• No results found

T he nucleus was once thought of as a non-structured amorphous

N/A
N/A
Protected

Academic year: 2021

Share "T he nucleus was once thought of as a non-structured amorphous"

Copied!
8
0
0

Loading.... (view fulltext now)

Full text

(1)

Visualization of gene activity in

living cells

Toshiro Tsukamoto*†, Noriyo Hashiguchi*†, Susan M. Janicki*, Tudorita Tumbar‡, Andrew S. Belmont‡ and David L. Spector*§ *Cold Spring Harbor Laboratory, One Bungtown Road, Cold Spring Harbor, New York 11724, USA † Department of Life Science, Himeji Institute of Technology, 3-2-1 Koto, Kamigori, Hyogo 678-1297, Japan ‡ Department of Cell and Structural Biology, University of Illinois, Urbana, Illinois 61801, USA § e-mail: spector@cshl.org

Chromatin structure is thought to play a critical role in gene expression. Using the lac operator/repressor system and two colour variants of green fluorescent protein (GFP), we developed a system to visualize a gene and its pro-tein product directly in living cells, allowing us to examine the spatial organization and timing of gene expressionin vivo. Dynamic morphological changes in chromatin structure, from a condensed to an open structure, were

observed upon gene activation, and targeting of the gene product, cyan fluorescent protein (CFP) reporter to peroxi-somes was visualized directly in living cells. We found that the integrated gene locus was surrounded by a promyelo-cytic leukaemia (PML) nuclear body. The association was transcription independent but was dependent upon the direct in vivo binding of specific proteins (EYFP/lac repressor, tetracycline receptor/VP16 transactivator) to the locus. The ability to visualize gene expression directly in living cells provides a powerful system with which to study the dynamics of nuclear events such as transcription, RNA processing and DNA repair.

T

he nucleus was once thought of as a non-structured amor-phous environment in which interactions occur in a rather random manner, but a high degree of functional organization has since been found within the nucleus (for reviews see refs 1,2). The ability to tag specific proteins or nucleic-acid sequences directly in living cells has greatly extended the ability of light microscopy to address structure–function questions relating to many fundamental

cellular processes. Most notable has been the revolutionary use of green fluorescent protein and its spectral variants to assess the dynamic aspects of cellular function (for reviews see refs 3–5).

Knowledge of the organization of chromatin in the interphase nucleus, above the level of the folded 30-nm fibre, has substantial-ly lagged behind our molecular and biochemical understanding of chromatin structure and function. This has been due in part to

pSV2-EYFP-lac repressor pTet-On

Transcription Translation Peroxisome

EYFP VP16 lac repressor Dox r tetR p3216PCβ Visualize gene p3216PCβ pTK-Hyg pTet-On pSV2-EYFP-lac repressor Inducible promoter by doxycycline Monitor transcription Peroxisome targeting signal (SKL)

lac op TRE Pmin CFP SKL β-globin Intron

x8 x6 x32 x16 a b Hygromycin B selection Doxycycline clone 2, 22, 102 BHK

Figure 1 Experimental design. a, Schematic representation of plasmid p3216PCβ. An approximately 10-kb lac-operator repeat is present at the 5‘ end of the plasmid, and is visualized in cells with EYFP/lac repressor expressed from the pSV2-EYFP/lac repressor plasmid. Transcription of CFP–SKL is activated by binding

selected with hygromycin B. Isolated clones were subjected to a second transfec-tion with pTet-On and pSV2-EYFP/lac repressor plasmids followed by inductransfec-tion with doxycycline. The integrated plasmid was visualized as a YFP signal in the nucleus, and the transcriptional state of the integrated locus was monitored by the

(2)

appear-limitations in preserving chromatin structure after fixation and antibody labelling. Belmont and colleagues have taken advantage of the thermodynamic properties of the lac operator/repressor inter-action to examine chromatin organization in vivo (for a review see ref. 6). Using lac operator–repressor localization combined with gene amplification, it was possible to visualize large-scale chromatin fibres directly in living cells7. Immunoelectron

microscopy showed that these fibres corresponded to previously described chromonema fibres of ~100 nm. Using this system, a 90-million-base-pair region of late-replicating DNA comprising a het-erochromatic homogeneously staining region was examined; this region was found to decondense just before DNA replication, and 4–6 hours into S phase it moves from a peripheral nuclear region to the nuclear interior8.

In a subsequent study, a striking large-scale decondensation of chromatin into extended chromonema fibres was observed9 by

specifically targeting the acidic activation domain of the herpes simplex virus transcriptional activator VP16 to two heterochro-matic amplified chromosome regions. These data led the authors to suggest a functional link between recruitment of the transcription-al machinery and changes in large-sctranscription-ale chromatin structure. Several other studies have made use of this system to examine the separation of sister chromatids, and chromosome dynamics in liv-ing yeast cells10,11.

Here we have extended these studies by examining chromatin organization and its relationship to transcriptional activity in the context of the synthesis of an inducible pre-mRNA in a mam-malian cell. We have managed to visualize gene expression directly Table 1 Correlation between chromatin organization and CFP/SKL gene expression

Time (h) DNA Peroxisomes 0 1 2 4 6 8 22 8(–) 22(–) Closed – 100 98 96 82 61 54 52 94 97 Open – 0 2 1 4 8 18 5 6 2 Closed + 0 0 2 2 2 0 5 0 0 Open + 0 0 1 12 29 28 38 0 1 Clone-2 cells were transfected with pTet-On and plated onto coverslips. Doxycycline was added 2 h after transfection. Cells were fixed at the times as indicated, and the gene locus was visualized with recombinant EGFP/lac repressor protein. We counted 100 nuclear dots. Homogeneous and round signals were scored as closed; heterogeneous signals were counted as open. According to our criteria, signals in Fig. 5a–c are closed and d–l are open (DNA was visualized by in vivo expression of EYFP/lac repressor). At early time points, the difference between open and closed cells was not significant.

Copy number

a b

c

Clone

100101102 103 2 22

BHK Clone 2 Clone 22 Clone 102

– + – + – + – + Dox Dox – + + + + + + – – 0 1 2 4 6 8 22 8 22 (h) 102 1 2 3 4 5 6 7 8 1 2 3 4 5 6 7 8 1 2 3 4 5 6 7 8 9 Size (kb) Mr (K) Mr (K) 23.1 9.4 6.6 4.4 2.3 2.0 0.56 194 120 87 64 52 39 26 21 194 120 87 64 52 39 26 21

Figure 2 Characterization of isolated clones. a, Southern blotting of isolated clones. Genomic DNA was isolated from parental BHK cells, and three isolated clones (2, 22 and 102) were examined by Southern blotting to derive estimates of plasmid copy number in each stable cell line. Lanes 1–4, BHK genomic DNA con-taining 6.2 pg, 62 pg, 620 pg or 6.2 ng of p3216PCβcorresponding to 1, 10, 100 and 1,000 copies per haploid genome; lane 5, Biotin-labelled λ-HindIII marker; lanes 6–8, genomic DNA of clones 2, 22 and 102, respectively. Lower panel, shorter exposure time. b, Induction of CFP–SKL protein by doxycycline. Cells were transfected with pTet-On and examined for induction of the CFP–SKL fusion protein

by immunoblotting with an anti-GFP antibody. Lanes 1 and 2, parental BHK; lanes 3 and 4, clone 2; lanes 5 and 6, clone 22; lanes 7 and 8, clone 102. Lanes 1, 3, 5 and 7, doxycycline (–); lanes 2, 4, 6 and 8, doxycycline (+). c, Time course of induction of clone-2 cells. Cells were transfected with pTet-On. Two independent transfections were pooled and plated into ten dishes. Doxycycline was added 2 h after transfection, as indicated. Cells were examined at 0 (lane 1), 1 (lane 2), 2 (lane 3), 4 (lane 4), 6 (lane 5), 8 (lane 6) and 22 h (lane 7), after addition of doxycy-cline; doxycycline-free samples were also prepared at 8 (lane 8) and 22 h (lane 9).

(3)

in living cells. Furthermore, we have demonstrated that the stably integrated gene locus associates with a PML nuclear body.

Results

A system for visualizing gene expression. To examine the dynam-ics of gene expression in living cells, we developed several baby hamster kidney (BHK) cell lines containing a stably integrated plas-mid composed of 256 copies of the lac operator sequence. Downstream of these repeats we inserted 96 copies of the tetracy-cline-responsive element controlling a minimal cytomegalovirus (CMV) promoter regulating the expression of a cyan fluorescent protein with a peroxisome targeting signal-1 (Ser-Lys-Leu (SKL) tripeptide) at its carboxy terminus12(Fig. 1a). The entire plasmid

(p3216PCβ) sequence encompassed 18.52 kilobases (kb) (Fig. 1a). The system was designed such that when the stable cells were tran-siently transfected with an EYFP/lac repressor plasmid, the expressed fusion protein would be able to localize to the stably inte-grated locus by association of the EYFP/lac repressor protein with the lac operator sequences. Alternatively, the integrated locus could be localized after fixation using a bacterially expressed EGFP/lac repressor fusion protein. The repressor binds to the operator with a Kdof 10–13 M (ref. 13). Co-transfection with the pTet-On plasmid

followed by addition of doxycycline to the culture medium acti-vates the tetracycline-responsive element, stimulating transcription of the CFP–SKL fusion protein. Localization of the fusion protein to peroxisomes confirms that transcription has occurred (Fig. 1b). Southern blot analysis was used to determine the relative copy number of the plasmid sequences in several clonal cell lines. PstI,

β

No transfection pTet-On, Dox(–) pTet-On, Dox(+)

BHK Repressor stain Clone 2 Repressor stain Clone 22 Repressor stain Clone 102 Repressor stain Clone 2 In vivo expression a b c d e f g h i j k l m n o

Figure 3 Visualization of the genetic locus. Parental BHK cells and each clone were examined for the localization of the genetic locus. Only cells expressing pTet-On and exposed to doxycycline exhibit a CFP–SKL signal (f, i, l, o). Overlay staining with recombinant EGFP/lac repressor (a–l) or in vivo expression of EYFP/lac

repres-sor signal (m–o, fixed with 4% formaldehyde) is represented as a yellow colour. The CFP–SKL signal is shown in cyan (a–o). The enhancement of each signal was com-parable among three panels of the same cell. The CFP signal in a–c was processed as in j–l. Scale bar represents 10µm.

Unfixed, Dox(–) Unfixed, Dox(+) 4%PFA

Saponin/4%PFA Triton/1.6%PFA Methanol

Saponin/4%PFA

repressor stain Triton/1.6%PFrepressor stain Methanol

a b c

d e f

g h i

Figure 4 Comparison of different methods of fixation and staining. The EYFP/lac repressor was expressed in a–f and doxycycline was added to all except a. The YFP signal was observed without fixation (a and b), after fixation with 4% formaldehyde (PFA) for 1 h (c), with saponin pretreatment before fixation (d and g), Triton X-100 pretreatment before fixation with 1.6% formaldehyde (e and h), or with ice-cold methanol for 5 min (f and i). EGFP/lac repressor overlay staining was done after different fixation methods (g–i) of cells transfected with pTet-On. Peroxisomal

(4)

the three clonal cell lines examined (Fig. 2a). For simplicity, we will refer to the integration site as a genetic locus. Integrated copy num-bers of the three clones were estimated as 1,000 (clone 2), 50 (clone 102) and 10 (clone 22) copies per haploid genome (Fig. 2a). We then examined the response of the integrated locus to doxycycline. All three clones that transiently expressed pTet-On showed a sig-nificant stimulation of transcription 24 hours after treatment of cells with doxycycline, as determined by immunoblot analysis using an anti-GFP antibody that recognized the CFP–SKL fusion protein (Fig. 2b). Minimal leakiness of the promoter was observed in all three clones. The parental BHK cell line showed no response to doxycycline treatment (Fig. 2b, lanes 1, 2).

To investigate the temporal response of the cells to doxycycline more precisely, cells from clone 2 were transfected with pTet-On and doxycycline was added 2 hours after transfection. Cells were collected at the indicated time points and examined by immunoblot analysis for the presence of the CFP–SKL fusion pro-tein. This was first detected 6 h after the addition of doxycycline (Fig. 2c, lane 5), and increased over time (Fig. 2c, lanes 6, 7). The number of peroxisome(+) cells also increased over time. The increase in the number of cells with an open chromatin structure and peroxisome labelling (open/peroxisome(+); Table 1) at 22 h

compared with the earlier time points may be the result of cells that were open/peroxisome(–) at 6–8 h not having accumulated detectable levels of CFP–SKL in their peroxisomes (Table 1). No protein product was detected at 8 or 22 h post-transfection in the absence of doxycycline (Fig. 2c, lanes 8, 9).

Chromatin organization and transcription. The stably integrated genetic locus was examined in all three clonal cell lines in the pres-ence or abspres-ence of transcription. Cells were either not transfected or transfected with pTet-On, and were then allowed to grow for 8 h in the absence or presence of doxycycline. Cells were subsequently fixed and the lac operator sequences localized by incubation with bacterially expressed EGFP/lac repressor. In all cases, the parental BHK cell line showed no nuclear EGFP/lac repressor signal and no CFP–SKL protein product in the cytoplasm (Fig. 3a–c). Cells from clones 2, 22 and 102 each showed a single site of integration of the genetic locus in the absence (Fig. 3d, g, j) or presence (Fig. 3e, h, k) of the pTet-On plasmid. Activation of the gene with doxycycline for 8 h resulted in the chromatin decondensing (Fig. 3f, i, l). This change in chromatin morphology was obvious in cells from clone 2 but was not as easily observed in cells from clones 22 or 102, which contain fewer integrated copies of the plasmid. In addition, the degree of decondensation varied among clone-2 cells. The presence of the

a b c d e f g h i j k l m n o p q r s 0 h 0.25 h 0.5 h 0.75 h 1 h 1.5 h 2 h 2.5 h 3 h 4 h 5 h 6 h Dox(–) 0 Dox(–) 1

Dox(+) 2 Dox(+) 8 Dox(–) 8

Dox(–) 3 Dox(–) 6

Figure 5 Changes in chromatin organization during gene activation. Chromatin organization was monitored in the same living cell for 6 h in the presence (a–l) or absence (m–p) of doxycycline. Note the nuclear rotation over this time period. The EYFP/lac repressor signal is strongly enhanced to show the position in the nucleus (yellow) in the main panels. Appropriately enhanced images are shown in the insets. CFP–SKL data were collected only in a, g, i, j, l, m and o–s. The enhancement con-ditions for CFP–SKL in a, g, i, m, o and p are the same; j and l are less enhanced.

Using fixed cells, CFP–SKL RNA in the nucleus was detectable 2 h (q) or 8 h (r) after doxycyline induction, but no signal was observed in the absence of doxycycline (s). The enhancement of the RNA FISH signal and the CFP–SKL signal are the same in q, r and s. Images in the cyan channel were taken only at the time points indicated in a, g, i, j, l, m and o–s. In q–s the RNA FISH signal is indicated in red, the EYFP/lac repressor signal in green and the CFP-SKL signal in cyan. Scale bar represents 10µm; insets, ×3 magnification.

(5)

CFP–SKL fusion protein in peroxisomes confirmed not only that doxycycline-dependent transcription had occurred in these cells, but also that the messenger RNA translation product correctly localized to its appropriate intracellular destination. Next, we visu-alized gene expression directly in clone-2 cells that transiently expressed the pSV2-EYFP/lac repressor under the pTet-On gene-expression system. As was observed above, activation of transcrip-tion resulted in chromatin decondensatranscrip-tion and the appearance of the CFP–SKL fusion protein in peroxisomes (Fig. 3m–o).

We then examined the effect of fixation conditions on the reor-ganization of chromatin structure after transcriptional activation. All fixation conditions used resulted in an extended chromatin structure similar to that seen in living cells (Fig. 4). However, formaldehyde fixation with or without prior permeabilization with detergent (Fig. 4c–e, g–h) provided images that most closely resem-bled those observed in living cells (Fig. 4b). Cells fixed in methanol (Fig. 4f, i) resulted in a more tightly packed chromatin structure, perhaps because of the dehydrating properties of this fixative. Subsequent experiments with fixed cells therefore used 4% formaldehyde in PBS. That the observed reorganization of chro-matin was not simply caused by binding of the EYFP/lac repressor to the locus in living cells is indicated by the finding of a similar morphology when fixed cells were stained with EGFP/lac repressor (Fig. 4g–i).

Spatial organization and timing of gene expression. The ability to visualize a gene and its protein product directly in living cells allowed us to examine the spatial organization and timing of gene expression in vivo. Clone-2 cells were co-transfected with pTet-On and pSV2-EYFP/lac repressor and allowed to attach to cover-slips fitted for an FCS2 live-cell chamber. Then, 1.5 h post-trans-fection, the coverslip was mounted into the chamber and an image of the inactive gene locus was acquired 30 min later (Fig. 5a, 0 h). Doxycycline was then perfused into the chamber to acti-vate transcription of the CFP–SKL fusion protein. Images were acquired for the gene and overlaid onto brightfield images of the cell (Fig. 5b–l) at the indicated times after the addition of doxy-cycline. At various time points, an additional image was acquired, showing the localization of the CFP–SKL fusion protein. Images of CFP–SKL were taken at limited time points to reduce the pos-sibility of phototoxicity.

Changes in chromatin organization were first detected about 30 min after the addition of doxycycline (Fig. 5c, see inset), and fully extended chromatin organization was observed about 4 h

doxycycline (Fig. 5i). The lag period from the first observation of a change in chromatin structure (+0.5 h) to the time the fusion protein was first detected (+3 h) is probably related to the time required for detectable levels of the fusion protein to accumulate in the peroxisomes. RNA was easily detected at the site of tran-scription in many cells by in situ hybridization 2 h after the addi-tion of doxycycline (Fig. 5q). For a more precise examinaaddi-tion of gene expression in living cells, images were acquired every 10 or 20 minutes over a 7-h period in both the YFP and CFP channels, and the data were converted into a movie (see Supplementary Information).

Over the time period that cells were observed, in many cases, the locus appeared to reposition itself upon transcriptional activa-tion (Fig. 5a–d). Cellular movements and nuclear rotaactiva-tion were also seen during the observation of living cells (compare the nuclear position in Fig. 5a, j, k). Cells that were not treated with doxycycline showed no change in chromatin organization (Fig. 5m–p), no RNA hybridization signal (Fig. 5s), or any CFP–SKL fusion protein signal (Fig. 5m–p, s), demonstrating the specificity and tight control of the gene locus.

To examine potential chromatin movements during transcrip-tion more closely, 8 h after the additranscrip-tion of doxycycline images were acquired every 15 s. Selected images from this time course are shown in Fig. 6. No change in chromatin organization was observed, and minimal movement of the locus was detected (com-pare Fig. 6a, b) over a 15-min time period. These data demonstrate that significant movement of chromatin is not necessary for the maintenance of gene activity.

Association with a PML body. Next, we examined the relationship of the stably integrated gene locus to the localization of other nuclear compartments. We investigated the relationship of the gene locus to nucleoli, Cajal (coiled) bodies, and PML bodies. No significant association was observed between the gene locus and nucleoli or Cajal bodies. However, we found an association between this locus and PML bodies in a significant number of cells of all three clonal cell lines upon expression of the EYFP/lac repressor and pTet-On (PML bodies have also been called ND10, PODs (for PML oncogenic domains), and Kremer bodies (for a review see ref. 14)). PML bodies vary in size from 0.3 to 1µm in diameter, and a typical mammalian nucleus contains 10–20 of these structures. PML bodies have been suggested to play a role in aspects of transcriptional regulation, and appear to be targets of viral infection (for reviews see refs 14,15).

To study the relationship between the integrated gene locus and PML bodies, we used clone-22 cells because the integrated copy number was small (10 copies, ~185 kb), so the signal appeared as a small dot, making the evaluation of co-localization more precise. Clone-22 cells transiently transfected with pTet-On and the pSV2-EYFP/lac repressor showed a significant degree of association with PML bodies, as detected by immunocytochem-istry, in the presence or absence of doxycycline, indicating that this association was not dependent upon ongoing transcription of the locus (Fig. 7a, b, Table 2). In cells transfected with only the pSV2-EYFP/lac repressor (Fig. 7c) or pTet-On plasmids (Fig. 7d), an association between the gene locus and PML bodies was still observed.

To address whether this association was directly due to the integration site of the gene locus or to the proteins associated with the locus, clone-22 cells were fixed and stained with recombinant EGFP/lac repressor protein. An association between the gene locus and PML bodies was not observed if pSV2-EYFP/lac repressor and pTet-On were not expressed in living cells (Fig. 7e). This finding demonstrates that the association was related to a local high con-centration of the expressed protein (EYFP/lac repressor or tetracy-cline receptor/VP16 transactivator) at the gene locus, rather than the locus itself. These data suggest that PML bodies may function

8h 0 min +15 min

a b

c d e f g

h i j k l

Figure 6 The open chromatin structure is static. At 8 h after addition of doxycy-cline, an actively transcribing locus was examined every 15 s for changes in chro-matin organization. This cell expressed CFP–SKL. The nucleus moved about 10 µm during observation because of cell movement, but the shape of the open chromatin region did not change. Scale bar represents 10µm; boxes in a and b indicate the regions of interest shown in c–l.

(6)

Discussion

We have developed a cell system that allows the observation of changes in chromatin organization related to gene expression, and provides the ability to visualize gene readout directly in the form of a CFP fusion protein with a peroxisome targeting signal. Using this system we have visualized directly gene expression in living cells.

The mechanisms by which genes are activated have been inves-tigated extensively using a variety of biochemical and genetic approaches16. These studies have revealed the involvement of

ATP-driven chromatin remodelling complexes, as well as the chem-ical modification of specific lysine residues on the amino-terminal histone tails by acetyltransferases (for review see refs 17–19). However, the remodelling and modification of chromatin that allow it to become transcriptionally competent must also be considered in light of the constraints placed by chromatin packaging on the acces-sibility of chromatin to activator proteins in the interphase nucleus. Although it has been difficult to accurately determine the packaging ratio of the same gene in both the inactive and active states20,

a b

c d

e f

Figure 7 Relationship between the integrated locus and PML bodies. PML bodies were immunolabelled with monoclonal antibody 5E10 and are shown as a red signal. The p3216PCβintegration site was visualized in clone-22 cells by in vivo expressed EYFP/lac repressor (a–c), by EGFP/lac repressor overlay staining (d, e)

or by RNA FISH (f) after the addition of doxycycline and shown as a green signal. CFP–SKL expression was shown as a cyan signal. Scale bar represents 10µm; insets, ×3 magnification.

Table 2 Association of a PML body with the integrated genetic locus

pTet-On pSV2-EYFP/lac repressor Dox PML(+)/Per(+) PML(+)/Per(–)

– + – 0/0 19/25 *

+ + – 0/0 17/25 *

+ + + 8/8 13/17 *

+ – + 7/7 2/18 **

– – – 0/0 0/25 **

PML body localization in relation to the integrated genetic locus was examined in 25 clone-22 cells. PML(+), PML body was colocalized with or adjacent to DNA signal; PML(+)/Per(–), PML body was co-localized with or adjacent to DNA signal in a peroxisome-negative cell. * Cells with EYFP/lac repressor signal were counted; only transfected cells were counted. ** Cells were stained with the EGFP/lac repressor protein. All cells with DNA signal were counted, whether transfected or not.

(7)

Robinett et al.7used the lac operator/repressor system to examine

chromosome regions containing several copies of a dihydrofolate reductase gene (a 90-million-base-pair amplified chromosome region) to visualize chromatin organization directly at the level of the 100–130-nm fibre in interphase cells. Remarkably, the observed chromatin region was shown to undergo precisely timed contrac-tions and expansions during the G1 and the late S/G2 phases8.

Tumbar et al.9 have since used this approach to observe the

unfolding and remodelling of a large heterochromatic chromo-some amplified region containing repetitive DNA sequences that represented the target of the herpes simplex virus acidic activation domain VP16. VP16 targeting resulted in a decondensation of the heterochromatic homogeneously staining region and to induce a strong increase in histone acetylation at the targeted chromosome site. In addition, hyperacetylation at H2A (Lys 5) and H4 was also observed at this site. However, this locus did not directly encode a protein that could be visualized in living cells, so only the result of activator binding was observed rather than the transcription of a specific locus. Here we have extended these studies by developing a system in which direct gene readout can be visualized in living cells. An important question with regard to the positioning of chro-matin within the interphase nucleus is whether a particular locus occupies a fixed interphase position. Using specific probes against centromere-associated α-satellite DNA sequences, chromosomal subdomains were found to exhibit changes in their intranuclear localization during the cell cycle21–24. For example, in G1 cells,

chromosome-8 centromeres localize adjacent to the nuclear periphery, whereas in G2 cells they localize to more internal nuclear regions23. In a study using the lac operator/repressor

sys-tem, a 90-million-base-pair region of late-replicating DNA was found to move from a peripheral nuclear region into the nuclear interior 4–6 h into S phase8. Dynamic aspects of chromatin have

also been studied by fluorescence recovery after photobleaching25,

and it was concluded that interphase chromatin is immobile over distances of 0.4µm or more25.

A second approach involved tagging specific chromosome sites in living cells of the yeast Saccharomyces cerevisiae with GFP and in

Drosophila with fluorescently labelled topoisomerase II26. It was

found that chromatin undergoes significant diffusive brownian motion within a restricted area with a radius of less than 0.3µm in yeast and 0.9µm in Drosophila. The observed differences between these reports may be due to differences in the sensitivities of the respective techniques or the cell systems used. However, these stud-ies indicate that chromatin does not appear to undergo any long-range movements over short time periods within the interphase nucleus. The present study, in which we examined a stably inte-grated and transcriptionally active genetic locus, indicates that in some cases there is chromatin movement when genes are activated within the interphase nucleus of mammalian cells. Consistent with this possibility, changes in centromere and chromosome distribu-tion have been observed over longer time periods or after alter-ations in physiological conditions, for example in response to cell differentiation27, transcription signals28, cell-cycle stage23,29–31, or

pathological state32.

We have found an association between the integrated genetic locus and PML bodies. This association is independent of the tran-scriptional activity of the locus and seems to be mediated through the transiently expressed proteins that associate with the integrated locus. These data lead us to propose that the PML body may act as a ‘sensor’ with the ability to detect and mark local accumulations of proteins or nucleic acids that are foreign or ‘suspect’ to the cell. In previous studies, interferon has been shown to cause an increase in the number and size of PML bodies by upregulating the PML-body-associated proteins Sp100 and PML33–36through activation of

an interferon response element. Based on this finding, it was sug-gested that these proteins, and the PML body itself, are part of an

37

and herpes simplex virus type 1) were shown to localize to PML bodies when cells were infected. In addition, several transiently expressed proteins (such as ISG20 and BRCA1) have also been shown to localize adjacent to PML bodies after an excess of the pro-tein is expressed38,39.

In summary, we have developed a cell system in which gene expression can be visualized directly in living cells and the dynam-ics of various nuclear proteins and structures can be studied in rela-tion to the site of gene expression. Using this system, we have found that gene expression induces dynamic changes in chromatin struc-ture and that the PML body may act as a nuclear sensor that can detect local concentrations of exogeneously introduced foreign proteins. In particular, this system will be useful for studying the spatial and temporal dynamics of nuclear proteins involved in such processes as transcription, RNA processing and DNA repair.

Methods

Plasmid construction.

The plasmids pTK-Hyg, pTet-On and pEYFP-C1 were purchased from Clontech (Palo Alto); pQE30 was obtained from Qiagen; and pCFP-C1 was obtained by inserting the NheI/ScaI fragment of pCFP-C3 in the NheI/blunted BspEI sites of pEYFP-C1. p3SS–EGFP dimer lac repressor and pSV2DHFR8.32 were described previously7,9. EV-124 encoding the tetracycline-responsive transcriptional activator

(Tet-Off) and pUHD10-4B were obtained from M. Wilkinson and J. Skowronski, respectively. pUHD10-4B is composed of a XhoI-tet responsive element (TRE)–SacI–KpnI–CMV minimal promoter–SacII–

XbaI–Nef cDNA–BamHI–rabbit β-globin intron–HindIII. p3216PCβconsists of 32 units of lac opera-tor, 16 units of TRE, the CMV minimal promoter (P), CFP with peroxisome-targeting signal (C) and rabbit β-globin gene intron (β), and was constructed as follows. The TRE fragment (blunted SacI/XhoI) of pUHD10-4B was subcloned in blunted SalI/XhoI sites of pBluescriptIIKS (-). TRE was amplified to TRE16 by four-cycle amplification using XhoI, SalI and EcoRI according to ref. 7. The CMV minimal promoter was excised from pUHD10-4B with blunted KpnI/SacII sites and inserted downstream (blunt-ed SalI/SacII) of TRE16. Nef cDNA was remov(blunt-ed from pUHD10-4B by XbaI/BamHI digestion follow(blunt-ed by blunting and self-ligation. The original TRE and CMV minimal promoter were replaced with TRE16 with a CMV promoter constructed in pBluescriptIIKS(-) using XhoI/SacII sites resulting in p16Pβ. The peroxisome targeting signal-1 was fused to the C terminus of CFP by subcloning the HaeIII/KpnI frag-ment encoding 25 amino acid residues of rat acetyl-CoA oxidase C terminus in blunted EcoRI/KpnI sites of pCFP–C1. CFP–SKL cDNA was then excised with NheI/blunted KpnI site and subcloned into the XbaI/blunted SpeI site of pBluescriptII KS (-). CFP–SKL cDNA was then introduced between the

SacII and BamHI sites of p16Pβ,resulting in p16PCβ. The BamHI site was removed by digestion, blunt-ended and self-ligated. The XhoI site was then converted to a BamHI site using the oligonucleotide link-er TCGAGGATCC, resulting in p16PCβ(B-/+). p16PCβ(B-/+) was digested with BamHI and partly filled-in in the presence of dGTP and dATP with Klenow DNA polymerase. A fragment of 32 copies of

lac operator sequence was excised with XhoI/SalI from pSV2-DHFR-8.32 and partly filled-in in the

presence of dCTP, and TTP and was inserted in the digested p16PCβ(B-/+), resulting in p3216PCβ. The

HindIII site of pSV2neo was converted to a NheI site by blunt-ending and self-ligation. pSV2-EYFP-C1

was made by replacing the CMV promoter (blunted AseI/NheI) of pEYFP-C1 with the SV2 promoter (PvuII/NheI) from pSV2neo with a NheI site. pSV2-EYFP/lac repressor was made by inserting the lac repressor fragment (blunted PstI/BamHI) of p3SS–EGFP dimer lac repressor between blunt-ended

EcoRI and BglII sites of the pSV2-EYFP-C1 vector. pQE-EGFP/lac repressor was made by trimolecular

ligation of pQE30 (digested with SalI/PstI), EGFP (SalI/BamHI fragment from p3SS–EGFP dimer lac repressor) and lac repressor (BamHI/PstI fragment from p3SS–EGFP dimer lac repressor).

Cell culture and isolation of p3216PCβstably integrated clones.

BHK cells were cultured in DMEM supplemented with 10% FBS (Tet System Approved, Clontech) and 25 mM HEPES/NaOH, pH 7.3 and 10% CO2. Cells were grown in 10-cm Petri dishes and transfected

with 18 µg p3216PCβand 2 µg pTK-Hyg or pSV2hyg by a modified calcium phosphate co-precipita-tion method40. Stable transformant clones were selected with 200 µg ml–1hygromycin B for 10 days.

Then 71 isolated clones were further transfected with EV-124. Cells were fixed 24 h after removing cal-cium phosphate precipitates from the medium, and the expression of CFP–SKL was examined by fluo-rescence microscopy. Five clones exhibited inducible expression of the integrated CFP–SKL reporter plasmid, and three of the five (clones 2, 22 and 102) showed good growth. Clones 2 and 22 were derived from transfection with pTK-Hyg and clone 102 was derived from pSV2hyg. These clones were cultured in DMEM containing 150 µg ml–1hygromycin B (Sigma).

Immunoblotting and Southern blotting

Immunoblot analysis was performed as previously described41. Genomic DNA was extracted from the

cells according to ref. 42 and digested with PstI. After agarose gel electrophoresis, DNA was transferred onto a Biodyne A membrane (Life Technologies, Rockville, MD). An oligonucleotide (CACATGTG-GAATTGTGAGCGGATAACAATTTGTG, corresponding to lac operator sequence) labelled with biotin at both the 3’ and 5’ ends was used as a probe. Washing was done with 1x SSC at 37 °C. Detection was done with the PHOTOGENE Nucleic Acid Detection System ver. 2 (Life Technologies).

Electroporation and activation of transcription.

Cells were detached from the Petri dish with PBS containing 1 mM EDTA, centrifuged and resuspend-ed in complete mresuspend-edium. pTet-On (20 µg) and pSV2-EYFP/lac represor (2 µg) were addresuspend-ed into a 200-µl

(8)

ed onto a coverslip coated with mouse type IV collagen (Life Technologies). Hygromycin B was not added after electroporation. For fixed-cell observation, doxycycline was added at a final concentration of 1 µg ml–12 h after inoculation. For living-cell observations, 50 µg pTet-On was used and the

cover-slip was set into the Focht Live-cell Chamber System 2 (Bioptechs Inc., Butler, PA.) 1.5 h after electro-poration and the chamber was placed on the microscope stage. Phenol red-free L15 medium (Life Technologies) supplemented with 10% FBS was used for cell culture in the chamber. At 2 h a fluores-cence image was taken and medium containing 1 µg ml–1doxycycline was perfused into the chamber.

Medium was changed every 3 h.

Immunofluorescence and RNA fluorescence in situ hybridization.

Cells were rinsed once with PBS and fixed with 4% formaldehyde in PBS for 1 h at room temperature. After washing 3 times with PBS, cells were treated with 0.1% Triton X-100 in PBS for 15 min at room temperature. To detect the PML body, monoclonal antibody 5E10 and Texas-red conjugated anti-mouse IgG (Jackson Labs, Bar Harbor) were used. For RNA detection, a 1.2-kb BamHI/HindIII frag-ment corresponding to the rabbit β-globin intron of p3216PCβwas labelled with 16-biotin dUTP by nick translation and detection was accomplished using Texas Red-avidin (Vector Labs, Burlingame). Hybridization was performed according to ref. 43. Cells were mounted in 90% glycerol/10% PBS con-taining 25 mg ml–11,4-diazabicyclo-[2.2.2] octane (DABCO).

Overlay staining of lac operator sequence with recombinant EGFP/lac repressor.

Escherichia coli JM109 was transformed with pQE-EGFP/lac repressor and cultured with 2X YT

medi-um containing 2% glucose to suppress the expression of recombinant protein. Histidine-tagged EGFP/lac repressor was produced by removing glucose at 25 °C overnight without the addition of IPTG. Protein purification was performed according to the manufacturer’s instructions (Qiagen) using PBS containing 10% glycerol, 0.5 M NaCl and 0.1 mM DTT to avoid aggregation of the recombinant protein. After elution with imidazole (400 mM), recombinant protein was precipitated with 33% ammonium sulphate and resuspended in 0.1 M Tris-HCl, pH 7.5, containing 33% glycerol.

To localize lac operator sequences, cells were rinsed once with PBS and fixed with cold 4% formaldehyde in PBS at 4 °C for 2 min. This brief fixation is needed to maintain peroxisome structure. Cells were washed twice with PBS containing 20 mM glycine at room temperature, incubated with 0.1% saponin in PBS at 4 °C for 5 min, then rinsed twice with PBS at room temperature for 1 min each. Cells were fixed with 4% formaldehyde in PBS at room temperature for 1 h. After the second fix-ation, cells were washed three times with PBS and blocked with 10% (w/v) skimmed milk in PBS for 2 h. Cells were then incubated with 8 µg ml–1recombinant EGFP/lac repressor in PBS containing 5%

(w/v) skimmed milk, 5 mM MgCl2, 0.1 mM EDTA for 30 min. After washing three times with PBS

containing 5 mM MgCl2and 0.1 mM EDTA, cells were mounted with 90% glycerol/10% PBS

contain-ing 25 mg ml–1DABCO.

Microscopic observation and image processing.

Live and fixed cells were observed using an Olympus IX-70 inverted fluorescence microscope equipped with a Hamamatsu C4742-95-12NR ORCA camera. Filters for Texas Red (exciter HQ560/55, emitter HQ645/75, beam-splitter Q595LP), YFP (HQ520/20X, HQ520LP, Q515LP; for double labelling with CFP), YFP (HQ520/20X, HQ560/40m, Q515LP; for double labelling with Texas Red), GFP (HQ470/40, HQ525/50, Q495LP), and CFP (D436/10, D470/30, 460DCLP) were obtained from Chroma Technology Corp., Brattleboro. For double labelling with GFP and CFP, the YFP filter was used instead of that for GFP to minimize spectral overlap. For living-cell observations, a 1.25% ND filter for YFP excitation and a 6% ND filter for CFP excitation were used. Exposure time for image acquisition was 0.5 s. To adjust the focus of the YFP image, a 0.36% ND filter was used and exposure time was less than 5 s at each data acquisition point. The CCD camera was controlled by OpenLab software (Improvision, Boston), and acquired images were processed using Adobe PhotoShop software.

RECEIVED 28 JUNE; REVISED 22 AUGUST; ACCEPTED 22 AUGUST; PUBLISHED 10 NOVEMBER 2000

1. Spector, D. L. Macromolecular domains within the cell nucleus. Annu. Rev. Cell Biol. 9, 265–315 (1993).

2. Lamond, A. I. & Earnshaw, W. C. Structure and function in the nucleus. Science 280, 547–553 (1998). 3. Misteli, T. & Spector, D. L. Applications of the green fluorescent protein in cell biology and

biotech-nology. Nature Biotechnol. 15, 961–964 (1997).

4. Tsien, R. Y. The green fluorescent protein. Annu. Rev. Biochem. 67, 509–544 (1998).

5. Ellenberg, J., Lippincott-Schwartz, J. & Presley, J. F. Dual-colour imaging with GFP variants. Trends

Cell. Biol. 9, 52–56 (1999).

6. Belmont, A. S. & Straight, A.F. In vivo visualization of chromosomes using lac operator–repressor binding. Trends Cell. Biol. 8, 121–124 (1998).

7. Robinett, C. C. et al. In vivo localization of DNA sequences and visualization of large-scale chro-matin organization using lac operator/repressor recognition. J. Cell Biol. 135, 1685–1700 (1996). 8. Li, G., Sudlow, G. & Belmont, A. S. Interphase cell cycle dynamics of a late-replicating,

heterochro-matic homogeneously staining region: precise choreography of condensation/decondensation and nuclear positioning. J. Cell Biol. 140, 975–989 (1998).

9. Tumbar, T., Sudlow, G. & Belmont, A. S. Large-scale chromatin unfolding and remodeling induced by VP16 acidic activation domain. J. Cell Biol. 145, 1341–1354 (1999).

10. Straight, A. F., Belmont, A. S., Robinett, C. C. & Murray, A. W. GFP tagging of budding yeast chro-mosomes reveals that protein-protein interactions can mediate sister chromatid cohesion. Curr.

Biol. 6, 1599–1608 (1996).

11. Minshull, J. et al. Protein phosphatase 2A regulates MPF activity and sister chromatid cohesion in budding yeast. Curr. Biol. 6, 1609–1620 (1996).

12. Miyazawa, S. et al. Peroxisome targeting signal of rat liver acyl-coenzyme A oxidase resides at the carboxy terminus. Mol. Cell. Biol. 9, 83–91 (1989).

13. Miller, J. H. & Reznikoff, W. A. The Operon, 17–220 (Cold Spring Harbor Laboratory Press, New York, 1980).

14. Maul, G. G., Negorev, D., Bell, P. & Ishov, A. M. Properties and assembly mechanisms of ND10, PML bodies, or PODs. J. Struct. Biol. 129, 278–287 (2000).

15. Zhong, S., Salomoni, P. & Pandolfi, P. P. The transcriptional role of PML and the nuclear body.

Nature Cell Biol. 2, E85–E90 (2000).

16. Weintraub, H. & Groudine, M. Chromosomal subunits in active genes have an altered conforma-tion. Science 193, 848–856 (1976).

17. Paranjape, S. M., Kamakaka, R. T. & Kadonaga, J. T. Role of chromatin structure in the regulation of transcription by RNA polymerase II. Annu. Rev. Biochem. 63, 265–297 (1994).

18. Armstrong, J. A. & Emerson, B. M. Transcription of chromatin: these are complex times. Curr.

Opin. Genet. Dev. 8, 165–172 (1998).

19. Kuo, M. H. & Allis, C. D. Roles of histone acetyltransferases and deacetylases in gene regulation.

Bioessays 20, 615–626 (1998).

20. Pederson, T. Chromatin structure and gene transcription: nucleosomes permit a new synthesis. Int.

Rev. Cytol. 55, 1–21 (1978).

21. Manuelidis, L. Different central nervous system cell types display distinct and nonrandom arrange-ments of satellite DNA sequences. Proc. Natl Acad. Sci. USA 81, 3123–3127 (1984).

22. Manuelidis, L. & Borden, J. Reproducible compartmentalization of individual chromosome domains in human CNS cells revealed by in situ hybridization and three-dimensional reconstruc-tion. Chromosoma 96, 397–410 (1988).

23. Ferguson, M. & Ward, D. C. Cell cycle dependent chromosomal movement in pre-mitotic human T-lymphocyte nuclei. Chromosoma 101, 557–565 (1992).

24. Dietzel, S. et al. Three-dimensional distribution of centromeric or paracentromeric heterochro-matin of chromosomes 1, 7, 15, and 17 in human lymphocyte nuclei studied with light microscopic axial tomography. Bioimaging 3, 121–133 (1995).

25. Abney, J. R., Cutler, B., Fillbach, M. L., Axelrod, D. & Scalettar, B. A. Chromatin dynamics in inter-phase nuclei and its implications for nuclear structure. J. Cell Biol. 137, 1459–1468 (1997). 26. Marshall, W. F. et al. Interphase chromosomes undergo constrained diffusional motion in living

cells. Curr. Biol. 7, 930–939 (1997).

27. Manuelidis, L. Individual interphase chromosome domains revealed by in-situ hybridization. Hum.

Genet. 71, 288–293 (1985).

28. Janevski, J., Park, P. C. & De Boni, U. Organization of centromeric domains in hepatocyte nuclei: rearrangement associated with de novo activation of the vitellogenin gene family in Xenopus laevis.

Exp. Cell Res. 217, 227–239 (1995).

29. Bartholdi, M. F. Nuclear distribution of centromeres during the cell cycle of human diploid fibrob-lasts. J. Cell Sci. 99, 255–263 (1991).

30. Funabiki, H., Hagan, I., Uzawa, S. & Yanagida, M. Cell cycle-dependent specific positioning and clustering of centromeres and telomeres in fission yeast. J. Cell Biol. 121, 961–976 (1993). 31. LaSalle, J. M. & Lalande, M. Homologous association of oppositely imprinted chromosomal

domains. Science 272, 725–728 (1996).

32. Borden, J. & Manuelidis, L. Movement of the X chromosome in epilepsy. Science 242, 1687–1691 (1988). 33. Guldner, H. H., Szostecki, C., Grotzinger, T. & Will, H. IFN enhance expression of Sp100, an

autoantigen in primary biliary cirrhosis. J. Immunol. 149, 4067–4073 (1992).

34. Koken, M. H. et al. The t(15;17) translocation alters a nuclear body in a retinoic acid-reversible fashion. EMBO J. 13, 1073–1083 (1994).

35. Korioth, F., Gieffers, C., Maul, G. G. & Frey, J. Molecular characterization of NDP52, a novel pro-tein of the nuclear domain 10, which is redistributed upon virus infection and interferon treat-ment. J. Cell Biol. 130, 1–13 (1995).

36. Maul, G. G., Yu, E., Ishov, A. M. & Epstein, A. L. Nuclear domain 10 (ND10) associated proteins are also present in nuclear bodies and redistribute to hundreds of nuclear sites after stress. J. Cell.

Biochem. 59, 498–513 (1995).

37. Ishov, A. M. & Maul, G. G. The periphery of nuclear domain 10 (ND10) as site of DNA virus depo-sition. J. Cell Biol. 134, 815–826 (1996).

38. Maul, G. G., Jensen, D. E., Ishov, A. M., Herlyn, M. & Rauscher, F. J. III Nuclear redistribution of BRCA1 during viral infection. Cell Growth Differ. 9, 743–755 (1998).

39. Gongora, C. et al. Molecular cloning of a new interferon-induced PML nuclear body-associated protein. J. Biol. Chem. 272, 19457–19463 (1997).

40. Chen, C. & Okayama, H. High-efficiency transformation of mammalian cells by plasmid DNA.

Mol. Cell. Biol. 7, 2745–2752 (1987).

41. Mintz, P. J., Patterson, S. D., Neuwald, A. F., Spahr, C. S. & Spector, D. L. Purification and biochemi-cal characterization of interchromatin granule clusters. EMBO J. 18, 4308–4320 (1999). 42. Sambrook, J., Fritsch, E. F. & Maniatis, T. Molecular Cloning (Cold Spring Harbor Laboratory Press,

New York, 1989).

43. Spector, D. L., Goldman, R. D. & Leinwand, L. A. Cells: A Laboratory Manual (Cold Spring Harbor Laboratory Press, New York, 1998).

ACKNOWLEDGEMENTS

We thank T. Misteli for discussions and P. Sacco-Bubulya and N. Saitoh for reviewing the manuscript. pW7-C3 (=pCFP-C3), which encoded CFP, was provided by R. Tsien, EV-124 was provided by M. Wilkinson and pUHD10-4B was provided by J. Skowronski, and monoclonal antibody 5E10 was obtained from R. van Driel. S.M.J. is supported by an NIH/NCI training grant 5T32CA09311. D.L.S. is funded by a grant from NIGMS (NIH 498100).

Correspondence and requests for materials should be addressed to D.L.S. Supplementary Information is available on Nature Cell Biology’s World-Wide Web site (http://cellbio.nature.com) or as paper copy from the London editorial office of Nature Cell Biology.

References

Related documents

Once this job is over then location master as per NSAP-MIS standard will be provided to states to further map NSAP-MIS location codes such as district code,

Automated preform manufacturing using HGT as heat source, resin infusion of preforms using hot VARTM, inspection of manufactured laminate quality, evaluation of

Figure 16: Plot of the theoretical electrochemical

departmental websites rests with the Digital Marketing team and the Marketing provisions within each individual School. Responsibility for the training and access of individual

Through the use of evaporation rate coefficients derived from formation free energies calculated by quantum chemi- cal methods for clusters containing dimethylamine or ammo- nia

• to aid teachers in understanding the role of student standards in language education • to provide instruction in training students effectively in the use of technology •

The risk assessment will guide the development of the center’s written plan for infection prevention goals, interventions, and evaluation to improve patient safety

Lebedev Physical Institute, Moscow, Russia 41: Also at Budker Institute of Nuclear Physics, Novosibirsk, Russia 42: Also at Faculty of Physics, University of Belgrade, Belgrade,