Sie sind auf Seite 1von 11

Vol.

11, 455– 465, August 2000 Cell Growth & Differentiation 455

Regulated Ran-binding Protein 1 Activity Is Required for


Organization and Function of the Mitotic Spindle in
Mammalian Cells in Vivo1

Giulia Guarguaglini,2 Luigina Renzi,3 nucleocytoplasmic traffic in several organisms (for reviews,
Filippo D’Ottavio, Barbara Di Fiore, see Refs. 1– 4). The biological activity of Ran is dependent on
Martina Casenghi,2 Enrico Cundari, and the turnover rate between the GTP- and GDP-bound state.
Patrizia Lavia4 Major regulators controlling the nucleotide-bound state of
National Research Council Centre of Evolutionary Genetics, c/o Ran include RanGAP,5 which catalyzes GTP hydrolysis yield-
Department of Genetics and Molecular Biology, University of Rome “La
Sapienza,” Rome 00185, Italy
ing Ran-GDP (5), and the RCC1 protein, which acts as the
guanine exchange factor for Ran and favors the formation of
Ran-GTP (6). RanBP1 interacts with GTP-bound Ran (7, 8)
Abstract and favors its conversion to Ran-GDP, at least in vitro, by
Ran-binding protein (RanBP) 1 is a major regulator of increasing the rate of GTP hydrolysis via RanGAP and by
the Ran GTPase and is encoded by a regulatory target inhibiting the exchange activity of RCC1 (9).
gene of E2F factors. The Ran GTPase network controls The role of Ran is particularly well documented in control
several cellular processes, including nucleocytoplasmic of nucleocytoplasmic traffic in interphase cells. The nucle-
transport and cell cycle progression, and has recently otide-bound state of Ran is crucial for the assembly and
also been shown to regulate microtubule nucleation disassembly of transport complexes (for recent reviews, see
and spindle assembly in Xenopus oocyte extracts. Here Refs. 10 –12). These findings have led to the proposal that
we report that RanBP1 protein levels are cell cycle many pleiotropic effects ascribed to the RanGTPase and its
regulated in mammalian cells, increase from S phase regulators may in fact reflect a primary effect of Ran over
to M phase, peak in metaphase, and abruptly decline in nuclear transport. However, recent lines of evidence increas-
late telophase. Overexpression of RanBP1 throughout ingly implicate the Ran network in mitotic control. In yeast, a
the cell cycle yields abnormal mitoses characterized by mutant allele of the RanBP1 gene (yrb1 in Saccharomyces
severe defects in spindle polarization. In addition, cerevisiae) causes mitotic spindle misalignment and cell cy-
microinjection of anti-RanBP1 antibody in mitotic cells cle arrest in late mitosis or G1, with no apparent perturbation
induces mitotic delay and abnormal nuclear division, of nuclear import (13). In mammalian cells, a Ran-interacting
reflecting an abnormal stabilization of the mitotic component named RanBPM localizes at centrosomes and
spindle. Thus, regulated RanBP1 activity is required for controls microtubule nucleation from centrosomes (14). Fur-
proper execution of mitosis in somatic cells. thermore, work with Xenopus egg extracts has depicted a
direct role of members of the Ran network in mitotic control
(reviewed in Refs. 3, 4, and 15): the addition of purified
Introduction
Ran-GTP or RCC1 (which generates Ran-GTP) promotes
The Ran GTPase network has been implicated in control of a
microtubule nucleation; whereas the addition of purified Ran-
puzzling variety of processes because mutations in the Ran
GDP, RanGAP, or RanBP1 (which favor Ran-GDP formation)
GTPase itself or in partner molecules affect cell cycle pro-
inhibits microtubule nucleation and spindle assembly (16 –
gression, chromosome stability, nuclear organization, and
19). Frog egg extracts provide a useful experimental system
to pinpoint biochemical requirements for the organization of
microtubules into a functional mitotic spindle. However, so-
Received 1/25/00; revised 4/17/00; accepted 6/1/00. matic cells often show higher regulatory constraints: specific
The costs of publication of this article were defrayed in part by the
payment of page charges. This article must therefore be hereby marked
components may be present in limited amounts and are
advertisement in accordance with 18 U.S.C. Section 1734 solely to indi- often subjected to regulated expression both in time, during
cate this fact. cell cycle progression, and in space, in specialized subcel-
1
This work was supported by grants from the Consiglio Nazionale delle
Ricerche and the European Union. G. G. and B. D. F. were supported by lular compartments. It is important to examine in vivo pro-
fellowships from the Ministero dell’Università e Ricerca Scientifica e Tec- cesses to assess whether components of the Ran network
nologica. L. R. was supported by a fellowship from the European Union/ are actually implicated in mitotic control in somatic cells in
Consiglio Nazionale delle Ricerche. F. D. was supported by a grant from
the Fondazione Buzzati-Traverso. G. G. and L. R. contributed equally to vivo.
this work.
2
Present address: Department of Cell Biology, Max-Planck Institute of
Biochemistry, Am Klopferspitz 18a, D-82152 Martinsried, Germany.
3
Present address: ENEA Centro Ricerche della Casaccia, Via Anguillar-
5
ese 301, S. Maria di Galeria, 00060 Rome, Italy. The abbreviations used are: RanGAP, Ran GTPase-activating protein;
4
To whom requests for reprints should be addressed, at National Re- RanBP, Ran-binding protein; RCC1, regulator of chromosome condensa-
search Council Centre of Evolutionary Genetics, c/o Department of Ge- tion; NOC, nocodazole; IF, immunofluorescence; NES, nuclear export
netics and Molecular Biology, University of Rome “La Sapienza,” Via degli signal; LMB, leptomycin B; GFP, green fluorescent protein; DAPI, 4⬘,6-
Apuli 4, Rome 00185, Italy. Phone: 39-06-4457528; Fax: 39-06-4457529; diamidino-2-phenylindole; GST, glutathione S-transferase; RBD, Ran-
E-mail: patrizia.lavia@uniroma1.it. binding domain; PBST, PBS containing 0.05% Tween 20.
456 Mitotic Functions of RanBP1

Fig. 1. Components of the Ran network during cell cycle progression. A, anti-RanBP1, anti-Ran, and anti-RCC1 antibodies (see “Materials and Methods”
for details) yield specific signals in murine NIH/3T3 cells. Thirty ␮g of whole cell extract were loaded in each lane. Antibodies were used alone (⫺) or after
preincubation (⫹) with immunogenic peptides. B, Western blot assays of protein extract from staged cells (40 ␮g/lane). Protein extracts from growth-
arrested (G0), G1, and S-phase cells were prepared from serum-starved and restimulated cultures harvested at the indicated times after serum addition.
To follow-up mitotic exit, extracts from M-phase cells were prepared from cultures grown in the presence of NOC and then released and harvested at the
indicated times after release of NOC arrest.

In previous work, we cloned and characterized the regu- tibody into living cells progressing through mitosis, when
latory features of the murine RanBP1 gene. We found that RanBP1 is maximally expressed. This induced mitotic delay,
RanBP1 mRNA transcription is subjected to both growth segregation defects, and incomplete separation of daughter
regulation in proliferating versus quiescent cells (20, 21) and nuclei, reflecting, at least in part, an abnormal stabilization of
cell cycle phase-specific regulation, being specifically up- the spindle microtubules. Both lines of evidence therefore
regulated at the G1-S-phase boundary (22). This control is indicate that RanBP1 dysfunction or deregulation impairs
exerted by E2F and retinoblastoma-related proteins (23), mitotic spindle function and hence proper mitotic division in
which act as major cell cycle regulators (see Refs. 24 –26 for mammalian cells in vivo.
reviews). Thus, E2F-dependent control couples RanBP1
transcription to that of many genes required for cell cycle
progression (reviewed in Ref. 27). This control is lost in Results
several tumor types, and RanBP1 is consistently overex- The RanBP1 Protein Is Present in a Limited Cell Cycle
pressed in several transformed cell lines (20). The link be- Window in Mammalian Cells. Before addressing the func-
tween RanBP1 gene expression and the cell cycle machinery tional role of RanBP1, we sought to compare the relative
suggests that RanBP1 protein plays an important role in cell distribution of components of the Ran network during the cell
cycle control. Such a role could be exerted either directly or cycle phases. Western immunoblotting experiments were
via modulation of the efficiency of nucleocytoplasmic trans- carried out with murine NIH/3T3 whole cell extract using
port during the cell cycle phases. Indeed, previous experi- commercial affinity-purified antibodies against Ran, RanBP1,
ments in which RanBP1 transcription was rendered consti- and RCC1. Highly specific signals were generated, which
tutive from a cell cycle-independent promoter yielded were abolished by preincubation with the immunogenic pep-
significant cell cycle alterations with a predominant impair- tides (Fig. 1A). Cell cultures were induced to reach growth
ment in mitotic exit (28). In retrospect, mitotic defects or arrest (G0) by serum starvation and subsequently stimulated
arrest may be consistent with the regulatory role over spindle to cycle by serum refeeding. To examine reentry into S phase
assembly ascribed to purified Ran network components in after mitotic completion and hence distinguish between cell
cell-free extracts. cycle phase-specific and growth-dependent regulation, cells
The experiments described here were undertaken to gain were also induced to accumulate in prometaphase in the
further insight into the mitotic role of RanBP1 in somatic cells presence of NOC and then released from prometaphase
in vivo. Two major experimental approaches were taken. arrest in NOC-free medium and allowed to progress into
Firstly, we sought to characterize cellular phenotypes gen- mitosis and into the following G1 phase. Most cells exited
erated in cell cultures constitutively expressing RanBP1. We mitosis and resumed a normal interphase appearance within
report that two major processes are impaired under those 2 h after release of the NOC block. Fig. 1B shows that
conditions: (a) mitotic spindles showed an abnormal number RanBP1 protein levels, but not those of Ran or RCC1, are low
of poles; and (b) after mitotic exit, the reorganization of in G0-early G1 cells and are up-regulated during progression
nuclear chromatin was aberrant. Given that both classes of from S phase to mitosis. RanBP1 levels are high in promet-
defects were typically found either in mitosis or in cells that aphase-arrested cells but decline within 1 h after release of
had just passed through mitosis, we further sought to estab- NOC arrest, i.e., when cells traverse the mitosis to G1 tran-
lish whether RanBP1 dysfunction directly affected mitotic sition. These results are specific by comparison with both
progression. To that end, we microinjected anti-RanBP1 an- phase-specific (i.e., E2F-1 in the case of release from serum
Cell Growth & Differentiation 457

Fig. 2. Immunolocalization of
RanBP1 during the cell cycle. A,
NIH/3T3 cell cultures stimulated
to cycle were examined at vari-
ous times after serum addition. a,
10 h (G1). b, 18 h (S/G2). c⫺e,
mitotic cells were seen after 24 h,
when synchronization is gradu-
ally lost. Cell spreads were incu-
bated with anti-RanBP1 antibody
followed by FITC-conjugated
secondary antibody and counter-
stained with DAPI. Bar, 10 ␮m. B,
RanBP1 during mitotic exit. Cells
were exposed to NOC for 9 h and
then fixed at regular intervals
from the block release and
stained with DAPI, MPM-2, and
anti-RanBP1 antibodies. A met-
aphase is shown in column a (20
min after release from NOC ar-
rest), anaphase and telophase
are shown in columns b (40 min
after release) and c (80 min after
release), and a late telophase is
shown in c (arrows). Bar, 10 ␮m.
C, RanBP1 transits through the
nucleus. Constructs encoding
wild-type (a, pRanBP1wt) or NES-
mutagenized (b, pNES) RanBP1
were transfected in cycling cells,
and the subcellular localization
of the protein was determined.
RanBP1 IF and DAPI staining
were performed as described for
A. RanBP1 is cytoplasmic in con-
trol cultures (c, ⫺LMB) but be-
comes nuclear within 2 h of LMB
addition (d, ⫹LMB). Bar, 10 ␮m.

starvation and cyclin B1 after release from NOC arrest) and became barely detectable in late telophase (compare Fig.
phase-independent (i.e., actin) protein markers (data not 2A, e with Fig. 2A, c and d). To follow-up mitotic exit more
shown). In contrast, both Ran and RCC1 are steadily ex- accurately, cells were cultured in NOC to induce promet-
pressed under the same conditions (Fig. 1B). These findings aphase accumulation and then released in NOC-free me-
delimit a specific cell cycle window during which RanBP1 is dium as described above. Cells were fixed during the follow-
present, whereas Ran and RCC1 are both expressed in a ing 2 h at regular intervals. To distinguish late telophase
constitutive manner. figures from early interphase figures, cells were labeled using
We next examined the RanBP1 protein in cell cultures MPM-2 antibody, which reacts with phosphoepitopes gen-
synchronized by serum starvation/restimulation using indi- erated in late G2 and mitosis (29). These experiments re-
rect IF. Consistent with the immunoblotting results, the vealed high levels of RanBP1 until early telophase (Fig. 2B, a
RanBP1 signal was barely detectable in early G1 cells (Fig. and b) and a massive decrease in RanBP1 levels in late
2A, a) yet was effectively visualized in cells beyond the G1-S telophase, concomitant with chromatin decondensation; for
transition (Fig. 2A, b). In metaphase and anaphase cells, example, the arrowed nuclei in Fig. 2B, c have reestablished
RanBP1 immunostaining reached the highest intensity and a chromatin organization resembling that of G1 nuclei yet
was confined to the mitotic cytoplasm (Fig. 2A, c and d). represent late telophase products based on MPM-2 reactiv-
However, the signal abruptly declined during mitotic exit and ity in the midbody (arrowed in the middle row). Thus, these
458 Mitotic Functions of RanBP1

Fig. 3. Ran is redistributed to the cy-


toplasm, whereas RCC1 colocalizes
with chromatin during mitosis. Cells
were synchronized by serum starva-
tion/restimulation as described in the
Fig. 2A legend. a and e were taken
from S/G2-enriched cultures 18 h after
cell cycle entry; a similar distribution
was detected throughout interphase.
Prophase cells in b and f, metaphase
cells in c and g, and anaphase cells in
d and h were seen among cultures har-
vested 24 h after cell cycle entry. Cell
spreads were fixed in paraformalde-
hyde and incubated with anti-Ran an-
tibody (a⫺d) or fixed with methanol
and incubated with anti-RCC1 anti-
body (e⫺h). FITC-conjugated second-
ary antibody was used, and DNA was
counterstained with DAPI. Bar, 10 ␮m.

experiments enabled us to visualize the stage at which Western blot analysis, with a predominantly nuclear location,
RanBP1 underwent down-regulation as revealed in Western as seen in other cell types (34, 35). Cytoplasmic signals were
blot assays (see Fig. 1B). also detected in certain cells, consistent with the intrinsic
In S and G2 phases, in which RanBP1 is abundantly ex- shuttling activity of the protein. In prophase, IF signals were
pressed, a predominant cytoplasmic distribution was ob- excluded from condensing chromatin and accumulated to-
served, yet nuclear signals were also apparent (see Fig. 2A, ward the nuclear periphery (Fig. 3b). Ran was massively
b). RanBP1 was previously reported to contain a NES (30, redistributed to the cytoplasm during mitotic progression
31). Indeed, by mutagenizing two crucial amino acids iden- (Fig. 3, c and d), with an increased intensity associated with
tified by Richards et al. (30) within the NES (i.e., L186A and the mitotic spindle in metaphase (Fig. 3c). In contrast, RCC1
V188A), we were able to visualize nuclear retention of the immunostaining coincided with chromatin at all interphase
protein (Fig. 2C, compare b with a). Furthermore, we ob- stages (examples of S-phase cells are shown in Fig. 3e) and
served nuclear accumulation of the endogenous RanBP1 in throughout mitotic progression, including the moment of
cell cultures exposed to LMB (Fig. 2C, compare d with c), chromosome alignment in metaphase (Fig. 3g). Chromo-
which blocks nuclear export by specifically inhibiting the somes were selectively stained by the RCC1 antibody in
CRM1/exportin 1 factor (32, 33). These results are consistent methanol-fixed cells (Fig. 3, f⫺h), whereas formaldehyde
with previous competition experiments with functional NES fixation enabled us to visualize a fraction of RCC1 in the
sequences that had implicated CRM1 in RanBP1 export (31). mitotic cytoplasm in addition to a high proportion of the
Together, the IF results indicate that RanBP1 does indeed RCC1 pool that remained chromosome associated through-
enter and exit nuclei in a regulated manner in the S and G2 out mitosis (data not shown), consistent with previous results
phases of the cell cycle. in human cells (36). Examination of isolated mitotic chromo-
Because no comparative analysis of Ran network compo- somes from human AHH1 cells confirmed that RCC1 is in-
nents has actually been carried out in somatic cells progress- deed retained on metaphase chromosomes (data not
ing through the cell cycle, IF methods were also used to shown).
examine staged cell cultures for their Ran and RCC1 content. In summary, only Ran and RCC1 are steadily expressed
Ran was detected throughout the cell cycle (examples of throughout the cell cycle, whereas RanBP1 levels are low in
S-phase cells are shown in Fig. 3a), consistent with the G1 and increase throughout S phase and G2 phase. In these
Cell Growth & Differentiation 459

Table 1 Cellular abnormalities in cultures overexpressing RanBP1 products (Ref. 28; data not shown). The frequency of cells
with chromatin abnormalities was statistically higher in cul-
Cellular phenotypes were scored among GFP-expressing cells in cul-
tures transfected with pX vector, pRanBP1 or pNES (6 ␮g of each con- tures transfected with pNES than in cultures transfected with
struct). Ps were calculated from the single comparison of either pRanBP1- pRanBP1 construct (Table 1) and increased with the dose of
or pNES-transfected cultures, with control cultures using the ␹2 test.
transfected construct (Fig. 4). Immunostaining of transfected
Cell types (%) pX pRanBP1 pNES cells using the MPM-2 antibody to visualize mitotic proteins
Normal interphases 81.63 66.5 47.64a revealed that nuclear abnormalities were typically associated
Normal mitoses 6.08 5.4 6.8 with expression of MPM-2 antigens and hence were gener-
Abnormal mitoses 0.36 5.2a 8.63a ated during mitosis. Actually, several pyknotic nuclei ap-
Pyknotic nuclei 7.5 13.75 21.99a
peared in paired figures (see Fig. 4A for example), suggesting
Aberrant nuclei 4.38 9.12 14.92a
Counted cells 822 670 764 that they derived from telophase cells that failed to decon-
dense chromatin.
P ⬍⬍0.001 ⬍⬍0.001
Examination of DAPI staining in GFP-positive cells further
a
Classes whose partial ␹2 values contribute most significantly to the
total ␹2.
revealed that abnormal mitotic figures also accumulated in
cultures transfected with either export-defective or wild-type
RanBP1 construct (Table 1). To characterize mitotic abnor-
phases, Ran and RCC1 are largely or almost exclusively malities in more detail, transfection experiments were re-
nuclear, whereas RanBP1 is largely cytoplasmic but appears peated, and cells were immunostained using an anti-␣-
to be able to shuttle between the nucleus and the cytoplasm. tubulin antibody to visualize the spindle. Severe spindle
In mitotic cells, all components are abundantly expressed defects were observed in the presence of either construct. In
and asymetrically redistributed: mitotic chromosomes only most cases, the number of spindle poles was abnormal;
retain RCC1 or at least a significant fraction(s) of the RCC1 monopolar (Fig. 5a) and multipolar (Fig. 5, b and c) spindles
pool; whereas Ran and RanBP1 massively localize to the were observed. In certain cases, no spindle structure could
mitotic cytoplasm. RanBP1 is present at high levels until late actually be recognized, and mitotic chromosomes were scat-
telophase and is dramatically down-regulated during mitotic tered around tubulin aggregates that were largely unfocused
exit, so that cells reentering G1 again express low levels of (Fig. 5d). Polarization defects were particularly evident in
RanBP1. metaphase cells and were accompanied by misaligned or
Overexpression of RanBP1 in Asynchronously Cycling unattached chromosomes. These defects persisted in an-
Cells Yields Aberrant Mitotic Spindles and Nuclear Ab- aphase/telophase, with figures often showing abnormal cen-
normalities. In previous work (28), we designed experi- tral spindles in both multipolar and bipolar cells, with shorter
ments to override cell cycle regulation of endogenous and thicker microtubules compared with control cells trans-
RanBP1 and forced constitutive transcription from a trans- fected with vector alone. Groups of chromatids segregated
fected cytomegalovirus-based vector: transfected cells abnormally in multipolar anaphase cells (Fig. 5e). The fre-
failed to reach the G0 state during serum starvation and quency of abnormal mitoses was highly significant (Table 1),
remained arrested at various stages of mitosis or during and their phenotype was indistinguishable in cultures over-
mitotic exit. To extend those earlier observations, we under- expressing either wild-type or export-defective RanBP1.
took the characterization of mitotic defects in cycling cells Microinjection of Anti-RanBP1 Antibody in Living Mi-
overexpressing RanBP1. Cells were cotransfected with totic Cells. Results thus far indicate that the mitotic spindle
mammalian pRanBP1 expression construct and with pGFP organization is sensitive to intracellular levels of RanBP1. To
plasmid, which enabled us to unambiguously identify trans- directly assess the role of RanBP1 in mitosis, we injected
fected cells. The export-defective RanBP1 derivative (pNES anti-RanBP1 antibody in living cells that were progressing
construct; see Fig. 2C) was used for comparison. GFP- through mitotic substages, when RanBP1 is maximally ex-
expressing cells were examined 36 – 48 h after transfection. pressed. To further ascertain that anti-RanBP1 antibody did
Data from eight independent experiments are summarized in not cross-react with conserved RBDs present in other cel-
Table 1. In cultures transfected with empty vector (pX), over lular proteins, we carried out preliminary Western blot exper-
80% of GFP-expressing cells had a normal interphase ap- iments with purified GST-fusion proteins expressing single
pearance; about 6% were normal mitoses, and around 12% RBDs (RBD1– 4) from the RanBP2 protein: no cross-reactiv-
showed an abnormal nuclear morphology and/or condensed ity was detected (data not shown). Microinjections were car-
chromatin. The proportion of normal interphase cells de- ried out during mitotic substages as indicated in Table 2.
creased in cultures overexpressing pRanBP1 (66.5%) and Control cells were injected with either PBS alone or mouse
decreased more dramatically in cultures transfected with IgG. After microinjection, cells were allowed to progress
pNES (47.6%). Concomitantly, abnormal mitotic figures and through mitosis, and the timing of each mitotic substage was
cells with aberrantly condensed nuclei accumulated in these recorded until completion of the mitotic division. We found
cultures. Nuclear abnormalities included pyknotic nuclei, in that anti-RanBP1 injection during metaphase and early an-
which chromatin was homogenously condensed (Fig. 4A), aphase delayed further mitotic progression (Table 2): the
and nuclei with irregular edges and blobs of condensed average length of metaphase and anaphase actually doubled
chromatin (Fig. 4B). Flow cytometry and terminal de- compared with that of control cells. One anti-RanBP1-
oxynucleotidyl transferase-mediated nick end labeling anal- injected cell was blocked in metaphase for as long as 94 min;
yses revealed that aberrant nuclei do not represent apoptotic however, that particular delay was unique and was not con-
460 Mitotic Functions of RanBP1

Fig. 4. Examples of nuclear defects induced


by overexpression of wild-type or export-
defective RanBP1. Asynchronously cycling
cell cultures were transfected with vector (pX),
wild-type pRanBP1, or pNES (4 and 6 ␮g of
each). Recorded defects include (A) pyknotic
nuclei with homogeneously hypercondensed
chromatin and (B) abnormal nuclei with irreg-
ular edges and blobs of chromatin. Right, de-
fects were quantified among 400 GFP-positive
cells cotransfected with pX vector, wild-type
pRanBP1, and pNES and stained with MPM-2
antibody. Histograms represent the percent-
age of observed defects using 4 or 6 ␮g of
each construct.

sidered in statistical calculations of the average mitotic delay the poles. As assessed by IF staining of ␣-tubulin, the anti-
in Table 2. Most importantly, after recovery of the delay and RanBP1 antibody does not appear to alter the spindle mor-
resumption of mitotic progression, most injected metaphase phology in microinjected cells (Fig. 7a). We therefore sought
and anaphase cells failed to achieve complete nuclear seg- to assess whether the spindle dynamics were affected as
regation. The formation of the contractile ring was apparently revealed by testing the sensitivity of mitotic microtubules to
not affected (examples of microinjected cells progressing the tubulin-depolymerizing activity of NOC in the presence or
through cytokinesis are shown in Fig. 6A), hence daughter absence of the anti-RanBP1 antibody. Metaphase cells were
cells began to part, but the nuclear contents remained partly microinjected with anti-RanBP1 antibody and immediately
or largely connected. This was best visualized by fixing the exposed to NOC. After 10 min, cells were fixed and labeled
cells after termination of the in vivo observation and staining using a secondary antibody to reveal the microinjected anti-
the DNA with DAPI. In control (PBS- or IgG-injected) cells, RanBP1 and anti-␣-tubulin antibody to visualize microtu-
daughter nuclei were fully separated, and the distance be- bules. Control metaphase cells injected with PBS alone
tween them clearly indicated that the spindle motion toward showed a disassembled spindle and a diffuse ␣-tubulin
the poles had proceeded normally (Fig. 6A, a). In contrast, staining throughout the mitotic cytoplasm within 10 min of
variable amounts of nuclear DNA remained trapped in the NOC addition (Fig. 7b), indicating that microtubules were
cleavage furrow in many anti-RanBP1-injected cells, so that completely depolymerized. In contrast, when anti-RanBP1-
the nuclear division was either incomplete (Fig. 6A, b) or microinjected metaphase cells were exposed to NOC (Fig.
ultimately failed (Fig. 6A, c). Abnormal mitotic products were 7c), microtubules remained polymerized and effectively
particularly frequent among cells that had been injected with stained. Thus, RanBP1 inactivation during metaphase coun-
anti-RanBP1 during metaphase and anaphase (Table 3). Ex- teracts the effect of NOC on tubulin depolymerization and
amples of defects of increasing severity are shown in Fig. 6B: prevents disassembly of the mitotic spindle.
micronuclei generated by chromosome loss were often pres-
ent in late telophase/early G1 cells (Fig. 6B, a); most anti-
RanBP1-injected cells showed chromatin bridges (Fig. 6B, b Discussion
and c); and, in extreme cases, daughter nuclei remained The present study reports several novel aspects of RanBP1
partly or largely connected, and their separation was pre- function in mitotic control in mammalian cells in vivo. We
vented (Fig. 6B, d and e). have found that regulated RanBP1 activity is required in at
The defects shown in Fig. 6 suggest that microinjection of least three important processes associated with mitotic di-
anti-RanBP1 antibody during or after metaphase impairs the vision, including the structural organization of the spindle,
spindle dynamics and hence impairs chromatid migration to control of microtubule dynamics in metaphase and an-
Cell Growth & Differentiation 461

GDP ratio at the G1-S and M-G1 transitions. Between S


phase and the nuclear envelope breakdown, RanBP1 tran-
siently enters the nucleus. RanBP1 export from the nucleus
depends on NES integrity and on the availability of functional
CRM1 (Refs. 30 and 31 and this study). The fact that we
never saw nuclear accumulation of the endogenous RanBP1
except in the presence of LMB suggests that rapid cycles of
import and export continuously take place, such that only a
fraction of the RanBP1 pool localizes to the nucleus at any
given time. During nuclear transit, RanBP1 may transiently
interact with RCC1 in vivo, consistent with the demonstrated
ability of both proteins to interact in vitro (9, 37). Such inter-
actions may be important to convey or modulate regulatory
signal(s) between the nuclear and cytoplasmic compart-
ments in S phase and G2.
To begin to pinpoint cell cycle events sensitive to alter-
ations in RanBP1 activity, overexpression experiments were
carried out in asynchronous cycling cells. Recorded defects
fall into two major classes: (a) mitotic spindles with abnormal
poles; and (b) completely or locally hypercondensed nuclei in
interphase cells. The latter were associated with MPM-2
reactivity, indicating that an aberrant chromatin organization
is established during mitosis and persists through mitotic
exit, preventing a normal interphase reorganization in daugh-
ter cells during the following cell cycle. Nuclear chromatin
organization is differentially sensitive to overexpression of
mislocalized as opposed to wild-type RanBP1.
Chromatin defects were observed previously in mamma-
lian cell cultures overexpressing RanBP1 during serum star-
vation, during which cells ought to have completed the mi-
totic division to enter G0 (28), and in Schizosaccharomyces
pombe strains carrying an imbalance among components of
the Ran network (38, 39) and defective in the mitosis-to-
interphase transition (40). In nuclear reconstitution experi-
ments with Xenopus laevis extracts, excess RanBP1 inter-
Fig. 5. Defects in spindle polarization after overexpression of wild-type
or export-defective RanBP1. Asynchronous cell cultures were transfected feres with nuclear reassembly and nuclear envelope
with vector, wild-type pRanBP1 (a and b), or pNES (c⫺e). In all cases, the reformation after mitosis (35, 41). In our experiments, this
pGFP expression plasmid was cotransfected. Cells were fixed with phenotype is induced with significantly higher frequency by
paraformaldehyde to visualize GFP, processed for IF using anti-␣-tubulin
antibody (left column), and counterstained with DAPI (middle column). export-defective RanBP1 as compared with wild-type
Merged figures in the right column show chromosome misalignment with RanBP1. Both constructs yielded continuous synthesis of
respect to aberrant spindles. Examples of spindle abnormalities are
shown: a, monopolar spindle; b, tripolar spindle; c, tetrapolar spindle, d, RanBP1, which persisted in late telophase, when the endog-
tubulin aggregates with no clear spindle structure; and e, tripolar enous protein normally disappears, and chromatin is due to
anaphase. decondense. Thus, continuous RanBP1 expression during
mitotic exit, particularly when accompanied by nuclear re-
tention, appears to interfere with the relocalization and/or
aphase, and nuclear chromatin reorganization after mitotic activity of components regulating cyclic condensation and
exit. decondensation of mitotic chromatin.
In mammalian cells, the RanBP1 protein is abundant in the A central aspect of the present work is represented by the
cell cycle window included between S phase and late telo- finding that the mitotic spindle is severely affected in cells in
phase, unlike Ran and RCC1, which are both steadily ex- which RanBP1 activity was deregulated during the preceding
pressed during the cell cycle. Given the antagonism between interphase or impaired during mitosis. Spindles with an ab-
RanBP1 and RCC1 in modulating Ran activity (9), these data normal number of poles were observed in cells that reached
suggest that two major changes in the ratio of RanBP1:RCC1 mitosis after continuous expression of RanBP1. Unlike chro-
take place during the cell cycle: (a) a first switch is expected matin defects, which are preferentially generated by export-
to occur at the onset of S phase, when RanBP1 begins to defective RanBP1, abnormal spindles were assembled in
accumulate; and (b) a second switch would take place in late cells overexpressing either form of the protein. Thus, main-
telophase, when RanBP1 is down-regulated. Cyclic fluctua- taining regulated RanBP1 levels appears to represent a more
tions in RanBP1 levels in nontransformed cells are expected critical requirement than correctly localizing the protein, as
to determine programmed changes in the Ran-GTP:Ran- far as the spindle structure is concerned. Overexpressed
462 Mitotic Functions of RanBP1

Table 2 Anti-RanBP1 microinjection in mitosis induces metaphase and anaphase delay

Microinjected cells Metaphase length (min) Anaphase length (min)


Injection
Stage No. of cells Mean ⫾ SD Range Mean ⫾ SD Range

PBS Prophase a
10 6.2 ⫾ 2.0 4–10 9.5 ⫾ 1.6 9–13
Prometaphase 10 6.2 ⫾ 2.3 3–10 10.3 ⫾ 13 9–12
Metaphase 10 5.2 ⫾ 2.9 2–10 9.0 ⫾ 2.7 7–15
Anaphase 5 13.8 ⫾ 4.7 7–21
IgG Prophasea 10 6.4 ⫾ 1.6 4–10 9.5 ⫾ 2.4 7–13
Prometaphase 10 5.6 ⫾ 1.7 3–9 10.7 ⫾ 3.1 8–15
Metaphase 10 5.6 ⫾ 1.6 2–8 12.4 ⫾ 2.8 8–16
Anaphase 5 12.7 ⫾ 2.6 9–16
RanBP1 antibody Prophasea 10 7.5 ⫾ 5.4 3–16 8.6 ⫾ 1.1 7–10
Prometaphase 10 7.0 ⫾ 3.5 3–15 13.6 ⫾ 9.1 7–34
Metaphase 10 10.9 ⫾ 5.1b,c 5–20 18.2 ⫾ 7.6d 11–30
Anaphase 5 24.0 ⫾ 4.6c 18–31
a
No difference between cytoplasmic and nuclear injection.
b
Because one cell was arrested for 94 min, mean ⫾ SD values were calculated from nine cells only.
c
P ⬍ 0.01 (differences in mitotic stage lengths were evaluated using the Student’s t test).
d
P ⬍ 0.05 (differences in mitotic stage lengths were evaluated using the Student’s t test).

Fig. 6. Examples of defects induced


by microinjecting RanBP1 antibody
into mitotic cells. Mitotic progression
was timed by live image recording for
at least 90 min. A, examples of mitotic
progression in microinjected cells in
vivo (phase-contrast microscopy).
DAPI-stained nuclei after fixation are
shown in the right column. a, PBS-
injected cell progressing through
complete cytokinesis and nuclear di-
vision. Anti-RanBP1-microinjected
cells, despite forming a normal cleav-
age furrow, show partly (b) or largely
(c) incomplete nuclear division. Bar,
10 ␮m. B, abnormal mitotic products
in anti-RanBP1-microinjected cells
fixed after mitotic completion and
stained with DAPI. a, micronucleus
(arrow); b and c, DNA bridges be-
tween daughter cells, with increasing
amounts of trapped chromatin in the
actin contractile ring (arrows); d, chro-
matid sets did not migrate at an-
aphase; e, chromosomes did not go
through metaphase or anaphase, and
chromatin blobs were visualized at
telophase. Bar, 10 ␮m.

wild-type RanBP1 will abnormally activate RanGAP in the regulation of nucleation activities (reviewed in Refs. 3, 4, and
cytoplasm, yielding an excess of Ran-GDP. On the other 15). RanBP1 may affect the organization of the mitotic spin-
hand, the export-defective version is expected to yield an dle in various ways: it may contribute to a regulatory pathway
excess of RanBP1/RCC1/Ran interacting complexes in the directly regulating the spindle structure [for example, the
nucleus. Based on biochemical evidence (9), RCC1 would centrosome-associated RanBPM protein (14) may be viewed
become inactive in those complexes, which would also yield as a putative target]; or else the spindle abnormalities may
an excess of Ran-GDP. Thus, RanBP1 overexpression could result from impaired transport of particular proteins, which
interfere through either mechanism with the timing of Ran- would in turn perturb spindle assembly. In either framework,
GTP-dependent interactions important for the spindle forma- the results in Fig. 5 link for the first time the requirement for
tion. Experiments with cell-free Xenopus oocyte extracts regulated levels of RanBP1 (a situation that in vivo is per-
indicate that the nucleotide-bound state of Ran is crucial for turbed in retinoblastoma-deficient cells and tumors; see
Cell Growth & Differentiation 463

Table 3 Defects in mitotic cells microinjected with anti-RanBP1


antibody

No. of
Stage of injected cells Observed defects
injection
Total Aberrant

Prophase 10 2 Chromosomes decondensed in the


absence of proper metaphase-to-
anaphase transition; chromatin
blobs formed; no cytokinesis
(Fig. 7B, p d)
1 Chromatin bridge between daughter
nuclei; no cytokinesis
1 Tripolar cell

Prometaphase 10 1 Chromatin bridge


2 Micronuclei
1 Chromosomes decondensed in the
absence of proper metaphase-to-
anaphase transition; chromatin
blobs formed; no cytokinesis
Fig. 7. Anti-RanBP1 microinjection stabilizes the mitotic spindle. Met-
aphase cells were injected with PBS or with anti-RanBP1 antibody and
Metaphase 10 2 Lagging chromosomes and exposed to NOC. After 10 min, cells were fixed and stained with anti-␣-
micronuclei (Fig. 7B, p a) tubulin antibody to reveal the spindle morphology and with secondary
2 Chromatin bridges (Fig. 7B, p b) antibody to visualize anti-RanBP1 antibody; chromosomes were stained
2 Chromatin bridges; no cytokinesis with DAPI. a, normal spindle in a metaphase cell injected anti-RanBP1
(p b in Fig. 7A and c in Fig. 7B) antibody; b, disassembled spindle in a PBS-injected metaphase cell ex-
1 Incomplete separation of posed to NOC; c, polymerized spindle in an anti-RanBP1-injected met-
chromosome sets aphase cell exposed to NOC. Bar, 10 ␮m.
1 Metaphase arrest for 94 min;
eventually, daughter nuclei
separated, forming one
micronucleus aphase alignment. Anti-RanBP1 microinjection prevented
the tubulin-depolymerizing effect of NOC on spindle micro-
Anaphase 5 2 Delay in chromosome migration, tubules. These results suggest that the role of the RanBP1
incomplete anaphase, chromosome
protein in vivo is exerted, at least in part, through control of
arms entangled with the contractile
ring, no cytokinesis (p c in Fig. 6A microtubule dynamics after metaphase alignment. At this
and e in Fig. 6B) stage, this effect is clearly transport independent. During
2 Chromatin bridges, no cytokinesis
mitosis, Ran and RanBP1 localize to the mitotic cytoplasm,
1 Progressed from anaphase onset to
anaphase B in 10 min; aberrant whereas RCC1 remains largely associated with chromo-
telophase after 31 min somes. Part of Ran colocalizes with the mitotic spindle.
RanGAP, although not examined here due to the lack of a
reliable antibody against the murine protein, was also shown
to colocalize with the spindle in human cells (43). Because
Refs. 20 and 23) to balanced chromosome segregation in microinjection results indicated that functional RanBP1 is
daughter cells. RanBP1 expression is normally induced at required in metaphase and anaphase, we tested the possi-
the G1-S transition under the control of E2F factors (23), as bility that Ran and RanBP1 act in control of the spindle
is the expression of several genes whose products are re- dynamics through a direct association with the spindle mi-
quired for DNA replication (27). The centrosome duplication crotubules. However, no component of the Ran network was
cycle has also been recently shown to implicate E2F1 and evidenced in coimmunoprecipitation assays with mitotic tu-
cyclin A/cyclin-dependent kinase 2 activities (42). It may be bulin; furthermore, only a minor fraction of Ran and RanBP1
speculated that coordinated control of RanBP1 and of genes cosedimented with in vitro polymerized microtubules pre-
involved in the DNA replication and centrosome duplication pared from mitotic extracts as described in Ref. 44 (data not
machineries is important for coupling formation of the spin- shown). Thus, the largest pool of Ran and RanBP1 does not
dle poles to the cell cycle. appear to be directly engaged in a structural interaction with
Direct evidence for a distinct role of RanBP1 in spindle tubulin in the spindle. These observations rather suggest that
control was shown by microinjecting anti-RanBP1 antibody RanBP1 activity (and hence Ran-GTP hydrolysis) near the
into mitotic cells. In these experiments, mitotic products spindle may be important to either inactivate a (⫹end-direct-
showed chromatin bridges and micronuclei reflecting chro- ed) protein important for microtubule growth until metaphase
mosome loss or failed separation in extreme cases in which or to activate a protein important for spindle motion toward
daughter nuclei remained mutually entangled. This in vivo the poles after metaphase. The presence of RCC1 on chro-
function is clearly distinct from the nucleation regulatory mosomes may be important to maintain local regions of high
activities ascribed to purified Ran network components in Ran-GTP near the sites of microtubule attachment to chro-
Xenopus oocyte extracts. In our experiments, the mitotic mosomes.
spindle had evidently been functional before microinjection In conclusion, the present results provide the first in vivo
of anti-RanBP1 antibody, to the point of orchestrating met- evidence that RanBP1 is directly implicated in mitotic control
464 Mitotic Functions of RanBP1

in mammalian cells. Because the RanBP1 gene is a regula- arations were incubated in 20% (v/v) FCS in PBST for 45 min. Antibodies
tory target of E2F- and retinoblastoma-related factors, which against Ran network components were used at 10 ␮g/ml, MPM-2 anti-
body was used at 1.3 ␮g/ml, and anti-␣-tubulin was used at 3 ␮g/ml.
are often deregulated in many tumors, these results may be
Primary antibodies were diluted in 5% (v/v) FCS in PBST, and coverslips
relevant in terms of regulatory mechanisms that may be were incubated for 45 min. After rinsing in PBST, coverslips were incu-
altered during oncogenesis. In addition to an abnormal rate bated for 30 min with FITC-conjugated antigoat antibody (4 ␮g/ml; Santa
of cell proliferation, many solid tumors in which members of Cruz Biotechnology) to detect Ran, RCC1, and RanBP1 or with Texas
the E2F/retinoblastoma pathway are unbalanced also show Red-conjugated antimouse antibody (15 ␮g/ml; Vector Laboratories) to
detect MPM-2 and ␣-tubulin. Incubations were carried out at 37°C in a
high frequencies of aneuploidy. The present findings, which humid chamber. After washing, coverslips were counterstained with DAPI
place RanBP1 in a crucial regulatory pathway for mitotic (0.5 ␮g/ml in distilled water) and mounted in Vectashield (Vector Labora-
execution, and the characterization of mitotic defects asso- tories). Images were taken using a Zeiss Axioplan microscope configured
ciated with RanBP1 deregulation or dysfunction may begin for epifluorescence with a ⫻100 oil immersion 1.3 objective and equipped
with a charge-coupled device camera (Photometrics).
to identify aberrant processes that may occur in tumors.
Mammalian Expression Constructs and Transfections. The
pRanBP1 wild-type construct was generated by inserting Htf9-a/RanBP1
cDNA (GenBank X56045) under the cytomegalovirus promoter/enhancer
Materials and Methods region in the pBluescript-derived pX vector (28). An export-defective
Cell Cultures and Cell Cycle Analysis. Murine NIH/3T3 embryo fibro- version (termed pNES) was synthesized using the Quick Change site-
blasts (ATCC CRL 1658) were routinely grown as described previously directed mutagenesis kit (Stratagene) and the oligonucleotide 5⬘-
(22). Cell cycle synchronization was obtained by maintaining the cultures GAGAAGCTGGAAGCCGCTTCAGCTAGGGAGGCCAGAGAG-3⬘. pNES
in low FCS (0.5%) for 48 h to induce growth arrest and subsequently carries two amino acidic substitutions, i.e., L186A and V188A, within the
stimulating synchronous cell cycle reentry by raising the serum to 15%. NES (30). NIH/3T3 cells were seeded in Petri dishes (21 cm2) onto sterile
Cells were collected at regular intervals after stimulation. Cultures were coverslips and transfected using FUGENE (Boehringer/Roche) and 4 or 6
also synchronized in prometaphase by exposure to 0.2– 0.5 ␮g/ml NOC ␮g of DNA from pX empty vector, wild-type pRanBP1, or pNES. pGFP
for 9 h and then released in NOC-free medium and harvested during the expression vector (2 ␮g/dish; Clontech) was also cotransfected. Cells
following 2 h at 20-min intervals. Cell cycle progression was analyzed by were collected 36 – 48 h after transfection and processed for indirect IF to
fluorescence-activated cell sorting using a FACStar Plus flow cytometer visualize either GFP and MPM-2 or GFP and ␣-tubulin.
(Becton Dickinson) as described previously (22). To inhibit nuclear export, Antibody Microinjection Experiments. Cells were cultured on
cells were exposed to 20 nM LMB [a kind gift from Minuro Yoshida etched coverslips, and the medium was changed just before injection. A
(Department of Biotechnology, University of Tokyo, Tokyo, Japan)] for 2 h. concentrated stock (2 mg/ml in PBS) of affinity-purified anti-RanBP1
Antibody Assays and Western Immunoblotting Experiments. The antibody (Santa Cruz Biotechnology) was used. Microinjections were
specificity of antibodies against components of the Ran network was performed using borosilicate glass microneedles mounted on a manual
assessed in Western blot assays with whole cell extract (30 ␮g) from micromanipulator and a microinjector (Narishige, Tokyo, Japan). Cells
NIH/3T3 cells by preincubating the antibodies with the corresponding were microinjected at different stages of mitosis, from prophase to early
immunogenic peptides (2.5 ␮g/ml). All antibodies and immunogenic pep- anaphase. PBS or mouse IgG (2 mg/ml) was used in control experiments.
tides were from Santa Cruz Biotechnology. Affinity-purified antibodies Live cell imaging was recorded using a Nikon Eclipse 300 inverted micro-
include the N-19 series (against NH2-terminal residues) and the C-20 scope with a heated stage equipped with ⫻40 (0.6) and ⫻60 (0.7) objec-
series (against COOH-terminal residues) for both murine Ran (sc-1155 tives and a Nikon F70 camera with ISO400 films. Images were taken at
and sc-1156) and RCC1 (sc-1161 and sc-1162) and the M-19 (sc-1159) 2-min intervals. The length of mitotic substages was calculated from live
antibody against the CKLEALSVREAREEAEEKSE unique peptide of mu- cell imaging. Mitotic progression of injected cells was followed in vivo until
rine RanBP1. To further ensure that the anti-RanBP1 antibody did not completion and in all cases for at least 90 min after injection. Cells were
react with other RBDs, Western blot experiments were set up using then fixed and stained with DAPI. In some experiments, cells were injected
GST-fusion proteins with single RBDs (RBD1– 4) from the RanBP2 protein, with the anti-RanBP1 antibody, exposed immediately to NOC, fixed after
and no cross-reactivity was detected. Purified GST-RBDs and their re- 10 min, and processed for immunostaining as described above using
spective antibodies were kindly provided by Paulo Ferreira (Pharmacology anti-␣-tubulin antibody to visualize the spindle and FITC-conjugated sec-
Department, Medical College of Wisconsin, Milwaukee, WI). Anti-p21 ondary antibody to reveal anti-RanBP1.
antibody (sc-397) was from Santa Cruz Biotechnology. Protein extracts
were prepared from staged cell cultures, electrophoresed, and electro-
blotted as described previously (21). Filters were incubated with 0.5 ␮g/ml
Acknowledgments
primary antibody in 5% low-fat milk in TBST buffer [10 mM Tris (pH 8.0,) We are grateful to Rosamaria Mangiacasale for help with flow cytom-
150 mM NaCl, and 0.1% Tween 20] for 1 h at room temperature, washed, etry and to Antonella Palena for many contributions to this work. We also
and further incubated for 40 min with secondary horseradish peroxidase- thank Minuro Yoshida for the gift of LMB, Paulo Ferreira for the gift of
conjugated antibody (Santa Cruz Biotechnology). Immunoblots were re- RanBP2-derived Ran-binding domains and antibodies, and Takeharu
vealed using the enhanced chemiluminescence detection system (Amer- Nishimoto for anti-RCC1 antibodies.
sham/Pharmacia).
Indirect IF. Components of the Ran network were examined using
various antibodies and fixation procedures. Affinity-purified antibodies
References
(Santa Cruz Biotechnology) are specified above; clones directed against 1. Rush, M. G., Drivas, G., and D’Eustachio, P. The small nuclear GTPase
the NH2-terminal and the COOH-terminal region of each protein were Ran: how much does it run? Bioessays, 18: 103–112, 1996.
used. RCC1 was also examined using antibodies against X. laevis RCC1 2. Sazer, S. The search for the primary function of the Ran GTPase
(45) kindly provided by Takeharu Nishimoto (Molecular Biology Depart- continues. Trends Cell Biol., 6: 81– 85, 1996.
ment, Graduate School of Medical Science, Fukuoka, Japan). Similarly, 3. Nishimoto, T. A new role of Ran GTPase. Biochem. Biophys. Res.
RanBP1 IF experiments were carried out using either affinity-purified Commun., 262: 571–574, 1999.
antipeptide antibody (M-19; Santa Cruz Biotechnology) or a polyclonal
4. Sazer, S., and Dasso, M. The Ran decathlon: multiple roles of Ran.
antibody against the entire murine RanBP1 generated in our laboratory
J. Cell Sci., 113: 1111–1118, 2000.
(28). All tested antibodies gave similar results. MPM-2 antibody against
G2/mitotic phosphoepitopes (29) was from DAKO. Anti-␣-tubulin antibody 5. Bischoff, R. F., Klebe, C., Kretschmer, J., Wittinghofer, A., and
was from Amersham/Pharmacia. Cells were fixed by two washes in cold Ponstingl, H. RanGAP induces GTPase activity of nuclear Ras-related
absolute methanol or, alternatively, 3% paraformaldehyde under the con- Ran. Proc. Natl. Acad. Sci. USA, 91: 2787–2791, 1994.
ditions described in Refs. 36 and 45. In cotransfection experiments with 6. Bischoff, F. R., and Ponstingl, H. Catalysis of guanine exchange on Ran
the pGFP construct, cells were fixed in 4% paraformaldehyde. Cell prep- by the mitotic regulator RCC1. Nature (Lond.), 354: 80 – 82, 1991.
Cell Growth & Differentiation 465

7. Coutavas, E., Ren, M., Oppenheim, J., D’Eustachio, P., and Rush, M. 28. Battistoni, A., Guarguaglini, G., Degrassi, F., Pittoggi, C., Palena, A.,
Characterization of proteins that interact with the cell-cycle regulatory Di Matteo, G., Pisano, C., Cundari, E., and Lavia, P. Deregulated expres-
protein Ran/TC4. Nature (Lond.), 366: 585–587, 1993. sion of the RanBP1 gene alters cell cycle progression in murine fibro-
8. Richards, S. A., Lounsbury, K., and Macara, I. The C-terminus of the blasts. J. Cell Sci., 110: 2345–2357, 1997.
nuclear Ran/TC4 GTPase stabilizes the GDP-bound state and mediates 29. Davis, F. M., Tsao, T. Y., Fowler, S. K., and Rao, P. N. Monoclonal
interaction with RCC1, Ran-GAP and Htf9-a/RanBP1. J. Biol. Chem., 270: antibodies to mitotic cells. Proc. Natl. Acad. Sci. USA, 80: 2926 –2930,
14405–14411, 1995. 1983.
9. Bischoff, R. F., Krebber, H., Smirnova, E., Dong, W., and Ponstingl, H. 30. Richards, S. A., Lounsbury, K. M., Carey, K. L., and Macara, I. A
Co-activation of RanGTPase and inhibition of GTP dissociation by Ran- nuclear export signal is essential for the cytosolic localization of the Ran
GTP binding protein RanBP1. EMBO J., 14: 705–715, 1995. binding protein, RanBP1. J. Cell Biol., 134: 1157–1168, 1996.
10. Dasso, M., and Pu, R. T. Nuclear transport: run by Ran? Am. J. Hum. 31. Pasquinelli, A. E., Powers, M. A., Lund, E., Forbes, D., and Dahlberg,
Genet., 63: 311–316, 1998. J. E. Inhibition of mRNA export in vertebrate cells by nuclear export signal
11. Adam, S. A. Transport pathways of macromolecules between the conjugates. Proc. Natl. Acad. Sci. USA, 94: 14394 –14399, 1997.
nucleus and the cytoplasm. Curr. Opin. Cell Biol., 11: 402– 406, 1999. 32. Fukuda, M., Asano, S., Nakamura, T., Adachi, M., Yoshida, M.,
12. Stochaj, U., and Rother, K. L. Nucleocytoplasmic trafficking of pro- Yanagida, M., and Nishida, E. CRM1 is responsible for intracellular trans-
teins: with or without Ran? Bioessays, 21: 579 –589, 1999. port mediated by the nuclear export signal. Nature (Lond.), 390: 308 –311,
13. Ouspenski, I. I. A RanBP1 mutation which does not visibly affect 1997.
nuclear import may reveal additional functions of the Ran GTPase system. 33. Kudo, N., Khochbin, S., Nishi, K., Kitano, K., Yanagida, M., Yoshida,
Exp. Cell Res., 244: 171–183, 1998. M., and Horinouchi, S. Molecular cloning and cell cycle-dependent ex-
14. Nakamura, M., Masuda, H., Horii, J., Kuma, K., Yokoyama, N., Ohba, pression of mammalian CRM1, a protein involved in nuclear export of
T., Nishitani, H., Miyata, T., Tanaka, M., and Nishimoto, T. When overex- proteins. J. Biol. Chem., 272: 29742–29751, 1997.
pressed, a novel centrosomal protein, RanBPM, causes ectopic microtu- 34. Ren, M., Drivas, G., D’Eustachio, P., and Rush, M. G. Ran/TC4: a
bule nucleation similar to ␥-tubulin. J. Cell Biol., 143: 1041–1052, 1998. small nuclear GTP-binding protein that regulates DNA synthesis. J. Cell
15. Kahana, J. A., and Cleveland, D. W. Beyond nuclear transport. Ran- Biol., 120: 313–323, 1993.
GTP as a determinant of spindle assembly. J. Cell Biol., 146: 1205–1210, 35. Zhang, C., Hughes, M., and Clarke, P. R. Ran-GTP stabilizes micro-
1999. tubule asters and inhibits nuclear assembly in Xenopus egg extracts.
16. Carazo-Salas, R. E., Guarguaglini, G., Gruss, O. J., Segref, A., J. Cell Sci., 112: 2453–2461, 1999.
Karsenti, E., and Mattaj, I. W. Generation of GTP-bound Ran by RCC1 is 36. Azuma, Y., Hachiya, T., and Nishimoto, T. Inhibition by anti-RCC1
required for chromatin-induced mitotic spindle formation. Nature (Lond.), monoclonal antibodies of RCC1-stimulated guanine nucleotide exchange
400: 178 –181, 1999. on Ran GTPase. J. Biochem. (Tokyo), 122: 1133–1138, 1997.
17. Kalab, P., Pu, R. T., and Dasso, M. The Ran GTPase regulates mitotic 37. Pu, R. T., and Dasso, M. The balance of RanBP1 and RCC1 is critical
spindle assembly. Curr. Biol., 9: 481– 484, 1999. for nuclear assembly and nuclear transport. Mol. Biol. Cell, 10: 1955–
18. Ohba, T., Nakamura, M., Nishitani, H., and Nishimoto, T. Self- 1970, 1997.
organization of microtubule asters induced in Xenopus egg extracts by 38. Demeter, J., Morphew, M., and Sazer, S. A mutation in the RCC1-
GTP-bound Ran. Science (Washington DC), 284: 1356 –1358, 1999. related protein pim1 results in nuclear envelope fragmentation in fission
19. Wilde, A., and Zheng, Y. Stimulation of microtubule aster formation yeast. Proc. Natl. Acad. Sci. USA, 92: 1436 –1440, 1995.
and spindle assembly by the small GTPase Ran. Science (Washington 39. He, X., Hayashi, N., Walcott, N. G., Azuma, Y., Patterson, T. E.,
DC), 284: 1359 –1362, 1999. Bischoff, F. R., Nishimoto, T., and Sazer, S. The identification of cDNAs
20. Di Matteo, G., Fuschi, P., Zerfass, K., Moretti, S., Ricordy, R., Cen- that affect the mitosis-to-interphase transition in Schizosaccharomyces
ciarelli, C., Tripodi, M., Jansen-Dürr, P., and Lavia, P. Transcriptional pombe, including sbp1, which encodes a spi1p-GTP-binding protein.
control of the Htf9-a/RanBP1 gene during the cell cycle. Cell Growth Genetics, 148: 645– 656, 1998.
Differ., 6: 1213–1224, 1995. 40. Matynia, A., Dimitrov, K., Mueller, U., He, X., and Sazer, S. Perturba-
21. Di Matteo, G., Salerno, M., Guarguaglini, G., Di Fiore, B., Palitti, F., tions in the spi1p GTPase cycle of Schizosaccharomyces pombe through
and Lavia, P. Interactions with single-stranded and double-stranded DNA- its GTPase-activating protein and guanine nucleotide exchange factor
binding factors and alternative promoter conformation upon transcrip- components result in similar phenotypic consequences. Mol. Cell. Biol.,
tional activation of the Htf9-a/RanBP1 and Htf9-c genes. J. Biol. Chem., 16: 6352– 6362, 1996.
273: 495–505, 1998. 41. Nicolas, F., Zhang, C., Hughes, M., Goldberg, M., Watton, S., and
22. Guarguaglini, G., Battistoni, A., Pittoggi, C., Di Matteo, G., Di Fiore, B., Clarke, P. Xenopus Ran-binding protein 1: molecular interactions and
and Lavia, P. Expression of the murine RanBP1 and Htf9-c genes is effects on nuclear assembly in Xenopus egg extracts. J. Cell Sci., 110:
regulated from a shared bidirectional promoter during cell cycle progres- 3019 –3330, 1997.
sion. Biochem. J., 325: 277–286, 1997. 42. Meraldi, P., Lukas, J., Fry, A. M., Bartek, J., and Nigg, E. A. Centro-
23. Di Fiore, B., Guarguaglini, G., Palena, A., Kerkhoven, R. M., Bernards, some duplication in mammalian somatic cells requires E2F and Cdk2-
R., and Lavia, P. Two E2F-binding sites independently control growth- cyclin A. Nat. Cell Biol., 1: 88 –93, 1999.
regulated and cell-cycle regulated transcription of the Htf9-a/RanBP1 43. Matunis, M. J., Coutavas, E., and Blobel, G. A novel ubiquitin-like
gene. J. Biol. Chem., 274: 10339 –10348, 1999. modification modulates the partitioning of the Ran-GTPase-activating
24. Muller, H., and Helin, K. The E2F transcription factors: key regulators protein RanGAP between the cytosol and the nuclear pore complex.
of cell proliferation. Biochim. Biophys. Acta, 1470: M1–M12, 2000. J. Cell Biol., 135: 1457–1470, 1996.
25. Dyson, N. The regulation of E2F by pRB-family proteins. Genes Dev., 44. Balczon, R., Varden, C. E., and Schroer, T. A. Role for microtubules in
12: 2245–2262, 1998. centrosome doubling in Chinese hamster ovary cells. Cell Motil. Cytoskel-
26. Nevins, J. R. Toward an understanding of the functional complexity of eton, 42: 60 –72, 1999.
the E2F and retinoblastoma families. Cell Growth Differ., 9: 585–593, 45. Ohtsubo, M., Yoshida, T., Seino, H., Nishitani, H., Clark, K. L.,
1998. Sprague, G. F., Jr., Frasch, M., and Nishimoto, T. Mutation of the hamster
27. Lavia, P., and Jansen-Dürr, P. E2F target genes and cell cycle check- cell cycle gene RCC1 is complemented by the homologous genes of
point control. Bioessays, 21: 221–230, 1999. Drosophila and S. cerevisiae. EMBO J., 10: 1265–1273, 1991.

Das könnte Ihnen auch gefallen