Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

DEVELOPMENTAL DYNAMICS 218:80 –93 (2000)

REVIEWS
A PEER REVIEWED FORUM

Integration of Epithelial Patterning and Morphogenesis in


Drosophila Ovarian Follicle Cells
LEONARD L. DOBENS AND LAUREL A. RAFTERY*
Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown,
Massachusetts

ABSTRACT Drosophila oogenesis involves ized structure of the eggshell. An accompanying review
the coordinated development of germ cells and discusses the establishment and maintenance of the
an overlying follicular epithelium. The follicle FC epithelium. Here, we focus on patterning of the FC
cells provide a genetically tractable system to fates and how it is linked to the cell shape changes and
investigate the cell biology of patterning and migrations that create the three-dimensional architec-
morphogenesis. Follicle cells initially form a ture of the eggshell.
cuboidal epithelium surrounding a syncytium of The follicle cells provide a tractable system to inves-
nurse cells and oocyte. Epithelial structure is tigate key questions about the coordination of cell fate
maintained as these cells reorganize to create and behavior within an intact epithelial sheet. FCs
the three dimensional architecture of the egg- first organize a simple cuboidal epithelium; epithelial
shell. Both long-range and short-range cell-cell structure is maintained through migrations that create
communications pattern the domains of follicle a three-dimensional architecture. FC development re-
cells that will create specific eggshell structures. quires many of the same genes that regulate morpho-
After terminal differentiation to deposit the egg- genesis of other organs (Table 1). How are domains of
shell proteins, the follicle cells die. This review cell fate patterned by local and regional cell-cell com-
summarizes recent progress in understanding munication? How are boundaries set, so that morpho-
the cell-cell communication that orchestrates fol- genesis is uniquely orchestrated within each domain?
licle cell patterning and migrations. DE-cad- How is tissue architecture formed, maintained, and
herin-mediated adhesion is important at several dissolved? Early on, cell contacts were recognized as
steps in egg chamber formation and follicle cell important signals in FC morphogenesis. More recently,
migration. Notch signaling is critical during each critical roles were demonstrated for secreted signals
successive round of patterning and migration. such as epidermal growth factor (EGF)-related pro-
Integration of bone morphogenetic protein teins and bone morphogenetic protein (BMP)-related
(BMP) and epidermal growth factor (EGF) sig- members of the transforming growth factor-␤ (TGF-␤)
nals patterns the elaborate structures of the dor- family. Notch signaling has been implicated in all as-
sal-anterior eggshell. Dev Dyn 2000;218:80 –93. pects of FC morphogenesis. Understanding the succes-
© 2000 Wiley-Liss, Inc. sive interactions of these patterning signals will shed
light on their interactions in morphogenesis of other
Key words: Drosophila melanogaster; follicle cells; tissues and other organisms.
oogenesis; cell migration; pattern
formation OVERVIEW OF OOGENESIS
Each Drosophila egg develops as a discrete structure
INTRODUCTION termed an egg chamber (Fig. 1; reviewed in King, 1970;
Spradling, 1993). Egg chambers are formed at the an-
The Drosophila egg is surrounded by an eggshell
terior of the ovary, and move posteriorly as they de-
with elaborate structures that enhance survival of the
velop. Thus, each ovariole contains a linear array of
developing embryo. This eggshell is the product of the
developmental stages. The follicular epithelium under-
somatic follicle cell epithelium, which develops in con-
cert with the oocyte. Oogenesis requires a series of
interactions between the follicle cells and an underly-
Grant sponsor: American Cancer Society; Grant number: RPG-96-
ing cyst of germ cells. At the same time, distinct do- 103-03-DDC; Grant sponsor: National Institutes of Health; Grant
mains of follicle cells (FCs) are patterned by series of number: GM60501.
local cell-cell interactions. Within each domain, the *Correspondence to: Laurel A. Raftery, Cutaneous Biology Research
Center, MGH-East 3rd Floor, Building 149 13th Street, Charlestown,
cells undergo coordinated shape changes and move- MA 02129. E-mail: laurel.raftery@cbrc2.mgh.harvard.edu
ments, followed by differentiation to create a special- Received 10 January 2000; Accepted 8 February 2000

© 2000 WILEY-LISS, INC.


REGULATION OF FOLLICLE CELL MORPHOGENESIS 81

TABLE 1. Genes Involved in Follicle Cell Morphogenesis

Gene name Ovarian requirementa Protein function Referenceb


EGFR signaling
Epidermal growth factor
receptor (EGFR, torpedo) S EGF receptor tyrosine kinase (RTK) 1
fs(1)K10 (K10) G Helix-turn-helix motif transcription factor 2
gurken (grk) G TGF␣ ligand 3
ras S Small GTPase, RTK signaling 4
Raf G/S Raf kinase, RTK signaling 5
argos (aos) S Secreted, EGFR antagonist 6
rhomboid (rho) S Transmembrane protein, RTK mediator 7
sprouty (sty) S Membrane protein, EGF antagonist 8
pointed (ptd) S ETS domain transcription factor 9
bunched (bun) S TSC-22 family transcription factor 10
Notch signaling
Notch (N) S Cell surface receptor 11
Delta (Dl) S Cell surface ligand 11
big brain (bib) S MIP family motif, aquaporin, channel 11
brainiac (brn) G Novel, secreted 12
daughterless (da) S bHLH motif transcription factor 13
cut S Homeodomain transcription factor 14
fs(1)Yb G Novel 15
toucan (toc) G Novel, coiled-coil motif 16
neuralized (neu) S Zinc finger transcription factor 11
egghead (egh) G Novel, secreted 17
BMP signaling
decapentaplegic (dpp) S BMP ligand 18
thick veins (tkv) S BMP receptor ser-thr kinase 18
saxophone (sax) S,G BMP receptor ser-thr kinase 18
Mothers against dpp (Mad) S Smad transcription factor 19
D-mef2 Myogenic transcription factor 20
Hedgehog signaling
hedgehog (hh) S Ligand 21
costal-2 (cos-2) S Kinesin domain, antagonist 22
cubitus interruptus (ci) S Zinc finger transcription factor 23
patched (ptc) S 7-pass transmembrane protein, antagonist 23
JUN kinase signaling
hemipterous (hep) S JUN kinase 39
puckered (puc) S JUN phosphatase 40
Wnt signaling
wingless (wg) S Wnt family ligand 23
Cytoskeletal organization
zipper (zip) S,G Myosin II heavy chain 25
spaghetti squash (sqh) S Myosin regulatory light chain 26
rap1, Dcdc42, Drac1, DRhoL S/G Small GTPase family members 27, 28
␣-spectrin S ␣-spectrin, membrane cytoskeleton 29
karst S ␤-H-spectrin, membrane cytoskeleton 30
Cell adhesion
Fasciclin III (FasIII) S Septate junctions; transmembrane 11
discs large (dlg) S,G Septate junctions, MAGUK family 31
shotgun (shg) S,G E-cadherin, cell adhesion 32
armadillo (arm) S ␤-catenin 24
Other genes and markers
stall S Unknown 33
slow border cells (slbo) S bZIP transcription factor 34
breathless (btl) S FGF receptor tyrosine kinase 35
93F S Stalk cell marker 33
fs(1)cup G Novel; coiled-coil motif membrane protein 36
bullwinkle (bwk) G HMG protein 37
dicephalic (dic) G Unknown 38
A359 S Centripetally migrating FC marker, unknown 19
a
Abbreviations: (G) Germ line, (S) soma.
b
All genes are described in FlyBase (http://flybase.bio.indiana.edu/) Representative references: 1. Price et al. (1989); 2. Cheung
et al. (1992); 3. Roth and Schüpbach (1994); 4. Lee and Montell (1997); 5. Brand and Perrimon (1994); 6. Wasserman and
Freeman (1998); 7. Ruohola-Baker et al. (1993); 8. Reich et al. (1999); 9. Morimota et al. (1996); 10. Dobens et al. (1997); 11.
Ruohola et al. (1991); 12. Goode et al. (1996a); 13. Cummings and Cronmiller (1994); 14. Jackson and Blochlinger (1997); 15.
King and Lin (1999); 16. Grammont et al. (1997); 17. Goode et al. (1996b); 18. Twombly et al. (1996); 19. Dobens et al. (2000);
20. Mantrova et al. (1999); 21. Forbes et al. (1996a); 22. Liu and Montell (1999); 23. Forbes et al. (1996b); 24. Peifer et al. (1993);
25. Young et al. (1993); 26. Edwards and Kiehart (1996); 27. Asha et al. (1999); 28. Murphy and Montell (1996); 29. Lee et al.
(1997); 30. Zarnescu and Thomas (1999); 31. Goode and Perrimon (1997); 32. Niewiadomska et al. (1999); 33. Tworoger et al.
(1999); 34. Montell et al. (1992); 35. Murphy et al. (1995); 36. Keyes and Spradling (1997); 37. Rittenhouse and Berg (1995);
38. González-Reyes et al. (1997); 39. S. Noselli, personal communication; 40. LLD and LAR, in preparation.
82 DOBENS AND RAFTERY

nurse cells and uptake of yolk produced in the fat body


(reviewed in Spradling, 1993). With the exception of
the polar cells, FC proliferation continues through
stage 6 (Orr-Weaver, 1991; Calvi et al., 1998). During
stages 7 to 10, FCs become polyploid through en-
doreduplication. Specific amplification of the chorion
genes occurs after stage 10B.
The architecture of the eggshell is laid out through a
stereotypic sequence of migrations. During stage 9,
most FCs shift posteriorly to form a columnar epithe-
lium over the oocyte (Fig. 2A–C). During this process,
about 30 anterior FCs flatten to form a squamous epi-
thelium over the nurse cells. Simultaneously, 6 –10
anterior FCs, the border cells, migrate between the
nurse cells to the oocyte. The anterior squamous and
posterior columnar FCs maintain a contiguous epithe-
Fig. 1. Follicle cell migrations in the germarium. Anterior is to the left lium over the germline cyst.
in these and all subsequent figures. Germ cells divide in region 1 to form Follicle cell behaviors are tightly coordinated with
a 16-cell cyst, which flattens as it exits region 2A. Under the influence of events in the germ cells during stage 10B. First, the
the terminal filament, FCs (depicted in gray) proliferate in region 2B, nurse cells dump their contents into the oocyte, causing
where they migrate to contact the posterior side of the flattened cyst. The
cyst is enveloped by FCs to form a stage 1 egg chamber as it leaves
it to swell rapidly. At the same time, FCs at the nurse
region 2B. FCs that do not contact the germ cyst interleave to form stalks cell-oocyte boundary migrate centripetally, to cover the
that connect to adjacent egg chambers at the polar cells. anterior end of the oocyte (Fig. 2D–F). The squamous
FCs shrink in size, maintaining their association with
the degenerating nurse cells. As the oocyte swells,
goes many changes after the egg chamber is formed. mainbody FCs stretch along the anterior/posterior
Terminally differentiated FCs die after depositing the axis, maintaining the epithelium. FCs surrounding the
eggshell. The structural details of the eggshell reveal oocyte deposit the eggshell. Two dorsal anterior FC
the organization of the final FC epithelium. subgroups migrate coordinately to produce the dorsal
appendages of the eggshell. Finally, FCs undergo apo-
Egg Chamber Formation ptosis (Chao and Nagoshi, 1999), and are resorbed as
The egg chamber forms from two distinct cell types: the mature egg exits the ovary.
the somatic follicle cells, derived from the gonadal me-
soderm, and the germ cells. Egg chambers are assem- Structure of the Eggshell
bled at the anterior tip of each ovariole, in a germarium The eggshell (Fig. 3A) is a multi-layered protein
(Fig. 1). Germline stem cells are maintained at the structure deposited by the FCs (Margaritis et al.,
distal tip (Lin and Spradling, 1993). Egg chambers 1980). The inner layers include the vitelline mem-
assemble in an adjacent tube of somatic sheath cells, brane, the waxy layer, and the inner chorion layer
which is divided into three regions. In region 1, the (ICL). The outer layers include the endochorion, which
pro-oocyte arises from an asymmetric stem cell divi- we focus on here, and the exochorion. The proteins that
sion, and undergoes four incomplete cell divisions to comprise each layer are deposited by the FCs under
form a syncytial cyst. As the cyst moves posteriorly strict temporal control. The roof of the endochorion
through region 2a, one germ cell is selected to become shows ridges that correspond to the junctions between
the oocyte. The remaining 15 germ cells become nurse FCs. Thus, FCs leave imprints on the eggshell, so that
cells. The cyst flattens and moves to region 2b, where a the final epithelial organization is evident in the result-
few FC stem cells reside (Margolis and Spradling, ant egg (Fig. 3B).
1995). FCs migrate to encase the cyst in a cuboidal The eggshell has regional specializations, both in its
epithelium. three-dimensional architecture and in the ultrastruc-
As the egg chamber buds off from the germarium, ture of the specific layers (Margaritis et al., 1980). The
five to eight FCs become stalk cells (Fig. 1). These cells most prominent features are the dorsal appendages,
form stacks that separate adjacent egg chambers. Stalk long tubes of chorion that project from the anterior-
cells do not contact the germ cells, and are not part of dorsal surface (Fig. 3C). The chorion of these filaments
the epithelium. Within the epithelium, the polar cells is uniquely porous (Fig. 3C inset), suggesting a role in
become distinct from other FCs (Margolis and Sprad- gas exchange. The anterior-most structure is the oper-
ling, 1995). These cells are important for subsequent culum, a flat plate ringed on three sides by a prominent
patterning of the egg chamber. ridge, the collar (Fig. 3A, E). The operculum plate
arises from the centripetally migrating FCs of stage
Morphogenesis of Eggshell Architecture 10B. The micropyle juts out near the ventral collar
After leaving the germarium, the egg chamber grows (Fig. 3E), and surrounds a canal for sperm entry,
in size. The germ cells enlarge from synthesis in the formed by the border cells. The operculum can be fur-
REGULATION OF FOLLICLE CELL MORPHOGENESIS 83

Fig. 2. Follicle cell rearrangements during mid-oogenesis. Anterior is to the left. ␣-spectrin staining reveals FC shapes (white; ␣-spectrin antiserum
from Pesacreta et al., 1989). A: Stage 8 egg chamber cuboidal epithelium covering nurse cells (NC) and oocyte (O; line marks the anterior edge).
Posterior terminal cells are more rectangular than others. B: Main body epithelium (MBE) changes shape (arrows) and shifts posteriorly (stage 9). C:
Border cells (bc; arrowhead) migrate through the nurse cells; the oocyte increases in size (stage 9). D: Boundary between squamous and columnar
cells (arrow) is the site of presumptive centripetally migrating FCs (CMFC, stage 10A). E: Centripetally migrating FCs further elongate and begin to
cover anterior of oocyte (stage 10B). F: Border cells shift slightly to the dorsal side.

ther subdivided into three regions that form concentric line stem cells can proliferate independently (Gateff,
arcs around the micropyle (Fig. 3E). The collar exhibits 1982; McKearin and Spradling, 1990; Margolis and
ultrastructural perforations in the chorion and Spradling, 1995), but may be coordinated (King and
vitelline membrane and is easily split. Thus, the oper- Lin, 1999). FC proliferation in the germarium is regu-
culum is an escape hatch for the larva. At the posterior lated by a hedgehog (hh) signal produced by the termi-
pole, the aeropyle includes two nested rings of distinct nal filament, and requires the signal transduction com-
cell imprints (Fig. 3D, F). FC patterning specifies the ponents patched and cubitus interruptus in the FCs
domains that will create each eggshell structure; FC (Forbes et al., 1996a; Forbes et al., 1996b). Hh signal-
migrations move these domains to the proper position. ing regulates both FC proliferation and specification of
The specialized ultrastructure of the chorion must the stalk/polar cell precursors, suggesting that these
arise from different programs of gene expression and processes are connected.
protein secretion as the differentiated FCs deposit the
eggshell. Follicle Cell-Germline Interactions Mediate
Cyst Envelopment
FOLLICLE CELL MORPHOGENESIS DURING Assembly of discrete egg chambers requires that FCs
EGG CHAMBER FORMATION recognize individual germline cysts and migrate be-
The formation of the egg chamber involves three tween them. Encapsulation of the germline cyst in-
interdependent processes: 1) FC proliferation from volves both the envelopment of the cyst and the sepa-
stem cells, 2) FC migration and envelopment of a germ- ration of egg chambers by differentiation of stalk cells.
line cyst, and 3) FC differentiation to create the stalks These processes are interdependent, as revealed by
that separate egg chambers. Some defects in these sensitive genetic interactions between several of the
processes lead to egg chambers with two or more com- genes involved.
plete germline cysts; in extreme cases, a single epithe- Proliferating FCs migrate between successive cysts,
lial bag full of cysts forms. Other defects can result in first contacting the posterior portion of the cyst disc
split cysts, in which an egg chamber with too few germ (Fig. 1) (González-Reyes and St. Johnston, 1998b) and
cells occurs next to one with too many. In either case, then surrounding the entire disc. A germ cell to FC
the egg chambers fail to develop, causing reduced fe- signal promotes initial FC migration and patterning;
cundity or sterility. this signal either activates or functions coordinately
with Notch signaling. Notch protein levels are tran-
Regulation of Stem Cell Proliferation siently elevated in posterior FCs of region 2b, and fused
A lifetime of egg production depends on continuous egg chambers are among the phenotypes engendered
production of FCs from a few stem cells in germarium by a temperature-sensitive mutation in Notch (Nts, Xu
region 2b (Fig. 1) (Margolis and Spradling, 1995). Re- et al., 1992). The use of temperature-sensitive muta-
duced FC proliferation results in fused egg chambers tions has been essential to determine the roles of Notch
and gaps in the FC epithelium. The somatic and germ- signaling, because it functions at multiple times during
84 DOBENS AND RAFTERY

Fig. 3. Specialized features of the Drosophila eggshell. (A–F) Scanning electron micrographs of eggshell chorion. A: Dorso-lateral view of entire
egg. Reference line marks the dorsal midline in this and other panels. B: Dorso-lateral close up of midbody imprints (d ⫽ dorsal; v ⫽ ventro-lateral
imprints). C: Close up views of a dorsal respiratory appendage (da). Inset shows the sharp boundary between specialized exochorion along the ventral
and dorsal sides of the appendage (arrowheads). D: Aeropyle at the posterior pole of the eggshell, with two types of distinct imprints, I and II. E: Close
up view of the anterior eggshell showing the micropyle (mp), dorsal appendages (da) and three regions of distinct cell imprints (I–III). F: Close up view
of posterior imprints shows thin eggshell of region I imprints and waved surface of region II imprints. Scanning electron micrographs are reprinted from
Margaritas et al., 1980, with permission from The Company of Biologists, Ltd.

oogenesis (Xu et al., 1992). Another neurogenic gene, cannot migrate between the germ cells within a wild-
brainiac (brn), is required in the germline and genet- type cyst (Rubsam et al., 1998). However, FCs that
ically interacts with Notch (Goode et al., 1996). Notch lack the Cut transcription factor can migrate be-
interacts with two other genes required in the germ- tween germ cells within a cyst (Jackson and Bloch-
line, egghead (egh) and toucan (toc) (Goode et al., linger, 1997), supporting the notion that FCs actively
1996a; Grammont et al., 1997). An alternative hy- distinguish the boundary of a cyst. Strikingly, re-
pothesis is that the germline genes are required to duced Notch levels suppress this phenotype of cut
strengthen germ cell-germ cell adhesion, so that FCs mutants.
REGULATION OF FOLLICLE CELL MORPHOGENESIS 85

The germ cell to FC signal may be DE-cadherin- formation requires additional genes, such as rap1 and
mediated adhesion, which is required for several steps stall (Asha et al., 1999; Tworoger et al., 1999), but their
of cyst envelopment (Oda et al., 1993). Encoded by the specific roles are unknown. Subtle differences in Notch
gene shotgun, DE-cadherin is an integral membrane and hh phenotypes indicate that these signals regulate
protein that mediates cell-cell adhesion through ho- different aspects of stalk formation.
mophilic binding. FCs first contact the posterior of the Polar cells are determined in the germarium, where
disc-shaped germline cyst, where the oocyte lies (Fig. they cease to divide. Visible differentiation of the polar
1). In the nascent egg chamber, DE-cadherin levels are cells occurs during stages 6 – 8, when these cells round
elevated in both the oocyte and the FCs at the anterior up and lose contact with the basal lamina. Polar cells
and posterior poles. Expression is required in both cell exhibit unusual subcellular localization of DE-cadherin
types to maintain the oocyte in this posterior position and the ␤-catenin Armadillo, and are otherwise dis-
(Godt and Tepass, 1998; González-Reyes and St. tinct in their subcellular morphology (Ruohola et al.,
Johnston, 1998b). When the posterior FCs lack DE- 1991; Peifer et al., 1993; Oda et al., 1997; González-
cadherin, the oocyte associates with DE-cadherin-ex- Reyes and St. Johnston, 1998b). The polar cells are the
pressing anterior FCs. The oocyte position determines keystone for FC patterning, for they nucleate subse-
the anterior-posterior polarity of the egg chamber, quent patterning of the terminal FCs (González-Reyes
which subsequently establishes the anterior-posterior and St. Johnston, 1998b; González-Reyes and St.
polarity of the embryo (Godt and Tepass, 1998; Gonzá- Johnston, 1998a).
lez-Reyes and Johnston, 1998).
ESTABLISHMENT OF TERMINAL AND
Encapsulation Involves Determination of Stalk MAINBODY FOLLICLE CELL DOMAINS
Cells and Polar Cells A signal from the oocyte specifies posterior FC iden-
Each egg chamber is attached to its neighbors by a tity, thus polarizing the anterior-posterior (A/P) axis of
stalk that forms as it leaves the germarium (Fig. 1). the epithelium (González-Reyes et al., 1995; Roth et al.,
Stalks are essential to maintain separate egg cham- 1995). Gurken (Grk) a TGF-␣-like ligand, is produced
bers. Fused egg chambers arise from mutations that in the oocyte (Neuman-Silberberg and Schüpbach,
cause the early absence of stalk cell precursors (Ruo- 1993). Both the EGF-receptor (EGFR) (Price et al.,
hola et al., 1991; Forbes et al., 1996a) or the later 1989; Clifford and Schüpbach, 1992) and the Ras signal
degeneration of the stalks (Edwards and Kiehart, transduction pathway are required in the FCs (Brand
1996). The stalks contact the FC epithelium through and Perrimon, 1994; Hsu and Perrimon, 1994; Lee et
two pairs of polar cells, situated at the anterior and al., 1996). In the absence of EGF signaling, posterior
posterior poles of the egg chamber. FCs express anterior FC markers (González-Reyes et
A single precursor cell type gives rise to stalk cells al., 1995; Roth et al., 1995). Conversely, global eleva-
and polar cells (Tworoger et al., 1999). The stalk/polar tion of Ras activity early in oogenesis can induce ante-
precursor is specified by a Hh signal produced from the rior FCs to transiently express the later posterior gene
terminal filament (Forbes et al., 1996a; Forbes et al., expression marker, pointed (ptd) (Lee and Montell,
1996b). Ectopic Hh expression can induce the forma- 1997). Furthermore, expression of constitutively active
tion of ectopic polar and stalk cells at any position in EGFR can induce anterior ptd expression and prevent
the follicular epithelium. The costal2 gene is required anterior decapentaplegic (dpp) expression (Larkin et
in the FC epithelium to prevent the polar cell fate al., 1999). Thus, EGFR signaling prevents posterior
(Forbes et al., 1996; Liu and Montell, 1999; Tworoger et FCs from adopting the anterior fate.
al., 1999). The FCs at the ends of the egg chamber, the terminal
The level of Notch signal is critical for the proper FCs, are distinct from FCs on the lateral surface of the
subdivision of stalk cells and epithelial FCs. Stalk cell egg chamber, the mainbody FCs (Fig. 4). Terminal FCs
marker expression is lost when Notch signal levels are are uniquely competent to express posterior markers in
reduced using temperature sensitive mutations in response to Ras activation. Global elevation of EGFR or
Notch or its ligand Delta (Ruohola et al., 1991). In Ras activity does not induce ptd expression in the
contrast, low level expression of activated Notch in- mainbody FCs (Lee and Montell, 1997; Larkin et al.,
duces extended stalk structures (Larkin et al., 1996). 1999). Further studies led González-Reyes and St.
At higher levels of activated Notch, stalk cells fail to Johnston (1998a) to propose that each pair of polar
fully differentiate. Both stalk cells and polar cells show cells specifies a flanking domain of terminal cells (Fig.
persistent expression of daughterless, which encodes a 4). Establishment of the terminal cells occurs by the
transcription factor required for Notch signaling in end of stage 6, is Notch dependent, and is independent
several tissues (Cummings and Cronmiller, 1994). of A/P axis polarization (González-Reyes and St.
Consistent with this role, daughterless mutations ge- Johnston, 1998a; Larkin et al., 1999). Although termi-
netically enhance both Notch and Delta mutant encap- nal specification is thought to precede A/P polarization,
sulation phenotypes. Other genes, such as fs(1)Yb the gene expression markers currently available for the
(Johnson et al., 1995) and cut (Cummings and Cron- terminal cells reflect A/P patterning. These markers
miller, 1994), show genetic interactions with Notch are first detected during stage 7, and include slow
that implicate them in stalk cell differentiation. Stalk border cells (slbo) and dpp enhancer traps in the ante-
86 DOBENS AND RAFTERY

rior and ptd enhancer traps in the posterior (Lee and bers, the presumptive border cells do not express slbo
Montell, 1997; Larkin et al., 1999). and do not delaminate from the epithelium (González-
Anterior FC become subdivided into multiple fates, Reyes and St. Johnston, 1998a). Specification of the
including the border cells, the squamous FCs and the border cell fate requires slbo, which encodes a bZIP
centripetally migrating FCs (González-Reyes et al., transcription factor (Montell et al., 1992). slbo is re-
1995; Roth et al., 1995; Lee et al., 1996). González- quired for expression of border cell-specific genes in-
Reyes and St. Johnston (1998a) suggest that posterior cluding the breathless (btl) FGF receptor and DE-cad-
terminal cells are subdivided into equivalent domains herin (Niewiadomska et al., 1999).
(Fig. 4), based on their observation that posterior cells Gratuitous Btl/FGF receptor expression rescues bor-
lacking the EGFR show position-specific expression of der cell migration in slbo mutants (Murphy et al.,
anterior markers. These authors propose that each ter- 1995). Because the FGF receptor signals through the
minal domain is patterned so that a series of concentric Ras pathway, further studies used overexpression of
competence domains surrounds the polar cells. constitutively active or dominant negative Ras trans-
Whether anterior or posterior, all terminal cells at a genes (Lee et al., 1996). These studies indicate that the
specific distance from the polar cells have a similar level and timing of Ras activity is critical to maintain
positional identity, and are competent to give rise to migration. Surprisingly, Slbo and Ras play antagonis-
the appropriate anterior cell type. EGFR activation tic roles during the initiation of migration (Lee et al.,
then diverts posterior terminal cells to a posterior fate 1996).
(Fig. 4). Consistent with this proposed positional spec- Migrating border cells form a highly organized clus-
ification, the number and terminal position of the an- ter, with the polar cells surrounded by a rosette of
terior border cells and squamous FCs are similar to the flanking FCs. Specialized cell junctions appear to hold
number and terminal position of the two posterior cell the cluster together as it migrates between the nurse
types that deposit the aeropyle (Fig. 1D, F). The cen- cells (Niewiadomska et al., 1999). Migration involves
tripetally migrating FCs were proposed as an outer- reorganization of the cytoskeleton; expression of dom-
most terminal domain (González-Reyes and St. inant negative Rac can block migration (Murphy and
Johnston, 1998a); however, their identity as a terminal Montell, 1996) and genes for cytoskeletal components
cell type remains in question. Mainbody FCs can ex- such as sqh and karst, are required (Edwards and
press a centripetal migration marker in response to Kiehart, 1996; Zarnescu and Thomas, 1999). Notch
ectopic Dpp (Dobens and Raftery, 1998; Dobens et al., signaling may be active in migrating border cells, for
2000), suggesting that this cell type is an indirect re- Notch protein levels are elevated (Xu et al., 1992).
sult of terminal patterning. Border cell delamination is regulated separately
from migration. discs large (dlg) inhibits delamination,
EPITHELIAL REORGANIZATION DURING but not migration (Goode and Perrimon, 1997). Many
MID-OOGENESIS FCs migrate along the normal border cell route in dlg
During stage 9, FCs reorganize into three physical mutants, even though they do not express border cell
domains: a small cluster of border cells inside the egg markers. In contrast, ectopic border cells do not mi-
chamber, a squamous epithelium over the nurse cells, grate (Liu and Montell, 1999). They delaminate, only to
and a columnar epithelium over the oocyte (Fig. 2). The remain between the FC epithelium and the nurse cells.
border cells detach from the anterior epithelium and These observations suggest that germ cells provide
migrate through the nurse cells to reach the oocyte. At guidance molecules for border cell migration.
the same time, mainbody FCs move posteriorly to form DE-cadherin is essential for migration (Oda et al.,
a columnar epithelium over the oocyte. The remaining 1997; Niewiadomska et al., 1999); furthermore, the
30 – 40 anterior terminal cells flatten over the nurse speed of migration appears to be dependent on DE-
cells. Border cell migration and columnar reorganiza- cadherin levels. Absence of DE-cadherin only in the
tion proceed in concert. Each migration involves many germ cells results in aberrant migration between the
of the same cell biological processes, so that some mu- FC epithelium and the nurse cells. The change in route
tations cause defects in multiple migrations (first noted suggests that border cells preferentially adhere to the
by Schüpbach and Wieschaus, 1986). cells with the highest level of DE-cadherin, indepen-
dent of tissue origin. Migration also requires DE-cad-
Border Cell Recruitment and Migration herin expression in the border cells. However, a cluster
During border cell migration, 5–10 FCs delaminate only needs a few DE-cadherin-expressing cells to per-
from the anterior pole to invade the nurse cell complex mit migration. Strikingly, mosaic clusters that migrate
(Fig. 2C–F). The anterior polar cells recruit flanking always have DE-cadherin positive cells at the leading
FCs to become border cells. Ectopic polar cells can edge of migration. Taken together, these data indicate
recruit additional border cells, but only in the anterior that homophilic adhesion through DE-cadherin is in-
half of the egg chamber (Liu and Montell, 1999). Con- volved in both guidance and movement.
versely, failure to specify the posterior FC fate leads to
posterior expression of early border cell markers, such Mainbody Follicle Cell Reorganization
as slbo (González-Reyes et al., 1995). Notch is impor- Posterior migration of the mainbody FCs is tightly
tant for specification of border cells; in Nts egg cham- linked to formation of a columnar epithelium over the
REGULATION OF FOLLICLE CELL MORPHOGENESIS 87

oocyte. Prior to stage 9, most FCs are cuboidal (Fig. position, the overlying mainbody FCs become columnar
2A). Early in stage 9, FCs over the oocyte are columnar; at stage 10 (González-Reyes and St. Johnston, 1998a).
mainbody FCs become columnar as they move posteri- All data support the model that contact with the oocyte
orly to contact the oocyte (Fig. 2B). At the end of this triggers elongation of posterior and mainbody FCs. The
reorganization, a sharp apposition of columnar and resultant compaction of the posterior epithelium may
squamous cells lies directly over the oocyte-nurse cell provide the force for posterior movement of the main-
junction (Fig. 2C). Local cell-cell adhesion and signal- body cells.
ing between the germline and the FCs is critical to
reorganize the epithelium overlying the oocyte; both Formation of the Anterior
DE-cadherin and Notch pathway genes are important Squamous Epithelium
for this process. Surprisingly, the posterior migration The flattening of anterior terminal FCs is a conse-
does not appear to involve actin-myosin-based motility quence of patterning. A few ectopic squamous cells
(Edwards and Kiehart, 1996), suggesting that this form near the posterior pole when the posterior FC fate
movement is driven by other forces. is not specified (González-Reyes et al., 1995; Roth et al.,
Formation of a columnar epithelium over the oocyte 1995). This indicates that the anterior cells are not
may be regulated differently in the mainbody cells and simply stretched as a secondary consequence of main-
the posterior terminal cells. The Rho family of small body cell migration. It also indicates that a signal from
GTPase proteins regulates cell morphology in many the nurse cells is not necessary for formation of the
systems (Hall, 1998); their roles in FC morphogenesis overlying squamous epithelium. Little is known about
have been examined through the overexpression of the mechanism of cell flattening. These cells remain
dominant negative and constitutively active trans- tightly associated with the nurse cells, thus they are
genes (Murphy and Montell, 1996). Overexpression of also called the nurse cell follicle cells. The nurse cell
dominant negative RhoL results in loss of epithelial FCs do not secrete chorion, and contract as the nurse
structure specifically in the posterior terminal cells; cells degenerate.
these FCs change shape and become multilayered. The The current model for A/P patterning predicts that
balance of Rho family GTPase activity may be critical, absence of a posterior signal leads to flattening of the
for overexpression of constitutively active RhoL pro- anterior terminal cells. However, early over-expression
duces a similar phenotype. However, these observa- of activated Ras is not sufficient to prevent stage 10
tions do not clarify the function of Rho GTPases during anterior cell flattening. Instead, anterior cells can be-
normal reorganization. Loss of ␣-spectrin also causes come columnar at stage 10 when activated Ras is ex-
loss of epithelial polarity specifically in posterior FCs pressed in a slbo heterozygous background (Lee and
(Lee et al., 1997). These authors speculated that the Montell, 1997). Similarly, small clones of posterior cells
curvature of the egg chamber in the posterior is a that lack EGFR can express anterior markers, but the
source of physical strain on the epithelium. In contrast, cells retain columnar morphology (González-Reyes and
the change in mainbody cell shape specifically requires St. Johnston, 1998a). These data suggest that an ante-
␤-H spectrin (Zarnescu and Thomas, 1999). rior signal is necessary for flattening. The BMP ligand
Notch is essential for posterior migration. Notch is Dpp is a candidate for this signal, for it directs other
apically localized in the FCs, with higher levels in the aspects of anterior FC patterning (Twombly et al.,
FCs over the oocyte than in the nurse cell-associated 1996; Deng and Bownes, 1997; Dobens and Raftery,
FCs (Xu et al., 1992). A prolonged period with reduced 1998; Dobens et al., 2000; Peri and Roth, 2000). Dpp is
Notch function, using Nts, leads to a complete block of expressed in the anterior terminal cells before and
reorganization; shorter periods result in migration of during flattening (Fig. 5).
fewer FCs, but otherwise normal reorganization of the
columnar epithelium (Xu et al., 1992). Reduced Notch MORPHOGENESIS OF THE
function can also cause specific loss of posterior epithe- EGGSHELL ARCHITECTURE
lial organization (Ruohola et al., 1991; Goode et al., In the last stages of oogenesis, a tightly timed se-
1996; González-Reyes and St. Johnston, 1998a). quence of patterning interactions and migrations cre-
Cell-cell contacts with the germ cells are critical to ates the elaborate architecture of the eggshell. We will
posterior migration. Reduction of DE-cadherin in the consider these final events in roughly sequential order.
germline causes a delay in posterior migration (Oda et
al., 1997). Similarly, germline expression of toc is es- Dorsal-Anterior Patterning of Columnar
sential for posterior movement (Grammont et al., Follicle Cells
1997). In contrast, reduction of either egh or brn in the Polarization of the dorsal-ventral (D/V) axis is in-
germline causes a significant acceleration of posterior duced by a localized Grk signal from the oocyte (Schüp-
migration, as well as aberrant FC shape (Goode et al., bach, 1987; Neuman-Silberberg and Schüpbach, 1993;
1996). When the oocyte is small, due to growth retar- Roth et al., 1999). Prior to stage 7, the oocyte nucleus
dation caused by fs(2)cup or Bicaudal D mutations, the resides in the extreme posterior of the cell. During
sharp demarcation between the squamous and colum- stages 7 and 8, the oocyte nucleus migrates to an asym-
nar epithelia is lost (Swan and Suter, 1996; Keyes and metric position in the anterior. This repositioning is
Spradling, 1997). When the oocyte resides in a central essential for D/V patterning of the FCs and the result-
88 DOBENS AND RAFTERY

Fig. 4. Axial patterning of the follicle cells. Mainbody FCs are white,
terminal FCs are colored. Polar cells are determined in the germarium
(Margolis and Spradling, 1995). Establishment of terminal cells is Notch-
dependent, and occurs before stage 7 of oogenesis (González-Reyes
and St. Johnston, 1998a; Larkin et al., 1999). Subdivision of the terminal
cells into competence domains is proposed to occur before the anterior
and posterior fates are established (González-Reyes and St. Johnston,
1998a). The posterior follicle cell fate is induced by a Gurken signal from
the oocyte (González-Reyes et al., 1995; Roth et al., 1995). Patterning of
anterior-dorsal eggshell structures involves combinatorial signaling by
EGF and BMP signals (Dobens et al., 2000; Peri and Roth, 2000). The
eggshell diagram is color-coded to indicate the follicle cell population that
makes each structure.

Fig. 5. Flattening of the nurse cell follicle cells. Spectrin staining outlines the FCs (red), propidium iodide
stains nuclear DNA in blue; dpp enhancer trap (nuclear ␤-galactosidase; green) stains nuclei of prospective
squamous follicle cells in cuboidal epithelium at stage 8 (A). Dpp expression persists in these cells as they
flatten over the nurse cell complex during stage 9 (B) and persists through stage 10B (C).

ant embryo (recently reviewed in Ray and Schüpbach, The dorsal Grk signal is required to induce two
1996). The dorsal mainbody FCs are exposed to Grk prominent dorsal-anterior fates, the operculum and the
signal as they pass near the oocyte nucleus in the dorsal appendages. These eggshell structures are also
posterior migration. dependent on anterior patterning. When the oocyte is
REGULATION OF FOLLICLE CELL MORPHOGENESIS 89

Fig. 6. Gene expression domains in the centripetally migrating cells. come to lie adjacent to the forming micropyle at stage 12 (bottom). B:
In all panels, anterior is to the left; immunofluorescence staining reveals A359 enhancer trap marks columnar FCs adjacent to Dpp-expressing
nuclear ␤-galactosidase expression from enhancer traps in red, MYC cells (top), which migrate to cover the anterior of the egg (bottom). C:
antigen expression used to mark FCs in green. A: Enhancer trap in dpp bunched enhancer trap expression is strictly excluded from the A359-
marks nurse cell FCs at stage 10 in this confocal cross section (top); expressing cells (top) and abuts the edge of the operculum after centrip-
dpp-expressing cells are at the leading edge of centripetal migration and etal migration (bottom). Figures reprinted from Dobens et al., 2000.

small, the anterior mainbody FCs do not cover the


oocyte and dorsal appendages are aberrant or lacking
(Swan and Suter, 1996). Several EGFR target genes
are only expressed in response to the dorsal Grk signal
and not in response to the posterior signal (Brand and
Perrimon, 1994; Queenan et al., 1997; Sapir et al.,
1998). Hyperactivation of the EGFR/Ras signal trans-
duction pathway induces ectopic dorsal appendages
only in the anterior eggshell and ectopic gene expres-
sion only in the anterior mainbody FCs (Brand and
Perrimon, 1994). Two models are consistent with these
data. First, a terminal pre-pattern may specify the ring
of cells competent to form dorsal appendages. Second,
an anterior signal from the terminal cells may syner-
gize with the Grk signal to induce dorsal anterior fates.
Experiments in which the EGF-R/Ras signal trans-
duction pathway is activated in grk mutants support
the latter hypothesis. Dorsal target genes are ex-
pressed in a punctate pattern throughout the main-
body cells, and patches of dorsal appendage material
occur randomly throughout the eggshell (Queenan et
al., 1997; Sapir et al., 1998). Similarly, subtle eleva-
Fig. 7. Follicle cell migrations during dorsal appendage formation. tions of EGFR signaling, for example in FC clones
Panels A–C show immunofluorescence staining of egg chambers with
anti-␣-spectrin (red) to show cell outlines, and nuclear ␤-galactosidase
lacking the EGF signaling antagonist Sprouty (Reich et
expressed from bunched-lacZ (green) marks elongating dorsal append- al., 1999), result in ectopic dorsal appendages only
age FCs prominently (arrows). DNA is stained by propidium iodide (blue). along the collar (see also fs(1)K10, Heinlin et al., 1987).
90 DOBENS AND RAFTERY

The BMP ligand Dpp is a good candidate for a col- expressed leucine zipper transcription factor, Bunched,
laborating anterior signal. Dpp is expressed in the an- antagonizes anterior patterning by Dpp (Dobens et al.,
terior terminal cells (Fig. 5; Twombly et al., 1996; Lar- 2000). Similarly, an anteriorly-expressed leucine zip-
kin et al., 1999). High levels of Dpp expand the per transcription factor, Slbo, antagonizes posterior
operculum at the expense of the dorsal appendages patterning directed by activated Ras (Lee et al., 1996).
(Dobens et al., 2000). Moderate elevation of Dpp levels Centripetal migration involves DE-cadherin medi-
can cause duplication of the dorsal appendages along ated recognition between the FCs and the oocyte. Lack
the edge of the collar (Twombly et al., 1996). The Dpp of DE-cadherin in the germline results in FC migration
type I receptor Thickveins and the BMP signal trans- between the nurse cells (Oda et al., 1997; Niewiadom-
ducer Mad (Raftery and Sutherland, 1999) are required ska et al., 1999). Migration also is disrupted when FCs
to repress more posterior gene expression markers in lacking DE-cadherin are in or near the presumptive
the anterior-most columnar follicle cells (Deng and centripetally migrating FCs (Niewiadomska et al.,
Bownes, 1997; Dobens et al., 2000). Intriguingly, Mad 1999). Migration requires myosin II, which accumu-
is also required for expression of the Broad-Complex lates at the apical tips of centripetally migrating cells
genes in the dorsal appendage primordia (Peri and to form a supracellular ring (Edwards and Kiehart,
Roth, 2000). Thus, it appears that combinatorial sig- 1996). Notch accumulates in high levels in the migrat-
naling by BMP and EGF ligands generates the pattern ing cells, and centripetal migration can be blocked in
of anterior eggshell structures. Nts experiments (Xu et al., 1992).
The cells at the leading edge of centripetal migration
Competence for Centripetal Migration come to rest at the border cells. The leading edge cells
At stage 10B, follicle cells migrate centripetally be- express Dpp during the migration and throughout mi-
tween the nurse cells and the oocyte, enclosing the cropyle formation (Fig. 6A; Twombly et al., 1996; Do-
anterior of the egg (Fig. 2E, F). The centripetally mi- bens et al., 2000). Although centripetal migration is
grating FCs arise at the border between the anterior radially symmetrical, the micropyle is situated at the
squamous FCs and the columnar mainbody FCs, which ventral edge of the mature operculum (Fig. 3E). For-
overlies the border between the nurse cells and the mation of the operculum is directed by both EGF and
oocyte (Fig. 2D). This migration occurs concurrently Dpp signals. High levels of Dpp expand an otherwise
with nurse cell dumping, so that the oocyte is capped by normal operculum (Twombly et al., 1996). Overexpres-
FCs after the nurse cells have expelled their contents. sion of an activated EGFR can expand the operculum
It is likely that the timing of centripetal migration is material, but it is aberrantly patterned and lacks a
critical for proper egg formation. If migration is com- collar (Queenan et al., 1997). bunched integrates these
pleted too soon, nurse cell contents may not be com- signals to pattern the collar structure (Dobens et al.,
pletely transferred to the oocyte. 2000). Together, the leading edge cells and the border
Competence to migrate centripetally is a conse- cells create the micropyle of the eggshell. The leading
quence of FC patterning. When the oocyte is small, the edge cells deposit the vitelline membrane and endocho-
junction between anterior terminal and mainbody FCs rion, whereas the border cells extend a process that
lies over the nurse cell complex, rather than the nurse forms the canal (Edwards et al., 1997).
cell-oocyte boundary. This sometimes leads to inappro-
priate migration between nurse cells, so that the egg- Formation of the Dorsal Appendages
shell encloses some nurse cells with the oocyte (Swan Continued EGF ligand signaling is required for for-
and Suter, 1996). Distinct domains of gene expression mation of the dorsal appendages. However, exposure to
markers are visible in the anterior columnar FCs prior the Grk signal is cut off after vitelline membrane dep-
to and during migration (Fig. 6) (Deng and Bownes, osition, which begins at stage 10. The dorsal Grk signal
1997; Dobens et al., 2000). In particular, Dpp is ex- induces local FC expression or activation of two addi-
pressed in the cells that lead migration (Fig. 6A; Twom- tional EGFR ligands, Vein and Spitz (Sapir et al., 1998;
bly et al., 1996); the following cells express the A359 Wasserman and Freeman, 1998). The two dorsal ap-
enhancer trap (Fig. 6B; Dobens et al., 2000). A359 is pendage primordia separate when peak EGFR signal-
expressed in a tight band of anterior columnar FCs, at ing at the dorsal anterior midline induces expression of
the boundary with the squamous FCs, prior to migra- an EGFR inhibitor, Argos (Wasserman and Freeman,
tion (Dobens and Raftery, 1998), thus marking the 1998). These EGFR feedback loops have been reviewed
presumptive centripetally migrating cells. in detail (Stevens, 1998). Argos modulation of EGFR
The bunched gene, which encodes a leucine zipper signaling creates the patch of type III operculum cells
transcription factor, acts in the mainbody FCs to limit that separates the dorsal appendages (Fig. 3E). High
the size of this anterior domain (Dobens et al., 2000). level EGFR signaling continues in two dorso-lateral
EGF signals stimulate bunched expression, but Dpp patches that will migrate to form the dorsal append-
represses bunched expression in anterior columnar ages during stages 11 through 14.
FCs (Fig. 6C). bunched inhibits the operculum fate in The dorso-lateral patches of FCs migrate anteriorly
more posterior mainbody FCs, and regulates expres- over the nurse cell FCs (Fig. 7) and form two stretching
sion of cytoskeletal components involved in migration tubes. This process is easily perturbed, causing dorsal
(LLD and LAR, unpublished data). Thus, a posteriorly- appendage fusions and shortening. FC patterning is
REGULATION OF FOLLICLE CELL MORPHOGENESIS 91

critical; germline expression of bullwinkle is also re- ever, the sequence and timing of subsequent pattern-
quired (Rittenhouse and Berg, 1995). Dorsal append- ing events remains unclear. Are terminal regions cryp-
age formation may be indirectly perturbed when nurse tically subdivided by an early positional signal, or do
cell dumping is blocked (Robinson and Cooley, 1997). later interactions at the terminal-mainbody cell bound-
However, both nurse cell dumping and dorsal append- aries organize new fates? Boundary formation is im-
age formation involve cytoskeletal rearrangements, so portant in creating organizing centers for morphogen-
that some genes may be independently required in both esis of other tissues (Dahmann and Basler, 1999); do
processes. For example, the requirement for myosin cell type boundaries serve as organizing centers in the
light chain in dorsal appendage formation can be sep- FC? Cell sorting has been proposed to be important at
arated in time from the requirement in nurse cell boundaries (Dahmann and Basler, 1999), and many FC
dumping (Edwards and Kiehart, 1996). JUN kinase migrations involve selective adhesion between cells ex-
signaling plays a role in dorsal appendage morphogen- pressing high levels of DE-cadherin. Do boundaries
esis (S. Noselli, personal communication). In contrast, between follicle cell types play a role in initiating mi-
the JUN kinase phosphatase gene puckered acts in the grations? In other tissues, Notch signaling plays a
centripetally migrating cells (LLD, A. Martin-Blanco, prominent role in creating organized boundaries be-
A. Martinez-Arias, F. Kafatos, and LAR, unpublished tween two cell types (Irvine, 1999). Understanding the
data). These observations suggest that the level of Jun multiple roles of Notch in FC patterning will be impor-
kinase signal transduction is important for orchestrat- tant to resolve this question.
ing each of these migrations.
PERSPECTIVE ACKNOWLEDGMENTS

Dissecting the molecular pathways that specify cell We thank W. Petri and L. Margaritis for generously
fate and coordinate movement requires a well-defined sharing electron micrographs of eggshell structures, S.
and simple tissue. The follicle cells provide a geneti- Noselli for discussing unpublished results, and D.
cally tractable system, where mosaic analysis can es- Branton for antisera. We are grateful to Flybase and
tablish the molecules that are necessary for an event The Interactive Fly for providing organized and up-to-
and clonal overexpression can establish the molecules date information on the Web. Members of the Raftery
that are sufficient to induce it. Egg chambers will de- and Perrimon labs provided useful discussions. The
velop in organ culture, so that genetic analysis can be Raftery lab is supported by grants from the American
coupled with in vitro approaches. Through these tech- Cancer Society (RPG-96-103-03-DDC) and NIH
nical advantages, Drosophila follicle cells offer a (GM60501).
unique opportunity to understand the cell biology of
epithelial morphogenesis. REFERENCES
The architecture of the follicle cell epithelium is Asha H, Ruiter ND, Guang M-D, Hariharan I. 1999. The Rap1 GT-
achieved through successive iterations of local induc- Pase functions as a regulator of morphogenesis in vivo. EMBO J
tive interactions between tissue layers, regional pat- 18:605– 615.
Brand A, Perrimon N. 1994. Raf acts downstream of the EGF receptor
terning signals from within the epithelium, and con-
to determine dorsoventral polarity during Drosophila oogenesis.
certed migrations of specific cell types. The regulatory Genes Dev 8:629 – 639.
pathways for these events are the same pathways that Buszczak M, Freeman M, Carlson J, Bender M, Cooley L, Segraves W.
pattern other organs in both invertebrates and verte- 1999. Ecdysone response genes govern egg chamber development
brates. Notch signaling and cadherin-mediated adhe- during mid-oogenesis in Drosophila. Development 126:4581– 4589.
Calvi B, Lilly M, Spradling A. 1998. Cell cycle control of chorion gene
sion are involved in many of the morphogenetic events.
amplification. Genes Dev 12:734 –744.
Late patterning of the FCs involves combinatorial BMP Chao S, Nagoshi R. 1999. Induction of apoptosis in the germline and
and EGFR signaling. follicle cell layers of Drosophila egg chambers. Mech Dev 88:159 –
Key questions that remain for FC morphogenesis 172.
reflect important questions for the field of developmen- Cheung HK, Serano TL, Cohen RS. 1992. Evidence for a highly
selective RNA transport system and its role in establishing the
tal biology. Morphogenesis of the FCs appears to in-
dorsoventral axis of the Drosophila egg. Development 114:653– 661.
volve temporal changes in competence in addition to Clifford R, Schüpbach T. 1992. The torpedo (DER) receptor tyrosine
positional signaling. Are these temporal changes due to kinase is required at multiple times during Drosophila embryogen-
a required sequence of patterning events or does an esis. Development 853– 872.
internal clock regulate events in the FCs? The timing of Cummings C, Cronmiller C. 1994. The daughterless gene functions
nurse cell dumping is regulated by the level of ecdysone together with Notch and Delta in the control of ovarian follicle cell
development in Drosophila. Development 120:381–394.
produced by the germ cells (Buszczak et al., 1999). Dahmann C, Basler K. 1999. Compartment boundaries: at the edge of
Ecdysone signaling may regulate dorsal appendage for- development. Trends Genet 15:320 –326.
mation as well (Tzolovsky et al., 1999). It will be im- Deng WM, Bownes M. 1997. Two signalling pathways specify lo-
portant to determine whether this signal acts as an calised expression of the Broad-Complex in Drosophila eggshell
internal clock to coordinate morphogenesis of the two patterning and morphogenesis. Development. 124:4639 – 4647.
Dobens LL, Hsu TH, Twombly V, Gelbart WM, Raftery LA, Kafatos
cell types. FC. 1997. The Drosophila bunched gene is a homologue of the
The evidence for subdivision of epithelial FCs into growth factor stimulated mammalian TSC-22 sequence and is re-
terminal and mainbody cell types is compelling. How- quired during oogenesis. Mech Dev 65:197–208.
92 DOBENS AND RAFTERY

Dobens L, Raftery L. 1998. Drosophila oogenesis: a model system to Larkin M, Holder K, Yost C, Gininger E, Ruohola-Baker H. 1996.
understand TGF-␤/Dpp directed cell morphogenesis. NY Acad Sci Expression of constitutively active Notch arrests follicle cells at
857:245–247. a precursor stage during Drosophila oogenesis and disrupts
Dobens L, Petersen J, Treisman J, Raftery L. 2000. Drosophila the anterior-posterior axis of the oocyte. Development 132:3639 –
bunched integrates opposing DPP and EGF signals to set the oper- 3650.
culum boundary. Development. 127:745–754. Larkin M, Deng W-M, Holder K, Tworoger M, Clegg N, Ruohola-
Edwards K, Kiehart D. 1996. Drosophila non-muscle myosin II has Baker H. 1999. Role of Notch pathway in terminal follicle cell
multiple essential roles in imaginal disc and egg chamber morpho- differentiation during Drosophila oogenesis. Dev Genes Evol 209:
genesis. Development 122:1499 –1511. 301–311.
Edwards K, Demsky M, Montague R, Weymouth N, Kiehart D. 1997. Lee JK, Brandin E, Branton D, Goldstein LSB. 1997. ␣-spectrin is
GFP-moesin illuminates actin cytoskeleton dynamics in living tis- required for follicle cell monolayer integrity in Drosophila melano-
sue and demonstrates cell shape changes during morphogenesis in gaster. Development 124:353–362.
Drosophila. Dev Biol 191:103–117. Lee T, Montell D. 1997. Multiple Ras signals pattern the Drosophila
Forbes A, Lin H, Ingham P, Spradling A. 1996a. hedgehog is required ovarian follicle cells. Dev Biol 185:25–33.
for the proliferation and specification of ovarian somatic cells prior Lee T, Feig L, Montell D. 1996. Two distinct roles for ras in a devel-
to egg chamber formation in Drosophila. Development 122:1125– opmentally regulated cell migration. Development 122:409 – 418.
1135. Lin H, Spradling A. 1993. Germ line stem cell division and egg
Forbes A, Spradling A, Ingham P, Lin H. 1996b. The role of segment chamber development in transplanted Drosophila germaria. Dev
polarity genes during early oogenesis in Drosophila. Development Biol 159:140 –152.
122:3283–3294. Liu Y, Montell D. 1999. Identification of mutations that cause cell
Gateff E. 1982. Gonial cell neoplasms of genetic origin affecting both migration defects in mosaic clones. Development 126:1869 –1878.
sexes for Drosophila melanogaster. In: Burger M, Weber R, editors. Mantrova EY, Schulz RA, Hsu T. 1999. Oogenic function of the myo-
Embryonic development, part B: cellular aspects. New York: Alan genic factor D-MEF2: negative regulation of the decapentaplegic
R. Liss. p 621– 632. receptor gene thick veins. Proc Natl Acad Sci USA 96:11889 –11894.
Godt D, Tepass U. 1998. Drosophila oocyte localization is mediated by Margaritis L, Kafatos F, Petri W. 1980. The eggshell of Drosophila
differential cadherin-based adhesion. Nature 395:387–391. melanogaster: I. Fine structure of the layers and regions of the
González-Reyes A, Elliott H, St. Johnston D. 1995. Polarization of wild-type eggshell. J Cell Sci 43:1–35.
both major body axes in Drosophila by gurken-torpedo signaling. Margolis J, Spradling A. 1995. Identification and behavior of epithe-
Nature 375:654 – 658. lial stem cells in the Drosophila ovary. Development 121:3797–
González-Reyes A, St. Johnston D. 1998a. Patterning of the follicle 3807.
cell epithelium along the anterior-posterior axis during Drosophila McKearin D, Spradling A. 1990. bag-o-marbles, a Drosophila gene
oogenesis. Development 125:2837–2846. required to initiate both male and female gametogenesis. Genes
González-Reyes A, St. Johnston D. 1998b. The Drosophila AP axis is Dev 4:2242–2251.
polarized by the cadherin-mediated positioning of the oocyte. De- Montell D, Rørth P, Spradling A. 1992. slow border cells, a locus
velopment 125:3635–3644. required for a developmentally regulated cell migration during
Goode S, Perrimon N. 1997. Inhibition of patterned cell shape change oogenesis, encodes Drosophila C/EBP. Cell 121:51– 62.
and cell invasion by discs large during Drosophila oogenesis. Genes Morimota A, Jordan K, Tietze K, Britton J, O’Neill E, Ruohola-Baker
Dev 11:2432–2444. H. 1996. Pointed, an ETS domain transcription factor, negatively
Goode S, Melnick M, Chou T-B, Perrimon N. 1996. The neurogenic regulates the EFG receptor pathway in Drosophila oogenesis. De-
genes egghead and brainiac define a novel signaling pathway es- velopment 122:3745–3754.
sential for epithelial morphogenesis during Drosophila oogenesis. Murphy A, Montell D. 1996. Cell type-specific roles for Cdc42, Rac,
Development 122:3863–3879. and RhoL in Drosophila oogenesis. J Cell Biol 133:617– 630.
Grammont M, Dastugue B, Couderc J-L. 1997. The Drosophila toucan Murphy A, Lee T, Andrews C, Shilo B, Montell D. 1995. The breathless
(toc) gene is required in germline cells for the somatic cell pattern- FGF receptor homologue, a downstream target of Drosophila
ing during oogenesis. Development 124:4917– 4926. C/EBP in the developmental control of cell migration. Development
Hall A. 1998. Rho GTPases and the actin cytoskeleton. Science 279: 121:2255–2263.
509 –514. Neuman-Silberberg FS, Schüpbach T. 1993. The Drosophila dorso-
Heinlin M, Roos C, Cassab A, Mohier E. 1987. Oocyte-specific tran- ventral patterning gene gurken produces a dorsally localized RNA
scription of fs(1)K10: a Drosophila gene affecting dorsal-ventral and encodes a TGF␣-like protein. Cell 75:165–174.
developmental polarity. EMBO J 6:801– 807. Niewiadomska P, Godt D, Tepass U. 1999. DE-Cadherin is required
Hsu J-C, Perrimon N. 1994. A temperature-sensitive MEK mutation for intercellular motility during Drosophila oogenesis. J Cell Biol
demonstrates the conservation of the signaling pathways activated 144:533–547.
by receptor tyrosine kinase pathways. Genes Dev 8:2176 –2187. Oda H, Uemura T, Shiomi K, Nagafuchi A, Tsokita S, Takeichi M.
Irvine K. 1999. Fringe, Notch, and making developmental boundaries. 1993. Identification of a Drosophila homolog of cadherin associated
Curr Opin Genet Dev 9:434 – 441. with Armadillo and essential for embryonic cell-cell adhesion. Dev
Jackson S, Blochlinger K. 1997. cut interacts with Notch and Protein Biol 165:716 –726.
kinase A to regulate egg chamber formation and to maintain germ- Oda H, Uemura T, Takeichi M. 1997. Phenotypic analysis of null
line cyst integrity during Drosophila oogenesis. Development 124: mutants for DE-cadherin and armadillo in Drosophila ovaries re-
3663–3672. veals distinct aspects of their functions in cell adhesion and cy-
Johnson E, Wayne S, Nagoshi R. 1995. fs(1)Yb is required for ovary toskeletal organization. Genes to Cells 2:29 – 40.
follicle cell differentiation in Drosophila melanogaster and has ge- Orr-Weaver T. 1991. Drosophila chorion genes: cracking the egg-
netic interactions with the Notch group of neurogenic genes. Genet- shell’s secret. BioEssays 13:97–105.
ics 140:207–217. Peifer M, Orsulic S, Sweeton D, Wieschaus E. 1993. A role for the
Keyes L, Spradling A. 1997. The Drosophila gene fs(2)cup interacts Drosophila segment polarity gene armadillo in cell adhesion and
with otu to define a cytoplasmic pathway required for the structure cytoskeletal integrity during oogenesis. Development 118:1191–
and function of germ-line chromosomes. Development 124:1419 – 1207.
1431. Peri F, Roth S. 2000. Combined activities of Gurken and Decapen-
King F, Lin H. 1999. Somatic signaling mediated by fs(1)Yb is essen- taplegic specify dorsal chorion structures of the Drosophila egg.
tial for germline stem cell maintenance during Drosophila oogene- Development 127:841– 850.
sis. Development 126:1833–1844. Pesacreta T, Byers T, Dubrueil R, Kiehart D, Branton D. 1989. Dro-
King R. 1970. Ovarian development in Drosophila melanogaster. New sophila spectrin: the membrane skeleton during embryogenesis.
York: Academic Press. J Cell Biol 108:1697–1709.
REGULATION OF FOLLICLE CELL MORPHOGENESIS 93

Price J, Clifford R, Schüpbach T. 1989. The maternal ventralizing Sapir A, Schweitzer R, Shilo B-Z. 1998. Sequential activation of the
locus torpedo is allelic to faint little ball, an embryonic lethal, EGF receptor pathway during Drosophila oogenesis establishes the
and encodes the Drosophila EGF receptor homolog. Cell 56:1085– dorsoventral axis. Development 125:191–200.
1092. Schüpbach T. 1987. Germ line and soma cooperate during oogenesis to
Queenan A, Ghabrial A, Schüpbach T. 1997. Ectopic activation of establish the dorsalventral pattern of egg shell and embryo in
torpedo/Egfr, a Drosophila receptor tyrosine kinase, dorsalizes both Drosophila melanogaster. Cell 49:699 –707.
the eggshell and the embryo. Development 124:3871–3880. Schüpbach T, Wieschaus E. 1986. Maternal-effect mutations altering
Raftery L, Sutherland D. 1999. TGF-␤ family signal transduction in the anterior-posterior pattern of the Drosophila embryo. Roux’s
Drosophila: from Mad to Smads. Dev Biol 210:251–268. Arch Dev Biol 195:302–317.
Ray RP, Schüpbach T. 1996. Intercellular signaling and the polariza- Spradling A. 1993. Developmental genetics of oogenesis. In: Bate
tion of body axes during Drosophila oogenesis. Genes Dev 10:1711– M, Martinez-Arias A, editors. The Development of Drosophila
1723. melanogaster. New York: Cold Spring Harbor Laboratory Press.
Reich A, Sapir A, Shilo B-Z. 1999. Sprouty is a general inhibitor of p 1–70.
receptor tyrosine kinase signaling. Development 126:4139 – 4147. Stevens L. 1998. Twin peaks: Spitz and Argos star in patterning of the
Rittenhouse K, Berg C. 1995. Mutations in the Drosophila gene bull- Drosophila egg. Cell 95:291–294.
winkle cause the formation of abnormal eggshell structures. Devel- Swan A, Suter B. 1996. Role of Bicaudal-D in patterning the Drosoph-
opment 121:3023–3033. ila egg chamber in mid-oogenesis. Development 122:3577–3586.
Robinson DN, Cooley L. 1997. Genetic analysis of the actin cytoskel- Twombly V, Blackman RK, Jin H, Padgett RW, Gelbart WM. 1996.
eton in the Drosophila ovary. Annu Rev Cell Dev Biol 13:147–170. The TGF-␤ signaling pathway is required in Drosophila oogenesis.
Roth S, Neuman-Silberberg FS, Barcelo G, Schüpbach T. 1995. cor- Development 122:1555–1565.
nichon and the EGF receptor signaling process are necessary for Tworoger M, Larkin M, Bryant Z, Ruohola-Baker H. 1999. Mosaic
both anterior-posterior and dorsal-ventral pattern formation in analysis in the Drosophila ovary reveals a common hedgehog-induc-
Drosophila. Cell 81:967–978. ible precursor stage for stalk and polar cells. Genetics 151:739 –748.
Roth S, Jordan P, Karess R. 1999. Binuclear Drosophila oocytes: Tzolovsky G, Deng W, Schlitt T, Bownes M. 1999. The function of the
consequences and implications for dorsal-ventral patterning in Broad-complex during Drosophila melanogaster oogenesis. Genetics
oogenesis and embryogenesis. Development 126:927–934. 153:1371–1383.
Rubsam R, Hollmann M, Simmerl E, Lammermann U, Schafer MA, Wasserman J, Freeman M. 1998. An autoregulatory cascade of EGF
Buning J, Schafer U. 1998. The egghead gene product influences receptor signaling patterns the Drosophila egg. Cell 95:355–364.
oocyte differentiation by follicle cell-germ cell interactions in Dro- Xu T, Caron LA, Fehon RG, Artavanis-Tsakonis S. 1992. The involve-
sophila melanogaster. Mech Dev 72:131– 40. ment of the Notch locus in Drosophila oogenesis. Development
Ruohola H, Bremer KA, Baker D, Swedlow JR, Jan LY, Jan YN. 115:913–922.
1991. Role of neurogenic genes in establishment of follicle cell Zarnescu D, Thomas G. 1999. Apical spectrin is essential for epithelial
fate and oocyte polarity during oogenesis in Drosophila. Cell. morphogenesis but not apicobasal polarity in Drosophila. J Cell Biol
433– 449. 146:1075–1086.

You might also like