ArticlePDF Available

CHO1, a mammalian kinesin-like protein, interacts with F-actin and is involved in the terminal phase of cytokinesis

Rockefeller University Press
Journal of Cell Biology (JCB)
Authors:

Abstract and Figures

CHO1 is a kinesin-like protein of the mitotic kinesin-like protein (MKLP)1 subfamily present in central spindles and midbodies in mammalian cells. It is different from other subfamily members in that it contains an extra approximately 300 bp in the COOH-terminal tail. Analysis of the chicken genomic sequence showed that heterogeneity is derived from alternative splicing, and exon 18 is expressed in only the CHO1 isoform. CHO1 and its truncated isoform MKLP1 are coexpressed in a single cell. Surprisingly, the sequence encoded by exon 18 possesses a capability to interact with F-actin, suggesting that CHO1 can associate with both microtubule and actin cytoskeletons. Microinjection of exon 18-specific antibodies did not result in any inhibitory effects on karyokinesis and early stages of cytokinesis. However, almost completely separated daughter cells became reunited to form a binulceate cell, suggesting that the exon 18 protein may not have a role in the formation and ingression of the contractile ring in the cortex. Rather, it might be involved directly or indirectly in the membrane events necessary for completion of the terminal phase of cytokinesis.
Content may be subject to copyright.
JCB
The Rockefeller University Press, 0021-9525/2002/3/783/8 $5.00
The Journal of Cell Biology, Volume 156, Number 5, March 4, 2002 783–790
http://www.jcb.org/cgi/doi/10.1083/jcb.200109090
783
Report
CHO1, a mammalian kinesin-like protein, interacts
with F-actin and is involved in the terminal phase
of cytokinesis
Ryoko Kuriyama, Charles Gustus, Yasuhiko Terada, Yumi Uetake, and Jurgita Matuliene
Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455
HO1 is a kinesin-like protein of the mitotic kinesin-like
protein (MKLP)1 subfamily present in central spindles
and midbodies in mammalian cells. It is different
from other subfamily members in that it contains an extra
300 bp in the COOH-terminal tail. Analysis of the chicken
genomic sequence showed that heterogeneity is derived
from alternative splicing, and exon 18 is expressed in only the
CHO1 isoform. CHO1 and its truncated isoform MKLP1 are
coexpressed in a single cell. Surprisingly, the sequence
encoded by exon 18 possesses a capability to interact
with F-actin, suggesting that CHO1 can associate with both
C
microtubule and actin cytoskeletons. Microinjection of
exon 18–specific antibodies did not result in any inhibitory
effects on karyokinesis and early stages of cytokinesis.
However, almost completely separated daughter cells
became reunited to form a binulceate cell, suggesting that
the exon 18 protein may not have a role in the formation
and ingression of the contractile ring in the cortex. Rather,
it might be involved directly or indirectly in the membrane
events necessary for completion of the terminal phase of
cytokinesis.
Introduction
To ensure high fidelity of chromosome segregation, cells
form a special device called mitotic spindles. The structural
framework of the spindle is microtubules, and their assembly
into the spindle and function in chromosome separation and
cytokinesis are largely dependent on microtubule-based
motor proteins associated with them. A number of kinesin-like
proteins have been identified and further divided into distinct
subclasses (Miki et al., 2001). One such subclass designated
as a mitotic kinesin-like protein (MKLP)*1 subfamily contains
plus-end–directed NH
2
-terminal motor proteins. Since the
first member (CHO1) was identified in CHO cells (Sellitto
and Kuriyama, 1988), other members of this subfamily have
been shown to exist in various organisms, including human
(MKLP1 [Nislow et al., 1992]),
Caenorhabditis elegans
(ZEN-4 [Raich et al., 1998]; CeMKLP1 [Powers et al.,
1998]),
Drosophila
(PAV-KLP [Adams, 1998]), zebrafish
(sequence data available from GenBank/EMBL/DDBJ under
accession no. AF139990), and sea urchin (Chui et al.,
2000). Although all of those proteins are composed of
well-conserved three subdomains (NH
2
-terminal motor
region and COOH-terminal globular tail connected with the
-helical coiled-coil central stalk), CHO1 has been noted to
be unique in a sense that it contains extra
100 amino acids
in the middle of the tail (Kuriyama et al., 1994). Since none
of the homologs have the CHO1-specific tail sequence, it is
not clear whether the proteins are heterogeneous among species
or different forms of MKLP1/CHO1 coexist in a single
cell type.
MKLP1/CHO1-related proteins show dynamic changes
in their subcellular distribution during the cell cycle. In
mammalian cells, CHO1 is present in interphase centrosomes
and nuclei and becomes associated with the mitotic spindle.
As chromosomes move toward poles, the protein shifts to
the midzone and eventually concentrates into a bright spot
in the middle of the intercellular bridge (Sellitto and
Kuriyama, 1988). Since it is a plus-end–directed motor
present in the interzonal region of the spindle, MKLP1/
CHO1 was originally thought to function in chromosome
separation and spindle elongation during karyokinesis
(Nislow et al., 1992). In fact, microinjection of CHO1
antibodies caused mitotic arrest in mammalian cells (Nislow
et al., 1990) and sea urchin embryos (Wright et al., 1993).
However, genetic analysis has suggested involvement of
zen-4
and
pavarotti
in cytokinesis rather than karyokinesis (Adams
Address correspondence to Ryoko Kuriyama, Dept. of Genetics, Cell
Biology, and Development, 6-160 Jackson Hall, 321 Church St. SE,
University of Minnesota, Minneapolis, MN 55455. Tel.: (612) 624-
0471. Fax: (612) 626-6140. E-mail: ryoko@lenti.med.umn.edu
*Abbreviations used in this paper: GFP, green fluorescent protein;
MKLP, mitotic kinesin-like protein.
Key words: actin; alternative splicing; cytokinesis; kinesin-like protein;
midbody
784 The Journal of Cell Biology
|
Volume 156, Number 5, 2002
et al., 1998; Powers et al., 1998; Raich et al., 1998). Evi-
dence has also been presented that overexpression of the mu-
tant CHO1 and RNA-mediated interference specifically
blocked completion of cytokinesis in mammalian cells (Ma-
tuliene and Kuriyama, 2002). The ability of the motor pro-
teins to organize central spindles and the midbody appears
to be essential for their function in cytokinesis, which could
be achieved through their interaction with Aurora kinase
AIR-2 (Schumacher et al., 1998; Severson et al., 2001) and
RhoGAP Cyk-4 (Jantsch-Plunger et al., 2000).
To clarify the nature of molecular diversity among species
and define the role of MKLP1/CHO1 during cell division,
we examined the chicken genomic sequence. Here we report
that heterogeneity of the COOH-terminal tail is derived
from alternative splicing and that the sequence encoded by
exon 18 is expressed in only the CHO1 isoform. Exon 18
includes a polypeptide capable of interaction with F-actin in
vivo and in vitro, and microinjection of exon 18–specific an-
tibodies blocked the terminal phase of cytokinesis. A possi-
bility of CHO1 involvement in membrane events, rather
than the actin-containing contractile ring in the cell cortex,
is discussed.
Results and discussion
Coexpression of CHO1 and MKLP1 isoforms generated
by alternative splicing
Although CHO1 is a member of the MKLP1 subfamily, it is
significantly larger in size than other subfamily members.
This suggested a possibility of alternative splicing, which was
confirmed by cloning of chicken genomic sequences (Fig.
1). The chicken CHO1/MKLP1 gene spans
17.1 kb in
which 22 introns intervene between 23 exons flanked by
GT-AG consensus splice sequences (unpublished data). The
coding sequences of the motor (amino acids 1–441), stalk
(amino acids 442–654), and tail (amino acids 655–954) do-
mains are covered by exons 1–13, 13–17, and 17–23, re-
spectively. By comparing with the human sequence available
in the genome databanks, it was concluded that all exon-
intron boundaries occur at the same position between two
species (Fig. 1). Thus, the splicing pattern of CHO1/MKLP1
gene is well conserved in higher eukaryotes. The major di-
versity resides in the tail domain at the COOH terminus
(Fig. 2). Particularly prominent is the difference between
CHO1 and MKLP1, and a nearly 100 amino acid sequence
encoded by exon 18 is expressed in only CHO1 but not the
MKLP1 isoform.
Chicken cDNA encoding both CHO1- and MKLP1-type
proteins was cloned from a library (Fig. 2, GgCHO1 and
GgMKLP1), suggesting that two isoforms are likely to be ex-
pressed in a single organism. Indeed, cDNA fragments cor-
responding to each isoform were identified in EST data-
banks of not only human but also mouse. To confirm
simultaneous expression of CHO1 and MKLP1, we double
labeled CHO (Fig. 3 A) and HeLa (Fig. 3 B) cells with mono-
clonal anti-CHO1 antibody (mAb) and polyclonal anti-
body (E18) specific to exon 18. CHO1 probed by E18
was seen at the center of spindles and midbodies along with
the antigen recognized by mAb, which is reactive to the
COOH-terminal end of the stalk sequence (unpublished
data). Midbodies isolated from CHO cells are also intensely
stained with both types of antibodies (Fig. 3, C and D).
Thus, CHO1 and MKLP1 are coexpressed and tightly at-
tached to the midbody.
mAb recognizes polypeptides of 95 and 105 kD in
CHO and HeLa cells (Fig. 3 E, lanes 1 and 3). In some
occasions, the polypeptides are resolved as multiple bands,
suggesting the possibility of protein phosphorylation (un-
published data). When the same protein fractions were
probed with E18, only the band with the higher molecu-
lar mass was detected (Fig. 3 E, lanes 2 and 4). Both types
of proteins are highly enriched in isolated midbodies (Fig.
3, lanes 5 and 6). It is notable that immunostaining (Fig.
3, A and B) and immunoblotting (Fig. 3 E, lanes 2 and 4)
signals in whole cells obtained with E18 are far less in-
tense than those with mAb. This may suggest that either
CHO1 is minor or midbody staining with E18 is less ef-
fective than with mAb, or both. It was also noted that nei-
Figure 1. MKLP1/CHO1 gene organization in chicken and human. The middle column represents the full-coding chicken cDNA. Numbers
1–23 indicate the position of exons intervening between 22 introns. Although human MKLP1/CHO1 gene (23 kb) is considerably larger
than chicken (17 kb), almost all splicing sites occur at the same position between two species. A shaded column is a CHO1-specific exon
18, and dotted lines represent the gap detected in the human genomic sequence (sequence data available from EMBL/GenBank/DDBJ under
accession no. NT_010222.1; mapped to chromosome 15). Bars: (top and bottom) 5 kb; (middle) 0.5 kb.
CHO1 isoform with an actin-interacting domain |
Kuriyama et al. 785
ther PAV-KLP nor ZEN-4/CeMKLP1 includes the se-
quence corresponding to exon 18 in their genomic
sequences. However, immunostaining of
Drosophila
cul-
tured cells revealed the presence of an E18-reactive mole-
cule (
200 kD) at the center of the intercellular bridge
(unpublished data). Thus, a CHO1-related molecule with
the exon 18 sequence might colocalize with PAV-KLP at
the midzone and midbody in
Drosophila
cells.
Figure 2. Sequence alignment of the tail among MKLP1 subfamily members. In chicken and human, the COOH-terminal tail is encoded by
exons 17–23, which are differently colored. Sequences are derived from chicken (GgCHO1 and GgMKLP1; this paper), CHO cells (CgCH:
sequence data available from EMBL/GenBank/DDBJ under accession no. X83575), human EST (HsCHO1: sequence data available from EMBL/
GenBank/DDBJ under accession nos. AU123817 and BF897861), mouse EST (MmCHO1: sequence data available from EMBL/GenBank/DDBJ
under accession nos. BE333860, AA856173, and BG068324; MmMKLP1: sequence data available from EMBL/GenBank/DDBJ under accession
nos. AA798124 and AW907863), HeLa (HsMKLP1: sequence data available from EMBL/GenBank/DDBJ under accession nos. X67155 and
S46300), zebrafish (DrMKLP1: sequence data available from EMBL/GenBank/DDBJ under accession no. AF139990), sea urchin (SpMKLP1:
sequence data available from EMBL/GenBank/DDBJ under accession no. AAG18582), Drosophila (PAV-KLP: sequence data available from EMBL/
GenBank/DDBJ under accession no. AJ224882), and C. elegans (ZEN-4: sequence data available from EMBL/GenBank/DDBJ under accession nos.
AF057567 and AF057568). The E18 antibody was raised against the oligopeptide sequence encoded by exon 18 (double underlined).
786 The Journal of Cell Biology
|
Volume 156, Number 5, 2002
CHO1-specific exon 18 encodes an
actin-interacting domain
To characterize the CHO1-specific sequence, we expressed
91 amino acids encoded by exon 18 in CHO cells. The green
fluorescent protein (GFP)-tagged exogenous polypeptide is
located inside the cytoplasm in association with cytoskeletal
fibers (Fig. 4 A). Double staining with Alexa-phalloidin
(
MFs
) provided evidence that the fibers to which the exon
18 sequence attaches are actin filaments. When cells were
treated with dihydrocytochalasin B, GFP was no longer de-
tected along cytoskeletal fibers (Fig. 4 B,
DHCB
). Instead,
actin-containing dots formed by depolymerization of F-actin
become visible by GFP fluorescence. In contrast, the exon 18
polypeptide does not reveal any affinity to the microtubule
network (Fig. 4 C). This is in good agreement with our pre-
vious observation that CHO1 interacts with microtubules
through microtubule-binding sites located at the NH
2
-termi-
nal half of the protein (Matuliene and Kuriyama, 2002).
Affinity of the exon 18 sequence for F-actin was further
confirmed by in vitro cosedimentation. Fig. 4 D shows
CHO (lanes 1–4) and insect Sf9 cell lysates (lanes 5–8) con-
taining the GFP-tagged exon 18 sequence mixed with F-actin
prepared from rabbit skeletal muscles: the polypeptide
was cosedimented with F-actin (lanes 4 and 8). Lanes 9–16
demonstrate interaction of F-actin with
35
S-labeled GFP–
exon 18 synthesized in vitro: the protein was recovered in
the pellet along with F-actin (lane 14), whereas the control
35
S-luciferase remained in the supernatant (lane 16). Associ-
ation of GFP–exon 18 was also confirmed with platelet
F-actin (unpublished data). These results clearly indicate
that the sequence unique to the CHO1 isoform encodes a
polypeptide capable of interaction with F-actin. Exon 18
does not appear to contain any known consensus motifs
shared among actin-binding proteins. Thus, the question of
whether CHO1 interacts with F-actin directly or indirectly
still remains to be answered.
CHO1 is seen inside the nucleus, which is due to the pres-
ence of a nuclear localization signal at the COOH-terminal
end of the tail (unpublished data). When the full-length
CHO1 lacking nuclear localization signal was expressed in
interphase cells, the tagged molecule was located in the cyto-
plasm in association with both microtubules and F-actin
(unpublished data). However, in mitotic cells the protein
was seen predominantly in the spindle, and virtually no fluo-
rescent signal was detected in the cortex where the actin-con-
taining contractile ring is assembled. Since the exon 18 se-
quence was targeted to the cortex, especially in the vicinity of
the furrow during cytokinesis in mitotic cells (unpublished
data), the interaction of CHO1 with actin-containing struc-
tures may be controlled in a cell cycle–dependent manner.
Figure 3. Midbody staining with domain-specific antibodies. (A–D) Localization of the CHO1 isoform in CHO (A), HeLa cells (B), and isolated
CHO midbodies (C and D) probed by E18 antibodies. The same cells/structures were also stained with either mAb (A, B, and D) or anti–-tubulin
antibody (C). Bars, 10 m. (E) Immunoblot analysis of whole CHO (lanes 1 and 2), HeLa cells (lanes 3and 4), and isolated midbodies (lanes 5
and 6) stained with mAb (lanes 1, 3, and 5) and E18 (lanes 2, 4, and 6).
CHO1 isoform with an actin-interacting domain |
Kuriyama et al. 787
Cells treated with E18 antibodies failed to complete
the terminal phase of cytokinesis
By mutating the mechanochemical motor domain of
CHO1, we have shown previously that the protein is re-
quired for completion of cytoplasmic division in mamma-
lian cells (Matuliene and Kuriyama, 2002). To examine
whether inhibition of the actin-interacting domain results
in the similar phenotypes, we microinjected the affinity
purified E18 antibody into PtK
1
cells. In Fig. 5, the
metaphase cell received E18 at time zero (Fig. 5 A) under-
went normal chromosomes separation (Fig. 5 C) and cyto-
plasmic division (Fig. 5 D). Although two daughter cells
appeared to be separated completely, the cell boundary be-
came unclear (Fig. 5, E and F), and the two cells eventually
merged together by
3 h after antibody injection (Fig. 5
G). mAb staining of the fixed cell clearly indicated the for-
mation of a midbody between two separated nuclei (Fig. 5,
arrows). Nonetheless, the furrow ultimately resumed to
produce a binucleate cell (Fig. 5, DAPI), suggesting that
E18 specifically inhibits the terminal phase of cytokinesis.
The effect of E18 was specific because no major inhibition
of cell division was observed in cells treated with injection
buffer alone, commercially available nonspecific rabbit
IgG, and protein A–purified E18 preimmune antibodies
(Table I). These results contrast with those of Nislow et al.
(1990) and Wright et al. (1993) who observed mitotic ar-
rest in mAb-injected cells. The difference may be attrib-
uted to the specificity of the antibodies injected into cells
(mouse monoclonal IgM recognizing the central stalk ver-
sus rabbit E18 IgG specific to the tail). The role of
MKLP1/CHO1 in early stages of mitosis is left unsolved.
Initiation and ingression of cleavage furrows proceeded
normally in E18-injected cells, suggesting that the actin-
interacting site of CHO1 may not be involved in the for-
mation of contractile ring and the process of cell cleavage.
Rather, the protein may function to link the microtubule-
containing central spindle to the cortex/membrane at the
cell equator during the late stage of cell division. Time-
lapse analysis of dividing cells suggested that the midbody
matrix could be a structure holding microtubules and the
cell cortex/membrane together after the cessation of the
furrowing and the disassembly of the contractile ring
(Mullins and Biesele, 1977). Both the motor activity and
microtubule-bundling capacity of CHO1 are required for
completion of cytokinesis by organizing midzone microtu-
bules and the electron-dense matrix in the center of the in-
tercellular bridge (Matuliene and Kuriyama, 2002). Old
and new observations clearly indicate that complete separa-
tion of two daughter cells is achieved through stretching
and breaking the intercellular bridge at the narrowest re-
gion flanking the midbody center (Mullins and Biesele,
1977; Piel et al., 2001). Since microtubules derived from
the remnant of the central spindle are still connected to
each daughter cell, the microtubule bundle forming a core
of intercellular bridge must be under tension between two
cells. Therefore, for successful separation of daughter cells
Figure 4. Interaction of GFP–exon 18
protein with F-actin. (A–C) CHO cells
expressing the GFP-tagged exon 18
polypeptide were stained with
Alexa-conjugated phalloidin (A and B)
and -tubulin antibodies (C). In B, the
cells were treated with 5 g/ml
dihydrocytochalasin B (DHCB) for 30
min to depolymerize F-actin before
fixation. Bar, 10 m. (D) The proteins
expressed in CHO (lanes 1–4), Sf9 cells
(lanes 5–8), and reticulocyte lysates
(lanes 9–16) were mixed with () or
without () F-actin. Lanes 11, 12, 15,
and 16 include
35
S-labeled luciferase
prepared as a control protein. GFP–exon
18 bands in supernatant (S) and pellet (P)
fractions were visualized by either
immunostaining with anti-GFP antibodies
(lanes 1–8) or autoradiography (lanes
9–16). Note GFP–exon 18 prepared in
the reticulocyte lysate (lane 10) was
more easily sedimentable than that
expressed in CHO (lane 2) and Sf9 cells
(lane 6) without incubation with F-actin.
788 The Journal of Cell Biology
|
Volume 156, Number 5, 2002
Figure 5. Microinjection of E18 antibodies into PtK
1
cells. Numbers at the corner are the time after antibody injection. After fixation with
cold methanol, the cells were stained with mAb (probed by FITC-conjugated secondary antibodies, but its color was converted from green to
red), DAPI, and anti-rabbit IgG secondary antibody (E18). The coverslip was further stained with anti–-tubulin and FITC-conjugated secondary
antibodies (MTs/mAb). Arrows indicate the position of a midbody formed between two separated nuclei. Bar, 50 m.
Table I.
Effects of antibody injection into mitotic PtK
1
cells
Cells counted Normal cell division Incomplete cell division
a
Two cells appositioned
b
Arrested/no changes
c
(%)
E18
d
73 6(8) 41(56) 22(30) 4(5)
E18-pre
e
29 26(90) 0(0) 1(3) 2(7)
Control
f
41 35(85) 5(12) 0(0) 1(2)
Injection buffer
g
12 9(75) 0(0) 0(0) 3(25)
a
Although the cells underwent normal chromosome segregation and cytokinesis, regression of the cleavage furrow and/or cell fusion resulted in the forma-
tion of multinucleate cells.
b
Two almost completely separated cells were closely aligned side-by-side. A part of the cell boundary was unclear, and the two cells appeared to be par-
tially connected to each other.
c
The cells either arrested at mitosis or entered interphase without cell division.
d
5–13 mg/ml affinity purified E18 rabbit immunoglobulin molecules were prepared in injection buffer and introduced into PtK
1
cells at prophase to
anaphase. The cells were fixed at 2–6 h after antibody injection.
e
Protein A–purified preimmune E18 antibodies.
f
Two different batches of rabbit IgG fractions, which are commercially available.
g
Injection buffer alone.
CHO1 isoform with an actin-interacting domain |
Kuriyama et al. 789
the microtubule rope must be tightly attached to the mem-
brane/cortex during this tug-of-war. It might be CHO1
that is responsible for connection between the midbody
matrix and cell membrane.
Alternatively, the motor protein could be involved in
membrane events necessary for completion of cytokinesis.
It is widely believed that targeted secretion, insertion, and
fusion of membrane vesicles play a central role in progres-
sion and completion of cleavage furrows (for review see
Straight and Field, 2000). Thus, cells lacking syntaxins and
their associated proteins (Lukowitz et al., 1996; Heese et
al., 2001), a phospholipid kinase (Brill et al., 2000), dy-
namin (Gu and Verma, 1996), or Golgi-associated pro-
teins (Sisson et al., 2000), are defective in cytokinesis in a
variety of cells. Rabkinesin-6/Rab6-KIFL, a Rab6-binding
kinesin-like protein, is required for not only membrane
traffic but also cytokinesis (Hill et al., 2000). Of particular
interest is that this motor protein shares the highest degree
of sequence identity to CHO1 (Echard et al., 1998). Skop
et al. (2001) have reported recently that brefeldin, a potent
inhibitor of vesicle secretion by targeting a small GTPase-
binding protein Arf, specifically blocks the terminal phase
of cytokinesis by regressing ingressed furrows in
C. elegans
.
The phenotype detected in brefeldin-treated embryos is re-
markably similar to what we saw in E18-injected mamma-
lian cells. Importantly, CHO1/MKLP1 is capable of bind-
ing Arfs through the sequence encoded by exons 19–22,
just downstream the actin-interacting domain of exon 18
(Boman et al., 1999). Since dividing cells contain dynamic
membrane phospholipids tightly coupled with the actin
cytoskeleton (Emoto and Umeda, 2000) and Arfs are be-
lieved to be involved in both membrane traffic and actin
dynamics, it is likely that the actin- and Arf-interacting do-
mains act in concert to achieve the CHO1 function during
the late stage of cytokinesis. Further analysis of CHO1
would be of great benefit for our understanding of the
mechanism and regulation of cell division.
Materials and methods
Cloning of cDNA and genomic sequences of
chicken CHO1 and MKLP1
cDNA encoding chicken CHO1- and MKLP1-type of motor proteins were
cloned by screening of a chicken cDNA library (Stratagene) with CHO1
probes derived from CHO cells (Kuriyama et al., 1994). To extend the 5
sequence, mRNA was purified from cultured chicken lymphocytes (DT40
cells) and used for 5
-RACE with specific and degenerate primers matching
the conserved ATP-binding consensus motif (amino acids 118–124). For
cloning of genomic DNA, 10 primers (nucleotide positions 14
42, 3891
3927, 3891
3927, 7031
7073, 6294
6328, 9635
9667, 9594
9633, 13863
13902, 13851
13892, and 17097
17138) were
designed based on the nucleotide sequence of CHO1/MKLP1, and PCR
was performed with DT40 genomic templates. Five fragments in a size be-
tween 3.2 and 4.3 kb were cloned and assembled after nucleotide se-
quence analysis.
Cell culture, protein expression, and immunostaining
CHO and HeLa cells were cultured in 10% FCS containing Ham’s F-10
and DME medium, respectively. The exon 18 coding sequence (amino
acid positions 696–786) was isolated by digestion of the full-length CHO-
CHO1 with SspI and Eam1104 I and ligated into the eukaryotic expression
vector, pEGFP-C1 (CLONTECH Laboratories, Inc.). CHO cells on a cover-
slip in a 35-mm dish were transfected by addition of 0.6–2
g/ml purified
plasmid DNA and cultured overnight (Matuliene and Kuriyama, 2002). To
enrich mitotic cell populations, cells were partially synchronized by treat-
ment with thymidine followed by accumulation at M phase by addition of
nocodazole at a final concentration of 0.05
g/ml (Sellitto and Kuriyama,
1988). After washing out the drug, cells were allowed to recover for 20–50
min before fixation with cold methanol.
For immunofluorescence staining, cells were rehydrated with 0.05%
Tween-20 containing PBS and incubated with primary and secondary anti-
bodies. Primary antibodies include monoclonal anti–
-tubulin antibodies
(Sigma-Aldrich), monoclonal CHO1 (mAb [Sellitto and Kuriyama, 1988]),
and rabbit polyclonal antibodies (E18) raised against 23 amino acids in
exon 18 at amino acid positions 755–777 of chicken CHO1 (Fig. 2, double
underlined). Immunoblotting analysis was done as described before
(Kuriyama et al., 1994).
In vitro cosedimentation with F-actin
Actin was prepared from rabbit skeletal muscles (a gift from Drs. Albina
Orlova and Ewa Prochniewicz, University of Minnesota, Minneapolis,
MN) (Orlova et al., 1995), and platelet actin was purchased from Cytoskel-
eton, Inc. G-actin was polymerized and sedimented in a medium contain-
ing 5 mM Tris-HCl, pH 7.8, 0.2 mM ATP, 0.1 mM CaCl
2,
150 mM KCl,
and 1 mM MgCl
2
. For protein expression in insect cells, cDNA encoding
GFP–exon 18 was subloned into pVL1392 (PharMingen) and used for in-
fection of Sf9 cells as before (Kuriyama et al., 1994).
35
S-labeled GFP–exon
18 was synthesized using a commercially available kit (TNT Coupled
Reticulocyte Lysate System; Promega). CHO and Sf9 cells expressing exog-
enous proteins were washed once with PBS and lysed for 30–60 min at
0
C in a medium containing 10 mM Tris-HCl, pH 7.8, 0.5% Nonidet NP-
40, and protease inhibitors. Cell extracts and reticulocyte lysates recovered
after centrifugation at 200,000
g
for 30 min were mixed with F-actin and
further incubated for 30 min on ice. After layering on a 20% sucrose cush-
ion, F-actin plus associated proteins were sedimented at 100,000
g
for 30
min at 4
C.
Antibody injection
Affinity purified E18 was prepared in injection buffer (140 mM KCl, 100
mM glutamic acid, 40 mM citric acid, 1 mM MgCl
2
, 1 mM EGTA, pH 7.4)
and injected into PtK
1
cells cultured on a photoetched coverslip (Bellco).
Microinjection was performed using a Narishige microinjector attached to
a Nikon Diaphot inverted microscope, and time-lapse images were re-
corded with a Nikon Eclipse TE200 microscope using ImagePro software
packages.
This work was supported by National Institutes of Health grant GM55735
to R. Kuriyama.
Submitted: 24 September 2001
Revised: 18 January 2002
Accepted: 18 January 2002
References
Adams, R.R., A.A. Tavares, A. Salzberg, H.J. Bellen, and D.M. Glover. 1998.
pa-
varotti
encodes a kinesin-like protein required to organize the central spindle
and contractile ring for cytokinesis.
Genes Dev.
12:1483–1494.
Boman, A.L., J. Kuai, X. Zhu, J. Chen, R. Kuriyama, and R.A. Kahn. 1999. Arf
proteins bind to mitotic kinesin-like protein 1 (MKLP1) in a GTP-depen-
dent fashion.
Cell Motil. Cytoskeleton.
44:119–132.
Brill, J.A., G.R. Hime, M. Scharer-Schuksz, and M.T. Fuller. 2000. A phospho-
lipid kinase regulates actin organization and intercellular bridge formation
during germline cytokinesis.
Development.
127:3855–3864.
Chui, K.K., G.C. Rogers, A.M. Kashina, K.P. Wedaman, D.J. Sharp, D.T.
Nguyen, F. Wilt, and J.M. Scholey. 2000. Roles of two homotetrameric ki-
nesins in sea urchin embryonic cell division.
J. Biol. Chem.
275:38005–
38011.
Echard, A., F. Jollivet, O. Martinez, J.-J. Lacapere, A. Rousselet, I. Janoueix-Lero-
sey, and B. Gould. 1998. Interaction of a Golgi-associated kinesin-like pro-
tein with Rab6.
Science.
279:580–585.
Emoto, K., and M. Umeda. 2000. An essential role for a membrane lipid in cyto-
kinesis: regulation of contractile ring disassembly by redistribution of phos-
phatidylethanolamine.
J. Cell Biol.
149:1215–1224.
Gu, X., and D.P. Verma. 1996. Phragmoplastin, a dynamin-like protein associated
with cell plate formation in plants.
EMBO J.
15:695–704.
Heese, M., X. Gansel, L. Sticher, P. Wick, M. Grebe, F. Granier, and G. Jurgens.
2001. Functional characterization of the KNOLLE-interacting t-SNARE
AtSNAP33 and its role in plant cytokinesis.
J. Cell Biol.
155:239–249.
790 The Journal of Cell Biology
|
Volume 156, Number 5, 2002
Hill, E., M. Clarke, and F.A. Barr. 2000. The Rab6-binding kinesin, Rab6-KIFL,
is required for cytokinesis.
EMBO J.
19:5711–5719.
Jantsch-Plunger, V., P. Gonczy, A. Romano, H. Schnabel, D. Hamill, R. Schnabel,
A.A. Hyman, and M. Glotzer. 2000. CYK-4: a Rho family GTPase activat-
ing protein (GAP) required for central spindle formation and cytokinesis.
J.
Cell Biol.
149:1391–1404.
Kuriyama, R., S. Dragas-Granoic, T. Maekawa, A. Vassilev, A. Khodjakov, and H.
Kobayashi. 1994. Heterogeneity and microtubule interaction of the CHO1
antigen, a mitosis-specific kinesin-like protein. Analysis of subdomains ex-
pressed in insect Sf9 cells.
J. Cell Sci.
107:3485–3499.
Lukowitz, W., U. Mayer, and G. Jurgens. 1996. Cytokinesis in the
Arabidopsis
em-
bryo involves the syntaxin-related KNOLLE gene product.
Cell.
84:61–71.
Matuliene, J., and R. Kuriyama. 2002. Kinesin-like motor protein CHO1 is re-
quired for the formation of midbody matrix and the completion of cytokine-
sis in mammalian cells.
Mol. Biol. Cell.
In press.
Miki, H., M. Setou, K. Kaneshiro, and N. Hirokawa. 2001. All kinesin superfam-
ily protein, KIF, genes in mouse and human.
Proc. Natl. Acad. Sci. USA.
98:
7004–7011.
Mullins, J.M., and J.J. Biesele. 1977. Terminal phase of cytokinesis in D-98s cells.
J. Cell Biol.
73:672–684.
Nislow, C., C. Sellitto, R. Kuriyama, and J.R. McIntosh. 1990. A monoclonal an-
tibody to a mitotic microtubule-associated protein blocks mitotic progres-
sion.
J. Cell Biol.
111:511–522.
Nislow, C., V.A. Lombillo, R. Kuriyama, and J.R. McIntosh. 1992. A plus-end-
directed motor enzyme that moves antiparallel microtubules in vitro localizes
to the interzone of mitotic spindles. Nature. 359:543–547.
Orlova, A., E. Prochniewicz, and E.H. Egelman. 1995. Structural dynamics of
F-actin: II. Cooperativity in structural transitions. J. Mol. Biol. 245:598–
607.
Piel, M., J. Nordberg, U. Euteneuer, and M. Bornens. 2001. Centrosome-depen-
dent exit of cytokinesis in animal cells. Science. 291:1550–1553.
Powers, J., O. Bossinger, D. Rose, S. Strome, and W. Saxton. 1998. A nematode
kinesin required for cleavage furrow advancement. Curr. Biol. 8:1133–1136.
Raich, W.B., A.N. Moran, J.H. Rothman, and J. Hardin. 1998. Cytokinesis and
midzone microtubule organization in Caenorhabditis elegans require the ki-
nesin-like protein ZEN-4. Mol. Biol. Cell. 9:2037–2049.
Sellitto, C., and R. Kuriyama. 1988. Distribution of a matrix component of the
midbody during the cell cycle in Chinese hamster ovary cells. J. Cell Biol.
106:431–439.
Schumacher, J.M., A. Golden, and P.J. Donovan. 1998. AIR-2: an aurora/Ipl1-
related protein kinase associated with chromosomes and midbody microtu-
bules is required for polar body extrusion and cytokinesis in Caenorhabditis
elegans embryos. J. Cell Biol. 143:1635–1646.
Severson, A.F., D.R. Hamill, J.C. Carter, J. Schumacher, and B. Bowerman. 2001.
The Aurora-related kinase AIR-2 recruits ZEN-4/CeMKLP1 to the mitotic
spindle at metaphase and is required for cytokinesis. Curr. Biol. 10:1162–
1171.
Sisson, J.C., C. Field, R. Ventura, A. Royou, and W. Sullivan. 2000. Lava Lamp, a
novel peripheral Golgi protein, is required for Drosophila melanogaster cellu-
larization. J. Cell Biol. 151:905–917.
Skop, A.R., D. Bergmann, W.A. Mohler, and J.G. White. 2001. Completion of
cytokinesis in C. elegans requires a brefeldin A-sensitive membrane accumu-
lation at the cleavage furrow apex. Curr. Biol. 11:735–746.
Straight, A.F., and C.M. Field. 2000. Microtubules, membranes and cytokinesis.
Curr. Biol. 10:R760–R770.
Wright, B.D., M. Terasaki, and J.M. Scholey. 1993. Roles of kinesin and kinesin-
like proteins in sea urchin embryonic cell division: evaluation using antibody
microinjection. J. Cell Biol. 123:681–689.
... CHOI and MKLPl are two simultaneously co-expressed isoforms of the same gene. Full-length CHOI contains the alternatively spliced F-actin-binding exon 18[234] and has an essential role in the organisation of the midbody matrix and in the completion of cytokinesis[278].The staining pattern o f annexin A ll during cell division bears striking similaritiesto that reported for CHOI[278]. To investigate a possible relationship between these two proteins, we compared the localisation of annexin A ll with that of CHOI during the cell cycle, using an antibody specific for the full-length CHOI isoform[234] (Figure 5.12).Both annexin A ll and CHOI are predominantly nuclear in interphase andprophase cells, and excluded from the nucleoli. ...
... Full-length CHOI contains the alternatively spliced F-actin-binding exon 18[234] and has an essential role in the organisation of the midbody matrix and in the completion of cytokinesis[278].The staining pattern o f annexin A ll during cell division bears striking similaritiesto that reported for CHOI[278]. To investigate a possible relationship between these two proteins, we compared the localisation of annexin A ll with that of CHOI during the cell cycle, using an antibody specific for the full-length CHOI isoform[234] (Figure 5.12).Both annexin A ll and CHOI are predominantly nuclear in interphase andprophase cells, and excluded from the nucleoli. In metaphase, CHOI localised along the microtubular spindle, while annexin A ll was initially distributed throughout the cell body, although with greater intensity along the spindle. ...
... The fusion process would be spatially constrained to the furrow by the regulatory action of molecules such as the septins, capable of interacting with the Sec6/8 or exocyst complex[261,267].After actomyosin ring disassembly, the plasma membrane must be tightly attached to the midbody for abscission to occur. CHOI has previously been suggested to be involved in establishing this connection[234], but it is not known how this kinesin is capable to bind to the cell membrane. The annexins are known to be Ca^^-dependent membrane-binding proteins[4]. ...
Thesis
Annexin A11 is a ubiquitously expressed member of the annexin family of Ca2+-dependent phospholipid binding proteins, which lies at the root of the vertebrate annexin evolutionary tree. Annexin A11 is known to be localised to the nuclei of cells in culture, and to interact Ca2+-dependently in vitro with the S100 protein S100A6 and the penta EF-hand protein ALG-2 through its long glycine, proline and tyrosine-rich N-terminal domain, but very little is known about the physiological functions performed by this annexin in vivo. An investigation into the cellular roles of annexin A11 both in interphase and during cell division is presented in this thesis. Using various molecular and cell biology approaches, annexin A11 was identified in the nucleus and in cytoplasmic vesicles that appear to be transported along the microtubular network, linking annexin A11 function with intracellular membrane trafficking processes. Annexin A11 has also been identified in membrane domains involved in signal transduction at the basolateral membrane of A431 cells. A role for this annexin in membrane remodelling steps during cell growth is proposed in view of the accumulation of annexin A11-containing vesicles in the cytoplasm of non-contact inhibited cells and at the leading edge of monolayer outgrowths, together with the concentration of annexin A11 at the cell-cell contact sites in the plasma membrane of confluent monolayers. Annexin A11 relocates to the nuclear envelope and is tyrosine-phosphorylated in the presence of calcium in interphase cells, and invades the nuclear envelope at the invaginations created during nuclear envelope breakdown at the onset of mitosis, and is also detected at the nuclear envelope during nuclear envelope reassembly after cell division, making the nuclear envelope a specific target for annexin A11. The study of the function of annexin A11 during cell division led to the identification of an essential role for annexin A11 in the terminal phase of cytokinesis. Annexin 11 was observed to translocate from the nucleus to the spindle poles in metaphase, and then to the spindle midzone in anaphase, and to be recruited to the midbody in late telophase where it co-localises and associates with the mitotic kinesin- like protein CHO1. Depletion of annexin A11 by RNA interference has shown that the absence of this annexin leads to failure to establish a functional midbody, incomplete daughter cell separation, and ultimately cell death by apoptosis.
... 3′-sequence of this transcript is not complete (Ensembl release 100, April 2020) and lacks exons 17 to 23 making impossible to conclude whether x8 is part of Kif23-Δ17-18 detected in this study. The functional heterogeneity of KIF23 (MKLP1/CHO1) due to alternative splicing was earlier demonstrated by interaction of F-actin with the sequence encoded by exon 18 (KIF23-FL) implying association with both microtubules and actin cytoskeletons [16]. Moreover, it was shown that the antibodies specific for exon 18 did affect only the terminal phase of cytokinesis resulting in bi-nucleated cells, the exact result seen in HeLa cells in rescue experiments with KIF23 p.P916R [3]. ...
... Dashed lines show conventional splicing and solid lines show alternative splicing. Exons are shown in white (exons7,8,16,18,18,19) and alternative exon 8 (x8) is shown in grey. The primer sequences are provided in Supplemental Material S1. ...
Article
Full-text available
Kinesin Family Member 23 (KIF23), a cell cycle regulator, has a key task in cytokinesis. KIF23 over-expression in cancer has been associated with tumor growth, progression, and poor prognosis, indicating a potential to be a cancer biomarker. A mutation in KIF23 (c.2747C > G, p.P916R) was shown to cause congenital dyserythropoietic anemia, type III (CDA III). To-date, fifteen KIF23 transcripts have been annotated, but their expression is poorly investigated. We hypothesized that tissue specific expression of a particular transcript can be critical for CDA III phenotype. In this study, we quantified expression of alternative Kif23 transcripts in a mouse model with human KIF23 mutation and investigated its association with a regulator of alternative splicing, serine/arginine-rich splicing factor 3 (Srsf3). We confirmed presence of an additional exon 8 in both human and mouse KIF23 transcripts. A transcript lacking exons 17 and 18 was ubiquitously expressed in mice while other isoforms were common in human tissues however in bone marrow of knock-in mice a transcript without exon 18 was prevalent as it was in bone marrow of a CDA III patient. We conclude that the possibility that the tissue specific expression of KIF23 alternative transcripts influence the CDA III phenotype cannot be neglected.
... PRR14L may be one such hit from our screen, where it ranked 7 th among enhancers (S2 Table). Little is known about the molecular function of the PRR14L protein, but it was reported to localize at the cytokinetic midbody in HEK293T cells and interact with KIF4A and KIF23/MKLP1, two other midbody proteins that function in cytokinesis [31][32][33][34]. KIF4A ranked 87 th among enhancers of PLK1 inhibition in our screen (S2 Table). ...
Article
Full-text available
Polo-like kinase 1 (PLK1) is a serine/threonine kinase required for mitosis and cytokinesis. As cancer cells are often hypersensitive to partial PLK1 inactivation, chemical inhibitors of PLK1 have been developed and tested in clinical trials. However, these small molecule inhibitors alone are not completely effective. PLK1 promotes numerous molecular and cellular events in the cell division cycle and it is unclear which of these events most crucially depend on PLK1 activity. We used a CRISPR-based genome-wide screening strategy to identify genes whose inactivation enhances cell proliferation defects upon partial chemical inhibition of PLK1. Genes identified encode proteins that are functionally linked to PLK1 in multiple ways, most notably factors that promote centromere and kinetochore function. Loss of the kinesin KIF18A or the outer kinetochore protein SKA1 in PLK1-compromised cells resulted in mitotic defects, activation of the spindle assembly checkpoint and nuclear reassembly defects. We also show that PLK1-dependent CENP-A loading at centromeres is extremely sensitive to partial PLK1 inhibition. Our results suggest that partial inhibition of PLK1 compromises the integrity and function of the centromere/kinetochore complex, rendering cells hypersensitive to different kinetochore perturbations. We propose that KIF18A is a promising target for combinatorial therapies with PLK1 inhibitors.
... For example, KIF23, a member of the kinesin-6 family, was also known as CHO1/MKLP1 (human), ZEN-4/CeMKLP1 (C. elegans), and Pavarotti/MKLP1 (Pav-KLP, Drosophila) (Kuriyama et al., 2002). KIFs appear to be present in all eukaryotes, from fungi to plants and animals, as well as in different cell types. ...
Article
Full-text available
In mammalian testes, the apical cytoplasm of each Sertoli cell holds up to several dozens of germ cells, especially spermatids that are transported up and down the seminiferous epithelium. The blood-testis barrier (BTB) established by neighboring Sertoli cells in the basal compartment restructures on a regular basis to allow preleptotene/leptotene spermatocytes to pass through. The timely transfer of germ cells and other cellular organelles such as residual bodies, phagosomes, and lysosomes across the epithelium to facilitate spermatogenesis is important and requires the microtubule-based cytoskeleton in Sertoli cells. Kinesins, a superfamily of the microtubule-dependent motor proteins, are abundantly and preferentially expressed in the testis, but their functions are poorly understood. This review summarizes recent findings on kinesins in mammalian spermatogenesis, highlighting their potential role in germ cell traversing through the BTB and the remodeling of Sertoli cell-spermatid junctions to advance spermatid transport. The possibility of kinesins acting as a mediator and/or synchronizer for cell cycle progression, germ cell transit, and junctional rearrangement and turnover is also discussed. We mostly cover findings in rodents, but we also make special remarks regarding humans. We anticipate that this information will provide a framework for future research in the field.
... Nislow first identified a human KIF23 homolog in 1992 (13), which was a plus-end-directed motor enzyme that translocates antiparallel microtubules in vitro. Following this, other reports confirmed that KIF23 as a key regulator of cytokinesis (12,(36)(37)(38)(39)(40). By screening a HeLa cell cDNA library, KIF23 (also known as mitotic kinesin-like protein-1, MKLP1) was separated and confirmed as a plus-end-directed motor enzyme that translocates antiparallel microtubules in vitro (41). ...
Article
Full-text available
Background: In recent research, high expression of kinesin family member 23 (KIF23), one of the kinesin motor proteins involved in the regulation of cytokinesis, has been shown to be related to poor prognosis in glioma and paclitaxel-resistant gastric cancer, as a results of the enhancement of proliferation, migration, and invasion. In this study, we analyzed the role of KIF23 in the progression of pancreatic ductal adenocarcinoma. Methods: A bioinformatic method was used to analyze the KIF23 mRNA level in pancreatic tumor tissues compared with normal pancreatic tissues and to analyze the connection between high KIF23 expression and prognosis. We examined the expression of KIF23 using immunohistochemistry and analyzed the connection between the expression of KIF23 and clinicopathological features in pancreatic ductal adenocarcinoma patients. In addition, a colony formation assay, MTT assay, and western blot assay were performed in vitro, along with a mouse xenograft model in vivo, to analyze the effect of KIF23 on proliferation. Further, the correlation between KIF23 and CDCA8 was analyzed by TCGA and immunohistochemical data. Results: Bioinformatic results showed that KIF23 mRNA expression was higher in pancreatic tumor tissues than in normal pancreatic tissues and a poor prognosis has been linked to the high expression of KIF23. Immunohistochemistry revealed that KIF23 was highly expressed at the protein level and high expression of KIF23 correlated with adverse clinicopathological features. Our experimental results demonstrated that knockdown of KIF23 could inhibit the proliferation of pancreatic cells. Further, a positive correlation between KIF23 and CDCA8 expression existed, and KIF23 might promote pancreatic cancer proliferation by affecting CDCA8 expression. Conclusions: Our data showed that high expression of KIF23 is associated with a poor prognosis, and KIF23 might be a potential therapeutic target for pancreatic ductal adenocarcinoma.
Article
Full-text available
Long ignored as a vestigial remnant of cytokinesis, the mammalian midbody (MB) is released post-abscission inside large extracellular vesicles called MB remnants (MBRs). Recent evidence suggests that MBRs can modulate cell proliferation and cell fate decisions. Here, we demonstrate that the MB matrix is the site of ribonucleoprotein assembly and is enriched in mRNAs that encode proteins involved in cell fate, oncogenesis, and pluripotency, which we are calling the MB granule. Both MBs and post-abscission MBRs are sites of spatiotemporally regulated translation, which is initiated when nascent daughter cells re-enter G1 and continues after extracellular release. MKLP1 and ARC are necessary for the localization and translation of RNA in the MB dark zone, whereas ESCRT-III is necessary to maintain translation levels in the MB. Our work reveals a unique translation event that occurs during abscission and within a large extracellular vesicle.
Preprint
Full-text available
The midbody (MB) is a transient structure at the spindle midzone that is required for cytokinesis, the terminal stage of cell division. Long ignored as a vestigial remnant of cytokinesis, we now know MBs are released post-abscission as extracellular vesicles called MB remnants (MBRs) and can modulate cell proliferation, fate decisions, tissue polarity, neuronal architecture, and tumorigenic behavior. Here, we demonstrate that the MB matrix—the structurally amorphous MB core of unknown composition—is the site of ribonucleoprotein assembly and is enriched in mRNAs that encode proteins involved in cell fate, oncogenesis, and pluripotency, that we are calling the MB granule. Using a quantitative transcriptomic approach, we identified a population of mRNAs enriched in mitotic MBs and confirmed their presence in signaling MBR vesicles released by abscission. The MB granule is unique in that it is translationally active, contains both small and large ribosomal subunits, and has both membrane-less and membrane-bound states. Both MBs and post-abscission MBRs are sites of spatiotemporally regulated translation, which is initiated when nascent daughter cells re-enter G1 and continues after extracellular release. We demonstrate that the MB is the assembly site of an RNP granule. MKLP1 and ARC are necessary for the localization and translation of RNA in the MB dark zone, whereas ESCRT-III was necessary to maintain translation levels in the MB. Our data suggest a model in which the MB functions as a novel RNA-based organelle with a uniquely complex life cycle. We present a model in which the assembly and transfer of RNP complexes are central to post-mitotic MBR function and suggest the MBR serves as a novel mode of RNA-based intercellular communication with a defined biogenesis that is coupled to abscission, and inherently links cell division status with signaling capacity. To our knowledge, this is the first example of an autonomous extracellular vesicle with active translation activity.
Preprint
Full-text available
The midbody (MB) is a transient structure at the spindle midzone that is required for cytokinesis, the terminal stage of cell division. Long ignored as a vestigial remnant of cytokinesis, we now know MBs are released post-abscission as extracellular vesicles called MB remnants (MBRs) and can modulate cell proliferation, fate decisions, tissue polarity, neuronal architecture, and tumorigenic behavior. Here, we demonstrate that the MB matrix—the structurally amorphous MB core of unknown composition—is the site of ribonucleoprotein assembly and is enriched in mRNAs that encode proteins involved in cell fate, oncogenesis, and pluripotency, that we are calling the MB granule. Using a quantitative transcriptomic approach, we identified a population of mRNAs enriched in mitotic MBs and confirmed their presence in signaling MBR vesicles released by abscission. The MB granule is unique in that it is translationally active, contains both small and large ribosomal subunits, and has both membrane-less and membrane-bound states. Both MBs and post-abscission MBRs are sites of spatiotemporally regulated translation, which is initiated when nascent daughter cells re-enter G1 and continues after extracellular release. We demonstrate that the MB is the assembly site of an RNP granule. MKLP1 and ARC are necessary for the localization and translation of RNA in the MB dark zone, whereas ESCRT-III was necessary to maintain translation levels in the MB. Our data suggest a model in which the MB functions as a novel RNA-based organelle with a uniquely complex life cycle. We present a model in which the assembly and transfer of RNP complexes are central to post-mitotic MBR function and suggest the MBR serves as a novel mode of RNA-based intercellular communication with a defined biogenesis that is coupled to abscission, and inherently links cell division status with signaling capacity. To our knowledge, this is the first example of an autonomous extracellular vesicle with active translation activity. Highlights The MB, the center region of the intercellular bridge, is the assembly site of a ribonucleoprotein granule, we call the MB granule Distinct oncogenic and pluripotent transcription factor RNAs, including Jun/Fos and KLF4 , are packaged in MBs and MBRs The MB granule is coincident with the MB matrix, or dark zone, of the MB The Kif23/MKLP1 kinesin is a core hexanediol-sensitive MB granule component The MB and MBR are site of active translation that begins in early G1 and continues post-mitotically MKLP1 and ARC are necessary for RNA targeting/maintenance and translation at the MB Depletion of ESCRT-III increases the levels of translation during abscission Abscission releases MBRs as MB granule-harboring, translating extracellular vesicles Multiple cell types including cancer, stem, neural stem, all have actively translating MBRs MBRs are proposed as a novel mode of intercellular communication by extracellular vesicle-mediated direct transfer of RNA
Article
Full-text available
Kinesin motor proteins are responsible for orchestrating a variety of microtubule-based processes including intracellular transport, cell division, cytoskeletal organization, and cilium function. Members of the kinesin-6 family play critical roles in anaphase and cytokinesis during cell division as well as in cargo transport and microtubule organization during interphase, however little is known about their motility properties. We find that truncated versions of MKLP1 (HsKIF23), MKLP2 (HsKIF20A), and HsKIF20B largely interact statically with microtubules as single molecules but can also undergo slow, processive motility, most prominently for MKLP2. In multi-motor assays, all kinesin-6 proteins were able to drive microtubule gliding and MKLP1 and KIF20B were also able to drive robust transport of both peroxisomes, a low-load cargo, and Golgi, a high-load cargo, in cells. In contrast, MKLP2 showed minimal transport of peroxisomes and was unable to drive Golgi dispersion. These results indicate that the three mammalian kinesin-6 motor proteins can undergo processive motility but differ in their ability to generate forces needed to drive cargo transport and microtubule organization in cells.
Article
Full-text available
Pavarotti, the Drosophila MKLP1 orthologue, is a kinesin-like protein that works with Tumbleweed (MgcRacGAP) as the centralspindlin complex. This complex is essential for cytokinesis, where it helps to organize the contractile actomyosin ring at the equator of dividing cells by activating the RhoGEF Pebble. Actomyosin rings also function as the driving force during cell wound repair. We previously showed that Tumbleweed and Pebble are required for the cell wound repair process. Here, we show that Pavarotti also functions during wound repair and confirm that while Pavarotti, Tumbleweed, and Pebble are all used during this cellular repair, each has a unique localization pattern and knockdown phenotype, demonstrating centralspindlin-independent functions. Surprisingly, we find that the classically microtubule-associated Pavarotti binds directly to actin in vitro and in vivo and has a noncanonical role directly regulating actin dynamics. Finally, we demonstrate that this actin regulation by Pavarotti is not specific to cellular wound repair but is also used in normal development.
Article
Full-text available
Arf proteins comprise a family of 21-kDa GTP-binding proteins with many proposed functions in mammalian cells, including the regulation of several steps of membrane transport, maintenance of organelle integrity, and activation of phospholipase D. We performed a yeast two-hybrid screen of human cDNA libraries using a dominant activating allele, [Q71L], of human Arf3 as bait. Eleven independent isolates contained plasmids encoding the C-terminal tail of mitotic kinesin-like protein-1 (MKLP1). Further deletion mapping allowed the identification of an 88 amino acid Arf3 binding domain in the C-terminus of MKLP1. This domain has no clear homology to other Arf binding proteins or to other proteins in the protein databases. The C-terminal domain of MKLP1 was expressed and purified from bacteria as a GST fusion protein and shown to bind Arf3 in a GTP-dependent fashion. A screen for mutations in Arf3 that specifically lost the ability to bind MKLP1 identified 10 of 14 point mutations in the GTP-sensitive switch I or switch II regions of Arf3. Two-hybrid assays of the C-terminal domain of MKLP1 with each of the human Arf isoforms revealed strong interaction with each. Taken together, these data are all supportive of the conclusion that activated Arf proteins bind to the C-terminal “tail” domain of MKLP1. Cell Motil. Cytoskeleton 44:119–132, 1999. © 1999 Wiley-Liss, Inc.
Article
Full-text available
During cytokinesis of animal cells, the mitotic spindle plays at least two roles. Initially, the spindle positions the contractile ring. Subsequently, the central spindle, which is composed of microtubule bundles that form during anaphase, promotes a late step in cytokinesis. How the central spindle assembles and functions in cytokinesis is poorly understood. The cyk-4 gene has been identified by genetic analysis in Caenorhabditis elegans. Embryos from cyk-4(t1689ts) mutant hermaphrodites initiate, but fail to complete, cytokinesis. These embryos also fail to assemble the central spindle. We show that the cyk-4 gene encodes a GTPase activating protein (GAP) for Rho family GTPases. CYK-4 activates GTP hydrolysis by RhoA, Rac1, and Cdc42 in vitro. RNA-mediated interference of RhoA, Rac1, and Cdc42 indicates that only RhoA is essential for cytokinesis and, thus, RhoA is the likely target of CYK-4 GAP activity for cytokinesis. CYK-4 and a CYK-4:GFP fusion protein localize to the central spindle and persist at cell division remnants. CYK-4 localization is dependent on the kinesin-like protein ZEN-4/CeMKLP1 and vice versa. These data suggest that CYK-4 and ZEN-4/CeMKLP1 cooperate in central spindle assembly. Central spindle localization of CYK-4 could accelerate GTP hydrolysis by RhoA, thereby allowing contractile ring disassembly and completion of cytokinesis.
Article
Full-text available
The events leading to the completion of cytokinesis after the formation of the midbody and intercellular bridge in D-98S cells were studied with light and electron microscopy. Pairs of daughter cells corresponding to different stages of cytokineses, as determined previously form time lapse films, were selected from embedded monolayers for serial sectioning. Separation of daughter cells is preceded by the reduction in diameter of the intercellular bridge from 1-1.5 mum to approx. 0.2 mum. Two processes contribute to this reduction: (a) The intercellular bridge becomes gradually thinner after telophase; a progressive breakdown of midbody structures accompanies this change; and (b) the more significant contribution to reduction in bridge diameter occurs through the localized constriction of a segment of the intercellular bridge.. The microtubules within the constricted portion of the bridge are forced closer together, and some microtubules disappear as this narrowing progresses. The plasma membrane over the narrowed segments is thrown into a series of wavelike ripples. Separation of daughter cells is achieved through movements of the cells which stretch and break the diameter-reduced bridge. The midbody is discarded after separation and begins to deteriorate. Occasional pairs of daughter cells were found in which incomplete karyokineses resulted in their nuclei being connected by a strand of nuclear material traversing the bridge and midbody. Such cells do not complete cytokinesis but merge together several hours after telophase. This merging of daughter cells coincides with the nearly complete breakdown of the midbody.
Article
Full-text available
A monoclonal antibody raised against mitotic spindles isolated from CHO cells ([CHO1], Sellitto, C., and R. Kuriyama. 1988. J. Cell Biol. 106:431-439) identifies an epitope that resides on polypeptides of 95 and 105 kD and is localized in the spindles of diverse organisms. The antigen is distributed throughout the spindle at metaphase but becomes concentrated in a progressively narrower zone on either side of the spindle midplane as anaphase progresses. Microinjection of CHO1, either as an ascites fluid or as purified IgM, results in mitotic inhibition in a stage-specific and dose-dependent manner. Parallel control injections with nonimmune IgMs do not yield significant mitotic inhibition. Immunofluorescence analysis of injected cells reveals that those which complete mitosis display normal localization of CHO1, whereas arrested cells show no specific localization of the CHO1 antigen within the spindle. Immunoelectron microscopic images of such arrested cells indicate aberrant microtubule organization. The CHO1 antigen in HeLa cell extracts copurifies with taxol-stabilized microtubules. Neither of the polypeptides bearing the antigen is extracted from microtubules by ATP or GTP, but both are approximately 60% extracted with 0.5 M NaCl. Sucrose gradient analysis reveals that the antigens sediment at approximately 11S. The CHO 1 antigen appears to be a novel mitotic MAP whose proper distribution within the spindle is required for mitosis. The properties of the antigen(s) suggest that the corresponding protein(s) are part of the mechanism that holds the antiparallel microtubules of the two interdigitating half spindles together during anaphase.
Article
Full-text available
Monoclonal antibodies were raised against isolated spindles of CHO (Chinese hamster ovary) cells to probe for molecular components specific to the mitotic apparatus. One of the antibodies, CHO1, recognized an antigen localized to the midbody during mitosis. Immunofluorescence staining of metaphase cells showed that although the total spindle area was labeled faintly, the antigen corresponding to CHO1 was preferentially localized in the equatorial region of the spindle. With the progression of mitosis, the antigen was further organized into discrete short lines along the spindle axis, and eventually condensed into a bright fluorescent dot at the midzone of the intercellular bridge between two daughter cells. Parallel immunostaining of tubulin showed that the CHO1-stained area corresponded to the dark region where microtubules are entrapped by the amorphous dense matrix components and possibly blocked from binding to tubulin antibody. Immunoblot analysis indicated that CHO1 recognized two polypeptides of mol wt 95,000 and 105,000. The immunoreaction was always stronger in preparations of isolated midbodies than in mitotic spindle fractions. The protein doublet was retained in the particulate matrix fraction after Sarkosyl extraction (Mullins, J. M., and J. R. McIntosh. 1982. J. Cell Biol. 94:654-661), suggesting that CHO1 antigen is indeed a component of the dense matrix. In addition to the equatorial region of spindles and midbodies, CHO1 also stained interphase centrosomes, and nuclei in a speckled pattern that was cell cycle-dependent. Thus, the midbody appears to share either common molecular component(s) or a similar epitope with interphase centrosomes and nuclei.
Article
Full-text available
The CHO1 antigen is a mitosis-specific kinesin-like motor located at the interzonal region of the spindle. The human cDNA coding for the antigen contains a domain with sequence similarity to the motor domain of kinesin-like protein (Nislow et al., Nature 359, 543, 1992). Here we cloned cDNAs encoding the CHO1 antigen by immunoscreening of a CHO Uni-Zap expression library, the same species in which the original monoclonal antibody was raised. cDNAs of CHO cells encode a 953 amino acid polypeptide with a calculated molecular mass of 109 kDa. The N-terminal 73% of the antigen was 87% identical to the human clone, whereas the remaining 27% of the coding region showed only 48% homology. Insect Sf9 cells infected with baculovirus containing the full-length insert produced 105 and 95 kDa polypeptides, the same doublet identified as the original antigen in CHO cells. Truncated polypeptides corresponding to the N-terminal motor and C-terminal tail produced a 56 and 54 kDa polypeptide in Sf9 cells, respectively. Full and N-terminal proteins co-sedimented with, and caused bundling of, brain microtubules in vitro, whereas the C-terminal polypeptide did not. Cells expressing the N terminus formed one or more cytoplasmic processes. Immunofluorescence as well as electron microscopic observations revealed the presence of thick bundles of microtubules, which were closely packed, forming a marginal ring just beneath the cell membrane and a core in the processes. The diffusion coefficient and sedimentation coefficient were determined for the native CHO1 antigen by gel filtration and sucrose density gradient centrifugation, respectively.(ABSTRACT TRUNCATED AT 250 WORDS)
Article
As an organelle coupling nuclear and cytoplasmic divisions, the centrosome is essential to mitotic fidelity, and its inheritance could be critical to understanding cell transformation. Investigating the behavior of the centrosome in living mitotic cells, we documented a transient and remarkable postanaphase repositioning of this organelle, which apparently controls the release of central microtubules from the midbody and the completion of cell division. We also observed that the absence of the centrosome leads to defects in cytokinesis. Together with recent results in yeasts, our data point to a conserved centrosome-dependent pathway that integrates spatial controls into the decision of completing cell division, which requires the repositioning of the centrosome organelle.
Article
A monoclonal antibody raised against mitotic spindles isolated from CHO cells ([CHO1], Sellitto, C., and R. Kuriyama. 1988. J. Cell Biol. 106:431-439) identifies an epitope that resides on polypeptides of 95 and 105 kD and is localized in the spindles of diverse organisms. The antigen is distributed throughout the spindle at metaphase but becomes concentrated in a progressively narrower zone on either side of the spindle midplane as anaphase progresses. Microinjection of CHO1, either as an ascites fluid or as purified IgM, results in mitotic inhibition in a stage-specific and dose-dependent manner. Parallel control injections with nonimmune IgMs do not yield significant mitotic inhibition. Immunofluorescence analysis of injected cells reveals that those which complete mitosis display normal localization of CHO1, whereas arrested cells show no specific localization of the CHO1 antigen within the spindle. Immunoelectron microscopic images of such arrested cells indicate aberrant microtubule organization. The CHO1 antigen in HeLa cell extracts copurifies with taxol-stabilized microtubules. Neither of the polypeptides bearing the antigen is extracted from microtubules by ATP or GTP, but both are approximately 60% extracted with 0.5 M NaCl. Sucrose gradient analysis reveals that the antigens sediment at approximately 11S. The CHO 1 antigen appears to be a novel mitotic MAP whose proper distribution within the spindle is required for mitosis. The properties of the antigen(s) suggest that the corresponding protein(s) are part of the mechanism that holds the antiparallel microtubules of the two interdigitating half spindles together during anaphase.
Article
Mitosis comprises a complex set of overlapping motile events, many of which involve microtubule-dependent motor enzymes. Here we describe a new member of the kinesin superfamily. The protein was originally identified as a spindle antigen by the CHO1 monoclonal antibody and shown to be required for mitotic progression. We have cloned the gene that encodes this antigen and found that its sequence contains a domain with strong sequence similarity to the motor domain of kinesin-like proteins. The product of this gene, expressed in bacteria, can cross-bridge antiparallel microtubules in vitro, and in the presence of Mg-ATP, microtubules slide over one another in a fashion reminiscent of microtubule movements during spindle elongation.
Article
Intracellular microtubule motor proteins may direct the motile properties and/or morphogenesis of the mitotic spindle (reviewed in ref. 3). The recent identification of kinesin-like proteins important for mitosis or meiosis indicates that kinesin-related proteins may play a universal role in eukaryotic cell division, but the precise function of such proteins in mitosis remains unknown. Here we use an in vitro assay for spindle assembly, derived from Xenopus egg extracts, to investigate the role of Eg5, a kinesin-like protein in Xenopus eggs. Eg5 is localized along spindle microtubules, and particularly enriched near spindle poles. Immunodepletion of Eg5 from egg extracts markedly reduces the extent of spindle formation in extracts, as does direct addition of anti-Eg5 antibodies. We also demonstrate that Eg5 is a plus-end-directed microtubule motor in vitro. Our results suggest a novel mechanism for the dynamic self-organization of spindle poles in mitosis.