Retinoic acid signalling in the development of branchial arches

https://doi.org/10.1016/j.gde.2004.07.012Get rights and content

Branchial arches develop through a complex sequence of interactions between migrating cells, derived from neural crest and mesoderm, and epithelia of ectodermal and endodermal origin, to yield a variety of derivatives, notably skeletal elements, arteries and glands. In all vertebrate species, dramatic malformations generated by experimental blocks or activations of retinoic acid signalling highlight key roles for this molecule in the endoderm for branchial arch formation and morphogenesis.

Introduction

Branchial arches (BAs) are transient bulges of the embryonic oropharyngeal region (Figure 1a–c), giving rise to specific sets of adult tissue derivatives (see Table 1 for examples), the formation of which critically depends upon retinoic acid (RA). This active metabolite of vitamin A (retinol) is indispensable for patterning the anteroposterior body axis, for morphogenesis and organogenesis [1]. Its tissue distribution results from the balancing activities of RA-synthesizing enzymes (including retinaldehyde dehydrogenases RALDH1 to RALDH4), and RA-catabolizing cytochrome P450 hydroxylases (CYP26A1, B1 and C1) 2., 3.. The pleiotropic effects of RA are mediated through ligand-dependent transcriptional regulators belonging to the nuclear receptor superfamily: the retinoic acid receptors (RARα, β and γ isotypes) and the retinoid X receptors (RXRα, β and γ isotypes) that bind as RXR/RAR heterodimers to response elements located in RA-responsive genes. A genetic dissection, through targeted mutagenesis performed in the mouse, has revealed that RXRα/RAR(α, β and γ) heterodimers represent the main functional units of the RA signalling pathway during embryonic development [1].

Section snippets

Retinoid signalling mediated by RXRα/RAR-heterodimers is indispensable for the development of branchial arches 3–6

When analyzed between embryonic day (E) 9.5 and E 10.5, mouse embryos carrying targeted inactivation of RARα and RARβ (RARαβ-null mutants), or RARα and RARγ (RARαγ-null mutants) genes, display hypoplasia of BA3–6 and their corresponding branchial pouches (BPs) 4., 5.. Similar hypoplasia of BA3 and BP3 is observed in vitamin-A-deficient (VAD) quails and rats 6., 7., 8., in RALDH2-null and hypomorphic (RALDH2hypo) mutant mice 9., 10., 11., 12.•, and in mice expressing a dominant negative (dn)

The development of branchial arches 1–2 involves retinoic acid-dependent cellular and molecular mechanisms that are distinct from those in caudal branchial arches

BA1 is never missing under experimental conditions resulting in blocks in RA signalling, and hypoplasia of BA1 only occurs in a context of general embryonic growth retardation caused by RA deprivation 7., 9.. However, several BA1-derivatives cannot develop properly when RA signalling is impaired. For instance, morphogenesis of molars, which are derived from BA1 ectoderm and mesenchyme, clearly requires RA (Figure 3a,b). Similarly, atavistic modifications of chondrogenesis in BA1 of RARα-null

The teratogenic effects of retinoic acid excess and deficiency involve distinct cellular and molecular mechanisms in the oropharyngeal region

Treatments with pharmacological doses of RA, or other retinoids, during pregnancy causes multiple malformations which are receptor-mediated, and whose nature depends on the time of RA administration. In humans, intake of isotretinoin (13-cis RA) during gastrulation and early organogenesis (gestational weeks 3–5) results in a spectrum of congenital malformations collectively referred to as RA-induced embryopathy (RAE) [46]. In animal models of RAE, fusion and hypoplasia of the first two BA

Conclusions

Malformations induced by excess RA resemble those generated upon silencing RA-dependent pathways. For instance, a CATCH22-like spectrum of defects is recapitulated in mouse fetuses exposed to excess vitamin A [51]. Such similarities have popularized the idea that both RA excess and RA deficiency impair developmental processes by disrupting identical mechanisms. With respect to the early development of the oropharyngeal region, this is, however, clearly not the case for the following reasons.

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

Acknowledgements

This review is dedicated to the memory of Professor Jean-Victor Ruch. We thank all past and current members of the group for exciting discussions. We apologize that, because of strict space limitations, many primary references could not be cited directly. They will be found in reviews cited in the text. This work was supported by funds from the Centre National de la Recherche Scientifique (CNRS), the Institut National de la Santé et de la Recherche Médicale (INSERM), the Collège de France, and

References (55)

  • G.B. Mulder et al.

    Effects of excess vitamin A on development of cranial neural crest-derived structures: a neonatal and embryologic study

    Teratology

    (2000)
  • K. Niederreither et al.

    Differential expression of retinoic acid-synthesizing (RALDH) enzymes during fetal development and organ differentiation in the mouse

    Mech Dev

    (2002)
  • M. Mark et al.

    Functions of RARs and RXRs in vivo: genetic dissection of the retinoid signaling pathway

    Pure Appl Chem

    (2003)
  • G. Duester

    Families of retinoid dehydrogenases regulating vitamin A function: production of visual pigment and retinoic acid

    Eur J Biochem

    (2000)
  • S. Reijntjes et al.

    Generating gradients of retinoic acid in the chick embryo: Cyp26C1 expression and a comparative analysis of the Cyp26 enzymes

    Dev Dyn

    (2004)
  • V. Dupé et al.

    Key roles of retinoic acid receptors alpha and beta in the patterning of the caudal hindbrain, pharyngeal arches and otocyst in the mouse

    Development

    (1999)
  • O. Wendling et al.

    Roles of retinoic acid receptors in early embryonic morphogenesis and hindbrain patterning

    Development

    (2001)
  • M. Maden et al.

    Vitamin A-deficient quail embryos have half a hindbrain and other neural defects

    Curr Biol

    (1996)
  • K. Niederreither et al.

    Embryonic retinoic acid synthesis is essential for early mouse post-implantation development

    Nat Genet

    (1999)
  • K. Niederreither et al.

    Retinoic acid synthesis and hindbrain patterning in the mouse embryo

    Development

    (2000)
  • F.A. Mic et al.

    Novel retinoic acid generating activities in the neural tube and heart identified by conditional rescue of Raldh2 null mutant mice

    Development

    (2002)
  • J. Vermot et al.

    Decreased embryonic retinoic acid synthesis results in a DiGeorge syndrome phenotype in newborn mice

    Proc Natl Acad Sci USA

    (2003)
  • A. Iulianella et al.

    Chimeric analysis of retinoic acid receptor function during cardiac looping

    Dev Biol

    (2002)
  • G. Begemann et al.

    The zebrafish neckless mutation reveals a requirement for raldh2 in mesodermal signals that pattern the hindbrain

    Development

    (2001)
  • J.M. Lampert et al.

    Provitamin A conversion to retinal via the beta,beta-carotene-15,15′-oxygenase (bcox) is essential for pattern formation and differentiation during zebrafish embryogenesis

    Development

    (2003)
  • J.G. Wilson et al.

    An analysis of the syndrome of malformations induced by maternal vitamin A deficiency. Effects of restoration of vitamin A at various times during gestation

    Am J Anat

    (1953)
  • C. Mendelsohn et al.

    Function of the retinoic acid receptors (RARs) during development (II). Multiple abnormalities at various stages of organogenesis in RAR double mutants

    Development

    (1994)
  • Cited by (71)

    • Synovial tissue metabolomics analysis of the therapeutic effects of stir-fried Xanthii Fructus on rheumatoid arthritis in rats

      2023, Journal of Chromatography B: Analytical Technologies in the Biomedical and Life Sciences
    • Retinoic Acid Organizes the Zebrafish Vagus Motor Topographic Map via Spatiotemporal Coordination of Hgf/Met Signaling

      2020, Developmental Cell
      Citation Excerpt :

      It is therefore unclear whether RA can influence the A-P identity of mature vagus neurons. To address this question, we first visualized a direct reporter for RA signaling (Tg(12xrare-ef1a:EGFP)sk71) (Mark et al., 2004; Waxman and Yelon, 2011). Consistent with a role for RA in specifying A-P mX identity, RARE:GFP is detectable in the posterior but not the anterior of the vagus nucleus (Figure 2F).

    • Pharmacokinetics and therapeutic efficiency of a novel cationic liposome nano-formulated all trans retinoic acid in lung cancer mice model

      2017, Journal of Drug Delivery Science and Technology
      Citation Excerpt :

      All trans retinoic acid (ATRA) has been reported to treat various types of cancers such as acute promyelocytic leukemia (APL), lung cancer, head and neck cancer, breast cancer [1–4]. Due to its pleotropic regulator function in cell, ATRA participates in many physiological pathways such as cell proliferation, differentiation, embryogenesis, morphogenesis, apoptosis, and inflammation [5]. ATRA being a lipid – acid, is highly lipophilic with low water solubility which is sensitive to light and heat [6].

    View all citing articles on Scopus
    View full text