma-2, sma-3, and sma-4, that ported characterization of complementary DNA were structurally similar to Mad of Drosophila (cDNA) coding for human TGF-b1 (Derynck (Savage et al. 1996). In frog, mouse, and human, et al. 1985). Subsequently, various approaches, genes structurally similar to Mad and sma were based on biochemical purification, developmen- subsequently identified, and the designation tal genetics, and/or targeted cDNA cloning, led “Smad” (Sma and Mad) was adopted. to the identification of polypeptides structurally binding to specific tetrameric type II/type I re- similar to TGF-b1, which together comprise ceptor complexes stabilizes and activates their the members of the TGF-b family. Now signaling capacities, and the receptors then that the human and mouse genome sequence transduce the signals by phosphorylating car- projects are completed, it is apparent that mam- boxy-terminal serine residues of receptor-regu- malian genomes encode 33 TGF-b-related poly- lated (R-) Smads. In most cell types, TGF-bs and peptides. Table 1 shows the 33 known human activins induce phosphorylation of Smad2 TGF-b family polypeptides, which include three and Smad3 (activin/TGF-b-specific R-Smads), TGF-b isoforms, activins, , morpho- and BMPs induce phosphorylation of Smad1, genetic proteins (BMPs), and growth and dif- Smad5, and Smad8 (BMP-specific R-Smads). ferentiation factors (GDFs). Although mostly The activated R-Smads form hetero-oligomeric studied as homodimers, various heterodimeric complexes with a common-partner (co-) Smad, combinations of these have also been identified that is, Smad4 in vertebrate cells (Lagna et al. and characterized as biologically active proteins. 1996; Zhang et al. 1996; Kawabata et al. 1998). In contrast to the large number of TGF-b The complexes translocate into the nucleus ligands, fewer receptors and downstream intra- where they regulate the expression of target cellular effectors, termed Smad proteins, medi- genes, such as those encoding inhibitory (I-) ate transduction of intracellular signaling. In Smads, namely, Smad6 and Smad7 in verte- 1991, a cDNA clone for an , cur- brates, which can inhibit R-Smad activation by rently known as ActRII, was isolated (Mathews the receptors. Finally, TGF-b family proteins and Vale 1991). The kinase domain of this re- were also shown to induce PI3K-Akt signaling ceptor closely resembled the Caenorhabditis and to activate the common mitogen-associated elegans daf-1, a transmembrane serine/threo- (MAP) kinase pathways that are activat- nine-specific protein kinase, which at that time ed by receptor tyrosine kinases, albeit, generally, was seen as an orphan receptor, and provided the to a lower extent. Now that essential players in first indication that TGF-b family members sig- the signaling pathways have been identified, one nal through transmembrane serine/threonine of the major questions to be addressed in this kinases. In the following years, seven type I re- field is to reveal the precise molecular mecha- ceptors and five type II receptors were identified nisms that define the context-dependent dual in mammals and were shown to form hetero- roles of TGF-b family members. meric type I/type II receptorcomplexes.Genetic In this review, we will introduce the TGF-b analysis in Drosophila and C. elegans led to a family members, which in mammals are encod- breakthrough in how signals are transduced ed by 33 genes. Wewill cluster them into several from the receptors to the nucleus. In Drosophila, subgroups based on the structural or sequence mothers against dpp (Mad) was identified as a similarities of the encoded polypeptides. We encoding a component that acts epistati- further focus on the three TGF-b isoforms, cally downstream from (Dpp; TGF-b1, -b2, and -b3, as the best-studied fac- Drosophila BMP-2/-4 ligand) (Raftery et al. tors among the TGF-b family proteins. In ad-

2 Cite this article as Cold Spring Harb Perspect Biol 2016;8:a021873 Downloaded from http://cshperspectives.cshlp.org/ on September 24, 2021 - Published by Cold Spring Harbor Laboratory Press

Roles of TGF-b in Cell and Tissue Physiology

Table 1. Names and genes for the TGF-b family proteins Official gene Protein name symbol (human) Protein name synonyms TGF-b1 TGFB1 CIF-A (cartilage-inducing factor-A), differentiation-inhibiting factor TGF-b2 TGFB2 G-TsF (glioblastoma-derived, T-cell suppressor factor), BSC-1 GI (BSC-1 cell-growth inhibitor), polyergin, CIF-B (cartilage- inducing factor-B) TGF-b3 TGFB3 Inhibin a INHA Inhibin A and Ba a,b Inhibin bA INHBA Inhibin A and activin A or AB, FRP (follicle-stimulating hormone-releasing protein), EDF (erythroid differentiation factor), XTC-MIF (Xenopus XTC cell -inducing factor) a,b Inhibin bB INHBB Inhibin B and activin B or AB, XTC-MIF c Inhibin bC INHBC Activin C c Inhibin bE INHBE Activin E Nodal NODAL BMP-16 (bone morphogenetic protein-16) MSTN GDF-8 (growth and differentiation factor-8) BMP-2 BMP2 BMP-3 BMP3 Osteogenin BMP-4 BMP4 BMP-2B BMP-5 BMP5 BMP-6 BMP6 Vgr1 (Vg1-related protein) BMP-7 BMP7 OP-1 (osteogenic protein-1) BMP-8A BMP8A OP-2 BMP-8B BMP8B OP-3 BMP-9 GDF2 GDF-2 BMP-10 BMP10 GDF-1 GDF1 GDF-3 GDF3 Vgr2 GDF-5 GDF5 CDMP-1 (cartilage-derived morphogenetic protein-1), BMP-14 GDF-6 GDF6 CDMP-2, BMP-13 GDF-7 GDF7 CDMP-3, BMP-12 GDF-9 GDF9 GDF-9B BMP15 BMP-15 GDF-10 GDF10 BMP-3b GDF-11 GDF11 BMP-11 GDF-15 GDF15 Placental TGF-b, placental BMP (PLAB), PDF (prostate-derived factor), NAG-1 (nonsteroidal anti-inflammatory drug-activated gene-1), MIC-1 (macrophage inhibitory cytokine-1) MIS (Mu¨llerian- AMH AMH (anti-Mu¨llerian hormone) inhibiting substance) A LEFTY2 EBAF (endometrial bleeding-associated factor), TGF-b4, Stra3 Lefty B LEFTY1 a Inhibin A or B is a heterodimer of inhibin a and inhibin bA or bB, respectively. b Activin A or B is a homodimer of inhibin bA or bB, respectively. Activin AB is a heterodimer of inhibin bA and bB. c Activin C or D is a homodimer of inhibin bC or bD, respectively.

Cite this article as Cold Spring Harb Perspect Biol 2016;8:a021873 3 Downloaded from http://cshperspectives.cshlp.org/ on September 24, 2021 - Published by Cold Spring Harbor Laboratory Press

M. Morikawa et al.

dition to the original “transforming” potential, lowed by a cleavage site for a furin protein con- we introduce their diverse roles in the control of vertase that precedes the carboxy-terminal ma- cell proliferation, differentiation, wound heal- ture TGF-b family polypeptide. In the case of ing, and the immune system, and TGF-b’s key the three TGF-b isoforms, the prosegments roles in the context of pathological processes in are also known as latency-associated peptides vivo, for example, connective tissue disorders, (LAPs). Following secretion, the LAPs remain fibrosis, and cancer. noncovalently associated with the mature TGF- b dimer and keep the TGF-bs latent, that is, unable to bind and activate the receptors, in TGF-b FAMILY LIGANDS IN MAMMALS complexes that are called “small latent complex- The name TGF-b derives from the transforming es” (SLCs). Whereas these prosegments are like- activity of the cytokine, which induces anchor- ly to play important roles in folding and trans- age-independent growth when administered to port of the TGF-b family proteins, they were cells together with EGF (Roberts et al. 1981) or shown to remain associated following secretion without the need to add EGF (Shipley et al. with a number of mature TGF-b family pro- 1984), depending on the cell system. At first, teins, such as with BMP-4, BMP-7, BMP-9 two distinct transforming growth factors, that (also known as GDF-2), BMP-10, GDF-5, is, TGF-a and -b, were identified and isolated. GDF-11, myostatin (also known as GDF-8), TGF-a is related to EGF and binds to the EGF and Mu¨llerian-inhibiting substance (MIS, also receptor, whereas TGF-b is structurally distinct known as anti-Mu¨llerian hormone, AMH) from TGF-a. The most striking characteristic, (Wilson et al. 1993; Wolfman et al. 2003; Brown which distinguished the two at that et al. 2005; Ge et al. 2005; Sengle et al. 2008). time, was that TGF-b is a 25-kd disulfide-linked However, only some of the TGF-b family mem- dimer that is reduced to a 12.5-kd band on gels bers are produced as latent forms that are unable following treatment with reducing agents, for to bind the receptors. With the exception of the example, b-mercaptoethanol (Roberts et al. TGF- b1 complex, the roles of the prosegments 1983), whereas TGF-a was a monomeric pro- for other TGF-b family proteins have been tein of smaller size (Roberts et al. 1980). poorly characterized, even for the closely related All TGF-b family members are encoded by TGF-b2 and -b3. much larger precursor polypeptides whose se- The latency and activation mechanisms that quences have been deduced through cDNA result in the release of active TGF-b protein have cloning. The precursor polypeptides are com- been primarily studied in the case of TGF-b1, posed of three segments: an amino-terminal with additional knowledge gained on the acti- signal peptide that is removed during translo- vation of myostatin (Wolfman et al. 2003). The cation of the protein into the lumen of the prosegment of TGF-b1 contains a proline-rich rough endoplasmic reticulum, a large precursor loop, or latency lasso, which surrounds a TGF- segment or prosegment, and the carboxy-ter- b1 monomer like a straitjacket (Shi et al. 2011). minal TGF-b family monomer polypeptide Comparison between TGF-b1 LAP and the for the active and fully mature TGF-b family BMP-9 prosegment, which does not confer la- protein (112 amino acid residues in the case of tency, suggests conformational differences (Mi TGF-b). The prosegments vary in length from et al. 2015). Further analysis of the prosegments 150 to 450 residues and are remarkably not con- of other TGF-b family members, especially served in sequence among TGF-b family mem- nonlatent ones, will reveal functional conserva- bers, even among the three TGF-b isoforms tion among them. (Derynck et al. 1988). Indeed, the sequence sim- For TGF-b to bind to its cell-surface recep- ilarities among TGF-b-related proteins pertain tors, the LAP needs to be released from the ma- exclusively to the sequences of the mature poly- ture peptide. Both the TGF-b1 LAPand -b3 LAP peptides that follow the prosegments. In the contain an integrin recognition motif Arg-Gly- precursor, the prosegment is immediately fol- Asp, or RGD, and bind to several different integ-

4 Cite this article as Cold Spring Harb Perspect Biol 2016;8:a021873 Downloaded from http://cshperspectives.cshlp.org/ on September 24, 2021 - Published by Cold Spring Harbor Laboratory Press

Roles of TGF-b in Cell and Tissue Physiology

rins, for example, avb6 and avb8. Biochemical formation of gastrointestinal tumors (Yoshi- and structural studies revealed that contractile naga et al. 2008). LTBPs are efficiently incorpo- force is necessary for the release of mature TGF- rated in the extracellular matrix (ECM) (Taipale b1 after complex formation between integrin et al. 1994; Dallas et al. 1995), where latent TGF- avb6 and TGF-b1 LAP (Annes et al. 2004; Shi b, as a part of LLC, is stored for future mobili- et al. 2011). In agreement with these findings, zation and activation. Thus, the regulation of mice with RGD-to-RGE mutation in TGF-b1 latency of TGF-b has substantial biological sig- LAP (Yanget al. 2007) and integrin b6-deficient nificance, although it has received little atten- mice (Itgb62/2) (Munger et al. 1999) recapitu- tion. The control of TGF-b latency has been late aspects of the phenotype of Tgfb12/2 mice, reviewed elsewhere (Horiguchi et al. 2012). indicating the importance of the interaction be- The mammalian genome encodes three dif- tween LAP and integrins in latent TGF-b1 acti- ferent TGF-b isoforms, TGF-b1, -b2, and -b3. vation. The SLC covalently attaches the large The mature proteins are highly conserved in latent TGF-b-binding protein (LTBP) through sequence and are marked by the presence of LAP, thus, forming the large latent complex nine cysteine residues. The three-dimensional (LLC). The association of LTBP with the SLC is structure of TGF-b2 reveals that four cysteine also important for proper TGF-b1 function. pairs are formed intramolecularly, but the sixth Mice expressing a mutant TGF-b1 precursor cysteine at position 77 forms the single inter- (C33S) with a prosegment that is unable to molecular disulfide bridge that results in dimer bind LTBP, instead of the wild-type TGF-b1 formation (Fig. 1) (Daopin et al. 1992; Schlu- precursor, are hypomorphic for TGF-b1; negger and Gru¨tter 1992). Tgfb1C33S/C33S mice show decreased levels of ac- Another distinct subfamily is the inhibin b tive TGF-b1, decreased TGF-b signaling, and (or activin b) family. Inhibins are heterodimeric

RXXR Intermolecular

TGF-β, inhibin β C CC CC CC CC

Myostatin, GDF-11 C CC CC CC CC

GDF-15 C CC CC CC CC

BMP/GDF, inhibin α C CC CC CC

BMP-8A, BMP-8B C CC C CC CC

GDF-3 C CCCC CC

GDF-9, GDF-9B C CC C CC

Lefty A, lefty B C CC CC C CC

Figure 1. Comparison of the polypeptide organization of TGF-b family proteins. The precursor protein is cleaved by a furin protein convertase, which mainly recognizes the Arg-X-X-Arg (RXXR) motif, but also the Arg-X-Lys/Arg-Arg (RXK/RR) motif. The mature polypeptides (white) contain seven or nine cysteines con- served in most TGF-b family members. These cysteines engage in inter- and intramolecular disulfide bonds. Human BMP-8A, BMP-8B, and GDF-3 have an additional cysteine residue, which is shown in gray, whereas five members lack the cysteine that mediates the disulfide bond in ligand dimerization. Signal peptide, gray; proseg- ment, black; mature polypeptide, white; TGF-b, transforming growth factor b; BMP, bone morphogenetic protein; GDF, growth and differentiation factor.

Cite this article as Cold Spring Harb Perspect Biol 2016;8:a021873 5 Downloaded from http://cshperspectives.cshlp.org/ on September 24, 2021 - Published by Cold Spring Harbor Laboratory Press

M. Morikawa et al.

proteins consisting of an inhibin a chain and et al. 1995; Israel et al. 1996). BMP-2/GDF-6 an inhibin b chain, whereas activins are homo- heterodimer is also able to regulate ectodermal dimers of inhibin/activin b chains. The four cell fate determination (Chang and Hemmati- known b chains are closely related to each other Brivanlou 1999), and BMP-7/GDF-7 hetero- and have the corresponding set of nine cys- dimer is more potent than the corresponding teines, which is apparent in the TGF-bs. Inhibin homodimers in neuronal differentiation (Butler a, in contrast, is more divergent and has seven and Dodd 2003). cysteines, corresponding to the seven carboxy- Finally, some TGF-b family proteins, lefty A terminal cysteines seen in all TGF-b proteins. and lefty B, GDF-3, and GDF-9 and GDF-9B All other ligands in the TGF-b family, with (also known as BMP-15), lack the fourth cys- five exceptions discussed further below, have teine in the seven-cysteine pattern and thus have a characteristic seven-cysteine pattern that cor- eight or six cysteines (Fig. 1). As this missing responds to the cysteine pattern observed in cysteine mediates in other ligands the disulfide TGF-b and inhibin b, but without its amino- bond dimerization, one should assume that terminal two cysteines (Fig. 1). In these cases, these TGF-b family proteins do not form disul- the disulfide-linked dimerization is mediated by fide-bonded dimers and have alternative means the fourth cysteine in each monomer. Many of of interacting with other TGF-b family pro- these proteins have been named BMP or GDF teins. Lefty is found to associate with nodal followed by a number, based on their sequence and to inhibit nodal signaling through a dual relationship to other BMPs or GDFs, which mechanism involving its interaction with no- were often identified in the same laboratories. dal and the EGF-CFC coreceptors, -1, Within this large group of ligands, there is a FRL-1, or Cryptic, which are required for no- “core” BMP/GDF subfamily of proteins with dal signaling (Chen and Shen 2004; Tabibzadeh similar or related activities. These are structur- and Hemmati-Brivanlou 2006). GDF-3, which ally closely related to each other and are shown also lacks the cysteine required for intermolec- as the top 12 ligands in Figure 2. These BMPs ular disulfide formation, may also function as and GDFs signal through heteromeric complex- an inhibitor of TGF-b/BMP signaling, similarly es that comprise the BMP type I receptors, in- to lefty. Indeed, GDF-3 was reported to func- cluding ALK-1 (activin receptor-like kinase 1), tion as a BMPantagonist (Levine and Brivanlou ALK-2, ALK-3/BMPRIA, or ALK-6/BMPRIB. 2006), whereas it was also reported to act as In contrast, some ligands that have the seven- a nodal-like ligand (Chen et al. 2006; Levine cysteine pattern, like the BMPs and GDFs, do et al. 2009) and to signal possibly through not signal through the same BMP/GDFreceptor ALK-7, a type I receptor for activins (Andersson complexes and intracellular mediators as BMPs et al. 2008). GDF-9B has been reported to and GDFs. Most notably, nodal signals through bind to BMP type I receptor ALK-6/BMPRIB ALK-4, the major type I receptor for activins, and phosphorylate Smad1, Smad5, and Smad8, which is also known as ActRIB, whereas myosta- whereas GDF-9 activates Smad2 through tin signals through ALK-4/ActRIB or ALK-5, ALK-5/TbRI (Moore et al. 2003; Mazerbourg the TGF-b type I receptor also known as TbRI. et al. 2004; Peng et al. 2013). In addition, a As with the heterodimeric inhibins, BMP GDF-9/-9B heterodimer is more bioactive for and/or GDF heterodimers have been identified ovarian functions (Peng et al. 2013). Further and shown to be fully active. In mesodermal characterization of the functions of this sub- differentiation, BMP-4/-7 and -2/-7 hetero- family of TGF-b family proteins is expected. dimers potently induce mesoderm in frog (Su- zuki et al. 1997; Eimon and Harland 1999) and COMPLEXITY OF NOMENCLATURE zebrafish (Little and Mullins 2009). In cultured cells, BMP-2/-7, -4/-7, and -2/-6 heterodimers The name “TGF-b family” derives from the are more active than the corresponding homo- identification of TGF-b1 as a founder member dimers in ectopic bone-formation assays (Aono and its activity in the initial phenotypic trans-

6 Cite this article as Cold Spring Harb Perspect Biol 2016;8:a021873 Downloaded from http://cshperspectives.cshlp.org/ on September 24, 2021 - Published by Cold Spring Harbor Laboratory Press

Roles of TGF-b in Cell and Tissue Physiology

BMP-5 BMP-6/Vgr1 BMP-7/OP-1 BMP-8A/OP-2 BMP-8B BMP-2 BMP-4 BMP-9/GDF-2 BMP-10 GDF-5/CDMP-1 GDF-6/CDMP-2/BMP-13 GDF-7/CDMP-3 GDF-1 GDF-3/Vgr2 BMP-3 GDF-10/BMP-3B Nodal GDF-9 GDF-9B/BMP-15 Lefty A Lefty B Inhibin α MIS/AMH GDF-15/placental TGF-β Inhibin βA Inhibin βB Inhibin βC Inhibin βE Myostatin/GDF-8 GDF-11/BMP-11 TGF-β1 TGF-β2 TGF-β3 0.1

Figure 2. Phylogenetic tree of the 33 TGF-b family polypeptides in human. The amino acid sequences of 33 TGF- b family polypeptides, encoded by their corresponding human genes, were obtained from NCBI’s protein database (www.ncbi.nlm.nih.gov), and the carboxy-terminal mature polypeptides were assigned based on validated or predicted furin cleavage sites. The sequences were aligned by Clustal Omega (www.clustal.org/ omega) (Sievers et al. 2011) and the phylogenetic tree was illustrated using FigTree v1.4.2 (tree.bio.ed.ac.uk/ software/figtree). Ligands that signal through activin- or TGF-b-activated R-Smads or BMP-activated R-Smads are shown in red or blue, respectively. Ligands that may activate these two types of R-Smads, but whose receptors and Smad-signaling pathways have not been fully determined, are shown in orange or light blue, respectively. BMP, Bone morphogenetic protein; OP, osteogenic protein; GDF, growth and differentiation factor; CDMP, cartilage-derived morphogenetic protein; MIS/AMH, Mu¨llerian-inhibiting substance/anti-Mu¨llerian hor- mone; TGF-b, transforming growth factor b.

formation assay. However, this name may be The identification of the BMP/GDF somewhat misleading. The three TGF-b iso- subfamily was based on the isolation and char- forms regulate a wide variety of cellular pro- acterization of BMP-2 and BMP-4 with their cesses, both in development and in the adult ability to elicit ectopic bone formation and af- organisms, and their effects are context-depen- fect skeletal morphogenesis. However, most dent. The three TGF-b isoforms have been es- BMP/GDF proteins were assigned their names tablished as some of the most potent growth because of their sequence similarities to the oth- inhibitors, and inhibit proliferation of most er BMPs/GDFs, whereas their activities remain types of cells. largely uncharacterized. It requires substantial

Cite this article as Cold Spring Harb Perspect Biol 2016;8:a021873 7 Downloaded from http://cshperspectives.cshlp.org/ on September 24, 2021 - Published by Cold Spring Harbor Laboratory Press

M. Morikawa et al.

caution to assume their biological functions older mice with GDF-11 improved muscle re- merely because they share a name, especially generation, a contradictory report claims that for BMPs or GDFs. Recently, several BMP/ serum levels of GDF-11 increase with age (Eger- GDF ligands were rediscovered as critical regu- man et al. 2015). Further work on the biological lators of distinct physiological and pathological activity of GDF-11 is required to confirm the processes. For example, BMP-9 and BMP-10, significance of GDF-11 as a potential youth- which were identified as ligands of the ALK-1 promoting factor. In contrast, GDF-5 expres- receptor (David et al. 2007), play essential roles sion was shown to be associated with muscle in the maintenance of homeostasis of vascular hypertrophy through activation of BMP sig- system and heart development (David et al. naling (Sartori et al. 2013). Another TGF-b 2009; Pardali et al. 2010). Additionally, BMP-6 family protein, MIS/AMH, is secreted by Ser- was identified as an endogenous ligand that reg- toli cells in the testis, and plays important roles ulates expression and iron metabolism in early gonadal development and sex differen- in vivo (Andriopoulos et al. 2009; Meynard et al. tiation through inhibition of the Mu¨llerian duct 2009). GDF-9 and the closely related GDF-9B development. MIS signals through its type II are secreted by oocytes and regulate female fer- receptor, MISRII, and BMPRIA/ALK-3 in tility in several mammals. vivo, which subsequently activates the Smad1 Myostatin/GDF-8 (encoded by the Mstn pathway (Jamin et al. 2002). Mutations in the gene) is now established as a key regulator of gene encoding MIS (Behringer et al. 1994) or mass; it inhibits muscle differ- MISRII (Imbeaud et al. 1995) result in persis- entiation and regeneration, and induces muscle tent Mu¨llerian duct syndrome. Intriguingly, the atrophy through receptor complexes of the sex-determining roles of MIS/MISRII signaling ActRII or ActRIIB type II receptors with ALK-4 are also conserved in fish, non-mammalian ver- or -5 type I receptors that activate the Smad2/ tebrates that lack a Mu¨llerian duct (Morinaga 3 pathway (McPherron et al. 1997; Lee and et al. 2007). McPherron 2001). As shown in Mstn-null Figure 2 and Table 1 show the 33 known mice and in loss-of-function mutations in cattle human TGF-b family polypeptides with their with double-muscling phenotype, mutations in evolutionary relationship calculated based on the Mstn gene result in increased muscle mass sequence similarities. Because multiple and of- with a combination of muscle cell hyperplasia ten parallel approaches have led to the identifi- and hypertrophy (Grobet et al. 1997; Kambadur cation of TGF-b family proteins, it is not sur- et al. 1997; McPherron and Lee 1997; McPher- prising that several ligands became known with ron et al. 1997). GDF-11, which is highly related multiple names (Table 1). Now, most investiga- to myostatin, also signals through ActRII and/ tors have settled on a common name for each or ActRIIB (Oh et al. 2002). Similar to myosta- individual TGF-b family protein; this nomen- tin, GDF-11 has been reported to inhibit myo- clature is used in Figure 2 and Table 1. blast differentiation and regulate the same tar- With genome-wide and transcriptome get genes (Egerman et al. 2015). It is worth analyses gaining in popularity, much research noting that soluble ActRIIB-Fc, a ligand trap has involved the control of TGF-b family mem- for myostatin, activin, and GDF-11, prevents bers. However, some discrepancies between the muscle wasting and reverses loss of skeletal mus- “official gene symbols” and the commonly used cle and cardiac atrophy in tumor-bearing mice names for TGF- b family proteins have caused with cachexia (Zhou et al. 2010). Recently, confusion for researchers and students who en- GDF-11 has been identified as a circulating fac- ter the field. For example, BMP-9 is the accepted tor in young mice, which reverses age-related and commonly used name for the protein, but dysfunction in mouse cardiac and skeletal mus- its official gene symbol is GDF2. Lefty A and cle (Loffredo et al. 2013; Sinha et al. 2014). lefty B are widely used names for the proteins, Whereas these reports claimed that serum levels but the corresponding official gene symbols are of GDF-11 decrease with age and that treating Lefty2 and Lefty1, respectively.

8 Cite this article as Cold Spring Harb Perspect Biol 2016;8:a021873 Downloaded from http://cshperspectives.cshlp.org/ on September 24, 2021 - Published by Cold Spring Harbor Laboratory Press

Roles of TGF-b in Cell and Tissue Physiology

TGF-b SIGNALING IN THE CONTROL thermore, chromatin immunoprecipitation OF GENE EXPRESSION (ChIP) with promoter array analysis (ChIP- chip) and ChIP followed by sequencing More than 30 years of research on these multi- (ChIP-seq) revealed chromatin-binding land- functional ligands has revealed different and scapes of Smad proteins (Chen et al. 2008; Koi- sometimes opposite responses to TGF-b, which numa et al. 2009a,b; Qin et al. 2009). Some strongly depend on the cellular context. Most of these are defined through Smad binding information on the activities of TGF-b ligands with lineage-specific transcription factors at derives from studies using cells cultured in me- corresponding enhancer regions (Chen et al. dia that are supplemented with cytokine-con- 2008; Lee et al. 2011; Morikawa et al. 2011; Mul- taining serum, thus allowing for signaling cross len et al. 2011; Trompouki et al. 2011). Forced talk. Although Smads directly target and bind expression of different lineage-specific tran- regulatory gene sequences, theyeitheractivate or scription factors then redirects Smads to novel repress gene expression in cooperationwith high binding regions of different lineages (Mullen affinity DNA-binding transcription factors and et al. 2011; Trompouki et al. 2011). These find- transcription coregulators that are controlled by ings suggest that the binding of Smad complex- and/or dependent on other signaling pathways, es to target DNA sequences is defined through therefore providing scenarios for highlycontext- cooperation with other DNA-binding tran- dependent transcription responses controlled scription cofactors in two different ways: (1) by signaling cross talk. Furthermore, Smad acti- cell-type- or lineage-specific transcription fac- vation and Smad activities are regulated byother tors, or pioneer factors, open up local chroma- kinase and ubiquitylation pathways. Addition- tin structure to make it accessible for Smads, ally, Smad activation in response to TGF-b and (2) DNA-binding cofactors that are in- ligand-induced receptor activation is comple- duced and activated in a context-dependent mented by TGF-b/BMP-induced activation of manner strengthen the interaction between non-Smad-signaling pathways that may be sep- Smads and DNA. The control of gene expres- arately controlled. The combination of this sig- sion by Smads has been reviewed elsewhere naling cross talk at multiple levels, together with (Morikawa et al. 2013; Gaarenstroom and Hill differences in the physiology of different cell 2014). types, provides the basis for the high context In addition to genes that encode proteins, dependence of the responses to TGF-b. TGF-b also regulates expression of noncoding Several genes, such as the SMAD7 gene RNAs, such as microRNAs (miRNAs) and long encoding the inhibitory Smad7, SERPINE1 en- noncoding RNAs (lncRNAs). miRNAs are coding plasminogen activator inhibitor 1, or 19–25 nucleotides long (with an average CDKN1A encoding the cyclin-dependent ki- of 22 nucleotides), and are transcribed as long nase inhibitor p21Cip1/Waf1, have been estab- primary transcripts with hairpin structures lished as direct Smad target genes that are (pri-miRNAs). Most pri-miRNAs derive from generally and rapidly induced after TGF-b introns of coding genes or are independently treatment in many cell types (Lund et al. 1987; transcribed as noncoding transcripts. In the Datto et al. 1995b; Nakao et al. 1997; Koinuma nucleus, a microprocessor complex contain- et al. 2009b). The development of DNA micro- ing the RNase III enzyme Drosha and the dou- arrays, which allows simultaneous monitoring ble-stranded RNA (dsRNA)-binding protein of thousands of transcripts, has revealed “syn- DGCR8 (DiGeorge syndrome critical region 8) expression groups” of genes that are coregulated specifically recognizes and cleaves pri-miRNAs (Niehrs and Pollet 1999). FoxO and C/EBPb to generate precursor miRNAs (pre-miRNAs), proteins were identified as some of the first which are exported to the cytoplasm, where examples of transcription factors that control another RNase III Dicer generates a duplex the expression of TGF-b/Smad-regulated syn- of 19–22 nucleotides long. One strand of the expression groups (Gomis et al. 2006a,b). Fur- duplex is incorporated into the RNA-induced

Cite this article as Cold Spring Harb Perspect Biol 2016;8:a021873 9 Downloaded from http://cshperspectives.cshlp.org/ on September 24, 2021 - Published by Cold Spring Harbor Laboratory Press

M. Morikawa et al.

silencing complex, which posttranscriptionally epithelial cells and keratinocytes (Siegel and regulates hundreds of target mRNAs (Blahna Massague´ 2003, 2008). The extent of the growth and Hata 2013). inhibitory response to TGF-b varies depending TGF-b regulates directly or indirectly the on the cell type and the particular cell system expression of pri-miRNA transcript and the studied, and reaches a growth arrest in some cell generation of mature miRNAs. Among them, types (Tucker et al. 1984). Confirming obser- miRNA-200 family (miR-200a, -200b, -200c, vations in cell culture, evidence for an antipro- -141, and -429) and miR-205 are markedly liferative effect of TGF-b1 in vivo was first down-regulated in cells that have undergone obtained with polymer beads impregnated with epithelial-mesenchymal transition (EMT) in TGF-b1 and implanted near the epithelial end response to TGF-b. TGF-b indirectly represses buds of immature mammary glands (Silberstein miR-200 family members through induction and Daniel 1987). Also, intravenous injection of of ZEB1 (also known as dEF1) and ZEB2 TGF-b1or-b2 inhibits proliferation of the re- (also known as Smad-interacting protein 1, or generating rat (Russell et al. 1988). The SIP1) transcription factors, and miR-200 family cytostatic effects by TGF-b are mediated mainly members target and inhibit the expression of through two sets of mechanisms: induction of ZEB proteins, which stabilize regulatory net- cyclin-dependent kinase inhibitors and elimi- works for TGF-b-induced EMT (Gregory et nation of proliferative drivers. TGF-b induces al. 2008; Park et al. 2008). In addition, TGF-b expression of p15Ink4b, p21Cip1/Waf1, and activates processing of pri-miRNAs. R-Smads p27Kip1 (Hannon and Beach 1994; Polyak et al. interact with RNA helicase p68 (also known 1994; Datto et al. 1995a), and inhibits the ex- as DDX5), a component of the Drosha micro- pression of mediators that contribute to cell processor complex, to promote pri-miRNA proliferation, such as c-Myc (Pietenpol et al. processing by Drosha (Davis et al. 2008). The 1990), Cdc25A (Iavarone and Massague´ 1997), role of TGF-b signaling in the control of and Id proteins (Kang et al. 2003; Anido et al. miRNA generation is more extensively dis- 2010). In addition to direct effects of TGF-b cussed elsewhere (Butz et al. 2012; Hata and signaling on cell-cycle control, TGF-b opposes Lieberman 2015). the action of specific mitogens, such as EGF in Similarly to miRNAs, the emerging physio- keratinocytes (Coffey et al. 1988), or platelet- logical and pathological roles of lncRNAs have derived growth factor (PDGF) in rat fibroblast started to attract the interest of investigators. and embryonic cells (Roberts et al. 1985; An- LncRNAs are noncoding RNAs usually longer zano et al. 1986). How TGF-b signaling inter- than 200 nucleotides. Several lncRNAs have feres with the growth stimulation resulting from been reported to play essential roles in TGF-b- growth factor-induced receptor tyrosine kinase regulated biological processes. Future work will signaling has remained largely unexplored. confirm their biological significance. In contrast to the growth inhibition in many cell types, TGF-b can also promote cell prolif- eration in other cell types under certain condi- CONTROL OF CELL PROLIFERATION tions, as initially reported (Roberts et al. 1981). BY TGF-b TGF-b has been shown to stimulate prolifera- TGF-b ligands convey strong growth inhibitory tion of several cell types, including chondro- activity in most of the cell types, and this inhi- cytes, osteoblasts under some conditions, mes- bition is reversible after ligand removal (Tucker enchymal stem cells (MSCs), some fibroblasts, et al. 1984; Roberts et al. 1985; Ohta et al. 1987; and endothelial cells under some conditions Kimchi et al. 1988). Although the antiprolifer- (Roberts et al. 1981; Goumans et al. 2002; Jian ative response is observed in many cell types, et al. 2006). As mentioned, the effects are often including epithelial, endothelial, hematopoiet- context-dependent because TGF-b can induce ic, and glial cells, most knowledge about the both growth promotion and growth inhibition molecular mechanisms comes from studies in in the same cells depending on the cell culture

10 Cite this article as Cold Spring Harb Perspect Biol 2016;8:a021873 Downloaded from http://cshperspectives.cshlp.org/ on September 24, 2021 - Published by Cold Spring Harbor Laboratory Press

Roles of TGF-b in Cell and Tissue Physiology

conditions (Roberts et al. 1985). Thus, not only phenotype in the immune system (Shull et al. may different cell types respond differently to 1992). In addition, mice deficient in TGF-b2 TGF-b, but also the same cell type may show and -b3 expression reveal defects in the central opposite responses under different experimen- nervous system (Vogel et al. 2010). tal conditions. The molecular mechanisms re- Among a wide variety of effects on many sponsible for inducing proliferation are less lineages, TGF-b ligands play essential roles in defined than those that lead to inhibition of determining the direction and extent of mesen- proliferation. In some contexts, the growth- chymal differentiation, as first shown in ecto- promoting effect seems to be secondary to in- derm explants from frog (Kimelman duction of other cytokines such as PDGF (Leof and Kirschner 1987; Rosa et al. 1988). In cul- et al. 1986; Matsuyama et al. 2003; Bruna et al. tured cells, TGF-b1 inhibits differentiation to- 2007). The control of cell proliferation has been ward adipocytes (Ignotz and Massague´ 1985) reviewed elsewhere (Siegel and Massague´ 2003, and skeletal myocytes (Florini et al. 1986; Mas- 2008). sague´ et al. 1986). In contrast, TGF-b1 and -b2 enhance differentiation toward chondrocytes, consistent with their independent identification CONTROL OF CELL DIFFERENTIATION and purification as cartilage-inducing factors A AND STEMNESS BY TGF-b PROTEINS and B (CIF-A and CIF-B), respectively (Seye- TGF-b family members control the develop- din et al. 1985). Additionally, TGF-b promotes ment, differentiation, and function of diverse or represses the expression of differentiation cell types. For example, activin A, which uses markers by bone matrix-depositing osteoblasts, similar repertoires of receptors and intracellular depending on their extent of differentiation. In- signaling pathways as TGF-b, was identified as a hibition of endogenous TGF-b and activin sig- critical mesoderm-inducing factor in vivo (Asa- naling in human MSCs by pharmacological in- shima et al. 1990) as well as an erythroid differ- hibition of the ALK-4, -5, and -7 type I receptor entiation factor (EDF) (Eto et al. 1987; Yu et al. kinases, induces osteoblast maturation (Maeda 1987) and a nerve cell survival factor (Schubert et al. 2004), and transcriptome analysis further et al. 1990). Additionally, the Spemann’s orga- supports the notion that inhibition of ALK- nizer in early secretes 5/TbRI-mediated TGF-b signaling enhances extracellular antagonists, such as , adipogenic and osteogenic differentiation, and which prevents activin signaling, and the BMP prevents chondrogenic differentiation (Ng et al. inhibitor (Smith and Harland 1992; 2008). Hemmati-Brivanlou et al. 1994), which are crit- Complementing the control of MSC differ- ical for the establishment of morphogen gradi- entiation toward specific lineages, TGF-b helps ents of activins and BMPs. maintain MSCs in an undifferentiated state Similar to the developmental roles of (Jian et al. 2006). In serum-free media with ba- activin, TGF-b ligands are implicated in differ- sic fibroblast growth factor (FGF-2), PDGF-BB, entiation toward a wide variety of lineages, in- and insulin, TGF-b1 enables multiple passages cluding immune cells (Li and Flavell 2008), of MSCs, and inhibition of any of these signal- blood cells (Blank and Karlsson 2015), and neu- ing pathways decreases proliferation of MSCs, ral/neuronal cells (Krieglstein et al. 2011). Con- suggesting a requirement of these tyrosine ki- sistently, TGF-b2-deficient mice show defects nase-activated pathways for MSC proliferation in multiple organs, including heart, , cra- (Ng et al. 2008). Thus, the cooperation of TGF- niofacial, limb, , eye, inner ear, and b with other cytokines plays important roles urogenital tracts (Sanford et al. 1997), and TGF- in the maintenance and expansion of MSC pop- b3-deficient mice display defects during pul- ulations before differentiation commitment, monary and palate development, suggesting while also restricting their differentiation po- its critical roles in EMT (Kaartinen et al. tential, inhibiting differentiation along the os- 1995), whereas TGF-b1-deficient mice show a teoblast, myoblast, and adipocyte lineages.

Cite this article as Cold Spring Harb Perspect Biol 2016;8:a021873 11 Downloaded from http://cshperspectives.cshlp.org/ on September 24, 2021 - Published by Cold Spring Harbor Laboratory Press

M. Morikawa et al.

TGF-b signaling also controls the mainte- the cancer-initiating cell populations in certain nance of the undifferentiated state and pluri- cancers, including breast cancer, pancreatic can- potency of embryonic stem cells (ESCs). In cer, and diffuse-type gastric cancer (Tang et al. mouse ESCs, leukemia inhibitory factor (LIF), 2007; Ehata et al. 2011; Hoshino et al. 2015). an interleukin 6 (IL-6) class cytokine, and These observations suggest that targeting the BMP-4 play essential roles in the maintenance TGF-b/activin-signaling pathways could be an of pluripotency. However, in this model of attractive therapeutic basis for some advanced “naı¨ve pluripotency,” the role of TGF-b and ac- cancers, although inhibition of these pathways tivin signaling remains to be better defined. in normal tissues may increase the risk for de- Whereas kinase inhibitors for TGF-b/activin velopment of other tumors. The roles of TGF-b type I receptors were reported to help maintain in cancer stem cells and cancer progression are the naı¨ve pluripotent state (Hassani et al. 2012), reviewed elsewhere (Caja et al. 2012; Katsuno nodal/activin signaling was also shown to en- et al. 2013). hance self-renewal of mouse ESCs (Ogawa et al. 2007). TGF-b CONTROLS WOUND HEALING Human ESCs, which share characteristics with mouse epiblast stem cells derived from Normal wound healing is a complex process postimplantation embryos, represent a more that involves (1) cell migration and inflamma- mature stage in pluripotency, and are “primed” tion, (2) proliferation of fibroblasts with forma- for differentiation. These cells are usually main- tion of granulation tissue and ECM deposition, tained with FGF-2 and activin A (Beattie et al. and (3) remodeling of scar tissue for an extend- 2005; James et al. 2005; Vallier et al. 2005), ed time period. TGF-bs have been shown to which stabilize the primed pluripotent state. regulate these different steps through effects They no longer require LIF, which has essential on multiple cell types, and to promote the roles in the naı¨ve state of mouse ESCs. Defined wound healing process in vivo. Indeed, early culture conditions were reported to achieve a studies, initiated shortly after the isolation and naı¨ve pluripotent state for human ESCs and, purification of TGF-b1, revealed that TGF-b1 in this context, two groups used TGF-b or acti- strongly accelerates wound healing in vivo vin ligands (Gafni et al. 2013; Theunissen et al. (Sporn et al. 1983). 2014) rather than their inhibitor(s). The roles of TGF-b1 expression and activation are rap- TGF-b family proteins in stem cells are reviewed idly induced in response to injury, progress- elsewhere (Oshimori and Fuchs 2012; Sakaki- ing outward from the site of injury (Kane Yumoto et al. 2013). et al. 1991). Platelets, which immediately after Accumulating evidence also indicates essen- wounding facilitate the formation of the hemo- tial roles of TGF-b/activin signaling in the static plug, store large amounts of TGF-b1 (As- maintenance of stem-cell-like properties of soian et al. 1983) that is then released at the site some cancer-initiating cell populations, such of injury. Because TGF-b1 acts as a potent che- as glioma-initiating cells (Ikushima et al. moattractant for monocytes (Wahl et al. 1987) 2009; Penuelas et al. 2009), mammary cancer and fibroblasts (Postlethwaite et al. 1987), the stem cells (Mani et al. 2008), pancreatic can- platelet-derived TGF-b1 is able to attract both cer–initiating cells (Lonardo et al. 2011), and cell populations to sites of inflammation and leukemia-initiating cells in chronic myeloid leu- repair. Accordingly, administration of TGF-b1 kemia (Naka et al. 2010). Consistent with these into wound chambers, or to incisional wounds, observations, pharmacological inhibition of stimulates the accumulation of granulation TGF-b/activin type I receptor kinases reduces tissue and the cellularization of the wound cancer progression in some animal models bed, and accelerates the wound healing re- (Ikushima et al. 2009; Penuelas et al. 2009; sponse in experimental models (Sporn et al. Naka et al. 2010; Lonardo et al. 2011). On the 1983; Roberts et al. 1986; Mustoe et al. 1987). other hand, TGF-b was also shown to reduce TGF-b1 is also a potent inducer of the expres-

12 Cite this article as Cold Spring Harb Perspect Biol 2016;8:a021873 Downloaded from http://cshperspectives.cshlp.org/ on September 24, 2021 - Published by Cold Spring Harbor Laboratory Press

Roles of TGF-b in Cell and Tissue Physiology

sion of major ECM proteins, such as fibronectin phylactic use of recombinant TGF-b3 ligand in and collagens (Ignotz and Massague´ 1986; Rob- phase I/II trial (Ferguson et al. 2009). erts et al. 1986), thus promoting ECM deposi- tion at the site of wound healing. Furthermore, TGF-b CONTROLS THE IMMUNE SYSTEM TGF-b also induces inhibition of ECM degra- dation as a result of repressed expression of TGF-b acts as a potent immunosuppressive cy- metalloproteinases and induction of tissue in- tokine through effects on both cell differentia- hibitor of metalloproteinase synthesis (Edwards tion and cell proliferation. For example, TGF-b et al. 1987). inhibits proliferation of T-lymphocytes (Kehrl The repression of the epithelial phenotype et al. 1986) and thymocytes (Ristow 1986), and with its apical-basal polarity and epithelial junc- TGF-b2 was isolated as glioblastoma-derived tion architecture in response to TGF-b, and T-cell suppressor factor, based on the observa- concomitant induction of mesenchymal traits tion that glioblastoma is frequently accompa- with increased cell motility also contribute to nied by immunosuppression (de Martin et al. epithelial wound healing. At the site of injury, 1987). The key roles of TGF-b1 in suppressing epithelial cells undergo partial or complete immune responses also explain the rapid neo- EMT as a result of increased TGF-b signaling, natal development of overt inflammation in therefore promoting epithelial resurfacing, and TGF-b1-deficient mice (Shull et al. 1992). then reacquire their epithelial phenotype (Kal- In contrast, TGF-b2- and -b3-deficient mice luri and Weinberg 2009). TGF-b has also been display developmental defects (Kaartinen et al. shown to induce the expression of a-smooth 1995; Sanford et al. 1997), emphasizing their muscle actin (Desmouliere et al. 1993), a marker roles in development rather than in immune of myofibroblasts. In addition, TGF-b treat- regulation. Consistent with the phenotype of ment enhances acquisition of the myofibroblas- Tgfb12/2 mice, T-cell-specific expression of a tic phenotype, and costimulation with FGF-2 is dominant-negative TGF-b type II receptor shown to inhibit the process (Shirakihara et al. (TbRII) mutant, and T-cell-specific ablation 2011). This and other observations raise the of TbRII or TbRI/ALK-5 show that inhibition possibility that increased TGF-b stimulation of TGF-b signaling in T cells causes neonatal during wound healing may result in or contrib- lethal inflammatory disease. ute to the generation of myofibroblasts. Conversely, TGF-b1 promotes T-cell differ- Although TGF-b1, -b2, and -b3 act through entiation. Similarities in the autoimmune phe- the same receptors and Smad2/3 pathway, they notype of mice lacking expression of TGF-b1 show differences in the control of wound heal- (Tgfb12/2) or Foxp3, the transcription factor ing that remain to be fully explained. Adminis- required for CD4þCD25þ regulatory T cells tration of TGF-b1or-b2 promotes ECM de- (Treg) (Brunkow et al. 2001), led to the discov- position in the early stages of wound healing, ery that TGF-b, in combination with IL-2, but wounds treated with TGF-b1or-b2donot induces Treg cell differentiation through in- differ from control wounds in the final quality of duction of Foxp3 expression in CD4þ T cells scarring (Shah et al. 1995). However, inhibition (Chen et al. 2003; Liu et al. 2008). TGF-b is of TGF-b/Smad3 signaling, for example, in the also essential for the generation of IL-17- absence of Smad3 expression or in the presence expressing proinflammatory T helper cells of an antibody to TGF-b1and-b2, enhances the (TH17), in combination with IL-6 or -21 (Veld- quality of wound healing and reduces scarring hoen et al. 2006; Korn et al. 2007). Thus, TGF-b (Ashcroft et al. 1999; Amendt et al. 2002). Ad- can induce both regulatory/inhibitory and ditionally, administration of TGF-b3, but not proinflammatory T cells, depending on whether TGF-b1nor-b2, also reduces cutaneous scarring proinflammatory cytokines such as IL-6 are (Shah et al. 1995). Thus, recombinant TGF-b3 present. The roles of TGF-b in the immune sys- could be considered to promote scarless wound tem are reviewed elsewhere (Li and Flavell 2008; healing, which is supported by results from pro- Flavell et al. 2010).

Cite this article as Cold Spring Harb Perspect Biol 2016;8:a021873 13 Downloaded from http://cshperspectives.cshlp.org/ on September 24, 2021 - Published by Cold Spring Harbor Laboratory Press

M. Morikawa et al.

ANOMALOUS TGF-b ACTIVATION rysms and dissections, and aneurysms-osteoar- IN CONNECTIVE TISSUE AND SKELETAL thritis syndrome, are also caused by enhanced DISEASES TGF-b signaling. Mutations that affect TGF-b- signaling pathway components have been re- Gene mutations that result in dysregulation of ported; these include mutations in loci encoding TGF-b latency and confer increased release of TGFB2 (Boileau et al. 2012; Lindsay et al. 2012) activated TGF-b have been implicated in the or TGFB3 (Bertoli-Avella et al. 2015), as well as pathogenesis of connective tissue diseases and other signaling components or regulators, that skeletal diseases. Camurati–Engelmann disease is, TGFBR1 (Loeys et al. 2005, 2006), TGFBR2 (CED) is characterized by a progressive diaphy- (Mizuguchi et al. 2004; Loeys et al. 2005, 2006), seal dysplasia with hyperostosis and sclerosis of SMAD3 (van de Laaret al. 2011), and SKI (Doyle the diaphyses of long . Most individuals in et al. 2012). These syndromes share some clinical CED families have a mutation in the region of features in the aorta, including dilation of the the TGFB1 gene that encodes LAP (Jans- aortic root or ascending aorta and degeneration sens et al. 2000; Kinoshita et al. 2000), which of the medial layer of the aorta. Another com- affects LAP dimerization and results in hyper- mon feature is paradoxical activation of TGF-b activation of TGF-b signaling (Saito et al. 2001; signaling in aortic lesions, although they harbor Tang et al. 2009), whereas no mutations are loss-of-function mutations in TGF-b-signaling found in the domain encoding the mature components. The molecular mechanisms that TGF-b1 peptide. As an underlying mechanism, explain the paradoxical activation have not increased TGF-b may promote migration of been elucidated. The dysregulation of TGF-b stromal cells to sites of bone re- signaling in connective tissue and skeletal dis- sorption (Tanget al. 2009). Accordingly, a type I eases has been discussed elswhere by Gallo receptor kinase inhibitor partially rescues the (Lindsay and Dietz 2011; Akhurst 2012). uncoupled bone remodeling in a mouse model for CED with the mutant TGF-b1 precursor b gene (Tang et al. 2009). INCREASED TGF- SIGNALING DRIVES FIBROSIS Mutations in the gene encoding fibrillin-1 have been causally linked to Marfan syndrome A pivotal role of TGF-b in fibrogenesis was first (Neptune et al. 2003), and their etiology high- revealed when subcutaneous injection of puri- lights important roles of TGF-b in Marfan fied TGF-b1 was seen to induce fibrotic lesions syndrome or Marfan-like connective tissue dis- at the injection site (Roberts et al. 1986). This orders. The fibrillins, which form extracellular potent effect was further confirmed by the fi- microfibrils, associate with LTBP-1, which in- brotic response in of rodents following teracts with the LAP in the LLC, consequently intratracheal administration of adenovirus that linking this complex to elastic microfibrils (Iso- expresses active TGF-b1 (Sime et al. 1997), gai et al. 2003). The absence of fibrillin-1 in whereas neutralization of TGF-b with antise- Fbn1-deficient mice results in excessive TGF-b rum ameliorated experimental fibrosis in the activity in vivo, which could be at the basis of kidney, heart, and liver (Border et al. 1990; De- the pathogenesis of emphysema and other man- nis 1994; Kuwahara et al. 2002). In addition, ifestations of Marfan syndrome (Neptune et al. inhibitors or antagonists of the TGF-b/Smad 2003). Additionally, mutant fibrillin-1 disturbs pathway, such as soluble TbRII (George et al. the microfibril structure (Judge et al. 2004; 1999) or adenovirally expressed Smad7 (Nakao Charbonneau et al. 2010) and activates TGF-b et al. 1999), were shown to prevent liver and signaling, which may then over time generate lung fibrosis in mice, respectively. Most fibro- the pathological features of Marfan syndrome. genic effects resulting from increased TGF-b Clinically overlapping conditions, which re- signaling are thought to be mediated by the sult in aortic aneurysms, for example, in Loeys– Smad pathway because Smad3-null mice are Dietz syndrome, familial thoracic aortic aneu- resistant to bleomycin-induced lung fibrosis

14 Cite this article as Cold Spring Harb Perspect Biol 2016;8:a021873 Downloaded from http://cshperspectives.cshlp.org/ on September 24, 2021 - Published by Cold Spring Harbor Laboratory Press

Roles of TGF-b in Cell and Tissue Physiology

(Zhao et al. 2002), radiation-induced skin fi- vitamin D receptor, PPAR-g (peroxisome pro- brosis (Flanders et al. 2002), and tubulo- liferator-activated receptor-g), and NR4A1, di- interstitial fibrosis following unilateral ureteral rectly repress fibroblast activation induced by obstruction (Sato et al. 2003). In addition, phe- TGF-b (Wu et al. 2009; Ding et al. 2013; Pa- notypes of integrin avb6-deficient mice support lumbo-Zerr et al. 2015). a role of integrin-mediated TGF-b activation in progressive fibrosis because bleomycin-induced TGF-b SIGNALING IN CANCER inflammation can occur without progression to PROGRESSION fibrosis in these mice (Munger et al. 1999). These and other observations suggest that dys- TGF-b induces an antiproliferative response in regulation and sustained activation of TGF-b/ many cell types, including both normal epithe- Smad3 signaling play essential roles in the ini- lial cells and transformed cells. Initial reports tiation and maintenance of the fibrotic tissue showed that antisense inhibition of TGF-b en- phenotype. hances tumorigenicity in vivo (Wu et al. 1992), The profibrotic effects of TGF-b involve a and that certain tumor cells can become un- combination of mechanisms and cell types, in- responsive to TGF-b, which was primarily cluding enhanced infiltration and/or prolifera- assessed by its effects on proliferation (Shipley tion of preexisting fibroblasts, generation of et al. 1986; Kimchi et al. 1988). It was, therefore, myofibroblasts, increased ECM synthesis, and assumed that disruption of TGF-b signaling is inhibition of collagenolysis, all of which are implicated in the pathogenesis of tumor devel- also apparent in wound healing. In addition, opment. In line with this hypothesis, inacti- lineage tracing strongly suggests important con- vating mutations and deletions in the TGFBR2 tributions of mesenchymal cells arising through and SMAD4 gene loci were found in cancers TGF-b-induced EMT. For example, in a model (Markowitz et al. 1995; Hahn et al. 1996) and of tubulointerstitial renal fibrosis, the use of the thought to drive tumorigenesis (reviewed in promoter region of the gene encoding g-gluta- Garraway and Lander 2013; Vogelstein et al. myltranspeptidase, a proximal tubule marker, 2013). for cell lineage tracing suggests that 36% of renal As advocated by Vogelstein and colleagues, fibroblasts derive through EMT from renal tu- the multistep tumorigenesis model (Fearon and bular epithelium (Iwano et al. 2002). Addition- Vogelstein 1990; Vogelstein et al. 2013) involves ally, using the promoter of surfactant protein the requirement of sequential genetic alter- C, a type II alveolar epithelial cell marker, for ations over time, which enables the develop- lineage tracing highlights a substantial con- ment of tumors and promotes cancer progres- tribution of EMT of alveolar epithelial cells sion. In colon cancers, for example, gatekeeping in TGF-b1-induced pulmonary fibrosis (Kim mutations most often occur in the adeno- et al. 2006). Endothelial cells have also been matous polyposis coli (APC) gene, and second identified as a source of fibroblasts in fibrosis. mutations in another gene, such as KRAS, then In heart fibrosis, for example, 27%–35% of all stimulate clonal growth and expansion. Muta- fibroblasts are estimated to originate from en- tions in genes such as PIK3CA, SMAD4, and dothelial cells through endothelial-mesenchy- TP53 follow this mutation process, resulting mal transition (Zeisberg et al. 2007). in more invasive and metastatic tumors. Con- Although TGF-b has been well established ceptually similarly, TGF-b’s function as a sup- as a key profibrotic cytokine, its activity is at- pressor of epithelial cell tumorigenesis may tenuated or aggravated by several other cyto- need to be alleviated at an early stage in tumor kines. For example, IL-1 and tumor necrosis development, which does not preclude roles of factor-a, which cross talk with TGF-b signaling TGF-b signaling at later stages. at multiple levels, are well-characterized profi- On the other hand, tumor cells show in- brotic cytokines. Conversely, some members of creased expression of TGF-b, most commonly the nuclear receptor superfamily, such as the TGF-b1, when compared with normal sur-

Cite this article as Cold Spring Harb Perspect Biol 2016;8:a021873 15 Downloaded from http://cshperspectives.cshlp.org/ on September 24, 2021 - Published by Cold Spring Harbor Laboratory Press

M. Morikawa et al.

rounding tissue, and secrete TGF-b ligands. Ad- multiple roles in developmental patterning, tis- ditionally, elevated TGF-b expression correlates sue differentiation, and maintenance of ho- with tumor progression and poorer prognosis, meostasis. As discussed, the biological effects indicating pro-oncogenic roles for TGF-b at of TGF-b family proteins are contextual, and late stages. Substantial evidence now supports even the same cell type may show different or the notion that the increased TGF-b expression opposite responses to the ligand under different by tumor cells promotes tumor progression by biological contexts. Some TGF-b family mem- enhancing migration, invasion, and survival of bers are made available to cells with concentra- tumor cells during later stages of tumorigenesis, tion gradients in vivo, resulting in graded effects through stimulating ECM deposition and tissue of cell differentiation and function by the re- fibrosis, perturbing immune and inflammatory sponsive cells. The activation of distinctive sets function, stimulating angiogenesis, promoting of genes by the ligand in a dosage-dependent EMT that enables increased migration and in- manner plays essential roles in the specification vasion, and maintaining cancer stem cells. of cell fates during normal development. Cells Platelets attached to tumor cells in the circula- also respond to TGF-b family proteins in com- tion provide an additional source of TGF-b that bination with other external stimuli. Costimu- promotes an invasive mesenchymal-like pheno- lation by other cytokines modifies the respon- type and enhances metastasis in vivo through siveness to the ligand, as observed in the stimulating the TGF-b/Smad and NF-kB path- immune system. In addition, the complexity of ways in the tumor cells (Labelle et al. 2011). the TGF-b family may reflect some functional With the sequential acquisition of genomic redundancy. It is, therefore, possible that some mutations, tumor cells were shown to display a family members could play redundant roles in range of mutations in different signaling path- the regulation of some biological processes. ways, which lead to changes in TGF-b respon- Therefore, detailed profiling of expression and siveness. This is illustrated by a model system activation of all TGF-b family ligands in each consisting of a set of human breast epithelial context could lead to a better understanding of MCF10A–derived cell lines, in which TGF-b the complex nature of TGF-b family members. functions as a tumor suppressor early in the car- Recent advances in single-cell assays have cinogenic process, but switches to a pro-onco- revealed intertumor heterogeneity between tu- genic agent in the later stage. Intriguingly, in the mors of the same histopathological subtype, most aggressive cell line, M-IV,which resembles and intratumor genetic and epigenetic hetero- a high-grade breast carcinoma, loss of TGF-b geneity within individual cancer masses. In ad- response does not affect primary tumorigenesis, dition, characterization of the transcriptome but suppresses metastasis (Tang et al. 2003). and histone modification markers in normal As illustrated with these observations, TGF- cells, such as mouse ESCs, similarly suggests b signaling controls both the initial develop- substantial cell-to-cell variability even in cells ment of tumors as well as the cancer progression with identical genetic backgrounds. It is, there- of a large variety of tumor types. Its roles in fore, possible that the heterogeneity could con- tumor suppression and its extensive roles in tribute to different responsiveness to TGF-b cancer progression through direct effects on family members. Thus, spatial and temporal the cancer cells themselves, as well as the micro- profiling of signaling at the single-cell level environment, are the subject of many reviews will help us to understand the complex mecha- (Bierie and Moses 2006; Ikushima and Miya- nisms of TGF-b family members. zono 2010; Pickup et al. 2013). REFERENCES CONCLUSIONS Akhurst RJ. 2012. The paradoxical TGF-b vasculopathies. Nat Genet 44: 838–839. The TGF-b family comprises a large number of Amendt C, Mann A, Schirmacher P, Blessing M. 2002. secreted and structurally related proteins with Resistance of keratinocytes to TGFb-mediated growth

16 Cite this article as Cold Spring Harb Perspect Biol 2016;8:a021873 Downloaded from http://cshperspectives.cshlp.org/ on September 24, 2021 - Published by Cold Spring Harbor Laboratory Press

Roles of TGF-b in Cell and Tissue Physiology

restriction and apoptosis induction accelerates re-epithe- Blank U, Karlsson S. 2015. TGF-b signaling in the control of lialization in skin wounds. J Cell Sci 115: 2189–2198. hematopoietic stem cells. Blood 125: 3542–3550. Andersson O, Korach-Andre M, Reissmann E, Ibanez CF, Boileau C, Guo DC, Hanna N, Regalado ES, Detaint D, Bertolino P.2008. Growth/differentiation factor 3 signals Gong L, Varret M, Prakash SK, Li AH, d’Indy H, et al. through ALK7 and regulates accumulation of adipose 2012. TGFB2 mutations cause familial thoracic aortic tissue and diet-induced obesity. Proc Natl Acad Sci 105: aneurysms and dissections associated with mild systemic 7252–7256. features of Marfan syndrome. Nat Genet 44: 916–921. Andriopoulos B Jr, Corradini E, Xia Y, Faasse SA, Chen S, Border WA,Okuda S, Languino LR, Sporn MB, Ruoslahti E. Grgurevic L, Knutson MD, Pietrangelo A, Vukicevic S, 1990. Suppression of experimental glomerulonephritis Lin HY, et al. 2009. BMP6 is a key endogenous regulator by antiserum against transforming growth factor b1. Na- of hepcidin expression and iron metabolism. Nat Genet ture 346: 371–374. 41: 482–487. Brown MA, Zhao Q, Baker KA, Naik C, Chen C, Pukac L, Anido J, Saez-Borderias A, Gonzalez-Junca A, Rodon L, Singh M, Tsareva T, Parice Y, Mahoney A, et al. 2005. Folch G, Carmona MA, Prieto-Sanchez RM, Barba I, Crystal structure of BMP-9 and functional interactions Martinez-Saez E, Prudkin L, et al. 2010. TGF-b receptor with pro-region and receptors. J Biol Chem 280: 25111– inhibitors target the CD44high/Id1high glioma-initiating 25118. cell population in human glioblastoma. Cancer Cell 18: Bruna A, Darken RS, Rojo F, Ocana A, Penuelas S, Arias A, 655–668. Paris R, Tortosa A, Mora J, Baselga J, et al. 2007. High Annes JP, Chen Y, Munger JS, Rifkin DB. 2004. Integrin TGFb-Smad activity confers poor prognosis in glioma aVb6-mediated activation of latent TGF-b requires the patients and promotes cell proliferation depending on latent TGF-b binding protein-1. J Cell Biol 165: 723–734. the methylation of the PDGF-B gene. Cancer Cell 11: Anzano MA, Roberts AB, Sporn MB. 1986. Anchorage-in- 147–160. dependent growth of primary rat cells is induced Brunkow ME, Jeffery EW, Hjerrild KA, Paeper B, Clark LB, by platelet-derived growth factor and inhibited by type-b Yasayko SA, Wilkinson JE, Galas D, Ziegler SF,Ramsdell F. transforming growth factor. J Cell Physiol 126: 312–318. 2001. Disruption of a new forkhead/winged-helix pro- Aono A, Hazama M, Notoya K, Taketomi S, Yamasaki H, tein, scurfin, results in the fatal lymphoproliferative dis- Tsukuda R, Sasaki S, Fujisawa Y. 1995. Potent ectopic order of the scurfy mouse. Nat Genet 27: 68–73. bone-inducing activity of bone morphogenetic protein- Butler SJ, Dodd J. 2003. A role for BMP heterodimers in roof 4/7 heterodimer. Biochem Biophys Res Commun 210: plate-mediated repulsion of commissural axons. Neuron 670–677. 38: 389–401. Asashima M, Nakano H, Shimada K, Kinoshita K, Ishii K, Butz H, Racz K, Hunyady L, Patocs A. 2012. Crosstalk be- Shibai H, Ueno N. 1990. Mesodermal induction in early tween TGF-b signaling and the microRNA machinery. amphibian embryos by activin A (erythroid differentia- Trends Pharmacol Sci 33: 382–393. tion factor). Rouxs Arch Dev Biol 198: 330–335. Caja L, Kahata K, Moustakas A. 2012. Context-dependent Ashcroft GS, Yang X, Glick AB, Weinstein M, Letterio JL, action of transforming growth factor b family members Mizel DE, Anzano M, Greenwell-Wild T, Wahl SM, Deng on normal and cancer stem cells. Curr Pharm Des 18: C, et al. 1999. Mice lacking Smad3 show accelerated 4072–4086. wound healing and an impaired local inflammatory re- Chang C, Hemmati-Brivanlou A. 1999. Xenopus GDF6, a sponse. Nat Cell Biol 1: 260–266. new antagonist of noggin and a partner of BMPs. Devel- Assoian RK, Komoriya A, Meyers CA, Miller DM, Sporn opment 126: 3347–3357. MB. 1983. Transforming growth factor-b in human Charbonneau NL, Carlson EJ, Tufa S, Sengle G, Manalo EC, platelets. Identification of a major storage site, purifica- Carlberg VM, Ramirez F, Keene DR, Sakai LY. 2010. In tion, and characterization. J Biol Chem 258: 7155–7160. vivo studies of mutant fibrillin-1 microfibrils. J Biol Beattie GM, Lopez AD, Bucay N, Hinton A, Firpo MT, King Chem 285: 24943–24955. CC, Hayek A. 2005. Activin A maintains pluripotency of Chen C, Shen MM. 2004. Two modes by which Lefty pro- human embryonic stem cells in the absence of feeder teins inhibit nodal signaling. Curr Biol 14: 618–624. layers. Stem Cells 23: 489–495. Chen W, Jin W, Hardegen N, Lei KJ, Li L, Marinos N, Behringer RR, Finegold MJ, Cate RL. 1994. Mu¨llerian-in- McGrady G, Wahl SM. 2003. Conversion of peripheral hibiting substance function during mammalian sexual CD4þCD252 naive T cells to CD4þCD25þ regulatory T development. Cell 79: 415–425. cells by TGF-b induction of transcription factor Foxp3. Bertoli-Avella AM, Gillis E, Morisaki H, Verhagen JM, de J Exp Med 198: 1875–1886. Graaf BM, van de Beek G, Gallo E, Kruithof BP,Venselaar Chen C, Ware SM, Sato A, Houston-Hawkins DE, Habas R, H, Myers LA, et al. 2015. Mutations in a TGF-b ligand, Matzuk MM, Shen MM, Brown CW. 2006. The Vg1-re- TGFB3, cause syndromic aortic aneurysms and dissec- lated protein Gdf3 acts in a in tions. J Am Coll Cardiol 65: 1324–1336. the pre- mouse embryo. Development 133: Bierie B, Moses HL. 2006. TGFb: The molecular Jekyll and 319–329. Hyde of cancer. Nat Rev Cancer 6: 506–520. Chen X, Xu H, Yuan P, Fang F, Huss M, Vega VB, Wong E, Blahna MT, Hata A. 2013. Regulation of miRNA biogenesis Orlov YL, Zhang W, Jiang J, et al. 2008. Integration of as an integrated component of growth factor signaling. external signaling pathways with the core transcriptional Curr Opin Cell Biol 25: 233–240. network in embryonic stem cells. Cell 133: 1106–1117.

Cite this article as Cold Spring Harb Perspect Biol 2016;8:a021873 17 Downloaded from http://cshperspectives.cshlp.org/ on September 24, 2021 - Published by Cold Spring Harbor Laboratory Press

M. Morikawa et al.

Coffey RJ Jr, Bascom CC, Sipes NJ, Graves-Deal R, Weiss- Edwards DR, Murphy G, Reynolds JJ, Whitham SE, Do- man BE, Moses HL. 1988. Selective inhibition of growth- cherty AJ, Angel P,Heath JK. 1987. Transforming growth related gene expression in murine keratinocytes by trans- factor b modulates the expression of collagenase and forming growth factor b. Mol Cell Biol 8: 3088–3093. metalloproteinase inhibitor. EMBO J 6: 1899–1904. Dallas SL, Miyazono K, Skerry TM, Mundy GR, Bonewald Egerman MA, Cadena SM, Gilbert JA, Meyer A, Nelson HN, LF. 1995. Dual role for the latent transforming growth Swalley SE, Mallozzi C, Jacobi C, Jennings LL, Clay I, et al. factor-b binding protein in storage of latent TGF-b in 2015. GDF11 increases with age and inhibits skeletal the extracellular matrix and as a structural matrix pro- muscle regeneration. Cell Metab 22: 164–174. tein. J Cell Biol 131: 539–549. Ehata S, Johansson E, Katayama R, Koike S, Watanabe A, Daopin S, Piez KA, Ogawa Y,Davies DR. 1992. Crystal struc- Hoshino Y,Katsuno Y,Komuro A, Koinuma D, Kano MR, ture of transforming growth factor-b2: An unusual fold et al. 2011. Transforming growth factor-b decreases the for the superfamily. Science 257: 369–373. cancer-initiating cell population within diffuse-type gas- Datto MB, Li Y,Panus JF,Howe DJ, Xiong Y,WangXF.1995a. tric carcinoma cells. 30: 1693–1705. Transforming growth factor b induces the cyclin-depen- Eimon PM, Harland RM. 1999. In Xenopus embryos, BMP dent kinase inhibitor p21 through a p53-independent heterodimers are not required for mesoderm induction, mechanism. Proc Natl Acad Sci 92: 5545–5549. but BMP activity is necessary for dorsal/ventral pattern- Datto MB, Yu Y,Wang XF. 1995b. Functional analysis of the ing. Dev Biol 216: 29–40. transforming growth factor b responsive elements in the Eto Y,Tsuji T, Takezawa M, Takano S, Yokogawa Y,Shibai H. WAF1/Cip1/p21 promoter. J Biol Chem 270: 28623– 1987. Purification and characterization of erythroid dif- 28628. ferentiation factor (EDF) isolated from human leukemia David L, Mallet C, Mazerbourg S, Feige JJ, Bailly S. 2007. cell line THP-1. Biochem Biophys Res Commun 142: Identification of BMP9 and BMP10 as functional activa- 1095–1103. tors of the orphan activin receptor-like kinase 1 (ALK1) Fearon ER, Vogelstein B. 1990. A genetic model for colorec- in endothelial cells. Blood 109: 1953–1961. tal tumorigenesis. Cell 61: 759–767. David L, Feige JJ, Bailly S. 2009. Emerging role of bone mor- Ferguson MW, Duncan J, Bond J, Bush J, Durani P, So K, phogenetic proteins in angiogenesis. Cytokine Growth Taylor L, Chantrey J, Mason T, James G, et al. 2009. Pro- Factor Rev 20: 203–212. phylactic administration of avotermin for improvement Davis BN, Hilyard AC, Lagna G, Hata A. 2008. SMAD pro- of skin scarring: Three double-blind, placebo-controlled, teins control DROSHA-mediated microRNA matura- phase I/II studies. Lancet 373: 1264–1274. tion. Nature 454: 56–61. Flanders KC, Sullivan CD, Fujii M, Sowers A, Anzano MA, de Martin R, Haendler B, Hofer-Warbinek R, Gaugitsch H, Arabshahi A, Major C, Deng C, Russo A, Mitchell JB, et Wrann M, Schlusener H, Seifert JM, Bodmer S, Fontana al. 2002. Mice lacking Smad3 are protected against cuta- A, Hofer E. 1987. Complementary DNA for human glio- neous injury induced by ionizing radiation. Am J Pathol blastoma-derived T cell suppressor factor, a novel mem- 160: 1057–1068. ber of the transforming growth factor-b gene family. EMBO J 6: 3673–3677. Flavell RA, Sanjabi S, Wrzesinski SH, Licona-Limon P.2010. The polarization of immune cells in the tumour environ- Denis M. 1994. Neutralization of transforming growth fac- ment by TGFb. Nat Rev Immunol 10: 554–567. tor-b1 in a mouse model of immune-induced lung fibro- sis. Immunology 82: 584–590. Florini JR, Roberts AB, Ewton DZ, Falen SL, Flanders KC, Sporn MB. 1986. Transforming growth factor-b.Avery Derynck R, Jarrett JA, Chen EY,Eaton DH, Bell JR, Assoian potent inhibitor of myoblast differentiation, identical to RK, Roberts AB, Sporn MB, Goeddel DV. 1985. Human the differentiation inhibitor secreted by Buffalo rat liver transforming growth factor-b complementary DNA se- cells. J Biol Chem 261: 16509–16513. quence and expression in normal and transformed cells. Nature 316: 701–705. Gaarenstroom T,Hill CS. 2014. TGF-b signaling to chroma- tin: How Smads regulate transcription during self-renew- Derynck R, Lindquist PB, Lee A, WenD, TammJ, Graycar JL, al and differentiation. Semin Cell Dev Biol 32: 107–118. Rhee L, Mason AJ, Miller DA, Coffey RJ, et al. 1988. A new type of transforming growth factor-b, TGF-b3. Gafni O, Weinberger L, Mansour AA, Manor YS, Chomsky EMBO J 7: 3737–3743. E, Ben-YosefD, Kalma Y,Viukov S, Maza I, Zviran A, et al. Desmouliere A, Geinoz A, Gabbiani F, Gabbiani G. 1993. 2013. Derivation of novel human ground state naive plu- Transforming growth factor-b1 induces a-smooth mus- ripotent stem cells. Nature 504: 282–286. cle actin expression in granulation tissue myofibroblasts Garraway LA, Lander ES. 2013. Lessons from the cancer and in quiescent and growing cultured fibroblasts. J Cell genome. Cell 153: 17–37. Biol 122: 103–111. Ge G, Hopkins DR, Ho WB, Greenspan DS. 2005. GDF11 Ding N, Yu RT, Subramaniam N, Sherman MH, Wilson C, forms a bone morphogenetic protein 1–activated latent Rao R, Leblanc M, Coulter S, He M, Scott C, et al. 2013. A complex that can modulate nerve growth factor-induced vitamin D receptor/SMAD genomic circuit gates hepatic differentiation of PC12 cells. Mol Cell Biol 25: 5846– fibrotic response. Cell 153: 601–613. 5858. Doyle AJ, Doyle JJ, Bessling SL, Maragh S, Lindsay ME, George J, Roulot D, Koteliansky VE, Bissell DM. 1999. In Schepers D, Gillis E, Mortier G, Homfray T, Sauls K, et vivo inhibition of rat stellate cell activation by soluble al. 2012. Mutations in the TGF-b repressor SKI cause transforming growth factor b type II receptor: A poten- Shprintzen–Goldberg syndrome with aortic aneurysm. tial new therapy for hepatic fibrosis. Proc Natl Acad Sci Nat Genet 44: 1249–1254. 96: 12719–12724.

18 Cite this article as Cold Spring Harb Perspect Biol 2016;8:a021873 Downloaded from http://cshperspectives.cshlp.org/ on September 24, 2021 - Published by Cold Spring Harbor Laboratory Press

Roles of TGF-b in Cell and Tissue Physiology

Gomis RR, Alarcon C, He W, Wang Q, Seoane J, Lash A, Ikushima H, Miyazono K. 2010. TGFb signalling: A com- Massague´ J. 2006a. A FoxO-Smad synexpression group in plex web in cancer progression. Nat Rev Cancer 10: 415– human keratinocytes. Proc Natl Acad Sci 103: 12747– 424. 12752. Ikushima H, Todo T, Ino Y, Takahashi M, Miyazawa K, Gomis RR, Alarcon C, Nadal C, Van Poznak C, Massague´ J. Miyazono K. 2009. Autocrine TGF-b signaling maintains 2006b. C/EBPb at the core of the TGFb cytostatic re- tumorigenicity of glioma-initiating cells through Sry-re- sponse and its evasion in metastatic breast cancer cells. lated HMG-box factors. Cell 5: 504–514. Cancer Cell 10: 203–214. Imbeaud S, Faure E, Lamarre I, Mattei MG, di Clemente N, Goumans MJ, Valdimarsdottir G, Itoh S, Rosendahl A, Side- Tizard R, Carre-Eusebe D, Belville C, Tragethon L, Ton- ras P, ten Dijke P. 2002. Balancing the activation state of kin C, et al. 1995. Insensitivity to anti-Mu¨llerian hor- the endothelium via two distinct TGF-b type I receptors. mone due to a mutation in the human anti-Mu¨llerian EMBO J 21: 1743–1753. hormone receptor. Nat Genet 11: 382–388. Gregory PA, Bert AG, Paterson EL, Barry SC, Tsykin A, Isogai Z, Ono RN, Ushiro S, Keene DR, Chen Y,Mazzieri R, Farshid G, Vadas MA, Khew-Goodall Y, Goodall GJ. Charbonneau NL, Reinhardt DP, Rifkin DB, Sakai LY. 2008. The miR-200 family and miR-205 regulate epithe- 2003. Latent transforming growth factor b-binding pro- lial to mesenchymal transition by targeting ZEB1 and tein 1 interacts with fibrillin and is a microfibril-associ- SIP1. Nat Cell Biol 10: 593–601. ated protein. J Biol Chem 278: 2750–2757. Grobet L, Martin LJ, Poncelet D, Pirottin D, Brouwers B, Israel DI, Nove J, Kerns KM, Kaufman RJ, Rosen V,Cox KA, Riquet J, Schoeberlein A, Dunner S, Menissier F, Massa- Wozney JM. 1996. Heterodimeric bone morphogenetic banda J, et al. 1997. A deletion in the bovine myostatin proteins show enhanced activity in vitro and in vivo. gene causes the double-muscled phenotype in cattle. Nat Growth Factors 13: 291–300. Genet 17: 71–74. Iwano M, Plieth D, Danoff TM, Xue C, Okada H, Neilson Hahn SA, Schutte M, Hoque AT,Moskaluk CA, da Costa LT, EG. 2002. Evidence that fibroblasts derive from epitheli- Rozenblum E, Weinstein CL, Fischer A, Yeo CJ, Hruban um during tissue fibrosis. J Clin Invest 110: 341–350. RH, et al. 1996. DPC4, a candidate tumor suppressor James D, Levine AJ, Besser D, Hemmati-Brivanlou A. 2005. gene at human 18q21.1. Science 271: TGFb/activin/nodal signaling is necessary for the main- 350–353. tenance of pluripotency in human embryonic stem cells. INK4B Development 132: 1273–1282. Hannon GJ, Beach D. 1994. p15 is a potential effector of TGF-b-induced cell cycle arrest. Nature 371: 257–261. Jamin SP,Arango NA, Mishina Y,Hanks MC, Behringer RR. 2002. Requirement of Bmpr1a for Mu¨llerian duct regres- Hassani SN, Totonchi M, Farrokhi A, Taei A, Larijani MR, sion during male sexual development. Nat Genet 32: Gourabi H, Baharvand H. 2012. Simultaneous suppres- 408–410. sion of TGF-b and ERK signaling contributes to the high- ly efficient and reproducible generation of mouse embry- Janssens K, Gershoni-Baruch R, Guanabens N, Migone N, onic stem cells from previously considered refractory and Ralston S, Bonduelle M, Lissens W, Van Maldergem L, non-permissive strains. Stem Cell Rev 8: 472–481. Vanhoenacker F, Verbruggen L, et al. 2000. Mutations in the gene encoding the latency-associated peptide of TGF- Hata A, Lieberman J. 2015. Dysregulation of microRNA b1 cause Camurati–Engelmann disease. Nat Genet 26: biogenesis and gene silencing in cancer. Sci Signal 8: re3. 273–275. Hemmati-Brivanlou A, Kelly OG, Melton DA. 1994. Folli- Jian H, Shen X, Liu I, Semenov M, He X, Wang XF. 2006. statin, an antagonist of activin, is expressed in the Spe- Smad3-dependent nuclear translocation of b-catenin is mann organizer and displays direct neuralizing activity. required for TGF-b1-induced proliferation of bone mar- Cell 77: 283–295. row-derived adult human mesenchymal stem cells. Genes Horiguchi M, Ota M, Rifkin DB. 2012. Matrix control of Dev 20: 666–674. transforming growth factor-b function. J Biochem 152: Judge DP, Biery NJ, Keene DR, Geubtner J, Myers L, Huso 321–329. DL, Sakai LY, Dietz HC. 2004. Evidence for a critical Hoshino Y, Nishida J, Katsuno Y, Koinuma D, Aoki T, Ko- contribution of haploinsufficiency in the complex path- kudo N, Miyazono K, Ehata S. 2015. Smad4 decreases the ogenesis of Marfan syndrome. J Clin Invest 114: 172–181. population of pancreatic cancer-initiating cells through Kaartinen V, Voncken JW, Shuler C, Warburton D, Bu D, transcriptional repression of ALDH1A1. Am J Pathol 185: Heisterkamp N, Groffen J. 1995. Abnormal lung devel- 1457–1470. opment and cleft palate in mice lacking TGF-b3 indicates Iavarone A, Massague´ J. 1997. Repression of the CDK acti- defects of epithelial-mesenchymal interaction. Nat Genet vator Cdc25A and cell-cycle arrest by cytokine TGF-b in 11: 415–421. cells lacking the CDK inhibitor p15. Nature 387: 417– Kalluri R, Weinberg RA. 2009. The basics of epithelial-mes- 422. enchymal transition. J Clin Invest 119: 1420–1428. Ignotz RA, Massague´ J. 1985. Type b transforming growth Kambadur R, Sharma M, Smith TP,Bass JJ. 1997. Mutations factor controls the adipogenic differentiation of 3T3 fi- in myostatin (GDF8) in double-muscled Belgian Blue broblasts. Proc Natl Acad Sci 82: 8530–8534. and Piedmontese cattle. Genome Res 7: 910–916. Ignotz RA, Massague´ J. 1986. Transforming growth factor b Kane CJ, Hebda PA, Mansbridge JN, Hanawalt PC. 1991. stimulates the expression of fibronectin and collagen and Direct evidence for spatial and temporal regulation of their incorporation into the extracellular matrix. J Biol transforming growth factor b1 expression during cuta- Chem 261: 4337–4345. neous wound healing. J Cell Physiol 148: 157–173.

Cite this article as Cold Spring Harb Perspect Biol 2016;8:a021873 19 Downloaded from http://cshperspectives.cshlp.org/ on September 24, 2021 - Published by Cold Spring Harbor Laboratory Press

M. Morikawa et al.

Kang Y, Chen CR, Massague´ J. 2003. A self-enabling TGFb Lagna G, Hata A, Hemmati-Brivanlou A, Massague´ J. 1996. response coupled to stress signaling: Smad engages stress Partnership between DPC4 and SMAD proteins in TGF- response factor ATF3for Id1 repression in epithelial cells. b signalling pathways. Nature 383: 832–836. Mol Cell 11: 915–926. Lee SJ, McPherron AC. 2001. Regulation of myostatin activ- Katsuno Y, Lamouille S, Derynck R. 2013. TGF-b signaling ity and muscle growth. Proc Natl Acad Sci 98: 9306–9311. and epithelial-mesenchymal transition in cancer progres- Lee KL, Lim SK, Orlov YL, Yit le Y, Yang H, Ang LT, Poel- sion. Curr Opin Oncol 25: 76–84. linger L, Lim B. 2011. Graded Nodal/Activin signaling Kawabata M, Inoue H, Hanyu A, Imamura T, Miyazono K. titrates conversion of quantitative phospho-Smad2 levels 1998. Smad proteins exist as monomers in vivo and un- into qualitative fate decisions. PLoS dergo homo- and hetero-oligomerization upon activa- Genet 7: e1002130. tion by serine/threonine kinase receptors. EMBO J 17: Leof EB, Proper JA, Goustin AS, Shipley GD, DiCorleto PE, 4056–4065. Moses HL. 1986. Induction of c-sis mRNA and activity similar to platelet-derived growth factor by transforming Kehrl JH, Wakefield LM, Roberts AB, Jakowlew S, Alvarez- growth factor b: A proposed model for indirect mito- Mon M, Derynck R, Sporn MB, Fauci AS. 1986. Produc- genesis involving autocrine activity. Proc Natl Acad Sci tion of transforming growth factor b by human T lym- 83: 2453–2457. phocytes and its potential role in the regulation of T cell growth. J Exp Med 163: 1037–1050. Levine AJ, Brivanlou AH. 2006. GDF3, a BMP inhibitor, regulates cell fate in stem cells and early embryos. Devel- Kim KK, Kugler MC, Wolters PJ, Robillard L, Galvez MG, opment 133: 209–216. Brumwell AN, Sheppard D, Chapman HA. 2006. Alveolar Levine AJ, Levine ZJ, Brivanlou AH. 2009. GDF3 is a BMP epithelial cell mesenchymal transition develops in vivo inhibitor that can activate Nodal signaling only at very during pulmonary fibrosis and is regulated by the extra- high doses. Dev Biol 325: 43–48. cellular matrix. Proc Natl Acad Sci 103: 13180–13185. Li MO, Flavell RA. 2008. TGF-b: A master of all T cell trades. Kimchi A, Wang XF, Weinberg RA, Cheifetz S, Massague´ J. Cell 134: 392–404. 1988. Absence of TGF-b receptors and growth inhibitory Lindsay ME, Dietz HC. 2011. Lessons on the pathogenesis of responses in retinoblastoma cells. Science 240: 196–199. aneurysm from heritable conditions. Nature 473: 308– Kimelman D, Kirschner M. 1987. Synergistic induction of 316. mesoderm by FGF and TGF-b and the identification of Lindsay ME, Schepers D, Bolar NA, Doyle JJ, Gallo E, Fert- an mRNA coding for FGF in the early Xenopus embryo. Bober J, Kempers MJ, Fishman EK, Chen Y,Myers L, et al. Cell 51: 869–877. 2012. Loss-of-function mutations in TGFB2 cause a syn- Kinoshita A, Saito T, Tomita H, Makita Y, Yoshida K, Gha- dromic presentation of thoracic aortic aneurysm. Nat dami M, YamadaK, Kondo S, Ikegawa S, Nishimura G, et Genet 44: 922–927. al. 2000. Domain-specific mutations in TGFB1 result in Little SC, Mullins MC. 2009. Bone morphogenetic protein Camurati-Engelmann disease. Nat Genet 26: 19–20. heterodimers assemble heteromeric type I receptor com- Koinuma D, Tsutsumi S, Kamimura N, Imamura T, Abu- plexes to pattern the dorsoventral axis. Nat Cell Biol 11: ratani H, Miyazono K. 2009a. Promoter-wide analysis of 637–643. Smad4 binding sites in human epithelial cells. Cancer Sci Liu Y,Zhang P,Li J, Kulkarni AB, Perruche S, Chen W.2008. 100: 2133–2142. A critical function for TGF-b signaling in the develop- þ þ þ Koinuma D, Tsutsumi S, Kamimura N, Taniguchi H, Miya- ment of natural CD4 CD25 Foxp3 regulatory T cells. zawa K, Sunamura M, Imamura T, Miyazono K, Abura- Nat Immunol 9: 632–640. tani H. 2009b. Chromatin immunoprecipitation on mi- Loeys BL, Chen J, Neptune ER, Judge DP, Podowski M, croarray analysis of Smad2/3 binding sites reveals roles Holm T, Meyers J, Leitch CC, Katsanis N, Sharifi N, et of ETS1 and TFAP2A in transforming growth factor b al. 2005. A syndrome of altered cardiovascular, craniofa- signaling. Mol Cell Biol 29: 172–186. cial, neurocognitive and skeletal development caused by Korn T, Bettelli E, Gao W, Awasthi A, Jager A, Strom TB, mutations in TGFBR1 or TGFBR2. Nat Genet 37: 275– Oukka M, Kuchroo VK. 2007. IL-21 initiates an alterna- 281. Loeys BL, Schwarze U, Holm T,Callewaert BL, Thomas GH, tive pathway to induce proinflammatory TH17 cells. Na- ture 448: 484–487. Pannu H, De Backer JF,Oswald GL, Symoens S, Manouv- rier S, et al. 2006. Aneurysm syndromes caused by mu- Krieglstein K, Zheng F,Unsicker K, Alzheimer C. 2011. More tations in the TGF-b receptor. N Engl J Med 355: 788– b/ than being protective: Functional roles for TGF- acti- 798. vin signaling pathways at central synapses. Trends Neuro- sci 34: 421–429. Loffredo FS, Steinhauser ML, Jay SM, Gannon J, Pancoast JR, YalamanchiP,Sinha M, Dall’Osso C, Khong D, Shad- Kuwahara F, Kai H, Tokuda K, Kai M, Takeshita A, Egashira rach JL, et al. 2013. Growth differentiation factor 11 is a K, Imaizumi T. 2002. Transforming growth factor-b circulating factor that reverses age-related cardiac hyper- function blocking prevents myocardial fibrosis and dia- trophy. Cell 153: 828–839. stolic dysfunction in pressure-overloaded rats. Circula- Lonardo E, Hermann PC, Mueller MT, Huber S, Balic A, tion 106: 130–135. Miranda-Lorenzo I, Zagorac S, Alcala S, Rodriguez-Ara- Labelle M, Begum S, Hynes RO. 2011. Direct signaling be- baolaza I, Ramirez JC, et al. 2011. Nodal/Activin signal- tween platelets and cancer cells induces an epithelial- ing drives self-renewal and tumorigenicity of pancreatic mesenchymal-like transition and promotes metastasis. cancer stem cells and provides a target for combined drug Cancer Cell 20: 576–590. therapy. Cell Stem Cell 9: 433–446.

20 Cite this article as Cold Spring Harb Perspect Biol 2016;8:a021873 Downloaded from http://cshperspectives.cshlp.org/ on September 24, 2021 - Published by Cold Spring Harbor Laboratory Press

Roles of TGF-b in Cell and Tissue Physiology

Lund LR, Riccio A, Andreasen PA, Nielsen LS, Kristensen P, Morikawa M, Koinuma D, Miyazono K, Heldin CH. 2013. Laiho M, Saksela O, Blasi F, Dano K. 1987. Transforming Genome-wide mechanisms of Smad binding. Oncogene growth factor-b is a strong and fast acting positive regu- 32: 1609–1615. lator of the level of type-1 plasminogen activator inhib- Morinaga C, Saito D, Nakamura S, Sasaki T, Asakawa S, itor mRNA in WI-38 human lung fibroblasts. EMBO J 6: Shimizu N, Mitani H, Furutani-Seiki M, Tanaka M, Kon- 1281–1286. doh H. 2007. The hotei mutation of medaka in the anti- Maeda S, Hayashi M, Komiya S, Imamura T, Miyazono K. Mu¨llerian hormone receptor causes the dysregulation of 2004. Endogenous TGF-b signaling suppresses matura- germ cell and sexual development. Proc Natl Acad Sci 104: tion of osteoblastic mesenchymal cells. EMBO J 23: 552– 9691–9696. 563. Mullen AC, Orlando DA, Newman JJ, Loven J, Kumar RM, Mani SA, Guo W, Liao MJ, Eaton EN, Ayyanan A, Zhou AY, Bilodeau S, Reddy J, Guenther MG, DeKoter RP, Young Brooks M, Reinhard F, Zhang CC, Shipitsin M, et al. RA. 2011. Master transcription factors determine cell- 2008. The epithelial-mesenchymal transition generates type-specific responses to TGF-b signaling. Cell 147: cells with properties of stem cells. Cell 133: 704–715. 565–576. Markowitz S, Wang J, Myeroff L, Parsons R, Sun L, Lutter- Munger JS, Huang X, Kawakatsu H, Griffiths MJ, Dalton SL, baugh J, Fan RS, Zborowska E, Kinzler KW,Vogelstein B, Wu J, Pittet JF, Kaminski N, Garat C, Matthay MA, et al. et al. 1995. Inactivation of the type II TGF-b receptor in 1999. The integrin avb6 binds and activates latent colon cancer cells with microsatellite instability. Science TGFb1: A mechanism for regulating pulmonary inflam- 268: 1336–1338. mation and fibrosis. Cell 96: 319–328. Massague´ J, Cheifetz S, Endo T, Nadal-Ginard B. 1986. Type Mustoe TA,Pierce GF,Thomason A, Gramates P,Sporn MB, b transforming growth factor is an inhibitor of myogenic Deuel TF.1987. Accelerated healing of incisional wounds differentiation. Proc Natl Acad Sci 83: 8206–8210. in rats induced by transforming growth factor-b. Science 237: 1333–1336. Mathews LS, Vale WW. 1991. Expression cloning of an acti- vin receptor, a predicted transmembrane serine kinase. Naka K, Hoshii T, Muraguchi T, Tadokoro Y, Ooshio T, Cell 65: 973–982. Kondo Y, Nakao S, Motoyama N, Hirao A. 2010. TGF- b-FOXO signalling maintains leukaemia-initiating cells Matsuyama S, Iwadate M, Kondo M, Saitoh M, Hanyu A, in chronic myeloid leukaemia. Nature 463: 676–680. Shimizu K, Aburatani H, Mishima HK, Imamura T, Miyazono K, et al. 2003. SB-431542 and Gleevec inhibit Nakao A, Afrakhte M, Moren A, Nakayama T, Christian JL, transforming growth factor-b-induced proliferation of Heuchel R, Itoh S, Kawabata M, Heldin NE, Heldin CH, human osteosarcoma cells. Cancer Res 63: 7791–7798. et al. 1997. Identification of Smad7, a TGFb-inducible antagonist of TGF-b signalling. Nature 389: 631–635. Mazerbourg S, Klein C, Roh J, Kaivo-Oja N, Mottershead DG, Korchynskyi O, Ritvos O, Hsueh AJ. 2004. Growth Nakao A, Fujii M, Matsumura R, Kumano K, Saito Y,Miya- differentiation factor-9 signaling is mediated by the type I zono K, Iwamoto I. 1999. Transient gene transfer and expression of Smad7 prevents bleomycin-induced lung receptor, activin receptor-like kinase 5. Mol Endocrinol fibrosis in mice. J Clin Invest 104: 5–11. 18: 653–665. Neptune ER, Frischmeyer PA, Arking DE, Myers L, Bunton McPherron AC, Lee SJ. 1997. Double muscling in cattle due TE, Gayraud B, Ramirez F, Sakai LY, Dietz HC. 2003. to mutations in the myostatin gene. Proc Natl Acad Sci 94: Dysregulation of TGF-b activation contributes to path- 12457–12461. ogenesis in Marfan syndrome. Nat Genet 33: 407–411. McPherron AC, Lawler AM, Lee SJ. 1997. Regulation of Ng F,Boucher S, Koh S, Sastry KS, Chase L, Lakshmipathy U, skeletal muscle mass in mice by a new TGF-b superfamily Choong C, Yang Z, Vemuri MC, Rao MS, et al. 2008. member. Nature 387: 83–90. PDGF,TGF-b, and FGF signaling is important for differ- Meynard D, Kautz L, Darnaud V,Canonne-Hergaux F,Cop- entiation and growth of mesenchymal stem cells (MSCs): pin H, Roth MP. 2009. Lack of the bone morphogenetic Transcriptional profiling can identify markers and sig- protein BMP6 induces massive iron overload. Nat Genet naling pathways important in differentiation of MSCs 41: 478–481. into adipogenic, chondrogenic, and osteogenic lineages. Mi LZ, Brown CT, Gao Y, Tian Y, Le VQ, Walz T, Springer Blood 112: 295–307. TA. 2015. Structure of bone morphogenetic protein 9 Niehrs C, Pollet N. 1999. Synexpression groups in eukary- procomplex. Proc Natl Acad Sci 112: 3710–3715. otes. Nature 402: 483–487. Mizuguchi T, Collod-Beroud G, Akiyama T, Abifadel M, Ogawa K, Saito A, Matsui H, Suzuki H, Ohtsuka S, Shimo- Harada N, Morisaki T, Allard D, Varret M, Claustres M, sato D, Morishita Y, Watabe T, Niwa H, Miyazono K. Morisaki H, et al. 2004. Heterozygous TGFBR2 muta- 2007. Activin-nodal signaling is involved in propagation tions in Marfan syndrome. Nat Genet 36: 855–860. of mouse embryonic stem cells. J Cell Sci 120: 55–65. Moore RK, Otsuka F, Shimasaki S. 2003. Molecular basis of Oh SP, Yeo CY, Lee Y, Schrewe H, Whitman M, Li E. 2002. bone morphogenetic protein-15 signaling in granulosa Activin type IIA and IIB receptors mediate Gdf11 signal- cells. J Biol Chem 278: 304–310. ing in axial vertebral patterning. Genes Dev 16: 2749– Morikawa M, Koinuma D, Tsutsumi S, Vasilaki E, Kanki Y, 2754. Heldin CH, Aburatani H, Miyazono K. 2011. ChIP-seq Ohta M, Greenberger JS, Anklesaria P,Bassols A, Massague´ reveals cell type-specific binding patterns of BMP-specif- J. 1987. Two forms of transforming growth factor-b dis- ic Smads and a novel binding motif. Nucleic Acids Res 39: tinguished by multipotential haematopoietic progenitor 8712–8727. cells. Nature 329: 539–541.

Cite this article as Cold Spring Harb Perspect Biol 2016;8:a021873 21 Downloaded from http://cshperspectives.cshlp.org/ on September 24, 2021 - Published by Cold Spring Harbor Laboratory Press

M. Morikawa et al.

Oshimori N, Fuchs E. 2012. The harmonies played by TGF- Roberts AB, Anzano MA, Meyers CA, Wideman J, Blacher R, b in stem cell biology. Cell Stem Cell 11: 751–764. Pan YC, Stein S, Lehrman SR, Smith JM, Lamb LC, et al. Palumbo-Zerr K, Zerr P, Distler A, Fliehr J, Mancuso R, 1983. Purification and properties of a type b transform- Huang J, Mielenz D, TomcikM, Furnrohr BG, Scholtysek ing growth factor from bovine kidney. Biochemistry 22: C, et al. 2015. Orphan nuclear receptor NR4A1 regulates 5692–5698. transforming growth factor-b signaling and fibrosis. Nat Roberts AB, Anzano MA, Wakefield LM, Roche NS, Stern Med 21: 150–158. DF,Sporn MB. 1985. Type b transforming growth factor: Pardali E, Goumans MJ, ten Dijke P. 2010. Signaling by A bifunctional regulator of cellular growth. Proc Natl members of the TGF-b family in vascular morphogenesis Acad Sci 82: 119–123. and disease. Trends Cell Biol 20: 556–567. Roberts AB, Sporn MB, Assoian RK, Smith JM, Roche NS, Park SM, Gaur AB, Lengyel E, Peter ME. 2008. The miR-200 Wakefield LM, Heine UI, Liotta LA, Falanga V,Kehrl JH, family determines the epithelial phenotype of cancer cells et al. 1986. Transforming growth factor type b: Rapid by targeting the E-cadherin repressors ZEB1 and ZEB2. induction of fibrosis and angiogenesis in vivo and stim- Genes Dev 22: 894–907. ulation of collagen formation in vitro. Proc Natl Acad Sci Peng J, Li Q, Wigglesworth K, Rangarajan A, Kattamuri C, 83: 4167–4171. Peterson RT,Eppig JJ, Thompson TB, Matzuk MM. 2013. Rosa F, Roberts AB, Danielpour D, Dart LL, Sporn MB, Growth differentiation factor 9:bone morphogenetic Dawid IB. 1988. Mesoderm induction in amphibians: protein 15 heterodimers are potent regulators of ovarian The role of TGF-b2-like factors. Science 239: 783–785. functions. Proc Natl Acad Sci 10: E776–E785. Russell WE, Coffey RJ Jr, Ouellette AJ, Moses HL. 1988. Type Penuelas S, Anido J, Prieto-Sanchez RM, Folch G, Barba I, b transforming growth factor reversibly inhibits the early Cuartas I, Garcia-Dorado D, Poca MA, Sahuquillo J, proliferative response to partial hepatectomy in the rat. Baselga J, et al. 2009. TGF-b increases glioma-initiating Proc Natl Acad Sci 85: 5126–5130. cell self-renewal through the induction of LIF in human Saito T,Kinoshita A, Yoshiura K, Makita Y,Wakui K, Honke glioblastoma. Cancer Cell 15: 315–327. K, Niikawa N, Taniguchi N. 2001. Domain-specific mu- Pickup M, Novitskiy S, Moses HL. 2013. The roles of TGFb tations of a transforming growth factor (TGF)-b1 la- in the tumour microenvironment. Nat Rev Cancer 13: tency-associated peptide cause Camurati–Engelmann 788–799. disease because of the formation of a constitutively active Pietenpol JA, Stein RW,Moran E, Yaciuk P,Schlegel R, Lyons form of TGF-b1. J Biol Chem 276: 11469–11472. RM, Pittelkow MR, Munger K, Howley PM, Moses HL. Sakaki-Yumoto M, Katsuno Y, Derynck R. 2013. TGF-b 1990. TGF-b1 inhibition of c-myc transcription and family signaling in stem cells. Biochim Biophys Acta growth in keratinocytes is abrogated by viral transform- 1830: 2280–2296. ing proteins with pRB binding domains. Cell 61: 777– Sanford LP,Ormsby I, Gittenberger-de Groot AC, Sariola H, 785. Friedman R, Boivin GP,Cardell EL, Doetschman T.1997. Polyak K, Kato JY, Solomon MJ, Sherr CJ, Massague´ J, Rob- TGFb2 knockout mice have multiple developmental de- erts JM, Koff A. 1994. p27Kip1, a cyclin-Cdk inhibitor, fects that are non-overlapping with other TGFb knock- links transforming growth factor-b and contact inhibi- out phenotypes. Development 124: 2659–2670. tion to cell cycle arrest. Genes Dev 8: 9–22. Sartori R, Schirwis E, Blaauw B, Bortolanza S, Zhao J, Enzo Postlethwaite AE, Keski-Oja J, Moses HL, Kang AH. 1987. E, Stantzou A, Mouisel E, Toniolo L, Ferry A, et al. 2013. Stimulation of the chemotactic migration of human fi- BMP signaling controls muscle mass. Nat Genet 45: broblasts by transforming growth factor b. J Exp Med 1309–1318. 165: 251–256. Sato M, Muragaki Y,Saika S, Roberts AB, Ooshima A. 2003. Qin H, Chan MW, Liyanarachchi S, Balch C, Potter D, Sou- Targeted disruption of TGF-b1/Smad3 signaling pro- riraj IJ, Cheng AS, Agosto-Perez FJ, Nikonova EV,YanPS, tects against renal tubulointerstitial fibrosis induced by et al. 2009. An integrative ChIP-chip and gene expression unilateral ureteral obstruction. J Clin Invest 112: 1486– profiling to model SMAD regulatory modules. BMC Syst 1494. Biol 3: 73. Savage C, Das P,Finelli AL, Townsend SR, Sun CY,Baird SE, Raftery LA, Twombly V, Wharton K, Gelbart WM. 1995. Padgett RW. 1996. Caenorhabditis elegans genes sma-2, Genetic screens to identify elements of the decapentaple- sma-3, and sma-4 define a conserved family of trans- gic signaling pathway in Drosophila. Genetics 139: 241– forming growth factor b pathway components. Proc 254. Natl Acad Sci 93: 790–794. Ristow HJ. 1986. BSC-1 growth inhibitor/type b transform- Schlunegger MP, Gru¨tter MG. 1992. An unusual feature ing growth factor is a strong inhibitor of thymocyte pro- revealed by the crystal structure at 2.2 A˚ resolution of liferation. Proc Natl Acad Sci 83: 5531–5533. human transforming growth factor-b2. Nature 358: Roberts AB, Lamb LC, Newton DL, Sporn MB, De Larco JE, 430–434. Todaro GJ. 1980. Transforming growth factors: Isolation Schubert D, Kimura H, LaCorbiere M, Vaughan J, Karr D, of polypeptides from virally and chemically transformed Fischer WH. 1990. Activin is a nerve cell survival mole- cells by acid/ethanol extraction. Proc Natl Acad Sci 77: cule. Nature 344: 868–870. 3494–3498. Sekelsky JJ, Newfeld SJ, Raftery LA, Chartoff EH, Gelbart Roberts AB, Anzano MA, Lamb LC, Smith JM, Sporn MB. WM. 1995. Genetic characterization and cloning of 1981. New class of transforming growth factors potenti- mothers against dpp, a gene required for decapentaplegic ated byepidermal growth factor: Isolation from non-neo- function in Drosophila melanogaster. Genetics 139: 1347– plastic tissues. Proc Natl Acad Sci 78: 5339–5343. 1358.

22 Cite this article as Cold Spring Harb Perspect Biol 2016;8:a021873 Downloaded from http://cshperspectives.cshlp.org/ on September 24, 2021 - Published by Cold Spring Harbor Laboratory Press

Roles of TGF-b in Cell and Tissue Physiology

Sengle G, Charbonneau NL, Ono RN, Sasaki T, Alvarez J, Sporn MB, Roberts AB, Shull JH, Smith JM, Ward JM, So- Keene DR, Bachinger HP, Sakai LY. 2008. Targeting of dek J. 1983. Polypeptide transforming growth factors iso- bone morphogenetic protein growth factor complexes lated from bovine sources and used for wound healing in to fibrillin. J Biol Chem 283: 13874–13888. vivo. Science 219: 1329–1331. Seyedin SM, Thomas TC, Thompson AY, Rosen DM, Piez Suzuki A, Kaneko E, Maeda J, Ueno N. 1997. Mesoderm KA. 1985. Purification and characterization of two carti- induction by BMP-4 and -7 heterodimers. Biochem Bio- lage-inducing factors from bovine demineralized bone. phys Res Commun 232: 153–156. Proc Natl Acad Sci 82: 2267–2271. Tabibzadeh S, Hemmati-Brivanlou A. 2006. Lefty at the Shah M, Foreman DM, Ferguson MW. 1995. Neutralisation crossroads of “stemness” and differentiative events. of TGF-b1 and TGF-b2 or exogenous addition of TGF- Stem Cells 24: 1998–2006. b3 to cutaneous rat wounds reduces scarring. J Cell Sci Taipale J, Miyazono K, Heldin CH, Keski-Oja J. 1994. Latent 108: 985–1002. transforming growth factor-b1 associates to fibroblast Shi M, Zhu J, Wang R, Chen X, Mi L, Walz T, Springer TA. extracellular matrix via latent TGF-b binding protein. 2011. Latent TGF-b structure and activation. Nature 474: J Cell Biol 124: 171–181. 343–349. Tang B, Vu M, Booker T, Santner SJ, Miller FR, Anver MR, Shipley GD, Childs CB, Volkenant ME, Moses HL. 1984. Wakefield LM. 2003. TGF-b switches from tumor sup- Differential effects of epidermal growth factor, trans- pressor to prometastatic factor in a model of breast can- forming growth factor, and insulin on DNA and protein cer progression. J Clin Invest 112: 1116–1124. synthesis and morphology in serum-free cultures of Tang B, Yoo N, Vu M, Mamura M, Nam JS, Ooshima A, Du AKR-2B cells. Cancer Res 44: 710–716. Z, Desprez PY, Anver MR, Michalowska AM, et al. 2007. Shipley GD, Pittelkow MR, Wille JJ Jr, Scott RE, Moses HL. Transforming growth factor-b can suppress tumorigen- 1986. Reversible inhibition of normal human prokerati- esis through effects on the putative cancer stem or early nocyte proliferation by type b transforming growth fac- progenitor cell and committed progeny in a breast cancer tor-growth inhibitor in serum-free medium. Cancer Res xenograft model. Cancer Res 67: 8643–8652. 46: 2068–2071. TangY,Wu X, Lei W,Pang L, WanC, Shi Z, Zhao L, Nagy TR, Shirakihara T,Horiguchi K, Miyazawa K, Ehata S, Shibata T, Peng X, Hu J, et al. 2009. TGF-b1-induced migration of Morita I, Miyazono K, Saitoh M. 2011. TGF-b regulates bone mesenchymal stem cells couples bone resorption isoform switching of FGF receptors and epithelial-mes- with formation. Nat Med 15: 757–765. enchymal transition. EMBO J 30: 783–795. Theunissen TW,Powell BE, Wang H, Mitalipova M, Faddah Shull MM, Ormsby I, Kier AB, Pawlowski S, Diebold RJ, Yin DA, Reddy J, Fan ZP,Maetzel D, Ganz K, Shi L, et al. 2014. M, Allen R, Sidman C, Proetzel G, Calvin D, et al. 1992. Systematic identification of culture conditions for induc- Targeted disruption of the mouse transforming growth tion and maintenance of naive human pluripotency. Cell factor-b1 gene results in multifocal inflammatory dis- Stem Cell 15: 471–487. ease. Nature 359: 693–699. Trompouki E, Bowman TV, Lawton LN, Fan ZP, Wu DC, Siegel PM, Massague´ J. 2003. Cytostatic and apoptotic ac- DiBiase A, Martin CS, Cech JN, Sessa AK, Leblanc JL, et tions of TGF-b in homeostasis and cancer. Nat Rev Can- al. 2011. Lineage regulators direct BMP and Wnt path- cer 3: 807–821. ways to cell-specific programs during differentiation and Siegel PM, Massague´ J. 2008. Growth control by TGF-b: regeneration. Cell 147: 577–589. Mechanisms controlling cell cycle progression and apo- Tucker RF,Shipley GD, Moses HL, Holley RW.1984. Growth ptosis. In The TGF-b family (eds. Derynck R, Miyazono inhibitor from BSC-1 cells closely related to platelet type K), pp. 333–362. Cold Spring Harbor Laboratory Press, b transforming growth factor. Science 226: 705–707. Cold Spring Harbor, NY. Vallier L, Alexander M, Pedersen RA. 2005. Activin/nodal Sievers F, Wilm A, Dineen D, Gibson TJ, Karplus K, Li W, and FGF pathways cooperate to maintain pluripotency of Lopez R, McWilliam H, Remmert M, Soding J, et al. 2011. human embryonic stem cells. J Cell Sci 118: 4495–4509. Fast, scalable generation of high-quality protein multiple van de Laar IM, Oldenburg RA, Pals G, Roos-Hesselink JW, sequence alignments using Clustal Omega. Mol Syst Biol de Graaf BM, Verhagen JM, Hoedemaekers YM, Willem- 7: 539. sen R, Severijnen LA, Venselaar H, et al. 2011. Mutations Silberstein GB, Daniel CW. 1987. Reversible inhibition of in SMAD3 cause a syndromic form of aortic aneurysms mammary gland growth by transforming growth fac- and dissections with early-onset osteoarthritis. Nat Genet tor-b. Science 237: 291–293. 43: 121–126. Sime PJ, Xing Z, Graham FL, Csaky KG, Gauldie J. 1997. Veldhoen M, Hocking RJ, Atkins CJ, Locksley RM, Stock- Adenovector-mediated gene transfer of active transform- inger B. 2006. TGFb in the context of an inflammatory ing growth factor-b1 induces prolonged severe fibrosis in cytokine milieu supports de novo differentiation of IL- rat lung. J Clin Invest 100: 768–776. 17-producing T cells. Immunity 24: 179–189. Sinha M, Jang YC, Oh J, Khong D, Wu EY, Manohar R, Vogel T, Ahrens S, Buttner N, Krieglstein K. 2010. Trans- Miller C, Regalado SG, Loffredo FS, Pancoast JR, et al. forming growth factor b promotes neuronal cell fate of 2014. Restoring systemic GDF11 levels reverses age-relat- mouse cortical and hippocampal progenitors in vitro and ed dysfunction in mouse skeletal muscle. Science 344: in vivo: Identification of Nedd9 as an essential signaling 649–652. component. Cereb Cortex 20: 661–671. Smith WC, Harland RM. 1992. Expression cloning of nog- Vogelstein B, Papadopoulos N, Velculescu VE, Zhou S, Diaz gin, a new dorsalizing factor localized to the Spemann LA Jr, Kinzler KW. 2013. Cancer genome landscapes. organizer in Xenopus embryos. Cell 70: 829–840. Science 339: 1546–1558.

Cite this article as Cold Spring Harb Perspect Biol 2016;8:a021873 23 Downloaded from http://cshperspectives.cshlp.org/ on September 24, 2021 - Published by Cold Spring Harbor Laboratory Press

M. Morikawa et al.

Wahl SM, Hunt DA, Wakefield LM, McCartney-Francis N, TGFb1 activation in vivo recapitulates the phenotype Wahl LM, Roberts AB, Sporn MB. 1987. Transforming of TGFb1-null mice. J Cell Biol 176: 787–793. growth factor type b induces monocyte chemotaxis and Yoshinaga K, Obata H, Jurukovski V, Mazzieri R, Chen Y, growth factor production. Proc Natl Acad Sci 84: 5788– Zilberberg L, Huso D, Melamed J, Prijatelj P,Todorovic V, 5792. et al. 2008. Perturbation of transforming growth factor Wilson CA, di Clemente N, Ehrenfels C, Pepinsky RB, Josso (TGF)-b1 association with latent TGF-b binding protein N, Vigier B, Cate RL. 1993. Mu¨llerian inhibiting sub- yields inflammation and tumors. Proc Natl Acad Sci 105: stance requires its N-terminal domain for maintenance 18758–18763. of biological activity, a novel finding within the trans- Yu J, Shao LE, Lemas V, Yu AL, Vaughan J, Rivier J, Vale W. forming growth factor-b superfamily. Mol Endocrinol 7: 1987. Importance of FSH-releasing protein and inhibin 247–257. in erythrodifferentiation. Nature 330: 765–767. Wolfman NM, McPherron AC, Pappano WN, Davies MV, Zeisberg EM, Tarnavski O, Zeisberg M, Dorfman AL, Song K, Tomkinson KN, Wright JF, Zhao L, Sebald SM, McMullen JR, Gustafsson E, Chandraker A, Yuan X, Pu Greenspan DS, et al. 2003. Activation of latent myostatin WT, Roberts AB, et al. 2007. Endothelial-to-mesenchy- by the BMP-1/tolloid family of metalloproteinases. Proc mal transition contributes to cardiac fibrosis. Nat Med Natl Acad Sci 100: 15842–15846. 13: 952–961. Wu SP,Theodorescu D, Kerbel RS, Willson JK, Mulder KM, Zhang Y, Feng X, We R, Derynck R. 1996. Receptor-associ- Humphrey LE, Brattain MG. 1992. TGF-b1 is an auto- ated Mad homologues synergize as effectors of the TGF-b crine-negative growth regulator of human colon carcino- response. Nature 383: 168–172. ma FET cells in vivo as revealed by transfection of an Zhao J, Shi W, Wang YL, Chen H, Bringas P Jr, Datto MB, antisense expression vector. J Cell Biol 116: 187–196. Frederick JP, Wang XF, Warburton D. 2002. Smad3 defi- Wu M, Melichian DS, Chang E, Warner-Blankenship M, ciency attenuates bleomycin-induced pulmonary fibrosis Ghosh AK, Varga J. 2009. Rosiglitazone abrogates bleo- in mice. Am J Physiol Lung Cell Mol Physiol 282: L585– mycin-induced scleroderma and blocks profibrotic L593. responses through peroxisome proliferator-activated re- Zhou X, Wang JL, Lu J, Song Y, Kwak KS, Jiao Q, Rosenfeld ceptor-g. Am J Pathol 174: 519–533. R, Chen Q, Boone T,Simonet WS, et al. 2010. Reversal of Yang Z, Mu Z, Dabovic B, Jurukovski V,Yu D, Sung J, Xiong cancer cachexia and muscle wasting by ActRIIB antago- X, Munger JS. 2007. Absence of integrin-mediated nism leads to prolonged survival. Cell 142: 531–543.

24 Cite this article as Cold Spring Harb Perspect Biol 2016;8:a021873 Downloaded from http://cshperspectives.cshlp.org/ on September 24, 2021 - Published by Cold Spring Harbor Laboratory Press

TGF-β and the TGF-β Family: Context-Dependent Roles in Cell and Tissue Physiology

Masato Morikawa, Rik Derynck and Kohei Miyazono

Cold Spring Harb Perspect Biol 2016; doi: 10.1101/cshperspect.a021873

Subject Collection The Biology of the TGF-β Family

TGF-β Family Signaling in Early Vertebrate TGF-β Family Signaling in Mesenchymal Development Differentiation Joseph Zinski, Benjamin Tajer and Mary C. Mullins Ingo Grafe, Stefanie Alexander, Jonathan R. Peterson, et al. Bone Morphogenetic Protein−Based Therapeutic TGF-β1 Signaling and Tissue Fibrosis Approaches Kevin K. Kim, Dean Sheppard and Harold A. Jonathan W. Lowery and Vicki Rosen Chapman TGF-β Family Signaling in Ductal Differentiation Bone Morphogenetic Proteins in Vascular and Branching Morphogenesis Homeostasis and Disease Kaoru Kahata, Varun Maturi and Aristidis Marie-José Goumans, An Zwijsen, Peter ten Dijke, Moustakas et al. TGF-β Signaling in Control of Cardiovascular TGF-β Family Signaling in Epithelial Function Differentiation and Epithelial−Mesenchymal Marie-José Goumans and Peter ten Dijke Transition Kaoru Kahata, Mahsa Shahidi Dadras and Aristidis Moustakas TGF-β Family Signaling in Tumor Suppression TGF-β Family Signaling in Connective Tissue and and Cancer Progression Skeletal Diseases Joan Seoane and Roger R. Gomis Elena Gallo MacFarlane, Julia Haupt, Harry C. Dietz, et al. Targeting TGF-β Signaling for Therapeutic Gain The TGF-β Family in the Reproductive Tract Rosemary J. Akhurst Diana Monsivais, Martin M. Matzuk and Stephanie A. Pangas Regulation of Hematopoiesis and Hematological TGF-β Family Signaling in Drosophila Disease by TGF- β Family Signaling Molecules Ambuj Upadhyay, Lindsay Moss-Taylor, Myung-Jun Kazuhito Naka and Atsushi Hirao Kim, et al.

For additional articles in this collection, see http://cshperspectives.cshlp.org/cgi/collection/

Copyright © 2016 Cold Spring Harbor Laboratory Press; all rights reserved Downloaded from http://cshperspectives.cshlp.org/ on September 24, 2021 - Published by Cold Spring Harbor Laboratory Press

TGF-β Family Signaling in Neural and Neuronal Signaling Cross Talk between TGF-β/Smad and Differentiation, Development, and Function Other Signaling Pathways Emily A. Meyers and John A. Kessler Kunxin Luo

For additional articles in this collection, see http://cshperspectives.cshlp.org/cgi/collection/

Copyright © 2016 Cold Spring Harbor Laboratory Press; all rights reserved