Ascidian embyogenesis

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 17

DEVELOPMENTAL DYNAMICS 233:1177–1193, 2005

REVIEWS–A PEER REVIEWED FORUM

Specification of Embryonic Axis and Mosaic


Development in Ascidians
Hiroki Nishida*

Setting up future body axes is the first important event before and at the beginning of embryogenesis. The
ascidian embryo is a classic model that has been used to gain insight into developmental processes for over
a century. This review summarizes advances made in this decade in our understanding of the
developmental processes involved in the specification of the embryonic axes and cell fates during early
ascidian embryogenesis. Maternal factors, including mRNAs, are translocated to specific regions of the egg
by cytoplasmic and cortical reorganization, so-called ooplasmic segregation, and specify the animal–vegetal
axis and the one perpendicular to it, which is defined as the anteroposterior axis in ascidians. Some
postplasmic/PEM RNAs that are anchored to cortical endoplasmic reticulum are brought to the future
posterior pole of fertilized eggs, and play crucial roles in posterior development. Following specification of
the animal–vegetal axis, nuclear localization of ␤-catenin takes place in the vegetal blastomeres; this
occurrence is important for the acquisition of the vegetal character of the blastomeres in later development.
Positioning of these maternal factors lead to subsequent cell interactions and zygotic gene expression
responsible for axis establishment and for cell fate specification. We describe how endoderm blastomeres in
the vegetal pole region emanate inductive signals mainly attributable to fibroblast growth factor. Marginal
blastomeres next to endoderm blastomeres respond differently in ways that are determined by intrinsic
competence factors. Expression patterns of developmentally important genes, including key transcription
factors of each tissue type, are also summarized. Developmental Dynamics 233:1177–1193, 2005.
© 2005 Wiley-Liss, Inc.

Key words: embryonic axis; cell fate specification; ascidian; maternal factor; localized mRNA; embryonic induction; cell
signaling; transcription factor

Received 4 January 2005; Revised 14 March 2005; Accepted 28 March 2005

INTRODUCTION ascidian embryo is a classic model 2001; Nishida, 2002a,b; Satoh,


that has been used to study axis and 2003a,b). Most of the recent concepts
The three-dimensional structure of or-
cell fate specification during embryo- and analysis of ascidian development
ganisms is characterized by the three
genesis for over a century (Chabry, derive from observations and experi-
perpendicular body axis, namely, the
anterior–posterior, dorsal–ventral, and 1887; Conklin, 1905; reviewed by Sa- mental works using four main evolu-
left–right axes. These axes generally toh, 1994; Nishida, 1997; Jeffery, tionarily distant species (Ciona intes-
emerge in the early embryonic stage, 2001). Recent advances in ascidian re- tinalis and savignyi, Halocynthia
and are sometimes already apparent search have placed this simple organ- roretzi, and Phallusia mammillata),
in eggs before fertilization. One of the ism in a unique position to contribute which have contributed complemen-
major issues in developmental biology to a detailed understanding of the cel- tary and/or converging or diverging
is to understand how axial polarity is lular and molecular processes of em- information.
achieved in a developing embryo. The bryogenesis (reviewed by Corbo et al., The restriction of cell fate proceeds

Department of Biology, Graduate School of Science, Osaka University, Osaka, Japan


Grant sponsors: MEXT; JSPS; Toray Science Foundation.
*Correspondence to: Hiroki Nishida, Department of Biology, Graduate School of Science, Osaka University, 1-1 Machi-
kaneyama-cho, Toyonaka, Osaka 560-0043, Japan. E-mail: hnishida@bio.sci.osaka-u.ac.jp
DOI 10.1002/dvdy.20469
Published online 22 June 2005 in Wiley InterScience (www.interscience.wiley.com).

© 2005 Wiley-Liss, Inc.


1178 NISHIDA

quickly in ascidian embryos. Just be- point (Arendt and Nübler-Jung, ventral midline at the boundary of
fore gastrulation is initiated at the 1997). In this sense, the definition of trunk and tail of the tadpole (Fig. 2).
110-cell stage, the developmental axes in early embryos is somewhat ar- What happens in the vegetal hemi-
fates of most blastomeres are re- tificial. In studies of ascidians, the an- sphere is not so simple (Fig. 1). Ante-
stricted such that a blastomere gives imal pole, indicated by the site of polar rior-vegetal blastomeres give rise
rise to a single cell type in the tadpole body extrusion, is always held up- mainly to endoderm, notochord, and
larva (Fig. 1; Nishida, 1987). Even ward. As axial structures such as no- nerve cord. Notochord and nerve cord
when blastomeres are isolated from tochord and nerve cord undergo con- cells are located in the larval tail. Pos-
110-cell embryos, they continue to de- vergent and extension movement, and terior-vegetal blastomeres develop
velop into differentiated cells accord- thus the dorsal blastopore lip of ascid- mainly into endoderm, mesenchyme,
ing to the fate map (Reverberi and ian gastrulae heads vegetally, the and muscle. Muscle cells are found in
Minganti, 1946; Nishida, 1992a). This original vegetal pole is covered by the tail, while endoderm and mesen-
intriguing cell-autonomous property these dorsal tissues. Thus, the vegetal chyme cells are found in the trunk
of ascidian embryos is referred to in pole corresponds to the future dorsal region. In the present paper, “ante-
many textbooks as a typical example side in ascidians. Looked at this way, rior” and “posterior” are used in the
of mosaic development. Therefore, it embryos lie in a somewhat unusual way these adjectives are traditionally
will be sufficient to take into account way on their back. This finding may applied to ascidian eggs and early em-
the events that happen before the 110- not matter to ascidian researchers, be- bryos. Thus, in the present context,
cell stage to fully understand axis cause ascidian embryos develop in any “anterior fate” is that of blastomeres
specification in this animal. This arti- orientation in seawater like sea ur- of the anterior half of cleavage-stage
cle summarizes the current knowl- chin embryos. In contrast, in a usual embryos and does not mean the fate
edge on maternal and zygotic pro- textbook drawing of amphibian gas- that gives rise to the anterior trunk
cesses related to embryonic axis trulation (e.g., Gilbert, 2003), the dor- structures of larvae, nor does “poste-
specification in ascidians. In this arti- sal blastopore lip is held in a constant rior fate” mean the fate that gives rise
cle, we first describe postfertilization position. In this presentation, one to the posterior tail structures.
processes involving maternal factors holds the region of dynamic cell move-
before zygotic gene expression is initi- ment and actually holds no cell POLARITY IN
ated. Then processes that require zy- constantly. With this as a fixed point,
gotic gene expression and cellular in- it is necessary to turn other regions
UNFERTILIZED EGGS
teractions after embryos become around such as the regions with Generally, the A-V axis is already
multicellular will be summarized. less dynamic cell movement, for exam- present in unfertilized eggs. The axis
ple, animal pole cells. This presenta- in pregastrula embryos is correlated
DEFINITION OF AXES IN tion makes sense in light of the fact with germ layer segregation. In as-
that amphibian embryos settle and cidians, ectoderm, mesoderm, and
EARLY EMBRYOS are oriented by gravity. When one endoderm territory is present along
The definition of embryonic axis in draws gastrulating embryos of either the A-V axis, as in amphibian (Fig. 1).
early embryos is somewhat artificial animal in another way, one can see Processes that involve maternally
and is a question of semantics. It is how morphogenetic movements are stored factors and occur before the ac-
difficult per se to superpose future em- conserved between ascidians and am- tivation of the zygotic genome are dis-
bryonic axes on eggs and embryos be- phibians. cussed in this and the following two
fore and during gastrulation, because In ascidians, the A-P axis of the chapters. The mature oocytes of most
dynamic movements and rearrange- early embryo (corresponding to the animals show radial symmetry with a
ments of cells occur in gastrulae. Dif- D-V axis in amphibians) is defined as single A-V axis. The animal pole is
ferences in axis definition between perpendicular to the A-V axis, as the where polar bodies form during meio-
amphibians and ascidians (which animal pole represents the reference sis. Unfertilized eggs of ascidians are
show similar morphogenetic move- point. But it does not correspond pre- in meiotic metaphase I with the mei-
ment) provide an example of the diffi- cisely to the A-P axis of larvae, be- otic spindle at the animal pole. In ad-
culties (Fig. 2). In ascidians, the ani- cause cells do not remain stationary dition to the spindle, ascidian eggs
mal–vegetal (A-V) axis conventionally during morphogenetic movements. already show various kinds of polar-
has been considered to be the future Complicated positional rearrange- ization along the A-V axis in the sub-
ventral– dorsal (V-D) axis, and the ment does not take place in the animal cortical and cortical domains (Fig. 3).
axis perpendicular to the A-V axis is hemisphere, and animal cells just No remarkable polarization in the
the anterior–posterior (A-P) axis cover the embryo to form ectoderm by central egg cytoplasm has been re-
(Conklin, 1905). By contrast, in Xeno- epiboly, so the A-P axis of eggs and ported. In the subcortical region, mi-
pus, the definition of embryonic axis is early embryos in the animal hemi- tochondria are enriched in the vegetal
the opposite. The A-V axis is assigned sphere corresponds precisely to the half forming a mitochondria-rich re-
as the future A-P axis, and the D-V A-P axis of larvae. Anterior-animal gion, called the myoplasm, from which
axis is perpendicular to it (Nieuwk- blastomeres give rise to head/trunk microtubules are excluded (Zalokar
oop, 1977). The difference comes from epidermis of larvae, and posterior-an- and Sardet, 1984). The localization of
the difference in the position of the imal blastomeres develop into tail epi- mitochondria in myoplasm would
developing embryo used as a reference dermis. The polar bodies attach to the have an adaptive significance in that
AXIS SPECIFICATION IN ASCIDIANS 1179

Fig. 1. Developmental fates of blastomeres of the ascidian embryo.


A: Fate restriction during cleavage stages. Blastomeres are colored
when the fate of the blastomere is restricted to give rise to a single kind
of tissue cell. The colors correspond to the colors of larval tissues
indicated in C. Fate restriction in ascidian embryos proceeds quickly in
the early embryo. Sister blastomeres of the previous cleavage are con-
nected by bars. B: Fate map of the 110-cell stage. Animal and vegetal
hemispheres. Names of blastomeres are indicated as “a8.19” and so on.
C: Organization of tail bud embryos. Mid-sagittal planes, sagittal planes,
and transverse sections of the tail. TCL, trunk lateral cell; TVC, trunk
ventral cell.

they are preferentially segregated into


larval muscle cells, which need vigorous
energy production. In the egg cortex,
microfilaments form a basket with its
opening at the animal pole region (Sar-
det et al., 1992). The microfilaments
gives a mechanical motive force for the
first phase of cytoplasmic and cortical
reorganization, called ooplasmic segre- Fig. 2. Schematic representation of morphogenetic movements during
gation, as mentioned two paragraphs gastrulation of ascidians (left) and frogs (right). Presumptive notochord
region is colored pink. Anterior and posterior neural tube is colored light
below. A network of tubules and sheets and dark blue. Polar bodies indicate the animal pole. Green asterisks
of cortical endoplasmic reticulum (cER) point the original vegetal pole. The red arrowhead represents the point
is present just beneath the plasma held in a constant position. Embryonic axes at tail bud stage are indi-
membrane, showing a gradient with the cated at the bottom. See text in detail.
highest density at the vegetal pole. The
cER is tethered to the plasma mem- ternal mRNAs, called type I postplas- 1996), show a polarized distribution
brane (Sardet et al., 1992). mic RNAs in Halocynthia (Sasakura with the highest concentration at the
In addition to these membranous and et al., 2000) or PEM (posterior end vegetal pole cortex (Fig. 3) (reviewed
cytoskeletal components, several ma- mark) RNAs in Ciona (Yoshida et al., in Nishida, 2002a; Sardet et al., 2005).
1180 NISHIDA

Fig. 3. Localization of maternal factors from fertilization to the 110-cell stage. Cytoplasmic and cortical reorganization (ooplasmic segregation)
proceeds in two major phases during the first cell cycle. The centrosome-attracting body (CAB) is present in the posterior region of the B4.1
blastomeres of the eight-cell embryos and is inherited by the posterior-most blastomeres during cleavages. Cortical endoplasmic reticulum (cER) and
postplasmic/PEM RNAs are concentrated into the CAB. The bottom part shows localization of various maternal determinants as inferred from ooplasmic
removal and transplantation experiments. Nuclear localization of ␤-catenin during cleavages is shown at the bottom right.

These type I postplasmic/PEM RNAs tion in the unfertilized egg cortex, but clearly involved in the formation of
encode a great variety of protein their role in the establishment of the the A-P axis, as their final destination
types. They show a polarized distribu- A-V axis is unclear. Some of them are after ooplasmic movements is the pos-
AXIS SPECIFICATION IN ASCIDIANS 1181

terior region, as discussed in the next dria, cER, postplasmic/PEM RNAs, amphibian and fish embryos, this ma-
section. The results of transplantation and muscle- and endoderm-forming ternal system specifies the D-V axis,
of the peripheral cytoplasm have also activity toward the vegetal pole region which is perpendicular to the A-V axis
revealed the polarized distribution of (Roegiers et al., 1999; Nishida, 1997; (Miller and Moon, 1996; Heasman,
tissue-forming substances in unfertil- Sardet et al., 2003). A microfilament 1997; Schneider et al., 1996; Miller et
ized eggs. Muscle and endoderm form- basket with its opening at the animal al., 1999). However, the same path-
ing activities are present in a gradient pole contracts toward the vegetal pole, way is used to establish the A-V axis
manner with the highest activity at bringing these components toward the in echinoderms (Wikramanayake et
the vegetal pole, while epidermis vegetal pole region. Eggs still show al., 1998; Emily-Fenouil et al., 1998;
forming activity is present in an oppo- radial symmetry along the A-V axis at Logan et al., 1999; Weitzel et al.,
site gradient (Nishida, 1992b, 1997, this stage, but the original polarity is 2003). The role of this pathway in
and references therein; Fig. 3). Thus, much intensified, as all components germ layer segregation would be con-
unfertilized eggs already show appar- mentioned above are highly concen- served among ancient metazoans, as
ent polarity; however, when and how trated at the vegetal pole region (Fig. Cnidaria embryos also shows nuclear
the polarity is established during oo- 3). In Halocynthia, the contraction ␤-catenin in cells that invaginate
genesis and oocyte maturation is un- pole is at the opposite position of the (Wikramanayake et al., 2003). In the
known. In Halocynthia oocytes, the polar bodies in most cases. However, canonical Wnt signaling pathway,
germinal vesicle is in an eccentric po- in Phallusia and Ciona, the contrac- Wnt proteins bind to the transmem-
sition (Numakunai, 2001), but this tion pole can be off the opposite posi- brane receptor Frizzled and then acti-
finding does not seem to be the case in tion by as much as 45– 60 degrees, vate Dsh, which in turn inhibits
Ciona and Phallusia oocytes. depending on the site of the sperm GSK-3 activity. GSK-3 destabilizes
entry (Roegiers et al., 1995), making it ␤-catenin protein. Hence, inhibition of
POSTFERTILIZATION difficult to define the A-V axis unam- GSK-3 activity results in ␤-catenin
biguously. stabilization and its translocation into
PROCESSES THAT
The second phase of reorganization nuclei. ␤-catenin forms a complex
ESTABLISH THE ANIMAL (85–110 min in Halocynthia, 25– 45 with the transcription factor Tcf to ac-
VS. VEGETAL CELL FATES min in Ciona and Phallusia) brings tivate specific gene expression (Cadi-
Cytoplasmic and Cortical most of these components toward the gan and Nusse, 1997). In the sea ur-
Reorganization During the
future posterior pole (Fig. 3; Sawada, chin, nuclear accumulation of ␤-catenin
1988; Sardet et al., 1989; Nishida, occurs in the vegetal region of cleaving
First Cell Cycle 1997; Roegiers et al., 1999) and is rel- embryos, and this occurrence is a cru-
Eggs of many kinds of animal show evant to specification of the A-P axis. cial event in endomesoderm (vegetal
cortical and cytoplasmic movements Exceptionally, factors involved in cell fates) specification. Dsh is re-
just after fertilization. These pro- endoderm formation and gastrulation quired for the stabilization of ␤-cate-
cesses have been extensively analyzed do not move toward the future poste- nin in vegetal cells. Green fluorescent
in Caenorhabditis elegans, Xenopus, rior pole and just expand their distri- protein–tagged Dsh protein is tar-
and ascidians. The mechanisms in- bution to the entire vegetal hemi- geted to the vegetal cortex of the un-
volved and factors translocated are di- sphere during the second phase (Fig. fertilized egg (Weitzel et al., 2003).
verged among these animals; how- 3; Bates and Jeffery, 1987; Nishida, Therefore, Dsh seems a key localized
ever, egg cortices commonly play a 1996). The significance of this second protein, although the authors argue
significant role. The cortex provides a phase in the A-P axis specification is the possibility that Dsh activator is
scaffold to anchor and translocate lo- discussed in the next chapter. also localized in the vegetal cortical
calized maternal factors, and supplies region, because overexpressed Dsh
spatial information for later embryo- ␤-Catenin and Vegetal Cell mRNA cannot promote vegetal fate in
genesis (reviewed by Sardet et al., animal blastomeres.
1994, 2002). Ascidian eggs undergo
Fate Specification In ascidians, Imai et al. (2000) re-
dramatic cytoplasmic and cortical re- Recently, molecular aspects of postfer- ported a role for ␤-catenin in vegetal
organizations between fertilization tilization processes that is involved in fate specification in Ciona intestinalis
and the beginning of the first cleav- establishment of the A-V axis in sea and C. savignyi embryos. In this re-
age; this process has been called oop- urchin embryos have been investi- gard, ascidian embryos show a simi-
lasmic segregation (Fig. 3; Conklin, gated intensively. Dishevelled (Dsh), larity to echinoderm embryos. Prefer-
1905; Sardet at al., 1989; Roegiers et glycogen synthetase kinase 3 (GSK-3), ential ␤-catenin nuclear localization
al., 1999). Reorganizations occurs in and ␤-catenin are involved. These occurred in the vegetal hemisphere in
two major phases. Shortly after fertil- molecules are signaling molecules in cleavage-stage embryos (Fig. 3). When
ization (0 –10 min after insemination the canonical Wnt pathway. The Wnt mRNA encoding the stabilized form of
in Halocynthia, 0 –5 min in Ciona and signaling pathway is generally used in ␤-catenin (lacking the phosphoryla-
Phallusia), the first phase of ooplas- cellular communication. However, tion sites for GSK-3) was injected into
mic segregation is accompanied by a early embryos use only a downstream eggs, nuclear ␤-catenin was also ob-
rapid contraction of the egg cortex and part of the intracellular signal trans- served in the animal hemisphere. In
plasma membrane, resulting in a seg- duction cascade in maternal mecha- these embryos, most embryonic cells
regation of microfilaments, mitochon- nisms that specify embryonic axes. In produced alkaline phosphatase, an
1182 NISHIDA

endoderm differentiation marker, present as triplicate copies) in the ge- Role of the Posterior-Vegetal
and the expression of an epidermis nome, and none of their maternal Cytoplasm/Cortex and
marker, Cs-Epi1, was down-regu- mRNAs are localized to the vegetal Postplasmic/PEM RNAs
lated. The only exception concerned pole (Takatori et al., 2004). This find-
primary muscle precursor cells, whose ing is consistent with the observation The posterior-vegetal cytoplasm/cor-
fate was not altered by the overpro- that mechanisms involved in specifica- tex (PVC) after the second phase of
duction of ␤-catenin. To inhibit ␤-cate- tion of vegetal fates may differ between segregation and before the first cleav-
nin function in nuclei, ␤-catenin was echinoderms/ascidians and amphibi- age plays crucial roles in specification
sequestered into a cell adhesion com- of the A-P axis. Removal of the PVC
ans.
plex by overproduction of cadherin. In (less than 10% of total egg volume)
these embryos, nuclear staining with results in radialization of the cleavage
␤-catenin antibody was abolished in pattern, producing a mirror image du-
the entire embryo. Alkaline phospha-
POSTFERTILIZATION plication of the anterior cleavage pat-
tase expression was lost, and Cs-Epi1 PROCESSES THAT SPECIFY tern in the original posterior half. It
expression expanded into the vegetal THE ANTEROPOSTERIOR has an effect not only on the cleavage
hemisphere. The embryos did not gas- AXIS pattern, but also on the developmen-
trulate. The development of muscle tal fates of blastomeres, as they also
cells was not perturbed in these cad- Cortical Movements show mirror image duplication of the
herin-overproducing embryos. Nei- Unfertilized eggs and eggs after the anterior half. Transplantation of the
ther ␤-catenin mRNA nor protein was first phase of ooplasmic segregation PVC to the anterior region brings
localized to the vegetal cytoplasmic re- are radially symmetrical along the about mirror image posteriorization of
gion of eggs and initial cleavage stage the original anterior half (Nishida,
A-V axis. This symmetry is broken
embryos. This finding suggests that 1994). It is worth noting that removal
during the second phase of ooplasmic
the localized endoderm determinants and transplantation of regional egg
segregation, and eggs become bilater-
may be molecules that stabilize cytoplasm other than the PVC has no
ally symmetrical before the first cleav-
␤-catenin in the vegetal hemisphere. effect on embryogenesis. These micro-
age, as cortical and subcortical compo- manipulative experiments demon-
A promising candidate is Dsh, as has
nents, such as postplasmic/PEM RNAs strate that some important factors are
been shown recently in sea urchins.
Identification of maternal endoderm that are located in the vegetal pole localized to the PVC and that no de-
determinants and gastrulation deter- region after the first phase, move to- velopmentally important factor is lo-
minants in ascidians remains elusive ward the future posterior pole (Fig. 3). calized to any small area other than
for the moment. This movement is directed by the cen- the PVC. During the second phase,
These observations indicate that trosome introduced by the sperm and cER and subcortical mitochondria re-
A-V axis establishment is mediated by depends on the sperm entry point locate posteriorly (Fig. 3). In contrast,
␤-catenin signaling. Endoderm (vege- (Sardet at al., 1989; Roegiers et al., the plasma membrane and cortical ac-
tal) fate requires ␤-catenin function, 1995). In Phallusia, the sperm tends tin cap do not move. In the past de-
whereas epidermis (animal) fate is to bind to the egg surface at the ani- cade, several maternal mRNAs that
promoted by suppressing ␤-catenin mal hemisphere (Speksnijder et al., are localized to the PVC have been
function. It is not known whether me- 1989) and is conveyed partway toward identified. The first example is the
soderm cells in the marginal zone of the vegetal pole when the plasma pem (posterior end mark) mRNA in
the vegetal hemisphere, such as noto- membrane and cortex contracts dur- Ciona savignyi, which was isolated us-
chord and mesenchyme precursors, ing the first phase, which occurs ing differential screening techniques
require intermediate stabilization of shortly after fertilization (Fig. 3). In with egg fragments prepared by cen-
␤-catenin within the cells or not. In contrast to this first phase, which is trifugation (Yoshida et al, 1996). Since
sea urchin, there is a gradient in driven by microfilaments, the motive then, nine mRNAs localized to the
␤-catenin stability along the A-V axis power for the second phase is princi-
PVC have been identified in Halocyn-
during early cleavage, and this gradi- thia roretzi (macho-1, Hr-PEM, Hr-
pally generated by microtubules of the
ent could be involved in specifying an PEM-3, Hr-Wnt-5, Hr-POPK-1, Hr-
sperm aster (Sawada and Schatten,
intermediate zone along the A-V axis GULT, Hr-PEN-1, Hr-PEN-2, Hr-
1988). Vegetally located cortical com-
(Weitzel et al., 2003). In ascidians, the ZF-1; reviewed in Nishida, 2002a;
ponents are then brought to the future
presence of such a gradient has not Sardet et al., 2005) by the collabora-
yet been investigated. posterior pole together with the sperm tive MAGEST project (cDNA project
In Xenopus, maternal VegT mRNA aster, which goes back just beneath of maternal mRNAs: http://www.
is localized to the vegetal pole of the the plasma membrane toward the genome.jp/magest/; Sasakura et al.,
egg. VegT encodes a T-box transcrip- equatorial region during the second 1998a,b, 2000; Makabe et al., 2001;
tion factor and is required for specifi- phase. The male pronucleus and Nakamura et al., 2003) and by sub-
cation of the endomesoderm germ sperm aster never cross or go to the traction hybridization screening be-
layer in the vegetal hemisphere opposite side of the vegetal pole; tween animal and vegetal blastomeres
(Zhang et al., 1998). However, in therefore, the future posterior side co- (Nishida and Sawada, 2001). These
Ciona, there are seven T-box tran- incides with the side of sperm entry RNAs have been called type I postplas-
scription factor genes (one of these is point. mic RNA in Halocynthia (Sasakura et
AXIS SPECIFICATION IN ASCIDIANS 1183

al., 2000). They are present in cortex of blastomeres (Kobayashi et al., 2003), during early cleavages (Sardet et al.,
the PVC. Some of them (macho-1, Hr- as discussed later. Thus macho-1 lo- 2003). During three rounds of succes-
PEM) have been shown to be anchored calization supports central roles in the sive unequal cleavages, the mRNAs
to and segregate with the cER (Sardet specification of cell fates of the poste- are segregated into the smaller
et al., 2003). The cER network present rior blastomeres. daughter cells located at the posterior
in unfertilized egg provides a scaffold to pole together with the CAB (Fig. 3).
anchor these maternal RNAs, and the Unequal cleavage. Thus, the CAB serves as the core
second phase of ooplasmic segregation structure of a multifunctional complex
brings this cER/mRNA domain into an The centrosome-attracting body (CAB) that operates cleavage planes and an-
asymmetric position along the A-P axis. was originally discovered as a subcel- chors postplasmic RNAs. Having both
Localization sequence elements of some lular structure involved in generation functions together, the CAB ensures
postplasmic RNAs were identified in of unequal cleavages (Hibino et al., that postplasmic RNAs are infallibly
their 3⬘-untranslated regions (Sasakura 1998; reviewed in Nishida et al., partitioned into one of the daughter
and Makabe, 2002). Every type I post- 1999). During unequal cleavages in cells after cell divisions.
plasmic mRNA shows identical regional the posterior-most blastomeres, mi-
localization during ooplasmic segrega- crotubule arrays extending from the Formation of primordial germ
tion and cleavage stages (Fig. 3). They posterior centrosome focus on the
cells.
encode variety of protein such as tran- CAB, which is present just beneath
scription factors, secreted signaling pro- plasma membrane at the posterior An electron microscopic study of 16-
tein, kinase, RNA-binding protein, and pole of the cleaving embryo. Then, as cell embryos revealed that the CAB is
proteins with no obvious similarity to the microtubule arrays shorten, the also characterized by an electron-
known proteins. interphase nucleus with the centro- dense matrix that resembles the germ
What are the functions of type I some shifts posteriorly and ap- plasm in other animals (Iseto and
postplasmic/PEM RNAs? Removal proaches the CAB. Consequently, an Nishida, 1999). A pair of the posterior-
and transplantation of the PVC of fer- asymmetrically located mitotic appa- most and smallest blastomeres (B7.6
tilized eggs, where type I postplasmic ratus is formed. Unequal division then cell pair, brown in Fig. 1) of the 64-cell
RNAs are localized, and other obser- takes place, producing a smaller embryos that inherit the CAB cease to
vations of events unique to the posterior daughter cell that inherits the CAB is divide during early embryogenesis
region of developing embryos indicate the posterior-most position in the veg- and give rise to two cells in the
the following possible functions. The etal half of the 16-cell embryo. This endodermal strand of the tadpole
suppression of functions of some of the process takes place three times from (Nishida, 1987). In the posterior-most
postplasmic RNAs by antisense oligo- the 8- to 64-cell stages, resulting in blastomeres, zygotic gene expression
nucleotides supports their roles in pos- three successive unequal cleavages is repressed (Tomioka et al., 2002), as
terior specification. only at the posterior pole. These un- observed in Drosophila and C. elegans
equal cleavages make the cleavage germline cells (Seydoux and Dunn,
Muscle specification. pattern of the posterior-vegetal region 1997; Van Doren et al., 1998). The ma-
unique, and different from the radial ternal vasa (Ci-DEAD1) mRNA is con-
A recent molecular study identified anterior cleavage pattern. The CAB is centrated in the B7.6 cells in Ciona,
macho-1 mRNA as the localized ma- not present in eggs. The PVC of fertil- and vasa protein also appears in the
ternal determinant of muscle forma- ized eggs is required and sufficient for descendant cells at tail bud stage (Fu-
tion in ascidians (Nishida and the formation of the CAB and conse- jimura and Takamura, 2000; Taka-
Sawada, 2001; Satou et al., 2002a). quent unequal cleavages (Nishikata et mura et al., 2002). After metamorpho-
macho-1 mRNA is a type I postplas- al., 1999). Results of knockdown of Hr- sis, Vasa-positive cells are found in
mic RNA, and encodes a Zn-finger pro- PEM mRNA function in Halocynthia the developing gonad. Vasa is a well-
tein that is transcriptional activator suggest that Hr-PEM is involved in the known marker of germ cells in many
(Sawada et al., 2005). Without ma- positioning of cleavage planes and un- animals (Ikenishi, 1998). These obser-
cho-1 function, muscle blastomeres equal cleavages (T. Negishi, K. Sawada, vations support the view that B7.6
assume nerve cord fate (Fig. 1; Koba- and H.N., unpublished observations). cells are primordial germ cells in as-
yashi et al., 2003). cidians and that the CAB, enriched in
Concentration of postplasmic/ postplasmic/PEM RNAs, contains
Responsiveness to inductive germ plasm.
PEM RNAs into the CAB.
signals.
Of interest, all postplasmic RNAs are Specification of trunk ventral
macho-1 is also involved in generating highly concentrated into a very re- cell precursors.
difference in responsiveness to induc- stricted posterior region of the poste-
tive signals between notochord and rior-vegetal (B4.1) blastomere pair by Precursor blastomeres of trunk ven-
mesenchyme precursor blastomeres. the eight-cell stage. This region is in- tral cells (B7.5 cells, sister cells of
macho-1 confers responsiveness to in- deed the CAB. The CAB is rich in ER B7.6 of the 64-cell embryo, purple in
duction of mesenchyme fate by extra- (Iseto and Nishida, 1999), and cER Fig. 1) form in the posterior region of
cellular fibroblast growth factor (FGF) bearing postplasmic RNAs in eggs is early embryos (Nishida, 1987). Trunk
signal on presumptive mesenchyme likely to be concentrated into the CAB ventral cells of tadpole larvae are pro-
1184 NISHIDA

TABLE 1. Key Transcription Factors for Tissue Formation in Ascidian Embryos*

DNA-binding
Tissue Gene domain Reference
Endoderm Lhx3 Homeobox Satou et al. [2001a]
TTF1 Homeobox Ristoratore et al. [1999]
Muscle Tbx6 T-box Mitani et al. [1999]
Notochord Brachyury T-box Yasuo and Satoh [1998]
Mesenchyme Twist-like1 bHLH Imai et al. [2003]
Trunk lateral cells Hand-like/NoTrlc bHLH Imai et al. [2003]
Trunk ventral cells Mesp bHLH Satou et al. [2004]
Brain Otx? Homeobox Wada et al. [2004]
Nerve cord ?
Epidermis ?

*Expression pattern of each transcription factor is indicated in Fig. 4.

genitors of body wall muscle and heart ZYGOTIC GENE One group represents genes that are
of metamorphosed juveniles (Hirano EXPRESSION THAT involved in endoderm formation
and Nishida, 1997; Davidson and Le- (FoxA5, Lhx3, TTF1). The second group
ESTABLISHES THE ANIMAL
vine, 2003). Recently, it was revealed includes genes that are relevant to em-
VS. VEGETAL CELL FATES bryonic induction of mesodermal tis-
that a Mesp ortholog in ascidians is
specifically and transiently expressed Zygotic Genes Downstream sues (Fgf9/16/20, chordin, FoxD, Zic,
in B7.5 cells (Fig. 4) and is essential of ␤-catenin Brachyury, twist-like1, Mesp). The
for the specification of heart precursor genes in the second group are discussed
When embryos become multicellular, in the next chapter. Third group con-
cells (Satou et al., 2004). Maternal
cell interactions and zygotic gene ex- tains genes whose functions have not
macho-1 at least is required for Mesp
pression start. So far, the earliest zy- been fully analyzed in ascidians (cad-
expression. Other postplasmic/PEM
gotic expression in ascidian has been herinII, protochadherin, Eph, lefty, dkk,
RNAs might be involved in the speci-
observed from the eight-cell stage. DMRT1, hairy, ELK, Fli, jun, msxb,
fication of the trunk ventral cells and
Then expression of various genes etc.). Essentially, the expression of
zygotic expression of Mesp gene.
starts at the 32-cell stage, correspond- these genes starts during cleavage
ing to the mid-blastula stage in ascid- stages in the vegetal hemisphere.
Control of cell cycle length. ians (e.g., Miya and Nishida, 2002).
Cell divisions are retarded in the pos- Several key transcription factors that
terior-vegetal blastomeres compared show tissue-specific zygotic expression Zygotic Genes for
with blastomeres in the other regions and are essential for the formation of Gastrulation and Endoderm
(Conklin, 1905). Postplasmic/PEM each tissue have been identified in as- Fate Specification
RNAs might be also involved in this cidians in this decade, as described The FoxA5 (former forkhead/HNF3␤)
retardation of cell cycle. below (Table 1). gene is expressed in vegetal blas-
Nuclear ␤-catenin, together with tomeres as early as the 16-cell stage
These events that are unique to the transcription factor Tcf, promotes zy-
posterior region and have not been (Mocu-FH1, Olsen and Jeffery, 1997;
gotic expression of various genes in Hr-HNF3, Shimauchi et al., 1997; Ci-
well analyzed yet, could be attributed the vegetal hemisphere. Genes down- fkh, Di Gregorio et al., 2001). Its expres-
to the postplasmic RNAs that also stream of ␤-catenin have been sur- sion is rather broad in the vegetal hemi-
have not been analyzed. It is not veyed in Ciona by subtraction between sphere (Fig. 4). When antisense oligo
known whether the A-P polarity of embryos overproducing ␤-catenin DNA interfered with the function of Mo-
eggs is accounted for only by the and those overproducing cadherin cuFH1, gastrulation movements were
asymmetric distribution of the post- (Satou et al., 2001a; Imai, 2003) and inhibited (Olsen and Jeffery, 1997). Shi-
plasmic/PEM RNAs, although the fol- by comparison of expression levels by mauchi et al. (2001a) suggested a role of
lowing discussion supports this possi- real-time reverse transcriptase-PCR FoxA5 also in notochord formation.
bility. Several postplasmic RNAs have for every transcription factor and sig- Endoderm precursors are formed
been found as described above, but no naling molecule in the Ciona genome from vegetal pole blastomeres (Fig. 1).
maternal mRNA localized to other re- in early gastrulae injected with a At the 32-cell stage, expression of a
gions of eggs has been reported so far. ␤-catenin morpholino antisense oligo- LIM-class homeobox gene, Lhx3, starts
This finding coincides with the results nucleotide (MO) (Imai et al., 2004). (Wada et al., 1995). Expression of the
of removal and transplantation of egg The downstream genes thus identified gene is not strictly restricted to
cytoplasm other than the PVC. can be classified into three groups. endoderm blastomeres at first, but then
Fig. 4. Zygotic gene expression of various developmentally important genes during cleavage stages. Expression of each gene is indicated by blue
dots on blastomeres. The light colors of blastomeres correspond to the colors used in the fate map shown in Figure 1. There are some differences
in the expressions of Tbx6, snail, and Zic genes between Ciona and Halocynthia, although the differences are trivial and not relevant to the discussion
in the review.
1186 NISHIDA

becomes restricted to endoderm blas- vegetal hemisphere shows high com- Directed Signaling and
tomeres by the 110-cell stage (Fig. 4). plexity and remarkable asymmetry Asymmetric Division
Satou et al. (2001a) showed that Lhx3 is along the A-P axis (Fig. 1). The mater-
required and sufficient for expression of nal factors in the PVC play crucial Then asymmetric cell divisions occur
late marker genes of endoderm differen- roles in making this region asymmet- in the marginal blastomeres. These
tiation. Overexpression of Lhx3 pro- blastomeres divide radially so that
ric. In addition, inductive cell interac-
motes ectopic endoderm formation two lines of blastomeres encircle the
tions contribute to generating cell di-
without ␤-catenin activity. Therefore, endoderm blastomeres at the 64-cell
versity in this area. The importance of
Lhx3 would be a key zygotic transcrip- stage (Fig. 1). Four anterior blas-
cell interactions has been revealed by
tion factor for endoderm formation. At tomeres of the 32-cell embryo divide
blastomere isolation and recombina-
the 64-cell stage, the expression of into four outer nerve cord precursors
tion and with inhibitors of cell signal-
TTF1 is initiated exclusively in (colored pale blue in Fig. 1) and four
ing (reviewed by Nishida, 2002a,b and
endoderm precursors (Fig. 4). TTF1 is a inner notochord precursors (pink) of
references therein). Thus, it is now re- the 64-cell embryo. (The central ner-
transcription factor containing an NK-
alized that the development of ascid- vous system of ascidian tadpole larvae
2–like homeodomain. When synthetic
ian embryos is not entirely mosaic as consists of an anterior brain in the
mRNA of TTF1 is injected into eggs,
has been hitherto thought. Key terms cranial region and a posterior nerve
ectopic endoderm cells are formed (Ris-
to understand what happens in this cord in the trunk and tail region [Fig.
toratore et al., 1999; Satou et al.,
area are “FGF signaling,” “directed 1]. Posterior-nerve cord precursors are
2001a). However, injection of its MO did
not affect endoderm differentiation (Sa- signaling and asymmetric division,” blastomeres in the vegetal hemi-
tou et al., 2001a), although the effi- and “cell responsiveness.” sphere and give rise to the posterior
ciency of the MO needs to be confirmed. neural tube in the trunk and tail re-
GATA3/4/5 and Sox17 genes have been gion.) Similarly in the posterior re-
FGF Signaling gion, four blastomeres of the 32-cell
reported to play important roles in
endoderm formation in various metazo- In the 32-cell embryos, central embryo divide into four outer muscle
ans (Laverriere et al., 1994; Hudson et endoderm blastomeres are flanked by precursors (red) and four inner mes-
al., 1997). However, these genes do a single circle of marginal blastomeres enchyme precursors (dark green). In
not seem to be significant in early (Fig. 1). The endoderm blastomeres these processes, outer nerve cord and
endoderm specification processes in emit an inductive signal toward these muscle fates represent the default
ascidians to date. Thus, ␤-catenin nu- marginal cells. FGF (an Fgf9/16/20 state and inner notochord and mesen-
clear localization promotes expression gene product) plays a major role as the chyme fates represent the induced
of some zygotic genes that are in- signaling molecule (Nakatani et al., fates. When the relevant blastomeres
volved in gastrulation and specifica- 1996; Kim et al., 2000; Satou et al., are isolated before induction at the
tion of endoderm precursors. 2002c; Imai et al., 2002a). Treatment 32-cell stage or treated with signaling
In the animal hemisphere, various of isolated blastomeres with recombi- inhibitors, both daughters of them as-
genes also start zygotic expression dur- nant FGF protein efficiently mimics sume the default fate. In contrast,
ing cleavage stages (e.g., Epi-C in Fig. 4; when isolated blastomeres are treated
the normal cell responses. Fgf9/16/20
Ishida et al., 1996; Miya and Nishida, with FGF dissolved in seawater, both
MO suppresses the inductions. Ex-
2002). However, much attention has not daughters take the induced fate (Kim
pression of the Fgf9/16/20 gene starts
been paid to the mechanisms of fate et al., 2000; Minokawa et al., 2001).
at the 16-cell stage (Fig. 4) and occurs
specification of epidermis cells. Thus, A conspicuous feature of these in-
downstream of ␤-catenin (Imai et al.,
the relationship of these zygotic genes ductions is the asymmetric cell divi-
2002a). Thus, acquisition of the induc-
to ␤-catenin nuclear localization and sions of the cells that received the in-
ing ability depends on the position of
their functions in epidermal develop- ductive signal. On the basis of the
blastomeres along the A-V axis. The
ment have not been analyzed. Key tran- results described above, it is possible
scription factors in epidermal formation inductive interactions take place at to imagine the following scenario. In
are not yet known. the 32-cell stage. Results of experi- normal embryos, the precursor cells
ments involving recombination of iso- of the 32-cell embryo receive an
lated blastomeres at various stages endoderm signal from the vegetal
CELL INTERACTIONS AND and of experiments determining the pole, and only one of the daughter
ZYGOTIC GENE periods of sensitivity to FGF treat- cells facing the endoderm assumes an
EXPRESSION ALONG THE ment and to signaling inhibitors all induced cell fate in the 64-cell embryo.
ANTEROPOSTERIOR AXIS support the idea that major processes Directed signals that emanate from
of induction take place at the 32-cell endoderm blastomeres may polarize
Induction of Notochord and stage (Nakatani et al., 1996; Kim and the responding blastomeres at the 32-
Mesenchyme Nishida, 2001). The expression of cell stage and promote asymmetric di-
Endoderm blastomeres are derived Fgf9/16/20 occurs at the right time visions that operate in both the ante-
from the most vegetal region. Meso- and in the right place. The signal-re- rior and posterior regions. This
derm and nerve cord precursors sur- ceiving blastomeres quickly lose their directed signaling and asymmetric di-
round the central endoderm blas- competence at the next cleavage (Na- vision model is supported by the fact
tomeres. This marginal area in the katani and Nishida, 1999). that treatment of isolated blastomeres
AXIS SPECIFICATION IN ASCIDIANS 1187

with FGF in seawater causes both chord in the posterior region in place tor for mesenchyme differentiation; it
daughters to assume induced fates, of mesenchyme. macho-1– overex- is specifically expressed in mesen-
because isolated mother blastomeres pressed embryos form mesenchyme in chyme precursors. The expression de-
receive the signal over the entire cell place of notochord. Most intriguing re- pends on macho-1 and FGF, and
surface (Kim et al., 2000; Minokawa et sult is that when macho-1 is overex- twist-like1 is essential for the expres-
al., 2001). The results of blastomere pressed in entire embryos and blas- sion of late mesenchyme marker
transplantation also support the tomeres of the animal hemisphere are genes (Fig. 4; Imai et al., 2003; To-
model (G.J. Kim and H.N., unpub- isolated and treated with FGF, those kuoka et al., 2004). snail also lies
lished). Presumably, FGF signaling cells develop into mesenchyme. ma- downstream of macho-1 (Kobayashi et
causes localized changes in the cho-1 is sufficient to confer compe- al., 2003) and is involved in the control
mother cell. The extracellular FGF tence for induction by FGF to form of responsiveness. snail encodes a Zn-
signal is transduced to the nuclei by mesenchyme even in animal blas- finger transcription repressor and is
conserved molecules in ascidians: tomeres. Therefore, without macho-1, expressed in muscle and mesenchyme
FGF receptor, ras, MEK, and MAPK the anterior marginal blastomeres di- precursors at the 32-cell stage (Fig. 4;
(Nakatani and Nishida, 1997; Kim vide asymmetrically into notochord Erives et al., 1998; Wada and Saiga,
and Nishida, 2001; Shimauchi et al., and nerve cord precursors. The poste- 1999). As discussed two paragraphs
2001b). The mRNAs of these genes are rior marginal blastomeres containing below, brachyury is a key transcrip-
supplied maternally. The simplest macho-1 divide into mesenchyme and tion factor for notochord differentia-
model is that MAPK is activated in muscle. tion in ascidians. Analysis of the cis-
one side of the signal-receiving cell In inductive interactions, extracel- regulatory region of the brachyury
that faces the inducer cell, and then lular signaling molecule and intrinsic gene and overexpression of snail
cleavage starts before the activation factors are combined in signal-receiv- mRNA indicate that snail represses
propagates into the entire cytoplasmic ing cells, conferring particular cell the activation of brachyury in the pos-
region of the cell. At least, activated fates. Intrinsic factors of responding terior blastomeres (Fujiwara et al.,
and phosphorylated MAPK is ob- cells play roles in defining the re- 1998; Kobayashi et al., 2003). This
served only in the nucleus of one sponse. Thus, in the case of these as- finding partly explains how the ma-
daughter cell that faces the endoderm cidian embryonic inductions, the ex- cho-1 transcription activator can sup-
(Nishida, 2003), although a high back- tracellular signal is transduced into press notochord fate in the posterior
ground of immunostaining unfortu- the Ets transcription factor, while the region.
nately prevented detection of asym- intrinsic competence factor is macho-1 Antibodies against macho-1 protein
metric activation of MAPK in the transcription factor. are not available yet, but evidence is
cytoplasm of the mother cells before accumulating that macho-1 transla-
the asymmetric cell division. Zygotic Expression of Key tion is initiated at the eight-cell stage,
Transcription Factors: and the protein diffuses into the entire
Responsiveness to Inductive cytoplasm of the posterior-vegetal
Muscle and Notochord (B4.1) blastomeres of the eight-cell
Signal embryo. The B4.1 blastomere gives
macho-1 is a transcription activator
Ets family transcription factors is and binds to specific DNA sequences. rise mainly to muscle, mesenchyme,
known as one of the targets of the Yagi et al. (2004) and Sawada et al. and endoderm. However, every de-
Ras–MAPK signaling pathway (re- (2005) have reported genes down- scendant of the B4.1 blastomere, even
viewed by Wasylyk et al., 1998; Yordy stream of macho-1. Various genes for endoderm-lineage cells, assumes mus-
and Muise-Helmericks, 2000). It has transcription factors and muscle cle fate without cell interaction (Kon-
been shown that Ets is also required structural protein have been found. doh et al., 2003). FGF and bone
for notochord and mesenchyme induc- Some of these genes have a macho- morphogenetic protein (BMP) are in-
tion in ascidian (Miya and Nishida, binding consensus sequence in their 5⬘ volved in formation of endoderm cells
2003). Ets mRNA is maternally upstream regions (Yagi et al., 2004). from the B4.1 blastomeres. By con-
present and ubiquitous. The same Among these genes, Tbx6 is a candi- trast, endoderm formation from the
FGF signal and the same intracellular date of a key transcription factor for A4.1 (anterior-vegetal) blastomeres,
signal transduction cascade down to muscle development, because misex- which do not have macho-1 products,
the Ets transcription factor induce dif- pression of Tbx6 causes ectopic ex- is a cell-autonomous process and does
ferent responses in the anterior and pression of muscle structural genes not require cell interaction. The cell
posterior marginal blastomeres. What (Mitani et al., 1999). Tbx6 is expressed interactions in the B4.1 descendants
causes this difference in responsive- in muscle precursor blastomeres at are necessary for the suppression of
ness? The difference must preexist cleavage stages (Fig. 4). The expres- macho-1 function in B-line endoderm
within the responding cells. Results of sion of a myogenic factor (AMD1 in precursors, because injection of ma-
removal and transfer of egg cytoplasm Halocynthia, Ci-MDF in Ciona; Satoh cho-1 MO makes cell interaction un-
indicate that the PVC is involved. In- et al., 1996; Meedel et al., 1997) is also necessary (Kondoh et al., 2003).
deed, macho-1 causes the difference muscle-specific at the cleavage stages, Therefore, macho-1 directs muscle
between anterior and posterior blas- although the function of it is not fully fate in posterior muscle cells; its func-
tomeres (Kobayashi et al., 2003). ma- analyzed yet. On the other hand, tion is likely modified by FGF signal-
cho-1– deficient embryos form noto- twist-like1 is a key transcription fac- ing to promote mesenchyme fate in
1188 NISHIDA

intermediate mesenchyme cells; and sors at the 64-cell stage. Finally, ex- domain and is specifically expressed
in central B-line endoderm cells its pression of brachyury, a key transcrip- in TLC precursors from the 64-cell
function is totally suppressed by FGF tion factor in notochord formation, stage (Fig. 4). FoxD activity is re-
and BMP. Probably there are intrinsic specifically starts in notochord precur- quired for the expression of the Hand-
differences in responsiveness to FGF sors of 64-cell embryos (Yasuo and like gene. Blastomere isolation and re-
between mesenchyme and endoderm Satoh, 1998). Injection of Zic MO combination experiments showed that
blastomeres, and the localization of eliminates brachyury expression and inductive interaction from the animal
endoderm determinants could account notochord formation (Wada and Saiga, blastomere at the 16-cell stage is impor-
for the differences. The cell interac- 2002; Imai et al., 2002c), and Zic over- tant and that only TLC-lineage blas-
tions would restrict the regions where expression ectopically activates the tomeres have competence (Kawami-
maternal determinants work, and brachyury promoter through the Zic- nami and Nishida, 1997).
play a role in marking a precise binding site (Yagi et al., 2003). Zic ap- TVC precursor blastomeres (B7.5)
boundary between precursor cells of pears to be a competence factor in no-
and putative germline precursor blas-
different tissue types. It would be tochord induction, although this needs
tomeres (B7.6) are present in the pos-
hard for embryos to precisely partition further investigation. Zic contains a
terior-most region (purple and brown
muscle determinants exclusively into Zn-finger domain that is closely re-
in Fig. 1, respectively). B7.5 and B7.6
multiple muscle precursor blas- lated to that of macho-1. Therefore, it
are sister cells that are generated by
tomeres through a complicated cleav- is interesting that similar Zn-finger
proteins with the same origin are used the final unequal cleavage to the 64-
age history. These findings suggest
that the cell interactions are also used in anterior (Zic) and posterior (ma- cell stage. B7.6 is the smaller blas-
to suppress functions of inappropri- cho-1) regions. Another interesting tomere; it is present at the posterior
ately distributed maternal determi- symmetry is seen in the key transcrip- pole and inherits the CAB and post-
nants after embryogenesis starts. tion factors. Both brachyury and Tbx6 plasmic/PEM RNAs as mentioned be-
FGF treatment can induce noto- belong to the T-box family, and they fore. On the other hand, B7.5 is pre-
chord fate only in presumptive noto- participate in notochord (anterior) cursor of TVCs of the larvae and adult
chord/nerve cord-lineage cells. Cells in and muscle (posterior) formation, re- heart. Mesp, which has a bHLH do-
the animal hemisphere never respond spectively. main, is a key transcription factor for
to FGF by assuming notochord fate. formation of TVCs and adult heart. It
This finding indicates that responsive- is specifically expressed in TVC pre-
ness to induction to form notochord is cursors from the 110-cell stage (Fig.
restricted to marginal notochord/
Zygotic Expression of Key 4). macho-1 is required for Mesp ex-
nerve cord-lineage cells. ␤-catenin– Transcription Factors: pression (Satou et al., 2004). In accor-
deficient embryos fail to develop noto- Mesenchyme, Trunk Ventral dance, every descendant of B7.5 as-
chord, as the downstream FGF9/ Cells, and Trunk Lateral sumes larval muscle fate without cell
16/20 is not expressed. However, even Cells interaction (Nishida, 1992a; Kondoh
if FGF9/16/20 is overexpressed in et al., 2003). In more detail, the B7.5
␤-catenin– deficient embryos, noto- Among mesodermal tissues, muscle blastomere divides into a larval mus-
chord is not formed (Imai et al., 2000, and notochord function at the swim- cle precursor and a TVC precursor at
2002a). This indicates that compe- ming tadpole stage. In addition, three the next division. Therefore, directed
tence to be induced to form notochord cell types in the larvae—mesenchyme, signaling and asymmetric division
is also lost. It has been suggested that trunk lateral cells (TLCs), and trunk might also operate in this cell division
␤-catenin, FoxD, and Zic are involved ventral cells (TVCs)—are preserved in (Fig. 1). Various cell interactions are
in the acquisition of competence (Fig. the larvae and function after meta- speculated to be involved in TVC spec-
4). MOs of these genes abolish noto- morphosis. Mesenchyme gives rise to
ification (Davidson and Levine, 2003).
chord. FoxD gene is a direct target of tunic cells in the juvenile. TLCs are
The competence factor for TVC induc-
␤-catenin, as its expression depends precursors of blood cells and body wall
tion may be one of the products of
on ␤-catenin and on a Tcf-binding site muscle. TVCs develop into heart and
postplasmic/PEM RNAs.
in its 5⬘ upstream cis-element (Imai et body wall muscle (Hirano and
The marginal zone of the vegetal
al., 2002b). FoxD expression is inde- Nishida, 1997). As mentioned above,
pendent of FGF9/16/20 and vice mesenchyme specification requires hemisphere is patterned along the A-P
versa. Expression of FoxD is transient macho-1 and FGF. Twist-like1 is a key axis, as discussed above. Localized
at the 16-and 32-cells stages in the transcription factor for mesenchyme. maternal factors are involved in the
vegetal hemisphere. FoxD is expressed Recent studies have shown that TLCs A-P polarization. Then, cellular inter-
in notochord/nerve cord/endoderm- and TVCs are specified during cleav- actions play important roles in this
lineage blastomeres at the 16-cell age stages. TLC precursor (A7.6) blas- patterning when embryos become
stage. Then Zic expression starts in tomeres of the 64-cell embryo form in multicellular. Zygotic gene expression
notochord/nerve cord-lineage cells at the lateral region of the vegetal hemi- is involved in production of both of
the 32-cell stage (Wada and Saiga, sphere (sky blue in Fig. 1). Hand-like/ inductive signals and competence fac-
2002; Imai et al., 2002c). The expres- NoTrlc is a key transcription factor for tors, and finally expression of cell-type
sion depends on FoxD and persists in TLC formation (Imai et al., 2003). It specific key transcription factors is
both notochord and nerve cord precur- has a basic helix–loop– helix (bHLH) bought about.
AXIS SPECIFICATION IN ASCIDIANS 1189

Animal Hemisphere have been analyzed blastomere by communication at early developmental


blastomere at the single-cell level in stages because of its limited cell num-
What happens in the animal hemi-
the ascidian embryo. This analysis ber and invariant mode of development.
sphere is much simpler. Most regions
combined with the stereotypic cleav-
of the animal hemisphere are occupied
age pattern makes studies of ascidian
by epidermis blastomeres. Only on the
embryogenesis easy to analyze and
Maternal Factors
anterior edge are brain precursors
understand. Ascidian embryogenesis Analysis of maternal processes is also
formed (Fig. 1). The formation of brain
has been hitherto regarded as a typi- under way, and the most pressing
precursors requires induction from
cal example of mosaic development, questions are: How is the A-V polarity
the vegetal blastomeres (reviewed by
but recent analyses revealed that cell established during oogenesis? Is Di-
Okamura et al., 1993; Lemaire et al.,
interactions play equally important shevelled a vegetal determinant in as-
2002; Meinertzhagen et al., 2004).
roles. In retrospect, there are two cidians? Is there a gradient of ␤-cate-
Otx, a putative key transcription fac-
main reasons for ascidian embryos to nin stabilization along the A-V axis,
tor for brain development, is ex-
be regarded as a mosaic. When blas- and how is the intermediate marginal
pressed in these blastomeres (Fig. 4).
tomeres are isolated at an early cleav- zone specified? What are the roles of
Otx is a homeobox transcription factor
age stage, for example at the eight-cell unanalyzed postplasmic/PEM RNAs
that plays important roles in brain de-
stage, cell interactions take place in establishment of the A-P axis? Cel-
velopment in Drosophila and in verte-
within the partial embryos derived lular mechanisms of unequal cleavage
brates (Simeone, 1998). The functions
from the isolated blastomeres. There- in relation to the CAB and anchoring
of otx have been investigated with MO
fore, the blastomeres can develop var- of postplasmic RNA to cER will be im-
(Wada et al., 2004). In the absence of
ious tissues as expected from the fate portant issues to pursue. In addition,
Otx function, brain development was
map, and spuriously behave in an au- spatiotemporal analysis of FGF signal
abrogated but expression of some
tonomous way. In other words, the transduction processes will be re-
brain-specific genes persisted. There-
blastomeres can develop autono- quired to fully understand the mecha-
fore, the idea that Otx is a key tran-
mously in isolation; however, tissue nisms of asymmetric divisions after
scription factor is still controversial.
formation is not cell-autonomous pro- cells receive directed signals.
FGF/Ets and GATA are crucial for the
cess in these cases.
expression in brain precursors (In-
The second reason is that ascidian
azawa et al., 1998; Miya and Nishida, Genome and ESTs
embryos do not compensate for lost
2003; Bertrand et al., 2003). In accor-
parts if blastomeres are ablated. This An evolutionary point of view is also
dance, Ets- and GATA-binding cis-el-
is because competence or responsive- interesting because of the phylogenic
ements are crucial for the expression
ness to be induced to certain tissues is position of ascidians (Di Gregorio and
of the Otx–reporter construct. Thus,
indeed restricted to the tissue precur- Levine, 2001; Corbo et al., 2001; Sa-
the FGF signal from the vegetal hemi-
sor blastomeres, as discussed in this toh, 2003a). So far, most research has
sphere induces brain precursors
article. Generally in animal develop- been concentrated on embryogenesis.
through activation of Ets. GATA gene
ment, various organisms use a limited However, analysis of mechanisms of
expression is maternal and ubiqui-
repertoire of signaling molecules for metamorphosis is also worth analysis
tous, but it is translated only in ani-
intercellular communication. The same (Hirano and Nishida, 1997, 2000; Eri
mal blastomeres at the late 32-cell
signals are used over and over again et al., 1999; Davidson and Swalla,
stage, when induction takes place.
in different cells at different stages, 2002).
These results suggest that GATA is
with different biological outcomes de- Recently, infrastructure for ascid-
the competence factor that restricts
pending on the spatial and temporal ian research has been set up expedi-
brain induction to the animal hemi-
context. To understand animal devel- tiously. A cDNA project involving
sphere (Bertrand et al., 2003). It is
opment, it is important to clarify how large-scale ESTs and in situ hybrid-
still not clear how the brain is induced
embryonic cells can respond differ- ization of maternal mRNA in Halocyn-
only at the anterior edge, although
ently to the same signal, and to under- thia roretzi (http://www.genome.jp/
blastomere recombination experi-
stand how competence is restricted to magest/; Makabe et al., 2001) and a
ments and treatment with inducing
specific cells. cDNA project on maternal and zygotic
substance demonstrated that the an-
Ascidian embryo provides a good transcripts in Ciona intestinalis (http://
terior and posterior blastomeres have
model for elucidating this issue. Cel- ghost.zool.kyoto-u.ac.jp/indexr1.html;
different efficiencies of brain induc-
lular simplicity and transparency of Satou et al., 2001b, 2002b) will help fu-
tion (Okado and Takahashi, 1993;
ascidian embryos have opened the ture studies. The genome sequence of C.
Hudson and Lemaire, 2001).
way to the use of sophisticated labeling intestinalis is completed (http://genome.
and imaging techniques, which allow jgi-psf.org/ciona4/ciona4.home.html; De-
SUMMARY AND FUTURE for three-dimensional and four-dimen- hal et al., 2002; Satoh, 2003b), and that
DIRECTIONS sional reconstruction of virtual em- of C. savignyi is nearly completed (http://
Mosaic Development and bryos. Hence, the imaging tools may www.broad.mit.edu/annotation/ciona/;
help access to cell contact surface or cell http://www2.bioinformatics.tll.org.sg:
Cell Interactions division features of neighboring blas- 8082/Ciona_savignyi/). Comparison of
As can be seen in Figures 1, 3, and 4, tomeres. Ascidian embryo is indeed a promoter sequences in these closely
fate specification and gene expression perfect system for elucidating cellular related species are useful for detecting
1190 NISHIDA

conserved regulatory cis-elements for simples. J Anat Physiol (Paris) 23:167– Fujimura M, Takamura K. 2000. Charac-
gene expression (Johnson et al., 2004). 319. terization of an ascidian DEAD-box
Conklin EG. 1905. The organization and gene, CiDEAD1: specific expression in
Developmentally important genes the germ cells and its mRNA localization
cell lineage of the ascidian egg. J Acad
are annotated in a special ascidian is- Nat Sci (Phila) 13:1–119. in the posterior-most blastomeres in
sue of Development Genes and Evolu- Corbo J, Di Gregorio A, Levine M. 2001. early embryos. Dev Genes Evol 210:64 –
tion (Volume 213, No. 5– 6, 2003). Re- The ascidian as a model organism in de- 72.
cently, Imai et al. (2004) listed every velopmental and evolutionary biology. Fujiwara S, Corbo JC, Levine M. 1998. The
Cell 106:535–538. snail repressor establishes a muscle/no-
gene for transcription factors and sig- tochord boundary in the Ciona embryo.
Davidson B, Levine M. 2003. Evolutionary
naling molecules in the C. intestinalis origins of the vertebrate heart: specifica- Development 125:2511–2520.
genome, and the expression pattern tion of the cardiac lineage in Ciona intes- Gilbert SF. 2003. Developmental biology.
during embryogenesis was compre- tinalis. Proc Natl Acad Sci U S A 100: 7th ed. Sunderland: Sinauer Associates.
hensively reported (http://ghost.zool. 11469 –11473. Heasman J. 1997. Patterning Xenopus
Davidson B, Swalla BJ. 2002. A molecular blastula. Development 124:1553–1560.
kyoto-u.ac.jp/tfst.html). The minimal analysis of ascidian metamorphosis re- Hibino T, Nishikata T, Nishida H. 1998.
gene redundancy in the ascidian ge- veals activation of an innate immune re- Centrosome-attracting body: a novel
nome will simplify our understanding sponse. Development 129:4739 –4751. structure closely related to unequal
of the expression patterns and the re- Dehal P, Satou Y, Campbell RK, Chapman cleavages in the ascidian embryo. Dev
J, Degnan B, De Tomaso A, Davidson B, Growth Differ 40:85–95.
sults of gene knockdown experiments.
Di Gregorio A, Gelpke M, Goodstein DM, Hirano T, Nishida H. 1997. Developmental
The wealth of genetic information on Harafuji N, Hastings KE, Ho I, Hotta K, fates of larval tissues after metamorpho-
ascidians will promote studies to en- Huang W, Kawashima T, Lemaire P, sis in ascidian Halocynthia roretzi. I. Or-
visage the molecular network during Martinez D, Meinertzhagen IA, Necula igin of mesodermal tissues of the juve-
ascidian embryogenesis. Genetic ap- S, Nonaka M, Putnam N, Rash S, Saiga nile. Dev Biol 192:199 –210.
H, Satake M, Terry A, Yamada L, Wang Hirano T, Nishida H. 2000. Developmental
proaches are now feasible in Ciona fates of larval tissues after metamorpho-
HG, Awazu S, Azumi K, Boore J, Branno
and have begun bearing fruit (e.g., M, Chin-Bow S, DeSantis R, Doyle S, sis in ascidian Halocynthia roretzi. II.
Nakatani et al., 1999; Awazu et al., Francino P, Keys DN, Haga S, Hayashi Origin of endodermal tissues of the juve-
2004). Another possible genetic ap- H, Hino K, Imai KS, Inaba K, Kano S, nile. Dev Gene Evol 210:55–63.
proach will be to use appendicularians Kobayashi K, Kobayashi M, Lee BI, Hudson C, Lemaire P. 2001. Induction of
Makabe KW, Manohar C, Matassi G, anterior neural fates in the ascidian
(Urochordata, a larvecian, Oiko- Medina M, Mochizuki Y, Mount S, Mor- Ciona intestinalis. Mech Dev 100:189 –
pleura), which have a simple chordate ishita T, Miura S, Nakayama A, Nish- 203.
body plan (Fenaux, 1998; Nishino and izaka S, Nomoto H, Ohta F, Oishi K, Hudson C, Clements D, Friday RV, Stott D,
Satoh, 2001), and their life cycle is Rigoutsos I, Sano M, Sasaki A, Sasakura Woodland HR. 1997. Xsox17alpha and
Y, Shoguchi E, Shin-i T, Spagnuolo A, -beta mediate endoderm formation in Xe-
only 4 to 7 days.
Stainier D, Suzuki MM, Tassy O, Taka- nopus. Cell 91:397–405.
tori N, Tokuoka M, Yagi K, Yoshizaki F, Ikenishi K. 1998. Germ plasm in Caeno-
Wada S, Zhang C, Hyatt PD, Larimer F, rhabditis elegans, Drosophila, and Xeno-
ACKNOWLEDGMENTS Detter C, Doggett N, Glavina T, pus. Dev Growth Differ 40:1–10.
I thank Dr. G. Kumano, Dr. J. Matsu- Hawkins T, Richardson P, Lucas S, Imai KS. 2003. Isolation and characteriza-
Kohara Y, Levine M, Satoh N, Rokhsar tion of ␤-catenin downstream genes in
moto, and Dr. C. Sardet for their valu- DS. 2002. The draft genome of Ciona early embryos of the ascidian Ciona sav-
able comments on the manuscript. intestinalis: insights into chordate and ignyi. Differentiation 71:346 –360.
vertebrate origins. Science 298:2157– Imai K, Takada N, Satoh N, Satou Y. 2000.
2167. ␤-catenin mediates the specification of
REFERENCES Di Gregorio A, Levine M. 2001. Ascidian endoderm cells in ascidian embryos. De-
embryogenesis and the origins of the velopment 127:3009 –3020.
Arendt D, Nübler-Jung K. 1997. Dorsal or chordate body plan. Curr Opin Genet Imai KS, Satoh N, Satou Y. 2002a. Early
ventral: similarities in fate maps and Dev 8:457–463. embryonic expression of FGF4/6/9 gene
gastrulation patterns in annelids, ar- Di Gregorio A, Corbo JC, Levine M. 2001. and its role in the induction of mesen-
thropods and chordates. Mech Dev 61:7– The regulation of forkhead/HNF-3␤ ex- chyme and notochord in Ciona savignyi
21. pression in the Ciona embryo. Dev Biol embryos. Development 129:1729 –1738.
Awazu S, Sasaki A, Matsuoka T, Satoh N, 229:31–43. Imai KS, Satoh N, Satou Y. 2002b. An es-
Sasakura Y. 2004. An enhancer trap in Emily-Fenouil F, Ghiglione C, Lhomond G, sential role of a FoxD gene in notochord
the ascidian Ciona intestinalis identifies Lepage T, Gache C. 1998. GSK3␤/shaggy induction in Ciona intestinalis. Develop-
enhancer of its Musashi orthologous mediates patterning along the animal- ment 129:3441–3453.
gene. Dev Biol 275:459 –472. vegetal axis of the sea urchin embryo. Imai KS, Satou Y, Satoh N. 2002c. Multiple
Bates WR, Jeffery WR. 1987. Alkaline Development 125:2489 –2498. functions of a Zic-like gene in the differ-
phosphatase expression in ascidian egg Eri R, Arnold JM, Hinman VF, Green KM, entiation of notochord, central nervous
fragments and andromerogons. Dev Biol Jones MK, Degnan BM, Lavin MF. 1999. system and muscle in Ciona savignyi
119:382–389. Hemps, a novel EGF-like protein, plays a embryos. Development 129:2723–2732.
Bertrand V, Hudson C, Caillol D, Popovici central role in ascidian metamorphosis. Imai KS, Satoh N, Satou Y. 2003. A twist-
C, Lemaire P. 2003. Neural tissue in as- Development 126:5809 –5818. like bHLH gene is a downstream factor
cidian embryos is induced by FGF9/16/ Erives A, Corbo JC, Levine M. 1998. Lin- of an endogenous FGF and determines
20, acting via a combination of maternal eage-specific regulation of the Ciona mesenchymal fate in the ascidian em-
GATA and Ets transcription factors. Cell snail gene in the embryonic mesoderm bryos. Development 130:4461–4472.
115:615–627. and neuroectoderm. Dev Biol 194:213– Imai KS, Hino K, Yagi K, Satoh N, Satou
Cadigan KM, Nusse R. 1997. Wnt signal- 225. Y. 2004. Gene expression profiles of tran-
ing: a common theme in animal develop- Fenaux R. 1998. Life history of the appen- scription factors and signaling molecules
ment. Genes Dev 11:3286 –3305. dicularia. In: Bone Q, editor. The biology in the ascidian embryo: towards a com-
Chabry L. 1887. Contribution l’embryologie of pelagic tunictes. New York: Oxford prehensive understanding of gene net-
normale et teratologique des Ascidies University Press. works. Development 131:4047–4058.
AXIS SPECIFICATION IN ASCIDIANS 1191

Inazawa T, Okamura Y, Takahashi K. pressed proteins: implications for the sociated cells of the ascidian embryo.
1998. Basic fibroblast growth factor in- evolution of chordate muscle gene regu- Rouxs Arch Dev Biol 201:81–87.
duction of neural ion channel expression lation. Development 124:1711–1721. Nishida H. 1992b. Regionality of egg cyto-
in ascidian ectodermal blastomeres. Meinertzhagen IA, Lemaire P, Okamura Y. plasm that promotes muscle differentia-
J Physiol 511:347–359. 2004. The neurobiology of the ascidian tion in embryo of the ascidian, Halocyn-
Iseto T, Nishida H. 1999. Ultrastructural tadpole larva: recent developments in an thia roretzi. Development 116:521–529.
studies on the centrosome-attracting ancient chordates. Annu Rev Neurosci Nishida H. 1994. Localization of determi-
body: electron-dense matrix and its role 27:453–485. nants for formation of the anterior-pos-
in unequal cleavage in ascidian embryos. Miller JR, Moon RT. 1996. Signal trans- terior axis in eggs of the ascidian Halo-
Dev Growth Differ 41:601–609. duction through ␤-catenin and specifica- cynthia roretzi. Development 120:3093–
Ishida K, Ueki T, Satoh N. 1996. Spatio- tion of cell fates during embryogenesis. 3104.
temporal expression patterns of eight Genes Dev 10:2527–2539. Nishida H. 1996. Vegetal egg cytoplasm
epidermis-specific genes in the ascidian Miller JR, Rowning BA, Larabell CA, promotes gastrulation and is responsible
embryo. Zool Sci 13:699 –709. Yang-Snyder JA, Bates RL, Moon RT. for specification of vegetal blastomeres
Jeffery WR. 2001. Determinants of cell and 1999. Establishment of the dorsal-ven- in embryos of the ascidian Halocynthia
positional fate in ascidian embryos. Int tral axis in Xenopus embryos coincides roretzi. Development 122:1271–1279.
Rev Cytol 203:3–62. with the dorsal enrichment of dishev- Nishida H. 1997. Cell-fate specification by
Johnson D, Davidson B, Brown CD, Smith elled that is dependent on cortical rota- localized cytoplasmic determinants and
WC, Sidow A. 2004. Noncoding regula- tion. J Cell Biol 146:427–437. cell interaction in ascidian embryos. Int
tory sequences of Ciona exhibit strong Minokawa T, Yagi K, Makabe KW, Nishida Rev Cytol 176:245–306.
correspondence between evolutionary H. 2001. Binary specification of nerve Nishida H. 2002a. Specification of develop-
constraint and functional importance. cord and notochord cell fates in ascidian mental fates in ascidian embryos: molec-
Genome Res 14:2448 –2456. embryos. Development 128:2007–2017. ular approach to maternal determinants
Kawaminami T, Nishida H. 1997. Induc- Mitani Y, Takahashi H, Satoh N. 1999. An and signaling molecules. Int Rev Cytol
tion of trunk lateral cells, the blood cell ascidian T-box gene As-T2 is related to 217:227–276.
precursors, during ascidian embryogen- the Tbx6 subfamily and is associated Nishida H. 2002b. Patterning the marginal
esis. Dev Biol 181:14 –20. with embryonic muscle cell differentia- zone of early ascidian embryos: localized
Kim GJ, Nishida H. 2001. Role of FGF and tion. Dev Dyn 215:62–68. maternal mRNA and inductive interac-
MEK signaling pathway in the ascidian Miya T, Nishida H. 2002. Isolation of cDNA tions. Bioessays 24:87–92.
embryo. Dev Growth Differ 43:521–533. clones for mRNAs transcribed zygoti- Nishida H. 2003. Spatio-temporal pattern
Kim GJ, Yamada A, Nishida H. 2000. An cally during cleavage in the ascidian, of the activation of MAP kinase in em-
FGF signal from endoderm and localized Halocynthia roretzi. Dev Genes Evol 212: bryos of the ascidian, Halocynthia ro-
factors in the posterior-vegetal egg cyto- 31–37. retzi. Dev Growth Differ 45:27–37.
plasm pattern the mesodermal tissues in Nishida H, Sawada K. 2001. macho-1 en-
Miya T, Nishida H. 2003. An Ets transcrip-
the ascidian. Development 127:2853– codes a localized mRNA in ascidian eggs
tion factor, HrEts, is target of FGF sig-
2862. that specifies muscle fate during embry-
naling and involved in induction of noto-
Kobayashi K, Sawada K, Yamamoto H, ogenesis. Nature 409:724 –729.
chord, mesenchyme, and brain in ascidian
Wada S, Saiga H, Nishida H. 2003. Ma- embryos. Dev Biol 261:25–38. Nishida H, Morokuma J, Nishikata T.
ternal macho-1 is an intrinsic factor that 1999. Maternal cytoplasmic factors for
Nakamura Y, Makabe KW, Nishida H.
makes cell response to the same FGF generation of unique cleavage pattern in
2003. Localization and expression pat-
signal differ between mesenchyme and animal embryos. Curr Top Dev Biol 46:
tern of type I postplasmic mRNAs in em-
notochord induction in ascidian embryos. 1–37.
bryos of the ascidian Halocynthia roretzi.
Development 130:5179 –5190. Nishikata T, Hibino T, Nishida H. 1999.
Gene Express Pattern 3:71–75.
Kondoh K, Kobayashi K, Nishida H. 2003. The centrosome-attracting body, micro-
Suppression of macho-1-directed muscle Nakatani Y, Nishida H. 1997. Ras is an tubule system, and posterior egg cyto-
fate by FGF and BMP is required for essential component for notochord for- plasm are involved in positioning of
formation of posterior endoderm in as- mation during ascidian embryogenesis. cleavage planes in the ascidian embryo.
cidian embryos. Development 130:3205– Mech Dev 68:81–89. Dev Biol 209:72–85.
3216. Nakatani Y, Nishida H. 1999. Duration of Nishino A, Satoh N. 2001. The simple tail
Laverriere AC, MacNiell C, Mueller C, competence and inducing capacity of of chordates: phylogenic significance of
Poelmann RE, Burch JBE, Evance T. blastomeres in notochord induction dur- appendicularians. Genesis 29:36 –45.
1994. GATA-4/5/6: a subfamily of three ing ascidian embryogenesis. Dev Growth Numakunai T. 2001. Oocyte maturation
transcription factors transcribed in the Differ 41:449 –453. and self-sterility by treatment with
developing heart and gut. J Biol Chem Nakatani Y, Yasuo H, Satoh N, Nishida H. ovary extracts of the ascidian, Halocyn-
269:23177–23184. 1996. Basic fibroblast growth factor in- thia roretzi. In: Sawada H, Yokosawa H,
Lemaire P, Bertrand V, Hudson C. 2002. duces notochord formation and the ex- Lambert CC, editors. The biology of as-
Early steps in the formation of neural pression of As-T, a Brachyury homolog, cidians. Tokyo: Springer-Verlag.
tissue in ascidian embryos. Dev Biol 252: during ascidian embryogenesis. Develop- Okado H, Takahashi K. 1993. Neural dif-
151–169. ment 122:2023–2031. ferentiation in cleavage-arrested ascid-
Logan CY, Miller JR, Ferkowicz MJ, Ma- Nakatani Y, Moody R, Smith WC. 1999. ian blastomeres induced by a proteolytic
Clay DR. 1999. Nuclear ␤-catenin is re- Mutations affecting tail and notochord enzyme. J Physiol 463:269 –290.
quired to specify vegetal cell fates in the development in the ascidian Ciona sav- Okamura Y, Okado H, Takahashi K. 1993.
sea urchin embryo. Development 126: ignyi. Development 126:3293–3301. The ascidian embryo as a prototype of
345–357. Nieuwkoop PD. 1977. Origin and establish- vertebrate neurogenesis. Bioessays 15:
Makabe KW, Kawashima T, Kawashima S, ment of embryonic polar axes in amphib- 723–729.
et al. 2001. Large-scale cDNA analysis of ian development. Curr Top Dev Biol 11: Olsen CL, Jeffery WR. 1997. A forkhead
the maternal genetic information in the 115–132. gene related to HNF-3b is required for
egg of the ascidian, Halocynthia roretzi, Nishida H. 1987. Cell lineage analysis in gastrulation and axis formation in the
for the gene expression catalog during ascidian embryos by intracellular injec- ascidianembryo.Development124:3609 –
development. Development 128:2555– tion of a tracer enzyme. III. Up to the 3619.
2567. tissue-restricted stage. Dev Biol 121:526 – Reverberi G, Minganti A. 1946. Fenomeni
Meedel TH, Farmer SC, Lee JJ. 1997. The 541. di evocatione nelle sviluppo dell’uovo di
single MyoD family genes of Ciona intes- Nishida H. 1992a. Developmental poten- ascidie. Pubbl Stn Zool Napoli 20:199 –
tinalis encodes two differentially ex- tial for tissue differentiation of fully dis- 252.
1192 NISHIDA

Ristoratore F, Spagnuolo A, Aniello F, Br- ferentiation of muscle cells in the ascid- cynthia roretzi. Development 128:2711–
anno M, Fabbrini F, Di Lauro R. 1999. ian embryo. Proc Natl Acad Sci U S A 2721.
Expression and functional analysis of 93:9315–9321. Simeone A. 1998. Otx1 and Otx2 in the
Cititf1, an ascidian NK-2 class gene, sug- Satou Y, Imai KS, Satoh N. 2001a. Early development and evolution of the mam-
gest its role in endoderm development. embryonic expression of a LIM-ho- malian brain. EMBO J 17:6790 –6798.
Development 126:5149 –5159. meobox gene Cs-lhx3 is downstream of Speksnijder J, Jaffe L, Sardet C. 1989. Po-
Roegiers F, McDougall A, Sardet C. 1995. ␤-catenin and responsible for the em- larity of sperm entry in the ascidian egg.
The sperm entry point defines the orien- doderm differentiation in Ciona savignyi Dev Biol 133:180 –184.
tation of the calcium-induced contraction embryos. Development 128:3559 –3570. Takamura K, Fujimura M, Yamaguchi Y.
wave that directs the first phase of cyto- Satou Y, Takatori N, Yamada L, Mochizuki 2002. Primordial germ cells originate
plasmic reorganization in the ascidian Y, Hamaguchi M, Ishikawa H, Chiba S, form the endodermal strand cells in the
egg. Development 121:3457–3466. Imai K, Kano S, Murakami SD, Na- ascidian Ciona intestinalis. Dev Genes
Roegiers F, Djediat C, Dumollard R, Rou- kayama A, Nishino A, Sasakura Y, Satoh Evol 212:11–18.
viére C, Sardet C. 1999. Phases of cyto- G, Shimotori T, Shin-I T, Shoguchi E, Takatori N, Hotta K, Mochizuki Y, Satoh
plasmic and cortical reorganizations of Suzuki MM, Takada N, Utsumi N, Yo- G, Mitani Y, Satoh N, Satou Y, Taka-
the ascidian zygote between fertilization shida N, Saiga H, Kohara Y, Satoh N. hashi H. 2004. T-box genes in the ascid-
andfirstdivision.Development126:3101– 2001b. Gene expression profiles in Ciona ian Ciona intestinalis: characterization
3117. intestinalis tailbud embryos. 128:2893– of cDNAs and spatial expression. Dev
Sardet C, Speksnijder J, Inoue S, Jaffe L. 2904. Dyn 230:743–753.
1989. Fertilization and ooplasmic move- Satou Y, Yagi K, Imai KS, Yamada L, Tokuoka M, Imai KS, Satou Y, Satoh N.
ments in the ascidian egg. Development Nishida H, Satoh N. 2002a. macho-1-re- 2004. Three distinct lineages of mesen-
105:237–249. lated genes in Ciona embryos. Dev chymal cells in Ciona intestinalis em-
Sardet C, Speksnijder J, Terasaki M, Genes Evol 212:87–92. bryos demonstrated by specific gene ex-
Chang P. 1992. Polarity of the ascidian Satou Y, Takatori N, Fujiwara S, Nish- pression. Dev Biol 274:211–224.
egg cortex before fertilization. Develop- ikata T, Saiga H, Kusakabe T, Shin-i T, Tomioka M, Miya T, Nishida H. 2002. Re-
ment 115:221–237. Kohara Y, Satoh N. 2002b. Ciona intes- pression of zygotic gene expression in the
Sardet C, McDougall A, Houliston E. 1994. tinalis cDNA projects: expressed se- putative germline cells in ascidian em-
Cytoplasmic domains in eggs. Trends quence tag analysis and gene expression bryos. Zool Sci 19:49 –55.
Cell Biol 4:166 –172. profiles during embryogenesis. Gene 287: Van Doren M, Williamson AL, Lehmann R.
Sardet C, Prodon F, Dumollard R, Chang 83–96. 1998. Regulation of zygotic gene expres-
P, Chenevert J. 2002. Structure and Satou Y, Imai KS, Satoh N. 2002c. Fgf sion in Drosophila primordial germ cells.
function of the egg cortex from oogenesis genes in the basal chordate Ciona in- Curr Biol 8:243–246.
through fertilization. Dev Biol 241:1–23. testinalis. Dev Genes Evol 212:432– Wada S, Saiga H. 1999. Cloning and embry-
Sardet C, Nishida H, Prodon F, Sawada K. 438. onic expression of Hrsna, a snail family
2003. Maternal mRNAs of PEM and ma- Satou Y, Imai KS, Satoh N. 2004. The as- gene of the ascidian Halocynthia roretzi:
cho-1, the ascidian muscle determinant, cidian Mesp gene specifies heart precur- implication in the origin of mechanisms
associate and move with a rough endo- sor cells. Development 131:2533–2541. for mesoderm specification and body axis
plasmic reticulum network in the egg Sawada T. 1988. The mechanism of ooplas- formation in chordates. Dev Growth Differ
cortex. Development 130:5839 –5849. mic segregation in the ascidian eggs. 41:9 –18.
Sardet C, Dru P, Prodon F. 2005. Maternal Zool Sci 5:667–675. Wada S, Saiga H. 2002. HrZicN, a new Zic
determinants and mRNA in the cortex of Sawada T, Schatten G. 1988. Microtubules family genes of ascidians, plays essential
ascidian oocytes, zygotes and embryos. in ascidian eggs during meiosis, fertiliza- roles in the neural tube and notochord
Biol Cell 97:1–15. tion, and mitosis. Cell Motil Cytoskele- development. Development 129:5597–
Sasakura T, Makabe KW. 2002. Identifica- ton 9:219 –230. 5608.
tion of cis elements which direct the lo- Sawada K, Fukushima Y, Nishida H. 2005. Wada S, Katsuyama Y, Yasugi S, Saiga H.
calization of maternal mRNAs to the Macho-1 functions as transcriptional ac- 1995. Spatially and temporally regulated
posterior pole of ascidian embryos. Dev tivator for muscle formation in embryos expression of the LIM class homeobox
Biol 250:128 –144. of the ascidian Halocynthia roretzi. Gene gene Hrlim suggests multiple distinct
Sasakura Y, Ogasawara M, Makabe KW. Express Pattern 5:429 –437. function in development of the ascidian,
1998a. HrWnt-5: a maternally expressed Schneider S, Steinbesser H, Warga RM, Halocynthia roretzi. Mech Dev 51:115–
ascidian Wnt gene with posterior local- Hausen P. 1996. ␤-catenin translocation 126.
ization in early embryos. Int J Dev Biol into nuclei demarcates the dorsalizing Wada S, Sudou N, Saiga H. 2004. Roles of
42:573–579. centers in frog and fish embryos. Mech HrOth, the ascidian otx gene, in the dif-
Sasakura Y, Ogasawara M, Makabe KW. Dev 57:191–198. ferentiation of the brain (sensory vesicle)
1998b. Maternally localized RNA encod- Seydoux G, Dunn MA. 1997. Transcrip- and anterior trunk epidermis in the lar-
ing a serine/threonine protein kinase in tionally repressed germ cells lack a sub- val development of Halocynthia roretzi.
the ascidian, Halocynthia roretzi. Mech population of phosphorylated RNA Mech Dev 121:463–474.
Dev 76:161–163. Polymerase II in early embryos of Cae- Wasylyk B, Hagman J, Gutierrez-Hart-
Sasakura Y, Ogasawara M, Makabe KW. norhabditis elegans and Drosophila mann A. 1998. Ets transcription factors:
2000. Two pathways of maternal RNA melanogaster. Development 121:2191– nuclear effectors of the Ras-MAP-kinase
localization at the posterior-vegetal cyto- 2201. signaling pathway. Trends Biochem Sci
plasm in early ascidian embryos. Dev Shimauchi Y, Yasuo H, Satoh N. 1997. 23:213–216.
Biol 220:365–378. Autonomy of ascidian fork head/HNF-3 Weitzel HE, Illies MR, Byrum CA, Xu R,
Satoh N. 1994. Developmental biology of gene expression. Mech Dev 69:143– Wikramanayake AH, Ettensohn C. 2003.
ascidians. Cambridge: Cambridge Uni- 154. Differential stability of ␤-catenin along
versity Press. Shimauchi Y, Chiba S, Satoh N. 2001a. the animal-vegetal axis of the sea urchin
Satoh N. 2003a. The ascidian tadpole lar- Synergistic action of HNF-3 and embryo mediated by disheveled. Devel-
va: comparative molecular development Brachyury in the notochord differentia- opment 131:2947-2956.
and genomics. Nat Rev Genet 4:285–295. tion of ascidian embryos. Int J Dev Biol Wikramanayake AH, Huang L, Klein WH.
Satoh N. 2003b. Ciona intestinalis: an 45:643–652. 1998. ␤-catenin is essential for pattern-
emerging model for whole-genome anal- Shimauchi Y, Murakami SD, Satoh N. ing the maternally specified animal-
ysis. Trends Genet 19:361–381. 2001b. FGF signals are involved in the vegetal axis in the sea urchin embryo.
Satoh N, Araki I, Satou Y. 1996. An intrin- differentiation of notochord cells and Proc Natl Acad Sci U S A 95:9343–
sic genetic program for autonomous dif- mesenchyme cells of the ascidian Halo- 9348.
AXIS SPECIFICATION IN ASCIDIANS 1193

Wikramanayake AH, Hong M, Lee PN, muscle determinant gene Ci-macho-1. gene encoding a localized factor in the
Pang K, Byrum CA, Bince JM, Xu R, Dev Biol 274:478 –489. ascidian embryo. Development 122:2005–
Martindale MQ. 2003. An ancient role Yasuo H, Satoh N. 1998. Conservation of 2012.
for nuclear ␤-catenin in the evolution of the developmental role of Brachyury in Zalokar M, Sardet C. 1984. Tracing of cell
axial polarity and germ layer segrega- notochord formation in a urochordate, lineage in embryonic development of
tion. Nature 426:446 –450. the ascidian Halocynthia roretzi. Dev Phallusia Mammillata (Ascidia) by vital
Yagi K, Satou Y, Satoh N. 2003. A zinc Biol 200:158 –170. staining of mitochondria. Dev Biol 102:
finger transcription factor, ZicL, is a di- Yordy JS, Muise-Helmericks RC. 2000. 195–205.
rect activator of Brachyury in the noto- Signal transduction and the Ets family Zhang J, Houston DW, King ML, Payne C,
chord specification of Ciona intestinalis. of transcription factors. Oncogene 19: Wylie C, Heasman J. 1998. The role of
Development 131:1279 –1288. 6503–6513. maternal VegT in establishing the pri-
Yagi K, Satoh N, Satou Y. 2004. Identifica- Yoshida S, Marikawa Y, Satoh N. 1996. mary germ layers in Xenopus embryos.
tion of downstream genes of the ascidian posterior end mark, a novel maternal Cell 94:515–524.

You might also like