<<

View metadata, citation and similar papers at core.ac.uk brought to you by CORE

provided by Elsevier - Publisher Connector

Developmental Biology 276 (2004) 1–15 www.elsevier.com/locate/ydbio Review Early development of the cranial sensory : from a common field to individual placodes

Andrea Streit

Department of Craniofacial Development, King’s College London, Guy’s Campus, London SE1 9RT, UK

Received for publication 23 April 2004, revised 20 August 2004, accepted 23 August 2004 Available online 30 September 2004

Abstract

Sensory placodes are unique columnar epithelia with neurogenic potential that develop in the vertebrate head ectoderm next to the . They contribute to the paired sensory organs and the cranial sensory ganglia generating a wide variety of cell types ranging from fibres to sensory receptor cells and neurons. Although progress has been made in recent years to identify the molecular players that mediate placode specification, induction and patterning, the processes that initiate placode development are not well understood. One hypothesis suggests that all placode precursors arise from a common territory, the pre-placodal region, which is then subdivided to generate placodes of specific character. This model implies that their induction begins through molecular and cellular mechanisms common to all placodes. Embryological and molecular evidence suggests that placode induction is a multi-step process and that the molecular networks establishing the pre-placodal domain as well as the acquisition of placodal identity are surprisingly similar to those used in Drosophila to specify sensory structures. D 2004 Elsevier Inc. All rights reserved.

Keywords: Placode; Development; Nervous system

Introduction olfaction and vision. While some placodes are simple neurogenic centres, others such as the otic and olfactory The vertebrate head ectoderm contains unique neuro- placodes generate a multitude of cell types and undergo genic regions: the cranial sensory placodes. Like the neural complex patterning events once a columnar epithelium has plate, which gives rise to the central nervous system (CNS), been established. placodes are transient columnar, pseudostratified epithelia. Despite their long history in experimental They arise at distinct rostrocaudal positions next to the (the term dplacodesT was coined in 1891 [von Kupffer, neural tube shortly after the start of and, because 1891]), the importance of placodes for sensory functions of their ability to generate neurons outside the central and their evolutionary significance as key structures nervous system, are excellent model systems to study both believed to have evolved along with the appearance of inductive interactions and . Placodal cells give higher chordates, placode development has been neglected rise to the majority of neurons that form the cranial sensory by modern developmental biologists until very recently. nervous system contributing to the cranial ganglia and the The availability of molecular markers specific for individ- special sense organs associated with hearing, balance, ual placodes as well as for particular cell types derived from them has now stimulated much new research in the field and has lead to exciting findings in placode induction, patterning and cell fate specification. An * Department of Craniofacial Development, King’s College London, Guy’s Hospital, Guy’s Tower Floor 27, St. Thomas Street, London SE1 excellent review has recently summarised in detail the 9RT, UK. Fax: +44 20 7188 1674. classical and current literature on tissue interactions and E-mail address: [email protected]. signals involved in the induction of individual placodes

0012-1606/$ - see front matter D 2004 Elsevier Inc. All rights reserved. doi:10.1016/j.ydbio.2004.08.037 2 A. Streit / 276 (2004) 1–15

(Baker and Bronner-Fraser, 2001). Here, I will concentrate on some of the earliest steps that initiate placode formation, asking the question of when and how placode induction begins and discussing the evidence for and against the view that all placodes arise from a common placodal field—the pre-placodal region —through a common molecular mechanism.

Cranial placodes: origin of diverse structures and cell types

Cranial placodes are regions of columnar epithelium that form between the 10- and 30-somite stage at precise positions along the developing brain (Alvarez and Navascues, 1990; Anniko and Wikstrom, 1984; Bancroft and Bellairs, 1977; Brunjes and Frazier, 1986; Croucher and Tickle, 1989; D’Amico-Martel and Noden, 1983; Hilfer et al., 1989; Mayordomo et al., 1998; Meier, 1978a,b; Mendoza et al., 1982; Romanoff, 1960; Schook, 1980; Van Campenhout, 1956; Wakely, 1976; Fig. 1B). The olfactory placode forms in the anteriormost position next to the forebrain; it gives rise to the olfactory and vomeronasal epithelium including their sensory neurons (Brunjes and Frazier, 1986), to a population of migratory GnRH neurons (Dellovade et al., 1998; Schwanzel- Fukuda and Pfaff, 1989; Wray et al., 1989) as well as to the glia that will ensheath the (Chuah and Au, 1991; Couly and Le Douarin, 1985; Norgren et Fig. 1. Position of the cranial placodes in a 1.5- and 3.5-day chick al., 1992). The lens placode forms next to the optic embryo. (A) At the 10- to 13-somite stage precursors for different vesicle and is the only cranial placode that does not give placodes are segregated and occupy unique positions in the head rise to neurons; it is included in this review by virtue of ectoderm. Fate maps combined from Bhattacharyya et al. (2004) and D’Amico-Martel and Noden (1983). (B) At 3.5 days of development, all its close association with the eye and because like all placodes can be recognised morphologically and some have begun to other placodes, it arises from the ectoderm close to the undergo morphogenesis or differentiation. The olfactory placode lies next . The next more caudal placode is the to the future olfactory bulb, while the lens is surrounded by the retina. trigeminal, which occupies a large region next to the The trigeminal placode is situated adjacent to the midbrain, while the otic mid- and anterior hindbrain and forms the trigeminal placode next to the hindbrain has already invaginated to form the . The epibranchial placodes are positioned just dorsal of the ganglion (V) which has somatosensory function and acts branchial clefts. as a relay station for stimuli such as temperature, pain and touch from the skin, jaws and teeth. Molecularly, the trigeminal placode can be subdivided into two separate post-otic lateral line placodes, which develop into entities: the maxillomandibular and the ophthalmic por- mechano- and electroreceptors as well as the neurons that tions (Baker et al., 1999; Begbie et al., 2002). In fact, in innervate them (Northcutt et al., 1994, 1995; Schlosser, some amphibians two ganglia initially develop and fuse 2002; Schlosser and Northcutt, 2000). These primordia later (Northcutt and Brandle, 1995), while in some lower undergo remarkable migration to deposit lines of lateral vertebrates, two distinct ganglia form (Northcutt and line organs in the ectoderm of the head and along the Bemis, 1993; Piotrowski and Northcutt, 1996). Caudal entire body axis. Finally, the three epibranchial placo- to the trigeminal, next to the hindbrain, lies the otic des—geniculate, petrosal and nodose—are located more placode; it gives rise to the specialised epithelia of the laterally, just dorsal to the branchial clefts, and form the inner ear, including endolymph secreting cells, supporting distal parts of the VII, IX and X cranial ganglia that cells, sensory hair cells for the detection of sound, balance innervate taste buds, visceral organs and the heart and acceleration and neurons forming the cochlear– (D’Amico-Martel and Noden, 1983; Schlosser and North- vestibular ganglion (for review, see Brown et al., 2003; cutt, 2000). It is important to note that all cranial ganglia Fritzsch et al., 1998, 2002; Rubel and Fritzsch, 2002; have a dual origin: while their distal portions are placodal Torres and Giraldez, 1998). In fish and aquatic amphib- derivatives, their proximal parts are of cell ians, it is surrounded by a variable number of pre- and origin. A. Streit / Developmental Biology 276 (2004) 1–15 3

The bplacodal fieldQ hypothesis—support from pathway, to which additional components were then added experimental embryology to specify the distinct identities of individual placodes. The idea of a common placodal field provides a plausible Despite the structural and functional diversity of sensory mechanism by which this could happen: the ancestral organs and cranial ganglia in the adult, it has been suggested placode might have expanded and subsequently subdivided that the induction of all placodes begins through a common into separate placodes each then acquiring a specific cellular and molecular mechanism, which involves the function. formation of a common bplacodal fieldQ—the pre-placodal In favour of the placodal field hypothesis, histological region (Baker and Bronner-Fraser, 2001; Jacobson, 1963c; studies in mouse, fish and some amphibian embryos have Torres and Giraldez, 1998). For such a field to exist two pre- revealed a bprimitive placodal thickeningQ that surrounds the requisites have to be fulfilled: first, all placodal precursors anterior neural plate at late gastrula to early stages should arise from a continuous domain of the embryonic (Knouff, 1935; Miyake et al., 1997; Platt, 1896; van ectoderm and second, this region should have unique Oostrom and Verwoerd, 1972; Verwoerd and van Oostrom, properties, being biased or specified towards a placodal 1979). This morphology is maintained in areas where fate, accompanied by the expression of a specific set of placodes form, while the ectoderm in the non-placode molecular markers. forming regions between them thins out. However, other The idea of a common pre-placodal region has been vertebrate embryos, like the chick, lack these morphological challenged recently based on developmental and evolu- characteristics. Based on previous findings by others and his tionary considerations (Begbie and Graham, 2001; Graham own experiments in amphibians, Jacobson (1963a,b,c, and Begbie, 2000). First, embryological findings suggested 1966) was the first to present a concise hypothesis for the that two types of placodes—the epibranchial and trigemi- complexity of cranial placode induction and the first to nal—can be induced through different tissue interactions suggest the existence of a common pre-placodal region. A and probably by different signalling molecules in a single large number of studies had determined that the induction of step and therefore do not share a common inductive different placodes involves similar tissue interactions: early mechanism (Baker et al., 1999; Begbie et al., 1999). signals from the endoderm and mesoderm promote the However, substantial evidence from both classical and formation of olfactory, lens and otic placodes (otic: modern experimental embryology indicates that placode Harrison, 1935; Orts-Llorca and Jimenez-Collado, 1971; induction is a drawn out process that involves a series of Stone, 1931; Waddington, 1937; Yntema, 1950; Zwilling, events before placodes become committed to their indivi- 1940a; see also Gallagher et al., 1996; nasal: Emerson, dual fates as exemplified for otic and lens induction (see 1945; Siggia, 1936; Yntema, 1955; Zwilling, 1940b; lens: below for detailed discussion). So far, there is no clear Jacobson, 1958; Liedke, 1942, 1951, 1955; see also Henry evidence that a specific placode can be induced in a single and Grainger, 1987, 1990). These signals, however, are not step from cells that during normal development never sufficient to trigger the development of a complete ear or contribute to the same or other placodes (see below). lens vesicle or a nasal sac. Later interactions with specific Furthermore, the fact that placode dinducingT signals or parts of the CNS seem to complete induction and to confer tissues differ from each other does not argue against a identity to individual placodes: while the forebrain promotes common pre-placodal state as they may represent final development of the , the triggers that confer placode identity onto cells that are promotes lens development and the hindbrain already biased towards a generic placodal fate. formation (Haggis, 1956; Harrison, 1920, 1936; Jacobson, Second, while a long-standing view holds that placodes, 1963a; Reyer, 1958a,b; Street, 1937). Thus, it seems that an like neural crest cells, evolved with the emergence of initial step common to all placodes involve signals from the vertebrates (Northcutt and Gans, 1983), structures analo- mesendoderm, while later, more region-specific signals gous to the otic and olfactory placodes were recently confer placode-specific properties. identified in non-vertebrate chordates based on the expres- In a second series of critical experiments, Jacobson sion of molecular markers and the presence of ciliated showed that within the ectoderm adjacent to the anterior sensory cells (reviewed in Graham and Begbie, 2000; neural plate, (the PPR) cells are competent to give rise to Holland and Holland, 2001; Manni et al., 2001; Shimeld any placode (Jacobson, 1963c): when future placode and Holland, 2000). So far, there is no evidence that lower territory is rotated along its rostrocaudal axis at open chordates possess structures homologous to epibranchial neural plate stages such that the future otic region is now placodes, suggesting that these evolved separately from the in the position of the olfactory placode and vice versa, remaining placodes. It has been argued that their independ- placodes develop according to their new position (Fig. 2). ent emergence during evolution precludes a common However, when rotated later, placode identity seems molecular mechanism initiating the induction of all verte- already determined, but interplacodal tissue remains com- brate placodes (Graham and Begbie, 2000). However, it is petent to form new placodes. Thus, at neurula stages, cells just as likely that once an ancestral placode developed new within the PPR are able to generate a placode different placodes could arise by co-option of the same molecular from their normal fate. These early findings highlight two 4 A. Streit / Developmental Biology 276 (2004) 1–15

mesoderm and the young neural plate, either of which alone is insufficient to trigger the formation of a mature lens (Grainger, 1996; Grainger et al., 1988, 1997; Henry and Grainger, 1990; Zygar et al., 1998; for review, see Chow and Lang, 2001). In the absence of the optic vesicle, molecular markers for the presumptive lens ectoderm, like Pax6, are maintained, however, the vesicle is crucial for the ectoderm to acquire placodal morphology as well as for lens maturation as reflected by the expression of lens-specific crystallins and lens fibre formation (Furuta and Hogan, 1998; Kamachi et al., 1998; Li et al., 1994; Porter et al., 1997). Bone Morphogenetic Protein (BMP) and Fibroblast Growth Factor (FGF) signalling have been implicated in mediating some of the interactions between the optic vesicle and the future lens ectoderm to form a mature placode (Faber et al., 2001; Furuta and Hogan, 1998; Wawersik et al., 1999; for review, see Chow and Lang, 2001), but the early signals responsible for setting up the presumptive lens region remain elusive. Fig. 2. Diagram of Jacobson’s rotation experiment in amphibians (Jacobson, 1963c). When the placodal domain of an open neural plate Induction of the otic placode depends on a similarly stage embryo was rotated along its rostrocaudal axis, placodes developed complex process: early signals from the mesoderm under- according to their new position (bottom left). Occasionally an extra otic lying the placode (Gallagher et al., 1996; Ladher et al., structure was observed next to the . When the same 2000; Mendonsa and Riley, 1999; Orts-Llorca and Jimenez- experiment was performed at late neurula stages (bottom right), placode Collado, 1971) seem to act in concert with signals from the identity was already established and placodes form according to their original fate. Some variation of the results is observed probably due to the neural tube to induce otic-specific markers and the otic exact timing and position of the transplant. Note, however, that even at later vesicle. The activity of the paraxial head mesoderm seems stages, inter-placodal ectoderm is still responsive to placode inducing to be mediated by FGF signalling, however, depending on signals and additional placodes are formed close to their normal position, the species, different members of the FGF family may be for example, the olfactory placode (diagram lower right). Olfactory: orange; involved (Ladher et al., 2000; Wright and Mansour, 2003; lens: blue; otic: purple. for discussion see Riley and Phillips, 2003). A role for FGF3 as one of the major otic inducers emanating from the important features of placode induction: its multi-step hindbrain is conserved among most vertebrates, while the nature and a potentially common inducing mechanism for involvement of other FGFs expressed in the hindbrain all placode precursors. seems to vary between species (Adamska et al., 2001; Alvarez et al., 2003; Leger and Brand, 2002; Lombardo and Slack, 1998; Mansour, 1994; Maroon et al., 2002; Phillips Placode induction—a multi-step process et al., 2001; Vendrell et al., 2000; Wright and Mansour, 2003). In addition, it has been proposed that in chick Multiple inducing tissues and signals hindbrain-derived Wnt signals act synergistically with FGFs to induce the otic placode (Ladher et al., 2000), although More recent findings clearly support the idea that placode this may not be the case in zebrafish (Phillips et al., 2004). induction is a complex multi-step process. For example, lens While it is clear that otic placode formation requires FGF induction was initially believed to depend on a single signalling, loss of function approaches have shown that inductive step, in which the optic vesicle instructs the even in the absence of at least two FGFs, or even most, if overlying ectoderm to develop into the lens placode (Lewis, not all, FGF signalling, some cells retain otic marker 1904, 1907; Spemann, 1901): extirpation of the vesicle led expression and may form a rudimentary otic epithelium to failure of lens formation, while its ectopic transplantation (Liu et al., 2003; Maroon et al., 2002; Phillips et al., 2001; appeared to induce lens from belly ectoderm. However, Wright and Mansour, 2003). This raises the possibility that soon thereafter conflicting evidence was obtained, showing additional upstream signals initiating otic development normal lens formation in the absence of the optic vesicle remain to be identified. (King, 1905; Mencl, 1903), and other tissues were In contrast, it has been suggested that the trigeminal and implicated in lens induction (Henry and Grainger, 1987; epibranchial placodes giving rise to cranial ganglia require Jacobson, 1958; Liedke, 1942, 1951, 1955). Recent studies less complexity of inductive interactions or signals. One or using molecular markers and labelling techniques to identify more unknown signal(s) from the neural tube (no other graft and host tissues unequivocally suggest that indeed, tissues being required) promote the formation of the early lens induction depends on signals from both the ophthalmic part of the trigeminal placode (Stark et al., A. Streit / Developmental Biology 276 (2004) 1–15 5

1997), and epibranchial placodes are generated through an transcription factors can initiate placode formation, they can interaction with pharyngeal endoderm, where it comes into only do so near cells that normally give rise to placodes, contact with the ectoderm just dorsal to the branchial clefts namely within the pre-placodal region. (Begbie et al., 1999). This endoderm expresses high levels In summary, the findings described above strongly of BMP7, and this factor can induce neuronal markers support the notion that formation of mature placodes is a characteristic of epibranchial placodes in isolated, non- multi-step process where different tissues and signals act epibranchial ectoderm, whereas inhibition of BMP signal- sequentially and/or cooperatively. So far, no experimental ling results in the loss of epibranchial neurons. Thus, BMP7 manipulation has yet led to the induction of placodes away is clearly involved in some step of epibranchial placode from their normal position, in ectoderm that does not specification. However, there is one caveat in interpreting normally contain placode precursors. Therefore, it is likely both experiments: the test tissue used to assay placode that some of the upstream signals that initiate placode induction is ectoderm either fated to become the same formation remain to be identified. Could one of the placode or ectoderm containing precursors for other upstream steps involve the formation of a pre-placodal placodes. It is therefore possible that these tissues had domain as suggested by Jacobson? already received other signals at the time of their isolation, which may have established a placodal bias. The pre-placodal region is defined by cell fates and Placode-specific transcription factors can only induce unique gene expression ectopic placodes within the pre-placodal domain The existence of a pre-placodal region implies that all While the studies summarised above focused on the placodal precursors arise from a continuous domain of the inducing signals and tissues, functional analysis of the embryonic ectoderm. Many of the fate map studies that transcription factors that integrate these signals within the localised future placodes to the ectoderm and outer neural placode have revealed similar levels of complexity. A large folds in the head have been performed after neurula stages number of transcription factors, among them members of when precursors for different placodes begin to be confined the Sox, Dlx, Fox and Pax gene families, have been to unique regions (Fig. 1A; Carpenter, 1937; Couly and Le identified that are expressed in one or more placodes (for Douarin, 1985, 1987, 1988; D’Amico-Martel and Noden, review see Baker and Bronner-Fraser, 2001). While loss-of- 1983; Rfhlich, 1931; for review, see Baker and Bronner- function approaches have clearly demonstrated that some of Fraser, 2001). A recent study in zebrafish shows a continuous these genes are essential for placode formation, so far gain- domain of placode precursors adjacent to the future brain at of-function experiments have failed to identify any factor gastrula stages (Kozlowski et al., 1997), while lineage sufficient to make non-placodal ectoderm (future epidermis) studies in the chick have identified a unique region of the acquire placodal characteristics. embryonic ectoderm that contains precursors for most, if not The two homeobox genes, Pax6 and Six3, are necessary all, placodes abutting the anterior neural plate slightly later for normal vertebrate eye and lens development (Carl et al., (Bhattacharyya et al., 2004; Streit, 2002; Fig. 3). In this 2002; Collinson et al., 2000, 2003; Grindley et al., 1997; territory, precursors for different placodes are intermingled Hogan et al., 1986; Lagutin et al., 2003; Quinn et al., 1996; and are recruited from a large area to their final position Wallis and Muenke, 1999; for review, see: Cvekl and (Bhattacharyya et al., 2004; Streit, 2002; Whitlock and Piatigorsky, 1996; Gehring and Ikeo, 1999; Kawakami et Westerfield, 2000). These studies first identify the pre- al., 2000; Oliver and Gruss, 1997). Overexpression of either placodal region at early neurula stages surrounding the neural factor results in ectopic lenses, which are, however, always plate from forebrain to hindbrain levels, reminiscent of the associated with neural structures (either ectopic retinae or a bprimitive placodal thickeningQ observed in mouse, fish and lens that develops very close to the host neural tube; some amphibians and of Jacobson’s bplacodal fieldQ. While Altmann et al., 1997; Chow et al., 1999; Lagutin et al., fate map studies cannot resolve the question of whether or 2001; Oliver et al., 1996). Expression of Six3 under the not different placodes share some inductive events, the fact control of regulatory elements that drive Pax2 expression in that their precursors initially overlap considerably is con- the otic territory leads to ectopic Pax6 expression in the sistent with the notion of a pre-placodal region. placode (Lagutin et al., 2001). Likewise, misexpression of If a common placode territory indeed exists, it should be the HMG-box factor Sox3 results in the formation of ectopic characterised not only by the position of relevant precursors, vesicles that express Pax6, but only next to endogenous but also by unique properties, like expression of a specific placodes (Koster et al., 2000). One of the earliest genes set of molecular markers that distinguish it from other expressed in the otic territory is the forkhead gene foxi1;itis regions of the ectoderm. Indeed, at neurula stages, members required for otic induction and can induce ectopic expres- of several gene families begin to be expressed or are up- sion of the otic marker Pax8 in a spatially restricted manner regulated in a horseshoe-shaped area encircling the anterior next to the neural tube, but not in ventral ectoderm (Nissen neural plate among them Six, Eya, Id, Dlx, Iro and Fox et al., 2003; Solomon et al., 2003a). Thus, while some genes (Akimenko et al., 1994; Esteve and Bovolenta, 1999; 6 A. Streit / Developmental Biology 276 (2004) 1–15

Fig. 3. Placode precursors at mid-gastrula and early neurula stages. (A) In zebrafish, placode precursors are found in the ectoderm adjacent to the future central nervous system (grey) from fore- to hindbrain level. Precursors for different placodes partially overlap, but are roughly organised according to their later position along the rostrocaudal axis. Vermillion: lens and olfactory placode, telencephalon; pale yellow: lens, olfactory epithelium, anterior cranial ganglia, otic vesicle, telencephalon; green: otic vesicle, posterior lateral line; dark grey: caudal lateral line; modified according to (Kozlowski et al., 1997); CNS fate map after (Woo and Fraser, 1995). (B) At the 0–1 somite stage in the chick, precursors for different placodes are intermingled in a horseshoe-shaped domain encircling the neural plate. Their posterior limit lies at the level of Hensen’s node adjacent to cells that will contribute to 6–7. Olfactory: orange; lens: blue; otic: purple; epibranchial: dark blue. Note: complete maps for epibranchial and trigeminal precursors are not available. Modified after Bhattacharyya et al. (2004) and Streit (2002).

Kee and Bronner-Fraser, 2001a,b; Liu and Harland, 2003; Thus, the position of placode precursors is demarcated by Luo et al., 2001a,b; McLarren et al., 2003; Mishima and co-expression of a specific set of genes in a continuous Tomarev, 1998; Nissen et al., 2003; Ohyama and Groves, domain surrounding the rostral neural plate, strongly 2004; Pandur and Moody, 2000; Papalopulu and Kintner, supporting the idea that different placodes initially share a 1993; Pera et al., 1999; Sahly et al., 1999; Schlosser and developmental programme. It is conceivable that cells Ahrens, 2004; Solomon and Fritz, 2002; Solomon et al., within this region have received signals to bias them 2003b; Streit, 2002; Wilson and Mohun, 1995). Some of towards a placodal fate, which in turn may account for the them, like Dlx genes, are not uniquely found in placodal fact that induction of ectopic placodes has so far only been cells but also encompass other ectodermal derivatives like achieved close to their normal position (see above). The future epidermis and/or neural crest. In contrast, Six and Eya precise function of Six and Eya proteins during these gene expression domains most closely reflect the location of processes is still unclear, however, it is possible that they placode precursors (compare Figs. 3 and 4) and members of specify cells as placodal without conferring placodal both families have been described in comparable patterns in identity. While several studies have addressed specification chick, fish and amphibian embryos (Esteve and Bovolenta, of individual placodes, we know nothing about the 1999; McLarren et al., 2003; Mishima and Tomarev, 1998; developmental state of pre-placodal cells. However, it is Pandur and Moody, 2000; Sahly et al., 1999; Schlosser and clear that this territory is unique due to its restriction to Ahrens, 2004; Streit, 2002). Although Xenopus fate map specific cell fates, its special properties and the expression data for the non-neural ectoderm at this stage are not of a unique set of molecular markers. available, a comparative expression analysis of placodal transcription factors relative to those found in neural and neural crest ectoderm supports the notion that Six and Eya The Six/Eya/Dach network in the pre-placodal region genes may represent pre-placodal markers (Schlosser and Ahrens, 2004). During later development, most of the genes Among the many genes expressed in the pre-placodal present in the placodal territory are either lost completely region, the expression of members of the Six and Eya from placodal tissue, maintained in only some placodes families most closely mirrors the cellular changes during (e.g., Fox) or down-regulated before re-appearing later in placode formation. Their Drosophila homologues, sine some mature placodes (e.g., Dlx). In contrast, mimicking oculis (so) and eyes absent (eya), form a regulatory network morphological changes of the ectoderm during placode that, together with the nuclear factor dachshund (dac), formation (see above), Six and Eya are the only transcripts controls specification of sensory structures, namely photo- to be maintained in all developing placodes, while being lost receptor cells (Bonini et al., 1993, 1997; Chen et al., 1997; Li from inter-placodal domains. et al., 2003; Mardon et al., 1994; Pignoni et al., 1997; Shen A. Streit / Developmental Biology 276 (2004) 1–15 7

Fig. 4. Expression of pre-placodal markers. Whole mount in situ hybridisation of 3–5 somite chick embryos using Six1 (A, aV), Eya2 (B, bV) and Dach1 (C, cV) antisense probes. Six1 (A, aV) and Eya2 (B, bV) expression in the ectoderm is confined to a domain surrounding the neural plate (np) from its most anterior edge to approximately node levels (arrow in A–C). This region corresponds to the position of placodal precursors, the pre-placodal domain (ppr; see Fig. 3). In contrast, Dach1 transcripts (C, cV) are distributed more widespread and found in the neural plate (np) as well as the pre-placodal domain (ppr), where they overlap with Six1 and Eya2. and Mardon, 1997; for review, see: Kumar and Moses, display inner ear abnormalities similar to the defects 2001c). Mutations in any of these genes leads to absence or observed in Eya1À/À mice: the otic vesicle fails to grow reduction of the Drosophila eye, while their overexpression and the cochlear duct and/or semicircular canals are absent can induce ectopic eye formation in a spatially restricted (Laclef et al., 2003; Li et al., 2003; Zheng et al., 2003). Like manner (Bonini et al., 1993; Mardon et al., 1994; Pignoni Eya1 mutant mice, Six1À/À mice lack both the cochlear– et al., 1997). In vertebrates, members of these families are not vestibular and petrosal ganglia. only expressed in the pre-placodal domain, but also later in However, a complete loss of placodal derivatives in any different combinations in each of the mature sensory of the mutants generated has not been observed as one placodes and some of their derivatives (for review, see might have expected if these genes were instrumental in Hanson, 2001; Kawakami et al., 2000). For example, while defining the pre-placodal domain. Since several members Eya1, Six1, Six4 and Dach1 are found in parts of the otic of the Six and Eya families are co-expressed in this region vesicle, the olfactory placode expresses Eya2, Six1 and Six3 and seem to share functional properties, the lack of an (Bovolenta et al., 1998; Davis et al., 1999; Ghanbari et al., early phenotype may be due to functional redundancy. 2001; Heanue et al., 2002; Mishima and Tomarev, 1998; There is good evidence that Six, Eya and Dac proteins Oliver et al., 1995; Pandur and Moody, 2000; Xu et al., form a regulatory network and that they interact physically 1997). to activate downstream target genes. In Drosophila, so, eya In addition, loss-of-function analysis has revealed an and dac expression is interdependent, while their combined important role for Six and Eya proteins in ear, eye and nasal misexpression in imaginal discs other than the eye disc development. Mice heterozygous for Eya1 show a con- shows strong synergistic effects with a dramatically ductive loss of hearing due to defects of the middle ear (Xu increased frequency of ectopic eye induction (Chen et al., et al., 1999), a condition very similar to the human 1997; Mardon et al., 1994; Pignoni et al., 1997; Shen and Branchio-Oto-Renal syndrome caused by a mutation in Mardon, 1997). Although the interdependence of gene the EYA1 gene (Abdelhak et al., 1997). Homozygous expression in vertebrates is not always conserved (Heanue Eya1À/À mice display severe inner ear defects: otic placode et al., 2002; Zheng et al., 2003; for review see: Hanson, formation is initiated normally but development arrests at 2001; Wawersik and Maas, 2000), the only case where the vesicle stage and the cochlear–vestibular ganglia do not compound homozygous mutations have been generated in form (Xu et al., 1999). In addition, the epibranchial placode mice, namely for Six1 and Eya1, shows a more severe ear derived petrosal ganglia are missing. Mutations in the phenotype than each mutation separately (Li et al., 2003; human EYA1 gene also lead to congenital eye defects Zheng et al., 2003), supporting the idea that they may act in (Azuma et al., 2000), however, the eye phenotypes in mice a molecular network and synergise with each other. In have been less well studied. Mice lacking Six1 function addition, in vitro studies have clearly demonstrated a 8 A. Streit / Developmental Biology 276 (2004) 1–15 physical interaction between Six and Eya as well as Dach has been conserved in vertebrates to regulate sensory organ and Eya proteins (Chen et al., 1997; Ikeda et al., 2002; Li development at different levels. et al., 2003; Ohto et al., 1999; Silver et al., 2003). While Six1 alone or Six1/Dach seems to act as transcriptional repressors, Eya recruits co-activators; due to their phospha- Signals and tissues mediating the induction of the tase activity, Eya proteins stabilise co-activator interactions pre-placodal domain and switch the complex to activate downstream target genes (Li et al., 2003). Thus, co-expression of Six, Eya and Dach How is the pre-placodal region established in the family members in the pre-placodal domain may be func- ectoderm adjacent to the rostral neural plate? As mentioned tionally significant and their role will need to be addressed at the beginning of this review, studies in amphibian in the future. embryos have implicated the endoderm, the presumptive In the Drosophila eye, the Six/Eya/Dach network heart mesoderm and the neural plate as early placode functions downstream of the Pax6 homologue eyeless inducing tissues, which may act in combination or (ey), while in other embryonic tissues, their expression is sequentially. Neither mesoderm nor endoderm alone, how- independent of Pax genes (Bonini et al., 1997; Halder et ever, appears to be sufficient to trigger the development of a al., 1998; Kumar and Moses, 2001b; Niimi et al., 1999). complete ear or lens vesicle or a nasal sac. Signals emitted Interestingly, during vertebrate placode development, Pax by these tissues may be responsible for setting up the initial gene expression (with the exception of Pax6 in the lens- common placodal field. olfactory domain; Bhattacharyya et al., 2004; Li et al., Neural crest cells arise at the border of the neural plate in 1994) is only initiated once placodes with characteristic close association with future placodes. Interaction of the identity begin to be specified. While Pax6 identifies the neural plate with future epidermis is sufficient to induce lens and later the olfactory placode (Grindley et al., 1995; neural crest at the interface (Dickinson et al., 1995; Mancilla Li et al., 1997; Nornes et al., 1998; Schlosser and Ahrens, and Mayor, 1996; Moury and Jacobson, 1990; Selleck and 2004; Walther and Gruss, 1991), Pax2 is specific for the Bronner-Fraser, 1995) and both Wnt and BMP pathways otic and epibranchial (Groves and Bronner-Fraser, 2000; have been implicated in this process (for review, see: Aybar Heller and Brandli, 1999; Krauss et al., 1991; Pfeffer et al., and Mayor, 2002; Knecht and Bronner-Fraser, 2002; 1998; Schlosser and Ahrens, 2004) and Pax3 for the LaBonne, 2002; Mayor and Aybar, 2001; Nieto, 2001; trigeminal placode (Stark et al., 1997). So far none of the Yanfeng et al., 2003). Are the same tissue interactions and Pax genes seems to be expressed in the lateral line signalling pathways responsible for placode induction? In placodes of amphibians and fish embryos (Schlosser and Drosophila, the expression of so, eya and dac during eye Ahrens, 2004). It is therefore tempting to speculate that the development is dependent on the presence of the BMP expression of members of the Six/Eya/Dach network in the homologue decapentaplegic (dpp) (Chen et al., 1999; Curtiss pre-placodal domain assembles some of the molecular and Mlodzik, 2000), while in the leg imaginal disc dac is network required for placode formation, but is not induced by a combination of dpp and the Wnt homologue sufficient by itself to complete the developmental pro- wingless (Wg) (Lecuit and Cohen, 1997). It will be gramme. In this scenario, Pax gene expression may initiate interesting to investigate whether similar upstream signalling the formation of a placode proper and together with other pathways control pre-placodal gene expression. Some transcription factors confer placode identity. Thus, one support for this idea comes from the finding that BMPs potential role of the Six/Eya/Dach network is to bestow participate in patterning the ectoderm during or shortly after placodal competence or bias to the ectoderm next to the gastrulation (for review, see: Mayor and Aybar, 2001; Sasai anterior neural plate. and De Robertis, 1997; Wilson and Hemmati-Brivanlou, A quite different possibility, however, is that the Six/Eya/ 1997). BMPs are thought to act as a morphogen gradient to Dach network is involved in regulating proliferation of pre- specify different cell types at distinct concentrations: high placodal cells. Loss-of-function alleles of so, eya and dac in levels of BMP activity promote the formation of epidermis, Drosophila produce a transient overgrowth within the eye intermediate levels the formation of neural crest, while low field, followed by extensive cell death resulting in reduction or no BMP signalling allows neural development (Barth or loss of the eye. In vertebrates, in vivo and in vitro data et al., 1999; Nguyen et al., 1998, 2000; Tribulo et al., 2003; suggest that lack of functional Six1, Eya2 or Eya3 leads to Wilson et al., 1997). A recent study revealed that expression reduced proliferation (Li et al., 2003; Pignoni et al., 1997; of the homeodomain protein Iro1, which labels the pre- Zheng et al., 2003). As a consequence, Six1 as well as Eya1 placodal region, is regulated by BMP (Glavic et al., 2004). mutant mice show severe inner ear phenotypes displaying However, the formation of olfactory and otic placodes is small otic vesicles that lack several otic placode-derived differentially affected in zebrafish mutants that show structures or cell types. moderate or severe disruption of the BMP signalling Regardless of the precise molecular function of the Six/ pathway (Nguyen et al., 1998), making it unlikely that a Eya/Dach network, it is remarkable that the same molecular unique threshold of BMP defines the placode territory. cassette used in Drosophila to specify photoreceptor cells Future studies will be necessary to unravel the signalling A. Streit / Developmental Biology 276 (2004) 1–15 9 pathways that specify the pre-placodal region and to these movements are random and that only cells that determine how these are integrated with signalling mecha- encounter placode-specific inducing signals from surround- nism that define the position of individual placodes. ing tissues are directed towards a specific fate. Alternatively, individual cells or groups of cells may already have acquired specific identities and according to these properties Transcription factor codes for different ectodermal move directionally to their final destination, sorting out domains? from their neighbours. Interestingly, some placode-specific genes like the otic marker Pax2 begin to be expressed in a One important question that remains open is how salt-and-pepper pattern (Streit, 2002). Thus, mosaic gene ectodermal patterning signals are interpreted by individual expression may reflect differential properties of individual cells to implement cell fate decisions. Some of the earliest cells in the pre-placodal territory. It will be necessary in the patterning events appear to begin before gastrulation, as future to investigate whether directional cues guide these indicated by the complex domains of gene expression in the precursors to their target position in the mature placode and ectoderm. Evidence from the chick suggests that before the degree to which these mosaic patterns of expression primitive streak formation the ectoderm is subdivided into might reflect differential competence to inducing signals, or two domains revealed by the mutually exclusive expression different states of commitment and differential responsive- of Sox3 and ERNI in one (Streit et al., 2000) and low levels ness to chemoattractants and/or repellents, or cell sorting of Msx1 (Streit and Stern, 1999), Dlx5 (Pera et al., 1999) mechanisms that would guide them to their appropriate and Gata2 and -3 (Sheng and Stern, 1999) in the other. destinations. Concomitant with the onset of expression of stable neural markers, like Sox2, in the future neural plate, Msx1 and the early neural crest marker Pax7 are up-regulated along its Parallels between fly and vertebrates: determination of border in a fairly broad stripe of cells. At the same time, pre- placode and imaginal disc identity placodal gene expression starts in the ectoderm lateral to the neural plate, overlapping medially with Msx1 and Pax7 Although vertebrate sensory placodes have no direct (Streit and Stern, 1999; Litsiou and Streit, unpublished structural or functional homologues in Drosophila and are observations). Shortly thereafter, however, early crest and unlikely to have evolved directly from sensory organs in the placodal markers are restricted to distinct regions of the fly, some of the molecular cassettes that regulate the ectoderm: Msx1 and Pax7 are confined to the neural folds development of such organs in both groups are surprisingly abutting Sox2 medially and Six1, Six4 and Eya2 laterally well conserved. The idea of a common domain for sensory (McLarren et al., 2003; Schlosser and Ahrens, 2004; Streit, placode precursors is remarkably similar to the situation in 2002). The successive restriction of transcription factors to Drosophila, where light, odour and sound perceiving cells distinct ectodermal domains may reflect the specification of arise from a common primordium, the eye-antennal disc. In different cell fates within the embryonic ectoderm. In the adult, photoreceptor cells are confined to the compound addition, these observations may point to negative as well eye, while the antenna contains cells with both auditory and as positive regulatory relationships between different tran- chemosensory function. Precursors for these cells arise from scription factors, which cooperate to integrate ectodermal the eye-antennal disc, a small group of epithelial cells that is patterning signals on a cell-by-cell basis. set aside during early development. During larval develop- ment eye and antennal precursors segregate until two separate entities are established that have distinct identities: Cell movements within the pre-placodal domain the eye and the antennal primordium. Some of the key regulators that confer identity to these primordia are the Once the pre-placodal domain is established, how do transcription factors distalless and eyeless (Cohen et al., precursors for individual placodes acquire distinct properties 1989; Dong et al., 2000; Halder et al., 1995, 1998; Sunkel and become different from each other? Within the pre- and Whittle, 1987; for review, see Gehring, 1996; Panga- placodal ectoderm, precursors for different placodes are niban and Rubenstein, 2002). Both proteins are initially co- initially interspersed (Bhattacharyya et al., 2004; Kozlowski expressed, but subsequently separate with eyeless being et al., 1997; Streit, 2002). Precursors for specific placodes confined to the eye primordium, while distalless is found in are recruited from a large area within this territory and the antennal part (Kumar and Moses, 2001a,b). It has been converge to their final position. Recent studies both in suggested that both transcription factors negatively cross zebrafish and chick suggest that they do so by extensive cell regulate each other to establish eye versus antennal cell fates movements while constantly changing their neighbours and (Kurata et al., 2000). Interestingly, a similar molecular undergoing cell rearrangements (Streit, 2002; Whitlock and mechanism may be involved in conferring lens versus Westerfield, 2000). In the chick, groups of cells from a olfactory placode identity in vertebrates. It is well estab- single location ultimately contribute to different placodes. lished that the vertebrate eyeless homologue, Pax6, is How is their segregation controlled? One possibility is that required in the presumptive lens ectoderm for development 10 A. Streit / Developmental Biology 276 (2004) 1–15 of a normal lens placode and a functional lens (Ashery- Precursors for different placodes arise from a common pre- Padan et al., 2000; for review, see Ashery-Padan and Gruss, placodal region characterised by the expression of a unique 2001; Callaerts et al., 1997; Gehring, 2002). Before placode set of molecular markers and by unique cellular properties. formation, however, Pax6 is expressed in the anterior Future studies will have to investigate the upstream signals ectoderm encircling the future forebrain—a region of the that establish the pre-placodal region as well as the role of ectoderm that contains cells fated to become lens and transcription factor networks implementing cell fate deci- olfactory placode (Bhattacharyya et al., 2004; Li et al., sions within this region. Molecular networks that govern 1994). Interestingly, the distalless homologue Dlx5 initially sensory organ formation in insects are remarkably con- colocalises with Pax6 in the ectoderm, however, as soon as served in vertebrates and may be used repeatedly to control the lens placode forms a columnar epithelium, Dlx protein is specification of the pre-placodal field, placode identity as lost form this territory, while Pax6 expression is maintained well as cell type specification within placodes. (Bhattacharyya et al., 2004). When lens precursors are forced to maintain Dlx5 beyond this time, they become excluded from the lens, cluster in the non-lens ectoderm and Acknowledgments acquire lens-specific gene expression (Bhattacharyya et al., 2004). These data suggest that the loss of Dlx protein is I am grateful to Claudio D. Stern for critical reading of required for cells to acquire lens identity in a process that the manuscript and would like to thank the anonymous parallels molecular events that lead to the manifestation of reviewers for their particularly constructive comments. imaginal disc identity in the fly. What is the cellular mechanism underlying this behav- iour? Like Dlx5 maintaining lens precursors, Pax6À/À cells References in mouse chimaeras are excluded from the lens territory (Collinson et al., 2000), suggesting that Pax6 expression Abdelhak, S., Kalatzis, V., Heilig, R., Compain, S., Samson, D., Vincent, C., Weil, D., Cruaud, C., Sahly, I., Leibovici, M., Bitner-Glindzicz, M., confers cells with adhesive properties different from their Francis, M., Lacombe, D., Vigneron, J., Charachon, R., Boven, K., neighbours. Similarly, in the leg imaginal disc of Droso- Bedbeder, P., Van Regemorter, N., Weissenbach, J., Petit, C., 1997. A / +/+ phila dllÀ À clones sort out from their dll neighbours human homologue of the Drosophila eyes absent gene underlies displaying apparent differential adhesion (Gorfinkiel et al., branchio-oto-renal (BOR) syndrome and identifies a novel gene family. 1997; Wu and Cohen, 1999; for review, see Panganiban Nat. Genet. 15, 157–164. Adamska, M., Herbrand, H., Adamski, M., Kruger, M., Braun, T., Bober, and Rubenstein, 2002). Interestingly, differential expres- E., 2001. FGFs control the patterning of the inner ear but are not able to sion of cell adhesion molecules is observed during lens induce the full ear program. Mech. Dev. 109, 303–313. placode formation in the mouse: placode cells acquire N- Akimenko, M.A., Ekker, M., Wegner, J., Lin, W., Westerfield, M., 1994. cadherin expression while concomitantly losing P-cadherin Combinatorial expression of the three zebrafish genes related to (Xu et al., 2002). Together, these findings raise the Distal-less: part of a homeobox gene code for the head. J. Neurosci. 14, 3475–3486. possibility that during vertebrate placode formation, a cell Altmann, C.R., Chow, R.L., Lang, R.A., Hemmati-Brivanlou, A., 1997. sorting mechanism acts to ensure that only cells with Lens induction by Pax-6 in Xenopus laevis. Dev. Biol. 185, 119–123. appropriate gene expression profiles are included in the Alvarez, I.S., Navascues, J., 1990. Shaping, invagination, and closure of the developing placode, while those that do not conform with chick embryo otic vesicle: scanning electron microscopic and quanti- their neighbours are excluded. Future experiments will tative study. Anat. Rec. 228, 315–326. Alvarez, Y., Alonso, M.T., Vendrell, V., Zelarayan, L.C., Chamero, P., need to address whether adhesive properties of placodal Theil, T., Bosl, M.R., Kato, S., Maconochie, M., Riethmacher, D., cells are under direct or indirect control of transcription Schimmang, T., 2003. Requirements for FGF3 and FGF10 during inner factors like Pax6 and Dlx5. ear formation. Development 130, 6329–6338. Thus, despite their developmental and structural diffe- Anniko, M., Wikstrom, S.O., 1984. Pattern formation of the otic placode rences, there is a surprising similarity in how cells and morphogenesis of the otocyst. Am. J. Otolaryngol. 5, 373–381. Ashery-Padan, R., Gruss, P., 2001. Pax6 lights-up the way for eye contributing to sensory organs are established in the fly development. Curr. Opin. Cell Biol. 13, 706–714. and vertebrates. Not only are the same molecular networks Ashery-Padan, R., Marquardt, T., Zhou, X., Gruss, P., 2000. Pax6 activity deployed to specify identity, but they may also act to in the lens primordium is required for lens formation and for correct segregate precursors destined to contribute to different placement of a single retina in the eye. Genes Dev. 14, 2701–2711. sensory structures. Aybar, M.J., Mayor, R., 2002. Early induction of neural crest cells: lessons learned from frog, fish and chick. Curr. Opin. Genet. Dev. 12, 452–458. Azuma, N., Hirakiyama, A., Inoue, T., Asaka, A., Yamada, M., 2000. Mutations of a human homologue of the Drosophila eyes absent gene Conclusion (EYA1) detected in patients with congenital cataracts and ocular anterior segment anomalies. Hum. Mol. Genet. 9, 363–366. In summary, it is evident that placode induction is a Baker, C.V.H., Bronner-Fraser, M., 2001. Vertebrate cranial placodes. Part I. Embryonic induction. Dev. Biol. 232, 1–61. complex process that involves signals emanating from Baker, C.V., Stark, M.R., Marcelle, C., Bronner-Fraser, M., 1999. different tissues and which act sequentially and/or in parallel Competence, specification and induction of Pax-3 in the trigeminal to bestow individual identity to each sensory placode. placode. Development 126, 147–156. A. Streit / Developmental Biology 276 (2004) 1–15 11

Bancroft, M., Bellairs, R., 1977. Placodes of the chick embryo studied by Couly, G.F., Le Douarin, N.M., 1987. Mapping of the early neural SEM. Anat. Embryol. (Berlin) 151, 97–108. primordium in quail-chick chimeras. II. The prosencephalic neural plate Barth, K.A., Kishimoto, Y., Rohr, K.B., Seydler, C., Schulte-Merker, S., and neural folds: implications for the genesis of cephalic human Wilson, S.W., 1999. Bmp activity establishes a gradient of positional congenital abnormalities. Dev. Biol. 120, 198–214. information throughout the entire neural plate. Development 126, Couly, G., Le Douarin, N.M., 1988. The fate map of the cephalic neural 4977–4987. primordium at the presomitic to the 3-somite stage in the avian embryo. Begbie, J., Graham, A., 2001. The ectodermal placodes: a dysfunctional Development 103, 101–113. family. Philos. Trans. R. Soc. Lond., Ser. B Biol. Sci. 356, 1655–1660. Croucher, S.J., Tickle, C., 1989. Characterization of epithelial domains in Begbie, J., Brunet, J.F., Rubenstein, J.L., Graham, A., 1999. Induction of the nasal passages of chick embryos: spatial and temporal mapping of a the epibranchial placodes. Development 126, 895–902. range of extracellular matrix and cell surface molecules during Begbie, J., Ballivet, M., Graham, A., 2002. Early steps in the production of development of the nasal placode. Development 106, 493–509. sensory neurons by the neurogenic placodes. Mol. Cell. Neurosci. 21, Curtiss, J., Mlodzik, M., 2000. Morphogenetic furrow initiation and 502–511. progression during in Drosophila: the roles of Bhattacharyya, S., Bailey, A.P., Bronner-Fraser, M., Streit, A., 2004. decapentaplegic, hedgehog and eyes absent. Development 127, Segregation of lens and olfactory precursors from a common territory: 1325–1336. cell sorting and reciprocity of Dlx5 and Pax6 expression. Dev. Biol. Cvekl, A., Piatigorsky, J., 1996. Lens development and crystallin gene 271, 403–414. expression: many roles for Pax-6. BioEssays 18, 621–630. Bonini, N.M., Leiserson, W.M., Benzer, S., 1993. The eyes absent gene: D’Amico-Martel, A., Noden, D.M., 1983. Contributions of placodal and genetic control of cell survival and differentiation in the developing neural crest cells to avian cranial peripheral ganglia. Am. J. Anat. 166, Drosophila eye. Cell 72, 379–395. 445–468. Bonini, N.M., Bui, Q.T., Gray-Board, G.L., Warrick, J.M., 1997. The Davis, R.J., Shen, W., Heanue, T.A., Mardon, G., 1999. Mouse Dach, a Drosophila eyes absent gene directs ectopic eye formation in a homologue of Drosophila dachshund, is expressed in the developing pathway conserved between flies and vertebrates. Development 124, retina, brain and limbs. Dev. Genes Evol. 209, 526–536. 4819–4826. Dellovade, T.L., Pfaff, D.W., Schwanzel-Fukuda, M., 1998. The gonado- Bovolenta, P., Mallamaci, A., Puelles, L., Boncinelli, E., 1998. Expression tropin-releasing hormone system does not develop in Small-Eye (Sey) pattern of cSix3, a member of the Six/sine oculis family of transcription mouse phenotype. Brain Res. Dev. Brain Res. 107, 233–240. factors. Mech. Dev. 70, 201–203. Dickinson, M.E., Selleck, M.A., McMahon, A.P., Bronner-Fraser, M., Brown, S.T., Martin, K., Groves, A.K., 2003. Molecular basis of inner ear 1995. Dorsalization of the neural tube by the non-neural ectoderm. induction. Curr. Top. Dev. Biol. 57, 115–149. Development 121, 2099–2106. Brunjes, P.C., Frazier, L.L., 1986. Maturation and plasticity in the olfactory Dong, P.D., Chu, J., Panganiban, G., 2000. Coexpression of the homeobox system of vertebrates. Brain Res. 396, 1–45. genes Distal-less and homothorax determines Drosophila antennal Callaerts, P., Halder, G., Gehring, W.J., 1997. PAX-6 in development and identity. Development 127, 209–216. evolution. Annu. Rev. Neurosci. 20, 483–532. Emerson, H.S., 1945. The development of late gastrula explants of rana Carl, M., Loosli, F., Wittbrodt, J., 2002. Six3 inactivation reveals its pipiens in salt solution. J. Exp. Zool. 100, 497–521. essential role for the formation and patterning of the vertebrate eye. Esteve, P., Bovolenta, P., 1999. cSix4, a member of the six gene family of Development 129, 4057–4063. transcription factors, is expressed during placode and somite develop- Carpenter, E., 1937. The head pattern in Ambystoma studied by vital ment. Mech. Dev. 85, 161–165. staining and transplantation methods. J. Exp. Zool. 75, 103–129. Faber, S.C., Dimanlig, P., Makarenkova, H.P., Shirke, S., Ko, K., Lang, Chen, R., Amoui, M., Zhang, Z., Mardon, G., 1997. Dachshund and eyes R.A., 2001. Fgf receptor signaling plays a role in lens induction. absent proteins form a complex and function synergistically to induce Development 128, 4425–4438. ectopic eye development in Drosophila [see comments]. Cell 91, Fritzsch, B., Barald, K.F., Lomax, M.I., 1998. Early embryology of the 893–903. vertebrate ear. In: Rubel, E.W., Popper, A.N., Fay, R.R. (Eds.), Dev. Chen, R., Halder, G., Zhang, Z., Mardon, G., 1999. Signaling by the TGF- Audit. Syst., vol. 9. Springer, New York, pp. 80–145. beta homolog decapentaplegic functions reiteratively within the net- Fritzsch, B., Beisel, K.W., Jones, K., Farinas, I., Maklad, A., Lee, J., work of genes controlling retinal cell fate determination in Drosophila. Reichardt, L.F., 2002. Development and evolution of inner ear sensory Development 126, 935–943. epithelia and their innervation. J. Neurobiol. 53, 143–156. Chow, R.L., Lang, R.A., 2001. Early eye development in vertebrates. Annu. Furuta, Y., Hogan, B.L.M., 1998. BMP4 is essential for lens induction in Rev. Cell Dev. Biol. 17, 255–296. the mouse embryo. Genes Dev. 12, 3764–3775. Chow, R.L., Altmann, C.R., Lang, R.A., Hemmati-Brivanlou, A., Gallagher, B.C., Henry, J.J., Grainger, R.M., 1996. Inductive processes 1999. Pax6 induces ectopic eyes in a vertebrate. Development 126, leading to inner ear formation during Xenopus development. Dev. Biol. 4213–4222. 175, 95–107. Chuah, M.I., Au, C., 1991. Olfactory Schwann cells are derived from Gehring, W.J., 1996. The master control gene for morphogenesis and precursor cells in the olfactory epithelium. J. Neurosci. Res. 29, evolution of the eye. Genes Cells 1, 11–15. 172–180. Gehring, W.J., 2002. The genetic control of eye development and its Cohen, S.M., Bronner, G., Kuttner, F., Jurgens, G., Jackle, H., 1989. Distal- implications for the evolution of the various eye-types. Int. J. Dev. Biol. less encodes a homeodomain protein required for limb development in 46, 65–73. Drosophila. Nature 338, 432–434. Gehring, W.J., Ikeo, K., 1999. Pax 6: mastering eye morphogenesis and eye Collinson, J.M., Hill, R.E., West, J.D., 2000. Different roles for Pax6 in the evolution. Trends Genet. 15, 371–377. optic vesicle and facial epithelium mediate early morphogenesis of the Ghanbari, H., Seo, H.C., Fjose, A., Brandli, A.W., 2001. Molecular cloning murine eye. Development 127, 945–956. and embryonic expression of Xenopus Six homeobox genes. Mech. Collinson, J.M., Quinn, J.C., Hill, R.E., West, J.D., 2003. The roles of Pax6 Dev. 101, 271–277. in the cornea, retina, and olfactory epithelium of the developing mouse Glavic, A., Maris Honore, S., Gloria Feijoo, C., Bastidas, F., Allende, M.L., embryo. Dev. Biol. 255, 303–312. Mayor, R., 2004. Role of BMP signaling and the homeoprotein iroquois Couly, G.F., Le Douarin, N.M., 1985. Mapping of the early neural in the specification of the cranial placodal field. Dev. Biol. 272, 89–103. primordium in quail-chick chimeras. I. Developmental relationships Gorfinkiel, N., Morata, G., Guerrero, I., 1997. The homeobox gene Distal- between placodes, facial ectoderm, and prosencephalon. Dev. Biol. 110, less induces ventral appendage development in Drosophila. Genes Dev. 422–439. 11, 2259–2271. 12 A. Streit / Developmental Biology 276 (2004) 1–15

Graham, A., Begbie, J., 2000. Neurogenic placodes: a common front. and ear. I. Interactions within the ectoderm, and between the ectoderm Trends Neurosci. 23, 313–316. and underlying tissues. J. Exp. Zool. 154, 273–283. Grainger, R.M., 1996. New perspectives on embryonic lens induction. Jacobson, A.G., 1963b. The determination and positioning of the , lens, Semin. Cell Dev. Biol. 7, 149–155. and ear. II. The role of the endoderm. J. Exp. Zool. 154, 285–291. Grainger, R.M., Herry, J.J., Henderson, R.A., 1988. Reinvestigation of the Jacobson, A.G., 1963c. The determination and positioning of the nose, lens, role of the optic vesicle in embryonic lens induction. Development 102, and ear. III. Effects of reversing the antero-posterior axis of epidermis, 517–526. neural plate and . J. Exp. Zool. 154, 293–303. Grainger, R.M., Mannion, J.E., Cook Jr., T.L., Zygar, C.A., 1997. Jacobson, A.G., 1966. Inductive processes in embryonic development. Defining intermediate stages in cell determination: acquisition of a Science 125, 25–34. lens-forming bias in head ectoderm during lens determination. Dev. Kamachi, Y., Uchikawa, M., Collignon, J., Lovell-Badge, R., Kondoh, H., Genet. 20, 246–257. 1998. Involvement of Sox1, 2 and 3 in the early and subsequent Grindley, J.C., Davidson, D.R., Hill, R.E., 1995. The role of Pax-6 in eye molecular events of lens induction. Development 125, 2521–2532. and nasal development. Development 121, 1433–1442. Kawakami, K., Sato, S., Ozaki, H., Ikeda, K., 2000. Six family genes— Grindley, J.C., Hargett, L.K., Hill, R.E., Ross, A., Hogan, B.L., 1997. Structure and function as transcription factors and their roles in Disruption of PAX6 function in mice homozygous for the Pax6Sey- development. BioEssays 22, 616–626. 1Neu mutation produces abnormalities in the early development and Kee, Y., Bronner-Fraser, M., 2001a. Temporally and spatially restricted regionalization of the diencephalon. Mech. Dev. 64, 111–126. expression of the helix–loop-helix transcriptional regulator Id1 during Groves, A.K., Bronner-Fraser, M., 2000. Competence, specification and avian embryogenesis. Mech. Dev. 109, 331–335. commitment in otic placode induction. Development 127, 3489–3499. Kee, Y., Bronner-Fraser, M., 2001b. The transcriptional regulator Id3 is Haggis, A.J., 1956. Analysis of the determination of the olfactory placode expressed in cranial sensory placodes during early avian embryonic in ambystoma punctatum. J. Embryol. Exp. Morphol. 4, 120–138. development. Mech. Dev. 109, 337–340. Halder, G., Callaerts, P., Gehring, W.J., 1995. Induction of ectopic eyes by King, H.D., 1905. Experimental studies on the eye of the frog embryo. targeted expression of the eyeless gene in Drosophila [see comments]. Arch. Entwicklungsmech. 19, 84–107. Science 267, 1788–1792. Knecht, A.K., Bronner-Fraser, M., 2002. Induction of the neural crest: a Halder, G., Callaerts, P., Flister, S., Walldorf, U., Kloter, U., Gehring, W.J., multigene process. Nat. Rev., Genet. 3, 453–461. 1998. Eyeless initiates the expression of both sine oculis and eyes Knouff, R.A., 1935. The developmental pattern of ectodermal placodes in absent during Drosophila compound eye development. Development Rana pipiens. J. Comp. Neurol. 62, 17–71. 125, 2181–2191. Koster, R.W., Kuhnlein, R.P., Wittbrodt, J., 2000. Ectopic sox3 activity Hanson, I.M., 2001. Mammalian homologues of the Drosophila eye elicits sensory placode formation. Mech. Dev. 95, 175–187. specification genes. Semin. Cell Dev. Biol. 12, 475–484. Kozlowski, D.J., Murakami, T., Ho, R.K., Weinberg, E.S., 1997. Regional Harrison, R.G., 1920. Experiments on the lens in Ambystoma. Proc. Soc. cell movement and tissue patterning in the zebrafish embryo revealed by Exp. Biol. Med. 17, 200. fate mapping with caged fluorescein. Biochem. Cell Biol. 75, 551–562. Harrison, R.G., 1935. Factors concerned in the development of the ear in Krauss, S., Johansen, T., Korzh, V., Fjose, A., 1991. Expression of the Ambystoma punctatum. Anat. Rec. 64, 38–39. zebrafish paired box gene pax[zf-b] during early neurogenesis. Harrison, R.G., 1936. Relations of symmetry in the developing ear of Development 113, 1193–1206. Ambystoma punctatum. Proc. Natl. Acad. Sci. U. S. A. 22, 238–247. Kumar, J.P., Moses, K., 2001a. The EGF receptor and notch signaling Heanue, T.A., Davis, R.J., Rowitch, D.H., Kispert, A., McMahon, A.P., pathways control the initiation of the morphogenetic furrow during Mardon, G., Tabin, C.J., 2002. Dach1, a vertebrate homologue of Drosophila eye development. Development 128, 2689–2697. Drosophila dachshund, is expressed in the developing eye and ear of Kumar, J.P., Moses, K., 2001b. Expression of evolutionarily conserved eye both chick and mouse and is regulated independently of Pax and Eya specification genes during Drosophila embryogenesis. Dev. Genes genes. Mech. Dev. 111, 75–87. Evol. 211, 406–414. Heller, N., Brandli, A.W., 1999. Xenopus Pax-2/5/8 orthologues: novel Kumar, J.P., Moses, K., 2001c. Eye specification in Drosophila: insights into Pax gene evolution and identification of Pax-8 as the perspectives and implications. Semin. Cell Dev. Biol. 12, 469–474. earliest marker for otic and pronephric cell lineages. Dev. Genet. 24, Kurata, S., Go, M.J., Artavanis-Tsakonas, S., Gehring, W.J., 2000. Notch 208–219. signaling and the determination of appendage identity. Proc. Natl. Acad. Henry, J.J., Grainger, R.M., 1987. Inductive interactions in the spatial and Sci. U. S. A. 97, 2117–2122. temporal restriction of lens-forming potential in embryonic ectoderm of LaBonne, C., 2002. Vertebrate development: wnt signals at the crest. Curr. Xenopus laevis. Dev. Biol. 124, 200–214. Biol. 12, R743–R744. Henry, J.J., Grainger, R.M., 1990. Early tissue interactions leading to Laclef, C., Souil, E., Demignon, J., Maire, P., 2003. Thymus, kidney and embryonic lens formation in Xenopus laevis. Dev. Biol. 141, 149–163. craniofacial abnormalities in Six 1 deficient mice. Mech. Dev. 120, Hilfer, S.R., Esteves, R.A., Sanzo, J.F., 1989. Invagination of the otic 669–679. placode: normal development and experimental manipulation. J. Exp. Ladher, R.K., Anakwe, K.U., Gurney, A.L., Schoenwolf, G.C., Francis- Zool. 251, 253–264. West, P.H., 2000. Identification of synergistic signals initiating inner ear Hogan, B.L., Horsburgh, G., Cohen, J., Hetherington, C.M., Fisher, development. Science 290, 1965–1968. G., Lyon, M.F., 1986. Small eyes (Sey): a homozygous lethal Lagutin, O., Zhu, C.C., Furuta, Y., Rowitch, D.H., McMahon, A.P., Oliver, mutation on chromosome 2 which affects the differentiation of both G., 2001. Six3 promotes the formation of ectopic optic vesicle-like lens and nasal placodes in the mouse. J. Embryol. Exp. Morphol. 97, structures in mouse embryos. Dev. Dyn. 221, 342–349. 95–110. Lagutin, O.V., Zhu, C.C., Kobayashi, D., Topczewski, J., Shimamura, K., Holland, L.Z., Holland, N.D., 2001. Evolution of neural crest and placodes: Puelles, L., Russell, H.R., McKinnon, P.J., Solnica-Krezel, L., Oliver, amphioxus as a model for the ancestral vertebrate? J. Anat. 199, 85–98. G., 2003. Six3 repression of Wnt signaling in the anterior neuro- Ikeda, K., Watanabe, Y., Ohto, H., Kawakami, K., 2002. Molecular ectoderm is essential for vertebrate forebrain development. Genes Dev. interaction and synergistic activation of a promoter by Six, Eya, and 17, 368–379. Dach proteins mediated through CREB binding protein. Mol. Cell. Biol. Lecuit, T., Cohen, S.M., 1997. Proximal–distal axis formation in the 22, 6759–6766. Drosophila leg. Nature 388, 139–145. Jacobson, A.G., 1958. The roles of neural and non-neural tissues in lens Leger, S., Brand, M., 2002. Fgf8 and Fgf3 are required for zebrafish ear induction. J. Exp. Zool. 139, 525–557. placode induction, maintenance and inner ear patterning. Mech. Dev. Jacobson, A.G., 1963a. The determination and positioning of the nose, lens, 119, 91–108. A. Streit / Developmental Biology 276 (2004) 1–15 13

Lewis, W.H., 1904. Experimental studies on the development of the eye in Mendonsa, E.S., Riley, B.B., 1999. Genetic analysis of tissue interactions amphibia. I. On the origin of the lens, Rana palustris. Am. J. Anat. 3, required for otic placode induction in the zebrafish. Dev. Biol. 206, 505–536. 100–112. Lewis, W.H., 1907. Experimental studies on the development of the eye in Mendoza, A.S., Breipohl, W., Miragall, F., 1982. Cell migration from the amphibia. III. On the origin and differentiation of the lens, Rana chick olfactory placode: a light and electron microscopic study. palustris. Am. J. Anat. 6, 473–509. J. Embryol. Exp. Morphol. 69, 47–59. Li, H.S., Yang, J.M., Jacobson, R.D., Pasko, D., Sundin, O., 1994. Pax-6 is Mishima, N., Tomarev, S., 1998. Chicken Eyes absent 2 gene: isolation first expressed in a region of ectoderm anterior to the early neural plate: and expression pattern during development. Int. J. Dev. Biol. 42, implications for stepwise determination of the lens. Dev. Biol. 162, 1109–1115. 181–194. Miyake, T., von Herbing, I.H., Hall, B.K., 1997. Neural ectoderm, neural Li, H., Tierney, C., Wen, L., Wu, J.Y., Rao, Y., 1997. A single crest, and placodes: contribution of the otic placode to the ectodermal morphogenetic field gives rise to two retina primordia under the lining of the embryonic opercular cavity in Atlantic cod (Teleostei). influence of the prechordal plate. Development 124, 603–615. J. Morphol. 231, 231–252. Li, X., Oghi, K.A., Zhang, J., Krones, A., Bush, K.T., Glass, C.K., Moury, J.D., Jacobson, A.G., 1990. The origins of neural crest cells in the Nigam, S.K., Aggarwal, A.K., Maas, R., Rose, D.W., Rosenfeld, axolotl. Dev. Biol. 141, 243–253. M.G., 2003. Eya protein phosphatase activity regulates Six1-Dach- Nguyen, V.H., Schmid, B., Trout, J., Connors, S.A., Ekker, M., Mullins, Eya transcriptional effects in mammalian organogenesis. Nature 426, M.C., 1998. Ventral and lateral regions of the zebrafish gastrula, 247–254. including the neural crest progenitors, are established by a bmp2b/swirl Liedke, K., 1942. Lens competence in Rana pipiens. J. Exp. Zool. 90, pathway of genes. Dev. Biol. 199, 93–110. 331–351. Nguyen, V.H., Trout, J., Connors, S.A., Andermann, P., Weinberg, E., Liedke, K., 1951. Lens competence in Ambystoma punctatum. J. Exp. Mullins, M.C., 2000. Dorsal and intermediate neuronal cell types of the Zool. 117, 573–591. spinal cord are established by a BMP signaling pathway. Development Liedke, K., 1955. Studies on lens induction in Ambystoma punctatum. 127, 1209–1220. J. Exp. Zool. 130, 353–379. Nieto, M.A., 2001. The early steps of neural crest development. Mech. Dev. Liu, K.J., Harland, R.M., 2003. Cloning and characterization of Xenopus 105, 27–35. Id4 reveals differing roles for Id genes. Dev. Biol. 264, 339–351. Niimi, T., Seimiya, M., Kloter, U., Flister, S., Gehring, W.J., 1999. Direct Liu, D., Chu, H., Maves, L., Yan, Y.L., Morcos, P.A., Postlethwait, J.H., regulatory interaction of the eyeless protein with an eye-specific Westerfield, M., 2003. Fgf3 and Fgf8 dependent and independent enhancer in the sine oculis gene during eye induction in Drosophila. transcription factors are required for otic placode specification. Development 126, 2253–2260. Development 130, 2213–2224. Nissen, R.M., Yan, J., Amsterdam, A., Hopkins, N., Burgess, S.M., 2003. Lombardo, A., Slack, J.M., 1998. Postgastrulation effects of fibroblast Zebrafish foxi one modulates cellular responses to Fgf signaling growth factor on Xenopus development. Dev. Dyn. 212, 75–85. required for the integrity of ear and jaw patterning. Development 130, Luo, T., Matsuo-Takasaki, M., Lim, J.H., Sargent, T.D., 2001a. Differential 2543–2554. regulation of Dlx gene expression by a BMP morphogenetic gradient. Norgren Jr., R.B., Ratner, N., Brackenbury, R., 1992. Development of Int. J. Dev. Biol. 45, 681–684. olfactory nerve glia defined by a monoclonal antibody specific for Luo, T., Matsuo-Takasaki, M., Sargent, T.D., 2001b. Distinct roles for Schwann cells. Dev. Dyn. 194, 231–238. Distal-less genes Dlx3 and Dlx5 in regulating ectodermal development Nornes, S., Clarkson, M., Mikkola, I., Pedersen, M., Bardsley, A., in Xenopus. Mol. Reprod. Dev. 60, 331–337. Martinez, J.P., Krauss, S., Johansen, T., 1998. Zebrafish contains two Mancilla, A., Mayor, R., 1996. Neural crest formation in Xenopus laevis: pax6 genes involved in eye development. Mech. Dev. 77, 185–196. mechanisms of Xslug induction. Dev. Biol. 177, 580–589. Northcutt, R.G., Bemis, W.E., 1993. Cranial nerves of the coelacanth, Manni, L., Lane, N.J., Burighel, P., Zaniolo, G., 2001. Are neural crest and Latimeria chalumnae [Osteichthyes: Sarcopterygii: Actinistia], and placodes exclusive to vertebrates? Evol. Dev. 3, 297–298. comparisons with other craniata. Brain. Behav. Evol. 42, 1–76 Mansour, S.L., 1994. Targeted disruption of int-2 (fgf-3) causes devel- (Suppl. 1). opmental defects in the tail and inner ear. Mol. Reprod. Dev. 39, 62–67. Northcutt, R.G., Brandle, K., 1995. Development of branchiomeric and (discussion 67-8). lateral line nerves in the axolotl. J. Comp. Neurol. 355, 427–454. Mardon, G., Solomon, N.M., Rubin, G.M., 1994. Dachshund encodes a Northcutt, R.G., Gans, C., 1983. The genesis of neural crest and epidermal nuclear protein required for normal eye and leg development in placodes: a reinterpretation of vertebrate origins. Q. Rev. Biol. 58, 1–28. Drosophila. Development 120, 3473–3486. Northcutt, R.G., Catania, K.C., Criley, B.B., 1994. Development of lateral Maroon, H., Walshe, J., Mahmood, R., Kiefer, P., Dickson, C., Mason, I., line organs in the axolotl. J. Comp. Neurol. 340, 480–514. 2002. Fgf3 and Fgf8 are required together for formation of the otic Northcutt, R.G., Brandle, K., Fritzsch, B., 1995. Electroreceptors and placode and vesicle. Development 129, 2099–2108. mechanosensory lateral line organs arise from single placodes in Mayor, R., Aybar, M.J., 2001. Induction and development of neural crest in axolotls. Dev. Biol. 168, 358–373. Xenopus laevis. Cell Tissue Res. 305, 203–209. Ohto, H., Kamada, S., Tago, K., Tominaga, S.I., Ozaki, H., Sato, S., Mayordomo, R., Rodriguez-Gallardo, L., Alvarez, I.S., 1998. Morpholog- Kawakami, K., 1999. Cooperation of six and eya in activation of their ical and quantitative studies in the otic region of the neural tube in chick target genes through nuclear translocation of Eya. Mol. Cell. Biol. 19, embryos suggest a neuroectodermal origin for the otic placode. J. Anat. 6815–6824. 193, 35–48. Ohyama, T., Groves, A.K., 2004. Expression of mouse Foxi class genes in McLarren, K.W., Litsiou, A., Streit, A., 2003. DLX5 positions the neural early craniofacial development. Dev. Dyn. (in press). crest and pre-placode region at the border of the neural plate. Dev. Biol. Oliver, G., Gruss, P., 1997. Current views on eye development. Trends 259, 34–47. Neurosci. 20, 415–421. Meier, S., 1978a. Development of the embryonic chick otic placode. I. Oliver, G., Mailhos, A., Wehr, R., Copeland, N.G., Jenkins, N.A., Gruss, P., Light microscopic analysis. Anat. Rec. 191, 447–458. 1995. Six3, a murine homologue of the sine oculis gene, demarcates the Meier, S., 1978b. Development of the embryonic chick otic placode. II. most anterior border of the developing neural plate and is expressed Electron microscopic analysis. Anat. Rec. 191, 459–477. during eye development. Development 121, 4045–4055. Mencl, E., 1903. Ein Fall von beiderseitiger Augenlinsenausbildung Oliver, G., Loosli, F., Koster, R., Wittbrodt, J., Gruss, P., 1996. Ectopic lens w7hrend der Abwesenheit von Augenblasen. Arch. Entwicklungsmech. induction in fish in response to the murine homeobox gene Six3. Mech. 16, 328–339. Dev. 60, 233–239. 14 A. Streit / Developmental Biology 276 (2004) 1–15

Orts-Llorca, F., Jimenez-Collado, J., 1971. Regulation of the embryo after Schwanzel-Fukuda, M., Pfaff, D.W., 1989. Origin of luteinizing hormone- the extirpation of Hensen’s node. Consequences on the differentiation releasing hormone neurons. Nature 338, 161–164. of the otic placode. Arch. Anat. Histol. Embryol. 54, 1–11. Selleck, M.A., Bronner-Fraser, M., 1995. Origins of the avian neural crest: the Pandur, P.D., Moody, S.A., 2000. Xenopus six1 gene is expressed in role of neural plate-epidermal interactions. Development 121, 525–538. neurogenic cranial placodes and maintained in the differentiating lateral Shen, W., Mardon, G., 1997. Ectopic eye development in Drosophila lines [In Process Citation]. Mech. Dev. 96, 253–257. induced by directed dachshund expression. Development 124, 45–52. Panganiban, G., Rubenstein, J.L., 2002. Developmental functions of the Sheng, G., Stern, C.D., 1999. Gata2 and gata3: novel markers for early Distal-less/Dlx homeobox genes. Development 129, 4371–4386. embryonic polarity and for non-neural ectoderm in the chick embryo [In Papalopulu, N., Kintner, C., 1993. Xenopus Distal-less related homeobox Process Citation]. Mech. Dev. 87, 213–216. genes are expressed in the developing forebrain and are induced by Shimeld, S.M., Holland, P.W., 2000. Vertebrate innovations. Proc. Natl. planar signals. Development 117, 961–975. Acad. Sci. U. S. A. 97, 4449–4452. Pera, E., Stein, S., Kessel, M., 1999. Ectodermal patterning in the avian Siggia, S., 1936. Contributi allo studio della determinazione del placode embryo: epidermis versus neural plate. Development 126, 63–73. olfattivo di Discoglossus pictus. Monit. Zool. Ital., 47. Pfeffer, P.L., Gerster, T., Lun, K., Brand, M., Busslinger, M., 1998. Silver, S.J., Davies, E.L., Doyon, L., Rebay, I., 2003. Functional dissection Characterization of three novel members of the zebrafish Pax2/5/8 of eyes absent reveals new modes of regulation within the retinal family: dependency of Pax5 and Pax8 expression on the Pax2.1 (noi) determination gene network. Mol. Cell. Biol. 23, 5989–5999. function. Development 125, 3063–3074. Solomon, K.S., Fritz, A., 2002. Concerted action of two dlx paralogs in Phillips, B.T., Bolding, K., Riley, B.B., 2001. Zebrafish fgf3 and fgf8 sensory placode formation. Development 129, 3127–3136. encode redundant functions required for otic placode induction. Dev. Solomon, K.S., Kudoh, T., Dawid, I.B., Fritz, A., 2003a. Zebrafish foxi1 Biol. 235, 351–365. mediates otic placode formation and jaw development. Development Phillips, B.T., Storch, E.M., Lekven, A.C., Riley, B.B., 2004. A direct 130, 929–940. role for Fgf but not Wnt in otic placode induction. Development 131, Solomon, K.S., Logsdon, J.M., Fritz, A., 2003b. Expression and 923–931. phylogenetic analyses of three zebrafish FoxI class genes. Dev. Pignoni, F., Hu, B., Zavitz, K.H., Xiao, J., Garrity, P.A., Zipursky, S.L., 1997. Dyn. 228, 301–307. The eye-specification proteins So and Eya form a complex and regulate Spemann, H., 1901. U¨ ber Korrelationen in der Entwicklung des Auges. multiple steps in Drosophila eye development [published erratum Verh. der Anat., 15. appears in Cell 1998 Feb 20;92(4): following 585]. Cell 91, 881–891. Stark, M.R., Sechrist, J., Bronner-Fraser, M., Marcelle, C., 1997. Neural Piotrowski, T., Northcutt, R.G., 1996. The cranial nerves of the Senegal tube-ectoderm interactions are required for trigeminal placode forma- bichir, Polypterus senegalus [osteichthyes: actinopterygii: cladistia]. tion. Development 124, 4287–4295. Brain Behav. Evol. 47, 55–102. Stone, L.S., 1931. Induction of the ear by the medulla and its relation to Platt, J.B., 1896. Ontogenetic differentiation of the ectoderm in Necturus. experiments of the lateralis system in amphibia. Science 74, 577. Study II. Development of the peripheral nervous system. Q. J. Nicr. Sci. Street, S.F., 1937. The differentiation of the nasal area of the chick embryo 38, 911–966. in grafts. J. Exp. Zool. 77, 49–80. Porter, F.D., Drago, J., Xu, Y., Cheema, S.S., Wassif, C., Huang, S.P., Lee, Streit, A., 2002. Extensive cell movements accompany formation of the otic E., Grinberg, A., Massalas, J.S., Bodine, D., Alt, F., Westphal, H., 1997. placode. Dev. Biol. 249, 237–254. Lhx2, a LIM homeobox gene, is required for eye, forebrain, and Streit, A., Stern, C.D., 1999. Establishment and maintenance of the border definitive erythrocyte development. Development 124, 2935–2944. of the neural plate in the chick: involvement of FGF and BMP activity. Quinn, J.C., West, J.D., Hill, R.E., 1996. Multiple functions for Pax6 in Mech. Dev. 82, 51–66. mouse eye and nasal development. Genes Dev. 10, 435–446. Streit, A., Berliner, A.J., Papanayotou, C., Sirulnik, A., Stern, C.D., 2000. Reyer, R., 1958a. Studies on lens induction in Ambystoma punctatum and Initiation of neural induction by FGF signalling before gastrulation. Triturus viridescens viridescens. I. Transplants of prospective belly Nature 406, 74–78. ectoderm. J. Exp. Zool. 138, 505–555. Sunkel, C.E., Whittle, J.R.S., 1987. Brista: a gene involved in the Reyer, R., 1958b. Studies on lens induction in Ambystoma punctatum and specification and differentiation of distal cephalic and thoracic Triturus viridescens viridescens. II. Transplants of prospective belly structures in Drosophila melanogaster. Wilhelm Roux’s Arch. Dev. ectoderm. J. Exp. Zool. 139, 137–179. Biol. 196, 124–132. Riley, B.B., Phillips, B.T., 2003. Ringing in the new ear: resolution of cell Torres, M., Giraldez, F., 1998. The development of the vertebrate inner ear. interactions in otic development. Dev. Biol. 261, 289–312. Mech. Dev. 71, 5–21. Rfhlich, K., 1931. Gestaltungsbewegungen der prasumptiven Epidermis Tribulo, C., Aybar, M.J., Nguyen, V.H., Mullins, M.C., Mayor, R., 2003. wahrend der Neurulation und Kopfbildung bei Triton taeniatus. Arch. Regulation of Msx genes by a Bmp gradient is essential for neural crest Entwicklungsmech. Org. 124, 66–81. specification. Development 130, 6441–6452. Romanoff, A.L., 1960. The Avian Embryo. Macmillan, New York. Van Campenhout, E., 1956. Le de´veloppment embryonnaire compare´ Rubel, E.W., Fritzsch, B., 2002. Auditory system development: primary des nerfs olfactif et auditif. Acta Oto-Rhino-Laryngol. Belg. 11, auditory neurons and their targets. Annu. Rev. Neurosci. 25, 51–101. 279–287. Sahly, I., Andermann, P., Petit, C., 1999. The zebrafish eya1 gene and its van Oostrom, C.G., Verwoerd, C.D., 1972. The origin of the olfactory expression pattern during embryogenesis. Dev. Genes Evol. 209, placode. Acta Morphol. Neerl.-Scand., 9. 399–410. Vendrell, V., Carnicero, E., Giraldez, F., Alonso, M.T., Schimmang, T., 2000. Sasai, Y., De Robertis, E.M., 1997. Ectodermal patterning in vertebrate Induction of inner ear fate by FGF3. Development 127, 2011–2019. embryos. Dev. Biol. 182, 5–20. Verwoerd, C.D., van Oostrom, C.G., 1979. Cephalic neural crest and Schlosser, G., 2002. Development and evolution of lateral line placodes in placodes. Adv. Anat. Embryol. Cell Biol. 58, 1–75. amphibians I. Development. Zoology 105, 119–146. von Kupffer, C., 1891. The development of the cranial nerves of Schlosser, G., Ahrens, K., 2004. Molecular of placode develop- vertebrates. J. Comp. Neurol. 1, 246–264, 315–332. ment in Xenopus laevis. Dev. Biol. 271, 439–466. Waddington, C.H., 1937. The determination of the auditory placode in the Schlosser, G., Northcutt, R.G., 2000. Development of neurogenic placodes chick. J. Exp. Biol. 14, 232–239. in Xenopus laevis. J. Comp. Neurol. 418, 121–146. Wakely, J., 1976. Scanning electron microscopy of lens placode invagina- Schook, P., 1980. Morphogenetic movements during the early development tion in the chick embryo. Exp. Eye Res. 22, 647–651. of the chick eye. A light microscopic and spatial reconstructive study. Wallis, D.E., Muenke, M., 1999. Molecular mechanisms of holoprosence- Acta Morphol. Neerl. Scand. 18, 1–30. phaly. Mol. Genet. Metab. 68, 126–138. A. Streit / Developmental Biology 276 (2004) 1–15 15

Walther, C., Gruss, P., 1991. Pax-6, a murine paired box gene, is expressed Xu, P.X., Woo, I., Her, H., Beier, D.R., Maas, R.L., 1997. Mouse in the developing CNS. Development 113, 1435–1449. Eya homologues of the Drosophila eyes absent gene require Wawersik, S., Maas, R.L., 2000. Vertebrate eye development as modeled in Pax6 for expression in lens and nasal placode. Development 124, Drosophila. Hum. Mol. Genet. 9, 917–925. 219–231. Wawersik, S., Purcell, P., Rauchman, M., Dudley, A.T., Robertson, E.J., Xu, P.X., Adams, J., Peters, H., Brown, M.C., Heaney, S., Maas, R., 1999. Maas, R., 1999. BMP7 acts in murine lens placode development. Dev. Eya1-deficient mice lack ears and kidneys and show abnormal Biol. 207, 176–188. apoptosis of organ primordia. Nat. Genet. 23, 113–117. Whitlock, K.E., Westerfield, M., 2000. The olfactory placodes of the Xu, L., Overbeek, P.A., Reneker, L.W., 2002. Systematic analysis of E-, N- zebrafish form by convergence of cellular fields at the edge of the and P-cadherin expression in mouse eye development. Exp. Eye Res. neural plate. Development 127, 3645–3653. 74, 753–760. Wilson, P.A., Hemmati-Brivanlou, A., 1997. Vertebrate neural induction: Yanfeng, W., Saint-Jeannet, J.P., Klein, P.S., 2003. Wnt-frizzled signaling in inducers, inhibitors, and a new synthesis. Neuron 18, 699–710. the induction and differentiation of the neural crest. BioEssays 25, Wilson, R., Mohun, T., 1995. XIdx, a dominant negative regulator of bHLH 317–325. function in early Xenopus embryos. Mech. Dev. 49, 211–222. Yntema, C.L., 1950. An analysis of induction of the ear from foreign Wilson, P.A., Lagna, G., Suzuki, A., Hemmati-Brivanlou, A., 1997. ectoderm in the salamander embryo. J. Exp. Zool. 113, 211–244. Concentration-dependent patterning of the Xenopus ectoderm by Yntema, C.L., 1955. Ear and nose. In: Willier, B.H., Weiss, P.A., BMP4 and its signal transducer Smad1. Development 124, 3177–3184. Hamburger, V. (Eds.), Analysis of Development. Saunders, Philadel- Woo, K., Fraser, S.E., 1995. Order and coherence in the fate map of the phia, pp. 415–428. zebrafish nervous system. Development 121, 2595–2609. Zheng, W., Huang, L., Wei, Z.B., Silvius, D., Tang, B., Xu, P.X., 2003. The Wray, S., Nieburgs, A., Elkabes, S., 1989. Spatiotemporal cell expression of role of Six1 in mammalian auditory system development. Development luteinizing hormone-releasing hormone in the prenatal mouse: evidence 130, 3989–4000. for an embryonic origin in the olfactory placode. Brain Res. Dev. Brain Zwilling, E., 1940a. The determination of the otic vesicle in Rana pipiens. Res. 46, 309–318. J. Exp. Zool. 86, 333–343. Wright, T.J., Mansour, S.L., 2003. Fgf3 and Fgf10 are required for mouse Zwilling, E., 1940b. An experimental analysis of the development of the otic placode induction. Development 130, 3379–3390. anuran olfactory organ. J. Exp. Zool. 84, 291–323. Wu, J., Cohen, S.M., 1999. Proximodistal axis formation in the Drosophila Zygar, C.A., Cook, T.L., Grainger Jr., R.M., 1998. Gene activation leg: subdivision into proximal and distal domains by Homothorax and during early stages of lens induction in Xenopus. Development 125, Distal-less. Development 126, 109–117. 3509–3519.