ArticlePDF AvailableLiterature Review

RNA localization mechanisms in oocytes

Authors:

Abstract and Figures

In many animals, normal development depends on the asymmetric distribution of maternal determinants, including various coding and noncoding RNAs, within the oocyte. The temporal and spatial distribution of localized RNAs is determined by intricate mechanisms that regulate their movement and anchoring. These mechanisms involve cis-acting sequences within the RNA molecules and a multitude of trans-acting factors, as well as a polarized cytoskeleton, molecular motors and specific transporting organelles. The latest studies show that the fates of localized RNAs within the oocyte cytoplasm are predetermined in the nucleus and that nuclear proteins, some of them deposited on RNAs during splicing, together with the components of the RNA-silencing pathway, dictate the proper movement, targeting, anchoring and translatability of localized RNAs.
Balbiani body in the oocytes of insects and Xenopus. (A) Fragment of the ovariole from the ovary of the cricket Acheta domesticus seen with a Nomarski contrast microscope. This is a panoistic type of ovary. In this type of ovary (unlike in the meroistic type found in Drosophila), there are no nurse cells, and the oocyte nucleus (the germinal vesicle, GV) is large and transcriptionally active. In the previtellogenic oocytes (on the left), there is one Balbiani body (arrow) located at the anterior pole of the oocyte. In older oocytes, there are two Balbiani bodies, one at the anterior and another at the posterior pole (marked by a star) (for details, see Bradley et al., 2001). (B) Balbiani bodies (mitochondrial clouds) in stage I and stage II Xenopus oocytes. Whole mount in situ hybridization with antisense Xcat2 probe shows that Xcat2 mRNA is localized in the mitochondrial cloud (arrow), which always faces the vegetal pole (star) of the oocyte. In stage I oocytes, the mitochondrial cloud is located close to the GV and, in stage II oocytes, the mitochondrial cloud translocates to the vegetal pole. (C) Mitochondrial cloud from stage I Xenopus oocyte. Threedimensional reconstruction from 21 serial electron microscopic sections of an oocyte hybridized to the Xcat2 RNA probe, artificially colored. Germinal granules (arrows indicate the individual granules), labeled with Xcat2 mRNA (red), are located between the mitochondria and concentrated in the METRO region at the vegetal apex (star) of the cloud (for details, see Kloc et al., 2002). Bar, 60 µm in A, 120 µm in B, and 6 µm in C.
… 
Model illustrating the dependence of the fate of localized endogenous and exogenous RNAs in Xenopus oogenesis on their nuclear/cytoplasmic history. Endogenous (A) and exogenous (B) early-and late-pathway RNAs. (1) In stage I oocytes, the earlypathway RNAs, such as Xcat2 mRNA, bind nuclear proteins (possibly Sm proteins) that facilitate transport (via nuclear pores) to the mitochondrial cloud (yellow) and germinal granules (red spheres). The late-pathway RNAs, such as Vg1 mRNA, bind to Vg1RPB/Vera and hnRNP, which form a core complex facilitating export from the nucleus, and diffuse uniformly within the oocyte cytoplasm. (2) Later in oogenesis (starting from late stage II or early stage III), Staufen and Prrp proteins are added to the core complex assembled on late-pathway RNAs. These bind to a molecular motor, such as kinesin I and/or II, that transports RNAs on the MT tracks that form a wedge around the remnants of the mitochondrial cloud. (3) Early-and late-pathway RNAs injected into the nuclei of stage I/early stage II oocytes that bind the appropriate nuclear factors and mimic the localization pattern of their endogenous counterparts. There is no information on the fate of early-or late-pathway RNAs injected into the cytoplasm of stage I oocytes. (4) Early-pathway RNAs injected into the nucleus or cytoplasm of stage III, or older, oocytes behave like late-pathway RNAs migrating on the MTs towards the vegetal cortex. This indicates that the early-pathway binding factors present in the nuclei of stage I/early stage II oocytes are either absent or unavailable in older oocytes. However, the early-pathway RNAs can bind some of the cytoplasmic factors of the late-pathway machinery, and they either mimic the movement of late-pathway RNAs or piggyback on late-pathway RNAs. Latepathway RNAs injected into the nuclei or cytoplasm of older oocytes bind the appropriate factors and after export into the cytoplasm behave like their endogenous counterparts-either assembling their own transport complexes or piggybacking on endogenous RNAs.
… 
Content may be subject to copyright.
Introduction
The early stages of embryogenesis are dependent on the proper
spatial and temporal distribution within the oocyte (and
ultimately within the egg) of the vast stockpile of maternal
molecules accrued during the elaborate and lengthy process of
oogenesis. Among these molecules, various localized RNAs
are crucial players involved in the generation of oocyte polarity
and/or patterning of the embryo. In addition, certain RNAs
localized in germinal granules (also know as P-granules or
polar granules) of the oocyte are involved in the specification
of germ cell fate. Some of these localized RNAs are noncoding
structural or regulatory RNAs, whereas others become
translated into localized proteins. Here, we discuss recent work
that has provided insight into the multifaceted phenomenon of
RNA localization in oocytes.
Roles of localized RNAs
In many invertebrates and vertebrates, the position of the
anterior-posterior and dorsal-ventral axes of the embryo and
the formation of the germ line are controlled by the asymmetric
localization of various RNAs (and/or their translation products)
in the oocyte (Hashimoto et al., 2004; Jansen, 2001; King et
al., 1999; Kloc et al., 2001; Kloc et al., 2002a; Palacios and
Johnston, 2001; Riechmann and Ephrussi, 2001; Zhou and
King, 2004). In Drosophila, the localization of nanos and oskar
mRNAs to the posterior pole of the oocyte, and bicoid RNA to
the anterior pole, plays a role in the formation of the anterior-
posterior body axis, pole plasm assembly and germ cell
function. Mislocalization of oskar RNA in females that have a
fourfold increase in osk gene dosage or in females bearing the
osk bcd 3UTR transgene (which contains the bicoid anterior
localization signal) results in misexpression of Oskar protein,
which in turn causes mislocalization of Nanos protein and
repression of bicoid. This results in a bicaudal embryo, its
anterior replaced by a mirror image of the posterior (Ephrussi
and Lehmann, 1992; Kim-Ha et al., 1991; Smith et al., 1992;
Wharton and Struhl, 1991). Another localized mRNA, gurken,
signals to the epithelial cells surrounding the oocyte for the
proper formation of the dorsal-ventral axis (Gonzales-Reyes et
al., 1995; Neuman-Silberberg and Shüpbach, 1993; Nilson and
Shüpbach, 1999).
In ascidians, the localized mRNAs macho-1 and pem
determine muscle fate and anterior and dorsal patterning,
respectively (Nishida and Sawada, 2001; Yoshida et al., 1996).
In sea urchin, the frog Xenopus laevis and zebrafish, subsets of
RNAs are also localized to the animal and vegetal poles of the
oocyte (Braat et al., 1999; Bruce et al., 2003; Di Carlo et al.,
2004; Howley and Ho, 2000; King et al., 1999; Kloc et al.,
2001; Kloc et al., 2002a). In Xenopus oocytes, the vegetally
localized mRNAs Vg1, VegT and Xwnt 11 are mesodermal and
endodermal determinants and determinants of the left-right
axis in the embryo (Hyatt and Yost, 1998; Joseph and Melton,
1998; Ku and Melton, 1993; Rebagliati et al., 1985; Thomsen
and Melton, 1993; Zhang et al., 1998; Zhang and King, 1996).
Other vegetally localized mRNAs in Xenopus, such as Xcat2
and Xdazl, are believed to play a role in germ cell
determination or migration (Houston et al., 1998; Mosquera et
al., 1993). In zebrafish, several mRNAs are localized either to
the animal or vegetal pole of the oocyte (Howley and Ho,
2000). Two of them: the mRNA encoding the DEAD-box RNA
helicase Vasa and the mRNA encoding the T-box gene product
eomesodermin are cortically localized in vitellogenic oocytes,
and might play a role in germ cell determination and the
specification of the organizer, respectively (Bruce et al., 2003;
Knaut et al., 2000; Knaut et al., 2002).
Mechanisms of RNA transport
RNA is exported from the oocyte nucleus through nuclear
pores. Once in the cytoplasm, RNA can be trapped by or
targeted to specialized transporting organelles or bind to
cytoskeletal elements that assist in transport of RNA to its
ultimate destination. Alternatively, asymmetrical distribution
269
In many animals, normal development depends on the
asymmetric distribution of maternal determinants,
including various coding and noncoding RNAs, within the
oocyte. The temporal and spatial distribution of localized
RNAs is determined by intricate mechanisms that regulate
their movement and anchoring. These mechanisms involve
cis-acting sequences within the RNA molecules and a
multitude of trans-acting factors, as well as a polarized
cytoskeleton, molecular motors and specific transporting
organelles. The latest studies show that the fates of localized
RNAs within the oocyte cytoplasm are predetermined in
the nucleus and that nuclear proteins, some of them
deposited on RNAs during splicing, together with the
components of the RNA-silencing pathway, dictate the
proper movement, targeting, anchoring and translatability
of localized RNAs.
Key words: RNA localization, Microtubules, Oocytes
Summary
RNA localization mechanisms in oocytes
Malgorzata Kloc and Laurence D. Etkin*
Department of Molecular Genetics, The University of Texas, M. D. Anderson Cancer Center, Houston, TX 77030, USA
*Author for correspondence (e-mail: lde@mdanderson.org)
Journal of Cell Science 118, 269-282 Published by The Company of Biologists 2005
doi:10.1242/jcs.01637
Commentary
Journal of Cell Science
270
can be achieved through a combination of RNA destabilization/
degradation and local protection (Tadros et al., 2003). In
certain animals, the oocyte nucleus is transcriptionally
quiescent and most RNAs are synthesized in the accessory cells
(called nurse cells in insects), from which they are exported to
the oocyte. In this case, independent mechanisms are often
responsible for RNA transport to and within the oocyte.
Specialized transporting organelles
Two pathways of RNA localization to the oocyte vegetal cortex
have been identified in Xenopus – the early pathway and the
late pathway (Fig. 1). The early pathway, also known as the
messenger transport organizer (METRO) pathway, functions in
early oogenesis (stages I and II) and transports germinal
granules, various RNAs (some of them
involved in germ cell specification), and
germline-specific mitochondria in a
specialized organelle that in Xenopus is
called the mitochondrial cloud (also known
as the Balbiani body) (Heasman et al., 1984;
Forristall et al., 1995; Kloc and Etkin, 1995)
(Figs 1 and 2). The Balbiani body was
discovered by von Wittich in 1845 in oocytes
of spiders. Between 1864-1893, Balbiani
conducted comprehensive studies of this
organelle in oocytes of myriapods and
spiders; later, this organelle was discovered
in oocytes of various invertebrates and
vertebrates, including humans (de Smedt et
al., 2000; Guraya, 1979; Heasman et al.,
1984; Kloc et al., 2004b). Interestingly, the
existence of the Balbiani body in the oocytes
of marsupials and eutherian mammals
(including nonhuman primates and humans)
is not widely known. This situation arises
primarily from the fact that the mouse, the
dominant model organism in mammalian
studies, does not have a Balbiani body in its
oocytes. In this respect, the mouse is
probably an exception among mammals.
However, as a consequence of its absence in
the mouse, there is not a single study that
experimentally addresses the role of the
Balbiani body in mammalian oocytes.
The Balbiani body is a spherical
membraneless structure that forms in contact
with the oocyte nucleus in early oogenesis,
and fragments and disperses in late
oogenesis (Figs 1 and 2). Its constant
components are mitochondria, endoplasmic
reticulum (ER), membranous vesicles and
lipid droplets. In Xenopus stage I oocytes,
the Balbiani body is ~40 µm in diameter,
contains half a million mitochondria, which
differ in morphology and metabolism from
those of cytoplasmic mitochondria, and is
rich in membranous vesicles, and ER
cysternae. The vegetal apex of the Xenopus
Balbiani body (called the METRO region)
contains germinal granules and various
localized RNAs (Fig. 2) (Forristall et al., 1995; Heasman et al.,
1984; Kloc et al., 2004b). For nearly 150 years, the role of the
Balbiani body remained a complete mystery, although on the
basis of its biochemical and ultrastrucural composition, it was
commonly believed that this organelle participates either in the
formation of lipids or in the multiplication of mitochondria
(Guraya, 1979). In 1993, Kloc et al. showed that the Balbiani
body/mitochondrial cloud in Xenopus is a vehicle that
transports localized RNA to the vegetal pole of the oocyte.
Cis-acting elements directing RNA to the mitochondrial
cloud (mitochondrial cloud localization elements, MCLEs)
have been identified in the 3UTR of Xcat2 mRNA and in the
noncoding Xlsirts RNA in Xenopus oocytes (Allen et al., 2003;
Kloc et al., 1993; Zhou and King, 1996). Xcat2 mRNA also
has a germinal granule localization element (GGLE) in its
Journal of Cell Science 118 (2)
A
B
Early (METRO) pathway
Late (Vg1) pathway
1
2
3
4
5
6
Fig. 1. Pathways of RNA localization in Xenopus oogenesis. (A) The early (METRO)
localization pathway operates in early oogenesis (stages I and II) and uses the
mitochondrial cloud (Balbiani body) to localize RNAs such as Xcat2 mRNA (red) and
noncoding Xlsirts RNA (blue) to the vegetal pole of the oocyte. (1) In stage I oocytes,
RNAs synthesized in the nucleus (yellow) enter (either via nuage or by the
diffusion/entrapment mechanism) the mitochondrial cloud (mitochondria shown in green),
which faces the vegetal pole of the oocyte. Xcat2 mRNA becomes localized to the
germinal granules (red spheres) and Xlsirts RNA is localized between the germinal
granules at the vegetal apex (METRO region) of the mitochondrial cloud. (2) In stage II
oocytes, the mitochondrial cloud moves to the vegetal cortex and starts to disperse. (3) In
stage III-VI oocytes, the mitochondrial cloud disperses, and germinal granules and
localized RNAs form a disc at the apex of the vegetal cortex. (B) The late (Vg1) pathway
localizes mRNAs such as Vg1 or VegT, using MTs, molecular motors and possibly the
ER. (4) In stage I oocytes, these RNAs (purple) are uniformly distributed within the
oocyte cytoplasm and are excluded from the mitochondrial cloud. (5) In late stage II
oocytes, RNAs concentrate, in a wedge, around the moving mitochondrial cloud and a
subdomain of ER, and translocate on MTs towards the vegetal pole. (6) Later in oogenesis
(stages III-VI), late-pathway RNAs localize and anchor at the cortex of the vegetal half of
the oocyte (for details, see Kloc and Etkin, 1995).
Journal of Cell Science
271
RNA localization mechanisms
3UTR (Kloc et al., 2000). Recently, Claussen et al. identified
a 300-nucleotide element in the 5UTR of XNIF mRNA that
directs it to the mitochondrial cloud (Claussen et al., 2004).
The localization of RNAs into the mitochondrial cloud is
independent of microfilaments and MTs and probably involves
a diffusion and entrapment mechanism (Kloc et al., 1996;
Chang et al., 2004). The movement of the mitochondrial cloud
to the vegetal cortex of the oocyte is also independent of
MTs and microfilaments, and the mechanism underlying this
movement remains a mystery; one possibility is passive
translocation along the cytoplasm streaming towards the
vegetal cortex (Kloc et al., 2004b).
The Balbiani body has recently been discovered in the
oocytes of several insects (Fig. 2) (Bradley et al., 2001; Cox
and Spradling, 2003; Jaglarz et al., 2003; Kloc et al., 2004b).
In Drosophila, it contains germline-specific mitochondria,
forms in the oocyte in early oogenesis, and might facilitate
localization of oskar mRNA to the posterior pole of the oocyte
(Cox and Spradling, 2003). The Balbiani body differs from
Drosophila sponge bodies, which were previously thought to
be equivalent to the Xenopus Balbiani body. The sponge bodies
form at the surface of nurse cell nuclei later in oogenesis,
associate with Exuperantia (Exu), a novel protein that is a core
component of a protein complex involved in the localization of
mRNAs within the nurse cells and oocyte, and mediate
transport of bicoid RNA (Wilsch-Brauninger et al., 1997).
Another ooplasmic organelle involved in the localization of
RNAs in oocytes is the ER. In ascidians, the maternal mRNAs
pem and macho-1 are localized in the egg on the network of
cortical rough ER, which compacts after fertilization to form
the centrosome-attracting body (CAB), which is responsible
for the unequal cleavages (Sardet et al., 2003). Recently, Chang
et al. showed that, in Xenopus, the ER vesicles are involved in
the entrapment of Xcat2 and Xdazl mRNA within the
mitochondrial cloud (Chang et al., 2004). Since these
experiments used injected synthetic fluorescent RNA
synthesized from the Xcat2 3UTR and Xdazl localization
element, it remains to be seen whether the same mechanism is
involved in the localization of endogenous Xcat2 and Xdazl
mRNAs to the mitochondrial cloud.
MT-dependent transport
Oocytes have a highly polarized system of MTs and molecular
motors that are responsible for the proper localization of most
localized RNAs in Drosophila and of late-pathway (also known
as the Vg1 pathway) RNAs in Xenopus (Fig. 1).
In typical somatic cells, MT minus-ends are arranged around
a MT-organizing center (MTOC) close to the nucleus, and MT
plus-ends occupy the cell periphery. This polarity is recognized
by molecular motors, which are typically unidirectional –
either minus-end directed or plus-end directed (Cohen, 2002;
Lopez de Heredia and Jansen, 2004; Welte, 2004). However,
traffic along the MTs in oocytes is much more complex than
previously thought. As a consequence, different experiments
on the same subject often yield contradictory results and
conclusions, depending on the method used by the researchers
(Stephenson, 2004). Thus, some of the original papers and even
some of the review articles on MT trafficking are extremely
confusing.
Injection of exogenous RNAs and analysis of mutants
GV
GV
C
GV
GV
A
B
*
*
*
*
*
Fig. 2. Balbiani body in the oocytes of insects and Xenopus.
(A) Fragment of the ovariole from the ovary of the cricket Acheta
domesticus seen with a Nomarski contrast microscope. This is a
panoistic type of ovary. In this type of ovary (unlike in the meroistic
type found in Drosophila), there are no nurse cells, and the oocyte
nucleus (the germinal vesicle, GV) is large and transcriptionally
active. In the previtellogenic oocytes (on the left), there is one
Balbiani body (arrow) located at the anterior pole of the oocyte. In
older oocytes, there are two Balbiani bodies, one at the anterior and
another at the posterior pole (marked by a star) (for details, see
Bradley et al., 2001). (B) Balbiani bodies (mitochondrial clouds) in
stage I and stage II Xenopus oocytes. Whole mount in situ
hybridization with antisense Xcat2 probe shows that Xcat2 mRNA is
localized in the mitochondrial cloud (arrow), which always faces the
vegetal pole (star) of the oocyte. In stage I oocytes, the mitochondrial
cloud is located close to the GV and, in stage II oocytes, the
mitochondrial cloud translocates to the vegetal pole.
(C) Mitochondrial cloud from stage I Xenopus oocyte. Three-
dimensional reconstruction from 21 serial electron microscopic
sections of an oocyte hybridized to the Xcat2 RNA probe, artificially
colored. Germinal granules (arrows indicate the individual granules),
labeled with Xcat2 mRNA (red), are located between the
mitochondria and concentrated in the METRO region at the vegetal
apex (star) of the cloud (for details, see Kloc et al., 2002). Bar, 60
µm in A, 120 µm in B, and 6 µm in C.
Journal of Cell Science
272
indicate that, in Drosophila, oskar mRNA is localized to the
posterior pole by plus-end-directed motors (kinesins), whereas
bicoid mRNA is localized to the anterior pole and gurken
mRNA is localized to the anterior-dorsal region by minus-end-
directed motors (dyneins) (Brendza et al., 2000; Cohen, 2002;
Lopez de Heredia and Jansen, 2004). These findings raise a
fascinating question: how can the same molecular motor direct
bicoid and gurken RNAs to different cellular locations?
Recently, MacDougall et al. used time-lapse photography to
track the localization of fluorescent gurken RNA in living
Drosophila oocytes (MacDougall et al., 2003). They showed
that the movement of gurken RNA is dependent on dynein and
MTs but occurs in two distinct steps using distinct arrays of
MTs. First, gurken particles move to the oocyte anterior; later,
they turn dorsally towards the nucleus. The authors suggested
that, in this instance, as well as in others, the use of distinct
MT networks might be responsible for transport by the same
motor of different cargos to different destinations. Recent
experiments showing that kinesin heavy chain (Khc) mutants
that exhibit impaired oskar localization also, unexpectedly,
exhibit impaired localization of gurken and bicoid mRNA
indicate that anterior and anterior-dorsal transport also require
kinesin (Duncan and Warrior, 2002; Welte, 2004). Several
models for cooperation between minus-end-directed and plus-
end-directed motors in the oocyte have been proposed. For
example, kinesin might indirectly activate dynein (through an
adaptor protein) or these two motors might cooperate by
recycling one another – i.e. kinesin could transport dynein to
MT plus-ends, where dynein would bind RNA cargo and
transport it to the minus-ends, and vice versa (Cohen, 2002;
Duncan and Warrior, 2002; Januschke et al., 2002; Welte,
2004).
Further complications arise because the polarity of the
Drosophila oocyte cytoskeleton changes during oogenesis
(which is divided into 14 stages) (Theurkauf et al., 1992). Just
after formation of the oocyte (stage 1), the MTOC assembles
at the anterior pole of the oocyte and MTs extend from it to
the nurse cells. The oocyte then grows by acquiring molecules
and organelles (such as centrosomes and mitochondria) from
the nurse cells by actin-based and minus-end-directed, MT-
based transport (Swan et al., 1999). In early oogenesis (stages
2-6), the MTOC shifts from the anterior to the posterior pole,
resulting in localization of oskar mRNA and Gurken protein as
a crescent at the oocyte posterior. Subsequently, Gurken signals
to overlying follicle cells and they adopt a posterior fate. The
genes required for the MTOC shift from the anterior to the
posterior pole in early oogenesis include the armitage gene,
which encodes a homolog of SDE3, an RNA helicase involved
in post-transcriptional gene silencing, and probably other genes
whose products are involved in RNA silencing (see below),
such as spindle-E, aubergine and maelstrom. In armitage
mutants, the MTOC does not switch to the posterior pole,
which impairs posterior localization of oskar mRNA and
causes its premature translation (Chekulaeva and Ephrussi,
2004; Cook et al., 2004; Tomari et al., 2004). It is possible that
this defect in MOTC switching is caused not by the mutation
in armitage but, directly or indirectly, by the premature
synthesis of Oskar protein. However, the fact that oocytes
overexpressing Oskar from the transgene have normal MT
organization (Riechmann et al., 2002) makes it very unlikely
that MT polarization and oskar silencing are co-dependent.
At the beginning (stage 7) of mid oogenesis (stages 7-10),
the posterior follicular cells send an unknown signal back to
the oocyte, causing a re-polarization of the oocyte MTs; the
MTOC at the oocyte posterior disassembles, and new MTs are
nucleated from the anterior and lateral oocyte cortex
(Schnorrer et al., 2002). At this stage, localized oskar mRNA
becomes translated at the posterior of the oocyte, bicoid mRNA
is localized at the oocyte anterior, and gurken mRNA and
protein become located at the anterior-dorsal corner of the
oocyte, which sets up the final polarity of the oocyte.
The polarization of the Drosophila cytoskeleton seems to
require more than just a simple switch in the position of the
MTOC; it also depends on the partitioning-defective (PAR)
proteins (Huynh and St Johnston, 2004). Evolutionary
conserved PAR proteins are involved in the establishment and
maintenance of cell polarity during development and in
somatic cells in various species. These proteins include PAR-
1, a serine/threonine kinase; PAR-2, a RING finger protein;
PAR-3, which has a PDZ domain [PDZ domains are
homologous in diverse signaling proteins including
postsynaptic density protein-95 (PSD-95), a protein involved
in signaling at the post-synaptic structures; DLG, the
Drosophila Discs Large protein; and ZO-1, the zonula
occludens 1 protein]; PAR-4, a homolog of mammalian kinase
LKB1; and PAR-5, a homolog of 14-3-3 (which are highly
conserved proteins involved in the regulation of protein
phosphorylation and mitogen-activated protein kinase
pathways). In Drosophila oocytes, PAR-1 regulates posterior
patterning through phosphorylation and thus stabilization of
Oskar (Riechmann et al., 2002) and, together with other PAR
proteins, PAR-1 might control the dynamics of MT assembly.
In par mutants, the MT minus-ends do not switch from the
anterior to the posterior oocyte cortex in early oogenesis
(Vaccari and Ephrussi, 2002), and there is a disorganization of
the MT cytoskeleton in mid oogenesis in which MT minus-
ends are located around the oocyte cortex and MT plus-ends
are directed towards the oocyte center (Benton et al., 2002;
Shulman et al., 2000). Although the molecular targets and
mode of action of PAR proteins within the oocyte cytoskeleton
are unknown, it is possible that PAR kinases, like the
mammalian PAR-1 homolog, MARK, regulate MT stability by
phosphorylation of MT-associated proteins (Benton et al.,
2002; Drewes et al., 1997).
In Xenopus oocytes, several mRNAs, including Vg1 mRNA,
localize to the vegetal cortex during mid and late oogenesis
(oocyte stages III-V) via the MT-dependent, late (or Vg1)
pathway (Fig. 1) (Kloc and Etkin, 1995; Melton, 1987; Weeks
and Melton, 1987; Yisraeli et al., 1990; Zhou and King, 1996;
Zhou and King, 2004). The MT-dependent localization of Vg1
mRNA starts with the ‘streaming’ of RNA towards the vegetal
cortex around the so-called wedge region of the oocyte, which
contains a subdomain of ER organized on top of the
mitochondrial cloud. Either the ER serves as a matrix for the
formation of polarized MT tracks that transport Vg1 mRNA,
or Vg1 binds to the ER vesicles (possibly through the
Vg1RBP/Vera protein), and the ER-Vg1 mRNA complexes are
transported on the MTs to the oocyte vegetal cortex (Deshler
et al., 1997; Kloc and Etkin, 1998).
There is some evidence that the movement of Vg1 mRNA
to the Xenopus oocyte vegetal cortex is mediated by the plus-
end-directed motor kinesin II (Betley et al., 2004) and possibly
Journal of Cell Science 118 (2)
Journal of Cell Science
273
RNA localization mechanisms
by kinesin I (Yoon and Mowry, 2004). Paradoxically, the
majority (around 96%) of MTs in Xenopus stage III and VI
oocytes have their minus-ends directed towards the cortex,
which is the reverse of what is typical for eukaryotic cells
(Pfeiffer and Gard, 1999). The majority of the molecules
transported to the cortex should therefore use minus-end-
directed motors, such as cytoplasmic dyneins or minus-end-
directed kinesins. Possible explanations for these apparently
contradictory findings (i.e. that movement of Vg1 to the vegetal
cortex is mediated by plus-end-directed motors but that most
MTs have minus-ends directed towards the cortex) are that, in
Xenopus, as in Drosophila, the polarity of the MTs shifts
during oogenesis and/or that the RNA-binding factors decide
the directionality of the RNA transport. Rat neurons provide
an example of this: the ribonucleoprotein granules of the RNA-
binding protein Staufen fused to green fluorescent protein
(GFP) are able to associate with both kinesin and dynein, and
occasionally reverse the direction of their movement on MTs
(Köhrmann et al., 1999; Villace et al., 2004). Although we do
not understand the molecular events that lead to the changes in
the direction of movement along MTs, the novel Halo-like
proteins discovered in mutants that exhibit defective transport
of lipid droplets might be involved. Halo proteins might affect
the duration of the particular run of the cargo by altering the
binding strength between a particular motor and MTs, which
would change the frequency of dissociation of motor proteins
from MTs, or by controlling the number of motors that bind to
MTs. Another possibility is that Halo proteins act in trans,
changing the net direction of the cargo translocation: in the
presence of Halo, the net movement of the cargo is plus-end
directed whereas, in the absence of Halo, the net movement of
cargo is minus-end directed (Fig. 3) (Gross et al., 2003; Cohen,
2003; Welte, 2004).
RNA localization via MTs depends on the presence of
cis-acting localization elements (LEs) located in the 3UTR
(common), the 5UTR (rare), or both (Table 1) (Betley et al.,
2002; Bubunenko et al., 2002; Claussen et al., 2004; Kloc
et al., 2002a; Sasakura and Makabe, 2002; Zhou and King,
1996). Proper secondary structure is often important for the
functioning of LEs, and several LEs can act redundantly. The
best characterized are those of: Vg1, VegT, fatvg, Xvelo1 and
XNIF mRNAs and noncoding Xlsirts RNA in Xenopus (Chan
et al., 1999; Claussen and Pieler, 2004; Claussen et al., 2004;
Kloc et al., 1993; Kwon et al., 2002; Mowry and Cote, 1999;
Yaniv and Yisraeli, 2001); oskar, nanos, gurken and bicoid
mRNAs in Drosophila (Bergsten and Gavis, 1999; Brunel and
Ehresmann, 2004; Bullock and Ish-Horowicz, 2001; Crucs et
al., 2000; Kim-Ha et al., 1993; Macdonald and Kerr, 1997;
Macdonald and Kerr, 1998; Macdonald and Struhl, 1988; Thio
et al., 2000; Wagner et al., 2004); and a group of maternal
mRNAs in zebrafish and ascidians (Knaut et al., 2002;
Sasakura and Makabe, 2002).
Multiple RNA-binding proteins that interact with these
elements, and thus link RNA, MTs and molecular motors, have
been identified (Table 1), primarily in Drosophila and Xenopus.
For example, Staufen, Mago nashi, Y14 (also known as
Tsunagi), Barentsz (Btz) and Hrp48 are components of the
oskar mRNA transport complex (Hachet and Ephrussi, 2001;
Huynh et al., 2004; Mohr et al., 2001; St Johnston et al., 1991;
van Eeden et al., 2001), and Egalitarian (Egl) and Bicaudal D
(BicD) are components of the dynein motor complex in
Drosophila oocytes (Navarro et al., 2004). Staufen, a double-
stranded-RNA-binding protein is essential for the localization
of oskar and bicoid mRNA in oocytes and prospero mRNA in
neuroblasts (Li et al., 1997; Zhou and King, 2004). Mago nashi
and Y14, a putative RNA-binding protein, are highly conserved
from Schizosaccharomyces pombe to humans, and in
Drosophila they are required for the correct localization of
oskar mRNA at the posterior pole. Mago nashi and Y14
interact both in vitro and in vivo. They also interact with the
mRNA export factor TAP, and are associated with mRNA
during its export from the nucleus to the cytoplasm (Hachet
and Ephrussi, 2001; Le Hir et al., 2001a; Le Hir et al., 2001b).
Btz, which shares some sequence similarity with the human
nucleo-cytoplasmic shuttling protein malignant lymph node 51
(MLN51), is essential for the localization of oskar mRNA to
the posterior pole of the Drosophila oocyte. When expressed
in Drosophila oocytes, the mammalian homolog of Btz
interacts with mammalian Staufen in an RNA-dependent
manner (Macchi et al., 2003; van Eeden et al., 2001).
Hrp48 is a member of the heterogeneous nuclear
ribonucleoprotein A/B (hnRNPA/B) family of RNA-binding
proteins that bind 5and 3regulatory regions of oskar mRNA.
Because the Hrp48 mutant exhibits defective formation of
GFP-Staufen particles, it is believed that HRP48 plays a role
in the assembly of Staufen–oskar mRNA transport particles
(Huynch et al., 2004). BicD is essential for the organization
and maintenance of the polarized MT network and the
plus-end motor
minus-end motor
coordination machinery
HALO-like protein
-
-
A
B
RNA cargo
+
+
Fig. 3. Model of the action of Halo-like proteins in bidirectional
transport on MTs. (A) In the absence of Halo proteins, plus-end and
minus-end motors alternate their binding to coordination machinery
(possibly containing dynactin) and move RNA cargo towards either
plus-ends or minus-ends of MTs. (B) Halo proteins change the steric
properties of the coordination machinery, which weakens its binding
to the minus-end motor and increases its binding to the plus-end
motor, and results in the movement of the cargo towards the MT
plus-end [adapted from Gross (Gross, 2003) and Gross et al. (Gross
et al., 2003)].
Journal of Cell Science
274 Journal of Cell Science 118 (2)
Localization elements (LEs) and trans-acting factors
RNA LE Trans-acting factor Motor References
Drosophila bicoid mRNA Five independent
domains (I-V) in
3UTR
Staufen, BicD, Egl, Swa Dynein-dynactin
complex and
kinesin I
Brunel and, Ehresmann, 2004; Duncan and
Warrior, 2002; Januschke et al., 2002;
Macdonald and Struhl, 1988; Schnorrer
et al., 2000
Drosophila gurken mRNA GLE2 in 5UTR BicD, Egl, Hrb27C, Sqd, Otu Dynein-dynactin
complex and
kinesin I
Duncan and Warrior, 2002; Filardo and
Ephrussi, 2003; Goodrich et al., 2004;
Januschke et al., 2002; Saunders and
Cohen, 1999
Drosophila nanos mRNA
(localization is mainly
based on
diffusion/entrapment, and
a small portion localizes to
the germ plasm, possibly
through a LE in the
3UTR)
Multiple redundant
subelements in 3UTR
p75 Unknown Bergsten and Gavis, 1999; Bergsten et al.,
2001; Forrest and Gavis, 2003
Drosophila oskar mRNA Multiple subelements in
3UTR
EJC, Staufen, Barentsz Mago
nashi, Y14 (Tsunagi), Hrp48,
Tropomyosin II (TmII),
Bruno (regulation of
translation)
Kinesin I Brendza et al., 2000; Filardo and Ephrussi,
2003; Hachet and Ephrussi, 2001; Huynh
et al., 2004; Januschke et al., 2002; Kim-
Ha et al., 1993; Mohr et al., 2001; St
Johnston et al., 1991; van Eeden et al.,
2001
Xenopus late-pathway RNAs:
Vg1 mRNA
VegT mRNA
Xvelo1 mRNA
FatVg mRNA
Multiple redundant
subelements in 3UTR
Reiterated VM1 and E2
elements in 3UTR
Multiple subelements in
3UTR
Multiple redundant
subelements in 3UTR
VgRBP/Vera
VgRBP60/hnRNPI
VgRBP71 (probable translation
activator not involved in
transport)
Prrp
Strong binding to Prrp, and
weak to VgRBP/Vera
Unknown
Kinesin I and/or
II through
Staufen
Unknown
Unknown
Bubunenko et al., 2002; Cote et al., 1999;
Deshler et al., 1997; Deshler et al., 1998;
Gautreau et al., 1997; Havin et al., 1998;
Kolev and Huber, 2003; Kroll et al.,
2002; Kwon et al., 2002; Mowry, 1996;
Mowry and Melton, 1992; Yaniv and
Ysraeli, 2001; Yoon and Mowry, 2004
Claussen and Pieler, 2004
Chan et al., 1999
Xenopus early-pathway RNAs when using late pathway:
Xcat2 mRNA, Xpat mRNA
and Xwnt11
XNIF mRNA
Xlsirts noncoding RNA
LE in 3UTR
LE in 5UTR
Repeat element
VegRBP/Vera
VgRBP71
VegRBP/Vera
VgRBP71
Prrp
Unknown
Kinesin II
Unknown
Unknown
Betley et al., 2004
Yoon and Mowry, 2004
Claussen et al., 2004
Allen et al., 2003; Kloc et al., 1993
Xenopus early-pathway RNAs:
Xcat2 mRNA
XNIF mRNA
Xlsirts noncoding RNA
MCLE and GGLE
within LE
MCLE within LE
Repeat element
Unknown
62 and 64 kDa proteins
Unknown
Unknown
Unknown
Unknown
Kloc et al., 2000; Zhou and King, 1996
Claussen et al., 2004;
Allen et al., 2003; Kloc et al., 1993
Xenopus early- and late-
pathway RNAs
CAC motif in LEs Unknown Unknown Betley et al., 2002
Zebrafish vasa mRNA
(vegetal localization after
injection into Xenopus
oocytes)
LE in 3UTR Unknown Unknown Knaut et al., 2002
Ascidians type I and type II
mRNAs: HrPOPK-1,
HrPet-1, Pet-2 and Pet-3,
HrWnt5, Hr2F-1
(posterior localization
after injection into eggs)
LE in 3UTR, different
subelements for type I
and type II RNAs
Unknown Unknown Sasakura and Makabe, 2002; Sasakura et
al., 2000
Journal of Cell Science
275
RNA localization mechanisms
structural integrity of Drosophila oocytes and nurse cells. BicD
protein contains four heptad-repeat domains typical of
intermediate filament proteins and might be an integral part of
the cytoskeleton (Oh and Steward, 2001). BicD forms a
complex with Egl, which interacts directly with dynein light
chain (Dlc) through an Egl domain distinct from that
responsible for the binding to BicD (Navarro et al., 2004).
In Xenopus, Vg1RBP (also known as Vera) binds to various
subelements of LEs in Vg1, VegT and Xvelo1 mRNAs
(Claussen and Pieler, 2004; Git and Standart, 2002; Kwon et
al., 2002). The Xenopus homolog of Drosophila Staufen
associates with kinesin I and the vegetally localized RNAs Vg1
and VegT in oocytes. The function of the Vg1 localization
element (VLE) in VglRNA is blocked by the expression in
oocytes of dominant-negative XStau234 (a mutant form
of XStau containing only a double-stranded-RNA-binding
domain) (Yoon and Mowry, 2004). The presence of Staufen in
RNA-transporting complexes in Drosophila and Xenopus
indicates common elements in localization mechanisms in
invertebrates and vertebrates.
Another example of the commonality of RNA localization
mechanisms between different organisms comes from the
study of zebrafish Vasa, whose RNA is a component of the
germ plasm in the embryo. In zebrafish stage I oocytes, vasa
mRNA is uniformly distributed within the cytoplasm. In stage
II oocytes, it is localized to the cortical cup; in stage III oocytes,
it is enriched in the animal cortex. In late-stage oocytes
(starting from stage IV), vasa mRNA remains cortical and is
localized to germ plasm granules. In cleaving embryos, vasa
mRNA segregates asymmetrically to the germ plasm and
subsequently to the founder population of primordial germ cell
(PGCs). Although nothing is known about the mechanisms
responsible for the localization of vasa mRNA in zebrafish
oocytes, the asymmetrical segregation of vasa mRNA to the
germ plasm in the embryos depends on intact MTs and also on
the presence of the product of the maternal-effect gene nebel
(Howley and Ho, 2000; Knaut et al., 2000). Recently, Knaut et
al. showed that when vasa mRNA 3UTR (which directs its
localization to the germ plasm in zebrafish embryos) is fused
to a GFP reporter and injected into Xenopus oocytes, it directs
reporter to Xenopus germ plasm (Knaut et al., 2002). Although
there is no obvious similarity between the 3UTR of zebrafish
vasa mRNA and the 3UTR of the Xenopus vasa-like
DEADSouth mRNA, both are able to direct transcripts to germ
plasm in either species (Knaut et al., 2002). The experiments
discussed above show not only the universality of the
localization mechanisms between oocytes and embryos and
between different species, but also the intrinsic interspecies
similarity of the germ plasm.
Anchoring RNA at its destination
Asymmetrically localized RNAs in the oocyte must be
anchored at their final destinations. RNA can be transported
directly to its anchor, it can be captured as it diffuses by, or a
combination of these two mechanisms can be involved.
Drosophila oocytes contain an actin-filament-based anchor at
the posterior pole that requires the F-actin-binding protein
Bifocal, tropomyosin II, Dmoesin and Cap for proper
organization (Babu et al., 2004; Baum, 2002). Dmoesin is
required for the crosslinking of F-actin to the membrane of the
posterior pole. In Dmoesin-deficient flies, which have defects
in localization of oskar mRNA, the actin filament network is
loose and detached from the plasma membrane (Jankovics et
al., 2002; Polesello et al., 2002). Recently, Cha et al., studying
the role of Khc in oskar mRNA localization in Drosophila, re-
evaluated the distribution and polarity of the MTs in the
Drosophila oocyte, and the role of the posterior anchor in the
localization of oskar mRNA (Cha et al., 2002). They concluded
that, during late oogenesis, the MT minus-ends are uniformly
distributed over the oocyte cortex, the lowest MT density being
at the posterior. In stage 8, the plus-end-directed movement
mediated by kinesin transports oskar mRNA towards the
oocyte interior, preventing its association with unwanted
cortical sites. In late stage 8 and early stage 9, the majority of
MTs at the posterior pole depolymerize, unmasking (making
available) the cortical actin anchor that tethers oskar mRNA
(Fig. 4). Entrapment at the posterior pole also mediates
localization of nanos mRNA in late oogenesis and possibly also
cyclin B, gcl (germ-cell-less) and pgc (noncoding polar granule
component) RNAs (Dalby and Glover, 1992; Forrest and
Gavis, 2003; Jongens et al., 1992; Nakamura et al., 1996).
Similarly, the microfilament-rich oocyte cortex is necessary for
the anchoring of vegetally localized RNAs in Xenopus oocytes
(Elinson et al., 1993; Forristall et al., 1995; Kloc and Etkin,
1995; Klymkowsky et al., 1991; Pondel and King, 1988;
Yisraeli et al., 1990). The disruption of actin and/or cytokeratin
filaments releases late-pathway RNAs from the vegetal cortex
(Alarcon, and Elinson, 2001). Interestingly, two vegetally
localized RNAs – early-pathway noncoding Xlsirts and late-
pathway VegT mRNA – are necessary for the anchoring of
other late-pathway RNAs to the oocyte cortex (Heasman et al.,
2001; Kloc and Etkin, 1994). Indeed, we and others have
shown that Xlsirts and VegT function as structural RNAs in the
organization of the cytokeratin cytoskeleton at the vegetal
cortex of Xenopus oocytes (M.K., K. Wilk, M. Bilinski and
L.D.E., unpublished).
The fate of a localized RNA depends on its nuclear
and/or cytoplasmic history
The latest theme in the RNA localization saga concerns how
the events preceding the export of RNA into the oocyte
cytoplasm – either events in the oocyte nucleus or events in the
nuclei and cytoplasm of the accessory/nurse cells – determine
the fate of RNA, including its stability, translatability, mode of
transport and destination. The most striking finding is the
discovery that the polarity of RNA transport depends on the
source of RNA and its binding partners. Fluorescent bicoid
RNA injected directly into Drosophila oocyte cytoplasm
moves on MTs to the closest cortical surface. However, if first
injected into the nurse cell cytoplasm and then withdrawn and
re-injected into the oocyte, the bicoid RNA moves to the
anterior. Transport to the proper destination depends on the
ability of Exu protein and bicoid mRNA to be assembled into
complexes in nurse cell cytoplasm. Moreover, this finding
indicates that the direction of transport along the oocyte
microtubular system in Drosophila depends on the identity of
the proteins bound to the RNA cargo (Cha et al., 2001).
Drosophila oskar provides another example. The hybrid
RNA lacZ/oskar-3UTR is efficiently transported to the
posterior pole of the oocyte (Kim-Ha et al., 1993), which
Journal of Cell Science
276
implies that the oskar RNA 3UTR contains all signals
necessary and sufficient to direct RNA to the posterior pole.
However, the hybrid RNA cannot move to the posterior pole
in the absence of endogenous, properly spliced oskar mRNA.
This leads to the conclusion that the hybrid molecule
piggybacks on the endogenous oskar mRNA and suggests that
the oskar RNA 3UTR, although necessary, is not sufficient for
transport to the posterior pole and that the splicing events in
the nucleus regulate cytoplasmic localization of mRNA
(Hachet and Ephrussi, 2004).
Splicing not only removes introns from RNA, but also
deposits a stable protein complex called the exon-exon
junction complex (EJC) upstream of mRNA exon-exon
junctions. In Drosophila and Xenopus, the EJC, besides
containing proteins directly involved in splicing, contains the
Y14-Mago heterodimer, which binds to the nucleo-
cytoplasmic shuttling complex TAP/p15, which transports
spliced RNA through the nuclear pores. In addition, the EJC
serves as an anchoring point for the factors responsible for the
nonsense-mediated decay (NMD) of mRNA containing
premature termination codons (le Hir et al., 2001a; le Hir et
al., 2001b; Wagner and Lykke-Andersen, 2002). Hachet and
Ephrussi showed that the Drosophila EJC, deposited on oskar
mRNA in a splicing-dependent manner in the nucleus, is
necessary for the oskar mRNA 3UTR to assemble into a
functional localization complex (Hachet and Ephrussi, 2004).
The authors suggested that the first EJC landmark on oskar
mRNA mediates subsequent interactions with other factors
and specifies the structure of the oskar mRNA localization
complex.
In Xenopus oocytes, the fate of localized RNA also
probably depends on its nuclear or cytoplasmic history.
When synthetic Xcat2 mRNA is injected into the nucleus
of stage I or early stage II Xenopus oocytes, it behaves
like endogenous Xcat2 mRNA, following the
mitochondrial cloud pathway and entering germinal
granules (Fig. 5 and Table 1) (Kloc et al., 1996; Kloc et
al., 2000). However, when synthetic Xcat2 RNA is
injected into the nuclei or the cytoplasm of older oocytes
(stage III/IV), it follows the MT-dependent Vg1 pathway
and probably uses a kinesin motor (Fig. 5 and Table 1)
(Betley et al., 2004; Kloc et al., 1996; Zhou and King,
1996). Similarly, when an early-pathway XNIF mRNA
is injected into stage I oocytes it moves to the
mitochondrial cloud, but when the same RNA is injected
into stage III oocytes it follows the late (Vg1) pathway,
and its LE binds to the same proteins that bind the Vg1-
LE (Table 1) (Claussen et al., 2004).
The findings discussed above suggest that the mode of
transport in Xenopus oocytes and Drosophila oocytes
depends not only on the cis-acting elements but also on
the identity and availability of the factors with which they
associate. Indeed, modification of the localized RNA
both in the nucleus and in the cytoplasm has recently
been observed in Xenopus. Modification of the late-
pathway RNAs Vg1 and VegT in stage III/IV oocytes
begins in the nucleus, in which hnRNP1 and Vg1RBP/
Vera bind to the RNA, forming a core complex. After
export into the cytoplasm, this complex undergoes
remodeling and recruitment of additional components
(the proline-rich, RNA-binding protein Prrp and XStau)
that lead to the formation of an RNP-transporting/anchoring
complex (Kress et al., 2004; Zhao et al., 2001).
Translational regulation of localized RNAs
Another process directed by the events in the nucleus is the
regulation of translation. To produce proteins at the right time
and place within the oocyte, localized mRNAs must be
translationally silent not only during their transport but also at
the final destination until their protein product is required. In
Drosophila oocytes, Oskar protein does not accumulate before
its mRNA becomes localized to the posterior pole in mid
oogenesis (Markussen et al., 1995; Rongo et al., 1995). The
fact that unlocalized oskar mRNA is associated with
polysomes indicates that either regulated protein degradation
or inhibition of translation is involved (Benton and St Johnston,
2002; Braat et al., 2004; Riechmann et al., 2002). The Bruno
protein binds to Bruno-response elements (BREs) in the oskar
mRNA 3UTR, and mutation of these elements causes
premature translation of oskar (Kim-Ha et al., 1995). Bruno
also binds to the gurken mRNA 3UTR and plays a role in its
translational regulation (Filardo and Ephrussi, 2003).
At least eight distinct transacting factors, including Btz,
Y14-Mago (a component of the EJC; see above), Yps and Cup,
assemble with oskar mRNA into RNP particles and participate
in oskar mRNA localization and its translational repression
(Hachet and Ephrussi, 2001; Palacios et al., 2004; van Eeden
et al., 2001; Wilhelm et al., 2000). Wilhelm et al. have
proposed that an interaction between Cup and eukaryotic
initiation factor 4E (eIF4E) blocks the initiation of oskar
Journal of Cell Science 118 (2)
12 3
Ring Canal
Kinesin
Oskar mRNA
Microtubule
Cortical actin anchor
Fig. 4. Transport and anchoring of oskar mRNA in Drosophila oogenesis.
(1) In early oogenesis (stage 2-6), the MTs extend from the oocyte posterior
to the nurse cells. Then, oskar mRNA is transported from the nurse cells, on
the MTs, via ring canals, to the posterior cortex of the oocyte. (2) In mid
oogenesis (stage 8), there is a re-polarization of the oocyte MTs, the MTOC
at the oocyte posterior disassembles, and new MTs are nucleated from most
of the oocyte cortex. The plus-end-directed motor kinesin transports oskar
mRNA away from the cortex and towards the oocyte interior. (3) Subsequent
destabilization of MTs at the oocyte posterior (late stage 8 and early stage 9)
uncovers the posterior actin anchor, leading to the entrapment and
concentration of oskar mRNA at the posterior pole [adapted from Cha et al.
(Cha et al., 2004)].
Journal of Cell Science
277
RNA localization mechanisms
mRNA translation in early oogenesis (Wilhelm et
al., 2003). In late oogenesis, after oskar becomes
localized at the posterior, Cup no longer
associates with eIF4E, and this de-represses
translation (Lasko, 2003; Nakamura et al., 2004;
Wilhelm et al., 2003). The translational regulation
of oskar and gurken mRNAs also depends on the
members of the hnRNP family, such as Hrp48/p50/Hrb27 and
Squid (Goodrich et al., 2004; Gunkel et al., 1998; Yano et al.,
2004).
Little is known about translational regulation of localized
RNA in Xenopus oocytes. Vg1 mRNA undergoes 3UTR-
dependent translational repression before being localized in
stage IV oocytes (Dale et al., 1989; King et al., 1999; Tannahill
and Melton, 1989). VgRBP71, a KH-domain protein that
interacts with the Vg1 mRNA LE, acts as a translational
activator of Vg1 mRNA by promoting the removal of its
translational repressor element. VgRBP71, which has RNA-
strand-separating activity, binds to the 3 end of Vg1LE,
inducing the cleavage at an adjacent polyadenylation signal. As
a consequence, the Vg1 mRNA becomes polyadenylated at this
site and the downstream translational repressor element is
removed (Kolev and Huber, 2003). Xcat2 and Xdazl mRNAs
are probably silent throughout oogenesis. However, exogenous
Xcat2 mRNA is translated upon injection into stage IV oocytes
(Zhou and King, 1996). Since endogenous Xcat2 mRNA is
sequestered within germinal granules (Kloc et al., 1998; Kloc
et al., 2002a; Kloc et al., 2002b), it might be inaccessible to
the components of the translation machinery. By contrast,
exogenous Xcat2 RNA injected into stage III/IV oocytes and
unable to enter germinal granules is readily available to
undergo translation.
The translational silencing in Xenopus oocytes might also
depend on Y-box proteins such as mRNP3 and FRGY2/
mRNP4, which are present in mRNP storage particles and
probably mask or inhibit translation (Bouvet and Wolfe,
1994; Darnbrough and Ford, 1981; Murray et al., 1992).
34
A
B
?? ??
??
Vg1RBP/Vera
hnRNP
Prrp
Staufen & motor
Sm proteins ?
Late-pathway RNA
Injected late pathway RNA
Injected early pathway RNA
?? ?? ??
12
Early-pathway RNA
Fig. 5. Model illustrating the dependence of the fate of
localized endogenous and exogenous RNAs in
Xenopus oogenesis on their nuclear/cytoplasmic
history. Endogenous (A) and exogenous (B) early- and
late-pathway RNAs. (1) In stage I oocytes, the early-
pathway RNAs, such as Xcat2 mRNA, bind nuclear
proteins (possibly Sm proteins) that facilitate transport
(via nuclear pores) to the mitochondrial cloud (yellow)
and germinal granules (red spheres). The late-pathway
RNAs, such as Vg1 mRNA, bind to Vg1RPB/Vera and
hnRNP, which form a core complex facilitating export
from the nucleus, and diffuse uniformly within the
oocyte cytoplasm. (2) Later in oogenesis (starting
from late stage II or early stage III), Staufen and Prrp
proteins are added to the core complex assembled on
late-pathway RNAs. These bind to a molecular motor,
such as kinesin I and/or II, that transports RNAs on the
MT tracks that form a wedge around the remnants of
the mitochondrial cloud. (3) Early- and late-pathway
RNAs injected into the nuclei of stage I/early stage II
oocytes that bind the appropriate nuclear factors and
mimic the localization pattern of their endogenous
counterparts. There is no information on the fate of
early- or late-pathway RNAs injected into the
cytoplasm of stage I oocytes. (4) Early-pathway RNAs
injected into the nucleus or cytoplasm of stage III, or
older, oocytes behave like late-pathway RNAs
migrating on the MTs towards the vegetal cortex. This
indicates that the early-pathway binding factors
present in the nuclei of stage I/early stage II oocytes
are either absent or unavailable in older oocytes.
However, the early-pathway RNAs can bind some of
the cytoplasmic factors of the late-pathway machinery,
and they either mimic the movement of late-pathway
RNAs or piggyback on late-pathway RNAs. Late-
pathway RNAs injected into the nuclei or cytoplasm of
older oocytes bind the appropriate factors and after
export into the cytoplasm behave like their
endogenous counterparts – either assembling their
own transport complexes or piggybacking on
endogenous RNAs.
Journal of Cell Science
278
Interestingly, the Y-box protein Yps, related to Xenopus FrgY2
(implicated in translational silencing), was discovered in an
RNP complex containing oskar mRNA (see above) (Wilhelm
et al., 2000). Recently, Tanaka et al. suggested that, in
ascidians, translational repression of the localized mRNA
cipem also depends on the formation of an RNP complex
containing the Y-box protein CiYB (Tanaka et al., 2004).
Components of the RNA-silencing pathways as
possible regulators of RNA localization and
translation
RNA silencing regulates a plethora of vital functions in a
variety of organisms, acting through small noncoding RNAs
[short interfering (si)RNAs and/or micro (mi)RNAs] that are
produced by DICER-mediated cleavage of long double-
stranded or hairpin RNA precursors, respectively. Single-
stranded miRNA or siRNA in RNA-induced silencing
complexes (RISCs) recognize complementary target mRNA.
Perfect complementarity results in target degradation, and
partial complementarity results in translational repression
(Bartel, 2004; Dykxhoorn et al., 2003; Finnegan and Matzke,
2003; Nolan and Cogoni, 2004).
Recent observations raise the exciting possibility that
components of the RNA-silencing pathway regulate localized
RNAs in oocytes. In Drosophila, the armitage gene, which
encodes a component of the RNA-silencing pathway that is
also involved in MT polarization (Cook et al., 2004; Tomari et
al., 2004), is required for oskar mRNA silencing. Armitage is
thought to participate in the assembly of the RISC and, by
colocalizing with the oskar mRNA, it can spatially restrict
RNA-silencing activity to its particular location (Cook et al.,
2004).
Another gene required for RNA silencing in Drosophila and
efficient translation of oskar mRNA is aubergine, which
encodes a member of the RNAi-defective/Argonaute1
(RDE1/AGO1) protein family (Wilson et al., 1996). In
addition, aubergine is involved in the localization of
Maelstrom, a member of the Drosophila spindle-class family,
to the nuage (Findley et al., 2003). The nuage is a specialized,
perinuclear, electron-dense structure present in germ cells
across the animal kingdom (Kloc et al., 2004b). It is believed
to be a precursor of germinal (polar/P) granules, and recent
studies indicate that it participates in the exchange of
components between the nucleus and the germinal granules
(Findley et al., 2003; Kloc et al., 2004a; Kloc et al., 2002b). In
animals ranging from nematodes to mammals, the conserved
component of nuage is the DEAD-box RNA helicase Vasa
(Raz, 2000; Snee and Macdonald, 2004), which is probably
involved in translational control of RNAs localized in the
nuage and also, indirectly, in RNA silencing (Findley et al.,
2003). In Drosophila, Maelstrom shuttles between the nucleus
and cytoplasm, possibly via the nuage and sponge bodies
(Findley et al., 2003). Maelstrom mutants also exhibit defective
processing of Vasa and cause the mislocalization of two
proteins involved in RNA silencing, Dicer and Argonaute2,
which suggests a possible connection between the nuage and
the RNA-silencing pathway in Drosophila oocytes (Findley et
al., 2003).
Recently, a Vasa-like DEAD-box helicase and components
of the splicing machinery, including Sm proteins, have been
found in the nuage in Xenopus oocytes (Bilinski et al., 2004).
This suggests that a subset of Sm proteins have a splicing-
independent role. The nuage thus might serve as a platform for
the shuttling of proteins and RNAs between the nucleus and
cytoplasm and for the assembly of RNP particles necessary for
the correct localization of various mRNAs into the germinal
granules. These results also link the nuage and the RNA-
silencing pathway (Barbee et al., 2002; Bilinski et al., 2004;
Findley et al., 2003). Another possibility is that, as in
Drosophila, there is a connection in Xenopus oocytes between
splicing events and the fate of mRNAs localized in the nuage
and germinal granules.
Perspective
The mechanisms governing RNA localization in oocytes are
clearly extremely complex. For example, the mechanisms
governing the localization of endogenous RNAs are very often
different from those governing the localization of exogenously
introduced counterparts. The development of new imaging
technologies and in vivo techniques allowing monitoring of the
transport of endogenous RNA in real time (Bratu et al., 2003;
Cha et al., 2001; Famulok, 2004; Forrest and Gavis, 2003; Snee
and Macdonald, 2004; Stephenson, 2004) should alleviate the
problems arising from the current heterogeneity of research
techniques.
The recent findings that the fate of a localized RNA depends
on its nuclear and/or cytoplasmic history and its connection to
the RNA-silencing pathways, and the discovery that coding
and noncoding localized RNAs can play a structural role in the
organization of the oocyte cytoskeleton, should open
completely new avenues in the study of RNA localization in
oocytes. The majority of RNA localization studies in oocytes
are limited to model organisms such as Drosophila and
Xenopus, which are not necessarily reflective of other dipterans
(Bullock et al., 2004) or amphibians. For example, in directly
developing frog Eleutherodactylus coqui, in contrast to
Xenopus, the mRNAs of the Vg1 and VegT orthologs are
localized at the animal pole of the oocyte (Beckham et al.,
2003). Findings like this indicate the necessity of widening
studies to other, lesser-known species and taxa. Such studies
could reveal which phenomena and mechanisms operating in
RNA localization are fundamental to a given group of
organisms and which evolved to accommodate the specific
developmental needs of particular species.
This work was supported by grants from NSF (L.D.E.).
References
Alarcon, V. B. and Elinson, R. P. (2001). RNA anchoring in the vegetal cortex
of the Xenopus oocyte. J. Cell Sci. 114, 1731-1741.
Allen, L., Kloc, M. and Etkin, L. D. (2003). Identification and
characterization of the Xlsirts cis-acting RNA localization element.
Differentiation 71, 311-321.
Babu, K., Cai, Y., Bahri, S., Yang, X. and Chia, W. (2004). Roles of Bifocal,
Homer, and F-actin in anchoring Oskar to the posterior cortex of Drosophila
oocytes. Genes Dev. 18, 138-143.
Barbee, S. A., Lublin, A. L. and Evans, T. C. (2002). A novel function for
the Sm proteins in germ granule localization during C. elegans
embryogenesis. Curr. Biol. 12, 1502-1506.
Bartel, D. (2004). MicroRNAs: genomics, biogenesis, mechanism and
function. Cell 116, 281-297.
Journal of Cell Science 118 (2)
Journal of Cell Science
279
RNA localization mechanisms
Baum, B. (2002). Drosophila oogenesis: generating an axis of polarity. Curr.
Biol. 12, R835-R837.
Beckham, Y. M., Nath, K. and Elinson, R. P. (2003). Localization of RNAs
in oocytes of Eleutherodactylus coqui, a direct developing frog, differs from
Xenopus laevis. Evol. Dev. 5, 562-571.
Benton, R. and St Johnston, D. (2002). Cell polarity: posterior Par-1 prevents
proteolysis. Curr. Biol. 12, R479-R481.
Benton, R., Palacios, I. M. and St Johnston, D. (2002). Drosophila 14-3-
3/PAR-5 is an essential mediator of PAR-1 function in axis formation. Dev.
Cell 3, 659-671.
Bergsten, S. E. and Gavis, E. R. (1999). Role for mRNA localization in
translational activation but not spatial restriction of nanos RNA.
Development 126, 659-669.
Bergsten, S. E., Huang, T., Chatterjee, S. and Gavis, E. R. (2001).
Recognition and long-range interactions of a minimal nanos RNA
localization signal element. Development 128, 427-435.
Betley, J. N., Frith, M. C., Graber, J. H., Choo, S. and Deshler, J. O. (2002).
A ubiquitous and conserved signal for RNA localization in chordates. Curr.
Biol. 12, 1756-1761.
Betley, J. N., Heinrich, B., Versnos, I., Sardet, C., Prodon, F. and Deshler,
J. O. (2004). Kinesin II mediates Vg1 mRNA transport in Xenopus oocytes.
Curr. Biol. 14, 219-224.
Bilinski, S. M., Jaglarz, M. K., Szymanska, B., Etkin, L. D. and Kloc, M.
(2004). Sm proteins, the constituents of the spliceosome, are components of
nuage and mitochondrial cement in Xenopus oocytes. Exp. Cell Res. 299,
171-178.
Bouvet, P. and Wolfe, A. P. (1994). A role for transcription and FRGY2 in
masking maternal mRNA within Xenopus oocytes. Cell 77, 931-941.
Braat, A. K., Zandbergen, T., van de Water, S., Goos, H. J. and Zivkovic,
D. (1999). Characterization of zebrafish primordial germ cells: morphology
and early distribution of vasa RNA. Dev. Dyn. 216, 153-167.
Braat, A. K., Yan, N., Arn, E., Harrison, D. and Macdonald, P. M. (2004).
Localization-dependent oskar protein accumulation: control after the
initiation of translation. Dev. Cell 7
, 125-131.
Bradley, J. T., Kloc, M., Wolfe, K. G., Estridge, B. H. and Bilinski, S. M.
(2001). Balbiani bodies in cricket oocytes: development, ultrastructure, and
presence of localized RNAs. Differentiation 67, 117-127.
Bratu, D. P., Cha, B. J., Mhlanga, M. M., Kramer, F. R. and Tyagi, S.
(2003). Visualizing the distribution and transport of mRNAs in living cells.
Proc. Natl. Acad. Sci. USA 100, 13308-13313.
Brendza, R. P., Serbus, L. R., Duffy, J. B. and Saxton, W. M. (2000).A
function for kinesin I in the posterior transport of oskar mRNA and Staufen
protein. Science 289, 2120-2122.
Bruce, A. E. E., Howley, C., Zhou, Y., Vickers, S. L., Silver, L. M., King,
M. L. and Ho, R. K. (2003). The maternally expressed zebrafish T-box gene
eomesodermin regulates organizer formation. Development 130, 5503-5517.
Brunel, C. and Ehresmann, C. (2004). Secondary structure of the 3UTR of
bicoid mRNA. Biochimie 86, 91-104.
Bubunenko, M., Kress, T. L., Vempati, U. D., Mowry, K. L. and King, M.
L. (2002). A consensus RNA signal that directs germ layer determinants to
the vegetal cortex of Xenopus oocytes. Dev. Biol. 248, 82-92.
Bullock, S. L. and Ish-Horowicz, D. (2001). Conserved signals and
machinery for RNA transport in Drosophila oogenesis and embryogenesis.
Nature 414, 611-616.
Bullock, S. L., Stauber, M., Prell, A., Hughes, J. R., Ish-Horowicz, D. and
Schmidt-Ott, U. (2004). Differential cytoplasmic mRNA localisation
adjusts pair-rule transcription factor activity to cytoarchitecture in dipteran
evolution. Development 131, 4251-4261.
Cha, B.-J., Koppetsch, B. S. and Theurkauf, W. E. (2001). In vivo analysis
of Drosophila bicoid mRNA localization reveals a novel microtubule-
dependent axis specification pathway. Cell 106, 35-46.
Cha, B.-J., Serbus, L. R., Koppetsch, B. S. and Theurkauf, W. E. (2002).
Kinesin I-dependent cortical exclusion restricts pole plasm to the oocyte
posterior. Nat. Cell Biol. 4, 592-598.
Chan, A. P., Kloc, M. and Etkin, L. D. (1999). Fatvg encodes a new localized
RNA that uses a 25 nucleotide element (FVLE1) to localize to the vegetal
cortex of Xenopus oocytes. Development 126, 4943-4953.
Chang, P., Torres, J., Lewis, R. A., Mowry, K. L., Houliston, E. and King,
M. L. (2004). Localization of RNAs to the mitochondrial cloud in Xenopus
oocytes by entrapment and association with endoplasmic reticulum. Mol.
Biol. Cell
15, 4669-4681.
Chekulaeva, M. and Ephrussi, A. (2004). Drosophila development: RNA
interference ab ovo. Curr. Biol. 14, R428-R430.
Claussen, M. and Pieler, T. (2004). Xvelo1 uses a novel 75-nucleotide signal
sequence that drives vegetal localization along the late pathway in Xenopus
oocytes. Dev. Biol. 266, 270-284.
Claussen, M., Horvay, K. and Pieler, T. (2004). Evidence for overlapping,
but not identical, protein machineries operating in vegetal RNA localization
along early and late pathways in Xenopus oocytes. Development 131, 4263-
4273.
Cohen, R. S. (2002). Oocyte patterning: dynein and kinesin, Inc. Curr. Biol.
12, R797-R799.
Cohen, R. S. (2003). Halo: a guiding light for transport. Curr. Biol. 13, R869-
R870.
Cook, H. A., Koppetsch, B. S., Wu, J. and Theurkauf, W. E. (2004). The
Drosophila SDE3 homolog armitage is required for oskar mRNA silencing
and embryonic axis specification. Cell 116, 817-829.
Cote, C. A., Gautreau, D., Denegre, J. M., Kress, T. L., Terry, N. A. and
Mowry, K. L. (1999). A Xenopus protein related to hnRNPI has a role in
cytoplasmic RNA localization. Mol. Cell 4, 431-437.
Cox, R. T. and Spradling, A. C. (2003). A Balbiani body and the fusome
mediate mitochondrial inheritance during Drosophila oogenesis.
Development 130, 1579-1590.
Crucs, S., Chatterjee, S. and Gavis, E. R. (2000). Overlapping but distinct
RNA elements control repression and activation of nanos translation. Mol.
Cell 5, 457-467.
Dalby, B. and Glover, D. M. (1992). 3 non-translated sequences in
Drosophila cyclin B transcripts direct posterior pole accumulation late in
oogenesis and peri-nuclear association in syncytial embryos. Development
115, 989-997.
Dale, L., Matthews, G., Tabe, L. and Colman, A. (1989). Developmental
expression of the protein product of Vg1, a localized maternal mRNA in the
frog Xenopus laevis. EMBO J. 8, 1057-1065.
Darnbrough, C. H. and Ford, P. J. (1981). Identification in Xenopus laevis
of a class of oocyte-specific proteins bound to messenger RNA. Eur. J.
Biochem. 107
, 45-56.
De Smedt, V., Szöllösi, D. and Kloc, M. (2000). The Balbiani body:
asymmetry in the mammalian oocyte. Genesis 26, 208-212.
Deshler, J. O., Highet, M. I. and Schnapp, B. J. (1997). Localization of
Xenopus Vg1 mRNA by Vera protein and the endoplasmic reticulum.
Science 276, 1128-1131.
Deshler, J. O., Highett, M. I., Abramson, T. and Schnapp, B. J. (1998). A
highly conserved RNA-binding protein for cytoplasmic mRNA localization
in vertebrates. Curr. Biol. 8, 489-496.
Drewes, G., Ebneth, A., Preuss, U., Mandelkow, E. M. and Mandelkow, E.
(1997). MARK, a novel family of protein kinases that phosphorylate
microtubule-associated protein and trigger microtubule disruption. Cell 89,
297-308.
Di Carlo, M., Montana, G. and Romancino, D. P. (2004). Paracentrotus
lividus eggs contain different RNAs at the animal and vegetal poles.
Biochem. Biophys. Res. Commun. 315, 1110-1119.
Duncan, J. E. and Warrior, R. (2002). The cytoplasmic dynein and kinesin
motors have interdependent roles in patterning the Drosophila oocyte. Curr.
Biol. 12, 1982-1991.
Dykxhoorn, D. M., Novina, C. D. and Sharp, P. A. (2003). Killing the
messenger: short RNAs that silence gene expression. Nat. Rev. Mol. Cell
Biol. 4, 457-467.
Elinson, R. P., King, M. L. and Forristall, C. (1993). Isolated vegetal cortex
from Xenopus oocytes selectively retains localized RNAs. Dev. Biol. 160,
554-562.
Ephrussi, A. and Lehmann, R. (1992). Induction of germ cell formation by
oskar. Nature 358, 387-392.
Famulok, M. (2004). Green fluorescent RNA. Nature 430, 976-977.
Filardo, P. and Ephrussi, A. (2003). Bruno regulates gurken during
Drosophila oogenesis. Mech. Dev. 120, 289-297.
Findley, S. D., Tamanaha, M., Clegg, N. J. and Ruohola-Baker, H. (2003).
Maelstrom, a Drosophila spindle-class gene, encodes a protein that
colocalizes with Vasa and RDE1/AGO1 homolog, Aubergine, in nuage.
Development 130, 859-871.
Finnegan, E. J. and Matzke, M. A. (2003). The small RNA world.
J. Cell
Sci. 116, 4689-4693.
Forrest, K. M. and Gavis, E. R. (2003). Live imaging of endogenous RNA
reveals a diffusion and entrapment mechanism for nanos mRNA localization
in Drosophila. Curr. Biol. 13, 1159-1168.
Forristall, C., Pondel, M., Chen, L. and King, M. L. (1995). Patterns of
localization and cytoskeletal association of two vegetally localized RNAs,
Vg1 and Xcat-2. Development 121, 201-208.
Gautreau, D., Cote, C. A. and Mowry, K. L. (1997). Two copies of a
Journal of Cell Science
280
subelement from the Vg1 RNA localization sequence are sufficient to direct
vegetal localization in Xenopus oocytes. Development 124, 5013-5020.
Git, A. and Standart, N. (2002). The KH domains of Xenopus Vg1RBP
mediate RNA binding and self-association. RNA 8, 1319-1333.
Gonzales-Reyes, A., Elliott, H. and St Johnston, D. (1995). Polarization of
both major body axes in Drosophila by Gurken-Torpedo signaling. Nature
375, 654-658.
Goodrich, J. S., Clouse, K. N. and Schupbach, T. (2004). Hrb27C, Sqd and
Otu cooperatively regulate gurken RNA localization and mediate nurse cell
chromosome dispersion in Drosophila oogenesis. Development 131, 1949-
1958.
Gross, S. P. (2003). Dynactin: coordinating motors with opposite inclinations.
Curr. Biol. 13, R320-R322.
Gross, S. P., Guo, Y., Martines, J. R. and Welte, M. A. (2003). A determinant
of directionality of organelle transport in Drosophila embryos. Curr. Biol.
13, 1660-1668.
Gunkel, N., Yano, T., Markussen, F. H., Olsen, L. C. and Ephrussi, A.
(1998). Localization-dependent translation requires a functional interaction
between the 5 and 3 ends of oscar mRNA. Genes Dev. 12, 1652-1664.
Guraya, S. S. (1979). Recent advances in the morphology, cytochemistry, and
function of Balbiani’s vitelline body in animal oocytes. Int. Rev. Cytol. 59,
249-321.
Hachet, O. and Ephrussi, A. (2001). Drosophila Y14 shuttles to the posterior
of the oocyte and is required for oskar mRNA transport. Curr. Biol. 11,
1666-1674.
Hachet, O. and Ephrussi, A. (2004). Splicing of oskar RNA in the nucleus
is coupled to its cytoplasmic localization. Nature 428, 959-963.
Hashimoto, Y., Maegawa, S., Nagai, T., Yamaha, E., Suzuki, H., Yasuda,
K. and Inoue, K. (2004). Localized maternal factors are required for
zebrafish germ cell formation. Dev. Biol. 268, 152-161.
Havin, L., Git, A., Elisha, Z., Oberman, F., Yaniv, K., Schwartz, S. P.,
Standart. N. and Yisraeli, J. K. (1998). RNA-binding protein conserved
in both microtubule- and microfilament-based RNA localization. Genes Dev.
12
, 1593-1598.
Heasman, J., Quarmby, J. and Wylie, C. C. (1984). The mitochondrial cloud
of Xenopus oocytes. The source of germinal granule material. Dev. Biol.
105, 458-469.
Heasman, J., Wessely, O., Langland, R., Craig, E. J. and Kessler, D. S.
(2001). Vegetal localization of maternal mRNAs is disrupted by VegT
depletion. Dev. Biol. 240, 377-386.
Houston, D. W., Zhang, J., Maines, J. Z., Wasserman, S. A. and King, M.
L. (1998). A Xenopus DAZ-like gene encodes an RNA component of germ
plasm and is a functional homolog of Drosophila boule. Development 125,
171-180.
Howley, C. and Ho, R. K. (2000). mRNA localization patterns in zebrafish
oocytes. Mech. Dev. 92, 305-309.
Huynh, J. R. and St Johnston, D. (2004). The origin of asymmetry: early
polarization of the Drosophila germline cyst and oocyte. Curr. Biol. 14,
R438-R449.
Huynh, J. R., Munro, T. P., Smith-Litiere, K., Lepesant, J. A. and St
Johnston, D. (2004). The Drosophila hnRNPA/B homolog, Hrp48, is
specifically required for a distinct step in osk mRNA localization. Dev. Cell
6, 625-635.
Hyatt, B. A. and Yost, J. (1998). The left-right coordinator: the role of Vg1
in organizing left-right axis formation. Cell 93, 37-46.
Jaglarz, M. K., Nowak, Z. and Bilinski, S. M. (2003). The Balbiani body
and generation of early asymmetry in the oocyte of tiger beetle.
Differentiation 71, 142-151.
Jankovics, F., Sinka, R., Lukacsovich, T. and Erdelyi, M. (2002). Moesin
crosslinks actin and cell membrane in Drosophila oocytes and is required
for oskar anchoring. Curr. Biol. 12, 2060-2065.
Jansen, R. P. (2001). mRNA localization: message on the move. Nat. Rev.
Mol. Cell Biol. 2, 247-256.
Januschke, J., Gervais, L., Dass, S., Kaltschmidt, J. A., Lopez-Schier, H.,
St Johnston, D., Brand, A. H., Roth, S. and Guichet, A. (2002). Polar
transport in the Drosophila oocyte requires dynein and kinesin I cooperation.
Curr. Biol. 12, 1971-1981.
Jongens, T. A., Hay, B., Jan, L. Y. and Jan, Y. N. (1992). The germ cell-less
gene product: a posteriorly localized component necessary for germ cell
development in Drosophila. Cell 70, 569-584.
Joseph, E. and Melton, D. (1998). Mutant Vg1 ligands disrupt endoderm and
mesoderm formation in Xenopus embryos. Development 125, 2677-2685.
Kim-Ha, J., Smith, J. L. and Macdonald, P. M. (1991). Oskar mRNA is
localized to the posterior pole of Drosophila oocyte. Cell 66, 23-35.
Kim-Ha, J., Webster, P. J., Smith, J. L. and Macdonald, P. M. (1993).
Multiple regulatory elements mediate distinct steps in localization of oskar
mRNA. Development 119, 169-178.
Kim-Ha, J., Kerr, K. and Macdonald, P. M. (1995). Translational regulation
of oskar mRNA by bruno, an ovarian RNA-binding protein, is essential. Cell
81, 403-412.
King, M. L., Zhou, Y. and Bubunenko, M. (1999). Polarizing genetic
information in the egg: RNA localization in the frog oocyte. Bioessays 21,
546-557.
Kloc, M. and Etkin, L. D. (1994). Delocalization of Vg1 mRNA from the
vegetal cortex in Xenopus oocytes after destruction of Xlsirt RNA. Science
265, 1101-1103.
Kloc, M. and Etkin, L. D. (1995). Two distinct pathways for the localization
of RNAs at the vegetal cortex in Xenopus oocytes. Development 121, 287-
297.
Kloc, M. and Etkin, L. D. (1998). Apparent continuity between the messenger
transport organizer and late RNA localization pathways during oogenesis in
Xenopus. Mech. Dev. 73, 95-106.
Kloc, M., Spohr, G. and Etkin, L. D. (1993). Translocation of repetitive RNA
sequences with the germ plasm in Xenopus oocytes. Science 262, 1712-
1714.
Kloc, M., Larabell, C. and Etkin, L. D. (1996). Elaboration of the messenger
transport organizer pathway for localization of RNA to the vegetal cortex
of Xenopus oocytes. Dev. Biol. 180, 119-130.
Kloc, M., Larabell, C., Chan, A. P. and Etkin, L. D. (1998). Contribution
of METRO pathway localized molecules to the organization of the germ cell
lineage. Mech. Dev. 75, 81-93.
Kloc, M., Bilinski, S., Chan, A. P. and Etkin, L. D. (2000). The targeting of
Xcat2 mRNA to the germinal granules depends on cis-acting germinal
granule localization element within the 3UTR. Dev. Biol. 217
, 221-229.
Kloc, M., Bilinski, S., Chan, A. P., Allen, L. H., Zearfoss, N. R. and Etkin,
L. D. (2001). RNA localization and germ cell determination in Xenopus. Int.
Rev. Cytol. 203, 63-91.
Kloc, M., Zearfoss, N. R. and Etkin, L. D. (2002a). Mechanisms of
subcellular mRNA localization. Cell 108, 533-544.
Kloc, M., Dougherty, M. T., Bilinski, S., Chan, A. P., Brey, E., King, M.
L., Patrick, C. W. and Etkin, L. D. (2002b). Three-dimensional
ultrastructural analysis of RNA distribution within germinal granules of
Xenopus. Dev. Biol. 241, 79-93.
Kloc, M., Bilinski, S., Dougherty, M. T., Brey, E. M. and Etkin, L. D.
(2004a). Formation, architecture and polarity of female germline cyst in
Xenopus. Dev. Biol. 266, 43-61.
Kloc, M., Bilinski, S. and Etkin, L. D. (2004b). The Balbiani body and germ
cell determinants: 150 years later. Curr. Top. Dev. Biol. 59, 1-36.
Klymkowsky, M. W., Maynell, L. A. and Nislow, C. (1991). Cytokeratin
phosphorylation, cytokeratin filament severing and the solubilization of the
maternal mRNA Vg1. J. Cell Biol. 114, 787-797.
Knaut, H., Pelegri, F., Bohmann, K., Schwarz, H. and Nüsslein-Volhard,
C. (2000). Zebrafish vasa RNA but not its protein is a component of the
germ plasm and segregates asymmetrically before germline specification. J.
Cell Biol. 149, 875-888.
Knaut, H., Steinbeisser, H., Schwarz, H. and Nüsslein-Volhard, C. (2002).
An evolutionary conserved region in the vasa 3UTR targets RNA translation
to the germ cells in the zebrafish. Curr. Biol. 12, 454-466.
Köhrmann, M., Luo, M., Kaether, C., DesGroseillers, L., Dotti, C. G. and
Kiebler, M. A. (1999). Microtubule-dependent recruitment of Staufen-green
fluorescent protein into large RNA-containing granules and subsequent
dendritic transport in living hippocampal neurons. Mol. Biol. Cell 10, 2945-
2953.
Kolev, N. G. and Huber, P. W. (2003). VgRBP71 stimulates cleavage at a
polyadenylation signal in Vg1 mRNA, resulting in the removal of a cis-
acting element that represses translation. Mol. Cell 11, 745-755.
Kress, T. L., Yoon, Y. J. and Mowry, K. L. (2004). Nuclear RNP complex
assembly initiates cytoplasmic RNA localization. J. Cell Biol. 165, 203-211.
Kroll, T. T., Zhao, W. M., Jiang, C. and Huber, P. W. (2002). A homolog
of FBP2/KSRP binds to localized mRNAs in Xenopus oocytes. Development
129, 5609-5619.
Ku, M. and Melton, D. A. (1993). Xwnt11: a maternally expressed wnt gene.
Development 119, 1161-1173.
Kwon, S., Abramson, T., Munro, T. P., John, C. M., Kohrmann, M. and
Schnapp, B. J. (2002). UUCAC- and vera-dependent localization of VegT
RNA in Xenopus oocytes. Curr. Biol. 12, 558-564.
Lasko, P. (2003). Cup-ling oskar RNA localization and translational control.
J. Cell Biol. 163, 1189-1191.
Journal of Cell Science 118 (2)
Journal of Cell Science
281
RNA localization mechanisms
Le Hir, H., Gatfield, D., Izaurralde, E. and Moore, M. J. (2001a). The exon-
exon junction complex provides a binding platform for factors involved in
mRNA export and nonsense-mediated mRNA decay. EMBO J. 20, 4987-
4997.
Le Hir, H., Gatfield, D., Braun, I. C., Forler, D. and Izaurralde, E. (2001b).
The protein Mago provides a link between splicing and mRNA localization.
EMBO Rep. 21, 1119-1124.
Li, P., Wasser, M., Cai, Y. and Chia, W. (1997). Inscuteable and Staufen
mediate asymmetric localization and segregation of prospero RNA during
Drosophila neuroblast cell divisions. Cell 90, 437-447.
Lopez de Heredia, M. and Jansen, R. P. (2004). mRNA localization and the
cytoskeleton. Curr. Opin. Cell Biol. 16, 80-85.
Macchi, P., Kroening, S., Palacios, I. M., Baldassa, S., Grunewald, B.,
Ambrosino, C., Goetze, B., Lupas, A., St Johnston, D. and Kiebler, M.
(2003). Barentsz, a new component of the Staufen-containing
ribonucleoprotein particles in mammalian cells, interacts with Staufen in an
RNA-dependent manner. J. Neurosci. 23, 5778-5788.
Macdonald, P. and Kerr, K. (1997). Redundant RNA recognition events in
bicoid mRNA localization. RNA 3, 1413-1420.
Macdonald, P. and Kerr, K. (1998). Mutational analysis of an RNA
recognition element that mediates localization of bicoid mRNA. Mol. Cell.
Biol. 18, 3788-3795.
Macdonald, P. and Struhl, G. (1988). Cis-actin sequences responsible for
localizing bicoid mRNA at the anterior pole of Drosophila embryos. Nature
336, 595-598.
MacDougall, N., Clark, A., MacDougall, E. and Davis, I. (2003).
Drosophila gurken (TGFalpha) mRNA localizes as particles that move
within the oocyte in two dynein-dependent steps. Dev. Cell. 4, 307-319.
Markussen, F. H., Michon, A. M., Breitwieser, W. and Ephrussi, A. (1995).
Translational control of oskar generates short OSK, the isoform that induces
pole plasm assembly. Development 121, 3723-3732.
Melton, D. A. (1987). Translocation of a localized maternal mRNA to the
vegetal pole of Xenopus oocytes. Nature 328, 80-82.
Mohr, S. E., Dillon, S. T. and Boswell, R. E. (2001). The RNA-binding
protein Tsunagi interacts with Mago Nashi to establish polarity and localize
oskar mRNA during Drosophila oogenesis. Genes Dev. 15, 2886-2899.
Mosquera, L., Forristall, C., Zhou, Y. and King, M. L. (1993). mRNA
localized to the vegetal cortex of Xenopus oocytes encodes a protein with a
nanos-like zinc finger domain. Development 117
, 377-386.
Mowry, K. L. (1996). Complex formation between stage-specific oocyte
factors and a Xenopus mRNA localization element. Proc. Natl. Acad. Sci.
USA 93, 14608-14613.
Mowry, K. L. and Cote, C. A. (1999). RNA sorting in Xenopus oocytes and
embryos. FASEB J. 13, 435-445.
Mowry, K. L. and Melton, D. A. (1992). Vegetal messenger RNA localization
directed by a 340-nt RNA sequence element in Xenopus oocytes. Science
255, 991-994.
Murray, M. T., Schiller, D. L. and Franke, W. W. (1992). Sequence
analysis of cytoplasmic mRNA binding proteins in Xenopus oocytes
identifies a family of RNA-binding proteins. Proc. Natl. Acad. Sci. USA
89, 11-15.
Nakamura, A., Amikura, R., Mukai, M., Kobayashi, S. and Lasko, P.
(1996). Requirement for a noncoding RNA in Drosophila polar granules for
germ cell establishment. Science 274, 2075-2079.
Nakamura, A., Sato, K. and Hanyu-Nakamura, K. (2004). Drosophila cup
is an elF4E binding protein that associates with Bruno and regulates oskar
mRNA translation in oogenesis. Dev. Cell 6, 69-78.
Navarro, C., Puthalakath, H., Adams, J. M., Strasser, A. and Lehmann,
R. (2004). Egalitarian binds dynein light chain to establish oocyte polarity
and maintain oocyte fate. Nat. Cell Biol. 6, 427-435.
Neuman-Silberberg, F. S. and Schüpbach, T. (1993). The Drosophila
dorsoventral patterning gene gurken produces a dorsally localized RNA and
encodes a TGF alpha-like protein. Cell 75, 165-174.
Nilson, L. A. and Schüpbach, T. (1999). EGF receptor signaling in
Drosophila oogenesis. Curr. Top. Dev. Biol. 44, 203-243.
Nishida, H. and Sawada, K. (2001). Macho-1 encodes a localized mRNA in
ascidian eggs that specifies muscle fate during embryogenesis. Nature 409,
724-729.
Nolan, T. and Cogoni, C. (2004). The long hand of the small RNAs reaches
into several levels of gene regulation. Biochem. Cell Biol. 82, 472-481.
Oh, J. and Steward, R. (2001). Bicaudal-D is essential for egg chamber
formation and cytoskeletal organization in drosophila oogenesis. Dev. Biol.
232, 91-104.
Palacios, I. M. and Johnston, D. S. (2001). Getting the message across: the
intracellular localization of mRNA in higher eukaryotes. Annu. Rev. Cell.
Dev. Biol. 17, 569-614.
Palacios, I. M., Gatfield, D., St Johnston, D. and Izaurralde, E.
(2004). An
elF4AIII-containing complex required for mRNA localization and nonsense
mediated mRNA decay. Nature 427, 753-757.
Pfeiffer, D. C. and Gard, D. L. (1999). Microtubules in Xenopus oocytes are
oriented with their minus-ends towards the cortex. Cell Motil. Cytoskeleton
44, 34-43.
Polesello, C., Delon, I., Valenti, P., Ferrer, P. and Payre, F. (2002). Dmoesin
controls actin based cell shape and polarity during Drosophila melanogaster
oogenesis. Nat. Cell Biol. 4, 782-789.
Pondel, M. D. and King, M. L. (1988). Localized maternal mRNA related to
transforming growth factor beta mRNA is concentrated in a cytokeratin-
enriched fraction from Xenopus oocytes. Proc. Natl. Acad. Sci. USA 85,
7612-7616.
Raz, E. (2000). The function and regulation of vasa-like genes in germ cell
development. Genome Biol. 1, 1017.1-1017.6.
Rebagliati, M. R., Weeks, D. L., Harvey, R. P. and Melton, D. A. (1985).
Identification and cloning of localized maternal RNAs from Xenopus eggs.
Cell 42, 769-777.
Riechann, V. and Ephrussi, A. (2001). Axis formation during Drosophila
oogenesis. Curr. Opin. Genet. Dev. 11, 374-383.
Riechmann, V., Gutierrez, G. J., Filardo, P., Nebreda, A. R. and Ephrussi,
A. (2002). Par-1 regulates stability of the posterior determinant Oskar by
phosphorylation. Nat. Cell Biol. 4, 337-342.
Rongo, C., Gavis, E. R. and Lehmann, R. (1995). Localization of oskar RNA
regulates oskar translation and requires Oskar protein. Development 121,
2737-2746.
Sardet, C., Nishida, H., Prodon, F. and Sawada, K. (2003). Maternal
mRNAs of PEM and macho 1, the ascidian muscle determinant, associate
and move with rough endoplasmic reticulum network in the egg cortex.
Development 130, 5839-5849.
Sasakura, Y. and Makabe, K. W. (2002). Identification of cis elements which
direct the localization of maternal mRNAs to the posterior pole of ascidian
embryos. Dev. Biol. 250, 128-144.
Sasakura, Y., Ogasawara, M. and Makabe, K. W. (2000). Two pathways of
maternal RNA localization at the posterior-vegetal cytoplasm in early
ascidian embryos. Dev. Biol. 220, 365-378.
Saunders, C. and Cohen, R. S. (1999). The role of oocyte transcription, the
5UTR, and translation repression and derepression in Drosophila gurken
mRNA and protein localization. Mol. Cell. 3, 43-54.
Schnorrer, F., Bohmann, K. and Nusslein-Volhard, C. (2000). The
molecular motor dynein is involved in targeting swallow and bicoid RNA
to the anterior pole of Drosophila oocytes. Nat. Cell Biol. 2, 185-190.
Schnorrer, F., Luschnig, S., Koch, I. and Nüsslein-Volhard, C. (2002). γ-
Tubulin37C and γ-tubulin ring complex protein 75 are essential for bicoid
RNA localization during Drosophila oogenesis. Dev. Cell 3, 685-696.
Shulman, J. M., Benton, R. and St Johnston, D. (2000). The Drosophila
homolog of C. elegans PAR-1 organizes the oocyte cytoskeleton and directs
oskar mRNA localisation to the posterior pole. Cell 101, 1-20.
Smith, J. L., Wilson, J. E. and Macdonald, P. M. (1992). Overexpression of
oskar directs ectopic activation of nanos and presumptive pole cell formation
in Drosophila embryos. Cell 70, 849-859.
Snee, M. J. and Macdonald, P. M. (2004). Live imaging of nuage and polar
granules: evidence against a precursor-product relationship and novel role
of Oskar in stabilizing of polar granule components. J. Cell Sci. 117, 2109-
2120.
St Johnston, D., Beuchle, D. and Nüslein-Volhard, C. (1991). Staufen, a
gene required to localize maternal RNAs in the Drsosophila egg. Cell 66,
51-63.
Stephenson, E. C. (2004). Localization of Swallow-Green fluorescent protein
in Drosophila oogenesis and implications for the role of Swallow in RNA
localization. Genesis 39, 280-287.
Swan, A., Nguyen, T. and Suter, B. (1999). Drosophila Lissencephaly-1
functions with Bic-D and dynein in oocyte determination and nuclear
positioning. Nat. Cell Biol. 1, 444-449.
Tadros, W., Houston, S. A., Bashirullah, A., Cooperstock, R. L., Semotok,
J. L., Reed, B. H. and Lipshitz, H. D. (2003). Regulation of maternal
transcript destabilization during egg activation in Drosophila. Genetics 164,
989-1001.
Tanaka, K. J., Matsumoto, K., Tsujimoto, M. and Nishikata, T. (2004).
CiYB1 is a major component of storage mRNPs in ascidian oocytes:
implications in translational regulation of localized mRNAs. Dev. Biol. 272,
217-230.
Journal of Cell Science
282
Tannahill, D. and Melton, D. A. (1989). Localized synthesis of the Vg1
protein during early Xenopus development. Development 106, 775-785.
Theurkauf, W. E., Smiley, S., Wong, M. L. and Alberts, B. M. (1992).
Reorganization of the cytoskeleton during Drosophila oogenesis:
implications for axis specification and intracellular transport. Development
115, 923-936.
Thio, G. L., Ray, R. P., Barcelo, G. and Schupbach, T. (2000). Localization
of gurken RNA in Drosophila oogenesis requires elements in the 5 and 3
regions of the transcript. Dev. Biol. 221, 435-446.
Thomsen, G. H. and Melton, D. A. (1993). Processed Vg1 protein is an axial
mesoderm inducer in Xenopus. Cell 74, 433-441.
Tomari, Y., Du, T., Haley, B., Schwarz, D. S., Bennett, R., Cook, H. A.,
Koppetsch, B. S., Theurkauf, W. E. and Zamore, P. D. (2004). RISC
assembly defects in the Drosophila RNAi mutant armitage. Cell 116, 831-
841.
Vaccari, T. and Ephrussi, A. (2002). The fusome and microtubules enrich
Par-1 in the oocyte, where it effects polarization in conjunction with Par-3:
BicD, Egl, and dynein. Curr. Biol. 12, 1524-1528.
van Eeden, F. J., Palacios, I. M., Petronczki, M., Weston, M. J. and St
Johnston, D. (2001). Barentsz is essential for the posterior localization of
oskar mRNA and colocalizes with it to the posterior pole. J. Cell Biol. 154,
511-523.
Villace, P., Marion, R. M. and Ortin, J. (2004). The composition of Staufen-
containing RNA granules from human cells indicates their role in the
regulated transport and translation of messenger RNAs. Nucleic Acids Res.
32, 2411-2420.
Wagner, C., Ehresmann, C., Ehresmann, B. and Brunel, C. (2004).
Mechanism of dimerization of bicoid mRNA: initiation and stabilization. J.
Biol. Chem. 279, 4560-4569.
Wagner, E. and Lykke-Andersen, J. (2002). MRNA surveillance: the perfect
persist. J. Cell Sci. 115, 3033-3038.
Weeks, D. L. and Melton, D. A. (1987). A maternal mRNA localized to the
vegetal hemisphere in Xenopus eggs codes for a growth factor related to
TGF-beta. Cell 51, 861-867.
Welte, M. A. (2004). Bidirectional transport along microtubules. Curr. Biol.
14, R525-R537.
Wharton, R. P. and Struhl, G. (1991). RNA regulatory elements mediate
control of Drosophila body pattern by posterior morphogen nanos. Cell 67,
955-967.
Wilhelm, J. E., Mansfield, N., Hom-Booher, S., Wang, C. W., Turck, T.,
Hazelrigg, T. and Vale, R. D. (2000). Isolation of a ribonucleoprotein
complex involved in mRNA localization in Drosophila oocytes. J. Cell Biol.
148, 427-440.
Wilhelm, J. E., Hilton, M., Amos, Q. and Henzel, W. J. (2003). Cup is an
elF4E binding protein required for both the translational repression of oskar
and the recruitment of Barentsz. J. Cell Biol. 163, 1197-1204.
Wilsch-Brauninger, M., Schwarz, H. and Nüsslein-Volhard, C. (1997). A
sponge-like structure involved in the association and transport of maternal
products during Drosophila oogenesis. J. Cell Biol. 139, 817-829.
Wilson, J. E., Connell, J. E. and Macdonald, P. M. (1996). aubergine
enhances oskar translation in the Drosophila ovary. Development 122, 1631-
1639.
Yaniv, K. and Yisraeli, J. K. (2001). Defining cis-acting elements and trans-
acting factors in RNA localization. Int. Rev. Cytol. 203, 521-539.
Yano, T., de Quinto, L. S., Matsui, Y., Shevchenko, A., Shevchenko, A. and
Ephrussi, A. (2004). Hrp48, a Drosophila hnRNPA/B homolog, binds and
regulates translation of oskar mRNA. Dev. Cell 6, 637-648.
Yisraeli, J. K., Sokol, S. and Melton, D. A. (1990). A two-step model for the
localization of maternal mRNA in Xenopus oocytes: involvement of
microtubules and microfilaments in the translocation and anchoring of Vg1
mRNA. Development 108, 289-298.
Yoon, Y. J. and Mowry, K. L. (2004). Xenopus Satufen is a component of a
ribonucleoprotein complex containing Vg1 RNA and kinesin. Development
131, 3035-3045.
Yoshida, S., Marikawa, Y. and Satoh, N. (1996). Posterior end mark, a novel
maternal gene encoding a localized factor in the ascidian embryo.
Development 122, 2005-2012.
Zhang, J. and King, M. L. (1996). Xenopus VegT RNA is localized to the
vegetal cortex during oogenesis and encodes a novel T-box transcription
factor involved in mesoderm patterning. Development 122, 4119-4129.
Zhang, J., Houston, D. W., King, M. L., Payne, C., Wylie, C. and Heasman,
L. (1998). The role of maternal VegT in establishing the primary germ layers
in Xenopus embryos. Cell 94, 515-524.
Zhao, W., Jiang, C., Kroll, T. T. and Huber, P. W. (2001). A proline-rich
protein binds to the localization element of Xenopus Vg1 mRNA and to
ligand involved in actin polymerization. EMBO J. 20, 2315-2325.
Zhou, Y. and King, M. L. (1996). RNA transport to the vegetal cortex of
Xenopus oocytes. Dev. Biol. 179, 173-183.
Zhou, Y. and King, M. L. (2004). Sending RNAs into the future: RNA
localization and germ cell fate. IUBMB Life 56, 19-27.
Journal of Cell Science 118 (2)
Journal of Cell Science
... Several mechanistic studies lead to the description of two molecular pathways for RNA localization to the vegetal pole: an early pathway that is active in stages I and II, and a late pathway, active in stages III to IV. Nonetheless, some publications suggest a functional continuum of these pathways (Claußen, Horvay and Pieler, 2004;Choo et al., 2005), leading to the state of sometimes contradictory and 'confusing' reports (Kloc and Etkin, 2005). The transport of RNA to the animal pole is largely not well understood (Sindelka et al., 2018). ...
... First described in the oocytes of spiders in 1845, it is present in all examined vertebrates, including humans (De Smedt, Szöllösi and Kloc, 2000). The Balbiani body is not present in mice, the predominant model for mammalian developmental studies, which likely contributes to the fact that its role is less well understood (Kloc and Etkin, 2005). ...
... RNA is then transported in larger complexes together with RNA binding proteins, motor proteins and ER to the vegetal cortex (Figure 7b, bottom) (Deshler, Highett and Schnapp, 1997;Kloc and Etkin, 1998). While the plus-end directed motor proteins kinesin I (Yoon and Mowry, 2004) and kinesin II (Betley et al., 2004) have been shown to be involved in microtubule-dependent, vegetally directed transport, it is 'paradox and confusing' (Kloc and Etkin, 2005) that around 96% of the microtubules have their minus ends directed towards the cortex at these stages. This stands in contrast to somatic eukaryotic cells, and possible explanations include the suggestion that RNA-binding proteins (RBPs) can change the directionality of the RNA transport (Kloc and Etkin, 2005). ...
Thesis
Die Lokalisierung von mRNA ist ein wichtiger regulativer Mechanismus in polarisierten Zellen und in frühen Embryonalstadien. Dort sind räumliche Muster maternaler mRNA für die korrekte Entwicklung der Körperachsen und die Spezifizierung der Keimzellen verantwortlich. Systematische Analysen dieser Prozesse wurden jedoch bisher limitiert durch einen Mangel an räumlicher und zeitlicher Auflösung von Einzelzell- Sequenzierungsdaten. Wir analysierten die Dynamik des räumlichen und zeitlichen Transkriptoms während frühen Embryonalstadien von Zebrafischen. Wir verbesserten Empfindlichkeit und Auflösung von tomo-seq und erfassten damit systematisch räumlich aufgelöste Transkriptome entlang der animal-vegetalen-Achse Embryonen im Einzell-Stadium und fanden 97 vegetal lokalisierte Gene. Außerdem etablierten wir eine Hochdurchsatz kompatible Variante der RNA-Markierungsmethode scSLAM-seq. Wir wendeten diese in Embryonen während der Gastrulation. Von den vegetal lokalisierten Genen waren 22 angereichert in Keimzellen, was eine funktionelle Rolle bei der Spezifizierung von Keimzellen nahelegt. Mit tomo-seq untersuchten wir die evolutionäre Konservierung der RNA-Lokalisierung zwischen Zebrafischen und gereiften Oozyten zweier Xenopus-Arten. Wir verglichen die lokalisierten Gene, suchten nach konservierten 3'UTR-Motiven, und fanden zum Teil überlappende Motive, was auf eine mögliche mechanistische Konservierung der Lokalisierungsmechanismen hinweist. Wir untersuchten auch RNA-Editierung von Adenin zu Inosin während der Embryonalentwicklung und in den Organen erwachsener Fische. In im Gehirn exprimierten Transkripten fanden wir 117 Editierstellen, die hauptsächlich für Ionentransporter kodieren und zum Teil zum Menschen konserviert sind. Die höchsten Editierraten konnten wir in Eierstöcken, Hoden und frühen Embryonen nachweisen, was auf eine mögliche Rolle bei der Regulierung der RNA-Stabilität hindeutet.
... Similarities and differences between the eggs and embryos are propagated in the particular distribution of biomolecules regulating development (reviewed by King et al., 2005; reviewed by Kloc and Etkin, 2005). Proteins, RNAs, cellular structures and organelles are distributed unevenly within the egg (reviewed by Marlow, 2010). ...
... C) Genomic and egg developmental properties for each model. mesodermal and endodermal determinants, and are also crucial for left-right axis formation in the embryo (Hyatt and Yost, 1998;Joseph and Melton, 1998;reviewed by King et al., 2005; reviewed by Kloc and Etkin, 2005;Ku and Melton, 1993;Thomsen and Melton, 1993;Zhang et al., 1998). Saito et al. (2014) demonstrated that in A. ruthenus, PGCs are formed in the vegetal pole as well. ...
Article
Asymmetrical localization of biomolecules inside the egg, results in uneven cell division and establishment of many biological processes, cell types and the body plan. However, our knowledge about evolutionary conservation of localized transcripts is still limited to a few models. Our goal was to compare localization profiles along the animal-vegetal axis of mature eggs from four vertebrate models, two amphibians (Xenopus laevis, Ambystoma mexicanum) and two fishes (Acipenser ruthenus, Danio rerio) using the spatial expression method called TOMO-Seq. We revealed that RNAs of many known important transcripts such as germ layer determinants, germ plasm factors and members of key signalling pathways, are localized in completely different profiles among the models. It was also observed that there was a poor correlation between the vegetally localized transcripts but a relatively good correlation between the animally localized transcripts. These findings indicate that the regulation of embryonic development within the animal kingdom is highly diverse and cannot be deduced based on a single model.
... The Bb varies in morphology and composition among species, consisting mainly of mitochondria, nuage and proteins which are precursors of germplasm. It also contains localized maternal messenger ribonucleic acids (mRNAs) and, in some cases, lipid droplets and lysosome-like organelles, endoplasmic reticulum, Golgi bodies and membranous vesicles Kloc et al., 2001Kloc et al., , 2004Kloc et al., , 2007Kloc et al., , 2008; Kloc & Etkin, 2005;Marlow & Mullins, 2008; which is critical for embryonic development because it establishes the embryonic axis (Colozza & De Robertis, 2014;Elkouby et al., 2016;Langdon & Mullins, 2011;Nojima et al., 2010). It transports the germplasm determinants to the vegetal pole (Marlow & Mullins, 2008;Saito et al., 2014;Škugor et al., 2016) and contains gene products required for RNA processing and storage, as well as transposon suppression (Marlow & Mullins, 2008). ...
... The Bb appears as a distinct structure in primary growth oocytes of most teleosts (e.g., Beams & Kessel, 1973;Domínguez-Castanedo & Uribe, 2019;Elkouby, 2017;Elkouby et al., 2016;Fusco et al., 2000;Grier, 2012;Grier et al., 2018;Lyman-Gingerich & Pelegri, 2007;Marlow & Mullins, 2008;_ Zelazowska & Halajian, 2019;Zhang et al., 2008), similarly to Xenopus laevis (Kloc et al., 2001Kloc & Etkin, 2005). In zebrafish, the Bb forms as a spherical structure in a nuclear cleft on the side of the centrosome, where a cluster of chromosomal telomeres forms during zygotene at prophase I of meiosis, gradually diverging from the perinuclear region to the periphery at the vegetal cortex, where it disassembles (Elkouby & Mullins, 2017). ...
Article
Full-text available
The Balbiani body (Bb) was examined in primary growth phase oocytes for the first time in two clupeoid fish species, the Mediterranean sardine, Sardina pilchardus, and the European anchovy, Engraulis encrasicolus, which belong to different families, Clupeidae and Engraulidae, respectively. Cytoplasmic morphological changes of early secondary growth oocytes were also investigated using confocal laser scanning microscopy, light and transmission electron microscopy. The ultrastructural observations showed that the two species develop a distinct spherical Bb. However, differences in the cytoplasm, mainly in the perinuclear area, were observed. Briefly, in sardine the Bb coexists with a thick perinuclear ring containing mitochondria, nuage, endoplasmic reticulum and small vesicles, while in anchovy this perinuclear ring is thinner, consisting of complexes of nuage and mitochondria. After the disassembly of the Bb, a prominent cytoplasmic zonation develops in the secondary growth oocytes of sardine and anchovy, although with different organelle distribution between the two species. Sardine oocytes exhibit a thick zone of endoplasmic reticulum around the nucleus, whereas in those of anchovy, a thick mitochondria‐rich ring surrounding the nucleus was observed. The cytoplasmic characteristics, such as the perinuclear ring in primary oocytes in sardine and the mitochondria‐rich ring of early secondary oocytes in anchovy, are also discernible in histological sections by standard procedures and could thus be used as indicators of maturity or imminent spawning period in routine light microscopy observations, providing a valuable tool for applied fisheries biology.
... rerio, and other species of vertebrates and invertebrates the Balbiani body is involved in the establishment of the animal-vegetal axis and formation of the germplasm (germ cells determinants; Aguero et al., 2017;Cox & Spradling, 2003;Elkouby, 2017;Elkouby et al., 2016;Escobar-Aguirre et al., 2017;Ikami et al., 2017;Kloc & Etkin, 2005;Kloc et al., 2001Kloc et al., , 2008Kloc et al., , 2014. It forms a size exclusion barrier for molecules over a certain size that cannot penetrate its ruthenus represent a stage little more advanced than those of albinos described above. ...
Article
Ovarian follicles of sterlets ( Acipenser ruthenus ) are composed of a single oocyte surrounded by follicular cells (FCs), basal lamina, and thecal cells. Previtellogenic oocytes are polarized. Homogeneous ooplasm (contains ribosomes) and granular ooplasm (comprises nuage aggregations of nuclear origin, rough endoplasmic reticulum (RER), Golgi complexes, ribosomes, and mitochondria) are distinguished. Granular ooplasm is initially located near the nucleus, contacts the plasma membrane of the oocyte (oolemma) and forms a thin layer underneath its entire perimeter. Next, a ring that surrounds the nucleus is formed and sends strands directed toward the oolemma. The lipid body composed of lipid droplets forms adjacent to this ring. Later, the granular ooplasm and strands enlarge toward the oolemma, lipid body disperses, and homogeneous ooplasm is no longer present. A thin cortical ooplasm is formed underneath the oolemma and does not contain any organelles. The oocyte nucleus moves to the center. The nucleoplasm contains lampbrush chromosomes, nuclear bodies, and multiple nucleoli. Early vitellogenic oocytes are polarized, too. Three regions in the ooplasm are distinguished: the perinuclear (contains lipid droplets near the nuclear envelope), the endoplasm (contains yolk platelets and lipid droplets), and the periplasm (contains yolk spheres, pigment granules, and microtubules). In all these regions the RER, Golgi complexes, nuage, and mitochondria are present. Micropinocytotic vesicles, Golgi vesicles and precursors of the internal layer of the egg envelope are in the cortical ooplasm. Some FCs delaminate from the follicular epithelium, degenerate and vesicles are released into the perioocytic space. They may contain precursors of egg envelope and may be involved in “cell–cell” communication. The egg envelope ( zona radiata, zona pellucida ) is made up of three layers: the vitelline envelope (inner layer), the middle layer, and the outer layer. In its deposition, both the oocyte and FCs are engaged.
... Previous research that concerned model species, zebrafish Danio rerio and African clawed frog Xenopus laevis, sturgeons, and invertebrates, revealed that formation of early asymmetry of the ooplasm is of vital importance for establishment of the animal-vegetal axis and formation of germplasm, which determines germline cells in the embryos (reviewed in Kloc and Etkin, 2005;Aguero et al., 2017;Escobar-Aguirre et al., 2017;Zelazowska et al., 2007;Saito et al., 2014;Kloc et al., 2014). This is manifested by several events that take place during primary growth. ...
Article
The ovaries of Sander lucioperca (Actinopterygii, Perciformes) are made up of the germinal epithelium and ovarian follicles, in which primary oocytes grow. Each follicle is composed of an oocyte surrounded by flattened follicular cells, the basal lamina, and thecal cells. The early stages of oocyte development (primary growth = previtellogenesis) are not fully explained in this species. The results of research with the use of stereoscopic, light, fluorescence, and transmission electron microscopes on ovarian follicles containing developing primary oocytes of S. lucioperca are presented. The polarization and ultrastructure of oocytes are described and discussed. The deposition of egg envelopes during the primary growth and the ultrastructure of the eggshell in maturing follicles of S. lucioperca are also presented. Nuclei in primary oocytes comprise lampbrush chromosomes, nuclear bodies, and nucleoli. Numerous additional nucleoli arise in the nucleoplasm during primary growth and locate close to the nuclear envelope. The Balbiani body in the cytoplasm of oocytes (ooplasm) is composed of nuage aggregations of nuclear origin and mitochondria, endoplasmic reticulum (ER), and Golgi apparatus. The presence of the Balbiani body was reported in oocytes of numerous species of Actinopterygii; however, its ultrastructure was investigated in a limited number of species. In primary oocytes of S. lucioperca, the Balbiani body is initially located in the perinuclear ooplasm on one side of the nucleus. Next, it surrounds the nucleus, expands toward the plasma membrane of oocytes (oolemma), and becomes fragmented. Within the Balbiani body, the granular nuage condenses in the form of threads, locates near the oolemma, at the vegetal oocyte pole, and then dissolves. Mitochondria and cisternae of the rough endoplasmic reticulum (RER) are present between the threads. During primary growth micropylar cells differentiate in the follicular epithelium. They contain cisternae and vesicles of the RER and Golgi apparatus as well as numerous dense vesicles suggesting high synthetic and secretory activity. During the final step of primary growth several follicular cells delaminate from the follicular epithelium, migrate toward the oocyte and submerge in the most external egg envelope. In the ooplasm, three regions are distinguished: perinuclear, endoplasm, and periplasm. Cortical alveoli arise in the perinuclear ooplasm and in the endoplasm as a result of the fusion of RER vesicles with Golgi ones. They are evenly distributed. Lamellar bodies in the periplasm store the plasma membrane and release it into a space between the follicular cells and the oocyte. The developing eggshell in this space is made up of two egg envelopes (the internal one and the external) that are pierced by canals formed around the microvilli of oocytes and the processes of follicular cells. In the deposition of egg envelopes the oocyte itself and follicular cells are engaged. In maturing ovarian follicles the eggshell is solid and the internal egg envelope is covered with protuberances.
... asymmetry of the previtellogenic oocytes. In all described species and also in the three osteoglossiforms studied here, the Balbiani body was always enriched in the germ plasm, mitochondria, ER and Golgi apparatus (Kloc & Etkin, 2005;_ Zelazowska et al., 2007). However, the formation of some structures varied among studied species (Table 1, Figures 8-11). ...
Article
During the early stages of oogenesis, the Balbiani body is formed in the primary oocytes. It consists of the Golgi apparatus, endoplasmic reticulum (ER) and numerous mitochondria aggregated with germ plasm, but its form may differ among animals. Hypothetically, during oogenesis oocytes become adapted to future development in two different environments depending on internal or external fertilization. We aimed to investigate, using light and transmission electron microscopy, the development of the Balbiani body during oogenesis in representatives of Osteoglossiformes, one of the most basal Teleostei groups. We analyzed the structure of oogonia and primary oocytes in the internally fertilizing butterflyfish Pantodon buchholzi and the externally fertilizing Osteoglossum bicirrhosum and Arapaima gigas to compare formation of the Balbiani body in relation to modes of fertilization. We demonstrated that the presence of the germ plasm as well as the fusion and fission of mitochondria are the conserved features of the Bb. However, each species exhibited also some peculiar features, including the presence of three types of ooplasm with different electron density and mitochondria‐associated membranes in P. buchholzi; annulate lamellae, complexes of the Golgi apparatus, ER network, and lysosome‐like bodies in O. bicirrhosum; as well as karmellae and whorls formed by the lamellae of the ER in A. gigas. Moreover, the form of the germ plasm observed in close contact with mitochondria differed between osteoglossiforms, with a “net‐like” structure in P. buchholzi, the presence of numerous strings in O. bicirrhosum, and irregular accumulations in A. gigas. These unique features indicate that the extreme diversity of gamete structure observed so far only in the spermatozoa of osteoglossiforms is also characteristic for oocyte development in these basal teleosts. Possible reason of this variability is a period of about 150 million years of independent evolution of the lineages.
... Across many biological contexts, cells must be able to sense external spatial cues and generate asymmetric distributions of their internal components. Anisotropic patterns of protein/RNA localization play crucial roles during embryo development (Kloc and Etkin, 2005;Sailer et al., 2015), and motile cells can migrate by generating persistent internal asymmetries even in a uniform environment (Prentice-Mott et al., 2016). It is often assumed that both the establishment and maintenance of these persistent spatial patterns require complex genetic and/or biochemical networks, such as Turing-like mechanisms that combine short-range positive feedback with longrange negative feedback (Gierer and Meinhardt, 1972;Turing, 1990) or stochastic processes that rely on depleting a limiting pool of proteins that participate in an auto-regulatory positive feedback loop (Altschuler et al., 2008). ...
Preprint
Full-text available
Protein/RNA clusters arise frequently in spatially-regulated biological processes, from the asymmetric distribution of P granules and PAR proteins in developing embryos to localized receptor oligomers in migratory cells. This co-occurrence suggests that protein clusters might possess intrinsic properties that make them a useful substrate for spatial regulation. Here, we demonstrate that protein droplets show a robust form of spatial memory, maintaining the spatial pattern of an inhibitor of droplet formation long after it has been removed. Despite this persistence, droplets can be highly dynamic, continuously exchanging monomers with the diffuse phase. We investigate the principles of biophysical spatial memory in three contexts: a computational model of phase separation; a novel optogenetic system where light can drive rapid, localized dissociation of liquid-like protein droplets; and membrane-localized signal transduction from clusters of receptor tyrosine kinases. Our results suggest that the persistent polarization underlying many cellular and developmental processes could arise through a simple biophysical process, without any additional requirement for biochemical positive and negative feedback loops. Highlights We introduce PixELLs, an optogenetic system for protein droplet disassembly. Modeling and experiments demonstrate long-term memory of local droplet dissociation. Droplets ‘remember’ spatial stimuli in nuclei, the cytosol and on cell membranes. FGFR-optoDroplets convert transient local inputs to persistent cytoskeletal responses.
Preprint
Full-text available
Axes of polarity (and primary morphogenetic gradients) are established in the oocyte - early embryo through active transport and localization of maternal factors. It is the oocyte - syncytial embryo of Drosophila (D. melanogaster) that is a model object for studying the molecular machinery of such transport systems. The attention of researchers is focused on the processes of formation, maintenance, and functioning of active transport systems of maternal mRNAs and proteins that are key for early Drosophila embryogenesis. Here we develop an approach for agent-based 3D modeling of the key components of transport by molecular motors (by elements of the cytoskeleton) of the Drosophila oocyte-syncytial embryo. The models were developed using Skeledyne software developed by Odell and Foe [Odell and Foe, 2008]. We start with the results of modeling transport along oriented microtubule (MT) bundles in the oocyte. This is a model of transport systems in the Drosophila oocyte, where three maternal mRNAs (bicoid (bcd), oskar, and gurken) that are key to embryonic polarity are transported along their oriented MT bundles. Then we consider models of oriented MT networks in the volume of a cell (oocyte) generated by a single microtubule organization center (or a pair of the centers). This model reproduces the formation of bcd mRNA intrusions deep into the cytoplasm in the head half of the early syncytial embryo. Finally, we consider models for the active transport of bcd mRNA in a syncytial embryo along a randomized network of many short MT strands. In conclusion, we consider the prospects for the implementation of cytoplasmic fountain flows in the active transport model.
Article
Full-text available
Ribonucleoprotein condensates can exhibit diverse physical states in vitro and in vivo. Despite considerable progress, the relevance of condensate physical states for in vivo biological function remains limited. Here, we investigated the physical properties of processing bodies (P bodies) and their impact on mRNA storage in mature Drosophila oocytes. We show that the conserved DEAD-box RNA helicase Me31B forms viscous P body condensates, which adopt an arrested physical state. We demonstrate that structurally distinct proteins and protein-protein interactions, together with RNA, regulate the physical properties of P bodies. Using live imaging and in situ hybridization, we show that the arrested state and integrity of P bodies support the storage of bicoid (bcd) mRNA and that egg activation modulates P body properties, leading to the release of bcd for translation in the early embryo. Together, this work provides an example of how physical states of condensates regulate cellular function in development.
Article
Full-text available
During meiotic maturation, the cortical cytokeratin filament system of the Xenopus oocyte disappears (Klymkowsky, M. W., and L. A. Maynell. 1989. Dev. Biol. 134:479). Here we demonstrate that this disappearance results from the severing of cytokeratin filaments into a heterogenous population of oligomers, with S- values ranging from 12S and greater. Cytokeratin filament severing correlates with the hyperphosphorylation of the type II cytokeratin of the oocyte. Both the severing of cytokeratin filaments and cytokeratin hyperphosphorylation are reversed by treatment with cycloheximide. These data suggest that fragmentation of cytokeratin filaments is controlled, at least in part, by the phosphorylation of the type II cytokeratin, and that the cytokeratin kinase activity responsible is biosynthetically labile. Cytokeratin filaments have been suggested to anchor the maternal mRNA Vg1 to the vegetal cortex of the oocyte (Pondel, M., and M. L. King. 1988. Proc. Natl. Acad. Sci. USA. 85:7216). By injecting fractions containing active maturation promoting factor or a purified, mutant cyclin protein, we find that the bulk of the Vg1 mRNA in the oocyte can be solubilized under conditions that block the fragmentation of cytokeratin filaments, and that the fragmentation of cytokeratin filaments itself leads to the solubilization of only a minor fraction of the Vg1 mRNA. Thus, at best, cytokeratin filaments directly anchor only a minor fraction of the Vg1 mRNA in the oocyte. Moreover, factors distinct from maturation promoting factor appear to be required for the complete solubilization of Vg1 mRNA during oocyte maturation.
Article
Full-text available
Ezrin, Radixin and Moesin (ERM) proteins are thought to constitute a bridge between the actin cytoskeleton and the plasma membrane (PM). Here we report a genetic analysis of Dmoesin, the sole member of the ERM family in Drosophila. We show that Dmoesin is required during oogenesis for anchoring microfilaments to the oocyte cortex. Alteration of the actin cytoskeleton resulting from Dmoesin mutations impairs the localization of maternal determinants, thus disrupting antero–posterior polarity. This study also demonstrates the requirement of Dmoesin for the specific organization of cortical microfilaments in nurse cells and, consequently, mutations in Dmoesin produce severe defects in cell shape.
Article
Full-text available
Despite lacking centrosomes, stage VI Xenopus oocytes contain extensive networks of cytoplasmic microtubules (MTs). To gain additional insight into the factors regulating MT organization during oogenesis, we have used electron microscopy and “hook decoration” to examine the distribution and orientation of MTs in Xenopus oocytes. A limited survey of two “undecorated” stage VI oocytes revealed 218 MTs in images covering ∼2,500 μm2, and indicated that the MT number density of the animal cytoplasm was greater than that of the vegetal cytoplasm. Examination of five “decorated” stage VI oocytes (three animal and five vegetal hemispheres) revealed 653 MTs. Of these, 76% could be scored as having exclusively counterclockwise (CCW) or clockwise (CW) hooks. In the animal hemispheres, 93% of the scored MTs exhibited CCW hooks when viewed from the direction of the cortex, indicating that most MTs were oriented with their minus-ends out. MT orientation appeared relatively uniform throughout the animal cytoplasm: more than 90% of the scored MTs in the cortical (90%), subcortical (96%), or perinuclear (98%) cytoplasm were oriented with their minus-ends out. In the vegetal hemispheres, ∼80% of the scored MTs exhibited CCW hooks, and thus were oriented with their minus-ends out; 96% of the scored MTs in stage III oocytes were oriented minus-end out. These observations support a model in which the cortex plays a significant role in MT nucleation and organization in Xenopus oocytes, and have significant implications for the MT-dependent transport and localization of cytoplasmic organelles and RNAs during oogenesis. Cell Motil. Cytoskeleton 44:34–43, 1999. © 1999 Wiley-Liss, Inc.
Article
Figure 1. Expression and Localization of Heterotrimeric Kinesin II mRNA and Protein during Oogenesis
Article
In many organisms, pattern formation in the embryo develops from the polarized distributions of messenger RNAs (mRNAs) in the egg. InXenopus, the mRNA encoding Vg1, a growth factor involved in mesoderm induction, is localized to the vegetal cortex of oocytes. A protein named Vera was shown to be involved in Vg1 mRNA localization. Vera cofractionates with endoplasmic reticulum (ER) membranes, and endogenous Vg1 mRNA is associated with a subcompartment of the ER. Vera may promote mRNA localization in Xenopus oocytes by mediating an interaction between the Vg1 3′ untranslated region and the ER subcompartment.
Article
In eukaryotic cells, many organelles are transported bidirectionally along microtubules by kinesin and dynein. These opposite-polarity motors appear to be coordinated to avoid interfering with each other's function. New work has provided the first molecular insight into how such coordination might occur.
Article
bicoid (bcd) RNA localization requires the activity of exuperantia and swallow at sequential steps of oogenesis and is microtubule dependent. In a genetic screen, we identified two novel genes essential for bcd RNA localization. They encode maternal γ-Tubulin37C (γTub37C) and γ-tubulin ring complex protein 75 (Dgrip75), both of which are γ-tubulin ring complex components. Mutations in these genes specifically affect bcd RNA localization, whereas other microtubule-dependent processes during oogenesis are not impaired. This provides direct evidence that a subset of microtubules organized by the γ-tubulin ring complex is essential for localization of bcd RNA. At stage 10b, we find γTub37C and Dgrip75 anteriorly concentrated and propose the formation of a microtubule-organizing center at the anterior pole of the oocyte.
Article
The proteins Mago and Y14 are evolutionarily conserved binding partners. Y14 is a component of the exon-exon junction complex (EJC), deposited by the spliceosome upstream of messenger RNA (mRNA) exon-exon junctions. The EJC is implicated in post-splicing events such as mRNA nuclear export and nonsense-mediated mRNA decay. Drosophila Mago is essential for the localization of oskar mRNA to the posterior pole of the oocyte, but the functional role of Mago in other species is unknown. We show that Mago is a bona fide component of the EJC. Like Y14, Mago escorts spliced mRNAs to the cytoplasm, providing a direct functional link between splicing and the downstream process of mRNA localization. Mago/Y14 heterodimers are essential in cultured Drosophila cells. Taken together, these results suggest that, in addition to its specialized function in mRNA localization, Mago plays an essential role in other steps of mRNA metabolism.