Download Nuclear and nucleolar targeting of human ribosomal protein

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

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

Document related concepts

SR protein wikipedia , lookup

Endomembrane system wikipedia , lookup

Magnesium transporter wikipedia , lookup

Protein phosphorylation wikipedia , lookup

Protein structure prediction wikipedia , lookup

Amitosis wikipedia , lookup

Protein moonlighting wikipedia , lookup

Protein wikipedia , lookup

Signal transduction wikipedia , lookup

Intrinsically disordered proteins wikipedia , lookup

Cyclol wikipedia , lookup

Cell nucleus wikipedia , lookup

Protein–protein interaction wikipedia , lookup

Nuclear magnetic resonance spectroscopy of proteins wikipedia , lookup

List of types of proteins wikipedia , lookup

Proteolysis wikipedia , lookup

Transcript
Oncogene (1999) 18, 1503 ± 1514
ã 1999 Stockton Press All rights reserved 0950 ± 9232/99 $12.00
http://www.stockton-press.co.uk/onc
Nuclear and nucleolar targeting of human ribosomal protein S25: Common
features shared with HIV-1 regulatory proteins
Satoshi Kubota1, Terry D Copeland2 and Roger J Pomerantz1
1
The Dorrance H. Hamilton Laboratories, Center for Human Virology, Division of Infectious Diseases, Department of Medicine,
Je€erson Medical College, Thomas Je€erson University, Philadelphia, Pennsylvania 19107; and 2ABL-Basic Research Program
NCI-Frederick Cancer Research and Development Center P.O. Box B, Frederick, Maryland 21702, USA
The nuclear and nucleolar targeting properties of human
ribosomal protein S25 (RPS25) were analysed by the
expression of epitope-tagged RPS25 cDNAs in Cos-1
cells. The tagged RPS25 was localized to the cell
nucleus, with a strong predominance in the nucleolus. At
the amino terminus of RPS25, two stretches of highly
basic residues juxtapose. This con®guration shares
common features with the nucleolar targeting signals
(NOS) of lentiviral RNA-binding transactivators, including human immunode®ciency viruses' (HIV) Rev proteins. Deletion and site-directed mutational analyses
demonstrated that the ®rst NOS-like stretch is dispensable for both nuclear and nucleolar localization of
RPS25, and that the nuclear targeting signal is located
within the second NOS-like stretch. It has also been
suggested that a set of continuous basic residues and the
total number of basic residues should be required for
nucleolar targeting. Signal-mediated nuclear/nucleolar
targeting was further characterized by the construction
and expression of a variety of chimeric constructs,
utilizing three di€erent backbones with RPS25 cDNA
fragments. Immuno¯uorescence analyses demonstrated a
17 residue peptide of RPS25 as a potential nuclear/
nucleolar targeting signal. The identi®ed peptide signal
may belong to a putative subclass of NOS, characterized
by compact structure, together with lentiviral RNAbinding transactivators.
Keywords: ribosome; protein; nuclear targeting; rev;
HIV-1
Introduction
The mechanism of nuclear translocation of proteins
has been characterized in signi®cant detail. It has been
shown that nuclear import through the nuclear pore
complexes consists of two distinct steps, in which
karyopherin/importin and a small GTP-binding protein, Ran/TC4, play central roles (Akey and Goldfarb,
1989; Melchior and Gerace, 1995; GoÈrlich and Mattaj,
1996). A number of nuclear localization signals (NLS)
have been identi®ed in a variety of nuclear proteins.
Recently, alternative pathways of nuclear import, one
of which is employed speci®cally by certain ribosomal
proteins in yeast, have been reported as well (Siomi
and Dreyfuss, 1995; Goldfarb, 1997; Rout et al., 1997).
Correspondence: RJ Pomerantz
Received 27 May 1998; revised 16 September 1998; accepted 16
September 1998
In contrast, targeting of proteins into the cell nucleolus
by single distinct signals has been described in a limited
number of proteins, primarily in RNA-binding
transactivator proteins of human retroviruses ±
human immunode®ciency virus type 1 (HIV-1) and
human T-cell leukemia virus type I (HTLV-I) (Siomi et
al., 1988, 1990; Kubota et al., 1989; Dang and Lee,
1989). The functional signi®cance of the nucleolar
localization of these proteins is still controversial,
although a possible role of the nucleolus as a storage
site for the proteins is suggested in the case of the HIV1 Rev protein (Kubota et al., 1996), which shuttles
between the cytoplasm and the nucleus, to enable the
nuclear export of unspliced viral mRNAs (Malim et
al., 1989a,b; Fischer et al., 1994; Kalland et al., 1994;
Meyer and Malim, 1994; Richard et al., 1994). Despite
its evident functional signi®cance as a pre-event of
ribosomal assembly, nucleolar targeting of human
ribosomal proteins has been characterized in only a
few members, S6, S14 and L7a (Schmidt et al., 1995;
Martin-Nieto and Roufa, 1997; Russo et al., 1997).
Therefore, further analyses of the nucleolar targeting of
other ribosomal proteins seem to be required to gain
insights into not only ribosomal biogenesis, but also
the role of the nucleolar localization of such RNAbinding transactivators.
Ribosomes can be regarded as huge RNA-protein
complexes, which are precisely organized to allow
ecient protein biosynthesis. In eukaryotic cells, the
biogenesis of ribosomes is carried out in the cell
nucleolus (Sommerville, 1986; Warner, 1990), under the
well-regulated expression of at least 80 ribosomal
proteins, which are coordinated with cellular metabolism (Mager, 1988). The expression of eukaryotic
ribosomal proteins has been shown to be controlled
at both transcriptional (Agrawal and Bowman, 1987;
Tasheva and Roufa, 1995) and post-transcriptional
sites (Gritz et al., 1985; Mager, 1988; Aloni et al.,
1992), predominantly at the level of translation. After
expression in the cytoplasm, ribosomal proteins
migrate into the nucleus, and reach the nucleolus.
The transcription of pre-ribosomal RNAs (rRNA) also
takes place in the nucleolus (Sollner-Webb and
Mougey, 1991). Afterwards, the ribosomal proteins
and rRNAs assemble together in the nucleolus.
Eventually, assembled ribosomal subunits are exported out of the nucleus to be engaged in protein
synthesis in the cytoplasm. Therefore, ribosomal
proteins are initially imported into the nucleus
towards the nucleolus, and later, return to the
cytoplasm as a part of ribosome. However, as stated
above, it has not been well-investigated how a vast
number of human ribosomal proteins can accumulate
Nuclear targeting of S25
S Kubota et al
1504
in the nucleolus, prior to ribosomal assembly, although
it is one of the fundamental steps to clarify in the
mechanism(s) of ribosomal biogenesis.
Human ribosomal protein S25 (RPS25) is one of the
highly basic members of human ribosomal proteins. Of
note, the RPS25 gene was reported to be highly overexpressed in human leukemia (HL60) cells isolated for
adriamycin-resistance, thus its cDNA was cloned and
identi®ed from those cells (Li et al., 1991). Cloning and
sequencing of the cDNA revealed RPS25 as a protein
of 125 amino acid residues, which shares 56%
homology to yeast ribosomal protein S31 (Li et al.,
1991). A recent report assigned the chromosomal
location of the RPS25 gene to 11q23.3 (Imai et al.,
1994). Nevertheless, no further investigation has been
reported until now. Recently, we have determined that
RPS25 possesses common features with the NOS of the
lentiviral RNA-binding transactivators, Rev proteins.
Most interestingly, another ribosomal protein, L32 of
yeast, has been shown to bind the structured target of
its own mRNA to inhibit splicing (Li et al., 1995),
which is quite similar to the function of the lentiviral
Rev proteins. As ®rst steps in the functional
characterization, we set up a system for the expression
and analysis of RPS25, focusing on the primary preassembly event, pursued its nuclear/nucleolar targeting
property, and demonstrated critical features involved in
nuclear/nucleolar targeting, in comparison with other
NOSs of lentiviral RNA-binding transactivators.
Results
Expression and characterization of an epitope-tagged
RPS25
In general, it is dicult to raise a highly speci®c
antibody against a ubiquitous protein in experimental
animals. Indeed, all the recent similar works on human
ribosomal proteins have employed either epitopetagging systems (Moreland et al., 1985; Michael and
Dreyfuss, 1996; Martin-Nieto and Roufa, 1997) or
even b-galactosidase fusions (Schmidt et al., 1995;
Russo et al., 1997). Following the more well-accepted
former strategy, an RPS25 expression vector, pcFLRPS25, was designed (Figure 1a). In our system, the
expressed RPS25 was tagged with an eight residue
epitope of a murine monoclonal antibody (Anti-FLAG
M2), to enable its detection. Cos-1 cells were
transfected with pcFL-RPS25, and the transient
expression of the FLAG-RPS25 protein was examined
by immunoblotting using the corresponding monoclonal antibody. As displayed in Figure 1b a distinct
signal was speci®cally detected in the lysate of Cos-1
cells transfected with pcFL-RPS25. As expected, the
apparent molecular weight of the expressed RPS25 in
SDS ± PAGE was reasonably larger than the calculated
molecular weight of RPS25 (14 KD), which is due to
the addition of the FLAG epitope and linker residues
(2 KD), and its high positive charge density. Immuno¯uorescence analyses of the transfected cells clearly
demonstrated the expression and nuclear localization
of RPS25, with a dominant accumulation in the cell
nucleoli (Figure 1c and d) and also weak signals from
the cytoplasm (Figures 1c and 3b). The expression of
exogenous FLAG-RPS25 did not seem to cause major
damage in nucleolar structure, which had been
observed by the over-expression of another nucleolar
protein, HIV-1 Rev (Nosaka et al., 1993). As
illustrated in Figure 1e, the morphology of the
nucleoli appeared to be unaltered with a variety of
levels of FLAG-RPS25 expression. Similar experiments
with human 293T cells also yielded the same results
(data not illustrated).
Since the added FLAG epitope [DYKDDDDK]
contains a net negative charge, it may be of concern
Nuclear targeting of S25
S Kubota et al
1505
Figure 1 Establishment of a FLAG-tagged RPS25 expression and detection system. (a) Structure of the plasmid for the expression
of a FLAG-tagged RPS25 (FL-RPS25). Abbreviations; CMV: cytomegalovirus promoter, FLAG: the FLAG epitope-encoding
region (in the stippled box). The cDNA for RPS25 was a composite of synthetic oligonucleotides (solid black box) and a DNA
fragment from a cDNA clone of HeLa cells (hatched box with the name of the clone). Numbers over a box indicate codon numbers
counted from the initiation codon of the cDNA. (b) Expression of RPS25 in Cos-1 cells detected by Western blotting analysis. An
arrowhead indicates the speci®c signal. Positions of protein molecular weight standards are shown on the left of the panel. (c)
Expression of RPS25 in Cos-1 cells detected by indirect immuno¯uorescence analysis. Non-transfected cells, around the three cells
with positive signal, serve as internal negative controls. Some cells in this ®eld also represent the morphological variation among
their two dimensional views. Apparent sizes of the cells and the nuclei are, frequently, not uniform in Cos-1 cell cultures. (d)
Immuno¯uorescence analysis of the Cos-1 cells expressing RPS25 at high power magni®cation (61000), in comparison with phasecontrast photomicrographs of the same cells. (e) No major morphological alteration of the nucleoli by the expression of FLAGRPS25 at a variety of levels. Individual cells expressing low to high (top to bottom) levels of FLAG-RPS25 are displayed. (f)
Veri®cation of the subcellular distribution of RPS25 by a Myc-tagged expression and detection system. Structure of the plasmid to
express a carboxy-terminal Myc-tagged RPS25 is shown at the top. Abbreviations; SV: SV40 promoter, Myc: an oligonucleotide
encoding a c-Myc epitope (Siomi and Dreyfuss, 1995; Dang and Lee, 1989; Kalderon et al., 1984). Numbers indicate the same items
as shown in a. Immunostaining of the transfected Cos-1 cells was performed with a murine monoclonal antibody (9E10) against the
Myc epitope and is illustrated
Nuclear targeting of S25
S Kubota et al
1506
that such an acidic stretch of amino acid residues
might have some interaction with a basic stretch of
amino acid residues of RPS25, to a€ect its native
subcellular localization. To verify if such an undesirable e€ect is caused by the FLAG epitope, control
experiments were carried out. The other RPS25
expression plasmid, entitled pCHRPS25-Myc, was
constructed to express an RPS25, tagged with an
epitope of c-Myc protein at the carboxy terminus
(Figure 1f). This Myc-tag can be detected by a murine
monoclonal antibody, which has been previously
utilized (Siomi and Dreyfuss, 1995; Dang and Lee,
1989). Transfection of pCHRPS25-Myc into Cos-1
cells and subsequent immuno¯uorescence analysis
revealed the same subcellular distribution of the Myctagged RPS25 as the FLAG-tagged RPS25, suggesting
that the FLAG epitope at the amino terminus should
not a€ect the intact subcellular localization of RPS25.
In addition, the e€ect of the FLAG epitope on another
nucleolar-localizing protein, HIV-1 Rev, was evaluated.
Of importance, the subcellular distribution of the Rev,
which was tagged by the FLAG epitope, was
demonstrated to be the same as the wild-type Rev
protein (data not illustrated). The results of these
control experiments strongly suggest the utility of the
FLAG tag in studying nuclear-nucleolar targeting of
proteins.
corresponding region, for nuclear/nucleolar localization, since a majority of classical NLSs do contain such
an arginine residue. In the mutant, pcFL-RPS25m1,
the arginine codon was changed to a glycine codon
(Figure 4a). Despite the mutation, in comparison with
the wild-type, no signi®cant change in subcellular
localization was observed (Figure 4b), which indicates
that the arginine residue itself is not critical for either
nuclear, or nucleolar, localization of RPS25.
In contrast to the m1-mutation, extensive mutation
of ten residues, including the arginine, to unrelated
eight residues (two residues deleted) has completely
abolished the dominant nuclear targeting of RPS25.
This second mutant, entitled pcFL-RPS25m2, expressed an RPS25 protein with a di€use intracellular
distribution, both inside or outside of the nuclear
membrane (Figure 4). Apparently, the immunofluorescent signal from and around the nucleus frequently
a
The region required for nuclear localization of RPS25
To initially estimate which motif is necessary for the
nuclear localization of RPS25, six deletion cDNA
mutants of the FLAG-RPS25 protein were constructed. Three constructs were generated to express
the RPS25 mutants with amino-terminal deletions, and
the others for carboxy-terminal deletion mutants
(Figures 2a and 3a). These constructs were transfected
and characterized in the same manner as the wild-type
protein. Unexpectedly, two amino-terminal deletion
mutants, dN1 and dN2, demonstrated approximately
the same subcellular distribution as the wild-type by
immuno¯uorescence analyses (Figure 2b). Namely, the
®rst lysine-rich stretch (residues 4 ± 20), which contains
nine lysine residues out of 17, was entirely dispensable
for nuclear/nucleolar targeting. Extension of the
deletion up to residue 43 completely abolished the
proper nuclear/nucleolar localization of RPS25, and a
di€use distribution throughout the cell was observed
(dN3: Figure 2b). Similar analyses with the other three
carboxy-terminal deletion mutants have revealed that
removal of the 53 carboxy-terminal residues did not
signi®cantly alter the nuclear/nucleolar targeting of
RPS25 (dC1 and dC2: Figure 3b), whereas the other
mutant which lacks the 94 carboxy-terminal residues
lost nuclear targeting ability (dC3: Figure 3b). Therefore, the nuclear localization signal (NLS) was thought
to reside between residues 21 and 72 of the RPS25
protein.
Extensive analyses were further carried out by sitedirected mutagenesis of RPS25 cDNA. Since there is
no NLS-like basic residue stretch between residues 42
and 72, we suspected a potential NLS/NOS between
residues 21 and 42 of RPS25. As such, the mutagenesis
was targeted to this region. The ®rst point mutant was
constructed to examine the requirements of the
arginine residue, which is the only one found in the
b
Figure 2 Amino-terminal deletion mutants of RPS25, and their
subcellular localization. (a) Schematic representation of the
primary structures of the deletion mutants. At the top, the
amino acid sequence of the trimmed area is shown in single letter
codes, in which basic residues are shown in bold letters. Numbers
above boxes denote residue numbers counted from the initiation
methionine of RPS25. Deleted regions are represented as solid
lines. (b) Immuno¯uorescence analysis of the amino-terminal
deletion mutants, by an anti-FLAG antibody. The names of the
expressed and detected proteins are shown above the photomicrographs
Nuclear targeting of S25
S Kubota et al
a
exclusion (Figure 4b and c). The distinct nuclear
localization and the intensity of the nuclear signal in
each cell, was not at all a€ected by this minor deletion,
which indicates the continuous cluster of lysine residues
plays a role, not in the nuclear, but rather in the
nucleolar localization of the RPS25 protein.
Nuclear/nucleolar targeting signal of RPS25
b
Figure 3 Carboxy-terminal deletion mutants of RPS25 and their
subcellular localization. (a) Representation of the primary
structures of the deletion mutants. Numbers above boxes denote
residue numbers counted from the initiation methionine of
RPS25. Deleted regions are represented as solid lines. At the
end of each line, some extra residues, which were added through
construction procedures, are also shown in single letter codes. (b)
Detection of the carboxy-terminal deletion mutants by immuno¯uorescence analyses. The names of the deletion mutants are
shown above the photomicrographs
seems to be relatively stronger, in the case of mutants
m2, as well as dN3 and dC3, which is probably due to
the even distribution inside and outside of the nuclei
and the three-dimensional shape of the cells (Figures 2,
3 and 4). Thus, at least some of the ten residues of
RPS25 are required for its nuclear localization.
Nucleolar targeting property of RPS25
As observed in Figure 2b, the removal of aminoterminal 43 residues caused nucleolar exclusion as well
as the loss of nuclear targeting in RPS25. Furthermore,
creation and characterization of another RPS25
mutant yielded yet another insight into a sequence
that a€ects, not the nuclear translocation, but only the
nucleolar accumulation of RPS25. The mutant,
RPS25m3, harbors a deletion of three continuous
lysine residues in this speci®c area (Figure 4a).
Expression and detection of this mutant showed
signi®cant attenuation of nucleolar accumulation
caused by the mutation, despite no evident nucleolar
In order to identify the NLS/NOS of RPS25, three
di€erent fusion constructs were generated. As an initial
step, the 41 amino-terminal residues with the FLAG
epitope, which includes two NOS-like motifs, were
fused to a short fragment of the rat preproinsulin two
gene at the amino terminus (Figure 5a). Expression
and immuno¯uorescence analysis demonstrated a clear
nuclear/nucleolar localization of this small fusion
protein, FL-NOQDINS (Figure 5b). As a control, a
similar fusion construct with the corresponding region
from an RPS25 mutant, pcFL-RPS25m2 (Figure 4),
the nuclear/nucleolar targeting property of which is
inactivated by mutation, was prepared (Figure 5a). A
di€use intracellular distribution pattern was observed
with transient expression and immuno¯uorescence
analysis of the mutant plasmid, entitled pcFLNOQm2DINS. These ®ndings indicate a NOS within
residues 1 ± 41 of RPS25. In order to further examine
the ability of the amino terminal 41 residues to
function as a NOS for a larger protein, E. coli bgalactosidase was next chosen as a fusion counterpart
for the RPS25 fragment (Figure 6a). The b-galactosidase is a bacterial 116 kD protein, which is thus found
outside of the nucleus when it is expressed in
eukaryotic cells. This was also reproducible in our
laboratory (Figure 6b: CH110). In contrast, once the
41 amino-terminal residues of RPS25 were fused to its
amino terminus, the fusion protein was transported
into the nucleus, and also accumulated in the
nucleolus, although at a reduced level compared to
the insulin fusion protein (Figure 6b: CHNOQ). This
®nding indicates that the RPS25 fragment functioned
as an active NLS/NOS at the amino terminus. The
other RPS25-b-galactosidase fusion protein, with
residues 1 ± 21 of RPS25, failed to localize in the
nucleus, in concordance with the mutational analysis of
RPS25 (Figure 6b: CH-NOH).
Although the b-galactosidase fusion system has
been widely accepted as a marker for nuclear
targeting of proteins, it has been suspected that this
prokaryotic protein partially migrates through the
nuclear membrane, which may make results ambiguous (Siomi and Dreyfuss, 1995; Kalderon et al.,
1984). Therefore, it may not be a perfect reporter for
nuclear/nucleolar targeting. Thus, to gain more
reliability using a di€erent system, we constructed
another fusion-protein expression plasmid, in the
backbone of chicken muscle pyruvate kinase (PK).
PK has been shown to be stably excluded from the
nucleus, hence the utility of a myc-tagged PK cDNA,
as a marker gene for nuclear targeting, has already
been established (Siomi and Dreyfuss, 1995; Dang
and Lee, 1989; Kalderon et al., 1984). Immunofluorescence analyses of PK, expressed in Cos-1 cells, has
shown extremely clear nuclear-exclusion, which seems
more striking and reliable than b-galactosidase
(Figure 7).
1507
Nuclear targeting of S25
S Kubota et al
1508
Based on the data from the amino-terminal
deletions of RPS25, RSP25-insulin and RPS25-bgalactosidase fusion proteins, we hypothesized that
residues 25 ± 41 of RPS25 should involve a NLS/NOS.
For veri®cation, we fused the RPS25 fragment to the
carboxy terminus of the Myc-PK protein (Myc-PKNOR) (Figure 7a). During the construction procedure
of pcDNA3Myc-PK-NOR, the carboxy terminal 86
residues of PK were removed. In such a case,
although unlikely, there still was a formal possibility
that removal of the 86 residues might a€ect the
subcellular localization of PK itself. Therefore, as a
further control experiment, another plasmid used to
express PK, with a carboxy terminal truncation of 87
residues, was constructed. Immunostaining of these
a
expressed proteins demonstrated that, as anticipated,
the fusion protein was capable of migrating into the
nucleus and accumulating in the nucleolus, while the
PK with carboxy terminal deletion revealed the same
intracellular distribution pattern, with clear nuclear
exclusion, as the full-length PK (Figure 7b). The
subcellular distribution of Myc-PK-NOR was not
precisely uniform throughout all of the expressing
cells. Some cells demonstrated complete nuclear
accumulation, while others still revealed a cytoplasmic signal, as well as a nuclear/nucleolar signal
(Figure 7). However, it was evident that residues
25 ± 41 of RPS25 acted as a NLS/NOS at the carboxy
terminus of Myc-PK, to actively guide this cytoplasmic protein into the nucleus and the nucleolus.
c
b
Figure 4 Site-directed mutational analyses targeted to the region
which is critical for the nuclear and nucleolar targeting of RPS25.
(a) Structure of the mutants. Primary structures of the native
RPS25 with the FLAG epitope, and mutants, are represented.
Numbers over a box denote residue numbers, as seen in Figures 1
and 2. Amino acid residues around the a€ected area are shown in
single letter codes. Basic and mutated residues are shown in bold
letters. Deletion and amino acid residue substitutions are
represented as a solid bar, and a hatched box, respectively. (b)
Subcellular distribution of the mutants in comparison with the
wild-type protein. Abbreviated names of intact and mutant
proteins are shown above the photomicrographs. (c) Attentuated
nucleolar accumulation of the mutant, m3. Two additional
examples with phase-contrast photomicrographs are shown
Nuclear targeting of S25
S Kubota et al
1509
a
b
Figure 5 Insulin fusions with the amino-terminal fragment of
RPS25 and its mutant. (a) Primary structures of the fusion
constructs. A crossed box with `DINS2' represents a carboxyterminal fragment of rat preproinsulin 2. Numbers over the boxes
and other abbreviations represent the same as in Figure 1.
Mutations introduced into FL-NOQm2DINS are described in the
middle of this panel in an analogous manner to Figure 4a. The
code `wt' stands for the wild-type RPS25. The insulin fusion
constructs express proteins with predicted molecular weights of
7.1 ± 7.3 kD. (b) Subcellular location of the fusion proteins
illustrated in a. The names of the fusion proteins are displayed
above each photomicrograph
Figure 7 Pyruvate kinase fusion with the NOS of RPS25. (a)
Primary structure of Myc-tagged chicken muscle pyruvate kinase
and its derivatives. PK and RPS are the abbreviations for
pyruvate kinase, and RPS25, respectively. Numbers above and
below boxes represent residue numbers corresponding to PK and
RPS25, respectively. (b) Subcellular distribution of Myc-tagged
wild-type or truncated PK, with or without the NOS of RPS25
(residue 25 ± 41). Two representative views are shown for the
Myc-tagged RPS25-PK-fusion protein. The name of the expressed
protein is given above each photomicrograph
Discussion
Figure 6 Fusion proteins of b-galactosidase with amino terminal
fragments of RPS25. (a) Primary structures of the b-galactosidase
fusion proteins. Numbers over the boxes represent residue
numbers of RPS25, counted from the initiation methionine.
Crossed boxes indicate assemblies of gene products of gpt, trpS,
and lacZ of E. coli which are involved in the parental and control
plasmid, pCH110 (Pharmacia). The lacZ fusion constructs express
proteins with predicted molecular weights of 116 ± 120 kD. (b)
Subcellular location of the fusion proteins illustrated in a. The
names of the fusion proteins are displayed above each
photomicrograph
The FLAG-epitope tagging system has been initially
developed and utilized to characterize the nuclear/
nucleolar targeting of human RPS25, and enabled us
to perform further mutational analyses to dissect its
structure-function relationships. Using this system, we
have demonstrated the distinct nuclear/nucleolar
localization of RPS25, analysed its sequence requirements, and identi®ed a new NOS in RPS25. This
system is shown to be excellent in analysing the ®rst
dynamic event of the long journey of RPS25 ± from
ribosome, back to ribosome, since nuclear/nucleolar
targeting itself is not to be a€ected by the FLAGRPS25 accumulation that occurs after this initial event.
The utility of this system for the analysis of late events,
after nucleolar-targeting of RPS25, is not yet established, although our preliminary data have suggested
considerable cytoplasmic FLAG-RPS25 (Figure 3b and
subcellular fractionation analysis-data not illustrated),
and a signi®cant, but limited amount of FLAG-RPS25
incorporated in puri®ed ribosomes (data not illustrated). The strong nuclear-dominant distribution of
FLAG-RPS25 may have been caused by the combination of Cos-1 (or 293T) cells and pcFL-RPS25
plasmids. This system yields high levels of transient
protein expression, through the intracellular amplification of plasmids supported by endogenous SV40 large
T antigen. However, along with cell growth, ribosomes
Nuclear targeting of S25
S Kubota et al
1510
are not synthesized in an unlimited fashion. Overexpression of exogenous FLAG-RPS25 should not
increase, or even may cause possible inhibitory e€ects
on overall ribosomal biogenesis; for synthesis of other
components are coordinately controlled. Consequently,
only a limited portion of FLAG-RPS25 could be
incorporated into ribosomes in the present system,
which caused strong nuclear accumulation of the
remaining FLAG-RPS25. Currently, we are improving
this system to obtain lower levels of stable exogenous
RPS25 expression for more natural analysis of postnucleolar events of RPS25, as previously performed for
another ribosomal protein (Martin-Nieto and Roufa,
1997). In those studies, comparative examination of the
distribution of nucleolar components, in the absence or
presence of exogenous RPS25 and its mutants, will be
involved as well, in order to estimate possible e€ects of
those proteins on nucleolar functions.
In this study, a short (17 residue) and distinct NOS
has been identi®ed. Such a compact NOS is not
commonly found in eukaryotic cellular proteins. More
generally, nucleolar localizing properties seem to be
acquired by cooperation of several functional domains
(Russo et al., 1997; Peculis and Gall, 1992; SchmidtZachmann and Nigg, 1993; Yan and MeÂleÁse, 1993;
Michael and Dreyfuss, 1996). Among these proteins,
no common or prototypic NOS motif has been
presented, suggesting the complexity of the nucleolar
localizing activity. Also, investigations of the other
human ribosomal proteins revealed such features in
their nucleolar targeting. In S6, portions of approximately 60 residues were shown as minimum continuous
fragments to confer nuclear/nucleolar targeting, in
which two independent basic amino acid stretches
seemed to cooperate (Schmidt et al., 1995). In the case
of L7a, the amino-terminal 100 residue fragment was
necessary and sucient for the nuclear/nucleolar
targeting, which was also enabled by the combination
of 17 residue amino terminal peptide and another
portion (residue number 52 ± 100) in the fragment
(Russo et al., 1997). However, most interestingly,
several small and distinct NOSs, which are similar to
the NLS/NOS of RPS25, have been identi®ed in
retroviral RNA-binding trans-regulator proteins. In
particular, the Rev protein of HIV-1 possesses a
NOS/RNA-binding domain, which contains a similar
motif to the NLS/NOS of RPS25 (Kubota et al., 1989)
(Figure 8a). HIV-1 Rev is a 116 amino acid nucleocytoplasmic shuttling protein with a predominant
localization in the nucleolus (Malim et al., 1989a;
Kalland et al., 1994; Meyer and Malim, 1994; Richard
et al., 1994), binds to a structured target on viral
unspliced and singly-spliced mRNAs, and enables these
incompletely-spliced RNAs to accumulate in the
cytoplasm for the expression of structural and
enzymatic viral protein precursors (Malim et al.,
1989b; Fischer et al., 1994; Zapp and Green, 1989;
Heaphy et al., 1990; Malim and Cullen, 1991).
The putative NLS/NOSs from the Rev proteins of
HIV-2 and simian immunode®ciency virus (SIV)mac also
retain the basic amino acid residue motif (Figure 8a).
Although belonging to another class of retroviruses,
human T-cell leukemia/lymphoma virus type I (HTLVI) possesses a functional equivalent of Rev, which is
termed Rex (Seiki et al., 1988). A small and distinct
NOS was ®rst identi®ed in this protein (Siomi et al.,
Figure 8 Comparative alignment of the NOS of RPS25 with other
ribosomal and retroviral proteins. Numbers over each amino acid
sequence represent residue numbers of each protein. (a) A basic
residue motif shared among the NOSs of RPS25 and the Rev
proteins of primate lentiviruses. NOSs of simian immunode®ciency
virus (SIV)mac and HIV-2 are putative. Identical residues are
indicated by bars, whereas non-identical basic residues are linked
by dots. Shared motif is indicated at the bottom. `B' stands for a
basic residue. Lower case letters represent the residues shared by
only three proteins. (b) Sequence comparison of identi®ed NOSs.
Continuous clusters of three or more basic amino acid residues are
boxed. Other basic residues are underlined. Total number of basic
residues involved in each NOS is also shown at the right. (c)
Conservation of the NOS sequence of human RPS25 in yeast
RPS31, and a common basic residue motif between a basic domain
of Visna virus Rev and the non-conserved, amino-terminal basic
residue cluster of RPS25. Identical residues and non-identical basic
residues are indicated by bars and dots, respectively. The region of
a basic residue motifs shared by Visna virus Rev and RPS25 and
by the NOS of human RPS25 and its homologue in yeast RPS31,
are framed by the large boxes. Sources of amino acid sequences are
as follows; human RPS25 and yeast RPS31: (Li et al., 1991) HIV-1
(strain BRU:GenBank accession #K02013), HIV-2 (strain
ROD:GenBank accession #M15390), and SIVmac (strain
MM142:GenBank accession #M16403): Visna virus: (Schoborg
and Clements, 1994)
1988). Such a compact NOS can be found in the other
RNA-binding trans-regulator of HIV-1, Tat (Dang and
Lee, 1989; Siomi et al., 1990). The major function of
Tat is to increase the expression of all viral genes
(Sodroski et al., 1985), through the binding to its
structured RNA target on viral gene transcripts. Taken
together, these compact NOSs are commonly characterized by the involvement of a functional NLS
herein, hence they do not require cooperation of the
other functional domain for nuclear targeting. In their
primary structures, although there are signi®cant
variations in sequence, a few common features can be
pointed out. First, all the identi®ed compact NOSs
retain continuous stretches of basic residues; one
continuous stretch of at least four basic residues, or
two continuous stretches of three basic residues.
Second, the total number of basic residues is more
Nuclear targeting of S25
S Kubota et al
than nine (Figure 8b). These features are necessary but
not sucient for nucleolar targeting property. For
example, the other basic residue stretch at the amino
terminus of RPS25 and a highly basic region in RPS6
which are not fully demonstrated as NOSs themselves
(Schmidt et al., 1995), also possess these characteristics.
However, although high positive charge density is not
sucient to provide nucleolar targeting ability,
accumulated mutational analysis data on the de®ned
compact NOSs have already pointed out the critical
requirement of these features for their function (Siomi
et al., 1988; Kubota et al., 1989; Dang and Lee, 1989;
Malim et al., 1989a). Also, in our study, removal of the
continuous lysine residues in the NOS caused
signi®cant loss of nucleolar accumulation. Therefore,
such NOSs, as shown in Figure 8, may be regarded as
members of the compact NOS family, a particular
subclass of NOSs whose majority are composed of
more than one domain which do not juxtapose. Unlike
nuclear localization, the mechanisms for nucleolar
localization appear to be complex. Since no membrane
exists around the nucleolus, it has been believed that
direct or indirect binding to nucleolar components
should play central roles in nucleolar localization.
Thus, nucleolar localization can be accomplished by
three di€erent types of interactions: nucleolar or
nucleolar-associated protein-mediated capture, rDNAmediated capture, and nucleolar RNA-mediated
capture of nucleolar localization signals. In the case
of RPS25, we hypothesized that the nucleolar
localization may be mediated by RNA-protein interactions, as the NOS of RPS25 shares common features
with the NOS of RNA-binding trans-regulators, which
are thought to function also as RNA-binding domains
(Tan et al., 1993). RNA binding analyses are being
performed to investigate interactions between RPS25
and speci®c RNAs. At present, preliminary results
indicate direct binding of the NOS of RPS25 to
ribosomal RNAs (data not illustrated). Our characterizations of RNA binding properties may clarify which
type of molecular interaction(s) plays a central role in
the nucleolar localization of RPS25.
Yeast ribosomal protein S31 (RPS31) is a homologue of human RPS25, with 56% homology in amino
acid sequence (Li et al., 1991). Comparison of the
primary structures of these two ribosomal proteins
reveals interesting characteristics. The NOS region,
particularly the basic residue motif, is quite wellconserved between these proteins (Figure 8c), which
suggests an indispensable functional role of this region
± probably in nuclear/nucleolar targeting for these
moieties. Nevertheless, only limited homology can be
found in the amino-termini of these proteins. Most of
the lysine residues in this area (residues 4 ± 20) of
human RPS25 are absent in the corresponding area of
yeast RPS31. Together with the fact that this region is
not de®nitely required for the nuclear/nucleolar
localization in human RPS25, it is hypothesized that
the region may have evolved towards an accessory
function, that is required only in higher eukaryotes.
Interestingly, this section of RPS25 shares a distinct
motif of basic residues, with the putative RNA-binding
domain of the Visna virus Rev protein (Figure 8c),
which has been shown to be required for multimerization and binding to the target RNA of the
Visna virus Rev protein (Tiley et al., 1991).
RNA-binding by a ribosomal protein is one of the
key interactions involved in the control of ribosomal
protein expression. Bacterial ribosomal proteins S4 and
S15 bind to their own mRNAs and repress translation
(Tang and Drapier, 1990). In yeast, ribosomal protein
L32 (RPL 32) binds to a purine-rich stem loop of its
unspliced mRNA, and blocks its splicing to prevent
over-production of RPL32 (Li et al., 1995; Eng and
Warner, 1991), which has similarities to the interaction
of the Rev proteins and the structured targets on their
unspliced viral mRNAs. Since introns are rare in yeast,
it is plausible that this kind of RNA-binding regulatory
function may have developed more extensively in higher
eukaryotes. As such, it can be suggested that the `Visna
Rev-like' region may have been furnished as an
alternative RNA-binding domain for a speci®c regulatory function, acquired during evolution. It can also be
suggested that the evolutional pathway of RNA-binding
ribosomal proteins, such as yeast L32, may be similar to
the Rev proteins of HIVs and other lenti-retroviruses.
Comparative investigation of the ribosomal proteins of
higher eukaryotes and RNA-binding lentiviral proteins
may yield comprehensive insights into the understanding of both critical families of proteins.
Materials and methods
Construction of an epitope-tagged expression system of RPS25
Basic RPS25 expression plasmid The major portion of
RPS25 cDNA, which encodes the carboxy-terminal 84
amino acid residues, was obtained from an isolated clone
of lZAP II cDNA library of HeLa cells (Stratagene). The
corresponding cDNA fragment was ampli®ed through a
polymerase chain reaction (PCR), using an RPS25-speci®c
sense primer with a ¯anking HincII site (RPC: 5'GACGTCGACAAGCTCAATAACTTAGTCTTG-3') and
an RPS25-speci®c anti-sense primer with a ¯anking XhoI
site (RPX: 5'-GAGCTCGAGTACAGCTGGTTGGACCTATTC-3'). The obtained amplicon was digested with
HincII and XhoI, subcloned into pBluescriptSK(7)
(Stratagene) and the resultant plasmid was designated,
pBSRPS-C. Between the upstream HindIII site and the
HincII site of pBSRPS-C, four synthetic oligonucleotides
were assembled, providing the amino-terminal 41 codons
of RPS25 (Li et al., 1991) to modify pBSRPS-C into
pBSRPS-F, that contains a full-length RPS25 cDNA.
Eukaryotic expression machinery was provided by
pcREVM1 (Malim et al., 1989a). At the unique BglII
site, which is immediately downstream of the initiation
codon, double-stranded synthetic oligonucleotides encoding the octapeptide [DYKDDDDK] entitled FLAG epitope
(IBI/Kodak) was inserted into pcREVM1. Utilizing the
internal HindIII site inscribed in the FLAG oligonucleotides, the HIV-1 Rev cDNA was replaced by the RPS25
cDNA through HindIII ± XhoI digestion and fragmentexchange with pBSRPS-F. The resultant plasmid,
pcFLRPS25, contains RPS25 cDNA with the FLAG
peptide at the amino terminus, which is driven by the
cytomegalovirus (CMV) promoter. The structure of pcFLRPS25 is illustrated in Figure 1a.
Myc-tagged RPS25 expression plasmid The full-length
cDNA of RPS25 was re-ampli®ed from pcFL-RPS25 by
PCR, in order to construct another plasmid for the
expression and characterization of RPS25 with a di€erent
tag-epitope. A sense primer (NRP: 5'-TAGGTACCAAGCTTAATGCCGCCTAAG-3'), which anneals from
the HindIII site in the FLAG region towards the RPS25
cDNA in pcFL-RPS25, and an anti-sense primer (RPE: 5'-
1511
Nuclear targeting of S25
S Kubota et al
1512
GGGGTACCGCATCTTCACCGGC-3') with an internal
KpnI site which is located immediately upstream of the
termination codon of RPS25 cDNA, were synthesized and
used for PCR ampli®cation. The amplicon was puri®ed,
digested by HindIII and KpnI and ligated between the
unique HindIII and KpnI sites of pCH110 (Pharmacia).
After digestion of this plasmid, entitled pCHRPS25, with
KpnI and MluI, a 4.7 kbp fragment was puri®ed and
ligated with a double-stranded synthetic oligonucleotide,
encoding a c-Myc epitope (Siomi and Dreyfuss, 1995) to be
pCHRPS25-Myc. As shown in Figure 1f, this construct
contains an RPS25 cDNA with a c-Myc epitope at the
carboxy terminus, which is driven by an SV40 promoter for
eukaryotic expression.
Deletion mutants Amino-terminal deletion mutants were
obtained, utilizing PCR. Three deletion mutants of RPS25
cDNA, each of which lacks 10, 20, or 43 amino-terminal
codons, respectively, were ampli®ed by corresponding
speci®c sense primers and RPX (see the ®rst subsection
of Materials and methods) from pcFL-RPS25. Plasmids
pcFL-RPS25 dN1, dN2, and dN3 were established by
substituting the RPS25 cDNA with these PCR-ampli®ed
cDNA mutants, utilizing the ¯anking restriction enzymatic
sites (Figure 2a).
Two carboxy-terminal deletion mutant expression vectors
were constructed by enzymatic digestion and re-ligation of
the basic plasmids, pBSRPS-F or pcFL-RPS25. RPS25
cDNA contains two internal SacI sites. Removal of a 40 bp
fragment between these SacI sites, by digestion and religation of pBSRPS-F, resulted in a frame shift at codon 91,
and an extra lysine and termination codons downstream. The
XhoI ± HindIII fragment of the pBSRPS-F variant, which
contained the deleted cDNA, was exchanged with the
corresponding fragment of pcFL-RPS25, and the resultant
plasmid was entitled pcFL-RPS25dC1. Similarly, using
internal three PvuII sites in RPS25 cDNA, pcFL-RPS25dC2
was constructed. Digestion and self-ligation of the major
fragment of pcFL-RPS25 resulted in the removal of 150 bp
and 30 bp internal fragments, which caused a frame-shift at
codon 72, which was followed by two extra and one nonsense
codons. Plasmid pcFL-RPS25dC3 was obtained via PCR. An
RPS25 cDNA fragment, which encodes the amino terminal
34 amino acid residues, was ampli®ed from pcFL-RPS25 by
corresponding speci®c primers with a HindIII site upstream
and an XhoI site downstream. The cDNA fragment was
exchanged with the full-length RPS25 cDNA in pcFL-RPS25
to obtain pcFL-RPS25dC3. In this construct, four extra
codons and a nonsense codon follows codon 34 of RPS25.
Structures of these carboxy-terminal deletion mutant proteins
are displayed in Figure 3a.
Internal site-directed mutants For the construction of
pcFL-RPS25m1, a point mutation was introduced into
pcFL-RPS25 by a PCR-mediated technique. NRP was
utilized for the sense primer here. As an anti-sense primer,
NRG: (5'-ACGCGTCAGGTACCGGCCCAACTTTGCC3') with a mutation at codon 41 and a HaeIII site, was
synthesized. PCR-ampli®cation of pcFL-RPS25 with these
primers yielded a mutated cDNA fragment encoding
residues 1 ± 41 of RPS25, with the 41st codon of a
glycine, instead of the arginine codon. The PCR product
was digested by HindIII and HaeIII, and ligated with
HindIII ± HincII digested pBSRPS-C to obtain a full-length
cDNA with the [41R-G] mutation. The mutant cDNA was
built in the same backbone as pcFL-RPS25, to become
pcFL-RPS25 m1.
In the process of verifying the nucleotide sequence of the
expected pcFL-RPS25dC1, one of the molecular clones was
shown to harbor unanticipated spontaneous minor deletions
in RPS25 cDNA. Five nucleotides [AAGAA] at codons 31 ±
34, and a single nucleotide [G] at codon 41, were missing.
These deletions caused an introduction of eight unrelated
amino acid residues between residues 32 ± 41 in the expressed
protein, instead of ten intact residues. This molecular clone
was designated as pcFL-RPS25dC1/m2. Since the plasmid
harbors a carboxy-terminal deletion, as well as the internal
mutation in RPS25 cDNA, a 0.6 kbp NcoI ± MscI fragment
of pcFL-RPS25dC1/m2, which included the mutated area,
was exchanged with the corresponding fragment of pcFLRPS25 to obtain pcFL-RPS25m2 lacking the carboxyterminal deletion.
Deletion of three internal codons for [31KKK] was
introduced by a ligation-mediated, multiple-step PCR
technique. A 57 base plus-strand oligonucleotide with a 3'
¯anking HaeIII site, which encodes residues 21 ± 41 except
31 ± 33 of RPS25, was synthesized. The oligonucleotide was
used to generate a double-stranded form, using a hemi-PCR
with a short primer (NRM: 5'-TTACGCGTCAGGTACCGGCCGAACTTTGCC-3') complementary to the 3'
end of the oligonucleotide, using Pfu polymerase (Stratagene)
to avoid extra additions of nucleotides at the 3' end. The
double-stranded oligonucleotide was ligated with the other
double-stranded oligonucleotides encoding residues 1 ± 20 of
RPS25, which had been used for the construction of intact
RPS25 cDNA. Fragments, that were ligated in order, were
then selectively ampli®ed by PCR with NRP and NRM
primers. The amplicon was digested with HindIII and HaeIII
and subcloned between the HindIII ± HincII sites of pBSRPSC. Transfer of the mutant cDNA to the eukaryotic
expression backbone was carried out, as described for the
wild-type. The obtained plasmid was designated as pcFLRPS25m3. Structures of the mutants described herein are
illustrated in Figure 4a.
All the RPS25 cDNA mutants were sequenced from both
directions to verify the proper introduction of mutations.
DNA sequencing was performed via an automated DNA
sequencing system (Applied Biosystems).
Structures of RPS25 fusion-protein expression plasmids
RPS25-insulin fusion construct Since the poly(A) addition
signal of pcFL-RPS25 originates in the rat preproinsulin
two gene, the plasmid contains the fragment of the 3' end of
the protein-encoding region of rat preproinsulin two gene,
as well as the poly(A) signal itself. By taking advantage of
this feature, a fusion cDNA was created. Ligation of the
assembled oligonucleotides with an upstream HindIII site
and a downstream HaeIII site, which encodes residues 1 ± 41
of RPS25, with a HindIII ± PvuII digested 3.5 kbp fragment
of pcFL-RPS25, yielded a fused open reading frame that
consists of the FLAG epitope, 41 amino-terminal residues
of RPS25, and 25 carboxy-terminal residues of rat
preproinsulin 2. Schematic representation of the fusion
protein is displayed in Figure 5a, as the translational
product of pcFL-NOQDINS.
As a control, a corresponding region of the cDNA of an
RPS25 mutant with abolished nuclear/nucleolar localization
was isolated and inserted in the same backbone as pcFLNOQDINS. An anti-sense primer for amplifying the mutated
cDNA fragment, corresponding to residues 1 ± 41 of RPS25,
from pcFL-RPS25m2, was designed and synthesized
(NRMM: 5'-GGGGCCAACTTTGCCTTTGGAC-3'). PCR
ampli®cation of pcFL-RPS25m2 with NRP and NRMM
yielded an RPS25m2 cDNA fragment corresponding to
residues 1 ± 41 of the wild-type (Figure 5a). The insulin
fusion construct was obtained through the same construction
procedure, as described above, in which the PCR product
was used, instead of the assembled oligonucleotides. In the
resultant fusion construct, pcFL-NOQm2DINS, the mutation
was copied from pcFL-RPS25m2, with an alternative codon
of tryptophan, instead of serine, at the forty-®rst codon of
the wild-type RPS25.
RPS25-b-galactosidase constructs The parental plasmid
for the expression of the bacterial lacZ gene was
Nuclear targeting of S25
S Kubota et al
commercially available (pCH110; Pharmacia). Two complementary oligonucleotides encoding residues 1 ± 21 of
RPS25 were synthesized and hybridized to each other to
form a double-stranded DNA fragment, with HindIII and
KpnI over-hangs. This fragment was ligated with a 6.9 kbp
HindIII ± KpnI fragment of pCH110 to establish pCHNOH. Similar procedures were applied to obtain pCHNOQ with a HindIII ± KpnI digested PCR amplicon, which
had been ampli®ed by NRP and NRM primers from pcFLRPS25. The expressed protein from pCH-NOQ contains
residues 1 ± 41 of RPS25 and b-galactosidase (Figure 6a).
Myc-tagged pyruvate kinase-RPS25 fusion construct The
construction of pcDNA3Myc-PK has been described
previously (Siomi and Dreyfuss, 1995). A sense primer
with a ¯anking KpnI site and an anti-sense primer with a
¯anking NotI site were designed to create a DNA
fragment, which encodes residues 25 ± 41 of RPS25. The
fragment was ampli®ed by PCR from pcFL-RPS25,
digested by KpnI and NotI and substituted with a 270 bp
KpnI ± NotI fragment of pcDNA3Myc-PK. The resultant
plasmid was designated, pcDNA3Myc-PK-NOR. An
alternative negative control, pcDNA3Myc-PK-C, was
constructed as follows. Original pcDNA3Myc-PK was
double-digested by KpnI and XbaI and the major fragment
was puri®ed. The isolated DNA fragment was treated with
T4 DNA polymerase for 15 min at 168C to trim and ®ll the
KpnI and XbaI overhangs, respectively, then self-ligated by
T4 ligase. This blunting ligation procedure removed the
tyrosine codon for residue 443 of PK, and a termination
codon that originated in the XbaI overhang was placed in
frame immediately downstream, which was con®rmed by
DNA sequencing. Primary structures of the expressed
proteins are illustrated in Figure 7a.
Cell culture and DNA transfection
Cos-1 and 293T cells were maintained in Dulbecco's
modi®ed minimum essential medium (D-MEM) supplemented with 10% fetal bovine serum (FBS) at 378C. A
liposome-mediated DNA transfection system (LipofectAMINE: GIBCO/BRL) was utilized for the immunofluorescence analyses. Twenty-four hours prior to transfection,
2.56104 Cos-1 or 293T cells were seeded into each well of
Lab-Tek eight-well chamber slides (Nunc, Inc.). DNA
transfection was carried out following a manufacturer's
optimized protocol with 200 ng of each plasmid DNA. For
immunoblotting, DEAE-dextran-mediated DNA transfection was performed, according to an established procedure
(Cullen, 1987). In this case, Cos-1 cells (26105 cells) were
prepared in 35 mm tissue culture plates, 24 h before
transfection. In each experiment, 500 ng of plasmid DNA
was used.
Antibodies
A murine monoclonal antibody for the detection of the
FLAG tag was purchased from IBI/Kodak (Anti FLAG
M2), and was used in immuno¯uorescence analyses at a
dilution of 1 : 100, or in immunoblotting at a dilution of
1 : 1000, respectively. A murine monoclonal anti-b-galactosidase IgG was obtained from Boehringer-Mannheim
Biochemicals, and used at a dilution of 1 : 10 000 for
immuno¯uorescence studies. The anti-Myc 9E10 murine
monoclonal antibody was kindly provided by Dr H Siomi
(University of Pennsylvania). For immuno¯uorescence,
9E10 was applied to the ®xed cells at a dilution of
1 : 1000. Fluorescein-isothiocyanate (FITC)-conjugated
anti-murine IgG serum, FITC-conjugated anti-rabbit IgG
serum, and horseradish peroxidase (HRP)-conjugated antimurine IgG serum were purchased (Sigma), and used as
detector antibodies for immuno¯uorescence (1 : 100) and
immunoblotting (1 : 1000), respectively.
Immunoblotting
Forty-eight hours after transfection, cells were washed
once and harvested in 500 ml of a RIPA bu€er (Adachi et
al., 1992). Proteins in 360 ml of the clari®ed lysate were
precipitated in 80% acetone, dissolved in 40 ml of
16sodium dodecyl sulfate (SDS) sample bu€er (NOVEX)
with 2.5% 2-mercapto-ethanol and separated using SDSpolyacrylamide gel electrophoresis (PAGE) through a 14%
pre-cast gel (NOVEX). Separated proteins were transferred
onto a PVDF membrane (Polyscreen : Dupont NEN),
blocked in 5% nonfat dry milk, and probed with the antiFLAG antibody for 1 h in 1% BSA/0.05% Tween 20/PBS
at room temperature. After extensive washes, protein
visualization was carried out following a manufacturer's
protocol, using a commercial kit (Renaissance: Dupont
NEN) with the HRP-conjugated antiserum.
Indirect immuno¯uorescence
Forty-eight hours after transfection, cells were washed
once with PBS, ®xed in 3.5% formaldehyde/PBS for
20 min and permeabilized in 0.1% NP-40/PBS for 10 min
at room temperature. Incubation in primary or secondary
antibodies in 3% BSA/PBS was performed for 1 h at 378C,
followed by extensive washes with PBS, as described
previously (Kubota et al., 1991). After the last wash, cells
were placed in a mounting medium (Omni¯uor: Virostat),
and viewed with an epi¯uorescence microscope (Olympus).
Generally, transfection eciency was approximately 15%,
as analysed by immuno¯uorescence microscopy. All the
photomicrographs were taken under a magni®cation
of6400, with the exception of Figure 1, in which a few of
these were taken at6200 or61000. Each photomicrograph
represents a vast majority of the cells with similar subcellular
localization, unless otherwise stated on the text.
Acknowledgements
The authors wish to thank Dr Haruhiko Siomi for the
murine monoclonal antibodies and pcDNA3Myc-PK, Dr
Bryan R Cullen for the anti-HIV-1-Rev serum, Dr Michael
H Malim for pcRev and pcREVM1, Ms Eva Majerova for
peptide synthesis, Ms Lisa Bobroski for digital photography and Ms Rita M Victor and Ms Brenda O Gordon
for excellent secretarial assistance. This work was supported in part by US PHS grant AI36552 to RJP, and the
National Cancer Institute, Department of Health and
Human Services under contract with ABL.
References
Adachi Y, Copeland TD, Takahashi C, Nosaka T, Ahmed A,
Oroszlan S and Hatanaka M. (1992). J. Biol. Chem., 263,
21977 ± 21981.
Agrawal MG and Bowman LH. (1987). J. Biol. Chem., 202,
4868 ± 4875.
Akey CW and Goldfarb DS. (1989). J. Cell Biol., 109, 971 ±
982.
Aloni R, Peleg D and Meyuhas O. (1992). Mol. Cell. Biol.,
12, 2203 ± 2212.
Cullen BR. (1987). Methods Enzymol., 152, 684 ± 704.
Dang C and Lee WMF. (1989). J. Biol. Chem., 264, 18019 ±
18023.
Eng FJ and Warner J. (1991). Cell, 65, 797 ± 804.
1513
Nuclear targeting of S25
S Kubota et al
1514
Fischer U, Meyer S, Teufel M, Heckel C, LuÈhrman R and
Rautmann G. (1994). EMBO J., 13, 4105 ± 4112.
Goldfarb DS. (1997). Curr. Biol., 7, 1213 ± 1216.
GoÈrlich D and Mattaj IW. (1996). Science, 271, 1513 ± 1518.
Gritz L, Abovich N, Teem JL and Rosbash M. (1985). Mol.
Cell. Biol., 5, 3436 ± 3442.
Heaphy S, Dingwall C, Ernberg I, Gait MJ, Green SM, Karn
J, Lowe AD, Singh M and Skinner MA. (1990). Cell, 60,
685 ± 693.
Imai T, Sudo K and Miwa T. (1994). Genomics, 20, 142 ± 143.
Kalderon D, Roberts BL, Richardson WD and Smith AE.
(1984). Cell, 39, 499 ± 509.
Kalland K-H, Szilvay AM, Brokstad KA, Sñtrevik W and
Haukenes G. (1994). Mol. Cell. Biol., 14, 7436 ± 7444.
Kubota S, Duan L-X, Furuta RA, Hatanaka M and
Pomerantz RJ. (1996). J. Virol., 70, 1282 ± 1287.
Kubota S, Nosaka T, Cullen BR, Maki M and Hatanaka M.
(1991). J. Virol., 65, 2452 ± 2456.
Kubota S, Siomi H, Satoh T, Endo S, Maki M and Hatanaka
M. (1989). Biochem. Biophys. Res. Comm., 162, 963 ± 970.
Li H, Dalal S, Kohler J, Vilardell J and White SA. (1995). J.
Mol. Biol., 250, 447 ± 459.
Li M, Latoud C and Center MS. (1991). Gene, 107, 329 ± 333.
Mager WH. (1988). Biochim. Biophys. Acta, 949, 1 ± 15.
Malim MH, BoÈhnlein S, Hauber J and Cullen BR. (1989a).
Cell, 58, 205 ± 214.
Malim MH and Cullen BR. (1991). Cell, 65, 241 ± 248.
Malim MH, Hauber J, Le S-H, Maizel JV and Cullen BR.
(1989b). Nature, 338, 254 ± 257.
Martin-Nieto J and Roufa DJ. (1997). J. Cell Sci., 110, 955 ±
965.
Melchior F and Gerace L. (1995). Curr. Opin. Cell Biol., 7,
310 ± 318.
Meyer BE and Malim MH. (1994). Genes & Develop., 8,
1538 ± 1547.
Michael WM and Dreyfuss G. (1996). J. Biol. Chem., 271,
11571 ± 11574.
Moreland RB, Nam HG, Hereford LM and Fried HM.
(1985). Proc. Natl. Acad. Sci. USA, 82, 6561 ± 6565.
Nosaka T, Takamatsu T, Miyazaki Y, Sano K, Sato A,
Kubota S, Sakurai M, Ariumi Y, Nakai M, Fujita S and
Hatanaka M. (1993). Exp. Cell Res., 209, 89 ± 102.
Peculis BA and Gall JG. (1992). J. Cell. Biol., 116, 1 ± 14.
Richard N, Iacampo S and Cochrane A. (1994). Virology,
204, 123 ± 131.
Rout MP, Blobel G and Aitchison JD. (1997). Cell, 89, 715 ±
725.
Russo G, Ricciardelli G and Concetta P. (1997). J. Biol.
Chem., 272, 5229 ± 5235.
Schmidt C, Lipsius E and Kruppa J. (1995). Mol. Biol. Cell.,
6, 1875 ± 1885.
Schmidt-Zachmann and Nigg EA. (1993). J. Cell Sci., 105,
799 ± 806.
Schoborg RV and Clements JE. (1994). Virology, 202, 485 ±
490.
Seiki M, Inoue J-I, Hidaka M and Yoshida M. (1988). Proc.
Natl. Acad. Sci. USA, 85, 7124 ± 7128.
Siomi H and Dreyfuss G. (1995). J. Cell Biol., 129, 551 ± 560.
Siomi H, Shida H, Nam SH, Nosaka T, Maki M and
Hatanaka M. (1988). Cell, 55, 197 ± 209.
Siomi S, Shida H, Maki M and Hatanaka M. (1990). J.
Virol., 64, 1803 ± 1807.
Sodroski J, Rosen C, Wong-Staal F, Salahuddin SZ, Popovic
M, Arya S, Gallo RC and Haseltine WA. (1985). Science,
227, 171 ± 173.
Sollner-Webb B and Mougey BB. (1991). Trends Biochem.
Sci., 16, 58 ± 62.
Sommerville J. (1986). Trends Biochem. Sci., 11, 438 ± 442.
Tan R, Chen L, Buettner JA, Hudson D and Frankel AD.
(1993). Cell, 73, 1031 ± 1040.
Tang CK and Drapier DE. (1990). Biochem., 29, 4434 ± 4439.
Tasheva ES and Roufa DJ. (1995). Genes & Develop., 9,
304 ± 316.
Tiley L, Malim M and Cullen BR. (1991). J. Virol., 65,
3877 ± 3881.
Warner JR. (1990). Curr. Opin. Cell Biol., 2, 521 ± 527.
Yan C and MeÂleÁse T. (1993). J. Cell Biol., 123, 1081 ± 1091.
Zapp ML and Green MR. (1989). Nature, 342, 714 ± 716.