Review
TGF-β signalling pathways in early Xenopus development

https://doi.org/10.1016/S0959-437X(00)00229-XGet rights and content

Abstract

Many different ligands of the TGF-β superfamily signal in the early Xenopus embryo and are required for the specification and patterning of the three germ layers as well as for gastrulation. Recent advances in the field are helping us understand how ligand activity is regulated both spatially and temporally, the mechanism by which the signals are transduced to the nucleus and how essentially the same signalling pathway can activate completely different sets of genes in different regions of the embryo.

Introduction

The members of the TGF-β superfamily that are important in early Xenopus embryos belong to two subfamilies: those functionally related to Activin, which include members of the Nodal family (Xnr1–6), Derrière, Vg-1, and Activin itself and members of the bone morphogenetic protein (BMP) subfamily, in particular BMP2, 4 and 7. The Activin-related ligands are required for mesoderm and endoderm specification and patterning, for promoting gastrulation movements and for establishing left–right asymmetry 1••, 2, 3. The BMPs control the fundamental decision between formation of neural versus other ectodermal cell fates and are also involved in patterning ventral and lateral mesoderm (reviewed in 4). Moreover, TGF-β superfamily members have the ability to act as morphogens, with different doses of ligand turning on particular combinations of downstream genes, and thus instructing different groups of cells to adopt distinct fates 5, 6, 7. Recent advances have now provided some insights into how gradients of active ligand might be established in the embryo. Furthermore, many components of the signalling pathways have now been identified in Xenopus, allowing us to trace the pathways from receptors to nucleus. With this information, it is becoming clear how the same signalling pathway can be involved in distinct biological responses.

Section snippets

Regulation of ligand activity or how to makegradients

Gradients of active ligands have long been thought to underlie the mechanism that establishes position in the developing embryo. Conceptually, gradients of active ligands can be established by a variety of mechanisms. The simplest model involves the production of the ligand at a local source from which it diffuses away to a more distant sink: high ligand concentrations are achieved close to the source and low concentrations at a distance. Alternatively, the ligand may be produced at an equal

Transducing the signals to the nucleus

TGF-β superfamily members function by signalling to the nucleus and regulating the transcription of target genes. A major breakthrough has been the identification of many components of the TGF-β signalling pathways, making it now possible to trace the signalling pathways from receptors to the nucleus, and to identify the critical points at which the pathway can be regulated.

Specificity of the responses

How can the same signalling pathway activate different genes in different regions of the embryo? Recent work 68••, 69, 70••, 71• has identified important determinants of specificity in the form of transcription factors that are required to recruit activated Smad complexes to specific promoter elements.

Smads bind DNA either very weakly (Smad1, 3, 4) or not at all (Smad2), and have a low level of sequence specificity, recognizing a 4 base-pair motif (GTCT) which will occur on average once every

Conclusions and future perspectives

I have focused here on the molecular details of the TGF-β signalling pathways in Xenopus embryos. Many components are known, although given the complexity of other signalling pathways, undoubtedly there are more to be discovered. The most striking feature of these pathways is that a host of different ligands activate a common short signal transduction pathway to elicit a complex array of transcriptional responses, which ultimately specify and pattern the germ layers of the embryo. The pathways

Update

Since completion of this manuscript, a paper from the Whitman group 75• has reported biochemical evidence for the role of the EGF-CFC family member Cripto as a co-receptor for Nodal. Cripto interacts with the type I receptor ALK4 which permits Nodal binding to the receptor complex, resulting in phosphorylation of Smad2. These experiments were performed in Xenopus embryos, but with the mouse family members. It therefore still remains to be demonstrated that FRL-1 plays this role in Xenopus.

In

Acknowledgements

I am very grateful to Mike Howell for Fig. 2 and thank him, Karolien De Bosscher, Les Dale, Gareth Inman, Helen McNeill, Francisco Nicolás, Becky Randall, Jim Smith and Astrid Steinmair for useful discussions and helpful comments on the manuscript. I thank Malcolm Whitman for generously sending a manuscript prior to publication.

References and recommended reading

Papers of particular interest, published within the annual period of review, have been highlighted as:

  • • of special interest

  • •• of outstanding interest

References (76)

  • S Piccolo et al.

    Cleavage of Chordin by Xolloid metalloprotease suggests a role for proteolytic processing in the regulation of Spemann organizer activity

    Cell

    (1997)
  • J.M Graff et al.

    Studies with a Xenopus BMP receptor suggest that ventral mesoderm-inducing signals override dorsal signals in vivo

    Cell

    (1994)
  • A Suzuki et al.

    Regulation of epidermal induction by BMP2 and BMP7 signaling

    Dev Biol

    (1997)
  • M.M Shen et al.

    The EGF-CFC gene family in vertebrate development

    Trends Genet

    (2000)
  • K Gritsman et al.

    The EGF-CFC protein one-eyed pinhead is essential for nodal signaling

    Cell

    (1999)
  • N Kinoshita et al.

    The identification of two novel ligands of the FGF receptor by a yeast screening method and their activity in Xenopus development

    Cell

    (1995)
  • J.M Graff et al.

    Xenopus Mad proteins transduce distinct subsets of signals for the TGF-β superfamily

    Cell

    (1996)
  • A Suzuki et al.

    Smad5 induces ventral fates in Xenopus embryo

    Dev Biol

    (1997)
  • M Howell et al.

    Xenopus Smad3 is specifically expressed in the chordoneural hinge, notochord and in the endocardium of the developing heart

    Mech Dev

    (2001)
  • T Tsukazaki et al.

    SARA, a FYVE domain protein that recruits Smad2 to the TGFβ receptor

    Cell

    (1998)
  • M Howell et al.

    Xenopus Smad4β is the co-Smad component of developmentally-regulated transcription factor complexes responsible for induction of early mesodermal genes

    Dev Biol

    (1999)
  • N Masuyama et al.

    Identification of two Smad4 proteins in Xenopus. Their common and distinct properties

    J Biol Chem

    (1999)
  • R Casellas et al.

    Xenopus Smad7 inhibits both the Activin and BMP pathways and acts as a neural inducer

    Dev Biol

    (1998)
  • A Bhushan et al.

    Smad7 inhibits mesoderm formation and promotes neural cell fate in Xenopus embryos

    Dev Biol

    (1998)
  • T Nakayama et al.

    Dissection of inhibitory Smad proteins: both N- and C-terminal domains are necessary for full activities of Xenopus Smad6 and Smad7

    Mech Dev

    (2001)
  • P Kavsak et al.

    Smad7 binds to Smurf2 to form an E3 ubiquitin ligase that targets the TGF β receptor for degradation

    Mol Cell

    (2000)
  • T Ebisawa et al.

    Smurf1 interacts with transforming growth factor-β type I receptor through Smad7 and induces receptor degradation

    J Biol Chem

    (2001)
  • T Kurata et al.

    Visualization of endogenous BMP signaling during Xenopus development

    Differentiation

    (2001)
  • A Hata et al.

    OAZ uses distinct DNA- and protein-binding zinc fingers in separate BMP-Smad and Olf signaling pathways

    Cell

    (2000)
  • C Yeo et al.

    Nodal signals to Smads through Cripto-dependent and Cripto-independent mechanisms

    Mol Cell

    (2001)
  • S Osada et al.

    Activin/Nodal responsiveness and asymmetric expression of a Xenopus nodal-related gene converge on a FAST-regulated module in intron 1

    Development

    (2000)
  • Smith JC, Howard JE: Mesoderm-inducing factors and the control of gastrulation. Development 1992,...
  • R Harland et al.

    Formation and function of Spemann's organizer

    Annu Rev Cell Dev Biol

    (1997)
  • P.A Wilson et al.

    Concentration-dependent patterning of the Xenopus ectoderm by BMP4 and its signal transducer Smad1

    Development

    (1997)
  • J.B Green et al.

    Graded changes in dose of a Xenopus Activin A homologue elicit stepwise transitions in embryonic cell fate

    Nature

    (1990)
  • J.B Gurdon et al.

    Activin signalling and response to a morphogen gradient

    Nature

    (1994)
  • C Niederlander et al.

    Arkadia enhances nodal-related signalling to induce mesendoderm

    Nature

    (2001)
  • R Dosch et al.

    Bmp-4 acts as a morphogen in dorsoventral mesoderm patterning in Xenopus

    Development

    (1997)
  • Cited by (77)

    • miR-430a regulates the development of left–right asymmetry by targeting sqt in the teleost

      2020, Gene
      Citation Excerpt :

      Most vertebrate body display bilateral symmetry, however, the visceral organs show left-right (L-R) asymmetric trait, such as heart and liver (Palmer 2004, Bisgrove et al., 2015). It has been reported that the development of endoderm and the establishment of L-R asymmetric pattern was influenced by nodal-related genes, which were expressed in left lateral plate mesoderm (Rebagliati et al., 1998a,b, Hill 2001, Long et al., 2003). There are three homologues of nodal in zebrafish: cyclops (cyc), squint (sqt) and southpaw (spaw) (Erter et al., 1998, Rebagliati et al., 1998a,b, Long et al., 2003).

    • Foxh1 Occupies cis-Regulatory Modules Prior to Dynamic Transcription Factor Interactions Controlling the Mesendoderm Gene Program

      2017, Developmental Cell
      Citation Excerpt :

      Vegt controls the zygotic transcription of the Xenopus Nodal ligands (Kofron et al., 1999; Xanthos et al., 2001), initiating Nodal signaling and mesendoderm development (reviewed in Whitman, 2001). The Nodal ligands activate the phosphorylation of R-Smad2/3, which complexes with co-Smad4 to activate target genes (reviewed in Hill, 2001). Foxh1 has been implicated as a critical Smad2/3 co-factor (Chen et al., 1996, 1997), and has an essential function during mesoderm and endoderm development based on studies in Xenopus (Howell et al., 2002; Kofron et al., 2004; Chiu et al., 2014), zebrafish (Pogoda et al., 2000; Kunwar et al., 2003; Sirotkin et al., 2000; Slagle et al., 2011), mouse (Hoodless et al., 2001; Yamamoto et al., 2001), and differentiated human embryonic stem cells (Yoon et al., 2011).

    • Small C-terminal domain phosphatase 3 dephosphorylates the linker sites of receptor-regulated smads (R-Smads) to ensure transforming growth factor β (TGFβ)-mediated germ layer induction in Xenopus embryos

      2015, Journal of Biological Chemistry
      Citation Excerpt :

      As such, shortly after the MBT, a good supply of responsive R-Smad allows for the rapid activation of TGFβ signaling. Spatially, SCP3 is enriched vegetally, consistent with the vegetally enriched active R-Smads at stage 9.5–10, during which Nodal/Activin and BMP signals start to be highly activated (1, 7, 10, 12). Compared with SCP3 in the vegetal part, the lower level of SCP3 in the animal part causes weaker but necessary dephosphorylation effects, possibly contributing to activation of R-Smads not restricted in the vegetal region, such as activated Smad1 in the ventral side across the animal-vegetal axis during gastrulation (10–12).

    View all citing articles on Scopus
    View full text