Download Determinants of Drosophila zw10 protein localization and function

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts

Cell encapsulation wikipedia , lookup

Signal transduction wikipedia , lookup

Magnesium transporter wikipedia , lookup

Cell nucleus wikipedia , lookup

Protein (nutrient) wikipedia , lookup

Hedgehog signaling pathway wikipedia , lookup

Protein phosphorylation wikipedia , lookup

Protein moonlighting wikipedia , lookup

Cell growth wikipedia , lookup

Cell cycle wikipedia , lookup

Nuclear magnetic resonance spectroscopy of proteins wikipedia , lookup

Meiosis wikipedia , lookup

List of types of proteins wikipedia , lookup

Amitosis wikipedia , lookup

Biochemical switches in the cell cycle wikipedia , lookup

Cytokinesis wikipedia , lookup

Kinetochore wikipedia , lookup

Mitosis wikipedia , lookup

Spindle checkpoint wikipedia , lookup

Transcript
785
Journal of Cell Science 107, 785-798 (1994)
Printed in Great Britain © The Company of Biologists Limited 1994
Determinants of Drosophila zw10 protein localization and function
Byron C. Williams and Michael L. Goldberg*
Section of Genetics and Development, Biotechnology Building, Cornell University, Ithaca, NY 14853-2703, USA
*Author for correspondence
SUMMARY
We have examined several issues concerning how the
Drosophila l(1)zw10 gene product functions to ensure
proper chromosome segregation. (a) We have found that in
zw10 mutant embryos and larval neuroblasts, absence of
the zw10 protein has no obvious effect on either the congression of chromosomes to the metaphase plate or the
morphology of the metaphase spindle, although many aberrations are observed subsequently in anaphase. This
suggests that activity of the zw10 protein becomes essential
at anaphase onset, a time at which the zw10 protein is redistributed to the kinetochore region of the chromosomes. (b)
The zw10 protein appears to bind to kinetochores in mitotically arrested cells, eventually accumulating to high levels
within the chromosome mass. Our results imply that zw10
may act as part of a novel feedback pathway that normally
renders sister chromatid separation dependent upon
spindle integrity. (c) The localization of zw10 protein is
altered by two mitotic mutations, rough deal and abnormal
anaphase resolution, that specifically disrupt anaphase.
These findings indicate that the zw10 protein functions as
part of a multicomponent mechanism ensuring proper
chromosome segregation at the beginning of anaphase.
INTRODUCTION
The relationship, if any, of this PSCS phenomenon to the generation of aneuploidy is unclear.
The zw10 protein is found at different locations at various
times during the Drosophila embryonic cell cycle. It is
excluded from the nuclei during interphase. The zw10 product
migrates into the nuclear volume at prometaphase, eventually
becoming part of a filamentous structure associated with a
portion of the spindle and extending through the metaphase
plate. Because the zw10 protein colocalizes with only a subset
of the tubulin found in the spindle, we have suggested that
zw10 is associated either with one class of microtubules (most
likely the kinetochore microtubules) or with an unknown
microtubule-independent structure within the spindle. Within
a few seconds of anaphase onset, zw10 protein relocalizes to
an area at or near the kinetochores of the separating sister chromatids and remains there until telophase (Williams et al.,
1992).
These observations together suggest that the movement of
zw10 protein to the vicinity of the kinetochores at the
metaphase-anaphase transition is important for subsequent
proper chromatid separation. However, these initial findings do
not exclude the possibility that zw10 function is important at
earlier times in mitosis. The localization of zw10 protein to
different structures through mitosis could indicate that it has
multiple sites of function. For example, zw10 protein could be
required for the bipolar attachment of chromosomes to the
spindle, for spindle assembly or integrity, for prometaphase
chromosome movements, or for the formation of the
metaphase plate. In addition, we have been unable to incorporate the phenomenon of PSCS into a consistent model of zw10
The end result of mitosis is the equal apportionment of sister
chromatids to two daughter cells. To avoid the deleterious consequences of aneuploidy, the movements of chromosomes and
their interactions with the mitotic apparatus must be coordinated precisely in space and in time. One strategy to identify
molecules required for proper mitotic chromosome segregation
and to help define their associated biochemical activities is the
study of mutations that result in altered karyotypes. A variety
of methods for the isolation of such mitotic mutations in
Drosophila have been developed (Gatti and Goldberg, 1991).
We have previously shown that mutations abolishing the
function of one such gene, l(1)zw10 (hereafter abbreviated
zw10), result in high levels of aneuploidy in larval neuroblast
cells (Williams et al., 1992). Aberrations occurring during
anaphase appear to be the immediate cause of chromosomal
mis-segregation: many mutant anaphase figures display
lagging chromatids that remain near the metaphase plate at a
time when the majority of chromatids have arrived at the poles.
In many cases, it appears that the separation of sister chromatids is delayed, resulting in the migration of both sister chromatids to the same pole. At times we observe anaphase figures
with more severe defects, such as the appearance of grossly
unequal chromosome complements at the two poles. Another
facet of the zw10 mutant phenotype is ‘precocious sister
chromatid separation’ (PSCS) triggered by mitotic arrest: the
connection between sister chromatids in severed in mutant, but
not wild-type larval neuroblast cells treated with the mitotic
poison colchicine (Smith et al., 1985; Williams et al., 1992).
Key words: kinetochore, centromere, mitosis, chromosome
segregation, microtubule poison, anaphase onset
786
B. C. Williams and M. L. Goldberg
Fig. 1. Non-specific spindle
abnormalities in zw10 mutant
germline clone embryos.
Embryos were stained with
Hoechst 33258 (A,C) and antiβ-tubulin antibodies (B,D; see
Materials and Methods).
(A,B) Wild-type syncytial
blastoderm embryo, which
contains a field of evenly
spaced metaphase nuclei (A). In
each mitotic figure, the spindle
(outline arrow, B) is organized
around a metaphase plate
(outline arrow, A) and has been
nucleated from centrosomes at
opposite poles as revealed by
the positions of the asters
(arrowheads, B). (C,D) In zw10
germline clone syncytial
blastoderm embryos, nuclei
commonly are unevenly spaced.
Defects in nuclear division in
mutant embryos can lead to free
asters (arrowheads in D; see
text) unassociated with nuclei
(arrowheads in C). These free
asters may in turn result in the formation of tripolar spindles when they become attached to nearby nuclei (outline arrow, C and D). The uneven
spacing of nuclei can lead to spindle fusions, since a single pole is shared by two spindles (long arrow, C and D). Bars, 10 µm.
action, because we did not know the fate of zw10 protein in
mitotically arrested cells.
In this paper, we describe more detailed analyses, both of
the mutant phenotype and of the cell cycle-dependent intracellular distribution of the zw10 protein, which provide clues
to the role played by zw10 in mitotic chromosome distribution.
We show that in zw10 mutants the first obvious mitotic defect
is seen in anaphase chromatid movement. Prometaphase centrosome movement, metaphase spindle structure, and
metaphase chromosome alignment all appear to be unaffected,
suggesting that zw10 function is not required prior to anaphase
onset.
We have furthermore made two observations of potential
relevance to the PSCS phenomenon described above. First, we
have found that the zw10 protein localizes to the region of the
kinetochores in cells mitotically arrested by either colchicine
or taxol treatment. Second, we demonstrate that PSCS in mitotically arrested zw10 mutant cells occurs in the absence of
cyclin B degradation. This supports the recent finding by
Holloway et al. (1993) that sister chromatid separation can
occur without MPF destruction. We believe that this PSCS
phenomenon indicates that the zw10 protein participates as part
of a novel checkpoint that keeps sister chromatids together
until the metaphase spindle is properly established.
Finally, we have studied zw10 localization in cells homozygous for several other Drosophila mitotic mutations that
disrupt accurate chromosome segregation (reviewed by Gatti
and Goldberg, 1991). Of these, mutations in abnormal
anaphase resolution (aar; Gomes et al., 1993; Mayer-Jaekel et
al., 1993) and rough deal (rod; Karess and Glover, 1989) significantly alter the localization of the zw10 protein. Both aar
and rod mutations result in aberrant anaphases similar to those
observed in zw10 mutants; it has been suggested that they
affect the behavior of chromatids at anaphase onset (Karess
and Glover, 1989; Mayer-Jaekel et al., 1993). Altogether, these
results suggest that the zw10 protein functions at the centromere/kinetochore as part of a signalling pathway that coordinates sister chromatid separation or movement at the
metaphase-anaphase transition.
MATERIALS AND METHODS
Examination of zw10 germline clone embryos
Homozygous zw10 germline clones were generated by the FLP
recombinase system (Chou and Perrimon, 1992). Three null alleles of
zw10 were used (zw10S1, zw10S2M and zw1065i20), all of which gave
the same germline clone phenotype. Briefly, the FRT site (the site of
FLP-catalyzed recombination) was crossed onto the zw10 chromosome by mating y,w,FRT(w+)/Y males with zw10/FM7a virgin
females. The female progeny of genotype y,w,FRT(w+)/zw10 were
then crossed to y,w,FRT(w+)/Y males. Female progeny from this
second cross included desired recombinants with the genotype
zw10,FRT(w+)/y,w,FRT(w+). These could be identified by individually mating female progeny with wild-type eye color to FM7a/Y
males. If crossing-over occurs so that zw10 is on the same chromosome as the FRT site, this last cross will yield no white-eyed males.
Furthermore, a stock can be established with the
zw10,FRT(w+)/FM7a females by a subsequent cross with FM7a/Y
males.
Once such lines were established, zw10,FRT/FM7a virgin females
were mated with ovoD1,FRT;FLP/FLP males in vials and their
progeny heat-shocked at 37°C in a water bath for two hours at the
third instar larval and pupal stages. The heat shock at 37°C activates
expression of the FLP recombinase under the control of the hsp70
promoter, which acts on the paired FRT sites to catalyze mitotic
Drosophila zw10 protein
787
Fig. 2. Mitotic abnormalities in zw10 embryos. A
syncytial blastoderm wild-type embryo (A) and
an embryo from a homozygous zw10S1 germline
clone (B) were fixed and stained with propidium
iodide for confocal microscopy (see Materials
and Methods). (A) Wild-type embryonic nuclei
present in a mitotic wave (Foe and Alberts,
1983). Nuclei can be seen in mid-anaphase
(outline arrow, left), late anaphase (long arrow,
middle), and telophase (arrowheads, right). Note
the equal and complete separation of chromatids.
(B) Aberrant mitoses in a typical zw10 embryo,
showing defects such as an unequal anaphase
(filled arrow, bottom left), and lagging chromatids or chromatin bridges at anaphase and telophase (outline arrows). Anti-tubulin staining (not
shown) is consistent with the staging of these nuclei at anaphase and telophase. Bars, 10 µm.
Fig. 3. Localization of zw10
protein in wild-type brain
neuroblasts. Untreated brains
were fixed and stained as
described (see Materials and
Methods) to visualize
chromosomes (blue) and zw10
antigen (red) in single optical
sections acquired by confocal
microscopy. (A) Prometaphase
(outline arrow, left) shows zw10
(red) localized near the
chromosomes (blue) and
beginning to form the filaments
seen at metaphase (filled arrow,
right) analogous to those seen in
the embryo (Williams et al.,
1992). (B,C) Metaphase plates
viewed head-on showing
chromosomes arranged in a ring
with their centromeres oriented
towards the center. zw10 protein
(red) occupies the central region
corresponding to the location of
the kinetochores. The zw10
staining pattern contrasts with the
overall microtubule distribution,
which is broader and
encompasses the entire
chromosome arrangement (not shown). (D) Early anaphase showing zw10 antigen at or near kinetochores of the separated chromatids (arrows).
(E) Mid-anaphase and (F) late anaphase figures with zw10 remaining at the kinetochore regions (arrows). Bars, 5 µM.
crossing-over (Chou and Perrimon, 1992). Any mitotic cross-over
products that can produce functional ovaries due to absence of ovoD1
will be simultaneously homozygous for zw10. Females
(zw10,FRT/ovoD1,FRT;FLP) were collected and mated to sibling
males (FM7a/Y;FLP), and allowed to lay eggs on yeasted grape agar
plates.
Embryos were collected, fixed with formaldehyde, and stained for
immunofluorescence according to previously described procedures
(Karr and Alberts, 1986; Theurkauf, 1992; Williams et al., 1992).
Fixed embryos were rehydrated in Tris-buffered saline + Triton
(TBST; 50 mM Tris-HCl, pH 7.4, 50 mM NaCl, 0.02% sodium azide,
0.1% Triton X-100) and incubated with a 1:10 dilution of anti-βtubulin monoclonal antibody (Calbiochem, La Jolla, CA) in the
presence of 1 µg/ml DNase-free RNase (Boehringer Mannheim)
overnight at 4°C. After washing with TBST, embryos were incubated
in a 1:100 dilution (12.5 µg/ml final concentration) of FITC-conjugated goat anti-mouse IgG (Boehringer Mannheim Biochemicals,
Indianapolis, IN). DNA was stained with both 1 µg/ml propidium
iodide and 0.05 µg/ml Hoechst 33258 for 5 minutes, and mounted in
80% glycerol containing 2% n-propyl gallate. For examination of
788
B. C. Williams and M. L. Goldberg
zw10 protein localization in embryonic polar body nuclei, wild-type
embryos were fixed with formaldehyde and incubated with affinitypurified anti-zw10 antibodies as previously described (Williams et al.,
1992).
Laser-scanning confocal microscopy was primarily carried out
using a Zeiss Axiovert 10 attached to a Bio-Rad MRC600 confocal
imaging system equipped with a Krypton/Argon laser (Bio-Rad Laboratories, Cambridge, MA) at Cornell (Biotechnology Flow
Cytometry and Imaging Facility), with additional images collected
using a similar system in the laboratory of Professor David Glover
(University of Dundee, Scotland). Image collection was performed
utilizing a no. 2 neutral density filter to attenuate photobleaching, and
assisted by Kalman averaging to improve the signal/noise ratio.
Double fluorescence images were collected simultaneously either as
two full screen images or on a split screen, and could be merged in
pseudocolor using COMOS software. Images were photographed with
a Screenstar camera (Presentation Technologies, Sunnyvale, CA).
Immunostaining of brain mitotic figures
Larval brains were dissected and fixed following the protocol of
González et al. (1990), except that, after fixation in formaldehyde,
brains were exposed to methanol for 2 minutes, which appeared to
improve the antibody staining. This methanol treatment was not used
for brains fixed for subsequent staining with anti-cyclin B antibodies.
For drug effect studies, brains were treated with 0.5×10−5 M
colchicine (Sigma Chemical Co., St. Louis, MO) in 0.7% NaCl for
1.5 hours, or 9 µM taxol (Molecular Probes, Eugene, OR) in 0.7%
NaCl, for 1 hour at 24°C before fixation. In certain cases (described
in Results), hypotonic treatment (8 minute incubation of the brains in
0.5% sodium citrate) preceded the fixation (Gatti and Goldberg,
1991). Control brains were incubated in 0.7% NaCl alone.
All antibody incubations were done at 4°C overnight in phosphatebuffered saline (PBS; formula of Karr and Alberts, 1986) containing
0.3% Triton-X (PBST), 10% fetal calf serum (FCS; Sigma Chemical
Co.), DNase-free RNase (1 µg/ml), and 0.02% sodium azide
(González et al., 1990). Washes were done for 30 minutes with several
changes of PBST lacking FCS and RNase. Prior to staining, brains
were first equilibrated in PBST, then blocked in PBST + FCS for 1
hour. For spindle staining, brains were incubated in a 1:5 dilution of
anti-β-tubulin monoclonal antibody (Calbiochem), washed, and
incubated in a 1:50 dilution (final concentration 25 µg/ml) of FITCconjugated goat anti-mouse IgG (Boehringer Mannheim). For zw10
staining, brains were incubated with either crude rabbit anti-zw10
serum (1:300 dilution), mouse anti-zw10 serum (1:100) or affinitypurified rabbit anti-zw10 antibody (1:50), all of which gave identical
results. After washing, the appropriate biotinylated goat anti-rabbit or
anti-mouse IgG (Vector Laboratories, Burlingame, CA) was added at
a dilution of 1:400 (to a final concentration of 4 µg/ml). After washing
again, brains were incubated in a 1:600 dilution of avidin-FITC (4
µg/ml final concentration; Vector Laboratories). After a final wash,
DNA was stained with propidium iodide, mounted, and examined by
confocal microscopy as for embryos (see above). Alternatively, rabbit
anti-zw10 antibodies were detected by TRITC-conjugated anti-rabbit
IgG antibodies (7.5 µg/ml; Jackson Immunoresearch) and DNA
stained with YOYO-1 (benzoxazolium-4-quinolinium dimer) iodide
(Molecular Probes, Eugene, OR) at 0.2 µM for 5 minutes, with
identical results. BX63, a mouse monoclonal antibody that stains centrosomes (Frasch et al., 1986), and Rb271, a rabbit polyclonal
antibody against cyclin B (Whitfield et al., 1990), were gifts from D.
Glover (University of Dundee, Scotland). For double labelling with
anti-mouse and anti-rabbit antibodies (e.g. anti-tubulin and anti-zw10)
the positions of the chromosomes were obtained by Hoechst 33258
fluorescence.
Brain squashes for immunostaining were prepared according to
González and Glover (1993). Brains were fixed as above, except that
incubation in the fixative was reduced to 30 minutes. Up to four brains
at a time were placed in 45% acetic acid for 30 seconds, then in drops
of 60% acetic acid for 3 minutes on a non-siliconized coverslip. A
glass slide was placed on the coverslip and the sandwich inverted;
excess liquid was removed and the brains were then vigorously
squashed. After freezing the slide in liquid nitrogen, the coverslip was
removed with a razor blade, the squash was dehydrated in ethanol for
30 minutes, immediately rehydrated in PBST without letting the slide
dry out, and stained with antibodies as above except that incubation
with the secondary antibodies was reduced to 2 hours at room temperature. When examining brains of other mitotic mutants, one wildtype brain was placed on each slide as a control for antibody staining.
RESULTS
The zw10 mutant phenotype and zw10 protein
distribution in syncytial and cellular mitoses
Previously, we examined the phenotypic effects of zw10
mutations in larval neuroblast (brain) cells, but determined the
cell cycle-dependent distribution of zw10 protein in syncytial
blastoderm embryos (Williams et al., 1992). Mitosis exhibits
significant differences in these two tissue types, including variations in cell cycle controls, source of mitotic components
(maternal vs zygotic), and syncytial vs cellularized modes of
division. To correlate these sets of observations preliminary to
the studies reported here, it was thus important to analyze the
phenotype and the distribution of zw10 gene product in both
tissues.
Phenotype of zw10 mutant embryos
The first 13 nuclear divisions of Drosophila embryogenesis
rely almost entirely on maternally supplied gene products
stored in the oocyte. Although it was difficult to obtain zw10
homozygotes given their almost complete lethality, in rare
instances such females can be obtained. Embryos collected
from these females showed a variety of mitotic defects at the
syncytial blastoderm stage, including uneven nuclear density
and aberrant anaphases often containing lagging chromatids or
chromatin bridging. Larger numbers of zw10-deficient
embryos, necessary for more detailed examination of the
mutant phenotype, can be generated in homozygous germline
clones produced from zw10/zw10+ heterozygous females using
the FLP recombinase system (Chou and Perrimon, 1992; see
Materials and Methods).
To determine the lethal phase, embryos derived from
homozygous zw10 germline clones were allowed to develop,
then fixed and stained with Hoechst 33258 to visualize nuclei.
zw10 embryos terminated development primarily in the late
syncytial blastoderm stages, though degenerating embryos
apparently representing earlier developmental stages are also
commonly found (data not shown). A subpopulation (15-25%)
develop through later stages of embryogenesis and die, though
a small fraction (1%) of them eventually hatch into larvae,
most likely due to paternal rescue of zw10 function by wildtype sperm.
To examine associated nuclear division abnormalities, zw10
mutant embryos were collected at timed intervals, fixed, and
stained to visualize DNA and microtubules (see Materials and
Methods). In wild type, the syncytial blastoderm stage is
typified by a spatially even layer of metasynchronously dividing
nuclei at the surface of the egg (Foe and Alberts, 1983).
Spindles are bipolar and prominent asters at the poles are seen
Drosophila zw10 protein
789
Fig. 4. Metaphases in zw10 mutant
embryos. zw10 mutations do not
directly affect the formation of the
metaphase plate or the metaphase
spindle as visualized here in fields of
nuclei stained with Hoechst (A,C)
and anti-tubulin antibodies (B,D) in
syncytial blastoderm embryos derived
from homozygous zw10S1 germline
clones. Metaphase plates (filled
arrows in A,C) appear to be normal
(refer to Fig. 1A,B for wild-type
metaphase plate) when the spindle is
bipolar (filled arrows in B,D).
However, anaphase chromatid
separation is often abnormal (outline
arrows in A; see also Fig. 2B). Other
abnormalities, such as spindles
lacking asters (arrowheads, B) and
free asters (B,D) can also be seen in
the same field but are not likely to be
direct effects of zw10 mutations (see
text). Bar, 10 µm.
(Fig. 1A,B). In zw10 embryos, however, mitotic synchrony is
lost. Nuclei often have a highly uneven distribution in the
cortex, and free centrosomes and multipolar spindles are
common (Fig. 1C,D). However, these defects are unlikely to be
the direct consequence of lesions in the zw10 gene. Many
mutations or drug treatments that affect virtually any aspect of
nuclear division produce similar effects (Freeman et al., 1986;
Raff and Glover, 1988; Sunkel and Glover, 1988; González et
al., 1990; Lin and Wolfner, 1991; Girdham and Glover, 1991;
Gomes et al., 1993). This is thought to be due to loss of coordination between the nuclear and centrosome cycles, which is
particularly sensitive to disruption during the rapid syncytial
divisions. More importantly, anaphase and post-anaphase aberrations similar to those seen in zw10 mutant brains (Williams
et al., 1992) are immediately visible in zw10-deficient embryos.
These include chromatin bridges between nuclei (Fig. 2B;
compare to wild-type in Fig. 2A), unequal segregation of chromatids at anaphase, and lagging chromatids at anaphase (Fig.
2B). We believe these phenomena to be the primary defects
associated with the absence of zw10 function in embryos. Additional aspects of this embryonic phenotype are discussed below.
Localization of zw10 protein in neuroblast cells
We have characterized the distribution of zw10 antigen during
mitosis in wild-type neuroblast cells by examining fixed,
whole-mounted brains stained with anti-zw10 antibody using
confocal microscopy. The localization of zw10 protein is
essentially identical to that previously described for syncytial
blastoderm embryos (Williams et al., 1992). The zw10 protein
remains primarily cytoplasmic at interphase and prophase (not
shown). By metaphase, it becomes associated with, or forms,
filaments that pass through the central region of the metaphase
plate (Fig. 3A-C). At anaphase, zw10 is found almost exclusively at the leading edges of the chromosomes in the region
of the kinetochores (Fig. 3D,E) until telophase when it is
dispersed into the cytoplasm (not shown). Importantly, none of
these structures was recognized by anti-zw10 antibodies in
brains of zw10S1/Y larva (not shown), which do not contain
zw10 protein on Western blots (Williams et al., 1992). This
last observation confirms that these structures do indeed
contain zw10 protein.
In summary, these results combined with previously
published observations (Williams et al., 1992) show that the
zw10 mutant phenotype and the cell cycle-dependent distribution of zw10 protein are similar in two very diverse types of
tissue. The biochemical function of zw10 is therefore likely to
be the same in many kinds of cell division.
Organization of mutant mitoses prior to anaphase
One of the central goals of this study was to determine whether
790
B. C. Williams and M. L. Goldberg
Fig. 5. Metaphases in zw10 mutant
brains. Metaphase plate (blue;
arrows) and spindle formation (green)
were assessed in whole mounted
brains examined by confocal
microscopy. Single optical sections
from typical wild-type (A) and
zw10S1/Y (B,C) neuroblasts are
shown; no differences between the
two are apparent. Head-on views of
the metaphase plate in zw10 brains
(C) show no alteration in the orientation of chromosomes (compare with wild-type in Fig. 3B,C above). Anti-tubulin staining of the mitotic
Fig. 7. Cyclin B levels in wild-type and zw10 brains
treated with colchicine. Cyclin B abundance in
mitotically arrested colchicine-treated neuroblasts
appears to be the same in (A) wild-type and (B)
zw10/Y mutant brains. Brains were treated with
colchicine as described (see Materials and Methods)
and immunofluorescence performed to show
chromosomes (blue) and cyclin B (purple). Wildtype and zw10/Y brains were incubated with
colchicine for the same duration, and all conditions
for staining were identical. Images were collected
using the same settings on the confocal microscope
and merged without adjustment. Individual
metaphase-arrested cells (arrows) are recognized by
their highly condensed chromosomes. Bars, 10 µm.
Fig. 8. Polar body localization of zw10
protein. Early embryos were collected
and fixed (see Materials and Methods),
and stained with propidium iodide to
visualize chromosomes (blue), and with
affinity-purified anti-zw10 antibody to
locate zw10 antigen (red). (A,B,C) are
superimposed views of zw10 staining and
chromosomes in single optical sections of
polar bodies visualized by confocal
microscopy. Arrows point to relatively
clear instances of zw10 antigen
localization to regions at or near
kinetochores. Bars, 5 µm.
absence of zw10 function disrupts processes that occur prior
to the onset of anaphase. Are there any obvious abnormalities
in the migration of chromosomes to the metaphase plate or in
metaphase spindle structure in either zw10-deficient embryos
or in mutant neuroblast cells?
Metaphase structures in zw10-deficient embryos
As noted above, embryos devoid of zw10 activity contain
many free centrosomes, which appears to be a non-specific
consequence of disrupted nuclear division. These free centrosomes are often shared between closely adjacent nuclei,
leading to ‘frayed’ or fused tri- and multi-polar spindles. Thus,
in order to assess the direct effect of zw10 mutations on
metaphase plate formation and the spindle in embryos, we
restricted our observations to nuclei that were in regions of
normal nuclear density and in which the spindles were clearly
Drosophila zw10 protein
791
Fig. 6. Effects of mitotic poisons
on spindle structure and zw10
distribution. Wild-type third
instar larval brains were treated
with colchicine (A-E) or with
taxol (F-I) as described in
Materials and Methods, and
stained for DNA (blue) and
zw10 antigen (red).
(A-C) Colchicine treated
neuroblasts in whole mounts
showing large accumulations of
zw10 antigen on, between and
around the condensed
chromosomes.
(D,E) Kinetochore region
localization of zw10 antigen
(arrows) in colchicine-treated
whole-mounted brains (D) and
colchicine-hypotonic-treated (E)
brain cells in a squashed
preparation. (F) Taxol-treated
neuroblast from a wholemounted brain showing a large
monoaster (green) with highly
condensed chromosomes (blue)
encircling the monoaster.
Compare with the untreated
spindle in Fig. 5A.
(G,H) Examples of zw10
localization in taxol-treated
neuroblasts showing in (G)
accumulations of zw10 antigen
around chromosomes, and in (H)
association of the zw10 protein
with the kinetochore regions.
(I) zw10 (red) in relation to a
large taxol-induced monoaster
(green; see arrow). zw10 is not
localized to the microtubules in the monoaster; instead, it remains associated with the mass of condensed chromosomes, which were visualized
separately with Hoechst staining (not shown). Same magnification in (A,B,C,D), in (G,H), and in (F,G). Bars, 5 µm.
nucleated from only two centrosomes. In these cases, the
spindles are comparable to wild-type in appearance (Fig. 4AD). All chromosomes congress to a tight metaphase plate; we
see no evidence of DNA that is unassociated with the
metaphase plate.
Metaphase structures in zw10 larval neuroblasts
Because of the complications introduced by free centrosomes
in syncytial mutant embryos, we wished to confirm our impressions about spindle and metaphase plate organization by
examining these structures in third instar larval brain tissue. In
this cellularized material, centrosomes cannot be shared by
adjacent cells, thus clarifying the analysis.
To this end, confocal microscopy of fixed, whole-mounted
zw10 brains were used to examine metaphase structures in
three dimensions. We looked in particular for chromosomal
abnormalities (defects in metaphase plate formation or the
presence of maloriented chromosomes) and mitotic spindle
defects (monopolar or multipolar spindles or aberrations in
gross morphology). Examination of a large population of cells
in 40 larval brains revealed that the overall appearances of the
metaphase plate and spindle in zw10 brains are comparable to
those in wild-type brains (Fig. 5A,B). Head-on views of the
metaphase plate in zw10 brains are particularly revealing,
showing the normal chromosome configuration with centromeres pointed inward, which are typically seen in wild-type
(Fig. 5C). We have never observed chromosomes that are
clearly unattached to the metaphase plate. Operationally, this
could be distinguished only if individual chromosomes were
separated from the metaphase plate by more than approximately one-fifth of the distance from the main mass of chromosomes to the pole. Such events were rarely, if ever, observed
in zw10 mutant brain metaphases. However, it should be noted
that subtle defects in zw10 brains, such as chromosomes at the
metaphase plate that are unconnected to kinetochore microtubules, or effects on small subpopulations of microtubules,
792
B. C. Williams and M. L. Goldberg
would not have been detectable in these immunofluorescence
experiments.
Interphase and prophase microtubule distribution in zw10
mutants appears to be normal as well; centrosome localization
is not obviously altered in any stage of the cell cycle (not
shown). In contrast, many anaphases in whole-mounted brains
contain lagging chromatids, as previously seen in squashed
preparations (Williams et al., 1992). The shape of these lagging
chromatids, indicative of kinetochore microtubule attachment,
suggests strongly that they are being translocated towards the
pole, albeit in a delayed fashion. Otherwise, anaphase spindle
morphology appears to be normal (not shown).
Effect of mitotic arrest on zw10 localization
Colchicine treatment
Colchicine, an inhibitor of microtubule assembly, causes cell
cycle arrest in wild-type brain cells; this block is generally
thought to be at prometaphase (Rieder and Palazzo, 1992).
Colchicine treatment, followed by a short incubation in
hypotonic solution, has long been used for examining karyotypes in many organisms (Hsu, 1952; Gatti and Goldberg,
1991). Chromosomes in cells treated in this manner are highly
condensed and do not assemble into a compact metaphase
plate. The sister chromatids of these chromosomes appear in
squashed preparations to remain attached at the centromeres,
suggesting that centromere separation is normally dependent
upon the action of the mitotic spindle (González et al., 1991).
In contrast, zw10 mutant brains treated in the same way exhibit
precocious sister chromatid separation (PSCS): sister chromatids are separated from each other in a high proportion (3040%) of mutant cells (Williams et al., 1992).
In order to understand the PSCS phenomenon in terms of
possible models for zw10 action, we used confocal microscopy
to determine where the zw10 product is found in colchicinetreated wild-type cells. Unexpectedly, this localization
primarily consists of very intensely staining concentrations of
zw10 antigen around, on, and between chromosomes (Fig. 6AC). Localization in the vicinity of kinetochores is often clearly
visible in whole mounts, especially with shorter times of incubation of brains in colchicine (Fig. 6D). This effect is particularly noticeable in squashed preparations, where the locations
of the centromeres are more easily distinguished (Fig. 6E).
Further characterization of these colchicine-treated cells
provides additional evidence that they are indeed in classical
prometaphase arrest. No organized spindles are observed; in
addition, centrosomes do not separate to opposite poles of the
cell (not shown).
Taxol treatment
Taxol, a drug that stabilizes microtubules and therefore
promotes microtubule assembly, is known to cause mitotic
arrest in mononuclear blood cells (Sondell et al., 1988). We
wished to determine whether taxol treatment of zw10 mutant
Drosophila brains would induce the PSCS phenomenon. When
zw10/Y brains are treated with 9 µM taxol for one hour,
followed by a short hypotonic shock, a far higher proportion
of nuclei display PSCS (60% of cells) than is the case in cells
in similarly treated wild-type brains (7%). We therefore
suggest that PSCS is a generalized response to mitotic arrest
in the absence of the zw10 gene product.
The state of cells arrested by taxol is different in many
respects from those arrested with colchicine. In wild type and
in zw10 mutant brains treated with taxol, mitotically arrested
cells contain large asters averaging 5-7 µm in diameter. Each
cell usually contains one aster; the condensed chromosomes
encircle or lie off to one side of the monoaster (Fig. 6F). In
69% of mitotically arrested cells, these large asters are associated with unseparated or unduplicated centrosomes (not
shown). In the remainder (31%), an abnormally short separation (0.5-2.0 µm) of centrosomes was observed, compared with
the approximately 10 µm separation of centrosomes at
metaphase in untreated brains (not shown). These observations
are in agreement with studies in other organisms, where taxol
has been reported to produce large monoasters and to prevent
the movement of centrioles to opposite poles (Daub and
Hauser, 1988; Rao et al., 1989; Buendia et al., 1990).
In spite of the differences in the appearance of the taxol- and
colchicine-arrested cells, the zw10 antigen is localized to
essentially the same positions in both types of drug-treated
brains. High concentrations of zw10 protein are again observed
within the chromosomal mass of taxol-arrested cells (Fig. 6G).
Occasionally, localization to the kinetochore regions of individual chromosomes can be seen (Fig. 6H). It is important to
note that, with taxol treatment, zw10 protein preferentially
localizes to the chromosomal region rather than to the
extensive array of microtubules in the monoaster (Fig. 6I).
Cyclin B levels in mitotically arrested zw10 brains
One possible explanation of the PSCS phenomenon is that
drug-treated zw10 mutant (but not wild-type) cells actually
transit through anaphase onset, as signified by their separated
sister chromatids. The cyclin B-cdc2 kinase complex (MPF or
maturation-promoting factor) would thus be expected to be
inactivated in these cells, an event that accompanies anaphase
onset (Whitfield et al., 1990). However, the subsequent events
of anaphase, such as chromatid movement, could not occur
because of the absence of spindle function in the presence of
microtubule poisons.
In this scenario, the zw10 protein’s normal function would
be to provide feedback control, ensuring that cells stay in
metaphase until assembly of the spindle and metaphase plate
is complete; the existence of such controls has previously been
inferred (Murray and Kirschner, 1989; Rieder and Alexander,
1989). The bub and mad gene products in Saccharomyces cerevisiae provide a precedent for this model of zw10 action. In
wild-type yeast, treatment with the microtubule poison
benomyl arrests the cell cycle in a metaphase-like state characterized by high MPF levels. When yeast carrying mutations
in their bub or mad genes are incubated in benomyl, the cells
exit from mitosis and MPF levels are not sustained. Because
the spindle is compromised by the drug, massive nondisjunction and cell death result (Li and Murray, 1991; Hoyt et al.,
1991).
To test if zw10 mutants were defective in this type of
feedback control of anaphase onset, levels of cyclin B protein
(the labile component of MPF that is degraded at the
metaphase-anaphase transition; Whitfield et al., 1990; Hunt et
al., 1992) were assayed by indirect immunofluorescence. Fig.
7A verifies previous reports (Whitfield et al., 1990) showing
that cyclin B levels remain high in colchicine-treated wild-type
brain cells that contain condensed chromosomes characteristic
Drosophila zw10 protein
of mitotic arrest. In zw10 brains incubated in colchicine, cyclin
B levels are approximately equivalent to those in treated wildtype cells (Fig. 7A,B). Furthermore, elevated cyclin B levels
are observed in both wild-type and zw10 neuroblasts arrested
with taxol (not shown). Thus, PSCS in mutant brains is
unlikely to result from a feedback control defect in which
events dependent upon MPF degradation occur in the absence
of a functional spindle.
zw10 localization in mitotically arrested polar bodies
As described above, one means of studying the influence of
mitotic arrest on zw10 localization is treatment with microtubule poisons. Another method is to examine zw10 in
naturally occurring mitotically arrested nuclei, such as those of
the early embryonic polar bodies. Polar bodies are the three
haploid products of meiosis that remain in the egg after
formation of the female pronucleus (Doane, 1960; CamposOrtega and Hartenstein, 1985; Matthews et al., 1993). They
move to the egg cortex and become arrested at metaphase,
often coalescing with one another so that sometimes only one
or two discrete polar bodies are seen (Doane, 1960). The chromosomes in a polar body are arranged with their centromeres
pointing inwards and telomeres directed towards the periphery,
suggesting a monopolar spindle (Hatsumi and Endow, 1992).
In our hands, anti-tubulin staining shows MTs originating at or
near the chromosomes and extending a short distance into the
surrounding cytoplasm. At the surface of the embryo, the polar
body is ‘capped’ by a proliferation of MTs (not shown).
The zw10 protein is localized at or near kinetochores on
polar body chromosomes (Fig. 8A-C). When compared with
the mitotically dividing nuclei at the syncytial blastoderm
stage, the immunofluorescent signal is much greater on polar
body chromosomes, suggesting a higher concentration of zw10
protein. The polar body chromosomes are condensed and presumably in a metaphase-arrested state, so it is possible that this
staining pattern reflects the concentrated localization of zw10
in larval neuroblast cells arrested in mitosis by spindle poisons
(see above).
Effect of other mitotic mutations on zw10
localization
To determine the role of the zw10 gene product with respect
to the action of other proteins important in chromosome segregation, the localization of the zw10 protein was examined in
brains of other Drosophila mitotic mutants. In particular, we
closely examined the effects of mutations in three genes
required for proper chromatid behavior during anaphase:
namely, rough deal (rod; Karess and Glover, 1989), lodestar
(Girdham and Glover, 1991), and abnormal anaphase resolution (aar; Gomes et al., 1993). At least 50 larval brains
homozygous for each mutation were examined for the localization of zw10 protein during the cell cycle. These observations are described for each mutant in the following sections.
rough deal (rod)
rod mutations result in levels of aneuploidy comparable to
those of zw10, and analogous anaphase defects such as lagging
chromatids are commonly observed (Fig. 9B; Karess and
Glover, 1989). Unlike zw10, however, nuclear bridging (where
a strand of chromatin stretches continuously from one nucleus
to the other during anaphase/telophase) is often seen in rod
793
brains; also, PSCS does not result when rod brains are exposed
to colchicine (Karess and Glover, 1989).
rod mutations severely affect zw10 protein localization. In
brains of larvae homozygous for either weak (X-6) or strong
(H4.8, X-5) alleles of rod, proper metaphase and anaphase
zw10 staining was greatly diminished or abolished (Fig. 9A,B;
compare with wild-type, Fig. 3A-F). In addition, the intense
staining of zw10 antigen near kinetochores seen in colchicinetreated wild-type brains (Fig. 6A-E) was clearly not evident in
colchicine-treated rod brains (Fig. 9C). Mutant rod alleles do
not affect the overall accumulation of the zw10 protein in the
larva, as detected on Western blots (Fig. 10). The zw10 protein
in rod neuroblasts is distributed uniformly throughout the
extranuclear volume (Fig. 9A). Thus, rod mutations most
likely result in defective zw10 protein aggregation or localization.
lodestar
Like rod, mutations at the lodestar locus also affect anaphase.
The primary defect of lodestar mitoses is the failure to separate
chromatin at anaphase, resulting in chromatin bridging and
fragmentation. The lodestar gene encodes a member of the
helicase superfamily (Girdham and Glover, 1991). Our examination of lodestar mutant brains revealed that zw10 localization was similar to that in wild-type throughout mitosis; even
anaphases with tangled chromatids contained the zw10 protein
correctly localized to the kinetochore regions (Fig. 9D).
However, zw10 protein was not localized on chromatin that
remained behind at the former position of the metaphase plate
during anaphase (Fig. 9E). The chromatin most likely represents chromosome fragments lacking centromeres previously
described in lodestar anaphases (Girdham and Glover, 1991).
In accordance with this idea, such chromatin exhibits no consistent poleward orientation, as would be expected if it
contained centromeres capable of attachment to the spindle
(Fig. 9E). These findings suggest that the lodestar helicase is
probably not required for proper zw10 localization during
metaphase and anaphase.
abnormal anaphase resolution (aar)
The aar1 mutant allele causes high levels of chromatin
bridging and lagging chromatids at anaphase. The latter class
of aar1 anaphase figures are identical in appearance to those
seen in zw10 mutants and have been demonstrated to contain
complete centromeres and telomeres (Gomes et al., 1993;
Mayer-Jaekel et al., 1993). The aar gene encodes the 55 kDa
regulatory subunit of protein phosphatase 2A involved in the
metaphase-anaphase transition (Mayer-Jaekel et al., 1993).
The localization of zw10 protein was altered in aberrant aar1
anaphase figures. Typically, chromatids that have moved
properly to the poles show zw10 occupying its normal position
at the kinetochore regions, but chromatids or chromosomes
lagging at the metaphase plate completely lack zw10 staining
(Fig. 9F-H). In aar1 anaphases, chromosome bridges, which
have been shown to contain intact centromeres (Mayer-Jaekel
et al., 1993), also lack zw10 antigen at the expected locations
(Fig. 9I). In aar1 brains, localizations of zw10 at other stages
in mitosis and during colchicine-induced mitotic arrest, were
comparable to those in wild-type (not shown). Thus, the mislocalization of zw10 antigen in aar1 cells appears to be specific
to chromosomes that mis-segregate during anaphase.
794
B. C. Williams and M. L. Goldberg
Drosophila zw10 protein
DISCUSSION
The role of the zw10 protein in syncytial and
cellularized mitoses
The experiments reported previously (Williams et al., 1992)
and those in this paper demonstrate that both the distribution
of the zw10 protein and the phenotype resulting from the
absence of zw10 gene product are similar in syncytial embryos
and in cellularized larval neuroblasts. Thus, the zw10 protein
plays a consistent and important role in governing accurate
mitotic chromosome segregation. This role is similar in both
syncytial and cellularized divisions, which differ in several
aspects of cell cycle control. Maternally and zygotically
supplied zw10 proteins thus appear to have identical properties.
Time and location of zw10 action
Our evidence strongly suggests that zw10 function is first
required at anaphase onset. Null mutations in the zw10 gene
appear to have no obvious effect on either the morphology of
metaphase spindles or the congression of chromosomes to the
metaphase plate in the two tissue types examined. It is nonetheless conceivable that the absence of zw10 activity could cause
subtle though important disruptions in structures or processes
prior to the start of anaphase. Moreover, it is possible that zw10
function might be required prior to anaphase onset, but the
effects of this activity would be undetectable until anaphase
onset. Despite these caveats, we believe it most likely that the
role of the zw10 protein first becomes critical at the metaphaseanaphase transition. This protein undergoes a rapid relocaliza-
Fig. 9. Effects of rod, lodestar and aar mutations on zw10 protein
localization. Larval brains from rodH4.8 (A-C), lodestar079-2 (D,E),
and aar1 (F-I) mutant homozygotes were fixed and stained (see
Materials and Methods) to detect zw10 (red) and DNA (blue).
Regions of overlap between zw10 and DNA appear white.
(A) Metaphase cell from rodH4.8 showing chromosomes situated on
the metaphase plate (arrow) accompanied by absence of zw10
staining in filaments that pass through the metaphase plate (see Fig.
3A). Instead, zw10 is diffuse in the cytoplasm. (B) Anaphase figure
from rodH4.8 (Karess and Glover, 1989) showing a lagging chromatid
(open arrow). zw10 protein is absent from kinetochore regions of all
chromatids. (C) Colchicine-treated rodH4.8 brain showing two cells
in mitotic arrest as indicated by their highly condensed chromosomes
(open arrows). zw10 protein does not accumulate in high levels
around kinetochores as in wild-type (Fig. 6A-E) but instead is
distributed diffusely in the cytoplasm. (D,E) Anaphase figures from
lodestar brains (Girdham and Glover, 1991) displaying chromatin
tangling (D, open arrow) and chromatin fragments that remained at
the metaphase plate (E, open arrow). In both instances, zw10 protein
is present at the kinetochore regions of properly situated chromatids
(filled arrows in D,E). (F-H) Anaphase figures from aar1 brains with
lagging chromatids (described by Gomes et al., 1993). Chromatids
that have moved properly to the poles contain zw10 on the
kinetochore regions (filled arrows) while lagging chromatids
completely lack zw10 staining (open arrows). (I) aar1 anaphase
figure with a stretched chromosome (chromosome bridge) between
the two poles (arrow). Whereas zw10 is properly localized on
separated chromatids at the poles, it is absent from the entire length
of the chromosome bridge. Similar chromosome bridges in aar1
anaphases have been shown to contain two centromeric regions
(Mayer-Jaekel et al., 1993). Bars, 5 µm.
795
tion within a few seconds of anaphase onset (Williams et al.,
1992), suggesting a mechanism by which mutant anaphase
figures can display aberrations while mutant metaphase figures
appear normal.
According to this hypothesis, the localization of zw10
antigen to spindle-associated filaments during normal
metaphase would essentially be for the purpose of sequestering this protein from its normal site of action near or at kinetochores, thereby preventing zw10 function until anaphase
onset. In support of this notion, our findings suggest that the
intrinsic affinity of zw10 protein is for the centromere/kinetochore. Localization of zw10 product to the spindle-associated
metaphase filaments is clearly not an obligatory precondition
for subsequent movement to the chromosomes, as zw10
protein is concentrated in the chromosomal mass even when
spindles are depolymerized by colchicine treatment. Moreover,
when faced with a decision between chromosomes and a large
population of monoaster microtubules in taxol-poisoned cells,
zw10 protein chooses the chromosomes.
The idea that zw10 protein plays no essential role prior to
anaphase will remain speculative until we understand at the
ultrastructural level where it is found in the region of the
metaphase spindle. We have previously suggested that during
metaphase, zw10 is a component of kinetochore microtubules,
consistent with its location in the central portion of the spindle
and with its rapid relocalization to kinetochore regions at
anaphase onset (Williams et al., 1992). However, it is by no
means clear that this supposition is correct. There is some
evidence for the existence of non-microtubule-associated
structures in the spindle (Paddy and Chelsky, 1991; Steffen and
Linck, 1992). In addition, preliminary results indicate that
zw10 is not purified on microtubule affinity columns (B.
Williams, J. Raff and T. Karr, unpublished). It should be noted
in particular that our indirect immunofluorescence studies to
date have insufficient resolution to dismiss the possibility that
some fraction of zw10 protein is located at the
centromere/kinetochores during prometaphase or metaphase,
even if the majority of the antigen at metaphase is found extrachromosomally on the spindle-associated filaments.
The zw10 protein and PSCS
PSCS occurs in zw10 mutant brain cells treated with colchicine
or taxol, both of which cause mitotic arrest. We propose that
the occurrence of PSCS in the absence of zw10 protein
function reflects the unusual localization of zw10 protein in
arrested cells. Our results suggest that zw10 first localizes to
the kinetochores and accumulates there over time, forming
large aggregations of zw10 antigen around the chromosomes.
This accumulation does not result from an increase in zw10
protein synthesis, since the zw10 protein levels in untreated
and colchicine-treated brains are indistinguishable on Western
blots (data not shown). It should be noted that other molecules,
including calmodulin, the ‘32-9’ antigen, and the INCENP
proteins, also redistribute to kinetochores after drug-induced
mitotic arrest (Welsh and Sweet, 1989; Balczon et al., 1990;
Earnshaw and Cooke, 1991).
At least in a formal sense, the chromosomal aggregation of
zw10 protein in mitotically arrested cells acts as part of a
checkpoint control that holds sister chromatids together when
the spindle is disrupted by microtubule poisons. Because cyclin
B levels remain high in these cells, it is unlikely that the zw10
796
B. C. Williams and M. L. Goldberg
be noted, however, that the rod phenotype cannot be completely explained by mislocalization of zw10 protein because
rod mutations do not appear to cause PSCS (Karess and
Glover, 1989).
Fig. 10. zw10 protein levels are unaffected by mutations in rod.
(A) Western blot showing amounts of the 85 kDa zw10 protein
(arrow) in: lane 1, two whole female wild-type larvae; lane 2, two
whole male wild-type larvae; lane 3, two whole homozygous rodX5
female larvae; and lane 4, two whole homozygous rodX5 male larvae.
The zw10 protein band does not show significant differences
between wild-type and rodX5 larvae of both sexes. Crude anti-zw10
antisera was used at 1:4000 dilution and the blot was processed by
enhanced chemiluminescence as described (Williams et al., 1992).
(B) Protein levels in these lanes were assayed by incubation with
antibodies to a 75 kDa Drosophila protein (R. Gandhi and M. L.
Goldberg, unpublished) as a loading control.
protein makes cell cycle progression dependent upon spindle
integrity, a checkpoint that is abolished by the yeast mad and
bub mutations (Li and Murray, 1991; Hoyt et al., 1991).
However, Holloway et al. (1993) have recently shown that
sister chromatids can separate even if MPF remains active. Our
finding that PSCS occurs while cyclin B levels remain high
verify their conclusions, at least in zw10 mutant cells. Thus, it
may be possible that the zw10 product participates in a previously unknown type of feedback control that renders sister
chromatid separation, but not other events that occur at or subsequent to anaphase onset, sensitive to the state of the spindle.
The rod+ product is required for zw10 localization
Mutations in the Drosophila mitotic gene rough deal (Karess
and Glover, 1989) eliminate proper zw10 protein localization
in untreated and in colchicine-treated cells. This effect appears
to be quite specific. Global accumulation of zw10 protein is
not reduced, nor is this due to general disruptions in the mitotic
apparatus: we have confirmed previous findings (Karess and
Glover, 1989) that rod metaphase and anaphase spindles have
normal morphology (data not shown). The complete inability
of zw10 to localize in rod mutants is unique among a large
number of Drosophila mitotic mutants examined (B. Williams
and M. L. Goldberg, unpublished results).
The phenotypic effects of rod and zw10 mutations have
strong similarities: both cause disruptions in anaphase leading
to high levels of aneuploidy. Preliminary examination of
zw10;rod double mutants shows that the levels of aneuploidy
are no worse than in either mutant alone, suggesting that both
genes may be part of the same pathway (R. Karess, personal
communication). We speculate that the rod protein may be
directly responsible for the attachment of zw10 protein to
specific sites within the spindle or on the kinetochore. It should
zw10 protein is missing from mis-segregating
chromosomes in aar anaphase figures
The aar gene encodes the 55 kDa regulatory subunit of the
serine/threonine-specific protein phosphatase 2A (MayerJaekel et al., 1993). Mutations in the aar gene result in the
appearance of lagging chromosomes and chromosome bridges
at anaphase (Gomes et al., 1993), which have been shown by
in situ hybridization procedures to contain intact centromeres
and telomeres (Mayer-Jaekel et al., 1993). It is remarkable that
zw10 protein is absent from the kinetochore region of these
missegregating chromosomes, while it is present on chromosomes that have properly migrated to the poles of the anaphase
spindle. This observation underscores the importance of the
zw10 protein for coordinating chromosome segregation.
Dephosphorylation of many mitotic phosphoproteins occurs
at anaphase onset (Vandre and Borisy, 1989). It thus seems
likely that the phosphorylation state of the zw10 protein, or of
some other component with which it interacts, is necessary for
the movement of zw10 protein to the kinetochore region and
its function there. Our results imply that protein phosphatase
2A participates in such a dephosphorylation event. It is surprising that the consequences of defects in dephosphorylation
are highly compartmentalized: zw10 protein is apparently
completely absent from particular chromosomes, yet it is
present on others. This suggests that highly cooperative interactions are required for zw10 localization to the kinetochore
region.
Possible functions of the zw10 protein
The cytological behavior of zw10 mutants (Williams et al.,
1992; this paper) clearly indicates that the zw10 gene product
plays an essential role in ensuring the proper segregation of
chromosomes during mitosis. The precise biochemical
function of this protein must be consistent with its site and
timing of action. In this paper, we present evidence that the
location of zw10 activity is at the centromere/kinetochore.
Normally, the function of this protein is not required until
anaphase onset, correlating with its movement to the centromere/kinetochore at this stage of mitosis. However, it
appears that the zw10 protein can have an effect prior to the
beginning of anaphase when the cell cycle is arrested by
treatment with microtubule poisons. Presumably, these cells
are blocked in a state similar to metaphase, as evidenced by
their condensed chromosomes and elevated levels of cyclin B.
However, in this abnormal circumstance, zw10 protein apparently again finds its way to the centromere/kinetochore, where
we suggest that its activity is manifested.
These observations are most simply explained by a role for
zw10 either in sister chromatid separation or in anaphase
chromatid movements. The latter possibility appears unlikely
because we have been unable to observe the presence of
tubulin at kinetochores in colchicine-treated Drosophila neuroblasts (not shown). Chromosome movement is dependent on
kinetochore microtubules, yet the absence of zw10 protein has
phenotypic consequences (i.e. PSCS) in cells without obvious
microtubules. However, it remains possible that we were
Drosophila zw10 protein
unable to detect a small population of short microtubules
attached to the kinetochores in colchicine-treated cells; such
microtubules have been observed in colchicine-treated
mammalian and plant cells (Mitchison and Kirschner, 1985;
Utrilla et al., 1989; Meininger et al., 1990).
Two considerations must be kept in mind while evaluating
any potential explanation of zw10 action, including the hypothesis that we favor: that zw10 participates in the process of sister
chromatid separation. First, mutants lacking zw10 gene product
show no evidence of a mitotic block in any stage of the cell
cycle (Williams et al., 1992). This suggests that zw10 is
required for the timing and/or accuracy of chromosome segregation, but is not essential for transit through the cell cycle or
for chromosome distribution to occur at all.
A second conceptual difficulty in understanding the role of
the zw10 protein is that zw10 mutations appear to have
opposite effects in the absence or presence of microtubule
poisons. For example, untreated zw10 mutant cells contain
anaphase figures suggesting a delay in sister chromatid separation, while mitotically arrested zw10 mutant cells show
PSCS. In another view of the same phenomenology, if the
arrival of zw10 protein at the centromere/kinetochore of wildtype untreated cells could be imagined to trigger sister
chromatid separation, then the presence of zw10 at these structures must inhibit sister chromatid separation in drug-arrested
wild-type cells. PSCS in mutants is thus paradoxically associated with the absence of zw10 product from the
centromere/kinetochore. One way to reconcile these observations is to suggest that zw10 activity can vary with the cell
cycle. The zw10 product, or some other molecule it associates
with, might be phosphorylated during normal or drug-blocked
metaphase, but then become dephosphorylated at anaphase.
This could lead to very different effects when the protein
reaches the centromere/kinetochore normally at anaphase
onset, or artefactually during drug-induced metaphase arrest.
The zw10 protein contains at least one potential phosphorylation target; we are currently investigating whether this site is
indeed employed to regulate zw10 activity.
We are grateful to David Glover, Cayetano Gonzalez and Hiroyuki
Ohkura for their help, and for providing protocols, fly stocks and antibodies. We are indebted to Roger Karess for sending rod alleles and
sharing unpublished data. We also thank Jim Slattery and Carol
Bayles for assistance with confocal microscopy, Ingrid Monteleone
for providing fly media, William Theurkauf for advice on embryo
fixation, and Joe Chou and Norbert Perrimon for supplying fly stocks.
This work was supported by NIH grant GM48430 to M.L.G., and in
part by NIH training grant GM07617 to B.C.W. Parts of this work
have been presented in the doctoral thesis ‘Studies on the Drosophila
l(1)zw10 gene and its product’, by B. C. Williams.
REFERENCES
Balczon, R., Accavitti, M. A. and Brinkley, B. R. (1990). Identification of a
40×103 Mr centromere-associated protein in cultured mammalian cells. J.
Cell Sci. 97, 705-713.
Buendia, B., Antony, C., Verde, F., Bornens, M. and Karsenti, E. (1990). A
centrosomal antigen localized on intermediate filaments and mitotic spindle
poles. J. Cell Sci. 97, 259-272.
Campos-Ortega, J. A. and Hartenstein, V. (1985). The Embryonic
Development of Drosophila melanogaster. Berlin: Springer-Verlag.
Chou, T.-B. and Perrimon, N. (1992). Use of a yeast site-specific recombinase
to produce female germline chimeras in Drosophila. Genetics 131, 643-653.
797
Daub, A. M. and Hauser, M. (1988). Taxol affects meiotic spindle function in
locust spermatocytes. Protoplasma 142, 147-155.
Doane, W. H. (1960). Completion of meiosis in uninseminated eggs of
Drosophila melanogaster. Science 132, 677-678.
Earnshaw, W. C. and Cooke, C. A. (1991). Analysis of the distribution of the
INCENPs throughout mitosis reveals the existence of a pathway of structural
changes in the chromosomes during metaphase and early events in cleavage
furrow formation. J. Cell Sci. 98, 443-461.
Foe, V. E. and Alberts, B. M. (1983). Studies of nuclear and cytoplasmic
behavior during the five mitotic cycles that precede gastrulation in
Drosophila embryogenesis. J. Cell Sci. 61, 31-70.
Frasch, M., Glover, D. M. and Saumweber, H. (1986). Nuclear antigens
follow different pathways into daughter nuclei during mitosis in early
Drosophila embryos. J. Cell Sci. 82, 155-172.
Freeman, M., Nusslein-Vollhard, C. and Glover, D. M. (1986). The
dissociation of nuclear and centrosomal division in gnu, a mutation causing
giant nuclei in Drosophila. Cell 46, 457-468.
Gatti, M. and Goldberg, M. L. (1991). Mutations affecting cell division in
Drosophila. Meth. Cell Biol. 35, 543-586.
Girdham, C. H. and Glover, D. M. (1991). Chromosome tangling and
breakage at anaphase result from mutations in lodestar, a Drosophila gene
encoding a putative nucleoside triphosphate binding protein. Genes Dev. 5,
1786-1789.
Gomes, R., Karess, R. E., Ohkura, H., Glover, D. M. and Sunkel, C. E.
(1993). Abnormal anaphase resolution (aar): a locus required for progression
through mitosis in Drosophila. J. Cell Sci. 104, 583-593.
González, C., Saunders, R. D. C., Casal, J., Molina, I., Carmena, M.,
Ripoll, P. and Glover, D. M. (1990). Mutations in the asp locus of
Drosophila lead to multiple free centrosomes in syncytial embryos, but
restrict centrosome duplication in larval neuroblasts. J. Cell Sci. 96, 605-616.
González, C., Jimenez, J. C., Ripoll, P. and Sunkel, C. E. (1991). The spindle
is required for the process of sister chromatid separation in Drosophila
neuroblasts. Exp. Cell Res. 192, 10-15.
González, C. and Glover, D. M. (1993). Methods for studying mutations in
Drosophila. In The Cell Cycle: A Practical Approach (ed. P. Fantes and R.
Brookes). IRL Press, Oxford, England (in press).
Hatsumi, M. and Endow, S. A. (1992). The Drosophila ncd microtubule
motor protein is spindle-associated in meiotic and mitotic cells. J. Cell Sci.
103, 1013-1020.
Holloway, S. L., Glotzer, M., King, R. W. and Murray, A. W. (1993).
Anaphase is initiated by proteolysis rather than by the inactivation of
maturation-promoting factor. Cell 73, 1393-1402.
Hoyt, M. A., Totis, L. and Roberts, B. T. (1991). S. cerevisiae genes required
for cell cycle arrest in response to loss of microtubule function. Cell 66, 507517.
Hsu, T. C. (1952). Mammalian chromosomes in vitro. I. The karyotype of man.
J. Hered. 43, 167-172.
Hunt, T., Luca, F. C. and Ruderman, J. V. (1992). The requirements for
protein synthesis and degradation, and the control of destruction of cyclins A
and B in the meiotic and mitotic cell cycles of the clam embryo. J. Cell Biol.
116, 707-724.
Karess, R. E. and Glover, D. M. (1989). rough deal: a gene required for proper
mitotic segregation in Drosophila. J. Cell Biol. 109, 2951-2961.
Karr, T. L. and Alberts, B. M. (1986). Organization of the cytoskeleton in
early Drosophila embryos. J. Cell Biol. 102, 1494-1509.
Li, R. and Murray, A. W. (1991). Feedback control of mitosis in budding
yeast. Cell 66, 519-531.
Lin, H. and Wolfner, M. (1991). The Drosophila maternal-effect gene fs(1)Ya
encodes a cell cycle-dependent nuclear envelope component required for
embryonic mitosis. Cell 64, 49-62.
Matthews, K. A., Rees, D. and Kaufman, T. C. (1993). A functionally
specialized α-tubulin is required for oocyte meiosis and cleavage mitoses in
Drosophila. Development 117, 977-991.
Mayer-Jaekel, R., Ohkura, H., Gomes, R., Sunkel, C. E., Baumgartner, S.,
Hemmings, B. A. and Glover, D. M. (1993). The 55 kd regulatory subunit
of Drosophila protein phosphatase 2A is required for anaphase. Cell 72, 621633.
Meininger, V., Binet, S., Chaineau, E. and Fellous, A. (1990). In situ
response to vinca alkaloids by microtubules in cultured post-implanted
mouse embryos. Biol. Cell 68, 21-30.
Mitchison, T. J. and Kirschner, M. W. (1985). Properties of the kinetochore in
vitro. I. Microtubule nucleation and tubulin binding. J. Cell Biol. 101, 755-765.
Murray, A. W. and Kirschner, M. W. (1989). Dominoes and clocks: the
union of two views of cell cycle regulation. Science 246, 614-621.
798
B. C. Williams and M. L. Goldberg
Paddy, M. R. and Chelsky, D. (1991). Spoke: a 120-kD protein associated
with a novel filamentous structure on or near kinetochore microtubules in the
mitotic spindle. J. Cell Biol. 113, 161-171.
Raff, J. W. and Glover, D. M. (1988). Nuclear and cytoplasmic mitotic cycles
continue in Drosophila embryos in which DNA synthesis is inhibited with
aphidicolin. J. Cell Biol. 107, 2009-2119.
Rao, P. N., Zhao, J.-Y., Ganju, R. K. and Ashorn, C. L. (1989). Monoclonal
antibody against the centrosome. J. Cell Sci. 93, 63-70.
Rieder, C. L. and Alexander, S. P. (1989). The attachment of chromosomes to
the mitotic spindle and the production of aneuploidy in newt lung cells. In
Mechanisms of Chromosome Distribution and Aneuploidy (ed. M. A.
Resnick and B. K. Vig), pp. 185-194. Alan R. Liss, Inc., New York.
Rieder, C. L. and Palazzo, R. E. (1992). Colcemid and the mitotic cycle. J.
Cell Sci. 102, 387-392.
Smith, D. A., Baker, B. S. and Gatti, M. (1985). Mutations in genes
controlling essential mitotic functions in Drosophila melanogaster. Genetics
110, 647-670.
Sondell, K., Anderson, C., Holmgren, G., Norberg, B. O. and Nordenson, I.
(1988). Cluster formation in PHA-stimulated mononuclear cells from
peripheral blood cells: effects of colcemid and taxol. Lymphology 21, 181186.
Steffen, W. and Linck, R. W. (1992). Evidence for a non-tubulin spindle
matrix and for spindle components immunologically related to tektin
filaments. J. Cell Sci. 101, 809-822.
Sunkel, C. E. and Glover, D. M. (1988). polo, a mitotic mutant of Drosophila
displaying abnormal spindle poles. J. Cell Sci. 89, 25-38.
Theurkauf, W. E. (1992). Behavior of structurally divergent α-tubulin
isotypes during Drosophila embryogenesis: evidence for post-translational
regulation of isotype abundance. Dev. Biol. 154, 205-217.
Utrilla, L., Sans, J. and De-la-Torre, C. (1989). Colchicine-resistant
assembly of tubulin in plant mitosis. Protoplasma 152, 101-108.
Vandre, D. D. and Borisy, G. G. (1989). Anaphase onset and
dephosphorylation of mitotic phosphoproteins occur concomitantly. J. Cell
Sci. 94, 245-58.
Welsh, M. J. and Sweet, S. C. (1989). Calmodulin regulation of spindle
function. In Mitosis, Molecules and Mechanisms (ed. J. S. Hyams and B. R.
Brinkley), pp. 203-240. Academic Press, San Diego.
Whitfield, W. G. F., González, C., Maldonado-Codina, G. and Glover, D.
M. (1990). The A and B type cyclins are accumulated and destroyed in
temporally distinct events that define separable phases of the G2/M
transition. EMBO J. 9, 2563-2572.
Williams, B. C., Karr, T. L., Montgomery, J. M. and Goldberg, M. L.
(1992). The Drosophila l(1)zw10 gene product, required for accurate mitotic
chromosome segregation, is redistributed at anaphase onset. J. Cell Biol. 118,
759-773.
(Received 3 August 1993 - Accepted, in revised form,
3 December 1993)