SlideShare a Scribd company logo
1 of 8
Download to read offline
Available online at www.sciencedirect.com
Early zebrafish development: It’s in the maternal genes
Elliott W Abrams and Mary C Mullins
The earliest stages of embryonic development in all animals
examined rely on maternal gene products that are generated
during oogenesis and supplied to the egg. The period of
maternal control of embryonic development varies among
animals according to the onset of zygotic transcription and the
persistence of maternal gene products. This maternal
regulation has been little studied in vertebrates, owing to the
difficulty in manipulating maternal gene function and lack of
basic molecular information. However, recent maternal-effect
screens in the zebrafish have generated more than 40 unique
mutants that are providing new molecular entry points to the
maternal control of early vertebrate development. Here we
discuss recent studies of 12 zebrafish mutant genes that
illuminate the maternal molecular controls on embryonic
development, including advances in the regulation of animal–
vegetal polarity, egg activation, cleavage development, body
plan formation, tissue morphogenesis, microRNA function and
germ cell development.
Address
University of Pennsylvania School of Medicine, Department of Cell and
Developmental Biology, 1211 BRB II/III, 421 Curie Blvd, Philadelphia,
PA 19104-6058, United States
Corresponding author: Mullins, Mary C (mullins@mail.med.upenn.edu)
Current Opinion in Genetics & Development 2009, 19:396–403
This review comes from a themed issue on
Pattern formation and developmental mechanisms
Edited by Kathryn Anderson and Kenneth Irvine
Available online 14th July 2009
0959-437X/$ – see front matter
# 2009 Elsevier Ltd. All rights reserved.
DOI 10.1016/j.gde.2009.06.002
Introduction
The zebrafish has emerged as a premiere genetic tool for
studying vertebrate development. In the 1990s forward
genetic screens identified numerous zygotic mutants
defective in key molecules important in early embryonic
development [1–7]. Recently, a major focus has shifted
towards more specialized screens, including the identi-
fication of maternal-effect mutations in adult screens in
the zebrafish. Since the earliest stages of development
are driven primarily by maternal gene products, the
identification of corresponding mutants is crucial to
provide genetic entry points to known maternally con-
trolled processes, which are still poorly understood in
vertebrates.
Maternal-effect screens have identified over 40 mutants
affecting many early developmental processes (Figure 1).
Collectively, these mutants have defects in oocyte de-
velopment [8], egg activation [8,9], embryonic cleavage
[8–11], patterning and morphogenesis [9–11]. New stu-
dies have focused on the molecular identification of the
corresponding mutant genes, a key step to understand-
ing the molecular mechanisms governing the very
earliest stages of embryogenesis. The results have
unveiled novel and known genes with unanticipated
roles in early development. Mutants generated by
reverse genetic TILLING methods in the zebrafish
have also recently revealed important roles for small
noncoding RNA molecules, miRNAs [12] and piRNAs
[13,14], in the regulation of maternal processes in zebra-
fish development.
In this review, we highlight recent contributions to the
molecular regulation of animal–vegetal polarity in the
oocyte and egg, maternal gene regulation of early embryo-
nic patterning, tissue morphogenesis and small noncoding
RNA molecules, which are emerging as important players
in germ line development.
Molecular insights into zebrafish animal–
vegetal polarity
In frogs and fish, the first developmental asymmetry of
the embryo is the animal–vegetal axis, which predicts the
anterior–posterior axis of the embryo. This asymmetry is
established during early stages of oogenesis and is first
marked by formation of the Balbiani body (Bb; also
referred to as the mitochondrial cloud) adjacent to the
germinal vesicle (the oocyte nucleus) in stage I oocytes.
The Bb position predicts the location of the vegetal pole,
but its presence is only transient, as it disassembles by
stage II of oogenesis. The Bb is composed of a collection
of mitochondria, ER, germinal granules and several germ
plasm mRNAs (reviewed in [15]). Recently, a thorough
study recapitulated in zebrafish transgenic constructs the
localization pattern of three Bb-localized transcripts. The
30
UTRs of nanos, vasa and dazl directed their localization
initially to the Bb and then to the vegetal cortex of the
oocyte [16
], probably via the METRO pathway
described in Xenopus [17]. The ensuing distribution at
the vegetal cortex differs among these transcripts: dazl
persists at the vegetal cortex, vasa extends around the
cortex, and nanos becomes unlocalized (Figure 2),
suggesting, unexpectedly, that coordinate localization
of germ plasm components is followed by their redistri-
bution to distinct locations. However, after fertilization of
the egg, these germ plasm RNAs reunite in the blastodisc
at the animal pole, where they localize to the germ plasm
Current Opinion in Genetics  Development 2009, 19:396–403 www.sciencedirect.com
Maternal Zebrafish Development Abrams and Mullins 397
Figure 1
Maternal-effect mutant genes in zebrafish disrupt development at distinct stages. Mutants discussed here are indicated in red. acytokinesis [11],
atomos [9], aura [9], barrette [9], bo peep [9], bedazzled [10], blistered [10], claustro [9], cobblestone [9], emulsion [8], golden gate [9], indivisible [8],
irreducible [8], jumpstart [8], kwai [9], misson impossible [9], nebel [9], over easy [8], poky [10], pollywog [10], pug [10], slow [10], ruehrei [8], souffle [8],
screeching halt [10], sunny side up [8], under repair [9], waldo [9], weeble [9].
Figure 2
mRNA localization during oocyte development. During stage I of oogenesis buc, nanos, vasa and dazl transcripts localize to the Balibani body (Bb,
pink), while cyclinB begins to be localized to the animal pole. By stage II of oogenesis the Bb has disassembled, leaving buc, vasa and dazl mRNAs at
the vegetal cortex, whereas nanos becomes unlocalized, and pou2 becomes localized to the animal pole. Note vasa has a broad vegetal cortical
domain at stage II. By stage III bruno-like and mago nashi become vegetally localized (late pathway); now buc, and Vg1 are localized to the animal pole
and vasa is localized radially at the cortex. Animal (An) pole is to top and vegetal (Ve) to bottom.
www.sciencedirect.com Current Opinion in Genetics  Development 2009, 19:396–403
at the cleavage furrows of the four-cell stage embryo
[16
,18–21].
Three regions within the dazl 30
UTR are crucial for (1)
localization to the Bb, (2) anchoring to the vegetal cortex,
and (3) localization to the cleavage furrows in the early
embryo [16
]. Interestingly, although the 30
UTR of the
nanos-related gene Xcat2 in Xenopus, also localizes it to
the Bb and vegetal cortex in zebrafish, little sequence
similarity is evident in the 30
UTRs of Xcat2, and zebrafish
nanos, dazl and vasa [16
], suggesting either small motifs,
or secondary/tertiary structure of the UTRs are con-
served. Thus, germ plasm mRNA localization occurs in
a stepwise, spatiotemporal fashion, with conserved fea-
tures to the transport systems in Xenopus [16
].
Recent studies have provided genetic access into the
mechanisms of Bb formation and germ plasm assembly
in the oocyte. The maternal-effect mutant bucky ball (buc),
identified through its animal–vegetal polarity egg pheno-
type [8], is defective in Bb formation and early oocyte
polarity, as animal pole markers are expanded radially and
vegetal pole markers are unlocalized [22
,23
]. Buc is
required for early vegetal pole mRNA localization that
occurs via the Bb, as well as late vegetal pole localization
that occurs after Bb dissociation, which is postulated to
depend on the early pathway [24,25]. Thus, the failure of
late vegetal pole mRNA localization in buc mutant
oocytes may be secondary to the lack of Bb formation.
Buc functions to promote vegetal and exclude animal
pole identity also in the surrounding follicle cell layer,
preventing the formation of multiple micropylar cells, an
animal pole-specific follicle cell fate. Thus, patterning of
the oocyte and surrounding somatic follicle cell layer
appears to be coordinated through an as yet unknown
signaling mechanism [22
].
Recently the molecular identity of buc was determined to
be a novel 639 amino acid protein [23
], first identified as
XVelo1 in Xenopus [26]. The predicted Buc protein
contains no known functional motifs. The localization
of buc mRNA is dynamic during oocyte development,
initially localizing to the Bb, moving vegetally and then
ultimately localizing to the animal pole during late
oogenesis [23
] (Figure 2). A Buc-GFP fusion also loca-
lizes to the Bb and, interestingly, to the germ plasm of the
embryo and can induce ectopic germ cells in the embryo
[23
]. Buc homologues in other vertebrates have low
predicted amino acid conservation, indicating that it
has evolved at a relatively accelerated rate. Interestingly,
the human version lacks a complete open reading frame,
suggesting that it has lost its function all together in
humans or functions as an RNA. In zebrafish Buc clearly
functions as a protein, since a nonsense mutation trun-
cating it by just 30 amino acids causes a failure in Bb
formation [23
]. However, the buc RNA could have
additional functions in Bb formation and animal–vegetal
polarity, similar to oskar in Drosophila [27], also a germ
plasm component. Future structure-function exper-
iments of the buc RNA/protein, together with the iso-
lation of interacting partners will unravel mechanistically
how this novel gene functions.
Egg activation molecular genetics
In zebrafish egg activation is marked by cortical granule
exocytosis (CGE), chorion elevation and the segregation
of cytoplasm from the yolk to the animal pole to form the
single cell blastodisc. Embryos derived from brom bones
(brb) mutant females are defective in this process [28].
The egg activation defect is a result of failure of inositol
1,4,5-triphosphate (IP3) signaling, which induces a Ca2+
wave crucial for normal egg activation in all animals
examined. The reinstatement of either IP3 or Ca2+
in
brb mutant eggs can rescue the egg activation defect [28].
brb was shown to encode heterogeneous nuclear ribonu-
cleoprotein I (hnRNP I), probably regulating during
oogenesis the production of an egg activation signaling
component. hnRNP I has been previously shown to be
important in a variety of developmental processes in-
cluding translational control in oogenesis [29], spermato-
genesis [30] and RNA localization [31–34]. Thus, brb
mutants reveal yet another developmental function for
hnRNP I, that is, a role in egg activation.
Cleavage stage molecular genetics
Maternal-effect screens have yielded a significant num-
ber of mutations affecting cleavage development [8–11],
although most of the mutant genes have yet to be cloned.
cellular atoll (cea) mutants fail to undergo cleavages at the
second cell division and beyond [8,35], and was recently
shown to encode the centriolar component sas-6 [35].
Interestingly, as sperm normally provide the centriole to
the zygote, cea also has a paternal-effect function,
whereby wild-type eggs fertilized with cea mutant sperm
result in inviable tetraploid embryos. Since the cleavage
stage of development is primarily under maternal control,
the eventual molecular identification of other maternal-
effect mutant genes functioning during this period will
enhance our understanding of this important stage of
development.
The maternal-effect cellular island (cei) mutant displays an
early defect in cleavage furrow formation [8]. cei encodes a
hypomorphic allele of Aurora B Kinase [36], a protein
previously shown to be important in several aspects of cell
division in other systems (reviewed in [37]). Aurora B
Kinase function is vital zygotically later in zebrafish
embryogenesis, as a null retroviral insertional allele
reveals furrow defects during this period [36,38]. Inter-
estingly, the maternal-effect cei allele causes a specific
defect in distal furrow formation during the early cleavage
stage, while medially positioned furrows can form, prob-
ably owing to intact mitotic spindle-derived signals med-
iating medial furrow formation [36]. The ability of the cei
398 Pattern formation and developmental mechanisms
Current Opinion in Genetics  Development 2009, 19:396–403 www.sciencedirect.com
mutant Aurora B Kinase protein to mediate all its cyto-
kinesis functions zygotically, but its inability to mediate
distal furrow formation in the cleavage embryo may be
related to unique requirements for astral microtubules
and Aurora B Kinase to divide the large cells of the very
early embryo [36].
Molecular genetic advances in maternal
regulated patterning and morphogenesis
In fish and amphibians, the dorsoventral embryonic axis is
established through a maternally regulated Wnt/bcatenin
pathway. Several zebrafish maternal-effect mutants with
defects in dorsoventral axis formation have been ident-
ified. The maternal-effect ventralized mutants hecate and
tokkaebe produce embryos lacking a dorsal organizer and
consequently are radially ventralized. Although hecate and
tokkaebe have not yet been molecular defined, injection/
rescue experiments with Wnt signaling pathway com-
ponents indicates that they act upstream of, or within a
Wnt/bcatenin signaling pathway, respectively, to induce
the dorsal organizer [39–41]. tokkaebe probably corre-
sponds to a novel component of the Wnt pathway, since
no known Wnt components are found within the 0.5 cM
interval containing the tokkaebe mutation [41].
The maternal-effect ventralized mutant ichabod is caused
by a specific loss of maternal bcatenin2 function [42].
Two bcatenin genes have been identified in the zebrafish
genome; however, only bcatenin2 is essential maternally
to induce the dorsal organizer through a Wnt signaling
pathway. Interestingly, the maternal, dorsal organizer
Wnt/bcatenin2 pathway also functions to repress expres-
sion of a later Wnt8/bcatenin pathway that opposes dorsal
specification ventrally [42] (Figure 3). In zebrafish this
late blastula Wnt pathway depends on both bcatenin1 and
bcatenin2 function and is mediated by zygotic Wnt8
signaling ventrally [42–45]. Thus in the absence of both
bcatenin1 and bcatenin2 function, early dorsal organizer
formation fails; however, at later stages the ventral Wnt8
pathway also fails to oppose dorsal fate specification
ventrally. The result is the formation of dorsal tissues
circumferentially. These results show, surprisingly, that
at a late blastula stage circumferential organizer-like
tissue can form in the absence of a maternal Wnt/bcatenin
pathway and independently of Wnt/bcatenin signaling
entirely.
The transcription factor pou2/oct4 also acts maternally in
dorsoventral patterning and morphogenesis [46,47],
revealing additional roles to its described maternal func-
tion in endoderm specification [48,49]. Loss of both
maternal and zygotic (MZ) Pou2 causes severe dorsaliza-
tion owing to failure to induce bmp ligand gene expres-
sion, which functions in ventrolateral tissue specification
[46]. These results demonstrate that ventral specification
via the BMP signaling pathway is not a default pathway,
as previously thought, but instead is initiated by maternal
Pou2/Oct4 in the early embryo [46] (Figure 3). MZPou2
Maternal Zebrafish Development Abrams and Mullins 399
Figure 3
The role of BMP and early and late Wnt signaling in dorsoventral patterning. (a) Maternal Pou2 induces early zygotic bmp ligand expression. bmp
expression is initially present throughout the blastoderm (grey). An early maternal Wnt signal from the yolk cell promotes future dorsal organizer (DO)
formation. (b) By the late blastula stage (50% epiboly), a BMP activity gradient (highest ventrally (black)) is established that promotes ventral fates. At
this stage Wnt 8 signaling opposes dorsal fate specification ventrally. Ventral is positioned to the left and dorsal to the right.
www.sciencedirect.com Current Opinion in Genetics  Development 2009, 19:396–403
also regulates the morphogenic process of epiboly, the
thinning and spreading of the blastoderm over the yolk
cell, through functions in yolk cell microtubule formation,
cell adhesive properties and blastoderm cell movements
via a cell non-autonomous mechanism [46,47]. Together,
these studies reveal that the renowned pou2/oct4 stem cell
gene in mammals is a key maternal regulator of early
zebrafish development.
The betty boop (bbp) mutant, identified in a maternal-
effect screen, is a strictly, maternally acting gene reg-
ulating the morphogenic process of epiboly [10,50
].
Embryos from bbp mothers develop normally until they
reach 50% epiboly at which point the embryo abruptly
bursts via a presumptive premature constriction of the
actin cytoskeleton in the yolk cell [50
]. Interestingly,
bbp was recently shown to encode the zebrafish homol-
ogue of Mitogen Activated Protein Kinase Activated
Protein Kinase 2 (MAPKAPK2), a target of p38 MAP
kinase (MAPK) in cell culture systems [51]. During
zebrafish epiboly, p38 MAPK also appears to activate
MAPKAPK2, as a dominant-negative p38 MAPK causes
the same epiboly defect as loss of bbp [50
]. Neither p38
MAPK, nor MAPKAPK2 have been previously impli-
cated in tissue morphogenesis. Thus, the identification
and cloning of the bbp gene is a model genetic case for an
unexpected pathway being placed in a developmental
process, in this case epiboly, which may not have
been considered in a candidate-gene reverse genetic
approach.
Small RNA molecules in early development
Small noncoding RNAs are emerging as important
players in early zebrafish development. Maternal-zygo-
tic (MZ) mutant embryos of the microRNA (miRNA)-
processing enzyme Dicer exhibit early embryonic
defects in gastrulation, somitogenesis, brain morpho-
genesis and heart development [52]. By removing the
strong maternal component of dicer, MZ-dicer mutant
embryos are completely devoid of miRNA processing
and therefore, devoid of all miRNA function. miRNAs
negatively regulate target genes by binding to their
30
UTR, promoting deadenylation, translational repres-
sion and/or ultimately degradation of the transcript
(reviewed in [53]). In zebrafish, the predominantly
expressed miRNA during early embryogenesis is miR-
430, which is first expressed at the mid-blastula tran-
sition (MBT), and is not expressed maternally [54]. A
central finding revealed by the loss of miR-430 through
MZ-dicer is its role in the clearance of maternal mRNAs
at the MBT. In MZ-dicer mutant embryos, maternal
mRNAs abnormally persist beyond the MBT [12]. To
date, no maternal miRNAs have been reported in the
zebrafish embryo [54]. Thus, it is unclear whether the
maternal role of dicer is to primarily process miR-430 at
the MBT or in addition, also process unidentified
maternal miRNA(s).
Inhibition of miRNA function plays a role in germ line
development. nanos1 is expressed early in germ line
development and a mutant, generated through TIL-
LING methods, demonstrates its maternal requirement
for PGC survival and a function in the adult in maintain-
ing oocyte production [55]. nanos1 is resistant to miRNA
repression in the germ line, but not in the soma, promot-
ing its specific expression in the germ line [56]. This
resistance is conferred, at least partly, by the maternally
expressed germ cell-localized RNA binding protein Dead
End (Dnd), which interacts with the nanos1 30
UTR,
presumably blocking the binding of miR-430 [57
]. Inter-
estingly, this resistance to miRNA repression is also found
in another germ plasm mRNA, Tudor-domain-contain-
ing-7 (Tdrd-7) [56], suggesting a general mechanism
involving Dnd in influencing germ cell-specific gene
expression (Figure 4).
Germ plasm mRNA resistance to miRNA regulation is
not universal, however, as the dnd 30
UTR itself lacks a
miR-430 site and therefore is presumably not repressed in
the soma by miRNAs [58]. Since dnd maternal transcripts
are dramatically eliminated in the soma about one hour
after the MBT [59], independently of miR-430 regula-
tion, it suggests the existence of additional mode(s) of
modulating maternal transcripts at this crucial develop-
400 Pattern formation and developmental mechanisms
Figure 4
miRNA regulation of germ line development. In the soma miR-430
inhibits expression of germ line specific genes (GLSGs). In the germ line
Dnd blocks miR-430-mediated repression of GLSGs. Note that an
unidentified factor X is required in the soma to silence GLSGs that are
not regulated by miR-430. Soma shown in grey; germ line in purple.
Current Opinion in Genetics  Development 2009, 19:396–403 www.sciencedirect.com
mental transition. Likewise, vasa removal from the soma
is also independent of miR-430 regulation [56]. Thus, an
unknown, possibly common additional mechanism elim-
inates dnd and vasa transcripts from the soma.
A Dicer-independent class of small noncoding RNAs,
known as piRNAs or Piwi associated RNAs, appear to
be germ line specific [13,14,60,61]. Piwi proteins are
important for target gene silencing [62]. Presumptive
null, zygotic mutations in either of two piwi homologues
in zebrafish, ziwi and zili, cause the progressive loss of
germ cells between three and seven weeks of age [13,14].
Mutant adults are phenotypically male, consistent with
recent studies demonstrating that zebrafish develop phe-
notypically as male when eliminating the germ line
[13,63,64]. Interestingly, there is a maternal-effect meio-
tic progression defect in zili hypomorphic mutants.
Although mutant eggs can be fertilized, they fail to
undergo meiosis I and II [13]. Piwi proteins in the mouse
function to repress transposon activity in the germ line
[65,66]. The meiotic defects in zili hypomorphs mutants,
however, are not linked to increased transposon activity as
measured by quantitative RT-PCR and in situ hybridiz-
ation [13]. These findings reveal a novel function for
piRNAs, whose function in vertebrates was previously
thought confined to regulating transposon activity in the
germ line [13].
Future outlook
A major hindrance to the molecular cloning of chemically
induced mutant genes in zebrafish is the incomplete
assembly of the genomic sequence. This obstacle is
becoming less of an issue with recent improvements to
the assembly (currently Zv8; www.sanger.ac.uk). As exist-
ing gaps are eliminated and the genome sequence is
completed, the molecular cloning of maternal-effect
mutant genes will be greatly accelerated.
The investigation of the maternal functions of essential
zygotic genes will be more difficult to study. Although
germ line chimeric analysis is a successful method to
examine the maternal function of zygotic lethal genes in
zebrafish [67], it is quite labour intensive and would be
impractical for large throughput analysis of such maternal
function. This problem may be overcome by employing
techniques that utilize mitotic recombination, a principle
heavily relied upon in Drosophila genetics [68]. RecQ
helicases are known to prevent recombination during
replication [69]. Induced mitotic recombination through
the suppression of RecQ helicases was recently demon-
strated in zebrafish [70]. This approach can generate
mutant clones from heterozygous cells at a significant
frequency (1.7–3.4%) [70]. With further improvements
in this technology, mitotic recombination could poten-
tially be used to generate homozygous mutant germ line
clones of zygotic lethal mutations from heterozygous
individuals. Such germ line mosaic females would gen-
erate maternally deficient eggs and embryos, allowing the
study of the maternal gene function.
Reverse genetic techniques are very valuable comp-
lements to forward genetic approaches in the zebrafish.
Antisense morpholino oligos are widely used to block
translation or disrupt splicing [71,72]. However, the func-
tion of maternal protein already present in the egg cannot
be blocked by morpholino injection into the egg,
although advances in oocyte cell culture methods may
make it possible to use this method in the future to
examine maternal gene functions. Reverse genetic
approaches that rely on induction of genomic sequence
alterations are considerably more laborious, but generate
robust loss-of-function reagents. Recently, the TILLING
(Target Induced Local Lesions in Genomes) approach
[13,14,55], as well as the zinc finger nuclease approach
designed to mutate a specific sequence of the genome
[73–75], have been very successful in generating mutants
in zebrafish. Hence, TILLING and zinc finger nuclease
strategies look very promising for eliminating gene func-
tion of suspected maternal-effect genes in zebrafish in the
future.
Acknowledgements
We thank Lee Kapp for comments on the manuscript, and Eric Weinberg
and Mate Varga for helpful discussion. Funding was provided by NIH grant
HD050901 to MCM.
References and recommended reading
Papers of particular interest, published within the period of review,
have been highlighted as:
 of special interest
 of outstanding interest
1. Driever W, Solnica-Krezel L, Schier AF, Neuhauss SC, Malicki J,
Stemple DL, Stainier DY, Zwartkruis F, Abdelilah S, Rangini Z et al.:
A genetic screen for mutations affecting embryogenesis in
zebrafish. Development 1996, 123:37-46.
2. Hammerschmidt M, Pelegri F, Mullins MC, Kane DA, Brand M, van
Eeden FJ, Furutani-Seiki M, Granato M, Haffter P, Heisenberg CP
et al.: Mutations affecting morphogenesis during gastrulation
and tail formation in the zebrafish, Danio rerio. Development
1996, 123:143-151.
3. Kane DA, Hammerschmidt M, Mullins MC, Maischein HM,
Brand M, van Eeden FJ, Furutani-Seiki M, Granato M, Haffter P,
Heisenberg CP et al.: The zebrafish epiboly mutants.
Development 1996, 123:47-55.
4. Kane DA, Maischein HM, Brand M, van Eeden FJ, Furutani-
Seiki M, Granato M, Haffter P, Hammerschmidt M, Heisenberg CP,
Jiang YJ et al.: The zebrafish early arrest mutants. Development
1996, 123:57-66.
5. Mullins MC, Hammerschmidt M, Kane DA, Odenthal J, Brand M,
van Eeden FJ, Furutani-Seiki M, Granato M, Haffter P,
Heisenberg CP et al.: Genes establishing dorsoventral pattern
formation in the zebrafish embryo: the ventral specifying
genes. Development 1996, 123:81-93.
6. Solnica-Krezel L, Stemple DL, Mountcastle-Shah E, Rangini Z,
Neuhauss SC, Malicki J, Schier AF, Stainier DY, Zwartkruis F,
Abdelilah S et al.: Mutations affecting cell fates and cellular
rearrangements during gastrulation in zebrafish. Development
1996, 123:67-80.
7. Stemple DL, Solnica-Krezel L, Zwartkruis F, Neuhauss SC,
Schier AF, Malicki J, Stainier DY, Abdelilah S, Rangini Z,
Maternal Zebrafish Development Abrams and Mullins 401
www.sciencedirect.com Current Opinion in Genetics  Development 2009, 19:396–403
Mountcastle-Shah E et al.: Mutations affecting development of
the notochord in zebrafish. Development 1996, 123:117-128.
8. Dosch R, Wagner DS, Mintzer KA, Runke G, Wiemelt AP,
Mullins MC: Maternal control of vertebrate development before
the midblastula transition: mutants from the zebrafish I. Dev
Cell 2004, 6:771-780.
9. Pelegri F, Dekens MP, Schulte-Merker S, Maischein HM, Weiler C,
Nusslein-Volhard C: Identification of recessive maternal-effect
mutations in the zebrafish using a gynogenesis-based
method. Dev Dyn 2004, 231:324-335.
10. Wagner DS, Dosch R, Mintzer KA, Wiemelt AP, Mullins MC:
Maternal control of development at the midblastula transition
and beyond: mutants from the zebrafish II. Dev Cell 2004,
6:781-790.
11. Kishimoto Y, Koshida S, Furutani-Seiki M, Kondoh H: Zebrafish
maternal-effect mutations causing cytokinesis defect without
affecting mitosis or equatorial vasa deposition. Mech Dev
2004, 121:79-89.
12. Giraldez AJ, Mishima Y, Rihel J, Grocock RJ, Van Dongen S,
Inoue K, Enright AJ, Schier AF: Zebrafish MiR-430 promotes
deadenylation and clearance of maternal mRNAs. Science
2006, 312:75-79.
13. Houwing S, Berezikov E, Ketting RF: Zili is required for germ cell
differentiation and meiosis in zebrafish. EMBO J 2008, 27:2702-
2711.
14. Houwing S, Kamminga LM, Berezikov E, Cronembold D, Girard A,
van den Elst H, Filippov DV, Blaser H, Raz E, Moens CB et al.: A
role for Piwi and piRNAs in germ cell maintenance and
transposon silencing in Zebrafish. Cell 2007, 129:69-82.
15. Kloc M, Bilinski S, Etkin LD: The Balbiani body and germ
cell determinants: 150 years later. Curr Top Dev Biol 2004,
59:1-36.
16.

Kosaka K, Kawakami K, Sakamoto H, Inoue K: Spatiotemporal
localization of germ plasm RNAs during zebrafish oogenesis.
Mech Dev 2007, 124:279-289.
A very thorough analysis of the 30
UTR of dazl involving a series of
transgenic animals demonstrates that distinct regions are required for
successive steps in dazl localization during early oogenesis and in germ
plasm localization in the embryo, implicating a more general utility of the
early Xenopus METRO oogenesis localization pathway in zebrafish.
17. Kloc M, Etkin LD: Two distinct pathways for the localization of
RNAs at the vegetal cortex in Xenopus oocytes. Development
1995, 121:287-297.
18. Theusch EV, Brown KJ, Pelegri F: Separate pathways of RNA
recruitment lead to the compartmentalization of the zebrafish
germ plasm. Dev Biol 2006, 292:129-141.
19. Knaut H, Pelegri F, Bohmann K, Schwarz H, Nusslein-Volhard C:
Zebrafish vasa RNA but not its protein is a component of the
germ plasm and segregates asymmetrically before germline
specification. J Cell Biol 2000, 149:875-888.
20. Koprunner M, Thisse C, Thisse B, Raz E: A zebrafish nanos-
related gene is essential for the development of primordial
germ cells. Genes Dev 2001, 15:2877-2885.
21. Yoon C, Kawakami K, Hopkins N: Zebrafish vasa homologue
RNA is localized to the cleavage planes of 2- and 4-cell-stage
embryos and is expressed in the primordial germ cells.
Development 1997, 124:3157-3165.
22.

Marlow FL, Mullins MC: Bucky ball functions in Balbiani body
assembly and animal-vegetal polarity in the oocyte and follicle
cell layer in zebrafish. Dev Biol 2008, 321:40-50.
This is a study of the first mutant in vertebrates known to be defective in
Balbiani formation, the earliest oocyte asymmetric structure in verte-
brates, which is conserved from insects to mammals and first described
over 150 years ago. The analysis implicates this gene in setting up the
animal–vegetal axis of early oocytes and shows that patterning of the
oocyte and surrounding follicle cell layer is coordinated, probably through
a signaling mechanism.
23.

Bontems F, Stein A, Marlow F, Lyautey J, Gupta T, Mullins MC,
Dosch R: Bucky ball organizes germ plasm assembly in
zebrafish. Curr Biol 2009, 19:414-422.
This study identifies the molecular nature of the bucky ball gene as a
novel, rapidly evolving gene and demonstrates that its overexpression
can result in ectopic germ cells, implicating it in germ plasm assembly.
24. Kloc M, Larabell C, Etkin LD: Elaboration of the messenger
transport organizer pathway for localization of RNA to the
vegetal cortex of Xenopus oocytes. Dev Biol 1996, 180:119-130.
25. King ML, Messitt TJ, Mowry KL: Putting RNAs in the right place
at the right time: RNA localization in the frog oocyte. Biol Cell
2005, 97:19-33.
26. Claussen M, Pieler T: Xvelo1 uses a novel 75-nucleotide signal
sequence that drives vegetal localization along the late
pathway in Xenopus oocytes. Dev Biol 2004, 266:270-284.
27. Jenny A, Hachet O, Zavorszky P, Cyrklaff A, Weston MD,
Johnston DS, Erdelyi M, Ephrussi A: A translation-independent
role of oskar RNA in early Drosophila oogenesis. Development
2006, 133:2827-2833.
28. Mei W, Lee KW, Marlow F, Miller AL, Mullins MC: hnRNP I/brom
bones is required to generate the Ca2+
signal that causes egg
activation in the zebrafish. Development, in press.
29. Besse F, Lopez de Quinto S, Marchand V, Trucco A, Ephrussi A:
Drosophila PTB promotes formation of high-order RNP
particles and represses oskar translation. Genes Dev 2009,
23:195-207.
30. Robida MD, Singh R: Drosophila polypyrimidine-tract binding
protein (PTB) functions specifically in the male germline.
EMBO J 2003, 22:2924-2933.
31. Cote CA, Gautreau D, Denegre JM, Kress TL, Terry NA, Mowry KL:
A Xenopus protein related to hnRNP I has a role in cytoplasmic
RNA localization. Mol Cell 1999, 4:431-437.
32. Kress TL, Yoon YJ, Mowry KL: Nuclear RNP complex assembly
initiates cytoplasmic RNA localization. J Cell Biol 2004,
165:203-211.
33. Czaplinski K, Mattaj IW: 40LoVe interacts with Vg1RBP/Vera
and hnRNP I in binding the Vg1-localization element. RNA
2006, 12:213-222.
34. Lewis RA, Gagnon JA, Mowry KL: PTB/hnRNP I is required for
RNP remodeling during RNA localization in Xenopus oocytes.
Mol Cell Biol 2008, 28:678-686.
35. Yabe T, Ge X, Pelegri F: The zebrafish maternal-effect gene
cellular atoll encodes the centriolar component sas-6 and
defects in its paternal function promote whole genome
duplication. Dev Biol 2007, 312:44-60.
36. Yabe T, Ge X, Lin R, Nair S, Runke G, Mullins MC, Pelegri F: The
maternal-effect gene cellular island encodes aurora B kinase
and is essential for furrow formation in the early zebrafish
embryo. PLoS Genet 2009, 5:e1000518.
37. Vader G, Medema RH, Lens SM: The chromosomal passenger
complex: guiding Aurora-B through mitosis. J Cell Biol 2006,
173:833-837.
38. Golling G, Amsterdam A, Sun Z, Antonelli M, Maldonado E,
Chen W, Burgess S, Haldi M, Artzt K, Farrington S et al.:
Insertional mutagenesis in zebrafish rapidly identifies genes
essential for early vertebrate development. Nat Genet 2002,
31:135-140.
39. Lyman Gingerich J, Westfall TA, Slusarski DC, Pelegri F: hecate, a
zebrafish maternal effect gene, affects dorsal organizer
induction and intracellular calcium transient frequency. Dev
Biol 2005, 286:427-439.
40. Lyman Gingerich J, Lindeman R, Putiri E, Stolzmann K, Pelegri F:
Analysis of axis induction mutant embryos reveals
morphogenetic events associated with zebrafish yolk
extension formation. Dev Dyn 2006, 235:2749-2760.
41. Nojima H, Shimizu T, Kim CH, Yabe T, Bae YK, Muraoka O, Hirata T,
Chitnis A, Hirano T, Hibi M: Genetic evidence for involvement of
maternally derived Wnt canonical signaling in dorsal
determination in zebrafish. Mech Dev 2004, 121:371-386.
42. Bellipanni G, Varga M, Maegawa S, Imai Y, Kelly C, Myers AP,
Chu F, Talbot WS, Weinberg ES: Essential and opposing roles of
402 Pattern formation and developmental mechanisms
Current Opinion in Genetics  Development 2009, 19:396–403 www.sciencedirect.com
zebrafish beta-catenins in the formation of dorsal
axial structures and neurectoderm. Development 2006,
133:1299-1309.
43. Erter CE, Wilm TP, Basler N, Wright CV, Solnica-Krezel L: Wnt8 is
required in lateral mesendodermal precursors for neural
posteriorization in vivo. Development 2001, 128:3571-3583.
44. Lekven AC, Thorpe CJ, Waxman JS, Moon RT: Zebrafish wnt8
encodes two wnt8 proteins on a bicistronic transcript and is
required for mesoderm and neurectoderm patterning. Dev Cell
2001, 1:103-114.
45. Varga M, Maegawa S, Bellipanni G, Weinberg ES: Chordin
expression, mediated by Nodal and FGF signaling, is
restricted by redundant function of two beta-catenins in the
zebrafish embryo. Mech Dev 2007, 124:775-791.
46. Reim G, Brand M: Maternal control of vertebrate dorsoventral
axis formation and epiboly by the POU domain protein Spg/
Pou2/Oct4. Development 2006, 133:2757-2770.
47. Lachnit M, Kur E, Driever W: Alterations of the cytoskeleton in all
three embryonic lineages contribute to the epiboly defect of
Pou5f1/Oct4 deficient MZspg zebrafish embryos. Dev Biol
2008, 315:1-17.
48. Lunde K, Belting HG, Driever W: Zebrafish pou5f1/pou2,
homolog of mammalian Oct4, functions in the endoderm
specification cascade. Curr Biol 2004, 14:48-55.
49. Reim G, Mizoguchi T, Stainier DY, Kikuchi Y, Brand M: The POU
domain protein spg (pou2/Oct4) is essential for endoderm
formation in cooperation with the HMG domain protein
casanova. Dev Cell 2004, 6:91-101.
50.

Holloway BA, Gomez de la Torre Canny S, Ye Y, Slusarski DC,
Freisinger CM, Dosch R, Chou MM, Wagner DS, Mullins MC: A
novel role for MAPKAPK2 in morphogenesis during zebrafish
development. PLoS Genet 2009, 5:e1000413.
This study identifies an unanticipated role for MAPKAPK2 and p38 MAP
kinase in modulating a marginal constrictive force acting in zebrafish
epiboly, providing novel molecular insight into this still poorly understood
tissue morphogenesis process.
51. Lukas SM, Kroe RR, Wildeson J, Peet GW, Frego L, Davidson W,
Ingraham RH, Pargellis CA, Labadia ME, Werneburg BG:
Catalysis and function of the p38 alpha, MK2a signaling
complex. Biochemistry 2004, 43:9950-9960.
52. Giraldez AJ, Cinalli RM, Glasner ME, Enright AJ, Thomson JM,
Baskerville S, Hammond SM, Bartel DP, Schier AF: MicroRNAs
regulate brain morphogenesis in zebrafish. Science 2005,
308:833-838.
53. Bushati N, Cohen SM: microRNA functions. Annu Rev Cell Dev
Biol 2007, 23:175-205.
54. Schier AF, Giraldez AJ: MicroRNA function and mechanism:
insights from zebra fish. Cold Spring Harb Symp Quant Biol
2006, 71:195-203.
55. Draper BW, McCallum CM, Moens CB: nanos1 is required to
maintain oocyte production in adult zebrafish. Dev Biol 2007,
305:589-598.
56. Mishima Y, Giraldez AJ, Takeda Y, Fujiwara T, Sakamoto H,
Schier AF, Inoue K: Differential regulation of germline mRNAs in
soma and germ cells by zebrafish miR-430. Curr Biol 2006,
16:2135-2142.
57.

Kedde M, Strasser MJ, Boldajipour B, Oude Vrielink JA,
Slanchev K, le Sage C, Nagel R, Voorhoeve PM, van Duijse J,
Orom UA et al.: RNA-binding protein Dnd1 inhibits microRNA
access to target mRNA. Cell 2007, 131:1273-1286.
This study demonstrates that the vertebrate-specific RNA binding protein
Dnd, essential for germ line development in zebrafish and mouse, but
previously of unknown molecular function, acts by blocking miRNA-
mediated gene silencing in zebrafish germ cells and in cultured human
cells. Dnd binds to a U-rich site in the 30
UTR of regulated transcripts that,
in a still unknown manner, blocks miRNA binding.
58. Slanchev K, Stebler J, Goudarzi M, Cojocaru V, Weidinger G,
Raz E: Control of dead end localization and activity–
implications for the function of the protein in antagonizing
miRNA function. Mech Dev 2009, 126:270-277.
59. Weidinger G, Stebler J, Slanchev K, Dumstrei K, Wise C, Lovell-
Badge R, Thisse C, Thisse B, Raz E: dead end, a novel vertebrate
germ plasm component, is required for zebrafish primordial
germ cell migration and survival. Curr Biol 2003, 13:1429-1434.
60. Kuramochi-Miyagawa S, Kimura T, Ijiri TW, Isobe T, Asada N,
Fujita Y, Ikawa M, Iwai N, Okabe M, Deng W et al.: Mili, a
mammalian member of piwi family gene, is essential for
spermatogenesis. Development 2004, 131:839-849.
61. Cox DN, Chao A, Lin H: piwi encodes a nucleoplasmic factor
whose activity modulates the number and division rate of
germline stem cells. Development 2000, 127:503-514.
62. Pal-Bhadra M, Bhadra U, Birchler JA: RNAi related mechanisms
affect both transcriptional and posttranscriptional transgene
silencing in Drosophila. Mol Cell 2002, 9:315-327.
63. Siegfried KR, Nusslein-Volhard C: Germ line control of female
sex determination in zebrafish. Dev Biol 2008, 324:277-287.
64. Slanchev K, Stebler J, de la Cueva-Mendez G, Raz E:
Development without germ cells: the role of the germ line in
zebrafish sex differentiation. Proc Natl Acad Sci U S A 2005,
102:4074-4079.
65. Aravin AA, Sachidanandam R, Girard A, Fejes-Toth K, Hannon GJ:
Developmentally regulated piRNA clusters implicate MILI in
transposon control. Science 2007, 316:744-747.
66. Carmell MA, Girard A, van de Kant HJ, Bourc’his D, Bestor TH, de
Rooij DG, Hannon GJ: MIWI2 is essential for spermatogenesis
and repression of transposons in the mouse male germline.
Dev Cell 2007, 12:503-514.
67. Ciruna B, Weidinger G, Knaut H, Thisse B, Thisse C, Raz E,
Schier AF: Production of maternal-zygotic mutant zebrafish by
germ-line replacement. Proc Natl Acad Sci U S A 2002,
99:14919-14924.
68. Theodosiou NA, Xu T: Use of FLP/FRT system to study
Drosophila development. Methods 1998, 14:355-365.
69. Khakhar RR, Cobb JA, Bjergbaek L, Hickson ID, Gasser SM: RecQ
helicases: multiple roles in genome maintenance. Trends Cell
Biol 2003, 13:493-501.
70. Xie J, Bessling SL, Cooper TK, Dietz HC, McCallion AS, Fisher S:
Manipulating mitotic recombination in the zebrafish embryo
through RecQ helicases. Genetics 2007, 176:1339-1342.
71. Nasevicius A, Ekker SC: Effective targeted gene ‘knockdown’ in
zebrafish. Nat Genet 2000, 26:216-220.
72. Draper BW, Morcos PA, Kimmel CB: Inhibition of zebrafish fgf8
pre-mRNA splicing with morpholino oligos: a quantifiable
method for gene knockdown. Genesis 2001, 30:154-156.
73. Doyon Y, McCammon JM, Miller JC, Faraji F, Ngo C, Katibah GE,
Amora R, Hocking TD, Zhang L, Rebar EJ et al.: Heritable
targeted gene disruption in zebrafish using designed zinc-
finger nucleases. Nat Biotechnol 2008, 26:702-708.
74. Meng X, Noyes MB, Zhu LJ, Lawson ND, Wolfe SA: Targeted
gene inactivation in zebrafish using engineered zinc-finger
nucleases. Nat Biotechnol 2008, 26:695-701.
75. Foley JE, Yeh JR, Maeder ML, Reyon D, Sander JD, Peterson RT,
Joung JK: Rapid mutation of endogenous zebrafish genes
using zinc finger nucleases made by Oligomerized Pool
ENgineering (OPEN). PLoS ONE 2009, 4:e4348.
Maternal Zebrafish Development Abrams and Mullins 403
www.sciencedirect.com Current Opinion in Genetics  Development 2009, 19:396–403

More Related Content

What's hot

Cell Fate by Progressive Determinants
Cell Fate by Progressive DeterminantsCell Fate by Progressive Determinants
Cell Fate by Progressive DeterminantsZoologist Pakistan
 
homeostasis and soma-germline interaction
homeostasis and soma-germline interactionhomeostasis and soma-germline interaction
homeostasis and soma-germline interactionBhavya Vashisht
 
Drosophila Melanogaster Genome And its developmental process
Drosophila Melanogaster  Genome And its developmental processDrosophila Melanogaster  Genome And its developmental process
Drosophila Melanogaster Genome And its developmental processSubhradeep sarkar
 
Primordial germ cells ps
Primordial germ cells psPrimordial germ cells ps
Primordial germ cells psParaniSankar
 
P S H S U P C A T Review Bio ( Part 1)
P S H S  U P C A T  Review    Bio ( Part 1)P S H S  U P C A T  Review    Bio ( Part 1)
P S H S U P C A T Review Bio ( Part 1)Kent Kawashima
 
Segmentation in Drosophila melanogaster
Segmentation in Drosophila melanogaster Segmentation in Drosophila melanogaster
Segmentation in Drosophila melanogaster Shreya Ahuja
 
Mesoderm induction see page 5
Mesoderm induction see page 5Mesoderm induction see page 5
Mesoderm induction see page 5Shoeb Ahmad
 
Morphogens, induction and cytoplasmic determinants
Morphogens, induction and cytoplasmic determinantsMorphogens, induction and cytoplasmic determinants
Morphogens, induction and cytoplasmic determinantsSakshi Saxena
 
Axis and pattern formation in amphibia
Axis and pattern formation in amphibiaAxis and pattern formation in amphibia
Axis and pattern formation in amphibiaShariqaJan
 
Vulva development - C.elegans
Vulva development - C.elegansVulva development - C.elegans
Vulva development - C.elegansnaren
 
Genetic control of animal development
Genetic control of animal developmentGenetic control of animal development
Genetic control of animal developmentHafiz M Waseem
 
Cleavage patterns, fate maps and cell lineages… What can early developmental ...
Cleavage patterns, fate maps and cell lineages… What can early developmental ...Cleavage patterns, fate maps and cell lineages… What can early developmental ...
Cleavage patterns, fate maps and cell lineages… What can early developmental ...ahejnol
 
Differential Cell Affinity & Cell Adhesion Molecules
Differential Cell Affinity & Cell Adhesion MoleculesDifferential Cell Affinity & Cell Adhesion Molecules
Differential Cell Affinity & Cell Adhesion MoleculesSyed Muhammad Khan
 
Morphogenesis and cell adhesion.
Morphogenesis and cell adhesion.Morphogenesis and cell adhesion.
Morphogenesis and cell adhesion.Bhupen Koch
 

What's hot (20)

Fabrizio 2008
Fabrizio 2008Fabrizio 2008
Fabrizio 2008
 
Cell Fate by Progressive Determinants
Cell Fate by Progressive DeterminantsCell Fate by Progressive Determinants
Cell Fate by Progressive Determinants
 
GenesDev_2008
GenesDev_2008GenesDev_2008
GenesDev_2008
 
homeostasis and soma-germline interaction
homeostasis and soma-germline interactionhomeostasis and soma-germline interaction
homeostasis and soma-germline interaction
 
Drosophila Melanogaster Genome And its developmental process
Drosophila Melanogaster  Genome And its developmental processDrosophila Melanogaster  Genome And its developmental process
Drosophila Melanogaster Genome And its developmental process
 
Primordial germ cells ps
Primordial germ cells psPrimordial germ cells ps
Primordial germ cells ps
 
P S H S U P C A T Review Bio ( Part 1)
P S H S  U P C A T  Review    Bio ( Part 1)P S H S  U P C A T  Review    Bio ( Part 1)
P S H S U P C A T Review Bio ( Part 1)
 
Segmentation in Drosophila melanogaster
Segmentation in Drosophila melanogaster Segmentation in Drosophila melanogaster
Segmentation in Drosophila melanogaster
 
Muzzopappa and Wappner Dev
Muzzopappa and Wappner DevMuzzopappa and Wappner Dev
Muzzopappa and Wappner Dev
 
Mesoderm induction see page 5
Mesoderm induction see page 5Mesoderm induction see page 5
Mesoderm induction see page 5
 
cell fates
cell fatescell fates
cell fates
 
Morphogens, induction and cytoplasmic determinants
Morphogens, induction and cytoplasmic determinantsMorphogens, induction and cytoplasmic determinants
Morphogens, induction and cytoplasmic determinants
 
Developmental genetics
Developmental genetics Developmental genetics
Developmental genetics
 
Axis and pattern formation in amphibia
Axis and pattern formation in amphibiaAxis and pattern formation in amphibia
Axis and pattern formation in amphibia
 
Vulva development - C.elegans
Vulva development - C.elegansVulva development - C.elegans
Vulva development - C.elegans
 
Genetic control of animal development
Genetic control of animal developmentGenetic control of animal development
Genetic control of animal development
 
PATTERN FORMATION
PATTERN FORMATIONPATTERN FORMATION
PATTERN FORMATION
 
Cleavage patterns, fate maps and cell lineages… What can early developmental ...
Cleavage patterns, fate maps and cell lineages… What can early developmental ...Cleavage patterns, fate maps and cell lineages… What can early developmental ...
Cleavage patterns, fate maps and cell lineages… What can early developmental ...
 
Differential Cell Affinity & Cell Adhesion Molecules
Differential Cell Affinity & Cell Adhesion MoleculesDifferential Cell Affinity & Cell Adhesion Molecules
Differential Cell Affinity & Cell Adhesion Molecules
 
Morphogenesis and cell adhesion.
Morphogenesis and cell adhesion.Morphogenesis and cell adhesion.
Morphogenesis and cell adhesion.
 

Similar to Early zebrafish development itis in

Developmental biology
Developmental biologyDevelopmental biology
Developmental biologyNova Corciega
 
SO MR1 Bioremediation
SO MR1 BioremediationSO MR1 Bioremediation
SO MR1 BioremediationMeg Vermilion
 
Practical manual of in vitro fertilization
Practical manual of in vitro fertilizationPractical manual of in vitro fertilization
Practical manual of in vitro fertilizationSpringer
 
Development in drosophila ppt
Development in drosophila pptDevelopment in drosophila ppt
Development in drosophila pptSujataRao11
 
Ptacin_et_al-2013-Cellular_Microbiology (review)
Ptacin_et_al-2013-Cellular_Microbiology (review)Ptacin_et_al-2013-Cellular_Microbiology (review)
Ptacin_et_al-2013-Cellular_Microbiology (review)Jerod Ptacin
 
How does the Drosophila embryo get its pair-rule stripes How is the .pdf
How does the Drosophila embryo get its pair-rule stripes How is the .pdfHow does the Drosophila embryo get its pair-rule stripes How is the .pdf
How does the Drosophila embryo get its pair-rule stripes How is the .pdfdeepakarora871
 
Ontogenetic Development of Drosophila
Ontogenetic Development of DrosophilaOntogenetic Development of Drosophila
Ontogenetic Development of DrosophilaSamchuchoo
 
14.2_ Frog Embryology - Biology LibreTexts.pdf
14.2_ Frog Embryology - Biology LibreTexts.pdf14.2_ Frog Embryology - Biology LibreTexts.pdf
14.2_ Frog Embryology - Biology LibreTexts.pdfGabrielChua12
 
B978-0-444-63956-1.00002-3.pdf
B978-0-444-63956-1.00002-3.pdfB978-0-444-63956-1.00002-3.pdf
B978-0-444-63956-1.00002-3.pdfsuresh163251
 
Early development of invertebrates
Early development of invertebratesEarly development of invertebrates
Early development of invertebratesMerlyn Denesia
 
Bio22 4th post lab discussion
Bio22 4th post lab discussionBio22 4th post lab discussion
Bio22 4th post lab discussionLani Manahan
 
Normal and abnormal oocyte morphology
Normal  and abnormal oocyte morphologyNormal  and abnormal oocyte morphology
Normal and abnormal oocyte morphologySumia abdulsalam
 
Gutell 086.bmc.evol.biol.2003.03.07
Gutell 086.bmc.evol.biol.2003.03.07Gutell 086.bmc.evol.biol.2003.03.07
Gutell 086.bmc.evol.biol.2003.03.07Robin Gutell
 

Similar to Early zebrafish development itis in (20)

Chap20
Chap20Chap20
Chap20
 
Trends Molecular Medicine 2015
Trends Molecular Medicine 2015Trends Molecular Medicine 2015
Trends Molecular Medicine 2015
 
Developmental biology
Developmental biologyDevelopmental biology
Developmental biology
 
Cell fate (fate maps)
Cell fate (fate maps)Cell fate (fate maps)
Cell fate (fate maps)
 
SO MR1 Bioremediation
SO MR1 BioremediationSO MR1 Bioremediation
SO MR1 Bioremediation
 
Practical manual of in vitro fertilization
Practical manual of in vitro fertilizationPractical manual of in vitro fertilization
Practical manual of in vitro fertilization
 
Development in drosophila ppt
Development in drosophila pptDevelopment in drosophila ppt
Development in drosophila ppt
 
Ptacin_et_al-2013-Cellular_Microbiology (review)
Ptacin_et_al-2013-Cellular_Microbiology (review)Ptacin_et_al-2013-Cellular_Microbiology (review)
Ptacin_et_al-2013-Cellular_Microbiology (review)
 
How does the Drosophila embryo get its pair-rule stripes How is the .pdf
How does the Drosophila embryo get its pair-rule stripes How is the .pdfHow does the Drosophila embryo get its pair-rule stripes How is the .pdf
How does the Drosophila embryo get its pair-rule stripes How is the .pdf
 
Ontogenetic Development of Drosophila
Ontogenetic Development of DrosophilaOntogenetic Development of Drosophila
Ontogenetic Development of Drosophila
 
14.2_ Frog Embryology - Biology LibreTexts.pdf
14.2_ Frog Embryology - Biology LibreTexts.pdf14.2_ Frog Embryology - Biology LibreTexts.pdf
14.2_ Frog Embryology - Biology LibreTexts.pdf
 
SICBposter_final2
SICBposter_final2SICBposter_final2
SICBposter_final2
 
B978-0-444-63956-1.00002-3.pdf
B978-0-444-63956-1.00002-3.pdfB978-0-444-63956-1.00002-3.pdf
B978-0-444-63956-1.00002-3.pdf
 
Early development of invertebrates
Early development of invertebratesEarly development of invertebrates
Early development of invertebrates
 
Brienoenriquez 2015
Brienoenriquez 2015Brienoenriquez 2015
Brienoenriquez 2015
 
Bio22 4th post lab discussion
Bio22 4th post lab discussionBio22 4th post lab discussion
Bio22 4th post lab discussion
 
Normal and abnormal oocyte morphology
Normal  and abnormal oocyte morphologyNormal  and abnormal oocyte morphology
Normal and abnormal oocyte morphology
 
Holubcova et al
Holubcova et alHolubcova et al
Holubcova et al
 
research paper
research paperresearch paper
research paper
 
Gutell 086.bmc.evol.biol.2003.03.07
Gutell 086.bmc.evol.biol.2003.03.07Gutell 086.bmc.evol.biol.2003.03.07
Gutell 086.bmc.evol.biol.2003.03.07
 

More from wujunbo1015

More from wujunbo1015 (8)

Cell reprogram
Cell reprogramCell reprogram
Cell reprogram
 
Cell review
Cell  reviewCell  review
Cell review
 
Mouse zar1 like (xm 359149) colocalizes with m-rna processing components and ...
Mouse zar1 like (xm 359149) colocalizes with m-rna processing components and ...Mouse zar1 like (xm 359149) colocalizes with m-rna processing components and ...
Mouse zar1 like (xm 359149) colocalizes with m-rna processing components and ...
 
7
77
7
 
6
66
6
 
5
55
5
 
4
44
4
 
3
33
3
 

Early zebrafish development itis in

  • 1. Available online at www.sciencedirect.com Early zebrafish development: It’s in the maternal genes Elliott W Abrams and Mary C Mullins The earliest stages of embryonic development in all animals examined rely on maternal gene products that are generated during oogenesis and supplied to the egg. The period of maternal control of embryonic development varies among animals according to the onset of zygotic transcription and the persistence of maternal gene products. This maternal regulation has been little studied in vertebrates, owing to the difficulty in manipulating maternal gene function and lack of basic molecular information. However, recent maternal-effect screens in the zebrafish have generated more than 40 unique mutants that are providing new molecular entry points to the maternal control of early vertebrate development. Here we discuss recent studies of 12 zebrafish mutant genes that illuminate the maternal molecular controls on embryonic development, including advances in the regulation of animal– vegetal polarity, egg activation, cleavage development, body plan formation, tissue morphogenesis, microRNA function and germ cell development. Address University of Pennsylvania School of Medicine, Department of Cell and Developmental Biology, 1211 BRB II/III, 421 Curie Blvd, Philadelphia, PA 19104-6058, United States Corresponding author: Mullins, Mary C (mullins@mail.med.upenn.edu) Current Opinion in Genetics & Development 2009, 19:396–403 This review comes from a themed issue on Pattern formation and developmental mechanisms Edited by Kathryn Anderson and Kenneth Irvine Available online 14th July 2009 0959-437X/$ – see front matter # 2009 Elsevier Ltd. All rights reserved. DOI 10.1016/j.gde.2009.06.002 Introduction The zebrafish has emerged as a premiere genetic tool for studying vertebrate development. In the 1990s forward genetic screens identified numerous zygotic mutants defective in key molecules important in early embryonic development [1–7]. Recently, a major focus has shifted towards more specialized screens, including the identi- fication of maternal-effect mutations in adult screens in the zebrafish. Since the earliest stages of development are driven primarily by maternal gene products, the identification of corresponding mutants is crucial to provide genetic entry points to known maternally con- trolled processes, which are still poorly understood in vertebrates. Maternal-effect screens have identified over 40 mutants affecting many early developmental processes (Figure 1). Collectively, these mutants have defects in oocyte de- velopment [8], egg activation [8,9], embryonic cleavage [8–11], patterning and morphogenesis [9–11]. New stu- dies have focused on the molecular identification of the corresponding mutant genes, a key step to understand- ing the molecular mechanisms governing the very earliest stages of embryogenesis. The results have unveiled novel and known genes with unanticipated roles in early development. Mutants generated by reverse genetic TILLING methods in the zebrafish have also recently revealed important roles for small noncoding RNA molecules, miRNAs [12] and piRNAs [13,14], in the regulation of maternal processes in zebra- fish development. In this review, we highlight recent contributions to the molecular regulation of animal–vegetal polarity in the oocyte and egg, maternal gene regulation of early embryo- nic patterning, tissue morphogenesis and small noncoding RNA molecules, which are emerging as important players in germ line development. Molecular insights into zebrafish animal– vegetal polarity In frogs and fish, the first developmental asymmetry of the embryo is the animal–vegetal axis, which predicts the anterior–posterior axis of the embryo. This asymmetry is established during early stages of oogenesis and is first marked by formation of the Balbiani body (Bb; also referred to as the mitochondrial cloud) adjacent to the germinal vesicle (the oocyte nucleus) in stage I oocytes. The Bb position predicts the location of the vegetal pole, but its presence is only transient, as it disassembles by stage II of oogenesis. The Bb is composed of a collection of mitochondria, ER, germinal granules and several germ plasm mRNAs (reviewed in [15]). Recently, a thorough study recapitulated in zebrafish transgenic constructs the localization pattern of three Bb-localized transcripts. The 30 UTRs of nanos, vasa and dazl directed their localization initially to the Bb and then to the vegetal cortex of the oocyte [16 ], probably via the METRO pathway described in Xenopus [17]. The ensuing distribution at the vegetal cortex differs among these transcripts: dazl persists at the vegetal cortex, vasa extends around the cortex, and nanos becomes unlocalized (Figure 2), suggesting, unexpectedly, that coordinate localization of germ plasm components is followed by their redistri- bution to distinct locations. However, after fertilization of the egg, these germ plasm RNAs reunite in the blastodisc at the animal pole, where they localize to the germ plasm Current Opinion in Genetics Development 2009, 19:396–403 www.sciencedirect.com
  • 2. Maternal Zebrafish Development Abrams and Mullins 397 Figure 1 Maternal-effect mutant genes in zebrafish disrupt development at distinct stages. Mutants discussed here are indicated in red. acytokinesis [11], atomos [9], aura [9], barrette [9], bo peep [9], bedazzled [10], blistered [10], claustro [9], cobblestone [9], emulsion [8], golden gate [9], indivisible [8], irreducible [8], jumpstart [8], kwai [9], misson impossible [9], nebel [9], over easy [8], poky [10], pollywog [10], pug [10], slow [10], ruehrei [8], souffle [8], screeching halt [10], sunny side up [8], under repair [9], waldo [9], weeble [9]. Figure 2 mRNA localization during oocyte development. During stage I of oogenesis buc, nanos, vasa and dazl transcripts localize to the Balibani body (Bb, pink), while cyclinB begins to be localized to the animal pole. By stage II of oogenesis the Bb has disassembled, leaving buc, vasa and dazl mRNAs at the vegetal cortex, whereas nanos becomes unlocalized, and pou2 becomes localized to the animal pole. Note vasa has a broad vegetal cortical domain at stage II. By stage III bruno-like and mago nashi become vegetally localized (late pathway); now buc, and Vg1 are localized to the animal pole and vasa is localized radially at the cortex. Animal (An) pole is to top and vegetal (Ve) to bottom. www.sciencedirect.com Current Opinion in Genetics Development 2009, 19:396–403
  • 3. at the cleavage furrows of the four-cell stage embryo [16 ,18–21]. Three regions within the dazl 30 UTR are crucial for (1) localization to the Bb, (2) anchoring to the vegetal cortex, and (3) localization to the cleavage furrows in the early embryo [16 ]. Interestingly, although the 30 UTR of the nanos-related gene Xcat2 in Xenopus, also localizes it to the Bb and vegetal cortex in zebrafish, little sequence similarity is evident in the 30 UTRs of Xcat2, and zebrafish nanos, dazl and vasa [16 ], suggesting either small motifs, or secondary/tertiary structure of the UTRs are con- served. Thus, germ plasm mRNA localization occurs in a stepwise, spatiotemporal fashion, with conserved fea- tures to the transport systems in Xenopus [16 ]. Recent studies have provided genetic access into the mechanisms of Bb formation and germ plasm assembly in the oocyte. The maternal-effect mutant bucky ball (buc), identified through its animal–vegetal polarity egg pheno- type [8], is defective in Bb formation and early oocyte polarity, as animal pole markers are expanded radially and vegetal pole markers are unlocalized [22 ,23 ]. Buc is required for early vegetal pole mRNA localization that occurs via the Bb, as well as late vegetal pole localization that occurs after Bb dissociation, which is postulated to depend on the early pathway [24,25]. Thus, the failure of late vegetal pole mRNA localization in buc mutant oocytes may be secondary to the lack of Bb formation. Buc functions to promote vegetal and exclude animal pole identity also in the surrounding follicle cell layer, preventing the formation of multiple micropylar cells, an animal pole-specific follicle cell fate. Thus, patterning of the oocyte and surrounding somatic follicle cell layer appears to be coordinated through an as yet unknown signaling mechanism [22 ]. Recently the molecular identity of buc was determined to be a novel 639 amino acid protein [23 ], first identified as XVelo1 in Xenopus [26]. The predicted Buc protein contains no known functional motifs. The localization of buc mRNA is dynamic during oocyte development, initially localizing to the Bb, moving vegetally and then ultimately localizing to the animal pole during late oogenesis [23 ] (Figure 2). A Buc-GFP fusion also loca- lizes to the Bb and, interestingly, to the germ plasm of the embryo and can induce ectopic germ cells in the embryo [23 ]. Buc homologues in other vertebrates have low predicted amino acid conservation, indicating that it has evolved at a relatively accelerated rate. Interestingly, the human version lacks a complete open reading frame, suggesting that it has lost its function all together in humans or functions as an RNA. In zebrafish Buc clearly functions as a protein, since a nonsense mutation trun- cating it by just 30 amino acids causes a failure in Bb formation [23 ]. However, the buc RNA could have additional functions in Bb formation and animal–vegetal polarity, similar to oskar in Drosophila [27], also a germ plasm component. Future structure-function exper- iments of the buc RNA/protein, together with the iso- lation of interacting partners will unravel mechanistically how this novel gene functions. Egg activation molecular genetics In zebrafish egg activation is marked by cortical granule exocytosis (CGE), chorion elevation and the segregation of cytoplasm from the yolk to the animal pole to form the single cell blastodisc. Embryos derived from brom bones (brb) mutant females are defective in this process [28]. The egg activation defect is a result of failure of inositol 1,4,5-triphosphate (IP3) signaling, which induces a Ca2+ wave crucial for normal egg activation in all animals examined. The reinstatement of either IP3 or Ca2+ in brb mutant eggs can rescue the egg activation defect [28]. brb was shown to encode heterogeneous nuclear ribonu- cleoprotein I (hnRNP I), probably regulating during oogenesis the production of an egg activation signaling component. hnRNP I has been previously shown to be important in a variety of developmental processes in- cluding translational control in oogenesis [29], spermato- genesis [30] and RNA localization [31–34]. Thus, brb mutants reveal yet another developmental function for hnRNP I, that is, a role in egg activation. Cleavage stage molecular genetics Maternal-effect screens have yielded a significant num- ber of mutations affecting cleavage development [8–11], although most of the mutant genes have yet to be cloned. cellular atoll (cea) mutants fail to undergo cleavages at the second cell division and beyond [8,35], and was recently shown to encode the centriolar component sas-6 [35]. Interestingly, as sperm normally provide the centriole to the zygote, cea also has a paternal-effect function, whereby wild-type eggs fertilized with cea mutant sperm result in inviable tetraploid embryos. Since the cleavage stage of development is primarily under maternal control, the eventual molecular identification of other maternal- effect mutant genes functioning during this period will enhance our understanding of this important stage of development. The maternal-effect cellular island (cei) mutant displays an early defect in cleavage furrow formation [8]. cei encodes a hypomorphic allele of Aurora B Kinase [36], a protein previously shown to be important in several aspects of cell division in other systems (reviewed in [37]). Aurora B Kinase function is vital zygotically later in zebrafish embryogenesis, as a null retroviral insertional allele reveals furrow defects during this period [36,38]. Inter- estingly, the maternal-effect cei allele causes a specific defect in distal furrow formation during the early cleavage stage, while medially positioned furrows can form, prob- ably owing to intact mitotic spindle-derived signals med- iating medial furrow formation [36]. The ability of the cei 398 Pattern formation and developmental mechanisms Current Opinion in Genetics Development 2009, 19:396–403 www.sciencedirect.com
  • 4. mutant Aurora B Kinase protein to mediate all its cyto- kinesis functions zygotically, but its inability to mediate distal furrow formation in the cleavage embryo may be related to unique requirements for astral microtubules and Aurora B Kinase to divide the large cells of the very early embryo [36]. Molecular genetic advances in maternal regulated patterning and morphogenesis In fish and amphibians, the dorsoventral embryonic axis is established through a maternally regulated Wnt/bcatenin pathway. Several zebrafish maternal-effect mutants with defects in dorsoventral axis formation have been ident- ified. The maternal-effect ventralized mutants hecate and tokkaebe produce embryos lacking a dorsal organizer and consequently are radially ventralized. Although hecate and tokkaebe have not yet been molecular defined, injection/ rescue experiments with Wnt signaling pathway com- ponents indicates that they act upstream of, or within a Wnt/bcatenin signaling pathway, respectively, to induce the dorsal organizer [39–41]. tokkaebe probably corre- sponds to a novel component of the Wnt pathway, since no known Wnt components are found within the 0.5 cM interval containing the tokkaebe mutation [41]. The maternal-effect ventralized mutant ichabod is caused by a specific loss of maternal bcatenin2 function [42]. Two bcatenin genes have been identified in the zebrafish genome; however, only bcatenin2 is essential maternally to induce the dorsal organizer through a Wnt signaling pathway. Interestingly, the maternal, dorsal organizer Wnt/bcatenin2 pathway also functions to repress expres- sion of a later Wnt8/bcatenin pathway that opposes dorsal specification ventrally [42] (Figure 3). In zebrafish this late blastula Wnt pathway depends on both bcatenin1 and bcatenin2 function and is mediated by zygotic Wnt8 signaling ventrally [42–45]. Thus in the absence of both bcatenin1 and bcatenin2 function, early dorsal organizer formation fails; however, at later stages the ventral Wnt8 pathway also fails to oppose dorsal fate specification ventrally. The result is the formation of dorsal tissues circumferentially. These results show, surprisingly, that at a late blastula stage circumferential organizer-like tissue can form in the absence of a maternal Wnt/bcatenin pathway and independently of Wnt/bcatenin signaling entirely. The transcription factor pou2/oct4 also acts maternally in dorsoventral patterning and morphogenesis [46,47], revealing additional roles to its described maternal func- tion in endoderm specification [48,49]. Loss of both maternal and zygotic (MZ) Pou2 causes severe dorsaliza- tion owing to failure to induce bmp ligand gene expres- sion, which functions in ventrolateral tissue specification [46]. These results demonstrate that ventral specification via the BMP signaling pathway is not a default pathway, as previously thought, but instead is initiated by maternal Pou2/Oct4 in the early embryo [46] (Figure 3). MZPou2 Maternal Zebrafish Development Abrams and Mullins 399 Figure 3 The role of BMP and early and late Wnt signaling in dorsoventral patterning. (a) Maternal Pou2 induces early zygotic bmp ligand expression. bmp expression is initially present throughout the blastoderm (grey). An early maternal Wnt signal from the yolk cell promotes future dorsal organizer (DO) formation. (b) By the late blastula stage (50% epiboly), a BMP activity gradient (highest ventrally (black)) is established that promotes ventral fates. At this stage Wnt 8 signaling opposes dorsal fate specification ventrally. Ventral is positioned to the left and dorsal to the right. www.sciencedirect.com Current Opinion in Genetics Development 2009, 19:396–403
  • 5. also regulates the morphogenic process of epiboly, the thinning and spreading of the blastoderm over the yolk cell, through functions in yolk cell microtubule formation, cell adhesive properties and blastoderm cell movements via a cell non-autonomous mechanism [46,47]. Together, these studies reveal that the renowned pou2/oct4 stem cell gene in mammals is a key maternal regulator of early zebrafish development. The betty boop (bbp) mutant, identified in a maternal- effect screen, is a strictly, maternally acting gene reg- ulating the morphogenic process of epiboly [10,50 ]. Embryos from bbp mothers develop normally until they reach 50% epiboly at which point the embryo abruptly bursts via a presumptive premature constriction of the actin cytoskeleton in the yolk cell [50 ]. Interestingly, bbp was recently shown to encode the zebrafish homol- ogue of Mitogen Activated Protein Kinase Activated Protein Kinase 2 (MAPKAPK2), a target of p38 MAP kinase (MAPK) in cell culture systems [51]. During zebrafish epiboly, p38 MAPK also appears to activate MAPKAPK2, as a dominant-negative p38 MAPK causes the same epiboly defect as loss of bbp [50 ]. Neither p38 MAPK, nor MAPKAPK2 have been previously impli- cated in tissue morphogenesis. Thus, the identification and cloning of the bbp gene is a model genetic case for an unexpected pathway being placed in a developmental process, in this case epiboly, which may not have been considered in a candidate-gene reverse genetic approach. Small RNA molecules in early development Small noncoding RNAs are emerging as important players in early zebrafish development. Maternal-zygo- tic (MZ) mutant embryos of the microRNA (miRNA)- processing enzyme Dicer exhibit early embryonic defects in gastrulation, somitogenesis, brain morpho- genesis and heart development [52]. By removing the strong maternal component of dicer, MZ-dicer mutant embryos are completely devoid of miRNA processing and therefore, devoid of all miRNA function. miRNAs negatively regulate target genes by binding to their 30 UTR, promoting deadenylation, translational repres- sion and/or ultimately degradation of the transcript (reviewed in [53]). In zebrafish, the predominantly expressed miRNA during early embryogenesis is miR- 430, which is first expressed at the mid-blastula tran- sition (MBT), and is not expressed maternally [54]. A central finding revealed by the loss of miR-430 through MZ-dicer is its role in the clearance of maternal mRNAs at the MBT. In MZ-dicer mutant embryos, maternal mRNAs abnormally persist beyond the MBT [12]. To date, no maternal miRNAs have been reported in the zebrafish embryo [54]. Thus, it is unclear whether the maternal role of dicer is to primarily process miR-430 at the MBT or in addition, also process unidentified maternal miRNA(s). Inhibition of miRNA function plays a role in germ line development. nanos1 is expressed early in germ line development and a mutant, generated through TIL- LING methods, demonstrates its maternal requirement for PGC survival and a function in the adult in maintain- ing oocyte production [55]. nanos1 is resistant to miRNA repression in the germ line, but not in the soma, promot- ing its specific expression in the germ line [56]. This resistance is conferred, at least partly, by the maternally expressed germ cell-localized RNA binding protein Dead End (Dnd), which interacts with the nanos1 30 UTR, presumably blocking the binding of miR-430 [57 ]. Inter- estingly, this resistance to miRNA repression is also found in another germ plasm mRNA, Tudor-domain-contain- ing-7 (Tdrd-7) [56], suggesting a general mechanism involving Dnd in influencing germ cell-specific gene expression (Figure 4). Germ plasm mRNA resistance to miRNA regulation is not universal, however, as the dnd 30 UTR itself lacks a miR-430 site and therefore is presumably not repressed in the soma by miRNAs [58]. Since dnd maternal transcripts are dramatically eliminated in the soma about one hour after the MBT [59], independently of miR-430 regula- tion, it suggests the existence of additional mode(s) of modulating maternal transcripts at this crucial develop- 400 Pattern formation and developmental mechanisms Figure 4 miRNA regulation of germ line development. In the soma miR-430 inhibits expression of germ line specific genes (GLSGs). In the germ line Dnd blocks miR-430-mediated repression of GLSGs. Note that an unidentified factor X is required in the soma to silence GLSGs that are not regulated by miR-430. Soma shown in grey; germ line in purple. Current Opinion in Genetics Development 2009, 19:396–403 www.sciencedirect.com
  • 6. mental transition. Likewise, vasa removal from the soma is also independent of miR-430 regulation [56]. Thus, an unknown, possibly common additional mechanism elim- inates dnd and vasa transcripts from the soma. A Dicer-independent class of small noncoding RNAs, known as piRNAs or Piwi associated RNAs, appear to be germ line specific [13,14,60,61]. Piwi proteins are important for target gene silencing [62]. Presumptive null, zygotic mutations in either of two piwi homologues in zebrafish, ziwi and zili, cause the progressive loss of germ cells between three and seven weeks of age [13,14]. Mutant adults are phenotypically male, consistent with recent studies demonstrating that zebrafish develop phe- notypically as male when eliminating the germ line [13,63,64]. Interestingly, there is a maternal-effect meio- tic progression defect in zili hypomorphic mutants. Although mutant eggs can be fertilized, they fail to undergo meiosis I and II [13]. Piwi proteins in the mouse function to repress transposon activity in the germ line [65,66]. The meiotic defects in zili hypomorphs mutants, however, are not linked to increased transposon activity as measured by quantitative RT-PCR and in situ hybridiz- ation [13]. These findings reveal a novel function for piRNAs, whose function in vertebrates was previously thought confined to regulating transposon activity in the germ line [13]. Future outlook A major hindrance to the molecular cloning of chemically induced mutant genes in zebrafish is the incomplete assembly of the genomic sequence. This obstacle is becoming less of an issue with recent improvements to the assembly (currently Zv8; www.sanger.ac.uk). As exist- ing gaps are eliminated and the genome sequence is completed, the molecular cloning of maternal-effect mutant genes will be greatly accelerated. The investigation of the maternal functions of essential zygotic genes will be more difficult to study. Although germ line chimeric analysis is a successful method to examine the maternal function of zygotic lethal genes in zebrafish [67], it is quite labour intensive and would be impractical for large throughput analysis of such maternal function. This problem may be overcome by employing techniques that utilize mitotic recombination, a principle heavily relied upon in Drosophila genetics [68]. RecQ helicases are known to prevent recombination during replication [69]. Induced mitotic recombination through the suppression of RecQ helicases was recently demon- strated in zebrafish [70]. This approach can generate mutant clones from heterozygous cells at a significant frequency (1.7–3.4%) [70]. With further improvements in this technology, mitotic recombination could poten- tially be used to generate homozygous mutant germ line clones of zygotic lethal mutations from heterozygous individuals. Such germ line mosaic females would gen- erate maternally deficient eggs and embryos, allowing the study of the maternal gene function. Reverse genetic techniques are very valuable comp- lements to forward genetic approaches in the zebrafish. Antisense morpholino oligos are widely used to block translation or disrupt splicing [71,72]. However, the func- tion of maternal protein already present in the egg cannot be blocked by morpholino injection into the egg, although advances in oocyte cell culture methods may make it possible to use this method in the future to examine maternal gene functions. Reverse genetic approaches that rely on induction of genomic sequence alterations are considerably more laborious, but generate robust loss-of-function reagents. Recently, the TILLING (Target Induced Local Lesions in Genomes) approach [13,14,55], as well as the zinc finger nuclease approach designed to mutate a specific sequence of the genome [73–75], have been very successful in generating mutants in zebrafish. Hence, TILLING and zinc finger nuclease strategies look very promising for eliminating gene func- tion of suspected maternal-effect genes in zebrafish in the future. Acknowledgements We thank Lee Kapp for comments on the manuscript, and Eric Weinberg and Mate Varga for helpful discussion. Funding was provided by NIH grant HD050901 to MCM. References and recommended reading Papers of particular interest, published within the period of review, have been highlighted as: of special interest of outstanding interest 1. Driever W, Solnica-Krezel L, Schier AF, Neuhauss SC, Malicki J, Stemple DL, Stainier DY, Zwartkruis F, Abdelilah S, Rangini Z et al.: A genetic screen for mutations affecting embryogenesis in zebrafish. Development 1996, 123:37-46. 2. Hammerschmidt M, Pelegri F, Mullins MC, Kane DA, Brand M, van Eeden FJ, Furutani-Seiki M, Granato M, Haffter P, Heisenberg CP et al.: Mutations affecting morphogenesis during gastrulation and tail formation in the zebrafish, Danio rerio. Development 1996, 123:143-151. 3. Kane DA, Hammerschmidt M, Mullins MC, Maischein HM, Brand M, van Eeden FJ, Furutani-Seiki M, Granato M, Haffter P, Heisenberg CP et al.: The zebrafish epiboly mutants. Development 1996, 123:47-55. 4. Kane DA, Maischein HM, Brand M, van Eeden FJ, Furutani- Seiki M, Granato M, Haffter P, Hammerschmidt M, Heisenberg CP, Jiang YJ et al.: The zebrafish early arrest mutants. Development 1996, 123:57-66. 5. Mullins MC, Hammerschmidt M, Kane DA, Odenthal J, Brand M, van Eeden FJ, Furutani-Seiki M, Granato M, Haffter P, Heisenberg CP et al.: Genes establishing dorsoventral pattern formation in the zebrafish embryo: the ventral specifying genes. Development 1996, 123:81-93. 6. Solnica-Krezel L, Stemple DL, Mountcastle-Shah E, Rangini Z, Neuhauss SC, Malicki J, Schier AF, Stainier DY, Zwartkruis F, Abdelilah S et al.: Mutations affecting cell fates and cellular rearrangements during gastrulation in zebrafish. Development 1996, 123:67-80. 7. Stemple DL, Solnica-Krezel L, Zwartkruis F, Neuhauss SC, Schier AF, Malicki J, Stainier DY, Abdelilah S, Rangini Z, Maternal Zebrafish Development Abrams and Mullins 401 www.sciencedirect.com Current Opinion in Genetics Development 2009, 19:396–403
  • 7. Mountcastle-Shah E et al.: Mutations affecting development of the notochord in zebrafish. Development 1996, 123:117-128. 8. Dosch R, Wagner DS, Mintzer KA, Runke G, Wiemelt AP, Mullins MC: Maternal control of vertebrate development before the midblastula transition: mutants from the zebrafish I. Dev Cell 2004, 6:771-780. 9. Pelegri F, Dekens MP, Schulte-Merker S, Maischein HM, Weiler C, Nusslein-Volhard C: Identification of recessive maternal-effect mutations in the zebrafish using a gynogenesis-based method. Dev Dyn 2004, 231:324-335. 10. Wagner DS, Dosch R, Mintzer KA, Wiemelt AP, Mullins MC: Maternal control of development at the midblastula transition and beyond: mutants from the zebrafish II. Dev Cell 2004, 6:781-790. 11. Kishimoto Y, Koshida S, Furutani-Seiki M, Kondoh H: Zebrafish maternal-effect mutations causing cytokinesis defect without affecting mitosis or equatorial vasa deposition. Mech Dev 2004, 121:79-89. 12. Giraldez AJ, Mishima Y, Rihel J, Grocock RJ, Van Dongen S, Inoue K, Enright AJ, Schier AF: Zebrafish MiR-430 promotes deadenylation and clearance of maternal mRNAs. Science 2006, 312:75-79. 13. Houwing S, Berezikov E, Ketting RF: Zili is required for germ cell differentiation and meiosis in zebrafish. EMBO J 2008, 27:2702- 2711. 14. Houwing S, Kamminga LM, Berezikov E, Cronembold D, Girard A, van den Elst H, Filippov DV, Blaser H, Raz E, Moens CB et al.: A role for Piwi and piRNAs in germ cell maintenance and transposon silencing in Zebrafish. Cell 2007, 129:69-82. 15. Kloc M, Bilinski S, Etkin LD: The Balbiani body and germ cell determinants: 150 years later. Curr Top Dev Biol 2004, 59:1-36. 16. Kosaka K, Kawakami K, Sakamoto H, Inoue K: Spatiotemporal localization of germ plasm RNAs during zebrafish oogenesis. Mech Dev 2007, 124:279-289. A very thorough analysis of the 30 UTR of dazl involving a series of transgenic animals demonstrates that distinct regions are required for successive steps in dazl localization during early oogenesis and in germ plasm localization in the embryo, implicating a more general utility of the early Xenopus METRO oogenesis localization pathway in zebrafish. 17. Kloc M, Etkin LD: Two distinct pathways for the localization of RNAs at the vegetal cortex in Xenopus oocytes. Development 1995, 121:287-297. 18. Theusch EV, Brown KJ, Pelegri F: Separate pathways of RNA recruitment lead to the compartmentalization of the zebrafish germ plasm. Dev Biol 2006, 292:129-141. 19. Knaut H, Pelegri F, Bohmann K, Schwarz H, Nusslein-Volhard C: Zebrafish vasa RNA but not its protein is a component of the germ plasm and segregates asymmetrically before germline specification. J Cell Biol 2000, 149:875-888. 20. Koprunner M, Thisse C, Thisse B, Raz E: A zebrafish nanos- related gene is essential for the development of primordial germ cells. Genes Dev 2001, 15:2877-2885. 21. Yoon C, Kawakami K, Hopkins N: Zebrafish vasa homologue RNA is localized to the cleavage planes of 2- and 4-cell-stage embryos and is expressed in the primordial germ cells. Development 1997, 124:3157-3165. 22. Marlow FL, Mullins MC: Bucky ball functions in Balbiani body assembly and animal-vegetal polarity in the oocyte and follicle cell layer in zebrafish. Dev Biol 2008, 321:40-50. This is a study of the first mutant in vertebrates known to be defective in Balbiani formation, the earliest oocyte asymmetric structure in verte- brates, which is conserved from insects to mammals and first described over 150 years ago. The analysis implicates this gene in setting up the animal–vegetal axis of early oocytes and shows that patterning of the oocyte and surrounding follicle cell layer is coordinated, probably through a signaling mechanism. 23. Bontems F, Stein A, Marlow F, Lyautey J, Gupta T, Mullins MC, Dosch R: Bucky ball organizes germ plasm assembly in zebrafish. Curr Biol 2009, 19:414-422. This study identifies the molecular nature of the bucky ball gene as a novel, rapidly evolving gene and demonstrates that its overexpression can result in ectopic germ cells, implicating it in germ plasm assembly. 24. Kloc M, Larabell C, Etkin LD: Elaboration of the messenger transport organizer pathway for localization of RNA to the vegetal cortex of Xenopus oocytes. Dev Biol 1996, 180:119-130. 25. King ML, Messitt TJ, Mowry KL: Putting RNAs in the right place at the right time: RNA localization in the frog oocyte. Biol Cell 2005, 97:19-33. 26. Claussen M, Pieler T: Xvelo1 uses a novel 75-nucleotide signal sequence that drives vegetal localization along the late pathway in Xenopus oocytes. Dev Biol 2004, 266:270-284. 27. Jenny A, Hachet O, Zavorszky P, Cyrklaff A, Weston MD, Johnston DS, Erdelyi M, Ephrussi A: A translation-independent role of oskar RNA in early Drosophila oogenesis. Development 2006, 133:2827-2833. 28. Mei W, Lee KW, Marlow F, Miller AL, Mullins MC: hnRNP I/brom bones is required to generate the Ca2+ signal that causes egg activation in the zebrafish. Development, in press. 29. Besse F, Lopez de Quinto S, Marchand V, Trucco A, Ephrussi A: Drosophila PTB promotes formation of high-order RNP particles and represses oskar translation. Genes Dev 2009, 23:195-207. 30. Robida MD, Singh R: Drosophila polypyrimidine-tract binding protein (PTB) functions specifically in the male germline. EMBO J 2003, 22:2924-2933. 31. Cote CA, Gautreau D, Denegre JM, Kress TL, Terry NA, Mowry KL: A Xenopus protein related to hnRNP I has a role in cytoplasmic RNA localization. Mol Cell 1999, 4:431-437. 32. Kress TL, Yoon YJ, Mowry KL: Nuclear RNP complex assembly initiates cytoplasmic RNA localization. J Cell Biol 2004, 165:203-211. 33. Czaplinski K, Mattaj IW: 40LoVe interacts with Vg1RBP/Vera and hnRNP I in binding the Vg1-localization element. RNA 2006, 12:213-222. 34. Lewis RA, Gagnon JA, Mowry KL: PTB/hnRNP I is required for RNP remodeling during RNA localization in Xenopus oocytes. Mol Cell Biol 2008, 28:678-686. 35. Yabe T, Ge X, Pelegri F: The zebrafish maternal-effect gene cellular atoll encodes the centriolar component sas-6 and defects in its paternal function promote whole genome duplication. Dev Biol 2007, 312:44-60. 36. Yabe T, Ge X, Lin R, Nair S, Runke G, Mullins MC, Pelegri F: The maternal-effect gene cellular island encodes aurora B kinase and is essential for furrow formation in the early zebrafish embryo. PLoS Genet 2009, 5:e1000518. 37. Vader G, Medema RH, Lens SM: The chromosomal passenger complex: guiding Aurora-B through mitosis. J Cell Biol 2006, 173:833-837. 38. Golling G, Amsterdam A, Sun Z, Antonelli M, Maldonado E, Chen W, Burgess S, Haldi M, Artzt K, Farrington S et al.: Insertional mutagenesis in zebrafish rapidly identifies genes essential for early vertebrate development. Nat Genet 2002, 31:135-140. 39. Lyman Gingerich J, Westfall TA, Slusarski DC, Pelegri F: hecate, a zebrafish maternal effect gene, affects dorsal organizer induction and intracellular calcium transient frequency. Dev Biol 2005, 286:427-439. 40. Lyman Gingerich J, Lindeman R, Putiri E, Stolzmann K, Pelegri F: Analysis of axis induction mutant embryos reveals morphogenetic events associated with zebrafish yolk extension formation. Dev Dyn 2006, 235:2749-2760. 41. Nojima H, Shimizu T, Kim CH, Yabe T, Bae YK, Muraoka O, Hirata T, Chitnis A, Hirano T, Hibi M: Genetic evidence for involvement of maternally derived Wnt canonical signaling in dorsal determination in zebrafish. Mech Dev 2004, 121:371-386. 42. Bellipanni G, Varga M, Maegawa S, Imai Y, Kelly C, Myers AP, Chu F, Talbot WS, Weinberg ES: Essential and opposing roles of 402 Pattern formation and developmental mechanisms Current Opinion in Genetics Development 2009, 19:396–403 www.sciencedirect.com
  • 8. zebrafish beta-catenins in the formation of dorsal axial structures and neurectoderm. Development 2006, 133:1299-1309. 43. Erter CE, Wilm TP, Basler N, Wright CV, Solnica-Krezel L: Wnt8 is required in lateral mesendodermal precursors for neural posteriorization in vivo. Development 2001, 128:3571-3583. 44. Lekven AC, Thorpe CJ, Waxman JS, Moon RT: Zebrafish wnt8 encodes two wnt8 proteins on a bicistronic transcript and is required for mesoderm and neurectoderm patterning. Dev Cell 2001, 1:103-114. 45. Varga M, Maegawa S, Bellipanni G, Weinberg ES: Chordin expression, mediated by Nodal and FGF signaling, is restricted by redundant function of two beta-catenins in the zebrafish embryo. Mech Dev 2007, 124:775-791. 46. Reim G, Brand M: Maternal control of vertebrate dorsoventral axis formation and epiboly by the POU domain protein Spg/ Pou2/Oct4. Development 2006, 133:2757-2770. 47. Lachnit M, Kur E, Driever W: Alterations of the cytoskeleton in all three embryonic lineages contribute to the epiboly defect of Pou5f1/Oct4 deficient MZspg zebrafish embryos. Dev Biol 2008, 315:1-17. 48. Lunde K, Belting HG, Driever W: Zebrafish pou5f1/pou2, homolog of mammalian Oct4, functions in the endoderm specification cascade. Curr Biol 2004, 14:48-55. 49. Reim G, Mizoguchi T, Stainier DY, Kikuchi Y, Brand M: The POU domain protein spg (pou2/Oct4) is essential for endoderm formation in cooperation with the HMG domain protein casanova. Dev Cell 2004, 6:91-101. 50. Holloway BA, Gomez de la Torre Canny S, Ye Y, Slusarski DC, Freisinger CM, Dosch R, Chou MM, Wagner DS, Mullins MC: A novel role for MAPKAPK2 in morphogenesis during zebrafish development. PLoS Genet 2009, 5:e1000413. This study identifies an unanticipated role for MAPKAPK2 and p38 MAP kinase in modulating a marginal constrictive force acting in zebrafish epiboly, providing novel molecular insight into this still poorly understood tissue morphogenesis process. 51. Lukas SM, Kroe RR, Wildeson J, Peet GW, Frego L, Davidson W, Ingraham RH, Pargellis CA, Labadia ME, Werneburg BG: Catalysis and function of the p38 alpha, MK2a signaling complex. Biochemistry 2004, 43:9950-9960. 52. Giraldez AJ, Cinalli RM, Glasner ME, Enright AJ, Thomson JM, Baskerville S, Hammond SM, Bartel DP, Schier AF: MicroRNAs regulate brain morphogenesis in zebrafish. Science 2005, 308:833-838. 53. Bushati N, Cohen SM: microRNA functions. Annu Rev Cell Dev Biol 2007, 23:175-205. 54. Schier AF, Giraldez AJ: MicroRNA function and mechanism: insights from zebra fish. Cold Spring Harb Symp Quant Biol 2006, 71:195-203. 55. Draper BW, McCallum CM, Moens CB: nanos1 is required to maintain oocyte production in adult zebrafish. Dev Biol 2007, 305:589-598. 56. Mishima Y, Giraldez AJ, Takeda Y, Fujiwara T, Sakamoto H, Schier AF, Inoue K: Differential regulation of germline mRNAs in soma and germ cells by zebrafish miR-430. Curr Biol 2006, 16:2135-2142. 57. Kedde M, Strasser MJ, Boldajipour B, Oude Vrielink JA, Slanchev K, le Sage C, Nagel R, Voorhoeve PM, van Duijse J, Orom UA et al.: RNA-binding protein Dnd1 inhibits microRNA access to target mRNA. Cell 2007, 131:1273-1286. This study demonstrates that the vertebrate-specific RNA binding protein Dnd, essential for germ line development in zebrafish and mouse, but previously of unknown molecular function, acts by blocking miRNA- mediated gene silencing in zebrafish germ cells and in cultured human cells. Dnd binds to a U-rich site in the 30 UTR of regulated transcripts that, in a still unknown manner, blocks miRNA binding. 58. Slanchev K, Stebler J, Goudarzi M, Cojocaru V, Weidinger G, Raz E: Control of dead end localization and activity– implications for the function of the protein in antagonizing miRNA function. Mech Dev 2009, 126:270-277. 59. Weidinger G, Stebler J, Slanchev K, Dumstrei K, Wise C, Lovell- Badge R, Thisse C, Thisse B, Raz E: dead end, a novel vertebrate germ plasm component, is required for zebrafish primordial germ cell migration and survival. Curr Biol 2003, 13:1429-1434. 60. Kuramochi-Miyagawa S, Kimura T, Ijiri TW, Isobe T, Asada N, Fujita Y, Ikawa M, Iwai N, Okabe M, Deng W et al.: Mili, a mammalian member of piwi family gene, is essential for spermatogenesis. Development 2004, 131:839-849. 61. Cox DN, Chao A, Lin H: piwi encodes a nucleoplasmic factor whose activity modulates the number and division rate of germline stem cells. Development 2000, 127:503-514. 62. Pal-Bhadra M, Bhadra U, Birchler JA: RNAi related mechanisms affect both transcriptional and posttranscriptional transgene silencing in Drosophila. Mol Cell 2002, 9:315-327. 63. Siegfried KR, Nusslein-Volhard C: Germ line control of female sex determination in zebrafish. Dev Biol 2008, 324:277-287. 64. Slanchev K, Stebler J, de la Cueva-Mendez G, Raz E: Development without germ cells: the role of the germ line in zebrafish sex differentiation. Proc Natl Acad Sci U S A 2005, 102:4074-4079. 65. Aravin AA, Sachidanandam R, Girard A, Fejes-Toth K, Hannon GJ: Developmentally regulated piRNA clusters implicate MILI in transposon control. Science 2007, 316:744-747. 66. Carmell MA, Girard A, van de Kant HJ, Bourc’his D, Bestor TH, de Rooij DG, Hannon GJ: MIWI2 is essential for spermatogenesis and repression of transposons in the mouse male germline. Dev Cell 2007, 12:503-514. 67. Ciruna B, Weidinger G, Knaut H, Thisse B, Thisse C, Raz E, Schier AF: Production of maternal-zygotic mutant zebrafish by germ-line replacement. Proc Natl Acad Sci U S A 2002, 99:14919-14924. 68. Theodosiou NA, Xu T: Use of FLP/FRT system to study Drosophila development. Methods 1998, 14:355-365. 69. Khakhar RR, Cobb JA, Bjergbaek L, Hickson ID, Gasser SM: RecQ helicases: multiple roles in genome maintenance. Trends Cell Biol 2003, 13:493-501. 70. Xie J, Bessling SL, Cooper TK, Dietz HC, McCallion AS, Fisher S: Manipulating mitotic recombination in the zebrafish embryo through RecQ helicases. Genetics 2007, 176:1339-1342. 71. Nasevicius A, Ekker SC: Effective targeted gene ‘knockdown’ in zebrafish. Nat Genet 2000, 26:216-220. 72. Draper BW, Morcos PA, Kimmel CB: Inhibition of zebrafish fgf8 pre-mRNA splicing with morpholino oligos: a quantifiable method for gene knockdown. Genesis 2001, 30:154-156. 73. Doyon Y, McCammon JM, Miller JC, Faraji F, Ngo C, Katibah GE, Amora R, Hocking TD, Zhang L, Rebar EJ et al.: Heritable targeted gene disruption in zebrafish using designed zinc- finger nucleases. Nat Biotechnol 2008, 26:702-708. 74. Meng X, Noyes MB, Zhu LJ, Lawson ND, Wolfe SA: Targeted gene inactivation in zebrafish using engineered zinc-finger nucleases. Nat Biotechnol 2008, 26:695-701. 75. Foley JE, Yeh JR, Maeder ML, Reyon D, Sander JD, Peterson RT, Joung JK: Rapid mutation of endogenous zebrafish genes using zinc finger nucleases made by Oligomerized Pool ENgineering (OPEN). PLoS ONE 2009, 4:e4348. Maternal Zebrafish Development Abrams and Mullins 403 www.sciencedirect.com Current Opinion in Genetics Development 2009, 19:396–403